1
|
Li J, Cai C, Teo WW, Chin KS, Naing YT, Song S, Nelson F, Qiang L, Xu D, Sun L. Isoform usage as a distinct regulatory layer driving nutrient-responsive metabolic adaptation. Cell Metab 2025; 37:772-787.e6. [PMID: 39938511 DOI: 10.1016/j.cmet.2025.01.009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/13/2023] [Revised: 08/16/2024] [Accepted: 01/10/2025] [Indexed: 02/14/2025]
Abstract
Transcriptome modulation is essential for metabolic adaptation to nutrient environments. However, the role of isoform usage, a crucial transcriptome component, is not yet fully understood. This study outlines the landscape of isoform-usage modulations across major metabolic organs in both mice and monkeys, spanning diverse metabolic states. Our in-depth analysis identifies numerous isoform-usage events, intricately influenced by nutrient challenges and largely independent of gene expression regulation. Comparative analyses of mice and monkeys highlight hundreds of conserved isoform events that exhibit consistent responses to nutrient challenges across species and correlate with human metabolic traits. When analyzing splicing factor-binding motifs in nutrient-regulated events, HuR emerges as the predominant orchestrator of the isoform network in adipocytes, which is validated using an adipose tissue-specific knockout and an Ap2-promoter-driven transgenic mouse model. In summary, our results offer a comprehensive perspective on isoform usage in metabolic regulation, setting a platform for future functional inquiries.
Collapse
Affiliation(s)
- Jia Li
- Cardiovascular and Metabolic Disorders Program, Duke-NUS Medical School, Singapore, Singapore
| | - Chaoqun Cai
- Cardiovascular and Metabolic Disorders Program, Duke-NUS Medical School, Singapore, Singapore
| | - Wei Wen Teo
- Cardiovascular and Metabolic Disorders Program, Duke-NUS Medical School, Singapore, Singapore
| | - Kai Shin Chin
- Cardiovascular and Metabolic Disorders Program, Duke-NUS Medical School, Singapore, Singapore
| | - Yadanar Than Naing
- Cardiovascular and Metabolic Disorders Program, Duke-NUS Medical School, Singapore, Singapore
| | - Shengren Song
- The Hormel Institute, University of Minnesota, Austin, MN, USA
| | - Franklin Nelson
- The Hormel Institute, University of Minnesota, Austin, MN, USA
| | - Li Qiang
- Naomi Berrie Diabetes Center and Department of Pathology and Cell Biology, Columbia University, New York, NY, USA
| | - Dan Xu
- The Hormel Institute, University of Minnesota, Austin, MN, USA.
| | - Lei Sun
- TaiKang Center for Life and Medical Sciences, TaiKang Medical School, Wuhan University, Wuhan, China; Cardiovascular and Metabolic Disorders Program, National Heart Research Institute, DUKE-NUS Medical School, Singapore, Singapore.
| |
Collapse
|
2
|
Liu Y, Shen S, Wang X, Chen H, Ren W, Wei H, Li K, Li L. GATA3-Driven ceRNA Network in Lung Adenocarcinoma Bone Metastasis Progression and Therapeutic Implications. Cancers (Basel) 2025; 17:559. [PMID: 39941924 PMCID: PMC11816722 DOI: 10.3390/cancers17030559] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2025] [Revised: 01/18/2025] [Accepted: 01/23/2025] [Indexed: 02/16/2025] Open
Abstract
Background/Objectives: Bone metastasis is a common and severe complication of lung adenocarcinoma (LUAD), impacting prognosis and treatment outcomes. Understanding the molecular mechanisms behind LUAD bone metastasis (LUADBM) is essential for developing new therapeutic strategies. The interactions between long non-coding RNAs (lncRNAs), microRNAs (miRNAs), and mRNAs in the competing endogenous RNA (ceRNA) network are crucial in cancer progression and metastasis, but the regulatory mechanisms in LUADBM remain unclear. Methods: Microarray analysis was performed on clinical samples, followed by weighted gene co-expression network analysis (WGCNA) and construction of a ceRNA network. Molecular mechanisms were validated using colony formation assays, transwell migration assays, wound healing assays to assess cell migration, and osteoclastogenesis assays to evaluate osteoclast differentiation. Potential therapeutic drugs and their binding affinities were predicted using the CMap database and Kdeep. The interaction between the small-molecule drug and its target protein was confirmed by surface plasmon resonance (SPR) and drug affinity responsive target stability (DARTS) assays. Mechanistic insights and therapeutic efficacy were further validated using patient-derived organoid (PDO) cultures, drug sensitivity assays, and in vivo drug treatments. Results: Our results identified the XLOC_006941/hsa-miR-543/NPRL3 axis as a key regulatory pathway in LUADBM. We also demonstrated that GATA3-driven Th2 cell infiltration creates an immunosuppressive microenvironment that promotes metastasis. Additionally, we confirmed that the inhibitor E7449 effectively targets NPRL3, and its combination with the IL4R-blocking antibody dupilumab resulted in improved therapeutic outcomes in LUADBM. Conclusions: These findings offer new insights into the molecular mechanisms of LUADBM and highlight potential therapeutic targets, including the XLOC_006941/miR-543/NPRL3 axis and GATA3-driven Th2 cell infiltration. The dual-target therapy combining E7449 with dupilumab shows promise for improving patient outcomes in LUADBM, warranting further clinical evaluation.
Collapse
Affiliation(s)
- Yun Liu
- Shanghai Key Laboratory of Regulatory Biology, Institute of Biomedical Sciences, East China Normal University, Shanghai 200241, China
| | - Shihui Shen
- Joint Center for Translational Medicine, Shanghai Fifth People’s Hospital, Fudan University and School of Life Science, East China Normal University, Shanghai 200240, China
- School of Life Sciences, East China Normal University, Shanghai 200241, China
| | - Xudong Wang
- Department of Orthopedic Oncology, Changzheng Hospital, Shanghai 200003, China
- Department of Orthopedics, 905th Hospital of PLA Navy, Shanghai 200030, China
| | - Hansen Chen
- Shanghai Key Laboratory of Regulatory Biology, Institute of Biomedical Sciences, East China Normal University, Shanghai 200241, China
| | - Wenjie Ren
- Shanghai Key Laboratory of Regulatory Biology, Institute of Biomedical Sciences, East China Normal University, Shanghai 200241, China
| | - Haifeng Wei
- Department of Orthopedic Oncology, Changzheng Hospital, Shanghai 200003, China
- Department of Orthopedics, 905th Hospital of PLA Navy, Shanghai 200030, China
| | - Kun Li
- Health Science Center, East China Normal University, Shanghai 200241, China
| | - Lei Li
- Joint Center for Translational Medicine, Shanghai Fifth People’s Hospital, Fudan University and School of Life Science, East China Normal University, Shanghai 200240, China
- School of Life Sciences, East China Normal University, Shanghai 200241, China
| |
Collapse
|
3
|
Chen X, Han W, Yang R, Zhu X, Li S, Wang Y, Sun X, Li Y, Bao L, Zhang L, Wang S, Wang J. Transcriptome Analysis Reveals the lncRNA-mRNA Co-expression Network Regulating the Aestivation of Sea Cucumber. MARINE BIOTECHNOLOGY (NEW YORK, N.Y.) 2024; 27:15. [PMID: 39611876 DOI: 10.1007/s10126-024-10388-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/16/2024] [Accepted: 10/03/2024] [Indexed: 11/30/2024]
Abstract
LncRNAs are long non-coding RNAs that are widely recognized as crucial regulators of gene expression and metabolic control, involved in numerous dormancy-related processes. Aestivation is a common hypometabolism strategy of sea cucumber (Apostichopus japonicus) in response to high-temperature conditions and is typically characterized by the degradation of the intestine and respiratory tree. Although the aestivation process has been extensively studied in sea cucumbers, the role of lncRNAs in the context of aestivation states remains a conspicuous knowledge gap. Here, we identified and characterized 14,711 lncRNAs in A. japonicus and analyzed their differential expression patterns during the aestivation process in the intestine and respiratory tree. The results revealed the physiological differences, especially the metabolic processes, between the intestine and respiratory tree during the aestivation. The co-expression network of lncRNA-mRNA suggested the dominant role of lncRNA in regulating the differential response of the intestine and respiratory trees. Differentially co-expressed factors were significantly enriched in the deep-aestivation stage-specific modules. Conserved co-expressed factors included several transcription factors known to be involved in rhythm regulation, such as Klf2 and Egr1. Furthermore, a specific trans-acting lncRNA (lncrna.1393.1) was identified as a potential regulator of Klf2 and Egr1. Overall, the systematic identification, characterization, and expression analysis of lncRNAs in A. japonicus enhanced our knowledge of long non-coding regulation of aestivation in sea cucumber and provided new clues for understanding the common "toolkit" of dormancy regulatory mechanisms.
Collapse
Affiliation(s)
- Xiaomei Chen
- Fang Zongxi Center for Marine Evo-Devo & MOE Key Laboratory of Marine Genetics and Breeding, Ocean University of China, Qingdao, 266003, China
- Laboratory for Marine Biology and Biotechnology, Qingdao National Laboratory for Marine Science and Technology, Qingdao, 266237, China
| | - Wentao Han
- Fang Zongxi Center for Marine Evo-Devo & MOE Key Laboratory of Marine Genetics and Breeding, Ocean University of China, Qingdao, 266003, China
| | - Rui Yang
- Fang Zongxi Center for Marine Evo-Devo & MOE Key Laboratory of Marine Genetics and Breeding, Ocean University of China, Qingdao, 266003, China
| | - Xuan Zhu
- Fang Zongxi Center for Marine Evo-Devo & MOE Key Laboratory of Marine Genetics and Breeding, Ocean University of China, Qingdao, 266003, China
| | - Shengwen Li
- Fang Zongxi Center for Marine Evo-Devo & MOE Key Laboratory of Marine Genetics and Breeding, Ocean University of China, Qingdao, 266003, China
- Laboratory for Marine Biology and Biotechnology, Qingdao National Laboratory for Marine Science and Technology, Qingdao, 266237, China
| | - Yangfan Wang
- Fang Zongxi Center for Marine Evo-Devo & MOE Key Laboratory of Marine Genetics and Breeding, Ocean University of China, Qingdao, 266003, China
| | - Xue Sun
- Fang Zongxi Center for Marine Evo-Devo & MOE Key Laboratory of Marine Genetics and Breeding, Ocean University of China, Qingdao, 266003, China
| | - Yuli Li
- Fang Zongxi Center for Marine Evo-Devo & MOE Key Laboratory of Marine Genetics and Breeding, Ocean University of China, Qingdao, 266003, China
| | - Lisui Bao
- Institute of Evolution & Marine Biodiversity, Ocean University of China, Qingdao, 266003, China
| | - Lingling Zhang
- Fang Zongxi Center for Marine Evo-Devo & MOE Key Laboratory of Marine Genetics and Breeding, Ocean University of China, Qingdao, 266003, China
| | - Shi Wang
- Southern Marine Science and Engineering Guangdong Laboratory, Guangzhou, 511458, China
- Key Laboratory of Tropical Aquatic Germplasm of Hainan Province, Sanya Oceanographic Institution, Ocean University of China, Sanya, 572000, China
| | - Jing Wang
- Fang Zongxi Center for Marine Evo-Devo & MOE Key Laboratory of Marine Genetics and Breeding, Ocean University of China, Qingdao, 266003, China.
- Laboratory for Marine Biology and Biotechnology, Qingdao National Laboratory for Marine Science and Technology, Qingdao, 266237, China.
| |
Collapse
|
4
|
Ngowi EE, Lu T, Liu Q, Xie X, Wang N, Luo L, Deng L, Zhou Y, Zhang Z, Qiao A. Biofluid-Derived Exosomal LncRNAs: Their Potential in Obesity and Related Comorbidities. BIOLOGY 2024; 13:976. [PMID: 39765643 PMCID: PMC11673191 DOI: 10.3390/biology13120976] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/05/2024] [Revised: 11/21/2024] [Accepted: 11/24/2024] [Indexed: 01/03/2025]
Abstract
Obesity has escalated into a critical global health crisis, tripling in prevalence since the mid-1970s. This increase mirrors the rise in metabolic-associated diseases such as type 2 diabetes (T2D) and its complications, certain cancers, and cardiovascular conditions. While substantial research efforts have enriched our understanding and led to the development of innovative management strategies for these diseases, the suboptimal response rates of existing therapies remain a major obstacle to effectively managing obesity and its associated conditions. Over the years, inter-organ communication (IOC) has emerged as a crucial factor in the development and progression of metabolic disorders. Exosomes, which are nano-sized vesicular couriers released by cells, play a significant role in this communication by transporting proteins, lipids, and nucleic acids across cellular landscapes. The available evidence indicates that exosomal RNAs present in biofluids such as blood, urine, milk, vitreous humor (VH), and cerebrospinal fluid (CSF) are altered in numerous diseases, suggesting their diagnostic and therapeutic potential. Long non-coding RNAs contained in exosomes (exo-lncRNAs) have attracted considerable interest, owing to their ability to interact with critical components involved in a multitude of metabolic pathways. Recent studies have found that alterations in exo-lncRNAs in biofluids correlate with several metabolic parameters in patients with metabolic-associated conditions; however, their exact roles remain largely unclear. This review highlights the diagnostic and therapeutic potential of exosomal lncRNAs in obesity and its associated conditions, emphasizing their role in IOC and disease progression, aiming to pave the way for further research in this promising domain.
Collapse
Affiliation(s)
- Ebenezeri Erasto Ngowi
- Zhongshan Institute for Drug Discovery, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Zhongshan 528400, China; (E.E.N.); (T.L.); (Q.L.); (X.X.); (N.W.); (L.L.); (L.D.) (Y.Z.); (Z.Z.)
- Shanghai Institute of Materia Medica, Chinese Academy of Sciences, 555 Zu Chong Zhi Road, Shanghai 201203, China
- University of Chinese Academy of Sciences, Beijing 101408, China
| | - Tuyan Lu
- Zhongshan Institute for Drug Discovery, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Zhongshan 528400, China; (E.E.N.); (T.L.); (Q.L.); (X.X.); (N.W.); (L.L.); (L.D.) (Y.Z.); (Z.Z.)
| | - Qing Liu
- Zhongshan Institute for Drug Discovery, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Zhongshan 528400, China; (E.E.N.); (T.L.); (Q.L.); (X.X.); (N.W.); (L.L.); (L.D.) (Y.Z.); (Z.Z.)
| | - Xianghong Xie
- Zhongshan Institute for Drug Discovery, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Zhongshan 528400, China; (E.E.N.); (T.L.); (Q.L.); (X.X.); (N.W.); (L.L.); (L.D.) (Y.Z.); (Z.Z.)
| | - Ning Wang
- Zhongshan Institute for Drug Discovery, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Zhongshan 528400, China; (E.E.N.); (T.L.); (Q.L.); (X.X.); (N.W.); (L.L.); (L.D.) (Y.Z.); (Z.Z.)
| | - Liping Luo
- Zhongshan Institute for Drug Discovery, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Zhongshan 528400, China; (E.E.N.); (T.L.); (Q.L.); (X.X.); (N.W.); (L.L.); (L.D.) (Y.Z.); (Z.Z.)
| | - Lijuan Deng
- Zhongshan Institute for Drug Discovery, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Zhongshan 528400, China; (E.E.N.); (T.L.); (Q.L.); (X.X.); (N.W.); (L.L.); (L.D.) (Y.Z.); (Z.Z.)
| | - Yinghua Zhou
- Zhongshan Institute for Drug Discovery, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Zhongshan 528400, China; (E.E.N.); (T.L.); (Q.L.); (X.X.); (N.W.); (L.L.); (L.D.) (Y.Z.); (Z.Z.)
| | - Zhihong Zhang
- Zhongshan Institute for Drug Discovery, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Zhongshan 528400, China; (E.E.N.); (T.L.); (Q.L.); (X.X.); (N.W.); (L.L.); (L.D.) (Y.Z.); (Z.Z.)
| | - Aijun Qiao
- Zhongshan Institute for Drug Discovery, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Zhongshan 528400, China; (E.E.N.); (T.L.); (Q.L.); (X.X.); (N.W.); (L.L.); (L.D.) (Y.Z.); (Z.Z.)
- Shanghai Institute of Materia Medica, Chinese Academy of Sciences, 555 Zu Chong Zhi Road, Shanghai 201203, China
- University of Chinese Academy of Sciences, Beijing 101408, China
| |
Collapse
|
5
|
Zhou Y, Zhang S, Qiu G, Wang X, Yonemura A, Xu H, Cui G, Deng S, Chun J, Chen N, Xu M, Song X, Wang J, Xu Z, Deng Y, Evert M, Calvisi DF, Lin S, Wang H, Chen X. TSC/mTORC1 mediates mTORC2/AKT1 signaling in c-MYC-induced murine hepatocarcinogenesis via centromere protein M. J Clin Invest 2024; 134:e174415. [PMID: 39325536 PMCID: PMC11563669 DOI: 10.1172/jci174415] [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: 08/14/2023] [Accepted: 09/20/2024] [Indexed: 09/28/2024] Open
Abstract
Activated mTORC2/AKT signaling plays a role in hepatocellular carcinoma (HCC). Research has shown that TSC/mTORC1 and FOXO1 are distinct downstream effectors of AKT signaling in liver regeneration and metabolism. However, the mechanisms by which these pathways mediate mTORC2/AKT activation in HCC are not yet fully understood. Amplification and activation of c-MYC are key molecular events in HCC. In this study, we explored the roles of tuberous sclerosis complex/mTORC1 (TSC/mTORC1) and FOXO1 as downstream effectors of mTORC2/AKT1 in c-MYC-induced hepatocarcinogenesis. Using various genetic approaches in mice, we found that manipulating the FOXO pathway had a minimal effect on c-MYC-induced HCC. In contrast, loss of mTORC2 inhibited c-MYC-induced HCC, an effect that was completely reversed by ablation of TSC2, which activated mTORC1. Additionally, we discovered that p70/RPS6 and 4EBP1/eIF4E acted downstream of mTORC1, regulating distinct molecular pathways. Notably, the 4EBP1/eIF4E cascade is crucial for cell proliferation and glycolysis in c-MYC-induced HCC. We also identified centromere protein M (CENPM) as a downstream target of the TSC2/mTORC1 pathway in c-MYC-driven hepatocarcinogenesis, and its ablation entirely inhibited c-MYC-dependent HCC formation. Our findings demonstrate that the TSC/mTORC1/CENPM pathway, rather than the FOXO cascade, is the primary signaling pathway regulating c-MYC-driven hepatocarcinogenesis. Targeting CENPM holds therapeutic potential for treating c-MYC-driven HCC.
Collapse
Affiliation(s)
- Yi Zhou
- Department of Infectious Diseases, the First Affiliated Hospital of Xi’an Jiaotong University, Xi’an, China
- Department of Bioengineering and Therapeutic Sciences and Liver Center, UCSF, San Francisco, California, USA
| | - Shu Zhang
- Department of Head and Neck Oncology, Cancer Center
- Department of Radiation Oncology, Cancer Center
| | - Guoteng Qiu
- Division of Liver Surgery, Department of General Surgery, and
- Laboratory of Liver Surgery, West China Hospital, Sichuan University, Chengdu, China
| | - Xue Wang
- Cancer Biology Program, University of Hawaii Cancer Center, Honolulu, Hawaii, USA
| | - Andrew Yonemura
- Cancer Biology Program, University of Hawaii Cancer Center, Honolulu, Hawaii, USA
| | - Hongwei Xu
- Department of Bioengineering and Therapeutic Sciences and Liver Center, UCSF, San Francisco, California, USA
- Division of Liver Surgery, Department of General Surgery, and
| | - Guofei Cui
- Department of Bioengineering and Therapeutic Sciences and Liver Center, UCSF, San Francisco, California, USA
- Cancer Biology Program, University of Hawaii Cancer Center, Honolulu, Hawaii, USA
| | - Shanshan Deng
- Department of Bioengineering and Therapeutic Sciences and Liver Center, UCSF, San Francisco, California, USA
- Cancer Biology Program, University of Hawaii Cancer Center, Honolulu, Hawaii, USA
| | - Joanne Chun
- Cancer Biology Program, University of Hawaii Cancer Center, Honolulu, Hawaii, USA
| | - Nianyong Chen
- Department of Head and Neck Oncology, Cancer Center
- Department of Radiation Oncology, Cancer Center
- Laboratory of Single Cell Research and Liquid Biopsy, Cancer Center, West China Hospital, Sichuan University, Chengdu, China
| | - Meng Xu
- Department of General Surgery, the Second Affiliated Hospital of Xi’an Jiaotong University, Xi’an, China
| | - Xinhua Song
- School of Traditional Chinese Medicine, Laboratory for Clinical Medicine, Capital Medical University, Beijing, China
| | - Jingwen Wang
- School of Traditional Chinese Medicine, Laboratory for Clinical Medicine, Capital Medical University, Beijing, China
| | - Zijing Xu
- School of Traditional Chinese Medicine, Laboratory for Clinical Medicine, Capital Medical University, Beijing, China
| | - Youping Deng
- Cancer Biology Program, University of Hawaii Cancer Center, Honolulu, Hawaii, USA
- Department of Quantitative Health Sciences, John A. Burns School of Medicine, Honolulu, Hawaii, USA
| | - Matthias Evert
- Institute of Pathology, University of Regensburg, Regensburg, Germany
| | - Diego F. Calvisi
- Institute of Pathology, University of Regensburg, Regensburg, Germany
| | - Shumei Lin
- Department of Infectious Diseases, the First Affiliated Hospital of Xi’an Jiaotong University, Xi’an, China
| | - Haichuan Wang
- Division of Liver Surgery, Department of General Surgery, and
- Laboratory of Liver Surgery, West China Hospital, Sichuan University, Chengdu, China
| | - Xin Chen
- Department of Bioengineering and Therapeutic Sciences and Liver Center, UCSF, San Francisco, California, USA
- Cancer Biology Program, University of Hawaii Cancer Center, Honolulu, Hawaii, USA
| |
Collapse
|
6
|
Jaso-Vera ME, Takaoka S, Patel I, Ruan X. Integrative regulation of hLMR1 by dietary and genetic factors in nonalcoholic fatty liver disease and hyperlipidemia. Hum Genet 2024; 143:897-906. [PMID: 38493444 DOI: 10.1007/s00439-024-02654-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2023] [Accepted: 02/05/2024] [Indexed: 03/19/2024]
Abstract
Long non-coding RNA (lncRNA) genes represent a large class of transcripts that are widely expressed across species. As most human lncRNAs are non-conserved, we recently employed a unique humanized liver mouse model to study lncRNAs expressed in human livers. We identified a human hepatocyte-specific lncRNA, hLMR1 (human lncRNA metabolic regulator 1), which is induced by feeding and promotes hepatic cholesterol synthesis. Recent genome-wide association studies (GWAS) found that several single-nucleotide polymorphisms (SNPs) from the hLMR1 gene locus are associated with blood lipids and markers of liver damage. These results suggest that dietary and genetic factors may regulate hLMR1 to affect disease progression. In this study, we first screened for nutritional/hormonal factors and found that hLMR1 was robustly induced by insulin/glucose in cultured human hepatocytes, and this induction is dependent on the transcription factor SREBP1. We then tested if GWAS SNPs genetically linked to hLMR1 could regulate hLMR1 expression. We found that DNA sequences flanking rs9653945, a SNP from the last exon of the hLMR1 gene, functions as an enhancer that can be robustly activated by SREBP1c depending on the presence of rs9653945 major allele (G). We further performed CRISPR base editing in human HepG2 cells and found that rs9653945 major (G) to minor (A) allele modification resulted in blunted insulin/glucose-induced expression of hLMR1. Finally, we performed genotyping and gene expression analyses using a published human NAFLD RNA-seq dataset and found that individuals homozygous for rs9653945-G have a higher expression of hLMR1 and risk of NAFLD. Taken together, our data support a model that rs9653945-G predisposes individuals to insulin/glucose-induced hLMR1, contributing to the development of hyperlipidemia and NAFLD.
Collapse
Affiliation(s)
- Marcos E Jaso-Vera
- Division of Endocrinology, Diabetes and Metabolism, Johns Hopkins University School of Medicine, Baltimore, MD, USA
- Institute for Fundamental Biomedical Research, Johns Hopkins All Childrens Hospital, 600 Fifth Street S., St. Petersburg, FL, 33701, USA
| | - Shohei Takaoka
- Division of Endocrinology, Diabetes and Metabolism, Johns Hopkins University School of Medicine, Baltimore, MD, USA
- Institute for Fundamental Biomedical Research, Johns Hopkins All Childrens Hospital, 600 Fifth Street S., St. Petersburg, FL, 33701, USA
| | - Ishika Patel
- Division of Endocrinology, Diabetes and Metabolism, Johns Hopkins University School of Medicine, Baltimore, MD, USA
- Institute for Fundamental Biomedical Research, Johns Hopkins All Childrens Hospital, 600 Fifth Street S., St. Petersburg, FL, 33701, USA
| | - Xiangbo Ruan
- Division of Endocrinology, Diabetes and Metabolism, Johns Hopkins University School of Medicine, Baltimore, MD, USA.
- Institute for Fundamental Biomedical Research, Johns Hopkins All Childrens Hospital, 600 Fifth Street S., St. Petersburg, FL, 33701, USA.
| |
Collapse
|
7
|
Li J, Huang Q, Nie Q, Luo Y, Zeng H, Zhang Y, He X, Liu J. Effects of myo-inositol on regulating glucose and lipid metabolism and alternative splicing events coexpressed with lncRNAs in the liver tissues of diabetic mice. Heliyon 2024; 10:e32460. [PMID: 38933931 PMCID: PMC11201111 DOI: 10.1016/j.heliyon.2024.e32460] [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: 04/19/2023] [Revised: 05/26/2024] [Accepted: 06/04/2024] [Indexed: 06/28/2024] Open
Abstract
Objective Recent studies have shown that gene alternative splicing (AS) and long noncoding RNAs (lncRNAs) are involved in diabetes mellitus (DM) and its complications. Currently, myo-inositol (MI) is considered as effective for the treatment of insulin resistance and lipid metabolism disorders in diabetes patients. We hope to better explore the potential roles of gene AS and lncRNAs in liver glucose and lipid metabolism in diabetes, as well as the effects of myo-inositol treatment, through transcriptome analysis. Methods This study analysed glucose and lipid metabolism-related biochemical indicators and liver HE staining in four groups of mice: the control group (Ctrl group), the diabetes group (DM group), the myo-inositol treatment group (MI group), and the metformin treatment group (Met group). The changes in relevant gene-regulated alternative splicing events (RASEs) and lncRNAs were analysed by RNA sequencing of liver tissue, and coexpression analysis and functional enrichment analysis were used to predict the possible lncRNAs and RASEs involved in liver glucose and lipid metabolism. Result Metformin and myo-inositol alleviated insulin resistance, lipid metabolism disorders, and hepatic steatosis in diabetic mice. Transcriptome sequencing analysis revealed differential splicing events of genes related to lipid metabolism and differentially expressed lncRNAs (DElncRNAs). Six different lncRNAs and their potentially interacting splicing events were predicted. Conclusion The present study revealed novel changes in RASEs and lncRNAs in the livers of diabetic mice following treatment with myo-inositol, which may shed light on the potential mechanisms by which myo-inositol delays and treats the progression of hepatic glucose and lipid metabolism in diabetes.
Collapse
Affiliation(s)
- Jin'e Li
- Department of Endocrinology and Metabolism of the Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, 330006, Jiangxi, China
- Institute for the Study of Endocrinology and Metabolism in Jiangxi Province, Nanchang, 330006, Jiangxi, China
| | - Qiulan Huang
- Department of Endocrinology and Metabolism of the Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, 330006, Jiangxi, China
| | - Qin Nie
- Zhongnan Hospital of Wuhan University, Wuhan, 430071, Hubei, China
| | - Yunfei Luo
- Department of Endocrinology and Metabolism of the Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, 330006, Jiangxi, China
| | - Haixia Zeng
- Department of Endocrinology and Metabolism of the Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, 330006, Jiangxi, China
- Institute for the Study of Endocrinology and Metabolism in Jiangxi Province, Nanchang, 330006, Jiangxi, China
| | - Yuying Zhang
- Department of Endocrinology and Metabolism of the Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, 330006, Jiangxi, China
- Institute for the Study of Endocrinology and Metabolism in Jiangxi Province, Nanchang, 330006, Jiangxi, China
| | - Xiaoju He
- Department of Endocrinology and Metabolism of the Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, 330006, Jiangxi, China
- Department of Obstetrics and Gynecology of the Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, 330006, Jiangxi, China
| | - Jianping Liu
- Department of Endocrinology and Metabolism of the Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, 330006, Jiangxi, China
- Institute for the Study of Endocrinology and Metabolism in Jiangxi Province, Nanchang, 330006, Jiangxi, China
| |
Collapse
|
8
|
Wu C, Zhu H, Zhang Y, Ding L, Wang J. Interference with mitochondrial metabolism could serve as a potential therapeutic strategy for advanced prostate cancer. PLoS One 2024; 19:e0290753. [PMID: 38598542 PMCID: PMC11006138 DOI: 10.1371/journal.pone.0290753] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2023] [Accepted: 08/15/2023] [Indexed: 04/12/2024] Open
Abstract
Metabolic reprogramming has been defined as a hallmark of malignancies. Prior studies have focused on the single nucleotide polymorphism (SNP) of POLG2 gene, which is reportedly responsible for encoding mitochondrial DNA genes and is implicated in the material and energy metabolism of tumor cells, whereas its function in prostate cancer has been elusive. Gene expression profile matrix and clinical information were downloaded from TCGA (The Cancer Genome Atlas) data portal, and GSE3325 and GSE8511 were retrieved from GEO (Gene Expression Omnibus) database. We conducted analysis of the relative expression of POLG2, clinical characterization, survival analysis, GO / KEGG and GSEA (Gene Set Enrichment Analysis) enrichment analysis in R and employed STRING portal to acquaint ourselves with the protein-protein interaction (PPI). IHC (Immunohistochemical) profiles of POLG2 protein between normal and cancerous tissues were consulted via HPA (Human protein atlas) database and the immunohistochemical POLG2 were verified between para-cancerous and cancerous tissues in tissue array. At the cellular level, Mitochondrial dysfunction assay, DNA synthesis test, wound healing assay, and invasion assay were implemented to further validate the phenotype of POLG2 knockdown in PCa cell lines. RT-qPCR and western blotting were routinely adopted to verify variations of molecular expression within epithelial mesenchymal transition (EMT). Results showed that POLG2 was over-expressed in most cancer types, and the over-expression of POLG2 was correlated with PCa progression and suggested poor OS (Overall Survival) and PFI (Progress Free Interval). Multivariate analysis showed that POLG2 might be an independent prognostic factor of prostate cancer. We also performed GO/KEGG, GSEA analysis, co-expression genes, and PPI, and observed the metabolism-related gene alterations in PCa. Furthermore, we verified that POLG2 knockdown had an inhibitory effect on mitochondrial function, proliferation, cell motility, and invasion, we affirmed POLG2 could affect the prognosis of advanced prostate cancer via EMT. In summary, our findings indicate that over-expressed POLG2 renders poor prognosis in advanced prostate cancer. This disadvantageous factor can serve as a potential indicator, making it possible to target mitochondrial metabolism to treat advanced prostate cancer.
Collapse
Affiliation(s)
- Chuang Wu
- Department of Urology, Jiangsu Province Geriatric Hospital, Nanjing, Jiangsu, China
| | - Huihuang Zhu
- Department of Urology, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, Jiangsu, China
| | - Yang Zhang
- Department of Urology, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, Jiangsu, China
| | - Li Ding
- Department of Urology, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, Jiangsu, China
| | - Junqi Wang
- Department of Urology, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, Jiangsu, China
| |
Collapse
|
9
|
Jiang C, Li P, Ma Y, Yoneda N, Kawai K, Uehara S, Ohnishi Y, Suemizu H, Cao H. Comprehensive gene profiling of the metabolic landscape of humanized livers in mice. J Hepatol 2024; 80:622-633. [PMID: 38049085 PMCID: PMC10947884 DOI: 10.1016/j.jhep.2023.11.020] [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] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/05/2023] [Revised: 10/12/2023] [Accepted: 11/06/2023] [Indexed: 12/06/2023]
Abstract
BACKGROUND & AIMS The human liver transcriptome is complex and highly dynamic, e.g. one gene may produce multiple distinct transcripts, each with distinct posttranscriptional modifications. Direct knowledge of transcriptome dynamics, however, is largely obscured by the inaccessibility of the human liver to treatments and the insufficient annotation of the human liver transcriptome at transcript and RNA modification levels. METHODS We generated mice that carry humanized livers of identical genetic background and subjected them to representative metabolic treatments. We then analyzed the humanized livers with nanopore single-molecule direct RNA sequencing to determine the expression level, m6A modification and poly(A) tail length of all RNA transcript isoforms. Our system allows for the de novo annotation of human liver transcriptomes to reflect metabolic responses and for the study of transcriptome dynamics in parallel. RESULTS Our analysis uncovered a vast number of novel genes and transcripts. Our transcript-level analysis of human liver transcriptomes also identified a multitude of regulated metabolic pathways that were otherwise invisible using conventional short-read RNA sequencing. We revealed for the first time the dynamic changes in m6A and poly(A) tail length of human liver transcripts, many of which are transcribed from key metabolic genes. Furthermore, we performed comparative analyses of gene regulation between humans and mice, and between two individuals using the liver-specific humanized mice, revealing that transcriptome dynamics are highly species- and genetic background-dependent. CONCLUSION Our work revealed a complex metabolic response landscape of the human liver transcriptome and provides a novel resource to understand transcriptome dynamics of the human liver in response to physiologically relevant metabolic stimuli (https://caolab.shinyapps.io/human_hepatocyte_landscape/). IMPACT AND IMPLICATIONS Direct knowledge of the human liver transcriptome is currently very limited, hindering the overall understanding of human liver pathophysiology. We combined a liver-specific humanized mouse model and long-read direct RNA sequencing technology to establish a de novo annotation of the human liver transcriptome and identified a multitude of regulated metabolic pathways that were otherwise invisible using conventional technologies. The extensive regulatory information on human genes we provided could enable basic scientists to infer the pathological relevance of their genes of interest and physician scientists to better pinpoint the changes in metabolic networks underlying a specific pathophysiology.
Collapse
Affiliation(s)
- Chengfei Jiang
- Cardiovascular Branch, National Heart, Lung and Blood Institute, National Institutes of Health, Bethesda, MD 20892, USA
| | - Ping Li
- Cardiovascular Branch, National Heart, Lung and Blood Institute, National Institutes of Health, Bethesda, MD 20892, USA
| | - Yonghe Ma
- Cardiovascular Branch, National Heart, Lung and Blood Institute, National Institutes of Health, Bethesda, MD 20892, USA
| | - Nao Yoneda
- Liver Engineering Laboratory, Department of Applied Research for Laboratory Animals, Central Institute for Experimental Animals (CIEA), 3-25-12 Tonomachi, Kawasaki-ku, Kawasaki 210-0821, Japan
| | - Kenji Kawai
- Pathology Center, Translational Research and Contract Research Service Division, Central Institute for Experimental Animals (CIEA), 3-25-12 Tonomachi, Kawasaki-ku, Kawasaki 210-0821, Japan
| | - Shotaro Uehara
- Liver Engineering Laboratory, Department of Applied Research for Laboratory Animals, Central Institute for Experimental Animals (CIEA), 3-25-12 Tonomachi, Kawasaki-ku, Kawasaki 210-0821, Japan
| | - Yasuyuki Ohnishi
- Liver Engineering Laboratory, Department of Applied Research for Laboratory Animals, Central Institute for Experimental Animals (CIEA), 3-25-12 Tonomachi, Kawasaki-ku, Kawasaki 210-0821, Japan
| | - Hiroshi Suemizu
- Liver Engineering Laboratory, Department of Applied Research for Laboratory Animals, Central Institute for Experimental Animals (CIEA), 3-25-12 Tonomachi, Kawasaki-ku, Kawasaki 210-0821, Japan
| | - Haiming Cao
- Cardiovascular Branch, National Heart, Lung and Blood Institute, National Institutes of Health, Bethesda, MD 20892, USA.
| |
Collapse
|
10
|
Schneider H, Krizanac AM, Falker-Gieske C, Heise J, Tetens J, Thaller G, Bennewitz J. Genomic dissection of the correlation between milk yield and various health traits using functional and evolutionary information about imputed sequence variants of 34,497 German Holstein cows. BMC Genomics 2024; 25:265. [PMID: 38461236 PMCID: PMC11385139 DOI: 10.1186/s12864-024-10115-6] [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: 08/17/2023] [Accepted: 02/13/2024] [Indexed: 03/11/2024] Open
Abstract
BACKGROUND Over the last decades, it was subject of many studies to investigate the genomic connection of milk production and health traits in dairy cattle. Thereby, incorporating functional information in genomic analyses has been shown to improve the understanding of biological and molecular mechanisms shaping complex traits and the accuracies of genomic prediction, especially in small populations and across-breed settings. Still, little is known about the contribution of different functional and evolutionary genome partitioning subsets to milk production and dairy health. Thus, we performed a uni- and a bivariate analysis of milk yield (MY) and eight health traits using a set of ~34,497 German Holstein cows with 50K chip genotypes and ~17 million imputed sequence variants divided into 27 subsets depending on their functional and evolutionary annotation. In the bivariate analysis, eight trait-combinations were observed that contrasted MY with each health trait. Two genomic relationship matrices (GRM) were included, one consisting of the 50K chip variants and one consisting of each set of subset variants, to obtain subset heritabilities and genetic correlations. In addition, 50K chip heritabilities and genetic correlations were estimated applying merely the 50K GRM. RESULTS In general, 50K chip heritabilities were larger than the subset heritabilities. The largest heritabilities were found for MY, which was 0.4358 for the 50K and 0.2757 for the subset heritabilities. Whereas all 50K genetic correlations were negative, subset genetic correlations were both, positive and negative (ranging from -0.9324 between MY and mastitis to 0.6662 between MY and digital dermatitis). The subsets containing variants which were annotated as noncoding related, splice sites, untranslated regions, metabolic quantitative trait loci, and young variants ranked highest in terms of their contribution to the traits` genetic variance. We were able to show that linkage disequilibrium between subset variants and adjacent variants did not cause these subsets` high effect. CONCLUSION Our results confirm the connection of milk production and health traits in dairy cattle via the animals` metabolic state. In addition, they highlight the potential of including functional information in genomic analyses, which helps to dissect the extent and direction of the observed traits` connection in more detail.
Collapse
Affiliation(s)
- Helen Schneider
- Institute of Animal Science, University of Hohenheim, 70599, Stuttgart, Germany.
| | - Ana-Marija Krizanac
- Department of Animal Sciences, University of Göttingen, 37077, Göttingen, Germany
| | | | - Johannes Heise
- Vereinigte Informationssysteme Tierhaltung w.V. (VIT), 27283, Verden, Germany
| | - Jens Tetens
- Department of Animal Sciences, University of Göttingen, 37077, Göttingen, Germany
| | - Georg Thaller
- Institute of Animal Breeding and Husbandry, Christian-Albrechts University of Kiel, 24098, Kiel, Germany
| | - Jörn Bennewitz
- Institute of Animal Science, University of Hohenheim, 70599, Stuttgart, Germany
| |
Collapse
|
11
|
Zailaie SA, Khoja BB, Siddiqui JJ, Mawardi MH, Heaphy E, Aljagthmi A, Sergi CM. Investigating the Role of Non-Coding RNA in Non-Alcoholic Fatty Liver Disease. Noncoding RNA 2024; 10:10. [PMID: 38392965 PMCID: PMC10891858 DOI: 10.3390/ncrna10010010] [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: 12/12/2023] [Revised: 01/22/2024] [Accepted: 01/27/2024] [Indexed: 02/25/2024] Open
Abstract
Non-coding RNAs (ncRNAs) are RNA molecules that do not code for protein but play key roles in regulating cellular processes. NcRNAs globally affect gene expression in diverse physiological and pathological contexts. Functionally important ncRNAs act in chromatin modifications, in mRNA stabilization and translation, and in regulation of various signaling pathways. Non-alcoholic fatty liver disease (NAFLD) is a set of conditions caused by the accumulation of triacylglycerol in the liver. Studies of ncRNA in NAFLD are limited but have demonstrated that ncRNAs play a critical role in the pathogenesis of NAFLD. In this review, we summarize NAFLD's pathogenesis and clinical features, discuss current treatment options, and review the involvement of ncRNAs as regulatory molecules in NAFLD and its progression to non-alcoholic steatohepatitis (NASH). In addition, we highlight signaling pathways dysregulated in NAFLD and review their crosstalk with ncRNAs. Having a thorough understanding of the disease process's molecular mechanisms will facilitate development of highly effective diagnostic and therapeutic treatments. Such insights can also inform preventive strategies to minimize the disease's future development.
Collapse
Affiliation(s)
- Samar A. Zailaie
- Research Center, King Faisal Specialist Hospital & Research Center-Jeddah (KFSHRC-J), Jeddah 21499, Saudi Arabia; (S.A.Z.); (B.B.K.); (E.H.); (A.A.)
| | - Basmah B. Khoja
- Research Center, King Faisal Specialist Hospital & Research Center-Jeddah (KFSHRC-J), Jeddah 21499, Saudi Arabia; (S.A.Z.); (B.B.K.); (E.H.); (A.A.)
| | - Jumana J. Siddiqui
- Biochemistry Department, Faculty of Science, King Abdulaziz University, Jeddah 21589, Saudi Arabia;
| | - Mohammad H. Mawardi
- Medicine Department, Gastroenterology Section, King Faisal Specialist Hospital & Research Center-Jeddah (KFSHRC-J), Jeddah 21499, Saudi Arabia;
| | - Emily Heaphy
- Research Center, King Faisal Specialist Hospital & Research Center-Jeddah (KFSHRC-J), Jeddah 21499, Saudi Arabia; (S.A.Z.); (B.B.K.); (E.H.); (A.A.)
| | - Amjad Aljagthmi
- Research Center, King Faisal Specialist Hospital & Research Center-Jeddah (KFSHRC-J), Jeddah 21499, Saudi Arabia; (S.A.Z.); (B.B.K.); (E.H.); (A.A.)
| | - Consolato M. Sergi
- Children’s Hospital of Eastern Ontario (CHEO), University of Ottawa, Ottawa, ON K1H 8L1, Canada
- Department of Laboratory Medicine and Pathology, University of Alberta, Edmonton, AB T6G 2R3, Canada
| |
Collapse
|
12
|
Liu C, Liu X, Li H, Kang Z. Advances in the regulation of adipogenesis and lipid metabolism by exosomal ncRNAs and their role in related metabolic diseases. Front Cell Dev Biol 2023; 11:1173904. [PMID: 37791070 PMCID: PMC10543472 DOI: 10.3389/fcell.2023.1173904] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2023] [Accepted: 08/15/2023] [Indexed: 10/05/2023] Open
Abstract
Exosomes are membrane-bound extracellular vesicles released following the fusion of multivesicular bodies (MVBs) with the cell membrane. Exosomes transport diverse molecules, including proteins, lipids, DNA and RNA, and regulate distant intercellular communication. Noncoding RNA (ncRNAs) carried by exosomes regulate cell-cell communication in tissues, including adipose tissue. This review summarizes the action mechanisms of ncRNAs carried by exosomes on adipocyte differentiation and modulation of adipogenesis by exosomal ncRNAs. This study aims to provide valuable insights for developing novel therapeutics.
Collapse
Affiliation(s)
- Cong Liu
- Department of Breast Surgery, China-Japan Union Hospital of Jilin University, Changchun, China
| | - Xilin Liu
- Department of Hand and Foot Surgery, China-Japan Union Hospital of Jilin University, Changchun, China
| | - Hong Li
- Department of Nursing, China-Japan Union Hospital of Jilin University, Changchun, China
| | - Zhichen Kang
- Department of Rehabilitation, The Second Hospital of Jilin University, Changchun, China
| |
Collapse
|
13
|
Zong Y, Wang X, Cui B, Xiong X, Wu A, Lin C, Zhang Y. Decoding the regulatory roles of non-coding RNAs in cellular metabolism and disease. Mol Ther 2023; 31:1562-1576. [PMID: 37113055 PMCID: PMC10277898 DOI: 10.1016/j.ymthe.2023.04.012] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2023] [Revised: 04/12/2023] [Accepted: 04/21/2023] [Indexed: 04/29/2023] Open
Abstract
Non-coding RNAs, including long non-coding RNAs (lncRNAs), microRNAs (miRNAs), and circular RNAs (circRNAs), are being studied extensively in a variety of fields. Their roles in metabolism have received increasing attention in recent years but are not yet clear. The regulation of glucose, fatty acid, and amino acid metabolism is an imperative physiological process that occurs in living organisms and takes part in cancer and cardiovascular diseases. Here, we summarize the important roles played by non-coding RNAs in glucose metabolism, fatty acid metabolism, and amino acid metabolism, as well as the mechanisms involved. We also summarize the therapeutic advances for non-coding RNAs in diseases such as obesity, cardiovascular disease, and some metabolic diseases. Overall, non-coding RNAs are indispensable factors in metabolism and have a significant role in the three major metabolisms, which may be exploited as therapeutic targets in the future.
Collapse
Affiliation(s)
- Yuru Zong
- Beijing Institute of Brain Disorders, Laboratory of Brain Disorders, Ministry of Science and Technology, Collaborative Innovation Center for Brain Disorders, Capital Medical University, Beijing 100069, China
| | - Xuliang Wang
- Department of Cardiology and Institute of Vascular Medicine, Peking University Third Hospital, Key Laboratory of Molecular Cardiovascular Science, Ministry of Education, Beijing 100191, China
| | - Bing Cui
- Beijing Institute of Brain Disorders, Laboratory of Brain Disorders, Ministry of Science and Technology, Collaborative Innovation Center for Brain Disorders, Capital Medical University, Beijing 100069, China
| | - Xiaowei Xiong
- Department of Cardiology and Macrovascular Disease, Beijing Tiantan Hospital, Capital Medical University, Beijing 100050, China
| | - Andrew Wu
- Department of Molecular and Cellular Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Chunru Lin
- Department of Molecular and Cellular Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA; The Graduate School of Biomedical Sciences, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA.
| | - Yaohua Zhang
- Beijing Institute of Brain Disorders, Laboratory of Brain Disorders, Ministry of Science and Technology, Collaborative Innovation Center for Brain Disorders, Capital Medical University, Beijing 100069, China.
| |
Collapse
|
14
|
Shi N, Sun K, Tang H, Mao J. The impact and role of identified long noncoding RNAs in nonalcoholic fatty liver disease: A narrative review. J Clin Lab Anal 2023; 37:e24943. [PMID: 37435630 PMCID: PMC10431402 DOI: 10.1002/jcla.24943] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2023] [Revised: 06/07/2023] [Accepted: 07/02/2023] [Indexed: 07/13/2023] Open
Abstract
BACKGROUND Nonalcoholic fatty liver disease (NAFLD) is the most common liver disease worldwide, but its mechanism and pathophysiology remain unclear. Long noncoding RNAs (lncRNAs) may exert a vital influence on regulating various biological functions in NAFLD. METHODS The databases such as Google Scholar, PubMed, and Medline were searched using the following keywords: nonalcoholic fatty liver disease, nonalcoholic fatty liver disease, NAFLD, nonalcoholic steatohepatitis, nonalcoholic steatohepatitis, NASH, long noncoding RNAs, and lncRNAs. Considering the titles and abstracts, unrelated studies were excluded. The authors evaluated the full texts of the remaining studies. RESULTS We summarized the current knowledge of lncRNAs and the main signaling pathways of lncRNAs involved in NAFLD explored in recent years. As a heterogeneous group of noncoding RNAs (ncRNAs), lncRNAs play crucial roles in biological processes underlying the pathophysiology of NAFLD. The mechanisms, particularly those associated with the regulation of the expression and activities of lncRNAs, play important roles in NAFLD. CONCLUSION A better comprehension of the mechanism controlled by lncRNAs in NAFLD is necessary for the identification of novel therapeutic targets for drug development and improved, noninvasive methods for diagnosis.
Collapse
Affiliation(s)
- Na Shi
- Department of GastroenterologyFirst Affiliated Hospital of Dalian Medical UniversityDalianChina
- Department of Internal MedicineThe Third People's Hospital of ChengduChengduChina
| | - Kang Sun
- Department of GastroenterologyFirst Affiliated Hospital of Dalian Medical UniversityDalianChina
| | - Haiying Tang
- Department of Respiratory and Critical Care MedicineFirst Affiliated Hospital of Dalian Medical UniversityDalianChina
| | - Jingwei Mao
- Department of GastroenterologyFirst Affiliated Hospital of Dalian Medical UniversityDalianChina
| |
Collapse
|
15
|
Sufianov A, Beilerli A, Kudriashov V, Ilyasova T, Liang Y, Mukhamedzyanov A, Bessonova M, Mashkin A, Beylerli O. The role of long non-coding RNAs in the development of adipose cells. Noncoding RNA Res 2023; 8:255-262. [PMID: 36890808 PMCID: PMC9988400 DOI: 10.1016/j.ncrna.2023.02.009] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2023] [Revised: 02/27/2023] [Accepted: 02/27/2023] [Indexed: 03/06/2023] Open
Abstract
In recent times, the rising prevalence of obesity and its associated comorbidities have had a severe impact on human health and social progress. Therefore, scientists are delving deeper into the pathogenesis of obesity, exploring the role of non-coding RNAs. Long non-coding RNAs (lncRNAs), once regarded as mere "noise" during genome transcription, have now been confirmed through numerous studies to regulate gene expression and contribute to the occurrence and progression of several human diseases. LncRNAs can interact with protein, DNA, and RNA, respectively, and participate in regulating gene expression by modulating the levels of visible modification, transcription, post-transcription, and biological environment. Increasingly, researchers have established the involvement of lncRNAs in regulating adipogenesis, development, and energy metabolism of adipose tissue (white and brown fat). In this article, we present a literature review of the role of lncRNAs in the development of adipose cells.
Collapse
Affiliation(s)
- Albert Sufianov
- Educational and Scientific Institute of Neurosurgery, Рeoples’ Friendship University of Russia (RUDN University), Moscow, Russia
- Department of Neurosurgery, Sechenov First Moscow State Medical University (Sechenov University), Moscow, Russia
| | - Aferin Beilerli
- Department of Obstetrics and Gynecology, Tyumen State Medical University, 54 Odesskaya Street, 625023, Tyumen, Russia
| | | | - Tatiana Ilyasova
- Department of Internal Diseases, Bashkir State Medical University, Ufa, Republic of Bashkortostan, 450008, Russia
| | - Yanchao Liang
- Department of Neurosurgery, The First Affiliated Hospital of Harbin Medical University, Harbin, 150001, China
| | | | - Marina Bessonova
- Tyumen Cardiology Research Center, Tomsk National Research Medical Center, Russian Academy of Science, Tomsk, Russia
| | - Andrey Mashkin
- Educational and Scientific Institute of Neurosurgery, Рeoples’ Friendship University of Russia (RUDN University), Moscow, Russia
| | - Ozal Beylerli
- Educational and Scientific Institute of Neurosurgery, Рeoples’ Friendship University of Russia (RUDN University), Moscow, Russia
- Corresponding author. Рeoples’ Friendship University of Russia (RUDN University), 6 Miklukho-Maklaya Street, Moscow, 117198, Russian Federation.
| |
Collapse
|
16
|
Cao L, Qu N, Wang X, Chen L, Liu M. The function of long non-coding RNA in non-alcoholic fatty liver disease. Clin Res Hepatol Gastroenterol 2023; 47:102095. [PMID: 36781069 DOI: 10.1016/j.clinre.2023.102095] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/07/2022] [Revised: 01/24/2023] [Accepted: 02/09/2023] [Indexed: 02/15/2023]
Abstract
Non-alcoholic fatty liver disease is a disease that is currently prevalent in the world, increasingly becoming the mainstay of liver diseases. And its prevalence is rapidly increasing, but its pathogenesis is not entirely understood. Long non-coding RNAs have increasingly gained attention as science has progressed in recent years. Studies have shown that long non-coding RNAs are involved in a variety of biological processes in vivo, such as proliferation, differentiation, and apoptosis, and can affect disease by regulating gene expression. This review explores the biological processes involving long non-coding RNAs, including lipid metabolism, glucose metabolism, liver fibrosis, and apoptosis. In addition, we summarize how the different long non-coding RNAs involved in each process function. Finally, the shortcomings of long non-coding RNAs as potential therapeutic targets are briefly described. In conclusion, this article provides a clear visualization of the link that exists between long non-coding RNAs and non-alcoholic fatty liver disease.
Collapse
Affiliation(s)
- Lianrui Cao
- School of Pharmaceutical Sciences, Liaoning University, No.66, Chongshan Mid Road, Shenyang 110036, China
| | - Na Qu
- School of Pharmaceutical Sciences, Liaoning University, No.66, Chongshan Mid Road, Shenyang 110036, China
| | - Xin Wang
- School of Pharmaceutical Sciences, Liaoning University, No.66, Chongshan Mid Road, Shenyang 110036, China
| | - Lijiang Chen
- School of Pharmaceutical Sciences, Liaoning University, No.66, Chongshan Mid Road, Shenyang 110036, China.
| | - Mingxia Liu
- School of Pharmaceutical Sciences, Liaoning University, No.66, Chongshan Mid Road, Shenyang 110036, China.
| |
Collapse
|
17
|
Kudriashov V, Sufianov A, Mashkin A, Beilerli A, Ilyasova T, Liang Y, Lyulin S, Beylerli O. The role of long non-coding RNAs in carbohydrate and fat metabolism in the liver. Noncoding RNA Res 2023; 8:294-301. [PMID: 36970373 PMCID: PMC10031277 DOI: 10.1016/j.ncrna.2023.03.003] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2023] [Revised: 02/20/2023] [Accepted: 03/13/2023] [Indexed: 03/17/2023] Open
Abstract
The metabolism of carbohydrates and lipids (fat) in the liver is closely interconnected both in physiological conditions and in pathology. This relationship in the body is possible due to the regulation by many factors, including epigenetic ones. Histone modifications, DNA methylation, and non-coding RNAs are considered to be the main epigenetic factors. Non-coding RNAs (ncRNAs) refers to ribonucleic acid (RNA) molecules that do not code for a protein. They cover a huge number of RNA classes and perform a wide range of biological functions such as regulating gene expression, protecting the genome from exogenous DNA, and directing DNA synthesis. One such class of ncRNAs that has been extensively studied are long non-coding RNAs (lncRNAs). The important role of lncRNAs in the formation and maintenance of normal homeostasis of biological systems, as well as participation in many pathological processes, has been proven. The results of recent studies indicate the importance of lncRNAs in lipid and carbohydrate metabolism. Modifications of lncRNAs expression can lead to disruption of biological processes in tissues, including fat and protein, such as adipocyte proliferation and differentiation, inflammation, and insulin resistance. Further study of lncRNAs made it possible to partly determine the regulatory mechanisms underlying the formation of an imbalance in carbohydrate and fat metabolism individually and in their relationship, and the degree of interaction between different types of cells involved in this process. This review will focus on the function of lncRNAs and its relation to hepatic carbohydrate and fat metabolism and related diseases in order to elucidate the underlying mechanisms and prospects for studies with lncRNAs.
Collapse
|
18
|
Hong Y, Zhang Y, Chen H, Tang X, Zhao H, Meng Z, Jia X, Liu W, Li X, Wang L, Zhong X, Bai X, Sun H, Kopylov P, Afina B, Shchekochikhin D, Zhang Y, Liu X, Fan Y. Genetic dissection of the impact of lncRNA AI662270 during the development of atherosclerosis. J Transl Med 2023; 21:97. [PMID: 36755320 PMCID: PMC9906833 DOI: 10.1186/s12967-023-03962-6] [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: 05/12/2022] [Accepted: 02/02/2023] [Indexed: 02/10/2023] Open
Abstract
BACKGROUND Atherosclerosis is driven by synergistic interactions between pathological biomechanical and lipid metabolic factors. Long noncoding RNAs (LncRNAs) have been implicated in atherogenesis. The purpose of this study was to investigate the potential mechanism of lncRNA AI662270 on macrophage cholesterol transport in atherosclerosis. METHODS Apolipoprotein E deficiency (ApoE-/-) mice were fed a high fat diet for 16 weeks to construct atherosclerotic model, and the mice were injected with recombinant lentivirus carrying AI662270 gene to overexpress AI662270. Macrophages were cleared by liposomal clondronate in vivo. Fundamental experiments and functional assays, hematoxylin and eosin staining, oil red O staining and others, were performed to evaluate the function of AI662270 on atherogenesis. Peritoneal macrophages were treated with oxidized low density lipoprotein (ox-LDL) to simulate in vitro model. Mechanism assays, RNA-interacting protein immunoprecipitation, RNA-protein pulldown and others, were performed to study the regulatory mechanism of AI662270 in macrophages. RESULTS The novel AI662270 was mainly enriched in macrophages, but not in endothelial cells, smooth muscle cells and fibroblasts of mouse atherosclerotic lesions and was upregulated by ox-LDL. Overexpression of AI662270 resulted in lipid accumulation, larger atherosclerotic plaques and cardiac dysfunction in vivo. After macrophages were removed, the pro-atherogenic effect of AI662270 disappeared. Downregulation of AI662270 in macrophages protected against foam cell formation by potentiating cholesterol efflux and reducing intracellular total cholesterol. The opposite effect was observed in macrophage-specific AI662270-overexpressed cells in vitro. AI662270 bound to adenosine triphosphate-binding cassette transporter A1 (Abca1) responsible for regulating cholesterol efflux in macrophages. Forced expression of AI662270 in macrophages decreased Abca1 expression. The reverse occurred when expression of AI662270 was repressed. CONCLUSION These findings reveal an essential role for AI662270 in atherosclerosis progression by regulating cholesterol efflux from macrophages.
Collapse
Affiliation(s)
- Yang Hong
- grid.410736.70000 0001 2204 9268Department of Pharmacology, State-Province Key Laboratories of Biomedicine-Pharmaceutics of China, Key Laboratory of Cardiovascular Medicine Research, Ministry of Education, College of Pharmacy, Harbin Medical University, Harbin, 150081 Heilongjiang People’s Republic of China
| | - Yue Zhang
- grid.410736.70000 0001 2204 9268Department of Pharmacology, State-Province Key Laboratories of Biomedicine-Pharmaceutics of China, Key Laboratory of Cardiovascular Medicine Research, Ministry of Education, College of Pharmacy, Harbin Medical University, Harbin, 150081 Heilongjiang People’s Republic of China
| | - Hui Chen
- grid.410736.70000 0001 2204 9268Department of Pharmacology, State-Province Key Laboratories of Biomedicine-Pharmaceutics of China, Key Laboratory of Cardiovascular Medicine Research, Ministry of Education, College of Pharmacy, Harbin Medical University, Harbin, 150081 Heilongjiang People’s Republic of China
| | - Xueqing Tang
- grid.410736.70000 0001 2204 9268Department of Pharmacology, State-Province Key Laboratories of Biomedicine-Pharmaceutics of China, Key Laboratory of Cardiovascular Medicine Research, Ministry of Education, College of Pharmacy, Harbin Medical University, Harbin, 150081 Heilongjiang People’s Republic of China
| | - Hongrui Zhao
- grid.410736.70000 0001 2204 9268Department of Pharmacology, State-Province Key Laboratories of Biomedicine-Pharmaceutics of China, Key Laboratory of Cardiovascular Medicine Research, Ministry of Education, College of Pharmacy, Harbin Medical University, Harbin, 150081 Heilongjiang People’s Republic of China
| | - Ziyu Meng
- grid.410736.70000 0001 2204 9268Department of Pharmacology, State-Province Key Laboratories of Biomedicine-Pharmaceutics of China, Key Laboratory of Cardiovascular Medicine Research, Ministry of Education, College of Pharmacy, Harbin Medical University, Harbin, 150081 Heilongjiang People’s Republic of China
| | - Xueling Jia
- grid.410736.70000 0001 2204 9268Department of Pathology and Pathophysiology, College of Basic Medical Sciences, Harbin Medical University-Daqing, Daqing, 163319 Heilongjiang People’s Republic of China
| | - Wenfeng Liu
- grid.410736.70000 0001 2204 9268Department of Pathology and Pathophysiology, College of Basic Medical Sciences, Harbin Medical University-Daqing, Daqing, 163319 Heilongjiang People’s Republic of China
| | - Xiaohan Li
- grid.410736.70000 0001 2204 9268Department of Pharmacology, State-Province Key Laboratories of Biomedicine-Pharmaceutics of China, Key Laboratory of Cardiovascular Medicine Research, Ministry of Education, College of Pharmacy, Harbin Medical University, Harbin, 150081 Heilongjiang People’s Republic of China
| | - Lin Wang
- grid.410736.70000 0001 2204 9268Department of Pathology and Pathophysiology, College of Basic Medical Sciences, Harbin Medical University-Daqing, Daqing, 163319 Heilongjiang People’s Republic of China
| | - Xinrui Zhong
- grid.410736.70000 0001 2204 9268Department of Pathology and Pathophysiology, College of Basic Medical Sciences, Harbin Medical University-Daqing, Daqing, 163319 Heilongjiang People’s Republic of China
| | - Xuefeng Bai
- grid.410736.70000 0001 2204 9268School of Medical Informatics, Harbin Medical University, Daqing Campus, Daqing, 163319 People’s Republic of China
| | - Heyang Sun
- grid.410736.70000 0001 2204 9268Department of Pharmacology, State-Province Key Laboratories of Biomedicine-Pharmaceutics of China, Key Laboratory of Cardiovascular Medicine Research, Ministry of Education, College of Pharmacy, Harbin Medical University, Harbin, 150081 Heilongjiang People’s Republic of China
| | - Philipp Kopylov
- grid.410736.70000 0001 2204 9268Department of Pharmacology, State-Province Key Laboratories of Biomedicine-Pharmaceutics of China, Key Laboratory of Cardiovascular Medicine Research, Ministry of Education, College of Pharmacy, Harbin Medical University, Harbin, 150081 Heilongjiang People’s Republic of China ,grid.448878.f0000 0001 2288 8774Department of Preventive and Emergency Cardiology, Sechenov First Moscow State Medical University, Moscow, Russian Federation
| | - Bestavashvili Afina
- grid.410736.70000 0001 2204 9268Department of Pharmacology, State-Province Key Laboratories of Biomedicine-Pharmaceutics of China, Key Laboratory of Cardiovascular Medicine Research, Ministry of Education, College of Pharmacy, Harbin Medical University, Harbin, 150081 Heilongjiang People’s Republic of China ,grid.448878.f0000 0001 2288 8774Department of Cardiology, Functional and Ultrasound Diagnostics, N.V. Sklifosofsky, I. M. Sechenov First Moscow State Medical University, Moscow, Russian Federation
| | - Dmitry Shchekochikhin
- grid.410736.70000 0001 2204 9268Department of Pharmacology, State-Province Key Laboratories of Biomedicine-Pharmaceutics of China, Key Laboratory of Cardiovascular Medicine Research, Ministry of Education, College of Pharmacy, Harbin Medical University, Harbin, 150081 Heilongjiang People’s Republic of China ,grid.448878.f0000 0001 2288 8774Department of Preventive and Emergency Cardiology, Sechenov First Moscow State Medical University, Moscow, Russian Federation
| | - Yong Zhang
- grid.410736.70000 0001 2204 9268Department of Pharmacology, State-Province Key Laboratories of Biomedicine-Pharmaceutics of China, Key Laboratory of Cardiovascular Medicine Research, Ministry of Education, College of Pharmacy, Harbin Medical University, Harbin, 150081 Heilongjiang People’s Republic of China
| | - Xin Liu
- Department of Pharmacology, State-Province Key Laboratories of Biomedicine-Pharmaceutics of China, Key Laboratory of Cardiovascular Medicine Research, Ministry of Education, College of Pharmacy, Harbin Medical University, Harbin, 150081, Heilongjiang, People's Republic of China.
| | - Yuhua Fan
- Department of Pathology and Pathophysiology, College of Basic Medical Sciences, Harbin Medical University-Daqing, Daqing, 163319, Heilongjiang, People's Republic of China. .,Department of Pharmacology, State-Province Key Laboratories of Biomedicine-Pharmaceutics of China, Key Laboratory of Cardiovascular Medicine Research, Ministry of Education, College of Pharmacy, Harbin Medical University, Harbin, 150081, Heilongjiang, People's Republic of China.
| |
Collapse
|
19
|
lncRNA CRNDE Affects Th17/IL-17A and Inhibits Epithelial-Mesenchymal Transition in Lung Epithelial Cells Reducing Asthma Signs. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2023; 2023:2092184. [PMID: 36743692 PMCID: PMC9897922 DOI: 10.1155/2023/2092184] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 08/09/2022] [Revised: 12/26/2022] [Accepted: 12/30/2022] [Indexed: 01/28/2023]
Abstract
Background Asthma treatment is difficult due to disease heterogeneity and comorbidities. In addition, the development of drugs targeting the underlying mechanisms of asthma remains slow. We planned to identify the most upregulated differentially expressed long noncoding RNA in asthma to explore its regulatory patterns and pathways in asthma. Methods We sensitized mice using a mixture of ovalbumin, house dust mites, and lipopolysaccharide to establish an asthma mouse model. We also sensitized asthma cells with TGF-β1 in an in vitro model. We performed a microarray analysis to identify the lncRNA with the differential expression level in model mice. We applied hematoxylin and eosin and Masson's trichrome stainings to mouse tissues to quantify the tissue damage extent. Next, we assess the levels of lncRNA CRNDE, miR-29a-3p, TGF-β1, MCL-1, E-cadherin, vimentin, and snail. We counted the percentages of Th17 cells using flow cytometry. Finally, we performed a dual-luciferase reporter assay to assess the association between lncRNA CRNDE and miR-29a-3p. Results We successfully established asthma mouse/cell models and selected the lncRNA CRNDE for our study. Transfection of si-CRNDE reduced the degree of injury and inflammation in the mouse model and reversed the TGF-β1-induced epithelial-mesenchymal transition (EMT) in the cell model. Moreover, the E-cadherin level was upregulated, and the levels of IL-17A, vimentin, snail, and α-SMA were downregulated. We also discovered that lncRNA CRNDE negatively regulated miR-29a-3p and that this one in turn inhibited MCL-1 in mice. After lncRNA CRNDE expression downregulation, the level of miR-29a-3p was increased, and we detected reduced levels of MCL-1 and EMTs. Conclusions lncRNA CRNDE expression downregulation led to reduced inflammation and reduced lung damage in mice with induced asthma, it inhibited the EMTs of lung epithelial cells via the miR-29a-3p/MCL-1 pathway, and it reduced the levels of Th17/IL-17A cells to reduce asthma signs.
Collapse
|
20
|
Wang A, Wang J, Mao M, Zhao X, Li Q, Xuan R, Li F, Chao T. Analyses of lncRNAs, circRNAs, and the Interactions between ncRNAs and mRNAs in Goat Submandibular Glands Reveal Their Potential Function in Immune Regulation. Genes (Basel) 2023; 14:187. [PMID: 36672927 PMCID: PMC9859278 DOI: 10.3390/genes14010187] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2022] [Revised: 01/01/2023] [Accepted: 01/06/2023] [Indexed: 01/13/2023] Open
Abstract
As part of one of the main ruminants, goat salivary glands hardly secrete digestive enzymes, but play an important role in immunity. The immune function of goat salivary glands significantly changes with age, while the expression profile and specific function of non-coding RNA during this process are unknown. In this study, transcriptome sequencing was performed on submandibular gland (SMG) tissues of 1-month-old, 12-month-old, and 24-month-old goats, revealing the expression patterns of lncRNA and circRNA at different ages. A total of 369 lncRNAs and 1699 circRNAs were found to be differentially expressed. Functional enrichment analyses showed that the lncRNA regulated target mRNAs and circRNA host genes were significantly enriched in immune-related GO terms and pathways. CeRNA network analysis showed that the key differentially expressed circRNAs and lncRNAs mainly regulate the key immune-related genes ITGB2, LCP2, PTPRC, SYK, and ZAP70 through competitive binding with miR-141-x, miR-29-y, and chi-miR-29b-3p, thereby affecting the natural killer cell-mediated cytotoxicity pathway, the T cell receptor signaling pathway, and other immune-related pathways. It should be noted that the expression of key circRNAs, lncRNAs, and key immune-related genes in goat SMGs decreased significantly with the growth of the goat. This is the first reporting of lncRNAs, circRNAs, and ceRNA network regulation in goat SMGs. Our study contributes to the knowledge of changes in the expression of non-coding RNAs during SMG development in goats and provides new insights into the relationship between non-coding RNAs and salivary gland immune function in goats.
Collapse
Affiliation(s)
- Aili Wang
- Shandong Provincial Key Laboratory of Animal Biotechnology and Disease Control and Prevention, College of Animal Science and Veterinary Medicine, Shandong Agricultural University, Taian 271000, China
| | - Jianmin Wang
- Shandong Provincial Key Laboratory of Animal Biotechnology and Disease Control and Prevention, College of Animal Science and Veterinary Medicine, Shandong Agricultural University, Taian 271000, China
- Key Laboratory of Efficient Utilization of Non-Grain Feed Resources (Co-Construction by Ministry and Province), Ministry of Agriculture and Rural Affairs, Shandong Agricultural University, Taian 271000, China
| | - Meina Mao
- Shandong Peninsula Engineering Research Center of Comprehensive Brine Utilization, Weifang University of Science and Technology, Shouguang 262700, China
| | - Xiaodong Zhao
- Shandong Provincial Key Laboratory of Animal Biotechnology and Disease Control and Prevention, College of Animal Science and Veterinary Medicine, Shandong Agricultural University, Taian 271000, China
- Shandong Vocational Animal Science and Veterinary College, Weifang 261000, China
| | - Qing Li
- Shandong Provincial Key Laboratory of Animal Biotechnology and Disease Control and Prevention, College of Animal Science and Veterinary Medicine, Shandong Agricultural University, Taian 271000, China
- Key Laboratory of Efficient Utilization of Non-Grain Feed Resources (Co-Construction by Ministry and Province), Ministry of Agriculture and Rural Affairs, Shandong Agricultural University, Taian 271000, China
| | - Rong Xuan
- Shandong Provincial Key Laboratory of Animal Biotechnology and Disease Control and Prevention, College of Animal Science and Veterinary Medicine, Shandong Agricultural University, Taian 271000, China
- Key Laboratory of Efficient Utilization of Non-Grain Feed Resources (Co-Construction by Ministry and Province), Ministry of Agriculture and Rural Affairs, Shandong Agricultural University, Taian 271000, China
| | - Fajun Li
- Shandong Peninsula Engineering Research Center of Comprehensive Brine Utilization, Weifang University of Science and Technology, Shouguang 262700, China
| | - Tianle Chao
- Shandong Provincial Key Laboratory of Animal Biotechnology and Disease Control and Prevention, College of Animal Science and Veterinary Medicine, Shandong Agricultural University, Taian 271000, China
- Key Laboratory of Efficient Utilization of Non-Grain Feed Resources (Co-Construction by Ministry and Province), Ministry of Agriculture and Rural Affairs, Shandong Agricultural University, Taian 271000, China
| |
Collapse
|
21
|
Su R, Zhang H, Zhang L, Khan AR, Zhang X, Wang R, Shao C, Wei X, Xu X. Systemic analysis identifying
PVT1
/
DUSP13
axis for microvascular invasion in hepatocellular carcinoma. Cancer Med 2022; 12:8937-8955. [PMID: 36524545 PMCID: PMC10134337 DOI: 10.1002/cam4.5546] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2022] [Revised: 10/26/2022] [Accepted: 11/04/2022] [Indexed: 12/23/2022] Open
Abstract
BACKGROUND Microvascular invasion (MVI) is an independent detrimental risk factor for tumor recurrence and poor survival in hepatocellular carcinoma (HCC). Competitive endogenous RNA (ceRNA) networks play a pivotal role in the modulation of carcinogenesis and progression among diverse tumor types. However, whether the ceRNA mechanisms are engaged in promoting the MVI process in patients with HCC remains unknown. METHODS A ceRNA regulatory network was constructed based on RNA-seq data of patients with HCC from The Cancer Genome Atlas (TCGA) database. In total, 10 hub genes of the ceRNA network were identified using four algorithms: "MCC," "Degree," "Betweenness," and "Stress." Transcriptional expressions were verified by in situ hybridization using clinical samples. Interactions between ceRNA modules were validated by luciferase reporting assay. Logistic regression analysis, correlation analysis, enrichment analysis, promoter region analysis, methylation analysis, and immune infiltration analysis were performed to further investigate the molecular mechanisms and clinical transformation value. RESULTS The ceRNA regulatory network featuring a tumor invasion phenotype consisting of 3 long noncoding RNAs, 3 microRNAs, and 93 mRNAs was constructed using transcriptional data from the TCGA database. Systemic analysis and experimentally validation identified a ceRNA network (PVT1/miR-1258/DUSP13 axis) characterized by lipid regulatory potential, immune properties, and abnormal methylation states in patients with HCC and MVI. Meanwhile, 28 transcriptional factors were identified as potential promotors of PVT1 with 3 transcriptional factors MXD3, ZNF580, and KDM1A promising as therapeutic targets in patients with HCC and MVI. Furthermore, miR-1258 was an independent predictor for MVI in patients with HCC. CONCLUSION The PVT1/DUSP13 axis is significantly associated with MVI progression in HCC patients. This study provides new insight into mechanisms related to lipids, immune phenotypes, and abnormal epigenetics in oncology research.
Collapse
Affiliation(s)
- Renyi Su
- Institute of Organ Transplantation, Zhejiang University Hangzhou China
- Key Laboratory of Integrated Oncology and Intelligent Medicine of Zhejiang Province, Department of Hepatobiliary and Pancreatic Surgery, Affiliated Hangzhou First People's Hospital Zhejiang University School of Medicine Hangzhou China
| | - Huizhong Zhang
- Key Laboratory of Integrated Oncology and Intelligent Medicine of Zhejiang Province, Department of Hepatobiliary and Pancreatic Surgery, Affiliated Hangzhou First People's Hospital Zhejiang University School of Medicine Hangzhou China
| | - Lincheng Zhang
- Institute of Organ Transplantation, Zhejiang University Hangzhou China
- Key Laboratory of Integrated Oncology and Intelligent Medicine of Zhejiang Province, Department of Hepatobiliary and Pancreatic Surgery, Affiliated Hangzhou First People's Hospital Zhejiang University School of Medicine Hangzhou China
| | - Abdul Rehman Khan
- Institute of Organ Transplantation, Zhejiang University Hangzhou China
- Key Laboratory of Integrated Oncology and Intelligent Medicine of Zhejiang Province, Department of Hepatobiliary and Pancreatic Surgery, Affiliated Hangzhou First People's Hospital Zhejiang University School of Medicine Hangzhou China
| | - Xuanyu Zhang
- Institute of Organ Transplantation, Zhejiang University Hangzhou China
- Department of Hepatobiliary and Pancreatic Surgery, The First Affiliated Hospital Zhejiang University School of Medicine Hangzhou China
| | - Rui Wang
- Institute of Organ Transplantation, Zhejiang University Hangzhou China
| | - Chuxiao Shao
- Department of Hepatobiliary and Pancreatic Surgery, Affiliated Lishui Hospital Zhejiang University School of Medicine Lishui China
| | - Xuyong Wei
- Institute of Organ Transplantation, Zhejiang University Hangzhou China
- Key Laboratory of Integrated Oncology and Intelligent Medicine of Zhejiang Province, Department of Hepatobiliary and Pancreatic Surgery, Affiliated Hangzhou First People's Hospital Zhejiang University School of Medicine Hangzhou China
| | - Xiao Xu
- Institute of Organ Transplantation, Zhejiang University Hangzhou China
- Key Laboratory of Integrated Oncology and Intelligent Medicine of Zhejiang Province, Department of Hepatobiliary and Pancreatic Surgery, Affiliated Hangzhou First People's Hospital Zhejiang University School of Medicine Hangzhou China
- Westlake Laboratory of Life Sciences and Biomedicine Hangzhou China
| |
Collapse
|
22
|
Corral A, Alcala M, Carmen Duran-Ruiz M, Arroba AI, Ponce-Gonzalez JG, Todorčević M, Serra D, Calderon-Dominguez M, Herrero L. Role of long non-coding RNAs in adipose tissue metabolism and associated pathologies. Biochem Pharmacol 2022; 206:115305. [DOI: 10.1016/j.bcp.2022.115305] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2022] [Revised: 10/11/2022] [Accepted: 10/12/2022] [Indexed: 11/17/2022]
|
23
|
Nadhan R, Dhanasekaran DN. Regulation of Tumor Metabolome by Long Non-Coding RNAs. J Mol Signal 2022. [DOI: 10.55233/1750-2187-16-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022] Open
|
24
|
Noncoding RNAs Associated with PPARs in Etiology of MAFLD as a Novel Approach for Therapeutics Targets. PPAR Res 2022; 2022:6161694. [PMID: 36164476 PMCID: PMC9509273 DOI: 10.1155/2022/6161694] [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: 03/02/2022] [Revised: 07/25/2022] [Accepted: 08/27/2022] [Indexed: 11/18/2022] Open
Abstract
Background. Metabolic associated fatty liver disease (MAFLD) is a complex disease that results from the accumulation of fat in the liver. MAFLD is directly associated with obesity, insulin resistance, diabetes, and metabolic syndrome. PPARγ ligands, including pioglitazone, are also used in the management of this disease. Noncoding RNAs play a critical role in various diseases such as diabetes, obesity, and liver diseases including MAFLD. However, there is no adequate knowledge about the translation of using these ncRNAs to the clinics, particularly in MAFLD conditions. The aim of this study was to identify ncRNAs in the etiology of MAFLD as a novel approach to the therapeutic targets. Methods. We collected human and mouse MAFLD gene expression datasets available in GEO. We performed pathway enrichment analysis of total mRNAs based on KEGG repository data to screen the most potential pathways in the liver of MAFLD human subjects and mice model, and analyzed pathway interconnections via ClueGO. Finally, we screened disease causality of the MAFLD ncRNAs, which were associated with PPARs, and then discussed the role of revealed ncRNAs in PPAR signaling and MAFLD. Results. We found 127 ncRNAs in MAFLD which 25 out of them were strongly validated before for regulation of PPARs. With a polypharmacology approach, we screened 51 ncRNAs which were causal to a subset of diseases related to MAFLD. Conclusion. This study revealed a subset of ncRNAs that could help in more clear and guided designation of preclinical and clinical studies to verify the therapeutic application of the revealed ncRNAs by manipulating the PPARs molecular mechanism in MAFLD.
Collapse
|
25
|
Rashidmayvan M, Sahebi R, Ghayour-Mobarhan M. Long non-coding RNAs: a valuable biomarker for metabolic syndrome. Mol Genet Genomics 2022; 297:1169-1183. [PMID: 35854006 DOI: 10.1007/s00438-022-01922-1] [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: 03/27/2022] [Accepted: 06/25/2022] [Indexed: 10/17/2022]
Abstract
Long non-coding RNAs (lncRNAs) have become important regulators of gene expression because they affect a wide range of biological processes, such as cell growth, death, differentiation, and aging. More and more evidence suggests that lncRNAs play a role in maintaining metabolic homeostasis. When certain lncRNAs are out of balance, metabolic disorders like diabetes, obesity, and heart disease get worse. In this review, we talk about what we know about how lncRNAs control metabolism, with a focus on diseases caused by long-term inflammation and the characteristics of the metabolic syndrome. We looked at lncRNAs and their molecular targets in the pathogenesis of signaling pathways. We also talked about how lncRNAs are becoming more and more interesting as diagnostic and therapeutic targets for improving metabolic homeostasis.
Collapse
Affiliation(s)
- Mohammad Rashidmayvan
- Department of Nutrition, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Reza Sahebi
- Metabolic Syndrome Research Center, School of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Majid Ghayour-Mobarhan
- Department of Nutrition, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran.
| |
Collapse
|
26
|
Lovell CD, Anguera MC. Long Noncoding RNAs That Function in Nutrition: Lnc-ing Nutritional Cues to Metabolic Pathways. Annu Rev Nutr 2022; 42:251-274. [PMID: 35436418 DOI: 10.1146/annurev-nutr-062220-030244] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Long noncoding RNAs (lncRNAs) are sensitive to changing environments and play key roles in health and disease. Emerging evidence indicates that lncRNAs regulate gene expression to shape metabolic processes in response to changing nutritional cues. Here we review various lncRNAs sensitive to fasting, feeding, and high-fat diet in key metabolic tissues (liver, adipose, and muscle), highlighting regulatory mechanisms that trigger expression changes of lncRNAs themselves, and how these lncRNAs regulate gene expression of key metabolic genes in specific cell types or across tissues. Determining how lncRNAs respond to changes in nutrition is critical for our understanding of the complex downstream cascades following dietary changes and can shape how we treat metabolic disease. Furthermore, investigating sex biases that might influence lncRNA-regulated responses will likely reveal contributions toward the observed disparities between the sexes in metabolic diseases.
Collapse
Affiliation(s)
- Claudia D Lovell
- Department of Biomedical Sciences, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA;
| | - Montserrat C Anguera
- Department of Biomedical Sciences, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA;
| |
Collapse
|
27
|
Piergentili R, Basile G, Nocella C, Carnevale R, Marinelli E, Patrone R, Zaami S. Using ncRNAs as Tools in Cancer Diagnosis and Treatment-The Way towards Personalized Medicine to Improve Patients' Health. Int J Mol Sci 2022; 23:9353. [PMID: 36012617 PMCID: PMC9409241 DOI: 10.3390/ijms23169353] [Citation(s) in RCA: 35] [Impact Index Per Article: 11.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2022] [Revised: 08/14/2022] [Accepted: 08/16/2022] [Indexed: 12/06/2022] Open
Abstract
Although the first discovery of a non-coding RNA (ncRNA) dates back to 1958, only in recent years has the complexity of the transcriptome started to be elucidated. However, its components are still under investigation and their identification is one of the challenges that scientists are presently facing. In addition, their function is still far from being fully understood. The non-coding portion of the genome is indeed the largest, both quantitatively and qualitatively. A large fraction of these ncRNAs have a regulatory role either in coding mRNAs or in other ncRNAs, creating an intracellular network of crossed interactions (competing endogenous RNA networks, or ceRNET) that fine-tune the gene expression in both health and disease. The alteration of the equilibrium among such interactions can be enough to cause a transition from health to disease, but the opposite is equally true, leading to the possibility of intervening based on these mechanisms to cure human conditions. In this review, we summarize the present knowledge on these mechanisms, illustrating how they can be used for disease treatment, the current challenges and pitfalls, and the roles of environmental and lifestyle-related contributing factors, in addition to the ethical, legal, and social issues arising from their (improper) use.
Collapse
Affiliation(s)
- Roberto Piergentili
- Institute of Molecular Biology and Pathology, Italian National Research Council (CNR-IBPM), 00185 Rome, Italy
| | - Giuseppe Basile
- Trauma Unit and Emergency Department, IRCCS Galeazzi Orthopedics Institute, 20161 Milan, Italy
- Head of Legal Medicine Unit, Clinical Institute San Siro, 20148 Milan, Italy
| | - Cristina Nocella
- Department of Clinical Internal, Anaesthesiological and Cardiovascular Sciences, “Sapienza” University of Rome, Viale del Policlinico, 155, 00161 Rome, Italy
| | - Roberto Carnevale
- Department of Medico-Surgical Sciences and Biotechnologies, “Sapienza” University of Rome, 04100 Latina, Italy
- Mediterranea Cardiocentro-Napoli, Via Orazio, 80122 Naples, Italy
| | - Enrico Marinelli
- Department of Medico-Surgical Sciences and Biotechnologies, “Sapienza” University of Rome, 04100 Latina, Italy
| | - Renato Patrone
- PhD ICTH, University of Federico II, HPB Department INT F. Pascale IRCCS of Naples, Via Mariano Semmola, 80131 Naples, Italy
| | - Simona Zaami
- Department of Anatomical, Histological, Forensic and Orthopedic Sciences, Section of Forensic Medicine, “Sapienza” University of Rome, 00161 Rome, Italy
| |
Collapse
|
28
|
Zhang P, Wu S, He Y, Li X, Zhu Y, Lin X, Chen L, Zhao Y, Niu L, Zhang S, Li X, Zhu L, Shen L. LncRNA-Mediated Adipogenesis in Different Adipocytes. Int J Mol Sci 2022; 23:ijms23137488. [PMID: 35806493 PMCID: PMC9267348 DOI: 10.3390/ijms23137488] [Citation(s) in RCA: 24] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2022] [Revised: 06/29/2022] [Accepted: 07/04/2022] [Indexed: 02/01/2023] Open
Abstract
Long-chain noncoding RNAs (lncRNAs) are RNAs that do not code for proteins, widely present in eukaryotes. They regulate gene expression at multiple levels through different mechanisms at epigenetic, transcription, translation, and the maturation of mRNA transcripts or regulation of the chromatin structure, and compete with microRNAs for binding to endogenous RNA. Adipose tissue is a large and endocrine-rich functional tissue in mammals. Excessive accumulation of white adipose tissue in mammals can cause metabolic diseases. However, unlike white fat, brown and beige fats release energy as heat. In recent years, many lncRNAs associated with adipogenesis have been reported. The molecular mechanisms of how lncRNAs regulate adipogenesis are continually investigated. In this review, we discuss the classification of lncRNAs according to their transcriptional location. lncRNAs that participate in the adipogenesis of white or brown fats are also discussed. The function of lncRNAs as decoy molecules and RNA double-stranded complexes, among other functions, is also discussed.
Collapse
Affiliation(s)
- Peiwen Zhang
- College of Animal Science and Technology, Sichuan Agricultural University, Chengdu 611130, China; (P.Z.); (S.W.); (Y.H.); (X.L.); (X.L.); (L.C.); (Y.Z.); (L.N.); (S.Z.); (X.L.)
- Farm Animal Genetic Resources Exploration and Innovation Key Laboratory of Sichuan Province, Sichuan Agricultural University, Chengdu 611130, China
| | - Shuang Wu
- College of Animal Science and Technology, Sichuan Agricultural University, Chengdu 611130, China; (P.Z.); (S.W.); (Y.H.); (X.L.); (X.L.); (L.C.); (Y.Z.); (L.N.); (S.Z.); (X.L.)
- Farm Animal Genetic Resources Exploration and Innovation Key Laboratory of Sichuan Province, Sichuan Agricultural University, Chengdu 611130, China
| | - Yuxu He
- College of Animal Science and Technology, Sichuan Agricultural University, Chengdu 611130, China; (P.Z.); (S.W.); (Y.H.); (X.L.); (X.L.); (L.C.); (Y.Z.); (L.N.); (S.Z.); (X.L.)
- Farm Animal Genetic Resources Exploration and Innovation Key Laboratory of Sichuan Province, Sichuan Agricultural University, Chengdu 611130, China
| | - Xinrong Li
- College of Animal Science and Technology, Sichuan Agricultural University, Chengdu 611130, China; (P.Z.); (S.W.); (Y.H.); (X.L.); (X.L.); (L.C.); (Y.Z.); (L.N.); (S.Z.); (X.L.)
- Farm Animal Genetic Resources Exploration and Innovation Key Laboratory of Sichuan Province, Sichuan Agricultural University, Chengdu 611130, China
| | - Yan Zhu
- College of Life Science, China West Normal University, Nanchong 637009, China;
| | - Xutao Lin
- College of Animal Science and Technology, Sichuan Agricultural University, Chengdu 611130, China; (P.Z.); (S.W.); (Y.H.); (X.L.); (X.L.); (L.C.); (Y.Z.); (L.N.); (S.Z.); (X.L.)
- Farm Animal Genetic Resources Exploration and Innovation Key Laboratory of Sichuan Province, Sichuan Agricultural University, Chengdu 611130, China
| | - Lei Chen
- College of Animal Science and Technology, Sichuan Agricultural University, Chengdu 611130, China; (P.Z.); (S.W.); (Y.H.); (X.L.); (X.L.); (L.C.); (Y.Z.); (L.N.); (S.Z.); (X.L.)
- Farm Animal Genetic Resources Exploration and Innovation Key Laboratory of Sichuan Province, Sichuan Agricultural University, Chengdu 611130, China
| | - Ye Zhao
- College of Animal Science and Technology, Sichuan Agricultural University, Chengdu 611130, China; (P.Z.); (S.W.); (Y.H.); (X.L.); (X.L.); (L.C.); (Y.Z.); (L.N.); (S.Z.); (X.L.)
- Farm Animal Genetic Resources Exploration and Innovation Key Laboratory of Sichuan Province, Sichuan Agricultural University, Chengdu 611130, China
| | - Lili Niu
- College of Animal Science and Technology, Sichuan Agricultural University, Chengdu 611130, China; (P.Z.); (S.W.); (Y.H.); (X.L.); (X.L.); (L.C.); (Y.Z.); (L.N.); (S.Z.); (X.L.)
- Farm Animal Genetic Resources Exploration and Innovation Key Laboratory of Sichuan Province, Sichuan Agricultural University, Chengdu 611130, China
| | - Shunhua Zhang
- College of Animal Science and Technology, Sichuan Agricultural University, Chengdu 611130, China; (P.Z.); (S.W.); (Y.H.); (X.L.); (X.L.); (L.C.); (Y.Z.); (L.N.); (S.Z.); (X.L.)
- Farm Animal Genetic Resources Exploration and Innovation Key Laboratory of Sichuan Province, Sichuan Agricultural University, Chengdu 611130, China
| | - Xuewei Li
- College of Animal Science and Technology, Sichuan Agricultural University, Chengdu 611130, China; (P.Z.); (S.W.); (Y.H.); (X.L.); (X.L.); (L.C.); (Y.Z.); (L.N.); (S.Z.); (X.L.)
- Farm Animal Genetic Resources Exploration and Innovation Key Laboratory of Sichuan Province, Sichuan Agricultural University, Chengdu 611130, China
| | - Li Zhu
- College of Animal Science and Technology, Sichuan Agricultural University, Chengdu 611130, China; (P.Z.); (S.W.); (Y.H.); (X.L.); (X.L.); (L.C.); (Y.Z.); (L.N.); (S.Z.); (X.L.)
- Farm Animal Genetic Resources Exploration and Innovation Key Laboratory of Sichuan Province, Sichuan Agricultural University, Chengdu 611130, China
- Correspondence: (L.Z.); (L.S.); Tel.: +86-28-8629-1133 (L.Z. & L.S.)
| | - Linyuan Shen
- College of Animal Science and Technology, Sichuan Agricultural University, Chengdu 611130, China; (P.Z.); (S.W.); (Y.H.); (X.L.); (X.L.); (L.C.); (Y.Z.); (L.N.); (S.Z.); (X.L.)
- Farm Animal Genetic Resources Exploration and Innovation Key Laboratory of Sichuan Province, Sichuan Agricultural University, Chengdu 611130, China
- Correspondence: (L.Z.); (L.S.); Tel.: +86-28-8629-1133 (L.Z. & L.S.)
| |
Collapse
|
29
|
Santoleri D, Lim HW, Emmett MJ, Stoute J, Gavin MJ, Sostre-Colón J, Uehara K, Welles JE, Liu KF, Lazar MA, Titchenell PM. Global-run on sequencing identifies Gm11967 as an Akt-dependent long noncoding RNA involved in insulin sensitivity. iScience 2022; 25:104410. [PMID: 35663017 PMCID: PMC9156944 DOI: 10.1016/j.isci.2022.104410] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2021] [Revised: 03/18/2022] [Accepted: 05/11/2022] [Indexed: 01/07/2023] Open
Abstract
The insulin responsive Akt and FoxO1 signaling axis is a key regulator of the hepatic transcriptional response to nutrient intake. Here, we used global run-on sequencing (GRO-seq) to measure the nascent transcriptional response to fasting and refeeding as well as define the specific role of hepatic Akt and FoxO1 signaling in mediating this response. We identified 599 feeding-regulated transcripts, as well as over 6,000 eRNAs, and mapped their dependency on Akt and FoxO1 signaling. Further, we identified several feeding-regulated lncRNAs, including the lncRNA Gm11967, whose expression was dependent upon the liver Akt-FoxO1 axis. Restoring Gm11967 expression in mice lacking liver Akt improved insulin sensitivity and induced glucokinase protein expression, indicating that Akt-dependent control of Gm11967 contributes to the translational control of glucokinase. More broadly, we have generated a unique genome-wide dataset that defines the feeding and Akt/FoxO1-dependent transcriptional changes in response to nutrient availability.
Collapse
Affiliation(s)
- Dominic Santoleri
- Biochemistry and Molecular Biophysics Graduate Group, University of Pennsylvania Biomedical Graduate Studies, Philadelphia, PA 19104, USA
- Institute of Diabetes, Obesity and Metabolism, Smilow Center for Translational Research, University of Pennsylvania Perelman School of Medicine, 3400 Civic Center Blvd, Building 421, Philadelphia, PA 19104, USA
| | - Hee-Woong Lim
- Division of Biomedical Informatics, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH 45229, USA
- Department of Pediatrics, University of Cincinnati, Cincinnati, OH 45229, USA
| | - Matthew J. Emmett
- Department of Medicine, Massachusetts General Hospital, Boston, MA 02114, USA
| | - Julian Stoute
- Biochemistry and Molecular Biophysics Graduate Group, University of Pennsylvania Biomedical Graduate Studies, Philadelphia, PA 19104, USA
- Department of Biochemistry and Biophysics, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA 19104, USA
| | - Matthew J. Gavin
- Institute of Diabetes, Obesity and Metabolism, Smilow Center for Translational Research, University of Pennsylvania Perelman School of Medicine, 3400 Civic Center Blvd, Building 421, Philadelphia, PA 19104, USA
| | - Jaimarie Sostre-Colón
- Institute of Diabetes, Obesity and Metabolism, Smilow Center for Translational Research, University of Pennsylvania Perelman School of Medicine, 3400 Civic Center Blvd, Building 421, Philadelphia, PA 19104, USA
| | - Kahealani Uehara
- Biochemistry and Molecular Biophysics Graduate Group, University of Pennsylvania Biomedical Graduate Studies, Philadelphia, PA 19104, USA
- Institute of Diabetes, Obesity and Metabolism, Smilow Center for Translational Research, University of Pennsylvania Perelman School of Medicine, 3400 Civic Center Blvd, Building 421, Philadelphia, PA 19104, USA
| | - Jaclyn E. Welles
- Institute of Diabetes, Obesity and Metabolism, Smilow Center for Translational Research, University of Pennsylvania Perelman School of Medicine, 3400 Civic Center Blvd, Building 421, Philadelphia, PA 19104, USA
| | - Kathy Fange Liu
- Biochemistry and Molecular Biophysics Graduate Group, University of Pennsylvania Biomedical Graduate Studies, Philadelphia, PA 19104, USA
- Department of Biochemistry and Biophysics, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA 19104, USA
| | - Mitchell A. Lazar
- Institute of Diabetes, Obesity and Metabolism, Smilow Center for Translational Research, University of Pennsylvania Perelman School of Medicine, 3400 Civic Center Blvd, Building 421, Philadelphia, PA 19104, USA
- Division of Endocrinology, Diabetes, and Metabolism, Department of Medicine, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Paul M. Titchenell
- Biochemistry and Molecular Biophysics Graduate Group, University of Pennsylvania Biomedical Graduate Studies, Philadelphia, PA 19104, USA
- Institute of Diabetes, Obesity and Metabolism, Smilow Center for Translational Research, University of Pennsylvania Perelman School of Medicine, 3400 Civic Center Blvd, Building 421, Philadelphia, PA 19104, USA
- Department of Physiology, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA 19104, USA
| |
Collapse
|
30
|
Bao G, Li S, Zhao F, Wang J, Liu X, Hu J, Shi B, Wen Y, Zhao L, Luo Y. Comprehensive Transcriptome Analysis Reveals the Role of lncRNA in Fatty Acid Metabolism in the Longissimus Thoracis Muscle of Tibetan Sheep at Different Ages. Front Nutr 2022; 9:847077. [PMID: 35369085 PMCID: PMC8964427 DOI: 10.3389/fnut.2022.847077] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/01/2022] [Accepted: 02/11/2022] [Indexed: 12/19/2022] Open
Abstract
Long noncoding RNA (lncRNA) plays an important regulatory role in mammalian adipogenesis and lipid metabolism. However, their function in the longissimus thoracis (LT) muscle of fatty acid metabolism of Tibetan sheep remains undefined. In this study, fatty acid and fat content in LT muscle of Tibetan sheep were determined, and RNA sequencing was performed to reveal the temporal regularity of lncRNA expression and the effect of lncRNA-miRNA-mRNA ceRNA regulatory network on lipid metabolism of LT muscle in Tibetan sheep at four growth stages (4-month-old, 4 m; 1.5-year-old, 1.5 y; 3.5-year-old, 3.5 y; 6-year-old, 6 y). The results indicated that the intramuscular fat (IMF) content was highest at 1.5 y. Moreover, the monounsaturated fatty acid (MUFA) content in 1.5 y of Tibetan sheep is significantly higher than those of the other groups (P < 0.05), and it was also rich in a variety of polyunsaturated fatty acids (PUFA). A total of 360 differentially expressed lncRNAs (DE lncRNAs) were identified from contiguous period transcriptome comparative groups of 4 m vs. 1.5 y, 1.5 y vs. 3.5 y, 3.5 y vs. 6 y, and 4 m vs. 6 y, respectively. Kyoto encyclopedia of genes and genomes (KEGG) enrichment analysis found that the target genes in lncRNA trans-mRNA were significantly related to the protein digestion, absorption, and fatty acid biosynthesis pathways (P < 0.05), which demonstrated that DE lncRNA trans-regulated the target genes, and further regulated the growth and development of the LT muscle and intramuscular fatty acid metabolism in Tibetan sheep. We further analyzed the role of the lncRNA-miRNA-mRNA regulatory network in the lipid metabolism of Tibetan sheep. Additionally, GPD2, LIPE (lipase E hormone-sensitive enzyme), TFDP2, CPT1A, ACACB, ADIPOQ, and other mRNA related to fatty acid and lipid metabolism and the corresponding lncRNA-miRNA regulatory pairs were identified. The enrichment analysis of mRNA in the regulatory network found that the AMPK signaling pathway was the most significantly enriched (P = 0.0000112361). Comprehensive transcriptome analysis found that the LIPE, ADIPOQ, ACACB, and CPT1A that were regulated by lncRNA might change the formation of energy metabolism in Tibetan sheep muscle through the AMPK signaling pathway, and oxidized muscle fibers are transformed into glycolytic muscle fibers, reduced IMF content, and the fatty acid profile also changed.
Collapse
Affiliation(s)
- Gaoliang Bao
- Gansu Key Laboratory of Herbivorous Animal Biotechnology, Faculty of Animal Science and Technology, Gansu Agricultural University, Lanzhou, China
| | - Shaobin Li
- Gansu Key Laboratory of Herbivorous Animal Biotechnology, Faculty of Animal Science and Technology, Gansu Agricultural University, Lanzhou, China
| | - Fangfang Zhao
- Gansu Key Laboratory of Herbivorous Animal Biotechnology, Faculty of Animal Science and Technology, Gansu Agricultural University, Lanzhou, China
| | - Jiqing Wang
- Gansu Key Laboratory of Herbivorous Animal Biotechnology, Faculty of Animal Science and Technology, Gansu Agricultural University, Lanzhou, China
| | - Xiu Liu
- Gansu Key Laboratory of Herbivorous Animal Biotechnology, Faculty of Animal Science and Technology, Gansu Agricultural University, Lanzhou, China
| | - Jiang Hu
- Gansu Key Laboratory of Herbivorous Animal Biotechnology, Faculty of Animal Science and Technology, Gansu Agricultural University, Lanzhou, China
| | - Bingang Shi
- Gansu Key Laboratory of Herbivorous Animal Biotechnology, Faculty of Animal Science and Technology, Gansu Agricultural University, Lanzhou, China
| | - Yuliang Wen
- Gansu Key Laboratory of Herbivorous Animal Biotechnology, Faculty of Animal Science and Technology, Gansu Agricultural University, Lanzhou, China
| | - Li Zhao
- Gansu Key Laboratory of Herbivorous Animal Biotechnology, Faculty of Animal Science and Technology, Gansu Agricultural University, Lanzhou, China
| | - Yuzhu Luo
- Gansu Key Laboratory of Herbivorous Animal Biotechnology, Faculty of Animal Science and Technology, Gansu Agricultural University, Lanzhou, China
| |
Collapse
|
31
|
Hennessy EJ. LncRNAs and Cardiovascular Disease. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2022; 1363:71-95. [PMID: 35220566 DOI: 10.1007/978-3-030-92034-0_5] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Indexed: 10/19/2022]
Abstract
A novel class of RNA molecule emerged from human transcriptome sequencing studies termed long non-coding RNAs. These RNA molecules differ from other classes of non-coding RNAs such as microRNAs in their sizes, sequence motifs and structures. Studies have demonstrated that long non-coding RNAs play a prominent role in the development and progression of cardiovascular disease. They provide the cell with tiered levels of gene regulation interacting with DNA, other RNA molecules or proteins acting in various capacities to control a variety of cellular mechanisms. Cell specificity is a hallmark of lncRNA studies and they have been identified in macrophages, smooth muscle cells, endothelial cells and hepatocytes. Recent lncRNA studies have uncovered functional micropeptides encoded within lncRNA genes that can have a different function to the lncRNA. Disease associated mutations in the genome tend to occur in non-coding regions signifying the importance of non-coding genes in disease associations. There is a great deal of work to be done in the non-coding RNA field and tremendous therapeutic potential due to their cell type specificity. A better understanding of the functions and interactions of lncRNAs will inevitably have clinical implications.
Collapse
Affiliation(s)
- Elizabeth J Hennessy
- University of Pennsylvania, Perelman School of Medicine, Institute for Translational Medicine and Therapeutics (ITMAT), Philadelphia, PA, USA.
| |
Collapse
|
32
|
Fontanini M, Cabiati M, Giacomarra M, Federico G, Del Ry S. Long non-Coding RNAs and Obesity: New Potential Pathogenic Biomarkers. Curr Pharm Des 2022; 28:1592-1605. [DOI: 10.2174/1381612828666220211153304] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2021] [Accepted: 01/17/2022] [Indexed: 11/22/2022]
Abstract
Background:
A portion of the human genome is characterized by long non-coding RNAs (lncRNAs), a class of non-coding RNA longer than 200 nucleotides. Recently, the development of new biomolecular methods, made it possible to delineate the involvement of lncRNAs in the regulation of different biological processes, both physiological and pathological, by acting within the cell with different regulatory mechanisms based on their specific target. To date, obesity is one of the most important health problems spread all over the world, including the child population: the search for new potential early biomarkers could open the doors to novel therapeutic strategies useful to fight the disease early in life and to reduce the risk of obesity-related co-morbidities.
Objective:
This review highlights the lncRNAs involved in obesity, in adipogenesis, and lipid metabolism, particularly in lipogenesis.
Conclusion:
LncRNAs involved in adipogenesis and lipogenesis, being at the cross-road of obesity, should be deeply analysed in this contest, allowing to understand possible causative actions in starting obesity and whether they might be helpful to treat obesity.
Collapse
Affiliation(s)
- Martina Fontanini
- CNR Institute of Clinical Physiology, Biochemistry and Molecular Biology laboratory, Via G. Moruzzi 1, 56124 Pisa Italy
| | - Manuela Cabiati
- CNR Institute of Clinical Physiology, Biochemistry and Molecular Biology laboratory, Via G. Moruzzi 1, 56124 Pisa Italy
| | - Manuel Giacomarra
- CNR Institute of Clinical Physiology, Biochemistry and Molecular Biology laboratory, Via G. Moruzzi 1, 56124 Pisa Italy
| | - Giovanni Federico
- Unit of Pediatric Endocrinology and Diabetes, Dep. Clinical and Experimental Medicine, University of Pisa, Via Roma n. 67 56126 Pisa, Italy
| | - Silvia Del Ry
- CNR Institute of Clinical Physiology, Biochemistry and Molecular Biology laboratory, Via G. Moruzzi 1, 56124 Pisa Italy
| |
Collapse
|
33
|
Desind SZ, Iacona JR, Yu CY, Mitrofanova A, Lutz CS. PACER lncRNA regulates COX-2 expression in lung cancer cells. Oncotarget 2022; 13:291-306. [PMID: 35136486 PMCID: PMC8815784 DOI: 10.18632/oncotarget.28190] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2021] [Accepted: 01/25/2022] [Indexed: 11/28/2022] Open
Abstract
Long noncoding RNAs (lncRNAs) are known to regulate gene expression; however, in many cases, the mechanism of this regulation is unknown. One novel lncRNA relevant to inflammation and arachidonic acid (AA) metabolism is the p50-associated COX-2 extragenic RNA (PACER). We focused our research on the regulation of PACER in lung cancer. While the function of PACER is not entirely understood, PACER is known to play a role in inflammation-associated conditions. Our data suggest that PACER is critically involved in COX-2 transcription and dysregulation in lung cancer cells. Our analysis of The Cancer Genome Atlas (TCGA) expression data revealed that PACER expression is significantly higher in lung adenocarcinomas than normal lung tissues. Additionally, we discovered that elevated PACER expression strongly correlates with COX-2 expression in lung adenocarcinoma patients. Specific siRNA-mediated knockdown of PACER decreases COX-2 expression indicating a direct relationship. Additionally, we show that PACER expression is induced upon treatment with proinflammatory cytokines to mimic inflammation. Treatment with prostaglandin E2 (PGE2) induces both PACER and COX-2 expression, suggesting a PGE2-mediated feedback loop. Inhibition of COX-2 with celecoxib decreased PACER expression, confirming this self-regulatory process. Significant overlap between the COX-2 promotor and the PACER promotor led us to investigate their transcriptional regulatory mechanisms. Treatment with pharmacologic inhibitors of NF-κB or AP-1 showed a modest effect on both PACER and COX-2 expression but did not eliminate expression. These data suggest that the regulation of expression of both PACER and COX-2 is complex and intricately linked.
Collapse
Affiliation(s)
- Samuel Z. Desind
- Department of Microbiology, Biochemistry, and Molecular Genetics, Rutgers Biomedical and Health Sciences, New Jersey Medical School, School of Graduate Studies, Newark, NJ 07103, USA
- These authors contributed equally to this work
| | - Joseph R. Iacona
- Department of Microbiology, Biochemistry, and Molecular Genetics, Rutgers Biomedical and Health Sciences, New Jersey Medical School, School of Graduate Studies, Newark, NJ 07103, USA
- These authors contributed equally to this work
| | - Christina Y. Yu
- Department of Health Informatics, Rutgers School of Health Professions, Rutgers Biomedical and Health Sciences, Newark, NJ 07107, USA
| | - Antonina Mitrofanova
- Department of Health Informatics, Rutgers School of Health Professions, Rutgers Biomedical and Health Sciences, Newark, NJ 07107, USA
- Rutgers Cancer Institute of New Jersey, Rutgers, The State University of New Jersey, New Brunswick, NJ 08901, USA
| | - Carol S. Lutz
- Department of Microbiology, Biochemistry, and Molecular Genetics, Rutgers Biomedical and Health Sciences, New Jersey Medical School, School of Graduate Studies, Newark, NJ 07103, USA
| |
Collapse
|
34
|
Mumtaz PT, Taban Q, Bhat B, Ahmad SM, Dar MA, Kashoo ZA, Ganie NA, Shah RA. Expression of lncRNAs in response to bacterial infections of goat mammary epithelial cells reveals insights into mammary gland diseases. Microb Pathog 2021; 162:105367. [PMID: 34963641 DOI: 10.1016/j.micpath.2021.105367] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2021] [Revised: 12/16/2021] [Accepted: 12/17/2021] [Indexed: 10/19/2022]
Abstract
Mastitis or inflammation of the mammary gland is a highly economic and deadly alarming disease for the dairy sector as well as policymakers caused by microbial infection. Transcriptomic and proteomic approaches have been widely employed to identify the underlying molecular mechanisms of bacterial infections in the mammary gland. Numerous differentially expressed mRNAs, miRNAs, and proteins together with their associated signaling pathways have been identified during bacterial infection, paving the way for analysis of their biological functions. Long noncoding RNAs (lncRNAs) are important regulators of multiple biological processes. However, little is known regarding their role in bacterial infection in mammary epithelial cells. Hence, RNA-sequencing was performed by infecting primary mammary epithelial cells (pMECs) with both gram-negative (E. coli) and gram-positive bacteria (S. aureus). Using stringent pipeline, a set of 1957 known and 1175 novel lncRNAs were identified, among which, 112 lncRNAs were found differentially expressed in bacteria challenged PMECs compared with the control. Additionally, potential targets of the lncRNAs were predicted in cis- and trans-configuration. KEGG analysis revealed that DE lncRNAs were associated with at least 15 immune-related pathways. Therefore, our study revealed that bacterial challenge triggers the expression of lncRNAs associated with immune response and defense mechanisms in goat mammary epithelial cells.
Collapse
Affiliation(s)
- Peerzada Tajamul Mumtaz
- Division of Animal Biotechnology, Faculty of Veterinary Sciences and Animal Husbandry, Shuhama, SKUAST-K, India; Department of Biochemistry, School of Life Sciences Jaipur National University, India
| | - Qamar Taban
- Division of Animal Biotechnology, Faculty of Veterinary Sciences and Animal Husbandry, Shuhama, SKUAST-K, India
| | - Basharat Bhat
- Division of Animal Biotechnology, Faculty of Veterinary Sciences and Animal Husbandry, Shuhama, SKUAST-K, India
| | - Syed Mudasir Ahmad
- Division of Animal Biotechnology, Faculty of Veterinary Sciences and Animal Husbandry, Shuhama, SKUAST-K, India.
| | - Mashooq Ahmad Dar
- Division of Animal Biotechnology, Faculty of Veterinary Sciences and Animal Husbandry, Shuhama, SKUAST-K, India
| | - Zahid Amin Kashoo
- Division of Veterinary Microbiology, Faculty of Veterinary Sciences and Animal Husbandry, Shuhama, SKUAST-K, India
| | - Nazir A Ganie
- Division of Animal Biotechnology, Faculty of Veterinary Sciences and Animal Husbandry, Shuhama, SKUAST-K, India
| | - Riaz Ahmad Shah
- Division of Animal Biotechnology, Faculty of Veterinary Sciences and Animal Husbandry, Shuhama, SKUAST-K, India
| |
Collapse
|
35
|
Xin M, Guo Q, Lu Q, Lu J, Wang PS, Dong Y, Li T, Chen Y, Gerhard GS, Yang XF, Autieri M, Yang L. Identification of Gm15441, a Txnip antisense lncRNA, as a critical regulator in liver metabolic homeostasis. Cell Biosci 2021; 11:208. [PMID: 34906243 PMCID: PMC8670210 DOI: 10.1186/s13578-021-00722-1] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2021] [Accepted: 11/30/2021] [Indexed: 12/12/2022] Open
Abstract
Background The majority of mammalian genome is composed of non-coding regions, where numerous long non-coding RNAs (lncRNAs) are transcribed. Although lncRNAs have been identified to regulate fundamental biological processes, most of their functions remain unknown, especially in metabolic homeostasis. Analysis of our recent genome-wide screen reveals that Gm15441, a thioredoxin-interacting protein (Txnip) antisense lncRNA, is the most robustly induced lncRNA in the fasting mouse liver. Antisense lncRNAs are known to regulate their sense gene expression. Given that Txnip is a critical metabolic regulator of the liver, we aimed to investigate the role of Gm15441 in the regulation of Txnip and liver metabolism. Methods We examined the response of Gm15441 and Txnip under in vivo metabolic signals such as fasting and refeeding, and in vitro signals such as insulin and key metabolic transcription factors. We investigated the regulation of Txnip expression by Gm15441 and the underlying mechanism in mouse hepatocytes. Using adenovirus-mediated liver-specific overexpression, we determined whether Gm15441 regulates Txnip in the mouse liver and modulates key aspects of liver metabolism. Results We found that the expression levels of Gm15441 and Txnip showed a similar response pattern to metabolic signals in vivo and in vitro, but that their functions were predicted to be opposite. Furthermore, we found that Gm15441 robustly reduced Txnip protein expression in vitro through sequence-specific regulation and translational inhibition. Lastly, we confirmed the Txnip inhibition by Gm15441 in vivo (mice) and found that Gm15441 liver-specific overexpression lowered plasma triglyceride and blood glucose levels and elevated plasma ketone body levels. Conclusions Our data demonstrate that Gm15441 is a potent Txnip inhibitor and a critical metabolic regulator in the liver. This study reveals the therapeutic potential of Gm15441 in treating metabolic diseases. Supplementary Information The online version contains supplementary material available at 10.1186/s13578-021-00722-1.
Collapse
Affiliation(s)
- Mingyang Xin
- Department of Medical Genetics and Molecular Biochemistry, Lewis Katz School of Medicine at Temple University, Philadelphia, PA, 19140, USA
| | - Qian Guo
- Department of Medical Genetics and Molecular Biochemistry, Lewis Katz School of Medicine at Temple University, Philadelphia, PA, 19140, USA
| | - Qingchun Lu
- Department of Medical Genetics and Molecular Biochemistry, Lewis Katz School of Medicine at Temple University, Philadelphia, PA, 19140, USA
| | - Juan Lu
- Department of Medical Genetics and Molecular Biochemistry, Lewis Katz School of Medicine at Temple University, Philadelphia, PA, 19140, USA.,Department of Intensive Care Unit, The First Hospital of Jilin University, Changchun, 130021, China
| | - Po-Shun Wang
- Department of Medical Genetics and Molecular Biochemistry, Lewis Katz School of Medicine at Temple University, Philadelphia, PA, 19140, USA
| | - Yun Dong
- Department of Medical Genetics and Molecular Biochemistry, Lewis Katz School of Medicine at Temple University, Philadelphia, PA, 19140, USA.,Department of Endocrinology, Nanxishan Hospital of Guangxi Zhuang Autonomous Region, Guilin, 541001, China
| | - Tao Li
- Department of Medical Genetics and Molecular Biochemistry, Lewis Katz School of Medicine at Temple University, Philadelphia, PA, 19140, USA.,Department of Infectious diseases, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, 250021, China
| | - Ye Chen
- Department of Mathematics and Statistics, Northern Arizona University, Flagsta, AZ, 86011, USA
| | - Glenn S Gerhard
- Department of Medical Genetics and Molecular Biochemistry, Lewis Katz School of Medicine at Temple University, Philadelphia, PA, 19140, USA
| | - Xiao-Feng Yang
- Center for Metabolic Disease Research, Lewis Katz School of Medicine at Temple University, Philadelphia, PA, 19140, USA
| | - Michael Autieri
- Cardiovascular Research Center, Lewis Katz School of Medicine at Temple University, Philadelphia, PA, 19140, USA
| | - Ling Yang
- Department of Medical Genetics and Molecular Biochemistry, Lewis Katz School of Medicine at Temple University, Philadelphia, PA, 19140, USA.
| |
Collapse
|
36
|
Zhu L, Li Y, Xia F, Xue M, Wang Y, Jia D, Gao Y, Li L, Shi Y, Chen S, Xu G, Yuan C. H19: A vital long noncoding RNA in the treatment of diabetes and diabetic complications. Curr Pharm Des 2021; 28:1011-1018. [PMID: 34895118 DOI: 10.2174/1381612827666211210123959] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2021] [Accepted: 10/29/2021] [Indexed: 11/22/2022]
Abstract
BACKGROUND Increasing academic efforts have been made to explore the correlation of long noncoding RNAs (lncRNAs) with human diseases, particularly metabolic diseases like diabetes mellitus. Taking lncRNA H19 as an example, this review intends to reveal the functions and mechanism of lncRNA H19 in diabetes mellitus and diabetic complications. METHODS The research results associated with lncRNA H19 and diabetes mellitus are collected and summarized on PubMed. CONCLUSION LncRNA H19 is a potential instructive marker for the treatment of diabetes mellitus and diabetic complications.
Collapse
Affiliation(s)
- Leiqi Zhu
- College of Medical Science, China Three Gorges University, Yichang 443002. China
| | - Yuanyang Li
- College of Medical Science, China Three Gorges University, Yichang 443002. China
| | - Fangqi Xia
- College of Medical Science, China Three Gorges University, Yichang 443002. China
| | - Mengzhen Xue
- College of Medical Science, China Three Gorges University, Yichang 443002. China
| | - Yaqi Wang
- College of Medical Science, China Three Gorges University, Yichang 443002. China
| | - Dengke Jia
- College of Medical Science, China Three Gorges University, Yichang 443002. China
| | - Yan Gao
- College of Medical Science, China Three Gorges University, Yichang 443002. China
| | - Luoying Li
- College of Medical Science, China Three Gorges University, Yichang 443002. China
| | - Yue Shi
- College of Medical Science, China Three Gorges University, Yichang 443002. China
| | - Silong Chen
- College of Medical Science, China Three Gorges University, Yichang 443002. China
| | - Guangfu Xu
- College of Medical Science, China Three Gorges University, Yichang 443002. China
| | - Chengfu Yuan
- College of Medical Science, China Three Gorges University, Yichang 443002. China
| |
Collapse
|
37
|
Nettore IC, Franchini F, Palatucci G, Macchia PE, Ungaro P. Epigenetic Mechanisms of Endocrine-Disrupting Chemicals in Obesity. Biomedicines 2021; 9:biomedicines9111716. [PMID: 34829943 PMCID: PMC8615468 DOI: 10.3390/biomedicines9111716] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2021] [Revised: 11/15/2021] [Accepted: 11/16/2021] [Indexed: 02/07/2023] Open
Abstract
The incidence of obesity has dramatically increased over the last decades. Recently, there has been a growing interest in the possible association between the pandemics of obesity and some endocrine-disrupting chemicals (EDCs), termed “obesogens”. These are a heterogeneous group of exogenous compounds that can interfere in the endocrine regulation of energy metabolism and adipose tissue structure. Oral intake, inhalation, and dermal absorption represent the major sources of human exposure to these EDCs. Recently, epigenetic changes such as the methylation of cytosine residues on DNA, post-translational modification of histones, and microRNA expression have been considered to act as an intermediary between deleterious effects of EDCs and obesity development in susceptible individuals. Specifically, EDCs exposure during early-life development can detrimentally affect individuals via inducing epigenetic modifications that can permanently change the epigenome in the germline, enabling changes to be transmitted to the next generations and predisposing them to a multitude of diseases. The purpose of this review is to analyze the epigenetic alterations putatively induced by chemical exposures and their ability to interfere with the control of energy metabolism and adipose tissue regulation, resulting in imbalances in the control of body weight, which can lead to obesity.
Collapse
Affiliation(s)
- Immacolata Cristina Nettore
- Dipartimento di Medicina Clinica e Chirurgia, Università degli Studi di Napoli Federico II, Via S. Pansini, 80131 Naples, Italy; (I.C.N.); (F.F.); (G.P.); (P.E.M.)
| | - Fabiana Franchini
- Dipartimento di Medicina Clinica e Chirurgia, Università degli Studi di Napoli Federico II, Via S. Pansini, 80131 Naples, Italy; (I.C.N.); (F.F.); (G.P.); (P.E.M.)
| | - Giuseppe Palatucci
- Dipartimento di Medicina Clinica e Chirurgia, Università degli Studi di Napoli Federico II, Via S. Pansini, 80131 Naples, Italy; (I.C.N.); (F.F.); (G.P.); (P.E.M.)
| | - Paolo Emidio Macchia
- Dipartimento di Medicina Clinica e Chirurgia, Università degli Studi di Napoli Federico II, Via S. Pansini, 80131 Naples, Italy; (I.C.N.); (F.F.); (G.P.); (P.E.M.)
| | - Paola Ungaro
- Istituto per l’Endocrinologia e l’Oncologia Sperimentale del CNR “G. Salvatore”, Via S. Pansini, 80131 Naples, Italy
- Correspondence: ; Tel.: +39-081-770-4795
| |
Collapse
|
38
|
Rey F, Zuccotti GV, Carelli S. Long non-coding RNAs in metabolic diseases: from bench to bedside. Trends Endocrinol Metab 2021; 32:747-749. [PMID: 34158225 DOI: 10.1016/j.tem.2021.05.009] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/12/2021] [Revised: 05/25/2021] [Accepted: 05/28/2021] [Indexed: 12/30/2022]
Abstract
Long non-coding RNAs (lncRNAs) are being widely studied for their implications in physiological and diseases contexts but their functions and therapeutic potentials in metabolic diseases are far from clarified. Here, we report a summary of current advances in lncRNAs identification, functions, role as biomarkers and therapeutic prospects in metabolic diseases.
Collapse
Affiliation(s)
- Federica Rey
- Department of Biomedical and Clinical Sciences, L. Sacco, University of Milan, Milan, Italy; Pediatric Clinical Research Center, Fondazione Romeo ed Enrica Invernizzi, University of Milan, Milan, Italy
| | - Gian Vincenzo Zuccotti
- Department of Biomedical and Clinical Sciences, L. Sacco, University of Milan, Milan, Italy; Pediatric Clinical Research Center, Fondazione Romeo ed Enrica Invernizzi, University of Milan, Milan, Italy; Department of Pediatrics, Children's Hospital V. Buzzi, Milan, Italy
| | - Stephana Carelli
- Department of Biomedical and Clinical Sciences, L. Sacco, University of Milan, Milan, Italy; Pediatric Clinical Research Center, Fondazione Romeo ed Enrica Invernizzi, University of Milan, Milan, Italy.
| |
Collapse
|
39
|
Ruan X, Li P, Ma Y, Jiang CF, Chen Y, Shi Y, Gupta N, Seifuddin F, Pirooznia M, Ohnishi Y, Yoneda N, Nishiwaki M, Dumbovic G, Rinn JL, Higuchi Y, Kawai K, Suemizu H, Cao H. Identification of human long noncoding RNAs associated with nonalcoholic fatty liver disease and metabolic homeostasis. J Clin Invest 2021; 131:136336. [PMID: 33048844 DOI: 10.1172/jci136336] [Citation(s) in RCA: 29] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2020] [Accepted: 10/09/2020] [Indexed: 12/11/2022] Open
Abstract
A growing number of long noncoding RNAs (lncRNAs) have emerged as vital metabolic regulators. However, most human lncRNAs are nonconserved and highly tissue specific, vastly limiting our ability to identify human lncRNA metabolic regulators (hLMRs). In this study, we established a pipeline to identify putative hLMRs that are metabolically sensitive, disease relevant, and population applicable. We first progressively processed multilevel human transcriptome data to select liver lncRNAs that exhibit highly dynamic expression in the general population, show differential expression in a nonalcoholic fatty liver disease (NAFLD) population, and respond to dietary intervention in a small NAFLD cohort. We then experimentally demonstrated the responsiveness of selected hepatic lncRNAs to defined metabolic milieus in a liver-specific humanized mouse model. Furthermore, by extracting a concise list of protein-coding genes that are persistently correlated with lncRNAs in general and NAFLD populations, we predicted the specific function for each hLMR. Using gain- and loss-of-function approaches in humanized mice as well as ectopic expression in conventional mice, we validated the regulatory role of one nonconserved hLMR in cholesterol metabolism by coordinating with an RNA-binding protein, PTBP1, to modulate the transcription of cholesterol synthesis genes. Our work overcame the heterogeneity intrinsic to human data to enable the efficient identification and functional definition of disease-relevant human lncRNAs in metabolic homeostasis.
Collapse
Affiliation(s)
- Xiangbo Ruan
- Cardiovascular Branch, National Heart, Lung, and Blood Institute, NIH, Bethesda, Maryland, USA
| | - Ping Li
- Cardiovascular Branch, National Heart, Lung, and Blood Institute, NIH, Bethesda, Maryland, USA
| | - Yonghe Ma
- Cardiovascular Branch, National Heart, Lung, and Blood Institute, NIH, Bethesda, Maryland, USA
| | - Cheng-Fei Jiang
- Cardiovascular Branch, National Heart, Lung, and Blood Institute, NIH, Bethesda, Maryland, USA
| | - Yi Chen
- Cardiovascular Branch, National Heart, Lung, and Blood Institute, NIH, Bethesda, Maryland, USA
| | - Yu Shi
- Cardiovascular Branch, National Heart, Lung, and Blood Institute, NIH, Bethesda, Maryland, USA
| | - Nikhil Gupta
- Cardiovascular Branch, National Heart, Lung, and Blood Institute, NIH, Bethesda, Maryland, USA
| | - Fayaz Seifuddin
- Bioinformatics and Computational Biology, National Heart, Lung, and Blood Institute, NIH, Bethesda, Maryland, USA
| | - Mehdi Pirooznia
- Bioinformatics and Computational Biology, National Heart, Lung, and Blood Institute, NIH, Bethesda, Maryland, USA
| | - Yasuyuki Ohnishi
- Laboratory Animal Research Department, Biomedical Research Laboratory, Central Institute for Experimental Animals, Kawasaki, Japan
| | - Nao Yoneda
- Laboratory Animal Research Department, Biomedical Research Laboratory, Central Institute for Experimental Animals, Kawasaki, Japan
| | - Megumi Nishiwaki
- Laboratory Animal Research Department, Biomedical Research Laboratory, Central Institute for Experimental Animals, Kawasaki, Japan.,Technical Service Department, CLEA Japan Inc., Shizuoka, Japan
| | - Gabrijela Dumbovic
- Department of Biochemistry and BioFrontiers, University of Colorado Boulder, Boulder, Colorado, USA
| | - John L Rinn
- Department of Biochemistry and BioFrontiers, University of Colorado Boulder, Boulder, Colorado, USA
| | - Yuichiro Higuchi
- Laboratory Animal Research Department, Biomedical Research Laboratory, Central Institute for Experimental Animals, Kawasaki, Japan
| | - Kenji Kawai
- Department Pathology Analysis Center, Central Institute for Experimental Animals, Kawasaki, Japan
| | - Hiroshi Suemizu
- Laboratory Animal Research Department, Biomedical Research Laboratory, Central Institute for Experimental Animals, Kawasaki, Japan
| | - Haiming Cao
- Cardiovascular Branch, National Heart, Lung, and Blood Institute, NIH, Bethesda, Maryland, USA
| |
Collapse
|
40
|
Di Mauro S, Salomone F, Scamporrino A, Filippello A, Morisco F, Guido M, Lembo V, Cossiga V, Pipitone RM, Grimaudo S, Malaguarnera R, Purrello F, Piro S. Coffee Restores Expression of lncRNAs Involved in Steatosis and Fibrosis in a Mouse Model of NAFLD. Nutrients 2021; 13:nu13092952. [PMID: 34578828 PMCID: PMC8467439 DOI: 10.3390/nu13092952] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2021] [Revised: 08/20/2021] [Accepted: 08/22/2021] [Indexed: 12/31/2022] Open
Abstract
Background and aim: Coffee intake exerts protective effects against non-alcoholic fatty liver disease (NAFLD), although without fully cleared mechanisms. In this study we aimed to assess whether coffee consumption may influence the expression of long non-coding RNAs (lncRNAs) in the liver. Methods: C57BL/6J mice were fed a 12-week standard diet (SD), high-fat diet (HFD) or HFD plus decaffeinated coffee solution (HFD + coffee). Expression of specific lncRNAs involved in NAFLD was analyzed by real-time PCR. For the most differentially expressed lncRNAs, the analysis was also extended to their mRNA targets. Results: Decaffeinated coffee intake reduced body weight gain, prevented NAFLD, lowered hyperglycemia and hypercholesterolemia. NAFLD was associated with lower hepatic expression of Gm16551, a lncRNA inhibiting de novo lipogenesis, and higher expression of H19, a lncRNA promoting fibrogenesis. Coffee intake restored Gm16551 to levels observed in lean mice and downregulated gene expression of its targets acetyl coenzyme A carboxylase 1 and stearoyl coenzyme A desaturase 1. Furthermore, coffee consumption markedly decreased hepatic expression of H19 and of its target gene collagen alpha-1(I) chain; consistently, in mice fed HFD + coffee liver expression of αSMA protein returned to levels of mice fed SD. Expression of lncRNA involved in circadian clock such as fatty liver-related lncRNA 1 (FLRL1) and fatty liver-related lncRNA 2 (FLRL2) were upregulated by HFD and were also modulated by coffee intake. Conclusion. Hepatoprotective effects of coffee may be depending on the modulation of lncRNAs involved in key pathways of NAFLD onset and progression.
Collapse
Affiliation(s)
- Stefania Di Mauro
- Department of Clinical and Experimental Medicine, University of Catania, 95122 Catania, Italy; (S.D.M.); (A.S.); (A.F.); (S.P.)
| | - Federico Salomone
- Division of Gastroenterology, Ospedale di Acireale, Azienda Sanitaria Provinciale di Catania, 95024 Catania, Italy;
| | - Alessandra Scamporrino
- Department of Clinical and Experimental Medicine, University of Catania, 95122 Catania, Italy; (S.D.M.); (A.S.); (A.F.); (S.P.)
| | - Agnese Filippello
- Department of Clinical and Experimental Medicine, University of Catania, 95122 Catania, Italy; (S.D.M.); (A.S.); (A.F.); (S.P.)
| | - Filomena Morisco
- Department of Clinical Medicine and Surgery, University of Naples “Federico II”, 80125 Naples, Italy; (F.M.); (V.L.); (V.C.)
| | - Maria Guido
- Department of Medicine, University of Padua, 35121 Padua, Italy;
| | - Vincenzo Lembo
- Department of Clinical Medicine and Surgery, University of Naples “Federico II”, 80125 Naples, Italy; (F.M.); (V.L.); (V.C.)
| | - Valentina Cossiga
- Department of Clinical Medicine and Surgery, University of Naples “Federico II”, 80125 Naples, Italy; (F.M.); (V.L.); (V.C.)
| | | | - Stefania Grimaudo
- Department PROMISE, University of Palermo, 90128 Palermo, Italy; (R.M.P.); (S.G.)
| | | | - Francesco Purrello
- Department of Clinical and Experimental Medicine, University of Catania, 95122 Catania, Italy; (S.D.M.); (A.S.); (A.F.); (S.P.)
- Correspondence: ; Tel.: +39-095-759-8401
| | - Salvatore Piro
- Department of Clinical and Experimental Medicine, University of Catania, 95122 Catania, Italy; (S.D.M.); (A.S.); (A.F.); (S.P.)
| |
Collapse
|
41
|
Waterlogging-Stress-Responsive LncRNAs, Their Regulatory Relationships with miRNAs and Target Genes in Cucumber ( Cucumis sativus L.). Int J Mol Sci 2021; 22:ijms22158197. [PMID: 34360961 PMCID: PMC8348067 DOI: 10.3390/ijms22158197] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2021] [Revised: 07/19/2021] [Accepted: 07/23/2021] [Indexed: 12/13/2022] Open
Abstract
Low oxygen level is a phenomenon often occurring during the cucumber cultivation period. Genes involved in adaptations to stress can be regulated by non-coding RNA. The aim was the identification of long non-coding RNAs (lncRNAs) involved in the response to long-term waterlogging stress in two cucumber haploid lines, i.e., DH2 (waterlogging tolerant—WL-T) and DH4 (waterlogging sensitive—WL-S). Plants, at the juvenile stage, were waterlogged for 7 days (non-primed, 1xH), and after a 14-day recovery period, plants were stressed again for another 7 days (primed, 2xH). Roots were collected for high-throughput RNA sequencing. Implementation of the bioinformatic pipeline made it possible to determine specific lncRNAs for non-primed and primed plants of both accessions, highlighting differential responses to hypoxia stress. In total, 3738 lncRNA molecules were identified. The highest number (1476) of unique lncRNAs was determined for non-primed WL-S plants. Seventy-one lncRNAs were depicted as potentially being involved in acquiring tolerance to hypoxia in cucumber. Understanding the mechanism of gene regulation under long-term waterlogging by lncRNAs and their interactions with miRNAs provides sufficient information in terms of adaptation to the oxygen deprivation in cucumber. To the best of our knowledge, this is the first report concerning the role of lncRNAs in the regulation of long-term waterlogging tolerance by priming application in cucumber.
Collapse
|
42
|
Chi K, Geng X, Liu C, Zhang Y, Cui J, Cai G, Chen X, Wang F, Hong Q. LncRNA-HOTAIR promotes endothelial cell pyroptosis by regulating the miR-22/NLRP3 axis in hyperuricaemia. J Cell Mol Med 2021; 25:8504-8521. [PMID: 34296520 PMCID: PMC8419175 DOI: 10.1111/jcmm.16812] [Citation(s) in RCA: 30] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2021] [Revised: 07/06/2021] [Accepted: 07/09/2021] [Indexed: 12/20/2022] Open
Abstract
Long non‐coding RNA (lncRNA) plays an important role in the renal inflammatory response caused by hyperuricaemia. However, the underlying molecular mechanisms through which lncRNA is involved in endothelial injury induced by hyperuricaemia remain unclear. In this study, we investigated the regulatory role of lncRNA‐HOTAIR in high concentration of uric acid (HUA)–induced renal injury. We established hyperuricaemia mouse model and an in vitro uric acid (UA)–induced human umbilical vein endothelial cell (HUVEC) injury model. In HUA‐treated HUVECs and hyperuricaemia mice, we observed increased HOTAIR and decreased miR‐22 expression. The expression of pyroptosis‐associated protein (NLRP3, Caspase‐1, GSDMD‐N, GSDMD‐FL) was increased. The release of LDH, IL‐1β and IL‐18 in cell supernatants and the sera of model mice was also increased. The proliferation of HUVECs stimulated by HUA was significantly inhibited, and the number of TUNEL‐positive cells in hyperuricaemia mouse kidney was increased. Bioinformatics analysis and luciferase reporter and RIP assays confirmed that HOTAIR promoted NLRP3 inflammasome activation by competitively binding miR‐22. In gain‐ or loss‐of‐function experiments, we found that HOTAIR and NLRP3 overexpression or miR‐22 knock down activated the NLRP3 inflammasome and promoted pyroptosis in HUA‐treated HUVECs, while NLRP3 and HOTAIR knockdown or a miR‐22 mimic exerted the opposite effects. Furthermore, in vivo experiments validated that HOTAIR knockdown alleviated renal inflammation in hyperuricaemia mice. In conclusion, we demonstrated that in hyperuricaemia, lncRNA‐HOTAIR promotes endothelial cell pyroptosis by competitively binding miR‐22 to regulate NLRP3 expression.
Collapse
Affiliation(s)
- Kun Chi
- Department of Nephrology, Chinese PLA General Hospital, Medical School of Chinese PLA, Chinese PLA Institute of Nephrology, State Key Laboratory of Kidney Diseases, National Clinical Research Center for Kidney Diseases, Beijing Key Laboratory of Kidney Diseases, Beijing, China
| | - Xiaodong Geng
- Department of Nephrology, Chinese PLA General Hospital, Medical School of Chinese PLA, Chinese PLA Institute of Nephrology, State Key Laboratory of Kidney Diseases, National Clinical Research Center for Kidney Diseases, Beijing Key Laboratory of Kidney Diseases, Beijing, China.,Beidaihe Rehabilitation and Recuperation Center, Chinese People's Liberation Army Joint Logistics Support Force Qinhuangdao, Qinhuangdao, China
| | - Chao Liu
- Department of Nephrology, Chinese PLA General Hospital, Medical School of Chinese PLA, Chinese PLA Institute of Nephrology, State Key Laboratory of Kidney Diseases, National Clinical Research Center for Kidney Diseases, Beijing Key Laboratory of Kidney Diseases, Beijing, China
| | - Yang Zhang
- Department of Nephrology, Chinese PLA General Hospital, Medical School of Chinese PLA, Chinese PLA Institute of Nephrology, State Key Laboratory of Kidney Diseases, National Clinical Research Center for Kidney Diseases, Beijing Key Laboratory of Kidney Diseases, Beijing, China
| | - Jie Cui
- Beidaihe Rehabilitation and Recuperation Center, Chinese People's Liberation Army Joint Logistics Support Force Qinhuangdao, Qinhuangdao, China
| | - GuangYan Cai
- Department of Nephrology, Chinese PLA General Hospital, Medical School of Chinese PLA, Chinese PLA Institute of Nephrology, State Key Laboratory of Kidney Diseases, National Clinical Research Center for Kidney Diseases, Beijing Key Laboratory of Kidney Diseases, Beijing, China
| | - Xiangmei Chen
- Department of Nephrology, Chinese PLA General Hospital, Medical School of Chinese PLA, Chinese PLA Institute of Nephrology, State Key Laboratory of Kidney Diseases, National Clinical Research Center for Kidney Diseases, Beijing Key Laboratory of Kidney Diseases, Beijing, China
| | - Fangfang Wang
- Department of Cardiology and Institute of Vascular Medicine, NHC Key Laboratory of Cardiovascular Molecular Biology and Regulatory Peptides, Key Laboratory of Molecular Cardiovascular Science, Ministry of Education, Beijing Key Laboratory of Cardiovascular Receptors Research., Peking University Third Hospital, Beijing, 100191, China
| | - Quan Hong
- Department of Nephrology, Chinese PLA General Hospital, Medical School of Chinese PLA, Chinese PLA Institute of Nephrology, State Key Laboratory of Kidney Diseases, National Clinical Research Center for Kidney Diseases, Beijing Key Laboratory of Kidney Diseases, Beijing, China
| |
Collapse
|
43
|
Abstract
BACKGROUND Long non-coding RNAs (lncRNAs) have evolved as a critical regulatory mechanism for almost all biological processes. By dynamically interacting with their molecular partners, lncRNAs regulate gene activity at multiple levels ranging from transcription, pre-mRNA splicing, RNA transporting, RNA decay, and translation of mRNA. RESULTS AND CONCLUSIONS Dysregulation of lncRNAs has been associated with human diseases, including cancer, neurodegenerative, and cardiometabolic diseases. However, as lncRNAs are usually much less conserved than mRNAs at the sequence level, most human lncRNAs are either primate or human specific. The pathophysiological significance of human lncRNAs is still mostly unclear due to the persistent limitations in studying human-specific genes. This review will focus on recent discoveries showing human lncRNAs' roles in regulating metabolic homeostasis and the potential of targeting this unique group of genes for treatment of cardiometabolic diseases.
Collapse
Affiliation(s)
- Xiangbo Ruan
- Cardiovascular Branch, National Heart, Lung and Blood Institute, National Institutes of Health, Bethesda, MD, 20892, USA.
| |
Collapse
|
44
|
Systematic Analysis of Long Noncoding RNA and mRNA in Granulosa Cells during the Hen Ovulatory Cycle. Animals (Basel) 2021; 11:ani11061533. [PMID: 34070248 PMCID: PMC8225051 DOI: 10.3390/ani11061533] [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: 05/07/2021] [Revised: 05/19/2021] [Accepted: 05/20/2021] [Indexed: 12/04/2022] Open
Abstract
Simple Summary Chicken is one of the most economically important farm poultry, and providing many food products, such as meat and eggs for human consumption. However, follicle transcriptome studies in chickens with timepoints relating to changes in luteinizing hormone level remain unknown. In this study, the largest preovulatory follicle of chicken underwent the early, middle, and terminal stages of ovulatory cycle. Our work provides a comprehensive analysis of lncRNAs and mRNAs in chicken granulosa cells during the ovulatory cycle. A total of 12,479 mRNAs and 7528 lncRNAs were identified among the three stages. Thousands of lncRNAs were annotated, and the most differentially abundant genes were detected in the luteinizing hormone surge stage. Functional features of the lncRNAs and mRNAs at each stage were revealed, which was also associated with the changes in serum luteinizing hormone level. Especially, genes related to oxidative stress, steroids regulation, and inflammatory process were enriched in the luteinizing hormone surge stage, The comprehensive data generated in this study provides the foundation for future investigations to improve the reproductive performance of chickens and explore the mechanisms responsible for female ovarian diseases. Abstract Long non-coding RNAs (lncRNAs) and mRNAs are temporally expressed during chicken follicle development. However, follicle transcriptome studies in chickens with timepoints relating to changes in luteinizing hormone (LH) levels are rare. In this study, gene expression in Rohman layers was investigated at three distinct stages of the ovulatory cycle: zeitgeber time 0 (ZT0, 9:00 a.m.), zeitgeber time 12 (ZT12, 9:00 p.m.), and zeitgeber time 20 (ZT20, 5:00 a.m.) representing the early, middle, and LH surge stages, respectively, of the ovulatory cycle. Gene expression profiles were explored during follicle development at ZT0, ZT12, and ZT20 using Ribo-Zero RNA sequencing. The three stages were separated into two major stages, including the pre-LH surge and the LH surge stages. A total of 12,479 mRNAs and 7528 lncRNAs were identified among the three stages, and 4531, 523 differentially expressed genes (DEGs) and 2367, 211 differentially expressed lncRNAs (DELs) were identified in the ZT20 vs. ZT12, and ZT12 vs. ZT0, comparisons. Functional enrichment analysis revealed that genes involved in cell proliferation and metabolism processes (lipid-related) were mainly enriched in the ZT0 and ZT12 stages, respectively, and genes related to oxidative stress, steroids regulation, and inflammatory process were enriched in the ZT20 stage. These findings provide the basis for further investigation of the specific genetic and molecular functions of follicle development in chickens.
Collapse
|
45
|
Batista TM, Haider N, Kahn CR. Defining the underlying defect in insulin action in type 2 diabetes. Diabetologia 2021; 64:994-1006. [PMID: 33730188 PMCID: PMC8916220 DOI: 10.1007/s00125-021-05415-5] [Citation(s) in RCA: 92] [Impact Index Per Article: 23.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/29/2020] [Accepted: 01/29/2021] [Indexed: 01/08/2023]
Abstract
Insulin resistance is one of the earliest defects in the pathogenesis of type 2 diabetes. Over the past 50 years, elucidation of the insulin signalling network has provided important mechanistic insights into the abnormalities of glucose, lipid and protein metabolism that underlie insulin resistance. In classical target tissues (liver, muscle and adipose tissue), insulin binding to its receptor initiates a broad signalling cascade mediated by changes in phosphorylation, gene expression and vesicular trafficking that result in increased nutrient utilisation and storage, and suppression of catabolic processes. Insulin receptors are also expressed in non-classical targets, such as the brain and endothelial cells, where it helps regulate appetite, energy expenditure, reproductive hormones, mood/behaviour and vascular function. Recent progress in cell biology and unbiased molecular profiling by mass spectrometry and DNA/RNA-sequencing has provided a unique opportunity to dissect the determinants of insulin resistance in type 2 diabetes and the metabolic syndrome; best studied are extrinsic factors, such as circulating lipids, amino acids and other metabolites and exosomal microRNAs. More challenging has been defining the cell-intrinsic factors programmed by genetics and epigenetics that underlie insulin resistance. In this regard, studies using human induced pluripotent stem cells and tissues point to cell-autonomous alterations in signalling super-networks, involving changes in phosphorylation and gene expression both inside and outside the canonical insulin signalling pathway. Understanding how these multi-layered molecular networks modulate insulin action and metabolism in different tissues will open new avenues for therapy and prevention of type 2 diabetes and its associated pathologies.
Collapse
Affiliation(s)
- Thiago M Batista
- Section of Integrative Physiology and Metabolism, Joslin Diabetes Center, Harvard Medical School, Boston, MA, USA
| | - Nida Haider
- Section of Integrative Physiology and Metabolism, Joslin Diabetes Center, Harvard Medical School, Boston, MA, USA
| | - C Ronald Kahn
- Section of Integrative Physiology and Metabolism, Joslin Diabetes Center, Harvard Medical School, Boston, MA, USA.
| |
Collapse
|
46
|
Aurilia C, Donati S, Palmini G, Miglietta F, Iantomasi T, Brandi ML. The Involvement of Long Non-Coding RNAs in Bone. Int J Mol Sci 2021; 22:ijms22083909. [PMID: 33920083 PMCID: PMC8069547 DOI: 10.3390/ijms22083909] [Citation(s) in RCA: 30] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2021] [Revised: 04/02/2021] [Accepted: 04/08/2021] [Indexed: 12/18/2022] Open
Abstract
A harmonious balance between osteoblast and osteoclast activity guarantees optimal bone formation and resorption, pathological conditions affecting the bone may arise. In recent years, emerging evidence has shown that epigenetic mechanisms play an important role during osteoblastogenesis and osteoclastogenesis processes, including long non-coding RNAs (lncRNAs). These molecules are a class of ncRNAs with lengths exceeding 200 nucleotides not translated into protein, that have attracted the attention of the scientific community as potential biomarkers to use for the future development of novel diagnostic and therapeutic approaches for several pathologies, including bone diseases. This review aims to provide an overview of the lncRNAs and their possible molecular mechanisms in the osteoblastogenesis and osteoclastogenesis processes. The deregulation of their expression profiles in common diseases associated with an altered bone turnover is also described. In perspective, lncRNAs could be considered potential innovative molecular biomarkers to help with earlier diagnosis of bone metabolism-related disorders and for the development of new therapeutic strategies.
Collapse
Affiliation(s)
- Cinzia Aurilia
- Department of Experimental and Clinical Biomedical Sciences, University of Florence, 50134 Florence, Italy; (C.A.); (S.D.); (G.P.); (F.M.); (T.I.)
| | - Simone Donati
- Department of Experimental and Clinical Biomedical Sciences, University of Florence, 50134 Florence, Italy; (C.A.); (S.D.); (G.P.); (F.M.); (T.I.)
| | - Gaia Palmini
- Department of Experimental and Clinical Biomedical Sciences, University of Florence, 50134 Florence, Italy; (C.A.); (S.D.); (G.P.); (F.M.); (T.I.)
| | - Francesca Miglietta
- Department of Experimental and Clinical Biomedical Sciences, University of Florence, 50134 Florence, Italy; (C.A.); (S.D.); (G.P.); (F.M.); (T.I.)
| | - Teresa Iantomasi
- Department of Experimental and Clinical Biomedical Sciences, University of Florence, 50134 Florence, Italy; (C.A.); (S.D.); (G.P.); (F.M.); (T.I.)
| | - Maria Luisa Brandi
- Department of Experimental and Clinical Biomedical Sciences, University of Florence, 50134 Florence, Italy; (C.A.); (S.D.); (G.P.); (F.M.); (T.I.)
- Fondazione Italiana Ricerca sulle Malattie dell’Osso (FIRMO Onlus), 50141 Florence, Italy
- Correspondence:
| |
Collapse
|
47
|
Ma Y, Harris J, Li P, Cao H. Long noncoding RNAs-a new dimension in the molecular architecture of the bile acid/FXR pathway. Mol Cell Endocrinol 2021; 525:111191. [PMID: 33539963 PMCID: PMC8437140 DOI: 10.1016/j.mce.2021.111191] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/22/2020] [Revised: 01/25/2021] [Accepted: 01/28/2021] [Indexed: 11/15/2022]
Abstract
Bile acids, regarded as the body's detergent for digesting lipids, also function as critical signaling molecules that regulate cholesterol and triglyceride levels in the body. Bile acids are the natural ligands of the nuclear receptor, FXR, which controls an intricate network of cellular pathways to maintain metabolic homeostasis. In recent years, growing evidence supports that many cellular actions of the bile acid/FXR pathway are mediated by long non-coding RNAs (lncRNAs), and lncRNAs are in turn powerful regulators of bile acid levels and FXR activities. In this review, we highlight the substantial progress made in the understanding of the functional and mechanistic role of lncRNAs in bile acid metabolism and how lncRNAs connect bile acid activity to additional metabolic processes. We also discuss the potential of lncRNA studies in elucidating novel molecular mechanisms of the bile acid/FXR pathway and the promise of lncRNAs as potential diagnostic markers and therapeutic targets for diseases associated with altered bile acid metabolism.
Collapse
Affiliation(s)
- Yonghe Ma
- Cardiovascular Branch, National Heart, Lung and Blood Institute, National Institutes of Health, Bethesda, MD, 20892, USA
| | - Jamie Harris
- Cardiovascular Branch, National Heart, Lung and Blood Institute, National Institutes of Health, Bethesda, MD, 20892, USA
| | - Ping Li
- Cardiovascular Branch, National Heart, Lung and Blood Institute, National Institutes of Health, Bethesda, MD, 20892, USA
| | - Haiming Cao
- Cardiovascular Branch, National Heart, Lung and Blood Institute, National Institutes of Health, Bethesda, MD, 20892, USA.
| |
Collapse
|
48
|
Wang Y, Mao Y, Zhao Y, Yi X, Ding G, Yu C, Sheng J, Liu X, Meng Y, Huang H. Early-life undernutrition induces enhancer RNA remodeling in mice liver. Epigenetics Chromatin 2021; 14:18. [PMID: 33789751 PMCID: PMC8011416 DOI: 10.1186/s13072-021-00392-w] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2020] [Accepted: 03/19/2021] [Indexed: 01/10/2023] Open
Abstract
Background Maternal protein restriction diet (PRD) increases the risk of metabolic dysfunction in adulthood, the mechanisms during the early life of offspring are still poorly understood. Apart from genetic factors, epigenetic mechanisms are crucial to offer phenotypic plasticity in response to environmental situations and transmission. Enhancer-associated noncoding RNAs (eRNAs) transcription serves as a robust indicator of enhancer activation, and have potential roles in mediating enhancer functions and gene transcription. Results Using global run-on sequencing (GRO-seq) of nascent RNA including eRNA and total RNA sequencing data, we show that early-life undernutrition causes remodeling of enhancer activity in mouse liver. Differentially expressed nascent active genes were enriched in metabolic pathways. Besides, our work detected a large number of high confidence enhancers based on eRNA transcription at the ages of 4 weeks and 7 weeks, respectively. Importantly, except for ~ 1000 remodeling enhancers, the early-life undernutrition induced instability of enhancer activity which decreased in 4 weeks and increased in adulthood. eRNA transcription mainly contributes to the regulation of some important metabolic enzymes, suggesting a link between metabolic dysfunction and enhancer transcriptional control. We discovered a novel eRNA that is positively correlated to the expression of circadian gene Cry1 with increased binding of epigenetic cofactor p300. Conclusions Our study reveals novel insights into mechanisms of metabolic dysfunction. Enhancer activity in early life acts on metabolism-associated genes, leading to the increased susceptibility of metabolic disorders. Supplementary Information The online version contains supplementary material available at 10.1186/s13072-021-00392-w.
Collapse
Affiliation(s)
- Yinyu Wang
- The International Peace Maternity and Child Health Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Yiting Mao
- The International Peace Maternity and Child Health Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Yiran Zhao
- The International Peace Maternity and Child Health Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Xianfu Yi
- School of Biomedical Engineering and Technology, Tianjin Medical University, Tianjin, China
| | - Guolian Ding
- The International Peace Maternity and Child Health Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China.,Shanghai Key Laboratory of Embryo Original Disease, Shanghai, China.,Institute of Embryo-Fetal Original Adult Disease Affiliated To Shanghai, Jiao Tong University School of Medicine, Shanghai, China
| | - Chuanjin Yu
- The International Peace Maternity and Child Health Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China.,Shanghai Key Laboratory of Embryo Original Disease, Shanghai, China.,Institute of Embryo-Fetal Original Adult Disease Affiliated To Shanghai, Jiao Tong University School of Medicine, Shanghai, China
| | - Jianzhong Sheng
- Shanghai Key Laboratory of Embryo Original Disease, Shanghai, China.,Department of Pathology and Pathophysiology, School of Medicine, Zhejiang University, Hangzhou, China
| | - Xinmei Liu
- The International Peace Maternity and Child Health Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China.,Shanghai Key Laboratory of Embryo Original Disease, Shanghai, China.,Institute of Embryo-Fetal Original Adult Disease Affiliated To Shanghai, Jiao Tong University School of Medicine, Shanghai, China
| | - Yicong Meng
- The International Peace Maternity and Child Health Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China. .,Shanghai Key Laboratory of Embryo Original Disease, Shanghai, China. .,Institute of Embryo-Fetal Original Adult Disease Affiliated To Shanghai, Jiao Tong University School of Medicine, Shanghai, China.
| | - Hefeng Huang
- The International Peace Maternity and Child Health Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China. .,Shanghai Key Laboratory of Embryo Original Disease, Shanghai, China. .,Institute of Embryo-Fetal Original Adult Disease Affiliated To Shanghai, Jiao Tong University School of Medicine, Shanghai, China.
| |
Collapse
|
49
|
Ma Y, Jiang CF, Li P, Cao H. In Vivo Functional Analysis of Nonconserved Human lncRNAs Using a Humanized Mouse Model. Methods Mol Biol 2021; 2254:339-347. [PMID: 33326086 DOI: 10.1007/978-1-0716-1158-6_21] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
LncRNAs (long noncoding RNAs) are transcripts that are at least 200 nucleotides long and lack any predicted coding potential. Whereas significant progress has been made in deciphering the function of mouse lncRNAs, critical gaps remain in understanding how human lncRNAs exercise their function in a physiological context. As most human lncRNAs are currently considered nonconserved and often do not have homologs in mouse, the technical bottleneck is the lack of a suitable model to study the physiological function. Chimeric mice with repopulated human hepatocytes have emerged as promising tools to study human-specific, liver enriched lncRNAs. Among all liver-specific humanized mouse models, TK-NOG is relatively easy to prepare and holds a higher repopulation rate for a prolonged period of time. In this chapter, we will illustrate how to establish humanized TK-NOG mice for in vivo analysis of human lncRNAs in detail.
Collapse
Affiliation(s)
- Yonghe Ma
- Cardiovascular Branch, National Heart, Lung and Blood Institute, National Institutes of Health, Bethesda, MD, USA
| | - Cheng-Fei Jiang
- Cardiovascular Branch, National Heart, Lung and Blood Institute, National Institutes of Health, Bethesda, MD, USA
| | - Ping Li
- Cardiovascular Branch, National Heart, Lung and Blood Institute, National Institutes of Health, Bethesda, MD, USA
| | - Haiming Cao
- Cardiovascular Branch, National Heart, Lung and Blood Institute, National Institutes of Health, Bethesda, MD, USA.
| |
Collapse
|
50
|
Identification of Accessible Hepatic Gene Signatures for Interindividual Variations in Nutrigenomic Response to Dietary Supplementation of Omega-3 Fatty Acids. Cells 2021; 10:cells10020467. [PMID: 33671567 PMCID: PMC7926558 DOI: 10.3390/cells10020467] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2021] [Revised: 02/15/2021] [Accepted: 02/17/2021] [Indexed: 12/29/2022] Open
Abstract
Dietary supplementation is a widely adapted strategy to maintain nutritional balance for improving health and preventing chronic diseases. Conflicting results in studies of similar design, however, suggest that there is substantial heterogenicity in individuals’ responses to nutrients, and personalized nutrition is required to achieve the maximum benefit of dietary supplementation. In recent years, nutrigenomics studies have been increasingly utilized to characterize the detailed genomic response to a specific nutrient, but it remains a daunting task to define the signatures responsible for interindividual variations to dietary supplements for tissues with limited accessibility. In this work, we used the hepatic response to omega-3 fatty acids as an example to probe such signatures. Through comprehensive analysis of nutrigenomic response to eicosapentaneoid acid (EPA) and/or docosahexaenoic acid (DHA) including both protein coding and long noncoding RNA (lncRNA) genes in human hepatocytes, we defined the EPA- and/or DHA-specific signature genes in hepatocytes. By analyzing gene expression variations in livers of healthy and relevant disease populations, we identified a set of protein coding and lncRNA signature genes whose responses to omega-3 fatty acid exhibit very high interindividual variabilities. The large variabilities of individual responses to omega-3 fatty acids were further validated in human hepatocytes from ten different donors. Finally, we profiled RNAs in exosomes isolated from the circulation of a liver-specific humanized mouse model, in which the humanized liver is the sole source of human RNAs, and confirmed the in vivo detectability of some signature genes, supporting their potential as biomarkers for nutrient response. Taken together, we have developed an efficient and practical procedure to identify nutrient-responsive gene signatures as well as accessible biomarkers for interindividual variations.
Collapse
|