1
|
Hu C, Yang M, Feng X, Wang S, Ma Y, Ma Y. miR-10167-3p targets TCF7L1 to inhibit bovine adipocyte differentiation and promote bovine adipocyte proliferation. Genomics 2024; 116:110903. [PMID: 39069233 DOI: 10.1016/j.ygeno.2024.110903] [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: 03/19/2024] [Revised: 07/09/2024] [Accepted: 07/24/2024] [Indexed: 07/30/2024]
Abstract
MicroRNAs (miRNAs) are widely involved in various lipogenic processes, including adipocyte proliferation and differentiation, lipid droplet formation, and adipocyte-specific gene activation. The present study aimed to investigate the gene expression profiles of bovine preadipocytes under high miR-10167-3p expression using the RNA-seq technique and to verify the functions of its downstream target genes on the proliferation and differentiation of bovine preadipocytes. First, RNA-seq identified 573 differentially expressed genes (DEGs), of which 243 were downregulated and 330 were upregulated. Then, Kyoto Encyclopedia of Genes and Genomes (KEGG) analysis revealed that 15.19% of the DEGs were enriched in pathways related to lipid metabolism. Meanwhile, dual-luciferase reporter gene assay verified the target-binding relationship between miR-10167-3p and TCF7L1. The function of TCF7L1 was assessed using several experiments in adipocytes with high TCF7L1 expression and RNA interference. The mRNA and protein expression of proliferation, differentiation, and apoptosis marker genes were detected using qPCR and western blot, respectively; lipid droplet synthesis was detected using oil red O, Nile red, and bodipy staining; adipocyte proliferation was detected by EdU; and apoptosis was detected using flow cytometry. The results revealed that TCF7L1 overexpression inhibited bovine preadipocyte differentiation and apoptosis and promoted their proliferation, with opposite results obtained with its RNA interference. These results may provide a reference for the subsequent investigation of the molecular mechanism of bovine fat deposition.
Collapse
Affiliation(s)
- Chunli Hu
- Key Laboratory of Ruminant Molecular Cell Breeding, Ningxia Hui Autonomous Region, College of Animal Science and Technology, Ningxia University, Yinchuan 750021, China
| | - Mengli Yang
- Key Laboratory of Ruminant Molecular Cell Breeding, Ningxia Hui Autonomous Region, College of Animal Science and Technology, Ningxia University, Yinchuan 750021, China
| | - Xue Feng
- Key Laboratory of Ruminant Molecular Cell Breeding, Ningxia Hui Autonomous Region, College of Animal Science and Technology, Ningxia University, Yinchuan 750021, China
| | - Shuzhe Wang
- Key Laboratory of Ruminant Molecular Cell Breeding, Ningxia Hui Autonomous Region, College of Animal Science and Technology, Ningxia University, Yinchuan 750021, China
| | - Yanfen Ma
- Key Laboratory of Ruminant Molecular Cell Breeding, Ningxia Hui Autonomous Region, College of Animal Science and Technology, Ningxia University, Yinchuan 750021, China.
| | - Yun Ma
- Key Laboratory of Ruminant Molecular Cell Breeding, Ningxia Hui Autonomous Region, College of Animal Science and Technology, Ningxia University, Yinchuan 750021, China.
| |
Collapse
|
2
|
Smith L, Santiago EG, Eke C, Gu W, Wang W, Llivichuzhca-Loja D, Kehoe T, St Denis K, Strine M, Taylor S, Tseng G, Konnikova L. Human Milk Supports Robust Intestinal Organoid Growth, Differentiation, and Homeostatic Cytokine Production. GASTRO HEP ADVANCES 2024; 3:1030-1042. [PMID: 39529649 PMCID: PMC11550179 DOI: 10.1016/j.gastha.2024.07.007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/08/2024] [Accepted: 07/11/2024] [Indexed: 11/16/2024]
Abstract
Background and Aims Necrotizing enterocolitis is a severe gastrointestinal complication of prematurity. Using small intestinal organoids derived from fetal tissue of a gestational age similar to an extremely preterm infant, this study aims to assess the effect of diet on intestinal epithelial growth and differentiation to elucidate the role nutrition type plays in intestinal development and modifies the risk for necrotizing enterocolitis. Methods Organoids were cultured for 5 days in growth media and 5 days in differentiation media supplemented 1:40 with 4 different diets: parental milk, donor human milk, standard formula, or extensively hydrolyzed formula. Images were captured daily and organoids were quantified. Organoids were preserved for RNA sequencing and immunofluorescence staining with Ki67, cleaved caspase 3, and chromogranin-A. Media was saved for cytokine/chemokine and growth factor analysis. Results Human milk supplementation improved growth and differentiation of intestinal organoids generating larger organoids during the growth phase and organoids with longer and wider buds during differentiation compared to formula. Ki67 staining confirmed the proliferative nature of milk-supplemented organoids and chromogranin A staining proved that MM-supplemented organoids induced highest enteroendocrine differentiation. Human milk supplementation also upregulated genes involved in Wnt signaling and fatty acid metabolism pathways and promoted a homeostatic immune landscape, including via increased secretion of tumor necrosis factor-related apoptosis-inducing ligand among other cytokines. Conversely, organoids supplemented with formula had a downregulation of cell-cycle-promoting genes and a more inflammatory immune signature, including a reduced level of leukemia inhibitory factor. Conclusion Our results demonstrate that parental milk, and to a lesser extent donor human milk, support robust intestinal epithelial proliferation, differentiation, and homeostatic cytokine production, suggesting a critical role for factors enriched in human milk in intestinal epithelial health.
Collapse
Affiliation(s)
- Lauren Smith
- Department of Pediatrics, Yale School of Medicine, New Haven, Connecticut
| | | | - Chino Eke
- Department of Pediatrics, Yale School of Medicine, New Haven, Connecticut
| | - Weihong Gu
- Department of Pediatrics, Yale School of Medicine, New Haven, Connecticut
| | - Wenjia Wang
- Department of Biostatistics, University of Pittsburgh, Pittsburgh, Pennsylvania
| | | | - Tessa Kehoe
- Department of Pediatrics, Yale School of Medicine, New Haven, Connecticut
| | - Kerri St Denis
- Department of Pediatrics, Yale School of Medicine, New Haven, Connecticut
| | - Madison Strine
- Department of Pediatrics, Yale School of Medicine, New Haven, Connecticut
| | - Sarah Taylor
- Department of Pediatrics, Yale School of Medicine, New Haven, Connecticut
| | - George Tseng
- Department of Biostatistics, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Liza Konnikova
- Department of Pediatrics, Yale School of Medicine, New Haven, Connecticut
- Department of Obstetrics, Gynecology and Reproductive Sciences, Yale School of Medicine, New Haven, Connecticut
- Department of Immunobiology, Yale School of Medicine, New Haven, Connecticut
- Program in Human and Translational Immunology, Yale School of Medicine, New Haven, Connecticut
- Program in Translational Biomedicine, Yale School of Medicine, New Haven, Connecticut
- Center for Systems and Engineering Immunology, Yale School of Medicine, New Haven, Connecticut
| |
Collapse
|
3
|
Zhao K, Li W, Yang Y, Hu X, Dai Y, Huang M, Luo J, Zhang K, Zhao N. Comprehensive analysis of m6A/m5C/m1A-related gene expression, immune infiltration, and sensitivity of antineoplastic drugs in glioma. Front Immunol 2022; 13:955848. [PMID: 36203569 PMCID: PMC9530704 DOI: 10.3389/fimmu.2022.955848] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2022] [Accepted: 09/07/2022] [Indexed: 11/20/2022] Open
Abstract
This research aims to develop a prognostic glioma marker based on m6A/m5C/m1A genes and investigate the potential role in the tumor immune microenvironment. Data for patients with glioma were downloaded from The Cancer Genome Atlas (TCGA) and Chinese Glioma Genome Atlas (CGGA). The expression of genes related to m6A/m5C/m1A was compared for normal and glioma groups. Gene Ontology and Kyoto Encyclopedia of Genes and Gene enrichment analysis of differentially expressed genes were conducted. Consistent clustering analysis was performed to obtain glioma subtypes and complete the survival analysis and immune analysis. Based on TCGA, Lasso regression analysis was used to obtain a prognostic model, and the CGGA database was used to validate the model. The model-based risk scores and the hub genes with the immune microenvironment, clinical features, and antitumor drug susceptibility were investigated. The clinical glioma tissues were collected to verify the expression of hub genes via immunohistochemistry. Twenty genes were differentially expressed, Consensus cluster analysis identified two molecular clusters. Overall survival was significantly higher in cluster 2 than in cluster 1. Immunological analysis revealed statistically significant differences in 26 immune cells and 17 immune functions between the two clusters. Enrichment analysis detected multiple meaningful pathways. We constructed a prognostic model that consists of WTAP, TRMT6, DNMT1, and DNMT3B. The high-risk and low-risk groups affected the survival prognosis and immune infiltration, which were related to grade, gender, age, and survival status. The prognostic value of the model was validated using another independent cohort CGGA. Clinical correlation and immune analysis revealed that four hub genes were associated with tumor grade, immune cells, and antitumor drug sensitivity, and WTAP was significantly associated with microsatellite instability(MSI). Immunohistochemistry confirmed the high expression of WTAP, DNMT1, and DNMT3B in tumor tissue, but the low expression of TRMT6. This study established a strong prognostic marker based on m6A/m5C/m1A methylation regulators, which can accurately predict the prognosis of patients with gliomas. m6A/m5C/m1A modification mode plays an important role in the tumor microenvironment, can provide valuable information for anti-tumor immunotherapy, and have a profound impact on the clinical characteristics.
Collapse
Affiliation(s)
- Kai Zhao
- Neurosurgery Department, the Second Affiliated Hospital of Kunming Medical University, Kunming, China
| | - Wenhu Li
- Neurosurgery Department, the Second Affiliated Hospital of Kunming Medical University, Kunming, China
| | - Yongtao Yang
- Cerebrovascular Disease Department, the Second Affiliated Hospital of Kunming Medical University, Kunming, China
| | - Xinyue Hu
- Department of Clinical Laboratory, Kunming First People’s Hospital, Kunming Medical University, Kunming, China
| | - Ying Dai
- Neurosurgery Department, the Second Affiliated Hospital of Kunming Medical University, Kunming, China
| | - Minhao Huang
- Neurosurgery Department, the Second Affiliated Hospital of Kunming Medical University, Kunming, China
| | - Ji Luo
- Neurosurgery Department, the Second Affiliated Hospital of Kunming Medical University, Kunming, China
| | - Kui Zhang
- Neurosurgery Department, the Second Affiliated Hospital of Kunming Medical University, Kunming, China
| | - Ninghui Zhao
- Neurosurgery Department, the Second Affiliated Hospital of Kunming Medical University, Kunming, China
- *Correspondence: Ninghui Zhao,
| |
Collapse
|