1
|
Ye Y, Zeng Y, Huang S, Zhu C, Wang Q. A Chemotherapy Response-Related Gene Signature and DNAJC8 as Key Mediators of Hepatocellular Carcinoma Progression and Drug Resistance. J Hepatocell Carcinoma 2025; 12:579-595. [PMID: 40130083 PMCID: PMC11932135 DOI: 10.2147/jhc.s506706] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2024] [Accepted: 02/21/2025] [Indexed: 03/26/2025] Open
Abstract
Background Chemotherapy resistance in hepatocellular carcinoma presents a significant challenge to improved patient outcomes. Identifying genes associated with chemotherapy response can enhance treatment strategies and prognostic models. Methods We analyzed the expression of chemotherapy response-related gene in hepatocellular carcinoma using TCGA and GSE109211 cohorts. We constructed a prognostic model using Least Absolute Shrinkage and Selection Operator (LASSO) analysis and assessed its efficacy using Kaplan-Meier survival analysis. Additionally, we evaluated the immune landscape and gene mutation profiles between different chemotherapy response-related gene (CRRG) subtypes. DNAJC8's role in hepatocellular carcinoma cell functions and chemotherapy resistance was further explored through gene knockdown experiments in vitro and in vivo. Results Differential expression analysis identified 220 common genes associated with chemotherapy response. The prognostic model incorporating seven key genes efficiently distinguished responders from non-responders and indicated poorer overall survival for the CRRG-high subtype. The CRRG value correlated with tumor stage and grade, and mutation profiles showed distinct patterns between CRRG subtypes. The CRRG-high subtype exhibited an immune-suppressive phenotype with higher expression of PD-L1 and CTLA-4. High DNAJC8 expression was linked to poor prognosis in multiple cohorts. Knocking down DNAJC8 significantly inhibited hepatocellular carcinoma cell proliferation, migration, invasion, and reduced sorafenib IC50. Conclusion The seven-gene CRRG model, particularly DNAJC8, holds potential for predicting chemotherapy response and serves as a therapeutic target in hepatocellular carcinoma.
Collapse
Affiliation(s)
- Yan Ye
- Ganzhou Key Laboratory of Molecular Medicine, The Affiliated Ganzhou Hospital of Nanchang University, Ganzhou, People’s Republic of China
| | - Yanmei Zeng
- Ganzhou Key Laboratory of Molecular Medicine, The Affiliated Ganzhou Hospital of Nanchang University, Ganzhou, People’s Republic of China
| | - Shenggang Huang
- Ganzhou Key Laboratory of Molecular Medicine, The Affiliated Ganzhou Hospital of Nanchang University, Ganzhou, People’s Republic of China
- Department of Gastroenterology, The Affiliated Ganzhou Hospital of Nanchang University, Ganzhou, People’s Republic of China
| | - Chunping Zhu
- Ganzhou Key Laboratory of Molecular Medicine, The Affiliated Ganzhou Hospital of Nanchang University, Ganzhou, People’s Republic of China
- Department of Gastroenterology, The Affiliated Ganzhou Hospital of Nanchang University, Ganzhou, People’s Republic of China
| | - Qingshui Wang
- College of Integrative Medicine, Academy of Integrative Medicine, Fujian University of Traditional Chinese Medicine, Fujian University of Traditional Chinese Medicine, Fuzhou, People’s Republic of China
| |
Collapse
|
2
|
Das J, Busia-Bourdain O, Khan KM, Wolfe AL. IMPlications of IMP2 in RNA Biology and Disease. Int J Mol Sci 2025; 26:2415. [PMID: 40141058 PMCID: PMC11942581 DOI: 10.3390/ijms26062415] [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: 01/16/2025] [Revised: 02/25/2025] [Accepted: 03/06/2025] [Indexed: 03/28/2025] Open
Abstract
Insulin-like growth factor 2 mRNA-binding protein 2 (IMP2) is an RNA-binding protein that positively regulates m6A-modified RNAs involved in critical cellular processes such as metabolism, oncogenesis, and immune function. Here, we elucidate facets of IMP2 biology, including several mechanisms of action on RNA, factors that regulate IMP2 expression, its relevant biological target RNAs, its role in normal development and disease, and its potential as a therapeutic target. IMP2 is a multi-level regulator of metabolism, influencing pathways linked to diabetes, obesity, and adipose function. Through genomic amplification and transcriptional overexpression in cancer cells, IMP2 can drive the initiation and progression of multiple cancer types, and high expression is associated with decreased overall survival of patients with cancer. IMP2 influences normal immune function, inflammation, macrophage polarization, and tumor immune evasion. IMP2 has emerged as a promising therapeutic target, particularly for cancers and metabolic diseases.
Collapse
Affiliation(s)
- Jessica Das
- Department of Biological Sciences, Hunter College, City University of New York, New York, NY 10065, USA
- Molecular, Cellular, and Developmental Biology Subprogram of the Biology Ph.D. Program, Graduate Center, City University of New York, New York, NY 10031, USA
| | - Ottavia Busia-Bourdain
- Department of Biological Sciences, Hunter College, City University of New York, New York, NY 10065, USA
| | - Khizr M. Khan
- Department of Biological Sciences, Hunter College, City University of New York, New York, NY 10065, USA
- New York Research and Mentoring for Postbaccalaureates (NY-RaMP) Program, Hunter College, New York, NY 10021, USA
| | - Andrew L. Wolfe
- Department of Biological Sciences, Hunter College, City University of New York, New York, NY 10065, USA
- Molecular, Cellular, and Developmental Biology Subprogram of the Biology Ph.D. Program, Graduate Center, City University of New York, New York, NY 10031, USA
- New York Research and Mentoring for Postbaccalaureates (NY-RaMP) Program, Hunter College, New York, NY 10021, USA
- Biochemistry Ph.D. Program, Graduate Center, City University of New York, New York, NY 10031, USA
- Department of Pharmacology, Weill Cornell Medicine, New York, NY 10021, USA
| |
Collapse
|
3
|
Kumari S, Peela S, Nagaraju GP, Srilatha M. Polysaccharides as therapeutic vehicles in pancreatic cancer. Drug Discov Today 2025; 30:104320. [PMID: 40024518 DOI: 10.1016/j.drudis.2025.104320] [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: 01/20/2025] [Revised: 02/13/2025] [Accepted: 02/25/2025] [Indexed: 03/04/2025]
Abstract
Pancreatic cancer (PC) is highly aggressive, with rising incidence and mortality rates. It has significant therapy obstacles due to the limited clinical options, late-stage identification, dense tumor microenvironment (TME), and resistance to therapy. Recent advances might improve treatment consequences in therapy strategies that target important TME components. Moreover, new polymeric drug delivery techniques based on polysaccharides such as polymeric micelles, liposomes, and nanoparticles enhance the solubility of drugs, drug stability, and tumor-specific targeting, which increase the chances of circumventing resistance and improving the efficacy of treatment. Preclinical research has suggested that by modulating the TME and enhancing the efficacy of chemotherapy, polysaccharide-based therapy, such as RP02-1 and DPLL-functionalized amylose, may help treat PC.
Collapse
Affiliation(s)
- Seema Kumari
- Department of Biotechnology, Dr. B.R. Ambedkar University, Srikakulam 532410 AP, India
| | - Sujatha Peela
- Department of Biotechnology, Dr. B.R. Ambedkar University, Srikakulam 532410 AP, India
| | - Ganji Purnachandra Nagaraju
- School of Medicine, Division of Hematology and Oncology, University of Alabama at Birmingham, Birmingham, AL 35233, USA
| | - Mundla Srilatha
- Department of Biotechnology, Sri Venkateswara University, Tirupati, Andhra Pradesh 517502, India.
| |
Collapse
|
4
|
Huang CY. The Loop-In Binding Mode of Dihydroorotase: Implications for Ligand Binding and Therapeutic Targeting. Int J Mol Sci 2025; 26:1359. [PMID: 39941127 PMCID: PMC11818841 DOI: 10.3390/ijms26031359] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2025] [Revised: 02/02/2025] [Accepted: 02/04/2025] [Indexed: 02/16/2025] Open
Abstract
Dihydroorotase (DHOase; EC 3.5.2.3) is a zinc-dependent metalloenzyme that plays a key role in the de novo pyrimidine biosynthesis pathway, catalyzing the reversible cyclization of N-carbamoyl aspartate to dihydroorotate. This reaction is essential for the production of uridine monophosphate, the precursor of all pyrimidine nucleotides required for DNA and RNA synthesis. Despite its conserved enzymatic function, DHOase exhibits significant structural diversity across species, particularly in its oligomeric states, gene fusion patterns, and active site architecture. A crucial structural feature of DHOase is its flexible active site loop, which undergoes dynamic conformational changes during catalysis. Previously, the loop-in conformation was associated with substrate binding, whereas the loop-out conformation was linked to product release and non-substrate ligand binding. However, recent crystallographic studies challenge this paradigm, revealing that certain non-substrate ligands and inhibitors, including malate, 5-fluoroorotate, plumbagin, 5-aminouracil, and 5-fluorouracil, interact with DHOase via a loop-in binding mechanism rather than the previously assumed loop-out mode. These findings necessitate a reassessment of the catalytic mechanism of DHOase and underscore the active site loop as a potential target for drug development. This review revisits the structural and biochemical mechanisms of DHOase, with a focus on recent crystallographic insights that redefine the loop-in binding mode for ligand interaction. By leveraging the unique conformational dynamics of the active site loop, novel inhibitors may be developed to selectively target pyrimidine biosynthesis in cancer cells and microbial pathogens. These insights emphasize the crucial role of structural biology in therapeutic design and highlight DHOase as a promising drug target.
Collapse
Affiliation(s)
- Cheng-Yang Huang
- Department of Biomedical Sciences, Chung Shan Medical University, Taichung City 402, Taiwan;
- Department of Medical Research, Chung Shan Medical University Hospital, Taichung City 402, Taiwan
| |
Collapse
|
5
|
Sharma N, Bhat SH, Mathew B, Yadav M, Tripathi G, Bindal V, Yadav S, Sharma N, Pandey S, Hemati H, Bohra D, Rana R, Sharma NK, Falari S, Pamecha V, Maras JS. Bile molecular landscape provides pathological insight and classifies signatures predictive of carcinoma of the gall bladder. MOLECULAR THERAPY. ONCOLOGY 2024; 32:200904. [PMID: 39640865 PMCID: PMC11617464 DOI: 10.1016/j.omton.2024.200904] [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: 06/26/2024] [Revised: 10/01/2024] [Accepted: 11/04/2024] [Indexed: 12/07/2024]
Abstract
Carcinoma of the gall bladder (CAGB) has a poor prognosis. Molecular analysis of bile could classify indicators of CAGB. Bile samples (n = 87; training cohort) were screened for proteomics and metabolomics signatures of cancer detection. In bile, CAGB showed distinct proteomic (217 upregulated, 258 downregulated) and metabolomic phenotypes (111 upregulated, 505 downregulated, p < 0.05, fold change > 1.5, false discovery rate <0.01) linked to significantly increased inflammation (coagulation, arachidonic acid, bile acid) and alternate energy pathways (pentose-phosphate pathway, amino acids, lipid metabolism); and decreased glycolysis, cholesterol metabolism, PPAR, RAS, and RAP1 signaling, oxidative phosphorylation, and others compared to gallstone or healthy controls (p < 0.05). Bile proteins/metabolites signatures showed significant correlation (r 2 > 0.5, p < 0.05) with clinical parameters. Metabolite/protein signature-based probability of detection for CAGB (cancer) was >90% (p < 0.05), with area under the receiver operating characteristic curve >0.94. Validation of the top four metabolites-toluene, 5,6-DHET, creatine, and phenylacetaldehyde-in separate cohorts (n = 80; bile [T1] and paired plasma [T2]) showed accuracy (99%) and sensitivity/specificity (>98%) for CAGB detection. Tissue validation showed bile 5,6-DHET positively correlated with tissue PCNA (proliferation), and caspase-3 linked to cancer development (r 2 >0.5, p < 0.05). In conclusion, the bile molecular landscape provides critical molecular understanding and outlines metabolomic indicator panels for early CAGB detection.
Collapse
Affiliation(s)
- Nupur Sharma
- Department of Molecular and Cellular Medicine, Institute of Liver and Biliary Sciences, New Delhi 110070, India
| | - Sadam H. Bhat
- Department of Molecular and Cellular Medicine, Institute of Liver and Biliary Sciences, New Delhi 110070, India
| | - Babu Mathew
- Department of Molecular and Cellular Medicine, Institute of Liver and Biliary Sciences, New Delhi 110070, India
| | - Manisha Yadav
- Department of Molecular and Cellular Medicine, Institute of Liver and Biliary Sciences, New Delhi 110070, India
| | - Gaurav Tripathi
- Department of Molecular and Cellular Medicine, Institute of Liver and Biliary Sciences, New Delhi 110070, India
| | - Vasundhra Bindal
- Department of Molecular and Cellular Medicine, Institute of Liver and Biliary Sciences, New Delhi 110070, India
| | - Sanju Yadav
- Department of Molecular and Cellular Medicine, Institute of Liver and Biliary Sciences, New Delhi 110070, India
| | - Neha Sharma
- Department of Molecular and Cellular Medicine, Institute of Liver and Biliary Sciences, New Delhi 110070, India
| | - Sushmita Pandey
- Department of Molecular and Cellular Medicine, Institute of Liver and Biliary Sciences, New Delhi 110070, India
| | - Hami Hemati
- Department of Toxicology and Cancer Biology, University of Kentucky, Lexington, KY 40506, USA
| | - Deepika Bohra
- Department of Research, GRIPMER, New Delhi 110060, India
| | - Rashmi Rana
- Department of Research, GRIPMER, New Delhi 110060, India
| | - Narendra Kumar Sharma
- Department of Bioscience and Biotechnology, Banasthali Vidyapith, Rajasthan 304022, India
| | - Sanyam Falari
- Department of Liver Transplant and HepatoPancreato Biliary Surgery, Institute of Liver and Biliary Sciences, New Delhi 110070, India
| | - Viniyendra Pamecha
- Department of Liver Transplant and HepatoPancreato Biliary Surgery, Institute of Liver and Biliary Sciences, New Delhi 110070, India
| | - Jaswinder Singh Maras
- Department of Molecular and Cellular Medicine, Institute of Liver and Biliary Sciences, New Delhi 110070, India
| |
Collapse
|
6
|
Baro M, Lee H, Kelley V, Lou R, Phoomak C, Politi K, Zeiss CJ, Van Zandt M, Contessa JN. OST Catalytic Subunit Redundancy Enables Therapeutic Targeting of N-Glycosylation. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.12.03.626593. [PMID: 39677793 PMCID: PMC11643024 DOI: 10.1101/2024.12.03.626593] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 12/17/2024]
Abstract
Protein asparagine (N)-glycosylation, which promotes folding and trafficking of cell surface receptors such as the EGFR, has not been considered a viable target in oncology due to the essential and non-redundant enzymatic activities required for glycan synthesis and transfer. In mammals an exception to this rule is the presence of the oligosaccharyltransferase (OST) catalytic subunit paralogs, STT3A and STT3B. Here we delineate the chemical biology of OST inhibitors and develop an approach for limited inhibition of N-glycosylation optimized for downstream effects on EGFR. Small molecules with enhanced pharmacokinetic properties and preferences for STT3A or STT3B were synthesized, characterized in vitro, and advanced to in vivo testing. The lead from this series, NGI-189, causes tumor regression or growth delay of patient derived and TKI resistant EGFR-mutant lung cancer xenografts without toxicity. Together these results suggest that bioavailable OST inhibitors can be developed as therapeutic agents for oncology.
Collapse
Affiliation(s)
- Marta Baro
- Department of Therapeutic Radiology, Yale University School of Medicine, New Haven, CT, 06510 USA
| | - Hojin Lee
- Department of Therapeutic Radiology, Yale University School of Medicine, New Haven, CT, 06510 USA
| | - Vanessa Kelley
- Department of Therapeutic Radiology, Yale University School of Medicine, New Haven, CT, 06510 USA
- Department of Pharmacology, Yale University School of Medicine, New Haven, CT, 06510 USA
| | - Rongliang Lou
- New England Discovery Partners, Branford, CT, 06405 USA
| | - Chatchai Phoomak
- Department of Therapeutic Radiology, Yale University School of Medicine, New Haven, CT, 06510 USA
- Department of Biology, Faculty of Science, Chulalongkorn University, Bangkok 10330 Thailand
| | - Katerina Politi
- Department of Internal Medicine (Section of Medical Oncology), Yale University School of Medicine, New Haven, CT, 06510 USA
| | - Caroline J. Zeiss
- Department of Comparative Medicine, Yale School of Medicine, New Haven, CT 06510, USA
| | | | - Joseph N. Contessa
- Department of Therapeutic Radiology, Yale University School of Medicine, New Haven, CT, 06510 USA
- Department of Pharmacology, Yale University School of Medicine, New Haven, CT, 06510 USA
- Lead contact
| |
Collapse
|
7
|
Nam M, Xia W, Mir AH, Jerrett A, Spinelli JB, Huang TT, Possemato R. Glucose limitation protects cancer cells from apoptosis induced by pyrimidine restriction and replication inhibition. Nat Metab 2024; 6:2338-2353. [PMID: 39592843 PMCID: PMC12019718 DOI: 10.1038/s42255-024-01166-w] [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: 10/23/2023] [Accepted: 10/10/2024] [Indexed: 11/28/2024]
Abstract
Cancer cells often experience nutrient-limiting conditions because of their robust proliferation and inadequate tumour vasculature, which results in metabolic adaptation to sustain proliferation. Most cancer cells rapidly consume glucose, which is severely reduced in the nutrient-scarce tumour microenvironment. In CRISPR-based genetic screens to identify metabolic pathways influenced by glucose restriction, we find that tumour-relevant glucose concentrations (low glucose) protect cancer cells from inhibition of de novo pyrimidine biosynthesis, a pathway that is frequently targeted by chemotherapy. We identify two mechanisms to explain this result, which is observed broadly across cancer types. First, low glucose limits uridine-5-diphosphate-glucose synthesis, preserving pyrimidine nucleotide availability and thereby prolonging the time to replication fork stalling. Second, low glucose directly modulates apoptosis downstream of replication fork stalling by suppressing BAK activation and subsequent cytochrome c release, key events that activate caspase-9-dependent mitochondrial apoptosis. These results indicate that the low glucose levels frequently observed in tumours may limit the efficacy of specific chemotherapeutic agents, highlighting the importance of considering the effects of the tumour nutrient environment on cancer therapy.
Collapse
Affiliation(s)
- Minwoo Nam
- Department of Pathology, New York University Grossman School of Medicine, New York, NY, USA
- Laura & Isaac Perlmutter Cancer Center, New York, NY, USA
| | - Wenxin Xia
- Laura & Isaac Perlmutter Cancer Center, New York, NY, USA
- Department of Biochemistry and Molecular Pharmacology, New York University Grossman School of Medicine, New York, NY, USA
| | - Abdul Hannan Mir
- Department of Pathology, New York University Grossman School of Medicine, New York, NY, USA
- Laura & Isaac Perlmutter Cancer Center, New York, NY, USA
| | | | | | - Tony T Huang
- Laura & Isaac Perlmutter Cancer Center, New York, NY, USA
- Department of Biochemistry and Molecular Pharmacology, New York University Grossman School of Medicine, New York, NY, USA
| | - Richard Possemato
- Department of Pathology, New York University Grossman School of Medicine, New York, NY, USA.
- Laura & Isaac Perlmutter Cancer Center, New York, NY, USA.
| |
Collapse
|
8
|
Long XB, Yao CR, Li SY, Zhang JG, Lu ZJ, Ma DD, Jiang YX, Ying GG, Shi WJ. Multiomics analysis reveal the impact of 17α-Ethinylestradiol on mortality in juvenile zebrafish. Comp Biochem Physiol C Toxicol Pharmacol 2024; 286:110027. [PMID: 39233286 DOI: 10.1016/j.cbpc.2024.110027] [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: 05/25/2024] [Revised: 08/11/2024] [Accepted: 09/01/2024] [Indexed: 09/06/2024]
Abstract
17α-Ethinylestradiol (EE2) is known for its endocrine-disrupting effects on embryonic and adult fish. However, its impact on juvenile zebrafish has not been well established. In this study, juvenile zebrafish were exposed to EE2 at concentrations of 5 ng/L (low dose, L), 10 ng/L (medium dose, M), and 50 ng/L (high dose, H) from 21 days post-fertilization (dpf) to 49 dpf. We assessed their growth, development, behavior, transcriptome, and metabolome. The findings showed that the survival rate in the EE2-H group was 66.8 %, with all surviving fish displaying stunted growth and swollen, transparent abdomens by 49 dpf. Moreover, severe organ deformities were observed in the gills, kidneys, intestines, and heart of fish in both the EE2-H and EE2-M groups. Co-expression analysis of mRNA and lncRNA revealed that EE2 downregulated the transcription of key genes involved in the cell cycle, DNA replication, and Fanconi anemia signaling pathways. Additionally, metabolomic analysis indicated that EE2 influenced metabolism and development-related signaling pathways. These pathways were also significantly identified based on the genes regulated by lncRNA. Consequently, EE2 induced organ deformities and mortality in juvenile zebrafish by disrupting signaling pathways associated with development and metabolism. The results of this study offer new mechanistic insights into the adverse effects of EE2 on juvenile zebrafish based on multiomics analysis. The juvenile zebrafish are highly sensitive to EE2 exposure, which is not limited to adult and embryonic stages. It is a potential model for studying developmental toxicity.
Collapse
Affiliation(s)
- Xiao-Bing Long
- SCNU Environmental Research Institute, Guangdong Provincial Key Laboratory of Chemical Pollution and Environmental Safety & MOE Key Laboratory of Theoretical Chemistry of Environment, South China Normal University, Guangzhou 510006, China; School of Environment, South China Normal University, University Town, Guangzhou 510006, China
| | - Chong-Rui Yao
- SCNU Environmental Research Institute, Guangdong Provincial Key Laboratory of Chemical Pollution and Environmental Safety & MOE Key Laboratory of Theoretical Chemistry of Environment, South China Normal University, Guangzhou 510006, China; School of Environment, South China Normal University, University Town, Guangzhou 510006, China
| | - Si-Ying Li
- SCNU Environmental Research Institute, Guangdong Provincial Key Laboratory of Chemical Pollution and Environmental Safety & MOE Key Laboratory of Theoretical Chemistry of Environment, South China Normal University, Guangzhou 510006, China; School of Environment, South China Normal University, University Town, Guangzhou 510006, China
| | - Jin-Ge Zhang
- SCNU Environmental Research Institute, Guangdong Provincial Key Laboratory of Chemical Pollution and Environmental Safety & MOE Key Laboratory of Theoretical Chemistry of Environment, South China Normal University, Guangzhou 510006, China; School of Environment, South China Normal University, University Town, Guangzhou 510006, China
| | - Zhi-Jie Lu
- SCNU Environmental Research Institute, Guangdong Provincial Key Laboratory of Chemical Pollution and Environmental Safety & MOE Key Laboratory of Theoretical Chemistry of Environment, South China Normal University, Guangzhou 510006, China; School of Environment, South China Normal University, University Town, Guangzhou 510006, China
| | - Dong-Dong Ma
- SCNU Environmental Research Institute, Guangdong Provincial Key Laboratory of Chemical Pollution and Environmental Safety & MOE Key Laboratory of Theoretical Chemistry of Environment, South China Normal University, Guangzhou 510006, China; School of Environment, South China Normal University, University Town, Guangzhou 510006, China
| | - Yu-Xia Jiang
- South China Institute of Environmental Sciences, Ministry of Ecology and Environment, Guangzhou 510535, China
| | - Guang-Guo Ying
- SCNU Environmental Research Institute, Guangdong Provincial Key Laboratory of Chemical Pollution and Environmental Safety & MOE Key Laboratory of Theoretical Chemistry of Environment, South China Normal University, Guangzhou 510006, China; School of Environment, South China Normal University, University Town, Guangzhou 510006, China
| | - Wen-Jun Shi
- SCNU Environmental Research Institute, Guangdong Provincial Key Laboratory of Chemical Pollution and Environmental Safety & MOE Key Laboratory of Theoretical Chemistry of Environment, South China Normal University, Guangzhou 510006, China; School of Environment, South China Normal University, University Town, Guangzhou 510006, China.
| |
Collapse
|
9
|
Xu L, Zhang W, Zhang H, Yang X, Ceccobelli S, Zhao Y, E G. Identification of Goat Supernumerary Teat Phenotype Using Wide-Genomic Copy Number Variants. Animals (Basel) 2024; 14:3252. [PMID: 39595305 PMCID: PMC11591440 DOI: 10.3390/ani14223252] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2024] [Revised: 11/08/2024] [Accepted: 11/12/2024] [Indexed: 11/28/2024] Open
Abstract
Supernumerary teats (SNTs) or nipples often emerge around the mammary line. This study performed a genome-wide selective sweep analysis (GWS) at the copy number variant (CNV) level using two selected signal calculation methods (VST and FST) to identify candidate genes associated with SNTs in goats. A total of 12,310 CNVs were identified from 37 animals and 123 CNVs, with the top 1% VST values including 84 candidate genes (CDGs). Of these CDGs, minichromosome maintenance complex component 3, ectodysplasin A receptor associated via death domain, and cullin 5 demonstrated functions closely related to mammary gland development. In addition, 123 CNVs with the top 1% FST values were annotated to 97 CDGs. 5-Hydroxytryptamine receptor 2A, CCAAT/enhancer-binding protein alpha, and the polymeric immunoglobulin receptor affect colostrum secretion through multiple signaling pathways. Two genes, namely, RNA-binding motif protein 46 and β-1,3-galactosyltransferase 5, showed a close relation to mammary gland development. Six CNVs were identified and annotated to five genes by intersecting the top 1% of candidate CNVs with both parameters. These genes include LOC102185621, LOC102190481, and UDP-glucose pyrophosphorylase 2, which potentially affect the occurrence of BC through multiple biological processes, such as cell detoxification, glycogen synthesis, and phospholipid metabolism. In conclusion, we discovered numerous genes related to mammary development and breast cancer (BC) through a GWS, which suggests the mechanism of SNTs in goats and a certain association between mammary cancer and SNTs.
Collapse
Affiliation(s)
- Lu Xu
- College of Animal Science and Technology, Southwest University, Chongqing 400715, China; (L.X.); (W.Z.); (H.Z.); (Y.Z.)
| | - Weiyi Zhang
- College of Animal Science and Technology, Southwest University, Chongqing 400715, China; (L.X.); (W.Z.); (H.Z.); (Y.Z.)
| | - Haoyuan Zhang
- College of Animal Science and Technology, Southwest University, Chongqing 400715, China; (L.X.); (W.Z.); (H.Z.); (Y.Z.)
| | - Xiuqin Yang
- College of Animal Science and Technology, Northeast Agricultural University, Harbin 150030, China;
| | - Simone Ceccobelli
- Department of Agricultural, Food and Environmental Sciences, Università Politecnica delle Marche, 60131 Ancona, Italy;
| | - Yongju Zhao
- College of Animal Science and Technology, Southwest University, Chongqing 400715, China; (L.X.); (W.Z.); (H.Z.); (Y.Z.)
| | - Guangxin E
- College of Animal Science and Technology, Southwest University, Chongqing 400715, China; (L.X.); (W.Z.); (H.Z.); (Y.Z.)
| |
Collapse
|
10
|
Jaiyesimi O, Kuppuswamy S, Zhang G, Batan S, Zhi W, Ganta VC. Glycolytic PFKFB3 and Glycogenic UGP2 Axis Regulates Perfusion Recovery in Experimental Hind Limb Ischemia. Arterioscler Thromb Vasc Biol 2024; 44:1764-1783. [PMID: 38934117 PMCID: PMC11323258 DOI: 10.1161/atvbaha.124.320665] [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: 01/03/2024] [Accepted: 06/03/2024] [Indexed: 06/28/2024]
Abstract
BACKGROUND Despite being in an oxygen-rich environment, endothelial cells (ECs) use anaerobic glycolysis (Warburg effect) as the primary metabolic pathway for cellular energy needs. PFKFB (6-phosphofructo-2-kinase/fructose-2,6-biphosphatase)-3 regulates a critical enzymatic checkpoint in glycolysis and has been shown to induce angiogenesis. This study builds on our efforts to determine the metabolic regulation of ischemic angiogenesis and perfusion recovery in the ischemic muscle. METHODS Hypoxia serum starvation (HSS) was used as an in vitro peripheral artery disease (PAD) model, and hind limb ischemia by femoral artery ligation and resection was used as a preclinical PAD model. RESULTS Despite increasing PFKFB3-dependent glycolysis, HSS significantly decreased the angiogenic capacity of ischemic ECs. Interestingly, inhibiting PFKFB3 significantly induced the angiogenic capacity of HSS-ECs. Since ischemia induced a significant in PFKFB3 levels in hind limb ischemia muscle versus nonischemic, we wanted to determine whether glucose bioavailability (rather than PFKFB3 expression) in the ischemic muscle is a limiting factor behind impaired angiogenesis. However, treating the ischemic muscle with intramuscular delivery of D-glucose or L-glucose (osmolar control) showed no significant differences in the perfusion recovery, indicating that glucose bioavailability is not a limiting factor to induce ischemic angiogenesis in experimental PAD. Unexpectedly, we found that shRNA-mediated PFKFB3 inhibition in the ischemic muscle resulted in an increased perfusion recovery and higher vascular density compared with control shRNA (consistent with the increased angiogenic capacity of PFKFB3 silenced HSS-ECs). Based on these data, we hypothesized that inhibiting HSS-induced PFKFB3 expression/levels in ischemic ECs activates alternative metabolic pathways that revascularize the ischemic muscle in experimental PAD. A comprehensive glucose metabolic gene qPCR arrays in PFKFB3 silenced HSS-ECs, and PFKFB3-knock-down ischemic muscle versus respective controls identified UGP2 (uridine diphosphate-glucose pyrophosphorylase 2), a regulator of protein glycosylation and glycogen synthesis, is induced upon PFKFB3 inhibition in vitro and in vivo. Antibody-mediated inhibition of UGP2 in the ischemic muscle significantly impaired perfusion recovery versus IgG control. Mechanistically, supplementing uridine diphosphate-glucose, a metabolite of UGP2 activity, significantly induced HSS-EC angiogenic capacity in vitro and enhanced perfusion recovery in vivo by increasing protein glycosylation (but not glycogen synthesis). CONCLUSIONS Our data present that inhibition of maladaptive PFKFB3-driven glycolysis in HSS-ECs is necessary to promote the UGP2-uridine diphosphate-glucose axis that enhances ischemic angiogenesis and perfusion recovery in experimental PAD.
Collapse
Affiliation(s)
- Olukemi Jaiyesimi
- Vascular Biology Center and Department of Medicine (J.O., S.K., G.Z., S.B., V.C.G.), Augusta University, GA
| | - Sivaraman Kuppuswamy
- Vascular Biology Center and Department of Medicine (J.O., S.K., G.Z., S.B., V.C.G.), Augusta University, GA
| | - Guangwei Zhang
- Vascular Biology Center and Department of Medicine (J.O., S.K., G.Z., S.B., V.C.G.), Augusta University, GA
| | - Sonia Batan
- Vascular Biology Center and Department of Medicine (J.O., S.K., G.Z., S.B., V.C.G.), Augusta University, GA
| | - Wenbo Zhi
- Department of Obstetrics and Gynecology, Center for Biotechnology and Genomic Medicine (W.Z.), Augusta University, GA
| | - Vijay C Ganta
- Vascular Biology Center and Department of Medicine (J.O., S.K., G.Z., S.B., V.C.G.), Augusta University, GA
| |
Collapse
|
11
|
Lam D, Arroyo B, Liberchuk AN, Wolfe AL. Effects of N361 Glycosylation on Epidermal Growth Factor Receptor Biological Function. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.07.12.603279. [PMID: 39071333 PMCID: PMC11275927 DOI: 10.1101/2024.07.12.603279] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 07/30/2024]
Abstract
Epidermal growth factor receptor (EGFR) is a transmembrane tyrosine kinase that is frequently modified by glycosylation post-translationally. In cancer, EGFR amplifications and hotspot mutations such as L858R that promote proliferation have been detected in a significant fraction of non-small cell lung carcinomas and breast adenocarcinomas. Molecular dynamic simulations suggested that glycosylation at asparagine residue 361 (N361) promotes dimerization and ligand binding. We stably expressed glycosylation-deficient mutant EGFR N361A, with or without the oncogenic mutation L858R. Immunofluorescence and flow cytometry demonstrated that the mutants were each well expressed at the cell membrane. N361A decreased proliferation relative to wild-type EGFR as well as decreased sensitivity to ligands. Proximity ligation assays measuring co-localization of EGFR with its binding partner HER2 in cells revealed that N361A mutations increased co-localization. N361A, located near the binding interface for the EGFR inhibitor necitumumab, desensitized cells expressing the oncogenic EGFR L858R to antibody-based inhibition. These findings underline the critical relevance of post-translational modifications on oncogene function.
Collapse
Affiliation(s)
- Dennis Lam
- Department of Biological Sciences, Hunter College of the City University of New York
- Department of Pharmacology, Weill Cornell Medicine
| | - Brandon Arroyo
- Department of Biological Sciences, Hunter College of the City University of New York
- Maximizing Access to Research Careers Program, Hunter College of the City University of New York
| | - Ariel N. Liberchuk
- Department of Biological Sciences, Hunter College of the City University of New York
- Macaulay Honors College, Hunter College of the City University of New York
| | - Andrew L. Wolfe
- Department of Biological Sciences, Hunter College of the City University of New York
- Department of Pharmacology, Weill Cornell Medicine
- Biochemistry Ph.D. Program, Graduate Center of the City University of New York
- Molecular, Cellular, and Developmental Biology Ph.D. Subprogram, Graduate Center of the City University of New York
| |
Collapse
|
12
|
Ou Y, Guo Y, Wang H, Guo Z, Zheng B. Porphyra haitanensis glycoprotein regulates glucose homeostasis: targeting the liver. Food Funct 2024; 15:7491-7508. [PMID: 38916282 DOI: 10.1039/d4fo01544d] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/26/2024]
Abstract
In this study, we investigated the effects of glycoprotein (PG)-mediated regulation of Porphyra haitanensis on liver glucose metabolism in hyperglycemic mouse models, and sought to establish the underlying mechanism, as determined by the changes in liver gene expression and metabolic profiles. The results showed that 30-300 mg kg-1 PG upregulated the expression of the liver genes Ins1, Ins2, Insr, Gys2, Gpi1, Gck, and downregulated the expression of G6pc, G6pc2, and G6pc3, in a concentration-dependent manner. 300 mg kg-1 PG downregulated the concentrations of glucose-related metabolites in the liver, but upregulated lactic acid, 2-aminoacetic acid, and glucose-1-phosphate concentrations. It was assumed that PG regulated liver glucose metabolism by enriching insulin secretion, glycolysis/gluconeogenesis, and the AMPK signaling pathway, and promoting insulin secretion, glycogen synthesis, and glycolysis. Our findings supported the development of P. haitanensis and its glycoproteins as novel natural antidiabetic compounds that regulated blood glucose homeostasis.
Collapse
Affiliation(s)
- Yujia Ou
- College of Food Science, Fujian Agriculture and Forestry University, Fuzhou 350002, China.
- Engineering Research Center of Fujian-Taiwan Special Marine Food Processing and Nutrition, Ministry of Education, Fuzhou 350002, China
| | - Yuehong Guo
- College of Food Science, Fujian Agriculture and Forestry University, Fuzhou 350002, China.
- Engineering Research Center of Fujian-Taiwan Special Marine Food Processing and Nutrition, Ministry of Education, Fuzhou 350002, China
| | - Haoyu Wang
- College of Food Science, Fujian Agriculture and Forestry University, Fuzhou 350002, China.
| | - Zebin Guo
- College of Food Science, Fujian Agriculture and Forestry University, Fuzhou 350002, China.
- Engineering Research Center of Fujian-Taiwan Special Marine Food Processing and Nutrition, Ministry of Education, Fuzhou 350002, China
| | - Baodong Zheng
- College of Food Science, Fujian Agriculture and Forestry University, Fuzhou 350002, China.
- Engineering Research Center of Fujian-Taiwan Special Marine Food Processing and Nutrition, Ministry of Education, Fuzhou 350002, China
| |
Collapse
|
13
|
Sahu U, Villa E, Reczek CR, Zhao Z, O’Hara BP, Torno MD, Mishra R, Shannon WD, Asara JM, Gao P, Shilatifard A, Chandel NS, Ben-Sahra I. Pyrimidines maintain mitochondrial pyruvate oxidation to support de novo lipogenesis. Science 2024; 383:1484-1492. [PMID: 38547260 PMCID: PMC11325697 DOI: 10.1126/science.adh2771] [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: 02/21/2023] [Accepted: 02/20/2024] [Indexed: 04/02/2024]
Abstract
Cellular purines, particularly adenosine 5'-triphosphate (ATP), fuel many metabolic reactions, but less is known about the direct effects of pyrimidines on cellular metabolism. We found that pyrimidines, but not purines, maintain pyruvate oxidation and the tricarboxylic citric acid (TCA) cycle by regulating pyruvate dehydrogenase (PDH) activity. PDH activity requires sufficient substrates and cofactors, including thiamine pyrophosphate (TPP). Depletion of cellular pyrimidines decreased TPP synthesis, a reaction carried out by TPP kinase 1 (TPK1), which reportedly uses ATP to phosphorylate thiamine (vitamin B1). We found that uridine 5'-triphosphate (UTP) acts as the preferred substrate for TPK1, enabling cellular TPP synthesis, PDH activity, TCA-cycle activity, lipogenesis, and adipocyte differentiation. Thus, UTP is required for vitamin B1 utilization to maintain pyruvate oxidation and lipogenesis.
Collapse
Affiliation(s)
- Umakant Sahu
- Department of Biochemistry and Molecular Genetics, Feinberg School of Medicine, Northwestern University, Chicago, IL, 60611, USA
- Robert H. Lurie Comprehensive Cancer Center, Northwestern University, Chicago IL, 60611 USA
| | - Elodie Villa
- Department of Biochemistry and Molecular Genetics, Feinberg School of Medicine, Northwestern University, Chicago, IL, 60611, USA
- Robert H. Lurie Comprehensive Cancer Center, Northwestern University, Chicago IL, 60611 USA
| | - Colleen R. Reczek
- Robert H. Lurie Comprehensive Cancer Center, Northwestern University, Chicago IL, 60611 USA
- Department of Medicine, Northwestern University Feinberg School of Medicine, Chicago, IL, 60611, USA
| | - Zibo Zhao
- Department of Biochemistry and Molecular Genetics, Feinberg School of Medicine, Northwestern University, Chicago, IL, 60611, USA
- Robert H. Lurie Comprehensive Cancer Center, Northwestern University, Chicago IL, 60611 USA
| | - Brendan P. O’Hara
- Department of Biochemistry and Molecular Genetics, Feinberg School of Medicine, Northwestern University, Chicago, IL, 60611, USA
- Robert H. Lurie Comprehensive Cancer Center, Northwestern University, Chicago IL, 60611 USA
| | - Michael D. Torno
- Department of Biochemistry and Molecular Genetics, Feinberg School of Medicine, Northwestern University, Chicago, IL, 60611, USA
- Robert H. Lurie Comprehensive Cancer Center, Northwestern University, Chicago IL, 60611 USA
| | | | | | - John M. Asara
- Mass Spectrometry Core, Beth Israel Deaconess Medical Center, Department of Medicine, Harvard Medical School, Boston, MA, 02115, USA
| | - Peng Gao
- Metabolomics Core Facility, Robert H. Lurie Comprehensive Cancer Center, Northwestern University, Chicago, IL, 60611, USA.2
| | - Ali Shilatifard
- Department of Biochemistry and Molecular Genetics, Feinberg School of Medicine, Northwestern University, Chicago, IL, 60611, USA
- Robert H. Lurie Comprehensive Cancer Center, Northwestern University, Chicago IL, 60611 USA
| | - Navdeep S. Chandel
- Department of Biochemistry and Molecular Genetics, Feinberg School of Medicine, Northwestern University, Chicago, IL, 60611, USA
- Robert H. Lurie Comprehensive Cancer Center, Northwestern University, Chicago IL, 60611 USA
- Department of Medicine, Northwestern University Feinberg School of Medicine, Chicago, IL, 60611, USA
| | - Issam Ben-Sahra
- Department of Biochemistry and Molecular Genetics, Feinberg School of Medicine, Northwestern University, Chicago, IL, 60611, USA
- Robert H. Lurie Comprehensive Cancer Center, Northwestern University, Chicago IL, 60611 USA
| |
Collapse
|
14
|
Chen L, Chu D, Li W, Zhang H. Non-small cell lung cancer patient with a rare UGP2-ALK fusion protein responded well to alectinib: a case report. Anticancer Drugs 2024; 35:97-100. [PMID: 37450292 PMCID: PMC10720821 DOI: 10.1097/cad.0000000000001531] [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: 06/14/2023] [Revised: 06/14/2023] [Indexed: 07/18/2023]
Abstract
Several rare anaplastic lymphoma kinase (ALK) fusions have been identified in patients with non-small cell lung cancer (NSCLC); however, their treatment is not currently uniform. alectinib has been commonly used to treat rare ALK fusions in patients with NSCLC. This is the first study to report the occurrence of a uridine diphosphate-glucose pyrophosphorylase 2 (UGP2)-ALK fusion in a patient with NSCLC. The patient, who was hospitalized because of shortness of breath lasting 20 days, showed hydrothorax of the left lung under a computerized tomography chest scan. Pathological histology revealed lung adenocarcinoma in the patient. The UGP2-ALK mutation was found by next-generation sequencing. Subsequently, the patient was administered alectinib, and thereafter, the tumor lesion was observed to gradually shrink over the follow-up period. Progression-free survival reached 10 months as of the follow-up date, with no adverse events detected. This case report provides valuable insights into the clinical management of NSCLC patients with UGP2-ALK fusions. Moreover, alectinib is confirmed to be an appropriate therapeutic agent for such patients.
Collapse
Affiliation(s)
- Liulin Chen
- Department of Respiratory and Critical Care Medicine, Tangdu Hospital, Air Force Military Medical University, Xi’an, China
| | - Daifang Chu
- Department of Respiratory and Critical Care Medicine, Tangdu Hospital, Air Force Military Medical University, Xi’an, China
| | - Wangping Li
- Department of Respiratory and Critical Care Medicine, Tangdu Hospital, Air Force Military Medical University, Xi’an, China
| | - Haitao Zhang
- Department of Respiratory and Critical Care Medicine, Tangdu Hospital, Air Force Military Medical University, Xi’an, China
| |
Collapse
|
15
|
Han X, Han B, Luo H, Ling H, Hu X. Integrated Multi-Omics Profiling of Young Breast Cancer Patients Reveals a Correlation between Galactose Metabolism Pathway and Poor Disease-Free Survival. Cancers (Basel) 2023; 15:4637. [PMID: 37760606 PMCID: PMC10526161 DOI: 10.3390/cancers15184637] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2023] [Revised: 09/08/2023] [Accepted: 09/13/2023] [Indexed: 09/29/2023] Open
Abstract
In recent years, there has been a notable rise in the incidence of breast cancer among young patients, who exhibit worse survival outcomes and distinct characteristics compared to intermediate and elderly patients. Therefore, it is imperative to identify the specific features unique to young patients, which could offer insights into potential therapeutic strategies and improving survival outcomes. In our study, we performed an integrative analysis of bulk transcriptional and genomic data from extensive clinical cohorts to identify the prognostic factotrs. Additionally, we analyzed the single-cell transcriptional data and conducted in vitro experiments. Our work confirmed that young patients exhibited higher grading, worse disease-free survival (DFS), a higher frequency of mutations in TP53 and BRCA1, a lower frequency of mutations in PIK3CA, and upregulation of eight metabolic pathways. Notably, the galactose metabolism pathway showed upregulation in young patients and was associated with worse DFS. Further analysis and experiments indicated that the galactose metabolism pathway may regulate the stemness of cancer cells and ultimately contribute to worse survival outcomes. In summary, our finding identified distinct clinicopathological, transcriptional, and genomics features and revealed a correlation between the galactose metabolism pathway, stemness, and poor disease-free survival of breast cancer in young patients.
Collapse
Affiliation(s)
- Xiangchen Han
- Department of Breast Surgery, Fudan University Shanghai Cancer Center, Shanghai 200032, China; (X.H.); (B.H.)
- Key Laboratory of Breast Cancer in Shanghai, Department of Oncology, Shanghai Medical College, Fudan University, Shanghai 200032, China
- Precision Cancer Medical Center Affiliated to Fudan University Shanghai Cancer Center, Shanghai 200032, China;
| | - Boyue Han
- Department of Breast Surgery, Fudan University Shanghai Cancer Center, Shanghai 200032, China; (X.H.); (B.H.)
- Key Laboratory of Breast Cancer in Shanghai, Department of Oncology, Shanghai Medical College, Fudan University, Shanghai 200032, China
- Precision Cancer Medical Center Affiliated to Fudan University Shanghai Cancer Center, Shanghai 200032, China;
| | - Hong Luo
- Precision Cancer Medical Center Affiliated to Fudan University Shanghai Cancer Center, Shanghai 200032, China;
| | - Hong Ling
- Department of Breast Surgery, Fudan University Shanghai Cancer Center, Shanghai 200032, China; (X.H.); (B.H.)
- Key Laboratory of Breast Cancer in Shanghai, Department of Oncology, Shanghai Medical College, Fudan University, Shanghai 200032, China
| | - Xin Hu
- Key Laboratory of Breast Cancer in Shanghai, Department of Oncology, Shanghai Medical College, Fudan University, Shanghai 200032, China
- Precision Cancer Medical Center Affiliated to Fudan University Shanghai Cancer Center, Shanghai 200032, China;
| |
Collapse
|
16
|
Nissa MU, Pinto N, Ghosh B, Singh U, Goswami M, Srivastava S. Proteomic analysis of liver tissue reveals Aeromonas hydrophila infection mediated modulation of host metabolic pathways in Labeo rohita. J Proteomics 2023; 279:104870. [PMID: 36906258 DOI: 10.1016/j.jprot.2023.104870] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2022] [Revised: 02/20/2023] [Accepted: 03/04/2023] [Indexed: 03/11/2023]
Abstract
Aeromonas hydrophila (Ah) is a Gram-negative bacterium and a serious global pathogen causing Motile Aeromonas Septicaemia (MAS) in fish leading to global loss in aquaculture. Investigation of the molecular alterations of host tissues such as liver could be a powerful approach to identify mechanistic and diagnostic immune signatures of disease pathogenesis. We performed a proteomic analysis of Labeo rohita liver tissue to examine the protein dynamics in the host cells during Ah infection. The proteomic data was acquired using two strategies; discovery and targeted proteomics. Label-free quantification was performed between Control and challenged group (AH) to identify the differentially expressed proteins (DEPs). A total of 2525 proteins were identified and 157 were DEPs. DEPs include metabolic enzymes (CS, SUCLG2), antioxidative proteins, cytoskeletal proteins and immune related proteins (TLR3, CLEC4E). Pathways like lysosome pathway, apoptosis, metabolism of xenobiotics by cytochrome P450 were enriched by downregulated proteins. However, upregulated proteins majorly mapped to innate immune system, signaling of B cell receptor, proteosome pathway, ribosome, carbon metabolism and protein processing in ER. Our study would help in exploring the role of Toll-like receptors, C-type lectins and, metabolic intermediates like citrate and succinate in Ah pathogenesis to understand the Ah infection in fish. SIGNIFICANCE: Bacterial diseases such as motile aeromonas septicaemia (MAS) are among the most serious problems in aquaculture industry. Small molecules that target the metabolism of the host have recently emerged as potential treatment possibilities in infectious diseases. However, the ability to develop new therapies is hampered due to lack of knowledge about pathogenesis mechanisms and host-pathogen interactions. We examined alterations in the host proteome during MAS caused by Aeromonas hydrophila (Ah) infection, in Labeo rohita liver tissue to find cellular proteins and processes affected by Ah infection. Upregulated proteins belong to innate immune system, signaling of B cell receptor, proteosome pathway, ribosome, carbon metabolism and protein processing. Our work is an important step towards leveraging host metabolism in targeting the disease by providing a bigger picture on proteome pathology correlation during Ah infection.
Collapse
Affiliation(s)
- Mehar Un Nissa
- Department of Biosciences and Bioengineering, Indian Institute of Technology Bombay, Powai, Mumbai 400076, India
| | - Nevil Pinto
- Central Institute of Fisheries Education, Indian Council of Agricultural Research, Versova, Mumbai, Maharashtra 400061, India
| | - Biplab Ghosh
- Regional Centre for Biotechnology, Faridabad 121001, India
| | - Urvi Singh
- Department of Biochemistry, Sri Venkateswara College, University of Delhi, 110034, India
| | - Mukunda Goswami
- Central Institute of Fisheries Education, Indian Council of Agricultural Research, Versova, Mumbai, Maharashtra 400061, India.
| | - Sanjeeva Srivastava
- Department of Biosciences and Bioengineering, Indian Institute of Technology Bombay, Powai, Mumbai 400076, India.
| |
Collapse
|
17
|
Young LEA, Conroy LR, Clarke HA, Hawkinson TR, Bolton KE, Sanders WC, Chang JE, Webb MB, Alilain WJ, Vander Kooi CW, Drake RR, Andres DA, Badgett TC, Wagner LM, Allison DB, Sun RC, Gentry MS. In situ mass spectrometry imaging reveals heterogeneous glycogen stores in human normal and cancerous tissues. EMBO Mol Med 2022; 14:e16029. [PMID: 36059248 PMCID: PMC9641418 DOI: 10.15252/emmm.202216029] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2022] [Revised: 07/25/2022] [Accepted: 08/03/2022] [Indexed: 01/19/2023] Open
Abstract
Glycogen dysregulation is a hallmark of aging, and aberrant glycogen drives metabolic reprogramming and pathogenesis in multiple diseases. However, glycogen heterogeneity in healthy and diseased tissues remains largely unknown. Herein, we describe a method to define spatial glycogen architecture in mouse and human tissues using matrix-assisted laser desorption/ionization mass spectrometry imaging. This assay provides robust and sensitive spatial glycogen quantification and architecture characterization in the brain, liver, kidney, testis, lung, bladder, and even the bone. Armed with this tool, we interrogated glycogen spatial distribution and architecture in different types of human cancers. We demonstrate that glycogen stores and architecture are heterogeneous among diseases. Additionally, we observe unique hyperphosphorylated glycogen accumulation in Ewing sarcoma, a pediatric bone cancer. Using preclinical models, we correct glycogen hyperphosphorylation in Ewing sarcoma through genetic and pharmacological interventions that ablate in vivo tumor growth, demonstrating the clinical therapeutic potential of targeting glycogen in Ewing sarcoma.
Collapse
Affiliation(s)
- Lyndsay E A Young
- Department of Molecular and Cellular Biochemistry, College of MedicineUniversity of KentuckyLexingtonKYUSA
- Markey Cancer CenterUniversity of KentuckyLexingtonKYUSA
| | - Lindsey R Conroy
- Markey Cancer CenterUniversity of KentuckyLexingtonKYUSA
- Department of Neuroscience, College of MedicineUniversity of KentuckyLexingtonKYUSA
| | - Harrison A Clarke
- Department of Neuroscience, College of MedicineUniversity of KentuckyLexingtonKYUSA
| | - Tara R Hawkinson
- Department of Neuroscience, College of MedicineUniversity of KentuckyLexingtonKYUSA
| | - Kayli E Bolton
- Department of Molecular and Cellular Biochemistry, College of MedicineUniversity of KentuckyLexingtonKYUSA
| | - William C Sanders
- Department of Molecular and Cellular Biochemistry, College of MedicineUniversity of KentuckyLexingtonKYUSA
| | - Josephine E Chang
- Department of Neuroscience, College of MedicineUniversity of KentuckyLexingtonKYUSA
| | - Madison B Webb
- Department of Molecular and Cellular Biochemistry, College of MedicineUniversity of KentuckyLexingtonKYUSA
| | - Warren J Alilain
- Department of Neuroscience, College of MedicineUniversity of KentuckyLexingtonKYUSA
- Spinal Cord and Brain Injury Research CenterUniversity of KentuckyLexingtonKYUSA
| | - Craig W Vander Kooi
- Department of Molecular and Cellular Biochemistry, College of MedicineUniversity of KentuckyLexingtonKYUSA
- Markey Cancer CenterUniversity of KentuckyLexingtonKYUSA
| | - Richard R Drake
- Cell and Molecular Pharmacology and Experimental TherapeuticsMedical University of South CarolinaCharlestonSCUSA
| | - Douglas A Andres
- Department of Molecular and Cellular Biochemistry, College of MedicineUniversity of KentuckyLexingtonKYUSA
| | - Tom C Badgett
- Pediatric Hematology‐Oncology, College of MedicineUniversity of KentuckyLexingtonKYUSA
| | - Lars M Wagner
- Pediatric Hematology‐OncologyDuke UniversityDurhamNCUSA
| | - Derek B Allison
- Department of Pathology and Laboratory Medicine, College of MedicineUniversity of KentuckyLexingtonKYUSA
| | - Ramon C Sun
- Markey Cancer CenterUniversity of KentuckyLexingtonKYUSA
- Department of Neuroscience, College of MedicineUniversity of KentuckyLexingtonKYUSA
- Spinal Cord and Brain Injury Research CenterUniversity of KentuckyLexingtonKYUSA
- Department of Biochemistry & Molecular Biology, College of MedicineUniversity of FloridaGainesvilleFLUSA
- Center for Advanced Spatial Biomolecule ResearchUniversity of FloridaGainesvilleFLUSA
| | - Matthew S Gentry
- Department of Molecular and Cellular Biochemistry, College of MedicineUniversity of KentuckyLexingtonKYUSA
- Markey Cancer CenterUniversity of KentuckyLexingtonKYUSA
- Department of Biochemistry & Molecular Biology, College of MedicineUniversity of FloridaGainesvilleFLUSA
- Center for Advanced Spatial Biomolecule ResearchUniversity of FloridaGainesvilleFLUSA
| |
Collapse
|
18
|
Maimaiti A, Tuersunniyazi A, Meng X, Pei Y, Ji W, Feng Z, Jiang L, Wang Z, Kasimu M, Wang Y, Shi X. N6-methyladenosine RNA methylation regulator-related alternative splicing gene signature as prognostic predictor and in immune microenvironment characterization of patients with low-grade glioma. Front Genet 2022; 13:872186. [PMID: 35937991 PMCID: PMC9355308 DOI: 10.3389/fgene.2022.872186] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2022] [Accepted: 07/01/2022] [Indexed: 11/13/2022] Open
Abstract
Background: N6-methyladenosine (m6A) RNA methylation is an important epigenetic modification affecting alternative splicing (AS) patterns of genes to regulate gene expression. AS drives protein diversity and its imbalance may be an important factor in tumorigenesis. However, the clinical significance of m6A RNA methylation regulator-related AS in the tumor microenvironment has not been investigated in low-grade glioma (LGG). Methods: We used 12 m6A methylation modulatory genes (WTAP, FTO, HNRNPC, YTHDF2, YTHDF1, YTHDC2, ALKBH5, YTHDC1, ZC3H13, RBM15, METTL14, and METTL3) from The Cancer Genome Atlas (TCGA) database as well as the TCGA-LGG (n = 502) dataset of AS events and transcriptome data. These data were downloaded and subjected to machine learning, bioinformatics, and statistical analyses, including gene ontology (GO) and the Kyoto Encyclopedia of Genes and Genomes (KEGG) pathway enrichment analysis. Univariate Cox, the Least Absolute Shrinkage and Selection Operator (LASSO), and multivariable Cox regression were used to develop prognostic characteristics. Prognostic values were validated using Kaplan-Maier survival analysis, proportional risk models, ROC curves, and nomograms. The ESTIMATE package, TIMER database, CIBERSORT method, and ssGSEA algorithm in the R package were utilized to explore the role of the immune microenvironment in LGG. Lastly, an AS-splicing factor (SF) regulatory network was examined in the case of considering the role of SFs in regulating AS events. Results: An aggregate of 3,272 m6A regulator-related AS events in patients with LGG were screened using six machine learning algorithms. We developed eight AS prognostic characteristics based on splice subtypes, which showed an excellent prognostic prediction performance. Furthermore, quantitative prognostic nomograms were developed and showed strong validity in prognostic prediction. In addition, prognostic signatures were substantially associated with tumor immune microenvironment diversity, ICB-related genes, and infiltration status of immune cell subtypes. Specifically, UGP2 has better promise as a prognostic factor for LGG. Finally, splicing regulatory networks revealed the potential functions of SFs. Conclusion: The present research offers a novel perspective on the role of AS in m6A methylation. We reveal that m6A methylation regulator-related AS events can mediate tumor progression through the immune-microenvironment, which could serve as a viable biological marker for clinical stratification of patients with LGG so as to optimize treatment regimens.
Collapse
Affiliation(s)
- Aierpati Maimaiti
- Department of Neurosurgery, Neurosurgery Centre, The First Affiliated Hospital of Xinjiang Medical University, Urumqi, China
| | | | - Xianghong Meng
- Department of Neurosurgery, Shenzhen University General Hospital and Shenzhen University Clinical Medical Academy Centre, Shenzhen University, Shenzhen, China
| | - Yinan Pei
- Department of Neurosurgery, Neurosurgery Centre, The First Affiliated Hospital of Xinjiang Medical University, Urumqi, China
| | - Wenyu Ji
- Department of Neurosurgery, Neurosurgery Centre, The First Affiliated Hospital of Xinjiang Medical University, Urumqi, China
| | - Zhaohai Feng
- Department of Neurosurgery, Neurosurgery Centre, The First Affiliated Hospital of Xinjiang Medical University, Urumqi, China
| | - Lei Jiang
- Department of Neurosurgery, Neurosurgery Centre, The First Affiliated Hospital of Xinjiang Medical University, Urumqi, China
| | - Zengliang Wang
- Department of Neurosurgery, Neurosurgery Centre, The First Affiliated Hospital of Xinjiang Medical University, Urumqi, China
| | - Maimaitijiang Kasimu
- Department of Neurosurgery, Neurosurgery Centre, The First Affiliated Hospital of Xinjiang Medical University, Urumqi, China
| | - Yongxin Wang
- Department of Neurosurgery, Neurosurgery Centre, The First Affiliated Hospital of Xinjiang Medical University, Urumqi, China
- *Correspondence: Xin Shi, ; Yongxin Wang,
| | - Xin Shi
- Department of Neurosurgery, Shenzhen University General Hospital and Shenzhen University Clinical Medical Academy Centre, Shenzhen University, Shenzhen, China
- *Correspondence: Xin Shi, ; Yongxin Wang,
| |
Collapse
|
19
|
Liu P, Zhou Q, Li J. Integrated Multi-Omics Data Analysis Reveals Associations Between Glycosylation and Stemness in Hepatocellular Carcinoma. Front Oncol 2022; 12:913432. [PMID: 35814473 PMCID: PMC9259879 DOI: 10.3389/fonc.2022.913432] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2022] [Accepted: 05/25/2022] [Indexed: 12/14/2022] Open
Abstract
Background Glycosylation plays an essential role in driving the progression and treatment resistance of hepatocellular carcinoma (HCC). However, its function in regulating the acquisition and maintenance of the cancer stemness-like phenotype in HCC remains largely unknown. There is also very little known about how CAD and other potential glycosylation regulators may influence stemness. This study explores the relationship between glycosylation and stemness in HCC. Methods Gene set variance analysis (GSVA) was used to assess the TCGA pan-cancer enrichment in glycosylation-related pathways. Univariate, LASSO, and multivariate COX regression were then used to identify prognostic genes in the TCGA-LIHC and construct a prognostic signature. HCC patients were classified into high- and low-risk subgroups based on the signature. The relationship between gene expression profiles and stemness was confirmed using bulk and single-cell RNA-sequencing data. The role of CAD and other genes in regulating the stemness of HCC was also validated by RT-qPCR, CCK-8, and colony formation assay. Copy number variation (CNV), immune infiltration, and clinical features were further analyzed in different subgroups and subsequent gene expression profiles. Sensitive drugs were also screened. Results In the pan-cancer analysis, HCC was shown to have specific glycosylation alterations. Five genes, CAD, SLC51B, LGALS3, B3GAT3, and MT3, identified from 572 glycosylation-related genes, were used to construct a gene signature and predict HCC patient survival in the TCGA cohort. The results demonstrated a significant positive correlation between patients in the high-risk group and both elevated gene expression and HCC dedifferentiation status. A significant reduction in the stemness-related markers, CD24, CD44, CD20, FOXM1, and EpCAM, was found after the knockdown of CAD and other genes in HepG2 and Huh7 cells. Frequent mutations increased CNVs, immune-suppressive responses, and poor prognosis were also associated with the high-risk profile. The ICGC-LIRI-JP cohort confirmed a similar relationship between glycosylation-related subtypes and stemness. Finally, 84 sensitive drugs were screened for abnormal glycosylation of HCC, and carfilzomib was most highly correlated with CAD. Conclusions Glycosylation-related molecular subtypes are associated with HCC stemness and disease prognosis. These results provide new directions for further research on the relationship between glycosylation and stemness phenotypes.
Collapse
Affiliation(s)
- Peiyan Liu
- Department of Hepatology, Second People’s Clinical College of Tianjin Medical University, Tianjin, China
- Department of Hepatology, Tianjin Second People’s Hospital, Tianjin, China
| | - Qi Zhou
- Department of Gastroenterology, The Third Affiliated Hospital of Sun Yat-Sen University, Guangzhou, China
- Guangdong Provincial Key Laboratory of Liver Disease Research, Guangzhou, China
| | - Jia Li
- Department of Hepatology, Second People’s Clinical College of Tianjin Medical University, Tianjin, China
- Department of Hepatology, Tianjin Second People’s Hospital, Tianjin, China
- *Correspondence: Jia Li,
| |
Collapse
|
20
|
Goel K, Birdi U, Menaker S, Bannykh SI, Patel C. Atypical Choroid Plexus Papilloma of the Fourth Ventricle in an Adult: A Case Report. Cureus 2022; 14:e25256. [PMID: 35755561 PMCID: PMC9217547 DOI: 10.7759/cureus.25256] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 05/23/2022] [Indexed: 11/05/2022] Open
|
21
|
Increased levels of acidic free-N-glycans, including multi-antennary and fucosylated structures, in the urine of cancer patients. PLoS One 2022; 17:e0266927. [PMID: 35413075 PMCID: PMC9004742 DOI: 10.1371/journal.pone.0266927] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2022] [Accepted: 03/29/2022] [Indexed: 12/01/2022] Open
Abstract
We recently reported increased levels of urinary free-glycans in some cancer patients. Here, we focused on cancer related alterations in the levels of high molecular weight free-glycans. The rationale for this study was that branching, elongation, fucosylation and sialylation, which lead to increases in the molecular weight of glycans, are known to be up-regulated in cancer. Urine samples from patients with gastric cancer, pancreatic cancer, cholangiocarcinoma and colorectal cancer and normal controls were analyzed. The extracted free-glycans were fluorescently labeled with 2-aminopyridine and analyzed by multi-step liquid chromatography. Comparison of the glycan profiles revealed increased levels of glycans in some cancer patients. Structural analysis of the glycans was carried out by performing chromatography and mass spectrometry together with enzymatic or chemical treatments. To compare glycan levels between samples with high sensitivity and selectivity, simultaneous measurements by reversed-phase liquid chromatography-selected ion monitoring of mass spectrometry were also performed. As a result, three lactose-core glycans and 78 free-N-glycans (one phosphorylated oligomannose-type, four sialylated hybrid-type and 73 bi-, tri- and tetra-antennary complex-type structures) were identified. Among them, glycans with α1,3-fucosylation ((+/− sialyl) Lewis X), triply α2,6-sialylated tri-antennary structures and/or a (Man3)GlcNAc1-core displayed elevated levels in cancer patients. However, simple α2,3-sialylation and α1,6-core-fucosylation did not appear to contribute to the observed increase in the level of glycans. Interestingly, one tri-antennary free-N-glycan that showed remarkable elevation in some cancer patients contained a unique Glcβ1-4GlcNAc-core instead of the common GlcNAc2-core at the reducing end. This study provides further insights into free-glycans as potential tumor markers and their processing pathways in cancer.
Collapse
|
22
|
Ding DX, Wang Y, Yan W, Fu WN. MYCT1 alters the glycogen shunt by regulating selective translation of RACK1-mediated enzymes. iScience 2022; 25:103955. [PMID: 35281731 PMCID: PMC8908216 DOI: 10.1016/j.isci.2022.103955] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2021] [Revised: 01/13/2022] [Accepted: 02/16/2022] [Indexed: 11/09/2022] Open
Abstract
MYCT1 has been shown to function as a tumor suppressor in various tumors, but its role in metabolism has never been reported. Here, we showed that global inactivation of Myct1 in mice led to progressive accumulation of glycogen in the liver, which was accompanied by aberrant changes in intermediates of the glycogen metabolic pathway. Mechanistically, MYCT1 appeared to promote translation efficiency of PGM1, UGP2 and GSK3A in hepatic cells in a RACK1-dependent manner. Consequently, upregulation of the three enzymes enhanced the glycogen shunt. Our data reveal a critical role of MYCT1 as a switch for the glycogen shunt in tumor cells. Myct1 depletion causes glycogen accumulation in mouse liver MYCT1 affects glycogen shunt in tumor and normal cells MYCT1 regulates translation efficiency of glycogen enzymes MYCT1 alters the glycogen shunt in a RACK1 dependent manner
Collapse
|
23
|
Kim S, Wolfe A, Kim SE. Targeting cancer's sweet spot: UGP2 as a therapeutic vulnerability. Mol Cell Oncol 2021; 8:1990676. [PMID: 35419477 PMCID: PMC8997258 DOI: 10.1080/23723556.2021.1990676] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2021] [Revised: 10/01/2021] [Accepted: 10/04/2021] [Indexed: 06/14/2023]
Abstract
Understanding the mechanisms governing metabolic reprogramming that underlie potential vulnerabilities in cancer cells is key to developing novel therapeutic strategies. The catalytic enzyme UDP-glucose pyrophosphorylase 2 (UGP2) drives the production of UDP-glucose. Our recent work demonstrated the crucial role of UGP2 in cancer growth and its regulation of cellular metabolic processes.
Collapse
Affiliation(s)
- Sunghoon Kim
- Department of Biosystems and Biomedical Sciences, College of Health Sciences, Korea University, Seoul, Republic of Korea
- Department of Integrated Biomedical and Life Sciences, College of Health Sciences, Korea University, Seoul, Republic of Korea
| | - Andrew Wolfe
- Department of Biological Sciences, Hunter College of the City University of New York, New York, NY, USA
- Departments of Biology and Biochemistry, The Graduate Center of the City University of New York, New York, NY, USA
| | - Sung Eun Kim
- Department of Biosystems and Biomedical Sciences, College of Health Sciences, Korea University, Seoul, Republic of Korea
- Department of Integrated Biomedical and Life Sciences, College of Health Sciences, Korea University, Seoul, Republic of Korea
| |
Collapse
|
24
|
Pyrimidine Biosynthetic Enzyme CAD: Its Function, Regulation, and Diagnostic Potential. Int J Mol Sci 2021; 22:ijms221910253. [PMID: 34638594 PMCID: PMC8508918 DOI: 10.3390/ijms221910253] [Citation(s) in RCA: 36] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2021] [Revised: 09/18/2021] [Accepted: 09/19/2021] [Indexed: 01/10/2023] Open
Abstract
CAD (Carbamoyl-phosphate synthetase 2, Aspartate transcarbamoylase, and Dihydroorotase) is a multifunctional protein that participates in the initial three speed-limiting steps of pyrimidine nucleotide synthesis. Over the past two decades, extensive investigations have been conducted to unmask CAD as a central player for the synthesis of nucleic acids, active intermediates, and cell membranes. Meanwhile, the important role of CAD in various physiopathological processes has also been emphasized. Deregulation of CAD-related pathways or CAD mutations cause cancer, neurological disorders, and inherited metabolic diseases. Here, we review the structure, function, and regulation of CAD in mammalian physiology as well as human diseases, and provide insights into the potential to target CAD in future clinical applications.
Collapse
|