1
|
Xu J, Wang Y, Zhang J, Tang J, Zhou Z. The role of branched-chain amino acids in cardio-oncology: A review. Life Sci 2025; 372:123614. [PMID: 40189196 DOI: 10.1016/j.lfs.2025.123614] [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: 02/04/2025] [Revised: 03/18/2025] [Accepted: 04/01/2025] [Indexed: 04/26/2025]
Abstract
Cancer and cardiovascular diseases (CVDs) are global health challenges. In cancer patients, CVD is the second leading cause of death following disease progression. There are few specialized services for cardio-oncology patients worldwide currently. Branched-chain amino acids (BCAAs) are essential amino acids that promote protein synthesis and energy homeostasis. The disruption of BCAAs metabolism facilitates the development of cancer and CVDs while the benefit of BCAA supplement is full of controversy. In this review, we summarized BCAA-related studies in cardiometabolism, cancer and chemotherapy-induced cardiotoxicity, and provided our perspectives on the roles of BCAAs in cardio-oncology. We find that supplementation of BCAAs presents protective effects in cardiometabolic diseases, while the influence on cancer is intricate and varies across different types of cancers. Large-scale clinical studies are needed to understand the long-term effects of BCAA intake and its impact on different stages of the disease. BCAAs have potential to mitigate chemotherapy-induced cardiotoxicity. Continued research is still essential to understand the precise mechanisms, determine optimal dosage and timing, and assess the effectiveness of BCAA supplement in cardio-oncology, in particular clinical research.
Collapse
Affiliation(s)
- Jiaqi Xu
- Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Yu Wang
- Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China; Department of Cardiology, The First Hospital of Hebei Medical University, Hebei, China
| | - Jing Zhang
- State Key Laboratory of Pharmaceutical Biotechnology, The University of Hong Kong, Hong Kong; Department of Pharmacology and Pharmacy, LKS Faculty of Medicine, The University of Hong Kong, Hong Kong
| | - Jingyi Tang
- Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China.
| | - Zhongyan Zhou
- Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China; State Key Laboratory of Pharmaceutical Biotechnology, The University of Hong Kong, Hong Kong; Department of Pharmacology and Pharmacy, LKS Faculty of Medicine, The University of Hong Kong, Hong Kong.
| |
Collapse
|
2
|
Majid H, Kohli S, Islam SU, Nidhi. The role of branched chain aminotransferase in the interrelated pathways of type 2 diabetes mellitus and Alzheimer's disease. J Diabetes Metab Disord 2025; 24:90. [PMID: 40151764 PMCID: PMC11936868 DOI: 10.1007/s40200-025-01597-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/10/2024] [Accepted: 02/23/2025] [Indexed: 03/29/2025]
Abstract
Objectives This review assessed the role of Branched-Chain Amino Acid Transaminase (BCAT) enzymes in human metabolism, and their involvement in the catabolism of branched-chain amino acids (BCAAs) and exploring the association between Type 2 Diabetes Mellitus (T2DM) and Alzheimer's disease (AD) through insulin resistance. Methods The analysis involves a comprehensive literature review of recent research findings related to BCAT enzymes, BCAA metabolism, T2DM, and AD. Relevant studies and articles were identified through systematic searches in databases such as PubMed, ScienceDirect, and other scholarly resources. Inclusion criteria encompassed research articles, reviews, and studies published in peer-reviewed journals, with a focus on human metabolism, BCAT enzymes, and the interplay between BCAA metabolism, T2DM, and AD. Results The association between T2DM and AD suggests a potential metabolic link, particularly through dysregulated BCAA metabolism leading to insulin resistance. The impact of impaired insulin signaling is implicated in brain function and the accumulation of amyloid plaques facilitated by BCAT. Conclusion The identified link between BCAT, BCAA metabolism, T2DM, and AD suggests that disruptions in BCAT levels could serve as valuable indicators for early detection of insulin resistance and cognitive impairment as observed in Type 3 Diabetes which may present a promising therapeutic target.
Collapse
Affiliation(s)
- Haya Majid
- Department of Translational and Clinical Research, School of Chemical and Life Sciences, Jamia Hamdard, New Delhi, 110062 India
| | - Sunil Kohli
- Department of Medicine and Diabetes Unit, Hamdard Institute of Medical Sciences and Research, Jamia Hamdard, New Delhi, 110062 India
| | - Sajad Ul Islam
- Department of Medicine and Diabetes Unit, Hamdard Institute of Medical Sciences and Research, Jamia Hamdard, New Delhi, 110062 India
| | - Nidhi
- Department of Translational and Clinical Research, School of Chemical and Life Sciences, Jamia Hamdard, New Delhi, 110062 India
| |
Collapse
|
3
|
Dare ES, Newman RH, Conway ME, Dong M. Crystal structures of the phosphorylation mimics of human cytosolic branched chain aminotransferase. Arch Biochem Biophys 2025; 770:110479. [PMID: 40414328 DOI: 10.1016/j.abb.2025.110479] [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: 04/23/2025] [Revised: 05/20/2025] [Accepted: 05/22/2025] [Indexed: 05/27/2025]
Abstract
The phosphorylation sites of the human cytosolic Branched Chain Aminotransferase (hBCATc) mediated by mitogen-activated protein kinase (MAPK)/extracellular-signal-regulated-kinase 2 (ERK2, also known as MAPK1) were mapped. The crystal structures of the phosphorylation mimics at T33 and T36 were determined. The modified transaminase activity of these variants was analyzed. Although there were no major conformational changes in the phosphorylation mimics of hBCAT, a regional conformational change at the interdomain loop was observed mainly in mutant T33E. Consistently, when the catalytic turnovers of the T33E and T36E mutants were comparable to the wild type of hBCATc, the KM dropped significantly compared to the wild type, indicating a shift of the substrate binding affinity in the mutants. Taken together, this indicated the phosphorylation of hBCATc by ERK2 is affecting the hBCATc's transaminase activity.
Collapse
Affiliation(s)
- Elizabeth S Dare
- Department of Chemistry and Biochemistry, University of North Carolina Wilmington, Wilmington, NC, USA
| | - Robert H Newman
- Department of Biology, North Carolina A&T State University, Greensboro, NC, USA
| | - Myra E Conway
- College of Health, Psychology and Social Care, University of Derby, Derby, UK
| | - Ming Dong
- Department of Chemistry and Biochemistry, University of North Carolina Wilmington, Wilmington, NC, USA.
| |
Collapse
|
4
|
Pedersen S, Ali Mohamed A, Krzyslak H, Al-Kaabi LSSA, Abuhaweeleh MN, Al Moustafa AE, Ghabreau L, Vranic S, Honoré B. Proteomic analysis reveals potential biomarker candidates in serous ovarian tumors - a preliminary study. Contemp Oncol (Pozn) 2025; 29:77-92. [PMID: 40330446 PMCID: PMC12051873 DOI: 10.5114/wo.2025.149180] [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: 12/03/2024] [Accepted: 01/18/2025] [Indexed: 05/08/2025] Open
Abstract
Introduction Ovarian serous cystadenocarcinoma (SCA), a deadly gynecologic cancer, often goes undetected until the late stages. Tissue proteomics unveils disease heterogeneity, enhancing tumor classification and enabling personalized treatments tailored to individual expression profiles. Material and methods Tissue samples from 46 serous ovarian tumors were quantified using label-free liquid chromatography-tandem mass spectrometry. We identified 80 proteins differentiating SCA from borderline tumors, 277 distinguishing SCA from benign tumors, and 195 between borderline and benign tumors. Ingenuity pathway analysis revealed increased cell proliferation and RNA processing in SCA and borderline tumors compared to benign tumors, with SCA showing greater oxidative phosphorylation than borderline tumors. Results Our comparative analysis indicates that upregulated (ASS1 - argininosuccinate synthase 1, CAPS, PPA1, BCAT1, MCM4) and downregulated proteins (MUC5B, SLC4A1, tenascin-XB - TNXB, carbonic anhydrase 1, hemoglobin β) may offer a robust panel for distinguishing SCA from benign and borderline ovarian tumors, potentially aiding in early diagnosis and disease monitoring. The cancer-associated proteins pyridoxal dependent decarboxylase domain containing 1 (AUC: 0.83, 95% CI: 0.66-1), GFPT1 (AUC: 0.84, CI: 0.70-0.89), and HYOU1 (AUC: 0.84, CI: 0.70-0.98) significantly differentiated between low-grade (LGSCA) and high-grade serous cystadenocarcinoma (HGSCA). Low-grade SCA showed significantly greater levels of MZB1 (log2 fold change (FC): -1.951, p-value: 0.0258), CRABP2 (FC: -2.34, p-value: 0.0016), and BCAM (FC: -1.945, p-value: 0.0197) than borderline cancers. Conclusions Argininosuccinate synthase 1 and TNXB showed potential as markers of disease progression. Elevated ASS1 was observed in borderline, LGSCA, and HGSCA tumors compared to benign tumors, while TNXB levels progressively declined from benign to borderline, LGSCA, and HGSCA tumors. Our study pinpoints critical biomarkers in serous ovarian tumors for HGSCA progression.
Collapse
Affiliation(s)
- Shona Pedersen
- College of Medicine, QU Health, Qatar University, Doha, Qatar
| | | | - Hubert Krzyslak
- Department of Clinical Biochemistry, Aalborg University Hospital, Aalborg, Denmark
| | | | | | | | - Lina Ghabreau
- Faculty of Medicine, University of Aleppo, Aleppo, Syria
| | - Semir Vranic
- College of Medicine, QU Health, Qatar University, Doha, Qatar
| | - Bent Honoré
- Department of Biomedicine, Aarhus University, Aarhus, Denmark
| |
Collapse
|
5
|
Tsai MJM, Kao HJ, Chen HH, Yu CH, Chien YH, Hwu WL, Kwok PY, Lee NC, Yang YL. Optical genome mapping with whole genome sequencing identifies complex chromosomal structural variations in acute leukemia. Front Genet 2025; 16:1496847. [PMID: 40242470 PMCID: PMC12000080 DOI: 10.3389/fgene.2025.1496847] [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: 09/15/2024] [Accepted: 03/19/2025] [Indexed: 04/18/2025] Open
Abstract
Introduction Chromosomal structural variations (SVs) play an important role in the formation of human cancers, including leukemias. However, many complex SVs cannot be identified by conventional tools, including karyotyping, fluorescence in situ hybridization, microarrays, and multiplex ligation-dependent probe amplification (MLPA). Methods Optical genome mapping (OGM) and whole genome sequencing (WGS) were employed to analyze five leukemia samples with SVs detected by karyotyping, MLPA, and RNA sequencing (RNA-seq). OGM was performed using the Saphyr chip on a Bionano Saphyr system. Copy number variation and rare variant assembly analyses were performed with Bionano software v3.7. WGS was analyzed by the Manta program for SVs. Results The leukemia samples had an average of 477 insertions, 457 deletions, and 32 inversions, which were significantly greater than those of the normal blood samples (p = 0.016, 0.028, and 0.028, respectively). In Case 1, OGM detected a sequential translocation between chromosomes 5, 8, 12, and 21 and ETV6::RUNX1 and BCAT1::BAALC gene fusions. Case 2 had two pathogenic SVs and a BCR::ABL1 fusion. Case 3 had one pathogenic SV and an IGH::DUSP22 fusion. Case 4 had two pathogenic SVs and a CBFB::MYH11 fusion. Case 5 had an STIL::TAL1 fusion. All breakpoint sequences were defined by WGS. An IGH::DUX4 fusion previously found by RNA-seq in Case 3 was not confirmed because DUX4, which has multiple pseudogenes, was refractory to OGM and WGS analyses. Conclusion OGM is a fundamental tool that complements G-banding analysis in identifying complex SVs in leukemia samples, and WGS effectively closes the gaps in OGM mapping.
Collapse
Affiliation(s)
- Meng-Ju Melody Tsai
- Department of Pediatrics, National Taiwan University Hospital and College of Medicine, National Taiwan University, Taipei, Taiwan
- Department of Pediatrics, National Taiwan University Hospital Yunlin Branch, Yunlin, Taiwan
| | - Hsiao-Jung Kao
- Institute of Biomedical Sciences, Academia Sinica, Taipei, Taiwan
| | - Hsiao-Huei Chen
- Institute of Biomedical Sciences, Academia Sinica, Taipei, Taiwan
| | - Chih-Hsiang Yu
- Institute of Statistical Science Academia Sinica, Taipei, Taiwan
| | - Yin-Hsiu Chien
- Department of Medical Genetics, National Taiwan University Hospital, Taipei, Taiwan
| | - Wuh-Liang Hwu
- Department of Pediatrics, National Taiwan University Hospital and College of Medicine, National Taiwan University, Taipei, Taiwan
- Department of Medical Genetics, National Taiwan University Hospital, Taipei, Taiwan
- Center for Precision Medicine, China Medical University Hospital, China Medical University, Taichung, Taiwan
| | - Pui-Yan Kwok
- Institute of Biomedical Sciences, Academia Sinica, Taipei, Taiwan
- Cardiovascular Research Institute, Institute for Human Genetics, University of California, San Francisco, San Francisco, CA, United States
- Department of Dermatology, University of California, San Francisco, San Francisco, CA, United States
| | - Ni-Chung Lee
- Department of Medical Genetics, National Taiwan University Hospital, Taipei, Taiwan
| | - Yung-Li Yang
- Department of Pediatrics, National Taiwan University Hospital and College of Medicine, National Taiwan University, Taipei, Taiwan
- Department of Laboratory Medicine, National Taiwan University Hospital, and Department of Laboratory Medicine, College of Medicine, National Taiwan University, Taipei, Taiwan
- Department of Laboratory Medicine, National Taiwan University Cancer Center, Taipei, Taiwan
| |
Collapse
|
6
|
Tesoriere G, Pilesi E, De Rosa M, Giampaoli O, Patriarca A, Spagnoli M, Chiocciolini F, Tramonti A, Contestabile R, Sciubba F, Vernì F. Vitamin B6 deficiency produces metabolic alterations in Drosophila. Metabolomics 2025; 21:42. [PMID: 40123014 PMCID: PMC11930875 DOI: 10.1007/s11306-025-02236-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/16/2024] [Accepted: 02/11/2025] [Indexed: 03/25/2025]
Abstract
INTRODUCTION Pyridoxal 5'-phosphate (PLP), the biologically active form of vitamin B6 is involved in 4% of cellular enzymatic activities and its deficiency is responsible for or contributes to several human diseases. The study of underlying mechanisms is still in its infancy and requires suitable model organisms. In Drosophila the deficiency of vitamin B6 produces chromosome aberrations and hallmarks of human diseases including diabetes and cancer. However, the effects of vitamin B6 deficiency have never been examined at a metabolic level. OBJECTIVES This study evaluates the metabolic changes in vitamin B6 deficient Drosophila larvae with the aim of validating flies as a suitable model for diseases associated to vitamin B6 deficiency. METHODS To induce vitamin B6 deficiency we fed Drosophila wild type larvae with 4-deoxypyridoxine (4DP), a PLP antagonist. By HPLC analysis we verified that the 4DP treatment was effective in inducing vitamin B6 deficiency. Using an NMR-based metabolomic approach we compared the metabolites in larval extracts from untreated and 4DP-fed larvae. RESULTS The NMR spectra analysis identified quantitative differences for sixteen metabolites out of forty, including branched chain and aromatic amino acids, glucose, and lipids, thus revealing interesting possible associations with the phenotypes showed by vitamin B6 deficient flies. CONCLUSIONS Our results validate Drosophila as a suitable model to study in depth the molecular mechanisms underlying human diseases associated with vitamin B6 deficiency and confirmed that 4DP treatment is effective in inducing vitamin B6 deficiency.
Collapse
Affiliation(s)
- Giulia Tesoriere
- Department of Biology and Biotechnology "Charles Darwin", Sapienza University of Rome, 00185, Rome, Italy
| | - Eleonora Pilesi
- Department of Biology and Biotechnology "Charles Darwin", Sapienza University of Rome, 00185, Rome, Italy
| | - Michele De Rosa
- Department of Chemistry, Sapienza University of Rome, Piazzale Aldo Moro 5, 00185, Rome, Italy
- NMR-Based Metabolomics Laboratory (NMLab), Sapienza University of Rome, Rome, Italy
| | - Ottavia Giampaoli
- NMR-Based Metabolomics Laboratory (NMLab), Sapienza University of Rome, Rome, Italy
- Department of Environmental Biology, Sapienza University of Rome, Piazzale Aldo Moro 5, 00185, Rome, Italy
| | - Adriano Patriarca
- Department of Chemistry, Sapienza University of Rome, Piazzale Aldo Moro 5, 00185, Rome, Italy
- NMR-Based Metabolomics Laboratory (NMLab), Sapienza University of Rome, Rome, Italy
- Department of Environmental Biology, Sapienza University of Rome, Piazzale Aldo Moro 5, 00185, Rome, Italy
| | - Mariangela Spagnoli
- Department of Occupational and Environmental Medicine, Epidemiology and Hygiene, INAIL, Via Fontana Candida 1, 00078, Monte Porzio Catone, Italy
| | - Federica Chiocciolini
- Institute of Molecular Biology and Pathology, Consiglio Nazionale delle Ricerche, 00185, Rome, Italy
| | - Angela Tramonti
- Institute of Molecular Biology and Pathology, Consiglio Nazionale delle Ricerche, 00185, Rome, Italy
| | - Roberto Contestabile
- Department of Biochemical Sciences "A. Rossi Fanelli", Sapienza, University of Rome, 00185, Rome, Italy
| | - Fabio Sciubba
- NMR-Based Metabolomics Laboratory (NMLab), Sapienza University of Rome, Rome, Italy
- Department of Environmental Biology, Sapienza University of Rome, Piazzale Aldo Moro 5, 00185, Rome, Italy
| | - Fiammetta Vernì
- Department of Biology and Biotechnology "Charles Darwin", Sapienza University of Rome, 00185, Rome, Italy.
| |
Collapse
|
7
|
Nakatsuka N, Adler D, Jiang L, Hartman A, Cheng E, Klann E, Satija R. A Reproducibility Focused Meta-Analysis Method for Single-Cell Transcriptomic Case-Control Studies Uncovers Robust Differentially Expressed Genes. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2024.10.15.618577. [PMID: 39463993 PMCID: PMC11507907 DOI: 10.1101/2024.10.15.618577] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 10/29/2024]
Abstract
We assessed the reproducibility of differentially expressed genes (DEGs) in previously published Alzheimer's (AD), Parkinson's (PD), Schizophrenia (SCZ), and COVID-19 scRNA-seq studies. While transcriptional scores from DEGs of individual PD and COVID-19 datasets had moderate predictive power for case-control status of other datasets (AUC=0.77 and 0.75), genes from individual AD and SCZ datasets had poor predictive power (AUC=0.68 and 0.55). We developed a non-parametric meta-analysis method, SumRank, based on reproducibility of relative differential expression ranks across datasets, and found DEGs with improved predictive power (AUC=0.88, 0.91, 0.78, and 0.62). By multiple other metrics, specificity and sensitivity of these genes were substantially higher than those discovered by dataset merging and inverse variance weighted p-value aggregation methods. The DEGs revealed known and novel biological pathways, and we validate BCAT1 as down-regulated in AD mouse oligodendrocytes. Lastly, we evaluate factors influencing reproducibility of individual studies as a prospective guide for experimental design.
Collapse
|
8
|
Zhu X, Peng A, Zou Y, Li Y, Wei H, Zheng X, Zhao Y. Impact of Cold Stress on Hepatopancreas Transcriptomic and Metabolomic in Red Swamp Crayfish Procambarus clarkii. Int J Mol Sci 2025; 26:1221. [PMID: 39940993 PMCID: PMC11818106 DOI: 10.3390/ijms26031221] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/25/2024] [Revised: 01/26/2025] [Accepted: 01/29/2025] [Indexed: 02/16/2025] Open
Abstract
The aquaculture industry of red swamp crayfish (RSC), Procambarus clarkii, has grown significantly in recent decades due to increasing market demand. In China, low water temperatures, particularly during overwintering, pose a challenge, hindering the development of the RSC aquaculture industry in northern regions. Understanding the molecular mechanism of RSCs' responses to cold stress could be beneficial for its aquaculture practices. In this study, we exposed RSCs to 4 °C (T4) and 22 °C (T22: control) for 96 h. Transcriptomic and metabolomic analyses of hepatopancreas tissues were performed to identify key genes and metabolites that participate in cold stress response. A total of 787 differentially expressed genes (DEGs) and 198 differentially expressed metabolites (DEMs) were identified between T4 and T22. DEGs were significantly enriched in KEGG pathways related to carbohydrate, lipid, amino acid, and nucleotide metabolism, immunity, and signaling, while DEMs were significantly enriched in pathways associated with lipid and amino acid metabolism and membrane transport. The results indicated that cold stress altered RSCs' metabolism and their innate immune system. This study provides valuable information to increase our understanding of cold stress responses in RSCs.
Collapse
Affiliation(s)
- Xiaochen Zhu
- Hebei Key Laboratory of the Bohai Sea Fish Germplasm Resources Conservation and Utilization, Beidaihe Central Experiment Station, Chinese Academy of Fishery Sciences, Qinhuangdao 066100, China
- College of Animal Science and Veterinary Medicine, Shenyang Agricultural University, Shenyang 110866, China
- College of Science and Engineering, Flinders University, Adelaide, SA 5042, Australia
| | - Aidi Peng
- College of Animal Science and Veterinary Medicine, Shenyang Agricultural University, Shenyang 110866, China
| | - Yueying Zou
- College of Animal Science and Veterinary Medicine, Shenyang Agricultural University, Shenyang 110866, China
| | - Yingdong Li
- College of Animal Science and Veterinary Medicine, Shenyang Agricultural University, Shenyang 110866, China
| | - Hua Wei
- College of Animal Science and Veterinary Medicine, Shenyang Agricultural University, Shenyang 110866, China
| | - Xianhu Zheng
- Key Laboratory of Freshwater Aquatic Biotechnology and Breeding, Ministry of Agriculture and Rural Affairs, Heilongjiang Fisheries Research Institute, Chinese Academy of Fishery Sciences, Harbin 150070, China
| | - Yingying Zhao
- College of Animal Science and Veterinary Medicine, Shenyang Agricultural University, Shenyang 110866, China
| |
Collapse
|
9
|
Aden D, Sureka N, Zaheer S, Chaurasia JK, Zaheer S. Metabolic Reprogramming in Cancer: Implications for Immunosuppressive Microenvironment. Immunology 2025; 174:30-72. [PMID: 39462179 DOI: 10.1111/imm.13871] [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: 05/18/2024] [Revised: 10/07/2024] [Accepted: 10/09/2024] [Indexed: 10/29/2024] Open
Abstract
Cancer is a complex and heterogeneous disease characterised by uncontrolled cell growth and proliferation. One hallmark of cancer cells is their ability to undergo metabolic reprogramming, which allows them to sustain their rapid growth and survival. This metabolic reprogramming creates an immunosuppressive microenvironment that facilitates tumour progression and evasion of the immune system. In this article, we review the mechanisms underlying metabolic reprogramming in cancer cells and discuss how these metabolic alterations contribute to the establishment of an immunosuppressive microenvironment. We also explore potential therapeutic strategies targeting metabolic vulnerabilities in cancer cells to enhance immune-mediated anti-tumour responses. TRIAL REGISTRATION: ClinicalTrials.gov identifier: NCT02044861, NCT03163667, NCT04265534, NCT02071927, NCT02903914, NCT03314935, NCT03361228, NCT03048500, NCT03311308, NCT03800602, NCT04414540, NCT02771626, NCT03994744, NCT03229278, NCT04899921.
Collapse
Affiliation(s)
- Durre Aden
- Department of Pathology, Hamdard Institute of Medical Science and Research, New Delhi, India
| | - Niti Sureka
- Department of Pathology, Vardhman Mahavir Medical College and Safdarjung Hospital, New Delhi, India
| | - Samreen Zaheer
- Department of Radiotherapy, Jawaharlal Nehru Medical College, AMU, Aligarh, India
| | | | - Sufian Zaheer
- Department of Pathology, Vardhman Mahavir Medical College and Safdarjung Hospital, New Delhi, India
| |
Collapse
|
10
|
Singer M, Hamdy R, Elsayed TM, Husseiny MI. The Mechanisms and Therapeutic Implications of Metabolic Communication in the Tumor-Immune Microenvironment. METABOLIC DYNAMICS IN HOST-MICROBE INTERACTION 2025:291-315. [DOI: 10.1007/978-981-96-1305-2_12] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/06/2025]
|
11
|
Al-Jaber H, Al-Muraikhy S, Jabr A, Yousef A, Anwardeen NR, Elrayess MA, Al-Mansoori L. Comparing Methods for Induction of Insulin Resistance in Mouse 3T3-L1 Cells. Curr Diabetes Rev 2025; 21:1-12. [PMID: 38204253 DOI: 10.2174/0115733998263359231211044539] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/06/2023] [Revised: 10/26/2023] [Accepted: 10/30/2023] [Indexed: 01/12/2024]
Abstract
Cell culture plays a crucial role in addressing fundamental research questions, particularly in studying insulin resistance (IR) mechanisms. Multiple in vitro models are utilized for this purpose, but their technical distinctions and relevance to in vivo conditions remain unclear. This study aims to assess the effectiveness of existing in vitro models in inducing IR and their ability to replicate in vivo IR conditions. BACKGROUND Insulin resistance (IR) is a cellular condition linked to metabolic disorders. Despite the utility of cell culture in IR research, questions persist regarding the suitability of various models. This study seeks to evaluate these models' efficiency in inducing IR and their ability to mimic in vivo conditions. Insights gained from this research could enhance our understanding of model strengths and limitations, potentially advancing strategies to combat IR and related disorders. OBJECTIVE 1- Investigate the technical differences between existing cell culture models used to study molecular mediators of insulin resistance (IR). 2- Compare the effectiveness of present in vitro models in inducing insulin resistance (IR). 3- Assess the relevance of the existing cell culture models in simulating the in vivo conditions and environment that provoke the induction of insulin resistance (IR). METHODS AND MATERIAL In vitro, eight sets of 3T3-L1 cells were cultured until they reached 90% confluence. Subsequently, adipogenic differentiation was induced using a differentiation cocktail (media). These cells were then divided into four groups, with four subjected to normal conditions and the other four to hypoxic conditions. Throughout the differentiation process, each cell group was exposed to specific factors known to induce insulin resistance (IR). These factors included 2.5 nM tumor necrosis factor-alpha (TNFα), 20 ng/ml interleukin-6 (IL-6), 10 micromole 4-hydroxynonenal (4HNE), and high insulin (HI) at a concentration of 100 nM. To assess cell proliferation, DAPI staining was employed, and the expression of genes associated with various metabolic pathways affected by insulin resistance was investigated using Real-Time PCR. Additionally, insulin signaling was examined using the Bio-plex Pro cell signaling Akt panel. RESULTS We induced insulin resistance in 3T3-L1 cells using IL-6, TNFα, 4HNE, and high insulin in both hypoxic and normoxic conditions. Hypoxia increased HIF1a gene expression by approximately 30% (P<0.01). TNFα reduced cell proliferation by 10-20%, and chronic TNFα treatment significantly decreased mature adipocytes due to its cytotoxicity. We assessed the impact of insulin resistance (IR) on metabolic pathways, focusing on genes linked to branched-chain amino acid metabolism, detoxification, and chemotaxis. Notably, ALDH6A1 and MCCC1 genes, related to amino acid metabolism, were significantly affected under hypoxic conditions. TNFα treatment notably influenced MCP-1 and MCP-2 genes linked to chemotaxis, with remarkable increases in MCP-1 levels and MCP-2 expression primarily under hypoxia. Detoxification-related genes showed minimal impact, except for a significant increase in MAOA expression under acute hypoxic conditions with TNFα treatment. Additional genes displayed varying effects, warranting further investigation. To investigate insulin signaling's influence in vitro by IRinducing factors, we assessed phospho-protein levels. Our results reveal a significant p-Akt induction with chronic high insulin (10%) and acute TNFα (12%) treatment under hypoxia (both P<0.05). Other insulin resistance-related phospho-proteins (GSK3B, mTOR, PTEN) increased with IL-6, 4HNE, TNFα, and high insulin under hypoxia, while p-IRS1 levels remained unaffected. CONCLUSION In summary, different in vitro models using inflammatory, oxidative stress, and high insulin conditions under hypoxic conditions can capture various aspects of in vivo adipose tissue insulin resistance (IR). Among these models, acute TNFα treatment may offer the most robust approach for inducing IR in 3T3-L1 cells.
Collapse
Affiliation(s)
- Hend Al-Jaber
- Biomedical Research Center, Qatar University, Doha, Qatar
| | | | - Aldana Jabr
- Biomedical Sciences Department, College of Health Sciences, QU Health, Qatar University, Doha, Qatar
| | - Aisha Yousef
- Biomedical Sciences Department, College of Health Sciences, QU Health, Qatar University, Doha, Qatar
| | | | | | | |
Collapse
|
12
|
Bo T, Fujii J. Primary Roles of Branched Chain Amino Acids (BCAAs) and Their Metabolism in Physiology and Metabolic Disorders. Molecules 2024; 30:56. [PMID: 39795113 PMCID: PMC11721030 DOI: 10.3390/molecules30010056] [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: 11/19/2024] [Revised: 12/25/2024] [Accepted: 12/25/2024] [Indexed: 01/13/2025] Open
Abstract
Leucine, isoleucine, and valine are collectively known as branched chain amino acids (BCAAs) and are often discussed in the same physiological and pathological situations. The two consecutive initial reactions of BCAA catabolism are catalyzed by the common enzymes referred to as branched chain aminotransferase (BCAT) and branched chain α-keto acid dehydrogenase (BCKDH). BCAT transfers the amino group of BCAAs to 2-ketoglutarate, which results in corresponding branched chain 2-keto acids (BCKAs) and glutamate. BCKDH performs an oxidative decarboxylation of BCKAs, which produces their coenzyme A-conjugates and NADH. BCAT2 in skeletal muscle dominantly catalyzes the transamination of BCAAs. Low BCAT activity in the liver reduces the metabolization of BCAAs, but the abundant presence of BCKDH promotes the metabolism of muscle-derived BCKAs, which leads to the production of glucose and ketone bodies. While mutations in the genes responsible for BCAA catabolism are involved in rare inherited disorders, an aberrant regulation of their enzymatic activities is associated with major metabolic disorders such as diabetes, cardiovascular disease, and cancer. Therefore, an understanding of the regulatory process of metabolic enzymes, as well as the functions of the BCAAs and their metabolites, make a significant contribution to our health.
Collapse
Affiliation(s)
- Tomoki Bo
- Laboratory Animal Center, Institute for Promotion of Medical Science Research, Faculty of Medicine, Yamagata University, Yamagata 990-9585, Japan
| | - Junichi Fujii
- Department of Biochemistry and Molecular Biology, Graduate School of Medical Science, Yamagata University, Yamagata 990-9585, Japan
| |
Collapse
|
13
|
Lu M, Luo D, Zhang Z, Ouyang F, Shi Y, Hu C, Su H, Li Y, Zhang J, Gui Q, Yang TS. Branched-chain amino acid catabolism promotes M2 macrophage polarization. Front Immunol 2024; 15:1469163. [PMID: 39582859 PMCID: PMC11582057 DOI: 10.3389/fimmu.2024.1469163] [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: 07/23/2024] [Accepted: 10/24/2024] [Indexed: 11/26/2024] Open
Abstract
Introduction During an immune response, macrophages undergo systematic metabolic rewiring tailored to support their functions. Branched-chain amino acid (BCAA) metabolism has been reported to modulate macrophage function; however, its role in macrophage alternative activation remain unclear. We aimed to investigate the role of BCAA metabolism in macrophage alternative activation. Method The metabolomics of BMDM-derived M0 and M2 macrophages were analyzed using LC-MS. BCAAs were supplemented and genes involved in BCAA catabolism were inhibited during M2 macrophage polarization. The expression of M2 marker genes was assessed through RT-qPCR, immunofluorescence, and flow cytometry. Results and discussion Metabolomic analysis identified increased BCAA metabolism as one of the most significantly rewired pathways upon alternative activation. M2 macrophages had significantly lower BCAA levels compared to controls. BCAA supplementation promoted M2 macrophage polarization both in vitro and in vivo and increased oxidative phosphorylation in M2 macrophages. Blocking BCAA entry into mitochondria by knockdown of SLC25A44 inhibited M2 macrophage polarization. Furthermore, M2 macrophages polarization was suppressed by knockdown of Branched-chain amino-acid transaminase 2 (BCAT2) and branched chain keto acid dehydrogenase E1 subunit alpha (BCKDHA), both of which are key enzymes involved in BCAA oxidation. Overall, our findings suggest that BCAA catabolism plays an important role in polarization toward M2 macrophages.
Collapse
Affiliation(s)
- Manxi Lu
- Shanghai Key Laboratory of Metabolic Remodeling and Health, Institute of Metabolism and Integrative Biology, Fudan University, Shanghai, China
| | - Da Luo
- Shanghai Key Laboratory of Metabolic Remodeling and Health, Institute of Metabolism and Integrative Biology, Fudan University, Shanghai, China
| | - Zixuan Zhang
- Shanghai Key Laboratory of Metabolic Remodeling and Health, Institute of Metabolism and Integrative Biology, Fudan University, Shanghai, China
| | - Feng Ouyang
- Shanghai Key Laboratory of Metabolic Remodeling and Health, Institute of Metabolism and Integrative Biology, Fudan University, Shanghai, China
| | - Yihong Shi
- Shanghai Key Laboratory of Metabolic Remodeling and Health, Institute of Metabolism and Integrative Biology, Fudan University, Shanghai, China
| | - Changyong Hu
- Shanghai Key Laboratory of Metabolic Remodeling and Health, Institute of Metabolism and Integrative Biology, Fudan University, Shanghai, China
| | - Hang Su
- Shanghai Key Laboratory of Metabolic Remodeling and Health, Institute of Metabolism and Integrative Biology, Fudan University, Shanghai, China
| | - Yining Li
- Shanghai Key Laboratory of Metabolic Remodeling and Health, Institute of Metabolism and Integrative Biology, Fudan University, Shanghai, China
| | - Jiayi Zhang
- Shanghai Key Laboratory of Metabolic Remodeling and Health, Institute of Metabolism and Integrative Biology, Fudan University, Shanghai, China
| | - Qian Gui
- Shanghai Key Laboratory of Metabolic Remodeling and Health, Institute of Metabolism and Integrative Biology, Fudan University, Shanghai, China
| | - Tian-Shu Yang
- Shanghai Key Laboratory of Metabolic Remodeling and Health, Institute of Metabolism and Integrative Biology, Fudan University, Shanghai, China
- School of Life Sciences, Tianjian Laboratory of Advanced Biomedical Sciences, Zhengzhou University, Zhengzhou, Henan, China
- Shanghai Key Laboratory of Lung Inflammation and Injury, Department of Pulmonary Medicine, Zhongshan Hospital, Fudan University, Shanghai, China
| |
Collapse
|
14
|
Cao Q, Fan J, Zou J, Wang W. Multi-omics analysis identifies BCAT2 as a potential pan-cancer biomarker for tumor progression and immune microenvironment modulation. Sci Rep 2024; 14:23371. [PMID: 39375392 PMCID: PMC11458862 DOI: 10.1038/s41598-024-74441-1] [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: 05/17/2024] [Accepted: 09/26/2024] [Indexed: 10/09/2024] Open
Abstract
Branched-chain amino acid transaminase 2 (BCAT2) encodes a crucial protein involved in the initial catalysis of branched-chain amino acid (BCAA) catabolism, with emerging evidence suggesting its association with tumor progression. This study explores BCAT2 in a pan-cancer multi-omics context and evaluates its prognostic significance. We utilized a multi-database approach, analyzing cBioPortal for genetic alterations, RNA-Seq data from TCGA and GTEx for expression patterns, and RSEM for transcript analysis. Protein expression and interaction networks were assessed using the Human Protein Atlas, UniProt, and STRING. Prognostic value was determined through Cox regression analysis of TCGA clinical survival data, while immune cell infiltration across various cancers was examined using TCGA data and the TIMER2 platform. Our results revealed that BCAT2 alterations are primarily amplifications and is upregulated in various tumors, correlating with poor survival rates in several tumor types, including GBMLGG, LGG, and UVM. Elevated BCAT2 protein levels were common in pan-cancer, interacting with a range of metabolic enzymes. Additionally, BCAT2 expression significantly influenced CD4+ T cells, CD8+ T cells, and Treg cells infiltration, with varied correlations across cancer types. These findings indicate BCAT2 as a potential biomarker for cancer diagnosis and therapy, potentially regulating key metabolic and immune factors to mediate tumor progression and the microenvironment.
Collapse
Affiliation(s)
- Qixuan Cao
- School of Public Health, Chongqing Medical University, Chongqing, 400016, China
| | - Jie Fan
- School of Public Health, Chongqing Medical University, Chongqing, 400016, China
| | - Jian Zou
- School of Public Health, Chongqing Medical University, Chongqing, 400016, China.
| | - Wei Wang
- School of Public Health, Chongqing Medical University, Chongqing, 400016, China.
| |
Collapse
|
15
|
Materniak-Kornas M, Ropka-Molik K, Piórkowska K, Kowalik J, Szmatoła T, Sikora J, Kawęcka A, Kuźmak J. Identification of New Single Nucleotide Polymorphisms Potentially Related to Small Ruminant Lentivirus Infection Susceptibility in Goats Based on Data Selected from High-Throughput Sequencing. Pathogens 2024; 13:830. [PMID: 39452702 PMCID: PMC11510762 DOI: 10.3390/pathogens13100830] [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: 07/11/2024] [Revised: 09/18/2024] [Accepted: 09/23/2024] [Indexed: 10/26/2024] Open
Abstract
Small ruminant lentivirus (SRLV) infections are spread in the flocks of sheep and goats all over the world, causing economic loss. Although only a fraction of infected animals develop disease symptoms, all of them may shed the virus, causing uncontrolled spread of the infection. Antibodies against the virus can be detected in the blood of infected animals and are the main marker of infection. Additionally, in most infected animals, proviral DNA can also be detected, but at different levels. Due to the lack of treatment or vaccines, the most effective strategy to prevent SRLV infections are control programmes introduced by several countries based on the elimination of seropositive individuals from the flock. An alternative approach, which has currently become the rationale, is an identification of host factors which may predispose certain individuals or breeds to resistance or susceptibility to small ruminant lentivirus infection. In our work, attention was paid to goats of the Carpathian breed infected with SRLV. Available RNA-seq results from the blood of 12 goats with a determined level of SRLV proviral load were used to analyse single nucleotide polymorphisms (SNPs) by the variant calling method. Six SNPs within five genes (POU2AF1, BCAT2, TMEM154, PARP14, UBASH3A) were selected for genotyping to determine their association with the level of small ruminant lentivirus proviral DNA in a group of 60 goats. Interestingly, in seronegative individuals, only the TT genotype of the PARP14 gene was observed, while the TMEM154 CC genotype was found only in seropositive goats. Both genes may be considered potential markers for resistance/susceptibility to SRLV infection. In contrast, polymorphisms identified in POU2AF1 and UBASH3A genes seemed to be deleterious for respective protein functions; therefore, these genes are less likely to be recognised as resistance/susceptibility markers of SRLV infection.
Collapse
Affiliation(s)
| | - Katarzyna Ropka-Molik
- Department of Animal Molecular Biology, National Research Institute of Animal Production, Krakowska 1, 32-083 Balice, Poland
| | - Katarzyna Piórkowska
- Department of Animal Molecular Biology, National Research Institute of Animal Production, Krakowska 1, 32-083 Balice, Poland
| | - Joanna Kowalik
- Department of Biochemistry, National Veterinary Research Institute, 24-100 Pulawy, Poland
| | - Tomasz Szmatoła
- Department of Animal Molecular Biology, National Research Institute of Animal Production, Krakowska 1, 32-083 Balice, Poland
- Center for Experimental and Innovative Medicine, University of Agriculture in Krakow, Rędzina 1c, 30-248 Krakow, Poland
| | - Jacek Sikora
- Department of Sheep and Goat Breeding, National Research Institute of Animal Production, Krakowska 1, 32-083 Balice, Poland
| | - Aldona Kawęcka
- Department of Sheep and Goat Breeding, National Research Institute of Animal Production, Krakowska 1, 32-083 Balice, Poland
| | - Jacek Kuźmak
- Department of Biochemistry, National Veterinary Research Institute, 24-100 Pulawy, Poland
| |
Collapse
|
16
|
Liu N, Li C, Yan C, Yan HC, Jin BX, Yang HR, Jiang GY, Gong HD, Li JY, Ma SJ, Liu HL, Gao C. BCAT1 alleviates early brain injury by inhibiting ferroptosis through PI3K/AKT/mTOR/GPX4 pathway after subarachnoid hemorrhage. Free Radic Biol Med 2024; 222:173-186. [PMID: 38871197 DOI: 10.1016/j.freeradbiomed.2024.05.045] [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: 12/05/2023] [Revised: 02/22/2024] [Accepted: 05/27/2024] [Indexed: 06/15/2024]
Abstract
Regulation of the redox system by branched-chain amino acid transferase 1 (BCAT1) is of great significance in the occurrence and development of diseases, but the relationship between BCAT1 and subarachnoid hemorrhage (SAH) is still unknown. Ferroptosis, featured by iron-dependent lipid peroxidation accompanied by the depletion of glutathione peroxidase 4 (GPX4), has been implicated in the pathological process of early brain injury after subarachnoid hemorrhage. This study established SAH model by endovascular perforation and adding oxyhemoglobin (Hb) to HT22 cells and delved into the mechanism of BCAT1 in SAH-induced ferroptotic neuronal cell death. It was found that SAH-induced neuronal ferroptosis could be inhibited by BCAT1 overexpression (OE) in rats and HT22 cells, and BCAT1 OE alleviated neurological deficits and cognitive dysfunction in rats after SAH. In addition, the effect of BCAT1 could be reversed by the Ly294002, a specific inhibitor of the PI3K pathway. In summary, our present study indicated that BCAT1 OE alleviated early brain injury EBI after SAH by inhibiting neuron ferroptosis via activation of PI3K/AKT/mTOR pathway and the elevation of GPX4. These results suggested that BCAT1 was a promising therapeutic target for subarachnoid hemorrhage.
Collapse
Affiliation(s)
- Nan Liu
- Department of Neurosurgery, First Affiliated Hospital of Harbin Medical University, Harbin, China; Key Colleges and Universities Laboratory of Neurosurgery in Heilongjiang Province, Harbin, China; Institute of Neuroscience, Sino-Russian Medical Research Center, Harbin Medical University, Harbin, China
| | - Chen Li
- Department of Neurosurgery, First Affiliated Hospital of Harbin Medical University, Harbin, China; Key Colleges and Universities Laboratory of Neurosurgery in Heilongjiang Province, Harbin, China; Institute of Neuroscience, Sino-Russian Medical Research Center, Harbin Medical University, Harbin, China
| | - Cong Yan
- Department of Neurosurgery, First Affiliated Hospital of Harbin Medical University, Harbin, China; Key Colleges and Universities Laboratory of Neurosurgery in Heilongjiang Province, Harbin, China; Institute of Neuroscience, Sino-Russian Medical Research Center, Harbin Medical University, Harbin, China
| | - Hao-Chen Yan
- Department of Neurosurgery, First Affiliated Hospital of Harbin Medical University, Harbin, China; Key Colleges and Universities Laboratory of Neurosurgery in Heilongjiang Province, Harbin, China; Institute of Neuroscience, Sino-Russian Medical Research Center, Harbin Medical University, Harbin, China
| | - Bing-Xuan Jin
- Department of Neurosurgery, First Affiliated Hospital of Harbin Medical University, Harbin, China; Key Colleges and Universities Laboratory of Neurosurgery in Heilongjiang Province, Harbin, China; Institute of Neuroscience, Sino-Russian Medical Research Center, Harbin Medical University, Harbin, China
| | - Hong-Rui Yang
- Department of Neurosurgery, First Affiliated Hospital of Harbin Medical University, Harbin, China; Key Colleges and Universities Laboratory of Neurosurgery in Heilongjiang Province, Harbin, China; Institute of Neuroscience, Sino-Russian Medical Research Center, Harbin Medical University, Harbin, China
| | - Guang-You Jiang
- Department of Neurosurgery, First Affiliated Hospital of Harbin Medical University, Harbin, China; Key Colleges and Universities Laboratory of Neurosurgery in Heilongjiang Province, Harbin, China; Institute of Neuroscience, Sino-Russian Medical Research Center, Harbin Medical University, Harbin, China
| | - Hai-Dong Gong
- Department of Neurosurgery, First Affiliated Hospital of Harbin Medical University, Harbin, China; Key Colleges and Universities Laboratory of Neurosurgery in Heilongjiang Province, Harbin, China; Institute of Neuroscience, Sino-Russian Medical Research Center, Harbin Medical University, Harbin, China
| | - Ji-Yi Li
- Department of Neurosurgery, First Affiliated Hospital of Harbin Medical University, Harbin, China; Key Colleges and Universities Laboratory of Neurosurgery in Heilongjiang Province, Harbin, China; Institute of Neuroscience, Sino-Russian Medical Research Center, Harbin Medical University, Harbin, China
| | - Sheng-Ji Ma
- Department of Neurosurgery, First Affiliated Hospital of Harbin Medical University, Harbin, China; Key Colleges and Universities Laboratory of Neurosurgery in Heilongjiang Province, Harbin, China; Institute of Neuroscience, Sino-Russian Medical Research Center, Harbin Medical University, Harbin, China
| | - Huai-Lei Liu
- Department of Neurosurgery, First Affiliated Hospital of Harbin Medical University, Harbin, China; Key Colleges and Universities Laboratory of Neurosurgery in Heilongjiang Province, Harbin, China; Institute of Neuroscience, Sino-Russian Medical Research Center, Harbin Medical University, Harbin, China.
| | - Cheng Gao
- Department of Neurosurgery, First Affiliated Hospital of Harbin Medical University, Harbin, China; Key Colleges and Universities Laboratory of Neurosurgery in Heilongjiang Province, Harbin, China; Institute of Neuroscience, Sino-Russian Medical Research Center, Harbin Medical University, Harbin, China.
| |
Collapse
|
17
|
Reifenberg P, Zimmer A. Branched-chain amino acids: physico-chemical properties, industrial synthesis and role in signaling, metabolism and energy production. Amino Acids 2024; 56:51. [PMID: 39198298 PMCID: PMC11358235 DOI: 10.1007/s00726-024-03417-2] [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: 06/13/2024] [Accepted: 08/20/2024] [Indexed: 09/01/2024]
Abstract
Branched-chain amino acids (BCAAs)-leucine (Leu), isoleucine (Ile), and valine (Val)-are essential nutrients with significant roles in protein synthesis, metabolic regulation, and energy production. This review paper offers a detailed examination of the physico-chemical properties of BCAAs, their industrial synthesis, and their critical functions in various biological processes. The unique isomerism of BCAAs is presented, focusing on analytical challenges in their separation and quantification as well as their solubility characteristics, which are crucial for formulation and purification applications. The industrial synthesis of BCAAs, particularly using bacterial strains like Corynebacterium glutamicum, is explored, alongside methods such as genetic engineering aimed at enhancing production, detailing the enzymatic processes and specific precursors. The dietary uptake, distribution, and catabolism of BCAAs are reviewed as fundamental components of their physiological functions. Ultimately, their multifaceted impact on signaling pathways, immune function, and disease progression is discussed, providing insights into their profound influence on muscle protein synthesis and metabolic health. This comprehensive analysis serves as a resource for understanding both the basic and complex roles of BCAAs in biological systems and their industrial application.
Collapse
Affiliation(s)
- Philipp Reifenberg
- Merck Life Science KGaA, Upstream R&D, Frankfurter Strasse 250, 64293, Darmstadt, Germany
- Institute for Organic Chemistry and Biochemistry, Technische Universität Darmstadt, Alarich‑Weiss‑Strasse 4, 64287, Darmstadt, Germany
| | - Aline Zimmer
- Merck Life Science KGaA, Upstream R&D, Frankfurter Strasse 250, 64293, Darmstadt, Germany.
| |
Collapse
|
18
|
Wang Y, Zhao N, Meng Y, Chen J, Qi C, Hu X, Zhu H, Yang D, Zhang X, Ma H, Zhao J, Di T, Li P, Wang Y. Bcat2-Mediated Branched-Chain Amino Acid Catabolism Is Linked to the Aggravated Inflammation in Obese with Psoriasis Mice. Mol Nutr Food Res 2024; 68:e2300720. [PMID: 38581348 DOI: 10.1002/mnfr.202300720] [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: 10/09/2023] [Revised: 02/07/2024] [Indexed: 04/08/2024]
Abstract
SCOPE The global prevalence of obesity has significantly increased, presenting a major health challenge. High-fat diet (HFD)-induced obesity is closely related to the disease severity of psoriasis, but the mechanism is not fully understood. METHODS AND RESULTS The study utilizes the HFD-induced obesity model along with an imiquimod (IMQ)-induced psoriasis-like mouse model (HFD-IMQ) to conduct transcriptomics and metabolomic analyses. HFD-induced obese mice exhibits more severe psoriasis-like lesions compared to normal diet (ND)-IMQ mice. The expression of genes of the IL-17 signaling pathway (IL-17A, IL-17F, S100A9, CCL20, CXCL1) is significantly upregulated, leading to an accumulation of T cells and neutrophils in the skin. Moreover, the study finds that there is an inhibition of the branched-chain amino acids (BCAAs) catabolism pathway, and the key gene branched-chain amino transferase 2 (Bcat2) is significantly downregulated, and the levels of leucine, isoleucine, and valine are elevated in the HFD-IMQ mice. Furthermore, the study finds that the peroxisome proliferator-activated receptor gamma (PPAR γ) is inhibited, while STAT3 activity is promoted in HFD-IMQ mice. CONCLUSION HFD-induced obesity significantly amplifies IL-17 signaling and exacerbates psoriasis, with a potential role played by Bcat2-mediated BCAAs metabolism. The study suggests that BCAA catabolism and PPAR γ-STAT3 exacerbate inflammation in psoriasis with obesity.
Collapse
Affiliation(s)
- Yazhuo Wang
- Beijing Hospital of Traditional Chinese Medicine, Capital Medical University, Beijing Institute of Traditional, Chinese Medicine, Beijing Key Laboratory of Clinic and Basic Research with Traditional Chinese Medicine on Psoriasis, Beijing, 100010, China
- Capital Medical University, Beijing, 100069, China
| | - Ning Zhao
- Beijing Hospital of Traditional Chinese Medicine, Capital Medical University, Beijing Institute of Traditional, Chinese Medicine, Beijing Key Laboratory of Clinic and Basic Research with Traditional Chinese Medicine on Psoriasis, Beijing, 100010, China
- Capital Medical University, Beijing, 100069, China
| | - Yujiao Meng
- Beijing Hospital of Traditional Chinese Medicine, Capital Medical University, Beijing Institute of Traditional, Chinese Medicine, Beijing Key Laboratory of Clinic and Basic Research with Traditional Chinese Medicine on Psoriasis, Beijing, 100010, China
| | - Jia Chen
- Beijing Hospital of Traditional Chinese Medicine, Capital Medical University, Beijing Institute of Traditional, Chinese Medicine, Beijing Key Laboratory of Clinic and Basic Research with Traditional Chinese Medicine on Psoriasis, Beijing, 100010, China
- Beijing University of Chinese Medicine, Beijing, 100029, China
| | - Cong Qi
- Beijing Hospital of Traditional Chinese Medicine, Capital Medical University, Beijing Institute of Traditional, Chinese Medicine, Beijing Key Laboratory of Clinic and Basic Research with Traditional Chinese Medicine on Psoriasis, Beijing, 100010, China
| | - Xueqing Hu
- Beijing Hospital of Traditional Chinese Medicine, Capital Medical University, Beijing Institute of Traditional, Chinese Medicine, Beijing Key Laboratory of Clinic and Basic Research with Traditional Chinese Medicine on Psoriasis, Beijing, 100010, China
| | - Haoyue Zhu
- Beijing Hospital of Traditional Chinese Medicine, Capital Medical University, Beijing Institute of Traditional, Chinese Medicine, Beijing Key Laboratory of Clinic and Basic Research with Traditional Chinese Medicine on Psoriasis, Beijing, 100010, China
- Capital Medical University, Beijing, 100069, China
| | - Danyang Yang
- Beijing Hospital of Traditional Chinese Medicine, Capital Medical University, Beijing Institute of Traditional, Chinese Medicine, Beijing Key Laboratory of Clinic and Basic Research with Traditional Chinese Medicine on Psoriasis, Beijing, 100010, China
- Capital Medical University, Beijing, 100069, China
| | - Xiawei Zhang
- Beijing Hospital of Traditional Chinese Medicine, Capital Medical University, Beijing Institute of Traditional, Chinese Medicine, Beijing Key Laboratory of Clinic and Basic Research with Traditional Chinese Medicine on Psoriasis, Beijing, 100010, China
- Capital Medical University, Beijing, 100069, China
| | - Huike Ma
- Beijing Hospital of Traditional Chinese Medicine, Capital Medical University, Beijing Institute of Traditional, Chinese Medicine, Beijing Key Laboratory of Clinic and Basic Research with Traditional Chinese Medicine on Psoriasis, Beijing, 100010, China
- Capital Medical University, Beijing, 100069, China
| | - Jingxia Zhao
- Beijing Hospital of Traditional Chinese Medicine, Capital Medical University, Beijing Institute of Traditional, Chinese Medicine, Beijing Key Laboratory of Clinic and Basic Research with Traditional Chinese Medicine on Psoriasis, Beijing, 100010, China
| | - Tingting Di
- Beijing Hospital of Traditional Chinese Medicine, Capital Medical University, Beijing Institute of Traditional, Chinese Medicine, Beijing Key Laboratory of Clinic and Basic Research with Traditional Chinese Medicine on Psoriasis, Beijing, 100010, China
| | - Ping Li
- Beijing Hospital of Traditional Chinese Medicine, Capital Medical University, Beijing Institute of Traditional, Chinese Medicine, Beijing Key Laboratory of Clinic and Basic Research with Traditional Chinese Medicine on Psoriasis, Beijing, 100010, China
| | - Yan Wang
- Beijing Hospital of Traditional Chinese Medicine, Capital Medical University, Beijing Institute of Traditional, Chinese Medicine, Beijing Key Laboratory of Clinic and Basic Research with Traditional Chinese Medicine on Psoriasis, Beijing, 100010, China
| |
Collapse
|
19
|
Isaiah S, Loots DT, van Furth AMT, Davoren E, van Elsland S, Solomons R, van der Kuip M, Mason S. Urinary markers of Mycobacterium tuberculosis and dysbiosis in paediatric tuberculous meningitis cases undergoing treatment. Gut Pathog 2024; 16:14. [PMID: 38475868 PMCID: PMC10936073 DOI: 10.1186/s13099-024-00609-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/11/2023] [Accepted: 02/29/2024] [Indexed: 03/14/2024] Open
Abstract
BACKGROUND The pathogenesis of tuberculous meningitis (TBM) involves infection by Mycobacterium tuberculosis in the meninges and brain. However, recent studies have shown that the immune response and inflammatory processes triggered by TBM can have significant effects on gut microbiota. Disruptions in the gut microbiome have been linked to various systemic consequences, including altered immunity and metabolic dysregulation. Inflammation caused by TBM, antibiotic treatment, and changes in host immunity can all influence the composition of gut microbes. This complex relationship between TBM and the gut microbiome is of great importance in clinical settings. To gain a deeper understanding of the intricate interactions between TBM and the gut microbiome, we report innovative insights into the development of the disease in response to treatment. Ultimately, this could lead to improved outcomes, management strategies and quality of life for individuals affected by TBM. METHOD We used a targeted liquid chromatography-tandem mass spectrometry (LC-MS/MS) approach to investigate metabolites associated with gut metabolism in paediatric participants by analysing the urine samples collected from a control group (n = 40), and an experimental group (n = 35) with confirmed TBM, which were subdivided into TBM stage 1 (n = 8), stage 2 (n = 11) and stage 3 (n = 16). FINDINGS Our metabolomics investigation showed that, of the 78 initially selected compounds of microbiome origin, eight unique urinary metabolites were identified: 2-methylbutyrlglycine, 3-hydroxypropionic acid, 3-methylcrotonylglycine, 4-hydroxyhippuric acid, 5-hydroxyindoleacetic acid, 5-hydroxyhexanoic acid, isobutyrylglycine, and phenylacetylglutamine as urinary markers of dysbiosis in TBM. CONCLUSION These results - which are supported by previous urinary studies of tuberculosis - highlight the importance of gut metabolism and of identifying corresponding microbial metabolites as novel points for the foundation of improved management of TBM patients.
Collapse
Affiliation(s)
- Simon Isaiah
- Human Metabolomics, Faculty of Natural and Agricultural Sciences, North-West University, Potchefstroom, South Africa
| | - Du Toit Loots
- Human Metabolomics, Faculty of Natural and Agricultural Sciences, North-West University, Potchefstroom, South Africa
| | - A Marceline Tutu van Furth
- Vrije Universiteit, Pediatric Infectious Diseases and Immunology, Amsterdam University Medical Centers, Emma Children's Hospital, De Boelelaan 1117, Amsterdam, The Netherlands
| | - Elmarie Davoren
- Centre for Human Metabolomics, North-West University, Potchefstroom, South Africa
| | - Sabine van Elsland
- MRC Centre for Global Infectious Disease Analysis, Imperial College London, London, UK
- Department of Paediatrics and Child Health, Faculty of Medicine and Health Sciences, Stellenbosch University, Cape Town, South Africa
| | - Regan Solomons
- Department of Paediatrics and Child Health, Faculty of Medicine and Health Sciences, Stellenbosch University, Cape Town, South Africa
| | - Martijn van der Kuip
- Vrije Universiteit, Pediatric Infectious Diseases and Immunology, Amsterdam University Medical Centers, Emma Children's Hospital, De Boelelaan 1117, Amsterdam, The Netherlands
| | - Shayne Mason
- Human Metabolomics, Faculty of Natural and Agricultural Sciences, North-West University, Potchefstroom, South Africa.
| |
Collapse
|
20
|
You H, Song G, Xu Z, Chen S, Shen W, Liu H, Deng B, Li J, Huang G. HuR promotes castration-resistant prostate cancer progression by altering ERK5 activation via posttranscriptional regulation of BCAT1. J Transl Med 2024; 22:178. [PMID: 38369471 PMCID: PMC10874581 DOI: 10.1186/s12967-024-04970-w] [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: 11/24/2023] [Accepted: 02/09/2024] [Indexed: 02/20/2024] Open
Abstract
BACKGROUND Castration-resistant prostate cancer (CRPC) is refractory to hormone treatment, and the underlying mechanism has not been fully elucidated. This study aimed to clarify the role and mechanism of Human antigen R (HuR) as a therapeutic target for CRPC progression. METHODS HuR was knocked out by Cas9 or inhibited by the HuR-specific inhibitor KH-3 in CRPC cell lines and in a mouse xenograft model. The effects of HuR inhibition on tumour cell behaviors and signal transduction were examined by proliferation, transwell, and tumour xenograft assays. Posttranscriptional regulation of BCAT1 by HuR was determined by half-life and RIP assays. RESULTS HuR knockout attenuated the proliferation, migration, and invasion of PC3 and DU145 cells in vitro and inhibited tumour progression in vivo. Moreover, BCAT1 was a direct target gene of HuR and mediated the oncogenic effect of HuR on CRPC. Mechanistically, HuR directly interacted with BCAT1 mRNA and upregulated BCAT1 expression by increasing the stability and translation of BCAT1, which activated ERK5 signalling. Additionally, the HuR-specific inhibitor KH-3 attenuated CRPC progression by disrupting the HuR-BCAT1 interaction. CONCLUSIONS We confirmed that the HuR/BCAT1 axis plays a crucial role in CRPC progression and suggest that inhibiting the HuR/BCAT1 axis is a promising therapeutic approach for suppressing CRPC progression.
Collapse
Affiliation(s)
- Hang You
- Department of Urologic Oncology Surgery, Chongqing University Cancer Hospital, HanYu Road 181, Chongqing, 400030, China
- School of Medicine, Chongqing University, Chongqing, 400030, China
- Department of Biochemistry and Molecular Biology, College of Basic Medical Science, Army Medical University, GaoTanYan Main Street 30, Chongqing, 400038, China
| | - Guojing Song
- Department of Biochemistry and Molecular Biology, College of Basic Medical Science, Army Medical University, GaoTanYan Main Street 30, Chongqing, 400038, China
- Department of Urology, Southwest Hospital, Amy Medical University, Chongqing, 400038, China
| | - Zhizhen Xu
- Department of Biochemistry and Molecular Biology, College of Basic Medical Science, Army Medical University, GaoTanYan Main Street 30, Chongqing, 400038, China
| | - Saipeng Chen
- Department of Urology, Southwest Hospital, Amy Medical University, Chongqing, 400038, China
| | - Wenhao Shen
- Department of Urology, Southwest Hospital, Amy Medical University, Chongqing, 400038, China
| | - Heting Liu
- Department of Biochemistry and Molecular Biology, College of Basic Medical Science, Army Medical University, GaoTanYan Main Street 30, Chongqing, 400038, China
| | - Bingqian Deng
- Department of Biochemistry and Molecular Biology, College of Basic Medical Science, Army Medical University, GaoTanYan Main Street 30, Chongqing, 400038, China
| | - Jun Li
- Department of Urologic Oncology Surgery, Chongqing University Cancer Hospital, HanYu Road 181, Chongqing, 400030, China.
| | - Gang Huang
- Department of Biochemistry and Molecular Biology, College of Basic Medical Science, Army Medical University, GaoTanYan Main Street 30, Chongqing, 400038, China.
| |
Collapse
|
21
|
Mi Y, Shan H, Wang B, Tang H, Jia J, Liu X, Yang Q. Genipin inhibits proliferation of gastric cancer cells by inducing ferroptosis: an integrated study of network pharmacology and bioinformatics study. Med Oncol 2024; 41:46. [PMID: 38175425 DOI: 10.1007/s12032-023-02283-4] [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: 11/01/2023] [Accepted: 12/07/2023] [Indexed: 01/05/2024]
Abstract
Ferroptosis has been demonstrated to suppress cancer development and is targeted for cancer therapy. Genipin, an iridoid constituent in Gardeniae Fructus, has been reported to exert anti-cancer abilities. However, whether genipin could induce ferroptosis remains unclear. The purpose of this study is to explore the anti-gastric cancer (GC) effects of genipin by inducing ferroptosis and to identify the potential targets. CCK-8 and colony formation assays were performed to evaluate the anti-GC effects of genipin. Flowcytometry and western blot were used to indicate ferroptosis-inducing ability of genipin. The potential targets of genipin were analyzed by network pharmacology, screened using UALCAN and KM-plotter database and evaluated by molecular docking. The results showed that genipin inhibited cell viability and proliferation of GC cells. Genipin treatment decreased levels of GPX4 and SLC7A11, induced accumulation of lipid peroxidation intracellularly and led to ferroptosis in GC cells. Network pharmacology analysis identified that lipid- and ROS-related pathways involved in ferroptosis ranked high among genipin-GC common targets. Data from UALCAN and KM-plotter database demonstrated that expression levels of ferroptosis-related targets, including AURKA, BCAT2, DHODH, and GPI, increased in GC tissues and the higher levels of the above four targets were related to tumor stage, tumor grade, and poor prognosis. Among these four targets, AURKA, BCAT2, and DHODH were confirmed by molecular docking with binding energies less than - 5. Taken together, our study demonstrates that genipin could exert anti-GC ability by inducing ferroptosis and provides evidence for the potential application of genipin in GC treatment.
Collapse
Affiliation(s)
- Yalu Mi
- College of Traditional Chinese Medicine, Shandong University of Traditional Chinese Medicine, Jinan, 250355, Shandong, China
- Institute of Pathogen Biology, School of Basic Medical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, 250012, Shandong, China
| | - Hui Shan
- Institute of Pathogen Biology, School of Basic Medical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, 250012, Shandong, China
| | - Bo Wang
- Department of Traditional Medicine, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Jinan, 250012, Shandong, China
| | - Huidi Tang
- Institute of Pathogen Biology, School of Basic Medical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, 250012, Shandong, China
| | - Jihui Jia
- Institute of Pathogen Biology, School of Basic Medical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, 250012, Shandong, China
- Key Laboratory for Experimental Teratology of the Chinese Ministry of Education, School of Basic Medical Science, Cheeloo College of Medicine, Shandong University, Jinan, 250012, Shandong, China
- Shandong Key Laboratory of Infection and Immunity, Shandong University, Jinan, 250012, Shandong, China
| | - Xijian Liu
- College of Traditional Chinese Medicine, Shandong University of Traditional Chinese Medicine, Jinan, 250355, Shandong, China.
| | - Qing Yang
- Institute of Pathogen Biology, School of Basic Medical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, 250012, Shandong, China.
- Key Laboratory for Experimental Teratology of the Chinese Ministry of Education, School of Basic Medical Science, Cheeloo College of Medicine, Shandong University, Jinan, 250012, Shandong, China.
- Shandong Key Laboratory of Infection and Immunity, Shandong University, Jinan, 250012, Shandong, China.
| |
Collapse
|
22
|
Carneiro TJ, Carvalho ALMB, Vojtek M, Carmo IF, Marques MPM, Diniz C, Gil AM. Disclosing a metabolic signature of cisplatin resistance in MDA-MB-231 triple-negative breast cancer cells by NMR metabolomics. Cancer Cell Int 2023; 23:310. [PMID: 38057765 DOI: 10.1186/s12935-023-03124-0] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2023] [Accepted: 11/03/2023] [Indexed: 12/08/2023] Open
Abstract
This work compared the metabolic profile of a parental MDA-MB-231 cisplatin-sensitive triple negative breast cancer (TNBC) cell line with that of a derived cisplatin-resistant line, to characterize inherent metabolic adaptations to resistance, as a means for marker and new TNBC therapies discovery. Supported by cytotoxic, microscopic and biochemical characterization of both lines, Nuclear Magnetic Resonance (NMR) metabolomics was employed to characterize cell polar extracts for the two cell lines, as a function of time (0, 24 and 48 h), and identify statistically relevant differences both between sensitive and resistant cells and their time course behavior. Biochemical results revealed a slight increase in activation of the NF-κB pathway and a marked decrease of the ERK signaling pathway in resistant cells. This was accompanied by lower glycolytic and glutaminolytic activities, possibly linked to glutamine being required to increase stemness capacity and, hence, higher survival to cisplatin. The TCA cycle dynamics seemed to be time-dependent, with an apparent activation at 48 h preferentially supported by anaplerotic aromatic amino acids, leucine and lysine. A distinct behavior of leucine, compared to the other branched-chain-amino-acids, suggested the importance of the recognized relationship between leucine and in mTOR-mediated autophagy to increase resistance. Suggested markers of MDA-MB-231 TNBC cisplatin-resistance included higher phosphocreatine/creatine ratios, hypotaurine/taurine-mediated antioxidant protective mechanisms, a generalized marked depletion in nucleotides/nucleosides, and a distinctive pattern of choline compounds. Although the putative hypotheses generated here require biological demonstration, they pave the way to the use of metabolites as markers of cisplatin-resistance in TNBC and as guidance to develop therapies.
Collapse
Affiliation(s)
- Tatiana J Carneiro
- Department of Chemistry and CICECO -Aveiro Institute of Materials, University of Aveiro, 3810-193, Aveiro, Portugal
- Molecular Physical-Chemistry R&D Unit, Department of Chemistry, University of Coimbra, 3004-535, Coimbra, Portugal
- LAQV/REQUIMTE, Laboratory of Pharmacology, Department of Drug Sciences, Faculty of Pharmacy, University of Porto, 4150-755, Porto, Portugal
| | - Ana L M Batista Carvalho
- Molecular Physical-Chemistry R&D Unit, Department of Chemistry, University of Coimbra, 3004-535, Coimbra, Portugal
| | - Martin Vojtek
- LAQV/REQUIMTE, Laboratory of Pharmacology, Department of Drug Sciences, Faculty of Pharmacy, University of Porto, 4150-755, Porto, Portugal
| | - Inês F Carmo
- Department of Chemistry and CICECO -Aveiro Institute of Materials, University of Aveiro, 3810-193, Aveiro, Portugal
| | - Maria Paula M Marques
- Molecular Physical-Chemistry R&D Unit, Department of Chemistry, University of Coimbra, 3004-535, Coimbra, Portugal
- Department of Life Sciences, Faculty of Science and Technology, University of Coimbra, 3000-456, Coimbra, Portugal
| | - Carmen Diniz
- LAQV/REQUIMTE, Laboratory of Pharmacology, Department of Drug Sciences, Faculty of Pharmacy, University of Porto, 4150-755, Porto, Portugal.
| | - Ana M Gil
- Department of Chemistry and CICECO -Aveiro Institute of Materials, University of Aveiro, 3810-193, Aveiro, Portugal.
| |
Collapse
|
23
|
Sneha NP, Dharshini SAP, Taguchi YH, Gromiha MM. Investigating Neuron Degeneration in Huntington's Disease Using RNA-Seq Based Transcriptome Study. Genes (Basel) 2023; 14:1801. [PMID: 37761940 PMCID: PMC10530489 DOI: 10.3390/genes14091801] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2023] [Revised: 09/02/2023] [Accepted: 09/11/2023] [Indexed: 09/29/2023] Open
Abstract
Huntington's disease (HD) is a progressive neurodegenerative disorder caused due to a CAG repeat expansion in the huntingtin (HTT) gene. The primary symptoms of HD include motor dysfunction such as chorea, dystonia, and involuntary movements. The primary motor cortex (BA4) is the key brain region responsible for executing motor/movement activities. Investigating patient and control samples from the BA4 region will provide a deeper understanding of the genes responsible for neuron degeneration and help to identify potential markers. Previous studies have focused on overall differential gene expression and associated biological functions. In this study, we illustrate the relationship between variants and differentially expressed genes/transcripts. We identified variants and their associated genes along with the quantification of genes and transcripts. We also predicted the effect of variants on various regulatory activities and found that many variants are regulating gene expression. Variants affecting miRNA and its targets are also highlighted in our study. Co-expression network studies revealed the role of novel genes. Function interaction network analysis unveiled the importance of genes involved in vesicle-mediated transport. From this unified approach, we propose that genes expressed in immune cells are crucial for reducing neuron death in HD.
Collapse
Affiliation(s)
- Nela Pragathi Sneha
- Department of Biotechnology, Bhupat and Jyoti Mehta School of Biosciences, Indian Institute of Technology Madras, Chennai 600036, Tamil Nadu, India; (N.P.S.); (S.A.P.D.)
| | - S. Akila Parvathy Dharshini
- Department of Biotechnology, Bhupat and Jyoti Mehta School of Biosciences, Indian Institute of Technology Madras, Chennai 600036, Tamil Nadu, India; (N.P.S.); (S.A.P.D.)
| | - Y.-h. Taguchi
- Department of Physics, Chuo University, Kasuga, Bunkyo-ku, Tokyo 112-8551, Japan;
| | - M. Michael Gromiha
- Department of Biotechnology, Bhupat and Jyoti Mehta School of Biosciences, Indian Institute of Technology Madras, Chennai 600036, Tamil Nadu, India; (N.P.S.); (S.A.P.D.)
| |
Collapse
|
24
|
Chavdoula E, Anastas V, Ferlita AL, Aldana J, Carota G, Spampinato M, Soysal B, Cosentini I, Parashar S, Sircar A, Nigita G, Sehgal L, Freitas MA, Tsichlis PN. Transcriptional regulation of amino acid metabolism by KDM2B, in the context of ncPRC1.1 and in concert with MYC and ATF4. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.07.07.548031. [PMID: 37461630 PMCID: PMC10350079 DOI: 10.1101/2023.07.07.548031] [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: 07/29/2023]
Abstract
Introduction KDM2B encodes a JmjC domain-containing histone lysine demethylase, which functions as an oncogene in several types of tumors, including TNBC. This study was initiated to address the cancer relevance of the results of our earlier work, which had shown that overexpression of KDM2B renders mouse embryonic fibroblasts (MEFs) resistant to oxidative stress by regulating antioxidant mechanisms. Methods We mainly employed a multi-omics strategy consisting of RNA-Seq, quantitative TMT proteomics, Mass-spectrometry-based global metabolomics, ATAC-Seq and ChIP-seq, to explore the role of KDM2B in the resistance to oxidative stress and intermediary metabolism. These data and data from existing patient datasets were analyzed using bioinformatic tools, including exon-intron-split analysis (EISA), FLUFF and clustering analyses. The main genetic strategy we employed was gene silencing with shRNAs. ROS were measured by flow cytometry, following staining with CellROX and various metabolites were measured with biochemical assays, using commercially available kits. Gene expression was monitored with qRT-PCR and immunoblotting, as indicated. Results The knockdown of KDM2B in basal-like breast cancer cell lines lowers the levels of GSH and sensitizes the cells to ROS inducers, GSH targeting molecules, and DUB inhibitors. To address the mechanism of GSH regulation, we knocked down KDM2B in MDA-MB-231 cells and we examined the effects of the knockdown, using a multi-omics strategy. The results showed that KDM2B, functioning in the context of ncPRC1.1, regulates a network of epigenetic and transcription factors, which control a host of metabolic enzymes, including those involved in the SGOC, glutamate, and GSH metabolism. They also showed that KDM2B enhances the chromatin accessibility and expression of MYC and ATF4, and that it binds in concert with MYC and ATF4, the promoters of a large number of transcriptionally active genes, including many, encoding metabolic enzymes. Additionally, MYC and ATF4 binding sites were enriched in genes whose accessibility depends on KDM2B, and analysis of a cohort of TNBCs expressing high or low levels of KDM2B, but similar levels of MYC and ATF4 identified a subset of MYC targets, whose expression correlates with the expression of KDM2B. Further analyses of basal-like TNBCs in the same cohort, revealed that tumors expressing high levels of all three regulators exhibit a distinct metabolic signature that carries a poor prognosis. Conclusions The present study links KDM2B, ATF4, and MYC in a transcriptional network that regulates the expression of multiple metabolic enzymes, including those that control the interconnected SGOC, glutamate, and GSH metabolic pathways. The co-occupancy of the promoters of many transcriptionally active genes, by all three factors, the enrichment of MYC binding sites in genes whose chromatin accessibility depends on KDM2B, and the correlation of the levels of KDM2B with the expression of a subset of MYC target genes in tumors that express similar levels of MYC, suggest that KDM2B regulates both the expression and the transcriptional activity of MYC. Importantly, the concerted expression of all three factors also defines a distinct metabolic subset of TNBCs with poor prognosis. Overall, this study identifies novel mechanisms of SGOC regulation, suggests novel KDM2B-dependent metabolic vulnerabilities in TNBC, and provides new insights into the role of KDM2B in the epigenetic regulation of transcription.
Collapse
Affiliation(s)
- Evangelia Chavdoula
- Department of Cancer Biology and Genetics, The Ohio State University, Columbus, OH, United States
- The Ohio State University, Comprehensive Cancer Center, Columbus, OH, United States
| | - Vollter Anastas
- Department of Cancer Biology and Genetics, The Ohio State University, Columbus, OH, United States
- The Ohio State University, Comprehensive Cancer Center, Columbus, OH, United States
- Tufts Graduate School of Biomedical Sciences, Program in Genetics, Boston, MA, United States
| | - Alessandro La Ferlita
- Department of Cancer Biology and Genetics, The Ohio State University, Columbus, OH, United States
- The Ohio State University, Comprehensive Cancer Center, Columbus, OH, United States
| | - Julian Aldana
- Department of Cancer Biology and Genetics, The Ohio State University, Columbus, OH, United States
- The Ohio State University, Comprehensive Cancer Center, Columbus, OH, United States
| | - Giuseppe Carota
- Department of Biomedical and Biotechnological Sciences, University of Catania, Catania, Italy
| | - Mariarita Spampinato
- Department of Biomedical and Biotechnological Sciences, University of Catania, Catania, Italy
| | - Burak Soysal
- Department of Cancer Biology and Genetics, The Ohio State University, Columbus, OH, United States
- The Ohio State University, Comprehensive Cancer Center, Columbus, OH, United States
| | - Ilaria Cosentini
- Department of Clinical and Experimental Medicine, Bioinformatics Unit, University of Catania, Catania, Italy
| | - Sameer Parashar
- Department of Cancer Biology and Genetics, The Ohio State University, Columbus, OH, United States
- The Ohio State University, Comprehensive Cancer Center, Columbus, OH, United States
| | - Anuvrat Sircar
- The Ohio State University, Comprehensive Cancer Center, Columbus, OH, United States
- Division of Hematology, Department of Internal Medicine, The Ohio State University, Columbus, OH, United States
| | - Giovanni Nigita
- Department of Cancer Biology and Genetics, The Ohio State University, Columbus, OH, United States
- The Ohio State University, Comprehensive Cancer Center, Columbus, OH, United States
| | - Lalit Sehgal
- The Ohio State University, Comprehensive Cancer Center, Columbus, OH, United States
- Division of Hematology, Department of Internal Medicine, The Ohio State University, Columbus, OH, United States
| | - Michael A. Freitas
- Department of Cancer Biology and Genetics, The Ohio State University, Columbus, OH, United States
- The Ohio State University, Comprehensive Cancer Center, Columbus, OH, United States
| | - Philip N. Tsichlis
- Department of Cancer Biology and Genetics, The Ohio State University, Columbus, OH, United States
- The Ohio State University, Comprehensive Cancer Center, Columbus, OH, United States
| |
Collapse
|
25
|
Guo L, Hu C, Yao M, Han G. Mechanism of sorafenib resistance associated with ferroptosis in HCC. Front Pharmacol 2023; 14:1207496. [PMID: 37351514 PMCID: PMC10282186 DOI: 10.3389/fphar.2023.1207496] [Citation(s) in RCA: 39] [Impact Index Per Article: 19.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2023] [Accepted: 05/26/2023] [Indexed: 06/24/2023] Open
Abstract
Hepatocellular carcinoma (HCC) is the most familiar primary hepatic malignancy with a poor prognosis. The incidence of HCC and the associated deaths have risen in recent decades. Sorafenib is the first drug to be approved by the Food and Drug Administration (FDA) for routine use in the first-line therapy of patients with advanced HCC. However, only about 30% of patients with HCC will be benefited from sorafenib therapy, and drug resistance typically develops within 6 months. In recent years, the mechanisms of resistance to sorafenib have gained the attention of a growing number of researchers. A promising field of current studies is ferroptosis, which is a novel form of cell death differing from apoptosis, necroptosis, and autophagy. This process is dependent on the accumulation of intracellular iron and reactive oxygen species (ROS). Furthermore, the increase in intracellular iron levels and ROS can be significantly observed in cells resistant to sorafenib. This article reviews the mechanisms of resistance to sorafenib that are related to ferroptosis, evaluates the relationship between ferroptosis and sorafenib resistance, and explores new therapeutic approaches capable of reversing sorafenib resistance in HCC through the modulation of ferroptosis.
Collapse
|
26
|
Zagubnaya OA, Nartsissov YR. MOLECULAR MECHANISMS UNDERLYING THERAPEUTIC ACTION OF VITAMIN B6. PHARMACY & PHARMACOLOGY 2023. [DOI: 10.19163/2307-9266-2022-10-6-500-514] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/17/2023]
Abstract
The aim of the study was to analyze the molecular mechanisms that determine the possibility of using vitamin B6 in clinical practice for the correction of various pathological conditions.Materials and methods. Information retrieval (Scopus, PubMed) and library (eLibrary) databases were used as research tools. In some cases, the ResearchGate application was used for a semantic search. The analysis and generalization of the scientific literature on the topic of research, covering the period from 1989 to the present, has been carried out in the work.Results. It has been shown that all chemical forms of vitamin B6 are able to penetrate the membranes of most cells by free diffusion, while forming phosphorylated forms inside. Pyridoxal phosphate is a biologically important metabolite that is directly involved as a cofactor in a variety of intracellular reactions. Requirements for this cofactor depend on the age, sex and condition of the patient. Pregnancy and lactation play a special role in the consumption of vitamin B6. In most cases, a balanced diet will provide an acceptable level of this vitamin. At the same time, its deficiency leads to the development of a number of pathological conditions, including neurodegenerative diseases, inflammations and diabetes. Negative manifestations from the central nervous system are also possible with an excessive consumption of B6.Conclusion. Replenishment of the vitamin B6 level in case of its identified deficiency is a necessary condition for the successful treatment of the central nervous system diseases, diabetes and correction of patients’ immune status. At the same time, it is necessary to observe a balanced intake of this cofactor in order to avoid negative effects on metabolism in case of its excess.
Collapse
Affiliation(s)
- O. A. Zagubnaya
- Institute of Cytochemistry and Molecular Pharmacology;
Biomedical Research Group, BiDiPharma GmbH
| | - Y. R. Nartsissov
- Institute of Cytochemistry and Molecular Pharmacology;
Biomedical Research Group, BiDiPharma GmbH
| |
Collapse
|
27
|
Leung RWH, Lee TKW. Wnt/β-Catenin Signaling as a Driver of Stemness and Metabolic Reprogramming in Hepatocellular Carcinoma. Cancers (Basel) 2022; 14:cancers14215468. [PMID: 36358885 PMCID: PMC9656505 DOI: 10.3390/cancers14215468] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2022] [Revised: 10/30/2022] [Accepted: 11/03/2022] [Indexed: 11/09/2022] Open
Abstract
Simple Summary Aberrant Wnt/β-catenin signaling has been reported to play crucial role in pathogenesis of hepatocellular carcinoma (HCC). In this review, we focus on the regulatory role of Wnt/β-catenin signaling in cancer stemness and metabolic reprogramming, which are two emerging hallmarks of cancer. Understanding the role of Wnt/β-catenin signaling in regulation of the above processes reveals novel therapeutic strategy against this deadly disease. Abstract Hepatocellular carcinoma (HCC) is a major cause of cancer death worldwide due to its high rates of tumor recurrence and metastasis. Aberrant Wnt/β-catenin signaling has been shown to play a significant role in HCC development, progression and clinical impact on tumor behavior. Accumulating evidence has revealed the critical involvement of Wnt/β-catenin signaling in driving cancer stemness and metabolic reprogramming, which are regarded as emerging cancer hallmarks. In this review, we summarize the regulatory mechanism of Wnt/β-catenin signaling and its role in HCC. Furthermore, we provide an update on the regulatory roles of Wnt/β-catenin signaling in metabolic reprogramming, cancer stemness and drug resistance in HCC. We also provide an update on preclinical and clinical studies targeting Wnt/β-catenin signaling alone or in combination with current therapies for effective cancer therapy. This review provides insights into the current opportunities and challenges of targeting this signaling pathway in HCC.
Collapse
Affiliation(s)
- Rainbow Wing Hei Leung
- Department of Applied Biology and Chemical Technology, The Hong Kong Polytechnic University, Hong Kong, China
| | - Terence Kin Wah Lee
- Department of Applied Biology and Chemical Technology, The Hong Kong Polytechnic University, Hong Kong, China
- State Key Laboratory of Chemical Biology and Drug Discovery, The Hong Kong Polytechnic University, Hong Kong, China
- Correspondence: ; Tel.: +852-3400-8799; Fax: +852-2364-9932
| |
Collapse
|