1
|
Huang G, Lin Y, Zhao J, Zhang J, Du Y, Xiao M, Li H, Chen Z, Kang N, Khan IA, Liu Y, Huang B, Xu Q. Corosolic acid and its derivatives targeting MCCC1 against insulin resistance and their hypoglycemic effect on type 2 diabetic mice. Eur J Med Chem 2025; 284:117184. [PMID: 39731787 DOI: 10.1016/j.ejmech.2024.117184] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2024] [Revised: 12/15/2024] [Accepted: 12/16/2024] [Indexed: 12/30/2024]
Abstract
Corosolic acid (CA), a natural triterpenoid, exhibits various biological activities and is often called as plant-derived insulin due to its significant hypoglycemic effects, making it especially beneficial for individuals with diabetes or high blood glucose levels. However, CA has notable in vitro toxicity, low water solubility, and poor pharmacokinetic properties. To address these limitations, a series of CA derivatives were synthesized, resulting in the identification of derivative H26, which demonstrates a significantly enhanced hypoglycemic effect, reduced toxicity, and improved pharmacokinetic characteristics compared to CA. To identify the target protein of CA and investigate its therapeutic potential, a chemical probe derived from natural products, called CA-biotin, was designed and synthesized. By employing an avidin-biotin affinity binding system, we distinguished the differential protein bands between CA-biotin and biotin. This quantitative proteomic analysis revealed, for the first time, that the biotin-containing enzyme methylcrotonoyl-CoA carboxylase 1 (MCCC1) directly binds to CA. The interaction between H26 and MCCC1 was examined in vitro. The research on the mechanisms by which CA and H26 address Type 2 diabetes mellitus (T2DM) focused on the insulin resistance signaling pathway, specifically targeting MCCC1. The results indicated that H26 shows significant promise as a potential hypoglycemic agent, while MCCC1 may serve as a valuable target for addressing insulin resistance. This presents a promising opportunity for developing new medications aimed at improving the health of patients with type 2 diabetes mellitus (T2DM) or hyperglycemia.
Collapse
Affiliation(s)
- Guiyan Huang
- College of Pharmaceutical Sciences, Soochow University, Suzhou, Jiangsu, 215123, China
| | - Yu Lin
- College of Pharmaceutical Sciences, Soochow University, Suzhou, Jiangsu, 215123, China
| | - Jianping Zhao
- National Center for Natural Products Research, School of Pharmacy, University of Mississippi, University, MS, 38677, USA
| | - Junlei Zhang
- College of Pharmaceutical Sciences, Soochow University, Suzhou, Jiangsu, 215123, China
| | - Yexin Du
- College of Pharmaceutical Sciences, Soochow University, Suzhou, Jiangsu, 215123, China
| | - Mingyue Xiao
- College of Pharmaceutical Sciences, Soochow University, Suzhou, Jiangsu, 215123, China
| | - Heng Li
- College of Pharmaceutical Sciences, Soochow University, Suzhou, Jiangsu, 215123, China
| | - Zhong Chen
- College of Pharmaceutical Sciences, Soochow University, Suzhou, Jiangsu, 215123, China
| | - Naixin Kang
- College of Pharmaceutical Sciences, Soochow University, Suzhou, Jiangsu, 215123, China
| | - Ikhlas A Khan
- National Center for Natural Products Research, School of Pharmacy, University of Mississippi, University, MS, 38677, USA
| | - Yanli Liu
- College of Pharmaceutical Sciences, Soochow University, Suzhou, Jiangsu, 215123, China
| | - Bin Huang
- College of Pharmaceutical Sciences, Soochow University, Suzhou, Jiangsu, 215123, China.
| | - Qiongming Xu
- College of Pharmaceutical Sciences, Soochow University, Suzhou, Jiangsu, 215123, China.
| |
Collapse
|
2
|
Ishiyama M, Gotoh H, Oe S, Nomura T, Kitada M, Ono K. Glycogenolysis-Induced Astrocytic Serping1 Expression Regulates Neuroinflammatory Effects on Hippocampal neuron. Mol Neurobiol 2025; 62:1373-1387. [PMID: 38985256 DOI: 10.1007/s12035-024-04345-8] [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: 12/01/2023] [Accepted: 07/02/2024] [Indexed: 07/11/2024]
Abstract
The bacterial pathogen, lipopolysaccharide (LPS), elicits microglial response and induces cytokine secretion that subsequently activates astrocytes. Recent findings have indicated that LPS-induced activation of postnatal glial cells has led to alterations in synapse formation in hippocampal and cortical neurons, thereby resulting in a prolonged increased risk for seizure or depression. Nevertheless, its mechanisms remain to be fully elucidated. Cellular metabolism has recently gained recognition as a critical regulatory mechanism for the activation of peripheral immune cells, as it supplies the requisite energy and metabolite for their activation. In the present study, we report that LPS did not change the expression of reported astrocyte-derived synaptogenic genes in the postnatal hippocampus; however, it induced upregulation of astrocytic complement component regulator Serping1 within the postnatal hippocampus. As a regulatory mechanism, activation of glycogen degradation (glycogenolysis) governs the expression of a subset of inflammatory-responsive genes including Serping1 through reactive oxygen species (ROS)-NF-κB axis. Our study further demonstrated that glycogenolysis is implicated in neurotoxic phenotypes of astrocytes, such as impaired neuronal synaptogenesis or cellular toxicity. These findings suggested that activation of glycogenolysis in postnatal astrocytes is an essential metabolic pathway for inducing responses in inflammatory astrocytes.
Collapse
Affiliation(s)
- Masahito Ishiyama
- Department of Biology, Kyoto Prefectural University of Medicine, Inamori Building, 1-5 Shimogamo Hanki-Cho, Sakyo-Ku, Kyoto City, 606-0823, Japan
| | - Hitoshi Gotoh
- Department of Biology, Kyoto Prefectural University of Medicine, Inamori Building, 1-5 Shimogamo Hanki-Cho, Sakyo-Ku, Kyoto City, 606-0823, Japan.
| | - Souichi Oe
- Department of Anatomy, Kansai Medical University, 2-5-1 Shinmachi, Hirakata City, Osaka, 573-1010, Japan
| | - Tadashi Nomura
- Applied Biology, Kyoto Institute of Technology, 1-5 Matsugasaki Hashikami-Cho, Sakyo-Ku, Kyoto City, 606-8585, Japan
| | - Masaaki Kitada
- Department of Anatomy, Kansai Medical University, 2-5-1 Shinmachi, Hirakata City, Osaka, 573-1010, Japan
| | - Katsuhiko Ono
- Department of Biology, Kyoto Prefectural University of Medicine, Inamori Building, 1-5 Shimogamo Hanki-Cho, Sakyo-Ku, Kyoto City, 606-0823, Japan
| |
Collapse
|
3
|
Yi Y, Wang G, Zhang W, Yu S, Fei J, An T, Yi J, Li F, Huang T, Yang J, Niu M, Wang Y, Xu C, Xiao ZXJ. Mitochondrial-cytochrome c oxidase II promotes glutaminolysis to sustain tumor cell survival upon glucose deprivation. Nat Commun 2025; 16:212. [PMID: 39747079 PMCID: PMC11695821 DOI: 10.1038/s41467-024-55768-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2023] [Accepted: 12/20/2024] [Indexed: 01/04/2025] Open
Abstract
Glucose deprivation, a hallmark of the tumor microenvironment, compels tumor cells to seek alternative energy sources for survival and growth. Here, we show that glucose deprivation upregulates the expression of mitochondrial-cytochrome c oxidase II (MT-CO2), a subunit essential for the respiratory chain complex IV, in facilitating glutaminolysis and sustaining tumor cell survival. Mechanistically, glucose deprivation activates Ras signaling to enhance MT-CO2 transcription and inhibits IGF2BP3, an RNA-binding protein, to stabilize MT-CO2 mRNA. Elevated MT-CO2 increases flavin adenosine dinucleotide (FAD) levels in activating lysine-specific demethylase 1 (LSD1) to epigenetically upregulate JUN transcription, consequently promoting glutaminase-1 (GLS1) and glutaminolysis for tumor cell survival. Furthermore, MT-CO2 is indispensable for oncogenic Ras-induced glutaminolysis and tumor growth, and elevated expression of MT-CO2 is associated with poor prognosis in lung cancer patients. Together, these findings reveal a role for MT-CO2 in adapting to metabolic stress and highlight MT-CO2 as a putative therapeutic target for Ras-driven cancers.
Collapse
Affiliation(s)
- Yong Yi
- Center of Growth, Metabolism and Aging, Key Laboratory of Bio-Resource and Eco-Environment, Ministry of Education, College of Life Sciences, Sichuan University, Chengdu, China.
| | - Guoqiang Wang
- Center of Growth, Metabolism and Aging, Key Laboratory of Bio-Resource and Eco-Environment, Ministry of Education, College of Life Sciences, Sichuan University, Chengdu, China
| | - Wenhua Zhang
- Center of Growth, Metabolism and Aging, Key Laboratory of Bio-Resource and Eco-Environment, Ministry of Education, College of Life Sciences, Sichuan University, Chengdu, China
| | - Shuhan Yu
- Center of Growth, Metabolism and Aging, Key Laboratory of Bio-Resource and Eco-Environment, Ministry of Education, College of Life Sciences, Sichuan University, Chengdu, China
- Department of Oncology & Cancer Institute, Department of Laboratory Medicine and Sichuan Provincial Key Laboratory for Human Disease Gene Study, Sichuan Academy of Medical Sciences, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu, China
| | - Junjie Fei
- Center of Growth, Metabolism and Aging, Key Laboratory of Bio-Resource and Eco-Environment, Ministry of Education, College of Life Sciences, Sichuan University, Chengdu, China
| | - Tingting An
- Center of Growth, Metabolism and Aging, Key Laboratory of Bio-Resource and Eco-Environment, Ministry of Education, College of Life Sciences, Sichuan University, Chengdu, China
| | - Jianqiao Yi
- Center of Growth, Metabolism and Aging, Key Laboratory of Bio-Resource and Eco-Environment, Ministry of Education, College of Life Sciences, Sichuan University, Chengdu, China
| | - Fengtian Li
- School of Biosciences and Technology, Chengdu Medical College, Chengdu, China
| | - Ting Huang
- Department of Oncology & Cancer Institute, Department of Laboratory Medicine and Sichuan Provincial Key Laboratory for Human Disease Gene Study, Sichuan Academy of Medical Sciences, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu, China
| | - Jian Yang
- Center of Growth, Metabolism and Aging, Key Laboratory of Bio-Resource and Eco-Environment, Ministry of Education, College of Life Sciences, Sichuan University, Chengdu, China
| | - Mengmeng Niu
- Center of Growth, Metabolism and Aging, Key Laboratory of Bio-Resource and Eco-Environment, Ministry of Education, College of Life Sciences, Sichuan University, Chengdu, China
| | - Yang Wang
- Center of Growth, Metabolism and Aging, Key Laboratory of Bio-Resource and Eco-Environment, Ministry of Education, College of Life Sciences, Sichuan University, Chengdu, China.
| | - Chuan Xu
- Department of Oncology & Cancer Institute, Department of Laboratory Medicine and Sichuan Provincial Key Laboratory for Human Disease Gene Study, Sichuan Academy of Medical Sciences, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu, China.
| | - Zhi-Xiong Jim Xiao
- Center of Growth, Metabolism and Aging, Key Laboratory of Bio-Resource and Eco-Environment, Ministry of Education, College of Life Sciences, Sichuan University, Chengdu, China.
- Department of Oncology & Cancer Institute, Department of Laboratory Medicine and Sichuan Provincial Key Laboratory for Human Disease Gene Study, Sichuan Academy of Medical Sciences, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu, China.
- State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, China.
| |
Collapse
|
4
|
Zhang R, Dai F, Deng S, Zeng Y, Wang J, Liu G. Reprogramming of Glucose Metabolism for Revisiting Hepatocellular Carcinoma Resistance to Transcatheter Hepatic Arterial Chemoembolization. Chembiochem 2025; 26:e202400719. [PMID: 39501124 DOI: 10.1002/cbic.202400719] [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: 09/01/2024] [Revised: 11/04/2024] [Indexed: 11/24/2024]
Abstract
Hepatocellular carcinoma (HCC) is recognized globally as one of the most lethal tumors, presenting a significant menace to patients' lives owing to its exceptional aggressiveness and tendency to recur. Transcatheter hepatic arterial chemoembolization (TACE) therapy, as a first-line treatment option for patients with advanced HCC, has been proven effective. However, it is disheartening that nearly 40 % of patients exhibit resistance to this therapy. Consequently, this review delves into the metabolic aspects of glucose metabolism to explore the underlying mechanisms behind TACE treatment resistance and to propose potentially fruitful therapeutic strategies. The ultimate objective is to present novel insights for the development of personalized treatment methods targeting HCC.
Collapse
Affiliation(s)
- Ruijie Zhang
- State Key Laboratory of Vaccines for Infectious Diseases, Xiang An Biomedicine Laboratory, National Innovation Platform for Industry-Education Integration in Vaccine Research, State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics, Center for Molecular Imaging and Translational Medicine, School of Public Health, Xiamen University, Xiamen, 361102, China
| | - Fan Dai
- State Key Laboratory of Vaccines for Infectious Diseases, Xiang An Biomedicine Laboratory, National Innovation Platform for Industry-Education Integration in Vaccine Research, State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics, Center for Molecular Imaging and Translational Medicine, School of Public Health, Xiamen University, Xiamen, 361102, China
| | - Songhan Deng
- State Key Laboratory of Cellular Stress Biology, School of Life Sciences, Faculty of Medicine and Life Sciences, Xiamen University, Xiamen, 361102, China
| | - Yun Zeng
- State Key Laboratory of Vaccines for Infectious Diseases, Xiang An Biomedicine Laboratory, National Innovation Platform for Industry-Education Integration in Vaccine Research, State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics, Center for Molecular Imaging and Translational Medicine, School of Public Health, Xiamen University, Xiamen, 361102, China
| | - Jinyang Wang
- State Key Laboratory of Vaccines for Infectious Diseases, Xiang An Biomedicine Laboratory, National Innovation Platform for Industry-Education Integration in Vaccine Research, State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics, Center for Molecular Imaging and Translational Medicine, School of Public Health, Xiamen University, Xiamen, 361102, China
| | - Gang Liu
- State Key Laboratory of Vaccines for Infectious Diseases, Xiang An Biomedicine Laboratory, National Innovation Platform for Industry-Education Integration in Vaccine Research, State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics, Center for Molecular Imaging and Translational Medicine, School of Public Health, Xiamen University, Xiamen, 361102, China
| |
Collapse
|
5
|
Li X, Zhu D. Role of disulfide death in cancer (Review). Oncol Lett 2025; 29:55. [PMID: 39606569 PMCID: PMC11600708 DOI: 10.3892/ol.2024.14801] [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: 07/24/2024] [Accepted: 10/24/2024] [Indexed: 11/29/2024] Open
Abstract
The research field of regulated cell death is growing extensively. Following the recognition of ferroptosis, other unique and distinct forms of regulated cell death, including cuproptosis and disulfide death, have been identified. Disulfide death occurs due to the abnormal accumulation of disulfides within cells in environments lacking glucose, leading to contraction of the actin cytoskeleton, which ultimately triggers various signaling pathways and cell death. The induction of disulfide death in the treatment of cancer may exhibit significant therapeutic potential. Therefore, in the present review, a comprehensive and critical analysis of the current understanding of the molecular mechanisms and regulatory networks of disulfide death is presented. In addition, the potential physiological functions of disulfide death in tumor suppression and immune surveillance as well as its pathological roles and therapeutic potential are described. The core focus areas for future research into this form of cell death are also explored. Given the current lack of extensive clinical findings and well-defined key concepts, these may be regarded as pivotal points of interest in future studies.
Collapse
Affiliation(s)
- Xue Li
- Oncology Department, The Third Affiliated Hospital of Soochow University, Changzhou, Jiangsu 213000, P.R. China
| | - Danxia Zhu
- Oncology Department, The Third Affiliated Hospital of Soochow University, Changzhou, Jiangsu 213000, P.R. China
| |
Collapse
|
6
|
Brown SD, Klimi E, Bakker WAM, Beqqali A, Baker AH. Non-coding RNAs to treat vascular smooth muscle cell dysfunction. Br J Pharmacol 2025; 182:246-280. [PMID: 38773733 DOI: 10.1111/bph.16409] [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: 11/17/2023] [Revised: 02/19/2024] [Accepted: 03/14/2024] [Indexed: 05/24/2024] Open
Abstract
Vascular smooth muscle cell (vSMC) dysfunction is a critical contributor to cardiovascular diseases, including atherosclerosis, restenosis and vein graft failure. Recent advances have unveiled a fascinating range of non-coding RNAs (ncRNAs) that play a pivotal role in regulating vSMC function. This review aims to provide an in-depth analysis of the mechanisms underlying vSMC dysfunction and the therapeutic potential of various ncRNAs in mitigating this dysfunction, either preventing or reversing it. We explore the intricate interplay of microRNAs, long-non-coding RNAs and circular RNAs, shedding light on their roles in regulating key signalling pathways associated with vSMC dysfunction. We also discuss the prospects and challenges associated with developing ncRNA-based therapies for this prevalent type of cardiovascular pathology. LINKED ARTICLES: This article is part of a themed issue Non-coding RNA Therapeutics. To view the other articles in this section visit http://onlinelibrary.wiley.com/doi/10.1111/bph.v182.2/issuetoc.
Collapse
MESH Headings
- Animals
- Humans
- Cardiovascular Diseases/drug therapy
- Cardiovascular Diseases/genetics
- Cardiovascular Diseases/metabolism
- Cardiovascular Diseases/pathology
- Muscle, Smooth, Vascular/cytology
- Muscle, Smooth, Vascular/drug effects
- Muscle, Smooth, Vascular/metabolism
- Myocytes, Smooth Muscle/drug effects
- Myocytes, Smooth Muscle/metabolism
- Myocytes, Smooth Muscle/pathology
- RNA, Circular/genetics
- RNA, Circular/metabolism
- RNA, Long Noncoding/genetics
- RNA, Long Noncoding/metabolism
- RNA, Untranslated/genetics
- RNA, Untranslated/metabolism
- RNA, Untranslated/pharmacology
- RNA, Untranslated/therapeutic use
Collapse
Affiliation(s)
- Simon D Brown
- BHF Centre for Cardiovascular Science, Queens Medical Research Institute, University of Edinburgh, Edinburgh, UK
| | - Eftychia Klimi
- BHF Centre for Cardiovascular Science, Queens Medical Research Institute, University of Edinburgh, Edinburgh, UK
| | | | - Abdelaziz Beqqali
- BHF Centre for Cardiovascular Science, Queens Medical Research Institute, University of Edinburgh, Edinburgh, UK
| | - Andrew H Baker
- BHF Centre for Cardiovascular Science, Queens Medical Research Institute, University of Edinburgh, Edinburgh, UK
- Cardiovascular Research Institute Maastricht (CARIM), Maastricht University Medical Centre, Maastricht, The Netherlands
| |
Collapse
|
7
|
Mahenge CM, Akasheh RT, Kinder B, Nguyen XV, Kalam F, Cheng TYD. CT-Scan-Assessed Body Composition and Its Association with Tumor Protein Expression in Endometrial Cancer: The Role of Muscle and Adiposity Quantities. Cancers (Basel) 2024; 16:4222. [PMID: 39766121 PMCID: PMC11674723 DOI: 10.3390/cancers16244222] [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: 11/03/2024] [Revised: 12/04/2024] [Accepted: 12/16/2024] [Indexed: 01/11/2025] Open
Abstract
Background: Endometrial cancer is strongly associated with obesity, and tumors often harbor mutations in major cancer signaling pathways. To inform the integration of body composition into targeted therapy paradigms, this hypothesis-generating study explores the association between muscle mass, body fat, and tumor proteomics. Methods: We analyzed data from 113 patients in The Cancer Genome Atlas (TCGA) and Cancer Proteomic Tumor Analysis Consortium (CPTAC) cohorts and their corresponding abdominal CT scans. Among these patients, tumor proteomics data were available for 45 patients, and 133 proteins were analyzed. Adiposity and muscle components were assessed at the L3 vertebral level on the CT scans. Patients were stratified into tertiles of muscle and fat mass and categorized into three groups: high muscle/low adiposity, high muscle/high adiposity, and low muscle/all adiposities. Linear and Cox regression models were adjusted for study cohort, stage, histology type, age, race, and ethnicity. Results: Compared with the high-muscle/low-adiposity group, both the high-muscle/high-adiposity (HR = 4.3, 95% CI = 1.0-29.0) and low-muscle (HR = 4.4, 95% CI = 1.3-14.9) groups experienced higher mortality. Low muscle was associated with higher expression of phospho-4EBP1(T37 and S65), phospho-GYS(S641) and phospho-MAPK(T202/Y204) but lower expression of ARID1A, CHK2, SYK, LCK, EEF2, CYCLIN B1, and FOXO3A. High muscle/high adiposity was associated with higher expression of phospho-4EBP1 (T37), phospho-GYS (S641), CHK1, PEA15, SMAD3, BAX, DJ1, GYS, PKM2, COMPLEX II Subunit 30, and phospho-P70S6K (T389) but with lower expression of CHK2, CRAF, MSH6, TUBERIN, PR, ERK2, beta-CATENIN, AKT, and S6. Conclusions: These findings demonstrate an association between body composition and proteins involved in key cancer signaling pathways, notably the PI3K/AKT/MTOR, MAPK/ERK, cell cycle regulation, DNA damage response, and mismatch repair pathways. These findings warrant further validation and assessment in relation to prognosis and outcomes in these patients.
Collapse
Affiliation(s)
- Cuthbert Mario Mahenge
- Division of Cancer Control and Prevention, Department of Internal Medicine, College of Medicine, The Ohio State University, 3650 Olentangy River Rd., Suite 200, Columbus, OH 43214, USA; (C.M.M.); (R.T.A.); (F.K.)
| | - Rand Talal Akasheh
- Division of Cancer Control and Prevention, Department of Internal Medicine, College of Medicine, The Ohio State University, 3650 Olentangy River Rd., Suite 200, Columbus, OH 43214, USA; (C.M.M.); (R.T.A.); (F.K.)
| | - Ben Kinder
- Division of Cancer Control and Prevention, Department of Internal Medicine, College of Medicine, The Ohio State University, 3650 Olentangy River Rd., Suite 200, Columbus, OH 43214, USA; (C.M.M.); (R.T.A.); (F.K.)
| | - Xuan Viet Nguyen
- Department of Radiology, College of Medicine, The Ohio State University, 395 W 12th Ave., Suite 486, Columbus, OH 43210, USA;
| | - Faiza Kalam
- Division of Cancer Control and Prevention, Department of Internal Medicine, College of Medicine, The Ohio State University, 3650 Olentangy River Rd., Suite 200, Columbus, OH 43214, USA; (C.M.M.); (R.T.A.); (F.K.)
| | - Ting-Yuan David Cheng
- Division of Cancer Control and Prevention, Department of Internal Medicine, College of Medicine, The Ohio State University, 3650 Olentangy River Rd., Suite 200, Columbus, OH 43214, USA; (C.M.M.); (R.T.A.); (F.K.)
| |
Collapse
|
8
|
Jeganathan A, Arunachalam K, Byju A, Rani George A, Sajeev S, Thangasamy K, Natesan G. Chitosan Nanoparticle-Mediated Delivery of Alstonia venenata R.Br. Root Methanolic Extract: A Promising Strategy for Breast Cancer Therapy in DMBA-Induced Breast Cancer in Sprague Dawley Rats. Antioxidants (Basel) 2024; 13:1513. [PMID: 39765841 PMCID: PMC11673636 DOI: 10.3390/antiox13121513] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2024] [Revised: 12/05/2024] [Accepted: 12/06/2024] [Indexed: 01/11/2025] Open
Abstract
Alstonia venenata R.Br., a plant native to the Western Ghats, is recognized for its diverse medicinal properties. The plant's extracts, particularly rich in alkaloids and other bioactive compounds, have shown potential anticancer activity. This study investigates the therapeutic potential of chitosan nanoparticles (CNPs) loaded with the root methanolic extract (RME) of A. venenata in combating breast cancer induced by dimethylbenz(a)anthracene (DMBA) in female Sprague Dawley rats. The RME-loaded chitosan nanoparticles (RME-EnCNPs) were synthesized and characterized, and their in vivo efficacy was evaluated. Treatment with RME-EnCNPs significantly inhibited tumor progression, which is evidenced by reduced tumor volume, burden, and incidence. Moreover, the nanoparticles demonstrated a sustained release of the active compounds, leading to marked improvements in various biochemical, enzymatic, and histopathological parameters. The study found that both RME and RME-EnCNPs effectively suppressed tumor growth, with RME-EnCNPs showing superior efficacy in modulating tumor progression. Antioxidant assays revealed that treatment with RME-EnCNPs (500 mg/kg) resulted in significant increases in total protein, superoxide dismutase (SOD), catalase, glutathione peroxidase (GPx), and glutathione (GSH) levels, alongside a marked reduction in lipid peroxidation (LPO) (p < 0.001). These findings suggest that RME-EnCNPs exert a potent antioxidant effect, mitigating oxidative stress within the tumor microenvironment. The root extract of A. venenata and its nanoparticle formulation hold promise as a potential therapeutic agent for breast cancer, warranting further investigation to isolate active bioactive compounds and elucidate their mechanisms of action.
Collapse
Affiliation(s)
- Aarthi Jeganathan
- Department of Botany, Bharathiar University, Coimbatore 641046, TN, India; (A.J.); (A.B.); (A.R.G.); (S.S.); (K.T.)
| | - Karuppusamy Arunachalam
- Center for Studies in Stem Cells, Cellular Therapy and Toxicological Genetics (CeTroGen), Faculty of Medicine (FAMED), Federal University of Mato Grosso do Sul (UFMS), Campo Grande 79070-900, MS, Brazil;
| | - Anju Byju
- Department of Botany, Bharathiar University, Coimbatore 641046, TN, India; (A.J.); (A.B.); (A.R.G.); (S.S.); (K.T.)
| | - Anju Rani George
- Department of Botany, Bharathiar University, Coimbatore 641046, TN, India; (A.J.); (A.B.); (A.R.G.); (S.S.); (K.T.)
| | - Sradha Sajeev
- Department of Botany, Bharathiar University, Coimbatore 641046, TN, India; (A.J.); (A.B.); (A.R.G.); (S.S.); (K.T.)
| | - Kavimani Thangasamy
- Department of Botany, Bharathiar University, Coimbatore 641046, TN, India; (A.J.); (A.B.); (A.R.G.); (S.S.); (K.T.)
| | - Geetha Natesan
- Department of Botany, Bharathiar University, Coimbatore 641046, TN, India; (A.J.); (A.B.); (A.R.G.); (S.S.); (K.T.)
| |
Collapse
|
9
|
Zhang X, Cheng T, Cho E, Lu W, Denoyer D, McMillan P, Shobhana K, Varshney S, Williamson NA, Stewart A. Nutritionally physiological cell culture medium and 3D culture influence breast tumour proteomics and anti-cancer drug effectiveness. Pharmacol Res 2024; 210:107519. [PMID: 39603575 DOI: 10.1016/j.phrs.2024.107519] [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: 08/27/2024] [Revised: 11/21/2024] [Accepted: 11/21/2024] [Indexed: 11/29/2024]
Abstract
Many drugs have been discontinued during phase II/III breast cancer clinical trials due to lack of clinical efficacy, indicating shortcomings in predictive value of preclinical data. Nutrient availability in the tumour cell microenvironment and the dimensionality of in vitro tumour cells likely impact on drug responsiveness. Global proteomics experiments were conducted to assess the impact of nutrient availability and dimensionality of culture. Protein set enrichment analyses identified "pathways in cancer", "focal adhesion" and "ECM receptor in interaction" related to cell culture dimensionality in MDA-MB-231 cells. In MCF-7 cells, 4 pathways were influenced by medium composition, and 2 pathways were influenced by cell culture dimensionality (2D vs. 3D). These pathways were also identified using KEGG analyses. Eight drugs were selected for investigation according to the differential expression of their putative or known target proteins. The influence of medium composition on drug effectiveness was explored using the "Melbourne Medium" (MM), developed to have nutritionally physiological levels of metabolites as compared with conventional (hyper-nutritional) cell culture medium (CM). The influence of dimensionality on drug effectiveness was also explored, using an innovative 3D viability assessment combining automated confocal microscopy and image analysis. Dimensionality of culture appeared to have a greater influence on the proteome and drug effects than variation in nutrient levels. The number of differentially expressed proteins in the different media was greater in 2D than 3D. We conclude that the risk of qualifying inactive compounds in preclinical assessment may be mitigated using additional models incorporating physiological media and 3-dimensionality.
Collapse
Affiliation(s)
- Xiaodan Zhang
- Department of Biochemistry and Pharmacology, The University of Melbourne, VIC, Australia; ARC Centre for Personalised Therapeutics Technologies, Melbourne, VIC, Australia
| | - Tianhong Cheng
- Department of Biochemistry and Pharmacology, The University of Melbourne, VIC, Australia; ARC Centre for Personalised Therapeutics Technologies, Melbourne, VIC, Australia
| | - Ellie Cho
- Department of Biochemistry and Pharmacology, The University of Melbourne, VIC, Australia; The Biological Optical Microscopy Platform (BOMP), The University of Melbourne, VIC, Australia
| | - Wenjia Lu
- Department of Biochemistry and Pharmacology, The University of Melbourne, VIC, Australia; ARC Centre for Personalised Therapeutics Technologies, Melbourne, VIC, Australia
| | - Delphine Denoyer
- Department of Biochemistry and Pharmacology, The University of Melbourne, VIC, Australia; ARC Centre for Personalised Therapeutics Technologies, Melbourne, VIC, Australia
| | - Paul McMillan
- Department of Biochemistry and Pharmacology, The University of Melbourne, VIC, Australia; The Biological Optical Microscopy Platform (BOMP), The University of Melbourne, VIC, Australia
| | - Kalyan Shobhana
- Department of Biochemistry and Pharmacology, The University of Melbourne, VIC, Australia; The Biological Optical Microscopy Platform (BOMP), The University of Melbourne, VIC, Australia
| | - Swati Varshney
- Melbourne Mass Spectrometry and Proteomics Facility, Bio21 Molecular Science & Biotechnology Institute, The University of Melbourne, VIC, Australia
| | - Nicholas A Williamson
- Melbourne Mass Spectrometry and Proteomics Facility, Bio21 Molecular Science & Biotechnology Institute, The University of Melbourne, VIC, Australia
| | - Alastair Stewart
- Department of Biochemistry and Pharmacology, The University of Melbourne, VIC, Australia; ARC Centre for Personalised Therapeutics Technologies, Melbourne, VIC, Australia.
| |
Collapse
|
10
|
Zhu D, Zhang X, Fang Y, Xu Z, Yu Y, Zhang L, Yang Y, Li S, Wang Y, Jiang C, Huang D. Identification of a lactylation-related gene signature as the novel biomarkers for early diagnosis of acute myocardial infarction. Int J Biol Macromol 2024; 282:137431. [PMID: 39521235 DOI: 10.1016/j.ijbiomac.2024.137431] [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: 09/29/2024] [Revised: 11/03/2024] [Accepted: 11/07/2024] [Indexed: 11/16/2024]
Abstract
Cardiovascular disease, particularly acute myocardial infarction (AMI), is a major global health concern. Current diagnostic methods for AMI lack sensitivity and specificity, necessitating novel biomarkers for early detection. In this study, we analyzed AMI gene expression datasets from the GEO database, employing Differential Gene Expression Analysis and WGCNA to identify key genes and co-expression modules. Lactylation-related genes (LRGs) from the MSigDB database were examined to identify those linked to AMI. Unsupervised consensus clustering classified AMI into subtypes, and machine learning models were developed for diagnosis. Immune cell infiltration was assessed using CIBERSORT, xCell, and MCPcounter, focusing on monocyte activation-related LRGs. We identified four LRGs (AMPD2, PYGL, SLC7A7, SAT1) significantly expressed in AMI, validated through in vitro experiments with primary cardiomyocytes from Sprague-Dawley rats. Our findings highlight LRGs as potential early AMI biomarkers and provide insights into myocardial repair mechanisms mediated by histone lactylation and monocytes.
Collapse
Affiliation(s)
- Dongfei Zhu
- The Department of Cardiology, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Xue Zhang
- The Department of Cardiology, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Yuan Fang
- The Department of Cardiology, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Ziyang Xu
- The Department of Surgery, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Yin Yu
- The Department of Cardiology, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Lili Zhang
- The Department of Cardiology, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - YanPing Yang
- The Department of Cardiology, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China; The Department of Emergency, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Shuai Li
- The Department of Cardiology, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Yanpeng Wang
- The Department of Cardiology, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Can Jiang
- The Department of Anesthesiology, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China.
| | - Dong Huang
- The Department of Cardiology, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China.
| |
Collapse
|
11
|
Alexopoulos S, McGawley M, Mathews R, Papakostopoulou S, Koulas S, Leonidas DD, Zwain T, Hayes JM, Skamnaki V. Evidence for the Quercetin Binding Site of Glycogen Phosphorylase as a Target for Liver-Isoform-Selective Inhibitors against Glioblastoma: Investigation of Flavanols Epigallocatechin Gallate and Epigallocatechin. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2024; 72:24070-24081. [PMID: 39433280 PMCID: PMC11528470 DOI: 10.1021/acs.jafc.4c06920] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/01/2024] [Revised: 10/07/2024] [Accepted: 10/08/2024] [Indexed: 10/23/2024]
Abstract
Glycogen phosphorylase (GP) is the rate-determining enzyme in glycogenolysis, and its druggability has been extensively studied over the years for the development of therapeutics against type 2 diabetes (T2D) and, more recently, cancer. However, the conservation of binding sites between the liver and muscle isoforms makes the inhibitor selectivity challenging. Using a combination of kinetic, crystallographic, modeling, and cellular studies, we have probed the binding of dietary flavonoids epigallocatechin gallate (EGCG) and epigallocatechin (EGC) to GP isoforms. The structures of rmGPb-EGCG and rmGPb-EGC complexes were determined by X-ray crystallography, showing binding at the quercetin binding site (QBS) in agreement with kinetic studies that revealed both compounds as noncompetitive inhibitors of GP, with EGCG also causing a significant reduction in cell viability and migration of U87-MG glioblastoma cells. Interestingly, EGCG exhibits different binding modes to GP isoforms, revealing QBS as a promising site for GP targeting, offering new opportunities for the design of liver-selective GP inhibitors.
Collapse
Affiliation(s)
- Serafeim Alexopoulos
- Department
of Biochemistry and Biotechnology, University
of Thessaly, Biopolis, Larisa 41500, Greece
| | - Megan McGawley
- School
of Pharmacy & Biomedical Sciences, University
of Central Lancashire, Preston PR1 2HE, U.K.
| | - Roshini Mathews
- School
of Pharmacy & Biomedical Sciences, University
of Central Lancashire, Preston PR1 2HE, U.K.
| | - Souzana Papakostopoulou
- Department
of Biochemistry and Biotechnology, University
of Thessaly, Biopolis, Larisa 41500, Greece
| | - Symeon Koulas
- Department
of Biochemistry and Biotechnology, University
of Thessaly, Biopolis, Larisa 41500, Greece
| | - Demetres D. Leonidas
- Department
of Biochemistry and Biotechnology, University
of Thessaly, Biopolis, Larisa 41500, Greece
| | - Tamara Zwain
- School
of Pharmacy & Biomedical Sciences, University
of Central Lancashire, Preston PR1 2HE, U.K.
| | - Joseph M. Hayes
- School
of Pharmacy & Biomedical Sciences, University
of Central Lancashire, Preston PR1 2HE, U.K.
| | - Vasiliki Skamnaki
- Department
of Biochemistry and Biotechnology, University
of Thessaly, Biopolis, Larisa 41500, Greece
| |
Collapse
|
12
|
Hicks E, Layosa MA, Andolino C, Truffer C, Song Y, Heden TD, Donkin SS, Teegarden D. Gluconeogenesis and glycogenolysis required in metastatic breast cancer cells. Front Oncol 2024; 14:1476459. [PMID: 39479019 PMCID: PMC11521782 DOI: 10.3389/fonc.2024.1476459] [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: 08/05/2024] [Accepted: 09/23/2024] [Indexed: 11/02/2024] Open
Abstract
Introduction Metabolic adaptability, including glucose metabolism, enables cells to survive multiple stressful environments. Glycogen may serve as a critical storage depot to provide a source of glucose during times of metabolic demand during the metastatic cascade; therefore, understanding glycogen metabolism is critical. Our goal was to determine mechanisms driving glycogen accumulation and its role in metastatic (MCF10CA1a) compared to nonmetastatic (MCF10A-ras) human breast cancer cells. Methodology 13C6-glucose flux analysis in combination with inhibitors of the gluconeogenic pathway via phosphoenolpyruvate carboxykinase (PCK), the anaplerotic enzyme pyruvate carboxylase (PC), and the rate-limiting enzyme of the pentose phosphate pathway (PPP) glucose 6-phosphate dehydrogenase (G6PD). To determine the requirement of glycogenolysis for migration or survival in extracellular matrix (ECM) detached conditions, siRNA inhibition of glycogenolysis (liver glycogen phosphorylase, PYGL) or glycophagy (lysosomal enzyme α-acid glucosidase, GAA) enzymes was utilized. Results Metastatic MCF10CA1a cells had 20-fold greater glycogen levels compared to non-metastatic MCF10A-ras cells. Most glucose incorporated into glycogen of the MCF10CA1a cells was in the five 13C-containing glucose (M+5) instead of the expected M+6 glycogen-derived glucose moiety, which occurs through direct glucose conversion to glycogen. Furthermore, 13C6-glucose in glycogen was quickly reduced (~50%) following removal of 13C-glucose. Incorporation of 13C6-glucose into the M+5 glucose in the glycogen stores was reduced by inhibition of PCK, with additional contributions from flux through the PPP. Further, inhibition of PC reduced total glycogen content. However, PCK inhibition increased total unlabeled glucose accumulation into glycogen, suggesting an alternative pathway to glycogen accumulation. Inhibition of the rate-limiting steps in glycogenolysis (PYGL) or glycophagy (GAA) demonstrated that both enzymes are necessary to support MCF10CA1a, but not MCF10A-ras, cell migration. GAA inhibition, but not PYGL, reduced viability of MCF10CA1a cells, but not MCF10A-ras, in ECM detached conditions. Conclusion Our results indicate that increased glycogen accumulation is primarily mediated through the gluconeogenesis pathway and that glycogen utilization is required for both migration and ECM detached survival of metastatic MCF10CA1a cells. These results suggest that glycogen metabolism may play an important role in the progression of breast cancer metastasis.
Collapse
Affiliation(s)
- Emily Hicks
- Department of Nutrition Science, Purdue University, West Lafayette, IN, United States
| | - Marjorie Anne Layosa
- Department of Nutrition Science, Purdue University, West Lafayette, IN, United States
- Purdue Institute for Cancer Research, Purdue University, West Lafayette, IN, United States
| | - Chaylen Andolino
- Department of Nutrition Science, Purdue University, West Lafayette, IN, United States
- Purdue Institute for Cancer Research, Purdue University, West Lafayette, IN, United States
| | - Caitlin Truffer
- Department of Nutrition Science, Purdue University, West Lafayette, IN, United States
| | - Yazhen Song
- Department of Nutrition Science, Purdue University, West Lafayette, IN, United States
- Purdue Institute for Cancer Research, Purdue University, West Lafayette, IN, United States
| | - Timothy D. Heden
- Pennington Biomedical Research Center, Baton Rouge, LA, United States
| | - Shawn S. Donkin
- Department of Animal Sciences, Purdue University, West Lafayette, IN, United States
- College of Agricultural Sciences, Oregon State University, Corvallis, OR, United States
| | - Dorothy Teegarden
- Department of Nutrition Science, Purdue University, West Lafayette, IN, United States
- Purdue Institute for Cancer Research, Purdue University, West Lafayette, IN, United States
| |
Collapse
|
13
|
Di Marco T, Mazzoni M, Greco A, Cassinelli G. Non-oncogene dependencies: Novel opportunities for cancer therapy. Biochem Pharmacol 2024; 228:116254. [PMID: 38704100 DOI: 10.1016/j.bcp.2024.116254] [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: 02/07/2024] [Revised: 04/22/2024] [Accepted: 04/30/2024] [Indexed: 05/06/2024]
Abstract
Targeting oncogene addictions have changed the history of subsets of malignancies and continues to represent an excellent therapeutic opportunity. Nonetheless, alternative strategies are required to treat malignancies driven by undruggable oncogenes or loss of tumor suppressor genes and to overcome drug resistance also occurring in cancers addicted to actionable drivers. The discovery of non-oncogene addiction (NOA) uncovered novel therapeutically exploitable "Achilles' heels". NOA refers to genes/pathways not oncogenic per sé but essential for the tumor cell growth/survival while dispensable for normal cells. The clinical success of several classes of conventional and molecular targeted agents can be ascribed to their impact on both tumor cell-associated intrinsic as well as microenvironment-related extrinsic NOA. The integration of genetic, computational and pharmacological high-throughput approaches led to the identification of an expanded repertoire of synthetic lethality interactions implicating NOA targets. Only a few of them have been translated into the clinics as most NOA vulnerabilities are not easily druggable or appealing targets. Nonetheless, their identification has provided in-depth knowledge of tumor pathobiology and suggested novel therapeutic opportunities. Here, we summarize conceptual framework of intrinsic and extrinsic NOA providing exploitable vulnerabilities. Conventional and emerging methodological approaches used to disclose NOA dependencies are reported together with their limits. We illustrate NOA paradigmatic and peculiar examples and outline the functional/mechanistic aspects, potential druggability and translational interest. Finally, we comment on difficulties in exploiting the NOA-generated knowledge to develop novel therapeutic approaches to be translated into the clinics and to fully harness the potential of clinically available drugs.
Collapse
Affiliation(s)
- Tiziana Di Marco
- Integrated Biology of Rare Tumors Unit, Experimental Oncology Department, Fondazione IRCCS Istituto Nazionale dei Tumori, Via Amadeo 42, 20133 Milan, Italy
| | - Mara Mazzoni
- Integrated Biology of Rare Tumors Unit, Experimental Oncology Department, Fondazione IRCCS Istituto Nazionale dei Tumori, Via Amadeo 42, 20133 Milan, Italy
| | - Angela Greco
- Integrated Biology of Rare Tumors Unit, Experimental Oncology Department, Fondazione IRCCS Istituto Nazionale dei Tumori, Via Amadeo 42, 20133 Milan, Italy
| | - Giuliana Cassinelli
- Molecular Pharmacology Unit, Experimental Oncology Department, Fondazione IRCCS Istituto Nazionale dei Tumori, Via Amadeo 42, 20133 Milan, Italy.
| |
Collapse
|
14
|
Zhao N, Shen Y, Shi D, Mao Y, Wang G, Xiao G, Xu D, Yan G. Advances in the Clinical Study of Nuclear Overhauser Enhancement. J Magn Reson Imaging 2024. [PMID: 39340226 DOI: 10.1002/jmri.29623] [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: 06/14/2024] [Revised: 08/06/2024] [Accepted: 08/06/2024] [Indexed: 09/30/2024] Open
Abstract
Nuclear overhauser enhancement is a confounding factor arising from the in vivo application of a chemical exchange saturation transfer technique in which two nuclei in close proximity undergo dipole cross-relaxation. Several studies have shown applicability and efficacy of nuclear overhauser enhancement in observing tumors and other lesions in vivo. Thus, this effect could become an emerging molecular imaging research tool for many diseases. Moreover, nuclear overhauser enhancement has the advantages of simplicity, noninvasiveness, and high resolution and has become a major focus of current research. In this review, we summarize the principles and applications of nuclear overhauser enhancement. LEVEL OF EVIDENCE: 2 TECHNICAL EFFICACY: Stage 1.
Collapse
Affiliation(s)
- Nannan Zhao
- Department of Center of Morphological Experiment, Medical college of Yanbian University, Yanji, China
- Department of Radiology, Xiamen Medical College Affiliated Second Hospital, Xiamen, China
| | - Yuanyu Shen
- Department of Radiology, Second Affiliated Hospital of Shantou University Medical College, Shantou, China
| | - Dafa Shi
- Department of Radiology, Second Affiliated Hospital of Shantou University Medical College, Shantou, China
| | - Yumeng Mao
- Department of Radiology, Second Affiliated Hospital of Shantou University Medical College, Shantou, China
| | - Guangsong Wang
- Department of Radiology, Xiang'an Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, China
| | - Gang Xiao
- School of Mathematics and Statistics, Hanshan Normal University, Chaozhou, China
| | - Dongyuan Xu
- Department of Center of Morphological Experiment, Medical college of Yanbian University, Yanji, China
| | - Gen Yan
- Department of Radiology, Xiamen Medical College Affiliated Second Hospital, Xiamen, China
| |
Collapse
|
15
|
Taneja N, Chauhan A, Kulshreshtha R, Singh S. HIF-1 mediated metabolic reprogramming in cancer: Mechanisms and therapeutic implications. Life Sci 2024; 352:122890. [PMID: 38971364 DOI: 10.1016/j.lfs.2024.122890] [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: 05/06/2024] [Revised: 06/24/2024] [Accepted: 07/03/2024] [Indexed: 07/08/2024]
Abstract
Cancer cells undergo metabolic reprogramming to survive in hypoxic conditions and meet the elevated energy demands of the cancer microenvironment. This metabolic alteration is orchestrated by hypoxia-inducible factor 1 (HIF-1), regulating various processes within cancer cells. The intricate metabolic modifications induced by hypoxia underscore the significance of HIF-1-induced metabolic reprogramming in promoting each aspect of cancer progression. The complex interactions between HIF-1 signalling and cellular metabolic processes in response to hypoxia are examined in this study, focusing on the metabolism of carbohydrates, nucleotides, lipids, and amino acids. Comprehending the various regulatory mechanisms controlled by HIF-1 in cellular metabolism sheds light on the intricate biology of cancer growth and offers useful insights for developing targeted treatments.
Collapse
Affiliation(s)
- Nikita Taneja
- Amity Institute of Health Allied Sciences, Amity University, Noida, Uttar Pradesh, India
| | - Akansha Chauhan
- Amity Institute of Health Allied Sciences, Amity University, Noida, Uttar Pradesh, India
| | - Ritu Kulshreshtha
- Department of Biochemical Engineering and Biotechnology, Indian Institute of Technology, New Delhi, India
| | - Sandhya Singh
- Amity Institute of Health Allied Sciences, Amity University, Noida, Uttar Pradesh, India.
| |
Collapse
|
16
|
Zhang F, Guo J, Yu S, Zheng Y, Duan M, Zhao L, Wang Y, Yang Z, Jiang X. Cellular senescence and metabolic reprogramming: Unraveling the intricate crosstalk in the immunosuppressive tumor microenvironment. Cancer Commun (Lond) 2024; 44:929-966. [PMID: 38997794 PMCID: PMC11492308 DOI: 10.1002/cac2.12591] [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: 11/26/2023] [Revised: 06/23/2024] [Accepted: 07/07/2024] [Indexed: 07/14/2024] Open
Abstract
The intrinsic oncogenic mechanisms and properties of the tumor microenvironment (TME) have been extensively investigated. Primary features of the TME include metabolic reprogramming, hypoxia, chronic inflammation, and tumor immunosuppression. Previous studies suggest that senescence-associated secretory phenotypes that mediate intercellular information exchange play a role in the dynamic evolution of the TME. Specifically, hypoxic adaptation, metabolic dysregulation, and phenotypic shifts in immune cells regulated by cellular senescence synergistically contribute to the development of an immunosuppressive microenvironment and chronic inflammation, thereby promoting the progression of tumor events. This review provides a comprehensive summary of the processes by which cellular senescence regulates the dynamic evolution of the tumor-adapted TME, with focus on the complex mechanisms underlying the relationship between senescence and changes in the biological functions of tumor cells. The available findings suggest that components of the TME collectively contribute to the progression of tumor events. The potential applications and challenges of targeted cellular senescence-based and combination therapies in clinical settings are further discussed within the context of advancing cellular senescence-related research.
Collapse
Affiliation(s)
- Fusheng Zhang
- Department of General SurgeryThe Fourth Affiliated Hospital of China Medical UniversityShenyangLiaoningP. R. China
- Department of Hepatobiliary and Pancreatic SurgeryPeking University First HospitalBeijingP. R. China
| | - Junchen Guo
- Department of RadiologyThe Fourth Affiliated Hospital of China Medical UniversityShenyangLiaoningP. R. China
| | - Shengmiao Yu
- Outpatient DepartmentThe Fourth Affiliated HospitalChina Medical UniversityShenyangLiaoningP. R. China
| | - Youwei Zheng
- Department of General SurgeryThe Fourth Affiliated Hospital of China Medical UniversityShenyangLiaoningP. R. China
| | - Meiqi Duan
- Department of General SurgeryThe Fourth Affiliated Hospital of China Medical UniversityShenyangLiaoningP. R. China
| | - Liang Zhao
- Department of General SurgeryThe Fourth Affiliated Hospital of China Medical UniversityShenyangLiaoningP. R. China
| | - Yihan Wang
- Department of General SurgeryThe Fourth Affiliated Hospital of China Medical UniversityShenyangLiaoningP. R. China
| | - Zhi Yang
- Department of General SurgeryThe Fourth Affiliated Hospital of China Medical UniversityShenyangLiaoningP. R. China
| | - Xiaofeng Jiang
- Department of General SurgeryThe Fourth Affiliated Hospital of China Medical UniversityShenyangLiaoningP. R. China
| |
Collapse
|
17
|
Basu B, Karwatka M, China B, McKibbin M, Khan K, Inglehearn CF, Ladbury JE, Johnson CA. Glycogen myophosphorylase loss causes increased dependence on glucose in iPSC-derived retinal pigment epithelium. J Biol Chem 2024; 300:107569. [PMID: 39009342 PMCID: PMC11342771 DOI: 10.1016/j.jbc.2024.107569] [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: 01/20/2024] [Revised: 06/07/2024] [Accepted: 06/27/2024] [Indexed: 07/17/2024] Open
Abstract
Loss of glycogen myophosphorylase (PYGM) expression results in an inability to break down muscle glycogen, leading to McArdle disease-an autosomal recessive metabolic disorder characterized by exercise intolerance and muscle cramps. While previously considered relatively benign, this condition has recently been associated with pattern dystrophy in the retina, accompanied by variable sight impairment, secondary to retinal pigment epithelial (RPE) cell involvement. However, the pathomechanism of this condition remains unclear. In this study, we generated a PYGM-null induced pluripotent stem cell line and differentiated it into mature RPE to examine structural and functional defects, along with metabolite release into apical and basal media. Mutant RPE exhibited normal photoreceptor outer segment phagocytosis but displayed elevated glycogen levels, reduced transepithelial resistance, and increased cytokine secretion across the epithelial layer compared to isogenic WT controls. Additionally, decreased expression of the visual cycle component, RDH11, encoding 11-cis-retinol dehydrogenase, was observed in PYGM-null RPE. While glycolytic flux and oxidative phosphorylation levels in PYGM-null RPE were near normal, the basal oxygen consumption rate was increased. Oxygen consumption rate in response to physiological levels of lactate was significantly greater in WT than PYGM-null RPE. Inefficient lactate utilization by mutant RPE resulted in higher glucose dependence and increased glucose uptake from the apical medium in the presence of lactate, suggesting a reduced capacity to spare glucose for photoreceptor use. Metabolic tracing confirmed slower 13C-lactate utilization by PYGM-null RPE. These findings have key implications for retinal health since they likely underlie the vision impairment in individuals with McArdle disease.
Collapse
Affiliation(s)
- Basudha Basu
- Division of Molecular Medicine, Leeds Institute of Medical Research, University of Leeds, Leeds, UK
| | - Magdalena Karwatka
- Division of Molecular Medicine, Leeds Institute of Medical Research, University of Leeds, Leeds, UK
| | - Becky China
- Division of Molecular Medicine, Leeds Institute of Medical Research, University of Leeds, Leeds, UK
| | - Martin McKibbin
- Division of Molecular Medicine, Leeds Institute of Medical Research, University of Leeds, Leeds, UK; Department of Ophthalmology, St James's University Hospital, Leeds, UK
| | - Kamron Khan
- Division of Molecular Medicine, Leeds Institute of Medical Research, University of Leeds, Leeds, UK
| | - Chris F Inglehearn
- Division of Molecular Medicine, Leeds Institute of Medical Research, University of Leeds, Leeds, UK
| | - John E Ladbury
- School of Molecular and Cellular Biology, University of Leeds, Leeds, UK
| | - Colin A Johnson
- Division of Molecular Medicine, Leeds Institute of Medical Research, University of Leeds, Leeds, UK.
| |
Collapse
|
18
|
Mestareehi A. Global Gene Expression Profiling and Bioinformatics Analysis Reveal Downregulated Biomarkers as Potential Indicators for Hepatocellular Carcinoma. ACS OMEGA 2024; 9:26075-26096. [PMID: 38911766 PMCID: PMC11191119 DOI: 10.1021/acsomega.4c01496] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/15/2024] [Revised: 05/22/2024] [Accepted: 05/24/2024] [Indexed: 06/25/2024]
Abstract
Objective: The study aimed to elucidate the significance of CLEC4G, CAMK2β, SLC22A1, CBFA2T3, and STAB2 in the prognosis of hepatocellular carcinoma (HCC) patients and their associated molecular biological characteristics. Additionally, the research sought to identify new potential biomarkers with therapeutic and diagnostic relevance for clinical applications. Methods and Materials: We utilized a publicly available high throughput phosphoproteomics and proteomics data set of HCC to focus on the analysis of 12 downregulated phosphoproteins in HCC. Our approach integrates bioinformatic analysis with pathway analysis, encompassing gene ontology (GO) analysis, Kyoto Encyclopedia of Genes and Genomes (KEGG) pathway analysis, and the construction of a protein-protein interaction (PPI) network. Results: In total, we quantified 11547 phosphorylation sites associated with 4043 phosphoproteins from a cohort of 159 HCC patients. Within this extensive data set, our specific focus was on 19 phosphorylation sites displaying significant downregulation (log2 FC ≤ -2 with p-values < 0.0001). Remarkably, our investigation revealed distinct pathways exhibiting differential regulation across multiple dimensions, including the genomic, transcriptomic, proteomic, and phosphoproteomic levels. These pathways encompass a wide range of critical cellular processes, including cellular component organization, cell cycle control, signaling pathways, transcriptional and translational control, and metabolism. Furthermore, our bioinformatics analysis unveiled noteworthy insights into the subcellular localizations, biological processes, and molecular functions associated with these proteins and phosphoproteins. Within the context of the PPI network, we identified 12 key genes CLEC4G, STAB2, ADH1A, ADH1B, CAMK2B, ADH4, CHGB, PYGL, ADH1C, AKAP12, CBFA2T3, and SLC22A1 as the top highly interconnected hub genes. Conclusions: The findings related to CLEC4G, ADH1B, SLC22A1, CAMK2β, CBFA2T3, and STAB2 indicate their reduced expression in HCC, which is associated with an unfavorable prognosis. Furthermore, the results of KEGG and GO pathway analyses suggest that these genes may impact liver cancer by engaging various targets and pathways, ultimately promoting the progression of hepatocellular carcinoma. These results underscore the significant potential of CLEC4G, ADH1B, SLC22A1, CAMK2β, CBFA2T3, and STAB2 as key contributors to HCC development and advancement. This insight holds promise for identifying therapeutic targets and charting research avenues to enhance our understanding of the intricate molecular mechanisms underlying hepatocellular carcinoma.
Collapse
Affiliation(s)
- Aktham Mestareehi
- Department
of Pharmaceutical Sciences, Faculty of Pharmacy, Isra University, P.O. Box 22, Amman 11622, Jordan
- School
of Medicine, The Ohio State University, Columbus, Ohio 43202, United States
- Department
of Pharmaceutical Sciences, School of Pharmacy and Health Sciences, Wayne State University, Detroit, Michigan 48201, United States
| |
Collapse
|
19
|
Chen X, Yang N, Wang Y, Yang S, Peng Y. PCK1-mediated glycogenolysis facilitates ROS clearance and chemotherapy resistance in cervical cancer stem cells. Sci Rep 2024; 14:13670. [PMID: 38871968 PMCID: PMC11176388 DOI: 10.1038/s41598-024-64255-6] [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: 01/16/2024] [Accepted: 06/06/2024] [Indexed: 06/15/2024] Open
Abstract
Cervical cancer, one of the most common gynecological cancers, is primarily caused by human papillomavirus (HPV) infection. The development of resistance to chemotherapy is a significant hurdle in treatment. In this study, we investigated the mechanisms underlying chemoresistance in cervical cancer by focusing on the roles of glycogen metabolism and the pentose phosphate pathway (PPP). We employed the cervical cancer cell lines HCC94 and CaSki by manipulating the expression of key enzymes PCK1, PYGL, and GYS1, which are involved in glycogen metabolism, through siRNA transfection. Our analysis included measuring glycogen levels, intermediates of PPP, NADPH/NADP+ ratio, and the ability of cells to clear reactive oxygen species (ROS) using biochemical assays and liquid chromatography-mass spectrometry (LC-MS). Furthermore, we assessed chemoresistance by evaluating cell viability and tumor growth in NSG mice. Our findings revealed that in drug-resistant tumor stem cells, the enzyme PCK1 enhances the phosphorylation of PYGL, leading to increased glycogen breakdown. This process shifts glucose metabolism towards PPP, generating NADPH. This, in turn, facilitates ROS clearance, promotes cell survival, and contributes to the development of chemoresistance. These insights suggest that targeting aberrant glycogen metabolism or PPP could be a promising strategy for overcoming chemoresistance in cervical cancer. Understanding these molecular mechanisms opens new avenues for the development of more effective treatments for this challenging malignancy.
Collapse
Affiliation(s)
- Xinxin Chen
- Department of Cadre Ward 2, the Second Affiliated Hospital of Harbin Medical University, Harbin, 150086, China
| | - Nan Yang
- Department of Cadre Ward 2, the Second Affiliated Hospital of Harbin Medical University, Harbin, 150086, China
| | - Ying Wang
- Department of Cadre Ward 2, the Second Affiliated Hospital of Harbin Medical University, Harbin, 150086, China
| | - Shuang Yang
- Department of Cadre Ward 2, the Second Affiliated Hospital of Harbin Medical University, Harbin, 150086, China
| | - Yuanhong Peng
- Department of Cadre Ward 1, the Second Affiliated Hospital of Harbin Medical University, Harbin, 150086, China.
| |
Collapse
|
20
|
Liu Y, Wu Z, Li Y, Chen Y, Zhao X, Wu M, Xia Y. Metabolic reprogramming and interventions in angiogenesis. J Adv Res 2024:S2090-1232(24)00178-4. [PMID: 38704087 DOI: 10.1016/j.jare.2024.05.001] [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: 03/15/2024] [Revised: 04/30/2024] [Accepted: 05/01/2024] [Indexed: 05/06/2024] Open
Abstract
BACKGROUND Endothelial cell (EC) metabolism plays a crucial role in the process of angiogenesis. Intrinsic metabolic events such as glycolysis, fatty acid oxidation, and glutamine metabolism, support secure vascular migration and proliferation, energy and biomass production, as well as redox homeostasis maintenance during vessel formation. Nevertheless, perturbation of EC metabolism instigates vascular dysregulation-associated diseases, especially cancer. AIM OF REVIEW In this review, we aim to discuss the metabolic regulation of angiogenesis by EC metabolites and metabolic enzymes, as well as prospect the possible therapeutic opportunities and strategies targeting EC metabolism. KEY SCIENTIFIC CONCEPTS OF REVIEW In this work, we discuss various aspects of EC metabolism considering normal and diseased vasculature. Of relevance, we highlight that the implications of EC metabolism-targeted intervention (chiefly by metabolic enzymes or metabolites) could be harnessed in orchestrating a spectrum of pathological angiogenesis-associated diseases.
Collapse
Affiliation(s)
- Yun Liu
- College of Animal Science and Technology, Southwest University, Chongqing 400715, China
| | - Zifang Wu
- College of Animal Science and Technology, Northwest A&F University, Yangling, Shaanxi 712100, China
| | - Yikun Li
- College of Animal Science and Technology, Southwest University, Chongqing 400715, China; College of Animal Science, South China Agricultural University, Guangzhou, Guangdong 510642, China
| | - Yating Chen
- College of Animal Science and Technology, Southwest University, Chongqing 400715, China
| | - Xuan Zhao
- College of Animal Science and Technology, Southwest University, Chongqing 400715, China.
| | - Miaomiao Wu
- Animal Nutritional Genome and Germplasm Innovation Research Center, College of Animal Science and Technology, Hunan Agricultural University, Changsha, Hunan 410128, China.
| | - Yaoyao Xia
- College of Animal Science and Technology, Southwest University, Chongqing 400715, China.
| |
Collapse
|
21
|
Xu G, Pan H, Fan L, Zhang L, Li J, Cheng S, Meng L, Shen N, Liu Y, Li Y, Huang T, Zhou L. Dietary Betaine Improves Glucose Metabolism in Obese Mice. J Nutr 2024; 154:1309-1320. [PMID: 38417550 DOI: 10.1016/j.tjnut.2024.02.025] [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: 11/26/2023] [Revised: 02/17/2024] [Accepted: 02/23/2024] [Indexed: 03/01/2024] Open
Abstract
BACKGROUND Obesity caused by the overconsumption of energy-dense foods high in fat and sugar has contributed to the growing prevalence of type 2 diabetes. Betaine, found in food or supplements, has been found to lower blood glucose concentrations, but its exact mechanism of action is not well understood. OBJECTIVES A comprehensive evaluation of the potential mechanisms by which betaine supplementation improves glucose metabolism. METHODS Hyperglycemic mice were fed betaine to measure the indexes of glucose metabolism in the liver and muscle. To explore the mechanism behind the regulation of betaine on glucose metabolism, Ribonucleic Acid-Seq was used to analyze the livers of the mice. In vitro, HepG2 and C2C12 cells were treated with betaine to more comprehensively evaluate the effect of betaine on glucose metabolism. RESULTS Betaine was added to the drinking water of high-fat diet-induced mice, and it was found to reduce blood glucose concentrations and liver triglyceride concentrations without affecting body weight, confirming its hypoglycemic effect. To investigate the specific mechanism underlying its hypoglycemic effect, protein-protein interaction enrichment analysis of the liver revealed key nodes associated with glucose metabolism, including cytochrome P450 family activity, insulin sensitivity, glucose homeostasis, and triglyceride concentrations. The Kyoto Encyclopedia of Genes and Genomes and gene ontogeny enrichment analyses showed significant enrichment of the Notch signaling pathway. These results provided bioinformatic evidence for specific pathways through which betaine regulates glucose metabolism. Key enzyme activities involved in glucose uptake, glycogen synthesis, and glycogenolysis pathways of the liver and muscle were measured, and improvements were observed in these pathways. CONCLUSIONS This study provides new insight into the mechanisms by which betaine improves glucose metabolism in the liver and muscle and supports its potential as a drug for the treatment of metabolic disorders related to glucose.
Collapse
Affiliation(s)
- Gaoxiao Xu
- Anhui Province Key Laboratory of Embryo Development and Reproductive Regulation, and Environmental Hormone and Reproduction, School of Biological and Food Engineering, Fuyang Normal University, Fuyang, China
| | - Hongyuan Pan
- College of Animal Science and Technology, Guangxi University, Nanning, China
| | - Liping Fan
- Anhui Province Key Laboratory of Embryo Development and Reproductive Regulation, and Environmental Hormone and Reproduction, School of Biological and Food Engineering, Fuyang Normal University, Fuyang, China
| | - Lifang Zhang
- College of Animal Science and Technology, Guangxi University, Nanning, China
| | - Jian Li
- Anhui Province Key Laboratory of Embryo Development and Reproductive Regulation, and Environmental Hormone and Reproduction, School of Biological and Food Engineering, Fuyang Normal University, Fuyang, China
| | - Shimei Cheng
- Anhui Province Key Laboratory of Embryo Development and Reproductive Regulation, and Environmental Hormone and Reproduction, School of Biological and Food Engineering, Fuyang Normal University, Fuyang, China
| | - Libing Meng
- Anhui Province Key Laboratory of Embryo Development and Reproductive Regulation, and Environmental Hormone and Reproduction, School of Biological and Food Engineering, Fuyang Normal University, Fuyang, China
| | - Nana Shen
- Anhui Province Key Laboratory of Embryo Development and Reproductive Regulation, and Environmental Hormone and Reproduction, School of Biological and Food Engineering, Fuyang Normal University, Fuyang, China
| | - Yong Liu
- Anhui Province Key Laboratory of Embryo Development and Reproductive Regulation, and Environmental Hormone and Reproduction, School of Biological and Food Engineering, Fuyang Normal University, Fuyang, China
| | - Yixing Li
- College of Animal Science and Technology, Guangxi University, Nanning, China
| | - Tengda Huang
- College of Animal Science and Technology, Guangxi University, Nanning, China.
| | - Lei Zhou
- Guangxi Academy of Medical Sciences, the People's Hospital of Guangxi Zhuang Autonomous Region, Nanning, China.
| |
Collapse
|
22
|
Ren LK, Lu RS, Fei XB, Chen SJ, Liu P, Zhu CH, Wang X, Pan YZ. Unveiling the role of PYGB in pancreatic cancer: a novel diagnostic biomarker and gene therapy target. J Cancer Res Clin Oncol 2024; 150:127. [PMID: 38483604 PMCID: PMC10940407 DOI: 10.1007/s00432-024-05644-2] [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: 12/12/2023] [Accepted: 02/05/2024] [Indexed: 03/17/2024]
Abstract
PURPOSE Pancreatic cancer (PC) is a highly malignant tumor that poses a severe threat to human health. Brain glycogen phosphorylase (PYGB) breaks down glycogen and provides an energy source for tumor cells. Although PYGB has been reported in several tumors, its role in PC remains unclear. METHODS We constructed a risk diagnostic model of PC-related genes by WGCNA and LASSO regression and found PYGB, an essential gene in PC. Then, we explored the pro-carcinogenic role of PYGB in PC by in vivo and in vitro experiments. RESULTS We found that PYGB, SCL2A1, and SLC16A3 had a significant effect on the diagnosis and prognosis of PC, but PYGB had the most significant effect on the prognosis. Pan-cancer analysis showed that PYGB was highly expressed in most of the tumors but had the highest correlation with PC. In TCGA and GEO databases, we found that PYGB was highly expressed in PC tissues and correlated with PC's prognostic and pathological features. Through in vivo and in vitro experiments, we found that high expression of PYGB promoted the proliferation, invasion, and metastasis of PC cells. Through enrichment analysis, we found that PYGB is associated with several key cell biological processes and signaling pathways. In experiments, we validated that the MAPK/ERK pathway is involved in the pro-tumorigenic mechanism of PYGB in PC. CONCLUSION Our results suggest that PYGB promotes PC cell proliferation, invasion, and metastasis, leading to poor patient prognosis. PYGB gene may be a novel diagnostic biomarker and gene therapy target for PC.
Collapse
Affiliation(s)
- Li-Kun Ren
- College of Clinical Medicine, Guizhou Medical University, Guiyang, 550000, Guizhou, China
| | - Ri-Shang Lu
- College of Clinical Medicine, Guizhou Medical University, Guiyang, 550000, Guizhou, China
| | - Xiao-Bin Fei
- College of Clinical Medicine, Guizhou Medical University, Guiyang, 550000, Guizhou, China
| | - Shao-Jie Chen
- College of Clinical Medicine, Guizhou Medical University, Guiyang, 550000, Guizhou, China
- Department of Hepatic-Biliary-Pancreatic Surgery, The Affiliated Hospital of Guizhou Medical University, Guiyang, 550000, China
| | - Peng Liu
- College of Clinical Medicine, Guizhou Medical University, Guiyang, 550000, Guizhou, China
- Department of Hepatic-Biliary-Pancreatic Surgery, The Affiliated Hospital of Guizhou Medical University, Guiyang, 550000, China
| | - Chang-Hao Zhu
- College of Clinical Medicine, Guizhou Medical University, Guiyang, 550000, Guizhou, China
- Department of Hepatic-Biliary-Pancreatic Surgery, The Affiliated Cancer Hospital of Guizhou Medical University, Guiyang, 550000, China
| | - Xing Wang
- College of Clinical Medicine, Guizhou Medical University, Guiyang, 550000, Guizhou, China.
- Department of Hepatic-Biliary-Pancreatic Surgery, The Affiliated Cancer Hospital of Guizhou Medical University, Guiyang, 550000, China.
| | - Yao-Zhen Pan
- College of Clinical Medicine, Guizhou Medical University, Guiyang, 550000, Guizhou, China.
- Department of Hepatic-Biliary-Pancreatic Surgery, The Affiliated Cancer Hospital of Guizhou Medical University, Guiyang, 550000, China.
| |
Collapse
|
23
|
Zhao X, Wang C, Zhao L, Tian Z. HBV DNA polymerase regulates tumor cell glycogen to enhance the malignancy of HCC cells. Hepatol Commun 2024; 8:e0387. [PMID: 38358372 PMCID: PMC10871796 DOI: 10.1097/hc9.0000000000000387] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/06/2023] [Accepted: 12/16/2023] [Indexed: 02/16/2024] Open
Abstract
BACKGROUND The essential function of HBV DNA polymerase (HBV-DNA-Pol) is to initiate viral replication by reverse transcription; however, the role of HBV-DNA-Pol in HBV-associated HCC has not been clarified. Glycogen phosphorylase L (PYGL) is a critical regulator of glycogenolysis and is involved in tumorigenesis, including HCC. However, it is unknown whether HBV-DNA-Pol regulates PYGL to contribute to HCC tumorigenesis. METHODS Bioinformatic analysis, real-time quantitative PCR, western blotting, and oncology functional assays were performed to determine the contribution of HBV-DNA-Pol and PYGL to HCC development and glycolysis. The mechanisms of co-immunoprecipitation and ubiquitination were employed to ascertain how HBV-DNA-Pol upregulated PYGL. RESULTS Overexpression of HBV-DNA-Pol enhanced HCC progression in vitro and in vivo. Mechanistically, HBV-DNA-Pol interacted with PYGL and increased PYGL protein levels by inhibiting PYGL ubiquitination, which was mediated by the E3 ligase TRIM21. HBV-DNA-Pol competitively impaired the binding of PYGL to TRIM21 due to its stronger binding affinity to TRIM21, suppressing the ubiquitination of PYGL. Moreover, HBV-DNA-Pol promoted glycogen decomposition by upregulating PYGL, which led to an increased flow of glucose into glycolysis, thereby promoting HCC development. CONCLUSIONS Our study reveals a novel mechanism by which HBV-DNA-Pol promotes HCC by controlling glycogen metabolism in HCC, establishing a direct link between HBV-DNA-Pol and the Warburg effect, thereby providing novel targets for HCC treatment and drug development.
Collapse
Affiliation(s)
- Xiaoqing Zhao
- Department of Clinical Laboratory, The Second Hospital, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Chunqing Wang
- Department of Clinical Laboratory Medicine, The First Affiliated Hospital of Shandong First Medical University & Shandong Provincial Qianfoshan Hospital, Shandong Medicine and Health Key Laboratory of Laboratory Medicine, Jinan, China
| | - Liqing Zhao
- Department of Pediatrics, Zaozhuang Municipal Hospital, Zaozhuang, China
| | - Zhongzheng Tian
- Shandong Agricultural Technology Extending Center, Jinan, China
| |
Collapse
|
24
|
Hagiwara K, Watanabe A, Harimoto N, Araki K, Yokobori T, Muranushi R, Hoshino K, Ishii N, Tsukagoshi M, Shirabe K. Liver regeneration after hepatectomy is significantly suppressed in a muscular atrophy mouse model. JOURNAL OF HEPATO-BILIARY-PANCREATIC SCIENCES 2024; 31:152-161. [PMID: 37909250 DOI: 10.1002/jhbp.1386] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/03/2022] [Revised: 08/31/2023] [Accepted: 09/07/2023] [Indexed: 11/02/2023]
Abstract
BACKGROUND Sarcopenia is a syndrome characterized by progressive and generalized loss of skeletal muscle mass and strength. As reported in previous studies, the loss of skeletal muscle mass is associated with poor liver regeneration after hepatectomy. It is considered important to clarify the effect of sarcopenia on liver regeneration; however, there are no reports about model animals for sarcopenia. We focused on the peroxisome proliferator-activated receptor-gamma coactivator-1alpha (PGC-1α) transgenic mice that overexpressed PGC-1α, specifically for skeletal muscle, and showed significant atrophy of type 2B fiber-rich muscles like sarcopenia. METHODS We performed 70% hepatectomy using PGC-1α transgenic mice and examined the liver regeneration rate and the effects of branched-chain amino acids (BCAA) after hepatectomy. RESULTS Liver regeneration after 70% hepatectomy was significantly suppressed in the PGC-1α transgenic mice. In addition, a decrease in the blood BCAA concentration and a decrease in the liver glycogen content after 70% hepatectomy were observed in the PGC-1α transgenic mice. By administering BCAA before and after surgery, it was clarified that a significant increase in the blood BCAA concentration was observed and the liver regeneration rate was improved in the PGC-1α transgenic mice. CONCLUSIONS BCAA administration may improve the suppression of liver regeneration in patients with sarcopenia.
Collapse
Affiliation(s)
- Kei Hagiwara
- Division of Hepatobiliary and Pancreatic Surgery, Department of General Surgical Science, Gunma University Graduate School of Medicine, Maebashi, Gunma, Japan
| | - Akira Watanabe
- Division of Hepatobiliary and Pancreatic Surgery, Department of General Surgical Science, Gunma University Graduate School of Medicine, Maebashi, Gunma, Japan
| | - Norifumi Harimoto
- Division of Hepatobiliary and Pancreatic Surgery, Department of General Surgical Science, Gunma University Graduate School of Medicine, Maebashi, Gunma, Japan
| | - Kenichiro Araki
- Division of Hepatobiliary and Pancreatic Surgery, Department of General Surgical Science, Gunma University Graduate School of Medicine, Maebashi, Gunma, Japan
| | - Takehiko Yokobori
- Division of Integrated Oncology Research, Gunma University Initiative for Advanced Research (GIAR), Maebashi, Gunma, Japan
| | - Ryo Muranushi
- Division of Hepatobiliary and Pancreatic Surgery, Department of General Surgical Science, Gunma University Graduate School of Medicine, Maebashi, Gunma, Japan
| | - Kouki Hoshino
- Division of Hepatobiliary and Pancreatic Surgery, Department of General Surgical Science, Gunma University Graduate School of Medicine, Maebashi, Gunma, Japan
| | - Norihiro Ishii
- Division of Hepatobiliary and Pancreatic Surgery, Department of General Surgical Science, Gunma University Graduate School of Medicine, Maebashi, Gunma, Japan
| | - Mariko Tsukagoshi
- Division of Hepatobiliary and Pancreatic Surgery, Department of General Surgical Science, Gunma University Graduate School of Medicine, Maebashi, Gunma, Japan
| | - Ken Shirabe
- Division of Hepatobiliary and Pancreatic Surgery, Department of General Surgical Science, Gunma University Graduate School of Medicine, Maebashi, Gunma, Japan
| |
Collapse
|
25
|
Xu S, Wang L, Zhao Y, Mo T, Wang B, Lin J, Yang H. Metabolism-regulating non-coding RNAs in breast cancer: roles, mechanisms and clinical applications. J Biomed Sci 2024; 31:25. [PMID: 38408962 PMCID: PMC10895768 DOI: 10.1186/s12929-024-01013-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: 11/17/2023] [Accepted: 01/17/2024] [Indexed: 02/28/2024] Open
Abstract
Breast cancer is one of the most common malignancies that pose a serious threat to women's health. Reprogramming of energy metabolism is a major feature of the malignant transformation of breast cancer. Compared to normal cells, tumor cells reprogram metabolic processes more efficiently, converting nutrient supplies into glucose, amino acid and lipid required for malignant proliferation and progression. Non-coding RNAs(ncRNAs) are a class of functional RNA molecules that are not translated into proteins but regulate the expression of target genes. NcRNAs have been demonstrated to be involved in various aspects of energy metabolism, including glycolysis, glutaminolysis, and fatty acid synthesis. This review focuses on the metabolic regulatory mechanisms and clinical applications of metabolism-regulating ncRNAs involved in breast cancer. We summarize the vital roles played by metabolism-regulating ncRNAs for endocrine therapy, targeted therapy, chemotherapy, immunotherapy, and radiotherapy resistance in breast cancer, as well as their potential as therapeutic targets and biomarkers. Difficulties and perspectives of current targeted metabolism and non-coding RNA therapeutic strategies are discussed.
Collapse
Affiliation(s)
- Shiliang Xu
- Department of Clinical Laboratory, The Second Affiliated Hospital of Soochow University, Suzhou, Jiangsu, 215004, People's Republic of China
| | - Lingxia Wang
- Department of Clinical Laboratory, The Second Affiliated Hospital of Soochow University, Suzhou, Jiangsu, 215004, People's Republic of China
| | - Yuexin Zhao
- Department of Clinical Laboratory, The Second Affiliated Hospital of Soochow University, Suzhou, Jiangsu, 215004, People's Republic of China
| | - Tong Mo
- Department of Clinical Laboratory, The Second Affiliated Hospital of Soochow University, Suzhou, Jiangsu, 215004, People's Republic of China
| | - Bo Wang
- Department of Oncology, The Second Affiliated Hospital of Soochow University, 1055 Sanxiang Road, Suzhou, Jiangsu, 215004, People's Republic of China
| | - Jun Lin
- Department of Clinical Laboratory, The Second Affiliated Hospital of Soochow University, Suzhou, Jiangsu, 215004, People's Republic of China.
| | - Huan Yang
- Department of Clinical Laboratory, The Second Affiliated Hospital of Soochow University, Suzhou, Jiangsu, 215004, People's Republic of China.
| |
Collapse
|
26
|
Chen J, Zhou Y, Liu Z, Lu Y, Jiang Y, Cao K, Zhou N, Wang D, Zhang C, Zhou N, Shi K, Zhang L, Zhou L, Wang Z, Zhang H, Tang K, Ma J, Lv J, Huang B. Hepatic glycogenesis antagonizes lipogenesis by blocking S1P via UDPG. Science 2024; 383:eadi3332. [PMID: 38359126 DOI: 10.1126/science.adi3332] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2023] [Accepted: 12/20/2023] [Indexed: 02/17/2024]
Abstract
The identification of mechanisms to store glucose carbon in the form of glycogen rather than fat in hepatocytes has important implications for the prevention of nonalcoholic fatty liver disease (NAFLD) and other chronic metabolic diseases. In this work, we show that glycogenesis uses its intermediate metabolite uridine diphosphate glucose (UDPG) to antagonize lipogenesis, thus steering both mouse and human hepatocytes toward storing glucose carbon as glycogen. The underlying mechanism involves transport of UDPG to the Golgi apparatus, where it binds to site-1 protease (S1P) and inhibits S1P-mediated cleavage of sterol regulatory element-binding proteins (SREBPs), thereby inhibiting lipogenesis in hepatocytes. Consistent with this mechanism, UDPG administration is effective at treating NAFLD in a mouse model and human organoids. These findings indicate a potential opportunity to ameliorate disordered fat metabolism in the liver.
Collapse
Affiliation(s)
- Jie Chen
- Department of Immunology and National Key Laboratory of Medical Molecular Biology, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100005, China
| | - Yabo Zhou
- Department of Immunology and National Key Laboratory of Medical Molecular Biology, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100005, China
| | - Zhuohang Liu
- Department of Immunology and National Key Laboratory of Medical Molecular Biology, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100005, China
| | - Yan Lu
- Department of Immunology and National Key Laboratory of Medical Molecular Biology, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100005, China
| | - Yishen Jiang
- Department of Immunology and National Key Laboratory of Medical Molecular Biology, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100005, China
| | - Kexin Cao
- Department of Immunology and National Key Laboratory of Medical Molecular Biology, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100005, China
| | - Nannan Zhou
- Department of Immunology and National Key Laboratory of Medical Molecular Biology, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100005, China
| | - Dianheng Wang
- Department of Immunology and National Key Laboratory of Medical Molecular Biology, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100005, China
| | - Chaoqi Zhang
- Department of Immunology and National Key Laboratory of Medical Molecular Biology, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100005, China
- Department of Thoracic Surgery, National Cancer Center/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100021, China
| | - Ning Zhou
- Department of Pathology, Sichuan Mianyang 404 Hospital, Sichuan 621000, China
| | - Keqing Shi
- Translational Medicine Laboratory, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou 325035, China
| | - Lu Zhang
- Tianjin Centers for Disease Control and Prevention, Tianjin 300011, China
| | - Li Zhou
- Department of Immunology and National Key Laboratory of Medical Molecular Biology, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100005, China
| | - Zhenfeng Wang
- Department of Immunology and National Key Laboratory of Medical Molecular Biology, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100005, China
| | - Huafeng Zhang
- Department of Pathology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Ke Tang
- Department of Biochemistry and Molecular Biology, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Jingwei Ma
- Department of Immunology, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Jiadi Lv
- Department of Immunology and National Key Laboratory of Medical Molecular Biology, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100005, China
| | - Bo Huang
- Department of Immunology and National Key Laboratory of Medical Molecular Biology, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100005, China
- Department of Biochemistry and Molecular Biology, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| |
Collapse
|
27
|
Yang Y, Teng H, Zhang Y, Wang F, Tang L, Zhang C, Hu Z, Chen Y, Ge Y, Wang Z, Yu Y. A glycosylation-related gene signature predicts prognosis, immune microenvironment infiltration, and drug sensitivity in glioma. Front Pharmacol 2024; 14:1259051. [PMID: 38293671 PMCID: PMC10824914 DOI: 10.3389/fphar.2023.1259051] [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: 07/15/2023] [Accepted: 12/11/2023] [Indexed: 02/01/2024] Open
Abstract
Glioma represents the most common primary cancer of the central nervous system in adults. Glycosylation is a prevalent post-translational modification that occurs in eukaryotic cells, leading to a wide array of modifications on proteins. We obtained the clinical information, bulk RNA-seq data, and single-cell RNA sequencing (scRNA-seq) from The Cancer Genome Atlas (TCGA), Chinese Glioma Genome Atlas (CGGA), Gene Expression Omnibus (GEO), and Repository of Molecular Brain Neoplasia Data (Rembrandt) databases. RNA sequencing data for normal brain tissues were accessed from the Genotype-Tissue Expression (GTEx) database. Then, the glycosylation genes that were differentially expressed were identified and further subjected to variable selection using a least absolute shrinkage and selection operator (LASSO)-regularized Cox model. We further conducted enrichment analysis, qPCR, nomogram, and single-cell transcriptome to detect the glycosylation signature. Drug sensitivity analysis was also conducted. A five-gene glycosylation signature (CHPF2, PYGL, GALNT13, EXT2, and COLGALT2) classified patients into low- or high-risk groups. Survival analysis, qPCR, ROC curves, and stratified analysis revealed worse outcomes in the high-risk group. Furthermore, GSEA and immune infiltration analysis indicated that the glycosylation signature has the potential to predict the immune response in glioma. In addition, four drugs (crizotinib, lapatinib, nilotinib, and topotecan) showed different responses between the two risk groups. Glioma cells had been classified into seven lines based on single-cell expression profiles. The five-gene glycosylation signature can accurately predict the prognosis of glioma and may offer additional guidance for immunotherapy.
Collapse
Affiliation(s)
- Yanbo Yang
- China-Japan Friendship Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Haiying Teng
- Department of Neurosurgery and Brain and Nerve Research Laboratory, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu, China
- Suzhou Medical College of Soochow University, Suzhou, Jiangsu, China
| | - Yulian Zhang
- Department of Neurosurgery, China-Japan Friendship Hospital, Beijing, China
| | - Fei Wang
- Department of Neurosurgery and Brain and Nerve Research Laboratory, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu, China
- Suzhou Medical College of Soochow University, Suzhou, Jiangsu, China
| | - Liyan Tang
- Department of Neurosurgery and Brain and Nerve Research Laboratory, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu, China
| | - Chuanpeng Zhang
- Department of Neurosurgery, China-Japan Friendship Hospital, Beijing, China
- Department of Neurosurgery, Peking University China-Japan Friendship School of Clinical Medicine, Beijing, China
| | - Ziyi Hu
- Department of Neurosurgery and Brain and Nerve Research Laboratory, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu, China
- Suzhou Medical College of Soochow University, Suzhou, Jiangsu, China
| | - Yuxuan Chen
- Department of Neurosurgery and Brain and Nerve Research Laboratory, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu, China
- Suzhou Medical College of Soochow University, Suzhou, Jiangsu, China
| | - Yi Ge
- The Affiliated Changzhou Second People’s Hospital of Nanjing Medical University, Nanjing, Jiangsu, China
| | - Zhong Wang
- Department of Neurosurgery and Brain and Nerve Research Laboratory, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu, China
| | - Yanbing Yu
- China-Japan Friendship Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| |
Collapse
|
28
|
Bizerra PFV, Gilglioni EH, Li HL, Go S, Oude Elferink RPJ, Verhoeven AJ, Chang JC. Opposite regulation of glycogen metabolism by cAMP produced in the cytosol and at the plasma membrane. BIOCHIMICA ET BIOPHYSICA ACTA. MOLECULAR CELL RESEARCH 2024; 1871:119585. [PMID: 37714306 DOI: 10.1016/j.bbamcr.2023.119585] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/24/2023] [Revised: 09/05/2023] [Accepted: 09/06/2023] [Indexed: 09/17/2023]
Abstract
Cyclic AMP is produced in cells by two different types of adenylyl cyclases: at the plasma membrane by the transmembrane adenylyl cyclases (tmACs, ADCY1~ADCY9) and in the cytosol by the evolutionarily more conserved soluble adenylyl cyclase (sAC, ADCY10). By employing high-resolution extracellular flux analysis in HepG2 cells to study glycogen breakdown in real time, we showed that cAMP regulates glycogen metabolism in opposite directions depending on its location of synthesis within cells and the downstream cAMP effectors. While the canonical tmAC-cAMP-PKA signaling promotes glycogenolysis, we demonstrate here that the non-canonical sAC-cAMP-Epac1 signaling suppresses glycogenolysis. Mechanistically, suppression of sAC-cAMP-Epac1 leads to Ser-15 phosphorylation and thereby activation of the liver-form glycogen phosphorylase to promote glycogenolysis. Our findings highlight the importance of cAMP microdomain organization for distinct metabolic regulation and establish sAC as a novel regulator of glycogen metabolism.
Collapse
Affiliation(s)
- Paulo F V Bizerra
- Tytgat Institute for Liver and Intestinal Research, Amsterdam UMC, University of Amsterdam, Amsterdam, the Netherlands; State University of Maringá, Paraná, Brazil
| | - Eduardo H Gilglioni
- Tytgat Institute for Liver and Intestinal Research, Amsterdam UMC, University of Amsterdam, Amsterdam, the Netherlands; Signal Transduction and Metabolism Laboratory, Université Libre de Bruxelles, Brussels, Belgium
| | - Hang Lam Li
- Tytgat Institute for Liver and Intestinal Research, Amsterdam UMC, University of Amsterdam, Amsterdam, the Netherlands; Amsterdam Gastroenterology Endocrinology Metabolism (AGEM) Research Institute, Amsterdam UMC, University of Amsterdam, Amsterdam, the Netherlands
| | - Simei Go
- Tytgat Institute for Liver and Intestinal Research, Amsterdam UMC, University of Amsterdam, Amsterdam, the Netherlands; Amsterdam Gastroenterology Endocrinology Metabolism (AGEM) Research Institute, Amsterdam UMC, University of Amsterdam, Amsterdam, the Netherlands; Department of Oncology, University of Oxford, Oxford, United Kingdom
| | - Ronald P J Oude Elferink
- Tytgat Institute for Liver and Intestinal Research, Amsterdam UMC, University of Amsterdam, Amsterdam, the Netherlands; Amsterdam Gastroenterology Endocrinology Metabolism (AGEM) Research Institute, Amsterdam UMC, University of Amsterdam, Amsterdam, the Netherlands
| | - Arthur J Verhoeven
- Tytgat Institute for Liver and Intestinal Research, Amsterdam UMC, University of Amsterdam, Amsterdam, the Netherlands
| | - Jung-Chin Chang
- Tytgat Institute for Liver and Intestinal Research, Amsterdam UMC, University of Amsterdam, Amsterdam, the Netherlands; Amsterdam Gastroenterology Endocrinology Metabolism (AGEM) Research Institute, Amsterdam UMC, University of Amsterdam, Amsterdam, the Netherlands; Division of Cell Biology, Metabolism & Cancer, Department of Biomolecular Health Sciences, Faculty of Veterinary Medicine, Utrecht University, Utrecht, the Netherlands.
| |
Collapse
|
29
|
Elkady EF, Ayoub HA, Ibrahim AM. Molluscicidal activity of calcium borate nanoparticles with kodom ball-flower structure on hematological, histological and biochemical parameters of Eobania vermiculata snails. PESTICIDE BIOCHEMISTRY AND PHYSIOLOGY 2024; 198:105716. [PMID: 38225073 DOI: 10.1016/j.pestbp.2023.105716] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/19/2023] [Revised: 11/16/2023] [Accepted: 11/27/2023] [Indexed: 01/17/2024]
Abstract
Land snails are the most harmful pests in agricultural fields. Eobania vermiculata is a widespread snail species that causes massive damage to all agricultural crops. Thus, the molluscicidal activity of calcium borate nanoparticles (CB-NPs) against Eobania vermiculata was evaluated and compared with metaldehyde (Gastrotox® E 5% G). The amorphous phase of CB-NPs was obtained after thermal treatment at a low temperature (500 °C) which conformed by X-ray diffraction (XRD) analysis. CB-NPs are composed of aggregated nano-sheets with an average thickness of 54 nm which enhanced their molluscicidal activity. These nano-sheets displayed meso-porous network architecture with pore diameters of 13.65 nm, and a 9.46 m2/g specific surface area. CB-NPs and metaldehyde (Gastrotox® E 5% G) exhibited molluscicidal effects on Eobania vermiculata snails with median lethal concentrations LC50 of 175.3 and 60.5 mg/l, respectively, after 72 h of exposure. The results also showed significant reductions of Eobania vermiculata snails hemocytes' mean total number, the levels of Testosterone (T) and Estrogen (E), alkaline phosphatase, acid phosphatase, albumin, and protein concentrations, succinate dehydrogenase, glucose, triglycerides and phospholipids levels, while significant increases in the phagocytic index and mortality index, both transaminases (ALT and AST) and glycogen phosphorylase concentration were observed after the exposure to LC50 of CB-NPs or metaldehyde (Gastrotox® E 5% G) compared to the control group. Therefore, CB-NPs could be used as an alternative molluscicide for controlling Eobania vermiculata, but further studies are needed to assess their effects on non-target organisms.
Collapse
Affiliation(s)
| | - Haytham A Ayoub
- Plant Protection Research Institute, Agricultural Research Center, Giza, Egypt
| | - Amina M Ibrahim
- Medical Malacology Department, Theodor Bilharz Research Institute, Giza, Egypt.
| |
Collapse
|
30
|
Davis S, Scott C, Oetjen J, Charles PD, Kessler BM, Ansorge O, Fischer R. Deep topographic proteomics of a human brain tumour. Nat Commun 2023; 14:7710. [PMID: 38001067 PMCID: PMC10673928 DOI: 10.1038/s41467-023-43520-8] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2023] [Accepted: 11/13/2023] [Indexed: 11/26/2023] Open
Abstract
The spatial organisation of cellular protein expression profiles within tissue determines cellular function and is key to understanding disease pathology. To define molecular phenotypes in the spatial context of tissue, there is a need for unbiased, quantitative technology capable of mapping proteomes within tissue structures. Here, we present a workflow for spatially-resolved, quantitative proteomics of tissue that generates maps of protein abundance across tissue slices derived from a human atypical teratoid-rhabdoid tumour at three spatial resolutions, the highest being 40 µm, to reveal distinct abundance patterns of thousands of proteins. We employ spatially-aware algorithms that do not require prior knowledge of the fine tissue structure to detect proteins and pathways with spatial abundance patterns and correlate proteins in the context of tissue heterogeneity and cellular features such as extracellular matrix or proximity to blood vessels. We identify PYGL, ASPH and CD45 as spatial markers for tumour boundary and reveal immune response-driven, spatially-organised protein networks of the extracellular tumour matrix. Overall, we demonstrate spatially-aware deep proteo-phenotyping of tissue heterogeneity, to re-define understanding tissue biology and pathology at the molecular level.
Collapse
Affiliation(s)
- Simon Davis
- Target Discovery Institute, Centre for Medicines Discovery, Nuffield Department of Medicine, University of Oxford, Roosevelt Drive, Oxford, OX3 7FZ, UK
- Chinese Academy for Medical Sciences Oxford Institute, Nuffield Department of Medicine, University of Oxford, Roosevelt Drive, Oxford, OX3 7FZ, UK
| | - Connor Scott
- Academic Unit of Neuropathology, Nuffield Department of Clinical Neurosciences, University of Oxford, John Radcliffe Hospital, Oxford, OX3 9DU, UK
| | - Janina Oetjen
- Bruker Daltonics GmbH & Co. KG, Fahrenheitstraße 4, 28359, Bremen, Germany
| | - Philip D Charles
- Target Discovery Institute, Centre for Medicines Discovery, Nuffield Department of Medicine, University of Oxford, Roosevelt Drive, Oxford, OX3 7FZ, UK
- Big Data Institute, Nuffield Department of Medicine, University of Oxford, Roosevelt Drive, Oxford, OX3 7FZ, UK
| | - Benedikt M Kessler
- Target Discovery Institute, Centre for Medicines Discovery, Nuffield Department of Medicine, University of Oxford, Roosevelt Drive, Oxford, OX3 7FZ, UK
- Chinese Academy for Medical Sciences Oxford Institute, Nuffield Department of Medicine, University of Oxford, Roosevelt Drive, Oxford, OX3 7FZ, UK
| | - Olaf Ansorge
- Academic Unit of Neuropathology, Nuffield Department of Clinical Neurosciences, University of Oxford, John Radcliffe Hospital, Oxford, OX3 9DU, UK
| | - Roman Fischer
- Target Discovery Institute, Centre for Medicines Discovery, Nuffield Department of Medicine, University of Oxford, Roosevelt Drive, Oxford, OX3 7FZ, UK.
- Chinese Academy for Medical Sciences Oxford Institute, Nuffield Department of Medicine, University of Oxford, Roosevelt Drive, Oxford, OX3 7FZ, UK.
| |
Collapse
|
31
|
Lee SCES, Pyo AHA, Koritzinsky M. Longitudinal dynamics of the tumor hypoxia response: From enzyme activity to biological phenotype. SCIENCE ADVANCES 2023; 9:eadj6409. [PMID: 37992163 PMCID: PMC10664991 DOI: 10.1126/sciadv.adj6409] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/07/2023] [Accepted: 10/23/2023] [Indexed: 11/24/2023]
Abstract
Poor oxygenation (hypoxia) is a common spatially heterogeneous feature of human tumors. Biological responses to tumor hypoxia are orchestrated by the decreased activity of oxygen-dependent enzymes. The affinity of these enzymes for oxygen positions them along a continuum of oxygen sensing that defines their roles in launching reactive and adaptive cellular responses. These responses encompass regulation of all steps in the central dogma, with rapid perturbation of the metabolome and proteome followed by more persistent reprogramming of the transcriptome and epigenome. Core hypoxia response genes and pathways are commonly regulated at multiple inflection points, fine-tuning the dependencies on oxygen concentration and hypoxia duration. Ultimately, shifts in the activity of oxygen-sensing enzymes directly or indirectly endow cells with intrinsic hypoxia tolerance and drive processes that are associated with aggressive phenotypes in cancer including angiogenesis, migration, invasion, immune evasion, epithelial mesenchymal transition, and stemness.
Collapse
Affiliation(s)
- Sandy Che-Eun S. Lee
- Princess Margaret Cancer Center, University Health Network, Toronto, Ontario, Canada
- Institute of Medical Science, University of Toronto, Toronto, Ontario, Canada
| | - Andrea Hye An Pyo
- Princess Margaret Cancer Center, University Health Network, Toronto, Ontario, Canada
- Institute of Medical Science, University of Toronto, Toronto, Ontario, Canada
| | - Marianne Koritzinsky
- Princess Margaret Cancer Center, University Health Network, Toronto, Ontario, Canada
- Institute of Medical Science, University of Toronto, Toronto, Ontario, Canada
- Department of Radiation Oncology, University of Toronto, Toronto, Ontario, Canada
- Department of Medical Biophysics, University of Toronto, Toronto, Ontario, Canada
| |
Collapse
|
32
|
Bertrand M, Szeremeta F, Hervouet-Coste N, Sarou-Kanian V, Landon C, Morisset-Lopez S, Decoville M. An adult Drosophila glioma model to highlight metabolic dysfunctions and evaluate the role of the serotonin 5-HT 7 receptor as a potential therapeutic target. FASEB J 2023; 37:e23230. [PMID: 37781977 DOI: 10.1096/fj.202300783rr] [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: 04/23/2023] [Revised: 08/31/2023] [Accepted: 09/18/2023] [Indexed: 10/03/2023]
Abstract
Gliomas account for 50% of brain cancers and are therefore the most common brain tumors. Molecular alterations involved in adult gliomas have been identified and mainly affect tyrosine kinase receptors with amplification and/or mutation of the epidermal growth factor receptor (EGFR) and its associated signaling pathways. Several targeted therapies have been developed, but current treatments remain ineffective for glioblastomas, the most severe forms. Thus, it is a priority to identify new pharmacological targets. Drosophila glioma models established in larvae and adults are useful to identify new genes and signaling pathways involved in glioma progression. Here, we used a Drosophila glioma model in adults, to characterize metabolic disturbances associated with glioma and assess the consequences of 5-HT7 R expression on glioma development. First, by using in vivo magnetic resonance imaging, we have shown that expression of the constitutively active forms of EGFR and PI3K in adult glial cells induces brain enlargement. Then, we explored altered cellular metabolism by using high-resolution magic angle spinning NMR and 1 H-13 C heteronuclear single quantum coherence solution states. Discriminant metabolites identified highlight the rewiring of metabolic pathways in glioma and associated cachexia phenotypes. Finally, the expression of 5-HT7 R in this adult model attenuates phenotypes associated with glioma development. Collectively, this whole-animal approach in Drosophila allowed us to provide several rapid and robust phenotype readouts, such as enlarged brain volume and glioma-associated cachexia, as well as to determine the metabolic pathways involved in glioma genesis and finally to confirm the interest of the 5-HT7 R in the treatment of glioma.
Collapse
Affiliation(s)
- Marylène Bertrand
- Centre de Biophysique Moléculaire-CBM, UPR 4301, CNRS, Orléans, France
| | | | | | - Vincent Sarou-Kanian
- Conditions Extrêmes et Matériaux: Haute Température et Irradiation-CEMHTI-CNRS UPR 3079, Orléans, France
| | - Céline Landon
- Centre de Biophysique Moléculaire-CBM, UPR 4301, CNRS, Orléans, France
| | | | - Martine Decoville
- Centre de Biophysique Moléculaire-CBM, UPR 4301, CNRS, Orléans, France
- UFR Sciences et Techniques, Université d'Orléans, Orléans, France
| |
Collapse
|
33
|
Mancini MC, Noland RC, Collier JJ, Burke SJ, Stadler K, Heden TD. Lysosomal glucose sensing and glycophagy in metabolism. Trends Endocrinol Metab 2023; 34:764-777. [PMID: 37633800 PMCID: PMC10592240 DOI: 10.1016/j.tem.2023.07.008] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/18/2023] [Revised: 07/27/2023] [Accepted: 07/28/2023] [Indexed: 08/28/2023]
Abstract
Lysosomes are cellular organelles that function to catabolize both extra- and intracellular cargo, act as a platform for nutrient sensing, and represent a core signaling node integrating bioenergetic cues to changes in cellular metabolism. Although lysosomal amino acid and lipid sensing in metabolism has been well characterized, lysosomal glucose sensing and the role of lysosomes in glucose metabolism is unrefined. This review will highlight the role of the lysosome in glucose metabolism with a focus on lysosomal glucose and glycogen sensing, glycophagy, and lysosomal glucose transport and how these processes impact autophagy and energy metabolism. Additionally, the role of lysosomal glucose metabolism in genetic and metabolic diseases will be briefly discussed.
Collapse
Affiliation(s)
- Melina C Mancini
- Pennington Biomedical Research Center, Baton Rouge, LA 70808, USA
| | - Robert C Noland
- Pennington Biomedical Research Center, Baton Rouge, LA 70808, USA
| | - J Jason Collier
- Pennington Biomedical Research Center, Baton Rouge, LA 70808, USA
| | - Susan J Burke
- Pennington Biomedical Research Center, Baton Rouge, LA 70808, USA
| | | | - Timothy D Heden
- Pennington Biomedical Research Center, Baton Rouge, LA 70808, USA.
| |
Collapse
|
34
|
Zhou Z, Zhang H, Tao Y, Zang J, Zhao J, Li H, Wang Y, Wang T, Zhao H, Wang F, Guo C, Zhu F, Mao H, Liu F, Zhang L, Wang Q. FGF21 alleviates adipose stem cell senescence via CD90 glycosylation-dependent glucose influx in remodeling healthy white adipose tissue. Redox Biol 2023; 67:102877. [PMID: 37690164 PMCID: PMC10497791 DOI: 10.1016/j.redox.2023.102877] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2023] [Revised: 09/04/2023] [Accepted: 09/04/2023] [Indexed: 09/12/2023] Open
Abstract
The senescence of adipose stem cells (ASCs) impairs healthy adipose tissue remodeling, causing metabolic maladaptation to energy surplus. The intrinsic molecular pathways and potential therapy targets for ASC senescence are largely unclear. Here, we showed that visceral ASCs were prone to senescence that was caused by reactive oxygen species (ROS) overload, especially mitochondrial ROS. These senescent ASCs failed to sustain efficient glucose influx, pentose phosphate pathway (PPP) and redox homeostasis. We showed that CD90 silence restricted the glucose uptake by ASCs and thus disrupted their PPP and anti-oxidant system, resulting in ASC senescence. Notably, fibroblast growth factor 21 (FGF21) treatment significantly reduced the senescent phenotypes of ASCs by augmenting CD90 protein via glycosylation, which promoted glucose influx via the AKT-GLUT4 axis and therefore mitigated ROS overload. For diet-induced obese mice, chronic administration of low-dose FGF21 relieved their visceral white adipose tissue (VAT) dysfunction and systemic metabolic disorders. In particular, VAT homeostasis was restored in FGF21-treated obese mice, where ASC repertoire was markedly recovered, accompanied by CD90 elevation and anti-senescent phenotypes in these ASCs. Collectively, we reveal a molecular mechanism of ASC senescence by which CD90 downregulation interferes glucose influx into PPP and redox homeostasis. And we propose a FGF21-based strategy for healthy VAT remodeling, which targets CD90 glycosylation to correct ASC senescence and therefore combat obesity-related metabolic dysfunction.
Collapse
Affiliation(s)
- Zixin Zhou
- Key Laboratory of Infection and Immunity of Shandong Province, Department of Immunology, School of Basic Medical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, 250012, China
| | - Huiying Zhang
- Key Laboratory of Infection and Immunity of Shandong Province, Department of Immunology, School of Basic Medical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, 250012, China
| | - Yan Tao
- Key Laboratory of Infection and Immunity of Shandong Province, Department of Immunology, School of Basic Medical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, 250012, China
| | - Jinhao Zang
- Key Laboratory of Infection and Immunity of Shandong Province, Department of Immunology, School of Basic Medical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, 250012, China
| | - Jingyuan Zhao
- Key Laboratory of Infection and Immunity of Shandong Province, Department of Immunology, School of Basic Medical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, 250012, China
| | - Huijie Li
- Key Laboratory of Infection and Immunity of Shandong Province, Department of Immunology, School of Basic Medical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, 250012, China
| | - Yalin Wang
- Key Laboratory of Infection and Immunity of Shandong Province, Department of Immunology, School of Basic Medical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, 250012, China
| | - Tianci Wang
- Key Laboratory of Infection and Immunity of Shandong Province, Department of Immunology, School of Basic Medical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, 250012, China
| | - Hui Zhao
- Department of Clinical Laboratory, The Second Hospital, Cheeloo College of Medicine, Shandong University, Jinan, 250033, China
| | - Fuwu Wang
- Key Laboratory for Experimental Teratology of Ministry of Education, Shandong Key Laboratory of Mental Disorders, Department of Histology and Embryology, School of Basic Medical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, 250012, China
| | - Chun Guo
- Key Laboratory of Infection and Immunity of Shandong Province, Department of Immunology, School of Basic Medical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, 250012, China
| | - Faliang Zhu
- Key Laboratory of Infection and Immunity of Shandong Province, Department of Immunology, School of Basic Medical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, 250012, China
| | - Haiting Mao
- Department of Clinical Laboratory, The Second Hospital, Cheeloo College of Medicine, Shandong University, Jinan, 250033, China
| | - Fengming Liu
- Key Laboratory of Infection and Immunity of Shandong Province, Department of Immunology, School of Basic Medical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, 250012, China
| | - Lining Zhang
- Key Laboratory of Infection and Immunity of Shandong Province, Department of Immunology, School of Basic Medical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, 250012, China
| | - Qun Wang
- Key Laboratory of Infection and Immunity of Shandong Province, Department of Immunology, School of Basic Medical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, 250012, China.
| |
Collapse
|
35
|
Pervin J, Asad M, Cao S, Jang GH, Feizi N, Haibe-Kains B, Karasinska JM, O’Kane GM, Gallinger S, Schaeffer DF, Renouf DJ, Zogopoulos G, Bathe OF. Clinically impactful metabolic subtypes of pancreatic ductal adenocarcinoma (PDAC). Front Genet 2023; 14:1282824. [PMID: 38028629 PMCID: PMC10643182 DOI: 10.3389/fgene.2023.1282824] [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: 08/24/2023] [Accepted: 10/06/2023] [Indexed: 12/01/2023] Open
Abstract
Background: Pancreatic ductal adenocarcinoma (PDAC) is a lethal disease characterized by a diverse tumor microenvironment. The heterogeneous cellular composition of PDAC makes it challenging to study molecular features of tumor cells using extracts from bulk tumor. The metabolic features in tumor cells from clinical samples are poorly understood, and their impact on clinical outcomes are unknown. Our objective was to identify the metabolic features in the tumor compartment that are most clinically impactful. Methods: A computational deconvolution approach using the DeMixT algorithm was applied to bulk RNASeq data from The Cancer Genome Atlas to determine the proportion of each gene's expression that was attributable to the tumor compartment. A machine learning algorithm designed to identify features most closely associated with survival outcomes was used to identify the most clinically impactful metabolic genes. Results: Two metabolic subtypes (M1 and M2) were identified, based on the pattern of expression of the 26 most important metabolic genes. The M2 phenotype had a significantly worse survival, which was replicated in three external PDAC cohorts. This PDAC subtype was characterized by net glycogen catabolism, accelerated glycolysis, and increased proliferation and cellular migration. Single cell data demonstrated substantial intercellular heterogeneity in the metabolic features that typified this aggressive phenotype. Conclusion: By focusing on features within the tumor compartment, two novel and clinically impactful metabolic subtypes of PDAC were identified. Our study emphasizes the challenges of defining tumor phenotypes in the face of the significant intratumoral heterogeneity that typifies PDAC. Further studies are required to understand the microenvironmental factors that drive the appearance of the metabolic features characteristic of the aggressive M2 PDAC phenotype.
Collapse
Affiliation(s)
- Jannat Pervin
- Department of Oncology, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada
| | - Mohammad Asad
- Department of Biochemistry and Molecular Biology, University of Calgary, Calgary, AB, Canada
| | - Shaolong Cao
- Department of Bioinformatics and Computational Biology, University of Texas MD Anderson Cancer Centre, Houston, TX, United States
| | - Gun Ho Jang
- Ontario Institute for Cancer Research, Toronto, ON, Canada
| | - Nikta Feizi
- Princess Margaret Cancer Centre, University Health Network, Toronto, ON, Canada
| | | | | | - Grainne M. O’Kane
- University Health Network, University of Toronto, Toronto, ON, Canada
| | | | - David F. Schaeffer
- Department of Pathology and Laboratory Medicine, University of British Columbia, Vancouver, BC, Canada
| | - Daniel J. Renouf
- Department of Medicine, University of British Columbia, Vancouver, BC, Canada
| | - George Zogopoulos
- Department of Surgery, McGill University Health Centre, McGill University, Montreal, QC, Canada
| | - Oliver F. Bathe
- Department of Oncology, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada
- Department of Surgery, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada
| |
Collapse
|
36
|
Angarola BL, Sharma S, Katiyar N, Gu Kang H, Nehar-Belaid D, Park S, Gott R, Eryilmaz GN, LaBarge MA, Palucka K, Chuang JH, Korstanje R, Ucar D, Anczukow O. Comprehensive single cell aging atlas of mammary tissues reveals shared epigenomic and transcriptomic signatures of aging and cancer. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.10.20.563147. [PMID: 37961129 PMCID: PMC10634680 DOI: 10.1101/2023.10.20.563147] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/15/2023]
Abstract
Aging is the greatest risk factor for breast cancer; however, how age-related cellular and molecular events impact cancer initiation is unknown. We investigate how aging rewires transcriptomic and epigenomic programs of mouse mammary glands at single cell resolution, yielding a comprehensive resource for aging and cancer biology. Aged epithelial cells exhibit epigenetic and transcriptional changes in metabolic, pro-inflammatory, or cancer-associated genes. Aged stromal cells downregulate fibroblast marker genes and upregulate markers of senescence and cancer-associated fibroblasts. Among immune cells, distinct T cell subsets (Gzmk+, memory CD4+, γδ) and M2-like macrophages expand with age. Spatial transcriptomics reveal co-localization of aged immune and epithelial cells in situ. Lastly, transcriptional signatures of aging mammary cells are found in human breast tumors, suggesting mechanistic links between aging and cancer. Together, these data uncover that epithelial, immune, and stromal cells shift in proportions and cell identity, potentially impacting cell plasticity, aged microenvironment, and neoplasia risk.
Collapse
Affiliation(s)
| | | | - Neerja Katiyar
- The Jackson Laboratory for Genomic Medicine, Farmington, CT, USA
| | - Hyeon Gu Kang
- The Jackson Laboratory for Genomic Medicine, Farmington, CT, USA
| | | | - SungHee Park
- The Jackson Laboratory for Genomic Medicine, Farmington, CT, USA
| | | | - Giray N Eryilmaz
- The Jackson Laboratory for Genomic Medicine, Farmington, CT, USA
| | - Mark A LaBarge
- Beckman Research Institute at City of Hope, Duarte, CA, USA
| | - Karolina Palucka
- The Jackson Laboratory for Genomic Medicine, Farmington, CT, USA
| | - Jeffrey H Chuang
- The Jackson Laboratory for Genomic Medicine, Farmington, CT, USA
| | | | - Duygu Ucar
- The Jackson Laboratory for Genomic Medicine, Farmington, CT, USA
- Department of Genetics and Genome Sciences, UConn Health, Farmington, CT, USA
- Institute for Systems Genomics, UConn Health, Farmington, CT, USA
| | - Olga Anczukow
- The Jackson Laboratory for Genomic Medicine, Farmington, CT, USA
- Department of Genetics and Genome Sciences, UConn Health, Farmington, CT, USA
- Institute for Systems Genomics, UConn Health, Farmington, CT, USA
| |
Collapse
|
37
|
Tsuji T, Tsunematsu H, Imanishi M, Denda M, Tsuchiya K, Otaka A. Enhanced tumor specific drug release by hypoxia sensitive dual-prodrugs based on 2-nitroimidazole. Bioorg Med Chem Lett 2023; 95:129484. [PMID: 37716415 DOI: 10.1016/j.bmcl.2023.129484] [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: 07/04/2023] [Revised: 09/01/2023] [Accepted: 09/11/2023] [Indexed: 09/18/2023]
Abstract
Hypoxia in cancer is important in the development of cancer-selective medicines. Here, a novel hypoxia-responsible dual-prodrug is described. We designed and synthesized 2-nitroimidazole derivatives which spontaneously release both a PYG inhibitor and gemcitabine under hypoxic conditions. One such derivative, a prodrug 9 was found to be stable against chemical and enzymatic hydrolysis, and upon chemical reduction of the nitro group on imidazole, successfully releases both drugs. In an in vitro proliferation assay using human pancreatic cells, compound 9 exhibited significant anti-proliferative effects in hypoxia but fewer effects in normoxia. Consequently, prodrug 9 should be useful for cancer treatment due to its improved cancer selectivity and potential to overcome drug resistance.
Collapse
Affiliation(s)
- Takashi Tsuji
- Institute of Biomedical Sciences and Graduate School of Pharmaceutical Sciences, Tokushima University, Tokushima 770-8505, Japan
| | - Honoka Tsunematsu
- Institute of Biomedical Sciences and Graduate School of Pharmaceutical Sciences, Tokushima University, Tokushima 770-8505, Japan
| | - Masaki Imanishi
- Institute of Biomedical Sciences and Graduate School of Pharmaceutical Sciences, Tokushima University, Tokushima 770-8505, Japan
| | - Masaya Denda
- Institute of Biomedical Sciences and Graduate School of Pharmaceutical Sciences, Tokushima University, Tokushima 770-8505, Japan
| | - Koichiro Tsuchiya
- Institute of Biomedical Sciences and Graduate School of Pharmaceutical Sciences, Tokushima University, Tokushima 770-8505, Japan
| | - Akira Otaka
- Institute of Biomedical Sciences and Graduate School of Pharmaceutical Sciences, Tokushima University, Tokushima 770-8505, Japan.
| |
Collapse
|
38
|
Zhou Q, Tao C, Yuan J, Pan F, Wang R. Ferroptosis, a subtle talk between immune system and cancer cells: To be or not to be? Biomed Pharmacother 2023; 165:115251. [PMID: 37523985 DOI: 10.1016/j.biopha.2023.115251] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2023] [Revised: 07/25/2023] [Accepted: 07/27/2023] [Indexed: 08/02/2023] Open
Abstract
Ferroptosis, an established form of programmed cell death discovered in 2012, is characterized by an imbalance in iron metabolism, lipid metabolism, and antioxidant metabolism. Activated CD8 + T cells can trigger ferroptosis in tumor cells by releasing interferon-γ, which initiates the ferroptosis program. Despite the remarkable progress made in treating various tumors with immunotherapy, such as anti-PD1/PDL1, there are still significant challenges to overcome, including limited treatment options and drug resistance. In this review, we exam the potential biological significance of the ferroptosis phenotype using bioinformatics and review the latest advancements in understanding the mechanism of ferroptosis-mediated anti-tumor immunotherapy. Furthermore, we revisit the host immune system, immune microenvironment, ferroptotic defense system, metabolic reprogramming, and key genes that regulate the occurrence and resistance of ferroptosis of tumor cell. Additionally, several immune-combined ferroptosis treatment strategies were put forward to improve immunotherapy efficacy and to provide new insights into reversing anti-tumor immune drug resistance.
Collapse
Affiliation(s)
- Qiong Zhou
- Department of Medical Oncology, Jinling Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, Jiangsu Province 210093, PR China.
| | - Chunyu Tao
- Department of Medical Oncology, Jinling Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, Jiangsu Province 210093, PR China.
| | - Jiakai Yuan
- Department of Medical Oncology, Jinling Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, Jiangsu Province 210093, PR China.
| | - Fan Pan
- Department of Medical Oncology, Jinling Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, Jiangsu Province 210093, PR China.
| | - Rui Wang
- Department of Medical Oncology, Jinling Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, Jiangsu Province 210093, PR China.
| |
Collapse
|
39
|
Mathomes RT, Koulas SM, Tsialtas I, Stravodimos G, Welsby PJ, Psarra AMG, Stasik I, Leonidas DD, Hayes JM. Multidisciplinary docking, kinetics and X-ray crystallography studies of baicalein acting as a glycogen phosphorylase inhibitor and determination of its' potential against glioblastoma in cellular models. Chem Biol Interact 2023; 382:110568. [PMID: 37277066 DOI: 10.1016/j.cbi.2023.110568] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2023] [Revised: 05/19/2023] [Accepted: 05/24/2023] [Indexed: 06/07/2023]
Abstract
Glycogen phosphorylase (GP) is the rate-determining enzyme in the glycogenolysis pathway. Glioblastoma (GBM) is amongst the most aggressive cancers of the central nervous system. The role of GP and glycogen metabolism in the context of cancer cell metabolic reprogramming is recognised, so that GP inhibitors may have potential treatment benefits. Here, baicalein (5,6,7-trihydroxyflavone) is studied as a GP inhibitor, and for its effects on glycogenolysis and GBM at the cellular level. The compound is revealed as a potent GP inhibitor against human brain GPa (Ki = 32.54 μM), human liver GPa (Ki = 8.77 μM) and rabbit muscle GPb (Ki = 5.66 μM) isoforms. It is also an effective inhibitor of glycogenolysis (IC50 = 119.6 μM), measured in HepG2 cells. Most significantly, baicalein demonstrated anti-cancer potential through concentration- and time-dependent decrease in cell viability for three GBM cell-lines (U-251 MG, U-87 MG, T98-G) with IC50 values of ∼20-55 μM (48- and 72-h). Its effectiveness against T98-G suggests potential against GBM with resistance to temozolomide (the first-line therapy) due to a positive O6-methylguanine-DNA methyltransferase (MGMT) status. The solved X-ray structure of rabbit muscle GP-baicalein complex will facilitate structure-based design of GP inhibitors. Further exploration of baicalein and other GP inhibitors with different isoform specificities against GBM is suggested.
Collapse
Affiliation(s)
- Rachel T Mathomes
- School of Pharmacy & Biomedical Sciences, University of Central Lancashire, Preston, PR1 2HE, United Kingdom
| | - Symeon M Koulas
- Department of Biochemistry & Biotechnology, University of Thessaly, Biopolis, 41500, Larissa, Greece
| | - Ioannis Tsialtas
- Department of Biochemistry & Biotechnology, University of Thessaly, Biopolis, 41500, Larissa, Greece
| | - George Stravodimos
- Department of Biochemistry & Biotechnology, University of Thessaly, Biopolis, 41500, Larissa, Greece
| | - Philip J Welsby
- Department of Postgraduate Medical Education, Edge Hill University, Ormskirk, L39 4QP, United Kingdom
| | - Anna-Maria G Psarra
- Department of Biochemistry & Biotechnology, University of Thessaly, Biopolis, 41500, Larissa, Greece
| | - Izabela Stasik
- School of Pharmacy & Biomedical Sciences, University of Central Lancashire, Preston, PR1 2HE, United Kingdom
| | - Demetres D Leonidas
- Department of Biochemistry & Biotechnology, University of Thessaly, Biopolis, 41500, Larissa, Greece.
| | - Joseph M Hayes
- School of Pharmacy & Biomedical Sciences, University of Central Lancashire, Preston, PR1 2HE, United Kingdom.
| |
Collapse
|
40
|
Mukherjee A, Bezwada D, Greco F, Zandbergen M, Shen T, Chiang CY, Tasdemir M, Fahrmann J, Grapov D, La Frano MR, Vu HS, Faubert B, Newman JW, McDonnell LA, Nezi L, Fiehn O, DeBerardinis RJ, Lengyel E. Adipocytes reprogram cancer cell metabolism by diverting glucose towards glycerol-3-phosphate thereby promoting metastasis. Nat Metab 2023; 5:1563-1577. [PMID: 37653041 DOI: 10.1038/s42255-023-00879-8] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/05/2021] [Accepted: 07/27/2023] [Indexed: 09/02/2023]
Abstract
In the tumor microenvironment, adipocytes function as an alternate fuel source for cancer cells. However, whether adipocytes influence macromolecular biosynthesis in cancer cells is unknown. Here we systematically characterized the bidirectional interaction between primary human adipocytes and ovarian cancer (OvCa) cells using multi-platform metabolomics, imaging mass spectrometry, isotope tracing and gene expression analysis. We report that, in OvCa cells co-cultured with adipocytes and in metastatic tumors, a part of the glucose from glycolysis is utilized for the biosynthesis of glycerol-3-phosphate (G3P). Normoxic HIF1α protein regulates the altered flow of glucose-derived carbons in cancer cells, resulting in increased glycerophospholipids and triacylglycerol synthesis. The knockdown of HIF1α or G3P acyltransferase 3 (a regulatory enzyme of glycerophospholipid synthesis) reduced metastasis in xenograft models of OvCa. In summary, we show that, in an adipose-rich tumor microenvironment, cancer cells generate G3P as a precursor for critical membrane and signaling components, thereby promoting metastasis. Targeting biosynthetic processes specific to adipose-rich tumor microenvironments might be an effective strategy against metastasis.
Collapse
Affiliation(s)
- Abir Mukherjee
- Department of Obstetrics and Gynecology/Section of Gynecologic Oncology-Center for Integrative Sciences, University of Chicago, Chicago, IL, USA
| | - Divya Bezwada
- Children's Medical Center Research Institute, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Francesco Greco
- Fondazione Pisana per la Scienza ONLUS, San Giuliano Terme, Italy
- Institute of Life Sciences, Sant'Anna School of Advanced Studies, Pisa, Italy
| | - Malu Zandbergen
- Department of Obstetrics and Gynecology/Section of Gynecologic Oncology-Center for Integrative Sciences, University of Chicago, Chicago, IL, USA
| | - Tong Shen
- NIH West Coast Metabolomics Center, University of California, Davis, CA, USA
| | - Chun-Yi Chiang
- Department of Obstetrics and Gynecology/Section of Gynecologic Oncology-Center for Integrative Sciences, University of Chicago, Chicago, IL, USA
| | - Medine Tasdemir
- Department of Obstetrics and Gynecology/Section of Gynecologic Oncology-Center for Integrative Sciences, University of Chicago, Chicago, IL, USA
| | - Johannes Fahrmann
- Department of Clinical Cancer Prevention, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Dmitry Grapov
- NIH West Coast Metabolomics Center, University of California, Davis, CA, USA
| | - Michael R La Frano
- NIH West Coast Metabolomics Center, University of California, Davis, CA, USA
| | - Hieu S Vu
- Children's Medical Center Research Institute, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Brandon Faubert
- Department of Medicine/Section of Hematology and Oncology, University of Chicago, Chicago, IL, USA
| | - John W Newman
- NIH West Coast Metabolomics Center, University of California, Davis, CA, USA
| | - Liam A McDonnell
- Institute of Life Sciences, Sant'Anna School of Advanced Studies, Pisa, Italy
| | - Luigi Nezi
- Department of Experimental Oncology, IRCCS European Institute of Oncology, Milano, Italy
| | - Oliver Fiehn
- NIH West Coast Metabolomics Center, University of California, Davis, CA, USA
| | - Ralph J DeBerardinis
- Children's Medical Center Research Institute, University of Texas Southwestern Medical Center, Dallas, TX, USA
- 9Howard Hughes Medical Institute, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Ernst Lengyel
- Department of Obstetrics and Gynecology/Section of Gynecologic Oncology-Center for Integrative Sciences, University of Chicago, Chicago, IL, USA.
| |
Collapse
|
41
|
Mahé M, Rios-Fuller TJ, Karolin A, Schneider RJ. Genetics of enzymatic dysfunctions in metabolic disorders and cancer. Front Oncol 2023; 13:1230934. [PMID: 37601653 PMCID: PMC10433910 DOI: 10.3389/fonc.2023.1230934] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2023] [Accepted: 07/19/2023] [Indexed: 08/22/2023] Open
Abstract
Inherited metabolic disorders arise from mutations in genes involved in the biogenesis, assembly, or activity of metabolic enzymes, leading to enzymatic deficiency and severe metabolic impairments. Metabolic enzymes are essential for the normal functioning of cells and are involved in the production of amino acids, fatty acids and nucleotides, which are essential for cell growth, division and survival. When the activity of metabolic enzymes is disrupted due to mutations or changes in expression levels, it can result in various metabolic disorders that have also been linked to cancer development. However, there remains much to learn regarding the relationship between the dysregulation of metabolic enzymes and metabolic adaptations in cancer cells. In this review, we explore how dysregulated metabolism due to the alteration or change of metabolic enzymes in cancer cells plays a crucial role in tumor development, progression, metastasis and drug resistance. In addition, these changes in metabolism provide cancer cells with a number of advantages, including increased proliferation, resistance to apoptosis and the ability to evade the immune system. The tumor microenvironment, genetic context, and different signaling pathways further influence this interplay between cancer and metabolism. This review aims to explore how the dysregulation of metabolic enzymes in specific pathways, including the urea cycle, glycogen storage, lysosome storage, fatty acid oxidation, and mitochondrial respiration, contributes to the development of metabolic disorders and cancer. Additionally, the review seeks to shed light on why these enzymes represent crucial potential therapeutic targets and biomarkers in various cancer types.
Collapse
Affiliation(s)
| | | | | | - Robert J. Schneider
- Department of Microbiology, Grossman NYU School of Medicine, New York, NY, United States
| |
Collapse
|
42
|
Copeland CA, Olenchock BA, Ziehr D, McGarrity S, Leahy K, Young JD, Loscalzo J, Oldham WM. MYC overrides HIF-1α to regulate proliferating primary cell metabolism in hypoxia. eLife 2023; 12:e82597. [PMID: 37428010 DOI: 10.7554/elife.82597] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2022] [Accepted: 06/27/2023] [Indexed: 07/11/2023] Open
Abstract
Hypoxia requires metabolic adaptations to sustain energetically demanding cellular activities. While the metabolic consequences of hypoxia have been studied extensively in cancer cell models, comparatively little is known about how primary cell metabolism responds to hypoxia. Thus, we developed metabolic flux models for human lung fibroblast and pulmonary artery smooth muscle cells proliferating in hypoxia. Unexpectedly, we found that hypoxia decreased glycolysis despite activation of hypoxia-inducible factor 1α (HIF-1α) and increased glycolytic enzyme expression. While HIF-1α activation in normoxia by prolyl hydroxylase (PHD) inhibition did increase glycolysis, hypoxia blocked this effect. Multi-omic profiling revealed distinct molecular responses to hypoxia and PHD inhibition, and suggested a critical role for MYC in modulating HIF-1α responses to hypoxia. Consistent with this hypothesis, MYC knockdown in hypoxia increased glycolysis and MYC over-expression in normoxia decreased glycolysis stimulated by PHD inhibition. These data suggest that MYC signaling in hypoxia uncouples an increase in HIF-dependent glycolytic gene transcription from glycolytic flux.
Collapse
Affiliation(s)
- Courtney A Copeland
- Department of Medicine, Brigham and Women's Hospital, Boston, United States
- Department of Medicine, Harvard Medical School, Boston, United States
| | - Benjamin A Olenchock
- Department of Medicine, Brigham and Women's Hospital, Boston, United States
- Department of Medicine, Harvard Medical School, Boston, United States
| | - David Ziehr
- Department of Medicine, Brigham and Women's Hospital, Boston, United States
- Department of Medicine, Harvard Medical School, Boston, United States
- Department of Medicine, Massachusetts General Hospital, Boston, United States
| | - Sarah McGarrity
- Department of Medicine, Brigham and Women's Hospital, Boston, United States
- Department of Medicine, Harvard Medical School, Boston, United States
- Center for Systems Biology, School of Health Sciences, University of Iceland, Reykjavik, Iceland
| | - Kevin Leahy
- Department of Medicine, Brigham and Women's Hospital, Boston, United States
- Department of Medicine, Harvard Medical School, Boston, United States
| | - Jamey D Young
- Departments of Chemical & Biomolecular Engineering and Molecular Physiology & Biophysics, Vanderbilt University, Nashville, United States
| | - Joseph Loscalzo
- Department of Medicine, Brigham and Women's Hospital, Boston, United States
- Department of Medicine, Harvard Medical School, Boston, United States
| | - William M Oldham
- Department of Medicine, Brigham and Women's Hospital, Boston, United States
- Department of Medicine, Harvard Medical School, Boston, United States
| |
Collapse
|
43
|
Pathmanapan S, Poon R, De Renshaw TB, Nadesan P, Nakagawa M, Seesankar GA, Ho Loe AK, Zhang HH, Guinovart JJ, Duran J, Newgard CB, Wunder JS, Alman BA. Mutant IDH regulates glycogen metabolism from early cartilage development to malignant chondrosarcoma formation. Cell Rep 2023; 42:112578. [PMID: 37267108 PMCID: PMC10592452 DOI: 10.1016/j.celrep.2023.112578] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2022] [Revised: 01/22/2023] [Accepted: 05/16/2023] [Indexed: 06/04/2023] Open
Abstract
Chondrosarcomas are the most common malignancy of cartilage and are associated with somatic mutations in isocitrate dehydrogenase 1 (IDH1) and IDH2 genes. Somatic IDH mutations are also found in its benign precursor lesion, enchondromas, suggesting that IDH mutations are early events in malignant transformation. Human mutant IDH chondrosarcomas and mutant Idh mice that develop enchondromas investigated in our studies display glycogen deposition exclusively in mutant cells from IDH mutant chondrosarcomas and Idh1 mutant murine growth plates. Pharmacologic blockade of glycogen utilization induces changes in tumor cell behavior, downstream energetic pathways, and tumor burden in vitro and in vivo. Mutant IDH1 interacts with hypoxia-inducible factor 1α (HIF1α) to regulate expression of key enzymes in glycogen metabolism. Here, we show a critical role for glycogen in enchondromas and chondrosarcomas, which is likely mediated through an interaction with mutant IDH1 and HIF1α.
Collapse
Affiliation(s)
- Sinthu Pathmanapan
- Developmental and Stem Cell Biology, Hospital for Sick Children, Toronto, ON, Canada; Institute of Medical Science, University of Toronto, Toronto, ON, Canada
| | - Raymond Poon
- Developmental and Stem Cell Biology, Hospital for Sick Children, Toronto, ON, Canada
| | | | | | - Makoto Nakagawa
- Department of Orthopaedic Surgery, Duke University, Durham, NC, USA
| | - Gireesh A Seesankar
- Developmental and Stem Cell Biology, Hospital for Sick Children, Toronto, ON, Canada
| | - Adrian Kwan Ho Loe
- Developmental and Stem Cell Biology, Hospital for Sick Children, Toronto, ON, Canada
| | - Hongyuan H Zhang
- Department of Orthopaedic Surgery, Duke University, Durham, NC, USA
| | - Joan J Guinovart
- Institute for Research in Biomedicine (IRB Barcelona) Barcelona, Barcelona, Spain
| | - Jordi Duran
- Institute for Research in Biomedicine (IRB Barcelona) Barcelona, Barcelona, Spain
| | - Christopher B Newgard
- Department of Pharmacology & Cancer Biology, Duke University, Durham, NC, USA; Sarah W. Stedman Nutrition and Metabolism Center and Duke Molecular Physiology Institute, Duke University Medical Center, Durham, NC, USA
| | - Jay S Wunder
- Lunenfeld-Tanenbaum Research Institute and the University Musculoskeletal Oncology Unit, Mount Sinai Hospital, Toronto, ON, Canada
| | - Benjamin A Alman
- Department of Orthopaedic Surgery, Duke University, Durham, NC, USA.
| |
Collapse
|
44
|
de Heer EC, Zois CE, Bridges E, van der Vegt B, Sheldon H, Veldman WA, Zwager MC, van der Sluis T, Haider S, Morita T, Baba O, Schröder CP, de Jong S, Harris AL, Jalving M. Glycogen synthase 1 targeting reveals a metabolic vulnerability in triple-negative breast cancer. J Exp Clin Cancer Res 2023; 42:143. [PMID: 37280675 PMCID: PMC10242793 DOI: 10.1186/s13046-023-02715-z] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2022] [Accepted: 05/18/2023] [Indexed: 06/08/2023] Open
Abstract
BACKGROUND Hypoxia-induced glycogen turnover is implicated in cancer proliferation and therapy resistance. Triple-negative breast cancers (TNBCs), characterized by a hypoxic tumor microenvironment, respond poorly to therapy. We studied the expression of glycogen synthase 1 (GYS1), the key regulator of glycogenesis, and other glycogen-related enzymes in primary tumors of patients with breast cancer and evaluated the impact of GYS1 downregulation in preclinical models. METHODS mRNA expression of GYS1 and other glycogen-related enzymes in primary breast tumors and the correlation with patient survival were studied in the METABRIC dataset (n = 1904). Immunohistochemical staining of GYS1 and glycogen was performed on a tissue microarray of primary breast cancers (n = 337). In four breast cancer cell lines and a mouse xenograft model of triple-negative breast cancer, GYS1 was downregulated using small-interfering or stably expressed short-hairpin RNAs to study the effect of downregulation on breast cancer cell proliferation, glycogen content and sensitivity to various metabolically targeted drugs. RESULTS High GYS1 mRNA expression was associated with poor patient overall survival (HR 1.20, P = 0.009), especially in the TNBC subgroup (HR 1.52, P = 0.014). Immunohistochemical GYS1 expression in primary breast tumors was highest in TNBCs (median H-score 80, IQR 53-121) and other Ki67-high tumors (median H-score 85, IQR 57-124) (P < 0.0001). Knockdown of GYS1 impaired proliferation of breast cancer cells, depleted glycogen stores and delayed growth of MDA-MB-231 xenografts. Knockdown of GYS1 made breast cancer cells more vulnerable to inhibition of mitochondrial proteostasis. CONCLUSIONS Our findings highlight GYS1 as potential therapeutic target in breast cancer, especially in TNBC and other highly proliferative subsets.
Collapse
Affiliation(s)
- E C de Heer
- Department of Medical Oncology, University of Groningen, University Medical Center Groningen, PO Box 30.001, 9700 RB, Groningen, The Netherlands
| | - C E Zois
- Department of Oncology, Weatherall Institute of Molecular Medicine, University of Oxford, Hypoxia and Angiogenesis Group, Cancer Research UK Molecular Oncology Laboratories, Oxford, OX3 9DS, UK.
- Department of Radiotherapy and Oncology, School of Health, Democritus University of Thrace, Alexandroupolis, Greece.
- Department of Oncology, MRC Weatherall Institute of Molecular Medicine, John Radcliffe Hospital, Molecular Oncology Laboratories, Oxford University, Oxford, OX3 9DS, UK.
| | - E Bridges
- Department of Oncology, Weatherall Institute of Molecular Medicine, University of Oxford, Hypoxia and Angiogenesis Group, Cancer Research UK Molecular Oncology Laboratories, Oxford, OX3 9DS, UK
| | - B van der Vegt
- Department of Pathology and Medical Biology, University of Groningen, University Medical Center Groningen, Groningen, the Netherlands
| | - H Sheldon
- Department of Oncology, Weatherall Institute of Molecular Medicine, University of Oxford, Hypoxia and Angiogenesis Group, Cancer Research UK Molecular Oncology Laboratories, Oxford, OX3 9DS, UK
| | - W A Veldman
- Department of Medical Oncology, University of Groningen, University Medical Center Groningen, PO Box 30.001, 9700 RB, Groningen, The Netherlands
| | - M C Zwager
- Department of Pathology and Medical Biology, University of Groningen, University Medical Center Groningen, Groningen, the Netherlands
| | - T van der Sluis
- Department of Pathology and Medical Biology, University of Groningen, University Medical Center Groningen, Groningen, the Netherlands
| | - S Haider
- The Breast Cancer Now Toby Robins Research Centre, The Institute of Cancer Research, London, UK
| | - T Morita
- Tokushima University Graduate School, 3-18-15, Kuramoto-Cho, Tokushima, 770-8504, Japan
| | - O Baba
- Tokushima University Graduate School, 3-18-15, Kuramoto-Cho, Tokushima, 770-8504, Japan
| | - C P Schröder
- Department of Medical Oncology, University of Groningen, University Medical Center Groningen, PO Box 30.001, 9700 RB, Groningen, The Netherlands
- Department of Medical Oncology, Antoni Van Leeuwenhoek-Netherlands Cancer Institute, Amsterdam, the Netherlands
| | - S de Jong
- Department of Medical Oncology, University of Groningen, University Medical Center Groningen, PO Box 30.001, 9700 RB, Groningen, The Netherlands
| | - A L Harris
- Department of Oncology, Weatherall Institute of Molecular Medicine, University of Oxford, Hypoxia and Angiogenesis Group, Cancer Research UK Molecular Oncology Laboratories, Oxford, OX3 9DS, UK
| | - M Jalving
- Department of Medical Oncology, University of Groningen, University Medical Center Groningen, PO Box 30.001, 9700 RB, Groningen, The Netherlands.
| |
Collapse
|
45
|
El Shami M, Savani MR, Gattie LC, Smith B, Hicks WH, Rich JN, Richardson TE, McBrayer SK, Abdullah KG. Human plasma-like medium facilitates metabolic tracing and enables upregulation of immune signaling pathways in glioblastoma explants. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.05.29.542774. [PMID: 37398280 PMCID: PMC10312566 DOI: 10.1101/2023.05.29.542774] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/04/2023]
Abstract
Purpose Metabolism within the tumor microenvironment (TME) represents an increasing area of interest to understand glioma initiation and progression. Stable isotope tracing is a technique critical to the study of tumor metabolism. Cell culture models of this disease are not routinely cultured under physiologically relevant nutrient conditions and do not retain cellular heterogeneity present in the parental TME. Moreover, in vivo, stable isotope tracing in intracranial glioma xenografts, the gold standard for metabolic investigation, is time consuming and technically challenging. To provide insights into glioma metabolism in the presence of an intact TME, we performed stable isotope tracing analysis of patient-derived, heterocellular Surgically eXplanted Organoid (SXO) glioma models in human plasma-like medium (HPLM). Methods Glioma SXOs were established and cultured in conventional media or transitioned to HPLM. We evaluated SXO cytoarchitecture and histology, then performed spatial transcriptomic profiling to identify cellular populations and differential gene expression patterns. We performed stable isotope tracing with 15N2-glutamine to evaluate intracellular metabolite labeling patterns. Results Glioma SXOs cultured in HPLM retain cytoarchitecture and cellular constituents. Immune cells in HPLM-cultured SXOs demonstrated increased transcription of immune-related signatures, including innate immune, adaptive immune, and cytokine signaling programs. 15N isotope enrichment from glutamine was observed in metabolites from diverse pathways, and labeling patterns were stable over time. Conclusion To enable ex vivo, tractable investigations of whole tumor metabolism, we developed an approach to conduct stable isotope tracing in glioma SXOs cultured under physiologically relevant nutrient conditions. Under these conditions, SXOs maintained viability, composition, and metabolic activity while exhibiting increased immune-related transcriptional programs.
Collapse
Affiliation(s)
- Mohamad El Shami
- Department of Neurosurgery, University of Pittsburgh School of Medicine, 200 Lothrop St, Pittsburgh, PA, 15213, USA
- Hillman Comprehensive Cancer Center, University of Pittsburgh Medical Center, 5115 Centre Ave, Pittsburgh, PA, 15232, USA
| | - Milan R Savani
- Children’s Medical Center Research Institute, University of Texas Southwestern Medical Center, 6000 Harry Hines Blvd, Dallas, TX, 75235, USA
| | - Lauren C Gattie
- Department of Neurosurgery, University of Pittsburgh School of Medicine, 200 Lothrop St, Pittsburgh, PA, 15213, USA
- Hillman Comprehensive Cancer Center, University of Pittsburgh Medical Center, 5115 Centre Ave, Pittsburgh, PA, 15232, USA
| | - Bailey Smith
- Children’s Medical Center Research Institute, University of Texas Southwestern Medical Center, 6000 Harry Hines Blvd, Dallas, TX, 75235, USA
| | - William H Hicks
- Department of Neurosurgery, University of Pittsburgh School of Medicine, 200 Lothrop St, Pittsburgh, PA, 15213, USA
- Hillman Comprehensive Cancer Center, University of Pittsburgh Medical Center, 5115 Centre Ave, Pittsburgh, PA, 15232, USA
| | - Jeremy N Rich
- Hillman Comprehensive Cancer Center, University of Pittsburgh Medical Center, 5115 Centre Ave, Pittsburgh, PA, 15232, USA
| | - Timothy E Richardson
- Department of Pathology, Molecular, and Cell-Based Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
| | - Samuel K McBrayer
- Children’s Medical Center Research Institute, University of Texas Southwestern Medical Center, 6000 Harry Hines Blvd, Dallas, TX, 75235, USA
- Simmons Comprehensive Cancer Center, University of Texas Southwestern Medical Center, Dallas, TX, 75390, USA
| | - Kalil G Abdullah
- Department of Neurosurgery, University of Pittsburgh School of Medicine, 200 Lothrop St, Pittsburgh, PA, 15213, USA
- Hillman Comprehensive Cancer Center, University of Pittsburgh Medical Center, 5115 Centre Ave, Pittsburgh, PA, 15232, USA
| |
Collapse
|
46
|
Koufaris C, Papandreou ME, Ellis JK, Nicolaidou V, Keun HC. miR-22-enriched breast cancer cells display repressed glycolytic metabolism, increased glycogen synthesis, and reduced survival in low glucose conditions. Mol Biol Rep 2023; 50:5185-5193. [PMID: 37119413 DOI: 10.1007/s11033-023-08458-6] [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: 02/15/2023] [Accepted: 04/12/2023] [Indexed: 05/01/2023]
Abstract
BACKGROUND Breast cancer (BC) is the second leading cause of cancer-related mortality among women. Beyond the established tumourigenic role of genetic mutations, metabolic reprogramming is another key cancer hallmark. Glucose metabolism in particular is known to be prominently altered in tumours, in order to support biomass accumulation and cancer cell survival. The tumor suppressor microRNA (miRNA) miR-22 has been previously associated with a plethora of BC phenotypes such as growth, invasion-metastasis, and regulation of metabolic phenotypes such as lipid and folate metabolism. In this study, we aimed to investigate the role of miR-22 in the regulation of glucose metabolism in BC cells. METHODS AND RESULTS Here we examined how miR-22 affects glucose metabolism in the MCF-7 BC cells. We found that over-expression of miR-22 caused a reduced glycolytic rate in these cells. Moreover, the miRNA also rendered MCF-7 cells more sensitive to lower glucose levels. We next unbiasedly screened the transcript levels of 84 genes relevant to glucose metabolism using the Human Glucose RT2 Profiler PCR Array. Interestingly, the strongest effect identified by this screen was the upregulation of genes involved in glycogen synthesis and the repression of gene involved in glycogen catabolism. Examination of publicly available transcriptomic datasets confirmed the correlations between expression of miR-22 and these glycogen metabolism genes in BC cells. CONCLUSION This study has generated evidence for a regulatory role of miR-22 in glucose and glycogen metabolism, expanding the involvement of this miRNA in BC metabolic reprogramming.
Collapse
Affiliation(s)
- Costas Koufaris
- Department of Biological Sciences, University of Cyprus, Nicosia, Cyprus.
| | | | - James K Ellis
- Department of Surgery & Cancer, Imperial College London, London, UK
| | - Vicky Nicolaidou
- Department of Life Sciences, University of Nicosia, Nicosia, Cyprus
- Non-Coding RNA Research Laboratory, School of Life and Health Sciences, University of Nicosia, Nicosia, Cyprus
| | - Hector C Keun
- Department of Surgery & Cancer, Imperial College London, London, UK
| |
Collapse
|
47
|
Kun S, Mathomes RT, Docsa T, Somsák L, Hayes JM. Design and Synthesis of 3-(β-d-Glucopyranosyl)-4-amino/4-guanidino Pyrazole Derivatives and Analysis of Their Glycogen Phosphorylase Inhibitory Potential. Molecules 2023; 28:3005. [PMID: 37049768 PMCID: PMC10095824 DOI: 10.3390/molecules28073005] [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: 02/27/2023] [Revised: 03/22/2023] [Accepted: 03/23/2023] [Indexed: 03/30/2023] Open
Abstract
Glycogen phosphorylase (GP) is a key regulator of glucose levels and, with that, an important target for the discovery of novel treatments against type 2 diabetes. β-d-Glucopyranosyl derivatives have provided some of the most potent GP inhibitors discovered to date. In this regard, C-β-d-glucopyranosyl azole type inhibitors proved to be particularly effective, with 2- and 4-β-d-glucopyranosyl imidazoles among the most potent designed to date. His377 backbone C=O hydrogen bonding and ion-ion interactions of the protonated imidazole with Asp283 from the 280s loop, stabilizing the inactive state, were proposed as crucial to the observed potencies. Towards further exploring these features, 4-amino-3-(β-d-glucopyranosyl)-5-phenyl-1H-pyrazole (3) and 3-(β-d-glucopyranosyl)-4-guanidino-5-phenyl-1H-pyrazole (4) were designed and synthesized with the potential to exploit similar interactions. Binding assay experiments against rabbit muscle GPb revealed 3 as a moderate inhibitor (IC50 = 565 µM), but 4 displayed no inhibition at 625 µM concentration. Towards understanding the observed inhibitions, docking and post-docking molecular mechanics-generalized Born surface area (MM-GBSA) binding free energy calculations were performed, together with Monte Carlo and density functional theory (DFT) calculations on the free unbound ligands. The computations revealed that while 3 was predicted to hydrogen bond with His377 C=O in its favoured tautomeric state, the interactions with Asp283 were not direct and there were no ion-ion interactions; for 4, the most stable tautomer did not have the His377 backbone C=O interaction and while ion-ion interactions and direct hydrogen bonding with Asp283 were predicted, the conformational strain and entropy loss of the ligand in the bound state was significant. The importance of consideration of tautomeric states and ligand strain for glucose analogues in the confined space of the catalytic site with the 280s loop in the closed position was highlighted.
Collapse
Affiliation(s)
- Sándor Kun
- Department of Organic Chemistry, University of Debrecen, P.O. Box 400, H-4002 Debrecen, Hungary
| | - Rachel T. Mathomes
- School of Pharmacy & Biomedical Sciences, University of Central Lancashire, Preston PR1 2HE, UK
| | - Tibor Docsa
- Department of Medical Chemistry, Faculty of Medicine, University of Debrecen, 4032 Debrecen, Hungary
| | - László Somsák
- Department of Organic Chemistry, University of Debrecen, P.O. Box 400, H-4002 Debrecen, Hungary
| | - Joseph M. Hayes
- School of Pharmacy & Biomedical Sciences, University of Central Lancashire, Preston PR1 2HE, UK
| |
Collapse
|
48
|
Replacing Fish Meal with Hydrolyzed Collagen Derived from Fish By-Products Improved Muscle Quality and Glycolipid Metabolism of Triploid Crucian Carp. Foods 2023; 12:foods12061235. [PMID: 36981161 PMCID: PMC10048121 DOI: 10.3390/foods12061235] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2023] [Revised: 03/07/2023] [Accepted: 03/12/2023] [Indexed: 03/17/2023] Open
Abstract
Fish by-products are rich in collagen. Hydrolyzed collagen derived from fish by-products was used to replace fish meal to evaluate the effects on muscle quality and glycolipid metabolism of juvenile triploid crucian carp. A total of 240 juvenile fish with body weight of 10.01 ± 0.02 g were divided into four groups and fed four diets for 66 days: fish meal (FM) replaced with hydrolyzed collagen (HC) in 0% (Control), 2% (2% HC), 4% (4% HC), and 6% (6% HC), respectively. The results were as follows: The increased proportion of fish meal replaced with hydrolyzed collagen linearly and quadratically decreased the specific growth rate (SGR) of triploid crucian carp (p < 0.05). Compared with the control group, the SGR and intestinal α-amylase, trypsin and lipase activities in the 4% and 6% HC groups significantly decreased (p < 0.05), while there was no significant difference between the control and 2% HC groups (p > 0.05). Total umami amino acids content, chewiness and myofiber density of muscle in the 4% and 6% HC groups, as well as the essential fatty acids content in all HC groups increased significantly (p < 0.05). All HC groups significantly increased the serum glutathione peroxidase (GSH-Px) activity and decreased the serum malondialdehyde (MDA) content (p < 0.05). When the replacement amount reached 4%, the serum glucose and liver glycogen content, the liver and serum triglyceride (TG) content, and serum total cholesterol (T-CHO) content were significantly reduced (p < 0.05). In addition, the expression levels of insulin-like growth factor-1 (IGF-1) of the liver in all HC groups and lipolysis-related genes (lipoprotein lipase (LPL), carnitine O-palmitoyltransferase 1 (CPT 1) and hydroxyacyl-coenzyme A dehydrogenase (HADH)) of the liver in the 6% of HC group increased significantly (p < 0.05), and the expression levels of lipogenesis-related genes (fatty acid synthase (FAS) and sterol regulatory element-binding protein 1 (SREBP 1)) of the liver in the 4% HC and 6% HC groups decreased significantly (p < 0.05). In conclusion, the replacement of 2% fish meal with hydrolyzed collagen had no negative effects on the growth of triploid crucian carp, while the replacement of 4% fish meal with hydrolyzed collagen decreased SGR, but improved the muscle quality and decreased glycolipid levels. The maximum proportion of hydrolyzed collagen replacing fish meal should not exceed 4%.
Collapse
|
49
|
Anand S, Khan MA, Zubair H, Sudan SK, Vikramdeo KS, Deshmukh SK, Azim S, Srivastava SK, Singh S, Singh AP. MYB sustains hypoxic survival of pancreatic cancer cells by facilitating metabolic reprogramming. EMBO Rep 2023; 24:e55643. [PMID: 36592158 PMCID: PMC9986821 DOI: 10.15252/embr.202255643] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2022] [Revised: 12/13/2022] [Accepted: 12/19/2022] [Indexed: 01/03/2023] Open
Abstract
Extensive desmoplasia and poor vasculature renders pancreatic tumors severely hypoxic, contributing to their aggressiveness and therapy resistance. Here, we identify the HuR/MYB/HIF1α axis as a critical regulator of the metabolic plasticity and hypoxic survival of pancreatic cancer cells. HuR undergoes nuclear-to-cytoplasmic translocation under hypoxia and stabilizes MYB transcripts, while MYB transcriptionally upregulates HIF1α. Upon MYB silencing, pancreatic cancer cells fail to survive and adapt metabolically under hypoxia, despite forced overexpression of HIF1α. MYB induces the transcription of several HIF1α-regulated glycolytic genes by directly binding to their promoters, thus enhancing the recruitment of HIF1α to hypoxia-responsive elements through its interaction with p300-dependent histone acetylation. MYB-depleted pancreatic cancer cells exhibit a dramatic reduction in tumorigenic ability, glucose-uptake and metabolism in orthotopic mouse model, even after HIF1α restoration. Together, our findings reveal an essential role of MYB in metabolic reprogramming that supports pancreatic cancer cell survival under hypoxia.
Collapse
Affiliation(s)
- Shashi Anand
- Department of Pathology, College of Medicine, University of South Alabama, Mobile, AL, USA
- Cancer Biology Program, Mitchell Cancer Institute, University of South Alabama, Mobile, AL, USA
| | - Mohammad Aslam Khan
- Department of Pathology, College of Medicine, University of South Alabama, Mobile, AL, USA
- Cancer Biology Program, Mitchell Cancer Institute, University of South Alabama, Mobile, AL, USA
| | - Haseeb Zubair
- Department of Pathology, College of Medicine, University of South Alabama, Mobile, AL, USA
- Cancer Biology Program, Mitchell Cancer Institute, University of South Alabama, Mobile, AL, USA
| | - Sarabjeet Kour Sudan
- Department of Pathology, College of Medicine, University of South Alabama, Mobile, AL, USA
- Cancer Biology Program, Mitchell Cancer Institute, University of South Alabama, Mobile, AL, USA
| | - Kunwar Somesh Vikramdeo
- Department of Pathology, College of Medicine, University of South Alabama, Mobile, AL, USA
- Cancer Biology Program, Mitchell Cancer Institute, University of South Alabama, Mobile, AL, USA
| | - Sachin Kumar Deshmukh
- Department of Pathology, College of Medicine, University of South Alabama, Mobile, AL, USA
- Cancer Biology Program, Mitchell Cancer Institute, University of South Alabama, Mobile, AL, USA
| | - Shafquat Azim
- Department of Pathology, College of Medicine, University of South Alabama, Mobile, AL, USA
- Cancer Biology Program, Mitchell Cancer Institute, University of South Alabama, Mobile, AL, USA
| | - Sanjeev Kumar Srivastava
- Department of Pathology, College of Medicine, University of South Alabama, Mobile, AL, USA
- Cancer Biology Program, Mitchell Cancer Institute, University of South Alabama, Mobile, AL, USA
| | - Seema Singh
- Department of Pathology, College of Medicine, University of South Alabama, Mobile, AL, USA
- Cancer Biology Program, Mitchell Cancer Institute, University of South Alabama, Mobile, AL, USA
- Department of Biochemistry and Molecular Biology, College of Medicine, University of South Alabama, Mobile, AL, USA
| | - Ajay Pratap Singh
- Department of Pathology, College of Medicine, University of South Alabama, Mobile, AL, USA
- Cancer Biology Program, Mitchell Cancer Institute, University of South Alabama, Mobile, AL, USA
- Department of Biochemistry and Molecular Biology, College of Medicine, University of South Alabama, Mobile, AL, USA
| |
Collapse
|
50
|
Jung J, Gokhale S, Xie P. TRAF3: A novel regulator of mitochondrial physiology and metabolic pathways in B lymphocytes. Front Oncol 2023; 13:1081253. [PMID: 36776285 PMCID: PMC9911533 DOI: 10.3389/fonc.2023.1081253] [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: 10/27/2022] [Accepted: 01/13/2023] [Indexed: 01/28/2023] Open
Abstract
Mitochondria, the organelle critical for cell survival and metabolism, are exploited by cancer cells and provide an important therapeutic target in cancers. Mitochondria dynamically undergo fission and fusion to maintain their diverse functions. Proteins controlling mitochondrial fission and fusion have been recognized as essential regulators of mitochondrial functions, mitochondrial quality control, and cell survival. In a recent proteomic study, we identified the key mitochondrial fission factor, MFF, as a new interacting protein of TRAF3, a known tumor suppressor of multiple myeloma and other B cell malignancies. This interaction recruits the majority of cytoplasmic TRAF3 to mitochondria, allowing TRAF3 to regulate mitochondrial morphology, mitochondrial functions, and mitochondria-dependent apoptosis in resting B lymphocytes. Interestingly, recent transcriptomic, metabolic and lipidomic studies have revealed that TRAF3 also vitally regulates multiple metabolic pathways in B cells, including phospholipid metabolism, glucose metabolism, and ribonucleotide metabolism. Thus, TRAF3 emerges as a novel regulator of mitochondrial physiology and metabolic pathways in B lymphocytes and B cell malignancies. Here we review current knowledge in this area and discuss relevant clinical implications.
Collapse
Affiliation(s)
- Jaeyong Jung
- Department of Cell Biology and Neuroscience, Rutgers University, Piscataway, NJ, United States.,Graduate Program in Cellular and Molecular Pharmacology, Rutgers University, Piscataway, NJ, United States
| | - Samantha Gokhale
- Department of Cell Biology and Neuroscience, Rutgers University, Piscataway, NJ, United States.,Graduate Program in Cellular and Molecular Pharmacology, Rutgers University, Piscataway, NJ, United States
| | - Ping Xie
- Department of Cell Biology and Neuroscience, Rutgers University, Piscataway, NJ, United States.,Rutgers Cancer Institute of New Jersey, New Brunswick, NJ, United States
| |
Collapse
|