1
|
Yu X, Yang T, Wu D, Xu C, Li Z, Sun A, Gao S, Li H, Fan Z, Huang R. PARP14 inhibits microglial activation via NNT to alleviate depressive-like behaviors in mice. Brain Behav Immun 2025; 126:235-246. [PMID: 39978699 DOI: 10.1016/j.bbi.2025.02.017] [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: 10/20/2024] [Revised: 02/03/2025] [Accepted: 02/17/2025] [Indexed: 02/22/2025] Open
Abstract
Microglial inflammation has been implicated in the pathophysiology of major depressive disorder; however, the underlying biological mechanisms remain inadequately understood. Consequently, we conducted a screening of the Poly ADP-ribose (PAR) polymerase (PARP) family expression in the hippocampus of chronic unpredictable stress (CUS) mouse models and investigated the specific role of PARP14 in microglial inflammation and its association with depression. Here, this study demonstrated the elevated PARP14 expression in the hippocampus of CUS mice. The knockdown of PARP14 in the hippocampus did not mitigate depressive-like behaviors in mice, whereas overexpression of PARP14 significantly mitigated these behaviors. Furthermore, PARP14 was abundant in microglia, and microglial-targeted PARP14 overexpression significantly alleviated depressive-behaviors in CUS, reduced microglial activation, and inhibited the central inflammatory responses. Mechanistically, PARP14 positively regulated nicotinamide nucleotide transhydrogenase (NNT) expression in microglia, and the inflammatory response of microglia induced by PARP14 knockdown was suppressed through NNT overexpression. Additionally, deficiency in NNT led to an accumulation of reactive oxygen species (ROS) and subsequent microglial inflammation, which was effectively inhibited by the ROS inhibitor N-Acetylcysteine. These findings suggest that PARP14 alleviates depressive-like behaviors in mice by inhibiting microglial activation via NTT-mediated clearance of ROS.
Collapse
Affiliation(s)
- Xiaoyu Yu
- Wuxi School of Medicine, Jiangnan University, Wuxi 214122, China
| | - Tingting Yang
- Wuxi School of Medicine, Jiangnan University, Wuxi 214122, China
| | - Di Wu
- Zhongda Hospital, Medical School, Southeast University, Nanjing 210009, China
| | - Chenxue Xu
- Wuxi School of Medicine, Jiangnan University, Wuxi 214122, China
| | - Zhuoran Li
- Wuxi School of Medicine, Jiangnan University, Wuxi 214122, China
| | - Ao Sun
- Wuxi School of Medicine, Jiangnan University, Wuxi 214122, China
| | - Shulei Gao
- Wuxi School of Medicine, Jiangnan University, Wuxi 214122, China
| | - Heng Li
- Wuxi School of Medicine, Jiangnan University, Wuxi 214122, China
| | - Zhenyu Fan
- Deparment of Pharmacy, Affiliated Hospital of Nantong University, Nantong 226001, China.
| | - Rongrong Huang
- Deparment of Pharmacy, Affiliated Hospital of Nantong University, Nantong 226001, China.
| |
Collapse
|
2
|
Wang K, Li X, Guo S, Chen J, Lv Y, Guo Z, Liu H. Metabolic reprogramming of glucose: the metabolic basis for the occurrence and development of hepatocellular carcinoma. Front Oncol 2025; 15:1545086. [PMID: 39980550 PMCID: PMC11839411 DOI: 10.3389/fonc.2025.1545086] [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: 12/14/2024] [Accepted: 01/20/2025] [Indexed: 02/22/2025] Open
Abstract
Primary liver cancer is a common malignant tumor of the digestive system, with hepatocellular carcinoma (HCC) being the most prevalent type. It is characterized by high malignancy, insidious onset, and a lack of specific early diagnostic and therapeutic markers, posing a serious threat to human health. The occurrence and development of HCC are closely related to its metabolic processes. Similar to other malignant tumors, metabolic reprogramming occurs extensively in tumor cells, with glucose metabolism reprogramming being particularly prominent. This is characterized by abnormal activation of glycolysis and inhibition of oxidative phosphorylation and gluconeogenesis, among other changes. Glucose metabolism reprogramming provides intermediates and energy for HCC to meet its demands for rapid growth, proliferation, and metastasis. Additionally, various enzymes and signaling molecules involved in glucose metabolism reprogramming play irreplaceable roles. Therefore, regulating key metabolic enzymes and pathways in these processes is considered an important target for the diagnosis and treatment of HCC. This paper reviews the current status and progress of glucose metabolism reprogramming in HCC, aiming to provide new insights for the diagnosis, detection, and comprehensive treatment strategies of HCC involving combined glucose metabolism intervention in clinical settings.
Collapse
Affiliation(s)
- Kai Wang
- Department of Colorectal Surgery, Heping Hospital Affiliated to Changzhi Medical College, Changzhi, Shanxi, China
| | - Xiaodan Li
- Department of Pediatric Health Care, Zhangzi County Maternal and Child Health Family Planning Service Center, Changzhi, Shanxi, China
| | - Shuwei Guo
- Department of Colorectal Surgery, Heping Hospital Affiliated to Changzhi Medical College, Changzhi, Shanxi, China
| | - Junsheng Chen
- Department of Colorectal Surgery, Heping Hospital Affiliated to Changzhi Medical College, Changzhi, Shanxi, China
| | - Yandong Lv
- Department of Colorectal Surgery, Heping Hospital Affiliated to Changzhi Medical College, Changzhi, Shanxi, China
| | - Zhiqiang Guo
- Department of Colorectal Surgery, Heping Hospital Affiliated to Changzhi Medical College, Changzhi, Shanxi, China
| | - Hongzhou Liu
- Department of Colorectal Surgery, Heping Hospital Affiliated to Changzhi Medical College, Changzhi, Shanxi, China
| |
Collapse
|
3
|
López-Cánovas JL, Naranjo-Martínez B, Diaz-Ruiz A. Fasting in combination with the cocktail Sorafenib:Metformin blunts cellular plasticity and promotes liver cancer cell death via poly-metabolic exhaustion. Cell Oncol (Dordr) 2025; 48:161-182. [PMID: 38990489 PMCID: PMC11850423 DOI: 10.1007/s13402-024-00966-2] [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] [Accepted: 05/31/2024] [Indexed: 07/12/2024] Open
Abstract
PURPOSE Dual-Interventions targeting glucose and oxidative metabolism are receiving increasing attention in cancer therapy. Sorafenib (S) and Metformin (M), two gold-standards in liver cancer, are known for their mitochondrial inhibitory capacity. Fasting, a glucose-limiting strategy, is also emerging as chemotherapy adjuvant. Herein, we explore the anti-carcinogenic response of nutrient restriction in combination with sorafenib:metformin (NR-S:M). RESULTS Our data demonstrates that, independently of liver cancer aggressiveness, fasting synergistically boosts the anti-proliferative effects of S:M co-treatment. Metabolic and Cellular plasticity was determined by the examination of mitochondrial and glycolytic activity, cell cycle modulation, activation of cellular apoptosis, and regulation of key signaling and metabolic enzymes. Under NR-S:M conditions, early apoptotic events and the pro-apoptotic Bcl-xS/Bcl-xL ratio were found increased. NR-S:M induced the highest retention in cellular SubG1 phase, consistent with the presence of DNA fragments from cellular apoptosis. Mitochondrial functionality, Mitochondrial ATP-linked respiration, Maximal respiration and Spare respiratory capacity, were all found blunted under NR-S:M conditions. Basal Glycolysis, Glycolytic reserve, and glycolytic capacity, together with the expression of glycogenic (PKM), gluconeogenic (PCK1 and G6PC3), and glycogenolytic enzymes (PYGL, PGM1, and G6PC3), were also negatively impacted by NR-S:M. Lastly, a TMT-proteomic approach corroborated the synchronization of liver cancer metabolic reprogramming with the activation of molecular pathways to drive a quiescent-like status of energetic-collapse and cellular death. CONCLUSION Altogether, we show that the energy-based polytherapy NR-S:M blunts cellular, metabolic and molecular plasticity of liver cancer. Notwithstanding the in vitro design of this study, it holds a promising therapeutic tool worthy of exploration for this tumor pathology.
Collapse
Affiliation(s)
- Juan L López-Cánovas
- Laboratory of Cellular and Molecular Gerontology, Precision Nutrition and Aging Program, Institute IMDEA Food (CEI UAM+CSIC), Crta. de Canto Blanco nº 8, Madrid, E-28049, Spain
| | - Beatriz Naranjo-Martínez
- Laboratory of Cellular and Molecular Gerontology, Precision Nutrition and Aging Program, Institute IMDEA Food (CEI UAM+CSIC), Crta. de Canto Blanco nº 8, Madrid, E-28049, Spain
| | - Alberto Diaz-Ruiz
- Laboratory of Cellular and Molecular Gerontology, Precision Nutrition and Aging Program, Institute IMDEA Food (CEI UAM+CSIC), Crta. de Canto Blanco nº 8, Madrid, E-28049, Spain.
- CIBER Pathophysiology of Obesity and Nutrition (CIBERobn), Córdoba, Spain.
| |
Collapse
|
4
|
Luo J, An J, Jia R, Liu C, Zhang Y. Identification and Verification of Metabolism-related Immunotherapy Features and Prognosis in Lung Adenocarcinoma. Curr Med Chem 2025; 32:1423-1441. [PMID: 38500277 DOI: 10.2174/0109298673293414240314043529] [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: 11/30/2023] [Revised: 02/21/2024] [Accepted: 03/04/2024] [Indexed: 03/20/2024]
Abstract
BACKGROUND Lung cancer is a frequent malignancy with a poor prognosis. Extensive metabolic alterations are involved in carcinogenesis and could, therefore, serve as a reliable prognostic phenotype. AIMS Our study aimed to develop a prognosis signature and explore the relationship between metabolic characteristic-related signature and immune infiltration in lung adenocarcinoma (LUAD). OBJECTIVE TCGA-LUAD and GSE31210 datasets were used as a training set and a validation set, respectively. METHODS A total of 513 LUAD samples collected from The Cancer Genome Atlas database (TCGA-LUAD) were used as a training dataset. Molecular subtypes were classified by consensus clustering, and prognostic genes related to metabolism were analyzed based on Differentially Expressed Genes (DEGs), Protein-Protein Interaction (PPI) network, the univariate/multivariate- and Lasso- Cox regression analysis. RESULTS Two molecular subtypes with significant survival differences were divided by the metabolism gene sets. The DEGs between the two subtypes were identified by integrated analysis and then used to develop an 8-gene signature (TTK, TOP2A, KIF15, DLGAP5, PLK1, PTTG1, ECT2, and ANLN) for predicting LUAD prognosis. Overexpression of the 8 genes was significantly correlated with worse prognostic outcomes. RiskScore was an independent factor that could divide LUAD patients into low- and high-risk groups. Specifically, high-risk patients had poorer prognoses and higher immune escape. The Receiver Operating Characteristic (ROC) curve showed strong performance of the RiskScore model in estimating 1-, 3- and 5-year survival in both training and validation sets. Finally, an optimized nomogram model was developed and contributed the most to the prognostic prediction in LUAD. CONCLUSION The current model could help effectively identify high-risk patients and suggest the most effective drug and treatment candidates for patients with LUAD.
Collapse
Affiliation(s)
- Junfang Luo
- Department of Geriatric Respiratory, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, China
| | - Jinlu An
- Department of Geriatric Respiratory, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, China
| | - Rongyan Jia
- Department of Anesthesiology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, China
| | - Cong Liu
- Department of Geriatric Respiratory, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, China
| | - Yang Zhang
- Department of Geriatric Respiratory, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, China
| |
Collapse
|
5
|
Kam CMT, Tauber AL, Zunk MS, McDermott CM, Levonis SM, Schweiker SS. Novel inhibitors of PARP1 and PARP14: design, synthesis, and potentiation of cisplatin efficacy in cancer. Future Med Chem 2025; 17:35-58. [PMID: 39691063 DOI: 10.1080/17568919.2024.2437972] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2024] [Accepted: 11/25/2024] [Indexed: 12/19/2024] Open
Abstract
BACKGROUND Poly(ADP-ribose) polymerase (PARP) is a superfamily of enzymes involved in cell survival. Both PARP1 and PARP14 are overexpressed in malignancies. No clinically approved PARP14 inhibitors are available, and PARP1 inhibitors are generally nonspecific, resulting in a need for a more diverse library of selective PARP1 and PARP14 inhibitors. MATERIALS AND METHODS Based on the previous lead compounds 1 and 2, 26 novel compounds were designed, synthesized, and screened against PARP1 and PARP14. Compounds with the best in vitro inhibitory results were further screened against PARP2, PARP3, PARP5a, PARP7, and PARP15. RESULTS AND CONCLUSION The 26 novel compounds demonstrated a lesser inhibitory effect than the lead compounds. Compounds 1 and 2 were further investigated using in vitro cell viability assays, which revealed that cells treated with either lead PARP inhibitor and cisplatin in combination had significantly lower survival rates than those treated with cisplatin alone. At 10 µM, the combination showed more significant cell survival reduction, suggesting greater inhibition of PARP increases lethality, particularly in HeLa and PC-3 cell lines at 96 h and beyond.
Collapse
Affiliation(s)
- Caleb M T Kam
- Medicinal Chemistry Group, Faculty of Health Sciences and Medicine, Bond University, Robina, Queensland, Australia
| | - Amanda L Tauber
- Medicinal Chemistry Group, Faculty of Health Sciences and Medicine, Bond University, Robina, Queensland, Australia
| | - Matthew S Zunk
- School of Pharmacy and Medical Sciences, Griffith University, Southport, Queensland, Australia
| | - Catherine M McDermott
- Centre for Urology Research, Faculty of Health Sciences and Medicine, Bond University, Robina, Queensland, Australia
| | - Stephan M Levonis
- Medicinal Chemistry Group, Faculty of Health Sciences and Medicine, Bond University, Robina, Queensland, Australia
| | - Stephanie S Schweiker
- Medicinal Chemistry Group, Faculty of Health Sciences and Medicine, Bond University, Robina, Queensland, Australia
| |
Collapse
|
6
|
Revert-Ros F, Ventura I, Prieto-Ruiz JA, Giner-Moreno E, Pérez-Cervera Á, Pérez-Rojas J, Revert F, Hernández-Andreu JM. Polyribonucleotide phosphorylase is overexpressed in hepatocellular cancer, promoting epithelial phenotype maintenance and tumor progression. Pathol Res Pract 2024; 264:155713. [PMID: 39522317 DOI: 10.1016/j.prp.2024.155713] [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: 10/02/2024] [Revised: 11/04/2024] [Accepted: 11/05/2024] [Indexed: 11/16/2024]
Abstract
Liver cancer, particularly hepatocellular carcinoma (HCC), is a major global health challenge, largely associated with cirrhosis caused by various factors. Prognosis is often guided by molecular and histological classifications. In this study, expression of Polyribonucleotide Phosphorylase (PNPT1) in HCC was investigated to better understand its role in tumor behavior and patient outcomes. The expression of the corresponding protein PNPase was assessed in HCC tissue samples using immunohistochemistry, while RNA-seq data from The Cancer Genome Atlas (TCGA) were analyzed via OncoDB. Additionally, PNPT1 silencing in HepG2 cells was followed by gene and protein expression analysis. The results revealed that PNPT1 is overexpressed in HCC tumors, particularly in those expressing E-cadherin. Notably, silencing PNPT1 in HepG2 cells triggered a shift towards a mesenchymal phenotype. In HCC tissues, PNPT1 expression was linked to markers of epithelial phenotype, oxidative stress, and poor prognosis, especially in non-viral HCC cases. These findings suggest that PNPase may play a crucial role in the progression of well-differentiated HCC tumors, serving as a poor prognostic biomarker.
Collapse
Affiliation(s)
- Francisco Revert-Ros
- Grupo de Medicina Molecular y Mitocondrial, Facultad de Medicina y Ciencias de la Salud, Universidad Católica de Valencia San Vicente Mártir, C/Quevedo 2, Valencia 46001, Spain
| | - Ignacio Ventura
- Grupo de Medicina Molecular y Mitocondrial, Facultad de Medicina y Ciencias de la Salud, Universidad Católica de Valencia San Vicente Mártir, C/Quevedo 2, Valencia 46001, Spain
| | - Jesús A Prieto-Ruiz
- Grupo de Medicina Molecular y Mitocondrial, Facultad de Medicina y Ciencias de la Salud, Universidad Católica de Valencia San Vicente Mártir, C/Quevedo 2, Valencia 46001, Spain
| | - Eduardo Giner-Moreno
- Department of Pathology, Hospital Universitario y Politécnico La Fe, Valencia, Spain
| | - Ángela Pérez-Cervera
- Grupo de Medicina Molecular y Mitocondrial, Facultad de Medicina y Ciencias de la Salud, Universidad Católica de Valencia San Vicente Mártir, C/Quevedo 2, Valencia 46001, Spain
| | - Judith Pérez-Rojas
- Department of Pathology, Hospital Universitario y Politécnico La Fe, Valencia, Spain
| | - Fernando Revert
- Grupo de Medicina Molecular y Mitocondrial, Facultad de Medicina y Ciencias de la Salud, Universidad Católica de Valencia San Vicente Mártir, C/Quevedo 2, Valencia 46001, Spain.
| | - José Miguel Hernández-Andreu
- Grupo de Medicina Molecular y Mitocondrial, Facultad de Medicina y Ciencias de la Salud, Universidad Católica de Valencia San Vicente Mártir, C/Quevedo 2, Valencia 46001, Spain
| |
Collapse
|
7
|
Khan ZF, Rathi A, Khan A, Anjum F, Chaudhury A, Taiyab A, Shamsi A, Hassan MI. Exploring PDK3 inhibition in lung cancer through drug repurposing for potential therapeutic interventions. Sci Rep 2024; 14:29672. [PMID: 39613779 DOI: 10.1038/s41598-024-78022-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2024] [Accepted: 10/28/2024] [Indexed: 12/01/2024] Open
Abstract
The pyruvate dehydrogenase kinase-3 (PDK3) plays an important role in the regulation of a variety of cancers, including lung, by inhibiting the pyruvate dehydrogenase complex (PDC), shifting energy production towards glycolysis necessary for cancer metabolism. In this study, we aimed to identify potential PDK3 inhibitors using a computer-based drug design approach. Virtual screening of the FDA-approved library of 3839 compounds was carried out, from which Bagrosin and Dehydrocholic acid appeared best due to their strong binding affinity, specific interactions, and potential biological characteristics, and thus were selected for further investigations. Both compounds show strong interactions with functionally important residues of the PDK3 with a binding affinity of - 10.6 and - 10.5 kcal/mol for Bagrosin and Dehydrocholic acid, respectively. MD simulation studies for 100 ns suggest the formation of stable complexes, which is evident from RMSD, RMSF, Rg, and SASA parameters. The PCA and FEL analysis suggested admirable global energy minima for the bagrosin-PDK3 and dehydrocholic acid-PDK3 complexes. Finally, we identified FDA-approved drugs, Bagrosin and Dehydrocholic acid, that offer valuable resources and potential therapeutic molecules for targeting lung cancer. Further clinical investigations are required to validate the clinical utility of selected molecules.
Collapse
Affiliation(s)
- Zeba Firdos Khan
- Department of Biosciences, Faculty of Life Sciences, Jamia Millia Islamia, Jamia Nagar, New Delhi, 110025, India
| | - Aanchal Rathi
- Department of Biotechnology, Faculty of Life Sciences, Jamia Millia Islamia, Jamia Nagar, New Delhi, 110025, India
| | - Afreen Khan
- Centre for Interdisciplinary Research in Basic Sciences, Jamia Millia Islamia, Jamia Nagar, New Delhi, 110025, India
| | - Farah Anjum
- Department of Clinical Laboratory Sciences, College of Applied Medical Sciences, Taif University, PO Box 11099, 21944, Taif, Saudi Arabia
| | - Arunabh Chaudhury
- Centre for Interdisciplinary Research in Basic Sciences, Jamia Millia Islamia, Jamia Nagar, New Delhi, 110025, India
| | - Aaliya Taiyab
- Centre for Interdisciplinary Research in Basic Sciences, Jamia Millia Islamia, Jamia Nagar, New Delhi, 110025, India
| | - Anas Shamsi
- Center of Medical and Bio-Allied Health Sciences Research (CMBHSR), Ajman University, Ajman, United Arab Emirates.
| | - Md Imtaiyaz Hassan
- Centre for Interdisciplinary Research in Basic Sciences, Jamia Millia Islamia, Jamia Nagar, New Delhi, 110025, India.
| |
Collapse
|
8
|
Ming Y, Gong Y, Fu X, Ouyang X, Peng Y, Pu W. Small-molecule-based targeted therapy in liver cancer. Mol Ther 2024; 32:3260-3287. [PMID: 39113358 PMCID: PMC11489561 DOI: 10.1016/j.ymthe.2024.08.001] [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: 11/22/2023] [Revised: 03/13/2024] [Accepted: 08/02/2024] [Indexed: 08/23/2024] Open
Abstract
Liver cancer is one of the most prevalent malignant tumors worldwide. According to the Barcelona Clinic Liver Cancer staging criteria, clinical guidelines provide tutorials to clinical management of liver cancer at their individual stages. However, most patients diagnosed with liver cancer are at advanced stage; therefore, many researchers conduct investigations on targeted therapy, aiming to improve the overall survival of these patients. To date, small-molecule-based targeted therapies are highly recommended (first line: sorafenib and lenvatinib; second line: regorafenib and cabozantinib) by current the clinical guidelines of the American Society of Clinical Oncology, European Society for Medical Oncology, and National Comprehensive Cancer Network. Herein, we summarize the small-molecule-based targeted therapies in liver cancer, including the approved and preclinical therapies as well as the therapies under clinical trials, and introduce their history of discovery, clinical trials, indications, and molecular mechanisms. For drug resistance, the revealed mechanisms of action and the combination therapies are also discussed. In fact, the known small-molecule-based therapies still have limited clinical benefits to liver cancer patients. Therefore, we analyze the current status and give our ideas for the urgent issues and future directions in this field, suggesting clues for novel techniques in liver cancer treatment.
Collapse
Affiliation(s)
- Yue Ming
- Laboratory of Molecular Oncology, Frontiers Science Center for Disease-related Molecular Network, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu 610064, China
| | - Yanqiu Gong
- National Clinical Research Center for Geriatrics and Department of General Practice, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Xuewen Fu
- Jinhua Huanke Environmental Technology Co., Ltd., Jinhua 321000, China
| | - Xinyu Ouyang
- Laboratory of Molecular Oncology, Frontiers Science Center for Disease-related Molecular Network, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu 610064, China; West China School of Medicine, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Yong Peng
- Laboratory of Molecular Oncology, Frontiers Science Center for Disease-related Molecular Network, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu 610064, China; Frontier Medical Center, Tianfu Jincheng Laboratory, Chengdu 610212, China.
| | - Wenchen Pu
- Laboratory of Molecular Oncology, Frontiers Science Center for Disease-related Molecular Network, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu 610064, China; West China School of Medicine, West China Hospital, Sichuan University, Chengdu 610041, China.
| |
Collapse
|
9
|
Chunlian Z, Qi W, Rui Z. The Role of Pyruvate Kinase M2 Posttranslational Modification in the Occurrence and Development of Hepatocellular Carcinoma. Cell Biochem Funct 2024; 42:e4125. [PMID: 39327771 DOI: 10.1002/cbf.4125] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2024] [Revised: 08/09/2024] [Accepted: 09/08/2024] [Indexed: 09/28/2024]
Abstract
Hepatocellular carcinoma (HCC) is one of the deadly malignant tumors that directly leads to the death of nearly one million people worldwide every year, causing a serious burden on society. In the presence of sufficient oxygen, HCC cells rapidly generate energy through aerobic glycolysis, which promotes tumor cell proliferation, immune evasion, metastasis, angiogenesis, and drug resistance. Pyruvate kinase M2 (PKM2) is a key rate-limiting enzyme in glycolysis. In recent years, studies have found that PKM2 not only exerts pyruvate kinase activity in the process of glucose metabolism, but also exerts protein kinase activity in non-metabolic pathways to affect tumor cell processes, and its activity is flexibly regulated by various posttranslational modifications such as acetylation, phosphorylation, lactylation, ubiquitination, SUMOylation, and so forth. This review summarizes the role of posttranslational modifications of PKM2-related sites in the development of HCC.
Collapse
Affiliation(s)
- Zhao Chunlian
- Second Hospital of Lanzhou University, Lanzhou, China
| | - Wan Qi
- Second Hospital of Lanzhou University, Lanzhou, China
| | - Zhao Rui
- Second Hospital of Lanzhou University, Lanzhou, China
| |
Collapse
|
10
|
Ke R, Kumar S, Singh SK, Rana A, Rana B. Molecular insights into the role of mixed lineage kinase 3 in cancer hallmarks. Biochim Biophys Acta Rev Cancer 2024; 1879:189157. [PMID: 39032538 DOI: 10.1016/j.bbcan.2024.189157] [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: 12/22/2023] [Revised: 07/14/2024] [Accepted: 07/17/2024] [Indexed: 07/23/2024]
Abstract
Mixed-lineage kinase 3 (MLK3) is a serine/threonine kinase of the MAPK Kinase kinase (MAP3K) family that plays critical roles in various biological processes, including cancer. Upon activation, MLK3 differentially activates downstream MAPKs, such as JNK, p38, and ERK. In addition, it regulates various non-canonical signaling pathways, such as β-catenin, AMPK, Pin1, and PAK1, to regulate cell proliferation, apoptosis, invasion, and metastasis. Recent studies have also uncovered other potentially diverse roles of MLK3 in malignancy, which include metabolic reprogramming, cancer-associated inflammation, and evasion of cancer-related immune surveillance. The role of MLK3 in cancer is complex and cancer-specific, and an understanding of its function at the molecular level aligned specifically with the cancer hallmarks will have profound therapeutic implications for diagnosing and treating MLK3-dependent cancers. This review summarizes the current knowledge about the effect of MLK3 on the hallmarks of cancer, providing insights into its potential as a promising anticancer drug target.
Collapse
Affiliation(s)
- Rong Ke
- Department of Surgery, Division of Surgical Oncology, University of Illinois at Chicago, Chicago, IL 60612, USA; Department of Biochemistry and Molecular Genetics, University of Illinois at Chicago, Chicago, IL 60607, USA
| | - Sandeep Kumar
- Department of Surgery, Division of Surgical Oncology, University of Illinois at Chicago, Chicago, IL 60612, USA; University of Illinois Hospital and Health Sciences System Cancer Center, University of Illinois at Chicago, Chicago, IL 60612, USA
| | - Sunil Kumar Singh
- Department of Surgery, Division of Surgical Oncology, University of Illinois at Chicago, Chicago, IL 60612, USA
| | - Ajay Rana
- Department of Surgery, Division of Surgical Oncology, University of Illinois at Chicago, Chicago, IL 60612, USA; University of Illinois Hospital and Health Sciences System Cancer Center, University of Illinois at Chicago, Chicago, IL 60612, USA; Jesse Brown VA Medical Center, Chicago, IL 60612, USA
| | - Basabi Rana
- Department of Surgery, Division of Surgical Oncology, University of Illinois at Chicago, Chicago, IL 60612, USA; University of Illinois Hospital and Health Sciences System Cancer Center, University of Illinois at Chicago, Chicago, IL 60612, USA; Jesse Brown VA Medical Center, Chicago, IL 60612, USA.
| |
Collapse
|
11
|
Gu W, Li C, Shen T, Tong L, Yuan W, Zheng X, Wang T, Wang S, Zhu B, Zhang C, Zhang C. NAT1 inhibits liver metastasis of colorectal cancer by regulating EMT and glycolysis. Aging (Albany NY) 2024; 16:10546-10562. [PMID: 38916406 PMCID: PMC11236320 DOI: 10.18632/aging.205957] [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: 01/03/2024] [Accepted: 05/29/2024] [Indexed: 06/26/2024]
Abstract
Metastasis is the primary cause of cancer-related deaths, and colorectal cancer (CRC) liver metastasis is a major poor prognostic factor in CRC. NAT1 (N-acetyltransferase 1) plays a crucial role in the invasive and metastatic processes of colorectal cancer. The role and molecular mechanism of NAT1 on tumor cells were verified by establishing a cell model of overexpression and knockdown of NAT1, and further verified by establishing a liver metastasis model of colorectal cancer for animal experiments. In vivo and in vitro experiments have demonstrated that overexpression of NAT1 reduces the ability of metastasis and invasion of colorectal cancer cells. NAT1 overexpression inhibits the PI3K/AKT/mTOR signaling pathway, thereby suppressing the EMT (epithelial-mesenchymal transition) process and glycolytic ability of tumor cells. Additionally, decreased glycolytic ability results in reduced VEGF (Vascular endothelial growth factor) expression in colorectal cancer cells. The decreased VEGF expression leads to decreased angiogenesis and vascular permeability in liver metastases, ultimately reducing the occurrence of liver metastasis. Our findings highlight that overexpression of NAT1 significantly inhibits the PI3K/AKT/mTOR signaling pathway, thereby suppressing EMT, glycolytic ability, and VEGF expression in colorectal cancer cells, collectively preventing the development of liver metastasis.
Collapse
Affiliation(s)
- Wang Gu
- Department of General Surgery, The First Affiliated Hospital of Anhui Medical University, Hefei, Anhui 230022, China
| | - Chen Li
- Department of Biology, Chemistry, Pharmacy, Free University of Berlin, Berlin 14195, Germany
| | - Tingting Shen
- Department of Pathology, The First Affiliated Hospital of Anhui Medical University, Hefei, Anhui 230022, China
| | - Li Tong
- Department of General Surgery, The First Affiliated Hospital of Anhui Medical University, Hefei, Anhui 230022, China
| | - Wenkang Yuan
- Department of General Surgery, The First Affiliated Hospital of Anhui Medical University, Hefei, Anhui 230022, China
| | - Xiaofei Zheng
- Department of General Surgery, The First Affiliated Hospital of Anhui Medical University, Hefei, Anhui 230022, China
| | - Tianqi Wang
- Department of General Surgery, The First Affiliated Hospital of Anhui Medical University, Hefei, Anhui 230022, China
| | - Siyu Wang
- Department of General Surgery, The First Affiliated Hospital of Anhui Medical University, Hefei, Anhui 230022, China
| | - Benshuai Zhu
- Department of General Surgery, The First Affiliated Hospital of Anhui Medical University, Hefei, Anhui 230022, China
| | - Chong Zhang
- Department of General Surgery, The First Affiliated Hospital of Anhui Medical University, Hefei, Anhui 230022, China
| | - Chao Zhang
- Department of General Surgery, The First Affiliated Hospital of Anhui Medical University, Hefei, Anhui 230022, China
| |
Collapse
|
12
|
Wang Y, Peng J, Yang D, Xing Z, Jiang B, Ding X, Jiang C, Ouyang B, Su L. From metabolism to malignancy: the multifaceted role of PGC1α in cancer. Front Oncol 2024; 14:1383809. [PMID: 38774408 PMCID: PMC11106418 DOI: 10.3389/fonc.2024.1383809] [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: 02/08/2024] [Accepted: 04/16/2024] [Indexed: 05/24/2024] Open
Abstract
PGC1α, a central player in mitochondrial biology, holds a complex role in the metabolic shifts seen in cancer cells. While its dysregulation is common across major cancers, its impact varies. In some cases, downregulation promotes aerobic glycolysis and progression, whereas in others, overexpression escalates respiration and aggression. PGC1α's interactions with distinct signaling pathways and transcription factors further diversify its roles, often in a tissue-specific manner. Understanding these multifaceted functions could unlock innovative therapeutic strategies. However, challenges exist in managing the metabolic adaptability of cancer cells and refining PGC1α-targeted approaches. This review aims to collate and present the current knowledge on the expression patterns, regulators, binding partners, and roles of PGC1α in diverse cancers. We examined PGC1α's tissue-specific functions and elucidated its dual nature as both a potential tumor suppressor and an oncogenic collaborator. In cancers where PGC1α is tumor-suppressive, reinstating its levels could halt cell proliferation and invasion, and make the cells more receptive to chemotherapy. In cancers where the opposite is true, halting PGC1α's upregulation can be beneficial as it promotes oxidative phosphorylation, allows cancer cells to adapt to stress, and promotes a more aggressive cancer phenotype. Thus, to target PGC1α effectively, understanding its nuanced role in each cancer subtype is indispensable. This can pave the way for significant strides in the field of oncology.
Collapse
Affiliation(s)
- Yue Wang
- Department of Surgery, Nanjing Central Hospital, Nanjing, China
| | - Jianing Peng
- Division of Biosciences, University College London, London, United Kingdom
| | - Dengyuan Yang
- Department of Surgery, Nanjing Central Hospital, Nanjing, China
| | - Zhongjie Xing
- Department of Surgery, Nanjing Central Hospital, Nanjing, China
| | - Bo Jiang
- Department of General Surgery, Nanjing Drum Tower Hospital, Clinical College of Nanjing Medical University, Nanjing, China
| | - Xu Ding
- Department of Surgery, Nanjing Central Hospital, Nanjing, China
| | - Chaoyu Jiang
- Department of General Surgery, Nanjing Drum Tower Hospital, Clinical College of Nanjing Medical University, Nanjing, China
| | - Bing Ouyang
- Department of Surgery, Nanjing Central Hospital, Nanjing, China
| | - Lei Su
- Department of General Surgery, Nanjing Drum Tower Hospital, Clinical College of Nanjing Medical University, Nanjing, China
- Department of General Surgery, Affiliated Drum Tower Hospital, Medical School of Nanjing University, Nanjing, China
| |
Collapse
|
13
|
Hashemi M, Daneii P, Asadalizadeh M, Tabari K, Matinahmadi A, Bidoki SS, Motlagh YSM, Jafari AM, Ghorbani A, Dehghanpour A, Nabavi N, Tan SC, Rashidi M, Taheriazam A, Entezari M, Goharrizi MASB. Epigenetic regulation of hepatocellular carcinoma progression: MicroRNAs as therapeutic, diagnostic and prognostic factors. Int J Biochem Cell Biol 2024; 170:106566. [PMID: 38513802 DOI: 10.1016/j.biocel.2024.106566] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2023] [Revised: 01/28/2024] [Accepted: 03/19/2024] [Indexed: 03/23/2024]
Abstract
Hepatocellular carcinoma (HCC), a significant challenge for public healthcare systems in developed Western countries including the USA, Canada, and the UK, is influenced by different risk factors including hepatitis virus infections, alcoholism, and smoking. The disruption in the balance of microRNAs (miRNAs) plays a vital function in tumorigenesis, given their function as regulators in numerous signaling networks. These miRNAs, which are mature and active in the cytoplasm, work by reducing the expression of target genes through their impact on mRNAs. MiRNAs are particularly significant in HCC as they regulate key aspects of the tumor, like proliferation and invasion. Additionally, during treatment phases such as chemotherapy and radiotherapy, the levels of miRNAs are key determinants. Pre-clinical experiments have demonstrated that altered miRNA expression contributes to HCC development, metastasis, drug resistance, and radio-resistance, highlighting related molecular pathways and processes like MMPs, EMT, apoptosis, and autophagy. Furthermore, the regulatory role of miRNAs in HCC extends beyond their immediate function, as they are also influenced by other epigenetic factors like lncRNAs and circular RNAs (circRNAs), as discussed in recent reviews. Applying these discoveries in predicting the prognosis of HCC could mark a significant advancement in the therapy of this disease.
Collapse
Affiliation(s)
- Mehrdad Hashemi
- Department of Genetics, Faculty of Advanced Science and Technology, Tehran Medical Sciences, Islamic Azad University, Tehran, Iran; Farhikhtegan Medical Convergence Sciences Research Center, Farhikhtegan Hospital Tehran Medical Sciences, Islamic Azad University, Tehran, Iran
| | - Pouria Daneii
- Farhikhtegan Medical Convergence Sciences Research Center, Farhikhtegan Hospital Tehran Medical Sciences, Islamic Azad University, Tehran, Iran
| | - Mahya Asadalizadeh
- Farhikhtegan Medical Convergence Sciences Research Center, Farhikhtegan Hospital Tehran Medical Sciences, Islamic Azad University, Tehran, Iran
| | - Kiana Tabari
- Farhikhtegan Medical Convergence Sciences Research Center, Farhikhtegan Hospital Tehran Medical Sciences, Islamic Azad University, Tehran, Iran
| | - Arash Matinahmadi
- Department of Cellular and Molecular Biology, Nicolaus Copernicus University, Torun, Poland
| | - Seyed Shahabadin Bidoki
- Faculty of medicine, School of Medicine, Shahid Sadoughi University of Medical Sciences, Yazd, Iran
| | | | - Ali Moghadas Jafari
- Farhikhtegan Medical Convergence Sciences Research Center, Farhikhtegan Hospital Tehran Medical Sciences, Islamic Azad University, Tehran, Iran
| | - Amin Ghorbani
- Farhikhtegan Medical Convergence Sciences Research Center, Farhikhtegan Hospital Tehran Medical Sciences, Islamic Azad University, Tehran, Iran
| | - Amir Dehghanpour
- Farhikhtegan Medical Convergence Sciences Research Center, Farhikhtegan Hospital Tehran Medical Sciences, Islamic Azad University, Tehran, Iran
| | - Noushin Nabavi
- Department of Urologic Sciences and Vancouver Prostate Centre, University of British Columbia, V6H3Z6, Vancouver, BC, Canada
| | - Shing Cheng Tan
- UKM Medical Molecular Biology Institute, Universiti Kebangsaan Malaysia, Kuala Lumpur, Malaysia
| | - Mohsen Rashidi
- Department Pharmacology, Faculty of Medicine, Mazandaran University of Medical Sciences, Sari, Iran; The Health of Plant and Livestock Products Research Center, Mazandaran University of Medical Sciences, Sari, Iran.
| | - Afshin Taheriazam
- Farhikhtegan Medical Convergence Sciences Research Center, Farhikhtegan Hospital Tehran Medical Sciences, Islamic Azad University, Tehran, Iran; Department of Orthopedics, Faculty of Medicine, Tehran Medical Sciences, Islamic Azad University, Tehran, Iran.
| | - Maliheh Entezari
- Department of Genetics, Faculty of Advanced Science and Technology, Tehran Medical Sciences, Islamic Azad University, Tehran, Iran; Farhikhtegan Medical Convergence Sciences Research Center, Farhikhtegan Hospital Tehran Medical Sciences, Islamic Azad University, Tehran, Iran.
| | | |
Collapse
|
14
|
Upadhyay S, Khan S, Hassan MI. Exploring the diverse role of pyruvate kinase M2 in cancer: Navigating beyond glycolysis and the Warburg effect. Biochim Biophys Acta Rev Cancer 2024; 1879:189089. [PMID: 38458358 DOI: 10.1016/j.bbcan.2024.189089] [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: 12/20/2023] [Revised: 02/25/2024] [Accepted: 03/03/2024] [Indexed: 03/10/2024]
Abstract
Pyruvate Kinase M2, a key enzyme in glycolysis, has garnered significant attention in cancer research due to its pivotal role in the metabolic reprogramming of cancer cells. Originally identified for its association with the Warburg effect, PKM2 has emerged as a multifaceted player in cancer biology. The functioning of PKM2 is intricately regulated at multiple levels, including controlling the gene expression via various transcription factors and non-coding RNAs, as well as adding post-translational modifications that confer distinct functions to the protein. Here, we explore the diverse functions of PKM2, encompassing newly emerging roles in non-glycolytic metabolic regulation, immunomodulation, inflammation, DNA repair and mRNA processing, beyond its canonical role in glycolysis. The ever-expanding list of its functions has recently grown to include roles in subcellular compartments such as the mitochondria and extracellular milieu as well, all of which make PKM2 an attractive drug target in the pursuit of therapeutics for cancer.
Collapse
Affiliation(s)
- Saurabh Upadhyay
- Kusuma School of Biological Sciences, Indian Institute of Technology Delhi, Hauz Khas, New Delhi 110016, India
| | - Shumayila Khan
- International Health Division, Indian Council of Medical Research, Ansari Nagar, New Delhi 110029, India
| | - Md Imtaiyaz Hassan
- Centre for Interdisciplinary Research in Basic Sciences, Jamia Millia Islamia, New Delhi 110025, India.
| |
Collapse
|
15
|
Medrano M, Contreras M, Caballero-Velázquez T, Martínez L, Bejarano-García JA, Calderón-Ruiz R, García-Calderón CB, Rosado IV, Pérez-Simón JA. Cannabinoids induce cell death in leukaemic cells through Parthanatos and PARP-related metabolic disruptions. Br J Cancer 2024; 130:1529-1541. [PMID: 38461169 PMCID: PMC11058274 DOI: 10.1038/s41416-024-02618-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2023] [Revised: 02/01/2024] [Accepted: 02/08/2024] [Indexed: 03/11/2024] Open
Abstract
BACKGROUND Several studies have described a potential anti-tumour effect of cannabinoids (CNB). CNB receptor 2 (CB2) is mostly present in hematopoietic stem cells (HSC). The present study evaluates the anti-leukaemic effect of CNB. METHODS Cell lines and primary cells from acute myeloid leukaemia (AML) patients were used and the effect of the CNB derivative WIN-55 was evaluated in vitro, ex vivo and in vivo. RESULTS We demonstrate a potent antileukemic effect of WIN-55 which is abolished with CB antagonists. WIN-treated mice, xenografted with AML cells, had better survival as compared to vehicle or cytarabine. DNA damage-related genes were affected upon exposure to WIN. Co-incubation with the PARP inhibitor Olaparib prevented WIN-induced cell death, suggesting PARP-mediated apoptosis which was further confirmed with the translocation of AIF to the nucleus observed in WIN-treated cells. Nicotinamide prevented WIN-related apoptosis, indicating NAD+ depletion. Finally, WIN altered glycolytic enzymes levels as well as the activity of G6PDH. These effects are reversed through PARP1 inhibition. CONCLUSIONS WIN-55 exerts an antileukemic effect through Parthanatos, leading to translocation of AIF to the nucleus and depletion of NAD+, which are reversed through PARP1 inhibition. It also induces metabolic disruptions. These effects are not observed in normal HSC.
Collapse
Affiliation(s)
- M Medrano
- Instituto de Biomedicina de Sevilla (IBIS/CSIC), Universidad de Sevilla, Seville, Spain
| | - M Contreras
- Instituto de Biomedicina de Sevilla (IBIS/CSIC), Universidad de Sevilla, Seville, Spain
| | - T Caballero-Velázquez
- Instituto de Biomedicina de Sevilla (IBIS/CSIC), Universidad de Sevilla, Seville, Spain
- Department of Hematology, University Hospital Virgen del Rocío, Universidad de Sevilla, Seville, Spain
| | - L Martínez
- Department of Medical Biochemistry, Molecular Biology and Immunology, Universidad de Sevilla, Seville, Spain
- Centro Andaluz de Biología Molecular y Medicina Regenerativa (CABIMER), Universidad de Sevilla-CSIC-Universidad Pablo de Olavide, Seville, Spain
| | - J A Bejarano-García
- Instituto de Biomedicina de Sevilla (IBIS/CSIC), Universidad de Sevilla, Seville, Spain
| | - R Calderón-Ruiz
- Instituto de Biomedicina de Sevilla (IBIS/CSIC), Universidad de Sevilla, Seville, Spain
| | - C B García-Calderón
- Instituto de Biomedicina de Sevilla (IBIS/CSIC), Universidad de Sevilla, Seville, Spain
| | - I V Rosado
- Centro Andaluz de Biología Molecular y Medicina Regenerativa (CABIMER), Universidad de Sevilla-CSIC-Universidad Pablo de Olavide, Seville, Spain
| | - J A Pérez-Simón
- Instituto de Biomedicina de Sevilla (IBIS/CSIC), Universidad de Sevilla, Seville, Spain.
- Department of Hematology, University Hospital Virgen del Rocío, Universidad de Sevilla, Seville, Spain.
| |
Collapse
|
16
|
Liu F, Chen Y, Qin D, Qian C. Interleukin-22 inhibits cardiac fibrosis by regulating fibroblast metabolic reprogramming in myocardial infarction. Pathol Res Pract 2024; 256:155256. [PMID: 38492359 DOI: 10.1016/j.prp.2024.155256] [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: 08/28/2023] [Revised: 01/09/2024] [Accepted: 03/06/2024] [Indexed: 03/18/2024]
Abstract
Cardiac fibrosis, a significant characteristic of cardiovascular diseases, leads to ventricular remodeling and impaired cardiac function. In this study, we aimed to investigate the role of Interleukin-22 (IL-22) in myocardial fibrosis following myocardial infarction (MI) and to explore the underlying metabolic mechanisms. Here we analyzed the single-cell sequencing data and found that the level of aerobic glycolysis was significantly higher in cardiac fibrosis in MI patient, which we validated through in vivo experiments. Utilizing MI mouse model, these experiments revealed decreased serum IL-22 levels and increased levels of AngII and TGF-β1. However, treatment with exogenous IL-22 reversed these changes, reduced infarct size, and fibrosis. In vitro experiments demonstrated that IL-22 inhibited AngII-induced fibroblast-to-myofibroblast transition (FMT) by suppressing the expression of α-SMA, Cola1, and Cola3. Metabolic analysis indicated that IL-22 decreased the expression of glycolytic enzymes and reduced lactate production in cardiac fibroblasts. Further in vivo experiments confirmed the inhibitory effect of IL-22 on Pyruvate kinase isoform M2 (PKM2) levels in heart tissue. Additionally, IL-22 activated the c-Jun N-terminal kinase (JNK) pathway, while inhibition of JNK partially reversed IL-22's effect on PKM2 activity. These findings suggest that IL-22 mitigates cardiac fibrosis and FMT by inhibiting aerobic glycolysis by activating the JNK/PKM2 pathway. Our study highlights IL-22 as a potential therapeutic target for myocardial fibrosis and cardiovascular diseases, providing insights into its role in regulating fibrosis and glycolysis. These findings pave the way for developing targeted therapies and investigating additional metabolic pathways for improved treatment outcomes in the field of cardiovascular diseases.
Collapse
Affiliation(s)
- Fang Liu
- Department of Vascular Surgery, Affiliated Hospital of Jiangsu University, Zhenjiang 212001, China; International Genome Center, Jiangsu University, Zhenjiang 212013, China.
| | - Yueqi Chen
- International Genome Center, Jiangsu University, Zhenjiang 212013, China
| | - Demeng Qin
- International Genome Center, Jiangsu University, Zhenjiang 212013, China
| | - Cheng Qian
- International Genome Center, Jiangsu University, Zhenjiang 212013, China
| |
Collapse
|
17
|
Fei M, Li X, Liang S, Zhou S, Wu H, Sun L, Liu Y, Hu Q, Liu L, Wang J. LncRNA PWRN1 inhibits the progression of hepatocellular carcinoma by activating PKM2 activity. Cancer Lett 2024; 584:216620. [PMID: 38218456 DOI: 10.1016/j.canlet.2024.216620] [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: 08/06/2023] [Revised: 12/14/2023] [Accepted: 12/23/2023] [Indexed: 01/15/2024]
Abstract
Hepatocellular carcinoma (HCC) is one of the most prevalent and leading causes of cancer-related mortality worldwide. Long non-coding RNAs (lncRNAs) have been demonstrated to play vital roles in cancer development and progression. The lncRNA PWRN1 (PWRN1), acts as a tumor suppressor factor, which is low expressed in some cancers. However, the molecular mechanisms underlying the effects of PWRN1, especially the regulatory relationship with RNA binding protein in HCC remain largely unknown. In the present study, we demonstrated that PWRN1 was significantly down-regulated in HCC and correlated with better prognosis; furthermore, gain-of-function experiments showed that PWRN1 inhibited the proliferation of HCC cells. We further found that PWRN1 up-regulated pyruvate kinase activity and thus hinders the proliferation of HCC in vitro and in vivo. Mechanistically, pyruvate kinase M2 (PKM2) was bound to it and maintained the high activity state of PKM2, thereby hindering PKM2 from entering the nucleus in the form of low-activity dimers, reducing the expression of c-Myc downstream gene LDHA, leading to a decrease in lactate levels, and inhibiting the growth of tumor cells. In addition, PWRN1 was found to inhibit aerobic glycolysis. Finally, TEPP-46, a pyruvate kinase activator, appeared to inhibit HCC proliferation by maintaining tetramer stability and increasing pyruvate kinase activity. Taken together, our results provide new insights into the biology hindering HCC proliferation and indicate that PWRN1 in combination with PKM2 activators might represent a novel therapeutic target for HCC.
Collapse
Affiliation(s)
- Mingming Fei
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Anhui Province Key Laboratory of Hepatopancreatobiliary Surgery, Anhui Provincial Clinical Research Center for Hepatobiliary Diseases, Swan Lake Road, Hefei, 230001, Anhui Province, China
| | - Xianying Li
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Anhui Province Key Laboratory of Hepatopancreatobiliary Surgery, Anhui Provincial Clinical Research Center for Hepatobiliary Diseases, Swan Lake Road, Hefei, 230001, Anhui Province, China
| | - Shuhang Liang
- Department of Gastrointestinal Surgery, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Anhui Province Key Laboratory of Hepatopancreatobiliary Surgery, Swan Lake Road, Hefei, 230001, Anhui Province, China
| | - Shuo Zhou
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Anhui Province Key Laboratory of Hepatopancreatobiliary Surgery, Anhui Provincial Clinical Research Center for Hepatobiliary Diseases, Swan Lake Road, Hefei, 230001, Anhui Province, China
| | - Huihui Wu
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Anhui Province Key Laboratory of Hepatopancreatobiliary Surgery, Anhui Provincial Clinical Research Center for Hepatobiliary Diseases, Swan Lake Road, Hefei, 230001, Anhui Province, China
| | - Linmao Sun
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Anhui Province Key Laboratory of Hepatopancreatobiliary Surgery, Anhui Provincial Clinical Research Center for Hepatobiliary Diseases, Swan Lake Road, Hefei, 230001, Anhui Province, China
| | - Yao Liu
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Anhui Province Key Laboratory of Hepatopancreatobiliary Surgery, Anhui Provincial Clinical Research Center for Hepatobiliary Diseases, Swan Lake Road, Hefei, 230001, Anhui Province, China
| | - Qingsong Hu
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Anhui Province Key Laboratory of Hepatopancreatobiliary Surgery, Anhui Provincial Clinical Research Center for Hepatobiliary Diseases, Swan Lake Road, Hefei, 230001, Anhui Province, China.
| | - Lianxin Liu
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Anhui Province Key Laboratory of Hepatopancreatobiliary Surgery, Anhui Provincial Clinical Research Center for Hepatobiliary Diseases, Swan Lake Road, Hefei, 230001, Anhui Province, China.
| | - Jiabei Wang
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Anhui Province Key Laboratory of Hepatopancreatobiliary Surgery, Anhui Provincial Clinical Research Center for Hepatobiliary Diseases, Swan Lake Road, Hefei, 230001, Anhui Province, China.
| |
Collapse
|
18
|
Dou Q, Grant AK, Coutinto de Souza P, Moussa M, Nasser I, Ahmed M, Tsai LL. Characterizing Metabolic Heterogeneity of Hepatocellular Carcinoma with Hyperpolarized 13C Pyruvate MRI and Mass Spectrometry. Radiol Imaging Cancer 2024; 6:e230056. [PMID: 38426887 PMCID: PMC10988335 DOI: 10.1148/rycan.230056] [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: 04/27/2023] [Revised: 12/16/2023] [Accepted: 01/11/2024] [Indexed: 03/02/2024]
Abstract
Purpose To characterize the metabolomic profiles of two hepatocellular carcinoma (HCC) rat models, track evolution of these profiles to a stimulated tumor state, and assess their effect on lactate flux with hyperpolarized (HP) carbon 13 (13C) MRI. Materials and Methods Forty-three female adult Fischer rats were implanted with N1S1 or McA-RH7777 HCC tumors. In vivo lactate-to-pyruvate ratio (LPR) was measured with HP 13C MRI at 9.4 T. Ex vivo mass spectrometry was used to measure intratumoral metabolites, and Ki67 labeling was used to quantify proliferation. Tumors were first compared with three normal liver controls. The tumors were then compared with stimulated variants via off-target hepatic thermal ablation treatment. All comparisons were made using the Mann-Whitney test. Results HP 13C pyruvate MRI showed greater LPR in N1S1 tumors compared with normal liver (mean [SD], 0.564 ± 0.194 vs 0.311 ± 0.057; P < .001 [n = 9]), but not for McA-RH7777 (P = .44 [n = 8]). Mass spectrometry confirmed that the glycolysis pathway was increased in N1S1 tumors and decreased in McA-RH7777 tumors. The pentose phosphate pathway was also decreased only in McA-RH7777 tumors. Increased proliferation in stimulated N1S1 tumors corresponded to a net increase in LPR (six stimulated vs six nonstimulated, 0.269 ± 0.148 vs 0.027 ± 0.08; P = .009), but not in McA-RH7777 (eight stimulated vs six nonstimulated, P = .13), despite increased proliferation and metastases. Mass spectrometry demonstrated relatively increased lactate production with stimulation in N1S1 tumors only. Conclusion Two HCC subtypes showed divergent glycolytic dependency at baseline and during transformation to a high proliferation state. This metabolic heterogeneity in HCC should be considered with use of HP 13C MRI for diagnosis and tracking. Keywords: Molecular Imaging-Probe Development, Liver, Abdomen/GI, Oncology, Hepatocellular Carcinoma © RSNA, 2024 See also commentary by Ohliger in this issue.
Collapse
Affiliation(s)
- Qianhui Dou
- From the Departments of Radiology (Q.D., A.K.G., M.M., M.A., L.L.T.)
and Pathology (I.N.), Beth Israel Deaconess Medical Center, Harvard Medical
School, 330 Brookline Ave, ANSIN 230, Boston, MA 02215; and Merck & Co,
Rahway, NJ (P.C.d.S.)
| | - Aaron K. Grant
- From the Departments of Radiology (Q.D., A.K.G., M.M., M.A., L.L.T.)
and Pathology (I.N.), Beth Israel Deaconess Medical Center, Harvard Medical
School, 330 Brookline Ave, ANSIN 230, Boston, MA 02215; and Merck & Co,
Rahway, NJ (P.C.d.S.)
| | - Patricia Coutinto de Souza
- From the Departments of Radiology (Q.D., A.K.G., M.M., M.A., L.L.T.)
and Pathology (I.N.), Beth Israel Deaconess Medical Center, Harvard Medical
School, 330 Brookline Ave, ANSIN 230, Boston, MA 02215; and Merck & Co,
Rahway, NJ (P.C.d.S.)
| | - Marwan Moussa
- From the Departments of Radiology (Q.D., A.K.G., M.M., M.A., L.L.T.)
and Pathology (I.N.), Beth Israel Deaconess Medical Center, Harvard Medical
School, 330 Brookline Ave, ANSIN 230, Boston, MA 02215; and Merck & Co,
Rahway, NJ (P.C.d.S.)
| | - Imad Nasser
- From the Departments of Radiology (Q.D., A.K.G., M.M., M.A., L.L.T.)
and Pathology (I.N.), Beth Israel Deaconess Medical Center, Harvard Medical
School, 330 Brookline Ave, ANSIN 230, Boston, MA 02215; and Merck & Co,
Rahway, NJ (P.C.d.S.)
| | - Muneeb Ahmed
- From the Departments of Radiology (Q.D., A.K.G., M.M., M.A., L.L.T.)
and Pathology (I.N.), Beth Israel Deaconess Medical Center, Harvard Medical
School, 330 Brookline Ave, ANSIN 230, Boston, MA 02215; and Merck & Co,
Rahway, NJ (P.C.d.S.)
| | - Leo L. Tsai
- From the Departments of Radiology (Q.D., A.K.G., M.M., M.A., L.L.T.)
and Pathology (I.N.), Beth Israel Deaconess Medical Center, Harvard Medical
School, 330 Brookline Ave, ANSIN 230, Boston, MA 02215; and Merck & Co,
Rahway, NJ (P.C.d.S.)
| |
Collapse
|
19
|
Luo Y, Yang Y, Ye M, Zuo J. Targeting metabolic reprogramming promotes the efficacy of transarterial chemoembolization in the rabbit VX2 liver tumor model. Oncol Lett 2024; 27:111. [PMID: 38304171 PMCID: PMC10831404 DOI: 10.3892/ol.2024.14244] [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: 07/25/2023] [Accepted: 11/30/2023] [Indexed: 02/03/2024] Open
Abstract
Transarterial chemoembolization (TACE) may prolong the survival of patients with hepatocellular carcinoma (HCC); however, its efficacy is limited due to the high rate of incomplete embolization. Hypoxia after embolization can cause a series of changes in the tumor microenvironment, including lactate dehydrogenase A (LDHA) upregulation. Therefore, the current study assessed the antitumor effect and the underlying mechanism of the LDHA inhibitor, sodium oxamate (Ox), combined with TACE, using the rabbit VX2 liver tumor model. VX2 liver tumor models were created in the left liver lobe of rabbits, and after 14 days of treatments, the rabbits were sacrificed for the collection of the tumor tissues and blood samples. The antitumor effects of Ox, and the combination of Ox and TACE, and changes in the tumor microenvironment after treatments were assessed by histopathological evaluation, and the safety of the treatments was analyzed by measuring changes in the serum levels of alanine aminotransferase, aspartate aminotransferase, blood urea nitrogen and creatinine. The results demonstrated that the combination of Ox and TACE notably improved antitumor effects compared with in the other groups, as it significantly inhibited tumor growth. Additionally, treatment with Ox + TACE downregulated vascular endothelial growth factor and matrix metalloproteinase-9, and enhanced the infiltration of CD3+ and CD8+ T cells into tumor tissues, thus suggesting that Ox + TACE may have a synergistic effect on increasing the infiltration of immune cells in the tumor microenvironment. With a well-tolerated and manageable impairment of hepatorenal function, targeting metabolic reprogramming could promote the efficacy of TACE, thus providing novel avenues for the future clinical management of patients with advanced HCC.
Collapse
Affiliation(s)
- Yi Luo
- Department of Interventional Radiology, The Second Hospital of Wuhan Iron and Steel (Group) Corp., Wuhan, Hubei 430022, P.R. China
| | - Yong Yang
- Department of Oncology, The Second Hospital of Wuhan Iron and Steel (Group) Corp., Wuhan, Hubei 430022, P.R. China
| | - Meize Ye
- Department of Interventional Radiology, Huazhong University of Science and Technology, Wuhan, Hubei 430000, P.R. China
| | - Jing Zuo
- Department of Oncology, The Second Hospital of Wuhan Iron and Steel (Group) Corp., Wuhan, Hubei 430022, P.R. China
| |
Collapse
|
20
|
Saleh H, Liloglou T, Rigden DJ, Parsons JL, Grundy GJ. KH-like Domains in PARP9/DTX3L and PARP14 Coordinate Protein-Protein Interactions to Promote Cancer Cell Survival. J Mol Biol 2024; 436:168434. [PMID: 38182103 PMCID: PMC11080071 DOI: 10.1016/j.jmb.2023.168434] [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: 09/18/2023] [Revised: 12/12/2023] [Accepted: 12/30/2023] [Indexed: 01/07/2024]
Abstract
Certain members of the ADP-ribosyltransferase superfamily (ARTD or PARP enzymes) catalyse ADP-ribosylation in response to cellular stress, DNA damage and viral infection and are upregulated in various tumours. PARP9, its binding partner DTX3L and PARP14 protein levels are significantly correlated in head and neck squamous cell carcinoma (HNSCC) and other tumour types though a mechanism where PARP9/DTX3L regulates PARP14 post-transcriptionally. Depleting PARP9, DTX3L or PARP14 expression in HNSCC or HeLa cell lines decreases cell survival through a reduction of proliferation and an increase in apoptosis. A partial rescue of survival was achieved by expressing a PARP14 truncation containing a predicted eukaryotic type I KH domain. KH-like domains were also found in PARP9 and in DTX3L and contributed to protein-protein interactions between PARP9-DTX3L and PARP14-DTX3L. Homodimerization of DTX3L was also coordinated by a KH-like domain and was disrupted by site-specific mutation. Although, cell survival promoted by PARP14 did not require ADP-ribosyltransferase activity, interaction of DTX3L in vitro suppressed PARP14 auto-ADP-ribosylation and promoted trans-ADP-ribosylation of PARP9 and DTX3L. In summary, we characterised PARP9-DTX3L-PARP14 interactions important to pro-survival signalling in HNSCC cells, albeit in PARP14 catalytically independent fashion.
Collapse
Affiliation(s)
- Hadil Saleh
- University of Liverpool, Department of Molecular and Clinical Cancer Medicine, 6 West Derby St, Liverpool L7 8TX, UK
| | - Triantafillos Liloglou
- Edge Hill University, Faculty of Health, Social Care & Medicine, St Helens Road, Ormskirk, Lancashire L39 4QP, UK
| | - Daniel J Rigden
- University of Liverpool, Department of Biochemistry, Cell and Systems Biology, Liverpool L69 7ZB, UK
| | - Jason L Parsons
- University of Birmingham, Institute of Cancer and Genomic Sciences, IBR West, Birmingham B15 2TT, UK
| | - Gabrielle J Grundy
- University of Liverpool, Department of Molecular and Clinical Cancer Medicine, 6 West Derby St, Liverpool L7 8TX, UK.
| |
Collapse
|
21
|
Chang X, Yan S, Zhang Y, Zhang Y, Li L, Gao Z, Lin X, Chi X. GINv2.0: a comprehensive topological network integrating molecular interactions from multiple knowledge bases. NPJ Syst Biol Appl 2024; 10:4. [PMID: 38218959 PMCID: PMC10787761 DOI: 10.1038/s41540-024-00330-y] [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: 08/19/2023] [Accepted: 01/02/2024] [Indexed: 01/15/2024] Open
Abstract
Knowledge bases have been instrumental in advancing biological research, facilitating pathway analysis and data visualization, which are now widely employed in the scientific community. Despite the establishment of several prominent knowledge bases focusing on signaling, metabolic networks, or both, integrating these networks into a unified topological network has proven to be challenging. The intricacy of molecular interactions and the diverse formats employed to store and display them contribute to the complexity of this task. In a prior study, we addressed this challenge by introducing a "meta-pathway" structure that integrated the advantages of the Simple Interaction Format (SIF) while accommodating reaction information. Nevertheless, the earlier Global Integrative Network (GIN) was limited to reliance on KEGG alone. Here, we present GIN version 2.0, which incorporates human molecular interaction data from ten distinct knowledge bases, including KEGG, Reactome, and HumanCyc, among others. We standardized the data structure, gene IDs, and chemical IDs, and conducted a comprehensive analysis of the consistency among the ten knowledge bases before combining all unified interactions into GINv2.0. Utilizing GINv2.0, we investigated the glycolysis process and its regulatory proteins, revealing coordinated regulations on glycolysis and autophagy, particularly under glucose starvation. The expanded scope and enhanced capabilities of GINv2.0 provide a valuable resource for comprehensive systems-level analyses in the field of biological research. GINv2.0 can be accessed at: https://github.com/BIGchix/GINv2.0 .
Collapse
Affiliation(s)
- Xiao Chang
- Department of Dermatology and Venereal Disease, Xuan Wu Hospital, Beijing, 100053, China
| | - Shen Yan
- Agricultural Information Institute, Chinese Academy of Agricultural Science, Beijing, 100081, China
| | - Yizheng Zhang
- CAS Key Laboratory of Genomic and Precision Medicine, Beijing Institute of Genomics, Chinese Academy of Sciences and China National Center for Bioinformation, Beijing, 100101, China
- University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Yingchun Zhang
- Key Laboratory of Plant Molecular Physiology, Institute of Botany, Chinese Academy of Sciences, Beijing, 100093, China
| | - Luyang Li
- CAS Key Laboratory of Genomic and Precision Medicine, Beijing Institute of Genomics, Chinese Academy of Sciences and China National Center for Bioinformation, Beijing, 100101, China
- University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Zhanyu Gao
- CAS Key Laboratory of Genomic and Precision Medicine, Beijing Institute of Genomics, Chinese Academy of Sciences and China National Center for Bioinformation, Beijing, 100101, China
- University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Xuefei Lin
- Department of Dermatology and Venereal Disease, Xuan Wu Hospital, Beijing, 100053, China
| | - Xu Chi
- CAS Key Laboratory of Genomic and Precision Medicine, Beijing Institute of Genomics, Chinese Academy of Sciences and China National Center for Bioinformation, Beijing, 100101, China.
| |
Collapse
|
22
|
Chen P, Lou L, Sharma B, Li M, Xie C, Yang F, Wu Y, Xiao Q, Gao L. Recent Advances on PKM2 Inhibitors and Activators in Cancer Applications. Curr Med Chem 2024; 31:2955-2973. [PMID: 37455458 DOI: 10.2174/0929867331666230714144851] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2022] [Revised: 05/28/2023] [Accepted: 06/05/2023] [Indexed: 07/18/2023]
Abstract
Metabolic reprogramming of cells, from the normal mode of glucose metabolism named glycolysis, is a pivotal characteristic of impending cancerous cells. Pyruvate kinase M2 (PKM2), an important enzyme that catalyzes the final rate-limiting stage during glycolysis, is highly expressed in numerous types of tumors and aids in development of favorable conditions for the survival of tumor cells. Increasing evidence has suggested that PKM2 is one of promising targets for innovative drug discovery, especially for the developments of antitumor therapeutics. Herein, we systematically summarize the recent advancement on PKM2 modulators including inhibitors and activators in cancer applications. We also discussed the classifications of pyruvate kinases in mammals and the biological functions of PKM2 in this review. We do hope that this review would provide a comprehensive understanding of the current research on PKM2 modulators, which may benefit the development of more potent PKM2-related drug candidates to treat PKM2-associated diseases including cancers in future.
Collapse
Affiliation(s)
- Peng Chen
- School of Pharmaceutical Sciences (Shenzhen), Shenzhen Campus of Sun Yat-Sen University, Shenzhen 518107, P.R. China
| | - Liang Lou
- School of Pharmaceutical Sciences (Shenzhen), Shenzhen Campus of Sun Yat-Sen University, Shenzhen 518107, P.R. China
| | - Bigyan Sharma
- School of Pharmaceutical Sciences (Shenzhen), Shenzhen Campus of Sun Yat-Sen University, Shenzhen 518107, P.R. China
| | - Mengchu Li
- School of Pharmaceutical Sciences (Shenzhen), Shenzhen Campus of Sun Yat-Sen University, Shenzhen 518107, P.R. China
| | - Chengliang Xie
- School of Pharmaceutical Sciences (Shenzhen), Shenzhen Campus of Sun Yat-Sen University, Shenzhen 518107, P.R. China
| | - Fen Yang
- School of Pharmaceutical Sciences (Shenzhen), Shenzhen Campus of Sun Yat-Sen University, Shenzhen 518107, P.R. China
| | - Yihang Wu
- School of Pharmaceutical Sciences (Shenzhen), Shenzhen Campus of Sun Yat-Sen University, Shenzhen 518107, P.R. China
| | - Qicai Xiao
- School of Pharmaceutical Sciences (Shenzhen), Shenzhen Campus of Sun Yat-Sen University, Shenzhen 518107, P.R. China
| | - Liqian Gao
- School of Pharmaceutical Sciences (Shenzhen), Shenzhen Campus of Sun Yat-Sen University, Shenzhen 518107, P.R. China
| |
Collapse
|
23
|
Wang B, Pu R. Association between glycolysis markers and prognosis of liver cancer: a systematic review and meta-analysis. World J Surg Oncol 2023; 21:390. [PMID: 38114977 PMCID: PMC10731852 DOI: 10.1186/s12957-023-03275-4] [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: 07/13/2023] [Accepted: 12/08/2023] [Indexed: 12/21/2023] Open
Abstract
BACKGROUND In recent years, the capacity of tumor cells to maintain high levels of glycolysis, even in the presence of oxygen, has emerged as one of the main metabolic traits and garnered considerable attention. The purpose of this meta-analysis is to investigate the prognostic value of glycolysis markers in liver cancer. METHODS PubMed, Embase, and Cochrane Library databases were searched for articles on glycolytic marker expression levels associated with the prognosis of liver cancer until April 2023. Stata SE14.0 was used to calculate the aggregate hazard ratios and 95% confidence intervals. RESULTS Thirty-five studies were included. The worse overall survival (OS) (P < 0.001), disease-free survival (DFS) (P = 0.001), recurrence-free survival (RFS) (P = 0.004), and time to recurrence (TTR) (P < 0.001) were significantly associated with elevated expression of glycolysis markers. Higher expression of PKM2 (P < 0.001), STMN1 (P = 0.002), MCT4 (P < 0.001), GLUT1 (P = 0.025), HK-2 (P < 0.001), and CA9 (P < 0.001) were significantly related to shorter OS. Increased levels of PKM2 (P < 0.001), CA9 (P = 0.005), and MCT4 (P < 0.001) were associated with worse DFS. Elevated PKM2 expression (P = 0.002) was also associated with poorer RFS in hepatocellular carcinoma patients. GLUT2 expression was not correlated with the prognosis of liver cancer (P = 0.134). CONCLUSIONS Elevated expression of glycolysis markers was associated with worse OS, DFS, RFS, and TTR in patients with liver cancer. Therefore, these glycolysis markers could serve as potential prognostic markers and therapeutic targets in liver cancer. TRIAL REGISTRATION PROSPERO registration: CRD42023469645.
Collapse
Affiliation(s)
- Boqin Wang
- Department of Clinical Laboratory, SSL Central Hospital of Dongguan City, Dongguan, Guangdong, China
| | - Rong Pu
- Department of Clinical Laboratory, SSL Central Hospital of Dongguan City, Dongguan, Guangdong, China.
| |
Collapse
|
24
|
Wang H, Luo S, Wu X, Ruan Y, Qiu L, Feng H, Zhu S, You Y, Li M, Yang W, Zhao Y, Tao X, Jiang H. Exploration of glycosyltransferases mutation status in cervical cancer reveals PARP14 as a potential prognostic marker. Glycoconj J 2023; 40:513-522. [PMID: 37650946 PMCID: PMC10638145 DOI: 10.1007/s10719-023-10134-7] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2023] [Revised: 07/31/2023] [Accepted: 08/17/2023] [Indexed: 09/01/2023]
Abstract
This study investigates the potential role of Glycosyltransferases (GTs) in the glycosylation process and their association with malignant tumors. Specifically, the study focuses on PARP14, a member of GTs, and its potential as a target for tumors in the diagnosis and treatment of cervical cancer. To gather data, the study used somatic mutation data, gene expression data and clinical information from TCGA-CESE dataset as well as tissue samples from cervical cancer patients. Further verification was conducted through RT-qPCR and immunohistochemistry staining on cervical cancer tissues to confirm the expression of PARP14. The study utilized Kaplan-Meier for survival analysis of cervical cancer patient and found significant mutational abnormalities in GTs. The high frequency mutated gene was identified as PARP14. RT-qPCR revealed significantly higher mRNA expression of PARP14 compared to precancerous tissue. Using IHC combined with Kaplan-Meier,patients in the PARP14 high expression group had a better prognosis than the low expression group. The study identified PARP14 as a frequently mutated gene in cervical cancer and proposed its potential role in diagnosis and treatment.
Collapse
Affiliation(s)
- Hui Wang
- Obstetrics & Gynecology Hospital of Fudan University, Shanghai, 200090, China
| | - Shen Luo
- Obstetrics & Gynecology Hospital of Fudan University, Shanghai, 200090, China
| | - Xin Wu
- Obstetrics & Gynecology Hospital of Fudan University, Shanghai, 200090, China
| | - Yuanyuan Ruan
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Fudan University, Shanghai, 200032, China
| | - Ling Qiu
- Obstetrics & Gynecology Hospital of Fudan University, Shanghai, 200090, China
| | - Hao Feng
- Obstetrics & Gynecology Hospital of Fudan University, Shanghai, 200090, China
| | - Shurong Zhu
- Obstetrics & Gynecology Hospital of Fudan University, Shanghai, 200090, China
| | - Yanan You
- Obstetrics & Gynecology Hospital of Fudan University, Shanghai, 200090, China
| | - Ming Li
- Obstetrics & Gynecology Hospital of Fudan University, Shanghai, 200090, China
| | - Wenting Yang
- Shanghai Genenexus healthcare technology company, Shanghai, 200433, China
| | - Yanding Zhao
- Department of Molecular and Systems Biology, The Geisel School of Medicine at Dartmouth, 03756, Lebanon, NH, USA
| | - Xiang Tao
- Obstetrics & Gynecology Hospital of Fudan University, Shanghai, 200090, China
| | - Hua Jiang
- Obstetrics & Gynecology Hospital of Fudan University, Shanghai, 200090, China.
| |
Collapse
|
25
|
Wong CW, Evangelou C, Sefton KN, Leshem R, Zhang W, Gopalan V, Chattrakarn S, Fernandez Carro ML, Uzuner E, Mole H, Wilcock DJ, Smith MP, Sergiou K, Telfer BA, Isaac DT, Liu C, Perl NR, Marie K, Lorigan P, Williams KJ, Rao PE, Nagaraju RT, Niepel M, Hurlstone AFL. PARP14 inhibition restores PD-1 immune checkpoint inhibitor response following IFNγ-driven acquired resistance in preclinical cancer models. Nat Commun 2023; 14:5983. [PMID: 37752135 PMCID: PMC10522711 DOI: 10.1038/s41467-023-41737-1] [Citation(s) in RCA: 19] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2022] [Accepted: 09/18/2023] [Indexed: 09/28/2023] Open
Abstract
Resistance mechanisms to immune checkpoint blockade therapy (ICBT) limit its response duration and magnitude. Paradoxically, Interferon γ (IFNγ), a key cytokine for cellular immunity, can promote ICBT resistance. Using syngeneic mouse tumour models, we confirm that chronic IFNγ exposure confers resistance to immunotherapy targeting PD-1 (α-PD-1) in immunocompetent female mice. We observe upregulation of poly-ADP ribosyl polymerase 14 (PARP14) in chronic IFNγ-treated cancer cell models, in patient melanoma with elevated IFNG expression, and in melanoma cell cultures from ICBT-progressing lesions characterised by elevated IFNγ signalling. Effector T cell infiltration is enhanced in tumours derived from cells pre-treated with IFNγ in immunocompetent female mice when PARP14 is pharmacologically inhibited or knocked down, while the presence of regulatory T cells is decreased, leading to restoration of α-PD-1 sensitivity. Finally, we determine that tumours which spontaneously relapse in immunocompetent female mice following α-PD-1 therapy upregulate IFNγ signalling and can also be re-sensitised upon receiving PARP14 inhibitor treatment, establishing PARP14 as an actionable target to reverse IFNγ-driven ICBT resistance.
Collapse
Affiliation(s)
- Chun Wai Wong
- Faculty of Biology, Medicine and Health, The University of Manchester, Manchester, M13 9PT, UK
- Lydia Becker Institute of Immunology, The University of Manchester, Manchester, M13 9PT, UK
| | - Christos Evangelou
- Faculty of Biology, Medicine and Health, The University of Manchester, Manchester, M13 9PT, UK
- Lydia Becker Institute of Immunology, The University of Manchester, Manchester, M13 9PT, UK
| | - Kieran N Sefton
- Faculty of Biology, Medicine and Health, The University of Manchester, Manchester, M13 9PT, UK
- Lydia Becker Institute of Immunology, The University of Manchester, Manchester, M13 9PT, UK
| | - Rotem Leshem
- Faculty of Biology, Medicine and Health, The University of Manchester, Manchester, M13 9PT, UK
- Lydia Becker Institute of Immunology, The University of Manchester, Manchester, M13 9PT, UK
| | - Wei Zhang
- Faculty of Biology, Medicine and Health, The University of Manchester, Manchester, M13 9PT, UK
| | - Vishaka Gopalan
- Cancer Data Science Laboratory, National Cancer Institute, Bethesda, MD, 20814, USA
| | - Sorayut Chattrakarn
- Faculty of Biology, Medicine and Health, The University of Manchester, Manchester, M13 9PT, UK
- Lydia Becker Institute of Immunology, The University of Manchester, Manchester, M13 9PT, UK
| | - Macarena Lucia Fernandez Carro
- Faculty of Biology, Medicine and Health, The University of Manchester, Manchester, M13 9PT, UK
- Lydia Becker Institute of Immunology, The University of Manchester, Manchester, M13 9PT, UK
| | - Erez Uzuner
- Faculty of Biology, Medicine and Health, The University of Manchester, Manchester, M13 9PT, UK
- Lydia Becker Institute of Immunology, The University of Manchester, Manchester, M13 9PT, UK
| | - Holly Mole
- Faculty of Biology, Medicine and Health, The University of Manchester, Manchester, M13 9PT, UK
| | - Daniel J Wilcock
- Faculty of Biology, Medicine and Health, The University of Manchester, Manchester, M13 9PT, UK
| | - Michael P Smith
- Faculty of Biology, Medicine and Health, The University of Manchester, Manchester, M13 9PT, UK
| | - Kleita Sergiou
- Faculty of Biology, Medicine and Health, The University of Manchester, Manchester, M13 9PT, UK
| | - Brian A Telfer
- Faculty of Biology, Medicine and Health, The University of Manchester, Manchester, M13 9PT, UK
| | - Dervla T Isaac
- Ribon Therapeutics Inc., 35 Cambridge Park Drive, Suite 300, Cambridge, MA, 02140, USA
| | - Chang Liu
- Ribon Therapeutics Inc., 35 Cambridge Park Drive, Suite 300, Cambridge, MA, 02140, USA
| | - Nicholas R Perl
- Ribon Therapeutics Inc., 35 Cambridge Park Drive, Suite 300, Cambridge, MA, 02140, USA
| | - Kerrie Marie
- Faculty of Biology, Medicine and Health, The University of Manchester, Manchester, M13 9PT, UK
| | - Paul Lorigan
- Faculty of Biology, Medicine and Health, The University of Manchester, Manchester, M13 9PT, UK
- Department of Medical Oncology, The Christie NHS Foundation Trust, Wilmslow Road, Withington, Manchester, M20 4BX, UK
| | - Kaye J Williams
- Faculty of Biology, Medicine and Health, The University of Manchester, Manchester, M13 9PT, UK
| | | | - Raghavendar T Nagaraju
- Faculty of Biology, Medicine and Health, The University of Manchester, Manchester, M13 9PT, UK
- Colorectal and Peritoneal Oncology Centre, The Christie NHS Foundation Trust, Wilmslow Road, Withington, Manchester, UK
| | - Mario Niepel
- Ribon Therapeutics Inc., 35 Cambridge Park Drive, Suite 300, Cambridge, MA, 02140, USA
| | - Adam F L Hurlstone
- Faculty of Biology, Medicine and Health, The University of Manchester, Manchester, M13 9PT, UK.
- Lydia Becker Institute of Immunology, The University of Manchester, Manchester, M13 9PT, UK.
| |
Collapse
|
26
|
Đukić N, Strømland Ø, Elsborg JD, Munnur D, Zhu K, Schuller M, Chatrin C, Kar P, Duma L, Suyari O, Rack JGM, Baretić D, Crudgington DRK, Groslambert J, Fowler G, Wijngaarden S, Prokhorova E, Rehwinkel J, Schüler H, Filippov DV, Sanyal S, Ahel D, Nielsen ML, Smith R, Ahel I. PARP14 is a PARP with both ADP-ribosyl transferase and hydrolase activities. SCIENCE ADVANCES 2023; 9:eadi2687. [PMID: 37703374 PMCID: PMC10499325 DOI: 10.1126/sciadv.adi2687] [Citation(s) in RCA: 23] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/14/2023] [Accepted: 08/10/2023] [Indexed: 09/15/2023]
Abstract
PARP14 is a mono-ADP-ribosyl transferase involved in the control of immunity, transcription, and DNA replication stress management. However, little is known about the ADP-ribosylation activity of PARP14, including its substrate specificity or how PARP14-dependent ADP-ribosylation is reversed. We show that PARP14 is a dual-function enzyme with both ADP-ribosyl transferase and hydrolase activity acting on both protein and nucleic acid substrates. In particular, we show that the PARP14 macrodomain 1 is an active ADP-ribosyl hydrolase. We also demonstrate hydrolytic activity for the first macrodomain of PARP9. We reveal that expression of a PARP14 mutant with the inactivated macrodomain 1 results in a marked increase in mono(ADP-ribosyl)ation of proteins in human cells, including PARP14 itself and antiviral PARP13, and displays specific cellular phenotypes. Moreover, we demonstrate that the closely related hydrolytically active macrodomain of SARS2 Nsp3, Mac1, efficiently reverses PARP14 ADP-ribosylation in vitro and in cells, supporting the evolution of viral macrodomains to counteract PARP14-mediated antiviral response.
Collapse
Affiliation(s)
- Nina Đukić
- Sir William Dunn School of Pathology, University of Oxford, Oxford OX1 3RE, UK
| | - Øyvind Strømland
- Sir William Dunn School of Pathology, University of Oxford, Oxford OX1 3RE, UK
- Department of Biomedicine, University of Bergen, 5020 Bergen, Norway
| | - Jonas Damgaard Elsborg
- Proteomics Program, Novo Nordisk Foundation Center for Protein Research, Faculty of Health and Medical Sciences, University of Copenhagen, Blegdamsvej 3B, 2200 Copenhagen, Denmark
| | - Deeksha Munnur
- Sir William Dunn School of Pathology, University of Oxford, Oxford OX1 3RE, UK
| | - Kang Zhu
- Sir William Dunn School of Pathology, University of Oxford, Oxford OX1 3RE, UK
| | - Marion Schuller
- Sir William Dunn School of Pathology, University of Oxford, Oxford OX1 3RE, UK
| | - Chatrin Chatrin
- Sir William Dunn School of Pathology, University of Oxford, Oxford OX1 3RE, UK
| | - Pulak Kar
- Sir William Dunn School of Pathology, University of Oxford, Oxford OX1 3RE, UK
| | - Lena Duma
- Sir William Dunn School of Pathology, University of Oxford, Oxford OX1 3RE, UK
| | - Osamu Suyari
- Sir William Dunn School of Pathology, University of Oxford, Oxford OX1 3RE, UK
| | - Johannes Gregor Matthias Rack
- Sir William Dunn School of Pathology, University of Oxford, Oxford OX1 3RE, UK
- MRC Centre for Medical Mycology, University of Exeter, Geoffrey Pope Building, Stocker Road, Exeter EX4 4QD, UK
| | - Domagoj Baretić
- Sir William Dunn School of Pathology, University of Oxford, Oxford OX1 3RE, UK
| | | | | | - Gerissa Fowler
- Medical Research Council Human Immunology Unit, Medical Research Council Weatherall Institute of Molecular Medicine, Radcliffe Department of Medicine, University of Oxford, Oxford OX3 9DS, UK
| | - Sven Wijngaarden
- Leiden Institute of Chemistry, Leiden University, Einsteinweg 55, 2333 CC Leiden, Netherlands
| | - Evgeniia Prokhorova
- Sir William Dunn School of Pathology, University of Oxford, Oxford OX1 3RE, UK
| | - Jan Rehwinkel
- Medical Research Council Human Immunology Unit, Medical Research Council Weatherall Institute of Molecular Medicine, Radcliffe Department of Medicine, University of Oxford, Oxford OX3 9DS, UK
| | - Herwig Schüler
- Center for Molecular Protein Science, Department of Chemistry, Lund University, 22100 Lund, Sweden
| | - Dmitri V. Filippov
- Leiden Institute of Chemistry, Leiden University, Einsteinweg 55, 2333 CC Leiden, Netherlands
| | - Sumana Sanyal
- Sir William Dunn School of Pathology, University of Oxford, Oxford OX1 3RE, UK
| | - Dragana Ahel
- Sir William Dunn School of Pathology, University of Oxford, Oxford OX1 3RE, UK
| | - Michael L Nielsen
- Proteomics Program, Novo Nordisk Foundation Center for Protein Research, Faculty of Health and Medical Sciences, University of Copenhagen, Blegdamsvej 3B, 2200 Copenhagen, Denmark
| | - Rebecca Smith
- Sir William Dunn School of Pathology, University of Oxford, Oxford OX1 3RE, UK
| | - Ivan Ahel
- Sir William Dunn School of Pathology, University of Oxford, Oxford OX1 3RE, UK
| |
Collapse
|
27
|
Pak JN, Lee HJ, Sim DY, Park JE, Ahn CH, Park SY, Khil JH, Shim B, Kim B, Kim SH. Anti-Warburg effect via generation of ROS and inhibition of PKM2/β-catenin mediates apoptosis of lambertianic acid in prostate cancer cells. Phytother Res 2023; 37:4224-4235. [PMID: 37235481 DOI: 10.1002/ptr.7903] [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: 02/06/2023] [Revised: 04/24/2023] [Accepted: 05/17/2023] [Indexed: 05/28/2023]
Abstract
To elucidate the underlying antitumor mechanism of lambertianic acid (LA) derived from Pinus koraiensis, the role of cancer metabolism related molecules was investigated in the apoptotic effect of LA in DU145 and PC3 prostate cancer cells. MTT assay for cytotoxicity, RNA interference, cell cycle analysis for sub G1 population, nuclear and cytoplasmic extraction, lactate, Glucose and ATP assay by ELISA, Measurement of reactive oxygen species (ROS) generation, Western blotting, and immunoprecipitation assay were conducted in DU145 and PC3 prostate cancer cells. Herein LA exerted cytotoxicity, increased sub G1 population and attenuated the expression of pro-Caspase3 and pro-poly (ADP-ribose) polymerase (pro-PARP) in DU145 and PC3 cells. Also, LA reduced the expression of lactate dehydrogenase A (LDHA), glycolytic enzymes such as hexokinase 2 and pyruvate kinase M2 (PKM2) with reduced production of lactate in DU145 and PC3 cells. Notably, LA decreased phosphorylation of PKM2 on Tyr105 and inhibited the expression of p-STAT3, cyclin D1, C-Myc, β-catenin, and p-GSK3β with the decrease of nuclear translocation of p-PKM2. Furthermore, LA disturbed the binding of p-PKM2 and β-catenin in DU145 cells, which was supported by Spearman coefficient (0.0463) of cBioportal database. Furthermore, LA generated ROS in DU145 and PC3 cells, while ROS scavenger NAC (N-acetyl L-cysteine) blocked the ability of LA to reduce p-PKM2, PKM2, β-catenin, LDHA, and pro-caspase3 in DU145 cells. Taken together, these findings provide evidence that LA induces apoptosis via ROS generation and inhibition of PKM2/β-catenin signaling in prostate cancer cells.
Collapse
Affiliation(s)
- Ji-Na Pak
- College of Korean Medicine, Kyung Hee University, Seoul, Republic of Korea
| | - Hyo-Jung Lee
- College of Korean Medicine, Kyung Hee University, Seoul, Republic of Korea
| | - Deok Yong Sim
- College of Korean Medicine, Kyung Hee University, Seoul, Republic of Korea
| | - Ji Eon Park
- College of Korean Medicine, Kyung Hee University, Seoul, Republic of Korea
| | - Chi-Hoon Ahn
- College of Korean Medicine, Kyung Hee University, Seoul, Republic of Korea
| | - Su-Yeon Park
- College of Korean Medicine, Kyung Hee University, Seoul, Republic of Korea
| | - Jae-Ho Khil
- Institute of Sports Science, Kyung Hee University, Yongin, Republic of Korea
| | - Bumsang Shim
- College of Korean Medicine, Kyung Hee University, Seoul, Republic of Korea
| | - Bonglee Kim
- College of Korean Medicine, Kyung Hee University, Seoul, Republic of Korea
| | - Sung-Hoon Kim
- College of Korean Medicine, Kyung Hee University, Seoul, Republic of Korea
| |
Collapse
|
28
|
Nizi MG, Sarnari C, Tabarrini O. Privileged Scaffolds for Potent and Specific Inhibitors of Mono-ADP-Ribosylating PARPs. Molecules 2023; 28:5849. [PMID: 37570820 PMCID: PMC10420676 DOI: 10.3390/molecules28155849] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2023] [Revised: 07/31/2023] [Accepted: 08/01/2023] [Indexed: 08/13/2023] Open
Abstract
The identification of new targets to address unmet medical needs, better in a personalized way, is an urgent necessity. The introduction of PARP1 inhibitors into therapy, almost ten years ago, has represented a step forward this need being an innovate cancer treatment through a precision medicine approach. The PARP family consists of 17 members of which PARP1 that works by poly-ADP ribosylating the substrate is the sole enzyme so far exploited as therapeutic target. Most of the other members are mono-ADP-ribosylating (mono-ARTs) enzymes, and recent studies have deciphered their pathophysiological roles which appear to be very extensive with various potential therapeutic applications. In parallel, a handful of mono-ARTs inhibitors emerged that have been collected in a perspective on 2022. After that, additional very interesting compounds were identified highlighting the hot-topic nature of this research field and prompting an update. From the present review, where we have reported only mono-ARTs inhibitors endowed with the appropriate profile of pharmacological tools or drug candidate, four privileged scaffolds clearly stood out that constitute the basis for further drug discovery campaigns.
Collapse
Affiliation(s)
- Maria Giulia Nizi
- Department of Pharmaceutical Sciences, University of Perugia, 06123 Perugia, Italy;
| | | | - Oriana Tabarrini
- Department of Pharmaceutical Sciences, University of Perugia, 06123 Perugia, Italy;
| |
Collapse
|
29
|
Dong Y, Liu Y, Liu X, Ma H, Liu Y, Lv G, Niu J. Multi-omics profiling of primary hepatic stellate cells from advanced liver fibrosis patients reveals distinctive molecular signatures. J Gastroenterol Hepatol 2023; 38:1416-1425. [PMID: 37226284 DOI: 10.1111/jgh.16221] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/19/2023] [Revised: 04/30/2023] [Accepted: 05/06/2023] [Indexed: 05/26/2023]
Abstract
BACKGROUND AND AIM Hepatic fibrosis is a common pathogenic outcome of almost all chronic liver diseases and a growing public health problem globally. However, the key genes or proteins driving liver fibrosis and cirrhosis are not well understood. We aimed to identify novel hepatic fibrosis genes of human primary hepatic stellate cells (HSCs). METHODS Human primary HSCs were isolated from surgically resected advanced fibrosis liver tissues (n = 6) and surgical resection of normal liver tissue around hemangioma (n = 5). Differences in the expression levels of mRNA and proteins from HSCs in advanced fibrosis group and the control group were analyzed using RNA sequencing and mass spectrometry as transcriptomic and proteomic approaches. The obtained biomarkers were further validated through real-time quantitative polymerase chain reaction (RT-qPCR), immunofluorescence, and Western blot. RESULTS A total of 2156 transcripts and 711 proteins were found to be differently expressed between the advanced fibrosis group and the control group patients. The Venn diagram shows that a total of 96 upregulated molecules are overlapped in both the transcriptomic and proteomic datasets. Gene Ontology enrichment analysis and Kyoto Encyclopedia of Genes and Genomes analysis indicated that those overlapped genes were mainly involved in wound healing, cell adhesion regulation, and actin binding, which reflects the major biological conversions in liver cirrhosis process. Pyruvate kinase M2 and EH domain-containing 2 were identified as potential new markers for advanced liver cirrhosis, which have been validated in primary human HSCs and in vitro cellular hepatic fibrosis model Lieming Xu-2 (LX-2) cells. CONCLUSIONS Our results revealed the major transcriptomic and proteomic changes during liver cirrhosis process and identified new biomarkers and potential therapeutic targets for advanced liver fibrosis.
Collapse
Affiliation(s)
- Yutong Dong
- Department of Hepatology, Center of Infectious Diseases and Pathogen Biology, The First Hospital of Jilin University, Changchun, China
- Key Laboratory of Zoonosis Research, Ministry of Education, Jilin University, Changchun, China
| | - Yuwei Liu
- Department of Hepatology, Center of Infectious Diseases and Pathogen Biology, The First Hospital of Jilin University, Changchun, China
| | - Xu Liu
- Department of Hepatology, Center of Infectious Diseases and Pathogen Biology, The First Hospital of Jilin University, Changchun, China
| | - Heming Ma
- Department of Hepatology, Center of Infectious Diseases and Pathogen Biology, The First Hospital of Jilin University, Changchun, China
| | - Yahui Liu
- Department of Hepatobiliary and Pancreatic Surgery, General Surgery Center, The First Hospital of Jilin University, Changchun, China
| | - Guoyue Lv
- Department of Hepatobiliary and Pancreatic Surgery, General Surgery Center, The First Hospital of Jilin University, Changchun, China
| | - Junqi Niu
- Department of Hepatology, Center of Infectious Diseases and Pathogen Biology, The First Hospital of Jilin University, Changchun, China
- Key Laboratory of Zoonosis Research, Ministry of Education, Jilin University, Changchun, China
| |
Collapse
|
30
|
Arévalo CM, Cruz-Rodriguez N, Quijano S, Fiorentino S. Plant-derived extracts and metabolic modulation in leukemia: a promising approach to overcome treatment resistance. Front Mol Biosci 2023; 10:1229760. [PMID: 37520325 PMCID: PMC10382028 DOI: 10.3389/fmolb.2023.1229760] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2023] [Accepted: 06/30/2023] [Indexed: 08/01/2023] Open
Abstract
Leukemic cells acquire complex and often multifactorial mechanisms of resistance to treatment, including various metabolic alterations. Although the use of metabolic modulators has been proposed for several decades, their use in clinical practice has not been established. Natural products, the so-called botanical drugs, are capable of regulating tumor metabolism, particularly in hematopoietic tumors, which could partly explain the biological activity attributed to them for a long time. This review addresses the most recent findings relating to metabolic reprogramming-Mainly in the glycolytic pathway and mitochondrial activity-Of leukemic cells and its role in the generation of resistance to conventional treatments, the modulation of the tumor microenvironment, and the evasion of immune response. In turn, it describes how the modulation of metabolism by plant-derived extracts can counteract resistance to chemotherapy in this tumor model and contribute to the activation of the antitumor immune system.
Collapse
Affiliation(s)
- Cindy Mayerli Arévalo
- Grupo de Inmunobiología y Biología Celular, Facultad de Ciencias, Pontificia Universidad Javeriana, Bogotá, Colombia
| | | | - Sandra Quijano
- Grupo de Inmunobiología y Biología Celular, Facultad de Ciencias, Pontificia Universidad Javeriana, Bogotá, Colombia
| | - Susana Fiorentino
- Grupo de Inmunobiología y Biología Celular, Facultad de Ciencias, Pontificia Universidad Javeriana, Bogotá, Colombia
| |
Collapse
|
31
|
Fernandez-Checa JC, Torres S, Garcia-Ruiz C. Hilpda, A New Player In Nash-Driven Hcc, Links Hypoxia Signaling With Ceramide Synthesis. J Hepatol 2023:S0168-8278(23)00337-9. [PMID: 37207912 DOI: 10.1016/j.jhep.2023.05.009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/26/2023] [Accepted: 05/02/2023] [Indexed: 05/21/2023]
Affiliation(s)
- Jose C Fernandez-Checa
- Department of Cell Death and Proliferation, Institute of Biomedical Research of Barcelona (IIBB), CSIC, Barcelona, Spain; Liver Unit, Hospital Clinic I Provincial de Barcelona, Barcelona, Spain; Instituto de Investigaciones Biomédicas August Pi I Sunyer (IDIBAPS), Barcelona; CIBEREHD, Madrid, Spain; Department of Medicine, Keck School of Division of Gastrointestinal and Liver Disease,Medicine, University of Southern California, Los Angeles, CA, United States.
| | - Sandra Torres
- Department of Cell Death and Proliferation, Institute of Biomedical Research of Barcelona (IIBB), CSIC, Barcelona, Spain; Instituto de Investigaciones Biomédicas August Pi I Sunyer (IDIBAPS), Barcelona; CIBEREHD, Madrid, Spain
| | - Carmen Garcia-Ruiz
- Department of Cell Death and Proliferation, Institute of Biomedical Research of Barcelona (IIBB), CSIC, Barcelona, Spain; Liver Unit, Hospital Clinic I Provincial de Barcelona, Barcelona, Spain; Instituto de Investigaciones Biomédicas August Pi I Sunyer (IDIBAPS), Barcelona; CIBEREHD, Madrid, Spain; Department of Medicine, Keck School of Division of Gastrointestinal and Liver Disease,Medicine, University of Southern California, Los Angeles, CA, United States.
| |
Collapse
|
32
|
Dong F, Li H, Liu L, Yao LL, Wang J, Xiang D, Ma J, Zhang G, Zhang S, Li J, Jiang SH, Hu X, Chen J, Bao Z. ACE2 negatively regulates the Warburg effect and suppresses hepatocellular carcinoma progression via reducing ROS-HIF1α activity. Int J Biol Sci 2023; 19:2613-2629. [PMID: 37215979 PMCID: PMC10197896 DOI: 10.7150/ijbs.81498] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2022] [Accepted: 04/30/2023] [Indexed: 05/24/2023] Open
Abstract
Aerobic glycolysis has pleiotropic roles in the pathogenesis of hepatocellular carcinoma (HCC). Emerging studies revealed key promoters of aerobic glycolysis, however, little is known about its negative regulators in HCC. In this study, an integrative analysis identifies a repertoire of differentially expressed genes (DNASE1L3, SLC22A1, ACE2, CES3, CCL14, GYS2, ADH4, and CFHR3) that are inversely associated with the glycolytic phenotype in HCC. ACE2, a member of the rennin-angiotensin system, is revealed to be downregulated in HCC and predicts a poor prognosis. ACE2 overexpression significantly inhibits the glycolytic flux as evidenced by reduced glucose uptake, lactate release, extracellular acidification rate, and the expression of glycolytic genes. Opposite results are noticed in loss-of-function studies. Mechanistically, ACE2 metabolizes Ang II to Ang-(1-7), which activates Mas receptor and leads to the phosphorylation of Src homology 2-containing inositol phosphatase 2 (SHP-2). SHP2 activation further blocks reactive oxygen species (ROS)-HIF1α signaling. Addition of Ang-(1-7) or the antioxidant N-acetylcysteine compromises in vivo additive tumor growth and aerobic glycolysis induced by ACE2 knockdown. Moreover, growth advantages afforded by ACE2 knockdown are largely glycolysis-dependent. In clinical settings, a close link between ACE2 expression and HIF1α or the phosphorated level of SHP2 is found. Overexpression of ACE2 significantly retards tumor growth in patient-derived xenograft model. Collectively, our findings suggest that ACE2 is a negative glycolytic regulator, and targeting the ACE2/Ang-(1-7)/Mas receptor/ROS/HIF1α axis may be a promising therapeutic strategy for HCC treatment.
Collapse
Affiliation(s)
- Fangyuan Dong
- Department of Gastroenterology, Huadong Hospital, Shanghai Medical College, Fudan University, Shanghai 200040, P.R. China
- Shanghai Key Laboratory of Clinical Geriatric Medicine, Shanghai 200040, P.R. China
- National Clinical Research Center for Aging and Medicine, Shanghai 200040, P.R. China
- Department of Geriatrics, Huadong Hospital, Shanghai Medical College, Fudan University Shanghai 200040, P.R. China
| | - Hui Li
- State Key Laboratory of Oncogenes and Related Genes, Shanghai Cancer Institute, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200240, P.R. China
| | - Limin Liu
- Department of Oral pathology, Ninth People's Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200011, P.R. China
| | - Lin-Li Yao
- State Key Laboratory of Oncogenes and Related Genes, Shanghai Cancer Institute, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200240, P.R. China
| | - Jiaofeng Wang
- Shanghai Key Laboratory of Clinical Geriatric Medicine, Shanghai 200040, P.R. China
- National Clinical Research Center for Aging and Medicine, Shanghai 200040, P.R. China
- Department of Geriatrics, Huadong Hospital, Shanghai Medical College, Fudan University Shanghai 200040, P.R. China
| | - Danni Xiang
- Department of Gastroenterology, Huadong Hospital, Shanghai Medical College, Fudan University, Shanghai 200040, P.R. China
| | - Jianxia Ma
- Department of Gastroenterology, Huadong Hospital, Shanghai Medical College, Fudan University, Shanghai 200040, P.R. China
| | - Gansheng Zhang
- Department of Gastroenterology, Huadong Hospital, Shanghai Medical College, Fudan University, Shanghai 200040, P.R. China
| | - Shan Zhang
- State Key Laboratory of Oncogenes and Related Genes, Shanghai Cancer Institute, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200240, P.R. China
| | - Jun Li
- State Key Laboratory of Oncogenes and Related Genes, Shanghai Cancer Institute, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200240, P.R. China
| | - Shu-Heng Jiang
- State Key Laboratory of Oncogenes and Related Genes, Shanghai Cancer Institute, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200240, P.R. China
| | - Xiaona Hu
- Department of Gastroenterology, Huadong Hospital, Shanghai Medical College, Fudan University, Shanghai 200040, P.R. China
- Shanghai Key Laboratory of Clinical Geriatric Medicine, Shanghai 200040, P.R. China
- National Clinical Research Center for Aging and Medicine, Shanghai 200040, P.R. China
- Department of Geriatrics, Huadong Hospital, Shanghai Medical College, Fudan University Shanghai 200040, P.R. China
| | - Jie Chen
- Department of Gastroenterology, Huadong Hospital, Shanghai Medical College, Fudan University, Shanghai 200040, P.R. China
- Shanghai Key Laboratory of Clinical Geriatric Medicine, Shanghai 200040, P.R. China
- National Clinical Research Center for Aging and Medicine, Shanghai 200040, P.R. China
- Department of Geriatrics, Huadong Hospital, Shanghai Medical College, Fudan University Shanghai 200040, P.R. China
| | - Zhijun Bao
- Department of Gastroenterology, Huadong Hospital, Shanghai Medical College, Fudan University, Shanghai 200040, P.R. China
- Shanghai Key Laboratory of Clinical Geriatric Medicine, Shanghai 200040, P.R. China
- National Clinical Research Center for Aging and Medicine, Shanghai 200040, P.R. China
- Department of Geriatrics, Huadong Hospital, Shanghai Medical College, Fudan University Shanghai 200040, P.R. China
| |
Collapse
|
33
|
Ye Y, Wang M, Wang G, Mai Z, Zhou B, Han Y, Zhuang J, Xia W. lncRNA miR4458HG modulates hepatocellular carcinoma progression by activating m6A-dependent glycolysis and promoting the polarization of tumor-associated macrophages. Cell Mol Life Sci 2023; 80:99. [PMID: 36933158 PMCID: PMC11072995 DOI: 10.1007/s00018-023-04741-8] [Citation(s) in RCA: 24] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2022] [Revised: 02/08/2023] [Accepted: 03/01/2023] [Indexed: 03/19/2023]
Abstract
Long non-coding RNAs (lncRNAs) play significant roles in different biological functions of cancers. However, their function in the metabolism of glucose in patients with human hepatocellular carcinoma (HCC) remains largely unknown. In this study, HCC and paired intact liver tissues were utilized to examine the miR4458HG expression using qRT-PCR and human HCC cell lines to examine cell proliferation, colony formation, and glycolysis after transfection of siRNAs targeting miR4458HG or miR4458HG vectors. The molecular mechanism of miR4458HG was clarified through in situ hybridization, Western blotting, qRT-PCR, RNA pull-down, and RNA immunoprecipitation analysis. The results showed that the miR4458HG affected HCC cell proliferation, activated the glycolysis pathway, and promoted the polarization of tumor-associated macrophage in vitro and in vivo models. Mechanistically, miR4458HG bound IGF2BP2 (a key RNA m6A reader) and facilitated IGF2BP2-mediated target mRNA stability, including HK2 and SLC2A1 (GLUT1), and consequently altered HCC glycolysis and tumor cell physiology. At the same time, HCC-derived miR4458HG could be wrapped in the exosomes and promoted the polarization of tumor-associated macrophage by increasing ARG1 expression. Hence, miR4458HG is oncogenic in nature among patients with HCC. To develop an effective treatment strategy of HCC patients presenting with high glucose metabolism, physicians should focus on miR4458HG and its pathway.
Collapse
Affiliation(s)
- Ying Ye
- Central Laboratory, The Seventh People's Hospital of Shanghai University of Traditional Chinese Medicine, Shanghai, 200137, China
| | - Menghan Wang
- Department of Nuclear Medicine, The Seventh People's Hospital, Shanghai University of Traditional Chinese Medicine, 358 Datong Rd, Pudong New Area, Shanghai, 200137, China
| | - Guoyu Wang
- Department of Nuclear Medicine, The Seventh People's Hospital, Shanghai University of Traditional Chinese Medicine, 358 Datong Rd, Pudong New Area, Shanghai, 200137, China
| | - Zhongchao Mai
- Department of Nuclear Medicine, The Seventh People's Hospital, Shanghai University of Traditional Chinese Medicine, 358 Datong Rd, Pudong New Area, Shanghai, 200137, China
| | - Borong Zhou
- Department of Nuclear Medicine, The Seventh People's Hospital, Shanghai University of Traditional Chinese Medicine, 358 Datong Rd, Pudong New Area, Shanghai, 200137, China
| | - Yang Han
- Department of Nuclear Medicine, The Seventh People's Hospital, Shanghai University of Traditional Chinese Medicine, 358 Datong Rd, Pudong New Area, Shanghai, 200137, China
| | - Juhua Zhuang
- Department of Nuclear Medicine, The Seventh People's Hospital, Shanghai University of Traditional Chinese Medicine, 358 Datong Rd, Pudong New Area, Shanghai, 200137, China
| | - Wei Xia
- Department of Nuclear Medicine, The Seventh People's Hospital, Shanghai University of Traditional Chinese Medicine, 358 Datong Rd, Pudong New Area, Shanghai, 200137, China.
| |
Collapse
|
34
|
Dhoonmoon A, Nicolae CM. Mono-ADP-ribosylation by PARP10 and PARP14 in genome stability. NAR Cancer 2023; 5:zcad009. [PMID: 36814782 PMCID: PMC9940457 DOI: 10.1093/narcan/zcad009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2022] [Revised: 01/09/2023] [Accepted: 02/02/2023] [Indexed: 02/22/2023] Open
Abstract
ADP-ribosylation is a post-translational modification involved in a variety of processes including DNA damage repair, transcriptional regulation, and cellular proliferation. Depending on the number of ADP moieties transferred to target proteins, ADP-ribosylation can be classified either as mono-ADP-ribosylation (MARylation) or poly-ADP-ribosylation (PARylation). This post-translational modification is catalyzed by enzymes known as ADP-ribosyltransferases (ARTs), which include the poly (ADP-ribose)-polymerase (PARP) superfamily of proteins. Certain members of the PARP family including PARP1 and PARP2 have been extensively studied and assessed as therapeutic targets. However, the other members of the PARP family of protein are not as well studied but have gained attention in recent years given findings suggesting their roles in an increasing number of cellular processes. Among these other members are PARP10 and PARP14, which have gradually emerged as key players in maintenance of genomic stability and carcinogenesis. PARP10 and PARP14 catalyze the transfer of a single ADP moiety to target proteins. Here, we summarize the current knowledge on MARylation in DNA repair and cancer, focusing on PARP10 and PARP14. We highlight the roles of PARP10 and PARP14 in cancer progression and response to chemotherapeutics and briefly discuss currently known PARP10 and PARP14 inhibitors.
Collapse
Affiliation(s)
- Ashna Dhoonmoon
- Department of Biochemistry and Molecular Biology, The Pennsylvania State University College of Medicine, Hershey, PA 17033, USA
| | - Claudia M Nicolae
- Department of Biochemistry and Molecular Biology, The Pennsylvania State University College of Medicine, Hershey, PA 17033, USA
| |
Collapse
|
35
|
Wang J, Cheng Q, Zhang Y, Hong C, Liu J, Liu X, Chang J. PARP16-Mediated Stabilization of Amyloid Precursor Protein mRNA Exacerbates Alzheimer's Disease Pathogenesis. Aging Dis 2023:AD.2023.0119. [PMID: 37163422 PMCID: PMC10389827 DOI: 10.14336/ad.2023.0119] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2022] [Accepted: 01/19/2023] [Indexed: 05/12/2023] Open
Abstract
The accumulation and deposition of beta-amyloid (Aβ) are key neuropathological hallmarks of Alzheimer's disease (AD). PARP16, a Poly(ADP-ribose) polymerase, is a known tail-anchored endoplasmic reticulum (ER) transmembrane protein that transduces ER stress during pathological processes. Here, we found that PARP16 was significantly increased in the hippocampi and cortices of APPswe/PS1dE9 (APP/PS1) mice and hippocampal neuronal HT22 cells exposed to Aβ, suggesting a positive correlation between the progression of AD pathology and the overexpression of PARP16. To define the effect of PARP16 on AD progression, adeno-associated virus mediated-PARP16 knockdown was used in APP/PS1 mice to investigate the role of PARP16 in spatial memory, amyloid burden, and neuroinflammation. Knockdown of PARP16 partly attenuated impaired spatial memory, as indicated by the Morris water maze test, and decreased amyloid deposition, neuronal apoptosis, and the production of inflammatory cytokines in the brains of APP/PS1 mice. In vitro experiments demonstrated that the knockdown of PARP16 expression rescued neuronal damage and ER stress triggered by Aβ. Furthermore, we discovered that intracellular PARP16 acts as an RNA-binding protein that regulates the mRNA stability of amyloid precursor protein (APP) and protects targeted APP from degradation, thereby increasing APP levels and AD pathology. Our findings revealed an unanticipated role of PARP16 in the pathogenesis of AD, and at least in part, its association with increased APP mRNA stability.
Collapse
Affiliation(s)
- Jinghuan Wang
- Pharmacophenomics Laboratory, Human Phenome Institute, Fudan University, Shanghai 201203, China
| | - Qianwen Cheng
- Pharmacophenomics Laboratory, Human Phenome Institute, Fudan University, Shanghai 201203, China
| | - Yuyu Zhang
- Pharmacophenomics Laboratory, Human Phenome Institute, Fudan University, Shanghai 201203, China
| | - Chen Hong
- Pharmacophenomics Laboratory, Human Phenome Institute, Fudan University, Shanghai 201203, China
| | - Jiayao Liu
- Pharmacophenomics Laboratory, Human Phenome Institute, Fudan University, Shanghai 201203, China
| | - Xinhua Liu
- Pharmacophenomics Laboratory, Human Phenome Institute, Fudan University, Shanghai 201203, China
| | - Jun Chang
- Pharmacophenomics Laboratory, Human Phenome Institute, Fudan University, Shanghai 201203, China
| |
Collapse
|
36
|
Kaci FN, Lepore A, Papa S, Bubici C. Screening Kinase-Dependent Phosphorylation of Key Metabolic Reprogramming Regulators. Methods Mol Biol 2023; 2675:205-218. [PMID: 37258766 DOI: 10.1007/978-1-0716-3247-5_16] [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] [Indexed: 06/02/2023]
Abstract
Aerobic glycolysis has been commonly linked to cell proliferation, especially in cancer cells where it serves to generate sufficient energy and biosynthesis of new cell constituents needed for cell growth and division. The M2 isoform of pyruvate kinase (PKM2) catalyzes the last reaction of the glycolytic process. PKM2 promotes the transfer of a phosphate group from phosphoenolpyruvate (PEP) to ADP, generating ATP and releasing pyruvate. This rate-limiting reaction relies therefore on the enzymatic activity of PKM2. The switching between the high- and low-activity states of PKM2 is subjected to a combination of allosteric mechanisms and fine-tuned regulation by oncogenes and tumor suppressor genes. These regulatory mechanisms involve primarily post-translational modifications of PKM2. Recent findings suggest that phosphorylation contributes to the regulation of PKM2 activity.Here, we describe an in vitro kinase assay we used to assess PKM2 phosphorylation by c-Jun N-terminal kinase (JNK), a master regulator of apoptosis, cell proliferation, and differentiation. While the use of phospho-specific antibodies gives information in terms of measuring the effects of a given kinase on its substrate, specific antibodies for newly identified phospho-groups are not readily available. The in vitro kinase assay allows the immediate measuring of phosphorylation of any substrate of interest. Although there are several options that do not use radioactive materials, we continue to rely on this biochemical method for robust quantitation of results. More interestingly, this protocol can be easily adapted to measure the activity of other kinases by using their specific substrates.
Collapse
Affiliation(s)
- Fatma Necmiye Kaci
- Leeds Institute of Medical Research, St. James's University Hospital, University of Leeds, Leeds, UK
| | - Alessio Lepore
- Leeds Institute of Medical Research, St. James's University Hospital, University of Leeds, Leeds, UK
| | - Salvatore Papa
- Leeds Institute of Medical Research, St. James's University Hospital, University of Leeds, Leeds, UK
| | - Concetta Bubici
- Department of Life Sciences, Center for Genome Engineering and Maintenance, College of Health, Medicine and Life Sciences, Brunel University London, London, UK.
| |
Collapse
|
37
|
Lepore A, Kaci FN, Bubici C, Papa S. An Integrated Methodology to Quantify the Glycolytic Stress in Plasma Cell Myeloma in Response to Cytotoxic Drugs. Methods Mol Biol 2023; 2675:285-296. [PMID: 37258771 DOI: 10.1007/978-1-0716-3247-5_21] [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] [Indexed: 06/02/2023]
Abstract
Multiple myeloma (MM) is an incurable plasma cell malignancy primarily localized within the bone marrow (BM). Myeloma plasma cells, like many other cancer cells, change their metabolism in response to internal and external stimuli. The main metabolic alterations of MM cells include deregulated glycolysis (commonly associated with enhanced uptake and utilization of glucose), lipid metabolism dysregulation, as well as deregulated mitochondrial respiration (commonly associated with the deregulated formation of reactive oxygen species). Over the past decade, the discovery of novel methodologies and the commercialization of sophisticated instrumentation and reagents have facilitated the detection of real-time changes in cellular bioenergetics. Of those, the Seahorse™ extracellular flux (XF) analyzer has been widely used to evaluate the glycolytic flux and mitochondrial respiration in many cell types. While adherent cell lines are easy to use with this technology, non-adherent suspension cells are more difficult to handle especially when their metabolic activities are being investigated in response to drug treatment. Here, we provide an integrated protocol that allows the detection of extracellular acidification rate (ECAR) of live myeloma plasma cells in response to chemotherapeutic drugs. Our optimized protocol consists of treating myeloma cells with cytotoxic drug of interest in a standard culture plate prior to the real-time analysis in the XF analyzer. Furthermore, we provide results of experiments in which the metabolic activities of myeloma cells in response to cytotoxic treatment were compared between the manufacturer's basic procedure and our optimized protocol. Our observations suggest that our integrated protocol can be used to achieve consistent, well-standardized results and thus it may have broad applications in studies focusing on the characterization of metabolic events in non-adherent suspension cells.
Collapse
Affiliation(s)
- Alessio Lepore
- Leeds Institute of Medical Research, St. James's University Hospital, University of Leeds, Leeds, UK
| | - Fatma Necmiye Kaci
- Leeds Institute of Medical Research, St. James's University Hospital, University of Leeds, Leeds, UK
| | - Concetta Bubici
- Department of Life Sciences, Center for Genome Engineering and Maintenance, College of Health, Medicine and Life Sciences, Brunel University London, London, UK
| | - Salvatore Papa
- Leeds Institute of Medical Research, St. James's University Hospital, University of Leeds, Leeds, UK.
| |
Collapse
|
38
|
Disorders of cancer metabolism: The therapeutic potential of cannabinoids. Biomed Pharmacother 2023; 157:113993. [PMID: 36379120 DOI: 10.1016/j.biopha.2022.113993] [Citation(s) in RCA: 47] [Impact Index Per Article: 23.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2022] [Revised: 11/07/2022] [Accepted: 11/07/2022] [Indexed: 11/13/2022] Open
Abstract
Abnormal energy metabolism, as one of the important hallmarks of cancer, was induced by multiple carcinogenic factors and tumor-specific microenvironments. It comprises aerobic glycolysis, de novo lipid biosynthesis, and glutamine-dependent anaplerosis. Considering that metabolic reprogramming provides various nutrients for tumor survival and development, it has been considered a potential target for cancer therapy. Cannabinoids have been shown to exhibit a variety of anticancer activities by unclear mechanisms. This paper first reviews the recent progress of related signaling pathways (reactive oxygen species (ROS), AMP-activated protein kinase (AMPK), mitogen-activated protein kinases (MAPK), phosphoinositide 3-kinase (PI3K), hypoxia-inducible factor-1alpha (HIF-1α), and p53) mediating the reprogramming of cancer metabolism (including glucose metabolism, lipid metabolism, and amino acid metabolism). Then we comprehensively explore the latest discoveries and possible mechanisms of the anticancer effects of cannabinoids through the regulation of the above-mentioned related signaling pathways, to provide new targets and insights for cancer prevention and treatment.
Collapse
|
39
|
Zhang J, Gao Y, Zhang Z, Zhao J, Jia W, Xia C, Wang F, Liu T. Multi-therapies Based on PARP Inhibition: Potential Therapeutic Approaches for Cancer Treatment. J Med Chem 2022; 65:16099-16127. [PMID: 36512711 DOI: 10.1021/acs.jmedchem.2c01352] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
The nuclear enzymes called poly(ADP-ribose)polymerases (PARPs) are known to catalyze the process of PARylation, which plays a vital role in various cellular functions. They have become important targets for the discovery of novel antitumor drugs since their inhibition can induce significant lethality in tumor cells. Therefore, researchers all over the world have been focusing on developing novel and potent PARP inhibitors for cancer therapy. Studies have shown that PARP inhibitors and other antitumor agents, such as EZH2 and EGFR inhibitors, play a synergistic role in cancer cells. The combined inhibition of PARP and the targets with synergistic effects may provide a rational strategy to improve the effectiveness of current anticancer regimens. In this Perspective, we sum up the recent advance of PARP-targeted agents, including single-target inhibitors/degraders and dual-target inhibitors/degraders, discuss the fundamental theory of developing these dual-target agents, and give insight into the corresponding structure-activity relationships of these agents.
Collapse
Affiliation(s)
- Jie Zhang
- Department of Medicinal Chemistry, School of Pharmacy, Shandong First Medical University and Shandong Academy of Medical Sciences, Taian, Shandong 271016, China
| | - Yuqi Gao
- College of Radiology, Shandong First Medical University and Shandong Academy of Medical Sciences, Taian, Shandong 271016, China.,Medical Science and Technology Innovation Center, Shandong First Medical University, Jinan, Shandong 250117, China
| | - Zipeng Zhang
- Medical Science and Technology Innovation Center, Shandong First Medical University, Jinan, Shandong 250117, China
| | - Jinbo Zhao
- Department of Medicinal Chemistry, School of Pharmacy, Shandong First Medical University and Shandong Academy of Medical Sciences, Taian, Shandong 271016, China.,Department of Chemistry and Biology, Jilin Provincial Key Laboratory of Carbon Fiber Development and Application, Changchun University of Technology, Changchun, Jilin 130012, China
| | - Wenshuang Jia
- Medical Science and Technology Innovation Center, Shandong First Medical University, Jinan, Shandong 250117, China
| | - Chengcai Xia
- Department of Medicinal Chemistry, School of Pharmacy, Shandong First Medical University and Shandong Academy of Medical Sciences, Taian, Shandong 271016, China
| | - Fugang Wang
- Department of Pharmacology, School of Pharmacy, Shandong First Medical University and Shandong Academy of Medical Sciences, Taian, Shandong 271016, China
| | - Tingting Liu
- Department of Medicinal Chemistry, School of Pharmacy, Shandong First Medical University and Shandong Academy of Medical Sciences, Taian, Shandong 271016, China
| |
Collapse
|
40
|
Systematic Analysis of Molecular Subtypes Based on the Expression Profile of Immune-Related Genes in Pancreatic Cancer. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2022; 2022:3124122. [PMID: 36567857 PMCID: PMC9780013 DOI: 10.1155/2022/3124122] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 08/13/2022] [Revised: 09/07/2022] [Accepted: 09/14/2022] [Indexed: 12/23/2022]
Abstract
Immunotherapy has a good therapeutic effect and provides a new approach for cancer treatment. However, only limited studies have focused on the use of molecular typing to construct an immune characteristic index for gene expression in pancreatic adenocarcinoma (PAAD) and to assess the effectiveness of immunotherapy in patients with PAAD. Clinical follow-up data and gene expression profile of PAAD patients were retrieved from The Cancer Genome Atlas (TCGA) database. Based on 184 immune features, molecular subtypes of pancreatic cancer were found by the "ConsensusClusterPlus" package, and the association between clinical features and immune cell subtype distribution was analysed. In addition, the relationship between the proportion of immune subtypes and the expression of immune checkpoints was analysed. The CIBERSORT algorithm was introduced to evaluate the immune scores of different molecular subtypes. We used the tumor immune dysfunction and exclusion (TIDE) algorithm to assess the potential clinical effect of immunotherapy interventions on single-molecule subtypes. In addition, the oxidative stress index was constructed by linear discriminant analysis DNA (LDA), and weighted correlation network analysis was performed to identify the core module of the index and its characteristic genes. Expression of hub genes was verified by immunohistochemical analysis in an independent PAAD cohort. Pancreatic cancer is divided into three molecular subtypes (IS1, IS2, and IS3), with significant differences in prognosis between multiple cohorts. Expression of immune checkpoint-associated genes was significantly reduced in IS3 and higher in IS1 and IS2, suggesting that the three subgroups have different responsiveness to immunotherapy interventions. The results of the CIBERSORT analysis showed that IS1 exhibited the highest levels of immune infiltration, whereas the results of the TIDE analysis showed that the T-cell dysfunction score of IS1 was higher than that of IS2 and IS3. Furthermore, IS3 was found to be more sensitive to 5-FU and to have a higher immune signature index than IS1 and IS2. Based on WGCNA analysis, 10 potential gene markers were identified, and their expression at the protein level was verified by immunohistochemical analysis. Specific molecular expression patterns in pancreatic cancer can predict the efficacy of immunotherapy and influence the prognosis of patients.
Collapse
|
41
|
Zhang X, Liu H, Wang H, Zhao R, Lu Q, Liu Y, Han Y, LuluRen, Pan H, Han W. B3galt5 deficiency attenuates hepatocellular carcinoma by suppressing mTOR/p70s6k-mediated glycolysis. Cell Mol Life Sci 2022; 80:8. [PMID: 36495345 PMCID: PMC11072394 DOI: 10.1007/s00018-022-04601-x] [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: 11/02/2021] [Revised: 10/12/2022] [Accepted: 10/14/2022] [Indexed: 12/13/2022]
Abstract
Hepatocellular carcinoma (HCC) is one of the most common malignancies with high morbidity and mortality. Beta-1,3-galactosyltransferase 5 (b3galt5) plays crucial roles in protein glycosylation, but its function in HCC remains unclear. Here, we investigated the role and underlying mechanism of b3galt5 in HCC. We found that b3galt5 is highly expressed and associated with a poor prognosis in HCC patients. In vitro studies showed that b3galt5 promoted the proliferation and survival of HCC cells. We also demonstrated that b3galt5 deficiency suppressed hepatocarcinogenesis in DEN/TCPOBOP-induced HCC. Further investigation confirmed that b3galt5 promoted aerobic glycolysis in HCC. Mechanistically, b3galt5 promoted glycolysis by activating the mTOR/p70s6k pathway through O-linked glycosylation modification on mTOR. Moreover, p70s6k inhibition reduced the expression of key glycolytic enzymes and the glycolysis rate in b3galt5-overexpressing cells. Our study uncovers a novel mechanism by which b3galt5 mediates glycolysis in HCC and highlights the b3galt5-mTOR/p70s6k axis as a potential target for HCC therapy.
Collapse
Affiliation(s)
- Xiaoling Zhang
- Department of Medical Oncology, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, 3# East Qingchun Road, Hangzhou, 310016, Zhejiang, China
- Department of Medical Oncology, Hangzhou First People's Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, China
| | - Hao Liu
- Department of Medical Oncology, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, 3# East Qingchun Road, Hangzhou, 310016, Zhejiang, China
- Laboratory of Cancer Biology, Institute of Clinical Science, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, China
| | - Haidong Wang
- Department of Medical Oncology, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, 3# East Qingchun Road, Hangzhou, 310016, Zhejiang, China
| | - Rongjie Zhao
- Department of Medical Oncology, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, 3# East Qingchun Road, Hangzhou, 310016, Zhejiang, China
| | - Qian Lu
- Department of Medical Oncology, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, 3# East Qingchun Road, Hangzhou, 310016, Zhejiang, China
| | - Yunlong Liu
- Department of Medical Oncology, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, 3# East Qingchun Road, Hangzhou, 310016, Zhejiang, China
- Laboratory of Cancer Biology, Institute of Clinical Science, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, China
| | - Yicheng Han
- Department of Medical Oncology, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, 3# East Qingchun Road, Hangzhou, 310016, Zhejiang, China
| | - LuluRen
- Department of Medical Oncology, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, 3# East Qingchun Road, Hangzhou, 310016, Zhejiang, China
| | - Hongming Pan
- Department of Medical Oncology, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, 3# East Qingchun Road, Hangzhou, 310016, Zhejiang, China.
| | - Weidong Han
- Department of Medical Oncology, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, 3# East Qingchun Road, Hangzhou, 310016, Zhejiang, China.
| |
Collapse
|
42
|
Hu F, Li C, Ye Y, Lu X, Alimujiang M, Bai N, Sun J, Ma X, Li X, Yang Y. PARP12 is required for mitochondrial function maintenance in thermogenic adipocytes. Adipocyte 2022; 11:379-388. [PMID: 35916471 PMCID: PMC9351573 DOI: 10.1080/21623945.2022.2091206] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/01/2022] Open
Abstract
PARP12 is a member of poly-ADP-ribosyl polymerase (PARPs), which has been characterized for its antiviral function. Yet its physiological implication in adipocytes remains unknown. Here, we report a central function of PARP12 in thermogenic adipocytes. We show that PARP12 is highly expressed in brown adipose tissue and is mainly localized to the mitochondria. Knockdown of PARP12 in vitro reduced UCP1 expression. In parallel, the deficiency of PARP12 reduced mitochondrial respiration in adipocytes, while overexpression of PARP12 reversed these effects.
Collapse
Affiliation(s)
- Fan Hu
- Shanghai Clinical Center for Diabetes, Shanghai Key Clinical Center for Metabolic Disease, Shanghai Diabetes Institute, Shanghai Key Laboratory of Diabetes Mellitus, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, China
| | - Chang Li
- Department of Endocrinology, Seventh People's Hospital Affiliated to Shanghai University of TCM, Shanghai, China
| | - Yafen Ye
- Shanghai Clinical Center for Diabetes, Shanghai Key Clinical Center for Metabolic Disease, Shanghai Diabetes Institute, Shanghai Key Laboratory of Diabetes Mellitus, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, China
| | - Xuhong Lu
- Shanghai Clinical Center for Diabetes, Shanghai Key Clinical Center for Metabolic Disease, Shanghai Diabetes Institute, Shanghai Key Laboratory of Diabetes Mellitus, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, China
| | - Miriayi Alimujiang
- Shanghai Clinical Center for Diabetes, Shanghai Key Clinical Center for Metabolic Disease, Shanghai Diabetes Institute, Shanghai Key Laboratory of Diabetes Mellitus, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, China
| | - Ningning Bai
- Shanghai Clinical Center for Diabetes, Shanghai Key Clinical Center for Metabolic Disease, Shanghai Diabetes Institute, Shanghai Key Laboratory of Diabetes Mellitus, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, China
| | - Jingjing Sun
- Shanghai Clinical Center for Diabetes, Shanghai Key Clinical Center for Metabolic Disease, Shanghai Diabetes Institute, Shanghai Key Laboratory of Diabetes Mellitus, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, China
| | - Xiaojing Ma
- Shanghai Clinical Center for Diabetes, Shanghai Key Clinical Center for Metabolic Disease, Shanghai Diabetes Institute, Shanghai Key Laboratory of Diabetes Mellitus, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, China
| | - Xiaohua Li
- Department of Endocrinology, Seventh People's Hospital Affiliated to Shanghai University of TCM, Shanghai, China
| | - Ying Yang
- Shanghai Clinical Center for Diabetes, Shanghai Key Clinical Center for Metabolic Disease, Shanghai Diabetes Institute, Shanghai Key Laboratory of Diabetes Mellitus, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, China
| |
Collapse
|
43
|
Choudhury D, Rong N, Ikhapoh I, Rajabian N, Tseropoulos G, Wu Y, Mehrotra P, Thiyagarajan R, Shahini A, Seldeen KL, Troen B, Lei P, Andreadis ST. Inhibition of glutaminolysis restores mitochondrial function in senescent stem cells. Cell Rep 2022; 41:111744. [PMID: 36450260 PMCID: PMC9809151 DOI: 10.1016/j.celrep.2022.111744] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2021] [Revised: 07/07/2022] [Accepted: 11/07/2022] [Indexed: 12/03/2022] Open
Abstract
Mitochondrial dysfunction, a hallmark of aging, has been associated with the onset of aging phenotypes and age-related diseases. Here, we report that impaired mitochondrial function is associated with increased glutamine catabolism in senescent human mesenchymal stem cells (MSCs) and myofibroblasts derived from patients suffering from Hutchinson-Gilford progeria syndrome. Increased glutaminase (GLS1) activity accompanied by loss of urea transporter SLC14A1 induces urea accumulation, mitochondrial dysfunction, and DNA damage. Conversely, blocking GLS1 activity restores mitochondrial function and leads to amelioration of aging hallmarks. Interestingly, GLS1 expression is regulated through the JNK pathway, as demonstrated by chemical and genetic inhibition. In agreement with our in vitro findings, tissues isolated from aged or progeria mice display increased urea accumulation and GLS1 activity, concomitant with declined mitochondrial function. Inhibition of glutaminolysis in progeria mice improves mitochondrial respiratory chain activity, suggesting that targeting glutaminolysis may be a promising strategy for restoring age-associated loss of mitochondrial function.
Collapse
Affiliation(s)
- Debanik Choudhury
- Department of Chemical and Biological Engineering, University at Buffalo, Buffalo, NY 14260
| | - Na Rong
- Department of Chemical and Biological Engineering, University at Buffalo, Buffalo, NY 14260
| | - Izuagie Ikhapoh
- Department of Chemical and Biological Engineering, University at Buffalo, Buffalo, NY 14260
| | - Nika Rajabian
- Department of Chemical and Biological Engineering, University at Buffalo, Buffalo, NY 14260
| | - Georgios Tseropoulos
- Department of Chemical and Biological Engineering, University at Buffalo, Buffalo, NY 14260
| | - Yulun Wu
- Department of Chemical and Biological Engineering, University at Buffalo, Buffalo, NY 14260
| | - Pihu Mehrotra
- Department of Chemical and Biological Engineering, University at Buffalo, Buffalo, NY 14260
| | - Ramkumar Thiyagarajan
- Department of Medicine, Division of Geriatrics and Palliative medicine, Buffalo, NY 14203
| | - Aref Shahini
- Department of Chemical and Biological Engineering, University at Buffalo, Buffalo, NY 14260
| | - Kenneth L. Seldeen
- Department of Medicine, Division of Geriatrics and Palliative medicine, Buffalo, NY 14203
| | - Bruce Troen
- Department of Medicine, Division of Geriatrics and Palliative medicine, Buffalo, NY 14203
| | - Pedro Lei
- Department of Chemical and Biological Engineering, University at Buffalo, Buffalo, NY 14260
| | - Stelios T. Andreadis
- Department of Chemical and Biological Engineering, University at Buffalo, Buffalo, NY 14260,Department of Biomedical Engineering, University at Buffalo, Buffalo, NY 14260,Center of Excellence in Bioinformatics and Life Sciences, University at Buffalo, Buffalo, NY 14263,Center for Cell, Gene and Tissue Engineering (CGTE), University at Buffalo, Buffalo, NY 14260,Address for all Correspondence: Stelios T. Andreadis, Ph.D., SUNY Distinguished Professor, Bioengineering Laboratory, 908 Furnas Hall, Department of Chemical and Biological Engineering, Department of Biomedical Engineering, and Center of Excellence in Bioinformatics and Life Sciences, Center for Cell, Gene and Tissue Engineering (CGTE), University at Buffalo, The State University of New York, Amherst, NY 14260-4200, USA, Tel: (716) 645-1202, Fax: (716) 645-3822,
| |
Collapse
|
44
|
Di Paola S, Matarese M, Barretta ML, Dathan N, Colanzi A, Corda D, Grimaldi G. PARP10 Mediates Mono-ADP-Ribosylation of Aurora-A Regulating G2/M Transition of the Cell Cycle. Cancers (Basel) 2022; 14:5210. [PMID: 36358629 PMCID: PMC9659153 DOI: 10.3390/cancers14215210] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2022] [Revised: 10/11/2022] [Accepted: 10/22/2022] [Indexed: 08/13/2023] Open
Abstract
Intracellular mono-ADP-ribosyltransferases (mono-ARTs) catalyze the covalent attachment of a single ADP-ribose molecule to protein substrates, thus regulating their functions. PARP10 is a soluble mono-ART involved in the modulation of intracellular signaling, metabolism and apoptosis. PARP10 also participates in the regulation of the G1- and S-phase of the cell cycle. However, the role of this enzyme in G2/M progression is not defined. In this study, we found that genetic ablation, protein depletion and pharmacological inhibition of PARP10 cause a delay in the G2/M transition of the cell cycle. Moreover, we found that the mitotic kinase Aurora-A, a previously identified PARP10 substrate, is actively mono-ADP-ribosylated (MARylated) during G2/M transition in a PARP10-dependent manner. Notably, we showed that PARP10-mediated MARylation of Aurora-A enhances the activity of the kinase in vitro. Consistent with an impairment in the endogenous activity of Aurora-A, cells lacking PARP10 show a decreased localization of the kinase on the centrosomes and mitotic spindle during G2/M progression. Taken together, our data provide the first evidence of a direct role played by PARP10 in the progression of G2 and mitosis, an event that is strictly correlated to the endogenous MARylation of Aurora-A, thus proposing a novel mechanism for the modulation of Aurora-A kinase activity.
Collapse
Affiliation(s)
- Simone Di Paola
- Institute of Experimental Endocrinology and Oncology “G. Salvatore” (IEOS), National Research Council (CNR), 80131 Naples, Italy
| | - Maria Matarese
- Institute of Experimental Endocrinology and Oncology “G. Salvatore” (IEOS), National Research Council (CNR), 80131 Naples, Italy
| | - Maria Luisa Barretta
- National Research Council (CNR), Piazzale Aldo Moro, 700185 Rome, Italy
- Steril Farma Srl, Via L. Da Vinci 128, 80055 Portici, Italy
| | - Nina Dathan
- Institute of Experimental Endocrinology and Oncology “G. Salvatore” (IEOS), National Research Council (CNR), 80131 Naples, Italy
| | - Antonino Colanzi
- Institute of Experimental Endocrinology and Oncology “G. Salvatore” (IEOS), National Research Council (CNR), 80131 Naples, Italy
| | - Daniela Corda
- Institute of Experimental Endocrinology and Oncology “G. Salvatore” (IEOS), National Research Council (CNR), 80131 Naples, Italy
| | - Giovanna Grimaldi
- Institute of Experimental Endocrinology and Oncology “G. Salvatore” (IEOS), National Research Council (CNR), 80131 Naples, Italy
| |
Collapse
|
45
|
Balakrishnan K. Hepatocellular carcinoma stage: an almost loss of fatty acid metabolism and gain of glucose metabolic pathways dysregulation. MEDICAL ONCOLOGY (NORTHWOOD, LONDON, ENGLAND) 2022; 39:247. [PMID: 36209296 DOI: 10.1007/s12032-022-01839-0] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/27/2022] [Accepted: 09/01/2022] [Indexed: 10/10/2022]
Abstract
Cancer cells rewire the metabolic processes beneficial for cancer cell proliferation, survival, and their progression. In this study, metabolic processes related to glucose, glutamine, and fatty acid metabolism signatures were collected from the molecular signatures database and investigated in the context of energy metabolic pathways through available genome-wide expression profiles of liver cancer cohorts by gene sets-based pathway activation scoring analysis. The outcomes of this study portray that the fatty acid metabolism, transport, and its storage related signatures are highly expressed across early stages of liver tumors and on the contrary, the gene sets related to glucose transport and glucose metabolism are prominently activated in the hepatocellular carcinoma (HCC) stage. Based on the results, these metabolic pathways are clearly dysregulated across specific stages of carcinogenesis. The identified dimorphic metabolic pathway dysregulation patterns are further reconfirmed by examining corresponding metabolic pathway genes expression patterns across various stages encompassing profiles. Recurrence is the primary concern in the carcinogenesis of liver tumors due to liver tissues regeneration. Hence, to further explore these dysregulation effects on recurrent cirrhosis and recurrent HCC sample containing profile GSE20140 was examined and interestingly, this result also reiterated these differential metabolic pathways dysregulation. In addition, a recently established metabolome profile for the massive panel of cancer cell-lines, including liver cancer cell-lines, was used for further exploration. These findings also reassured those differential metabolites abundance of the fatty acid and glucose metabolic pathways enlighten those dimorphic metabolic pathways dysregulation. Moreover, ROC curves of fatty acid metabolic pathway genes such as acetyl-CoA carboxylase (ACACB), acyl-CoA dehydrogenase long chain (ACADL), and acyl-CoA dehydrogenase medium chain (ACADM) as well as glucose metabolic pathway genes such as phosphoglycerate kinase (PGK1), pyruvate dehydrogenase (PDHA1), pyruvate dehydrogenase kinase (PDK1) demonstrated greater sensitivity and specificity in the corresponding stage-specific tumors with significant p-values (p < 0.05). Furthermore, overall survival (OS) and recurrence-free survival (RFS) studies also reconfirmed that the rate-limiting genes expression of fatty acid and glucose metabolic pathways reveal better and poor survival in HCC patient cohorts, respectively. In conclusion, all these results clearly show that metabolic rewiring and the existence of two diverse metabolic pathways dysregulation involving fatty acid and glucose metabolism across the stages of liver tumors have been identified. These findings might be useful for developing therapeutic target treatments in stage-specific tumors.
Collapse
Affiliation(s)
- Karthik Balakrishnan
- Department of Biotechnology, Saroj Institute of Technology and Management (SITM), 12th KM Stone, Lucknow-Sultanpur Road, Lucknow, Uttar Pradesh, 226002, India.
| |
Collapse
|
46
|
Mashimo M, Shimizu A, Mori A, Hamaguchi A, Fukushima K, Seira N, Fujii T, Fujino H. PARP14 regulates EP4 receptor expression in human colon cancer HCA-7 cells. Biochem Biophys Res Commun 2022; 623:133-139. [DOI: 10.1016/j.bbrc.2022.07.055] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2022] [Accepted: 07/14/2022] [Indexed: 11/02/2022]
|
47
|
Dhoonmoon A, Nicolae CM, Moldovan GL. The KU-PARP14 axis differentially regulates DNA resection at stalled replication forks by MRE11 and EXO1. Nat Commun 2022; 13:5063. [PMID: 36030235 PMCID: PMC9420157 DOI: 10.1038/s41467-022-32756-5] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2022] [Accepted: 08/12/2022] [Indexed: 11/24/2022] Open
Abstract
Suppression of nascent DNA degradation has emerged as an essential role of the BRCA pathway in genome protection. In BRCA-deficient cells, the MRE11 nuclease is responsible for both resection of reversed replication forks, and accumulation of single stranded DNA gaps behind forks. Here, we show that the mono-ADP-ribosyltransferase PARP14 is a critical co-factor of MRE11. PARP14 is recruited to nascent DNA upon replication stress in BRCA-deficient cells, and through its catalytic activity, mediates the engagement of MRE11. Loss or inhibition of PARP14 suppresses MRE11-mediated fork degradation and gap accumulation, and promotes genome stability and chemoresistance of BRCA-deficient cells. Moreover, we show that the KU complex binds reversed forks and protects them against EXO1-catalyzed degradation. KU recruits the PARP14-MRE11 complex, which initiates partial resection to release KU and allow long-range resection by EXO1. Our work identifies a multistep process of nascent DNA processing at stalled replication forks in BRCA-deficient cells. Protection of replication forks against nucleolytic degradation is crucial for genome stability. Here, Dhoonmoon et al identify PARP14 and the KU complex as essential regulators of fork degradation by MRE11 and EXO1 nucleases in BRCA-deficient cells.
Collapse
Affiliation(s)
- Ashna Dhoonmoon
- Department of Biochemistry and Molecular Biology, The Pennsylvania State University College of Medicine, Hershey, PA, 17033, USA
| | - Claudia M Nicolae
- Department of Biochemistry and Molecular Biology, The Pennsylvania State University College of Medicine, Hershey, PA, 17033, USA.
| | - George-Lucian Moldovan
- Department of Biochemistry and Molecular Biology, The Pennsylvania State University College of Medicine, Hershey, PA, 17033, USA.
| |
Collapse
|
48
|
Li P, Lei Y, Qi J, Liu W, Yao K. Functional roles of ADP-ribosylation writers, readers and erasers. Front Cell Dev Biol 2022; 10:941356. [PMID: 36035988 PMCID: PMC9404506 DOI: 10.3389/fcell.2022.941356] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2022] [Accepted: 07/20/2022] [Indexed: 11/17/2022] Open
Abstract
ADP-ribosylation is a reversible post-translational modification (PTM) tightly regulated by the dynamic interplay between its writers, readers and erasers. As an intricate and versatile PTM, ADP-ribosylation plays critical roles in various physiological and pathological processes. In this review, we discuss the major players involved in the ADP-ribosylation cycle, which may facilitate the investigation of the ADP-ribosylation function and contribute to the understanding and treatment of ADP-ribosylation associated disease.
Collapse
|
49
|
Zhu Y, Liu Z, Wan Y, Zou L, Liu L, Ding S, Lu C, Qiu F. PARP14 promotes the growth and glycolysis of acute myeloid leukemia cells by regulating HIF-1α expression. Clin Immunol 2022; 242:109094. [DOI: 10.1016/j.clim.2022.109094] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2022] [Revised: 07/29/2022] [Accepted: 08/01/2022] [Indexed: 11/03/2022]
|
50
|
Nizi M, Maksimainen MM, Lehtiö L, Tabarrini O. Medicinal Chemistry Perspective on Targeting Mono-ADP-Ribosylating PARPs with Small Molecules. J Med Chem 2022; 65:7532-7560. [PMID: 35608571 PMCID: PMC9189837 DOI: 10.1021/acs.jmedchem.2c00281] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2022] [Indexed: 12/13/2022]
Abstract
Major advances have recently defined functions for human mono-ADP-ribosylating PARP enzymes (mono-ARTs), also opening up potential applications for targeting them to treat diseases. Structural biology combined with medicinal chemistry has allowed the design of potent small molecule inhibitors which typically bind to the catalytic domain. Most of these inhibitors are at the early stages, but some have already a suitable profile to be used as chemical tools. One compound targeting PARP7 has even progressed to clinical trials. In this review, we collect inhibitors of mono-ARTs with a typical "H-Y-Φ" motif (Φ = hydrophobic residue) and focus on compounds that have been reported as active against one or a restricted number of enzymes. We discuss them from a medicinal chemistry point of view and include an analysis of the available crystal structures, allowing us to craft a pharmacophore model that lays the foundation for obtaining new potent and more specific inhibitors.
Collapse
Affiliation(s)
- Maria
Giulia Nizi
- Department
of Pharmaceutical Sciences, University of
Perugia, 06123 Perugia, Italy
| | - Mirko M. Maksimainen
- Faculty
of Biochemistry and Molecular Medicine & Biocenter Oulu, University of Oulu, 5400 Oulu, Finland
| | - Lari Lehtiö
- Faculty
of Biochemistry and Molecular Medicine & Biocenter Oulu, University of Oulu, 5400 Oulu, Finland
| | - Oriana Tabarrini
- Department
of Pharmaceutical Sciences, University of
Perugia, 06123 Perugia, Italy
| |
Collapse
|