101
|
Kim E, Hwang Y, Kim H, Kim GU, Ryu YC, Yoon M, Choi KY. Pyruvate Kinase M2 Accelerates Cutaneous Wound Healing via Glycolysis and Wnt/β-Catenin Signaling. Pharmaceutics 2023; 15:2028. [PMID: 37631242 PMCID: PMC10458512 DOI: 10.3390/pharmaceutics15082028] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2023] [Revised: 07/20/2023] [Accepted: 07/25/2023] [Indexed: 08/27/2023] Open
Abstract
Cutaneous wound healing is a complex and dynamic process with high energy demand. The activation of glycolysis is essential for restoring the structure and function of injured tissues in wounds. Pyruvate kinase M2 (PKM2) is an enzyme that plays a crucial role in the last step of glycolysis. PKM2-mediated glycolysis is known to play an important role in diseases related to regeneration and inflammation. However, the role of PKM2 in wound healing has not been fully elucidated. In this study, we found that PKM2 expression and pyruvate kinase (PK) activity were increased with the activation of Wnt/β-catenin signaling during wound healing in mice. TEPP-46, an allosteric activator of PKM2, enhanced HaCaT human keratinocyte migration and cutaneous wound healing with an increment of PK activity. Moreover, we confirmed the effect of co-treatment with TEPP-46 and KY19382, a Wnt/β-catenin signaling activator through the interference with the CXXC-type zinc finger protein 5 (CXXC5) Dishevelled interaction, on wound healing. The combination treatment significantly accelerated wound healing, which was confirmed by the expression level of PCNA, keratin 14, and α-SMA. Furthermore, co-treatment induced angiogenesis in the wound beds. Overall, activation of both glycolysis and Wnt/β-catenin signaling has the potential to be used as a therapeutic approach for wound healing.
Collapse
Affiliation(s)
- Eunhwan Kim
- Department of Biotechnology, College of Life Science and Biotechnology, Yonsei University, Seoul 03722, Republic of Korea; (E.K.); (Y.H.); (H.K.); (G.-U.K.); (Y.C.R.); (M.Y.)
| | - Yumi Hwang
- Department of Biotechnology, College of Life Science and Biotechnology, Yonsei University, Seoul 03722, Republic of Korea; (E.K.); (Y.H.); (H.K.); (G.-U.K.); (Y.C.R.); (M.Y.)
| | - Heejene Kim
- Department of Biotechnology, College of Life Science and Biotechnology, Yonsei University, Seoul 03722, Republic of Korea; (E.K.); (Y.H.); (H.K.); (G.-U.K.); (Y.C.R.); (M.Y.)
| | - Geon-Uk Kim
- Department of Biotechnology, College of Life Science and Biotechnology, Yonsei University, Seoul 03722, Republic of Korea; (E.K.); (Y.H.); (H.K.); (G.-U.K.); (Y.C.R.); (M.Y.)
| | - Yeong Chan Ryu
- Department of Biotechnology, College of Life Science and Biotechnology, Yonsei University, Seoul 03722, Republic of Korea; (E.K.); (Y.H.); (H.K.); (G.-U.K.); (Y.C.R.); (M.Y.)
| | - Minguen Yoon
- Department of Biotechnology, College of Life Science and Biotechnology, Yonsei University, Seoul 03722, Republic of Korea; (E.K.); (Y.H.); (H.K.); (G.-U.K.); (Y.C.R.); (M.Y.)
| | - Kang-Yell Choi
- Department of Biotechnology, College of Life Science and Biotechnology, Yonsei University, Seoul 03722, Republic of Korea; (E.K.); (Y.H.); (H.K.); (G.-U.K.); (Y.C.R.); (M.Y.)
- CK Regeon Inc., Seoul 03722, Republic of Korea
| |
Collapse
|
102
|
Qu H, Liu J, Zhang D, Xie R, Wang L, Hong J. Glycolysis in Chronic Liver Diseases: Mechanistic Insights and Therapeutic Opportunities. Cells 2023; 12:1930. [PMID: 37566009 PMCID: PMC10417805 DOI: 10.3390/cells12151930] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2023] [Revised: 07/17/2023] [Accepted: 07/21/2023] [Indexed: 08/12/2023] Open
Abstract
Chronic liver diseases (CLDs) cover a spectrum of liver diseases, ranging from nonalcoholic fatty liver disease to liver cancer, representing a growing epidemic worldwide with high unmet medical needs. Glycolysis is a conservative and rigorous process that converts glucose into pyruvate and sustains cells with the energy and intermediate products required for diverse biological activities. However, abnormalities in glycolytic flux during CLD development accelerate the disease progression. Aerobic glycolysis is a hallmark of liver cancer and is responsible for a broad range of oncogenic functions including proliferation, invasion, metastasis, angiogenesis, immune escape, and drug resistance. Recently, the non-neoplastic role of aerobic glycolysis in immune activation and inflammatory disorders, especially CLD, has attracted increasing attention. Several key mediators of aerobic glycolysis, including HIF-1α and pyruvate kinase M2 (PKM2), are upregulated during steatohepatitis and liver fibrosis. The pharmacological inhibition or ablation of PKM2 effectively attenuates hepatic inflammation and CLD progression. In this review, we particularly focused on the glycolytic and non-glycolytic roles of PKM2 in the progression of CLD, highlighting the translational potential of a glycolysis-centric therapeutic approach in combating CLD.
Collapse
Affiliation(s)
| | | | | | | | | | - Jian Hong
- Department of Pathophysiology, School of Medicine, Jinan University, Guangzhou 510632, China; (H.Q.)
| |
Collapse
|
103
|
He P, Dai Q, Wu X. New insight in urological cancer therapy: From epithelial-mesenchymal transition (EMT) to application of nano-biomaterials. ENVIRONMENTAL RESEARCH 2023; 229:115672. [PMID: 36906272 DOI: 10.1016/j.envres.2023.115672] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/20/2023] [Revised: 03/07/2023] [Accepted: 03/09/2023] [Indexed: 05/21/2023]
Abstract
A high number of cancer-related deaths (up to 90) are due to metastasis and simple definition of metastasis is new colony formation of tumor cells in a secondary site. In tumor cells, epithelial-mesenchymal transition (EMT) stimulates metastasis and invasion, and it is a common characteristic of malignant tumors. Prostate cancer, bladder cancer and renal cancer are three main types of urological tumors that their malignant and aggressive behaviors are due to abnormal proliferation and metastasis. EMT has been well-documented as a mechanism for promoting invasion of tumor cells and in the current review, a special attention is directed towards understanding role of EMT in malignancy, metastasis and therapy response of urological cancers. The invasion and metastatic characteristics of urological tumors enhance due to EMT induction and this is essential for ensuring survival and ability in developing new colonies in neighboring and distant tissues and organs. When EMT induction occurs, malignant behavior of tumor cells enhances and their tend in developing therapy resistance especially chemoresistance promotes that is one of the underlying reasons for therapy failure and patient death. The lncRNAs, microRNAs, eIF5A2, Notch-4 and hypoxia are among common modulators of EMT mechanism in urological tumors. Moreover, anti-tumor compounds such as metformin can be utilized in suppressing malignancy of urological tumors. Besides, genes and epigenetic factors modulating EMT mechanism can be therapeutically targeted for interfering malignancy of urological tumors. Nanomaterials are new emerging agents in urological cancer therapy that they can improve potential of current therapeutics by their targeted delivery to tumor site. The important hallmarks of urological cancers including growth, invasion and angiogenesis can be suppressed by cargo-loaded nanomaterials. Moreover, nanomaterials can improve chemotherapy potential in urological cancer elimination and by providing phototherapy, they mediate synergistic tumor suppression. The clinical application depends on development of biocompatible nanomaterials.
Collapse
Affiliation(s)
- Peng He
- Department of Urology, Southwest Hospital, Army Medical University (Third Military Medical University), Chongqing, 400038, China
| | - Qiang Dai
- Department of Urology, Southwest Hospital, Army Medical University (Third Military Medical University), Chongqing, 400038, China
| | - Xiaojun Wu
- Department of Urology, Southwest Hospital, Army Medical University (Third Military Medical University), Chongqing, 400038, China.
| |
Collapse
|
104
|
Paciotti S, Wojdała AL, Bellomo G, Toja A, Chipi E, Piersma SR, Pham TV, Gaetani L, Jimenez CR, Parnetti L, Chiasserini D. Potential diagnostic value of CSF metabolism-related proteins across the Alzheimer's disease continuum. Alzheimers Res Ther 2023; 15:124. [PMID: 37454217 DOI: 10.1186/s13195-023-01269-8] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2023] [Accepted: 07/04/2023] [Indexed: 07/18/2023]
Abstract
BACKGROUND Alzheimer's disease (AD) cerebrospinal fluid (CSF) core biomarkers (Aβ42/40 ratio, p-tau, and t-tau) provide high diagnostic accuracy, even at the earliest stage of disease. However, these markers do not fully reflect the complex AD pathophysiology. Recent large scale CSF proteomic studies revealed several new AD candidate biomarkers related to metabolic pathways. In this study we measured the CSF levels of four metabolism-related proteins not directly linked to amyloid- and tau-pathways (i.e., pyruvate kinase, PKM; aldolase, ALDO; ubiquitin C-terminal hydrolase L1, UCHL1, and fatty acid-binding protein 3, FABP3) across the AD continuum. We aimed at validating the potential value of these proteins as new CSF biomarkers for AD and their possible involvement in AD pathogenesis, with specific interest on the preclinical phase of the disease. METHODS CSF PKM and ALDO activities were measured with specific enzyme assays while UCHL1 and FABP3 levels were measured with immunoassays in a cohort of patients composed as follows: preclinical AD (pre-AD, n = 19, cognitively unimpaired), mild cognitive impairment due to AD (MCI-AD, n = 50), dementia due to AD (ADdem, n = 45), and patients with frontotemporal dementia (FTD, n = 37). Individuals with MCI not due to AD (MCI, n = 30) and subjective cognitive decline (SCD, n = 52) with negative CSF AD-profile, were enrolled as control groups. RESULTS CSF UCHL1 and FABP3 levels, and PKM activity were significantly increased in AD patients, already at the pre-clinical stage. CSF PKM activity was also increased in FTD patients compared with control groups, being similar between AD and FTD patients. No difference was found in ALDO activity among the groups. UCHL1 showed good performance in discriminating early AD patients (pre-AD and MCI-AD) from controls (AUC ~ 0.83), as assessed by ROC analysis. Similar results were obtained for FABP3. Conversely, PKM provided the best performance when comparing FTD vs. MCI (AUC = 0.80). Combination of PKM, FABP3, and UCHL1 improved the diagnostic accuracy for the detection of patients within the AD continuum when compared with single biomarkers. CONCLUSIONS Our study confirmed the potential role of UCHL1 and FABP3 as neurodegenerative biomarkers for AD. Furthermore, our results validated the increase of PKM activity in CSF of AD patients, already at the preclinical phase of the disease. Increased PKM activity was observed also in FTD patients, possibly underlining similar alterations in energy metabolism in AD and FTD.
Collapse
Affiliation(s)
- Silvia Paciotti
- Section of Physiology and Biochemistry, Department of Medicine and Surgery, University of Perugia, Perugia, Italy
| | - Anna Lidia Wojdała
- Laboratory of Clinical Neurochemistry, Department of Medicine and Surgery, University of Perugia, Perugia, Italy
| | - Giovanni Bellomo
- Laboratory of Clinical Neurochemistry, Department of Medicine and Surgery, University of Perugia, Perugia, Italy
| | - Andrea Toja
- Section of Neurology, Department of Medicine and Surgery, University of Perugia, Perugia, Italy
| | - Elena Chipi
- Section of Neurology, Department of Medicine and Surgery, University of Perugia, Perugia, Italy
| | - Sander R Piersma
- OncoProteomics Laboratory, Laboratory Medical Oncology, Amsterdam University Medical Center, Amsterdam, The Netherlands
| | - Thang V Pham
- OncoProteomics Laboratory, Laboratory Medical Oncology, Amsterdam University Medical Center, Amsterdam, The Netherlands
| | - Lorenzo Gaetani
- Section of Neurology, Department of Medicine and Surgery, University of Perugia, Perugia, Italy
| | - Connie R Jimenez
- OncoProteomics Laboratory, Laboratory Medical Oncology, Amsterdam University Medical Center, Amsterdam, The Netherlands
| | - Lucilla Parnetti
- Section of Neurology, Department of Medicine and Surgery, University of Perugia, Perugia, Italy.
| | - Davide Chiasserini
- Section of Physiology and Biochemistry, Department of Medicine and Surgery, University of Perugia, Perugia, Italy.
| |
Collapse
|
105
|
Zhang Q, Liu J, Lin H, Lin B, Zhu M, Li M. Glucose metabolism reprogramming promotes immune escape of hepatocellular carcinoma cells. EXPLORATION OF TARGETED ANTI-TUMOR THERAPY 2023; 4:519-536. [PMID: 37455832 PMCID: PMC10344893 DOI: 10.37349/etat.2023.00149] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2022] [Accepted: 03/13/2023] [Indexed: 07/18/2023] Open
Abstract
Hepatocellular carcinoma (HCC) is a complex process that plays an important role in its progression. Abnormal glucose metabolism in HCC cells can meet the nutrients required for the occurrence and development of liver cancer, better adapt to changes in the surrounding microenvironment, and escape the attack of the immune system on the tumor. There is a close relationship between reprogramming of glucose metabolism and immune escape. This article reviews the current status and progress of glucose metabolism reprogramming in promoting immune escape in liver cancer, aiming to provide new strategies for clinical immunotherapy of liver cancer.
Collapse
Affiliation(s)
- Qiuyue Zhang
- Hainan Provincial Key Laboratory of Carcinogenesis and Intervention, Hainan Medical College, Haikou 571199, Hainan Province, China
| | - Jinchen Liu
- Hainan Provincial Key Laboratory of Carcinogenesis and Intervention, Hainan Medical College, Haikou 571199, Hainan Province, China
| | - Haifeng Lin
- Department of Medical Oncology, Second Affiliated Hospital, Hainan Medical College, Haikou 570216, Hainan Province, China
| | - Bo Lin
- Hainan Provincial Key Laboratory of Carcinogenesis and Intervention, Hainan Medical College, Haikou 571199, Hainan Province, China
| | - Mingyue Zhu
- Hainan Provincial Key Laboratory of Carcinogenesis and Intervention, Hainan Medical College, Haikou 571199, Hainan Province, China
| | - Mengsen Li
- Hainan Provincial Key Laboratory of Carcinogenesis and Intervention, Hainan Medical College, Haikou 571199, Hainan Province, China
- Department of Medical Oncology, Second Affiliated Hospital, Hainan Medical College, Haikou 570216, Hainan Province, China
- Institution of Tumor, Hainan Medical College, Haikou 570102, Hainan Province, China
| |
Collapse
|
106
|
Yang Q, Zou Y, Wei X, Ye P, Wu Y, Ai H, Zhang Z, Tan J, Zhou J, Yang Y, Dai Q, Dou C, Luo F. PTP1B knockdown alleviates BMSCs senescence via activating AMPK-mediated mitophagy and promotes osteogenesis in senile osteoporosis. Biochim Biophys Acta Mol Basis Dis 2023:166795. [PMID: 37385514 DOI: 10.1016/j.bbadis.2023.166795] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2023] [Revised: 06/09/2023] [Accepted: 06/21/2023] [Indexed: 07/01/2023]
Abstract
The senescence of bone marrow mesenchymal stem cells (BMSCs) is the basis of senile osteoporosis (SOP). Targeting BMSCs senescence is of paramount importance for developing anti-osteoporotic strategy. In this study, we found that protein tyrosine phosphatase 1B (PTP1B), an enzyme responsible for tyrosine dephosphorylation, was significantly upregulated in BMSCs and femurs with advancing chronological age. Therefore, the potential role of PTP1B in BMSCs senescence and senile osteoporosis was studied. Firstly, significantly upregulated PTP1B expression along with impaired osteogenic differentiation capacity was observed in D-galactose (D-gal)-induced BMSCs and naturally-aged BMSCs. Furthermore, PTP1B silencing could effectively alleviate senescence, improve mitochondrial dysfunction, and restore osteogenic differentiation in aged BMSCs, which was attributable to enhanced mitophagy mediated by PKM2/AMPK pathway. In addition, hydroxychloroquine (HCQ), an autophagy inhibitor, significantly reversed the protective effects from PTP1B knockdown. In SOP animal model, transplantation of LVsh-PTP1B-transfected D-gal-induced BMSCs harvested double protective effects, including increased bone formation and reduced osteoclastogenesis. Similarly, HCQ treatment remarkably suppressed osteogenesis of LVsh-PTP1B-transfected D-gal-induced BMSCs in vivo. Taken together, our data demonstrated that PTP1B silencing protects against BMSCs senescence and mitigates SOP via activating AMPK-mediated mitophagy. Targeting PTP1B may represent a promising interventional strategy to attenuate SOP.
Collapse
Affiliation(s)
- QianKun Yang
- National & Regional United Engineering Lab of Tissue Engineering, Department of Orthopedics, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, 400038, China
| | - YuChi Zou
- National & Regional United Engineering Lab of Tissue Engineering, Department of Orthopedics, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, 400038, China
| | - XiaoYu Wei
- National & Regional United Engineering Lab of Tissue Engineering, Department of Orthopedics, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, 400038, China
| | - Peng Ye
- National & Regional United Engineering Lab of Tissue Engineering, Department of Orthopedics, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, 400038, China
| | - YuTong Wu
- National & Regional United Engineering Lab of Tissue Engineering, Department of Orthopedics, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, 400038, China
| | - HongBo Ai
- National & Regional United Engineering Lab of Tissue Engineering, Department of Orthopedics, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, 400038, China
| | - Zhao Zhang
- National & Regional United Engineering Lab of Tissue Engineering, Department of Orthopedics, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, 400038, China; Orthopedics Department, The General Hospital of Western Theater Command PLA, Chengdu 610083, Sichuan Province, China
| | - JiuLin Tan
- National & Regional United Engineering Lab of Tissue Engineering, Department of Orthopedics, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, 400038, China
| | - Jiangling Zhou
- National & Regional United Engineering Lab of Tissue Engineering, Department of Orthopedics, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, 400038, China
| | - YuSheng Yang
- National & Regional United Engineering Lab of Tissue Engineering, Department of Orthopedics, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, 400038, China
| | - QiJie Dai
- National & Regional United Engineering Lab of Tissue Engineering, Department of Orthopedics, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, 400038, China
| | - Ce Dou
- National & Regional United Engineering Lab of Tissue Engineering, Department of Orthopedics, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, 400038, China
| | - Fei Luo
- National & Regional United Engineering Lab of Tissue Engineering, Department of Orthopedics, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, 400038, China.
| |
Collapse
|
107
|
Wang L, Jiang Q, Chen S, Wang S, Lu J, Gao X, Zhang D, Jin X. Natural epidithiodiketopiperazine alkaloids as potential anticancer agents: Recent mechanisms of action, structural modification, and synthetic strategies. Bioorg Chem 2023; 137:106642. [PMID: 37276722 DOI: 10.1016/j.bioorg.2023.106642] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2023] [Revised: 05/18/2023] [Accepted: 05/27/2023] [Indexed: 06/07/2023]
Abstract
Cancer has become a grave health crisis that threatens the lives of millions of people worldwide. Because of the drawbacks of the available anticancer drugs, the development of novel and efficient anticancer agents should be encouraged. Epidithiodiketopiperazine (ETP) alkaloids with a 2,5-diketopiperazine (DKP) ring equipped with transannular disulfide or polysulfide bridges or S-methyl moieties constitute a special subclass of fungal natural products. Owing to their privileged sulfur units and intriguing architectural structures, ETP alkaloids exhibit excellent anticancer activities by regulating multiple cancer proteins/signaling pathways, including HIF-1, NF-κB, NOTCH, Wnt, and PI3K/AKT/mTOR, or by inducing cell-cycle arrest, apoptosis, and autophagy. Furthermore, a series of ETP alkaloid derivatives obtained via structural modification showed more potent anticancer activity than natural ETP alkaloids. To solve supply difficulties from natural resources, the total synthetic routes for several ETP alkaloids have been designed. In this review, we summarized several ETP alkaloids with anticancer properties with particular emphasis on their underlying mechanisms of action, structural modifications, and synthetic strategies, which will offer guidance to design and innovate potential anticancer drugs.
Collapse
Affiliation(s)
- Lin Wang
- School of Pharmacy, China Medical University, Shenyang 110122, China
| | - Qinghua Jiang
- Department of Pharmacy, Shengjing Hospital of China Medical University, Shenyang 110004, China
| | - Siyu Chen
- China Medical University-Queen's University of Belfast Joint College, China Medical University, Shenyang 110122, China
| | - Siyi Wang
- The 1st Clinical Department, China Medical University, Shenyang 110122, China
| | - Jingyi Lu
- School of Pharmacy, China Medical University, Shenyang 110122, China
| | - Xun Gao
- Jiangsu Institute Marine Resources Development, Jiangsu Ocean University, Lianyungang 222005, China
| | - Dongfang Zhang
- School of Pharmacy, China Medical University, Shenyang 110122, China.
| | - Xin Jin
- School of Pharmacy, China Medical University, Shenyang 110122, China.
| |
Collapse
|
108
|
Zhan M, Ding Y, Huang S, Liu Y, Xiao J, Yu H, Lu L, Wang X. Lysyl oxidase-like 3 restrains mitochondrial ferroptosis to promote liver cancer chemoresistance by stabilizing dihydroorotate dehydrogenase. Nat Commun 2023; 14:3123. [PMID: 37253718 DOI: 10.1038/s41467-023-38753-6] [Citation(s) in RCA: 29] [Impact Index Per Article: 14.5] [Reference Citation Analysis] [Abstract] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2022] [Accepted: 05/11/2023] [Indexed: 06/01/2023] Open
Abstract
To overcome chemotherapy resistance, novel strategies sensitizing cancer cells to chemotherapy are required. Here, we screen the lysyl-oxidase (LOX) family to clarify its contribution to chemotherapy resistance in liver cancer. LOXL3 depletion significantly sensitizes liver cancer cells to Oxaliplatin by inducing ferroptosis. Chemotherapy-activated EGFR signaling drives LOXL3 to interact with TOM20, causing it to be hijacked into mitochondria, where LOXL3 lysyl-oxidase activity is reinforced by phosphorylation at S704. Metabolic adenylate kinase 2 (AK2) directly phosphorylates LOXL3-S704. Phosphorylated LOXL3-S704 targets dihydroorotate dehydrogenase (DHODH) and stabilizes it by preventing its ubiquitin-mediated proteasomal degradation. K344-deubiquitinated DHODH accumulates in mitochondria, in turn inhibiting chemotherapy-induced mitochondrial ferroptosis. CRISPR-Cas9-mediated site-mutation of mouse LOXL3-S704 to D704 causes a reduction in lipid peroxidation. Using an advanced liver cancer mouse model, we further reveal that low-dose Oxaliplatin in combination with the DHODH-inhibitor Leflunomide effectively inhibit liver cancer progression by inducing ferroptosis, with increased chemotherapy sensitivity and decreased chemotherapy toxicity.
Collapse
Affiliation(s)
- Meixiao Zhan
- Zhuhai Interventional Medical Center, Guangdong Provincial Key Laboratory of Tumour Interventional Diagnosis and Treatment, Zhuhai People's Hospital, Zhuhai Hospital affiliated with Jinan University, Zhuhai, 519000, Guangdong, China
| | - Yufeng Ding
- Precise Genome Engineering Center, School of Life Sciences, Guangzhou University, 510006, Guangzhou, China.
| | - Shanzhou Huang
- Department of General Surgery, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, 510080, Guangzhou, China
| | - Yuhang Liu
- Precise Genome Engineering Center, School of Life Sciences, Guangzhou University, 510006, Guangzhou, China
| | - Jing Xiao
- Zhuhai Interventional Medical Center, Guangdong Provincial Key Laboratory of Tumour Interventional Diagnosis and Treatment, Zhuhai People's Hospital, Zhuhai Hospital affiliated with Jinan University, Zhuhai, 519000, Guangdong, China
| | - Hua Yu
- Precise Genome Engineering Center, School of Life Sciences, Guangzhou University, 510006, Guangzhou, China.
| | - Ligong Lu
- Zhuhai Interventional Medical Center, Guangdong Provincial Key Laboratory of Tumour Interventional Diagnosis and Treatment, Zhuhai People's Hospital, Zhuhai Hospital affiliated with Jinan University, Zhuhai, 519000, Guangdong, China.
| | - Xiongjun Wang
- Precise Genome Engineering Center, School of Life Sciences, Guangzhou University, 510006, Guangzhou, China.
| |
Collapse
|
109
|
Lu B, Tang L, Li L, Zhou X, Leng Y, Quan C. Phosphorylated PKM2 regulates endothelium-dependent vasodilation in diabetes. ZHONG NAN DA XUE XUE BAO. YI XUE BAN = JOURNAL OF CENTRAL SOUTH UNIVERSITY. MEDICAL SCIENCES 2023; 48:663-670. [PMID: 37539568 PMCID: PMC10930405 DOI: 10.11817/j.issn.1672-7347.2023.220541] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Subscribe] [Scholar Register] [Received: 10/19/2022] [Indexed: 08/05/2023]
Abstract
OBJECTIVES Endothelium-dependent vasodilation dysfunction is the pathological basis of diabetic macroangiopathy. The utilization and adaptation of endothelial cells to high glucose determine the functional status of endothelial cells. Glycolysis pathway is the major energy source for endothelial cells. Abnormal glycolysis plays an important role in endothelium-dependent vasodilation dysfunction induced by high glucose. Pyruvate kinase isozyme type M2 (PKM2) is one of key enzymes in glycolysis pathway, phosphorylation of PKM2 can reduce the activity of pyruvate kinase and affect the glycolysis process of glucose. TEPP-46 can stabilize PKM2 in its tetramer form, reducing its dimer formation and phosphorylation. Using TEPP-46 as a tool drug to inhibit PKM2 phosphorylation, this study aims to explore the impact and potential mechanism of phosphorylated PKM2 (p-PKM2) on endothelial dependent vasodilation function in high glucose, and to provide a theoretical basis for finding new intervention targets for diabetic macroangiopathy. METHODS The mice were divided into 3 groups: a wild-type (WT) group (a control group, C57BL/6 mice) and a db/db group (a diabetic group, db/db mice), which were treated with the sodium carboxymethyl cellulose solution (solvent) by gavage once a day, and a TEPP-46 group (a treatment group, db/db mice+TEPP-46), which was gavaged with TEPP-46 (30 mg/kg) and sodium carboxymethyl cellulose solution once a day. After 12 weeks of treatment, the levels of p-PKM2 and PKM2 protein in thoracic aortas, plasma nitric oxide (NO) level and endothelium-dependent vasodilation function of thoracic aortas were detected. High glucose (30 mmol/L) with or without TEPP-46 (10 μmol/L), mannitol incubating human umbilical vein endothelial cells (HUVECs) for 72 hours, respectively. The level of NO in supernatant, the content of NO in cells, and the levels of p-PKM2 and PKM2 protein were detected. Finally, the effect of TEPP-46 on endothelial nitric oxide synthase (eNOS) phosphorylation was detected at the cellular and animal levels. RESULTS Compared with the control group, the levels of p-PKM2 in thoracic aortas of the diabetic group increased (P<0.05). The responsiveness of thoracic aortas in the diabetic group to acetylcholine (ACh) was 47% lower than that in the control group (P<0.05), and that in TEPP-46 treatment group was 28% higher than that in the diabetic group (P<0.05), while there was no statistically significant difference in the responsiveness of thoracic aortas to sodium nitroprusside (SNP). Compared with the control group, the plasma NO level of mice decreased in the diabetic group, while compared with the diabetic group, the phosphorylation of PKM2 in thoracic aortas decreased and the plasma NO level increased in the TEPP-46 group (both P<0.05). High glucose instead of mannitol induced the increase of PKM2 phosphorylation in HUVECs and reduced the level of NO in supernatant (both P<0.05). HUVECs incubated with TEPP-46 and high glucose reversed the reduction of NO production and secretion induced by high glucose while inhibiting PKM2 phosphorylation (both P<0.05). At the cellular and animal levels, TEPP-46 reversed the decrease of eNOS (ser1177) phosphorylation induced by high glucose (both P<0.05). CONCLUSIONS p-PKM2 may be involved in the process of endothelium-dependent vasodilation dysfunction in Type 2 diabetes by inhibiting p-eNOS (ser1177)/NO pathway.
Collapse
Affiliation(s)
- Bin Lu
- Vascular Disease and Transformation Medical Center, Third Xiangya Hospital, Central South University, Changsha 410013.
| | - Lei Tang
- Vascular Disease and Transformation Medical Center, Third Xiangya Hospital, Central South University, Changsha 410013
| | - Le Li
- Department of Cardiology, Affiliated Hospital of Southwest Medical University, Luzhou Sichuan 646000
| | - Xiaoyu Zhou
- Vascular Disease and Transformation Medical Center, Third Xiangya Hospital, Central South University, Changsha 410013
| | - Yiping Leng
- Research Center for Phase Ⅰ Clinical Trials, Affiliated Changsha Central Hospital, University of South China, Changsha 410004
| | - Chengxuan Quan
- Vascular Disease and Transformation Medical Center, Third Xiangya Hospital, Central South University, Changsha 410013.
- Department of Anesthesiology, Third Xiangya Hospital, Central South University, Changsha 410013, China.
| |
Collapse
|
110
|
Iacobini C, Vitale M, Pugliese G, Menini S. The "sweet" path to cancer: focus on cellular glucose metabolism. Front Oncol 2023; 13:1202093. [PMID: 37305566 PMCID: PMC10248238 DOI: 10.3389/fonc.2023.1202093] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2023] [Accepted: 05/17/2023] [Indexed: 06/13/2023] Open
Abstract
The hypoxia-inducible factor-1α (HIF-1α), a key player in the adaptive regulation of energy metabolism, and the M2 isoform of the glycolytic enzyme pyruvate kinase (PKM2), a critical regulator of glucose consumption, are the main drivers of the metabolic rewiring in cancer cells. The use of glycolysis rather than oxidative phosphorylation, even in the presence of oxygen (i.e., Warburg effect or aerobic glycolysis), is a major metabolic hallmark of cancer. Aerobic glycolysis is also important for the immune system, which is involved in both metabolic disorders development and tumorigenesis. More recently, metabolic changes resembling the Warburg effect have been described in diabetes mellitus (DM). Scientists from different disciplines are looking for ways to interfere with these cellular metabolic rearrangements and reverse the pathological processes underlying their disease of interest. As cancer is overtaking cardiovascular disease as the leading cause of excess death in DM, and biological links between DM and cancer are incompletely understood, cellular glucose metabolism may be a promising field to explore in search of connections between cardiometabolic and cancer diseases. In this mini-review, we present the state-of-the-art on the role of the Warburg effect, HIF-1α, and PKM2 in cancer, inflammation, and DM to encourage multidisciplinary research to advance fundamental understanding in biology and pathways implicated in the link between DM and cancer.
Collapse
|
111
|
Liang J, Ye C, Chen K, Gao Z, Lu F, Wei K. Non-coding RNAs in breast cancer: with a focus on glucose metabolism reprogramming. Discov Oncol 2023; 14:72. [PMID: 37204526 DOI: 10.1007/s12672-023-00687-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/02/2023] [Accepted: 05/10/2023] [Indexed: 05/20/2023] Open
Abstract
Breast cancer is the tumor with the highest incidence in women worldwide. According to research, the poor prognosis of breast cancer is closely related to abnormal glucose metabolism in tumor cells. Changes in glucose metabolism in tumor cells are an important feature. When sufficient oxygen is available, cancer cells tend to undergo glycolysis rather than oxidative phosphorylation, which promotes rapid proliferation and invasion of tumor cells. As research deepens, targeting the glucose metabolism pathway of tumor cells is seen as a promising treatment. Non-coding RNAs (ncRNAs), a recent focus of research, are involved in the regulation of enzymes of glucose metabolism and related cancer signaling pathways in breast cancer cells. This article reviews the regulatory effect and mechanism of ncRNAs on glucose metabolism in breast cancer cells and provides new ideas for the treatment of breast cancer.
Collapse
Affiliation(s)
- Junjie Liang
- Medical School, Hunan University of Chinese Medicine, Changsha, 410208, China
| | - Chun Ye
- Medical School, Hunan University of Chinese Medicine, Changsha, 410208, China
| | - Kaiqin Chen
- Medical School, Hunan University of Chinese Medicine, Changsha, 410208, China
| | - Zihan Gao
- Medical School, Hunan University of Chinese Medicine, Changsha, 410208, China
| | - Fangguo Lu
- Medical School, Hunan University of Chinese Medicine, Changsha, 410208, China
| | - Ke Wei
- Medical School, Hunan University of Chinese Medicine, Changsha, 410208, China.
- Hunan Province Key Laboratory of Integrative Pathogen Biology, Hunan University of Chinese Medicine, Changsha, 410208, Hunan, China.
| |
Collapse
|
112
|
Ko Y, Hong M, Lee S, Kumar M, Ibrahim L, Nutsch K, Stanton C, Sondermann P, Sandoval B, Bulos ML, Iaconelli J, Chatterjee AK, Wiseman RL, Schultz PG, Bollong MJ. S-lactoyl modification of KEAP1 by a reactive glycolytic metabolite activates NRF2 signaling. Proc Natl Acad Sci U S A 2023; 120:e2300763120. [PMID: 37155889 PMCID: PMC10193962 DOI: 10.1073/pnas.2300763120] [Citation(s) in RCA: 18] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2023] [Accepted: 04/05/2023] [Indexed: 05/10/2023] Open
Abstract
KEAP1 (Kelch-like ECH-associated protein), a cytoplasmic repressor of the oxidative stress responsive transcription factor Nuclear factor erythroid 2-related factor 2 (NRF2), senses the presence of electrophilic agents by modification of its sensor cysteine residues. In addition to xenobiotics, several reactive metabolites have been shown to covalently modify key cysteines on KEAP1, although the full repertoire of these molecules and their respective modifications remain undefined. Here, we report the discovery of sAKZ692, a small molecule identified by high-throughput screening that stimulates NRF2 transcriptional activity in cells by inhibiting the glycolytic enzyme pyruvate kinase. sAKZ692 treatment promotes the buildup of glyceraldehyde 3-phosphate, a metabolite which leads to S-lactate modification of cysteine sensor residues of KEAP1, resulting in NRF2-dependent transcription. This work identifies a posttranslational modification of cysteine derived from a reactive central carbon metabolite and helps further define the complex relationship between metabolism and the oxidative stress-sensing machinery of the cell.
Collapse
Affiliation(s)
- Yeonjin Ko
- Department of Chemistry, The Scripps Research Institute, San Diego, CA92037
| | - Mannkyu Hong
- Department of Chemistry, The Scripps Research Institute, San Diego, CA92037
| | - Seungbeom Lee
- Department of Chemistry, The Scripps Research Institute, San Diego, CA92037
| | - Manoj Kumar
- Calibr, A Division of Scripps Research, San Diego, CA92037
| | - Lara Ibrahim
- Department of Chemistry, The Scripps Research Institute, San Diego, CA92037
- Department of Molecular Medicine, The Scripps Research Institute, San Diego, CA92037
| | - Kayla Nutsch
- Department of Chemistry, The Scripps Research Institute, San Diego, CA92037
| | - Caroline Stanton
- Department of Chemistry, The Scripps Research Institute, San Diego, CA92037
- Department of Molecular Medicine, The Scripps Research Institute, San Diego, CA92037
| | - Phillip Sondermann
- Department of Chemistry, The Scripps Research Institute, San Diego, CA92037
| | - Braddock Sandoval
- Department of Chemistry, The Scripps Research Institute, San Diego, CA92037
| | - Maya L. Bulos
- Department of Chemistry, The Scripps Research Institute, San Diego, CA92037
| | - Jonathan Iaconelli
- Department of Chemistry, The Scripps Research Institute, San Diego, CA92037
| | | | - R. Luke Wiseman
- Department of Molecular Medicine, The Scripps Research Institute, San Diego, CA92037
| | - Peter G. Schultz
- Department of Chemistry, The Scripps Research Institute, San Diego, CA92037
- Calibr, A Division of Scripps Research, San Diego, CA92037
| | - Michael J. Bollong
- Department of Chemistry, The Scripps Research Institute, San Diego, CA92037
| |
Collapse
|
113
|
Li Y, Tan R, Li R, Tian R, Liu Z, Wang X, Chen E, Pan T, Qu H. PKM2/STAT1-mediated PD-L1 upregulation on neutrophils during sepsis promotes neutrophil organ accumulation by serving an anti-apoptotic role. J Inflamm (Lond) 2023; 20:16. [PMID: 37131151 PMCID: PMC10155438 DOI: 10.1186/s12950-023-00341-2] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2022] [Accepted: 04/25/2023] [Indexed: 05/04/2023] Open
Abstract
BACKGROUND Delayed neutrophil apoptosis during sepsis may impact neutrophil organ accumulation and tissue immune homeostasis. Elucidating the mechanisms underlying neutrophil apoptosis may help identify potential therapeutic targets. Glycolysis is critical to neutrophil activities during sepsis. However, the precise mechanisms through which glycolysis regulates neutrophil physiology remain under-explored, especially those involving the non-metabolic functions of glycolytic enzymes. In the present study, the impact of programmed death ligand-1 (PD-L1) on neutrophil apoptosis was explored. The regulatory effect of the glycolytic enzyme, pyruvate kinase M2 (PKM2), whose role in septic neutrophils remains unaddressed, on neutrophil PD-L1 expression was also explored. METHODS Peripheral blood neutrophils were isolated from patients with sepsis and healthy controls. PD-L1 and PKM2 levels were determined by flow cytometry and Western blotting, respectively. Dimethyl sulfoxide (DMSO)-differentiated HL-60 cells were stimulated with lipopolysaccharide (LPS) as an in vitro simulation of septic neutrophils. Cell apoptosis was assessed by annexin V/propidium iodide (annexin V/PI) staining, as well as determination of protein levels of cleaved caspase-3 and myeloid cell leukemia-1 (Mcl-1) by Western blotting. An in vivo model of sepsis was constructed by intraperitoneal injection of LPS (5 mg/kg) for 16 h. Pulmonary and hepatic neutrophil infiltration was assessed by flow cytometry or immunohistochemistry. RESULTS PD-L1 level was elevated on neutrophils under septic conditions. Administration of neutralizing antibodies against PD-L1 partially reversed the inhibitory effect of LPS on neutrophil apoptosis. Neutrophil infiltration into the lung and liver was also reduced in PD-L1-/- mice 16 h after sepsis induction. PKM2 was upregulated in septic neutrophils and promoted neutrophil PD-L1 expression both in vitro and in vivo. In addition, PKM2 nuclear translocation was increased after LPS stimulation, which promoted PD-L1 expression by directly interacting with and activating signal transducer and activator of transcription 1 (STAT1). Inhibition of PKM2 activity or STAT1 activation also led to increased neutrophil apoptosis. CONCLUSION In this study, a PKM2/STAT1-mediated upregulation of PD-L1 on neutrophils and the anti-apoptotic effect of upregulated PD-L1 on neutrophils during sepsis were identified, which may result in increased pulmonary and hepatic neutrophil accumulation. These findings suggest that PKM2 and PD-L1 could serve as potential therapeutic targets.
Collapse
Affiliation(s)
- Yinjiaozhi Li
- Department of Critical Care Medicine, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, 197 Ruijin Er Road, Shanghai, 200025, China
| | - Ruoming Tan
- Department of Critical Care Medicine, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, 197 Ruijin Er Road, Shanghai, 200025, China
| | - Ranran Li
- Department of Critical Care Medicine, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, 197 Ruijin Er Road, Shanghai, 200025, China
| | - Rui Tian
- Department of Critical Care Medicine, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, 197 Ruijin Er Road, Shanghai, 200025, China
| | - Zhaojun Liu
- Department of Critical Care Medicine, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, 197 Ruijin Er Road, Shanghai, 200025, China
| | - Xiaoli Wang
- Department of Critical Care Medicine, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, 197 Ruijin Er Road, Shanghai, 200025, China
| | - Erzhen Chen
- Department of Emergency Medicine, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, 197 Ruijin Er Road, Shanghai, 200025, China
| | - Tingting Pan
- Department of Critical Care Medicine, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, 197 Ruijin Er Road, Shanghai, 200025, China.
| | - Hongping Qu
- Department of Critical Care Medicine, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, 197 Ruijin Er Road, Shanghai, 200025, China.
| |
Collapse
|
114
|
Kim JH, Han J, Afridi R, Kim JH, Rahman MH, Park DH, Lee WS, Song GJ, Suk K. A multiplexed siRNA screen identifies key kinase signaling networks of brain glia. Life Sci Alliance 2023; 6:e202201605. [PMID: 36878638 PMCID: PMC9990460 DOI: 10.26508/lsa.202201605] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2022] [Revised: 02/22/2023] [Accepted: 02/22/2023] [Indexed: 03/08/2023] Open
Abstract
The dynamic behaviors of brain glial cells in various neuroinflammatory conditions and neurological disorders have been reported; however, little is known about the underlying intracellular signaling pathways. Here, we developed a multiplexed kinome-wide siRNA screen to identify the kinases regulating several inflammatory phenotypes of mouse glial cells in culture, including inflammatory activation, migration, and phagocytosis of glia. Subsequent proof-of-concept experiments involving genetic and pharmacological inhibitions indicated the importance of T-cell receptor signaling components in microglial activation and a metabolic shift from glycolysis to oxidative phosphorylation in astrocyte migration. This time- and cost-effective multiplexed kinome siRNA screen efficiently provides exploitable drug targets and novel insight into the mechanisms underlying the phenotypic regulation of glial cells and neuroinflammation. Moreover, the kinases identified in this screen may be relevant in other inflammatory diseases and cancer, wherein kinases play a critical role in disease signaling pathways.
Collapse
Affiliation(s)
- Jong-Heon Kim
- Brain Science & Engineering Institute, Kyungpook National University, Daegu, Republic of Korea
| | - Jin Han
- Department of Biomedical Science, Department of Pharmacology, School of Medicine, Kyungpook National University, Daegu, Republic of Korea
| | - Ruqayya Afridi
- Department of Biomedical Science, Department of Pharmacology, School of Medicine, Kyungpook National University, Daegu, Republic of Korea
| | - Jae-Hong Kim
- Brain Science & Engineering Institute, Kyungpook National University, Daegu, Republic of Korea
| | - Md Habibur Rahman
- Brain Science & Engineering Institute, Kyungpook National University, Daegu, Republic of Korea
- Department of Neurology, The Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Dong Ho Park
- Department of Ophthalmology, School of Medicine, Kyungpook National University, Kyungpook National University Hospital, Daegu, Republic of Korea
| | - Won Suk Lee
- Neuracle Science Co., Ltd. Seoul, Republic Korea
| | - Gyun Jee Song
- Department of Medical Science, College of Medicine, Catholic Kwandong University, Gangneung-si, Republic Korea; Translational Brain Research Center, International St. Mary's Hospital, Catholic Kwandong University, Incheon, Republic Korea
| | - Kyoungho Suk
- Brain Science & Engineering Institute, Kyungpook National University, Daegu, Republic of Korea
- Department of Biomedical Science, Department of Pharmacology, School of Medicine, Kyungpook National University, Daegu, Republic of Korea
| |
Collapse
|
115
|
Dai Y, Liu P, Wen W, Li P, Yang C, Wang P, Xu S. Sarsasapogenin, a principal active component absorbed into blood of total saponins of Anemarrhena, attenuates proliferation and invasion in rheumatoid arthritis fibroblast-like synoviocytes through downregulating PKM2 inhibited pathological glycolysis. Phytother Res 2023; 37:1951-1967. [PMID: 36631974 DOI: 10.1002/ptr.7712] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2022] [Revised: 12/12/2022] [Accepted: 12/12/2022] [Indexed: 01/13/2023]
Abstract
Increased glycolytic in fibroblast-like synoviocytes (FLS) of rheumatoid arthritis (RA) not only contributes to early-stage disease pathogenesis but leads to sustained proliferation of FLS. Given the importance of PKM2 in glycolysis and apoptosis, PKM2 is considered a potential therapeutic and drug discovery target in RA. Total saponins of anemarrhena (TSA), a class of steroid saponins, originated from Anemarrhena asphodeloides Bge. In this study, we verified that 200 mg/kg TSA could significantly alleviate inflammation and the pathological characteristics of RA and inhibit synovial hyperplasia in AA rats. We confirmed that sarsasapogenin (SA) was the principal active ingredient absorbed into the blood of TSA by the UPLC/Q Exactive MS test. Then we used TNF-α-induced MH7A to get the conclusion that 20 μM SA could effectively inhibit the glycolysis by inhibiting the activity of PKM2 tetramer and glucose uptake. Moreover, 20 μM SA could suppress proliferation, migration, invasion, and cytokine release of FLS, interfere with the growth cycle of FLS, and induce FLS apoptosis by depressing the phosphorylation of PKM2. At last, In-1, a potent inhibitor of the PKM2 was used to reverse verify the above results. Taken together, the key mechanisms of SA on RA treatment through downregulating the activity of PKM2 tetramer and phosphorylation of PKM2 inhibited pathological glycolysis and induced apoptosis to exert inhibition on the proliferation and invasion of RA FLS.
Collapse
Affiliation(s)
- Yuan Dai
- School of Health Preservation and Rehabilitation, Chengdu University of Traditional Chinese Medicine, Chengdu, China
- Institute of Meterial Medica Integration and Transformation for Brain Disorders, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Panwang Liu
- Institute of Meterial Medica Integration and Transformation for Brain Disorders, Chengdu University of Traditional Chinese Medicine, Chengdu, China
- School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Wen Wen
- Institute of Meterial Medica Integration and Transformation for Brain Disorders, Chengdu University of Traditional Chinese Medicine, Chengdu, China
- School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Ping Li
- Institute of Meterial Medica Integration and Transformation for Brain Disorders, Chengdu University of Traditional Chinese Medicine, Chengdu, China
- School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Chen Yang
- Antibiotics Research and Re-evaluation Key Laboratory of Sichuan Province, Sichuan Industrial Institute of Antibiotics, Chengdu University, Chengdu, China
| | - Ping Wang
- Institute of Meterial Medica Integration and Transformation for Brain Disorders, Chengdu University of Traditional Chinese Medicine, Chengdu, China
- School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Shijun Xu
- Institute of Meterial Medica Integration and Transformation for Brain Disorders, Chengdu University of Traditional Chinese Medicine, Chengdu, China
- School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| |
Collapse
|
116
|
Cheng Z, Huang H, Li M, Liang X, Tan Y, Chen Y. Lactylation-Related Gene Signature Effectively Predicts Prognosis and Treatment Responsiveness in Hepatocellular Carcinoma. Pharmaceuticals (Basel) 2023; 16:ph16050644. [PMID: 37242427 DOI: 10.3390/ph16050644] [Citation(s) in RCA: 23] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2023] [Revised: 04/20/2023] [Accepted: 04/23/2023] [Indexed: 05/28/2023] Open
Abstract
BACKGROUND Hepatocellular carcinoma (HCC) is a malignant tumor associated with high morbidity and mortality. Therefore, it is of great importance to develop effective prognostic models and guide clinical treatment in HCC. Protein lactylation is found in HCC tumors and is associated with HCC progression. METHODS The expression levels of lactylation-related genes were identified from the TCGA database. A lactylation-related gene signature was constructed using LASSO regression. The prognostic value of the model was assessed and further validated in the ICGC cohort, with the patients split into two groups based on risk score. Glycolysis and immune pathways, treatment responsiveness, and the mutation of signature genes were analyzed. The correlation between PKM2 expression and the clinical characteristics was investigated. RESULTS Sixteen prognostic differentially expressed lactylation-related genes were identified. An 8-gene signature was constructed and validated. Patients with higher risk scores had poorer clinical outcomes. The two groups were different in immune cell abundance. The high-risk group patients were more sensitive to most chemical drugs and sorafenib, while the low-risk group patients were more sensitive to some targeted drugs such as lapatinib and FH535. Moreover, the low-risk group had a higher TIDE score and was more sensitive to immunotherapy. PKM2 expression correlated with clinical characteristics and immune cell abundance in the HCC samples. CONCLUSIONS The lactylation-related model exhibited robust predictive efficiency in HCC. The glycolysis pathway was enriched in the HCC tumor samples. A low-risk score indicated better treatment response to most targeted drugs and immunotherapy. The lactylation-related gene signature could be used as a biomarker for the effective clinical treatment of HCC.
Collapse
Affiliation(s)
- Zhe Cheng
- Department of Oncology, NHC Key Laboratory of Cancer Proteomics, Laboratory of Structural Biology, National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha 410008, China
| | - Huichao Huang
- Department of Infectious Disease, NHC Key Laboratory of Cancer Proteomics, Laboratory of Structural Biology, National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha 410008, China
| | - Maoyu Li
- Department of Oncology, NHC Key Laboratory of Cancer Proteomics, Laboratory of Structural Biology, National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha 410008, China
| | - Xujun Liang
- Department of Oncology, NHC Key Laboratory of Cancer Proteomics, Laboratory of Structural Biology, National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha 410008, China
| | - Yuying Tan
- Department of Oncology, NHC Key Laboratory of Cancer Proteomics, Laboratory of Structural Biology, National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha 410008, China
| | - Yongheng Chen
- Department of Oncology, NHC Key Laboratory of Cancer Proteomics, Laboratory of Structural Biology, National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha 410008, China
| |
Collapse
|
117
|
Ren C, Tan P, Gao L, Zeng Y, Hu S, Chen C, Tang N, Chen Y, Zhang W, Qin Y, Zhang X, Du S. Melatonin reduces radiation-induced ferroptosis in hippocampal neurons by activating the PKM2/NRF2/GPX4 signaling pathway. Prog Neuropsychopharmacol Biol Psychiatry 2023; 126:110777. [PMID: 37100272 DOI: 10.1016/j.pnpbp.2023.110777] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/13/2022] [Revised: 04/19/2023] [Accepted: 04/21/2023] [Indexed: 04/28/2023]
Abstract
Ferroptosis is a type of regulated cell death that is dependent on iron and reactive oxygen species (ROS). Melatonin (N-acetyl-5-methoxytryptamine) reduces hypoxic-ischemic brain damage via mechanisms that involve free radical scavenging. How melatonin regulates radiation-induced ferroptosis of hippocampal neurons is yet to be elucidated. In this study, the mouse hippocampal neuronal cell line HT-22 was treated with 20μM melatonin before being stimulated with a combination of irradiation and 100 μM FeCl3. Furthermore, in vivo experiments were performed in mice treated with melatonin via intraperitoneal injection, which was followed by radiation exposure. A series of functional assays, including CCK-8, DCFH-DA kit, flow cytometry, TUNEL staining, iron estimations, and transmission electron microscopy, were performed on cells as well as hippocampal tissues. The interactions between PKM2 and NRF2 proteins were detected using a coimmunoprecipitation (Co-IP) assay. Moreover, chromatin immunoprecipitation (ChIP), a luciferase reporter assay, and an electrophoretic mobility shift assay (EMSA) were performed to explore the mechanism by which PKM2 regulates the NRF2/GPX4 signaling pathway. The spatial memory of mice was evaluated using the Morris Water Maze test. Hematoxylin-eosin and Nissl staining were performed for histological examination. The results revealed that melatonin protected HT-22 neuronal cells from radiation-induced ferroptosis, as inferred from increased cell viability, decreased ROS production, reduced number of apoptotic cells, and less cristae, higher electron density in mitochondria. In addition, melatonin induced PKM2 nuclear transference, while PKM2 inhibition reversed the effects of melatonin. Further experiments demonstrated that PKM2 bound to and induced the nuclear translocation of NRF2, which regulated GPX4 transcription. Ferroptosis enhanced by PKM2 inhibition was also converted by NRF2 overexpression. In vivo experiments indicated that melatonin alleviated radiation-induced neurological dysfunction and injury in mice. In conclusion, melatonin suppressed ferroptosis to decrease radiation-induced hippocampal neuronal injury by activating the PKM2/NRF2/GPX4 signaling pathway.
Collapse
Affiliation(s)
- Chen Ren
- Department of Radiation Oncology, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou 510080, Guangdong, China
| | - Peixin Tan
- Department of Radiation Oncology, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou 510080, Guangdong, China
| | - Lianxuan Gao
- Department of Radiation Oncology, Nanfang Hospital, Southern Medical University, Guangzhou 510515, Guangdong, China
| | - Yingying Zeng
- Department of Radiation Oncology, Nanfang Hospital, Southern Medical University, Guangzhou 510515, Guangdong, China
| | - Shushu Hu
- Department of Radiation Oncology, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou 510080, Guangdong, China
| | - Chen Chen
- Department of Radiation Oncology, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou 510080, Guangdong, China
| | - Nan Tang
- Department of Radiation Oncology, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou 510080, Guangdong, China
| | - Yulei Chen
- Department of Radiation Oncology, Nanfang Hospital, Southern Medical University, Guangzhou 510515, Guangdong, China
| | - Wan Zhang
- Department of Radiation Oncology, Nanfang Hospital, Southern Medical University, Guangzhou 510515, Guangdong, China
| | - Yue Qin
- Department of Radiation Oncology, Nanfang Hospital, Southern Medical University, Guangzhou 510515, Guangdong, China
| | - Xiaonan Zhang
- Department of Radiation Oncology, Nanfang Hospital, Southern Medical University, Guangzhou 510515, Guangdong, China
| | - Shasha Du
- Department of Radiation Oncology, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou 510080, Guangdong, China.
| |
Collapse
|
118
|
Buneeva O, Kopylov A, Gnedenko O, Medvedeva M, Veselovsky A, Ivanov A, Zgoda V, Medvedev A. Proteomic Profiling of Mouse Brain Pyruvate Kinase Binding Proteins: A Hint for Moonlighting Functions of PKM1? Int J Mol Sci 2023; 24:ijms24087634. [PMID: 37108803 PMCID: PMC10143413 DOI: 10.3390/ijms24087634] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2023] [Revised: 04/18/2023] [Accepted: 04/19/2023] [Indexed: 04/29/2023] Open
Abstract
Affinity-based proteomic profiling is widely used for the identification of proteins involved in the formation of various interactomes. Since protein-protein interactions (PPIs) reflect the role of particular proteins in the cell, identification of interaction partners for a protein of interest can reveal its function. The latter is especially important for the characterization of multifunctional proteins, which can play different roles in the cell. Pyruvate kinase (PK), a classical glycolytic enzyme catalyzing the last step of glycolysis, exists in four isoforms: PKM1, PKM2, PKL, and PKR. The enzyme isoform expressed in actively dividing cells, PKM2, exhibits many moonlighting (noncanonical) functions. In contrast to PKM2, PKM1, predominantly expressed in adult differentiated tissues, lacks well-documented moonlighting functions. However, certain evidence exists that it can also perform some functions unrelated to glycolysis. In order to evaluate protein partners, bound to PKM1, in this study we have combined affinity-based separation of mouse brain proteins with mass spectrometry identification. The highly purified PKM1 and a 32-mer synthetic peptide (PK peptide), sharing high sequence homology with the interface contact region of all PK isoforms, were used as the affinity ligands. This proteomic profiling resulted in the identification of specific and common proteins bound to both affinity ligands. Quantitative affinity binding to the affinity ligands of selected identified proteins was validated using a surface plasmon resonance (SPR) biosensor. Bioinformatic analysis has shown that the identified proteins, bound to both full-length PKM1 and the PK peptide, form a protein network (interactome). Some of these interactions are relevant for the moonlighting functions of PKM1. The proteomic dataset is available via ProteomeXchange with the identifier PXD041321.
Collapse
Affiliation(s)
- Olga Buneeva
- Institute of Biomedical Chemistry, 10 Pogodinskaya Street, Moscow 119121, Russia
| | - Arthur Kopylov
- Institute of Biomedical Chemistry, 10 Pogodinskaya Street, Moscow 119121, Russia
| | - Oksana Gnedenko
- Institute of Biomedical Chemistry, 10 Pogodinskaya Street, Moscow 119121, Russia
| | - Marina Medvedeva
- Department of Biochemistry, School of Biology, Moscow State University, Moscow 119991, Russia
| | - Alexander Veselovsky
- Institute of Biomedical Chemistry, 10 Pogodinskaya Street, Moscow 119121, Russia
| | - Alexis Ivanov
- Institute of Biomedical Chemistry, 10 Pogodinskaya Street, Moscow 119121, Russia
| | - Victor Zgoda
- Institute of Biomedical Chemistry, 10 Pogodinskaya Street, Moscow 119121, Russia
| | - Alexei Medvedev
- Institute of Biomedical Chemistry, 10 Pogodinskaya Street, Moscow 119121, Russia
| |
Collapse
|
119
|
Marcucci F, Rumio C. On the Role of Glycolysis in Early Tumorigenesis-Permissive and Executioner Effects. Cells 2023; 12:cells12081124. [PMID: 37190033 DOI: 10.3390/cells12081124] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2023] [Revised: 03/26/2023] [Accepted: 04/06/2023] [Indexed: 05/17/2023] Open
Abstract
Reprogramming energy production from mitochondrial respiration to glycolysis is now considered a hallmark of cancer. When tumors grow beyond a certain size they give rise to changes in their microenvironment (e.g., hypoxia, mechanical stress) that are conducive to the upregulation of glycolysis. Over the years, however, it has become clear that glycolysis can also associate with the earliest steps of tumorigenesis. Thus, many of the oncoproteins most commonly involved in tumor initiation and progression upregulate glycolysis. Moreover, in recent years, considerable evidence has been reported suggesting that upregulated glycolysis itself, through its enzymes and/or metabolites, may play a causative role in tumorigenesis, either by acting itself as an oncogenic stimulus or by facilitating the appearance of oncogenic mutations. In fact, several changes induced by upregulated glycolysis have been shown to be involved in tumor initiation and early tumorigenesis: glycolysis-induced chromatin remodeling, inhibition of premature senescence and induction of proliferation, effects on DNA repair, O-linked N-acetylglucosamine modification of target proteins, antiapoptotic effects, induction of epithelial-mesenchymal transition or autophagy, and induction of angiogenesis. In this article we summarize the evidence that upregulated glycolysis is involved in tumor initiation and, in the following, we propose a mechanistic model aimed at explaining how upregulated glycolysis may play such a role.
Collapse
Affiliation(s)
- Fabrizio Marcucci
- Department of Pharmacological and Biomolecular Sciences, University of Milan, Via Trentacoste 2, 20134 Milan, Italy
| | - Cristiano Rumio
- Department of Pharmacological and Biomolecular Sciences, University of Milan, Via Trentacoste 2, 20134 Milan, Italy
| |
Collapse
|
120
|
Nong S, Han X, Xiang Y, Qian Y, Wei Y, Zhang T, Tian K, Shen K, Yang J, Ma X. Metabolic reprogramming in cancer: Mechanisms and therapeutics. MedComm (Beijing) 2023; 4:e218. [PMID: 36994237 PMCID: PMC10041388 DOI: 10.1002/mco2.218] [Citation(s) in RCA: 43] [Impact Index Per Article: 21.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2022] [Revised: 01/22/2023] [Accepted: 01/30/2023] [Indexed: 03/29/2023] Open
Abstract
Cancer cells characterized by uncontrolled growth and proliferation require altered metabolic processes to maintain this characteristic. Metabolic reprogramming is a process mediated by various factors, including oncogenes, tumor suppressor genes, changes in growth factors, and tumor-host cell interactions, which help to meet the needs of cancer cell anabolism and promote tumor development. Metabolic reprogramming in tumor cells is dynamically variable, depending on the tumor type and microenvironment, and reprogramming involves multiple metabolic pathways. These metabolic pathways have complex mechanisms and involve the coordination of various signaling molecules, proteins, and enzymes, which increases the resistance of tumor cells to traditional antitumor therapies. With the development of cancer therapies, metabolic reprogramming has been recognized as a new therapeutic target for metabolic changes in tumor cells. Therefore, understanding how multiple metabolic pathways in cancer cells change can provide a reference for the development of new therapies for tumor treatment. Here, we systemically reviewed the metabolic changes and their alteration factors, together with the current tumor regulation treatments and other possible treatments that are still under investigation. Continuous efforts are needed to further explore the mechanism of cancer metabolism reprogramming and corresponding metabolic treatments.
Collapse
Affiliation(s)
- Shiqi Nong
- State Key Laboratory of Oral DiseasesWest China Hospital of StomatologyWest China School of StomatologyNational Clinical Research Center for Oral DiseasesSichuan UniversityChengduSichuanChina
| | - Xiaoyue Han
- State Key Laboratory of Oral DiseasesWest China Hospital of StomatologyWest China School of StomatologyNational Clinical Research Center for Oral DiseasesSichuan UniversityChengduSichuanChina
| | - Yu Xiang
- Department of BiotherapyCancer CenterWest China HospitalSichuan UniversityChengduSichuanChina
| | - Yuran Qian
- State Key Laboratory of Oral DiseasesWest China Hospital of StomatologyWest China School of StomatologyNational Clinical Research Center for Oral DiseasesSichuan UniversityChengduSichuanChina
| | - Yuhao Wei
- Department of Clinical MedicineWest China School of MedicineWest China HospitalSichuan UniversityChengduSichuanChina
| | - Tingyue Zhang
- State Key Laboratory of Oral DiseasesWest China Hospital of StomatologyWest China School of StomatologyNational Clinical Research Center for Oral DiseasesSichuan UniversityChengduSichuanChina
| | - Keyue Tian
- State Key Laboratory of Oral DiseasesWest China Hospital of StomatologyWest China School of StomatologyNational Clinical Research Center for Oral DiseasesSichuan UniversityChengduSichuanChina
| | - Kai Shen
- Department of OncologyFirst Affiliated Hospital of Nanjing Medical UniversityNanjingChina
| | - Jing Yang
- State Key Laboratory of Oncology in South ChinaCollaborative Innovation Center for Cancer MedicineSun Yat‐sen University Cancer CenterGuangzhouChina
| | - Xuelei Ma
- State Key Laboratory of Oral DiseasesWest China Hospital of StomatologyWest China School of StomatologyNational Clinical Research Center for Oral DiseasesSichuan UniversityChengduSichuanChina
- Department of Biotherapy and Cancer CenterState Key Laboratory of BiotherapyCancer CenterWest China HospitalSichuan UniversityChengduSichuanChina
| |
Collapse
|
121
|
Kuo CW, Chen DH, Tsai MT, Lin CC, Cheng HW, Tsay YG, Wang HT. Pyruvate kinase M2 modification by a lipid peroxidation byproduct acrolein contributes to kidney fibrosis. Front Med (Lausanne) 2023; 10:1151359. [PMID: 37007793 PMCID: PMC10050374 DOI: 10.3389/fmed.2023.1151359] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2023] [Accepted: 02/22/2023] [Indexed: 03/17/2023] Open
Abstract
Renal fibrosis is a hallmark of diabetic nephropathy (DN) and is characterized by an epithelial-to-mesenchymal transition (EMT) program and aberrant glycolysis. The underlying mechanisms of renal fibrosis are still poorly understood, and existing treatments are only marginally effective. Therefore, it is crucial to comprehend the pathophysiological mechanisms behind the development of renal fibrosis and to generate novel therapeutic approaches. Acrolein, an α-,β-unsaturated aldehyde, is endogenously produced during lipid peroxidation. Acrolein shows high reactivity with proteins to form acrolein-protein conjugates (Acr-PCs), resulting in alterations in protein function. In previous research, we found elevated levels of Acr-PCs along with kidney injuries in high-fat diet-streptozotocin (HFD-STZ)-induced DN mice. This study used a proteomic approach with an anti-Acr-PC antibody followed by liquid chromatography–tandem mass spectrometry (LC–MS/MS) analysis to identify several acrolein-modified protein targets. Among these protein targets, pyruvate kinase M2 (PKM2) was found to be modified by acrolein at Cys358, leading to the inactivation of PKM2 contributing to the pathogenesis of renal fibrosis through HIF1α accumulation, aberrant glycolysis, and upregulation of EMT in HFD-STZ-induced DN mice. Finally, PKM2 activity and renal fibrosis in DN mice can be reduced by acrolein scavengers such as hydralazine and carnosine. These results imply that acrolein-modified PKM2 contributes to renal fibrosis in the pathogenesis of DN.
Collapse
Affiliation(s)
- Chin-Wei Kuo
- Institute of Pharmacology, College of Medicine, National Yang Ming Chiao Tung University, Taipei, Taiwan
| | - Dong-Hao Chen
- Molecular Medicine Program, National Yang Ming Chiao Tung University, Taipei, Taiwan
| | - Ming-Tsun Tsai
- Division of Nephrology, Department of Medicine, Taipei Veterans General Hospital, Taipei, Taiwan
- School of Medicine, College of Medicine, National Yang Ming Chiao Tung University, Taipei, Taiwan
| | - Chih-Ching Lin
- Division of Nephrology, Department of Medicine, Taipei Veterans General Hospital, Taipei, Taiwan
- School of Medicine, College of Medicine, National Yang Ming Chiao Tung University, Taipei, Taiwan
| | - Hsiao-Wei Cheng
- Institute of Pharmacology, College of Medicine, National Yang Ming Chiao Tung University, Taipei, Taiwan
| | - Yeou-Guang Tsay
- Institute of Biochemistry and Molecular Biology, College of Life Science, National Yang Ming Chiao Tung University, Taipei, Taiwan
| | - Hsiang-Tsui Wang
- Institute of Pharmacology, College of Medicine, National Yang Ming Chiao Tung University, Taipei, Taiwan
- Institute of Food Safety and Health Risk Assessment, National Yang Ming Chiao Tung University, Taipei, Taiwan
- Doctor Degree Program in Toxicology, Kaohsiung Medical University, Kaohsiung, Taiwan
- *Correspondence: Hsiang-Tsui Wang,
| |
Collapse
|
122
|
Fang Y, Zhao T, Ni H, Li Y, Zhu Y, Gao R, Zhang L, Jia Z, Chen G. USP11 exacerbates neuronal apoptosis after traumatic brain injury via PKM2-mediated PI3K/AKT signaling pathway. Brain Res 2023; 1807:148321. [PMID: 36898475 DOI: 10.1016/j.brainres.2023.148321] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2023] [Revised: 03/05/2023] [Accepted: 03/06/2023] [Indexed: 03/12/2023]
Abstract
Ubiquitin-specific protease 11 (USP11) is a ubiquitin-specific protease involved in the regulation of protein ubiquitination. However, its role in traumatic brain injury (TBI) remains unclear. This experiment suggests that USP11 is possibly involved in regulating neuronal apoptosis in TBI. Therefore, we use precision impactor device to established a TBI rat model and assayed the role of USP11 by overexpressing and inhibiting USP11. We found that Usp11 expression increased after TBI. In addition, we hypothesized that pyruvate kinase M2 (PKM2) is a potential USP11 target and experimentally confirmed that upregulation of Usp11 increased Pkm2 expression. Furthermore, elevated USP11 levels exacerbate blood-brain barrier damage, brain edema, and neurobehavioral impairment and cause apoptosis induction through Pkm2 upregulation. Moreover, we hypothesize that PKM2-induced neuronal apoptosis is mediated by the phosphoinositide 3-kinase (PI3K)/protein kinase B (AKT) signaling pathway. Our findings were confirmed by changes in Pi3k and Akt expression with Usp11 upregulation and downregulation and PKM2 inhibition. In conclusion, our findings show that USP11 exacerbates injury in TBI through PKM2 and causes neurological impairment and neuronal apoptosis through the PI3K/AKT signaling pathway.
Collapse
Affiliation(s)
- Yiling Fang
- Department of General Practice, The Affiliated Zhangjiagang Hospital of Soochow University, Suzhou, China
| | - Tianheng Zhao
- Department of Neurosurgery and Brain and Nerve Research Laboratory, The First Affiliated Hospital of Soochow University, Suzhou, China
| | - Haibo Ni
- Department of Neurosurgery, The Affiliated Zhangjiagang Hospital of Soochow University, Suzhou, China
| | - Yajun Li
- Department of Orthopedics, Zhangjiagang TCM Hospital Affiliated to Nanjing University of Chinese Medicine, Suzhou, China
| | - Yongkui Zhu
- Department of Intensive Care Unit, The Affiliated Zhangjiagang Hospital of Soochow University, Suzhou, China
| | - Rong Gao
- Department of Neurosurgery, The Affiliated Zhangjiagang Hospital of Soochow University, Suzhou, China
| | - Li Zhang
- Department of Neurosurgery, The Affiliated Zhangjiagang Hospital of Soochow University, Suzhou, China.
| | - Zhenyu Jia
- Department of General Practice, The Affiliated Zhangjiagang Hospital of Soochow University, Suzhou, China.
| | - Gang Chen
- Department of Neurosurgery and Brain and Nerve Research Laboratory, The First Affiliated Hospital of Soochow University, Suzhou, China
| |
Collapse
|
123
|
Wu H, Jiao Y, Zhou C, Guo X, Wu Z, Lv Q. miR-140-3p/usp36 axis mediates ubiquitination to regulate PKM2 and suppressed the malignant biological behavior of breast cancer through Warburg effect. Cell Cycle 2023; 22:680-692. [PMID: 36305548 PMCID: PMC9980702 DOI: 10.1080/15384101.2022.2139554] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2022] [Revised: 10/10/2022] [Accepted: 10/19/2022] [Indexed: 11/03/2022] Open
Abstract
Breast cancer is a phenomenon in which breast epithelial cells proliferate out of control under the action of various carcinogenic factors. However, the role of USP36 in breast cancer is unknown. We analyzed the expression of USP36 in breast cancer and its association with poor prognosis in breast cancer patients. The effect of USP36 on malignant biological behavior of breast cancer was verified by cell functional experiments. The upstream regulatory mechanism of USP36 was analyzed by Western blot and quantitative RT-qPCR. The influence of USP36 on the Warburg effect of breast cancer was analyzed by detecting the metabolism of cellular energy substances. We found that USP36 is highly expressed in breast tumor tissues and breast cancer cell lines. High expression of USP36 predicts poor prognosis in patients with breast cancer. Effectively reducing the expression of USP36 can significantly inhibit the proliferation, invasion and migration of breast cancer cells, and promote the apoptosis of breast cancer cells. Meanwhile, inhibiting the expression of USP36 can significantly inhibit the production of ATP, lactate, pyruvate and glucose uptake in breast cancer cells. miR-140-3p is an upstream regulator of USP36, which can partially reverse the regulatory effect of USP36 on breast cancer cells. Importantly, USP36 regulates the expression of PKM2 through ubiquitination, which plays a role in regulating the Warburg effect. We confirmed that miR-140-3p regulates the expression of USP36, which mediates ubiquitination and regulates the expression of PKM2, and regulates the malignant biological behavior of breast cancer through the energy metabolism process.
Collapse
Affiliation(s)
- Hao Wu
- Department of Breast Surgery, West China Hospital, Sichuan University, Chengdu, China
- Laboratory of Pathology, West China Hospital, Sichuan University, Chengdu, China
| | - Yile Jiao
- Department of Breast Surgery, West China Hospital, Sichuan University, Chengdu, China
| | - Chen Zhou
- Department of Breast Surgery, West China Hospital, Sichuan University, Chengdu, China
| | - Xinyi Guo
- Department of Breast Surgery, West China Hospital, Sichuan University, Chengdu, China
| | - Zhenru Wu
- Laboratory of Pathology, West China Hospital, Sichuan University, Chengdu, China
| | - Qing Lv
- Department of Breast Surgery, West China Hospital, Sichuan University, Chengdu, China
| |
Collapse
|
124
|
Borbor M, Yin D, Brockmeier U, Wang C, Doeckel M, Pillath-Eilers M, Kaltwasser B, Hermann DM, Dzyubenko E. Neurotoxicity of ischemic astrocytes involves STAT3-mediated metabolic switching and depends on glycogen usage. Glia 2023; 71:1553-1569. [PMID: 36810803 DOI: 10.1002/glia.24357] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2022] [Revised: 02/08/2023] [Accepted: 02/10/2023] [Indexed: 02/24/2023]
Abstract
Astrocytic responses are critical for the maintenance of neuronal networks in health and disease. In stroke, reactive astrocytes undergo functional changes potentially contributing to secondary neurodegeneration, but the mechanisms of astrocyte-mediated neurotoxicity remain elusive. Here, we investigated metabolic reprogramming in astrocytes following ischemia-reperfusion in vitro, explored their role in synaptic degeneration, and verified the key findings in a mouse model of stroke. Using indirect cocultures of primary mouse astrocytes and neurons, we demonstrate that transcription factor STAT3 controls metabolic switching in ischemic astrocytes promoting lactate-directed glycolysis and hindering mitochondrial function. Upregulation of astrocytic STAT3 signaling associated with nuclear translocation of pyruvate kinase isoform M2 and hypoxia response element activation. Reprogrammed thereby, the ischemic astrocytes induced mitochondrial respiration failure in neurons and triggered glutamatergic synapse loss, which was prevented by inhibiting astrocytic STAT3 signaling with Stattic. The rescuing effect of Stattic relied on the ability of astrocytes to utilize glycogen bodies as an alternative metabolic source supporting mitochondrial function. After focal cerebral ischemia in mice, astrocytic STAT3 activation was associated with secondary synaptic degeneration in the perilesional cortex. Inflammatory preconditioning with LPS increased astrocytic glycogen content, reduced synaptic degeneration, and promoted neuroprotection post stroke. Our data indicate the central role of STAT3 signaling and glycogen usage in reactive astrogliosis and suggest novel targets for restorative stroke therapy.
Collapse
Affiliation(s)
- Mina Borbor
- Department of Neurology and Center for Translational Neuro- and Behavioral Sciences (C-TNBS), University Hospital Essen, Essen, Germany
| | - Dongpei Yin
- Department of Neurology and Center for Translational Neuro- and Behavioral Sciences (C-TNBS), University Hospital Essen, Essen, Germany
| | - Ulf Brockmeier
- Department of Neurology and Center for Translational Neuro- and Behavioral Sciences (C-TNBS), University Hospital Essen, Essen, Germany
| | - Chen Wang
- Department of Neurology and Center for Translational Neuro- and Behavioral Sciences (C-TNBS), University Hospital Essen, Essen, Germany
| | - Marius Doeckel
- Department of Neurology and Center for Translational Neuro- and Behavioral Sciences (C-TNBS), University Hospital Essen, Essen, Germany
| | - Matthias Pillath-Eilers
- Department of Neurology and Center for Translational Neuro- and Behavioral Sciences (C-TNBS), University Hospital Essen, Essen, Germany
| | - Britta Kaltwasser
- Department of Neurology and Center for Translational Neuro- and Behavioral Sciences (C-TNBS), University Hospital Essen, Essen, Germany
| | - Dirk M Hermann
- Department of Neurology and Center for Translational Neuro- and Behavioral Sciences (C-TNBS), University Hospital Essen, Essen, Germany
| | - Egor Dzyubenko
- Department of Neurology and Center for Translational Neuro- and Behavioral Sciences (C-TNBS), University Hospital Essen, Essen, Germany
| |
Collapse
|
125
|
Li X, Ma TK, Wang M, Zhang XD, Liu TY, Liu Y, Huang ZH, Zhu YH, Zhang S, Yin L, Xu YY, Ding H, Liu C, Shi H, Fan QL. YY1-induced upregulation of LncRNA-ARAP1-AS2 and ARAP1 promotes diabetic kidney fibrosis via aberrant glycolysis associated with EGFR/PKM2/HIF-1α pathway. Front Pharmacol 2023; 14:1069348. [PMID: 36874012 PMCID: PMC9974832 DOI: 10.3389/fphar.2023.1069348] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2022] [Accepted: 02/06/2023] [Indexed: 02/17/2023] Open
Abstract
Objectives: Dimeric pyruvate kinase (PK) M2 (PKM2) plays an important role in promoting the accumulation of hypoxia-inducible factor (HIF)-1α, mediating aberrant glycolysis and inducing fibrosis in diabetic kidney disease (DKD). The aim of this work was to dissect a novel regulatory mechanism of Yin and Yang 1 (YY1) on lncRNA-ARAP1-AS2/ARAP1 to regulate EGFR/PKM2/HIF-1α pathway and glycolysis in DKD. Materials and methods: We used adeno-associated virus (AAV)-ARAP1 shRNA to knocked down ARAP1 in diabetic mice and overexpressed or knocked down YY1, ARAP1-AS2 and ARAP1 expression in human glomerular mesangial cells. Gene levels were assessed by Western blotting, RT-qPCR, immunofluorescence staining and immunohistochemistry. Molecular interactions were determined by RNA pull-down, co-immunoprecipitation, ubiquitination assay and dual-luciferase reporter analysis. Results: YY1, ARAP1-AS2, ARAP1, HIF-1α, glycolysis and fibrosis genes expressions were upregulated and ARAP1 knockdown could inhibit dimeric PKM2 expression and partly restore tetrameric PKM2 formation, while downregulate HIF-1α accumulation and aberrant glycolysis and fibrosis in in-vivo and in-vitro DKD models. ARAP1 knockdown attenuates renal injury and renal dysfunction in diabetic mice. ARAP1 maintains EGFR overactivation in-vivo and in-vitro DKD models. Mechanistically, YY1 transcriptionally upregulates ARAP1-AS2 and indirectly regulates ARAP1 and subsequently promotes EGFR activation, HIF-1α accumulation and aberrant glycolysis and fibrosis. Conclusion: Our results first highlight the role of the novel regulatory mechanism of YY1 on ARAP1-AS2 and ARAP1 in promoting aberrant glycolysis and fibrosis by EGFR/PKM2/HIF-1α pathway in DKD and provide potential therapeutic strategies for DKD treatments.
Collapse
Affiliation(s)
- Xin Li
- Department of Nephrology, First Hospital of China Medical University, Shenyang, China
- Department of Nephrology, Fourth Hospital of China Medical University, Shenyang, China
| | - Tian-Kui Ma
- Department of Nephrology, First Hospital of China Medical University, Shenyang, China
| | - Min Wang
- Department of Nephrology, First Hospital of China Medical University, Shenyang, China
| | - Xiao-Dan Zhang
- Department of Nephrology, First Hospital of China Medical University, Shenyang, China
| | - Tian-Yan Liu
- Department of Nephrology, First Hospital of China Medical University, Shenyang, China
| | - Yue Liu
- Department of Nephrology, First Hospital of China Medical University, Shenyang, China
| | - Zhao-Hui Huang
- Department of Nephrology, First Hospital of China Medical University, Shenyang, China
| | - Yong-Hong Zhu
- Department of Nephrology, First Hospital of China Medical University, Shenyang, China
| | - Shuang Zhang
- Department of Nephrology, Fourth Hospital of China Medical University, Shenyang, China
| | - Li Yin
- Department of Nephrology, Fourth Hospital of China Medical University, Shenyang, China
| | - Yan-Yan Xu
- Department of Nephrology, Fourth Hospital of China Medical University, Shenyang, China
| | - Hong Ding
- Department of Nephrology, Fourth Hospital of China Medical University, Shenyang, China
| | - Cong Liu
- Department of General Surgery, First Hospital of Harbin Medical University, Harbin, China
| | - Hang Shi
- Department of Intensive Care Unit, Sun Yat-sen Memorial Hospital of Sun Yat-sen University, Guangzhou, China
| | - Qiu-Ling Fan
- Department of Nephrology, First Hospital of China Medical University, Shenyang, China
- Department of Nephrology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| |
Collapse
|
126
|
Li W, Chen W, Peng H, Xiao Z, Liu J, Zeng Y, Huang T, Song Q, Wang X, Xiao Y. Shikonin improves pulmonary vascular remodeling in monocrotaline‑induced pulmonary arterial hypertension via regulation of PKM2. Mol Med Rep 2023; 27:60. [PMID: 36734266 PMCID: PMC9936259 DOI: 10.3892/mmr.2023.12947] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2022] [Accepted: 01/10/2023] [Indexed: 02/04/2023] Open
Abstract
Pulmonary arterial hypertension (PAH), a fatal disease with an insidious onset and rapid progression, shows characteristics such as increases in pulmonary circulatory resistance and pulmonary arterial pressure, and progressive right heart failure. Shikonin can reduce right ventricular systolic pressure in chronically hypoxic mice. However, the mechanisms underlying the protective effect of shikonin against PAH pathogenesis have only been sporadically identified. The present study evaluated whether inhibiting the expression of pyruvate kinase M2 (PKM2) contributed to the improvement of pulmonary vascular remodeling in PAH rats induced by monocrotaline (MCT) treatment. Hemodynamic parameters were assessed using echocardiography and right ventricular catheterization. Right ventricular hypertrophy index analysis and hematoxylin and eosin staining were used to evaluate the degree of pulmonary vascular and right heart remodeling. Moreover, PKM2, p‑PKM2, ERK, p‑ERK, glucose transporter 1 (GLUT1), lactate dehydrogenase A (LDHA) protein expression levels were semi‑quantified using western blotting. The expression and distribution of PKM2 were assessed using immunofluorescence microscopy. The present study demonstrated that MCT treatment caused pulmonary arterial hypertension and pulmonary vascular remodeling in experimental rats. Shikonin improved hemodynamics and pulmonary vascular remodeling in MCT‑induced PAH rats, decreased aerobic glycolysis and downregulated PKM2, p‑PKM2, p‑ERK, GLUT 1 and LDHA protein expression levels. Shikonin improved experimental pulmonary arterial hypertension hemodynamics and pulmonary vascular remodeling at least partly through the inhibition of PKM2 and the resultant suppression of aerobic glycolysis. These results provide a novel understanding of possible new treatment targets for PAH.
Collapse
Affiliation(s)
- Wenfeng Li
- Department of Ultrasound, Hunan Children's Hospital, Changsha, Hunan 410007, P.R. China
| | - Wenjuan Chen
- Department of Ultrasound, Hunan Children's Hospital, Changsha, Hunan 410007, P.R. China
| | - Hongyan Peng
- Hunan Children's Research Institute, Hunan Children's Hospital, Changsha, Hunan 410007, P.R. China
| | - Zhenghui Xiao
- Intensive Care Unit, Hunan Children's Hospital, Changsha, Hunan 410007, P.R. China
| | - Jinqiao Liu
- Department of Ultrasound, Hunan Children's Hospital, Changsha, Hunan 410007, P.R. China
| | - Yunhong Zeng
- Department of Cardiology, Hunan Children's Hospital, Changsha, Hunan 410007, P.R. China
| | - Ting Huang
- Department of Ultrasound, Hunan Children's Hospital, Changsha, Hunan 410007, P.R. China
| | - Qingqing Song
- Department of Cardiology, Hunan Children's Hospital, Changsha, Hunan 410007, P.R. China
| | - Xun Wang
- Department of Cardiology, Hunan Children's Hospital, Changsha, Hunan 410007, P.R. China
| | - Yunbin Xiao
- Department of Cardiology, Hunan Children's Hospital, Changsha, Hunan 410007, P.R. China,Correspondence to: Dr Yunbin Xiao, Department of Cardiology, Hunan Children's Hospital, 86 Zi Yuan Road, Yuhua, Changsha, Hunan 410007, P.R. China, E-mail:
| |
Collapse
|
127
|
Zhang Q, Zhang J, Yao A, Tian X, Han Z, Yuan Y, Tao K, Yang X. OTUB2 promotes the progression of endometrial cancer by regulating the PKM2-mediated PI3K/AKT signaling pathway. Cell Biol Int 2023; 47:428-438. [PMID: 36316812 DOI: 10.1002/cbin.11950] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2022] [Revised: 09/27/2022] [Accepted: 10/01/2022] [Indexed: 11/07/2022]
Abstract
Endometrial carcinoma (EC) morbidity and mortality have been increasing in recent years. Otubain 2 (OTUB2) was shown to be upregulated in EC patients, so the aim of this study was to explore the role of OTUB2 in EC. Cell Counting Kit-8 (CCK-8), colony formation, enzyme-linked immunosorbent assay, the wound healing assay, and Transwell invasion assays were used to investigate the specific role of OTUB2 in EC tumorigenesis. Real-time polymerase chain reaction and western blot analysis were used to detect the expression of OTUB2 in EC tissues and cells. OTUB2 is upregulated in EC patients and cell lines and is associated with a poor prognosis. The overexpression of OTUB2 promoted glycolysis and induced the proliferation, migration, and invasion of endometrial cancer cells. The silencing of OTUB2 had the opposite effect. In addition, the silencing of OTUB2 significantly suppressed the expression levels of PKM2. Importantly, inhibition of the PKM2/PI3K/AKT signaling pathway significantly reversed the promoting effect of OTUB2 overexpression on EC. OTUB2 regulated the proliferation and invasion of EC cells by regulating the PKM2/PI3K/AKT signaling pathway. OTUB2 may serve as a potential prognostic and therapeutic target in EC.
Collapse
Affiliation(s)
- Qian Zhang
- Department of The First of Internal Medicine, Shaanxi Provincial Cancer Hospital, Xi'an, Shannxi, People's Republic of China
| | - Jing Zhang
- Department of The Fourth of Gynecologic Oncology, Shaanxi Provincial Cancer Hospital, Xi'an, Shannxi, People's Republic of China
| | - Anmei Yao
- Department of The Second of Gynecologic Oncology, Shaanxi Provincial Cancer Hospital, Xi'an, Shannxi, People's Republic of China
| | - Xiaofei Tian
- Department of The Second of Gynecologic Oncology, Shaanxi Provincial Cancer Hospital, Xi'an, Shannxi, People's Republic of China
| | - Zhihong Han
- Department of The Second of Gynecologic Oncology, Shaanxi Provincial Cancer Hospital, Xi'an, Shannxi, People's Republic of China
| | - Yuan Yuan
- Department of The Second of Gynecologic Oncology, Shaanxi Provincial Cancer Hospital, Xi'an, Shannxi, People's Republic of China
| | - Kai Tao
- Department of The Second of Gynecologic Oncology, Shaanxi Provincial Cancer Hospital, Xi'an, Shannxi, People's Republic of China
| | - Xuemei Yang
- Department of The Second of Gynecologic Oncology, Shaanxi Provincial Cancer Hospital, Xi'an, Shannxi, People's Republic of China
| |
Collapse
|
128
|
Ahmed D, Al-Daraawi M, Cassol E. Innate sensing and cellular metabolism: role in fine tuning antiviral immune responses. J Leukoc Biol 2023; 113:164-190. [PMID: 36822175 DOI: 10.1093/jleuko/qiac011] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2022] [Indexed: 01/19/2023] Open
Abstract
Several studies over the last decade have identified intimate links between cellular metabolism and macrophage function. Metabolism has been shown to both drive and regulate macrophage function by producing bioenergetic and biosynthetic precursors as well as metabolites (and other bioactive molecules) that regulate gene expression and signal transduction. Many studies have focused on lipopolysaccharide-induced reprogramming, assuming that it is representative of most inflammatory responses. However, emerging evidence suggests that diverse pathogen-associated molecular patterns (PAMPs) are associated with unique metabolic profiles, which may drive pathogen specific immune responses. Further, these metabolic pathways and processes may act as a rheostat to regulate the magnitude of an inflammatory response based on the biochemical features of the local microenvironment. In this review, we will discuss recent work examining the relationship between cellular metabolism and macrophage responses to viral PAMPs and describe how these processes differ from lipopolysaccharide-associated responses. We will also discuss how an improved understanding of the specificity of these processes may offer new insights to fine-tune macrophage function during viral infections or when using viral PAMPs as therapeutics.
Collapse
Affiliation(s)
- Duale Ahmed
- Department of Health Sciences, Carleton University, Ottawa, Ontario, Canada.,Department of Biology, Carleton University, Ottawa, Ontario, Canada
| | - Malak Al-Daraawi
- Department of Health Sciences, Carleton University, Ottawa, Ontario, Canada
| | - Edana Cassol
- Department of Health Sciences, Carleton University, Ottawa, Ontario, Canada.,Centre for Infection, Immunity and Inflammation, University of Ottawa, Ottawa, Ontario, Canada
| |
Collapse
|
129
|
Park YS, Han JH, Park JH, Choi JS, Kim SH, Kim HS. Pyruvate Kinase M2: A New Biomarker for the Early Detection of Diabetes-Induced Nephropathy. Int J Mol Sci 2023; 24:ijms24032683. [PMID: 36769016 PMCID: PMC9916947 DOI: 10.3390/ijms24032683] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2022] [Revised: 01/25/2023] [Accepted: 01/28/2023] [Indexed: 02/04/2023] Open
Abstract
Diabetic nephropathy (DN) is a common complication of diabetes. DN progresses to end-stage renal disease, which has a high mortality rate. Current research is focused on identifying non-invasive potential biomarkers in the early stage of DN. We previously indicated that pyruvate kinase M2 (PKM2) is excreted in the urine of rats after cisplatin-induced acute kidney injury (AKI). However, it has not been reported whether PKM2 can be used as a biomarker to diagnose DN. Therefore, we try to compare whether the protein PKM2 can be detected in the urine samples from diabetic patients as shown in the results of DN models. In this study, high-fat diet (HFD)-induced Zucker diabetic fatty (ZDF) rats were used for DN phenotyping. After 19 weeks of receiving a HFD, the DN model's blood glucose, blood urea nitrogen, and serum creatinine levels were significantly increased; severe tubular and glomerular damages were also noted. The following protein-based biomarkers were increased in the urine of these models: kidney injury molecule-1 (KIM-1), neutrophil gelatinase-associated lipocalin (NGAL), and PKM2. PKM2 had the earliest detection rate. In the urine samples of patients, PKM2 protein was highly detected in the urine of diabetic patients but was not excreted in the urine of normal subjects. Therefore, PKM2 was selected as the new biomarker for the early diagnosis of DN. Our results reflect current knowledge on the role of PKM2 in DN.
Collapse
Affiliation(s)
- Yeon Su Park
- School of Pharmacy, Sungkyunkwan University, Suwon 16419, Republic of Korea
| | - Joo Hee Han
- School of Pharmacy, Sungkyunkwan University, Suwon 16419, Republic of Korea
| | - Jae Hyeon Park
- School of Pharmacy, Sungkyunkwan University, Suwon 16419, Republic of Korea
| | - Ji Soo Choi
- School of Pharmacy, Sungkyunkwan University, Suwon 16419, Republic of Korea
| | - Seung Hyeon Kim
- St. Mark’s School, 25 Marlboro Rd, Southborough, MA 01772, USA
| | - Hyung Sik Kim
- School of Pharmacy, Sungkyunkwan University, Suwon 16419, Republic of Korea
- Correspondence: ; Tel.:+82-31-290-7789
| |
Collapse
|
130
|
Epigenetics and Metabolism Reprogramming Interplay into Glioblastoma: Novel Insights on Immunosuppressive Mechanisms. Antioxidants (Basel) 2023; 12:antiox12020220. [PMID: 36829778 PMCID: PMC9952003 DOI: 10.3390/antiox12020220] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2022] [Revised: 01/09/2023] [Accepted: 01/16/2023] [Indexed: 01/19/2023] Open
Abstract
The central nervous system represents a complex environment in which glioblastoma adapts skillfully, unleashing a series of mechanisms suitable for its efficient development and diffusion. In particular, changes in gene expression and mutational events that fall within the domain of epigenetics interact complexly with metabolic reprogramming and stress responses enacted in the tumor microenvironment, which in turn fuel genomic instability by providing substrates for DNA modifications. The aim of this review is to analyze this complex interaction that consolidates several conditions that confer a state of immunosuppression and immunoevasion, making glioblastoma capable of escaping attack and elimination by immune cells and therefore invincible against current therapies. The progressive knowledge of the cellular mechanisms that underlie the resistance of the glioblastoma represents, in fact, the only weapon to unmask its weak points to be exploited to plan successful therapeutic strategies.
Collapse
|
131
|
Du X, Wei H, Zhang B, Wang B, Li Z, Pang LK, Zhao R, Yao W. Molecular mechanisms of osteosarcoma metastasis and possible treatment opportunities. Front Oncol 2023; 13:1117867. [PMID: 37197432 PMCID: PMC10183593 DOI: 10.3389/fonc.2023.1117867] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2022] [Accepted: 04/18/2023] [Indexed: 05/19/2023] Open
Abstract
In osteosarcoma patients, metastasis of the primary cancer is the leading cause of death. At present, management options to prevent metastasis are limited and non-curative. In this study, we review the current state of knowledge on the molecular mechanisms of metastasis and discuss promising new therapies to combat osteosarcoma metastasis. Genomic and epigenomic changes, metabolic reprogramming, transcription factors, dysregulation of physiologic pathways, and alterations to the tumor microenvironment are some of the changes reportedly involved in the regulation of osteosarcoma metastasis. Key factors within the tumor microenvironment include infiltrating lymphocytes, macrophages, cancer-associated fibroblasts, platelets, and extracellular components such as vesicles, proteins, and other secreted molecules. We conclude by discussing potential osteosarcoma-limiting agents and their clinical studies.
Collapse
Affiliation(s)
- Xinhui Du
- Bone Soft Tissue Department, The Affiliated Cancer Hospital of Zhengzhou University and Henan Cancer Hospital, Zhengzhou, China
- Key Laboratory for Digital Assessment of Spinal-Pelvic Tumor and Surgical Aid Tools Design (Zhengzhou), Zhengzhou, Henan, China
- Key Laboratory for Perioperative Digital Assessment of Bone Tumors (Henan), Zhengzhou, Henan, China
- *Correspondence: Xinhui Du,
| | - Hua Wei
- Department of Anesthesiology, Pain and Perioperative Medicine, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Boya Zhang
- Bone Soft Tissue Department, The Affiliated Cancer Hospital of Zhengzhou University and Henan Cancer Hospital, Zhengzhou, China
- Key Laboratory for Digital Assessment of Spinal-Pelvic Tumor and Surgical Aid Tools Design (Zhengzhou), Zhengzhou, Henan, China
- Key Laboratory for Perioperative Digital Assessment of Bone Tumors (Henan), Zhengzhou, Henan, China
| | - Bangmin Wang
- Bone Soft Tissue Department, The Affiliated Cancer Hospital of Zhengzhou University and Henan Cancer Hospital, Zhengzhou, China
- Key Laboratory for Digital Assessment of Spinal-Pelvic Tumor and Surgical Aid Tools Design (Zhengzhou), Zhengzhou, Henan, China
- Key Laboratory for Perioperative Digital Assessment of Bone Tumors (Henan), Zhengzhou, Henan, China
| | - Zhehuang Li
- Bone Soft Tissue Department, The Affiliated Cancer Hospital of Zhengzhou University and Henan Cancer Hospital, Zhengzhou, China
- Key Laboratory for Digital Assessment of Spinal-Pelvic Tumor and Surgical Aid Tools Design (Zhengzhou), Zhengzhou, Henan, China
- Key Laboratory for Perioperative Digital Assessment of Bone Tumors (Henan), Zhengzhou, Henan, China
| | - Lon Kai Pang
- Baylor College of Medicine, Houston, TX, United States
| | - Ruiying Zhao
- Department of Integrative Biology and Pharmacology, McGovern Medical School, The University of Texas Health Science Center at Houston, Houston, TX, United States
| | - Weitao Yao
- Bone Soft Tissue Department, The Affiliated Cancer Hospital of Zhengzhou University and Henan Cancer Hospital, Zhengzhou, China
- Key Laboratory for Digital Assessment of Spinal-Pelvic Tumor and Surgical Aid Tools Design (Zhengzhou), Zhengzhou, Henan, China
- Key Laboratory for Perioperative Digital Assessment of Bone Tumors (Henan), Zhengzhou, Henan, China
| |
Collapse
|
132
|
Kapoor S, Chatterjee DR, Chowdhury MG, Das R, Shard A. Roadmap to Pyruvate Kinase M2 Modulation - A Computational Chronicle. Curr Drug Targets 2023; 24:464-483. [PMID: 36998144 DOI: 10.2174/1389450124666230330103126] [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: 10/01/2022] [Revised: 01/14/2023] [Accepted: 02/10/2023] [Indexed: 04/01/2023]
Abstract
Pyruvate kinase M2 (PKM2) has surfaced as a potential target for anti-cancer therapy. PKM2 is known to be overexpressed in the tumor cells and is a critical metabolic conduit in supplying the augmented bioenergetic demands of the recalcitrant cancer cells. The presence of PKM2 in structurally diverse tetrameric as well as dimeric forms has opened new avenues to design novel modulators. It is also a truism to state that drug discovery has advanced significantly from various computational techniques like molecular docking, virtual screening, molecular dynamics, and pharmacophore mapping. The present review focuses on the role of computational tools in exploring novel modulators of PKM2. The structural features of various isoforms of PKM2 have been discussed along with reported modulators. An extensive analysis of the structure-based and ligand- based in silico methods aimed at PKM2 modulation has been conducted with an in-depth review of the literature. The role of advanced tools like QSAR and quantum mechanics has been established with a brief discussion of future perspectives.
Collapse
Affiliation(s)
- Saumya Kapoor
- Department of Medicinal Chemistry, National Institute of Pharmaceutical Education and Research, Ahmedabad, Opposite Air force Station Palaj, Gandhinagar-382355, Gujarat, India
| | - Deep Rohan Chatterjee
- Department of Medicinal Chemistry, National Institute of Pharmaceutical Education and Research, Ahmedabad, Opposite Air force Station Palaj, Gandhinagar-382355, Gujarat, India
| | - Moumita Ghosh Chowdhury
- Department of Medicinal Chemistry, National Institute of Pharmaceutical Education and Research, Ahmedabad, Opposite Air force Station Palaj, Gandhinagar-382355, Gujarat, India
| | - Rudradip Das
- Department of Medicinal Chemistry, National Institute of Pharmaceutical Education and Research, Ahmedabad, Opposite Air force Station Palaj, Gandhinagar-382355, Gujarat, India
| | - Amit Shard
- Department of Medicinal Chemistry, National Institute of Pharmaceutical Education and Research, Ahmedabad, Opposite Air force Station Palaj, Gandhinagar-382355, Gujarat, India
| |
Collapse
|
133
|
Shi Y, Li Z, Wang B, Shi X, Ye H, Delafield DG, Lv L, Ye Z, Chen Z, Ma F, Li L. Enabling Global Analysis of Protein Citrullination via Biotin Thiol Tag-Assisted Mass Spectrometry. Anal Chem 2022; 94:17895-17903. [PMID: 36512406 DOI: 10.1021/acs.analchem.2c03844] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Citrullination is a key post-translational modification (PTM) that affects protein structures and functions. Although it has been linked to various biological processes and disease pathogenesis, the underlying mechanism remains poorly understood due to a lack of effective tools to enrich, detect, and localize this PTM. Herein, we report the design and development of a biotin thiol tag that enables derivatization, enrichment, and confident identification of citrullination via mass spectrometry. We perform global mapping of the citrullination proteome of mouse tissues. In total, we identify 691 citrullination sites from 432 proteins which represents the largest data set to date. We discover novel distribution and functions of this PTM. This study depicts a landscape of protein citrullination and lays the foundation for further deciphering their physiological and pathological roles.
Collapse
Affiliation(s)
- Yatao Shi
- School of Pharmacy, University of Wisconsin─Madison, Madison, Wisconsin 53705, United States
| | - Zihui Li
- Department of Chemistry, University of Wisconsin─Madison, Madison, Wisconsin 53706, United States
| | - Bin Wang
- School of Pharmacy, University of Wisconsin─Madison, Madison, Wisconsin 53705, United States
| | - Xudong Shi
- Department of Surgery, School of Medicine and Public Health, University of Wisconsin─Madison, Madison, Wisconsin 53792, United States
| | - Hui Ye
- School of Pharmaceutical Sciences, China Pharmaceutical University, Nanjing, 210009, China
| | - Daniel G Delafield
- Department of Chemistry, University of Wisconsin─Madison, Madison, Wisconsin 53706, United States
| | - Langlang Lv
- School of Pharmaceutical Sciences, China Pharmaceutical University, Nanjing, 210009, China
| | - Zhengqing Ye
- Medicinal Chemistry Center, School of Pharmacy, University of Wisconsin─Madison, Madison, Wisconsin 53705, United States
| | - Zhengwei Chen
- Department of Chemistry, University of Wisconsin─Madison, Madison, Wisconsin 53706, United States
| | - Fengfei Ma
- School of Pharmacy, University of Wisconsin─Madison, Madison, Wisconsin 53705, United States
| | - Lingjun Li
- School of Pharmacy, University of Wisconsin─Madison, Madison, Wisconsin 53705, United States.,Department of Chemistry, University of Wisconsin─Madison, Madison, Wisconsin 53706, United States
| |
Collapse
|
134
|
Yang Y, Qin H, Ding M, Ji C, Chen W, Diao W, Yin H, Chen M, Gan W, Guo H. Small ankyrin 1 (sANK1) promotes docetaxel resistance in castration-resistant prostate cancer cells by enhancing oxidative phosphorylation. FEBS Open Bio 2022; 13:257-269. [PMID: 36508323 PMCID: PMC9900087 DOI: 10.1002/2211-5463.13535] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2022] [Revised: 11/09/2022] [Accepted: 12/11/2022] [Indexed: 12/14/2022] Open
Abstract
Docetaxel (DTX) plays an important role in treating advanced prostate cancer (PCa). However, nearly all patients receiving DTX therapy ultimately progress to DTX resistance. How to address DTX resistance in PCa remains a key challenge for all urologists. Small ankyrin 1 (sAnk1) is an integral membrane protein in the endoplasmic reticulum. In the present study, we identified that sAnk1 is upregulated in PCa tissues and is positively associated with DTX therapy resistance in PCa. Further investigation demonstrated that overexpression of sAnk1 can significantly increase the DTX-resistant ability of PCa cells in vitro and in vivo. In addition, overexpression of sAnk1 could enhance oxidative phosphorylation (OXPHOS) levels in PCa cells, which was consistent with the higher OXPHOS levels observed in DTX-resistant PCa cells as compared to DTX-sensitive PCa cells. sAnk1 was also found to interact with polypyrimidine-tract-binding protein (PTBP1), an alternative splicing factor, and suppressed PTBP1-mediated alternative splicing of the pyruvate kinase gene (PKM). Thus, overexpression of sAnk1 decreased the ratio of PKM2/PKM1, enhanced the OXPHOS level, and ultimately promoted the resistance of PCa cells to DTX. In summary, our data suggest that sAnk1 enhances DTX resistance in PCa cells.
Collapse
Affiliation(s)
- Yang Yang
- Department of Urology, Drum Tower Hospital, Medical School of Nanjing University, Institute of UrologyNanjing UniversityChina
| | - Haixiang Qin
- Department of Urology, Drum Tower Hospital, Medical School of Nanjing University, Institute of UrologyNanjing UniversityChina
| | - Meng Ding
- Department of Urology, Drum Tower Hospital, Medical School of Nanjing University, Institute of UrologyNanjing UniversityChina
| | - Changwei Ji
- Department of Urology, Drum Tower Hospital, Medical School of Nanjing University, Institute of UrologyNanjing UniversityChina
| | - Wei Chen
- Department of Urology, Drum Tower Hospital, Medical School of Nanjing University, Institute of UrologyNanjing UniversityChina
| | - Wenli Diao
- Department of Urology, Drum Tower Hospital, Medical School of Nanjing University, Institute of UrologyNanjing UniversityChina
| | - Haoli Yin
- Department of Urology, Drum Tower Hospital, Medical School of Nanjing University, Institute of UrologyNanjing UniversityChina
| | - Mengxia Chen
- Department of Urology, Drum Tower Hospital, Medical School of Nanjing University, Institute of UrologyNanjing UniversityChina
| | - Weidong Gan
- Department of Urology, Drum Tower Hospital, Medical School of Nanjing University, Institute of UrologyNanjing UniversityChina
| | - Hongqian Guo
- Department of Urology, Drum Tower Hospital, Medical School of Nanjing University, Institute of UrologyNanjing UniversityChina
| |
Collapse
|
135
|
Abdollahi S, Hasanpour Ardekanizadeh N, Poorhosseini SM, Gholamalizadeh M, Roumi Z, Goodarzi MO, Doaei S. Unraveling the Complex Interactions between the Fat Mass and Obesity-Associated (FTO) Gene, Lifestyle, and Cancer. Adv Nutr 2022; 13:2406-2419. [PMID: 36104156 PMCID: PMC9776650 DOI: 10.1093/advances/nmac101] [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: 04/12/2022] [Revised: 06/28/2022] [Accepted: 09/12/2022] [Indexed: 01/29/2023] Open
Abstract
Carcinogenesis is a complicated process and originates from genetic, epigenetic, and environmental factors. Recent studies have reported a potential critical role for the fat mass and obesity-associated (FTO) gene in carcinogenesis through different signaling pathways such as mRNA N6-methyladenosine (m6A) demethylation. The most common internal modification in mammalian mRNA is the m6A RNA methylation that has significant biological functioning through regulation of cancer-related cellular processes. Some environmental factors, like physical activity and dietary intake, may influence signaling pathways engaged in carcinogenesis, through regulating FTO gene expression. In addition, people with FTO gene polymorphisms may be differently influenced by cancer risk factors, for example, FTO risk allele carriers may need a higher intake of nutrients to prevent cancer than others. In order to obtain a deeper viewpoint of the FTO, lifestyle, and cancer-related pathway interactions, this review aims to discuss upstream and downstream pathways associated with the FTO gene and cancer. The present study discusses the possible mechanisms of interaction of the FTO gene with various cancers and provides a comprehensive picture of the lifestyle factors affecting the FTO gene as well as the possible downstream pathways that lead to the effect of the FTO gene on cancer.
Collapse
Affiliation(s)
- Sepideh Abdollahi
- Department of Medical Genetics, School of Medicine, Tehran University of
Medical Sciences, Tehran, Iran
| | - Naeemeh Hasanpour Ardekanizadeh
- Department of Clinical Nutrition, School of Nutrition and Food Sciences,
Shiraz University of Medical Sciences, Shiraz, Iran
| | | | - Maryam Gholamalizadeh
- Cancer Research Center, Shahid Beheshti University of Medical
Sciences, Tehran, Iran
| | - Zahra Roumi
- Department of Nutrition, Science and Research Branch, Islamic Azad
University, Tehran, Iran
| | - Mark O Goodarzi
- Division of Endocrinology, Diabetes and Metabolism, Department of Medicine,
Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - Saeid Doaei
- Department of Community Nutrition, School of Nutrition and Food Sciences,
Shahid Beheshti University of Medical Sciences, Tehran, Iran
| |
Collapse
|
136
|
Pyruvate Kinase M2 Promotes Hair Regeneration by Connecting Metabolic and Wnt/β-Catenin Signaling. Pharmaceutics 2022; 14:pharmaceutics14122774. [PMID: 36559274 PMCID: PMC9781674 DOI: 10.3390/pharmaceutics14122774] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2022] [Revised: 12/03/2022] [Accepted: 12/08/2022] [Indexed: 12/15/2022] Open
Abstract
Hair follicle stem cells (HFSCs) utilize glycolytic metabolism during their activation and anagen induction. However, the role of pyruvate kinase M2 (PKM2), which catalyzes the final step of glycolysis, in hair regeneration has not been elucidated. In this study, we investigated the expression pattern and activity of PKM2 during the depilation-induced anagen progression in mice. We found that TEPP-46, a selective activator of PKM2, enhanced hair re-growth and proliferation of HFSCs. PKM2 expression was increased via up-regulation of Wnt/β-catenin signaling, which is involved in hair regeneration. Moreover, a combined treatment with KY19382, a small molecule that activates Wnt/β-catenin signaling, and TEPP-46 significantly enhanced hair re-growth and wound-induced hair follicle neogenesis (WIHN). These results indicate that simultaneous activation of the PKM2 and Wnt/β-catenin signaling could be a potential strategy for treating alopecia patients.
Collapse
|
137
|
Magadum A. Modified mRNA Therapeutics for Heart Diseases. Int J Mol Sci 2022; 23:ijms232415514. [PMID: 36555159 PMCID: PMC9779737 DOI: 10.3390/ijms232415514] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2022] [Revised: 11/26/2022] [Accepted: 11/27/2022] [Indexed: 12/13/2022] Open
Abstract
Cardiovascular diseases (CVD) remain a substantial global health problem and the leading cause of death worldwide. Although many conventional small-molecule treatments are available to support the cardiac function of the patient with CVD, they are not effective as a cure. Among potential targets for gene therapy are severe cardiac and peripheral ischemia, heart failure, vein graft failure, and some forms of dyslipidemias. In the last three decades, multiple gene therapy tools have been used for heart diseases caused by proteins, plasmids, adenovirus, and adeno-associated viruses (AAV), but these remain as unmet clinical needs. These gene therapy methods are ineffective due to poor and uncontrolled gene expression, low stability, immunogenicity, and transfection efficiency. The synthetic modified mRNA (modRNA) presents a novel gene therapy approach which provides a transient, stable, safe, non-immunogenic, controlled mRNA delivery to the heart tissue without any risk of genomic integration, and achieves a therapeutic effect in different organs, including the heart. The mRNA translation starts in minutes, and remains stable for 8-10 days (pulse-like kinetics). The pulse-like expression of modRNA in the heart induces cardiac repair, cardiomyocyte proliferation and survival, and inhibits cardiomyocyte apoptosis post-myocardial infarction (MI). Cell-specific (cardiomyocyte) modRNA translation developments established cell-specific modRNA therapeutics for heart diseases. With these laudable characteristics, combined with its expression kinetics in the heart, modRNA has become an attractive therapeutic for the treatment of CVD. This review discusses new developments in modRNA therapy for heart diseases.
Collapse
Affiliation(s)
- Ajit Magadum
- Center for Translational Medicine, Temple University, Philadelphia, PA 19140, USA
| |
Collapse
|
138
|
Qing L, Li Q, Yang Y, Xu W, Dong Z. A prognosis marker MUC1 correlates with metabolism and drug resistance in bladder cancer: a bioinformatics research. BMC Urol 2022; 22:114. [PMID: 35879749 PMCID: PMC9309451 DOI: 10.1186/s12894-022-01067-8] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2022] [Accepted: 07/19/2022] [Indexed: 12/12/2022] Open
Abstract
Abstract
Background
MUC1 is a type I transmembrane protein that plays an important role in tumor cell signal transduction. Although current studies have shown that MUC1 is upregulated in bladder cancer (BC), the specific mechanism is still unclear.
Methods
We performed expression analysis, gene set enrichment analysis, survival analysis, immune infiltration analysis, drug sensitivity analysis, and metabolism-related gene expression analysis on TCGA-BLCA, GES31684 and GSE13507.
Results
The expression of MUC1 in the tumor and lymphatic metastasis positive samples was significantly increased. Genes related to MUC1 expression were significantly enriched in immune response, ribosomes, exosomes, and energy metabolism. The results of the immune infiltration analysis showed that M1 macrophages in BC with high MUC1 expression were significantly decreased. Expression of MUC1 increases drug resistance in BC patients. In addition, MUC1 increases glycolysis, glucose uptake, and lactate production by inducing metabolic reprogramming.
Conclusion
MUC1 has a significant effect on the metabolism and immune cell infiltration of BC, which may be the cause of increased drug resistance, and can be used as a molecular target for the diagnosis and treatment of BC.
Collapse
|
139
|
Quercetin protects against LPS-induced lung injury in mice via SIRT1-mediated suppression of PKM2 nuclear accumulation. Eur J Pharmacol 2022; 936:175352. [PMID: 36309049 DOI: 10.1016/j.ejphar.2022.175352] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2022] [Revised: 10/19/2022] [Accepted: 10/21/2022] [Indexed: 11/24/2022]
|
140
|
Guo Y, Hu H, Xu S, Xia W, Li H. Useful genes for predicting the efficacy of transarterial chemoembolization in hepatocellular carcinoma. J Cancer Res Ther 2022; 18:1860-1866. [PMID: 36647943 DOI: 10.4103/jcrt.jcrt_1479_22] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023]
Abstract
Transarterial chemoembolization (TACE) is generally used to treat patients with hepatocellular carcinoma (HCC), a common and deadly cancer; however, its efficacy varies according to factors such as tumor volume, stage, serum alpha-fetoprotein level, and chosen feeding artery. In addition, gene-related factors have been recently suggested to be involved in the regulation and prediction of TACE outcomes. Accordingly, genes could serve as effective biomarkers to select patients who can benefit from TACE. These gene-related factors can activate signaling pathways affecting cancer cell survival while regulating the epithelial-mesenchymal transition, angiogenesis, and the tumor microenvironment, all directly associated with tumor progression, thereby affecting TACE efficacy. Moreover, this disordered gene expression is associated with poor prognosis in patients with HCC, including TACE resistance, postoperative recurrence, and metastasis. To identify the exact relationship between various genes and TACE efficacy, this review summarizes the involvement of protein-coding and non-coding genes and single nucleotide polymorphisms in TACE efficacy for predicting the efficacy of TACE; the present findings may help improve the efficacy of TACE in clinical settings.
Collapse
Affiliation(s)
- Yuan Guo
- Department of Minimal Invasive Intervention, The Affiliated Cancer Hospital of Zhengzhou University, Henan Cancer Hospital, Zhengzhou, China
| | - Hongtao Hu
- Department of Minimal Invasive Intervention, The Affiliated Cancer Hospital of Zhengzhou University, Henan Cancer Hospital, Zhengzhou, China
| | - Shijun Xu
- Department of Radiology, The Affiliated Cancer Hospital of Zhengzhou University, Henan Cancer Hospital, Zhengzhou, China
| | - Weili Xia
- Department of Minimal Invasive Intervention, The Affiliated Cancer Hospital of Zhengzhou University, Henan Cancer Hospital, Zhengzhou, China
| | - Hailiang Li
- Department of Minimal Invasive Intervention, The Affiliated Cancer Hospital of Zhengzhou University, Henan Cancer Hospital, Zhengzhou, China
| |
Collapse
|
141
|
Yang Y, Ren P, Liu X, Sun X, Zhang C, Du X, Xing B. PPP1R26 drives hepatocellular carcinoma progression by controlling glycolysis and epithelial-mesenchymal transition. J Exp Clin Cancer Res 2022; 41:101. [PMID: 35292107 PMCID: PMC8922775 DOI: 10.1186/s13046-022-02302-8] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2021] [Accepted: 02/25/2022] [Indexed: 01/17/2023] Open
Abstract
Background Hepatocellular carcinoma (HCC) is usually diagnosed at an advanced stage due to rapid progression. Glycolysis supports anabolic growth and metastasis to promote HCC progression. However, the molecular mechanisms linking glycolysis and metastasis in HCC are not completely defined. Methods The expression of PPP1R26 in human HCC tissues was evaluated by immunohistochemistry, and the clinical significance of PPP1R26 in the progression and prognosis of the HCC patients were analyzed. The PPP1R26-binding proteins were determined by mass spectrometry analysis. The function of PPP1R26 in glycolysis, EMT and tumorigenesis were evaluated in HCC cells. Glucose uptake and tumor growth were evaluated using PET imaging in mouse xenografts in vivo. Protein binding was confirmed by co-immunoprecipitation and immunofluorescence co-localization. Protein-RNA binding was determined by RNA-immunoprecipitation (RIP) experiment. The binding of protein on the promoter was evaluated by chromatin immunoprecipitation assay (ChIP). Results PPP1R26 is upregulated in human HCC tissues and its upregulation is significantly associated with metastasis and the poor survival of the patients. PPP1R26 activates glycolysis in HCC cells and in mouse xenografts in vivo. PPP1R26 drives glycolysis by binding to PTBP1 to facilitate the mRNA splicing of PKM2. Simultaneously, overexpressed PPP1R26 induces the nuclear accumulation of PKM2 to inhibit the expression of E-cadherin further to drive EMT. Mechanistically, PPP1R26 binds with Ser37-phosphorylated PKM2 and TGIF2 in the nucleus and blocks the binding of TGIF2 with CDH1 promoter to inhibit the transcription of CDH1. Conclusion PPP1R26 promotes glycolysis by enhancing PKM2 splicing and simultaneously activates EMT by forming a PPP1R26-PKM2-TGIF2 complex to drive HCC progression. Therefore, targeting PPP1R26 attenuates HCC progression and provides a potential therapeutic strategy for the HCC patients with upregulation of PPP1R26. Supplementary Information The online version contains supplementary material available at 10.1186/s13046-022-02302-8.
Collapse
|
142
|
Abstract
Lysine succinylation is a novel, broad-spectrum, dynamic, non-enzymatic protein post-translational modification (PTM). Succinylation is essential for the regulation of protein function and control of various signaling and regulatory pathways. It is involved in several life activities, including glucose metabolism, amino acid metabolism, fatty acid metabolism, ketone body synthesis, and reactive oxygen species clearance, by regulating protease activity and gene expression. The level of succinylation is mainly regulated by succinyl donor, succinyltransferase, and desuccinylase. Many studies have confirmed that succinylation plays a role in tumorigenesis by creating tissue heterogeneity, and can promote or inhibit various cancers via the regulation of different substrate targets or signaling pathways. The mechanism of action of some antineoplastic drugs is related to succinylation. To better understand the role of succinylation modification in cancer development and treatment, the present study reviewed the current research content and latest progress of succinylation modification in cancer, which might provide a new direction and target for the prevention and treatment of cancer.
Collapse
Affiliation(s)
- Keer Lu
- Department of Prescription Science, Heilongjiang University of Chinese Medicine, Harbin, Heilongjiang, China
| | - Dongwei Han
- Department of Prescription Science, Heilongjiang University of Chinese Medicine, Harbin, Heilongjiang, China
- *Correspondence: Dongwei Han, Department of Prescription Science, Heilongjiang University of Chinese Medicine, No. 24 Heping Road, Harbin, Heilongjiang 150040, China (e-mail: )
| |
Collapse
|
143
|
Zhang Z, Zheng Y, Chen Y, Yin Y, Chen Y, Chen Q, Hou Y, Shen S, Lv M, Wang T. Gut fungi enhances immunosuppressive function of myeloid-derived suppressor cells by activating PKM2-dependent glycolysis to promote colorectal tumorigenesis. Exp Hematol Oncol 2022; 11:88. [DOI: 10.1186/s40164-022-00334-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2022] [Accepted: 10/07/2022] [Indexed: 11/09/2022] Open
Abstract
Abstract
Background
Accumulating evidence implicates that gut fungi are associated with the pathogenesis of colorectal cancer (CRC). Our previous study has revealed that Candida tropicalis (C. tropicalis) promotes colorectal tumorigenesis by enhancing immunosuppressive function of myeloid-derived suppressor cells (MDSCs) and increasing accumulation of MDSCs, but the underlying mechanisms remain unestablished.
Methods
Bone marrow–derived MDSCs were stimulated with C. tropicalis. RNA-sequencing analysis was performed to screen the differentially expressed genes. Quantitative real-time PCR and western blot were used to measure the expression of related proteins. Co-culture assay of MDSCs and CD8+ T cells was used to determine the immunosuppressive ability of MDSCs. Metabolomic analysis was conducted to detect metabolic reprogramming of MDSCs. Aerobic glycolysis of MDSCs was assessed by extracellular acidification rate (ECAR), glucose consumption and lactate production. A CAC mouse model was induced by AOM and DSS to determine the therapeutic action of TEPP-46. IHC and immunofluorescence were performed to examine the expression of PKM2, PKM2 (p-Y105) and iNOS in human CRC-infiltrated MDSCs.
Results
C. tropicalis facilitates immunosuppressive function of MDSCs by increasing the expression of iNOS, COX2 and NOX2, production of nitric oxide (NO) and reactive oxygen species (ROS). Mechanistically, C. tropicalis facilitates the immunosuppressive function of MDSCs through the C-type lectin receptors Dectin-3 and Syk. C. tropicalis-enhanced immunosuppressive function of MDSCs is further dependent on aerobic glycolysis. On the one hand, NO produced by MDSCs enhanced aerobic glycolysis in a positive feedback manner. On the other hand, C. tropicalis promotes p-Syk binding to PKM2, which results in PKM2 Tyr105 phosphorylation and PKM2 nuclear translocation in MDSCs. Nuclear PKM2 interacts with HIF-1α and subsequently upregulates the expression of HIF-1α target genes encoding glycolytic enzymes, GLUT1, HK2, PKM2, LDHA and PDK1, which are required for the C. tropicalis-induced aerobic glycolysis of MDSCs. Blockade of PKM2 nuclear translocation attenuates C. tropicalis-mediated colorectal tumorigenesis. The high expression of PKM2, PKM2 (p-Y105) and iNOS in CRC-infiltrated MDSCs correlates with the development of human CRC.
Conclusion
C. tropicalis enhances immunosuppressive function of MDSCs via Syk-PKM2-HIF-1α-glycolysis signaling axis, which drives CRC. Therefore, we identify the Syk-PKM2-HIF-1α-glycolysis signaling axis as a potential therapeutic target for CRC.
Collapse
|
144
|
Lorenzana-Carrillo MA, Gopal K, Byrne NJ, Tejay S, Saleme B, Das SK, Zhang Y, Haromy A, Eaton F, Mendiola Pla M, Bowles DE, Dyck JR, Ussher JR, Michelakis ED, Sutendra G. TRIM35-mediated degradation of nuclear PKM2 destabilizes GATA4/6 and induces P53 in cardiomyocytes to promote heart failure. Sci Transl Med 2022; 14:eabm3565. [DOI: 10.1126/scitranslmed.abm3565] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
Abstract
Pyruvate kinase M2 (PKM2) is a glycolytic enzyme that translocates to the nucleus to regulate transcription factors in different tissues or pathologic states. Although studied extensively in cancer, its biological role in the heart remains unresolved. PKM1 is more abundant than the PKM2 isoform in cardiomyocytes, and thus, we speculated that PKM2 is not genetically redundant to PKM1 and may be critical in regulating cardiomyocyte-specific transcription factors important for cardiac survival. Here, we showed that nuclear PKM2 (
S37
P-PKM2) in cardiomyocytes interacts with prosurvival and proapoptotic transcription factors, including GATA4, GATA6, and P53. Cardiomyocyte-specific PKM2-deficient mice (
Pkm2
Mut Cre
+
) developed age-dependent dilated cardiac dysfunction and had decreased amounts of GATA4 and GATA6 (GATA4/6) but increased amounts of P53 compared to Control Cre
+
hearts. Nuclear PKM2 prevented caspase-1–dependent cleavage and degradation of GATA4/6 while also providing a molecular platform for MDM2-mediated reduction of P53. In a preclinical heart failure mouse model, nuclear PKM2 and GATA4/6 were decreased, whereas P53 was increased in cardiomyocytes. Loss of nuclear PKM2 was ubiquitination dependent and associated with the induction of the E3 ubiquitin ligase TRIM35. In mice, cardiomyocyte-specific TRIM35 overexpression resulted in decreased
S37
P-PKM2 and GATA4/6 along with increased P53 in cardiomyocytes compared to littermate controls and similar cardiac dysfunction to
Pkm2
Mut Cre
+
mice. In patients with dilated left ventricles, increase in TRIM35 was associated with decreased
S37
P-PKM2 and GATA4/6 and increased P53. This study supports a previously unrecognized role for PKM2 as a molecular platform that mediates cell signaling events essential for cardiac survival.
Collapse
Affiliation(s)
- Maria Areli Lorenzana-Carrillo
- Department of Medicine, University of Alberta, Edmonton, AB T6G 2R3, Canada
- Mazankowski Alberta Heart Institute, University of Alberta, Edmonton, AB T6G 2B7, Canada
- Cardiovascular Research Centre, University of Alberta, Edmonton, AB T6G 1C9, Canada
| | - Keshav Gopal
- Mazankowski Alberta Heart Institute, University of Alberta, Edmonton, AB T6G 2B7, Canada
- Cardiovascular Research Centre, University of Alberta, Edmonton, AB T6G 1C9, Canada
- Faculty of Pharmacy and Pharmaceutical Sciences, University of Alberta, Edmonton, AB T6G 2H1, Canada
| | - Nikole J. Byrne
- Mazankowski Alberta Heart Institute, University of Alberta, Edmonton, AB T6G 2B7, Canada
- Cardiovascular Research Centre, University of Alberta, Edmonton, AB T6G 1C9, Canada
- Department of Pediatrics, University of Alberta, Edmonton, AB T6G 1C9, Canada
| | - Saymon Tejay
- Department of Medicine, University of Alberta, Edmonton, AB T6G 2R3, Canada
- Mazankowski Alberta Heart Institute, University of Alberta, Edmonton, AB T6G 2B7, Canada
- Cardiovascular Research Centre, University of Alberta, Edmonton, AB T6G 1C9, Canada
| | - Bruno Saleme
- Department of Medicine, University of Alberta, Edmonton, AB T6G 2R3, Canada
- Mazankowski Alberta Heart Institute, University of Alberta, Edmonton, AB T6G 2B7, Canada
- Cardiovascular Research Centre, University of Alberta, Edmonton, AB T6G 1C9, Canada
| | - Subhash K. Das
- Department of Medicine, University of Alberta, Edmonton, AB T6G 2R3, Canada
- Mazankowski Alberta Heart Institute, University of Alberta, Edmonton, AB T6G 2B7, Canada
- Cardiovascular Research Centre, University of Alberta, Edmonton, AB T6G 1C9, Canada
| | - Yongneng Zhang
- Department of Medicine, University of Alberta, Edmonton, AB T6G 2R3, Canada
- Mazankowski Alberta Heart Institute, University of Alberta, Edmonton, AB T6G 2B7, Canada
- Cardiovascular Research Centre, University of Alberta, Edmonton, AB T6G 1C9, Canada
| | - Alois Haromy
- Department of Medicine, University of Alberta, Edmonton, AB T6G 2R3, Canada
- Mazankowski Alberta Heart Institute, University of Alberta, Edmonton, AB T6G 2B7, Canada
- Cardiovascular Research Centre, University of Alberta, Edmonton, AB T6G 1C9, Canada
| | - Farah Eaton
- Mazankowski Alberta Heart Institute, University of Alberta, Edmonton, AB T6G 2B7, Canada
- Cardiovascular Research Centre, University of Alberta, Edmonton, AB T6G 1C9, Canada
- Faculty of Pharmacy and Pharmaceutical Sciences, University of Alberta, Edmonton, AB T6G 2H1, Canada
| | | | - Dawn E. Bowles
- Department of Surgery, Duke University, Durham, NC 27710, USA
| | - Jason R. B. Dyck
- Mazankowski Alberta Heart Institute, University of Alberta, Edmonton, AB T6G 2B7, Canada
- Cardiovascular Research Centre, University of Alberta, Edmonton, AB T6G 1C9, Canada
- Department of Pediatrics, University of Alberta, Edmonton, AB T6G 1C9, Canada
| | - John R. Ussher
- Mazankowski Alberta Heart Institute, University of Alberta, Edmonton, AB T6G 2B7, Canada
- Cardiovascular Research Centre, University of Alberta, Edmonton, AB T6G 1C9, Canada
- Faculty of Pharmacy and Pharmaceutical Sciences, University of Alberta, Edmonton, AB T6G 2H1, Canada
| | - Evangelos D. Michelakis
- Department of Medicine, University of Alberta, Edmonton, AB T6G 2R3, Canada
- Mazankowski Alberta Heart Institute, University of Alberta, Edmonton, AB T6G 2B7, Canada
- Cardiovascular Research Centre, University of Alberta, Edmonton, AB T6G 1C9, Canada
| | - Gopinath Sutendra
- Department of Medicine, University of Alberta, Edmonton, AB T6G 2R3, Canada
- Mazankowski Alberta Heart Institute, University of Alberta, Edmonton, AB T6G 2B7, Canada
- Cardiovascular Research Centre, University of Alberta, Edmonton, AB T6G 1C9, Canada
- Cancer Research Institute of Northern Alberta, University of Alberta, Edmonton, AB T6G 2E1, Canada
| |
Collapse
|
145
|
Zhou N, Shen B, Bai C, Ma L, Wang S, Wu D. Nutritional deficiency induces nucleus pulposus cell apoptosis via the ATF4-PKM2-AKT signal axis. BMC Musculoskelet Disord 2022; 23:946. [PMID: 36324122 PMCID: PMC9628105 DOI: 10.1186/s12891-022-05853-1] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/11/2022] [Accepted: 09/19/2022] [Indexed: 11/05/2022] Open
Abstract
Background The intervertebral disc is the largest avascular tissue in the human body. The nucleus pulposus (NP) consumes glucose and oxygen to generate energy to maintain cellular metabolism via nutrients that diffuse from the cartilage endplate. The microenvironment in the intervertebral disc becomes nutritionally deficient during degeneration, and nutritional deficiency has been shown to inhibit the viability and proliferation of NP cells. Methods To investigate the molecular mechanism by which nutritional deficiency reduces viability and decreases proliferation, we created an in vitro model by using decreasing serum concentration percentages. Results In this study, we found that nutritional deficiency reduced NP cell viability and increased cell apoptosis and that the upregulation of ATF4 expression and the downregulation of PKM2 expression were involved in this process. Moreover, we found that PKM2 inhibition can reduce the cell apoptosis induced by ATF4 silence under nutritional deficiency. Conclusion Our findings revealed that PKM2 inhibition reduces the cell apoptosis induced by ATF4 silence under nutritional deficiency by inhibiting AKT phosphate. Revealing the function and mechanism of NP cell development under nutritional deficiency will provide new insights into the etiology, diagnosis, and treatment of intervertebral disc and related diseases. Supplementary Information The online version contains supplementary material available at 10.1186/s12891-022-05853-1.
Collapse
Affiliation(s)
- Ningfeng Zhou
- grid.24516.340000000123704535Department of Spinal Surgery, Shanghai East Hospital, Tongji University School of Medicine, Shanghai, China
| | - Bin Shen
- grid.24516.340000000123704535Department of Spinal Surgery, Shanghai East Hospital, Tongji University School of Medicine, Shanghai, China
| | - Chong Bai
- grid.24516.340000000123704535Department of Spinal Surgery, Shanghai East Hospital, Tongji University School of Medicine, Shanghai, China
| | - Liang Ma
- grid.24516.340000000123704535Department of Spinal Surgery, Shanghai East Hospital, Tongji University School of Medicine, Shanghai, China
| | - Shanjin Wang
- grid.24516.340000000123704535Department of Spinal Surgery, Shanghai East Hospital, Tongji University School of Medicine, Shanghai, China
| | - Desheng Wu
- grid.24516.340000000123704535Department of Spinal Surgery, Shanghai East Hospital, Tongji University School of Medicine, Shanghai, China
| |
Collapse
|
146
|
Lu GF, Geng F, Deng LP, Lin DC, Huang YZ, Lai SM, Lin YC, Gui LX, Sham JSK, Lin MJ. Reduced CircSMOC1 Level Promotes Metabolic Reprogramming via PTBP1 (Polypyrimidine Tract-Binding Protein) and miR-329-3p in Pulmonary Arterial Hypertension Rats. Hypertension 2022; 79:2465-2479. [PMID: 35997022 DOI: 10.1161/hypertensionaha.122.19183] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2022] [Accepted: 08/01/2022] [Indexed: 02/05/2023]
Abstract
BACKGROUND Pulmonary arterial hypertension maintains rapid cell proliferation and vascular remodeling through metabolic reprogramming. Recent studies suggested that circRNAs play important role in pulmonary vascular remodeling and pulmonary arterial smooth muscle cells proliferation. However, the relationship between circRNA, cell proliferation, and metabolic reprogramming in pulmonary arterial hypertension has not been investigated. METHODS RNA-seq and qRT-PCR reveal the differential expression profile of circRNA in pulmonary arteries of pulmonary arterial hypertension rat models. Transfection was used to examine the effects of circSMOC1 on pulmonary artery smooth muscle cells, and the roles of circSMOC1 in vivo were investigated by adenoassociated virus. Mass spectrometry, RNA pull-down, RNA immunoprecipitation, and dual-luciferase reporter assay were performed to investigate the signaling pathway of circSMOC1 regulating the metabolic reprogramming. RESULTS CircSMOC1 was significantly downregulated in pulmonary arteries of pulmonary arterial hypertension rats. CircSMOC1 knockdown promoted proliferation and migration and enhanced aerobic glycolysis of pulmonary artery smooth muscle cells. CircSMOC1 overexpression in vivo alleviates pulmonary vascular remodeling, right ventricular pressure, and right heart hypertrophy. In the nucleus, circSMOC1 directly binds to PTBP1 (polypyrimidine tract-binding protein), competitively inhibits the specific splicing of PKM (pyruvate kinase M) premRNA, resulting in the upregulation of PKM2 (pyruvate kinase M2), the key enzyme of aerobic glycolysis, to enhance glycolysis. In the cytoplasm, circSMOC1 acted as a miR-329-3p sponge, and its reduction in pulmonary arterial hypertension suppressed PDHB (pyruvate dehydrogenase E1 subunit beta) expression, leading to the impairment of mitochondrial oxidative phosphorylation. CONCLUSIONS circSMOC1 is crucially involved in the metabolic reprogramming of pulmonary artery smooth muscle cells through PTBP1 and miR-329-3p to regulate pulmonary vascular remodeling in pulmonary arterial hypertension.
Collapse
Affiliation(s)
- Gui-Feng Lu
- Key Laboratory of Fujian Province Universities on Ion Channel and Signal Transduction in Cardiovascular Diseases, Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Fujian Medical University, Fuzhou, Fujian Province, People’s Republic of China
| | - Fei Geng
- Department of Physiology and Guangdong Provincial Key Laboratory of Infectious Diseases and Molecular Immunopathology, Shantou University Medical College, Shantou, Guangdong province, People’s Republic of China
| | - Li-Ping Deng
- Key Laboratory of Fujian Province Universities on Ion Channel and Signal Transduction in Cardiovascular Diseases, Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Fujian Medical University, Fuzhou, Fujian Province, People’s Republic of China
| | - Da-Cen Lin
- Department of Epidemiology and Health Statistics, School of Public Health, Fujian Medical University, Fuzhou, Fujian Province, People’s Republic of China
| | - Yan-Zhen Huang
- Key Laboratory of Fujian Province Universities on Ion Channel and Signal Transduction in Cardiovascular Diseases, Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Fujian Medical University, Fuzhou, Fujian Province, People’s Republic of China
| | - Su-Mei Lai
- Key Laboratory of Fujian Province Universities on Ion Channel and Signal Transduction in Cardiovascular Diseases, Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Fujian Medical University, Fuzhou, Fujian Province, People’s Republic of China
| | - Yi-Chen Lin
- Key Laboratory of Fujian Province Universities on Ion Channel and Signal Transduction in Cardiovascular Diseases, Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Fujian Medical University, Fuzhou, Fujian Province, People’s Republic of China
| | - Long-Xin Gui
- Key Laboratory of Fujian Province Universities on Ion Channel and Signal Transduction in Cardiovascular Diseases, Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Fujian Medical University, Fuzhou, Fujian Province, People’s Republic of China
| | - James S K Sham
- Division of Pulmonary and Critical Care Medicine, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Mo-Jun Lin
- Key Laboratory of Fujian Province Universities on Ion Channel and Signal Transduction in Cardiovascular Diseases, Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Fujian Medical University, Fuzhou, Fujian Province, People’s Republic of China
| |
Collapse
|
147
|
Zandi M, Shokri S, Mahmoudvand S, Hosseinzadeh Adli A, Mohammadi R, Haddadi A. Interplay between cellular metabolism and DNA viruses. J Med Virol 2022; 94:5163-5173. [PMID: 35869415 DOI: 10.1002/jmv.28018] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2022] [Revised: 07/15/2022] [Accepted: 07/21/2022] [Indexed: 12/15/2022]
Abstract
Viruses as intracellular pathogens take over the host metabolism and reprogram to facilitate optimal virus production. DNA viruses can cause alterations in several metabolic pathways, including aerobic glycolysis also known as the Warburg effect, pentose phosphate pathway activation, and amino acid catabolism such as glutaminolysis, nucleotide biosynthesis, lipid metabolism, and amino acid biosynthesis. The available energy for productive infection can be increased in infected cells via modification of different carbon source utilization. This review discusses the metabolic alterations of the DNA viruses that will be the basis for future novel therapeutic approaches.
Collapse
Affiliation(s)
- Milad Zandi
- Department of Virology, School of Public Health, Tehran University of Medical Sciences, Tehran, Iran
| | - Somayeh Shokri
- Research Center for Molecular Medicine, Hamadan University of Medical Sciences, Hamadan, Iran.,Department of Medical Virology, Faculty of Medicine, Hamadan University of Medical Sciences, Hamadan, Iran
| | - Shahab Mahmoudvand
- Research Center for Molecular Medicine, Hamadan University of Medical Sciences, Hamadan, Iran.,Department of Medical Virology, Faculty of Medicine, Hamadan University of Medical Sciences, Hamadan, Iran
| | - Ahmad Hosseinzadeh Adli
- Department of Virology, School of Public Health, Tehran University of Medical Sciences, Tehran, Iran.,Department of Bacteriology and Virology, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Ramin Mohammadi
- College of Pharmacy and Nutrition, University of Saskatchewan, Saskatoon, Saskatchewan, Canada
| | - Azita Haddadi
- College of Pharmacy and Nutrition, University of Saskatchewan, Saskatoon, Saskatchewan, Canada
| |
Collapse
|
148
|
Panax notoginseng saponins alleviates inflammation induced by microglial activation and protects against ischemic brain injury via inhibiting HIF-1α/PKM2/STAT3 signaling. Biomed Pharmacother 2022; 155:113479. [DOI: 10.1016/j.biopha.2022.113479] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2022] [Revised: 07/22/2022] [Accepted: 07/26/2022] [Indexed: 11/02/2022] Open
|
149
|
Dai T, Zhang X, Zhou X, Hu X, Huang X, Xing F, Tian H, Li Y. Long non-coding RNA VAL facilitates PKM2 enzymatic activity to promote glycolysis and malignancy of gastric cancer. Clin Transl Med 2022; 12:e1088. [PMID: 36229913 PMCID: PMC9561166 DOI: 10.1002/ctm2.1088] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2022] [Revised: 09/25/2022] [Accepted: 09/29/2022] [Indexed: 01/28/2023] Open
Abstract
BACKGROUND Gastric cancer (GC) is one of the most common types of cancer worldwide, which leads to more than 10% of cancer-related deaths. Metabolism reprogramming presents as a pivotal event in cancer initiation and progression through enhancing aerobic glycolysis and anabolic metabolism. However, the underlying regulatory mechanisms in GC remain unknown. METHODS VAL was identified by bioinformatics analyses in GC. Cell-based assays and mouse model illustrate the role of VAL in GC. RNA pull-down, immunoprecipitation assay and Western blot elucidate the interaction between VAL and PKM2. Pyruvate kinase activity, ECAR and OCR were measured to validate aerobic glycolysis of GC cells. RESULTS Long non-coding RNA (lncRNA) VAL is significantly upregulated in GCs and indicates poor prognosis. Functional assays showed that VAL promotes GC malignant progression. Mechanistically, VAL strengthens the enzymatic activity of PKM2 and aerobic glycolysis of GC cells through directly binding with PKM2 to abrogate the PKM2-Parkin interaction, and to suppress Parkin-induced polyubiquitination of PKM2. In addition, glucose starvation induces VAL expression to enhance this process. CONCLUSIONS Our study provides an insight into an lncRNA-dependent regulation on the enzymatic activity of PKM2, and suggests a potential of targeting VAL or PKM2 as promising biomarkers in GC diagnosis and treatment.
Collapse
Affiliation(s)
- Ting Dai
- Institute of Tissue Transplantation and Immunology, Department of ImmunobiologyJinan UniversityGuangzhouGuangdongChina,GMU‐GIBH Joint School of Life SciencesGuangzhou Medical UniversityGuangzhouGuangdongChina
| | - Xin Zhang
- Clinical Experimental Center, Jiangmen Key Laboratory of Clinical Biobanks and Translational Research, Jiangmen Central HospitalAffiliated Jiangmen Hospital of Sun Yat‐sen UniversityJiangmenChina
| | - Xiang Zhou
- Department of Microsurgery, Trauma and Hand SurgeryThe First Affiliated Hospital of Sun Yat‐sen UniversityGuangzhouGuangdongChina
| | - Xiaoxia Hu
- Institute of Tissue Transplantation and Immunology, Department of ImmunobiologyJinan UniversityGuangzhouGuangdongChina
| | - Xiaodi Huang
- Institute of Tissue Transplantation and Immunology, Department of ImmunobiologyJinan UniversityGuangzhouGuangdongChina
| | - Feiyue Xing
- Institute of Tissue Transplantation and Immunology, Department of ImmunobiologyJinan UniversityGuangzhouGuangdongChina
| | - Han Tian
- Zhongshan School of MedicineSun Yat‐sen UniversityGuangzhouGuangdongChina
| | - Yun Li
- Institute of Tissue Transplantation and Immunology, Department of ImmunobiologyJinan UniversityGuangzhouGuangdongChina
| |
Collapse
|
150
|
Zhou J, Lei N, Tian W, Guo R, Chen M, Qiu L, Wu F, Li Y, Chang L. Recent progress of the tumor microenvironmental metabolism in cervical cancer radioresistance. Front Oncol 2022; 12:999643. [PMID: 36313645 PMCID: PMC9597614 DOI: 10.3389/fonc.2022.999643] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2022] [Accepted: 09/27/2022] [Indexed: 08/01/2023] Open
Abstract
Radiotherapy is widely used as an indispensable treatment option for cervical cancer patients. However, radioresistance always occurs and has become a big obstacle to treatment efficacy. The reason for radioresistance is mainly attributed to the high repair ability of tumor cells that overcome the DNA damage caused by radiotherapy, and the increased self-healing ability of cancer stem cells (CSCs). Accumulating findings have demonstrated that the tumor microenvironment (TME) is closely related to cervical cancer radioresistance in many aspects, especially in the metabolic processes. In this review, we discuss radiotherapy in cervical cancer radioresistance, and focus on recent research progress of the TME metabolism that affects radioresistance in cervical cancer. Understanding the mechanism of metabolism in cervical cancer radioresistance may help identify useful therapeutic targets for developing novel therapy, overcome radioresistance and improve the efficacy of radiotherapy in clinics and quality of life of patients.
Collapse
Affiliation(s)
- Junying Zhou
- Department of Obstetrics and Gynecology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Ningjing Lei
- School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, China
| | - Wanjia Tian
- Department of Obstetrics and Gynecology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Ruixia Guo
- Department of Obstetrics and Gynecology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Mengyu Chen
- Department of Obstetrics and Gynecology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Luojie Qiu
- Department of Obstetrics and Gynecology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Fengling Wu
- Department of Obstetrics and Gynecology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Yong Li
- Cancer Care Centre, St George Hospital, Kogarah, NSW, Australia
- St George and Sutherland Clinical Campuses, School of Clinical Medicine, University of New South Wales (UNSW) Sydney, Kensington, NSW, Australia
| | - Lei Chang
- Department of Obstetrics and Gynecology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| |
Collapse
|