1
|
Hong Z, Wang J, Hu B, Tu X, Yang J, Sun W, Duan X. Esculetin inhibits liver cancer by targeting glucose-6-phosphate isomerase mediated glycolysis. Biomed Pharmacother 2025; 188:118118. [PMID: 40373632 DOI: 10.1016/j.biopha.2025.118118] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2025] [Revised: 04/26/2025] [Accepted: 04/30/2025] [Indexed: 05/17/2025] Open
Abstract
BACKGROUND Liver cancer is challenging to detect in its early stages, and the global incidence rate and mortality associated with this disease have reached alarming levels. Currently, treatment options for liver cancer are limited, and there is a significant lack of safe and effective therapeutic agents. Esculetin is a natural product, exhibits almost non-toxic and inhibitory properties against various malignancies, making it a subject worthy of further investigation in liver cancer. METHODS In this study, potential targets of esculetin in liver cancer were identified through transcriptomics, network pharmacology, and molecular docking technologies, and gene interference. Direct binding targets of esculetin were identified using surface plasmon resonance (SPR). The molecular mechanisms by which esculetin affects glucose metabolism in liver cancer were also explored. Finally, the activity against liver cancer and mechanisms of action of esculetin were validated in vivo using a mouse tumor model. RESULTS Glucose-6-phosphate isomerase (GPI) was shown to have a direct binding affinity for this compound. Esculetin inhibits glycolysis in liver cancer through its interaction with GPI and it was shown to exert a significant inhibitory effect on the genes and proteins associated with glycolysis such as ALDOA, ENO1, GAPDH, LDHA, PFKL, PGAM1, PGK1, and PKM2. Furthermore, esculetin not only suppresses the growth of liver cancer cells in vitro but also exhibits notable anti-tumor effects in vivo. CONCLUSIONS This study demonstrated the inhibitory effects of esculetin against liver cancer both in vitro and in vivo, demonstrating inhibition of glycolysis in liver cancer cells. In addition, the key glycolysis enzyme GPI was identified as a direct target of esculetin.
Collapse
Affiliation(s)
- Zongchao Hong
- Wuling Mountain Traditional Chinese Medicine Inspection and Testing Center, Hubei Minzu University, Enshi, China; Health Science Center, Hubei Minzu University, Enshi, China; Hubei Provincial Key Laboratory of Occurrence and Intervention of Rheumatic diseases,Hubei Minzu University, Enshi, China.
| | - Jingbo Wang
- Department of Integrated Traditional Chinese and Western Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.
| | - Baodan Hu
- The Central Hospital of Enshi Tujia and Miao Autonomous Prefecture, Enshi, China.
| | - Xin Tu
- Wuling Mountain Traditional Chinese Medicine Inspection and Testing Center, Hubei Minzu University, Enshi, China
| | - Jin Yang
- Health Science Center, Hubei Minzu University, Enshi, China
| | - Wanjin Sun
- Hubei Key Laboratory of theory and application research of liver and kidney in traditional Chinese medicine, Hubei Provincial Hospital of Traditional Chinese Medicine, Wuhan, China.
| | - Xueyun Duan
- Hubei Key Laboratory of theory and application research of liver and kidney in traditional Chinese medicine, Hubei Provincial Hospital of Traditional Chinese Medicine, Wuhan, China.
| |
Collapse
|
2
|
Pan Z, Liu Y, Li H, Qiu H, Zhang P, Li Z, Wang X, Tian Y, Feng Z, Zhu S, Wang X. The role and mechanism of aerobic glycolysis in nasopharyngeal carcinoma. PeerJ 2025; 13:e19213. [PMID: 40191756 PMCID: PMC11971989 DOI: 10.7717/peerj.19213] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2024] [Accepted: 03/05/2025] [Indexed: 04/09/2025] Open
Abstract
This review delves into the pivotal role and intricate mechanisms of aerobic glycolysis in nasopharyngeal carcinoma (NPC). NPC, a malignancy originating from the nasopharyngeal epithelium, displays distinct geographical and clinical features. The article emphasizes the significance of aerobic glycolysis, a pivotal metabolic alteration in cancer cells, in NPC progression. Key enzymes such as hexokinase 2, lactate dehydrogenase A, phosphofructokinase 1, and pyruvate kinase M2 are discussed for their regulatory functions in NPC glycolysis through signaling pathways like PI3K/Akt and mTOR. Further, the article explores how oncogenic signaling pathways and transcription factors like c-Myc and HIF-1α modulate aerobic glycolysis, thereby affecting NPC's proliferation, invasion, metastasis, angiogenesis, and immune evasion. By elucidating these mechanisms, the review aims to advance research and clinical practice in NPC, informing the development of targeted therapeutic strategies that enhance treatment precision and reduce side effects. Overall, this review offers a broad understanding of the multifaceted role of aerobic glycolysis in NPC and its potential impact on therapeutic outcomes.
Collapse
Affiliation(s)
- Zhiyong Pan
- Department of Radiotherapy, Affiliated Qingyuan Hospital, Guangzhou Medical University, Qingyuan, Guangdong, China
| | - Yuyi Liu
- Department of Radiotherapy, Affiliated Qingyuan Hospital, Guangzhou Medical University, Qingyuan, Guangdong, China
| | - Hui Li
- Department of Ophthalmology, Affiliated Qingyuan Hospital, Guangzhou Medical University, Qingyuan, Guangdong, China
| | - Huisi Qiu
- Department of Radiotherapy, Affiliated Qingyuan Hospital, Guangzhou Medical University, Qingyuan, Guangdong, China
| | - Pingmei Zhang
- Department of Radiotherapy, Affiliated Qingyuan Hospital, Guangzhou Medical University, Qingyuan, Guangdong, China
| | - Zhiying Li
- Department of Radiotherapy, Affiliated Qingyuan Hospital, Guangzhou Medical University, Qingyuan, Guangdong, China
| | - Xinyu Wang
- Department of Radiotherapy, Affiliated Qingyuan Hospital, Guangzhou Medical University, Qingyuan, Guangdong, China
| | - Yuxiao Tian
- Department of Radiotherapy, Affiliated Qingyuan Hospital, Guangzhou Medical University, Qingyuan, Guangdong, China
| | - Zhengfu Feng
- Department of Radiotherapy, Affiliated Qingyuan Hospital, Guangzhou Medical University, Qingyuan, Guangdong, China
| | - Song Zhu
- Department of Radiotherapy, Affiliated Qingyuan Hospital, Guangzhou Medical University, Qingyuan, Guangdong, China
| | - Xin Wang
- Department of Radiotherapy, Affiliated Qingyuan Hospital, Guangzhou Medical University, Qingyuan, Guangdong, China
| |
Collapse
|
3
|
Jing D, Wu W, Huang X, Zhang Z, Chen X, Huang F, Liu J, Zhang Z, Shao Z, Pu F. Transketolase promotes osteosarcoma progression through the YY1-PAK4 axis. FEBS J 2025; 292:1798-1812. [PMID: 39853945 DOI: 10.1111/febs.17375] [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: 10/23/2023] [Revised: 05/18/2024] [Accepted: 12/16/2024] [Indexed: 01/26/2025]
Abstract
Osteosarcoma, a malignant bone tumor that occurs in adolescents, proliferates and is prone to pulmonary metastasis. Osteosarcoma is characterized by high genotypic heterogeneity, making it difficult to identify reliable anti-osteosarcoma targets. The genotype of osteosarcoma may be highly dynamic, but its high dependence on energy remains constant. Fortunately, tumors tend to have relatively consistent metabolic types. Targeting metabolism with anti-tumor therapies is a new strategy for treating tumors. Genes related to carbohydrate metabolism are widely and highly expressed in tumor tissues. Transketolase (TKT), a key enzyme at the non-oxidative stage of the pentose phosphate pathway, is up-regulated in various tumors. In the present study, TKT promoted osteosarcoma cell proliferation non-metabolically. Specifically, TKT bound directly to amino acid residues of Yin Yang 1 (YY1) at amino acids 201-228, stimulating YY1 to bind to the promoter of P21 activated kinase 4 (PAK4) and resulting in PAK4 expression and activation of the phosphoinositide 3-kinase-Akt signaling pathway. Additionally, we designed a peptide, YY1-PEP, based on the exact mechanism of how TKT promotes osteosarcoma. Per in vivo and in vitro experiments, YY1-PEP displayed anti-osteosarcoma properties. The present study provides a new feasible strategy against osteosarcoma progression.
Collapse
Affiliation(s)
- Doudou Jing
- Department of Orthopaedics, The Second Hospital of Shanxi Medical University, Taiyuan, China
- Department of Orthopedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Wei Wu
- Department of Orthopedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Xin Huang
- Department of Orthopedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Zhenhao Zhang
- Department of Orthopedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Xuanzuo Chen
- Department of Orthopedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Fuhua Huang
- Department of Orthopedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Jianxiang Liu
- Department of Orthopedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Zhicai Zhang
- Department of Orthopedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Zengwu Shao
- Department of Orthopedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Feifei Pu
- Department of Orthopedics, Wuhan Hospital of Traditional Chinese and Western Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Department of Orthopedics, Wuhan No.1 Hospital, China
| |
Collapse
|
4
|
Rojas-Pirela M, Andrade-Alviárez D, Rojas V, Marcos M, Salete-Granado D, Chacón-Arnaude M, Pérez-Nieto MÁ, Kemmerling U, Concepción JL, Michels PAM, Quiñones W. Exploring glycolytic enzymes in disease: potential biomarkers and therapeutic targets in neurodegeneration, cancer and parasitic infections. Open Biol 2025; 15:240239. [PMID: 39904372 PMCID: PMC11793985 DOI: 10.1098/rsob.240239] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2024] [Revised: 12/11/2024] [Accepted: 12/16/2024] [Indexed: 02/06/2025] Open
Abstract
Glycolysis, present in most organisms, is evolutionarily one of the oldest metabolic pathways. It has great relevance at a physiological level because it is responsible for generating ATP in the cell through the conversion of glucose into pyruvate and reducing nicotinamide adenine dinucleotide (NADH) (that may be fed into the electron chain in the mitochondria to produce additional ATP by oxidative phosphorylation), as well as for producing intermediates that can serve as substrates for other metabolic processes. Glycolysis takes place through 10 consecutive chemical reactions, each of which is catalysed by a specific enzyme. Although energy transduction by glucose metabolism is the main function of this pathway, involvement in virulence, growth, pathogen-host interactions, immunomodulation and adaptation to environmental conditions are other functions attributed to this metabolic pathway. In humans, where glycolysis occurs mainly in the cytosol, the mislocalization of some glycolytic enzymes in various other subcellular locations, as well as alterations in their expression and regulation, has been associated with the development and progression of various diseases. In this review, we describe the role of glycolytic enzymes in the pathogenesis of diseases of clinical interest. In addition, the potential role of these enzymes as targets for drug development and their potential for use as diagnostic and prognostic markers of some pathologies are also discussed.
Collapse
Affiliation(s)
- Maura Rojas-Pirela
- Instituto de Investigación Biomédica de Salamanca (IBSAL), Salamanca37007, Spain
- Unidad de Medicina Molecular, Departamento de Medicina, Universidad de Salamanca, Salamanca37007, Spain
- Servicio de Medicina Interna, Hospital Universitario de Salamanca, Salamanca37007, Spain
| | - Diego Andrade-Alviárez
- Laboratorio de Enzimología de Parásitos, Departamento de Biología, Facultad de Ciencias, Universidad de Los Andes, Mérida5101, Venezuela
| | - Verónica Rojas
- Instituto de Biología, Facultad de Ciencias, Pontificia Universidad Católica de Valparaíso, Valparaíso2373223, Chile
| | - Miguel Marcos
- Instituto de Investigación Biomédica de Salamanca (IBSAL), Salamanca37007, Spain
- Unidad de Medicina Molecular, Departamento de Medicina, Universidad de Salamanca, Salamanca37007, Spain
- Servicio de Medicina Interna, Hospital Universitario de Salamanca, Salamanca37007, Spain
| | - Daniel Salete-Granado
- Instituto de Investigación Biomédica de Salamanca (IBSAL), Salamanca37007, Spain
- Unidad de Medicina Molecular, Departamento de Medicina, Universidad de Salamanca, Salamanca37007, Spain
| | - Marirene Chacón-Arnaude
- Laboratorio de Enzimología de Parásitos, Departamento de Biología, Facultad de Ciencias, Universidad de Los Andes, Mérida5101, Venezuela
| | - María Á. Pérez-Nieto
- Unidad de Medicina Molecular, Departamento de Medicina, Universidad de Salamanca, Salamanca37007, Spain
- Fundación Instituto de Estudios de Ciencias de la Salud de Castilla y León, Soria42002, Spain
| | - Ulrike Kemmerling
- Instituto de Ciencias Biomédicas, Universidad de Chile, Facultad de Medicina, Santiago de Chile8380453, Chile
| | - Juan Luis Concepción
- Laboratorio de Enzimología de Parásitos, Departamento de Biología, Facultad de Ciencias, Universidad de Los Andes, Mérida5101, Venezuela
| | - Paul A. M. Michels
- School of Biological Sciences, University of Edinburgh, The King’s Buildings, EdinburghEH9 3FL, UK
| | - Wilfredo Quiñones
- Laboratorio de Enzimología de Parásitos, Departamento de Biología, Facultad de Ciencias, Universidad de Los Andes, Mérida5101, Venezuela
| |
Collapse
|
5
|
Suh JY, Sim DY, Ahn CH, Park SY, Shim BS, Kim B, Lee DY, Jeong HB, Lee HE, Kim SH. Crucial Role of c-Myc/Monocarboxylate Transporter 4 Signaling in Capsaicin Induced Apoptotic and Anti-Warburg Effects in Hepatocellular Carcinoma. Phytother Res 2025; 39:536-547. [PMID: 39655472 DOI: 10.1002/ptr.8388] [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: 06/05/2024] [Revised: 10/10/2024] [Accepted: 10/29/2024] [Indexed: 01/30/2025]
Abstract
Though Capsaicin from chili peppers was known to have antitumor effects in several cancers, the underlying antitumor pathogenesis of Capsaicin is not clear to date. Thus, the antitumor mechanism of Capsaicin was explored in Hep3B and Huh7 hepatocellular carcinoma (HCC) cells in relation to c-Myc/monocarboxylate transporter 4 (MCT4) signaling. To elucidate the antitumor mechanism of capsaicin, cytotoxicity assay, cell cycle analysis, Western blotting, RT-qPCR, RNA interference, ELISA, immunoprecipitation, and mouse xenograft model were used in this work. Capsaicin increased the cytotoxicity, subG1 population, and the number of TUNEL-positive bodies in Huh7 and Hep3B cells. Consistently, Capsaicin diminished the expression of pro-PARP, HK2, PKM2, LDHA, glucose transporter type 1 (Glut1), c-Myc, and monocarboxylate transporter 4 (MCT4) in Huh7 and Hep3B cells, along with decreased production of glucose, lactate, and ATP. However, a glycolysis end product pyruvate treatment reversed the capacity of Capsaicin to attenuate the expression of pro-PARP, HK2, c-Myc, and MCT4 in Hep3B cells. Furthermore, Capsaicin reduced c-Myc stability in the presence of cycloheximide and induced c-Myc ubiquitination in Hep3B cells, while c-Myc directly binds to MCT4 as a lactate transporter and downstream of c-Myc in Hep3B cells by immunoprecipitation and correlation factor (Spearman efficient = 0.0027). Furthermore, a preliminary analysis of an animal study reveals that Capsaicin significantly suppressed the growth of Hep3B cells inoculated in BALB/c nude mice without hurting body weight, liver, and spleen. Our findings provide novel evidence that Capsaicin exerts apoptotic and anti-Warburg effect via c-Myc/MCT4 signaling axis as a potent anticancer candidate for liver cancer therapy.
Collapse
Affiliation(s)
- Jin Young Suh
- College of Korean Medicine, Kyung Hee University, Seoul, Republic of Korea
| | - Deok Yong Sim
- College of Korean Medicine, Kyung Hee University, Seoul, Republic of Korea
| | - Chi-Hoon Ahn
- College of Korean Medicine, Kyung Hee University, Seoul, Republic of Korea
| | - Su-Yeon Park
- College of Korean Medicine, Kyung Hee University, Seoul, Republic of Korea
| | - Bum-Sang Shim
- College of Korean Medicine, Kyung Hee University, Seoul, Republic of Korea
| | - Bonglee Kim
- College of Korean Medicine, Kyung Hee University, Seoul, Republic of Korea
| | - Dae Young Lee
- BK21 FOUR KNU Creative BioResearch Group, School of Life Sciences, Kyungpook National University, Daegu, Republic of Korea
| | - Hyo Bong Jeong
- Department of Horticultural Crop Research, National Institute of Horticultural and Herbal Science, Rural Development Administration, Wanju, Republic of Korea
| | - Hye Eun Lee
- Department of Horticultural Crop Research, National Institute of Horticultural and Herbal Science, Rural Development Administration, Wanju, Republic of Korea
| | - Sung-Hoon Kim
- College of Korean Medicine, Kyung Hee University, Seoul, Republic of Korea
| |
Collapse
|
6
|
Juretić D, Bonačić Lošić Ž. Theoretical Improvements in Enzyme Efficiency Associated with Noisy Rate Constants and Increased Dissipation. ENTROPY (BASEL, SWITZERLAND) 2024; 26:151. [PMID: 38392406 PMCID: PMC10888251 DOI: 10.3390/e26020151] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/30/2023] [Revised: 01/18/2024] [Accepted: 02/05/2024] [Indexed: 02/24/2024]
Abstract
Previous studies have revealed the extraordinarily large catalytic efficiency of some enzymes. High catalytic proficiency is an essential accomplishment of biological evolution. Natural selection led to the increased turnover number, kcat, and enzyme efficiency, kcat/KM, of uni-uni enzymes, which convert a single substrate into a single product. We added or multiplied random noise with chosen rate constants to explore the correlation between dissipation and catalytic efficiency for ten enzymes: beta-galactosidase, glucose isomerase, β-lactamases from three bacterial strains, ketosteroid isomerase, triosephosphate isomerase, and carbonic anhydrase I, II, and T200H. Our results highlight the role of biological evolution in accelerating thermodynamic evolution. The catalytic performance of these enzymes is proportional to overall entropy production-the main parameter from irreversible thermodynamics. That parameter is also proportional to the evolutionary distance of β-lactamases PC1, RTEM, and Lac-1 when natural or artificial evolution produces the optimal or maximal possible catalytic efficiency. De novo enzyme design and attempts to speed up the rate-limiting catalytic steps may profit from the described connection between kinetics and thermodynamics.
Collapse
Affiliation(s)
- Davor Juretić
- Mediterranean Institute for Life Sciences, Šetalište Ivana Meštrovića 45, 21000 Split, Croatia
- Faculty of Science, University of Split, Ruđera Boškovića 33, 21000 Split, Croatia
| | | |
Collapse
|
7
|
Xiao QH, Li ZZ, Ren L, Wang SY, Li XQ, Bai HX, Qiao RZ, Tang N, Liu WJ, Wang JM, Ma GY, Dong DC, Wu KH, Cao W. α-Glucan derivatives as selective blockers of aldolase A: Computer-aided structure optimization and the effects on HCC. Carbohydr Polym 2024; 325:121566. [PMID: 38008473 DOI: 10.1016/j.carbpol.2023.121566] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2023] [Revised: 10/16/2023] [Accepted: 11/04/2023] [Indexed: 11/28/2023]
Abstract
Aldolase A (ALDOA) promotes hepatocellular carcinoma (HCC) growth and is a potential therapeutic target. A previous study found an α-D-glucan (α-D-(1,6)-Glcp-α-D-(1,4)-Glcp, 10.0:1.0), named HDPS-4II, that could specifically inhibit ALDOA but its activity was not high enough. In this study, the derivatives of α-D-glucan binding to ALDOA were optimized using molecular docking, and its sulfated modification demonstrated the highest affinity with ALDOA among sulfated, carboxylated, and aminated derivatives. Sulfated HDPS-4II and dextrans with different molecular weights (1000 Da, 3000 Da, and 4000 Da) were prepared. Using MST assay, 3-O-sulfated HDPS-4II (SHDPS-4II) and 1000 Da dextran (SDextran1) showed higher affinities to ALDOA with Kd of 1.83 μM and 85.04 μM, respectively. Furthermore, SHDPS-4II and SDextran1 markedly inhibited the proliferation of HCC cells both in vitro and in vivo by blocking ALDOA. These results demonstrate that sulfated modification of α-D-glucans could enhance their affinities with ALDOA and anti-HCC effects.
Collapse
Affiliation(s)
- Qian-Han Xiao
- Shaanxi Key Laboratory of Natural Products & Chemical Biology, School of Chemistry & Pharmacy, Northwest A&F University, Yangling 712100, China
| | - Ze-Zhi Li
- Shaanxi Key Laboratory of Natural Products & Chemical Biology, School of Chemistry & Pharmacy, Northwest A&F University, Yangling 712100, China
| | - Li Ren
- Shaanxi Key Laboratory of Natural Products & Chemical Biology, School of Chemistry & Pharmacy, Northwest A&F University, Yangling 712100, China
| | - Shu-Yao Wang
- Shaanxi Key Laboratory of Natural Products & Chemical Biology, School of Chemistry & Pharmacy, Northwest A&F University, Yangling 712100, China
| | - Xiao-Qiang Li
- Department of Pharmacology, Key Laboratory of Gastrointestinal Pharmacology of Chinese Materia Medica of the State Administration of Traditional Chinese Medicine, School of Pharmacy, Air Force Medical University, Xi'an, China
| | - Hong-Xin Bai
- Shaanxi Key Laboratory of Natural Products & Chemical Biology, School of Chemistry & Pharmacy, Northwest A&F University, Yangling 712100, China
| | - Rui-Zhi Qiao
- Shaanxi Key Laboratory of Natural Products & Chemical Biology, School of Chemistry & Pharmacy, Northwest A&F University, Yangling 712100, China
| | - Na Tang
- Shaanxi Key Laboratory of Natural Products & Chemical Biology, School of Chemistry & Pharmacy, Northwest A&F University, Yangling 712100, China
| | - Wen-Juan Liu
- Shaanxi Key Laboratory of Natural Products & Chemical Biology, School of Chemistry & Pharmacy, Northwest A&F University, Yangling 712100, China
| | - Jing-Mei Wang
- Shaanxi Key Laboratory of Natural Products & Chemical Biology, School of Chemistry & Pharmacy, Northwest A&F University, Yangling 712100, China
| | - Guang-Yuan Ma
- Shaanxi Key Laboratory of Natural Products & Chemical Biology, School of Chemistry & Pharmacy, Northwest A&F University, Yangling 712100, China
| | - Dian-Chao Dong
- Shaanxi Key Laboratory of Natural Products & Chemical Biology, School of Chemistry & Pharmacy, Northwest A&F University, Yangling 712100, China
| | - Ke-Han Wu
- Shaanxi Key Laboratory of Natural Products & Chemical Biology, School of Chemistry & Pharmacy, Northwest A&F University, Yangling 712100, China
| | - Wei Cao
- Shaanxi Key Laboratory of Natural Products & Chemical Biology, School of Chemistry & Pharmacy, Northwest A&F University, Yangling 712100, China; Department of Pharmacology, Key Laboratory of Gastrointestinal Pharmacology of Chinese Materia Medica of the State Administration of Traditional Chinese Medicine, School of Pharmacy, Air Force Medical University, Xi'an, China.
| |
Collapse
|
8
|
Yan L, Sun H, Chen Y, Yu X, Zhang J, Li P. FOXP2 suppresses the proliferation, invasion, and aerobic glycolysis of hepatocellular carcinoma cells by regulating the KDM5A/FBP1 axis. ENVIRONMENTAL TOXICOLOGY 2024; 39:341-356. [PMID: 37713600 DOI: 10.1002/tox.23971] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/06/2023] [Revised: 08/04/2023] [Accepted: 08/30/2023] [Indexed: 09/17/2023]
Abstract
The Warburg effect is the preference of cancer cells to use glycolysis rather than oxidative phosphorylation to generate energy. Accumulating evidence suggests that aerobic glycolysis is widespread in hepatocellular carcinoma (HCC) and closely related to tumorigenesis. The purpose of this study was to investigate the role and mechanism of forkhead box P2 (FOXP2) in aerobic glycolysis and tumorigenesis in HCC. Here, we found that FOXP2 was lower expressed in HCC tissues and cells than in nontumor tissues and normal hepatocytes. Overexpression of FOXP2 suppressed cell proliferation and invasion of HCC cells and promoted cell apoptosis in vitro, and hindered the growth of mouse xenograft tumors in vivo. Further researches showed that FOXP2 inhibited the Warburg effect in HCC cells. Moreover, we demonstrated that FOXP2 up-regulated the expression of fructose-1, 6-diphosphatase (FBP1), and the inhibitory effect of FOXP2 on glycolysis was dependent on FBP1. Mechanistically, as a transcription factor, FOXP2 negatively regulated the transcription of lysine-specific demethylase 5A (KDM5A), and then blocked KDM5A-induced H3K4me3 demethylation in FBP1 promoter region, thereby promoting the expression of FBP1. Consistently, overexpressing KDM5A or silencing FBP1 effectively reversed the inhibitory effect of FOXP2 on HCC progression. Together, our findings revealed the mechanistic role of the FOXP2/KDM5A/FBP1 axis in glycolysis and malignant progression of HCC cells, providing a potential molecular target for the therapy of HCC.
Collapse
Affiliation(s)
- Lijing Yan
- Department of Endocrinology, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Huanhuan Sun
- Department of Gastroenterology, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Yuling Chen
- Department of Gastroenterology, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Xiaohui Yu
- Department of Respiratory Medicine, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Jingru Zhang
- Department of Gastroenterology, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Peijie Li
- Department of Gastroenterology, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| |
Collapse
|
9
|
Trejo-Solis C, Silva-Adaya D, Serrano-García N, Magaña-Maldonado R, Jimenez-Farfan D, Ferreira-Guerrero E, Cruz-Salgado A, Castillo-Rodriguez RA. Role of Glycolytic and Glutamine Metabolism Reprogramming on the Proliferation, Invasion, and Apoptosis Resistance through Modulation of Signaling Pathways in Glioblastoma. Int J Mol Sci 2023; 24:17633. [PMID: 38139462 PMCID: PMC10744281 DOI: 10.3390/ijms242417633] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2023] [Revised: 12/11/2023] [Accepted: 12/14/2023] [Indexed: 12/24/2023] Open
Abstract
Glioma cells exhibit genetic and metabolic alterations that affect the deregulation of several cellular signal transduction pathways, including those related to glucose metabolism. Moreover, oncogenic signaling pathways induce the expression of metabolic genes, increasing the metabolic enzyme activities and thus the critical biosynthetic pathways to generate nucleotides, amino acids, and fatty acids, which provide energy and metabolic intermediates that are essential to accomplish the biosynthetic needs of glioma cells. In this review, we aim to explore how dysregulated metabolic enzymes and their metabolites from primary metabolism pathways in glioblastoma (GBM) such as glycolysis and glutaminolysis modulate anabolic and catabolic metabolic pathways as well as pro-oncogenic signaling and contribute to the formation, survival, growth, and malignancy of glioma cells. Also, we discuss promising therapeutic strategies by targeting the key players in metabolic regulation. Therefore, the knowledge of metabolic reprogramming is necessary to fully understand the biology of malignant gliomas to improve patient survival significantly.
Collapse
Affiliation(s)
- Cristina Trejo-Solis
- Laboratorio Experimental de Enfermedades Neurodegenerativas, Laboratorio de Reprogramación Celular, Departamento de Neurofisiología, Instituto Nacional de Neurología y Neurocirugía, Ciudad de Mexico 14269, Mexico; (D.S.-A.); (N.S.-G.); (R.M.-M.)
| | - Daniela Silva-Adaya
- Laboratorio Experimental de Enfermedades Neurodegenerativas, Laboratorio de Reprogramación Celular, Departamento de Neurofisiología, Instituto Nacional de Neurología y Neurocirugía, Ciudad de Mexico 14269, Mexico; (D.S.-A.); (N.S.-G.); (R.M.-M.)
| | - Norma Serrano-García
- Laboratorio Experimental de Enfermedades Neurodegenerativas, Laboratorio de Reprogramación Celular, Departamento de Neurofisiología, Instituto Nacional de Neurología y Neurocirugía, Ciudad de Mexico 14269, Mexico; (D.S.-A.); (N.S.-G.); (R.M.-M.)
| | - Roxana Magaña-Maldonado
- Laboratorio Experimental de Enfermedades Neurodegenerativas, Laboratorio de Reprogramación Celular, Departamento de Neurofisiología, Instituto Nacional de Neurología y Neurocirugía, Ciudad de Mexico 14269, Mexico; (D.S.-A.); (N.S.-G.); (R.M.-M.)
| | - Dolores Jimenez-Farfan
- Laboratorio de Inmunología, División de Estudios de Posgrado e Investigación, Facultad de Odontología, Universidad Nacional Autónoma de México, Ciudad de Mexico 04510, Mexico;
| | - Elizabeth Ferreira-Guerrero
- Centro de Investigación Sobre Enfermedades Infecciosas, Instituto Nacional de Salud Pública, Cuernavaca 62100, Mexico; (E.F.-G.); (A.C.-S.)
| | - Arturo Cruz-Salgado
- Centro de Investigación Sobre Enfermedades Infecciosas, Instituto Nacional de Salud Pública, Cuernavaca 62100, Mexico; (E.F.-G.); (A.C.-S.)
| | | |
Collapse
|
10
|
Hamza A, Cho JY, Cap KC, Hossain AJ, Kim JG, Park JB. Extracellular pyruvate kinase M2 induces cell migration through p-Tyr42 RhoA-mediated superoxide generation and epithelial-mesenchymal transition. Free Radic Biol Med 2023; 208:614-629. [PMID: 37722568 DOI: 10.1016/j.freeradbiomed.2023.09.016] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/25/2023] [Revised: 09/12/2023] [Accepted: 09/15/2023] [Indexed: 09/20/2023]
Abstract
In the tumor microenvironment (TME), communication between cancer cells and tumor-associated macrophages (TAMs) through secreted extracellular proteins promotes cancer progression. Here, we observed that co-culturing cancer cells (4T1) and macrophage cells (Raw264.7) significantly enhanced superoxide production in both cell types. Using MALDI-TOF, we identified PKM2 as a highly secreted protein by Raw264.7 cells and bone marrow-derived monocytes. The extracellular recombinant PKM2 protein not only enhanced cancer cell migration and invasion but also increased superoxide production. Additionally, PKM2 was found to associate with the cell surface, and its binding to integrin α5/β1 receptor was inhibited by antibodies specifically targeting it. Furthermore, we investigated downstream signaling pathways involved in PKM2-induced superoxide production. We found that knock-down of RhoA and p47phox using siRNAs effectively abolished superoxide generation in response to extracellular PKM2. Notably, extracellular PKM2 triggered the phosphorylation of p47phox at Ser345 residue and RhoA at Tyr42 residue (p-Tyr42 RhoA). Moreover, extracellular PKM2 exerted regulatory control over the expression of key epithelial-mesenchymal transition (EMT) markers, including ZEB1, Snail1, vimentin, and E-cadherin. Interestingly, p-Tyr42 RhoA translocated to the nucleus, where it bound to the ZEB1 promoter region. In light of these findings, we propose that extracellular PKM2 within the TME plays a critical role in tumorigenesis by promoting cancer cell migration and invasion through RhoA/p47phox signaling pathway.
Collapse
Affiliation(s)
- Amir Hamza
- Department of Biochemistry, Hallym University College of Medicine, Chuncheon, Kangwon-do, 24252, Republic of Korea
| | - Jung Yoon Cho
- Institute of Cell Differentiation and Aging, Hallym University College of Medicine, Chuncheon, Kangwon-do, 24252, Republic of Korea
| | - Kim Cuong Cap
- Department of Biochemistry, Hallym University College of Medicine, Chuncheon, Kangwon-do, 24252, Republic of Korea; Institute of Cell Differentiation and Aging, Hallym University College of Medicine, Chuncheon, Kangwon-do, 24252, Republic of Korea
| | - Abu Jubayer Hossain
- Department of Biochemistry, Hallym University College of Medicine, Chuncheon, Kangwon-do, 24252, Republic of Korea; Institute of Cell Differentiation and Aging, Hallym University College of Medicine, Chuncheon, Kangwon-do, 24252, Republic of Korea
| | - Jae-Gyu Kim
- Department of Biochemistry, Hallym University College of Medicine, Chuncheon, Kangwon-do, 24252, Republic of Korea; Institute of Cell Differentiation and Aging, Hallym University College of Medicine, Chuncheon, Kangwon-do, 24252, Republic of Korea
| | - Jae-Bong Park
- Department of Biochemistry, Hallym University College of Medicine, Chuncheon, Kangwon-do, 24252, Republic of Korea; Institute of Cell Differentiation and Aging, Hallym University College of Medicine, Chuncheon, Kangwon-do, 24252, Republic of Korea.
| |
Collapse
|
11
|
Centola CL, Dasso ME, Soria JD, Riera MF, Meroni SB, Galardo MN. Glycolysis as key regulatory step in FSH-induced rat Sertoli cell proliferation: Role of the mTORC1 pathway. Biochimie 2023; 214:145-156. [PMID: 37442535 DOI: 10.1016/j.biochi.2023.07.007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2023] [Revised: 06/09/2023] [Accepted: 07/08/2023] [Indexed: 07/15/2023]
Abstract
The definitive number of Sertoli cells (SCs), achieved during the proliferative periods, defines the spermatogenic capacity in adulthood. It is recognized that FSH is the main mitogen targeting SC and that it exerts its action, at least partly, through the activation of the PI3K/Akt/mTORC1 pathway. mTORC1 controls a large number of cellular functions, including glycolysis and cell proliferation. Interestingly, recent evidence revealed that the glycolytic flux might modulate mTORC1 activity and, consequently, cell cycle progression. Although mature SC metabolism has been thoroughly studied, several aspects of metabolism regulation in proliferating SC are still to be elucidated. The objective of this study was to explore whether aerobic glycolysis is regulated by FSH through mTORC1 pathway in proliferating SC, and to assess the involvement of glycolysis in the regulation of SC proliferation. The present study was carried out utilizing 8-day-old rat SC cultures. The results obtained show that FSH enhances glycolytic flux through the induction of 6-phosphofructo-2-kinase/fructose-2,6-bisphosphatase 3 (PFKFB3) and lactate dehydrogenase A (LDHA) in an mTORC1 dependent manner. In addition, PFKFB3 and LDH inhibitors prevent FSH from activating mTORC1 and stimulating SC proliferation and glycolysis, presumably through mTORC1 pathway inhibition. In summary, FSH simultaneously regulates SC proliferation and glycolysis in an mTORC1 dependent manner, and glycolysis seems to cooperate with FSH in the stimulation of both cellular functions through the modulation of the same signalling pathway. Therefore, a positive feedback between the mTORC1 pathway and glycolysis triggered by FSH is hypothesized.
Collapse
Affiliation(s)
- Cecilia Lucia Centola
- Centro de Investigaciones Endocrinológicas "Dr. César Bergadá" (CEDIE) CONICET - FEI - División de Endocrinología, Hospital de Niños Ricardo Gutiérrez, Gallo 1330, C1425EFD, Ciudad Autónoma de Buenos Aires, Argentina
| | - Marina Ercilia Dasso
- Centro de Investigaciones Endocrinológicas "Dr. César Bergadá" (CEDIE) CONICET - FEI - División de Endocrinología, Hospital de Niños Ricardo Gutiérrez, Gallo 1330, C1425EFD, Ciudad Autónoma de Buenos Aires, Argentina
| | - Julio Daniel Soria
- Centro de Investigaciones Endocrinológicas "Dr. César Bergadá" (CEDIE) CONICET - FEI - División de Endocrinología, Hospital de Niños Ricardo Gutiérrez, Gallo 1330, C1425EFD, Ciudad Autónoma de Buenos Aires, Argentina
| | - Maria Fernanda Riera
- Centro de Investigaciones Endocrinológicas "Dr. César Bergadá" (CEDIE) CONICET - FEI - División de Endocrinología, Hospital de Niños Ricardo Gutiérrez, Gallo 1330, C1425EFD, Ciudad Autónoma de Buenos Aires, Argentina
| | - Silvina Beatriz Meroni
- Centro de Investigaciones Endocrinológicas "Dr. César Bergadá" (CEDIE) CONICET - FEI - División de Endocrinología, Hospital de Niños Ricardo Gutiérrez, Gallo 1330, C1425EFD, Ciudad Autónoma de Buenos Aires, Argentina
| | - Maria Noel Galardo
- Centro de Investigaciones Endocrinológicas "Dr. César Bergadá" (CEDIE) CONICET - FEI - División de Endocrinología, Hospital de Niños Ricardo Gutiérrez, Gallo 1330, C1425EFD, Ciudad Autónoma de Buenos Aires, Argentina.
| |
Collapse
|
12
|
Zhang Y, Barberis M. Exploring cell cycle-mediated regulations of glycolysis in budding yeast. Front Microbiol 2023; 14:1270487. [PMID: 37886071 PMCID: PMC10598772 DOI: 10.3389/fmicb.2023.1270487] [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: 07/31/2023] [Accepted: 09/25/2023] [Indexed: 10/28/2023] Open
Abstract
Coordination of cell cycle with metabolism exists in all cell types that grow by division. It serves to build a new cell, (i) fueling building blocks for the synthesis of proteins, nucleic acids, and membranes, and (ii) producing energy through glycolysis. Cyclin-dependent kinases (Cdks) play an essential role in this coordination, thereby in the regulation of cell division. Cdks are functional homologs across eukaryotes and are the engines that drive cell cycle events and the clocks that time them. Their function is counteracted by stoichiometric inhibitors; specifically, inhibitors of cyclin-cyclin dependent kinase (cyclin/Cdk) complexes allow for their activity at specific times. Here, we provide a new perspective about the yet unknown cell cycle mechanisms impacting on metabolism. We first investigated the effect of the mitotic cyclin/Cdk1 complex Cyclin B/Cdk1-functional homolog in mammalian cells of the budding yeast Clb2/Cdk1-on yeast metabolic enzymes of, or related to, the glycolysis pathway. Six glycolytic enzymes (Glk1, Hxk2, Pgi1, Fba1, Tdh1, and Pgk1) were subjected to in vitro Cdk-mediated phosphorylation assays. Glucose-6-phosphate dehydrogenase (Zwf1), the first enzyme in the pentose phosphate pathway that is important for NADPH production, and 6-phospho-fructo-2-kinase (Pfk27), which catalyzes fructose-2,6-bisphosphate synthesis, a key regulator of glycolysis, were also included in the study. We found that, among these metabolic enzymes, Fba1 and Pgk1 may be phosphorylated by Cdk1, in addition to the known Cdk1-mediated phosphorylation of Gph1. We then investigated the possible effect of Sic1, stoichiometric inhibitor of mitotic cyclin/Cdk1 complexes in budding yeast, on the activities of three most relevant glycolytic enzymes: Hxk2, Glk1, and Tdh1. We found that Sic1 may have a negative effect on Hxk2. Altogether, we reveal possible new routes, to be further explored, through which cell cycle may regulate cellular metabolism. Because of the functional homology of cyclin/Cdk complexes and their stoichiometric inhibitors across evolution, our findings may be relevant for the regulation of cell division in eukaryotes.
Collapse
Affiliation(s)
- Yanfei Zhang
- Molecular Systems Biology, School of Biosciences, Faculty of Health and Medical Sciences, University of Surrey, Guildford, United Kingdom
- Synthetic Systems Biology and Nuclear Organization, Swammerdam Institute for Life Sciences, University of Amsterdam, Amsterdam, Netherlands
| | - Matteo Barberis
- Molecular Systems Biology, School of Biosciences, Faculty of Health and Medical Sciences, University of Surrey, Guildford, United Kingdom
- Synthetic Systems Biology and Nuclear Organization, Swammerdam Institute for Life Sciences, University of Amsterdam, Amsterdam, Netherlands
- Centre for Mathematical and Computational Biology, CMCB, University of Surrey, Guildford, United Kingdom
| |
Collapse
|
13
|
Polónia B, Xavier CPR, Kopecka J, Riganti C, Vasconcelos MH. The role of Extracellular Vesicles in glycolytic and lipid metabolic reprogramming of cancer cells: Consequences for drug resistance. Cytokine Growth Factor Rev 2023; 73:150-162. [PMID: 37225643 DOI: 10.1016/j.cytogfr.2023.05.001] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2023] [Revised: 05/10/2023] [Accepted: 05/12/2023] [Indexed: 05/26/2023]
Abstract
In order to adapt to a higher proliferative rate and an increased demand for energy sources, cancer cells rewire their metabolic pathways, a process currently recognized as a hallmark of cancer. Even though the metabolism of glucose is perhaps the most discussed metabolic shift in cancer, lipid metabolic alterations have been recently recognized as relevant players in the growth and proliferation of cancer cells. Importantly, some of these metabolic alterations are reported to induce a drug resistant phenotype in cancer cells. The acquisition of drug resistance traits severely hinders cancer treatment, being currently considered one of the major challenges of the oncological field. Evidence suggests that Extracellular Vesicles (EVs), which play a crucial role in intercellular communication, may act as facilitators of tumour progression, survival and drug resistance by modulating several aspects involved in the metabolism of cancer cells. This review aims to gather and discuss relevant data regarding metabolic reprograming in cancer, particularly involving the glycolytic and lipid alterations, focusing on its influence on drug resistance and highlighting the relevance of EVs as intercellular mediators of this process.
Collapse
Affiliation(s)
- Bárbara Polónia
- i3S - Instituto de Investigação e Inovação em Saúde, Universidade do Porto, 4200-135 Porto, Portugal; Cancer Drug Resistance Group, IPATIMUP - Institute of Molecular Pathology and Immunology, University of Porto, Portugal, 4200-135 Porto, Portugal; Department of Biological Sciences, FFUP - Faculty of Pharmacy of the University of Porto, Porto, Portugal
| | - Cristina P R Xavier
- i3S - Instituto de Investigação e Inovação em Saúde, Universidade do Porto, 4200-135 Porto, Portugal; Cancer Drug Resistance Group, IPATIMUP - Institute of Molecular Pathology and Immunology, University of Porto, Portugal, 4200-135 Porto, Portugal
| | - Joanna Kopecka
- Department of Oncology, University of Torino, 10126 Torino, Italy
| | - Chiara Riganti
- Department of Oncology, University of Torino, 10126 Torino, Italy; Interdepartmental Research Center for Molecular Biotechnology "G. Tarone", University of Torino, 10126 Torino, Italy
| | - M Helena Vasconcelos
- i3S - Instituto de Investigação e Inovação em Saúde, Universidade do Porto, 4200-135 Porto, Portugal; Cancer Drug Resistance Group, IPATIMUP - Institute of Molecular Pathology and Immunology, University of Porto, Portugal, 4200-135 Porto, Portugal; Department of Biological Sciences, FFUP - Faculty of Pharmacy of the University of Porto, Porto, Portugal.
| |
Collapse
|
14
|
Arietta-García AG, Calzada F, Ramírez-Sánchez I, Bautista E, García-Hernandez N, Ordoñez-Razo RM. Advances in the Properties of Incomptine A: Cytotoxic Activity and Downregulation of Hexokinase II in Breast Cancer Cell Lines. Int J Mol Sci 2023; 24:12406. [PMID: 37569780 PMCID: PMC10419048 DOI: 10.3390/ijms241512406] [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: 06/30/2023] [Revised: 07/25/2023] [Accepted: 07/28/2023] [Indexed: 08/13/2023] Open
Abstract
Breast cancer treatments are limited by the cancer subtype and its selectivity towards tumor cells, hence the importance of finding compounds that increase the survival of healthy cells and target any subtype. Incomptine A (IA) is a sesquiterpene lactone with demonstrated cytotoxic activity. In this study, through in vitro assays, it was observed that IA has similar cytotoxic activity between the subtypes triple negative, HER2+, and luminal A of the breast cancer cell lines. IA cytotoxic activity is higher in cancer than in nontumorigenic cells, and its selectivity index for cancer cells is more than that of the drug doxorubicin. Molecular docking and its in silico comparison with the 2-Deoxyglucose inhibitor suggest that IA could bind to Hexokinase II (HKII), decreasing its expression. Since we did not find changes in the expression of the glycolytic pathway, we suppose that IA could affect the antiapoptotic function of HKII in cancer cells. The IA-HKII union would activate the voltage-gated anion channel 1 (VDAC1), resuming apoptosis. Therefore, we suggest that IA could be used against almost any subtype and that its cytotoxic effect could be due to the reactivation of apoptosis in breast cancer cells.
Collapse
Affiliation(s)
- Angel Giovanni Arietta-García
- Unidad de Investigación Médica en Genética Humana, UMAE Hospital de Pediatría 2° Piso, Centro Médico Nacional Siglo XXI, Instituto Mexicano del Seguro Social, Av. Cuauhtémoc 330, Col. Doctores, Mexico City CP 06725, Mexico; (A.G.A.-G.); (N.G.-H.)
| | - Fernando Calzada
- Unidad de Investigación Médica en Farmacología, UMAE Hospital de Especialidades, 2° Piso CORSE, Centro Médico Nacional Siglo XXI, Instituto Mexicano del Seguro Social, Av. Cuauhtémoc 330, Col. Doctores, Mexico City CP 06725, Mexico;
| | - Israel Ramírez-Sánchez
- Sección de Estudios de Posgrado e Investigación, Escuela Superior de Medicina, Instituto Politécnico Nacional, Mexico City CP 07738, Mexico;
| | - Elihú Bautista
- CONAHCYT—Consorcio de Investigación, Innovación y Desarrollo para las Zonas Áridas, Instituto Potosino de Investigación Científica y Tecnológica A.C., San Luis Potosí CP 78216, Mexico;
| | - Normand García-Hernandez
- Unidad de Investigación Médica en Genética Humana, UMAE Hospital de Pediatría 2° Piso, Centro Médico Nacional Siglo XXI, Instituto Mexicano del Seguro Social, Av. Cuauhtémoc 330, Col. Doctores, Mexico City CP 06725, Mexico; (A.G.A.-G.); (N.G.-H.)
| | - Rosa María Ordoñez-Razo
- Unidad de Investigación Médica en Genética Humana, UMAE Hospital de Pediatría 2° Piso, Centro Médico Nacional Siglo XXI, Instituto Mexicano del Seguro Social, Av. Cuauhtémoc 330, Col. Doctores, Mexico City CP 06725, Mexico; (A.G.A.-G.); (N.G.-H.)
| |
Collapse
|
15
|
Xu S, Lin J, Chen R, Xie J, Yuan E, Cen F, Kong F. LINC00174 Promotes Colon Cancer Progression by Regulating Inflammation and Glycolysis by Targeting the MicroRNA-2467-3p/Enolase 3 Axis. Mediators Inflamm 2023; 2023:8052579. [PMID: 37448887 PMCID: PMC10338131 DOI: 10.1155/2023/8052579] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2023] [Revised: 05/03/2023] [Accepted: 05/23/2023] [Indexed: 07/15/2023] Open
Abstract
OBJECTIVE To elucidate the mechanism by which LINC00174 promotes colon cancer progression by targeting the microRNA-2467-3p (miR-2467-3p)/enolase 3 (ENO3) axis to regulate inflammation and glycolysis. METHODS The expression of LINC00174 and ENO3 in colon cancer tissues, its relationship with survival rate, and correlation were analyzed using bioinformatic analysis. The effects of LINC00174 overexpression and silencing on the biological behavior of and inflammation in colon cancer cells were analyzed via transfection experiments. The target relationships between miR-2467-3p or LINC00174 and ENO3 were verified using sequence prediction and the dual-luciferase reporter assay, respectively. Furthermore, LINC00174- and/or miR-2467-3p-overexpressing cells were prepared to determine the effects on ENO3 protein levels and glycolysis. Finally, the effects of LINC00174 and/or miR-2467-3p overexpression on colon cancer, ENO3 protein levels, and inflammation were analyzed using a tumor-bearing mice model. RESULTS LINC00174 and ENO3 were overexpressed and associated with a lower survival rate. LINC00174 was positively correlated with ENO3 in colon cancer tissues. Furthermore, the overexpression of LINC00174 in colon cancer cell lines promoted the proliferation, migration, and invasion of colon cancer cells and inflammation but inhibited apoptosis. The overexpression of miR-2467-3p inhibited ENO3 protein levels, which was attenuated via LINC00174 overexpression. Furthermore, it inhibited the biological behavior of and inflammation and glycolysis in colon cancer cells and blocked their LINC00174-induced promotion. Moreover, using animal experiments, the regulatory effects of LINC00174 on tumor growth, ENO3 protein levels, and inflammation via miR-2467-3p were confirmed. CONCLUSION LINC00174 promotes the glycolysis, inflammation, proliferation, migration, and invasion of colon cancer cells and inhibits apoptosis. The cancer-promoting mechanism of LINC00174 is related to targeting miR-2467-3p to promote ENO3 protein levels.
Collapse
Affiliation(s)
- Sheng Xu
- Department of General Surgery, People's Hospital of Guangxi Zhuang Autonomous Region, 530021 Nanning, China
| | - Jiawei Lin
- Department of General Surgery, People's Hospital of Guangxi Zhuang Autonomous Region, 530021 Nanning, China
| | - Rong Chen
- Department of General Surgery, People's Hospital of Guangxi Zhuang Autonomous Region, 530021 Nanning, China
| | - Junjie Xie
- Oncology Department, General Hospital of Central Theater Command, Wuluo 627, Wuhan, 430070 Hubei Province, China
| | - Enquan Yuan
- Department of General Surgery, People's Hospital of Guangxi Zhuang Autonomous Region, 530021 Nanning, China
| | - Fajie Cen
- Department of General Surgery, People's Hospital of Guangxi Zhuang Autonomous Region, 530021 Nanning, China
| | - Fanbiao Kong
- Department of General Surgery, People's Hospital of Guangxi Zhuang Autonomous Region, 530021 Nanning, China
| |
Collapse
|
16
|
Shegay PV, Shatova OP, Zabolotneva AA, Shestopalov AV, Kaprin AD. Moonlight functions of glycolytic enzymes in cancer. Front Mol Biosci 2023; 10:1076138. [PMID: 37449059 PMCID: PMC10337784 DOI: 10.3389/fmolb.2023.1076138] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2022] [Accepted: 06/19/2023] [Indexed: 07/18/2023] Open
Abstract
Since an extensive genome research has started, basic principle "one gene-one protein-one function" was significantly revised. Many proteins with more than one function were identified and characterized as "moonlighting" proteins, which activity depend not only on structural peculiarities but also on compartmentation and metabolic environment. It turned out that "housekeeping" glycolytic enzymes show important moonlight functions such as control of development, proliferation, apoptosis, migration, regulation of transcription and cell signaling. Glycolytic enzymes emerged very early in evolution and because of the limited content of genomes, they could be used as ancient regulators for intercellular and intracellular communication. The multifunctionality of the constitutively expressed enzymes began to serve cancer cell survival and growth. In the present review we discuss some moonlight functions of glycolytic enzymes that important for malignant transformation and tumor growth.
Collapse
Affiliation(s)
- Petr V. Shegay
- Federal State Budget Institution, National Medical Research Radiology Center of the Ministry of Healthcare of the Russian Federation, Moscow, Russia
| | - Olga P. Shatova
- Department of Biochemistry and Molecular Biology, Faculty of Medicine, Pirogov Russian National Research Medical University, Moscow, Russia
- Biochemistry Department, Peoples’ Friendship University of Russia, Moscow, Russia
| | - Anastasia A. Zabolotneva
- Department of Biochemistry and Molecular Biology, Faculty of Medicine, Pirogov Russian National Research Medical University, Moscow, Russia
- National Medical Research Centre for Endocrinology, Laboratory of Biochemistry of Signaling Pathways, Moscow, Russia
| | - Aleksandr V. Shestopalov
- Department of Biochemistry and Molecular Biology, Faculty of Medicine, Pirogov Russian National Research Medical University, Moscow, Russia
- National Medical Research Centre for Endocrinology, Laboratory of Biochemistry of Signaling Pathways, Moscow, Russia
- Dmitry Rogachev National Medical Research Center of Pediatric Hematology, Oncology and Immunology, Ministry of Health of the Russian Federation, Moscow, Russia
| | - Andrei D. Kaprin
- Federal State Budget Institution, National Medical Research Radiology Center of the Ministry of Healthcare of the Russian Federation, Moscow, Russia
- Biochemistry Department, Peoples’ Friendship University of Russia, Moscow, Russia
| |
Collapse
|
17
|
Hong Z, Lu Y, Liu B, Ran C, Lei X, Wang M, Wu S, Yang Y, Wu H. Glycolysis, a new mechanism of oleuropein against liver tumor. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2023; 114:154770. [PMID: 36963367 DOI: 10.1016/j.phymed.2023.154770] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/17/2022] [Revised: 01/10/2023] [Accepted: 03/13/2023] [Indexed: 06/18/2023]
Abstract
BACKGROUND Benign and malignant liver tumors are prevalent worldwide. However, there is no effective and comprehensive treatment option for many patients with malignant tumors. Thus, it is critical to prevent benign tumors from worsening, increasing the number of treatment options and effective medications against malignant liver tumors. Oleuropein is a natural and non-toxic product and inhibits tumor growth in various ways. METHODS We employed bioinformatics analysis and molecular docking to identify potential targets of oleuropein. Surface plasmon resonance (SPR) was used to determine the direct binding strength of the target and compounds. Essential functionalities of the targets were analyzed using gene interference approaches. Transcriptomic studies were performed to observe the global genomic alterations occurring inside cells. Changes in glycolytic metabolites and gene and protein expressions were also detected. The anti-tumor benefits of oleuropein in vivo were determined using a tumor-bearing mouse model. RESULTS Glucose-6-phosphate isomerase (GPI) was found to be a direct target of oleuropein. GPI discontinuation in liver tumor cells altered the expression of many genes, causing glycogenolysis. GPI interference was associated with PYGM and PFKFB4 inhibitors to inhibit glycolysis in liver tumors. Oleuropein inhibited glycolysis and showed good anti-tumor activity in vivo without adverse side effects. CONCLUSIONS GPI is a crucial enzyme in glycolysis and the immediate target of oleuropein. GPI expression inside tumor cells affects different physiological functions and signal transduction. Oleuropein has depicted anti-tumor action in vivo without harmful side effects. Moreover, it can control tumor glycolysis through GPI.
Collapse
Affiliation(s)
- Zongchao Hong
- Faculty of Pharmacy, Hubei University of Chinese Medicine, No. 16, Huangjiahu West Road, Hongshan District, Wuhan, Hubei 430065, China
| | - Yi Lu
- Faculty of Pharmacy, Hubei University of Chinese Medicine, No. 16, Huangjiahu West Road, Hongshan District, Wuhan, Hubei 430065, China
| | - Bo Liu
- Faculty of Pharmacy, Hubei University of Chinese Medicine, No. 16, Huangjiahu West Road, Hongshan District, Wuhan, Hubei 430065, China
| | - Chongwang Ran
- Faculty of Pharmacy, Hubei University of Chinese Medicine, No. 16, Huangjiahu West Road, Hongshan District, Wuhan, Hubei 430065, China
| | - Xia Lei
- Faculty of Pharmacy, Hubei University of Chinese Medicine, No. 16, Huangjiahu West Road, Hongshan District, Wuhan, Hubei 430065, China
| | - Mengfan Wang
- Faculty of Pharmacy, Hubei University of Chinese Medicine, No. 16, Huangjiahu West Road, Hongshan District, Wuhan, Hubei 430065, China
| | - Songtao Wu
- Faculty of Pharmacy, Hubei University of Chinese Medicine, No. 16, Huangjiahu West Road, Hongshan District, Wuhan, Hubei 430065, China.
| | - Yanfang Yang
- Faculty of Pharmacy, Hubei University of Chinese Medicine, No. 16, Huangjiahu West Road, Hongshan District, Wuhan, Hubei 430065, China; Key Laboratory of Traditional Chinese Medicine Resources and Chemistry of Hubei Province, Wuhan, China.
| | - Hezhen Wu
- Faculty of Pharmacy, Hubei University of Chinese Medicine, No. 16, Huangjiahu West Road, Hongshan District, Wuhan, Hubei 430065, China; Key Laboratory of Traditional Chinese Medicine Resources and Chemistry of Hubei Province, Wuhan, China; Modern Engineering Research Center of Traditional Chinese Medicine and Ethnic Medicine of Hubei Province, Wuhan, 430065, China.
| |
Collapse
|
18
|
Duan X, Chen Y, Zhang K, Chen W, Zhao J, Dai X, Cao W, Dong Z, Mo S, Lu J. PHGDH promotes esophageal squamous cell carcinoma progression via Wnt/β-catenin pathway. Cell Signal 2023:110736. [PMID: 37263462 DOI: 10.1016/j.cellsig.2023.110736] [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: 02/23/2023] [Revised: 05/05/2023] [Accepted: 05/26/2023] [Indexed: 06/03/2023]
Abstract
PURPOSE Esophageal squamous carcinoma (ESCC) with a high incidence in China, lacks effective therapeutic targets. Phosphoglycerate dehydrogenase (PHGDH) is a key enzyme in serine biosynthesis. However, the biological role of PHGDH in ESCC has not been revealed. METHODS The expression of PHGDH in ESCC was investigated by UALCAN. The relationship between PHGDH expression and its prognostic value was analyzed by Kaplan-Meier and univariate Cox regression. Further, the potential functions of PHGDH involved in ESCC were explored through DAVID database and GSEA software. In addition, the expression of PHGDH was verified in ESCC. Then, the effects of PHGDH knockdown on ESCC were evaluated in vitro and in vivo by cell proliferation, clone formation, cell cycle, apoptosis, tube formation assays and ESCC cells derived xenograft model. In addition, western blotting and immunohistochemistry were used to detect the expression of Wnt/β-catenin pathway which was associated with PHGDH. RESULTS Bioinformatics analysis found that PHGDH was highly expressed in ESCC, and meaningfully, patients with high PHGDH expression had a poor prognosis. Moreover, the overexpression of PHGDH was verified in ESCC. Afterwards, PHGDH knockdown inhibited the cell proliferation, induced cell cycle arrest and apoptosis in ESCC cells, and inhibited the angiogenesis of HUVECs induced by ESCC conditioned medium, as well as inhibited the growth of xenograft tumor. Mechanistically, PHGDH knockdown inhibited Wnt/β-catenin signaling pathway in ESCC. CONCLUSION High expression of PHGDH predicts a poor prognosis for ESCC. PHGDH knockdown inhibits ESCC progression by suppressing Wnt/β-catenin signaling pathway, indicating that PHGDH might be a potential target for ESCC therapy.
Collapse
Affiliation(s)
- Xiaoxuan Duan
- Department of Pathophysiology, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, Henan Province 450001, PR China
| | - Yihuan Chen
- Department of Pathophysiology, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, Henan Province 450001, PR China
| | - Kai Zhang
- Department of Pathophysiology, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, Henan Province 450001, PR China
| | - Wei Chen
- Department of Pathophysiology, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, Henan Province 450001, PR China
| | - Jun Zhao
- Department of Oncology, Changzhi People's Hospital, Changzhi, Shanxi 046000, PR China
| | - Xiaoshuo Dai
- Department of Pathophysiology, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, Henan Province 450001, PR China
| | - Wenbo Cao
- Department of Pathophysiology, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, Henan Province 450001, PR China; Collaborative Innovation Center of Henan Province for Cancer Chemoprevention, Zhengzhou University, Zhengzhou, Henan Province 450001, PR China; State Key Laboratory of Esophageal Cancer Prevention & Treatment, Zhengzhou University, Zhengzhou, Henan Province 450052, PR China
| | - Ziming Dong
- Department of Pathophysiology, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, Henan Province 450001, PR China; Collaborative Innovation Center of Henan Province for Cancer Chemoprevention, Zhengzhou University, Zhengzhou, Henan Province 450001, PR China; State Key Laboratory of Esophageal Cancer Prevention & Treatment, Zhengzhou University, Zhengzhou, Henan Province 450052, PR China
| | - Saijun Mo
- Department of Pathophysiology, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, Henan Province 450001, PR China; Collaborative Innovation Center of Henan Province for Cancer Chemoprevention, Zhengzhou University, Zhengzhou, Henan Province 450001, PR China; State Key Laboratory of Esophageal Cancer Prevention & Treatment, Zhengzhou University, Zhengzhou, Henan Province 450052, PR China.
| | - Jing Lu
- Department of Pathophysiology, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, Henan Province 450001, PR China; Collaborative Innovation Center of Henan Province for Cancer Chemoprevention, Zhengzhou University, Zhengzhou, Henan Province 450001, PR China; State Key Laboratory of Esophageal Cancer Prevention & Treatment, Zhengzhou University, Zhengzhou, Henan Province 450052, PR China.
| |
Collapse
|
19
|
Sawant Dessai A, Kalhotra P, Novickis AT, Dasgupta S. Regulation of tumor metabolism by post translational modifications on metabolic enzymes. Cancer Gene Ther 2023; 30:548-558. [PMID: 35999357 PMCID: PMC9947196 DOI: 10.1038/s41417-022-00521-x] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2022] [Revised: 07/05/2022] [Accepted: 08/04/2022] [Indexed: 11/09/2022]
Abstract
Metabolic reprogramming is a hallmark of cancer development, progression, and metastasis. Several metabolic pathways such as glycolysis, tricarboxylic acid (TCA) cycle, lipid metabolism, and glutamine catabolism are frequently altered to support cancer growth. Importantly, the activity of the rate-limiting metabolic enzymes in these pathways are specifically modulated in cancer cells. This is achieved by transcriptional, translational, and post translational regulations that enhance the expression, activity, stability, and substrate sensitivity of the rate-limiting enzymes. These mechanisms allow the enzymes to retain increased activity supporting the metabolic needs of rapidly growing tumors, sustain their survival in the hostile tumor microenvironments and in the metastatic lesions. In this review, we primarily focused on the post translational modifications of the rate-limiting enzymes in the glucose and glutamine metabolism, TCA cycle, and fatty acid metabolism promoting tumor progression and metastasis.
Collapse
Affiliation(s)
- Abhisha Sawant Dessai
- Department of Cell Stress Biology, Roswell Park Comprehensive Cancer Center, Buffalo, NY, 14263, USA
| | - Poonam Kalhotra
- Department of Cell Stress Biology, Roswell Park Comprehensive Cancer Center, Buffalo, NY, 14263, USA
| | - Aaron T Novickis
- Department of Cell Stress Biology, Roswell Park Comprehensive Cancer Center, Buffalo, NY, 14263, USA
| | - Subhamoy Dasgupta
- Department of Cell Stress Biology, Roswell Park Comprehensive Cancer Center, Buffalo, NY, 14263, USA.
| |
Collapse
|
20
|
Li L, Liu C, Qin Y, Gao F, Wang Q, Zhu Y. Identification of cancer protein biomarker based on cell specific peptide and its potential role in predicting tumor metastasis. J Proteomics 2023; 275:104826. [PMID: 36708809 DOI: 10.1016/j.jprot.2023.104826] [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: 02/18/2022] [Revised: 11/19/2022] [Accepted: 01/13/2023] [Indexed: 01/28/2023]
Abstract
The identification of tumor related membrane protein is important for both cancer diagnosis and targeted therapy. Currently, the number of ideal clinical biomarkers is still limited partially because of lacking efficient methods in biomarker discovery. Targeting peptides are generated by library screening and can recognize their cognate targets with high specificity and affinity. In addition, these functional peptides have been considered to be a valuable molecule for both imaging detection and targeting therapy in oncology. The selected peptides can be used to identify cell-surface protein biomarkers of cancer cells. In our study, the peptide (VECYLIRDNLCIY) derived from the bacteria displaying library worked as a bait to capture its binding partner and aldolase A was identified as the candidate. The results indicated that aldolase A' expression level on the cell membrane was regulated by PI3K and aldolase A located on the membrane could inhibit the aggression of tumors through mediating cell metabolic pathway. Aldolase A could work as the joint for the metabolic and signal pathways related to tumor progression. In our work, we demonstrated a promising technology for selecting and identifying binding partners for cell-specific peptides that enables discovery of new tumor biomarkers, showing great scientific study values and clinical translation potencies. SIGNIFICANCE: MS-based cancer biomarker discovery provides promising target candidates for cancer diagnosis and therapy. However, the inevitable limits make it inconvenient in the process of sample preparation and data analysis. In this way, the small molecular probes show some advantages due to their readily availability and specific binding affinity such as the aptamers screened with SELEX technology and peptides derived from displaying libraries. In the present study, aldolase A was proved to be the membrane binding partner of a specific peptidic ligand towards ZR-75-1 tumor cell. It was discovered that membrane aldolase A was more sensitive and observable than other subcellular fractions in response to cellular metabolic state alteration or glucose availability. In addition, the reduced membrane-localized aldolase A expression indicated a more aggressive tumor phenotype and was accompanied by the upregulation of MMP-2/MMP-9. The non-glycolysis activity endowed it with potential utility as a tumor diagnostic marker and therapeutic target. This work demonstrates the practicability of screened peptide in cancer biomarker discovery, facilitating the development of diagnostic tools and therapeutic approaches to cancer, and markedly improves our understanding of cancer biology.
Collapse
Affiliation(s)
- Lin Li
- Key Laboratory of Nano-Bio Interface Research, Division of Nano biomedicine, Suzhou Institute of Nano-Tech and Nano-Bionics, Chinese Academy of Sciences, Suzhou 215123, China
| | - Cuijuan Liu
- Key Laboratory of Nano-Bio Interface Research, Division of Nano biomedicine, Suzhou Institute of Nano-Tech and Nano-Bionics, Chinese Academy of Sciences, Suzhou 215123, China; School of Nano-Tech and Nano-Bionics, University of Science and Technology of China, Hefei 230026, China
| | - Yingzhou Qin
- Key Laboratory of Nano-Bio Interface Research, Division of Nano biomedicine, Suzhou Institute of Nano-Tech and Nano-Bionics, Chinese Academy of Sciences, Suzhou 215123, China; School of Nano-Tech and Nano-Bionics, University of Science and Technology of China, Hefei 230026, China
| | - Fan Gao
- Key Laboratory of Nano-Bio Interface Research, Division of Nano biomedicine, Suzhou Institute of Nano-Tech and Nano-Bionics, Chinese Academy of Sciences, Suzhou 215123, China; School of Nano-Tech and Nano-Bionics, University of Science and Technology of China, Hefei 230026, China
| | - Qianqian Wang
- Key Laboratory of Nano-Bio Interface Research, Division of Nano biomedicine, Suzhou Institute of Nano-Tech and Nano-Bionics, Chinese Academy of Sciences, Suzhou 215123, China; School of Nano-Tech and Nano-Bionics, University of Science and Technology of China, Hefei 230026, China
| | - Yimin Zhu
- Key Laboratory of Nano-Bio Interface Research, Division of Nano biomedicine, Suzhou Institute of Nano-Tech and Nano-Bionics, Chinese Academy of Sciences, Suzhou 215123, China; School of Nano-Tech and Nano-Bionics, University of Science and Technology of China, Hefei 230026, China.
| |
Collapse
|
21
|
Liu Y, Feng Z, Zhang P, Chen H, Zhu S, Wang X. Advances in the study of aerobic glycolytic effects in resistance to radiotherapy in malignant tumors. PeerJ 2023; 11:e14930. [PMID: 36811010 PMCID: PMC9939019 DOI: 10.7717/peerj.14930] [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: 09/22/2022] [Accepted: 01/30/2023] [Indexed: 02/18/2023] Open
Abstract
Aerobic glycolysis is a metabolic mode of tumor cells different from normal cells that plays an important role in tumor proliferation and distant metastasis. Radiotherapy has now become a routine and effective treatment for many malignancies, however, resistance to radiotherapy remains a major challenge in the treatment of malignant tumors. Recent studies have found that the abnormal activity of the aerobic glycolysis process in tumor cells is most likely involved in regulating chemoresistance and radiation therapy resistance in malignant tumors. However, research on the functions and mechanisms of aerobic glycolysis in the molecular mechanisms of resistance to radiotherapy in malignant tumors is still in its early stages. This review collects recent studies on the effects of aerobic glycolysis and radiation therapy resistance in malignant tumors, to further understand the progress in this area. This research may more effectively guide the clinical development of more powerful treatment plans for radiation therapy resistant subtypes of cancer patients, and take an important step to improve the disease control rate of radiation therapy resistant subtypes of cancer patients.
Collapse
|
22
|
Gjorgjieva M, Ay AS, Correia de Sousa M, Delangre E, Dolicka D, Sobolewski C, Maeder C, Fournier M, Sempoux C, Foti M. MiR-22 Deficiency Fosters Hepatocellular Carcinoma Development in Fatty Liver. Cells 2022; 11:cells11182860. [PMID: 36139435 PMCID: PMC9496902 DOI: 10.3390/cells11182860] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2022] [Revised: 09/02/2022] [Accepted: 09/09/2022] [Indexed: 12/24/2022] Open
Abstract
MiR-22 is mostly considered as a hepatic tumor-suppressor microRNA based on in vitro analyses. Yet, whether miR-22 exerts a tumor-suppressive function in the liver has not been investigated in vivo. Herein, in silico analyses of miR-22 expression were performed in hepatocellular carcinomas from human patient cohorts and different mouse models. Diethylnitrosamine-induced hepatocellular carcinomas were then investigated in lean and diet-induced obese miR-22-deficient mice. The proteome of liver tissues from miR-22-deficient mice prior to hepatocellular carcinoma development was further analyzed to uncover miR-22 regulated factors that impact hepatocarcinogenesis with miR-22 deficiency. MiR-22 downregulation was consistently observed in hepatocellular carcinomas from all human cohorts and mouse models investigated. The time of appearance of the first tumors was decreased and the number of tumoral foci induced by diethylnitrosamine was significantly increased by miR-22-deficiency in vivo, two features which were further drastically exacerbated with diet-induced obesity. At the molecular level, we provide evidence that the loss of miR-22 significantly affects the energetic metabolism and mitochondrial functions of hepatocytes, and the expression of tumor-promoting factors such as thrombospondin-1. Our study demonstrates that miR-22 acts as a hepatic tumor suppressor in vivo by restraining pro-carcinogenic metabolic deregulations through pleiotropic mechanisms and the overexpression of relevant oncogenes.
Collapse
Affiliation(s)
- Monika Gjorgjieva
- Department of Cell Physiology and Metabolism, Faculty of Medicine, University of Geneva, CH-1211 Geneva, Switzerland
| | - Anne-Sophie Ay
- Department of Cell Physiology and Metabolism, Faculty of Medicine, University of Geneva, CH-1211 Geneva, Switzerland
| | - Marta Correia de Sousa
- Department of Cell Physiology and Metabolism, Faculty of Medicine, University of Geneva, CH-1211 Geneva, Switzerland
| | - Etienne Delangre
- Department of Cell Physiology and Metabolism, Faculty of Medicine, University of Geneva, CH-1211 Geneva, Switzerland
| | - Dobrochna Dolicka
- Department of Cell Physiology and Metabolism, Faculty of Medicine, University of Geneva, CH-1211 Geneva, Switzerland
| | - Cyril Sobolewski
- Department of Cell Physiology and Metabolism, Faculty of Medicine, University of Geneva, CH-1211 Geneva, Switzerland
| | - Christine Maeder
- Department of Cell Physiology and Metabolism, Faculty of Medicine, University of Geneva, CH-1211 Geneva, Switzerland
| | - Margot Fournier
- Department of Cell Physiology and Metabolism, Faculty of Medicine, University of Geneva, CH-1211 Geneva, Switzerland
| | - Christine Sempoux
- Service of Clinical Pathology, Institute of Pathology, Lausanne University Hospital, University of Lausanne, CH-1011 Lausanne, Switzerland
| | - Michelangelo Foti
- Department of Cell Physiology and Metabolism, Faculty of Medicine, University of Geneva, CH-1211 Geneva, Switzerland
- Translational Research Centre in Onco-Haematology, Faculty of Medicine, University of Geneva, CH-1211 Geneva, Switzerland
- Correspondence:
| |
Collapse
|
23
|
Konieczka P, Żelechowska E, Przybylski W, Jaworska D, Sałek P, Kinsner M, Jankowski J. The Sarcoplasmic Protein Profile of Breast Muscle in Turkeys in Response to Different Dietary Ratios of Limiting Amino Acids and Clostridium perfringens-Induced Inflammation. Poult Sci 2022; 101:102195. [PMID: 36257075 PMCID: PMC9574763 DOI: 10.1016/j.psj.2022.102195] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2022] [Revised: 09/15/2022] [Accepted: 09/16/2022] [Indexed: 12/23/2022] Open
Abstract
In this study, the effects of the Arginine/Lysine (Arg/Lys) ratio in low- and high-methionine (Met) diets on the sarcoplasmic protein profile of breast muscles from turkeys reared under optimal or challenge (Clostridium perfringens infection) conditions were determined. One-day-old Hybrid Converter female turkey poults (216 in total) obtained from a commercial hatchery on hatching day, and on the basis of their average initial body weight were randomly allocated to 12 pens (4 m2 each; 2.0 m × 2.0 m) containing litter bedding and were reared over a 42-day experimental period. Diets with high levels of Lys contained approximately 1.80% and 1.65% Lys and were offered in two successive feeding periods (days 1–28 and days 29–42). The supplemental levels of Lys were consistent with the nutritional specifications for birds at their respective ages as established in the Management Guidelines for Raising Commercial Turkeys. The experiment was based on a completely randomized 3 × 2 × 2 factorial design with three levels of Arg (90%, 100% and 110%) relative to the content of dietary Met (30 or 45%) and without (−) or with (+) C. perfringens challenge at 34, 36, or 37 d of age. Meat samples were investigated in terms of pH, color, and sarcoplasmic protein profile. The experimental factors did not influence meat quality but the dietary Arg content affected meat color. The sarcoplasmic protein profile was influenced by all studied factors, and glycolytic enzymes were the most abundant. This study evidenced strong association between the challenge conditions and the involvement of glycolytic enzymes in cell metabolism, particularly in inflammatory processes, and DNA replication and maintenance in turkeys. The results showed an effect of C. perfringens infection and feeding with different doses of Arg and Met may lead to significant consequences in cell metabolism.
Collapse
Affiliation(s)
- Paweł Konieczka
- Department of Poultry Science and Apiculture, University of Warmia and Mazury, Olsztyn, Poland.
| | - Elżbieta Żelechowska
- Institute of Human Nutrition Sciences, Warsaw University of Life Sciences, Warsaw, Poland
| | - Wiesław Przybylski
- Institute of Human Nutrition Sciences, Warsaw University of Life Sciences, Warsaw, Poland
| | - Danuta Jaworska
- Institute of Human Nutrition Sciences, Warsaw University of Life Sciences, Warsaw, Poland
| | - Piotr Sałek
- Institute of Human Nutrition Sciences, Warsaw University of Life Sciences, Warsaw, Poland
| | - Misza Kinsner
- Department of Animal Nutrition, The Kielanowski Institute of Animal Physiology and Nutrition, Polish Academy of Sciences, Jabłonna, Poland
| | - Jan Jankowski
- Department of Poultry Science and Apiculture, University of Warmia and Mazury, Olsztyn, Poland
| |
Collapse
|
24
|
Peng F, Liao M, Qin R, Zhu S, Peng C, Fu L, Chen Y, Han B. Regulated cell death (RCD) in cancer: key pathways and targeted therapies. Signal Transduct Target Ther 2022; 7:286. [PMID: 35963853 PMCID: PMC9376115 DOI: 10.1038/s41392-022-01110-y] [Citation(s) in RCA: 407] [Impact Index Per Article: 135.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2022] [Revised: 07/04/2022] [Accepted: 07/05/2022] [Indexed: 02/07/2023] Open
Abstract
Regulated cell death (RCD), also well-known as programmed cell death (PCD), refers to the form of cell death that can be regulated by a variety of biomacromolecules, which is distinctive from accidental cell death (ACD). Accumulating evidence has revealed that RCD subroutines are the key features of tumorigenesis, which may ultimately lead to the establishment of different potential therapeutic strategies. Hitherto, targeting the subroutines of RCD with pharmacological small-molecule compounds has been emerging as a promising therapeutic avenue, which has rapidly progressed in many types of human cancers. Thus, in this review, we focus on summarizing not only the key apoptotic and autophagy-dependent cell death signaling pathways, but the crucial pathways of other RCD subroutines, including necroptosis, pyroptosis, ferroptosis, parthanatos, entosis, NETosis and lysosome-dependent cell death (LCD) in cancer. Moreover, we further discuss the current situation of several small-molecule compounds targeting the different RCD subroutines to improve cancer treatment, such as single-target, dual or multiple-target small-molecule compounds, drug combinations, and some new emerging therapeutic strategies that would together shed new light on future directions to attack cancer cell vulnerabilities with small-molecule drugs targeting RCD for therapeutic purposes.
Collapse
Affiliation(s)
- Fu Peng
- West China School of Pharmacy, State Key Laboratory of Biotherapy and Cancer Center, Department of Gastrointestinal Surgery, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Minru Liao
- West China School of Pharmacy, State Key Laboratory of Biotherapy and Cancer Center, Department of Gastrointestinal Surgery, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Rui Qin
- State Key Laboratory of Southwestern Chinese Medicine Resources, Hospital of Chengdu University of Traditional Chinese Medicine, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, 611137, China
| | - Shiou Zhu
- West China School of Pharmacy, State Key Laboratory of Biotherapy and Cancer Center, Department of Gastrointestinal Surgery, West China Hospital, Sichuan University, Chengdu, 610041, China
- Sichuan Engineering Research Center for Biomimetic Synthesis of Natural Drugs, School of Life Science and Engineering, Southwest Jiaotong University, Chengdu, 610031, China
| | - Cheng Peng
- State Key Laboratory of Southwestern Chinese Medicine Resources, Hospital of Chengdu University of Traditional Chinese Medicine, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, 611137, China
| | - Leilei Fu
- Sichuan Engineering Research Center for Biomimetic Synthesis of Natural Drugs, School of Life Science and Engineering, Southwest Jiaotong University, Chengdu, 610031, China.
| | - Yi Chen
- West China School of Pharmacy, State Key Laboratory of Biotherapy and Cancer Center, Department of Gastrointestinal Surgery, West China Hospital, Sichuan University, Chengdu, 610041, China.
| | - Bo Han
- State Key Laboratory of Southwestern Chinese Medicine Resources, Hospital of Chengdu University of Traditional Chinese Medicine, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, 611137, China.
| |
Collapse
|
25
|
Wang L, Wang W, Cai Y, Zhou Y, Jiang J, Ning Y, Shui C, Sun R, Wang Y, Li C. RETRACTED ARTICLE: Circ-NUP214 Promotes Papillary Thyroid Carcinoma Tumorigenesis by Regulating HK2 Expression Through miR-15a-5p. Biochem Genet 2022; 60:1408. [PMID: 35099648 DOI: 10.1007/s10528-022-10192-w] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2021] [Accepted: 01/18/2022] [Indexed: 01/15/2023]
Affiliation(s)
- Ling Wang
- Department of Otorhinolaryngology Head and Neck Surgery, Affiliated Hospital of Southwest Medical University, No. 55, 4th Section of Southern Renmin Road, Chengdu, Sichuan, China
- Department of Otorhinolaryngology Head and Neck Surgery, The First Affiliated Hospital of Chengdu Medical College, Chengdu, Sichuan, China
| | - Wei Wang
- Department of Head and Neck Surgery, Sichuan Cancer Hospital, Sichuan Cancer Research Institute, Sichuan Cancer Prevention and Cure Center, Cancer Hospital Affiliate to School of Medicine, Chengdu, Sichuan, China
| | - Yongcong Cai
- Department of Head and Neck Surgery, Sichuan Cancer Hospital, Sichuan Cancer Research Institute, Sichuan Cancer Prevention and Cure Center, Cancer Hospital Affiliate to School of Medicine, Chengdu, Sichuan, China
| | - Yuqiu Zhou
- Department of Head and Neck Surgery, Sichuan Cancer Hospital, Sichuan Cancer Research Institute, Sichuan Cancer Prevention and Cure Center, Cancer Hospital Affiliate to School of Medicine, Chengdu, Sichuan, China
| | - Jian Jiang
- Department of Head and Neck Surgery, Sichuan Cancer Hospital, Sichuan Cancer Research Institute, Sichuan Cancer Prevention and Cure Center, Cancer Hospital Affiliate to School of Medicine, Chengdu, Sichuan, China
| | - Yudong Ning
- Department of Head and Neck Surgery, Sichuan Cancer Hospital, Sichuan Cancer Research Institute, Sichuan Cancer Prevention and Cure Center, Cancer Hospital Affiliate to School of Medicine, Chengdu, Sichuan, China
| | - Chunyan Shui
- Department of Head and Neck Surgery, Sichuan Cancer Hospital, Sichuan Cancer Research Institute, Sichuan Cancer Prevention and Cure Center, Cancer Hospital Affiliate to School of Medicine, Chengdu, Sichuan, China
| | - Ronghao Sun
- Department of Head and Neck Surgery, Sichuan Cancer Hospital, Sichuan Cancer Research Institute, Sichuan Cancer Prevention and Cure Center, Cancer Hospital Affiliate to School of Medicine, Chengdu, Sichuan, China
| | - Yi Wang
- Department of Otorhinolaryngology Head and Neck Surgery, The First Affiliated Hospital of Chengdu Medical College, Chengdu, Sichuan, China
| | - Chao Li
- Department of Otorhinolaryngology Head and Neck Surgery, Affiliated Hospital of Southwest Medical University, No. 55, 4th Section of Southern Renmin Road, Chengdu, Sichuan, China.
- Department of Head and Neck Surgery, Sichuan Cancer Hospital, Sichuan Cancer Research Institute, Sichuan Cancer Prevention and Cure Center, Cancer Hospital Affiliate to School of Medicine, Chengdu, Sichuan, China.
| |
Collapse
|
26
|
Icard P, Simula L, Fournel L, Leroy K, Lupo A, Damotte D, Charpentier MC, Durdux C, Loi M, Schussler O, Chassagnon G, Coquerel A, Lincet H, De Pauw V, Alifano M. The strategic roles of four enzymes in the interconnection between metabolism and oncogene activation in non-small cell lung cancer: Therapeutic implications. Drug Resist Updat 2022; 63:100852. [PMID: 35849943 DOI: 10.1016/j.drup.2022.100852] [Citation(s) in RCA: 33] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
NSCLC is the leading cause of cancer mortality and represents a major challenge in cancer therapy. Intrinsic and acquired anticancer drug resistance are promoted by hypoxia and HIF-1α. Moreover, chemoresistance is sustained by the activation of key signaling pathways (such as RAS and its well-known downstream targets PI3K/AKT and MAPK) and several mutated oncogenes (including KRAS and EGFR among others). In this review, we highlight how these oncogenic factors are interconnected with cell metabolism (aerobic glycolysis, glutaminolysis and lipid synthesis). Also, we stress the key role of four metabolic enzymes (PFK1, dimeric-PKM2, GLS1 and ACLY), which promote the activation of these oncogenic pathways in a positive feedback loop. These four tenors orchestrating the coordination of metabolism and oncogenic pathways could be key druggable targets for specific inhibition. Since PFK1 appears as the first tenor of this orchestra, its inhibition (and/or that of its main activator PFK2/PFKFB3) could be an efficacious strategy against NSCLC. Citrate is a potent physiologic inhibitor of both PFK1 and PFKFB3, and NSCLC cells seem to maintain a low citrate level to sustain aerobic glycolysis and the PFK1/PI3K/EGFR axis. Awaiting the development of specific non-toxic inhibitors of PFK1 and PFK2/PFKFB3, we propose to test strategies increasing citrate levels in NSCLC tumors to disrupt this interconnection. This could be attempted by evaluating inhibitors of the citrate-consuming enzyme ACLY and/or by direct administration of citrate at high doses. In preclinical models, this "citrate strategy" efficiently inhibits PFK1/PFK2, HIF-1α, and IGFR/PI3K/AKT axes. It also blocks tumor growth in RAS-driven lung cancer models, reversing dedifferentiation, promoting T lymphocytes tumor infiltration, and increasing sensitivity to cytotoxic drugs.
Collapse
Affiliation(s)
- Philippe Icard
- Thoracic Surgery Department, Paris Center University Hospitals, AP-HP, Paris, France; Normandie Univ, UNICAEN, CHU de Caen Normandie, Unité de recherche BioTICLA INSERM U1086, 14000 Caen, France.
| | - Luca Simula
- Department of Infection, Immunity and Inflammation, Cochin Institute, INSERM U1016, CNRS UMR8104, Paris University, Paris 75014, France
| | - Ludovic Fournel
- Thoracic Surgery Department, Paris Center University Hospitals, AP-HP, Paris, France; INSERM UMR-S 1124, Cellular Homeostasis and Cancer, University of Paris, Paris, France
| | - Karen Leroy
- Department of Genomic Medicine and Cancers, Georges Pompidou European Hospital, APHP, Paris, France
| | - Audrey Lupo
- Pathology Department, Paris Center University Hospitals, AP-HP, Paris, France; INSERM U1138, Integrative Cancer Immunology, University of Paris, 75006 Paris, France
| | - Diane Damotte
- Pathology Department, Paris Center University Hospitals, AP-HP, Paris, France; INSERM U1138, Integrative Cancer Immunology, University of Paris, 75006 Paris, France
| | | | - Catherine Durdux
- Radiation Oncology Department, Georges Pompidou European Hospital, APHP, Paris, France
| | - Mauro Loi
- Radiotherapy Department, University of Florence, Florence, Italy
| | - Olivier Schussler
- Thoracic Surgery Department, Paris Center University Hospitals, AP-HP, Paris, France
| | | | - Antoine Coquerel
- INSERM U1075, COMETE " Mobilités: Attention, Orientation, Chronobiologie", Université Caen, France
| | - Hubert Lincet
- ISPB, Faculté de Pharmacie, Lyon, France, Université Lyon 1, Lyon, France; INSERM U1052, CNRS UMR5286, Cancer Research Center of Lyon (CRCL), France
| | - Vincent De Pauw
- Thoracic Surgery Department, Paris Center University Hospitals, AP-HP, Paris, France
| | - Marco Alifano
- Thoracic Surgery Department, Paris Center University Hospitals, AP-HP, Paris, France; INSERM U1138, Integrative Cancer Immunology, University of Paris, 75006 Paris, France
| |
Collapse
|
27
|
Xie K, Feng J, Fan D, Wang S, Luo J, Ren Z, Zheng C, Diao Y, De Mello RA, Tavolari S, Brandi G, Roden AC, Ren B, Shen Y, Xu L. BARX2/FOXA1/HK2 axis promotes lung adenocarcinoma progression and energy metabolism reprogramming. Transl Lung Cancer Res 2022; 11:1405-1419. [PMID: 35958341 PMCID: PMC9359959 DOI: 10.21037/tlcr-22-465] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2022] [Accepted: 07/15/2022] [Indexed: 11/25/2022]
Abstract
BACKGROUND Metabolic reprogramming is an emerging cancer feature that has recently drawn special attention since it promotes tumor cell growth and proliferation. However, the mechanism of the Warburg effect is still largely unknown. This research aimed to reveal the effects of BarH-like homeobox 2 (BARX2) in regulating tumor progression and glucose metabolism in lung adenocarcinoma (LUAD). METHODS Expression of BARX2 was measured by quantitative real-time polymerase chain reaction (qRT-PCR) in LUAD cell line and tissues, and the tumor-promoting function of BARX2 in LUAD cells was detected in vitro and in vivo xenograft models. The metabolic effects of BARX2 were examined by detecting glucose uptake, the production levels of lactate and pyruvate, and the extracellular acidification rate (ECAR). Chromatin immunoprecipitation (ChIP) assay and luciferase reporter gene assay were used to identify the underlying molecular mechanism of BARX2 regulation of HK2. Further studies showed that transcription factor FOXA1 directly interacts with BARX2 and promotes the transcriptional activity of BARX2. RESULTS BARX2 was remarkably up-regulated in LUAD tissues and positively linked to advanced clinical stage and poor prognosis. In vitro and in vivo data indicated ectopic expression of BARX2 enhanced cell proliferation and tumorigenesis, whereas BARX2 knockdown suppressed these effects. Metabolic-related experiments showed BARX2 promoted the reprogramming of glucose metabolism. Mechanistically, the BARX2/FOXA1/HK2 axis promoted LUAD progression and energy metabolism reprogramming. CONCLUSIONS In summary, our research first defined BARX2 as a tumor-promoting factor in LUAD and that it may act as a novel prognostic biomarker and new therapeutic target for the disease.
Collapse
Affiliation(s)
- Kai Xie
- Department of Cardiothoracic Surgery, Jinling Hospital, School of Nanjing Medical University, Nanjing, China
- Department of Thoracic Surgery, The Affiliated Cancer Hospital of Nanjing Medical University & Jiangsu Cancer Hospital & Jiangsu Institute of Cancer Research, Jiangsu Key Laboratory of Molecular and Translational Cancer Research, Nanjing, China
| | - Jian Feng
- Department of Thoracic Surgery, The Affiliated Cancer Hospital of Nanjing Medical University & Jiangsu Cancer Hospital & Jiangsu Institute of Cancer Research, Jiangsu Key Laboratory of Molecular and Translational Cancer Research, Nanjing, China
- Department of Thoracic Surgery, Shanghai Chest Hospital, Shanghai Jiao Tong University, Shanghai, China
| | - Dingwei Fan
- Department of Thoracic Surgery, The Affiliated Cancer Hospital of Nanjing Medical University & Jiangsu Cancer Hospital & Jiangsu Institute of Cancer Research, Jiangsu Key Laboratory of Molecular and Translational Cancer Research, Nanjing, China
| | - Shi Wang
- Department of Thoracic Surgery, The Affiliated Cancer Hospital of Nanjing Medical University & Jiangsu Cancer Hospital & Jiangsu Institute of Cancer Research, Jiangsu Key Laboratory of Molecular and Translational Cancer Research, Nanjing, China
| | - Jing Luo
- Department of Cardiothoracic Surgery, Jinling Hospital, Medical School of Nanjing University, Nanjing, China
| | - Zhijian Ren
- Department of Thoracic Surgery, The Affiliated Cancer Hospital of Nanjing Medical University & Jiangsu Cancer Hospital & Jiangsu Institute of Cancer Research, Jiangsu Key Laboratory of Molecular and Translational Cancer Research, Nanjing, China
| | - Chao Zheng
- Department of Cardiothoracic Surgery, Jinling Hospital, School of Medicine, Southeast University, Nanjing, China
| | - Yifei Diao
- Department of Cardiothoracic Surgery, Jinling Hospital, Medical School of Nanjing University, Nanjing, China
| | - Ramon Andrade De Mello
- Division of Medical Oncology, Hospital 9 de Julho, São Paulo, SP, Brazil
- Post-Graduation Programme in Medicine, School of Medicine, Nove de Julho University (UNINOVE), São Paulo, SP, Brazil
- Algarve Biomedical Centre, Department of Biomedical Sciences and Medicine, University of Algarve, Faro, Portugal
| | - Simona Tavolari
- Department of Experimental Diagnostic and Specialty Medicine, S. Orsola-Malpighi University Hospital, Bologna, Italy
- Center for Applied Biomedical Research, S. Orsola-Malpighi University Hospital, Bologna, Italy
| | - Giovanni Brandi
- Department of Experimental, Diagnostic and Speciality Medicine, ‘L. & A. Seragnoli’ Institute of Hematology and Medical Oncology, Sant’Orsola-Malpighi Hospital, Bologna, Italy
| | - Anja C. Roden
- Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, MN, USA
| | - Binhui Ren
- Department of Thoracic Surgery, The Affiliated Cancer Hospital of Nanjing Medical University & Jiangsu Cancer Hospital & Jiangsu Institute of Cancer Research, Jiangsu Key Laboratory of Molecular and Translational Cancer Research, Nanjing, China
| | - Yi Shen
- Department of Cardiothoracic Surgery, Jinling Hospital, School of Nanjing Medical University, Nanjing, China
- Department of Cardiothoracic Surgery, Jinling Hospital, Medical School of Nanjing University, Nanjing, China
- Department of Cardiothoracic Surgery, Jinling Hospital, School of Medicine, Southeast University, Nanjing, China
| | - Lin Xu
- Department of Thoracic Surgery, The Affiliated Cancer Hospital of Nanjing Medical University & Jiangsu Cancer Hospital & Jiangsu Institute of Cancer Research, Jiangsu Key Laboratory of Molecular and Translational Cancer Research, Nanjing, China
| |
Collapse
|
28
|
Wu H, Zhou H, Zhang W, Jin P, Shi Q, Miao Z, Wang H, Zha Z. Three birds with one stone: co-encapsulation of diclofenac and DL-menthol for realizing enhanced energy deposition, glycolysis inhibition and anti-inflammation in HIFU surgery. J Nanobiotechnology 2022; 20:215. [PMID: 35524259 PMCID: PMC9074192 DOI: 10.1186/s12951-022-01437-2] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2021] [Accepted: 04/25/2022] [Indexed: 01/12/2023] Open
Abstract
Despite attracting increasing attention in clinic, non-invasive high-intensity focused ultrasound (HIFU) surgery still commonly suffers from tumor recurrence and even matastasis due to the generation of thermo-resistance in non-apoptotic tumor cells and adverse therapy-induced inflammation with enhanced secretion of growth factors in irradiated region. In this work, inspired by the intrinsic property that the expression of thermo-resistant heat shock proteins (HSPs) is highly dependent with adenosine triphosphate (ATP), dual-functionalized diclofenac (DC) with anti-inflammation and glycolysis-inhibition abilities was successfully co-encapsulated with phase-change dl-menthol (DLM) in poly(lactic-co-glycolic acid) nanoparticles (DC/DLM@PLGA NPs) to realize improved HIFU surgery without causing adverse inflammation. Both in vitro and in vivo studies demonstrated the great potential of DC/DLM@PLGA NPs for serving as an efficient synergistic agent for HIFU surgery, which can not only amplify HIFU ablation efficacy through DLM vaporization-induced energy deposition but also simultaneously sensitize tumor cells to hyperthermia by glycolysis inhibition as well as diminished inflammation. Thus, our study provides an efficient strategy for simultaneously improving the curative efficiency and diminishing the harmful inflammatory responses of clinical HIFU surgery.
Collapse
Affiliation(s)
- Haitao Wu
- School of Food and Biological Engineering, School of Instrument Science and Opto-Electronics Engineering, Hefei University of Technology, Anhui, 230009, Hefei, China
| | - Hu Zhou
- Shenzhen Maternity and Child Healthcare Hospital, The First School of Clinical Medicine, Southern Medical University, Shenzhen, 518028, Guangdong, China
| | - Wenjie Zhang
- School of Food and Biological Engineering, School of Instrument Science and Opto-Electronics Engineering, Hefei University of Technology, Anhui, 230009, Hefei, China
| | - Ping Jin
- Shenzhen Maternity and Child Healthcare Hospital, The First School of Clinical Medicine, Southern Medical University, Shenzhen, 518028, Guangdong, China.
| | - Qianqian Shi
- School of Food and Biological Engineering, School of Instrument Science and Opto-Electronics Engineering, Hefei University of Technology, Anhui, 230009, Hefei, China
| | - Zhaohua Miao
- School of Food and Biological Engineering, School of Instrument Science and Opto-Electronics Engineering, Hefei University of Technology, Anhui, 230009, Hefei, China
| | - Hua Wang
- Department of Oncology, Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, The First Affiliated Hospital of Anhui Medical University, Hefei, 230022, Anhui, China.
| | - Zhengbao Zha
- School of Food and Biological Engineering, School of Instrument Science and Opto-Electronics Engineering, Hefei University of Technology, Anhui, 230009, Hefei, China.
| |
Collapse
|
29
|
Osteoblasts induce glucose-derived ATP perturbations in chondrocytes through noncontact communication. Acta Biochim Biophys Sin (Shanghai) 2022; 54:625-636. [PMID: 35593470 PMCID: PMC9828329 DOI: 10.3724/abbs.2022042] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023] Open
Abstract
Cartilage and subchondral bone communicate with each other through material and signal exchanges. However, direct evidence provided by experimental studies on their interactions is insufficient. In the present study, we establish a noncontact co-culture model with a transwell chamber to explore the energetic perturbations in chondrocytes influenced by osteoblasts. Our results indicate that osteoblasts induce more ATP generation in chondrocytes through an energetic shift characterized by enhanced glycolysis and impaired mitochondrial tricarboxylic acid cycle. Enhanced glycolysis is shown by an increase of secreted lactate and the upregulation of glycolytic enzymes, including glucose-6-phosphate isomerase (Gpi), liver type ATP-dependent 6-phosphofructokinase (Pfkl), fructose-bisphosphate aldolase C (Aldoc), glyceraldehyde-3-phosphate dehydrogenase (Gapdh), triosephosphate isomerase (Tpi1), and phosphoglycerate kinase 1 (Pgk1). Impaired mitochondrial tricarboxylic acid cycle is characterized by the downregulation of cytoplasmic aspartate aminotransferase (Got1) and mitochondrial citrate synthase (Cs). Osteoblasts induce the activation of Akt and P38 signaling to mediate ATP perturbations in chondrocytes. This study may deepen our understanding of the maintenance of metabolic homeostasis in the bone-cartilage unit.
Collapse
|
30
|
Abstract
Over the years, the engineering aspect of nanotechnology has been significantly exploited. Medical intervention strategies have been developed by leveraging existing molecular biology knowledge and combining it with nanotechnology tools to improve outcomes. However, little attention has been paid to harnessing the strengths of nanotechnology as a biological discovery tool. Fundamental understanding of controlling dynamic biological processes at the subcellular level is key to developing personalized therapeutic and diagnostic interventions. Single-cell analyses using intravital microscopy, expansion microscopy, and microfluidic-based platforms have been helping to better understand cell heterogeneity in healthy and diseased cells, a major challenge in oncology. Also, single-cell analysis has revealed critical signaling pathways and biological intracellular components with key biological functions. The physical manipulation enabled by nanotools can allow real-time monitoring of biological changes at a single-cell level by sampling intracellular fluid from the same cell. The formation of intercellular highways by nanotube-like structures has important clinical implications such as metastasis development. The integration of nanomaterials into optical and molecular imaging techniques has rendered valuable morphological, structural, and biological information. Nanoscale imaging unravels mechanisms of temporality by enabling the visualization of nanoscale dynamics never observed or measured between individual cells with standard biological techniques. The exceptional sensitivity of nanozymes, artificial enzymes, make them perfect components of the next-generation mobile diagnostics devices. Here, we highlight these impactful cancer-related biological discoveries enabled by nanotechnology and producing a paradigm shift in cancer research and oncology.
Collapse
Affiliation(s)
- Carolina Salvador-Morales
- Nanodelivery Systems and Devices Branch, Cancer Imaging Program, Division of Cancer Treatment and Diagnosis, National Cancer Institute, National Institutes of Health, Rockville, Maryland 20850, United States
| | - Piotr Grodzinski
- Nanodelivery Systems and Devices Branch, Cancer Imaging Program, Division of Cancer Treatment and Diagnosis, National Cancer Institute, National Institutes of Health, Rockville, Maryland 20850, United States
| |
Collapse
|
31
|
da Silva EL, Mesquita FP, de Sousa Portilho AJ, Bezerra ECA, Daniel JP, Aranha ESP, Farran S, de Vasconcellos MC, de Moraes MEA, Moreira-Nunes CA, Montenegro RC. Differences in glucose concentration shows new perspectives in gastric cancer metabolism. Toxicol In Vitro 2022; 82:105357. [PMID: 35427737 DOI: 10.1016/j.tiv.2022.105357] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2021] [Revised: 03/16/2022] [Accepted: 04/07/2022] [Indexed: 12/06/2022]
Abstract
Gastric cancer (GC) is among the deadliest cancers worldwide despite available therapies, highlighting the need for novel therapies and pharmacological agents. Metabolic deregulation is a potential study area for new anticancer targets, but the in vitro metabolic studies are controversial, as different ranges of glucose used in the culture medium can influence results. In this study, we evaluated cellular viability, glucose uptake, and LDH activity in gastric cell lines when exposed to different glucose concentrations: high (HG, 25 mM), low (LG, 5.5 mM), and free (FG, 0 mM) glucose mediums. Moreover, we evaluated how glucose variations may influence cellular phenotype and the expression of genes related to epithelial-mesenchymal transition (EMT), metabolism, and cancer development in metastatic GC cells (AGP-01). Results showed that in the FG metastatic cells evidenced higher viability when compared with other cell lines and that when exposed to either LG or HG mediums most of the phenotypic assays did not differ. However, cells exposed to LG increased colony formation and mRNA levels of metabolic-related genes when compared to HG medium. Our results recommend LG medium to metabolic studies once glucose concentration is closer to physiological levels. These findings are important to point out new relevant targets in metabolic reprogramming that can be alternatives to current chemotherapies in patients with metastatic GC.
Collapse
Affiliation(s)
- Emerson Lucena da Silva
- Laboratory of Pharmacogenetics, Drug Research and Development Center (NPDM), Federal University of Ceará, Cel. Nunes de Melo, 1000 - Rodolfo Teófilo, Fortaleza, Brazil
| | - Felipe Pantoja Mesquita
- Laboratory of Pharmacogenetics, Drug Research and Development Center (NPDM), Federal University of Ceará, Cel. Nunes de Melo, 1000 - Rodolfo Teófilo, Fortaleza, Brazil
| | - Adrhyann Jullyanne de Sousa Portilho
- Laboratory of Pharmacogenetics, Drug Research and Development Center (NPDM), Federal University of Ceará, Cel. Nunes de Melo, 1000 - Rodolfo Teófilo, Fortaleza, Brazil
| | - Emanuel Cintra Austregésilo Bezerra
- Laboratory of Pharmacogenetics, Drug Research and Development Center (NPDM), Federal University of Ceará, Cel. Nunes de Melo, 1000 - Rodolfo Teófilo, Fortaleza, Brazil
| | - Julio Paulino Daniel
- Laboratory of Pharmacogenetics, Drug Research and Development Center (NPDM), Federal University of Ceará, Cel. Nunes de Melo, 1000 - Rodolfo Teófilo, Fortaleza, Brazil
| | - Elenn Suzany Pereira Aranha
- Biological Activity Laboratory, Faculty of Pharmaceutical Sciences, Federal University of Amazonas, Av. General Rodrigo Octavio Jordão Ramos, 1200 - Coroado, Manaus, Brazil
| | - Sarah Farran
- Department of Pathology and Laboratory Medicine, American University of Beirut Medical Center - Riad El-Solh, Beirut, Lebanon
| | - Marne Carvalho de Vasconcellos
- Biological Activity Laboratory, Faculty of Pharmaceutical Sciences, Federal University of Amazonas, Av. General Rodrigo Octavio Jordão Ramos, 1200 - Coroado, Manaus, Brazil
| | - Maria Elisabete Amaral de Moraes
- Laboratory of Pharmacogenetics, Drug Research and Development Center (NPDM), Federal University of Ceará, Cel. Nunes de Melo, 1000 - Rodolfo Teófilo, Fortaleza, Brazil
| | - Caroline Aquino Moreira-Nunes
- Laboratory of Pharmacogenetics, Drug Research and Development Center (NPDM), Federal University of Ceará, Cel. Nunes de Melo, 1000 - Rodolfo Teófilo, Fortaleza, Brazil
| | - Raquel Carvalho Montenegro
- Laboratory of Pharmacogenetics, Drug Research and Development Center (NPDM), Federal University of Ceará, Cel. Nunes de Melo, 1000 - Rodolfo Teófilo, Fortaleza, Brazil.
| |
Collapse
|
32
|
Naturally occurring deamidated triosephosphate isomerase is a promising target for cell-selective therapy in cancer. Sci Rep 2022; 12:4028. [PMID: 35256749 PMCID: PMC8901631 DOI: 10.1038/s41598-022-08051-0] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2021] [Accepted: 02/28/2022] [Indexed: 11/08/2022] Open
Abstract
Human triosephosphate isomerase (HsTIM) is a central glycolytic enzyme and is overexpressed in cancer cells with accelerated glycolysis. Triple-negative breast cancer is highly dependent on glycolysis and is typically treated with a combination of surgery, radiation therapy, and chemotherapy. Deamidated HsTIM was recently proposed as a druggable target. Although thiol-reactive drugs affect cell growth in deamidated HsTIM-complemented cells, the role of this protein as a selective target has not been demonstrated. To delve into the usefulness of deamidated HsTIM as a selective target, we assessed its natural accumulation in breast cancer cells. We found that deamidated HsTIM accumulates in breast cancer cells but not in noncancerous cells. The cancer cells are selectively programmed to undergo cell death with thiol-reactive drugs that induced the production of methylglyoxal (MGO) and advanced glycation-end products (AGEs). In vivo, a thiol-reactive drug effectively inhibits the growth of xenograft tumors with an underlying mechanism involving deamidated HsTIM. Our findings demonstrate the usefulness of deamidated HsTIM as target to develop new therapeutic strategies for the treatment of cancers and other pathologies in which this post translationally modified protein accumulates.
Collapse
|
33
|
Li H, Kim H, Xu F, Han J, Yao Q, Wang J, Pu K, Peng X, Yoon J. Activity-based NIR fluorescent probes based on the versatile hemicyanine scaffold: design strategy, biomedical applications, and outlook. Chem Soc Rev 2022; 51:1795-1835. [PMID: 35142301 DOI: 10.1039/d1cs00307k] [Citation(s) in RCA: 198] [Impact Index Per Article: 66.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
The discovery of a near-infrared (NIR, 650-900 nm) fluorescent chromophore hemicyanine dye with high structural tailorability is of great significance in the field of detection, bioimaging, and medical therapeutic applications. It exhibits many outstanding advantages including absorption and emission in the NIR region, tunable spectral properties, high photostability as well as a large Stokes shift. These properties are superior to those of conventional fluorogens, such as coumarin, fluorescein, naphthalimides, rhodamine, and cyanine. Researchers have made remarkable progress in developing activity-based multifunctional fluorescent probes based on hemicyanine skeletons for monitoring vital biomolecules in living systems through the output of fluorescence/photoacoustic signals, and integration of diagnosis and treatment of diseases using chemotherapy or photothermal/photodynamic therapy or combination therapy. These achievements prompted researchers to develop more smart fluorescent probes using a hemicyanine fluorogen as a template. In this review, we begin by describing the brief history of the discovery of hemicyanine dyes, synthetic approaches, and design strategies for activity-based functional fluorescent probes. Then, many selected hemicyanine-based probes that can detect ions, small biomolecules, overexpressed enzymes and diagnostic reagents for diseases are systematically highlighted. Finally, potential drawbacks and the outlook for future investigation and clinical medicine transformation of hemicyanine-based activatable functional probes are also discussed.
Collapse
Affiliation(s)
- Haidong Li
- State Key Laboratory of Fine Chemicals, Dalian University of Technology, 2 Linggong Road, Dalian 116024, China. .,School of Bioengineering, Dalian University of Technology, 2 Linggong Road, Dalian 116024, China
| | - Heejeong Kim
- Department of Chemistry and Nano Science, Ewha Womans University, Seoul 03760, Korea.
| | - Feng Xu
- State Key Laboratory of Fine Chemicals, Dalian University of Technology, 2 Linggong Road, Dalian 116024, China. .,The Key Laboratory of Laboratory Medicine, Ministry of Education, School of Laboratory Medicine and Life Science, Wenzhou Medical University, Wenzhou 325035, China
| | - Jingjing Han
- Department of Chemistry and Nano Science, Ewha Womans University, Seoul 03760, Korea.
| | - Qichao Yao
- State Key Laboratory of Fine Chemicals, Dalian University of Technology, 2 Linggong Road, Dalian 116024, China.
| | - Jingyun Wang
- State Key Laboratory of Fine Chemicals, Dalian University of Technology, 2 Linggong Road, Dalian 116024, China. .,School of Bioengineering, Dalian University of Technology, 2 Linggong Road, Dalian 116024, China
| | - Kanyi Pu
- School of Chemical and Biomedical Engineering, Nanyang Technological University, 70 Nanyang Drive, 637457, Singapore. .,Division of Chemistry and Biological Chemistry, School of Physical and Mathematical Sciences, Nanyang Technological University, 21 Nanyang Link, Singapore
| | - Xiaojun Peng
- State Key Laboratory of Fine Chemicals, Dalian University of Technology, 2 Linggong Road, Dalian 116024, China. .,Research Institute of Dalian University of Technology in Shenzhen, Nanshan District, Shenzhen 518057, China
| | - Juyoung Yoon
- Department of Chemistry and Nano Science, Ewha Womans University, Seoul 03760, Korea.
| |
Collapse
|
34
|
Tehlan A, Bhowmick K, Kumar A, Subbarao N, Dhar SK. The tetrameric structure of Plasmodium falciparum phosphoglycerate mutase is critical for optimal enzymatic activity. J Biol Chem 2022; 298:101713. [PMID: 35150741 PMCID: PMC8913309 DOI: 10.1016/j.jbc.2022.101713] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2021] [Revised: 02/02/2022] [Accepted: 02/03/2022] [Indexed: 11/29/2022] Open
Abstract
The glycolytic enzyme phosphoglycerate mutase (PGM) is of utmost importance for overall cellular metabolism and has emerged as a novel therapeutic target in cancer cells. This enzyme is also conserved in the rapidly proliferating malarial parasite Plasmodium falciparum, which have a similar metabolic framework as cancer cells and rely on glycolysis as the sole energy-yielding process during intraerythrocytic development. There is no redundancy among the annotated PGM enzymes in Plasmodium, and PfPGM1 is absolutely required for the parasite survival as evidenced by conditional knockdown in our study. A detailed comparison of PfPGM1 with its counterparts followed by in-depth structure-function analysis revealed unique attributes of this parasitic protein. Here, we report for the first time the importance of oligomerization for the optimal functioning of the enzyme in vivo, as earlier studies in eukaryotes only focused on the effects in vitro. We show that single point mutation of the amino acid residue W68 led to complete loss of tetramerization and diminished catalytic activity in vitro. Additionally, ectopic expression of the WT PfPGM1 protein enhanced parasite growth, whereas the monomeric form of PfPGM1 failed to provide growth advantage. Furthermore, mutation of the evolutionarily conserved residue K100 led to a drastic reduction in enzymatic activity. The indispensable nature of this parasite enzyme highlights the potential of PfPGM1 as a therapeutic target against malaria, and targeting the interfacial residues critical for oligomerization can serve as a focal point for promising drug development strategies that may not be restricted to malaria only.
Collapse
Affiliation(s)
- Ankita Tehlan
- Special Centre for Molecular Medicine, Jawaharlal Nehru University, New Delhi 110067
| | - Krishanu Bhowmick
- Special Centre for Molecular Medicine, Jawaharlal Nehru University, New Delhi 110067
| | - Amarjeet Kumar
- School of Computational and Integrative Sciences, Jawaharlal Nehru University, New Delhi, 110067, India
| | - Naidu Subbarao
- School of Computational and Integrative Sciences, Jawaharlal Nehru University, New Delhi, 110067, India
| | - Suman Kumar Dhar
- Special Centre for Molecular Medicine, Jawaharlal Nehru University, New Delhi 110067.
| |
Collapse
|
35
|
A non-canonical role for pyruvate kinase M2 as a functional modulator of Ca 2+ signalling through IP 3 receptors. BIOCHIMICA ET BIOPHYSICA ACTA. MOLECULAR CELL RESEARCH 2022; 1869:119206. [PMID: 35026348 DOI: 10.1016/j.bbamcr.2021.119206] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/01/2021] [Revised: 12/20/2021] [Accepted: 12/23/2021] [Indexed: 12/22/2022]
Abstract
Pyruvate kinase isoform M2 (PKM2) is a rate-limiting glycolytic enzyme that is widely expressed in embryonic tissues. The expression of PKM2 declines in some tissues following embryogenesis, while other pyruvate kinase isozymes are upregulated. However, PKM2 is highly expressed in cancer cells and is believed to play a role in supporting anabolic processes during tumour formation. In this study, PKM2 was identified as an inositol 1,4,5-trisphosphate receptor (IP3R)-interacting protein by mass spectrometry. The PKM2:IP3R interaction was further characterized by pull-down and co-immunoprecipitation assays, which showed that PKM2 interacted with all three IP3R isoforms. Moreover, fluorescence microscopy indicated that both IP3R and PKM2 localized at the endoplasmic reticulum. PKM2 binds to IP3R at a highly conserved 21-amino acid site (corresponding to amino acids 2078-2098 in mouse type 1 IP3R isoform). Synthetic peptides (denoted 'TAT-D5SD' and 'D5SD'), based on the amino acid sequence at this site, disrupted the PKM2:IP3R interaction and potentiated IP3R-mediated Ca2+ release both in intact cells (TAT-D5SD peptide) and in a unidirectional 45Ca2+ flux assay on permeabilized cells (D5SD peptide). The TAT-D5SD peptide did not affect the enzymatic activity of PKM2. Reducing PKM2 protein expression using siRNA increased IP3R-mediated Ca2+ signalling in intact cells without altering the ER Ca2+ content. These data identify PKM2 as an IP3R-interacting protein that inhibits intracellular Ca2+ signalling. The elevated expression of PKM2 in cancer cells is therefore not solely connected to its canonical role in glycolytic metabolism, rather PKM2 also has a novel non-canonical role in regulating intracellular signalling.
Collapse
|
36
|
Zhang S, Hua Z, Ba G, Xu N, Miao J, Zhao G, Gong W, Liu Z, Thiele CJ, Li Z. Antitumor effects of the small molecule DMAMCL in neuroblastoma via suppressing aerobic glycolysis and targeting PFKL. Cancer Cell Int 2021; 21:619. [PMID: 34819091 PMCID: PMC8613996 DOI: 10.1186/s12935-021-02330-y] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2021] [Accepted: 11/10/2021] [Indexed: 12/20/2022] Open
Abstract
Background Neuroblastoma (NB) is a common solid malignancy in children that is associated with a poor prognosis. Although the novel small molecular compound Dimethylaminomicheliolide (DMAMCL) has been shown to induce cell death in some tumors, little is known about its role in NB. Methods We examined the effect of DMAMCL on four NB cell lines (NPG, AS, KCNR, BE2). Cellular confluence, survival, apoptosis, and glycolysis were detected using Incucyte ZOOM, CCK-8 assays, Annexin V-PE/7-AAD flow cytometry, and Seahorse XFe96, respectively. Synergistic effects between agents were evaluated using CompuSyn and the effect of DMAMCL in vivo was evaluated using a xenograft mouse model. Phosphofructokinase-1, liver type (PFKL) expression was up- and down-regulated using overexpression plasmids or siRNA. Results When administered as a single agent, DMAMCL decreased cell proliferation in a time- and dose-dependent manner, increased the percentage of cells in SubG1 phase, and induced apoptosis in vitro, as well as inhibiting tumor growth and prolonging survival in tumor-bearing mice (NGP, BE2) in vivo. In addition, DMAMCL exerted synergistic effects when combined with etoposide or cisplatin in vitro and displayed increased antitumor effects when combined with etoposide in vivo compared to either agent alone. Mechanistically, DMAMCL suppressed aerobic glycolysis by decreasing glucose consumption, lactate excretion, and ATP production, as well as reducing the expression of PFKL, a key glycolysis enzyme, in vitro and in vivo. Furthermore, PFKL overexpression attenuated DMAMCL-induced cell death, whereas PFKL silencing promoted NB cell death. Conclusions The results of this study suggest that DMAMCL exerts antitumor effects on NB both in vitro and in vivo by suppressing aerobic glycolysis and that PFKL could be a potential target of DMAMCL in NB. Supplementary Information The online version contains supplementary material available at 10.1186/s12935-021-02330-y.
Collapse
Affiliation(s)
- Simeng Zhang
- Department of Pediatrics, Shengjing Hospital of China Medical University, Shenyang, China.,Medical Research Center, Liaoning Key Laboratory of Research and Application of Animal Models for Environment and Metabolic Diseases, Shengjing Hospital of China Medical University, #36 Sanhao Street, Heping District, Shenyang, 110004, China
| | - Zhongyan Hua
- Department of Pediatrics, Shengjing Hospital of China Medical University, Shenyang, China.,Medical Research Center, Liaoning Key Laboratory of Research and Application of Animal Models for Environment and Metabolic Diseases, Shengjing Hospital of China Medical University, #36 Sanhao Street, Heping District, Shenyang, 110004, China
| | - Gen Ba
- Department of Pediatrics, Shengjing Hospital of China Medical University, Shenyang, China.,Medical Research Center, Liaoning Key Laboratory of Research and Application of Animal Models for Environment and Metabolic Diseases, Shengjing Hospital of China Medical University, #36 Sanhao Street, Heping District, Shenyang, 110004, China
| | - Ning Xu
- Department of Pediatrics, Shengjing Hospital of China Medical University, Shenyang, China.,Medical Research Center, Liaoning Key Laboratory of Research and Application of Animal Models for Environment and Metabolic Diseases, Shengjing Hospital of China Medical University, #36 Sanhao Street, Heping District, Shenyang, 110004, China
| | - Jianing Miao
- Department of Pediatrics, Shengjing Hospital of China Medical University, Shenyang, China.,Medical Research Center, Liaoning Key Laboratory of Research and Application of Animal Models for Environment and Metabolic Diseases, Shengjing Hospital of China Medical University, #36 Sanhao Street, Heping District, Shenyang, 110004, China
| | - Guifeng Zhao
- Department of Pediatrics, Shengjing Hospital of China Medical University, Shenyang, China.,Medical Research Center, Liaoning Key Laboratory of Research and Application of Animal Models for Environment and Metabolic Diseases, Shengjing Hospital of China Medical University, #36 Sanhao Street, Heping District, Shenyang, 110004, China
| | - Wei Gong
- Department of Pediatrics, Shengjing Hospital of China Medical University, Shenyang, China.,Medical Research Center, Liaoning Key Laboratory of Research and Application of Animal Models for Environment and Metabolic Diseases, Shengjing Hospital of China Medical University, #36 Sanhao Street, Heping District, Shenyang, 110004, China
| | - Zhihui Liu
- Cellular & Molecular Biology Section, Pediatric Oncology Branch, National Cancer Institute, National Institutes of Health Bethesda, Bethesda, MD, 20892, USA
| | - Carol J Thiele
- Cellular & Molecular Biology Section, Pediatric Oncology Branch, National Cancer Institute, National Institutes of Health Bethesda, Bethesda, MD, 20892, USA
| | - Zhijie Li
- Department of Pediatrics, Shengjing Hospital of China Medical University, Shenyang, China. .,Medical Research Center, Liaoning Key Laboratory of Research and Application of Animal Models for Environment and Metabolic Diseases, Shengjing Hospital of China Medical University, #36 Sanhao Street, Heping District, Shenyang, 110004, China.
| |
Collapse
|
37
|
Zuo J, Tang J, Lu M, Zhou Z, Li Y, Tian H, Liu E, Gao B, Liu T, Shao P. Glycolysis Rate-Limiting Enzymes: Novel Potential Regulators of Rheumatoid Arthritis Pathogenesis. Front Immunol 2021; 12:779787. [PMID: 34899740 PMCID: PMC8651870 DOI: 10.3389/fimmu.2021.779787] [Citation(s) in RCA: 51] [Impact Index Per Article: 12.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2021] [Accepted: 11/02/2021] [Indexed: 01/10/2023] Open
Abstract
Rheumatoid arthritis (RA) is a classic autoimmune disease characterized by uncontrolled synovial proliferation, pannus formation, cartilage injury, and bone destruction. The specific pathogenesis of RA, a chronic inflammatory disease, remains unclear. However, both key glycolysis rate-limiting enzymes, hexokinase-II (HK-II), phosphofructokinase-1 (PFK-1), and pyruvate kinase M2 (PKM2), as well as indirect rate-limiting enzymes, 6-phosphofructo-2-kinase/fructose-2,6-bisphosphatase 3 (PFKFB3), are thought to participate in the pathogenesis of RA. In here, we review the latest literature on the pathogenesis of RA, introduce the pathophysiological characteristics of HK-II, PFK-1/PFKFB3, and PKM2 and their expression characteristics in this autoimmune disease, and systematically assess the association between the glycolytic rate-limiting enzymes and RA from a molecular level. Moreover, we highlight HK-II, PFK-1/PFKFB3, and PKM2 as potential targets for the clinical treatment of RA. There is great potential to develop new anti-rheumatic therapies through safe inhibition or overexpression of glycolysis rate-limiting enzymes.
Collapse
Affiliation(s)
- Jianlin Zuo
- Department of Orthopeadics, China-Japan Union Hospital of Jilin University, Changchun, China
| | - Jinshuo Tang
- Department of Orthopeadics, China-Japan Union Hospital of Jilin University, Changchun, China
| | - Meng Lu
- Department of Nursing, The First Bethune Hospital of Jilin University, Changchun, China
| | - Zhongsheng Zhou
- Department of Orthopeadics, China-Japan Union Hospital of Jilin University, Changchun, China
| | - Yang Li
- Scientific Research Center, China-Japan Union Hospital of Jilin University, Changchun, China
| | - Hao Tian
- Department of Orthopeadics, China-Japan Union Hospital of Jilin University, Changchun, China
| | - Enbo Liu
- Department of Orthopeadics, China-Japan Union Hospital of Jilin University, Changchun, China
| | - Baoying Gao
- Department of Cardiology, China-Japan Union Hospital of Jilin University, Changchun, China
| | - Te Liu
- Scientific Research Center, China-Japan Union Hospital of Jilin University, Changchun, China
| | - Pu Shao
- Department of Orthopeadics, China-Japan Union Hospital of Jilin University, Changchun, China
- Scientific Research Center, China-Japan Union Hospital of Jilin University, Changchun, China
| |
Collapse
|
38
|
Rodriguez S, Skeet K, Mehmetoglu-Gurbuz T, Goldfarb M, Karri S, Rocha J, Shahinian M, Yazadi A, Poudel S, Subramani R. Phytochemicals as an Alternative or Integrative Option, in Conjunction with Conventional Treatments for Hepatocellular Carcinoma. Cancers (Basel) 2021; 13:cancers13225753. [PMID: 34830907 PMCID: PMC8616323 DOI: 10.3390/cancers13225753] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2021] [Revised: 11/10/2021] [Accepted: 11/15/2021] [Indexed: 12/12/2022] Open
Abstract
Simple Summary Hepatocellular carcinoma (HCC) is globally ranked as the sixth most diagnosed cancer, and the second most deadly cancer. To worsen matters, there are only limited therapeutic options currently available; therefore, it is necessary to find a reservoir from which new HCC treatments may be acquired. The field of phytomedicine may be the solution to this problem, as it offers an abundance of plant-derived molecules, which show capabilities of being effective against HCC proliferation, invasion, migration, and metastasis. In our review, we collect and analyze current evidence regarding these promising phytochemical effects on HCC, and delve into their potential as future chemotherapies. Additionally, information on the signaling behind these numerous phytochemicals is provided, in an attempt to understand their mechanisms. This review makes accessible the current body of knowledge pertaining to phytochemicals as HCC treatments, in order to serve as a reference and inspiration for further research into this subject. Abstract Hepatocellular carcinoma (HCC) is the most abundant form of liver cancer. It accounts for 75–85% of liver cancer cases and, though it ranks globally as the sixth most common cancer, it ranks second in cancer-related mortality. Deaths from HCC are usually due to metastatic spread of the cancer. Unfortunately, there are many challenges and limitations with the latest HCC therapies and medications, making it difficult for patients to receive life-prolonging care. As there is clearly a high demand for alternative therapy options for HCC, it is prudent to turn to plants for the solution, as their phytochemicals have long been used and revered for their many medicinal purposes. This review explores the promising phytochemical compounds identified from pre-clinical and clinical trials being used either independently or in conjunction with already existing cancer therapy treatments. The phytochemicals discussed in this review were classified into several categories: lipids, polyphenols, alkaloids, polysaccharides, whole extracts, and phytochemical combinations. Almost 80% of the compounds failed to progress into clinical studies due to lack of information regarding the toxicity to normal cells and bioavailability. Although large obstacles remain, phytochemicals can be used either as an alternative or integrative therapy in conjunction with existing HCC chemotherapies. In conclusion, phytochemicals have great potential as treatment options for hepatocellular carcinoma.
Collapse
Affiliation(s)
- Sheryl Rodriguez
- Center of Emphasis in Cancer Research, Department of Molecular and Translational Medicine, Paul L. Foster School of Medicine, Texas Tech University Health Sciences Center El Paso, El Paso, TX 79905, USA; (S.R.); (T.M.-G.); (S.P.)
| | - Kristy Skeet
- Graduate School of Biomedical Sciences, Texas Tech University Health Sciences Center, El Paso, TX 79905, USA; (K.S.); (J.R.); (M.S.); (A.Y.)
| | - Tugba Mehmetoglu-Gurbuz
- Center of Emphasis in Cancer Research, Department of Molecular and Translational Medicine, Paul L. Foster School of Medicine, Texas Tech University Health Sciences Center El Paso, El Paso, TX 79905, USA; (S.R.); (T.M.-G.); (S.P.)
| | - Madeline Goldfarb
- Paul L. Foster School of Medicine, Texas Tech University Health Sciences Center El Paso, El Paso, TX 79905, USA; (M.G.); (S.K.)
| | - Shri Karri
- Paul L. Foster School of Medicine, Texas Tech University Health Sciences Center El Paso, El Paso, TX 79905, USA; (M.G.); (S.K.)
| | - Jackelyn Rocha
- Graduate School of Biomedical Sciences, Texas Tech University Health Sciences Center, El Paso, TX 79905, USA; (K.S.); (J.R.); (M.S.); (A.Y.)
| | - Mark Shahinian
- Graduate School of Biomedical Sciences, Texas Tech University Health Sciences Center, El Paso, TX 79905, USA; (K.S.); (J.R.); (M.S.); (A.Y.)
| | - Abdallah Yazadi
- Graduate School of Biomedical Sciences, Texas Tech University Health Sciences Center, El Paso, TX 79905, USA; (K.S.); (J.R.); (M.S.); (A.Y.)
| | - Seeta Poudel
- Center of Emphasis in Cancer Research, Department of Molecular and Translational Medicine, Paul L. Foster School of Medicine, Texas Tech University Health Sciences Center El Paso, El Paso, TX 79905, USA; (S.R.); (T.M.-G.); (S.P.)
| | - Ramadevi Subramani
- Center of Emphasis in Cancer Research, Department of Molecular and Translational Medicine, Paul L. Foster School of Medicine, Texas Tech University Health Sciences Center El Paso, El Paso, TX 79905, USA; (S.R.); (T.M.-G.); (S.P.)
- Graduate School of Biomedical Sciences, Texas Tech University Health Sciences Center, El Paso, TX 79905, USA; (K.S.); (J.R.); (M.S.); (A.Y.)
- Correspondence: ; Tel.: +1-915-215-6851
| |
Collapse
|
39
|
Alvarez R, Mandal D, Chittiboina P. Canonical and Non-Canonical Roles of PFKFB3 in Brain Tumors. Cells 2021; 10:cells10112913. [PMID: 34831136 PMCID: PMC8616071 DOI: 10.3390/cells10112913] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2021] [Revised: 10/18/2021] [Accepted: 10/18/2021] [Indexed: 12/27/2022] Open
Abstract
PFKFB3 is a bifunctional enzyme that modulates and maintains the intracellular concentrations of fructose-2,6-bisphosphate (F2,6-P2), essentially controlling the rate of glycolysis. PFKFB3 is a known activator of glycolytic rewiring in neoplastic cells, including central nervous system (CNS) neoplastic cells. The pathologic regulation of PFKFB3 is invoked via various microenvironmental stimuli and oncogenic signals. Hypoxia is a primary inducer of PFKFB3 transcription via HIF-1alpha. In addition, translational modifications of PFKFB3 are driven by various intracellular signaling pathways that allow PFKFB3 to respond to varying stimuli. PFKFB3 synthesizes F2,6P2 through the phosphorylation of F6P with a donated PO4 group from ATP and has the highest kinase activity of all PFKFB isoenzymes. The intracellular concentration of F2,6P2 in cancers is maintained primarily by PFKFB3 allowing cancer cells to evade glycolytic suppression. PFKFB3 is a primary enzyme responsible for glycolytic tumor metabolic reprogramming. PFKFB3 protein levels are significantly higher in high-grade glioma than in non-pathologic brain tissue or lower grade gliomas, but without relative upregulation of transcript levels. High PFKFB3 expression is linked to poor survival in brain tumors. Solitary or concomitant PFKFB3 inhibition has additionally shown great potential in restoring chemosensitivity and radiosensitivity in treatment-resistant brain tumors. An improved understanding of canonical and non-canonical functions of PFKFB3 could allow for the development of effective combinatorial targeted therapies for brain tumors.
Collapse
Affiliation(s)
- Reinier Alvarez
- Department of Neurological Surgery, University of Colorado School of Medicine, Aurora, CO 80045, USA;
- Neurosurgery Unit for Pituitary and Inheritable Disorders, National Institute of Neurological Disorders and Stroke, Bethesda, MD 20824, USA;
| | - Debjani Mandal
- Neurosurgery Unit for Pituitary and Inheritable Disorders, National Institute of Neurological Disorders and Stroke, Bethesda, MD 20824, USA;
| | - Prashant Chittiboina
- Neurosurgery Unit for Pituitary and Inheritable Disorders, National Institute of Neurological Disorders and Stroke, Bethesda, MD 20824, USA;
- Surgical Neurology Branch, National Institute of Neurological Disorders and Stroke, Bethesda, MD 20824, USA
- Correspondence:
| |
Collapse
|
40
|
Daks A, Shuvalov O, Fedorova O, Petukhov A, Lezina L, Zharova A, Baidyuk E, Khudiakov A, Barlev NA. p53-Independent Effects of Set7/9 Lysine Methyltransferase on Metabolism of Non-Small Cell Lung Cancer Cells. Front Oncol 2021; 11:706668. [PMID: 34692483 PMCID: PMC8528242 DOI: 10.3389/fonc.2021.706668] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2021] [Accepted: 09/14/2021] [Indexed: 11/13/2022] Open
Abstract
Set7/9 is a lysine-specific methyltransferase, which regulates the functioning of both the histone and non-histone substrates, thereby significantly affecting the global gene expression landscape. Using microarray expression profiling, we have identified several key master regulators of metabolic networks, including c-Myc, that were affected by Set7/9 status. Consistent with this observation, c-Myc transcriptional targets-genes encoding the glycolytic enzymes hexokinase (HK2), aldolase (ALDOB), and lactate dehydrogenase (LDHA)-were upregulated upon Set7/9 knockdown (Set7/9KD). Importantly, we showed the short hairpin RNA (shRNA)-mediated attenuation of Set7/9 augmented c-Myc, GLUT1, HK2, ALDOA, and LDHA expression in non-small cell lung cancer (NSCLC) cell lines, not only at the transcriptional but also at the protein level. In line with this observation, Set7/9KD significantly augmented the membrane mitochondrial potential (MMP), glycolysis, respiration, and the proliferation rate of NSCLC cells. Importantly, all these effects of Set7/9 on cell metabolism were p53-independent. Bioinformatic analysis has shown a synergistic impact of Set7/9 together with either GLUT1, HIF1A, HK2, or LDHA on the survival of lung cancer patients. Based on these evidence, we hypothesize that Set7/9 can be an important regulator of energy metabolism in NSCLC.
Collapse
Affiliation(s)
- Alexandra Daks
- Institute of Cytology, Russian Academy of Sciences, St Petersburg, Russia
| | - Oleg Shuvalov
- Institute of Cytology, Russian Academy of Sciences, St Petersburg, Russia
| | - Olga Fedorova
- Institute of Cytology, Russian Academy of Sciences, St Petersburg, Russia
| | - Alexey Petukhov
- Institute of Cytology, Russian Academy of Sciences, St Petersburg, Russia.,Institute of Molecular Biology and Genetics, Almazov National Medical Research Centre, St Petersburg, Russia
| | - Larissa Lezina
- Regulation of Cell Signaling Laboratory, Moscow Institute of Physics and Technology, Dolgoprudny, Russia
| | - Arsenia Zharova
- Institute of Cytology, Russian Academy of Sciences, St Petersburg, Russia
| | - Ekaterina Baidyuk
- Institute of Cytology, Russian Academy of Sciences, St Petersburg, Russia
| | - Alexander Khudiakov
- Institute of Molecular Biology and Genetics, Almazov National Medical Research Centre, St Petersburg, Russia
| | - Nickolai A Barlev
- Institute of Cytology, Russian Academy of Sciences, St Petersburg, Russia.,Regulation of Cell Signaling Laboratory, Moscow Institute of Physics and Technology, Dolgoprudny, Russia
| |
Collapse
|
41
|
Soni VK, Mehta A, Ratre YK, Chandra V, Shukla D, Kumar A, Vishvakarma NK. Counteracting Action of Curcumin on High Glucose-Induced Chemoresistance in Hepatic Carcinoma Cells. Front Oncol 2021; 11:738961. [PMID: 34692517 PMCID: PMC8526934 DOI: 10.3389/fonc.2021.738961] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2021] [Accepted: 09/10/2021] [Indexed: 12/11/2022] Open
Abstract
Along with direct anticancer activity, curcumin hinders the onset of chemoresistance. Among many, high glucose condition is a key driving factor for chemoresistance. However, the ability of curcumin remains unexplored against high glucose-induced chemoresistance. Moreover, chemoresistance is major hindrance in effective clinical management of liver cancer. Using hepatic carcinoma HepG2 cells, the present investigation demonstrates that high glucose induces chemoresistance, which is averted by the simultaneous presence of curcumin. Curcumin obviated the hyperglycemia-induced modulations like elevated glucose consumption, lactate production, and extracellular acidification, and diminished nitric oxide and reactive oxygen species (ROS) production. Modulated molecular regulators are suggested to play a crucial role as curcumin pretreatment also prevented the onset of chemoresistance by high glucose. High glucose instigated suppression in the intracellular accumulation of anticancer drug doxorubicin and drug-induced chromatin compactness along with declined expression of drug efflux pump MDR-1 and transcription factors and signal transducers governing the survival, aggressiveness, and apoptotic cell death (p53, HIF-1α, mTOR, MYC, STAT3). Curcumin alleviated the suppression of drug retention and nuclear condensation along with hindering the high glucose-induced alterations in transcription factors and signal transducers. High glucose-driven resistance in cancer cells was associated with elevated expression of metabolic enzymes HKII, PFK1, GAPDH, PKM2, LDH-A, IDH3A, and FASN. Metabolite transporters and receptors (GLUT-1, MCT-1, MCT-4, and HCAR-1) were also found upregulated in high glucose exposed HepG2 cells. Curcumin inhibited the elevated expression of these enzymes, transporters, and receptors in cancer cells. Curcumin also uplifted the SDH expression, which was inhibited in high glucose condition. Taken together, the findings of the present investigation first time demonstrate the ability of curcumin against high glucose-induced chemoresistance, along with its molecular mechanism. This will have implication in therapeutic management of malignancies in diabetic conditions.
Collapse
Affiliation(s)
- Vivek Kumar Soni
- Department of Biotechnology, Guru Ghasidas Vishwavidyalaya, Bilaspur, India
| | - Arundhati Mehta
- Department of Biotechnology, Guru Ghasidas Vishwavidyalaya, Bilaspur, India
| | | | - Vikas Chandra
- Department of Biotechnology, Guru Ghasidas Vishwavidyalaya, Bilaspur, India
| | - Dhananjay Shukla
- Department of Biotechnology, Guru Ghasidas Vishwavidyalaya, Bilaspur, India
| | - Ajay Kumar
- Department of Zoology, Banaras Hindu University, Varanasi, India
| | | |
Collapse
|
42
|
Bisht VS, Giri K, Kumar D, Ambatipudi K. Oxygen and metabolic reprogramming in the tumor microenvironment influences metastasis homing. Cancer Biol Ther 2021; 22:493-512. [PMID: 34696706 DOI: 10.1080/15384047.2021.1992233] [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: 10/20/2022] Open
Abstract
Tumor metastasis is the leading cause of cancer mortality, often characterized by abnormal cell growth and invasion to distant organs. The cancer invasion due to epithelial to mesenchymal transition is affected by metabolic and oxygen availability in the tumor-associated micro-environment. A precise alteration in oxygen and metabolic signaling between healthy and metastatic cells is a substantial probe for understanding tumor progression and metastasis. Molecular heterogeneity in the tumor microenvironment help to sustain the metastatic cell growth during their survival shift from low to high metabolic-oxygen-rich sites and reinforces the metastatic events. This review highlighted the crucial role of oxygen and metabolites in metastatic progression and exemplified the role of metabolic rewiring and oxygen availability in cancer cell adaptation. Furthermore, we have also addressed potential applications of altered oxygen and metabolic networking with tumor type that could be a signature pattern to assess tumor growth and chemotherapeutics efficacy in managing cancer metastasis.
Collapse
Affiliation(s)
- Vinod S Bisht
- Department of Biosciences and Bioengineering, Indian Institute of Technology Roorkee, Roorkee, India
| | - Kuldeep Giri
- Department of Biosciences and Bioengineering, Indian Institute of Technology Roorkee, Roorkee, India
| | - Deepak Kumar
- Department of Cancer Biology, Central Drug Research Institute, Lucknow, India.,Academy of Scientific & Innovative Research, New Delhi, India
| | - Kiran Ambatipudi
- Department of Biosciences and Bioengineering, Indian Institute of Technology Roorkee, Roorkee, India
| |
Collapse
|
43
|
Hua H, Zhang H, Chen J, Wang J, Liu J, Jiang Y. Targeting Akt in cancer for precision therapy. J Hematol Oncol 2021; 14:128. [PMID: 34419139 PMCID: PMC8379749 DOI: 10.1186/s13045-021-01137-8] [Citation(s) in RCA: 144] [Impact Index Per Article: 36.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2021] [Accepted: 08/03/2021] [Indexed: 02/08/2023] Open
Abstract
Biomarkers-guided precision therapeutics has revolutionized the clinical development and administration of molecular-targeted anticancer agents. Tailored precision cancer therapy exhibits better response rate compared to unselective treatment. Protein kinases have critical roles in cell signaling, metabolism, proliferation, survival and migration. Aberrant activation of protein kinases is critical for tumor growth and progression. Hence, protein kinases are key targets for molecular targeted cancer therapy. The serine/threonine kinase Akt is frequently activated in various types of cancer. Activation of Akt promotes tumor progression and drug resistance. Since the first Akt inhibitor was reported in 2000, many Akt inhibitors have been developed and evaluated in either early or late stage of clinical trials, which take advantage of liquid biopsy and genomic or molecular profiling to realize personalized cancer therapy. Two inhibitors, capivasertib and ipatasertib, are being tested in phase III clinical trials for cancer therapy. Here, we highlight recent progress of Akt signaling pathway, review the up-to-date data from clinical studies of Akt inhibitors and discuss the potential biomarkers that may help personalized treatment of cancer with Akt inhibitors. In addition, we also discuss how Akt may confer the vulnerability of cancer cells to some kinds of anticancer agents.
Collapse
Affiliation(s)
- Hui Hua
- State Key Laboratory of Biotherapy, Laboratory of Stem Cell Biology, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, China
| | - Hongying Zhang
- State Key Laboratory of Biotherapy, Laboratory of Oncogene, Cancer Center, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Jingzhu Chen
- State Key Laboratory of Biotherapy, Laboratory of Oncogene, Cancer Center, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Jiao Wang
- School of Basic Medicine, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Jieya Liu
- State Key Laboratory of Biotherapy, Laboratory of Oncogene, Cancer Center, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Yangfu Jiang
- State Key Laboratory of Biotherapy, Laboratory of Oncogene, Cancer Center, West China Hospital, Sichuan University, Chengdu, 610041, China.
| |
Collapse
|
44
|
Lu J, Wang Y, Hu Y, Yang B. Lnc-H19 enhances anaerobic glycolysis of keloid fibroblasts by targeting the miR-214-5p/FGF2 axis. Burns 2021:S0305-4179(21)00200-X. [PMID: 34887123 DOI: 10.1016/j.burns.2021.07.015] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2020] [Revised: 07/23/2021] [Accepted: 07/27/2021] [Indexed: 01/12/2023]
Abstract
Long non-coding RNA (lncRNA) H19 has been demonstrated as vital regulator in tumors. However, whether lnc-H19 mediated the development of keloid fibroblasts (KD) was unknown, this study was aimed to clarify the role and molecular mechanisms of lnc-H19 in KD. We have investigated the expression levels of lnc-H19, miR-214-5p and fibroblast growth factor 2 (FGF2) in KD skin samples and normal skin tissues as well as matched cells by real-time quantitative polymerase chain reaction (RT-qPCR) assay. The glycolysis ability of keloid fibroblasts was assessed by measuring glucose consumption, lactate production, and ATP level. The western blot assay was used to assay the expression levels of FGF2 and hexokinase 2 (HK2). Migration and invasion were analyzed by transwell in keloid fibroblasts. The bioinformatics database and dual-luciferase reporter assay were used to search and identify the target of miR-214-5p and lnc-H19. Lnc-H19 was overexpressed in KD tissues and keloid fibroblasts than normal skin tissues and normal fibroblasts, respectively. Small interfering RNA of lnc-H19 treatment markedly inhibited glycolysis, migration and invasion of keloid fibroblasts exposed to hypoxia, which was reserved by silencing of miR-214-5p or upregulation of FGF2. Mechanistically, lnc-H19 regulated KD development by regulation of miR-214-5p/FGF2 axis. In summary, lnc-H19 may exert regulatory functions in KD by targeting miR-214-5p/FGF2 axis, further regulated glycolysis, migration and invasion in keloid fibroblasts exposed to hypoxia, which might be a potential marker of KD diagnosis or progression.
Collapse
Affiliation(s)
- Junxu Lu
- Department of Plastic Surgery, Weifang Medical University, Weifang, 261000, Shandong, China
| | - Yuanbo Wang
- Department of Plastic Surgery, Weifang Medical University, Weifang, 261000, Shandong, China
| | - Yanuan Hu
- Department of Plastic Surgery, Weifang Medical University, Weifang, 261000, Shandong, China
| | - Biaobing Yang
- Department of Plastic Surgery, Weifang Medical University, Weifang, 261000, Shandong, China.
| |
Collapse
|
45
|
Traxler L, Lagerwall J, Eichhorner S, Stefanoni D, D'Alessandro A, Mertens J. Metabolism navigates neural cell fate in development, aging and neurodegeneration. Dis Model Mech 2021; 14:dmm048993. [PMID: 34345916 PMCID: PMC8353098 DOI: 10.1242/dmm.048993] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
An uninterrupted energy supply is critical for the optimal functioning of all our organs, and in this regard the human brain is particularly energy dependent. The study of energy metabolic pathways is a major focus within neuroscience research, which is supported by genetic defects in the oxidative phosphorylation mechanism often contributing towards neurodevelopmental disorders and changes in glucose metabolism presenting as a hallmark feature in age-dependent neurodegenerative disorders. However, as recent studies have illuminated roles of cellular metabolism that span far beyond mere energetics, it would be valuable to first comprehend the physiological involvement of metabolic pathways in neural cell fate and function, and to subsequently reconstruct their impact on diseases of the brain. In this Review, we first discuss recent evidence that implies metabolism as a master regulator of cell identity during neural development. Additionally, we examine the cell type-dependent metabolic states present in the adult brain. As metabolic states have been studied extensively as crucial regulators of malignant transformation in cancer, we reveal how knowledge gained from the field of cancer has aided our understanding in how metabolism likewise controls neural fate determination and stability by directly wiring into the cellular epigenetic landscape. We further summarize research pertaining to the interplay between metabolic alterations and neurodevelopmental and psychiatric disorders, and expose how an improved understanding of metabolic cell fate control might assist in the development of new concepts to combat age-dependent neurodegenerative diseases, particularly Alzheimer's disease.
Collapse
Affiliation(s)
- Larissa Traxler
- Neural Aging Laboratory, Institute of Molecular Biology, CMBI, Leopold-Franzens-University Innsbruck, Tyrol 6020, Austria
| | - Jessica Lagerwall
- Neural Aging Laboratory, Institute of Molecular Biology, CMBI, Leopold-Franzens-University Innsbruck, Tyrol 6020, Austria
| | - Sophie Eichhorner
- Neural Aging Laboratory, Institute of Molecular Biology, CMBI, Leopold-Franzens-University Innsbruck, Tyrol 6020, Austria
| | - Davide Stefanoni
- Department of Biochemistry and Molecular Genetics, University of Colorado Denver, Aurora, CO 80045, USA
| | - Angelo D'Alessandro
- Department of Biochemistry and Molecular Genetics, University of Colorado Denver, Aurora, CO 80045, USA
| | - Jerome Mertens
- Neural Aging Laboratory, Institute of Molecular Biology, CMBI, Leopold-Franzens-University Innsbruck, Tyrol 6020, Austria
- Laboratory of Genetics, The Salk Institute for Biological Studies, La Jolla, CA 92037, USA
| |
Collapse
|
46
|
Liu KX, Everdell E, Pal S, Haas-Kogan DA, Milligan MG. Harnessing Lactate Metabolism for Radiosensitization. Front Oncol 2021; 11:672339. [PMID: 34367959 PMCID: PMC8343095 DOI: 10.3389/fonc.2021.672339] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2021] [Accepted: 07/12/2021] [Indexed: 11/13/2022] Open
Abstract
Cancer cells rewire their metabolism to promote cell proliferation, invasion, and metastasis. Alterations in the lactate pathway have been characterized in diverse cancers, correlate with outcomes, and lead to many downstream effects, including decreasing oxidative stress, promoting an immunosuppressive tumor microenvironment, lipid synthesis, and building chemo- or radio-resistance. Radiotherapy is a key modality of treatment for many cancers and approximately 50% of patients with cancer will receive radiation for cure or palliation; thus, overcoming radio-resistance is important for improving outcomes. Growing research suggests that important molecular controls of the lactate pathway may serve as novel therapeutic targets and in particular, radiosensitizers. In this mini-review, we will provide an overview of lactate metabolism in cancer, discuss three important contributors to lactate metabolism (lactate dehydrogenase, monocarboxylate transporters, and mitochondrial pyruvate carrier), and present data that inhibition of these three pathways can lead to radiosensitization. Future research is needed to further understand critical regulators of lactate metabolism and explore clinical safety and efficacy of inhibitors of lactate dehydrogenase, monocarboxylate transporters, and mitochondrial pyruvate carrier alone and in combination with radiation.
Collapse
Affiliation(s)
- Kevin X Liu
- Department of Radiation Oncology, Brigham and Women's Hospital, Dana-Farber Cancer Institute, Boston Children's Hospital, Harvard Medical School, Boston, MA, United States
| | | | - Sharmistha Pal
- Department of Radiation Oncology, Dana-Farber Cancer Institute, Boston, MA, United States
| | - Daphne A Haas-Kogan
- Department of Radiation Oncology, Brigham and Women's Hospital, Dana-Farber Cancer Institute, Boston Children's Hospital, Harvard Medical School, Boston, MA, United States
| | - Michael G Milligan
- Department of Radiation Oncology, Brigham and Women's Hospital, Dana-Farber Cancer Institute, Boston Children's Hospital, Harvard Medical School, Boston, MA, United States
| |
Collapse
|
47
|
Pawestri AR, Arjkongharn N, Suvannaboon R, Tuekprakhon A, Srimuninnimit V, Udompunthurak S, Atchaneeyasakul LO, Koolvisoot A, Trinavarat A. Autoantibody profiles and clinical association in Thai patients with autoimmune retinopathy. Sci Rep 2021; 11:15047. [PMID: 34294798 PMCID: PMC8298708 DOI: 10.1038/s41598-021-94377-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2021] [Accepted: 07/09/2021] [Indexed: 11/10/2022] Open
Abstract
Autoimmune retinopathy (AIR) is a rare immune-mediated inflammation of the retina. The autoantibodies against retinal proteins and glycolytic enzymes were reported to be involved in the pathogenesis. This retrospective cohort study assessed the antiretinal autoantibody profiles and their association with clinical outcomes of AIR patients in Thailand. We included 44 patients, 75% were females, with the overall median age of onset of 48 (17-74, IQR 40-55.5) years. Common clinical presentations were nyctalopia (65.9%), blurred vision (52.3%), constricted visual field (43.2%), and nonrecordable electroretinography (65.9%). Underlying malignancy and autoimmune diseases were found in 2 and 12 female patients, respectively. We found 41 autoantibodies, with anti-α-enolase (65.9%) showing the highest prevalence, followed by anti-CAII (43.2%), anti-aldolase (40.9%), and anti-GAPDH (36.4%). Anti-aldolase was associated with male gender (P = 0.012, OR 7.11, 95% CI 1.54-32.91). Anti-CAII showed significant association with age of onset (P = 0.025, 95% CI - 17.28 to - 1.24), while anti-α-enolase (P = 0.002, OR 4.37, 95% CI 1.83-10.37) and anti-GAPDH (P = 0.001, OR 1.87, 95% CI 1.32-2.64) were significantly associated with nonrecordable electroretinography. Association between the antibody profiles and clinical outcomes may be used to direct and adjust the treatment plans and provide insights in the pathogenesis of AIR.
Collapse
Affiliation(s)
| | - Niracha Arjkongharn
- Department of Ophthalmology, Faculty of Medicine Siriraj Hospital, Mahidol University, 2 Wanglang Road, Bangkok Noi, Bangkok, 10700, Thailand
| | - Ragkit Suvannaboon
- Department of Ophthalmology, Faculty of Medicine Siriraj Hospital, Mahidol University, 2 Wanglang Road, Bangkok Noi, Bangkok, 10700, Thailand
- Research Division, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok, Thailand
| | - Aekkachai Tuekprakhon
- Department of Ophthalmology, Faculty of Medicine Siriraj Hospital, Mahidol University, 2 Wanglang Road, Bangkok Noi, Bangkok, 10700, Thailand
- Nuffield Department of Medicine, Welcome Center for Human Genetics, University of Oxford, Oxford, UK
| | - Vichien Srimuninnimit
- Division of Medical Oncology, Department of Medicine, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok, Thailand
| | - Suthipol Udompunthurak
- Clinical Epidemiology Division, Siriraj Medical Research Center, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok, Thailand
| | - La-Ongsri Atchaneeyasakul
- Department of Ophthalmology, Faculty of Medicine Siriraj Hospital, Mahidol University, 2 Wanglang Road, Bangkok Noi, Bangkok, 10700, Thailand
| | - Ajchara Koolvisoot
- Division of Rheumatology, Department of Medicine, Faculty of Medicine Siriraj Hospital, Mahidol University, 2 Wanglang Road, Bangkok Noi, Bangkok, 10700, Thailand.
| | - Adisak Trinavarat
- Department of Ophthalmology, Faculty of Medicine Siriraj Hospital, Mahidol University, 2 Wanglang Road, Bangkok Noi, Bangkok, 10700, Thailand.
| |
Collapse
|
48
|
Understanding the Central Role of Citrate in the Metabolism of Cancer Cells and Tumors: An Update. Int J Mol Sci 2021; 22:ijms22126587. [PMID: 34205414 PMCID: PMC8235534 DOI: 10.3390/ijms22126587] [Citation(s) in RCA: 74] [Impact Index Per Article: 18.5] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2021] [Revised: 06/14/2021] [Accepted: 06/16/2021] [Indexed: 02/07/2023] Open
Abstract
Citrate plays a central role in cancer cells’ metabolism and regulation. Derived from mitochondrial synthesis and/or carboxylation of α-ketoglutarate, it is cleaved by ATP-citrate lyase into acetyl-CoA and oxaloacetate. The rapid turnover of these molecules in proliferative cancer cells maintains a low-level of citrate, precluding its retro-inhibition on glycolytic enzymes. In cancer cells relying on glycolysis, this regulation helps sustain the Warburg effect. In those relying on an oxidative metabolism, fatty acid β-oxidation sustains a high production of citrate, which is still rapidly converted into acetyl-CoA and oxaloacetate, this latter molecule sustaining nucleotide synthesis and gluconeogenesis. Therefore, citrate levels are rarely high in cancer cells. Resistance of cancer cells to targeted therapies, such as tyrosine kinase inhibitors (TKIs), is frequently sustained by aerobic glycolysis and its key oncogenic drivers, such as Ras and its downstream effectors MAPK/ERK and PI3K/Akt. Remarkably, in preclinical cancer models, the administration of high doses of citrate showed various anti-cancer effects, such as the inhibition of glycolysis, the promotion of cytotoxic drugs sensibility and apoptosis, the neutralization of extracellular acidity, and the inhibition of tumors growth and of key signalling pathways (in particular, the IGF-1R/AKT pathway). Therefore, these preclinical results support the testing of the citrate strategy in clinical trials to counteract key oncogenic drivers sustaining cancer development and resistance to anti-cancer therapies.
Collapse
|
49
|
Liu B, Zhang Y, Suo J. Increased Expression of PDK4 Was Displayed in Gastric Cancer and Exhibited an Association With Glucose Metabolism. Front Genet 2021; 12:689585. [PMID: 34220962 PMCID: PMC8248380 DOI: 10.3389/fgene.2021.689585] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2021] [Accepted: 05/12/2021] [Indexed: 01/15/2023] Open
Abstract
Previous studies reported that pyruvate dehydrogenase kinase 4 (PDK4) is closely related to diabetes, heart disease, and carcinomas. Nevertheless, the role of PDK4 in gastric cancer (GC) occurrence and development is yet poorly understood. Our experiments were taken to evaluate PDK4's function in GC. The Cancer Genome Atlas tumor genome map database was employed to validate the levels of PDK family in different grades and stages of GC. The survival ratio of PDK families in GC was detected by the Kaplan-Meier plotter database. The links existing in the expression of PDK family and the level of tumor-infiltrating immune cells were investigated by tumor immunity assessment resource (TIMER). PDK4-associated signal pathways in GC were analyzed by the Kyoto Encyclopedia of Genes and Genomes pathway analysis. PDK4 mRNA level in the GC cells was measured by qRT-PCR. Cell counting kit-8 and Transwell assays were separately carried out to evaluate PDK4-induced influence on GC cell proliferation, migration, and invasion. Our data suggested that GC cells highly expressed PDK4, and PDK4 expression presented a significant relation with the staging, grade, and survival rate of GC. PDK4 expression presented a positive correlation with the types of different infiltrating immune cells, comprising B cells, CD4+ T cells, and dendritic cells. Meanwhile, PDK4 expression exhibited a strong association with macrophages. Survival analysis revealed that the expression of PDK4 displayed a relationship with the prognosis of patients. Therefore, PDK4 was liable to be a biomarker for prognosis. Our results further displayed that PDK4 might modulate the glycolysis level in GC cells, and its expression was associated with GC cell proliferation, migration, and invasion. These data may provide insights into designing a new treatment strategy for GC.
Collapse
Affiliation(s)
| | | | - Jian Suo
- Department of Gastrocolorectal Surgery, The First Hospital of Jilin University, Changchun, China
| |
Collapse
|
50
|
Mir MA, Mehraj U, Sheikh BA. Recent Advances in Chemotherapeutic Implications of Deguelin: A Plant-Derived Retinoid. ACTA ACUST UNITED AC 2021. [DOI: 10.2174/2210315510666200128125950] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Deguelin, a plant retinoid has emerged to be a promising therapeutic agent in the treatment
of different cancers. Recent studies demonstrate that deguelin has potential as an angiogenesis
antagonist in malignant and endothelial cells by specifically targeting HGF-c-Met and VEGFVEGFR
pathways. It is reported to have profound therapeutic effects in pancreatic cancer by inactivation
of the hedgehog (Hh) signalling pathway and suppresses the expression of matrix metalloproteinases
such as MMP-2 and MMP-9. The basic underlying mechanisms for deguelin mediated anti-
NSCLC effects were uncovered through its induction of elevated intracellular Reactive Oxygen Species
(ROS) levels and suppression of the PI3K /Akt-HK2 signalling pathway. Deguelin induces cell
apoptosis by targeting various pathways most notably regulating the expression of galectin-1 and
binding directly to anti-apoptotic Bcl-2 (B-cell lymphoma 2), Bcl-xl (B-cell lymphoma-extralarge)
and Mcl-1 (Myeloid Cell Leukemia Sequence 1) in the hydrophobic grooves thereby liberating BAD
and BAX from binding with these proteins. These results derived from the effect of Deguelin on various
cancer cell lines have further elucidated its role as a novel anti-tumorigenic agent targeting angiogenesis,
apoptosis, cell proliferation and migration for cancer chemoprevention. In this review, an
attempt has been made to highlight the potential therapeutic effects of Deguelin in destroying the
cancer cells by inhibiting various tumour promoting pathways and its uses as a therapeutic agent
alone or in combination.
Collapse
Affiliation(s)
- Manzoor A. Mir
- Department of Bioresources, School of Biological Sciences, University of Kashmir, Srinagar-190006, India
| | - Umar Mehraj
- Department of Bioresources, School of Biological Sciences, University of Kashmir, Srinagar-190006, India
| | - Bashir A. Sheikh
- Department of Bioresources, School of Biological Sciences, University of Kashmir, Srinagar-190006, India
| |
Collapse
|