1
|
Shan W, Zhang SL, Assaraf YG, Tam KY. Combined inhibition of hexokinase 2 and pyruvate dehydrogenase surmounts SHP2 inhibitor resistance in non-small cell lung cancer with hybrid metabolic state harboring KRAS Q61H mutation. Biochim Biophys Acta Mol Basis Dis 2025; 1871:167859. [PMID: 40250775 DOI: 10.1016/j.bbadis.2025.167859] [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: 11/02/2024] [Revised: 03/02/2025] [Accepted: 04/15/2025] [Indexed: 04/20/2025]
Abstract
KRAS Q61H is an aggressive oncogenic driver mutation rendering cancer cells drug resistant to SHP2 inhibitors (SHP2i). Some metastatic and chemoresistant non-small cell lung cancer (NSCLC) cells, exhibit a hybrid metabolic state in which both glycolysis and oxidative phosphorylation (OXPHOS) coexist. Hence, we evaluated the in vitro and in vivo efficacy of a combination of hexokinase 2 (HK2) and pyruvate dehydrogenase (PDH) inhibitors, benserazide (Benz) and CPI-613, respectively, against NSCLC NCI-H460 cells harboring the driver KRAS Q61H mutation. This combination synergistically disrupted the hybrid metabolic state, inhibited NCI-H460 cell proliferation in vitro, and markedly suppressed tumor growth in NCI-H460 cell xenograft model in mice. The molecular basis underlying this antitumor activity was apparently due to suppression of SHP2/SOS1/RAS/MAPK signaling pathways, leading to enhanced apoptosis. Moreover, this drug combination restored the sensitivity to SHP2i. Consistently, SHP2 overexpression in NCI-H460 cells abrogated the antitumor activity of this drug combination. These findings reveal that the combination of Benz and CPI-613 targets the metabolic vulnerability of KRAS Q61H mutant-bearing NSCLC tumors. These results offer a combination therapeutic strategy for the possible treatment of cancer cells displaying a hybrid metabolic state, thereby surmounting chemoresistance.
Collapse
Affiliation(s)
- Wenying Shan
- Faculty of Health Sciences, University of Macau, Taipa, Macau
| | - Shao-Lin Zhang
- School of Pharmaceutical Sciences, Chongqing Key Laboratory of Natural Product Synthesis and Drug Research, Chongqing University, Chongqing 401331, PR China
| | - Yehuda G Assaraf
- The Fred Wyszkowski Cancer Research Laboratory, Faculty of Biology, Technion-Israel Institute of Technology, Haifa 3200003, Israel.
| | - Kin Yip Tam
- Faculty of Health Sciences, University of Macau, Taipa, Macau.
| |
Collapse
|
2
|
Yang K, Yue B, Tian H, Wang L, Yang X, Zhang W. Paeonol inhibits the Glycolysis in oral squamous cell carcinoma though suppressing NAT10-mediated ac 4C modification. BMC Cancer 2025; 25:629. [PMID: 40197308 PMCID: PMC11977892 DOI: 10.1186/s12885-025-14000-7] [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: 11/27/2024] [Accepted: 03/24/2025] [Indexed: 04/10/2025] Open
Abstract
BACKGROUND Oral squamous cell carcinoma (OSCC) is the most common malignant tumor of the oral and maxillofacial regions. Paeonol, derived from Moutan Cortex, has diverse pharmacological effects including anti-inflammatory and anticancer activities. The N-acetyltransferase 10 (NAT10)-mediated N4-acetylcytidine (ac4C) modification is a newly discovered RNA epigenetic mechanism. This study aimed to investigate the role of paeonol in OSCC and its underlying mechanisms of action. METHODS Cell viability and migration were assessed using a cell counting kit-8 and transwell migration assays. Glycolysis-related indices were detected using commercial kits. The interaction between NAT10 and hexokinase 2 (HK2) was examined using RNA immunoprecipitation and dual-luciferase reporter assays. A tumor-bearing mouse model was established. RESULTS The results showed that paeonol treatment decreased the viability, migration, and glycolysis of OSCC cells. Moreover, paeonol treatment inhibited NAT10-mediated ac4C modifications in OSCC cells. In addition, NAT10 overexpression upregulates glycolysis and cell migration in OSCC cells. Moreover, NAT10 upregulated ac4C levels of HK2 in OSCC cells. Animal studies have revealed that paeonol treatment decreases OSCC tumor growth. CONCLUSION This study revealed that paeonol inhibited glycolysis and cell migration in OSCC by suppressing the NAT10-mediated ac4C modification of HK2.
Collapse
Affiliation(s)
- Kang Yang
- Department of Pharmacy, Xi'an Hospital of Traditional Chinese Medicine, No.69, Fengcheng 8th Road, Weiyang District, Xi'an, 710021, China
| | - Baosen Yue
- Department of Pharmacy, Xi'an Hospital of Traditional Chinese Medicine, No.69, Fengcheng 8th Road, Weiyang District, Xi'an, 710021, China
| | - Huan Tian
- Department of Pharmacy, Xi'an Hospital of Traditional Chinese Medicine, No.69, Fengcheng 8th Road, Weiyang District, Xi'an, 710021, China
| | - Lu Wang
- Xi'an Beilin Zhongqingyun Dental Clinic, Xi'an, 710032, China
| | - Xiao Yang
- Department of Pharmacy, Xi'an Hospital of Traditional Chinese Medicine, No.69, Fengcheng 8th Road, Weiyang District, Xi'an, 710021, China
| | - Weiying Zhang
- Department of Pharmacy, Xi'an Hospital of Traditional Chinese Medicine, No.69, Fengcheng 8th Road, Weiyang District, Xi'an, 710021, China.
| |
Collapse
|
3
|
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] [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
|
4
|
Zhang Y, He R, She Z, Yin X, Li X, Yao S, Du M, An S. PBAN regulates sex pheromone biosynthesis by Ca 2+/CaN/ACC and Ca 2+/PKC/HK2 signal pathways in Spodoptera litura. INSECT MOLECULAR BIOLOGY 2025; 34:347-362. [PMID: 39548954 DOI: 10.1111/imb.12976] [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: 07/29/2024] [Accepted: 11/01/2024] [Indexed: 11/18/2024]
Abstract
Sex pheromones emitted by female moths play important roles in mate attraction. The molecular mechanism underlying pheromone biosynthesis activating neuropeptide (PBAN)-regulated sex pheromone biosynthesis has been well elucidated in many moth species, although this mechanism is species-dependent. Spodoptera litura, an important pest, has caused serious economic losses to agricultural production, yet the mechanism for its sex pheromone biosynthesis has not been fully identified. The present study investigates in detail mechanism underlying PBAN-regulated sex pheromone biosynthesis in S. litura. The transcriptome sequencing of S. litura pheromone glands (PGs) was analysed to identify a serial of candidate genes potentially involved in sex pheromone biosynthesis. Further investigation revealed a bimodal pattern in both sex pheromone release and mating frequency. PBAN was found to regulate sex pheromone biosynthesis via its receptor by using Ca2+ as a secondary messenger, as demonstrated by RNA interference and the application of pharmacological inhibitors. Furthermore, PBAN/Ca2+ signalling activated calcineurin (CaN) and acetyl-CoA carboxylase (ACC), which mediated sex pheromone biosynthesis in response to PBAN stimulation. Mostly importantly, hexokinase 2 (HK2) was confirmed to be activated by PBAN/PBANR /Ca2+/PKC signalling via phosphorylation at two specific sites (ser423 and ser434 sites of HK2). Overall, our findings shed light on the intricate processes involved in sex pheromone production in S. litura, in which PBAN regulates sex pheromone biosynthesis through PBAN/PBANR/Ca2+/CaN/ACC and PBAN/PBANR/Ca2+/PKC/HK2 signalling pathways. These insights significantly contribute to our comprehension of the specific mechanisms underlying sex pheromone biosynthesis in this moth species.
Collapse
Affiliation(s)
- Yao Zhang
- State Key Laboratory of Wheat and Maize Crop Science/Henan International Laboratory for Green Pest Control/College of Plant Protection, Henan Agricultural University, Zhengzhou, China
| | - Ruolan He
- State Key Laboratory of Wheat and Maize Crop Science/Henan International Laboratory for Green Pest Control/College of Plant Protection, Henan Agricultural University, Zhengzhou, China
| | - Zelong She
- State Key Laboratory of Wheat and Maize Crop Science/Henan International Laboratory for Green Pest Control/College of Plant Protection, Henan Agricultural University, Zhengzhou, China
| | - Xinming Yin
- State Key Laboratory of Wheat and Maize Crop Science/Henan International Laboratory for Green Pest Control/College of Plant Protection, Henan Agricultural University, Zhengzhou, China
| | - Xiang Li
- State Key Laboratory of Wheat and Maize Crop Science/Henan International Laboratory for Green Pest Control/College of Plant Protection, Henan Agricultural University, Zhengzhou, China
| | - Shuangyan Yao
- State Key Laboratory of Wheat and Maize Crop Science/Henan International Laboratory for Green Pest Control/College of Plant Protection, Henan Agricultural University, Zhengzhou, China
| | - Mengfang Du
- State Key Laboratory of Wheat and Maize Crop Science/Henan International Laboratory for Green Pest Control/College of Plant Protection, Henan Agricultural University, Zhengzhou, China
| | - Shiheng An
- State Key Laboratory of Wheat and Maize Crop Science/Henan International Laboratory for Green Pest Control/College of Plant Protection, Henan Agricultural University, Zhengzhou, China
| |
Collapse
|
5
|
Gore M, Kabekkodu SP, Chakrabarty S. Exploring the metabolic alterations in cervical cancer induced by HPV oncoproteins: From mechanisms to therapeutic targets. Biochim Biophys Acta Rev Cancer 2025; 1880:189292. [PMID: 40037419 DOI: 10.1016/j.bbcan.2025.189292] [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: 09/12/2024] [Revised: 02/24/2025] [Accepted: 02/25/2025] [Indexed: 03/06/2025]
Abstract
The role of human Papillomavirus (HPV) in metabolic reprogramming is implicated in the development and progression of cervical cancer. During carcinogenesis, cancer cells modify various metabolic pathways to generate energy and sustain their growth and development. Cervical cancer, one of the most prevalent malignancies affecting women globally, involves metabolic alterations such as increased glycolysis, elevated lactate production, and lipid accumulation. The oncoproteins, primarily E6 and E7, which are encoded by high-risk HPVs, facilitate the accumulation of several cancer markers, promoting not only the growth and development of cancer but also metastasis, immune evasion, and therapy resistance. HPV oncoproteins interact with cellular MYC (c-MYC), retinoblastoma protein (pRB), p53, and hypoxia-inducible factor 1α (HIF-1α), leading to the induction of metabolic reprogramming and favour the Warburg effect. Metabolic reprogramming enables HPV to persist for an extended period and accelerates the progression of cervical cancer. This review summarizes the role of HPV oncoproteins in metabolic reprogramming and their contributions to the development and progression of cervical cancer. Additionally, this review provides insights into how metabolic reprogramming opens avenues for novel therapeutic strategies, including the discovery of new and repurposed drugs that could be applied to treat cervical cancer.
Collapse
Affiliation(s)
- Mrudula Gore
- Department of Cell and Molecular Biology, Manipal School of Life Sciences, Manipal Academy of Higher Education, Manipal 576104, Karnataka, India
| | - Shama Prasada Kabekkodu
- Department of Cell and Molecular Biology, Manipal School of Life Sciences, Manipal Academy of Higher Education, Manipal 576104, Karnataka, India.
| | - Sanjiban Chakrabarty
- Department of Public Health Genomics, Manipal School of Life Sciences, Manipal Academy of Higher Education, Manipal 576104, Karnataka, India
| |
Collapse
|
6
|
Jiang X, Ge X, Huang Y, Xie F, Chen C, Wang Z, Tao W, Zeng S, Lv L, Zhan Y, Bao L. Drug resistance in TKI therapy for hepatocellular carcinoma: Mechanisms and strategies. Cancer Lett 2025; 613:217472. [PMID: 39832650 DOI: 10.1016/j.canlet.2025.217472] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2024] [Revised: 01/14/2025] [Accepted: 01/16/2025] [Indexed: 01/22/2025]
Abstract
Tyrosine kinase inhibitors (TKIs) are such as sorafenib the first-line therapeutic drugs for patients with advanced hepatocellular carcinoma. However, patients with TKI-resistant advanced liver cancer are insensitive to TKI treatment, resulting in limited survival benefits. This paper comprehensively reviewed the mechanisms underlying TKI resistance in hepatocytes, investigating activation of tumor signaling pathways, epigenetic regulation, tumor microenvironment, and metabolic reprogramming. Based on resistance mechanisms, it also reviews preclinical and clinical studies of drug resistance strategies and summarizes targeted therapy combined with immunotherapy currently in investigational clinical trials. Understanding the interactions and clinical studies of these resistance mechanisms offers new hope for improving and prolonging patient survival.
Collapse
Affiliation(s)
- Xue Jiang
- Department of Pharmacy, Shanghai Eastern Hepatobiliary Surgery Hospital, Shanghai, 200438, China.
| | - Xiaoying Ge
- Department of Pharmacy, Shanghai Eastern Hepatobiliary Surgery Hospital, Shanghai, 200438, China.
| | - Yueying Huang
- Department of Pharmacy, Shanghai Eastern Hepatobiliary Surgery Hospital, Shanghai, 200438, China.
| | - Fangyuan Xie
- Department of Pharmacy, Shanghai Eastern Hepatobiliary Surgery Hospital, Shanghai, 200438, China.
| | - Chun Chen
- Department of Pharmacy, Shanghai Eastern Hepatobiliary Surgery Hospital, Shanghai, 200438, China.
| | - Zijun Wang
- Department of Pharmacy, Shanghai Eastern Hepatobiliary Surgery Hospital, Shanghai, 200438, China.
| | - Wanru Tao
- Department of Pharmacy, Shanghai Eastern Hepatobiliary Surgery Hospital, Shanghai, 200438, China.
| | - Sailiang Zeng
- Department of Pharmacy, Shanghai Eastern Hepatobiliary Surgery Hospital, Shanghai, 200438, China.
| | - Lei Lv
- Department of Pharmacy, Shanghai Eastern Hepatobiliary Surgery Hospital, Shanghai, 200438, China.
| | - Yangyang Zhan
- Department of Pharmacy, Shanghai Eastern Hepatobiliary Surgery Hospital, Shanghai, 200438, China.
| | - Leilei Bao
- Department of Pharmacy, Shanghai Eastern Hepatobiliary Surgery Hospital, Shanghai, 200438, China.
| |
Collapse
|
7
|
Yang L, Shi W, Li D, Shen Y, Li N, Meng Z. Study on the mechanism of 17-Hydroxy-jolkinolide B on anaplastic thyroid cancer cell. Am J Med Sci 2025; 369:405-412. [PMID: 39326738 DOI: 10.1016/j.amjms.2024.09.004] [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: 04/19/2024] [Revised: 09/13/2024] [Accepted: 09/16/2024] [Indexed: 09/28/2024]
Abstract
BACKGROUND Anaplastic thyroid cancer (ATC) has a dismal prognosis, and the optimal treatment has not yet been confirmed. Euphorbia fischeriana Steud has been proven to exhibit pharmacological properties, including various antitumor effects, that can be used to treat numerous diseases and has been used to treat cancer. 17-Hydroxy-jolkinolide B (17-HJB) is one of the major diterpenoids produced from plants, but little research has investigated how it affects cancer. METHODS MTT assays, glucose and lactate concentration detection, Annexin V-FITC detection via cytometry, and Western blotting were performed to research the mechanism of 17-HJB. RESULTS Cell viability was inhibited in a concentration-dependent manner after 17-HJB treatment. 17-HJB inhibited glucose consumption and lactate production, and the expression of the glucose transporter GLUT1 and proteins associated with glycolysis, HK2, PFK1, and PKM2, was significantly downregulated. 17-HJB induced apoptosis, and the expression of signaling proteins related to apoptosis, such as Caspase-3 and cleaved Caspase-3, was upregulated. In vivo, 17-HJB effectively inhibited the growth of ATC tumors. The results of the expression of glycolysis-related enzyme proteins and apoptosis signaling proteins were consistent with those in vitro. CONCLUSIONS 17-HJB inhibited the growth of ATCs both in vivo and in vitro. The mechanism may be related to the effects on glucose metabolism and the inhibition of aerobic glycolysis. 17-HJB also induced ATC apoptosis.
Collapse
Affiliation(s)
- Lei Yang
- Tianjin Key Laboratory of Acute Abdomen Disease Associated Organ Injury and ITCWM Repair, Tianjin NanKai Hospital, Tianjin Medical University, Tianjin 300100, China
| | - Wanying Shi
- Department of Nuclear Medicine, Tianjin Medical University General Hospital, Tianjin 300052, China; Werner Siemens Imaging Center, Department of Preclinical Imaging and Radiopharmacy, Eberhard KarlsD University of Tuebingen, Tuebingen 72076, Germany
| | - Dihua Li
- Tianjin Key Laboratory of Acute Abdomen Disease Associated Organ Injury and ITCWM Repair, Tianjin NanKai Hospital, Tianjin Medical University, Tianjin 300100, China
| | - Yiming Shen
- Department of Nuclear Medicine, Tianjin Medical University General Hospital, Tianjin 300052, China
| | - Ning Li
- Department of Nuclear Medicine, Tianjin Medical University General Hospital, Tianjin 300052, China
| | - Zhaowei Meng
- Department of Nuclear Medicine, Tianjin Medical University General Hospital, Tianjin 300052, China; Tianjin Key Lab of Functional Imaging & Tianjin Institute of Radiology, Tianjin Medical University General Hospital, Tianjin 300052, China.
| |
Collapse
|
8
|
Andryszkiewicz W, Gąsiorowska J, Kübler M, Kublińska K, Pałkiewicz A, Wiatkowski A, Szwedowicz U, Choromańska A. Glucose Metabolism and Tumor Microenvironment: Mechanistic Insights and Therapeutic Implications. Int J Mol Sci 2025; 26:1879. [PMID: 40076506 PMCID: PMC11900028 DOI: 10.3390/ijms26051879] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2025] [Revised: 02/17/2025] [Accepted: 02/19/2025] [Indexed: 03/14/2025] Open
Abstract
Metabolic reprogramming in cancer cells involves changes in glucose metabolism, glutamine utilization, and lipid production, as well as promoting increased cell proliferation, survival, and immune resistance by altering the tumor microenvironment. Our study analyzes metabolic reprogramming in neoplastically transformed cells, focusing on changes in glucose metabolism, glutaminolysis, and lipid synthesis. Moreover, we discuss the therapeutic potential of targeting cancer metabolism, focusing on key enzymes involved in glycolysis, the pentose phosphate pathway, and amino acid metabolism, including lactate dehydrogenase A, hexokinase, phosphofructokinase and others. The review also highlights challenges such as metabolic heterogeneity, adaptability, and the need for personalized therapies to overcome resistance and minimize adverse effects in cancer treatment. This review underscores the significance of comprehending metabolic reprogramming in cancer cells to engineer targeted therapies, personalize treatment methodologies, and surmount challenges, including metabolic plasticity and therapeutic resistance.
Collapse
Affiliation(s)
- Wiktoria Andryszkiewicz
- Faculty of Medicine, Wroclaw Medical University, Pasteura 1, 50-367 Wroclaw, Poland; (W.A.); (J.G.); (M.K.); (K.K.); (A.P.); (A.W.)
| | - Julia Gąsiorowska
- Faculty of Medicine, Wroclaw Medical University, Pasteura 1, 50-367 Wroclaw, Poland; (W.A.); (J.G.); (M.K.); (K.K.); (A.P.); (A.W.)
| | - Maja Kübler
- Faculty of Medicine, Wroclaw Medical University, Pasteura 1, 50-367 Wroclaw, Poland; (W.A.); (J.G.); (M.K.); (K.K.); (A.P.); (A.W.)
| | - Karolina Kublińska
- Faculty of Medicine, Wroclaw Medical University, Pasteura 1, 50-367 Wroclaw, Poland; (W.A.); (J.G.); (M.K.); (K.K.); (A.P.); (A.W.)
| | - Agata Pałkiewicz
- Faculty of Medicine, Wroclaw Medical University, Pasteura 1, 50-367 Wroclaw, Poland; (W.A.); (J.G.); (M.K.); (K.K.); (A.P.); (A.W.)
| | - Adam Wiatkowski
- Faculty of Medicine, Wroclaw Medical University, Pasteura 1, 50-367 Wroclaw, Poland; (W.A.); (J.G.); (M.K.); (K.K.); (A.P.); (A.W.)
| | - Urszula Szwedowicz
- Department of Molecular and Cellular Biology, Faculty of Pharmacy, Wroclaw Medical University, Borowska 211A, 50-556 Wroclaw, Poland;
| | - Anna Choromańska
- Department of Molecular and Cellular Biology, Faculty of Pharmacy, Wroclaw Medical University, Borowska 211A, 50-556 Wroclaw, Poland;
| |
Collapse
|
9
|
Zhang S, Wang Y, Sun B, Zhu S, Jia Z, Liu L, Liu L. Regulation of Glycolysis by SMAD5 in Glioma Cells: Implications for Tumor Growth and Apoptosis. Neurochem Res 2025; 50:101. [PMID: 39964587 DOI: 10.1007/s11064-025-04352-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2024] [Revised: 02/04/2025] [Accepted: 02/05/2025] [Indexed: 04/26/2025]
Abstract
The Warburg effect serves as a crucial aspect of tumor metabolism, where tumor cells preferentially rely on glycolysis, despite its lower efficiency, over oxidative phosphorylation for energy production even under aerobic conditions. This reprogramming of glucose metabolism confers glioma cells with the capacity for survival and proliferation. Serving as a messenger for regulating transforming growth factor beta, intracellular pH, cell metabolism maintaining cellular bioenergetic homeostasis, SMAD family member 5 (SMAD5) plays a pivotal role in the malignant progression of glioma cells and aerobic glycolysis. Hence, we have identified the expression and function of SMAD5 in human glioma cells, aiming to clarify its role in glycolysis. qRT-PCR and Western blot, reveal that SMAD5 is significantly overexpressed in glioma cells. Knocking down SMAD5 can effectively suppress the proliferation and invasion of glioma cells, while promoting apoptosis, furthermore, downregulation of SMAD5 in vivo has been shown to significantly reduce the growth of xenograft tumors. Conversely, overexpressing SMAD5 enhances the proliferative and invasive capabilities of glioma cells, while suppressing apoptosis. Concurrently, alterations in the expression level of SMAD5 exert an impact on the expression of glucose transporter GLUT1 and crucial enzymes involved in glycolysis, namely HK2 and PKM2, ultimately influencing the glycolytic capability of glioma cells. Specifically, knockdown of SMAD5 suppresses glycolysis, whereas its overexpression enhances glycolytic activity. In conclusion, our data demonstrate that SMAD5 can influence the proliferation, invasion, and apoptosis of glioma cells by modulating glycolysis. This finding holds potential for the development of novel metabolic treatment strategies for glioma.
Collapse
Affiliation(s)
- Shiyang Zhang
- Department of Neurosurgery, The Second Hospital of Hebei Medical University, Shijiazhuang, 050000, China
| | - Yizheng Wang
- Department of Pain and Rehabilitation, The Fourth Hospital of Hebei Medical University, Shijiazhuang, 050000, China
| | - Boyu Sun
- Department of Neurosurgery, The Second Hospital of Hebei Medical University, Shijiazhuang, 050000, China
| | - Siyu Zhu
- Department of Neurosurgery, The Second Hospital of Hebei Medical University, Shijiazhuang, 050000, China
| | - Ziyang Jia
- Department of Neurosurgery, The Second Hospital of Hebei Medical University, Shijiazhuang, 050000, China
| | - Liqiang Liu
- Department of Neurosurgery, The Second Hospital of Hebei Medical University, Shijiazhuang, 050000, China.
| | - Lixin Liu
- Department of Neurology, The Third Hospital of Shijiazhuang City, Shijiazhuang, 050000, China.
| |
Collapse
|
10
|
Dong XM, Chen L, Xu YX, Wu P, Xie T, Liu ZQ. Exploring metabolic reprogramming in esophageal cancer: the role of key enzymes in glucose, amino acid, and nucleotide pathways and targeted therapies. Cancer Gene Ther 2025; 32:165-183. [PMID: 39794467 DOI: 10.1038/s41417-024-00858-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2024] [Revised: 11/08/2024] [Accepted: 11/14/2024] [Indexed: 01/13/2025]
Abstract
Esophageal cancer (EC) is one of the most common malignancies worldwide with the character of poor prognosis and high mortality. Despite significant advancements have been achieved in elucidating the molecular mechanisms of EC, for example, in the discovery of new biomarkers and metabolic pathways, effective treatment options for patients with advanced EC are still limited. Metabolic heterogeneity in EC is a critical factor contributing to poor clinical outcomes. This heterogeneity arises from the complex interplay between the tumor microenvironment and genetic factors of tumor cells, which drives significant metabolic alterations in EC, a process known as metabolic reprogramming. Understanding the mechanisms of metabolic reprogramming is essential for developing new antitumor therapies and improving treatment outcomes. Targeting the distinct metabolic alterations in EC could enable more precise and effective therapies. In this review, we explore the complex metabolic changes in glucose, amino acid, and nucleotide metabolism during the progression of EC, and how these changes drive unique nutritional demands in cancer cells. We also evaluate potential therapies targeting key metabolic enzymes and their clinical applicability. Our work will contribute to enhancing knowledge of metabolic reprogramming in EC and provide new insights and approaches for the clinical treatment of EC.
Collapse
Affiliation(s)
- Xue-Man Dong
- Xiangya School of Pharmaceutical Sciences, Central South University, Changsha, China
- School of Pharmacy, Hangzhou Normal University, Hangzhou, China
| | - Lin Chen
- School of Pharmacy, Hangzhou Normal University, Hangzhou, China
| | - Yu-Xin Xu
- School of Pharmacy, Hangzhou Normal University, Hangzhou, China
| | - Pu Wu
- Xiangya School of Pharmaceutical Sciences, Central South University, Changsha, China
- School of Pharmacy, Hangzhou Normal University, Hangzhou, China
| | - Tian Xie
- School of Pharmacy, Hangzhou Normal University, Hangzhou, China.
| | - Zhao-Qian Liu
- Xiangya School of Pharmaceutical Sciences, Central South University, Changsha, China.
| |
Collapse
|
11
|
Aden D, Sureka N, Zaheer S, Chaurasia JK, Zaheer S. Metabolic Reprogramming in Cancer: Implications for Immunosuppressive Microenvironment. Immunology 2025; 174:30-72. [PMID: 39462179 DOI: 10.1111/imm.13871] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2024] [Revised: 10/07/2024] [Accepted: 10/09/2024] [Indexed: 10/29/2024] Open
Abstract
Cancer is a complex and heterogeneous disease characterised by uncontrolled cell growth and proliferation. One hallmark of cancer cells is their ability to undergo metabolic reprogramming, which allows them to sustain their rapid growth and survival. This metabolic reprogramming creates an immunosuppressive microenvironment that facilitates tumour progression and evasion of the immune system. In this article, we review the mechanisms underlying metabolic reprogramming in cancer cells and discuss how these metabolic alterations contribute to the establishment of an immunosuppressive microenvironment. We also explore potential therapeutic strategies targeting metabolic vulnerabilities in cancer cells to enhance immune-mediated anti-tumour responses. TRIAL REGISTRATION: ClinicalTrials.gov identifier: NCT02044861, NCT03163667, NCT04265534, NCT02071927, NCT02903914, NCT03314935, NCT03361228, NCT03048500, NCT03311308, NCT03800602, NCT04414540, NCT02771626, NCT03994744, NCT03229278, NCT04899921.
Collapse
Affiliation(s)
- Durre Aden
- Department of Pathology, Hamdard Institute of Medical Science and Research, New Delhi, India
| | - Niti Sureka
- Department of Pathology, Vardhman Mahavir Medical College and Safdarjung Hospital, New Delhi, India
| | - Samreen Zaheer
- Department of Radiotherapy, Jawaharlal Nehru Medical College, AMU, Aligarh, India
| | | | - Sufian Zaheer
- Department of Pathology, Vardhman Mahavir Medical College and Safdarjung Hospital, New Delhi, India
| |
Collapse
|
12
|
Singer M, Hamdy R, Elsayed TM, Husseiny MI. The Mechanisms and Therapeutic Implications of Metabolic Communication in the Tumor-Immune Microenvironment. METABOLIC DYNAMICS IN HOST-MICROBE INTERACTION 2025:291-315. [DOI: 10.1007/978-981-96-1305-2_12] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/06/2025]
|
13
|
Kumari S, Gupta S, Jamil A, Tabatabaei D, Karakashev S. Exploring Metabolic Approaches for Epithelial Ovarian Cancer Therapy. J Cell Physiol 2025; 240:e31495. [PMID: 39676338 DOI: 10.1002/jcp.31495] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2024] [Revised: 10/21/2024] [Accepted: 11/18/2024] [Indexed: 12/17/2024]
Abstract
Epithelial ovarian cancer (EOC) has the highest mortality rate among malignant tumors of the female reproductive system and the lowest survival rate. This poor prognosis is due to the aggressive nature of EOC, its late-stage diagnosis, and the tumor's ability to adapt to stressors through metabolic reprogramming. EOC cells sustain their rapid proliferation by altering the uptake, utilization, and regulation of carbohydrates, lipids, and amino acids. These metabolic changes support tumor growth and contribute to metastasis, chemotherapy resistance, and immune evasion. Targeting these metabolic vulnerabilities has shown promise in preclinical studies, with some therapies advancing to clinical trials. However, challenges remain due to tumor heterogeneity, adaptive resistance mechanisms, and the influence of the tumor microenvironment. This review provides a comprehensive summary of metabolic targets for EOC treatment and offers an overview of the current landscape of clinical trials focusing on ovarian cancer metabolism. Future efforts should prioritize combination therapies that integrate metabolic inhibitors with immunotherapies or chemotherapy. Advances in precision medicine and multi-omics approaches will be crucial for identifying patient-specific metabolic dependencies and improving outcomes. By addressing these challenges, metabolism-based therapies can significantly transform the treatment of this devastating disease.
Collapse
Affiliation(s)
- Sangeeta Kumari
- Fels Cancer Institute for Personalized Medicine and Department of Cancer and Cellular Biology, Lewis Katz School of Medicine, Temple University, Philadelphia, Pennsylvania, USA
| | - Shraddha Gupta
- Fels Cancer Institute for Personalized Medicine and Department of Cancer and Cellular Biology, Lewis Katz School of Medicine, Temple University, Philadelphia, Pennsylvania, USA
| | - Aisha Jamil
- Fels Cancer Institute for Personalized Medicine and Department of Cancer and Cellular Biology, Lewis Katz School of Medicine, Temple University, Philadelphia, Pennsylvania, USA
| | - Deyana Tabatabaei
- Fels Cancer Institute for Personalized Medicine and Department of Cancer and Cellular Biology, Lewis Katz School of Medicine, Temple University, Philadelphia, Pennsylvania, USA
- Department of Biology, College of Science and Technology, Temple University, Philadelphia, Pennsylvania, USA
| | - Sergey Karakashev
- Fels Cancer Institute for Personalized Medicine and Department of Cancer and Cellular Biology, Lewis Katz School of Medicine, Temple University, Philadelphia, Pennsylvania, USA
- Nuclear Dynamics and Cancer Program, Fox Chase Cancer Center, Philadelphia, Pennsylvania, USA
- Cancer Epigenetics Institute, Fox Chase Cancer Center, Philadelphia, Pennsylvania, USA
| |
Collapse
|
14
|
Chen S, Hu X, Yi X, Deng X, Xiong T, Ou Y, Liu S, Li C, Yan X, Hao L. USP22 Promotes Osteosarcoma Progression by Stabilising β-Catenin and Upregulating HK2 and Glycolysis. J Cell Mol Med 2024; 28:e70239. [PMID: 39661501 PMCID: PMC11633763 DOI: 10.1111/jcmm.70239] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2024] [Revised: 10/26/2024] [Accepted: 11/14/2024] [Indexed: 12/13/2024] Open
Abstract
Osteosarcoma is a primary malignancy that is difficult to treat and is prone to developing resistance to chemotherapy. As such, it is necessary to continuously explore novel therapeutic targets. Ubiquitin-specific protease 22 (USP22) is an ubiquitin-specific protease that has been demonstrated to have potent carcinogenic effects on a variety of cancers and is involved in several biological processes. Studies have demonstrated that reprogramming of glucose metabolism is a major factor in the development and progression of osteosarcoma, and that USP22 is strongly associated with the metabolism of glucose in osteosarcoma. However, it is still unknown how precisely USP22 works in osteosarcoma. To further elucidate the expression and specific molecular mechanisms of USP22 in osteosarcoma. The results of Western blot analysis and quantitative reverse transcription polymerase chain reaction (qRT-PCR) showed that the expression of USP22 in osteosarcoma tissues was significantly higher than that in adjacent healthy tissues. In addition, the expression of USP22 promotes the proliferation of osteosarcoma cells in a glycolytic dependent manner both in vitro and in vivo, while the knockout of USP22 is the opposite. In addition, USP22 knockout reduced the protein expression of β-catenin and hexokinase 2 (HK2) in osteosarcoma cells. In addition, the regulation of HK2 expression induced by USP22 depends on β-catenin. Mechanistically, USP22 regulates HK2 by deubiquitination and stabilising the expression of β-catenin, thereby controlling glycolysis in osteosarcoma cells.
Collapse
Affiliation(s)
- Shenliang Chen
- Department of Orthopedics, the 2nd Affiliated Hospital, Jiangxi Medical CollegeNanchang UniversityNanchangChina
- University of NanchangNanchangChina
| | - Xin Hu
- Jiangxi Pingxiang People's HospitalPingxiangChina
| | - Xuan Yi
- Department of Orthopedics, the 2nd Affiliated Hospital, Jiangxi Medical CollegeNanchang UniversityNanchangChina
| | - Xueqiang Deng
- Department of Orthopedics, the 2nd Affiliated Hospital, Jiangxi Medical CollegeNanchang UniversityNanchangChina
| | - Ting Xiong
- Department of Orthopedics, the 2nd Affiliated Hospital, Jiangxi Medical CollegeNanchang UniversityNanchangChina
- University of NanchangNanchangChina
| | - Yanghuan Ou
- Department of Orthopedics, the 2nd Affiliated Hospital, Jiangxi Medical CollegeNanchang UniversityNanchangChina
- University of NanchangNanchangChina
| | - Shuaigang Liu
- Department of Orthopedics, the 2nd Affiliated Hospital, Jiangxi Medical CollegeNanchang UniversityNanchangChina
| | - Chen Li
- Department of Orthopedics, the 2nd Affiliated Hospital, Jiangxi Medical CollegeNanchang UniversityNanchangChina
| | - Xiaohua Yan
- University of NanchangNanchangChina
- Department of Biochemistry and Molecular Biology, School of Basic Medical SciencesNanchang University Jiangxi Medical CollegeNanchangChina
| | - Liang Hao
- Department of Orthopedics, the 2nd Affiliated Hospital, Jiangxi Medical CollegeNanchang UniversityNanchangChina
| |
Collapse
|
15
|
Liu S, Zhang X, Wang W, Li X, Sun X, Zhao Y, Wang Q, Li Y, Hu F, Ren H. Metabolic reprogramming and therapeutic resistance in primary and metastatic breast cancer. Mol Cancer 2024; 23:261. [PMID: 39574178 PMCID: PMC11580516 DOI: 10.1186/s12943-024-02165-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2024] [Accepted: 10/31/2024] [Indexed: 11/25/2024] Open
Abstract
Metabolic alterations, a hallmark of cancer, enable tumor cells to adapt to their environment by modulating glucose, lipid, and amino acid metabolism, which fuels rapid growth and contributes to treatment resistance. In primary breast cancer, metabolic shifts such as the Warburg effect and enhanced lipid synthesis are closely linked to chemotherapy failure. Similarly, metastatic lesions often display distinct metabolic profiles that not only sustain tumor growth but also confer resistance to targeted therapies and immunotherapies. The review emphasizes two major aspects: the mechanisms driving metabolic resistance in both primary and metastatic breast cancer, and how the unique metabolic environments in metastatic sites further complicate treatment. By targeting distinct metabolic vulnerabilities at both the primary and metastatic stages, new strategies could improve the efficacy of existing therapies and provide better outcomes for breast cancer patients.
Collapse
Affiliation(s)
- Shan Liu
- Department of oncological surgery, Harbin Medical University Cancer Hospital, Harbin, China
| | - Xingda Zhang
- Department of oncological surgery, Harbin Medical University Cancer Hospital, Harbin, China
| | - Wenzheng Wang
- Department of oncological surgery, Harbin Medical University Cancer Hospital, Harbin, China
| | - Xue Li
- Department of oncological surgery, Harbin Medical University Cancer Hospital, Harbin, China
| | - Xue Sun
- Department of oncological surgery, Harbin Medical University Cancer Hospital, Harbin, China
| | - Yuqian Zhao
- Department of oncological surgery, Harbin Medical University Cancer Hospital, Harbin, China
| | - Qi Wang
- Department of oncological surgery, Harbin Medical University Cancer Hospital, Harbin, China
| | - Yingpu Li
- Department of oncological surgery, Harbin Medical University Cancer Hospital, Harbin, China.
| | - Fangjie Hu
- Department of Gastroenterology, Beijing Chaoyang Hospital, Capital Medical University, Beijing, China.
| | - He Ren
- Department of oncological surgery, Harbin Medical University Cancer Hospital, Harbin, China.
| |
Collapse
|
16
|
Zhang Y, Lu P, Jin S, Zhang J, Chen X. Transcriptional activation of SIRT5 by FOXA1 reprograms glycolysis to facilitate the malignant progression of diffuse large B-cell lymphoma. Cell Signal 2024; 123:111356. [PMID: 39173857 DOI: 10.1016/j.cellsig.2024.111356] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2024] [Revised: 08/06/2024] [Accepted: 08/18/2024] [Indexed: 08/24/2024]
Abstract
Diffuse large B-cell lymphoma (DLBCL) is the most common diagnosed subtype of lymphoma with high invasiveness and heterogeneity. Glycolysis is involved in regulating DLBCL progression. We aimed to explore the role of forkhead box protein A1 (FOXA1) in DLBCL and the mechanisms related to sirtuine5 (SIRT5) and glycolysis. FOXA1 expression in DLBCL cells was analyzed. Then, the proliferation and apoptosis of DLBCL cells were detected using Cell Counting Kit-8 (CCK-8), 5-ethynyl-2'-deoxyuridine (EDU) staining and flow cytometry analysis following FOXA1 or SIRT5 knockdown. The glycolysis was assessed by measuring extracellular acidification rate (ECAR), glucose consumption and lactate secretion. Immunoblotting was employed to examine the expression of apoptosis- and glycolysis-related proteins. Additionally, luciferase reporter assay and chromatin immunoprecipitation (ChIP) assay were conducted to test the combination of FOXA1 to SIRT5 promotor region. Subsequently, SIRT5 expression was upregulated to conduct rescue assays. Finally, the effects of FOXA1 downregulation on the growth and glycolysis in OCI-ly7 tumor-bearing mice were examined. As a result, FOXA1 was upregulated in DLBCL cells and FOXA1 or SIRT5 knockdown inhibited the proliferation, accelerated the apoptosis and suppressed glycolysis reprograming in DLBCL cells. Importantly, FOXA1 could transcriptionally activate SIRT5 expression in DLBCL cells. Besides, SIRT5 overexpression counteracted the effects of FOXA1 deficiency on the proliferation, apoptosis and glycolysis reprogramming in DLBCL cells. Furthermore, FOXA1 knockdown inhibited the tumor growth, suppressed the glycolysis reprogramming and downregulated SIRT5 expression in vivo. In summary, FOXA1 could transcriptionally activate SIRT5 to reprogram glycolysis, thereby facilitating the malignant progression of DLBCL.
Collapse
Affiliation(s)
- Ye Zhang
- Department of Hematology, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou City, Zhejiang Province 310016, PR China.
| | - Peng Lu
- Department of Neurosurgery, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou City, Zhejiang Province 310016, PR China
| | - Shenhe Jin
- Department of Hematology, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou City, Zhejiang Province 310016, PR China
| | - Jin Zhang
- Department of Hematology, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou City, Zhejiang Province 310016, PR China
| | - Xiaochang Chen
- Department of Hematology, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou City, Zhejiang Province 310016, PR China
| |
Collapse
|
17
|
Wang H. The interplay of EBV virus and cell metabolism in lung cancer. J Cell Mol Med 2024; 28:e70088. [PMID: 39601114 PMCID: PMC11599874 DOI: 10.1111/jcmm.70088] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2024] [Revised: 08/27/2024] [Accepted: 09/03/2024] [Indexed: 11/29/2024] Open
Abstract
Epstein-Barr virus infection has been implicated in various cancers, including lung cancer, where it influences cellular metabolism to promote tumorigenesis. This review examines the complex interplay between Epstein-Barr virus and cell metabolism in lung cancer, highlighting viral mechanisms of metabolic reprogramming and their implications for therapeutic strategies. Key viral proteins such as LMP1 and LMP2A manipulate glycolysis, glutaminolysis and lipid metabolism to support viral replication and immune evasion within the tumour microenvironment. Understanding these interactions provides insights into novel therapeutic approaches targeting viral-induced metabolic vulnerabilities in Epstein-Barr virus-associated lung cancer.
Collapse
Affiliation(s)
- Hongwei Wang
- Shanxi Province Cancer Hospital/Shanxi Hospital Affiliated to Cancer HospitalChinese Academy of Medical Sciences/Cancer Hospital Affiliated to Shanxi Medical UniversityTaiyuanShanxiChina
| |
Collapse
|
18
|
Zhou Y, Peng X, Fang C, Peng X, Tang J, Wang Z, Long Y, Chen J, Peng Y, Zhang Z, Zhou Y, Tang J, Liao J, Xiao D, Tao Y, Shi Y, Liu S. Histones Methyltransferase NSD3 Inhibits Lung Adenocarcinoma Glycolysis Through Interacting with PPP1CB to Decrease STAT3 Signaling Pathway. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2024; 11:e2400381. [PMID: 39119928 PMCID: PMC11481231 DOI: 10.1002/advs.202400381] [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: 01/10/2024] [Revised: 06/28/2024] [Indexed: 08/10/2024]
Abstract
Histones methyltransferase NSD3 targeting H3K36 is frequently disordered and mutant in various cancers, while the function of NSD3 during cancer initiation and progression remains unclear. In this study, it is proved that downregulated level of NSD3 is linked to clinical features and poor survival in lung adenocarcinoma. In vivo, NSD3 inhibited the proliferation, immigration, and invasion ability of lung adenocarcinoma. Meanwhile, NSD3 suppressed glycolysis by inhibiting HK2 translation, transcription, glucose uptake, and lactate production in lung adenocarcinoma. Mechanistically, as an intermediary, NSD3 binds to PPP1CB and p-STAT3 in protein levels, thus forming a trimer to dephosphorylate the level of p-STAT3 by PPP1CB, leading to the suppression of HK2 transcription. Interestingly, the phosphorylation function of PPP1CB is related to the concentration of carbon dioxide and pH value in the culture environment. Together, this study revealed the critical non-epigenetic role of NSD3 in the regulation of STAT3-dependent glycolysis, providing a piece of compelling evidence for targeting the NSD3/PPP1CB/p-STAT3 in lung adenocarcinoma.
Collapse
Affiliation(s)
- Yanling Zhou
- Department of OncologyInstitute of Medical SciencesNational Clinical Research Center for Geriatric DisordersInstitue of Medical SciencesXiangya Hospital, Central South UniversityChangshaHunan410008China
- Department of HematologyXiangya Hospital, Central South UniversityChangshaHunan410008China
| | - Xintong Peng
- Key Laboratory of Carcinogenesis and Cancer InvasionMinistry of Education, Department of PathologyXiangya Hospital, Central South UniversityChangshaHunan410008China
- Cancer Research InstituteSchool of Basic MedicineCentral South UniversityChangshaHunan410028China
| | - Cheng Fang
- Department of Cardiac SurgeryXiangya Hospital, Central South UniversityChangshaHunan410008China
| | - Xin Peng
- Department of PathologyXiangya Hospital, Central South UniversityChangshaHunan410008China
| | - Jianing Tang
- Department of Liver SurgeryXiangya Hospital, Central South UniversityChangshaHunan410008China
| | - Zuli Wang
- Center for Tissue Engineering and Stem Cell ResearchGuizhou Medical UniversityGuiyangGuizhou561113China
| | - Yao Long
- Key Laboratory of Carcinogenesis and Cancer InvasionMinistry of Education, Department of PathologyXiangya Hospital, Central South UniversityChangshaHunan410008China
- Cancer Research InstituteSchool of Basic MedicineCentral South UniversityChangshaHunan410028China
| | - Jielin Chen
- Department of PathologyXiangya Hospital, Central South UniversityChangshaHunan410008China
| | - Yuanhao Peng
- Key Laboratory of Carcinogenesis and Cancer InvasionMinistry of Education, Department of PathologyXiangya Hospital, Central South UniversityChangshaHunan410008China
- Cancer Research InstituteSchool of Basic MedicineCentral South UniversityChangshaHunan410028China
| | - Zewen Zhang
- Key Laboratory of Carcinogenesis and Cancer InvasionMinistry of Education, Department of PathologyXiangya Hospital, Central South UniversityChangshaHunan410008China
- Cancer Research InstituteSchool of Basic MedicineCentral South UniversityChangshaHunan410028China
| | - Yanmin Zhou
- Department of OncologyInstitute of Medical SciencesNational Clinical Research Center for Geriatric DisordersInstitue of Medical SciencesXiangya Hospital, Central South UniversityChangshaHunan410008China
| | - Jun Tang
- Key Laboratory of Carcinogenesis and Cancer InvasionMinistry of Education, Department of PathologyXiangya Hospital, Central South UniversityChangshaHunan410008China
- Cancer Research InstituteSchool of Basic MedicineCentral South UniversityChangshaHunan410028China
| | - Jingzhong Liao
- Department of Laboratory MedicineXiangya Hospital, Central South UniversityChangshaHunan410008China
| | - Desheng Xiao
- Department of PathologyXiangya Hospital, Central South UniversityChangshaHunan410008China
| | - Yongguang Tao
- Key Laboratory of Carcinogenesis and Cancer InvasionMinistry of Education, Department of PathologyXiangya Hospital, Central South UniversityChangshaHunan410008China
- Cancer Research InstituteSchool of Basic MedicineCentral South UniversityChangshaHunan410028China
| | - Ying Shi
- Key Laboratory of Carcinogenesis and Cancer InvasionMinistry of Education, Department of PathologyXiangya Hospital, Central South UniversityChangshaHunan410008China
- Cancer Research InstituteSchool of Basic MedicineCentral South UniversityChangshaHunan410028China
| | - Shuang Liu
- Department of OncologyInstitute of Medical SciencesNational Clinical Research Center for Geriatric DisordersInstitue of Medical SciencesXiangya Hospital, Central South UniversityChangshaHunan410008China
- Key Laboratory of Carcinogenesis and Cancer InvasionMinistry of Education, Department of PathologyXiangya Hospital, Central South UniversityChangshaHunan410008China
- Department of PathologyXiangya Hospital, Central South UniversityChangshaHunan410008China
| |
Collapse
|
19
|
Zhu Y, Wang Z, Li H, Ren Z, Zi T, Qin X, Sun W, Chen X, Wu G. LncRNA HCG18 promotes prostate cancer progression by regulating the miR-512-3p/HK-2 axis. Asian J Urol 2024; 11:575-585. [PMID: 39533994 PMCID: PMC11551385 DOI: 10.1016/j.ajur.2024.01.007] [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: 05/07/2023] [Accepted: 10/10/2023] [Indexed: 11/16/2024] Open
Abstract
Objective Long non-coding RNAs (lncRNAs) play an important role in tumor progression. Numerous studies show that lncRNAs are strongly associated with prostate cancer (PCa) progression. The aim of this study was to investigate the pathway through which lncRNA HCG18 regulates PCa progression by bioinformatics analysis and experiments. Methods We compared HCG18 expression in PCa versus normal tissue and cells by data and cell lines, followed by comparing the changes in tumor cell proliferation, migration, and invasive ability after knockdown of HCG18. Then we searched for its downstream pathway by database and validated the pathway in vivo and in vitro. Results HCG18 was highly expressed in PCa and has the ability to promote tumor proliferation, migration, and invasion; knockdown of HCG18 led to a decrease in the ability of cells to do so, which can be reversed by knockdown of miR-512-3p or overexpression of hexokinase 2. Conclusion Our in vivo and in vitro experiments suggest that HCG18 can play a role in promoting PCa progression by blocking the inhibition of hexokinase 2 by miR-512-3p via sponge adsorption.
Collapse
Affiliation(s)
- Yaru Zhu
- Department of Urology, Tongji Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Zhijing Wang
- Department of Gastroenterology and Hepatology, Tongji Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Haopeng Li
- Department of Urology, Tongji Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Zhen Ren
- Department of Medicine, Dalian University of Technology, Dalian, China
| | - Tong Zi
- Department of Urology, Tongji Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Xin Qin
- Department of Urology, Tongji Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Wenhuizi Sun
- Department of Obstetrics and Gynecology, Tongji Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Xi Chen
- Department of Urology, Tongji Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Gang Wu
- Department of Urology, Tongji Hospital, School of Medicine, Tongji University, Shanghai, China
| |
Collapse
|
20
|
Zhang L, Li M, Li X, Xiao T, Zhou H, Zhang W, Wang P. Deciphering the role of PLCD3 in lung cancer: A gateway to glycolytic reprogramming via PKC-Rap1 activation. Heliyon 2024; 10:e37063. [PMID: 39296221 PMCID: PMC11408031 DOI: 10.1016/j.heliyon.2024.e37063] [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: 05/13/2024] [Revised: 08/21/2024] [Accepted: 08/27/2024] [Indexed: 09/21/2024] Open
Abstract
PLCD3 belongs to the phospholipase C delta group and is involved in numerous biological functions, including cell growth, programmed cell death, and specialization. However, the role of PLCD3 in lung cancer still needs further investigation. This research aimed to investigate if PLCD3 influences glycolytic reprogramming and lung cancer development through the PKC-dependent Rap1 signaling pathway. This study found that PLCD3 was increased in lung cancer tissues. PLCD3 promotes the proliferation and invasion of lung cancer cells by activating the PKC-dependent Rap1 pathway. The detailed process involves PLCD3 triggering PKC, which subsequently stimulates the Rap1 pathway, leading to glycolytic reprogramming that supplies adequate energy and metabolic substrates necessary for the growth and spread of lung cancer cells. Moreover, PLCD3 can also promote the metastasis and invasion of lung cancer cells by activating the Rap1 pathway. This study reveals the mechanism of PLCD3 in lung cancer and provides new ideas for the treatment of lung cancer. Inhibiting PLCD3, PKC, and the Rap1 pathway may be an effective strategy for treating lung cancer.
Collapse
Affiliation(s)
- Liang Zhang
- Tianjin Medical University Cancer Institute & Hospital, Tianjin, PR China
- Department of Thoracic Surgery, Tianjin First Central Hospital, Tianjin, 300192, PR China
| | - Mingjiang Li
- Department of Thoracic Surgery, Tianjin First Central Hospital, Tianjin, 300192, PR China
| | - Xiaoping Li
- Department of Thoracic Surgery, Tianjin First Central Hospital, Tianjin, 300192, PR China
| | - Ting Xiao
- College of Pharmacy and Key Laboratory of Molecular Drug Research, Nankai University, Tianjin, PR China
| | - Honggang Zhou
- College of Pharmacy and Key Laboratory of Molecular Drug Research, Nankai University, Tianjin, PR China
| | - Weidong Zhang
- Department of Thoracic Surgery, Tianjin First Central Hospital, Tianjin, 300192, PR China
| | - Ping Wang
- Tianjin Medical University Cancer Institute & Hospital, Tianjin, PR China
| |
Collapse
|
21
|
He XY, Liang JT, Xiao JY, Li X, Zhang XB, Chen DY, Wu LJ. Dahuang Zhechong Pill Improves Pulmonary Fibrosis through miR-29b-2-5p/HK2 Mediated Glycolysis Pathway. Chin J Integr Med 2024:10.1007/s11655-024-3765-x. [PMID: 39231918 DOI: 10.1007/s11655-024-3765-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/17/2024] [Indexed: 09/06/2024]
Abstract
OBJECTIVE To explore the preventive and therapeutic effects of Dahuang Zhechong Pill (DZP) on pulmonary fibrosis and the underlying mechanisms. METHODS The first key rate-limiting enzyme hexokinase 2 (HK2) of glycolysis was silenced and over-expressed through small interfering RNA and lentivirus using lung fibroblast MRC-5 cell line, respectively. The cell viability, migration, invasion and proliferation were detected by cell counting kit-8, wound healing assay, transwell assay, and flow cytometry. The mRNA and protein expression levels of HK2 were detected by RT-PCR and Western blotting, respectively. The contents of glucose, adenosine triphosphate (ATP) and lactate in MRC-5 cells were determined by enzyme-linked immunosorbnent assay (ELISA). Then, the relationship between miR-29b-2-5p and HK2 was explored by luciferase reporter gene assay. Pulmonary fibrosis cell model was induced by transforming growth factor-β 1 (TGF-β 1) in MRC-5 cells, and the medicated serum of DZP (DMS) was prepared in rats. MRC-5 cells were divided into control, TGF-β 1, TGF-β 1+10% DMS, TGF-β 1+10% DMS+miR-29b-2-5p inhibitor, TGF-β 1+10% DMS+inhibitor negative control, TGF-β 1+10% DMS+miR-29b-2-5p mimic and TGF-β 1+10% DMS+mimic negative control groups. After miR-29b-2-5p mimics and inhibitors were transfected into MRC-5 cells, all groups except control and model group were treated with DMS. The effect of DMS on MRC-5 cells were detected using aforementioned methods and immunofluorescence. Similarly, the contents of glucose, ATP and lactate in each group were measured by ELISA. RESULTS The mRNA and protein expressions of HK2 in MRC-5 cells were successfully silenced and overexpressed through si-HK2-3 and lentiviral transfection, respectively. After silencing HK2, the mRNA and protein expressions of HK2 were significantly decreased (P<0.01), and the concentrations of glucose, ATP and lactate were also significantly decreased (P<0.05). The proliferation, migration and invasion of MRC-5 cells were significantly declined (P<0.05 or P<0.01), while the apoptosis of MRC-5 cells was significantly increased (P<0.01). After overexpressing HK2, the mRNA and protein expressions of HK2 were significantly increased (P<0.05), and the concentrations of glucose, ATP and lactate were also significantly increased (P<0.05 or P<0.01). The proliferation, migration and invasion of MRC-5 cells were significantly increased (P<0.05 or P<0.01), while the apoptosis of MRC-5 cells was significantly decreased (P<0.05). The relative luciferase activity of 3'UTR-WT+hsa-miR-29b-2-5p transfected with HK2 was significantly decreased (P<0.01). After miR-29b-2-5p mimic and inhibitor were transfected into the MRC-5 cells, DMS intervention could significantly reduce the concentration of glucose, ATP and lactate, and the mRNA and proteins expressions of HK2, phosphofructokinase and pyruvate kinase isoform M2 (P<0.05 or P<0.01). The proliferation, migration and invasion of MRC-5 cells were alleviated (P<0.05 or P<0.01), and the deposition of fibronectin, α-smooth muscle actin, and collagen I were significantly decreased (P<0.05 or P<0.01). CONCLUSIONS Glycolysis is closely related to pulmonary fibrosis. DZP reduced glycolysis and inhibited fibroblasts' excessive differentiation and abnormal collagen deposition through the miR-29b-2-5p/HK2 pathway, which played a role in delaying the process of pulmonary fibrosis.
Collapse
Affiliation(s)
- Xiao-Yan He
- College of Public Health, Chengdu University of Traditional Chinese Medicine, Chengdu, 611137, China
| | - Jing-Tao Liang
- Department of Neurology, Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, 610072, China
| | - Jing-Yi Xiao
- College of Public Health, Chengdu University of Traditional Chinese Medicine, Chengdu, 611137, China
| | - Xin Li
- College of Public Health, Chengdu University of Traditional Chinese Medicine, Chengdu, 611137, China
| | - Xiao-Bo Zhang
- College of Public Health, Chengdu University of Traditional Chinese Medicine, Chengdu, 611137, China
| | - Da-Yi Chen
- College of Public Health, Chengdu University of Traditional Chinese Medicine, Chengdu, 611137, China
| | - Li-Juan Wu
- College of Public Health, Chengdu University of Traditional Chinese Medicine, Chengdu, 611137, China.
| |
Collapse
|
22
|
Zhang H, Li S, Wang D, Liu S, Xiao T, Gu W, Yang H, Wang H, Yang M, Chen P. Metabolic reprogramming and immune evasion: the interplay in the tumor microenvironment. Biomark Res 2024; 12:96. [PMID: 39227970 PMCID: PMC11373140 DOI: 10.1186/s40364-024-00646-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2024] [Accepted: 08/24/2024] [Indexed: 09/05/2024] Open
Abstract
Tumor cells possess complex immune evasion mechanisms to evade immune system attacks, primarily through metabolic reprogramming, which significantly alters the tumor microenvironment (TME) to modulate immune cell functions. When a tumor is sufficiently immunogenic, it can activate cytotoxic T-cells to target and destroy it. However, tumors adapt by manipulating their metabolic pathways, particularly glucose, amino acid, and lipid metabolism, to create an immunosuppressive TME that promotes immune escape. These metabolic alterations impact the function and differentiation of non-tumor cells within the TME, such as inhibiting effector T-cell activity while expanding regulatory T-cells and myeloid-derived suppressor cells. Additionally, these changes lead to an imbalance in cytokine and chemokine secretion, further enhancing the immunosuppressive landscape. Emerging research is increasingly focusing on the regulatory roles of non-tumor cells within the TME, evaluating how their reprogrammed glucose, amino acid, and lipid metabolism influence their functional changes and ultimately aid in tumor immune evasion. Despite our incomplete understanding of the intricate metabolic interactions between tumor and non-tumor cells, the connection between these elements presents significant challenges for cancer immunotherapy. This review highlights the impact of altered glucose, amino acid, and lipid metabolism in the TME on the metabolism and function of non-tumor cells, providing new insights that could facilitate the development of novel cancer immunotherapies.
Collapse
Affiliation(s)
- Haixia Zhang
- The Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Hunan Cancer Hospital, Changsha, China
- Department of Pediatrics, Third Xiangya Hospital, Central South University, Changsha, China
| | - Shizhen Li
- The Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Hunan Cancer Hospital, Changsha, China
| | - Dan Wang
- Department of Pediatrics, Third Xiangya Hospital, Central South University, Changsha, China
| | - Siyang Liu
- Department of Pediatrics, Third Xiangya Hospital, Central South University, Changsha, China
| | - Tengfei Xiao
- The Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Hunan Cancer Hospital, Changsha, China
| | - Wangning Gu
- The Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Hunan Cancer Hospital, Changsha, China
| | - Hongmin Yang
- The Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Hunan Cancer Hospital, Changsha, China
| | - Hui Wang
- The Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Hunan Cancer Hospital, Changsha, China.
| | - Minghua Yang
- Department of Pediatrics, Third Xiangya Hospital, Central South University, Changsha, China.
| | - Pan Chen
- The Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Hunan Cancer Hospital, Changsha, China.
| |
Collapse
|
23
|
Tong Y, Liu X, Wu L, Xiang Y, Wang J, Cheng Y, Zhang C, Han B, Wang L, Yan D. Hexokinase 2 nonmetabolic function-mediated phosphorylation of IκBα enhances pancreatic ductal adenocarcinoma progression. Cancer Sci 2024; 115:2673-2685. [PMID: 38801832 PMCID: PMC11309947 DOI: 10.1111/cas.16204] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2023] [Revised: 04/15/2024] [Accepted: 04/18/2024] [Indexed: 05/29/2024] Open
Abstract
Aberrant signaling in tumor cells induces nonmetabolic functions of some metabolic enzymes in many cellular activities. As a key glycolytic enzyme, the nonmetabolic function of hexokinase 2 (HK2) plays a role in tumor immune evasion. However, whether HK2, dependent of its nonmetabolic activity, plays a role in human pancreatic ductal adenocarcinoma (PDAC) tumorigenesis remains unclear. Here, we demonstrated that HK2 acts as a protein kinase and phosphorylates IκBα at T291 in PDAC cells, activating NF-κB, which enters the nucleus and promotes the expression of downstream targets under hypoxia. HK2 nonmetabolic activity-promoted activation of NF-κB promotes the proliferation, migration, and invasion of PDAC cells. These findings provide new insights into the multifaceted roles of HK2 in tumor development and underscore the potential of targeting HK2 protein kinase activity for PDAC treatment.
Collapse
Affiliation(s)
- Yingying Tong
- Cancer Center, Beijing Luhe HospitalCapital Medical UniversityBeijingChina
| | - Xin Liu
- Cancer Center, Beijing Luhe HospitalCapital Medical UniversityBeijingChina
| | - Lihui Wu
- Zhejiang Provincial Key Laboratory of Pancreatic DiseaseThe First Affiliated Hospital, Zhejiang University School of MedicineHangzhouChina
| | - Yaoxian Xiang
- Cancer Center, Beijing Luhe HospitalCapital Medical UniversityBeijingChina
| | - Jing Wang
- Cancer Center, Beijing Luhe HospitalCapital Medical UniversityBeijingChina
| | - Yurong Cheng
- Cancer Center, Beijing Luhe HospitalCapital Medical UniversityBeijingChina
| | - Chan Zhang
- Cancer Center, Beijing Luhe HospitalCapital Medical UniversityBeijingChina
| | - Baojuan Han
- Cancer Center, Beijing Luhe HospitalCapital Medical UniversityBeijingChina
| | - Li Wang
- Cancer Center, Beijing Luhe HospitalCapital Medical UniversityBeijingChina
| | - Dong Yan
- Cancer Center, Beijing Luhe HospitalCapital Medical UniversityBeijingChina
| |
Collapse
|
24
|
Tian Z, Dai Q, Liu B, Lin H, Ou H. LncARSR promotes glioma tumor growth by mediating glycolysis through the STAT3/HK2 axis. Cytokine 2024; 180:156663. [PMID: 38815522 DOI: 10.1016/j.cyto.2024.156663] [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: 03/15/2024] [Revised: 05/20/2024] [Accepted: 05/21/2024] [Indexed: 06/01/2024]
Abstract
BACKGROUND Glioma represents the predominant malignant brain tumor. This investigation endeavors to elucidate the impact and prospective mechanisms of glycolysis-related lncARSR on glioma. METHODS LncARSR level was assessed in normal glial cells and glioma cells. Cell proliferation, migration, and invasion measurements were conducted through CCK-8, wound healing, and transwell assay. Flow cytometry was utilized to measure cell apoptosis and cell cycle. Biochemical assay kits and immunoblotting were employed to measure the content of glycolysis-related indicators and protein expression, respectively. We analyzed the impact of both lncARSR knockdown and overexpression of the Signal Transducer and Activator of Transcription 3 (STAT3) on Hexokinase 2 (HK2) using dual luciferase reporter assays and Chromatin Immunoprecipitation (ChIP) experiments. Further assessment of the impact of lncARSR on glioma progression was conducted through animal experiments. RESULTS LncARSR was expressed at elevated levels in glioma cells compared to normal glial cells. Overexpressing lncARSR enhanced proliferation, migration, invasion, and G2/M phase arrest in glioma cells and also increased glucose, lactate, ATP production, as well as the expression of HK2, PFK1, PKM2, GLUT1, and LDHA. STAT3 binding to the HK2 gene promoter was weakened following the knockdown of lncARSR. Upregulation of STAT3 reversed the suppressed functions of knocking down lncARSR on cell proliferation, migration, invasion, G2/M phase arrest, and glycolysis and counteracted its promotional effect on cell apoptosis. In vivo, knocking down lncARSR inhibits glioma growth and ki67 and PCNA expression. CONCLUSION LncARSR promotes the development of glioma by enhancing glycolysis through the STAT3-HK2 axis.
Collapse
Affiliation(s)
- Zhenyang Tian
- School of Pharmacy, Hunan Traditional Chinese Medicinal College, Zhuzhou, Hunan, China
| | - Qi Dai
- School of Traditional Chinese Medicine, Hunan Traditional Chinese Medicinal College, Zhuzhou, Hunan, China
| | - Bin Liu
- School of Traditional Chinese Medicine, Hunan Traditional Chinese Medicinal College, Zhuzhou, Hunan, China
| | - Hui Lin
- School of Traditional Chinese Medicine, Hunan Traditional Chinese Medicinal College, Zhuzhou, Hunan, China.
| | - Huiping Ou
- School of Traditional Chinese Medicine, Hunan Traditional Chinese Medicinal College, Zhuzhou, Hunan, China.
| |
Collapse
|
25
|
Spinelli S, Barbieri F, Averna M, Florio T, Pedrazzi M, Tremonti BF, Capraro M, De Tullio R. Expression of calpastatin hcast 3-25 and activity of the calpain/calpastatin system in human glioblastoma stem cells: possible involvement of hcast 3-25 in cell differentiation. Front Mol Biosci 2024; 11:1359956. [PMID: 39139809 PMCID: PMC11319182 DOI: 10.3389/fmolb.2024.1359956] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2023] [Accepted: 07/03/2024] [Indexed: 08/15/2024] Open
Abstract
Glioblastoma (GBM) is the most malignant brain tumor, characterized by cell heterogeneity comprising stem cells (GSCs) responsible for aggressiveness. The calpain/calpastatin (calp/cast) proteolytic system is involved in critical physiological processes and cancer progression. In this work we showed the expression profile of hcast 3-25 (a Type III calpastatin variant devoid of inhibitory units) and the members of the system in several patient-derived GSCs exploring the relationship between hcast 3-25 and activation/activity of calpains. Each GSC shows a peculiar calp/cast mRNA and protein expression pattern, and hcast 3-25 is the least expressed. Differentiation promotes upregulation of all the calp/cast system components except hcast 3-25 mRNA, which increased or decreased depending on individual GSC culture. Transfection of hcast 3-25-V5 into two selected GSCs indicated that hcast 3-25 effectively associates with calpains, supporting the digestion of selected calpain targets. Hcast 3-25 possibly affects the stem state promoting a differentiated, less aggressive phenotype.
Collapse
Affiliation(s)
- Sonia Spinelli
- IRCCS Istituto Giannina Gaslini, Laboratory of Molecular Nephrology, Genova, Italy
- Department of Experimental Medicine (DIMES), Section of Biochemistry, University of Genova, Genova, Italy
| | - Federica Barbieri
- Department of Internal Medicine (DIMI), Section of Pharmacology, University of Genova, Genova, Italy
- IRCCS Ospedale Policlinico San Martino, Genova, Italy
| | - Monica Averna
- Department of Experimental Medicine (DIMES), Section of Biochemistry, University of Genova, Genova, Italy
| | - Tullio Florio
- Department of Internal Medicine (DIMI), Section of Pharmacology, University of Genova, Genova, Italy
- IRCCS Ospedale Policlinico San Martino, Genova, Italy
| | - Marco Pedrazzi
- Department of Experimental Medicine (DIMES), Section of Biochemistry, University of Genova, Genova, Italy
| | - Beatrice F. Tremonti
- Department of Internal Medicine (DIMI), Section of Pharmacology, University of Genova, Genova, Italy
| | - Michela Capraro
- Department of Experimental Medicine (DIMES), Section of Biochemistry, University of Genova, Genova, Italy
| | - Roberta De Tullio
- Department of Experimental Medicine (DIMES), Section of Biochemistry, University of Genova, Genova, Italy
| |
Collapse
|
26
|
Codocedo JF, Mera-Reina C, Bor-Chian Lin P, Fallen PB, Puntambekar SS, Casali BT, Jury-Garfe N, Martinez P, Lasagna-Reeves CA, Landreth GE. Therapeutic targeting of immunometabolism reveals a critical reliance on hexokinase 2 dosage for microglial activation and Alzheimer's progression. Cell Rep 2024; 43:114488. [PMID: 39002124 PMCID: PMC11398604 DOI: 10.1016/j.celrep.2024.114488] [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: 03/01/2022] [Revised: 03/14/2024] [Accepted: 06/25/2024] [Indexed: 07/15/2024] Open
Abstract
Neuroinflammation is a prominent feature of Alzheimer's disease (AD). Activated microglia undergo a reprogramming of cellular metabolism necessary to power their cellular activities during disease. Thus, selective targeting of microglial immunometabolism might be of therapeutic benefit for treating AD. In the AD brain, the levels of microglial hexokinase 2 (HK2), an enzyme that supports inflammatory responses by promoting glycolysis, are significantly increased. In addition, HK2 displays non-metabolic activities that extend its inflammatory role beyond glycolysis. The antagonism of HK2 affects microglial phenotypes and disease progression in a gene-dose-dependent manner. HK2 complete loss fails to improve pathology by exacerbating inflammation, while its haploinsufficiency reduces pathology in 5xFAD mice. We propose that the partial antagonism of HK2 is effective in slowing disease progression by modulating NF-κB signaling through its cytosolic target, IKBα. The complete loss of HK2 affects additional inflammatory mechanisms related to mitochondrial dysfunction.
Collapse
Affiliation(s)
- Juan F Codocedo
- Stark Neurosciences Research Institute, Indiana University School of Medicine, Indianapolis, IN 46202, USA; Department of Anatomy, Cell Biology and Physiology, Indiana University School of Medicine, Indianapolis, IN 46202, USA
| | - Claudia Mera-Reina
- Stark Neurosciences Research Institute, Indiana University School of Medicine, Indianapolis, IN 46202, USA; Department of Anatomy, Cell Biology and Physiology, Indiana University School of Medicine, Indianapolis, IN 46202, USA
| | - Peter Bor-Chian Lin
- Stark Neurosciences Research Institute, Indiana University School of Medicine, Indianapolis, IN 46202, USA
| | - Paul B Fallen
- Stark Neurosciences Research Institute, Indiana University School of Medicine, Indianapolis, IN 46202, USA; Department of Anatomy, Cell Biology and Physiology, Indiana University School of Medicine, Indianapolis, IN 46202, USA
| | - Shweta S Puntambekar
- Stark Neurosciences Research Institute, Indiana University School of Medicine, Indianapolis, IN 46202, USA; Department of Medical and Molecular Genetics, Indiana University School of Medicine, Indianapolis, IN 46202, USA
| | - Brad T Casali
- Stark Neurosciences Research Institute, Indiana University School of Medicine, Indianapolis, IN 46202, USA; Department of Anatomy, Cell Biology and Physiology, Indiana University School of Medicine, Indianapolis, IN 46202, USA
| | - Nur Jury-Garfe
- Stark Neurosciences Research Institute, Indiana University School of Medicine, Indianapolis, IN 46202, USA; Department of Anatomy, Cell Biology and Physiology, Indiana University School of Medicine, Indianapolis, IN 46202, USA
| | - Pablo Martinez
- Stark Neurosciences Research Institute, Indiana University School of Medicine, Indianapolis, IN 46202, USA; Department of Anatomy, Cell Biology and Physiology, Indiana University School of Medicine, Indianapolis, IN 46202, USA
| | - Cristian A Lasagna-Reeves
- Stark Neurosciences Research Institute, Indiana University School of Medicine, Indianapolis, IN 46202, USA; Department of Anatomy, Cell Biology and Physiology, Indiana University School of Medicine, Indianapolis, IN 46202, USA
| | - Gary E Landreth
- Stark Neurosciences Research Institute, Indiana University School of Medicine, Indianapolis, IN 46202, USA; Department of Anatomy, Cell Biology and Physiology, Indiana University School of Medicine, Indianapolis, IN 46202, USA.
| |
Collapse
|
27
|
Singla P, Jain A. Deciphering the complex landscape of post-translational modifications on PKM2: Implications in head and neck cancer pathogenesis. Life Sci 2024; 349:122719. [PMID: 38759866 DOI: 10.1016/j.lfs.2024.122719] [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: 03/07/2024] [Revised: 05/03/2024] [Accepted: 05/13/2024] [Indexed: 05/19/2024]
Abstract
In the vast landscape of human health, head and neck cancer (HNC) poses a significant health burden globally, necessitating the exploration of novel diagnostics and therapeutics. Metabolic alterations occurring within tumor microenvironment are crucial to understand the foundational cause of HNC. Post-translational modifications (PTMs) have recently emerged as a silent foe exerting a significantly heightened influence on various aspects of the biological processes associated with the onset and advancement of cancer, particularly in the context of HNC. There are numerous targets involved in HNC but recently, the enzyme pyruvate kinase M2 (PKM2) has come out as a hot target due to its involvement in glycolysis resulting in metabolic reprogramming of cancer cells. Various PTMs have been reported to affect the structure and function of PKM2 by modulating its activity. This review aims to investigate the impact of PTMs on the interaction between PKM2 and several signaling pathways and transcription factors in the context of HNC. These interactions possess significant ramification for cellular proliferation, apoptosis, angiogenesis and metastasis. This review primarily explores the role of PTMs influencing PKM2 and its involvement in tumor development. While acknowledging the significance of PKM2 interactions with other tumor regulators, the emphasis lies on dissecting PTM-related mechanisms rather than solely scrutinizing individual regulators. It lays the framework for the development of more sophisticated diagnostic tools and uncovers exciting possibilities for precision medicine essential for effectively addressing the complexity of this malignancy in a precise and focused manner.
Collapse
Affiliation(s)
- Palak Singla
- Department of Bioengineering and Biotechnology, Birla Institute of Technology Mesra, Ranchi 835215, Jharkhand, India
| | - Alok Jain
- Department of Bioengineering and Biotechnology, Birla Institute of Technology Mesra, Ranchi 835215, Jharkhand, India.
| |
Collapse
|
28
|
Xu J, Lu W, Wei X, Zhang B, Yang H, Tu M, Chen X, Wu S, Guo T. Single-cell transcriptomics reveals the aggressive landscape of high-grade serous carcinoma and therapeutic targets in tumor microenvironment. Cancer Lett 2024; 593:216928. [PMID: 38714290 DOI: 10.1016/j.canlet.2024.216928] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2024] [Revised: 03/29/2024] [Accepted: 04/26/2024] [Indexed: 05/09/2024]
Abstract
High-grade serous carcinoma (HGSC) is characterized by early abdominal metastasis, leading to a dismal prognosis. In this study, we conducted single-cell RNA sequencing on 109,573 cells from 34 tumor samples of 18 HGSC patients, including both primary tumors and their metastatic sites. Our analysis revealed a distinct S100A9+ tumor cell subtype present in both primary and metastatic sites, strongly associated with poor overall survival. This subtype exhibited high expression of S100A8, S100A9, ADGRF1, CEACAM6, CST6, NDRG2, MUC4, PI3, SDC1, and C15orf48. Individual knockdown of these ten marker genes, validated through in vitro and in vivo models, significantly inhibited ovarian cancer growth and invasion. Around S100A9+ tumor cells, a population of HK2+_CAF was identified, characterized by activated glycolysis metabolism, correlating with shorter overall survival in patients. Notably, similar to CAFs, immunosuppressive tumor-associated macrophage (TAM) subtypes underwent glycolipid metabolism reprogramming via PPARgamma regulation, promoting tumor metastasis. These findings shed light on the mechanisms driving the aggressiveness of HGSC, offering crucial insights for the development of novel therapeutic targets against this formidable cancer.
Collapse
Affiliation(s)
- Junfen Xu
- Department of Gynecologic Oncology, Women's Hospital, Zhejiang University School of Medicine, Hangzhou, 310006, Zhejiang, China; Zhejiang Provincial Clinical Research Center for Obstetrics and Gynecology, Hangzhou, 310006, Zhejiang, China.
| | - Weiguo Lu
- Department of Gynecologic Oncology, Women's Hospital, Zhejiang University School of Medicine, Hangzhou, 310006, Zhejiang, China; Zhejiang Provincial Clinical Research Center for Obstetrics and Gynecology, Hangzhou, 310006, Zhejiang, China
| | - Xinyi Wei
- Department of Gynecologic Oncology, Women's Hospital, Zhejiang University School of Medicine, Hangzhou, 310006, Zhejiang, China
| | - Bo Zhang
- Novel Bioinformatics Co., Ltd, Shanghai, China
| | - Haihua Yang
- Novel Bioinformatics Co., Ltd, Shanghai, China
| | - Mengyan Tu
- Department of Gynecologic Oncology, Women's Hospital, Zhejiang University School of Medicine, Hangzhou, 310006, Zhejiang, China
| | - Xin Chen
- Department of Gynecologic Oncology, Women's Hospital, Zhejiang University School of Medicine, Hangzhou, 310006, Zhejiang, China
| | - Shenglong Wu
- Department of Gynecologic Oncology, Women's Hospital, Zhejiang University School of Medicine, Hangzhou, 310006, Zhejiang, China
| | - Tianchen Guo
- Department of Gynecologic Oncology, Women's Hospital, Zhejiang University School of Medicine, Hangzhou, 310006, Zhejiang, China
| |
Collapse
|
29
|
Fu Y, Chen B, Gao T, Wang Z. CircSLC25A16 facilitates the development of non-small-cell lung cancer through the miR-335-5p/CISD2 axis. Thorac Cancer 2024; 15:1490-1501. [PMID: 38803052 PMCID: PMC11219286 DOI: 10.1111/1759-7714.15163] [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: 09/04/2023] [Revised: 10/31/2023] [Accepted: 11/02/2023] [Indexed: 05/29/2024] Open
Abstract
BACKGROUND Non-small-cell lung cancer (NSCLC) is a common malignancy with high morbidity and mortality. Circular RNAs are widely involved in NSCLC progression. However, the mechanism of circSLC25A16 in NSCLC has not been reported. METHODS The expressions of circSLC25A16, microRNA-335-5p (miR-335-5p), and CDGSH iron-sulfur domain-containing protein 2 (CISD2) were monitored by quantitative real-time fluorescence polymerase chain reaction. Western blot was also carried out to measure the protein levels of CISD2, hexokinase 2 (HK2), and lactate dehydrogenase A (LDHA). For functional analysis, cell counting kit-8 assay, 5-ethynyl-2'-deoxyuridine, flow cytometry, transwell, and wound healing assays were utilized to examine cell proliferation, apoptosis, and migration. Glucose uptake and lactate production were detected using commercial kits. The relationship between miR-335-5p and circSLC25A16 or CISD2 was verified by dual-luciferase reporter and RNA immunoprecipitation assays. Furthermore, tumor xenograft was established to explore the function of circSLC25A16 in vivo. RESULTS CircSLC25A16 and CISD2 were overexpressed in NSCLC, but miR-335-5p was downregulated. CircSLC25A16 acted as a miR-335-5p sponge, and silencing of circSLC25A16 arrested cell proliferation, migration, and glycolysis, and promoted apoptosis, but these impacts were resumed by miR-335-5p inhibition. CISD2 was a miR-335-5p target, and overexpression of CISD2 abolished the suppressive function of miR-335-5p mimic on the malignant behavior of NSCLC cells. CircSLC25A16 could adsorb miR-335-5p to mediate CISD2 expression. Additionally, silencing circSLC25A16 restrained the growth of NSCLC tumor xenograft in vivo. CONCLUSION CircSLC25A16 facilitated NSCLC progression via the miR-335-5p/CISD2 axis, implying that circSLC25A16 may serve as a novel biomarker for NSCLC treatment.
Collapse
Affiliation(s)
- Yu Fu
- Department of Respiratory MedicineYiwu Fuyuan Private HospitalYiwu CityChina
| | - Bin Chen
- Department of PharmacyYiwu Fuyuan Private HospitalYiwu CityChina
| | - Tao Gao
- Department of Respiratory MedicineYiwu Fuyuan Private HospitalYiwu CityChina
| | - Zhenglong Wang
- Department of Respiratory MedicineYiwu Fuyuan Private HospitalYiwu CityChina
| |
Collapse
|
30
|
Gao W, Wang J, Xu Y, Yu H, Yi S, Bai C, Cong Q, Zhu Y. Research progress in the metabolic reprogramming of hepatocellular carcinoma (Review). Mol Med Rep 2024; 30:131. [PMID: 38818815 PMCID: PMC11148525 DOI: 10.3892/mmr.2024.13255] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2023] [Accepted: 05/03/2024] [Indexed: 06/01/2024] Open
Abstract
Hepatocellular carcinoma (HCC) is the most common primary liver malignancy and its morbidity is increasing worldwide due to increasing prevalence. Metabolic reprogramming has been recognized as a hallmark of cancer and serves a role in cancer progression. Glucose, lipids and amino acids are three major components whose altered metabolism can directly affect the energy production of cells, including liver cancer cells. Nutrients and energy are indispensable for the growth and proliferation of cancer cells, thus altering the metabolism of hepatoma cells can inhibit the progression of HCC. The present review summarizes recent studies on tumour regulatory molecules, including numerous noncoding RNAs, oncogenes and tumour suppressors, which regulate the metabolic activities of glucose, lipids and amino acids by targeting key enzymes, signalling pathways or interactions between the two. These regulatory molecules can regulate the rapid proliferation of cancer cells, tumour progression and treatment resistance. It is thought that these tumour regulatory factors may serve as therapeutic targets or valuable biomarkers for HCC, with the potential to mitigate HCC drug resistance. Furthermore, the advantages and disadvantages of metabolic inhibitors as a treatment approach for HCC, as well as possible solutions are discussed, providing insights for developing more effective treatment strategies for HCC.
Collapse
Affiliation(s)
- Wenyue Gao
- Department of Infectious Diseases, The First Affiliated Hospital of Dalian Medical University, Dalian, Liaoning 116000, P.R China
| | - Jing Wang
- Department of Infectious Diseases, The First Affiliated Hospital of Dalian Medical University, Dalian, Liaoning 116000, P.R China
| | - Yuting Xu
- Department of Infectious Diseases, The First Affiliated Hospital of Dalian Medical University, Dalian, Liaoning 116000, P.R China
| | - Hongbo Yu
- Department of Infectious Diseases, The First Affiliated Hospital of Dalian Medical University, Dalian, Liaoning 116000, P.R China
| | - Sitong Yi
- Department of Infectious Diseases, The First Affiliated Hospital of Dalian Medical University, Dalian, Liaoning 116000, P.R China
| | - Changchuan Bai
- Internal Department of Chinese Medicine, Dalian Hospital of Traditional Chinese Medicine, Dalian, Liaoning 116000, P.R China
| | - Qingwei Cong
- Department of Infectious Diseases, The First Affiliated Hospital of Dalian Medical University, Dalian, Liaoning 116000, P.R China
| | - Ying Zhu
- Department of Infectious Diseases, The First Affiliated Hospital of Dalian Medical University, Dalian, Liaoning 116000, P.R China
| |
Collapse
|
31
|
Lin F, Long Y, Li M, Cai C, Wu Y, You X, Tian X, Zhou Q. Xihuang pills targeting the Warburg effect through inhibition of the Wnt/β-catenin pathway in prostate cancer. Heliyon 2024; 10:e32914. [PMID: 38994113 PMCID: PMC11237975 DOI: 10.1016/j.heliyon.2024.e32914] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2023] [Revised: 06/11/2024] [Accepted: 06/11/2024] [Indexed: 07/13/2024] Open
Abstract
Objective Prostate cancer, marked by a high incidence and mortality rate, presents a significant challenge, especially in the context of castration-resistant prostate cancer (CRPC) with limited treatment options due to drug resistance. This study aims to explore the anti-tumor effects of Xihuang Pills (XHP) on CRPC, focusing on metabolic reprogramming and the Wnt/β-catenin pathway. Methods In vitro and in vivo biofunctional assays were employed to assess the efficacy and mechanisms of XHP. Subcutaneous xenografts of PC3 in mice served as an in vivo model to evaluate XHP's anti-tumor activity. Tumor volume, weight, proliferation, and apoptosis were monitored. Various assays, including CCK8, TUNEL assay, QRT-PCR, and Western Blotting, were conducted to measure metabolic reprogramming, proliferation, apoptosis, and cell cycle in prostate cancer cells. RNA-seq analysis predicted XHP's impact on prostate cancer, validating the expression of Wnt/β-catenin-related proteins and mRNA. Additionally, 58 compounds in XHP were identified via LC-MS/MS, and molecular docking analysis connected these compounds to key genes. Results In vitro and in vivo experiments demonstrated that XHP significantly inhibited CRPC cell viability, induced apoptosis, and suppressed invasion and migration. mRNA sequencing revealed differentially expressed genes, with functional enrichment analysis indicating modulation of key biological processes. XHP treatment downregulated Wnt signaling pathway-related genes, including CCND2, PRKCG, and CCN4. Moreover, XHP effectively inhibited glucose uptake and lactate production, leading to reduced HIF-1α and glycolytic enzymes (GLUT1, HK2, PKM2), suggesting its potential in attenuating the Warburg effect. Molecular docking analysis suggested a plausible interaction between XHP's active compounds and Wnt1 protein, indicating a mechanism through which XHP modulates the Wnt/β-catenin pathway. Conclusion XHP demonstrated remarkable efficacy in suppressing the growth, proliferation, apoptosis, migration, and invasiveness of prostate tumors. The interaction between XHP's active constituents and Wnt1 was evident, leading to the inhibition of Wnt1 and downstream anti-carcinogenic factors, thereby influencing the β-catenin/HIF-1α-mediated glycolysis.
Collapse
Affiliation(s)
- Fengxia Lin
- Department of Andrology, The First Affiliated Hospital of Hunan University of Chinese Medicine, Changsha, 410007, Hunan Province, China
- Department of Cardiovascular, Shenzhen Bao'an Chinese Medicine Hospital, Guangzhou University of Chinese Medicine, Shenzhen, 518000, Guangdong Province, China
- Graduate School of Hunan University of Chinese Medicine, Changsha, 410208, Hunan Province, China
| | - Yan Long
- Department of Andrology, The First Affiliated Hospital of Hunan University of Chinese Medicine, Changsha, 410007, Hunan Province, China
- Graduate School of Hunan University of Chinese Medicine, Changsha, 410208, Hunan Province, China
| | - Mingyue Li
- Department of Pharmacy, Shenzhen Bao'an Chinese Medicine Hospital, Guangzhou University of Chinese Medicine, Shenzhen, 518000, Guangdong Province, China
| | - Changlong Cai
- Department of Urology, Shenzhen Bao'an Chinese Medicine Hospital, Guangzhou University of Chinese Medicine, Shenzhen, 518000, Guangdong Province, China
| | - Yongrong Wu
- School of Traditional Chinese Medicine, Hunan University of Chinese Medicine, Changsha, 410208, Hunan Province, China
| | - Xujun You
- Department of Andrology, Shenzhen Bao'an Chinese Medicine Hospital, Guangzhou University of Chinese Medicine, Shenzhen, 518000, Guangdong Province, China
| | - Xuefei Tian
- College of Integrated Chinese and Western Medicine, Hunan University of Chinese Medicine, Changsha, 410208, Hunan Province, China
| | - Qing Zhou
- Department of Andrology, The First Affiliated Hospital of Hunan University of Chinese Medicine, Changsha, 410007, Hunan Province, China
| |
Collapse
|
32
|
Wang C, Tan J, Jin Y, Li Z, Yang J, Jia Y, Xia Y, Gong B, Dong Q, Zhao Q. A mitochondria-related genes associated neuroblastoma signature - based on bulk and single-cell transcriptome sequencing data analysis, and experimental validation. Front Immunol 2024; 15:1415736. [PMID: 38962012 PMCID: PMC11220120 DOI: 10.3389/fimmu.2024.1415736] [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: 04/11/2024] [Accepted: 06/03/2024] [Indexed: 07/05/2024] Open
Abstract
Background Neuroblastoma (NB), characterized by its marked heterogeneity, is the most common extracranial solid tumor in children. The status and functionality of mitochondria are crucial in regulating NB cell behavior. While the significance of mitochondria-related genes (MRGs) in NB is still missing in key knowledge. Materials and methods This study leverages consensus clustering and machine learning algorithms to construct and validate an MRGs-related signature in NB. Single-cell data analysis and experimental validation were employed to characterize the pivotal role of FEN1 within NB cells. Results MRGs facilitated the classification of NB patients into 2 distinct clusters with considerable differences. The constructed MRGs-related signature and its quantitative indicators, mtScore and mtRisk, effectively characterize the MRGs-related patient clusters. Notably, the MRGs-related signature outperformed MYCN in predicting NB patient prognosis and was adept at representing the tumor microenvironment (TME), tumor cell stemness, and sensitivity to the chemotherapeutic agents Cisplatin, Topotecan, and Irinotecan. FEN1, identified as the most contributory gene within the MRGs-related signature, was found to play a crucial role in the communication between NB cells and the TME, and in the developmental trajectory of NB cells. Experimental validations confirmed FEN1's significant influence on NB cell proliferation, apoptosis, cell cycle, and invasiveness. Conclusion The MRGs-related signature developed in this study offers a novel predictive tool for assessing NB patient prognosis, immune infiltration, stemness, and chemotherapeutic sensitivity. Our findings unveil the critical function of FEN1 in NB, suggesting its potential as a therapeutic target.
Collapse
Affiliation(s)
- Chaoyu Wang
- Department of Pediatric Oncology, Tianjin Medical University Cancer Institute & Hospital, National Clinical Research Center for Cancer, Tianjin's Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin, China
| | - Jiaxiong Tan
- Department of Pediatric Oncology, Tianjin Medical University Cancer Institute & Hospital, National Clinical Research Center for Cancer, Tianjin's Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin, China
| | - Yan Jin
- Department of Pediatric Oncology, Tianjin Medical University Cancer Institute & Hospital, National Clinical Research Center for Cancer, Tianjin's Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin, China
| | - Zongyang Li
- Department of Pediatric Oncology, Tianjin Medical University Cancer Institute & Hospital, National Clinical Research Center for Cancer, Tianjin's Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin, China
| | - Jiaxing Yang
- Department of Pediatric Oncology, Tianjin Medical University Cancer Institute & Hospital, National Clinical Research Center for Cancer, Tianjin's Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin, China
| | - Yubin Jia
- Department of Pediatric Oncology, Tianjin Medical University Cancer Institute & Hospital, National Clinical Research Center for Cancer, Tianjin's Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin, China
| | - Yuren Xia
- Department of Pediatric Oncology, Tianjin Medical University Cancer Institute & Hospital, National Clinical Research Center for Cancer, Tianjin's Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin, China
| | - Baocheng Gong
- Department of Pediatric Oncology, Tianjin Medical University Cancer Institute & Hospital, National Clinical Research Center for Cancer, Tianjin's Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin, China
| | - Qiuping Dong
- Department of Tumor Cell Biology, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Tianjin's Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin, China
| | - Qiang Zhao
- Department of Pediatric Oncology, Tianjin Medical University Cancer Institute & Hospital, National Clinical Research Center for Cancer, Tianjin's Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin, China
| |
Collapse
|
33
|
Li W, Wang Y, Li X, Wu H, Jia L. Dexmedetomidine hydrochloride plus sufentanil citrate inhibits glucose metabolism and epithelial‑mesenchymal transition in human esophageal squamous carcinoma KYSE30 cells by modulating the JAK/STAT3/HIF‑1α axis. Oncol Lett 2024; 27:273. [PMID: 38686357 PMCID: PMC11056934 DOI: 10.3892/ol.2024.14406] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2023] [Accepted: 11/10/2023] [Indexed: 05/02/2024] Open
Abstract
Dexmedetomidine hydrochloride (DEX-HCl) and sufentanil citrate (SFC) are commonly used anesthetic drugs for esophageal cancer (EC) surgery. The present study was performed to investigate the effect of DEX-HCl and SFC treatment on glucose metabolism and epithelial-mesenchymal transition in EC. Cell counting kit-8 (CCK8), clonogenic, wound healing and Transwell migration assays were performed to assess the effects of the DEX-HCl and SFC on KYSE30 cell proliferation, invasion and migration. Changes in lactate and glucose levels in KYSE30 cells were also detected. Western blot analysis was used to determine the protein expression levels of the JAK/STAT signaling pathway and glucose metabolism-related proteins. The results of CCK8, clonogenic and wound healing assays demonstrated that DEX-HCl and SFC inhibited KYSE30 cell proliferation, invasion and migration. Similarly, the combined DEX-HCl and SFC treatment significantly reduced lactate production, ATP production and glucose levels in KYSE30 cells. Western blotting indicated that DEX-HCl and SFC could reduce JAK/STAT and metastasis-related protein expression. Demonstrating a reduction in Hexokinase 2, matrix metallopeptidase 2 and 9, N-cadherin and lactate dehydrogenase A protein expression levels. The effects of DEX-HCl and SFC combined treatment were counteracted by the addition of JAK/STAT pathway activator RO8191, which suggested that DEX-HCl and SFC could serve a role in mediating the JAK/STAT signaling pathway in KYSE30 cells.
Collapse
Affiliation(s)
- Weijing Li
- Department of Anesthesiology, The Fourth Hospital of Hebei Medical University, Shijiazhuang, Hebei 050011, P.R. China
| | - Yong Wang
- Department of Anesthesiology, The Fourth Hospital of Hebei Medical University, Shijiazhuang, Hebei 050011, P.R. China
| | - Xiaolin Li
- Department of Anesthesiology, The Second Hospital of Hebei Medical University, Shijiazhuang, Hebei 050000, P.R. China
| | - Han Wu
- Department of Anesthesiology, The Fourth Hospital of Hebei Medical University, Shijiazhuang, Hebei 050011, P.R. China
| | - Li Jia
- Department of Anesthesiology, The Fourth Hospital of Hebei Medical University, Shijiazhuang, Hebei 050011, P.R. China
| |
Collapse
|
34
|
Lin X, Zheng J, Cai X, Liu L, Jiang S, Liu Q, Sun Y. Glycometabolism and lipid metabolism related genes predict the prognosis of endometrial carcinoma and their effects on tumor cells. BMC Cancer 2024; 24:571. [PMID: 38720279 PMCID: PMC11080313 DOI: 10.1186/s12885-024-12327-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2023] [Accepted: 05/01/2024] [Indexed: 05/12/2024] Open
Abstract
BACKGROUND Glycometabolism and lipid metabolism are critical in cancer metabolic reprogramming. The primary aim of this study was to develop a prognostic model incorporating glycometabolism and lipid metabolism-related genes (GLRGs) for accurate prognosis assessment in patients with endometrial carcinoma (EC). METHODS Data on gene expression and clinical details were obtained from publicly accessible databases. GLRGs were obtained from the Genecards database. Through nonnegative matrix factorization (NMF) clustering, molecular groupings with various GLRG expression patterns were identified. LASSO Cox regression analysis was employed to create a prognostic model. Use rich algorithms such as GSEA, GSVA, xCELL ssGSEA, EPIC,CIBERSORT, MCPcounter, ESTIMATE, TIMER, TIDE, and Oncoppredict to analyze functional pathway characteristics of the forecast signal, immune status, anti-tumor therapy, etc. The expression was assessed using Western blot and quantitative real-time PCR techniques. A total of 113 algorithm combinations were combined to screen out the most significant GLRGs in the signature for in vitro experimental verification, such as colony formation, EdU cell proliferation, wound healing, apoptosis, and Transwell assays. RESULTS A total of 714 GLRGs were found, and 227 of them were identified as prognostic-related genes. And ten GLRGs (AUP1, ESR1, ERLIN2, ASS1, OGDH, BCKDHB, SLC16A1, HK2, LPCAT1 and PGR-AS1) were identified to construct the prognostic model of patients with EC. Based on GLRGs, the risk model's prognosis and independent prognostic value were established. The signature of GLRGs exhibited a robust correlation with the infiltration of immune cells and the sensitivity to drugs. In cytological experiments, we selected HK2 as candidate gene to verify its value in the occurrence and development of EC. Western blot and qRT-PCR revealed that HK2 was substantially expressed in EC cells. According to in vitro experiments, HK2 knockdown can increase EC cell apoptosis while suppressing EC cell migration, invasion, and proliferation. CONCLUSION The GLRGs signature constructed in this study demonstrated significant prognostic value for patients with endometrial carcinoma, thereby providing valuable guidance for treatment decisions.
Collapse
Affiliation(s)
- Xuefen Lin
- Department of Gynecology, Clinical Oncology School of Fujian Medical University, Fujian Cancer Hospital, No.420, Fuma Road, Jin'an District, Fuzhou City, Fujian Province, 350014, P. R. China
- Fujian Provincial Key Laboratory of Tumor Biotherapy, Clinical Oncology School of Fujian Medical University, Fujian Cancer Hospital, Fuzhou, 350014, China
| | - Jianfeng Zheng
- Department of Gynecology, Clinical Oncology School of Fujian Medical University, Fujian Cancer Hospital, No.420, Fuma Road, Jin'an District, Fuzhou City, Fujian Province, 350014, P. R. China
- Fujian Provincial Key Laboratory of Tumor Biotherapy, Clinical Oncology School of Fujian Medical University, Fujian Cancer Hospital, Fuzhou, 350014, China
| | - Xintong Cai
- Department of Gynecology, Clinical Oncology School of Fujian Medical University, Fujian Cancer Hospital, No.420, Fuma Road, Jin'an District, Fuzhou City, Fujian Province, 350014, P. R. China
| | - Li Liu
- Department of Gynecology, Clinical Oncology School of Fujian Medical University, Fujian Cancer Hospital, No.420, Fuma Road, Jin'an District, Fuzhou City, Fujian Province, 350014, P. R. China
| | - Shan Jiang
- Department of Gynecology, Clinical Oncology School of Fujian Medical University, Fujian Cancer Hospital, No.420, Fuma Road, Jin'an District, Fuzhou City, Fujian Province, 350014, P. R. China
- Fujian University of Chinese Medicine, Fuzhou, 350014, China
| | - Qinying Liu
- Fujian Provincial Key Laboratory of Tumor Biotherapy, Clinical Oncology School of Fujian Medical University, Fujian Cancer Hospital, Fuzhou, 350014, China
| | - Yang Sun
- Department of Gynecology, Clinical Oncology School of Fujian Medical University, Fujian Cancer Hospital, No.420, Fuma Road, Jin'an District, Fuzhou City, Fujian Province, 350014, P. R. China.
- Fujian Provincial Key Laboratory of Tumor Biotherapy, Clinical Oncology School of Fujian Medical University, Fujian Cancer Hospital, Fuzhou, 350014, China.
| |
Collapse
|
35
|
Yadav D, Yadav A, Bhattacharya S, Dagar A, Kumar V, Rani R. GLUT and HK: Two primary and essential key players in tumor glycolysis. Semin Cancer Biol 2024; 100:17-27. [PMID: 38494080 DOI: 10.1016/j.semcancer.2024.03.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2024] [Revised: 03/02/2024] [Accepted: 03/09/2024] [Indexed: 03/19/2024]
Abstract
Cancer cells reprogram their metabolism to become "glycolysis-dominant," which enables them to meet their energy and macromolecule needs and enhancing their rate of survival. This glycolytic-dominancy is known as the "Warburg effect", a significant factor in the growth and invasion of malignant tumors. Many studies confirmed that members of the GLUT family, specifically HK-II from the HK family play a pivotal role in the Warburg effect, and are closely associated with glucose transportation followed by glucose metabolism in cancer cells. Overexpression of GLUTs and HK-II correlates with aggressive tumor behaviour and tumor microenvironment making them attractive therapeutic targets. Several studies have proven that the regulation of GLUTs and HK-II expression improves the treatment outcome for various tumors. Therefore, small molecule inhibitors targeting GLUT and HK-II show promise in sensitizing cancer cells to treatment, either alone or in combination with existing therapies including chemotherapy, radiotherapy, immunotherapy, and photodynamic therapy. Despite existing therapies, viable methods to target the glycolysis of cancer cells are currently lacking to increase the effectiveness of cancer treatment. This review explores the current understanding of GLUT and HK-II in cancer metabolism, recent inhibitor developments, and strategies for future drug development, offering insights into improving cancer treatment efficacy.
Collapse
Affiliation(s)
- Dhiraj Yadav
- Amity Institute of Molecular Medicine and Stem Cell Research, Amity University, Noida, Uttar Pradesh 201303, India; Drug Discovery, Jubilant Biosys, Greater Noida, Noida, Uttar Pradesh, India
| | - Anubha Yadav
- Amity Institute of Molecular Medicine and Stem Cell Research, Amity University, Noida, Uttar Pradesh 201303, India
| | - Sujata Bhattacharya
- Amity Institute of Molecular Medicine and Stem Cell Research, Amity University, Noida, Uttar Pradesh 201303, India
| | - Akansha Dagar
- Graduate School of Nanobioscience, Yokohama City University, 22-2 Seto, Kanazawa-Ku, Yokohama 236-0027, Japan
| | - Vinit Kumar
- Amity Institute of Molecular Medicine and Stem Cell Research, Amity University, Noida, Uttar Pradesh 201303, India.
| | - Reshma Rani
- Drug Discovery, Jubilant Biosys, Greater Noida, Noida, Uttar Pradesh, India.
| |
Collapse
|
36
|
Chen Y, Chen L, Wu J, Su D. Hsa_circ_0087862 contributes to the progression of colorectal cancer through regulating miR-512-3p/HK2 axis. Pathol Res Pract 2024; 257:155281. [PMID: 38669868 DOI: 10.1016/j.prp.2024.155281] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/21/2023] [Revised: 03/15/2024] [Accepted: 03/30/2024] [Indexed: 04/28/2024]
Abstract
BACKGROUND Colorectal cancer (CRC) theratened thousands of people every year. Emerging evidences suggested that circular RNAs (circRNAs) were involved in CRC malignancies. However, the underlying mechanisms have yet not been revealed. METHODS Quantitative real-time PCR (qRT-PCR) was used to determine the expression of circ_0087862 and microRNA-512-3p (miR-512-3p). Western blot was performed to measure the protein expression of hexokinase 2 (HK2), B-cell lymphoma-2 (Bcl-2), BCL2-associated X (Bax) and BCL2 antagonist/killer 1 (Bak). Moreover, 3-(4, 5-dimethyl-2-thiazolyl)-2, 5-diphenyl-2-H-tetrazolium bromide (MTT) assay, colony formation and 5-ethynyl-2'-deoxyuridine (EdU) assay were employed to assess CRC cell proliferation. Also, migration/invasion abilities and apoptosis rates were investigated by transwell assay and flow cytometry. Glucose consumption, lactate production and ATP production were detected using the corresponding kits. Dual-luciferase reporter analysis and RNA immunoprecipitation (RIP) experiments were utilized to analyze the target association of miR-512-3p and circ_0087862 or HK2. Finally, xenograft assay was carried out to analyze the function of circ_0087862 in tumor growth in vivo. RESULTS Circ_0087862 expression was elevated in CRC tissues and cells. Circ_0087862 silencing repressed cell viabilities, proliferation, migration/invasion and glycolysis, and reinforced cell apoptosis. However, HK2 could weaken these impacts. Additionally, miR-512-3p targeted HK2, and circ_0087862 could regulate HK2 expression by miR-512-3p. Furthermore, circ_0087862 silencing decreased CRC cell xenograft tumor growth. CONCLUSION Collectively, our data suggested that circ_0087862 knockdown impeded cell viabilities, proliferation, and glycolysis, and contributed to cell apoptosis in CRC, indicating circ_0087862 as a promising tumor promoter.
Collapse
Affiliation(s)
- Yu Chen
- Department of Gastroenterology, The First Affiliated Hospital of Hubei University of Science and Technology, The Central Hospital of Xianning, Xianning, China
| | - Lu Chen
- Department of Gastroenterology, The First Affiliated Hospital of Hubei University of Science and Technology, The Central Hospital of Xianning, Xianning, China
| | - Jieyun Wu
- Department of Gastroenterology, The First Affiliated Hospital of Hubei University of Science and Technology, The Central Hospital of Xianning, Xianning, China
| | - Dazhi Su
- Department of Gastroenterology, The First Affiliated Hospital of Hubei University of Science and Technology, The Central Hospital of Xianning, Xianning, China.
| |
Collapse
|
37
|
Cai Y, Li H, Xie D, Zhu Y. AKR1B10 accelerates glycolysis through binding HK2 to promote the malignant progression of oral squamous cell carcinoma. Discov Oncol 2024; 15:132. [PMID: 38671310 PMCID: PMC11052964 DOI: 10.1007/s12672-024-00996-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/02/2024] [Accepted: 04/23/2024] [Indexed: 04/28/2024] Open
Abstract
BACKGROUND Oral squamous cell carcinoma (OSCC) remains a rampant oral cavity neoplasm with high degree of aggressiveness. Aldo-keto reductase 1B10 (AKR1B10) that is an oxidoreductase dependent on nicotinamide adenine dinucleotide phosphate (NADPH) has been introduced to possess prognostic potential in OSCC. The present work was focused on specifying the involvement of AKR1B10 in the process of OSCC and its latent functional mechanism. METHODS AKR1B10 expression in OSCC tissues and cells were detected by RT-qPCR and Western blot analysis. CCK-8 method, EdU staining, wound healing and transwell assays respectively assayed cell viability, proliferation, migration and invasion. Immunofluorescence staining and Western blot evaluated epithelial mesenchymal transition (EMT). Adenosine triphosphate (ATP) contents, glucose consumption and extracellular acidification rate (ECAR) were measured by relevant commercially available kits and Seahorse XF96 Glycolysis Analyzer, severally. The expressions of proteins associated with metastasis and glycolysis were examined with Western blot. Co-IP assay confirmed the binding between AKR1B10 and hexokinase 2 (HK2). RESULTS It was observed that AKR1B10 expression was increased in OSCC tissues and cells. After AKR1B10 was knocked down, the proliferation, migration, invasion and EMT of OSCC cells were all hampered. Additionally, AKR1B10 silencing suppressed glycolysis and bound to HK2 in OSCC cells. Up-regulation of HK2 partially abolished the hampered glycolysis, proliferation, migration, invasion and EMT of AKR1B10-silenced OSCC cells. CONCLUSION To sum up, AKR1B10 could bind to HK2 to accelerate glycolysis, thereby facilitating the proliferation, migration, invasion and EMT of OSCC cells.
Collapse
Affiliation(s)
- Ye Cai
- Department of Endodontics, Nanjing Stomatological Hospital, Affiliated Hospital of Medical School, Research Institute of Stomatology, Nanjing University, 30 Zhongyang Road, Nanjing, Jiangsu, 210008, People's Republic of China
| | - Huiling Li
- Department of Oral Pathology, Nanjing Stomatological Hospital, Affiliated Hospital of Medical School, Research Institute of Stomatology, Nanjing University, Nanjing, Jiangsu, 210008, People's Republic of China
| | - Diya Xie
- Department of Oral and Maxillofacial Surgery, Nanjing Stomatological Hospital, Affiliated Hospital of Medical School, Research Institute of Stomatology, Nanjing University, Nanjing, Jiangsu, 210008, People's Republic of China
| | - Yanan Zhu
- Department of Endodontics, Nanjing Stomatological Hospital, Affiliated Hospital of Medical School, Research Institute of Stomatology, Nanjing University, 30 Zhongyang Road, Nanjing, Jiangsu, 210008, People's Republic of China.
| |
Collapse
|
38
|
Blázquez-García I, Guerrero L, Cacho-Navas C, Djouder N, Millan J, Paradela A, Carmona-Rodríguez L, Corrales FJ. Molecular Insights of Cholestasis in MDR2 Knockout Murine Liver Organoids. J Proteome Res 2024; 23:1433-1442. [PMID: 38488493 PMCID: PMC11002922 DOI: 10.1021/acs.jproteome.3c00900] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2023] [Revised: 02/29/2024] [Accepted: 03/06/2024] [Indexed: 04/06/2024]
Abstract
MDR3 (multidrug resistance 3) deficiency in humans (MDR2 in mice) causes progressive familial intrahepatic cholestasis type 3 (PFIC3). PFIC3 is a lethal disease characterized by an early onset of intrahepatic cholestasis progressing to liver cirrhosis, a preneoplastic condition, putting individuals at risk of hepatocellular carcinoma (HCC). Hepatocyte-like organoids from MDR2-deficient mice (MDR2KO) were used in this work to study the molecular alterations caused by the deficiency of this transporter. Proteomic analysis by mass spectrometry allowed characterization of 279 proteins that were differentially expressed in MDR2KO compared with wild-type organoids. Functional enrichment analysis indicated alterations in three main cellular functions: (1) interaction with the extracellular matrix, (2) remodeling intermediary metabolism, and (3) cell proliferation and differentiation. The affected cellular processes were validated by orthogonal molecular biology techniques. Our results point to molecular mechanisms associated with PFIC3 that may drive the progression to liver cirrhosis and HCC and suggest proteins and cellular processes that could be targeted for the development of early detection strategies for these severe liver diseases.
Collapse
Affiliation(s)
- Irene Blázquez-García
- Functional
Proteomics Laboratory, Centro Nacional de
Biotecnología (CSIC), Madrid 28049, Spain
| | - Laura Guerrero
- Functional
Proteomics Laboratory, Centro Nacional de
Biotecnología (CSIC), Madrid 28049, Spain
| | | | - Nabil Djouder
- Centro
Nacional de Investigaciones Oncológicas (CNIO), Madrid 28029, Spain
| | - Jaime Millan
- Centro
de Biología Molecular Severo Ochoa (CBMSO), Madrid 28049, Spain
| | - Alberto Paradela
- Functional
Proteomics Laboratory, Centro Nacional de
Biotecnología (CSIC), Madrid 28049, Spain
| | | | - Fernando J. Corrales
- Functional
Proteomics Laboratory, Centro Nacional de
Biotecnología (CSIC), Madrid 28049, Spain
| |
Collapse
|
39
|
Li Z, Sang R, Feng G, Feng Y, Zhang R, Yan X. Microbiological and metabolic pathways analysing the mechanisms of alfalfa polysaccharide and sulfated alfalfa polysaccharide in alleviating obesity. Int J Biol Macromol 2024; 263:130334. [PMID: 38387635 DOI: 10.1016/j.ijbiomac.2024.130334] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2023] [Revised: 02/10/2024] [Accepted: 02/19/2024] [Indexed: 02/24/2024]
Abstract
Alfalfa polysaccharide (AP) and sulfated alfalfa polysaccharide (SAP) exhibit potential for alleviating obesity. This study aimed to analyze the mechanism of action of AP and SAP in alleviating obesity through combined microbiomics and metabolomics. The research selected validated optimal AP and SAP concentration for experiment. The results showed that AP and SAP down-regulated colonic inflammatory gene expression, regulated intestinal pH to normal, and restored intestinal growth. Microbial sequencing showed that AP and SAP altered the microbial composition ratio. AP increased the relative abundance of Muribaculaceae and Romboutsia. SAP increased the relative abundance of Dubosiella, Fecalibaculum and Desulfovibrionaceae. Metabolomic analysis showed that AP regulated steroid hormone biosynthesis, neuroactive ligand-receptor interactions and bile secretion pathways. SAP focuses more on pathways related to amino acid metabolism. Meanwhile, AP and SAP down-regulated the mRNA expression of colonic COX-2, PepT-1 and HK2 and up-regulated the mRNA expression of TPH1. Correlation analysis showed a strong correlation between metabolites and gut bacteria. Dubosiella, Faecalibaculum may be the critical marker flora for polysaccharides to alleviate obesity. This study indicates that AP and SAP alleviate obesity through different pathways and that specific polysaccharide modifications affect characteristic microbial and metabolic pathways, providing new insights into polysaccharide modifications.
Collapse
Affiliation(s)
- Zhiwei Li
- College of Animal Science and Technology, Yangzhou University, Yangzhou, Jiangsu Province 225009, China
| | - Ruxue Sang
- College of Animal Science and Technology, Yangzhou University, Yangzhou, Jiangsu Province 225009, China
| | - Guilan Feng
- College of Animal Science and Technology, Yangzhou University, Yangzhou, Jiangsu Province 225009, China
| | - Yuxi Feng
- College of Animal Science and Technology, Yangzhou University, Yangzhou, Jiangsu Province 225009, China
| | - Ran Zhang
- College of Animal Science and Technology, Yangzhou University, Yangzhou, Jiangsu Province 225009, China
| | - Xuebing Yan
- College of Animal Science and Technology, Yangzhou University, Yangzhou, Jiangsu Province 225009, China; Joint International Research Laboratory of Agriculture and Agri-Product Safety, the Ministry of Education of China, Yangzhou University, Yangzhou, Jiangsu Province 225009, China.
| |
Collapse
|
40
|
Wang Y, Chen X, Yang Y. CircRNA-regulated glucose metabolism in ovarian cancer: an emerging landscape for therapeutic intervention. Clin Transl Oncol 2024; 26:584-596. [PMID: 37578652 DOI: 10.1007/s12094-023-03285-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2023] [Accepted: 07/11/2023] [Indexed: 08/15/2023]
Abstract
Ovarian cancer (OC) has the highest mortality rate among female reproductive system tumours, with limited efficacy of traditional treatments and 5-year survival rates that rarely exceed 40%. Circular RNA (circRNA) is a stable endogenous circular RNA that typically regulates protein expression by binding to downstream miRNA. It has been demonstrated that circRNAs play an important role in the proliferation, migration, and glucose metabolism (such as the Warburg effect) of OC and can regulate the expression of glucose metabolism-related proteins such as GLUT1 and HK2, promoting anaerobic glycolysis of cancer cells, increasing glucose uptake and ATP production, and affecting energy supply and biosynthetic substances to support tumour growth and invasion. This review summarises the formation and characteristics of circRNAs and focuses on their role in regulating glucose metabolism in OC cells and their potential therapeutic value, providing insights for identifying new therapeutic targets.
Collapse
Affiliation(s)
- Yaolong Wang
- Department of Obstetrics and Gynecology, The First Hospital of Lanzhou University, Lanzhou, 730000, China
- Key Laboratory of Gynecological Oncology of Gansu Province, Lanzhou, Gansu, China
- The First Clinical Medical College of Lanzhou University, Lanzhou, Gansu, China
| | - Xi Chen
- Department of Obstetrics and Gynecology, The First Hospital of Lanzhou University, Lanzhou, 730000, China
- Key Laboratory of Gynecological Oncology of Gansu Province, Lanzhou, Gansu, China
- The First Clinical Medical College of Lanzhou University, Lanzhou, Gansu, China
| | - Yongxiu Yang
- Department of Obstetrics and Gynecology, The First Hospital of Lanzhou University, Lanzhou, 730000, China.
- Key Laboratory of Gynecological Oncology of Gansu Province, Lanzhou, Gansu, China.
- The First Clinical Medical College of Lanzhou University, Lanzhou, Gansu, China.
| |
Collapse
|
41
|
Liao M, Yao D, Wu L, Luo C, Wang Z, Zhang J, Liu B. Targeting the Warburg effect: A revisited perspective from molecular mechanisms to traditional and innovative therapeutic strategies in cancer. Acta Pharm Sin B 2024; 14:953-1008. [PMID: 38487001 PMCID: PMC10935242 DOI: 10.1016/j.apsb.2023.12.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2023] [Revised: 11/09/2023] [Accepted: 11/14/2023] [Indexed: 03/17/2024] Open
Abstract
Cancer reprogramming is an important facilitator of cancer development and survival, with tumor cells exhibiting a preference for aerobic glycolysis beyond oxidative phosphorylation, even under sufficient oxygen supply condition. This metabolic alteration, known as the Warburg effect, serves as a significant indicator of malignant tumor transformation. The Warburg effect primarily impacts cancer occurrence by influencing the aerobic glycolysis pathway in cancer cells. Key enzymes involved in this process include glucose transporters (GLUTs), HKs, PFKs, LDHs, and PKM2. Moreover, the expression of transcriptional regulatory factors and proteins, such as FOXM1, p53, NF-κB, HIF1α, and c-Myc, can also influence cancer progression. Furthermore, lncRNAs, miRNAs, and circular RNAs play a vital role in directly regulating the Warburg effect. Additionally, gene mutations, tumor microenvironment remodeling, and immune system interactions are closely associated with the Warburg effect. Notably, the development of drugs targeting the Warburg effect has exhibited promising potential in tumor treatment. This comprehensive review presents novel directions and approaches for the early diagnosis and treatment of cancer patients by conducting in-depth research and summarizing the bright prospects of targeting the Warburg effect in cancer.
Collapse
Affiliation(s)
- Minru Liao
- Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Dahong Yao
- School of Pharmaceutical Sciences, Shenzhen Technology University, Shenzhen 518118, China
| | - Lifeng Wu
- Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Chaodan Luo
- Department of Psychology, University of Southern California, Los Angeles, CA 90089, USA
| | - Zhiwen Wang
- Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu 610041, China
- School of Pharmaceutical Sciences, Shenzhen Technology University, Shenzhen 518118, China
- School of Pharmacy, Shenzhen University Medical School, Shenzhen University, Shenzhen 518055, China
| | - Jin Zhang
- School of Pharmacy, Shenzhen University Medical School, Shenzhen University, Shenzhen 518055, China
| | - Bo Liu
- Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu 610041, China
| |
Collapse
|
42
|
Zhang N, Zhang H, Yang X, Xue Q, Wang Q, Chang R, Zhu L, Chen Z, Liu X. USP14 exhibits high expression levels in hepatocellular carcinoma and plays a crucial role in promoting the growth of liver cancer cells through the HK2/AKT/P62 axis. BMC Cancer 2024; 24:237. [PMID: 38383348 PMCID: PMC10880281 DOI: 10.1186/s12885-024-12009-y] [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: 08/31/2023] [Accepted: 02/15/2024] [Indexed: 02/23/2024] Open
Abstract
BACKGROUND Hepatocellular carcinoma (HCC) is a common malignant tumor with strong invasiveness and poor prognosis. Previous studies have demonstrated the significant role of USP14 in various solid tumors. However, the role of USP14 in the regulation of HCC development and progression remains unclear. METHODS We discovered through GEO and TCGA databases that USP14 may play an important role in liver cancer. Using bioinformatics analysis based on the Cancer Genome Atlas (TCGA) database, we screened and identified USP14 as highly expressed in liver cancer. We detected the growth and metastasis of HCC cells promoted by USP14 through clone formation, cell counting kit 8 assay, Transwell assay, and flow cytometry. In addition, we detected the impact of USP14 on the downstream protein kinase B (AKT) and epithelial-mesenchymal transition (EMT) pathways using western blotting. The interaction mechanism between USP14 and HK2 was determined using immunofluorescence and coimmunoprecipitation (CO-IP) experiments. RESULTS We found that sh-USP14 significantly inhibits the proliferation, invasion, and invasion of liver cancer cells, promoting apoptosis. Further exploration revealed that sh-USP14 significantly inhibited the expression of HK2. Sh-USP14 can significantly inhibit the expression of AKT and EMT signals. Further verification through immunofluorescence and CO-IP experiments revealed that USP14 co-expressed with HK2. Further research has found that USP14 regulates the glycolytic function of liver cancer cells by the deubiquitination of HK2. USP14 regulates the autophagy function of liver cancer cells by regulating the interaction between SQSTM1/P62 and HK2. CONCLUSIONS Our results indicate that USP14 plays a crucial role in the carcinogenesis of liver cancer. We also revealed the protein connections between USP14, HK2, and P62 and elucidated the potential mechanisms driving cancer development. The USP14/HK2/P62 axis may be a new therapeutic biomarker for the diagnosis and treatment of HCC.
Collapse
Affiliation(s)
- Nannan Zhang
- Medical College of Nantong University, Nantong, Jiangsu, 226000, China
- Department of General Surgery, Affiliated Hospital of Nantong University, Nantong, Jiangsu, 226000, China
| | - Hui Zhang
- Department of Radiation Oncology, Affiliated Hospital of Nantong University, Nantong, 226000, China
| | - Xiaobing Yang
- Department of General Surgery, Huaian Hospital of Huaian City, Huaian, Jiangsu, 223200, China
| | - Qiang Xue
- Department of Radiation Oncology, Affiliated Hospital of Nantong University, Nantong, 226000, China
| | - Quhui Wang
- Department of General Surgery, Affiliated Hospital of Nantong University, Nantong, Jiangsu, 226000, China
| | - Renan Chang
- Department of General Surgery, Affiliated Hospital of Nantong University, Nantong, Jiangsu, 226000, China
| | - Lirong Zhu
- Department of General Surgery, Affiliated Hospital of Nantong University, Nantong, Jiangsu, 226000, China
| | - Zhong Chen
- Department of General Surgery, Affiliated Hospital of Nantong University, Nantong, Jiangsu, 226000, China.
| | - Xiancheng Liu
- Department of Radiation Oncology, Affiliated Hospital of Nantong University, Nantong, 226000, China.
| |
Collapse
|
43
|
Juszczak K, Szczepankiewicz W, Walczak K. Synthesis and Primary Activity Assay of Novel Benitrobenrazide and Benserazide Derivatives. Molecules 2024; 29:629. [PMID: 38338374 PMCID: PMC10856005 DOI: 10.3390/molecules29030629] [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: 01/02/2024] [Revised: 01/17/2024] [Accepted: 01/19/2024] [Indexed: 02/12/2024] Open
Abstract
Schiff bases attract research interest due to their applications in chemical synthesis and medicinal chemistry. In recent years, benitrobenrazide and benserazide containing imine moiety have been synthesized and characterized as promising inhibitors of hexokinase 2 (HK2), an enzyme overexpressed in most cancer cells. Benserazide and benitrobenrazide possess a common structural fragment, a 2,3,4-trihydroxybenzaldehyde moiety connected through a hydrazone or hydrazine linker acylated on an N' nitrogen atom by serine or a 4-nitrobenzoic acid fragment. To avoid the presence of a toxicophoric nitro group in the benitrobenrazide molecule, we introduced common pharmacophores such as 4-fluorophenyl or 4-aminophenyl substituents. Modification of benserazide requires the introduction of other endogenous amino acids instead of serine. Herein, we report the synthesis of benitrobenrazide and benserazide analogues and preliminary results of inhibitory activity against HK2 evoked by these structural changes. The derivatives contain a fluorine atom or amino group instead of a nitro group in BNB and exhibit the most potent inhibitory effects against HK2 at a concentration of 1 µM, with HK2 inhibition rates of 60% and 54%, respectively.
Collapse
Affiliation(s)
| | | | - Krzysztof Walczak
- Department of Organic Chemistry, Bioorganic Chemistry and Biotechnology, Faculty of Chemistry, Silesian University of Technology, 44-100 Gliwice, Poland; (K.J.); (W.S.)
| |
Collapse
|
44
|
Lei Y, He L, Li Y, Hou J, Zhang H, Li G. PDLIM1 interacts with HK2 to promote gastric cancer progression through enhancing the Warburg effect via Wnt/β-catenin signaling. Cell Tissue Res 2024; 395:105-116. [PMID: 37930472 DOI: 10.1007/s00441-023-03840-z] [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: 04/27/2023] [Accepted: 10/27/2023] [Indexed: 11/07/2023]
Abstract
PDZ and LIM domain protein 1 (PDLIM1) is a cytoskeletal protein and is associated with the malignant pathological features of several tumors. However, the prognostic value of PDLIM1 and the molecular mechanisms by which it is involved in the metabolism and progression in gastric cancer (GC) are still unclear. The GEPIA database was used to predict the expression and prognosis of PDLIM1 in GC. qRT-PCR and western blot assays were applied to detect the mRNA and protein expression in GC tissues and cells. Loss- and gain-of-function experiments were performed to evaluate the biological role of PDLIM1 in GC cells. The Warburg effect was detected by a battery of glycolytic indicators. The interaction of PDLIM1 and hexokinase 2 (HK2) was determined by a co-immunoprecipitation assay. Furthermore, the modulatory effects of PDLIM1 and HK2 on Wnt/β-catenin signaling were assessed. The results showed that PDLIM1 expression was upregulated in GC tissues and cells and was associated with a poor prognosis for GC patients. PDLIM1 inhibition reduced GC cell proliferation, migration and invasion and promoted cell apoptosis. In the glucose deprivation (GLU-D) condition, the PDLIM1 level was reduced and PDLIM1 overexpression led to an increase in glycolysis. Besides, mechanistic investigation showed that PDLIM1 interacted with HK2 to mediate biological behaviors and the glycolysis of GC through Wnt/β-catenin signaling under glucose deprivation. In conclusion, PDLIM1 interacts with HK2 to promote gastric cancer progression by enhancing the Warburg effect via Wnt/β-catenin signaling.
Collapse
Affiliation(s)
- Yunpeng Lei
- Department of Gastrointestinal Surgery, Peking University Shenzhen Hospital, NO. 1120, Lianhua Road, Futian District, Shenzhen, Guangdong, 518036, China
| | - Lirui He
- Department of Gastrointestinal Surgery, Peking University Shenzhen Hospital, NO. 1120, Lianhua Road, Futian District, Shenzhen, Guangdong, 518036, China
| | - Yue Li
- Department of Gastrointestinal Surgery, Peking University Shenzhen Hospital, NO. 1120, Lianhua Road, Futian District, Shenzhen, Guangdong, 518036, China
| | - Jianing Hou
- Department of Gastrointestinal Surgery, Peking University Shenzhen Hospital, NO. 1120, Lianhua Road, Futian District, Shenzhen, Guangdong, 518036, China
| | - Haoran Zhang
- Department of Gastrointestinal Surgery, Peking University Shenzhen Hospital, NO. 1120, Lianhua Road, Futian District, Shenzhen, Guangdong, 518036, China
| | - Guan Li
- Department of Gastrointestinal Surgery, Peking University Shenzhen Hospital, NO. 1120, Lianhua Road, Futian District, Shenzhen, Guangdong, 518036, China.
| |
Collapse
|
45
|
Yang J, Jiang G, Huang L, Liu Z, Jiang R, Cao G, Cao J, Zhu H, Chen L, Chen X, Pei F. The Long non-coding RNA MALAT1 functions as a competing endogenous RNA to regulate vascular remodeling by sponging miR-145-5p/HK2 in hypertension. Clin Exp Hypertens 2023; 45:2284658. [PMID: 38010958 DOI: 10.1080/10641963.2023.2284658] [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: 09/28/2023] [Accepted: 11/11/2023] [Indexed: 11/29/2023]
Abstract
Long non-coding RNAs (LncRNAs) have been found to play a regulatory role in the pathophysiology of vascular remodeling-associated illnesses through the lncRNA-microRNA (miRNA) regulation axis. LncRNA metastasis-associated lung adenocarcinoma transcript 1 (MALAT1) is thought to be involved in proliferation, migration, apoptosis, and calcification of vascular smooth muscle cells (VSMCs). The purpose of this study was to investigate the regulatory role of MALAT1 on vascular remodeling in hypertension. Our data indicate that the expression of MALAT1 is significantly upregulated in hypertensive aortic smooth muscle. Knockdown of MALAT1 inhibited the proliferation, migration, and phenotypic transition of VSMCs induced by Ang II. Bioinformatics analysis was used to predict the complementary binding of miR-145-5p to the 3'-untranslated region of MALAT1. Besides, the expressions of MALAT1 and miR-145-5p were negatively correlated, while luciferase reporter assays and RNA immunoprecipitation assay validated the interaction between miR-145-5p and MALAT1. The proliferation, migration and phenotypic transformation of VSMCs induced by overexpression of MALAT1 were reversed in the presence of miR-145-5p. Furthermore, we verified that miR-145-5p could directly target and bind to hexokinase 2 (HK2) mRNA, and that HK2 expression was negatively correlated with miR-145-5p in VSMCs. Knockdown of HK2 significantly inhibited the effects of overexpression of MALAT1 on Ang II-induced VSMCs proliferation, migration and phenotypic transformation. Taken together, the MALAT1/miR-145-5p/HK2 axis may play a critical regulatory role in the vascular remodeling of VSMCs in hypertension.
Collapse
Affiliation(s)
- Jiangyong Yang
- Department of Cardiology, Ganzhou Hospital of Guangdong Provincial People's Hospital, Ganzhou Municipal Hospital, Ganzhou, Jiangxi, China
| | - Guojun Jiang
- Department of Pharmacy, The Second Affiliated Hospital of Guilin Medical University, Guilin, Guangxi, China
| | - Ling Huang
- Department of Cardiology, Ganzhou Hospital of Guangdong Provincial People's Hospital, Ganzhou Municipal Hospital, Ganzhou, Jiangxi, China
| | - Zhongyi Liu
- Department of Medical Research, Ganzhou Hospital of Guangdong Provincial People's Hospital, Ganzhou Municipal Hospital, Ganzhou, Jiangxi, China
| | - Rengui Jiang
- Department of Cardiology, Ganzhou Hospital of Guangdong Provincial People's Hospital, Ganzhou Municipal Hospital, Ganzhou, Jiangxi, China
| | - Gang Cao
- Department of Cardiology, Ganzhou Hospital of Guangdong Provincial People's Hospital, Ganzhou Municipal Hospital, Ganzhou, Jiangxi, China
| | - Jun Cao
- Department of Cardiology, Ganzhou Hospital of Guangdong Provincial People's Hospital, Ganzhou Municipal Hospital, Ganzhou, Jiangxi, China
| | - Hengqing Zhu
- Department of Cardiology, Ganzhou Hospital of Guangdong Provincial People's Hospital, Ganzhou Municipal Hospital, Ganzhou, Jiangxi, China
| | - Lemei Chen
- Department of Medical Research, Ganzhou Hospital of Guangdong Provincial People's Hospital, Ganzhou Municipal Hospital, Ganzhou, Jiangxi, China
| | - Xiaoming Chen
- Department of Medical Research, Ganzhou Hospital of Guangdong Provincial People's Hospital, Ganzhou Municipal Hospital, Ganzhou, Jiangxi, China
| | - Fang Pei
- Department of Cardiology, Ganzhou Hospital of Guangdong Provincial People's Hospital, Ganzhou Municipal Hospital, Ganzhou, Jiangxi, China
| |
Collapse
|
46
|
Guo Y, Lu X, Zhou Y, Chen WH, Tam KY. Combined inhibition of pyruvate dehydrogenase kinase 1 and hexokinase 2 induces apoptsis in non-small cell lung cancer cell models. Exp Cell Res 2023; 433:113830. [PMID: 37913974 DOI: 10.1016/j.yexcr.2023.113830] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2023] [Revised: 09/25/2023] [Accepted: 10/23/2023] [Indexed: 11/03/2023]
Abstract
Many cancer cells exhibit enhanced glycolysis, which is seen as one of the hallmark metabolic alterations, known as Warburg effect. Substantial evidence shows that upregulated glycolytic enzymes are often linked to malignant growth. Using glycolytic inhibitors for anticancer treatment has become appealing in recent years for therapeutic intervention in cancers with highly glycolytic characteristic, including non-small cell lung cancer (NSCLC). In this work, we studied the anticancer effects and the underlying mechanisms of combination of benzerazide hydrocholoride (Benz), a hexokinase 2 (HK2) inhibitor and 64, a pyruvate dehydrogenase kinase 1 (PDK1) inhibitor, in several NSCLC cell lines. We found that combination of Benz and 64 exhibited strong synergistic anticancer effects in NCI-H1975, HCC827, NCI-H1299 and SK-LU-1 cell lines. With this combination treatment, we observed changes of certain mechanistic determinants associated with metabolic stress caused by glycolysis restriction, such as mitochondrial membrane potential depolarization, overproduction of reactive oxygen species [1], activation of AMPK and down-regulation of mTOR, which contributed to enhanced apoptosis. Moreover, Benz and 64 together significantly suppressed the tumor growth in HCC827 cell mouse xenograft model. Taken together, our study may suggest that combined inhibition of HK2 and PDK1 using Benz and 64 could be a viable anticancer strategy for NSCLC.
Collapse
Affiliation(s)
- Yizhen Guo
- Faculty of Health Sciences, University of Macau, Taipa, Macau
| | - Xianchen Lu
- School of Biotechnology and Health Sciences, Wuyi University, Jiangmen, Guangdong, 529020, PR China
| | - Yan Zhou
- Faculty of Health Sciences, University of Macau, Taipa, Macau
| | - Wen-Hua Chen
- School of Biotechnology and Health Sciences, Wuyi University, Jiangmen, Guangdong, 529020, PR China.
| | - Kin Yip Tam
- Faculty of Health Sciences, University of Macau, Taipa, Macau.
| |
Collapse
|
47
|
Zheng C, Li R, Zheng S, Fang H, Xu M, Zhong L. The knockdown of lncRNA DLGAP1-AS2 suppresses osteosarcoma progression by inhibiting aerobic glycolysis via the miR-451a/HK2 axis. Cancer Sci 2023; 114:4747-4762. [PMID: 37817462 PMCID: PMC10728003 DOI: 10.1111/cas.15989] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2023] [Revised: 08/30/2023] [Accepted: 09/20/2023] [Indexed: 10/12/2023] Open
Abstract
Osteosarcoma (OS) is one of the most aggressive bone tumors worldwide. Emerging documents have shown that long noncoding RNAs (lncRNAs) elicit crucial regulatory functions in the process of tumorigenesis. LncRNA DLGAP1-AS2 is recognized as a regulator in several types of cancers, but its biological functions and molecular mechanisms in OS remain to be elucidated. RT-qPCR and In situ hybridization (ISH) were used to evaluate DLGAP1-AS2 expression in OS samples. Western blotting was used for the measurement of the protein levels of hexokinase 2 (HK2) and epithelial-mesenchymal transition (EMT)-related markers. The proliferation of OS cells was determined using a CCK-8 assay and EdU assay. TUNEL assay and flow cytometry were performed to assess OS cell apoptosis. Glucose metabolism in vitro assays were used. The binding relations among miR-451a, HK2, and DLGAP1-AS2 were validated by luciferase reporter assay. The cellular distribution of DLGAP1-AS2 in OS cells was determined by FISH and subcellular fractionation assays. Mouse xenograft models were established to perform the experiments in vivo. We found that DLGAP1-AS2 expression was upregulated in OS tissues and cells. Downregulation of DLGAP1-AS2 expression suppressed the malignancy of OS cells by restraining cell proliferation, the EMT process, invasiveness, migration, and aerobic glycolysis and accelerating apoptotic behaviors. Of note, silenced DLGAP1-AS2 restrained tumor growth and metastasis in vivo. However, DLGAP1-AS2 overexpression accelerated the progression of OS. We further found that DLGAP1-AS2 upregulation was induced by hypoxia and low glucose. Additionally, DLGAP1-AS2 bound to miR-451a to upregulate HK2 expression. Rescue assays revealed that the DLGAP1-AS2/miR-451a/HK2 axis contributed to OS cell malignancy by promoting aerobic glucose metabolism. Overall, these findings revealed a new regulatory pathway where DLGAP1-AS2 upregulated HK2 expression by sponging miR-451a to accelerate OS development.
Collapse
Affiliation(s)
- Changjun Zheng
- Department of OrthopedicsThe Second Hospital of Jilin UniversityChangchunChina
| | - Ronghang Li
- Department of OrthopedicsThe Second Hospital of Jilin UniversityChangchunChina
| | - Shuang Zheng
- Department of OrthopedicsThe Second Hospital of Jilin UniversityChangchunChina
| | - Hongjuan Fang
- Department of Electric DiagnosticThe Fourth Hospital of Jilin UniversityChangchunChina
| | - Meng Xu
- Department of OrthopedicsThe Second Hospital of Jilin UniversityChangchunChina
| | - Lei Zhong
- Department of OrthopedicsThe Second Hospital of Jilin UniversityChangchunChina
| |
Collapse
|
48
|
Li C, Liu FY, Shen Y, Tian Y, Han FJ. Research progress on the mechanism of glycolysis in ovarian cancer. Front Immunol 2023; 14:1284853. [PMID: 38090580 PMCID: PMC10715264 DOI: 10.3389/fimmu.2023.1284853] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2023] [Accepted: 11/10/2023] [Indexed: 12/18/2023] Open
Abstract
Glycolysis is the preferred energy metabolism pathway in cancer cells even when the oxygen content is sufficient. Through glycolysis, cancer cells convert glucose into pyruvic acid and then lactate to rapidly produce energy and promote cancer progression. Changes in glycolysis activity play a crucial role in the biosynthesis and energy requirements of cancer cells needed to maintain growth and metastasis. This review focuses on ovarian cancer and the significance of key rate-limiting enzymes (hexokinase, phosphofructokinase, and pyruvate kinase, related signaling pathways (PI3K-AKT, Wnt, MAPK, AMPK), transcription regulators (HIF-1a), and non-coding RNA in the glycolytic pathway. Understanding the relationship between glycolysis and these different mechanisms may provide new opportunities for the future treatment of ovarian cancer.
Collapse
Affiliation(s)
- Chan Li
- Heilongjiang University of Traditional Chinese Medicine (TCM), Harbin, Heilongjiang, China
| | - Fang-Yuan Liu
- Heilongjiang University of Traditional Chinese Medicine (TCM), Harbin, Heilongjiang, China
| | - Ying Shen
- Heilongjiang University of Traditional Chinese Medicine (TCM), Harbin, Heilongjiang, China
- The First Affiliated Hospital of Heilongjiang University of Traditional Chinese Medicine (TCM), Harbin, Heilongjiang, China
| | - Yuan Tian
- Zhejiang University of Chinese Medicine, Hangzhou, Zhejiang, China
| | - Feng-Juan Han
- The First Affiliated Hospital of Heilongjiang University of Traditional Chinese Medicine (TCM), Harbin, Heilongjiang, China
| |
Collapse
|
49
|
Shuvalov O, Kirdeeva Y, Daks A, Fedorova O, Parfenyev S, Simon HU, Barlev NA. Phytochemicals Target Multiple Metabolic Pathways in Cancer. Antioxidants (Basel) 2023; 12:2012. [PMID: 38001865 PMCID: PMC10669507 DOI: 10.3390/antiox12112012] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2023] [Revised: 11/09/2023] [Accepted: 11/15/2023] [Indexed: 11/26/2023] Open
Abstract
Cancer metabolic reprogramming is a complex process that provides malignant cells with selective advantages to grow and propagate in the hostile environment created by the immune surveillance of the human organism. This process underpins cancer proliferation, invasion, antioxidant defense, and resistance to anticancer immunity and therapeutics. Perhaps not surprisingly, metabolic rewiring is considered to be one of the "Hallmarks of cancer". Notably, this process often comprises various complementary and overlapping pathways. Today, it is well known that highly selective inhibition of only one of the pathways in a tumor cell often leads to a limited response and, subsequently, to the emergence of resistance. Therefore, to increase the overall effectiveness of antitumor drugs, it is advisable to use multitarget agents that can simultaneously suppress several key processes in the tumor cell. This review is focused on a group of plant-derived natural compounds that simultaneously target different pathways of cancer-associated metabolism, including aerobic glycolysis, respiration, glutaminolysis, one-carbon metabolism, de novo lipogenesis, and β-oxidation of fatty acids. We discuss only those compounds that display inhibitory activity against several metabolic pathways as well as a number of important signaling pathways in cancer. Information about their pharmacokinetics in animals and humans is also presented. Taken together, a number of known plant-derived compounds may target multiple metabolic and signaling pathways in various malignancies, something that bears great potential for the further improvement of antineoplastic therapy.
Collapse
Affiliation(s)
- Oleg Shuvalov
- Institute of Cytology of the Russian Academy of Sciences, St. Petersburg 194064, Russia; (Y.K.); (A.D.); (O.F.)
| | - Yulia Kirdeeva
- Institute of Cytology of the Russian Academy of Sciences, St. Petersburg 194064, Russia; (Y.K.); (A.D.); (O.F.)
| | - Alexandra Daks
- Institute of Cytology of the Russian Academy of Sciences, St. Petersburg 194064, Russia; (Y.K.); (A.D.); (O.F.)
| | - Olga Fedorova
- Institute of Cytology of the Russian Academy of Sciences, St. Petersburg 194064, Russia; (Y.K.); (A.D.); (O.F.)
| | - Sergey Parfenyev
- Institute of Cytology of the Russian Academy of Sciences, St. Petersburg 194064, Russia; (Y.K.); (A.D.); (O.F.)
| | - Hans-Uwe Simon
- Institute of Pharmacology, University of Bern, 3010 Bern, Switzerland;
- Institute of Fundamental Medicine and Biology, Kazan Federal University, Kazan 420008, Russia
| | - Nickolai A. Barlev
- Institute of Cytology of the Russian Academy of Sciences, St. Petersburg 194064, Russia; (Y.K.); (A.D.); (O.F.)
- Institute of Fundamental Medicine and Biology, Kazan Federal University, Kazan 420008, Russia
- Department of Biomedical Sciences, School of Medicine, Nazarbayev University, Astana 20000, Kazakhstan
| |
Collapse
|
50
|
Codocedo JF, Mera-Reina C, Lin PBC, Puntambekar SS, Casali BT, Jury N, Martinez P, Lasagna-Reeves CA, Landreth GE. Therapeutic targeting of immunometabolism in Alzheimer's disease reveals a critical reliance on Hexokinase 2 dosage on microglial activation and disease progression. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.11.11.566270. [PMID: 38014106 PMCID: PMC10680613 DOI: 10.1101/2023.11.11.566270] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/29/2023]
Abstract
Microgliosis and neuroinflammation are prominent features of Alzheimer's disease (AD). Disease-responsive microglia meet their increased energy demand by reprogramming metabolism, specifically, switching to favor glycolysis over oxidative phosphorylation. Thus, targeting of microglial immunometabolism might be of therapeutic benefit for treating AD, providing novel and often well understood immune pathways and their newly recognized actions in AD. We report that in the brains of 5xFAD mice and postmortem brains of AD patients, we found a significant increase in the levels of Hexokinase 2 (HK2), an enzyme that supports inflammatory responses by rapidly increasing glycolysis. Moreover, binding of HK2 to mitochondria has been reported to regulate inflammation by preventing mitochondrial dysfunction and NLRP3 inflammasome activation, suggesting that its inflammatory role extends beyond its glycolytic activity. Here we report, that HK2 antagonism selectively affects microglial phenotypes and disease progression in a gene-dose dependent manner. Paradoxically, complete loss of HK2 fails to improve AD progression by exacerbating inflammasome activity while its haploinsufficiency results in reduced pathology and improved cognition in the 5XFAD mice. We propose that the partial antagonism of HK2, is effective in slowed disease progression and inflammation through a non-metabolic mechanism associated with the modulation of NFKβ signaling, through its cytosolic target IKBα. The complete loss of HK2 affects additional inflammatory mechanisms associated to mitochondrial dysfunction.
Collapse
Affiliation(s)
- Juan F Codocedo
- Stark Neurosciences Research Institute, Indiana University, School of Medicine, Indianapolis, IN 46202, USA
| | - Claudia Mera-Reina
- Stark Neurosciences Research Institute, Indiana University, School of Medicine, Indianapolis, IN 46202, USA
| | - Peter Bor-Chian Lin
- Stark Neurosciences Research Institute, Indiana University, School of Medicine, Indianapolis, IN 46202, USA
| | - Shweta S Puntambekar
- Stark Neurosciences Research Institute, Indiana University, School of Medicine, Indianapolis, IN 46202, USA
| | - Brad T Casali
- Stark Neurosciences Research Institute, Indiana University, School of Medicine, Indianapolis, IN 46202, USA
| | - Nur Jury
- Stark Neurosciences Research Institute, Indiana University, School of Medicine, Indianapolis, IN 46202, USA
| | - Pablo Martinez
- Stark Neurosciences Research Institute, Indiana University, School of Medicine, Indianapolis, IN 46202, USA
| | - Cristian A Lasagna-Reeves
- Stark Neurosciences Research Institute, Indiana University, School of Medicine, Indianapolis, IN 46202, USA
| | - Gary E Landreth
- Stark Neurosciences Research Institute, Indiana University, School of Medicine, Indianapolis, IN 46202, USA
| |
Collapse
|