1
|
Xu Y, Hao X, Ren Y, Xu Q, Liu X, Song S, Wang Y. Research progress of abnormal lactate metabolism and lactate modification in immunotherapy of hepatocellular carcinoma. Front Oncol 2023; 12:1063423. [PMID: 36686771 PMCID: PMC9853001 DOI: 10.3389/fonc.2022.1063423] [Citation(s) in RCA: 8] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2022] [Accepted: 12/19/2022] [Indexed: 01/09/2023] Open
Abstract
Tumors meet their energy, biosynthesis, and redox demands through metabolic reprogramming. This metabolic abnormality results in elevated levels of metabolites, particularly lactate, in the tumor microenvironment. Immune cell reprogramming and cellular plasticity mediated by lactate and lactylation increase immunosuppression in the tumor microenvironment and are emerging as key factors in regulating tumor development, metastasis, and the effectiveness of immunotherapies such as immune checkpoint inhibitors. Reprogramming of glucose metabolism and the "Warburg effect" in hepatocellular carcinoma (HCC) lead to the massive production and accumulation of lactate, so lactate modification in tumor tissue is likely to be abnormal as well. This article reviews the immune regulation of abnormal lactate metabolism and lactate modification in hepatocellular carcinoma and the therapeutic strategy of targeting lactate-immunotherapy, which will help to better guide the medication and treatment of patients with hepatocellular carcinoma.
Collapse
Affiliation(s)
- Yiwei Xu
- Marine College, Shandong University, Weihai, China
| | - Xiaodong Hao
- Department of Clinical Laboratory, The Second Hospital, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, China
| | - Yidan Ren
- Department of Clinical Laboratory, The Second Hospital, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, China
| | - Qinchen Xu
- Department of Clinical Laboratory, The Second Hospital, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, China
| | - Xiaoyan Liu
- Department of Clinical Laboratory, The Second Hospital, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, China
| | - Shuliang Song
- Marine College, Shandong University, Weihai, China,*Correspondence: Shuliang Song, ; Yunshan Wang,
| | - Yunshan Wang
- Department of Clinical Laboratory, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, China,*Correspondence: Shuliang Song, ; Yunshan Wang,
| |
Collapse
|
2
|
Why may citrate sodium significantly increase the effectiveness of transarterial chemoembolization in hepatocellular carcinoma? Drug Resist Updat 2021; 59:100790. [PMID: 34924279 DOI: 10.1016/j.drup.2021.100790] [Citation(s) in RCA: 28] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2021] [Revised: 09/01/2021] [Accepted: 09/04/2021] [Indexed: 02/07/2023]
Abstract
Hepatocellular carcinoma (HCC) represents the third cause of cancer death in men worldwide, and its increasing incidence can be explained by the increasing occurrence of non-alcoholic steatohepatitis (NASH). HCC prognosis is poor, as its 5-year overall survival is approximately 18 % and most cases are diagnosed at an inoperable advanced stage. Moreover, tumor sensitivity to conventional chemotherapeutics (particularly to cisplatin-based regimen), trans-arterial chemoembolization (cTACE), tyrosine kinase inhibitors, anti-angiogenic molecules and immune checkpoint inhibitors is limited. Oncogenic signaling pathways, such as HIF-1α and RAS/PI3K/AKT, may provoke drug resistance by enhancing the aerobic glycolysis ("Warburg effect") in cancer cells. Indeed, this metabolism, which promotes cancer cell development and aggressiveness, also induces extracellular acidity. In turn, this acidity promotes the protonation of drugs, hence abrogating their internalization, since they are most often weakly basic molecules. Consequently, targeting the Warburg effect in these cancer cells (which in turn would reduce the extracellular acidification) could be an effective strategy to increase the delivery of drugs into the tumor. Phosphofructokinase-1 (PFK1) and its activator PFK2 are the main regulators of glycolysis, and they also couple the enhancement of glycolysis to the activation of key signaling cascades and cell cycle progression. Therefore, targeting this "Gordian Knot" in HCC cells would be of crucial importance. Here, we suggest that this could be achieved by citrate administration at high concentration, because citrate is a physiologic inhibitor of PFK1 and PFK2. As shown in various in vitro studies, including HCC cell lines, administration of high concentrations of citrate inhibits PFK1 and PFK2 (and consequently glycolysis), decreases ATP production, counteracts HIF-1α and PI3K/AKT signaling, induces apoptosis, and sensitizes cells to cisplatin treatment. Administration of high concentrations of citrate in animal models (including Ras-driven tumours) has been shown to effectively inhibit cancer growth, reverse cell dedifferentiation, and neutralize intratumor acidity, without apparent toxicity in animal studies. Citrate may also induce a rapid secretion of pro-inflammatory cytokines by macrophages, and it could favour the destruction of cancer stem cells (CSCs) sustaining tumor recurrence. Consequently, this "citrate strategy" could improve the tumor sensitivity to current treatments of HCC by reducing the extracellular acidity, thus enhancing the delivery of chemotherapeutic drugs into the tumor. Therefore, we propose that this strategy should be explored in clinical trials, in particular to enhance cTACE effectiveness.
Collapse
|
3
|
Hsu YK, Chen HY, Wu CC, Huang YC, Hsieh CP, Su PF, Huang YF. Butein induces cellular senescence through reactive oxygen species-mediated p53 activation in osteosarcoma U-2 OS cells. ENVIRONMENTAL TOXICOLOGY 2021; 36:773-781. [PMID: 33325610 DOI: 10.1002/tox.23079] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/17/2020] [Accepted: 12/06/2020] [Indexed: 06/12/2023]
Abstract
Butein is a flavonoid isolated from various medicinal plants. It is known to have different biological activities including anti-inflammation, anti-adipogenesis, and anti-angiogenesis. In the study, we demonstrated the anti-proliferative effect of butein in human osteosarcoma U-2 OS cells. Our data showed that butein significantly suppressed the viability and colony formation ability of U-2 OS cells. Further experiments revealed butein exposure resulted in a cell cycle arrest at S and G2/M phase in U-2 OS cells. Importantly, we found that butein activated the tumor suppressor p53, and trigged a p53-dependent senescence in U-2 OS cells. Knockdown of p53 suppressed the senescence and rescued the viability in butein-treated U-2 OS cells. Furthermore, we observed that butein exposure significantly enhanced reactive oxygen species (ROS) levels in U-2 OS cells. Co-administration of the ROS inhibitor NAC largely abolished the up-regulated p53 protein level, and rescued the suppressed viability and colony formation ability in butein-exposed U-2 OS cells. Taken together, our data proposed the increased ROS by butein exposure activated p53, and the activated p53 was involved in the anti-proliferative effect of butein via inducing senescence in U-2 OS cells. This report suggests that butein is a promising candidate for cancer therapy against osteosarcoma.
Collapse
Affiliation(s)
- Yung-Ken Hsu
- Department of Orthopedic Surgery, Changhua Christian Hospital, Changhua, Taiwan
| | - Hsuan-Ying Chen
- Orthopedics and Sports Medicine Laboratory, Changhua Christian Hospital, Changhua, Taiwan
| | - Chia-Chieh Wu
- Department of Orthopedic Surgery, Changhua Christian Hospital, Changhua, Taiwan
- School of Medicine, Kaohsiung Medical University, Kaohsiung, Taiwan
| | - Ying-Chih Huang
- Department of Research, Changhua Christian Hospital, Changhua, Taiwan
| | - Cheng-Pu Hsieh
- Department of Orthopedic Surgery, Changhua Christian Hospital, Changhua, Taiwan
- Orthopedics and Sports Medicine Laboratory, Changhua Christian Hospital, Changhua, Taiwan
| | - Po-Feng Su
- Department of Orthopedic Surgery, Changhua Christian Hospital, Changhua, Taiwan
| | - Yi-Fu Huang
- Orthopedics and Sports Medicine Laboratory, Changhua Christian Hospital, Changhua, Taiwan
| |
Collapse
|
4
|
Hu M, Fu Q, Jing C, Zhang X, Qin T, Pan Y. LncRNA HOTAIR knockdown inhibits glycolysis by regulating miR-130a-3p/HIF1A in hepatocellular carcinoma under hypoxia. Biomed Pharmacother 2020; 125:109703. [PMID: 32062551 DOI: 10.1016/j.biopha.2019.109703] [Citation(s) in RCA: 44] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2019] [Revised: 11/14/2019] [Accepted: 11/24/2019] [Indexed: 12/20/2022] Open
Abstract
High rate of glycolysis supports hepatocellular carcinoma (HCC) cell growth even in a hypoxic environment. However, the mechanism underlying glycolysis under hypoxia remains largely unknown. Long noncoding RNAs (lncRNAs) play essential roles in regulating glucose metabolism in cancers. This study aimed to explore the role of lncRNA homeobox transcript antisense RNA (HOTAIR) in HCC glycolysis under hypoxia. Thirty-eight HCC patients were recruited. HepG2 and Huh7 cells were used for study in vitro. The expression levels of HOTAIR, microRNA-130a-3p (miR-130a-3p) and hypoxia inducible factor 1 alpha (HIF1A) were measured by quantitative real-time polymerase chain reaction and western blot, respectively. The glycolysis under hypoxia (1 % O2) condition was investigated by glucose consumption, lactate production and hexokinase 2 (HK2) level. The target interaction between miR-130a-3p and HOTIR or HIF1A was analyzed by bioinformatics analysis, luciferase assay, RNA pull-down and RNA immunoprecipitation. We found that HOTAIR expression was enhanced in HCC tissues and cells. Under hypoxia condition, HOTAIR expression was increased and its knockdown inhibited glycolysis in HCC cells. HOTAIR was validated as a decoy of miR-130a-3p and miR-130a-3p deficiency reversed the suppressive effect of HOTAIR silence on glycolysis under hypoxia. HIF1A was indicated as a target of miR-130a-3p and miR-130a-3p overexpression repressed glycolysis under hypoxia by targeting HIF1A. Moreover, HIF1A expression was regulated by HOTAIR and miR-130a-3p. In conclusion, knockdown of HOTAIR suppressed glycolysis by regulating miR-130a-3p and HIF1A in HCC cells treated by hypoxia, elucidating a novel mechanism in HCC glycolysis.
Collapse
Affiliation(s)
- Mingxing Hu
- Department of Hepatobiliary and Pancreatic Surgery, the Henan Provincial People's Hospital, Zhengzhou, Henan, China; Department of Hepatobiliary and Pancreatic Surgery, the People's Hospital of Zhengzhou University, Zhengzhou, Henan, China; Department of Hepatobiliary and Pancreatic Surgery, the School of Clinical Medicine, Henan University, Zhengzhou, Henan, China
| | - Qiang Fu
- Department of Hepatobiliary and Pancreatic Surgery, the Henan Provincial People's Hospital, Zhengzhou, Henan, China; Department of Hepatobiliary and Pancreatic Surgery, the People's Hospital of Zhengzhou University, Zhengzhou, Henan, China; Department of Hepatobiliary and Pancreatic Surgery, the School of Clinical Medicine, Henan University, Zhengzhou, Henan, China
| | - Chan Jing
- Department of Hepatobiliary and Pancreatic Surgery, the Henan Provincial People's Hospital, Zhengzhou, Henan, China; Department of Hepatobiliary and Pancreatic Surgery, the People's Hospital of Zhengzhou University, Zhengzhou, Henan, China; Department of Hepatobiliary and Pancreatic Surgery, the School of Clinical Medicine, Henan University, Zhengzhou, Henan, China
| | - Xu Zhang
- Department of Hepatobiliary and Pancreatic Surgery, the Henan Provincial People's Hospital, Zhengzhou, Henan, China; Department of Hepatobiliary and Pancreatic Surgery, the People's Hospital of Zhengzhou University, Zhengzhou, Henan, China; Department of Hepatobiliary and Pancreatic Surgery, the School of Clinical Medicine, Henan University, Zhengzhou, Henan, China
| | - Tao Qin
- Department of Hepatobiliary and Pancreatic Surgery, the Henan Provincial People's Hospital, Zhengzhou, Henan, China; Department of Hepatobiliary and Pancreatic Surgery, the People's Hospital of Zhengzhou University, Zhengzhou, Henan, China; Department of Hepatobiliary and Pancreatic Surgery, the School of Clinical Medicine, Henan University, Zhengzhou, Henan, China
| | - Yanfeng Pan
- Department of Infectious Diseases, the First Affiliated Hospital of Zhengzhou University, No. Jianshe East Road, Zhengzhou 450000, Henan, China.
| |
Collapse
|
5
|
Ma D, Li Y, Xiao W, Peng L, Wang L, Liao Z, Hu L. Achyranthes bidentata extract protects chondrocytes functions through suppressing glycolysis and apoptosis via MAPK/AKT signaling axis. Am J Transl Res 2020; 12:142-152. [PMID: 32051743 PMCID: PMC7013231] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2019] [Accepted: 12/06/2019] [Indexed: 06/10/2023]
Abstract
Osteoarthritis (OA) is considered to be a joint-associated disorder and one of leading reasons for disability, however, potential mechanism has never been clarified. The purpose of this research was to evaluate protective-effects of Achyranthes Bidentata extracts (ABE) on chondrocytes function in osteoarthritis. We performed a systematic investigation of transcriptional and proteomic landscapes to identify the underlying mechanisms behind effects of ABE on chondrocytic functions. OA animal models were generated in the present research. Chondrocytes were isolated and cultured, and then prepared for GeneChip analysis. Two-dimensional gel electrophoresis and LC-MS/MS analysis were conducted to analyze samples. Quantitative real-time PCR (qRT-PCR) and western blotting were used to evaluate expression of protein kinase B (AKT), β-tubulin and β-action. Apoptosis and glycolysis pathway were significantly compromised in chondrocytes with ABE stimulation as revealed by both transcriptional and proteomic data. Consistently, ABE suppressed chondrocytes apoptosis and glycolytic activity in vitro through modulating multiple genes, such as Plk2, Casp1/12 and Cers1 as well as Pkm2, Eno1/3 and Pgk2. Mechanically, ABE activated MAPK signaling pathway and suppressed AKT signaling pathway, therefore, reducing the glycolysis to provide survival benefits. We extended our analysis by verifying insulin-like growth factor 1 (IGF-1) and MAP kinase 1 (MEK1) in chondrocytes function. Depletion of either IGF-1 or MEK1 impaired AKT expression and phosphorylation, leading to the enhanced chondrocyte apoptosis and reduced cell proliferation. In conclusion, our study provided systematic view and molecular basis for ABE to serve as potential intervention of OA via suppressing AKT signaling.
Collapse
Affiliation(s)
- Dujun Ma
- Shenzhen Traditional Chinese Medicine HospitalShenzhen 518033, China
| | - Yuan Li
- The Second People’s Hospital of Futian District ShenzhenShenzhen 518000, China
| | - Wei Xiao
- Shenzhen Traditional Chinese Medicine HospitalShenzhen 518033, China
| | - Liping Peng
- Shenzhen Traditional Chinese Medicine HospitalShenzhen 518033, China
| | - Lixin Wang
- Shenzhen Traditional Chinese Medicine HospitalShenzhen 518033, China
| | - Zhouwei Liao
- Shenzhen Traditional Chinese Medicine HospitalShenzhen 518033, China
| | - Liekui Hu
- The Fourth Clinical Medical College of Guangzhou University of Chinese MedicineShenzhen 518033, China
| |
Collapse
|
6
|
Bordoloi D, Monisha J, Roy NK, Padmavathi G, Banik K, Harsha C, Wang H, Kumar AP, Arfuso F, Kunnumakkara AB. An Investigation on the Therapeutic Potential of Butein, A Tretrahydroxychalcone Against Human Oral Squamous Cell Carcinoma. Asian Pac J Cancer Prev 2019; 20:3437-3446. [PMID: 31759370 PMCID: PMC7063020 DOI: 10.31557/apjcp.2019.20.11.3437] [Citation(s) in RCA: 39] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2019] [Indexed: 12/11/2022] Open
Abstract
Background: Oral squamous cell carcinoma (OSCC) is one of the most predominant cancers in India. With advances in the field of oncology, a number of therapies have emerged; however, they are minimally effective. Consequently, there is a need to develop safe and effective regimens for the treatment of OSCC. Butein, a tetrahydroxychalcone has been found to exhibit potent antioxidant, anti-inflammatory, and also anti-tumor effects against several cancer types. However, its effect on OSCC is not studied yet. Methods: The effect of butein on the viability, apoptosis, migration and invasion of OSCC cells was evaluated using MTT, colony formation, PI/FACS, live and dead, scratch wound healing, and matrigel invasion assays. Further Western blot analysis was done to evaluate the expression of different proteins involved in the regulation of cancer hallmarks. Results: This is the first report exemplifying the anti-cancer effect of butein against OSCC. Our results showed that butein exhibited potent anti-proliferative, cytotoxic, anti-migratory, and anti-invasive effects in OSCC cells. It suppressed the expression of NF-κB and NF-κB-regulated gene products such as COX-2, survivin and MMP-9 which are involved in the regulation of different processes like proliferation, survival, invasion, and metastasis of OSCC cells. Conclusion Collectively, these results suggest that butein has immense potential in the management of OSCC. Nonetheless, in vivo validation is critical before moving to clinical trials.
Collapse
Affiliation(s)
- Devivasha Bordoloi
- Cancer Biology Laboratory, & DAILAB, DBT-AIST International Center for Translational & Environmental Research (DAICENTER), Department of Biosciences and Bioengineering, Indian Institute of Technology Guwahati, Assam, India
| | - Javadi Monisha
- Cancer Biology Laboratory, & DAILAB, DBT-AIST International Center for Translational & Environmental Research (DAICENTER), Department of Biosciences and Bioengineering, Indian Institute of Technology Guwahati, Assam, India
| | - Nand Kishor Roy
- Cancer Biology Laboratory, & DAILAB, DBT-AIST International Center for Translational & Environmental Research (DAICENTER), Department of Biosciences and Bioengineering, Indian Institute of Technology Guwahati, Assam, India
| | - Ganesan Padmavathi
- Cancer Biology Laboratory, & DAILAB, DBT-AIST International Center for Translational & Environmental Research (DAICENTER), Department of Biosciences and Bioengineering, Indian Institute of Technology Guwahati, Assam, India
| | - Kishore Banik
- Cancer Biology Laboratory, & DAILAB, DBT-AIST International Center for Translational & Environmental Research (DAICENTER), Department of Biosciences and Bioengineering, Indian Institute of Technology Guwahati, Assam, India
| | - Choudhary Harsha
- Cancer Biology Laboratory, & DAILAB, DBT-AIST International Center for Translational & Environmental Research (DAICENTER), Department of Biosciences and Bioengineering, Indian Institute of Technology Guwahati, Assam, India
| | - Hong Wang
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore.,Singapore Nuclear Research and Safety Initiative, National University of Singapore, Singapore
| | - Alan Prem Kumar
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore
| | - Frank Arfuso
- Stem Cell and Cancer Biology Laboratory, School of Pharmacy and Biomedical Sciences, Curtin Health Innovation Research Institute, Curtin University, Perth, WA 6102, Australia
| | | |
Collapse
|
7
|
Zhang C, Zong X, Han Y. Effect of butein and glucose on oxidative stress and p38 activation marker in non-small cell lung cancer cell. Hum Exp Toxicol 2019; 38:1155-1167. [PMID: 31215238 DOI: 10.1177/0960327119851250] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
INTRODUCTION Tumor microenvironment is known to alter the anticancer drug efficiency. One of the factors that get altered in cancer microenvironment is glucose concentration. Butein, an active principle from plant, known to have anticancer effect against different types of tumor. The objective of the study is to determine the effect of butein on glucose exposed non-small cell lung cancer cells (NSCLCCs). METHODS The current study deals with the effect of butein (6.25-50μM) on NSCLCCs treated with different concentrations (0-40 mM) of glucose. RESULTS AND DISCUSSION Glucose concentration, 0 mM and 40 mM, was found to be lethal at 72 h. Viable cell numbers were statistically increased in 5-mM, 10-mM, and 20-mM glucose-treated cells. Butein at 12.5 µM inhibits (p < 0.05) glucose-induced cell proliferation. Butein inhibits glucose-induced proliferation through DNA damage and oxidative stress. Mitochondrial reactive oxygen species (ROS) level was elevated in 20-mM glucose-treated cells when compared to 5-mM glucose-treated cells, whereas butein treatment further increases glucose-induced mitochondrial ROS. Pharmacological inhibitor of glycolysis, such as 2-deoxy glucose (2-DG), was found to inhibit (p < 0.05) glucose-induced cells proliferation. Furthermore, 2-DG and butein showed synergistic anticancer effect. Butein treatment increases p38 phosphorylation. Inhibition of p38 phosphorylation and antioxidant pretreatment partially revert the glucose-induced cell proliferation. However, inhibition of p38 phosphorylation combined with antioxidant pretreatment completely reverts the anticancer effect of butein. The present study concludes through the evidence that butein could serve as a potential anticancer compound in tumor microenvironment.
Collapse
Affiliation(s)
- C Zhang
- 1 Department of Oncology, Affiliated Hospital of Jining Medical University, Jining, China
| | - X Zong
- 2 Department of Gastroenterology, People's Hospital of Jiaxiang, Jiaxiang, China
| | - Y Han
- 3 Department of Ultrasound, Affiliated Hospital of Jining Medical University, Jining, China
| |
Collapse
|
8
|
Pandey G, Marimuthu M, Kanagavalli P, Ravichandiran V, Balamurugan K, Veerapandian M. Chitosanylated MoO3–Ruthenium(II) Nanocomposite as Biocompatible Probe for Bioimaging and Herbaceutical Detection. ACS Biomater Sci Eng 2019; 5:3606-3617. [DOI: 10.1021/acsbiomaterials.9b00575] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Affiliation(s)
- Gaurav Pandey
- Department of Natural Products, National Institute of Pharmaceutical Education and Research, Kolkata, West Bengal 700 054, India
| | - Mohana Marimuthu
- Department of Biotechnology, Alagappa University, Science Campus, Karaikudi, Tamil Nadu 630 003, India
| | | | - Velayutham Ravichandiran
- Department of Natural Products, National Institute of Pharmaceutical Education and Research, Kolkata, West Bengal 700 054, India
| | - Krishnaswamy Balamurugan
- Department of Biotechnology, Alagappa University, Science Campus, Karaikudi, Tamil Nadu 630 003, India
| | | |
Collapse
|