101
|
Liu H, Chen X, Wang P, Chen M, Deng C, Qian X, Bai J, Li Z, Yu X. PRMT1-mediated PGK1 arginine methylation promotes colorectal cancer glycolysis and tumorigenesis. Cell Death Dis 2024; 15:170. [PMID: 38402202 PMCID: PMC10894231 DOI: 10.1038/s41419-024-06544-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2023] [Revised: 02/05/2024] [Accepted: 02/06/2024] [Indexed: 02/26/2024]
Abstract
Many types of cancer cells, including colorectal cancer cells (CRC), can simultaneously enhance glycolysis and repress the mitochondrial tricarboxylic acid (TCA) cycle, which is called the Warburg effect. However, the detailed mechanisms of abnormal activation of the glycolysis pathway in colorectal cancer are largely unknown. In this study, we reveal that the protein arginine methyltransferase 1 (PRMT1) promotes glycolysis, proliferation, and tumorigenesis in CRC cells. Mechanistically, PRMT1-mediated arginine asymmetric dimethylation modification of phosphoglycerate kinase 1 (PGK1, the first ATP-producing enzyme in glycolysis) at R206 (meR206-PGK1) enhances the phosphorylation level of PGK1 at S203 (pS203-PGK1), which inhibits mitochondrial function and promotes glycolysis. We found that PRMT1 and meR206-PGK1 expression were positively correlated with pS203-PGK1 expression in tissues from colorectal cancer patients. Furthermore, we also confirmed that meR206-PGK1 expression is positively correlated with the poor survival of patients with colorectal cancer. Our findings show that PRMT1 and meR206-PGK1 may become promising predictive biomarkers for the prognosis of patients with CRC and that arginine methyltransferase inhibitors have great potential in colorectal cancer treatment.
Collapse
Affiliation(s)
- Hao Liu
- School of Medicine, Nankai University, Tianjin, China
- Cancer Institute, Xuzhou Medical University, Xuzhou, Jiangsu, China
| | - Xintian Chen
- Department of Gastroenterology, the Affiliated Hospital of Guangdong Medical University, Zhanjiang, Guangdong, China
| | - Pengfei Wang
- Department of Gastroenterology, the First People's Hospital of Shuyang County, Suqian, Jiangsu, China
| | - Miaolei Chen
- Cancer Institute, Xuzhou Medical University, Xuzhou, Jiangsu, China
| | - Chuyin Deng
- Cancer Institute, Xuzhou Medical University, Xuzhou, Jiangsu, China
| | - Xingyou Qian
- Cancer Institute, Xuzhou Medical University, Xuzhou, Jiangsu, China
| | - Jin Bai
- Cancer Institute, Xuzhou Medical University, Xuzhou, Jiangsu, China.
- Center of Clinical Oncology, the Affiliated Hospital of Xuzhou Medical University, Xuzhou, Jiangsu, China.
- Jiangsu Center for the Collaboration and Innovation of Cancer Biotherapy, Cancer Institute, Xuzhou Medical University, Xuzhou, Jiangsu, China.
| | - Zhongwei Li
- Cancer Institute, Xuzhou Medical University, Xuzhou, Jiangsu, China.
- Laboratory of Tumor Epigenetics, Department of Pathophysiology, School of Basic Medical Sciences, Wannan Medical College, Wuhu, Anhui, China.
| | - Xiangyang Yu
- School of Medicine, Nankai University, Tianjin, China.
- Department of Gastrointestinal Surgery, the Hospital of Integrated Chinese and Western Medicine, Tianjin, China.
| |
Collapse
|
102
|
Jardim-Perassi BV, Irrera P, Oluwatola OE, Abrahams D, Estrella VC, Ordway B, Byrne SR, Ojeda AA, Whelan CJ, Kim J, Beatty MS, Damgaci-Erturk S, Longo DL, Gaspar KJ, Siegers GM, Centeno BA, Lau JYC, Pilon-Thomas SA, Ibrahim-Hashim A, Gillies RJ. L-DOS47 Elevates Pancreatic Cancer Tumor pH and Enhances Response to Immunotherapy. Biomedicines 2024; 12:461. [PMID: 38398062 PMCID: PMC10886509 DOI: 10.3390/biomedicines12020461] [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: 12/14/2023] [Revised: 01/17/2024] [Accepted: 01/25/2024] [Indexed: 02/25/2024] Open
Abstract
Acidosis is an important immunosuppressive mechanism that leads to tumor growth. Therefore, we investigated the neutralization of tumor acidity to improve immunotherapy response. L-DOS47, a new targeted urease immunoconjugate designed to neutralize tumor acidity, has been well tolerated in phase I/IIa trials. L-DOS47 binds to CEACAM6, a cell-surface protein that is highly expressed in gastrointestinal cancers, allowing urease to cleave endogenous urea into two NH4+ and one CO2, thereby raising local pH. To test the synergetic effect of neutralizing tumor acidity with immunotherapy, we developed a pancreatic orthotopic murine tumor model (KPC961) expressing human CEACAM6. Using chemical exchange saturation transfer-magnetic resonance imaging (CEST-MRI) to measure the tumor extracellular pH (pHe), we confirmed that L-DOS47 raises the tumor pHe from 4 h to 96 h post injection in acidic tumors (average increase of 0.13 units). Additional studies showed that combining L-DOS47 with anti-PD1 significantly increases the efficacy of the anti-PD1 monotherapy, reducing tumor growth for up to 4 weeks.
Collapse
Affiliation(s)
- Bruna Victorasso Jardim-Perassi
- Department of Metabolism and Physiology, H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL 33612, USA (P.I.); (B.O.); (S.D.-E.)
| | - Pietro Irrera
- Department of Metabolism and Physiology, H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL 33612, USA (P.I.); (B.O.); (S.D.-E.)
| | - Oluwaseyi E. Oluwatola
- Department of Immunology, H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL 33612, USA; (O.E.O.)
- Department of Molecular Medicine, University of South Florida, Tampa, FL 33620, USA
| | | | - Veronica C. Estrella
- Department of Metabolism and Physiology, H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL 33612, USA (P.I.); (B.O.); (S.D.-E.)
| | - Bryce Ordway
- Department of Metabolism and Physiology, H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL 33612, USA (P.I.); (B.O.); (S.D.-E.)
- Massachusetts General Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Samantha R. Byrne
- Department of Immunology, H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL 33612, USA; (O.E.O.)
| | - Andrew A. Ojeda
- Department of Immunology, H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL 33612, USA; (O.E.O.)
| | - Christopher J. Whelan
- Department of Metabolism and Physiology, H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL 33612, USA (P.I.); (B.O.); (S.D.-E.)
- Department of Biological Sciences, University of Illinois, Chicago, IL 60607, USA
| | - Jongphil Kim
- Department of Biostatistics and Bioinformatics, H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL 33612, USA
| | - Matthew S. Beatty
- Department of Immunology, H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL 33612, USA; (O.E.O.)
| | - Sultan Damgaci-Erturk
- Department of Metabolism and Physiology, H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL 33612, USA (P.I.); (B.O.); (S.D.-E.)
| | - Dario Livio Longo
- Institute of Biostructures and Bioimaging (IBB), National Research Council of Italy (CNR), 10126 Turin, Italy
| | - Kim J. Gaspar
- Helix BioPharma Corp., Bay Adelaide Centre-North Tower, 40 Temperance Street, Suite 2700, Toronto, ON M5H 0B4, Canada
| | - Gabrielle M. Siegers
- Helix BioPharma Corp., Bay Adelaide Centre-North Tower, 40 Temperance Street, Suite 2700, Toronto, ON M5H 0B4, Canada
| | - Barbara A. Centeno
- Department of Pathology, H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL 33612, USA
| | - Justin Y. C. Lau
- Small Animal Imaging Laboratory (SAIL), H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL 33612, USA;
| | - Shari A. Pilon-Thomas
- Department of Immunology, H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL 33612, USA; (O.E.O.)
| | - Arig Ibrahim-Hashim
- Department of Metabolism and Physiology, H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL 33612, USA (P.I.); (B.O.); (S.D.-E.)
| | - Robert J. Gillies
- Department of Metabolism and Physiology, H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL 33612, USA (P.I.); (B.O.); (S.D.-E.)
| |
Collapse
|
103
|
Stocker M, Blancke Soares A, Liebsch G, Meier RJ, Canis M, Gires O, Haubner F. Quantification of oxygen consumption in head and neck cancer using fluorescent sensor foil technology. Front Oncol 2024; 14:1002798. [PMID: 38390268 PMCID: PMC10882065 DOI: 10.3389/fonc.2024.1002798] [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: 07/25/2022] [Accepted: 01/22/2024] [Indexed: 02/24/2024] Open
Abstract
Introduction Head and neck squamous cell carcinoma (HNSCC) patients suffer from frequent local recurrences that negatively impact on prognosis. Hence, distinguishing tumor and normal tissue is of clinical importance as it may improve the detection of residual tumor tissue in surgical resection margins and during imaging-based surgery planning. Differences in O2 consumption (OC) can be used to this aim, as they provide options for improved surgical, image-guided approaches. Methods In the present study, the potential of a fluorescent sensor foil-based technology to quantify OC in HNSCC was evaluated in an in vitro 3D model and in situ in patients. Results In vitro measurements of OC using hypopharyngeal and esophageal cell lines allowed a specific detection of tumor cell spheroids embedded together with cancer-associated fibroblasts in type I collagen extracellular matrix down to a diameter of 440 µm. Pre-surgery in situ measurements were conducted with a handheld recording device and sensor foils with an oxygen permeable membrane and immobilized O2-reactive fluorescent dyes. Lateral tongue carcinoma and carcinoma of the floor of the mouth were chosen for analysis owing to their facilitated accessibility. OC was evaluated over a time span of 60 seconds and was significantly higher in tumor tissue compared to healthy mucosa in the vicinity of the tumor. Discussion Hence, OC quantification using fluorescent sensor foil-based technology is a relevant parameter for the differentiation of tumor tissue of the head and neck region and may support surgery planning.
Collapse
Affiliation(s)
- Magdalena Stocker
- Department of Otorhinolaryngology, Head and Neck Surgery, University Hospital, Ludwig Maximilians University (LMU) Munich, Munich, Germany
| | - Alexandra Blancke Soares
- Department of Otorhinolaryngology, Head and Neck Surgery, University Hospital, Ludwig Maximilians University (LMU) Munich, Munich, Germany
| | - Gregor Liebsch
- PreSens Precision Sensing GmbH, Imaging Solutions, Regensburg, Germany
| | - Robert J Meier
- PreSens Precision Sensing GmbH, Imaging Solutions, Regensburg, Germany
| | - Martin Canis
- Department of Otorhinolaryngology, Head and Neck Surgery, University Hospital, Ludwig Maximilians University (LMU) Munich, Munich, Germany
| | - Olivier Gires
- Department of Otorhinolaryngology, Head and Neck Surgery, University Hospital, Ludwig Maximilians University (LMU) Munich, Munich, Germany
| | - Frank Haubner
- Department of Otorhinolaryngology, Head and Neck Surgery, University Hospital, Ludwig Maximilians University (LMU) Munich, Munich, Germany
| |
Collapse
|
104
|
Wang W, Zhou Y, Li W, Quan C, Li Y. Claudins and hepatocellular carcinoma. Biomed Pharmacother 2024; 171:116109. [PMID: 38185042 DOI: 10.1016/j.biopha.2023.116109] [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: 10/04/2023] [Revised: 12/23/2023] [Accepted: 12/28/2023] [Indexed: 01/09/2024] Open
Abstract
Hepatocellular carcinoma (HCC) has a high incidence and dismal prognosis, making it a significant global health burden. To change this, the development of new therapeutic strategies is imminent. The claudin (CLDN) family, as key components of tight junctions (TJs), plays an important role in the initiation and development of cancer. Dysregulated expression of CLDNs leads to loss of intercellular adhesion and aberrant cell signaling, which are closely related to cancer cell invasion, migration, and epithelial-mesenchymal transition (EMT). CLDN1, CLDN3, CLDN4, CLDN5, CLDN6, CLDN7, CLDN9, CLDN10, CLDN11, CLDN14, and CLDN17 are aberrantly expressed in HCC, which drives the progression of the disease. Consequently, they have tremendous potential as prognostic indicators and therapeutic targets. This article summarizes the aberrant expression, molecular mechanisms, and clinical application studies of different subtypes of CLDNs in HCC, with a particular emphasis on CLDN1.
Collapse
Affiliation(s)
- Wentao Wang
- The Key Laboratory of Pathobiology, Ministry of Education, College of Basic Medical Sciences, Jilin University, 126 Xinmin Avenue, Changchun, Jilin 130021, China; The Second Norman Bethune College of Clinical Medicine, Jilin University, Changchun 130021, China
| | - Yi Zhou
- The Key Laboratory of Pathobiology, Ministry of Education, College of Basic Medical Sciences, Jilin University, 126 Xinmin Avenue, Changchun, Jilin 130021, China; The First Norman Bethune College of Clinical Medicine, Jilin University, Changchun 130021, China
| | - Wei Li
- The Key Laboratory of Pathobiology, Ministry of Education, College of Basic Medical Sciences, Jilin University, 126 Xinmin Avenue, Changchun, Jilin 130021, China
| | - Chengshi Quan
- The Key Laboratory of Pathobiology, Ministry of Education, College of Basic Medical Sciences, Jilin University, 126 Xinmin Avenue, Changchun, Jilin 130021, China
| | - Yanru Li
- The Key Laboratory of Pathobiology, Ministry of Education, College of Basic Medical Sciences, Jilin University, 126 Xinmin Avenue, Changchun, Jilin 130021, China.
| |
Collapse
|
105
|
Zhang S, Wang Y, Luo D, Cheng Z, Zeng Q, Wang G, Chen M, Zhang S, Luo P. Pirfenidone inhibits TGF-β1-induced metabolic reprogramming during epithelial-mesenchymal transition in non-small cell lung cancer. J Cell Mol Med 2024; 28:e18059. [PMID: 38140828 PMCID: PMC10844763 DOI: 10.1111/jcmm.18059] [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: 04/13/2023] [Revised: 11/13/2023] [Accepted: 11/16/2023] [Indexed: 12/24/2023] Open
Abstract
Metastasis is an important contributor to increased mortality rates in non-small cell lung cancer (NSCLC). The TGF-β signalling pathway plays a crucial role in facilitating tumour metastasis through epithelial-mesenchymal transition (EMT). Glycolysis, a key metabolic process, is strongly correlated with NSCLC metastasis. Pirfenidone (PFD) has been shown to safely and effectively inhibit TGF-β1 in patients with lung diseases. Furthermore, TGF-β1 and glycolysis demonstrate an interdependent relationship within the tumour microenvironment. Our previous study demonstrated that PFD effectively inhibited glycolysis in NSCLC cells, prompting further investigation into its potential antitumour effects in this context. Therefore, the present study aims to investigate the potential antitumour effect of PFD in NSCLC and explore the relationship among TGF-β1, glycolysis and EMT through further experimentation. The antitumour effects of PFD were evaluated using five different NSCLC cell lines and a xenograft tumour model. Notably, PFD demonstrated a significant antitumour effect specifically in highly glycolytic H1299 cells. To elucidate the underlying mechanism, we compared the efficacy of PFD after pretreatment with either TGF-β1 or a TGF-β receptor inhibitor (LY2109761). The energy metabolomics analysis of tumour tissue demonstrated that PFD, a chemosensitizing agent, reduced lactate and ATP production, thereby inhibiting glycolysis and exerting synergistic antineoplastic effects. Additionally, PFD combined with cisplatin targeted TGF-β1 to inhibit glycolysis during EMT and enhanced the chemosensitization of A549 and H1299 cells. The magnitude of the anticancer effect exhibited by PFD was intricately linked to its metabolic properties.
Collapse
Affiliation(s)
- Shuling Zhang
- Guizhou Medical UniversityGuiyangChina
- Key Laboratory of Environmental Pollution Monitoring and Disease Control, Ministry of EducationSchool of Public HealthGuiyangChina
- State Key Laboratory of Functions and Applications of Medicinal PlantsGuizhou Medical UniversityGuiyangChina
- Guizhou Provincial Engineering Research Center of Food Nutrition and HealthGuizhou Medical UniversityGuiyangChina
| | - Yuanmei Wang
- Guizhou Medical UniversityGuiyangChina
- Key Laboratory of Environmental Pollution Monitoring and Disease Control, Ministry of EducationSchool of Public HealthGuiyangChina
| | | | | | - Qibing Zeng
- Guizhou Medical UniversityGuiyangChina
- Key Laboratory of Environmental Pollution Monitoring and Disease Control, Ministry of EducationSchool of Public HealthGuiyangChina
- State Key Laboratory of Functions and Applications of Medicinal PlantsGuizhou Medical UniversityGuiyangChina
- Guizhou Provincial Engineering Research Center of Food Nutrition and HealthGuizhou Medical UniversityGuiyangChina
| | - Guoze Wang
- Guizhou Medical UniversityGuiyangChina
- Key Laboratory of Environmental Pollution Monitoring and Disease Control, Ministry of EducationSchool of Public HealthGuiyangChina
- State Key Laboratory of Functions and Applications of Medicinal PlantsGuizhou Medical UniversityGuiyangChina
- Guizhou Provincial Engineering Research Center of Food Nutrition and HealthGuizhou Medical UniversityGuiyangChina
| | | | - Shuai Zhang
- Guizhou Medical UniversityGuiyangChina
- Key Laboratory of Environmental Pollution Monitoring and Disease Control, Ministry of EducationSchool of Public HealthGuiyangChina
| | - Peng Luo
- Guizhou Medical UniversityGuiyangChina
- Key Laboratory of Environmental Pollution Monitoring and Disease Control, Ministry of EducationSchool of Public HealthGuiyangChina
- State Key Laboratory of Functions and Applications of Medicinal PlantsGuizhou Medical UniversityGuiyangChina
- Guizhou Provincial Engineering Research Center of Food Nutrition and HealthGuizhou Medical UniversityGuiyangChina
| |
Collapse
|
106
|
Mushtaq A, Li L, Grøndahl L, A A. Targeted Nanoparticles Based on Alendronate Polyethylene Glycol Conjugated Chitosan for the Delivery of siRNA and Curcumin for Bone Metastasized Breast Cancer Applications. Macromol Biosci 2024; 24:e2300268. [PMID: 37794635 DOI: 10.1002/mabi.202300268] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2023] [Revised: 09/22/2023] [Indexed: 10/06/2023]
Abstract
Bone metastasized breast cancer reduces the quality of life and median survival. Targeted delivery of small interfering RNA (siRNA) and chemotherapeutic drugs using nanoparticles (NPs) is a promising strategy to overcome current limitations in treating these metastatic breast cancers. This research develops alendronate conjugated polyethylene glycol functionalized chitosan (ALD-PEG-CHI) NP for the delivery of cell death siRNA (CD-siRNA) and curcumin (CUR) and explores its targeting ability and in vitro cell cytotoxicity. Polyethylene glycol functionalized CHI (mPEG-CHI) NPs serve as control. The size of CD-siRNA loaded NPs is below 100 nm while CUR loaded NPs is below 200 nm, with near neutral zeta potential for all NPs. The CUR encapsulation efficiency (EE) is 70% and 88% for targeted and control NPs, respectively, while complete encapsulation of CD-siRNA is achieved in both NP systems. The bone targeting ability of CY5-dsDNA loaded ALD-PEG-CHI NPs using hydroxyapatite discs is fivefold compared to control indicating ALD presentation at the targeting NP surface. Delivery of CD-siRNA loaded NPs and CUR loaded NPs show synergistic and additive growth inhibition effects against MCF-7 cells by mPEG-CHI and ALD-PEG-CHI NPs, respectively. Overall, these in vitro results illustrate the potential of the targeted NPs as an effective therapeutic system toward bone metastasized breast cancer.
Collapse
Affiliation(s)
- Asim Mushtaq
- School of Chemistry and Molecular Biosciences, The University of Queensland, Cooper Road, Brisbane, Queensland, 4072, Australia
| | - Li Li
- Australian Institute for Bioengineering and Nanotechnology, The University of Queensland, Corner of College and Cooper Road, Brisbane, Queensland, 4072, Australia
| | - Lisbeth Grøndahl
- School of Chemistry and Molecular Biosciences, The University of Queensland, Cooper Road, Brisbane, Queensland, 4072, Australia
- Australian Institute for Bioengineering and Nanotechnology, The University of Queensland, Corner of College and Cooper Road, Brisbane, Queensland, 4072, Australia
| | - Anitha A
- School of Chemistry and Molecular Biosciences, The University of Queensland, Cooper Road, Brisbane, Queensland, 4072, Australia
| |
Collapse
|
107
|
Chen PC, Ning Y, Li H, Su JG, Shen JB, Feng QC, Jiang SH, Shi PD, Guo RS. Targeting ONECUT3 blocks glycolytic metabolism and potentiates anti-PD-1 therapy in pancreatic cancer. Cell Oncol (Dordr) 2024; 47:81-96. [PMID: 37606818 DOI: 10.1007/s13402-023-00852-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 07/31/2023] [Indexed: 08/23/2023] Open
Abstract
BACKGROUND Reprogramming glucose metabolism, also known as the Warburg effect (aerobic glycolysis), is a hallmark of cancers. Increased tumor glycolysis not only favors rapid cancer cell proliferation but reprograms the immune microenvironment to enable tumor progression. The transcriptional factor ONECUT3 plays key roles in the development of the liver and pancreas, however, limited is known about its oncogenic roles, particularly metabolic reprogramming. METHODS Immunohistochemistry and Western blotting are applied to determine the expression pattern of ONECUT3 and its clinical relevance in pancreatic ductal adenocarcinoma (PDAC). Knockdown and overexpression strategies are employed to determine the in vitro and in vivo functions of ONECUT3. Chromatin immunoprecipitation, luciferase reporter assay, and gene set enrichment analysis are used to decipher the molecular mechanisms. RESULTS The glycolytic metabolism is inversely associated with T-cell infiltration in PDAC. ONECUT3 is identified as a key regulator for PDAC glycolysis and CD8+ T-cell infiltration. Genetic silencing of ONECUT3 inhibits cell proliferation, promotes cell apoptosis, and reduces glycolytic metabolism as evidenced by glucose uptake, lactate production, and extracellular acidification rate. Opposite effects of ONECUT3 are observed in overexpression studies. ONECUT3 enhances aerobic glycolysis via transcriptional regulation of PDK1. Targeting ONECUT3 effectively suppresses tumor growth, increases CD8+ T-cell infiltration, and potentiates anti-PD-1 therapy in PDAC. Pharmacological inhibition of PDK1 also shows a synergistic effect with anti-PD-1 therapy. In clinical setting, ONECUT3 is closely associated with PDK1 expression and T-cell infiltration in PDAC and acts as an independent prognostic factor. CONCLUSIONS Our study reveals a previous unprecedented regulatory role of ONECUT3 in PDAC glycolysis and provides in vivo evidence that increased glycolysis is linked to an immunosuppressive microenvironment. Moreover, targeting ONECUT3-PDK1 axis may serve as a promising therapeutic approach for the treatment of PDAC.
Collapse
Affiliation(s)
- Peng-Cheng Chen
- Department of General Surgery, Jiading District Central Hospital Affiliated Shanghai University of Medicine & Health Sciences, Shanghai, 201800, P.R. China
| | - Yong Ning
- Department of General Surgery, Jiading District Central Hospital Affiliated Shanghai University of Medicine & Health Sciences, Shanghai, 201800, P.R. China
| | - Hui Li
- State Key Laboratory of Oncogenes and Related Genes, Ren Ji Hospital, School of Medicine, Shanghai Cancer Institute, Shanghai Jiao Tong University, Shanghai, 200240, P.R. China
| | - Jin-Gen Su
- Department of General Surgery, Jiading District Central Hospital Affiliated Shanghai University of Medicine & Health Sciences, Shanghai, 201800, P.R. China
| | - Jiang-Bo Shen
- Department of General Surgery, Jiading District Central Hospital Affiliated Shanghai University of Medicine & Health Sciences, Shanghai, 201800, P.R. China
| | - Qing-Chun Feng
- Department of General Surgery, Jiading District Central Hospital Affiliated Shanghai University of Medicine & Health Sciences, Shanghai, 201800, P.R. China
| | - Shu-Heng Jiang
- State Key Laboratory of Oncogenes and Related Genes, Ren Ji Hospital, School of Medicine, Shanghai Cancer Institute, Shanghai Jiao Tong University, Shanghai, 200240, P.R. China
| | - Pei-Dong Shi
- Department of General Surgery, Jiading District Central Hospital Affiliated Shanghai University of Medicine & Health Sciences, Shanghai, 201800, P.R. China.
| | - Run-Sheng Guo
- Department of General Surgery, Jiading District Central Hospital Affiliated Shanghai University of Medicine & Health Sciences, Shanghai, 201800, P.R. China.
| |
Collapse
|
108
|
Zhang YH, Liu XS, Gao Y, Yuan LL, Huang ZM, Zhang Y, Liu ZY, Yang Y, Liu XY, Ke CB, Pei ZJ. SFXN1 as a potential diagnostic and prognostic biomarker of LUAD is associated with 18F-FDG metabolic parameters. Lung Cancer 2024; 188:107449. [PMID: 38184958 DOI: 10.1016/j.lungcan.2023.107449] [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: 10/28/2023] [Revised: 12/16/2023] [Accepted: 12/21/2023] [Indexed: 01/09/2024]
Abstract
BACKGROUND Sideroflexin 1 (SFXN1) has been discovered as a novel tumor marker for lung adenocarcinoma, but data on its importance in the development of lung adenocarcinoma is still limited. This study evaluated the correlation between SFXN1 and parameters related to 18F-flurodeoxyglucose (18F-FDG) positron emission tomography/computed tomography (PET/CT), and further explored the role of SFXN1 in the value-added and glycolytic processes of LUAD. METHOD The expression and prognostic value of SFXN1 mRNA in LUAD were analyzed using The Cancer Genome Atlas (TCGA) and Gene Expression Omnibus (GEO) data base. Retrospective analysis of 18F-FDG PET imaging and metabolic parameters in 42 patients to explore the relationship between the expression of SFXN1 and glucose metabolism levels in lung adenocarcinoma and its clinical significance. H1975 cells were selected as the in vitro research object, and the biological effects of SFXN1 on LUAD were further elucidated through Edu proliferation assay, CCK8 activity assay, wound healing experiment, and cell flow cytometry. RESULT SFXN1 is highly expressed in various tumors, including LUAD, and its high expression can serve as an independent predictor of overall survival in lung adenocarcinoma. In addition, the expression of SFXN1 in LUAD was significantly correlated with 18F-FDG PET/CT parameters: maximum and average standardized uptake values (SUVmax and SUVmean), as well as total lesion glycolysis (TLG) (rho = 0.574, 0.589, and 0.338, p < 0.05), which can predict the expression of SFXN1 with an accuracy of 0.934. In vitro functional experiments have shown that knocking down SFXN1 inhibits the proliferation and migration of LUAD cells, promotes cell apoptosis, and may inhibit tumor activity by regulating the expression of glycolytic related genes SLC2A1, HK2, GPI, ALDOA, GAPDH, ENO1, PKM, and LDHA. CONCLUSION The overexpression of SFXN1 is closely related to FDG uptake, and SFXN1, as a promising prognostic biomarker, may mediate the development of LUAD through the glycolytic pathway.
Collapse
Affiliation(s)
- Yao-Hua Zhang
- Department of Nuclear Medicine and Institute of Anesthesiology and Pain, Hubei Provincial Clinical Research Center for Precision Diagnosis and Treatment of Liver Cancer, Taihe Hospital, Hubei University of Medicine, Shiyan 442000, China
| | - Xu-Sheng Liu
- Department of Nuclear Medicine and Institute of Anesthesiology and Pain, Hubei Provincial Clinical Research Center for Precision Diagnosis and Treatment of Liver Cancer, Taihe Hospital, Hubei University of Medicine, Shiyan 442000, China; Hubei Provincial Clinical Research Center for Umbilical Cord Blood Hematopoietic Stem Cells, Taihe Hospital, Hubei University of Medicine, Shiyan 442000, China; Hubei Key Laboratory of Embryonic Stem Cell Research, Shiyan 442000, Hubei, China
| | - Yan Gao
- Department of Nuclear Medicine and Institute of Anesthesiology and Pain, Hubei Provincial Clinical Research Center for Precision Diagnosis and Treatment of Liver Cancer, Taihe Hospital, Hubei University of Medicine, Shiyan 442000, China
| | - Ling-Ling Yuan
- Department of Pathology, Taihe Hospital, Hubei University of Medicine, Shiyan 442000, China
| | - Zhong-Min Huang
- Department of Medical Ultrasound, Taihe Hospital, Hubei University of Medicine, Shiyan 442000, China
| | - Yu Zhang
- Department of Nuclear Medicine and Institute of Anesthesiology and Pain, Hubei Provincial Clinical Research Center for Precision Diagnosis and Treatment of Liver Cancer, Taihe Hospital, Hubei University of Medicine, Shiyan 442000, China
| | - Zi-Yue Liu
- Department of Nuclear Medicine and Institute of Anesthesiology and Pain, Hubei Provincial Clinical Research Center for Precision Diagnosis and Treatment of Liver Cancer, Taihe Hospital, Hubei University of Medicine, Shiyan 442000, China
| | - Yi Yang
- Department of Nuclear Medicine and Institute of Anesthesiology and Pain, Hubei Provincial Clinical Research Center for Precision Diagnosis and Treatment of Liver Cancer, Taihe Hospital, Hubei University of Medicine, Shiyan 442000, China
| | - Xiao-Yu Liu
- Department of Nuclear Medicine and Institute of Anesthesiology and Pain, Hubei Provincial Clinical Research Center for Precision Diagnosis and Treatment of Liver Cancer, Taihe Hospital, Hubei University of Medicine, Shiyan 442000, China
| | - Chang-Bin Ke
- Department of Nuclear Medicine and Institute of Anesthesiology and Pain, Hubei Provincial Clinical Research Center for Precision Diagnosis and Treatment of Liver Cancer, Taihe Hospital, Hubei University of Medicine, Shiyan 442000, China.
| | - Zhi-Jun Pei
- Department of Nuclear Medicine and Institute of Anesthesiology and Pain, Hubei Provincial Clinical Research Center for Precision Diagnosis and Treatment of Liver Cancer, Taihe Hospital, Hubei University of Medicine, Shiyan 442000, China; Hubei Provincial Clinical Research Center for Umbilical Cord Blood Hematopoietic Stem Cells, Taihe Hospital, Hubei University of Medicine, Shiyan 442000, China; Hubei Key Laboratory of Embryonic Stem Cell Research, Shiyan 442000, Hubei, China.
| |
Collapse
|
109
|
Wang S, Hou Y. New Types of Magnetic Nanoparticles for Stimuli-Responsive Theranostic Nanoplatforms. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2024; 11:e2305459. [PMID: 37988692 PMCID: PMC10885654 DOI: 10.1002/advs.202305459] [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: 08/07/2023] [Revised: 10/21/2023] [Indexed: 11/23/2023]
Abstract
Magnetic nanomaterials have played a crucial role in promoting the application of nanotechnology in the biomedical field. Although conventional magnetic nanomaterials such as iron oxide nanoparticles (NPs) are used as biosensors, drug delivery vehicles, diagnostic and treatment agents for several diseases, the persistent pursuit of high-performance technologies has prompted researchers to continuously develop new types of magnetic nanomaterials such as iron carbide NPs. Considering their potential application in biomedicine, magnetic NPs responsive to exogenous or endogenous stimuli are developed, thereby enhancing their applicability in more complex versatile scenarios. In this review, the synthesis and surface modification of magnetic NPs are focused, particularly iron carbide NPs. Subsequently, exogenous and endogenous stimuli-responsive magnetic NP-based theranostic platforms are introduced, particularly focusing on nanozyme-based technologies and magnetic NP-mediated immunotherapy, which are emerging stimuli-responsive treatments. Finally, the challenges and perspectives of magnetic NPs to accelerate future research in this field are discussed.
Collapse
Affiliation(s)
- Shuren Wang
- Beijing Key Laboratory for Magnetoelectric Materials and Devices, School of Materials Science and Engineering, Peking University, Beijing, 100871, China
| | - Yanglong Hou
- Beijing Key Laboratory for Magnetoelectric Materials and Devices, School of Materials Science and Engineering, Peking University, Beijing, 100871, China
- School of Materials, Sun Yat-Sen University, Shenzhen, 518107, China
| |
Collapse
|
110
|
Ye S, Zhong K, Huang Y, Zhang G, Sun C, Jiang J. Artificial Intelligence-based Amide-II Infrared Spectroscopy Simulation for Monitoring Protein Hydrogen Bonding Dynamics. J Am Chem Soc 2024; 146:2663-2672. [PMID: 38240637 DOI: 10.1021/jacs.3c12258] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/01/2024]
Abstract
The structurally sensitive amide II infrared (IR) bands of proteins provide valuable information about the hydrogen bonding of protein secondary structures, which is crucial for understanding protein dynamics and associated functions. However, deciphering protein structures from experimental amide II spectra relies on time-consuming quantum chemical calculations on tens of thousands of representative configurations in solvent water. Currently, the accurate simulation of amide II spectra for whole proteins remains a challenge. Here, we present a machine learning (ML)-based protocol designed to efficiently simulate the amide II IR spectra of various proteins with an accuracy comparable to experimental results. This protocol stands out as a cost-effective and efficient alternative for studying protein dynamics, including the identification of secondary structures and monitoring the dynamics of protein hydrogen bonding under different pH conditions and during protein folding process. Our method provides a valuable tool in the field of protein research, focusing on the study of dynamic properties of proteins, especially those related to hydrogen bonding, using amide II IR spectroscopy.
Collapse
Affiliation(s)
- Sheng Ye
- School of Artificial Intelligence, Anhui University, Hefei, Anhui 230601, People's Republic of China
| | - Kai Zhong
- Zernike Institute for Advanced Materials, University of Groningen, Nijenborgh 4, Groningen 9747AG, Netherlands
| | - Yan Huang
- Key Laboratory of Precision and Intelligent Chemistry, School of Chemistry and Materials Science, University of Science and Technology of China, Hefei, Anhui 230026, China
| | - Guozhen Zhang
- Hefei National Research Center of Physical Sciences at the Microscale, School of Chemistry and Materials Science, University of Science and Technology of China, Hefei, Anhui 230026, China
- Hefei National Laboratory, University of Science and Technology of China, Hefei 230088, China
| | - Changyin Sun
- School of Artificial Intelligence, Anhui University, Hefei, Anhui 230601, People's Republic of China
| | - Jun Jiang
- Key Laboratory of Precision and Intelligent Chemistry, School of Chemistry and Materials Science, University of Science and Technology of China, Hefei, Anhui 230026, China
- Hefei National Laboratory, University of Science and Technology of China, Hefei 230088, China
| |
Collapse
|
111
|
Javani Jouni F, Rastegar-Pouyani N, Najjar N, Nasirpour M, Payez A, Kashi G, Zafari J. Evaluation of the effects of photodynamic therapy consisted of the blue laser and zinc oxide QDs on MDA-MB-231 cancer cells by inhibiting cancer markers and inducing apoptosis. Lasers Med Sci 2024; 39:28. [PMID: 38216721 DOI: 10.1007/s10103-024-03977-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2023] [Accepted: 01/02/2024] [Indexed: 01/14/2024]
Abstract
The increasing number of cancer patients has cast attention on developing new anti-cancer modalities. Photodynamic therapy is a safe anti-cancer approach, which encompasses (1) local administration of a photosensitizer and (2) light irradiation. Zinc oxide (ZnO) quantum dots (QDs) are photosensitizers that can be utilized for this purpose. In the present study, to better appreciate the likely more efficient cytotoxic effect of the combination of ZnO QDs and the visible 470-nm blue light in comparison to the QDs alone, several assays were to be conducted upon breast cancer MDA-MB 231 cells. MTT assay showed that in certain groups the combination displayed higher cytotoxic effects compared to those following QD treatment alone. LDH leakage and lipid peroxidation rates by the combination were significantly higher than treatment with either the blue laser or QDs. Although the combination managed to meaningfully reduce the number of colonies and CAT activity compared to QD treatment, there were no palpable differences between them. Lastly, the combination was able to increase the apoptotic genes, including BAX, TP53, caspase 3, and caspase 9 compared to QD, while, in the case of Bcl-2, an anti-apoptotic gene, none of the groups managed to make any tangible differences on its expression levels. Our findings propose that there may be synergistic effects between the blue laser and QD that can possibly be adopted in anti-cancer therapy in the future. However, further investigations regarding this matter are of the essence.
Collapse
Affiliation(s)
- Fatemeh Javani Jouni
- Department of Biochemistry and Biophysics, Faculty of Advanced Sciences and Technology, Tehran Medical Sciences, Islamic Azad University, Tehran, Iran
| | - Nima Rastegar-Pouyani
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Tehran University of Medical Sciences, Tehran, Iran
| | - Nabaa Najjar
- Laser Application in Medical Sciences Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Mohammad Nasirpour
- Department of Medical Genetics, Institute of Medical Biotechnology, National Institute of Genetic Engineering and Biotechnology (NIGEB), Tehran, Iran
| | - Atefeh Payez
- Department of Biology Education, Farhangian University, P.O. Box, Tehran, 14665-889, Iran
| | - Giti Kashi
- Department of Environmental Health Engineering, Faculty of Health and Medical Engineering, Tehran Medical Sciences, Islamic Azad University, Tehran, Iran
- Water Purification Research Center, Tehran Medical Sciences, Islamic Azad University, Tehran, Iran
| | - Jaber Zafari
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Tehran Medical Sciences, Islamic Azad University, Tehran, Iran.
| |
Collapse
|
112
|
Zhu L, Wu W. Dual/Multi-Modal Image-Guided Diagnosis and Therapy Based on Luminogens with Aggregation-Induced Emission. Molecules 2024; 29:371. [PMID: 38257284 PMCID: PMC10819122 DOI: 10.3390/molecules29020371] [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: 12/11/2023] [Revised: 01/03/2024] [Accepted: 01/08/2024] [Indexed: 01/24/2024] Open
Abstract
The combination of multiple imaging methods has made an indelible contribution to the diagnosis, surgical navigation, treatment, and prognostic evaluation of various diseases. Due to the unique advantages of luminogens with aggregation-induced emission (AIE), their progress has been significant in the field of organic fluorescent contrast agents. Herein, this manuscript summarizes the recent advancements in AIE molecules as contrast agents for optical image-based dual/multi-modal imaging. We particularly focus on the exceptional properties of each material and the corresponding application in the diagnosis and treatment of diseases.
Collapse
Affiliation(s)
| | - Wenbo Wu
- Department of Chemistry, Institute of Molecular Aggregation Science, Tianjin University, Tianjin 300072, China;
| |
Collapse
|
113
|
Park W, Han JH, Wei S, Yang ES, Cheon SY, Bae SJ, Ryu D, Chung HS, Ha KT. Natural Product-Based Glycolysis Inhibitors as a Therapeutic Strategy for Epidermal Growth Factor Receptor-Tyrosine Kinase Inhibitor-Resistant Non-Small Cell Lung Cancer. Int J Mol Sci 2024; 25:807. [PMID: 38255882 PMCID: PMC10815680 DOI: 10.3390/ijms25020807] [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: 12/05/2023] [Revised: 01/02/2024] [Accepted: 01/05/2024] [Indexed: 01/24/2024] Open
Abstract
Non-small cell lung cancer (NSCLC) is a leading cause of cancer-related deaths worldwide. Targeted therapy against the epidermal growth factor receptor (EGFR) is a promising treatment approach for NSCLC. However, resistance to EGFR tyrosine kinase inhibitors (TKIs) remains a major challenge in its clinical management. EGFR mutation elevates the expression of hypoxia-inducible factor-1 alpha to upregulate the production of glycolytic enzymes, increasing glycolysis and tumor resistance. The inhibition of glycolysis can be a potential strategy for overcoming EGFR-TKI resistance and enhancing the effectiveness of EGFR-TKIs. In this review, we specifically explored the effectiveness of pyruvate dehydrogenase kinase inhibitors and lactate dehydrogenase A inhibitors in combating EGFR-TKI resistance. The aim was to summarize the effects of these natural products in preclinical NSCLC models to provide a comprehensive understanding of the potential therapeutic effects. The study findings suggest that natural products can be promising inhibitors of glycolytic enzymes for the treatment of EGFR-TKI-resistant NSCLC. Further investigations through preclinical and clinical studies are required to validate the efficacy of natural product-based glycolytic inhibitors as innovative therapeutic modalities for NSCLC.
Collapse
Affiliation(s)
- Wonyoung Park
- Department of Korean Medical Science, School of Korean Medicine, Pusan National University, Yangsan 50612, Republic of Korea;
- Korean Medical Research Center for Healthy Aging, Pusan National University, Yangsan 50612, Republic of Korea; (E.-S.Y.); (S.-Y.C.)
| | - Jung Ho Han
- Korean Medicine Application Center, Korea Institute of Oriental Medicine, Daegu 41062, Republic of Korea;
| | - Shibo Wei
- Department of Molecular Cell Biology, School of Medicine, Sungkyunkwan University, Suwon 16419, Republic of Korea;
| | - Eun-Sun Yang
- Korean Medical Research Center for Healthy Aging, Pusan National University, Yangsan 50612, Republic of Korea; (E.-S.Y.); (S.-Y.C.)
| | - Se-Yun Cheon
- Korean Medical Research Center for Healthy Aging, Pusan National University, Yangsan 50612, Republic of Korea; (E.-S.Y.); (S.-Y.C.)
| | - Sung-Jin Bae
- Department of Molecular Biology and Immunology, Kosin University College of Medicine, Busan 49267, Republic of Korea;
| | - Dongryeol Ryu
- Department of Biomedical Science and Engineering, Gwangju Institute of Science and Technology, Gwangju 61005, Republic of Korea;
| | - Hwan-Suck Chung
- Korean Medicine Application Center, Korea Institute of Oriental Medicine, Daegu 41062, Republic of Korea;
| | - Ki-Tae Ha
- Department of Korean Medical Science, School of Korean Medicine, Pusan National University, Yangsan 50612, Republic of Korea;
- Korean Medical Research Center for Healthy Aging, Pusan National University, Yangsan 50612, Republic of Korea; (E.-S.Y.); (S.-Y.C.)
| |
Collapse
|
114
|
Chen B, Wang Y, Wu Y, Xu T. Effect of HPV Oncoprotein on Carbohydrate and Lipid Metabolism in Tumor Cells. Curr Cancer Drug Targets 2024; 24:987-1004. [PMID: 38284713 DOI: 10.2174/0115680096266981231215111109] [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: 07/09/2023] [Revised: 09/29/2023] [Accepted: 11/13/2023] [Indexed: 01/30/2024]
Abstract
High-risk HPV infection accounts for 99.7% of cervical cancer, over 90% of anal cancer, 50% of head and neck cancers, 40% of vulvar cancer, and some cases of vaginal and penile cancer, contributing to approximately 5% of cancers worldwide. The development of cancer is a complex, multi-step process characterized by dysregulation of signaling pathways and alterations in metabolic pathways. Extensive research has demonstrated that metabolic reprogramming plays a key role in the progression of various cancers, such as cervical, head and neck, bladder, and prostate cancers, providing the material and energy foundation for rapid proliferation and migration of cancer cells. Metabolic reprogramming of tumor cells allows for the rapid generation of ATP, aiding in meeting the high energy demands of HPV-related cancer cell proliferation. The interaction between Human Papillomavirus (HPV) and its associated cancers has become a recent focus of investigation. The impact of HPV on cellular metabolism has emerged as an emerging research topic. A significant body of research has shown that HPV influences relevant metabolic signaling pathways, leading to cellular metabolic alterations. Exploring the underlying mechanisms may facilitate the discovery of biomarkers for diagnosis and treatment of HPV-associated diseases. In this review, we introduced the molecular structure of HPV and its replication process, discussed the diseases associated with HPV infection, described the energy metabolism of normal cells, highlighted the metabolic features of tumor cells, and provided an overview of recent advances in potential therapeutic targets that act on cellular metabolism. We discussed the potential mechanisms underlying these changes. This article aims to elucidate the role of Human Papillomavirus (HPV) in reshaping cellular metabolism and the application of metabolic changes in the research of related diseases. Targeting cancer metabolism may serve as an effective strategy to support traditional cancer treatments, as metabolic reprogramming is crucial for malignant transformation in cancer.
Collapse
Affiliation(s)
- Biqing Chen
- The Second Hospital of Jilin University, Changchun, China
| | - Yichao Wang
- The Second Hospital of Jilin University, Changchun, China
| | - Yishi Wu
- The Second Hospital of Jilin University, Changchun, China
| | - Tianmin Xu
- The Second Hospital of Jilin University, Changchun, China
| |
Collapse
|
115
|
Vara-Ciruelos D, Malumbres M. Cross Talk Between Metabolism and the Cell Division Cycle. Methods Mol Biol 2024; 2740:141-154. [PMID: 38393474 DOI: 10.1007/978-1-0716-3557-5_9] [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] [Indexed: 02/25/2024]
Abstract
Cell division requires a massive rewiring of cellular pathways, including molecular routes involved in providing energy for cell survival and functionality. The energetic requirements and the metabolic opportunities for generating energy change during the different phases of the cell cycle and how these processes are connected is still poorly understood. This chapter discusses basic concepts for a coordinated analysis of cell cycle progression and metabolism and provides specific protocols for studying these two connected processes in mammalian cells.
Collapse
Affiliation(s)
- Diana Vara-Ciruelos
- Cell Division and Cancer group, Spanish National Cancer Research Center (CNIO), Madrid, Spain
| | - Marcos Malumbres
- Cell Division and Cancer group, Spanish National Cancer Research Center (CNIO), Madrid, Spain.
- Cancer Cell Cycle group, Vall d'Hebron Institute of Oncology (VHIO), Barcelona, Spain.
- ICREA, Barcelona, Spain.
| |
Collapse
|
116
|
Rezaei M, Ghasemitarei M, Razzokov J, Yusupov M, Ghorbanalilu M, Ejtehadi MR. In silico study of the impact of oxidation on pyruvate transmission across the hVDAC1 protein channel. Arch Biochem Biophys 2024; 751:109835. [PMID: 38000492 DOI: 10.1016/j.abb.2023.109835] [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: 09/14/2023] [Revised: 10/25/2023] [Accepted: 11/20/2023] [Indexed: 11/26/2023]
Abstract
The overexpression of voltage dependent anion channels (VDACs), particularly VDAC1, in cancer cells compared to normal cells, plays a crucial role in cancer cell metabolism, apoptosis regulation, and energy homeostasis. In this study, we used molecular dynamics (MD) simulations to investigate the effect of a low level of VDAC1 oxidation (induced e.g., by cold atmospheric plasma (CAP)) on the pyruvate (Pyr) uptake by VDAC1. Inhibiting Pyr uptake through VDAC1 can suppress cancer cell proliferation. Our primary target was to study the translocation of Pyr across the native and oxidized forms of hVDAC1, the human VDAC1. Specifically, we employed MD simulations to analyze the hVDAC1 structure by modifying certain cysteine residues to cysteic acids and methionine residues to methionine sulfoxides, which allowed us to investigate the effect of oxidation. Our results showed that the free energy barrier for Pyr translocation through the native and oxidized channel was approximately 4.3 ± 0.7 kJ mol-1 and 10.8 ± 1.8 kJ mol-1, respectively. An increase in barrier results in a decrease in rate of Pyr permeation through the oxidized channel. Thus, our results indicate that low levels of CAP oxidation reduce Pyr translocation, resulting in decreased cancer cell proliferation. Therefore, low levels of oxidation are likely sufficient to treat cancer cells given the inhibition of Pyr uptake.
Collapse
Affiliation(s)
- Mahsa Rezaei
- Department of Physics, Shahid Beheshti University, Tehran, 19839-69411, Iran
| | - Maryam Ghasemitarei
- Department of Physics, Sharif University of Technology, 14588-89694, Tehran, Iran; Research Group PLASMANT, Department of Chemistry, University of Antwerp, 2610, Antwerp, Belgium.
| | - Jamoliddin Razzokov
- Institute of Fundamental and Applied Research, National Research University TIIAME, 100000, Tashkent, Uzbekistan; School of Engineering, Central Asian University, Tashkent, 111221, Uzbekistan; Laboratory of Experimental Biophysics, Centre for Advanced Technologies, 100174, Tashkent, Uzbekistan; Department of Chemistry, Termez State University, 190111, Termez, Uzbekistan
| | - Maksudbek Yusupov
- School of Engineering, New Uzbekistan University, 100000, Tashkent, Uzbekistan; Department of Information Technologies, Tashkent International University of Education, 100207, Tashkent, Uzbekistan; Laboratory of Thermal Physics of Multiphase Systems, Arifov Institute of Ion-Plasma and Laser Technologies, Academy of Sciences of Uzbekistan, 100125, Tashkent, Uzbekistan; Department of Power Supply and Renewable Energy Sources, National Research University TIIAME, 100000, Tashkent, Uzbekistan
| | | | | |
Collapse
|
117
|
Chu D, Qu H, Huang X, Shi Y, Li K, Lin W, Xu Z, Li D, Chen H, Gao L, Wang W, Wang H. Manganese Amplifies Photoinduced ROS in Toluidine Blue Carbon Dots to Boost MRI Guided Chemo/Photodynamic Therapy. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2024; 20:e2304968. [PMID: 37715278 DOI: 10.1002/smll.202304968] [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: 06/13/2023] [Revised: 09/04/2023] [Indexed: 09/17/2023]
Abstract
The contrast agents and tumor treatments currently used in clinical practice are far from satisfactory, due to the specificity of the tumor microenvironment (TME). Identification of diagnostic and therapeutic reagents with strong contrast and therapeutic effect remains a great challenge. Herein, a novel carbon dot nanozyme (Mn-CD) is synthesized for the first time using toluidine blue (TB) and manganese as raw materials. As expected, the enhanced magnetic resonance (MR) imaging capability of Mn-CDs is realized in response to the TME (acidity and glutathione), and r1 and r2 relaxation rates are enhanced by 224% and 249%, respectively. In addition, the photostability of Mn-CDs is also improved, and show an efficient singlet oxygen (1 O2 ) yield of 1.68. Moreover, Mn-CDs can also perform high-efficiency peroxidase (POD)-like activity and catalyze hydrogen peroxide to hydroxyl radicals, which is greatly improved under the light condition. The results both in vitro and in vivo demonstrate that the Mn-CDs are able to achieve real-time MR imaging of TME responsiveness through aggregation of the enhanced permeability and retention effect at tumor sites and facilitate light-enhanced chemodynamic and photodynamic combination therapies. This work opens a new perspective in terms of the role of carbon nanomaterials in integrated diagnosis and treatment of diseases.
Collapse
Affiliation(s)
- Dongchuan Chu
- Affiliated Hospital of Yangzhou University, Yangzhou University, Yangzhou, 225001, China
| | - Hang Qu
- Affiliated Hospital of Yangzhou University, Yangzhou University, Yangzhou, 225001, China
| | - Xueping Huang
- Institute of Translational Medicine, Medical College, Yangzhou University, Yangzhou, 225001, China
| | - Yu Shi
- Affiliated Hospital of Yangzhou University, Yangzhou University, Yangzhou, 225001, China
- Institute of Translational Medicine, Medical College, Yangzhou University, Yangzhou, 225001, China
| | - Ke Li
- Affiliated Hospital of Yangzhou University, Yangzhou University, Yangzhou, 225001, China
- Institute of Translational Medicine, Medical College, Yangzhou University, Yangzhou, 225001, China
| | - Wenzheng Lin
- Affiliated Hospital of Yangzhou University, Yangzhou University, Yangzhou, 225001, China
- Institute of Translational Medicine, Medical College, Yangzhou University, Yangzhou, 225001, China
| | - Zhuobin Xu
- Institute of Translational Medicine, Medical College, Yangzhou University, Yangzhou, 225001, China
| | - Dandan Li
- Institute of Translational Medicine, Medical College, Yangzhou University, Yangzhou, 225001, China
| | - Hao Chen
- Affiliated Hospital of Yangzhou University, Yangzhou University, Yangzhou, 225001, China
- Institute of Translational Medicine, Medical College, Yangzhou University, Yangzhou, 225001, China
| | - Lizeng Gao
- CAS Engineering Laboratory for Nanozyme, Institute of Biophysics, Chinese Academy of Sciences, Beijing, 100101, China
| | - Wei Wang
- Affiliated Hospital of Yangzhou University, Yangzhou University, Yangzhou, 225001, China
| | - Huihui Wang
- Affiliated Hospital of Yangzhou University, Yangzhou University, Yangzhou, 225001, China
- Institute of Translational Medicine, Medical College, Yangzhou University, Yangzhou, 225001, China
| |
Collapse
|
118
|
Moloudi K, Abrahamse H, George BP. Nanotechnology-mediated photodynamic therapy: Focus on overcoming tumor hypoxia. WILEY INTERDISCIPLINARY REVIEWS. NANOMEDICINE AND NANOBIOTECHNOLOGY 2024; 16:e1937. [PMID: 38072393 DOI: 10.1002/wnan.1937] [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: 06/15/2023] [Revised: 10/05/2023] [Accepted: 11/20/2023] [Indexed: 03/09/2024]
Abstract
The oxygen level in the tumor is a critical marker that determines response to different treatments. Cancerous cells can adapt to hypoxia and low pH conditions within the tumor microenvironment (TME) to regulate tumor metabolism, proliferation, and promote tumor metastasis as well as angiogenesis, consequently leading to treatment failure and recurrence. In recent years, widespread attempts have been made to overcome tumor hypoxia through different methods, such as hyperbaric oxygen therapy (HBOT), hyperthermia, O2 carriers, artificial hemoglobin, oxygen generator hydrogels, and peroxide materials. While oxygen is found to be an essential agent to improve the treatment response of photodynamic therapy (PDT) and other cancer treatment modalities, the development of hypoxia within the tumor is highly associated with PDT failure. Recently, the use of nanoparticles has been a hot topic for researchers and exploited to overcome hypoxia through Oxygen-generating hydrogels, O2 nanocarriers, and O2 -generating nanoparticles. This review aimed to discuss the role of nanotechnology in tumor oxygenation and highlight the challenges, prospective, and recent advances in this area to improve PDT outcomes. This article is categorized under: Nanotechnology Approaches to Biology > Cells at the Nanoscale Nanotechnology Approaches to Biology > Nanoscale Systems in Biology Therapeutic Approaches and Drug Discovery > Nanomedicine for Oncologic Disease.
Collapse
Affiliation(s)
- Kave Moloudi
- Laser Research Centre (LRC), Faculty of Health Sciences, University of Johannesburg, Johannesburg, South Africa
| | - Heidi Abrahamse
- Laser Research Centre (LRC), Faculty of Health Sciences, University of Johannesburg, Johannesburg, South Africa
| | - Blassan P George
- Laser Research Centre (LRC), Faculty of Health Sciences, University of Johannesburg, Johannesburg, South Africa
| |
Collapse
|
119
|
Liu F, Xiang Z, Li Q, Fang X, Zhou J, Yang X, Lin H, Yang Q. 18F-FDG PET/CT-based radiomics model for predicting the degree of pathological differentiation in non-small cell lung cancer: a multicentre study. Clin Radiol 2024; 79:e147-e155. [PMID: 37884401 DOI: 10.1016/j.crad.2023.09.017] [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: 04/28/2023] [Revised: 09/18/2023] [Accepted: 09/20/2023] [Indexed: 10/28/2023]
Abstract
AIM To explore the value of 2-[18F]-fluoro-2-deoxy-d-glucose (FDG) positron-emission tomography (PET)/computed tomography (CT)-based radiomics model for predicting the degree of pathological differentiation in non-small-cell lung cancer (NSCLC). MATERIALS AND METHODS Clinical characteristics of 182 NSCLC patients from four centres were collected, and radiomics features were extracted from 18F-FDG PET/CT images. Three logistic regression prediction models were established: clinical model; radiomics model; and nomogram combining radiomics signatures and clinical features. The predictive ability of the models was assessed using receiver operating characteristics curve analysis. RESULTS Patients from centre 1 were assigned randomly to the training and internal validation cohorts (7:3 ratio); patients from centres 2-4 served as the external validation cohort. The area under the curve (AUC) values for the clinical model in the training, internal validation, and external validation cohort were 0.74 (95% confidence interval [CI] = 0.64-0.84), 0.64 (95% CI = 0.46-0.81), and 0.74 (95% CI = 0.60-0.88), respectively. In the training (AUC: 0.84 [95% CI = 0.77-0.92]), internal validation (AUC: 0.81 [95% CI = 0.67-0.95]), and external validation cohorts (AUC: 0.74 [95% CI = 0.58-0.89]), the radiomics model showed good predictive ability for differentiation. Compared to the clinical and radiomics models, the nomogram has relatively better diagnostic performance, and the AUC values for nomogram in the training, internal validation, and external validation cohort were 0.86 (95% CI = 0.78-0.93), 0.83 (95% CI = 0.70-0.96), and 0.77 (95% CI = 0.62-0.92), respectively. CONCLUSIONS The 18F-FDG PET/CT-based radiomics model showed good ability for predicting the degree of differentiation of NSCLC. The nomogram combining the radiomics signature and clinical features has relatively better diagnostic performance.
Collapse
Affiliation(s)
- F Liu
- Department of Radiology, The Second Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang, Hunan 421001, China
| | - Z Xiang
- Department of Radiology, The Second Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang, Hunan 421001, China
| | - Q Li
- Department of Radiology, The Second Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang, Hunan 421001, China
| | - X Fang
- Department of Radiology, The Second Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang, Hunan 421001, China.
| | - J Zhou
- The Affiliated Hospital of Jiangsu University, Zhenjiang, Jiangsu 212001, China
| | - X Yang
- Sichuan Science City Hospital, Mianyang, Sichuan 621000, China
| | - H Lin
- Department of Pharmaceutical Diagnosis, GE Healthcare, Changsha 410005, China
| | - Q Yang
- Center for Molecular Imaging Probe, Hunan Province Key Laboratory of Tumour Cellular and Molecular Pathology, Cancer Research Institute, Hengyang Medical School, University of South China, Hengyang, Hunan 421001, China.
| |
Collapse
|
120
|
Ma PQ, Huang FW, Xie YQ, Li HR, Li HD, Ye BC, Yin BC. Universal DNA-Based Sensing Toolbox for Programming Cell Functions. J Am Chem Soc 2023; 145:28224-28232. [PMID: 38108623 DOI: 10.1021/jacs.3c11232] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2023]
Abstract
By recombining natural cell signaling systems and further reprogramming cell functions, use of genetically engineered cells and bacteria as therapies is an innovative emerging concept. However, the inherent properties and structures of the natural signal sensing and response pathways constrain further development. We present a universal DNA-based sensing toolbox on the cell surface to endow new signal sensing abilities for cells, control cell states, and reprogram multiple cell functions. The sensing toolbox contains a triangular-prismatic-shaped DNA origami framework and a sensing core anchored inside the internal confined space to enhance the specificity and efficacy of the toolbox. As a proof of principle, the sensing toolbox uses the customizable sensing core with signal sensing switches and converters to recognize unconventional signal inputs, deliver functional components to cells, and then control cell responses, including specific tumor cell death, immune cell disinhibition and adhesion, and bacterial expression. This work expands the diversity of cell sensing signals and reprograms biological functions by constructing nanomechanical-natural hybrid cells, providing new strategies for engineering cells and bacteria in diagnosis and treatment applications.
Collapse
Affiliation(s)
- Pei-Qiang Ma
- Lab of Biosystem and Microanalysis, State Key Laboratory of Bioreactor Engineering, Shanghai Collaborative Innovation Center for Biomanufacturing Technology, East China University of Science and Technology, Shanghai 200237, China
| | - Fu-Wen Huang
- Lab of Biosystem and Microanalysis, State Key Laboratory of Bioreactor Engineering, Shanghai Collaborative Innovation Center for Biomanufacturing Technology, East China University of Science and Technology, Shanghai 200237, China
| | - Ya-Qi Xie
- Lab of Biosystem and Microanalysis, State Key Laboratory of Bioreactor Engineering, Shanghai Collaborative Innovation Center for Biomanufacturing Technology, East China University of Science and Technology, Shanghai 200237, China
| | - Hong-Rui Li
- Lab of Biosystem and Microanalysis, State Key Laboratory of Bioreactor Engineering, Shanghai Collaborative Innovation Center for Biomanufacturing Technology, East China University of Science and Technology, Shanghai 200237, China
| | - Hua-Dong Li
- Lab of Biosystem and Microanalysis, State Key Laboratory of Bioreactor Engineering, Shanghai Collaborative Innovation Center for Biomanufacturing Technology, East China University of Science and Technology, Shanghai 200237, China
| | - Bang-Ce Ye
- Lab of Biosystem and Microanalysis, State Key Laboratory of Bioreactor Engineering, Shanghai Collaborative Innovation Center for Biomanufacturing Technology, East China University of Science and Technology, Shanghai 200237, China
- Institute of Engineering Biology and Health, Collaborative Innovation Center of Yangtze River Delta Region Green Pharmaceuticals, College of Pharmaceutical Sciences, Zhejiang University of Technology, Hangzhou, Zhejiang 310014, China
| | - Bin-Cheng Yin
- Lab of Biosystem and Microanalysis, State Key Laboratory of Bioreactor Engineering, Shanghai Collaborative Innovation Center for Biomanufacturing Technology, East China University of Science and Technology, Shanghai 200237, China
- School of Chemistry and Chemical Engineering, Shihezi University, Shihezi, Xinjiang 832000, China
| |
Collapse
|
121
|
Isowa M, Hamaguchi R, Narui R, Morikawa H, Okamoto T, Wada H. Potential of Alkalization Therapy for the Management of Metastatic Pancreatic Cancer: A Retrospective Study. Cancers (Basel) 2023; 16:61. [PMID: 38201489 PMCID: PMC10777900 DOI: 10.3390/cancers16010061] [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: 10/18/2023] [Revised: 12/07/2023] [Accepted: 12/11/2023] [Indexed: 01/12/2024] Open
Abstract
Current treatments for patients with pancreatic cancer offer limited benefits. In this study, we applied alkalization therapy, which was efficacious for other solid tumors at our clinic, to stage 4 pancreatic cancer patients, and investigated its effect on disease prognosis. Patients with metastatic pancreatic cancer who were treated at Karasuma Wada Clinic in Kyoto, Japan, between January 2011 and April 2022, were included in the study. All patients received alkalization therapy (a combination of an alkaline diet, bicarbonate, and citric acid administration), alongside standard chemotherapy. Urine samples were collected to assess urine pH as a marker of whole-body alkalization. In the 98 patients analyzed, the median overall survival (OS) from the time of diagnosis was 13.2 months. Patients with a mean urine pH of 7.5 or greater had a median OS of 29.9 months, compared with 15.2 months for those with a mean urine pH of 6.5 to 7.5, and 8.0 months for those with a mean urine pH of less than 6.5, which suggests a trend of a longer OS in patients with a higher urine pH (p = 0.0639). Alkalization therapy may offer a viable approach to extending the survival of stage 4 pancreatic cancer patients, who typically have an unfavorable prognosis.
Collapse
Affiliation(s)
- Masahide Isowa
- Japanese Society on Inflammation and Metabolism in Cancer, 119 Nishioshikouji-cho, Nakagyo-ku, Kyoto 604-0842, Japan; (M.I.); (R.N.); (H.M.); (H.W.)
| | - Reo Hamaguchi
- Japanese Society on Inflammation and Metabolism in Cancer, 119 Nishioshikouji-cho, Nakagyo-ku, Kyoto 604-0842, Japan; (M.I.); (R.N.); (H.M.); (H.W.)
| | - Ryoko Narui
- Japanese Society on Inflammation and Metabolism in Cancer, 119 Nishioshikouji-cho, Nakagyo-ku, Kyoto 604-0842, Japan; (M.I.); (R.N.); (H.M.); (H.W.)
| | - Hiromasa Morikawa
- Japanese Society on Inflammation and Metabolism in Cancer, 119 Nishioshikouji-cho, Nakagyo-ku, Kyoto 604-0842, Japan; (M.I.); (R.N.); (H.M.); (H.W.)
| | - Toshihiro Okamoto
- Department of Thoracic and Cardiovascular Surgery, Cleveland Clinic, Cleveland, OH 44195, USA;
- Department of Inflammation and Immunology, Lerner Research Institute, Cleveland Clinic, Cleveland, OH 44195, USA
- Transplant Center, Cleveland Clinic, Cleveland, OH 44195, USA
| | - Hiromi Wada
- Japanese Society on Inflammation and Metabolism in Cancer, 119 Nishioshikouji-cho, Nakagyo-ku, Kyoto 604-0842, Japan; (M.I.); (R.N.); (H.M.); (H.W.)
| |
Collapse
|
122
|
Schmidt K, Thatcher A, Grobe A, Hicks L, Gu H, Sears DD, Ellies LG, Kalachev L, Kroll E. The Combined Treatment with Ketogenic Diet and Metformin Slows Tumor Growth in Two Mouse Models of Triple Negative Breast Cancer. RESEARCH SQUARE 2023:rs.3.rs-3664129. [PMID: 38196628 PMCID: PMC10775859 DOI: 10.21203/rs.3.rs-3664129/v1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/11/2024]
Abstract
BACKGROUND Many tumors contain hypoxic microenvironments caused by inefficient tumor vascularization. Hypoxic tumors have been shown to resist conventional cancer therapies. Hypoxic cancer cells rely on glucose to meet their energetic and anabolic needs to fuel uncontrolled proliferation and metastasis. This glucose dependency is linked to a metabolic shift in response to hypoxic conditions. METHODS To leverage the glucose dependency of hypoxic tumor cells, we assessed the effects of a controlled reduction in systemic glucose by combining dietary carbohydrate restriction, using a ketogenic diet, with gluconeogenesis inhibition, using metformin, on two mouse models of triple-negative breast cancer (TNBC). RESULTS We confirmed that MET - 1 breast cancer cells require abnormally high glucose concentrations to survive in a hypoxic environment in vitro. Then, we showed that, compared to a ketogenic diet or metformin alone, animals treated with the combination regimen showed significantly lower tumor burden, higher tumor latency and slower tumor growth. As a result, lowering systemic glucose by this combined dietary and pharmacologic approach improved overall survival in our mouse model by 31 days, which is approximately equivalent to 3 human years. CONCLUSION This is the first preclinical study to demonstrate that reducing systemic glucose by combining a ketogenic diet and metformin significantly inhibits tumor proliferation and increases overall survival. Our findings suggest a possible treatment for a broad range of hypoxic and glycolytic tumor types, one that can also augment existing treatment options to improve patient outcomes.
Collapse
Affiliation(s)
- Karen Schmidt
- University of Montana Division of Biological Sciences
| | | | | | - Linda Hicks
- University of Montana Division of Biological Sciences
| | - Haiwei Gu
- Arizona State University School of Life Sciences
| | | | | | | | - Eugene Kroll
- University of Montana Missoula: University of Montana
| |
Collapse
|
123
|
Trejo-Solis C, Silva-Adaya D, Serrano-García N, Magaña-Maldonado R, Jimenez-Farfan D, Ferreira-Guerrero E, Cruz-Salgado A, Castillo-Rodriguez RA. Role of Glycolytic and Glutamine Metabolism Reprogramming on the Proliferation, Invasion, and Apoptosis Resistance through Modulation of Signaling Pathways in Glioblastoma. Int J Mol Sci 2023; 24:17633. [PMID: 38139462 PMCID: PMC10744281 DOI: 10.3390/ijms242417633] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2023] [Revised: 12/11/2023] [Accepted: 12/14/2023] [Indexed: 12/24/2023] Open
Abstract
Glioma cells exhibit genetic and metabolic alterations that affect the deregulation of several cellular signal transduction pathways, including those related to glucose metabolism. Moreover, oncogenic signaling pathways induce the expression of metabolic genes, increasing the metabolic enzyme activities and thus the critical biosynthetic pathways to generate nucleotides, amino acids, and fatty acids, which provide energy and metabolic intermediates that are essential to accomplish the biosynthetic needs of glioma cells. In this review, we aim to explore how dysregulated metabolic enzymes and their metabolites from primary metabolism pathways in glioblastoma (GBM) such as glycolysis and glutaminolysis modulate anabolic and catabolic metabolic pathways as well as pro-oncogenic signaling and contribute to the formation, survival, growth, and malignancy of glioma cells. Also, we discuss promising therapeutic strategies by targeting the key players in metabolic regulation. Therefore, the knowledge of metabolic reprogramming is necessary to fully understand the biology of malignant gliomas to improve patient survival significantly.
Collapse
Affiliation(s)
- Cristina Trejo-Solis
- Laboratorio Experimental de Enfermedades Neurodegenerativas, Laboratorio de Reprogramación Celular, Departamento de Neurofisiología, Instituto Nacional de Neurología y Neurocirugía, Ciudad de Mexico 14269, Mexico; (D.S.-A.); (N.S.-G.); (R.M.-M.)
| | - Daniela Silva-Adaya
- Laboratorio Experimental de Enfermedades Neurodegenerativas, Laboratorio de Reprogramación Celular, Departamento de Neurofisiología, Instituto Nacional de Neurología y Neurocirugía, Ciudad de Mexico 14269, Mexico; (D.S.-A.); (N.S.-G.); (R.M.-M.)
| | - Norma Serrano-García
- Laboratorio Experimental de Enfermedades Neurodegenerativas, Laboratorio de Reprogramación Celular, Departamento de Neurofisiología, Instituto Nacional de Neurología y Neurocirugía, Ciudad de Mexico 14269, Mexico; (D.S.-A.); (N.S.-G.); (R.M.-M.)
| | - Roxana Magaña-Maldonado
- Laboratorio Experimental de Enfermedades Neurodegenerativas, Laboratorio de Reprogramación Celular, Departamento de Neurofisiología, Instituto Nacional de Neurología y Neurocirugía, Ciudad de Mexico 14269, Mexico; (D.S.-A.); (N.S.-G.); (R.M.-M.)
| | - Dolores Jimenez-Farfan
- Laboratorio de Inmunología, División de Estudios de Posgrado e Investigación, Facultad de Odontología, Universidad Nacional Autónoma de México, Ciudad de Mexico 04510, Mexico;
| | - Elizabeth Ferreira-Guerrero
- Centro de Investigación Sobre Enfermedades Infecciosas, Instituto Nacional de Salud Pública, Cuernavaca 62100, Mexico; (E.F.-G.); (A.C.-S.)
| | - Arturo Cruz-Salgado
- Centro de Investigación Sobre Enfermedades Infecciosas, Instituto Nacional de Salud Pública, Cuernavaca 62100, Mexico; (E.F.-G.); (A.C.-S.)
| | | |
Collapse
|
124
|
Tanaka N, Okada H, Yamaguchi K, Seki M, Matsubara D, Gotoh N, Suzuki Y, Furukawa Y, Yamashita T, Inoue JI, Kaneko S, Sakamoto T. Mint3-depletion-induced energy stress sensitizes triple-negative breast cancer to chemotherapy via HSF1 inactivation. Cell Death Dis 2023; 14:815. [PMID: 38081808 PMCID: PMC10713533 DOI: 10.1038/s41419-023-06352-4] [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: 05/08/2023] [Revised: 11/23/2023] [Accepted: 11/29/2023] [Indexed: 12/18/2023]
Abstract
Given the lack of therapeutic targets, the conventional approach for managing triple-negative breast cancer (TNBC) involves the utilization of cytotoxic chemotherapeutic agents. However, most TNBCs acquire resistance to chemotherapy, thereby lowering the therapeutic outcome. In addition to oncogenic mutations in TNBC, microenvironment-induced mechanisms render chemoresistance more complex and robust in vivo. Here, we aimed to analyze whether depletion of Munc18-1 interacting protein 3 (Mint3), which activates hypoxia-inducible factor 1 (HIF-1) during normoxia, sensitizes TNBC to chemotherapy. We found that Mint3 promotes the chemoresistance of TNBC in vivo. Mint3 depletion did not affect the sensitivity of human TNBC cell lines to doxorubicin and paclitaxel in vitro but sensitized tumors of these cells to chemotherapy in vivo. Transcriptome analyses revealed that the Mint3-HIF-1 axis enhanced heat shock protein 70 (HSP70) expression in tumors of TNBC cells. Administering an HSP70 inhibitor enhanced the antitumor activity of doxorubicin in TNBC tumors, similar to Mint3 depletion. Mint3 expression was also correlated with HSP70 expression in human TNBC specimens. Mechanistically, Mint3 depletion induces glycolytic maladaptation to the tumor microenvironment in TNBC tumors, resulting in energy stress. This energy stress by Mint3 depletion inactivated heat shock factor 1 (HSF-1), the master regulator of HSP expression, via the AMP-activated protein kinase/mechanistic target of the rapamycin pathway following attenuated HSP70 expression. In conclusion, Mint3 is a unique regulator of TNBC chemoresistance in vivo via metabolic adaptation to the tumor microenvironment, and a combination of Mint3 inhibition and chemotherapy may be a good strategy for TNBC treatment.
Collapse
Affiliation(s)
- Noritaka Tanaka
- Department of Cancer Biology, Institute of Biomedical Science, Kansai Medical University, Osaka, Japan
| | - Hikari Okada
- Information-Based Medicine Development, Graduate School of Medical Sciences, Kanazawa University, Ishikawa, Japan
| | - Kiyoshi Yamaguchi
- Division of Clinical Genome Research, the Institute of Medical Science, The University of Tokyo, Tokyo, Japan
| | - Masahide Seki
- Department of Computational Biology and Medical Sciences, Graduate School of Frontier Sciences, The University of Tokyo, Chiba, Japan
| | | | - Noriko Gotoh
- Division of Cancer Cell Biology, Cancer Research Institute, Kanazawa University, Ishikawa, Japan
| | - Yutaka Suzuki
- Department of Computational Biology and Medical Sciences, Graduate School of Frontier Sciences, The University of Tokyo, Chiba, Japan
| | - Yoichi Furukawa
- Division of Clinical Genome Research, the Institute of Medical Science, The University of Tokyo, Tokyo, Japan
| | - Taro Yamashita
- Department of System Biology, Institute of Medical, Pharmaceutical and Health Sciences, Kanazawa University, Ishikawa, Japan
| | - Jun-Ichiro Inoue
- The University of Tokyo Pandemic Preparedness, Infection and Advanced Research Center (UTOPIA), Tokyo, Japan
| | - Shuichi Kaneko
- Information-Based Medicine Development, Graduate School of Medical Sciences, Kanazawa University, Ishikawa, Japan
| | - Takeharu Sakamoto
- Department of Cancer Biology, Institute of Biomedical Science, Kansai Medical University, Osaka, Japan.
- Department of System Biology, Institute of Medical, Pharmaceutical and Health Sciences, Kanazawa University, Ishikawa, Japan.
| |
Collapse
|
125
|
Singh N, Marwaha D, Gautam S, Rai N, Tiwari P, Sharma M, Shukla RP, Mugale MN, Kumar A, Mishra PR. Surface-Modified Lyotropic Crystalline Nanoconstructs Bearing Doxorubicin and Buparvaquone Target Sigma Receptors through pH-Sensitive Charge Conversion to Improve Breast Cancer Therapy. Biomacromolecules 2023; 24:5780-5796. [PMID: 38006339 DOI: 10.1021/acs.biomac.3c00795] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2023]
Abstract
In the current study, we aimed to develop lyotropic crystalline nanoconstructs (LCNs) based on poly(l-glutamic acid) (PLG) with a two-tier strategy. The first objective was to confer pH-responsive charge conversion properties to facilitate the delivery of both doxorubicin (DOX) and buparvaquone (BPQ) in combination (B + D@LCNs) to harness their synergistic effects. The second goal was to achieve targeted delivery to sigma receptors within the tumor tissues. To achieve this, we designed a pH-responsive charge conversion system using a polymer consisting of poly(ethylenimine), poly(l-lysine), and poly(l-glutamic acid) (PLG), which was then covalently coupled with methoxybenzamide (MBA) for potential sigma receptor targeting. The resulting B + D@LCNs were further modified by surface functionalization with PLG-MBA to confer both sigma receptor targeting and pH-responsive charge conversion properties. Our observations indicated that at physiological pH 7.4, P/B + D-MBA@LCNs exhibited a negative charge, while under acidic conditions (pH 5.5, characteristic of the tumor microenvironment), they acquired a positive charge. The particle size of P/B + D-MBA@LCNs was determined to be 168.23 ± 2.66 nm at pH 7.4 and 201.23 ± 1.46 nm at pH 5.5. The crystalline structure of the LCNs was confirmed through small-angle X-ray scattering (SAXS) diffraction patterns. Receptor-mediated endocytosis, facilitated by P/B + D-MBA@LCNs, was confirmed using confocal laser scanning microscopy and flow cytometry. The P/B + D-MBA@LCNs formulation demonstrated a higher rate of G2/M phase arrest (55.20%) compared to free B + D (37.50%) and induced mitochondrial depolarization (59.39%) to a greater extent than P/B + D@LCNs (45.66%). Pharmacokinetic analysis revealed significantly improved area under the curve (AUC) values for both DOX and BPQ when administered as P/B + D-MBA@LCNs, along with enhanced tumor localization. Tumor regression studies exhibited a substantial reduction in tumor size, with P/B + D-MBA@LCNs leading to 3.2- and 1.27-fold reductions compared to B + D and nontargeted P/B + D@LCNs groups, respectively. In summary, this two-tier strategy demonstrates substantial promise for the delivery of a drug combination through the prototype formulation. It offers a potential chemotherapeutic option by minimizing toxic effects on healthy cells while maximizing therapeutic efficacy.
Collapse
Affiliation(s)
- Neha Singh
- Division of Pharmaceutics and Pharmacokinetics, CSIR-Central Drug Research Institute, Lucknow 226031, India
- Academy of Scientific and Innovation Research (AcSIR), Ghaziabad 201002, U.P., India
| | - Disha Marwaha
- Division of Pharmaceutics and Pharmacokinetics, CSIR-Central Drug Research Institute, Lucknow 226031, India
| | - Shalini Gautam
- Division of Pharmaceutics and Pharmacokinetics, CSIR-Central Drug Research Institute, Lucknow 226031, India
| | - Nikhil Rai
- Division of Pharmaceutics and Pharmacokinetics, CSIR-Central Drug Research Institute, Lucknow 226031, India
| | - Pratiksha Tiwari
- Division of Pharmaceutics and Pharmacokinetics, CSIR-Central Drug Research Institute, Lucknow 226031, India
| | - Madhu Sharma
- Division of Pharmaceutics and Pharmacokinetics, CSIR-Central Drug Research Institute, Lucknow 226031, India
| | - Ravi Prakash Shukla
- Division of Pharmaceutics and Pharmacokinetics, CSIR-Central Drug Research Institute, Lucknow 226031, India
| | | | - Akhilesh Kumar
- Division of Toxicology, CSIR-Central Drug Research Institute, Lucknow 226031, U.P., India
| | - Prabhat Ranjan Mishra
- Division of Pharmaceutics and Pharmacokinetics, CSIR-Central Drug Research Institute, Lucknow 226031, India
- Academy of Scientific and Innovation Research (AcSIR), Ghaziabad 201002, U.P., India
| |
Collapse
|
126
|
Yang Q, Meng X, Chen J, Li X, Huang Y, Xiao X, Li R, Wu X. RPLP2 activates TLR4 in an autocrine manner and promotes HIF-1α-induced metabolic reprogramming in hepatocellular carcinoma. Cell Death Discov 2023; 9:440. [PMID: 38052785 DOI: 10.1038/s41420-023-01719-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2023] [Revised: 11/04/2023] [Accepted: 11/13/2023] [Indexed: 12/07/2023] Open
Abstract
Metabolic reprogramming is a major feature of cancer, and aerobic glycolysis is one of the most widely studied metabolic reprogramming processes. Acidic ribosome protein P2 (RPLP2) is associated with both tumorigenesis and endoplasmic reticulum stress. However, limited knowledge exists regarding the role of RPLP2 in hepatocellular carcinoma (HCC) progression. In the present study, we observed a significant upregulation of RPLP2 in HCC tissues. Moreover, RPLP2 expression is closely correlated with patient prognosis and survival. The subsequent experimental validation demonstrated that RPLP2 exerted a regulatory effect on the expression of glycolytic enzymes and lactate production, thereby facilitating HCC cell proliferation. Mechanistically, the PI3K/AKT signalling pathway was found to play an important role in the regulation of hypoxia-inducible factor-1α (HIF-1α)-mediated aerobic glycolysis and cell growth. RPLP2 activates TLR4 on the surface of HCC cells and the downstream PI3K/AKT pathway through autocrine signalling. This activation then facilitates the entry of HIF-1α into the nucleus, enabling it to fulfil its transcriptional function. In conclusion, our findings suggested that RPLP2 induces a metabolic shift towards aerobic glycolysis and facilitates the progression of HCC through TLR4-dependent activation of the PI3K/AKT/HIF-1α pathway. Our study revealed the novel mechanism by which the ribosomal protein RPLP2 regulates glycolysis to promote HCC progression. These findings may offer a potential therapeutic target for HCC treatment.
Collapse
Affiliation(s)
- Qingqing Yang
- Department of Gastroenterology, The Yancheng Clinical College of Xuzhou Medical University, 224006, Yancheng, Jiangsu, China
| | - Xiangrui Meng
- Yancheng Medical Research Center of Nanjing University Medical School, Yancheng First Hospital, Affiliated Hospital of Nanjing University Medical School, The First People's Hospital of Yancheng, 224006, Yancheng, Jiangsu, China
| | - Jin Chen
- Department of Gastroenterology, The Yancheng Clinical College of Xuzhou Medical University, 224006, Yancheng, Jiangsu, China
| | - Xiangsu Li
- Department of Gastroenterology, The Yancheng Clinical College of Xuzhou Medical University, 224006, Yancheng, Jiangsu, China
| | - Yang Huang
- Department of Gastroenterology, The Yancheng Clinical College of Xuzhou Medical University, 224006, Yancheng, Jiangsu, China
| | - Xueyi Xiao
- Department of Gastroenterology, The Yancheng Clinical College of Xuzhou Medical University, 224006, Yancheng, Jiangsu, China
| | - Rongqing Li
- Department of Medical Genetics and Prenatal Diagnosis, The Affiliated Taizhou People's Hospital of Nanjing Medical University, 225399, Taizhou, Jiangsu, China.
| | - Xudong Wu
- Department of Gastroenterology, The Yancheng Clinical College of Xuzhou Medical University, 224006, Yancheng, Jiangsu, China.
| |
Collapse
|
127
|
Abstract
Cancers undergo sequential changes to proton (H+) concentration and sensing that are consequences of the disease and facilitate its further progression. The impact of protonation state on protein activity can arise from alterations to amino acids or their titration. Indeed, many cancer-initiating mutations influence pH balance, regulation or sensing in a manner that enables growth and invasion outside normal constraints as part of oncogenic transformation. These cancer-supporting effects become more prominent when tumours develop an acidic microenvironment owing to metabolic reprogramming and disordered perfusion. The ensuing intracellular and extracellular pH disturbances affect multiple aspects of tumour biology, ranging from proliferation to immune surveillance, and can even facilitate further mutagenesis. As a selection pressure, extracellular acidosis accelerates disease progression by favouring acid-resistant cancer cells, which are typically associated with aggressive phenotypes. Although acid-base disturbances in tumours often occur alongside hypoxia and lactate accumulation, there is now ample evidence for a distinct role of H+-operated responses in key events underpinning cancer. The breadth of these actions presents therapeutic opportunities to change the trajectory of disease.
Collapse
Affiliation(s)
- Pawel Swietach
- Department of Physiology, Anatomy and Genetics, University of Oxford, Oxford, UK.
| | - Ebbe Boedtkjer
- Department of Biomedicine, Aarhus University, Aarhus, Denmark.
| | - Stine Falsig Pedersen
- Department of Biology, University of Copenhagen, University of Copenhagen, Faculty of Science, København, Denmark.
| |
Collapse
|
128
|
Wang M, He T, Meng D, Lv W, Ye J, Cheng L, Hu J. BZW2 Modulates Lung Adenocarcinoma Progression through Glycolysis-Mediated IDH3G Lactylation Modification. J Proteome Res 2023; 22:3854-3865. [PMID: 37955350 DOI: 10.1021/acs.jproteome.3c00518] [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] [Indexed: 11/14/2023]
Abstract
Histone lactylation (Hla) is a metabolically stress-related histone modification that featured in specific gene expression regulation. However, the role of Hla in the pathogenesis of lung adenocarcinoma (LUAD) remains unexplored. Through bioinformatics analysis, we found that BZW2 exhibited an elevated level of expression in LUAD tissues, which was associated with a poor prognosis. Flow cytometry and TUNEL assay were used to analyze the apoptosis of LUAD cells and tissues, respectively. The effect of the cell function experiment on the LUAD cell phenotype was analyzed. An XF 96 Extracellular Flux Analyzer measured the ECAR value, and kits were used to detect lactate production and glucose consumption. Animal experiments were performed for further verification. Cell experiments showed that BZW2 fostered the malignant progression of LUAD by promoting glycolysis-mediated lactate production and lactylation of IDH3G. In a compelling in vivo validation, the inhibition of Hla could suppress the malignant progression of LUAD. Knockdown of BZW2 combined with 2-DG treatment significantly repressed tumor growth in mice. BZW2 could regulate the progression of LUAD through glycolysis-mediated IDH3G lactylation, offering a theoretical basis for the targeted treatment of LUAD with glycolysis and Hla.
Collapse
Affiliation(s)
- Ming Wang
- Zhejiang University, Hangzhou 310058, China
- Department of Thoracic Surgery, Shulan (Hangzhou) Hospital, Hangzhou 310000, China
- Zhejiang Shuren University, Hangzhou 310015, China
| | - Tianyu He
- Department of Thoracic Surgery, The First Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou 310000, China
| | - Di Meng
- Department of Thoracic Surgery, The First Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou 310000, China
| | - Wang Lv
- Department of Thoracic Surgery, The First Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou 310000, China
| | - Jiayue Ye
- Department of Thoracic Surgery, The First Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou 310000, China
| | - Ling Cheng
- Academician Expert Workstation of Zhejiang Luo Xi Medical Technology Co., Ltd, Shaoxing City 312030, China
| | - Jian Hu
- Department of Thoracic Surgery, The First Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou 310000, China
- Key Laboratory of Clinical Evaluation Technology for Medical Device of Zhejiang Province, Hangzhou 310000, China
| |
Collapse
|
129
|
Patrucco D, Cutrin JC, Longo DL, Botto E, Cong L, Aime S, Delli Castelli D. In Situ Insonation of Alkaline Buffer Containing Liposomes Leads to a Net Improvement of the Therapeutic Outcome in a Triple Negative Breast Cancer Murine Model. Adv Healthc Mater 2023; 12:e2301480. [PMID: 37709294 DOI: 10.1002/adhm.202301480] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2023] [Revised: 09/12/2023] [Indexed: 09/16/2023]
Abstract
Breast cancer is characterized by an acidic micro-environment. Acidic extracellular pH gives cancer cells an evolutionary advantage, hence, neutralization of the extracellular pH has been considered as a potential therapeutic strategy. To address the issue of systemic pH alteration, an approach based on the targeted delivery of the buffering solution to the tumor region is investigated. The method relies on the use of low frequency ultrasound and sono-sensitive liposomes loaded with buffers at alkaline pH (LipHUS). After the i.v. injection of LipHUS, the application of ultrasound (US) at the sites of the pathology induces a local increase of pH that results highly effective in i) inhibiting primary tumor growth, ii) reducing tumor recurrence after surgery, and iii) suppressing metastases' formation. The experiments are carried out on a triple negative breast cancer mouse model. The results obtained demonstrate that localized and triggered release of bicarbonate or PBS buffer from sonosensitive liposomes represents an efficient therapeutic tool for treating triple-negative breast cancer. This approach holds promise for potential clinical translation.
Collapse
Affiliation(s)
- Deyssy Patrucco
- Department of Molecular Biotechnology and Health Science, University of Turin, Via Nizza 52, Turin, 10126, Italy
| | - Juan Carlos Cutrin
- Department of Molecular Biotechnology and Health Science, University of Turin, Via Nizza 52, Turin, 10126, Italy
| | - Dario Livio Longo
- Istituto di Biostrutture e Bioimmagini (IBB), Consiglio Nazionale delle Ricerche (CNR), Via Tommaso De Amicis, 95, Naples, 80145, Italy
| | - Elena Botto
- Istituto di Biostrutture e Bioimmagini (IBB), Consiglio Nazionale delle Ricerche (CNR), Via Tommaso De Amicis, 95, Naples, 80145, Italy
| | - Li Cong
- Key Laboratory of Smart Drug Delivery, Ministry of Education, School of Pharmacy, Fudan University, Shanghai, 201203, China
| | - Silvio Aime
- IRCCS SDN, SYNLAB, Via Gianturco 113, Naples, 80143, Italy
| | - Daniela Delli Castelli
- Department of Molecular Biotechnology and Health Science, University of Turin, Via Nizza 52, Turin, 10126, Italy
| |
Collapse
|
130
|
Yu M, Pan Q, Li W, Du T, Huang F, Wu H, He Y, Wu X, Shi H. Isoliquiritigenin inhibits gastric cancer growth through suppressing GLUT4 mediated glucose uptake and inducing PDHK1/PGC-1α mediated energy metabolic collapse. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2023; 121:155045. [PMID: 37742526 DOI: 10.1016/j.phymed.2023.155045] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/05/2023] [Revised: 08/12/2023] [Accepted: 08/24/2023] [Indexed: 09/26/2023]
Abstract
BACKGROUND Isoliquiritigenin (ISL), a natural flavonoid, has anti-tumor activity. But, the understanding of the impact and molecular mechanism of ISL on the growth of gastric cancer (GC) remains limited. PURPOSE The study was to explore the tumor suppressive effect of ISL on GC growth both in vitro and in vivo, meanwhile, clarify its molecular mechanisms. METHODS Cell viability was detected by cell counting kit-8 (CCK-8) assay. Apoptotic cells in vitro were monitored by Hoechst 33,342 solution. Protein expression was assessed by Western blot. Reactive oxygen species (ROS) level was evaluated by utilizing 2',7'- dichlorofluorescin diacetate (DCFH-DA). Lactic acid level was detected with L-lactate assay kit. Glucose uptake was monitored with fluorescently tagged glucose 2-[N-(7-nitrobenz-2-oxa-1,3-diaxol-4-yl)amino]-2-deoxyglucose (2-NBDG). Glycolytic proton efflux rate (GlycoPER) was evaluated by glycolytic rate assay kit. Oxygen consumption rate (OCR) was conducted by mito stress test kit. A nude mouse model of gastric cancer cell xenograft was established by subcutaneous injection with MGC803 cells. Pathological changes were evaluated by using H&E staining. Cell apoptosis in vivo was evaluated by terminal deoxy-nucleotide transferase mediated dUTP nick end labeling (TUNEL) assay. RESULTS ISL remarkably suppressed GC growth and increased cell apoptosis. It regulated apoptosis-related and metabolism-related protein expression both in vitro and in vivo. ISL blocked glucose uptake and suppressed production and secretion of lactic acid, which was accompanied with suppressed mitochondrial oxidative phosphorylation (OXPHOS) and glycolysis but increased ROS accumulation. Overexpression of peroxisome proliferator-activated receptor gamma coactivator-1 alpha (PGC-1α), cellular-myelocytomatosis viral oncogene (c-Myc), hypoxia inducible factor-1α (HIF-1α), glucose transporter 4 (GLUT4) or pyruvate dehydrogenase kinase 1 (PDHK1), could abolish ISL-induced inhibition of cell viability in GC cells. CONCLUSION These findings implicated that ISL inhibits GC growth by decreasing GLUT4 mediated glucose uptake and inducing PDHK1/PGC-1α-mediated energy metabolic collapse through depressing protein expression of c-Myc and HIF-1α in GC, suggesting its potential application for GC treatment.
Collapse
Affiliation(s)
- Mingzhu Yu
- Shanghai Key Laboratory of Compound Chinese Medicines, the Ministry of Education (MOE) Key Laboratory for Standardization of Chinese Medicines, the SATCM Key Laboratory for New Resources & Quality Evaluation of Chinese Medicine, Research Center of Shanghai Traditional Chinese Medicine Standardization, Institute of Chinese Materia Medica, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
| | - Qiaoling Pan
- Shanghai Key Laboratory of Compound Chinese Medicines, the Ministry of Education (MOE) Key Laboratory for Standardization of Chinese Medicines, the SATCM Key Laboratory for New Resources & Quality Evaluation of Chinese Medicine, Research Center of Shanghai Traditional Chinese Medicine Standardization, Institute of Chinese Materia Medica, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
| | - Wenbiao Li
- Shanghai Key Laboratory of Compound Chinese Medicines, the Ministry of Education (MOE) Key Laboratory for Standardization of Chinese Medicines, the SATCM Key Laboratory for New Resources & Quality Evaluation of Chinese Medicine, Research Center of Shanghai Traditional Chinese Medicine Standardization, Institute of Chinese Materia Medica, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
| | - Tingting Du
- Shanghai Key Laboratory of Compound Chinese Medicines, the Ministry of Education (MOE) Key Laboratory for Standardization of Chinese Medicines, the SATCM Key Laboratory for New Resources & Quality Evaluation of Chinese Medicine, Research Center of Shanghai Traditional Chinese Medicine Standardization, Institute of Chinese Materia Medica, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
| | - Fei Huang
- Shanghai Key Laboratory of Compound Chinese Medicines, the Ministry of Education (MOE) Key Laboratory for Standardization of Chinese Medicines, the SATCM Key Laboratory for New Resources & Quality Evaluation of Chinese Medicine, Research Center of Shanghai Traditional Chinese Medicine Standardization, Institute of Chinese Materia Medica, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
| | - Hui Wu
- Shanghai Key Laboratory of Compound Chinese Medicines, the Ministry of Education (MOE) Key Laboratory for Standardization of Chinese Medicines, the SATCM Key Laboratory for New Resources & Quality Evaluation of Chinese Medicine, Research Center of Shanghai Traditional Chinese Medicine Standardization, Institute of Chinese Materia Medica, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
| | - Yixin He
- Shanghai Key Laboratory of Compound Chinese Medicines, the Ministry of Education (MOE) Key Laboratory for Standardization of Chinese Medicines, the SATCM Key Laboratory for New Resources & Quality Evaluation of Chinese Medicine, Research Center of Shanghai Traditional Chinese Medicine Standardization, Institute of Chinese Materia Medica, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
| | - Xiaojun Wu
- Shanghai Key Laboratory of Compound Chinese Medicines, the Ministry of Education (MOE) Key Laboratory for Standardization of Chinese Medicines, the SATCM Key Laboratory for New Resources & Quality Evaluation of Chinese Medicine, Research Center of Shanghai Traditional Chinese Medicine Standardization, Institute of Chinese Materia Medica, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China.
| | - Hailian Shi
- Shanghai Key Laboratory of Compound Chinese Medicines, the Ministry of Education (MOE) Key Laboratory for Standardization of Chinese Medicines, the SATCM Key Laboratory for New Resources & Quality Evaluation of Chinese Medicine, Research Center of Shanghai Traditional Chinese Medicine Standardization, Institute of Chinese Materia Medica, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China.
| |
Collapse
|
131
|
Xu F, Yu F. Sensing and regulation of plant extracellular pH. TRENDS IN PLANT SCIENCE 2023; 28:1422-1437. [PMID: 37596188 DOI: 10.1016/j.tplants.2023.06.015] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/03/2023] [Revised: 06/03/2023] [Accepted: 06/19/2023] [Indexed: 08/20/2023]
Abstract
In plants, pH determines nutrient acquisition and sensing, and triggers responses to osmotic stress, whereas pH homeostasis protects the cellular machinery. Extracellular pH (pHe) controls the chemistry and rheology of the cell wall to adjust its elasticity and regulate cell expansion in space and time. Plasma membrane (PM)-localized proton pumps, cell-wall components, and cell wall-remodeling enzymes jointly maintain pHe homeostasis. To adapt to their environment and modulate growth and development, plant cells must sense subtle changes in pHe caused by the environment or neighboring cells. Accumulating evidence indicates that PM-localized cell-surface peptide-receptor pairs sense pHe. We highlight recent advances in understanding how plants perceive and maintain pHe, and discuss future perspectives.
Collapse
Affiliation(s)
- Fan Xu
- State Key Laboratory of Chemo/Biosensing and Chemometrics, College of Biology, and Hunan Key Laboratory of Plant Functional Genomics and Developmental Regulation, Hunan University, Changsha 410082, PR China
| | - Feng Yu
- State Key Laboratory of Chemo/Biosensing and Chemometrics, College of Biology, and Hunan Key Laboratory of Plant Functional Genomics and Developmental Regulation, Hunan University, Changsha 410082, PR China.
| |
Collapse
|
132
|
Fu L, Qi C, Sun T, Huang K, Lin J, Huang P. Glucose oxidase-instructed biomineralization of calcium-based biomaterials for biomedical applications. EXPLORATION (BEIJING, CHINA) 2023; 3:20210110. [PMID: 38264686 PMCID: PMC10742215 DOI: 10.1002/exp.20210110] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 09/06/2022] [Accepted: 02/22/2023] [Indexed: 01/25/2024]
Abstract
In recent years, glucose oxidase (GOx) has aroused great research interest in the treatment of diseases related to abnormal glucose metabolisms like cancer and diabetes. However, as a kind of endogenous oxido-reductase, GOx suffers from poor stability and system toxicity in vivo. In order to overcome this bottleneck, GOx is encapsulated in calcium-based biomaterials (CaXs) such as calcium phosphate (CaP) and calcium carbonate (CaCO3) by using it as a biotemplate to simulate the natural biomineralization process. The biomineralized GOx holds improved stability and reduced side effects, due to the excellent bioactivity, biocompatibitliy, and biodegradability of CaXs. In this review, the state-of-the-art studies on GOx-mineralized CaXs are introduced with an emphasis on their application in various biomedical fields including disease diagnosis, cancer treatment, and diabetes management. The current challenges and future perspectives of GOx-mineralized CaXs are discussed, which is expected to promote further studies on these smart GOx-mineralized CaXs biomaterials for practical applications.
Collapse
Affiliation(s)
- Lian‐Hua Fu
- Marshall Laboratory of Biomedical EngineeringInternational Cancer Center, Laboratory of Evolutionary Theranostics (LET)School of Biomedical EngineeringShenzhen University Medical SchoolShenzhen UniversityShenzhenChina
| | - Chao Qi
- Marshall Laboratory of Biomedical EngineeringInternational Cancer Center, Laboratory of Evolutionary Theranostics (LET)School of Biomedical EngineeringShenzhen University Medical SchoolShenzhen UniversityShenzhenChina
| | - Tuanwei Sun
- Marshall Laboratory of Biomedical EngineeringInternational Cancer Center, Laboratory of Evolutionary Theranostics (LET)School of Biomedical EngineeringShenzhen University Medical SchoolShenzhen UniversityShenzhenChina
| | - Kai Huang
- Department of Materials Science and EngineeringUniversity of TorontoTorontoOntarioCanada
| | - Jing Lin
- Marshall Laboratory of Biomedical EngineeringInternational Cancer Center, Laboratory of Evolutionary Theranostics (LET)School of Biomedical EngineeringShenzhen University Medical SchoolShenzhen UniversityShenzhenChina
| | - Peng Huang
- Marshall Laboratory of Biomedical EngineeringInternational Cancer Center, Laboratory of Evolutionary Theranostics (LET)School of Biomedical EngineeringShenzhen University Medical SchoolShenzhen UniversityShenzhenChina
| |
Collapse
|
133
|
Henn JG, Bernardes Ferro M, Lopes Alves GA, Pires Peña F, de Oliveira JVR, de Souza BM, da Silva LF, Rapack Jacinto Silva V, Silva Pinheiro AC, Steffens Reinhardt L, Morás AM, Nugent M, da Rosa RG, Silveira Aguirre TA, Moura DJ. Development and characterization of a temozolomide-loaded nanoemulsion and the effect of ferrocene pre and co-treatments in glioblastoma cell models. Pharmacol Rep 2023; 75:1597-1609. [PMID: 37837521 DOI: 10.1007/s43440-023-00537-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2023] [Revised: 09/25/2023] [Accepted: 09/26/2023] [Indexed: 10/16/2023]
Abstract
BACKGROUND Glioblastoma is a severe brain tumor that requires aggressive treatment involving surgery, radiotherapy, and chemotherapy, offering a survival rate of only 15 months. Fortunately, recent nanotechnology progress has enabled novel approaches and, alongside ferrocenes' unique properties of cytotoxicity, sensitization, and interaction with reactive oxygen species, have brought new possibilities to complement chemotherapy in nanocarrier systems, enhancing treatment results. METHODS In this work, we developed and characterized a temozolomide-loaded nanoemulsion and evaluated its cytotoxic potential in combination with ferrocene in the temozolomide-resistant T98G and temozolomide-sensitive U87 cell lines. The effects of the treatments were assessed through acute assays of cell viability, cell death, mitochondrial alterations, and a treatment protocol simulation based on different two-cycle regimens. RESULTS Temozolomide nanoemulsion showed a z-average diameter of 173.37 ± 0.86 nm and a zeta potential of - 6.53 ± 1.13 mV. Physicochemical characterization revealed that temozolomide is probably associated with nanoemulsion droplets instead of being entrapped within the nanostructure, allowing a rapid drug release. In combination with ferrocene, temozolomide nanoemulsion reduced glioblastoma cell viability in both acute and two-cycle regimen assays. The combined treatment approach also reversed T98G's temozolomide-resistant profile by altering the mitochondrial membrane potential of the cells, thus increasing reactive oxygen species generation, and ultimately inducing cell death. CONCLUSIONS Altogether, our results indicate that using nanoemulsion containing temozolomide in combination with ferrocene is an effective approach to improve glioblastoma therapy outcomes.
Collapse
Affiliation(s)
- Jeferson Gustavo Henn
- Laboratório de Genética Toxicológica, Universidade Federal de Ciências da Saúde de Porto Alegre, 245 Sarmento Leite Street, Lab. 714, Porto Alegre, Rio Grande do Sul, 90050-170, Brazil
- Materials Research Institute, Technological University of the Shannon: Midlands Midwest, Athlone, Co. Westmeath, N37HD68, Ireland
| | - Matheus Bernardes Ferro
- Laboratório de Genética Toxicológica, Universidade Federal de Ciências da Saúde de Porto Alegre, 245 Sarmento Leite Street, Lab. 714, Porto Alegre, Rio Grande do Sul, 90050-170, Brazil
| | - Gabriel Antonio Lopes Alves
- Laboratório de Farmacociências, Universidade Federal de Ciências da Saúde de Porto Alegre, 245 Sarmento Leite Street, Porto Alegre, Rio Grande do Sul, 90050-170, Brazil
| | - Flávia Pires Peña
- Laboratório de Farmacociências, Universidade Federal de Ciências da Saúde de Porto Alegre, 245 Sarmento Leite Street, Porto Alegre, Rio Grande do Sul, 90050-170, Brazil
| | - João Vitor Raupp de Oliveira
- Laboratório de Farmacociências, Universidade Federal de Ciências da Saúde de Porto Alegre, 245 Sarmento Leite Street, Porto Alegre, Rio Grande do Sul, 90050-170, Brazil
| | - Bárbara Müller de Souza
- Departamento de Química Inorgânica, Universidade Federal do Rio Grande do Sul, 9500 Bento Gonçalves Avenue, Porto Alegre, Rio Grande do Sul, 91501-970, Brazil
| | - Leonardo Fonseca da Silva
- Departamento de Química Inorgânica, Universidade Federal do Rio Grande do Sul, 9500 Bento Gonçalves Avenue, Porto Alegre, Rio Grande do Sul, 91501-970, Brazil
| | - Victória Rapack Jacinto Silva
- Laboratório de Genética Toxicológica, Universidade Federal de Ciências da Saúde de Porto Alegre, 245 Sarmento Leite Street, Lab. 714, Porto Alegre, Rio Grande do Sul, 90050-170, Brazil
| | - Ana Carolina Silva Pinheiro
- Laboratório de Genética Toxicológica, Universidade Federal de Ciências da Saúde de Porto Alegre, 245 Sarmento Leite Street, Lab. 714, Porto Alegre, Rio Grande do Sul, 90050-170, Brazil
| | - Luiza Steffens Reinhardt
- Laboratório de Genética Toxicológica, Universidade Federal de Ciências da Saúde de Porto Alegre, 245 Sarmento Leite Street, Lab. 714, Porto Alegre, Rio Grande do Sul, 90050-170, Brazil
| | - Ana Moira Morás
- Laboratório de Genética Toxicológica, Universidade Federal de Ciências da Saúde de Porto Alegre, 245 Sarmento Leite Street, Lab. 714, Porto Alegre, Rio Grande do Sul, 90050-170, Brazil
| | - Michael Nugent
- Materials Research Institute, Technological University of the Shannon: Midlands Midwest, Athlone, Co. Westmeath, N37HD68, Ireland
| | - Ricardo Gomes da Rosa
- Departamento de Química Inorgânica, Universidade Federal do Rio Grande do Sul, 9500 Bento Gonçalves Avenue, Porto Alegre, Rio Grande do Sul, 91501-970, Brazil
| | - Tanira Alessandra Silveira Aguirre
- Laboratório de Farmacociências, Universidade Federal de Ciências da Saúde de Porto Alegre, 245 Sarmento Leite Street, Porto Alegre, Rio Grande do Sul, 90050-170, Brazil
| | - Dinara Jaqueline Moura
- Laboratório de Genética Toxicológica, Universidade Federal de Ciências da Saúde de Porto Alegre, 245 Sarmento Leite Street, Lab. 714, Porto Alegre, Rio Grande do Sul, 90050-170, Brazil.
| |
Collapse
|
134
|
Hunt H, Leape S, Sidhu JS, Ajmera I, Lynch JP, Ratcliffe RG, Sweetlove LJ. A role for fermentation in aerobic conditions as revealed by computational analysis of maize root metabolism during growth by cell elongation. THE PLANT JOURNAL : FOR CELL AND MOLECULAR BIOLOGY 2023; 116:1553-1570. [PMID: 37831626 DOI: 10.1111/tpj.16478] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/31/2023] [Revised: 09/07/2023] [Accepted: 09/11/2023] [Indexed: 10/15/2023]
Abstract
The root is a well-studied example of cell specialisation, yet little is known about the metabolism that supports the transport functions and growth of different root cell types. To address this, we used computational modelling to study metabolism in the elongation zone of a maize lateral root. A functional-structural model captured the cell-anatomical features of the root and modelled how they changed as the root elongated. From these data, we derived constraints for a flux balance analysis model that predicted metabolic fluxes of the 11 concentric rings of cells in the root. We discovered a distinct metabolic flux pattern in the cortical cell rings, endodermis and pericycle (but absent in the epidermis) that involved a high rate of glycolysis and production of the fermentation end-products lactate and ethanol. This aerobic fermentation was confirmed experimentally by metabolite analysis. The use of fermentation in the model was not obligatory but was the most efficient way to meet the specific demands for energy, reducing power and carbon skeletons of expanding cells. Cytosolic acidification was avoided in the fermentative mode due to the substantial consumption of protons by lipid synthesis. These results expand our understanding of fermentative metabolism beyond that of hypoxic niches and suggest that fermentation could play an important role in the metabolism of aerobic tissues.
Collapse
Affiliation(s)
- Hilary Hunt
- Department of Biology, University of Oxford, South Parks Road, Oxford, OX1 3RB, UK
| | - Stefan Leape
- Department of Biology, University of Oxford, South Parks Road, Oxford, OX1 3RB, UK
| | - Jagdeep Singh Sidhu
- Department of Plant Science, The Pennsylvania State University, University Park, Pennsylvania, 16802, USA
| | - Ishan Ajmera
- Department of Plant Science, The Pennsylvania State University, University Park, Pennsylvania, 16802, USA
| | - Jonathan P Lynch
- Department of Plant Science, The Pennsylvania State University, University Park, Pennsylvania, 16802, USA
| | - R George Ratcliffe
- Department of Biology, University of Oxford, South Parks Road, Oxford, OX1 3RB, UK
| | - Lee J Sweetlove
- Department of Biology, University of Oxford, South Parks Road, Oxford, OX1 3RB, UK
| |
Collapse
|
135
|
Song S, Xie S, Liu X, Li S, Wang L, Jiang X, Lu D. miR-3200 accelerates the growth of liver cancer cells by enhancing Rab7A. Noncoding RNA Res 2023; 8:675-685. [PMID: 37860266 PMCID: PMC10582768 DOI: 10.1016/j.ncrna.2023.10.005] [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: 07/11/2023] [Revised: 10/07/2023] [Accepted: 10/07/2023] [Indexed: 10/21/2023] Open
Abstract
Researches indicate miR-3200 is closely related to tumorigenesis, However, the role of miR-3200 in human hepatocarcinogenesis is still unclear. In this study, we clearly demonstrate that miR-3200 accelerates the growth of liver cancer cells in vivo and in vitro. Obviously, these findings are noteworthy that miR-3200 affects the transcriptional regulation for several genes, including DSP,BABAM2, Rab7A,SQSTM1,PRKAG2,CDK1,ABCE1,BECN1,PTEN,UPRT. And miR-3200 affects the transcriptional ability of several genes, such as, upregulating CADPS, DSP,FBXO32, PPCA,SGK1, PATXN7L1, PLK2,ITGB5,FZD3,HOXC8,HSPA1A,C-Myc,CyclnD1,CyclinE,PCNA and down -regulating SUV39H1, MYO1G, OLFML3, CBX5, PPDE2A, HOXA7, RAD54L, CDC45,SHMT7,MAD2L1,P27,IQGAP3,PTEN,P57,SCAMP3,etc...On the other hand, it is obvious that miR-3200 affects the translational ability of several genes, such as, upregulating GNS,UPRT,EIFAD,YOS1,SGK1,K-Ras,PKM2,C-myc,Pim1,CyclinD1,mTOR,erbB-2,CyclinE,PCNA,RRAS,ARAF,RAPH1,etc.. and down-regulating KDM2A, AATF, TMM17B, RAB8B, MYO1G,P21WAF1/Cip1,GADD45,PTEN,P27,P18,P57,SERBP1,RPL34,UFD1,Bax,ANXA6,GSK3β. Strikingly, miR-3200 affects some signaling pathway in liver cancer, including carbon metabolism signaling pathway, DNA replication pathway, FoxO signaling pathway, Hippo signaling pathway, serine and threonine metabolism signaling pathway, mTOR signaling pathway, Fatty acid biosynthesis signaling pathway, carcinogenesis-receptor activation signaling pathway, autophagy signaling pathway. Furthermore, our results suggest that miR-3200 enhances expression of RAB7A, and then Rab7A regulates the carcinogenic function of miR-3200 by increasing telomere remodeling in human liver cancer. These results are of great significance for the prevention and treatment of human liver cancer.
Collapse
Affiliation(s)
| | | | | | - Shujie Li
- Shanghai Putuo People's Hospital, School of Life Science and Technology, Tongji University, Shanghai, 200092, China
| | - Liyan Wang
- Shanghai Putuo People's Hospital, School of Life Science and Technology, Tongji University, Shanghai, 200092, China
| | - Xiaoxue Jiang
- Shanghai Putuo People's Hospital, School of Life Science and Technology, Tongji University, Shanghai, 200092, China
| | - Dongdong Lu
- Shanghai Putuo People's Hospital, School of Life Science and Technology, Tongji University, Shanghai, 200092, China
| |
Collapse
|
136
|
Huang PC, Chang CW, Lin YC, Chen CY, Chen TY, Chuang LT, Liu CJ, Huang CL, Li WC. Pyruvate Kinase Differentially Alters Metabolic Signatures during Head and Neck Carcinogenesis. Int J Mol Sci 2023; 24:16639. [PMID: 38068962 PMCID: PMC10706023 DOI: 10.3390/ijms242316639] [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: 10/16/2023] [Revised: 11/13/2023] [Accepted: 11/16/2023] [Indexed: 12/18/2023] Open
Abstract
During glycolysis, the muscle isoform of pyruvate kinase PKM2 produces ATP in exchange for dephosphorylation of phosphoenolpyruvate (PEP) into pyruvate. PKM2 has been considered as a tumor-promoting factor in most cancers, whereas the regulatory role of PKM2 during head and neck carcinogenesis remained to be delineated. PKM2 mRNA and protein expression was examined in head and neck tumorous specimens. The role of PKM2 in controlling cellular malignancy was determined in shRNA-mediated PKM2-deficient head and neck squamous cell carcinoma (HNSC) cells. In agreement with the results in other cancers, PKM2 expression is enriched in both mouse and human HNSC tissues. Nevertheless, PKM2 mRNA expression reversely correlated with tumor stage, and greater recurrence-free survival rates are evident in the PKM2high HNSC population, arguing that PKM2 may be tumor-suppressive. Multifaceted analyses showed a greater in vivo xenografic tumor growth and an enhanced cisplatin resistance in response to PKM2 loss, whereas PKM2 silencing led to reduced cell motility. At the molecular level, metabolic shifts towards mitochondrial metabolism and activation of oncogenic Protein kinase B (PKB/Akt) and extracellular signal-regulated kinase (ERK) signals were detected in PKM2-silencing HNSC cells. In sum, our findings demonstrated that PKM2 differentially modulated head and neck tumorigenicity via metabolic reprogramming.
Collapse
Affiliation(s)
- Pei-Chun Huang
- Institute of Oral Biology, College of Dentistry, National Yang Ming Chiao Tung University, Taipei 11221, Taiwan; (P.-C.H.); (C.-Y.C.); (T.-Y.C.)
| | - Ching-Wen Chang
- Graduate Institute of Metabolism and Obesity Sciences (GIMOS), College of Nutrition, Taipei Medical University, Taipei 11031, Taiwan;
- Taipei Cancer Center, Taipei Medical University, Taipei 11031, Taiwan
| | - Yu-Cheng Lin
- Department of Dentistry, College of Dentistry, National Yang Ming Chiao Tung University, Taipei 11221, Taiwan; (Y.-C.L.); (C.-J.L.)
- Oral Medicine Innovation Center (OMIC), National Yang Ming Chiao Tung University, Taipei 11221, Taiwan
| | - Chang-Yi Chen
- Institute of Oral Biology, College of Dentistry, National Yang Ming Chiao Tung University, Taipei 11221, Taiwan; (P.-C.H.); (C.-Y.C.); (T.-Y.C.)
| | - Tsai-Ying Chen
- Institute of Oral Biology, College of Dentistry, National Yang Ming Chiao Tung University, Taipei 11221, Taiwan; (P.-C.H.); (C.-Y.C.); (T.-Y.C.)
| | - Lu-Te Chuang
- Department of Biotechnology and Pharmaceutical Technology, Yuanpei University of Medical Technology, Hsinchu 30015, Taiwan;
| | - Chung-Ji Liu
- Department of Dentistry, College of Dentistry, National Yang Ming Chiao Tung University, Taipei 11221, Taiwan; (Y.-C.L.); (C.-J.L.)
- Department of Oral and Maxillofacial Surgery, MacKay Memorial Hospital, Taipei 10449, Taiwan
- Department of Medical Research, MacKay Memorial Hospital, Taipei 10449, Taiwan
| | - Chien-Ling Huang
- Department of Health Technology and Informatics (HTI), The Hong Kong Polytechnic University (PolyU), Hung Hom, Kowloon, Hong Kong SAR, China;
| | - Wan-Chun Li
- Institute of Oral Biology, College of Dentistry, National Yang Ming Chiao Tung University, Taipei 11221, Taiwan; (P.-C.H.); (C.-Y.C.); (T.-Y.C.)
- Department of Dentistry, College of Dentistry, National Yang Ming Chiao Tung University, Taipei 11221, Taiwan; (Y.-C.L.); (C.-J.L.)
- Oral Medicine Innovation Center (OMIC), National Yang Ming Chiao Tung University, Taipei 11221, Taiwan
| |
Collapse
|
137
|
Sun X, Sui W, Mu Z, Xie S, Deng J, Li S, Seki T, Wu J, Jing X, He X, Wang Y, Li X, Yang Y, Huang P, Ge M, Cao Y. Mirabegron displays anticancer effects by globally browning adipose tissues. Nat Commun 2023; 14:7610. [PMID: 37993438 PMCID: PMC10665320 DOI: 10.1038/s41467-023-43350-8] [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: 03/29/2023] [Accepted: 11/07/2023] [Indexed: 11/24/2023] Open
Abstract
Metabolic reprogramming in malignant cells is a hallmark of cancer that relies on augmented glycolytic metabolism to support their growth, invasion, and metastasis. However, the impact of global adipose metabolism on tumor growth and the drug development by targeting adipose metabolism remain largely unexplored. Here we show that a therapeutic paradigm of drugs is effective for treating various cancer types by browning adipose tissues. Mirabegron, a clinically available drug for overactive bladders, displays potent anticancer effects in various animal cancer models, including untreatable cancers such as pancreatic ductal adenocarcinoma and hepatocellular carcinoma, via the browning of adipose tissues. Genetic deletion of the uncoupling protein 1, a key thermogenic protein in adipose tissues, ablates the anticancer effect. Similarly, the removal of brown adipose tissue, which is responsible for non-shivering thermogenesis, attenuates the anticancer activity of mirabegron. These findings demonstrate that mirabegron represents a paradigm of anticancer drugs with a distinct mechanism for the effective treatment of multiple cancers.
Collapse
Affiliation(s)
- Xiaoting Sun
- Oujiang Laboratory (Zhejiang Lab for Regenerative Medicine, Vison and Brain Health), School of Pharmaceutical Science, Wenzhou Medical University, Wenzhou, China
- Department of Microbiology, Tumor and Cell Biology, Karolinska Institutet, 171 65, Solna, Sweden
| | - Wenhai Sui
- National Key Laboratory for Innovation and Transformation of Luobing Theory National Key Laboratory for Innovation and Transformation of Luobing Theory; The Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education, Chinese National Health Commission and Chinese Academy of Medical Sciences, The State and Shandong Province Joint Key Laboratory of Translational Cardiovascular Medicine, Department of Cardiology, Qilu Hospital of Shandong University, 250012, Jinan, China
| | - Zepeng Mu
- Department of Endocrinology, Affiliated Hospital of Medical College Qingdao University, Qingdao, China
| | - Sisi Xie
- Department of Cellular and Genetic Medicine, School of Basic Medical Sciences, Fudan University, 200032, Shanghai, China
| | - Jinxiu Deng
- Department of Cellular and Genetic Medicine, School of Basic Medical Sciences, Fudan University, 200032, Shanghai, China
| | - Sen Li
- Department of Cellular and Genetic Medicine, School of Basic Medical Sciences, Fudan University, 200032, Shanghai, China
| | - Takahiro Seki
- Department of Microbiology, Tumor and Cell Biology, Karolinska Institutet, 171 65, Solna, Sweden
| | - Jieyu Wu
- Department of Microbiology, Tumor and Cell Biology, Karolinska Institutet, 171 65, Solna, Sweden
| | - Xu Jing
- Department of Microbiology, Tumor and Cell Biology, Karolinska Institutet, 171 65, Solna, Sweden
- Department of Head and Neck Surgery, Center of Otolaryngology-Head and Neck Surgery, Zhejiang Provincial People's Hospital, People's Hospital of Hangzhou Medical College, Hangzhou, China
| | - Xingkang He
- Department of Gastroenterology, Sir Run Run Shaw Hospital, Zhejiang University Medical School, Hangzhou, 310016, China
| | - Yangang Wang
- Department of Endocrinology, Affiliated Hospital of Medical College Qingdao University, Qingdao, China
| | - Xiaokun Li
- Oujiang Laboratory (Zhejiang Lab for Regenerative Medicine, Vison and Brain Health), School of Pharmaceutical Science, Wenzhou Medical University, Wenzhou, China
| | - Yunlong Yang
- Department of Cellular and Genetic Medicine, School of Basic Medical Sciences, Fudan University, 200032, Shanghai, China.
| | - Ping Huang
- Department of Pharmacy, Zhejiang Provincial People's Hospital, People's Hospital of Hangzhou Medical College, Hangzhou, China.
- Key Laboratory of Endocrine Gland Diseases of Zhejiang Province, Hangzhou, China.
| | - Minghua Ge
- Department of Head and Neck Surgery, Center of Otolaryngology-Head and Neck Surgery, Zhejiang Provincial People's Hospital, People's Hospital of Hangzhou Medical College, Hangzhou, China.
| | - Yihai Cao
- Department of Microbiology, Tumor and Cell Biology, Karolinska Institutet, 171 65, Solna, Sweden.
| |
Collapse
|
138
|
Nakahara R, Aki S, Sugaya M, Hirose H, Kato M, Maeda K, Sakamoto DM, Kojima Y, Nishida M, Ando R, Muramatsu M, Pan M, Tsuchida R, Matsumura Y, Yanai H, Takano H, Yao R, Sando S, Shibuya M, Sakai J, Kodama T, Kidoya H, Shimamura T, Osawa T. Hypoxia activates SREBP2 through Golgi disassembly in bone marrow-derived monocytes for enhanced tumor growth. EMBO J 2023; 42:e114032. [PMID: 37781951 PMCID: PMC10646561 DOI: 10.15252/embj.2023114032] [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: 03/15/2023] [Revised: 08/22/2023] [Accepted: 08/28/2023] [Indexed: 10/03/2023] Open
Abstract
Bone marrow-derived cells (BMDCs) infiltrate hypoxic tumors at a pre-angiogenic state and differentiate into mature macrophages, thereby inducing pro-tumorigenic immunity. A critical factor regulating this differentiation is activation of SREBP2-a well-known transcription factor participating in tumorigenesis progression-through unknown cellular mechanisms. Here, we show that hypoxia-induced Golgi disassembly and Golgi-ER fusion in monocytic myeloid cells result in nuclear translocation and activation of SREBP2 in a SCAP-independent manner. Notably, hypoxia-induced SREBP2 activation was only observed in an immature lineage of bone marrow-derived cells. Single-cell RNA-seq analysis revealed that SREBP2-mediated cholesterol biosynthesis was upregulated in HSCs and monocytes but not in macrophages in the hypoxic bone marrow niche. Moreover, inhibition of cholesterol biosynthesis impaired tumor growth through suppression of pro-tumorigenic immunity and angiogenesis. Thus, our findings indicate that Golgi-ER fusion regulates SREBP2-mediated metabolic alteration in lineage-specific BMDCs under hypoxia for tumor progression.
Collapse
Affiliation(s)
- Ryuichi Nakahara
- Division of Nutriomics and Oncology, RCASTThe University of TokyoTokyoJapan
- Department of Chemistry and Biotechnology, Graduate School of EngineeringThe University of TokyoTokyoJapan
| | - Sho Aki
- Division of Nutriomics and Oncology, RCASTThe University of TokyoTokyoJapan
- Department of Chemistry and Biotechnology, Graduate School of EngineeringThe University of TokyoTokyoJapan
| | - Maki Sugaya
- Division of Nutriomics and Oncology, RCASTThe University of TokyoTokyoJapan
| | - Haruka Hirose
- Department of Systems Biology, Graduate School of MedicineNagoya UniversityNagoyaJapan
- Present address:
Department of Computational and Systems Biology, Medical Research InstituteTokyo Medical and Dental UniversityTokyoJapan
| | - Miki Kato
- Division of Nutriomics and Oncology, RCASTThe University of TokyoTokyoJapan
| | - Keisuke Maeda
- Division of Nutriomics and Oncology, RCASTThe University of TokyoTokyoJapan
| | - Daichi M Sakamoto
- Department of Chemistry and Biotechnology, Graduate School of EngineeringThe University of TokyoTokyoJapan
| | - Yasuhiro Kojima
- Department of Systems Biology, Graduate School of MedicineNagoya UniversityNagoyaJapan
| | - Miyuki Nishida
- Division of Nutriomics and Oncology, RCASTThe University of TokyoTokyoJapan
| | - Ritsuko Ando
- Division of Nutriomics and Oncology, RCASTThe University of TokyoTokyoJapan
| | - Masashi Muramatsu
- Division of Molecular and Vascular Biology, IRDAKumamoto UniversityKumamotoJapan
| | - Melvin Pan
- Division of Nutriomics and Oncology, RCASTThe University of TokyoTokyoJapan
| | - Rika Tsuchida
- Division of Nutriomics and Oncology, RCASTThe University of TokyoTokyoJapan
| | | | - Hideyuki Yanai
- Department of Inflammology, RCASTThe University of TokyoTokyoJapan
| | - Hiroshi Takano
- Department of Cell BiologyJapanese Foundation for Cancer ResearchTokyoJapan
| | - Ryoji Yao
- Department of Cell BiologyJapanese Foundation for Cancer ResearchTokyoJapan
| | - Shinsuke Sando
- Department of Chemistry and Biotechnology, Graduate School of EngineeringThe University of TokyoTokyoJapan
- Department of Bioengineering, Graduate School of EngineeringThe University of TokyoTokyoJapan
| | - Masabumi Shibuya
- Institute of Physiology and MedicineJobu UniversityTakasakiJapan
| | - Juro Sakai
- Division of Metabolic Medicine, RCASTThe University of TokyoTokyoJapan
- Division of Molecular Physiology and Metabolism, Graduate School of MedicineTohoku UniversitySendaiJapan
| | - Tatsuhiko Kodama
- Division of Nutriomics and Oncology, RCASTThe University of TokyoTokyoJapan
| | - Hiroyasu Kidoya
- Department of Signal Transduction, RIMDOsaka UniversityOsakaJapan
- Department of Integrative Vascular Biology, Faculty of Medical SciencesUniversity of FukuiFukuiJapan
| | - Teppei Shimamura
- Department of Systems Biology, Graduate School of MedicineNagoya UniversityNagoyaJapan
- Present address:
Department of Computational and Systems Biology, Medical Research InstituteTokyo Medical and Dental UniversityTokyoJapan
| | - Tsuyoshi Osawa
- Division of Nutriomics and Oncology, RCASTThe University of TokyoTokyoJapan
- Department of Chemistry and Biotechnology, Graduate School of EngineeringThe University of TokyoTokyoJapan
| |
Collapse
|
139
|
Luo Y, Xie Y, Wu D, Wang B, Lu H, Wang Z, Quan Y, Han B. AL360181.1 promotes proliferation and invasion in colon cancer and is one of ten m6A-related lncRNAs that predict overall survival. PeerJ 2023; 11:e16123. [PMID: 37953780 PMCID: PMC10638913 DOI: 10.7717/peerj.16123] [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: 06/21/2023] [Accepted: 08/27/2023] [Indexed: 11/14/2023] Open
Abstract
Background N6-methyladenosine (m6A) exerted a pivotal role in colon cancer. Nevertheless, the long non-coding RNAs (lncRNAs) associated with this process have yet to be elucidated. Methods The open-access data used for analysis was downloaded from The Cancer Genome Atlas (TCGA) database for analysis, employing the R software for computational evaluations. The RNA level of specific molecules was assessed using the quantitative real-time PCR. CCK8, colony formation and transwell assay were used to evaluate the proliferation, invasion and migration ability of colon cancer cells. Results Here, we identified the m6A regulators from TCGA data and subsequently pinpointed lncRNAs with a -Cor- > 0.3 and P < 0.05, categorizing them as m6A-associated lncRNAs. Moreover, we formulated a prognosis signature rooted in ten m6A-related lncRNAs, consisting of AL360181.1, PCAT6, SNHG26, AC016876.1, AC104667.2, AL114730.3, LINC02257, AC147067.1, AP006621.3 and AC009237.14. This signature exhibited notable predictive accuracy in gauging patient survival. Immune-related evaluations revealed varied immune cell infiltration patterns across different risk groups, with our findings suggesting superior immunotherapy response in low-risk patients. Biological enrichment analysis indicated that the high-risk patients had a higher activity of multiple carcinogenic pathways, including glycolysis. The previously unreported lncRNA, AL360181.1, displayed a connection to glycolytic activity and diminished survival rates, warranting further investigation. The result indicated that AL360181.1 was correlated with more aggressive clinical characteristics. Immune infiltration assessments found AL360181.1 to have a positive correlation with Tcm infiltration, but an inverse relationship with entities like Th2 cells, T cells, neutrophils and macrophages. Biological enrichment analysis indicated that the pathways of WNT/β-catenin, pancreas beta cells, hedgehog signaling and some metabolism pathways were upregulated in high AL360181.1 patients. In vitro experiments showed that AL360181.1 was upregulated in the colon cancer cells. Moreover, AL360181.1 significantly promotes the proliferation, invasion and migration of colon cancer cells. Conclusions Our results can provide direction for future studies on m6A-related lncRNA in colon cancer.
Collapse
Affiliation(s)
- Yi Luo
- Tongren Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Yayun Xie
- Tongren Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Dejun Wu
- Shanghai Pudong Hospital, Fudan University Pudong Medical Center, Shanghai, China
| | - Bingyi Wang
- Tongren Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Helei Lu
- Tongren Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Zhiqiang Wang
- Tongren Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Yingjun Quan
- Tongren Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Bo Han
- Tongren Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| |
Collapse
|
140
|
Arutyunyan I, Jumaniyazova E, Makarov A, Fatkhudinov T. In Vitro Models of Head and Neck Cancer: From Primitive to Most Advanced. J Pers Med 2023; 13:1575. [PMID: 38003890 PMCID: PMC10672510 DOI: 10.3390/jpm13111575] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2023] [Revised: 10/31/2023] [Accepted: 11/01/2023] [Indexed: 11/26/2023] Open
Abstract
For several decades now, researchers have been trying to answer the demand of clinical oncologists to create an ideal preclinical model of head and neck squamous cell carcinoma (HNSCC) that is accessible, reproducible, and relevant. Over the past years, the development of cellular technologies has naturally allowed us to move from primitive short-lived primary 2D cell cultures to complex patient-derived 3D models that reproduce the cellular composition, architecture, mutational, or viral load of native tumor tissue. Depending on the tasks and capabilities, a scientific laboratory can choose from several types of models: primary cell cultures, immortalized cell lines, spheroids or heterospheroids, tissue engineering models, bioprinted models, organoids, tumor explants, and histocultures. HNSCC in vitro models make it possible to screen agents with potential antitumor activity, study the contribution of the tumor microenvironment to its progression and metastasis, determine the prognostic significance of individual biomarkers (including using genetic engineering methods), study the effect of viral infection on the pathogenesis of the disease, and adjust treatment tactics for a specific patient or groups of patients. Promising experimental results have created a scientific basis for the registration of several clinical studies using HNSCC in vitro models.
Collapse
Affiliation(s)
- Irina Arutyunyan
- Research Institute of Molecular and Cellular Medicine, RUDN University, 6 Miklukho-Maklaya Street, 117198 Moscow, Russia; (I.A.); (A.M.); (T.F.)
- National Medical Research Center for Obstetrics, Gynecology and Perinatology Named after Academician V.I. Kulakov Ministry of Healthcare of the Russian Federation, 4 Oparina Street, 117997 Moscow, Russia
| | - Enar Jumaniyazova
- Research Institute of Molecular and Cellular Medicine, RUDN University, 6 Miklukho-Maklaya Street, 117198 Moscow, Russia; (I.A.); (A.M.); (T.F.)
| | - Andrey Makarov
- Research Institute of Molecular and Cellular Medicine, RUDN University, 6 Miklukho-Maklaya Street, 117198 Moscow, Russia; (I.A.); (A.M.); (T.F.)
- Histology Department, Pirogov Russian National Research Medical University, Ministry of Healthcare of the Russian Federation, 117997 Moscow, Russia
| | - Timur Fatkhudinov
- Research Institute of Molecular and Cellular Medicine, RUDN University, 6 Miklukho-Maklaya Street, 117198 Moscow, Russia; (I.A.); (A.M.); (T.F.)
- Avtsyn Research Institute of Human Morphology of Federal State Budgetary Scientific Institution Petrovsky National Research Centre of Surgery, 3 Tsyurupy Street, 117418 Moscow, Russia
| |
Collapse
|
141
|
Yang F, Hilakivi-Clarke L, Shaha A, Wang Y, Wang X, Deng Y, Lai J, Kang N. Metabolic reprogramming and its clinical implication for liver cancer. Hepatology 2023; 78:1602-1624. [PMID: 36626639 PMCID: PMC10315435 DOI: 10.1097/hep.0000000000000005] [Citation(s) in RCA: 18] [Impact Index Per Article: 18.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/19/2022] [Accepted: 09/28/2022] [Indexed: 01/12/2023]
Abstract
Cancer cells often encounter hypoxic and hypo-nutrient conditions, which force them to make adaptive changes to meet their high demands for energy and various biomaterials for biomass synthesis. As a result, enhanced catabolism (breakdown of macromolecules for energy production) and anabolism (macromolecule synthesis from bio-precursors) are induced in cancer. This phenomenon is called "metabolic reprogramming," a cancer hallmark contributing to cancer development, metastasis, and drug resistance. HCC and cholangiocarcinoma (CCA) are 2 different liver cancers with high intertumoral heterogeneity in terms of etiologies, mutational landscapes, transcriptomes, and histological representations. In agreement, metabolism in HCC or CCA is remarkably heterogeneous, although changes in the glycolytic pathways and an increase in the generation of lactate (the Warburg effect) have been frequently detected in those tumors. For example, HCC tumors with activated β-catenin are addicted to fatty acid catabolism, whereas HCC tumors derived from fatty liver avoid using fatty acids. In this review, we describe common metabolic alterations in HCC and CCA as well as metabolic features unique for their subsets. We discuss metabolism of NAFLD as well, because NAFLD will likely become a leading etiology of liver cancer in the coming years due to the obesity epidemic in the Western world. Furthermore, we outline the clinical implication of liver cancer metabolism and highlight the computation and systems biology approaches, such as genome-wide metabolic models, as a valuable tool allowing us to identify therapeutic targets and develop personalized treatments for liver cancer patients.
Collapse
Affiliation(s)
- Flora Yang
- BA/MD Joint Admission Scholars Program, University of Minnesota, Minneapolis, Minnesota
| | - Leena Hilakivi-Clarke
- Food Science and Nutrition Section, The Hormel Institute, University of Minnesota, Austin, Minnesota
| | - Aurpita Shaha
- Tumor Microenvironment and Metastasis Section, the Hormel Institute, University of Minnesota, Austin, Minnesota
| | - Yuanguo Wang
- Tumor Microenvironment and Metastasis Section, the Hormel Institute, University of Minnesota, Austin, Minnesota
| | - Xianghu Wang
- Tumor Microenvironment and Metastasis Section, the Hormel Institute, University of Minnesota, Austin, Minnesota
| | - Yibin Deng
- Department of Urology, Masonic Cancer Center, The University of Minnesota Medical School, Minneapolis, Minnesota
| | - Jinping Lai
- Department of Pathology and Laboratory Medicine, Kaiser Permanente Sacramento Medical Center, Sacramento, California
| | - Ningling Kang
- Tumor Microenvironment and Metastasis Section, the Hormel Institute, University of Minnesota, Austin, Minnesota
| |
Collapse
|
142
|
Mansur MB, deSouza NM, Natrajan R, Abegglen LM, Schiffman JD, Greaves M. Evolutionary determinants of curability in cancer. Nat Ecol Evol 2023; 7:1761-1770. [PMID: 37620552 DOI: 10.1038/s41559-023-02159-w] [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: 01/23/2023] [Accepted: 07/05/2023] [Indexed: 08/26/2023]
Abstract
The emergence of drug-resistant cells, most of which have a mutated TP53 gene, prevents curative treatment in most advanced and common metastatic cancers of adults. Yet, a few, rarer malignancies, all of which are TP53 wild type, have high cure rates. In this Perspective, we discuss how common features of curable cancers offer insights into the evolutionary and developmental determinants of drug resistance. Acquired loss of TP53 protein function is the most common genetic change in cancer. This probably reflects positive selection in the context of strong ecosystem pressures including microenvironmental hypoxia. Loss of TP53's functions results in multiple fitness benefits and enhanced evolvability of cancer cells. TP53-null cells survive apoptosis, and tolerate potent oncogenic signalling, DNA damage and genetic instability. In addition, critically, they provide an expanded pool of self-renewing, or stem, cells, the primary units of evolutionary selection in cancer, making subsequent adaptation to therapeutic challenge by drug resistance highly probable. The exceptional malignancies that are curable, including the common genetic subtype of childhood acute lymphoblastic leukaemia and testicular seminoma, differ from the common adult cancers in originating prenatally from embryonic or fetal cells that are developmentally primed for TP53-dependent apoptosis. Plus, they have other genetic and phenotypic features that enable dissemination without exposure to selective pressures for TP53 loss, retaining their intrinsic drug hypersensitivity.
Collapse
Affiliation(s)
| | - Nandita M deSouza
- Division of Radiotherapy and Imaging, The Institute of Cancer Research, London, UK
- Department of Imaging, The Royal Marsden National Health Service (NHS) Foundation Trust, London, UK
| | - Rachael Natrajan
- The Breast Cancer Now Toby Robins Research Centre, Division of Breast Cancer, The Institute of Cancer Research, London, UK
| | - Lisa M Abegglen
- Department of Pediatrics and Huntsman Cancer Institute, University of Utah, Salt Lake City, UT, USA
| | - Joshua D Schiffman
- Department of Pediatrics and Huntsman Cancer Institute, University of Utah, Salt Lake City, UT, USA
- Peel Therapeutics, Inc., Salt Lake City, UT, USA
| | - Mel Greaves
- Centre for Evolution and Cancer, The Institute of Cancer Research, London, UK.
| |
Collapse
|
143
|
Khorshid M, Varshosaz J, Rostami M, Haghiralsadat F, Akbari V, Khorshid P. Anti HER-2 aptamer functionalized gold nanoparticles of dasatinib for targeted chemo-radiotherapy in breast cancer cells. BIOMATERIALS ADVANCES 2023; 154:213591. [PMID: 37611441 DOI: 10.1016/j.bioadv.2023.213591] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/28/2023] [Revised: 08/07/2023] [Accepted: 08/13/2023] [Indexed: 08/25/2023]
Abstract
In the present study, gold nanoparticles functionalized with anti HER-2 aptamer were designed for effective targeted delivery of dasatinib (DSB) to breast cancer cells. Anti HER-2 aptamer attached to porous or plain gold nanoparticles were compared for dasatinib delivery. Activated drug with succinic anhydride and L-cysteine linker was used for conjugation of DSB to gold nanoparticles. The loading efficiency of the activated drug on plain and porous gold nanoparticles was 52 and 68 %, respectively, which was significantly more than the loading of free DSB in gold nanoparticles (1-2.5 %). The anti HER-2 aptamer was conjugated to porous gold nanoparticles loaded with the activated drug. Various characterization techniques such as FESEM, TEM, AFM, zeta potential and ICP-MS were used to confirm the binding of the drug to gold nanoparticles. 1HNMR and FTIR spectroscopic analyses were employed to examine the structural characteristics of the conjugated drug. These analytical techniques confirmed the successful incorporation of succinyl and thiol groups onto the drug molecule. The amount of aptamer binding to different types of gold nanoparticles was obtained from the intensity of the light emitted from the bands observed in electrophoresis gel and due to the presence of porosity in porous gold nanoparticles, the amount of aptamer conjugation on porous gold nanoparticles increased compared to plain ones. Cell cytotoxicity and cellular uptake were evaluated by MTT assay and TEM in BT-474 and MCF-7 cells. Aptamer-functionalized porous gold nanoparticles containing activated dasatinib showed higher cytotoxicity and cellular uptake than modified DSB-loaded nanoparticles and un-activated DSB. The combination of radiation therapy with the modified dasatinib attached to porous gold nanoparticles and aptamer demonstrated a notable reduction in the IC50 values for both the BT-474 and MCF-7 cell lines. Specifically, the IC50 value for the BT-474 cells decreased from 6.95 μM (for unmodified dasatinib) to 2.57 μM, while for the MCF-7 cells, it decreased from 13.97 μM to 8.57 μM. These findings indicate a significant improvement in the efficacy of the modified dasatinib compared to its unmodified counterpart when used in conjunction with radiation therapy.
Collapse
Affiliation(s)
- Mahdis Khorshid
- Department of Pharmaceutics, Faculty of Pharmacy and Novel Drug Delivery Systems Research Centre, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Jaleh Varshosaz
- Department of Pharmaceutics, Faculty of Pharmacy and Novel Drug Delivery Systems Research Centre, Isfahan University of Medical Sciences, Isfahan, Iran.
| | - Mahboubeh Rostami
- Department of Medicinal Chemistry, School of Pharmacy and Pharmaceutical Sciences, Isfahan University of Medical Sciences, Isfahan, Iran.
| | - Fateme Haghiralsadat
- Department of Advanced Medical Sciences, School of Paramedicine, Shahid Sadoughi University of Medical Sciences, Yazd, Iran
| | - Vajihe Akbari
- Department of Pharmaceutical Biotechnology, School of Pharmacy and Pharmaceutical Sciences, Isfahan University of Medical Sciences, Isfahan, Iran.
| | - Pardis Khorshid
- Department of Medicinal Chemistry, Faculty of Pharmacy, Shahid Sadoughi University of Medical Sciences, Yazd, Iran
| |
Collapse
|
144
|
Pandkar MR, Sinha S, Samaiya A, Shukla S. Oncometabolite lactate enhances breast cancer progression by orchestrating histone lactylation-dependent c-Myc expression. Transl Oncol 2023; 37:101758. [PMID: 37572497 PMCID: PMC10425713 DOI: 10.1016/j.tranon.2023.101758] [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: 06/15/2023] [Revised: 07/22/2023] [Accepted: 08/07/2023] [Indexed: 08/14/2023] Open
Abstract
Due to the enhanced glycolytic rate, cancer cells generate lactate copiously, subsequently promoting the lactylation of histones. While previous studies have explored the impact of histone lactylation in modulating gene expression, the precise role of this epigenetic modification in regulating oncogenes is largely unchartered. In this study, using breast cancer cell lines and their mutants exhibiting lactate-deficient metabolome, we have identified that an enhanced rate of aerobic glycolysis supports c-Myc expression via promoter-level histone lactylation. Interestingly, c-Myc further transcriptionally upregulates serine/arginine splicing factor 10 (SRSF10) to drive alternative splicing of MDM4 and Bcl-x in breast cancer cells. Moreover, our results reveal that restricting the activity of critical glycolytic enzymes affects the c-Myc-SRSF10 axis to subside the proliferation of breast cancer cells. Our findings provide novel insights into the mechanisms by which aerobic glycolysis influences alternative splicing processes that collectively contribute to breast tumorigenesis. Furthermore, we also envisage that chemotherapeutic interventions attenuating glycolytic rate can restrict breast cancer progression by impeding the c-Myc-SRSF10 axis.
Collapse
Affiliation(s)
- Madhura R Pandkar
- Department of Biological Sciences, Indian Institute of Science Education and Research Bhopal, Bhopal, Madhya Pradesh 462066, India. https://twitter.com/https://twitter.com/MadhuraPandkar
| | - Sommya Sinha
- Department of Biological Sciences, Indian Institute of Science Education and Research Bhopal, Bhopal, Madhya Pradesh 462066, India. https://twitter.com/https://twitter.com/sinha_sommya
| | - Atul Samaiya
- Department of Surgical Oncology, Bansal Hospital, Bhopal, Madhya Pradesh 462016, India
| | - Sanjeev Shukla
- Department of Biological Sciences, Indian Institute of Science Education and Research Bhopal, Bhopal, Madhya Pradesh 462066, India.
| |
Collapse
|
145
|
Huang SS, Wu LY, Qiu Y, Xie Y, Wu H, Li YQ, Xie XH. Identification of lactate-related subgroups and prognostic model in triple-negative breast cancer. J Cancer Res Clin Oncol 2023; 149:13107-13122. [PMID: 37474680 DOI: 10.1007/s00432-023-05171-6] [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: 06/08/2023] [Accepted: 07/09/2023] [Indexed: 07/22/2023]
Abstract
BACKGROUND Triple-negative breast cancer (TNBC) is a highly aggressive subtype of breast cancer that exhibits elevated glycolytic capacity. Lactate, as a byproduct of glycolysis, is considered a major oncometabolite that plays an important role in oncogenesis and remodeling of the tumor microenvironment. However, the potential roles of lactate in TNBC are not yet fully understood. In this study, our goal was to identify prognosis-related lactate genes (PLGs) and construct a lactate-related prognostic model (LRPM) for TNBC. METHODS First, we applied lactate-related genes to classify TNBC samples using a hierarchical clustering algorithm. Then, we performed the log-rank analysis and the least absolute shrinkage and selection operator analysis to screen PLGs and construct the LRPM. The biological functions of the identified PLGs in TNBC were investigated using CCK8 assay and clone formation assay. Finally, we constructed a nomogram based on the lactate-risk score and tumor clinical stage. We used the operating characteristic curve and decision curve analysis to evaluate the predictive capability of the nomogram. RESULTS Our results showed that the TNBC samples could be classified into two subgroups with different survival probabilities. Three genes (NDUFAF3, CARS2 and FH), which can suppress TNBC cell proliferation, were identified as PLGs. Moreover, the LRPM and nomogram exhibited excellent predictive performance for TNBC patient prognosis. CONCLUSION We have developed a novel LRPM that enables risk stratification and identification of poor molecular subtypes in TNBC patients, showing great potential in clinical practice.
Collapse
Affiliation(s)
- Shan-Shan Huang
- Department of Breast Oncology, State Key Laboratory of Oncology in South China, Sun Yat-Sen University Cancer Center, 651 Dongfeng Road East, Guangzhou, 510060, China
| | - Lin-Yu Wu
- Department of Breast Oncology, State Key Laboratory of Oncology in South China, Sun Yat-Sen University Cancer Center, 651 Dongfeng Road East, Guangzhou, 510060, China
| | - Yu Qiu
- Department of Breast Oncology, State Key Laboratory of Oncology in South China, Sun Yat-Sen University Cancer Center, 651 Dongfeng Road East, Guangzhou, 510060, China
| | - Yi Xie
- Department of Breast Oncology, State Key Laboratory of Oncology in South China, Sun Yat-Sen University Cancer Center, 651 Dongfeng Road East, Guangzhou, 510060, China
| | - Hao Wu
- Department of Breast Oncology, State Key Laboratory of Oncology in South China, Sun Yat-Sen University Cancer Center, 651 Dongfeng Road East, Guangzhou, 510060, China
| | - Ying-Qing Li
- Outpatient Department, State Key Laboratory of Oncology in South China, Sun Yat-Sen University Cancer Center, 651 Dongfeng Road East, Guangzhou, 510060, China.
| | - Xin-Hua Xie
- Department of Breast Oncology, State Key Laboratory of Oncology in South China, Sun Yat-Sen University Cancer Center, 651 Dongfeng Road East, Guangzhou, 510060, China.
| |
Collapse
|
146
|
Su Z, Zhang G, Li X, Zhang H. Inverse correlation between Alzheimer's disease and cancer from the perspective of hypoxia. Neurobiol Aging 2023; 131:59-73. [PMID: 37572528 DOI: 10.1016/j.neurobiolaging.2023.07.002] [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: 01/08/2023] [Revised: 06/02/2023] [Accepted: 07/03/2023] [Indexed: 08/14/2023]
Abstract
Sporadic Alzheimer's disease and cancer remain epidemiologically inversely related, and exploring the reverse pathogenesis is important for our understanding of both. Cognitive dysfunctions in Alzheimer's disease (AD) might result from the depletion of adaptive reserves in the brain. Energy storage in the brain is limited and is dynamically regulated by neurovascular and neurometabolic coupling. The research on neurodegenerative diseases has been dominated by the neurocentric view that neuronal defects cause the diseases. However, the proposal of the 2-hit vascular hypothesis in AD led us to focus on alterations in the vasculature, especially hypoperfusion. Chronic hypoxia is a feature shared by AD and cancer. It is interesting how contradicting chronic hypoxia's effects on both cancer and AD are. In this article, we discuss the potential links between the 2 diseases' etiology, from comparable upstream circumstances to diametrically opposed downstream effects. We suggest opposing potential mechanisms, including upregulation and downregulation of hypoxia-inducible factor-1α, the Warburg and reverse-Warburg effects, lactate-mediated intracellular acidic and alkaline conditions, and VDAC1-mediated apoptosis and antiapoptosis, and search for regulators that may be identified as the crossroads between cancer and AD.
Collapse
Affiliation(s)
- Zhan Su
- Department of Neurology and Neuroscience Centre, The First Hospital of Jilin University, Changchun, China
| | - Guimei Zhang
- Department of Neurology and Neuroscience Centre, The First Hospital of Jilin University, Changchun, China
| | - Xiangting Li
- Department of Neurology and Neuroscience Centre, The First Hospital of Jilin University, Changchun, China
| | - Haining Zhang
- Department of Neurology and Neuroscience Centre, The First Hospital of Jilin University, Changchun, China.
| |
Collapse
|
147
|
Hoh Kam J, Mitrofanis J. Glucose Improves the Efficacy of Photobiomodulation in Changing ATP and ROS Levels in Mouse Fibroblast Cell Cultures. Cells 2023; 12:2533. [PMID: 37947612 PMCID: PMC10648764 DOI: 10.3390/cells12212533] [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: 09/26/2023] [Revised: 10/16/2023] [Accepted: 10/25/2023] [Indexed: 11/12/2023] Open
Abstract
In this study, we tested the idea that photobiomodulation-the application of red to near infrared light (~λ = 600-1300 nm) to body tissues-is more effective in influencing cell metabolism when glucose is readily available. To this end, we used a mouse fibroblast (L-929) cell culture model and had two sets of conditions: non-stressed (10% FBS (foetal bovine serum)) and stressed (1% FBS), both either with or without glucose. We treated (or not) cells with photobiomodulation using an 810 nm laser at 15 mW/cm2 (~7.2 J/cm2). Our results showed that photobiomodulation was neither cytotoxic nor effective in enhancing measures of cell viability and proliferation, together with protein levels in any of the cell cultures. Photobiomodulation was, however, effective in increasing adenosine triphosphate (ATP) and decreasing reactive oxygen species (ROS) levels and this was-most importantly-only in conditions where glucose was present; corresponding cultures that did not contain glucose did not show these changes. In summary, we found that the benefits of photobiomodulation, in particular in changing ATP and ROS levels, were induced only when there was glucose available. Our findings lay a template for further explorations into the mechanisms of photobiomodulation, together with having considerable experimental and clinical implications.
Collapse
Affiliation(s)
- Jaimie Hoh Kam
- Fonds de Dotation Clinatec, Grenoble Alpes University, 38000 Grenoble, France;
| | - John Mitrofanis
- Fonds de Dotation Clinatec, Grenoble Alpes University, 38000 Grenoble, France;
- Institute of Ophthalmology, University College London, London EC1V 9EL, UK
| |
Collapse
|
148
|
Yang X, Ma H, Zhang M, Wang R, Li X. TRIM32 promotes oral squamous cell carcinoma progression by enhancing FBP2 ubiquitination and degradation. Biochem Biophys Res Commun 2023; 678:165-172. [PMID: 37640002 DOI: 10.1016/j.bbrc.2023.08.030] [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: 07/31/2023] [Accepted: 08/16/2023] [Indexed: 08/31/2023]
Abstract
The aberrant expression of TRIM32, an E3 ubiquitin ligase, has been identified in multiple malignant cancer types. Nevertheless, the functional roles and detailed mechanisms of TRIM32 in oral squamous cell carcinoma (OSCC) remain to be elucidated. Here, we investigated TRIM32 expression and its functional role in OSCC. TRIM32 expression was consistently elevated in OSCC tissues, particularly in samples from patients with advanced clinical grades. Functionally, silencing TRIM32 dampened OSCC cell growth, migration and invasion. Additionally, a xenograft tumor model suggested that TRIM32 knockdown suppressed in vivo OSCC tumor growth and lung metastasis formation. Mechanistically, we discovered that TRIM32 directly bound to the FBP2 protein via mass spectrometry and co-immunoprecipitation. TRIM32 could interact with FBP2 and accelerates its degradation, eventually enhancing glycolysis in OSCC cell lines. Importantly, rescue assays demonstrated that FBP2 silencing could at least partially offset the tumor-suppressive and aerobic glycolysis inhibition effect induced by TRIM32 knockdown. Thus, our findings demonstrate that TRIM32 plays a crucial role in promoting tumor growth and enhancing glycolysis through FBP2 inhibition. Given OSCC is associated with increased glycolysis levels, our study suggests potential therapeutic targets for OSCC treatment.
Collapse
Affiliation(s)
- Xiao Yang
- Department of Stomatology, Affiliated Hospital of Jining Medical University, No. 89, Guhuai Road, Jining, 272000, Shandong, China; Postdoctoral Mobile Station of Shandong University of Traditional Chinese Medicine, No. 4655, Daxue Road, Changqing District, Jinan, 250399, Shandong, China.
| | - Haifeng Ma
- Department of Stomatology, Affiliated Hospital of Jining Medical University, No. 89, Guhuai Road, Jining, 272000, Shandong, China.
| | - Min Zhang
- Department of Stomatology, Affiliated Hospital of Jining Medical University, No. 89, Guhuai Road, Jining, 272000, Shandong, China.
| | - Renzhong Wang
- Department of Otolaryngology, Affiliated Hospital of Shandong University of Traditional Chinese Medicine, No. 42, Wenhua Road, Jinan, 250011, Shandong, China.
| | - Xiaoyu Li
- Department of Otolaryngology, Affiliated Hospital of Jining Medical University, No. 89, Guhuai Road, Jining, 272000, Shandong, China.
| |
Collapse
|
149
|
Skandalis SS. CD44 Intracellular Domain: A Long Tale of a Short Tail. Cancers (Basel) 2023; 15:5041. [PMID: 37894408 PMCID: PMC10605500 DOI: 10.3390/cancers15205041] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2023] [Revised: 10/10/2023] [Accepted: 10/16/2023] [Indexed: 10/29/2023] Open
Abstract
CD44 is a single-chain transmembrane receptor that exists in multiple forms due to alternative mRNA splicing and post-translational modifications. CD44 is the main cell surface receptor of hyaluronan as well as other extracellular matrix molecules, cytokines, and growth factors that play important roles in physiological processes (such as hematopoiesis and lymphocyte homing) and the progression of various diseases, the predominant one being cancer. Currently, CD44 is an established cancer stem cell marker in several tumors, implying a central functional role in tumor biology. The present review aims to highlight the contribution of the CD44 short cytoplasmic tail, which is devoid of any enzymatic activity, in the extraordinary functional diversity of the receptor. The interactions of CD44 with cytoskeletal proteins through specific structural motifs within its intracellular domain drives cytoskeleton rearrangements and affects the distribution of organelles and transport of molecules. Moreover, the CD44 intracellular domain specifically interacts with various cytoplasmic effectors regulating cell-trafficking machinery, signal transduction pathways, the transcriptome, and vital cell metabolic pathways. Understanding the cell type- and context-specificity of these interactions may unravel the high complexity of CD44 functions and lead to novel improved therapeutic interventions.
Collapse
Affiliation(s)
- Spyros S Skandalis
- Biochemistry, Biochemical Analysis & Matrix Pathobiology Res. Group, Laboratory of Biochemistry, Department of Chemistry, University of Patras, 26504 Patras, Greece
| |
Collapse
|
150
|
Duan SL, Wu M, Zhang ZJ, Chang S. The potential role of reprogrammed glucose metabolism: an emerging actionable codependent target in thyroid cancer. J Transl Med 2023; 21:735. [PMID: 37853445 PMCID: PMC10585934 DOI: 10.1186/s12967-023-04617-2] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2023] [Accepted: 10/11/2023] [Indexed: 10/20/2023] Open
Abstract
Although the incidence of thyroid cancer is increasing year by year, most patients, especially those with differentiated thyroid cancer, can usually be cured with surgery, radioactive iodine, and thyroid-stimulating hormone suppression. However, treatment options for patients with poorly differentiated thyroid cancers or radioiodine-refractory thyroid cancer have historically been limited. Altered energy metabolism is one of the hallmarks of cancer and a well-documented feature in thyroid cancer. In a hypoxic environment with extreme nutrient deficiencies resulting from uncontrolled growth, thyroid cancer cells utilize "metabolic reprogramming" to satisfy their energy demand and support malignant behaviors such as metastasis. This review summarizes past and recent advances in our understanding of the reprogramming of glucose metabolism in thyroid cancer cells, which we expect will yield new therapeutic approaches for patients with special pathological types of thyroid cancer by targeting reprogrammed glucose metabolism.
Collapse
Affiliation(s)
- Sai-Li Duan
- Department of General Surgery, Xiangya Hospital Central South University, Changsha, 410008, Hunan, People's Republic of China
| | - Min Wu
- Department of General Surgery, Xiangya Hospital Central South University, Changsha, 410008, Hunan, People's Republic of China
| | - Zhe-Jia Zhang
- Department of General Surgery, Xiangya Hospital Central South University, Changsha, 410008, Hunan, People's Republic of China.
| | - Shi Chang
- Department of General Surgery, Xiangya Hospital Central South University, Changsha, 410008, Hunan, People's Republic of China.
- Xiangya Hospital, National Clinical Research Center for Geriatric Disorders, Changsha, 410008, Hunan, People's Republic of China.
- Clinical Research Center for Thyroid Disease in Hunan Province, Changsha, 410008, Hunan, People's Republic of China.
- Hunan Provincial Engineering Research Center for Thyroid and Related Diseases Treatment Technology, Changsha, 410008, Hunan, People's Republic of China.
| |
Collapse
|