1
|
Gorodetska I, Samusieva A, Lahuta T, Ponomarova O, Socha O, Kozeretska I. Exploring New Frontiers: Alternative Breast Cancer Treatments Through Glycocalyx Research. Breast J 2025; 2025:9952727. [PMID: 40443562 PMCID: PMC12122162 DOI: 10.1155/tbj/9952727] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2024] [Accepted: 05/03/2025] [Indexed: 06/02/2025]
Abstract
Breast cancer (BC) treatment is developing toward more precise and personalized care through the approval of different comprehensive approaches. Clinical practice emphasizes significant patient-to-patient variability in treatment response among patients, even those with similar clinical and biological profiles. Recent studies have demonstrated that the glycocalyx is an essential organelle that plays an important role in many cellular processes and can be a promising target for treatment. The glycocalyx of cancer cells is a key component influencing the interaction between the tumor and the immune system. Glycan modifications attached to glycoproteins and glycolipids are a common characteristic of the transition to malignancy. We review how the specific structure and function of the glycocalyx are regulated at the molecular level, contribute to immune evasion, and can be overcome by using both traditional drugs and combination therapies, as well as drugs not previously used in standard cancer treatments, to address treatment resistance associated with glycocalyx alterations. Trial Registration: ClinicalTrials.gov identifier: NCT00770354, NCT00925548, NCT01731587, NCT00088413, NCT00179309, NCT00986609, NCT05812326, NCT04020575, NCT05239143, NCT01279603, and NCT03562637.
Collapse
Affiliation(s)
- Ielizaveta Gorodetska
- Institute of Radiooncology – OncoRay, Helmholtz-Zentrum Dresden-Rossendorf, Dresden, Germany
| | - Anastasiia Samusieva
- Department of Oncology, Shupyk National Healthcare University of Ukraine, Kyiv, Ukraine
| | - Tetiana Lahuta
- Department of General and Molecular Pathophysiology, Bogomoletz Institute of Physiology, Kyiv, Ukraine
| | - Olga Ponomarova
- Department of Oncology, Shupyk National Healthcare University of Ukraine, Kyiv, Ukraine
- Department of Chemotherapy #1, Kyiv Municipal Clinical Oncological Center, Kyiv, Ukraine
| | - Oleg Socha
- Provincial Hospital. St. Padre Pio, Przemyśl, Poland
| | | |
Collapse
|
2
|
Xu J, Xu X, Zhang H, Wu J, Pan R, Zhang B. Tumor-associated inflammation: The role and research progress in tumor therapy. J Drug Deliv Sci Technol 2024; 102:106376. [DOI: 10.1016/j.jddst.2024.106376] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2025]
|
3
|
Xue S, Cai Y, Liu J, Ji K, Yi P, Long H, Zhang X, Li P, Song Y. Dysregulation of phosphoenolpyruvate carboxykinase in cancers: A comprehensive analysis. Cell Signal 2024; 120:111198. [PMID: 38697449 DOI: 10.1016/j.cellsig.2024.111198] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2024] [Revised: 04/02/2024] [Accepted: 04/29/2024] [Indexed: 05/05/2024]
Abstract
BACKGROUND Phosphoenolpyruvate carboxykinase (PEPCK) plays a crucial role in gluconeogenesis, glycolysis, and the tricarboxylic acid cycle by converting oxaloacetate into phosphoenolpyruvate. Two distinct isoforms of PEPCK, specifically cytosolic PCK1 and mitochondrial PCK2, have been identified. Nevertheless, the comprehensive understanding of their dysregulation in pan-cancer and their potential mechanism contributing to signaling transduction pathways remains elusive. METHODS We conducted comprehensive analyses of PEPCK gene expression across 33 diverse cancer types using data from The Cancer Genome Atlas (TCGA). Multiple public databases such as HPA, TIMER 2.0, GEPIA2, cBioPortal, UALCAN, CancerSEA, and String were used to investigate protein levels, prognostic significance, clinical associations, genetic mutations, immune cell infiltration, single-cell sequencing, and functional enrichment analysis in patients with pan-cancer. PEPCK expression was analyzed about different clinical and genetic factors of patients using data from TCGA, GEO, and CGGA databases. Furthermore, the role of PCK2 in Glioma was examined using both in vitro and in vivo experiments. RESULTS The analysis we conducted revealed that the expression of PEPCK is involved in both clinical outcomes and immune cell infiltration. Initially, we verified the high expression of PCK2 in GBM cells and its role in metabolic reprogramming and proliferation in GBM. CONCLUSION Our study showed a correlation between PEPCK (PCK1 and PCK2) expression with clinical prognosis, gene mutation, and immune infiltrates. These findings identified two possible predictive biomarkers across different cancer types, as well as a comprehensive analysis of PCK2 expression in various tumors, with a focus on GBM.
Collapse
Affiliation(s)
- Shuaishuai Xue
- Department of Neurosurgery, Nanfang Hospital, Southern Medical University, Guangzhou 510515, China
| | - Yonghua Cai
- Department of Neurosurgery, Nanfang Hospital, Southern Medical University, Guangzhou 510515, China
| | - Jun Liu
- Department of Neurosurgery, The 2(nd) affiliated hospital Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi 330000, China
| | - Ke Ji
- Department of Neurosurgery, Nanfang Hospital, Southern Medical University, Guangzhou 510515, China
| | - Peiyao Yi
- Department of Neurosurgery, Nanfang Hospital, Southern Medical University, Guangzhou 510515, China
| | - Hao Long
- Department of Neurosurgery, Nanfang Hospital, Southern Medical University, Guangzhou 510515, China
| | - Xian Zhang
- Department of Neurosurgery, Nanfang Hospital, Southern Medical University, Guangzhou 510515, China.
| | - Peng Li
- Department of Neurosurgery, Nanfang Hospital, Southern Medical University, Guangzhou 510515, China.
| | - Ye Song
- Department of Neurosurgery, Nanfang Hospital, Southern Medical University, Guangzhou 510515, China.
| |
Collapse
|
4
|
Tajaldini M, Poorkhani A, Amiriani T, Amiriani A, Javid H, Aref P, Ahmadi F, Sadani S, Khori V. Strategy of targeting the tumor microenvironment via inhibition of fibroblast/fibrosis remodeling new era to cancer chemo-immunotherapy resistance. Eur J Pharmacol 2023; 957:175991. [PMID: 37619785 DOI: 10.1016/j.ejphar.2023.175991] [Citation(s) in RCA: 17] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2023] [Revised: 08/02/2023] [Accepted: 08/10/2023] [Indexed: 08/26/2023]
Abstract
The use of repurposing drugs that may have neoplastic and anticancer effects increases the efficiency and decrease resistance to chemotherapy drugs through a biochemical and mechanical transduction mechanisms through modulation of fibroblast/fibrosis remodeling in tumor microenvironment (TME). Interestingly, fibroblast/fibrosis remodeling plays a vital role in mediating cancer metastasis and drug resistance after immune chemotherapy. The most essential hypothesis for induction of chemo-immunotherapy resistance is via activation of fibroblast/fibrosis remodeling and preventing the infiltration of T cells after is mainly due to the interference between cytoskeleton, mechanical, biochemical, metabolic, vascular, and remodeling signaling pathways in TME. The structural components of the tumor that can be targeted in the fibroblast/fibrosis remodeling include the depletion of the TME components, targeting the cancer-associated fibroblasts and tumor associated macrophages, alleviating the mechanical stress within the ECM, and normalizing the blood vessels. It has also been found that during immune-chemotherapy, TME injury and fibroblast/fibrosis remodeling causes the up-regulation of inhibitory signals and down-regulation of activated signals, which results in immune escape or chemo-resistance of the tumor. In this regard, repurposing or neo-adjuvant drugs with various transduction signaling mechanisms, including anti-fibrotic effects, are used to target the TME and fibroblast/fibrosis signaling pathway such as angiotensin 2, transforming growth factor-beta, physical barriers of the TME, cytokines and metabolic factors which finally led to the reverse of the chemo-resistance. Consistent to many repurposing drugs such as pirfenidone, metformin, losartan, tranilast, dexamethasone and pentoxifylline are used to decrease immune-suppression by abrogation of TME inhibitory signal that stimulates the immune system and increases efficiency and reduces resistance to chemotherapy drugs. To overcome immunosuppression based on fibroblast/fibrosis remodeling, in this review, we focus on inhibitory signal transduction, which is the physical barrier, alleviates mechanical stress and prevents mechano-metabolic activation.
Collapse
Affiliation(s)
- Mahboubeh Tajaldini
- Ischemic Disorder Research Center, Golestan University of Medical Sciences, Gorgan, Iran
| | - Amirhoushang Poorkhani
- Ischemic Disorder Research Center, Golestan University of Medical Sciences, Gorgan, Iran
| | - Taghi Amiriani
- Ischemic Disorder Research Center, Golestan University of Medical Sciences, Gorgan, Iran
| | - Amirhossein Amiriani
- Ischemic Disorder Research Center, Golestan University of Medical Sciences, Gorgan, Iran
| | - Hossein Javid
- Department of Medical Laboratory Sciencess, Catastega Institue of Medical Sciences, Mashhad, Iran
| | - Parham Aref
- Ischemic Disorder Research Center, Golestan University of Medical Sciences, Gorgan, Iran
| | - Farahnazsadat Ahmadi
- Ischemic Disorder Research Center, Golestan University of Medical Sciences, Gorgan, Iran
| | - Somayeh Sadani
- Ischemic Disorder Research Center, Golestan University of Medical Sciences, Gorgan, Iran.
| | - Vahid Khori
- Ischemic Disorder Research Center, Golestan University of Medical Sciences, Gorgan, Iran.
| |
Collapse
|
5
|
Melnik D, Cortés-Sánchez JL, Sandt V, Kahlert S, Kopp S, Grimm D, Krüger M. Dexamethasone Selectively Inhibits Detachment of Metastatic Thyroid Cancer Cells during Random Positioning. Cancers (Basel) 2023; 15:cancers15061641. [PMID: 36980530 PMCID: PMC10046141 DOI: 10.3390/cancers15061641] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2023] [Revised: 02/27/2023] [Accepted: 03/06/2023] [Indexed: 03/11/2023] Open
Abstract
We recently reported that synthetic glucocorticoid dexamethasone (DEX) is able to suppress metastasis-like spheroid formation in a culture of follicular thyroid cancer (FTC)-133 cells cultured under random positioning. We now show that this inhibition was selective for two metastatic thyroid carcinoma cells, FTC-133 and WRO, whereas benign Nthy-ori 3-1 thyrocytes and recurrent ML-1 follicular thyroid cancer cells were not affected by DEX. We then compare Nthy-ori 3-1 and FTC-133 cells concerning their adhesion and mechanosignaling. We demonstrate that DEX disrupts random positioning-triggered p38 stress signaling in FTC-133 cells, thereby antagonizing a variety of biological functions. Thus, DEX treatment of FTC-133 cells is associated with increased adhesiveness, which is mainly caused by the restored, pronounced formation of a normal number of tight junctions. Moreover, we show that Nthy-ori 3-1 and ML-1 cells upregulate the anti-adhesion protein mucin-1 during random positioning, presumably as a protection against mechanical stress. In summary, mechanical stress seems to be an important component in this metastasis model system that is processed differently by metastatic and healthy cells. The balance between adhesion, anti-adhesion and cell–cell connections enables detachment of adherent human cells on the random positioning machine—or not, allowing selective inhibition of thyroid in vitro metastasis by DEX.
Collapse
Affiliation(s)
- Daniela Melnik
- Department of Microgravity and Translational Regenerative Medicine, Otto von Guericke University, 39106 Magdeburg, Germany
- Research Group “Magdeburger Arbeitsgemeinschaft für Forschung unter Raumfahrt- und Schwerelosigkeitsbedingungen” (MARS), Otto von Guericke University, 39106 Magdeburg, Germany
| | - José Luis Cortés-Sánchez
- Department of Microgravity and Translational Regenerative Medicine, Otto von Guericke University, 39106 Magdeburg, Germany
| | - Viviann Sandt
- Department of Microgravity and Translational Regenerative Medicine, Otto von Guericke University, 39106 Magdeburg, Germany
| | - Stefan Kahlert
- Research Group “Magdeburger Arbeitsgemeinschaft für Forschung unter Raumfahrt- und Schwerelosigkeitsbedingungen” (MARS), Otto von Guericke University, 39106 Magdeburg, Germany
- Institute of Anatomy, Otto von Guericke University, 39120 Magdeburg, Germany
| | - Sascha Kopp
- Research Group “Magdeburger Arbeitsgemeinschaft für Forschung unter Raumfahrt- und Schwerelosigkeitsbedingungen” (MARS), Otto von Guericke University, 39106 Magdeburg, Germany
- Core Facility Tissue Engineering, Otto von Guericke University, 39106 Magdeburg, Germany
| | - Daniela Grimm
- Department of Microgravity and Translational Regenerative Medicine, Otto von Guericke University, 39106 Magdeburg, Germany
- Research Group “Magdeburger Arbeitsgemeinschaft für Forschung unter Raumfahrt- und Schwerelosigkeitsbedingungen” (MARS), Otto von Guericke University, 39106 Magdeburg, Germany
- Department of Biomedicine, Aarhus University, 8000 Aarhus, Denmark
| | - Marcus Krüger
- Department of Microgravity and Translational Regenerative Medicine, Otto von Guericke University, 39106 Magdeburg, Germany
- Research Group “Magdeburger Arbeitsgemeinschaft für Forschung unter Raumfahrt- und Schwerelosigkeitsbedingungen” (MARS), Otto von Guericke University, 39106 Magdeburg, Germany
- Correspondence: ; Tel.: +49-391-6757471
| |
Collapse
|
6
|
Dexamethasone Is Not Sufficient to Facilitate Tenogenic Differentiation of Dermal Fibroblasts in a 3D Organoid Model. Biomedicines 2023; 11:biomedicines11030772. [PMID: 36979751 PMCID: PMC10044928 DOI: 10.3390/biomedicines11030772] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2022] [Revised: 01/11/2023] [Accepted: 02/22/2023] [Indexed: 03/08/2023] Open
Abstract
Self-assembling three-dimensional organoids that do not rely on an exogenous scaffold but maintain their native cell-to-cell and cell-to-matrix interactions represent a promising model in the field of tendon tissue engineering. We have identified dermal fibroblasts (DFs) as a potential cell type for generating functional tendon-like tissue. The glucocorticoid dexamethasone (DEX) has been shown to regulate cell proliferation and facilitate differentiation towards other mesenchymal lineages. Therefore, we hypothesized that the administration of DEX could reduce excessive DF proliferation and thus, facilitate the tenogenic differentiation of DFs using a previously established 3D organoid model combined with dose-dependent application of DEX. Interestingly, the results demonstrated that DEX, in all tested concentrations, was not sufficient to notably induce the tenogenic differentiation of human DFs and DEX-treated organoids did not have clear advantages over untreated control organoids. Moreover, high concentrations of DEX exerted a negative impact on the organoid phenotype. Nevertheless, the expression profile of tendon-related genes of untreated and 10 nM DEX-treated DF organoids was largely comparable to organoids formed by tendon-derived cells, which is encouraging for further investigations on utilizing DFs for tendon tissue engineering.
Collapse
|
7
|
T Cell Energy Metabolism Is a Target of Glucocorticoids in Mice, Healthy Humans, and MS Patients. Cells 2023; 12:cells12030450. [PMID: 36766792 PMCID: PMC9914408 DOI: 10.3390/cells12030450] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2022] [Revised: 01/24/2023] [Accepted: 01/29/2023] [Indexed: 01/31/2023] Open
Abstract
Glucocorticoids (GCs) are used to treat inflammatory disorders such as multiple sclerosis (MS) by exerting prominent activities in T cells including apoptosis induction and suppression of cytokine production. However, little is known about their impact on energy metabolism, although it is widely accepted that this process is a critical rheostat of T cell activity. We thus tested the hypothesis that GCs control genes and processes involved in nutrient transport and glycolysis. Our experiments revealed that escalating doses of dexamethasone (Dex) repressed energy metabolism in murine and human primary T cells. This effect was mediated by the GC receptor and unrelated to both apoptosis induction and Stat1 activity. In contrast, treatment of human T cells with rapamycin abolished the repression of metabolic gene expression by Dex, unveiling mTOR as a critical target of GC action. A similar phenomenon was observed in MS patients after intravenous methylprednisolon (IVMP) pulse therapy. The expression of metabolic genes was reduced in the peripheral blood T cells of most patients 24 h after GC treatment, an effect that correlated with disease activity. Collectively, our results establish the regulation of T cell energy metabolism by GCs as a new immunomodulatory principle.
Collapse
|
8
|
Afshari AR, Sanati M, Aminyavari S, Shakeri F, Bibak B, Keshavarzi Z, Soukhtanloo M, Jalili-Nik M, Sadeghi MM, Mollazadeh H, Johnston TP, Sahebkar A. Advantages and drawbacks of dexamethasone in glioblastoma multiforme. Crit Rev Oncol Hematol 2022; 172:103625. [PMID: 35158070 DOI: 10.1016/j.critrevonc.2022.103625] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2021] [Revised: 02/01/2022] [Accepted: 02/07/2022] [Indexed: 12/25/2022] Open
Abstract
The most widespread, malignant, and deadliest type of glial tumor is glioblastoma multiforme (GBM). Despite radiation, chemotherapy, and radical surgery, the median survival of afflicted individuals is about 12 months. Unfortunately, existing therapeutic interventions are abysmal. Dexamethasone (Dex), a synthetic glucocorticoid, has been used for many years to treat brain edema and inflammation caused by GBM. Several investigations have recently shown that Dex also exerts antitumoral effects against GBM. On the other hand, more recent disputed findings have questioned the long-held dogma of Dex treatment for GBM. Unfortunately, steroids are associated with various undesirable side effects, including severe immunosuppression and metabolic changes like hyperglycemia, which may impair the survival of GBM patients. Current ideas and concerns about Dex's effects on GBM cerebral edema, cell proliferation, migration, and its clinical outcomes were investigated in this study.
Collapse
Affiliation(s)
- Amir R Afshari
- Department of Physiology and Pharmacology, Faculty of Medicine, North Khorasan University of Medical Sciences, Bojnurd, Iran
| | - Mehdi Sanati
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Birjand University of Medical Sciences, Birjand, Iran
| | - Samaneh Aminyavari
- Department of Neuroscience and Addiction Studies, School of Advanced Technologies in Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Farzaneh Shakeri
- Department of Physiology and Pharmacology, Faculty of Medicine, North Khorasan University of Medical Sciences, Bojnurd, Iran
| | - Bahram Bibak
- Department of Physiology and Pharmacology, Faculty of Medicine, North Khorasan University of Medical Sciences, Bojnurd, Iran
| | - Zakieh Keshavarzi
- Department of Physiology and Pharmacology, Faculty of Medicine, North Khorasan University of Medical Sciences, Bojnurd, Iran
| | - Mohammad Soukhtanloo
- Department of Clinical Biochemistry, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Mohammad Jalili-Nik
- Department of Clinical Biochemistry, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Mohammad Montazami Sadeghi
- Department of Physiology and Pharmacology, Faculty of Medicine, North Khorasan University of Medical Sciences, Bojnurd, Iran
| | - Hamid Mollazadeh
- Department of Physiology and Pharmacology, Faculty of Medicine, North Khorasan University of Medical Sciences, Bojnurd, Iran.
| | - Thomas P Johnston
- Division of Pharmacology and Pharmaceutical Sciences, School of Pharmacy, University of Missouri-Kansas City, Kansas City, MO, USA
| | - Amirhossein Sahebkar
- Biotechnology Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran; Applied Biomedical Research Center, Mashhad University of Medical Sciences, Mashhad, Iran; School of Medicine, The University of Western Australia, Perth, Australia; Department of Biotechnology, School of Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran.
| |
Collapse
|
9
|
Pu J, Zhou X, Liu J, Hou P, Ji M. Therapeutic potential and deleterious effect of glucocorticoids on azoxymethane/dextran sulfate sodium-induced colorectal cancer in mice. Am J Cancer Res 2021; 11:4866-4883. [PMID: 34765297 PMCID: PMC8569368] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2021] [Accepted: 09/15/2021] [Indexed: 06/13/2023] Open
Abstract
Glucocorticoids (GCs) are widely used in the treatment of various autoimmune and inflammatory diseases, including inflammatory bowel disease (IBD). However, the effect of GCs on the progression of colitis-associated colorectal cancer (CAC) has not been well explored. In this study, we first established a colorectal cancer model induced by azoxymethane and dextran sulfate sodium (AOM/DSS) and a colitis model induced by DSS in mice. Dexamethasone (DEX) was then administered at different periods of time to determine its effect on tumorigenesis and tumor progression. Meanwhile, body weight, stool property and fecal blood of mice were recorded. At the end of this study, the number and load of tumors were evaluated, and the expression of proteins associated with cell proliferation was analyzed. To evaluate the inflammation in colon, we detected the level of pro-inflammatory cytokine TNFα, and the mucosal infiltration of inflammatory cells. Our results revealed that AOM injection followed by three cycles of drinking water containing 1.5% DSS successfully induced multiple tumor formation in mouse colon and rectum. Both early and late DEX intervention suppressed tumor growth in mouse colorectum, and downregulated the expression of PCNA and cyclin D1. Moreover, DEX treatment significantly inhibited TNFα production, mucosal infiltration of inflammatory cells, and the activity of MAPK/JNK pathway, particularly early DEX intervention. However, we also found that DEX treatment deteriorated the general state of mouse manifested by greater loss of body weight and rectal bleeding. In summary, both early and late DEX interventions significantly ameliorate colonic inflammation and inhibit the progression of AOM/DSS-induced colorectal cancer, at least partly due to the inhibition of MAPK/JNK pathway. It is noteworthy that the deleterious effect on the general condition of mouse may limit the duration of GCs treatment.
Collapse
Affiliation(s)
- Jun Pu
- Key Laboratory for Tumor Precision Medicine of Shaanxi Province, The First Affiliated Hospital of Xi’an Jiaotong UniversityXi’an 710061, Shaanxi, China
| | - Xinrui Zhou
- Key Laboratory for Tumor Precision Medicine of Shaanxi Province, The First Affiliated Hospital of Xi’an Jiaotong UniversityXi’an 710061, Shaanxi, China
| | - Jiaxin Liu
- Key Laboratory for Tumor Precision Medicine of Shaanxi Province, The First Affiliated Hospital of Xi’an Jiaotong UniversityXi’an 710061, Shaanxi, China
| | - Peng Hou
- Key Laboratory for Tumor Precision Medicine of Shaanxi Province, The First Affiliated Hospital of Xi’an Jiaotong UniversityXi’an 710061, Shaanxi, China
- Department of Endocrinology, The First Affiliated Hospital of Xi’an Jiaotong UniversityXi’an 710061, Shaanxi, China
| | - Meiju Ji
- Key Laboratory for Tumor Precision Medicine of Shaanxi Province, The First Affiliated Hospital of Xi’an Jiaotong UniversityXi’an 710061, Shaanxi, China
- Center for Translational Medicine, The First Affiliated Hospital of Xi’an Jiaotong UniversityXi’an 710061, Shaanxi, China
| |
Collapse
|
10
|
Xu M, Yang J, Sun J, Xing X, Liu Z, Liu T. A novel mutation in PCK2 gene causes primary angle-closure glaucoma. Aging (Albany NY) 2021; 13:23338-23347. [PMID: 34650006 PMCID: PMC8544327 DOI: 10.18632/aging.203627] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2021] [Accepted: 10/03/2021] [Indexed: 12/25/2022]
Abstract
Primary angle-closure glaucoma (PACG) is an ophthalmic genetic disease characterized by direct contact between the iris and trabecular meshwork, resulting in an obstructed outflow of aqueous humor from the eye. However, it is unclear as to what role genetics plays in the development of PACG. The present study investigated the disease-causing mutation in a five-generation Chinese PACG family using whole-genome sequencing. A novel heterozygous missense mutation c.977C>T in PCK2 gene was identified in five affected family members, but not in any unaffected and 86 unrelated healthy individuals. This nucleotide substitute is predicted to result in a proline to leucine substitution p.Pro326Leu. Furthermore, the function of this mutation was analyzed through various in vitro assays using the RGC-5 cell line. Our results demonstrate that the p.Pro326Leu mutation induces RGC-5 cell cycle arrest and apoptosis with a decreased BcL-XL. The increasing P53, P27, P21, AKT, and P-GSK3α were also detected in the cells transfected with c.977C>T mutation, suggesting that this mutation within PCK2 gene cause PACG through impairment of AKT/GSK3α signaling pathway.
Collapse
Affiliation(s)
- Menghan Xu
- Department of Ophthalmology, 3201 Hospital, Xi'an Jiaotong University Health Science Center, Hanzhong, Shaanxi 723000, China.,The First People's Hospital of Xianyang, Xianyang, Shaanxi 712000, China
| | - Jin Yang
- Department of Ophthalmology, 3201 Hospital, Xi'an Jiaotong University Health Science Center, Hanzhong, Shaanxi 723000, China.,Department of Ophthalmology, Shaanxi Provincial People's Hospital, Xi'an, Shaanxi 710068, China
| | - Jiayue Sun
- Department of Ophthalmology, 3201 Hospital, Xi'an Jiaotong University Health Science Center, Hanzhong, Shaanxi 723000, China
| | - Xuemei Xing
- Department of Ophthalmology, 3201 Hospital, Xi'an Jiaotong University Health Science Center, Hanzhong, Shaanxi 723000, China
| | - Zheng Liu
- College of Medical Laboratory Science, Guilin Medical University, Guilin, Guangxi 541004, China
| | - Tao Liu
- Department of Ophthalmology, 3201 Hospital, Xi'an Jiaotong University Health Science Center, Hanzhong, Shaanxi 723000, China.,Guihang Guiyang Hospital Affiliated to Zunyi Medical University, Guiyang, Guizhou 550000, China
| |
Collapse
|
11
|
Kuncharoenwirat N, Chatuphonprasert W, Jarukamjorn K. Differential Impacts of Phenol Red on Benzo[ a]pyrene and Dexamethasone-Modified Cytochrome P450s in Human Cancer Cells. Pak J Biol Sci 2021; 24:790-800. [PMID: 34486298 DOI: 10.3923/pjbs.2021.790.800] [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: 11/15/2022]
Abstract
<b>Background and Objective:</b> Phenol red, the pH indicator in cell culture media, influences the expression of cytochrome P450s (CYPs) in cell lines. This study aimed to examine how phenol red modified CYP induction by benzo[<i>a</i>]pyrene and dexamethasone in human hepatocarcinoma (HepG2), colorectal adenocarcinoma (Caco-2) and choriocarcinoma (BeWo) cells. <b>Materials and Methods:</b> The cells (1×10<sup>5</sup> cells/well in a 24-well plate) were incubated with benzo[<i>a</i>]pyrene (0.1, 1 and 10 μM) or dexamethasone (1, 5 and 10 μM) in either phenol red or phenol red-free media for 24 hrs. The mRNA expression of CYPs was determined by Real-Time Polymerase Chain Reaction (RT/qPCR). <b>Results:</b> Phenol red enhanced expression of benzo[<i>a</i>]pyrene-induced CYP1A2 inHepG2 and BeWo cells and suppressed benzo[<i>a</i>]pyrene-induced CYP2A6 expression in HepG2 and Caco-2 cells, benzo[<i>a</i>]pyrene induced CYP2B6 expression in HepG2 cells and benzo[<i>a</i>]pyrene- and dexamethasone-induced CYP3A4 expression in HepG2 and Caco-2 cells. The expression of CYP3A5 was affected differently in HepG2 and Caco-2 cell lines. Phenol red enhanced benzo[<i>a</i>]pyrene- and dexamethasone-induced CYP3A5 expression in Caco-2 cells but suppressed benzo[<i>a</i>]pyrene- and dexamethasone-induced CYP3A5 expression in HepG2 cells. <b>Conclusion:</b> Phenol red differentially influenced expression of benzo[<i>a</i>]pyrene- and dexamethasone-induced CYP1A2, CYP2A6, CYP2B6, CYP3A4 and CYP3A5 mRNAs in HepG2, Caco-2 and BeWo cells. Therefore, the inclusion of phenol red in cell culture media is of concern in studies of drug and xenobiotic metabolism via CYPs in human cell line models.
Collapse
|
12
|
Lin J, Wu S, Ye S, Papa APD, Yang J, Huang S, Arthur G, Zhuge Q, Zhang Y. Oridonin interrupts cellular bioenergetics to suppress glioma cell growth by down-regulating PCK2. Phytother Res 2021; 35:2624-2638. [PMID: 33438793 DOI: 10.1002/ptr.7009] [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: 05/15/2020] [Revised: 12/04/2020] [Accepted: 12/16/2020] [Indexed: 12/24/2022]
Abstract
We aim to evaluate the tumor metabolic suppressive activity of Oridonin (extract of Rabdosia rubescens) in glioma and elucidate its potential mechanism. Effects of Oridonin on U251/U87 cells were determined by CCK8, RTCA, colony formation, flow cytometry, wound healing, and Transwell assay. Xenograft tumor model to evaluate the effect of Oridonin on glioma cells in vivo. Cellular bioenergetics were measured by Seahorse. RNA-seq was performed to screen potential biological pathways in Oridonin treated cells. Bioinformatics analysis of PCK2 in glioma was performed based on TCGA/CGGA. Endogenous PCK2 was knocked-down by lentivirus packaged shRNA. We found Oridonin significantly inhibited cell growth in U251/U87 in vitro and in vivo. Both oxygen consumption rate (OCR) and extracellular acidification rate (ECAR) were decreased in Oridonin-treated U251/U87 cells. Oridonin treatment led to PCK2 down-regulation. Additionally, PCK2 was up-regulated in higher grade glioma and correlated with poor outcomes. Furthermore, PCK2 depletion significantly inhibited cell growth and decreased OCR/ECAR in U251/U87 which coincided with the effects of Oridonin. Therefore, we evaluated the potent anti-tumor property of Oridonin in glioma. Importantly, we demonstrated that PCK2 might be a novel target of Oridonin on glioma by inducing energy crisis and increasing oxidative stress.
Collapse
Affiliation(s)
- Jianhu Lin
- Zhejiang Provincial Key Laboratory of Aging and Neurological Disorder Research, Department of Neurosurgery, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Shanshan Wu
- Zhejiang Provincial Key Laboratory of Aging and Neurological Disorder Research, Department of Neurosurgery, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Sisi Ye
- Zhejiang Provincial Key Laboratory of Aging and Neurological Disorder Research, Department of Neurosurgery, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Akuetteh Percy David Papa
- Zhejiang Provincial Key Laboratory of Aging and Neurological Disorder Research, Department of Neurosurgery, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Jianjing Yang
- Zhejiang Provincial Key Laboratory of Aging and Neurological Disorder Research, Department of Neurosurgery, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Shengwei Huang
- Zhejiang Provincial Key Laboratory of Aging and Neurological Disorder Research, Department of Neurosurgery, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | | | - Qichuan Zhuge
- Zhejiang Provincial Key Laboratory of Aging and Neurological Disorder Research, Department of Neurosurgery, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Yu Zhang
- Zhejiang Provincial Key Laboratory of Aging and Neurological Disorder Research, Department of Neurosurgery, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| |
Collapse
|
13
|
Donia T, Khedr S, Salim EI, Hessien M. Trichostatin A sensitizes hepatoma cells to Taxol more than 5-Aza-dC and dexamethasone. Drug Metab Pers Ther 2021; 36:299-309. [PMID: 34773731 DOI: 10.1515/dmpt-2020-0186] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2020] [Accepted: 03/16/2021] [Indexed: 11/15/2022]
Abstract
OBJECTIVES This work was designed to compare the sensitizing effects of epigenetic modifiers on cancer cells vs. that of glucocorticoids. Also, to evaluate their effects on genes involved in epigenetic changes and drug metabolism. METHODS Hepatoma cells (HepG2) were treated with the anticancer drug (Taxol), with a histone deacetylase inhibitor (Trichostatin A [TSA]), DNA methyltransferase inhibitor (5-Aza-dC) or dexamethasone (DEX). Cytotoxicity was assessed by MTT assay and the apoptosis was determined by Annexin V-FITC. The expression levels of HDAC1, HDAC3, Dnmt1, Dnmt3α, CYP1A2, CYP3A4, CYP2B6, CYP2C19 and CYP2D6 were monitored by qRT-PCR. RESULTS TSA, synergistically enhanced cells sensitivity with the anticancer effect of Taxol more than 5-Aza-dC and DEX. This was evidenced by the relative decrease in IC50 in cells cotreated with Taxol + TSA, Taxol + 5-Aza-dC or Taxol + DEX. Apoptosis was induced in 51.2, 16.9 and 41.3% of cells, respectively. In presence of Taxol, TSA induced four-fold increase in the expression of HDAC1 and downregulated Dnmt1&3α genes. CYP2D6 demonstrated progressive expression (up to 28-fold) with the increasing number of drugs. Moreover, the isoform overexpressed in cells treated with TSA + Taxol > DEX + Taxol > 5-Aza-dC + Taxol (6.4, 4.6 and 2.99, respectively). The investigated genes were clustered in two distinct subsets, where no coregulation was observed between HDAC1 and HDAC3. However, tight pairwise correlation-based cluster was seen between (CYP3A4/Dnmt3α and CYP2D6/CYP2C19). CONCLUSIONS The data reflects the sensitizing effect of acetylation modification by TSA on the responsiveness of hepatoma cells to anticancer therapy. The effect of histone deacetylase inhibition was more than hypomethylation and glucocorticoid effects. TSA exerts its role through its modulatory role on epigenetics and drugs metabolizing genes. Other modifiers (5-Aza-dC and DEX), however may adopt different mechanisms.
Collapse
Affiliation(s)
- Thoria Donia
- Division of Biochemistry, Department of Chemistry, Faculty of Science, Tanta University, Tanta, Egypt
| | - Sherien Khedr
- College of Pharmacy, Arab Academy for Science, Technology & Maritime Transport, Alexandria, Egypt
| | - Elsayed I Salim
- Department of Zoology, Faculty of Science, Tanta University, Tanta, Egypt
| | - Mohamed Hessien
- Division of Biochemistry, Department of Chemistry, Faculty of Science, Tanta University, Tanta, Egypt
| |
Collapse
|
14
|
Donia T, Khedr S, Salim EI, Hessien M. Trichostatin A sensitizes hepatoma cells to Taxol more than 5-Aza-dC and dexamethasone. Drug Metab Pers Ther 2021; 0:dmdi-2020-0186. [PMID: 33818027 DOI: 10.1515/dmdi-2020-0186] [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: 12/05/2020] [Accepted: 03/16/2021] [Indexed: 11/15/2022]
Abstract
OBJECTIVES This work was designed to compare the sensitizing effects of epigenetic modifiers on cancer cells vs. that of glucocorticoids. Also, to evaluate their effects on genes involved in epigenetic changes and drug metabolism. METHODS Hepatoma cells (HepG2) were treated with the anticancer drug (Taxol), with a histone deacetylase inhibitor (Trichostatin A [TSA]), DNA methyltransferase inhibitor (5-Aza-dC) or dexamethasone (DEX). Cytotoxicity was assessed by MTT assay and the apoptosis was determined by Annexin V-FITC. The expression levels of HDAC1, HDAC3, Dnmt1, Dnmt3α, CYP1A2, CYP3A4, CYP2B6, CYP2C19 and CYP2D6 were monitored by qRT-PCR. RESULTS TSA, synergistically enhanced cells sensitivity with the anticancer effect of Taxol more than 5-Aza-dC and DEX. This was evidenced by the relative decrease in IC50 in cells cotreated with Taxol + TSA, Taxol + 5-Aza-dC or Taxol + DEX. Apoptosis was induced in 51.2, 16.9 and 41.3% of cells, respectively. In presence of Taxol, TSA induced four-fold increase in the expression of HDAC1 and downregulated Dnmt1&3α genes. CYP2D6 demonstrated progressive expression (up to 28-fold) with the increasing number of drugs. Moreover, the isoform overexpressed in cells treated with TSA + Taxol > DEX + Taxol > 5-Aza-dC + Taxol (6.4, 4.6 and 2.99, respectively). The investigated genes were clustered in two distinct subsets, where no coregulation was observed between HDAC1 and HDAC3. However, tight pairwise correlation-based cluster was seen between (CYP3A4/Dnmt3α and CYP2D6/CYP2C19). CONCLUSIONS The data reflects the sensitizing effect of acetylation modification by TSA on the responsiveness of hepatoma cells to anticancer therapy. The effect of histone deacetylase inhibition was more than hypomethylation and glucocorticoid effects. TSA exerts its role through its modulatory role on epigenetics and drugs metabolizing genes. Other modifiers (5-Aza-dC and DEX), however may adopt different mechanisms.
Collapse
Affiliation(s)
- Thoria Donia
- Division of Biochemistry, Department of Chemistry, Faculty of Science, Tanta University, Tanta, Egypt
| | - Sherien Khedr
- Division of Biochemistry, Department of Chemistry, Faculty of Science, Tanta University, Tanta, Egypt
| | - Elsayed I Salim
- Department of Zoology, Faculty of Science, Tanta University, Tanta, Egypt
| | - Mohamed Hessien
- Division of Biochemistry, Department of Chemistry, Faculty of Science, Tanta University, Tanta, Egypt
| |
Collapse
|
15
|
Luvanda MK, Posch W, Vosper J, Zaderer V, Noureen A, Lass-Flörl C, Wilflingseder D. Dexamethasone Promotes Aspergillus fumigatus Growth in Macrophages by Triggering M2 Repolarization via Targeting PKM2. J Fungi (Basel) 2021; 7:70. [PMID: 33498318 PMCID: PMC7909285 DOI: 10.3390/jof7020070] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2020] [Revised: 01/13/2021] [Accepted: 01/14/2021] [Indexed: 01/06/2023] Open
Abstract
Since long-term corticosteroid treatment is associated with emerging opportunistic fungal infections causing high morbidity and mortality in immune-suppressed individuals, here we characterized the impact of dexamethasone (Dex) treatment on Aspergillus fumigatus-related immune modulation. We found by high content screening and flow cytometric analyses that during monocyte-to-macrophage differentiation, as little as 0.1 µg/mL Dex resulted in a shift in macrophage polarization from M1 to M2-like macrophages. This macrophage repolarization mediated via Dex was characterized by significant upregulation of the M2 marker CD163 and downmodulation of M1 markers CD40 and CD86 as well as changes in phenotypic properties and adherence. These Dex-mediated phenotypic alterations were furthermore associated with a metabolic switch in macrophages orchestrated via PKM2. Such treated macrophages lost their ability to prevent Aspergillus fumigatus germination, which was correlated with accelerated fungal growth, destruction of macrophages, and induction of an anti-inflammatory cytokine profile. Taken together, repolarization of macrophages following corticosteroid treatment and concomitant switch to an anti-inflammatory phenotype might play a prominent role in triggering invasive aspergillosis (IA) due to suppression of innate immunological responses necessary to combat extensive fungal outgrowth.
Collapse
Affiliation(s)
- Maureen K. Luvanda
- Institute of Hygiene and Medical Microbiology, Medical University of Innsbruck, 6020 Innsbruck, Austria; (M.K.L.); (W.P.); (V.Z.); (A.N.); (C.L.-F.)
| | - Wilfried Posch
- Institute of Hygiene and Medical Microbiology, Medical University of Innsbruck, 6020 Innsbruck, Austria; (M.K.L.); (W.P.); (V.Z.); (A.N.); (C.L.-F.)
| | - Jonathan Vosper
- Institute of Medical Biochemistry, Medical University of Innsbruck, 6020 Innsbruck, Austria;
| | - Viktoria Zaderer
- Institute of Hygiene and Medical Microbiology, Medical University of Innsbruck, 6020 Innsbruck, Austria; (M.K.L.); (W.P.); (V.Z.); (A.N.); (C.L.-F.)
| | - Asma Noureen
- Institute of Hygiene and Medical Microbiology, Medical University of Innsbruck, 6020 Innsbruck, Austria; (M.K.L.); (W.P.); (V.Z.); (A.N.); (C.L.-F.)
| | - Cornelia Lass-Flörl
- Institute of Hygiene and Medical Microbiology, Medical University of Innsbruck, 6020 Innsbruck, Austria; (M.K.L.); (W.P.); (V.Z.); (A.N.); (C.L.-F.)
| | - Doris Wilflingseder
- Institute of Hygiene and Medical Microbiology, Medical University of Innsbruck, 6020 Innsbruck, Austria; (M.K.L.); (W.P.); (V.Z.); (A.N.); (C.L.-F.)
| |
Collapse
|
16
|
Brunekreeft KL, Paijens ST, Wouters MC, Komdeur FL, Eggink FA, Lubbers JM, Workel HH, Van Der Slikke EC, Pröpper NE, Leffers N, Adam J, Pijper H, Plat A, Kol A, Nijman HW, De Bruyn M. Deep immune profiling of ovarian tumors identifies minimal MHC-I expression after neoadjuvant chemotherapy as negatively associated with T-cell-dependent outcome. Oncoimmunology 2020; 9:1760705. [PMID: 32923120 PMCID: PMC7458665 DOI: 10.1080/2162402x.2020.1760705] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2019] [Accepted: 01/30/2020] [Indexed: 12/12/2022] Open
Abstract
Epithelial Ovarian cancer (EOC) is the most lethal gynecological malignancy and has limited curative therapeutic options. Immunotherapy for EOC is promising, but clinical efficacy remains restricted to a small percentage of patients. Several lines of evidence suggest that the low response rate might be improved by combining immunotherapy with carboplatin and paclitaxel, the standard-of-care chemotherapy for EOC. Here, we assessed the immune contexture of EOC tumors, draining lymph nodes, and peripheral blood mononuclear cells during carboplatin/paclitaxel chemotherapy. We observed that the immune contexture of EOC patients is defined by the tissue of origin, independent of exposure to chemotherapy. Summarized, draining lymph nodes were characterized by a quiescent microenvironment composed of mostly non-proliferating naïve CD4 + T cells. Circulating T cells shared phenotypic features of both lymph nodes and tumor-infiltrating immune cells. Immunologically 'hot' ovarian tumors were characterized by ICOS, GITR, and PD-1 expression on CD4 + and CD8 + cells, independent of chemotherapy. The presence of PD-1 + cells in tumors prior to, but not after, chemotherapy was associated with disease-specific survival (DSS). Accordingly, we observed high MHC-I expression in tumors prior to chemotherapy, but minimal MHC-I expression in tumors after neoadjuvant chemotherapy, even though there were no differences in the number of tumor-infiltrating lymphocytes (TIL) in both groups. We therefore speculate that the TIL influx into the chemotherapy tumor microenvironment may be a consequence of the general inflammatory nature of chemotherapy-experienced tumors. Strategies to upregulate MHC-I during or after neoadjuvant chemotherapy may thus improve treatment outcome in these patients.
Collapse
Affiliation(s)
- Kim L. Brunekreeft
- Department of Obstetrics and Gynecology, University of Groningen, University Medical Center Groningen, The Netherlands
| | - Sterre T. Paijens
- Department of Obstetrics and Gynecology, University of Groningen, University Medical Center Groningen, The Netherlands
| | | | - Fenne L. Komdeur
- Department of Obstetrics and Gynecology, University of Groningen, University Medical Center Groningen, The Netherlands
| | - Florine A. Eggink
- Department of Obstetrics and Gynecology, University of Groningen, University Medical Center Groningen, The Netherlands
| | - Joyce M. Lubbers
- Department of Obstetrics and Gynecology, University of Groningen, University Medical Center Groningen, The Netherlands
| | - Hagma H. Workel
- Department of Obstetrics and Gynecology, University of Groningen, University Medical Center Groningen, The Netherlands
| | - Elisabeth C. Van Der Slikke
- Department of Obstetrics and Gynecology, University of Groningen, University Medical Center Groningen, The Netherlands
| | - Noor E.J. Pröpper
- Department of Obstetrics and Gynecology, University of Groningen, University Medical Center Groningen, The Netherlands
| | - Ninke Leffers
- Department of Obstetrics and Gynecology, University of Groningen, University Medical Center Groningen, The Netherlands
| | - Julien Adam
- Department of Clinical Biology, Institut De Cancérologie Gustave Roussy, Paris, France
| | - Harry Pijper
- Department of Obstetrics and Gynecology, University of Groningen, University Medical Center Groningen, The Netherlands
| | - Annechien Plat
- Department of Obstetrics and Gynecology, University of Groningen, University Medical Center Groningen, The Netherlands
| | - Arjan Kol
- Department of Obstetrics and Gynecology, University of Groningen, University Medical Center Groningen, The Netherlands
| | - Hans W. Nijman
- Department of Obstetrics and Gynecology, University of Groningen, University Medical Center Groningen, The Netherlands
| | - Marco De Bruyn
- Department of Obstetrics and Gynecology, University of Groningen, University Medical Center Groningen, The Netherlands
| |
Collapse
|
17
|
High-Dose Dexamethasone Manipulates the Tumor Microenvironment and Internal Metabolic Pathways in Anti-Tumor Progression. Int J Mol Sci 2020; 21:ijms21051846. [PMID: 32156004 PMCID: PMC7084511 DOI: 10.3390/ijms21051846] [Citation(s) in RCA: 32] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2020] [Revised: 03/04/2020] [Accepted: 03/05/2020] [Indexed: 12/13/2022] Open
Abstract
High-dose dexamethasone (DEX) is used to treat chemotherapy-induced nausea and vomiting or to control immunotherapy-related autoimmune diseases in clinical practice. However, the underlying mechanisms of high-dose DEX in tumor progression remain unaddressed. Therefore, we explored the effects of high-dose DEX on tumor progression and the potential mechanisms of its anti-tumor function using immunohistochemistry, histological examination, real-time quantitative PCR (qPCR), and Western blotting. Tumor volume, blood vessel invasion, and levels of the cell proliferation markers Ki67 and c-Myc and the anti-apoptotic marker Bcl2 decreased in response to high-dose DEX. However, the cell apoptosis marker cleaved caspase 3 increased significantly in mice treated with 50 mg/kg DEX compared with controls. Some genes associated with immune responses were significantly downregulated following treatment with 50 mg/kg DEX e.g., Cxcl9, Cxcl10, Cd3e, Gzmb, Ifng, Foxp3, S100a9, Arg1, and Mrc1. In contrast, the M1-like tumor-associated macrophages (TAMs) activation marker Nos2 was shown to be increased. Moreover, the expression of peroxisome proliferator-activated receptors α and γ (Pparα and Pparg, respectively) was shown to be significantly upregulated in livers or tumors treated with DEX. However, high-dose DEX treatment decreased the expression of glucose and lipid metabolic pathway-related genes such as glycolysis-associated genes (Glut1, Hk2, Pgk1, Idh3a), triglyceride (TG) synthesis genes (Gpam, Agpat2, Dgat1), exogenous free fatty acid (FFA) uptake-related genes (Fabp1, Slc27a4, and CD36), and fatty acid oxidation (FAO) genes (Acadm, Acaa1, Cpt1a, Pnpla2). In addition, increased serum glucose and decreased serum TG and non-esterified fatty acid (NEFA) were observed in DEX treated-xenografted tumor mice. These findings indicate that high-dose DEX-inhibited tumor progression is a complicated process, not only activated by M1-like TAMs, but also decreased by the uptake and consumption of glucose and lipids that block the raw material and energy supply of cancer cells. Activated M1-like TAMs and inefficient glucose and lipid metabolism delayed tumor cell growth and promoted apoptosis. These findings have important implications for the application of DEX combined with drugs that target key metabolism pathways for tumor therapy in clinical practice.
Collapse
|
18
|
Chen Q, Xia R, Zheng W, Zhang L, Li P, Sun X, Shi J. Metronomic paclitaxel improves the efficacy of PD-1 monoclonal antibodies in breast cancer by transforming the tumor immune microenvironment. Am J Transl Res 2020; 12:519-530. [PMID: 32194900 PMCID: PMC7061845] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2019] [Accepted: 01/22/2020] [Indexed: 06/10/2023]
Abstract
The clinical efficacy of PD-1/PD-L1 monoclonal antibodies (mAbs) in triple-negative breast cancer (TNBC) is unsatisfactory. Immunotherapy combined with chemotherapy shows good therapeutic potential. Preclinical and clinical studies have shown that metronomic chemotherapy may stimulate anticancer immune responses. We aimed to verify whether metronomic paclitaxel (PTX, TAX) treatment can improve the efficacy of a PD-1 mAb in a TNBC mouse model and to explore the potential mechanism. After constructing the TNBC mouse model and treating with PD-1 mAb, metronomic PTX chemotherapy or combined therapy, the differences in the efficacy of each treatment group were compared and analyzed. Our findings suggested that the combination of metronomic PTX chemotherapy and PD-1 mAb produces a potent antitumor effect. Further experiments demonstrated that metronomic PTX chemotherapy changed the immune cell population in tumor tissues. These data suggest that metronomic PTX improves the efficacy of the PD-1 mAb in TNBC by transforming the tumor immune microenvironment, and these results provide strong evidence for the use of this treatment in TNBC patients in the future.
Collapse
Affiliation(s)
- Qian Chen
- Department of Oncology, The Affiliated Suzhou Hospital of Nanjing Medical University242 Guangji Road, Suzhou 215008, Jiangsu, China
- Central Laboratory, The Affiliated Suzhou Hospital of Nanjing Medical University242 Guangji Road, Suzhou 215008, Jiangsu, China
| | - Rui Xia
- Department of Oncology, The Second Affiliated Hospital of Soochow UniversitySuzhou 215004, Jiangsu, China
| | - Weiwei Zheng
- Central Laboratory, The Affiliated Suzhou Hospital of Nanjing Medical University242 Guangji Road, Suzhou 215008, Jiangsu, China
| | - Luyao Zhang
- Department of Oncology, The Affiliated Suzhou Hospital of Nanjing Medical University242 Guangji Road, Suzhou 215008, Jiangsu, China
| | - Ping Li
- Central Laboratory, The Affiliated Suzhou Hospital of Nanjing Medical University242 Guangji Road, Suzhou 215008, Jiangsu, China
| | - Xingwei Sun
- Department of Intervention, The Second Affiliated Hospital of Soochow UniversitySuzhou 215004, Jiangsu, China
| | - Jianming Shi
- Department of Oncology, The Affiliated Suzhou Hospital of Nanjing Medical University242 Guangji Road, Suzhou 215008, Jiangsu, China
- Central Laboratory, The Affiliated Suzhou Hospital of Nanjing Medical University242 Guangji Road, Suzhou 215008, Jiangsu, China
| |
Collapse
|