1
|
Duan X, Hou R, Huang Y, Wang C, Liu L, Du H, Shi J. Comprehensive expression, prognostic and validation analysis of necroptosis-related lncRNAs in esophageal cancer. Transl Oncol 2024; 46:101983. [PMID: 38797018 PMCID: PMC11152745 DOI: 10.1016/j.tranon.2024.101983] [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: 12/27/2023] [Revised: 04/12/2024] [Accepted: 04/30/2024] [Indexed: 05/29/2024] Open
Abstract
BACKGROUND Previous studies have shown that necroptosis-related long noncoding RNA (lncRNA) risk models can be used to predict prognosis and immune infiltration in patients with esophageal cancer. However, further analysis of the regulatory mechanisms of necroptosis-related lncRNAs used in risk models remains to be conducted. The purpose of the present study was to identify valuable necroptosis-related lncRNAs in esophageal cancer and to verify their molecular and cellular functions. METHODS Esophageal cancer data were downloaded from The Cancer Genome Atlas (TCGA). The expression of eight genes (LINC00299, AC090912.2, AC244197.2, AL158166.1, AC079684.1, AP003696.1, AC079684.1 and AP003696.1) in the necroptosis-related lncRNA risk model, their relationships with clinicopathological stage, and their diagnostic receiver operating characteristic (ROC) curves were analyzed. The prognostic value of these lncRNAs for overall survival (OS) and disease specific survival (DSS) was analyzed, and time-dependent ROC curves were generated. The AP003696.1 target gene (lncRNA ENSG00000253385.1) was further investigated through immune infiltration analysis, Gene Ontology/Kyoto Encyclopedia of Genes and Genomes (GO/KEGG) enrichment analyses, and gene coexpression analysis. Finally, in vitro functional assays based on lncRNA ENSG00000253385.1 were conducted to explore its regulatory role in esophageal cancer. RESULTS A bioinformatics approach was used to study the eight genes in the necroptosis-related lncRNA risk model. AP003696.1 (lncRNA ENSG00000253385.1) was highly expressed in esophageal cancer tissues, and its high expression was correlated with poor OS and DFdS. Both univariate and multivariate Cox regression analyses revealed that lncRNA ENSG00000253385.1 is an independent prognostic factor. The lncRNA ENSG00000253385.1 gene was demonstrated to play a definite role in the invasion of esophageal cancer immune cells and in signaling pathways in these cells. In vitro cell functional assays revealed that lncRNA ENSG00000253385.1 expression was elevated in the KYSE150 and KYSE410 esophageal cancer cell lines. Small interfering RNA (siRNA)-mediated silencing of lncRNA ENSG00000253385.1 significantly inhibited the proliferation, migration, and invasion of KYSE150 and KYSE410 cells, as well as promoted their apoptosis. CONCLUSIONS The ENSG00000253385.1 gene may be a key gene in the occurrence, development, and prognosis of esophageal cancer. These findings provide new ideas and references for the screening of therapeutic targets, as well as the development of targeted drugs, for esophageal cancer treatment.
Collapse
Affiliation(s)
| | - Ran Hou
- Departments of Medical Oncology, PR China
| | | | | | - Lie Liu
- Hebei Medical University, Shijiazhuang, Hebei 050000, PR China
| | - Huazhen Du
- Emergency department The Fourth Hospital of Hebei Medical University, Shijiazhuang, Hebei 050011, PR China.
| | - Jian Shi
- Departments of Medical Oncology, PR China.
| |
Collapse
|
2
|
Robinette FN, Valentine NP, Sehler KM, Medeck AM, Reynolds KE, Lane SN, Price AN, Cavanaugh IG, Shell SM, Ashford DL. Modulating Excited State Properties and Ligand Ejection Kinetics in Ruthenium Polypyridyl Complexes Designed to Mimic Photochemotherapeutics. Inorg Chem 2024; 63:8426-8439. [PMID: 38662617 DOI: 10.1021/acs.inorgchem.4c00922] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/07/2024]
Abstract
Ruthenium(II) polypyridyl complexes have gained significant interest as photochemotherapeutics (PCTs) due to their synthetic viability, strong light absorption, well understood excited state properties, and high phototoxicity indexes. Herein, we report the synthesis, characterization, electrochemical, spectrochemical, and preliminary cytotoxicity analyses of three series of ruthenium(II) polypyridyl complexes designed to mimic PCTs. The three series have the general structure of [Ru(bpy)2(N-N)]2+ (Series 1), [Ru(bpy)(dmb)(N-N)]2+ (Series 2), and [Ru(dmb)2(N-N)]2+ (Series 3, where N-N is a bidentate polypyridyl ligand, bpy = 2,2'-bipyridine, and dmb = 6,6'-dimethyl-2,2'-bipyridine). In the three series, the N-N ligand was systematically modified to incorporate increased conjugation and/or electronegative heteroatoms to increase dπ-π* backbonding, red-shifting the lowest energy metal-to-ligand charge transfer (MLCT) absorptions from λmax = 454 to λmax = 580 nm, nearing the therapeutic window for PCTs (600-1100 nm). In addition, steric bulk was systematically introduced through the series, distorting the Ru(II) octahedra, making the dissociative 3dd* state thermally accessible at room and body temperatures. This resulted in a 4 orders of magnitude increase in photoinduced ligand ejection kinetics, and demonstrates the ability to modulate both the MLCT* and dd* manifolds in the complexes, which is critical in PCT drug design. Preliminary cell viability assays suggest that the increased steric bulk to lower the 3dd* states may interfere with the cytotoxicity mechanism, limiting photoinitiated toxicity of the complexes. This work demonstrates the importance of understanding both the MLCT* and dd* manifolds and how they impact the ability of a complex to act as a PCT agent.
Collapse
Affiliation(s)
- Faith N Robinette
- Department of Natural Sciences, Tusculum University, Greeneville, Greeneville, Tennessee 37745, United States
| | - Nathaniel P Valentine
- Department of Natural Sciences, Tusculum University, Greeneville, Greeneville, Tennessee 37745, United States
| | - Konrad M Sehler
- Department of Natural Sciences, Tusculum University, Greeneville, Greeneville, Tennessee 37745, United States
| | - Andrew M Medeck
- Department of Natural Sciences, Tusculum University, Greeneville, Greeneville, Tennessee 37745, United States
| | - Keylon E Reynolds
- Department of Natural Sciences, Tusculum University, Greeneville, Greeneville, Tennessee 37745, United States
| | - Skylar N Lane
- Department of Natural Sciences, Tusculum University, Greeneville, Greeneville, Tennessee 37745, United States
| | - Averie N Price
- Department of Natural Sciences, Tusculum University, Greeneville, Greeneville, Tennessee 37745, United States
| | - Ireland G Cavanaugh
- Department of Natural Sciences, Tusculum University, Greeneville, Greeneville, Tennessee 37745, United States
| | - Steven M Shell
- Department of Natural Sciences, University of Virginia College at Wise, Wise, Virginia 24293, United States
| | - Dennis L Ashford
- Department of Natural Sciences, Tusculum University, Greeneville, Greeneville, Tennessee 37745, United States
| |
Collapse
|
3
|
Özaslan MS. Investigation of Potential Effects of Some Indole Compounds on the Glutathione S-Transferase Enzyme. BIOCHEMISTRY. BIOKHIMIIA 2024; 89:553-561. [PMID: 38648772 DOI: 10.1134/s0006297924030131] [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: 08/23/2023] [Revised: 02/23/2024] [Accepted: 02/27/2024] [Indexed: 04/25/2024]
Abstract
Glutathione S-transferases (GSTs) belong to the superfamily of multifunctional detoxification isoenzymes with an important role in cellular signaling. They can prevent reactive electrophilic compounds from harming the body by covalently binding identical type of moleculs to each other. GSTs can be used alone or in combination for cancer detection or diagnosis, in addition to therapeutic interventions. In recent years, indoles have become important due to their structural properties and biological activities such as antitubercular, antiulcer, anti-oxidant, and antidiabetic, as well as for the development of new anticancer agents. The current research investigated effects of some indoles with 3-carboxaldehyde structure on the GST enzyme activity. Impacts of various concentrations of indoles on the in vitro GST activity were examined. While IC50 values for the compounds ranged from 0.042 to 1.570 mM, Ki values changed between 0.018 ± 0.01 and 1.110 ± 0.15 mM. 6-Methylindole-3-carboxaldehyde (1b) exhibited the highest inhibitory effect among the indoles examined. Indole derivatives used in the study can be evaluated in further pharmacological studies due to their effects on GST activity.
Collapse
Affiliation(s)
- Muhammet Serhat Özaslan
- Department of Pharmacy Services, Nihat Delibalta Göle Vocational High School, Ardahan University, Ardahan, 75700, Turkey.
| |
Collapse
|
4
|
Li Z, Li X, Lu Y, Zhu X, Zheng W, Chen K, Liu S, Wu J, Guan W. Improved Photodynamic Therapy Based on Glutaminase Blockage via Tumor Membrane Coated CB-839/IR-780 Nanoparticles. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2024; 20:e2305174. [PMID: 37875654 DOI: 10.1002/smll.202305174] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/20/2023] [Revised: 10/12/2023] [Indexed: 10/26/2023]
Abstract
Photodynamic therapy (PDT) has promising applications. However, the lethal function of reactive oxygen species (ROS) produced during PDT is typically limited. This restriction is induced by oxygen shortage in the tumor microenvironment due to tumor cell hypermetabolism and reductive chemicals overexpression in tumor tissues. Glutamine (Gln) metabolism is crucial for malignancy development and is closely associated with redox. Herein, a novel nanoparticle (NP) named IRCB@M is constructed to boost PDT through dual effects. This NP simultaneously blocks aerobic respiration and inhibits cellular reduced substances by blocking the Gln metabolic pathway. Within the nanocomplex, a photosensitizer (IR-780) and a glutaminase inhibitor (CB-839) are self-assembled and then encapsulated by cancer cell membranes for homologous targeting. The Gln metabolism intervention relieves hypoxia and decreases the levels of nicotinamide adenine dinucleotide phosphate (NADPH) as well as reduced glutathione (GSH) in vitro and in vivo, which are the dual amplification effects on the IR-780-mediated lethal PDT. The antitumor effects against gastric cancer are ultimately evoked in vivo, thus offering a novel concept for enhancing PDT and other ROS-dependent therapeutic approaches.
Collapse
Affiliation(s)
- Zhiyan Li
- Division of Gastric Surgery, Department of General Surgery, Nanjing Drum Tower Hospital, the Affiliated Hospital of Medical School, Nanjing University, Nanjing, 210008, China
- State Key Laboratory of Pharmaceutical Biotechnology, Medical School, Nanjing University, Nanjing, 210093, China
| | - Xianghui Li
- Division of Gastric Surgery, Department of General Surgery, Nanjing Drum Tower Hospital, the Affiliated Hospital of Medical School, Nanjing University, Nanjing, 210008, China
- State Key Laboratory of Pharmaceutical Biotechnology, Medical School, Nanjing University, Nanjing, 210093, China
- Department of Dermatology, First Affiliated Hospital of Guangxi Medical University, Nanning, 530021, China
| | - Yanjun Lu
- Division of Gastric Surgery, Department of General Surgery, Nanjing Drum Tower Hospital, the Affiliated Hospital of Medical School, Nanjing University, Nanjing, 210008, China
| | - Xudong Zhu
- Division of Gastric Surgery, Department of General Surgery, Nanjing Drum Tower Hospital, the Affiliated Hospital of Medical School, Nanjing University, Nanjing, 210008, China
| | - Wenxuan Zheng
- Division of Gastric Surgery, Department of General Surgery, Nanjing Drum Tower Hospital, the Affiliated Hospital of Medical School, Nanjing University, Nanjing, 210008, China
| | - Kai Chen
- Division of Gastric Surgery, Department of General Surgery, Nanjing Drum Tower Hospital, the Affiliated Hospital of Medical School, Nanjing University, Nanjing, 210008, China
| | - Song Liu
- Division of Gastric Surgery, Department of General Surgery, Nanjing Drum Tower Hospital, the Affiliated Hospital of Medical School, Nanjing University, Nanjing, 210008, China
| | - Jinhui Wu
- State Key Laboratory of Pharmaceutical Biotechnology, Medical School, Nanjing University, Nanjing, 210093, China
- Chemistry and Biomedicine Innovation Center, Nanjing University, Nanjing, 210093, China
- Jiangsu Key Laboratory for Nano Technology, Nanjing University, Nanjing, 210093, China
| | - Wenxian Guan
- Division of Gastric Surgery, Department of General Surgery, Nanjing Drum Tower Hospital, the Affiliated Hospital of Medical School, Nanjing University, Nanjing, 210008, China
| |
Collapse
|
5
|
Fu Q, Yang X, Wang M, Zhu K, Wang Y, Song J. Activatable Probes for Ratiometric Imaging of Endogenous Biomarkers In Vivo. ACS NANO 2024; 18:3916-3968. [PMID: 38258800 DOI: 10.1021/acsnano.3c10659] [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] [Indexed: 01/24/2024]
Abstract
Dynamic variations in the concentration and abnormal distribution of endogenous biomarkers are strongly associated with multiple physiological and pathological states. Therefore, it is crucial to design imaging systems capable of real-time detection of dynamic changes in biomarkers for the accurate diagnosis and effective treatment of diseases. Recently, ratiometric imaging has emerged as a widely used technique for sensing and imaging of biomarkers due to its advantage of circumventing the limitations inherent to conventional intensity-dependent signal readout methods while also providing built-in self-calibration for signal correction. Here, the recent progress of ratiometric probes and their applications in sensing and imaging of biomarkers are outlined. Ratiometric probes are classified according to their imaging mechanisms, and ratiometric photoacoustic imaging, ratiometric optical imaging including photoluminescence imaging and self-luminescence imaging, ratiometric magnetic resonance imaging, and dual-modal ratiometric imaging are discussed. The applications of ratiometric probes in the sensing and imaging of biomarkers such as pH, reactive oxygen species (ROS), reactive nitrogen species (RNS), glutathione (GSH), gas molecules, enzymes, metal ions, and hypoxia are discussed in detail. Additionally, this Review presents an overview of challenges faced in this field along with future research directions.
Collapse
Affiliation(s)
- Qinrui Fu
- Institute for Translational Medicine, The Affiliated Hospital of Qingdao University, College of Medicine, Qingdao University, Qingdao, Shandong 266021, China
| | - Xiao Yang
- Institute for Translational Medicine, The Affiliated Hospital of Qingdao University, College of Medicine, Qingdao University, Qingdao, Shandong 266021, China
| | - Mengzhen Wang
- Institute for Translational Medicine, The Affiliated Hospital of Qingdao University, College of Medicine, Qingdao University, Qingdao, Shandong 266021, China
| | - Kang Zhu
- State Key Laboratory of Chemical Resource Engineering, Beijing University of Chemical Technology, Beijing, 100029, China
| | - Yin Wang
- Institute for Translational Medicine, The Affiliated Hospital of Qingdao University, College of Medicine, Qingdao University, Qingdao, Shandong 266021, China
| | - Jibin Song
- State Key Laboratory of Chemical Resource Engineering, Beijing University of Chemical Technology, Beijing, 100029, China
| |
Collapse
|
6
|
Kaimal R, Dube A, Souwaileh AA, Wu JJ, Anandan S. A copper metal-organic framework-based electrochemical sensor for identification of glutathione in pharmaceutical samples. Analyst 2024; 149:947-957. [PMID: 38197180 DOI: 10.1039/d3an01714a] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2024]
Abstract
The construction of a new electrochemical sensing platform based on a copper metal-organic framework (Cu-MOF) heterostructure is described in this paper. Drop-casting Cu-MOF suspension onto the electrode surface primed the sensor for glutathione detection. The composition and morphology of the Cu-MOF heterostructure were investigated using scanning electron microscopy (SEM), high-resolution transmission electron microscopy (HR-TEM), X-ray diffraction spectroscopy (XRD), Fourier transform infrared spectroscopy (FT-IR), and UV-visible spectroscopy. The Cu-MOF heterostructure can identify glutathione (GSH) with an enhanced sensitivity of 0.0437 μA μM-1 at the detection limit (LOD; 0.1 ± 0.005 μM) and a large dynamic range of 0.1-20 μM. Boosting the conductivity and surface area enhances electron transport and promotes redox processes. The constructed sensors were also adequately selective against interference from other contaminants in a similar potential window. Furthermore, the Cu-MOF heterostructure has outstanding selectivity, long-term stability, and repeatability, and the given sensors have demonstrated their capacity to detect GSH with high accuracy (recovery range = 98.2-100.8%) in pharmaceutical samples.
Collapse
Affiliation(s)
- Reshma Kaimal
- Nanomaterials & Solar Energy Conversion Lab, Department of Chemistry, National Institute of Technology, Tiruchirappalli-620015, India.
| | - Aashutosh Dube
- Nanomaterials & Solar Energy Conversion Lab, Department of Chemistry, National Institute of Technology, Tiruchirappalli-620015, India.
| | - Abdullah Al Souwaileh
- Department of Chemistry, College of Science, King Saud University, Riyadh 11451, Saudi Arabia
| | - Jerry J Wu
- Department of Environmental Engineering & Science, Feng Chia University, Taichung-407, Taiwan
| | - Sambandam Anandan
- Nanomaterials & Solar Energy Conversion Lab, Department of Chemistry, National Institute of Technology, Tiruchirappalli-620015, India.
| |
Collapse
|
7
|
Zhou Y, Tan F, Wang Z, Zhou G, Yuan C. The Pivotal Function of SLC16A1 and SLC16A1-AS1 in Cancer Progress: Molecular Pathogenesis and Prognosis. Mini Rev Med Chem 2024; 24:1685-1700. [PMID: 38616756 DOI: 10.2174/0113895575284780240327103039] [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: 10/27/2023] [Revised: 02/15/2024] [Accepted: 03/06/2024] [Indexed: 04/16/2024]
Abstract
More than 300 membranes make up the SLC family of transporters, utilizing an ion gradient or electrochemical potential difference to move their substrates across biological membranes. The SLC16 gene family contains fourteen members. Proton-linked transportation of monocarboxylates can be promoted by the transporters MCT1, which the SLC16A1 gene family encodes. Glycolysis is constitutively up-regulated in cancer cells, and the amount of lactate produced as a result is correlated with prognosis. Further speaking, SLC16A1 plays an essential role in controlling the growth and spread of tumors, according to mounting evidence. Additionally, LncRNAs are the collective term for all genes that produce RNA transcripts longer than 200 nucleotides but do not convert into proteins. It has steadily developed into a hub for research, offering an innovative approach to tumor study as technology related to molecular biology advances. The growing study has uncovered SLC16A1-AS1, an RNA that acts as an antisense to SLC16A1, which is erroneously expressed in various types of cancers. Therefore, we compiled the most recent information on the physiological functions and underlying processes of SLC16A1 and the LncRNA SLC16A1-AS1 during tumor development to explore their impact on cancer treatment and prognosis. We compiled the most recent information on the physiological functions and underlying processes of SLC16A1 and the LncRNA SLC16A1-AS1 during tumor development to explore their impact on cancer treatment and prognosis. Relevant studies were retrieved and collected through the PubMed system. After determining SLC16A1 and SLC16A1-AS1 as the research object, we found a close relationship between SLC16A1 and tumorigenesis as well as the influencing factors through the analysis of the research articles. SLC16A1 regulates lactate chemotaxis while uncovering SLC16A1- AS1 as an antisense RNA acting through multiple pathways; they affect the metabolism of tumor cells and have an impact on the prognosis of patients with various cancers.
Collapse
Affiliation(s)
- Yunxi Zhou
- Hubei Key Laboratory of Tumor Microenvironment and Immunotherapy, China Three Gorges University, Tichang 443002, China
- College of Basic Medical Science, China Three Gorges University, Yichang 443002, China
- Thirdgrade Pharmacological Laboratory on Traditional Chinese Medicine, State Administration of Traditional Chinese Medicine, China Three Gorges University, China
| | - Fangshun Tan
- Hubei Key Laboratory of Tumor Microenvironment and Immunotherapy, China Three Gorges University, Tichang 443002, China
- College of Basic Medical Science, China Three Gorges University, Yichang 443002, China
- Thirdgrade Pharmacological Laboratory on Traditional Chinese Medicine, State Administration of Traditional Chinese Medicine, China Three Gorges University, China
| | - Zhuowei Wang
- Hubei Key Laboratory of Tumor Microenvironment and Immunotherapy, China Three Gorges University, Tichang 443002, China
- College of Basic Medical Science, China Three Gorges University, Yichang 443002, China
- Thirdgrade Pharmacological Laboratory on Traditional Chinese Medicine, State Administration of Traditional Chinese Medicine, China Three Gorges University, China
| | - Gang Zhou
- College of Traditional Chinese Medicine, China Three Gorges University, Yichang, 443002, China
- Yichang Hospital of Traditional Chinese Medicine, Yichang, 443002, China
| | - Chengfu Yuan
- Hubei Key Laboratory of Tumor Microenvironment and Immunotherapy, China Three Gorges University, Tichang 443002, China
- College of Basic Medical Science, China Three Gorges University, Yichang 443002, China
- Thirdgrade Pharmacological Laboratory on Traditional Chinese Medicine, State Administration of Traditional Chinese Medicine, China Three Gorges University, China
| |
Collapse
|
8
|
Scott EN, Joseph AA, Dhanda A, Tanoshima R, Brooks B, Rassekh SR, Ross CJD, Carleton BC, Loucks CM. Systematic Critical Review of Genetic Factors Associated with Cisplatin-induced Ototoxicity: Canadian Pharmacogenomics Network for Drug Safety 2022 Update. Ther Drug Monit 2023; 45:714-730. [PMID: 37726872 DOI: 10.1097/ftd.0000000000001113] [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: 11/23/2022] [Accepted: 02/01/2023] [Indexed: 09/21/2023]
Abstract
BACKGROUND Cisplatin is commonly used to treat solid tumors; however, its use can be complicated by drug-induced hearing loss (ie, ototoxicity). The presence of certain genetic variants has been associated with the development/occurrence of cisplatin-induced ototoxicity, suggesting that genetic factors may be able to predict patients who are more likely to develop ototoxicity. The authors aimed to review genetic associations with cisplatin-induced ototoxicity and discuss their clinical relevance. METHODS An updated systematic review was conducted on behalf of the Canadian Pharmacogenomics Network for Drug Safety, based on the Preferred Reporting Items for Systematic reviews and Meta-Analyses 2020 statement. Pharmacogenomic studies that reported associations between genetic variation and cisplatin-induced ototoxicity were included. The evidence on genetic associations was summarized and evaluated, and knowledge gaps that can be used to inform future pharmacogenomic studies identified. RESULTS Overall, 40 evaluated reports, considering 47 independent patient populations, captured associations involving 24 genes. Considering GRADE criteria, genetic variants in 2 genes were strongly (ie, odds ratios ≥3) and consistently (ie, replication in ≥3 independent populations) predictive of cisplatin-induced ototoxicity. Specifically, an ACYP2 variant has been associated with ototoxicity in both children and adults, whereas TPMT variants are relevant in children. Encouraging evidence for associations involving several other genes also exists; however, further research is necessary to determine potential clinical relevance. CONCLUSIONS Genetic variation in ACYP2 and TPMT may be helpful in predicting patients at the highest risk of developing cisplatin-induced ototoxicity. Further research (including replication studies considering diverse pediatric and adult patient populations) is required to determine whether genetic variation in additional genes may help further identify patients most at risk.
Collapse
Affiliation(s)
- Erika N Scott
- BC Children's Hospital Research Institute, Vancouver, British Columbia, Canada
- Department of Medical Genetics, Faculty of Medicine, University of British Columbia (UBC), Vancouver, British Columbia, Canada
| | - Akshaya A Joseph
- BC Children's Hospital Research Institute, Vancouver, British Columbia, Canada
- Division of Translational Therapeutics, Department of Pediatrics, Faculty of Medicine, UBC, Vancouver, British Columbia, Canada
| | - Angie Dhanda
- BC Children's Hospital Research Institute, Vancouver, British Columbia, Canada
- Division of Translational Therapeutics, Department of Pediatrics, Faculty of Medicine, UBC, Vancouver, British Columbia, Canada
| | - Reo Tanoshima
- Department of Pediatrics, Yokohama City University Hospital, Yokohama, Japan
- YCU Center for Novel and Exploratory Clinical Trials, Yokohama City University Hospital, Yokohama, Japan
| | - Beth Brooks
- Audiology and Speech Pathology Department, British Columbia Children's Hospital, Vancouver, British Columbia, Canada
- School of Audiology and Speech Science, UBC, Vancouver, British Columbia, Canada
| | - S Rod Rassekh
- BC Children's Hospital Research Institute, Vancouver, British Columbia, Canada
- Division of Oncology, Hematology and Bone Marrow Transplant, British Columbia Children's Hospital and UBC, Vancouver, British Columbia, Canada
| | - Colin J D Ross
- BC Children's Hospital Research Institute, Vancouver, British Columbia, Canada
- Department of Medical Genetics, Faculty of Medicine, University of British Columbia (UBC), Vancouver, British Columbia, Canada
- Faculty of Pharmaceutical Sciences, UBC, Vancouver, British Columbia, Canada
| | - Bruce C Carleton
- BC Children's Hospital Research Institute, Vancouver, British Columbia, Canada
- Department of Medical Genetics, Faculty of Medicine, University of British Columbia (UBC), Vancouver, British Columbia, Canada
- Division of Translational Therapeutics, Department of Pediatrics, Faculty of Medicine, UBC, Vancouver, British Columbia, Canada
- Pharmaceutical Outcomes Programme, British Columbia Children's Hospital, Vancouver, British Columbia, Canada; and
| | - Catrina M Loucks
- BC Children's Hospital Research Institute, Vancouver, British Columbia, Canada
- Division of Translational Therapeutics, Department of Pediatrics, Faculty of Medicine, UBC, Vancouver, British Columbia, Canada
- Department of Anesthesiology, Pharmacology & Therapeutics, Faculty of Medicine, UBC, Vancouver, British Columbia, Canada
| |
Collapse
|
9
|
Xu J, Zhang J, Song J, Liu Y, Li J, Wang X, Tang R. Construction of multifunctional mesoporous silicon nano-drug delivery system and study of dual sensitization of chemo-photodynamic therapy in vitro and in vivo. J Colloid Interface Sci 2022; 628:271-285. [PMID: 35998453 DOI: 10.1016/j.jcis.2022.08.100] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2022] [Revised: 08/12/2022] [Accepted: 08/15/2022] [Indexed: 11/28/2022]
Abstract
This study was conducted to construct a multifunctional nanodrug delivery system (NDDS) to deplete glutathione (GSH) in tumor cells and amplify oxidative stress, enhancing the synergistic effect of chemotherapy and photodynamic therapy (PDT). l-Buthionine-sulfoximine (BSO) and chlorin e6 (Ce6) were loaded into mesoporous silicon nanoparticles (MSN), and then MSN were modified with oxidized hyaluronic acid (OHA) as a pore-blocking agent. Cisplatin (Pt(II)) was further loaded by a coordination reaction with carboxyl groups in OHA to yield a multifunctional NDDS (denoted as MSN@OHA-Ce6/BSO/Pt). The physicochemical properties and antitumor activity of the prepared nanoparticles were characterized in detail. In vitro and in vivo experiments demonstrated that OHA was shed from MSN@OHA-Ce6/BSO/Pt under acidic conditions in tumors, resulting in the release of free BSO, Ce6, and Pt(II). The released BSO could reduce intracellular GSH expression by 48.8 %, effectively enhancing the PDT effect of Ce6 and the chemotherapy effect of Pt(II). Finally, the tumor inhibitory rate (vs saline) reached 73.8 % ± 2.5 % for MSN@OHA-Ce6/BSO/Pt in A549/DDP tumor-bearing nude mice. Therefore, the multifunctional NDDS significantly enhanced the synergistic effect of PDT and chemotherapy.
Collapse
Affiliation(s)
- Jiaxi Xu
- Engineering Research Center for Biomedical Materials, Anhui Key Laboratory of Modern Biomanufacturing, School of Life Sciences, Anhui University, 111 Jiulong Road, Hefei, Anhui Province 230601, PR China
| | - Jingwen Zhang
- Engineering Research Center for Biomedical Materials, Anhui Key Laboratory of Modern Biomanufacturing, School of Life Sciences, Anhui University, 111 Jiulong Road, Hefei, Anhui Province 230601, PR China
| | - Jinping Song
- Engineering Research Center for Biomedical Materials, Anhui Key Laboratory of Modern Biomanufacturing, School of Life Sciences, Anhui University, 111 Jiulong Road, Hefei, Anhui Province 230601, PR China
| | - Yufei Liu
- Engineering Research Center for Biomedical Materials, Anhui Key Laboratory of Modern Biomanufacturing, School of Life Sciences, Anhui University, 111 Jiulong Road, Hefei, Anhui Province 230601, PR China
| | - Junnan Li
- Engineering Research Center for Biomedical Materials, Anhui Key Laboratory of Modern Biomanufacturing, School of Life Sciences, Anhui University, 111 Jiulong Road, Hefei, Anhui Province 230601, PR China
| | - Xin Wang
- Engineering Research Center for Biomedical Materials, Anhui Key Laboratory of Modern Biomanufacturing, School of Life Sciences, Anhui University, 111 Jiulong Road, Hefei, Anhui Province 230601, PR China.
| | - Rupei Tang
- Engineering Research Center for Biomedical Materials, Anhui Key Laboratory of Modern Biomanufacturing, School of Life Sciences, Anhui University, 111 Jiulong Road, Hefei, Anhui Province 230601, PR China.
| |
Collapse
|
10
|
Lin C, Chen Y, Zhu M, Pei J, Zhou Y, Gou M, Ouyang L. A sulfhydryl blocking reagent BT-4 sensitizes cisplatin-based micelle prodrugs for efficient treatment of breast cancer. Int J Pharm 2022; 626:122187. [PMID: 36100145 DOI: 10.1016/j.ijpharm.2022.122187] [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: 05/01/2022] [Revised: 09/01/2022] [Accepted: 09/05/2022] [Indexed: 10/14/2022]
Abstract
Detoxification of glutathione (GSH) and insufficient cellular uptake of cisplatin (CDDP) severely compromised the therapeutic efficacy of CDDP. Here, a nano-delivery system (BT-4@PtPPNPs) for CDDP prodrug (C16-Pt(Ⅳ)-PEG) based on a novel sulfhydryl blocking reagent methyl 2-(methylsulfonyl) benzothiazole-6-carboxylate (BT-4) was developed. On the one hand, BT-4 can deplete GSH in tumor cells by directly interacting with reactive sulfhydryl group on GSH, thereby increasing the cytotoxicity of CDDP. On the other hand, the CDDP prodrug carrier C16-Pt(IV)-PEG can promote the distribution of CDDP in tumors, reduce the probability of unexpected inactivation of CDDP, and reduce the content of GSH in tumor cells during the conversion to CDDP, thereby making CDDP more effective for treatment. The results showed that the optimized BT-4@PtPPNPs with a small particle size (130 nm) exhibited notable cytotoxicity and apoptosis of 4T1 cells. BT-4@PtPPNPs not only significantly improved the uptake of drugs by tumor cells, but also rapidly targeted and accumulated in the tumors for a long time. Moreover, in vivo efficacy studies showed that BT-4@PtPPNPs could effectively inhibit tumor growth, inhibiting 60.85 % of tumors in a 4T1 breast cancer mice model, showing superior antitumor activity, which can be attributed to GSH-triggered CDDP tolerance reversal. Overall, this study provides an attractive and simple strategy to combine novel sulfhydryl blockers and CDDP prodrugs to potentiate the efficacy of CDDP in breast cancer.
Collapse
Affiliation(s)
- Congcong Lin
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Collaborative Innovation Center for Biotherapy, Chengdu 610041, China; Department of Medicinal Chemistry and Natural Medicine Chemistry, College of Pharmacy, Harbin Medical University, Harbin 150081, China.
| | - Yuxiu Chen
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Collaborative Innovation Center for Biotherapy, Chengdu 610041, China.
| | - Mengli Zhu
- Core Facilities of West China Hospital, Chengdu 610041, China.
| | - Junping Pei
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Collaborative Innovation Center for Biotherapy, Chengdu 610041, China.
| | - Yang Zhou
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Collaborative Innovation Center for Biotherapy, Chengdu 610041, China.
| | - Maling Gou
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Collaborative Innovation Center for Biotherapy, Chengdu 610041, China.
| | - Liang Ouyang
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Collaborative Innovation Center for Biotherapy, Chengdu 610041, China.
| |
Collapse
|
11
|
Korkmaz IN, Özdemir H. Synthesis and Anticancer Potential of New Hydroxamic Acid Derivatives as Chemotherapeutic Agents. Appl Biochem Biotechnol 2022; 194:6349-6366. [PMID: 35917102 DOI: 10.1007/s12010-022-04107-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 07/15/2022] [Indexed: 11/25/2022]
Abstract
Histone deacetylase (HDAC) inhibitors have been shown to induce differentiation, cell cycle arrest, and apoptosis due to their low toxicity, inhibiting migration, invasion, and angiogenesis in many cancer cells. Studies show that hydroxamic acids are generally used as anticancers. For this reason, it is aimed to synthesize new derivatives of hydroxamic acids, to examine the anticancer properties of these candidate inhibitors, and to investigate the inhibition effects on some enzymes that cause multidrug resistance in cancer cells. For this reason, new (4-amino-2-methoxy benzohydroxamic acid (a), 4-amino-3-methyl benzohydroxamic acid (b), 3-amino-5-methyl benzohydroxamic acid (c)) amino benzohydroxamic acid derivatives were synthesized in this study. The effects on healthy fibroblast, lung (A549), and cervical (HeLa) cancer cells were investigated. In addition, their effects on TRXR1, GST, and GR activities, which are important for the development of chemotherapeutic strategies, were also examined. It was determined that molecule b was the most effective molecule in HeLa cancer cells with the lowest IC50 value of 0.54. It was determined that molecule c was the most effective molecules for A549 and HeLa cancer cells, with the lowest IC50 values of 0.78 mM and 0.25 mM, respectively. It was determined that b and c molecules directed cancer cells to necrosis rather than apoptosis. c molecule showed anticancer effect in A549 and HeLa cancer cells. It was found that molecule c significantly suppressed both GR and TRXR1 activities. In GST activities, however, inhibitors did not have a significant effect on cancer cells.
Collapse
Affiliation(s)
- Işıl Nihan Korkmaz
- Faculty of Science, Department of Chemistry, Atatürk University, Erzurum, 25240, Turkey
| | - Hasan Özdemir
- Faculty of Science, Department of Chemistry, Atatürk University, Erzurum, 25240, Turkey.
| |
Collapse
|
12
|
Saini AK, Sahoo SK. Fluorescent pH sensing and MnO2 nanosphere directed turn-on sensing of glutathione using pyridoxal 5′-phosphate modified polydopamine nanoparticles. INORG CHEM COMMUN 2022. [DOI: 10.1016/j.inoche.2022.109677] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
|
13
|
McCullough AB, Chen J, Valentine NP, Franklin TM, Cantrell AP, Darnell VM, Qureshi Q, Hanson K, Shell SM, Ashford DL. Balancing the interplay between ligand ejection and therapeutic window light absorption in ruthenium polypyridyl complexes. Dalton Trans 2022; 51:10186-10197. [PMID: 35735218 DOI: 10.1039/d2dt01237e] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
Ruthenium polypyridyl complexes have gained significant interest as photochemotherapies (PCTs) where their excited-state properties play a critical role in the photo-cytotoxicity mechanism and efficacy. Herein we report a systematic electrochemical, spectrochemical, and photophysical analysis of a series of ruthenium(II) polypyridyl complexes of the type [Ru(bpy)2(N-N)]2+ (where bpy = 2,2'-bipyridine; N-N is a bidentate polypyridyl ligand) designed to mimic PCTs. In this series, the N-N ligand was modified through increased conjugation and/or incorporation of electronegative heteroatoms to shift the metal-to-ligand charge-transfer (MLCT) absorptions near the therapeutic window for PCTs (600-1100 nm) while incorporating steric bulk to trigger photoinduced ligand dissociation. The lowest energy MLCT absorptions were red-shifted from λmax = 454 nm to 564 nm, with emission energies decreasing from λmax = 620 nm to 850 nm. Photoinduced ligand ejection and temperature-dependent emission studies revealed an important interplay between red-shifting MLCT absorptions and accessing the dissociative 3dd* states, with energy barriers between the 3MLCT* and 3dd* states ranging from 850 cm-1 to 2580 cm-1 for the complexes measured. This work demonstrates the importance of understanding both the MLCT manifold and 3dd* state energy levels in the future design of ligands and complexes for PCT.
Collapse
Affiliation(s)
- Annie B McCullough
- Department of Natural Sciences, Tusculum University, Greeneville, Tennessee, 37745, USA.
| | - Jiaqi Chen
- Department of Chemistry & Biochemistry, Florida State University, Tallahassee, Florida 32306, USA
| | - Nathaniel P Valentine
- Department of Natural Sciences, Tusculum University, Greeneville, Tennessee, 37745, USA.
| | - Toney M Franklin
- Department of Natural Sciences, Tusculum University, Greeneville, Tennessee, 37745, USA.
| | - Andrew P Cantrell
- Department of Natural Sciences, Tusculum University, Greeneville, Tennessee, 37745, USA.
| | - Vayda M Darnell
- Department of Natural Sciences, Tusculum University, Greeneville, Tennessee, 37745, USA.
| | - Qasim Qureshi
- Department of Natural Sciences, University of Virginia's College at Wise, Wise, Virginia, 24293, USA
| | - Kenneth Hanson
- Department of Chemistry & Biochemistry, Florida State University, Tallahassee, Florida 32306, USA
| | - Steven M Shell
- Department of Natural Sciences, University of Virginia's College at Wise, Wise, Virginia, 24293, USA
| | - Dennis L Ashford
- Department of Natural Sciences, Tusculum University, Greeneville, Tennessee, 37745, USA.
| |
Collapse
|
14
|
Wang S, Chen H, Yang H, Zhou K, Bai F, Wu X, Xu H. Gut Microbiome Was Highly Related to the Regulation of Metabolism in Lung Adenocarcinoma Patients. Front Oncol 2022; 12:790467. [PMID: 35592677 PMCID: PMC9113755 DOI: 10.3389/fonc.2022.790467] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2021] [Accepted: 03/21/2022] [Indexed: 11/27/2022] Open
Abstract
Background Lung adenocarcinoma (LUAD) is one of the most predominant subtypes of lung cancer. The gut microbiome plays a vital role in the pathophysiological processes of various diseases, including cancers. Methods In the study, 100 individuals were enrolled. In total 75 stool and blood samples were analyzed with 16s-rRNA gene sequencing and metabolomics (30 from healthy individuals (H); 45 from LUAD patients). In addition, 25 stool samples were analyzed with metagenomics (10 from H; 15 from LUAD). The linear discriminant analysis (LDA) effect size (LefSe) and logistic regression analysis were applied to identify biomarkers’ taxa and develop a diagnostic model. The diagnostic power of the model was estimated with the receiver operating characteristic curve (ROC) by comparing the area under the ROC (AUC). The correlation between biomarker’s taxa and metabolites was calculated using the Spearman analysis. Results The α and β diversity demonstrated the composition and structure of the gut microbiome in LUAD patients were different from those in healthy people. The top three abundance of genera were Bacteroides (25.06%), Faecalibacterium (11.00%), and Prevotella (5.94%). The LefSe and logistic regression analysis identified three biomarker taxa (Bacteroides, Pseudomonas, and Ruminococcus gnavus group) and constructed a diagnostic model. The AUCs of the diagnostic model in 16s-rRNA gene sequencing and metagenomics were 0.852 and 0.841, respectively. A total of 102 plasma metabolites were highly related to those three biomarkers’ taxa. Seven metabolic pathways were enriched by 102 plasma metabolites, including the Pentose phosphate pathway, Glutathione metabolism. Conclusions In LUAD patients, the gut microbiome profile has significantly changed. We used three biomarkers taxa to develop a diagnostic model, which was accurate and suitable for the diagnosis of LUAD. Gut microbes, especially those three biomarkers’ taxa, may participate in regulating metabolism-related pathways in LUAD patients, such as the pentose phosphate pathway and glutathione metabolism.
Collapse
Affiliation(s)
- Sheng Wang
- Department of Respiratory, Jinhua Guangfu Hospital, Jinhua, China
| | - Huachun Chen
- Department of Respiratory, Jinhua Guangfu Hospital, Jinhua, China
| | - Huizhen Yang
- Department of Respiratory, Jinhua Guangfu Hospital, Jinhua, China
| | - Kejin Zhou
- Department of Respiratory, Jinhua Guangfu Hospital, Jinhua, China
| | - Fan Bai
- Department of Respiratory, Jinhua Guangfu Hospital, Jinhua, China
| | - Xiaoyu Wu
- Department of Respiratory, Jinhua Guangfu Hospital, Jinhua, China
| | - Hanwen Xu
- Department of Respiratory, Jinhua Guangfu Hospital, Jinhua, China
| |
Collapse
|
15
|
Sengupta B, Biswas P, Roy D, Lovett J, Simington L, Fry DR, Travis K. Anticancer Properties of Kaempferol on Cellular Signaling Pathways. Curr Top Med Chem 2022; 22:2474-2482. [PMID: 36082856 PMCID: PMC10754212 DOI: 10.2174/1568026622666220907112822] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2022] [Revised: 08/01/2022] [Accepted: 08/12/2022] [Indexed: 01/20/2023]
Abstract
Polyhydroxy compounds are secondary metabolites that are ubiquitous in plants of higher genera. They possess therapeutic properties against a wide spectrum of diseases, including cancers, neurodegenerative disorders, atherosclerosis, as well as cardiovascular disease. The phytochemical flavonol (a type of flavonoid) kaempferol (KMP) (3,5,7-trihydroxy-2-(4-hydroxyphenyl)- 4Hchromen-4-one) is abundant in cruciferous vegetables, including broccoli, kale, spinach, and watercress, as well as in herbs like dill, chives, and tarragon. KMP is predominantly hydrophobic in nature due to its diphenylpropane structure (a characteristic feature of flavonoids). Recent findings have indicated the promise of applying KMP in disease prevention due to its potential antioxidant, antimutagenic, antifungal, and antiviral activities. In the literature, there is evidence that KMP exerts its anticancer effects by modulating critical elements in cellular signal transduction pathways linked to apoptosis, inflammation, angiogenesis, and metastasis in cancer cells without affecting the viability of normal cells. It has been shown that KMP triggers cancer cell death by several mechanisms, including cell cycle arrest, caspase activation, metabolic alteration, and impacting human telomerase reverse-transcriptase gene expression. This review is aimed at providing critical insights into the influence of KMP on the intracellular cascades that regulate metabolism and signaling in breast, ovarian, and cervical cancer cells.
Collapse
Affiliation(s)
- Bidisha Sengupta
- Department of Chemistry and Biochemistry, Stephen F. Austin State University, Nacogdoches, Texas, USA
| | - Pragnya Biswas
- School of Biotechnology, Presidency University, Kolkata, India
| | - Debarshi Roy
- Department of Biological Sciences, Alcorn State University, Lorman, Mississippi, USA
| | - Justin Lovett
- Department of Chemistry and Biochemistry, Stephen F. Austin State University, Nacogdoches, Texas, USA
| | - Laken Simington
- Department of Chemistry and Biochemistry, Stephen F. Austin State University, Nacogdoches, Texas, USA
| | - Darrell R. Fry
- Department of Chemistry and Biochemistry, Stephen F. Austin State University, Nacogdoches, Texas, USA
| | - Kaelin Travis
- Center of Biotechnology, Alcorn State University, Lorman, Mississippi, USA
| |
Collapse
|
16
|
Cheng X, Xu HD, Ran HH, Liang G, Wu FG. Glutathione-Depleting Nanomedicines for Synergistic Cancer Therapy. ACS NANO 2021; 15:8039-8068. [PMID: 33974797 DOI: 10.1021/acsnano.1c00498] [Citation(s) in RCA: 192] [Impact Index Per Article: 48.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/12/2023]
Abstract
Cancer cells frequently exhibit resistance to various molecular and nanoscale drugs, which inevitably affects the drugs' therapeutic outcomes. Overexpression of glutathione (GSH) has been observed in many cancer cells, and solid evidence has corroborated the resulting tumor resistance to a variety of anticancer therapies, suggesting that this biochemical characteristic of cancer cells can be developed as a potential target for cancer treatments. The single treatment of GSH-depleting agents can potentiate the responses of the cancer cells to different cell death stimuli; therefore, as an adjunctive strategy, GSH depletion is usually combined with mainstream cancer therapies for enhancing the therapeutic outcomes. Propelled by the rapid development of nanotechnology, GSH-depleting agents can be readily constructed into anticancer nanomedicines, which have shown a steep rise over the past decade. Here, we review the common GSH-depleting nanomedicines which have been widely applied in synergistic cancer treatments in recent years. Some current challenges and future perspectives for GSH depletion-based cancer therapies are also presented. With the understanding of the structure-property relationship and action mechanisms of these biomaterials, we hope that the GSH-depleting nanotechnology will be further developed to realize more effective disease treatments and even achieve successful clinical translations.
Collapse
Affiliation(s)
- Xiaotong Cheng
- State Key Laboratory of Bioelectronics, School of Biological Science and Medical Engineering, Southeast University, 2 Sipailou Road, Nanjing 210096, P.R. China
| | - Hai-Dong Xu
- State Key Laboratory of Bioelectronics, School of Biological Science and Medical Engineering, Southeast University, 2 Sipailou Road, Nanjing 210096, P.R. China
| | - Huan-Huan Ran
- State Key Laboratory of Bioelectronics, School of Biological Science and Medical Engineering, Southeast University, 2 Sipailou Road, Nanjing 210096, P.R. China
| | - Gaolin Liang
- State Key Laboratory of Bioelectronics, School of Biological Science and Medical Engineering, Southeast University, 2 Sipailou Road, Nanjing 210096, P.R. China
| | - Fu-Gen Wu
- State Key Laboratory of Bioelectronics, School of Biological Science and Medical Engineering, Southeast University, 2 Sipailou Road, Nanjing 210096, P.R. China
| |
Collapse
|
17
|
The metabolic reprogramming in acute myeloid leukemia patients depends on their genotype and is a prognostic marker. Blood Adv 2021; 5:156-166. [PMID: 33570627 DOI: 10.1182/bloodadvances.2020002981] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2020] [Accepted: 11/12/2020] [Indexed: 12/19/2022] Open
Abstract
Leukemic cells display some alterations in metabolic pathways, which play a role in leukemogenesis and in patients' prognosis. To evaluate the characteristics and the impact of this metabolic reprogramming, we explore the bone marrow samples from 54 de novo acute myeloid leukemia (AML) patients, using an untargeted metabolomics approach based on proton high-resolution magic angle spinning-nuclear magnetic resonance. The spectra obtained were subjected to multivariate statistical analysis to find specific metabolome alterations and biomarkers correlated to clinical features. We found that patients display a large diversity of metabolic profiles, according to the different AML cytologic subtypes and molecular statuses. The link between metabolism and molecular status was particularly strong for the oncometabolite 2-hydroxyglutarate (2-HG), whose intracellular production is directly linked to the presence of isocitrate dehydrogenase mutations. Moreover, patients' prognosis was strongly impacted by several metabolites, such as 2-HG that appeared as a good prognostic biomarker in our cohort. Conversely, deregulations in phospholipid metabolism had a negative impact on prognosis through 2 main metabolites (phosphocholine and phosphoethanolamine), which could be potential aggressiveness biomarkers. Finally, we highlighted an overexpression of glutathione and alanine in chemoresistant patients. Overall, our results demonstrate that different metabolic pathways could be activated in leukemic cells according to their phenotype and maturation levels. This confirms that metabolic reprogramming strongly influences prognosis of patients and underscores a particular role of certain metabolites and associated pathways in AML prognosis, suggesting common mechanisms developed by leukemic cells to maintain their aggressiveness even after well-conducted induction chemotherapy.
Collapse
|
18
|
Głowacki S, Synowiec E, Szwed M, Toma M, Skorski T, Śliwiński T. Relationship between Oxidative Stress and Imatinib Resistance in Model Chronic Myeloid Leukemia Cells. Biomolecules 2021; 11:biom11040610. [PMID: 33924068 PMCID: PMC8074285 DOI: 10.3390/biom11040610] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2021] [Revised: 04/18/2021] [Accepted: 04/19/2021] [Indexed: 01/19/2023] Open
Abstract
Chronic myeloid leukemia (CML) develops due to the presence of the BCR-ABL1 protein, a target of tyrosine kinase inhibitors (TKIs), such as imatinib (IM), used in a CML therapy. CML eradication is a challenge due to developing resistance to TKIs. BCR-ABL1 induces endogenous oxidative stress leading to genomic instability and development of TKI resistance. Model CML cells susceptible or resistant to IM, as well as wild-type, non-cancer cells without the BCR-ABL1 protein were treated with IM, hydrogen peroxide (H2O2) as a model trigger of external oxidative stress, or with IM+H2O2. Accumulation of reactive oxygen species (ROS), DNA damage, activity of selected antioxidant enzymes and glutathione (GSH), and mitochondrial potential (MMP) were assessed. We observed increase in ROS accumulation in BCR-ABL1 positive cells and distinct levels of ROS accumulation in IM-susceptible cells when compared to IM-resistant ones, as well as increased DNA damage caused by IM action in sensitive cells. Depletion of GSH levels and a decreased activity of glutathione peroxidase (GPx) in the presence of IM was higher in the cells susceptible to IM. IM-resistant cells showed an increase of catalase activity and a depletion of MMP. BCR-ABL1 kinase alters ROS metabolism, and IM resistance is accompanied by the changes in activity of GPx, catalase, and alterations in MMP.
Collapse
MESH Headings
- Animals
- Antineoplastic Agents/toxicity
- Catalase/metabolism
- Cell Line, Tumor
- DNA Damage
- Drug Resistance, Neoplasm
- Fusion Proteins, bcr-abl/genetics
- Glutathione/metabolism
- Glutathione Peroxidase/metabolism
- Imatinib Mesylate/toxicity
- Leukemia, Myelogenous, Chronic, BCR-ABL Positive/genetics
- Leukemia, Myelogenous, Chronic, BCR-ABL Positive/metabolism
- Membrane Potential, Mitochondrial
- Mice
- Oxidative Stress
Collapse
Affiliation(s)
- Sylwester Głowacki
- Laboratory of Medical Genetics, Faculty of Biology and Environmental Protection, University of Lodz, Pomorska 141/143 Street, 90-236 Lodz, Poland; (S.G.); (E.S.); (M.T.)
| | - Ewelina Synowiec
- Laboratory of Medical Genetics, Faculty of Biology and Environmental Protection, University of Lodz, Pomorska 141/143 Street, 90-236 Lodz, Poland; (S.G.); (E.S.); (M.T.)
| | - Marzena Szwed
- Department of Medical Biophysics, Faculty of Biology and Environmental Protection, University of Lodz, Pomorska 141/143 Street, 90-236 Lodz, Poland;
| | - Monika Toma
- Laboratory of Medical Genetics, Faculty of Biology and Environmental Protection, University of Lodz, Pomorska 141/143 Street, 90-236 Lodz, Poland; (S.G.); (E.S.); (M.T.)
| | - Tomasz Skorski
- Fels Cancer Institute for Personalized Medicine, Temple University Lewis Katz School of Medicine, Philadelphia, PA 19140, USA;
| | - Tomasz Śliwiński
- Laboratory of Medical Genetics, Faculty of Biology and Environmental Protection, University of Lodz, Pomorska 141/143 Street, 90-236 Lodz, Poland; (S.G.); (E.S.); (M.T.)
- Correspondence:
| |
Collapse
|
19
|
Integrated multi-omics uncovers reliable potential biomarkers and adverse effects of zinc deficiency. Clin Nutr 2021; 40:2683-2696. [PMID: 33933734 DOI: 10.1016/j.clnu.2021.03.019] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2020] [Revised: 01/18/2021] [Accepted: 03/12/2021] [Indexed: 11/24/2022]
Abstract
BACKGROUND Zinc deficiency is a worldwide public health problem. Currently, there are no established biomarkers available for the accurate diagnosis of zinc-deficiency in individuals. Additionally, a comprehensive view of the adverse effects of zinc deficiency is lacking. Our aim was to identify superior biomarkers of zinc deficiency and uncover the adverse effects of zinc deficiency. METHODS We performed multi-omics analysis using serum proteomics-metabolomics and liver proteomics on zinc-deficient rats to identify candidate biomarkers and reveal the associated adverse effects of zinc deficiency. Secondly, the candidate biomarkers were validated in two zinc-deficient populations and an RCT zinc supplementation trial on a zinc-deficient population. RESULTS Our integrated multi-omics approach revealed numerous biomarkers (>2000) and glutathione metabolism as the most important changed pathway in zinc deficiency. Three candidate biomarkers from glutathione metabolism were validated in repeated zinc-deficient rats by quantitative analysis. Only glutathione sulfotransferase omega-1 (GSTO1) (among 3 candidate biomarkers) was validated in the two zinc-deficient populations and zinc-supplemented population. Compared with serum zinc, serum GSTO1 yielded a better response to zinc supplementation and a higher correlation coefficient with zinc intake and the AUC value and has the potential for diagnosing zinc deficiency. By integrated multi-omics, we identified both established and novel adverse effects of zinc deficiency. CONCLUSIONS Our integrated multi-omics analysis revealed more complete information about zinc deficiency; GSTO1 was found to be a reliable potential biomarker for diagnosis of zinc deficiency. This trial is registered at http://www.chictr.org.cn/registry.aspx as ChiCTR1900028162.
Collapse
|
20
|
Raju B, Choudhary S, Narendra G, Verma H, Silakari O. Molecular modeling approaches to address drug-metabolizing enzymes (DMEs) mediated chemoresistance: a review. Drug Metab Rev 2021; 53:45-75. [PMID: 33535824 DOI: 10.1080/03602532.2021.1874406] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
Resistance against clinically approved anticancer drugs is the main roadblock in cancer treatment. Drug metabolizing enzymes (DMEs) that are capable of metabolizing a variety of xenobiotic get overexpressed in malignant cells, therefore, catalyzing drug inactivation. As evident from the literature reports, the levels of DMEs increase in cancer cells that ultimately lead to drug inactivation followed by drug resistance. To puzzle out this issue, several strategies inclusive of analog designing, prodrug designing, and inhibitor designing have been forged. On that front, the implementation of computational tools can be considered a fascinating approach to address the problem of chemoresistance. Various research groups have adopted different molecular modeling tools for the investigation of DMEs mediated toxicity problems. However, the utilization of these in-silico tools in maneuvering the DME mediated chemoresistance is least considered and yet to be explored. These tools can be employed in the designing of such chemotherapeutic agents that are devoid of the resistance problem. The current review canvasses various molecular modeling approaches that can be implemented to address this issue. Special focus was laid on the development of specific inhibitors of DMEs. Additionally, the strategies to bypass the DMEs mediated drug metabolism were also contemplated in this report that includes analogs and pro-drugs designing. Different strategies discussed in the review will be beneficial in designing novel chemotherapeutic agents that depreciate the resistance problem.
Collapse
Affiliation(s)
- Baddipadige Raju
- Molecular Modeling Lab (MML), Department of Pharmaceutical Sciences and Drug Research, Punjabi University, Patiala, India
| | - Shalki Choudhary
- Molecular Modeling Lab (MML), Department of Pharmaceutical Sciences and Drug Research, Punjabi University, Patiala, India
| | - Gera Narendra
- Molecular Modeling Lab (MML), Department of Pharmaceutical Sciences and Drug Research, Punjabi University, Patiala, India
| | - Himanshu Verma
- Molecular Modeling Lab (MML), Department of Pharmaceutical Sciences and Drug Research, Punjabi University, Patiala, India
| | - Om Silakari
- Molecular Modeling Lab (MML), Department of Pharmaceutical Sciences and Drug Research, Punjabi University, Patiala, India
| |
Collapse
|
21
|
Bravo-Cuellar A, Ortiz-Lazareno PC, Sierra-Díaz E, Solorzano-Ibarra F, Méndez-Clemente AS, Aguilar-Lemarroy A, Jave-Suárez LF, Ruiz Velazco-Niño É, Hernández-Flores G. Pentoxifylline Sensitizes Cisplatin-Resistant Human Cervical Cancer Cells to Cisplatin Treatment: Involvement of Mitochondrial and NF-Kappa B Pathways. Front Oncol 2020; 10:592706. [PMID: 33680921 PMCID: PMC7931705 DOI: 10.3389/fonc.2020.592706] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2020] [Accepted: 10/30/2020] [Indexed: 12/24/2022] Open
Abstract
Background Cervical cancer continues to be a major public health problem worldwide, and Cisplatin is used as first-line chemotherapy for this cancer; however, malignant cells exposed to CISplatin (CIS) become insensitive to the effects of this drug. PenToXifylline (PTX) is a xanthine that sensitizes several types of tumor cells to apoptosis induced by antitumor drugs, such as Adriamycin, Carboplatin, and CIS. The effects of PTX on tumor cells have been related to the disruption of the NF-κB pathway, thus preventing the activation of cell survival mechanisms such as the expression of anti-apoptotic genes, the secretion of proinflammatory interleukins, and growth factors. Objective In this work, we studied the antitumor proprieties of PTX in human SiHa cervical carcinoma cells resistant to CIS. Materials and Methods SiHa and HeLa cervical cancer cells and their CIS-resistant derived cell lines (SiHaCIS-R and HeLaCIS-R, respectively) were used as in-vitro models. We studied the effects of PTX alone or in combination with CIS on cell viability, apoptosis, caspase-3, caspase-8, and caspase-9 activity, cleaved PARP-1, anti-apoptotic protein (Bcl-2 and Bcl-xL) levels, p65 phosphorylation, cadmium chloride (CdCl2) sensitivity, Platinum (Pt) accumulation, and glutathione (GSH) levels, as well as on the gene expression of GSH and drug transporters (influx and efflux). Results PTX sensitized SiHaCIS-R cells to the effects of CIS by inducing apoptosis, caspase activation, and PARP-1 cleavage. PTX treatment also decreased p65 phosphorylation, increased Pt levels, depleted GSH, and downregulated the expression of the ATP7A, ATP7B, GSR, and MGST1 genes. Conclusion PTX reverses the acquired phenotype of CIS resistance close to the sensitivity of parental SiHa cells.
Collapse
Affiliation(s)
- Alejandro Bravo-Cuellar
- División de Inmunología, Centro de Investigación Biomédica de Occidente (CIBO), Instituto Mexicano del Seguro Social (IMSS), Guadalajara, Mexico.,Departamento de Ciencias de la Salud, Centro Universitario de los Altos, Universidad de Guadalajara, Tepatitlán de Morelos, Mexico
| | - Pablo Cesar Ortiz-Lazareno
- División de Inmunología, Centro de Investigación Biomédica de Occidente (CIBO), Instituto Mexicano del Seguro Social (IMSS), Guadalajara, Mexico
| | - Erick Sierra-Díaz
- Departamento de Urología, Hospital de Especialidades, Centro Médico Nacional de Occidente, Instituto Mexicano del Seguro Social (IMSS), Guadalajara, Mexico
| | - Fabiola Solorzano-Ibarra
- Programa de Doctorado en Ciencias Biomédicas Orientación Inmunología, Centro Universitario de Ciencias de la Salud (CUCS), Universidad de Guadalajara (UdeG), Guadalajara, Mexico
| | - Anibal Samael Méndez-Clemente
- División de Inmunología, Centro de Investigación Biomédica de Occidente (CIBO), Instituto Mexicano del Seguro Social (IMSS), Guadalajara, Mexico.,Programa de Doctorado en Ciencias Biomédicas Orientación Inmunología, Centro Universitario de Ciencias de la Salud (CUCS), Universidad de Guadalajara (UdeG), Guadalajara, Mexico
| | - Adriana Aguilar-Lemarroy
- División de Inmunología, Centro de Investigación Biomédica de Occidente (CIBO), Instituto Mexicano del Seguro Social (IMSS), Guadalajara, Mexico
| | - Luis Felipe Jave-Suárez
- División de Inmunología, Centro de Investigación Biomédica de Occidente (CIBO), Instituto Mexicano del Seguro Social (IMSS), Guadalajara, Mexico
| | - Édgar Ruiz Velazco-Niño
- División de Inmunología, Centro de Investigación Biomédica de Occidente (CIBO), Instituto Mexicano del Seguro Social (IMSS), Guadalajara, Mexico
| | - Georgina Hernández-Flores
- División de Inmunología, Centro de Investigación Biomédica de Occidente (CIBO), Instituto Mexicano del Seguro Social (IMSS), Guadalajara, Mexico
| |
Collapse
|
22
|
Casal-Mouriño A, Ruano-Ravina A, Torres-Durán M, Parente-Lamelas I, Provencio-Pulla M, Castro-Añón O, Vidal-García I, Abal-Arca J, Piñeiro-Lamas M, Fernández-Villar A, Valdés-Cuadrado L, Barros-Dios JM, Pérez-Ríos M. Polymorphisms in the BER and NER pathways and their influence on survival and toxicity in never-smokers with lung cancer. Sci Rep 2020; 10:21147. [PMID: 33273562 PMCID: PMC7713126 DOI: 10.1038/s41598-020-78051-5] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2020] [Accepted: 11/11/2020] [Indexed: 01/05/2023] Open
Abstract
Polymorphisms in DNA repair pathways may play a relevant role in lung cancer survival in never-smokers. Furthermore, they could be implicated in the response to chemotherapy and toxicity of platinum agents. The aim of this study was to evaluate the influence of various genetic polymorphisms in the BER and NER DNA repair pathways on survival and toxicity in never-smoker LC patients. The study included never-smokers LC cases diagnosed from 2011 through 2019, belonging to the Lung Cancer Research In Never Smokers study. A total of 356 never-smokers cases participated (79% women; 83% adenocarcinoma and 65% stage IV). Survival at 3 and 5 years from diagnosis was not associated with genetic polymorphisms, except in the subgroup of patients who received radiotherapy or chemo-radiotherapy, and presented with ERCC1 rs3212986 polymorphism. There was greater toxicity in those presenting OGG1 rs1052133 (CG) and ERCC1 rs11615 polymorphisms among patients treated with radiotherapy or chemo-radiotherapy, respectively. In general, polymorphisms in the BER and NER pathways do not seem to play a relevant role in survival and response to treatment among never-smoker LC patients.
Collapse
Affiliation(s)
- Ana Casal-Mouriño
- Department of Pneumology, Santiago de Compostela University Clinical Teaching Hospital, Santiago de Compostela, Spain.,Department of Preventive Medicine and Public Health, University of Santiago de Compostela, C/San Francisco s/n, 15782, Santiago de Compostela, Spain
| | - Alberto Ruano-Ravina
- Department of Preventive Medicine and Public Health, University of Santiago de Compostela, C/San Francisco s/n, 15782, Santiago de Compostela, Spain. .,Consortium for Biomedical Research in Epidemiology and Public Health (CIBER en Epidemiología and Salud Pública-CIBERESP), Madrid, Spain. .,C013 Group-Health Research Institute of Santiago de Compostela (Instituto Investigación Sanitaria de Santiago de Compostela/IDIS), Santiago de Compostela, Spain.
| | - María Torres-Durán
- Department of Pneumology, Vigo University Teaching Hospital Complex, Vigo, Spain
| | | | | | | | - Iria Vidal-García
- Department of Pneumology, A Coruña University Teaching Hospital Complex, Vigo, Spain
| | - José Abal-Arca
- Department of Pneumology, Ourense University Teaching Hospital Complex, Ourense, Spain
| | - María Piñeiro-Lamas
- Consortium for Biomedical Research in Epidemiology and Public Health (CIBER en Epidemiología and Salud Pública-CIBERESP), Madrid, Spain
| | | | - Luis Valdés-Cuadrado
- Department of Pneumology, Santiago de Compostela University Clinical Teaching Hospital, Santiago de Compostela, Spain.,Interdisciplinary Neumology Research Group, Health Research Institute of Santiago de Compostela (Instituto Investigación Sanitaria de Santiago de Compostela/IDIS), Santiago de Compostela, Spain
| | - Juan Miguel Barros-Dios
- Department of Preventive Medicine and Public Health, University of Santiago de Compostela, C/San Francisco s/n, 15782, Santiago de Compostela, Spain.,Consortium for Biomedical Research in Epidemiology and Public Health (CIBER en Epidemiología and Salud Pública-CIBERESP), Madrid, Spain
| | - Mónica Pérez-Ríos
- Department of Preventive Medicine and Public Health, University of Santiago de Compostela, C/San Francisco s/n, 15782, Santiago de Compostela, Spain.,Consortium for Biomedical Research in Epidemiology and Public Health (CIBER en Epidemiología and Salud Pública-CIBERESP), Madrid, Spain.,C013 Group-Health Research Institute of Santiago de Compostela (Instituto Investigación Sanitaria de Santiago de Compostela/IDIS), Santiago de Compostela, Spain
| |
Collapse
|
23
|
Tian M, Liu Y, Jiang FL. On the Route to Quantitative Detection and Real-Time Monitoring of Glutathione in Living Cells by Reversible Fluorescent Probes. Anal Chem 2020; 92:14285-14291. [PMID: 33063515 DOI: 10.1021/acs.analchem.0c03418] [Citation(s) in RCA: 33] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
In the last few decades, growing numbers of fluorescent probes have been developed to detect intracellular GSH. However, the majority of probes for GSH were irreversible without monitoring the changes of intracellular GSH concentration. Therefore, recently, fluorescent probes for monitoring concentrations of GSH in real-time in living cells have come into being to address this challenge. This Perspective aimed at the development of reversible probes for GSH was organized by structural features, chemical reactions, and physicochemical properties. The reversible probes designed by a coumarin skeleton as a read-out fluorophore and the Michael addition reaction as a response mechanism accounted for most of the reported reversible probes. The performances of reversible fluorescent probes based on Michael addition could be roughly predicted by fundamental laws of kinetics and thermodynamics in physical chemistry. Essentially, the design principles included a highly reactive site for GSH, a small thermodynamic driving force, a desirable Kd of 1-10 mM, and excellent cell membrane permeability. Prospectively, the development of various mechanisms and fluorophores will be effective measures to enrich the types of reversible probes for GSH.
Collapse
Affiliation(s)
- Ming Tian
- Sauvage Center for Molecular Sciences, College of Chemistry and Molecular Sciences, Wuhan University, Wuhan 430072, P. R. China
| | - Yi Liu
- Sauvage Center for Molecular Sciences, College of Chemistry and Molecular Sciences, Wuhan University, Wuhan 430072, P. R. China.,Hubei Province Key Laboratory for Coal Conversion and New Carbon Materials, School of Chemistry and Chemical Engineering, Wuhan University of Science and Technology, Wuhan 430081, P. R. China.,Guangxi Key Laboratory of Natural Polymer Chemistry, College of Chemistry and Materials Science, Nanning Normal University, Nanning 530001, P. R. China
| | - Feng-Lei Jiang
- Sauvage Center for Molecular Sciences, College of Chemistry and Molecular Sciences, Wuhan University, Wuhan 430072, P. R. China
| |
Collapse
|
24
|
Sun Y, Li C, Pang S, Yao Q, Chen L, Li Y, Zeng R. Kinase-substrate Edge Biomarkers Provide a More Accurate Prognostic Prediction in ER-negative Breast Cancer. GENOMICS, PROTEOMICS & BIOINFORMATICS 2020; 18:525-538. [PMID: 33450402 PMCID: PMC8377385 DOI: 10.1016/j.gpb.2019.11.012] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/19/2018] [Revised: 08/27/2019] [Accepted: 11/11/2019] [Indexed: 11/19/2022]
Abstract
The estrogen receptor (ER)-negative breast cancer subtype is aggressive with few treatment options available. To identify specific prognostic factors for ER-negative breast cancer, this study included 705,729 and 1034 breast invasive cancer patients from the Surveillance, Epidemiology, and End Results (SEER) and The Cancer Genome Atlas (TCGA) databases, respectively. To identify key differential kinase-substrate node and edge biomarkers between ER-negative and ER-positive breast cancer patients, we adopted a network-based method using correlation coefficients between molecular pairs in the kinase regulatory network. Integrated analysis of the clinical and molecular data revealed the significant prognostic power of kinase-substrate node and edge features for both subtypes of breast cancer. Two promising kinase-substrate edge features, CSNK1A1-NFATC3 and SRC-OCLN, were identified for more accurate prognostic prediction in ER-negative breast cancer patients.
Collapse
Affiliation(s)
- Yidi Sun
- CAS Key Laboratory of Systems Biology, CAS Center for Excellence in Molecular Cell Science, Institute of Biochemistry and Cell Biology, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai 200031, China; University of Chinese Academy of Sciences, Shanghai 200031, China
| | - Chen Li
- CAS Key Laboratory of Systems Biology, CAS Center for Excellence in Molecular Cell Science, Institute of Biochemistry and Cell Biology, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai 200031, China
| | - Shichao Pang
- Deptartment of Statistics, School of Mathematical Sciences, Shanghai Jiao Tong University, Shanghai 200240, China
| | - Qianlan Yao
- School of Life Sciences and Biotechnology, Shanghai Jiao Tong University, Shanghai 200240, China
| | - Luonan Chen
- CAS Key Laboratory of Systems Biology, CAS Center for Excellence in Molecular Cell Science, Institute of Biochemistry and Cell Biology, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai 200031, China; Department of Life Sciences, ShanghaiTech University, Shanghai 201210, China; CAS Center for Excellence in Animal Evolution and Genetics, Chinese Academy of Sciences, Kunming 650223, China.
| | - Yixue Li
- School of Life Sciences and Biotechnology, Shanghai Jiao Tong University, Shanghai 200240, China; Department of Life Sciences, ShanghaiTech University, Shanghai 201210, China; Bio-Med Big Data Center, Key Laboratory of Computational Biology, CAS-MPG Partner Institute for Computational Biology, Shanghai Institute of Nutrition and Health, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai 200031, China; Collaborative Innovation Center for Genetics and Development, Fudan University, Shanghai 200032, China; Shanghai Center for Bioinformation Technology, Shanghai Academy of Science & Technology, Shanghai 201203, China.
| | - Rong Zeng
- CAS Key Laboratory of Systems Biology, CAS Center for Excellence in Molecular Cell Science, Institute of Biochemistry and Cell Biology, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai 200031, China; Department of Life Sciences, ShanghaiTech University, Shanghai 201210, China.
| |
Collapse
|
25
|
Tian M, Liu XY, He H, Ma XZ, Liang C, Liu Y, Jiang FL. Real-Time Imaging of Intracellular Glutathione Levels Based on a Ratiometric Fluorescent Probe with Extremely Fast Response. Anal Chem 2020; 92:10068-10075. [PMID: 32538069 DOI: 10.1021/acs.analchem.0c01881] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
Glutathione (GSH), the most abundant nonprotein thiol found in living organisms, are involved in the etiology and progression of many human diseases including cancer. So, monitoring changes of cellular GSH levels has an important guiding significance. To date, however, majority of probes can only qualitatively detect GSH in living cells. Herein, with coumarin as the read-out fluorophore and Michael addition as the sensing mechanism, six fluorescent probes were designed and synthesized. Among them, RP-2 exhibited a reversible and extremely fast response toward GSH (half time: ∼3 s), which endowed RP-2 the capacity of real-time imaging. Among the reversible probes based on Michael addition, RP-2 had both the largest forward and reverse rate constants thus far. The reaction between RP-2 and GSH was studied in detail by density functional theory and fluorescence spectroscopy. Real-time imaging of GSH levels in living cells was achieved with a temporal resolution of seconds. To simplify the processing of images, a program was developed and validated. RP-2 was expected to serve as a new fluorescent imaging tool to understand the function of intracellular GSH in the future.
Collapse
Affiliation(s)
- Ming Tian
- Sauvage Center for Molecular Sciences, College of Chemistry and Molecular Sciences, Wuhan University, Wuhan 430072, P. R. China
| | - Xing-Yu Liu
- Sauvage Center for Molecular Sciences, College of Chemistry and Molecular Sciences, Wuhan University, Wuhan 430072, P. R. China
| | - Huan He
- Sauvage Center for Molecular Sciences, College of Chemistry and Molecular Sciences, Wuhan University, Wuhan 430072, P. R. China
| | - Xian-Zheng Ma
- National Engineering Research Center for Multimedia Software (NERCMS), School of Computer Science, Wuhan University, Wuhan 430072, P. R. China
| | - Chao Liang
- National Engineering Research Center for Multimedia Software (NERCMS), School of Computer Science, Wuhan University, Wuhan 430072, P. R. China
| | - Yi Liu
- Sauvage Center for Molecular Sciences, College of Chemistry and Molecular Sciences, Wuhan University, Wuhan 430072, P. R. China.,Hubei Province Key Laboratory for Coal Conversion and New Carbon Materials, School of Chemistry and Chemical Engineering, Wuhan University of Science and Technology, Wuhan 430081, P. R. China.,Guangxi Key Laboratory of Natural Polymer Chemistry, College of Chemistry and Materials Science, Nanning Normal University, Nanning 530001, P. R. China
| | - Feng-Lei Jiang
- Sauvage Center for Molecular Sciences, College of Chemistry and Molecular Sciences, Wuhan University, Wuhan 430072, P. R. China
| |
Collapse
|
26
|
Novel Liquid Biomarker Panels for A Very Early Response Capturing of NSCLC Therapies in Advanced Stages. Cancers (Basel) 2020; 12:cancers12040954. [PMID: 32290637 PMCID: PMC7226444 DOI: 10.3390/cancers12040954] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2020] [Revised: 04/09/2020] [Accepted: 04/10/2020] [Indexed: 12/28/2022] Open
Abstract
Computed tomography (CT) scans are the gold standard to measure treatment success of non-small cell lung cancer (NSCLC) therapies. Here, we investigated the very early tumor response of patients receiving chemotherapy or targeted therapies using a panel of already established and explorative liquid biomarkers. Blood samples from 50 patients were taken at baseline and at three early time points after therapy initiation. DNA mutations, a panel of 17 microRNAs, glycodelin, glutathione disulfide, glutathione, soluble caspase-cleaved cytokeratin 18 (M30 antigen), and soluble cytokeratin 18 (M65 antigen) were measured in serum and plasma samples. Baseline and first follow-up CT scans were evaluated and correlated with biomarker data. The detection rate of the individual biomarkers was between 56% and 100%. While only keratin 18 correlated with the tumor load at baseline, we found several individual markers correlating with the tumor response to treatment for each of the three time points of blood draws. A combination of the five best markers at each time point resulted in highly significant marker panels indicating therapeutic response (R2 = 0.78, R2 = 0.71, and R2 = 0.71). Our study demonstrates that an early measurement of biomarkers immediately after therapy start can assess tumor response to treatment and might support an adaptation of treatment to improve patients’ outcome.
Collapse
|
27
|
Huang Y, Lü X, Chen R, Chen Y. Comparative study of the effects of gold and silver nanoparticles on the metabolism of human dermal fibroblasts. Regen Biomater 2020; 7:221-232. [PMID: 32296541 PMCID: PMC7147366 DOI: 10.1093/rb/rbz051] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2019] [Revised: 11/25/2019] [Accepted: 12/04/2019] [Indexed: 12/26/2022] Open
Abstract
The purpose of this article was to explore the effects of gold nanoparticles (GNPs) and silver nanoparticles (SNPs) with different cytotoxicities on human dermal fibroblasts (HDFs) at the metabolic level. First, ∼20 nm of GNPs and SNPs were prepared, and their effects on the proliferation of HDFs were evaluated. Then, a metabolomics technique was used to analyse the effects of GNPs and SNPs on the expression profiles of metabolites in HDFs after 4, 8 and 24 h of treatment. Furthermore, the key metabolites and key metabolic pathways involved in the interaction of GNPs and SNPs with HDFs were identified through expression pattern analysis and metabolic pathway analysis of differentially expressed metabolites and were finally verified by experiments. The results of the cytotoxicity experiments showed that there was no cytotoxicity after the treatment of GNPs for 72 h, while the cytotoxicity of the SNPs reached grade 1 after 72 h. By using metabolomics analysis, 29, 30 and 27 metabolites were shown to be differentially expressed in HDFs after GNP treatment, while SNPs induced the differential expression of 13, 33 and 22 metabolites after 4, 8 and 24 h of treatment, respectively. Six and four candidate key metabolites in the GNP and SNP groups were identified by expression pattern analysis and metabolic pathway analysis, respectively. The key metabolic pathways in the GNP and SNP groups were identified as the glutathione metabolic pathway (the key metabolite of which was glutathione) and the citrate cycle pathway (the key metabolite of which was malic acid). Based on the experiments used to verify the key metabolites and key metabolic pathways, it was found that the increase in glutathione after GNP treatment might trigger an oxidative stress protection mechanism and thus avoid cytotoxicity. After exposure to SNPs, the citric acid content was increased, mainly through the citrate cycle pathway, thereby inhibiting the synthesis of malic acid to affect the formation of ATP and finally leading to cytotoxicity.
Collapse
Affiliation(s)
- Yan Huang
- State Key Laboratory of Bioelectronics, School of Biological Science and Medical Engineering, Southeast University, Nanjing 210096, P.R. China
| | - Xiaoying Lü
- State Key Laboratory of Bioelectronics, School of Biological Science and Medical Engineering, Southeast University, Nanjing 210096, P.R. China
| | - Rong Chen
- State Key Laboratory of Bioelectronics, School of Biological Science and Medical Engineering, Southeast University, Nanjing 210096, P.R. China
| | - Ye Chen
- State Key Laboratory of Bioelectronics, School of Biological Science and Medical Engineering, Southeast University, Nanjing 210096, P.R. China
| |
Collapse
|
28
|
Chang L, Fang S, Gu W. The Molecular Mechanism of Metabolic Remodeling in Lung Cancer. J Cancer 2020; 11:1403-1411. [PMID: 32047547 PMCID: PMC6995370 DOI: 10.7150/jca.31406] [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: 11/11/2018] [Accepted: 10/23/2019] [Indexed: 12/11/2022] Open
Abstract
Metabolic remodeling is a key phenomenon in the occurrence and development of tumors. It not only offers materials and energy for the survival and proliferation of tumor cells, but also protects tumor cells so that they may survive, proliferate and transfer in the harsh microenvironment. This paper attempts to reveal the role of abnormal metabolism in the development of lung cancer by considering the processes of glycolysis and lipid metabolism, Identification of the molecules that are specifically used in the processes of glycolysis and lipid metabolism, and their underlying molecular mechanisms, is of great clinical and theoretical significance. We will focus on the recent progress in elucidating the molecular mechanism of metabolic remodeling in lung cancer.
Collapse
Affiliation(s)
| | | | - Wei Gu
- Department of Respiratory Medicine, Nanjing First Hospital, Nanjing Medical University. No. 68 Changle Road, Qinhuai District, Nanjing 210001,People's Republic of China
| |
Collapse
|
29
|
Chaudhari KR, Kunwar A, Bhuvanesh N, Dey S. Synthesis and anti-proliferative activities of amine capped Pd and Pt macrocycles of 4,4′-dipyridylselenides. NEW J CHEM 2020. [DOI: 10.1039/c9nj06052a] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
Symmetric macrocyclic complexes characterized as dimeric and their oligomeric form in water and the solid state exhibit high in vitro anticancer activities.
Collapse
Affiliation(s)
- K. R. Chaudhari
- Chemistry Division
- Bhabha Atomic Research Centre
- Mumbai 400 085
- India
| | - A. Kunwar
- Homi Bhabha National Institute
- Training School Complex
- Mumbai 400 094
- India
- Radiation and Photo Chemistry Division
| | - N. Bhuvanesh
- Department of Chemistry
- Texas A&M University
- College Station
- USA
| | - S. Dey
- Chemistry Division
- Bhabha Atomic Research Centre
- Mumbai 400 085
- India
- Homi Bhabha National Institute
| |
Collapse
|
30
|
Sharma M, Anirudh CR. In silico characterization of residues essential for substrate binding of human cystine transporter, xCT. J Mol Model 2019; 25:336. [PMID: 31705320 DOI: 10.1007/s00894-019-4233-y] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2019] [Accepted: 10/14/2019] [Indexed: 02/06/2023]
Abstract
xCT is a sodium-independent amino acid antiporter that imports L-cystine and exports L-glutamate in a 1:1 ratio. It is a component of heterodimeric amino acid transporter system Xc- working at the cross-roads of maintaining neurological processes and regulating antioxidant defense. The transporter has 12 transmembrane domains with intracellular N- and C-termini, and like other transporter proteins can undergo various conformational changes while switching the ligand accessibilities from intracellular to extracellular site. In the present study, we generated two homology models of human xCT in two distinct conformations: inward-facing occluded state and outward-facing open state. Our results indicated the substrate translocation channel composed of transmembrane helices TMs 1, 3, 6, 8, and 10. We docked anionic L-cystine and L-glutamate within the cavities to assess the two distinct binding scenarios for xCT as antiporter. We also assessed the interactions between the ligands and transporter and observed that ligands bind to similar residues within the channel. Using MM-PBSA/MM-GBSA approach, we computed the binding energies of these ligands to different conformational states. Cystine and glutamate bind xCT with favorable binding energies, with more favorable binding observed in inward occluded state than in outward open state. We further computed the residue-wise decomposition of these binding energies and identified the residues as essential for substrate binding/permeation. Filtering the residues that form favorable energetic contributions to the ligand binding in both the states, our studies suggest T56, A60, R135, A138, V141, Y244, A247, F250, S330, L392, and R396 as critical residues for ligand binding as well as ligand transport for any conformational state adopted by xCT during its transport cycle. .Graphical Abstract.
Collapse
Affiliation(s)
- Monika Sharma
- Department of Chemical Sciences, Indian Institute of Science Education and Research (IISER), Sector 81, Knowledge City, SAS, Nagar, Punjab, India.
| | - C R Anirudh
- Department of Chemical Sciences, Indian Institute of Science Education and Research (IISER), Sector 81, Knowledge City, SAS, Nagar, Punjab, India
| |
Collapse
|
31
|
Bahar E, Kim JY, Yoon H. Chemotherapy Resistance Explained through Endoplasmic Reticulum Stress-Dependent Signaling. Cancers (Basel) 2019; 11:cancers11030338. [PMID: 30857233 PMCID: PMC6468910 DOI: 10.3390/cancers11030338] [Citation(s) in RCA: 66] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2019] [Revised: 03/01/2019] [Accepted: 03/01/2019] [Indexed: 02/06/2023] Open
Abstract
Cancers cells have the ability to develop chemotherapy resistance, which is a persistent problem during cancer treatment. Chemotherapy resistance develops through different molecular mechanisms, which lead to modification of the cancer cells signals needed for cellular proliferation or for stimulating an immune response. The endoplasmic reticulum (ER) is an important organelle involved in protein quality control, by promoting the correct folding of protein and ER-mediated degradation of unfolded or misfolded protein, namely, ER-associated degradation. Disturbances of the normal ER functions causes an accumulation of unfolded or misfolded proteins in the ER lumen, resulting in a condition called “ER stress (ERS).” ERS triggers the unfolded protein response (UPR)—also called the ERS response (ERSR)—to restore homeostasis or activate cell death. Although the ERSR is one emerging potential target for chemotherapeutics to treat cancer, it is also critical for chemotherapeutics resistance, as well. However, the detailed molecular mechanism of the relationship between the ERSR and tumor survival or drug resistance remains to be fully understood. In this review, we aim to describe the most vital molecular mechanism of the relationship between the ERSR and chemotherapy resistance. Moreover, the review also discusses the molecular mechanism of ER stress-mediated apoptosis on cancer treatments.
Collapse
Affiliation(s)
- Entaz Bahar
- College of Pharmacy, Research Institute of Pharmaceutical Sciences, Gyeongsang National University, Jinju 52828, Gyeongnam, Korea.
| | - Ji-Ye Kim
- Department of Pathology, College of Medicine, Yonsei University, Seoul 03722, Korea.
- Department of Pathology, Ilsan Paik Hospital, Inje University, Goyang 10381, Gyeonggi-do, Korea.
- Department of Pathology, National Cancer Center, Goyang 10408, Gyeonggi-do, Korea.
| | - Hyonok Yoon
- College of Pharmacy, Research Institute of Pharmaceutical Sciences, Gyeongsang National University, Jinju 52828, Gyeongnam, Korea.
| |
Collapse
|
32
|
Hanko M, Švorc Ľ, Planková A, Mikuš P. Overview and recent advances in electrochemical sensing of glutathione - A review. Anal Chim Acta 2019; 1062:1-27. [PMID: 30947984 DOI: 10.1016/j.aca.2019.02.052] [Citation(s) in RCA: 62] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2018] [Revised: 02/20/2019] [Accepted: 02/22/2019] [Indexed: 12/13/2022]
Abstract
The present paper is aimed at providing an overview of the recent advances in the electrochemical sensing of glutathione (GSH), an important electrochemically and biologically active molecule, for the period 2012-2018. Herein, the analytical performances of newly developed electrochemical methods, procedures and protocols for GSH sensing are comprehensively and critically discussed with respect to the type of method, electrodes used (new electrode modifications, advanced materials and formats), sample matrices, and basic validation parameters obtained (limit of detection, linear dynamic range, precision, selectivity/evaluation of interferences). This paper considers electrochemical methods used alone as well as the hyphenated methods with electrochemical detection (ECD), such as HPLC-ECD or CE-ECD. The practical applicability of the platforms developed for GSH detection and quantification is mostly focused on pharmaceutical and biomedical analysis. The most significant electrochemical approaches for GSH detection in multicomponent analyte samples and multicomponent matrices and for real-time in vivo GSH analysis are highlighted. The great variability in the electrochemical techniques, electrode approaches, and obtainable performance parameters, discussed in this review, brought new insights not only on current GSH and glutathione disulfide (GSSG) determinations, but, along with this, on the advances in electrochemical analysis from a more general point of view.
Collapse
Affiliation(s)
- Michal Hanko
- Comenius University in Bratislava, Faculty of Pharmacy, Department of Pharmaceutical Analysis and Nuclear Pharmacy, Odbojárov 10, SK-832 32, Bratislava, Slovak Republic
| | - Ľubomír Švorc
- Slovak University of Technology in Bratislava, Faculty of Chemical and Food Technology, Institute of Analytical Chemistry, Radlinského 9, SK-812 37, Bratislava, Slovak Republic
| | - Alexandra Planková
- Comenius University in Bratislava, Faculty of Pharmacy, Department of Pharmaceutical Analysis and Nuclear Pharmacy, Odbojárov 10, SK-832 32, Bratislava, Slovak Republic
| | - Peter Mikuš
- Comenius University in Bratislava, Faculty of Pharmacy, Department of Pharmaceutical Analysis and Nuclear Pharmacy, Odbojárov 10, SK-832 32, Bratislava, Slovak Republic; Comenius University in Bratislava, Faculty of Pharmacy, Toxicological and Antidoping Center, Odbojárov 10, SK-832 32, Bratislava, Slovak Republic.
| |
Collapse
|
33
|
Lehmann SG, Seve M, Vanwonterghem L, Michelland S, Cunin V, Coll JL, Hurbin A, Bourgoin-Voillard S. A large scale proteome analysis of the gefitinib primary resistance overcome by KDAC inhibition in KRAS mutated adenocarcinoma cells overexpressing amphiregulin. J Proteomics 2019; 195:114-124. [PMID: 30660770 DOI: 10.1016/j.jprot.2019.01.009] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2018] [Accepted: 01/14/2019] [Indexed: 02/06/2023]
Abstract
KDAC inhibitors (KDACi) overcome gefitinib primary resistance in non-small cell lung cancer (NSCLC) including mutant-KRAS lung adenocarcinoma. To identify which proteins are involved in the restoration of this sensitivity and to provide new therapeutic targets for mutant-KRAS lung adenocarcinoma, we performed an iTRAQ quantitative proteomic analysis after subcellular fractionation of H358-NSCLC treated with gefitinib and KDACi (TSA/NAM) versus gefitinib alone. The 86 proteins found to have been significantly dysregulated between the two conditions, were mainly involved in cellular metabolism and cell transcription processes. As expected, the pathway related to histone modifications was affected by the KDACi. Pathways known for controlling tumor development and (chemo)-resistance (miRNA biogenesis/glutathione metabolism) were affected by the KDACi/gefitinib treatment. Moreover, 57 dysregulated proteins were upstream of apoptosis (such as eEF1A2 and STAT1) and hence provide potential therapeutic targets. The inhibition by siRNA of eEF1A2 expression resulted in a slight decrease in H358-NSCLC viability. In addition, eEF1A2 and STAT1 siRNA transfections suggested that both STAT1 and eEF1A2 prevent AKT phosphorylation known for enhancing gefitinib resistance in NSCLC. Therefore, altogether our data provide new insights into proteome regulations in the context of overcoming the NSCLC resistance to gefitinib through KDACi in H358 KRAS mutated and amphiregulin-overexpressing NSCLC cells.
Collapse
Affiliation(s)
- Sylvia G Lehmann
- Univ. Grenoble Alpes, LBFA and BEeSy, PROMETHEE Proteomic Platform, Grenoble, France; Inserm, U1055, PROMETHEE Proteomic Platform, Grenoble, France; CHU Grenoble Alpes, Institut de Biologie et de Pathologie, PROMETHEE Proteomic Platform, Grenoble, France; Univ. Grenoble Alpes, ISTerre, F-38000 Grenoble, France
| | - Michel Seve
- Univ. Grenoble Alpes, LBFA and BEeSy, PROMETHEE Proteomic Platform, Grenoble, France; Inserm, U1055, PROMETHEE Proteomic Platform, Grenoble, France; CHU Grenoble Alpes, Institut de Biologie et de Pathologie, PROMETHEE Proteomic Platform, Grenoble, France
| | - Laetitia Vanwonterghem
- Cancer target and experimental therapeutics, Institute for Advanced Biosciences, INSERM U1209, CNRS UMR5301, Univ. Grenoble Alpes, F-38000 Grenoble, France
| | - Sylvie Michelland
- Univ. Grenoble Alpes, LBFA and BEeSy, PROMETHEE Proteomic Platform, Grenoble, France; Inserm, U1055, PROMETHEE Proteomic Platform, Grenoble, France; CHU Grenoble Alpes, Institut de Biologie et de Pathologie, PROMETHEE Proteomic Platform, Grenoble, France
| | - Valérie Cunin
- Univ. Grenoble Alpes, LBFA and BEeSy, PROMETHEE Proteomic Platform, Grenoble, France; Inserm, U1055, PROMETHEE Proteomic Platform, Grenoble, France; CHU Grenoble Alpes, Institut de Biologie et de Pathologie, PROMETHEE Proteomic Platform, Grenoble, France
| | - Jean-Luc Coll
- Cancer target and experimental therapeutics, Institute for Advanced Biosciences, INSERM U1209, CNRS UMR5301, Univ. Grenoble Alpes, F-38000 Grenoble, France
| | - Amandine Hurbin
- Cancer target and experimental therapeutics, Institute for Advanced Biosciences, INSERM U1209, CNRS UMR5301, Univ. Grenoble Alpes, F-38000 Grenoble, France.
| | - Sandrine Bourgoin-Voillard
- Univ. Grenoble Alpes, LBFA and BEeSy, PROMETHEE Proteomic Platform, Grenoble, France; Inserm, U1055, PROMETHEE Proteomic Platform, Grenoble, France; CHU Grenoble Alpes, Institut de Biologie et de Pathologie, PROMETHEE Proteomic Platform, Grenoble, France.
| |
Collapse
|
34
|
Jin X, Liu X, Zhang Z, Guan Y, Xv R, Li J. Identification of key pathways and genes in lung carcinogenesis. Oncol Lett 2018; 16:4185-4192. [PMID: 30250533 PMCID: PMC6144915 DOI: 10.3892/ol.2018.9203] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2016] [Accepted: 03/14/2018] [Indexed: 01/19/2023] Open
Abstract
The present study aimed to identify key pathways and genes in the pathogenesis of lung cancer. The GSE10072 dataset was downloaded from the Gene Expression Omnibus database. Protein-protein interaction data were collected from Human Protein Reference Database, and 201 pathways were downloaded from the Kyoto Encyclopedia of Genes and Genomes database. Signaling network impact analysis was performed to identify enriched pathways, followed by the construction of a pathway-pathway crosstalk network. Benzopyrene was used to treat normal human lung cells at concentrations of 0.01, 0.1, 1 and 10 µM, and cell viability was measured. Furthermore, growth arrest and DNA damage inducible β (GADD45B), p53, cyclin B, Akt and nuclear factor (NF)-κB protein levels were also measured via western blotting. Impact analysis identified 11 enriched lung cancer-associated KEGG pathways, including 'complement and coagulation cascades', 'ECM-receptor interaction', 'P53 signaling pathway', 'cell adhesion molecules' and 'focal adhesion'. In addition, cell cycle, 'drug metabolism-cytochrome P450', 'metabolic pathways', 'pathways in cancer', 'focal adhesion' and 'antigen processing and presentation' were central in the pathway-pathway cross-talk network. Furthermore, the upregulated gene GADD45B was associated with three of the pathways, including an activated pathway ('MAPK signaling pathway') and two repressed pathways ('cell cycle' and 'P53 pathway'). Western blotting demonstrated that the expression of NF-κB, Akt and GADD45B increased over time in lung cells treated with benzopyrene, whereas the expression levels of cyclin B and P53 decreased. In conclusion, GADD45B may contribute to lung carcinogenesis via affecting the MAPK, P53 signaling and cell cycle pathways.
Collapse
Affiliation(s)
- Xiang Jin
- Department of Respiration, The First Hospital of Jilin University, Changchun, Jilin 130021, P.R. China
| | - Xingang Liu
- Department of ICU, The First Hospital of Jilin University, Changchun, Jilin 130021, P.R. China
| | - Zhen Zhang
- Department of PICU, The First Hospital of Jilin University, Changchun, Jilin 130021, P.R. China
| | - Yinghui Guan
- Department of Respiration, The First Hospital of Jilin University, Changchun, Jilin 130021, P.R. China
| | - Ren Xv
- Department of Respiration, The First Hospital of Jilin University, Changchun, Jilin 130021, P.R. China
| | - Jun Li
- Department of Pneumology, Jinan Central Hospital, Jinan, Shandong 250013, P.R. China
| |
Collapse
|
35
|
Hadami K, Dakka N, Bensaid M, El Ahanidi H, Ameur A, Chahdi H, Oukabli M, Al Bouzidi A, Attaleb M, El Mzibri M. Evaluation of glutathione S-transferase pi 1 expression and gene promoter methylation in Moroccan patients with urothelial bladder cancer. Mol Genet Genomic Med 2018; 6:819-827. [PMID: 30043549 PMCID: PMC6160697 DOI: 10.1002/mgg3.449] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2018] [Revised: 05/29/2018] [Accepted: 06/13/2018] [Indexed: 12/23/2022] Open
Abstract
BACKGROUND Glutathione S-transferase pi 1 (GSTP1) is a cytosolic detoxifying enzyme that protects cells against deleterious effects of oxidative stress. Deregulated expression of GSTP1 protein and aberrant promoter methylation of GSTP1 gene were reported in various human tumors and were shown to be involved in the molecular pathway for cancer development. AIMS AND METHODS In this study, we aimed to determine the expression status of GSTP1 in relation to its gene promoter methylation in Moroccan population of 30 bladder cancer (BC) patients and in two noncancerous bladder tissues used as controls. GSTP1 expression was assessed by immunohistochemistry and GSTP1 gene promoter methylation status was studied by methylation-specific PCR (MS-PCR). RESULTS Glutathione S-transferase pi 1 was expressed in the two normal tissues. In BC cases, GSTP1 expression was strong in 23.33% (7/30), moderate in 60% (18/30), and weak in 13.33% (4/30) of cases, while GSTP1 was not expressed in one cancer case (3.33%). Variability of GSTP1 expression does not correlate with high-grade cancer or invasive-stage (p > 0.05). No GSTP1 gene promoter methylation was detected in all control and cancer cases. CONCLUSION Our data suggest that GSTP1 expression is not associated with BC development, limiting its use as a biomarker for BC management in Morocco. Moreover, difference in GSTP1 expression among BC cases is not due to GSTP1 promoter methylation.
Collapse
Affiliation(s)
- Khaoula Hadami
- Biology and Medical Research UnitCNESTENRabatMorocco
- Biology of Human Pathologies LaboratoryFaculty of SciencesMohammed V UniversityRabatMorocco
| | - Nadia Dakka
- Biology of Human Pathologies LaboratoryFaculty of SciencesMohammed V UniversityRabatMorocco
| | | | | | | | | | | | | | | | | |
Collapse
|
36
|
Ruano-Ravina A, García-Basteiro AL, Pérez-Ríos M, Gómez-Mosquera A, Cerdeira-Caramés S, Barros-Dios JM. Lung cancer survival and deletion of GSTM1 and GSTT1 genes. A case-series from Spain. TUMORI JOURNAL 2018; 99:445-51. [DOI: 10.1177/030089161309900401] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
Aims and background Lung cancer has a high incidence, and only 15% of all cases are alive 5 years after the diagnosis. Identifying the role of the genes implicated in the success of chemotherapy agents is crucial to predict survival. The objective of the study was to assess the effect of GSTM1 and GSTT1 gene deletion on lung cancer survival. Methods and study design A consecutive sampling of lung cancer cases was performed in 1999–2000 at the University Hospital of Santiago de Compostela, Spain, and GSTM1 and GSTT1 genes were genotyped. The effect of GSTM1 and GSTT1 deletion on survival was analyzed with the logrank test and with Cox regression. Results A total of 132 individuals were included, and more than half of them had stage IV lung cancer at diagnosis. Survival was similar irrespective of the presence or absence of the GSTM1 gene, whereas those with deleted GSTT1 had a significantly shorter survival. In multivariate Cox regression analysis, no significant effect was observed for the deletion of any of the genes, although there was a tendency towards a worse survival for those with deletion of GSTT1. The main limitation was that stage at diagnosis could not be considered in the analysis since most of the patients were diagnosed at stage IV. Conclusions GSTT1 appears to influence lung cancer survival whereas GSTM1 seems to have no effect.
Collapse
Affiliation(s)
- Alberto Ruano-Ravina
- Department of Preventive Medicine and Public Health, University of Santiago de Compostela, Santiago de Compostela
- CIBER de Epidemiología y Salud Pública, Madrid
| | | | - Mónica Pérez-Ríos
- Department of Preventive Medicine and Public Health, University of Santiago de Compostela, Santiago de Compostela
- CIBER de Epidemiología y Salud Pública, Madrid
| | - Antón Gómez-Mosquera
- Service of Obstetrics and Gynecology, University Hospital of Vigo, Galician Health Service, Vigo
| | - Sara Cerdeira-Caramés
- Service of Epidemiology, Department of Health, Xunta de Galicia, Santiago de Compostela
| | - Juan Miguel Barros-Dios
- Department of Preventive Medicine and Public Health, University of Santiago de Compostela, Santiago de Compostela
- CIBER de Epidemiología y Salud Pública, Madrid
- Service of Preventive Medicine, Clinic University Hospital of Santiago de Compostela, Santiago de Compostela, Spain
| |
Collapse
|
37
|
Clinical Practice Recommendations for the Management and Prevention of Cisplatin-Induced Hearing Loss Using Pharmacogenetic Markers. Ther Drug Monit 2017; 38:423-31. [PMID: 26960170 DOI: 10.1097/ftd.0000000000000298] [Citation(s) in RCA: 35] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Currently no pharmacogenomics-based criteria exist to guide clinicians in identifying individuals who are at risk of hearing loss from cisplatin-based chemotherapy. This review summarizes findings from pharmacogenomic studies that report genetic polymorphisms associated with cisplatin-induced hearing loss and aims to (1) provide up-to-date information on new developments in the field, (2) provide recommendations for the use of pharmacogenetic testing in the prevention, assessment, and management of cisplatin-induced hearing loss in children and adults, and (3) identify knowledge gaps to direct and prioritize future research. These practice recommendations for pharmacogenetic testing in the context of cisplatin-induced hearing loss reflect a review and evaluation of recent literature, and are designed to assist clinicians in providing optimal clinical care for patients receiving cisplatin-based chemotherapy.
Collapse
|
38
|
Deletion of GSTM1 and GSTT1 genes and lung cancer survival: a systematic review. TUMORI JOURNAL 2017; 103:338-344. [PMID: 28315511 DOI: 10.5301/tj.5000621] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 02/23/2017] [Indexed: 01/05/2023]
Abstract
PURPOSE The mechanisms of lung carcinogenesis are not fully understood. Not all smokers develop lung cancer, indicating that genetic variations and other environmental factors may play an important role in its development. The human glutathione S-transferases (GSTs) have been associated with an increased risk of lung cancer. Glutathione S-transferases are phase II biotransformation enzymes that play a role in detoxifying a wide range of exogenous agents including carcinogens but also anticarcinogenic drugs. METHODS We assessed the effect of allelic deletions in the GSTM1 and GSTT1 genotypes on lung cancer overall survival through a systematic review of the scientific literature after applying predefined inclusion and exclusion criteria. RESULTS Most of the included studies found no effect or a tendency to worse survival for individuals with deletion of GSTs. CONCLUSIONS Further studies are necessary to understand the magnitude of the effect of the deletion of both genes on lung cancer survival.
Collapse
|
39
|
Yardım Y, Vandeput M, Çelebi M, Şentürk Z, Kauffmann JM. A Reduced Graphene Oxide-based Electrochemical DNA Biosensor for the Detection of Interaction between Cisplatin and DNA based on Guanine and Adenine Oxidation Signals. ELECTROANAL 2017. [DOI: 10.1002/elan.201600804] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/17/2023]
Affiliation(s)
- Yavuz Yardım
- Yuzuncu Yil University; Faculty of Pharmacy, Department of Analytical Chemistry; 65080 Van Turkey
| | - Marie Vandeput
- Free University of Brussels; ULB, Faculty of Pharmacy, Laboratory of Instrumental Analysis and Bioelectrochemistry, ULB 205/6, Campus Plaine; B-1050 Brussels Belgium
| | - Metin Çelebi
- Yuzuncu Yil University; Faculty of Science, Department of Inorganic Chemistry; 65080 Van Turkey
| | - Zuhre Şentürk
- Yuzuncu Yil University; Faculty of Science, Department of Analytical Chemistry; 65080 Van Turkey
| | - Jean-Michel Kauffmann
- Free University of Brussels; ULB, Faculty of Pharmacy, Laboratory of Instrumental Analysis and Bioelectrochemistry, ULB 205/6, Campus Plaine; B-1050 Brussels Belgium
| |
Collapse
|
40
|
Identifying reproducible cancer-associated highly expressed genes with important functional significances using multiple datasets. Sci Rep 2016; 6:36227. [PMID: 27796338 PMCID: PMC5086981 DOI: 10.1038/srep36227] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2016] [Accepted: 10/12/2016] [Indexed: 01/08/2023] Open
Abstract
Identifying differentially expressed (DE) genes between cancer and normal tissues is of basic importance for studying cancer mechanisms. However, current methods, such as the commonly used Significance Analysis of Microarrays (SAM), are biased to genes with low expression levels. Recently, we proposed an algorithm, named the pairwise difference (PD) algorithm, to identify highly expressed DE genes based on reproducibility evaluation of top-ranked expression differences between paired technical replicates of cells under two experimental conditions. In this study, we extended the application of the algorithm to the identification of DE genes between two types of tissue samples (biological replicates) based on several independent datasets or sub-datasets of a dataset, by constructing multiple paired average gene expression profiles for the two types of samples. Using multiple datasets for lung and esophageal cancers, we demonstrated that PD could identify many DE genes highly expressed in both cancer and normal tissues that tended to be missed by the commonly used SAM. These highly expressed DE genes, including many housekeeping genes, were significantly enriched in many conservative pathways, such as ribosome, proteasome, phagosome and TNF signaling pathways with important functional significances in oncogenesis.
Collapse
|
41
|
Shen F, Li J, Zhu Y, Wang Z. Systematic investigation of metabolic reprogramming in different cancers based on tissue-specific metabolic models. J Bioinform Comput Biol 2016; 14:1644001. [PMID: 27760488 DOI: 10.1142/s0219720016440017] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
Cancer cells have different metabolism in contrast to normal cells. The advancement in omics measurement technology enables the genome-wide characterization of altered cellular processes in cancers, but the metabolic flux landscape of cancer is still far from understood. In this study, we compared the well-reconstructed tissue-specific models of five cancers, including breast, liver, lung, renal, and urothelial cancer, and their corresponding normal cells. There are similar patterns in majority of significantly regulated pathways and enriched pathways in correlated reaction sets. But the differences among cancers are also explicit. The renal cancer demonstrates more dramatic difference with other cancer models, including the smallest number of reactions, flux distribution patterns, and specifically correlated pathways. We also validated the predicted essential genes and revealed the Warburg effect by in silico simulation in renal cancer, which are consistent with the measurements for renal cancer. In conclusion, the tissue-specific metabolic model is more suitable to investigate the cancer metabolism. The similarity and heterogenicity of metabolic reprogramming in different cancers are crucial for understanding the aberrant mechanisms of cancer proliferation, which is fundamental for identifying drug targets and biomarkers.
Collapse
Affiliation(s)
- Fangzhou Shen
- 1 School of Life Sciences and Biotechnology, Shanghai Jiao Tong University, 800 Dongchuan Road, Shanghai 200240, P. R. China
| | - Jian Li
- 1 School of Life Sciences and Biotechnology, Shanghai Jiao Tong University, 800 Dongchuan Road, Shanghai 200240, P. R. China
| | - Ying Zhu
- 1 School of Life Sciences and Biotechnology, Shanghai Jiao Tong University, 800 Dongchuan Road, Shanghai 200240, P. R. China
| | - Zhuo Wang
- 1 School of Life Sciences and Biotechnology, Shanghai Jiao Tong University, 800 Dongchuan Road, Shanghai 200240, P. R. China
| |
Collapse
|
42
|
Stewart PA, Parapatics K, Welsh EA, Müller AC, Cao H, Fang B, Koomen JM, Eschrich SA, Bennett KL, Haura EB. A Pilot Proteogenomic Study with Data Integration Identifies MCT1 and GLUT1 as Prognostic Markers in Lung Adenocarcinoma. PLoS One 2015; 10:e0142162. [PMID: 26539827 PMCID: PMC4634858 DOI: 10.1371/journal.pone.0142162] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2015] [Accepted: 10/19/2015] [Indexed: 11/19/2022] Open
Abstract
We performed a pilot proteogenomic study to compare lung adenocarcinoma to lung squamous cell carcinoma using quantitative proteomics (6-plex TMT) combined with a customized Affymetrix GeneChip. Using MaxQuant software, we identified 51,001 unique peptides that mapped to 7,241 unique proteins and from these identified 6,373 genes with matching protein expression for further analysis. We found a minor correlation between gene expression and protein expression; both datasets were able to independently recapitulate known differences between the adenocarcinoma and squamous cell carcinoma subtypes. We found 565 proteins and 629 genes to be differentially expressed between adenocarcinoma and squamous cell carcinoma, with 113 of these consistently differentially expressed at both the gene and protein levels. We then compared our results to published adenocarcinoma versus squamous cell carcinoma proteomic data that we also processed with MaxQuant. We selected two proteins consistently overexpressed in squamous cell carcinoma in all studies, MCT1 (SLC16A1) and GLUT1 (SLC2A1), for further investigation. We found differential expression of these same proteins at the gene level in our study as well as in other public gene expression datasets. These findings combined with survival analysis of public datasets suggest that MCT1 and GLUT1 may be potential prognostic markers in adenocarcinoma and druggable targets in squamous cell carcinoma. Data are available via ProteomeXchange with identifier PXD002622.
Collapse
Affiliation(s)
- Paul A. Stewart
- Department of Thoracic Oncology, H. Lee Moffitt Cancer Center and Research Institute, Tampa, Florida, United States of America 33612
| | - Katja Parapatics
- CeMM Research Center for Molecular Medicine of the Austrian Academy of Sciences, Lazarettgasse 14, 1090 Vienna, Austria
| | - Eric A. Welsh
- Cancer Informatics Core Facility, H. Lee Moffitt Cancer Center and Research Institute, Tampa, Florida, United States of America 33612
| | - André C. Müller
- CeMM Research Center for Molecular Medicine of the Austrian Academy of Sciences, Lazarettgasse 14, 1090 Vienna, Austria
| | - Haoyun Cao
- Department of Biostatistics and Bioinformatics, H. Lee Moffitt Cancer Center and Research Institute, Tampa, Florida, United States of America 33612
| | - Bin Fang
- Proteomics Core Facility, H. Lee Moffitt Cancer Center and Research Institute, Tampa, Florida, United States of America 33612
| | - John M. Koomen
- Proteomics Core Facility, H. Lee Moffitt Cancer Center and Research Institute, Tampa, Florida, United States of America 33612
- Department of Molecular Oncology, H. Lee Moffitt Cancer Center and Research Institute, Tampa, Florida, United States of America 33612
| | - Steven A. Eschrich
- Cancer Informatics Core Facility, H. Lee Moffitt Cancer Center and Research Institute, Tampa, Florida, United States of America 33612
- Department of Biostatistics and Bioinformatics, H. Lee Moffitt Cancer Center and Research Institute, Tampa, Florida, United States of America 33612
| | - Keiryn L. Bennett
- CeMM Research Center for Molecular Medicine of the Austrian Academy of Sciences, Lazarettgasse 14, 1090 Vienna, Austria
| | - Eric B. Haura
- Department of Thoracic Oncology, H. Lee Moffitt Cancer Center and Research Institute, Tampa, Florida, United States of America 33612
- * E-mail:
| |
Collapse
|
43
|
Insights from the Molecular Docking of Hydrolytic Products of Methyl Isocyanate (MIC) to Inhibition of Human Immune Proteins. Interdiscip Sci 2015; 7:287-94. [DOI: 10.1007/s12539-015-0012-3] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2013] [Revised: 02/08/2014] [Accepted: 07/08/2014] [Indexed: 01/07/2023]
|
44
|
Dai L, Noverr MC, Parsons C, Kaleeba JAR, Qin Z. xCT, not just an amino-acid transporter: a multi-functional regulator of microbial infection and associated diseases. Front Microbiol 2015; 6:120. [PMID: 25745420 PMCID: PMC4333839 DOI: 10.3389/fmicb.2015.00120] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2014] [Accepted: 01/30/2015] [Indexed: 12/23/2022] Open
Abstract
Expression of xCT, a component of the xc– amino-acid transporter, is essential for the uptake of cystine required for intracellular glutathione (GSH) synthesis and maintenance of the intracellular redox balance. Therefore, xCT plays an important role not only in the survival of somatic and immune cells, but also in other aspects of tumorigenesis, including the growth and malignant progression of cancer cells, resistance to anticancer drugs, and protection of normal cells against oxidative damage induced by carcinogens. xCT also functions as a factor required for infection by Kaposi’s sarcoma-associated herpesvirus (KSHV), the causative agent of Kaposi’s sarcoma (KS) and other lymphoproliferative diseases associated with HIV/AIDS. In spite of these advances, our understanding of the role of xCT in the pathogenesis of infectious diseases is still limited. Therefore, this review will summarize recent findings about the functions of xCT in diseases associated with microbial (bacterial or viral) infections, in particular KSHV-associated malignancies. We will also discuss the remaining questions, future directions, as well as evidence that supports the potential benefits of exploring system xc– as a target for prevention and clinical management of microbial diseases and cancer.
Collapse
Affiliation(s)
- Lu Dai
- Research Center for Translational Medicine and Key Laboratory of Arrhythmias of the Ministry of Education of China, East Hospital, Tongji University School of Medicine , Shanghai, China ; Department of Medicine, Louisiana State University Health Sciences Center, Louisiana Cancer Research Center , New Orleans, LA, USA
| | - Mairi C Noverr
- Department of Oral and Craniofacial Biology, Louisiana State University Health Sciences Center, Louisiana Cancer Research Center , New Orleans, LA, USA
| | - Chris Parsons
- Department of Medicine, Louisiana State University Health Sciences Center, Louisiana Cancer Research Center , New Orleans, LA, USA
| | - Johnan A R Kaleeba
- Department of Microbiology and Immunology, Uniformed Services University of the Health Sciences , Bethesda, MD, USA
| | - Zhiqiang Qin
- Research Center for Translational Medicine and Key Laboratory of Arrhythmias of the Ministry of Education of China, East Hospital, Tongji University School of Medicine , Shanghai, China ; Department of Microbiology, Immunology, and Parasitology, Louisiana State University Health Sciences Center, Louisiana Cancer Research Center , New Orleans, LA, USA
| |
Collapse
|
45
|
Johnson C, Pankratz VS, Velazquez AI, Aakre JA, Loprinzi CL, Staff NP, Windebank AJ, Yang P. Candidate pathway-based genetic association study of platinum and platinum-taxane related toxicity in a cohort of primary lung cancer patients. J Neurol Sci 2015; 349:124-8. [PMID: 25586538 PMCID: PMC4334320 DOI: 10.1016/j.jns.2014.12.041] [Citation(s) in RCA: 46] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2014] [Revised: 12/19/2014] [Accepted: 12/29/2014] [Indexed: 11/18/2022]
Abstract
BACKGROUND Chemotherapy-induced peripheral neuropathy (CIPN) is a common toxicity secondary to chemotherapy. Genetic factors may be important in predisposing patients to this adverse effect. PATIENTS AND METHODS We studied 950 primary lung cancer patients, who received platinum or platinum-combination drug chemotherapy and who had DNA available for study. We analyzed epidemiological risk factors in 279 CIPN patients and 456 non-CIPN patients and genetic risk factors in 141 CIPN patients and 259 non-CIPN patients. The risk factors studied included demographic, diagnostic, and treatment data, as well as 174 tag SNPs (single nucleotide polymorphisms) across 43 candidate genes in the glutathione, cell cycle, DNA repair, cell signaling, and apoptosis pathways. RESULTS Patients who had diabetes mellitus were more likely to have CIPN (p=0.0002). Other epidemiologic risk factors associated with CIPN included number of cycles (p=0.0004) and type of concurrent chemotherapy (p<0.001). SNPs most associated with CIPN were in glutathione peroxidase 7 (GPX7) gene (p values 0.0015 and 0.0028, unadjusted and adjusted) and in ATP-binding cassette sub-family C member 4 (ABCC4) gene (p values 0.037 and 0.006, unadjusted and adjusted). We also found other suggestive associations in methyl-o-guanine-methyl-transferase (MGMT) and glutathione-S-transferase (GST) isoforms. CONCLUSIONS Epidemiological and genetic risk factors associated with CIPN in this cohort, included the type of chemotherapy drug, intensity of chemotherapy treatment, and genes known to be associated with chemotherapy resistance. These findings suggest that differentiating between cytotoxic and neurotoxic mechanisms of chemotherapy drugs is challenging but represents an important step toward individualized therapy and improving quality of life for patients.
Collapse
Affiliation(s)
- Cassandra Johnson
- Departments of Neurology, Oncology, Health Sciences Research, Division of Biomedical Statistics and Informatics, Mayo Clinic College of Medicine, Rochester, MN, USA
| | - Vernon S Pankratz
- Departments of Neurology, Oncology, Health Sciences Research, Division of Biomedical Statistics and Informatics, Mayo Clinic College of Medicine, Rochester, MN, USA
| | - Ana I Velazquez
- Departments of Neurology, Oncology, Health Sciences Research, Division of Biomedical Statistics and Informatics, Mayo Clinic College of Medicine, Rochester, MN, USA
| | - Jeremiah A Aakre
- Departments of Neurology, Oncology, Health Sciences Research, Division of Biomedical Statistics and Informatics, Mayo Clinic College of Medicine, Rochester, MN, USA
| | - Charles L Loprinzi
- Departments of Neurology, Oncology, Health Sciences Research, Division of Biomedical Statistics and Informatics, Mayo Clinic College of Medicine, Rochester, MN, USA
| | - Nathan P Staff
- Departments of Neurology, Oncology, Health Sciences Research, Division of Biomedical Statistics and Informatics, Mayo Clinic College of Medicine, Rochester, MN, USA
| | - Anthony J Windebank
- Departments of Neurology, Oncology, Health Sciences Research, Division of Biomedical Statistics and Informatics, Mayo Clinic College of Medicine, Rochester, MN, USA.
| | - Ping Yang
- Departments of Neurology, Oncology, Health Sciences Research, Division of Biomedical Statistics and Informatics, Mayo Clinic College of Medicine, Rochester, MN, USA
| |
Collapse
|
46
|
Tripathi MK, Yasir M, Gurjar VS, Bose P, Dubey A, Shrivastava R. Insights from the molecular docking of hydrolytic products of methyl iso cyanate (MIC) to inhibition of human immune proteins. Interdiscip Sci 2015. [PMID: 25595583 DOI: 10.1007/s12539-013-0217-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2013] [Revised: 02/08/2014] [Accepted: 07/08/2014] [Indexed: 06/04/2023]
Abstract
This study is an attempt to find the reason for immunological suppression in victims of Bhopal gas tragedy during 1984 against Mycobacterium Tuberculosis (Mtb) infection. Here we tried to understand this problem by studying interactions between immune proteins associated with susceptibility to Tuberculosis and hydrolytic products of methyl isocyanate (MIC) released during the tragedy.The hydrolytic products of methyl isocyanate (MIC) i.e. dimethyl urea, trimethyl urea and trimethyl isocyanurate was docked to different human immune proteins against Mtb using autodock 4.0. Results shows that all hydrolytic product (dimethyl urea, trimethyl urea and trimethylisocyanurate) strongly inhibits to CD40 ligand and their binding energies were found to be ΔG -3.51, -3.79, -4.55 (Kcal/Mole) respectively. Further to check the stability of docked complex we performed the molecular dynamics simulation study which also shows that CD40 Ligand was maximum inhibited by trimethylisocyanurate, has a role in the macrophage activation for the destruction of Mycobacterium tuberculosis. The present study may lead to better understanding of human immune protein inhibition by hydrolytic product of methyl isocyanate (MIC).
Collapse
Affiliation(s)
- Manish Kumar Tripathi
- Department of Biological Sciences and Engineering, Maulana Azad National Institute of Technology, Bhopal, 462051, India
| | | | | | | | | | | |
Collapse
|
47
|
Genetic variations underlying self-reported physical functioning: a review. Qual Life Res 2014; 24:1163-77. [PMID: 25387867 DOI: 10.1007/s11136-014-0844-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/29/2014] [Indexed: 10/24/2022]
Abstract
PURPOSE Genetic associations with self-reported physical functioning (SPF) are less well-studied than genetic associations with performance-measured physical functioning (PPF). We review the literature on the associations of genetic variations on SPF. We provide an overview of SPF assessment, genetic contributions to SPF including heritability, effects of genetic variations and mutations, and effects of interventions on the gene-SPF relationship. We also aim to provide directions for future research. METHODS A computerized literature search using PubMed, Web of Science, and PsychInfo was conducted to select relevant literature published up to November 2013. Inclusion criteria were the use of an SPF questionnaire, original articles in English on human subjects, published in peer-reviewed journals and reporting significant associations between SPF and the genome. RESULTS Nineteen articles were included. SPF was commonly assessed with the Short Form-36 questionnaire involving mainly convenience samples of either older persons or chronically ill. Heritability estimates were 10-30 %. Candidate genes associated with SPF could be ascribed to biological pathways associated with neurodegeneration, physiological systems regulation, or cell regulation. The APOE gene associated with neurodegeneration was most studied (n = 3). Three papers included both SPF and PPF assessments. No genome-wide association study on SPF has been conducted. CONCLUSIONS Associations between SPF and the genome have been investigated in selected populations in a limited number of publications. Future research should consider increasing sample variation and incorporate both SPF and PPF assessments. Also, longitudinal studies should be conducted in order to elicit stronger conclusions regarding the genetic associations with SPF.
Collapse
|
48
|
Papadaki C, Sfakianaki M, Lagoudaki E, Giagkas G, Ioannidis G, Trypaki M, Tsakalaki E, Voutsina A, Koutsopoulos A, Mavroudis D, Georgoulias V, Souglakos J. PKM2 as a biomarker for chemosensitivity to front-line platinum-based chemotherapy in patients with metastatic non-small-cell lung cancer. Br J Cancer 2014; 111:1757-64. [PMID: 25233397 PMCID: PMC4453739 DOI: 10.1038/bjc.2014.492] [Citation(s) in RCA: 42] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Revised: 07/17/2014] [Accepted: 08/13/2014] [Indexed: 01/06/2023] Open
Abstract
BACKGROUND Tumour cells exclusively express the embryonic M2 isoform of pyruvate kinase (PKM2). PKM2 expression levels have been correlated with the effect of platinum compounds in cancer cell lines and xenograft models. The potential predictive role of PKM2 in patients with metastatic/advanced non-small-cell lung cancer (NSCLC) receiving platinum-based chemotherapy as first-line was investigated. METHODS Quantitative real-time PCR was used to assess the expression of PKM2 in tumour samples from 148 and 157 NSCLC patients in the training and the validation set, respectively. All patients received front-line platinum-based chemotherapy. PKM2 mRNA expression was also analysed in a control group of 85 NSCLC patients treated with non-platinum containing regimens. RESULTS In the training set, high PKM2 mRNA levels were associated with decreased progression-free survival (PFS; 4.9 months vs 6.4, P=0.006), overall survival (OS; 10.1 vs 17.0 months, P=0.01) and disease control rate (DCR; 57.7% vs 74.3%; P=0.021) compared to patients with low PKM2 levels. In the validation set, high PKM2 mRNA levels were also associated with deceased PFS (3.7 vs 5.9 months, P=0.006), OS (8.3 vs 16.8 months, P=0.003) and DCR (57.7% vs 70.9%; P=0.049) compared to those with low PKM2 mRNA levels. There was no correlation between the PKM2 mRNA levels and the PFS (5.6 vs 5.9, P=0.43) or the OS (9.8 vs 10.1, P=0.51) in the control group. Multivariate analysis revealed high PKM2 mRNA expression as an independent predictive factor for the poor patients' outcome. CONCLUSIONS PKM2 expression may be a predictive biomarker of platinum sensitivity in advanced NSCLC patients treated with platinum-based chemotherapy.
Collapse
MESH Headings
- Adult
- Aged
- Aged, 80 and over
- Antibodies, Monoclonal, Humanized/administration & dosage
- Antineoplastic Combined Chemotherapy Protocols/therapeutic use
- Bevacizumab
- Biomarkers, Tumor/genetics
- Biomarkers, Tumor/metabolism
- Carcinoma, Non-Small-Cell Lung/drug therapy
- Carcinoma, Non-Small-Cell Lung/metabolism
- Carcinoma, Non-Small-Cell Lung/mortality
- Carcinoma, Non-Small-Cell Lung/secondary
- Carcinoma, Squamous Cell/drug therapy
- Carcinoma, Squamous Cell/metabolism
- Carcinoma, Squamous Cell/mortality
- Carcinoma, Squamous Cell/secondary
- Carrier Proteins/genetics
- Carrier Proteins/metabolism
- Deoxycytidine/administration & dosage
- Deoxycytidine/analogs & derivatives
- Docetaxel
- Female
- Follow-Up Studies
- Glutamates/administration & dosage
- Guanine/administration & dosage
- Guanine/analogs & derivatives
- Humans
- Immunoenzyme Techniques
- Lung Neoplasms/drug therapy
- Lung Neoplasms/metabolism
- Lung Neoplasms/mortality
- Lung Neoplasms/pathology
- Lymphatic Metastasis
- Male
- Membrane Proteins/genetics
- Membrane Proteins/metabolism
- Middle Aged
- Neoplasm Staging
- Pemetrexed
- Platinum/administration & dosage
- Prognosis
- RNA, Messenger/genetics
- Real-Time Polymerase Chain Reaction
- Reverse Transcriptase Polymerase Chain Reaction
- Survival Rate
- Taxoids/administration & dosage
- Thyroid Hormones/genetics
- Thyroid Hormones/metabolism
- Gemcitabine
- Thyroid Hormone-Binding Proteins
Collapse
Affiliation(s)
- C Papadaki
- Laboratory of Tumor Cell Biology, School of Medicine, University of Crete, Heraklion, 71003 Crete, Greece
| | - M Sfakianaki
- Laboratory of Tumor Cell Biology, School of Medicine, University of Crete, Heraklion, 71003 Crete, Greece
| | - E Lagoudaki
- Department of Pathology, University General Hospital of Heraklion, Heraklion, 71110 Crete, Greece
| | - G Giagkas
- Laboratory of Tumor Cell Biology, School of Medicine, University of Crete, Heraklion, 71003 Crete, Greece
| | - G Ioannidis
- Laboratory of Tumor Cell Biology, School of Medicine, University of Crete, Heraklion, 71003 Crete, Greece
| | - M Trypaki
- Laboratory of Tumor Cell Biology, School of Medicine, University of Crete, Heraklion, 71003 Crete, Greece
| | - E Tsakalaki
- Laboratory of Tumor Cell Biology, School of Medicine, University of Crete, Heraklion, 71003 Crete, Greece
| | - A Voutsina
- Laboratory of Tumor Cell Biology, School of Medicine, University of Crete, Heraklion, 71003 Crete, Greece
| | - A Koutsopoulos
- Department of Pathology, University General Hospital of Heraklion, Heraklion, 71110 Crete, Greece
| | - D Mavroudis
- Laboratory of Tumor Cell Biology, School of Medicine, University of Crete, Heraklion, 71003 Crete, Greece
- Department of Medical Oncology, University General Hospital of Heraklion, Heraklion, 71110 Crete, Greece
| | - V Georgoulias
- Laboratory of Tumor Cell Biology, School of Medicine, University of Crete, Heraklion, 71003 Crete, Greece
- Department of Medical Oncology, University General Hospital of Heraklion, Heraklion, 71110 Crete, Greece
| | - J Souglakos
- Laboratory of Tumor Cell Biology, School of Medicine, University of Crete, Heraklion, 71003 Crete, Greece
- Department of Medical Oncology, University General Hospital of Heraklion, Heraklion, 71110 Crete, Greece
| |
Collapse
|
49
|
Bonder MJ, Kasela S, Kals M, Tamm R, Lokk K, Barragan I, Buurman WA, Deelen P, Greve JW, Ivanov M, Rensen SS, van Vliet-Ostaptchouk JV, Wolfs MG, Fu J, Hofker MH, Wijmenga C, Zhernakova A, Ingelman-Sundberg M, Franke L, Milani L. Genetic and epigenetic regulation of gene expression in fetal and adult human livers. BMC Genomics 2014; 15:860. [PMID: 25282492 PMCID: PMC4287518 DOI: 10.1186/1471-2164-15-860] [Citation(s) in RCA: 99] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2014] [Accepted: 09/24/2014] [Indexed: 01/07/2023] Open
Abstract
Background The liver plays a central role in the maintenance of homeostasis and health in general. However, there is substantial inter-individual variation in hepatic gene expression, and although numerous genetic factors have been identified, less is known about the epigenetic factors. Results By analyzing the methylomes and transcriptomes of 14 fetal and 181 adult livers, we identified 657 differentially methylated genes with adult-specific expression, these genes were enriched for transcription factor binding sites of HNF1A and HNF4A. We also identified 1,000 genes specific to fetal liver, which were enriched for GATA1, STAT5A, STAT5B and YY1 binding sites. We saw strong liver-specific effects of single nucleotide polymorphisms on both methylation levels (28,447 unique CpG sites (meQTL)) and gene expression levels (526 unique genes (eQTL)), at a false discovery rate (FDR) < 0.05. Of the 526 unique eQTL associated genes, 293 correlated significantly not only with genetic variation but also with methylation levels. The tissue-specificities of these associations were analyzed in muscle, subcutaneous adipose tissue and visceral adipose tissue. We observed that meQTL were more stable between tissues than eQTL and a very strong tissue-specificity for the identified associations between CpG methylation and gene expression. Conclusions Our analyses generated a comprehensive resource of factors involved in the regulation of hepatic gene expression, and allowed us to estimate the proportion of variation in gene expression that could be attributed to genetic and epigenetic variation, both crucial to understanding differences in drug response and the etiology of liver diseases. Electronic supplementary material The online version of this article (doi:10.1186/1471-2164-15-860) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | - Lude Franke
- University of Groningen, University Medical Center Groningen, Department of Genetics, Hanzeplein 1, 9700 RB Groningen, the Netherlands.
| | | |
Collapse
|
50
|
Singly protonated dehydronorcantharidin silver coordination polymer induces apoptosis of lung cancer cells via reactive oxygen species-mediated mitochondrial pathway. Eur J Med Chem 2014; 86:1-11. [DOI: 10.1016/j.ejmech.2014.08.052] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2013] [Revised: 07/11/2014] [Accepted: 08/14/2014] [Indexed: 12/25/2022]
|