1
|
López-Estévez AM, Sanjurjo L, Turrero Á, Arriaga I, Abrescia NGA, Poveda A, Jiménez-Barbero J, Vidal A, Torres D, Alonso MJ. Nanotechnology-assisted intracellular delivery of antibody as a precision therapy approach for KRAS-driven tumors. J Control Release 2024; 373:277-292. [PMID: 39019086 DOI: 10.1016/j.jconrel.2024.07.032] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2024] [Revised: 07/03/2024] [Accepted: 07/12/2024] [Indexed: 07/19/2024]
Abstract
The Kirsten Rat Sarcoma Virus (KRAS) oncoprotein, one of the most prevalent mutations in cancer, has been deemed undruggable for decades. The hypothesis of this work was that delivering anti-KRAS monoclonal antibody (mAb) at the intracellular level could effectively target the KRAS oncoprotein. To reach this goal, we designed and developed tLyP1-targeted palmitoyl hyaluronate (HAC16)-based nanoassemblies (HANAs) adapted for the association of bevacizumab as a model mAb. Selected candidates with adequate physicochemical properties (below 150 nm, neutral surface charge), and high drug loading capacity (>10%, w/w) were adapted to entrap the antiKRASG12V mAb. The resulting antiKRASG12V-loaded HANAs exhibited a bilayer composed of HAC16 polymer and phosphatidylcholine (PC) enclosing a hydrophilic core, as evidenced by cryogenic-transmission electron microscopy (cryo-TEM) and X-ray photoelectron spectroscopy (XPS). Selected prototypes were found to efficiently engage the target KRASG12V and, inhibit proliferation and colony formation in KRASG12V-mutated lung cancer cell lines. In vivo, a selected formulation exhibited a tumor growth reduction in a pancreatic tumor-bearing mouse model. In brief, this study offers evidence of the potential to use nanotechnology for developing anti-KRAS precision therapy and provides a rational framework for advancing mAb intracellular delivery against intracellular targets.
Collapse
Affiliation(s)
- Ana M López-Estévez
- Center for Research in Molecular Medicine and Chronic Diseases (CiMUS), University of Santiago de Compostela, 15782 Santiago de Compostela, Spain; Health Research Institute of Santiago de Compostela, 15782 Santiago de Compostela, Spain; Department of Pharmacology, Pharmacy and Pharmaceutical Technology, School of Pharmacy, University of Santiago de Compostela, 15782 Santiago de Compostela, Spain
| | - Lucía Sanjurjo
- Center for Research in Molecular Medicine and Chronic Diseases (CiMUS), University of Santiago de Compostela, 15782 Santiago de Compostela, Spain; Health Research Institute of Santiago de Compostela, 15782 Santiago de Compostela, Spain
| | - Ángela Turrero
- Center for Research in Molecular Medicine and Chronic Diseases (CiMUS), University of Santiago de Compostela, 15782 Santiago de Compostela, Spain
| | - Iker Arriaga
- Structure and Cell Biology of Viruses Lab, CIC bioGUNE, Basque Research and Technology Alliance (BRTA), 48160 Derio, Spain
| | - Nicola G A Abrescia
- Structure and Cell Biology of Viruses Lab, CIC bioGUNE, Basque Research and Technology Alliance (BRTA), 48160 Derio, Spain; Ikerbasque, Basque Foundation for Science, 48009 Bilbao, Spain
| | - Ana Poveda
- Chemical Glycobiology Laboratory, CIC bioGUNE, Basque Research and Technology Alliance (BRTA), 48160 Derio, Spain
| | - Jesús Jiménez-Barbero
- Ikerbasque, Basque Foundation for Science, 48009 Bilbao, Spain; Chemical Glycobiology Laboratory, CIC bioGUNE, Basque Research and Technology Alliance (BRTA), 48160 Derio, Spain
| | - Anxo Vidal
- Center for Research in Molecular Medicine and Chronic Diseases (CiMUS), University of Santiago de Compostela, 15782 Santiago de Compostela, Spain
| | - Dolores Torres
- Department of Pharmacology, Pharmacy and Pharmaceutical Technology, School of Pharmacy, University of Santiago de Compostela, 15782 Santiago de Compostela, Spain
| | - María José Alonso
- Center for Research in Molecular Medicine and Chronic Diseases (CiMUS), University of Santiago de Compostela, 15782 Santiago de Compostela, Spain; Health Research Institute of Santiago de Compostela, 15782 Santiago de Compostela, Spain; Department of Pharmacology, Pharmacy and Pharmaceutical Technology, School of Pharmacy, University of Santiago de Compostela, 15782 Santiago de Compostela, Spain.
| |
Collapse
|
2
|
Zhou Q, Xiang J, Qiu N, Wang Y, Piao Y, Shao S, Tang J, Zhou Z, Shen Y. Tumor Abnormality-Oriented Nanomedicine Design. Chem Rev 2023; 123:10920-10989. [PMID: 37713432 DOI: 10.1021/acs.chemrev.3c00062] [Citation(s) in RCA: 53] [Impact Index Per Article: 26.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/17/2023]
Abstract
Anticancer nanomedicines have been proven effective in mitigating the side effects of chemotherapeutic drugs. However, challenges remain in augmenting their therapeutic efficacy. Nanomedicines responsive to the pathological abnormalities in the tumor microenvironment (TME) are expected to overcome the biological limitations of conventional nanomedicines, enhance the therapeutic efficacies, and further reduce the side effects. This Review aims to quantitate the various pathological abnormalities in the TME, which may serve as unique endogenous stimuli for the design of stimuli-responsive nanomedicines, and to provide a broad and objective perspective on the current understanding of stimuli-responsive nanomedicines for cancer treatment. We dissect the typical transport process and barriers of cancer drug delivery, highlight the key design principles of stimuli-responsive nanomedicines designed to tackle the series of barriers in the typical drug delivery process, and discuss the "all-into-one" and "one-for-all" strategies for integrating the needed properties for nanomedicines. Ultimately, we provide insight into the challenges and future perspectives toward the clinical translation of stimuli-responsive nanomedicines.
Collapse
Affiliation(s)
- Quan Zhou
- Zhejiang Key Laboratory of Smart Biomaterials and Center for Bionanoengineering, College of Chemical and Biological Engineering, Zhejiang University, Hangzhou 310058, China
- Key Laboratory of Biomass Chemical Engineering of Ministry of Education, College of Chemical and Biological Engineering, Zhejiang University, Hangzhou 310058, China
- Department of Cell Biology, Zhejiang University School of Medicine, Zhejiang University, Hangzhou 310058, China
| | - Jiajia Xiang
- Zhejiang Key Laboratory of Smart Biomaterials and Center for Bionanoengineering, College of Chemical and Biological Engineering, Zhejiang University, Hangzhou 310058, China
- Key Laboratory of Biomass Chemical Engineering of Ministry of Education, College of Chemical and Biological Engineering, Zhejiang University, Hangzhou 310058, China
- Department of Cell Biology, Zhejiang University School of Medicine, Zhejiang University, Hangzhou 310058, China
| | - Nasha Qiu
- Zhejiang Key Laboratory of Smart Biomaterials and Center for Bionanoengineering, College of Chemical and Biological Engineering, Zhejiang University, Hangzhou 310058, China
- Key Laboratory of Biomass Chemical Engineering of Ministry of Education, College of Chemical and Biological Engineering, Zhejiang University, Hangzhou 310058, China
| | - Yechun Wang
- Department of Cell Biology, Zhejiang University School of Medicine, Zhejiang University, Hangzhou 310058, China
| | - Ying Piao
- Zhejiang Key Laboratory of Smart Biomaterials and Center for Bionanoengineering, College of Chemical and Biological Engineering, Zhejiang University, Hangzhou 310058, China
- Key Laboratory of Biomass Chemical Engineering of Ministry of Education, College of Chemical and Biological Engineering, Zhejiang University, Hangzhou 310058, China
| | - Shiqun Shao
- Zhejiang Key Laboratory of Smart Biomaterials and Center for Bionanoengineering, College of Chemical and Biological Engineering, Zhejiang University, Hangzhou 310058, China
- Key Laboratory of Biomass Chemical Engineering of Ministry of Education, College of Chemical and Biological Engineering, Zhejiang University, Hangzhou 310058, China
| | - Jianbin Tang
- Zhejiang Key Laboratory of Smart Biomaterials and Center for Bionanoengineering, College of Chemical and Biological Engineering, Zhejiang University, Hangzhou 310058, China
- Key Laboratory of Biomass Chemical Engineering of Ministry of Education, College of Chemical and Biological Engineering, Zhejiang University, Hangzhou 310058, China
| | - Zhuxian Zhou
- Zhejiang Key Laboratory of Smart Biomaterials and Center for Bionanoengineering, College of Chemical and Biological Engineering, Zhejiang University, Hangzhou 310058, China
- Key Laboratory of Biomass Chemical Engineering of Ministry of Education, College of Chemical and Biological Engineering, Zhejiang University, Hangzhou 310058, China
| | - Youqing Shen
- Zhejiang Key Laboratory of Smart Biomaterials and Center for Bionanoengineering, College of Chemical and Biological Engineering, Zhejiang University, Hangzhou 310058, China
- Key Laboratory of Biomass Chemical Engineering of Ministry of Education, College of Chemical and Biological Engineering, Zhejiang University, Hangzhou 310058, China
- State Key Laboratory of Chemical Engineering, Zhejiang University, Hangzhou 310058, China
| |
Collapse
|
3
|
Noreen S, Hasan S, Ishtiaq M, Ghumman SA. Phospholipases in cancer progression and metastasis. PHOSPHOLIPASES IN PHYSIOLOGY AND PATHOLOGY 2023:303-313. [DOI: 10.1016/b978-0-323-95697-0.00021-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/03/2025]
|
4
|
Wang H, Luo Y, Chen H, Hou H, Hu Q, Ji M. Non-Targeted Serum Lipidomics Analysis and Potential Biomarkers of Laryngeal Cancer Based on UHPLC-QTOF-MS. Metabolites 2022; 12:1087. [PMID: 36355170 PMCID: PMC9695307 DOI: 10.3390/metabo12111087] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2022] [Revised: 11/03/2022] [Accepted: 11/07/2022] [Indexed: 08/31/2023] Open
Abstract
Laryngeal cancer is a common head and neck malignant cancer type. However, effective biomarkers for diagnosis are lacking and pathogenesis is unclear. Lipidomics is a powerful tool for identifying biomarkers and explaining disease mechanisms. Hence, in this study, non-targeted lipidomics based on ultra-performance liquid chromatography-quadrupole time of flight-mass spectrometry (UHPLC-QTOF-MS) were applied to screen the differential lipid metabolites in serum and allowed for exploration of the remodeled lipid metabolism of laryngeal cancer, laryngeal benign tumor patients, and healthy crowds. Multivariate analysis and univariate analysis were combined to screen for differential lipid metabolites among the three groups. The results showed that, across a total of 57 lipid metabolic markers that were screened, the regulation of the lipid metabolism network occurred mainly in phosphatidylcholine (PC), lysophosphatidylcholine (LPC), and sphingomyelin (SM) metabolism. Of note, the concentration levels of sphingolipids 42:2 (SM 42:2) and sphingolipids 42:3 (SM 42:3) correlated with laryngeal cancer progression and were both significantly different among the three groups. Both of them could be considered as potential biomarkers for diagnosis and indicators for monitoring the progression of laryngeal cancer. From the perspective of lipidomics, this study not only revealed the regulatory changes in the lipid metabolism network, but also provided a new possibility for screening biomarkers in laryngeal cancer.
Collapse
Affiliation(s)
- Haoyue Wang
- Anhui Institute of Optics and Fine Mechanics, Hefei Institutes of Physical Science, Chinese Academy of Sciences, Hefei 230031, China
- Science Island Branch, Graduate School of University of Science and Technology of China, Hefei 230026, China
- Key Laboratory of Tobacco Biological Effects, China National Tobacco Quality Supervision & Test Center, Zhengzhou 450001, China
| | - Yanbo Luo
- Key Laboratory of Tobacco Biological Effects, China National Tobacco Quality Supervision & Test Center, Zhengzhou 450001, China
| | - Huan Chen
- Key Laboratory of Tobacco Biological Effects, China National Tobacco Quality Supervision & Test Center, Zhengzhou 450001, China
| | - Hongwei Hou
- Key Laboratory of Tobacco Biological Effects, China National Tobacco Quality Supervision & Test Center, Zhengzhou 450001, China
| | - Qingyuan Hu
- Key Laboratory of Tobacco Biological Effects, China National Tobacco Quality Supervision & Test Center, Zhengzhou 450001, China
| | - Min Ji
- Anhui Institute of Optics and Fine Mechanics, Hefei Institutes of Physical Science, Chinese Academy of Sciences, Hefei 230031, China
| |
Collapse
|
5
|
Xiang J, Liu X, Yuan G, Zhang R, Zhou Q, Xie T, Shen Y. Nanomedicine from amphiphilizedprodrugs: Concept and clinical translation. Adv Drug Deliv Rev 2021; 179:114027. [PMID: 34732344 DOI: 10.1016/j.addr.2021.114027] [Citation(s) in RCA: 39] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2021] [Revised: 09/30/2021] [Accepted: 10/27/2021] [Indexed: 12/15/2022]
Abstract
Nanomedicines generally consisting of carrier materials with small fractions of active pharmaceutical ingredients (API) have long been used to improve the pharmacokinetics and biodistributions, augment the therapeutic efficacies and mitigate the side effects. Amphiphilizing hydrophobic/hydrophilic drugs to prodrugs capable of self-assembly into well-defined nanostructures has emerged as a facile approach to fabricating nanomedicines because this amphiphilized prodrug (APD) strategy presents many advantages, including minimized use of inert carrier materials, well-characterized prodrug structures, fixed and high drug loading contents, 100% loading efficiency, and burst-free but controlled drug release. This review comprehensively summarizes recent advances in APDs and their nanomedicines, from the rationale and the stimuli-responsive linker chemistry for on-demand drug release to their progress to the clinics, clinical performance of APDs, as well as the challenges and perspective on future development.
Collapse
|
6
|
Wang Q, Wei N, Guo J, Feng K, Wong YK, Zhang J, Wang J, Sun X. Hemin-lipid assembly as an artemisinin oral delivery system for enhanced cancer chemotherapy and immunotherapy. NANOSCALE 2021; 13:13231-13240. [PMID: 34477731 DOI: 10.1039/d1nr01302e] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/13/2023]
Abstract
Although artemisinin (ART) has shown initial promise in cancer therapy, its therapeutic efficacy is limited by its low tumor inhibitory efficacy and unfavorable distribution. Considering the important role of heme in the specific parasite-killing effect of ART, we designed a liposomal nanostructure self-assembled from hemin-lipid (Hemesome) to co-deliver ART and hemin for cancer therapy. The synergistic chemotherapeutic and immunotherapeutic effects of hemin and ART were demonstrated both in vitro and in vivo. The liposome-like structure was relatively stable in the blood circulation and gastrointestinal tract environment, but dissociated in the tumor cell environment. The folic acid (FA) modification not only increased their efficiency for transport across the epithelium, but also increased their tumor accumulation. In mouse models, following oral administration of FA-Hemesome-ART nanoparticles (5 mg kg-1 ART in total) every other day and intraperitoneal injection with a programmed death-ligand 1 antibody (aPD-L1, 70 μg per mouse in total), MC38 tumors were completely inhibited within 30 days. The cured mice remained tumor-free 30 days after rechallenging them with another inoculation of MC38 cells due to the strong immune memory effect.
Collapse
Affiliation(s)
- Qing Wang
- State Key Laboratory of Natural Medicines, Key Laboratory of Drug Quality Control and Pharmacovigilance, Department of Pharmaceutical Analysis, China Pharmaceutical University, Nanjing 210009, China
| | | | | | | | | | | | | | | |
Collapse
|
7
|
Jiang N, Zhang Z, Chen X, Zhang G, Wang Y, Pan L, Yan C, Yang G, Zhao L, Han J, Xue T. Plasma Lipidomics Profiling Reveals Biomarkers for Papillary Thyroid Cancer Diagnosis. Front Cell Dev Biol 2021; 9:682269. [PMID: 34235148 PMCID: PMC8255691 DOI: 10.3389/fcell.2021.682269] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2021] [Accepted: 05/28/2021] [Indexed: 11/30/2022] Open
Abstract
The objective of this study was to identify potential biomarkers and possible metabolic pathways of malignant and benign thyroid nodules through lipidomics study. A total of 47 papillary thyroid carcinomas (PTC) and 33 control check (CK) were enrolled. Plasma samples were collected for UPLC-Q-TOF MS system detection, and then OPLS-DA model was used to identify differential metabolites. Based on classical statistical methods and machine learning, potential biomarkers were characterized and related metabolic pathways were identified. According to the metabolic spectrum, 13 metabolites were identified between PTC group and CK group, and a total of five metabolites were obtained after further screening. Its metabolic pathways were involved in glycerophospholipid metabolism, linoleic acid metabolism, alpha-linolenic acid metabolism, glycosylphosphatidylinositol (GPI)—anchor biosynthesis, Phosphatidylinositol signaling system and the metabolism of arachidonic acid metabolism. The metabolomics method based on PROTON nuclear magnetic resonance (NMR) had great potential for distinguishing normal subjects from PTC. GlcCer(d14:1/24:1), PE-NME (18:1/18:1), SM(d16:1/24:1), SM(d18:1/15:0), and SM(d18:1/16:1) can be used as potential serum markers for the diagnosis of PTC.
Collapse
Affiliation(s)
- Nan Jiang
- Department of General Surgery, First Hospital of Tsinghua University, Beijing, China
| | - Zhenya Zhang
- Department of General Surgery, First Hospital of Tsinghua University, Beijing, China
| | - Xianyang Chen
- BaoFeng Key Laboratory of Genetics and Metabolism, Beijing, China
| | - Guofen Zhang
- Department of General Surgery, First Hospital of Tsinghua University, Beijing, China
| | - Ying Wang
- Department of Oncology, Tai'an City Central Hospital, Tai'an, China
| | - Lijie Pan
- Department of General Surgery, First Hospital of Tsinghua University, Beijing, China
| | - Chengping Yan
- Department of General Surgery, First Hospital of Tsinghua University, Beijing, China
| | - Guoshan Yang
- Department of General Surgery, First Hospital of Tsinghua University, Beijing, China
| | - Li Zhao
- Department of General Surgery, First Hospital of Tsinghua University, Beijing, China
| | - Jiarui Han
- BaoFeng Key Laboratory of Genetics and Metabolism, Beijing, China
| | - Teng Xue
- Zhongguancun Biological and Medical Big Data Center, Beijing, China
| |
Collapse
|
8
|
Gergen AK, Jarrett MJ, Li A, White AM, Meng X, Fullerton DA, Weyant MJ. Secretory Phospholipase A 2 Inhibition Attenuates Adhesive Properties of Esophageal Barrett's Cells. J Surg Res 2020; 259:562-568. [PMID: 33261858 DOI: 10.1016/j.jss.2020.10.018] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2020] [Revised: 09/10/2020] [Accepted: 10/31/2020] [Indexed: 11/16/2022]
Abstract
BACKGROUND Gastroesophageal reflux and Barrett's esophagus are significant risk factors for the development of esophageal adenocarcinoma. Group IIa secretory phospholipase A2 (sPLA2) catalyzes the production of various proinflammatory metabolites and plays a critical role in promoting reflux-induced inflammatory changes within the distal esophagus. We hypothesized that inhibition of sPLA2 in human Barrett's cells would attenuate adhesion molecule expression via decreased activation of nuclear factor kappa B (NF-κB) and decrease cell proliferation, possibly mitigating the invasive potential of Barrett's esophagus. MATERIALS AND METHODS Normal human esophageal epithelial cells (HET1A) and Barrett's cells (CPB) were assayed for baseline sPLA2 expression. CPB cells were treated with a specific inhibitor of sPLA2 followed by tumor necrosis factor-α. Protein expression was evaluated using immunoblotting. Cell proliferation was assessed using an MTS cell proliferation assay kit. Statistical analysis was performed using the Student's t-test or analysis of variance, where appropriate. RESULTS CPB cells demonstrated higher baseline sPLA2 expression than HET1A cells (P = 0.0005). Treatment with 30 μM sPLA2 inhibitor significantly attenuated intercellular adhesion molecule-1 (P = 0.004) and vascular cell adhesion molecule-1 (P < 0.0001) expression as well as decreased NF-κB activation (P = 0.002). sPLA2 inhibition decreased cell proliferation in a dose-dependent manner (P < 0.001 for 15, 20, and 30 μM doses). CONCLUSIONS sPLA2 inhibition in human Barrett's cells decreases cellular adhesive properties and NF-κB activation as well as decreases cell proliferation, signifying downregulation of the inflammatory response and possible attenuation of cellular malignant potential. These findings identify sPLA2 inhibition as a potential chemopreventive target for premalignant lesions of the esophagus.
Collapse
Affiliation(s)
- Anna K Gergen
- University of Colorado School of Medicine, Department of Surgery, Division of Cardiothoracic Surgery, Aurora, Colorado.
| | - Michael J Jarrett
- University of Colorado School of Medicine, Department of Surgery, Division of Cardiothoracic Surgery, Aurora, Colorado
| | - Anqi Li
- University of Colorado School of Medicine, Department of Surgery, Division of Cardiothoracic Surgery, Aurora, Colorado
| | - Allana M White
- University of Colorado School of Medicine, Department of Surgery, Division of Cardiothoracic Surgery, Aurora, Colorado
| | - Xianzhong Meng
- University of Colorado School of Medicine, Department of Surgery, Division of Cardiothoracic Surgery, Aurora, Colorado
| | - David A Fullerton
- University of Colorado School of Medicine, Department of Surgery, Division of Cardiothoracic Surgery, Aurora, Colorado
| | - Michael J Weyant
- University of Colorado School of Medicine, Department of Surgery, Division of Cardiothoracic Surgery, Aurora, Colorado
| |
Collapse
|
9
|
Cheng F, Wen Z, Feng X, Wang X, Chen Y. A serum lipidomic strategy revealed potential lipid biomarkers for early-stage cervical cancer. Life Sci 2020; 260:118489. [PMID: 32976882 DOI: 10.1016/j.lfs.2020.118489] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2020] [Revised: 09/06/2020] [Accepted: 09/10/2020] [Indexed: 12/15/2022]
Abstract
AIMS Cervical cancer (CC) is a common tumor of women worldwide. Here, we conducted a non-targeted lipidomic study to discover novel lipid biomarkers for early-stage CC. MAIN METHODS The lipidomic analysis of 71 samples in discovery set and 72 samples in validation set were performed by coupling ultra-high-pressure liquid chromatography (UHPLC) with quadrupole time-of-flight tandem mass spectrometry (Q-TOF-MS). Lipids with variable importance (VIP) values greater than 1, adj. p < 0.05 (the adjusted p value obtained from false discovery rate correction) and fold change (FC) higher than 1.5 were reserved as potential biomarkers. Subsequently, receiver operating characteristic (ROC) curve and binary logistic regression were implemented to assess the diagnostic potential of these biomarkers and to acquire the best biomarker combination. KEY FINDINGS A lipid biomarker panel, including phosphatidylcholine (PC, PC 14:0/18:2) and phosphatidylethanolamine (PE, PE 15:1e/22:6 and PE 16:1e/18:2), was established. This panel was effective in distinguishing between CC and non-CC (squamous intraepithelial lesions [SIL] and healthy controls) within the area under the ROC curve (AUC), sensitivity, and specificity reaching 0.966, 0.952, and 0.860 for discovery set and 0.961, 0.920, and 0.915 for external validation set. Furthermore, this panel was also capable of discriminating early-stage CC from SIL with AUC, sensitivity, and specificity reaching 0.946, 0.952, and 0.800 for discovery set and 0.956, 0.960, and 0.815 for external validation set. SIGNIFICANCE The combination of PC 14:0/18:2, PE 15:1e/22:6, and PE 16:1e/18:2 could serve as a promising serum biomarker for discriminating early-stage CC from SIL and healthy subjects.
Collapse
Affiliation(s)
- Feng Cheng
- Department of Clinical Laboratory, Women's Hospital of Nanjing Medical University, Nanjing Maternity and Child Health Care Hospital, Nanjing 210000, Jiangsu, China.
| | - Zhifa Wen
- Department of Clinical Laboratory, Women's Hospital of Nanjing Medical University, Nanjing Maternity and Child Health Care Hospital, Nanjing 210000, Jiangsu, China.
| | - Xiaodan Feng
- Department of Clinical Laboratory, Women's Hospital of Nanjing Medical University, Nanjing Maternity and Child Health Care Hospital, Nanjing 210000, Jiangsu, China
| | - Xiaoman Wang
- Department of Clinical Laboratory, Women's Hospital of Nanjing Medical University, Nanjing Maternity and Child Health Care Hospital, Nanjing 210000, Jiangsu, China
| | - Yajun Chen
- Department of Clinical Laboratory, Women's Hospital of Nanjing Medical University, Nanjing Maternity and Child Health Care Hospital, Nanjing 210000, Jiangsu, China.
| |
Collapse
|
10
|
Zheng K, Liu H, Liu X, Jiang L, Li L, Wu X, Guo N, Ding C, Huang M. Photo-triggered release of doxorubicin from liposomes formulated by amphiphilic phthalocyanines for combination therapy to enhance antitumor efficacy. J Mater Chem B 2020; 8:8022-8036. [PMID: 32766661 DOI: 10.1039/d0tb01093f] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
Multidrug combination therapy based on drug delivery systems (DDSs) has great potential for cancer treatment. Stimuli-sensitive DDSs further enhance therapeutic efficacy by providing controllable drug delivery. Herein, the phospholipid compound DPPC (1,2-dipalmitoyl-sn-glycero-3-phosphocholine) was used to construct thermosensitive liposomes to load the photosensitizer ZnPc(PEG)4 (zinc phthalocyanine substituted by tetraethylene glycol) for molecular imaging, and photodynamic and photothermal therapy, together with doxorubicin (DOX) for chemotherapy. Interestingly, ZnPc(PEG)4 as an amphipathic molecule was found to be important in the construction of the liposomes, and it provided liposomes with improved stability. The thus-obtained liposomes ZnPc(PEG)4:DOX@LiPOs were demonstrated to have enhanced ROS production capacity, heat generation properties and a photo-triggered doxorubicin release effect, and, in cellular experiments, increased cytotoxicity and apoptotic cell proportions, compared to ZnPc(PEG)4@LiPOs and DOX@LiPOs. ZnPc(PEG)4 loaded in lipid bilayers showed stronger intracellular ROS production ability compared to free ZnPc(PEG)4. In vivo studies indicated that ZnPc(PEG)4:DOX@LiPOs exhibited enhanced tumor accumulation, increased anti-cancer effects and reduced liver retention. These photo-triggered liposomes constructed by the photosensitizer ZnPc(PEG)4 can also be used to package other cargo for combined target tumor therapy and molecular imaging.
Collapse
Affiliation(s)
- Ke Zheng
- Key Laboratory of Optic-electric Sensing and Analytical Chemistry for Life Science, Ministry of Education, Chemical Engineering College, Qingdao University of Science and Technology, Qingdao 266042, China.
| | | | | | | | | | | | | | | | | |
Collapse
|
11
|
Tjørnelund HD, Madsen JJ, Peters GHJ. Water-Intake and Water-Molecule Paths to the Active Site of Secretory Phospholipase A 2 Studied Using MD Simulations and the Tracking Tool AQUA-DUCT. J Phys Chem B 2020; 124:1881-1891. [PMID: 32064878 DOI: 10.1021/acs.jpcb.9b10837] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
Secretory phospholipases A2 (sPLA2s) are a subclass of enzymes that catalyze the hydrolysis at the sn-2 position of glycerophospholipids, producing free fatty acids and lysophospholipids. In this study, different phospholipids with structural modifications close to the scissile sn-2 ester bond were studied to determine the effect of the structural changes on the formation of the Michaelis-Menten complex and the water entry/exit pathways using molecular dynamics simulations and the computational tracking tool AQUA-DUCT. Structural modifications include methylation, dehydrogenation, and polarization close to the sn-2 scissile bond. We found that all water molecules reaching the active site of sPLA2-IIA pass by the aromatic residues Phe5 and Tyr51 and enter the active site through an active-site cleft. The relative amount of water available for the enzymatic reaction of the different phospholipid-sPLA2 complexes was determined together with the distance between key atoms in the catalytic machinery. The results showed that (Z)-unsaturated phospholipid is a good substrate for sPLA2-IIA. The computational results are in good agreement with previously reported experimental data on the ability of sPLA2-IIA to hydrolyze liposomes made from the different phospholipids, and the results provide new insights into the necessary active-site solvation of the Michaelis-Menten complex and can pave the road for rational design in engineering applications.
Collapse
Affiliation(s)
- Helena D Tjørnelund
- Department of Chemistry, Technical University of Denmark, DK-2800 Kgs. Lyngby, Denmark
| | - Jesper J Madsen
- Department of Global Health, College of Public Health, University of South Florida, Tampa, Florida 33620, United States
| | - Günther H J Peters
- Department of Chemistry, Technical University of Denmark, DK-2800 Kgs. Lyngby, Denmark
| |
Collapse
|
12
|
Tunset HM, Feuerherm AJ, Selvik LKM, Johansen B, Moestue SA. Cytosolic Phospholipase A2 Alpha Regulates TLR Signaling and Migration in Metastatic 4T1 Cells. Int J Mol Sci 2019; 20:ijms20194800. [PMID: 31569627 PMCID: PMC6801560 DOI: 10.3390/ijms20194800] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2019] [Accepted: 09/14/2019] [Indexed: 12/02/2022] Open
Abstract
Metastatic disease is the leading cause of death in breast cancer patients. Disrupting the cancer cell’s ability to migrate may be a strategy for hindering metastasis. Cytosolic phospholipase A2 α (cPLA2α), along with downstream proinflammatory and promigratory metabolites, has been implicated in several aspects of tumorigenesis, as well as metastasis, in various types of cancer. In this study, we aim to characterize the response to reduced cPLA2α activity in metastatic versus non-metastatic cells. We employ an isogenic murine cell line pair displaying metastatic (4T1) and non-metastatic (67NR) phenotype to investigate the role of cPLA2α on migration. Furthermore, we elucidate the effect of reduced cPLA2α activity on global gene expression in the metastatic cell line. Enzyme inhibition is achieved by using a competitive pharmacological inhibitor, cPLA2α inhibitor X (CIX). Our data show that 4T1 expresses significantly higher cPLA2α levels as compared to 67NR, and the two cell lines show different sensitivity to the CIX treatment with regards to metabolism and proliferation. Inhibition of cPLA2α at nontoxic concentrations attenuates migration of highly metastatic 4T1 cells, but not non-metastatic 67NR cells. Gene expression analysis indicates that processes such as interferon type I (IFN-I) signaling and cell cycle regulation are key processes regulated by cPLA2a in metastatic 4T1 cells, supporting the findings from the biological assays. This study demonstrates that two isogenic cancer cell lines with different metastatic potential respond differently to reduced cPLA2α activity. In conclusion, we argue that cPLA2α is a potential therapeutic target in cancer and that enzyme inhibition may inhibit metastasis through an anti-migratory mechanism, possibly involving Toll-like receptor signaling and type I interferons.
Collapse
Affiliation(s)
- Hanna Maja Tunset
- Department of Circulation and Medical Imaging, Faculty of Medicine, Norwegian University of Science and Technology, P.O. Box 8905, 7491 Trondheim, Norway.
| | - Astrid Jullumstrø Feuerherm
- Center for Oral Health Services and Research (TkMidt), 7030 Trondheim, Norway.
- Department of Biology, Norwegian University of Science and Technology, Realfagbygget, 7491 Trondheim, Norway.
| | - Linn-Karina Myrland Selvik
- Department of Biology, Norwegian University of Science and Technology, Realfagbygget, 7491 Trondheim, Norway.
| | - Berit Johansen
- Department of Biology, Norwegian University of Science and Technology, Realfagbygget, 7491 Trondheim, Norway.
| | - Siver Andreas Moestue
- Department of Circulation and Medical Imaging, Faculty of Medicine, Norwegian University of Science and Technology, P.O. Box 8905, 7491 Trondheim, Norway.
- Department of Health Sciences, Nord University, P.O. Box 1490, 8049 Bodø, Norway.
| |
Collapse
|
13
|
Markovic M, Ben-Shabat S, Keinan S, Aponick A, Zimmermann EM, Dahan A. Molecular Modeling-Guided Design of Phospholipid-Based Prodrugs. Int J Mol Sci 2019; 20:ijms20092210. [PMID: 31060339 PMCID: PMC6538990 DOI: 10.3390/ijms20092210] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2019] [Revised: 04/29/2019] [Accepted: 05/01/2019] [Indexed: 02/06/2023] Open
Abstract
The lipidic prodrug approach is an emerging field for improving a number of biopharmaceutical and drug delivery aspects. Owing to their structure and nature, phospholipid (PL)-based prodrugs may join endogenous lipid processing pathways, and hence significantly improve the pharmacokinetics and/or bioavailability of the drug. Additional advantages of this approach include drug targeting by enzyme-triggered drug release, blood–brain barrier permeability, lymphatic targeting, overcoming drug resistance, or enabling appropriate formulation. The PL-prodrug design includes various structural modalities-different conjugation strategies and/or the use of linkers between the PL and the drug moiety, which considerably influence the prodrug characteristics and the consequent effects. In this article, we describe how molecular modeling can guide the structural design of PL-based prodrugs. Computational simulations can predict the extent of phospholipase A2 (PLA2)-mediated activation, and facilitate prodrug development. Several computational methods have been used to facilitate the design of the pro-drugs, which will be reviewed here, including molecular docking, the free energy perturbation method, molecular dynamics simulations, and free density functional theory. Altogether, the studies described in this article indicate that computational simulation-guided PL-based prodrug molecular design correlates well with the experimental results, allowing for more mechanistic and less empirical development. In the future, the use of molecular modeling techniques to predict the activity of PL-prodrugs should be used earlier in the development process.
Collapse
Affiliation(s)
- Milica Markovic
- Department of Clinical Pharmacology, School of Pharmacy, Faculty of Health Sciences, Ben-Gurion University of the Negev, Beer-Sheva 8410501, Israel.
| | - Shimon Ben-Shabat
- Department of Clinical Pharmacology, School of Pharmacy, Faculty of Health Sciences, Ben-Gurion University of the Negev, Beer-Sheva 8410501, Israel.
| | | | - Aaron Aponick
- Department of Chemistry, University of Florida, Gainesville, FL 32603, USA.
| | - Ellen M Zimmermann
- Department of Medicine, Division of Gastroenterology, University of Florida, Gainesville, FL 32608, USA.
| | - Arik Dahan
- Department of Clinical Pharmacology, School of Pharmacy, Faculty of Health Sciences, Ben-Gurion University of the Negev, Beer-Sheva 8410501, Israel.
| |
Collapse
|
14
|
Wu C, Su J, Wang X, Wang J, Xiao K, Li Y, Xiao Q, Ling M, Xiao Y, Qin C, Long W, Zhang F, Pan Y, Xiang F, Liu Q. Overexpression of the phospholipase A2 group V gene in glioma tumors is associated with poor patient prognosis. Cancer Manag Res 2019; 11:3139-3152. [PMID: 31114356 PMCID: PMC6489671 DOI: 10.2147/cmar.s199207] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2018] [Accepted: 03/17/2019] [Indexed: 11/23/2022] Open
Abstract
Purpose: Gliomas are the most common primary malignant neoplasms of the central nervous system. Secreted phospholipases A2 (sPLA2s) are known to play an important role in various physiological processes, including bioactive lipid production, defense mechanisms, and cell signaling. However, their roles and clinical importance in gliomas remain unclear. Patients and methods: In this study, we analyzed the association between the expression of various sPLA2-encoding genes and the clinicopathology of gliomas, using the data of 1047 patients obtained from a public database. Immunohistochemical analysis of 82 glioma tissues was also carried out to assess the relationship between phospholipase A2 group V (PLA2G5) protein expression and the World Health Organization (WHO) glioma grades. Results: We found that high PLA2G5 gene expression was associated with unfavorable prognosis in both low-grade and high-grade gliomas. The immunohistochemistry of the 82 glioma tissues further confirmed that PLA2G5 protein expression was dependent on the WHO glioma grade. In addition, we found a correlation between PLA2G5 gene expression and both epithelial-mesenchymal transition and the isocitrate dehydrogenase 1 mutation status in these tumors. Conclusion: Our results indicate that PLA2G5 could be a potential biomarker for predicting poor prognosis in patients with gliomas.
Collapse
Affiliation(s)
- Changwu Wu
- Department of Neurosurgery, Xiangya Hospital, Central-South University, Changsha, Hunan, People's Republic of China
| | - Jun Su
- Department of Neurosurgery, Xiangya Hospital, Central-South University, Changsha, Hunan, People's Republic of China
| | - Xiangyu Wang
- Department of Neurosurgery, Xiangya Hospital, Central-South University, Changsha, Hunan, People's Republic of China
| | - Junquan Wang
- Department of Neurosurgery, Xiangya Hospital, Central-South University, Changsha, Hunan, People's Republic of China
| | - Kai Xiao
- Department of Neurosurgery, Xiangya Hospital, Central-South University, Changsha, Hunan, People's Republic of China
| | - Yang Li
- Department of Neurosurgery, Xiangya Hospital, Central-South University, Changsha, Hunan, People's Republic of China
| | - Qun Xiao
- Department of Neurosurgery, Xiangya Hospital, Central-South University, Changsha, Hunan, People's Republic of China
| | - Min Ling
- Department of Neurosurgery, Xiangya Hospital, Central-South University, Changsha, Hunan, People's Republic of China
| | - Yao Xiao
- Department of Neurosurgery, Xiangya Hospital, Central-South University, Changsha, Hunan, People's Republic of China
| | - Chaoying Qin
- Department of Neurosurgery, Xiangya Hospital, Central-South University, Changsha, Hunan, People's Republic of China
| | - Wenyong Long
- Department of Neurosurgery, Xiangya Hospital, Central-South University, Changsha, Hunan, People's Republic of China.,Institute of Skull Base Surgery and Neuro-Oncology at Hunan Neurosurgery Institute of Central South University, Changsha, Hunan, People's Republic of China
| | - Fengqi Zhang
- Department of Neurosurgery, Xiangya Hospital, Central-South University, Changsha, Hunan, People's Republic of China
| | - Yimin Pan
- Department of Neurosurgery, Xiangya Hospital, Central-South University, Changsha, Hunan, People's Republic of China
| | - Feng Xiang
- Department of Neurosurgery, Xiangya Hospital, Central-South University, Changsha, Hunan, People's Republic of China
| | - Qing Liu
- Department of Neurosurgery, Xiangya Hospital, Central-South University, Changsha, Hunan, People's Republic of China.,Institute of Skull Base Surgery and Neuro-Oncology at Hunan Neurosurgery Institute of Central South University, Changsha, Hunan, People's Republic of China
| |
Collapse
|
15
|
Li Y, Du L, Wu C, Yu B, Zhang H, An F. Peptide Sequence-Dominated Enzyme-Responsive Nanoplatform for Anticancer Drug Delivery. Curr Top Med Chem 2019; 19:74-97. [PMID: 30686257 DOI: 10.2174/1568026619666190125144621] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2018] [Revised: 11/06/2018] [Accepted: 11/23/2018] [Indexed: 02/08/2023]
Abstract
Enzymatic dysregulation in tumor and intracellular microenvironments has made this property
a tremendously promising responsive element for efficient diagnostics, carrier targeting, and drug
release. When combined with nanotechnology, enzyme-responsive drug delivery systems (DDSs) have
achieved substantial advancements. In the first part of this tutorial review, changes in tumor and intracellular
microenvironmental factors, particularly the enzymatic index, are described. Subsequently, the
peptide sequences of various enzyme-triggered nanomaterials are summarized for their uses in various
drug delivery applications. Then, some other enzyme responsive nanostructures are discussed. Finally,
the future opportunities and challenges are discussed. In brief, this review can provide inspiration and
impetus for exploiting more promising internal enzyme stimuli-responsive nanoDDSs for targeted tumor
diagnosis and treatment.
Collapse
Affiliation(s)
- Yanan Li
- First Clinical Medical College, Shanxi Medical University, Taiyuan, Shanxi, 030001, China
| | - Liping Du
- Institute of Medical Engineering, Department of Biophysics, School of Basic Medical Science, Health Science Center, Xi’an Jiaotong University, No.76 Yanta West Road, Xi'an, Shaanxi 710061, China
| | - Chunsheng Wu
- Institute of Medical Engineering, Department of Biophysics, School of Basic Medical Science, Health Science Center, Xi’an Jiaotong University, No.76 Yanta West Road, Xi'an, Shaanxi 710061, China
| | - Bin Yu
- School of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou, 450001, China
| | - Hui Zhang
- First Clinical Medical College, Shanxi Medical University, Taiyuan, Shanxi, 030001, China
| | - Feifei An
- Institute of Medical Engineering, Department of Biophysics, School of Basic Medical Science, Health Science Center, Xi’an Jiaotong University, No.76 Yanta West Road, Xi'an, Shaanxi 710061, China
| |
Collapse
|
16
|
Markovic M, Ben-Shabat S, Keinan S, Aponick A, Zimmermann EM, Dahan A. Prospects and Challenges of Phospholipid-Based Prodrugs. Pharmaceutics 2018; 10:pharmaceutics10040210. [PMID: 30388756 PMCID: PMC6321354 DOI: 10.3390/pharmaceutics10040210] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2018] [Revised: 10/24/2018] [Accepted: 10/26/2018] [Indexed: 12/19/2022] Open
Abstract
Nowadays, the prodrug approach is used already at the early stages of drug development. Lipidic prodrug approach is a growing field for improving a number of drug properties/delivery/therapy aspects, and can offer solutions for various unmet needs. This approach includes drug moiety bound to the lipid carrier, which can be triglyceride, fatty acids, steroid, or phospholipid (PL). The focus of this article is PL-based prodrugs, which includes a PL carrier covalently bound to the active drug moiety. An overview of relevant physiological lipid processing pathways and absorption barriers is provided, followed by drug delivery/therapeutic application of PL-drug conjugates, as well as computational modeling techniques, and a modern bioinformatics tool that can aid in the optimization of PL conjugates. PL-based prodrugs have increased lipophilicity comparing to the parent drug, and can therefore significantly improve the pharmacokinetic profile and overall bioavailability of the parent drug, join the endogenous lipid processing pathways and therefore accomplish drug targeting, e.g., by lymphatic transport, drug release at specific target site(s), or passing the blood-brain barrier. Moreover, an exciting gateway for treating inflammatory diseases and cancer is presented, by utilizing the PL sn-2 position in the prodrug design, aiming for PLA₂-mediated activation. Overall, a PL-based prodrug approach shows great potential in improving different drug delivery/therapy aspects, and is expected to grow.
Collapse
Affiliation(s)
- Milica Markovic
- Department of Clinical Pharmacology, School of Pharmacy, Faculty of Health Sciences, Ben-Gurion University of the Negev, Beer-Sheva 8410501, Israel.
| | - Shimon Ben-Shabat
- Department of Clinical Pharmacology, School of Pharmacy, Faculty of Health Sciences, Ben-Gurion University of the Negev, Beer-Sheva 8410501, Israel.
| | | | - Aaron Aponick
- Department of Chemistry, University of Florida, Gainesville, FL 32603, USA.
| | - Ellen M Zimmermann
- Department of Medicine, Division of Gastroenterology, University of Florida, Gainesville, FL 32610, USA.
| | - Arik Dahan
- Department of Clinical Pharmacology, School of Pharmacy, Faculty of Health Sciences, Ben-Gurion University of the Negev, Beer-Sheva 8410501, Israel.
| |
Collapse
|
17
|
Bayci AWL, Baker DA, Somerset AE, Turkoglu O, Hothem Z, Callahan RE, Mandal R, Han B, Bjorndahl T, Wishart D, Bahado-Singh R, Graham SF, Keidan R. Metabolomic identification of diagnostic serum-based biomarkers for advanced stage melanoma. Metabolomics 2018; 14:105. [PMID: 30830422 DOI: 10.1007/s11306-018-1398-9] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/20/2018] [Accepted: 07/18/2018] [Indexed: 01/02/2023]
Abstract
INTRODUCTION Melanoma is a highly aggressive malignancy and is currently one of the fastest growing cancers worldwide. While early stage (I and II) disease is highly curable with excellent prognosis, mortality rates rise dramatically after distant spread. We sought to identify differences in the metabolome of melanoma patients to further elucidate the pathophysiology of melanoma and identify potential biomarkers to aid in earlier detection of recurrence. METHODS Using 1H NMR and DI-LC-MS/MS, we profiled serum samples from 26 patients with stage III (nodal metastasis) or stage IV (distant metastasis) melanoma and compared their biochemical profiles with 46 age- and gender-matched controls. RESULTS We accurately quantified 181 metabolites in serum using a combination of 1H NMR and DI-LC-MS/MS. We observed significant separation between cases and controls in the PLS-DA scores plot (permutation test p-value = 0.002). Using the concentrations of PC-aa-C40:3, DL-carnitine, octanoyl-L-carnitine, ethanol, and methylmalonyl-L-carnitine we developed a diagnostic algorithm with an AUC (95% CI) = 0.822 (0.665-0.979) with sensitivity and specificity of 100 and 56%, respectively. Furthermore, we identified arginine, proline, tryptophan, glutamine, glutamate, glutathione and ornithine metabolism to be significantly perturbed due to disease (p < 0.05). CONCLUSION Targeted metabolomic analysis demonstrated significant differences in metabolic profiles of advanced stage (III and IV) melanoma patients as compared to controls. These differences may represent a potential avenue for the development of multi-marker serum-based assays for earlier detection of recurrences, allow for newer, more effective targeted therapy when tumor burden is less, and further elucidate the pathophysiologic changes that occur in melanoma.
Collapse
Affiliation(s)
- A W L Bayci
- Department of General Surgery, Beaumont Health, Royal Oak, MI, USA
| | - D A Baker
- Department of General Surgery, Beaumont Health, Royal Oak, MI, USA.
- Department of Surgery, Beaumont Health, 3601 W. 13 Mile Rd., Royal Oak, MI, 48073, USA.
| | - A E Somerset
- Department of General Surgery, Beaumont Health, Royal Oak, MI, USA
| | - O Turkoglu
- Department of Obstetrics and Gynecology, Beaumont Health, Royal Oak, MI, USA
| | - Z Hothem
- Department of General Surgery, Beaumont Health, Royal Oak, MI, USA
| | - R E Callahan
- Department of General Surgery, Beaumont Health, Royal Oak, MI, USA
| | - R Mandal
- Department of Biological and Computing Sciences, University of Alberta Edmonton, Edmonton, AB, Canada
| | - B Han
- Department of Biological and Computing Sciences, University of Alberta Edmonton, Edmonton, AB, Canada
| | - T Bjorndahl
- Department of Biological and Computing Sciences, University of Alberta Edmonton, Edmonton, AB, Canada
| | - D Wishart
- Department of Biological and Computing Sciences, University of Alberta Edmonton, Edmonton, AB, Canada
| | - R Bahado-Singh
- Department of Obstetrics and Gynecology, Beaumont Health, Royal Oak, MI, USA
| | - S F Graham
- Department of Obstetrics and Gynecology, Beaumont Health, Royal Oak, MI, USA
| | - R Keidan
- Department of General Surgery, Beaumont Health, Royal Oak, MI, USA
| |
Collapse
|
18
|
Zhang D, Li J, Sun S, Huang C. The inhibitory effect of saPLIγ, a snake sourced PLA 2 inhibitor on carrageenan-induced inflammation in mice. Toxicon 2018; 151:89-95. [PMID: 30003915 DOI: 10.1016/j.toxicon.2018.07.002] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2018] [Revised: 06/07/2018] [Accepted: 07/05/2018] [Indexed: 12/23/2022]
Abstract
SaPLIγ is a natural phospholipase A2 (PLA2) inhibitor, isolated from Sinonatrix annularis, that has been demonstrated to protect against envenomation by other venomous snakes. As snake venom PLA2s and mammalian secretory PLA2s are similar, saPLIγ is thought to have potential to alleviate inflammatory reactions in which PLA2s act as a key enzyme for arachidonic acid release. The aim of this study was to investigate the anti-inflammatory effects and mechanisms of action of saPLIγ in an animal model of carrageenan-induced acute inflammation. The results indicated that saPLIγ inhibited PLA2 subtypes extensively, especially IIA-PLA2, in a dose-dependent manner. Paw swelling in mice was reduced markedly by intraperitoneal saPLIγ 2.5 mg/kg, and the effect was significantly better than observed with dexamethasone at the same dose. Lower neutrophil infiltration and tissue edema was observed in the paws of saPLIγ-treated mice. Additionally, carrageenan-induced cyclooxygenase-2 (COX-2) and pro-inflammatory cytokines (TNFα and IL-1β) were also significantly down-regulated by saPLIγ in a dose-dependent manner. These results suggested that saPLIγ had effective anti-inflammatory effects in vivo, and these were produced by blocking mammalian IB, IIA, V and X sPLA2 subtypes.
Collapse
Affiliation(s)
- Denghong Zhang
- Department of Biochemistry, College of Basic Medical Science, Nanchang University, Jiangxi province, China
| | - Jingjing Li
- Department of Biochemistry, College of Basic Medical Science, Nanchang University, Jiangxi province, China
| | - Shimin Sun
- Department of Biochemistry, College of Basic Medical Science, Nanchang University, Jiangxi province, China
| | - Chunhong Huang
- Department of Biochemistry, College of Basic Medical Science, Nanchang University, Jiangxi province, China.
| |
Collapse
|
19
|
Chen X, Chen H, Dai M, Ai J, Li Y, Mahon B, Dai S, Deng Y. Plasma lipidomics profiling identified lipid biomarkers in distinguishing early-stage breast cancer from benign lesions. Oncotarget 2017; 7:36622-36631. [PMID: 27153558 PMCID: PMC5095026 DOI: 10.18632/oncotarget.9124] [Citation(s) in RCA: 84] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2016] [Accepted: 04/16/2016] [Indexed: 12/29/2022] Open
Abstract
BACKGROUND Breast cancer is very common and highly fatal in women. Current non-invasive detection methods like mammograms are unsatisfactory. Lipidomics, a promising detection method, may serve as a novel prognostic approach for breast cancer in high-risk patients. RESULTS According the predictive model, the combination of 15 lipid species had high diagnostic value. In the training set, sensitivity, specificity, positive predictive value (PPV) and negative predictive value (NPV) of the combination of these 15 lipid species were 83.3%, 92.7%, 89.7%, and 87.9%, respectively. The AUC in the training set was 0.926 (95% CI 0.869-0.982). Similar results were found in the validation set, with the sensitivity, specificity, PPV and NPV at 81.0%, 94.5%, 91.9%, and 86.7%, respectively. The AUC was 0.938 (95% CI 0.889-0.986) in the validation set. METHODS Using triple quadrupole liquid chromatography electrospray ionization tandem mass spectrometry, this study was to detect global lipid profiling of a total of 194 plasma samples from 84 patients with early-stage breast cancer (stage 0-II) and 110 patients with benign breast disease included in a training set and a validation set. A binary logistic regression was used to build a predictive model for evaluating the lipid species as potential biomarkers in the diagnosis of breast cancer. CONCLUSIONS The combination of these 15 lipid species as a panel could be used as plasma biomarkers for the diagnosis of breast cancer.
Collapse
Affiliation(s)
- Xiaoli Chen
- Department of Clinical Laboratory, The Fourth Hospital Affiliated to Guangxi Medical University, Liuzhou City, Guangxi Province, China.,Department of Medicine, Rush University Medical Center, Chicago, Illinois, USA
| | - Hankui Chen
- Department of Medicine, Rush University Medical Center, Chicago, Illinois, USA
| | - Meiyu Dai
- Department of Clinical Laboratory, The Fourth Hospital Affiliated to Guangxi Medical University, Liuzhou City, Guangxi Province, China.,Department of Medicine, Rush University Medical Center, Chicago, Illinois, USA
| | - Junmei Ai
- Department of Medicine, Rush University Medical Center, Chicago, Illinois, USA
| | - Yan Li
- Department of Medicine, Rush University Medical Center, Chicago, Illinois, USA
| | - Brett Mahon
- Department of Pathology, Rush University Medical Center, Chicago, Illinois, USA
| | - Shengming Dai
- Department of Clinical Laboratory, The Fourth Hospital Affiliated to Guangxi Medical University, Liuzhou City, Guangxi Province, China
| | - Youping Deng
- Department of Medicine, Rush University Medical Center, Chicago, Illinois, USA.,Department of Anatomy and Cell Biology, Rush University Medical Center, Chicago, Illinois, USA.,Medical College, Wuhan University of Science and Technology, Wuhan, Hubei Province, China
| |
Collapse
|
20
|
Phospholipases during membrane dynamics in malaria parasites. Int J Med Microbiol 2017; 308:129-141. [PMID: 28988696 DOI: 10.1016/j.ijmm.2017.09.015] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2017] [Revised: 09/15/2017] [Accepted: 09/19/2017] [Indexed: 12/26/2022] Open
Abstract
Plasmodium parasites, the causative agents of malaria, display a well-regulated lipid metabolism required to ensure their survival in the human host as well as in the mosquito vector. The fine-tuning of lipid metabolic pathways is particularly important for the parasites during the rapid erythrocytic infection cycles, and thus enzymes involved in lipid metabolic processes represent prime targets for malaria chemotherapeutics. While plasmodial enzymes involved in lipid synthesis and acquisition have been studied in the past, to date not much is known about the roles of phospholipases for proliferation and transmission of the malaria parasite. These phospholipid-hydrolyzing esterases are crucial for membrane dynamics during host cell infection and egress by the parasite as well as for replication and cell signaling, and thus they are considered important virulence factors. In this review, we provide a comprehensive bioinformatic analysis of plasmodial phospholipases identified to date. We further summarize previous findings on the lipid metabolism of Plasmodium, highlight the roles of phospholipases during parasite life-cycle progression, and discuss the plasmodial phospholipases as potential targets for malaria therapy.
Collapse
|
21
|
Jiang N, Zhang G, Pan L, Yan C, Zhang L, Weng Y, Wang W, Chen X, Yang G. Potential plasma lipid biomarkers in early-stage breast cancer. Biotechnol Lett 2017; 39:1657-1666. [PMID: 28828718 DOI: 10.1007/s10529-017-2417-z] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2017] [Accepted: 08/20/2017] [Indexed: 02/07/2023]
Abstract
OBJECTIVE To find new biomarkers for early diagnosis of breast cancer. RESULTS 847 lipid species were identified from 78 plasma samples (37 breast cancer samples and 41 healthy controls) by ultra HPLC coupled with quadrupole time-of-flight tandem mass spectrometry. These include 321 glycerophospholipids (GPs), 265 glycerolipids (GLs), 91 sphingolipids (SPs), 77 fatty acyls (FAs), 68 sterol lipids (STs), 18 prenol lipids (PRs), 6 polyketides (PKs), and 1 saccharolipid (SL). Separation was observed from an orthogonal signal correction Partial Least Square Discrimination Analysis model. Based on this analysis, six differentiating lipids were identified: PC (20:2/20:5), PC (22:0/24:1), TG (12:0/14:1), and DG (18:1/18:2) had high levels, whereas PE (15:0/19:1) and N-palmitoyl proline had low levels in the breast cancer samples compared with the healthy controls. Furthermore, significant differences in metabolites were found among some clinical characteristics. CONCLUSIONS Our results reveal that six specific lipids could serve as potential biomarkers for early diagnosis of breast cancer.
Collapse
Affiliation(s)
- Nan Jiang
- Department of General Surgery, First Hospital of Tsinghua University, Beijing, 100016, China
| | - Guofen Zhang
- Department of General Surgery, First Hospital of Tsinghua University, Beijing, 100016, China
| | - Lijie Pan
- Department of General Surgery, First Hospital of Tsinghua University, Beijing, 100016, China
| | - Chengping Yan
- Department of General Surgery, First Hospital of Tsinghua University, Beijing, 100016, China
| | - Liwei Zhang
- Department of General Surgery, First Hospital of Tsinghua University, Beijing, 100016, China
| | - Yan Weng
- Department of Pathology, First Hospital of Tsinghua University, Beijing, 100016, China
| | - Wenjun Wang
- Beijing Qiji Biotechnology Company, Beijing, 100193, China
| | - Xianyang Chen
- Beijing Qiji Biotechnology Company, Beijing, 100193, China
| | - Guoshan Yang
- Department of General Surgery, First Hospital of Tsinghua University, Beijing, 100016, China.
| |
Collapse
|
22
|
Loffredo S, Borriello F, Iannone R, Ferrara AL, Galdiero MR, Gigantino V, Esposito P, Varricchi G, Lambeau G, Cassatella MA, Granata F, Marone G. Group V Secreted Phospholipase A 2 Induces the Release of Proangiogenic and Antiangiogenic Factors by Human Neutrophils. Front Immunol 2017; 8:443. [PMID: 28458672 PMCID: PMC5394767 DOI: 10.3389/fimmu.2017.00443] [Citation(s) in RCA: 59] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2017] [Accepted: 03/30/2017] [Indexed: 02/05/2023] Open
Abstract
Secreted phospholipases A2 (sPLA2s) are extracellular enzymes that catalyze the release of free fatty acids and lysophospholipids from membrane phospholipids and also bind to different receptors (e.g., PLA2R1 or integrins). To date, 12 mammalian sPLA2s have been identified, which play a critical role in pathophysiological processes including inflammation and cancer. sPLA2s activate immune cells such as human neutrophils (PMNs) by enzymatic activity- or receptor-mediated mechanisms. In addition, human PMNs synthesize and store human group V (hGV) and human group X (hGX) sPLA2s in their granules, but only the former is released upon cellular activation. We investigated the effects of sPLA2s on the release of proangiogenic and antiangiogenic factors by PMNs. We found that exogenous hGV and hGX sPLA2s induce the release of vascular endothelial growth factor (VEGF)-A, angiopoietin 1 (Ang1), and CXCL8/IL-8. Only hGV induces the secretion of the antiangiogenic isoform of VEGF-A, namely, VEGF-A165b. While the release of VEGF-A, Ang1, and CXCL8/IL-8 was likely mediated by hGV enzymatic activity and/or binding to PLA2R1 and heparan sulfate proteoglycans, the release of VEGF-A165b requires the interaction with αVβ3 and α4β1 integrins. We also provide evidence that endogenous hGV released by N-formyl-met-leu-phe (fMLF)-activated PMNs is involved in the release of angiogenic factors. The translational relevance of these data is supported by our findings that hGV expression is increased in human samples of lung cancer which are infiltrated by PMNs. Overall, our results suggest that the hGV-neutrophil axis may play a relevant role in the modulation of cancer-related inflammation and angiogenesis.
Collapse
Affiliation(s)
- Stefania Loffredo
- Division of Clinical Immunology and Allergy, Center for Basic and Clinical Immunology Research (CISI), University of Naples Federico II, Naples, Italy
| | - Francesco Borriello
- Division of Clinical Immunology and Allergy, Center for Basic and Clinical Immunology Research (CISI), University of Naples Federico II, Naples, Italy
- Division of Infectious Diseases, Department of Medicine, Boston Children’s Hospital, Boston, MA, USA
| | - Raffaella Iannone
- Division of Clinical Immunology and Allergy, Center for Basic and Clinical Immunology Research (CISI), University of Naples Federico II, Naples, Italy
| | - Anne L. Ferrara
- Division of Clinical Immunology and Allergy, Center for Basic and Clinical Immunology Research (CISI), University of Naples Federico II, Naples, Italy
| | - Maria R. Galdiero
- Division of Clinical Immunology and Allergy, Center for Basic and Clinical Immunology Research (CISI), University of Naples Federico II, Naples, Italy
| | - Vincenzo Gigantino
- Pathology Unit, Istituto Nazionale Tumori Fondazione “G. Pascale”, Naples, Italy
| | - Pasquale Esposito
- U.O.C. Immunohematology and Transfusion Medicine, University of Naples Federico II, Naples, Italy
| | - Gilda Varricchi
- Division of Clinical Immunology and Allergy, Center for Basic and Clinical Immunology Research (CISI), University of Naples Federico II, Naples, Italy
| | - Gerard Lambeau
- CNRS, Institut de Pharmacologie Moléculaire et Cellulaire, Université Côte d’Azur, Valbonne Sophia Antipolis, France
| | - Marco A. Cassatella
- Department of Medicine, Division of General Pathology, University of Verona, Verona, Italy
| | - Francescopaolo Granata
- Division of Clinical Immunology and Allergy, Center for Basic and Clinical Immunology Research (CISI), University of Naples Federico II, Naples, Italy
| | - Gianni Marone
- Division of Clinical Immunology and Allergy, Center for Basic and Clinical Immunology Research (CISI), University of Naples Federico II, Naples, Italy
- CNR Institute of Experimental Endocrinology and Oncology “G. Salvatore”, Naples, Italy
| |
Collapse
|
23
|
Madsen JJ, Fristrup P, Peters GH. Theoretical Assessment of Fluorinated Phospholipids in the Design of Liposomal Drug-Delivery Systems. J Phys Chem B 2016; 120:9661-71. [DOI: 10.1021/acs.jpcb.6b07206] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Affiliation(s)
- Jesper J. Madsen
- Department of Chemistry, Technical University of Denmark, DK-2800 Kgs. Lyngby, Denmark
| | - Peter Fristrup
- Department of Chemistry, Technical University of Denmark, DK-2800 Kgs. Lyngby, Denmark
| | - Günther H. Peters
- Department of Chemistry, Technical University of Denmark, DK-2800 Kgs. Lyngby, Denmark
| |
Collapse
|
24
|
Biological activity and binding properties of [Ru(II)(dcbpy)2Cl2] complex to bovine serum albumin, phospholipase A2 and glutathione. Biometals 2016; 29:921-33. [PMID: 27515969 DOI: 10.1007/s10534-016-9964-y] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2016] [Accepted: 07/31/2016] [Indexed: 02/02/2023]
Abstract
Ruthenium compounds are highly regarded as metallo-drug candidates. Many studies have focused their attention on the interaction between ruthenium complexes with their possible biological targets. The interaction of ruthenium complexes with transport proteins, enzymes and peptides is of great importance for understanding their biodistribution and mechanism of action, therefore, the development of an anti-cancer therapy involving ruthenium complexes has recently shifted from DNA targeting towards protein targeting. With the aim of gaining insight into possible interactions between ruthenium complexes with biologically relevant proteins, we have studied the interaction of cis-dichlorobis(2,2'-bipyridyl-4,4'-dicarboxylic acid)ruthenium(II) complex [Ru(II)(dcbpy)2Cl2], which previously showed good potency in photo-dynamic chemotherapy, with bovine serum albumin (BSA), phospholipase A2 (PLA2) and glutathione (GSH). Binding constants and possible number of binding sites to mentioned proteins and peptide are investigated by ultraviolet-visible spectroscopy and Matrix-Assisted Laser Desorption Ionization Mass Spectrometry (MALDI TOF MS). The complex binding affinities were in the following order: PLA2 > BSA > GSH. Moreover, genotoxic profile of the complex, tested on peripheral blood lymphocytes as a model system, was also promising.
Collapse
|
25
|
Szcześ A. Effect of the enzymatically modified supported dipalmitoylphosphatidylcholine (DPPC) bilayers on calcium carbonate formation. Colloid Polym Sci 2016; 294:409-419. [PMID: 26855469 PMCID: PMC4733140 DOI: 10.1007/s00396-015-3796-0] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2015] [Revised: 09/21/2015] [Accepted: 10/20/2015] [Indexed: 11/25/2022]
Abstract
After an hour contact with a phospholipase A2 (PLA2) solution, only the outer leaflet of the dipalmitoylphosphatidylcholine (DPPC) bilayers supported on mica surface underwent hydrolysis whose products, i.e., palmitic acid and lysophospholipid, accumulated on the bilayer surface. Only calcite was present on the bare mica and enzymatically unmodified and modified supported DPPC bilayers soaked for 2 weeks at 25 and 37 °C in a solution of initial pH equals to 7.4 and 9.2 containing calcium and bicarbonate ions at their concentrations about those of human blood plasma. The DPPC bilayers accelerate the crystal growth at lower pH and favors CaCO3 nucleation at higher pH. Enzymatic modification of bilayers does not affect crystal morphology and its organization on the examined surface but causes a slight crystal size increase at lower pH and significantly reduces crystal size at alkaline pH. The temperature increase leads to the formation of bigger crystals under physiological pH and has almost no effect on crystal size at alkaline pH. The obtained results are probably attributed to Ca2+ interaction with a specific polar site on the surface of the membrane and DPPC hydrolysis products acting as nucleation centers.
Collapse
Affiliation(s)
- Aleksandra Szcześ
- Department of Physical Chemistry-Interfacial Phenomena, Faculty of Chemistry, Maria Curie-Skłodowska University, Lublin, 20-031 Poland
| |
Collapse
|
26
|
Li J, Zhang Y, Ai J, Gao Q, Qi H, Zhang C, Cheng Z. Quantum dot cluster (QDC)-loaded phospholipid micelles as a FRET probe for phospholipase A 2 detection. RSC Adv 2016; 6:15895-15899. [PMID: 26998254 PMCID: PMC4792299 DOI: 10.1039/c5ra25292j] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/22/2022] Open
Abstract
A simple assay for phospholipase A2 (PLA2) enzyme was developed based on a fluorescence resonance energy transfer (FRET) probe using the quantum dot cluster (QDC)-loaded phospholipid micelles. The probe was prepared by encapsulating many small hydrophobic quantum dots (QDs) within the hydrophobic core of micelles that were formed from the coassembly of hydrogenated soy phosphatidylcholine phospholipids (HSPC) and fluorescent lipids (NBD-PC). QDCs formed within the micelle core served as the substrate for NBD fluorescence quenching through FRET. The QDC-loaded micelles showed very low background fluorescence. As the PLA2 enzyme selectively digested lipids, the NBD fluorescence was recovered from its quenched state, leading to the sensitive detection of PLA2. This assay provided a limit of detection (at a signal-to-noise ratio of 3) of 3 U/L for PLA2. In the presence of a PLA2 inhibitor, the fluorescent response of the sensor for PLA2 decreased, indicating that the assay could also be used for screening the PLA2 inhibitors.
Collapse
Affiliation(s)
- Junling Li
- Key Laboratory of Applied Surface and Colloid Chemistry, Ministry of Education, School of Chemistry and Chemical Engineering, Shaanxi Normal University, Xi'an, 710062, China
| | - Yonghua Zhang
- Key Laboratory of Applied Surface and Colloid Chemistry, Ministry of Education, School of Chemistry and Chemical Engineering, Shaanxi Normal University, Xi'an, 710062, China
| | - Junjie Ai
- Key Laboratory of Applied Surface and Colloid Chemistry, Ministry of Education, School of Chemistry and Chemical Engineering, Shaanxi Normal University, Xi'an, 710062, China
| | - Qiang Gao
- Key Laboratory of Applied Surface and Colloid Chemistry, Ministry of Education, School of Chemistry and Chemical Engineering, Shaanxi Normal University, Xi'an, 710062, China
| | - Honglan Qi
- Key Laboratory of Applied Surface and Colloid Chemistry, Ministry of Education, School of Chemistry and Chemical Engineering, Shaanxi Normal University, Xi'an, 710062, China
| | - Chengxiao Zhang
- Key Laboratory of Applied Surface and Colloid Chemistry, Ministry of Education, School of Chemistry and Chemical Engineering, Shaanxi Normal University, Xi'an, 710062, China
| | - Zhiliang Cheng
- Department of Bioengineering, University of Pennsylvania, Philadelphia, PA 19104, USA
| |
Collapse
|
27
|
Loza-Mejía MA, Salazar JR. Sterols and triterpenoids as potential anti-inflammatories: Molecular docking studies for binding to some enzymes involved in inflammatory pathways. J Mol Graph Model 2015; 62:18-25. [PMID: 26342572 DOI: 10.1016/j.jmgm.2015.08.010] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2015] [Revised: 08/24/2015] [Accepted: 08/26/2015] [Indexed: 12/12/2022]
Abstract
Triterpenes and sterols are good candidates for the development of anti-inflammatory drugs and use in chemoprevention or chemotherapy of cancer via the interaction with therapeutic targets related to inflammation, such as COX-1 and -2; LOX-5; MPO, PLA2 and i-NOS. In this study, we use molecular docking to evaluate the potential binding of a database of selected sterol and triterpenoid compounds with several skeletons against enzymes related to inflammation to propose structural requirements beneficial for anti-inflammatory activity that can be used for the design of more potent and selective anti-inflammatory and antitumor drugs. Our results suggest that the substitution pattern is important and that there is an important relationship between the class of sterol or triterpenoid skeleton and enzyme binding.
Collapse
Affiliation(s)
- Marco A Loza-Mejía
- Facultad de Ciencias Químicas, Universidad La Salle, Benjamín Franklin 47, 06140 México City, Mexico.
| | - Juan Rodrigo Salazar
- Facultad de Ciencias Químicas, Universidad La Salle, Benjamín Franklin 47, 06140 México City, Mexico.
| |
Collapse
|
28
|
Ram A, Mabalirajan U, Jaiswal A, Rehman R, Singh VP, Ghosh B. Parabromophenacyl bromide inhibits subepithelial fibrosis by reducing TGF-β1 in a chronic mouse model of allergic asthma. Int Arch Allergy Immunol 2015; 167:110-8. [PMID: 26303861 DOI: 10.1159/000434679] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2014] [Accepted: 05/28/2015] [Indexed: 11/19/2022] Open
Abstract
BACKGROUND Our previous study showed that parabromophenacyl bromide (PBPB) inhibits the features of allergic airway inflammation and airway hyperresponsiveness (AHR). However, its effect on airway remodeling, e.g. subepithelial fibrosis in a chronic allergic asthma model, was not investigated. We examined this issue in this study. METHODS PBPB was administered to mice with an induced chronic asthmatic condition. AHR was estimated at the end of the experiment, followed by euthanasia. Lung sections were stained with hematoxylin and eosin, periodic acid-Schiff and Masson's trichrome to determine airway inflammation, goblet cell metaplasia and subepithelial fibrosis, respectively. Transforming growth factor-β1 (TGF-β1) was estimated in lung homogenates. To determine the effect of PBPB on smooth-muscle hyperplasia, immunohistochemistry against α-smooth-muscle actin was performed on the lung sections. RESULTS Chronic ovalbumin challenges in a mouse model of allergic asthma caused significant subepithelial fibrosis and elevated TGF-β1, along with significant AHR. PBPB attenuated subepithelial fibrosis with a reduction of lung TGF-β1, airway inflammation and AHR without affecting goblet cell metaplasia. It also attenuated smooth-muscle hyperplasia with a reduction in the expression of α-smooth-muscle actin in the lungs. CONCLUSION Our findings indicate that PBPB attenuates some crucial features of airway remodeling such as subepithelial fibrosis and smooth-muscle hyperplasia. These data suggest that PBPB could therefore be a therapeutic drug for chronic asthma.
Collapse
Affiliation(s)
- Arjun Ram
- CSIR Institute of Genomics and Integrative Biology, Delhi, India
| | | | | | | | | | | |
Collapse
|
29
|
Ramanadham S, Ali T, Ashley JW, Bone RN, Hancock WD, Lei X. Calcium-independent phospholipases A2 and their roles in biological processes and diseases. J Lipid Res 2015; 56:1643-68. [PMID: 26023050 DOI: 10.1194/jlr.r058701] [Citation(s) in RCA: 146] [Impact Index Per Article: 14.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2015] [Indexed: 12/24/2022] Open
Abstract
Among the family of phospholipases A2 (PLA2s) are the Ca(2+)-independent PLA2s (iPLA2s) and they are designated group VI iPLA2s. In relation to secretory and cytosolic PLA2s, the iPLA2s are more recently described and details of their expression and roles in biological functions are rapidly emerging. The iPLA2s or patatin-like phospholipases (PNPLAs) are intracellular enzymes that do not require Ca(2+) for activity, and contain lipase (GXSXG) and nucleotide-binding (GXGXXG) consensus sequences. Though nine PNPLAs have been recognized, PNPLA8 (membrane-associated iPLA2γ) and PNPLA9 (cytosol-associated iPLA2β) are the most widely studied and understood. The iPLA2s manifest a variety of activities in addition to phospholipase, are ubiquitously expressed, and participate in a multitude of biological processes, including fat catabolism, cell differentiation, maintenance of mitochondrial integrity, phospholipid remodeling, cell proliferation, signal transduction, and cell death. As might be expected, increased or decreased expression of iPLA2s can have profound effects on the metabolic state, CNS function, cardiovascular performance, and cell survival; therefore, dysregulation of iPLA2s can be a critical factor in the development of many diseases. This review is aimed at providing a general framework of the current understanding of the iPLA2s and discussion of the potential mechanisms of action of the iPLA2s and related involved lipid mediators.
Collapse
Affiliation(s)
- Sasanka Ramanadham
- Cell, Developmental, and Integrative Biology, University of Alabama at Birmingham, Birmingham, AL 35294 Comprehensive Diabetes Center, University of Alabama at Birmingham, Birmingham, AL 35294
| | - Tomader Ali
- Undergraduate Research Office, University of Alabama at Birmingham, Birmingham, AL 35294
| | - Jason W Ashley
- Department of Orthopaedic Surgery, University of Pennsylvania, Philadelphia, PA 19104
| | - Robert N Bone
- Cell, Developmental, and Integrative Biology, University of Alabama at Birmingham, Birmingham, AL 35294 Comprehensive Diabetes Center, University of Alabama at Birmingham, Birmingham, AL 35294
| | - William D Hancock
- Cell, Developmental, and Integrative Biology, University of Alabama at Birmingham, Birmingham, AL 35294 Comprehensive Diabetes Center, University of Alabama at Birmingham, Birmingham, AL 35294
| | - Xiaoyong Lei
- Cell, Developmental, and Integrative Biology, University of Alabama at Birmingham, Birmingham, AL 35294 Comprehensive Diabetes Center, University of Alabama at Birmingham, Birmingham, AL 35294
| |
Collapse
|
30
|
Abstract
Enzymes underpin physiological function and exhibit dysregulation in many disease-associated microenvironments and aberrant cell processes. Exploiting altered enzyme activity and expression for diagnostics, drug targeting, and drug release is tremendously promising. When combined with booming research in nanobiotechnology, enzyme-responsive nanomaterials used for controlled drug release have achieved significant development and have been studied as an important class of drug delivery strategies in nanomedicine. In this review, we describe enzymes such as proteases, phospholipases and oxidoreductases that serve as delivery triggers. Subsequently, we explore recently developed enzyme-responsive nanomaterials with versatile applications for extracellular and intracellular drug delivery. We conclude by discussing future opportunities and challenges in this area.
Collapse
Affiliation(s)
- Quanyin Hu
- Joint Department of Biomedical Engineering, University of North Carolina at Chapel Hill and North Carolina State University, Raleigh, NC 27695, USA.
| | | | | |
Collapse
|
31
|
|
32
|
Kamčeva T, Radisavljević M, Vukićević I, Arnhold J, Petković M. Interactions of platinum and ruthenium coordination complexes with pancreatic phospholipase A(2) and phospholipids investigated by MALDI TOF mass spectrometry. Chem Biodivers 2014; 10:1972-86. [PMID: 24243606 DOI: 10.1002/cbdv.201300141] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2013] [Indexed: 11/07/2022]
Abstract
Phospholipase A2 is involved in propagation of inflammatory processes and carcinogenesis through its role in phospholipid metabolism, and release of arachidonic acid and lysophospholipids. Recent findings on correlation between elevated PLA2 activity and metastatic cancer render this enzyme an attractive target for cancer therapy. On the other hand, due to a broad range of oxidation states under physiological conditions and a high affinity for protein binding, platinum and ruthenium coordination complexes are promising candidates for PLA2 inhibitors. In this article, we discuss the interactions of Pt and Ru coordination complexes with PLA2 and phospholipids, as well as the application of MALDI-TOF mass spectrometry for screening PLA2 inhibitors. Owing to the ability of this technique to simultaneously detect and monitor changes in substrate and product concentrations, the inhibitor mechanisms of both Pt and Ru complexes with various ligands were determined.
Collapse
Affiliation(s)
- Tina Kamčeva
- 'Vinča' Institute of Nuclear Sciences, Department of Physical Chemistry, University of Belgrade, Mike Petrovića Alasa 12-14, RS-11001 Belgrade (phone: +381 11 3408 64; fax: +381 11 8066 434); Haukeland University Hospital, Laboratory of Clinical Biochemistry, Section of Clinical Pharmacology, Jonas Lies Vei 65, NO-5020 Bergen (phone: +47 46 572612; fax:+47 55 290 718).
| | | | | | | | | |
Collapse
|
33
|
Porterfield M, Zhao P, Han H, Cunningham J, Aoki K, Von Hoff DD, Demeure MJ, Pierce JM, Tiemeyer M, Wells L. Discrimination between adenocarcinoma and normal pancreatic ductal fluid by proteomic and glycomic analysis. J Proteome Res 2014; 13:395-407. [PMID: 24328148 PMCID: PMC3946306 DOI: 10.1021/pr400422g] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/24/2023]
Abstract
Sensitive and specific biomarkers for pancreatic cancer are currently unavailable. The high mortality associated with adenocarcinoma of the pancreatic epithelium justifies the broadest possible search for new biomarkers that can facilitate early detection or monitor treatment efficacy. Protein glycosylation is altered in many cancers, leading many to propose that glycoproteomic changes may provide suitable biomarkers. In order to assess this possibility for pancreatic cancer, we have performed an in-depth LC-MS/MS analysis of the proteome and MS(n)-based characterization of the N-linked glycome of a small set of pancreatic ductal fluid obtained from normal, pancreatitis, intraductal papillary mucinous neoplasm (IPMN), and pancreatic adenocarcinoma patients. Our results identify a set of seven proteins that were consistently increased in cancer ductal fluid compared to normal (AMYP, PRSS1, GP2-1, CCDC132, REG1A, REG1B, and REG3A) and one protein that was consistently decreased (LIPR2). These proteins are all directly or indirectly associated with the secretory pathway in normal pancreatic cells. Validation of these changes in abundance by Western blotting revealed increased REG protein glycoform diversity in cancer. Characterization of the total N-linked glycome of normal, IPMN, and adenocarcinoma ductal fluid clustered samples into three discrete groups based on the prevalence of six dominant glycans. Within each group, the profiles of less prevalent glycans were able to distinguish normal from cancer on this small set of samples. Our results emphasize that individual variation in protein glycosylation must be considered when assessing the value of a glycoproteomic marker, but also indicate that glycosylation diversity across human subjects can be reduced to simpler clusters of individuals whose N-linked glycans share structural features.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | | | - Lance Wells
- Co-corresponding authors: Michael Tiemeyer, and Lance Wells,
| |
Collapse
|
34
|
Sundarraj S, Thangam R, Sujitha MV, Vimala K, Kannan S. Ligand-conjugated mesoporous silica nanorattles based on enzyme targeted prodrug delivery system for effective lung cancer therapy. Toxicol Appl Pharmacol 2014; 275:232-43. [PMID: 24467950 DOI: 10.1016/j.taap.2014.01.012] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2013] [Revised: 01/03/2014] [Accepted: 01/14/2014] [Indexed: 10/25/2022]
Abstract
Epidermal growth factor receptor antibody (EGFRAb) conjugated silica nanorattles (SNs) were synthesized and used to develop receptor mediated endocytosis for targeted drug delivery strategies for cancer therapy. The present study determined that the rate of internalization of silica nanorattles was found to be high in lung cancer cells when compared with the normal lung cells. EGFRAb can specifically bind to EGFR, a receptor that is highly expressed in lung cancer cells, but is expressed at low levels in other normal cells. Furthermore, in vitro studies clearly substantiated that the cPLA2α activity, arachidonic acid release and cell proliferation were considerably reduced by pyrrolidine-2 loaded EGFRAb-SN in H460 cells. The cytotoxicity, cell cycle arrest and apoptosis were significantly induced by the treatment of pyrrolidine-2 loaded EGFRAb-SN when compared with free pyrrolidine-2 and pyrrolidine-2 loaded SNs in human non-small cell lung cancer cells. An in vivo toxicity assessment showed that silica nanorattles and EGFRAb-SN-pyrrolidine-2 exhibited low systemic toxicity in healthy Balb/c mice. The EGFRAb-SN-pyrrolidine-2 showed a much better antitumor activity (38%) with enhanced tumor inhibition rate than the pyrrolidine-2 on the non-small cell lung carcinoma subcutaneous model. Thus, the present findings validated the low toxicity and high therapeutic potentials of EGFRAb-SN-pyrrolidine-2, which may provide a convincing evidence of the silica nanorattles as new potential carriers for targeted drug delivery systems.
Collapse
Affiliation(s)
- Shenbagamoorthy Sundarraj
- Proteomics and Molecular Cell Physiology Laboratory, Department of Zoology, Bharathiar University, Coimbatore 641 046, TN, India.
| | - Ramar Thangam
- Proteomics and Molecular Cell Physiology Laboratory, Department of Zoology, Bharathiar University, Coimbatore 641 046, TN, India; Department of Virology, King Institute of Preventive Medicine and Research, Guindy, Chennai 600 032, TN, India
| | - Mohanan V Sujitha
- Proteomics and Molecular Cell Physiology Laboratory, Department of Zoology, Bharathiar University, Coimbatore 641 046, TN, India
| | - Karuppaiya Vimala
- Proteomics and Molecular Cell Physiology Laboratory, Department of Zoology, Bharathiar University, Coimbatore 641 046, TN, India
| | - Soundarapandian Kannan
- Proteomics and Molecular Cell Physiology Laboratory, Department of Zoology, Bharathiar University, Coimbatore 641 046, TN, India; Department of Zoology, Periyar University, Salem 636 011, TN, India.
| |
Collapse
|
35
|
Menschikowski M, Hagelgans A, Fuessel S, Mareninova OA, Asatryan L, Wirth MP, Siegert G. Serum amyloid A, phospholipase A2-IIA and C-reactive protein as inflammatory biomarkers for prostate diseases. Inflamm Res 2013; 62:1063-72. [DOI: 10.1007/s00011-013-0665-5] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2013] [Accepted: 09/04/2013] [Indexed: 11/29/2022] Open
|
36
|
Menschikowski M, Hagelgans A, Schuler U, Froeschke S, Rosner A, Siegert G. Plasma Levels of Phospholipase A2-IIA in Patients with Different Types of Malignancies: Prognosis and Association with Inflammatory and Coagulation Biomarkers. Pathol Oncol Res 2013; 19:839-46. [DOI: 10.1007/s12253-013-9652-y] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/12/2012] [Accepted: 05/05/2013] [Indexed: 12/13/2022]
|
37
|
Mass spectrometry-based quantitative metabolomics revealed a distinct lipid profile in breast cancer patients. Int J Mol Sci 2013; 14:8047-61. [PMID: 23584023 PMCID: PMC3645730 DOI: 10.3390/ijms14048047] [Citation(s) in RCA: 82] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2013] [Revised: 03/30/2013] [Accepted: 04/01/2013] [Indexed: 11/16/2022] Open
Abstract
Breast cancer accounts for the largest number of newly diagnosed cases in female cancer patients. Although mammography is a powerful screening tool, about 20% of breast cancer cases cannot be detected by this method. New diagnostic biomarkers for breast cancer are necessary. Here, we used a mass spectrometry-based quantitative metabolomics method to analyze plasma samples from 55 breast cancer patients and 25 healthy controls. A number of 30 patients and 20 age-matched healthy controls were used as a training dataset to establish a diagnostic model and to identify potential biomarkers. The remaining samples were used as a validation dataset to evaluate the predictive accuracy for the established model. Distinct separation was obtained from an orthogonal partial least squares-discriminant analysis (OPLS-DA) model with good prediction accuracy. Based on this analysis, 39 differentiating metabolites were identified, including significantly lower levels of lysophosphatidylcholines and higher levels of sphingomyelins in the plasma samples obtained from breast cancer patients compared with healthy controls. Using logical regression, a diagnostic equation based on three metabolites (lysoPC a C16:0, PC ae C42:5 and PC aa C34:2) successfully differentiated breast cancer patients from healthy controls, with a sensitivity of 98.1% and a specificity of 96.0%.
Collapse
|
38
|
Sundarraj S, Kannan S, Thangam R, Gunasekaran P. Effects of the inhibition of cytosolic phospholipase A(2)α in non-small cell lung cancer cells. J Cancer Res Clin Oncol 2012; 138:827-35. [PMID: 22274867 DOI: 10.1007/s00432-012-1157-7] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2011] [Accepted: 01/10/2012] [Indexed: 10/14/2022]
Abstract
PURPOSE The aim of this study was to investigate the expression of cPLA(2)α in non-small lung cancer cell lines and tissues, and we sought to determine the in vitro effects of the pyrrolidine-2 inhibitor on cPLA(2)α sensitivity in three different non-small lung cancer cell lines. METHODS The expression of cPLA(2)α was determined in lung cancer cells by Western blot. Cytotoxicity, cell growth and inhibition of cPLA(2)α activity were determined in relation to the concentration of pyrrolidine-2. Finally, this study investigated immunohistochemical expressions of cPLA(2)α in 23 species of human non-small lung cancer and 5 species of human normal lung to assess their clinicopathological relevance. RESULTS cPLA(2)α is expressed in A549 and H460, however, no expression in H661 cells. Pyrrolidine-2 demonstrated a dose-dependent inhibitory effect on cell growth and its significantly inhibited BrdU incorporation of human non-small lung cancer cells. Inhibition with pyrrolidine-2 results in reduction in cPLA(2)α activity in A549 and H460 lung cancer cells by 50% when present at IC(50) concentration in arachidonoyl thio-PC assay. Immunohistochemistry of human lung tissue revealed that cPLA(2)α is increased in lung cancer tissues. CONCLUSIONS Pyrrolidine-2 is a more potent and specific cPLA(2)α inhibitor than MAFP and AACOCF3 and represents an excellent pharmacological tool to investigate the biosynthesis and the biological roles of cancer. The present study suggests that pyrrolidine-2 could be a potential therapeutic agent for cancer therapy.
Collapse
Affiliation(s)
- Shenbagamoorthy Sundarraj
- Proteomics and Molecular Cell Physiology Laboratory, Department of Zoology, School of Life Sciences, Bharathiar University, Coimbatore, TN, India.
| | | | | | | |
Collapse
|
39
|
Serum Levels of Secreted Group IIA Phospholipase A2 in Benign Prostatic Hyperplasia and Prostate Cancer: A Biomarker for Inflammation or Neoplasia? Inflammation 2011; 35:1113-8. [DOI: 10.1007/s10753-011-9418-1] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
|
40
|
Dennis EA, Cao J, Hsu YH, Magrioti V, Kokotos G. Phospholipase A2 enzymes: physical structure, biological function, disease implication, chemical inhibition, and therapeutic intervention. Chem Rev 2011; 111:6130-85. [PMID: 21910409 PMCID: PMC3196595 DOI: 10.1021/cr200085w] [Citation(s) in RCA: 856] [Impact Index Per Article: 61.1] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Affiliation(s)
- Edward A. Dennis
- Department of Chemistry and Biochemistry and Pharmacology, School of Medicine, University of California, San Diego, La Jolla, California 92093-0601
| | - Jian Cao
- Department of Chemistry and Biochemistry and Pharmacology, School of Medicine, University of California, San Diego, La Jolla, California 92093-0601
| | - Yuan-Hao Hsu
- Department of Chemistry and Biochemistry and Pharmacology, School of Medicine, University of California, San Diego, La Jolla, California 92093-0601
| | - Victoria Magrioti
- Laboratory of Organic Chemistry, Department of Chemistry, University of Athens, Panepistimiopolis, Athens 15771, Greece
| | - George Kokotos
- Laboratory of Organic Chemistry, Department of Chemistry, University of Athens, Panepistimiopolis, Athens 15771, Greece
| |
Collapse
|
41
|
Towards novel radiosensitizing agents: the role of cytosolic PLA2α in combined modality cancer therapy. Future Med Chem 2011; 3:835-43. [PMID: 21644828 DOI: 10.4155/fmc.11.38] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
The radioresistant nature of some tumors serves as an obstacle to curative therapy for several poor-prognosis malignancies. The radiosensitivity of a cancer is dependent not only on the intrinsic ability of tumor cells to recover from radiation-induced damage, but also the ability of stromal elements (e.g., vasculature) in the tumor microenvironment to survive and continue proliferating in the face of ionizing radiation. In this regard, it is important to understand the initial events activating radiation-induced signal transduction pathways. Among these events is the activation of cytosolic phospholipase A2 α and the subsequent production of the lipid second messengers. These events occur within minutes following exposure to ionizing radiation, and have been shown to enhance cell viability through a number of prosurvival signaling pathways. Furthermore, inhibition of cytosolic phospholipase A2 α has now been shown to reduce the viability of endothelial cells in culture after exposure to ionizing radiation, as well as slowing the growth of tumors in animal models of cancer.
Collapse
|
42
|
Xing XF, Li H, Zhong XY, Zhang LH, Wang XH, Liu YQ, Jia SQ, Shi T, Niu ZJ, Peng Y, Du H, Zhang GG, Hu Y, Lu AP, Li JY, Chen S, Ji JF. Phospholipase A2 group IIA expression correlates with prolonged survival in gastric cancer. Histopathology 2011; 59:198-206. [DOI: 10.1111/j.1365-2559.2011.03913.x] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023]
|
43
|
Glunde K, Jiang L, Moestue SA, Gribbestad IS. MRS and MRSI guidance in molecular medicine: targeting and monitoring of choline and glucose metabolism in cancer. NMR IN BIOMEDICINE 2011; 24:673-90. [PMID: 21793073 PMCID: PMC3146026 DOI: 10.1002/nbm.1751] [Citation(s) in RCA: 44] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/22/2023]
Abstract
MRS and MRSI are valuable tools for the detection of metabolic changes in tumors. The currently emerging era of molecular medicine, which is shaped by molecularly targeted anticancer therapies combined with molecular imaging of the effects of such therapies, requires powerful imaging technologies that are able to detect molecular information. MRS and MRSI are such technologies that are able to detect metabolites arising from glucose and choline metabolism in noninvasive in vivo settings and at higher resolution in tissue samples. The roles played by MRS and MRSI in the diagnosis of different types of cancer, as well as in the early monitoring of the tumor response to traditional chemotherapies, are reviewed. The emerging roles of MRS and MRSI in the development and detection of novel targeted anticancer therapies that target oncogenic signaling pathways or markers in choline or glucose metabolism are discussed.
Collapse
Affiliation(s)
- Kristine Glunde
- Johns Hopkins University In Vivo Cellular and Molecular Imaging Center, Russell H. Morgan, Department of Radiology and Radiological Science, The Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Lu Jiang
- Johns Hopkins University In Vivo Cellular and Molecular Imaging Center, Russell H. Morgan, Department of Radiology and Radiological Science, The Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Siver A. Moestue
- Department of Circulation and Medical Imaging, Norwegian University of Science and Technology (NTNU), 7491 Trondheim, Norway
| | - Ingrid S. Gribbestad
- Department of Circulation and Medical Imaging, Norwegian University of Science and Technology (NTNU), 7491 Trondheim, Norway
| |
Collapse
|
44
|
Arouri A, Mouritsen OG. Anticancer double lipid prodrugs: liposomal preparation and characterization. J Liposome Res 2011; 21:296-305. [DOI: 10.3109/08982104.2011.563365] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
|
45
|
Caiazza F, McCarthy NS, Young L, Hill ADK, Harvey BJ, Thomas W. Cytosolic phospholipase A2-α expression in breast cancer is associated with EGFR expression and correlates with an adverse prognosis in luminal tumours. Br J Cancer 2010; 104:338-44. [PMID: 21119660 PMCID: PMC3031888 DOI: 10.1038/sj.bjc.6606025] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022] Open
Abstract
Background: The eicosanoid signalling pathway promotes the progression of malignancies through the production of proliferative prostaglandins (PGs). Cytosolic phospholipase A2α (cPLA2α) activity provides the substrate for cyclooxygenase-dependent PG release, and we have previously found that cPLA2α expression correlated with EGFR/HER2 over-expression in a small number of breast cancer cell lines. Methods: The importance of differential cPLA2α activity in clinical breast cancer was established by relating the expression of cPLA2α in tissue samples from breast cancer patients, and two microarray-based gene expression datasets to different clinicopathological and therapeutic parameters. Results: High cPLA2α mRNA expression correlated with clinical parameters of poor prognosis, which are characteristic of highly invasive tumours of the HER2-positive and basal-like subtype, including low oestrogen receptor expression and high EGFR expression. High cPLA2α expression decreased overall survival in patients with luminal cancers, and correlated with a reduced effect of tamoxifen treatment. The cPLA2α expression was an independent predictive parameter of poor response to endocrine therapy in the first 5 years of follow-up. Conclusion: This study shows a role of cPLA2α in luminal breast cancer progression, in which the enzyme could represent a novel therapeutic target and a predictive marker.
Collapse
Affiliation(s)
- F Caiazza
- Department of Molecular Medicine, Royal College of Surgeons in Ireland, Education and Research Centre, Beaumont Hospital, Dublin 9, Ireland
| | | | | | | | | | | |
Collapse
|
46
|
Early cessation of the clinical development of LiPlaCis, a liposomal cisplatin formulation. Eur J Cancer 2010; 46:3016-21. [DOI: 10.1016/j.ejca.2010.07.015] [Citation(s) in RCA: 63] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2010] [Accepted: 07/14/2010] [Indexed: 11/16/2022]
|
47
|
Andresen TL, Thompson DH, Kaasgaard T. Enzyme-triggered nanomedicine: drug release strategies in cancer therapy. Mol Membr Biol 2010; 27:353-63. [PMID: 20939771 DOI: 10.3109/09687688.2010.515950] [Citation(s) in RCA: 123] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
Abstract
Nanomedicine as a field has emerged from the early success of nanoparticle-based drug delivery systems, in particular for treatment of cancer, and the advances made in nano- and biotechnology over the past decade. A prerequisite for nanoparticle-based drug delivery systems to be effective is that the drug payload is released at the target site. A large number of drug release strategies have been proposed that can be classified into certain areas. The simplest and most successful strategy so far, probably due to relative simplicity, is based on utilizing certain physico-chemical characteristics of drugs to obtain a slow drug leakage from the formulations after accumulation in the cancerous site. However, this strategy is only applicable to a relatively small range of drugs and cannot be applied to biologicals. Many advanced drug release strategies have therefore been investigated. Such strategies include utilization of heat, light and ultrasound sensitive systems and in particular pH sensitive systems where the lower pH in endosomes induces drug release. Highly interesting are enzyme sensitive systems where over-expressed disease-associated enzymes are utilized to trigger drug release. The enzyme-based strategies are particularly interesting as they require no prior knowledge of the tumour localization. The basis of this review is an evaluation of the current status of drug delivery strategies focused on triggered drug release by disease-associated enzymes. We limit ourselves to reviewing the liposome field, but the concepts and conclusions are equally important for polymer-based systems.
Collapse
Affiliation(s)
- Thomas L Andresen
- Technical University of Denmark, DTU Nanotech, Department of Micro- and Nanotechnology, Roskilde, Denmark.
| | | | | |
Collapse
|
48
|
The expression and distribution of group IIA phospholipase A2 in human colorectal tumours. Virchows Arch 2010; 457:659-67. [PMID: 20938784 DOI: 10.1007/s00428-010-0992-7] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2010] [Revised: 09/01/2010] [Accepted: 09/29/2010] [Indexed: 10/19/2022]
Abstract
Secretory phospholipase A2 group IIA (IIA PLA2) is a protein shown to be increased in various human malignancies. The expression profile of this protein, however, is controversial in colorectal carcinoma. The aim of this study was to examine the distribution and expression of IIA PLA2 protein in benign, premalignant and malignant colorectal tumours as well as in peritumoural mucosa. Seven hyperplastic polyps, 24 adenomas and 83 colorectal carcinomas were stained with immunohistochemistry (IHC) for IIA PLA2. Four hyperplastic polyps, 12 adenomas and nine carcinomas were also evaluated for the sites of IIA PLA2 expression using mRNA in situ hybridisation (ISH). There was no immunoreactivity for IIA PLA2 in hyperplastic polyps. A total of 79% of adenomas and 31% of carcinomas showed IIA PLA2-immunopositive tumour cells in IHC, and the expression was localised to epithelial cells with ISH. In carcinomas, IIA PLA2-immunopositive apoptotic cells and necrosis were also found. The epithelial cells in the peritumoural mucosa showed immunopositivity for IIA PLA2 in 96% of cases, with considerably stronger intensity adjacent to carcinoma than in the more distal mucosa. Moreover, IIA PLA2-immunopositive malignant epithelial cells were found in 44% of cases in the invasive front of carcinomas. Our results suggest that the IIA PLA2 protein content is dramatically decreased in malignant colorectal tumours as compared with adenomas. The protein is also found in the apoptotic cells, necrosis, peritumoural mucosa and in the invasive front of carcinomas.
Collapse
|
49
|
Lim SC, Cho H, Lee TB, Choi CH, Min YD, Kim SS, Kim KJ. Impacts of cytosolic phospholipase A2, 15-prostaglandin dehydrogenase, and cyclooxygenase-2 expressions on tumor progression in colorectal cancer. Yonsei Med J 2010; 51:692-699. [PMID: 20635443 PMCID: PMC2908865 DOI: 10.3349/ymj.2010.51.5.692] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/07/2009] [Revised: 11/27/2009] [Accepted: 12/11/2009] [Indexed: 02/07/2023] Open
Abstract
PURPOSE In addition to cyclooxygenase-2 (COX-2) which is related to prostaglandin E2 synthesis, other enzymes such as cytosolic phospholipase A2 (cPLA2), microsomal prostaglandin E2 synthase-1 (mPGES-1), and 15-prostaglandin dehydrogenase (15-PGDH) have been suggested to be related to carcinogenesis of colorectal cancer (CRC). The aim of this study was to investigate the roles of cPLA2, COX-2, mPGES-1, and 15-PGDH in tumor progression. MATERIALS AND METHODS cPLA2, COX-2, mPGES-1, 15-PGDH, and vascular endothelial growth factor (VEGF) expressions were immunohistochemically examined in 89 CRC, and their expressions were compared with each other or clinicopathologic parameters as well as VEGF as tumor progression parameters. RESULTS cPLA2 was expressed in 54.5%, COX-2 in 80.5%, mPGES-1 in 96.4%, 15-PGDH in 46.1%, and VEGF in 65.9%. The expression of cPLA2 correlated with VEGF expression. COX-2 expression was correlated with the depth of invasion, tumor stage, cPLA2, and VEGF expressions. Moreover, VEGF revealed the highest expression in the tissues positive for both cPLA2 and COX-2. Furthermore, 15-PGDH expression was inversely correlated with VEGF expression. CONCLUSION The present study demonstrates that cPLA2 and mPGES-1, in addition to COX-2, are constitutively overexpressed, and that 15-PGDH might be attenuated in colorectal cancer. Furthermore, cPLA2 and 15-PGDH as well as COX-2 could have an important role in tumor progression.
Collapse
Affiliation(s)
- Sung Chul Lim
- Department of Pathology, Chosun University, Gwangju, Korea
- Research Center for Resistant Cells, Chosun University, Gwangju, Korea
| | - Hoon Cho
- Research Center for Resistant Cells, Chosun University, Gwangju, Korea
- Department of Polymer Science & Engineering, Chosun University, Gwangju, Korea
| | - Tae Bum Lee
- Research Center for Resistant Cells, Chosun University, Gwangju, Korea
| | - Cheol Hee Choi
- Research Center for Resistant Cells, Chosun University, Gwangju, Korea
| | - Young Don Min
- Department of Surgery, College of Medicine, Chosun University, Gwangju, Korea
| | - Sung Soo Kim
- Department of Surgery, College of Medicine, Chosun University, Gwangju, Korea
| | - Kyung Jong Kim
- Department of Surgery, College of Medicine, Chosun University, Gwangju, Korea
| |
Collapse
|
50
|
Mauchley D, Meng X, Babu A, Gamboni-Robertson F, Yang X, Banerjee A, Fullerton DA, Weyant MJ. Gastroduodenal reflux induces group IIa secretory phospholipase A(2) expression and activity in murine esophagus. Dis Esophagus 2010; 23:430-6. [PMID: 20095996 DOI: 10.1111/j.1442-2050.2009.01034.x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Exposure of esophageal epithelium to gastric and duodenal contents results in the histologic changes of hyperproliferation and mucosal thickening. We have previously shown that presence of secretory phospholipase A(2) (sPLA(2)) is necessary to produce these histologic changes in a murine model of gastroduodenal reflux. We sought to determine the influence of gastroduodenal reflux (GDR) on sPLA(2) protein and mRNA levels as well as enzyme activity in esophageal tissue. BALB/c (sPLA(2)(+/+)) mice (n= 28) underwent side-to-side surgical anastomosis of the first portion of the duodenum and GE junction (DGEA) resulting in continuous exposure of esophageal mucosa to mixed gastric and duodenal contents. Sham control mice (n= 14) underwent laparotomy, esophagotomy and closure. Real-time RT PCR was used to quantitate the influence of GDR on group IIa sPLA(2) expression. Immunofluorescent staining was quantitated by digital microscopy using a specific antibody to identify and locate sPLA(2) protein. A colorimetric assay was used to quantify total sPLA(2) activity after standardization of protein levels. Statistical analysis was conducted using Student's t-test. Group IIa sPLA(2) mRNA and protein levels were increased at 4 and 8 weeks compared with sham controls. This increase occurred in a time-dependent manner and correlated with esophageal mucosal thickness. Furthermore, sPLA(2) enzyme activity was increased significantly at 4 and 8 weeks compared with untreated controls. The expression of group IIa sPLA(2) as well as sPLA(2) activity is induced by GDR. This novel finding indicates that sPLA(2) may play a role in the development of the histologic changes produced by GDR in esophageal mucosa.
Collapse
Affiliation(s)
- David Mauchley
- Department of Surgery, Division of Cardiothoracic Surgery, University of Colorado at Denver and Health Sciences Center, Denver, Colorado, USA
| | | | | | | | | | | | | | | |
Collapse
|