1
|
Yuan Y, Lin Q, Feng HY, Zhang Y, Lai X, Zhu MH, Wang J, Shi J, Huang Y, Zhang L, Lu Q, Yuan Z, Lovell JF, Chen HZ, Sun P, Fang C. A multistage drug delivery approach for colorectal primary tumors and lymph node metastases. Nat Commun 2025; 16:1439. [PMID: 39920155 PMCID: PMC11806101 DOI: 10.1038/s41467-025-56768-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2024] [Accepted: 01/23/2025] [Indexed: 02/09/2025] Open
Abstract
The presence of lymph node (LN) metastases guides cancer staging and worsens prognoses. Incomplete lymphadenectomy of metastatic LNs may end up with disease recurrence, while excessive resection can result in increased postoperative complications with even no survival benefit. Thus, effective non-invasive methods to treat metastatic LNs would be highly desirable. Here, we develop an enzyme-responsive formulation of small-sized doxorubicin-loaded mesoporous silica nanoparticles (DMSN, 40 nm) encapsulated in nanoliposomes (DMSN@Pla-Lipo, 160 nm). The liposomal membrane contains 1,2-dipalmitoyl-sn-glycero-3-phospho-rac-(1-glycerol) (DPPG) and 1,2-dipalmitoyl-sn-glycero-3-phosphocholine (DPPC), two phospholipids sensitive to secreted phospholipase A2 in human colorectal tumors. In an orthotopic colorectal murine tumor model, phospholipase-induced membrane permeabilization triggers the liberation of DMSN from liposomes for enhanced tumor penetration, conferring an enhanced suppression for the primary tumor. Furthermore, through translocation into metastatic LNs via tumor lymphatics, metastatic tumor cells in LNs are eradicated. Metastases to other major organs are also suppressed, which can be ascribed to the inhibition of colorectal cancer metastasis-associated TGF-β, Wnt, and Hippo signaling pathways in metastatic LNs. The treatment confers an 80% 90-day survival rate in this aggressive tumor model. Taken together, this study demonstrates a deliberate treatment approach for management of both primary tumors and metastatic LNs through multistage drug delivery.
Collapse
Affiliation(s)
- Yihang Yuan
- Hongqiao International Institute of Medicine, Tongren Hospital and State Key Laboratory of Systems Medicine for Cancer, Department of Pharmacology and Chemical Biology, Shanghai Jiao Tong University School of Medicine (SJTU-SM), Shanghai, 200025, China
| | - Quanjun Lin
- Department of General Surgery, Tongren Hospital, SJTU-SM, Shanghai, 200336, China
| | - Hai-Yi Feng
- Hongqiao International Institute of Medicine, Tongren Hospital and State Key Laboratory of Systems Medicine for Cancer, Department of Pharmacology and Chemical Biology, Shanghai Jiao Tong University School of Medicine (SJTU-SM), Shanghai, 200025, China
| | - Yunpeng Zhang
- Department of General Surgery, Tongren Hospital, SJTU-SM, Shanghai, 200336, China
| | - Xing Lai
- Hongqiao International Institute of Medicine, Tongren Hospital and State Key Laboratory of Systems Medicine for Cancer, Department of Pharmacology and Chemical Biology, Shanghai Jiao Tong University School of Medicine (SJTU-SM), Shanghai, 200025, China
| | - Mao-Hua Zhu
- Hongqiao International Institute of Medicine, Tongren Hospital and State Key Laboratory of Systems Medicine for Cancer, Department of Pharmacology and Chemical Biology, Shanghai Jiao Tong University School of Medicine (SJTU-SM), Shanghai, 200025, China
| | - Jue Wang
- Department of General Surgery, Tongren Hospital, SJTU-SM, Shanghai, 200336, China
| | - Jiangpei Shi
- Hongqiao International Institute of Medicine, Tongren Hospital and State Key Laboratory of Systems Medicine for Cancer, Department of Pharmacology and Chemical Biology, Shanghai Jiao Tong University School of Medicine (SJTU-SM), Shanghai, 200025, China
| | - Yanhu Huang
- Hongqiao International Institute of Medicine, Tongren Hospital and State Key Laboratory of Systems Medicine for Cancer, Department of Pharmacology and Chemical Biology, Shanghai Jiao Tong University School of Medicine (SJTU-SM), Shanghai, 200025, China
| | - Lele Zhang
- Hongqiao International Institute of Medicine, Tongren Hospital and State Key Laboratory of Systems Medicine for Cancer, Department of Pharmacology and Chemical Biology, Shanghai Jiao Tong University School of Medicine (SJTU-SM), Shanghai, 200025, China
| | - Qin Lu
- Hongqiao International Institute of Medicine, Tongren Hospital and State Key Laboratory of Systems Medicine for Cancer, Department of Pharmacology and Chemical Biology, Shanghai Jiao Tong University School of Medicine (SJTU-SM), Shanghai, 200025, China
| | - Zeli Yuan
- Key Laboratory of Basic Pharmacology of Ministry of Education & Joint International Research Laboratory of Ethnomedicine of Ministry of Education, Zunyi Medical University, Zunyi, 563003, China
| | - Jonathan F Lovell
- Department of Biomedical Engineering, University at Buffalo, State University of New York, Buffalo, NY, 14260, USA
| | - Hong-Zhuan Chen
- Shanghai Frontiers Science Center of TCM Chemical Biology, Institute of Interdisciplinary Integrative Biomedical Research, Shuguang Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China
| | - Peng Sun
- Department of General Surgery, Tongren Hospital, SJTU-SM, Shanghai, 200336, China.
| | - Chao Fang
- Hongqiao International Institute of Medicine, Tongren Hospital and State Key Laboratory of Systems Medicine for Cancer, Department of Pharmacology and Chemical Biology, Shanghai Jiao Tong University School of Medicine (SJTU-SM), Shanghai, 200025, China.
- Key Laboratory of Basic Pharmacology of Ministry of Education & Joint International Research Laboratory of Ethnomedicine of Ministry of Education, Zunyi Medical University, Zunyi, 563003, China.
| |
Collapse
|
2
|
Xie J, Pink DL, Jayne Lawrence M, Lorenz CD. Digestion of lipid micelles leads to increased membrane permeability. NANOSCALE 2024; 16:2642-2653. [PMID: 38229565 DOI: 10.1039/d3nr05083a] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/18/2024]
Abstract
Lipid-based drug carriers are an attractive option to solubilise poorly water soluble therapeutics. Previously, we reported that the digestion of a short tail PC lipid (2C6PC) by the PLA2 enzyme has a significant effect on the structure and stability of the micelles it forms. Here, we studied the interactions of micelles of varying composition representing various degrees of digestion with a model ordered (70 mol% DPPC & 30 mol% cholesterol) and disordered (100% DOPC) lipid membrane. Micelles of all compositions disassociated when interacting with the two different membranes. As the percentage of digestion products (C6FA and C6LYSO) in the micelle increased, the disassociation occurred more rapidly. The C6FA inserts preferentially into both membranes. We find that all micelle components increase the area per lipid, increase the disorder and decrease the thickness of the membranes, and the 2C6PC lipid molecules have the most significant impact. Additionally, there is an increase in permeation of water into the membrane that accompanies the insertion of C6FA into the DOPC membranes. We show that the natural digestion of lipid micelles result in molecular species that can enhance the permeability of lipid membranes that in turn result in an enhanced delivery of drugs.
Collapse
Affiliation(s)
- Jun Xie
- Biological & Soft Matter Research Group, Department of Physics, Faculty of Natural, Mathematical & Engineering Sciences, King's College London, London, UK.
| | - Demi L Pink
- Biological & Soft Matter Research Group, Department of Physics, Faculty of Natural, Mathematical & Engineering Sciences, King's College London, London, UK.
| | - M Jayne Lawrence
- Division of Pharmacy and Optometry, School of Health Sciences, Faculty of Biology, Medicine and Health, University of Manchester, Stopford Building, Oxford Road, Manchester, UK
| | - Christian D Lorenz
- Biological & Soft Matter Research Group, Department of Physics, Faculty of Natural, Mathematical & Engineering Sciences, King's College London, London, UK.
| |
Collapse
|
3
|
Liang J, Li L, Li L, Zhou X, Zhang Z, Huang Y, Xiao X. Lipid metabolism reprogramming in head and neck cancer. Front Oncol 2023; 13:1271505. [PMID: 37927468 PMCID: PMC10622980 DOI: 10.3389/fonc.2023.1271505] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2023] [Accepted: 10/09/2023] [Indexed: 11/07/2023] Open
Abstract
Lipid metabolism reprogramming is one of the most prominent metabolic anomalies in cancer, wherein cancer cells undergo dysregulation of lipid metabolism to acquire adequate energy, cell membrane building blocks, as well as signaling molecules essential for cell proliferation, survival, invasion, and metastasis. These adaptations enable cancer cells to effectively respond to challenges posed by the tumor microenvironment, leading to cancer therapy resistance and poor cancer prognosis. Head and neck cancer, ranking as the seventh most prevalent cancer, exhibits numerous abnormalities in lipid metabolism. Nevertheless, the precise role of lipid metabolic rewiring in head and neck cancer remains unclear. In line with the LIPID MAPS Lipid Classification System and cancer risk factors, the present review delves into the dysregulated molecules and pathways participating in the process of lipid uptake, biosynthesis, transportation, and catabolism. We also present an overview of the latest advancements in understanding alterations in lipid metabolism and how they intersect with the carcinogenesis, development, treatment, and prognosis of head and neck cancer. By shedding light on the significance of metabolic therapy, we aspire to improve the overall prognosis and treatment outcomes of head and neck cancer patients.
Collapse
Affiliation(s)
- Jinfeng Liang
- Department of Otolaryngology-Head and Neck Surgery, First Affiliated Hospital of Guangxi Medical University, Nanning, China
| | - Lin Li
- Department of Otolaryngology-Head and Neck Surgery, First Affiliated Hospital of Guangxi Medical University, Nanning, China
| | - Limei Li
- Department of Pediatric Dentistry, College & Hospital of Stomatology, Guangxi Medical University, Nanning, China
| | - Xiaoying Zhou
- Key Laboratory of Early Prevention and Treatment for Regional High-Frequency Tumor, Guangxi Medical University, Ministry of Education, Nanning, China
| | - Zhe Zhang
- Department of Otolaryngology-Head and Neck Surgery, First Affiliated Hospital of Guangxi Medical University, Nanning, China
- Key Laboratory of Early Prevention and Treatment for Regional High-Frequency Tumor, Guangxi Medical University, Ministry of Education, Nanning, China
| | - Yi Huang
- Department of Otolaryngology-Head and Neck Surgery, First Affiliated Hospital of Guangxi Medical University, Nanning, China
| | - Xue Xiao
- Department of Otolaryngology-Head and Neck Surgery, First Affiliated Hospital of Guangxi Medical University, Nanning, China
- Key Laboratory of Early Prevention and Treatment for Regional High-Frequency Tumor, Guangxi Medical University, Ministry of Education, Nanning, China
| |
Collapse
|
4
|
Nel J, Elkhoury K, Velot É, Bianchi A, Acherar S, Francius G, Tamayol A, Grandemange S, Arab-Tehrany E. Functionalized liposomes for targeted breast cancer drug delivery. Bioact Mater 2023; 24:401-437. [PMID: 36632508 PMCID: PMC9812688 DOI: 10.1016/j.bioactmat.2022.12.027] [Citation(s) in RCA: 59] [Impact Index Per Article: 29.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2022] [Revised: 12/05/2022] [Accepted: 12/25/2022] [Indexed: 01/03/2023] Open
Abstract
Despite the exceptional progress in breast cancer pathogenesis, prognosis, diagnosis, and treatment strategies, it remains a prominent cause of female mortality worldwide. Additionally, although chemotherapies are effective, they are associated with critical limitations, most notably their lack of specificity resulting in systemic toxicity and the eventual development of multi-drug resistance (MDR) cancer cells. Liposomes have proven to be an invaluable drug delivery system but of the multitudes of liposomal systems developed every year only a few have been approved for clinical use, none of which employ active targeting. In this review, we summarize the most recent strategies in development for actively targeted liposomal drug delivery systems for surface, transmembrane and internal cell receptors, enzymes, direct cell targeting and dual-targeting of breast cancer and breast cancer-associated cells, e.g., cancer stem cells, cells associated with the tumor microenvironment, etc.
Collapse
Affiliation(s)
- Janske Nel
- Université de Lorraine, LIBio, F-54000, Nancy, France
| | | | - Émilie Velot
- Université de Lorraine, CNRS, IMoPA, F-54000, Nancy, France
| | - Arnaud Bianchi
- Université de Lorraine, CNRS, IMoPA, F-54000, Nancy, France
| | - Samir Acherar
- Université de Lorraine, CNRS, LCPM, F-54000, Nancy, France
| | | | - Ali Tamayol
- Department of Biomedical Engineering, University of Connecticut Health Center, Farmington, CT, 06030, USA
| | | | | |
Collapse
|
5
|
Khan SA, Ilies MA. The Phospholipase A2 Superfamily: Structure, Isozymes, Catalysis, Physiologic and Pathologic Roles. Int J Mol Sci 2023; 24:ijms24021353. [PMID: 36674864 PMCID: PMC9862071 DOI: 10.3390/ijms24021353] [Citation(s) in RCA: 66] [Impact Index Per Article: 33.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2022] [Revised: 12/23/2022] [Accepted: 01/06/2023] [Indexed: 01/13/2023] Open
Abstract
The phospholipase A2 (PLA2) superfamily of phospholipase enzymes hydrolyzes the ester bond at the sn-2 position of the phospholipids, generating a free fatty acid and a lysophospholipid. The PLA2s are amphiphilic in nature and work only at the water/lipid interface, acting on phospholipid assemblies rather than on isolated single phospholipids. The superfamily of PLA2 comprises at least six big families of isoenzymes, based on their structure, location, substrate specificity and physiologic roles. We are reviewing the secreted PLA2 (sPLA2), cytosolic PLA2 (cPLA2), Ca2+-independent PLA2 (iPLA2), lipoprotein-associated PLA2 (LpPLA2), lysosomal PLA2 (LPLA2) and adipose-tissue-specific PLA2 (AdPLA2), focusing on the differences in their structure, mechanism of action, substrate specificity, interfacial kinetics and tissue distribution. The PLA2s play important roles both physiologically and pathologically, with their expression increasing significantly in diseases such as sepsis, inflammation, different cancers, glaucoma, obesity and Alzheimer's disease, which are also detailed in this review.
Collapse
|
6
|
Thangam C, Cyril R, Sekar R, Jayasree R, Ramachandran V, Langeswaran K, Asir AB, Subbaraj GK. Role of phospholipase A2 in squamous cell carcinoma and breast cancer. PHOSPHOLIPASES IN PHYSIOLOGY AND PATHOLOGY 2023:315-335. [DOI: 10.1016/b978-0-323-95697-0.00010-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/02/2025]
|
7
|
Jiang Y, Jiang Z, Wang M, Ma L. Current understandings and clinical translation of nanomedicines for breast cancer therapy. Adv Drug Deliv Rev 2022; 180:114034. [PMID: 34736986 DOI: 10.1016/j.addr.2021.114034] [Citation(s) in RCA: 32] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2021] [Revised: 10/20/2021] [Accepted: 10/28/2021] [Indexed: 02/08/2023]
Abstract
Breast cancer is one of the most frequently diagnosed cancers that is threatening women's life. Current clinical treatment regimens for breast cancer often involve neoadjuvant and adjuvant systemic therapies, which somewhat are associated with unfavorable features. Also, the heterogeneous nature of breast cancers requires precision medicine that cannot be fulfilled by a single type of systemically administered drug. Taking advantage of the nanocarriers, nanomedicines emerge as promising therapeutic agents for breast cancer that could resolve the defects of drugs and achieve precise drug delivery to almost all sites of primary and metastatic breast tumors (e.g. tumor vasculature, tumor stroma components, breast cancer cells, and some immune cells). Seven nanomedicines as represented by Doxil® have been approved for breast cancer clinical treatment so far. More nanomedicines including both non-targeting and active targeting nanomedicines are being evaluated in the clinical trials. However, we have to realize that the translation of nanomedicines, particularly the active targeting nanomedicines is not as successful as people have expected. This review provides a comprehensive landscape of the nanomedicines for breast cancer treatment, from laboratory investigations to clinical applications. We also highlight the key advances in the understanding of the biological fate and the targeting strategies of breast cancer nanomedicine and the implications to clinical translation.
Collapse
|
8
|
The Use of Nanomedicine to Target Signaling by the PAK Kinases for Disease Treatment. Cells 2021; 10:cells10123565. [PMID: 34944073 PMCID: PMC8700304 DOI: 10.3390/cells10123565] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2021] [Revised: 12/14/2021] [Accepted: 12/15/2021] [Indexed: 12/11/2022] Open
Abstract
P21-activated kinases (PAKs) are serine/threonine kinases involved in the regulation of cell survival, proliferation, inhibition of apoptosis, and the regulation of cell morphology. Some members of the PAK family are highly expressed in several types of cancer, and they have also been implicated in several other medical disorders. They are thus considered to be good targets for treatment of cancer and other diseases. Although there are several inhibitors of the PAKs, the utility of some of these inhibitors is reduced for several reasons, including limited metabolic stability. One way to overcome this problem is the use of nanoparticles, which have the potential to increase drug delivery. The overall goals of this review are to describe the roles for PAK kinases in cell signaling and disease, and to describe how the use of nanomedicine is a promising new method for administering PAK inhibitors for the purpose of disease treatment and research. We discuss some of the basic mechanisms behind nanomedicine technology, and we then describe how these techniques are being used to package and deliver PAK inhibitors.
Collapse
|
9
|
Scott KF, Mann TJ, Fatima S, Sajinovic M, Razdan A, Kim RR, Cooper A, Roohullah A, Bryant KJ, Gamage KK, Harman DG, Vafaee F, Graham GG, Church WB, Russell PJ, Dong Q, de Souza P. Human Group IIA Phospholipase A 2-Three Decades on from Its Discovery. Molecules 2021; 26:molecules26237267. [PMID: 34885848 PMCID: PMC8658914 DOI: 10.3390/molecules26237267] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2021] [Revised: 11/21/2021] [Accepted: 11/22/2021] [Indexed: 12/13/2022] Open
Abstract
Phospholipase A2 (PLA2) enzymes were first recognized as an enzyme activity class in 1961. The secreted (sPLA2) enzymes were the first of the five major classes of human PLA2s to be identified and now number nine catalytically-active structurally homologous proteins. The best-studied of these, group IIA sPLA2, has a clear role in the physiological response to infection and minor injury and acts as an amplifier of pathological inflammation. The enzyme has been a target for anti-inflammatory drug development in multiple disorders where chronic inflammation is a driver of pathology since its cloning in 1989. Despite intensive effort, no clinically approved medicines targeting the enzyme activity have yet been developed. This review catalogues the major discoveries in the human group IIA sPLA2 field, focusing on features of enzyme function that may explain this lack of success and discusses future research that may assist in realizing the potential benefit of targeting this enzyme. Functionally-selective inhibitors together with isoform-selective inhibitors are necessary to limit the apparent toxicity of previous drugs. There is also a need to define the relevance of the catalytic function of hGIIA to human inflammatory pathology relative to its recently-discovered catalysis-independent function.
Collapse
Affiliation(s)
- Kieran F. Scott
- School of Medicine, Western Sydney University, Campbelltown, NSW 2560, Australia; (T.J.M.); (S.F.); (A.C.); (A.R.); (P.d.S.)
- Ingham Institute of Applied Medical Research, Liverpool, NSW 2170, Australia; (M.S.); (A.R.)
- Correspondence: ; Tel.: +61-2-8738-9026
| | - Timothy J. Mann
- School of Medicine, Western Sydney University, Campbelltown, NSW 2560, Australia; (T.J.M.); (S.F.); (A.C.); (A.R.); (P.d.S.)
- Ingham Institute of Applied Medical Research, Liverpool, NSW 2170, Australia; (M.S.); (A.R.)
| | - Shadma Fatima
- School of Medicine, Western Sydney University, Campbelltown, NSW 2560, Australia; (T.J.M.); (S.F.); (A.C.); (A.R.); (P.d.S.)
- Ingham Institute of Applied Medical Research, Liverpool, NSW 2170, Australia; (M.S.); (A.R.)
- School of Biotechnology and Biological Sciences, University of New South Wales (UNSW Sydney), Sydney, NSW 2052, Australia;
| | - Mila Sajinovic
- Ingham Institute of Applied Medical Research, Liverpool, NSW 2170, Australia; (M.S.); (A.R.)
| | - Anshuli Razdan
- Ingham Institute of Applied Medical Research, Liverpool, NSW 2170, Australia; (M.S.); (A.R.)
| | - Ryung Rae Kim
- School of Pharmacy, Faculty of Medicine and Health, University of Sydney, Sydney, NSW 2006, Australia; (R.R.K.); (W.B.C.)
| | - Adam Cooper
- School of Medicine, Western Sydney University, Campbelltown, NSW 2560, Australia; (T.J.M.); (S.F.); (A.C.); (A.R.); (P.d.S.)
- Ingham Institute of Applied Medical Research, Liverpool, NSW 2170, Australia; (M.S.); (A.R.)
- Liverpool Cancer Therapy Centre, Liverpool Hospital, Liverpool, NSW 2170, Australia
| | - Aflah Roohullah
- School of Medicine, Western Sydney University, Campbelltown, NSW 2560, Australia; (T.J.M.); (S.F.); (A.C.); (A.R.); (P.d.S.)
- Ingham Institute of Applied Medical Research, Liverpool, NSW 2170, Australia; (M.S.); (A.R.)
- Liverpool Cancer Therapy Centre, Liverpool Hospital, Liverpool, NSW 2170, Australia
| | - Katherine J. Bryant
- School of Photovoltaic and Renewable Energy Engineering, UNSW Sydney, Sydney, NSW 2052, Australia;
| | - Kasuni K. Gamage
- School of Science, Western Sydney University, Campbelltown, NSW 2560, Australia; (K.K.G.); (D.G.H.)
| | - David G. Harman
- School of Science, Western Sydney University, Campbelltown, NSW 2560, Australia; (K.K.G.); (D.G.H.)
| | - Fatemeh Vafaee
- School of Biotechnology and Biological Sciences, University of New South Wales (UNSW Sydney), Sydney, NSW 2052, Australia;
- UNSW Data Science Hub, UNSW Sydney, Sydney, NSW 2052, Australia
| | - Garry G. Graham
- Department of Clinical Pharmacology, St Vincent’s Hospital Sydney, Darlinghurst, NSW 2010, Australia;
- School of Medical Sciences, UNSW Sydney, Sydney, NSW 2052, Australia
| | - W. Bret Church
- School of Pharmacy, Faculty of Medicine and Health, University of Sydney, Sydney, NSW 2006, Australia; (R.R.K.); (W.B.C.)
| | - Pamela J. Russell
- Australian Prostate Cancer Research Centre—QUT, Brisbane, QLD 4102, Australia;
| | - Qihan Dong
- Chinese Medicine Anti-Cancer Evaluation Program, Greg Brown Laboratory, Central Clinical School and Charles Perkins Centre, The University of Sydney, Sydney, NSW 2006, Australia;
| | - Paul de Souza
- School of Medicine, Western Sydney University, Campbelltown, NSW 2560, Australia; (T.J.M.); (S.F.); (A.C.); (A.R.); (P.d.S.)
- Ingham Institute of Applied Medical Research, Liverpool, NSW 2170, Australia; (M.S.); (A.R.)
- School of Medicine, UNSW Sydney, Sydney, NSW 2052, Australia
| |
Collapse
|
10
|
Phospholipase Signaling in Breast Cancer. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2021. [PMID: 33983572 DOI: 10.1007/978-981-32-9620-6_2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/23/2023]
Abstract
Breast cancer progression results from subversion of multiple intra- or intercellular signaling pathways in normal mammary tissues and their microenvironment, which have an impact on cell differentiation, proliferation, migration, and angiogenesis. Phospholipases (PLC, PLD and PLA) are essential mediators of intra- and intercellular signaling. They hydrolyze phospholipids, which are major components of cell membrane that can generate many bioactive lipid mediators, such as diacylglycerol, phosphatidic acid, lysophosphatidic acid, and arachidonic acid. Enzymatic processing of phospholipids by phospholipases converts these molecules into lipid mediators that regulate multiple cellular processes, which in turn can promote breast cancer progression. Thus, dysregulation of phospholipases contributes to a number of human diseases, including cancer. This review describes how phospholipases regulate multiple cancer-associated cellular processes, and the interplay among different phospholipases in breast cancer. A thorough understanding of the breast cancer-associated signaling networks of phospholipases is necessary to determine whether these enzymes are potential targets for innovative therapeutic strategies.
Collapse
|
11
|
Rapid Screening and Identification of Antitumor Ingredients from the Mangrove Endophytic Fungus Using an Enzyme-Immobilized Magnetic Nanoparticulate System. Molecules 2021; 26:molecules26082255. [PMID: 33924693 PMCID: PMC8069786 DOI: 10.3390/molecules26082255] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2021] [Revised: 04/07/2021] [Accepted: 04/07/2021] [Indexed: 12/15/2022] Open
Abstract
As a consequence of recent progression in biomedicine and nanotechnology, nanoparticle-based systems have evolved as a new method with extensive applications in responsive therapy, multimodal imaging, drug delivery and natural product separation. Meanwhile, the magnetic nanoparticulate system has aroused great interest for separation and purification because of its excellent magnetic properties. Phospholipase A2 (PLA2) is a highly expressed regulator to promote the growth of various cancers and is an ideal target to treat cancers. In this study, a novel strategy based on ligand–receptor interactions to discover novel PLA2 inhibitors was established, in which PLA2-functionalized Fe3O4@PLGA-PEG-NH2 magnetic nanoparticles were used as a supporting material combined with high-performance liquid chromatography–mass spectrometry, aiming to accelerate the discovery of novel PLA2 inhibitors from natural sources such as mangrove endophytic fungi. Under the optimized ligand fishing conditions, six target compounds were ultimately fished and identified to be cyclic peptides (1–3) and sterols (4–6), which compounds 1, 2 and 4–6 have well-documented cytotoxicities. Compound 3 exerted better inhibitory effect on A549 cells by experiment. In conclusion, PLA2-functionalized Fe3O4@PLGA-PEG-NH2 magnetic nanoparticles-based ligand fishing provided a feasible, selective and effective platform for the efficient screening and identification of antitumor components from natural products.
Collapse
|
12
|
Analytical Platforms for the Determination of Phospholipid Turnover in Breast Cancer Tissue: Role of Phospholipase Activity in Breast Cancer Development. Metabolites 2021; 11:metabo11010032. [PMID: 33406793 PMCID: PMC7824782 DOI: 10.3390/metabo11010032] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2020] [Revised: 12/26/2020] [Accepted: 12/31/2020] [Indexed: 12/31/2022] Open
Abstract
Altered lipid metabolism has been associated with the progression of various cancers, and aberrant expression of enzymes involved in the lipid metabolism has been detected in different stages of cancer. Breast cancer (BC) is one of the cancer types known to be associated with alterations in the lipid metabolism and overexpression of enzymes involved in this metabolism. It has been demonstrated that inhibition of the activity of certain enzymes, such as that of phospholipase A2 in BC cell lines sensitizes these cells and decreases the IC50 values for forthcoming therapy with traditional drugs, such as doxorubicin and tamoxifen. Moreover, other phospholipases, such as phospholipase C and D, are involved in intracellular signal transduction, which emphasizes their importance in cancer development. Finally, BC is assumed to be dependent on the diet and the composition of lipids in nutrients. Despite their importance, analytical approaches that can associate the activity of phospholipases with changes in the lipid composition and distribution in cancer tissues are not yet standardized. In this review, an overview of various analytical platforms that are applied on the study of lipids and phospholipase activity in BC tissues will be given, as well as their association with cancer diagnosis and tumor progression. The methods that are applied to tissues obtained from the BC patients will be emphasized and critically evaluated, regarding their applicability in oncology.
Collapse
|
13
|
Pan M, Qin C, Han X. Lipid Metabolism and Lipidomics Applications in Cancer Research. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2021; 1316:1-24. [PMID: 33740240 PMCID: PMC8287890 DOI: 10.1007/978-981-33-6785-2_1] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/12/2023]
Abstract
Lipids are the critical components of cellular and plasma membrane, which constitute an impermeable barrier of cellular compartments, and play important roles on numerous cellular processes including cell growth, proliferation, differentiation, and signaling. Alterations in lipid metabolism have been implicated in the development and progression of cancers. However, unlike other biomolecules, the diversity in the structures and characteristics of lipid species results in the limited understanding of their metabolic alterations in cancers. Lipidomics is an emerging discipline that studies lipids in a large scale based on analytical chemistry principles and technological tools. Multidimensional mass spectrometry-based shotgun lipidomics (MDMS-SL) uses direct infusion to avoid difficulties from alterations in concentration, chromatographic anomalies, and ion-pairing alterations to improve resolution and achieve rapid and accurate qualitative and quantitative analysis. In this chapter, lipids and lipid metabolism relevant to cancer research are introduced, followed by a brief description of MDMS-SL and other shotgun lipidomics techniques and some applications for cancer research.
Collapse
Affiliation(s)
- Meixia Pan
- Barshop Institute for Longevity and Aging Studies, San Antonio, TX, USA
| | - Chao Qin
- Barshop Institute for Longevity and Aging Studies, San Antonio, TX, USA
| | - Xianlin Han
- Barshop Institute for Longevity and Aging Studies, San Antonio, TX, USA.
- Department of Medicine - Diabetes, University of Texas Health Science Center at San Antonio, San Antonio, TX, USA.
| |
Collapse
|
14
|
Liposomes Targeting P21 Activated Kinase-1 (PAK-1) and Selective for Secretory Phospholipase A 2 (sPLA 2) Decrease Cell Viability and Induce Apoptosis in Metastatic Triple-Negative Breast Cancer Cells. Int J Mol Sci 2020; 21:ijms21249396. [PMID: 33321758 PMCID: PMC7764208 DOI: 10.3390/ijms21249396] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2020] [Revised: 12/04/2020] [Accepted: 12/07/2020] [Indexed: 12/12/2022] Open
Abstract
P21 activated kinases (or group I PAKs) are serine/threonine kinases whose expression is altered in prostate and breast cancers. PAK-1 activity is inhibited by the small molecule "Inhibitor targeting PAK-1 activation-3" (IPA-3), which has selectivity for PAK-1 but is metabolically unstable. Secretory Group IIA phospholipase A2 (sPLA2) expression correlates to increased metastasis and decreased survival in many cancers. We previously designed novel liposomal formulations targeting both PAK-1 and sPLA2, called Secretory Phospholipase Responsive liposomes or SPRL-IPA-3, and demonstrated their ability to alter prostate cancer growth. The efficacy of SPRL against other types of cancers is not well understood. We addressed this limitation by determining the ability of SPRL to induce cell death in a diverse panel of cells representing different stages of breast cancer, including the invasive but non-metastatic MCF-7 cells, and metastatic triple-negative breast cancer (TNBC) cells such as MDA-MB-231, MDA-MB-468, and MDA-MB-435. We investigated the role of sPLA2 in the disposition of these liposomes by comparing the efficacy of SPRL-IPA-3 to IPA-3 encapsulated in sterically stabilized liposomes (SSL-IPA-3), a formulation shown to be less sensitive to sPLA2. Both SSL-IPA-3 and SPRL-IPA-3 induced time- and dose-dependent decreases in MTT staining in all cell lines tested, but SPRL-IPA-3-induced effects in metastatic TNBC cell lines were superior over SSL-IPA-3. The reduction in MTT staining induced by SPRL-IPA-3 correlated to the expression of Group IIA sPLA2. sPLA2 expression also correlated to increased induction of apoptosis in TNBC cell lines by SPRL-IPA-3. These data suggest that SPRL-IPA-3 is selective for metastatic TNBC cells and that the efficacy of SPRL-IPA-3 is mediated, in part, by the expression of Group IIA sPLA2.
Collapse
|
15
|
Verma A, Najahi-Missaoui W, Cummings BS, Somanath PR. Sterically stabilized liposomes targeting P21 (RAC1) activated kinase-1 and secreted phospholipase A 2 suppress prostate cancer growth and metastasis. Oncol Lett 2020; 20:179. [PMID: 32934746 PMCID: PMC7471734 DOI: 10.3892/ol.2020.12040] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2020] [Accepted: 07/22/2020] [Indexed: 12/11/2022] Open
Abstract
Metastatic prostate cancer (PCa) has a very high mortality rate in men, in Western countries and lacks reliable treatment. The advanced-stage PCa cells overexpress P21 (RAC1) activated kinase-1 (PAK1) and secreted phospholipase A2 (sPLA2) suggesting the potential utility of pharmacologically targeting these molecules to treat metastatic PCa. The small molecule, inhibitor targeting PAK1 activation-3 (IPA3) is a highly specific allosteric inhibitor of PAK1; however, it is metabolically unstable once in the plasma thus, limiting its utility as a chemotherapeutic agent. In the present study, the efficacy and specificity of IPA3 were combined with the stability and the sPLA2-targeted delivery method of two sterically stabilized liposomes [sterically stabilized long-circulating liposomes (SSL)-IPA3 and sPLA2 responsive liposomes (SPRL)-IPA3, respectively] to inhibit PCa growth and metastasis. It was found that twice-a-week administration of either SSL-IPA3 or SPRL-IPA3 for 3 weeks effectively suppressed the growth of PC-3 cell tumor xenografts implanted in athymic nude mice. Both drug formulations also inhibited the metastasis of intravenously administered murine RM1 PCa cells to the lungs of C57BL/6 mice. Whereas the twice-a-week administration of SSL-IPA3 significantly inhibited the spontaneous PCa metastasis to the lungs in Transgenic Adenocarcinoma of the Mouse Prostate mice, the administration of free IPA3 had no significant therapeutic benefit. The results present two novel IPA3 encapsulated liposomes to treat metastatic PCa.
Collapse
Affiliation(s)
- Arti Verma
- Program in Clinical and Experimental Therapeutics, University of Georgia, Augusta, GA 30912, USA
- Charlie Norwood Veterans Affairs Medical Center, Augusta, GA 30904, USA
| | - Wided Najahi-Missaoui
- Department of Pharmaceutical and Biomedical Sciences, University of Georgia, Athens, GA 30602, USA
| | - Brian S. Cummings
- Department of Pharmaceutical and Biomedical Sciences, University of Georgia, Athens, GA 30602, USA
- Interdisciplinary Toxicology Program, University of Georgia, Augusta, GA 30602, USA
| | - Payaningal R. Somanath
- Program in Clinical and Experimental Therapeutics, University of Georgia, Augusta, GA 30912, USA
- Charlie Norwood Veterans Affairs Medical Center, Augusta, GA 30904, USA
- Department of Medicine and Cancer Center, Augusta University, Augusta, GA 30602, USA
| |
Collapse
|
16
|
Peng Z, Chang Y, Fan J, Ji W, Su C. Phospholipase A2 superfamily in cancer. Cancer Lett 2020; 497:165-177. [PMID: 33080311 DOI: 10.1016/j.canlet.2020.10.021] [Citation(s) in RCA: 74] [Impact Index Per Article: 14.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2020] [Revised: 10/11/2020] [Accepted: 10/14/2020] [Indexed: 12/12/2022]
Abstract
Phospholipase A2 enzymes (PLA2s) comprise a superfamily that is generally divided into six subfamilies known as cytosolic PLA2s (cPLA2s), calcium-independent PLA2s (iPLA2s), secreted PLA2s (sPLA2s), lysosomal PLA2s, platelet-activating factor (PAF) acetylhydrolases, and adipose specific PLA2s. Each subfamily consists of several isozymes that possess PLA2 activity. The first three PLA2 subfamilies play important roles in inflammation-related diseases and cancer. In this review, the roles of well-studied enzymes sPLA2-IIA, cPLA2α and iPLA2β in carcinogenesis and cancer development were discussed. sPLA2-IIA seems to play conflicting roles and can act as a tumor suppressor or a tumor promoter according to the cancer type, but cPLA2α and iPLA2β play protumorigenic role in most cancers. The mechanisms of PLA2-mediated signal transduction and crosstalk between cancer cells and endothelial cells in the tumor microenvironment are described. Moreover, the mechanisms by which PLA2s mediate lipid reprogramming and glycerophospholipid remodeling in cancer cells are illustrated. PLA2s as the upstream regulators of the arachidonic acid cascade are generally high expressed and activated in various cancers. Therefore, they can be considered as potential pharmacological targets and biomarkers in cancer. The detailed information summarized in this review may aid in understanding the roles of PLA2s in cancer, and provide new clues for the development of novel agents and strategies for tumor prevention and treatment.
Collapse
Affiliation(s)
- Zhangxiao Peng
- Department of Molecular Oncology, Eastern Hepatobiliary Surgical Hospital & National Center for Liver Cancer, Navy Military Medical University, Shanghai, 200438, China.
| | - Yanxin Chang
- Department of Biliary Tract Surgery IV, Eastern Hepatobiliary Surgical Hospital, Navy Military Medical University, Shanghai, 200438, China.
| | - Jianhui Fan
- Mengchao Hepatobiliary Hospital, Fujian Medical University, Fuzhou, 350025, Fujian Province, China.
| | - Weidan Ji
- Department of Molecular Oncology, Eastern Hepatobiliary Surgical Hospital & National Center for Liver Cancer, Navy Military Medical University, Shanghai, 200438, China.
| | - Changqing Su
- Department of Molecular Oncology, Eastern Hepatobiliary Surgical Hospital & National Center for Liver Cancer, Navy Military Medical University, Shanghai, 200438, China.
| |
Collapse
|
17
|
Liu G, Lovell JF, Zhang L, Zhang Y. Stimulus-Responsive Nanomedicines for Disease Diagnosis and Treatment. Int J Mol Sci 2020; 21:E6380. [PMID: 32887466 PMCID: PMC7504550 DOI: 10.3390/ijms21176380] [Citation(s) in RCA: 34] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2020] [Revised: 08/26/2020] [Accepted: 08/31/2020] [Indexed: 02/07/2023] Open
Abstract
Stimulus-responsive drug delivery systems generally aim to release the active pharmaceutical ingredient (API) in response to specific conditions and have recently been explored for disease treatments. These approaches can also be extended to molecular imaging to report on disease diagnosis and management. The stimuli used for activation are based on differences between the environment of the diseased or targeted sites, and normal tissues. Endogenous stimuli include pH, redox reactions, enzymatic activity, temperature and others. Exogenous site-specific stimuli include the use of magnetic fields, light, ultrasound and others. These endogenous or exogenous stimuli lead to structural changes or cleavage of the cargo carrier, leading to release of the API. A wide variety of stimulus-responsive systems have been developed-responsive to both a single stimulus or multiple stimuli-and represent a theranostic tool for disease treatment. In this review, stimuli commonly used in the development of theranostic nanoplatforms are enumerated. An emphasis on chemical structure and property relationships is provided, aiming to focus on insights for the design of stimulus-responsive delivery systems. Several examples of theranostic applications of these stimulus-responsive nanomedicines are discussed.
Collapse
Affiliation(s)
- Gengqi Liu
- School of Chemical Engineering and Technology, Tianjin University, Tianjin 300350, China;
- Key Laboratory of Systems Bioengineering (Ministry of Education), Tianjin University, Tianjin 300072, China
| | - Jonathan F. Lovell
- Department of Biomedical Engineering, The State University of New York at Buffalo, Buffalo, NY 14260, USA;
| | - Lei Zhang
- School of Chemical Engineering and Technology, Tianjin University, Tianjin 300350, China;
- Key Laboratory of Systems Bioengineering (Ministry of Education), Tianjin University, Tianjin 300072, China
| | - Yumiao Zhang
- School of Chemical Engineering and Technology, Tianjin University, Tianjin 300350, China;
- Key Laboratory of Systems Bioengineering (Ministry of Education), Tianjin University, Tianjin 300072, China
| |
Collapse
|
18
|
Silva AAR, Cardoso MR, Rezende LM, Lin JQ, Guimaraes F, Silva GRP, Murgu M, Priolli DG, Eberlin MN, Tata A, Eberlin LS, Derchain SFM, Porcari AM. Multiplatform Investigation of Plasma and Tissue Lipid Signatures of Breast Cancer Using Mass Spectrometry Tools. Int J Mol Sci 2020; 21:E3611. [PMID: 32443844 PMCID: PMC7279467 DOI: 10.3390/ijms21103611] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2020] [Revised: 05/02/2020] [Accepted: 05/08/2020] [Indexed: 02/06/2023] Open
Abstract
Plasma and tissue from breast cancer patients are valuable for diagnostic/prognostic purposes and are accessible by multiple mass spectrometry (MS) tools. Liquid chromatography-mass spectrometry (LC-MS) and ambient mass spectrometry imaging (MSI) were shown to be robust and reproducible technologies for breast cancer diagnosis. Here, we investigated whether there is a correspondence between lipid cancer features observed by desorption electrospray ionization (DESI)-MSI in tissue and those detected by LC-MS in plasma samples. The study included 28 tissues and 20 plasma samples from 24 women with ductal breast carcinomas of both special and no special type (NST) along with 22 plasma samples from healthy women. The comparison of plasma and tissue lipid signatures revealed that each one of the studied matrices (i.e., blood or tumor) has its own specific molecular signature and the full interposition of their discriminant ions is not possible. This comparison also revealed that the molecular indicators of tissue injury, characteristic of the breast cancer tissue profile obtained by DESI-MSI, do not persist as cancer discriminators in peripheral blood even though some of them could be found in plasma samples.
Collapse
Affiliation(s)
- Alex Ap. Rosini Silva
- Postgraduate Program of Health Sciences, São Francisco University, Bragança Paulista SP 12916-900, Brazil; (A.A.R.S.); (D.G.P.)
| | - Marcella R. Cardoso
- Department of Gynecological and Breast Oncology, Women’s Hospital (CAISM), Faculty of Medical Sciences, State University of Campinas (UNICAMP), Campinas SP 13083-881, Brazil; (M.R.C.); (L.M.R.); (F.G.); (S.F.M.D.)
| | - Luciana Montes Rezende
- Department of Gynecological and Breast Oncology, Women’s Hospital (CAISM), Faculty of Medical Sciences, State University of Campinas (UNICAMP), Campinas SP 13083-881, Brazil; (M.R.C.); (L.M.R.); (F.G.); (S.F.M.D.)
| | - John Q. Lin
- Department of Chemistry, The University of Texas at Austin, Austin, TX 78712, USA; (J.Q.L.); (L.S.E.)
| | - Fernando Guimaraes
- Department of Gynecological and Breast Oncology, Women’s Hospital (CAISM), Faculty of Medical Sciences, State University of Campinas (UNICAMP), Campinas SP 13083-881, Brazil; (M.R.C.); (L.M.R.); (F.G.); (S.F.M.D.)
| | - Geisilene R. Paiva Silva
- Laboratory of Molecular and Investigative Pathology—LAPE, Women’s Hospital (CAISM), Faculty of Medical Sciences, State University of Campinas (UNICAMP), Campinas SP 13083-881, Brazil;
| | - Michael Murgu
- Waters Corporation, São Paulo, SP 13083-970, Brazil;
| | - Denise Gonçalves Priolli
- Postgraduate Program of Health Sciences, São Francisco University, Bragança Paulista SP 12916-900, Brazil; (A.A.R.S.); (D.G.P.)
| | - Marcos N. Eberlin
- School of Engineering, Mackenzie Presbyterian University, São Paulo SP 01302-907, Brazil;
| | - Alessandra Tata
- Laboratorio di Chimica Sperimentale, Istituto Zooprofilattico Sperimentale delle Venezie, Viale Fiume 78, 36100 Vicenza, Italy;
| | - Livia S. Eberlin
- Department of Chemistry, The University of Texas at Austin, Austin, TX 78712, USA; (J.Q.L.); (L.S.E.)
| | - Sophie F. M. Derchain
- Department of Gynecological and Breast Oncology, Women’s Hospital (CAISM), Faculty of Medical Sciences, State University of Campinas (UNICAMP), Campinas SP 13083-881, Brazil; (M.R.C.); (L.M.R.); (F.G.); (S.F.M.D.)
| | - Andreia M. Porcari
- Postgraduate Program of Health Sciences, São Francisco University, Bragança Paulista SP 12916-900, Brazil; (A.A.R.S.); (D.G.P.)
| |
Collapse
|
19
|
Silva CL, Perestrelo R, Sousa-Ferreira I, Capelinha F, Câmara JS, Petković M. Lipid biosignature of breast cancer tissues by matrix-assisted laser desorption/ionization time-of-flight mass spectrometry. Breast Cancer Res Treat 2020; 182:9-19. [DOI: 10.1007/s10549-020-05672-9] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2020] [Accepted: 05/06/2020] [Indexed: 12/24/2022]
|
20
|
Xiao D, Zhou R. Advances in the Application of Liposomal Nanosystems in Anticancer Therapy. Curr Stem Cell Res Ther 2020; 16:14-22. [PMID: 32324519 DOI: 10.2174/1574888x15666200423093906] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2019] [Revised: 02/03/2020] [Accepted: 02/17/2020] [Indexed: 02/08/2023]
Abstract
Cancer is the disease with the highest mortality rate, which poses a great threat to people's lives. Cancer caused approximately 3.4 million death worldwide annually. Surgery, chemotherapy and radiotherapy are the main therapeutic methods in clinical practice. However, surgery is only suitable for patients with early-stage cancers, and chemotherapy as well as radiotherapy have various side effects, both of which limit the application of available therapeutic methods. In 1965, liposome was firstly developed to form new drug delivery systems given the unique properties of nanoparticles, such as enhanced permeability and retention effect. During the last 5 decades, liposome has been widely used for the purpose of anticancer drug delivery, and several advances have been made regarding liposomal technology, including long-circulating liposomes, active targeting liposomes and triggered release liposomes, while problems exist all along. This review introduced the advances as well as the problems during the development of liposomal nanosystems for cancer therapy in recent years.
Collapse
Affiliation(s)
- Dexuan Xiao
- State Key Laboratory of Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, China
| | - Ronghui Zhou
- State Key Laboratory of Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, China
| |
Collapse
|
21
|
Hossain S, Pai KR, Piyasena ME. Fluorescent Lipo-Beads for the Sensitive Detection of Phospholipase A 2 and Its Inhibitors. ACS Biomater Sci Eng 2020; 6:1989-1997. [PMID: 33455318 PMCID: PMC10012499 DOI: 10.1021/acsbiomaterials.9b01720] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023]
Abstract
Phospholipase A2 (PLA2) is a membrane lytic enzyme that is present in many organisms. Human PLA2 has emerged as a potential biomarker as well as a therapeutic target for several diseases including cancer, cardiovascular diseases, and some inflammatory diseases. The current study focuses on the development of lipo-beads that are very reactive and highly sensitive to PLA2. To develop the best supported lipid bilayer formulation, several lipid combinations were investigated using 10 μm porous silica beads. The reactivity of PLA2 was monitored via the decrease in particle fluorescence because of the release of entrapped fluorescent dye from the particle pores or the disintegration of a fluorescent lipid constituted on the bilayer upon lipid hydrolysis using flow cytometry. The enzyme binding studies indicate that lipo-beads with bulky fluorescent tags in the lipid head group and anionic lipids produce a more pronounced response. The kinetic studies suggest that these lipo-beads are very reactive with PLA2 and can generate a detectable signal in less than 5 min. The enzyme inhibition studies were also conducted with two known PLA2 inhibitors, varespladib and quercetin. We find that quercetin can hydrolyze the supported membrane, and thus inhibition of PLA2 is not observed; however, varespladib has shown significant PLA2 inhibition on lipo-beads. We have demonstrated that our lipo-bead-based approach can detect annexin-3, a known disease biomarker, as low as 10 nM within 5 min after incubation.
Collapse
Affiliation(s)
- Shahriare Hossain
- Department of Chemistry, New Mexico Institute of Mining and Technology, 801, Leroy Place, Socorro, New Mexico 87801, United States
| | - Kalika R Pai
- Department of Chemistry, New Mexico Institute of Mining and Technology, 801, Leroy Place, Socorro, New Mexico 87801, United States
| | - Menake E Piyasena
- Department of Chemistry, New Mexico Institute of Mining and Technology, 801, Leroy Place, Socorro, New Mexico 87801, United States
| |
Collapse
|
22
|
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
|
23
|
Sales TA, Marcussi S, Ramalho TC. Current Anti-Inflammatory Therapies and the Potential of Secretory Phospholipase A2 Inhibitors in the Design of New Anti-Inflammatory Drugs: A Review of 2012 - 2018. Curr Med Chem 2020; 27:477-497. [PMID: 30706775 DOI: 10.2174/0929867326666190201120646] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2018] [Revised: 11/12/2018] [Accepted: 12/11/2018] [Indexed: 01/30/2023]
Abstract
The inflammatory process is a natural self-defense response of the organism to damage agents and its action mechanism involves a series of complex reactions. However, in some cases, this process can become chronic, causing much harm to the body. Therefore, over the years, many anti-inflammatory drugs have been developed aiming to decrease the concentrations of inflammatory mediators in the organism, which is a way of controlling these abnormal chain reactions. The main target of conventional anti-inflammatory drugs is the cyclooxygenase (COX) enzyme, but its use implies several side effects. Thus, based on these limitations, many studies have been performed, aiming to create new drugs, with new action mechanisms. In this sense, the phospholipase A2 (PLA2) enzymes stand out. Among all the existing isoforms, secretory PLA2 is the major target for inhibitor development, since many studies have proven that this enzyme participates in various inflammatory conditions, such as cancer, Alzheimer and arthritis. Finally, for the purpose of developing anti-inflammatory drugs that are sPLA2 inhibitors, many molecules have been designed. Accordingly, this work presents an overview of inflammatory processes and mediators, the current available anti-inflammatory drugs, and it briefly covers the PLA2 enzymes, as well as the diverse structural array of the newest sPLA2 inhibitors as a possible target for the production of new anti-inflammatory drugs.
Collapse
Affiliation(s)
- Thais A Sales
- Molecular Modeling Laboratory, Chemistry Department, Federal University of Lavras, 37200-000 Lavras, Brazil
| | - Silvana Marcussi
- Biochemistry Laboratory, Chemistry Department, Federal University of Lavras, 37200-000 Lavras, Brazil
| | - Teodorico C Ramalho
- Molecular Modeling Laboratory, Chemistry Department, Federal University of Lavras, 37200-000 Lavras, Brazil.,Center for Basic and Applied Research, Faculty of Informatics and Management, University of Hradec Kralove, 62, 50003 Rokitanskeho, Czech Republic
| |
Collapse
|
24
|
Zhang P, Villanueva V, Kalkowski J, Liu C, Pham T, Perez-Salas U, Bu W, Lin B, Liu Y. Polyunsaturated Phospholipid Modified Membrane Degradation Catalyzed by a Secreted Phospholipase A2. LANGMUIR : THE ACS JOURNAL OF SURFACES AND COLLOIDS 2019; 35:11643-11650. [PMID: 31401834 DOI: 10.1021/acs.langmuir.9b01476] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/10/2023]
Abstract
To optimize the compositions of the lipid-based nanomedicine and to advance understanding of the roles of polyunsaturated phospholipids in biological membranes, this study examined the effects of polyunsaturated phospholipids on the degradation of giant unilamellar vesicles catalyzed by a secreted phospholipase A2 (sPLA2) using fluorescence microscopy. Molecular interfacial packing, interaction, and degradation of the films containing various mixing ratios of saturated and polyunsaturated phospholipids were quantified using a Langmuir trough integrated with synchrotron X-ray surface scattering techniques. It was found that a high molar fraction (0.63 and above) of polyunsaturated phospholipids not only enhanced the rate of sPLA2-catalyzed vesicle degradation but also changed the vesicle deformation process and degradation product morphology. Hydrolysis of the saturated phospholipids generated highly ordered liquid crystal domains, which was reduced or prohibited by the presence of the polyunsaturated phospholipids in the reactant film.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Wei Bu
- NSF's ChemMatCARS , University of Chicago , Chicago , Illinois 60637 , United States
| | - Binhua Lin
- NSF's ChemMatCARS , University of Chicago , Chicago , Illinois 60637 , United States
| | | |
Collapse
|
25
|
Zhang P, Villanueva V, Kalkowski J, Liu C, Donovan AJ, Bu W, Schlossman ML, Lin B, Liu Y. Molecular interactions of phospholipid monolayers with a model phospholipase. SOFT MATTER 2019; 15:4068-4077. [PMID: 30958491 DOI: 10.1039/c8sm01154k] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/09/2023]
Abstract
The intrinsic overexpression of secretory phospholipase A2 (sPLA2) in various pro-inflammatory diseases and cancers has the potential to be exploited as a therapeutic strategy for diagnostics and treatment. To explore this potential and advance our knowledge of the role of sPLA2 in related diseases, it is necessary to systematically investigate the molecular interaction of the enzyme with lipids. By employing a Langmuir trough integrated with X-ray reflectivity and grazing incidence X-ray diffraction techniques, this study examined the molecular packing structure of 1,2-palmitoyl-sn-glycero-3-phosphocholine (DPPC) films before and after enzyme adsorption and enzyme-catalyzed degradation. Molecular interaction of sPLA2 (from bee venom) with the DPPC monolayer exhibited Ca2+ dependence. DPPC molecules at the interface without Ca2+ retained a monolayer organization; upon adsorption of sPLA2 to the monolayer the packing became tighter. In contrast, sPLA2-catalyzed degradation of DPPC occurred in the presence of Ca2+, leading to disruption of the ordered monolayer structure of DPPC. The interfacial film became a mixture of highly ordered multilayer domains of palmitic acid (PA) and loosely packed monolayer phase of 1-palmitoyl-2-hydroxy-sn-glycero-3-phosphocholine (lysoPC) that potentially contained the remaining un-degraded DPPC. The redistribution of lipid degradation products into the third dimension, which produced multilayer PA domains, damaged the structural integrity of the original lipid layer and may explain the bursting of liposomes observed in other studies after a latency period of mixing liposomes with sPLA2. A quantitative understanding of the lipid packing and lipid-enzyme interaction provides an intuitive means of designing and optimizing lipid-related drug delivery systems.
Collapse
Affiliation(s)
- Pin Zhang
- Department of Chemical Engineering, University of Illinois at Chicago, Chicago, IL 60607, USA.
| | | | | | | | | | | | | | | | | |
Collapse
|
26
|
Zhang S, Li Y, Qin Z, Tu G, Chen G, Yan A. SAR study on inhibitors of GIIA secreted phospholipase A
2
using machine learning methods. Chem Biol Drug Des 2019; 93:666-684. [DOI: 10.1111/cbdd.13470] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2018] [Revised: 12/14/2018] [Accepted: 12/17/2018] [Indexed: 12/24/2022]
Affiliation(s)
- Shengde Zhang
- State Key Laboratory of Chemical Resource EngineeringDepartment of Pharmaceutical EngineeringBeijing University of Chemical Technology Beijing China
| | - Yang Li
- Institute of Science and TechnologyShandong University of Traditional Chinese Medicine Ji'nan Shandong China
| | - Zijian Qin
- State Key Laboratory of Chemical Resource EngineeringDepartment of Pharmaceutical EngineeringBeijing University of Chemical Technology Beijing China
| | - Guipin Tu
- State Key Laboratory of Chemical Resource EngineeringDepartment of Pharmaceutical EngineeringBeijing University of Chemical Technology Beijing China
| | - Guang Chen
- College of Life Science and TechnologyBeijing University of Chemical Technology Beijing China
| | - Aixia Yan
- State Key Laboratory of Chemical Resource EngineeringDepartment of Pharmaceutical EngineeringBeijing University of Chemical Technology Beijing China
| |
Collapse
|
27
|
Olusanya TOB, Haj Ahmad RR, Ibegbu DM, Smith JR, Elkordy AA. Liposomal Drug Delivery Systems and Anticancer Drugs. Molecules 2018; 23:E907. [PMID: 29662019 PMCID: PMC6017847 DOI: 10.3390/molecules23040907] [Citation(s) in RCA: 344] [Impact Index Per Article: 49.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2018] [Revised: 03/29/2018] [Accepted: 04/09/2018] [Indexed: 12/27/2022] Open
Abstract
Cancer is a life-threatening disease contributing to ~3.4 million deaths worldwide. There are various causes of cancer, such as smoking, being overweight or obese, intake of processed meat, radiation, family history, stress, environmental factors, and chance. The first-line treatment of cancer is the surgical removal of solid tumours, radiation therapy, and chemotherapy. The systemic administration of the free drug is considered to be the main clinical failure of chemotherapy in cancer treatment, as limited drug concentration reaches the tumour site. Most of the active pharmaceutical ingredients (APIs) used in chemotherapy are highly cytotoxic to both cancer and normal cells. Accordingly, targeting the tumour vasculatures is essential for tumour treatment. In this context, encapsulation of anti-cancer drugs within the liposomal system offers secure platforms for the targeted delivery of anti-cancer drugs for the treatment of cancer. This, in turn, can be helpful for reducing the cytotoxic side effects of anti-cancer drugs on normal cells. This short-review focuses on the use of liposomes in anti-cancer drug delivery.
Collapse
Affiliation(s)
- Temidayo O B Olusanya
- School of Pharmacy and Pharmaceutical Sciences, University of Sunderland, Sunderland SR1 3SD, UK.
| | - Rita Rushdi Haj Ahmad
- School of Pharmacy and Pharmaceutical Sciences, University of Sunderland, Sunderland SR1 3SD, UK.
| | - Daniel M Ibegbu
- Department of Medical Biochemistry, College of Medicine, University of Nigeria Enugu Campus, Nigeria.
| | - James R Smith
- School of Pharmacy and Biomedical Sciences, University of Portsmouth, Portsmouth PO1 2DT, UK.
| | - Amal Ali Elkordy
- School of Pharmacy and Pharmaceutical Sciences, University of Sunderland, Sunderland SR1 3SD, UK.
| |
Collapse
|
28
|
Cala MP, Aldana J, Medina J, Sánchez J, Guio J, Wist J, Meesters RJW. Multiplatform plasma metabolic and lipid fingerprinting of breast cancer: A pilot control-case study in Colombian Hispanic women. PLoS One 2018; 13:e0190958. [PMID: 29438405 PMCID: PMC5810980 DOI: 10.1371/journal.pone.0190958] [Citation(s) in RCA: 46] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2017] [Accepted: 12/22/2017] [Indexed: 01/22/2023] Open
Abstract
Breast cancer (BC) is a highly heterogeneous disease associated with metabolic reprogramming. The shifts in the metabolome caused by BC still lack data from Latin populations of Hispanic origin. In this pilot study, metabolomic and lipidomic approaches were performed to establish a plasma metabolic fingerprint of Colombian Hispanic women with BC. Data from 1H-NMR, GC-MS and LC-MS were combined and compared. Statistics showed discrimination between breast cancer and healthy subjects on all analytical platforms. The differentiating metabolites were involved in glycerolipid, glycerophospholipid, amino acid and fatty acid metabolism. This study demonstrates the usefulness of multiplatform approaches in metabolic/lipid fingerprinting studies to broaden the outlook of possible shifts in metabolism. Our findings propose relevant plasma metabolites that could contribute to a better understanding of underlying metabolic shifts driven by BC in women of Colombian Hispanic origin. Particularly, the understanding of the up-regulation of long chain fatty acyl carnitines and the down-regulation of cyclic phosphatidic acid (cPA). In addition, the mapped metabolic signatures in breast cancer were similar but not identical to those reported for non-Hispanic women, despite racial differences.
Collapse
Affiliation(s)
- Mónica P. Cala
- Department of Chemistry, Grupo de Investigación en Química Analítica y Bioanalítica (GABIO), Universidad de los Andes, Bogotá D.C., Colombia
| | - Julian Aldana
- Department of Chemistry, Grupo de Investigación en Química Analítica y Bioanalítica (GABIO), Universidad de los Andes, Bogotá D.C., Colombia
| | - Jessica Medina
- Department of Chemistry, Universidad del Valle, Cali, Colombia
| | - Julián Sánchez
- Liga contra el Cáncer Seccional Bogotá, Bogotá, Colombia
| | - José Guio
- Liga contra el Cáncer Seccional Bogotá, Bogotá, Colombia
| | - Julien Wist
- Department of Chemistry, Universidad del Valle, Cali, Colombia
| | - Roland J. W. Meesters
- Department of Chemistry, Grupo de Investigación en Química Analítica y Bioanalítica (GABIO), Universidad de los Andes, Bogotá D.C., Colombia
| |
Collapse
|
29
|
Stephenson DJ, Hoeferlin LA, Chalfant CE. Lipidomics in translational research and the clinical significance of lipid-based biomarkers. Transl Res 2017; 189:13-29. [PMID: 28668521 PMCID: PMC5659874 DOI: 10.1016/j.trsl.2017.06.006] [Citation(s) in RCA: 92] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/09/2017] [Accepted: 06/08/2017] [Indexed: 02/08/2023]
Abstract
Lipidomics is a rapidly developing field of study that focuses on the identification and quantitation of various lipid species in the lipidome. Lipidomics has now emerged in the forefront of scientific research due to the importance of lipids in metabolism, cancer, and disease. Using both targeted and untargeted mass spectrometry as a tool for analysis, progress in the field has rapidly progressed in the last decade. Having the ability to assess these small molecules in vivo has led to better understanding of several lipid-driven mechanisms and the identification of lipid-based biomarkers in neurodegenerative disease, cancer, sepsis, wound healing, and pre-eclampsia. Biomarker identification and mechanistic understanding of specific lipid pathways linked to a disease's pathologies can form the foundation in the development of novel therapeutics in hopes of curing human disease.
Collapse
Affiliation(s)
- Daniel J Stephenson
- Department of Biochemistry and Molecular Biology, Virginia Commonwealth University (VCU), Richmond, Va
| | - L Alexis Hoeferlin
- Department of Biochemistry and Molecular Biology, Virginia Commonwealth University (VCU), Richmond, Va
| | - Charles E Chalfant
- Department of Biochemistry and Molecular Biology, Virginia Commonwealth University (VCU), Richmond, Va; Research Service, Hunter Holmes McGuire Veterans Administration Medical Center, Richmond, Va; VCU Massey Cancer Center, Cancer Cell Signaling Program, Virginia Commonwealth University, Richmond, Va; VCU Institute of Molecular Medicine, Richmond, Va; VCU Johnson Center for Critical Care and Pulmonary Research, Richmond, Va.
| |
Collapse
|
30
|
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
|
31
|
Antitumoral effects of γCdcPLI, a PLA 2 inhibitor from Crotalus durissus collilineatus via PI3K/Akt pathway on MDA-MB-231 breast cancer cell. Sci Rep 2017; 7:7077. [PMID: 28765552 PMCID: PMC5539153 DOI: 10.1038/s41598-017-07082-2] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2017] [Accepted: 06/22/2017] [Indexed: 12/23/2022] Open
Abstract
Phospholipases A2(PLA2s) overexpression is closely associated with the malignant potential of breast cancers. Here, we showed for the first the antitumoral effects of γCdcPLI, a PLA2 inhibitor from Crotalus durissus collilineatus via PI3K/Akt pathway on MDA-MB-231 cell. Firstly, γCdcPLI was more cytotoxic to MDA-MB-231 breast cancer cells than other cell lines (MCF-7, HeLa, PC3 and A549) and did not affect the viability of non-tumorigenic breast cell (MCF 10A). In addition, γCdcPLI induced modulation of important mediators of apoptosis pathways such as p53, MAPK-ERK, BIRC5 and MDM2. γCdcPLI decreased MDA-MB-231 adhesion, migration and invasion. Interestingly, the γCdcPLI also inhibited the adhesion and migration of endothelial cells and blocked angiogenesis by inhibiting tube formation by HUVECs in vitro and sprouting elongation on aortic ring assay ex vivo. Furthermore, γCdcPLI reduced the production of vascular endothelial growth factor (VEGF). γCdcPLI was also able to decrease PGE2 levels in MDA-MB-231 and inhibited gene and protein expression of the PI3K/Akt pathway. In conclusion, γCdcPLI showed in vitro antitumoral, antimestatatic and anti-angiogenic potential effects and could be an attractive approach for futures studies in cancer therapy.
Collapse
|
32
|
cPLA2α mediates TGF-β-induced epithelial-mesenchymal transition in breast cancer through PI3k/Akt signaling. Cell Death Dis 2017; 8:e2728. [PMID: 28383549 PMCID: PMC5477578 DOI: 10.1038/cddis.2017.152] [Citation(s) in RCA: 46] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2016] [Revised: 02/21/2017] [Accepted: 03/08/2017] [Indexed: 12/29/2022]
Abstract
A high incidence of tumor recurrence and metastasis has been reported in breast cancer patients; nevertheless, the underlying molecular mechanisms are largely unknown. Epithelial–mesenchymal transition (EMT), which is induced by transforming growth factor-β (TGF-β), has been implicated in tumorigenesis and breast cancer metastasis. EMT events are now directly associated with tumor metastasis, and this progress is dependent on the inflammatory microenvironment. Cytosolic phospholipase A2α (cPLA2α) has been shown to participate in a series of biological processes including inflammation and cancer development. However, the role and molecular mechanism of cPLA2α in breast cancer EMT and metastasis remain enigmatic. In this study, we found that cPLA2α was commonly overexpressed in most human breast cancer tissues and significantly correlated with a poor prognosis for human breast cancer. Functional studies demonstrated that cPLA2α overexpression was significantly associated with elevated migration and invasion in MDA-MB-231 and T47D cells. Conversely, reduced cPLA2α expression strongly attenuated metastasis and the EMT program of MDA-MB-231 cells. Further study found that knockdown of cPLA2α in MDA-MB-231 cells inhibited TGF-β-induced EMT through the PI3K/Akt signaling pathway. Animal experiments revealed that cPLA2α downregulation in MDA-MB-231 cells markedly restrained tumorigenesis and metastasis in vivo. This study indicates the potential role of cPLA2α in breast cancer metastasis and indicates that this molecule is a promising therapeutic target for breast cancer.
Collapse
|
33
|
Fouladi F, Steffen KJ, Mallik S. Enzyme-Responsive Liposomes for the Delivery of Anticancer Drugs. Bioconjug Chem 2017; 28:857-868. [PMID: 28201868 DOI: 10.1021/acs.bioconjchem.6b00736] [Citation(s) in RCA: 111] [Impact Index Per Article: 13.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/23/2023]
Abstract
Liposomes are nanocarriers that deliver the payloads at the target site, leading to therapeutic drug concentrations at the diseased site and reduced toxic effects in healthy tissues. Several approaches have been used to enhance the ability of the nanocarrier to target the specific tissues, including ligand-targeted liposomes and stimuli-responsive liposomes. Ligand-targeted liposomes exhibit higher uptake by the target tissue due to the targeting ligand attached to the surface, while the stimuli-responsive liposomes do not release their cargo unless they expose to an endogenous or exogenous stimulant at the target site. In this review, we mainly focus on the liposomes that are responsive to pathologically increased levels of enzymes at the target site. Enzyme-responsive liposomes release their cargo upon contact with the enzyme through several destabilization mechanisms: (1) structural perturbation in the lipid bilayer, (2) removal of a shielding polymer from the surface and increased cellular uptake, (3) cleavage of a lipopeptide or lipopolymer incorporated in the bilayer, and (4) activation of a prodrug in the liposomes.
Collapse
Affiliation(s)
- Farnaz Fouladi
- Department of Pharmaceutical Sciences, North Dakota State University , Fargo, North Dakota 58108, United States
| | - Kristine J Steffen
- Department of Pharmaceutical Sciences, North Dakota State University , Fargo, North Dakota 58108, United States
| | - Sanku Mallik
- Department of Pharmaceutical Sciences, North Dakota State University , Fargo, North Dakota 58108, United States
| |
Collapse
|
34
|
Fabrication of liposomal doxorubicin exhibiting ultrasensitivity against phospholipase A 2 for efficient pulmonary drug delivery to lung cancers. Int J Pharm 2017; 517:35-41. [DOI: 10.1016/j.ijpharm.2016.11.039] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2016] [Revised: 11/10/2016] [Accepted: 11/15/2016] [Indexed: 12/21/2022]
|
35
|
Activity-based targeting of secretory phospholipase A 2 enzymes: A fatty-acid-binding-protein assisted approach. Chem Phys Lipids 2016; 202:38-48. [PMID: 27894770 DOI: 10.1016/j.chemphyslip.2016.11.006] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2016] [Revised: 10/20/2016] [Accepted: 11/18/2016] [Indexed: 12/26/2022]
Abstract
Syntheses and enzymological characterization of fluorogenic substrate probes targeting secretory phospholipase A2 (sPLA2) for detection and quantitative assays are presented. Three fluorogenic phosphatidylcholine analogs PC-1, PC-2, and PC-3 each containing the duo of 7-mercapto-4-methyl-coumarin fluorophore and 2,4-dinitroanaline quencher on either tail were synthesized from (R)-3-amino-1,2-propanediol and R-(-)-2,2-dimethyl-1,3-dioxolane-4-methanol. These small reporter groups are advantageous in preserving natural membrane integrity. Phosphocholine was incorporated into the sn-3 position of the glycerol backbone. Acyl amino group at the sn-1 position in PC-1 and PC-2 is meant to block sPLA1. The sn-1 and sn-2 positions of the glycerol backbone in PC-1 have a quencher terminated 12-carbon chain and fluorophore terminated 11-carbon chain respectively. PC-2 has a quencher terminated 3-carbon chain at the sn-2 and chain terminating fluorescent reporter at the sn-1 positions. PC-3 resembles PC-1 except for an ester instead of amide at the sn-1 position, because of which it is more similar to natural phospholipids than PC-1. It was designed to elucidate the effect of replacing the ester group with amide by comparing its hydrolysis rate with that of PC-1. Design principles apply to synthesis of other labeled phospholipids. Enzymological characterization using bee-venom sPLA2 was performed by a fatty-acid-binding-protein fluorescence assay and by pH-Stat method in which the amount of fatty acid released by hydrolysis is given by the amount of base required to maintain a constant pH of 8.0. Hydrolytic activity toward PC-1 and PC-3 were each about 238±25μmol/mg/min and 537μmol/mg/min on unmodified phospholipid. Ester to amide change did not affect hydrolysis rates. Activity toward PC-2 was about 45-μmol/mg/min. PC-1 and PC-3 show potential for targeted real-time spectrophotometric assay of sPLA2.
Collapse
|
36
|
Yarla NS, Bishayee A, Sethi G, Reddanna P, Kalle AM, Dhananjaya BL, Dowluru KSVGK, Chintala R, Duddukuri GR. Targeting arachidonic acid pathway by natural products for cancer prevention and therapy. Semin Cancer Biol 2016; 40-41:48-81. [PMID: 26853158 DOI: 10.1016/j.semcancer.2016.02.001] [Citation(s) in RCA: 255] [Impact Index Per Article: 28.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2015] [Revised: 01/23/2016] [Accepted: 02/01/2016] [Indexed: 12/16/2022]
Abstract
Arachidonic acid (AA) pathway, a metabolic process, plays a key role in carcinogenesis. Hence, AA pathway metabolic enzymes phospholipase A2s (PLA2s), cyclooxygenases (COXs) and lipoxygenases (LOXs) and their metabolic products, such as prostaglandins and leukotrienes, have been considered novel preventive and therapeutic targets in cancer. Bioactive natural products are a good source for development of novel cancer preventive and therapeutic drugs, which have been widely used in clinical practice due to their safety profiles. AA pathway inhibitory natural products have been developed as chemopreventive and therapeutic agents against several cancers. Curcumin, resveratrol, apigenin, anthocyans, berberine, ellagic acid, eugenol, fisetin, ursolic acid, [6]-gingerol, guggulsteone, lycopene and genistein are well known cancer chemopreventive agents which act by targeting multiple pathways, including COX-2. Nordihydroguaiaretic acid and baicalein can be chemopreventive molecules against various cancers by inhibiting LOXs. Several PLA2s inhibitory natural products have been identified with chemopreventive and therapeutic potentials against various cancers. In this review, we critically discuss the possible utility of natural products as preventive and therapeutic agents against various oncologic diseases, including prostate, pancreatic, lung, skin, gastric, oral, blood, head and neck, colorectal, liver, cervical and breast cancers, by targeting AA pathway. Further, the current status of clinical studies evaluating AA pathway inhibitory natural products in cancer is reviewed. In addition, various emerging issues, including bioavailability, toxicity and explorability of combination therapy, for the development of AA pathway inhibitory natural products as chemopreventive and therapeutic agents against human malignancy are also discussed.
Collapse
Affiliation(s)
- Nagendra Sastry Yarla
- Department of Biochemisty/Bionformatics, Institute of Science, GITAM University, Rushikonda, Visakhapatnam 530 045, Adhra Pradesh, India
| | - Anupam Bishayee
- Department of Pharmaceutical Sciences, College of Pharmacy, Larkin Health Sciences Institute, 18301 N. Miami Avenue, Miami, FL 33169, USA.
| | - Gautam Sethi
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117600, Singapore; School of Biomedical Sciences, Curtin Health Innovation Research Institute, Biosciences Research Precinct, Curtin University, Western Australia 6009, Australia
| | - Pallu Reddanna
- Department of Animal Biology, School of Life Sciences, University of Hyderabad, Gachibowli, Hyderabad 500 046, Telagana, India
| | - Arunasree M Kalle
- Department of Animal Biology, School of Life Sciences, University of Hyderabad, Gachibowli, Hyderabad 500 046, Telagana, India; Department of Environmental Health Sciences, Laboratory of Human Environmental Epigenomes, Bloomberg School of Public Health, Johns Hopkins University, Baltimore, MD 21205, USA
| | - Bhadrapura Lakkappa Dhananjaya
- Toxinology/Toxicology and Drug Discovery Unit, Center for Emerging Technologies, Jain Global Campus, Jain University, Kanakapura Taluk, Ramanagara 562 112, Karnataka, India
| | - Kaladhar S V G K Dowluru
- Department of Biochemisty/Bionformatics, Institute of Science, GITAM University, Rushikonda, Visakhapatnam 530 045, Adhra Pradesh, India; Department of Microbiology and Bioinformatics, Bilaspur University, Bilaspur 495 001, Chhattisgarh, India
| | - Ramakrishna Chintala
- Department of Environmental Sciences, Institute of Science, GITAM University, Rushikonda, Visakhapatnam 530 045, Adhra Pradesh, India
| | - Govinda Rao Duddukuri
- Department of Biochemisty/Bionformatics, Institute of Science, GITAM University, Rushikonda, Visakhapatnam 530 045, Adhra Pradesh, India.
| |
Collapse
|
37
|
Pedada SR, Yarla NS, Tambade PJ, Dhananjaya BL, Bishayee A, Arunasree KM, Philip GH, Dharmapuri G, Aliev G, Putta S, Rangaiah G. Synthesis of new secretory phospholipase A2-inhibitory indole containing isoxazole derivatives as anti-inflammatory and anticancer agents. Eur J Med Chem 2016; 112:289-297. [PMID: 26907155 DOI: 10.1016/j.ejmech.2016.02.025] [Citation(s) in RCA: 53] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2015] [Revised: 02/07/2016] [Accepted: 02/08/2016] [Indexed: 11/25/2022]
Abstract
Secretory phospholipase A2 (sPLA2) is an important enzyme that plays a key role in various inflammatory diseases including cancer and its inhibitors have been developed as preventive or therapeutic agents. In the present study, a series of new indole containing isoxazole derivatives (10a-10o) is synthesized and evaluated for their sPLA2 inhibitory activities. All compounds (10a-10o) showed significant sPLA2 inhibition activities both in vitro and in vivo studies which is substantiated in in silico studies. Among all the tested compounds, 10o showed potent sPLA2 inhibition activity, that is comparable or more to ursolic acid (positive control). Further studies demonstrated that 10o showed in vitro antiproliferative activity when tested against MCF-7 breast and DU145 prostate cancer cells. Furthermore, compounds 10a-10o obeyed lipinsky's rule of 5 and suggesting druggable properties. The in vitro, in vivo and in silico results are encouraging and warrant pre-clinical studies to develop sPLA2-inhibitory compound 10o as novel therapeutic agent for various inflammatory disorders and several malignancies.
Collapse
Affiliation(s)
- Srinivasa Rao Pedada
- Centre for Research and Development, PRIST University, Thanjavur 613 403, Tamilnadu, India
| | - Nagendra Sastry Yarla
- Department of Biochemistry, Institute of Science, GITAM University, Visakhapatnam 530 045, Andhra Pradesh, India
| | - Pawan J Tambade
- Department of Chemistry, Arts, Commerce and Science College, Nandgaon 423 196, Maharashtra, India
| | - Bhadrapura Lakkappa Dhananjaya
- Toxinology/Toxicology and Drug Discovery Unit, Center for Emerging Technologies, Jain Global Campus, Jain University, Ramanagara 562 112, Karnataka, India
| | - Anupam Bishayee
- Department of Pharmaceutical Sciences, College of Pharmacy, Larkin Health Sciences Institute, Miami, FL 33169, USA
| | - Kalle M Arunasree
- Department of Animal Biology, School of Life Sciences, University of Hyderabad, Hyderabad 500 046, Telangana, India
| | - Gundala Harold Philip
- Department of Biotechnology, Sri Krishnadevaraya University, Anantapuramu 515 003, Andhra Pradesh, India
| | - Gangappa Dharmapuri
- Department of Animal Biology, School of Life Sciences, University of Hyderabad, Hyderabad 500 046, Telangana, India; Department of Biotechnology, Sri Krishnadevaraya University, Anantapuramu 515 003, Andhra Pradesh, India
| | - Gjumrach Aliev
- "GALLY" International Biomedical Research Consulting LLC, San Antonio, TX 78229, USA; School of Health Sciences and Healthcare Administration, University of Atlanta, E. Johns Crossing, #175, Johns Creek, GA 30097, USA; Institute of Physiologically Active Compounds, Russian Academy of Sciences, Chernogolovka 142432, Russia
| | - Swathi Putta
- Pharmacology Division, University College of Pharmaceutical Sciences, Andhra University, Visakhapatnam 530003, Andhra Pradesh, India
| | - Gururaja Rangaiah
- Department of Biochemistry, Sarada Vilas College, Mysore 570004, Karnataka, India.
| |
Collapse
|
38
|
ZHANG CHENGWEI, YU HAIPENG, XU HAIYAN, YANG LANLAN. Expression of secreted phospholipase A2-Group IIA correlates with prognosis of gastric adenocarcinoma. Oncol Lett 2015; 10:3050-3058. [PMID: 26722288 PMCID: PMC4665698 DOI: 10.3892/ol.2015.3736] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2014] [Accepted: 08/17/2015] [Indexed: 12/14/2022] Open
Abstract
The present study investigated the expression of secretory phospholipase A2-Group IIA (sPLA2-II) in gastric adenocarcinoma, in order to evaluate the correlation between sPLA2-II expression, and the clinicopathological features and prognosis of patients with gastric adenocarcinoma. Between January 2007 and April 2010, data were collected from 65 patients (44 males, 21 females; age range, 30-79 years; mean 66.7 ± 10.7 years). All patients exhibited a pathologically confirmed diagnosis of gastric adenocarcinoma. Endoscopic biopsy specimens of normal gastric mucosa from 11 of these patients were used as controls. Patients were subsequently followed-up at 3-month intervals, and survival data were recorded until April 2010. Expression of sPLA2-II in 65 gastric adenocarcinoma and 11 normal gastric mucosa specimens was evaluated via immunohistochemistry. A semi-quantitative method, consisting of evaluation of staining percentage and intensity, was utilized for immunohistochemical scoring, and the receiver operating characteristic curve method was applied to select a cut-off score for high and low sPLA2-II expression. The value of 8 was selected as the cut-off score, with maximum sensitivity and specificity. High sPLA2-II expression was observed in stage III/IV cases (83.3%; 40/48) and poorly differentiated cells (94.1%; 32/34), while sPLA2-II expression levels were observed to be significantly lower in stage I/II cases (52.9%; 9/17) and well and moderately differentiated cells (54.8%; 17/31; P=0.021 and P<0.001, respectively). There were no significant correlations observed between sPLA2-II expression and any other clinicopathological parameters, including gender, age, tumor diameter and Helicobacter pylori infection. Patients exhibiting low sPLA2-II expression experienced significantly improved overall survival (OS) and disease-free survival (DFS), compared with those exhibiting high sPLA2-II expression (P=0.043 and P=0.035, respectively). Multivariate analysis confirmed that high sPLA2-II expression may be an independent prognostic factor for OS [relative risk, 2.849; 95% confidence interval (CI), 1.088-7.459; P=0.033] and DFS (relative risk, 2.735; 95% CI, 1.104-6.776; P=0.030) in gastric adenocarcinoma. Therefore, sPLA2-II may be correlated with the histogenesis of gastric adenocarcinoma, and increased sPLA2-II expression may be an indicator of poor prognosis.
Collapse
Affiliation(s)
- CHENGWEI ZHANG
- Department of Anesthesiology, The Second Hospital of Jilin University, Changchun, Jilin 130041, P.R. China
| | - HAIPENG YU
- Department of Interventional Therapy, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center of Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin 300060, P.R. China
| | - HAIYAN XU
- Department of Physiology, College of Basic Medical Sciences, Jilin University, Changchun, Jilin 130021, P.R. China
| | - LANLAN YANG
- Department of Biliary and Pancreatic Internal Medicine, The Second Hospital of Jilin University, Changchun, Jilin 130041, P.R. China
| |
Collapse
|
39
|
Hansen AH, Mouritsen OG, Arouri A. Enzymatic action of phospholipase A2 on liposomal drug delivery systems. Int J Pharm 2015; 491:49-57. [DOI: 10.1016/j.ijpharm.2015.06.005] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2015] [Revised: 06/03/2015] [Accepted: 06/04/2015] [Indexed: 12/17/2022]
|
40
|
Deletion of phospholipase A2 group IVc induces apoptosis in rat mammary tumour cells by the nuclear factor-κB/lipocalin 2 pathway. Biochem J 2015; 469:315-24. [PMID: 26013918 DOI: 10.1042/bj20150064] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2015] [Accepted: 05/27/2015] [Indexed: 12/15/2022]
Abstract
Although some forms of phospholipase A2, the initiator of the arachidonic acid cascade, contribute to carcinogenesis in many organs, the contribution of phospholipase A2 group IVc (Pla2g4c) remains to be clarified and the function of the enzyme in cancer development is unknown. The Hirosaki hairless rat (HHR), a mutant rat strain with autosomal recessive inheritance, derived spontaneously from the Sprague-Dawley rat (SDR). The HHRs showed a lower incidence and much smaller volume of mammary tumours induced by 7,12-dimethylbenz[a]anthracene, and a markedly increased number of TUNEL (terminal deoxynucleotidyltransferase-mediated dUTP nick-end labelling)-positive apoptotic cells was detected. Array comparative genomic hybridization and PCR analyses revealed the deletion of 50-kb genomic DNA on 1q21, including Pla2g4c, in HHRs. The Pla2g4c gene was expressed in the ductal carcinoma cells and myoepithelial cells in SDRs, but not in HHRs. The direct involvement of Pla2g4c in the prevention of cell death was demonstrated through the inhibition of its expression in rat mammary tumour RMT-1 cells using siRNA. This treatment also induced expression of lipocalin 2 (Lcn2) and other NF-κB (nuclear factor κB)-related genes. siRNA-induced apoptosis was inhibited by Lcn2 repression or NF-κB inhibitors. This is the first report on Pla2g4c gene-deficient rats and their low susceptibility to mammary carcinogenesis by enhancing NF-κB/Lcn2-induced apoptosis.
Collapse
|
41
|
Arouri A, Trojnar J, Schmidt S, Hansen AH, Mollenhauer J, Mouritsen OG. Development of a cell-based bioassay for phospholipase A2-triggered liposomal drug release. PLoS One 2015; 10:e0125508. [PMID: 25945937 PMCID: PMC4422686 DOI: 10.1371/journal.pone.0125508] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2015] [Accepted: 03/21/2015] [Indexed: 01/16/2023] Open
Abstract
The feasibility of exploiting secretory phospholipase A2 (sPLA2) enzymes, which are overexpressed in tumors, to activate drug release from liposomes precisely at the tumor site has been demonstrated before. Although the efficacy of the developed formulations was evaluated using in vitro and in vivo models, the pattern of sPLA2-assisted drug release is unknown due to the lack of a suitable bio-relevant model. We report here on the development of a novel bioluminescence living-cell-based luciferase assay for the monitoring of sPLA2-triggered release of luciferin from liposomes. To this end, we engineered breast cancer cells to produce both luciferase and sPLA2 enzymes, where the latter is secreted to the extracellular medium. We report on setting up a robust and reproducible bioassay for testing sPLA2-sensitive, luciferin remote-loaded liposomal formulations, using 1,2-distearoyl-sn-glycero-3-phosphatidylcholine/1,2-distearoyl-sn-glycero-3-phosphatidylglycerol (DSPC/DSPG) 7:3 and DSPC/DSPG/cholesterol 4:3:3 as initial test systems. Upon their addition to the cells, the liposomes were degraded almost instantaneously by sPLA2 releasing the encapsulated luciferin, which provided readout from the luciferase-expressing cells. Cholesterol enhanced the integrity of the formulation without affecting its susceptibility to sPLA2. PEGylation of the liposomes only moderately broadened the release profile of luciferin. The provided bioassay represents a useful tool for monitoring active drug release in situ in real time as well as for testing and optimizing of sPLA2-sensitive lipid formulations. In addition, the bioassay will pave the way for future in-depth in vitro and in vivo studies.
Collapse
Affiliation(s)
- Ahmad Arouri
- MEMPHYS-Center for Biomembrane Physics, Department of Physics, Chemistry, and Pharmacy, University of Southern Denmark, Odense, Denmark
- Lundbeckfonden Center of Excellence NanoCAN, Institute for Molecular Medicine, University of Southern Denmark, Odense, Denmark
- * E-mail:
| | - Jakub Trojnar
- Lundbeckfonden Center of Excellence NanoCAN, Institute for Molecular Medicine, University of Southern Denmark, Odense, Denmark
- Molecular Oncology Group, Institute for Molecular Medicine, University of Southern Denmark, Odense, Denmark
| | - Steffen Schmidt
- Lundbeckfonden Center of Excellence NanoCAN, Institute for Molecular Medicine, University of Southern Denmark, Odense, Denmark
- Molecular Oncology Group, Institute for Molecular Medicine, University of Southern Denmark, Odense, Denmark
| | - Anders H. Hansen
- MEMPHYS-Center for Biomembrane Physics, Department of Physics, Chemistry, and Pharmacy, University of Southern Denmark, Odense, Denmark
- Lundbeckfonden Center of Excellence NanoCAN, Institute for Molecular Medicine, University of Southern Denmark, Odense, Denmark
| | - Jan Mollenhauer
- Lundbeckfonden Center of Excellence NanoCAN, Institute for Molecular Medicine, University of Southern Denmark, Odense, Denmark
- Molecular Oncology Group, Institute for Molecular Medicine, University of Southern Denmark, Odense, Denmark
| | - Ole G. Mouritsen
- MEMPHYS-Center for Biomembrane Physics, Department of Physics, Chemistry, and Pharmacy, University of Southern Denmark, Odense, Denmark
- Lundbeckfonden Center of Excellence NanoCAN, Institute for Molecular Medicine, University of Southern Denmark, Odense, Denmark
| |
Collapse
|
42
|
Pace-Asciak CR. Pathophysiology of the hepoxilins. Biochim Biophys Acta Mol Cell Biol Lipids 2015; 1851:383-96. [DOI: 10.1016/j.bbalip.2014.09.007] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2014] [Revised: 09/06/2014] [Accepted: 09/10/2014] [Indexed: 10/24/2022]
|
43
|
Brglez V, Lambeau G, Petan T. Secreted phospholipases A2 in cancer: Diverse mechanisms of action. Biochimie 2014; 107 Pt A:114-23. [DOI: 10.1016/j.biochi.2014.09.023] [Citation(s) in RCA: 45] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2014] [Accepted: 09/25/2014] [Indexed: 12/24/2022]
|
44
|
Halama A. Metabolomics in cell culture--a strategy to study crucial metabolic pathways in cancer development and the response to treatment. Arch Biochem Biophys 2014; 564:100-9. [PMID: 25218088 DOI: 10.1016/j.abb.2014.09.002] [Citation(s) in RCA: 66] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2014] [Revised: 09/01/2014] [Accepted: 09/02/2014] [Indexed: 12/11/2022]
Abstract
Metabolomics is a comprehensive tool for monitoring processes within biological systems. Thus, metabolomics may be widely applied to the determination of diagnostic biomarkers for certain diseases or treatment outcomes. There is significant potential for metabolomics to be implemented in cancer research because cancer may modify metabolic pathways in the whole organism. However, not all biological questions can be answered solely by the examination of small molecule composition in biofluids; in particular, the study of cellular processes or preclinical drug testing requires ex vivo models. The major objective of this review was to summarise the current achievement in the field of metabolomics in cancer cell culture-focusing on the metabolic pathways regulated in different cancer cell lines-and progress that has been made in the area of drug screening and development by the implementation of metabolomics in cell lines.
Collapse
Affiliation(s)
- Anna Halama
- Department of Physiology and Biophysics, Weill Cornell Medical College-Qatar, Doha, Qatar.
| |
Collapse
|
45
|
Peptidophospholipids: synthesis, phospholipase A2 catalyzed hydrolysis, and application to development of phospholipid prodrugs. Chem Phys Lipids 2014; 183:110-6. [PMID: 24905766 DOI: 10.1016/j.chemphyslip.2014.06.001] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2014] [Revised: 05/29/2014] [Accepted: 06/01/2014] [Indexed: 12/16/2022]
Abstract
New phospholipid analogues incorporating sn-2-peptide substituents have been prepared to probe the fundamental structural requirements for phospholipase A2 catalyzed hydrolysis of PLA2-directed synthetic substrates. Two structurally different antiviral oligopeptides with C-terminal glycine were introduced separately at the sn-2-carboxylic ester position of phospholipids to assess the role of the α-methylene group adjacent to the ester carbonyl in allowing hydrolytic cleavage by the enzyme. The oligopeptide-carrying phospholipid derivatives were readily incorporated into mixed micelles consisting of natural phospholipid (dipalmitoyl phosphatidylcholine, DPPC) and Triton X-100 as surfactant. Hydrolytic cleavage of the synthetic peptidophospholipids by the phospholipase A2 occurred slower, but within the same order of magnitude as the natural substrate alone. The results provide useful information toward better understanding the mechanism of action of the enzyme, and to improve the design and synthesis of phospholipid prodrugs targeted at secretory PLA2 enzymes.
Collapse
|
46
|
Quach ND, Arnold RD, Cummings BS. Secretory phospholipase A2 enzymes as pharmacological targets for treatment of disease. Biochem Pharmacol 2014; 90:338-48. [PMID: 24907600 DOI: 10.1016/j.bcp.2014.05.022] [Citation(s) in RCA: 114] [Impact Index Per Article: 10.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2014] [Revised: 05/28/2014] [Accepted: 05/28/2014] [Indexed: 02/03/2023]
Abstract
Phospholipase A2 (PLA2) cleave phospholipids preferentially at the sn-2 position, liberating free fatty acids and lysophospholipids. They are classified into six main groups based on size, location, function, substrate specificity and calcium requirement. These classes include secretory PLA2 (sPLA2), cytosolic (cPLA2), Ca(2+)-independent (iPLA2), platelet activating factor acetylhydrolases (PAF-AH), lysosomal PLA2 (LyPLA2) and adipose specific PLA2 (AdPLA2). It is hypothesized that PLA2 can serve as pharmacological targets for the therapeutic treatment of several diseases, including cardiovascular diseases, atherosclerosis, immune disorders and cancer. Special emphasis has been placed on inhibitors of sPLA2 isoforms as pharmacological moieties, mostly due to the fact that these enzymes are activated during inflammatory events and because their expression is increased in several diseases. This review focuses on understanding how sPLA2 isoform expression is altered during disease progression and the possible therapeutic interventions to specifically target sPLA2 isoforms, including new approaches using nano-particulate-based strategies.
Collapse
Affiliation(s)
- Nhat D Quach
- Pharmaceutical and Biomedical Sciences, University of Georgia, Athens, GA 30602, United States
| | - Robert D Arnold
- Department of Drug Discovery & Development, Harrison School of Pharmacy, Auburn University, Auburn, AL 36849-5503, United States
| | - Brian S Cummings
- Pharmaceutical and Biomedical Sciences, University of Georgia, Athens, GA 30602, United States.
| |
Collapse
|
47
|
Wang T, Goodman MA, McGough RL, Weiss KR, Rao UNM. Immunohistochemical Analysis of Expressions of RB1, CDK4, HSP90, cPLA2G4A, and CHMP2B Is Helpful in Distinction between Myxofibrosarcoma and Myxoid Liposarcoma. Int J Surg Pathol 2014; 22:589-99. [DOI: 10.1177/1066896914532539] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
The role and diagnostic efficacy of gene and protein products RB1, CDK4, CHMP2B, HSP90, and cPLA2G4A, all previously shown to be involved in tumor genesis and cell proliferation, were examined by immunohistochemical techniques in 32 cases of myxofibrosarcomas and 29 myxoid liposarcomas (all diagnosis had been confirmed by fluorescence in situ hybridization). HSP90 demonstrated strong nuclear and cytoplasmic positivity in all myxoid liposarcoma cases, while only 4 myxofibrosarcomas showed scattered HSP90 positivity. All but 4 cases of myxofibrosarcoma displayed strong positivity for cPLA2G4A, while only 2 myxoid liposarcoma cases were cPLA2G4A positive and both were CHMP2B negative. Overexpression of both cPLA2G4A and CHMP2B also suggested higher tumor grade. In conclusion, HSP90 and cPLA2G4A immunohistochemical stains are useful markers to distinguish myxofibrosarcoma from myxoid liposarcoma.
Collapse
Affiliation(s)
- Tao Wang
- University of Pittsburgh Medical Center–Shadyside Campus, Pittsburgh, PA, USA
| | - Mark A. Goodman
- University of Pittsburgh Medical Center–Shadyside Campus, Pittsburgh, PA, USA
| | - Richard L. McGough
- University of Pittsburgh Medical Center–Shadyside Campus, Pittsburgh, PA, USA
| | - Kurt R. Weiss
- University of Pittsburgh Medical Center–Shadyside Campus, Pittsburgh, PA, USA
| | - Uma N. M. Rao
- University of Pittsburgh Medical Center–Shadyside Campus, Pittsburgh, PA, USA
| |
Collapse
|
48
|
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
|
49
|
Brglez V, Pucer A, Pungerčar J, Lambeau G, Petan T. Secreted phospholipases A₂are differentially expressed and epigenetically silenced in human breast cancer cells. Biochem Biophys Res Commun 2014; 445:230-5. [PMID: 24508801 DOI: 10.1016/j.bbrc.2014.01.182] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2014] [Accepted: 01/29/2014] [Indexed: 02/06/2023]
Abstract
Secreted phospholipases A2 (sPLA2s) have recently been associated with several cancers, but their role in breast cancer is unknown. Here we demonstrate that mRNA expression of group IIA, III and X sPLA2s differs both in vivo in tumour biopsies and in breast cancer cells in vitro. Their expression is differentially regulated by DNA methylation and histone acetylation and, significantly, all three genes are silenced in aggressive triple negative cells due to both mechanisms. The transcription start site promoter region and the upstream CpG islands, exclusive to the group X sPLA2 gene, have variable roles in the regulation of sPLA2 expression. Our results suggest that the differential expression of hGIIA, hGIII and hGX sPLA2s in breast cancer cells is a consequence of various degrees of epigenetic silencing due to DNA hypermethylation and histone deacetylation.
Collapse
Affiliation(s)
- Vesna Brglez
- Department of Molecular and Biomedical Sciences, Jožef Stefan Institute, Ljubljana, Slovenia
| | - Anja Pucer
- Department of Molecular and Biomedical Sciences, Jožef Stefan Institute, Ljubljana, Slovenia
| | - Jože Pungerčar
- Department of Molecular and Biomedical Sciences, Jožef Stefan Institute, Ljubljana, Slovenia
| | - Gérard Lambeau
- Institut de Pharmacologie Moléculaire et Cellulaire, CNRS et Université de Nice Sophia Antipolis, UMR 6097, Sophia Antipolis, Valbonne, France
| | - Toni Petan
- Department of Molecular and Biomedical Sciences, Jožef Stefan Institute, Ljubljana, Slovenia.
| |
Collapse
|
50
|
Arouri A, Hansen AH, Rasmussen TE, Mouritsen OG. Lipases, liposomes and lipid-prodrugs. Curr Opin Colloid Interface Sci 2013. [DOI: 10.1016/j.cocis.2013.06.001] [Citation(s) in RCA: 47] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
|