1
|
Thim EA, Fox T, Deering T, Vass LR, Sheybani ND, Kester M, Price RJ. Solid tumor treatment via augmentation of bioactive C6 ceramide levels with thermally ablative focused ultrasound. Drug Deliv Transl Res 2023; 13:3145-3153. [PMID: 37335416 PMCID: PMC11423265 DOI: 10.1007/s13346-023-01377-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/05/2023] [Indexed: 06/21/2023]
Abstract
Sparse scan partial thermal ablation (TA) with focused ultrasound (FUS) may be deployed to treat solid tumors and increase delivery of systemically administered therapeutics. Furthermore, C6-ceramide-loaded nanoliposomes (CNLs), which rely upon the enhanced-permeation and retention (EPR) effect for delivery, have shown promise for treating solid tumors and are being tested in clinical trials. Here, our objective was to determine whether CNLs synergize with TA in the control of 4T1 breast tumors. CNL monotherapy of 4T1 tumors yielded significant intratumoral bioactive C6 accumulation by the EPR effect, but tumor growth was not controlled. TA increased bioactive C6 accumulation by ~ 12.5-fold over the EPR effect. In addition, TA + CNL caused shifts in long-chain to very-long-chain ceramide ratios (i.e., C16/24 and C18/C24) that could potentially contribute to tumor control. Nonetheless, these changes in intratumoral ceramide levels were still insufficient to confer tumor growth control beyond that achieved when combining with TA with control "ghost" nanoliposomes (GNL). While this lack of synergy could be due to increased "pro-tumor" sphingosine-1-phosphate (S1P) levels, this is unlikely because S1P levels exhibited only a moderate and statistically insignificant increase with TA + CNL. In vitro studies showed that 4T1 cells are highly resistant to C6, offering the most likely explanation for the inability of TA to synergize with CNL. Thus, while our results show that sparse scan TA is a powerful approach for markedly enhancing CNL delivery and generating "anti-tumor" shifts in long-chain to very-long-chain ceramide ratios, resistance of the tumor to C6 can still be a rate-limiting factor for some solid tumor types.
Collapse
Affiliation(s)
- E Andrew Thim
- Department of Biomedical Engineering, University of Virginia, Box 800759, Charlottesville, VA, 22908, USA
| | - Todd Fox
- Department of Pharmacology, University of Virginia, Charlottesville, VA, 22908, USA
| | - Tye Deering
- Department of Pharmacology, University of Virginia, Charlottesville, VA, 22908, USA
| | - Luke R Vass
- Department of Pathology, University of Virginia, Charlottesville, VA, 22908, USA
| | - Natasha D Sheybani
- Department of Biomedical Engineering, University of Virginia, Box 800759, Charlottesville, VA, 22908, USA
| | - Mark Kester
- Department of Biomedical Engineering, University of Virginia, Box 800759, Charlottesville, VA, 22908, USA
- Department of Pharmacology, University of Virginia, Charlottesville, VA, 22908, USA
| | - Richard J Price
- Department of Biomedical Engineering, University of Virginia, Box 800759, Charlottesville, VA, 22908, USA.
| |
Collapse
|
2
|
Zhu H, Chen HJ, Wen HY, Wang ZG, Liu SL. Engineered Lipidic Nanomaterials Inspired by Sphingomyelin Metabolism for Cancer Therapy. Molecules 2023; 28:5366. [PMID: 37513239 PMCID: PMC10383197 DOI: 10.3390/molecules28145366] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2023] [Revised: 07/08/2023] [Accepted: 07/10/2023] [Indexed: 07/30/2023] Open
Abstract
Sphingomyelin (SM) and its metabolites are crucial regulators of tumor cell growth, differentiation, senescence, and programmed cell death. With the rise in lipid-based nanomaterials, engineered lipidic nanomaterials inspired by SM metabolism, corresponding lipid targeting, and signaling activation have made fascinating advances in cancer therapeutic processes. In this review, we first described the specific pathways of SM metabolism and the roles of their associated bioactive molecules in mediating cell survival or death. We next summarized the advantages and specific applications of SM metabolism-based lipidic nanomaterials in specific cancer therapies. Finally, we discussed the challenges and perspectives of this emerging and promising SM metabolism-based nanomaterials research area.
Collapse
Affiliation(s)
- Han Zhu
- State Key Laboratory of Medicinal Chemical Biology, Tianjin Key Laboratory of Biosensing and Molecular Recognition, Research Center for Analytical Sciences, College of Chemistry, and School of Medicine, Nankai University, Tianjin 300071, China
| | - Hua-Jie Chen
- Engineering Research Center of Nano-Geomaterials of Ministry of Education, Faculty of Materials Science and Chemistry, China University of Geosciences, Wuhan 430074, China
| | - Hai-Yan Wen
- Engineering Research Center of Nano-Geomaterials of Ministry of Education, Faculty of Materials Science and Chemistry, China University of Geosciences, Wuhan 430074, China
| | - Zhi-Gang Wang
- State Key Laboratory of Medicinal Chemical Biology, Tianjin Key Laboratory of Biosensing and Molecular Recognition, Research Center for Analytical Sciences, College of Chemistry, and School of Medicine, Nankai University, Tianjin 300071, China
| | - Shu-Lin Liu
- State Key Laboratory of Medicinal Chemical Biology, Tianjin Key Laboratory of Biosensing and Molecular Recognition, Research Center for Analytical Sciences, College of Chemistry, and School of Medicine, Nankai University, Tianjin 300071, China
- Engineering Research Center of Nano-Geomaterials of Ministry of Education, Faculty of Materials Science and Chemistry, China University of Geosciences, Wuhan 430074, China
| |
Collapse
|
3
|
Singh AK, Malviya R, Prajapati B, Singh S, Yadav D, Kumar A. Nanotechnology-Aided Advancement in Combating the Cancer Metastasis. Pharmaceuticals (Basel) 2023; 16:899. [PMID: 37375846 PMCID: PMC10304141 DOI: 10.3390/ph16060899] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2023] [Revised: 05/28/2023] [Accepted: 06/14/2023] [Indexed: 06/29/2023] Open
Abstract
Modern medicine has been working to find a cure for cancer for almost a century, but thus far, they have not been very successful. Although cancer treatment has come a long way, more work has to be carried out to boost specificity and reduce systemic toxicity. The diagnostic industry is on the cusp of a technological revolution, and early diagnosis is essential for improving prognostic outlook and patient quality of life. In recent years, nanotechnology's use has expanded, demonstrating its efficacy in enhancing fields such as cancer treatment, radiation therapy, diagnostics, and imaging. Applications for nanomaterials are diverse, ranging from enhanced radiation adjuvants to more sensitive early detection instruments. Cancer, particularly when it has spread beyond the original site of cancer, is notoriously tough to combat. Many people die from metastatic cancer, which is why it remains a huge issue. Cancer cells go through a sequence of events known as the "metastatic cascade" throughout metastasis, which may be used to build anti-metastatic therapeutic techniques. Conventional treatments and diagnostics for metastasis have their drawbacks and hurdles that must be overcome. In this contribution, we explore in-depth the potential benefits that nanotechnology-aided methods might offer to the detection and treatment of metastatic illness, either alone or in conjunction with currently available conventional procedures. Anti-metastatic drugs, which can prevent or slow the spread of cancer throughout the body, can be more precisely targeted and developed with the help of nanotechnology. Furthermore, we talk about how nanotechnology is being applied to the treatment of patients with cancer metastases.
Collapse
Affiliation(s)
- Arun Kumar Singh
- Department of Pharmacy, School of Medical and Allied Sciences, Galgotias University, Greater Noida 203201, India; (A.K.S.); (D.Y.)
| | - Rishabha Malviya
- Department of Pharmacy, School of Medical and Allied Sciences, Galgotias University, Greater Noida 203201, India; (A.K.S.); (D.Y.)
| | - Bhupendra Prajapati
- Shree S. K. Patel College of Pharmaceutical Education and Research, Ganpat University, Kherva 384012, India
| | - Sudarshan Singh
- Department of Pharmaceutical Sciences, Faculty of Pharmacy, Chiang Mai University, Chiang Mai 50200, Thailand
| | - Deepika Yadav
- Department of Pharmacy, School of Medical and Allied Sciences, Galgotias University, Greater Noida 203201, India; (A.K.S.); (D.Y.)
| | - Arvind Kumar
- Chandigarh Engineering College, Jhanjeri, Mohali 140307, India;
| |
Collapse
|
4
|
Corsetto PA, Zava S, Rizzo AM, Colombo I. The Critical Impact of Sphingolipid Metabolism in Breast Cancer Progression and Drug Response. Int J Mol Sci 2023; 24:ijms24032107. [PMID: 36768427 PMCID: PMC9916652 DOI: 10.3390/ijms24032107] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2022] [Revised: 01/11/2023] [Accepted: 01/17/2023] [Indexed: 01/25/2023] Open
Abstract
Breast cancer is the second leading cause of cancer-related death in women in the world, and its management includes a combination of surgery, radiation therapy, chemotherapy, and immunotherapy, whose effectiveness depends largely, but not exclusively, on the molecular subtype (Luminal A, Luminal B, HER2+ and Triple Negative). All breast cancer subtypes are accompanied by peculiar and substantial changes in sphingolipid metabolism. Alterations in sphingolipid metabolite levels, such as ceramides, dihydroceramide, sphingosine, sphingosine-1-phosphate, and sphingomyelin, as well as in their biosynthetic and catabolic enzymatic pathways, have emerged as molecular mechanisms by which breast cancer cells grow, respond to or escape therapeutic interventions and could take on diagnostic and prognostic value. In this review, we summarize the current landscape around two main themes: 1. sphingolipid metabolites, enzymes and transport proteins that have been found dysregulated in human breast cancer cells and/or tissues; 2. sphingolipid-driven mechanisms that allow breast cancer cells to respond to or evade therapies. Having a complete picture of the impact of the sphingolipid metabolism in the development and progression of breast cancer may provide an effective means to improve and personalize treatments and reduce associated drug resistance.
Collapse
|
5
|
Linzer RW, Guida DL, Aminov J, Snider JM, Khalife G, Buyukbayraktar AB, Alhaddad C, Resnick AE, Wang P, Pan CH, Allopenna JJ, Clarke CJ. Dihydroceramide desaturase 1 (DES1) promotes anchorage-independent survival downstream of HER2-driven glucose uptake and metabolism. FASEB J 2022; 36:e22558. [PMID: 36165222 PMCID: PMC9597949 DOI: 10.1096/fj.202200748r] [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: 05/19/2022] [Revised: 08/03/2022] [Accepted: 09/07/2022] [Indexed: 11/11/2022]
Abstract
Oncogenic reprogramming of cellular metabolism is a hallmark of many cancers, but our mechanistic understanding of how such dysregulation is linked to tumor behavior remains poor. In this study, we have identified dihydroceramide desaturase (DES1)-which catalyzes the last step in de novo sphingolipid synthesis-as necessary for the acquisition of anchorage-independent survival (AIS), a key cancer enabling biology, and establish DES1 as a downstream effector of HER2-driven glucose uptake and metabolism. We further show that DES1 is sufficient to drive AIS and in vitro tumorigenicity and that increased DES1 levels-found in a third of HER2+ breast cancers-are associated with worse survival outcomes. Taken together, our findings reveal a novel pro-tumor role for DES1 as a transducer of HER2-driven glucose metabolic signals and provide evidence that targeting DES1 is an effective approach for overcoming AIS. Results further suggest that DES1 may have utility as a biomarker of aggressive and metastasis-prone HER2+ breast cancer.
Collapse
Affiliation(s)
- Ryan W Linzer
- Department of Medicine and the Cancer Center, Stony Brook University, Stony Brook, NY, USA
| | - Danielle L Guida
- Department of Medicine and the Cancer Center, Stony Brook University, Stony Brook, NY, USA
| | - Jonathan Aminov
- Department of Medicine and the Cancer Center, Stony Brook University, Stony Brook, NY, USA
| | - Justin M Snider
- Department of Medicine and the Cancer Center, Stony Brook University, Stony Brook, NY, USA
| | - Gabrielle Khalife
- Department of Medicine and the Cancer Center, Stony Brook University, Stony Brook, NY, USA
| | - A Burak Buyukbayraktar
- Department of Medicine and the Cancer Center, Stony Brook University, Stony Brook, NY, USA
| | - Charbel Alhaddad
- Department of Medicine and the Cancer Center, Stony Brook University, Stony Brook, NY, USA
| | - Andrew E Resnick
- Department of Medicine and the Cancer Center, Stony Brook University, Stony Brook, NY, USA
| | - Pule Wang
- Department of Medicine and the Cancer Center, Stony Brook University, Stony Brook, NY, USA
| | - Chun-Hao Pan
- Department of Medicine and the Cancer Center, Stony Brook University, Stony Brook, NY, USA
| | - Janet J Allopenna
- Department of Medicine and the Cancer Center, Stony Brook University, Stony Brook, NY, USA
| | - Christopher J Clarke
- Department of Medicine and the Cancer Center, Stony Brook University, Stony Brook, NY, USA
| |
Collapse
|
6
|
Salata GC, Lopes LB. Phosphatidylcholine-Based Nanoemulsions for Paclitaxel and a P-Glycoprotein Inhibitor Delivery and Breast Cancer Intraductal Treatment. Pharmaceuticals (Basel) 2022; 15:ph15091110. [PMID: 36145331 PMCID: PMC9503599 DOI: 10.3390/ph15091110] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2022] [Revised: 08/26/2022] [Accepted: 08/31/2022] [Indexed: 01/09/2023] Open
Abstract
In this study, incorporation of the cytotoxic agent paclitaxel and the P-glycoprotein inhibitor elacridar in hyaluronic acid (HA)-modified nanoemulsions was studied for intraductal delivery and breast cancer localized treatment. To improve cytotoxicity, we investigated the incorporation of perillyl alcohol or tributyrin as components of the nanoemulsion oil phase. The nanoemulsions presented size <180 nm and negative zeta potential. Both tributyrin and perillyl alcohol increased nanoemulsion cytotoxicity in MCF-7 cells, but not in MDA-MB-231. However, perillyl alcohol reduced nanoemulsion stability in the presence of the drugs. Concomitant incorporation of paclitaxel and elacridar in HA- and tributyrin-containing nanoemulsions (PE-NETri) increased cytotoxicity and reduced IC50 by 1.6 to 3-fold in MCF-7 and MDA-MB-231 cells compared to the nanoemulsion containing only paclitaxel (P-NE). This nanoemulsion also produced a 3.3-fold reduction in the viability of MDA-MB-231 spheroids. Elacridar incorporated in the nanoemulsion was capable of inhibiting P-glycoprotein in membranes. In vivo intraductal administration of the NE containing HA resulted in a three-fold higher retention of a fluorescent marker compared to a solution or nanoemulsion without HA, demonstrating the importance of HA. The nanoemulsion produced no histological changes in the mammary tissue. These results support the potential applicability of the nanoemulsion for local breast cancer management.
Collapse
|
7
|
Mondal K, Porter H, Cole J, Pandya HK, Basu SK, Khanam S, Chiu CY, Shah V, Stephenson DJ, Chalfant CE, Mandal N. Hydroxychloroquine Causes Early Inner Retinal Toxicity and Affects Autophagosome-Lysosomal Pathway and Sphingolipid Metabolism in the Retina. Mol Neurobiol 2022; 59:3873-3887. [PMID: 35426574 PMCID: PMC10259418 DOI: 10.1007/s12035-022-02825-3] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2022] [Accepted: 04/01/2022] [Indexed: 01/21/2023]
Abstract
Hydroxychloroquine (HCQ) is an anti-malarial drug but also widely used to treat autoimmune diseases like arthritis and lupus. Although there have been multiple reports of the adverse effect of prolonged HCQ usage on the outer retina, leading to bull's-eye maculopathy, the effect of HCQ toxicity on the inner retina as well as on overall visual functions has not been explored in detail. Furthermore, lack of an established animal model of HCQ toxicity hinders our understanding of the underlying molecular mechanisms. Here, using a small clinical study, we confirmed the effect of HCQ toxicity on the inner retina, in particular the reduction in central inner retinal thickness, and established a mouse model of chronic HCQ toxicity that recapitulates the effects observed in human retina. Using the mouse model, we demonstrated that chronic HCQ toxicity results in loss of inner retinal neurons and retinal ganglion cells (RGC) and compromises visual functions. We further established that HCQ treatment prevents autophagosome-lysosome fusion and alters the sphingolipid homeostasis in mouse retina. Our results affirm the notion that HCQ treatment causes early damage to the inner retina and affects visual functions before leading to characteristic toxicity in the macular region of the outer retina, 'bull's-eye maculopathy.' We also provide insights into the underlying molecular mechanisms of HCQ retinal toxicity that may involve autophagy-lysosomal defects and alterations in sphingolipid metabolism.
Collapse
Affiliation(s)
- Koushik Mondal
- Department of Ophthalmology, Hamilton Eye Institute, The University of Tennessee Health Science Center, Memphis, TN, 38163, USA
| | - Hunter Porter
- Department of Ophthalmology, Dean McGee Eye Institute, The University of Oklahoma Health Sciences Center, Oklahoma City, OK-73104, USA
| | - Jerome Cole
- Department of Ophthalmology, Hamilton Eye Institute, The University of Tennessee Health Science Center, Memphis, TN, 38163, USA
| | - Hemang K Pandya
- Department of Ophthalmology, Dean McGee Eye Institute, The University of Oklahoma Health Sciences Center, Oklahoma City, OK-73104, USA
| | - Sandip K Basu
- Department of Ophthalmology, Hamilton Eye Institute, The University of Tennessee Health Science Center, Memphis, TN, 38163, USA
| | - Sufiya Khanam
- Department of Ophthalmology, Hamilton Eye Institute, The University of Tennessee Health Science Center, Memphis, TN, 38163, USA
| | - Chi-Yang Chiu
- Division of Biostatistics, Department of Preventive Medicine, University of Tennessee Health Science Center, Memphis, TN, 38163, USA
| | - Vinay Shah
- Department of Ophthalmology, Dean McGee Eye Institute, The University of Oklahoma Health Sciences Center, Oklahoma City, OK-73104, USA
| | - Daniel J Stephenson
- Department of Cell Biology, Microbiology and Molecular Biology, University of South Florida, Tampa, FL, 33620, USA
| | - Charles E Chalfant
- Department of Cell Biology, Microbiology and Molecular Biology, University of South Florida, Tampa, FL, 33620, USA
- The Moffitt Cancer Center, Tampa, FL, 33620, USA
- Research Service, James A. Haley Veterans Hospital, Tampa, FL, 33612, USA
| | - Nawajes Mandal
- Department of Ophthalmology, Hamilton Eye Institute, The University of Tennessee Health Science Center, Memphis, TN, 38163, USA.
- Department of Ophthalmology, Dean McGee Eye Institute, The University of Oklahoma Health Sciences Center, Oklahoma City, OK-73104, USA.
- Department of Anatomy and Neurobiology, The University of Tennessee Health Science Center, Memphis, TN, 38163, USA.
- Department of Pharmaceutical Sciences, College of Pharmacy, The University of Tennessee Health Science Center, Memphis, TN, 38163, USA.
- Memphis VA Medical Center, Memphis, TN, 38104, USA.
| |
Collapse
|
8
|
Avula LR, Grodzinski P. Nanotechnology-aided advancement in the combating of cancer metastasis. Cancer Metastasis Rev 2022; 41:383-404. [PMID: 35366154 PMCID: PMC8975728 DOI: 10.1007/s10555-022-10025-7] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/16/2021] [Accepted: 03/09/2022] [Indexed: 02/03/2023]
Abstract
Cancer, especially when it has metastasized to different locations in the body, is notoriously difficult to treat. Metastatic cancer accounts for most cancer deaths and thus remains an enormous challenge. During the metastasis process, cancer cells negotiate a series of steps termed the “metastatic cascadeˮ that offer potential for developing anti-metastatic therapy strategies. Currently available conventional treatment and diagnostic methods addressing metastasis come with their own pitfalls and roadblocks. In this contribution, we comprehensively discuss the potential improvements that nanotechnology-aided approaches are able to bring, either alone or in combination with the existing conventional techniques, to the identification and treatment of metastatic disease. We tie specific nanotechnology-aided strategies to the complex biology of the different steps of the metastatic cascade in order to open up new avenues for fine-tuned targeting and development of anti-metastatic agents designed specifically to prevent or mitigate the metastatic outgrowth of cancer. We also present a viewpoint on the progress of translation of nanotechnology into cancer metastasis patient care.
Collapse
Affiliation(s)
- Leela Rani Avula
- Nanodelivery Systems and Devices Branch, Cancer Imaging Program, Division of Cancer Treatment and Diagnosis, National Cancer Institute, National Institutes of Health, Rockville, MD, USA.
| | - Piotr Grodzinski
- Nanodelivery Systems and Devices Branch, Cancer Imaging Program, Division of Cancer Treatment and Diagnosis, National Cancer Institute, National Institutes of Health, Rockville, MD, USA
| |
Collapse
|
9
|
Targeted liposomal doxorubicin/ceramides combinations: the importance to assess the nature of drug interaction beyond bulk tumor cells. Eur J Pharm Biopharm 2022; 172:61-77. [PMID: 35104605 DOI: 10.1016/j.ejpb.2022.01.006] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2021] [Revised: 01/14/2022] [Accepted: 01/18/2022] [Indexed: 12/26/2022]
Abstract
One of the major assets of anticancer nanomedicine is the ability to co-deliver drug combinations, as it enables targeting of different cellular populations and/or signaling pathways implicated in tumorigenesis and thus tackling tumor heterogeneity. Moreover, drug resistance can be circumvented, for example, upon co-encapsulation and delivery of doxorubicin and sphingolipids, as ceramides. Herein, the impact of short (C6) and long (C18) alkyl chain length ceramides on the nature of drug interaction, within the scope of combination with doxorubicin, was performed in bulk triple-negative breast cancer (TNBC) cells, as well as on the density of putative cancer stem cells and phenotype, including live single-cell tracking. C6- or C18-ceramide enabled a synergistic drug interaction in all conditions and (bulk) cell lines tested. However, differentiation among these two ceramides was reflected on the migratory potential of cancer cells, particularly significant against the highly motile MDA-MB-231 cells. This effect was supported by the downregulation of the PI3K/Akt pathway enabled by C6-ceramide and in contrast with C18-ceramide. The decrease of the migratory potential enabled by the targeted liposomal combinations is of high relevance in the context of TNBC, due to the underlying metastatic potential. Surprisingly, the nature of the drug interaction assessed at the level of bulk cancer cells revealed to be insufficient to predict whether a drug combination enables a decrease in the percentage of the master regulators of tumor relapse as ALDH+/high putative TNBC cancer stem cells, suggesting, for the first time, that it should be extended further down to this level.
Collapse
|
10
|
Piazzesi A, Afsar SY, van Echten‐Deckert G. Sphingolipid metabolism in the development and progression of cancer: one cancer's help is another's hindrance. Mol Oncol 2021; 15:3256-3279. [PMID: 34289244 PMCID: PMC8637577 DOI: 10.1002/1878-0261.13063] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2021] [Revised: 06/17/2021] [Accepted: 07/19/2021] [Indexed: 11/27/2022] Open
Abstract
Cancer development is a multistep process in which cells must overcome a series of obstacles before they can become fully developed tumors. First, cells must develop the ability to proliferate unchecked. Once this is accomplished, they must be able to invade the neighboring tissue, as well as provide themselves with oxygen and nutrients. Finally, they must acquire the ability to detach from the newly formed mass in order to spread to other tissues, all the while evading an immune system that is primed for their destruction. Furthermore, increased levels of inflammation have been shown to be linked to the development of cancer, with sites of chronic inflammation being a common component of tumorigenic microenvironments. In this Review, we give an overview of the impact of sphingolipid metabolism in cancers, from initiation to metastatic dissemination, as well as discussing immune responses and resistance to treatments. We explore how sphingolipids can either help or hinder the progression of cells from a healthy phenotype to a cancerous one.
Collapse
Affiliation(s)
- Antonia Piazzesi
- LIMES Institute for Membrane Biology and Lipid BiochemistryUniversity of BonnGermany
| | - Sumaiya Yasmeen Afsar
- LIMES Institute for Membrane Biology and Lipid BiochemistryUniversity of BonnGermany
| | | |
Collapse
|
11
|
Cruz AF, Caleiras MB, Fonseca NA, Gonçalves N, Mendes VM, Sampaio SF, Moura V, Melo JB, Almeida RD, Manadas B, Simões S, Moreira JN. The Enhanced Efficacy of Intracellular Delivery of Doxorubicin/C6-Ceramide Combination Mediated by the F3 Peptide/Nucleolin System Is Supported by the Downregulation of the PI3K/Akt Pathway. Cancers (Basel) 2021; 13:cancers13123052. [PMID: 34207464 PMCID: PMC8235382 DOI: 10.3390/cancers13123052] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2021] [Revised: 06/11/2021] [Accepted: 06/14/2021] [Indexed: 12/16/2022] Open
Abstract
Simple Summary Targeted nanomedicine-based approaches that aim at the simultaneous delivery of synergistic drug combinations to multiple cellular populations are of high relevance for tackling heterogeneity on solid tumors. Considering that cancer stem cells (CSC) may originate from non-stem cancer cells, single-drug regimens targeting only one of these cell populations could enable tumors to evade treatments. As such, the identification of a common marker, such as nucleolin, might result in a therapeutic advantage. The results herein generated suggested a transversal role of nucleolin in the internalization of F3 peptide-targeted pegylated pH-sensitive liposomes into bulk ovarian cancer cells, including putative CSC-enriched ovarian cells. The intracellular delivery of a drug combination such as the one tested herein was relevant in the context of cell lines with higher intrinsic resistances to doxorubicin. The enhanced efficacy of the F3 peptide-targeted liposomal combination of doxorubicin/C6-ceramide was supported by the downregulation of the Akt pathway, within a specific range of basal level of expression. Abstract Targeting multiple cellular populations is of high therapeutic relevance for the tackling of solid tumors heterogeneity. Herein, the ability of pegylated and pH-sensitive liposomes, functionalized with the nucleolin-binding F3 peptide and containing doxorubicin (DXR)/C6-ceramide synergistic combination, to target, in vitro, ovarian cancer, including ovarian cancer stem cells (CSC), was assessed. The underlying molecular mechanism of action of the nucleolin-mediated intracellular delivery of C6-ceramide to cancer cells was also explored. The assessment of overexpression of surface nucleolin expression by flow cytometry was critical to dissipate differences identified by Western blot in membrane/cytoplasm of SKOV-3, OVCAR-3 and TOV-112D ovarian cancer cell lines. The former was in line with the significant extent of uptake into (bulk) ovarian cancer cells, relative to non-targeted and non-specific counterparts. This pattern of uptake was recapitulated with putative CSC-enriched ovarian SKOV-3 and OVCAR-3 sub-population (EpCAMhigh/CD44high). Co-encapsulation of DXR:C6-ceramide into F3 peptide-targeted liposomes improved cytotoxic activity relative to liposomes containing DXR alone, in an extent that depended on the intrinsic resistance to DXR and on the incubation time. The enhanced cytotoxicity of the targeted combination was mechanistically supported by the downregulation of PI3K/Akt pathway by C6-ceramide, only among the nucleolin-overexpressing cancer cells presenting a basal p-Akt/total Akt ratio lower than 1.
Collapse
Affiliation(s)
- Ana F. Cruz
- CNC—Center for Neurosciences and Cell Biology, Center for Innovative Biomedicine and Biotechnology (CIBB), University of Coimbra, Faculty of Medicine (Polo 1), Rua Larga, 3004-504 Coimbra, Portugal; (A.F.C.); (M.B.C.); (N.A.F.); (N.G.); (V.M.M.); (S.F.S.); (V.M.); (R.D.A.); (B.M.); (S.S.)
- Univ Coimbra—University of Coimbra, CIBB, Faculty of Pharmacy, Pólo das Ciências da Saúde, Azinhaga de Santa Comba, 3000-548 Coimbra, Portugal
| | - Mariana B. Caleiras
- CNC—Center for Neurosciences and Cell Biology, Center for Innovative Biomedicine and Biotechnology (CIBB), University of Coimbra, Faculty of Medicine (Polo 1), Rua Larga, 3004-504 Coimbra, Portugal; (A.F.C.); (M.B.C.); (N.A.F.); (N.G.); (V.M.M.); (S.F.S.); (V.M.); (R.D.A.); (B.M.); (S.S.)
- Univ Coimbra—University of Coimbra, CIBB, Faculty of Pharmacy, Pólo das Ciências da Saúde, Azinhaga de Santa Comba, 3000-548 Coimbra, Portugal
| | - Nuno A. Fonseca
- CNC—Center for Neurosciences and Cell Biology, Center for Innovative Biomedicine and Biotechnology (CIBB), University of Coimbra, Faculty of Medicine (Polo 1), Rua Larga, 3004-504 Coimbra, Portugal; (A.F.C.); (M.B.C.); (N.A.F.); (N.G.); (V.M.M.); (S.F.S.); (V.M.); (R.D.A.); (B.M.); (S.S.)
- TREAT U, SA—Parque Industrial de Taveiro, Lote 44, 3045-508 Coimbra, Portugal
| | - Nélio Gonçalves
- CNC—Center for Neurosciences and Cell Biology, Center for Innovative Biomedicine and Biotechnology (CIBB), University of Coimbra, Faculty of Medicine (Polo 1), Rua Larga, 3004-504 Coimbra, Portugal; (A.F.C.); (M.B.C.); (N.A.F.); (N.G.); (V.M.M.); (S.F.S.); (V.M.); (R.D.A.); (B.M.); (S.S.)
| | - Vera M. Mendes
- CNC—Center for Neurosciences and Cell Biology, Center for Innovative Biomedicine and Biotechnology (CIBB), University of Coimbra, Faculty of Medicine (Polo 1), Rua Larga, 3004-504 Coimbra, Portugal; (A.F.C.); (M.B.C.); (N.A.F.); (N.G.); (V.M.M.); (S.F.S.); (V.M.); (R.D.A.); (B.M.); (S.S.)
| | - Susana F. Sampaio
- CNC—Center for Neurosciences and Cell Biology, Center for Innovative Biomedicine and Biotechnology (CIBB), University of Coimbra, Faculty of Medicine (Polo 1), Rua Larga, 3004-504 Coimbra, Portugal; (A.F.C.); (M.B.C.); (N.A.F.); (N.G.); (V.M.M.); (S.F.S.); (V.M.); (R.D.A.); (B.M.); (S.S.)
- Univ Coimbra—University of Coimbra, CIBB, Institute for Interdisciplinary Research (IIIUC), 3030-789 Coimbra, Portugal
| | - Vera Moura
- CNC—Center for Neurosciences and Cell Biology, Center for Innovative Biomedicine and Biotechnology (CIBB), University of Coimbra, Faculty of Medicine (Polo 1), Rua Larga, 3004-504 Coimbra, Portugal; (A.F.C.); (M.B.C.); (N.A.F.); (N.G.); (V.M.M.); (S.F.S.); (V.M.); (R.D.A.); (B.M.); (S.S.)
- TREAT U, SA—Parque Industrial de Taveiro, Lote 44, 3045-508 Coimbra, Portugal
| | - Joana B. Melo
- iCBR—Coimbra Institute for Clinical and Biomedical Research, CIBB, Center of Investigation on Environment Genetics and Oncobiology (CIMAGO), Pólo das Ciências da Saúde, Azinhaga de Santa Comba, 3000-548 Coimbra, Portugal;
- Univ Coimbra—University of Coimbra, Clinical Academic Center of Coimbra (CACC), Faculty of Medicine, Pólo das Ciências da Saúde, Azinhaga de Santa Comba, 3000-548 Coimbra, Portugal
| | - Ramiro D. Almeida
- CNC—Center for Neurosciences and Cell Biology, Center for Innovative Biomedicine and Biotechnology (CIBB), University of Coimbra, Faculty of Medicine (Polo 1), Rua Larga, 3004-504 Coimbra, Portugal; (A.F.C.); (M.B.C.); (N.A.F.); (N.G.); (V.M.M.); (S.F.S.); (V.M.); (R.D.A.); (B.M.); (S.S.)
| | - Bruno Manadas
- CNC—Center for Neurosciences and Cell Biology, Center for Innovative Biomedicine and Biotechnology (CIBB), University of Coimbra, Faculty of Medicine (Polo 1), Rua Larga, 3004-504 Coimbra, Portugal; (A.F.C.); (M.B.C.); (N.A.F.); (N.G.); (V.M.M.); (S.F.S.); (V.M.); (R.D.A.); (B.M.); (S.S.)
| | - Sérgio Simões
- CNC—Center for Neurosciences and Cell Biology, Center for Innovative Biomedicine and Biotechnology (CIBB), University of Coimbra, Faculty of Medicine (Polo 1), Rua Larga, 3004-504 Coimbra, Portugal; (A.F.C.); (M.B.C.); (N.A.F.); (N.G.); (V.M.M.); (S.F.S.); (V.M.); (R.D.A.); (B.M.); (S.S.)
- Univ Coimbra—University of Coimbra, CIBB, Faculty of Pharmacy, Pólo das Ciências da Saúde, Azinhaga de Santa Comba, 3000-548 Coimbra, Portugal
| | - João N. Moreira
- CNC—Center for Neurosciences and Cell Biology, Center for Innovative Biomedicine and Biotechnology (CIBB), University of Coimbra, Faculty of Medicine (Polo 1), Rua Larga, 3004-504 Coimbra, Portugal; (A.F.C.); (M.B.C.); (N.A.F.); (N.G.); (V.M.M.); (S.F.S.); (V.M.); (R.D.A.); (B.M.); (S.S.)
- Univ Coimbra—University of Coimbra, CIBB, Faculty of Pharmacy, Pólo das Ciências da Saúde, Azinhaga de Santa Comba, 3000-548 Coimbra, Portugal
- Correspondence:
| |
Collapse
|
12
|
Giacone DV, Dartora VFMC, de Matos JKR, Passos JS, Miranda DAG, de Oliveira EA, Silveira ER, Costa-Lotufo LV, Maria-Engler SS, Lopes LB. Effect of nanoemulsion modification with chitosan and sodium alginate on the topical delivery and efficacy of the cytotoxic agent piplartine in 2D and 3D skin cancer models. Int J Biol Macromol 2020; 165:1055-1065. [PMID: 32987080 DOI: 10.1016/j.ijbiomac.2020.09.167] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2020] [Revised: 08/24/2020] [Accepted: 09/20/2020] [Indexed: 01/11/2023]
Abstract
Due to the limited options for topical management of skin cancer, this study aimed at developing and evaluating nanoemulsions (NE) for topical delivery of the cytotoxic agent piplartine (piperlongumine). NEs were modified with chitosan or sodium alginate, and the effects on the physicochemical properties, piplartine delivery and formulation efficacy were evaluated. The nanoemulsion droplets displayed similar size (96-112 nm), but opposite charge; the polysaccharides improved piplartine penetration into and across the skin (1.3-1.9-fold) in a similar manner, increasing the ratio "drug in the skin/receptor phase" by 1.4-1.5-fold compared to the plain NE and highlighting their relevance for cutaneous localization. Oleic acid addition to the chitosan-containing NE further increased drug penetration (~1.9-2.0-fold), as did increases in drug content from 0.5 to 1%. The cytotoxicity of piplartine was ~2.8-fold higher when the drug was incorporated in the chitosan-containing NE compared to its solution (IC50 = 14.6 μM) against melanoma cells. The effects of this nanocarrier on 3D melanoma tissues were concentration-related; at 1%, piplartine elicited marked epidermis destruction. These results support the potential applicability of the chitosan-modified nanoemulsion containing piplartine as a new strategy for local management of skin cancer.
Collapse
Affiliation(s)
- Daniela V Giacone
- Institute of Biomedical Sciences, University of São Paulo, São Paulo, SP, Brazil
| | | | | | - Julia S Passos
- Institute of Biomedical Sciences, University of São Paulo, São Paulo, SP, Brazil
| | - Daniel A G Miranda
- Institute of Biomedical Sciences, University of São Paulo, São Paulo, SP, Brazil
| | - Erica A de Oliveira
- School of Pharmaceutical Sciences, University of São Paulo, São Paulo, SP, Brazil
| | - Edilberto R Silveira
- Department of Inorganic and Organic Chemistry, Federal University of Ceará, Fortaleza, CE, Brazil
| | | | | | - Luciana B Lopes
- Institute of Biomedical Sciences, University of São Paulo, São Paulo, SP, Brazil.
| |
Collapse
|
13
|
Huang Y, Zhao X, Zhang ZT, Chen SS, Li SS, Shi Z, Jing J, Huang A, Guo YM, Bai ZF, Zou ZS, Xiao XH, Wang JB, Niu M. Metabolomics Profiling and Diagnosis Biomarkers Searching for Drug-Induced Liver Injury Implicated to Polygonum multiflorum: A Cross-Sectional Cohort Study. Front Med (Lausanne) 2020; 7:592434. [PMID: 33330552 PMCID: PMC7734208 DOI: 10.3389/fmed.2020.592434] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2020] [Accepted: 10/30/2020] [Indexed: 12/17/2022] Open
Abstract
Aim: The diagnosis of drug-induced liver injury (DILI) remains a challenge and the cases of Polygonum multiflorum Thunb. (PM) induced DILI (PM-DILI) have received much attention This study aimed to identify a simple and high-efficiency approach to PM-DILI diagnosis via metabolomics analysis. Methods: Plasma metabolites in 13 PM-DILI patients were profiled by liquid chromatography along with high-resolution mass spectrometry. Meanwhile, the metabolic characteristics of the PM-DILI were compared with that of autoimmune hepatitis (AIH), hepatitis B (HBV), and healthy volunteers. Results: Twenty-four metabolites were identified to present significantly different levels in PM-DILI patients compared with HBV and AIH groups. These metabolites were enriched into glucose, amino acids, and sphingolipids metabolisms. Among these essential metabolites, the ratios of P-cresol sulfate vs. phenylalanine and inosine vs. bilirubin were further selected using a stepwise decision tree to construct a classification model in order to differentiate PM-DILI from HBV and AIH. The model was highly effective with sensitivity of 92.3% and specificity of 88.9%. Conclusions: This study presents an integrated view of the metabolic features of PM-DILI induced by herbal medicine, and the four-metabolite decision tree technique imparts a potent tool in clinical diagnosis.
Collapse
Affiliation(s)
- Ying Huang
- College of Pharmacy, Hunan University of Chinese Medicine, Changsha, China.,China Military Institute of Chinese Medicine, Fifth Medical Center of Chinese PLA General Hospital, Beijing, China
| | - Xu Zhao
- China Military Institute of Chinese Medicine, Fifth Medical Center of Chinese PLA General Hospital, Beijing, China
| | - Zi-Teng Zhang
- China Military Institute of Chinese Medicine, Fifth Medical Center of Chinese PLA General Hospital, Beijing, China
| | - Shuai-Shuai Chen
- China Military Institute of Chinese Medicine, Fifth Medical Center of Chinese PLA General Hospital, Beijing, China
| | - Shan-Shan Li
- China Military Institute of Chinese Medicine, Fifth Medical Center of Chinese PLA General Hospital, Beijing, China
| | - Zhuo Shi
- China Military Institute of Chinese Medicine, Fifth Medical Center of Chinese PLA General Hospital, Beijing, China
| | - Jing Jing
- China Military Institute of Chinese Medicine, Fifth Medical Center of Chinese PLA General Hospital, Beijing, China
| | - Ang Huang
- Center for Non-Infectious Liver Disease, Fifth Medical Center of Chinese PLA General Hospital, Beijing, China
| | - Yu-Ming Guo
- China Military Institute of Chinese Medicine, Fifth Medical Center of Chinese PLA General Hospital, Beijing, China
| | - Zhao-Fang Bai
- China Military Institute of Chinese Medicine, Fifth Medical Center of Chinese PLA General Hospital, Beijing, China
| | - Zheng-Sheng Zou
- Center for Non-Infectious Liver Disease, Fifth Medical Center of Chinese PLA General Hospital, Beijing, China
| | - Xiao-He Xiao
- China Military Institute of Chinese Medicine, Fifth Medical Center of Chinese PLA General Hospital, Beijing, China
| | - Jia-Bo Wang
- China Military Institute of Chinese Medicine, Fifth Medical Center of Chinese PLA General Hospital, Beijing, China
| | - Ming Niu
- China Military Institute of Chinese Medicine, Fifth Medical Center of Chinese PLA General Hospital, Beijing, China.,Department of Poisoning Treatment, Fifth Medical Center of Chinese PLA General Hospital, Beijing, China
| |
Collapse
|
14
|
Shaw JJP, Boyer TL, Venner E, Beck PJ, Slamowitz T, Caste T, Hickman A, Raymond MH, Costa-Pinheiro P, Jameson MJ, Fox TE, Kester M. Inhibition of Lysosomal Function Mitigates Protective Mitophagy and Augments Ceramide Nanoliposome-Induced Cell Death in Head and Neck Squamous Cell Carcinoma. Mol Cancer Ther 2020; 19:2621-2633. [PMID: 33087509 DOI: 10.1158/1535-7163.mct-20-0182] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2020] [Revised: 07/03/2020] [Accepted: 09/28/2020] [Indexed: 12/24/2022]
Abstract
Therapies for head and neck squamous cell carcinoma (HNSCC) are, at best, moderately effective, underscoring the need for new therapeutic strategies. Ceramide treatment leads to cell death as a consequence of mitochondrial damage by generating oxidative stress and causing mitochondrial permeability. However, HNSCC cells are able to resist cell death through mitochondria repair via mitophagy. Through the use of the C6-ceramide nanoliposome (CNL) to deliver therapeutic levels of bioactive ceramide, we demonstrate that the effects of CNL are mitigated in drug-resistant HNSCC via an autophagic/mitophagic response. We also demonstrate that inhibitors of lysosomal function, including chloroquine (CQ), significantly augment CNL-induced death in HNSCC cell lines. Mechanistically, the combination of CQ and CNL results in dysfunctional lysosomal processing of damaged mitochondria. We further demonstrate that exogenous addition of methyl pyruvate rescues cells from CNL + CQ-dependent cell death by restoring mitochondrial functionality via the reduction of CNL- and CQ-induced generation of reactive oxygen species and mitochondria permeability. Taken together, inhibition of late-stage protective autophagy/mitophagy augments the efficacy of CNL through preventing mitochondrial repair. Moreover, the combination of inhibitors of lysosomal function with CNL may provide an efficacious treatment modality for HNSCC.
Collapse
Affiliation(s)
- Jeremy J P Shaw
- Department of Pathology, University of Virginia, Charlottesville, Virginia
| | - Timothy L Boyer
- Department of Biomedical Engineering, University of Virginia, Charlottesville, Virginia
| | - Emily Venner
- Department of Biology, University of Virginia, Charlottesville, Virginia
| | - Patrick J Beck
- Department of Biomedical Engineering, University of Virginia, Charlottesville, Virginia
| | - Tristen Slamowitz
- Department of Biomedical Engineering, University of Virginia, Charlottesville, Virginia
| | - Tara Caste
- Department of Biomedical Engineering, University of Virginia, Charlottesville, Virginia
| | - Alexandra Hickman
- Department of Biomedical Engineering, University of Virginia, Charlottesville, Virginia
| | - Michael H Raymond
- Neuroscience Graduate Program, University of Virginia, Charlottesville, Virginia
| | | | - Mark J Jameson
- Department of Otolaryngology-Head and Neck Surgery, University of Virginia, Charlottesville, Virginia
| | - Todd E Fox
- Department of Pharmacology, University of Virginia, Charlottesville, Virginia
| | - Mark Kester
- Department of Biomedical Engineering, University of Virginia, Charlottesville, Virginia. .,Department of Pharmacology, University of Virginia, Charlottesville, Virginia
| |
Collapse
|
15
|
Preta G. New Insights Into Targeting Membrane Lipids for Cancer Therapy. Front Cell Dev Biol 2020; 8:571237. [PMID: 32984352 PMCID: PMC7492565 DOI: 10.3389/fcell.2020.571237] [Citation(s) in RCA: 77] [Impact Index Per Article: 15.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2020] [Accepted: 08/13/2020] [Indexed: 12/15/2022] Open
Abstract
Modulation of membrane lipid composition and organization is currently developing as an effective therapeutic strategy against a wide range of diseases, including cancer. This field, known as membrane-lipid therapy, has risen from new discoveries on the complex organization of lipids and between lipids and proteins in the plasma membranes. Membrane microdomains present in the membrane of all eukaryotic cells, known as lipid rafts, have been recognized as an important concentrating platform for protein receptors involved in the regulation of intracellular signaling, apoptosis, redox balance and immune response. The difference in lipid composition between the cellular membranes of healthy cells and tumor cells allows for the development of novel therapies based on targeting membrane lipids in cancer cells to increase sensitivity to chemotherapeutic agents and consequently defeat multidrug resistance. In the current manuscript strategies based on influencing cholesterol/sphingolipids content will be presented together with innovative ones, more focused in changing biophysical properties of the membrane bilayer without affecting the composition of its constituents.
Collapse
Affiliation(s)
- Giulio Preta
- Institute of Biochemistry, Life Sciences Center, Vilnius University, Vilnius, Lithuania
| |
Collapse
|
16
|
Kurmi BD, Patel P, Paliwal R, Paliwal SR. Molecular approaches for targeted drug delivery towards cancer: A concise review with respect to nanotechnology. J Drug Deliv Sci Technol 2020. [DOI: 10.1016/j.jddst.2020.101682] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
|
17
|
Exogenous Liposomal Ceramide-C6 Ameliorates Lipidomic Profile, Energy Homeostasis, and Anti-Oxidant Systems in NASH. Cells 2020; 9:cells9051237. [PMID: 32429478 PMCID: PMC7290333 DOI: 10.3390/cells9051237] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2020] [Revised: 05/05/2020] [Accepted: 05/13/2020] [Indexed: 02/07/2023] Open
Abstract
In non-alcoholic steatohepatitis (NASH), many lines of investigation have reported a dysregulation in lipid homeostasis, leading to intrahepatic lipid accumulation. Recently, the role of dysfunctional sphingolipid metabolism has also been proposed. Human and animal models of NASH have been associated with elevated levels of long chain ceramides and pro-apoptotic sphingolipid metabolites, implicated in regulating fatty acid oxidation and inflammation. Importantly, inhibition of de novo ceramide biosynthesis or knock-down of ceramide synthases reverse some of the pathology of NASH. In contrast, cell permeable, short chain ceramides have shown anti-inflammatory actions in multiple models of inflammatory disease. Here, we investigated non-apoptotic doses of a liposome containing short chain C6-Ceramide (Lip-C6) administered to human hepatic stellate cells (hHSC), a key effector of hepatic fibrogenesis, and an animal model characterized by inflammation and elevated liver fat content. On the basis of the results from unbiased liver transcriptomic studies from non-alcoholic fatty liver disease patients, we chose to focus on adenosine monophosphate activated kinase (AMPK) and nuclear factor-erythroid 2-related factor (Nrf2) signaling pathways, which showed an abnormal profile. Lip-C6 administration inhibited hHSC proliferation while improving anti-oxidant protection and energy homeostasis, as indicated by upregulation of Nrf2, activation of AMPK and an increase in ATP. To confirm these in vitro data, we investigated the effect of a single tail-vein injection of Lip-C6 in the methionine-choline deficient (MCD) diet mouse model. Lip-C6, but not control liposomes, upregulated phospho-AMPK, without inducing liver toxicity, apoptosis, or exacerbating inflammatory signaling pathways. Alluding to mechanism, mass spectrometry lipidomics showed that Lip-C6-treatment reversed the imbalance in hepatic phosphatidylcholines and diacylglycerides species induced by the MCD-fed diet. These results reveal that short-term Lip-C6 administration reverses energy/metabolic depletion and increases protective anti-oxidant signaling pathways, possibly by restoring homeostatic lipid function in a model of liver inflammation with fat accumulation.
Collapse
|
18
|
Ghosh S, Juin SK, Nandi P, Majumdar SB, Bose A, Baral R, Sil PC, Majumdar S. PKCζ mediated anti-proliferative effect of C2 ceramide on neutralization of the tumor microenvironment and melanoma regression. Cancer Immunol Immunother 2020; 69:611-627. [PMID: 31996991 PMCID: PMC11027884 DOI: 10.1007/s00262-020-02492-0] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2019] [Accepted: 01/18/2020] [Indexed: 12/12/2022]
Abstract
Immunotherapy, which has advantages over chemotherapy due to lesser toxicity and higher specificity, is on the rise to treat cancer. Recently, pro-apoptotic glycolipid, ceramide has emerged as a key regulator in cancer immunotherapy. The present study elucidated the potential anti-melanoma efficacy of cell-permeable, exogenous C2 ceramide on cell death and amelioration of tumor microenvironment (TME). We, for the first time, demonstrated that C2 ceramide triggered apoptosis of melanoma cells by augmenting PKCζ along with pro-inflammatory cytokines and signaling factors. C2 ceramide showed a PKCζ-mediated tumor-suppressive role in melanoma without exhibiting hepatotoxicity and nephrotoxicity. Moreover, PKCζ was revealed as one of the key regulators of Akt and ceramide during C2 ceramide-mediated apoptosis. C2 ceramide was effective in repolarization of M2 macrophage phenotype and reduction of angiogenic factors such as VEGF, VEGFR1, VEGFR2, HIF1α. Interestingly, PKCζ knockdown attenuated C2 ceramide-mediated inhibition of melanoma progression. Restoration of the Th1 type TME by C2 ceramide enhanced cytotoxic T cell-mediated killing of melanoma cells. Altogether, the study unraveled that C2 ceramide-induced PKCζ was associated with favorable immune cell functioning in TME leading to melanoma regression. Thus, our findings explored a novel mechanistic insight into C2 ceramide as a promising immunotherapeutic agent in melanoma treatment.
Collapse
Affiliation(s)
- Sweta Ghosh
- Division of Molecular Medicine, Bose Institute, P-1/12, CIT Scheme VII M, Kolkata, 700054, India
| | - Subir Kumar Juin
- Division of Molecular Medicine, Bose Institute, P-1/12, CIT Scheme VII M, Kolkata, 700054, India
| | - Partha Nandi
- Department of Immunoregulation and Immunodiagnostics, Chittaranjan National Cancer Institute (CNCI), Kolkata, 700026, India
| | | | - Anamika Bose
- Department of Immunoregulation and Immunodiagnostics, Chittaranjan National Cancer Institute (CNCI), Kolkata, 700026, India
| | - Rathindranath Baral
- Department of Immunoregulation and Immunodiagnostics, Chittaranjan National Cancer Institute (CNCI), Kolkata, 700026, India
| | - Parames C Sil
- Division of Molecular Medicine, Bose Institute, P-1/12, CIT Scheme VII M, Kolkata, 700054, India
| | - Subrata Majumdar
- Division of Molecular Medicine, Bose Institute, P-1/12, CIT Scheme VII M, Kolkata, 700054, India.
| |
Collapse
|
19
|
Visentin S, Sedić M, Pavelić SK, Pavelić K. Targeting Tumour Metastasis: The Emerging Role of Nanotechnology. Curr Med Chem 2020; 27:1367-1381. [DOI: 10.2174/0929867326666181220095343] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2017] [Revised: 02/13/2018] [Accepted: 11/12/2018] [Indexed: 12/29/2022]
Abstract
The metastatic process has still not been completely elucidated, probably due to insufficient knowledge of the underlying mechanisms. Here, we provide an overview of the current findings that shed light on specific molecular alterations associated with metastasis and present novel concepts in the treatment of the metastatic process. In particular, we discuss novel pharmacological approaches in the clinical setting that target metastatic progression. New insights into the process of metastasis allow optimisation and design of new treatment strategies, especially in view of the fact that metastatic cells share common features with stem cells. Nano- and micro-technologies are herein elaborated in details as a promising therapeutic concept in targeted drug delivery for metastatic cancer. Progression in the field could provide a more efficient way to tackle metastasis and thus bring about advancements in the treatment and management of patients with advanced cancer.
Collapse
Affiliation(s)
- Sarah Visentin
- Department of Biotechnology, University of Rijeka, Centre for High-Throughput Technologies, Radmile Matejcic 2, 51 000 Rijeka, Croatia
| | - Mirela Sedić
- Department of Biotechnology, University of Rijeka, Centre for High-Throughput Technologies, Radmile Matejcic 2, 51 000 Rijeka, Croatia
| | - Sandra Kraljević Pavelić
- Department of Biotechnology, University of Rijeka, Centre for High-Throughput Technologies, Radmile Matejcic 2, 51 000 Rijeka, Croatia
| | - Krešimir Pavelić
- Faculty of Medicine, Juraj Dobrila University of Pula, Zagrebacka 30, 52 100 Pula, Croatia
| |
Collapse
|
20
|
Choi MK, Song IS. Recent advances in the formulation of sphingolipid anticancer therapeutics. JOURNAL OF PHARMACEUTICAL INVESTIGATION 2020. [DOI: 10.1007/s40005-020-00475-y] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
|
21
|
Interaction of Synaptosomal-Associated Protein 25 with Neutral Sphingomyelinase 2: Functional Impact on the Sphingomyelin Pathway. Neuroscience 2020; 427:1-15. [PMID: 31765623 DOI: 10.1016/j.neuroscience.2019.08.015] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2019] [Revised: 08/06/2019] [Accepted: 08/07/2019] [Indexed: 11/22/2022]
Abstract
Neurotransmitter release is mediated by ceramide, which is generated by sphingomyelin hydrolysis. In the present study, we examined whether synaptosomal-associated protein 25 (SNAP-25) is involved in ceramide production and exocytosis. Neutral sphingomyelinase 2 (nSMase2) was partially purified from bovine brain and we found that SNAP-25 was enriched in the nSMase2-containing fractions. In rat synaptosomes and PC12 cells, the immunoprecipitation pellet of anti-SNAP-25 antibody showed higher nSMase activity than the immunoprecipitation pellet of anti-nSMase2 antibody. In PC12 cells, SNAP-25 was colocalized with nSMase2. Transfection of SNAP-25 small interfering RNA (siRNA) significantly inhibited nSMase2 translocation to the plasma membrane. A23187-induced ceramide production was concomitantly reduced in SNAP-25 siRNA-transfected PC12 cells compared with that in scrambled siRNA-transfected cells. Moreover, transfection of SNAP-25 siRNA inhibited dopamine release, whereas addition of C6-ceramide to the siRNA-treated cells moderately reversed this inhibition. Additionally, nSMase2 inhibition reduced dopamine release. Collectively, our results indicate that SNAP-25 interacts with nSMase2 during ceramide production, which mediates exocytosis and neurotransmitter release.
Collapse
|
22
|
Alrbyawi H, Poudel I, Dash RP, Srinivas NR, Tiwari AK, Arnold RD, Babu RJ. Role of Ceramides in Drug Delivery. AAPS PharmSciTech 2019; 20:287. [PMID: 31410612 DOI: 10.1208/s12249-019-1497-6] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2019] [Accepted: 07/31/2019] [Indexed: 12/20/2022] Open
Abstract
Ceramides belong to the sphingolipid group of lipids, which serve as both intracellular and intercellular messengers and as regulatory molecules that play essential roles in signal transduction, inflammation, angiogenesis, and metabolic disorders such as diabetes, neurodegenerative diseases, and cancer cell degeneration. Ceramides also play an important structural role in cell membranes by increasing their rigidity, creating micro-domains (rafts and caveolae), and altering membrane permeability; all these events are involved in the cell signaling. Ceramides constitute approximately half of the lipid composition in the human skin contributing to barrier function as well as epidermal signaling as they affect both proliferation and apoptosis of keratinocytes. Incorporation of ceramides in topical preparations as functional lipids appears to alter skin barrier functions. Ceramides also appear to enhance the bioavailability of drugs by acting as lipid delivery systems. They appear to regulate the ocular inflammation signaling, and external ceramides have shown relief in the anterior and posterior eye disorders. Ceramides play a structural role in liposome formulations and enhance the cellular uptake of amphiphilic drugs, such as chemotherapies. This review presents an overview of the various biological functions of ceramides, and their utility in topical, oral, ocular, and chemotherapeutic drug delivery.
Collapse
|
23
|
Kudaibergenova M, Perissinotti LL, Noskov SY. Lipid roles in hERG function and interactions with drugs. Neurosci Lett 2019; 700:70-77. [DOI: 10.1016/j.neulet.2018.05.019] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2018] [Revised: 05/08/2018] [Accepted: 05/11/2018] [Indexed: 01/29/2023]
|
24
|
Mojeiko G, de Brito M, Salata GC, Lopes LB. Combination of microneedles and microemulsions to increase celecoxib topical delivery for potential application in chemoprevention of breast cancer. Int J Pharm 2019; 560:365-376. [DOI: 10.1016/j.ijpharm.2019.02.011] [Citation(s) in RCA: 36] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2018] [Revised: 01/23/2019] [Accepted: 02/06/2019] [Indexed: 12/16/2022]
|
25
|
Chen L, Alrbyawi H, Poudel I, Arnold RD, Babu RJ. Co-delivery of Doxorubicin and Ceramide in a Liposomal Formulation Enhances Cytotoxicity in Murine B16BL6 Melanoma Cell Lines. AAPS PharmSciTech 2019; 20:99. [PMID: 30719596 DOI: 10.1208/s12249-019-1316-0] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2018] [Accepted: 01/22/2019] [Indexed: 02/07/2023] Open
Abstract
This study reports co-delivery of doxorubicin (DOX) and ceramide in a liposomal system in B16BL6 melanoma cell lines for enhanced cytotoxic effects. Different types of ceramides (C6-ceramide, C8-ceramide, and C8-glucosylceramide) and lipids (1,2-dioleoyl-3-trimethylammonium-propane (DOTAP), 1,2-dipalmitoyl-sn-glycero-3-phosphocholine (DPPC), 1,2-distearoyl-sn-glycero-3-phosphocholine (DSPC), 1,2-distearoyl-sn-glycero-3-phosphoethanolamine (DSPE)) were considered in the preparation of liposomes. DOX was encapsulated within liposome, and ceramide was used as the component of the lipid bilayer. The formulations were optimized for size and size distribution, zeta potential, and DOX encapsulation efficiency (EE). Cytotoxic effect on B16BL6 melanoma cell lines was measured by 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide (MTT) assay. The ceramide based liposome formulations generally provided a mean diameter < 181 nm, a zeta potential, + 35 mV, and EE > 90% DOX EE. Co-delivery of DOX and C8-ceramide with DOTAP liposomes demonstrated significantly higher cytotoxicity as compared to DOX liposomes without ceramide (P < 0.001), and also showed enhanced cellular uptake by B16BL6 cell lines. This study provides basis for developing a co-delivery system of DOX and ceramide for lowering the dose and dose-related side effects of DOX for the treatment of melanoma.
Collapse
|
26
|
Xu J, Zhao W, Sun J, Huang Y, Wang P, Venkataramanan R, Yang D, Ma X, Rana A, Li S. Novel glucosylceramide synthase inhibitor based prodrug copolymer micelles for delivery of anticancer agents. J Control Release 2018; 288:212-226. [PMID: 30223045 PMCID: PMC6177216 DOI: 10.1016/j.jconrel.2018.09.011] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2018] [Revised: 09/07/2018] [Accepted: 09/12/2018] [Indexed: 01/04/2023]
Abstract
In order to improve the efficacy of chemotherapy for cancers, we have developed a novel prodrug micellar formulation to co-deliver ceramide-generating anticancer agents and ceramide degradation inhibitor (PPMP). The prodrug nanocarrier is based on a well-defined POEG-b-PPPMP diblock copolymer. The hydrophilic block of POEG-b-PPPMP is POEG, and the hydrophobic block is composed of a number of PPMP units, which could work synergistically with loaded anticancer drugs. POEG-b-PPPMP was readily synthesized via a one-step reversible addition-fragment transfer (RAFT) polymerization from a PPMP monomer. The newly synthesized polymers were self-assembled into micelles and served as a carrier for several hydrophobic anticancer drugs including DOX, PTX and C6-ceramide. POEG-b-PPPMP prodrug polymer exhibited intrinsic antitumor activity in vitro and in vivo. In addition, POEG-b-PPPMP prodrug polymer was comparable to free PPMP in selectively enhancing the production of pro-apoptotic ceramide species as well as down-regulating the mRNA expression of GCS. DOX-loaded POEG-b-PPPMP micelles exhibited an excellent stability of 42 days at 4 °C. Moreover, DOX loaded in POEG-b-PPPMP micelles showed higher levels of cytotoxicity than DOX loaded in a pharmacologically inert polymer (POEG-b-POM) and Doxil formulation in several tumor cell lines. Consistently, in a 4T1.2 murine breast cancer model, the tumor inhibition followed the order of DOX/POEG-b-PPPMP > DOX/POEG-b-POM ≥ Doxil > POEG-b-PPPMP > POEG-b-POM. Our data suggest that POEG-b-PPPMP micelles are a promising dual-functional carrier that warrants more studies in the future.
Collapse
Affiliation(s)
- Jieni Xu
- Center for Pharmacogenetics, University of Pittsburgh, Pittsburgh, PA 15261, USA; Department of Pharmaceutical Sciences, School of Pharmacy, University of Pittsburgh, Pittsburgh, PA 15261, USA; University of Pittsburgh Cancer Institute, University of Pittsburgh, Pittsburgh, PA 15261, USA
| | - Whenchen Zhao
- Department of Pharmaceutical Sciences, School of Pharmacy, University of Pittsburgh, Pittsburgh, PA 15261, USA
| | - Jingjing Sun
- Center for Pharmacogenetics, University of Pittsburgh, Pittsburgh, PA 15261, USA; Department of Pharmaceutical Sciences, School of Pharmacy, University of Pittsburgh, Pittsburgh, PA 15261, USA; University of Pittsburgh Cancer Institute, University of Pittsburgh, Pittsburgh, PA 15261, USA
| | - Yixian Huang
- Center for Pharmacogenetics, University of Pittsburgh, Pittsburgh, PA 15261, USA; Department of Pharmaceutical Sciences, School of Pharmacy, University of Pittsburgh, Pittsburgh, PA 15261, USA; University of Pittsburgh Cancer Institute, University of Pittsburgh, Pittsburgh, PA 15261, USA
| | - Pengcheng Wang
- Center for Pharmacogenetics, University of Pittsburgh, Pittsburgh, PA 15261, USA; Department of Pharmaceutical Sciences, School of Pharmacy, University of Pittsburgh, Pittsburgh, PA 15261, USA
| | - Raman Venkataramanan
- Department of Pharmaceutical Sciences, School of Pharmacy, University of Pittsburgh, Pittsburgh, PA 15261, USA
| | - Da Yang
- Center for Pharmacogenetics, University of Pittsburgh, Pittsburgh, PA 15261, USA; Department of Pharmaceutical Sciences, School of Pharmacy, University of Pittsburgh, Pittsburgh, PA 15261, USA
| | - Xiaochao Ma
- Center for Pharmacogenetics, University of Pittsburgh, Pittsburgh, PA 15261, USA; Department of Pharmaceutical Sciences, School of Pharmacy, University of Pittsburgh, Pittsburgh, PA 15261, USA
| | - Ajay Rana
- Department of Surgery/Surgical Oncology, College of Medicine, University of Illinois at Chicago, Chicago, IL 60612, USA
| | - Song Li
- Center for Pharmacogenetics, University of Pittsburgh, Pittsburgh, PA 15261, USA; Department of Pharmaceutical Sciences, School of Pharmacy, University of Pittsburgh, Pittsburgh, PA 15261, USA; University of Pittsburgh Cancer Institute, University of Pittsburgh, Pittsburgh, PA 15261, USA.
| |
Collapse
|
27
|
Migotto A, Carvalho VFM, Salata GC, da Silva FWM, Yan CYI, Ishida K, Costa-Lotufo LV, Steiner AA, Lopes LB. Multifunctional nanoemulsions for intraductal delivery as a new platform for local treatment of breast cancer. Drug Deliv 2018; 25:654-667. [PMID: 29495885 PMCID: PMC7011997 DOI: 10.1080/10717544.2018.1440665] [Citation(s) in RCA: 47] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
Considering that breast cancer usually begins in the lining of the ducts, local drug administration into the ducts could target cancers and pre-tumor lesions locally while reducing systemic adverse effects. In this study, a cationic bioadhesive nanoemulsion was developed for intraductal administration of C6 ceramide, a sphingolipid that mediates apoptotic and non-apoptotic cell death. Bioadhesive properties were obtained by surface modification with chitosan. The optimized nanoemulsion displayed size of 46.3 nm and positive charge, properties that were not affected by ceramide encapsulation (0.4%, w/w). C6 ceramide concentration necessary to reduce MCF-7 cells viability to 50% (EC50) decreased by 4.5-fold with its nanoencapsulation compared to its solution; a further decrease (2.6-fold) was observed when tributyrin (a pro-drug of butyric acid) was part of the oil phase of the nanocarrier, a phenomenon attributed to synergism. The unloaded nanocarrier was considered safe, as indicated by a score <0.1 in HET-CAM models, by the high survival rates of Galleria mellonella larvae exposed to concentrations ≤500 mg/mL, and absence of histological changes when intraductally administered in rats. Intraductal administration of the nanoemulsion prolonged drug localization for more than 120 h in the mammary tissue compared to its solution. These results support the advantage of the optimized nanoemulsion to enable mammary tissue localization of C6 ceramide.
Collapse
Affiliation(s)
- Amanda Migotto
- a Department of Pharmacology , Institute of Biomedical Sciences, University of Sao Paulo , Sao Paulo , Brazil
| | - Vanessa F M Carvalho
- a Department of Pharmacology , Institute of Biomedical Sciences, University of Sao Paulo , Sao Paulo , Brazil
| | - Giovanna C Salata
- a Department of Pharmacology , Institute of Biomedical Sciences, University of Sao Paulo , Sao Paulo , Brazil
| | - Fernanda W M da Silva
- b Department of Microbiology , Institute of Biomedical Sciences, University of Sao Paulo , Sao Paulo , Brazil
| | - Chao Yun Irene Yan
- c Department of Cell and Developmental Biology , Institute of Biomedical Sciences, University of Sao Paulo , Sao Paulo , Brazil
| | - Kelly Ishida
- b Department of Microbiology , Institute of Biomedical Sciences, University of Sao Paulo , Sao Paulo , Brazil
| | - Leticia V Costa-Lotufo
- a Department of Pharmacology , Institute of Biomedical Sciences, University of Sao Paulo , Sao Paulo , Brazil
| | - Alexandre A Steiner
- d Department of Immunology , Institute of Biomedical Sciences, University of Sao Paulo , Sao Paulo , Brazil
| | - Luciana B Lopes
- a Department of Pharmacology , Institute of Biomedical Sciences, University of Sao Paulo , Sao Paulo , Brazil
| |
Collapse
|
28
|
Purwaha P, Gu F, Piyarathna DWB, Rajendiran T, Ravindran A, Omilian AR, Jiralerspong S, Das G, Morrison C, Ambrosone C, Coarfa C, Putluri N, Sreekumar A. Unbiased Lipidomic Profiling of Triple-Negative Breast Cancer Tissues Reveals the Association of Sphingomyelin Levels with Patient Disease-Free Survival. Metabolites 2018; 8:metabo8030041. [PMID: 30011843 PMCID: PMC6161031 DOI: 10.3390/metabo8030041] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2018] [Revised: 07/02/2018] [Accepted: 07/04/2018] [Indexed: 12/26/2022] Open
Abstract
The reprogramming of lipid metabolism is a hallmark of many cancers that has been shown to promote breast cancer progression. While several lipid signatures associated with breast cancer aggressiveness have been identified, a comprehensive lipidomic analysis specifically targeting the triple-negative subtype of breast cancer (TNBC) may be required to identify novel biomarkers and therapeutic targets for this most aggressive subtype of breast cancer that still lacks effective therapies. In this current study, our global LC-MS-based lipidomics platform was able to measure 684 named lipids across 15 lipid classes in 70 TNBC tumors. Multivariate survival analysis found that higher levels of sphingomyelins were significantly associated with better disease-free survival in TNBC patients. Furthermore, analysis of publicly available gene expression datasets identified that decreased production of ceramides and increased accumulation of sphingoid base intermediates by metabolic enzymes were associated with better survival outcomes in TNBC patients. Our LC-MS lipidomics profiling of TNBC tumors has, for the first time, identified sphingomyelins as a potential prognostic marker and implicated enzymes involved in sphingolipid metabolism as candidate therapeutic targets that warrant further investigation.
Collapse
Affiliation(s)
- Preeti Purwaha
- Alkek Center for Molecular Discovery and Department of Molecular and Cell Biology, Baylor College of Medicine, Houston, TX 77030, USA.
| | - Franklin Gu
- Alkek Center for Molecular Discovery and Department of Molecular and Cell Biology, Baylor College of Medicine, Houston, TX 77030, USA.
- Verna and Mars McLean Department of Biochemistry and Molecular Biology, Baylor College of Medicine, Houston, TX 77030, USA.
| | | | | | - Anindita Ravindran
- Alkek Center for Molecular Discovery and Department of Molecular and Cell Biology, Baylor College of Medicine, Houston, TX 77030, USA.
| | - Angela R Omilian
- Roswell Park Comprehensive Cancer Center, Buffalo, NY 14263, USA.
| | - Sao Jiralerspong
- Alkek Center for Molecular Discovery and Department of Molecular and Cell Biology, Baylor College of Medicine, Houston, TX 77030, USA.
| | - Gokul Das
- Roswell Park Comprehensive Cancer Center, Buffalo, NY 14263, USA.
| | - Carl Morrison
- Roswell Park Comprehensive Cancer Center, Buffalo, NY 14263, USA.
| | | | - Cristian Coarfa
- Alkek Center for Molecular Discovery and Department of Molecular and Cell Biology, Baylor College of Medicine, Houston, TX 77030, USA.
| | - Nagireddy Putluri
- Alkek Center for Molecular Discovery and Department of Molecular and Cell Biology, Baylor College of Medicine, Houston, TX 77030, USA.
| | - Arun Sreekumar
- Alkek Center for Molecular Discovery and Department of Molecular and Cell Biology, Baylor College of Medicine, Houston, TX 77030, USA.
- Verna and Mars McLean Department of Biochemistry and Molecular Biology, Baylor College of Medicine, Houston, TX 77030, USA.
| |
Collapse
|
29
|
Yin X, Xiao Y, Han L, Zhang B, Wang T, Su Z, Zhang N. Ceramide-Fabricated Co-Loaded Liposomes for the Synergistic Treatment of Hepatocellular Carcinoma. AAPS PharmSciTech 2018; 19:2133-2143. [PMID: 29714002 DOI: 10.1208/s12249-018-1005-4] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2017] [Accepted: 03/24/2018] [Indexed: 02/06/2023] Open
Abstract
Combination therapy is one of the important methods to improve therapeutic effect on the treatment of hepatocellular carcinoma (HCC). Sorafenib (SF) is a canonical US Food and Drug Administration-approved multikinase molecule inhibitor against HCC. However, therapeutic benefit with Sorafenib alone was usually unsatisfactory. Ceramide (CE) is an endogenous bioactive sphingolipid, which has a strong potential to suppress various tumors. The combination of SF and CE was hoping to exert maximum synergistic antitumor effect through different tumor-suppressible mechanisms. In this respect, SF and CE co-loaded liposomes (SF/CE-liposomes) were developed to verify synergistic antitumor efficacy. The optimal molar ratio of SF and CE was determined through combination index. SF/CE-liposomes were prepared by thin-film hydration method, which exhibited spherical or ellipsoidal shape. Particle size of SF/CE-liposomes was 174 ± 4 nm with homogeneous distribution. Release profile of SF demonstrated that addition of CE imposed no significant impact on the release of SF. SF/CE-liposomes exhibited acceptable stability in different media and desirable storage stability over 30 days at 4°C. In vitro cellular uptake confirmed that SF/CE-liposomes could be efficiently internalized into HepG2 cells. In vitro cytotoxicity evaluation indicated that SF/CE-liposomes exhibited higher cytotoxicity on HepG2 cells. IC50 value of SF/CE-liposomes was 11.5 ± 0.44 μM, which was significantly lower than that of SF-liposomes (**p < 0.01). Evaluation of in vivo synergistic effect on H22-bearing mice verified that SF/CE-liposomes achieved robust antitumor activity in preventing tumor growth. All results suggested that SF/CE-liposomes might be served as an efficient co-delivery system for improving therapeutic efficacy of HCC.
Collapse
|
30
|
Zhang X, Kitatani K, Toyoshima M, Ishibashi M, Usui T, Minato J, Egiz M, Shigeta S, Fox T, Deering T, Kester M, Yaegashi N. Ceramide Nanoliposomes as a MLKL-Dependent, Necroptosis-Inducing, Chemotherapeutic Reagent in Ovarian Cancer. Mol Cancer Ther 2018; 17:50-59. [PMID: 29079707 PMCID: PMC5752574 DOI: 10.1158/1535-7163.mct-17-0173] [Citation(s) in RCA: 43] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2017] [Revised: 08/28/2017] [Accepted: 10/10/2017] [Indexed: 12/12/2022]
Abstract
Ceramides are bioactive lipids that mediate cell death in cancer cells, and ceramide-based therapy is now being tested in dose-escalating phase I clinical trials as a cancer treatment. Multiple nanoscale delivery systems for ceramide have been proposed to overcome the inherent toxicities, poor pharmacokinetics, and difficult biophysics associated with ceramide. Using the ceramide nanoliposomes (CNL), we now investigate the therapeutic efficacy and signaling mechanisms of this nanoscale delivery platform in refractory ovarian cancer. Treatment of ovarian cancer cells with CNL decreased the number of living cells through necroptosis but not apoptosis. Mechanistically, dying SKOV3 ovarian cancer cells exhibit activation of pseudokinase mixed lineage kinase domain-like (MLKL) as evidenced by oligomerization and relocalization to the blebbing membranes, showing necroptotic characteristics. Knockdown of MLKL, but not its upstream protein kinases such as receptor-interacting protein kinases, with siRNA significantly abolished CNL-induced cell death. Monomeric MLKL protein expression inversely correlated with the IC50 values of CNL in distinct ovarian cancer cell lines, suggesting MLKL as a possible determinant for CNL-induced cell death. Finally, systemic CNL administration suppressed metastatic growth in an ovarian cancer cell xenograft model. Taken together, these results suggest that MLKL is a novel pronecroptotic target for ceramide in ovarian cancer models. Mol Cancer Ther; 17(1); 50-59. ©2017 AACR.
Collapse
Affiliation(s)
- Xuewei Zhang
- Department of Obstetrics and Gynecology, Tohoku University School of Medicine, Tohoku University, Sendai, Japan
| | - Kazuyuki Kitatani
- Department of Obstetrics and Gynecology, Tohoku University School of Medicine, Tohoku University, Sendai, Japan.
- Tohoku Medical Megabank Organization, Tohoku University, Sendai, Japan
| | - Masafumi Toyoshima
- Department of Obstetrics and Gynecology, Tohoku University School of Medicine, Tohoku University, Sendai, Japan.
| | - Masumi Ishibashi
- Department of Obstetrics and Gynecology, Tohoku University School of Medicine, Tohoku University, Sendai, Japan
| | - Toshinori Usui
- Tohoku Medical Megabank Organization, Tohoku University, Sendai, Japan
| | - Junko Minato
- Department of Obstetrics and Gynecology, Tohoku University School of Medicine, Tohoku University, Sendai, Japan
| | - Mahy Egiz
- Department of Obstetrics and Gynecology, Tohoku University School of Medicine, Tohoku University, Sendai, Japan
| | - Shogo Shigeta
- Department of Obstetrics and Gynecology, Tohoku University School of Medicine, Tohoku University, Sendai, Japan
| | - Todd Fox
- Department of Pharmacology, University of Virginia, Charlottesville, Virginia
| | - Tye Deering
- Department of Pharmacology, University of Virginia, Charlottesville, Virginia
| | - Mark Kester
- Department of Pharmacology, University of Virginia, Charlottesville, Virginia
| | - Nobuo Yaegashi
- Department of Obstetrics and Gynecology, Tohoku University School of Medicine, Tohoku University, Sendai, Japan
- Tohoku Medical Megabank Organization, Tohoku University, Sendai, Japan
| |
Collapse
|
31
|
Minnelli C, Cianfruglia L, Laudadio E, Galeazzi R, Pisani M, Crucianelli E, Bizzaro D, Armeni T, Mobbili G. Selective induction of apoptosis in MCF7 cancer-cell by targeted liposomes functionalised with mannose-6-phosphate. J Drug Target 2017; 26:242-251. [PMID: 28795851 DOI: 10.1080/1061186x.2017.1365873] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
Liposomes are versatile platforms to carry anticancer drugs in targeted drug delivery; they can be surface modified by different strategies and, when coupled with targeting ligands, are able to increase cellular internalisation and organelle-specific drug delivery. An interesting strategy of antitumoral therapy could involve the use of lysosomotropic ligand-targeted liposomes loaded with molecules, which can induce lysosomal membrane permeabilization (LMP), leakage of cathepsins into the cytoplasm and subsequent apoptosis. We have previously demonstrated the ability of liposomes functionalised with a mannose-6-phosphate to reach lysosomes; in this research we compare the behaviour of M6P-modified and non-functionalised liposomes in MCF7 tumour cell and in HDF normal cells. With this aim, we first demonstrated by Western blotting the overexpression of mannose-6-phosphate/insulin-like growth factor (M6P/IGF-II) receptor in MCF7. Then, we prepared calcein-loaded liposomes and we revealed the increased uptake of M6P-functionalised liposomes in MCF7 cells respect to HDF cells by flow cytometry analysis. Finally, we loaded functionalised and not functionalised liposomes with N-hexanoyl-d-erythro-sphingosine (C6Cer), able to initiate LMP-induced apoptosis; after having studied the stability of both vesicles in the presence of serum by Dynamic Light Scattering and Spectrophotometric turbidity measurements, we showed that ceramide-loaded M6P-liposomes significantly increased apoptosis in MCF7 with respect to HDF cells.
Collapse
Affiliation(s)
- Cristina Minnelli
- a Department of Life and Environmental Sciences , Università Politecnica delle Marche , Ancona , Italy
| | - Laura Cianfruglia
- b Department of Clinical Sciences, Section of Biochemistry, Biology and Physics , Università Politecnica delle Marche , Ancona , Italy
| | - Emiliano Laudadio
- a Department of Life and Environmental Sciences , Università Politecnica delle Marche , Ancona , Italy
| | - Roberta Galeazzi
- a Department of Life and Environmental Sciences , Università Politecnica delle Marche , Ancona , Italy
| | - Michela Pisani
- c Department of Materials, Environmental Sciences and Urban Planning , Università Politecnica delle Marche , Ancona , Italy
| | - Emanuela Crucianelli
- a Department of Life and Environmental Sciences , Università Politecnica delle Marche , Ancona , Italy
| | - Davide Bizzaro
- a Department of Life and Environmental Sciences , Università Politecnica delle Marche , Ancona , Italy
| | - Tatiana Armeni
- b Department of Clinical Sciences, Section of Biochemistry, Biology and Physics , Università Politecnica delle Marche , Ancona , Italy
| | - Giovanna Mobbili
- a Department of Life and Environmental Sciences , Università Politecnica delle Marche , Ancona , Italy
| |
Collapse
|
32
|
Singh MS, Tammam SN, Shetab Boushehri MA, Lamprecht A. MDR in cancer: Addressing the underlying cellular alterations with the use of nanocarriers. Pharmacol Res 2017; 126:2-30. [PMID: 28760489 DOI: 10.1016/j.phrs.2017.07.023] [Citation(s) in RCA: 44] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/17/2017] [Revised: 06/29/2017] [Accepted: 07/26/2017] [Indexed: 01/02/2023]
Abstract
Multidrug resistance (MDR) is associated with a wide range of pathological changes at different cellular and intracellular levels. Nanoparticles (NPs) have been extensively exploited as the carriers of MDR reversing payloads to resistant tumor cells. However, when properly formulated in terms of chemical composition and physicochemical properties, NPs can serve as beyond delivery systems and help overcome MDR even without carrying a load of chemosensitizers or MDR reversing molecular cargos. Whether serving as drug carriers or beyond, a wise design of the nanoparticulate systems to overcome the cellular and intracellular alterations underlying the resistance is imperative. Within the current review, we will initially discuss the cellular changes occurring in resistant cells and how such changes lead to chemotherapy failure and cancer cell survival. We will then focus on different mechanisms through which nanosystems with appropriate chemical composition and physicochemical properties can serve as MDR reversing units at different cellular and intracellular levels according to the changes that underlie the resistance. Finally, we will conclude by discussing logical grounds for a wise and rational design of MDR reversing nanoparticulate systems to improve the cancer therapeutic approaches.
Collapse
Affiliation(s)
- Manu S Singh
- Department of Pharmaceutical Technology and Biopharmceutics, University of Bonn, Germany
| | - Salma N Tammam
- Department of Pharmaceutical Technology and Biopharmceutics, University of Bonn, Germany; Department of Pharmaceutical Technology, German University of Cairo, Egypt
| | | | - Alf Lamprecht
- Department of Pharmaceutical Technology and Biopharmceutics, University of Bonn, Germany; Laboratory of Pharmaceutical Engineering (EA4267), University of Franche-Comté, Besançon, France.
| |
Collapse
|
33
|
Dany M. Sphingosine metabolism as a therapeutic target in cutaneous melanoma. Transl Res 2017; 185:1-12. [PMID: 28528915 DOI: 10.1016/j.trsl.2017.04.005] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/15/2017] [Revised: 03/26/2017] [Accepted: 04/25/2017] [Indexed: 12/19/2022]
Abstract
Melanoma is by far the most aggressive type of skin cancer with a poor prognosis in its advanced stages. Understanding the mechanisms involved in melanoma pathogenesis, response, and resistance to treatment has gained a lot of attention worldwide. Recently, the role of sphingolipid metabolism has been studied in cutaneous melanoma. Sphingolipids are bioactive lipid effector molecules involved in the regulation of various cellular signaling pathways such as inflammation, cancer cell proliferation, death, senescence, and metastasis. Recent studies suggest that sphingolipid metabolism impacts melanoma pathogenesis and is a potential therapeutic target. This review focuses on defining the role of sphingolipid metabolism in melanoma carcinogenesis, discussing sphingolipid-based therapeutic approaches, and highlighting the areas that require more extensive research.
Collapse
Affiliation(s)
- Mohammed Dany
- College of Medicine, Medical University of South Carolina, Charleston, SC.
| |
Collapse
|
34
|
Lee JJ, Saiful Yazan L, Che Abdullah CA. A review on current nanomaterials and their drug conjugate for targeted breast cancer treatment. Int J Nanomedicine 2017; 12:2373-2384. [PMID: 28392694 PMCID: PMC5376210 DOI: 10.2147/ijn.s127329] [Citation(s) in RCA: 64] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022] Open
Abstract
Breast cancer is the most common malignancy worldwide, especially among women, with substantial after-treatment effects. The survival rates of breast cancer have decreased over the years even with the existence of various therapeutic strategies, specifically, chemotherapy. Clinical drugs administered for breast cancer appear to be non-targeting to specific cancer sites leading to severe side effects and potentially harming healthy cells instead of just killing cancer cells. This leads to the need for designing a targeted drug delivery system. Nanomaterials, both organic and inorganic, are potential drug nanocarriers with the ability of targeting, imaging and tracking. Various types of nanomaterials have been actively researched together with their drug conjugate. In this review, we focus on selected nanomaterials, namely solid-lipid, liposomal, polymeric, magnetic nanoparticles, quantum dots, and carbon nanotubes and their drug conjugates, for breast cancer studies. Their advantages, disadvantages and previously conducted studies were highlighted.
Collapse
|
35
|
Ma YY, Mou XZ, Ding YH, Zou H, Huang DS. Delivery systems of ceramide in targeted cancer therapy: ceramide alone or in combination with other anti-tumor agents. Expert Opin Drug Deliv 2016; 13:1397-406. [PMID: 27168034 DOI: 10.1080/17425247.2016.1188803] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Affiliation(s)
- Ying-Yu Ma
- Clinical Research Institute, Zhejiang Provincial People’s Hospital, Hangzhou, China
- Medical School and Jiangsu Laboratory of Molecular Medicine, Nanjing University, Nanjing, China
| | - Xiao-Zhou Mou
- Clinical Research Institute, Zhejiang Provincial People’s Hospital, Hangzhou, China
| | - Ya-Hui Ding
- Department of Cardiology, Zhejiang Provincial People’s Hospital, Hangzhou, China
| | - Hai Zou
- Department of Cardiology, Zhejiang Provincial People’s Hospital, Hangzhou, China
| | - Dong-Sheng Huang
- Clinical Research Institute, Zhejiang Provincial People’s Hospital, Hangzhou, China
- Department of Hepatobiliary Surgery, Zhejiang Provincial People’s Hospital, Hangzhou, China
| |
Collapse
|
36
|
Kitatani K, Usui T, Sriraman SK, Toyoshima M, Ishibashi M, Shigeta S, Nagase S, Sakamoto M, Ogiso H, Okazaki T, Hannun YA, Torchilin VP, Yaegashi N. Ceramide limits phosphatidylinositol-3-kinase C2β-controlled cell motility in ovarian cancer: potential of ceramide as a metastasis-suppressor lipid. Oncogene 2015; 35:2801-12. [PMID: 26364609 PMCID: PMC4791218 DOI: 10.1038/onc.2015.330] [Citation(s) in RCA: 40] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2014] [Revised: 06/19/2015] [Accepted: 07/17/2015] [Indexed: 12/15/2022]
Abstract
Targeting cell motility, which is required for dissemination and metastasis, has therapeutic potential for ovarian cancer metastasis, and regulatory mechanisms of cell motility need to be uncovered for developing novel therapeutics. Invasive ovarian cancer cells spontaneously formed protrusions, such as lamellipodia, which are required for generating locomotive force in cell motility. Short interfering RNA screening identified class II phosphatidylinositol 3-kinase C2β (PI3KC2β) as the predominant isoform of PI3K involved in lamellipodia formation of ovarian cancer cells. The bioactive sphingolipid ceramide has emerged as an antitumorigenic lipid, and treatment with short-chain C6-ceramide decreased the number of ovarian cancer cells with PI3KC2β-driven lamellipodia. Pharmacological analysis demonstrated that long-chain ceramide regenerated from C6-ceramide through the salvage/recycling pathway, at least in part, mediated the action of C6-ceramide. Mechanistically, ceramide was revealed to interact with the PIK-catalytic domain of PI3KC2β and affect its compartmentalization, thereby suppressing PI3KC2β activation and its driven cell motility. Ceramide treatment also suppressed cell motility promoted by epithelial growth factor, which is a prometastatic factor. To examine the role of ceramide in ovarian cancer metastasis, ceramide liposomes were employed and confirmed to suppress cell motility in vitro. Ceramide liposomes had an inhibitory effect on peritoneal metastasis in a murine xenograft model of human ovarian cancer. Metastasis of PI3KC2β knocked-down cells was insensitive to treatment with ceramide liposomes, suggesting specific involvement of ceramide interaction with PI3KC2β in metastasis suppression. Our study identified ceramide as a bioactive lipid that limits PI3KC2β-governed cell motility, and ceramide is proposed to serve as a metastasis-suppressor lipid in ovarian cancer. These findings could be translated into developing ceramide-based therapy for metastatic diseases.
Collapse
Affiliation(s)
- K Kitatani
- Tohoku Medical Megabank Organization, Tohoku University Graduate School of Medicine, Tohoku University, Sendai, Japan.,Department of Obstetrics and Gynecology, Tohoku University Graduate School of Medicine, Tohoku University, Sendai, Japan
| | - T Usui
- Tohoku Medical Megabank Organization, Tohoku University Graduate School of Medicine, Tohoku University, Sendai, Japan
| | - S K Sriraman
- Department of Pharmaceutical Sciences, Center for Pharmaceutical Biotechnology and Nanomedicine, Northeastern University, Boston, MA, USA
| | - M Toyoshima
- Department of Obstetrics and Gynecology, Tohoku University Graduate School of Medicine, Tohoku University, Sendai, Japan
| | - M Ishibashi
- Department of Obstetrics and Gynecology, Tohoku University Graduate School of Medicine, Tohoku University, Sendai, Japan
| | - S Shigeta
- Department of Obstetrics and Gynecology, Tohoku University Graduate School of Medicine, Tohoku University, Sendai, Japan
| | - S Nagase
- Department of Obstetrics and Gynecology, Yamagata University, Yamagata, Japan
| | - M Sakamoto
- Department of Obstetrics and Gynecology, Tohoku University Graduate School of Medicine, Tohoku University, Sendai, Japan
| | - H Ogiso
- Department of Life Science, Medical Research Institute, Kanazawa Medical University, Ishikawa, Japan
| | - T Okazaki
- Department of Life Science, Medical Research Institute, Kanazawa Medical University, Ishikawa, Japan.,Department of Medicine, Division of Hematology/Immunology, Kanazawa Medical University, Ishikawa, Japan
| | - Y A Hannun
- Stony Brook Cancer Center and Department of Medicine, Stony Brook University, Stony Brook, NY, USA
| | - V P Torchilin
- Department of Pharmaceutical Sciences, Center for Pharmaceutical Biotechnology and Nanomedicine, Northeastern University, Boston, MA, USA
| | - N Yaegashi
- Tohoku Medical Megabank Organization, Tohoku University Graduate School of Medicine, Tohoku University, Sendai, Japan.,Department of Obstetrics and Gynecology, Tohoku University Graduate School of Medicine, Tohoku University, Sendai, Japan
| |
Collapse
|
37
|
C6-ceramide nanoliposome suppresses tumor metastasis by eliciting PI3K and PKCζ tumor-suppressive activities and regulating integrin affinity modulation. Sci Rep 2015; 5:9275. [PMID: 25792190 PMCID: PMC4366857 DOI: 10.1038/srep09275] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2014] [Accepted: 02/02/2015] [Indexed: 01/07/2023] Open
Abstract
Nanoliposomal formulation of C6-ceramide, a proapoptotic sphingolipid metabolite, presents an effective way to treat malignant tumor. Here, we provide evidence that acute treatment (30 min) of melanoma and breast cancer cells with nanoliposomal C6-ceramide (NaL-C6) may suppress cell migration without inducing cell death. By employing a novel flow migration assay, we demonstrated that NaL-C6 decreased tumor extravasation under shear conditions. Compared with ghost nanoliposome, NaL-C6 triggered phosphorylation of PI3K and PKCζ and dephosphorylation of PKCα. Concomitantly, activated PKCζ translocated into cell membrane. siRNA knockdown or pharmacological inhibition of PKCζ or PI3K rescued NaL-C6-mediated suppression of tumor migration. By inducing dephosphorylation of paxillin, PKCζ was responsible for NaL-C6-mediated stress fiber depolymerization and focal adhesion disassembly in the metastatic tumor cells. PKCζ and PI3K regulated cell shear-resistant adhesion in a way that required integrin αvβ3 affinity modulation. In conclusion, we identified a novel role of acute nanoliposomal ceramide treatment in reducing integrin affinity and inhibiting melanoma metastasis by conferring PI3K and PKCζ tumor-suppressive activities.
Collapse
|
38
|
Haakenson JK, Khokhlatchev AV, Choi YJ, Linton SS, Zhang P, Zaki PM, Fu C, Cooper TK, Manni A, Zhu J, Fox TE, Dong C, Kester M. Lysosomal degradation of CD44 mediates ceramide nanoliposome-induced anoikis and diminished extravasation in metastatic carcinoma cells. J Biol Chem 2015; 290:8632-43. [PMID: 25681441 DOI: 10.1074/jbc.m114.609677] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
The ceramide nanoliposome (CNL) has shown promise in being able to treat a variety of primary tumors. However, its potential for treating metastatic cancer remains unknown. In this study, we demonstrate that CNL increases anoikis while preventing cancer cell extravasation under both static and physiological fluid flow conditions. Mechanistically, CNL limits metastases by decreasing CD44 protein levels in human breast and pancreatic cancer cells via lysosomal degradation of CD44, independent of palmitoylation or proteasome targeting. siRNA down-regulation of CD44 mimics CNL-induced anoikis and diminished extravasation of cancer cells. Taken together, our data indicate that ceramide limits CD44-dependent cancer cell migration, suggesting that CNL could be used to prevent and treat solid tumor metastasis.
Collapse
Affiliation(s)
| | - Andrei V Khokhlatchev
- the Department of Pharmacology, University of Virginia, Charlottesville, Virginia 22908, and
| | - Younhee J Choi
- the Department of Pharmacology, University of Virginia, Charlottesville, Virginia 22908, and
| | | | - Pu Zhang
- the Department of Bioengineering, Pennsylvania State University, State College, Pennsylvania 16801
| | - Peter M Zaki
- the Department of Bioengineering, Pennsylvania State University, State College, Pennsylvania 16801
| | - Changliang Fu
- the Department of Bioengineering, Pennsylvania State University, State College, Pennsylvania 16801
| | | | | | - Junjia Zhu
- Public Health Sciences, Pennsylvania State University College of Medicine, Hershey, Pennsylvania 17033
| | - Todd E Fox
- the Department of Pharmacology, University of Virginia, Charlottesville, Virginia 22908, and
| | - Cheng Dong
- the Department of Bioengineering, Pennsylvania State University, State College, Pennsylvania 16801
| | - Mark Kester
- From the Departments of Pharmacology, the Department of Pharmacology, University of Virginia, Charlottesville, Virginia 22908, and
| |
Collapse
|
39
|
Dany M, Ogretmen B. Ceramide induced mitophagy and tumor suppression. BIOCHIMICA ET BIOPHYSICA ACTA-MOLECULAR CELL RESEARCH 2015; 1853:2834-45. [PMID: 25634657 DOI: 10.1016/j.bbamcr.2014.12.039] [Citation(s) in RCA: 89] [Impact Index Per Article: 8.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/09/2014] [Revised: 12/09/2014] [Accepted: 12/25/2014] [Indexed: 12/11/2022]
Abstract
Sphingolipids are bioactive lipid effectors, which are involved in the regulation of various cellular signaling pathways. Sphingolipids play essential roles in controlling cell inflammation, proliferation, death, migration, senescence, metastasis and autophagy. Alterations in sphingolipid metabolism have been also implicated in many human cancers. Macroautophagy (referred to here as autophagy) is a form of nonselective sequestering of cytosolic materials by double membrane structures, autophagosomes, which can be either protective or lethal for cells. Ceramide, a central molecule of sphingolipid metabolism is involved in the regulation of autophagy at various levels, including the induction of lethal mitophagy, a selective autophagy process to target and eliminate damaged mitochondria. In this review, we focused on recent studies with regard to the regulation of autophagy, in particular lethal mitophagy, by ceramide, and aimed at providing discussion points for various context-dependent roles and mechanisms of action of ceramide in controlling mitophagy.
Collapse
Affiliation(s)
- Mohammed Dany
- Department of Biochemistry and Molecular Biology, Medical University of South Carolina, Charleston, SC 29425, USA; Hollings Cancer Center, Medical University of South Carolina, 86 Jonathan Lucas Street, Charleston, SC 29425, USA
| | - Besim Ogretmen
- Department of Biochemistry and Molecular Biology, Medical University of South Carolina, Charleston, SC 29425, USA; Hollings Cancer Center, Medical University of South Carolina, 86 Jonathan Lucas Street, Charleston, SC 29425, USA.
| |
Collapse
|
40
|
Synergistic enhancement of cancer therapy using a combination of ceramide and docetaxel. Int J Mol Sci 2014; 15:4201-20. [PMID: 24619193 PMCID: PMC3975392 DOI: 10.3390/ijms15034201] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2014] [Revised: 02/19/2014] [Accepted: 02/21/2014] [Indexed: 12/12/2022] Open
Abstract
Ceramide (CE)-based combination therapy (CE combination) as a novel therapeutic strategy has attracted great attention in the field of anti-cancer therapy. The principal purposes of this study were to investigate the synergistic effect of CE in combination with docetaxel (DTX) (CE + DTX) and to explore the synergy mechanisms of CE + DTX. The 3-(4,5-Dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide (MTT) and combination index (CI) assay showed that simultaneous administration of CE and DTX with a molar ratio of 0.5:1 could generate the optimal synergistic effect on murine malignant melanoma cell (B16, CI = 0.31) and human breast carcinoma cell (MCF-7, CI = 0.48). The apoptosis, cell cycle, and cytoskeleton destruction study demonstrated that CE could target and destruct the microfilament actin, subsequently activate Caspase-3 and induce apoptosis. Meanwhile, DTX could target and disrupt the microtubules cytoskeleton, leading to a high proportion of cancer cells in G2/M-phase arrest. Moreover, CE plus DTX could cause a synergistic destruction of cytoskeleton, which resulted in a significantly higher apoptosis and a significantly higher arrest in G2/M arrest comparing with either agent alone (p < 0.01). The in vivo antitumor study evaluated in B16 tumor-bearing mice also validated the synergistic effects. All these results suggested that CE could enhance the antitumor activity of DTX in a synergistic manner, which suggest promising application prospects of CE + DTX combination treatment.
Collapse
|
41
|
Abstract
Ceramide serves as a central mediator in sphingolipid metabolism and signaling pathways, regulating many fundamental cellular responses. It is referred to as a 'tumor suppressor lipid', since it powerfully potentiates signaling events that drive apoptosis, cell cycle arrest, and autophagic responses. In the typical cancer cell, ceramide levels and signaling are usually suppressed by overexpression of ceramide-metabolizing enzymes or downregulation of ceramide-generating enzymes. However, chemotherapeutic drugs as well as radiotherapy increase intracellular ceramide levels, while exogenously treating cancer cells with short-chain ceramides leads to anticancer effects. All evidence currently points to the fact that the upregulation of ceramide levels is a promising anticancer strategy. In this review, we exhibit many anticancer ceramide analogs as downstream receptor agonists and ceramide-metabolizing enzyme inhibitors.
Collapse
|
42
|
Dhule SS, Penfornis P, He J, Harris MR, Terry T, John V, Pochampally R. The combined effect of encapsulating curcumin and C6 ceramide in liposomal nanoparticles against osteosarcoma. Mol Pharm 2014; 11:417-27. [PMID: 24380633 DOI: 10.1021/mp400366r] [Citation(s) in RCA: 68] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
This study examines the antitumor potential of curcumin and C6 ceramide (C6) against osteosarcoma (OS) cell lines when both are encapsulated in the bilayer of liposomal nanoparticles. Three liposomal formulations were prepared: curcumin liposomes, C6 liposomes and C6-curcumin liposomes. Curcumin in combination with C6 showed 1.5 times enhanced cytotoxic effect in the case of MG-63 and KHOS OS cell lines, in comparison with curcumin liposomes alone. Importantly, C6-curcumin liposomes were found to be less toxic on untransformed primary human cells (human mesenchymal stem cells) in comparison to OS cell lines. In addition, cell cycle assays on a KHOS cell line after treatment revealed that curcumin only liposomes induced G2/M arrest by upregulation of cyclin B1, while C6 only liposomes induced G1 arrest by downregulation of cyclin D1. C6-curcumin liposomes induced G2/M arrest and showed a combined effect in the expression levels of cyclin D1 and cyclin B1. The efficiency of the preparations was tested in vivo using a human osteosarcoma xenograft assay. Using pegylated liposomes to increase the plasma half-life and tagging with folate (FA) for targeted delivery in vivo, a significant reduction in tumor size was observed with C6-curcumin-FA liposomes. The encapsulation of two water insoluble drugs, curcumin and C6, in the lipid bilayer of liposomes enhances the cytotoxic effect and validates the potential of combined drug therapy.
Collapse
Affiliation(s)
- Santosh S Dhule
- Department of Chemical and Biomolecular Engineering, Tulane University , New Orleans, Louisiana 70118, United States
| | | | | | | | | | | | | |
Collapse
|
43
|
PLGA/liposome hybrid nanoparticles for short-chain ceramide delivery. Pharm Res 2013; 31:684-93. [PMID: 24065591 DOI: 10.1007/s11095-013-1190-5] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2013] [Accepted: 08/09/2013] [Indexed: 12/29/2022]
Abstract
PURPOSE Rapid premature release of lipophilic drugs from liposomal lipid bilayer to plasma proteins and biological membranes is a challenge for targeted drug delivery. The purpose of this study is to reduce premature release of lipophilic short-chain ceramides by encapsulating ceramides into liposomal aqueous interior with the aid of poly (lactic-coglycolicacid) (PLGA). METHODS BODIPY FL labeled ceramide (FL-ceramide) and BODIPY-TR labeled ceramide (TR-ceramide) were encapsulated into carboxy-terminated PLGA nanoparticles. The negatively charged PLGA nanoparticles were then encapsulated into cationic liposomes to obtain PLGA/liposome hybrids. As a control, FL-ceramide and/or TR ceramide co-loaded liposomes without PLGA were prepared. The release of ceramides from PLGA/liposome hybrids and liposomes in rat plasma, cultured MDA-MB-231 cells, and rat blood circulation was compared using fluorescence resonance energy transfer (FRET) between FL-ceramide (donor) and TR-ceramide (acceptor). RESULTS FRET analysis showed that FL-ceramide and TR-ceramide in liposomal lipid bilayer were rapidly released during incubation with rat plasma. In contrast, the FL-ceramide and TR-ceramide in PLGA/liposome hybrids showed extended release. FRET images of cells revealed that ceramides in liposomal bilayer were rapidly transferred to cell membranes. In contrast, ceramides in PLGA/liposome hybrids were internalized into cells with nanoparticles simultaneously. Upon intravenous administration to rats, ceramides encapsulated in liposomal bilayer were completely released in 2 min. In contrast, ceramides encapsulated in the PLGA core were retained in PLGA/liposome hybrids for 4 h. CONCLUSIONS The PLGA/liposome hybrid nanoparticles reduced in vitro and in vivo premature release of ceramides and offer a viable platform for targeted delivery of lipophilic drugs.
Collapse
|
44
|
Adiseshaiah PP, Clogston JD, McLeland CB, Rodriguez J, Potter TM, Neun BW, Skoczen SL, Shanmugavelandy SS, Kester M, Stern ST, McNeil SE. Synergistic combination therapy with nanoliposomal C6-ceramide and vinblastine is associated with autophagy dysfunction in hepatocarcinoma and colorectal cancer models. Cancer Lett 2013; 337:254-65. [PMID: 23664889 PMCID: PMC3722309 DOI: 10.1016/j.canlet.2013.04.034] [Citation(s) in RCA: 60] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2012] [Revised: 03/14/2013] [Accepted: 04/29/2013] [Indexed: 11/24/2022]
Abstract
Autophagy, a catabolic survival pathway, is gaining attention as a potential target in cancer. In human liver and colon cancer cells, treatment with an autophagy inducer, nanoliposomal C6-ceramide, in combination with the autophagy maturation inhibitor, vinblastine, synergistically enhanced apoptotic cell death. Combination treatment resulted in a marked increase in autophagic vacuole accumulation and decreased autophagy maturation, without diminution of the autophagy flux protein P62. In a colon cancer xenograft model, a single intravenous injection of the drug combination significantly decreased tumor growth in comparison to the individual treatments. Most importantly, the combination treatment did not result in increased toxicity as assessed by body weight loss. The mechanism of combination treatment-induced cell death both in vitro and in vivo appeared to be apoptosis. Supportive of autophagy flux blockade as the underlying synergy mechanism, treatment with other autophagy maturation inhibitors, but not autophagy initiation inhibitors, were similarly synergistic with C6-ceramide. Additionally, knockout of the autophagy protein Beclin-1 suppressed combination treatment-induced apoptosis in vitro. In conclusion, in vitro and in vivo data support a synergistic antitumor activity of the nanoliposomal C6-ceramide and vinblastine combination, potentially mediated by an autophagy mechanism.
Collapse
Affiliation(s)
- Pavan P. Adiseshaiah
- Nanotechnology Characterization Laboratory, Advanced Technology Program, SAIC-Frederick, Inc., Frederick National Laboratory for Cancer Research, Frederick, MD 21702
| | - Jeffrey D. Clogston
- Nanotechnology Characterization Laboratory, Advanced Technology Program, SAIC-Frederick, Inc., Frederick National Laboratory for Cancer Research, Frederick, MD 21702
| | - Christopher B. McLeland
- Nanotechnology Characterization Laboratory, Advanced Technology Program, SAIC-Frederick, Inc., Frederick National Laboratory for Cancer Research, Frederick, MD 21702
| | - Jamie Rodriguez
- Nanotechnology Characterization Laboratory, Advanced Technology Program, SAIC-Frederick, Inc., Frederick National Laboratory for Cancer Research, Frederick, MD 21702
| | - Timothy M. Potter
- Nanotechnology Characterization Laboratory, Advanced Technology Program, SAIC-Frederick, Inc., Frederick National Laboratory for Cancer Research, Frederick, MD 21702
| | - Barry W. Neun
- Nanotechnology Characterization Laboratory, Advanced Technology Program, SAIC-Frederick, Inc., Frederick National Laboratory for Cancer Research, Frederick, MD 21702
| | - Sarah L. Skoczen
- Nanotechnology Characterization Laboratory, Advanced Technology Program, SAIC-Frederick, Inc., Frederick National Laboratory for Cancer Research, Frederick, MD 21702
| | | | - Mark Kester
- Department of Pharmacology, Pennsylvania State University College of Medicine, Hershey, PA 17033
| | - Stephan T. Stern
- Nanotechnology Characterization Laboratory, Advanced Technology Program, SAIC-Frederick, Inc., Frederick National Laboratory for Cancer Research, Frederick, MD 21702
| | - Scott E. McNeil
- Nanotechnology Characterization Laboratory, Advanced Technology Program, SAIC-Frederick, Inc., Frederick National Laboratory for Cancer Research, Frederick, MD 21702
| |
Collapse
|
45
|
Abstract
Ceramide, a bioactive sphingolipid, is now at the forefront of cancer research. Classically, ceramide is thought to induce death, growth inhibition, and senescence in cancer cells. However, it is now clear that this simple picture of ceramide no longer holds true. Recent studies suggest that there are diverse functions of endogenously generated ceramides, which seem to be context dependent, regulated by subcellular/membrane localization and presence/absence of direct targets of these lipid molecules. For example, different fatty-acid chain lengths of ceramide, such as C(16)-ceramide that can be generated by ceramide synthase 6 (CerS6), have been implicated in cancer cell proliferation, whereas CerS1-generated C(18)-ceramide mediates cell death. The dichotomy of ceramides' function in cancer cells makes some of the metabolic enzymes of ceramide synthesis potential drug targets (such as Cers6) to prevent cancer growth in breast and head and neck cancers. Conversely, activation of CerS1 could be a new therapeutic option for the development of novel strategies against lung and head and neck cancers. This chapter focuses on recent discoveries about the mechanistic details of mainly de novo-generated ceramides and their signaling functions in cancer pathogenesis, and about how these mechanistic information can be translated into clinically relevant therapeutic options for the treatment of cancer.
Collapse
|
46
|
Sukumaran P, Lönnfors M, Långvik O, Pulli I, Törnquist K, Slotte JP. Complexation of c6-ceramide with cholesteryl phosphocholine - a potent solvent-free ceramide delivery formulation for cells in culture. PLoS One 2013; 8:e61290. [PMID: 23620740 PMCID: PMC3631171 DOI: 10.1371/journal.pone.0061290] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2012] [Accepted: 03/06/2013] [Indexed: 12/15/2022] Open
Abstract
Ceramides are potent bioactive molecules in cells. However, they are very hydrophobic molecules, and difficult to deliver efficiently to cells. We have made fluid bilayers from a short-chain D-erythro-ceramide (C6-Cer) and cholesteryl phosphocholine (CholPC), and have used this as a formulation to deliver ceramide to cells. C6-Cer complexed with CholPC led to much larger biological effects in cultured cells (rat thyroid FRTL-5 and human HeLa cells in culture) compared to C6-Cer dissolved in dimethyl sulfoxide (DMSO). Inhibition of cell proliferation and induction of apoptosis was significantly more efficient by C6-Cer/CholPC compared to C6-Cer dissolved in DMSO. C6-Cer/CholPC also permeated cell membranes and caused mitochondrial Ca2+ influx more efficiently than C6-Cer in DMSO. Even though CholPC was taken up by cells to some extent (from C6-Cer/CholPC bilayers), and was partially hydrolyzed to free cholesterol (about 9%), none of the antiproliferative effects were due to CholPC or excess cholesterol. The ceramide effect was not limited to D-erythro-C6-Cer, since L-erythro-C6-Cer and D-erythro-C6-dihydroCer also inhibited cell priolifereation and affected Ca2+ homeostasis. We conclude that C6-Cer complexed to CholPC increased the bioavailability of the short-chain ceramide for cells, and potentiated its effects in comparison to solvent-dissolved C6-Cer. This new ceramide formulation appears to be superior to previous solvent delivery approaches, and may even be useful with longer-chain ceramides.
Collapse
Affiliation(s)
| | - Max Lönnfors
- Biochemistry, Department of Biosciences, Åbo Akademi University, Turku, Finland
| | - Otto Långvik
- Laboratory of Organic Chemistry, Department of Natural Sciences, Åbo Akademi University, Turku, Finland
| | - Ilari Pulli
- Cell Biology, Åbo Akademi University, Turku, Finland
| | - Kid Törnquist
- Cell Biology, Åbo Akademi University, Turku, Finland
- Minerva Foundation Institute of Medical Research, Biomedicum Helsinki, Helsinki, Finland
- * E-mail: (KT); (JPS)
| | - J. Peter Slotte
- Biochemistry, Department of Biosciences, Åbo Akademi University, Turku, Finland
- * E-mail: (KT); (JPS)
| |
Collapse
|
47
|
Yao L, Daniels J, Wijesinghe D, Andreev OA, Reshetnyak YK. pHLIP®-mediated delivery of PEGylated liposomes to cancer cells. J Control Release 2013; 167:228-37. [PMID: 23416366 DOI: 10.1016/j.jconrel.2013.01.037] [Citation(s) in RCA: 64] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2012] [Accepted: 01/29/2013] [Indexed: 01/12/2023]
Abstract
We develop a method for pH-dependent fusion between liposomes and cellular membranes using pHLIP® (pH Low Insertion Peptide), which inserts into lipid bilayer of membrane only at low pH. Previously we establish the molecular mechanism of peptide action and show that pHLIP can target acidic diseased tissue. Here we investigate how coating of PEGylated liposomes with pHLIP might affect liposomal uptake by cells. The presence of pHLIP on the surface of PEGylated-liposomes enhanced membrane fusion and lipid exchange in a pH dependent fashion, leading to increase of cellular uptake and payload release, and inhibition of cell proliferation by liposomes containing ceramide. A novel type of pH-sensitive, "fusogenic" pHLIP-liposomes was developed, which could be used to selectively deliver various diagnostic and therapeutic agents to acidic diseased cells.
Collapse
Affiliation(s)
- Lan Yao
- Physics Department, University of Rhode Island, 2 Lippitt Road, Kingston, RI 02881, USA
| | | | | | | | | |
Collapse
|
48
|
Leblanc F, Zhang D, Liu X, Loughran TP. Large granular lymphocyte leukemia: from dysregulated pathways to therapeutic targets. Future Oncol 2013; 8:787-801. [PMID: 22830400 DOI: 10.2217/fon.12.75] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023] Open
Abstract
Large granular lymphocyte (LGL) leukemia is a clonal lymphoproliferative disorder of cytotoxic lymphocytes characterized by an expansion of CD3(+) cytotoxic T lymphocytes or CD3(-) natural killer cells. Patients present with various cytopenias including neutropenia, anemia and thrombocytopenia. In addition, there is an association of T-cell large granular lymphocytic leukemia with rheumatoid arthritis. It is believed that LGL leukemia begins as an antigen-driven immune response with subsequent constitutive activation of cytotoxic T lymphocytes or natural killer cells through PDGF and IL-15 contributing to their survival. Consequently, this leads to a dysregulation of apoptosis and dysfunction of the activation-induced cell death pathway. Treatment of LGL leukemia is based on a low-dose immunosuppressive regimen using methotrexate or cyclophosphamide. However, no standard of therapy has been established, as large prospective trials have not been conducted. In addition, some patients are refractory to treatment. The lack of a curative therapy for LGL leukemia means that new treatment options are needed. Insight into the various dysregulated signaling pathways in LGL leukemia may provide novel therapeutic treatment modalities.
Collapse
Affiliation(s)
- Francis Leblanc
- Penn State Hershey Cancer Institute, Experimental Therapeutics, Room 4427, 500 University Drive, PO Box 850, Hershey, PA 17033-0850, USA
| | | | | | | |
Collapse
|
49
|
Metronomic ceramide analogs inhibit angiogenesis in pancreatic cancer through up-regulation of caveolin-1 and thrombospondin-1 and down-regulation of cyclin D1. Neoplasia 2013; 14:833-45. [PMID: 23019415 DOI: 10.1593/neo.12772] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2012] [Revised: 07/30/2012] [Accepted: 07/30/2012] [Indexed: 02/07/2023] Open
Abstract
AIMS To evaluate the antitumor and antiangiogenic activity of metronomic ceramide analogs and their relevant molecular mechanisms. METHODS Human endothelial cells [human dermal microvascular endothelial cells and human umbilical vascular endothelial cell (HUVEC)] and pancreatic cancer cells (Capan-1 and MIA PaCa-2) were treated with the ceramide analogs (C2, AL6, C6, and C8), at low concentrations for 144 hours to evaluate any antiproliferative and proapoptotic effects and inhibition of migration and to measure the expression of caveolin-1 (CAV-1) and thrombospondin-1 (TSP-1) mRNAs by real-time reverse transcription-polymerase chain reaction. Assessment of extracellular signal-regulated kinases 1 and 2 (ERK1/2) and Akt phosphorylation and of CAV-1 and cyclin D1 protein expression was performed by ELISA. Maximum tolerated dose (MTD) gemcitabine was compared against metronomic doses of the ceramide analogs by evaluating the inhibition of MIA PaCa-2 subcutaneous tumor growth in nude mice. RESULTS Metronomic ceramide analogs preferentially inhibited cell proliferation and enhanced apoptosis in endothelial cells. Low concentrations of AL6 and C2 caused a significant inhibition of HUVEC migration. ERK1/2 and Akt phosphorylation were significantly decreased after metronomic ceramide analog treatment. Such treatment caused the overexpression of CAV-1 and TSP-1 mRNAs and proteins in endothelial cells, whereas cyclin D1 protein levels were reduced. The antiangiogenic and antitumor impact in vivo of metronomic C2 and AL6 regimens was similar to that caused by MTD gemcitabine. CONCLUSIONS Metronomic C2 and AL6 analogs have antitumor and antiangiogenic activity, determining the up-regulation of CAV-1 and TSP-1 and the suppression of cyclin D1.
Collapse
|
50
|
Morad SAF, Madigan JP, Levin JC, Abdelmageed N, Karimi R, Rosenberg DW, Kester M, Shanmugavelandy SS, Cabot MC. Tamoxifen magnifies therapeutic impact of ceramide in human colorectal cancer cells independent of p53. Biochem Pharmacol 2013; 85:1057-65. [PMID: 23353700 DOI: 10.1016/j.bcp.2013.01.015] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2012] [Revised: 01/16/2013] [Accepted: 01/18/2013] [Indexed: 12/28/2022]
Abstract
Poor prognosis in patients with later stage colorectal cancer (CRC) necessitates the search for new treatment strategies. Ceramide, because of its role in orchestrating death cascades in cancer cells, is a versatile alternative. Ceramide can be generated by exposure to chemotherapy or ionizing radiation, or it can be administered in the form of short-chain analogs (C6-ceramide). Because intracellular P-glycoprotein (P-gp) plays a role in catalyzing the conversion of ceramide to higher sphingolipids, we hypothesized that administration of P-gp antagonists with C6-ceramide would magnify cell death cascades. Human CRC cell lines were employed, HCT-15, HT-29, and LoVo. The addition of either tamoxifen, VX-710, verapamil, or cyclosporin A, antagonists of P-gp, enhanced C6-ceramide cytotoxicity in all cell lines. In depth studies with C6-ceramide and tamoxifen in LoVo cells showed the regimen induced PARP cleavage, caspase-dependent apoptosis, mitochondrial membrane permeabilization (MMP), and cell cycle arrest at G1 and G2. At the molecular level, the regimen, but not single agents, induced time-dependent upregulation of tumor suppressor protein p53; however, introduction of a p53 inhibitor staved neither MMP nor apoptosis. Nanoliposomal formulations of C6-ceramide and tamoxifen were also effective, yielding synergistic cell kill. We conclude that tamoxifen is a favorable adjuvant for enhancing C6-ceramide cytotoxicity in CRC, and demonstrates uniquely integrated effects. The high frequency of expression of P-gp in CRC presents an adventitious target for complementing ceramide-based therapies, a strategy that could hold promise for treatment of resistant disease.
Collapse
Affiliation(s)
- Samy A F Morad
- John Wayne Cancer Institute, Santa Monica, CA 90404, USA
| | | | | | | | | | | | | | | | | |
Collapse
|