1
|
Wang R, Yan Q, Liu X, Wu J. Unraveling lipid metabolism reprogramming for overcoming drug resistance in melanoma. Biochem Pharmacol 2024; 223:116122. [PMID: 38467377 DOI: 10.1016/j.bcp.2024.116122] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2024] [Revised: 02/27/2024] [Accepted: 03/07/2024] [Indexed: 03/13/2024]
Abstract
Cutaneous melanoma is the deadliest form of skin cancer, and its incidence is continuing to increase worldwide in the last decades. Traditional therapies for melanoma can easily cause drug resistance, thus the treatment of melanoma remains a challenge. Various studies have focused on reversing the drug resistance. As tumors grow and progress, cancer cells face a constantly changing microenvironment made up of different nutrients, metabolites, and cell types. Multiple studies have shown that metabolic reprogramming of cancer is not static, but a highly dynamic process. There is a growing interest in exploring the relationship between melanoma andmetabolic reprogramming, one of which may belipid metabolism. This review frames the recent research progresses on lipid metabolism in melanoma.In addition, we emphasize the dynamic ability of metabolism during tumorigenesis as a target for improving response to different therapies and for overcoming drug resistance in melanoma.
Collapse
Affiliation(s)
- Ruilong Wang
- Department of Dermatology, Huashan Hospital, Fudan University, Shanghai, China
| | - Qin Yan
- Shanghai General Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China
| | - Xiao Liu
- Department of Dermatology, Huashan Hospital, Fudan University, Shanghai, China.
| | - Jinfeng Wu
- Department of Dermatology, Huashan Hospital, Fudan University, Shanghai, China.
| |
Collapse
|
2
|
Rubinstein JC, Domanskyi S, Sheridan TB, Sanderson B, Park S, Kaster J, Li H, Anczukow O, Herlyn M, Chuang JH. Spatiotemporal profiling defines persistence and resistance dynamics during targeted treatment of melanoma. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.02.02.577085. [PMID: 38370717 PMCID: PMC10871267 DOI: 10.1101/2024.02.02.577085] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/20/2024]
Abstract
Resistance of BRAF-mutant melanomas to targeted therapy arises from the ability of cells to enter a persister state, evade treatment with relative dormancy, and repopulate the tumor when reactivated. Using spatial transcriptomics in patient derived xenograft models, we capture clonal lineage evolution during treatment, finding the persister state to show increased oxidative phosphorylation, decreased proliferation, and increased invasive capacity, with central-to-peripheral gradients. Phylogenetic tracing identifies intrinsic- and acquired-resistance mechanisms (e.g. dual specific phosphatases, Reticulon-4, CDK2) and suggests specific temporal windows of potential therapeutic efficacy. Using deep learning to analyze histopathological slides, we find morphological features of specific cell states, demonstrating that juxtaposition of transcriptomics and histology data enables identification of phenotypically-distinct populations using imaging data alone. In summary, we define state change and lineage selection during melanoma treatment with spatiotemporal resolution, elucidating how choice and timing of therapeutic agents will impact the ability to eradicate resistant clones. Statement of Significance Tumor evolution is accelerated by application of anti-cancer therapy, resulting in clonal expansions leading to dormancy and subsequently resistance, but the dynamics of this process are incompletely understood. Tracking clonal progression during treatment, we identify conserved, global transcriptional changes and local clone-clone and spatial patterns underlying the emergence of resistance.
Collapse
|
3
|
Targeting Protein Translation in Melanoma by Inhibiting EEF-2 Kinase Regulates Cholesterol Metabolism though SREBP2 to Inhibit Tumour Development. Int J Mol Sci 2022; 23:ijms23073481. [PMID: 35408842 PMCID: PMC8998919 DOI: 10.3390/ijms23073481] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2022] [Revised: 03/04/2022] [Accepted: 03/15/2022] [Indexed: 01/13/2023] Open
Abstract
Decreasing the levels of certain proteins has been shown to be important for controlling cancer but it is currently unknown whether proteins could potentially be targeted by the inhibiting of protein synthesis. Under this circumstance, targeting protein translation could preferentially affect certain pathways, which could then be of therapeutic advantage when treating cancer. In this report, eukaryotic elongation factor-2 kinase (EEF2K), which is involved in protein translation, was shown to regulate cholesterol metabolism. Targeting EEF2K inhibited key parts of the cholesterol pathway in cancer cells, which could be rescued by the addition of exogenous cholesterol, suggesting that it is a potentially important pathway modulated by targeting this process. Specifically, targeting EEF2K significantly suppressed tumour cell growth by blocking mRNA translation of the cholesterol biosynthesis transcription factor, sterol regulatory element-binding protein (SREBP) 2, and the proteins it regulates. The process could be rescued by the addition of LDL cholesterol taken into the cells via non-receptor-mediated-uptake, which negated the need for SREBP2 protein. Thus, the levels of SREBP2 needed for cholesterol metabolism in cancer cells are therapeutically vulnerable by targeting protein translation. This is the first report to suggest that targeting EEF2K can be used to modulate cholesterol metabolism to treat cancer.
Collapse
|
4
|
Leelamine Modulates STAT5 Pathway Causing Both Autophagy and Apoptosis in Chronic Myelogenous Leukemia Cells. BIOLOGY 2022; 11:biology11030366. [PMID: 35336740 PMCID: PMC8945775 DOI: 10.3390/biology11030366] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/17/2022] [Revised: 02/23/2022] [Accepted: 02/23/2022] [Indexed: 01/07/2023]
Abstract
Simple Summary Autophagy is a cellular mechanism that is essential for removing misfolded proteins and damaged organelles. Moreover, the aberrant activation of signal transducer and activator of transcription 5 (STAT5), which can regulate cellular survival and homeostasis, has been often observed in different malignancies. In this study, we demonstrate that leelamine inhibits the STAT5 phosphorylation while inducing autophagy as well as apoptosis in chronic myeloid leukemia cells. Leelamine induces autophagy by stimulating the expression of Atg7, beclin-1, and the production of autophagosomes, which leads to substantial inhibition of STAT5 activation. Abstract Leelamine (LEE) has recently attracted significant attention for its growth inhibitory effects against melanoma, breast cancer, and prostate cancer cells; however, its impact on hematological malignancies remains unclear. Here, we first investigate the cytotoxic effects of LEE on several human chronic myeloid leukemia (CML) cells. We noted that LEE stimulated both apoptosis and autophagy in CML cells. In addition, the constitutive activation of signal transducer and activator of transcription 5 (STAT5) was suppressed substantially upon LEE treatment. Moreover, STAT5 knockdown with small interfering RNA (siRNA) increased LEE-induced apoptosis as well as autophagy and affected the levels of various oncogenic proteins. Thus, the targeted mitigation of STAT5 activation by LEE can contribute to its diverse anticancer effects by enhancing two distinct cell death pathways.
Collapse
|
5
|
Zhang KL, Zhu WW, Wang SH, Gao C, Pan JJ, Du ZG, Lu L, Jia HL, Dong QZ, Chen JH, Lu M, Qin LX. Organ-specific cholesterol metabolic aberration fuels liver metastasis of colorectal cancer. Am J Cancer Res 2021; 11:6560-6572. [PMID: 33995676 PMCID: PMC8120208 DOI: 10.7150/thno.55609] [Citation(s) in RCA: 51] [Impact Index Per Article: 12.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2020] [Accepted: 04/09/2021] [Indexed: 01/28/2023] Open
Abstract
Rationale: Metastasis, the development of secondary malignant growth at a distance from a primary tumor, is the main cause of cancer-associated death. However, little is known about how metastatic cancer cells adapt to and colonize in the new organ environment. Here we sought to investigate the functional mechanism of cholesterol metabolic aberration in colorectal carcinoma (CRC) liver metastasis. Methods: The expression of cholesterol metabolism-related genes in primary colorectal tumors (PT) and paired liver metastases (LM) were examined by RT-PCR. The role of SREBP2-dependent cholesterol biosynthesis pathway in cell growth and CRC liver metastasis were determined by SREBP2 silencing in CRC cell lines and experimental metastasis models including, intra-splenic injection models and liver orthotropic injection model. Growth factors treatment and co-culture experiment were performed to reveal the mechanism underlying the up-regulation of SREBP2 in CRC liver metastases. The in vivo efficacy of inhibition of cholesterol biosynthesis pathway by betulin or simvastatin were evaluated in experimental metastasis models. Results: In the present study, we identify a colorectal cancer (CRC) liver metastasis-specific cholesterol metabolic pathway involving the activation of SREBP2-dependent cholesterol biosynthesis, which is required for the colonization and growth of metastatic CRC cells in the liver. Inhibiting this cholesterol biosynthesis pathway suppresses CRC liver metastasis. Mechanically, hepatocyte growth factor (HGF) from liver environment activates SREBP2-dependent cholesterol biosynthesis pathway by activating c-Met/PI3K/AKT/mTOR axis in CRC cells. Conclusion: Our findings support the notion that CRC liver metastases show a specific cholesterol metabolic aberration. Targeting this cholesterol biosynthesis pathway could be a promising treatment for CRC liver metastasis.
Collapse
|
6
|
Chen YC, Dinavahi SS, Feng Q, Gowda R, Ramisetti S, Xia X, LaPenna KB, Chirasani VR, Cho SH, Hafenstein SL, Battu MB, Berg A, Sharma AK, Kirchhausen T, Dokholyan NV, Amin S, He P, Robertson GP. Activating Sphingosine-1-phospahte signaling in endothelial cells increases myosin light chain phosphorylation to decrease endothelial permeability thereby inhibiting cancer metastasis. Cancer Lett 2021; 506:107-119. [PMID: 33600895 DOI: 10.1016/j.canlet.2021.01.004] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2020] [Revised: 12/22/2020] [Accepted: 01/04/2021] [Indexed: 12/23/2022]
Abstract
Targeting the metastatic process to prevent disease dissemination in cancer remains challenging. One step in the metastatic cascade involves cancer cells transiting through the vascular endothelium after inflammation has increased the permeability of this cellular layer. Reducing inflammation-mediated gaps in the vascular endothelium could potentially be used to retard metastasis. This study describes the development of a novel ASR396-containing nanoparticle designed to activate the Sphingosine-1-Phosphate Receptor 1 (S1PR1) in order to tighten the junctions between the endothelial cells lining the vascular endothelium thereby inhibiting metastasis. ASR396 was derived from the S1PR1 agonist SEW2871 through chemical modification enabling the new compound to be loaded into a nanoliposome. ASR396 retained S1PR1 binding activity and the nanoliposomal formulation (nanoASR396) made it systemically bioavailable upon intravenous injection. Studies conducted in microvessels demonstrated that nanoASR396 significantly attenuated inflammatory mediator-induced permeability increase through the S1PR1 activation. Similarly, nanoASR396 inhibited gap formation mediated by inflammatory agents on an endothelial cell monolayer by decreasing levels of phosphorylated myosin light chain protein thereby inhibiting cellular contractility. In animal models, nanoASR396 inhibited lung metastasis by up to 80%, indicating its potential for retarding melanoma metastasis. Thus, a novel bioavailable nanoparticle-based S1PR1 agonist has been developed to negate the effects of inflammatory mediators on the vascular endothelium in order to reduce the metastatic dissemination of cancer cells.
Collapse
Affiliation(s)
- Yu-Chi Chen
- Departments of Pharmacology, The Pennsylvania State University College of Medicine, Hershey, PA, 17033, USA
| | - Saketh S Dinavahi
- Departments of Pharmacology, The Pennsylvania State University College of Medicine, Hershey, PA, 17033, USA
| | - Qilong Feng
- Departments of Cellular and Molecular Physiology, The Pennsylvania State University College of Medicine, Hershey, PA, 17033, USA
| | - Raghavendra Gowda
- Departments of Pharmacology, The Pennsylvania State University College of Medicine, Hershey, PA, 17033, USA
| | - Srinivasa Ramisetti
- Departments of Pharmacology, The Pennsylvania State University College of Medicine, Hershey, PA, 17033, USA
| | - Xinghai Xia
- Departments of Cellular and Molecular Physiology, The Pennsylvania State University College of Medicine, Hershey, PA, 17033, USA
| | - Kyle B LaPenna
- Departments of Cellular and Molecular Physiology, The Pennsylvania State University College of Medicine, Hershey, PA, 17033, USA
| | - Venkat R Chirasani
- Departments of Pharmacology, The Pennsylvania State University College of Medicine, Hershey, PA, 17033, USA
| | - Sung Hyun Cho
- The Huck Institutes of the Life Sciences, Pennsylvania State University, University Park, PA, 16802, USA
| | - Susan L Hafenstein
- Departments of Medicine, The Pennsylvania State University College of Medicine, Hershey, PA, 17033, USA; Department of Biochemistry and Molecular Biology, Pennsylvania State University, University Park, PA, 16802, USA
| | | | - Arthur Berg
- Departments of Public Health Sciences, The Pennsylvania State University College of Medicine, Hershey, PA, 17033, USA
| | - Arun K Sharma
- Departments of Pharmacology, The Pennsylvania State University College of Medicine, Hershey, PA, 17033, USA
| | - Tom Kirchhausen
- Departments of Cell Biology, Harvard Medical School and Program in Cellular and Molecular Medicine at Boston Children's Hospital, MA, 02115, USA
| | - Nikolay V Dokholyan
- Departments of Pharmacology, The Pennsylvania State University College of Medicine, Hershey, PA, 17033, USA; Departments of Biochemistry and Molecular Biology, The Pennsylvania State University College of Medicine, Hershey, PA, 17033, USA
| | - Shantu Amin
- Departments of Pharmacology, The Pennsylvania State University College of Medicine, Hershey, PA, 17033, USA.
| | - Pingnian He
- Departments of Cellular and Molecular Physiology, The Pennsylvania State University College of Medicine, Hershey, PA, 17033, USA.
| | - Gavin P Robertson
- Departments of Pharmacology, The Pennsylvania State University College of Medicine, Hershey, PA, 17033, USA; Departments of Departments of Pathology, The Pennsylvania State University College of Medicine, Hershey, PA, 17033, USA; Departments of Dermatology, The Pennsylvania State University College of Medicine, Hershey, PA, 17033, USA; Departments of Surgery, The Pennsylvania State University College of Medicine, Hershey, PA, 17033, USA; The Foreman Foundation for Melanoma Research, The Pennsylvania State University College of Medicine, Hershey, PA, 17033, USA; The Melanoma and Skin Cancer Center, The Pennsylvania State University College of Medicine, Hershey, PA, 17033, USA; The Melanoma Therapeutics Program, The Pennsylvania State University College of Medicine, Hershey, PA, 17033, USA.
| |
Collapse
|
7
|
Lipid metabolic Reprogramming: Role in Melanoma Progression and Therapeutic Perspectives. Cancers (Basel) 2020; 12:cancers12113147. [PMID: 33121001 PMCID: PMC7692067 DOI: 10.3390/cancers12113147] [Citation(s) in RCA: 33] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2020] [Revised: 10/21/2020] [Accepted: 10/23/2020] [Indexed: 02/07/2023] Open
Abstract
Simple Summary Melanoma is a devastating skin cancer characterized by an impressive metabolic plasticity. Melanoma cells are able to adapt to the tumor microenvironment by using a variety of fuels that contribute to tumor growth and progression. In this review, the authors summarize the contribution of the lipid metabolic network in melanoma plasticity and aggressiveness, with a particular attention to specific lipid classes such as glycerophospholipids, sphingolipids, sterols and eicosanoids. They also highlight the role of adipose tissue in tumor progression as well as the potential antitumor role of drugs targeting critical steps of lipid metabolic pathways in the context of melanoma. Abstract Metabolic reprogramming contributes to the pathogenesis and heterogeneity of melanoma. It is driven both by oncogenic events and the constraints imposed by a nutrient- and oxygen-scarce microenvironment. Among the most prominent metabolic reprogramming features is an increased rate of lipid synthesis. Lipids serve as a source of energy and form the structural foundation of all membranes, but have also emerged as mediators that not only impact classical oncogenic signaling pathways, but also contribute to melanoma progression. Various alterations in fatty acid metabolism have been reported and can contribute to melanoma cell aggressiveness. Elevated expression of the key lipogenic fatty acid synthase is associated with tumor cell invasion and poor prognosis. Fatty acid uptake from the surrounding microenvironment, fatty acid β-oxidation and storage also appear to play an essential role in tumor cell migration. The aim of this review is (i) to focus on the major alterations affecting lipid storage organelles and lipid metabolism. A particular attention has been paid to glycerophospholipids, sphingolipids, sterols and eicosanoids, (ii) to discuss how these metabolic dysregulations contribute to the phenotype plasticity of melanoma cells and/or melanoma aggressiveness, and (iii) to highlight therapeutic approaches targeting lipid metabolism that could be applicable for melanoma treatment.
Collapse
|
8
|
Gowda R, Robertson BM, Iyer S, Barry J, Dinavahi SS, Robertson GP. The role of exosomes in metastasis and progression of melanoma. Cancer Treat Rev 2020; 85:101975. [PMID: 32050108 DOI: 10.1016/j.ctrv.2020.101975] [Citation(s) in RCA: 63] [Impact Index Per Article: 12.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2019] [Revised: 01/16/2020] [Accepted: 01/18/2020] [Indexed: 12/21/2022]
Abstract
The mechanisms of melanoma metastasis have been the subject of extensive research for decades. Improved diagnostic and therapeutic strategies are of increasing importance for the treatment of melanoma due to its high burden of mortality in the advanced stages of the disease. Intercellular communication is a critical event for the progression of cancer. Collective evidence suggests that exosomes, small extracellular membrane vesicles released by the cells, are important facilitators of intercellular communication between the cells and the surrounding environment. Although the emerging field of exosomes is rapidly gaining traction in the scientific community, there is limited knowledge regarding the role of exosomes in melanoma. This review discusses the multifaceted role of melanoma-derived exosomes in promoting the process of metastasis by modulating the invasive and angiogenic capacity of malignant cells. The future implications of exosome research and the therapeutic potential of exosomes are also discussed.
Collapse
Affiliation(s)
- Raghavendra Gowda
- Departments of Pharmacology, The Pennsylvania State University College of Medicine, Hershey, PA 17033, United States; Penn State Melanoma and Skin Cancer Center, The Pennsylvania State University College of Medicine, Hershey, PA 17033, United States; Penn State Melanoma Therapeutics Program, The Pennsylvania State University College of Medicine, Hershey, PA 17033, United States; Foreman Foundation for Melanoma Research, The Pennsylvania State University College of Medicine, Hershey, PA 17033, United States
| | - Bailey M Robertson
- Departments of Pharmacology, The Pennsylvania State University College of Medicine, Hershey, PA 17033, United States
| | - Soumya Iyer
- Departments of Pharmacology, The Pennsylvania State University College of Medicine, Hershey, PA 17033, United States; Penn State Melanoma and Skin Cancer Center, The Pennsylvania State University College of Medicine, Hershey, PA 17033, United States
| | - John Barry
- Departments of Pharmacology, The Pennsylvania State University College of Medicine, Hershey, PA 17033, United States
| | - Saketh S Dinavahi
- Departments of Pharmacology, The Pennsylvania State University College of Medicine, Hershey, PA 17033, United States; Penn State Melanoma and Skin Cancer Center, The Pennsylvania State University College of Medicine, Hershey, PA 17033, United States
| | - Gavin P Robertson
- Departments of Pharmacology, The Pennsylvania State University College of Medicine, Hershey, PA 17033, United States; Departments of Pathology, The Pennsylvania State University College of Medicine, Hershey, PA 17033, United States; Departments of Dermatology, The Pennsylvania State University College of Medicine, Hershey, PA 17033, United States; Departments of Surgery, The Pennsylvania State University College of Medicine, Hershey, PA 17033, United States; Penn State Melanoma and Skin Cancer Center, The Pennsylvania State University College of Medicine, Hershey, PA 17033, United States; Penn State Melanoma Therapeutics Program, The Pennsylvania State University College of Medicine, Hershey, PA 17033, United States; Foreman Foundation for Melanoma Research, The Pennsylvania State University College of Medicine, Hershey, PA 17033, United States.
| |
Collapse
|
9
|
Fu Y, Rathod D, Abo-Ali EM, Dukhande VV, Patel K. EphA2-Receptor Targeted PEGylated Nanoliposomes for the Treatment of BRAF V600E Mutated Parent- and Vemurafenib-Resistant Melanoma. Pharmaceutics 2019; 11:pharmaceutics11100504. [PMID: 31581483 PMCID: PMC6836218 DOI: 10.3390/pharmaceutics11100504] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2019] [Revised: 09/18/2019] [Accepted: 09/19/2019] [Indexed: 12/29/2022] Open
Abstract
The clinical outcomes of malignant melanoma have improved with the introduction of mitogen-activated protein kinase kinase (MEK) inhibitors. However, off-target toxicities of the MEK inhibitor trametinib (TMB) often result in dose interruption and discontinuation of therapy. The purpose of this study was to anchor a physically stable EphrinA1-mimicking peptide known as YSA (YSAYPDSVPMMS) on TMB-loaded PEGylated nanoliposomes (YTPLs), and evaluate them in BRAFV600E-mutated parent cells (lines A375 and SK-MEL-28) and vemurafenib-resistant cells lines (A375R and SK-MEL-28R) in melanoma. TMB-loaded PEGylated liposomes (TPL) functionalized with nickel-chelated phospholipids were prepared using a modified hydration method. The hydrodynamic diameter and zeta potential values of optimized YTPL were 91.20 ± 12.16 nm and –0.92 ± 3.27 mV, respectively. The drug release study showed TPL did not leak or burst release in 24 h. The hemolysis observed was negligible at therapeutic concentrations of TMB. A differential scanning calorimetry (DSC) study confirmed that TMB was retained in a solubilized state within lipid bilayers. YTPL showed higher intracellular uptake in parental cell lines compared to vemurafenib-resistant cell lines. Western blot analysis and a cytotoxicity study with the EphA2 inhibitor confirmed a reduction in EphA2 expression in resistant cell lines. Thus, EphA2 receptor-targeted nanoliposomes can be useful for metastatic melanoma-specific delivery of TMB.
Collapse
Affiliation(s)
- Yige Fu
- Pharmaceutical Sciences, St. John's University, Queens, NY 11439, USA
| | - Drishti Rathod
- Pharmaceutical Sciences, St. John's University, Queens, NY 11439, USA
| | - Ehab M Abo-Ali
- Pharmaceutical Sciences, St. John's University, Queens, NY 11439, USA
| | - Vikas V Dukhande
- Pharmaceutical Sciences, St. John's University, Queens, NY 11439, USA
| | - Ketan Patel
- Pharmaceutical Sciences, St. John's University, Queens, NY 11439, USA.
| |
Collapse
|
10
|
Merarchi M, Jung YY, Fan L, Sethi G, Ahn KS. A Brief Overview of the Antitumoral Actions of Leelamine. Biomedicines 2019; 7:biomedicines7030053. [PMID: 31330969 PMCID: PMC6783843 DOI: 10.3390/biomedicines7030053] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2019] [Revised: 07/09/2019] [Accepted: 07/15/2019] [Indexed: 12/27/2022] Open
Abstract
For the last couple of decades, natural products, either applied singly or in conjunction with other cancer therapies including chemotherapy and radiotherapy, have allowed us to combat different types of human cancers through the inhibition of their initiation and progression. The principal sources of these useful compounds are isolated from plants that were described in traditional medicines for their curative potential. Leelamine, derived from the bark of pine trees, was previously reported as having a weak agonistic effect on cannabinoid receptors and limited inhibitory effects on pyruvate dehydrogenase kinases (PDKs). It has been reported to possess a strong lysosomotropic property; this feature enables its assembly inside the acidic compartments within a cell, such as lysosomes, which may eventually hinder endocytosis. In this review, we briefly highlight the varied antineoplastic actions of leelamine that have found implications in pharmacological research, and the numerous intracellular targets affected by this agent that can effectively negate the oncogenic process.
Collapse
Affiliation(s)
- Myriam Merarchi
- Faculty of Pharmacy, University of Paris Descartes, 75006 Paris, France
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117600, Singapore
| | - Young Yun Jung
- College of Korean Medicine, Kyung Hee University, 24 Kyungheedae-ro, Dongdaemun-gu, Seoul 02447, Korea
| | - Lu Fan
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117600, Singapore
| | - Gautam Sethi
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117600, Singapore.
| | - Kwang Seok Ahn
- College of Korean Medicine, Kyung Hee University, 24 Kyungheedae-ro, Dongdaemun-gu, Seoul 02447, Korea.
| |
Collapse
|
11
|
Machelart A, Salzano G, Li X, Demars A, Debrie AS, Menendez-Miranda M, Pancani E, Jouny S, Hoffmann E, Deboosere N, Belhaouane I, Rouanet C, Simar S, Talahari S, Giannini V, Villemagne B, Flipo M, Brosch R, Nesslany F, Deprez B, Muraille E, Locht C, Baulard AR, Willand N, Majlessi L, Gref R, Brodin P. Intrinsic Antibacterial Activity of Nanoparticles Made of β-Cyclodextrins Potentiates Their Effect as Drug Nanocarriers against Tuberculosis. ACS NANO 2019; 13:3992-4007. [PMID: 30822386 PMCID: PMC6718168 DOI: 10.1021/acsnano.8b07902] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/16/2018] [Accepted: 03/01/2019] [Indexed: 05/23/2023]
Abstract
Multi-drug-resistant tuberculosis (TB) is a major public health problem, concerning about half a million cases each year. Patients hardly adhere to the current strict treatment consisting of more than 10 000 tablets over a 2-year period. There is a clear need for efficient and better formulated medications. We have previously shown that nanoparticles made of cross-linked poly-β-cyclodextrins (pβCD) are efficient vehicles for pulmonary delivery of powerful combinations of anti-TB drugs. Here, we report that in addition to being efficient drug carriers, pβCD nanoparticles are endowed with intrinsic antibacterial properties. Empty pβCD nanoparticles are able to impair Mycobacterium tuberculosis (Mtb) establishment after pulmonary administration in mice. pβCD hamper colonization of macrophages by Mtb by interfering with lipid rafts, without inducing toxicity. Moreover, pβCD provoke macrophage apoptosis, leading to depletion of infected cells, thus creating a lung microenvironment detrimental to Mtb persistence. Taken together, our results suggest that pβCD nanoparticles loaded or not with antibiotics have an antibacterial action on their own and could be used as a carrier in drug regimen formulations effective against TB.
Collapse
Affiliation(s)
- Arnaud Machelart
- Université
de Lille, CNRS, INSERM, CHU Lille, Institut
Pasteur de Lille, U1019 - UMR 8204 - CIIL
- Center for Infection and Immunity of Lille, F-59000 Lille, France
| | - Giuseppina Salzano
- Université
Paris Sud, Université Paris-Saclay, CNRS, UMR 8214 - Institute for Molecular Sciences of Orsay (ISMO), F-91405 Orsay, France
| | - Xue Li
- Université
Paris Sud, Université Paris-Saclay, CNRS, UMR 8214 - Institute for Molecular Sciences of Orsay (ISMO), F-91405 Orsay, France
| | - Aurore Demars
- Research
Unit in Microorganisms Biology (URBM), Laboratory of Immunology and
Microbiology, Université de Namur, Narilis, B-5000 Namur, Belgium
| | - Anne-Sophie Debrie
- Université
de Lille, CNRS, INSERM, CHU Lille, Institut
Pasteur de Lille, U1019 - UMR 8204 - CIIL
- Center for Infection and Immunity of Lille, F-59000 Lille, France
| | - Mario Menendez-Miranda
- Université
Paris Sud, Université Paris-Saclay, CNRS, UMR 8214 - Institute for Molecular Sciences of Orsay (ISMO), F-91405 Orsay, France
| | - Elisabetta Pancani
- Université
Paris Sud, Université Paris-Saclay, CNRS, UMR 8214 - Institute for Molecular Sciences of Orsay (ISMO), F-91405 Orsay, France
| | - Samuel Jouny
- Université
de Lille, CNRS, INSERM, CHU Lille, Institut
Pasteur de Lille, U1019 - UMR 8204 - CIIL
- Center for Infection and Immunity of Lille, F-59000 Lille, France
| | - Eik Hoffmann
- Université
de Lille, CNRS, INSERM, CHU Lille, Institut
Pasteur de Lille, U1019 - UMR 8204 - CIIL
- Center for Infection and Immunity of Lille, F-59000 Lille, France
| | - Nathalie Deboosere
- Université
de Lille, CNRS, INSERM, CHU Lille, Institut
Pasteur de Lille, U1019 - UMR 8204 - CIIL
- Center for Infection and Immunity of Lille, F-59000 Lille, France
| | - Imène Belhaouane
- Université
de Lille, CNRS, INSERM, CHU Lille, Institut
Pasteur de Lille, U1019 - UMR 8204 - CIIL
- Center for Infection and Immunity of Lille, F-59000 Lille, France
| | - Carine Rouanet
- Université
de Lille, CNRS, INSERM, CHU Lille, Institut
Pasteur de Lille, U1019 - UMR 8204 - CIIL
- Center for Infection and Immunity of Lille, F-59000 Lille, France
| | - Sophie Simar
- Université
de Lille, Institut Pasteur de Lille, EA 4483, F-59000 Lille, France
| | - Smaïl Talahari
- Université
de Lille, Institut Pasteur de Lille, EA 4483, F-59000 Lille, France
| | - Valerie Giannini
- Institut
Pasteur, Unit for Integrated
Mycobacterial Pathogenomics, Paris, CNRS
UMR 3525, 25 Rue du Dr. Roux, F-75015 Paris, France
| | - Baptiste Villemagne
- Université
de Lille, INSERM, Institut Pasteur de Lille, U1177 - Drugs and Molecules for living Systems, F-59000 Lille, France
| | - Marion Flipo
- Université
de Lille, INSERM, Institut Pasteur de Lille, U1177 - Drugs and Molecules for living Systems, F-59000 Lille, France
| | - Roland Brosch
- Institut
Pasteur, Unit for Integrated
Mycobacterial Pathogenomics, Paris, CNRS
UMR 3525, 25 Rue du Dr. Roux, F-75015 Paris, France
| | - Fabrice Nesslany
- Université
de Lille, Institut Pasteur de Lille, EA 4483, F-59000 Lille, France
| | - Benoit Deprez
- Université
de Lille, INSERM, Institut Pasteur de Lille, U1177 - Drugs and Molecules for living Systems, F-59000 Lille, France
| | - Eric Muraille
- Research
Unit in Microorganisms Biology (URBM), Laboratory of Immunology and
Microbiology, Université de Namur, Narilis, B-5000 Namur, Belgium
- Laboratory
of Parasitology, Faculty of Medicine, Université
Libre de Bruxelles, B-1070 Brussels, Belgium
| | - Camille Locht
- Université
de Lille, CNRS, INSERM, CHU Lille, Institut
Pasteur de Lille, U1019 - UMR 8204 - CIIL
- Center for Infection and Immunity of Lille, F-59000 Lille, France
| | - Alain R. Baulard
- Université
de Lille, CNRS, INSERM, CHU Lille, Institut
Pasteur de Lille, U1019 - UMR 8204 - CIIL
- Center for Infection and Immunity of Lille, F-59000 Lille, France
| | - Nicolas Willand
- Université
de Lille, INSERM, Institut Pasteur de Lille, U1177 - Drugs and Molecules for living Systems, F-59000 Lille, France
| | - Laleh Majlessi
- Institut
Pasteur, Unit for Integrated
Mycobacterial Pathogenomics, Paris, CNRS
UMR 3525, 25 Rue du Dr. Roux, F-75015 Paris, France
| | - Ruxandra Gref
- Université
Paris Sud, Université Paris-Saclay, CNRS, UMR 8214 - Institute for Molecular Sciences of Orsay (ISMO), F-91405 Orsay, France
| | - Priscille Brodin
- Université
de Lille, CNRS, INSERM, CHU Lille, Institut
Pasteur de Lille, U1019 - UMR 8204 - CIIL
- Center for Infection and Immunity of Lille, F-59000 Lille, France
| |
Collapse
|
12
|
Betancourt LH, Pawłowski K, Eriksson J, Szasz AM, Mitra S, Pla I, Welinder C, Ekedahl H, Broberg P, Appelqvist R, Yakovleva M, Sugihara Y, Miharada K, Ingvar C, Lundgren L, Baldetorp B, Olsson H, Rezeli M, Wieslander E, Horvatovich P, Malm J, Jönsson G, Marko-Varga G. Improved survival prognostication of node-positive malignant melanoma patients utilizing shotgun proteomics guided by histopathological characterization and genomic data. Sci Rep 2019; 9:5154. [PMID: 30914758 PMCID: PMC6435712 DOI: 10.1038/s41598-019-41625-z] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2018] [Accepted: 03/13/2019] [Indexed: 12/18/2022] Open
Abstract
Metastatic melanoma is one of the most common deadly cancers, and robust biomarkers are still needed, e.g. to predict survival and treatment efficiency. Here, protein expression analysis of one hundred eleven melanoma lymph node metastases using high resolution mass spectrometry is coupled with in-depth histopathology analysis, clinical data and genomics profiles. This broad view of protein expression allowed to identify novel candidate protein markers that improved prediction of survival in melanoma patients. Some of the prognostic proteins have not been reported in the context of melanoma before, and few of them exhibit unexpected relationship to survival, which likely reflects the limitations of current knowledge on melanoma and shows the potential of proteomics in clinical cancer research.
Collapse
Affiliation(s)
| | - Krzysztof Pawłowski
- Lund University, Lund, Sweden.
- Warsaw University of Life Sciences SGGW, Warszawa, Poland.
| | | | - A Marcell Szasz
- Lund University, Lund, Sweden
- National Koranyi Institute of Pulmonology, Budapest, Hungary
- Semmelweis University, Budapest, Hungary
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | - Peter Horvatovich
- Lund University, Lund, Sweden
- University of Groningen, Groningen, The Netherlands
| | | | | | | |
Collapse
|
13
|
Kim YS, Lee YM, Oh TI, Shin DH, Kim GH, Kan SY, Kang H, Kim JH, Kim BM, Yim WJ, Lim JH. Emodin Sensitizes Hepatocellular Carcinoma Cells to the Anti-Cancer Effect of Sorafenib through Suppression of Cholesterol Metabolism. Int J Mol Sci 2018; 19:ijms19103127. [PMID: 30321984 PMCID: PMC6213641 DOI: 10.3390/ijms19103127] [Citation(s) in RCA: 62] [Impact Index Per Article: 8.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2018] [Revised: 10/01/2018] [Accepted: 10/10/2018] [Indexed: 12/12/2022] Open
Abstract
Reduced therapeutic efficacy of sorafenib, a first-generation multikinase inhibitor, is often observed during the treatment of advanced hepatocellular carcinoma (HCC). Emodin is an active component of Chinese herbs, and is effective against leukemia, lung cancer, colon cancer, pancreatic cancer, and HCC; however, the sensitizing effect of emodin on sorafenib-based HCC therapy has not been evaluated. Here, we demonstrate that emodin significantly improved the anti-cancer effect of sorafenib in HCC cells, such as HepG2, Hep3B, Huh7, SK-HEP-1, and PLC/PRF5. Mechanistically, emodin inhibits sterol regulatory element-binding protein-2 (SREBP-2) transcriptional activity, which suppresses cholesterol biosynthesis and oncogenic protein kinase B (AKT) signaling. Additionally, attenuated cholesterol synthesis and oncogenic AKT signaling inactivated signal transducer and activator of transcription 3 (STAT3), an oncogenic transcription factor. Furthermore, emodin synergistically increased cell cycle arrest in the G1 phase and apoptotic cells in the presence of sorafenib. Animal models xenografted with HepG2 or SK-HEP-1 cells also showed that the combination of emodin and sorafenib was sufficient to inhibit tumor growth. Overall, these results suggested that the combination of emodin and sorafenib may offer a potential therapy for patients with advanced HCC.
Collapse
Affiliation(s)
- Young-Seon Kim
- Department of Biomedical Chemistry, College of Biomedical & Health Science, Konkuk University, Chungju 27478, Chungbuk, Korea.
| | - Yoon-Mi Lee
- Department of Food Bioscience, College of Biomedical & Health Science, Konkuk University, Chungju 27478, Chungbuk, Korea.
| | - Taek-In Oh
- Department of Biomedical Chemistry, College of Biomedical & Health Science, Konkuk University, Chungju 27478, Chungbuk, Korea.
| | - Dong Hoon Shin
- Research Institute, National Cancer Center, Department of Cancer Biomedical Science, National Cancer Center Graduate School of Cancer Science and Policy, Goyang 10408, Korea.
| | - Geon-Hee Kim
- Department of Biomedical Chemistry, College of Biomedical & Health Science, Konkuk University, Chungju 27478, Chungbuk, Korea.
| | - Sang-Yeon Kan
- Department of Biomedical Chemistry, College of Biomedical & Health Science, Konkuk University, Chungju 27478, Chungbuk, Korea.
| | - Hyeji Kang
- Department of Biomedical Chemistry, College of Biomedical & Health Science, Konkuk University, Chungju 27478, Chungbuk, Korea.
| | - Ji Hyung Kim
- College of Life Sciences and Biotechnology, Korea University, Seoul 02841, Korea.
| | - Byeong Mo Kim
- Severance Integrative Research Institute for Cerebral & Cardiovascular Diseases (SIRIC), Yonsei University College of Medicine, Seodaemun-gu, Seoul 03722, Korea.
| | - Woo Jong Yim
- Jung-Ang Microbe Research Institute (JM), 398, Jikji-daero, Heungdeok-gu, Cheongju 28576, Chungbuk, Korea.
| | - Ji-Hong Lim
- Department of Biomedical Chemistry, College of Biomedical & Health Science, Konkuk University, Chungju 27478, Chungbuk, Korea.
- Diabetes and Bio-Research Center, Konkuk University, Chungju 27478, Chungbuk, Korea.
| |
Collapse
|
14
|
Syntheses of C-ring modified dehydroabietylamides and their cytotoxic activity. Eur J Med Chem 2018; 156:861-870. [PMID: 30056282 DOI: 10.1016/j.ejmech.2018.07.051] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2018] [Revised: 07/15/2018] [Accepted: 07/19/2018] [Indexed: 11/22/2022]
Abstract
Due to their auspicious pharmacological efficacy as future drug candidates, natural products have been attracting scientific interest for centuries. An interesting field of research concerns the natural product class of terpenes. In this regard, a multitude of studies have already shown their promising biological potential. Therefore, a set of 27 derivatives of the diterpene dehydroabietylamine was synthesized, focusing on C-ring modifications and the derivatization of the amino moiety at C-18. Subsequent screening of the compounds in colorimetric sulforhodamine B-assays revealed an in vitro cytotoxicity especially towards malignant cell line MCF7. Particularly, 12-hydroxy-N-(isonicotinoyl)dehydroabietylamine and N-(4-methoxybenzoyl)dehydroabietylamine showed good cytotoxic activities (EC50 (MCF7) = 4.3 ± 0.2 μM and EC50 (MCF7) = 4.5 ± 1.5 μM, respectively) and significant selectivities (SI = 6.2 and SI = 8.8, respectively) towards malignant cell lines.
Collapse
|