1
|
Lazo PA. VRK2 kinase pathogenic pathways in cancer and neurological diseases. BIOCHIMICA ET BIOPHYSICA ACTA. MOLECULAR CELL RESEARCH 2025; 1872:119949. [PMID: 40187568 DOI: 10.1016/j.bbamcr.2025.119949] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/14/2025] [Revised: 03/07/2025] [Accepted: 04/01/2025] [Indexed: 04/07/2025]
Abstract
The VRK2 ser-thr kinase, belonging to the dark kinome, is implicated in the pathogenesis of cancer progression, neurological and psychiatric diseases. The VRK2 gene codes for two isoforms. The main isoform (VRK2A) is mainly located in the cytoplasm, and anchored to different types of membranes, such as the endoplasmic reticulum, mitochondria and nuclear envelope. The VRK2A isoform interacts with signaling modules assembled on scaffold proteins such as JIP1 or KSR1, forming stable complexes and blocking the activation of regulatory signaling pathways by altering their intracellular localization and the balance among them. VRK2 regulates apoptosis, nuclear membrane organization, immune responses, and Cajal bodies. Wild-type VRK2 is overexpressed in tumors and contributes to cancer development. In cells and tumors with low levels of nuclear VRK1, VRK2 generates by alternative splicing a shorter isoform (VRK2B) that lacks the C-terminal hydrophobic tail and permits its relocation to nuclei. Furthermore, rare VRK2 gene variants are associated with different neurological or psychiatric diseases such as schizophrenia, epilepsy, bipolar disorder, depression, autism, circadian clock alterations and insomnia, but their pathogenic mechanism is unknown. These diseases are a likely consequence of an altered balance among different signaling pathways that are regulated by VRK2.
Collapse
Affiliation(s)
- Pedro A Lazo
- Instituto de Biología Molecular y Celular del Cáncer (IBMCC), Consejo Superior de Investigaciones Científicas (CSIC), Universidad de Salamanca, 37007 Salamanca, Spain; Instituto de Investigación Biomédica de Salamanca (IBSAL), Hospital Universitario de Salamanca, 37007 Salamanca, Spain.
| |
Collapse
|
2
|
Awad MA, Soliman HSM, El-Mashtoly SF, Mansour YE, El-Deeb B, Hammad SF. Isolation and characterization of a bioactive compound from Sphingomonas sanguinis DM with cytotoxic and molecular docking analysis. Sci Rep 2025; 15:16049. [PMID: 40341615 PMCID: PMC12062506 DOI: 10.1038/s41598-025-99178-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2024] [Accepted: 04/17/2025] [Indexed: 05/10/2025] Open
Abstract
Datura metel, a common plant in the Solanaceae family, is known for its valuable medicinal properties. The metabolites created by its rhizosphere bacterium, Sphingomonas sanguinis DM, have garnered interest for their potential biological effects. This study will discuss the steps involved in fermenting and processing a bacterial strain to extract potent secondary metabolites. The ethyl acetate extract of the propagated strain was subjected to fractionation and purification through various chromatographic techniques. The purified compound was characterized through multiple spectroscopic methods for structure elucidation, including UV, MS, 1D, and 2D-NMR. Its cytotoxic activity was assessed on malignant skin cells (A-431) using the MTT test compared with normal melanocytes (HFB 4). Furthermore, A-431 cells were double-stained with PI and annexin V-FITC and analyzed by flow cytometry to detect Apoptosis. Molecular investigations include PCR screening to detect genes related to the biosynthesis of bioactive metabolites, such as NRPS and lipopeptide ItuD genes. A prospective effective strategy to overcome tumor plasticity in melanoma is to target the Wnt signaling pathways. Molecular docking studies were conducted in the different proteins (Fz4-CRD, LRP6, GSK3β) of the Wnt signaling pathway and Protein Kinase B/Akt for the isolated compound to investigate the possible pathway to inhibit melanoma. Sphingomonas sanguinis DM produced bis (2-methylheptyl) benzene-1,4-dicarboxylate isolated for the first time from a natural source. It was cytotoxic against the A-431 human skin carcinoma cell line (IC50 = 191.61 µg/mL) but less effective against HFB 4 human normal melanocytes (IC50 = 416.23 µg/mL; selectivity index = 2.17). The A-431 cells showed a significant increase in early Apoptosis and a moderate rise in late Apoptosis. PCR amplification confirmed genes encoding A domain and Iturin A. Bacterial sequences are available in NCBI GenBank with accession codes OR597597 and OR597598. Consequently, Sphingomonas sanguinis DM synthesized a cytotoxic natural terephthalate diester derivative, along with the host specificity of the strain.
Collapse
Affiliation(s)
- Mohamed A Awad
- Biotechnology Program, Institute of Basic and Applied Science, Egypt-Japan University of Science and Technology (E-JUST), New Borg El-Arab City, 21934, Alexandria, Egypt
- Botany and Microbiology Department, Faculty of Science, Sohag University, Sohag, 82524, Egypt
| | - Hesham S M Soliman
- Department of Pharmacognosy, Faculty of Pharmacy, Helwan University, Ain-Helwan, Cairo, 11795, Egypt.
- PharmD Program, Egypt-Japan University of Science and Technology (E-JUST), New Borg El-Arab City, 21934, Alexandria, Egypt.
| | - Samir F El-Mashtoly
- Biotechnology Program, Institute of Basic and Applied Science, Egypt-Japan University of Science and Technology (E-JUST), New Borg El-Arab City, 21934, Alexandria, Egypt
- Leibniz Institute of Photonic Technology, Albert-Einstein-Straße, 07745, Jena, Germany
| | - Yara E Mansour
- Pharmaceutical Organic Chemistry Department, Faculty of Pharmacy, Helwan University, Ain-Helwan, Cairo, 11795, Egypt
| | - Bahig El-Deeb
- Botany and Microbiology Department, Faculty of Science, Sohag University, Sohag, 82524, Egypt
| | - Sherif F Hammad
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, Helwan University, Ain-Helwan, Cairo, 11795, Egypt
- PharmD Program, Egypt-Japan University of Science and Technology (E-JUST), New Borg El-Arab City, 21934, Alexandria, Egypt
| |
Collapse
|
3
|
Fu L, Yong JM, Yeh R, Bartlett F, Whitelock JM, Lord MS. Functionalized Cerium Oxide Nanoparticles Enhance Penetration into Melanoma Spheroids In Vivo through Angiogenesis. Adv Healthc Mater 2025; 14:e2405129. [PMID: 40109098 PMCID: PMC12057615 DOI: 10.1002/adhm.202405129] [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: 12/18/2024] [Revised: 02/27/2025] [Indexed: 03/22/2025]
Abstract
Angiogenesis is a crucial step in tumor progression, including melanoma, making anti-angiogenic strategies a widely explored treatment approach. However, both innate and acquired resistance to these therapies suggest that this approach may need re-evaluation. Nanoparticles have gained attention for their potential to enhance drug delivery and retention within tumors via the bloodstream. However, the in vitro screening of nanoparticles is limited by the inability of preclinical models to replicate the complex tumor microenvironment, especially the blood supply. Here, it is demonstrated that melanoma cells embedded in Matrigel spheroids can engraft in and be vascularized by the chorioallantoic membrane (CAM) of fertilized chicken eggs. This model allows for the assessment of nanoparticle toxicity and accumulation in tumor spheroids, as well as functional effects such as angiogenesis. Cerium oxide nanoparticles (nanoceria) and their surface functionalized derivatives are widely explored for biomedical applications due to their ability to modulate oxidative stress and angiogenesis. Here, it is observed that heparin functionalized nanoceria penetrate melanoma spheroids in the CAM and promote spheroid vascularization to a greater extent than nanoceria alone. This study aids in the development of preclinical cancer models for nanoparticle screening and provides new insight into the interplay between nanoparticle surface coatings and biological effects.
Collapse
Affiliation(s)
- Lu Fu
- Graduate School of Biomedical EngineeringUniversity of New South WalesSydneyNSW2052Australia
| | - Joel M. Yong
- Graduate School of Biomedical EngineeringUniversity of New South WalesSydneyNSW2052Australia
| | - Robyn Yeh
- Graduate School of Biomedical EngineeringUniversity of New South WalesSydneyNSW2052Australia
| | - Florence Bartlett
- Katherina Gaus Light Microscopy FacilityMark Wainwright Analytical CentreUniversity of New South WalesSydneyNSW2052Australia
| | - John M. Whitelock
- Graduate School of Biomedical EngineeringUniversity of New South WalesSydneyNSW2052Australia
| | - Megan S. Lord
- Graduate School of Biomedical EngineeringUniversity of New South WalesSydneyNSW2052Australia
| |
Collapse
|
4
|
El Halawany M, Sharaky M, Aziz D. Stearyl amine tailored spanlastics embedded within tetronic ® nanogel for boosting the repurposed anticancer potential of mebendazole: formulation, in vitro profiling, cytotoxicity assessment, and in vivo permeation analysis. Daru 2025; 33:17. [PMID: 40156679 PMCID: PMC11954785 DOI: 10.1007/s40199-025-00560-3] [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: 07/12/2024] [Accepted: 03/06/2025] [Indexed: 04/01/2025] Open
Abstract
BACKGROUND Mebendazole (MBZ) is an anthelmintic drug that was repurposed as an anti-cancer agent. OBJECTIVES This study aimed at formulating MBZ into stearylamine tailored spanlastics dispersed in nanogel for enhancing MBZ anti-tumor efficacy against skin cancer. METHODS MBZ spanlastics were prepared by thin film hydration using 21 × 31 factorial design. The formulation variables were the total amount (mg) of Span 60 and Tween 80 in the formulations and the ratio between Span 60 and Tween 80. RESULTS Optimal spanlastics formulation was composed of 400 mg of Span 60 and Tween 80 in a ratio of 2:1 and showed EE% of 78 ± 2.9% and PS of 284.00 ± 35.36 nm. Stearylamine (20 mg) was added to the optimized formulation and showed acceptable positive charge (zeta potential = 47.53 ± 1.50 mV). It was dispersed in 30% Tetronic®1107 solution to form a nanogel. MBZ nanogel was assessed for their cytotoxic effect on cell proliferation against human malignant melanoma and epidermoid carcinoma cell lines and showed 38.70 ± 1.70% and 48.60 ± 0.50% (respectively) cell proliferation compared to the control group (100%). Finally, its permeation through Wistar rat skin was tested. CONCLUSION SA-spanlastics nanogel holds potential as an effective nanocarrier for boosting MBZ anti-cancer efficacy.
Collapse
Affiliation(s)
- Mai El Halawany
- Pharmaceutics and Industrial Pharmacy Department, Faculty of Pharmacy, Cairo University, Kasr El-Ainy Street 11562, Cairo, Egypt
| | - Marwa Sharaky
- Pharmacology Unit, Cancer Biology Department, National Cancer Institute, Cairo University, Kasr El-Ainy Street 11562, Cairo, Egypt
- Pharmacology and Toxicology Department, Faculty of Pharmacy, Ahram Canadian University, 6Th October City 12573, Giza, Egypt
| | - Diana Aziz
- Pharmaceutics and Industrial Pharmacy Department, Faculty of Pharmacy, Cairo University, Kasr El-Ainy Street 11562, Cairo, Egypt.
| |
Collapse
|
5
|
Arnold MR, Chen S, Unni VK. Alpha-synuclein knockout impairs melanoma development and alters DNA damage repair in the TG3 mouse model in a sex-dependent manner. Front Oncol 2025; 15:1554059. [PMID: 40182046 PMCID: PMC11967197 DOI: 10.3389/fonc.2025.1554059] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2024] [Accepted: 02/26/2025] [Indexed: 04/05/2025] Open
Abstract
Introduction Strong evidence suggests links between Parkinson's Disease (PD) and melanoma, as studies have found that people with PD are at an increased risk of developing melanoma and those with melanoma are at increased risk of developing PD. Although these clinical associations are well-established, the cellular and molecular pathways linking these diseases are poorly understood. Recent studies have found a previously unrecognized role for the neurodegeneration-associated protein alpha-synuclein (αSyn) in melanoma; the overexpression of αSyn promotes melanoma cell proliferation and metastasis. However, to our knowledge, no studies have investigated the role of αSyn in in vivo melanoma models outside of a xenograft paradigm. Methods Our study created and characterized Snca knockout in the spontaneously developing melanoma TG3 mouse line, TG3+/+Snca-/-. Results We show that αSyn loss-of-function significantly delays melanoma onset and slows tumor growth in vivo in males. Furthermore, decreased tumor volume is correlated with a decreased DNA damage signature and increased apoptotic markers, indicating a role for αSyn in modulating the DNA damage response (DDR) pathway. Discussion Overall, our study may suggest that targeting αSyn and its role in modulating the DDR and melanomagenesis could serve as a promising new therapeutic target.
Collapse
Affiliation(s)
- Moriah R. Arnold
- Medical Scientist Training Program, Oregon Health and Science University, Portland, OR, United States
- Department of Neurology and Jungers Center for Neurosciences Research, Oregon Health and Science University, Portland, OR, United States
| | - Suzie Chen
- Department of Chemical Biology, Ernest Mario School of Pharmacy, Rutgers University, Piscataway, NJ, United States
| | - Vivek K. Unni
- Department of Neurology and Jungers Center for Neurosciences Research, Oregon Health and Science University, Portland, OR, United States
- OHSU Parkinson Center, Oregon Health and Science University, Portland, OR, United States
| |
Collapse
|
6
|
Arnold MR, Chen S, Unni VK. Alpha-synuclein knockout impairs melanoma development and alters DNA damage repair in the TG3 mouse model in a sex-dependent manner. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.12.01.626256. [PMID: 39677631 PMCID: PMC11642733 DOI: 10.1101/2024.12.01.626256] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 12/17/2024]
Abstract
Strong evidence suggests links between Parkinson's Disease (PD) and melanoma, as studies have found that people with PD are at an increased risk of developing melanoma and those with melanoma are at increased risk of developing PD. Although these clinical associations are well-established, the cellular and molecular pathways linking these diseases are poorly understood. Recent studies have found a previously unrecognized role for the neurodegeneration-associated protein alpha-synuclein (αSyn) in melanoma; the overexpression of αSyn promotes melanoma cell proliferation and metastasis. However, to our knowledge, no studies have investigated the role of αSyn in in vivo melanoma models outside of a xenograft paradigm. Our study created and characterized Snca knockout in the spontaneously developing melanoma TG3 mouse line, TG3+/+Snca-/-. We show that αSyn loss-of-function significantly delays melanoma onset and slows tumor growth in vivo. Furthermore, decreased tumor volume is correlated with a decreased DNA damage signature and increased apoptotic markers, indicating a role for αSyn in modulating the DNA damage response (DDR) pathway. Overall, our study provides evidence that targeting αSyn and its role in modulating the DDR and melanomagenesis could serve as a promising new therapeutic target.
Collapse
Affiliation(s)
- Moriah R. Arnold
- Medical Scientist Training Program, Oregon Health and Science University, Portland, OR, USA
- Department of Neurology and Jungers Center for Neurosciences Research, Oregon Health and Science University, Portland, OR, USA
| | - Suzie Chen
- Departments of Chemical Biology, Ernest Mario School of Pharmacy, Rutgers University, NJ, USA
| | - Vivek K. Unni
- Department of Neurology and Jungers Center for Neurosciences Research, Oregon Health and Science University, Portland, OR, USA
- OHSU Parkinson Center, Oregon Health and Science University, Portland, OR, USA
| |
Collapse
|
7
|
Tósaki Á, Szabó Z, Király J, Lőrincz EB, Vass V, Tánczos B, Bereczki I, Herczegh P, Remenyik É, Tósaki Á, Szabó E. A new cannabigerol derivative, LE-127/2, induces autophagy mediated cell death in human cutaneous melanoma cells. Eur J Pharm Sci 2024; 203:106920. [PMID: 39357769 DOI: 10.1016/j.ejps.2024.106920] [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: 06/19/2024] [Revised: 09/26/2024] [Accepted: 09/29/2024] [Indexed: 10/04/2024]
Abstract
Despite the targeted- and immunotherapies used in the past decade, survival rate among patients with metastatic melanoma remains low, therefore, melanoma is responsible for the majority of skin cancer-related deaths. The ongoing investigation of natural antitumor agents, the nonpsychoactive cannabinoid, cannabigerol (CBG) found in Cannabis sativa is emerging as a promising candidate. CBG offers a potential therapeutic role in the treatment of melanoma demonstrating cell growth inhibition in some tumors. Its low water solubility and bioavailability hinder the potential effectiveness. To address these challenges, a modified CBG, namely LE-127/2 was synthesized by Mannich-type reaction. The aim was to investigate the effect of this novel compound on cell proliferation as well as the mechanism of cell death with a particular focus on autophagy and apoptosis. Human cutan melanoma cell lines, WM35, A2058 and WM3000 were utilized for the present study. Cell proliferation of the cells after the treatment with LE-127/2, parent CBG or vemurafenib was assessed by Cell Titer Blue Assay. Cells were treated with a 1.25-80 µM of the above-mentioned compounds, and it was found that at 20 μM of all drugs showed a comparable effective inhibition of cell proliferation, however, vemurafenib and CBG proved to be more effective than LE-127/2. In addition, clonogenic cell survival assays were performed to examine the inhibitory effect of LE-127/2 on the colony formation ability of melanoma cell lines. Cells treated with 20 µM of LE-127/2 for 14 days showed about a 50% suppression of clonogenic cell survival. LE-127/2 exerted the most intensive inhibition on A2058 cell colonies. Furthermore, notably, LDH cytotoxicity assay performed on HaCaT cell line, proved LE-127/2 to be cytotoxic only at higher concentration, such as 80 μM, while the parent CBG was cytotoxic at concentration as low as 5 μM, suggesting that the new CBG derivative as a drug candidate may be applied in human pharmacotherapy without causing a substantial damage in intact epidermal cells. Analysis of protein expression revealed the impact of LE-127/2 on the expression of basic proteins (LC-3, Beclin-1 and p62) involved in the process of autophagy in the three different melanoma cell lines studied. Elevated expression of these proteins was detected as a result of LE-127/2 (20 µM) treatment. LE-127/2 also induced the expression of some proteins involved in apoptosis, and it is particularly noteworthy the increased level of cleaved PARP. Based on the results obtained, it can be concluded that LE-127/2 induced autophagy could lead to the inhibition of cell proliferation and death in melanoma cells.
Collapse
Affiliation(s)
- Ágnes Tósaki
- Department of Dermatology, Faculty of Medicine, University of Debrecen, Debrecen, Hungary.
| | - Zsuzsanna Szabó
- Department of Biopharmacy, Faculty of Pharmacy, University of Debrecen, Debrecen, Hungary.
| | - József Király
- Department of Biopharmacy, Faculty of Pharmacy, University of Debrecen, Debrecen, Hungary.
| | - Eszter Boglárka Lőrincz
- Doctoral School of Pharmaceutical Sciences, University of Debrecen, Debrecen, Hungary; Department of Pharmaceutical Chemistry, Faculty of Pharmacy, University of Debrecen, Debrecen, Hungary.
| | - Virág Vass
- Doctoral School of Pharmaceutical Sciences, University of Debrecen, Debrecen, Hungary; Department of Pharmacology, Faculty of Pharmacy, University of Debrecen, Debrecen, Hungary.
| | - Bence Tánczos
- Department of Pharmacology, Faculty of Pharmacy, University of Debrecen, Debrecen, Hungary; HUN-REN-DE Pharmamodul Research Group, University of Debrecen, 4032 Debrecen, Nagyerdei krt. 98, Hungary.
| | - Ilona Bereczki
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, University of Debrecen, Debrecen, Hungary; HUN-REN-DE Pharmamodul Research Group, University of Debrecen, 4032 Debrecen, Nagyerdei krt. 98, Hungary.
| | - Pál Herczegh
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, University of Debrecen, Debrecen, Hungary; HUN-REN-DE Pharmamodul Research Group, University of Debrecen, 4032 Debrecen, Nagyerdei krt. 98, Hungary.
| | - Éva Remenyik
- Department of Dermatology, Faculty of Medicine, University of Debrecen, Debrecen, Hungary.
| | - Árpád Tósaki
- Department of Pharmacology, Faculty of Pharmacy, University of Debrecen, Debrecen, Hungary; HUN-REN-DE Pharmamodul Research Group, University of Debrecen, 4032 Debrecen, Nagyerdei krt. 98, Hungary.
| | - Erzsébet Szabó
- Department of Pharmacology, Faculty of Pharmacy, University of Debrecen, Debrecen, Hungary; HUN-REN-DE Pharmamodul Research Group, University of Debrecen, 4032 Debrecen, Nagyerdei krt. 98, Hungary.
| |
Collapse
|
8
|
Leroy M, Desmedt E, Deramoudt L, Vasseur M, Odou P, Béhal H, Décaudin B, Mortier L, Simon N. Retrospective comparison of a weight-based dose every 2 weeks with a fixed dose every month: a real-life analysis of nivolumab in the treatment of advanced melanoma. Melanoma Res 2024; 34:258-264. [PMID: 38489575 DOI: 10.1097/cmr.0000000000000965] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/17/2024]
Abstract
Nivolumab was first authorized at a weight-based dose (WBD) of 3 mg/kg every two weeks (Q2W). Since 2017, a fixed dose (FD) regimen [first 240 mg Q2W and then 480 mg per month (Q4W)] was allowed. The objective of the study was to compare a WBD regimen and an FD regimen with regard to effectiveness and safety. We conducted a single-center, retrospective, real-life study of consecutive adult patients who had received a WBD of nivolumab or an FD of 480 mg Q4W. The primary endpoint was the occurrence of grade ≥3 immune-related adverse events (irAEs). The secondary endpoints were overall survival and cost of the treatment. In all, 342 patients were included: 71 in the WBD cohort and 271 in the FD cohort. Of these patients, 201 patients (59.6%) experienced an irAE, and 24 of these events were graded as ≥3. At 12 months, there was no significant difference in irAE occurrence between the two cohorts [hazard ratio (95% confidence interval): 0.54 (0.21-1.36), P = 0.19]. The 12-month overall survival rate was significantly lower in the WBD cohort ( P < 0.001). Switching from a fortnightly weight dose to a fixed monthly dose halves the cost of hospitalization. Our results did not show a significant difference between WBD and FD cohort in the occurrence of severe irAEs. However overall survival appeared to be significantly higher in FD group. Some clinical trials are investigating a hybrid dosing regimen in which a WBD is capped by an FD. The present results need to be confirmed in prospective studies.
Collapse
Affiliation(s)
| | - Eve Desmedt
- Service de Dermatologie, Hôpital C. Huriez, CHU de Lille
| | - Laure Deramoudt
- Institut de Pharmacie, CHU Lille
- Univ. Lille, ULR 7365 - GRITA - Groupe de Recherche sur les formes Injectables et les Technologies Associées
| | - Michèle Vasseur
- Institut de Pharmacie, CHU Lille
- Univ. Lille, ULR 7365 - GRITA - Groupe de Recherche sur les formes Injectables et les Technologies Associées
| | - Pascal Odou
- Institut de Pharmacie, CHU Lille
- Univ. Lille, ULR 7365 - GRITA - Groupe de Recherche sur les formes Injectables et les Technologies Associées
| | | | - Bertrand Décaudin
- Institut de Pharmacie, CHU Lille
- Univ. Lille, ULR 7365 - GRITA - Groupe de Recherche sur les formes Injectables et les Technologies Associées
| | - Laurent Mortier
- Department of Dermatology, Claude Huriez Hospital, CARADERM and University of Lille, Lille, France
| | - Nicolas Simon
- Institut de Pharmacie, CHU Lille
- Univ. Lille, ULR 7365 - GRITA - Groupe de Recherche sur les formes Injectables et les Technologies Associées
| |
Collapse
|
9
|
Hutchenreuther J, Nguyen J, Quesnel K, Vincent KM, Petitjean L, Bourgeois S, Boyd M, Bou-Gharios G, Postovit LM, Leask A. Cancer-associated Fibroblast-specific Expression of the Matricellular Protein CCN1 Coordinates Neovascularization and Stroma Deposition in Melanoma Metastasis. CANCER RESEARCH COMMUNICATIONS 2024; 4:556-570. [PMID: 38363129 PMCID: PMC10898341 DOI: 10.1158/2767-9764.crc-23-0571] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/18/2023] [Revised: 01/19/2024] [Accepted: 02/09/2024] [Indexed: 02/17/2024]
Abstract
Melanoma is the leading cause of skin cancer-related death. As prognosis of patients with melanoma remains problematic, identification of new therapeutic targets remains essential. Matricellular proteins are nonstructural extracellular matrix proteins. They are secreted into the tumor microenvironment to coordinate behavior among different cell types, yet their contribution to melanoma is underinvestigated. Examples of matricellular proteins include those comprising the CCN family. The CCN family member, CCN1, is highly proangiogenic. Herein, we show that, in human patients with melanoma, although found in several tumor cell types, CCN1 is highly expressed by a subset of cancer-associated fibroblasts (CAF) in patients with melanoma and this expression correlates positively with expression of proangiogenic genes and progressive disease/resistance to anti-PD1 checkpoint inhibitors. Consistent with these observations, in a syngeneic C57BL6 mouse model of melanoma, loss of CCN1 expression from Col1A2-Cre-, herein identified as "universal," fibroblasts, impaired metastasis of subcutaneously injected B16F10 tumor cells to lung, concomitant with disrupted neovascularization and collagen organization. Disruption of the extracellular matrix in the loss of CCN1 was validated using a novel artificial intelligence-based image analysis platform that revealed significantly decreased phenotypic fibrosis and composite morphometric collagen scores. As drug resistance is linked to matrix deposition and neoangiogenesis, these data suggest that CCN1, due to its multifaceted role, may represent a novel therapeutic target for drug-resistant melanoma. Our data further emphasize the essential role that cancer-associated, (universal) Col1A2-Cre-fibroblasts and extracellular matrix remodeling play in coordinating behavior among different cell types within the tumor microenvironment. SIGNIFICANCE In human patients, the expression of proangiogenic matricellular protein CCN1 in CAFs correlates positively with expression of stroma and angiogenic markers and progressive disease/resistance to checkpoint inhibitor therapy. In an animal model, loss of CCN1 from CAFs impaired metastasis of melanoma cells, neovascularization, and collagen deposition, emphasizing that CAFs coordinate cellular behavior in a tumor microenvironment and that CCN1 may be a novel target.
Collapse
Affiliation(s)
- James Hutchenreuther
- Department of Physiology and Pharmacology, University of Western Ontario, London, Ontario, Canada
| | - John Nguyen
- College of Dentistry, University of Saskatchewan, Saskatoon, Saskatchewan, Canada
| | - Katherine Quesnel
- Department of Dentistry, University of Western Ontario, London, Ontario, Canada
| | - Krista M. Vincent
- Department of Oncology, University of Alberta, Edmonton, Alberta, Canada
- Department of Medical Genetics, Children's Hospital of Eastern Ontario, Ottawa, Ontario, Canada
| | | | - Sophia Bourgeois
- Department of Biology, University of Western Ontario, London, Ontario, Canada
| | - Mark Boyd
- Office of the Vice President of Research, University of Saskatchewan, Saskatoon, Saskatchewan, Canada
| | - George Bou-Gharios
- Department of Musculoskeletal and Ageing Science, Institute of Life Course and Medical Sciences, University of Liverpool, Liverpool, United Kingdom
| | - Lynne-Marie Postovit
- Department of Biomedical and Molecular Sciences, Queens University, Kingston, Ontario, Canada
| | - Andrew Leask
- College of Dentistry, University of Saskatchewan, Saskatoon, Saskatchewan, Canada
| |
Collapse
|
10
|
Avola R, Graziano ACE, Madrid A, Clericuzio M, Cardile V, Russo A. Pholiotic acid promotes apoptosis in human metastatic melanoma cells. Chem Biol Interact 2024; 390:110894. [PMID: 38301881 DOI: 10.1016/j.cbi.2024.110894] [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: 11/23/2023] [Revised: 12/30/2023] [Accepted: 01/29/2024] [Indexed: 02/03/2024]
Abstract
Mushrooms produce a great variety of secondary metabolites that can be successful in both prevention and treatment of various cancers. In particular, higher Basidiomycete mushrooms contain various types of biologically active low-molecular compounds in fruiting bodies with suggested anticarcinogenic effects. The polyamine analogue {(2R)-2-[(S)-3-hydroxy-3-methylglutaryloxy] putrescine dicinnamamide} indicated with the name pholiotic acid, isolated for the first time by us from the fruiting bodies of the Basidiomycete Pholiota spumosa (Fr.) Sing. (Strophariaceae), inhibited the viability of human prostate cancer cells, such as other polyamine synthetic analogues that have shown antitumor activity in several types of cancer, including melanoma. Melanoma is an aggressive skin cancer that can metastasize to other organs and presents a high resistance to conventional therapies. In light of these considerations, the present study was therefore designed to assess whether this putrescine derivative could inhibit the growth of human metastatic melanoma cell lines, M14 and A2058. The results obtained demonstrate that this natural compound, at 12.5-50 μM concentration, was able to reduce cell viability of both cancer cells inducing cell death by intrinsic apoptotic pathway that probably involves PTEN activity, inhibition of Hsp70 expression and reactive oxygen species production. On the other hand, the increased expression of enzymes involved in polyamine catabolism trigger apoptotic cell death leading to polyamine depletion and generation of reactive oxygen species as by-products. In conclusion, these findings, starting point for further investigation, implement available our data to support pholiotic acid as an attractive potential chemopreventive agent, and provide a basis for further research into the use of this polyamine derivative as potential anticancer agent for melanoma in combination with existing therapies to improve treatment efficacy and overcome the obstacle of drug resistance.
Collapse
Affiliation(s)
- R Avola
- Faculty of Medicine and Surgery, University of Enna "Kore", 94100, Enna, Italy
| | - A C E Graziano
- Faculty of Medicine and Surgery, University of Enna "Kore", 94100, Enna, Italy
| | - A Madrid
- Dept. de Química, Facultad de Ciencias Naturales y Exactas, Universidad de Playa Ancha, Avda. Leopoldo Carvallo 270, Playa Ancha, Valparaíso, 2340000, Chile
| | - M Clericuzio
- Dept. of Science and Technological Innovation, University of Piemonte Orientale, V.le T. Michel 11, 15121, Alessandria, Italy
| | - V Cardile
- Dept. of Biomedical and Biotechnological Sciences, University of Catania, Via S. Sofia, 89, 95123, Catania, Italy
| | - A Russo
- Dept. of Drug and Health Sciences, University of Catania, V.le A. Doria 6, 95125, Catania, Italy.
| |
Collapse
|
11
|
Yazdani Z, Pasandi MS, Golpour M, Eslami M, Rafiei A. Effect of cold atmospheric plasma on changing of biomolecular structures involved in apoptosis pathways of melanoma cancer. Skin Res Technol 2024; 30:e13544. [PMID: 38174746 PMCID: PMC10765363 DOI: 10.1111/srt.13544] [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: 05/31/2023] [Accepted: 11/22/2023] [Indexed: 01/05/2024]
Abstract
BACKGROUND Cold atmospheric plasma (CAP), is a technology based on non-thermal ionized gas that is used for cancer therapy in research. We evaluated the effect of CAP on malignant melanoma cancer cell line (B16) in comparison with normal cells (L929). METHODS The effect of CAP on the cytotoxicity of B16 and L929 cell lines was assayed by the MTT method and inverted microscopy. The induction of apoptosis in cells was evaluated using a fluorescence microscope. FTIR monitored the CAP effect in biomacromolecules changes in these cell lines. QPCR assayed gene expression of BAX, BCL-2, and Caspase-3 (CASP-3). RESULTS The results of the MTT test showed CAP has a cytotoxic effect on the B16 cancer cell line more than L929 normal cells (p < 0.0001). The results of invert and fluorescence microscopy showed CAP-induced apoptotic morphology on cancerous cells. FTIR spectroscopy indicated CAP changes biomacromolecules structure. Evaluation of gene expression showed CAP increased BAX and CASP-3 gene expression. Also, it decreased BCL-2 gene expression. CONCLUSIONS Taken together, CAP may change biomacromolecule structures involved in apoptosis pathways, decrease proliferation and induce apoptosis in cancer cells.
Collapse
Affiliation(s)
- Zahra Yazdani
- Department of ImmunologyMolecular and Cell Biology Research Center, School of Medicine, Mazandaran University of Medical SciencesSariIran
- Student Research CommitteeSchool of Medicine, Mazandaran University of Medical SciencesSariIran
| | - Marzieh Sharifi Pasandi
- Department of ImmunologyMolecular and Cell Biology Research Center, School of Medicine, Mazandaran University of Medical SciencesSariIran
| | - Monireh Golpour
- Department of ImmunologyMolecular and Cell Biology Research Center, School of Medicine, Mazandaran University of Medical SciencesSariIran
- Student Research CommitteeSchool of Medicine, Mazandaran University of Medical SciencesSariIran
| | - Mina Eslami
- Department of ImmunologyMolecular and Cell Biology Research Center, School of Medicine, Mazandaran University of Medical SciencesSariIran
| | - Alireza Rafiei
- Department of ImmunologyMolecular and Cell Biology Research Center, School of Medicine, Mazandaran University of Medical SciencesSariIran
| |
Collapse
|
12
|
Januszewski J, Forma A, Zembala J, Flieger M, Tyczyńska M, Dring JC, Dudek I, Świątek K, Baj J. Nutritional Supplements for Skin Health-A Review of What Should Be Chosen and Why. MEDICINA (KAUNAS, LITHUANIA) 2023; 60:68. [PMID: 38256329 PMCID: PMC10820017 DOI: 10.3390/medicina60010068] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/31/2023] [Revised: 12/13/2023] [Accepted: 12/15/2023] [Indexed: 01/24/2024]
Abstract
Supplementation of micronutrients is considered to be crucial in the reinforcement of the skin's barrier. In this paper, 14 nutritional compounds commonly used in food or pharmaceutic industries were analyzed in terms of influencing skin conditions. The major objective of this paper was to provide a narrative review of the available literature regarding several chosen compounds that are currently widely recommended as supplements that aim to maintain proper and healthy skin conditions. We conducted a review of the literature from PubMed, Scopus, and Web of Science until September 2023 without any other restrictions regarding the year of the publication. Ultimately, we reviewed 238 articles, including them in this review. Each of the reviewed compounds, including vitamin A, vitamin C, vitamin D, vitamin E, curcumin, chlorella, Omega-3, biotin,Ppolypodium leucotomos, Simmondsia chinesis, gamma oryzanol, olive leaf extract, spirulina, and astaxanthin, was observed to present some possible effects with promising benefits for a skin condition, i.e., photoprotective radiation. Adding them to the diet or daily routine might have a positive influence on some skin inflammatory diseases such as atopic dermatitis or psoriasis. Further, UV radiation protection facilitated by some supplements and their impact on human cells might be helpful during chemotherapy or in preventing melanoma development. Further research is needed because of the lack of clear consensus regarding the doses of the described compounds that could provide desirable effects on the skin.
Collapse
Affiliation(s)
- Jacek Januszewski
- Chair and Department of Anatomy, Medical University of Lublin, 20-090 Lublin, Poland; (J.J.); (M.T.); (J.C.D.); (K.Ś.)
| | - Alicja Forma
- Department of Forensic Medicine, Medical University of Lublin, ul. Jaczewskiego 8b, 20-090 Lublin, Poland; (M.F.); (I.D.)
| | - Julita Zembala
- University Clinical Center, Medical University of Warsaw, Lindleya 4, 02-004 Warsaw, Poland;
| | - Michał Flieger
- Department of Forensic Medicine, Medical University of Lublin, ul. Jaczewskiego 8b, 20-090 Lublin, Poland; (M.F.); (I.D.)
| | - Magdalena Tyczyńska
- Chair and Department of Anatomy, Medical University of Lublin, 20-090 Lublin, Poland; (J.J.); (M.T.); (J.C.D.); (K.Ś.)
| | - James Curtis Dring
- Chair and Department of Anatomy, Medical University of Lublin, 20-090 Lublin, Poland; (J.J.); (M.T.); (J.C.D.); (K.Ś.)
| | - Iga Dudek
- Department of Forensic Medicine, Medical University of Lublin, ul. Jaczewskiego 8b, 20-090 Lublin, Poland; (M.F.); (I.D.)
| | - Kamila Świątek
- Chair and Department of Anatomy, Medical University of Lublin, 20-090 Lublin, Poland; (J.J.); (M.T.); (J.C.D.); (K.Ś.)
| | - Jacek Baj
- Chair and Department of Anatomy, Medical University of Lublin, 20-090 Lublin, Poland; (J.J.); (M.T.); (J.C.D.); (K.Ś.)
| |
Collapse
|
13
|
Guidotti IL, Neis A, Martinez DP, Seixas FK, Machado K, Kremer FS. Bambu and its applications in the discovery of active molecules against melanoma. J Mol Graph Model 2023; 124:108564. [PMID: 37453311 DOI: 10.1016/j.jmgm.2023.108564] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2023] [Revised: 06/14/2023] [Accepted: 07/10/2023] [Indexed: 07/18/2023]
Abstract
PURPOSE OR OBJECTIVE Melanoma is one of the most dangerous forms of skin cancer and the discovery of novel drugs is an ongoing effort. Quantitative Structure Activity Relationship (QSAR) is a computational method that allows the estimation of the properties of a molecule, including its biological activity. QSAR models have been widely employed in the search for potential drug candidates, but also for agrochemicals and other molecules with applications in different branches of the industry. Here we present Bambu, a simple command line tool to generate QSAR models from high-throughput screening bioassays datasets. METHODS The tool was developed using the Python programming language and relies mainly on RDKit for molecule data manipulation, FLAML for automated machine learning and the PubChem REST API for data retrieval. As a proof-of-concept we have employed the tool to generate QSAR models for melanoma cell growth inhibition based on HTS data and used them to screen libraries of FDA-approved drugs and natural compounds. Additionally, Bambu was compared to QSAR-Co, another automated tool for QSAR model generation. RESULTS based on the developed tool we were able to produce QSAR models and identify a wide variety of molecules with potential melanoma cell growth inhibitors, many of which with anti-tumoral activity already described. The QSAR models are available through the URL http://caramel.ufpel.edu.br, and all data and code used to generate its models are available at Zenodo (https://doi.org/10.5281/zenodo.7495214). Bambu source code is available at GitHub (https://github.com/omixlab/bambu-v2). In the benchmark, Bambu was able to produce models with higher accuracy, recall, F1 and ROC AUC when compared to QSAR-Co for the selected datasets. CONCLUSIONS Bambu is an free and open source tool which facilitates the creation of QSAR models and can be futurely applied in a wide variety of drug discovery projects.
Collapse
Affiliation(s)
- Isadora Leitzke Guidotti
- Centro de Desenvolvimento Tecnológico, Universidade Federal de Pelotas, Pelotas, Rio Grande do Sul, Brazil
| | - Alessandra Neis
- Centro de Desenvolvimento Tecnológico, Universidade Federal de Pelotas, Pelotas, Rio Grande do Sul, Brazil
| | - Daniela Peres Martinez
- Centro de Desenvolvimento Tecnológico, Universidade Federal de Pelotas, Pelotas, Rio Grande do Sul, Brazil
| | - Fabiana Kömmling Seixas
- Centro de Desenvolvimento Tecnológico, Universidade Federal de Pelotas, Pelotas, Rio Grande do Sul, Brazil
| | - Karina Machado
- Centro de Ciências Computacionais, Universidade Federal do Rio Grande, Rio Grande, Rio Grande do Sul, Brazil
| | - Frederico Schmitt Kremer
- Centro de Desenvolvimento Tecnológico, Universidade Federal de Pelotas, Pelotas, Rio Grande do Sul, Brazil.
| |
Collapse
|
14
|
Ma W, Zhang X, Zhuang L. Exogenous Hydrogen Sulfide Induces A375 Melanoma Cell Apoptosis Through Overactivation of the Unfolded Protein Response. Clin Cosmet Investig Dermatol 2023; 16:1641-1651. [PMID: 37396710 PMCID: PMC10314752 DOI: 10.2147/ccid.s412588] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2023] [Accepted: 06/10/2023] [Indexed: 07/04/2023]
Abstract
Purpose Melanomas are highly malignant and rapidly develop drug resistance due to dysregulated apoptosis. Therefore, pro-apoptotic agents could be effective for the management of melanoma. Hydrogen sulfide is ubiquitous in the body, and exogenous hydrogen sulfide has been reported to show inhibitory and pro-apoptotic effects on cancer cells. However, whether high concentrations of exogenous hydrogen sulfide have pro-apoptotic effects on melanoma and its mechanisms remain unknown. Hence, this study aimed to explore the pro-apoptotic effects and mechanisms of exogenous hydrogen sulfide on the A375 melanoma cell line treated with a hydrogen sulfide donor (NaHS). Methods The cell proliferation test, flow cytometric analysis, Hoechst 33258 staining, and Western blotting of B-cell lymphoma 2 and cleaved caspase-3 were used to explore the pro-apoptotic effects of hydrogen sulfide on A375 cells. The transcriptional profile of NaHS-treated A375 cells was further explored via high-throughput sequencing. Western blotting of phosphorylated inositol-requiring enzyme 1α (p-IRE1α), phosphorylated protein kinase R-like ER kinase (p-PERK), phosphorylated eukaryotic translation initiation factor 2α (p-eIF2α), C/EBP homologous protein, glucose-regulating protein 78, IRE1α, PERK, and eIF2α was performed to verify the changes in the transcriptional profile. Results NaHS inhibited A375 melanoma cell proliferation and induced apoptosis. The endoplasmic reticulum stress unfolded protein response and apoptosis-associated gene expression was upregulated in NaHS-treated A375 melanoma cells. The overactivation of the unfolded protein response and increase in endoplasmic reticulum stress was verified at the protein level. Conclusion Treatment with NaHS increased endoplasmic reticulum stress, which triggered the overactivation of the unfolded protein response and ultimately lead to melanoma cell apoptosis. The pro-apoptotic effect of NaHS suggests that it can be explored as a potential therapeutic agent in melanoma.
Collapse
Affiliation(s)
- Weiyuan Ma
- Department of Dermatology, Affiliated Hospital of Weifang Medical University, Weifang, Shandong Province, People’s Republic of China
| | - Xiuwen Zhang
- Department of Dermatology, Weihai Municipal Hospital, Weihai, Shandong Province, People’s Republic of China
| | - Le Zhuang
- Department of Dermatology, Central Hospital Affiliated to Shandong First Medical University, Jinan, Shandong Province, People’s Republic of China
| |
Collapse
|
15
|
Zhao Z, Ding Y, Tran LJ, Chai G, Lin L. Innovative breakthroughs facilitated by single-cell multi-omics: manipulating natural killer cell functionality correlates with a novel subcategory of melanoma cells. Front Immunol 2023; 14:1196892. [PMID: 37435067 PMCID: PMC10332463 DOI: 10.3389/fimmu.2023.1196892] [Citation(s) in RCA: 52] [Impact Index Per Article: 26.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2023] [Accepted: 06/12/2023] [Indexed: 07/13/2023] Open
Abstract
Background Melanoma is typically regarded as the most dangerous form of skin cancer. Although surgical removal of in situ lesions can be used to effectively treat metastatic disease, this condition is still difficult to cure. Melanoma cells are removed in great part due to the action of natural killer (NK) and T cells on the immune system. Still, not much is known about how the activity of NK cell-related pathways changes in melanoma tissue. Thus, we performed a single-cell multi-omics analysis on human melanoma cells in this study to explore the modulation of NK cell activity. Materials and methods Cells in which mitochondrial genes comprised > 20% of the total number of expressed genes were removed. Gene ontology (GO), gene set enrichment analysis (GSEA), gene set variation analysis (GSVA), and AUCcell analysis of differentially expressed genes (DEGs) in melanoma subtypes were performed. The CellChat package was used to predict cell-cell contact between NK cell and melanoma cell subtypes. Monocle program analyzed the pseudotime trajectories of melanoma cells. In addition, CytoTRACE was used to determine the recommended time order of melanoma cells. InferCNV was utilized to calculate the CNV level of melanoma cell subtypes. Python package pySCENIC was used to assess the enrichment of transcription factors and the activity of regulons in melanoma cell subtypes. Furthermore, the cell function experiment was used to confirm the function of TBX21 in both A375 and WM-115 melanoma cell lines. Results Following batch effect correction, 26,161 cells were separated into 28 clusters and designated as melanoma cells, neural cells, fibroblasts, endothelial cells, NK cells, CD4+ T cells, CD8+ T cells, B cells, plasma cells, monocytes and macrophages, and dendritic cells. A total of 10137 melanoma cells were further grouped into seven subtypes, i.e., C0 Melanoma BIRC7, C1 Melanoma CDH19, C2 Melanoma EDNRB, C3 Melanoma BIRC5, C4 Melanoma CORO1A, C5 Melanoma MAGEA4, and C6 Melanoma GJB2. The results of AUCell, GSEA, and GSVA suggested that C4 Melanoma CORO1A may be more sensitive to NK and T cells through positive regulation of NK and T cell-mediated immunity, while other subtypes of melanoma may be more resistant to NK cells. This suggests that the intratumor heterogeneity (ITH) of melanoma-induced activity and the difference in NK cell-mediated cytotoxicity may have caused NK cell defects. Transcription factor enrichment analysis indicated that TBX21 was the most important TF in C4 Melanoma CORO1A and was also associated with M1 modules. In vitro experiments further showed that TBX21 knockdown dramatically decreases melanoma cells' proliferation, invasion, and migration. Conclusion The differences in NK and T cell-mediated immunity and cytotoxicity between C4 Melanoma CORO1A and other melanoma cell subtypes may offer a new perspective on the ITH of melanoma-induced metastatic activity. In addition, the protective factors of skin melanoma, STAT1, IRF1, and FLI1, may modulate melanoma cell responses to NK or T cells.
Collapse
Affiliation(s)
- Zhijie Zhao
- Department of Plastic and Reconstructive Surgery, Shanghai Ninth People’s Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Yantao Ding
- Department of Dermatology, The First Affiliated Hospital, Institute of Dermatology, Anhui Medical University, Hefei, Anhui, China
- China Key Laboratory of Dermatology, Ministry of Education, Anhui Medical University, Hefei, Anhui, China
| | - Lisa Jia Tran
- Department of General, Visceral, and Transplant Surgery, Ludwig-Maximilians-University Munich, Munich, Germany
| | - Gang Chai
- Department of Plastic and Reconstructive Surgery, Shanghai Ninth People’s Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Li Lin
- Department of Plastic and Reconstructive Surgery, Shanghai Ninth People’s Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Shanghai Jiao Tong University School of Medicine, Shanghai, China
| |
Collapse
|
16
|
Foster HM, Carle MN, Jira LR, Koh DW. TRPM2 Channels: A Potential Therapeutic Target in Melanoma? Int J Mol Sci 2023; 24:10437. [PMID: 37445615 DOI: 10.3390/ijms241310437] [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: 04/30/2023] [Revised: 06/09/2023] [Accepted: 06/15/2023] [Indexed: 07/15/2023] Open
Abstract
The transient receptor potential, the melastatin (TRPM) subfamily, which consists of eight known members, appears to have significant importance in melanoma progression, treatment, and prognosis. As several members were originally cloned from cancerous tissue, initial studies aimed towards identifying TRPM involvement in cancer progression and tumorigenesis. For relevance in skin cancer, previous research has shown roles for several TRPM members in skin cancer progression, growth, and patient prognosis. One unique member, TRPM2, appears to have notable therapeutic potential in the treatment of melanoma. Previous and recent studies have demonstrated increased TRPM2 expression levels in melanoma, as well as important roles for TRPM2 in melanoma growth, proliferation, and survival. TRPM2 is thus an emerging target in the treatment of melanoma, where TRPM2 antagonism may offer an additional treatment option for melanoma patients in the future.
Collapse
Affiliation(s)
- Hattie M Foster
- Department of Pharmaceutical & Biomedical Sciences, Ohio Northern University, Ada, OH 45810, USA
| | - McKenzie N Carle
- Department of Pharmaceutical & Biomedical Sciences, Ohio Northern University, Ada, OH 45810, USA
| | - Lukas R Jira
- Department of Pharmaceutical & Biomedical Sciences, Ohio Northern University, Ada, OH 45810, USA
| | - David W Koh
- Department of Pharmaceutical & Biomedical Sciences, Ohio Northern University, Ada, OH 45810, USA
| |
Collapse
|
17
|
Cunha C, Marinheiro D, Ferreira BJML, Oliveira H, Daniel-da-Silva AL. Morin Hydrate Encapsulation and Release from Mesoporous Silica Nanoparticles for Melanoma Therapy. Molecules 2023; 28:4776. [PMID: 37375331 DOI: 10.3390/molecules28124776] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2023] [Revised: 06/12/2023] [Accepted: 06/13/2023] [Indexed: 06/29/2023] Open
Abstract
Melanoma incidence, a type of skin cancer, has been increasing worldwide. There is a strong need to develop new therapeutic strategies to improve melanoma treatment. Morin is a bioflavonoid with the potential for use in the treatment of cancer, including melanoma. However, therapeutic applications of morin are restrained owing to its low aqueous solubility and limited bioavailability. This work investigates morin hydrate (MH) encapsulation in mesoporous silica nanoparticles (MSNs) to enhance morin bioavailability and consequently increase the antitumor effects in melanoma cells. Spheroidal MSNs with a mean size of 56.3 ± 6.5 nm and a specific surface area of 816 m2/g were synthesized. MH was successfully loaded (MH-MSN) using the evaporation method, with a loading capacity of 28.3% and loading efficiency of 99.1%. In vitro release studies showed that morin release from MH-MSNs was enhanced at pH 5.2, indicating increased flavonoid solubility. The in vitro cytotoxicity of MH and MH-MSNs on human A375, MNT-1 and SK-MEL-28 melanoma cell lines was investigated. Exposure to MSNs did not affect the cell viability of any of the cell lines tested, suggesting that the nanoparticles are biocompatible. The effect of MH and MH-MSNs on reducing cell viability was time- and concentration-dependent in all melanoma cell lines. The A375 and SK-MEL-28 cell lines were slightly more sensitive than MNT-1 cells in both the MH and MH-MSN treatments. Our findings suggest that MH-MSNs are a promising delivery system for the treatment of melanoma.
Collapse
Affiliation(s)
- Catarina Cunha
- Department of Biology, CESAM-Centre for Environmental and Marine Studies, University of Aveiro, 3810-193 Aveiro, Portugal
- Department of Chemistry, CICECO-Aveiro Institute of Materials, University of Aveiro, 3810-193 Aveiro, Portugal
| | - Diogo Marinheiro
- Department of Chemistry, CICECO-Aveiro Institute of Materials, University of Aveiro, 3810-193 Aveiro, Portugal
| | - Bárbara J M L Ferreira
- Department of Chemistry, CICECO-Aveiro Institute of Materials, University of Aveiro, 3810-193 Aveiro, Portugal
| | - Helena Oliveira
- Department of Biology, CESAM-Centre for Environmental and Marine Studies, University of Aveiro, 3810-193 Aveiro, Portugal
| | - Ana L Daniel-da-Silva
- Department of Chemistry, CICECO-Aveiro Institute of Materials, University of Aveiro, 3810-193 Aveiro, Portugal
| |
Collapse
|
18
|
Rahimi A, Esmaeili Y, Dana N, Dabiri A, Rahimmanesh I, Jandaghain S, Vaseghi G, Shariati L, Zarrabi A, Javanmard SH, Cordani M. A comprehensive review on novel targeted therapy methods and nanotechnology-based gene delivery systems in melanoma. Eur J Pharm Sci 2023:106476. [PMID: 37236377 DOI: 10.1016/j.ejps.2023.106476] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2023] [Revised: 05/17/2023] [Accepted: 05/22/2023] [Indexed: 05/28/2023]
Abstract
Melanoma, a malignant form of skin cancer, has been swiftly increasing in recent years. Although there have been significant advancements in clinical treatment underlying a well-understanding of melanoma-susceptible genes and the molecular basis of melanoma pathogenesis, the permanency of response to therapy is frequently constrained by the emergence of acquired resistance and systemic toxicity. Conventional therapies, including surgical resection, chemotherapy, radiotherapy, and immunotherapy, have already been used to treat melanoma and are dependent on the cancer stage. Nevertheless, ineffective side effects and the heterogeneity of tumors pose major obstacles to the therapeutic treatment of malignant melanoma through such strategies. In light of this, advanced therapies including nucleic acid therapies (ncRNA, aptamers), suicide gene therapies, and gene therapy using tumor suppressor genes, have lately gained immense attention in the field of cancer treatment. Furthermore, nanomedicine and targeted therapy based on gene editing tools have been applied to the treatment of melanoma as potential cancer treatment approaches nowadays. Indeed, nanovectors enable delivery of the therapeutic agents into the tumor sites by passive or active targeting, improving therapeutic efficiency and minimizing adverse effects. Accordingly, in this review, we summarized the recent findings related to novel targeted therapy methods as well as nanotechnology-based gene systems in melanoma. We also discussed current issues along with potential directions for future research, paving the way for the next-generation of melanoma treatments.
Collapse
Affiliation(s)
- Azadeh Rahimi
- Applied Physiology Research Center, Cardiovascular Research Institute, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Yasaman Esmaeili
- Biosensor Research Center, School of Advanced Technologies in Medicine, Isfahan University of Medical Sciences, Isfahan 8174673461, Iran
| | - Nasim Dana
- Applied Physiology Research Center, Cardiovascular Research Institute, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Arezou Dabiri
- Applied Physiology Research Center, Cardiovascular Research Institute, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Ilnaz Rahimmanesh
- Applied Physiology Research Center, Cardiovascular Research Institute, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Setareh Jandaghain
- Applied Physiology Research Center, Cardiovascular Research Institute, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Golnaz Vaseghi
- Applied Physiology Research Center, Cardiovascular Research Institute, Isfahan University of Medical Sciences, Isfahan, Iran; Isfahan Cardiovascular Research Center, Cardiovascular Research Institute, Isfahan University of Medical Sciences, Isfahan 8158388994, Iran
| | - Laleh Shariati
- Department of Biomaterials, Nanotechnology and Tissue Engineering, School of Advanced Technologies in Medicine, Isfahan University of Medical Sciences, Isfahan 8174673461, Iran
| | - Ali Zarrabi
- Department of Biomedical Engineering, Faculty of Engineering & Natural Sciences, Istinye University, Istanbul 34396, Turkey
| | - Shaghayegh Haghjooy Javanmard
- Applied Physiology Research Center, Cardiovascular Research Institute, Isfahan University of Medical Sciences, Isfahan, Iran.
| | - Marco Cordani
- Department of Biochemistry and Molecular Biology, Faculty of Biology, Complutense University, 28040 Madrid, Spain; Instituto de Investigaciones Sanitarias San Carlos (IdISSC), 28040 Madrid, Spain.
| |
Collapse
|
19
|
Mukherjee S, Bhatti GK, Chhabra R, Reddy PH, Bhatti JS. Targeting mitochondria as a potential therapeutic strategy against chemoresistance in cancer. Biomed Pharmacother 2023; 160:114398. [PMID: 36773523 DOI: 10.1016/j.biopha.2023.114398] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2022] [Revised: 01/21/2023] [Accepted: 02/08/2023] [Indexed: 02/11/2023] Open
Abstract
The importance of mitochondria is not only limited to energy generation but also in several physical and chemical processes critical for cell survival. Mitochondria play an essential role in cellular apoptosis, calcium ion transport and cellular metabolism. Mutation in the nuclear and mitochondrial genes, altered oncogenes/tumor suppressor genes, and deregulated signalling for cell viability are major reasons for cancer progression and chemoresistance. The development of drug resistance in cancer patients is a major challenge in cancer treatment as the resistant cells are often more aggressive. The drug resistant cells of numerous cancer types exhibit the deregulation of mitochondrial function. The increased biogenesis of mitochondria and its dynamic alteration contribute to developing resistance. Further, a small subpopulation of cancer stem cells in the heterogeneous tumor is primarily responsible for chemoresistance and has an attribute of mitochondrial dysfunction. This review highlights the critical role of mitochondrial dysfunction in chemoresistance in cancer cells through the processes of apoptosis, autophagy/mitophagy, and cancer stemness. Mitochondria-targeted therapeutic strategies might help reduce cancer progression and chemoresistance induced by various cancer drugs.
Collapse
Affiliation(s)
- Soumi Mukherjee
- Laboratory of Translational Medicine and Nanotherapeutics, Department of Human Genetics and Molecular Medicine, School of Health Sciences, Central University of Punjab, Bathinda, India.
| | - Gurjit Kaur Bhatti
- Department of Medical Lab Technology, University Institute of Applied Health Sciences, Chandigarh University, Mohali, India.
| | - Ravindresh Chhabra
- Department of Biochemistry, School of Basic Sciences, Central University of Punjab, Bathinda, India.
| | - P Hemachandra Reddy
- Department of Internal Medicine, Texas Tech University Health Sciences Center, Lubbock, TX 79430, USA; Department of Pharmacology and Neuroscience and Garrison Institute on Aging, Texas Tech University Health Sciences Center, Lubbock, TX 79430, USA; Department of Public Health, Graduate School of Biomedical Sciences, Texas Tech University Health Sciences Center, Lubbock, TX 79430, USA; Department of Neurology, Texas Tech University Health Sciences Center, Lubbock, TX 79430, USA; Department of Speech, Language, and Hearing Sciences, Texas Tech University Health Sciences Center, Lubbock, TX 79430, USA.
| | - Jasvinder Singh Bhatti
- Laboratory of Translational Medicine and Nanotherapeutics, Department of Human Genetics and Molecular Medicine, School of Health Sciences, Central University of Punjab, Bathinda, India.
| |
Collapse
|
20
|
Jędrzejewski T, Pawlikowska M, Sobocińska J, Wrotek S. COVID-19 and Cancer Diseases-The Potential of Coriolus versicolor Mushroom to Combat Global Health Challenges. Int J Mol Sci 2023; 24:ijms24054864. [PMID: 36902290 PMCID: PMC10003402 DOI: 10.3390/ijms24054864] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2022] [Revised: 02/09/2023] [Accepted: 02/28/2023] [Indexed: 03/06/2023] Open
Abstract
Coriolus versicolor (CV) is a common species from the Polyporaceae family that has been used in traditional Chinese herbal medicine for over 2000 years. Among well-described and most active compounds identified in CV are polysaccharopeptides, such as polysaccharide peptide (PSP) and Polysaccharide-K (PSK, krestin), which, in some countries, are already used as an adjuvant agent in cancer therapy. In this paper, research advances in the field of anti-cancer and anti-viral action of CV are analyzed. The results of data obtained in in vitro and in vivo studies using animal models as well as in clinical research trials have been discussed. The present update provides a brief overview regarding the immunomodulatory effects of CV. A particular focus has been given to the mechanisms of direct effects of CV on cancer cells and angiogenesis. A potential use of CV compounds in anti-viral treatment, including therapy against COVID-19 disease, has also been analyzed based on the most recent literature. Additionally, the significance of fever in viral infection and cancer has been debated, providing evidence that CV affects this phenomenon.
Collapse
|
21
|
Malakoutikhah Z, Mohajeri Z, Dana N, Haghjooy Javanmard S. The dual role of Nrf2 in melanoma: a systematic review. BMC Mol Cell Biol 2023; 24:5. [PMID: 36747120 PMCID: PMC9900951 DOI: 10.1186/s12860-023-00466-5] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2022] [Accepted: 02/01/2023] [Indexed: 02/08/2023] Open
Abstract
Melanoma is the most lethal type of skin cancer that originates from the malignant transformation of melanocytes. Although novel treatments have improved patient survival in melanoma, the overall prognosis remains poor. To improve current therapies and patients outcome, it is necessary to identify the influential elements in the development and progression of melanoma.Due to UV exposure and melanin synthesis, the melanocytic lineage seems to have a higher rate of ROS (reactive oxygen species) formation. Melanoma has been linked to an increased oxidative state, and all facets of melanoma pathophysiology rely on redox biology. Several redox-modulating pathways have arisen to resist oxidative stress. One of which, the Nrf2 (nuclear factor erythroid 2-related factor 2), has been recognized as a master regulator of cellular response to oxidative or electrophilic challenges. The activation of Nrf2 signaling causes a wide range of antioxidant and detoxification enzyme genes to be expressed. As a result, this transcription factor has lately received a lot of interest as a possible cancer treatment target.On the other hand, Nrf2 has been found to have a variety of activities in addition to its antioxidant abilities, constant Nrf2 activation in malignant cells may accelerate metastasis and chemoresistance. Hence, based on the cell type and context, Nrf2 has different roles in either preventing or promoting cancer. In this study, we aimed to systematically review all the studies discussing the function of Nrf2 in melanoma and the factors determining its alteration.
Collapse
Affiliation(s)
- Zahra Malakoutikhah
- Applied Physiology Research Center, Cardiovascular Research Institute, Isfahan University of Medical Sciences, Isfahan, Iran.
| | - Zahra Mohajeri
- Applied Physiology Research Center, Cardiovascular Research Institute, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Nasim Dana
- Applied Physiology Research Center, Cardiovascular Research Institute, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Shaghayegh Haghjooy Javanmard
- Applied Physiology Research Center, Cardiovascular Research Institute, Isfahan University of Medical Sciences, Isfahan, Iran.
| |
Collapse
|
22
|
ARMS-NF-κB signaling regulates intracellular ROS to induce autophagy-associated cell death upon oxidative stress. iScience 2023; 26:106005. [PMID: 36798436 PMCID: PMC9926119 DOI: 10.1016/j.isci.2023.106005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2022] [Revised: 11/23/2022] [Accepted: 01/13/2023] [Indexed: 01/19/2023] Open
Abstract
Ankyrin repeat-rich membrane spanning (ARMS) plays roles in neural development, neuropathies, and tumor formation. Such pleiotropic function of ARMS is often attributed to diverse ARMS-interacting molecules in different cell context. However, it might be achieved by ARMS' effect on global biological mediator like reactive oxygen species (ROS). We established ARMS-knockdown in melanoma cells (siARMS) and in Drosophila eyes (GMR>dARMS RNAi ) and challenged them with H2O2. Decreased ARMS in both systems compromises nuclear translocation of NF-κB and induces ROS, which in turn augments autophagy flux and confers susceptibility to H2O2-triggered autophagic cell death. Resuming NF-κB activity or reducing ROS by antioxidants in siARMS cells and GMR>dARMS RNAi fly decreases intracellular peroxides level concurrent with reduced autophagy and attenuated cell death. Conversely, blocking NF-κB activity in wild-type flies/melanoma enhances ROS and induces autophagy with cell death. We thus uncover intracellular ROS modulated by ARMS-NFκB signaling primes autophagy for autophagic cell death upon oxidative stress.
Collapse
|
23
|
Sun Y, Lei S, Luo X, Jiang C, Li Z. The value of cuproptosis-related differential genes in guiding prognosis and immune status in patients with skin cutaneous melanoma. Front Pharmacol 2023; 14:1129544. [PMID: 37138850 PMCID: PMC10149708 DOI: 10.3389/fphar.2023.1129544] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2022] [Accepted: 04/03/2023] [Indexed: 05/05/2023] Open
Abstract
Background: Skin cutaneous melanoma (SKCM) is one of the most common cutaneous malignancies, which incidence is increasing. Cuproptosis is a new type of programming cell death recently reported, which may affect the progression of SKCM. Method: The mRNA expression data of melanoma were obtained from the Gene Expression Omnibus and the Cancer Genome Atlas databases. We constructed a prognostic model according to the cuproptosis-related differential genes in SKCM. Finally, real-time quantitative PCR was performed to verify the expression of cuproptosis-related differential genes in patients with different stages of cutaneous melanoma. Results: We detected 767 cuproptosis-related differential genes based on 19 cuproptosis-related genes, and screened out 7 differential genes to construct a prognostic model, which including three high-risk differential genes (SNAI2, RAP1GAP, BCHE), and four low-risk differential genes (JSRP1, HAPLN3, HHEX, ERAP2). Kaplan-Meier analysis indicated that SKCM patients with low-risk differential genes signals had better prognosis. The Encyclopedia of Genomes results manifested that cuproptosis-related differential genes are not only involved in T cell receptor signaling channel, natural killer cell mediated cytotoxicity, but also chemokine signaling pathway and B cell receptor signaling pathway. In our risk scoring model, the receiver operating characteristic (ROC) values of the three-time nodes are 0.669 (1-year), 0.669 (3-year) and 0.685 (5-year), respectively. Moreover, the tumor burden mutational and immunology function, cell stemness characteristics and drug sensitivity have significant differences between low-risk group and high-risk group. The mRNA level of SNAI2, RAP1GAP and BCHE in stage Ⅲ+Ⅳ SKCM patients was significantly higher than that in stage Ⅰ+Ⅱ patients, while the level of JSRP1, HAPLN3, HHEX and ERAP2 in stage Ⅰ+Ⅱ SKCM patients was more remarkable higher than that in stage Ⅲ+Ⅳ SKCM patients. Conclusion: In summary, we suggest that cuproptosis can not only regulate the tumor immune microenvironment but also affect the prognosis of SKCM patients, and may offer a basic theory for SKCM patients survival studies and clinical decision-making with potentially therapeutic drugs.
Collapse
|
24
|
Fraxetin Interacts Additively with Cisplatin and Mitoxantrone, Antagonistically with Docetaxel in Various Human Melanoma Cell Lines-An Isobolographic Analysis. Int J Mol Sci 2022; 24:ijms24010212. [PMID: 36613654 PMCID: PMC9820609 DOI: 10.3390/ijms24010212] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2022] [Revised: 12/13/2022] [Accepted: 12/19/2022] [Indexed: 12/25/2022] Open
Abstract
Malignant melanoma is a skin cancer characterized by rapid development, poor prognosis and high mortality. Due to the frequent drug resistance and/or early metastases in melanoma, new therapeutic methods are urgently needed. The study aimed at assessing the cytotoxic and antiproliferative effects of scoparone and fraxetin in vitro, when used alone and in combination with three cytostatics: cisplatin, mitoxantrone, and docetaxel in four human melanoma cell lines. Our experiments showed that scoparone in the concentration range tested up to 200 µM had no significant effect on the viability of human malignant melanoma (therefore, it was not possible to evaluate it in combination with other cytostatics), while fraxetin inhibited cell proliferation with IC50 doses in the range of 32.42-73.16 µM, depending on the cell line. Isobolographic analysis allowed for the assessment of the interactions between the studied compounds. Importantly, fraxetin was not cytotoxic to normal keratinocytes (HaCaT) and melanocytes (HEMa-LP), although it slightly inhibited their viability at high concentrations. The combination of fraxetin with cisplatin and mitoxantrone showed the additive interaction, which seems to be a promising direction in melanoma therapy. Unfortunately, the combination of fraxetin with docetaxel may not be beneficial due to the antagonistic antiproliferative effect of both drugs used in the mixture.
Collapse
|
25
|
Bharti V, Watkins R, Kumar A, Shattuck-Brandt RL, Mossing A, Mittra A, Shen C, Tsung A, Davies AE, Hanel W, Reneau JC, Chung C, Sizemore GM, Richmond A, Weiss VL, Vilgelm AE. BCL-xL inhibition potentiates cancer therapies by redirecting the outcome of p53 activation from senescence to apoptosis. Cell Rep 2022; 41:111826. [PMID: 36543138 PMCID: PMC10030045 DOI: 10.1016/j.celrep.2022.111826] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2022] [Revised: 10/26/2022] [Accepted: 11/21/2022] [Indexed: 12/24/2022] Open
Abstract
Cancer therapies trigger diverse cellular responses, ranging from apoptotic death to acquisition of persistent therapy-refractory states such as senescence. Tipping the balance toward apoptosis could improve treatment outcomes regardless of therapeutic agent or malignancy. We find that inhibition of the mitochondrial protein BCL-xL increases the propensity of cancer cells to die after treatment with a broad array of oncology drugs, including mitotic inhibitors and chemotherapy. Functional precision oncology and omics analyses suggest that BCL-xL inhibition redirects the outcome of p53 transcriptional response from senescence to apoptosis, which likely occurs via caspase-dependent down-modulation of p21 and downstream cytostatic proteins. Consequently, addition of a BCL-2/xL inhibitor strongly improves melanoma response to the senescence-inducing drug targeting mitotic kinase Aurora kinase A (AURKA) in mice and patient-derived organoids. This study shows a crosstalk between the mitochondrial apoptotic pathway and cell cycle regulation that can be targeted to augment therapeutic efficacy in cancers with wild-type p53.
Collapse
Affiliation(s)
- Vijaya Bharti
- Department of Pathology, The Ohio State University, 460 W. 12th Avenue, Office 496, Columbus, OH, USA; The Ohio State University Comprehensive Cancer Center - Arthur G. James Cancer Hospital and Richard J. Solove Research Institute, Columbus, OH, USA
| | - Reese Watkins
- Department of Pathology, The Ohio State University, 460 W. 12th Avenue, Office 496, Columbus, OH, USA; The Ohio State University Comprehensive Cancer Center - Arthur G. James Cancer Hospital and Richard J. Solove Research Institute, Columbus, OH, USA
| | - Amrendra Kumar
- Department of Pathology, The Ohio State University, 460 W. 12th Avenue, Office 496, Columbus, OH, USA; The Ohio State University Comprehensive Cancer Center - Arthur G. James Cancer Hospital and Richard J. Solove Research Institute, Columbus, OH, USA
| | - Rebecca L Shattuck-Brandt
- Department of Veterans Affairs, Tennessee Valley Healthcare System, Nashville, TN, USA; Department of Pharmacology, Vanderbilt University School of Medicine, Nashville, TN, USA
| | - Alexis Mossing
- The Ohio State University Comprehensive Cancer Center - Arthur G. James Cancer Hospital and Richard J. Solove Research Institute, Columbus, OH, USA; Department of Radiation Oncology, The Ohio State University, Columbus, OH, USA
| | - Arjun Mittra
- The Ohio State University Comprehensive Cancer Center - Arthur G. James Cancer Hospital and Richard J. Solove Research Institute, Columbus, OH, USA; Division of Medical Oncology, The Ohio State University, Columbus, OH, USA
| | - Chengli Shen
- Department of Surgery, University of Virginia, Charlottesville, VA, USA
| | - Allan Tsung
- Department of Surgery, University of Virginia, Charlottesville, VA, USA
| | - Alexander E Davies
- Department of Veterinary Biosciences, The Ohio State University, Columbus, OH, USA
| | - Walter Hanel
- The Ohio State University Comprehensive Cancer Center - Arthur G. James Cancer Hospital and Richard J. Solove Research Institute, Columbus, OH, USA
| | - John C Reneau
- The Ohio State University Comprehensive Cancer Center - Arthur G. James Cancer Hospital and Richard J. Solove Research Institute, Columbus, OH, USA
| | - Catherine Chung
- Department of Pathology, The Ohio State University, 460 W. 12th Avenue, Office 496, Columbus, OH, USA; The Ohio State University Comprehensive Cancer Center - Arthur G. James Cancer Hospital and Richard J. Solove Research Institute, Columbus, OH, USA
| | - Gina M Sizemore
- The Ohio State University Comprehensive Cancer Center - Arthur G. James Cancer Hospital and Richard J. Solove Research Institute, Columbus, OH, USA; Department of Radiation Oncology, The Ohio State University, Columbus, OH, USA
| | - Ann Richmond
- Department of Veterans Affairs, Tennessee Valley Healthcare System, Nashville, TN, USA; Department of Pharmacology, Vanderbilt University School of Medicine, Nashville, TN, USA
| | - Vivian L Weiss
- Department of Pathology, Microbiology, and Immunology, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Anna E Vilgelm
- Department of Pathology, The Ohio State University, 460 W. 12th Avenue, Office 496, Columbus, OH, USA; The Ohio State University Comprehensive Cancer Center - Arthur G. James Cancer Hospital and Richard J. Solove Research Institute, Columbus, OH, USA.
| |
Collapse
|
26
|
Jung D, Shin S, Kang S, Jung I, Ryu S, Noh S, Choi S, Jeong J, Lee BY, Kim K, Kim CS, Yoon JH, Lee C, Bucher F, Kim Y, Im S, Song B, Yea K, Baek M. Reprogramming of T cell-derived small extracellular vesicles using IL2 surface engineering induces potent anti-cancer effects through miRNA delivery. J Extracell Vesicles 2022; 11:e12287. [PMID: 36447429 PMCID: PMC9709340 DOI: 10.1002/jev2.12287] [Citation(s) in RCA: 50] [Impact Index Per Article: 16.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2022] [Revised: 09/28/2022] [Accepted: 11/14/2022] [Indexed: 12/03/2022] Open
Abstract
T cell-derived small extracellular vesicles (sEVs) exhibit anti-cancer effects. However, their anti-cancer potential should be reinforced to enhance clinical applicability. Herein, we generated interleukin-2-tethered sEVs (IL2-sEVs) from engineered Jurkat T cells expressing IL2 at the plasma membrane via a flexible linker to induce an autocrine effect. IL2-sEVs increased the anti-cancer ability of CD8+ T cells without affecting regulatory T (Treg ) cells and down-regulated cellular and exosomal PD-L1 expression in melanoma cells, causing their increased sensitivity to CD8+ T cell-mediated cytotoxicity. Its effect on CD8+ T and melanoma cells was mediated by several IL2-sEV-resident microRNAs (miRNAs), whose expressions were upregulated by the autocrine effects of IL2. Among the miRNAs, miR-181a-3p and miR-223-3p notably reduced the PD-L1 protein levels in melanoma cells. Interestingly, miR-181a-3p increased the activity of CD8+ T cells while suppressing Treg cell activity. IL2-sEVs inhibited tumour progression in melanoma-bearing immunocompetent mice, but not in immunodeficient mice. The combination of IL2-sEVs and existing anti-cancer drugs significantly improved anti-cancer efficacy by decreasing PD-L1 expression in vivo. Thus, IL2-sEVs are potential cancer immunotherapeutic agents that regulate both immune and cancer cells by reprogramming miRNA levels.
Collapse
Affiliation(s)
- Dokyung Jung
- Department of Molecular Medicine, CMRI, Exosome Convergence Research Center (ECRC), School of MedicineKyungpook National UniversityDaeguRepublic of Korea
| | - Sanghee Shin
- Department of New BiologyDGISTDaeguRepublic of Korea
| | - Sung‐Min Kang
- Department of Molecular Medicine, CMRI, Exosome Convergence Research Center (ECRC), School of MedicineKyungpook National UniversityDaeguRepublic of Korea
| | - Inseong Jung
- Department of New BiologyDGISTDaeguRepublic of Korea
| | - Suyeon Ryu
- Department of Molecular Medicine, CMRI, Exosome Convergence Research Center (ECRC), School of MedicineKyungpook National UniversityDaeguRepublic of Korea
| | - Soojeong Noh
- Department of New BiologyDGISTDaeguRepublic of Korea
| | - Sung‐Jin Choi
- Department of New BiologyDGISTDaeguRepublic of Korea
| | - Jongwon Jeong
- Department of New BiologyDGISTDaeguRepublic of Korea
| | - Beom Yong Lee
- Department of New BiologyDGISTDaeguRepublic of Korea
| | - Kwang‐Soo Kim
- Department of New BiologyDGISTDaeguRepublic of Korea
| | | | - Jong Hyuk Yoon
- Department of Neural Development and DiseaseKorea Brain Research InstituteDaeguRepublic of Korea
| | - Chan‐Hyeong Lee
- Department of Molecular Medicine, CMRI, Exosome Convergence Research Center (ECRC), School of MedicineKyungpook National UniversityDaeguRepublic of Korea
| | - Felicitas Bucher
- Eye Center, Medical Center, Faculty of MedicineUniversity of FreiburgFreiburgGermany
| | - Yong‐Nyun Kim
- Division of Translational ScienceNational Cancer Center 323Ilsan‐ro, Ilsandong‐guGoyang‐siGyeonggi‐doRepublic of Korea
| | - Sin‐Hyeog Im
- Department of Life SciencesPohang University of Science and Technology (POSTECH)Gyeongsangbuk‐doRepublic of Korea
- Institute of Convergence ScienceYonsei UniversitySeoulRepublic of Korea
- ImmunoBiomePohangRepublic of Korea
| | - Byoung‐Joon Song
- Section of Molecular Pharmacology and Toxicology, Laboratory of Membrane Biochemistry and BiophysicsNational Institute on Alcohol Abuse and Alcoholism (NIAAA)BethesdaMarylandUSA
| | - Kyungmoo Yea
- Department of New BiologyDGISTDaeguRepublic of Korea
- New Biology Research CenterDGISTDaeguRepublic of Korea
| | - Moon‐Chang Baek
- Department of Molecular Medicine, CMRI, Exosome Convergence Research Center (ECRC), School of MedicineKyungpook National UniversityDaeguRepublic of Korea
| |
Collapse
|
27
|
Carotenuto P, Romano A, Barbato A, Quadrano P, Brillante S, Volpe M, Ferrante L, Tammaro R, Morleo M, De Cegli R, Iuliano A, Testa M, Andreone F, Ciliberto G, Clery E, Troncone G, Palma G, Arra C, Barbieri A, Capone M, Madonna G, Ascierto PA, Lanfrancone L, Indrieri A, Franco B. Targeting the MITF/APAF-1 axis as salvage therapy for MAPK inhibitors in resistant melanoma. Cell Rep 2022; 41:111601. [DOI: 10.1016/j.celrep.2022.111601] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2021] [Revised: 02/09/2022] [Accepted: 10/13/2022] [Indexed: 11/09/2022] Open
|
28
|
Cunha C, Daniel-da-Silva AL, Oliveira H. Drug Delivery Systems and Flavonoids: Current Knowledge in Melanoma Treatment and Future Perspectives. MICROMACHINES 2022; 13:1838. [PMID: 36363859 PMCID: PMC9693869 DOI: 10.3390/mi13111838] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 09/30/2022] [Revised: 10/18/2022] [Accepted: 10/25/2022] [Indexed: 06/16/2023]
Abstract
Melanoma is an aggressive form of skin cancer with a high prevalence in the population. An early diagnosis is crucial to cure this disease. Still, when this is not possible, combining potent pharmacological agents and effective drug delivery systems is essential to achieve optimal treatment and improve patients' quality of life. Nanotechnology application in biomedical sciences to encapsulate anticancer drugs, including flavonoids, in order to enhance therapeutic efficacy has attracted particular interest. Flavonoids have shown effectiveness against various types of cancers including in melanoma, but they show low aqueous solubility, low stability and very poor oral bioavailability. The utilization of novel drug delivery systems could increase flavonoid bioavailability, thereby potentiating its antitumor effects in melanoma. This review summarizes the potential of different flavonoids in melanoma treatment and the several nanosystems used to improve their biological activity, considering published information that reported improved biological and pharmacological properties of encapsulated flavonoids.
Collapse
Affiliation(s)
- Catarina Cunha
- Department of Biology, CESAM—Centre for Environmental and Marine Studies, University of Aveiro, 3810-193 Aveiro, Portugal
- Department of Chemistry, CICECO—Aveiro Institute of Materials, University of Aveiro, 3810-193 Aveiro, Portugal
| | - Ana L. Daniel-da-Silva
- Department of Chemistry, CICECO—Aveiro Institute of Materials, University of Aveiro, 3810-193 Aveiro, Portugal
| | - Helena Oliveira
- Department of Biology, CESAM—Centre for Environmental and Marine Studies, University of Aveiro, 3810-193 Aveiro, Portugal
| |
Collapse
|
29
|
Yang LK, Lin CX, Li SH, Liang JJ, Xiao LL, Xie GH, Liu HW, Liao X. Novel IKZF3 transcriptomic signature correlates with positive outcomes of skin cutaneous melanoma: A pan-cancer analysis. Front Genet 2022; 13:1036402. [PMID: 36353107 PMCID: PMC9638148 DOI: 10.3389/fgene.2022.1036402] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2022] [Accepted: 10/07/2022] [Indexed: 11/27/2022] Open
Abstract
To investigate the potential relationship between Ikaros family genes and skin cutaneous melanoma (SKCM), we undertook a pan-cancer analysis of the transcriptional signature and clinical data of melanoma through multiple databases. First, 10,327 transcriptomic samples from different cancers were included to determine the overall characteristics and clinical prognoses associated with Ikaros gene expression across cancer types. Second, differentially expressed genes analysis, prognostic evaluation, and gene set enrichment analysis were employed to investigate the role of Ikaros (IKZF) genes in SKCM. Third, we evaluated the relationship between Ikaros family genes and SKCM immune infiltrates and verified the findings using the GEO single-cell sequencing dataset. The results show that Ikaros genes were widely expressed among different cancer types with independently similar patterns as follows: 1. IKZF1 and IKZF3, and 2. IKZF2 and IKZF4–5. IKZF2 and IKZF5 were downregulated in the primary tumor, and IKZF1–3 expression decreased significantly as the T-stage or metastasis increased in SKCM. Moreover, high IKZF1–3 expression was associated with better overall survival, disease-specific survival, and progression-free interval. IKZF3 is an independent prognostic factor of SKCM. Among Ikaros genes, the expression of IKZF1 and IKZF3 positively correlated with the infiltration level of CD4+ T cells and CD8+ T cells, B cells, and Tregs in SKCM and negatively correlated with the infiltration level of M0 and M1 macrophages. Moreover, single-cell sequencing data analysis revealed that IKZF1 and IKZF3 were mainly expressed by immune cells. Correlation analysis shows the immune factors and drug responses associated with IKZF3 expression. In conclusion, the present study is the first, to our knowledge, to identify a pan-cancer genomic signature of the Ikaros gene family among different cancers. Expression of these family members, particularly high levels of IKZF3, indicate positive immunological status and beneficial clinical outcomes of SKCM. IKZF3 may therefore serve as potential targets for immunotherapy of melanoma.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | - Xuan Liao
- *Correspondence: Hong-Wei Liu, ; Xuan Liao,
| |
Collapse
|
30
|
Yang F, Bettadapura SN, Smeltzer MS, Zhu H, Wang S. Pyroptosis and pyroptosis-inducing cancer drugs. Acta Pharmacol Sin 2022; 43:2462-2473. [PMID: 35288674 PMCID: PMC9525650 DOI: 10.1038/s41401-022-00887-6] [Citation(s) in RCA: 78] [Impact Index Per Article: 26.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/14/2021] [Accepted: 02/07/2022] [Indexed: 02/07/2023]
Abstract
Pyroptosis, an inflammatory form of lytic cell death, is a type of cell death mediated by the gasdermin (GSDM) protein family. Upon recognizing exogenous or endogenous signals, cells undergo inflammasome assembly, GSDM cleavage, the release of proinflammatory cytokines and other cellular contents, eventually leading to inflammatory cell death. In this review, we discuss the roles of the GSDM family for anti-cancer functions and various antitumor drugs that could activate the pyroptosis pathways.
Collapse
Affiliation(s)
- Fan Yang
- Healthville LLC, Little Rock, AR, 72204, USA
| | - Sahana N Bettadapura
- Biology Department, University of Arkansas at Little Rock, Little Rock, AR, 72204, USA
| | - Mark S Smeltzer
- Department of Microbiology and Immunology, University of Arkansas for Medical Sciences, Little Rock, AR, 72205, USA
| | - Hua Zhu
- Department of Surgery, The Ohio State University Wexner Medical Center, Columbus, OH, 43210, USA.
| | - Shanzhi Wang
- Chemistry Department, University of Arkansas at Little Rock, Little Rock, AR, 72204, USA.
| |
Collapse
|
31
|
Evyapan G, Luleyap U, Kaplan HM, Kara IO. Ornidazole suppresses CD133+ melanoma stem cells via inhibiting hedgehog signaling pathway and inducing multiple death pathways in a mouse model. Croat Med J 2022; 63. [PMID: 36325671 PMCID: PMC9648086 DOI: 10.3325/cmj.2022.63.461] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022] Open
Abstract
AIM To evaluate the inhibitory effects of ornidazole on the proliferation and migration of metastatic melanoma cell line (B16F10) in vitro and its anti-cancer effects in vivo using a melanoma mouse model. METHODS We investigated the effects of ornidazole on cell viability (Crystal Violet and MTT assay) and migration ability (wound-healing assay) of B16F10 melanoma cells, and its ability to trigger DNA damage (Comet assay) in vitro. We also sorted CD133+ and CD133- cells from B16F10 melanoma cell line and injected them subcutaneously into Swiss albino mice to induce tumor formation. Tumor-bearing mice were divided into control and treatment groups. Treatment group received intraperitoneal ornidazole injections. Tumors were resected. Real-time polymerase chain reaction was used to determine the expression of genes involved into Sonic hedgehog (Shh) signaling pathway, stemness, apoptosis, endoplasmic reticulum (ER) stress, ER stress-mediated apoptosis, and autophagy. Shh signaling pathway-related proteins and CD133 protein were analyzed by ELISA. RESULTS Ornidazole effectively induced DNA damage in CD133+ melanoma cells and reduced their viability and migration ability in vitro. Moreover, it significantly suppressed tumor growth in melanoma mouse model seemingly by inhibiting the Shh signaling pathway and ER-stress mediated autophagy, as well as by activating multiple apoptosis pathways. CONCLUSIONS Our preclinical findings suggest the therapeutic potential of ornidazole in the treatment of metastatic melanoma. However, larger and more comprehensive studies are required to validate our results and to further explore the safety and clinical effectiveness of ornidazole.
Collapse
Affiliation(s)
- Gulsah Evyapan
- Department of Medical Biology, Cukurova University Faculty of Medicine, Adana, Turkey
| | - Umit Luleyap
- Department of Medical Biology, Cukurova University Faculty of Medicine, Adana, Turkey
| | - Halil Mahir Kaplan
- Department of Pharmacology, Cukurova University Faculty of Medicine, Adana, Turkey
| | - Ismail Oguz Kara
- Department of Medical Oncology, Cukurova University Faculty of Medicine, Adana, Turkey
| |
Collapse
|
32
|
Synthesis of naphthalimide derivatives bearing benzothiazole and thiazole moieties: In vitro anticancer and in silico ADMET study. J Mol Struct 2022. [DOI: 10.1016/j.molstruc.2022.133173] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022]
|
33
|
Montaseri H, Nkune NW, Abrahamse H. Active targeted photodynamic therapeutic effect of silver-based nanohybrids on melanoma cancer cells. JOURNAL OF PHOTOCHEMISTRY AND PHOTOBIOLOGY 2022. [DOI: 10.1016/j.jpap.2022.100136] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023] Open
|
34
|
Fernandes S, Vyas C, Lim P, Pereira RF, Virós A, Bártolo P. 3D Bioprinting: An Enabling Technology to Understand Melanoma. Cancers (Basel) 2022; 14:cancers14143535. [PMID: 35884596 PMCID: PMC9318274 DOI: 10.3390/cancers14143535] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2022] [Revised: 07/04/2022] [Accepted: 07/12/2022] [Indexed: 02/06/2023] Open
Abstract
Melanoma is a potentially fatal cancer with rising incidence over the last 50 years, associated with enhanced sun exposure and ultraviolet radiation. Its incidence is highest in people of European descent and the ageing population. There are multiple clinical and epidemiological variables affecting melanoma incidence and mortality, such as sex, ethnicity, UV exposure, anatomic site, and age. Although survival has improved in recent years due to advances in targeted and immunotherapies, new understanding of melanoma biology and disease progression is vital to improving clinical outcomes. Efforts to develop three-dimensional human skin equivalent models using biofabrication techniques, such as bioprinting, promise to deliver a better understanding of the complexity of melanoma and associated risk factors. These 3D skin models can be used as a platform for patient specific models and testing therapeutics.
Collapse
Affiliation(s)
- Samantha Fernandes
- Department of Mechanical, Aerospace and Civil Engineering, University of Manchester, Oxford Road, Manchester M13 9PL, UK; (S.F.); (C.V.); (P.L.)
| | - Cian Vyas
- Department of Mechanical, Aerospace and Civil Engineering, University of Manchester, Oxford Road, Manchester M13 9PL, UK; (S.F.); (C.V.); (P.L.)
- Singapore Centre for 3D Printing, School of Mechanical and Aerospace Engineering, Nanyang Technological University, Singapore 639798, Singapore
| | - Peggy Lim
- Department of Mechanical, Aerospace and Civil Engineering, University of Manchester, Oxford Road, Manchester M13 9PL, UK; (S.F.); (C.V.); (P.L.)
| | - Rúben F. Pereira
- ICBAS—Instituto de Ciências Biomédicas Abel Salazar, Universidade do Porto, 4050-313 Porto, Portugal;
- i3S—Instituto de Investigação e Inovação em Saúde, Universidade do Porto, 4200-135 Porto, Portugal
- INEB—Instituto de Engenharia Biomédica, Universidade do Porto, 4200-135 Porto, Portugal
| | - Amaya Virós
- Skin Cancer and Ageing Laboratory, Cancer Research UK Manchester Institute, University of Manchester, Oxford Road, Manchester M13 9PL, UK;
| | - Paulo Bártolo
- Department of Mechanical, Aerospace and Civil Engineering, University of Manchester, Oxford Road, Manchester M13 9PL, UK; (S.F.); (C.V.); (P.L.)
- Singapore Centre for 3D Printing, School of Mechanical and Aerospace Engineering, Nanyang Technological University, Singapore 639798, Singapore
- Correspondence: or
| |
Collapse
|
35
|
|
36
|
LINC00518 Promotes Cell Malignant Behaviors via Influencing EIF4A3-Mediated mRNA Stability of MITF in Melanoma. BIOMED RESEARCH INTERNATIONAL 2022; 2022:3546795. [PMID: 35813236 PMCID: PMC9262545 DOI: 10.1155/2022/3546795] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/20/2022] [Revised: 04/22/2022] [Accepted: 05/03/2022] [Indexed: 12/21/2022]
Abstract
Melanoma has become the most severe sort of skin cancer, deriving from the pigment-producing melanocytes. Existing research has validated that long noncoding RNAs (lncRNAs) have critical function in the progression of cancers. LINC00518 has been studied in cutaneous melanoma; however, the molecular mechanism of LINC00518 in melanoma needs in-depth investigation. In our study, LINC00518 was revealed to be upregulated in melanoma tissues and cells, and melanoma patients in high LINC00518 expression group had poorer prognosis as depicted in GEPIA database. Functional assays revealed that LINC00518 depletion inhibited cell proliferation, migration, invasion, and epithelial-mesenchymal transition (EMT). Furthermore, MITF was confirmed to be upregulated in melanoma tissues and cells, and melanoma patients in high MITF expression group had poorer prognosis as displayed in GEPIA database. MITF expression was positively connected to LINC00518 expression. Additionally, results of mechanism assays uncovered EIF4A3 could bind with LINC00518 and MITF, and LINC00518 recruited EIF4A3 to stabilize MITF mRNA. Finally, it was demonstrated that upregulation of MITF could partially abrogate the inhibitory impact of LINC00518 knockdown on melanoma cell malignant behaviors. To summarize, LINC00518 promotes the malignant processes of melanoma cells through targeting EIF4A3/MITF axis, which might provide novel potential biomarkers for melanoma prognosis.
Collapse
|
37
|
Marzęda P, Wróblewska-Łuczka P, Drozd M, Florek-Łuszczki M, Załuska-Ogryzek K, Łuszczki JJ. Cannabidiol Interacts Antagonistically with Cisplatin and Additively with Mitoxantrone in Various Melanoma Cell Lines-An Isobolographic Analysis. Int J Mol Sci 2022; 23:ijms23126752. [PMID: 35743195 PMCID: PMC9224300 DOI: 10.3390/ijms23126752] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2022] [Revised: 06/11/2022] [Accepted: 06/15/2022] [Indexed: 02/07/2023] Open
Abstract
The medical application of cannabidiol (CBD) has been gathering increasing attention in recent years. This non-psychotropic cannabis-derived compound possesses antiepileptic, antipsychotic, anti-inflammatory and anxiolytic properties. Recent studies report that it also exerts antineoplastic effects in multiple types of cancers, including melanoma. In this in vitro study we tried to reveal the anticancer properties of CBD in malignant melanoma cell lines (SK-MEL 28, A375, FM55P and FM55M2) administered alone, as well as in combination with mitoxantrone (MTX) or cisplatin (CDDP). The effects of CBD on the viability of melanoma cells were measured by the MTT assay; cytotoxicity was determined in the LDH test and proliferation in the BrdU test. Moreover, the safety of CBD was tested in human keratinocytes (HaCaT) in LDH and MTT tests. Results indicate that CBD reduces the viability and proliferation of melanoma-malignant cells and exerts additive interactions with MTX. Unfortunately, CBD produced antagonistic interaction when combined with CDDP. CBD does not cause significant cytotoxicity in HaCaT cell line. In conclusion, CBD may be considered as a part of melanoma multi-drug therapy when combined with MTX. A special attention should be paid to the combination of CBD with CDDP due to the antagonistic interaction observed in the studied malignant melanoma cell lines.
Collapse
Affiliation(s)
- Paweł Marzęda
- Department of Pathophysiology, Medical University of Lublin, 20-090 Lublin, Poland; (P.M.); (P.W.-Ł.); (M.D.); (K.Z.-O.)
| | - Paula Wróblewska-Łuczka
- Department of Pathophysiology, Medical University of Lublin, 20-090 Lublin, Poland; (P.M.); (P.W.-Ł.); (M.D.); (K.Z.-O.)
| | - Małgorzata Drozd
- Department of Pathophysiology, Medical University of Lublin, 20-090 Lublin, Poland; (P.M.); (P.W.-Ł.); (M.D.); (K.Z.-O.)
| | | | - Katarzyna Załuska-Ogryzek
- Department of Pathophysiology, Medical University of Lublin, 20-090 Lublin, Poland; (P.M.); (P.W.-Ł.); (M.D.); (K.Z.-O.)
| | - Jarogniew J. Łuszczki
- Department of Pathophysiology, Medical University of Lublin, 20-090 Lublin, Poland; (P.M.); (P.W.-Ł.); (M.D.); (K.Z.-O.)
- Correspondence: ; Tel.: +48-81-448-6500; Fax: +48-81-448-6501
| |
Collapse
|
38
|
Daoud M, Broxtermann PN, Schorn F, Werthenbach JP, Seeger JM, Schiffmann LM, Brinkmann K, Vucic D, Tüting T, Mauch C, Kulms D, Zigrino P, Kashkar H. XIAP promotes melanoma growth by inducing tumour neutrophil infiltration. EMBO Rep 2022; 23:e53608. [PMID: 35437868 PMCID: PMC9171690 DOI: 10.15252/embr.202153608] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2021] [Revised: 03/21/2022] [Accepted: 03/31/2022] [Indexed: 12/22/2022] Open
Abstract
Elevated expression of the X‐linked inhibitor of apoptosis protein (XIAP) has been frequently reported in malignant melanoma suggesting that XIAP renders apoptosis resistance and thereby supports melanoma progression. Independent of its anti‐apoptotic function, XIAP mediates cellular inflammatory signalling and promotes immunity against bacterial infection. The pro‐inflammatory function of XIAP has not yet been considered in cancer. By providing detailed in vitro analyses, utilising two independent mouse melanoma models and including human melanoma samples, we show here that XIAP is an important mediator of melanoma neutrophil infiltration. Neutrophils represent a major driver of melanoma progression and are increasingly considered as a valuable therapeutic target in solid cancer. Our data reveal that XIAP ubiquitylates RIPK2, involve TAB1/RIPK2 complex and induce the transcriptional up‐regulation and secretion of chemokines such as IL8, that are responsible for intra‐tumour neutrophil accumulation. Alteration of the XIAP‐RIPK2‐TAB1 inflammatory axis or the depletion of neutrophils in mice reduced melanoma growth. Our data shed new light on how XIAP contributes to tumour growth and provides important insights for novel XIAP targeting strategies in cancer.
Collapse
Affiliation(s)
- Mila Daoud
- Faculty of Medicine and University Hospital of Cologne, Institute for Molecular Immunology, University of Cologne, Cologne, Germany.,Faculty of Medicine and University Hospital of Cologne, Center for Molecular Medicine Cologne (CMMC), University of Cologne, Cologne, Germany.,Cologne Excellence Cluster on Cellular Stress Responses in Aging-Associated Diseases (CECAD), University of Cologne, Cologne, Germany
| | - Pia Nora Broxtermann
- Faculty of Medicine and University Hospital of Cologne, Institute for Molecular Immunology, University of Cologne, Cologne, Germany.,Faculty of Medicine and University Hospital of Cologne, Center for Molecular Medicine Cologne (CMMC), University of Cologne, Cologne, Germany.,Cologne Excellence Cluster on Cellular Stress Responses in Aging-Associated Diseases (CECAD), University of Cologne, Cologne, Germany
| | - Fabian Schorn
- Faculty of Medicine and University Hospital of Cologne, Institute for Molecular Immunology, University of Cologne, Cologne, Germany.,Faculty of Medicine and University Hospital of Cologne, Center for Molecular Medicine Cologne (CMMC), University of Cologne, Cologne, Germany.,Cologne Excellence Cluster on Cellular Stress Responses in Aging-Associated Diseases (CECAD), University of Cologne, Cologne, Germany
| | - J Paul Werthenbach
- Faculty of Medicine and University Hospital of Cologne, Institute for Molecular Immunology, University of Cologne, Cologne, Germany.,Faculty of Medicine and University Hospital of Cologne, Center for Molecular Medicine Cologne (CMMC), University of Cologne, Cologne, Germany.,Cologne Excellence Cluster on Cellular Stress Responses in Aging-Associated Diseases (CECAD), University of Cologne, Cologne, Germany
| | - Jens Michael Seeger
- Faculty of Medicine and University Hospital of Cologne, Institute for Molecular Immunology, University of Cologne, Cologne, Germany.,Faculty of Medicine and University Hospital of Cologne, Center for Molecular Medicine Cologne (CMMC), University of Cologne, Cologne, Germany.,Cologne Excellence Cluster on Cellular Stress Responses in Aging-Associated Diseases (CECAD), University of Cologne, Cologne, Germany
| | - Lars M Schiffmann
- Faculty of Medicine and University Hospital of Cologne, Institute for Molecular Immunology, University of Cologne, Cologne, Germany.,Cologne Excellence Cluster on Cellular Stress Responses in Aging-Associated Diseases (CECAD), University of Cologne, Cologne, Germany.,Faculty of Medicine and University Hospital of Cologne, Department of General, Visceral, Cancer and Transplant Surgery, University of Cologne, Cologne, Germany
| | - Kerstin Brinkmann
- The Walter & Eliza Hall Institute of Medical Research (WEHI) and Department of Medical Biology, University of Melbourne, Melbourne, Vic., Australia
| | - Domagoj Vucic
- Department of Early Discovery Biochemistry, Genentech, South San Francisco, CA, USA
| | - Thomas Tüting
- Laboratory of Experimental Dermatology, Department of Dermatology, University Hospital Magdeburg, Magdeburg, Germany
| | - Cornelia Mauch
- Faculty of Medicine and University Hospital of Cologne, Department of Dermatology and Venereology, University of Cologne, Cologne, Germany
| | - Dagmar Kulms
- Department of Dermatology, Experimental Dermatology, TU-Dresden, Dresden, Germany.,National Center for Tumor Diseases Dresden, TU-Dresden, Dresden, Germany
| | - Paola Zigrino
- Faculty of Medicine and University Hospital of Cologne, Department of Dermatology and Venereology, University of Cologne, Cologne, Germany
| | - Hamid Kashkar
- Faculty of Medicine and University Hospital of Cologne, Institute for Molecular Immunology, University of Cologne, Cologne, Germany.,Faculty of Medicine and University Hospital of Cologne, Center for Molecular Medicine Cologne (CMMC), University of Cologne, Cologne, Germany.,Cologne Excellence Cluster on Cellular Stress Responses in Aging-Associated Diseases (CECAD), University of Cologne, Cologne, Germany
| |
Collapse
|
39
|
Esmailzadeh A, Shanei A, Attaran N, Hejazi SH, Hemati S. Sonodynamic Therapy Using Dacarbazine-Loaded AuSiO 2 Nanoparticles for Melanoma Treatment: An In-Vitro Study on the B16F10 Murine Melanoma Cell Line. ULTRASOUND IN MEDICINE & BIOLOGY 2022; 48:1131-1142. [PMID: 35307236 DOI: 10.1016/j.ultrasmedbio.2022.02.015] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/12/2021] [Revised: 02/09/2022] [Accepted: 02/13/2022] [Indexed: 06/14/2023]
Abstract
The use of nanoparticles as a sonosensitizer in cancer sonodynamic therapy has been gaining attention because of their great advantages in drug delivery applications. By conjugating chemotherapy agents with nanoparticles, we can develop a drug delivery platform, control drug release and improve the outcome of treatments. The in-vitro study described here evaluates the combination of AuSiO2 nanoparticles and dacarbazine (DTIC@AuSiO2) as a sonosensitizer for sonodynamic therapy of melanoma. 3-(4,5-Dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide (MTT) and flow cytometry assays revealed that the viability of B16F10 melanoma cells was significantly inhibited by the increase in apoptosis induction in treatment with DTIC@AuSiO2 nanoparticles under ultrasound exposure compared with treatment with the free DTIC or AuSiO2 nanoparticles. The sonosensitization activity of AuSiO2 nanoparticles and greater uptake of DTIC by tumor cells after loading in DTIC@AuSiO2 nanoparticles inhibited the proliferation of melanoma tumor cells effectively. In conclusion, the DTIC@AuSiO2 nanoparticles established in this study could represent a good drug delivery and sonosensitizer platform for use in melanoma sonodynamic therapy.
Collapse
Affiliation(s)
- Arman Esmailzadeh
- Department of Medical Physics, School of Medicine, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Ahmad Shanei
- Department of Medical Physics, School of Medicine, Isfahan University of Medical Sciences, Isfahan, Iran.
| | - Neda Attaran
- Department of Medical Nanotechnology, Applied Biophotonics Research Center, Science and Research Branch, Islamic Azad University, Tehran, Iran
| | - Seyed Hossein Hejazi
- Department of Parasitology and Mycology, School of Medicine, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Simin Hemati
- Department of Radiation Oncology, School of Medicine, Seyyed Al-Shohada Hospital, Isfahan University of Medical Sciences, Isfahan, Iran
| |
Collapse
|
40
|
Hsu CH, Lee KJ, Chiu YH, Huang KC, Wang GS, Chen LP, Liao KW, Lin CS. The Lysosome in Malignant Melanoma: Biology, Function and Therapeutic Applications. Cells 2022; 11:1492. [PMID: 35563798 PMCID: PMC9103375 DOI: 10.3390/cells11091492] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2022] [Revised: 04/21/2022] [Accepted: 04/26/2022] [Indexed: 02/04/2023] Open
Abstract
Lysosomes are membrane-bound vesicles that play roles in the degradation and recycling of cellular waste and homeostasis maintenance within cells. False alterations of lysosomal functions can lead to broad detrimental effects and cause various diseases, including cancers. Cancer cells that are rapidly proliferative and invasive are highly dependent on effective lysosomal function. Malignant melanoma is the most lethal form of skin cancer, with high metastasis characteristics, drug resistance, and aggressiveness. It is critical to understand the role of lysosomes in melanoma pathogenesis in order to improve the outcomes of melanoma patients. In this mini-review, we compile our current knowledge of lysosomes' role in tumorigenesis, progression, therapy resistance, and the current treatment strategies related to lysosomes in melanoma.
Collapse
Affiliation(s)
- Chia-Hsin Hsu
- Department of Veterinary Medicine, School of Veterinary Medicine, National Taiwan University, Taipei 10617, Taiwan;
- Department of Biomedical Sciences, Cornell University, Ithaca, NY 14853, USA
| | - Keng-Jung Lee
- Department of Biomedical Engineering, Carnegie Mellon University, Pittsburgh, PA 15213, USA;
| | - Yi-Han Chiu
- Department of Microbiology, Soochow University, Taipei 10617, Taiwan;
| | - Kuo-Ching Huang
- Holistic Education Center, Mackay Medical College, New Taipei City 25245, Taiwan;
| | - Guo-Shou Wang
- Department of Biological Science and Technology, National Yang Ming Chiao Tung University, Hsinchu 30068, Taiwan; (G.-S.W.); (K.-W.L.)
| | - Lei-Po Chen
- Ph.D. Degree Program of Biomedical Science and Engineering, National Yang Ming Chiao Tung University, Hsinchu 30068, Taiwan;
| | - Kuang-Wen Liao
- Department of Biological Science and Technology, National Yang Ming Chiao Tung University, Hsinchu 30068, Taiwan; (G.-S.W.); (K.-W.L.)
| | - Chen-Si Lin
- Department of Veterinary Medicine, School of Veterinary Medicine, National Taiwan University, Taipei 10617, Taiwan;
| |
Collapse
|
41
|
Peng L, Qiu J, Liu L, Li X, Liu X, Zhang Y. Preparation of PEG/ZIF-8@HF drug delivery system for melanoma treatment via oral administration. Drug Deliv 2022; 29:1075-1085. [PMID: 35373691 PMCID: PMC8986218 DOI: 10.1080/10717544.2022.2058649] [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] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
Melanoma is one of the highly malignant tumors whose incidence and fatality rates have been increased year by year. However, in addition to early surgical resection, there still lacks specific targeted drugs and treatment strategies. In this study, it was discovered that hinokiflavone (HF) encapsulated in zeolitic imidazolate framework-8 (ZIF-8) exhibited a superior anti-melanoma effect in vitro and in vivo. HF was encapsulated in ZIF-8 through a one-step synthesis method, and polyethylene glycol (PEG-2000) was used to optimize the size and dispersion of the drug-loaded complex (PEG/ZIF-8@HF). The results show that the prepared PEG/ZIF-8@HF has a high encapsulation efficiency (92.12%) and can achieve selective drug release in an acidic microenvironment. The results of in vitro anti-melanoma experiments indicate that PEG/ZIF-8@HF shows up-regulation of reactive oxygen species (ROS) levels and can restrain the migration and invasion of B16F10 cells. Moreover, in vivo animal experiments further confirm that PEG/ZIF-8@HF shows anti-tumor effect by up-regulating the pro-apoptotic proteins caspase-3 and caspase-8, and down-regulating the migration-promoting invasion protein MMP-9. This study developed a safe and effective oral administration of HF based on the high-efficiency delivery ZIF-8 system, which provides an effective treatment strategy for melanoma.
Collapse
Affiliation(s)
- Luxi Peng
- The Third Affiliated Hospital of School of Medicine, Shihezi University, Shihezi, China.,The State Key Lab of High Performance Ceramics and Superfine Microstructure, Shanghai Institute of Ceramics, Chinese Academy of Sciences, Shanghai, China
| | - Jiajun Qiu
- The State Key Lab of High Performance Ceramics and Superfine Microstructure, Shanghai Institute of Ceramics, Chinese Academy of Sciences, Shanghai, China
| | - Lidan Liu
- The State Key Lab of High Performance Ceramics and Superfine Microstructure, Shanghai Institute of Ceramics, Chinese Academy of Sciences, Shanghai, China
| | - Xiaoyu Li
- Department of Pharmacy, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Xuanyong Liu
- The State Key Lab of High Performance Ceramics and Superfine Microstructure, Shanghai Institute of Ceramics, Chinese Academy of Sciences, Shanghai, China
| | - Yongjun Zhang
- The Third Affiliated Hospital of School of Medicine, Shihezi University, Shihezi, China
| |
Collapse
|
42
|
Kimsa-Dudek M, Synowiec-Wojtarowicz A, Krawczyk A, Kosowska A, Kimsa-Furdzik M, Francuz T. The Apoptotic Effect of Caffeic or Chlorogenic Acid on the C32 Cells That Have Simultaneously Been Exposed to a Static Magnetic Field. Int J Mol Sci 2022; 23:ijms23073859. [PMID: 35409218 PMCID: PMC8999068 DOI: 10.3390/ijms23073859] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2022] [Revised: 03/28/2022] [Accepted: 03/29/2022] [Indexed: 12/04/2022] Open
Abstract
The induction of apoptosis is one of the main goals of the designed anti-cancer therapies. In recent years, increased attention has been paid to the physical factors such as magnetic fields and to the natural bioactive compounds and the possibilities using them in medicine. Hence, the aim of this study was to evaluate the anti-tumor effect of caffeic or chlorogenic acid in combination with a moderate-strength static magnetic field on C32 melanoma cells by assessing the effect of both factors on the apoptotic process. The apoptosis of the C32 cells was evaluated using a flow cytometry analysis. The expression of the apoptosis-associated genes was determined using the RT-qPCR technique. The caspase activity and the concentration of the oxidative damage markers were also measured. It was found that phenolic acids and a static magnetic field trigger the apoptosis of the C32 cells and also affect the expression of the genes encoding the apoptosis regulatory proteins. In conclusion, our study indicated that both of the phenolic acids and a static magnetic field can be used supportively in the treatment of melanoma and that caffeic acid is more pro-apoptotic than chlorogenic acid.
Collapse
Affiliation(s)
- Magdalena Kimsa-Dudek
- Department of Nutrigenomics and Bromatology, Faculty of Pharmaceutical Sciences in Sosnowiec, Medical University of Silesia, Katowice, Jednosci 8, 41-200 Sosnowiec, Poland; (A.S.-W.); (A.K.)
- Correspondence: ; Tel.: +48-32-364-11-72
| | - Agnieszka Synowiec-Wojtarowicz
- Department of Nutrigenomics and Bromatology, Faculty of Pharmaceutical Sciences in Sosnowiec, Medical University of Silesia, Katowice, Jednosci 8, 41-200 Sosnowiec, Poland; (A.S.-W.); (A.K.)
| | - Agata Krawczyk
- Department of Nutrigenomics and Bromatology, Faculty of Pharmaceutical Sciences in Sosnowiec, Medical University of Silesia, Katowice, Jednosci 8, 41-200 Sosnowiec, Poland; (A.S.-W.); (A.K.)
| | - Agnieszka Kosowska
- Department of Biochemistry, Faculty of Medical Sciences in Katowice, Medical University of Silesia, Medyków 18, 40-752 Katowice, Poland; (A.K.); (M.K.-F.); (T.F.)
| | - Małgorzata Kimsa-Furdzik
- Department of Biochemistry, Faculty of Medical Sciences in Katowice, Medical University of Silesia, Medyków 18, 40-752 Katowice, Poland; (A.K.); (M.K.-F.); (T.F.)
| | - Tomasz Francuz
- Department of Biochemistry, Faculty of Medical Sciences in Katowice, Medical University of Silesia, Medyków 18, 40-752 Katowice, Poland; (A.K.); (M.K.-F.); (T.F.)
| |
Collapse
|
43
|
Patel M, Prabhu A. Smart nanocomposite assemblies for multimodal cancer theranostics. Int J Pharm 2022; 618:121697. [PMID: 35337903 DOI: 10.1016/j.ijpharm.2022.121697] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2022] [Revised: 03/18/2022] [Accepted: 03/19/2022] [Indexed: 12/28/2022]
Abstract
Despite great strides in anticancer research, performance statistics of current treatment modalities remain dismal, highlighting the need for safe, efficacious strategies for tumour mitigation. Non-invasive fusion technology platforms combining photodynamic, photothermal and hyperthermia therapies have emerged as alternate strategies with potential to meet many of the unmet clinical demands in the domain of cancer. These therapies make use of metallic and magnetic nanoparticles with light absorbing properties, which are manipulated to generate either reactive cytotoxic oxygen species or heat for tumour ablation. Combination therapies integrating light, heat and magnetism-mediated nanoplatforms with the conventional approaches of chemotherapy, radiotherapy and surgery are emerging as precision medicine for targeted interventions against cancer. This article aims to compile recent developments of advanced nanocomposite assemblies that integrate multimodal therapeutics for cancer treatment. Amalgamation of various effective, non-invasive technological platforms such as photodynamic therapy (PDT), photothermal therapy (PTT), magnetic hyperthermia (MHT), and chemodynamic therapy (CDT) have tremendous potential in presenting safe and efficacious solutions to the formidable challenges in cancer therapeutics.
Collapse
Affiliation(s)
- Manshi Patel
- Department of Pharmaceutical Chemistry & Quality Assurance, SVKM's Dr. Bhanuben Nanavati College of Pharmacy, Mumbai, India
| | - Arati Prabhu
- Department of Pharmaceutical Chemistry & Quality Assurance, SVKM's Dr. Bhanuben Nanavati College of Pharmacy, Mumbai, India.
| |
Collapse
|
44
|
Carpenter EL, Becker AL, Indra AK. NRF2 and Key Transcriptional Targets in Melanoma Redox Manipulation. Cancers (Basel) 2022; 14:cancers14061531. [PMID: 35326683 PMCID: PMC8946769 DOI: 10.3390/cancers14061531] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2022] [Revised: 03/13/2022] [Accepted: 03/14/2022] [Indexed: 02/04/2023] Open
Abstract
Melanocytes are dendritic, pigment-producing cells located in the skin and are responsible for its protection against the deleterious effects of solar ultraviolet radiation (UVR), which include DNA damage and elevated reactive oxygen species (ROS). They do so by synthesizing photoprotective melanin pigments and distributing them to adjacent skin cells (e.g., keratinocytes). However, melanocytes encounter a large burden of oxidative stress during this process, due to both exogenous and endogenous sources. Therefore, melanocytes employ numerous antioxidant defenses to protect themselves; these are largely regulated by the master stress response transcription factor, nuclear factor erythroid 2-related factor 2 (NRF2). Key effector transcriptional targets of NRF2 include the components of the glutathione and thioredoxin antioxidant systems. Despite these defenses, melanocyte DNA often is subject to mutations that result in the dysregulation of the proliferative mitogen-activated protein kinase (MAPK) pathway and the cell cycle. Following tumor initiation, endogenous antioxidant systems are co-opted, a consequence of elevated oxidative stress caused by metabolic reprogramming, to establish an altered redox homeostasis. This altered redox homeostasis contributes to tumor progression and metastasis, while also complicating the application of exogenous antioxidant treatments. Further understanding of melanocyte redox homeostasis, in the presence or absence of disease, would contribute to the development of novel therapies to aid in the prevention and treatment of melanomas and other skin diseases.
Collapse
Affiliation(s)
- Evan L. Carpenter
- Department of Pharmaceutical Sciences, College of Pharmacy, Oregon State University, Corvallis, OR 97331, USA; (E.L.C.); (A.L.B.)
| | - Alyssa L. Becker
- Department of Pharmaceutical Sciences, College of Pharmacy, Oregon State University, Corvallis, OR 97331, USA; (E.L.C.); (A.L.B.)
- John A. Burns School of Medicine, University of Hawaii, Honolulu, HI 96813, USA
| | - Arup K. Indra
- Department of Pharmaceutical Sciences, College of Pharmacy, Oregon State University, Corvallis, OR 97331, USA; (E.L.C.); (A.L.B.)
- Knight Cancer Institute, Oregon Health & Science University, Portland, OR 97239, USA
- Department of Biochemistry and Biophysics, Oregon State University, Corvallis, OR 97331, USA
- Linus Pauling Science Center, Oregon State University, Corvallis, OR 97331, USA
- Department of Dermatology, Oregon Health & Science University, Portland, OR 97239, USA
- Correspondence:
| |
Collapse
|
45
|
McKamey SG, Jira LR, Tweed CM, Blake SD, Powell DP, Daghistani AT, Koh DW. Antagonism of the transient receptor potential melastatin‑2 channel leads to targeted antitumor effects in primary human malignant melanoma cells. Int J Oncol 2022; 60:43. [PMID: 35234266 DOI: 10.3892/ijo.2022.5333] [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: 09/07/2021] [Accepted: 02/01/2022] [Indexed: 11/06/2022] Open
Abstract
Melanoma continues to be the most aggressive and devastating form of skin cancer for which the development of novel therapies is required. The present study aimed to determine the effects of antagonism of the transient receptor potential melastatin‑2 (TRPM2) ion channel in primary human malignant melanoma cells. TRPM2 antagonism via use of the antifungal agent, clotrimazole, led to decreases in cell proliferation, as well as dose‑dependent increases in cell death in all melanoma cell lines investigated. The targeting of TRPM2 channels was verified using TRPM2 knockdown, where treatment with TRPM2 small‑interfering RNA led to similar levels of cell death in all melanoma cell lines when compared with clotrimazole treatment. Minimal effects on proliferation and cell death were observed following antagonism or knockdown of TRPM2 in non‑cancerous human keratinocytes. Moreover, characteristics of TRPM2 were explored in these melanoma cells and the results demonstrated that TRPM2, localized to the plasma membrane as a non‑specific ion channel in non‑cancerous cells, displayed a nuclear localization in all human melanoma cell lines analyzed. Additional characterization of these melanoma cell lines confirmed that each expressed one or more established multidrug resistance genes. Results of the present study therefore indicated that antagonism of the TRPM2 channel led to antitumor effects in human melanoma cells, including those that are potentially unresponsive to current treatments due to the expression of drug resistance genes. The unique cellular localization of TRPM2 and the specificity of the antitumor effects elicited by TRPM2 antagonism suggested that TRPM2 possesses a unique role in melanoma cells. Collectively, the targeting of TRPM2 represents a potentially novel, efficacious and readily accessible treatment option for patients with melanoma.
Collapse
Affiliation(s)
- Shelby G McKamey
- Department of Pharmaceutical and Biomedical Sciences, Ohio Northern University, Ada, OH 45810, USA
| | - Lukas R Jira
- Department of Pharmaceutical and Biomedical Sciences, Ohio Northern University, Ada, OH 45810, USA
| | - Christopher M Tweed
- Department of Pharmaceutical and Biomedical Sciences, Ohio Northern University, Ada, OH 45810, USA
| | - Steven D Blake
- Department of Pharmaceutical and Biomedical Sciences, Ohio Northern University, Ada, OH 45810, USA
| | - Daniel P Powell
- Department of Pharmaceutical and Biomedical Sciences, Ohio Northern University, Ada, OH 45810, USA
| | - Ayah T Daghistani
- Department of Pharmaceutical and Biomedical Sciences, Ohio Northern University, Ada, OH 45810, USA
| | - David W Koh
- Department of Pharmaceutical and Biomedical Sciences, Ohio Northern University, Ada, OH 45810, USA
| |
Collapse
|
46
|
Haussmann PB, Pavani C, Marcolongo-Pereira C, Bellettini-Santos T, da Silva BS, Benedito IF, Freitas ML, Baptista MS, Chiarelli-Neto O. Melanin photosensitization by green light reduces melanoma tumor size. JOURNAL OF PHOTOCHEMISTRY AND PHOTOBIOLOGY 2022. [DOI: 10.1016/j.jpap.2021.100092] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022] Open
|
47
|
Yong JM, Fu L, Tang F, Yu P, Kuchel RP, Whitelock JM, Lord MS. ROS-Mediated Anti-Angiogenic Activity of Cerium Oxide Nanoparticles in Melanoma Cells. ACS Biomater Sci Eng 2022; 8:512-525. [PMID: 34989230 DOI: 10.1021/acsbiomaterials.1c01268] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Angiogenesis plays a key role in cancer progression, including transition to the metastatic phase via reactive oxygen species (ROS)-dependent pathways, among others. Antivascular endothelial growth factor (VEGF) antibodies have been trialed as an anti-angiogenic therapy for cancer but are associated with high cost, limited efficacy, and side effects. Cerium oxide nanoparticles (nanoceria) are promising nanomaterials for biomedical applications due to their ability to modulate intracellular ROS. Nanoceria can be produced by a range of synthesis methods, with chemical precipitation as the most widely explored. It has been reported that chemical precipitation can fine-tune primary particle size where a limited number of synthesis parameters were varied. Here, we explore the effect of temperature, precipitating agent concentration and rate of addition, stirring rate, and surfactant concentration on nanoceria primary particle size using a fractional factorial experimental design approach. We establish a robust synthesis method for faceted nanoceria with primary particle diameters of 5-6 nm. The nanoceria are not cytotoxic to a human melanoma cell line (Mel1007) at doses up to 400 μg/mL and are dose-dependently internalized by the cells. The intracellular ROS level for some cells that internalized the nanoceria is reduced, which correlates with a dose-dependent reduction in angiogenic gene expression including VEGF. These findings contribute to our knowledge of the anti-angiogenic effects of nanoceria and help to develop our understanding of potentially new anti-angiogenic agents for combination cancer therapies.
Collapse
Affiliation(s)
- Joel M Yong
- Graduate School of Biomedical Engineering, Level 5, Samuels Building, UNSW Sydney, Sydney, NSW 2052, Australia
| | - Lu Fu
- Graduate School of Biomedical Engineering, Level 5, Samuels Building, UNSW Sydney, Sydney, NSW 2052, Australia
| | - Fengying Tang
- Graduate School of Biomedical Engineering, Level 5, Samuels Building, UNSW Sydney, Sydney, NSW 2052, Australia.,Department of Comparative Medicine, University of Washington, Seattle, Washington 98195, United States
| | - Peimin Yu
- Graduate School of Biomedical Engineering, Level 5, Samuels Building, UNSW Sydney, Sydney, NSW 2052, Australia
| | - Rhiannon P Kuchel
- Electron Microscope Unit, Basement, Chemical Sciences Building, UNSW Sydney, Sydney, NSW 2052, Australia
| | - John M Whitelock
- Graduate School of Biomedical Engineering, Level 5, Samuels Building, UNSW Sydney, Sydney, NSW 2052, Australia
| | - Megan S Lord
- Graduate School of Biomedical Engineering, Level 5, Samuels Building, UNSW Sydney, Sydney, NSW 2052, Australia
| |
Collapse
|
48
|
Khan H, Alam W, Alsharif KF, Aschner M, Pervez S, Saso L. Alkaloids and Colon Cancer: Molecular Mechanisms and Therapeutic Implications for Cell Cycle Arrest. Molecules 2022; 27:molecules27030920. [PMID: 35164185 PMCID: PMC8838632 DOI: 10.3390/molecules27030920] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2021] [Revised: 01/21/2022] [Accepted: 01/25/2022] [Indexed: 12/18/2022] Open
Abstract
Cancer is the second most fatal disease worldwide, with colon cancer being the third most prevalent and fatal form of cancer in several Western countries. The risk of acquisition of resistance to chemotherapy remains a significant hurdle in the management of various types of cancer, especially colon cancer. Therefore, it is essential to develop alternative treatment modalities. Naturally occurring alkaloids have been shown to regulate various mechanistic pathways linked to cell proliferation, cell cycle, and metastasis. This review aims to shed light on the potential of alkaloids as anti-colon-cancer chemotherapy agents that can modulate or arrest the cell cycle. Preclinical investigated alkaloids have shown anti-colon cancer activities and inhibition of cancer cell proliferation via cell cycle arrest at different stages, suggesting that alkaloids may have the potential to act as anticancer molecules.
Collapse
Affiliation(s)
- Haroon Khan
- Department of Pharmacy, Abdul Wali Khan University Mardan, Mardan 23200, Pakistan;
- Correspondence: or
| | - Waqas Alam
- Department of Pharmacy, Abdul Wali Khan University Mardan, Mardan 23200, Pakistan;
| | - Khalaf F. Alsharif
- Department of Clinical Laboratory, College of Applied Medical Science, Taif University, P.O. Box 11099,Taif 21944, Saudi Arabia;
| | - Michael Aschner
- Department of Molecular Pharmacology, Albert Einstein College of Medicine, Bronx, NY 10461, USA;
| | - Samreen Pervez
- Department of Pharmacy, Qurtuba University of Science and Information Technology, Peshawar 29050, Pakistan;
| | - Luciano Saso
- Department of Physiology and Pharmacology “Vittorio Erspamer”, Sapienza University of Rome, 00185 Rome, Italy;
| |
Collapse
|
49
|
Santourlidis S, Schulz WA, Araúzo-Bravo MJ, Gerovska D, Ott P, Bendhack ML, Hassan M, Erichsen L. Epigenetics in the Diagnosis and Therapy of Malignant Melanoma. Int J Mol Sci 2022; 23:ijms23031531. [PMID: 35163453 PMCID: PMC8835790 DOI: 10.3390/ijms23031531] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2022] [Revised: 01/24/2022] [Accepted: 01/26/2022] [Indexed: 12/15/2022] Open
Abstract
Epigenetic mechanisms are fundamentally important for cancer initiation and development. However, a survey of the literature reveals that, to date, they appear less comprehensively investigated in melanoma than in many other cancers, e.g., prostate, breast, and colon carcinoma. The aim of this review is to provide a short summary of epigenetic aspects of functional relevance for melanoma pathogenesis. In addition, some new perspectives from epigenetic research in other cancers with potential for melanoma diagnosis and therapy are introduced. For example, the PrimeEpiHit hypothesis in urothelial carcinoma, which, similarly to malignant melanoma, can also be triggered by a single exogenous noxa, states that one of the first steps for cancer initiation could be epigenetic changes in key genes of one-carbon metabolism. The application of such insights may contribute to further progress in the diagnosis and therapy of melanoma, a deadly type of cancer.
Collapse
Affiliation(s)
- Simeon Santourlidis
- Epigenetics Core Laboratory, Institute of Transplantation Diagnostics and Cell Therapeutics, Medical Faculty, Heinrich-Heine University Duesseldorf, 40225 Duesseldorf, Germany; (S.S.); (P.O.)
| | - Wolfgang A. Schulz
- Department of Urology, Medical Faculty, Heinrich-Heine University Duesseldorf, 40225 Duesseldorf, Germany;
| | - Marcos J. Araúzo-Bravo
- Group of Computational Biology and Systems Biomedicine, Biodonostia Health Research Institute, 20014 San Sebastián, Spain; (M.J.A.-B.); (D.G.)
- IKERBASQUE, Basque Foundation for Science, 48009 Bilbao, Spain
| | - Daniela Gerovska
- Group of Computational Biology and Systems Biomedicine, Biodonostia Health Research Institute, 20014 San Sebastián, Spain; (M.J.A.-B.); (D.G.)
| | - Pauline Ott
- Epigenetics Core Laboratory, Institute of Transplantation Diagnostics and Cell Therapeutics, Medical Faculty, Heinrich-Heine University Duesseldorf, 40225 Duesseldorf, Germany; (S.S.); (P.O.)
| | - Marcelo L. Bendhack
- Department of Urology, University Hospital, Positivo University, Curitiba 80030-200, Brazil;
| | - Mohamed Hassan
- Department of Surgery, Tulane University School of Medicine, New Orleans, LA 70112, USA;
- Institut National de la Santé et de la Recherché Médicale, University of Strasbourg, 67000 Strasbourg, France
| | - Lars Erichsen
- Institute for Stem Cell Research and Regenerative Medicine, Medical Faculty, Heinrich-Heine University Düsseldorf, 40225 Duesseldorf, Germany
- Correspondence: ; Tel.: +49-0211-81-16905
| |
Collapse
|
50
|
Wang L, Lu D, Huo M, Xu H. Oligomycin A Induces Apoptosis‐to‐Pyroptosis Switch against Melanoma with Sensitized Immunotherapy. ADVANCED FUNCTIONAL MATERIALS 2022; 32. [DOI: 10.1002/adfm.202106332] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/01/2021] [Indexed: 01/11/2025]
Abstract
AbstractInduction of mitochondrial‐targeted therapies is demonstrated to be predominantly effective in malignant melanoma therapeutics mediated by programmed cell death (PCD). The pivotal control to switch cell apoptosis to pyroptosis is exhibited to overcome the apoptosis‐resisting characters and induce immune responses against the melanoma. Yet approaches to trigger this switch are limited by far. In the present work, it is found that oligomycin A (OA), a mitochondrial respiratory inhibitor, can effectively suppress mitochondrial functionalities and induce the intracellular oxidative stresses in situ, ultimately inducing apoptosis‐to‐pyroptosis switch against melanoma. The authors have encapsulated OA into the designed cyclic argine‐glycine‐aspartic (c(RGDyC)) decorated iron constructed metal‐organic framework (MIL101‐NH2‐Fe) to develop tumorous glutathione responsive nanocatalytic therapeutics, aiming to trigger intensive apoptosis‐to‐pyroptosis PCD against melanoma, accompanied with the sensitized immunotherapy via the immune checkpoint blockade. The present work offers a prominent PCD switch to destruct melanoma with promising clinical perspectives.
Collapse
Affiliation(s)
- Liying Wang
- Department of Medical Ultrasound Shanghai Tenth People's Hospital Ultrasound Research and Education Institute Tongji University School of Medicine Shanghai Engineering Research Center of Ultrasound Diagnosis and Treatment National Clinical Research Center of Interventional Medicine Shanghai 200072 P. R. China
| | - Dan Lu
- Department of Medical Ultrasound Shanghai Tenth People's Hospital Ultrasound Research and Education Institute Tongji University School of Medicine Shanghai Engineering Research Center of Ultrasound Diagnosis and Treatment National Clinical Research Center of Interventional Medicine Shanghai 200072 P. R. China
| | - Minfeng Huo
- Shanghai Tenth People's Hospital Tongji University School of Medicine Shanghai 200436 P. R. China
- State Key Laboratory of High Performance Ceramics and Superfine Microstructure Shanghai Institute of Ceramics Chinese Academy of Sciences Shanghai 200050 P. R. China
| | - Huixiong Xu
- Department of Medical Ultrasound Shanghai Tenth People's Hospital Ultrasound Research and Education Institute Tongji University School of Medicine Shanghai Engineering Research Center of Ultrasound Diagnosis and Treatment National Clinical Research Center of Interventional Medicine Shanghai 200072 P. R. China
| |
Collapse
|