1
|
Dai L, Chen L, Wang W, Lin P. Resveratrol inhibits ACHN cells via regulation of histone acetylation. PHARMACEUTICAL BIOLOGY 2020; 58:231-238. [PMID: 32202448 PMCID: PMC7144206 DOI: 10.1080/13880209.2020.1738503] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/27/2019] [Revised: 01/20/2020] [Accepted: 03/01/2020] [Indexed: 06/10/2023]
Abstract
Context: The relationship between resveratrol and histone acetylation in renal cell carcinoma (RCC) has not yet been reported.Objective: To explore the functional role of resveratrol in RCC.Materials and methods: Functional experiments were performed to determine proliferatio n of ACHN cells with treatment of resveratrol (0, 7.8125, 15.625, 31.25 and 62.5 μg/mL, for 12, 24 and 48 h of culture) or 0.1 μM SAHA. The enzyme activities of MMP-2/-9 were measured by gelatine zymography and histone acetylation by Western blot.Results: When the cells were treated with 15.625, 31.25 and 62.5 μg/mL resveratrol, ACHN cells viability was 73.2 ± 3.5%, 61.4 ± 3.1%, 50.2 ± 4.7% for 12 h, 62.7 ± 4.5%, 52.4 ± 5.5%, 40.2 ± 3.8% for 24 h, and 60.8 ± 3.7%, 39.4 ± 5.1%, 37.6 ± 2.7% for 48 h, and the wound closure (%) of migration was increased from 0.6 to 0.7, 0.85, 0.9 for 12 h and from 0.23 to 0.3, 0.48, 0.59 for 24 h. The invasion rate was 8.5 ± 0.9%, 7.4 ± 0.3% and 5.8 ± 0.6%, and cell cycle was arrested at G1 from 42.5 ± 2.9% to 55.3 ± 5.7%, 59.8 ± 3.4%, 68.7 ± 4.6%. MMP-2/-9 expression (p < 0.05) was inhibited by resveratrol. The protein levels of histone acetylation (p < 0.01) was increased by resveratrol.Discussion and conclusions: Our results suggest that these effects might be related to a high level of histone acetylation, and resveratrol can be considered as an alternative treatment for RCC.
Collapse
Affiliation(s)
- Lili Dai
- Department of Science and Education, Jiujiang University Clinical Medical College, Jiujiang, China
| | - Lingyan Chen
- Department of Rehabilitation, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Wenjing Wang
- TCM Basic Clinical Research Office, Guiyang University of Chinese Medicine, Guiyang, China
| | - Peizheng Lin
- Department of Encephalopathy, The First Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, China
| |
Collapse
|
2
|
From Seabed to Bedside: A Review on Promising Marine Anticancer Compounds. Biomolecules 2020; 10:biom10020248. [PMID: 32041255 PMCID: PMC7072248 DOI: 10.3390/biom10020248] [Citation(s) in RCA: 28] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2019] [Revised: 01/29/2020] [Accepted: 02/04/2020] [Indexed: 02/08/2023] Open
Abstract
The marine environment represents an outstanding source of antitumoral compounds and, at the same time, remains highly unexplored. Organisms living in the sea synthesize a wide variety of chemicals used as defense mechanisms. Interestingly, a large number of these compounds exert excellent antitumoral properties and have been developed as promising anticancer drugs that have later been approved or are currently under validation in clinical trials. However, due to the high need for these compounds, new methodologies ensuring its sustainable supply are required. Also, optimization of marine bioactives is an important step for their success in the clinical setting. Such optimization involves chemical modifications to improve their half-life in circulation, potency and tumor selectivity. In this review, we outline the most promising marine bioactives that have been investigated in cancer models and/or tested in patients as anticancer agents. Moreover, we describe the current state of development of anticancer marine compounds and discuss their therapeutic limitations as well as different strategies used to overcome these limitations. The search for new marine antitumoral agents together with novel identification and chemical engineering approaches open the door for novel, more specific and efficient therapeutic agents for cancer treatment.
Collapse
|
3
|
Zhou R, Wu J, Tang X, Wei X, Ju C, Zhang F, Sun J, Shuai D, Zhang Z, Liu Q, Lv XB. Histone deacetylase inhibitor AR-42 inhibits breast cancer cell growth and demonstrates a synergistic effect in combination with 5-FU. Oncol Lett 2018; 16:1967-1974. [PMID: 30008890 DOI: 10.3892/ol.2018.8854] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2017] [Accepted: 04/06/2018] [Indexed: 12/30/2022] Open
Abstract
AR-42 is a member of a novelly discovered class of phenylbutyrate-derived histone deacetylase inhibitors, and has a number of antitumor effects in a variety of tumor types; however, the role of AR-42 and its possible mechanisms have not been reported in the treatment of breast cancer. The aim of the present study was to investigate the antitumor effects of AR-42 and its associated mechanisms in breast cancer. MTT assays and colony formation assays were conducted to measure the proliferation of MCF-7 cells, and flow cytometry was used to analyze cell apoptosis. The results revealed that AR-42 induced cell apoptosis and suppressed cell growth in a dose- and time-dependent manner. Mechanistically, AR-42 treatment increased the acetylation of the p53 protein and prolonged the half-life of the p53 protein; furthermore, AR-42 treatment upregulated p21 and PUMA expression. Notably, AR-42 had a synergistic effect on MCF-7 cells in combination with fluorouracil, which is one of the most commonly used chemotherapeutic agents. In conclusion, the results indicated that AR-42 inhibits breast cancer cell proliferation and induces apoptosis, indicating that AR-42 is a potential therapeutic agent.
Collapse
Affiliation(s)
- Ruihao Zhou
- Nanchang Key Laboratory of Cancer Pathogenesis and Translational Research, The Third Affiliated Hospital, Nanchang University, Nanchang, Jiangxi 330008, P.R. China.,First Clinical Department, Medical School of Nanchang University, Nanchang, Jiangxi 330006, P.R. China
| | - Juan Wu
- Guangzhou Key Laboratory of Translational Medicine on Malignant Tumor Treatment, Affiliated Tumor Hospital of Guangzhou Medical University, Guangzhou, Guangdong 510095, P.R. China
| | - Xiaofeng Tang
- Nanchang Key Laboratory of Cancer Pathogenesis and Translational Research, The Third Affiliated Hospital, Nanchang University, Nanchang, Jiangxi 330008, P.R. China
| | - Xin Wei
- Nanchang Key Laboratory of Cancer Pathogenesis and Translational Research, The Third Affiliated Hospital, Nanchang University, Nanchang, Jiangxi 330008, P.R. China
| | - Cheng Ju
- Nanchang Key Laboratory of Cancer Pathogenesis and Translational Research, The Third Affiliated Hospital, Nanchang University, Nanchang, Jiangxi 330008, P.R. China
| | - Feifei Zhang
- Nanchang Key Laboratory of Cancer Pathogenesis and Translational Research, The Third Affiliated Hospital, Nanchang University, Nanchang, Jiangxi 330008, P.R. China
| | - Jun Sun
- Nanchang Key Laboratory of Cancer Pathogenesis and Translational Research, The Third Affiliated Hospital, Nanchang University, Nanchang, Jiangxi 330008, P.R. China
| | - Deyong Shuai
- Nanchang Key Laboratory of Cancer Pathogenesis and Translational Research, The Third Affiliated Hospital, Nanchang University, Nanchang, Jiangxi 330008, P.R. China
| | - Zhiping Zhang
- Nanchang Key Laboratory of Cancer Pathogenesis and Translational Research, The Third Affiliated Hospital, Nanchang University, Nanchang, Jiangxi 330008, P.R. China
| | - Qiong Liu
- Department of Cardiovascular Medicine, Xiangya Hospital of Central South University, Changsha, Hunan 410008, P.R. China
| | - Xiao-Bin Lv
- Nanchang Key Laboratory of Cancer Pathogenesis and Translational Research, The Third Affiliated Hospital, Nanchang University, Nanchang, Jiangxi 330008, P.R. China
| |
Collapse
|
4
|
Zalfa F, Panasiti V, Carotti S, Zingariello M, Perrone G, Sancillo L, Pacini L, Luciani F, Roberti V, D'Amico S, Coppola R, Abate SO, Rana RA, De Luca A, Fiers M, Melocchi V, Bianchi F, Farace MG, Achsel T, Marine JC, Morini S, Bagni C. The fragile X mental retardation protein regulates tumor invasiveness-related pathways in melanoma cells. Cell Death Dis 2017; 8:e3169. [PMID: 29144507 PMCID: PMC5775405 DOI: 10.1038/cddis.2017.521] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2017] [Revised: 08/24/2017] [Accepted: 08/25/2017] [Indexed: 02/06/2023]
Abstract
The fragile X mental retardation protein (FMRP) is lacking or mutated in patients with the fragile X syndrome (FXS), the most frequent form of inherited intellectual disability. FMRP affects metastasis formation in a mouse model for breast cancer. Here we show that FMRP is overexpressed in human melanoma with high Breslow thickness and high Clark level. Furthermore, meta-analysis of the TCGA melanoma data revealed that high levels of FMRP expression correlate significantly with metastatic tumor tissues, risk of relapsing and disease-free survival. Reduction of FMRP in metastatic melanoma cell lines impinges on cell migration, invasion and adhesion. Next-generation sequencing in human melanoma cells revealed that FMRP regulates a large number of mRNAs involved in relevant processes of melanoma progression. Our findings suggest an association between FMRP levels and the invasive phenotype in melanoma and might open new avenues towards the discovery of novel therapeutic targets.
Collapse
Affiliation(s)
- Francesca Zalfa
- Department of Medicine, Campus Bio-Medico University, via Alvaro del Portillo 21, 00128 Rome, Italy
| | - Vincenzo Panasiti
- Department of Medicine, Campus Bio-Medico University, via Alvaro del Portillo 21, 00128 Rome, Italy
| | - Simone Carotti
- Department of Medicine, Campus Bio-Medico University, via Alvaro del Portillo 21, 00128 Rome, Italy
| | - Maria Zingariello
- Department of Medicine, Campus Bio-Medico University, via Alvaro del Portillo 21, 00128 Rome, Italy
| | - Giuseppe Perrone
- Department of Medicine, Campus Bio-Medico University, via Alvaro del Portillo 21, 00128 Rome, Italy
| | - Laura Sancillo
- Department of Medicine and Science of Aging, University of Chieti 'G d'Annunzio', via dei Vestini 31, 66100 Chieti-Pescara, Italy
| | - Laura Pacini
- Department of Biomedicine and Prevention, University of Rome 'Tor Vergata', via Montpellier 1, 00133 Rome, Italy
| | - Flavie Luciani
- VIB/Center for the Biology of Disease, KU Leuven, O&N 4, Herestraat 49 Box 602, 3000, Leuven, Belgium.,Center for Human Genetics, Leuven Institute for Neuroscience and Disease, KU Leuven, O&N 4, Herestraat 49 Box 602, Leuven, 3000, Belgium
| | - Vincenzo Roberti
- Department of Dermatology, University of Rome 'La Sapienza', viale dell'Università 1, 00185 Rome, Italy
| | - Silvia D'Amico
- Department of Biomedicine and Prevention, University of Rome 'Tor Vergata', via Montpellier 1, 00133 Rome, Italy
| | - Rosa Coppola
- Department of Medicine, Campus Bio-Medico University, via Alvaro del Portillo 21, 00128 Rome, Italy
| | - Simona Osella Abate
- Department of Medical Science and Human Oncology, Section of Dermato-Oncology, University of Turin, via Verdi 8, 10124 Turin, Italy
| | - Rosa Alba Rana
- Department of Medicine and Science of Aging, University of Chieti 'G d'Annunzio', via dei Vestini 31, 66100 Chieti-Pescara, Italy
| | - Anastasia De Luca
- Department of Biomedicine and Prevention, University of Rome 'Tor Vergata', via Montpellier 1, 00133 Rome, Italy
| | - Mark Fiers
- VIB/Center for the Biology of Disease, KU Leuven, O&N 4, Herestraat 49 Box 602, 3000, Leuven, Belgium.,Center for Human Genetics, Leuven Institute for Neuroscience and Disease, KU Leuven, O&N 4, Herestraat 49 Box 602, Leuven, 3000, Belgium
| | - Valentina Melocchi
- ISBREMIT, Institute for Stem-cell Biology, Regenerative Medicine and Innovative Therapies, IRCCS Casa Sollievo della Sofferenza, viale Padre Pio 7, 71013 San Giovanni Rotondo (FG), Italy
| | - Fabrizio Bianchi
- ISBREMIT, Institute for Stem-cell Biology, Regenerative Medicine and Innovative Therapies, IRCCS Casa Sollievo della Sofferenza, viale Padre Pio 7, 71013 San Giovanni Rotondo (FG), Italy
| | - Maria Giulia Farace
- Department of Biomedicine and Prevention, University of Rome 'Tor Vergata', via Montpellier 1, 00133 Rome, Italy
| | - Tilmann Achsel
- VIB/Center for the Biology of Disease, KU Leuven, O&N 4, Herestraat 49 Box 602, 3000, Leuven, Belgium.,Center for Human Genetics, Leuven Institute for Neuroscience and Disease, KU Leuven, O&N 4, Herestraat 49 Box 602, Leuven, 3000, Belgium
| | - Jean-Christophe Marine
- VIB/Center for the Biology of Disease, KU Leuven, O&N 4, Herestraat 49 Box 602, 3000, Leuven, Belgium.,Center for Human Genetics, Leuven Institute for Neuroscience and Disease, KU Leuven, O&N 4, Herestraat 49 Box 602, Leuven, 3000, Belgium
| | - Sergio Morini
- Department of Medicine, Campus Bio-Medico University, via Alvaro del Portillo 21, 00128 Rome, Italy
| | - Claudia Bagni
- Department of Biomedicine and Prevention, University of Rome 'Tor Vergata', via Montpellier 1, 00133 Rome, Italy.,VIB/Center for the Biology of Disease, KU Leuven, O&N 4, Herestraat 49 Box 602, 3000, Leuven, Belgium.,Center for Human Genetics, Leuven Institute for Neuroscience and Disease, KU Leuven, O&N 4, Herestraat 49 Box 602, Leuven, 3000, Belgium.,Department of Fundamental Neuroscience, University of Lausanne, Rue du Bugnon 9, 1005 Lausanne, Switzerland
| |
Collapse
|
5
|
Sarkar D, Leung EY, Baguley BC, Finlay GJ, Askarian-Amiri ME. Epigenetic regulation in human melanoma: past and future. Epigenetics 2015; 10:103-21. [PMID: 25587943 PMCID: PMC4622872 DOI: 10.1080/15592294.2014.1003746] [Citation(s) in RCA: 216] [Impact Index Per Article: 21.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
The development and progression of melanoma have been attributed to independent or combined genetic and epigenetic events. There has been remarkable progress in understanding melanoma pathogenesis in terms of genetic alterations. However, recent studies have revealed a complex involvement of epigenetic mechanisms in the regulation of gene expression, including methylation, chromatin modification and remodeling, and the diverse activities of non-coding RNAs. The roles of gene methylation and miRNAs have been relatively well studied in melanoma, but other studies have shown that changes in chromatin status and in the differential expression of long non-coding RNAs can lead to altered regulation of key genes. Taken together, they affect the functioning of signaling pathways that influence each other, intersect, and form networks in which local perturbations disturb the activity of the whole system. Here, we focus on how epigenetic events intertwine with these pathways and contribute to the molecular pathogenesis of melanoma.
Collapse
Key Words
- 5hmC, 5-hydroxymethylcytosine
- 5mC, 5-methylcytosine
- ACE, angiotensin converting enzyme
- ANCR, anti-differentiation non-coding RNA
- ANRIL, antisense noncoding RNA in INK4 locus
- ASK1, apoptosis signal-regulating kinase 1
- ATRA, all-trans retinoic acid
- BANCR, BRAF-activated non-coding RNA
- BCL-2, B-cell lymphoma 2
- BRAF, B-Raf proto-oncogene, serine/threonine kinase
- BRG1, ATP-dependent helicase SMARCA4
- CAF-1, chromatin assembly factor-1
- CBX7, chromobox homolog 7
- CCND1, cyclin D1
- CD28, cluster of differentiation 28
- CDK, cyclin-dependent kinase
- CDKN2A/B, cyclin-dependent kinase inhibitor 2A/B
- CHD8, chromodomain-helicase DNA-binding protein 8
- CREB, cAMP response element-binding protein
- CUDR, cancer upregulated drug resistant
- Cdc6, cell division cycle 6
- DNA methylation/demethylation
- DNMT, DNA methyltransferase
- EMT, epithelial-mesenchymal transition
- ERK, extracellular signal-regulated kinase
- EZH2, enhancer of zeste homolog 2
- GPCRs, G-protein coupled receptors
- GSK3a, glycogen synthase kinase 3 α
- GWAS, genome-wide association study
- HDAC, histone deacetylase
- HOTAIR, HOX antisense intergenic RNA
- IAP, inhibitor of apoptosis
- IDH2, isocitrate dehydrogenase
- IFN, interferon, interleukin 23
- JNK, Jun N-terminal kinase
- Jak/STAT, Janus kinase/signal transducer and activator of transcription
- MAFG, v-maf avian musculoaponeurotic fibrosarcoma oncogene homolog G
- MALAT1, metastasis-associated lung adenocarcinoma transcript 1
- MAPK, mitogen-activated protein kinase
- MC1R, melanocortin-1 receptor
- MGMT, O6-methylguanine-DNA methyltransferase
- MIF, macrophage migration inhibitory factor
- MITF, microphthalmia-associated transcription factor
- MRE, miRNA recognition element
- MeCP2, methyl CpG binding protein 2
- NF-κB, nuclear factor kappa-light-chain-enhancer of activated B cells
- NOD, nucleotide-binding and oligomerization domain
- PBX, pre-B-cell leukemia homeobox
- PEDF, pigment epithelium derived factor
- PI3K, phosphatidylinositol-4, 5-bisphosphate 3-kinase
- PIB5PA, phosphatidylinositol-4, 5-biphosphate 5-phosphatase A
- PKA, protein kinase A
- PRC, polycomb repressor complex
- PSF, PTB associated splicing factor
- PTB, polypyrimidine tract-binding
- PTEN, phosphatase and tensin homolog
- RARB, retinoic acid receptor-β2
- RASSF1A, Ras association domain family 1A
- SETDB1, SET Domain, bifurcated 1
- SPRY4, Sprouty 4
- STAU1, Staufen1
- SWI/SNF, SWItch/Sucrose Non-Fermentable
- TCR, T-cell receptor
- TET, ten eleven translocase
- TGF β, transforming growth factor β
- TINCR, tissue differentiation-inducing non-protein coding RNA
- TOR, target of rapamycin
- TP53, tumor protein 53
- TRAF6, TNF receptor-associated factor 6
- UCA1, urothelial carcinoma-associated 1
- ceRNA, competitive endogenous RNAs
- chromatin modification
- chromatin remodeling
- epigenetics
- gene regulation
- lncRNA, long ncRNA
- melanoma
- miRNA, micro RNA
- ncRNA, non-coding RNA
- ncRNAs
- p14ARF, p14 alternative reading frame
- p16INK4a, p16 inhibitor of CDK4
- pRB, retinoblastoma protein
- snoRNA, small nucleolar RNA
- α-MSHm, α-melanocyte stimulating hormone
Collapse
Affiliation(s)
- Debina Sarkar
- a Auckland Cancer Society Research Center ; University of Auckland ; Auckland , New Zealand
| | | | | | | | | |
Collapse
|
6
|
Toxic epidermal necrolysis induced by vemurafenib. ACTAS DERMO-SIFILIOGRAFICAS 2015. [DOI: 10.1016/j.adengl.2015.07.014] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022] Open
|
7
|
Lapresta A, Dotor A, González-Herrada C. Toxic epidermal necrolysis induced by vemurafenib. ACTAS DERMO-SIFILIOGRAFICAS 2015; 106:682-3. [PMID: 26051238 DOI: 10.1016/j.ad.2015.03.008] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2014] [Revised: 01/09/2015] [Accepted: 03/05/2015] [Indexed: 11/26/2022] Open
Affiliation(s)
- A Lapresta
- Servicio de Dermatología, Hospital Universitario de Getafe, Getafe, Madrid, España.
| | - A Dotor
- Servicio de Anatomía Patológica, Hospital Universitario de Getafe, Getafe, Madrid, España
| | - C González-Herrada
- Servicio de Dermatología, Hospital Universitario de Getafe, Getafe, Madrid, España
| |
Collapse
|
8
|
Bhatia S, Emdad L, Das SK, Hamed H, Dent P, Sarkar D, Fisher PB. Non-BRAF targeted therapies for melanoma: protein kinase inhibitors in Phase II clinical trials. Expert Opin Investig Drugs 2014; 23:489-500. [DOI: 10.1517/13543784.2014.884558] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
|
9
|
Abstract
The PI3K-PTEN-AKT signaling pathway is involved in various cellular activities, including proliferation, migration, cell growth, cell survival and differentiation during adult homeostasis as well as in tumorigenesis. It has been suggested that the constitutive activation of PI3K/AKT signaling with concurrent loss of function of the tumor suppressor molecule PTEN contributes to cancer formation. Members of the PI3K-PTEN-AKT pathway, including these proteins and mTOR, are altered in melanoma tumors and cell lines. A hallmark of activation of the pathway is the loss of function of PTEN. Indeed, loss of heterozygosity of PTEN has been observed in approximately 30% of human melanomas, implicating this signaling pathway in this cancer. PI3K signaling activation, via loss of PTEN function, can inhibit proapoptotic genes such as the FoxO family of transcription factors, while inducing cell growth- and cell survival-related elements such as p70S6K and AKT. Determining how the PI3K-PTEN-AKT signaling pathway, alone or in cooperation with other pathways, orchestrates the induction of target genes involved in a diverse range of activities is a major challenge in research into melanoma initiation and progression. Moreover, the acquisition of basic knowledge will help patient management with appropriate therapies that are already, or will shortly be, on the market.
Collapse
Affiliation(s)
- Alejandro Conde-Perez
- Institut Curie, Developmental Genetics of Melanocytes, Bat. 110, 91405, Orsay, France
| | | |
Collapse
|
10
|
Machado D, Shishido SM, Queiroz KCS, Oliveira DN, Faria ALC, Catharino RR, Spek CA, Ferreira CV. Irradiated riboflavin diminishes the aggressiveness of melanoma in vitro and in vivo. PLoS One 2013; 8:e54269. [PMID: 23342114 PMCID: PMC3546980 DOI: 10.1371/journal.pone.0054269] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2012] [Accepted: 12/10/2012] [Indexed: 02/01/2023] Open
Abstract
Melanoma is one of the most aggressive skin cancers due to its high capacity to metastasize. Treatment of metastatic melanomas is challenging for clinicians, as most therapeutic agents have failed to demonstrate improved survival. Thus, new candidates with antimetastatic activity are much needed. Riboavin (RF) is a component of the vitamin B complex and a potent photosensitizer. Previously, our group showed that the RF photoproducts (iRF) have potential as an antitumoral agent. Hence, we investigated the capacity of iRF on modulating melanoma B16F10 cells aggressiveness in vitro and in vivo. iRF decreases B16F10 cells survival by inhibiting mTOR as well as Src kinase. Moreover, melanoma cell migration was disrupted after treatment with iRF, mainly by inhibition of metalloproteinase (MMP) activity and expression, and by increasing TIMP expression. Interestingly, we observed that the Hedgehog (HH) pathway was inhibited by iRF. Two mediators of HH signaling, GLI1 and PTCH, were downregulated, while SUFU expression (an inhibitor of this cascade) was enhanced. Furthermore, inhibition of HH pathway signaling by cyclopamine and Gant 61 potentiated the antiproliferative action of RF. Accordingly, when a HH ligand was applied, the effect of iRF was almost completely abrogated. Our findings indicate that Hedgehog pathway is involved on the modulation of melanoma cell aggressiveness by iRF. Moreover, iRF treatment decreased pulmonary tumor formation in a murine experimental metastasis model. Research to clarify the molecular action of flavins, in vivo, is currently in progress. Taken together, the present data provides evidence that riboflavin photoproducts may provide potential candidates for improving the efficiency of melanoma treatment.
Collapse
Affiliation(s)
- Daisy Machado
- Laboratory of Bioassays and Signal Transduction, Department of Biochemistry, Institute of Biology, University of Campinas, Campinas, São Paulo, Brazil
| | - Silvia M. Shishido
- Laboratory of Bioassays and Signal Transduction, Department of Biochemistry, Institute of Biology, University of Campinas, Campinas, São Paulo, Brazil
| | - Karla C. S. Queiroz
- Center for Experimental and Molecular Medicine, Academic Medical Center, University of Amsterdam, Meibergdreef, Amsterdam, The Netherlands
| | - Diogo N. Oliveira
- Laboratory Innovare of Biomarkers, Faculty of Medical Sciences, University of Campinas, Campinas, São Paulo, Brazil
| | - Ana L. C. Faria
- Laboratory Innovare of Biomarkers, Faculty of Medical Sciences, University of Campinas, Campinas, São Paulo, Brazil
| | - Rodrigo R. Catharino
- Laboratory Innovare of Biomarkers, Faculty of Medical Sciences, University of Campinas, Campinas, São Paulo, Brazil
| | - C. Arnold Spek
- Center for Experimental and Molecular Medicine, Academic Medical Center, University of Amsterdam, Meibergdreef, Amsterdam, The Netherlands
| | - Carmen V. Ferreira
- Laboratory of Bioassays and Signal Transduction, Department of Biochemistry, Institute of Biology, University of Campinas, Campinas, São Paulo, Brazil
| |
Collapse
|