EXPERIMENTAL RESEARCH
 
KEYWORDS
TOPICS
ABSTRACT
Introduction:
Drug resistance has become a huge challenge in melanoma. This study aimed to explore the molecular mechanism by which acetylshikonin (AceS) inhibits melanoma with drug resistance to BRAF inhibitor (BRAFi).

Material and methods:
To identify potential targets of AceS in drug-resistant melanoma, we intersected AceS targets predicted by SwissTargetPrediction and differentially expressed genes in drug-resistant melanoma identified from the GSE203545 dataset. Drug-resistant M14 cells were treated with AceS with or without PLX4720, a BRAFi. Cell proliferation, migration, invasion, and apoptosis were investigated. MMP3 was quantified by qPCR and immunofluorescence. Extracellular matrix (ECM) was evaluated by MMP1 and MMP9. Also, drug-resistant M14 cells were transfected by MMP3 siRNA. Moreover, drug-resistant M14 cells with MMP3 overexpression were treated with AceS. In vivo, a subcutaneous tumor-bearing nude mouse model was established to validate the effects of AceS in drug-resistant melanoma.

Results:
MMP3 was found to be the key target of AceS in drug-resistant melanoma. AceS significantly inhibited proliferation, migration, and invasion (p < 0.05), and promoted apoptosis (p < 0.05); it could increase the sensitivity of drug-resistant M14 cells to PLX4720 (p < 0.05). MMP3 silencing contributed to the decreases in proliferation, migration, and invasion, and the increased apoptosis (p < 0.05). Significantly, MMP3 overexpression reversed the effects caused by AceS (p < 0.05). AceS inhibited tumor growth; it reduced MMP1, MMP3, and MMP9 in vivo. AceS promoted tumor sensitivity to PLX4720 in vivo.

Conclusions:
AceS downregulated MMP3 to modulate ECM remodeling, thereby inhibiting drug-resistant melanoma, and enhanced the cell response to BRAFi. Our findings provide intriguing insights into drug-resistant melanoma.
REFERENCES (37)
1.
Bray F, Laversanne M, Sung H, et al. Global cancer statistics 2022: GLOBOCAN estimates of incidence and mortality worldwide for 36 cancers in 185 countries. CA Cancer J Clin 2024; 74: 229-63.
 
2.
Long GV, Swetter SM, Menzies AM, Gershenwald JE, Scolyer RA. Cutaneous melanoma. Lancet 2023; 402: 485-502.
 
3.
Timar J, Ladanyi A. Molecular pathology of skin melanoma: epidemiology, differential diagnostics, prognosis and therapy prediction. Int J Mol Sci 2022; 23: 5384.
 
4.
Colombino M, Casula M, Paliogiannis P, et al. Heterogeneous pathogenesis of melanoma: BRAF mutations and beyond. Crit Rev Oncol Hematol 2024; 201: 104435.
 
5.
Zhong J, Yan W, Wang C, et al. BRAF inhibitor resistance in melanoma: mechanisms and alternative therapeutic strategies. Curr Treat Options Oncol 2022; 23: 1503-21.
 
6.
Chen W, Park JI. Tumor cell resistance to the inhibition of BRAF and MEK1/2. Int J Mol Sci 2023; 24: 14837.
 
7.
Suhaimi SA, Chan SC, Rosli R. Matrix metallopeptidase 3 polymorphisms: emerging genetic markers in human breast cancer metastasis. J Breast Cancer 2020; 23: 1-9.
 
8.
Shao L, Liu W, Zhang C, et al. The role and function of secretory protein matrix metalloproteinase-3 (MMP3) in cervical cancer. Iran J Public Health 2024; 53: 855-66.
 
9.
Seehawer M, Li Z, Nishida J, et al. Loss of Kmt2c or Kmt2d drives brain metastasis via KDM6A-dependent upregulation of MMP3. Nat Cell Biol 2024; 26: 1165-75.
 
10.
Quinones-Diaz BI, Reyes-Gonzalez JM, Sanchez-Guzman V, et al. MicroRNA-18a-5p suppresses tumor growth via targeting matrix metalloproteinase-3 in cisplatin-resistant ovarian cancer. Front Oncol 2020; 10: 602670.
 
11.
Lawicki P, Malinowski P, Motyka J, et al. Plasma levels of metalloproteinase 3 (MMP-3) and metalloproteinase 7 (MMP-7) as new candidates for tumor biomarkers in diagnostic of breast cancer patients. J Clin Med 2023; 12: 2618.
 
12.
Nunomura J, Nakano R, Naruke A, et al. Interleukin-1beta triggers matrix metalloprotease-3 expression through p65/RelA activation in melanoma cells. PLoS One 2022; 17: e0278220.
 
13.
Zhou S, Lu J, Liu S, et al. Role of the tumor microenvironment in malignant melanoma organoids during the development and metastasis of tumors. Front Cell Dev Biol 2023; 11: 1166916.
 
14.
Kalli M, Poskus MD, Stylianopoulos T, Zervantonakis IK. Beyond matrix stiffness: targeting force-induced cancer drug resistance. Trends Cancer 2023; 9: 937-54.
 
15.
Marusak C, Thakur V, Li Y, et al. Targeting extracellular matrix remodeling restores BRAF inhibitor sensitivity in BRAFi-resistant melanoma. Clin Cancer Res 2020; 26: 6039-50.
 
16.
Pittayapruek P, Meephansan J, Prapapan O, Komine M, Ohtsuki M. Role of matrix metalloproteinases in photoaging and photocarcinogenesis. Int J Mol Sci 2016; 17: 868.
 
17.
Lin H, Ma X, Yang X, et al. Natural shikonin and acetyl-shikonin improve intestinal microbial and protein composition to alleviate colitis-associated colorectal cancer. Int Immunopharmacol 2022; 111: 109097.
 
18.
Zhang Z, Shen C, Zhou F, Zhang Y. Shikonin potentiates therapeutic efficacy of oxaliplatin through reactive oxygen species-mediated intrinsic apoptosis and endoplasmic reticulum stress in oxaliplatin-resistant colorectal cancer cells. Drug Dev Res 2023; 84: 542-55.
 
19.
Lin SS, Chang TM, Wei AI, et al. Acetylshikonin induces necroptosis via the RIPK1/RIPK3-dependent pathway in lung cancer. Aging 2023; 15: 14900-14.
 
20.
Cha HS, Lee HK, Park SH, Nam MJ. Acetylshikonin induces apoptosis of human osteosarcoma U2OS cells by triggering ROS-dependent multiple signal pathways. Toxicol In Vitro 2023; 86: 105521.
 
21.
Jiang S, Wang H, Guo Y, Liu Z, Song W. Acetylshikonin inhibits the migration and invasion of A375 cells by reversing EMT process via the PI3K/Akt/mTOR pathway. Biotechnol Biotechnol Equipment 2019; 33: 699-706.
 
22.
Rossi A, Roberto M, Panebianco M, et al. Drug resistance of BRAF-mutant melanoma: review of up-to-date mechanisms of action and promising targeted agents. Eur J Pharmacol 2019; 862: 172621.
 
23.
Zhang Z, Bai J, Zeng Y, et al. Pharmacology, toxicity and pharmacokinetics of acetylshikonin: a review. Pharm Biol 2020; 58: 950-8.
 
24.
Shon JC, Phuc NM, Kim WC, et al. Acetylshikonin is a novel non-selective cytochrome P450 inhibitor. Biopharm Drug Dispos 2017; 38: 553-6.
 
25.
Hao G, Zhai J, Jiang H, et al. Acetylshikonin induces apoptosis of human leukemia cell line K562 by inducing S phase cell cycle arrest, modulating ROS accumulation, depleting Bcr-Abl and blocking NF-kappaB signaling. Biomed Pharmacother 2020; 122: 109677.
 
26.
Wang J, Iannarelli R, Pucciarelli S, et al. Acetylshikonin isolated from Lithospermum erythrorhizon roots inhibits dihydrofolate reductase and hampers autochthonous mammary carcinogenesis in Delta16HER2 transgenic mice. Pharmacol Res 2020; 161: 105123.
 
27.
Olczak M, Orzechowska MJ, Bednarek AK, Lipinski M. The transcriptomic profiles of ESR1 and MMP3 stratify the risk of biochemical recurrence in primary prostate cancer beyond clinical features. Int J Mol Sci 2023; 24: 8399.
 
28.
Polz A, Morshed K, Drop B, Polz-Dacewicz M. Could MMP3 and MMP9 serve as biomarkers in EBV-related oropharyngeal cancer. Int J Mol Sci 2024; 25: 2561.
 
29.
Hu HF, Xu WW, Zhang WX, et al. Identification of miR-515-3p and its targets, vimentin and MMP3, as a key regulatory mechanism in esophageal cancer metastasis: functional and clinical significance. Signal Transduct Target Ther 2020; 5: 271.
 
30.
Liang M, Wang J, Wu C, et al. Targeting matrix metalloproteinase MMP3 greatly enhances oncolytic virus mediated tumor therapy. Transl Oncol 2021; 14: 101221.
 
31.
Taha EA, Sogawa C, Okusha Y, et al. Knockout of MMP3 weakens solid tumor organoids and cancer extracellular vesicles. Cancers 2020; 12: 1260.
 
32.
Yuan Z, Li Y, Zhang S, et al. Extracellular matrix remodeling in tumor progression and immune escape: from mechanisms to treatments. Mol Cancer 2023; 22: 48.
 
33.
Prakash J, Shaked Y. The interplay between extracellular matrix remodeling and cancer therapeutics. Cancer Discov 2024; 14: 1375-88.
 
34.
Popovic A, Tartare-Deckert S. Role of extracellular matrix architecture and signaling in melanoma therapeutic resistance. Front Oncol 2022; 12: 924553.
 
35.
Vempati P, Karagiannis ED, Popel AS. A biochemical model of matrix metalloproteinase 9 activation and inhibition. J Biol Chem 2007; 282: 37585-96.
 
36.
Steenport M, Khan KM, Du B, et al. Matrix metalloproteinase (MMP)-1 and MMP-3 induce macrophage MMP-9: evidence for the role of TNF-alpha and cyclooxygenase-2. J Immunol 2009; 183: 8119-27.
 
37.
Alge-Priglinger CS, Kreutzer T, Obholzer K, et al. Oxidative stress-mediated induction of MMP-1 and MMP-3 in human RPE cells. Invest Ophthalmol Vis Sci 2009; 50: 5495-503.
 
eISSN:1896-9151
ISSN:1734-1922
Journals System - logo
Scroll to top