1
|
Pacifici F, Chiereghin F, D’Orazio M, Malatesta G, Infante M, Fazio F, Bertinato C, Donadel G, Martinelli E, De Lorenzo A, Della-Morte D, Pastore D. Patch-Based Far-Infrared Radiation (FIR) Therapy Does Not Impact Cell Tracking or Motility of Human Melanoma Cells In Vitro. Curr Issues Mol Biol 2024; 46:10026-10037. [PMID: 39329951 PMCID: PMC11429816 DOI: 10.3390/cimb46090599] [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: 08/05/2024] [Revised: 08/31/2024] [Accepted: 09/05/2024] [Indexed: 09/28/2024] Open
Abstract
Far-Infrared Radiation (FIR) is emerging as a novel non-invasive tool for mitigating inflammation and oxidative stress, offering potential benefits for certain medical conditions such as cardiovascular disease and chronic inflammatory disorders. We previously demonstrated that the application of patch-based FIR therapy on human umbilical vein endothelial cells (HUVECs) reduced the expression of inflammatory biomarkers and the levels of reactive oxygen species (ROS). Several in vitro studies have shown the inhibitory effects of FIR therapy on cell growth in different cancer cells (including murine melanoma cells), mainly using the wound healing assay, without direct cell motility or tracking analysis. The main objective of the present study was to conduct an in-depth analysis of single-cell motility and tracking during the wound healing assay, using an innovative high-throughput technique in the human melanoma cell line M14/C2. This technique evaluates various motility descriptors, such as average velocity, average curvature, average turning angle, and diffusion coefficient. Our results demonstrated that patch-based FIR therapy did not impact cell proliferation and viability or the activation of mitogen-activated protein kinases (MAPKs) in the human melanoma cell line M14/C2. Moreover, no significant differences in cell motility and tracking were observed between control cells and patch-treated cells. Altogether, these findings confirm the beneficial effects of the in vitro application of patch-based FIR therapy in human melanoma cell lines, although such effects need to be confirmed in future in vivo studies.
Collapse
Affiliation(s)
- Francesca Pacifici
- Department of Human Sciences and Promotion of the Quality of Life, San Raffaele Roma Open University, 00166 Rome, Italy; (F.P.); (F.C.); (D.D.-M.)
- Interdisciplinary Center for Advanced Studies on Lab-on-Chip and Organ-on-Chip Applications (IC-LOC), University of Rome Tor Vergata, 00133 Rome, Italy; (M.D.); (E.M.)
| | - Francesca Chiereghin
- Department of Human Sciences and Promotion of the Quality of Life, San Raffaele Roma Open University, 00166 Rome, Italy; (F.P.); (F.C.); (D.D.-M.)
| | - Michele D’Orazio
- Interdisciplinary Center for Advanced Studies on Lab-on-Chip and Organ-on-Chip Applications (IC-LOC), University of Rome Tor Vergata, 00133 Rome, Italy; (M.D.); (E.M.)
- Department of Electronic Engineering, University of Rome Tor Vergata, 00133 Rome, Italy
| | - Gina Malatesta
- Department of Biomedicine and Prevention, University of Rome Tor Vergata, 00133 Rome, Italy; (G.M.); (A.D.L.)
| | - Marco Infante
- Section of Diabetes & Metabolic Disorders, UniCamillus, Saint Camillus International University of Health Sciences, Via di Sant’Alessandro 8, 00131 Rome, Italy;
| | - Federica Fazio
- Department of Medical and Surgery Sciences, University “Magna Graecia” of Catanzaro, 8810 Catanzaro, Italy;
| | - Chiara Bertinato
- Department of Cellular, Computational and Integrative Biology-CIBO, University of Trento, 38123 Trento, Italy;
| | - Giulia Donadel
- Department of Clinical Sciences and Translational Medicine, University of Rome Tor Vergata, 00133 Rome, Italy;
| | - Eugenio Martinelli
- Interdisciplinary Center for Advanced Studies on Lab-on-Chip and Organ-on-Chip Applications (IC-LOC), University of Rome Tor Vergata, 00133 Rome, Italy; (M.D.); (E.M.)
- Department of Electronic Engineering, University of Rome Tor Vergata, 00133 Rome, Italy
| | - Antonino De Lorenzo
- Department of Biomedicine and Prevention, University of Rome Tor Vergata, 00133 Rome, Italy; (G.M.); (A.D.L.)
| | - David Della-Morte
- Department of Human Sciences and Promotion of the Quality of Life, San Raffaele Roma Open University, 00166 Rome, Italy; (F.P.); (F.C.); (D.D.-M.)
- Interdisciplinary Center for Advanced Studies on Lab-on-Chip and Organ-on-Chip Applications (IC-LOC), University of Rome Tor Vergata, 00133 Rome, Italy; (M.D.); (E.M.)
- Department of Biomedicine and Prevention, University of Rome Tor Vergata, 00133 Rome, Italy; (G.M.); (A.D.L.)
- Department of Neurology, Evelyn F. McKnight Brain Institute, Miller School of Medicine, University of Miami, Miami, FL 33136, USA
| | - Donatella Pastore
- Department of Human Sciences and Promotion of the Quality of Life, San Raffaele Roma Open University, 00166 Rome, Italy; (F.P.); (F.C.); (D.D.-M.)
- Interdisciplinary Center for Advanced Studies on Lab-on-Chip and Organ-on-Chip Applications (IC-LOC), University of Rome Tor Vergata, 00133 Rome, Italy; (M.D.); (E.M.)
| |
Collapse
|
2
|
Kot M, Simiczyjew A, Wądzyńska J, Ziętek M, Matkowski R, Nowak D. Characterization of two melanoma cell lines resistant to BRAF/MEK inhibitors (vemurafenib and cobimetinib). Cell Commun Signal 2024; 22:410. [PMID: 39175042 PMCID: PMC11342534 DOI: 10.1186/s12964-024-01788-3] [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: 02/22/2024] [Accepted: 08/12/2024] [Indexed: 08/24/2024] Open
Abstract
BACKGROUND BRAF (v-raf murine sarcoma viral oncogene homolog B1)/MEK (mitogen-activated protein kinase kinase) inhibitors are used for melanoma treatment. Unfortunately, patients treated with this combined therapy develop resistance to treatment quite quickly, but the mechanisms underlying this phenomenon are not yet fully understood. Here, we report and characterize two melanoma cell lines (WM9 and Hs294T) resistant to BRAF (vemurafenib) and MEK (cobimetinib) inhibitors. METHODS Cell viability was assessed via the XTT test. The level of selected proteins as well as activation of signaling pathways were evaluated using Western blotting. The expression of the chosen genes was assessed by RT-PCR. The distribution of cell cycle phases was analyzed by flow cytometry, and confocal microscopy was used to take photos of spheroids. The composition of cytokines secreted by cells was determined using a human cytokine array. RESULTS The resistant cells had increased survival and activation of ERK kinase in the presence of BRAF/MEK inhibitors. The IC50 values for these cells were over 1000 times higher than for controls. Resistant cells also exhibited elevated activation of AKT, p38, and JNK signaling pathways with increased expression of EGFR, ErbB2, MET, and PDGFRβ receptors as well as reduced expression of ErbB3 receptor. Furthermore, these cells demonstrated increased expression of genes encoding proteins involved in drug transport and metabolism. Resistant cells also exhibited features of epithelial-mesenchymal transition and cancer stem cells as well as reduced proliferation rate and elevated cytokine secretion. CONCLUSIONS In summary, this work describes BRAF/MEK-inhibitor-resistant melanoma cells, allowing for better understanding the underlying mechanisms of resistance. The results may thus contribute to the development of new, more effective therapeutic strategies.
Collapse
Affiliation(s)
- Magdalena Kot
- Department of Cell Pathology, Faculty of Biotechnology, University of Wroclaw, Joliot-Curie 14a, Wroclaw, 50-383, Poland
| | - Aleksandra Simiczyjew
- Department of Cell Pathology, Faculty of Biotechnology, University of Wroclaw, Joliot-Curie 14a, Wroclaw, 50-383, Poland.
| | - Justyna Wądzyńska
- Department of Cell Pathology, Faculty of Biotechnology, University of Wroclaw, Joliot-Curie 14a, Wroclaw, 50-383, Poland
| | - Marcin Ziętek
- Department of Oncology, Division of Surgical Oncology, Wroclaw Medical University, Plac Hirszfelda 12, Wroclaw, 53-413, Poland
- Lower Silesian Oncology, Pulmonology, and Hematology Center, Plac Hirszfelda 12, Wroclaw, 53-413, Poland
| | - Rafał Matkowski
- Department of Oncology, Division of Surgical Oncology, Wroclaw Medical University, Plac Hirszfelda 12, Wroclaw, 53-413, Poland
- Lower Silesian Oncology, Pulmonology, and Hematology Center, Plac Hirszfelda 12, Wroclaw, 53-413, Poland
| | - Dorota Nowak
- Department of Cell Pathology, Faculty of Biotechnology, University of Wroclaw, Joliot-Curie 14a, Wroclaw, 50-383, Poland
| |
Collapse
|
3
|
Sağraç D, Aydın S, Kırbaş OK, Öztürkoğlu D, Şahin F. Extracellular vesicles derived from human foreskin cells (hFS-Exo) accelerate cell migration and angiogenesis through MAPK pathway: an in vitro study. Mol Biol Rep 2024; 51:471. [PMID: 38551706 DOI: 10.1007/s11033-024-09378-9] [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: 10/31/2023] [Accepted: 02/26/2024] [Indexed: 04/02/2024]
Abstract
BACKGROUND Wound healing is one of the important processes in the body. Attempts to create new drugs are of interest due to the side effects of natural and chemical wound healing compounds. To overcome this obstacle, stem cells have been used as healing agents. However, both difficulties in collection and risks such as rejection and teratoma in the recipient body have limited the use of stem cells, directly. Since the potential content of the stem cells can be transferred to the recipient cells by vesicles, small extracellular vesicles have recently become prominent agents. METHODS AND RESULTS The wound-healing effect of extracellular vesicles derived from foreskin cells was investigated in both keratinocyte and endothelial cells. Migration assay, RT-PCR, Col1a1 ELISA and Western Blot experiments were utilized to reveal healing effect of EVs and its possible molecular pathways. EV-treated groups exhibited more proliferative, invasive, and migrative characteristics. When comparing to the control group, new vessel formation was induced in EV groups. An increase in gene levels of growth factors related to wound healing and change in the mitogen-activated protein kinase (MAPK) signaling pathway proteins in EV-treated groups were determined. Possible molecular mechanisms underlying cell movements were associated with the MAPK pathway. It was found that human foreskin cell EVs (hFS-Exo) may have a potential to heal wounds in a short period of time by triggering the MAPK pathway. CONCLUSIONS hFS-Exo could be a new promising wound healing agent in the future.
Collapse
Affiliation(s)
- Derya Sağraç
- Department of Genetics and Bioengineering, Faculty of Engineering, Yeditepe University, İstanbul, Turkey
| | - Safa Aydın
- Department of Genetics and Bioengineering, Faculty of Engineering, Yeditepe University, İstanbul, Turkey
| | - Oğuz Kaan Kırbaş
- Department of Genetics and Bioengineering, Faculty of Engineering, Yeditepe University, İstanbul, Turkey
| | - Dilek Öztürkoğlu
- Department of Genetics and Bioengineering, Faculty of Engineering, Yeditepe University, İstanbul, Turkey
| | - Fikrettin Şahin
- Department of Genetics and Bioengineering, Faculty of Engineering, Yeditepe University, İstanbul, Turkey.
| |
Collapse
|
4
|
Singvogel K, Schittek B. Dormancy of cutaneous melanoma. Cancer Cell Int 2024; 24:88. [PMID: 38419052 PMCID: PMC10903048 DOI: 10.1186/s12935-024-03278-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2023] [Accepted: 02/19/2024] [Indexed: 03/02/2024] Open
Abstract
Many cancer-related deaths including melanoma result from metastases that develop months or years after the initial cancer therapy. Even the most effective drugs and immune therapies rarely eradicate all tumor cells. Instead, they strongly reduce cancer burden, permitting dormant cancer cells to persist in niches, where they establish a cellular homeostasis with their host without causing clinical symptoms. Dormant cancers respond poorly to most drugs and therapies since they do not proliferate and hide in niches. It therefore remains a major challenge to develop novel therapies for dormant cancers. In this review we focus on the mechanisms regulating the initiation of cutaneous melanoma dormancy as well as those which are involved in reawakening of dormant cutaneous melanoma cells. In recent years the role of neutrophils and niche components in reawakening of melanoma cells came into focus and indicate possible future therapeutic applications. Sophisticated in vitro and in vivo melanoma dormancy models are needed to make progress in this field and are discussed.
Collapse
Affiliation(s)
- Kathrin Singvogel
- Division of Dermatooncology, Department of Dermatology, University of Tübingen, Liebermeisterstr. 25, D -72076 , Tübingen, Germany
| | - Birgit Schittek
- Division of Dermatooncology, Department of Dermatology, University of Tübingen, Liebermeisterstr. 25, D -72076 , Tübingen, Germany.
- Cluster of Excellence iFIT (EXC 2180) "Image-Guided and Functionally Instructed Tumor Therapies", University of Tübingen, Tübingen, Germany.
| |
Collapse
|
5
|
Duan L, Zhao Y, Jia J, Chao T, Wang H, Liang Y, Lou Y, Zheng Q, Wang H. Myeloid-restricted CD68 deficiency attenuates atherosclerosis via inhibition of ROS-MAPK-apoptosis axis. Biochim Biophys Acta Mol Basis Dis 2023; 1869:166698. [PMID: 36965676 DOI: 10.1016/j.bbadis.2023.166698] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2022] [Revised: 02/24/2023] [Accepted: 03/19/2023] [Indexed: 03/27/2023]
Abstract
In atherosclerosis, macrophages derived from blood monocytes contribute to non-resolving inflammation, which subsequently primes necrotic core formation, and ultimately triggers acute thrombotic vascular disease. Nevertheless, little is known about how inflammatory cells, especially the macrophages fuel atherosclerosis. CD68, a unique class D scavenger receptor (SRD) family member, is specifically expressed in monocytes/macrophages and remarkably up-regulated upon oxidized low-density lipoprotein (ox-LDL) stimulation. Nonetheless, whether and how myeloid-specific CD68 affects atherosclerosis remains to be defined. To determine the essential in vivo role and mechanism linking CD68 to atherosclerosis, we engineered global and myeloid-specific CD68-deficient mice on an ApoE background. On Western diet, both the mice with global and the myeloid-restricted deletion of CD68 on ApoE background attenuated atherosclerosis, accompanied by diminished immune/inflammatory cell burden and necrotic core content, but increased smooth muscle cell content in atherosclerotic plaques. In vitro experiments revealed that CD68 deficiency in macrophages resulted in attenuated ox-LDL-induced macrophage apoptosis. Additionally, CD68 deficiency suppressed ROS production, while removal of ROS can markedly reversed this effect. We further showed that CD68 deficiency affected apoptosis through inactivation of the mitogen-activated protein kinase (MAPK) pathway. Our findings establish CD68 as a macrophage lineage-specific regulator of "ROS-MAPK-apoptosis" axis, thus providing a previously unknown mechanism for the prominence of CD68 as a risk factor for coronary artery disease. Its therapeutic inhibition may provide a potent lever to alleviate the cardiovascular disease.
Collapse
Affiliation(s)
- Liangwei Duan
- Henan Key Laboratory of Immunology and Targeted Drugs, Xinxiang Medical University, Xinxiang, Henan, China; Henan Collaborative Innovation Center of Molecular Diagnosis and Laboratory Medicine, School of Laboratory Medicine, Xinxiang Medical University, Xinxiang, Henan, China
| | - Yucong Zhao
- Henan Key Laboratory of Immunology and Targeted Drugs, Xinxiang Medical University, Xinxiang, Henan, China; Henan Collaborative Innovation Center of Molecular Diagnosis and Laboratory Medicine, School of Laboratory Medicine, Xinxiang Medical University, Xinxiang, Henan, China
| | - Jing Jia
- Henan Key Laboratory of Immunology and Targeted Drugs, Xinxiang Medical University, Xinxiang, Henan, China; Henan Collaborative Innovation Center of Molecular Diagnosis and Laboratory Medicine, School of Laboratory Medicine, Xinxiang Medical University, Xinxiang, Henan, China
| | - Tianzhu Chao
- College of Food Science and Pharmaceutical Engineering, Zaozhuang University, Zaozhuang, Shandong, China
| | - Hao Wang
- Henan Key Laboratory of Immunology and Targeted Drugs, Xinxiang Medical University, Xinxiang, Henan, China; Henan Collaborative Innovation Center of Molecular Diagnosis and Laboratory Medicine, School of Laboratory Medicine, Xinxiang Medical University, Xinxiang, Henan, China
| | - Yinming Liang
- Henan Key Laboratory of Immunology and Targeted Drugs, Xinxiang Medical University, Xinxiang, Henan, China; Henan Collaborative Innovation Center of Molecular Diagnosis and Laboratory Medicine, School of Laboratory Medicine, Xinxiang Medical University, Xinxiang, Henan, China
| | - Yunwei Lou
- Henan Key Laboratory of Immunology and Targeted Drugs, Xinxiang Medical University, Xinxiang, Henan, China; Henan Collaborative Innovation Center of Molecular Diagnosis and Laboratory Medicine, School of Laboratory Medicine, Xinxiang Medical University, Xinxiang, Henan, China
| | - Qianqian Zheng
- Henan Key Laboratory of Immunology and Targeted Drugs, Xinxiang Medical University, Xinxiang, Henan, China; Henan Collaborative Innovation Center of Molecular Diagnosis and Laboratory Medicine, School of Laboratory Medicine, Xinxiang Medical University, Xinxiang, Henan, China.
| | - Hui Wang
- Henan Key Laboratory of Immunology and Targeted Drugs, Xinxiang Medical University, Xinxiang, Henan, China; Henan Collaborative Innovation Center of Molecular Diagnosis and Laboratory Medicine, School of Laboratory Medicine, Xinxiang Medical University, Xinxiang, Henan, China.
| |
Collapse
|
6
|
Bai S, Hu S, Dai Y, Jin R, Zhang C, Yao F, Weng Q, Zhai P, Zhao B, Wu X, Chen Y. NRAS promotes the proliferation of melanocytes to increase melanin deposition in Rex rabbits. Genome 2023; 66:1-10. [PMID: 36440769 DOI: 10.1139/gen-2021-0111] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
Melanocytes play a major role in the formation of mammalian fur color and are regulated by several genes. Despite playing the pivotal role in the study of melanoma, the mechanistic role of NRAS (neuroblastoma RAS viral oncogene homolog) in the formation of mammalian epidermal color is still elusive. First of all, the expression levels of NRAS mRNA and protein in the dorsal skin of different colored Rex rabbits were detected by qRT-PCR and Western blot. Then, the subcellular localization of NRAS was identified in melanocytes by indirect immunofluorescence. Next, the expression of NRAS was overexpressed and knocked down in melanocytes, and its efficiency was verified by qRT-PCR and Western blot. Subsequently, NaOH, CCK-8, and Annexin V-FITC were used to verify the changes in melanin content, proliferation, and apoptosis in melanocytes. Finally, we analyzed the regulation of NRAS on other genes (MITF, TYR, DCT, PMEL, and CREB) that affect melanin production. In silico studies showed NRAS as a stable and hydrophilic protein, and it is localized in the cytoplasm and nucleus of melanocytes. The mRNA and protein expression levels of NRAS were significantly different in skin of different colored Rex rabbits, and the highest level was found in black skin (P < 0.01). Moreover, the NRAS demonstrated impact on the proliferation, apoptosis, and melanin production of melanocytes (P < 0.05), and the strong correlation of NRAS with melanin-related genes was evidently observed (P < 0.05). Our results suggested that NRAS can be used as a gene that regulates melanin production and controls melanocyte proliferation and apoptosis, providing a new theoretical basis for studying the mechanism of mammalian fur color formation.
Collapse
Affiliation(s)
- Shaocheng Bai
- College of Animal Science and Technology, Yangzhou University, Yangzhou 225000, China
| | - Shuaishuai Hu
- College of Animal Science and Technology, Yangzhou University, Yangzhou 225000, China
| | - Yingying Dai
- College of Animal Science and Technology, Yangzhou University, Yangzhou 225000, China
| | - Rongshuai Jin
- College of Animal Science and Technology, Yangzhou University, Yangzhou 225000, China
| | - Chen Zhang
- College of Animal Science and Technology, Yangzhou University, Yangzhou 225000, China
| | - Fan Yao
- College of Animal Science and Technology, Yangzhou University, Yangzhou 225000, China
| | - Qiaoqin Weng
- Yuyao Xinnong Rabbit Industry Co., Ltd, Ningbo 315400, China
| | - Pin Zhai
- Jiangsu Academy of Agricultural Sciences, Nanjing 210000, China
| | - Bohao Zhao
- College of Animal Science and Technology, Yangzhou University, Yangzhou 225000, China
| | - Xinsheng Wu
- College of Animal Science and Technology, Yangzhou University, Yangzhou 225000, China.,Joint International Research Laboratory of Agriculture & Agri-Product Safety, Yangzhou University, Yangzhou 225000, China
| | - Yang Chen
- College of Animal Science and Technology, Yangzhou University, Yangzhou 225000, China.,Joint International Research Laboratory of Agriculture & Agri-Product Safety, Yangzhou University, Yangzhou 225000, China
| |
Collapse
|
7
|
Zheng G, Bouamar H, Cserhati M, Zeballos CR, Mehta I, Zare H, Broome L, Hu R, Lai Z, Chen Y, Sharkey FE, Rani M, Halff GA, Cigarroa FG, Sun LZ. Integrin alpha 6 is upregulated and drives hepatocellular carcinoma progression through integrin α6β4 complex. Int J Cancer 2022; 151:930-943. [PMID: 35657344 PMCID: PMC9329238 DOI: 10.1002/ijc.34146] [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: 06/25/2021] [Revised: 05/16/2022] [Accepted: 05/17/2022] [Indexed: 12/24/2022]
Abstract
Integrin α6 (ITGA6) forms integrin receptors with either integrin β1 (ITGB1) or integrin β4 (ITGB4). How it functions to regulate hepatocellular carcinoma (HCC) progression is not well-elucidated. We found that ITGA6 RNA and protein expression levels are significantly elevated in human HCC tissues in comparison with paired adjacent nontumor tissues by RNA sequencing, RT-qPCR, Western blotting and immunofluorescence staining. Stable knockdown of ITGA6 with different ITGA6 shRNA expression lentivectors significantly inhibited proliferation, migration and anchorage-independent growth of HCC cell lines in vitro, and xenograft tumor growth in vivo. The inhibition of anchorage-dependent and -independent growth of HCC cell lines was also confirmed with anti-ITGA6 antibody. ITGA6 knockdown was shown to induce cell-cycle arrest at G0/G1 phase. Immunoprecipitation assay revealed apparent interaction of ITGA6 with ITGB4, but not ITGB1. Expression studies showed that ITGA6 positively regulates the expression of ITGB4 with no or negative regulation of ITGB1 expression. Finally, while high levels of ITGA6 and ITGB4 together were associated with significantly worse survival of HCC patients in TCGA data set, the association was not significant for high levels of ITGA6 and ITGB1. In conclusion, ITGA6 is upregulated in HCC tumors and has a malignant promoting role in HCC cells through integrin α6β4 complex. Thus, integrin α6β4 may be a therapeutic target for treating patients with HCC.
Collapse
Affiliation(s)
- Guixi Zheng
- Department of Cell Systems & Anatomy, University of Texas Health Science Center at San Antonio, TX
- Department of Clinical Laboratory, Qilu Hospital of Shandong University, China
| | - Hakim Bouamar
- Department of Cell Systems & Anatomy, University of Texas Health Science Center at San Antonio, TX
| | - Matyas Cserhati
- Department of Cell Systems & Anatomy, University of Texas Health Science Center at San Antonio, TX
| | - Carla R. Zeballos
- Department of Cell Systems & Anatomy, University of Texas Health Science Center at San Antonio, TX
| | - Isha Mehta
- Department of Cell Systems & Anatomy, University of Texas Health Science Center at San Antonio, TX
| | - Habil Zare
- Department of Cell Systems & Anatomy, University of Texas Health Science Center at San Antonio, TX
| | - Larry Broome
- Department of Cell Systems & Anatomy, University of Texas Health Science Center at San Antonio, TX
| | - Ruolei Hu
- Department of Cell Systems & Anatomy, University of Texas Health Science Center at San Antonio, TX
| | - Zhao Lai
- Greehey Children’s Cancer Research Institute, University of Texas Health Science Center at San Antonio, TX
- Department of Molecular Medicine, University of Texas Health Science Center at San Antonio, TX
| | - Yidong Chen
- Greehey Children’s Cancer Research Institute, University of Texas Health Science Center at San Antonio, TX
- Department of Molecular Medicine, University of Texas Health Science Center at San Antonio, TX
- Department of Population Health Sciences, University of Texas Health Science Center at San Antonio, TX
| | - Francis E. Sharkey
- Department of Pathology and Laboratory Medicine, University of Texas Health Science Center at San Antonio, TX
| | - Meenakshi Rani
- Transplant Center, University of Texas Health Science Center at San Antonio, TX
| | - Glenn A. Halff
- Transplant Center, University of Texas Health Science Center at San Antonio, TX
| | | | - Lu-Zhe Sun
- Department of Cell Systems & Anatomy, University of Texas Health Science Center at San Antonio, TX
| |
Collapse
|
8
|
Mizuta K, Matsubara T, Goto A, Addison WN, Nakatomi M, Matsuo K, Tada-Shigeyama Y, Yaginuma T, Honda H, Yoshioka I, Kokabu S. Plectin promotes tumor formation by B16 mouse melanoma cells via regulation of Rous sarcoma oncogene activity. BMC Cancer 2022; 22:936. [PMID: 36038818 PMCID: PMC9426213 DOI: 10.1186/s12885-022-10033-4] [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: 04/01/2022] [Accepted: 08/24/2022] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Melanoma is a malignant tumor characterized by high proliferation and aggressive metastasis. To address the molecular mechanisms of the proto-oncogene, Rous sarcoma oncogene (Src), which is highly activated and promotes cell proliferation, migration, adhesion, and metastasis in melanoma. Plectin, a cytoskeletal protein, has recently been identified as a Src-binding protein that regulates Src activity in osteoclasts. Plectin is a candidate biomarker of certain tumors because of its high expression and the target of anti-tumor reagents such as ruthenium pyridinecarbothioamide. The molecular mechanisms by which plectin affects melanoma is still unclear. In this study, we examined the role of plectin in melanoma tumor formation. METHODS We used CRISPR/Cas9 gene editing to knock-out plectin in B16 mouse melanoma cells. Protein levels of plectin and Src activity were examined by western blotting analysis. In vivo tumor formation was assessed by subcutaneous injection of B16 cells into nude mice and histological analysis performed after 2 weeks by Hematoxylin-Eosin (H&E) staining. Cell proliferation was evaluated by direct cell count, cell counting kit-8 assays, cyclin D1 mRNA expression and Ki-67 immunostaining. Cell aggregation and adhesion were examined by spheroid formation, dispase-based dissociation assay and cell adhesion assays. RESULTS In in vivo tumor formation assays, depletion of plectin resulted in low-density tumors with large intercellular spaces. In vitro experiments revealed that plectin-deficient B16 cells exhibit reduced cell proliferation and reduced cell-to-cell adhesion. Since Src activity is reduced in plectin-deficient melanomas, we examined the relationship between plectin and Src signaling. Src overexpression in plectin knockout B16 cells rescued cell proliferation and improved cell-to-cell adhesion and cell to extracellular matrix adhesion. CONCLUSION These results suggest that plectin plays critical roles in tumor formation by promoting cell proliferation and cell-to-cell adhesion through Src signaling activity in melanoma cells.
Collapse
Affiliation(s)
- Kana Mizuta
- Division of Molecular Signaling and Biochemistry, Department of Health Improvement, Kyushu Dental University, Kitakyushu, Japan.,Division of Oral Medicine, Department of Science of Physical Function, Kyushu Dental University, Kitakyushu, Japan
| | - Takuma Matsubara
- Division of Molecular Signaling and Biochemistry, Department of Health Improvement, Kyushu Dental University, Kitakyushu, Japan.
| | - Akino Goto
- Division of Molecular Signaling and Biochemistry, Department of Health Improvement, Kyushu Dental University, Kitakyushu, Japan
| | - William N Addison
- Division of Molecular Signaling and Biochemistry, Department of Health Improvement, Kyushu Dental University, Kitakyushu, Japan
| | - Mitsushiro Nakatomi
- Department of Human, Information and Life Sciences, School of Health Sciences, University of Occupational and Environmental Health, Kitakyushu, Japan
| | - Kou Matsuo
- Division of Oral Pathology, Department of Health Improvement, Kyushu Dental University, Kitakyushu, Japan
| | - Yukiyo Tada-Shigeyama
- Division of Dental Anesthesiology, Department of Science of Physical Function, Kyushu Dental University, Kitakyushu, Japan
| | - Tatsuki Yaginuma
- Division of Oral and Maxillofacial Surgery, Department of Science and Physical Function, Kyushu Dental University, Kitakyushu, Japan
| | - Hiromi Honda
- School of Oral Health Sciences, Kyushu Dental University, Kitakyushu, Japan
| | - Izumi Yoshioka
- Division of Oral Medicine, Department of Science of Physical Function, Kyushu Dental University, Kitakyushu, Japan
| | - Shoichiro Kokabu
- Division of Molecular Signaling and Biochemistry, Department of Health Improvement, Kyushu Dental University, Kitakyushu, Japan.
| |
Collapse
|
9
|
Chousakos E, Katsoulas N, Kavantzas N, Stratigos A, Lazaris AC. The role of dual-specificity phosphatase 3 in melanocytic oncogenesis. Exp Dermatol 2022; 31:1466-1476. [PMID: 35899430 DOI: 10.1111/exd.14653] [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: 12/17/2021] [Revised: 07/08/2022] [Accepted: 07/25/2022] [Indexed: 12/01/2022]
Abstract
Dual-specificity phosphatase 3 (DUSP3), also known as Vaccinia H1-related phosphatase, is a protein tyrosine phosphatase that typically performs its major role in the regulation of multiple cellular functions through the dephosphorylation of its diverse and constantly expanding range of substrates. Many of the substrates described so far as well as alterations in the expression or the activity of DUSP3 itself are associated with the development and progression of various types of neoplasms, indicating that DUSP3 may be an important player in oncogenesis and a promising therapeutic target. This review focuses exclusively on DUSP3's contribution to either benign or malignant melanocytic oncogenesis, as many of the established culprit pathways and mechanisms constitute DUSP3's regulatory targets, attempting to synthesize the current knowledge on the matter. The spectrum of the DUSP3 interactions analyzed in this review covers substrates implicated in cellular growth, cell cycle, proliferation, survival, apoptosis, genomic stability/repair, adhesion and migration of tumor melanocytes. Furthermore, the speculations raised, based on the evidence to date, may be considered a fundament for potential research regarding the oncogenesis, evolution, management and therapeutics of melanocytic tumors.
Collapse
Affiliation(s)
- Emmanouil Chousakos
- 1st Department of Pathology, Medical School, National and Kapodistrian University of Athens
| | - Nikolaos Katsoulas
- 1st Department of Pathology, Medical School, National and Kapodistrian University of Athens
| | - Nikolaos Kavantzas
- 1st Department of Pathology, Medical School, National and Kapodistrian University of Athens
| | - Alexandros Stratigos
- 1st Department of Dermatology-Venereology, "Andreas Syggros" Hospital, Medical School, National and Kapodistrian University of Athens
| | - Andreas C Lazaris
- 1st Department of Pathology, Medical School, National and Kapodistrian University of Athens
| |
Collapse
|
10
|
Targeting GPCRs and Their Signaling as a Therapeutic Option in Melanoma. Cancers (Basel) 2022; 14:cancers14030706. [PMID: 35158973 PMCID: PMC8833576 DOI: 10.3390/cancers14030706] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2022] [Revised: 01/27/2022] [Accepted: 01/27/2022] [Indexed: 12/10/2022] Open
Abstract
Simple Summary Sixteen G-protein-coupled receptors (GPCRs) have been involved in melanogenesis or melanomagenesis. Here, we review these GPCRs, their associated signaling, and therapies. Abstract G-protein-coupled receptors (GPCRs) serve prominent roles in melanocyte lineage physiology, with an impact at all stages of development, as well as on mature melanocyte functions. GPCR ligands are present in the skin and regulate melanocyte homeostasis, including pigmentation. The role of GPCRs in the regulation of pigmentation and, consequently, protection against external aggression, such as ultraviolet radiation, has long been established. However, evidence of new functions of GPCRs directly in melanomagenesis has been highlighted in recent years. GPCRs are coupled, through their intracellular domains, to heterotrimeric G-proteins, which induce cellular signaling through various pathways. Such signaling modulates numerous essential cellular processes that occur during melanomagenesis, including proliferation and migration. GPCR-associated signaling in melanoma can be activated by the binding of paracrine factors to their receptors or directly by activating mutations. In this review, we present melanoma-associated alterations of GPCRs and their downstream signaling and discuss the various preclinical models used to evaluate new therapeutic approaches against GPCR activity in melanoma. Recent striking advances in our understanding of the structure, function, and regulation of GPCRs will undoubtedly broaden melanoma treatment options in the future.
Collapse
|
11
|
Jee HY, Lee YG, Lee S, Elvira R, Seo HE, Lee JY, Han J, Lee K. Activation of ERK and p38 Reduces AZD8055-Mediated Inhibition of Protein Synthesis in Hepatocellular Carcinoma HepG2 Cell Line. Int J Mol Sci 2021; 22:ijms222111824. [PMID: 34769253 PMCID: PMC8584319 DOI: 10.3390/ijms222111824] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2021] [Revised: 10/27/2021] [Accepted: 10/29/2021] [Indexed: 02/07/2023] Open
Abstract
Protein synthesis is important for maintaining cellular homeostasis under various stress responses. In this study, we screened an anticancer drug library to select compounds with translational repression functions. AZD8055, an ATP-competitive mechanistic target of rapamycin complex 1/2 (mTORC1/2) inhibitor, was selected as a translational suppressor. AZD8055 inhibited protein synthesis in mouse embryonic fibroblasts and hepatocellular carcinoma HepG2 cells. Extracellular signal-regulated kinase (ERK) and p38 mitogen-activated protein kinase (MAPK) were activated during the early phase of mTORC1/2 inhibition by AZD8055 treatment. Combined treatment of AZD8055 with the MAPK kinase1/2 (MEK1/2) inhibitor refametinib or the p38 inhibitor SB203580 markedly decreased translation in HepG2 cells. Thus, the inhibition of ERK1/2 or p38 may enhance the efficacy of AZD8055-mediated inhibition of protein synthesis. In addition, AZD8055 down-regulated the phosphorylation of eukaryotic initiation factor 4E-binding protein 1 (4E-BP1), and AZD8055-induced phosphorylation of ERK1/2 and p38 had no effect on phosphorylation status of 4E-BP1. Interestingly, AZD8055 modulated the 4E-BP1 mRNA pool by up-regulating ERK1/2 and p38 pathways. Together, these results suggest that AZD8055-induced activation of MAPKs interferes with inhibition of protein synthesis at an early stage of mTORC1/2 inhibition, and that it may contribute to the development of resistance to mTORC1/2 inhibitors.
Collapse
Affiliation(s)
- Ha-yeon Jee
- Department of Biological Sciences, Konkuk University, Seoul 05029, Korea; (H.-y.J.); (Y.-G.L.); (S.L.); (H.-e.S.); (J.-Y.L.)
| | - Yoon-Gyeong Lee
- Department of Biological Sciences, Konkuk University, Seoul 05029, Korea; (H.-y.J.); (Y.-G.L.); (S.L.); (H.-e.S.); (J.-Y.L.)
| | - Sol Lee
- Department of Biological Sciences, Konkuk University, Seoul 05029, Korea; (H.-y.J.); (Y.-G.L.); (S.L.); (H.-e.S.); (J.-Y.L.)
| | - Rosalie Elvira
- Soonchunhyang Institute of Medi-Bio Science (SIMS), Soonchunhyang University, Cheonan 31151, Korea; (R.E.); (J.H.)
| | - Hye-eun Seo
- Department of Biological Sciences, Konkuk University, Seoul 05029, Korea; (H.-y.J.); (Y.-G.L.); (S.L.); (H.-e.S.); (J.-Y.L.)
| | - Ji-Yeon Lee
- Department of Biological Sciences, Konkuk University, Seoul 05029, Korea; (H.-y.J.); (Y.-G.L.); (S.L.); (H.-e.S.); (J.-Y.L.)
| | - Jaeseok Han
- Soonchunhyang Institute of Medi-Bio Science (SIMS), Soonchunhyang University, Cheonan 31151, Korea; (R.E.); (J.H.)
| | - Kyungho Lee
- Department of Biological Sciences, Konkuk University, Seoul 05029, Korea; (H.-y.J.); (Y.-G.L.); (S.L.); (H.-e.S.); (J.-Y.L.)
- Korea Hemp Institute, Konkuk University, Seoul 05029, Korea
- Correspondence: ; Tel.: +82-2-450-3423; Fax: +82-2-3436-5432
| |
Collapse
|
12
|
Fang S, Sun S, Cai H, Zou X, Wang S, Hao X, Wan X, Tian J, Li Z, He Z, Huang W, Liang C, Zhang Z, Yang L, Tian J, Yu B, Sun B. IRGM/Irgm1 facilitates macrophage apoptosis through ROS generation and MAPK signal transduction: Irgm1 +/- mice display increases atherosclerotic plaque stability. Theranostics 2021; 11:9358-9375. [PMID: 34646375 PMCID: PMC8490524 DOI: 10.7150/thno.62797] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2021] [Accepted: 09/02/2021] [Indexed: 02/04/2023] Open
Abstract
Rationale: Atherosclerosis plaque rupture (PR) is the pathological basis and chief culprit of most acute cardiovascular events and death. Given the complex and important role of macrophage apoptosis and autophagy in affecting plaque stability, an important unanswered question include is whether, and how, immunity-related GTPase family M protein (IRGM) and its mouse orthologue IRGM1 affect macrophage survival and atherosclerotic plaque stability. Methods: To investigate whether serum IRGM of ST-segment elevation myocardial infarction (STEMI) patients is related to plaque morphology, we divided 85 STEMI patients into those with and without plaque rupture (PR and non-PR, respectively) based on OCT image analysis, and quantified the patients' serum IRGM levels. Next, we engineered Irgm1 deficient mice (Irgm1+/-) and chimera mice with Irgm1 deficiency in the bone marrow on an ApoE-/- background, which were then fed a high-fat diet for 16 weeks. Pathological staining was used to detect necrotic plaque cores, ratios of neutral lipids and cholesterol crystal, as well as collagen fiber contents in these mice to characterize plaque stability. In addition, immunofluorescence, immunohistochemical staining and western blot were used to detect the apoptosis of macrophages in the plaques. In vitro, THP-1 and RAW264.7 cells were stimulated with ox-LDL to mimic the in vivo environment, and IRGM/IRGM1 expression were modified by specific siRNA (knockdown) or IRGM plasmid (knocked-in). The effect of IRGM/Irgm1 on autophagy and apoptosis of macrophages induced by ox-LDL was then evaluated. In addition, we introduced inhibitors of the JNK/p38/ERK signaling pathway to verify the specific mechanism by which Irgm1 regulates RAW264.7 cell apoptosis. Results: The serum IRGM levels of PR patients is significantly higher than that of non-PR patients and healthy volunteers, which may be an effective predictor of PR. On a high-fat diet, Irgm1-deficient mice exhibit reduced necrotic plaque cores, as well as neutral lipid and cholesterol crystal ratios, with increased collagen fiber content. Additionally, macrophage apoptosis is inhibited in the plaques of Irgm1-deficient mice. In vitro, IRGM/Irgm1 deficiency rapidly inhibits ox-LDL-induced macrophage autophagy while inhibiting ox-LDL-induced macrophage apoptosis in late stages. Additionally, IRGM/Irgm1 deficiency suppresses reactive oxygen species (ROS) production in macrophages, while removal of ROS effectively inhibits macrophage apoptosis induced by IRGM overexpression. We further show that Irgm1 can affect macrophage apoptosis by regulating JNK/p38/ERK phosphorylation in the MAPK signaling pathway. Conclusions: Serum IRGM may be related to the process of PR in STEMI patients, and IRGM/Irgm1 deficiency increases plaque stability. In addition, IRGM/Irgm1 deficiency suppresses macrophage apoptosis by inhibiting ROS generation and MAPK signaling transduction. Cumulatively, these results suggest that targeting IRGM may represent a new treatment strategy for the prevention and treatment of acute cardiovascular deaths caused by PR.
Collapse
|
13
|
Vandyck HHLD, Hillen LM, Bosisio FM, van den Oord J, zur Hausen A, Winnepenninckx V. Rethinking the biology of metastatic melanoma: a holistic approach. Cancer Metastasis Rev 2021; 40:603-624. [PMID: 33870460 PMCID: PMC8213587 DOI: 10.1007/s10555-021-09960-8] [Citation(s) in RCA: 30] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/15/2021] [Accepted: 03/29/2021] [Indexed: 02/06/2023]
Abstract
Over the past decades, melanoma-related mortality has remained nearly stable. The main reason is treatment failure of metastatic disease and the inherently linked knowledge gap regarding metastasis formation. In order to elicit invasion, melanoma cells manipulate the tumor microenvironment, gain motility, and adhere to the extracellular matrix and cancer-associated fibroblasts. Melanoma cells thereby express different cell adhesion molecules like laminins, integrins, N-cadherin, and others. Epithelial-mesenchymal transition (EMT) is physiological during embryologic development, but reactivated during malignancy. Despite not being truly epithelial, neural crest-derived malignancies like melanoma share similar biological programs that enable tumorigenesis, invasion, and metastasis. This complex phenomenon is termed phenotype switching and is intertwined with oncometabolism as well as dormancy escape. Additionally, it has been shown that primary melanoma shed exosomes that create a favorable premetastatic niche in the microenvironment of secondary organs and lymph nodes. Although the growing body of literature describes the aforementioned concepts separately, an integrative holistic approach is missing. Using melanoma as a tumor model, this review will shed light on these complex biological principles in an attempt to clarify the mechanistic metastatic pathways that dictate tumor and patient fate.
Collapse
Affiliation(s)
- Hendrik HLD Vandyck
- Department of Pathology, GROW-School for Oncology & Developmental Biology, Maastricht University Medical Center, MUMC+, PO Box 5800, 6202 AZ Maastricht, The Netherlands
| | - Lisa M Hillen
- Department of Pathology, GROW-School for Oncology & Developmental Biology, Maastricht University Medical Center, MUMC+, PO Box 5800, 6202 AZ Maastricht, The Netherlands
| | - Francesca M Bosisio
- Laboratory of Translational Cell and Tissue Research (TCTR), Department of Pathology, KU Leuven and UZ Leuven, Leuven, Belgium
| | - Joost van den Oord
- Laboratory of Translational Cell and Tissue Research (TCTR), Department of Pathology, KU Leuven and UZ Leuven, Leuven, Belgium
| | - Axel zur Hausen
- Department of Pathology, GROW-School for Oncology & Developmental Biology, Maastricht University Medical Center, MUMC+, PO Box 5800, 6202 AZ Maastricht, The Netherlands
| | - Véronique Winnepenninckx
- Department of Pathology, GROW-School for Oncology & Developmental Biology, Maastricht University Medical Center, MUMC+, PO Box 5800, 6202 AZ Maastricht, The Netherlands
| |
Collapse
|
14
|
Zhou S, Ouyang W, Zhang X, Liao L, Pi X, Yang R, Mei B, Xu H, Xiang S, Li J. UTRN inhibits melanoma growth by suppressing p38 and JNK/c-Jun signaling pathways. Cancer Cell Int 2021; 21:88. [PMID: 33632212 PMCID: PMC7905598 DOI: 10.1186/s12935-021-01768-4] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2020] [Accepted: 01/10/2021] [Indexed: 01/02/2023] Open
Abstract
Background Utrophin (UTRN), as a tumor suppressor gene, is involved in various cancer progression. The function of UTRN in the melanoma process and the related molecular mechanisms are still unclear. Herein, we studied the function of UTRN in melanoma growth and the relevant molecular mechanisms. Methods Using the GEO database and UCSC Xena project, we compared the expression of UTRN in non-cancerous and melanoma tissues. Immunohistochemistry (IHC) staining, qRT-PCR and Western Blot (WB) were performed to evaluate UTRN expression in clinical samples. A total of 447 cases with UTRN expression data, patient characteristics and survival data were extracted from TCGA database and analyzed. After stable transduction and single cell cloning, the proliferation ability of A375 human melanoma cells was analyzed by Cell Counting Kit‑8 (CCK) and 5‑ethynyl‑2′‑deoxyuridine (EdU) incorporation assays. GSEA was performed to predict the mechanism by which UTRN regulated melanoma growth. Then WB analysis was used to assess the protein expression levels of pathway signaling in overexpression (EXP) melanoma cells. Epac activator 8-pCPT-2′-O-Me-cAMP was then used to evaluate the proliferation ability by activation of p38 and JNK/c-Jun signaling pathways. Results Data from GEO and UCSC Xena project indicated that UTRN expression was decreased in melanoma. Experiment on clinical samples further confirmed our finding. TCGA results showed that a reduced expression of UTRN in 447 melanoma samples was associated with advanced clinical characteristics (T stage, Clark level, ulceration), shorter survival time and poorer prognosis. In addition, up-regulated UTRN expression inhibited melanoma cell proliferation when compared to control group. MAPK signaling pathway was presented in both KEGG and BioCarta databases by using GSEA tool. WB results confirmed the down-regulated expression of p38, JNK1 and c-Jun in EXP group when compared to control group. Epac activator 8-pCPT-2′-O-Me-cAMP treatment could partially rescue proliferation of tumor cells. Conclusion We have demonstrated that reduced UTRN predicted poorer prognosis and UTRN inhibited melanoma growth via p38 and JNK1/c-Jun pathways. Therefore, UTRN could serve as a tumor suppressor and novel prognostic biomarker for melanoma patients.
Collapse
Affiliation(s)
- Sitong Zhou
- Department of Dermatology, The First People's Hospital of Foshan, 81 Lingnan Avenue North, Foshan, 528000, Guangdong, China
| | - Wen Ouyang
- The Second Clinical Medical College, Zhujiang Hospital, Southern Medical University, Guangzhou, Guangdong, China
| | - Xi Zhang
- Department of Dermatology, Binzhou Medical University Hospital, Binzhou, Shandong, China
| | - Lexi Liao
- Department of Dermatology, Cosmetology and Venereology, Shenzhen Hospital, Southern Medical University, Shenzhen, Guangdong, China
| | - Xiaobing Pi
- Department of Dermatology, The First People's Hospital of Foshan, 81 Lingnan Avenue North, Foshan, 528000, Guangdong, China
| | - Ronghua Yang
- Department of Burn Surgery and Skin Regeneration, The First People's Hospital of Foshan, Foshan, Guangdong, China
| | - Baiqiang Mei
- Department of Cardiovascular Disease, The First People's Hospital of Foshan, Foshan, Guangdong, China
| | - Huaiyuan Xu
- Department of Bone and Soft Tissue Surgery, Sun Yat-Sen University Cancer Center, Guangzhou, Guangdong, China
| | - Shijian Xiang
- Department of Pharmacy, Seventh Affiliated Hospital of Sun Yat-Sen University, Shenzhen, 518107, China
| | - Jiehua Li
- Department of Dermatology, The First People's Hospital of Foshan, 81 Lingnan Avenue North, Foshan, 528000, Guangdong, China.
| |
Collapse
|
15
|
Huang XF, Gao HW, Lee SC, Chang KF, Tang LT, Tsai NM. Juniperus indica Bertol. extract synergized with cisplatin against melanoma cells via the suppression of AKT/mTOR and MAPK signaling and induction of cell apoptosis. Int J Med Sci 2021; 18:157-168. [PMID: 33390784 PMCID: PMC7738970 DOI: 10.7150/ijms.49423] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/12/2020] [Accepted: 10/15/2020] [Indexed: 01/05/2023] Open
Abstract
Juniperus indica Bertol. is an herbal plant that belongs to the genus Juniperus, which is commonly used in traditional medicine to refresh the mind and for diuretic use. However, few studies have reported the function of J. indica Bertol. Hence, this study aimed to investigate the anti-tumor and synergistic potential of J. indica Bertol. extract (JIB extract) for melanoma cells. Our results indicated the anti-melanoma activity of JIB extract. JIB extract induced cell cycle arrest at the G0/G1 phase and decreased cyclin and cdk protein expressions. In addition, AKT/mTOR signaling and MAPK signaling were inhibited by JIB extract to suppress melanoma cell growth and proliferation. Additionally, JIB extract induced B16/F10 cell apoptosis via the caspase cascade. According to the JIB extract's anti-melanoma capacity, to assess the synergistic effects of cisplatin and JIB extract. The results demonstrated that JIB extract combined with cisplatin enhanced the inhibition of cell growth, proliferation, and survival through the obstruction of cell cycle progression and AKT/mTOR and MAPK signaling as well as the induction of cell apoptosis. Collectively, our results indicate that JIB extract showed anti-tumor effects and synergized with cisplatin against B16/F10 cells, indicating the possibility of JIB extract to be developed as adjuvant therapy for melanoma.
Collapse
Affiliation(s)
- Xiao-Fan Huang
- Institute of Medicine, Chung Shan Medical University, Taichung, 40201, Taiwan, ROC.,Department of Medical Laboratory and Biotechnology, Chung Shan Medical University, Taichung, 40201, Taiwan, ROC
| | - Hong-Wei Gao
- Department of Pathology, Tri-Service General Hospital, National Defense Medical Center, Taipei, 11490, Taiwan, ROC
| | - Shan-Chih Lee
- Department of Medical Imaging and Radiological Sciences, Chung Shan Medical University, Taichung, 40201, Taiwan, ROC.,Department of Medical Imaging, Chung Shan Medical University Hospital, Taichung, 40201, Taiwan, ROC
| | - Kai-Fu Chang
- Institute of Medicine, Chung Shan Medical University, Taichung, 40201, Taiwan, ROC.,Department of Medical Laboratory and Biotechnology, Chung Shan Medical University, Taichung, 40201, Taiwan, ROC
| | - Li-Ting Tang
- Department of Medical Laboratory and Biotechnology, Chung Shan Medical University, Taichung, 40201, Taiwan, ROC
| | - Nu-Man Tsai
- Department of Medical Laboratory and Biotechnology, Chung Shan Medical University, Taichung, 40201, Taiwan, ROC.,Clinical Laboratory, Chung Shan Medical University Hospital, Taichung, 40201, Taiwan, ROC
| |
Collapse
|
16
|
Banik I, Cheng PF, Dooley CM, Travnickova J, Merteroglu M, Dummer R, Patton EE, Busch-Nentwich EM, Levesque MP. NRAS Q61K melanoma tumor formation is reduced by p38-MAPK14 activation in zebrafish models and NRAS-mutated human melanoma cells. Pigment Cell Melanoma Res 2020; 34:150-162. [PMID: 32910840 DOI: 10.1111/pcmr.12925] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2020] [Revised: 09/02/2020] [Accepted: 09/02/2020] [Indexed: 01/05/2023]
Abstract
Oncogenic BRAF and NRAS mutations drive human melanoma initiation. We used transgenic zebrafish to model NRAS-mutant melanoma, and the rapid tumor onset allowed us to study candidate tumor suppressors. We identified P38α-MAPK14 as a potential tumor suppressor in The Cancer Genome Atlas melanoma cohort of NRAS-mutant melanomas, and overexpression significantly increased the time to tumor onset in transgenic zebrafish with NRAS-driven melanoma. Pharmacological activation of P38α-MAPK14 using anisomycin reduced in vitro viability of melanoma cultures, which we confirmed by stable overexpression of p38α. We observed that the viability of MEK inhibitor resistant melanoma cells could be reduced by combined treatment of anisomycin and MEK inhibition. Our study demonstrates that activating the p38α-MAPK14 pathway in the presence of oncogenic NRAS abrogates melanoma in vitro and in vivo. SIGNIFICANCE: The significance of our study is in the accountability of NRAS mutations in melanoma. We demonstrate here that activation of p38α-MAPK14 pathway can abrogate NRAS-mutant melanoma which is contrary to the previously published role of p38α-MAPK14 pathway in BRAF mutant melanoma. These results implicate that BRAF and NRAS-mutant melanoma may not be identical biologically. We also demonstrate the translational benefit of our study by using a small molecule compound-anisomycin (already in use for other diseases in clinical trials) to activate p38α-MAPK14 pathway.
Collapse
Affiliation(s)
- Ishani Banik
- University Hospital Zurich, University of Zurich, Zurich, Switzerland
| | - Phil F Cheng
- University Hospital Zurich, University of Zurich, Zurich, Switzerland
| | - Christopher M Dooley
- Wellcome Sanger Institute, Hinxton, Cambridge, UK.,Max Planck Institute for Developmental Biology, Tübingen, Germany
| | - Jana Travnickova
- MRC Human Genetics Unit and Cancer Research, UK Edinburgh Centre, MRC Institute of Genetics and Molecular Medicine, University of Edinburgh, Western General Hospital, Edinburgh, UK
| | - Munise Merteroglu
- Wellcome Sanger Institute, Hinxton, Cambridge, UK.,Cambridge Institute of Therapeutic Immunology & Infectious Disease (CITIID), Department of Medicine, University of Cambridge, Cambridge, UK
| | - Reinhard Dummer
- University Hospital Zurich, University of Zurich, Zurich, Switzerland
| | - Elizabeth E Patton
- MRC Human Genetics Unit and Cancer Research, UK Edinburgh Centre, MRC Institute of Genetics and Molecular Medicine, University of Edinburgh, Western General Hospital, Edinburgh, UK
| | - Elisabeth M Busch-Nentwich
- Wellcome Sanger Institute, Hinxton, Cambridge, UK.,Cambridge Institute of Therapeutic Immunology & Infectious Disease (CITIID), Department of Medicine, University of Cambridge, Cambridge, UK
| | | |
Collapse
|
17
|
Obrador E, Salvador R, López-Blanch R, Jihad-Jebbar A, Alcácer J, Benlloch M, Pellicer JA, Estrela JM. Melanoma in the liver: Oxidative stress and the mechanisms of metastatic cell survival. Semin Cancer Biol 2020; 71:109-121. [PMID: 32428715 DOI: 10.1016/j.semcancer.2020.05.001] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2020] [Revised: 05/03/2020] [Accepted: 05/03/2020] [Indexed: 12/16/2022]
Abstract
Metastatic melanoma is a fatal disease with a rapid systemic dissemination. The most frequent target sites are the liver, bone, and brain. Melanoma metastases represent a heterogeneous cell population, which associates with genomic instability and resistance to therapy. Interaction of melanoma cells with the hepatic sinusoidal endothelium initiates a signaling cascade involving cytokines, growth factors, bioactive lipids, and reactive oxygen and nitrogen species produced by the cancer cell, the endothelium, and also by different immune cells. Endothelial cell-derived NO and H2O2 and the action of immune cells cause the death of most melanoma cells that reach the hepatic microvascularization. Surviving melanoma cells attached to the endothelium of pre-capillary arterioles or sinusoids may follow two mechanisms of extravasation: a) migration through vessel fenestrae or b) intravascular proliferation followed by vessel rupture and microinflammation. Invading melanoma cells first form micrometastases within the normal lobular hepatic architecture via a mechanism regulated by cross-talk with the stroma and multiple microenvironment-related molecular signals. In this review special emphasis is placed on neuroendocrine (systemic) mechanisms as potential promoters of liver metastatic growth. Growing metastatic cells undergo functional and metabolic changes that increase their capacity to withstand oxidative/nitrosative stress, which favors their survival. This adaptive process also involves upregulation of Bcl-2-related antideath mechanisms, which seems to lead to the generation of more resistant cell subclones.
Collapse
Affiliation(s)
- Elena Obrador
- Department of Physiology, University of Valencia, 46010, Valencia, Spain
| | - Rosario Salvador
- Department of Physiology, University of Valencia, 46010, Valencia, Spain
| | | | - Ali Jihad-Jebbar
- Department of Physiology, University of Valencia, 46010, Valencia, Spain
| | - Javier Alcácer
- Pathology Laboratory, Quirón Hospital, 46010, Valencia, Spain
| | - María Benlloch
- Department of Health & Functional Valorization, San Vicente Martir Catholic University, 46001, Valencia, Spain
| | - José A Pellicer
- Department of Physiology, University of Valencia, 46010, Valencia, Spain
| | - José M Estrela
- Department of Physiology, University of Valencia, 46010, Valencia, Spain.
| |
Collapse
|
18
|
P38 MAPK Promotes Migration and Metastatic Activity of BRAF Mutant Melanoma Cells by Inducing Degradation of PMCA4b. Cells 2020; 9:cells9051209. [PMID: 32414111 PMCID: PMC7290426 DOI: 10.3390/cells9051209] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2020] [Revised: 05/10/2020] [Accepted: 05/11/2020] [Indexed: 02/07/2023] Open
Abstract
Metastatic melanoma is the most aggressive type of skin cancer. Previously, we identified the plasma membrane Ca2+ pump isoform 4b (PMCA4b or ATP2B4) as a putative metastasis suppressor in BRAF mutant melanoma cells. Metastasis suppressors are often downregulated in cancer, therefore, it is important to identify the pathways involved in their degradation. Here, we studied the role of p38 MAPK in PMCA4b degradation and its effect on melanoma metastasis. We found that activation of p38 MAPK induces internalization and subsequent degradation of PMCA4b through the endo/lysosomal system that contributes to the low PMCA4b steady-state protein level of BRAF mutant melanoma cells. Moreover, BRAF wild type cell models including a doxycycline-inducible HEK cell system revealed that p38 MAPK is a universal modulator of PMCA4b endocytosis. Inhibition of the p38 MAPK pathway markedly reduced migration, colony formation and metastatic activity of BRAF mutant cells in vitro partially through an increase in PMCA4b and a decrease in β4 integrin abundance. In conclusion, our data suggest that the p38 MAPK pathway plays a key role in PMCA4b degradation and inhibition of this pathway—by increasing the stability of PMCA4b—may provide a potential therapeutic target for inhibition of melanoma progression and metastasis.
Collapse
|
19
|
Duval KEA, Vernice NA, Wagner RJ, Fiering SN, Petryk JD, Lowry GJ, Tau SS, Yin J, Houde GR, Chaudhry AS, Hoopes PJ. Immunogenetic effects of low dose (CEM43 30) magnetic nanoparticle hyperthermia and radiation in melanoma cells. Int J Hyperthermia 2020; 36:37-46. [PMID: 31795829 PMCID: PMC6943912 DOI: 10.1080/02656736.2019.1627433] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023] Open
Abstract
Objective: In this in vitro study we have used an RNA quantification technique, nanoString, and a conventional protein analysis technique (Western Blot) to assess the genetic and protein expression of B16 murine melanoma cells following a modest magnetic nanoparticle hyperthermia (mNPH) dose equivalent to 30 minutes @ 43°C (CEM43 30) and/or a clinically relevant 8 Gy radiation dose. Methods: Melanoma cells with mNPs(2.5 μg Fe/106 cells) were pelleted and exposed to an alternating magnetic field (AMF) to generate the targeted thermal dose. Thermal dose was accurately monitored by a fiber optic probe and automatically maintained at CEM43 30. All cells were harvested 24 hours after treatment. Results: The mNPH dose demonstrated notable elevations in the thermotolerance/immunogenic HSP70 gene and a number of chemoattractant and toll-like receptor gene pathways. The 8 Gy dose also upregulated a number of important immune and cytotoxic genetic and protein pathways. However, the mNPH/radiation combination was the most effective stimulator of a wide variety of immune and cytotoxic genes including HSP70, cancer regulating chemokines CXCL10, CXCL11, the T-cell trafficking chemokine CXCR3, innate immune activators TLR3, TLR4, the MDM2 and mTOR negative regulator of p53, the pro-apoptotic protein PUMA, and the cell death receptor Fas. Importantly a number of the genetic changes were accurately validated by protein expression changes, i.e., HSP70, p-mTOR, p-MDM2. Conclusion: These results not only show that low dose mNPH and radiation independently increase the expression of important immune and cytotoxic genes but that the effect is greatly enhanced when they are used in combination.
Collapse
Affiliation(s)
- Kayla E A Duval
- Thayer School of Engineering, Dartmouth College, Hanover, NH, USA
| | | | - Robert J Wagner
- Geisel School of Medicine, Dartmouth College, Hanover, NH, USA
| | | | - James D Petryk
- Geisel School of Medicine, Dartmouth College, Hanover, NH, USA
| | | | - Steven S Tau
- Geisel School of Medicine, Dartmouth College, Hanover, NH, USA
| | - John Yin
- Geisel School of Medicine, Dartmouth College, Hanover, NH, USA
| | - Georgia R Houde
- Geisel School of Medicine, Dartmouth College, Hanover, NH, USA
| | | | - P Jack Hoopes
- Thayer School of Engineering, Dartmouth College, Hanover, NH, USA.,Geisel School of Medicine, Dartmouth College, Hanover, NH, USA
| |
Collapse
|
20
|
Wenzina J, Holzner S, Puujalka E, Cheng PF, Forsthuber A, Neumüller K, Schossleitner K, Lichtenberger BM, Levesque MP, Petzelbauer P. Inhibition of p38/MK2 Signaling Prevents Vascular Invasion of Melanoma. J Invest Dermatol 2020; 140:878-890.e5. [DOI: 10.1016/j.jid.2019.08.451] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2019] [Revised: 07/29/2019] [Accepted: 08/22/2019] [Indexed: 12/20/2022]
|
21
|
Li J, Jia Y, An L, Niu C, Cong X, Zhao Y. Uncoupling protein 2 is upregulated in melanoma cells and contributes to the activation of Akt/mTOR and ERK signaling. Int J Oncol 2020; 56:1252-1261. [PMID: 32319575 DOI: 10.3892/ijo.2020.5010] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2019] [Accepted: 12/12/2019] [Indexed: 11/05/2022] Open
Abstract
The aim of the present study was to characterize the expression of uncoupling protein 2 (UCP2) in melanoma and to study the potential mechanisms underlying the involvement of UCP2 in melanomagenesis using human melanoma cell lines. The expression of UCP2 was evaluated in specimens from normal control subjects, patients with compound nevus, and patients with cutaneous and mucosal melanoma. Stable knockdown of UCP2 was achieved in human melanoma cell lines, which were used to examine whether UCP2 knockdown affects the mitochondrial membrane potential and intracellular levels of ATP, reactive oxygen species and lactate. Cell proliferation, invasion, spheroid formation and cisplatin sensitivity were also evaluated in the UCP2 knockdown cells. Finally, the effects of UCP2 knockdown on the Akt/mammalian target of rapamycin (mTOR) and extracellular signal‑regulated kinase (ERK) pathways, which are important oncogenic pathways during melanomagenesis, were evaluated. Relatively high expression of UCP2 was detected in human melanoma specimens, which was correlated with Clark level and Breslow thickness. Knockdown of UCP2 suppressed cell proliferation, invasion and spheroid formation, and increased the sensitivity of melanoma cells to cisplatin. Furthermore, the UCP2 knockdown cells exhibited inhibition of Akt/mTOR signaling and ERK activation. Therefore, human melanoma tissues exhibit relatively high UCP2 expression, which may be implicated in the mechanisms underlying tumor progression. The potential role of UCP2 in melanomagenesis may involve enhancing the Akt/mTOR and mitogen‑activated protein kinase/ERK pathways.
Collapse
Affiliation(s)
- Jinran Li
- Department of Dermatology, China‑Japan Union Hospital, Jilin University, Changchun, Jilin 130033, P.R. China
| | - Yuxi Jia
- Department of Dermatology, China‑Japan Union Hospital, Jilin University, Changchun, Jilin 130033, P.R. China
| | - Lin An
- Department of Dermatology, China‑Japan Union Hospital, Jilin University, Changchun, Jilin 130033, P.R. China
| | - Chunbo Niu
- Department of Pathology, China‑Japan Union Hospital, Jilin University, Changchun, Jilin 130033, P.R. China
| | - Xianling Cong
- Department of Dermatology, China‑Japan Union Hospital, Jilin University, Changchun, Jilin 130033, P.R. China
| | - Yunfeng Zhao
- Department of Pharmacology, Toxicology and Neurosciences, LSU Health Sciences Center, Shreveport, LA 71130, USA
| |
Collapse
|
22
|
Donohoe F, Wilkinson M, Baxter E, Brennan DJ. Mitogen-Activated Protein Kinase (MAPK) and Obesity-Related Cancer. Int J Mol Sci 2020; 21:ijms21041241. [PMID: 32069845 PMCID: PMC7072904 DOI: 10.3390/ijms21041241] [Citation(s) in RCA: 44] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2019] [Revised: 02/06/2020] [Accepted: 02/12/2020] [Indexed: 12/13/2022] Open
Abstract
Obesity is a major public health concern worldwide. The increased risk of certain types of cancer is now an established deleterious consequence of obesity, although the molecular mechanisms of this are not completely understood. In this review, we aim to explore the links between MAPK signalling and obesity-related cancer. We focus mostly on p38 and JNK MAPK, as the role of ERK remains unclear. These links are seen through the implication of MAPK in obesity-related immune paralysis as well as through effects on the endoplasmic reticulum stress response and activation of aromatase. By way of example, we highlight areas of interest and possibilities for future research in endometrioid endometrial cancer and hepatocellular carcinoma associated with non-alcoholic fatty liver disease (NAFLD), non-alcoholic steatohepatitis (NASH) and MAPK.
Collapse
Affiliation(s)
- Fionán Donohoe
- Ireland East Hospital Gynaeoncology Group, UCD School of Medicine, Mater Misericordiae University, D07R2WY Dublin 7, Ireland; (F.D.); (M.W.)
| | - Michael Wilkinson
- Ireland East Hospital Gynaeoncology Group, UCD School of Medicine, Mater Misericordiae University, D07R2WY Dublin 7, Ireland; (F.D.); (M.W.)
| | - Eva Baxter
- Queensland Centre for Gynaecological Cancer Research, The University of Queensland, Brisbane QLD 4029, Australia;
| | - Donal J. Brennan
- Ireland East Hospital Gynaeoncology Group, UCD School of Medicine, Mater Misericordiae University, D07R2WY Dublin 7, Ireland; (F.D.); (M.W.)
- Systems Biology Ireland, UCD School of Medicine, Belfield, D04V1W8 Dublin 4, Ireland
- Correspondence: ; Tel.: +353-1-7164567
| |
Collapse
|
23
|
Ko G, Kim T, Ko E, Park D, Lee Y. Synergistic Enhancement of Paclitaxel-induced Inhibition of Cell Growth by Metformin in Melanoma Cells. Dev Reprod 2019; 23:119-128. [PMID: 31321352 PMCID: PMC6635613 DOI: 10.12717/dr.2019.23.2.119] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2019] [Revised: 05/27/2019] [Accepted: 06/08/2019] [Indexed: 11/25/2022]
Abstract
Melanoma is one of the most aggressive and treatment-resistant malignancies.
Antidiabetic drug metformin has been reported to inhibit cell proliferation and
metastasis in many cancers, including melanoma. Metformin suppresses the
mammalian target of rapamycin (mTOR) and our previous study showed that it also
inhibits the activity of extracellular signal-regulated kinase (ERK). Paclitaxel
is currently prescribed for treatment of melanoma. However, paclitaxel induced
the activation of ERK/mitogen-activated protein kinase (MAPK) pathway, a cell
signaling pathway implicated in cell survival and proliferation. Therefore, we
reasoned that combined treatment of paclitaxel with metformin could be more
effective in the suppression of cell proliferation than treatment of paclitaxel
alone. Here, we investigated the combinatory effect of paclitaxel and metformin
on the cell survival in SK-MEL-28 melanoma cell line. Our study shows that the
combination of paclitaxel and metformin has synergistic effect on cell survival
and suppresses the expression of proteins involved in cancer metastasis. These
findings suggest that the combination of paclitaxel and metformin can be a
possible therapeutic option for treatment of melanoma.
Collapse
Affiliation(s)
- Gihyun Ko
- Dept. of Medicine, Jeju National University School of Medicine, Jeju 63243, Korea
| | - Taehyung Kim
- Dept. of Medicine, Jeju National University School of Medicine, Jeju 63243, Korea
| | - Eunjeong Ko
- Dept. of Medicine, Jeju National University School of Medicine, Jeju 63243, Korea
| | - Deokbae Park
- Histology, Jeju National University School of Medicine, Jeju 63243, Korea
| | - Youngki Lee
- Histology, Jeju National University School of Medicine, Jeju 63243, Korea
| |
Collapse
|
24
|
Sustained ERK activation-mediated proliferation inhibition of farrerol on human gastric carcinoma cell line by G0/G1-phase cell-cycle arrest. Eur J Cancer Prev 2018; 25:490-9. [PMID: 26656929 DOI: 10.1097/cej.0000000000000212] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/22/2022]
Abstract
Current cancer treatment is partly limited by chemotherapy-induced vascular toxicity associated with damage to vascular endothelial cells. In this study, the cytotoxicity of farrerol against SGC7901 gastric cancer cells and human umbilical vein endothelial cells (HUVECs) in vitro was investigated along with the underlying mechanisms of its growth-inhibitory effect against SGC7901 cells. MTT assays showed that farrerol inhibited SGC7901 cell growth, but exerted no cytotoxicity against HUVECs. Flow cytometry showed that treatment of SGC7901 cells with farrerol (5, 40, or 160 μmol/l) for 24 h caused G0/G1 cell cycle arrest in a concentration-dependent manner. Western blotting indicated that exposure of SGC7901 cells to farrerol resulted in significant upregulation of p27KIP1 (p27), accompanied by sustained activation of ERK1/2 and p38 MAPK instead of JNK. Farrerol-stimulated p27 expression, p38 MAPK activation, and cell growth inhibition were attenuated by pretreatment with U0126, an MEK1/2 inhibitor. In conclusion, this study indicates the selective cytotoxicity of farrerol against SGC7901 cells, but not HUVECs. Furthermore, it provides the first evidence that farrerol could induce cancer cell growth inhibition by G0/G1-phase cell-cycle arrest mediated by sustained ERK activation. The findings show the potential of farrerol as a chemotherapeutic agent without vascular toxicity for use against gastric cancer.
Collapse
|
25
|
Macrovipecetin, a C-type lectin from Macrovipera lebetina venom, inhibits proliferation migration and invasion of SK-MEL-28 human melanoma cells and enhances their sensitivity to cisplatin. Biochim Biophys Acta Gen Subj 2017; 1862:600-614. [PMID: 29196192 DOI: 10.1016/j.bbagen.2017.11.019] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2017] [Revised: 10/05/2017] [Accepted: 11/27/2017] [Indexed: 12/29/2022]
Abstract
BACKGROUND The resistance of melanoma cells to cisplatin restricts its clinical use. Therefore, the search for novel tumor inhibitors and effective combination treatments that sensitize tumor cells to this drug are still needed. We purified macrovipecetin, a novel heterodimeric C-type lectin, from Macrovipera lebetina snake venom and investigated its anti-tumoral effect on its own or combined with cisplatin, in human melanoma cells. METHODS Biochemical characterization, in vitro cells assays such as viability, apoptosis, adhesion, migration, invasion, Western blotting and in silico analysis were used in this study. RESULTS Macrovipecetin decreased melanoma cell viability 100 times more than cisplatin. Interestingly, when combined with the drug, macrovipecetin enhanced the sensitivity of SK-MEL-28 cells by augmenting their apoptosis through increased expression of the apoptosis inducing factor (AIF) and activation of ERK1/2, p38, AKT and NF-κB. Moreover, macrovipecetin alone or combined with cisplatin induced the expression of TRADD, p53, Bax, Bim and Bad and down-regulated the Bcl-2 expression and ROS levels in SK-MEL-28 cells. Interestingly, these treatments impaired SK-MEL-28 cell adhesion, migration and invasion through modulating the function and expression of αvβ3 integrin along with regulating E-cadherin, vimentin, β-catenin, c-Src and RhoA expression. In silico study suggested that only the α chain of macrovipecetin interacts with a region overlapping the RGD motif binding site on this integrin. CONCLUSIONS We validated the antitumor effect of macrovipecetin when combined, or not, with cisplatin on SK-MEL-28 cells. GENERAL SIGNIFICANCE The presented work proposes the potential use of macrovipecetin and cisplatin in combination as an effective anti-melanoma treatment.
Collapse
|
26
|
The crosstalk between p38 and Akt signaling pathways orchestrates EMT by regulating SATB2 expression in NSCLC cells. Tumour Biol 2017; 39:1010428317706212. [DOI: 10.1177/1010428317706212] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
Epithelial–mesenchymal transition is a crucial event for metastasis and could be mediated by several pathways such as phosphoinositide 3-kinase/Akt, mitogen-activated protein kinases, as well as many epigenetic regulators. Special AT-rich sequence-binding protein 2 is an epigenetic regulator involved in epithelial–mesenchymal transition and osteoblastic differentiation. It has been reported that the crosstalk between several pathways is responsible for the regulation of epithelial–mesenchymal transition in cancer cells. However, crosstalks between p38 and Akt pathways involved in epithelial–mesenchymal transition are still unknown. We recently reported that there is a crosstalk between p38 and Akt pathways in non-small-cell lung carcinoma cells, and this crosstalk is associated with E-cadherin and special AT-rich sequence-binding protein 2 expressions. Therefore, we aimed to determine whether this crosstalk has a mediator role in the regulation of epithelial–mesenchymal transition in non-small-cell lung carcinoma. Our results showed that inhibition of p38 leads to the disruption of this crosstalk via decreased expression of phosphatase and tensin homolog (PTEN) and subsequently increased activation of Akt in non-small-cell lung carcinoma cells. Then, we found that p38 inhibition upregulated special AT-rich sequence-binding protein 2 expression and reversed epithelial–mesenchymal transition in non-small-cell lung carcinoma cells. Furthermore, special AT-rich sequence-binding protein 2 knockdown abolished the effect of p38 inhibition on epithelial–mesenchymal transition in non-small-cell lung carcinoma cells. In conclusion, our results strongly indicate that the crosstalk between p38 and Akt pathways can determine special AT-rich sequence-binding protein 2 expression and epithelial character of non-small-cell lung carcinoma cells, and special AT-rich sequence-binding protein 2 is a critical epigenetic regulator for epithelial–mesenchymal transition mediated by p38 pathway in non-small-cell lung carcinoma. Our findings will contribute to illuminate the molecular mechanisms of the epithelial–mesenchymal transition process that has a critical significance for lung cancer metastasis.
Collapse
|
27
|
Moriwaki K, Asahi M. Augmented TME O-GlcNAcylation Promotes Tumor Proliferation through the Inhibition of p38 MAPK. Mol Cancer Res 2017; 15:1287-1298. [PMID: 28536142 DOI: 10.1158/1541-7786.mcr-16-0499] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2016] [Revised: 03/13/2017] [Accepted: 05/18/2017] [Indexed: 11/16/2022]
Abstract
O-GlcNAcylation is a dynamic O-linked glycosylation event that plays a crucial role in regulating cellular signaling. Recent studies indicate that increased O-GlcNAcylation is a general feature in cancer and contributes to various cancer phenotypes, including cell proliferation, survival, invasion, metastasis, and energy metabolism. However, the role of O-GlcNAcylation in the tumor microenvironment (TME) is not fully elucidated. Here, B16 melanoma cells were subcutaneously transplanted into O-GlcNAc transferase transgenic (Ogt-Tg) mice exhibiting elevated O-GlcNAcylation to examine the effect of O-GlcNAcylation in the TME on tumor progression. In this model system, B16 tumor growth was significantly higher in Ogt-Tg/+ mice compared with wild-type (WT) mice. The tumors grown in Ogt-Tg/+ mice showed significant downregulation of p38 MAPK activity and upregulation of the ERK1/2 signaling pathway. In addition, proinflammatory cytokine production was significantly lower in the tumor tissues from Ogt-Tg/+ mice than in those from WT mice. Activation of NF-κB, a key regulator in the cytokine production, was downregulated in the macrophages of the tumor tissues grown in Ogt-Tg/+ mice. These data reveal that elevated O-GlcNAcylation in the TME reduces the production of inflammatory cytokines and promotes cancer progression through downregulation of p38 MAPK activity and subsequent upregulation of the ERK1/2 signaling pathway.Implications: The reduced production of inflammatory cytokines by augmented O-GlcNAcylation in the TME, mainly macrophages, promotes tumor proliferation through the inhibition of p38 MAPK and suggests a possible cause of increased morbidity and mortality rates for various cancers in diabetic patients. Mol Cancer Res; 15(9); 1287-98. ©2017 AACR.
Collapse
Affiliation(s)
- Kazumasa Moriwaki
- Department of Pharmacology, Faculty of Medicine, Osaka Medical College, Takatsuki, Osaka, Japan
| | - Michio Asahi
- Department of Pharmacology, Faculty of Medicine, Osaka Medical College, Takatsuki, Osaka, Japan.
| |
Collapse
|
28
|
Fong Y, Wu CY, Chang KF, Chen BH, Chou WJ, Tseng CH, Chen YC, Wang HMD, Chen YL, Chiu CC. Dual roles of extracellular signal-regulated kinase (ERK) in quinoline compound BPIQ-induced apoptosis and anti-migration of human non-small cell lung cancer cells. Cancer Cell Int 2017; 17:37. [PMID: 28286419 PMCID: PMC5339964 DOI: 10.1186/s12935-017-0403-0] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2016] [Accepted: 02/16/2017] [Indexed: 02/06/2023] Open
Abstract
Background 2,9-Bis[2-(pyrrolidin-1-yl)ethoxy]-6-{4-[2-(pyrrolidin-1-yl)ethoxy] phenyl}-11H-indeno[1,2-c]quinoline-11-one (BPIQ), is a synthetic quinoline analog. A previous study showed the anti-cancer potential of BPIQ through modulating mitochondrial-mediated apoptosis. However, the effect of BPIQ on cell migration, an index of cancer metastasis, has not yet been examined. Furthermore, among signal pathways involved in stresses, the members of the mitogen-activated protein kinase (MAPK) family are crucial for regulating the survival and migration of cells. In this study, the aim was to explore further the role of MAPK members, including JNK, p38 and extracellular signal-regulated kinase (ERK) in BPIQ-induced apoptosis and anti-migration of human non-small cell lung cancer (NSCLC) cells. Methods Western Blot assay was performed for detecting the activation of MAPK members in NSCLC H1299 cells following BPIQ administration. Cellular proliferation was determined using a trypan blue exclusion assay. Cellular apoptosis was detected using flow cytometer-based Annexin V/propidium iodide dual staining. Cellular migration was determined using wound-healing assay and Boyden’s chamber assay. Zymography assay was performed for examining MMP-2 and -9 activities. The assessment of MAPK inhibition was performed for further validating the role of JNK, p38, and ERK in BPIQ-induced growth inhibition, apoptosis, and migration of NSCLC cells. Results Western Blot assay showed that BPIQ treatment upregulates the phosphorylated levels of both MAPK proteins JNK and ERK. However, only ERK inhibitor rescues BPIQ-induced growth inhibition of NSCLC H1299 cells. The results of Annexin V assay further confirmed the pro-apoptotic role of ERK in BPIQ-induced cell death of H1299 cells. The results of wound healing and Boyden chamber assays showed that sub-IC50 (sub-lethal) concentrations of BPIQ cause a significant inhibition of migration in H1299 cells accompanied with downregulating the activity of MMP-2 and -9, the motility index of cancer cells. Inhibition of ERK significantly enhances BPIQ-induced anti-migration of H1299 cells. Conclusions Our results suggest ERK may play dual roles in BPIQ-induced apoptosis and anti-migration, and it would be worthwhile further developing strategies for treating chemoresistant lung cancers through modulating ERK activity. Electronic supplementary material The online version of this article (doi:10.1186/s12935-017-0403-0) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Yao Fong
- Department of Thoracic Surgery, Chi-Mei Medical Center, Tainan, 710 Taiwan
| | - Chang-Yi Wu
- Department of Biological Sciences, National Sun Yat-sen University, Kaohsiung, 804 Taiwan.,Department of Biotechnology, Kaohsiung Medical University, Kaohsiung, 807 Taiwan
| | - Kuo-Feng Chang
- Department of Biotechnology, Kaohsiung Medical University, Kaohsiung, 807 Taiwan
| | - Bing-Hung Chen
- Department of Biotechnology, Kaohsiung Medical University, Kaohsiung, 807 Taiwan.,The Institute of Biomedical Sciences, National Sun Yat-Sen University, Kaohsiung, 804 Taiwan
| | - Wan-Ju Chou
- Department of Biotechnology, Kaohsiung Medical University, Kaohsiung, 807 Taiwan
| | - Chih-Hua Tseng
- School of Pharmacy, Kaohsiung Medical University, Kaohsiung, 807 Taiwan
| | - Yen-Chun Chen
- Department of Biotechnology, Kaohsiung Medical University, Kaohsiung, 807 Taiwan
| | - Hui-Min David Wang
- Graduate Institute of Biomedical Engineering, National Chung Hsing University, Taichung, 402 Taiwan
| | - Yeh-Long Chen
- Department of Medicinal and Applied Chemistry, Kaohsiung Medical University, Kaohsiung, 807 Taiwan
| | - Chien-Chih Chiu
- Department of Biological Sciences, National Sun Yat-sen University, Kaohsiung, 804 Taiwan.,Department of Biotechnology, Kaohsiung Medical University, Kaohsiung, 807 Taiwan.,Translational Research Center, Cancer Center and Department of Medical Research, Kaohsiung Medical University Hospital, Kaohsiung, 807 Taiwan.,Research Center for Environment Medicine, Kaohsiung Medical University, Kaohsiung, 807 Taiwan.,Graduate Institute of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung, 807 Taiwan
| |
Collapse
|
29
|
Berzaghi R, Maia VSC, Pereira FV, Melo FM, Guedes MS, Origassa CST, Scutti JB, Matsuo AL, Câmara NOS, Rodrigues EG, Travassos LR. SOCS1 favors the epithelial-mesenchymal transition in melanoma, promotes tumor progression and prevents antitumor immunity by PD-L1 expression. Sci Rep 2017; 7:40585. [PMID: 28079159 PMCID: PMC5227698 DOI: 10.1038/srep40585] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2016] [Accepted: 12/02/2016] [Indexed: 01/05/2023] Open
Abstract
Silencing of SOCS1 protein with shRNAi lentivirus (shR-SOCS1) led to partial reversion of the tumorigenic phenotype of B16F10-Nex2 melanoma cells. SOCS1 silencing inhibited cell migration and invasion as well as in vitro growth by cell cycle arrest at S phase with increased cell size and nuclei. Down-regulation of SOCS1 decreased the expression of epidermal growth factor receptor, Ins-Rα, and fibroblast growth factor receptors. The present work aimed at analyzing the SOCS1 cell signaling and expression of proteins relevant to tumor development. An RNA microarray analysis of B16F10-Nex2 melanoma cells with SOCS1 silenced by shRNAi-SOCS1 was undertaken in comparison with cells transduced with the empty vector. Among 609 differentially expressed genes, c-Kit, Met and EphA3 cytokine/tyrosine-kinase (TK) receptors were down regulated. A significant decrease in the expression of TK receptors, the phosphorylation of mediators of ERK1/2 and p38 pathways and STAT3 (S727) were observed. Subcutaneous immunization with shR-SOCS1-transduced viable tumor cells rendered protection against melanoma in a syngeneic model, with decreased expression of PD-L1 and of matrix metallo-proteinases (MMPs) and CD-10 in those cells. The present work shows the role of SOCS1 in murine melanoma development and the potential of SOCS1-silenced tumor cells in raising an effective anti-melanoma immune response.
Collapse
Affiliation(s)
- R. Berzaghi
- Experimental Oncology Unit, Department of Microbiology, Immunology and Parasitology, University of São Paulo, São Paulo, Brazil
| | | | - F. V. Pereira
- Laboratory of Cancer Immunobiology, University of São Paulo, São Paulo, Brazil
| | - F. M. Melo
- Immunology Department, Federal University of São Paulo, São Paulo, Brazil
| | - M. S. Guedes
- Experimental Oncology Unit, Department of Microbiology, Immunology and Parasitology, University of São Paulo, São Paulo, Brazil
| | - C. S. T. Origassa
- Laboratory of Cancer Immunobiology, University of São Paulo, São Paulo, Brazil
| | - J. B. Scutti
- Immunotherapy Platform, Department of Immunology, MD Anderson Cancer Center, Houston Texas, USA
| | - A. L. Matsuo
- Interdepartmental Group of Health Economics (Grides), Federal University of São Paulo, SP, Brazil
| | - N. O. S. Câmara
- Immunology Department, Biomedical Sciences Institute IV, University of São Paulo, São Paulo, Brazil
| | - E. G. Rodrigues
- Laboratory of Cancer Immunobiology, University of São Paulo, São Paulo, Brazil
| | - L. R. Travassos
- Experimental Oncology Unit, Department of Microbiology, Immunology and Parasitology, University of São Paulo, São Paulo, Brazil
- Recepta Biopharma São Paulo, Brazil
| |
Collapse
|
30
|
Li Y, Liu L, Yin Z, Xu H, Li S, Tao W, Cheng H, Du L, Zhou X, Zhang B. Effect of targeted silencing of IL-8 on in vitro migration and invasion of SKOV3 ovarian cancer cells. Oncol Lett 2016; 13:567-572. [PMID: 28356930 PMCID: PMC5351404 DOI: 10.3892/ol.2016.5511] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2016] [Accepted: 05/13/2016] [Indexed: 12/14/2022] Open
Abstract
The aim of the study was to determine whether interleukin-8 (IL-8) affects human SKOV3 ovarian cancer cell migration and invasion by targeting silencing of IL-8 expression. Silencing small-interfering RNA (siRNA) targeting IL-8 gene was constructed to infect SKOV3 cells by lentiviral vector. The expression of IL-8 and p-nuclear factor (NF)-κB protein was detected by western blot analysis. The wound scratch and Transwell tests were used to assay the cell migration and invasiveness of SKOV3 cells infected with lentiviral vector targeting IL-8 gene siRNA. The levels of IL-8 protein expressed by SKOV3 cells infected by lentiviral vector targeting IL-8 gene siRNA decreased by 72.3%. IL-8 (50 ng/ml) increased the ability of SKOV3 cells to suppress cell migration (p<0.01). Cisplatin and silencing of IL-8 achieved the ability to inhibit SKOV3 cell invasion (p<0.01), and 100 ng/ml concentration of IL-8 enhanced the ability of SKOV3 invasion (p<0.01). Silencing of IL-8 to a certain extent reduced the expression of p-NF-κB proteins, but it was not statistically significant. In conclusion, silencing of IL-8 may inhibit the migration and invasion of SKOV3 cells, which may be independent of the p-NF-κB protein.
Collapse
Affiliation(s)
- Yanyu Li
- Department of Gynaecology and Obstetrics, Xuzhou Central Hospital, Xuzhou, Jiangsu 221009, P.R. China; Xuzhou Clinical School, Xuzhou Medical College, Xuzhou, Jiangsu 221000, P.R. China
| | - Ling Liu
- Department of Pharmacology, Xuzhou Central Hospital, Xuzhou, Jiangsu 221009, P.R. China
| | - Zeyuan Yin
- Xuzhou Clinical School, Xuzhou Medical College, Xuzhou, Jiangsu 221000, P.R. China
| | - Hui Xu
- Department of Gynaecology and Obstetrics, Xuzhou Central Hospital, Xuzhou, Jiangsu 221009, P.R. China
| | - Shuang Li
- Department of Gynaecology and Obstetrics, Xuzhou Central Hospital, Xuzhou, Jiangsu 221009, P.R. China
| | - Wei Tao
- Department of Gynaecology and Obstetrics, Xuzhou Central Hospital, Xuzhou, Jiangsu 221009, P.R. China
| | - Hui Cheng
- Department of Gynaecology and Obstetrics, Xuzhou Central Hospital, Xuzhou, Jiangsu 221009, P.R. China
| | - Lei Du
- Jiangsu Key Laboratory of New Drug Research and Clinical Pharmacy, Xuzhou Medical College, Xuzhou, Jiangsu 221000, P.R. China
| | - Xueyuan Zhou
- Jiangsu Key Laboratory of New Drug Research and Clinical Pharmacy, Xuzhou Medical College, Xuzhou, Jiangsu 221000, P.R. China
| | - Bei Zhang
- Department of Gynaecology and Obstetrics, Xuzhou Central Hospital, Xuzhou, Jiangsu 221009, P.R. China; Xuzhou Clinical School, Xuzhou Medical College, Xuzhou, Jiangsu 221000, P.R. China
| |
Collapse
|
31
|
Domingue JC, Ao M, Sarathy J, Rao MC. Chenodeoxycholic acid requires activation of EGFR, EPAC, and Ca2+ to stimulate CFTR-dependent Cl- secretion in human colonic T84 cells. Am J Physiol Cell Physiol 2016; 311:C777-C792. [PMID: 27558159 DOI: 10.1152/ajpcell.00168.2016] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2016] [Accepted: 08/19/2016] [Indexed: 12/12/2022]
Abstract
Bile acids are known to initiate intricate signaling events in a variety of tissues, primarily in the liver and gastrointestinal tract. Of the known bile acids, only the 7α-dihydroxy species, deoxycholic acid and chenodeoxycholic acid (CDCA), and their conjugates, activate processes that stimulate epithelial Cl- secretion. We have previously published that CDCA acts in a rapid manner to stimulate colonic ion secretion via protein kinase A (PKA)-mediated activation of the dominant Cl- channel, the cystic fibrosis transmembrane conductance regulator (CFTR) (Ao M, Sarathy J, Domingue J, Alrefai WA, and Rao MC. Am J Physiol Cell Physiol 305: C447-C456, 2013); however, PKA signaling did not account for the entire CDCA response. Here we show that in human colonic T84 cells, CDCA's induction of CFTR activity, measured as changes in short-circuit current (Isc), is dependent on epidermal growth factor receptor (EGFR) activation and does not involve the bile acid receptors TGR5 or farnesoid X receptor. CDCA activation of Cl- secretion does not require Src, mitogen-activated protein kinases, or phosphoinositide 3-kinase downstream of EGFR but does require an increase in cytosolic Ca2+ In addition to PKA signaling, we found that the CDCA response requires the novel involvement of the exchange protein directly activated by cAMP (EPAC). EPAC is a known hub for cAMP and Ca2+ cross talk. Downstream of EPAC, CDCA activates Rap2, and changes in free cytosolic Ca2+ were dependent on both EPAC and EGFR activation. This study establishes the complexity of CDCA signaling in the colonic epithelium and shows the contribution of EGFR, EPAC, and Ca2+ in CDCA-induced activation of CFTR-dependent Cl- secretion.
Collapse
Affiliation(s)
- Jada C Domingue
- Department of Physiology and Biophysics, University of Illinois at Chicago, Chicago, Illinois
| | - Mei Ao
- Department of Physiology and Biophysics, University of Illinois at Chicago, Chicago, Illinois
| | - Jayashree Sarathy
- Department of Physiology and Biophysics, University of Illinois at Chicago, Chicago, Illinois.,Department of Biology, Benedictine University, Lisle, Illinois
| | - Mrinalini C Rao
- Department of Physiology and Biophysics, University of Illinois at Chicago, Chicago, Illinois; .,Department of Medicine, University of Illinois at Chicago, Chicago, Illinois; and
| |
Collapse
|
32
|
Senft D, Ronai ZA. Immunogenic, cellular, and angiogenic drivers of tumor dormancy--a melanoma view. Pigment Cell Melanoma Res 2015; 29:27-42. [PMID: 26514653 DOI: 10.1111/pcmr.12432] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2015] [Accepted: 10/27/2015] [Indexed: 12/27/2022]
Abstract
In tumor cells, the ability to maintain viability over long time periods without proliferation is referred to as a state of dormancy. Maintenance of dormancy is controlled by numerous cellular and environmental factors, from immune surveillance and tumor-stroma interaction to intracellular signaling. Interference of dormancy (to an 'awaken' state) is associated with reduced response to therapy, resulting in relapse or in metastatic burst. Thus, maintaining a dormant state should prolong therapeutic responses and delay metastasis. Technical obstacles in studying tumor dormancy have limited our understanding of underlying mechanisms and hampered our ability to target dormant cells. In this review, we summarize the progress of research in the field of immunogenic, angiogenic, and cellular dormancy in diverse malignancies with particular attention to our current understanding in melanoma.
Collapse
Affiliation(s)
- Daniela Senft
- Cancer Center, Sanford Burnham Prebys Medical Discovery Institute, La Jolla, CA, USA
| | - Ze'ev A Ronai
- Cancer Center, Sanford Burnham Prebys Medical Discovery Institute, La Jolla, CA, USA
| |
Collapse
|
33
|
George AL, Suriano R, Rajoria S, Osso MC, Tuli N, Hanly E, Geliebter J, Arnold AN, Wallack M, Tiwari RK. PLX4032 Mediated Melanoma Associated Antigen Potentiation in Patient Derived Primary Melanoma Cells. J Cancer 2015; 6:1320-30. [PMID: 26640592 PMCID: PMC4643088 DOI: 10.7150/jca.11126] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2014] [Accepted: 06/19/2015] [Indexed: 01/15/2023] Open
Abstract
Over expression of various immunogenic melanoma associated antigens (MAAs) has been exploited in the development of immunotherapeutic melanoma vaccines. Expression of MAAs such as MART-1 and gp100 is modulated by the MAPK signaling pathway, which is often deregulated in melanoma. The protein BRAF, a member of the MAPK pathway, is mutated in over 60% of melanomas providing an opportunity for the identification and approval by the FDA of a small molecule MAPK signaling inhibitor PLX4032 that functions to inactivate mutant BRAF(V600E). To this end, we characterized five patient derived primary melanoma cell lines with respect to treatment with PLX4032. Cells were treated with 5μM PLX4032 and harvested. Western blotting analysis, RT-PCR and in vitro transwell migration and invasion assays were utilized to determine treatment effects. PLX4032 treatment modulated phosphorylation of signaling proteins belonging to the MAPK pathway including BRAF, MEK, and ERK and abrogated cell phenotypic characteristics such as migration and invasion. Most significantly, PLX4032 led to an up regulation of many MAA proteins in three of the four BRAF mutated cell lines, as determined at the protein and RNA level. Interestingly, MAGE-A1 protein and mRNA levels were reduced upon PLX4032 treatment in two of the primary lines. Taken together, our findings suggest that the BRAF(V600E) inhibitor PLX4032 has therapeutic potential over and above its known target and in combination with specific melanoma targeting vaccine strategies may have further clinical utility.
Collapse
Affiliation(s)
- Andrea L George
- 1. New York Medical College, Department of Microbiology and Immunology, Valhalla, New York, 10595
| | - Robert Suriano
- 2. College of Mount Saint Vincent, Division of Natural Sciences, Bronx, New York, 10471
| | - Shilpi Rajoria
- 1. New York Medical College, Department of Microbiology and Immunology, Valhalla, New York, 10595
| | - Maria C Osso
- 4. McDaniel College, Westminster, Maryland 21157
| | - Neha Tuli
- 1. New York Medical College, Department of Microbiology and Immunology, Valhalla, New York, 10595
| | - Elyse Hanly
- 1. New York Medical College, Department of Microbiology and Immunology, Valhalla, New York, 10595
| | - Jan Geliebter
- 1. New York Medical College, Department of Microbiology and Immunology, Valhalla, New York, 10595
| | - Angelo N Arnold
- 5. Westchester Medical Center, Transplant Immunogenetics Laboratory, Valhalla, New York, 10595
| | - Marc Wallack
- 1. New York Medical College, Department of Microbiology and Immunology, Valhalla, New York, 10595; ; 3. Metropolitan Hospital Center GNS, Department of Surgery, New York, New York, 10029
| | - Raj K Tiwari
- 1. New York Medical College, Department of Microbiology and Immunology, Valhalla, New York, 10595
| |
Collapse
|
34
|
Corazzari M, Rapino F, Ciccosanti F, Giglio P, Antonioli M, Conti B, Fimia GM, Lovat PE, Piacentini M. Oncogenic BRAF induces chronic ER stress condition resulting in increased basal autophagy and apoptotic resistance of cutaneous melanoma. Cell Death Differ 2015; 22:946-58. [PMID: 25361077 PMCID: PMC4423179 DOI: 10.1038/cdd.2014.183] [Citation(s) in RCA: 121] [Impact Index Per Article: 12.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2014] [Revised: 09/29/2014] [Accepted: 09/30/2014] [Indexed: 01/11/2023] Open
Abstract
The notorious unresponsiveness of metastatic cutaneous melanoma to current treatment strategies coupled with its increasing incidence constitutes a serious worldwide clinical problem. Moreover, despite recent advances in targeted therapies for patients with BRAF(V600E) mutant melanomas, acquired resistance remains a limiting factor and hence emphasises the acute need for comprehensive pre-clinical studies to increase the biological understanding of such tumours in order to develop novel effective and longlasting therapeutic strategies. Autophagy and ER stress both have a role in melanoma development/progression and chemoresistance although their real impact is still unclear. Here, we show that BRAF(V600E) induces a chronic ER stress status directly increasing basal cell autophagy. BRAF(V600E)-mediated p38 activation stimulates both the IRE1/ASK1/JNK and TRB3 pathways. Bcl-XL/Bcl-2 phosphorylation by active JNK releases Beclin1 whereas TRB3 inhibits the Akt/mTor axes, together resulting in an increase in basal autophagy. Furthermore, we demonstrate chemical chaperones relieve the BRAF(V600E)-mediated chronic ER stress status, consequently reducing basal autophagic activity and increasing the sensitivity of melanoma cells to apoptosis. Taken together, these results suggest enhanced basal autophagy, typically observed in BRAF(V600E) melanomas, is a consequence of a chronic ER stress status, which ultimately results in the chemoresistance of such tumours. Targeted therapies that attenuate ER stress may therefore represent a novel and more effective therapeutic strategy for BRAF mutant melanoma.
Collapse
Affiliation(s)
- M Corazzari
- Department of Biology, University of Rome ‘Tor Vergata', Rome, Italy
- National Institute for Infectious Diseases IRCCS ‘L. Spallanzani', Rome, Italy
| | - F Rapino
- National Institute for Infectious Diseases IRCCS ‘L. Spallanzani', Rome, Italy
| | - F Ciccosanti
- National Institute for Infectious Diseases IRCCS ‘L. Spallanzani', Rome, Italy
| | - P Giglio
- Department of Biology, University of Rome ‘Tor Vergata', Rome, Italy
| | - M Antonioli
- National Institute for Infectious Diseases IRCCS ‘L. Spallanzani', Rome, Italy
| | - B Conti
- National Institute for Infectious Diseases IRCCS ‘L. Spallanzani', Rome, Italy
| | - G M Fimia
- National Institute for Infectious Diseases IRCCS ‘L. Spallanzani', Rome, Italy
- Department of Biological and Environmental Science and Technology (Di.S.Te.B.A.), University of Salento, Lecce, Italy
| | - P E Lovat
- Dermatological Sciences Institute of Cellular Medicine, Newcastle University, Newcastle upon Tyne, UK
| | - M Piacentini
- Department of Biology, University of Rome ‘Tor Vergata', Rome, Italy
- National Institute for Infectious Diseases IRCCS ‘L. Spallanzani', Rome, Italy
| |
Collapse
|
35
|
Kim HG, Shi C, Bode AM, Dong Z. p38α MAPK is required for arsenic-induced cell transformation. Mol Carcinog 2015; 55:910-7. [PMID: 25969347 DOI: 10.1002/mc.22331] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2014] [Revised: 03/18/2015] [Accepted: 04/02/2015] [Indexed: 12/19/2022]
Abstract
Arsenic exposure has been reported to cause neoplastic transformation through the activation of PcG proteins. In the present study, we show that activation of p38α mitogen-activated protein kinase (MAPK) is required for arsenic-induced neoplastic transformation. Exposure of cells to 0.5 μM arsenic increased CRE and c-Fos promoter activities that were accompanied by increases in p38α MAPK and CREB phosphorylation and expression levels concurrently with AP-1 activation. Introduction of short hairpin (sh) RNA-p38α into BALB/c 3T3 cells markedly suppressed arsenic-induced colony formation compared with wildtype cells. CREB phosphorylation and AP-1 activation were decreased in p38α knockdown cells after arsenic treatment. Arsenic-induced AP-1 activation, measured as c-Fos and CRE promoter activities, and CREB phosphorylation were attenuated by p38 inhibition in BALB/c 3T3 cells. Thus, p38α MAPK activation is required for arsenic-induced neoplastic transformation mediated through CREB phosphorylation and AP-1 activation.
Collapse
Affiliation(s)
- Hong-Gyum Kim
- The Hormel Institute, University of Minnesota, Austin, Minnesota
| | - Chengcheng Shi
- The Hormel Institute, University of Minnesota, Austin, Minnesota.,The First Affiliated Hospital, Zhengzhou University, Zhengzhou, China
| | - Ann M Bode
- The Hormel Institute, University of Minnesota, Austin, Minnesota
| | - Zigang Dong
- The Hormel Institute, University of Minnesota, Austin, Minnesota
| |
Collapse
|
36
|
The receptor for advanced glycation end products influences the expression of its S100 protein ligands in melanoma tumors. Int J Biochem Cell Biol 2014; 57:54-62. [PMID: 25310905 DOI: 10.1016/j.biocel.2014.10.001] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2014] [Revised: 09/02/2014] [Accepted: 10/01/2014] [Indexed: 10/24/2022]
Abstract
Recent studies have suggested that the receptor for advanced glycation end products (RAGE) participates in melanoma progression by promoting tumor growth. However, the mechanisms of RAGE activation in melanoma tumors are not clearly understood. To get deeper insights into these mechanisms, we transfected a melanoma cell line, which was established from a human melanoma primary tumor, with RAGE, and studied the effect of RAGE overexpression on cell proliferation and migration in vitro. We observed that overexpression of RAGE in these cells not only resulted in significantly increased migration rates compared to control cells, but also in decreased proliferation rates (Meghnani et al., 2014). In the present study, we compared the growth of xenograft tumors established from RAGE overexpressing WM115 cells, to that of control cells. We observed that when implanted in mice, RAGE overexpressing cells generated tumors faster than control cells. Analysis of protein tumor extracts showed increased levels of the RAGE ligands S100B, S100A2, S100A4, S100A6 and S100A10 in RAGE overexpressing tumors compared to control tumors. We show that the tumor growth was significantly reduced when the mice were treated with anti-RAGE antibodies, suggesting that RAGE, and probably several S100 proteins, were involved in tumor growth. We further demonstrate that the anti-RAGE antibody treatment significantly enhanced the efficacy of the alkylating drug dacarbazine in reducing the growth rate of RAGE overexpressing tumors.
Collapse
|
37
|
Lidsky M, Antoun G, Speicher P, Adams B, Turley R, Augustine C, Tyler D, Ali-Osman F. Mitogen-activated protein kinase (MAPK) hyperactivation and enhanced NRAS expression drive acquired vemurafenib resistance in V600E BRAF melanoma cells. J Biol Chem 2014; 289:27714-26. [PMID: 25063807 PMCID: PMC4183808 DOI: 10.1074/jbc.m113.532432] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2013] [Revised: 07/15/2014] [Indexed: 01/10/2023] Open
Abstract
Although targeting the V600E activating mutation in the BRAF gene, the most common genetic abnormality in melanoma, has shown clinical efficacy in melanoma patients, response is, invariably, short lived. To better understand mechanisms underlying this acquisition of resistance to BRAF-targeted therapy in previously responsive melanomas, we induced vemurafenib resistance in two V600E BRAF+ve melanoma cell lines, A375 and DM443, by serial in vitro vemurafenib exposure. The resulting approximately 10-fold more vemurafenib-resistant cell lines, A375rVem and D443rVem, had higher growth rates and showed differential collateral resistance to cisplatin, melphalan, and temozolomide. The acquisition of vemurafenib resistance was associated with significantly increased NRAS levels in A375rVem and D443rVem, increased activation of the prosurvival protein, AKT, and the MAPKs, ERK, JNK, and P38, which correlated with decreased levels of the MAPK inhibitor protein, GSTP1. Despite the increased NRAS, whole exome sequencing showed no NRAS gene mutations. Inhibition of all three MAPKs and siRNA-mediated NRAS suppression both reversed vemurafenib resistance significantly in A375rVem and DM443rVem. Together, the results indicate a mechanism of acquired vemurafenib resistance in V600E BRAF+ve melanoma cells that involves increased activation of all three human MAPKs and the PI3K pathway, as well as increased NRAS expression, which, contrary to previous reports, was not associated with mutations in the NRAS gene. The data highlight the complexity of the acquired vemurafenib resistance phenotype and the challenge of optimizing BRAF-targeted therapy in this disease. They also suggest that targeting the MAPKs and/or NRAS may provide a strategy to mitigate such resistance in V600E BRAF+ve melanoma.
Collapse
Affiliation(s)
| | - Gamil Antoun
- the Division of Surgical Sciences, Department of Surgery, Duke University Medical Center, Durham, North Carolina 27710
| | | | - Bartley Adams
- the Division of Surgical Sciences, Department of Surgery, Duke University Medical Center, Durham, North Carolina 27710
| | | | | | | | - Francis Ali-Osman
- the Division of Surgical Sciences, Department of Surgery, Duke University Medical Center, Durham, North Carolina 27710
| |
Collapse
|
38
|
Tian Y, Guan Y, Jia Y, Meng Q, Yang J. Chloride Intracellular Channel 1 Regulates Prostate Cancer Cell Proliferation and Migration Through the MAPK/ERK Pathway. Cancer Biother Radiopharm 2014; 29:339-44. [PMID: 25279971 DOI: 10.1089/cbr.2014.1666] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022] Open
Affiliation(s)
- Yudong Tian
- Department of Urology Surgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Yanbin Guan
- School of Pharmacy, Henan University of Tradition Chinese Medicine, Zhengzhou, China
| | - Yongyan Jia
- School of Pharmacy, Henan University of Tradition Chinese Medicine, Zhengzhou, China
| | - Qingjun Meng
- Department of Urology Surgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Jinjian Yang
- Department of Urology Surgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| |
Collapse
|
39
|
Inhibition of tumor-associated αvβ3 integrin regulates the angiogenic switch by enhancing expression of IGFBP-4 leading to reduced melanoma growth and angiogenesis in vivo. Angiogenesis 2014; 18:31-46. [PMID: 25249331 DOI: 10.1007/s10456-014-9445-2] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2014] [Accepted: 09/16/2014] [Indexed: 12/20/2022]
Abstract
A more complete understanding of the mechanisms that regulate the angiogenic switch, which contributes to the conversion of small dormant tumors to actively growing malignancies, is important for the development of more effective anti-angiogenic strategies for cancer therapy. While significant progress has been made in understanding the complex mechanisms by which integrin αvβ3 expressed in endothelial cells governs angiogenesis, less is known concerning the ability of αvβ3 expressed within the tumor cell compartment to modulate the angiogenic output of a tumor. Here we provide evidence that αvβ3 expressed in melanoma cells may contribute to the suppression of IGFBP-4, an important negative regulator of IGF-1 signaling. Given the multiple context-dependent roles for αvβ3 in angiogenesis and tumor progression, our novel findings provide additional molecular insight into how αvβ3 may govern the angiogenic switch by a mechanism associated with a p38 MAPK and matrix metalloproteinases-dependent regulation of the endogenous angiogenesis inhibitor IGFBP-4.
Collapse
|
40
|
Lopez-Bergami P. The role of mitogen- and stress-activated protein kinase pathways in melanoma. Pigment Cell Melanoma Res 2014; 24:902-21. [PMID: 21914141 DOI: 10.1111/j.1755-148x.2011.00908.x] [Citation(s) in RCA: 48] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
Recent discoveries have increased our comprehension of the molecular signaling events critical for melanoma development and progression. Many oncogenes driving melanoma have been identified, and most of them exert their oncogenic effects through the activation of the RAF/MEK/ERK mitogen-activated protein kinase (MAPK) pathway. The c-Jun N-terminal kinase (JNK) and p38 MAPK pathways are also important in melanoma, but their precise role is not clear yet. This review summarizes our current knowledge on the role of the three main MAPK pathways, extracellular regulated kinase (ERK), JNK, and p38, and their impact on melanoma biology. Although the results obtained with BRAF inhibitors in melanoma patients are impressive, several mechanisms of acquired resistance have emerged. To overcome this obstacle constitutes the new challenge in melanoma therapy. Given the major role that MAPKs play in melanoma, understanding their functions and the interconnection among them and with other signaling pathways represents a step forward toward this goal.
Collapse
Affiliation(s)
- Pablo Lopez-Bergami
- Instituto de Medicina y Biología Experimental, CONICET, Buenos Aires, Argentina.
| |
Collapse
|
41
|
Muthusamy V, Piva TJ. UVB-stimulated TNFα release from human melanocyte and melanoma cells is mediated by p38 MAPK. Int J Mol Sci 2013; 14:17029-54. [PMID: 23965971 PMCID: PMC3759950 DOI: 10.3390/ijms140817029] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2013] [Revised: 08/05/2013] [Accepted: 08/09/2013] [Indexed: 01/18/2023] Open
Abstract
Ultraviolet (UV) radiation activates cell signaling pathways in melanocytes. As a result of altered signaling pathways and UV-induced cellular damage, melanocytes can undergo oncogenesis and develop into melanomas. In this study, we investigated the effect of UV-radiation on p38 MAPK (mitogen-activated protein kinase), JNK and NFκB pathways to determine which plays a major role in stimulating TNFα secretion in human HEM (melanocytes) and MM96L (melanoma) cells. MM96L cells exhibited 3.5-fold higher p38 activity than HEM cells at 5 min following UVA + B radiation and 1.6-fold higher JNK activity at 15–30 min following UVB+A radiation, while NFκB was minimally activated in both cells. Irradiated HEM cells had the greatest fold of TNFα secretion (UVB: 109-fold, UVA + B: 103-fold & UVB+A: 130-fold) when co-exposed to IL1α. The p38 inhibitor, SB202190, inhibited TNFα release by 93% from UVB-irradiated HEM cells. In the UVB-irradiated MM96L cells, both SB202190 and sulfasalazine (NFκB inhibitor) inhibited TNFα release by 52%. Although, anisomycin was a p38 MAPK activator, it inhibited TNFα release in UV-irradiated cells. This suggests that UV-mediated TNFα release may occur via different p38 pathway intermediates compared to those stimulated by anisomycin. As such, further studies into the functional role p38 MAPK plays in regulating TNFα release in UV-irradiated melanocyte-derived cells are warranted.
Collapse
Affiliation(s)
- Visalini Muthusamy
- School of Medical Sciences, RMIT University, PO Box 71, Bundoora VIC 3083, Australia.
| | | |
Collapse
|
42
|
Selective regulation of p38β protein and signaling by integrin-linked kinase mediates bladder cancer cell migration. Oncogene 2013; 33:690-701. [DOI: 10.1038/onc.2013.20] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2012] [Revised: 11/13/2012] [Accepted: 12/17/2012] [Indexed: 12/13/2022]
|
43
|
Charbaji N, Schäfer-Korting M, Küchler S. Morphine stimulates cell migration of oral epithelial cells by delta-opioid receptor activation. PLoS One 2012; 7:e42616. [PMID: 22900034 PMCID: PMC3416801 DOI: 10.1371/journal.pone.0042616] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2012] [Accepted: 07/09/2012] [Indexed: 01/13/2023] Open
Abstract
Oral mucositis is one of the most common side effects of chemoradiation regimens and manifestation can be dose-limiting for the therapy, can impair the patient's nutritional condition and quality of life due to severe pain. The therapeutic options are limited; often only an alleviation of the symptoms such as pain reduction by using systemic opioids is possible. Stimulating opioid receptors on peripheral neurons and dermal tissue, potent analgesic effects are induced e.g. in skin grafted patients. Advantageous effects on the cell migration and, thus, on the wound healing process are described, too. In this study, we investigated whether opioid receptors are also expressed on oral epithelial cells and if morphine can modulate their cell migration behavior. The expression of the opioid receptors MOR, DOR and KOR on primary human oral epithelial cells was verified. Furthermore, a significantly accelerated cell migration was observed following incubation with morphine. The effect even slightly exceeded the cell migration stimulating effect of TGF-ß: After 14 h of morphine treatment about 86% of the wound area was closed, whereas TGF-ß application resulted in a closed wound area of 80%. With respect to morphine stimulated cell migration we demonstrate that DOR plays a key role and we show the involvement of the MAPK members Erk 1/2 and p38 using Western blot analysis.Further studies in more complex systems in vitro and in vivo are required. Nevertheless, these findings might open up a new therapeutic option for the treatment of oral mucositis.
Collapse
Affiliation(s)
- Nada Charbaji
- Institute of Pharmacy, Freie Universität Berlin, Berlin, Germany
| | | | - Sarah Küchler
- Institute of Pharmacy, Freie Universität Berlin, Berlin, Germany
| |
Collapse
|
44
|
Abstract
Selective BRAF inhibitors have recently emerged as a new standard treatment for patients with metastatic melanoma harboring activating BRAF mutations. Inhibition of the MAP kinase pathway and initial evidence of antitumor effects are very reliably observed. However, many patients experience short-lived responses, whereas others are durable. An overall survival benefit has been established for them, BRAF in it, the agents that have advanced furthest in clinical development. Nonetheless, attention has immediately turned to understanding de novo and acquired resistance and effort to develop rational combination therapy that will further improve patient outcomes. Opportunities for combining BRAF inhibitors with other signal transduction inhibitors as well as targeted therapies with distinct mechanisms of action are discussed.
Collapse
|
45
|
Zheng ST, Zhang CS, Qin X, Gen YH, Liu T, Sheyhidin I, Lu XM. The status of phosphorylated p38 in esophageal squamous cell carcinoma. Mol Biol Rep 2012; 39:5315-5321. [PMID: 22167332 DOI: 10.1007/s11033-011-1330-0] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2011] [Accepted: 12/03/2011] [Indexed: 12/23/2022]
Abstract
The p38 mitogen-activated protein kinase (MAPK) is a member of the MAPK family, which is initially found to be activated by stress stimuli, proinflammatory cytokines, and growth factors. However, its role in the pathogenesis of esophageal squamous cell carcinoma (ESCC) is largely unkown, so we investigate the role of phosphorylated p38 (p-p38) MAPK in ESCC. First of all, in vitro cell line ECa109, SB203580 as selective inhibitor of p38, can suppress the growth of esophageal cancer cell in a dose- and time-dependent way, suggesting that ECa109 cell growth and proliferation was closely associated with p-p38. Using western-blot analysis of fresh 16 paired surgically resected ESCC and matched non-tumor adjacent tissues (NAT), we showed that p-p38 was significantly expressed higher in NAT compared to ESCC. Moreover, expressions of p-p38 were further confirmed by 162 paired of formalin-fixed paraffin-embedded (FFPE) ESCC and NAT by immunohistochemistry, the same trend result was obtained through statistical analysis that there was increased expression of p-p38 in NAT as compared with ESCC (P < 0.01), and expression of p-p38 was not significantly associated with lymph nodes metastasis (P > 0.05) and ESCC differentiation degree (P > 0.05). Taken together, all the results we obtained demonstrated that p-p38 plays a key role in the malignant transformation of ESCC.
Collapse
Affiliation(s)
- Shu-Tao Zheng
- Medical Research Center, The Affiliated Hospital, Xinjiang Medical University, Urumqi, 830054, Xinjiang Uygur Autonomous Region, People's Republic of China
| | | | | | | | | | | | | |
Collapse
|
46
|
Sunaga N, Imai H, Shimizu K, Shames DS, Kakegawa S, Girard L, Sato M, Kaira K, Ishizuka T, Gazdar AF, Minna JD, Mori M. Oncogenic KRAS-induced interleukin-8 overexpression promotes cell growth and migration and contributes to aggressive phenotypes of non-small cell lung cancer. Int J Cancer 2011; 130:1733-44. [PMID: 21544811 DOI: 10.1002/ijc.26164] [Citation(s) in RCA: 65] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2011] [Accepted: 04/21/2011] [Indexed: 12/21/2022]
Abstract
The CXC chemokine interleukin-8 (IL-8) is an angiogenic growth factor that is overexpressed in various cancers, including non-small cell lung cancer (NSCLC). Previously, IL-8 was shown as a transcriptional target of RAS signaling, raising the possibility of its role in oncogenic KRAS-driven NSCLC. Using microarray analysis, we identified IL-8 as the most downregulated gene by shRNA-mediated KRAS knockdown in NCI-H1792 NSCLC cells where IL-8 is overexpressed. NSCLC cell lines harboring KRAS or EGFR mutations overexpressed IL-8, while IL-8 levels were more prominent in KRAS mutants compared to EGFR mutants. IL-8 expression was downregulated by shRNA-mediated KRAS knockdown in KRAS mutants or by treatment with EGFR tyrosine kinase inhibitors and EGFR siRNAs in EGFR mutants. In our analysis of the relationship of IL-8 expression with clinical parameters and mutation status of KRAS or EGFR in 89 NSCLC surgical specimens, IL-8 expression was shown to be significantly higher in NSCLCs of males, smokers, and elderly patients and those with pleural involvement and KRAS mutated adenocarcinomas. In KRAS mutant cells, the MEK inhibitor markedly decreased IL-8 expression, while the p38 inhibitor increased IL-8 expression. Attenuation of IL-8 function by siRNAs or a neutralizing antibody inhibited cell proliferation and migration of KRAS mutant/IL-8 overexpressing NSCLC cells. These results indicate that activating mutations of KRAS or EGFR upregulate IL-8 expression in NSCLC; IL-8 is highly expressed in NSCLCs from males, smokers, elderly patients, NSCLCs with pleural involvement, and KRAS-mutated adenocarcinomas; and IL-8 plays a role in cell growth and migration in oncogenic KRAS-driven NSCLC.
Collapse
Affiliation(s)
- Noriaki Sunaga
- Department of Medicine and Molecular Science, Gunma University Graduate School of Medicine, 3-39-15 Showa-machi, Maebashi, Gunma, Japan.
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
47
|
Hu N, Li Y, Zhao Y, Wang Q, You JC, Zhang XD, Ye LH. A novel positive feedback loop involving FASN/p-ERK1/2/5-LOX/LTB4/FASN sustains high growth of breast cancer cells. Acta Pharmacol Sin 2011; 32:921-9. [PMID: 21643005 DOI: 10.1038/aps.2011.40] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/16/2023] Open
Abstract
AIM To investigate the endogenous signaling pathways associated with high proliferation potential of breast cancer cells. METHODS Breast cancer cell lines LM-MCF-7 and MCF-7 with high and low proliferation capability were used. The promoter activity of fatty acid synthase (FASN) was examined using luciferase reporter gene assay. The expression level of FASN mRNA was measured using RT-PCR and real time PCR, respectively. The level of leukotriene B4 (LTB4) was determined with ELISA. The expression levels of 5-lipoxygenase (5-LOX) was analyzed using RT-PCR and Western blot, respectively. 5-Bromo-20-deoxyuridine (BrdU) incorporation assay was used to study the proliferation of LM-MCF-7 and MCF-7 cells. RESULTS The promoter activity of FASN was significantly higher in LM-MCF-7 cells than MCF-7 cells. Treatment of LM-MCF-7 cells with ERK1/2 inhibitor PD98059 (30-50 μmol/L) or LOX inhibitor NDGA (25 μmol/L) abolished the activation of FASN. Moreover, treatment of LM-MCF-7 cells with the specific 5-LOX inhibitor MK-886 (20-40 μmol/L) or 5-LOX siRNA (50-100 nmol/L) decreased the promoter activity of FASN. The level of LTB4, the final metabolite produced by 5-LOX, was significantly higher in LM-MCF-7 cells than MCF-7 cells. Administration of exogenous LTB4 (1-10 nmol/L) was able to stimulate the promoter activity of FASN in MCF-7 cells. Treatment of LM-MCF-7 cells with the FASN inhibitor cerulenin (10 μmol/L) reduced all the levels of p-ERK1/2, 5-LOX, and LTB4. Treatment of LM-MCF-7 cells with cerulenin, PD98059, or MK-886 abolished the proliferation. Administration of exogenous LTB4 (10 nmol/L) significantly increased BrdU incorporation in MCF-7 cells. CONCLUSION THESE results suggest a novel positive feedback loop involving FASN/p-ERK1/2/5-LOX/LTB4/FASN contributes to the sustaining growth of breast cancer LM-MCF-7 cells.
Collapse
|
48
|
Luan Q, Sun J, Li C, Zhang G, Lv Y, Wang G, Li C, Ma C, Gao T. Mutual enhancement between heparanase and vascular endothelial growth factor: a novel mechanism for melanoma progression. Cancer Lett 2011; 308:100-11. [PMID: 21624769 DOI: 10.1016/j.canlet.2011.04.019] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2010] [Revised: 03/23/2011] [Accepted: 04/28/2011] [Indexed: 12/17/2022]
Abstract
Heparanase is closely related to growth factors in the role of promoting tumor progression. Among them, vascular endothelial growth factor (VEGF) is necessary for tumor vascularity and metastasis. Release of VEGF by heparanase can initiate relative signaling pathways, resulting in an up-regulation of transcriptional factors related with heparanase. Therefore, VEGF likely has a potential function as a regulator of heparanase expression in melanoma. We hypothesized that a novel mechanism exists where heparanase and VEGF are mutually enhanced in melanoma. Our study was conducted to validate the hypothetical mutual enhancement and elucidate its effect on melanoma progression. We found that the addition of exogenous VEGF and its cDNA transfection induce heparanase over-expression by means of western-blot and real-time RT-PCR, while anti-VEGF siRNA reduces heparanase expression in A2058 and WM793 melanoma cell lines. Likewise, VEGF expression is also regulated by heparanase in these two cell lines. Additionally, the cells with mutual enhancement phenotypes exhibit higher proliferation and transmigration capacity. PD98059, a specific inhibitor of the MEK/ERK signaling pathway, is involved in this mutual enhancement. These data are the first to show that heparanase and VEGF have a mutual enhancement in melanoma cells, which may be a novel mechanism for promoting melanoma progression.
Collapse
Affiliation(s)
- Qi Luan
- Department of Dermatology, Xijing Hospital, Fourth Military Medical University, Xi'an, Shaanxi Province, China
| | | | | | | | | | | | | | | | | |
Collapse
|
49
|
Zhang YY, Wu JW, Wang ZX. Mitogen-activated protein kinase (MAPK) phosphatase 3-mediated cross-talk between MAPKs ERK2 and p38alpha. J Biol Chem 2011; 286:16150-62. [PMID: 21454500 PMCID: PMC3091224 DOI: 10.1074/jbc.m110.203786] [Citation(s) in RCA: 50] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023] Open
Abstract
MAPK phosphatase 3 (MKP3) is highly specific for ERK1/2 inactivation via dephosphorylation of both phosphotyrosine and phosphothreonine critical for enzymatic activation. Here, we show that MKP3 is able to effectively dephosphorylate the phosphotyrosine, but not phosphothreonine, in the activation loop of p38α in vitro and in intact cells. The catalytic constant of the MKP3 reaction for p38α is comparable with that for ERK2. Remarkably, MKP3, ERK2, and phosphorylated p38α can form a stable ternary complex in solution, and the phosphatase activity of MKP3 toward p38α substrate is allosterically regulated by ERK2-MKP3 interaction. This suggests that MKP3 not only controls the activities of ERK2 and p38α but also mediates cross-talk between these two MAPK pathways. The crystal structure of bisphosphorylated p38α has been determined at 2.1 Å resolution. Comparisons between the phosphorylated MAPK structures reveal the molecular basis of MKP3 substrate specificity.
Collapse
Affiliation(s)
- Yuan-Yuan Zhang
- From the Ministry of Education Key Laboratory of Bioinformatics, School of Life Sciences, Tsinghua University, Beijing 100084, China and
| | - Jia-Wei Wu
- From the Ministry of Education Key Laboratory of Bioinformatics, School of Life Sciences, Tsinghua University, Beijing 100084, China and
| | - Zhi-Xin Wang
- From the Ministry of Education Key Laboratory of Bioinformatics, School of Life Sciences, Tsinghua University, Beijing 100084, China and ,the Institute of Biophysics, Chinese Academy of Sciences, Beijing 100101, China, To whom correspondence should be addressed. Tel.: 86-10-62785505; Fax: 86-10-62792826; E-mail:
| |
Collapse
|
50
|
Loughlin DT, Artlett CM. Modification of collagen by 3-deoxyglucosone alters wound healing through differential regulation of p38 MAP kinase. PLoS One 2011; 6:e18676. [PMID: 21573155 PMCID: PMC3089600 DOI: 10.1371/journal.pone.0018676] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2010] [Accepted: 03/15/2011] [Indexed: 02/05/2023] Open
Abstract
Background Wound healing is a highly dynamic process that requires signaling from the extracellular matrix to the fibroblasts for migration and proliferation, and closure of the wound. This rate of wound closure is impaired in diabetes, which may be due to the increased levels of the precursor for advanced glycation end products, 3-deoxyglucosone (3DG). Previous studies suggest a differential role for p38 mitogen-activated kinase (MAPK) during wound healing; whereby, p38 MAPK acts as a growth kinase during normal wound healing, but acts as a stress kinase during diabetic wound repair. Therefore, we investigated the signaling cross-talk by which p38 MAPK mediates wound healing in fibroblasts cultured on native collagen and 3DG-collagen. Methodology/Principal Findings Using human dermal fibroblasts cultured on 3DG-collagen as a model of diabetic wounds, we demonstrated that p38 MAPK can promote either cell growth or cell death, and this was dependent on the activation of AKT and ERK1/2. Wound closure on native collagen was dependent on p38 MAPK phosphorylation of AKT and ERK1/2. Furthermore, proliferation and collagen production in fibroblasts cultured on native collagen was dependent on p38 MAPK regulation of AKT and ERK1/2. In contrast, 3DG-collagen decreased fibroblast migration, proliferation, and collagen expression through ERK1/2 and AKT downregulation via p38 MAPK. Conclusions/Significance Taken together, the present study shows that p38 MAPK is a key signaling molecule that plays a significantly opposite role during times of cellular growth and cellular stress, which may account for the differing rates of wound closure seen in diabetic populations.
Collapse
Affiliation(s)
- Danielle T. Loughlin
- Department of Microbiology and Immunology, Drexel University College of Medicine, Philadelphia, Pennsylvania, United States of America
| | - Carol M. Artlett
- Department of Microbiology and Immunology, Drexel University College of Medicine, Philadelphia, Pennsylvania, United States of America
- * E-mail:
| |
Collapse
|