1
|
Lee H, Nguyen AT, Choi H, Kim KY, Kim H. Anti-cancer Effects of 1,4-Dialkoxynaphthalene-Imidazolium Salt Derivatives through ERK5 kinase activity inhibition. Sci Rep 2025; 15:13648. [PMID: 40254676 PMCID: PMC12009964 DOI: 10.1038/s41598-025-96306-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2024] [Accepted: 03/27/2025] [Indexed: 04/22/2025] Open
Abstract
ERK5 has emerged as a promising therapeutic target in cancer treatment due to its pivotal role in regulating tumor cell proliferation and survival. In this study, we synthesized novel derivatives of 1,4-dialkoxynaphthalene-2-acyl or 2-alkyl-imidazolium salt (NAIMS), assessed their binding affinity with the ERK5 protein through molecular modeling, and evaluated their anti-cancer activity through the ERK5 kinase assay. Based on the MTT assay and qRT-PCR analysis of 21 synthesized NAIMS, the IC50 values for 4c, 4e, and 4k (8.5 μM, 6.8 μM, and 8.9 μM, respectively) and the inhibition rate of the expression of PCNA for 4c, 4e, and 4k (50%, 61.1%, and 70.2% of 5 μM respectively) were chosen for comprehensive biological research. Further analyses including DAPI staining, and flow cytometry confirmed that 4c, 4e, and 4k induced late-stage apoptosis, and triggered cell cycle arrest in the G2/M phase of HeLa cells. Moreover, molecular modeling analysis showed that 4e exhibited strong and stable molecular interactions at the ERK5 ATP-binding site. Our results strongly suggest that NAIMS compounds, especially 4e, could serve as novel inhibitors of ERK5, presenting promising lead compound to develop for cancer treatment.
Collapse
Affiliation(s)
- Haena Lee
- Department of Applied Chemistry, Global Center for Pharmaceutical Ingredient Materials, Kyung Hee University, Giheung, Yongin, Gyeonggi-Do, 1732, Republic of Korea
| | - Anh-Thu Nguyen
- Department of Genetics and Biotechnology, College of Life Science, Graduate School of Biotechnology, Kyung Hee University, Giheung, Yongin, Gyeonggi-Do, 1732, Republic of Korea
| | - Hyunkyung Choi
- Division of Medical Oncology, Department of Internal Medicine, Korea University College of Medicine, Korea University, Seoul, 02841, Republic of Korea.
- Department of Biomedical Research Center, Korea University Guro Hospital, Korea University, Seoul, 08308, Republic of Korea.
| | - Ki-Young Kim
- Department of Genetics and Biotechnology, College of Life Science, Graduate School of Biotechnology, Kyung Hee University, Giheung, Yongin, Gyeonggi-Do, 1732, Republic of Korea.
| | - Hakwon Kim
- Department of Applied Chemistry, Global Center for Pharmaceutical Ingredient Materials, Kyung Hee University, Giheung, Yongin, Gyeonggi-Do, 1732, Republic of Korea.
| |
Collapse
|
2
|
Liu X, Dong X, Hu Y, Dong C, Wu S, Fang Y, Hu Y. TXN promotes tumorigenesis by activating the ERK1/2 and ERK5 signaling pathways regulating c-Myc in non-small cell lung cancer. Cell Signal 2024; 125:111517. [PMID: 39571701 DOI: 10.1016/j.cellsig.2024.111517] [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: 07/28/2024] [Revised: 11/01/2024] [Accepted: 11/18/2024] [Indexed: 11/29/2024]
Abstract
Lung cancer is the primary cause of cancer-related deaths worldwide, particularly for non-small cell lung cancer (NSCLC). However, the exact mechanism underlying tumor formation remains unclear. It is widely acknowledged that inflammation and oxidative stress occur in the tumor microenvironment, promoting cell malignant growth and metastasis. Thioredoxin-1 (TXN), the main regulator of oxidative stress, plays a significant role in the development of NSCLC. However, the specific tumor-promoting mechanism is still being investigated. This study aimed to examine the function and mechanism of TXN in NSCLC. The effects of knockdown or overexpression TXN on cell proliferation, invasion and apoptosis were evaluated by Cell Counting Kit-8, colony formation, wound healing, transwell, TUNEL staining, and flow cytometric assays. Western blotting was performed to analyze the regulation of TXN and downstream proteins suppressed by genes and pharmacology. TXN knockdown significantly suppressed cell proliferation, invasion and promoted apoptosis both in vitro and in vivo, whereas TXN overexpression reversed these malignant phenotypes. We found that TXN regulated c-Myc expression through ERK1/2 and ERK5 signaling pathways. Suppressing ERK1/2 led to the compensatory activation of ERK5, and simultaneously inhibiting ERK1/2 and ERK5 synergistically reduced c-Myc expression, further attenuating cell proliferation, invasion and enhanced apoptosis. Our results indicated tumor promotion of TXN in NSCLC and TXN regulated c-Myc in the interest of tumorigenesis through ERK1/2 and ERK5 signaling pathways. Targeting TXN and blocking the ERK1/2 and ERK5 pathways could potentially offer new therapeutic strategies for NSCLC.
Collapse
Affiliation(s)
- Xiaoting Liu
- Department of Respiratory, The Second Affiliated Hospital of Xi'an Jiaotong University, No. 157 Xiwu Road, Xi'an 710004, Shaanxi Province, China
| | - Xilin Dong
- Department of Respiratory, The Second Affiliated Hospital of Xi'an Jiaotong University, No. 157 Xiwu Road, Xi'an 710004, Shaanxi Province, China.
| | - YiFan Hu
- Department of Respiratory, The Second Affiliated Hospital of Xi'an Jiaotong University, No. 157 Xiwu Road, Xi'an 710004, Shaanxi Province, China
| | - Cong Dong
- Department of Rehabilitation, The Second Affiliated Hospital of Xi'an Jiaotong University, No. 157 Xiwu Road, Xi'an 710004, Shaanxi Province, China
| | - Sanzhu Wu
- School of Computer Science, Xi'an Shiyou University, No. 18, East Section, Electronic Second Road, Xi'an 710065, Shaanxi Province, China
| | - Yanan Fang
- Department of Respiratory, The Second Affiliated Hospital of Xi'an Jiaotong University, No. 157 Xiwu Road, Xi'an 710004, Shaanxi Province, China
| | - Yaxin Hu
- Department of Respiratory, The Second Affiliated Hospital of Xi'an Jiaotong University, No. 157 Xiwu Road, Xi'an 710004, Shaanxi Province, China
| |
Collapse
|
3
|
Dong H, Han J, Chen X, Sun H, Han M, Wang W. LncRNA ZNF649-AS1 promotes trastuzumab resistance and TAM-dependent PD-L1 expression in breast cancer by regulating EXOC7 alternative splicing. Arch Biochem Biophys 2024; 761:110128. [PMID: 39159899 DOI: 10.1016/j.abb.2024.110128] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2024] [Revised: 07/31/2024] [Accepted: 08/16/2024] [Indexed: 08/21/2024]
Abstract
BACKGROUND Trastuzumab resistance is a serious clinical problem in the treatment of HER2-positive breast cancer (BC). The lncRNA ZNF649-AS1 was previously found to promote HER2-positive BC trastuzumab resistance. The study aims to explore the molecular mechanism of ZNF649-AS1 in HER2-positive BC trastuzumab resistance. METHODS Tumor tissue and peripheral blood samples were collected from 20 HER2-positive BC patients with trastuzumab-resistant and non-resistant, respectively. Trastuzumab-resistant BC cell lines SKBR-3-TR and BT474-TR were established. RIP was employed to confirm the binding of ZNF649-AS1, PRPF8 and exocyst complex component 7 (EXOC7). RNA expression of EXOC7-L (Full length of EXOC7) and EXOC7-S (Spliceosome of EXOC7) were detected using agarose gel electrophoresis. Expressions of macrophage markers CD68+ CD206+ were measured by flow cytometry. RESULTS ZNF649-AS1 expression was upregulated in HER2-positive BC trastuzumab resistance. ZNF649-AS1 downregulation inhibited trastuzumab resistance in HER2-positive BC. ZNF649-AS1 regulated EXOC7 alternative splicing by binding with PRPF8. EXOC7-S knockdown suppressed trastuzumab resistance and TAM-dependent PD-L1 expression in HER2-positive BC. EXOC7-S overexpression abolished the effects of ZNF649-AS1 knockdown on trastuzumab resistance and TAM-dependent PD-L1 expression in HER2-positive BC. CONCLUSION ZNF649-AS1 promoted trastuzumab resistance and TAM-dependent PD-L1 expression in HER2-positive BC via promoting alternative splicing of EXOC7 by PRPF8.
Collapse
Affiliation(s)
- Huaying Dong
- Department of General Surgery, Hainan General Hospital, Hainan Affiliated Hospital of Hainan Medical University, Haikou, 570311, Hainan Province, PR China
| | - Jing Han
- Department of General Surgery, Hainan General Hospital, Hainan Affiliated Hospital of Hainan Medical University, Haikou, 570311, Hainan Province, PR China
| | - Xiang Chen
- Department of General Surgery, Hainan General Hospital, Hainan Affiliated Hospital of Hainan Medical University, Haikou, 570311, Hainan Province, PR China
| | - Hening Sun
- Department of General Surgery, Hainan General Hospital, Hainan Affiliated Hospital of Hainan Medical University, Haikou, 570311, Hainan Province, PR China
| | - Mingli Han
- Department of Breast Surgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, Henan Province, PR China.
| | - Wei Wang
- Department of General Surgery, Hainan General Hospital, Hainan Affiliated Hospital of Hainan Medical University, Haikou, 570311, Hainan Province, PR China.
| |
Collapse
|
4
|
Dai X, Mizukami Y, Watanabe K, Tsuda T, Shidahara M, Yoshida S, Yatsuzuka K, Shiraishi K, Mori H, Murakami M, Kawakami R, Imamura T, Fujisawa Y, Muto J. Trehalose Prevents IL-4/IL-13-Induced Skin Barrier Impairment by Suppressing IL-33 Expression and Increasing NRF2 Activation in Human Keratinocytes In Vitro. J Invest Dermatol 2024:S0022-202X(24)02175-4. [PMID: 39384017 DOI: 10.1016/j.jid.2024.08.038] [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: 10/07/2023] [Revised: 08/16/2024] [Accepted: 08/25/2024] [Indexed: 10/11/2024]
Abstract
Skin barrier dysfunction initiates or deteriorates various cutaneous problems, such as atopic dermatitis. At high concentrations, the nonreducing disaccharide trehalose (α-d-glucopyranosyl α-d-glucopyranoside) induces a transient senescence-like state in fibroblasts and promotes wound repair. In this study, we investigated the effect of trehalose on normal human keratinocytes and demonstrated its specific role in the skin barrier. RNA-sequencing analysis revealed that trehalose regulates the expression of many skin barrier-associated genes. T helper 2 cytokines IL-4/IL-13 were observed to downregulate several differentiation markers (FLG, loricrin, keratin 1, and keratin 10) and epidermal antimicrobial proteins in monolayer-cultured keratinocytes and living skin equivalents and impaired skin barrier function in living skin equivalents, all of which were significantly upregulated or restored by trehalose. Trehalose inhibited IL-33 expression and reduced nuclear IL-33 levels by activating MAPK/extracellular signal-regulated kinase kinase 5-extracellular signal-regulated kinase 5 and suppressing extracellular signal-regulated kinase kinase 1/2-extracellular signal-regulated kinase pathway. It also increased NRF2 activation to trigger antioxidant enzyme production through JNK, thus neutralizing IL-4/IL-13-mediated oxidative stress. Trehalose prevented IL-4/IL-13-mediated signal transducer and activator of transcription 3/signal transducer and activator of transcription 6 activation and restored IL-4/IL-13-suppressed skin barrier molecules through IL-33 downregulation and NRF2 activation. This study demonstrated that trehalose may play a role in skin barrier repair in atopic dermatitis.
Collapse
Affiliation(s)
- Xiuju Dai
- Department of Dermatology, Ehime University Graduate School of Medicine, Ehime, Japan; Department of Dermatology, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiaotong University, Shanghai, China
| | - Yoichi Mizukami
- Institute of Gene Research, Yamaguchi University Science Research Center, Yamaguchi, Japan
| | - Kenji Watanabe
- Institute of Gene Research, Yamaguchi University Science Research Center, Yamaguchi, Japan
| | - Teruko Tsuda
- Department of Dermatology, Ehime University Graduate School of Medicine, Ehime, Japan
| | - Mutsumi Shidahara
- Department of Dermatology, Ehime University Graduate School of Medicine, Ehime, Japan
| | - Satoshi Yoshida
- Department of Dermatology, Ehime University Graduate School of Medicine, Ehime, Japan
| | - Kazuki Yatsuzuka
- Department of Dermatology, Ehime University Graduate School of Medicine, Ehime, Japan
| | - Ken Shiraishi
- Department of Dermatology, Ehime University Graduate School of Medicine, Ehime, Japan
| | - Hideki Mori
- Department of Dermatology, Ehime University Graduate School of Medicine, Ehime, Japan
| | - Masamoto Murakami
- Department of Dermatology, Ehime University Graduate School of Medicine, Ehime, Japan
| | - Ryosuke Kawakami
- Department of Molecular Medicine for Pathogenesis, Ehime University Graduate School of Medicine, Ehime, Japan
| | - Takeshi Imamura
- Department of Molecular Medicine for Pathogenesis, Ehime University Graduate School of Medicine, Ehime, Japan; Translational Research Center, Ehime University Hospital, Ehime, Japan
| | - Yasuhiro Fujisawa
- Department of Dermatology, Ehime University Graduate School of Medicine, Ehime, Japan
| | - Jun Muto
- Department of Dermatology, Ehime University Graduate School of Medicine, Ehime, Japan.
| |
Collapse
|
5
|
Filippelli A, Ciccone V, Del Gaudio C, Simonis V, Frosini M, Tusa I, Menconi A, Rovida E, Donnini S. ERK5 mediates pro-tumorigenic phenotype in non-small lung cancer cells induced by PGE2. BIOCHIMICA ET BIOPHYSICA ACTA. MOLECULAR CELL RESEARCH 2024; 1871:119810. [PMID: 39128596 DOI: 10.1016/j.bbamcr.2024.119810] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/24/2024] [Revised: 07/25/2024] [Accepted: 08/04/2024] [Indexed: 08/13/2024]
Abstract
Lung cancer is the leading cause of cancer-related deaths worldwide, with non-small cell lung cancer (NSCLC) constituting approximately 84 % of all lung cancer cases. The role of inflammation in the initiation and progression of NSCLC tumors has been the focus of extensive research. Among the various inflammatory mediators, prostaglandin E2 (PGE2) plays a pivotal role in promoting the aggressiveness of epithelial tumors through multiple mechanisms, including the stimulation of growth, evasion of apoptosis, invasion, and induction of angiogenesis. The Extracellular signal-Regulated Kinase 5 (ERK5), the last discovered member among conventional mitogen-activated protein kinases (MAPK), is implicated in cancer-associated inflammation. In this study, we explored whether ERK5 is involved in the process of tumorigenesis induced by PGE2. Using A549 and PC9 NSCLC cell lines, we found that PGE2 triggers the activation of ERK5 via the EP1 receptor. Moreover, both genetic and pharmacological inhibition of ERK5 reduced PGE2-induced proliferation, migration, invasion and stemness of A549 and PC9 cells, indicating that ERK5 plays a critical role in PGE2-induced tumorigenesis. In summary, our study underscores the pivotal role of the PGE2/EP1/ERK5 axis in driving the malignancy of NSCLC cells in vitro. Targeting this axis holds promise as a potential avenue for developing novel therapeutic strategies aimed at controlling the advancement of NSCLC.
Collapse
Affiliation(s)
| | - Valerio Ciccone
- Department of Life Sciences, University of Siena, 53100 Siena, Italy
| | - Cinzia Del Gaudio
- Department of Life Sciences, University of Siena, 53100 Siena, Italy
| | - Vittoria Simonis
- Department of Life Sciences, University of Siena, 53100 Siena, Italy
| | - Maria Frosini
- Department of Life Sciences, University of Siena, 53100 Siena, Italy
| | - Ignazia Tusa
- Department of Experimental and Clinical Biomedical Sciences "Mario Serio", University of Florence, 50134 Florence, Italy
| | - Alessio Menconi
- Department of Experimental and Clinical Biomedical Sciences "Mario Serio", University of Florence, 50134 Florence, Italy
| | - Elisabetta Rovida
- Department of Experimental and Clinical Biomedical Sciences "Mario Serio", University of Florence, 50134 Florence, Italy.
| | - Sandra Donnini
- Department of Life Sciences, University of Siena, 53100 Siena, Italy.
| |
Collapse
|
6
|
Mondru AK, Wilkinson B, Aljasir MA, Alrumayh A, Greaves G, Emmett M, Albohairi S, Pritchard-Jones R, Cross MJ. The ERK5 pathway in BRAFV600E melanoma cells plays a role in development of acquired resistance to dabrafenib but not vemurafenib. FEBS Lett 2024; 598:2011-2027. [PMID: 38977937 DOI: 10.1002/1873-3468.14960] [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: 01/23/2024] [Revised: 04/28/2024] [Accepted: 05/14/2024] [Indexed: 07/10/2024]
Abstract
Malignant melanoma, an aggressive skin cancer with a poor prognosis, frequently features BRAFV600E mutation resulting in activation of the MAPK pathway and melanocyte proliferation and survival. BRAFV600E inhibitors like vemurafenib and dabrafenib have enhanced patient survival, yet drug resistance remains a significant challenge. We investigated the role of the ERK5 pathway in BRAFV600E melanoma cells and cells with acquired resistance to PLX4720 (vemurafenib) and dabrafenib. In BRAFV600E melanoma, ERK5 inhibition minimally affected viability compared to ERK1/2 inhibition. In vemurafenib-resistant cells, ERK5 inhibition alone didn't impact viability or restore drug sensitivity to vemurafenib. However, in dabrafenib-resistant cells, ERK5 inhibition reduced viability and enhanced the anti-proliferative effect of MEK1/2 inhibition. Targeting the ERK5 pathway may represent a therapeutic opportunity in dabrafenib-resistant melanoma.
Collapse
Affiliation(s)
- Anil Kumar Mondru
- Department of Pharmacology and Therapeutics, Institute of Systems, Molecular and Integrative Biology, University of Liverpool, UK
| | - Beth Wilkinson
- Department of Pharmacology and Therapeutics, Institute of Systems, Molecular and Integrative Biology, University of Liverpool, UK
| | - Mohammad A Aljasir
- Department of Pharmacology and Therapeutics, Institute of Systems, Molecular and Integrative Biology, University of Liverpool, UK
| | - Ahmed Alrumayh
- Department of Pharmacology and Therapeutics, Institute of Systems, Molecular and Integrative Biology, University of Liverpool, UK
| | - Georgia Greaves
- Department of Pharmacology and Therapeutics, Institute of Systems, Molecular and Integrative Biology, University of Liverpool, UK
| | - Maxine Emmett
- Department of Pharmacology and Therapeutics, Institute of Systems, Molecular and Integrative Biology, University of Liverpool, UK
| | - Saad Albohairi
- Department of Pharmacology and Therapeutics, Institute of Systems, Molecular and Integrative Biology, University of Liverpool, UK
| | - Rowan Pritchard-Jones
- Department of Molecular and Clinical Cancer Medicine, Institute of Systems, Molecular and Integrative Biology, University of Liverpool, UK
| | - Michael J Cross
- Department of Pharmacology and Therapeutics, Institute of Systems, Molecular and Integrative Biology, University of Liverpool, UK
| |
Collapse
|
7
|
Nussinov R, Yavuz BR, Jang H. Anticancer drugs: How to select small molecule combinations? Trends Pharmacol Sci 2024; 45:503-519. [PMID: 38782689 PMCID: PMC11162304 DOI: 10.1016/j.tips.2024.04.012] [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: 03/20/2024] [Revised: 04/26/2024] [Accepted: 04/29/2024] [Indexed: 05/25/2024]
Abstract
Small molecules are at the forefront of anticancer therapies. Successive treatments with single molecules incur drug resistance, calling for combination. Here, we explore the tough choices oncologists face - not just which drugs to use but also the best treatment plans, based on factors such as target proteins, pathways, and gene expression. We consider the reality of cancer's disruption of normal cellular processes, highlighting why it's crucial to understand the ins and outs of current treatment methods. The discussion on using combination drug therapies to target multiple pathways sheds light on a promising approach while also acknowledging the hurdles that come with it, such as dealing with pathway crosstalk. We review options and provide examples and the mechanistic basis, altogether providing the first comprehensive guide to combinatorial therapy selection.
Collapse
Affiliation(s)
- Ruth Nussinov
- Computational Structural Biology Section, Frederick National Laboratory for Cancer Research, Frederick, MD 21702, USA; Cancer Innovation Laboratory, National Cancer Institute at Frederick, Frederick, MD 21702, USA; Department of Human Molecular Genetics and Biochemistry, Sackler School of Medicine, Tel Aviv University, Tel Aviv 69978, Israel.
| | - Bengi Ruken Yavuz
- Cancer Innovation Laboratory, National Cancer Institute at Frederick, Frederick, MD 21702, USA
| | - Hyunbum Jang
- Computational Structural Biology Section, Frederick National Laboratory for Cancer Research, Frederick, MD 21702, USA; Cancer Innovation Laboratory, National Cancer Institute at Frederick, Frederick, MD 21702, USA
| |
Collapse
|
8
|
Soares Martins T, Pelech S, Ferreira M, Pinho B, Leandro K, de Almeida LP, Breitling B, Hansen N, Esselmann H, Wiltfang J, da Cruz e Silva OAB, Henriques AG. Phosphoproteome Microarray Analysis of Extracellular Particles as a Tool to Explore Novel Biomarker Candidates for Alzheimer's Disease. Int J Mol Sci 2024; 25:1584. [PMID: 38338863 PMCID: PMC10855802 DOI: 10.3390/ijms25031584] [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: 11/30/2023] [Revised: 01/18/2024] [Accepted: 01/22/2024] [Indexed: 02/12/2024] Open
Abstract
Phosphorylation plays a key role in Alzheimer's disease (AD) pathogenesis, impacting distinct processes such as amyloid-beta (Aβ) peptide production and tau phosphorylation. Impaired phosphorylation events contribute to senile plaques and neurofibrillary tangles' formation, two major histopathological hallmarks of AD. Blood-derived extracellular particles (bdEP) can represent a disease-related source of phosphobiomarker candidates, and hence, in this pilot study, bdEP of Control and AD cases were analyzed by a targeted phosphoproteomics approach using a high-density microarray that featured at least 1145 pan-specific and 913 phosphosite-specific antibodies. This approach, innovatively applied to bdEP, allowed the identification of 150 proteins whose expression levels and/or phosphorylation patterns were significantly altered across AD cases. Gene Ontology enrichment and Reactome pathway analysis unraveled potentially relevant molecular targets and disease-associated pathways, and protein-protein interaction networks were constructed to highlight key targets. The discriminatory value of both the total proteome and the phosphoproteome was evaluated by univariate and multivariate approaches. This pilot experiment supports that bdEP are enriched in phosphotargets relevant in an AD context, holding value as peripheral biomarker candidates for disease diagnosis.
Collapse
Affiliation(s)
- Tânia Soares Martins
- Neurosciences and Signaling Group, Institute of Biomedicine (iBiMED), Department of Medical Sciences, University of Aveiro, 3810-193 Aveiro, Portugal; (T.S.M.)
| | - Steven Pelech
- Department of Medicine, University of British Columbia, Vancouver, BC V5Z 1M9, Canada
- Kinexus Bioinformatics Corporation, Vancouver, BC V6P 6T3, Canada
| | - Maria Ferreira
- Neurosciences and Signaling Group, Institute of Biomedicine (iBiMED), Department of Medical Sciences, University of Aveiro, 3810-193 Aveiro, Portugal; (T.S.M.)
| | - Beatriz Pinho
- Neurosciences and Signaling Group, Institute of Biomedicine (iBiMED), Department of Medical Sciences, University of Aveiro, 3810-193 Aveiro, Portugal; (T.S.M.)
| | - Kevin Leandro
- Center for Neuroscience and Cell Biology, Faculty of Pharmacy, University of Coimbra, 3004-504 Coimbra, Portugal
- ViraVector–Viral Vector for Gene Transfer Core Facility, University of Coimbra, 3004-504 Coimbra, Portugal
| | - Luís Pereira de Almeida
- Center for Neuroscience and Cell Biology, Faculty of Pharmacy, University of Coimbra, 3004-504 Coimbra, Portugal
- ViraVector–Viral Vector for Gene Transfer Core Facility, University of Coimbra, 3004-504 Coimbra, Portugal
| | - Benedict Breitling
- Department of Psychiatry and Psychotherapy, University Medical Center Goettingen (UMG), Georg-August University, 37075 Goettingen, Germany
| | - Niels Hansen
- Department of Psychiatry and Psychotherapy, University Medical Center Goettingen (UMG), Georg-August University, 37075 Goettingen, Germany
| | - Hermann Esselmann
- Department of Psychiatry and Psychotherapy, University Medical Center Goettingen (UMG), Georg-August University, 37075 Goettingen, Germany
| | - Jens Wiltfang
- Neurosciences and Signaling Group, Institute of Biomedicine (iBiMED), Department of Medical Sciences, University of Aveiro, 3810-193 Aveiro, Portugal; (T.S.M.)
- Department of Psychiatry and Psychotherapy, University Medical Center Goettingen (UMG), Georg-August University, 37075 Goettingen, Germany
- German Center for Neurodegenerative Diseases (DZNE), 37075 Goettingen, Germany
| | - Odete A. B. da Cruz e Silva
- Neurosciences and Signaling Group, Institute of Biomedicine (iBiMED), Department of Medical Sciences, University of Aveiro, 3810-193 Aveiro, Portugal; (T.S.M.)
| | - Ana Gabriela Henriques
- Neurosciences and Signaling Group, Institute of Biomedicine (iBiMED), Department of Medical Sciences, University of Aveiro, 3810-193 Aveiro, Portugal; (T.S.M.)
| |
Collapse
|
9
|
Shi A, Liu L, Li S, Qi B. Natural products targeting the MAPK-signaling pathway in cancer: overview. J Cancer Res Clin Oncol 2024; 150:6. [PMID: 38193944 PMCID: PMC10776710 DOI: 10.1007/s00432-023-05572-7] [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: 10/24/2023] [Accepted: 11/17/2023] [Indexed: 01/10/2024]
Abstract
PURPOSE This article summarizes natural products that target the MAPK-signaling pathway in cancer therapy. The classification, chemical structures, and anti-cancer mechanisms of these natural products are elucidated, and comprehensive information is provided on their potential use in cancer therapy. METHODS Using the PubMed database, we searched for keywords, including "tumor", "cancer", "natural product", "phytochemistry", "plant chemical components", and "MAPK-signaling pathway". We also screened for compounds with well-defined structures that targeting the MAPK-signaling pathway and have anti-cancer effects. We used Kingdraw software and Adobe Photoshop software to draw the chemical compound structural diagrams. RESULTS A total of 131 papers were searched, from which 85 compounds with well-defined structures were selected. These compounds have clear mechanisms for targeting cancer treatment and are mainly related to the MAPK-signaling pathway. Examples include eupatilin, carvacrol, oridonin, sophoridine, diosgenin, and juglone. These chemical components are classified as flavonoids, phenols, terpenoids, alkaloids, steroidal saponins, and quinones. CONCLUSIONS Certain MAPK pathway inhibitors have been used for clinical treatment. However, the clinical feedback has not been promising because of genomic instability, drug resistance, and side effects. Natural products have few side effects, good medicinal efficacy, a wide range of sources, individual heterogeneity of biological activity, and are capable of treating disease from multiple targets. These characteristics make natural products promising drugs for cancer treatment.
Collapse
Affiliation(s)
- Aiwen Shi
- Changchun University of Chinese Medicine, School of Phharmacy, 1035 Boshuo Road, Jingyue Street, Nanguan District, Changchun City, Jilin Province, China
| | - Li Liu
- Changchun University of Chinese Medicine, School of Phharmacy, 1035 Boshuo Road, Jingyue Street, Nanguan District, Changchun City, Jilin Province, China.
| | - Shuang Li
- Changchun University of Chinese Medicine, School of Phharmacy, 1035 Boshuo Road, Jingyue Street, Nanguan District, Changchun City, Jilin Province, China
| | - Bin Qi
- Changchun University of Chinese Medicine, School of Phharmacy, 1035 Boshuo Road, Jingyue Street, Nanguan District, Changchun City, Jilin Province, China.
| |
Collapse
|
10
|
Gureghian V, Herbst H, Kozar I, Mihajlovic K, Malod-Dognin N, Ceddia G, Angeli C, Margue C, Randic T, Philippidou D, Nomigni MT, Hemedan A, Tranchevent LC, Longworth J, Bauer M, Badkas A, Gaigneaux A, Muller A, Ostaszewski M, Tolle F, Pržulj N, Kreis S. A multi-omics integrative approach unravels novel genes and pathways associated with senescence escape after targeted therapy in NRAS mutant melanoma. Cancer Gene Ther 2023; 30:1330-1345. [PMID: 37420093 PMCID: PMC10581906 DOI: 10.1038/s41417-023-00640-z] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2023] [Revised: 05/19/2023] [Accepted: 06/21/2023] [Indexed: 07/09/2023]
Abstract
Therapy Induced Senescence (TIS) leads to sustained growth arrest of cancer cells. The associated cytostasis has been shown to be reversible and cells escaping senescence further enhance the aggressiveness of cancers. Chemicals specifically targeting senescent cells, so-called senolytics, constitute a promising avenue for improved cancer treatment in combination with targeted therapies. Understanding how cancer cells evade senescence is needed to optimise the clinical benefits of this therapeutic approach. Here we characterised the response of three different NRAS mutant melanoma cell lines to a combination of CDK4/6 and MEK inhibitors over 33 days. Transcriptomic data show that all cell lines trigger a senescence programme coupled with strong induction of interferons. Kinome profiling revealed the activation of Receptor Tyrosine Kinases (RTKs) and enriched downstream signaling of neurotrophin, ErbB and insulin pathways. Characterisation of the miRNA interactome associates miR-211-5p with resistant phenotypes. Finally, iCell-based integration of bulk and single-cell RNA-seq data identifies biological processes perturbed during senescence and predicts 90 new genes involved in its escape. Overall, our data associate insulin signaling with persistence of a senescent phenotype and suggest a new role for interferon gamma in senescence escape through the induction of EMT and the activation of ERK5 signaling.
Collapse
Affiliation(s)
- Vincent Gureghian
- Department of Life Sciences and Medicine, University of Luxembourg, 6, Avenue du Swing, L-4367, Belvaux, Luxembourg
| | - Hailee Herbst
- Department of Life Sciences and Medicine, University of Luxembourg, 6, Avenue du Swing, L-4367, Belvaux, Luxembourg
| | - Ines Kozar
- Laboratoire National de Santé, Dudelange, Luxembourg
| | | | | | - Gaia Ceddia
- Barcelona Supercomputing Center, 08034, Barcelona, Spain
| | - Cristian Angeli
- Department of Life Sciences and Medicine, University of Luxembourg, 6, Avenue du Swing, L-4367, Belvaux, Luxembourg
| | - Christiane Margue
- Department of Life Sciences and Medicine, University of Luxembourg, 6, Avenue du Swing, L-4367, Belvaux, Luxembourg
| | - Tijana Randic
- Department of Life Sciences and Medicine, University of Luxembourg, 6, Avenue du Swing, L-4367, Belvaux, Luxembourg
| | - Demetra Philippidou
- Department of Life Sciences and Medicine, University of Luxembourg, 6, Avenue du Swing, L-4367, Belvaux, Luxembourg
| | - Milène Tetsi Nomigni
- Department of Life Sciences and Medicine, University of Luxembourg, 6, Avenue du Swing, L-4367, Belvaux, Luxembourg
| | - Ahmed Hemedan
- Luxembourg Centre for Systems Biomedicine, University of Luxembourg, Esch-sur-Alzette, Luxembourg
| | - Leon-Charles Tranchevent
- Luxembourg Centre for Systems Biomedicine, University of Luxembourg, Esch-sur-Alzette, Luxembourg
| | - Joseph Longworth
- Experimental and Molecular Immunology, Department of Infection and Immunity, Luxembourg Institute of Health, Esch-sur-Alzette, Luxembourg
| | - Mark Bauer
- Department of Life Sciences and Medicine, University of Luxembourg, 6, Avenue du Swing, L-4367, Belvaux, Luxembourg
| | - Apurva Badkas
- Department of Life Sciences and Medicine, University of Luxembourg, 6, Avenue du Swing, L-4367, Belvaux, Luxembourg
| | - Anthoula Gaigneaux
- Department of Life Sciences and Medicine, University of Luxembourg, 6, Avenue du Swing, L-4367, Belvaux, Luxembourg
| | - Arnaud Muller
- LuxGen, TMOH and Bioinformatics platform, Data Integration and Analysis unit, Luxembourg Institute of Health, Esch-sur-Alzette, Luxembourg
| | - Marek Ostaszewski
- Luxembourg Centre for Systems Biomedicine, University of Luxembourg, Esch-sur-Alzette, Luxembourg
| | - Fabrice Tolle
- Department of Life Sciences and Medicine, University of Luxembourg, 6, Avenue du Swing, L-4367, Belvaux, Luxembourg
| | - Nataša Pržulj
- Barcelona Supercomputing Center, 08034, Barcelona, Spain
- Department of Computer Science, University College London, London, WC1E 6BT, UK
- ICREA, Pg. Lluís Companys 23, 08010, Barcelona, Spain
| | - Stephanie Kreis
- Department of Life Sciences and Medicine, University of Luxembourg, 6, Avenue du Swing, L-4367, Belvaux, Luxembourg.
| |
Collapse
|
11
|
Hwang J, Moon H, Kim H, Kim KY. Identification of a Novel ERK5 (MAPK7) Inhibitor, MHJ-627, and Verification of Its Potent Anticancer Efficacy in Cervical Cancer HeLa Cells. Curr Issues Mol Biol 2023; 45:6154-6169. [PMID: 37504304 PMCID: PMC10377775 DOI: 10.3390/cimb45070388] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2023] [Revised: 07/19/2023] [Accepted: 07/21/2023] [Indexed: 07/29/2023] Open
Abstract
Extracellular signal-regulated kinase 5 (ERK5), a member of the mitogen-activated protein kinase (MAPK) family, is involved in key cellular processes. However, overexpression and upregulation of ERK5 have been reported in various cancers, and ERK5 is associated with almost every biological characteristic of cancer cells. Accordingly, ERK5 has become a novel target for the development of anticancer drugs as inhibition of ERK5 shows suppressive effects of the deleterious properties of cancer cells. Herein, we report the synthesis and identification of a novel ERK5 inhibitor, MHJ-627, and verify its potent anticancer efficacy in a yeast model and the cervical cancer HeLa cell line. MHJ-627 successfully inhibited the kinase activity of ERK5 (IC50: 0.91 μM) and promoted the mRNA expression of tumor suppressors and anti-metastatic genes. Moreover, we observed significant cancer cell death, accompanied by a reduction in mRNA levels of the cell proliferation marker, proliferating cell nuclear antigen (PCNA), following ERK5 inhibition due to MHJ-627 treatment. We expect this finding to serve as a lead compound for further identification of inhibitors for ERK5-directed novel approaches for oncotherapy with increased specificity.
Collapse
Affiliation(s)
- Jeonghye Hwang
- Department of Genetics and Biotechnology, Kyung Hee University, Yongin 17104, Republic of Korea
| | - Hyejin Moon
- Department of Applied Chemistry, Global Center for Pharmaceutical Ingredient Materials, Kyung Hee University, Yongin 17104, Republic of Korea
| | - Hakwon Kim
- Department of Applied Chemistry, Global Center for Pharmaceutical Ingredient Materials, Kyung Hee University, Yongin 17104, Republic of Korea
| | - Ki-Young Kim
- Department of Genetics and Biotechnology, Kyung Hee University, Yongin 17104, Republic of Korea
- Graduate School of Biotechnology, Kyung Hee University, Yongin 17104, Republic of Korea
| |
Collapse
|
12
|
Sánchez-Fdez A, Matilla-Almazán S, Del Carmen S, Abad M, Arconada-Luque E, Jiménez-Suárez J, Chinchilla-Tábora LM, Ruíz-Hidalgo MJ, Sánchez-Prieto R, Pandiella A, Esparís-Ogando A. Etiopathogenic role of ERK5 signaling in sarcoma: prognostic and therapeutic implications. Exp Mol Med 2023; 55:1247-1257. [PMID: 37332046 PMCID: PMC10317974 DOI: 10.1038/s12276-023-01008-x] [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: 08/09/2022] [Revised: 02/28/2023] [Accepted: 03/07/2023] [Indexed: 06/20/2023] Open
Abstract
Sarcomas constitute a heterogeneous group of rare and difficult-to-treat tumors that can affect people of all ages, representing one of the most common forms of cancer in childhood and adolescence. Little is known about the molecular entities involved in sarcomagenesis. Therefore, the identification of processes that lead to the development of the disease may uncover novel therapeutic opportunities. Here, we show that the MEK5/ERK5 signaling pathway plays a critical role in the pathogenesis of sarcomas. By developing a mouse model engineered to express a constitutively active form of MEK5, we demonstrate that the exclusive activation of the MEK5/ERK5 pathway can promote sarcomagenesis. Histopathological analyses identified these tumors as undifferentiated pleomorphic sarcomas. Bioinformatic studies revealed that sarcomas are the tumors in which ERK5 is most frequently amplified and overexpressed. Moreover, analysis of the impact of ERK5 protein expression on overall survival in patients diagnosed with different sarcoma types in our local hospital showed a 5-fold decrease in median survival in patients with elevated ERK5 expression compared with those with low expression. Pharmacological and genetic studies revealed that targeting the MEK5/ERK5 pathway drastically affects the proliferation of human sarcoma cells and tumor growth. Interestingly, sarcoma cells with knockout of ERK5 or MEK5 were unable to form tumors when engrafted into mice. Taken together, our results reveal a role of the MEK5/ERK5 pathway in sarcomagenesis and open a new scenario to be considered in the treatment of patients with sarcoma in which the ERK5 pathway is pathophysiologically involved.
Collapse
Affiliation(s)
- Adrián Sánchez-Fdez
- Instituto de Investigación Biomédica de Salamanca (IBSAL), Salamanca, Spain
- Instituto de Biología Molecular y Celular del Cáncer (IBMCC)-CSIC, Salamanca, Spain
- Centro de Investigación Biomédica en Red de Cáncer (CIBERONC), CSIC-Universidad de Salamanca, Salamanca, Spain
- Moores Cancer Center, University of California San Diego, La Jolla, CA, USA
| | - Sofía Matilla-Almazán
- Instituto de Investigación Biomédica de Salamanca (IBSAL), Salamanca, Spain
- Instituto de Biología Molecular y Celular del Cáncer (IBMCC)-CSIC, Salamanca, Spain
- Centro de Investigación Biomédica en Red de Cáncer (CIBERONC), CSIC-Universidad de Salamanca, Salamanca, Spain
| | - Sofía Del Carmen
- Instituto de Investigación Biomédica de Salamanca (IBSAL), Salamanca, Spain
- Departmento de Patología, Hospital Universitario de Salamanca, Universidad de Salamanca, Salamanca, Spain
| | - Mar Abad
- Instituto de Investigación Biomédica de Salamanca (IBSAL), Salamanca, Spain
- Departmento de Patología, Hospital Universitario de Salamanca, Universidad de Salamanca, Salamanca, Spain
| | - Elena Arconada-Luque
- Universidad de Castilla-La Mancha, Laboratorio de Oncología Molecular, Unidad de Medicina Molecular, Centro Regional de Investigaciones Biomédicas, Unidad Asociada de Biomedicina UCLM, Unidad asociada al CSIC, Albacete, Spain
| | - Jaime Jiménez-Suárez
- Universidad de Castilla-La Mancha, Laboratorio de Oncología Molecular, Unidad de Medicina Molecular, Centro Regional de Investigaciones Biomédicas, Unidad Asociada de Biomedicina UCLM, Unidad asociada al CSIC, Albacete, Spain
| | - Luis Miguel Chinchilla-Tábora
- Instituto de Investigación Biomédica de Salamanca (IBSAL), Salamanca, Spain
- Departmento de Patología, Hospital Universitario de Salamanca, Universidad de Salamanca, Salamanca, Spain
| | - Mª José Ruíz-Hidalgo
- Universidad de Castilla-La Mancha, Laboratorio de Oncología Molecular, Unidad de Medicina Molecular, Centro Regional de Investigaciones Biomédicas, Unidad Asociada de Biomedicina UCLM, Unidad asociada al CSIC, Albacete, Spain
- Universidad de Castilla-La Mancha, Departamento de Química Inorgánica, Orgánica y Bioquímica, Área de Bioquímica y Biología Molecular. Facultad de Medicina, Albacete, Spain
| | - Ricardo Sánchez-Prieto
- Universidad de Castilla-La Mancha, Laboratorio de Oncología Molecular, Unidad de Medicina Molecular, Centro Regional de Investigaciones Biomédicas, Unidad Asociada de Biomedicina UCLM, Unidad asociada al CSIC, Albacete, Spain
- Universidad de Castilla-La Mancha, Departamento de Ciencias Médicas, Facultad de Medicina, Albacete, Spain
- Departamento de Biología del Cáncer, Instituto de Investigaciones Biomédicas 'Alberto Sols' (CSIC-UAM), Unidad Asociada de Biomedicina UCLM, Unidad Asociada al CSIC, Madrid, Spain
- Instituto de Investigaciones Biomédicas 'Alberto Sols', Consejo Superior de Investigaciones Científicas (IIBM-CSIC)-Universidad de Castilla-La Mancha (UCLM), Albacete, Spain
| | - Atanasio Pandiella
- Instituto de Investigación Biomédica de Salamanca (IBSAL), Salamanca, Spain
- Instituto de Biología Molecular y Celular del Cáncer (IBMCC)-CSIC, Salamanca, Spain
- Centro de Investigación Biomédica en Red de Cáncer (CIBERONC), CSIC-Universidad de Salamanca, Salamanca, Spain
| | - Azucena Esparís-Ogando
- Instituto de Investigación Biomédica de Salamanca (IBSAL), Salamanca, Spain.
- Instituto de Biología Molecular y Celular del Cáncer (IBMCC)-CSIC, Salamanca, Spain.
- Centro de Investigación Biomédica en Red de Cáncer (CIBERONC), CSIC-Universidad de Salamanca, Salamanca, Spain.
| |
Collapse
|
13
|
López-Borrego S, Campos-Silva C, Sandúa A, Camino T, Téllez-Pérez L, Alegre E, Beneitez A, Jara-Acevedo R, Paschen A, Pardo M, González Á, Valés-Gómez M. MAPK inhibitors dynamically affect melanoma release of immune NKG2D-ligands, as soluble protein and extracellular vesicle-associated. Front Cell Dev Biol 2023; 10:1055288. [PMID: 36726591 PMCID: PMC9884675 DOI: 10.3389/fcell.2022.1055288] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2022] [Accepted: 12/13/2022] [Indexed: 01/18/2023] Open
Abstract
Metastatic melanoma presents, in many cases, oncogenic mutations in BRAF, a MAPK involved in proliferation of tumour cells. BRAF inhibitors, used as therapy in patients with these mutations, often lead to tumour resistance and, thus, the use of MEK inhibitors was introduced in clinics. BRAFi/MEKi, a combination that has modestly increased overall survival in patients, has been proven to differentially affect immune ligands, such as NKG2D-ligands, in drug-sensitive vs. drug-resistant cells. However, the fact that NKG2D-ligands can be released as soluble molecules or in extracellular vesicles represents an additional level of complexity that has not been explored. Here we demonstrate that inhibition of MAPK using MEKi, and the combination of BRAFi with MEKi in vitro, modulates NKG2D-ligands in BRAF-mutant and WT melanoma cells, together with other NK activating ligands. These observations reinforce a role of the immune system in the generation of resistance to directed therapies and support the potential benefit of MAPK inhibition in combination with immunotherapies. Both soluble and EV-associated NKG2D-ligands, generally decreased in BRAF-mutant melanoma cell supernatants after MAPKi in vitro, replicating cell surface expression. Because potential NKG2D-ligand fluctuation during MAPKi treatment could have different consequences for the immune response, a pilot study to measure NKG2D-ligand variation in plasma or serum from metastatic melanoma patients, at different time points during MAPKi treatment, was performed. Not all NKG2D-ligands were equally detected. Further, EV detection did not parallel soluble protein. Altogether, our data confirm the heterogeneity between melanoma lesions, and suggest testing several NKG2D-ligands and other melanoma antigens in serum, both as soluble or vesicle-released proteins, to help classifying immune competence of patients.
Collapse
Affiliation(s)
- Silvia López-Borrego
- Department of Immunology and Oncology, National Center for Biotechnology (CNB), Spanish National Research Council (CSIC), Cantoblanco, Madrid, Spain
| | - Carmen Campos-Silva
- Department of Immunology and Oncology, National Center for Biotechnology (CNB), Spanish National Research Council (CSIC), Cantoblanco, Madrid, Spain
| | | | - Tamara Camino
- Health Research Institute of Santiago de Compostela (IDIS), Santiago de Compostela, Galicia, Spain
| | - Lucía Téllez-Pérez
- Department of Immunology and Oncology, National Center for Biotechnology (CNB), Spanish National Research Council (CSIC), Cantoblanco, Madrid, Spain
| | | | | | | | - Annette Paschen
- Clinic for Dermatology University Hospital of Essen, Essen, North RhineWestphalia, Germany
| | - María Pardo
- Health Research Institute of Santiago de Compostela (IDIS), Santiago de Compostela, Galicia, Spain
| | | | - Mar Valés-Gómez
- Department of Immunology and Oncology, National Center for Biotechnology (CNB), Spanish National Research Council (CSIC), Cantoblanco, Madrid, Spain,*Correspondence: Mar Valés-Gómez,
| |
Collapse
|
14
|
Ma W, Han X, Shasaltaneh MD, Hosseinifard H, Maghsoudloo M, Zhang Y, Weng Q, Wang Q, Wen Q, Imani S. The p110α/ΔNp63α complex mutations in triple-negative breast cancer: Potential targets for transcriptional-based therapies. Tumour Biol 2023; 45:127-146. [PMID: 37980588 DOI: 10.3233/tub-230013] [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] [Indexed: 11/21/2023] Open
Abstract
BACKGROUND Hotspot mutations occurring in the p110α domain of the PIK3CA gene, specifically p110αH1047R/L increase tumor metastasis and cell motility in triple-negative breast cancer (TNBC). These mutations also affect the transcriptional regulation of ΔNp63α, a significant isoform of the p53 protein involved in cancer progression. This study attempts to investigate the transcriptional impact of p110αH1047R/L mutations on the PIK3CA/ΔNp63α complex in TNBC carcinogenesis. METHODS We performed site-directed mutagenesis to introduce p110αH1047R/L mutations and evaluated their oncogenic effects on the growth, invasion, migration, and apoptosis of three different TNBC cell lines in vitro. We investigated the impact of these mutations on the p110α/ΔNp63α complex and downstream transcriptional signaling pathways at the gene and protein levels. Additionally, we used bioinformatics techniques such as molecular dynamics simulations and protein-protein docking to gain insight into the stability and structural changes induced by the p110αH1047R/L mutations in the p110α/ΔNp63α complex and downstream signaling pathway. RESULTS The presence of PIK3CA oncogenic hotspot mutations in the p110α/ΔNp63α complex led to increased scattering of TNBC cells during growth, migration, and invasion. Our in vitro mutagenesis assay showed that the p110αH1047R/L mutations activated the PI3K-Akt-mTOR and tyrosine kinase receptor pathways, resulting in increased cell proliferation, invasion, and apoptosis in TNBC cells. These mutations decreased the repressing effect of ΔNp63α on the p110α kinase domain, leading to the enhancement of downstream signaling pathways of PI3K and tyrosine kinase receptors and oncogenic transformation in TNBC. Additionally, our findings suggest that the physical interaction between the DNA binding domain of ΔNp63α and the kinase domain of p110α may be partially impaired, potentially leading to alterations in the conformation of the p110α/ΔNp63α complex. CONCLUSION Our findings suggest that targeting the p110αH1047R/L mutations in TNBC could be a promising strategy for developing transcriptional-based therapies. Restoring the interaction between ΔNp63α and the p110α kinase domain, which is disrupted by these mutations, may provide a new approach to treating TNBC.
Collapse
Affiliation(s)
- Wenqiong Ma
- Department of Oncology, The Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan, China
| | - Xingping Han
- Department of Oncology, The Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan, China
| | | | - Hossein Hosseinifard
- Department of Biostatistics, School of Public Health, Hamadan University of Medical Sciences, Hamadan, Iran
| | - Mazaher Maghsoudloo
- Key Laboratory of Epigenetics and Oncology, the Research Center for Preclinical Medicine, Southwest Medical University, Luzhou, Sichuan, China
| | - Yuqin Zhang
- Department of Oncology, The Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan, China
| | - Qiao Weng
- Department of Obstetrics & Gynecology, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing, China
| | - Qingjing Wang
- Shulan International Medical College, Zhejiang Shuren University, Hangzhou, Zhejiang, China
| | - QingLian Wen
- Department of Oncology, The Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan, China
| | - Saber Imani
- Shulan International Medical College, Zhejiang Shuren University, Hangzhou, Zhejiang, China
| |
Collapse
|