1
|
Kagemichi N, Umemura M, Ishikawa S, Iida Y, Takayasu S, Nagasako A, Nakakaji R, Akimoto T, Ohtake M, Horinouchi T, Yamamoto T, Ishikawa Y. Cytotoxic effects of the cigarette smoke extract of heated tobacco products on human oral squamous cell carcinoma: the role of reactive oxygen species and CaMKK2. J Physiol Sci 2024; 74:35. [PMID: 38918702 PMCID: PMC11197199 DOI: 10.1186/s12576-024-00928-1] [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: 03/26/2024] [Accepted: 06/08/2024] [Indexed: 06/27/2024]
Abstract
BACKGROUND The increasing prevalence of heated tobacco products (HTPs) has heightened concerns regarding their potential health risks. Previous studies have demonstrated the toxicity of cigarette smoke extract (CSE) from traditional tobacco's mainstream smoke, even after the removal of nicotine and tar. Our study aimed to investigate the cytotoxicity of CSE derived from HTPs and traditional tobacco, with a particular focus on the role of reactive oxygen species (ROS) and intracellular Ca2+. METHODS A human oral squamous cell carcinoma (OSCC) cell line, HSC-3 was utilized. To prepare CSE, aerosols from HTPs (IQOS) and traditional tobacco products (1R6F reference cigarette) were collected into cell culture media. A cell viability assay, apoptosis assay, western blotting, and Fluo-4 assay were conducted. Changes in ROS levels were measured using electron spin resonance spectroscopy and the high-sensitivity 2',7'-dichlorofluorescein diacetate assay. We performed a knockdown of calcium/calmodulin-dependent protein kinase kinase 2 (CaMKK2) by shRNA lentivirus in OSCC cells. RESULTS CSE from both HTPs and traditional tobacco exhibited cytotoxic effects in OSCC cells. Exposure to CSE from both sources led to an increase in intracellular Ca2+ concentration and induced p38 phosphorylation. Additionally, these extracts prompted cell apoptosis and heightened ROS levels. N-acetylcysteine (NAC) mitigated the cytotoxic effects and p38 phosphorylation. Furthermore, the knockdown of CaMKK2 in HSC-3 cells reduced cytotoxicity, ROS production, and p38 phosphorylation in response to CSE. CONCLUSION Our findings suggest that the CSE from both HTPs and traditional tobacco induce cytotoxicity. This toxicity is mediated by ROS, which are regulated through Ca2+ signaling and CaMKK2 pathways.
Collapse
Affiliation(s)
- Nagao Kagemichi
- Cardiovascular Research Institute, Yokohama City University Graduate School of Medicine, Yokohama, Kanagawa, Japan
- Neurosurgery, Yokohama City University Graduate School of Medicine, Yokohama, Kanagawa, Japan
| | - Masanari Umemura
- Cardiovascular Research Institute, Yokohama City University Graduate School of Medicine, Yokohama, Kanagawa, Japan.
| | - Soichiro Ishikawa
- Cardiovascular Research Institute, Yokohama City University Graduate School of Medicine, Yokohama, Kanagawa, Japan
- Oral and Maxillofacial Surgery, Yokohama City University Graduate School of Medicine, Yokohama, Kanagawa, Japan
| | - Yu Iida
- Cardiovascular Research Institute, Yokohama City University Graduate School of Medicine, Yokohama, Kanagawa, Japan
- Neurosurgery, Yokohama City University Graduate School of Medicine, Yokohama, Kanagawa, Japan
| | - Shota Takayasu
- Cardiovascular Research Institute, Yokohama City University Graduate School of Medicine, Yokohama, Kanagawa, Japan
| | - Akane Nagasako
- Cardiovascular Research Institute, Yokohama City University Graduate School of Medicine, Yokohama, Kanagawa, Japan
| | - Rina Nakakaji
- Cardiovascular Research Institute, Yokohama City University Graduate School of Medicine, Yokohama, Kanagawa, Japan
- Oral and Maxillofacial Surgery, Yokohama City University Graduate School of Medicine, Yokohama, Kanagawa, Japan
| | - Taisuke Akimoto
- Neurosurgery, Yokohama City University Graduate School of Medicine, Yokohama, Kanagawa, Japan
| | - Makoto Ohtake
- Neurosurgery, Yokohama City University Graduate School of Medicine, Yokohama, Kanagawa, Japan
| | - Takahiro Horinouchi
- Cellular Pharmacology, Hokkaido University Graduate School of Medicine, Sapporo, Hokkaido, Japan
| | - Tetsuya Yamamoto
- Neurosurgery, Yokohama City University Graduate School of Medicine, Yokohama, Kanagawa, Japan
| | - Yoshihiro Ishikawa
- Cardiovascular Research Institute, Yokohama City University Graduate School of Medicine, Yokohama, Kanagawa, Japan.
| |
Collapse
|
2
|
Rahman SMT, Zhou W, Deiters A, Haugh JM. Dissection of MKK6 and p38 Signaling Using Light-Activated Protein Kinases. Chembiochem 2024; 25:e202300551. [PMID: 37856284 DOI: 10.1002/cbic.202300551] [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: 08/04/2023] [Revised: 10/18/2023] [Accepted: 10/18/2023] [Indexed: 10/21/2023]
Abstract
Stress-activated signaling pathways orchestrate cellular behaviors and fates. Studying the precise role(s) of stress-activated protein kinases is challenging, because stress conditions induce adaptation and impose selection pressure. To meet this challenge, we have applied an optogenetic system with a single plasmid to express light-activated p38α or its upstream activator, MKK6, in conjunction with live-cell fluorescence microscopy. In starved cells, decaging of constitutively active p38α or MKK6 by brief exposure to UV light elicits rapid p38-mediated signaling, release of cytochrome c from mitochondria, and apoptosis with different kinetics. In parallel, light activation of p38α also suppresses autophagosome formation, similarly to stimulation with growth factors that activate PI3K/Akt/mTORC1 signaling. Active MKK6 negatively regulates serum-induced ERK activity, which is p38-independent as previously reported. Here, we reproduce that result with the one plasmid system and show that although decaging active p38α does not reduce basal ERK activity in our cells, it can block growth factor-stimulated ERK signaling in serum-starved cells. These results clarify the roles of MKK6 and p38α in dynamic signaling programs, which act in concert to actuate apoptotic death while suppressing cell survival mechanisms.
Collapse
Affiliation(s)
- Shah Md Toufiqur Rahman
- Department of Chemical and Biomolecular Engineering, North Carolina State University, Campus Box 7905, 911 Partners Way, Raleigh, NC, 27695, USA
| | - Wenyuan Zhou
- Department of Chemistry, University of Pittsburgh, Pittsburgh, PA, 15260, USA
| | - Alexander Deiters
- Department of Chemistry, University of Pittsburgh, Pittsburgh, PA, 15260, USA
| | - Jason M Haugh
- Department of Chemical and Biomolecular Engineering, North Carolina State University, Campus Box 7905, 911 Partners Way, Raleigh, NC, 27695, USA
| |
Collapse
|
3
|
Alatawi FS, Faridi U. Anticancer and anti-metastasis activity of 1,25 dihydroxycholecalciferols and genistein in MCF-7 and MDA-MB-231 breast cancer cell lines. Heliyon 2023; 9:e21975. [PMID: 38034665 PMCID: PMC10682641 DOI: 10.1016/j.heliyon.2023.e21975] [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: 11/28/2022] [Revised: 11/01/2023] [Accepted: 11/01/2023] [Indexed: 12/02/2023] Open
Abstract
A powerful steroid hormone precursor, 1,25 dihydroxycholecalciferols (1,25(OH)2D3), and dietary phytoestrogen (genistein) are essential compounds that act by binding to nuclear receptors and altering gene expression. They have many biological benefits, some of which have anticancer properties. We studied the impact of 1,25(OH)2D3 and genistein on the proliferation, progression, and metastasis of MCF-7 and MDA-MB-231 cells when they were used alone or in combination and investigated whether there was a synergistic effect between genistein and 1,25(OH)2D3. To achieve these goals, a variety of assays, including flow cytometry, cell invasion assays, cell adhesion assays, Western blotting, and RT‒PCR, were used. Our findings showed that genistein, 1,25(OH)2D3, and the two combined all effectively declined the growth of MCF-7 and MDA-MB-231 cells by arresting the cells in the G0/G1 phase and inducing an apoptotic pathway. Stimulation of apoptosis was achieved by upregulating the expression of BAX and CASP3 genes and downregulating the expression levels of BCL-2 gene. Furthermore, both compounds suppress metastasis by reducing cell adhesion and cell migration/invasion by elevating the expression level of E-cadherin and reducing the expression level of P-cadherin and N-cadherin. Additionally, both genistein and 1,25(OH)2D3 increased the expression level of ERK1 and reduced the expression levels of JNK, p38, Ras, and MEK proteins, which reduced metastasis, enhanced the response to cancer treatment, and improved overall survival. Thus, genistein and 1,25(OH)2D3 can both be considered key candidates in the search for new breast cancer treatments.
Collapse
Affiliation(s)
- Fatema Suliman Alatawi
- Faculty of Sciences, Biochemistry Department, Science College, University of Tabuk, Tabuk Saudi Arabia
| | - Uzma Faridi
- Faculty of Sciences, Biochemistry Department, Science College, University of Tabuk, Tabuk Saudi Arabia
| |
Collapse
|
4
|
Natarajan P, Manne M, Koduru SK, Bokkasam TS. 3-deazaadenosine: A promising novel p38γ antagonist with potential as a breast cancer therapeutic agent. Cancer Treat Res Commun 2023; 36:100744. [PMID: 37481995 DOI: 10.1016/j.ctarc.2023.100744] [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: 05/31/2023] [Revised: 07/07/2023] [Accepted: 07/13/2023] [Indexed: 07/25/2023]
Abstract
Human p38γ protein kinase, or MAPK12, is a crucial signaling protein that is important in channelizing membrane signals to the nucleus in the MAPK cascade pathway, associated with breast and colorectal cancer, besides other forms of malignancies and atherosclerotic lesions too. P38γ has a significant contribution to the progression of breast carcinoma due to its multifaceted functions. Targeting p38γ for defining potent antagonists against p38γ can turn out to be an attractive and novel means of breast cancer therapeutics. Novel and potent lead molecules were designed utilizing computational drug design methodologies. Using high-throughput virtual screening, 1909 geometrically similar analogs of known inhibitors were generated, primarily using BIRB796, SB202190, ANP, CHEBI: 620708, and CHEBI: 524699. Chemical correctness was ensured using LigPrep for the standalone library, and Prep Wizard for p38γ using Maestro v.11.5. Using the Glide v5.5 flexible docking procedure on a standalone library of p38γ binding sites, we defined 18 potential leads and assessed their ADMET properties. Lead "1", among the proposed four p38γ antagonists with high-scoring and favorable interactions, was considered for 100 ns molecular dynamics simulations. Among the four proposed leads, Lead '1' displayed consistent and stable bonding interactions with p38γ throughout the 100 ns molecular dynamics (MD) simulations. Additionally, it formed water bridges, contributing to its strong association with the protein. Notably, Lead '1' (3-deazaadenosine) exhibited favorable root-mean-square deviation (RMSD) and root-mean-square fluctuation (RMSF) within the acceptable range of pharmacological properties. Thus, 3-deazaadenosine and its mimetic might be promising new directions for developing a novel class of antagonists for breast cancer treatment.
Collapse
Affiliation(s)
- Pradeep Natarajan
- Bioinformatics Center, Department of Biotechnology, Anna University, Chennai, Tamil Nadu 600025, India.
| | - Munikumar Manne
- Clinical Division, ICMR-National Institute of Nutrition, Jamai-Osmania (Post), Hyderabad, 500007 Telangana, India.
| | - Swetha Kumari Koduru
- Department of Bio-sciences and Sericulture, Sri Padmavati Mahila Visvavidyalayam Women's University, Tirupati, Andhra Pradesh 517502, India
| | - Teja Sree Bokkasam
- Department of Biotechnology, Sri Padmavati Mahila Visvavidyalayam Women's University, Tirupati, Andhra Pradesh 517502, India
| |
Collapse
|
5
|
Zhang NZ, Zhao LF, Zhang Q, Fang H, Song WL, Li WZ, Ge YS, Gao P. Core fucosylation and its roles in gastrointestinal glycoimmunology. World J Gastrointest Oncol 2023; 15:1119-1134. [PMID: 37546555 PMCID: PMC10401475 DOI: 10.4251/wjgo.v15.i7.1119] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/18/2023] [Revised: 02/28/2023] [Accepted: 05/08/2023] [Indexed: 07/12/2023] Open
Abstract
Glycosylation is a common post-translational modification in eukaryotic cells. It is involved in the production of many biologically active glycoproteins and the regulation of protein structure and function. Core fucosylation plays a vital role in the immune response. Most immune system molecules are core fucosylated glycoproteins such as complements, cluster differentiation antigens, immunoglobulins, cytokines, major histocompatibility complex molecules, adhesion molecules, and immune molecule synthesis-related transcription factors. These core fucosylated glycoproteins play important roles in antigen recognition and clearance, cell adhesion, lymphocyte activation, apoptosis, signal transduction, and endocytosis. Core fucosylation is dominated by fucosyltransferase 8 (Fut8), which catalyzes the addition of α-1,6-fucose to the innermost GlcNAc residue of N-glycans. Fut8 is involved in humoral, cellular, and mucosal immunity. Tumor immunology is associated with aberrant core fucosylation. Here, we summarize the roles and potential modulatory mechanisms of Fut8 in various immune processes of the gastrointestinal system.
Collapse
Affiliation(s)
- Nian-Zhu Zhang
- Clinical Laboratory, The Second Hospital of Dalian Medical University, Dalian 116023, Liaoning Province, China
| | - Li-Fen Zhao
- Clinical Laboratory, The Second Hospital of Dalian Medical University, Dalian 116023, Liaoning Province, China
| | - Qian Zhang
- Department of Cell Therapy, Shanghai Tianze Yuntai Biomedical Co., Ltd., Shanghai 200100, China
| | - Hui Fang
- Graduate School of Comprehensive Human Sciences, University of Tsukuba, Tsukuba 305-0005, Ibaraki, Japan
| | - Wan-Li Song
- Guangdong Provincial Key Laboratory of Infectious Diseases and Molecular Immunopathology, Shantou University Medical College, Shantou 515041, Guangdong Province, China
| | - Wen-Zhe Li
- Guangdong Provincial Key Laboratory of Infectious Diseases and Molecular Immunopathology, Shantou University Medical College, Shantou 515041, Guangdong Province, China
| | - Yu-Song Ge
- Department of Neurology, The Second Hospital of Dalian Medical University, Dalian 116023, Liaoning Province, China
| | - Peng Gao
- Clinical Laboratory, The Second Hospital of Dalian Medical University, Dalian 116023, Liaoning Province, China
| |
Collapse
|
6
|
Hankittichai P, Thaklaewphan P, Wikan N, Ruttanapattanakul J, Potikanond S, Smith DR, Nimlamool W. Resveratrol Enhances Cytotoxic Effects of Cisplatin by Inducing Cell Cycle Arrest and Apoptosis in Ovarian Adenocarcinoma SKOV-3 Cells through Activating the p38 MAPK and Suppressing AKT. Pharmaceuticals (Basel) 2023; 16:ph16050755. [PMID: 37242538 DOI: 10.3390/ph16050755] [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: 03/23/2023] [Revised: 05/12/2023] [Accepted: 05/15/2023] [Indexed: 05/28/2023] Open
Abstract
In the current study, we identified a mechanism of resveratrol (RES) underlying its anti-cancer properties against human ovarian adenocarcinoma SKOV-3 cells. We investigated its anti-proliferative and apoptosis-inducing effects in combination with cisplatin, using cell viability assay, flow cytometry, immunofluorescence study and Western blot analysis. We discovered that RES suppressed cancer cell proliferation and stimulated apoptosis, especially when combined with cisplatin. This compound also inhibited SKOV-3 cell survival, which may partly be due to its potential to inhibit protein kinase B (AKT) phosphorylation and induce the S-phase cell cycle arrest. RES in combination with cisplatin strongly induced cancer cell apoptosis through activating the caspase-dependent cascade, which was associated with its ability to stimulate nuclear phosphorylation of p38 mitogen-activated protein kinase (MAPK), well recognized to be involved in transducing environmental stress signals. RES-induced p38 phosphorylation was very specific, and the activation status of extracellular signal-regulated kinase 1/2 (ERK1/2) and c-Jun N-terminal kinase (JNK) was not mainly affected. Taken together, our study provides accumulated evidence that RES represses proliferation and promotes apoptosis in SKOV-3 ovarian cancer cells through activating the p38 MAPK pathway. It is interesting that this active compound may be used as an effective agent to sensitize ovarian cancer to apoptosis induced by standard chemotherapies.
Collapse
Affiliation(s)
- Phateep Hankittichai
- Department of Pharmacology, Faculty of Medicine, Chiang Mai University, Chiang Mai 50200, Thailand
| | - Phatarawat Thaklaewphan
- Department of Pharmacology, Faculty of Medicine, Chiang Mai University, Chiang Mai 50200, Thailand
| | - Nitwara Wikan
- Department of Pharmacology, Faculty of Medicine, Chiang Mai University, Chiang Mai 50200, Thailand
| | | | - Saranyapin Potikanond
- Department of Pharmacology, Faculty of Medicine, Chiang Mai University, Chiang Mai 50200, Thailand
- Research Center of Pharmaceutical Nanotechnology, Faculty of Pharmacy, Chiang Mai University, Chiang Mai 50200, Thailand
| | - Duncan R Smith
- Institute of Molecular Biosciences, Mahidol University, Salaya, Nakhon Pathom 73170, Thailand
| | - Wutigri Nimlamool
- Department of Pharmacology, Faculty of Medicine, Chiang Mai University, Chiang Mai 50200, Thailand
- Research Center of Pharmaceutical Nanotechnology, Faculty of Pharmacy, Chiang Mai University, Chiang Mai 50200, Thailand
| |
Collapse
|
7
|
Luo L, Pasquali L, Srivastava A, Freisenhausen JC, Pivarcsi A, Sonkoly E. The Long Noncoding RNA LINC00958 Is Induced in Psoriasis Epidermis and Modulates Epidermal Proliferation. J Invest Dermatol 2023; 143:999-1010. [PMID: 36641130 DOI: 10.1016/j.jid.2022.12.011] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2022] [Revised: 12/07/2022] [Accepted: 12/09/2022] [Indexed: 01/13/2023]
Abstract
Psoriasis is a common, immune-mediated skin disease characterized by epidermal hyperproliferation and chronic skin inflammation. Long noncoding RNAs are >200 nucleotide-long transcripts that possess important regulatory functions. To date, little is known about the contribution of long noncoding RNAs to psoriasis. In this study, we identify LINC00958 as a long noncoding RNA overexpressed in keratinocytes (KCs) from psoriasis skin lesions, in a transcriptomic screen performed on KCs sorted from psoriasis and healthy skin. Increased levels of LINC00958 in psoriasis KCs were confirmed by RT-qPCR and single-molecule in situ hybridization. Confocal microscopy and analysis of subcellular fractions showed that LINC00958 is mainly localized in the cytoplasm of KCs. IL-17A, a key psoriasis cytokine, induced LINC00958 in KCs through C/EBP-β and the p38 pathway. The inhibition of LINC00958 led to decreased proliferation as measured by Ki-67 expression, IncuCyte imaging, and 5-ethynyl-2-deoxyuridine assays. Transcriptomic analysis of LINC00958-depleted KCs revealed enrichment of proliferation- and cell cycle‒related genes among differentially expressed transcripts. Moreover, LINC00958 depletion led to decreased basal and IL-17A‒induced phosphorylation of p38. Furthermore, IL-17A‒induced KC proliferation was counteracted by the inhibition of LINC00958. In summary, our data support a role for the IL-17A‒induced long noncoding RNA, LINC00958, in the pathological circuits of psoriasis by reinforcing IL-17A‒induced epidermal hyperproliferation.
Collapse
Affiliation(s)
- Longlong Luo
- Dermatology and Venereology Division, Department of Medicine, Solna, Karolinska Institutet, Solna, Sweden; Center for Molecular Medicine, Karolinska University Hospital, Solna, Sweden; Dermatology and Venereology, Department of Medical Sciences, Uppsala University, Uppsala, Sweden
| | - Lorenzo Pasquali
- Dermatology and Venereology Division, Department of Medicine, Solna, Karolinska Institutet, Solna, Sweden; Center for Molecular Medicine, Karolinska University Hospital, Solna, Sweden
| | - Ankit Srivastava
- Dermatology and Venereology Division, Department of Medicine, Solna, Karolinska Institutet, Solna, Sweden; Center for Molecular Medicine, Karolinska University Hospital, Solna, Sweden; Science for Life Laboratory, Department of Microbiology, Tumor and Cell Biology, Karolinska Institute, Solna, Sweden; Program in Epithelial Biology, Stanford University School of Medicine, Stanford, California, USA
| | - Jan C Freisenhausen
- Dermatology and Venereology Division, Department of Medicine, Solna, Karolinska Institutet, Solna, Sweden; Center for Molecular Medicine, Karolinska University Hospital, Solna, Sweden; Dermatology and Venereology, Department of Medical Sciences, Uppsala University, Uppsala, Sweden
| | - Andor Pivarcsi
- Dermatology and Venereology Division, Department of Medicine, Solna, Karolinska Institutet, Solna, Sweden; Center for Molecular Medicine, Karolinska University Hospital, Solna, Sweden; Department of Medical Biochemistry and Microbiology, Uppsala University, Uppsala, Sweden
| | - Enikö Sonkoly
- Dermatology and Venereology Division, Department of Medicine, Solna, Karolinska Institutet, Solna, Sweden; Center for Molecular Medicine, Karolinska University Hospital, Solna, Sweden; Dermatology and Venereology, Department of Medical Sciences, Uppsala University, Uppsala, Sweden.
| |
Collapse
|
8
|
Kaufman T, Nitzan E, Firestein N, Ginzberg MB, Iyengar S, Patel N, Ben-Hamo R, Porat Z, Hunter J, Hilfinger A, Rotter V, Kafri R, Straussman R. Visual barcodes for clonal-multiplexing of live microscopy-based assays. Nat Commun 2022; 13:2725. [PMID: 35585055 PMCID: PMC9117331 DOI: 10.1038/s41467-022-30008-0] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2020] [Accepted: 04/06/2022] [Indexed: 12/12/2022] Open
Abstract
While multiplexing samples using DNA barcoding revolutionized the pace of biomedical discovery, multiplexing of live imaging-based applications has been limited by the number of fluorescent proteins that can be deconvoluted using common microscopy equipment. To address this limitation, we develop visual barcodes that discriminate the clonal identity of single cells by different fluorescent proteins that are targeted to specific subcellular locations. We demonstrate that deconvolution of these barcodes is highly accurate and robust to many cellular perturbations. We then use visual barcodes to generate ‘Signalome’ cell-lines by mixing 12 clones of different live reporters into a single population, allowing simultaneous monitoring of the activity in 12 branches of signaling, at clonal resolution, over time. Using the ‘Signalome’ we identify two distinct clusters of signaling pathways that balance growth and proliferation, emphasizing the importance of growth homeostasis as a central organizing principle in cancer signaling. The ability to multiplex samples in live imaging applications, both in vitro and in vivo may allow better high-content characterization of complex biological systems. Multiplex analyses of samples allow understanding complex processes in cancer initiation, progression and therapy response. Here, the authors present a fluorescence imaging-based visual barcode for livecell clonal-multiplexing which allows identifying signalling pathways clusters in response to different chemotherapy compounds.
Collapse
Affiliation(s)
- Tom Kaufman
- Department of Molecular Cell Biology, Weizmann Institute of Science, Rehovot, Israel
| | - Erez Nitzan
- Department of Molecular Cell Biology, Weizmann Institute of Science, Rehovot, Israel
| | - Nir Firestein
- Department of Molecular Cell Biology, Weizmann Institute of Science, Rehovot, Israel
| | | | - Seshu Iyengar
- Department of Chemical and Physical Sciences, University of Toronto, Toronto, ON, Canada
| | - Nish Patel
- Programme in Cell Biology, The Hospital for Sick Children, Toronto, ON, Canada
| | - Rotem Ben-Hamo
- Department of Molecular Cell Biology, Weizmann Institute of Science, Rehovot, Israel
| | - Ziv Porat
- Flow Cytometry Unit, Life Sciences Core Facilities, Weizmann Institute of Science, Rehovot, Israel
| | - Jaryd Hunter
- Programme in Cell Biology, The Hospital for Sick Children, Toronto, ON, Canada.,Department of Molecular Genetics, University of Toronto, Toronto, ON, Canada
| | - Andreas Hilfinger
- Department of Chemical and Physical Sciences, University of Toronto, Toronto, ON, Canada
| | - Varda Rotter
- Department of Molecular Cell Biology, Weizmann Institute of Science, Rehovot, Israel
| | - Ran Kafri
- Programme in Cell Biology, The Hospital for Sick Children, Toronto, ON, Canada. .,Department of Molecular Genetics, University of Toronto, Toronto, ON, Canada.
| | - Ravid Straussman
- Department of Molecular Cell Biology, Weizmann Institute of Science, Rehovot, Israel.
| |
Collapse
|
9
|
Smith YE, Wang G, Flynn CL, Madden SF, MacEneaney O, Cruz RGB, Richards CE, Jahns H, Brennan M, Cremona M, Hennessy BT, Sheehan K, Casucci A, Sani FA, Hudson L, Fay J, Vellanki SH, O’Flaherty S, Devocelle M, Hill ADK, Brennan K, Sukumar S, Hopkins AM. Functional Antagonism of Junctional Adhesion Molecule-A (JAM-A), Overexpressed in Breast Ductal Carcinoma In Situ (DCIS), Reduces HER2-Positive Tumor Progression. Cancers (Basel) 2022; 14:cancers14051303. [PMID: 35267611 PMCID: PMC8909510 DOI: 10.3390/cancers14051303] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2022] [Revised: 02/24/2022] [Accepted: 02/25/2022] [Indexed: 02/05/2023] Open
Abstract
Simple Summary Specific drug targets for breast ductal carcinoma in situ (DCIS) remain elusive, despite increasing disease prevalence and burden to healthcare services. Estrogen receptor (ER)-negative HER2-positive DCIS, associated with the poorest patient prognosis, is in particular need of novel therapeutic avenues. This report provides the first evidence that a cell surface protein called JAM-A is upregulated on human DCIS patient tissues and can be readily targeted by a novel JAM-A-binding peptide inhibitor in separate in vivo models of DCIS. The anti-tumor efficacy and lack of systemic toxicity of this lead inhibitor, coupled with early indications of potential signaling pathways implicated, support the value of future studies investigating JAM-A as a novel drug target in DCIS patients. Abstract Breast ductal carcinoma in situ (DCIS) is clinically challenging, featuring high diagnosis rates and few targeted therapies. Expression/signaling from junctional adhesion molecule-A (JAM-A) has been linked to poor prognosis in invasive breast cancers, but its role in DCIS is unknown. Since progression from DCIS to invasive cancer has been linked with overexpression of the human epidermal growth factor receptor-2 (HER2), and JAM-A regulates HER2 expression, we evaluated JAM-A as a therapeutic target in DCIS. JAM-A expression was immunohistochemically assessed in patient DCIS tissues. A novel JAM-A antagonist (JBS2) was designed and tested alone/in combination with the HER2 kinase inhibitor lapatinib, using SUM-225 cells in vitro and in vivo as validated DCIS models. Murine tumors were proteomically analyzed. JAM-A expression was moderate/high in 96% of DCIS patient tissues, versus 23% of normal adjacent tissues. JBS2 bound to recombinant JAM-A, inhibiting cell viability in SUM-225 cells and a primary DCIS culture in vitro and in a chick embryo xenograft model. JBS2 reduced tumor progression in in vivo models of SUM-225 cells engrafted into mammary fat pads or directly injected into the mammary ducts of NOD-SCID mice. Preliminary proteomic analysis revealed alterations in angiogenic and apoptotic pathways. High JAM-A expression in aggressive DCIS lesions and their sensitivity to treatment by a novel JAM-A antagonist support the viability of testing JAM-A as a novel therapeutic target in DCIS.
Collapse
Affiliation(s)
- Yvonne E. Smith
- Department of Surgery, RCSI University of Medicine and Health Sciences, Beaumont Hospital, Dublin 9, Ireland; (Y.E.S.); (C.L.F.); (R.G.B.C.); (C.E.R.); (L.H.); (S.H.V.); (A.D.K.H.); (K.B.)
| | - Guannan Wang
- Department of Oncology, Johns Hopkins University School of Medicine, Baltimore, MD 21231, USA; (G.W.); (S.S.)
| | - Ciara L. Flynn
- Department of Surgery, RCSI University of Medicine and Health Sciences, Beaumont Hospital, Dublin 9, Ireland; (Y.E.S.); (C.L.F.); (R.G.B.C.); (C.E.R.); (L.H.); (S.H.V.); (A.D.K.H.); (K.B.)
| | - Stephen F. Madden
- Data Science Centre, RCSI University of Medicine and Health Sciences, Dublin 2, Ireland;
| | - Owen MacEneaney
- Department of Pathology, RCSI University of Medicine and Health Sciences, Beaumont Hospital, Dublin 9, Ireland; (O.M.); (K.S.); (J.F.)
| | - Rodrigo G. B. Cruz
- Department of Surgery, RCSI University of Medicine and Health Sciences, Beaumont Hospital, Dublin 9, Ireland; (Y.E.S.); (C.L.F.); (R.G.B.C.); (C.E.R.); (L.H.); (S.H.V.); (A.D.K.H.); (K.B.)
| | - Cathy E. Richards
- Department of Surgery, RCSI University of Medicine and Health Sciences, Beaumont Hospital, Dublin 9, Ireland; (Y.E.S.); (C.L.F.); (R.G.B.C.); (C.E.R.); (L.H.); (S.H.V.); (A.D.K.H.); (K.B.)
| | - Hanne Jahns
- School of Veterinary Medicine, University College Dublin, Dublin 4, Ireland;
| | - Marian Brennan
- School of Pharmacy and Biomolecular Sciences, RCSI University of Medicine and Health Sciences, Dublin 2, Ireland;
| | - Mattia Cremona
- Department of Medical Oncology, RCSI University of Medicine and Health Sciences, Beaumont Hospital, Dublin 9, Ireland; (M.C.); (B.T.H.)
| | - Bryan T. Hennessy
- Department of Medical Oncology, RCSI University of Medicine and Health Sciences, Beaumont Hospital, Dublin 9, Ireland; (M.C.); (B.T.H.)
| | - Katherine Sheehan
- Department of Pathology, RCSI University of Medicine and Health Sciences, Beaumont Hospital, Dublin 9, Ireland; (O.M.); (K.S.); (J.F.)
| | - Alexander Casucci
- School of Medicine, RCSI University of Medicine and Health Sciences, Dublin 2, Ireland; (A.C.); (F.A.S.)
| | - Faizah A. Sani
- School of Medicine, RCSI University of Medicine and Health Sciences, Dublin 2, Ireland; (A.C.); (F.A.S.)
| | - Lance Hudson
- Department of Surgery, RCSI University of Medicine and Health Sciences, Beaumont Hospital, Dublin 9, Ireland; (Y.E.S.); (C.L.F.); (R.G.B.C.); (C.E.R.); (L.H.); (S.H.V.); (A.D.K.H.); (K.B.)
| | - Joanna Fay
- Department of Pathology, RCSI University of Medicine and Health Sciences, Beaumont Hospital, Dublin 9, Ireland; (O.M.); (K.S.); (J.F.)
| | - Sri H. Vellanki
- Department of Surgery, RCSI University of Medicine and Health Sciences, Beaumont Hospital, Dublin 9, Ireland; (Y.E.S.); (C.L.F.); (R.G.B.C.); (C.E.R.); (L.H.); (S.H.V.); (A.D.K.H.); (K.B.)
| | - Siobhan O’Flaherty
- Department of Chemistry, RCSI University of Medicine and Health Sciences, Dublin 2, Ireland; (S.O.); (M.D.)
| | - Marc Devocelle
- Department of Chemistry, RCSI University of Medicine and Health Sciences, Dublin 2, Ireland; (S.O.); (M.D.)
| | - Arnold D. K. Hill
- Department of Surgery, RCSI University of Medicine and Health Sciences, Beaumont Hospital, Dublin 9, Ireland; (Y.E.S.); (C.L.F.); (R.G.B.C.); (C.E.R.); (L.H.); (S.H.V.); (A.D.K.H.); (K.B.)
| | - Kieran Brennan
- Department of Surgery, RCSI University of Medicine and Health Sciences, Beaumont Hospital, Dublin 9, Ireland; (Y.E.S.); (C.L.F.); (R.G.B.C.); (C.E.R.); (L.H.); (S.H.V.); (A.D.K.H.); (K.B.)
| | - Saraswati Sukumar
- Department of Oncology, Johns Hopkins University School of Medicine, Baltimore, MD 21231, USA; (G.W.); (S.S.)
| | - Ann M. Hopkins
- Department of Surgery, RCSI University of Medicine and Health Sciences, Beaumont Hospital, Dublin 9, Ireland; (Y.E.S.); (C.L.F.); (R.G.B.C.); (C.E.R.); (L.H.); (S.H.V.); (A.D.K.H.); (K.B.)
- Correspondence: ; Tel.: +353-1-809-3858
| |
Collapse
|
10
|
Kumar AA, Buckley BJ, Ranson M. The Urokinase Plasminogen Activation System in Pancreatic Cancer: Prospective Diagnostic and Therapeutic Targets. Biomolecules 2022; 12:152. [PMID: 35204653 PMCID: PMC8961517 DOI: 10.3390/biom12020152] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2021] [Revised: 01/13/2022] [Accepted: 01/16/2022] [Indexed: 02/07/2023] Open
Abstract
Pancreatic cancer is a highly aggressive malignancy that features high recurrence rates and the poorest prognosis of all solid cancers. The urokinase plasminogen activation system (uPAS) is strongly implicated in the pathophysiology and clinical outcomes of patients with pancreatic ductal adenocarcinoma (PDAC), which accounts for more than 90% of all pancreatic cancers. Overexpression of the urokinase-type plasminogen activator (uPA) or its cell surface receptor uPAR is a key step in the acquisition of a metastatic phenotype via multiple mechanisms, including the increased activation of cell surface localised plasminogen which generates the serine protease plasmin. This triggers multiple downstream processes that promote tumour cell migration and invasion. Increasing clinical evidence shows that the overexpression of uPA, uPAR, or of both is strongly associated with worse clinicopathological features and poor prognosis in PDAC patients. This review provides an overview of the current understanding of the uPAS in the pathogenesis and progression of pancreatic cancer, with a focus on PDAC, and summarises the substantial body of evidence that supports the role of uPAS components, including plasminogen receptors, in this disease. The review further outlines the clinical utility of uPAS components as prospective diagnostic and prognostic biomarkers for PDAC, as well as a rationale for the development of novel uPAS-targeted therapeutics.
Collapse
Affiliation(s)
- Ashna A. Kumar
- Illawarra Health and Medical Research Institute, Wollongong, NSW 2522, Australia; (A.A.K.); (B.J.B.)
- School of Chemistry and Molecular Biosciences, Faculty of Science, Medicine and Health, University of Wollongong, Wollongong, NSW 2522, Australia
| | - Benjamin J. Buckley
- Illawarra Health and Medical Research Institute, Wollongong, NSW 2522, Australia; (A.A.K.); (B.J.B.)
- School of Chemistry and Molecular Biosciences, Faculty of Science, Medicine and Health, University of Wollongong, Wollongong, NSW 2522, Australia
| | - Marie Ranson
- Illawarra Health and Medical Research Institute, Wollongong, NSW 2522, Australia; (A.A.K.); (B.J.B.)
- School of Chemistry and Molecular Biosciences, Faculty of Science, Medicine and Health, University of Wollongong, Wollongong, NSW 2522, Australia
| |
Collapse
|
11
|
Galindo CM, Oliveira Ganzella FAD, Klassen G, Souza Ramos EAD, Acco A. Nuances of PFKFB3 signaling in breast cancer. Clin Breast Cancer 2022; 22:e604-e614. [DOI: 10.1016/j.clbc.2022.01.002] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2021] [Revised: 12/28/2021] [Accepted: 01/09/2022] [Indexed: 02/08/2023]
|
12
|
Wu L, Zhou F, Xin W, Li L, Liu L, Yin X, Xu X, Wang Y, Hua Z. MAGP2 induces tumor progression by enhancing uPAR-mediated cell proliferation. Cell Signal 2021; 91:110214. [PMID: 34915136 DOI: 10.1016/j.cellsig.2021.110214] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2021] [Revised: 12/03/2021] [Accepted: 12/08/2021] [Indexed: 12/14/2022]
Abstract
Microfibril-associated glycoprotein 2 (MAGP2) plays an important role in regulating cell signaling and acts as a biomarker to predict the prognostic effect of tumor therapy. However, research on MAGP2 mostly focuses on its extracellular signal transmission features, and its potential intracellular function is rarely reported. Here, we reported that intracellular MAGP2 increased the stability of urokinase-type plasminogen activator receptor (uPAR) in the cell by direct interaction which inhibits the lysosomal-mediated degradation of uPAR. Furthermore, with the detection of protein content changes and proteomics analysis, we found that highly expressed MAGP2 promoted the proliferation of tumor cells through uPAR-mediated p38-NF-ĸB signaling axis activation, enhancement of DNA damage repair and reduction of cell stagnation in the S phase of the cell cycle. In the nude mouse xenograft model of colorectal cancer, the upregulation of MAGP2 in tumor cells significantly promoted tumor progression, while the downregulation of uPAR significantly attenuated tumor progression. These studies elucidate the role of MAGP2 inside the cell and provide a new explanation for why patients with higher MAGP2 expression in tumors are associated with a worse prognosis. In addition, we also determined a mechanism for the stable existence of uPAR in the cell, providing information for the development of tumor drugs targeting uPAR.
Collapse
Affiliation(s)
- Leyang Wu
- The State Key Laboratory of Pharmaceutical Biotechnology, School of Life Sciences, Nanjing University, Nanjing, 210023, Jiangsu, China
| | - Feng Zhou
- The State Key Laboratory of Pharmaceutical Biotechnology, School of Life Sciences, Nanjing University, Nanjing, 210023, Jiangsu, China
| | - Wenjie Xin
- The State Key Laboratory of Pharmaceutical Biotechnology, School of Life Sciences, Nanjing University, Nanjing, 210023, Jiangsu, China
| | - Lin Li
- The State Key Laboratory of Pharmaceutical Biotechnology, School of Life Sciences, Nanjing University, Nanjing, 210023, Jiangsu, China
| | - Lina Liu
- The State Key Laboratory of Pharmaceutical Biotechnology, School of Life Sciences, Nanjing University, Nanjing, 210023, Jiangsu, China
| | - Xingpeng Yin
- The State Key Laboratory of Pharmaceutical Biotechnology, School of Life Sciences, Nanjing University, Nanjing, 210023, Jiangsu, China
| | - Xuebo Xu
- The State Key Laboratory of Pharmaceutical Biotechnology, School of Life Sciences, Nanjing University, Nanjing, 210023, Jiangsu, China
| | - Yao Wang
- Division of Critical Care and Surgery, St. George Hospital, University of New South Wales, Sydney, NSW 2217, Australia
| | - Zichun Hua
- The State Key Laboratory of Pharmaceutical Biotechnology, School of Life Sciences, Nanjing University, Nanjing, 210023, Jiangsu, China; Changzhou High-Tech Research Institute of Nanjing University and Jiangsu TargetPharma Laboratories Inc., Changzhou 213164, Jiangsu, China; School of Biopharmacy, China Pharmaceutical University, Nanjing 210023, Jiangsu, China.
| |
Collapse
|
13
|
Huang Y, Wang Y, Wang Y, Wang N, Duan Q, Wang S, Liu M, Bilal MA, Zheng Y. LPCAT1 Promotes Cutaneous Squamous Cell Carcinoma via EGFR-Mediated Protein Kinase B/p38MAPK Signaling Pathways. J Invest Dermatol 2021; 142:303-313.e9. [PMID: 34358528 DOI: 10.1016/j.jid.2021.07.163] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2021] [Revised: 07/10/2021] [Accepted: 07/12/2021] [Indexed: 02/07/2023]
Abstract
Cutaneous squamous cell carcinoma (cSCC) is the second most common form of skin cancer. LPCAT1, a lysophosphatidylcholine acyltransferase, takes a center stage in membrane lipid remodeling. LPCAT1 is elevated in several cancers and contributes to cancer development. However, its role and molecular mechanisms in cSCC remain to be elucidated. In this study, we found that LPCAT1 was upregulated in cSCC tissues and in cell lines. In vitro, loss-of-function and gain-of-function experiments demonstrated that LPCAT1 facilitated cSCC cell proliferation, protected cells against apoptosis, accelerated epithelial‒mesenchymal transition, and enhanced cell metastasis. Mechanistically, LPCAT1 regulated EGFR signaling. The oncogenic effect of LPCAT1 was mediated by EGFR/protein kinase B and EGFR/p38MAPK pathways in cSCC. Using the xenograft mouse model, we consolidated the results mentioned earlier. In conclusion, LPCAT1 contributed to cSCC progression through EGFR-mediated protein kinase B and p38MAPK signaling pathways. LPCAT1 may serve as a target for therapeutic intervention in cSCC.
Collapse
Affiliation(s)
- Yingjian Huang
- Department of Dermatology, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Yuqian Wang
- Department of Dermatology, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Yan Wang
- Center for Mitochondrial Biology and Medicine, The Key Laboratory of Biomedical Information Engineering of Ministry of Education, School of Life Science and Technology, Xi'an Jiaotong University, Xi'an, China
| | - Ning Wang
- Department of Dermatology, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Qiqi Duan
- Department of Dermatology, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Shengbang Wang
- Department of Dermatology, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Meng Liu
- Department of Dermatology, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Muhammad Ahsan Bilal
- Department of Dermatology, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Yan Zheng
- Department of Dermatology, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China.
| |
Collapse
|
14
|
Synthesis and Biological Evaluation of 2,3,4-Triaryl-1,2,4-oxadiazol-5-ones as p38 MAPK Inhibitors. Molecules 2021; 26:molecules26061745. [PMID: 33804659 PMCID: PMC8003627 DOI: 10.3390/molecules26061745] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2021] [Revised: 03/12/2021] [Accepted: 03/18/2021] [Indexed: 12/13/2022] Open
Abstract
A series of azastilbene derivatives, characterized by the presence of the 1,2,4-oxadiazole-5-one system as a linker of the two aromatic rings of stilbenes, have been prepared as novel potential inhibitors of p38 MAPK. Biological assays indicated that some of the synthesized compounds are endowed with good inhibitory activity towards the kinase. Molecular modeling data support the biological results showing that the designed compounds possess a reasonable binding mode in the ATP binding pocket of p38α kinase with a good binding affinity.
Collapse
|
15
|
Synthesis and evaluation of the epithelial-to- mesenchymal inhibitory activity of indazole-derived imidazoles as dual ALK5/p38α MAP inhibitors. Eur J Med Chem 2021; 216:113311. [PMID: 33677350 DOI: 10.1016/j.ejmech.2021.113311] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2020] [Revised: 02/15/2021] [Accepted: 02/15/2021] [Indexed: 01/02/2023]
Abstract
Drugs of targeting both activin receptor-like kinase 5 (ALK5) and p38α have therapeutic advantages, making them attractive treatment options for tumors. Two series of 4-(1H-indazol-5-yl)-5-(6-methylpyridin-2-yl)-1H-imidazoles 13a-g and 4-(1-methyl-1H-indazol-5-yl)-5-(6-methylpyridin-2-yl)-1H-imidazoles 20a-g were synthesized and evaluated for ALK5 and p38α mitogen-activated protein kinase inhibitory activity. The most potent compound, 13c (J-1090), inhibited ALK5- and p38α-mediated phosphorylation with half-maximal inhibitor concentrations of 0.004 μM and 0.004 μM, respectively, in the enzymatic assay. In this study, the effectiveness of 13c in transforming growth factor (TGF-β)-exposed U87MG cells was investigated using western blotting, immunofluorescence assays, cell migration assay, invasion assay, and RT-PCR analysis. 13c inhibited the protein expression of Slug and the protein and RNA expression of the mesenchymal-related proteins N-cadherin and vimentin. Furthermore, 13c markedly suppressed TGF-β-induced epithelial-to-mesenchymal transition (EMT), migration, and invasion in U87MG cells. These results suggest that 13c is a novel inhibitor of ALK5 with potential utility in the treatment of human glioma.
Collapse
|
16
|
Wang X, Veeraraghavan J, Liu CC, Cao X, Qin L, Kim JA, Tan Y, Loo SK, Hu Y, Lin L, Lee S, Shea MJ, Mitchell T, Li S, Ellis MJ, Hilsenbeck SG, Schiff R, Wang XS. Therapeutic Targeting of Nemo-like Kinase in Primary and Acquired Endocrine-resistant Breast Cancer. Clin Cancer Res 2021; 27:2648-2662. [PMID: 33542078 DOI: 10.1158/1078-0432.ccr-20-2961] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2020] [Revised: 11/29/2020] [Accepted: 02/01/2021] [Indexed: 11/16/2022]
Abstract
PURPOSE Endocrine resistance remains a major clinical challenge in estrogen receptor (ER)-positive breast cancer. Despite the encouraging results from clinical trials for the drugs targeting known survival signaling, relapse is still inevitable. There is an unmet need to discover new drug targets in the unknown escape pathways. Here, we report Nemo-like kinase (NLK) as a new actionable kinase target that endows previously uncharacterized survival signaling in endocrine-resistant breast cancer. EXPERIMENTAL DESIGN The effects of NLK inhibition on the viability of endocrine-resistant breast cancer cell lines were examined by MTS assay. The effect of VX-702 on NLK activity was verified by kinase assay. The modulation of ER and its coactivator, SRC-3, by NLK was examined by immunoprecipitation, kinase assay, luciferase assay, and RNA sequencing. The therapeutic effects of VX-702 and everolimus were tested on cell line- and patient-derived xenograft (PDX) tumor models. RESULTS NLK overexpression endows reduced endocrine responsiveness and is associated with worse outcome of patients treated with tamoxifen. Mechanistically, NLK may function, at least in part, via enhancing the phosphorylation of ERα and its key coactivator, SRC-3, to modulate ERα transcriptional activity. Through interrogation of a kinase profiling database, we uncovered and verified a highly selective dual p38/NLK inhibitor, VX-702. Coadministration of VX-702 with the mTOR inhibitor, everolimus, demonstrated a significant therapeutic effect in cell line-derived xenograft and PDX tumor models of acquired or de novo endocrine resistance. CONCLUSIONS Together, this study reveals the potential of therapeutic modulation of NLK for the management of the endocrine-resistant breast cancers with active NLK signaling.
Collapse
Affiliation(s)
- Xian Wang
- UPMC Hillman Cancer Center, University of Pittsburgh, Pittsburgh, Pennsylvania.,Department of Pathology, University of Pittsburgh, Pittsburgh, Pennsylvania.,Women's Cancer Research Center, Magee-Womens Research Institute, Pittsburgh, Pennsylvania.,Lester & Sue Smith Breast Center, Baylor College of Medicine, Houston, Texas.,Dan L. Duncan Comprehensive Cancer Center, Baylor College of Medicine, Houston, Texas.,Department of Medicine, Baylor College of Medicine, Houston, Texas.,Department of Molecular & Cellular Biology, Baylor College of Medicine, Houston, Texas
| | - Jamunarani Veeraraghavan
- Lester & Sue Smith Breast Center, Baylor College of Medicine, Houston, Texas.,Dan L. Duncan Comprehensive Cancer Center, Baylor College of Medicine, Houston, Texas.,Department of Medicine, Baylor College of Medicine, Houston, Texas.,Department of Molecular & Cellular Biology, Baylor College of Medicine, Houston, Texas
| | - Chia-Chia Liu
- UPMC Hillman Cancer Center, University of Pittsburgh, Pittsburgh, Pennsylvania.,Department of Pathology, University of Pittsburgh, Pittsburgh, Pennsylvania.,Women's Cancer Research Center, Magee-Womens Research Institute, Pittsburgh, Pennsylvania
| | - Xixi Cao
- Lester & Sue Smith Breast Center, Baylor College of Medicine, Houston, Texas.,Dan L. Duncan Comprehensive Cancer Center, Baylor College of Medicine, Houston, Texas.,Department of Medicine, Baylor College of Medicine, Houston, Texas.,Department of Molecular & Cellular Biology, Baylor College of Medicine, Houston, Texas
| | - Lanfang Qin
- Lester & Sue Smith Breast Center, Baylor College of Medicine, Houston, Texas.,Dan L. Duncan Comprehensive Cancer Center, Baylor College of Medicine, Houston, Texas.,Department of Medicine, Baylor College of Medicine, Houston, Texas.,Department of Molecular & Cellular Biology, Baylor College of Medicine, Houston, Texas
| | - Jin-Ah Kim
- Lester & Sue Smith Breast Center, Baylor College of Medicine, Houston, Texas.,Dan L. Duncan Comprehensive Cancer Center, Baylor College of Medicine, Houston, Texas.,Department of Medicine, Baylor College of Medicine, Houston, Texas.,Department of Molecular & Cellular Biology, Baylor College of Medicine, Houston, Texas
| | - Ying Tan
- Lester & Sue Smith Breast Center, Baylor College of Medicine, Houston, Texas.,Dan L. Duncan Comprehensive Cancer Center, Baylor College of Medicine, Houston, Texas.,Department of Medicine, Baylor College of Medicine, Houston, Texas.,Department of Molecular & Cellular Biology, Baylor College of Medicine, Houston, Texas
| | - Suet Kee Loo
- UPMC Hillman Cancer Center, University of Pittsburgh, Pittsburgh, Pennsylvania.,Department of Pathology, University of Pittsburgh, Pittsburgh, Pennsylvania.,Women's Cancer Research Center, Magee-Womens Research Institute, Pittsburgh, Pennsylvania
| | - Yiheng Hu
- UPMC Hillman Cancer Center, University of Pittsburgh, Pittsburgh, Pennsylvania.,Department of Pathology, University of Pittsburgh, Pittsburgh, Pennsylvania.,Women's Cancer Research Center, Magee-Womens Research Institute, Pittsburgh, Pennsylvania.,Lester & Sue Smith Breast Center, Baylor College of Medicine, Houston, Texas.,Dan L. Duncan Comprehensive Cancer Center, Baylor College of Medicine, Houston, Texas.,Department of Medicine, Baylor College of Medicine, Houston, Texas.,Department of Molecular & Cellular Biology, Baylor College of Medicine, Houston, Texas
| | - Ling Lin
- UPMC Hillman Cancer Center, University of Pittsburgh, Pittsburgh, Pennsylvania.,Department of Pathology, University of Pittsburgh, Pittsburgh, Pennsylvania.,Women's Cancer Research Center, Magee-Womens Research Institute, Pittsburgh, Pennsylvania
| | - Sanghoon Lee
- UPMC Hillman Cancer Center, University of Pittsburgh, Pittsburgh, Pennsylvania.,Department of Biomedical Informatics, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Martin J Shea
- Lester & Sue Smith Breast Center, Baylor College of Medicine, Houston, Texas.,Dan L. Duncan Comprehensive Cancer Center, Baylor College of Medicine, Houston, Texas.,Department of Medicine, Baylor College of Medicine, Houston, Texas.,Department of Molecular & Cellular Biology, Baylor College of Medicine, Houston, Texas
| | - Tamika Mitchell
- Lester & Sue Smith Breast Center, Baylor College of Medicine, Houston, Texas.,Dan L. Duncan Comprehensive Cancer Center, Baylor College of Medicine, Houston, Texas.,Department of Medicine, Baylor College of Medicine, Houston, Texas.,Department of Molecular & Cellular Biology, Baylor College of Medicine, Houston, Texas
| | - Shunqiang Li
- Department of Medicine, Washington University School of Medicine at St Louis, St. Louis, Missouri
| | - Matthew J Ellis
- Lester & Sue Smith Breast Center, Baylor College of Medicine, Houston, Texas.,Dan L. Duncan Comprehensive Cancer Center, Baylor College of Medicine, Houston, Texas.,Department of Medicine, Baylor College of Medicine, Houston, Texas.,Department of Molecular & Cellular Biology, Baylor College of Medicine, Houston, Texas
| | - Susan G Hilsenbeck
- Lester & Sue Smith Breast Center, Baylor College of Medicine, Houston, Texas.,Dan L. Duncan Comprehensive Cancer Center, Baylor College of Medicine, Houston, Texas.,Department of Medicine, Baylor College of Medicine, Houston, Texas
| | - Rachel Schiff
- Lester & Sue Smith Breast Center, Baylor College of Medicine, Houston, Texas.,Dan L. Duncan Comprehensive Cancer Center, Baylor College of Medicine, Houston, Texas.,Department of Medicine, Baylor College of Medicine, Houston, Texas.,Department of Molecular & Cellular Biology, Baylor College of Medicine, Houston, Texas
| | - Xiao-Song Wang
- UPMC Hillman Cancer Center, University of Pittsburgh, Pittsburgh, Pennsylvania. .,Department of Pathology, University of Pittsburgh, Pittsburgh, Pennsylvania.,Women's Cancer Research Center, Magee-Womens Research Institute, Pittsburgh, Pennsylvania.,Lester & Sue Smith Breast Center, Baylor College of Medicine, Houston, Texas.,Dan L. Duncan Comprehensive Cancer Center, Baylor College of Medicine, Houston, Texas.,Department of Medicine, Baylor College of Medicine, Houston, Texas.,Department of Molecular & Cellular Biology, Baylor College of Medicine, Houston, Texas
| |
Collapse
|
17
|
Jimenez T, Friedman T, Vadgama J, Singh V, Tucker A, Collazo J, Sinha S, Hikim AS, Singh R, Pervin S. Nicotine Synergizes with High-Fat Diet to Induce an Anti-Inflammatory Microenvironment to Promote Breast Tumor Growth. Mediators Inflamm 2020; 2020:5239419. [PMID: 33414685 PMCID: PMC7752272 DOI: 10.1155/2020/5239419] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2020] [Revised: 10/26/2020] [Accepted: 11/25/2020] [Indexed: 01/03/2023] Open
Abstract
Breast cancer results from a complex interplay of genetics and environment that alters immune and inflammatory systems to promote tumorigenesis. Obesity and cigarette smoking are well-known risk factors associated breast cancer development. Nicotine known to decrease inflammatory signals also modulates immune responses that favor breast cancer development. However, the mechanisms by which nicotine and obesity contribute to breast cancer remain poorly understood. In this study, we examined potential mechanisms by which nicotine (NIC) and high-fat diet (HFD) promote growth of HCC70 and HCC1806 xenografts from African American (AA) triple negative (TN) breast cancer cells. Immunodeficient mice fed on HFD and treated with NIC generated larger HCC70 and HCC1806 tumors when compared to NIC or HFD alone. Increased xenograft growth in the presence of NIC and HFD was accompanied by higher levels of tissue-resident macrophage markers and anti-inflammatory cytokines including IL4, IL13, and IL10. We further validated the involvement of these players by in vitro and ex vivo experiments. We found a proinflammatory milieu with increased expression of IL6 and IL12 in xenografts with HFD. In addition, nicotine or nicotine plus HFD increased a subset of mammary cancer stem cells (MCSCs) and key adipose browning markers CD137 and TMEM26. Interestingly, there was upregulation of stress-induced pp38 MAPK and pERK1/2 in xenografts exposed to HFD alone or nicotine plus HFD. Scratch-wound assay showed marked reduction in proliferation/migration of nicotine and palmitate-treated breast cancer cells with mecamylamine (MEC), a nicotine acetylcholine receptor (nAchR) antagonist. Furthermore, xenograft development in immune-deficient mice, fed HFD plus nicotine, was reduced upon cotreatment with MEC and SB 203580, a pp38MAPK inhibitor. Our study demonstrates the presence of nicotine and HFD in facilitating an anti-inflammatory tumor microenvironment that influences breast tumor growth. This study also shows potential efficacy of combination therapy in obese breast cancer patients who smoke.
Collapse
Affiliation(s)
- Thalia Jimenez
- Division of Endocrinology and Metabolism, Department of Medicine, Charles R. Drew University of Medicine and Science, Los Angeles, CA 90059, USA
| | - Theodore Friedman
- Division of Endocrinology and Metabolism, Department of Medicine, Charles R. Drew University of Medicine and Science, Los Angeles, CA 90059, USA
- Department of Medicine, David Geffen School of Medicine at UCLA, Los Angeles, CA 90095, USA
| | - Jaydutt Vadgama
- Department of Medicine, David Geffen School of Medicine at UCLA, Los Angeles, CA 90095, USA
- Jonsson Comprehensive Cancer Center, David Geffen School of Medicine at UCLA, Los Angeles, CA 90095, USA
- Division of Cancer Research and Training, Department of Medicine, Charles R. Drew University of Medicine and Science, Los Angeles, CA 90059, USA
| | - Vineeta Singh
- Division of Endocrinology and Metabolism, Department of Medicine, Charles R. Drew University of Medicine and Science, Los Angeles, CA 90059, USA
| | - Alexandria Tucker
- Division of Endocrinology and Metabolism, Department of Medicine, Charles R. Drew University of Medicine and Science, Los Angeles, CA 90059, USA
| | - Javier Collazo
- Division of Endocrinology and Metabolism, Department of Medicine, Charles R. Drew University of Medicine and Science, Los Angeles, CA 90059, USA
| | - Satyesh Sinha
- Division of Endocrinology and Metabolism, Department of Medicine, Charles R. Drew University of Medicine and Science, Los Angeles, CA 90059, USA
- Department of Medicine, David Geffen School of Medicine at UCLA, Los Angeles, CA 90095, USA
| | - Amiya Sinha Hikim
- Division of Endocrinology and Metabolism, Department of Medicine, Charles R. Drew University of Medicine and Science, Los Angeles, CA 90059, USA
- Department of Medicine, David Geffen School of Medicine at UCLA, Los Angeles, CA 90095, USA
| | - Rajan Singh
- Division of Endocrinology and Metabolism, Department of Medicine, Charles R. Drew University of Medicine and Science, Los Angeles, CA 90059, USA
- Jonsson Comprehensive Cancer Center, David Geffen School of Medicine at UCLA, Los Angeles, CA 90095, USA
- Department of Obstetrics and Gynecology, David Geffen School of Medicine at UCLA, Los Angeles, CA 90095, USA
| | - Shehla Pervin
- Division of Endocrinology and Metabolism, Department of Medicine, Charles R. Drew University of Medicine and Science, Los Angeles, CA 90059, USA
- Jonsson Comprehensive Cancer Center, David Geffen School of Medicine at UCLA, Los Angeles, CA 90095, USA
- Department of Obstetrics and Gynecology, David Geffen School of Medicine at UCLA, Los Angeles, CA 90095, USA
| |
Collapse
|
18
|
Bhargavi M, Vhora N, Lanka G, Somadi G, Kanth SS, Jain A, Potlapally SR. Homology modelling and virtual screening to explore potent inhibitors for MAP2K3 protein. Struct Chem 2020. [DOI: 10.1007/s11224-020-01667-w] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022]
|
19
|
Yang J, Li Y, Wang L, Zhang Z, Li Z, Jia Q. LncRNA H19 aggravates TNF-α-induced inflammatory injury via TAK1 pathway in MH7A cells. Biofactors 2020; 46:813-820. [PMID: 32525617 DOI: 10.1002/biof.1659] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/13/2019] [Revised: 01/19/2020] [Accepted: 01/21/2020] [Indexed: 02/06/2023]
Abstract
Rheumatoid arthritis (RA) is a common chronic autoimmune disease in women. This research aims to disclose the probable function of lncRNA H19 in MH7A cells. The influences of tumor necrosis factor-α (TNF-α) on cell viability, apoptosis, and inflammatory factor expression were, respectively, detected through cell counting kit-8 (CCK-8), flow cytometry, quantitative reverse transcription polymerase chain reaction (qRT-PCR), enzyme-linked immunosorbent assay (ELISA) assay and Western Blot. The levels of H19 and TAK1 were, respectively, tested through qRT-PCR and Western blot. The expression of NF-κB and JNK/p38MAPK pathway-associated proteins was tested through Western blot. We found that TNF-α reduced MH7A cell viability in a concentration-dependent manner and facilitated apoptosis and IL-8, IL-1β, and IL-6 production. Besides, TNF-α treatment raised the level of H19 in MH7A cells. Moreover, H19 silence reduced the levels of inflammatory cytokines, while overexpression of H19 reversed this effect. TNF-α treatment elevated the expression of inflammatory cytokines by up-regulating H19. Furthermore, overexpression of H19 promoted TAK1 phosphorylation. Following studies revealed that H19 activated NF-κB and JNK/p38 MAPK pathways by promoting TAK1 phosphorylation.
Collapse
Affiliation(s)
- Jialiang Yang
- Department of Rheumatology and Immunology, Linyi People's Hospital, Linyi, Shandong, China
| | - Yixuan Li
- Department of Critical Care Medicine, Linyi People's Hospital, Linyi, Shandong, China
| | - Lili Wang
- Department of Rheumatology and Immunology, Linyi People's Hospital, Linyi, Shandong, China
| | - Zhenchun Zhang
- Department of Rheumatology and Immunology, Linyi People's Hospital, Linyi, Shandong, China
| | - Zunzhong Li
- Department of Rheumatology and Immunology, Linyi People's Hospital, Linyi, Shandong, China
| | - Qian Jia
- Department of Rheumatology and Immunology, Linyi People's Hospital, Linyi, Shandong, China
| |
Collapse
|
20
|
Yang J, Wang T, Zhao L, Rajasekhar VK, Joshi S, Andreou C, Pal S, Hsu HT, Zhang H, Cohen IJ, Huang R, Hendrickson RC, Miele MM, Pei W, Brendel MB, Healey JH, Chiosis G, Kircher MF. Gold/alpha-lactalbumin nanoprobes for the imaging and treatment of breast cancer. Nat Biomed Eng 2020; 4:686-703. [PMID: 32661307 PMCID: PMC8255032 DOI: 10.1038/s41551-020-0584-z] [Citation(s) in RCA: 48] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2018] [Accepted: 06/11/2020] [Indexed: 02/03/2023]
Abstract
Theranostic agents should ideally be renally cleared and biodegradable. Here, we report the synthesis, characterization and theranostic applications of fluorescent ultrasmall gold quantum clusters that are stabilized by the milk metalloprotein alpha-lactalbumin. We synthesized three types of these nanoprobes that together display fluorescence across the visible and near-infrared spectra when excited at a single wavelength through optical colour coding. In live tumour-bearing mice, the near-infrared nanoprobe generates contrast for fluorescence, X-ray computed tomography and magnetic resonance imaging, and exhibits long circulation times, low accumulation in the reticuloendothelial system, sustained tumour retention, insignificant toxicity and renal clearance. An intravenously administrated near-infrared nanoprobe with a large Stokes shift facilitated the detection and image-guided resection of breast tumours in vivo using a smartphone with modified optics. Moreover, the partially unfolded structure of alpha-lactalbumin in the nanoprobe helps with the formation of an anti-cancer lipoprotein complex with oleic acid that triggers the inhibition of the MAPK and PI3K-AKT pathways, immunogenic cell death and the recruitment of infiltrating macrophages. The biodegradability and safety profile of the nanoprobes make them suitable for the systemic detection and localized treatment of cancer.
Collapse
Affiliation(s)
- Jiang Yang
- Center for Molecular Imaging and Nanotechnology (CMINT), Memorial Sloan Kettering Cancer Center, New York, NY, USA
- Department of Radiology, Memorial Sloan Kettering Cancer Center, New York, NY, USA
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou, China
| | - Tai Wang
- Chemical Biology Program, Sloan Kettering Institute, New York, NY, USA
| | - Lina Zhao
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, Institute of High Energy Physics, Chinese Academy of Sciences, Beijing, China
| | | | - Suhasini Joshi
- Chemical Biology Program, Sloan Kettering Institute, New York, NY, USA
| | - Chrysafis Andreou
- Center for Molecular Imaging and Nanotechnology (CMINT), Memorial Sloan Kettering Cancer Center, New York, NY, USA
- Department of Radiology, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Suchetan Pal
- Center for Molecular Imaging and Nanotechnology (CMINT), Memorial Sloan Kettering Cancer Center, New York, NY, USA
- Department of Radiology, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Hsiao-Ting Hsu
- Center for Molecular Imaging and Nanotechnology (CMINT), Memorial Sloan Kettering Cancer Center, New York, NY, USA
- Department of Radiology, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Hanwen Zhang
- Center for Molecular Imaging and Nanotechnology (CMINT), Memorial Sloan Kettering Cancer Center, New York, NY, USA
- Department of Radiology, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Ivan J Cohen
- Center for Molecular Imaging and Nanotechnology (CMINT), Memorial Sloan Kettering Cancer Center, New York, NY, USA
- Louis V. Gerstner Jr Graduate School of Biomedical Sciences, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Ruimin Huang
- Center for Molecular Imaging and Nanotechnology (CMINT), Memorial Sloan Kettering Cancer Center, New York, NY, USA
- Department of Radiology, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Ronald C Hendrickson
- Proteomics and Microchemistry Core Laboratory, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Matthew M Miele
- Proteomics and Microchemistry Core Laboratory, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Wenbo Pei
- Chemical Biology Program, Sloan Kettering Institute, New York, NY, USA
| | - Matthew B Brendel
- Molecular Cytology Core Laboratory, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - John H Healey
- Department of Surgery, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Gabriela Chiosis
- Chemical Biology Program, Sloan Kettering Institute, New York, NY, USA
- Breast Cancer Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Moritz F Kircher
- Center for Molecular Imaging and Nanotechnology (CMINT), Memorial Sloan Kettering Cancer Center, New York, NY, USA.
- Department of Radiology, Memorial Sloan Kettering Cancer Center, New York, NY, USA.
- Molecular Pharmacology Program, Sloan Kettering Institute, New York, NY, USA.
- Department of Radiology, Weill Cornell Medical College, New York, NY, USA.
- Department of Imaging, Dana-Farber Cancer Institute and Harvard Medical School, Boston, MA, USA.
- Department of Radiology, Brigham & Women's Hospital and Harvard Medical School, Boston, MA, USA.
| |
Collapse
|
21
|
Höving AL, Schmidt KE, Merten M, Hamidi J, Rott AK, Faust I, Greiner JFW, Gummert J, Kaltschmidt B, Kaltschmidt C, Knabbe C. Blood Serum Stimulates p38-Mediated Proliferation and Changes in Global Gene Expression of Adult Human Cardiac Stem Cells. Cells 2020; 9:cells9061472. [PMID: 32560212 PMCID: PMC7349155 DOI: 10.3390/cells9061472] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2020] [Revised: 06/05/2020] [Accepted: 06/13/2020] [Indexed: 12/18/2022] Open
Abstract
During aging, senescent cells accumulate in various tissues accompanied by decreased regenerative capacities of quiescent stem cells, resulting in deteriorated organ function and overall degeneration. In this regard, the adult human heart with a generally low regenerative potential is of extreme interest as a target for rejuvenating strategies with blood borne factors that might be able to activate endogenous stem cell populations. Here, we investigated for the first time the effects of human blood plasma and serum on adult human cardiac stem cells (hCSCs) and showed significantly increased proliferation capacities and metabolism accompanied by a significant decrease of senescent cells, demonstrating a beneficial serum-mediated effect that seemed to be independent of age and sex. However, RNA-seq analysis of serum-treated hCSCs revealed profound effects on gene expression depending on the age and sex of the plasma donor. We further successfully identified key pathways that are affected by serum treatment with p38-MAPK playing a regulatory role in protection from senescence and in the promotion of proliferation in a serum-dependent manner. Inhibition of p38-MAPK resulted in a decline of these serum-mediated beneficial effects on hCSCs in terms of decreased proliferation and accelerated senescence. In summary, we provide new insights in the regulatory networks behind serum-mediated protective effects on adult human cardiac stem cells.
Collapse
Affiliation(s)
- Anna L. Höving
- Department of Cell Biology, University of Bielefeld, 33615 Bielefeld, Germany; (K.E.S.); (J.H.); (A.-K.R.); (J.F.W.G.)
- Institute for Laboratory- and Transfusion Medicine, Heart and Diabetes Centre NRW, Ruhr University Bochum, 32545 Bad Oeynhausen, Germany; (I.F.); (C.K.)
- Correspondence: (A.L.H.); (C.K.)
| | - Kazuko E. Schmidt
- Department of Cell Biology, University of Bielefeld, 33615 Bielefeld, Germany; (K.E.S.); (J.H.); (A.-K.R.); (J.F.W.G.)
- Institute for Laboratory- and Transfusion Medicine, Heart and Diabetes Centre NRW, Ruhr University Bochum, 32545 Bad Oeynhausen, Germany; (I.F.); (C.K.)
| | - Madlen Merten
- AG Molecular Neurobiology, University of Bielefeld, 33615 Bielefeld, Germany; (M.M.); (B.K.)
| | - Jassin Hamidi
- Department of Cell Biology, University of Bielefeld, 33615 Bielefeld, Germany; (K.E.S.); (J.H.); (A.-K.R.); (J.F.W.G.)
| | - Ann-Katrin Rott
- Department of Cell Biology, University of Bielefeld, 33615 Bielefeld, Germany; (K.E.S.); (J.H.); (A.-K.R.); (J.F.W.G.)
| | - Isabel Faust
- Institute for Laboratory- and Transfusion Medicine, Heart and Diabetes Centre NRW, Ruhr University Bochum, 32545 Bad Oeynhausen, Germany; (I.F.); (C.K.)
| | - Johannes F. W. Greiner
- Department of Cell Biology, University of Bielefeld, 33615 Bielefeld, Germany; (K.E.S.); (J.H.); (A.-K.R.); (J.F.W.G.)
| | - Jan Gummert
- Department of Thoracic and Cardiovascular surgery, Heart and Diabetes Centre NRW, Ruhr-University Bochum, 32545 Bad Oeynhausen, Germany;
| | - Barbara Kaltschmidt
- AG Molecular Neurobiology, University of Bielefeld, 33615 Bielefeld, Germany; (M.M.); (B.K.)
| | - Christian Kaltschmidt
- Department of Cell Biology, University of Bielefeld, 33615 Bielefeld, Germany; (K.E.S.); (J.H.); (A.-K.R.); (J.F.W.G.)
- Correspondence: (A.L.H.); (C.K.)
| | - Cornelius Knabbe
- Institute for Laboratory- and Transfusion Medicine, Heart and Diabetes Centre NRW, Ruhr University Bochum, 32545 Bad Oeynhausen, Germany; (I.F.); (C.K.)
| |
Collapse
|
22
|
Dasatinib prevents skeletal metastasis of osteotropic MDA-MB-231 cells in a xenograft mouse model. Arch Gynecol Obstet 2020; 301:1493-1502. [PMID: 32170411 DOI: 10.1007/s00404-020-05496-4] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2019] [Accepted: 03/05/2020] [Indexed: 10/25/2022]
Abstract
PURPOSE Bone metastasis in breast cancer has been linked to activity of c-Src kinase, one of the extensively explored tyrosine kinases in cell biology. The impact of TNF-related apoptosis inducing ligand (TRAIL) and TRAIL receptors has just recently been integrated into this conception. METHODS An osteotropic clone of MDA-MB-231 cells simulated a model for bone metastasis of triple-negative breast cancer (TNBC). The effects of Dasatinib, a clinically established inhibitor of Src kinases family and Abl were evaluated in vitro and in vivo. In vivo effects of Dasatinib treatment on the occurrence of skeletal metastases were tested in a xenograft mouse model after intra-cardiac injection of osteotropic MDA-MB-231-cells. Ex vivo analyses of the bone sections confirmed intraosseous growth of metastases and allowed determination of osteoclastic activity. RESULTS Treatment of osteotropic MDA-MB-231 cells with Dasatinib inhibited proliferation rates in vitro. A shift in TRAIL-receptor expression towards an induction of oncogenic TRAIL-R2 was observed. In vivo, 15 of 30 mice received an intra-peritoneal treatment with Dasatinib. These mice showed significantly less skeletal metastases in bioluminescence scans. Moreover, a pronounced increase in bone volume was observed in the treatment group, as detected by µ-Computed Tomography. Dasatinib treatment also led to a greater increase in bone density in tibiae without metastatic affection, which was accompanied by reduced recruitment of osteoclasts. CONCLUSION Our observations support the concept of utilizing Dasatinib in targeting early-stage bone metastatic TNBC and sustaining bone health.
Collapse
|
23
|
Liu J, Yu X, Yu H, Liu B, Zhang Z, Kong C, Li Z. Knockdown of MAPK14 inhibits the proliferation and migration of clear cell renal cell carcinoma by downregulating the expression of CDC25B. Cancer Med 2019; 9:1183-1195. [PMID: 31856414 PMCID: PMC6997073 DOI: 10.1002/cam4.2795] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2019] [Revised: 11/22/2019] [Accepted: 12/06/2019] [Indexed: 12/19/2022] Open
Abstract
Mitogen‐activated protein kinase 14 (MAPK14), which plays an important role in DNA damage and repair, is activated by various environmental stress and proinflammatory cytokines. It is highly active in a variety of tumors, acting as a tumor promoter or suppressor, but its role in clear cell renal cell carcinoma (ccRCC) has not been elucidated. Cell division cycle 25B (CDC25B) is involved in cell cycle regulation and is highly expressed in many malignant tumors. The transcription levels of MAPK14 and CDC25B in 72 pairs of ccRCC and adjacent healthy tissues from the cancer genome atlas database and the protein expression levels in 66 pairs of clinical samples were analyzed in this study. After MAPK14 was knocked down by small interfering RNA (siRNA), P‐MAPK14 and CDC25B protein levels decreased. Subsequently, Western blot and co‐immunoprecipitation demonstrated that P‐MAPK14 could bind to CDC25B, potentially maintaining its stability. The proliferation and migration of ccRCC cell lines were suppressed by siRNA knockdown of MAPK14, however, that could be partially reversed by the overexpression of CDC25B. These results suggest that downregulation of MAPK14 and P‐MAPK14 could inhibit the proliferation and migration of ccRCC by downregulating CDC25B.
Collapse
Affiliation(s)
- Junlong Liu
- Department of Urology, The First Hospital of China Medical University, Shenyang, P. R. China
| | - Xiuyue Yu
- Department of Urology, The First Hospital of China Medical University, Shenyang, P. R. China
| | - Hongyuan Yu
- Department of Urology, The First Hospital of China Medical University, Shenyang, P. R. China
| | - Bitian Liu
- Department of Urology, Shengjing Hospital of China Medical University, Shenyang, P. R. China
| | - Zhe Zhang
- Department of Urology, The First Hospital of China Medical University, Shenyang, P. R. China
| | - Chuize Kong
- Department of Urology, The First Hospital of China Medical University, Shenyang, P. R. China
| | - Zhenhua Li
- Department of Urology, The First Hospital of China Medical University, Shenyang, P. R. China
| |
Collapse
|
24
|
Li YW, Li XY, Li S, Zhao LM, Ma J, Piao HR, Jiang Z, Jin CH, Jin X. Synthesis and evaluation of the HIF-1α inhibitory activity of 3(5)-substituted-4-(quinolin-4-yl)- and 4-(2-phenylpyridin-4-yl)pyrazoles as inhibitors of ALK5. Bioorg Med Chem Lett 2019; 30:126822. [PMID: 31810777 DOI: 10.1016/j.bmcl.2019.126822] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2019] [Revised: 10/03/2019] [Accepted: 11/10/2019] [Indexed: 12/27/2022]
Abstract
The transcription factor hypoxia-inducible factor-1α (HIF-1α) plays an important role in apoptosis, metastasis, and proliferation and is recognized as an important potential therapeutic target for cancer. Six series of 3(5)-(6-methylpyridin-2-yl)-4-(quinolin-4-yl)pyrazoles (11a-d, 12a-d, and 18a-d) and 3(5)-(6-methylpyridin-2-yl)-4-(2-phenyl-pyridin-4-yl)pyrazoles (19a-d, 20a-d, and 21a-d) were synthesized and evaluated for activin receptor-like kinase 5 (ALK5) and HIF-1α inhibitory activity at the enzyme and cell levels. The effect of the lead compound 20d (J-1012) on HIF-1α activation in HCT116 cells was investigated. J-1012 markedly decreased the hypoxia-induced or TNF-induced accumulation of HIF-1α protein dose-dependently. Analysis revealed that J-1012 inhibited HIF-1α protein synthesis, without affecting the degradation of HIF-1α protein. Furthermore, by inhibiting the activation of HIF-1α, J-1012 suppressed the metastasis and proliferation and promoted apoptosis of HCT116 cells. These results suggest that J-1012 may be a potential therapeutic agent against human colon cancer.
Collapse
Affiliation(s)
- Yan-Wei Li
- Key Laboratory of Natural Resources of Changbai Mountain and Functional Molecules, Ministry of Education, Molecular Medicine Research Center, College of Pharmacy, Yanbian University, Yanji 133002, PR China; Pharmacy Intravenous Admixture Services, Yanbian University Hospital, Yanji 133000, PR China
| | - Xiang-Yu Li
- Key Laboratory of Natural Resources of Changbai Mountain and Functional Molecules, Ministry of Education, Molecular Medicine Research Center, College of Pharmacy, Yanbian University, Yanji 133002, PR China
| | - Shanji Li
- The School of Public Health, Jilin Medical University, Jilin 132013, PR China
| | - Li-Min Zhao
- Key Laboratory of Natural Resources of Changbai Mountain and Functional Molecules, Ministry of Education, Molecular Medicine Research Center, College of Pharmacy, Yanbian University, Yanji 133002, PR China
| | - Juan Ma
- Key Laboratory of Natural Resources of Changbai Mountain and Functional Molecules, Ministry of Education, Molecular Medicine Research Center, College of Pharmacy, Yanbian University, Yanji 133002, PR China
| | - Hu-Ri Piao
- Key Laboratory of Natural Resources of Changbai Mountain and Functional Molecules, Ministry of Education, Molecular Medicine Research Center, College of Pharmacy, Yanbian University, Yanji 133002, PR China
| | - Zhe Jiang
- Pharmacy Intravenous Admixture Services, Yanbian University Hospital, Yanji 133000, PR China.
| | - Cheng Hua Jin
- Key Laboratory of Natural Resources of Changbai Mountain and Functional Molecules, Ministry of Education, Molecular Medicine Research Center, College of Pharmacy, Yanbian University, Yanji 133002, PR China.
| | - Xuejun Jin
- Key Laboratory of Natural Resources of Changbai Mountain and Functional Molecules, Ministry of Education, Molecular Medicine Research Center, College of Pharmacy, Yanbian University, Yanji 133002, PR China.
| |
Collapse
|
25
|
Lin A, Giuliano CJ, Palladino A, John KM, Abramowicz C, Yuan ML, Sausville EL, Lukow DA, Liu L, Chait AR, Galluzzo ZC, Tucker C, Sheltzer JM. Off-target toxicity is a common mechanism of action of cancer drugs undergoing clinical trials. Sci Transl Med 2019; 11:eaaw8412. [PMID: 31511426 PMCID: PMC7717492 DOI: 10.1126/scitranslmed.aaw8412] [Citation(s) in RCA: 389] [Impact Index Per Article: 77.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2019] [Revised: 06/19/2019] [Accepted: 08/01/2019] [Indexed: 12/14/2022]
Abstract
Ninety-seven percent of drug-indication pairs that are tested in clinical trials in oncology never advance to receive U.S. Food and Drug Administration approval. While lack of efficacy and dose-limiting toxicities are the most common causes of trial failure, the reason(s) why so many new drugs encounter these problems is not well understood. Using CRISPR-Cas9 mutagenesis, we investigated a set of cancer drugs and drug targets in various stages of clinical testing. We show that-contrary to previous reports obtained predominantly with RNA interference and small-molecule inhibitors-the proteins ostensibly targeted by these drugs are nonessential for cancer cell proliferation. Moreover, the efficacy of each drug that we tested was unaffected by the loss of its putative target, indicating that these compounds kill cells via off-target effects. By applying a genetic target-deconvolution strategy, we found that the mischaracterized anticancer agent OTS964 is actually a potent inhibitor of the cyclin-dependent kinase CDK11 and that multiple cancer types are addicted to CDK11 expression. We suggest that stringent genetic validation of the mechanism of action of cancer drugs in the preclinical setting may decrease the number of therapies tested in human patients that fail to provide any clinical benefit.
Collapse
Affiliation(s)
- Ann Lin
- Cold Spring Harbor Laboratory, Cold Spring Harbor, NY 11724, USA
- Stony Brook University, Stony Brook, NY 11794, USA
| | - Christopher J Giuliano
- Cold Spring Harbor Laboratory, Cold Spring Harbor, NY 11724, USA
- Stony Brook University, Stony Brook, NY 11794, USA
| | - Ann Palladino
- Cold Spring Harbor Laboratory, Cold Spring Harbor, NY 11724, USA
| | - Kristen M John
- Cold Spring Harbor Laboratory, Cold Spring Harbor, NY 11724, USA
- Hofstra University, Hempstead, NY 11549, USA
| | - Connor Abramowicz
- Cold Spring Harbor Laboratory, Cold Spring Harbor, NY 11724, USA
- New York Institute of Technology, Glen Head, NY 11545, USA
| | - Monet Lou Yuan
- Cold Spring Harbor Laboratory, Cold Spring Harbor, NY 11724, USA
- Syosset High School, Syosset, NY 11791, USA
| | - Erin L Sausville
- Cold Spring Harbor Laboratory, Cold Spring Harbor, NY 11724, USA
| | - Devon A Lukow
- Cold Spring Harbor Laboratory, Cold Spring Harbor, NY 11724, USA
- Stony Brook University, Stony Brook, NY 11794, USA
| | - Luwei Liu
- Cold Spring Harbor Laboratory, Cold Spring Harbor, NY 11724, USA
- Stony Brook University, Stony Brook, NY 11794, USA
| | | | | | - Clara Tucker
- Cold Spring Harbor Laboratory, Cold Spring Harbor, NY 11724, USA
- Stony Brook University, Stony Brook, NY 11794, USA
| | - Jason M Sheltzer
- Cold Spring Harbor Laboratory, Cold Spring Harbor, NY 11724, USA.
| |
Collapse
|
26
|
Brattås MK, Reikvam H, Tvedt THA, Bruserud Ø. Precision medicine for TP53-mutated acute myeloid leukemia. EXPERT REVIEW OF PRECISION MEDICINE AND DRUG DEVELOPMENT 2019. [DOI: 10.1080/23808993.2019.1644164] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Affiliation(s)
| | - Håkon Reikvam
- Department of Medicine, Haukeland University Hospital, Bergen, Norway
- Section for Hematology, Department of Clinical Science, University of Bergen, Bergen, Norway
| | | | - Øystein Bruserud
- Department of Medicine, Haukeland University Hospital, Bergen, Norway
- Section for Hematology, Department of Clinical Science, University of Bergen, Bergen, Norway
| |
Collapse
|
27
|
Mazumdar A, Tahaney WM, Reddy Bollu L, Poage G, Hill J, Zhang Y, Mills GB, Brown PH. The phosphatase PPM1A inhibits triple negative breast cancer growth by blocking cell cycle progression. NPJ Breast Cancer 2019; 5:22. [PMID: 31372497 PMCID: PMC6659706 DOI: 10.1038/s41523-019-0118-6] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2018] [Accepted: 06/26/2019] [Indexed: 12/31/2022] Open
Abstract
Estrogen receptor (ER)-negative, progesterone receptor (PR)-negative and HER2-negative, or "triple negative," breast cancer (TNBC) is a poor prognosis clinical subtype that occurs more frequently in younger women and is commonly treated with toxic chemotherapy. Effective targeted therapy for TNBC is urgently needed. Our previous studies have identified several kinases critical for TNBC growth. Since phosphatases regulate the function of kinase signaling pathways, we sought to identify critical growth-regulatory phosphatases that are expressed differentially in ER-negative, as compared to ER-positive, breast cancers. In this study, we examined the role of one of these differentially expressed phosphatases, the protein phosphatase Mg + 2/Mn + 2 dependent 1A (PPM1A) which is underexpressed in ER-negative breast cancer as compared to ER-positive breast cancers, in regulating TNBC growth. We found that PPM1A is deleted in ~40% of ER-negative breast cancers, and that induced expression of PPM1A suppresses in vitro and in vivo growth of TNBC cells. This study demonstrates that induction of PPM1A expression blocks the cell cycle and reduces CDK and Rb phosphorylation. These results suggest PPM1A is a crucial regulator of cell cycle progression in triple negative breast cancer. Our results also suggest that PPM1A loss should be explored as a predictive biomarker of CDK inhibitor sensitivity.
Collapse
Affiliation(s)
- Abhijit Mazumdar
- Department of Clinical Cancer Prevention, The University of Texas M.D. Anderson Cancer Center, Texas, USA
| | - William M. Tahaney
- Department of Clinical Cancer Prevention, The University of Texas M.D. Anderson Cancer Center, Texas, USA
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX 77030 USA
| | - Lakshmi Reddy Bollu
- Department of Clinical Cancer Prevention, The University of Texas M.D. Anderson Cancer Center, Texas, USA
| | | | - Jamal Hill
- Department of Clinical Cancer Prevention, The University of Texas M.D. Anderson Cancer Center, Texas, USA
| | - Yun Zhang
- Department of Clinical Cancer Prevention, The University of Texas M.D. Anderson Cancer Center, Texas, USA
| | - Gordon B. Mills
- Department of Cell, Developmental & Cancer Biology, Oregon Health & Science University, Oregon, USA
| | - Powel H. Brown
- Department of Clinical Cancer Prevention, The University of Texas M.D. Anderson Cancer Center, Texas, USA
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX 77030 USA
| |
Collapse
|
28
|
Zarredar H, Farajnia S, Ansarin K, Baradaran B, Aria M, Asadi M. Synergistic Effect of Novel EGFR Inhibitor AZD8931 and p38α siRNA in Lung Adenocarcinoma Cancer Cells. Anticancer Agents Med Chem 2019; 19:638-644. [DOI: 10.2174/1871520619666190301125203] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2018] [Revised: 04/26/2018] [Accepted: 02/13/2019] [Indexed: 12/19/2022]
Abstract
Background:Lung cancer is the leading cause of cancer-related death with less than 5-year survival rate for both men and women worldwide. EGFR and MAPK signaling pathways have a critical role in proliferation and progression of various cancers, including lung cancer. P38 map kinase plays different role in various tissue hence showing a tissue-dependent behavior. It acts as an oncogene in some tissues while plays as tumor suppressor in some other tissues. The aim of this study was to investigate the combined effect of P38 αspecific siRNA and EGFR inhibitor on apoptosis and proliferation of A549 lung cancer cell line.Objective:This article is dedicated to the synergistic effect of novel EGFR inhibitor AZD8931 and P38 α siRNA in lung adenocarcinoma cancer cells proliferation and apoptosis.Methods and Materials:The A549 lung cancer cells were treated with P38 α- siRNA and EGFR inhibitor alone or in combination. The cytotoxic effects of P38 α- siRNA and EGFR inhibitor were determined using MTT assay. Relative P38 α and EGFR mRNA levels were measured by QRT-PCR. Induction of apoptosis were measured by FACS analysis.Results:The expression of mRNA related to P38 α, EGFR, and Her2 genes was reduced to 23.4%, 52.4%, and 75, respectively, after treatment of their inhibitors. Also, MTT assay showed that the cell viability after treatment with p38 α SiRNA, EGFR inhibitor and their combination was reduced to 51.02%, 48.9%, and 25.11%, respectively. FACS results indicated that p38 α siRNA, EGFR inhibitor and their combination, reduced the population of live cells to 49.5%, 32.2% and 14.3% in comparison to the population of untreated control cells (99.5%).Conclusion:The results of this study indicated that p38 α and EGFR might play an important role in the development and growth of lung cancer and might be a potential therapeutic target for the treatment of lung cancer.
Collapse
Affiliation(s)
- Habib Zarredar
- Tuberculosis and Lung Disease Research Center, Tabriz University of Medical Science. Tabriz, Iran
| | - Safar Farajnia
- Tuberculosis and Lung Disease Research Center, Tabriz University of Medical Science. Tabriz, Iran
| | - Khalil Ansarin
- Tuberculosis and Lung Disease Research Center, Tabriz University of Medical Science. Tabriz, Iran
| | - Behzad Baradaran
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Maryam Aria
- Drug Applied Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Milad Asadi
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| |
Collapse
|
29
|
Ma Y, Lai W, Zhao M, Yue C, Shi F, Li R, Hu Z. Plastin 3 down-regulation augments the sensitivity of MDA-MB-231 cells to paclitaxel via the p38 MAPK signalling pathway. ARTIFICIAL CELLS NANOMEDICINE AND BIOTECHNOLOGY 2019; 47:685-695. [PMID: 30829071 DOI: 10.1080/21691401.2019.1576707] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/02/2023]
Abstract
Plastin 3 (PLS3) overexpression may serve as a marker for predicting chemotherapeutic outcomes in drug-resistant cancer cells, but the mechanism is unclear. Herein, we show that the down-regulation of PLS3 by PLS3 gene silencing augments the sensitivity of MDA-MB-231 triple-negative breast cancer cells to paclitaxel. Interestingly, a low concentration of paclitaxel was able to induce strong apoptosis in the PLS3-silenced cells. Further study revealed that p38 MAPK signalling was responsible for the increased sensitivity to paclitaxel in these cells, as the p38 MAPK inhibitor SB203580 impaired the changes mediated by PLS3 down-regulation in response to paclitaxel. Therefore, our study identifies PLS3 as a potential target for enhancing the p38 MAPK-mediated apoptosis induced by paclitaxel. Unlike paclitaxel, Abraxane was unable to induce strong apoptosis in the PLS3-silenced cells. As PLS3 was found to be involved in the process of endocytosis in breast cancer cells, the reliance of cellular Abraxane uptake on this process may render it not as efficient as paclitaxel in PLS3-depleted tumour cells. The finding that PLS3 could be a critical regulator of paclitaxel sensitivity may have important implications for breast cancer chemotherapy.
Collapse
Affiliation(s)
- Yan Ma
- a CAS Key Laboratory of Standardization and Measurement for Nanotechnology, CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety , CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology of China , Beijing , China
| | - Wenjia Lai
- a CAS Key Laboratory of Standardization and Measurement for Nanotechnology, CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety , CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology of China , Beijing , China
| | - Minzhi Zhao
- a CAS Key Laboratory of Standardization and Measurement for Nanotechnology, CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety , CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology of China , Beijing , China
| | - Chunyan Yue
- a CAS Key Laboratory of Standardization and Measurement for Nanotechnology, CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety , CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology of China , Beijing , China.,b Sino-Danish College , University of Chinese Academy of Sciences , Beijing , China
| | - Fanghao Shi
- a CAS Key Laboratory of Standardization and Measurement for Nanotechnology, CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety , CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology of China , Beijing , China.,b Sino-Danish College , University of Chinese Academy of Sciences , Beijing , China
| | - Ren Li
- a CAS Key Laboratory of Standardization and Measurement for Nanotechnology, CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety , CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology of China , Beijing , China.,c Academy for Advanced Interdisciplinary Studies , Peking University , Beijing , China.,d University of Chinese Academy of Sciences , Beijing , China
| | - Zhiyuan Hu
- a CAS Key Laboratory of Standardization and Measurement for Nanotechnology, CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety , CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology of China , Beijing , China.,b Sino-Danish College , University of Chinese Academy of Sciences , Beijing , China.,e Center for Neuroscience Research, School of Basic Medical Sciences , Fujian Medical University , Fuzhou , China
| |
Collapse
|
30
|
Li X, Wang L, Ma H. Betaine alleviates high glucose‑induced mesangial cell proliferation by inhibiting cell proliferation and extracellular matrix deposition via the AKT/ERK1/2/p38 MAPK pathway. Mol Med Rep 2019; 20:1754-1760. [PMID: 31257485 PMCID: PMC6625408 DOI: 10.3892/mmr.2019.10391] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2018] [Accepted: 05/14/2019] [Indexed: 12/16/2022] Open
Abstract
Diabetic nephropathy (DN) is a major cause of chronic renal failure in diabetic patients worldwide. Betaine, a zwitterionic quaternary ammonium salt compound, is involved in numerous biological processes. The present study aimed to investigate the effects of betaine on mouse mesangial cells (MMCs) cultured under high glucose (HG) conditions and its underlying mechanisms. MMCs were treated with betaine under HG conditions. Cell proliferation and the cell cycle distribution were investigated with an MTT assay and flow cytometry, respectively. Western blotting and reverse transcription‑quantitative polymerase chain reaction analyses were applied to respectively determine protein and mRNA expression levels. The results suggested that betaine decreased cell proliferation in a dose‑dependent manner, while G1‑phase arrest was significantly induced in MMCs. Compared with the control group, the expression levels of p21 and p27 decreased under HG conditions, but were reversed by betaine. Furthermore, the expression levels of fibronectin and type IV collagen were significantly decreased in cells treated with betaine compared with the HG group. Additionally, betaine decreased the phosphorylation of Akt, extracellular‑signal‑regulated kinase (Erk)1/2 and p38 mitogen‑activated protein kinase (MAPK), but was enhanced under HG conditions. Overall, the results of the present study indicated that betaine serves a protective role in HG‑induced MMCs by inhibiting cell proliferation and extracellular matrix deposition via regulating regulation of the Akt/Erk1/2/p38 MAPK signaling pathway.
Collapse
Affiliation(s)
- Xianhui Li
- Department of Traditional Chinese Medicine, Tianjin Key Laboratory of Artificial Cell, Tianjin Institute of Hepatobiliary Disease, Artificial Cell Engineering Technology Research Center of Public Health Ministry, Third Central Hospital of Tianjin, Tianjin 300170, P.R. China
| | - Li Wang
- Department of Basic Medicine, Tianjin Medical College, Tianjin 300222, P.R. China
| | - Huining Ma
- Department of Traditional Chinese Medicine, Tianjin 4th Centre Hospital, Tianjin 300140, P.R. China
| |
Collapse
|
31
|
Abdelhafez OM, Ahmed EY, Abdel Latif NA, Arafa RK, Abd Elmageed ZY, Ali HI. Design and molecular modeling of novel P38α MAPK inhibitors targeting breast cancer, synthesized from oxygen heterocyclic natural compounds. Bioorg Med Chem 2019; 27:1308-1319. [DOI: 10.1016/j.bmc.2019.02.027] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2018] [Revised: 01/30/2019] [Accepted: 02/14/2019] [Indexed: 01/06/2023]
|
32
|
Sahu V, Mohan A, Dey S. p38 MAP kinases: plausible diagnostic and prognostic serum protein marker of non small cell lung cancer. Exp Mol Pathol 2019; 107:118-123. [PMID: 30771292 DOI: 10.1016/j.yexmp.2019.01.009] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2018] [Revised: 01/05/2019] [Accepted: 01/23/2019] [Indexed: 12/14/2022]
Abstract
INTRODUCTION p38 MAPK signaling molecules plays a dual role in cancer, both progression and suppression. Elevated expression of p38α was reported in lung cancer tissue in rat model. Our objective was to explore the concentration of all 4 isoforms of p38MAPK in serum of Non Small Cell Lung Cancer (NSCLC). MATERIAL AND METHODS The blood samples were collected from 77 NSCLC patients, 52 ethically matched healthy controls and 18 follow up patients were collected as some patients expired and some discontinued the treatment. The concentration of all isoforms of p38 (p38α, p38β, p38γ, and p38δ) were evaluated by Surface Plasmon Resonance (SPR) technology. RESULT The levels of all isoforms of serum p38 were significantly elevated at pre-therapy compare to control. Only p38α expression was significantly associated with tumor stage and its expression reduced after treatment which is then validated by western blot. However, no changes were observed in other isoforms after therapy. CONCLUSION Our study revealed that, p38α is more efficient among all the isoform to predict the disease accurately and it can be concluded that p38 MAPK may be used as diagnostic as well as prognostic marker of NSCLC disease.
Collapse
Affiliation(s)
- Vishal Sahu
- Department of Biophysics, All India Institute of Medical Sciences, New Delhi, India
| | - Anant Mohan
- Department of Pulmonary Medicine and Sleep Disorder, All India Institute of Medical Sciences, New Delhi, India
| | - Sharmistha Dey
- Department of Biophysics, All India Institute of Medical Sciences, New Delhi, India.
| |
Collapse
|
33
|
Zhang D, Huang S, Mei H, Kevin M, Shi T, Chen L. Protein-ligand interaction fingerprints for accurate prediction of dissociation rates of p38 MAPK Type II inhibitors. Integr Biol (Camb) 2019; 11:53-60. [PMID: 30855664 DOI: 10.1093/intbio/zyz004] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2018] [Revised: 11/20/2018] [Accepted: 02/01/2019] [Indexed: 12/22/2022]
Abstract
Binding/unbinding kinetics are key determinants of drug potencies. However, there are still a lot of challenges in predicting kinetic properties during early-stage drug development. In this work, position-restrained molecular dynamics simulations combined with energy decomposition were applied to extract protein-ligand interaction (PLI) fingerprints along the unbinding pathway of 20 p38 mitogen-activated protein kinase (p38 MAPK) Type II inhibitors. The results showed that the electrostatic and/or van der Waals interaction fingerprints at three key positions can be used for accurate prediction of the dissociation rate constants (koff) of p38 MAPK Type II inhibitors. The strategy proposed in this paper can provide not only an efficient method of predicting the dissociation rates of the p38 MAPK Type II inhibitors, but also the atom-level mechanism of enthalpy-driven unbinding process.
Collapse
Affiliation(s)
- Duo Zhang
- Key Laboratory of Biorheological Science and Technology (Ministry of Education), Chongqing University, Chongqing, China
- College of Bioengineering, Chongqing University, Chongqing, China
| | - Shuheng Huang
- Key Laboratory of Biorheological Science and Technology (Ministry of Education), Chongqing University, Chongqing, China
- College of Bioengineering, Chongqing University, Chongqing, China
| | - Hu Mei
- Key Laboratory of Biorheological Science and Technology (Ministry of Education), Chongqing University, Chongqing, China
- College of Bioengineering, Chongqing University, Chongqing, China
| | | | - Tingting Shi
- College of Bioengineering, Chongqing University, Chongqing, China
| | - Linxin Chen
- College of Bioengineering, Chongqing University, Chongqing, China
| |
Collapse
|
34
|
Zhao J, Liu Y, Lin F, Wang W, Yang S, Ge Y, Chen PR. Bioorthogonal Engineering of Bacterial Effectors for Spatial-Temporal Modulation of Cell Signaling. ACS CENTRAL SCIENCE 2019; 5:145-152. [PMID: 30693333 PMCID: PMC6346392 DOI: 10.1021/acscentsci.8b00751] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/15/2018] [Indexed: 05/02/2023]
Abstract
The complicated and entangled cell signaling network is dynamically regulated by a wide array of enzymes such as kinases. It remains desirable but challenging to specifically modulate individual, endogenous kinases within a cell, particularly in a spatial-temporally controlled fashion. Current strategies toward regulating the intracellular functions of a kinase of interest either lack specificity or require genetic engineering that may perturb its physiological activity. Herein, we harnessed a bacterial effector OspF for optical and chemical modulation of the endogenous mitogen-activated protein kinase (MAPK) cascade in living cells and mice. The phospho-lyase OspF provided high specificity and spatial resolution toward the desired kinase such as the extracellular signal-regulated kinase (ERK), while the genetically encoded bioorthogonal decaging strategy enabled its temporal activation in living systems. The photocaged OspF (OspF*) was applied to dissect the subcellular signaling roles of ERK in nucleus as opposed to cytoplasm, while the chemically caged OspF (OspFc) was introduced into living mice to modulate ERK-mediated gene expression. Finally, our spatially and chemically controlled OspFc was further used to precisely tune immune responses in T cells. Together, our bioorthogonal engineering strategy on bacterial effectors offers a general tool to modulate cell signaling with high specificity and spatial-temporal resolution.
Collapse
Affiliation(s)
- Jingyi Zhao
- Beijing
National Laboratory for Molecular Sciences, Key Laboratory of Bioorganic
Chemistry and Molecular Engineering of Ministry of Education, College
of Chemistry and Molecular Engineering, Peking University, Beijing 100871, China
- Peking-Tsinghua
Center for Life Sciences, Peking University, Beijing 100871, China
| | - Yanjun Liu
- Beijing
National Laboratory for Molecular Sciences, Key Laboratory of Bioorganic
Chemistry and Molecular Engineering of Ministry of Education, College
of Chemistry and Molecular Engineering, Peking University, Beijing 100871, China
| | - Feng Lin
- Beijing
National Laboratory for Molecular Sciences, Key Laboratory of Bioorganic
Chemistry and Molecular Engineering of Ministry of Education, College
of Chemistry and Molecular Engineering, Peking University, Beijing 100871, China
- Academy
for Advanced Interdisciplinary Studies, Peking University, Beijing 100871, China
| | - Weixia Wang
- Beijing
National Laboratory for Molecular Sciences, Key Laboratory of Bioorganic
Chemistry and Molecular Engineering of Ministry of Education, College
of Chemistry and Molecular Engineering, Peking University, Beijing 100871, China
| | - Shaojun Yang
- Beijing
National Laboratory for Molecular Sciences, Key Laboratory of Bioorganic
Chemistry and Molecular Engineering of Ministry of Education, College
of Chemistry and Molecular Engineering, Peking University, Beijing 100871, China
| | - Yun Ge
- Beijing
National Laboratory for Molecular Sciences, Key Laboratory of Bioorganic
Chemistry and Molecular Engineering of Ministry of Education, College
of Chemistry and Molecular Engineering, Peking University, Beijing 100871, China
| | - Peng R. Chen
- Beijing
National Laboratory for Molecular Sciences, Key Laboratory of Bioorganic
Chemistry and Molecular Engineering of Ministry of Education, College
of Chemistry and Molecular Engineering, Peking University, Beijing 100871, China
- Peking-Tsinghua
Center for Life Sciences, Peking University, Beijing 100871, China
- Academy
for Advanced Interdisciplinary Studies, Peking University, Beijing 100871, China
- E-mail:
| |
Collapse
|
35
|
Zarredar H, Pashapour S, Farajnia S, Ansarin K, Baradaran B, Ahmadzadeh V, Safari F. Targeting the KRAS, p38α, and NF-κB in lung adenocarcinoma cancer cells: The effect of combining RNA interferences with a chemical inhibitor. J Cell Biochem 2019; 120:10670-10677. [PMID: 30656741 DOI: 10.1002/jcb.28357] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2018] [Accepted: 11/29/2018] [Indexed: 12/28/2022]
Abstract
BACKGROUND Lung cancer is the leading cause of cancer-related death with less than 5-year survival rate for both men and women worldwide. KRAS (Kirsten rat sarcoma), nuclear factor-κB (NF-κB), and mitogen-activated protein kinase (MAPK) signaling pathways have a critical role in the proliferation and progression of various cancers, including lung cancer. The p38 MAPK plays a different role in various tissue hence show a tissue-dependent behavior. It acts as an oncogene in some tissues while plays as a tumor suppressor in some other tissues. Also, KRAS and NF-κB act as an oncogene in various cancer. This study was dedicated to analyzing the combined effect of NF-κB inhibitor, specific KRAS, and p38α small interfering RNA (siRNA) in A549 cell line. MATERIALS AND METHODS The cytotoxic effects of p38α siRNA, KRAS siRNA, and NF-κB inhibitor were determined using the 3-(4,5-dimethylthiazol-2-yl)-2,5 diphenyl tetrazolium bromide (MTT) assay. Relative p38α, KRAS, and NF-κB messenger RNA (mRNA) levels were measured by quantitative reverse-transcription polymerase chain reaction. Induction of apoptosis by treatments was measured by fluorescence-activated cell sorting (FACS) analysis. RESULTS The expression of mRNA related to p38α and KRAS genes was reduced to 23.4% and 26.7%, respectively, after treatment with specific siRNAs. Also, MTT assay showed that the cell viability after treatment with p38α siRNA, KRAS siRNA, NF-κB inhibitor and their combination was reduced. FACS results indicated that p38α siRNA, KRAS siRNA, and NF-κB inhibitor, and their combination, reduced the population of live cells in comparison with the population of untreated control cells (99.5%). The results are expressed as mean ± SD (n = 3); *P < 0.05; ** P < 0.01; *** P < 0.001; **** P < 0.0001 vs control group. CONCLUSION The results of this study indicated that p38α, KRAS, and NF-κB signaling pathways might play an important role in the development and growth of lung cancer and might be a potential therapeutic target for treatment of lung cancer.
Collapse
Affiliation(s)
- Habib Zarredar
- Tuberculosis and Lung Disease Research Center, Tabriz University of Medical Science, Tabriz, Iran.,Students Research Committee, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Shadi Pashapour
- Tuberculosis and Lung Disease Research Center, Tabriz University of Medical Science, Tabriz, Iran.,Drug Applied Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Safar Farajnia
- Tuberculosis and Lung Disease Research Center, Tabriz University of Medical Science, Tabriz, Iran.,Biotechnology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Khalil Ansarin
- Tuberculosis and Lung Disease Research Center, Tabriz University of Medical Science, Tabriz, Iran
| | - Behzad Baradaran
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Vahideh Ahmadzadeh
- Biotechnology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran.,Drug Applied Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Fatemeh Safari
- Biotechnology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran.,Drug Applied Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| |
Collapse
|
36
|
Khedri A, Khaghani S, Kheirollah A, Babaahmadi-Rezaei H, Shadboorestan A, Zangooei M, Afra HS, Meshkani R, Ghahremani MH. Signaling Crosstalk of FHIT, p53, and p38 in etoposide-induced apoptosis in MCF-7 cells. J Cell Biochem 2019; 120:9125-9137. [PMID: 30614034 DOI: 10.1002/jcb.28188] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2018] [Accepted: 11/12/2018] [Indexed: 12/13/2022]
Abstract
Fragile histidine trail (FHIT) is a tumor suppressor in response to DNA damage which has been deleted in various tumors. However, the signaling mechanisms and interactions of FHIT with regard to apoptotic proteins including p53 and p38 in the DNA damage-induced apoptosis are not well described. In the present study, we used etoposide-induced DNA damage in MCF-7 as a model to address these crosstalks. The time course study showed that the expression of FHIT, p53, and p38MAPK started after 1 hour following etoposide treatment. FHIT overexpression led to increase p53 expression, p38 activation, and augmented apoptosis following etoposide-induced DNA damage compared to wild-type cells. However, FHIT knockdown blocked p53 expression, delayed p38 activation, and completely inhibited etoposide-induced apoptosis. Inhibition of p38 activity prevented induction of p53, FHIT, and apoptosis in this model. Thus, activation of p38 upon etoposide treatment leads to increase in FHIT and p53 expression. In p53 knockdown MCF-7, the FHIT induction was hampered but p38 activation was induced in lower doses of etoposide. In p53 knockdown cells, inhibition of p38 induced FHIT expression and apoptosis. Our data demonstrated that the exposure of MCF-7 cells to etoposide increases apoptosis through a mechanism involving the activation of the p38-FHIT-p53 pathway. Moreover, our findings suggest signaling interaction for these pathways may represent a promising therapy for breast cancer.
Collapse
Affiliation(s)
- Azam Khedri
- Department of Biochemistry, Faculty of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Shahnaz Khaghani
- Department of Biochemistry, Faculty of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Alireza Kheirollah
- Cellular and Molecular Research Center, Department of Clinical Biochemistry, Faculty of Medicine, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| | - Hossein Babaahmadi-Rezaei
- Hyperlipidemia Research Center, Department of Clinical Biochemistry, Faculty of Medicine, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| | - Amir Shadboorestan
- Department of Toxicology Pharmacology, Faculty of Pharmacy, Tehran University of Medical Sciences, Tehran, Iran
| | - Mohammad Zangooei
- Department of Biochemistry, Faculty of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Hajar Shokri Afra
- Department of Biochemistry, Faculty of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Reza Meshkani
- Department of Biochemistry, Faculty of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Mohammad Hossein Ghahremani
- Department of Toxicology Pharmacology, Faculty of Pharmacy, Tehran University of Medical Sciences, Tehran, Iran
| |
Collapse
|
37
|
Dabiri Y, Schmid A, Theobald J, Blagojevic B, Streciwilk W, Ott I, Wölfl S, Cheng X. A Ruthenium(II) N-Heterocyclic Carbene (NHC) Complex with Naphthalimide Ligand Triggers Apoptosis in Colorectal Cancer Cells via Activating the ROS-p38 MAPK Pathway. Int J Mol Sci 2018; 19:ijms19123964. [PMID: 30544880 PMCID: PMC6320930 DOI: 10.3390/ijms19123964] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2018] [Revised: 12/04/2018] [Accepted: 12/06/2018] [Indexed: 12/14/2022] Open
Abstract
The p38 MAPK pathway is known to influence the anti-tumor effects of several chemotherapeutics, including that of organometallic drugs. Previous studies have demonstrated the important role of p38 both as a regulator and a sensor of cellular reactive oxygen species (ROS) levels. Investigating the anti-cancer properties of novel 1,8-naphthalimide derivatives containing Rh(I) and Ru(II) N-heterocyclic carbene (NHC) ligands, we observed a profound induction of ROS by the complexes, which is most likely generated from mitochondria (mtROS). Further analyses revealed a rapid and consistent activation of p38 signaling by the naphthalimide-NHC conjugates, with the Ru(II) analogue—termed MC6—showing the strongest effect. In view of this, genetic as well as pharmacological inhibition of p38α, attenuated the anti-proliferative and pro-apoptotic effects of MC6 in HCT116 colon cancer cells, highlighting the involvement of this signaling molecule in the compound’s toxicity. Furthermore, the influence of MC6 on p38 signaling appeared to be dependent on ROS levels as treatment with general- and mitochondria-targeted anti-oxidants abrogated p38 activation in response to MC6 as well as the molecule’s cytotoxic- and apoptogenic response in HCT116 cells. Altogether, our results provide new insight into the molecular mechanisms of naphthalimide-metal NHC analogues via the ROS-induced activation of p38 MAPK, which may have therapeutic interest for the treatment of various cancer types.
Collapse
Affiliation(s)
- Yasamin Dabiri
- Institute of Pharmacy and Molecular Biotechnology, Heidelberg University, Im Neuenheimer Feld 364, 69120 Heidelberg, Germany.
| | - Alice Schmid
- Institute of Pharmacy and Molecular Biotechnology, Heidelberg University, Im Neuenheimer Feld 364, 69120 Heidelberg, Germany.
| | - Jannick Theobald
- Institute of Pharmacy and Molecular Biotechnology, Heidelberg University, Im Neuenheimer Feld 364, 69120 Heidelberg, Germany.
| | - Biljana Blagojevic
- Institute of Pharmacy and Molecular Biotechnology, Heidelberg University, Im Neuenheimer Feld 364, 69120 Heidelberg, Germany.
| | - Wojciech Streciwilk
- Institute of Medicinal and Pharmaceutical Chemistry, Technische Universität Braunschweig, Beethovenstraße 55, 38106 Braunschweig, Germany.
| | - Ingo Ott
- Institute of Medicinal and Pharmaceutical Chemistry, Technische Universität Braunschweig, Beethovenstraße 55, 38106 Braunschweig, Germany.
| | - Stefan Wölfl
- Institute of Pharmacy and Molecular Biotechnology, Heidelberg University, Im Neuenheimer Feld 364, 69120 Heidelberg, Germany.
| | - Xinlai Cheng
- Institute of Pharmacy and Molecular Biotechnology, Heidelberg University, Im Neuenheimer Feld 364, 69120 Heidelberg, Germany.
| |
Collapse
|
38
|
Mun JG, Kee JY, Han YH, Lee S, Park SH, Jeon HD, Hong SH. Galla Rhois water extract inhibits lung metastasis by inducing AMPK‑mediated apoptosis and suppressing metastatic properties of colorectal cancer cells. Oncol Rep 2018; 41:202-212. [PMID: 30365120 PMCID: PMC6278418 DOI: 10.3892/or.2018.6812] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2018] [Accepted: 10/09/2018] [Indexed: 12/12/2022] Open
Abstract
Galla Rhois is a commonly used medicine in East Asia for the treatment of several diseases. However, the effects of Galla Rhois on the metastasis of colorectal cancer (CRC) and the underlying molecular mechanisms have not been studied. We investigated the anti-metastatic properties of Galla Rhois water extract (GRWE) on metastatic CRC cells. The effect of GRWE on the viability of colon 26 (CT26) cells was evaluated using WST-8 assay. Annexin V assay and western blot analysis were performed to elucidate the underlying molecular mechanisms involved in apoptosis. GRWE suppressed viability of CT26 cells by inducing apoptosis through the cleavage of caspase-3 and PARP, downregulation of caspase-8, caspase-9, Bcl-2 and Bcl-xL, and upregulation of Bax. Metastatic phenotypes such as epithelial-mesenchymal transition (EMT), migration, and invasion of CRC cells were investigated by real-time reverse transcription polymerase chain reaction, wound healing assay, and matrigel invasion assay, respectively. Non-cytotoxic concentrations of GRWE inhibited EMT in CRC cells by regulating the expression of EMT markers. GRWE attenuated cell migration and invasion through the inhibition of matrix metalloproteinase (MMP)-2 and MMP-9 activity. Moreover, GRWE suppressed colorectal lung metastasis in vivo, suggestive of its potential application for the treatment of colorectal metastasis.
Collapse
Affiliation(s)
- Jeong-Geon Mun
- Department of Oriental Pharmacy, College of Pharmacy, Wonkwang‑Oriental Medicines Research Institute, Wonkwang University, Iksan, Jeonbuk 54538, Republic of Korea
| | - Ji-Ye Kee
- Department of Oriental Pharmacy, College of Pharmacy, Wonkwang‑Oriental Medicines Research Institute, Wonkwang University, Iksan, Jeonbuk 54538, Republic of Korea
| | - Yo-Han Han
- Department of Oriental Pharmacy, College of Pharmacy, Wonkwang‑Oriental Medicines Research Institute, Wonkwang University, Iksan, Jeonbuk 54538, Republic of Korea
| | - Sullim Lee
- Department of Life Sciences, College of Bio‑Nano Technology, Gachon University, Seongnam 13120, Republic of Korea
| | - Seong-Hwan Park
- Department of Oriental Pharmacy, College of Pharmacy, Wonkwang‑Oriental Medicines Research Institute, Wonkwang University, Iksan, Jeonbuk 54538, Republic of Korea
| | - Hee Dong Jeon
- Department of Oriental Pharmacy, College of Pharmacy, Wonkwang‑Oriental Medicines Research Institute, Wonkwang University, Iksan, Jeonbuk 54538, Republic of Korea
| | - Seung-Heon Hong
- Department of Oriental Pharmacy, College of Pharmacy, Wonkwang‑Oriental Medicines Research Institute, Wonkwang University, Iksan, Jeonbuk 54538, Republic of Korea
| |
Collapse
|
39
|
Md Aksam VK, Chandrasekaran VM, Pandurangan S. Topological alternate centrality measure capturing drug targets in the network of MAPK pathways. IET Syst Biol 2018; 12:226-232. [PMID: 30259868 PMCID: PMC8687289 DOI: 10.1049/iet-syb.2017.0058] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2017] [Revised: 04/04/2018] [Accepted: 04/30/2018] [Indexed: 12/18/2022] Open
Abstract
A new centrality of the nodes in the network is proposed called alternate centrality, which can isolate effective drug targets in the complex signalling network. Alternate centrality metric defined over the network substructure (four nodes - motifs). The nodes involving in alternative activation in the motifs gain in metric values. Targeting high alternative centrality nodes hypothesised to be destructive free to the network due to their alternative activation mechanism. Overlapping and crosstalk among the gene products in the conserved network of MAPK pathways selected for the study. In silico knock-out of high alternate centrality nodes causing rewiring in the network is investigated using MCF-7 breast cancer cell line-based data. Degree of top alternate centrality nodes lies between the degree of bridging and pagerank nodes. Node deletion of high alternate centrality on the centralities such as eccentricity, closeness, betweenness, stress, centroid and radiality causes low perturbation. The authors identified the following alternate centrality nodes ERK1, ERK2, MEKK2, MKK5, MKK4, MLK3, MLK2, MLK1, MEKK4, MEKK1, TAK1, P38alpha, ZAK, DLK, LZK, MLTKa/b and P38beta as efficient drug targets for breast cancer. Alternate centrality identifies effective drug targets and is free from intertwined biological processes and lethality.
Collapse
Affiliation(s)
- V K Md Aksam
- School of Advanced Sciences, VIT University, Vellore 632014, India
| | | | | |
Collapse
|
40
|
Anwar T, Arellano-Garcia C, Ropa J, Chen YC, Kim HS, Yoon E, Grigsby S, Basrur V, Nesvizhskii AI, Muntean A, Gonzalez ME, Kidwell KM, Nikolovska-Coleska Z, Kleer CG. p38-mediated phosphorylation at T367 induces EZH2 cytoplasmic localization to promote breast cancer metastasis. Nat Commun 2018; 9:2801. [PMID: 30022044 PMCID: PMC6051995 DOI: 10.1038/s41467-018-05078-8] [Citation(s) in RCA: 82] [Impact Index Per Article: 13.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2017] [Accepted: 05/31/2018] [Indexed: 12/19/2022] Open
Abstract
Overexpression of EZH2 in estrogen receptor negative (ER-) breast cancer promotes metastasis. EZH2 has been mainly studied as the catalytic component of the Polycomb Repressive Complex 2 (PRC2) that mediates gene repression by trimethylating histone H3 at lysine 27 (H3K27me3). However, how EZH2 drives metastasis despite the low H3K27me3 levels observed in ER- breast cancer is unknown. Here we show that in human invasive carcinomas and distant metastases, cytoplasmic EZH2 phosphorylated at T367 is significantly associated with ER- disease and low H3K27me3 levels. p38-mediated EZH2 phosphorylation at T367 promotes EZH2 cytoplasmic localization and potentiates EZH2 binding to vinculin and other cytoskeletal regulators of cell migration and invasion. Ectopic expression of a phospho-deficient T367A-EZH2 mutant is sufficient to inhibit EZH2 cytoplasmic expression, disrupt binding to cytoskeletal regulators, and reduce EZH2-mediated adhesion, migration, invasion, and development of spontaneous metastasis. These results point to a PRC2-independent non-canonical mechanism of EZH2 pro-metastatic function.
Collapse
MESH Headings
- Animals
- Breast Neoplasms/genetics
- Breast Neoplasms/mortality
- Breast Neoplasms/pathology
- Breast Neoplasms/therapy
- Carcinoma, Ductal, Breast/genetics
- Carcinoma, Ductal, Breast/mortality
- Carcinoma, Ductal, Breast/secondary
- Carcinoma, Ductal, Breast/therapy
- Cell Line, Tumor
- Cell Movement
- Enhancer of Zeste Homolog 2 Protein/antagonists & inhibitors
- Enhancer of Zeste Homolog 2 Protein/genetics
- Enhancer of Zeste Homolog 2 Protein/metabolism
- Estrogen Receptor alpha/genetics
- Estrogen Receptor alpha/metabolism
- Female
- Gene Expression Regulation, Neoplastic
- Heterografts
- Histones/genetics
- Histones/metabolism
- Humans
- Lung Neoplasms/genetics
- Lung Neoplasms/mortality
- Lung Neoplasms/secondary
- Lung Neoplasms/therapy
- Mice
- Mice, SCID
- Phosphorylation
- Polycomb Repressive Complex 2/genetics
- Polycomb Repressive Complex 2/metabolism
- RNA, Small Interfering/genetics
- RNA, Small Interfering/metabolism
- Survival Analysis
- Threonine
- p38 Mitogen-Activated Protein Kinases/antagonists & inhibitors
- p38 Mitogen-Activated Protein Kinases/genetics
- p38 Mitogen-Activated Protein Kinases/metabolism
Collapse
Affiliation(s)
- Talha Anwar
- Department of Pathology, University of Michigan Medical School, Ann Arbor, MI, 48109, USA
- Molecular Cellular and Pathology Training Program, University of Michigan, Ann Arbor, MI, 48109, USA
- Rogel Cancer Center, University of Michigan, Ann Arbor, MI, 48109, USA
- Medical Scientist Training Program, University of Michigan, Ann Arbor, MI, 48109, USA
| | - Caroline Arellano-Garcia
- Michigan Post-baccalaureate Research Education Program, University of Michigan, Ann Arbor, MI, 48109, USA
| | - James Ropa
- Department of Pathology, University of Michigan Medical School, Ann Arbor, MI, 48109, USA
- Molecular Cellular and Pathology Training Program, University of Michigan, Ann Arbor, MI, 48109, USA
| | - Yu-Chih Chen
- Rogel Cancer Center, University of Michigan, Ann Arbor, MI, 48109, USA
- Department of Electrical Engineering and Computer Science, University of Michigan, Ann Arbor, MI, 48109, USA
| | - Hong Sun Kim
- Department of Pathology, University of Michigan Medical School, Ann Arbor, MI, 48109, USA
- Rogel Cancer Center, University of Michigan, Ann Arbor, MI, 48109, USA
| | - Euisik Yoon
- Department of Electrical Engineering and Computer Science, University of Michigan, Ann Arbor, MI, 48109, USA
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, MI, 48109, USA
| | - Sierrah Grigsby
- Department of Pathology, University of Michigan Medical School, Ann Arbor, MI, 48109, USA
- Molecular Cellular and Pathology Training Program, University of Michigan, Ann Arbor, MI, 48109, USA
| | - Venkatesha Basrur
- Department of Pathology, University of Michigan Medical School, Ann Arbor, MI, 48109, USA
| | - Alexey I Nesvizhskii
- Department of Pathology, University of Michigan Medical School, Ann Arbor, MI, 48109, USA
| | - Andrew Muntean
- Department of Pathology, University of Michigan Medical School, Ann Arbor, MI, 48109, USA
| | - Maria E Gonzalez
- Department of Pathology, University of Michigan Medical School, Ann Arbor, MI, 48109, USA
- Rogel Cancer Center, University of Michigan, Ann Arbor, MI, 48109, USA
| | - Kelley M Kidwell
- Department of Biostatistics, University of Michigan, Ann Arbor, MI, 48109, USA
| | | | - Celina G Kleer
- Department of Pathology, University of Michigan Medical School, Ann Arbor, MI, 48109, USA.
- Rogel Cancer Center, University of Michigan, Ann Arbor, MI, 48109, USA.
| |
Collapse
|
41
|
Cánovas B, Igea A, Sartori AA, Gomis RR, Paull TT, Isoda M, Pérez-Montoyo H, Serra V, González-Suárez E, Stracker TH, Nebreda AR. Targeting p38α Increases DNA Damage, Chromosome Instability, and the Anti-tumoral Response to Taxanes in Breast Cancer Cells. Cancer Cell 2018; 33:1094-1110.e8. [PMID: 29805078 DOI: 10.1016/j.ccell.2018.04.010] [Citation(s) in RCA: 63] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/22/2017] [Revised: 01/18/2018] [Accepted: 04/25/2018] [Indexed: 12/18/2022]
Abstract
Breast cancer is the second leading cause of cancer-related death among women. Here we report a role for the protein kinase p38α in coordinating the DNA damage response and limiting chromosome instability during breast tumor progression, and identify the DNA repair regulator CtIP as a p38α substrate. Accordingly, decreased p38α signaling results in impaired ATR activation and homologous recombination repair, with concomitant increases in replication stress, DNA damage, and chromosome instability, leading to cancer cell death and tumor regression. Moreover, we show that pharmacological inhibition of p38α potentiates the effects of taxanes by boosting chromosome instability in murine models and patient-derived xenografts, suggesting the potential interest of combining p38α inhibitors with chemotherapeutic drugs that induce chromosome instability.
Collapse
Affiliation(s)
- Begoña Cánovas
- Institute for Research in Biomedicine (IRB Barcelona), Barcelona Institute of Science and Technology, Barcelona 08028, Spain
| | - Ana Igea
- Institute for Research in Biomedicine (IRB Barcelona), Barcelona Institute of Science and Technology, Barcelona 08028, Spain
| | - Alessandro A Sartori
- University of Zurich, Institute of Molecular Cancer Research, Zurich 8057, Switzerland
| | - Roger R Gomis
- Institute for Research in Biomedicine (IRB Barcelona), Barcelona Institute of Science and Technology, Barcelona 08028, Spain; ICREA, Pg. Lluís Companys 23, Barcelona 08010, Spain; Universitat de Barcelona and CIBERONC, Barcelona, Spain
| | - Tanya T Paull
- Howard Hughes Medical Institute, University of Texas at Austin, Austin, TX 78712, USA
| | - Michitaka Isoda
- Institute for Research in Biomedicine (IRB Barcelona), Barcelona Institute of Science and Technology, Barcelona 08028, Spain
| | - Héctor Pérez-Montoyo
- Cancer Epigenetics and Biology Program (PEBC), Bellvitge Biomedical Research Institute (IDIBELL), Barcelona 08908, Spain
| | - Violeta Serra
- Vall d'Hebron Institute of Oncology (VHIO), Barcelona 08035, Spain
| | - Eva González-Suárez
- Cancer Epigenetics and Biology Program (PEBC), Bellvitge Biomedical Research Institute (IDIBELL), Barcelona 08908, Spain
| | - Travis H Stracker
- Institute for Research in Biomedicine (IRB Barcelona), Barcelona Institute of Science and Technology, Barcelona 08028, Spain
| | - Angel R Nebreda
- Institute for Research in Biomedicine (IRB Barcelona), Barcelona Institute of Science and Technology, Barcelona 08028, Spain; ICREA, Pg. Lluís Companys 23, Barcelona 08010, Spain.
| |
Collapse
|
42
|
Carrascal MA, Silva M, Ramalho JS, Pen C, Martins M, Pascoal C, Amaral C, Serrano I, Oliveira MJ, Sackstein R, Videira PA. Inhibition of fucosylation in human invasive ductal carcinoma reduces E-selectin ligand expression, cell proliferation, and ERK1/2 and p38 MAPK activation. Mol Oncol 2018; 12:579-593. [PMID: 29215790 PMCID: PMC5928367 DOI: 10.1002/1878-0261.12163] [Citation(s) in RCA: 49] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2017] [Revised: 10/09/2017] [Accepted: 11/13/2017] [Indexed: 01/08/2023] Open
Abstract
Breast cancer tissue overexpresses fucosylated glycans, such as sialyl-Lewis X/A (sLeX/A ), and α-1,3/4-fucosyltransferases (FUTs) in relation to increased disease progression and metastasis. These glycans in tumor circulating cells mediate binding to vascular E-selectin, initiating tumor extravasation. However, their role in breast carcinogenesis is still unknown. Here, we aimed to define the contribution of the fucosylated structures, including sLeX/A , to cell adhesion, cell signaling, and cell proliferation in invasive ductal carcinomas (IDC), the most frequent type of breast cancer. We first analyzed expression of E-selectin ligands in IDC tissue and established primary cell cultures from the tissue. We observed strong reactivity with E-selectin and anti-sLeX/A antibodies in both IDC tissue and cell lines, and expression of α-1,3/4 FUTs FUT4, FUT5, FUT6, FUT10, and FUT11. To further assess the role of fucosylation in IDC biology, we immortalized a primary IDC cell line with human telomerase reverse transcriptase to create the 'CF1_T cell line'. Treatment with 2-fluorofucose (2-FF), a fucosylation inhibitor, completely abrogated its sLeX/A expression and dramatically reduced adherence of CF1_T cells to E-selectin under hemodynamic flow conditions. In addition, 2-FF-treated CF1_T cells showed a reduced migratory ability, as well as decreased cell proliferation rate. Notably, 2-FF treatment lowered the growth factor expression of CF1_T cells, prominently for FGF2, vascular endothelial growth factor, and transforming growth factor beta, and negatively affected activation of signal-regulating protein kinases 1 and 2 and p38 mitogen-activated protein kinase signaling pathways. These data indicate that fucosylation licenses several malignant features of IDC, such as cell adhesion, migration, proliferation, and growth factor expression, contributing to tumor progression.
Collapse
Affiliation(s)
- Mylène A. Carrascal
- UCIBIODepartamento Ciências da VidaFaculdade de Ciências e TecnologiaUniversidade Nova de LisboaPortugal
- CEDOCChronic Diseases Research CenterNOVA Medical School/Faculdade de Ciências MédicasUniversidade Nova de LisboaPortugal
| | - Mariana Silva
- CEDOCChronic Diseases Research CenterNOVA Medical School/Faculdade de Ciências MédicasUniversidade Nova de LisboaPortugal
- Departments of Dermatology and MedicineBrigham & Women's HospitalBostonMAUSA
- Harvard Medical SchoolProgram of Excellence in GlycosciencesBostonMAUSA
| | - José S. Ramalho
- CEDOCChronic Diseases Research CenterNOVA Medical School/Faculdade de Ciências MédicasUniversidade Nova de LisboaPortugal
| | - Cláudia Pen
- Centro Hospitalar de Lisboa CentralEPE – Serviço de Anatomia PatológicaLisbonPortugal
| | - Manuela Martins
- Centro Hospitalar de Lisboa CentralEPE – Serviço de Anatomia PatológicaLisbonPortugal
| | - Carlota Pascoal
- UCIBIODepartamento Ciências da VidaFaculdade de Ciências e TecnologiaUniversidade Nova de LisboaPortugal
| | - Constança Amaral
- UCIBIODepartamento Ciências da VidaFaculdade de Ciências e TecnologiaUniversidade Nova de LisboaPortugal
| | | | - Maria José Oliveira
- New Therapies GroupINEB‐Institute for Biomedical EngineeringPortoPortugal
- Instituto de Investigação e Inovação em SaúdeUniversidade do PortoPortugal
| | - Robert Sackstein
- Departments of Dermatology and MedicineBrigham & Women's HospitalBostonMAUSA
- Harvard Medical SchoolProgram of Excellence in GlycosciencesBostonMAUSA
| | - Paula A. Videira
- UCIBIODepartamento Ciências da VidaFaculdade de Ciências e TecnologiaUniversidade Nova de LisboaPortugal
- CEDOCChronic Diseases Research CenterNOVA Medical School/Faculdade de Ciências MédicasUniversidade Nova de LisboaPortugal
- CDG & Allies – PPAIN Congenital Disorders of Glycosylation Professionals and Patient Associations International NetworkCaparicaPortugal
| |
Collapse
|
43
|
García-Aranda M, Redondo M. Protein Kinase Targets in Breast Cancer. Int J Mol Sci 2017; 18:ijms18122543. [PMID: 29186886 PMCID: PMC5751146 DOI: 10.3390/ijms18122543] [Citation(s) in RCA: 47] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2017] [Revised: 11/20/2017] [Accepted: 11/22/2017] [Indexed: 01/10/2023] Open
Abstract
With 1.67 million new cases and 522,000 deaths in the year 2012, breast cancer is the most common type of diagnosed malignancy and the second leading cause of cancer death in women around the world. Despite the success of screening programs and the development of adjuvant therapies, a significant percentage of breast cancer patients will suffer a metastatic disease that, to this day, remains incurable and justifies the research of new therapies to improve their life expectancy. Among the new therapies that have been developed in recent years, the emergence of targeted therapies has been a milestone in the fight against cancer. Over the past decade, many studies have shown a causal role of protein kinase dysregulations or mutations in different human diseases, including cancer. Along these lines, cancer research has demonstrated a key role of many protein kinases during human tumorigenesis and cancer progression, turning these molecules into valid candidates for new targeted therapies. The subsequent discovery and introduction in 2001 of the kinase inhibitor imatinib, as a targeted treatment for chronic myelogenous leukemia, revolutionized cancer genetic pathways research, and lead to the development of multiple small-molecule kinase inhibitors against various malignancies, including breast cancer. In this review, we analyze studies published to date about novel small-molecule kinase inhibitors and evaluate if they would be useful to develop new treatment strategies for breast cancer patients.
Collapse
Affiliation(s)
- Marilina García-Aranda
- Biochemistry Department, Hospital Costa del Sol, Carretera de Cádiz km, 187, 29600 Marbella, Málaga, Spain.
| | - Maximino Redondo
- Biochemistry Department, Hospital Costa del Sol, Carretera de Cádiz km, 187, 29600 Marbella, Málaga, Spain.
- Biochemistry Department, Facultad de Medicina de la Universidad de Málaga, Bulevar Louis Pasteur 32, 29010 Málaga, Spain.
| |
Collapse
|
44
|
Mora Vidal R, Regufe da Mota S, Hayden A, Markham H, Douglas J, Packham G, Crabb SJ. Epidermal Growth Factor Receptor Family Inhibition Identifies P38 Mitogen-activated Protein Kinase as a Potential Therapeutic Target in Bladder Cancer. Urology 2017; 112:225.e1-225.e7. [PMID: 29154981 DOI: 10.1016/j.urology.2017.10.041] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2017] [Revised: 09/29/2017] [Accepted: 10/28/2017] [Indexed: 11/30/2022]
Abstract
OBJECTIVE To investigate perturbations in downstream signaling pathway activation and potential resistance mechanisms to epidermal growth factor receptor (EGFR) or human epidermal growth factor receptor 2 (HER2) inhibition in cell line models of bladder cancer. METHODS We undertook a structured screening approach by phosphokinase array, followed by validation steps, to detect activated downstream signaling pathway nodes after therapeutic inhibition of EGFR or HER2 in bladder cancer cell lines. RESULTS Erlotinib treatment of RT112 cells induced phosphorylation of 9 activated phosphoprotein targets (p38 mitogen-activated protein kinase [MAPK] [Thr180/Tyr182], GSK-3α/β [Ser21/9], MEK1/2 [Ser218/222, Ser222/226], Akt (protein kinase B) [Ser473], TOR [target of rapamycin] [Ser2448], Src [Tyr419], p27 [Thr198], p27 [Thr157], and PLCγ-1 [Tyr783]), whereas STAT4 (signal transducer and activator of transcription 4) (Tyr693) phosphorylation was reduced. Of these, p38 MAPK phosphorylation was confirmed to occur in response to inhibition of either EGFR or HER2 signaling through multiple validation steps, including differing bladder cancer cell lines (RT112, UM-UC-3, and T24) and methods of receptor pathway inhibition (erlotinib, lapatinib, and siRNA depletion of EGFR or HER2). Chemical inhibition of p38 MAPK with SB203580 led to inhibition of proliferation in RT112, UM-UC-3, and T24 cell lines (IC50 20.85, 76.78, and 79.12 µM, respectively). Fractional effect analyses indicated a synergistic interaction for inhibition of cell proliferation when combining SB203580 with lapatinib. CONCLUSION p38 MAPK is a potential therapeutic target in bladder cancer and this strategy warrants further development in this disease. It may also allow combination therapy strategies to be developed in conjunction with EGFR or HER2 inhibition.
Collapse
Affiliation(s)
- Regina Mora Vidal
- Cancer Sciences Unit, University of Southampton, Southampton, United Kingdom
| | | | - Annette Hayden
- Cancer Sciences Unit, University of Southampton, Southampton, United Kingdom
| | - Hannah Markham
- Department of Histopathology, University Hospital Southampton NHS Foundation Trust, Southampton, United Kingdom
| | - James Douglas
- Cancer Sciences Unit, University of Southampton, Southampton, United Kingdom
| | - Graham Packham
- Cancer Sciences Unit, University of Southampton, Southampton, United Kingdom
| | - Simon J Crabb
- Cancer Sciences Unit, University of Southampton, Southampton, United Kingdom.
| |
Collapse
|
45
|
Zhao D, Tahaney WM, Mazumdar A, Savage MI, Brown PH. Molecularly targeted therapies for p53-mutant cancers. Cell Mol Life Sci 2017; 74:4171-4187. [PMID: 28643165 PMCID: PMC5664959 DOI: 10.1007/s00018-017-2575-0] [Citation(s) in RCA: 64] [Impact Index Per Article: 9.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2017] [Revised: 05/30/2017] [Accepted: 06/15/2017] [Indexed: 02/08/2023]
Abstract
The tumor suppressor p53 is lost or mutated in approximately half of human cancers. Mutant p53 not only loses its anti-tumor transcriptional activity, but also often acquires oncogenic functions to promote tumor proliferation, invasion, and drug resistance. Traditional strategies have been taken to directly target p53 mutants through identifying small molecular compounds to deplete mutant p53, or to restore its tumor suppressive function. Accumulating evidence suggest that cancer cells with mutated p53 often exhibit specific functional dependencies on secondary genes or pathways to survive, providing alternative targets to indirectly treat p53-mutant cancers. Targeting these genes or pathways, critical for survival in the presence of p53 mutations, holds great promise for cancer treatment. In addition, mutant p53 often exhibits novel gain-of-functions to promote tumor growth and metastasis. Here, we review and discuss strategies targeting mutant p53, with focus on targeting the mutant p53 protein directly, and on the progress of identifying genes and pathways required in p53-mutant cells.
Collapse
Affiliation(s)
- Dekuang Zhao
- Department of Clinical Cancer Prevention, University of Texas MD Anderson Cancer Center, 1515 Holcombe Blvd, Unit Number: 1360, Room Number: CPB6.3468, Houston, TX, 77030, USA
| | - William M Tahaney
- Department of Clinical Cancer Prevention, University of Texas MD Anderson Cancer Center, 1515 Holcombe Blvd, Unit Number: 1360, Room Number: CPB6.3468, Houston, TX, 77030, USA
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX, USA
| | - Abhijit Mazumdar
- Department of Clinical Cancer Prevention, University of Texas MD Anderson Cancer Center, 1515 Holcombe Blvd, Unit Number: 1360, Room Number: CPB6.3468, Houston, TX, 77030, USA
| | - Michelle I Savage
- Department of Clinical Cancer Prevention, University of Texas MD Anderson Cancer Center, 1515 Holcombe Blvd, Unit Number: 1360, Room Number: CPB6.3468, Houston, TX, 77030, USA
| | - Powel H Brown
- Department of Clinical Cancer Prevention, University of Texas MD Anderson Cancer Center, 1515 Holcombe Blvd, Unit Number: 1360, Room Number: CPB6.3468, Houston, TX, 77030, USA.
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX, USA.
| |
Collapse
|
46
|
Growth differentiation factor 15 mediates epithelial mesenchymal transition and invasion of breast cancers through IGF-1R-FoxM1 signaling. Oncotarget 2017; 8:94393-94406. [PMID: 29212236 PMCID: PMC5706882 DOI: 10.18632/oncotarget.21765] [Citation(s) in RCA: 38] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2017] [Accepted: 09/15/2017] [Indexed: 02/07/2023] Open
Abstract
Expression of the inflammatory cytokine growth differentiation factor 15 (GDF15) is significantly elevated in many tumor types in association with epithelial mesenchymal transition (EMT), drug resistance, and progressive disease. However, few studies have examined GDF15 expression, signaling, or function in breast cancer. In the current study, we demonstrate that GDF15 is associated with high tumor grade, ER-negativity, and HER2 overexpression in patients with breast cancer. Stable overexpression of GDF15 upregulates expression of mesenchymal markers and transcription factors, including FoxM1, and increases cellular invasion. GDF15 stable clones and breast cancer cells stimulated with recombinant human GDF15 (rhGDF15) demonstrate activation of insulin-like growth factor-1 receptor (IGF-1R), EMT, and invasion. Pharmacologic inhibition of IGF-1R reduces GDF15-mediated EMT and invasion in stable clones, and FoxM1 knockdown rescues invasion and EMT in GDF15 stable clones and rhGDF15-stimulated cells. These data suggest that IGF-1R-FoxM1 signaling is a potential mechanism through which GDF15 drives EMT and invasion of breast cancers. Further, GDF15 knockdown significantly inhibits invasion of HER2-overexpressing and triple-negative breast cancer cells, supporting further preclinical investigation of GDF15-targeted therapies.
Collapse
|
47
|
Wada M, Canals D, Adada M, Coant N, Salama MF, Helke KL, Arthur JS, Shroyer KR, Kitatani K, Obeid LM, Hannun YA. P38 delta MAPK promotes breast cancer progression and lung metastasis by enhancing cell proliferation and cell detachment. Oncogene 2017; 36:6649-6657. [PMID: 28783172 PMCID: PMC5746050 DOI: 10.1038/onc.2017.274] [Citation(s) in RCA: 53] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2017] [Revised: 06/30/2017] [Accepted: 07/03/2017] [Indexed: 12/18/2022]
Abstract
The protein p38 mitogen-activated protein kinase delta isoform (p38δ) is a poorly studied member of the MAPK family. Data analysis from The Cancer Genome Atlas (TCGA) database revealed that p38δ is highly expressed in all types of human breast cancers. Using a human breast cancer tissue array, we confirmed elevation in cancer tissue. The breast cancer mouse model, MMTV-PyMT (PyMT), developed breast tumors with lung metastasis; however, mice deleted in p38δ (PyMT/p38δ−/−) exhibited delayed primary tumor formation and highly reduced lung metastatic burden. At the cellular level, we demonstrate that targeting of p38δ in breast cancer cells, MCF-7 and MDA-MB-231 resulted in a reduced rate of cell proliferation. Additionally, cells lacking p38δ also displayed an increased cell-matrix adhesion and reduced cell detachment. This effect on cell adhesion was molecularly supported by the regulation of the focal adhesion kinase (FAK) by p38δ in the human breast cell lines. These studies define a previously unappreciated role for p38δ in breast cancer development and evolution by regulating tumor growth and altering metastatic properties.
Collapse
Affiliation(s)
- M Wada
- Department of Medicine, Stony Brook University, Stony Brook, NY, USA
| | - D Canals
- Department of Medicine, Stony Brook University, Stony Brook, NY, USA.,Stony Brook Cancer Center, Stony Brook University, Stony Brook, NY, USA
| | - M Adada
- Department of Medicine, Stony Brook University, Stony Brook, NY, USA.,Stony Brook Cancer Center, Stony Brook University, Stony Brook, NY, USA
| | - N Coant
- Department of Medicine, Stony Brook University, Stony Brook, NY, USA
| | - M F Salama
- Department of Medicine, Stony Brook University, Stony Brook, NY, USA.,Faculty of Veterinary Medicine, Department of Biochemistry, Mansoura University, Mansoura, Egypt
| | - K L Helke
- Department of Comparative Medicine, Medical University of South Carolina, Charleston, SC, USA
| | - J S Arthur
- MRC Protein Phosphorylation Unit, School of Life Sciences, University of Dundee, Dundee, UK
| | - K R Shroyer
- Department of Pathology, Stony Brook University, Stony Brook, NY, USA
| | - K Kitatani
- Tohoku Medical Megabank Organization and Department of Obstetrics and Gynecology, Tohoku University Graduate School of Medicine, Sendai, Japan
| | - L M Obeid
- Department of Medicine, Stony Brook University, Stony Brook, NY, USA.,Stony Brook Cancer Center, Stony Brook University, Stony Brook, NY, USA.,Northport VA Medical Center, Northport, NY, USA
| | - Y A Hannun
- Department of Medicine, Stony Brook University, Stony Brook, NY, USA.,Stony Brook Cancer Center, Stony Brook University, Stony Brook, NY, USA
| |
Collapse
|
48
|
Cai X, Cao C, Li J, Chen F, Zhang S, Liu B, Zhang W, Zhang X, Ye L. Inflammatory factor TNF-α promotes the growth of breast cancer via the positive feedback loop of TNFR1/NF-κB (and/or p38)/p-STAT3/HBXIP/TNFR1. Oncotarget 2017; 8:58338-58352. [PMID: 28938560 PMCID: PMC5601656 DOI: 10.18632/oncotarget.16873] [Citation(s) in RCA: 61] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2016] [Accepted: 03/15/2017] [Indexed: 01/07/2023] Open
Abstract
In the connection between inflammation and cancer development, tumor necrosis factor-alpha (TNF-α) contributes to the tumorigenesis. However, the underlying mechanism remains poorly understood. In this study, we report that TNF-α enhances the growth of breast cancer through up-regulation of oncoprotein hepatitis B X-interacting protein (HBXIP). Our data showed that the levels of TNF-α were positively related to those of HBXIP in clinical breast cancer tissues. Moreover, TNF-α could up-regulate HBXIP in breast cancer cells. Interestingly, silencing of TNF-α receptor 1 (TNFR1) blocked the effect of TNF-α on HBXIP. Mechanistically, we revealed that TNF-α could increase the activities of HBXIP promoter through activating transcriptional factor signal transducer and activator of transcription 3 (STAT3). In addition, nuclear factor kappa B (NF-κB) and/or p38 signaling increased the levels of p-STAT3 in the cells. Strikingly, HBXIP could also up-regulate TNFR1, forming a positive feedback loop of TNFR1/NF-κB (and/or p38)/p-STAT3/HBXIP/TNFR1. Notably, TNF-α was able to up-regulate TNFR1 through driving the loop. In function, we demonstrated that the knockdown of HBXIP remarkably abolished the growth of breast cancer mediated by TNF-α in vitro and in vivo. Thus, we conclude that TNF-α promotes the growth of breast cancer through the positive feedback loop of TNFR1/NF-κB (and/or p38)/p-STAT3/HBXIP/TNFR1.Our finding provides new insights into the mechanism by which TNF-α drives oncoprotein HBXIP in the development of breast cancer.
Collapse
Affiliation(s)
- Xiaoli Cai
- State Key Laboratory of Medicinal Chemical Biology, Department of Biochemistry, College of Life Sciences, Nankai University, Tianjin 300071, China
| | - Can Cao
- State Key Laboratory of Medicinal Chemical Biology, Department of Biochemistry, College of Life Sciences, Nankai University, Tianjin 300071, China
| | - Jiong Li
- State Key Laboratory of Medicinal Chemical Biology, Department of Cancer Research, College of Life Sciences, Nankai University, Tianjin 300071, China
| | - Fuquan Chen
- State Key Laboratory of Medicinal Chemical Biology, Department of Cancer Research, College of Life Sciences, Nankai University, Tianjin 300071, China
| | - Shuqin Zhang
- State Key Laboratory of Medicinal Chemical Biology, Department of Cancer Research, College of Life Sciences, Nankai University, Tianjin 300071, China
| | - Bowen Liu
- State Key Laboratory of Medicinal Chemical Biology, Department of Biochemistry, College of Life Sciences, Nankai University, Tianjin 300071, China
| | - Weiying Zhang
- State Key Laboratory of Medicinal Chemical Biology, Department of Biochemistry, College of Life Sciences, Nankai University, Tianjin 300071, China
| | - Xiaodong Zhang
- State Key Laboratory of Medicinal Chemical Biology, Department of Cancer Research, College of Life Sciences, Nankai University, Tianjin 300071, China
| | - Lihong Ye
- State Key Laboratory of Medicinal Chemical Biology, Department of Biochemistry, College of Life Sciences, Nankai University, Tianjin 300071, China
| |
Collapse
|
49
|
Amin KM, Syam YM, Anwar MM, Ali HI, Abdel-Ghani TM, Serry AM. Synthesis and molecular docking studies of new furochromone derivatives as p38α MAPK inhibitors targeting human breast cancer MCF-7 cells. Bioorg Med Chem 2017; 25:2423-2436. [PMID: 28291685 DOI: 10.1016/j.bmc.2017.02.065] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2017] [Accepted: 02/27/2017] [Indexed: 12/12/2022]
Abstract
Based on the reported high expression of p38α MAP kinase in invasive breast cancers and the activity of different functionalized chromone derivatives as p38α inhibitors, a new set of 4,9-dimethoxy/4-methoxy-7-methyl-5-oxo-5H-furo[3,2-g]chromone derivatives were efficiently synthesized aiming to introduce new p38α MAP kinase suppressors as new anti-breast cancer tools. Using GOLD program, molecular docking study of the target compounds into p38α MAP kinase binding pocket was performed to highlight their scores, mode of binding and the important interactions to the amino acid residues of the enzyme. MTT assay investigated that fifteen target compounds produced marked cytotoxic potency higher than that obtained by Doxorubicin against MCF-7 cancer cells of IC50 values ranging from 0.007 to 0.17μM vs IC50; 0.62μM of doxorubicin. Eleven selected compounds were evaluated for their inhibitory potency against p38α MAPK kinase. The derivatives IVa, Va,b, VIa, IXb and XIIIa represented significant activity (IC50; 0.19-0.67μM) comparing to the reference drug SB203580 (IC50; 0.50μM). In virtue of its promising cytotoxic and p38α MAP kinase inhibition potency, the furochromone derivative IXb was selected as a representative example to investigate its mechanistic effects on cell cycle progression and induction of apoptosis in MCF-7 cell lines. The results showed that the compound IXb induced cell cycle cessation at G2/M phase preventing its mitotic cycle, alongside its noteworthy activation of caspases-9 and -3 which might mediate the apoptosis of MCF-7 cells.
Collapse
Affiliation(s)
- Kamelia M Amin
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, Cairo University, Egypt
| | - Yasmin M Syam
- Department of Therapeutical Chemistry, National Research Centre, Dokki, Cairo 12622, Egypt.
| | - Manal M Anwar
- Department of Therapeutical Chemistry, National Research Centre, Dokki, Cairo 12622, Egypt
| | - Hamed I Ali
- Pharmaceutical Sciences Dept., Irma Lerma Rangel College of Pharmacy, Texas A&M Health Science Center, TX, USA
| | | | | |
Collapse
|
50
|
Huth HW, Santos DM, Gravina HD, Resende JM, Goes AM, de Lima ME, Ropert C. Upregulation of p38 pathway accelerates proliferation and migration of MDA-MB-231 breast cancer cells. Oncol Rep 2017; 37:2497-2505. [PMID: 28260101 DOI: 10.3892/or.2017.5452] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2016] [Accepted: 01/13/2017] [Indexed: 11/06/2022] Open
Abstract
Tumor cells capture the signaling pathways used by normal tissue to promote their own survival and dissemination and among them, the NF-κB and MAPK pathways (ERK, JNK and p38). MAPK activation has ambiguous effects on tumor cell fate depending on cell type, cancer stage and the engaged MAPK isoforms. A synthetic peptide named LyeTx II, derived from the venom of the Brazilian spider Lycosa erythrognatha, was capable of increasing MDA-MB-231 aggressive breast cancer cell proliferation as indicated by MTT and BrdU (5-bromo-2'-deoxyuridine) incorporation assay and cell migration. A correlation has been established between the accelerated proliferation and migration observed in the presence of LyeTx II and the upregulation of p38 MAPK phosphorylation. The use of the selective inhibitor of p38α/β (SB203580) abrogated the peptide effect in MDA-MB-231 cells. Besides, an augment of the canonical NF-κB pathway activation considered as crucial in cancer progression was noted after cell incubation with LyeTx II. Importantly, activation of p38 and NF-κB pathways was dependent on TAK1 activity. Together, these data suggest that TAK1-p38 pathway may represent an interesting target for treatment of aggressive breast cancers.
Collapse
Affiliation(s)
- Hugo W Huth
- Department of Physiology and Pharmacology, Federal University of Minas Gerais, Belo Horizonte, Minas Gerais 31270-910, Brazil
| | - Daniel M Santos
- Department of Biochemistry and Immunology, Federal University of Minas Gerais, Belo Horizonte, Minas Gerais 31270-910, Brazil
| | - Humberto D Gravina
- Department of Biochemistry and Immunology, Federal University of Minas Gerais, Belo Horizonte, Minas Gerais 31270-910, Brazil
| | - Jarbas M Resende
- Department of Chemistry, Federal University of Minas Gerais, Belo Horizonte, Minas Gerais 31270-910, Brazil
| | - Alfredo M Goes
- Department of Biochemistry and Immunology, Federal University of Minas Gerais, Belo Horizonte, Minas Gerais 31270-910, Brazil
| | - Maria Elena de Lima
- Department of Biochemistry and Immunology, Federal University of Minas Gerais, Belo Horizonte, Minas Gerais 31270-910, Brazil
| | - Catherine Ropert
- Department of Biochemistry and Immunology, Federal University of Minas Gerais, Belo Horizonte, Minas Gerais 31270-910, Brazil
| |
Collapse
|