1
|
Gorgoń S, Billing O, Eriksson AU, Hemmingsson O. A Drug Discovery Pipeline for MAPK/ERK Pathway Inhibitors in Caenorhabditis elegans. CANCER RESEARCH COMMUNICATIONS 2024; 4:2454-2462. [PMID: 39212544 PMCID: PMC11409438 DOI: 10.1158/2767-9764.crc-24-0221] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/15/2024] [Revised: 07/03/2024] [Accepted: 08/28/2024] [Indexed: 09/04/2024]
Abstract
Oncogenic signaling through the MAPK/ERK pathway drives tumor progression in many cancers. Although targeted MAPK/ERK pathway inhibitors improve survival in selected patients, most tumors are resistant. New drugs could be identified in small-animal models that, unlike in vitro models, can address oral uptake, compound bioavailability, and toxicity. This requires pharmacologic conformity between human and model MAPK/ERK pathways and available phenotypic assays. In this study, we test if the conserved MAPK/ERK pathway in Caenorhabditis elegans could serve as a model for pharmacological inhibition and develop in vivo pipelines for high-throughput compound screens. Using fluorescence-based image analysis of vulva development as a readout for MAPK/ERK activity, we obtained excellent assay Z-scores for the MEK inhibitors trametinib (Z = 0.95), mirdametinib (Z = 0.93), and AZD8330 (Z = 0.87), as well as the ERK inhibitor temuterkib (Z = 0.86). The throughput was 800 wells per hour, with an average seed density of 25.5 animals per well. Readouts included drug efficacy, toxicity, and pathway specificity, which was tested against pathway activating upstream (lin-15)- and downstream (lin-1) mutants. To validate the model in a high-throughput setting, we screened a blinded library of 433 anticancer compounds and identified four MEK inhibitors among seven positive hits. Our results highlight a high degree of pharmacological conformity between C. elegans and human MAPK/ERK pathways, and the presented high-throughput pipeline may discover and characterize novel inhibitors in vivo. SIGNIFICANCE Many tumors depend on MAPK/ERK signaling to sustain growth, avoid cell death, and metastasize. We show that specific and clinically relevant MAPK/ERK signaling inhibitors can be discovered in vivo with a high-throughput screening pipeline in small animals.
Collapse
Affiliation(s)
- Szymon Gorgoń
- Department of Diagnostics and Intervention, Surgery, Umeå University, Umeå, Sweden.
| | - Ola Billing
- Department of Diagnostics and Intervention, Surgery, Umeå University, Umeå, Sweden.
| | - Anna U. Eriksson
- Chemical Biology Consortium Sweden, Umeå University, Umeå, Sweden.
| | - Oskar Hemmingsson
- Department of Diagnostics and Intervention, Surgery, Umeå University, Umeå, Sweden.
- Wallenberg Centre for Molecular Medicine, Umeå University, Umeå, Sweden.
| |
Collapse
|
2
|
Makhoul P, Galas S, Paniagua-Gayraud S, Deleuze-Masquefa C, Hajj HE, Bonnet PA, Richaud M. Uncovering the Molecular Pathways Implicated in the Anti-Cancer Activity of the Imidazoquinoxaline Derivative EAPB02303 Using a Caenorhabditis elegans Model. Int J Mol Sci 2024; 25:7785. [PMID: 39063027 PMCID: PMC11277376 DOI: 10.3390/ijms25147785] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2024] [Revised: 07/05/2024] [Accepted: 07/14/2024] [Indexed: 07/28/2024] Open
Abstract
Imiqualines are analogues of the immunomodulatory drug imiquimod. EAPB02303, the lead of the second-generation imiqualines, is characterized by significant anti-tumor effects with IC50s in the nanomolar range. We used Caenorhabditis elegans transgenic and mutant strains of two key signaling pathways (PI3K-Akt and Ras-MAPK) disrupted in human cancers to investigate the mode of action of EAPB02303. The ability of this imiqualine to inhibit the insulin/IGF1 signaling (IIS) pathway via the PI3K-Akt kinase cascade was explored through assessing the lifespan of wild-type worms. Micromolar doses of EAPB02303 significantly enhanced longevity of N2 strain and led to the nuclear translocation and subsequent activation of transcription factor DAF-16, the only forkhead box transcription factor class O (Fox O) homolog in C. elegans. Moreover, EAPB02303 significantly reduced the multivulva phenotype in let-60/Ras mutant strains MT2124 and MT4698, indicative of its mode of action through the Ras pathway. In summary, we showed that EAPB02303 potently reduced the activity of IIS and Ras-MAPK signaling in C. elegans. Our results revealed the mechanism of action of EAPB02303 against human cancers associated with hyperactivated IIS pathway and oncogenic Ras mutations.
Collapse
Affiliation(s)
- Perla Makhoul
- Institut des Biomolécules Max Mousseron (IBMM), UMR 5247, CNRS, ENSCM, Université de Montpellier, 34090 Montpellier, France; (P.M.); (S.G.); (S.P.-G.); (C.D.-M.)
- Department of Biology, Faculty of Sciences, GSBT Laboratory, Lebanese University, R. Hariri Campus, Hadath 1533, Lebanon
| | - Simon Galas
- Institut des Biomolécules Max Mousseron (IBMM), UMR 5247, CNRS, ENSCM, Université de Montpellier, 34090 Montpellier, France; (P.M.); (S.G.); (S.P.-G.); (C.D.-M.)
| | - Stéphanie Paniagua-Gayraud
- Institut des Biomolécules Max Mousseron (IBMM), UMR 5247, CNRS, ENSCM, Université de Montpellier, 34090 Montpellier, France; (P.M.); (S.G.); (S.P.-G.); (C.D.-M.)
| | - Carine Deleuze-Masquefa
- Institut des Biomolécules Max Mousseron (IBMM), UMR 5247, CNRS, ENSCM, Université de Montpellier, 34090 Montpellier, France; (P.M.); (S.G.); (S.P.-G.); (C.D.-M.)
| | - Hiba El Hajj
- Department of Experimental Pathology, Immunology and Microbiology, Faculty of Medicine, American University of Beirut, Riad El-Solh, P.O. Box 11-0236, Beirut 1107, Lebanon;
| | - Pierre-Antoine Bonnet
- Institut des Biomolécules Max Mousseron (IBMM), UMR 5247, CNRS, ENSCM, Université de Montpellier, 34090 Montpellier, France; (P.M.); (S.G.); (S.P.-G.); (C.D.-M.)
| | - Myriam Richaud
- Institut des Biomolécules Max Mousseron (IBMM), UMR 5247, CNRS, ENSCM, Université de Montpellier, 34090 Montpellier, France; (P.M.); (S.G.); (S.P.-G.); (C.D.-M.)
| |
Collapse
|
3
|
Uram Ł, Wróbel K, Walczak M, Szymaszek Ż, Twardowska M, Wołowiec S. Exploring the Potential of Lapatinib, Fulvestrant, and Paclitaxel Conjugated with Glycidylated PAMAM G4 Dendrimers for Cancer and Parasite Treatment. Molecules 2023; 28:6334. [PMID: 37687164 PMCID: PMC10489794 DOI: 10.3390/molecules28176334] [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: 07/16/2023] [Revised: 08/06/2023] [Accepted: 08/22/2023] [Indexed: 09/10/2023] Open
Abstract
Fulvestrant (F), lapatinib (L), and paclitaxel (P) are hydrophobic, anticancer drugs used in the treatment of estrogen receptor (ER) and epidermal growth factor receptor (EGFR)-positive breast cancer. In this study, glycidylated PAMAM G4 dendrimers, substituted with F, L, and/or P and targeting tumor cells, were synthesized and characterized, and their antitumor activity against glioma U-118 MG and non-small cell lung cancer A549 cells was tested comparatively with human non-tumorogenic keratinocytes (HaCaT). All cell lines were ER+ and EGFR+. In addition, the described drugs were tested in the context of antinematode therapy on C. elegans. The results show that the water-soluble conjugates of G4P, G4F, G4L, and G4PFL actively entered the tested cells via endocytosis due to the positive zeta potential (between 13.57-40.29 mV) and the nanoparticle diameter of 99-138 nm. The conjugates of G4P and G4PFL at nanomolar concentrations were the most active, and the least active conjugate was G4F. The tested conjugates inhibited the proliferation of HaCaT and A549 cells; in glioma cells, cytotoxicity was associated mainly with cell damage (mitochondria and membrane transport). The toxicity of the conjugates was proportional to the number of drug residues attached, with the exception of G4L; its action was two- and eight-fold stronger against glioma and keratinocytes, respectively, than the equivalent of lapatinib alone. Unfortunately, non-cancer HaCaT cells were the most sensitive to the tested constructs, which forced a change in the approach to the use of ER and EGFR receptors as a goal in cancer therapy. In vivo studies on C. elegans have shown that all compounds, most notably G4PFL, may be potentially useful in anthelmintic therapy.
Collapse
Affiliation(s)
- Łukasz Uram
- Faculty of Chemistry, Rzeszów University of Technology, 6 Powstańcow Warszawy Ave., 35-959 Rzeszów, Poland; (Ł.U.); (M.W.); (Ż.S.); (M.T.)
| | - Konrad Wróbel
- Medical College, Rzeszów University, 1a Warzywna Street, 35-310 Rzeszów, Poland;
| | - Małgorzata Walczak
- Faculty of Chemistry, Rzeszów University of Technology, 6 Powstańcow Warszawy Ave., 35-959 Rzeszów, Poland; (Ł.U.); (M.W.); (Ż.S.); (M.T.)
| | - Żaneta Szymaszek
- Faculty of Chemistry, Rzeszów University of Technology, 6 Powstańcow Warszawy Ave., 35-959 Rzeszów, Poland; (Ł.U.); (M.W.); (Ż.S.); (M.T.)
| | - Magdalena Twardowska
- Faculty of Chemistry, Rzeszów University of Technology, 6 Powstańcow Warszawy Ave., 35-959 Rzeszów, Poland; (Ł.U.); (M.W.); (Ż.S.); (M.T.)
| | - Stanisław Wołowiec
- Medical College, Rzeszów University, 1a Warzywna Street, 35-310 Rzeszów, Poland;
| |
Collapse
|
4
|
Abstract
After decades of research, our knowledge of the complexity of cancer mechanisms, elegantly summarized as 'hallmarks of cancer', is expanding, as are the therapeutic opportunities that this knowledge brings. However, cancer still needs intense research to diminish its tremendous impact. In this context, the use of simple model organisms such as Caenorhabditis elegans, in which the genetics of the apoptotic pathway was discovered, can facilitate the investigation of several cancer hallmarks. Amenable for genetic and drug screens, convenient for fast and efficient genome editing, and aligned with the 3Rs ('Replacement, Reduction and Refinement') principles for ethical animal research, C. elegans plays a significant role in unravelling the intricate network of cancer mechanisms and presents a promising option in clinical diagnosis and drug discovery.
Collapse
Affiliation(s)
- Julián Cerón
- Modeling Human Diseases in C. elegans Group – Genes, Disease and Therapy Program, Bellvitge Biomedical Research Institute – IDIBELL, 08908 L'Hospitalet de Llobregat, Barcelona, Spain
| |
Collapse
|
5
|
Wu KC, Chu PC, Cheng YJ, Li CI, Tian J, Wu HY, Wu SH, Lai YC, Kao HH, Hsu AL, Lin HW, Lin CH. Development of a traditional Chinese medicine-based agent for the treatment of cancer cachexia. J Cachexia Sarcopenia Muscle 2022; 13:2073-2087. [PMID: 35718751 PMCID: PMC9397559 DOI: 10.1002/jcsm.13028] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/22/2021] [Revised: 04/19/2022] [Accepted: 05/13/2022] [Indexed: 11/06/2022] Open
Abstract
BACKGROUND Despite recent advances in understanding the pathophysiology of cancer cachexia, prevention/treatment of this debilitating disease remains an unmet medical need. METHODS We developed an integrated, multi-tiered strategy involving both in vitro and in vivo muscle atrophy platforms to identify traditional Chinese medicine (TCM)-based anti-cachectic agents. In the initial screening, we used inflammatory cytokine-induced atrophy of C2C12 myotubes as a phenotypic screening platform to assess the protective effects of TCMs. The selected TCMs were then evaluated for their abilities to protect Caenorhabditis elegans from age-related reduction of mobility and contractility, followed by the C-26 colon adenocarcinoma mouse model of cachexia to confirm the anti-muscle atrophy effects (body/skeletal muscle weights, fibre size distribution, grip strengths, and serum IL-6). Transcriptome analysis, quantitative real-time polymerase chain reaction, and immunoblotting were performed to gain understanding of the potential mechanism(s) by which effective TCM protected against C26 tumour-induced muscle atrophy. RESULTS Of 29 widely used TCMs, Dioscorea radix (DR) and Mu Dan Pi (MDP) showed a complete protection (all P values, 0.0002) vis-à-vis C26 conditioned medium control in the myotube atrophy platform. MDP exhibited a unique ability to ameliorate age-associated decreases in worm mobility, accompanied by improved total body contractions, relative to control (P < 0.0001 and <0.01, respectively), which, however, was not noted with DR. This differential in vivo protective effect between MDP and DR was also confirmed in the C-26 mouse model. MDP at 1000 mg/kg (MDP-H) was effective in protecting body weight loss (P < 0.05) in C-26 tumour-bearing mice without changing food or water intake, accompanied by the restoration of the fibre size distribution of hindleg skeletal muscles (P < 0.0001) and the forelimb grip strength (P < 0.05). MDP-treated C-26-tumour-bearing mice were alert, showed normal posture and better body conditions, and exhibited lower serum IL-6 levels (P = 0.06) relative to vehicle control. This decreased serum IL-6 was associated with the in vitro suppressive effect of MDP (25 and 50 μg/mL) on IL-6 secretion into culture medium by C26 cells. RNA-seq analysis, followed by quantitative real-time polymerase chain reaction and/or immunoblotting, shows that MDP's anti-cachectic effect was attributable to its ability to reverse the C-26 tumour-induced re-programming of muscle homoeostasis-associated gene expression, including that of two cachexia drivers (MuRF1 and Atrogin-1), in skeletal muscles. CONCLUSIONS All these findings suggest the translational potential of MDP to foster new strategies for the prevention and/or treatment of cachexia. The protective effect of MDP on other types of muscle atrophy such as sarcopenia might warrant investigations.
Collapse
Affiliation(s)
- Kun-Chang Wu
- School of Pharmacy, College of Pharmacy, China Medical University, Taichung, Taiwan.,Research Center for Healthy Aging, China Medical University, Taichung, Taiwan
| | - Po-Chen Chu
- Department of Cosmeceutics and Graduate Institute of Cosmeceutics, China Medical University, Taichung, Taiwan
| | - Yu-Jung Cheng
- Department of Physical Therapy and Graduate Institute of Rehabilitation Science, China Medical University, Taichung, Taiwan.,Department of Rehabilitation, China Medical University Hospital, Taichung, Taiwan
| | - Chia-Ing Li
- School of Medicine, College of Medicine, China Medical University, Taichung, Taiwan.,Department of Medical Research, China Medical University Hospital, Taichung, Taiwan
| | - Jingkui Tian
- Institute of Cancer and Basic Medicine, Chinese Academy of Sciences, Cancer Hospital of the University of Chinese Academy of Sciences, Zhejiang Cancer Hospital, Hangzhou, China.,College of Biomedical Engineering & Instrument Science, Zhejiang University, Hangzhou, China
| | - Hsing-Yu Wu
- Institute of Biological Chemistry, Academia Sinica, Taipei, Taiwan
| | - Szu-Hsien Wu
- School of Pharmacy, College of Pharmacy, China Medical University, Taichung, Taiwan.,Institute of New Drug Development, China Medical University, Taichung, Taiwan
| | - Yi-Chun Lai
- Research Center for Healthy Aging, China Medical University, Taichung, Taiwan
| | - Hsiang-Han Kao
- Department of Family Medicine, China Medical University Hospital, Taichung, Taiwan
| | - Ao-Lin Hsu
- Research Center for Healthy Aging, China Medical University, Taichung, Taiwan.,PhD Program for Aging, China Medical University, Taichung, Taiwan.,Department of Internal Medicine, Division of Geriatrics & Palliative Medicine, University of Michigan Medical School, Ann Arbor, MI, USA
| | - Hsiang-Wen Lin
- School of Pharmacy, College of Pharmacy, China Medical University, Taichung, Taiwan.,Department of Pharmacy, China Medical University Hospital, Taichung, Taiwan.,Department of Pharmacy System, Outcomes and Policy, College of Pharmacy, University of Illinois at Chicago, Chicago, IL, USA
| | - Chih-Hsueh Lin
- School of Medicine, College of Medicine, China Medical University, Taichung, Taiwan.,Department of Geriatric Medicine, China Medical University Hospital, Taichung, Taiwan
| |
Collapse
|
6
|
Knox J, Joly N, Linossi EM, Carmona-Negrón JA, Jura N, Pintard L, Zuercher W, Roy PJ. A survey of the kinome pharmacopeia reveals multiple scaffolds and targets for the development of novel anthelmintics. Sci Rep 2021; 11:9161. [PMID: 33911106 PMCID: PMC8080662 DOI: 10.1038/s41598-021-88150-6] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2020] [Accepted: 04/08/2021] [Indexed: 11/10/2022] Open
Abstract
Over one billion people are currently infected with a parasitic nematode. Symptoms can include anemia, malnutrition, developmental delay, and in severe cases, death. Resistance is emerging to the anthelmintics currently used to treat nematode infection, prompting the need to develop new anthelmintics. Towards this end, we identified a set of kinases that may be targeted in a nematode-selective manner. We first screened 2040 inhibitors of vertebrate kinases for those that impair the model nematode Caenorhabditis elegans. By determining whether the terminal phenotype induced by each kinase inhibitor matched that of the predicted target mutant in C. elegans, we identified 17 druggable nematode kinase targets. Of these, we found that nematode EGFR, MEK1, and PLK1 kinases have diverged from vertebrates within their drug-binding pocket. For each of these targets, we identified small molecule scaffolds that may be further modified to develop nematode-selective inhibitors. Nematode EGFR, MEK1, and PLK1 therefore represent key targets for the development of new anthelmintic medicines.
Collapse
Affiliation(s)
- Jessica Knox
- Department of Molecular Genetics, University of Toronto, Toronto, ON, M5S 1A8, Canada.,The Donnelly Centre for Cellular and Biomolecular Research, University of Toronto, Toronto, ON, M5S 3E1, Canada
| | - Nicolas Joly
- Programme Équipe Labellisée Ligue Contre Le Cancer, Institut Jacques Monod, UMR7592, Université de Paris, CNRS, Paris, France
| | - Edmond M Linossi
- Cardiovascular Research Institute, University of California San Francisco, San Francisco, CA, 94158, USA
| | - José A Carmona-Negrón
- Cardiovascular Research Institute, University of California San Francisco, San Francisco, CA, 94158, USA
| | - Natalia Jura
- Cardiovascular Research Institute, University of California San Francisco, San Francisco, CA, 94158, USA.,Department of Cellular and Molecular Pharmacology, University of California San Francisco, San Francisco, CA, 94158, USA
| | - Lionel Pintard
- Programme Équipe Labellisée Ligue Contre Le Cancer, Institut Jacques Monod, UMR7592, Université de Paris, CNRS, Paris, France
| | - William Zuercher
- School of Pharmacy, UNC Eshelman, University of North Carolina at Chapel Hill, Chapel Hill, NC, 27599, USA
| | - Peter J Roy
- Department of Molecular Genetics, University of Toronto, Toronto, ON, M5S 1A8, Canada. .,The Donnelly Centre for Cellular and Biomolecular Research, University of Toronto, Toronto, ON, M5S 3E1, Canada. .,Department of Pharmacology and Toxicology, University of Toronto, Toronto, ON, M5S 1A8, Canada.
| |
Collapse
|
7
|
Giunti S, Andersen N, Rayes D, De Rosa MJ. Drug discovery: Insights from the invertebrate Caenorhabditis elegans. Pharmacol Res Perspect 2021; 9:e00721. [PMID: 33641258 PMCID: PMC7916527 DOI: 10.1002/prp2.721] [Citation(s) in RCA: 50] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2020] [Accepted: 01/06/2021] [Indexed: 12/18/2022] Open
Abstract
Therapeutic drug development is a long, expensive, and complex process that usually takes 12-15 years. In the early phases of drug discovery, in particular, there is a growing need for animal models that ensure the reduction in both cost and time. Caenorhabditis elegans has been traditionally used to address fundamental aspects of key biological processes, such as apoptosis, aging, and gene expression regulation. During the last decade, with the advent of large-scale platforms for screenings, this invertebrate has also emerged as an essential tool in the pharmaceutical research industry to identify novel drugs and drug targets. In this review, we discuss the reasons why C. elegans has been positioned as an outstanding cost-effective option for drug discovery, highlighting both the advantages and drawbacks of this model. Particular attention is paid to the suitability of this nematode in large-scale genetic and pharmacological screenings. High-throughput screenings in C. elegans have indeed contributed to the breakthrough of a wide variety of candidate compounds involved in extensive fields including neurodegeneration, pathogen infections and metabolic disorders. The versatility of this nematode, which enables its instrumentation as a model of human diseases, is another attribute also herein underscored. As illustrative examples, we discuss the utility of C. elegans models of both human neurodegenerative diseases and parasitic nematodes in the drug discovery industry. Summing up, this review aims to demonstrate the impact of C. elegans models on the drug discovery pipeline.
Collapse
Affiliation(s)
- Sebastián Giunti
- Instituto de Investigaciones Bioquímicas de Bahía Blanca (INIBIBB) CCT UNS‐CONICETBahía BlancaArgentina
- Dpto de Biología, Bioquímica y FarmaciaUniversidad Nacional del SurBahía BlancaArgentina
| | - Natalia Andersen
- Instituto de Investigaciones Bioquímicas de Bahía Blanca (INIBIBB) CCT UNS‐CONICETBahía BlancaArgentina
- Dpto de Biología, Bioquímica y FarmaciaUniversidad Nacional del SurBahía BlancaArgentina
| | - Diego Rayes
- Instituto de Investigaciones Bioquímicas de Bahía Blanca (INIBIBB) CCT UNS‐CONICETBahía BlancaArgentina
- Dpto de Biología, Bioquímica y FarmaciaUniversidad Nacional del SurBahía BlancaArgentina
| | - María José De Rosa
- Instituto de Investigaciones Bioquímicas de Bahía Blanca (INIBIBB) CCT UNS‐CONICETBahía BlancaArgentina
- Dpto de Biología, Bioquímica y FarmaciaUniversidad Nacional del SurBahía BlancaArgentina
| |
Collapse
|
8
|
Lee JH, Park SY, Hwang W, Sung JY, Cho ML, Shim J, Kim YN, Yoon K. Isoharringtonine Induces Apoptosis of Non-Small Cell Lung Cancer Cells in Tumorspheroids via the Intrinsic Pathway. Biomolecules 2020; 10:E1521. [PMID: 33172112 PMCID: PMC7694770 DOI: 10.3390/biom10111521] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2020] [Revised: 10/30/2020] [Accepted: 11/04/2020] [Indexed: 12/24/2022] Open
Abstract
Lung cancer is the major cause of cancer-associated death worldwide, and development of new therapeutic drugs is needed to improve treatment outcomes. Three-dimensional (3D) tumorspheroids offer many advantages over conventional two-dimensional cell cultures due to the similarities to in vivo tumors. We found that isoharringtonine, a natural product purified from Cephalotaxus koreana Nakai, significantly inhibited the growth of tumorspheroids with NCI-H460 cells in a dose-dependent manner and induced apoptotic cell death in our 3D cell culture system. On the other hand, A549 tumorspheroids displayed low sensitivity to isoharringtonine-induced apoptosis. Nuclear receptor subfamily 4 group A member 1 (NR4A1) is an orphan nuclear receptor known to regulate proliferation and apoptosis of cancer cells. We observed that knockdown of NR4A1 dramatically increased isoharringtonine-induced cancer cell death in A549 tumorspheroids by activating the intrinsic apoptosis pathway. Furthermore, treatment with combined isoharringtonine and iNR4A1 significantly inhibited multivulva formation in a Caenorhabditis elegans model and tumor development in a xenograft mouse model. Taken together, our data suggest that isoharringtonine is a potential natural product for treatment of non-small cell lung cancers, and inhibition of NR4A1 sensitizes cancer cells to anti-cancer treatment.
Collapse
Affiliation(s)
- Ji Hae Lee
- Division of Translational Research, National Cancer Center, Goyang 10408, Korea; (J.H.L.); (S.-Y.P.); (W.H.); (J.Y.S.); (J.S.); (Y.-N.K.)
- College of Pharmacy, Graduate School of Pharmaceutical Sciences, Ewha Womans University, Seoul 03760, Korea
| | - So-Young Park
- Division of Translational Research, National Cancer Center, Goyang 10408, Korea; (J.H.L.); (S.-Y.P.); (W.H.); (J.Y.S.); (J.S.); (Y.-N.K.)
| | - Wonbin Hwang
- Division of Translational Research, National Cancer Center, Goyang 10408, Korea; (J.H.L.); (S.-Y.P.); (W.H.); (J.Y.S.); (J.S.); (Y.-N.K.)
| | - Jee Young Sung
- Division of Translational Research, National Cancer Center, Goyang 10408, Korea; (J.H.L.); (S.-Y.P.); (W.H.); (J.Y.S.); (J.S.); (Y.-N.K.)
| | - Myoung-Lae Cho
- National Institute for Korean Medicine Development, Gyeongsan 38540, Korea;
| | - Jaegal Shim
- Division of Translational Research, National Cancer Center, Goyang 10408, Korea; (J.H.L.); (S.-Y.P.); (W.H.); (J.Y.S.); (J.S.); (Y.-N.K.)
| | - Yong-Nyun Kim
- Division of Translational Research, National Cancer Center, Goyang 10408, Korea; (J.H.L.); (S.-Y.P.); (W.H.); (J.Y.S.); (J.S.); (Y.-N.K.)
| | - Kyungsil Yoon
- Division of Translational Research, National Cancer Center, Goyang 10408, Korea; (J.H.L.); (S.-Y.P.); (W.H.); (J.Y.S.); (J.S.); (Y.-N.K.)
| |
Collapse
|
9
|
Choi J, Sung JY, Lee S, Yoo J, Rongo C, Kim YN, Shim J. Rab8 and Rabin8-Mediated Tumor Formation by Hyperactivated EGFR Signaling via FGFR Signaling. Int J Mol Sci 2020; 21:ijms21207770. [PMID: 33092268 PMCID: PMC7589727 DOI: 10.3390/ijms21207770] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2020] [Revised: 10/13/2020] [Accepted: 10/19/2020] [Indexed: 02/08/2023] Open
Abstract
The epidermal growth factor receptor (EGFR) signaling is important for normal development, such as vulval development in Caenorhabditis elegans, and hyperactivation of the EGFR is often associated with cancer development. Our previous report demonstrated the multivulva (Muv) phenotype, a tumor model in C. elegans (jgIs25 strain) by engineering LET-23/EGFR with a TKI-resistant human EGFR T790-L858 mutant. Because Rab proteins regulate vesicle transport, which is important for receptor signaling, we screened the RNAi in the jgIs25 strain to find the Rabs critical for Muv formation. Herein, we show that rab-8 RNAi and the rab-8 (-/-) mutation effectively reduce Muv formation. We demonstrate that RABN-8, an ortholog of Rabin8, known as a GEF for Rab8, is also required for Muv formation by promoting the secretion of EGL-17/FGF from vulval precursor cells. In addition, FGFR inhibitors decreased Muv formation mediated by mutant EGFR. Our data suggest that Rab8 and Rabin8 mediate Muv formation through FGF secretion in the EGFR-TKI-resistant nematode model. Furthermore, FGFR-TKIs more effectively inhibit the growth of lung cancer cell lines in H1975 (EGFR T790M-L858R; EGFR-TKI-resistant) than H522 (wild-type EGFR) and H1650 (EGFR exon 19 deletion; EGFR-TKI-sensitive) cells, suggesting that FGFR-TKIs could be used to control cancers with EGFR-TKI-resistant mutations.
Collapse
Affiliation(s)
- Junghwa Choi
- Research Institute of National Cancer Center, 323 Ilsan-ro, Goyang, Gyeonggi-do 10408, Korea; (J.C.); (J.Y.S.); (S.L.); (J.Y.)
| | - Jee Young Sung
- Research Institute of National Cancer Center, 323 Ilsan-ro, Goyang, Gyeonggi-do 10408, Korea; (J.C.); (J.Y.S.); (S.L.); (J.Y.)
| | - Saerom Lee
- Research Institute of National Cancer Center, 323 Ilsan-ro, Goyang, Gyeonggi-do 10408, Korea; (J.C.); (J.Y.S.); (S.L.); (J.Y.)
| | - Jungyoen Yoo
- Research Institute of National Cancer Center, 323 Ilsan-ro, Goyang, Gyeonggi-do 10408, Korea; (J.C.); (J.Y.S.); (S.L.); (J.Y.)
| | - Christopher Rongo
- Waksman Institute of Microbiology, Rutgers, The State University of New Jersey, 190 Frelinghuysen Road, Piscataway, NJ 08854, USA;
| | - Yong-Nyun Kim
- Research Institute of National Cancer Center, 323 Ilsan-ro, Goyang, Gyeonggi-do 10408, Korea; (J.C.); (J.Y.S.); (S.L.); (J.Y.)
- Correspondence: (Y.-N.K.); (J.S.); Tel.: +82-31-920-2415 (Y.-N.K.); +82-31-920-2262 (J.S.)
| | - Jaegal Shim
- Research Institute of National Cancer Center, 323 Ilsan-ro, Goyang, Gyeonggi-do 10408, Korea; (J.C.); (J.Y.S.); (S.L.); (J.Y.)
- Correspondence: (Y.-N.K.); (J.S.); Tel.: +82-31-920-2415 (Y.-N.K.); +82-31-920-2262 (J.S.)
| |
Collapse
|
10
|
Mao YQ, Han SF, Zhang SL, Zhang ZY, Kong CY, Chen HL, Li ZM, Cai PR, Han B, Wang LS. An approach using Caenorhabditis elegans screening novel targets to suppress tumour cell proliferation. Cell Prolif 2020; 53:e12832. [PMID: 32452127 PMCID: PMC7309951 DOI: 10.1111/cpr.12832] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2020] [Revised: 04/07/2020] [Accepted: 04/29/2020] [Indexed: 12/12/2022] Open
Abstract
OBJECTIVES Tumour cell proliferation requires high metabolism to meet the bioenergetics and biosynthetic needs. Dauer in Caenorhabditis elegans is characterized by lower metabolism, and we established an approach with C elegans to find potential tumour therapy targets. MATERIALS AND METHODS RNAi screening was used to find dauer-related genes, and these genes were further analysed in glp-1(-) mutants for tumour-suppressing testing. The identified tumour-related genes were verified in clinical tumour tissues. RESULTS The lifespan of glp-1(-) mutants was found to be extended by classical dauer formation signalling. Then, 61 of 287 kinase-coding genes in Caenorhabditis elegans were identified as dauer-related genes, of which 27 were found to be homologous to human oncogenes. Furthermore, 12 dauer-related genes were randomly selected for tumour-suppressing test, and six genes significantly extended the lifespan of glp-1(-) mutants. Of these six genes, F47D12.9, W02B12.12 and gcy-21 were newly linked to dauer formation. These three new dauer-related genes significantly suppressed tumour cell proliferation and thus extended the lifespan of glp-1(-) mutants in a longevity- or dauer-independent manner. The mRNA expression profiles indicated that these dauer-related genes trigged similar low metabolism pattern in glp-1(-) mutants. Notably, the expression of homolog gene DCAF4L2/F47D12.9, TSSK6/W02B12.12 and NPR1/gcy-21 was found to be higher in glioma compared with adjacent normal tissue. In addition, the high expression of TSSK6/W02B12.12 and NPR1/gcy-21 correlated with a worse survival in glioma patients. CONCLUSIONS Dauer gene screening in combination with tumour-suppressing test in glp-1(-) mutants provided a useful approach to find potential targets for tumour therapy via suppressing tumour cell proliferation and rewiring tumour cell metabolism.
Collapse
Affiliation(s)
- Yu-Qin Mao
- Key Laboratory of Whole-period Monitoring and Precise Intervention of Digestive Cancer (SMHC), Minhang Hospital, Fudan University, Shanghai, China.,Institute of Fudan-Minhang Academic Health System, Minhang Hospital, Fudan University, Shanghai, China
| | - San-Feng Han
- Key Laboratory of Whole-period Monitoring and Precise Intervention of Digestive Cancer (SMHC), Minhang Hospital, Fudan University, Shanghai, China.,Institute of Fudan-Minhang Academic Health System, Minhang Hospital, Fudan University, Shanghai, China
| | - Shi-Long Zhang
- Key Laboratory of Whole-period Monitoring and Precise Intervention of Digestive Cancer (SMHC), Minhang Hospital, Fudan University, Shanghai, China.,Institute of Fudan-Minhang Academic Health System, Minhang Hospital, Fudan University, Shanghai, China
| | - Zheng-Yan Zhang
- Key Laboratory of Whole-period Monitoring and Precise Intervention of Digestive Cancer (SMHC), Minhang Hospital, Fudan University, Shanghai, China.,Institute of Fudan-Minhang Academic Health System, Minhang Hospital, Fudan University, Shanghai, China
| | - Chao-Yue Kong
- Key Laboratory of Whole-period Monitoring and Precise Intervention of Digestive Cancer (SMHC), Minhang Hospital, Fudan University, Shanghai, China.,Institute of Fudan-Minhang Academic Health System, Minhang Hospital, Fudan University, Shanghai, China
| | - Hui-Ling Chen
- Key Laboratory of Whole-period Monitoring and Precise Intervention of Digestive Cancer (SMHC), Minhang Hospital, Fudan University, Shanghai, China.,Institute of Fudan-Minhang Academic Health System, Minhang Hospital, Fudan University, Shanghai, China
| | - Zhan-Ming Li
- Key Laboratory of Whole-period Monitoring and Precise Intervention of Digestive Cancer (SMHC), Minhang Hospital, Fudan University, Shanghai, China.,Institute of Fudan-Minhang Academic Health System, Minhang Hospital, Fudan University, Shanghai, China
| | - Pei-Ran Cai
- Key Laboratory of Whole-period Monitoring and Precise Intervention of Digestive Cancer (SMHC), Minhang Hospital, Fudan University, Shanghai, China.,Institute of Fudan-Minhang Academic Health System, Minhang Hospital, Fudan University, Shanghai, China
| | - Bing Han
- Key Laboratory of Whole-period Monitoring and Precise Intervention of Digestive Cancer (SMHC), Minhang Hospital, Fudan University, Shanghai, China.,Institute of Fudan-Minhang Academic Health System, Minhang Hospital, Fudan University, Shanghai, China
| | - Li-Shun Wang
- Key Laboratory of Whole-period Monitoring and Precise Intervention of Digestive Cancer (SMHC), Minhang Hospital, Fudan University, Shanghai, China.,Institute of Fudan-Minhang Academic Health System, Minhang Hospital, Fudan University, Shanghai, China
| |
Collapse
|
11
|
Yun UJ, Lee JH, Shim J, Yoon K, Goh SH, Yi EH, Ye SK, Lee JS, Lee H, Park J, Lee IH, Kim YN. Anti-cancer effect of doxorubicin is mediated by downregulation of HMG-Co A reductase via inhibition of EGFR/Src pathway. J Transl Med 2019; 99:1157-1172. [PMID: 30700846 DOI: 10.1038/s41374-019-0193-1] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2018] [Revised: 11/30/2018] [Accepted: 12/19/2018] [Indexed: 02/06/2023] Open
Abstract
Doxorubicin is a widely used DNA damage-inducing anti-cancer drug. However, its use is limited by its dose-dependent side effects, such as cardiac toxicity. Cholesterol-lowering statin drugs increase the efficacy of some anti-cancer drugs. Cholesterol is important for cell growth and a critical component of lipid rafts, which are plasma membrane microdomains important for cell signaling. 3-hydroxy-3-methylglutaryl coenzyme A (HMG-CoA) reductase (HMG-CR) is a critical enzyme in cholesterol synthesis. Here, we show that doxorubicin downregulated HMG-CR protein levels and thus reduced levels of cholesterol and lipid rafts. Cholesterol addition attenuated doxorubicin-induced cell death, and cholesterol depletion enhanced it. Reduction of HMG-CR activity by simvastatin, a statin that acts as an HMG-CR inhibitor, or by siRNA-mediated HMG-CR knockdown enhanced doxorubicin cytotoxicity. Doxorubicin-induced HMG-CR downregulation was associated with inactivation of the EGFR-Src pathway. Furthermore, a high-cholesterol-diet attenuated the anti-cancer activity of doxorubicin in a tumor xenograft mouse model. In a multivulva model of Caenorhabditis elegans expressing an active-EGFR mutant, doxorubicin decreased hyperplasia more efficiently in the absence than in the presence of cholesterol. These data indicate that EGFR/Src/HMG-CR is a new pathway mediating doxorubicin-induced cell death and that cholesterol control could be combined with doxorubicin treatment to enhance efficacy and thus reduce side effects.
Collapse
Affiliation(s)
- Un-Jung Yun
- Comparative Biomedicine Research Branch, Division of Translational Science, National Cancer Center, Goyang, Korea
| | - Ji-Hye Lee
- Comparative Biomedicine Research Branch, Division of Translational Science, National Cancer Center, Goyang, Korea
| | - Jaegal Shim
- Comparative Biomedicine Research Branch, Division of Translational Science, National Cancer Center, Goyang, Korea
| | - Kyungsil Yoon
- Comparative Biomedicine Research Branch, Division of Translational Science, National Cancer Center, Goyang, Korea
| | - Sung-Ho Goh
- Therapeutic Target Discovery Branch, Division of Precision Medicine, National Cancer Center, Goyang, Korea
| | - Eun Hee Yi
- Department of Pharmacology, College of Medicine, Seoul National University, Seoul, Korea
| | - Sang-Kyu Ye
- Department of Pharmacology, College of Medicine, Seoul National University, Seoul, Korea
| | - Jae-Seon Lee
- Department of Molecular medicine, College of Medicine, Inha University, Incheon, Korea
| | - Hyunji Lee
- Department of Pharmacology and Medical Science, Metabolic Syndrome and Cell Signaling Laboratory, Institute for Cancer Research, College of Medicine, Chungnam National University, Daejeon, Korea
| | - Jongsun Park
- Department of Pharmacology and Medical Science, Metabolic Syndrome and Cell Signaling Laboratory, Institute for Cancer Research, College of Medicine, Chungnam National University, Daejeon, Korea
| | - In Hye Lee
- Department of Life Science, Ewha Womans University, Seoul, Korea.
| | - Yong-Nyun Kim
- Comparative Biomedicine Research Branch, Division of Translational Science, National Cancer Center, Goyang, Korea.
| |
Collapse
|
12
|
Ji J, Yuan J, Guo X, Ji R, Quan Q, Ding M, Li X, Liu Y. Harmine suppresses hyper-activated Ras-MAPK pathway by selectively targeting oncogenic mutated Ras/Raf in Caenorhabditis elegans. Cancer Cell Int 2019; 19:159. [PMID: 31198408 PMCID: PMC6558680 DOI: 10.1186/s12935-019-0880-4] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2019] [Accepted: 06/03/2019] [Indexed: 11/10/2022] Open
Abstract
Background Mutationally activated Ras proteins are closely linked to a wide variety of human cancers. Hence, there has been an intensive search for anti-Ras therapies for cancer treatment. The sole Ras gene, which encodes LET-60, in Caenorhabditis elegans regulates vulval development. While the loss of let-60 function leads to failure of vulva formation, the let-60(n1046gf) allele, which contains a missense mutation mimicking a Ras codon 13 mutation found in human cancers, results in extra vulval tissue, a phenotype named Muv (multiple vulvas). Methods By taking advantage of the easy-to-score Muv phenotype of let-60(n1046gf), we used a step-by-step screening approach (from crude extract to active fraction to active natural compound) to search for inhibitors of oncogenic Ras. Mutants of other key components in the Ras-mitogen-activated protein kinase (MAPK) pathway were used to identify other candidate targets. Results The natural compound harmine, isolated from the plant Peganum harmala, was found to suppress the Muv phenotype of let-60(n1046gf). In addition, harmine targets the hyper-activation of the Ras/MAPK pathway specifically caused by overexpression or mutated forms of LET-60/Ras and its immediate downstream molecule LIN-45/Raf. Finally, harmine can be absorbed into the worm body and probably functions in its native form, rather than requiring metabolic activation. Conclusion In sum, we have revealed for the first time the anti-Ras activity of harmine in a C. elegans model system. Our results revealed the potential anti-cancer mechanism of harmine, which may be useful for the treatment of specific human cancers that are associated with oncogenic Ras mutations.
Collapse
Affiliation(s)
- Jiaojiao Ji
- 1Beijing University of Chinese Medicine, Beijing, China
| | - Jiang Yuan
- 1Beijing University of Chinese Medicine, Beijing, China
| | - Xiaoyu Guo
- 1Beijing University of Chinese Medicine, Beijing, China
| | - Ruifang Ji
- 1Beijing University of Chinese Medicine, Beijing, China
| | - Qinghua Quan
- 1Beijing University of Chinese Medicine, Beijing, China
| | - Mei Ding
- 2State Key Laboratory of Molecular Developmental Biology, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing, China
| | - Xia Li
- 2State Key Laboratory of Molecular Developmental Biology, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing, China
| | - Yonggang Liu
- 1Beijing University of Chinese Medicine, Beijing, China
| |
Collapse
|
13
|
Akhoon BA, Rathor L, Pandey R. Withanolide A extends the lifespan in human EGFR-driven cancerous Caenorhabditis elegans. Exp Gerontol 2018; 104:113-117. [DOI: 10.1016/j.exger.2018.02.004] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2017] [Revised: 01/31/2018] [Accepted: 02/04/2018] [Indexed: 12/17/2022]
|
14
|
Ma L, Zhao Y, Chen Y, Cheng B, Peng A, Huang K. Caenorhabditis elegans as a model system for target identification and drug screening against neurodegenerative diseases. Eur J Pharmacol 2017; 819:169-180. [PMID: 29208474 DOI: 10.1016/j.ejphar.2017.11.051] [Citation(s) in RCA: 56] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2017] [Accepted: 11/30/2017] [Indexed: 12/12/2022]
Abstract
Over the past decades, Caenorhabditis elegans (C. elegans) has been widely used as a model system because of its small size, transparent body, short generation time and lifespan (~3 days and 3 weeks, respectively), completely sequenced genome and tractability to genetic manipulation. Protein misfolding and aggregation are key pathological features in neurodegenerative diseases such as Alzheimer's disease, Parkinson's disease, Huntington's disease and Amyotrophic lateral sclerosis. Animal models, including C. elegans, have been extensively used to discover and validate new drugs against neurodegenerative diseases. The well-defined and genetically tractable nervous system of C. elegans offers an effective model to explore basic mechanistic pathways of neurodegenerative diseases. Recent progress in high-throughput drug screening also provides a powerful approach for identifying chemical modulators of biological processes. Here, we summarize the latest progress of using C. elegans as a model system for target identification and drug screening in neurodegenerative diseases.
Collapse
Affiliation(s)
- Liang Ma
- Tongji School of Pharmacy, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Yudan Zhao
- Tongji School of Pharmacy, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Yuchen Chen
- Tongji School of Pharmacy, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Biao Cheng
- Department of Pharmacy, The Central Hospital of Wuhan, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430014, China
| | - Anlin Peng
- Department of Pharmacy, The Third Hospital of Wuhan, Wuhan 430060, China
| | - Kun Huang
- Tongji School of Pharmacy, Huazhong University of Science and Technology, Wuhan 430030, China; Center for Biomedicine Research, Wuhan Institute of Biotechnology, Wuhan 430075, China.
| |
Collapse
|
15
|
Huang G, Tong Y, He Q, Wang J, Chen Z. Aucklandia lappa DC. extract enhances gefitinib efficacy in gefitinib-resistance secondary epidermal growth factor receptor mutations. JOURNAL OF ETHNOPHARMACOLOGY 2017; 206:353-362. [PMID: 28619365 DOI: 10.1016/j.jep.2017.06.011] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/09/2017] [Revised: 06/06/2017] [Accepted: 06/10/2017] [Indexed: 06/07/2023]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Aucklandia lappa DC. is a widely used medicinal plant in China, India and Pakistan for a long time. Previously, a number of different pharmacological experiments in vitro and in vivo have convincingly demonstrated the abilities of it to exhibit anticancer activities. Reynoutria japonica Houtt. has also been widely used as traditional Chinese medicinal plant. Previous studies have demonstrated that it is bioactive to exhibit anticancer activities. AIM OF THE STUDY This study aims to investigate whether the extracts of Aucklandia lappa DC. and Reynoutria japonica Houtt. are capable of treating drug-resistant non-small cell lung cancer (NSCLC), providing support for novel usage beyond traditional uses. MATERIALS AND METHODS Extracts combined with gefitinib have been tested taking the vulval development of transgenic C. elegans (jgIs25) as an effective and simple in vivo model system, evaluating their efficacy against acquired NSCLC. Synchronous larval 1 (L1) larvae were treated with extracts plus gefitinib and cultured to obtain mainly L4 larvae. The multivulva (Muv) phenotype was recorded at the adult stage. RESULTS Our data showed that Aucklandia lappa DC. extract could significantly enhance the efficacy of gefitinib, suppressing the Muv phenotype of jgIs25. Meanwhile, it could also down-regulate the mRNA and protein expression of EGFR in jgIs25. Collectively, our results verified that the capability of Aucklandia lappa DC. to inhibit Muv phenotype may be based on the EGFR signaling pathway inhibition. CONCLUSION We demonstrated that the co-administration of Aucklandia lappa DC. with gefitinib may provide an effective strategy for the therapy of EGFR inhibitor resistant NSCLCs.
Collapse
Affiliation(s)
- Guan Huang
- School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou 510006, China
| | - Yanli Tong
- Department of Pharmacy, Guangdong Second Provincial General Hospital, Guangzhou 510317, China.
| | - Qidi He
- School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou 510006, China
| | - Jie Wang
- School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou 510006, China
| | - Zuanguang Chen
- School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou 510006, China
| |
Collapse
|
16
|
Beyond mouse cancer models: Three-dimensional human-relevant in vitro and non-mammalian in vivo models for photodynamic therapy. MUTATION RESEARCH-REVIEWS IN MUTATION RESEARCH 2017; 773:242-262. [DOI: 10.1016/j.mrrev.2016.09.002] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/29/2016] [Accepted: 09/09/2016] [Indexed: 02/08/2023]
|
17
|
Romancino DP, Anello L, Lavanco A, Buffa V, Di Bernardo M, Bongiovanni A. A sea urchin in vivo model to evaluate Epithelial-Mesenchymal Transition. Dev Growth Differ 2017; 59:141-151. [PMID: 28436008 DOI: 10.1111/dgd.12353] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2016] [Revised: 03/30/2017] [Accepted: 03/31/2017] [Indexed: 12/20/2022]
Abstract
Epithelial-mesenchymal transition (EMT) is an evolutionarily conserved cellular program, which is a prerequisite for the metastatic cascade in carcinoma progression. Here, we evaluate the EMT process using the sea urchin Paracentrotus lividus embryo. In sea urchin embryos, the earliest EMT event is related to the acquisition of a mesenchymal phenotype by the spiculogenetic primary mesenchyme cells (PMCs) and their migration into the blastocoel. We investigated the effect of inhibiting the epidermal growth factor (EGF) signaling pathway on this process, and we observed that mesenchyme cell differentiation was blocked. In order to extend and validate our studies, we investigated the migratory capability and the level of potential epidermal growth factor receptor (EGFr) targets in a breast cancer cell line after EGF modulation. Altogether, our data highlight the sensitivity of the sea urchin embryo to anti-EMT drugs and pinpoint the sea urchin embryo as a valuable in vivo model system for studying EMT and the screening of anti-EMT candidates.
Collapse
Affiliation(s)
- Daniele P Romancino
- Institute of Biomedicine and Molecular Immunology (IBIM), National Research Council (CNR), via Ugo La Malfa, 153 - 90146, Palermo, Italy
| | - Letizia Anello
- Institute of Biomedicine and Molecular Immunology (IBIM), National Research Council (CNR), via Ugo La Malfa, 153 - 90146, Palermo, Italy
| | - Antonella Lavanco
- Institute of Biomedicine and Molecular Immunology (IBIM), National Research Council (CNR), via Ugo La Malfa, 153 - 90146, Palermo, Italy
| | - Valentina Buffa
- Institute of Biomedicine and Molecular Immunology (IBIM), National Research Council (CNR), via Ugo La Malfa, 153 - 90146, Palermo, Italy
| | - Maria Di Bernardo
- Institute of Biomedicine and Molecular Immunology (IBIM), National Research Council (CNR), via Ugo La Malfa, 153 - 90146, Palermo, Italy
| | - Antonella Bongiovanni
- Institute of Biomedicine and Molecular Immunology (IBIM), National Research Council (CNR), via Ugo La Malfa, 153 - 90146, Palermo, Italy
| |
Collapse
|
18
|
Gauthier K, Rocheleau CE. C. elegans Vulva Induction: An In Vivo Model to Study Epidermal Growth Factor Receptor Signaling and Trafficking. Methods Mol Biol 2017; 1652:43-61. [PMID: 28791633 DOI: 10.1007/978-1-4939-7219-7_3] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
Epidermal growth factor receptor (EGFR)-mediated activation of the canonical Ras/MAPK signaling cascade is responsible for cell proliferation and cell growth. This signaling pathway is frequently overactivated in epithelial cancers; therefore, studying regulation of this pathway is crucial not only for our fundamental understanding of cell biology but also for our ability to treat EGFR-related disease. Genetic model organisms such as Caenorhabditis elegans, a hermaphroditic nematode, played a vital role in identifying components of the EGFR/Ras/MAPK pathway and delineating their order of function, and continues to play a role in identifying novel regulators of the pathway. Polarized activation of LET-23, the C. elegans homolog of EGFR, is responsible for induction of the vulval cell fate; perturbations in this signaling pathway produce either a vulvaless or multivulva phenotype. The translucent cuticle of the nematode facilitates in vivo visualization of the receptor, revealing that localization of LET-23 EGFR is tightly regulated and linked to its function. In this chapter, we review the methods used to harness vulva development as a tool for studying EGFR signaling and trafficking in C. elegans.
Collapse
Affiliation(s)
- Kimberley Gauthier
- Department of Anatomy and Cell Biology, McGill University, Montreal, Canada
- Centre for Translational Biology, Research Institute of the McGill University Health Centre, Montreal, Canada
| | - Christian E Rocheleau
- Department of Anatomy and Cell Biology, McGill University, Montreal, Canada.
- Centre for Translational Biology, Research Institute of the McGill University Health Centre, Montreal, Canada.
- Division of Endocrinology and Metabolism, Department of Medicine, McGill University, Montreal, Canada.
| |
Collapse
|
19
|
Kyriakakis E, Markaki M, Tavernarakis N. Caenorhabditis elegans as a model for cancer research. Mol Cell Oncol 2015; 2:e975027. [PMID: 27308424 PMCID: PMC4905018 DOI: 10.4161/23723556.2014.975027] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2014] [Revised: 09/18/2014] [Accepted: 09/18/2014] [Indexed: 04/21/2023]
Abstract
The term cancer describes a group of multifaceted diseases characterized by an intricate pathophysiology. Despite significant advances in the fight against cancer, it remains a key public health concern and burden on societies worldwide. Elucidation of key molecular and cellular mechanisms of oncogenic diseases will facilitate the development of better intervention strategies to counter or prevent tumor development. In vivo and in vitro models have long been used to delineate distinct biological processes involved in cancer such as apoptosis, proliferation, angiogenesis, invasion, metastasis, genome instability, and metabolism. In this review, we introduce Caenorhabditis elegans as an emerging animal model for systematic dissection of the molecular basis of tumorigenesis, focusing on the well-established processes of apoptosis and autophagy. Additionally, we propose that C. elegans can be used to advance our understanding of cancer progression, such as deregulation of energy metabolism, stem cell reprogramming, and host-microflora interactions.
Collapse
Affiliation(s)
- Emmanouil Kyriakakis
- Institute of Molecular Biology and Biotechnology; Foundation for Research and Technology-Hellas
| | - Maria Markaki
- Institute of Molecular Biology and Biotechnology; Foundation for Research and Technology-Hellas
| | - Nektarios Tavernarakis
- Institute of Molecular Biology and Biotechnology; Foundation for Research and Technology-Hellas
- Department of Basic Sciences; Faculty of Medicine; University of Crete Heraklion; Crete, Greece
- Correspondence to: N. Tavernarakis;
| |
Collapse
|
20
|
Kobet RA, Pan X, Zhang B, Pak SC, Asch AS, Lee MH. Caenorhabditis elegans: A Model System for Anti-Cancer Drug Discovery and Therapeutic Target Identification. Biomol Ther (Seoul) 2014; 22:371-83. [PMID: 25414766 PMCID: PMC4201220 DOI: 10.4062/biomolther.2014.084] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2014] [Revised: 08/14/2014] [Accepted: 08/18/2014] [Indexed: 01/27/2023] Open
Abstract
The nematode Caenorhabditis elegans (C. elegans) offers a unique opportunity for biological and basic medical researches due to its genetic tractability and well-defined developmental lineage. It also provides an exceptional model for genetic, molecular, and cellular analysis of human disease-related genes. Recently, C. elegans has been used as an ideal model for the identification and functional analysis of drugs (or small-molecules) in vivo. In this review, we describe conserved oncogenic signaling pathways (Wnt, Notch, and Ras) and their potential roles in the development of cancer stem cells. During C. elegans germline development, these signaling pathways regulate multiple cellular processes such as germline stem cell niche specification, germline stem cell maintenance, and germ cell fate specification. Therefore, the aberrant regulations of these signaling pathways can cause either loss of germline stem cells or overproliferation of a specific cell type, resulting in sterility. This sterility phenotype allows us to identify drugs that can modulate the oncogenic signaling pathways directly or indirectly through a high-throughput screening. Current in vivo or in vitro screening methods are largely focused on the specific core signaling components. However, this phenotype-based screening will identify drugs that possibly target upstream or downstream of core signaling pathways as well as exclude toxic effects. Although phenotype-based drug screening is ideal, the identification of drug targets is a major challenge. We here introduce a new technique, called Drug Affinity Responsive Target Stability (DARTS). This innovative method is able to identify the target of the identified drug. Importantly, signaling pathways and their regulators in C. elegans are highly conserved in most vertebrates, including humans. Therefore, C. elegans will provide a great opportunity to identify therapeutic drugs and their targets, as well as to understand mechanisms underlying the formation of cancer.
Collapse
Affiliation(s)
- Robert A Kobet
- Department of Medicine, Department of Oncology, Division of Hematology/Oncology, Brody School of Medicine, East Carolina University, Greenville, NC 27834
| | - Xiaoping Pan
- Department of Biology, East Carolina University, Greenville, NC 27858
| | - Baohong Zhang
- Department of Biology, East Carolina University, Greenville, NC 27858
| | - Stephen C Pak
- Department of Pediatrics, University of Pittsburgh School of Medicine, Children's Hospital of Pittsburgh of UPMC, 4401 Penn Avenue, Pittsburgh, PA 15224
| | - Adam S Asch
- Department of Medicine, Department of Oncology, Division of Hematology/Oncology, Brody School of Medicine, East Carolina University, Greenville, NC 27834 ; Lineberger Comprehensive Cancer Center, University of North Carolina-Chapel Hill, Chapel Hill, NC 27599 ; Current address: Department of Medicine, Division of Hematology/Oncology, University of Oklahoma Health Science Center, Oklahoma City, OK 73104, USA
| | - Myon-Hee Lee
- Department of Medicine, Department of Oncology, Division of Hematology/Oncology, Brody School of Medicine, East Carolina University, Greenville, NC 27834 ; Lineberger Comprehensive Cancer Center, University of North Carolina-Chapel Hill, Chapel Hill, NC 27599
| |
Collapse
|
21
|
Choi DY, You S, Jung JH, Lee JC, Rho JK, Lee KY, Freeman MR, Kim KP, Kim J. Extracellular vesicles shed from gefitinib-resistant nonsmall cell lung cancer regulate the tumor microenvironment. Proteomics 2014; 14:1845-56. [PMID: 24946052 DOI: 10.1002/pmic.201400008] [Citation(s) in RCA: 44] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2014] [Revised: 05/23/2014] [Accepted: 06/11/2014] [Indexed: 12/13/2022]
Abstract
Epidermal growth factor receptor (EGFR)-tyrosine kinase inhibitors (TKIs), including gefitinib, are the first-line treatment of choice for nonsmall cell lung cancer patients who harbor activating EGFR mutations, however, acquired resistance to EGFR-TKIs is inevitable. The main objective of this study was to identify informative protein signatures of extracellular vesicles (EV) derived from gefitinib-resistant nonsmall cell lung cancer cells using proteomics analysis. Nano-LC-MS/MS analysis identified with high confidence (false discovery rate < 0.05, fold change ≥2) 664 EV proteins enriched in PC9R cells, which are resistant to gefitinib due to EGFR T790M mutation. Computational analyses suggested components of several signal transduction mechanisms including the AKT (also PKB, protein kinase B)/mTOR (mechanistic target of rapamycin) pathway are overrepresented in EV from PC9R cells. Treatment of recipient cells with EV harvested from PC9R cells increased phosphorylation of signaling molecules, and enhanced proliferation, invasion, and drug resistance to gefitinib-induced apoptosis. Dose- and time-dependent pharmaceutical inhibition of AKT/mTOR pathway overcame drug resistance of PC9R cells and those of H1975 exhibiting EGFR T790M mutation. Our findings provide new insight into an oncogenic EV protein signature regulating tumor microenvironment, and will aid in the development of novel diagnostic strategies for prediction and assessment of gefitinib resistance.
Collapse
Affiliation(s)
- Do-Young Choi
- Department of Applied Chemistry, College of Applied Science, Kyung Hee University, Yongin, Republic of Korea
| | | | | | | | | | | | | | | | | |
Collapse
|