1
|
Matsuda A, Masuzawa R, Takahashi K, Takano K, Endo T. MEK inhibitors and DA-Raf, a dominant-negative antagonist of the Ras-ERK pathway, prevent the migration and invasion of KRAS-mutant cancer cells. Cytoskeleton (Hoboken) 2025; 82:32-44. [PMID: 38872577 DOI: 10.1002/cm.21881] [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: 04/05/2024] [Revised: 05/08/2024] [Accepted: 05/16/2024] [Indexed: 06/15/2024]
Abstract
The Ras-induced ERK pathway (Raf-MEK-ERK signaling cascade) regulates a variety of cellular responses including cell proliferation, survival, and migration. Activating mutations in RAS genes, particularly in the KRAS gene, constitutively activate the ERK pathway, resulting in tumorigenesis, cancer cell invasion, and metastasis. DA-Raf1 (DA-Raf) is a splicing isoform of A-Raf and contains the Ras-binding domain but lacks the kinase domain. Consequently, DA-Raf antagonizes the Ras-ERK pathway in a dominant-negative manner and can serve as a tumor suppressor that targets mutant Ras protein-induced tumorigenesis. We show here that MEK inhibitors and DA-Raf interfere with the in vitro collective cell migration and invasion of human KRAS-mutant carcinoma cell lines, the lung adenocarcinoma A549, colorectal carcinoma HCT116, and pancreatic carcinoma MIA PaCa-2 cells. DA-Raf expression was silenced in these cancer cell lines. All these cell lines had high collective migration abilities and invasion properties in Matrigel, compared with nontumor cells. Their migration and invasion abilities were impaired by suppressing the ERK pathway with the MEK inhibitors U0126 and trametinib, an approved anticancer drug. Expression of DA-Raf in MIA PaCa-2 cells reduced the ERK activity and hindered the migration and invasion abilities. Therefore, DA-Raf may function as an invasion suppressor protein in the KRAS-mutant cancer cells by blocking the Ras-ERK pathway when DA-Raf expression is induced in invasive cancer cells.
Collapse
Affiliation(s)
- Aoi Matsuda
- Department of Biology, Graduate School of Science, Chiba University, Chiba, Chiba, Japan
| | - Ryuichi Masuzawa
- Department of Biology, Graduate School of Science, Chiba University, Chiba, Chiba, Japan
| | - Kazuya Takahashi
- Department of Biology, Graduate School of Science, Chiba University, Chiba, Chiba, Japan
| | - Kazunori Takano
- Department of Biology, Graduate School of Science, Chiba University, Chiba, Chiba, Japan
| | - Takeshi Endo
- Department of Biology, Graduate School of Science, Chiba University, Chiba, Chiba, Japan
| |
Collapse
|
2
|
Chen Y, Jiang L, Li M, Shen Y, Liu S, Yang D. Huanglian Jiedu decoction alleviates neurobehavioral damage in mice with chronic alcohol exposure through the RAS-RAF-MEK-ERK pathway. Heliyon 2024; 10:e29556. [PMID: 38644875 PMCID: PMC11033144 DOI: 10.1016/j.heliyon.2024.e29556] [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: 12/03/2023] [Revised: 04/09/2024] [Accepted: 04/10/2024] [Indexed: 04/23/2024] Open
Abstract
Objective Long-term alcohol consumption can cause organic damage to the brain, resulting in mental and nervous system abnormalities and intellectual impairment. Huanglian Jiedu decoction (HLJDD) is the classic representative of clearing heat and detoxifying. This study aimed to explore the effects and possible mechanisms of HLJDD on brain injury in chronic alcohol-exposed mice. Methods The alcohol-exposed mice were treated with different doses of HLJDD to observe behavioral changes, hippocampal Aβ1-42 deposition, number and ultrastructural changes of neurons in the hippocampus and prefrontal cortex, and expressions of synaptic proteins. On this basis, transcriptome sequencing was used to analyze the differentially expressed genes in different treatment groups, and functional enrichment analysis was performed. Then, WB and RT-PCR were used to verify the expression of the pathway. Results Chronic alcohol exposure reduced body weight in mice, led to motor cognitive impairment, increased Aβ1-42 in the hippocampus, decreased the number of neurons in the hippocampus and prefrontal cortex, and the expression of PSD95 and SYN in the hippocampus. HLJDD significantly improved the cognitive dysfunction of mice and alleviated the damage of the hippocampus and prefrontal cortex. Transcriptome sequencing results showed that the regulatory effects of HLJDD on chronic alcohol-exposed mice may be related to the RAS pathway. Further experiments confirmed that chronic alcohol exposure caused a significant increase in protein and gene expressions of the RAS-RAF-MEK-ERK pathway in mouse, and this activation was reversed by HLJDD. Conclusion HLJDD may ameliorate brain damage caused by chronic alcohol exposure by regulating the RAS-RAF-MEK-ERK pathway.
Collapse
Affiliation(s)
- Yun Chen
- Department of Neurology, Chengdu University of Traditional Chinese Medicine, Sichuan, PR China
- Department of Neurology, The First People's Hospital of Bijie City, Guizhou, PR China
| | - Lianyan Jiang
- Department of Neurology, Chengdu University of Traditional Chinese Medicine, Sichuan, PR China
| | - Mao Li
- Department of Neurology, The First Affiliated Hospital of Henan University of Traditional Chinese Medicine, Zhengzhou, PR China
| | - Yuling Shen
- Department of Neurology, Chengdu University of Traditional Chinese Medicine, Sichuan, PR China
| | - Shanyu Liu
- Department of Neurology, Chengdu University of Traditional Chinese Medicine, Sichuan, PR China
| | - Dongdong Yang
- Department of Neurology, The Affiliated Hospital of Chengdu University of Traditional Chinese Medicine, Sichuan, PR China
| |
Collapse
|
3
|
Rohrer L, Spohr C, Beha C, Griffin R, Braun S, Halbach S, Brummer T. Analysis of RAS and drug induced homo- and heterodimerization of RAF and KSR1 proteins in living cells using split Nanoluc luciferase. Cell Commun Signal 2023; 21:136. [PMID: 37316874 DOI: 10.1186/s12964-023-01146-9] [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: 11/11/2022] [Accepted: 04/27/2023] [Indexed: 06/16/2023] Open
Abstract
The dimerization of RAF kinases represents a key event in their activation cycle and in RAS/ERK pathway activation. Genetic, biochemical and structural approaches provided key insights into this process defining RAF signaling output and the clinical efficacy of RAF inhibitors (RAFi). However, methods reporting the dynamics of RAF dimerization in living cells and in real time are still in their infancy. Recently, split luciferase systems have been developed for the detection of protein-protein-interactions (PPIs), incl. proof-of-concept studies demonstrating the heterodimerization of the BRAF and RAF1 isoforms. Due to their small size, the Nanoluc luciferase moieties LgBiT and SmBiT, which reconstitute a light emitting holoenzyme upon fusion partner promoted interaction, appear as well-suited to study RAF dimerization. Here, we provide an extensive analysis of the suitability of the Nanoluc system to study the homo- and heterodimerization of BRAF, RAF1 and the related KSR1 pseudokinase. We show that KRASG12V promotes the homo- and heterodimerization of BRAF, while considerable KSR1 homo- and KSR1/BRAF heterodimerization already occurs in the absence of this active GTPase and requires a salt bridge between the CC-SAM domain of KSR1 and the BRAF-specific region. We demonstrate that loss-of-function mutations impairing key steps of the RAF activation cycle can be used as calibrators to gauge the dynamics of heterodimerization. This approach identified the RAS-binding domains and the C-terminal 14-3-3 binding motifs as particularly critical for the reconstitution of RAF mediated LgBiT/SmBiT reconstitution, while the dimer interface was less important for dimerization but essential for downstream signaling. We show for the first time that BRAFV600E, the most common BRAF oncoprotein whose dimerization status is controversially portrayed in the literature, forms homodimers in living cells more efficiently than its wildtype counterpart. Of note, Nanoluc activity reconstituted by BRAFV600E homodimers is highly sensitive to the paradox-breaking RAFi PLX8394, indicating a dynamic and specific PPI. We report the effects of eleven ERK pathway inhibitors on RAF dimerization, incl. third-generation compounds that are less-defined in terms of their dimer promoting abilities. We identify Naporafenib as a potent and long-lasting dimerizer and show that the split Nanoluc approach discriminates between type I, I1/2 and II RAFi. Video Abstract.
Collapse
Affiliation(s)
- Lino Rohrer
- Institute of Molecular Medicine and Cell Research (IMMZ), Zentrum für Biochemie und Molekulare Zellforschung (ZBMZ), Faculty of Medicine, University of Freiburg, Stefan-Meier-Str. 17, Freiburg, 79104, Germany
| | - Corinna Spohr
- Institute of Molecular Medicine and Cell Research (IMMZ), Zentrum für Biochemie und Molekulare Zellforschung (ZBMZ), Faculty of Medicine, University of Freiburg, Stefan-Meier-Str. 17, Freiburg, 79104, Germany
| | - Carina Beha
- Institute of Molecular Medicine and Cell Research (IMMZ), Zentrum für Biochemie und Molekulare Zellforschung (ZBMZ), Faculty of Medicine, University of Freiburg, Stefan-Meier-Str. 17, Freiburg, 79104, Germany
| | - Ricarda Griffin
- Institute of Molecular Medicine and Cell Research (IMMZ), Zentrum für Biochemie und Molekulare Zellforschung (ZBMZ), Faculty of Medicine, University of Freiburg, Stefan-Meier-Str. 17, Freiburg, 79104, Germany
| | - Sandra Braun
- Institute of Molecular Medicine and Cell Research (IMMZ), Zentrum für Biochemie und Molekulare Zellforschung (ZBMZ), Faculty of Medicine, University of Freiburg, Stefan-Meier-Str. 17, Freiburg, 79104, Germany
| | - Sebastian Halbach
- Institute of Molecular Medicine and Cell Research (IMMZ), Zentrum für Biochemie und Molekulare Zellforschung (ZBMZ), Faculty of Medicine, University of Freiburg, Stefan-Meier-Str. 17, Freiburg, 79104, Germany
- German Cancer Consortium (DKTK), Partner Site Freiburg and German Cancer Research Center (DKFZ), Heidelberg, 69120, Germany
| | - Tilman Brummer
- Institute of Molecular Medicine and Cell Research (IMMZ), Zentrum für Biochemie und Molekulare Zellforschung (ZBMZ), Faculty of Medicine, University of Freiburg, Stefan-Meier-Str. 17, Freiburg, 79104, Germany.
- German Cancer Consortium (DKTK), Partner Site Freiburg and German Cancer Research Center (DKFZ), Heidelberg, 69120, Germany.
- Comprehensive Cancer Center Freiburg (CCCF), Medical Center, University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, 79106, Germany.
- Center for Biological Signalling Studies BIOSS, University of Freiburg, Freiburg, 79104, Germany.
| |
Collapse
|
4
|
Lackner A, Müller M, Gamperl M, Stoeva D, Langmann O, Papuchova H, Roitinger E, Dürnberger G, Imre R, Mechtler K, Latos PA. The Fgf/Erf/NCoR1/2 repressive axis controls trophoblast cell fate. Nat Commun 2023; 14:2559. [PMID: 37137875 DOI: 10.1038/s41467-023-38101-8] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2022] [Accepted: 04/15/2023] [Indexed: 05/05/2023] Open
Abstract
Placental development relies on coordinated cell fate decisions governed by signalling inputs. However, little is known about how signalling cues are transformed into repressive mechanisms triggering lineage-specific transcriptional signatures. Here, we demonstrate that upon inhibition of the Fgf/Erk pathway in mouse trophoblast stem cells (TSCs), the Ets2 repressor factor (Erf) interacts with the Nuclear Receptor Co-Repressor Complex 1 and 2 (NCoR1/2) and recruits it to key trophoblast genes. Genetic ablation of Erf or Tbl1x (a component of the NCoR1/2 complex) abrogates the Erf/NCoR1/2 interaction. This leads to mis-expression of Erf/NCoR1/2 target genes, resulting in a TSC differentiation defect. Mechanistically, Erf regulates expression of these genes by recruiting the NCoR1/2 complex and decommissioning their H3K27ac-dependent enhancers. Our findings uncover how the Fgf/Erf/NCoR1/2 repressive axis governs cell fate and placental development, providing a paradigm for Fgf-mediated transcriptional control.
Collapse
Affiliation(s)
- Andreas Lackner
- Center for Anatomy and Cell Biology, Medical University of Vienna, A-1090, Vienna, Austria
| | - Michael Müller
- Center for Anatomy and Cell Biology, Medical University of Vienna, A-1090, Vienna, Austria
| | - Magdalena Gamperl
- Center for Anatomy and Cell Biology, Medical University of Vienna, A-1090, Vienna, Austria
| | - Delyana Stoeva
- Center for Anatomy and Cell Biology, Medical University of Vienna, A-1090, Vienna, Austria
| | - Olivia Langmann
- Center for Anatomy and Cell Biology, Medical University of Vienna, A-1090, Vienna, Austria
| | - Henrieta Papuchova
- Center for Anatomy and Cell Biology, Medical University of Vienna, A-1090, Vienna, Austria
| | | | | | - Richard Imre
- Institute of Molecular Pathology, A-1030, Vienna, Austria
| | - Karl Mechtler
- Institute of Molecular Pathology, A-1030, Vienna, Austria
| | - Paulina A Latos
- Center for Anatomy and Cell Biology, Medical University of Vienna, A-1090, Vienna, Austria.
| |
Collapse
|
5
|
Dorard C, Madry C, Buhard O, Toifl S, Didusch S, Ratovomanana T, Letourneur Q, Dolznig H, Garnett MJ, Duval A, Baccarini M. RAF1 contributes to cell proliferation and STAT3 activation in colorectal cancer independently of microsatellite and KRAS status. Oncogene 2023; 42:1649-1660. [PMID: 37020037 PMCID: PMC10181936 DOI: 10.1038/s41388-023-02683-w] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2022] [Revised: 03/09/2023] [Accepted: 03/24/2023] [Indexed: 04/07/2023]
Abstract
More than 30% of all human cancers are driven by RAS mutations and activating KRAS mutations are present in 40% of colorectal cancer (CRC) in the two main CRC subgroups, MSS (Microsatellite Stable) and MSI (Microsatellite Instable). Studies in RAS-driven tumors have shown essential roles of the RAS effectors RAF and specifically of RAF1, which can be dependent or independent of RAF's ability to activate the MEK/ERK module. In this study, we demonstrate that RAF1, but not its kinase activity, plays a crucial role in the proliferation of both MSI and MSS CRC cell line-derived spheroids and patient-derived organoids, and independently of KRAS mutation status. Moreover, we could define a RAF1 transcriptomic signature which includes genes that contribute to STAT3 activation, and could demonstrate that RAF1 ablation decreases STAT3 phosphorylation in all CRC spheroids tested. The genes involved in STAT3 activation as well as STAT3 targets promoting angiogenesis were also downregulated in human primary tumors expressing low levels of RAF1. These results indicate that RAF1 could be an attractive therapeutic target in both MSI and MSS CRC regardless of their KRAS status and support the development of selective RAF1 degraders rather than RAF1 inhibitors for clinical use in combination therapies.
Collapse
Affiliation(s)
- Coralie Dorard
- Department of Microbiology, Immunology and Genetics, Center of Molecular Biology, University of Vienna, Max Perutz Labs, Doktor-Bohr-Gasse 9, 1030, Vienna, Austria.
- Sorbonne Université, INSERM, Unité Mixte de Recherche Scientifique 938 and SIRIC CURAMUS, Centre de Recherche Saint-Antoine (CRSA), Equipe Instabilité des Microsatellites et Cancer, Equipe Labellisée par la Ligue Nationale Contre le Cancer, F-75012, Paris, France.
| | - Claire Madry
- Sorbonne Université, INSERM, Unité Mixte de Recherche Scientifique 938 and SIRIC CURAMUS, Centre de Recherche Saint-Antoine (CRSA), Equipe Instabilité des Microsatellites et Cancer, Equipe Labellisée par la Ligue Nationale Contre le Cancer, F-75012, Paris, France
| | - Olivier Buhard
- Sorbonne Université, INSERM, Unité Mixte de Recherche Scientifique 938 and SIRIC CURAMUS, Centre de Recherche Saint-Antoine (CRSA), Equipe Instabilité des Microsatellites et Cancer, Equipe Labellisée par la Ligue Nationale Contre le Cancer, F-75012, Paris, France
| | - Stefanie Toifl
- Department of Microbiology, Immunology and Genetics, Center of Molecular Biology, University of Vienna, Max Perutz Labs, Doktor-Bohr-Gasse 9, 1030, Vienna, Austria
| | - Sebastian Didusch
- Department of Microbiology, Immunology and Genetics, Center of Molecular Biology, University of Vienna, Max Perutz Labs, Doktor-Bohr-Gasse 9, 1030, Vienna, Austria
| | - Toky Ratovomanana
- Sorbonne Université, INSERM, Unité Mixte de Recherche Scientifique 938 and SIRIC CURAMUS, Centre de Recherche Saint-Antoine (CRSA), Equipe Instabilité des Microsatellites et Cancer, Equipe Labellisée par la Ligue Nationale Contre le Cancer, F-75012, Paris, France
| | - Quentin Letourneur
- Sorbonne Université, INSERM, Unité Mixte de Recherche Scientifique 938 and SIRIC CURAMUS, Centre de Recherche Saint-Antoine (CRSA), Equipe Instabilité des Microsatellites et Cancer, Equipe Labellisée par la Ligue Nationale Contre le Cancer, F-75012, Paris, France
| | - Helmut Dolznig
- Institute of Medical Genetics, Medical University of Vienna, Waehringer Straße 10, A-1090, Vienna, Austria
| | | | - Alex Duval
- Sorbonne Université, INSERM, Unité Mixte de Recherche Scientifique 938 and SIRIC CURAMUS, Centre de Recherche Saint-Antoine (CRSA), Equipe Instabilité des Microsatellites et Cancer, Equipe Labellisée par la Ligue Nationale Contre le Cancer, F-75012, Paris, France
| | - Manuela Baccarini
- Department of Microbiology, Immunology and Genetics, Center of Molecular Biology, University of Vienna, Max Perutz Labs, Doktor-Bohr-Gasse 9, 1030, Vienna, Austria
| |
Collapse
|
6
|
Lozano A, Souche FR, Chavey C, Dardalhon V, Ramirez C, Vegna S, Desandre G, Riviere A, Zine El Aabidine A, Fort P, Akkari L, Hibner U, Grégoire D. Ras/MAPK signalling intensity defines subclonal fitness in a mouse model of hepatocellular carcinoma. eLife 2023; 12:76294. [PMID: 36656749 PMCID: PMC9891719 DOI: 10.7554/elife.76294] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2021] [Accepted: 01/18/2023] [Indexed: 01/20/2023] Open
Abstract
Quantitative differences in signal transduction are to date an understudied feature of tumour heterogeneity. The MAPK Erk pathway, which is activated in a large proportion of human tumours, is a prototypic example of distinct cell fates being driven by signal intensity. We have used primary hepatocyte precursors transformed with different dosages of an oncogenic form of Ras to model subclonal variations in MAPK signalling. Orthotopic allografts of Ras-transformed cells in immunocompromised mice gave rise to fast-growing aggressive tumours, both at the primary location and in the peritoneal cavity. Fluorescent labelling of cells expressing different oncogene levels, and consequently varying levels of MAPK Erk activation, highlighted the selection processes operating at the two sites of tumour growth. Indeed, significantly higher Ras expression was observed in primary as compared to secondary, metastatic sites, despite the apparent evolutionary trade-off of increased apoptotic death in the liver that correlated with high Ras dosage. Analysis of the immune tumour microenvironment at the two locations suggests that fast peritoneal tumour growth in the immunocompromised setting is abrogated in immunocompetent animals due to efficient antigen presentation by peritoneal dendritic cells. Furthermore, our data indicate that, in contrast to the metastatic-like outgrowth, strong MAPK signalling is required in the primary liver tumours to resist elimination by NK (natural killer) cells. Overall, this study describes a quantitative aspect of tumour heterogeneity and points to a potential vulnerability of a subtype of hepatocellular carcinoma as a function of MAPK Erk signalling intensity.
Collapse
Affiliation(s)
- Anthony Lozano
- Institut de Génétique Moléculaire de Montpellier, University of MontpellierMontpellierFrance
| | - Francois-Régis Souche
- Institut de Génétique Moléculaire de Montpellier, University of MontpellierMontpellierFrance
- Department of surgery and liver transplantation, Hopital Saint Eloi Hopitaux universitaires de MontpelierMontpellierFrance
| | - Carine Chavey
- Institut de Génétique Moléculaire de Montpellier, University of MontpellierMontpellierFrance
| | - Valérie Dardalhon
- Institut de Génétique Moléculaire de Montpellier, University of MontpellierMontpellierFrance
| | - Christel Ramirez
- Division of Tumor Biology and Immunology, Netherlands Cancer Institute, Oncode InstituteAmsterdamNetherlands
| | - Serena Vegna
- Division of Tumor Biology and Immunology, Netherlands Cancer Institute, Oncode InstituteAmsterdamNetherlands
| | - Guillaume Desandre
- Institut de Génétique Moléculaire de Montpellier, University of MontpellierMontpellierFrance
| | - Anaïs Riviere
- Institut de Génétique Moléculaire de Montpellier, University of MontpellierMontpellierFrance
| | - Amal Zine El Aabidine
- Institut de Génétique Moléculaire de Montpellier, University of MontpellierMontpellierFrance
| | - Philippe Fort
- Centre de Recherche en Biologie Cellulaire de Montpellier (CRBM), University of Montpellier, CNRSMontpellierFrance
| | - Leila Akkari
- Division of Tumor Biology and Immunology, Netherlands Cancer Institute, Oncode InstituteAmsterdamNetherlands
| | - Urszula Hibner
- Institut de Génétique Moléculaire de Montpellier, University of MontpellierMontpellierFrance
| | - Damien Grégoire
- Institut de Génétique Moléculaire de Montpellier, University of MontpellierMontpellierFrance
| |
Collapse
|
7
|
Abstract
Bao G, Pan W, Huang J, Zhou T. K-RasG12V/T35S -ERK1/2 pathway regulates H2BS14ph through Mst1 to facilitate the advancement of breast cancer cells. BioFactors. 2023;49:202. https://doi.org/10.1002/biof.1589 This article, published online on 28 November 2019 in Wiley Online Library, has been retracted by agreement between the International Union of Biochemistry and Molecular Biology, the Editor in Chief (Dr. Angelo Azzi), and Wiley Periodicals LLC. The retraction has been agreed following an investigation based on allegations raised by a third party. Evidence for image manipulation was found in figures 1, 4, 5, and 6. As a result, the conclusions of this article are considered to be invalid.
Collapse
|
8
|
Gunderwala A, Cope N, Wang Z. Mechanism and inhibition of BRAF kinase. Curr Opin Chem Biol 2022; 71:102205. [PMID: 36067564 PMCID: PMC10396080 DOI: 10.1016/j.cbpa.2022.102205] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2022] [Revised: 07/29/2022] [Accepted: 07/30/2022] [Indexed: 01/27/2023]
Abstract
The role of BRAF in tumor initiation has been established, however, the precise mechanism of autoinhibition has only been illustrated recently by several structural studies. These structures uncovered the basis by which the regulatory domains engage in regulating the activity of BRAF kinase domain, which lead to a more complete picture of the regulation cycle of RAF kinases. Small molecule BRAF inhibitors developed specifically to target BRAFV600E have proven effective at inhibiting the most dominant BRAF mutant in melanomas, but are less potent against other BRAF mutants in RAS-driven diseases due to paradoxical activation of the MAPK pathway. A variety of new generation inhibitors that do not show paradoxical activation have been developed. Alternatively, efforts have begun to develop inhibitors targeting the dimer interface of BRAF. A deeper understanding of BRAF regulation together with more diverse BRAF inhibitors will be beneficial for drug development in RAF or RASdriven cancers.
Collapse
Affiliation(s)
- Amber Gunderwala
- Department of Chemistry & Biochemistry, College of Science and Mathematics, Rowan University, Glassboro, NJ, USA
| | - Nicholas Cope
- Department of Chemistry & Biochemistry, College of Science and Mathematics, Rowan University, Glassboro, NJ, USA
| | - Zhihong Wang
- Department of Chemistry & Biochemistry, College of Science and Mathematics, Rowan University, Glassboro, NJ, USA.
| |
Collapse
|
9
|
Avery TY, Köhler N, Zeiser R, Brummer T, Ruess DA. Onco-immunomodulatory properties of pharmacological interference with RAS-RAF-MEK-ERK pathway hyperactivation. Front Oncol 2022; 12:931774. [PMID: 35965494 PMCID: PMC9363660 DOI: 10.3389/fonc.2022.931774] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2022] [Accepted: 06/30/2022] [Indexed: 12/25/2022] Open
Abstract
Hyperactivation of the RAS-RAF-MEK-ERK cascade - a mitogen-activated protein kinase pathway – has a well-known association with oncogenesis of leading tumor entities, including non-small cell lung cancer, colorectal carcinoma, pancreatic ductal adenocarcinoma, and malignant melanoma. Increasing evidence shows that genetic alterations leading to RAS-RAF-MEK-ERK pathway hyperactivation mediate contact- and soluble-dependent crosstalk between tumor, tumor microenvironment (TME) and the immune system resulting in immune escape mechanisms and establishment of a tumor-sustaining environment. Consequently, pharmacological interruption of this pathway not only leads to tumor-cell intrinsic disruptive effects but also modification of the TME and anti-tumor immunomodulation. At the same time, the importance of ERK signaling in immune cell physiology and potentiation of anti-tumor immune responses through ERK signaling inhibition within immune cell subsets has received growing appreciation. Specifically, a strong case was made for targeted MEK inhibition due to promising associated immune cell intrinsic modulatory effects. However, the successful transition of therapeutic agents interrupting RAS-RAF-MEK-ERK hyperactivation is still being hampered by significant limitations regarding durable efficacy, therapy resistance and toxicity. We here collate and summarize the multifaceted role of RAS-RAF-MEK-ERK signaling in physiology and oncoimmunology and outline the rationale and concepts for exploitation of immunomodulatory properties of RAS-RAF-MEK-ERK inhibition while accentuating the role of MEK inhibition in combinatorial and intermittent anticancer therapy. Furthermore, we point out the extensive scientific efforts dedicated to overcoming the challenges encountered during the clinical transition of various therapeutic agents in the search for the most effective and safe patient- and tumor-tailored treatment approach.
Collapse
Affiliation(s)
- Thomas Yul Avery
- Department of General and Visceral Surgery, Center of Surgery, Medical Center University of Freiburg, Freiburg, Germany
- *Correspondence: Thomas Yul Avery, ; Dietrich Alexander Ruess,
| | - Natalie Köhler
- Department of Medicine I - Medical Center, Medical Center University of Freiburg, Freiburg, Germany
- CIBSS - Centre for Integrative Biological Signalling Studies, University of Freiburg, Freiburg, Germany
| | - Robert Zeiser
- Department of Medicine I - Medical Center, Medical Center University of Freiburg, Freiburg, Germany
- German Cancer Consortium Deutsches Konsortium Translationale Krebsforschung (DKTK), partner site Freiburg, German Cancer Research Center Deutsches Krebsforschungszentrum (DKFZ), Heidelberg, Germany
| | - Tilman Brummer
- German Cancer Consortium Deutsches Konsortium Translationale Krebsforschung (DKTK), partner site Freiburg, German Cancer Research Center Deutsches Krebsforschungszentrum (DKFZ), Heidelberg, Germany
- Institute of Molecular Medicine and Cell Research (IMMZ), Faculty of Medicine, University of Freiburg, Freiburg, Germany
- Comprehensive Cancer Center Freiburg (CCCF), Faculty of Medicine, Medical Center University of Freiburg, Freiburg, Germany
| | - Dietrich Alexander Ruess
- Department of General and Visceral Surgery, Center of Surgery, Medical Center University of Freiburg, Freiburg, Germany
- German Cancer Consortium Deutsches Konsortium Translationale Krebsforschung (DKTK), partner site Freiburg, German Cancer Research Center Deutsches Krebsforschungszentrum (DKFZ), Heidelberg, Germany
- *Correspondence: Thomas Yul Avery, ; Dietrich Alexander Ruess,
| |
Collapse
|
10
|
Kuang P, Xie A, Deng J, Tang J, Wang P, Yu F. GTP-binding protein Di-RAS3 diminishes the migration and invasion of non-small cell lung cancer by inhibiting the RAS/extracellular-regulated kinase pathway. Bioengineered 2022; 13:5663-5674. [PMID: 35170376 PMCID: PMC8973588 DOI: 10.1080/21655979.2022.2031671] [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] [Indexed: 11/08/2022] Open
Abstract
The GTP-binding protein Di-Ras3 (DIRAS3) has been established as a maternally imprinted tumor suppressor gene. Growing evidence has correlated the DIRAS3 gene with tumor progression, but its role in non-small cell lung cancer (NSCLC) is rarely reported. Accordingly, the current study sought to evaluate the role and mechanism of DIRAS3 in NSCLC cell progression. First, we uncovered that DIRAS3 was poorly expressed in NSCLC tissues and cells. Subsequently, we examined the effect of DIRAS3 over-expression or knockdown in different lung cancer cells on their malignant phenotypes, with the help of transwell cell migration and invasion assays, and Western blot analyses. It was found that the over-expression of DIRAS3 inhibited the migration and invasion of A549 cells or H520 cells, whereas knockdown of DIRAS3 led to opposing trends. In addition, over-expression of DIRAS3 attenuated the tumor growth and reduced the number of lung tumor nodules. Mechanistically, DIRAS3 may inhibit the migration and invasion of NSCLC cells by inhibiting the RAS/extracellular-regulated kinase (ERK) signaling pathway. Collectively, our findings indicate that DIRAS3 could serve as a potential therapeutic target biomarker for NSCLC.
Collapse
Affiliation(s)
- Peng Kuang
- Department of Oncology, The First Affiliated Hospital of Nanchang University, Nanchang, China
| | - An Xie
- Jiangxi Institute of Urology, The First Affiliated Hospital of Nanchang University, China
| | - Jianxiong Deng
- Department of Oncology, The First Affiliated Hospital of Nanchang University, Nanchang, China
| | - Jiaming Tang
- Department of Oncology, The First Affiliated Hospital of Nanchang University, Nanchang, China
| | - Peijun Wang
- Department of Oncology, The First Affiliated Hospital of Nanchang University, Nanchang, China
| | - Feng Yu
- Department of Oncology, The First Affiliated Hospital of Nanchang University, Nanchang, China
| |
Collapse
|
11
|
Li SZ, Zhang ZY, Chen J, Dong MY, Du XH, Gao J, Shu QP, Li C, Liang XY, Ding ZH, Du RL, Wang J, Zhang XD. NLK is required for Ras/ERK/SRF/ELK signaling to tune skeletal muscle development by phosphorylating SRF and antagonizing the SRF/MKL pathway. Cell Death Dis 2022; 8:4. [PMID: 35013153 PMCID: PMC8748963 DOI: 10.1038/s41420-021-00774-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2021] [Revised: 11/15/2021] [Accepted: 11/25/2021] [Indexed: 11/23/2022]
Abstract
Serum response factor (SRF) regulates differentiation and proliferation by binding to RhoA-actin-activated MKL or Ras-MAPK-activated ELK transcriptional coactivators, but the molecular mechanisms responsible for SRF regulation remain unclear. Here, we show that Nemo-like kinase (NLK) is required for the promotion of SRF/ELK signaling in human and mouse cells. NLK was found to interact with and phosphorylate SRF at serine residues 101/103, which in turn enhanced the association between SRF and ELK. The enhanced affinity of SRF/ELK antagonized the SRF/MKL pathway and inhibited mouse myoblast differentiation in vitro. In a skeletal muscle-specific Nlk conditional knockout mouse model, forming muscle myofibers underwent hypertrophic growth, resulting in an increased muscle and body mass phenotype. We propose that both phosphorylation of SRF by NLK and phosphorylation of ELKs by MAPK are required for RAS/ELK signaling, confirming the importance of this ancient pathway and identifying an important role for NLK in modulating muscle development in vivo.
Collapse
Affiliation(s)
- Shang-Ze Li
- Hubei Key Laboratory of Cell Homeostasis, College of Life Sciences, Wuhan University, 430072, Wuhan, Hubei, China.,School of Medicine, Chongqing University, 400030, Chongqing, China
| | - Ze-Yan Zhang
- Hubei Key Laboratory of Cell Homeostasis, College of Life Sciences, Wuhan University, 430072, Wuhan, Hubei, China
| | - Jie Chen
- Hubei Key Laboratory of Cell Homeostasis, College of Life Sciences, Wuhan University, 430072, Wuhan, Hubei, China
| | - Ming-You Dong
- Reproductive genetics laboratory, Affiliated hospital of Youjiang Medical University for Nationalities, 533000, Baise, Guangxi, China
| | - Xue-Hua Du
- Hubei Key Laboratory of Cell Homeostasis, College of Life Sciences, Wuhan University, 430072, Wuhan, Hubei, China
| | - Jie Gao
- Hubei Key Laboratory of Cell Homeostasis, College of Life Sciences, Wuhan University, 430072, Wuhan, Hubei, China
| | - Qi-Peng Shu
- Hubei Key Laboratory of Cell Homeostasis, College of Life Sciences, Wuhan University, 430072, Wuhan, Hubei, China
| | - Chao Li
- Hubei Key Laboratory of Cell Homeostasis, College of Life Sciences, Wuhan University, 430072, Wuhan, Hubei, China
| | - Xin-Yi Liang
- Hubei Key Laboratory of Cell Homeostasis, College of Life Sciences, Wuhan University, 430072, Wuhan, Hubei, China
| | - Zhi-Hao Ding
- Hubei Key Laboratory of Cell Homeostasis, College of Life Sciences, Wuhan University, 430072, Wuhan, Hubei, China
| | - Run-Lei Du
- Hubei Key Laboratory of Cell Homeostasis, College of Life Sciences, Wuhan University, 430072, Wuhan, Hubei, China
| | - Junli Wang
- School of Medicine, Chongqing University, 400030, Chongqing, China. .,Reproductive genetics laboratory, Affiliated hospital of Youjiang Medical University for Nationalities, 533000, Baise, Guangxi, China.
| | - Xiao-Dong Zhang
- Hubei Key Laboratory of Cell Homeostasis, College of Life Sciences, Wuhan University, 430072, Wuhan, Hubei, China. .,Reproductive genetics laboratory, Affiliated hospital of Youjiang Medical University for Nationalities, 533000, Baise, Guangxi, China.
| |
Collapse
|
12
|
Sheppard PAS, Puri TA, Galea LAM. Sex Differences and Estradiol Effects in MAPK and Akt Cell Signaling across Subregions of the Hippocampus. Neuroendocrinology 2022; 112:621-635. [PMID: 34407537 DOI: 10.1159/000519072] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/01/2021] [Accepted: 08/16/2021] [Indexed: 11/19/2022]
Abstract
INTRODUCTION Rapid effects of estrogens within the hippocampus of rodents are dependent upon cell-signaling cascades, and activation of these cascades by estrogens varies by sex. Whether these pathways are rapidly activated within the dentate gyrus (DG) and CA1 by estrogens across sex and the anatomical longitudinal axis has been overlooked. METHODS Gonadally intact female and male rats were given either vehicle or physiological systemic low (1.1 µg/kg) or high (37.3 µg/kg) doses of 17β-estradiol 30 min prior to tissue collection. To control for the effects of circulating estrogens, an additional group of female rats was ovariectomized (OVX) and administered 17β-estradiol. Brains were extracted, and tissue punches of the CA1 and DG were taken along the longitudinal hippocampal axis (dorsal and ventral) and analyzed for key mitogen-activated protein kinase (MAPK) and protein kinase B (Akt) cascade phosphoproteins. RESULTS Intact females had higher Akt pathway phosphoproteins (pAkt, pGSK-3β, and pp70S6K) than males in the DG (dorsal and ventral) and lower pERK1/2 in the dorsal DG. Most effects of 17β-estradiol on cell signaling occurred in OVX animals. In OVX animals, 17β-estradiol increased cell signaling of MAPK and Akt phosphoproteins (pERK1/2, pJNK, pAkt, and pGSK-3β) in the CA1 and pERK1/2 and pJNK DG. DISCUSSION/CONCLUSIONS Systemic 17β-estradiol treatment rapidly alters phosphoprotein levels in the hippocampus, dependent on reproductive status, and intact females have greater expression of Akt phosphoproteins than that in intact males in the DG. These findings shed light on underlying mechanisms of sex differences in hippocampal function and response to interventions that affect MAPK or Akt signaling.
Collapse
Affiliation(s)
- Paul A S Sheppard
- Department of Psychology, University of British Columbia, Vancouver, British Columbia, Canada
- Djavad Mowafaghian Centre for Brain Health, University of British Columbia, Vancouver, British Columbia, Canada
| | - Tanvi A Puri
- Djavad Mowafaghian Centre for Brain Health, University of British Columbia, Vancouver, British Columbia, Canada
- Graduate Program in Neuroscience, University of British Columbia, Vancouver, British Columbia, Canada
| | - Liisa A M Galea
- Department of Psychology, University of British Columbia, Vancouver, British Columbia, Canada
- Djavad Mowafaghian Centre for Brain Health, University of British Columbia, Vancouver, British Columbia, Canada
- Graduate Program in Neuroscience, University of British Columbia, Vancouver, British Columbia, Canada
| |
Collapse
|
13
|
Sun Q, Zhou Y, Xiong M, Chen Y, Tan WS, Cai H. MEK1 activation enhances the ex vivo proliferation of haematopoietic stem/progenitor cell. Cell Biochem Funct 2021; 40:79-89. [PMID: 34855220 DOI: 10.1002/cbf.3677] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2021] [Revised: 11/12/2021] [Accepted: 11/16/2021] [Indexed: 11/11/2022]
Abstract
Haematopoietic stem/progenitor cell (HSPC) integrates intracellular signal network from growth factors (GFs) and utilizes its proliferation feature to generate high yields of transplantable cells upon ex vivo culture. However, the molecular basis for HSPC activation and proliferation is not completely understood. The goal of this study was to investigate proliferation regulator in the downstream of GFs and develop HSPC expansion strategy. Microarray and Ingenuity Pathway Analysis were performed to evaluate differentially expressed genes in cytokine-induced CD34+ cells after ex vivo culture. We identified that MEK1 was a potential HSPC proliferation regulator, which represented indispensable roles and MEK1 silence attenuated the proliferation of HSPC. Notably, 500 nM MEK1 agonist, PAF C-16, increased the numbers of phenotypic HSPC and induced cell cycling of HSPC. The PAF C-16 expanded HSPC demonstrated comparative clonal formation ability and secondary expansion capacity compared to the vehicle control. Our results provide insights into regulating the balance between proliferation and commitment of HSPC by targeting the HSPC proliferation-controlling network. This study demonstrates that MEK1 critically regulates HSPC proliferation and cell production in the ex vivo condition for transplantation.
Collapse
Affiliation(s)
- Qihao Sun
- State Key Laboratory of Bioreactor Engineering, East China University of Science and Technology, Shanghai, China
| | - Yiran Zhou
- State Key Laboratory of Bioreactor Engineering, East China University of Science and Technology, Shanghai, China
| | - Minghao Xiong
- State Key Laboratory of Bioreactor Engineering, East China University of Science and Technology, Shanghai, China
| | - Yuying Chen
- State Key Laboratory of Bioreactor Engineering, East China University of Science and Technology, Shanghai, China
| | - Wen-Song Tan
- State Key Laboratory of Bioreactor Engineering, East China University of Science and Technology, Shanghai, China
| | - Haibo Cai
- State Key Laboratory of Bioreactor Engineering, East China University of Science and Technology, Shanghai, China
| |
Collapse
|
14
|
Ma Y, Zhang H, Li X, Liu Y. HAGLROS promotes cell proliferation and angiogenesis and inhibits apoptosis by activating multiple signaling pathways in LSCC cells. J Oral Pathol Med 2021; 51:510-519. [PMID: 34634160 DOI: 10.1111/jop.13249] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2021] [Revised: 09/07/2021] [Accepted: 10/08/2021] [Indexed: 02/01/2023]
Abstract
BACKGROUND HAGLROS is a long noncoding RNA involving in the development of a variety of cancers, but its mechanism of action in laryngeal squamous cell carcinomas (LSCC) is still unclear. We aim to unveil the effect and mechanism of HAGLROS on LSCC. METHODS The expression of HAGLROS in LSCC patients' tissues, serum, and LSCC cell lines was quantified by quantitative real-time PCR. AMC-HN-8 and SNU-46 cells were transfected with the overexpression plasmid of HAGLROS and shHAGLROS, and the functional assay (colony formation assays, flow cytometry, and tube formation) was performed. Western blot was used to determine the expressions of vascular endothelial growth factor (VEGF), proliferating cell nuclear antigen (PCNA), P27 and cleaved caspase-3, as well as phosphorylated-c-Jun-N-terminal kinase (p-JNK), JNK, phosphorylated-extracellular signal-regulated kinase 1/2 (p-Erk1/2), Erk1/2, phosphorylated-protein kinase B (p-AKT) and AKT. RESULTS HAGLROS was highly expressed in LSCC tissues and cells, and it was correlated to lymph node, tumor depth, and clinical stage of LSCC patients. The proliferation ability of LSCC cells was higher than that of HuLa-PC cells. Meanwhile, HAGLROS overexpression promoted the abilities of proliferation and angiogenesis and reduced apoptosis, whereas silencing of HAGLROS exerted the opposite effects in LSCC cell lines. Moreover, overexpressed HAGLROS upregulated the expressions of VEGF and PCNA yet downregulated the expressions of P27 and cleaved caspase-3 by activating Erk1/2 and AKT or JNK signaling pathways in different LSCC cell lines. CONCLUSION Overexpressed HAGLROS promoted the proliferation and angiogenesis yet inhibited apoptosis of LSCC cells by activating Erk1/2 and AKT or JNK signaling pathways.
Collapse
Affiliation(s)
- Yunxia Ma
- Department of Otorhinolaryngology Head and Neck Surgery, The First Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Hui Zhang
- Department of Radiology, The First Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Xiaohong Li
- Department of Otorhinolaryngology Head and Neck Surgery, The First Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Yehai Liu
- Department of Otorhinolaryngology Head and Neck Surgery, The First Affiliated Hospital of Anhui Medical University, Hefei, China
| |
Collapse
|
15
|
Shi W, Wu H, Liu S, Wu Z, Wu H, Liu J, Hou Y. Progesterone Suppresses Cholesterol Esterification in APP/PS1 mice and a cell model of Alzheimer's Disease. Brain Res Bull 2021; 173:162-173. [PMID: 34044033 DOI: 10.1016/j.brainresbull.2021.05.020] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2021] [Revised: 04/27/2021] [Accepted: 05/21/2021] [Indexed: 01/01/2023]
Abstract
AIMS Cholesteryl ester(CE), generated from the mitochondria associated membrane (MAM), is involved in the pathogenesis of Alzheimer's Disease (AD). In theory, the different neuroprotective effects of progesterone in AD are all linked to MAM, yet the effect on cholesterol esterification has not been reported. Therefore, this study was aimed to investigate the regulation of progesterone on intracerebral CE in AD models and the underlying mechanism. METHODS APP/PS1 mice and AD cell model induced by Aβ 25-35 were selected as the research objects. APP/PS1 mice were daily administrated intragastrically with progesterone and The Morris Water Maze test was performed to detect the learning and memory abilities. Intracellular cholesterol was measured by Cholesterol/Cholesteryl Ester Quantitation Assay. The structure of MAMs were observed with transmission electron microscopy. The expression of acyl-CoA: cholesterol acyltransferase 1 (ACAT1), ERK1/2 and p-ERK1/2 were detected with western blotting, immunohistochemistry or immunofluorescence. RESULTS Progesterone suppressed the accumulation of intracellular CE, shortened the length of abnormally prolonged MAM in cortex of APP/PS1 mice. Progesterone decreased the expression of ACAT1, which could be blocked by progesterone receptor membrane component 1 (PGRMC1) inhibitor AG205. The ERK1/2 pathway maybe involved in the progesterone mediated regulation of ACAT1 in AD models, rather than the PI3K/Akt and the P38 MEPK pathways. SIGNIFICANCE The results supported a line of evidence that progesterone regulates CE level and the structure of MAM in neurons of AD models, providing a promising treatment against AD on the dysfunction of cholesterol metabolism.
Collapse
Affiliation(s)
- Wenjing Shi
- Department of Pharmacology, Hebei Medical University, Shijiazhuang 050017, Hebei Province, China; Department of Pharmacy, Hebei General Hospital, Shijiazhuang 050051, Hebei Province, China.
| | - Hang Wu
- Department of Pharmacy, Heze University, Heze 274000, Shandong Province, China.
| | - Sha Liu
- Department of Pharmacy, the Third Hospital of Shijiazhuang, Shijiazhuang 050000, Hebei Province, China.
| | - Zhigang Wu
- Department of Pharmacy, Hebei North University, Hebei Key Laboratory of Neuropharmacology, Zhangjiakou 075000, China.
| | - Honghai Wu
- Department of Pharmacy, Bethune International Peace Hospital, Shijiazhuang 050082, Hebei Province, China.
| | - Jianfang Liu
- Department of Pharmacy, Bethune International Peace Hospital, Shijiazhuang 050082, Hebei Province, China.
| | - Yanning Hou
- Department of Pharmacology, Hebei Medical University, Shijiazhuang 050017, Hebei Province, China; Department of Pharmacy, Bethune International Peace Hospital, Shijiazhuang 050082, Hebei Province, China.
| |
Collapse
|
16
|
Scheffler L, Feicht S, Babushku T, Kuhn LB, Ehrenberg S, Frankenberger S, Lehmann FM, Hobeika E, Jungnickel B, Baccarini M, Bornkamm GW, Strobl LJ, Zimber-Strobl U. ERK phosphorylation is RAF independent in naïve and activated B cells but RAF dependent in plasma cell differentiation. Sci Signal 2021; 14:eabc1648. [PMID: 33975980 DOI: 10.1126/scisignal.abc1648] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/02/2022]
Abstract
Members of the RAF family of serine-threonine kinases are intermediates in the mitogen-activated protein kinase and extracellular signal-regulated kinase (MAPK-ERK) signaling pathway, which controls key differentiation processes in B cells. By analyzing mice with B cell-specific deletion of Raf1, Braf, or both, we showed that Raf-1 and B-Raf acted together in mediating the positive selection of pre-B and transitional B cells as well as in initiating plasma cell differentiation. However, genetic or chemical inactivation of RAFs led to increased ERK phosphorylation in mature B cells. ERK activation in the absence of Raf-1 and B-Raf was mediated by multiple RAF-independent pathways, with phosphoinositide 3-kinase (PI3K) playing an important role. Furthermore, we found that ERK phosphorylation strongly increased during the transition from activated B cells to pre-plasmablasts. This increase in ERK phosphorylation did not occur in B cells lacking both Raf-1 and B-Raf, which most likely explains the partial block of plasma cell differentiation in mice lacking both RAFs. Collectively, our data indicate that B-Raf and Raf-1 are not necessary to mediate ERK phosphorylation in naïve or activated B cells but are essential for mediating the marked increase in ERK phosphorylation during the transition from activated B cells to pre-plasmablasts.
Collapse
Affiliation(s)
- Laura Scheffler
- Research Unit of Gene Vectors, Helmholtz Center Munich, German Research Center for Environmental Health GmbH, Marchioninistrasse 25, D-81377 Munich, Germany
| | - Samantha Feicht
- Research Unit of Gene Vectors, Helmholtz Center Munich, German Research Center for Environmental Health GmbH, Marchioninistrasse 25, D-81377 Munich, Germany
- Institute for Clinical Molecular Biology and Tumor Genetics, Helmholtz Center Munich, German Research Center for Environmental Health GmbH, Marchioninistrasse 25, D-81377 Munich, Germany
| | - Tea Babushku
- Research Unit of Gene Vectors, Helmholtz Center Munich, German Research Center for Environmental Health GmbH, Marchioninistrasse 25, D-81377 Munich, Germany
| | - Laura B Kuhn
- Research Unit of Gene Vectors, Helmholtz Center Munich, German Research Center for Environmental Health GmbH, Marchioninistrasse 25, D-81377 Munich, Germany
| | - Stefanie Ehrenberg
- Research Unit of Gene Vectors, Helmholtz Center Munich, German Research Center for Environmental Health GmbH, Marchioninistrasse 25, D-81377 Munich, Germany
| | - Samantha Frankenberger
- Research Unit of Gene Vectors, Helmholtz Center Munich, German Research Center for Environmental Health GmbH, Marchioninistrasse 25, D-81377 Munich, Germany
| | - Frank M Lehmann
- Institute for Clinical Molecular Biology and Tumor Genetics, Helmholtz Center Munich, German Research Center for Environmental Health GmbH, Marchioninistrasse 25, D-81377 Munich, Germany
| | - Elias Hobeika
- Molecular Immunology, Max Planck Institute of Immunobiology and Epigenetics, Stübeweg 51, D-79108 Freiburg, Germany
- Institute of Immunology, Ulm University Medical Center, Albert-Einstein-Allee 11, D-89070 Ulm, Germany
| | - Berit Jungnickel
- Institute for Clinical Molecular Biology and Tumor Genetics, Helmholtz Center Munich, German Research Center for Environmental Health GmbH, Marchioninistrasse 25, D-81377 Munich, Germany
- Department of Cell Biology, Institute of Biochemistry and Biophysics, Center for Molecular Biomedicine, Friedrich-Schiller University Jena, Hans-Knoell-Strasse 2, D-07745 Jena, Germany
| | - Manuela Baccarini
- Department of Microbiology, Immunobiology, and Genetics, Center for Molecular Biology of the University of Vienna, Max Perutz Labs, Dr. Bohr-Gasse 9, 1030 Vienna, Austria
| | - Georg W Bornkamm
- Institute for Clinical Molecular Biology and Tumor Genetics, Helmholtz Center Munich, German Research Center for Environmental Health GmbH, Marchioninistrasse 25, D-81377 Munich, Germany
| | - Lothar J Strobl
- Research Unit of Gene Vectors, Helmholtz Center Munich, German Research Center for Environmental Health GmbH, Marchioninistrasse 25, D-81377 Munich, Germany
| | - Ursula Zimber-Strobl
- Research Unit of Gene Vectors, Helmholtz Center Munich, German Research Center for Environmental Health GmbH, Marchioninistrasse 25, D-81377 Munich, Germany.
| |
Collapse
|
17
|
Fleischmann J, Feichtner A, DeFalco L, Kugler V, Schwaighofer S, Huber RG, Stefan E. Allosteric Kinase Inhibitors Reshape MEK1 Kinase Activity Conformations in Cells and In Silico. Biomolecules 2021; 11:518. [PMID: 33808483 PMCID: PMC8065916 DOI: 10.3390/biom11040518] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2021] [Revised: 03/26/2021] [Accepted: 03/27/2021] [Indexed: 01/26/2023] Open
Abstract
Mutations at different stages of the mitogen-activated protein kinase (MAPK) signaling pathway lead to aberrant activation of the involved protein kinase entities. These oncogenic modifications alter signal propagation which converge on the gatekeeper kinases MEK1/2, transmitting the input signal to ERK1/2. Thus, targeted MEK inhibition causes qualitative alterations of carcinogenic MAPK signals. Phosphorylation of the MEK1 activation loop at the positions S218 and S222 by RAF kinases triggers the conformational alignment of MEK's catalytic pocket to enable ATP-binding and substrate phosphorylation. We have extended a kinase conformation (KinCon) biosensor platform to record MEK1 activity dynamics. In addition to MEK phosphorylation by BRAF, the integration of the phosphorylation-mimetic mutations S218D/S222D triggered opening of the kinase. Structural rearrangement may involve the flexibility of the N terminal MEK1 A-helix. Application of the allosterically acting MEK inhibitors (MEKi) trametinib, cobimentinib, refametinib, and selumetinib converted activated MEK1 KinCon reporters back into a more closed inactive conformation. We confirmed MEK1 KinCon activity dynamics upon drug engagement using the patient-derived melanoma cell line A2058, which harbors the V600E hotspot BRAF mutation. In order to confirm biosensor dynamics, we simulated structure dynamics of MEK1 kinase in the presence and absence of mutations and/or MEKi binding. We observed increased dynamics for the S218D/S222D double mutant particularly in the region of the distal A-helix and alpha-C helix. These data underline that MEK1 KinCon biosensors have the potential to be subjected to MEKi efficacy validations in an intact cell setting.
Collapse
Affiliation(s)
- Jakob Fleischmann
- Institute of Biochemistry and Center for Molecular Biosciences Innsbruck, University of Innsbruck, Innrain 80/82, 6020 Innsbruck, Austria; (J.F.); (A.F.); (V.K.); (S.S.)
| | - Andreas Feichtner
- Institute of Biochemistry and Center for Molecular Biosciences Innsbruck, University of Innsbruck, Innrain 80/82, 6020 Innsbruck, Austria; (J.F.); (A.F.); (V.K.); (S.S.)
| | - Louis DeFalco
- Bioinformatics Institute (BII), Agency for Science Technology and Research (A*STAR), Singapore 138671, Singapore; (L.D.); (R.G.H.)
| | - Valentina Kugler
- Institute of Biochemistry and Center for Molecular Biosciences Innsbruck, University of Innsbruck, Innrain 80/82, 6020 Innsbruck, Austria; (J.F.); (A.F.); (V.K.); (S.S.)
| | - Selina Schwaighofer
- Institute of Biochemistry and Center for Molecular Biosciences Innsbruck, University of Innsbruck, Innrain 80/82, 6020 Innsbruck, Austria; (J.F.); (A.F.); (V.K.); (S.S.)
- Tyrolean Cancer Research Institute, Innrain 66, 6020 Innsbruck, Austria
| | - Roland G Huber
- Bioinformatics Institute (BII), Agency for Science Technology and Research (A*STAR), Singapore 138671, Singapore; (L.D.); (R.G.H.)
| | - Eduard Stefan
- Institute of Biochemistry and Center for Molecular Biosciences Innsbruck, University of Innsbruck, Innrain 80/82, 6020 Innsbruck, Austria; (J.F.); (A.F.); (V.K.); (S.S.)
- Tyrolean Cancer Research Institute, Innrain 66, 6020 Innsbruck, Austria
| |
Collapse
|
18
|
Koyama-Honda I, Fujiwara TK, Kasai RS, Suzuki KGN, Kajikawa E, Tsuboi H, Tsunoyama TA, Kusumi A. High-speed single-molecule imaging reveals signal transduction by induced transbilayer raft phases. J Cell Biol 2021; 219:211461. [PMID: 33053147 PMCID: PMC7563750 DOI: 10.1083/jcb.202006125] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2020] [Revised: 08/09/2020] [Accepted: 09/14/2020] [Indexed: 12/21/2022] Open
Abstract
Using single-molecule imaging with enhanced time resolutions down to 5 ms, we found that CD59 cluster rafts and GM1 cluster rafts were stably induced in the outer leaflet of the plasma membrane (PM), which triggered the activation of Lyn, H-Ras, and ERK and continually recruited Lyn and H-Ras right beneath them in the inner leaflet with dwell lifetimes <0.1 s. The detection was possible due to the enhanced time resolutions employed here. The recruitment depended on the PM cholesterol and saturated alkyl chains of Lyn and H-Ras, whereas it was blocked by the nonraftophilic transmembrane protein moiety and unsaturated alkyl chains linked to the inner-leaflet molecules. Because GM1 cluster rafts recruited Lyn and H-Ras as efficiently as CD59 cluster rafts, and because the protein moieties of Lyn and H-Ras were not required for the recruitment, we conclude that the transbilayer raft phases induced by the outer-leaflet stabilized rafts recruit lipid-anchored signaling molecules by lateral raft-lipid interactions and thus serve as a key signal transduction platform.
Collapse
Affiliation(s)
- Ikuko Koyama-Honda
- Department of Biochemistry and Molecular Biology, Graduate School and Faculty of Medicine, University of Tokyo, Tokyo, Japan
| | - Takahiro K Fujiwara
- Institute for Integrated Cell-Material Sciences, Kyoto University, Kyoto, Japan
| | - Rinshi S Kasai
- Institute for Frontier Life and Medical Sciences, Kyoto University, Kyoto, Japan
| | - Kenichi G N Suzuki
- Institute for Integrated Cell-Material Sciences, Kyoto University, Kyoto, Japan.,Institute for Glyco-core Research, Gifu University, Nagoya, Japan.,Center for Highly Advanced Integration of Nano and Life Sciences, Gifu University, Gifu, Japan
| | - Eriko Kajikawa
- Laboratory for Organismal Patterning, Center for Biosystems Dynamics Research, RIKEN Kobe, Kobe, Japan
| | - Hisae Tsuboi
- Membrane Cooperativity Unit, Okinawa Institute of Science and Technology Graduate University, Onna-son, Okinawa, Japan
| | - Taka A Tsunoyama
- Membrane Cooperativity Unit, Okinawa Institute of Science and Technology Graduate University, Onna-son, Okinawa, Japan
| | - Akihiro Kusumi
- Membrane Cooperativity Unit, Okinawa Institute of Science and Technology Graduate University, Onna-son, Okinawa, Japan
| |
Collapse
|
19
|
Abstract
Introduction Neurofibromin, a protein encoded by the NF1 gene, is mutated in neurofibromatosis 1, one of the most common genetic diseases. Oral manifestations are common and a high prevalence of hyposalivation was recently described in individuals with neurofibromatosis 1. Although neurofibromin is ubiquitously expressed, its expression levels vary depending on the tissue type and developmental stage of the organism. The role of neurofibromin in the development, morphology, and physiology of salivary glands is unknown and a detailed expression of neurofibromin in human normal salivary glands has never been investigated. Aim To investigate the expression levels and distribution of neurofibromin in acinar and ductal cells of major and minor salivary glands of adult individuals without NF1. Material and method Ten samples of morphologically normal major and minor salivary glands (three samples of each gland: parotid, submandibular and minor salivary; and one sample of sublingual gland) from individuals without neurofibromatosis 1 were selected to assess neurofibromin expression through immunohistochemistry. Immunoquantification was performed by a digital method. Results Neurofibromin was expressed in the cytoplasm of both serous and mucous acinar cells, as well as in ducts from all the samples of salivary glands. Staining intensity varied from mild to strong depending on the type of salivary gland and region (acini or ducts). Ducts had higher neurofibromin expression than acinar cells (p = 0.003). There was no statistical association between the expression of neurofibromin and the type of the salivary gland, considering acini (p = 0.09) or ducts (p = 0.50) of the four salivary glands (parotid, submandibular, minor salivary, and sublingual gland). Similar results were obtained comparing the acini (p = 0.35) and ducts (p = 0.50) of minor and major salivary glands. Besides, there was no correlation between the expression of neurofibromin and age (p = 0.08), and sex (p = 0.79) of the individuals, considering simultaneously the neurofibromin levels of acini and duct (n = 34). Conclusion Neurofibromin is expressed in the cytoplasm of serous and mucous acinar cells, and ductal cells of salivary glands, suggesting that this protein is important for salivary gland function.
Collapse
|
20
|
Myricetin: A review of the most recent research. Biomed Pharmacother 2020; 134:111017. [PMID: 33338751 DOI: 10.1016/j.biopha.2020.111017] [Citation(s) in RCA: 162] [Impact Index Per Article: 32.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2020] [Revised: 11/09/2020] [Accepted: 11/11/2020] [Indexed: 12/13/2022] Open
Abstract
Myricetin(MYR) is a flavonoid compound widely found in many natural plants including bayberry. So far, MYR has been proven to have multiple biological functions and it is a natural compound with promising research and development prospects. This review comprehensively retrieved and collected the latest pharmacological abstracts on MYR, and discussed the potential molecular mechanisms of its effects. The results of our review indicated that MYR has a therapeutic effect on many diseases, including tumors of different types, inflammatory diseases, atherosclerosis, thrombosis, cerebral ischemia, diabetes, Alzheimer's disease and pathogenic microbial infections. Furthermore, it regulates the expression of Hippo, MAPK, GSK-3β, PI3K/AKT/mTOR, STAT3, TLR, IκB/NF-κB, Nrf2/HO-1, ACE, eNOS / NO, AChE and BrdU/NeuN. MYR also enhances the immunomodulatory functions, suppresses cytokine storms, improves cardiac dysfunction, possesses an antiviral potential, can be used as an adjuvant treatment against cancer, cardiovascular injury and nervous system diseases, and it may be a potential drug against COVID-19 and other viral infections. Generally, this article provides a theoretical basis for the clinical application of MYR and a reference for its further use.
Collapse
|
21
|
Li M, Song SW, Ge Y, Jin JY, Li XY, Tan XD. The Ras-ERK signaling pathway regulates acetylated activating transcription factor 2 via p300 in pancreatic cancer cells. ANNALS OF TRANSLATIONAL MEDICINE 2020; 8:1234. [PMID: 33178766 PMCID: PMC7607129 DOI: 10.21037/atm-20-5880] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
Background Activating transcription factor 2 (ATF2) regulates the expression of downstream target genes and is phosphorylated by the Ras-extracellular-signal-regulated kinase (ERK) pathway. Acetylation of ATF2 is necessary for this type of regulation. However, the molecular mechanism by which the Ras-ERK pathway mediates the regulation of acetylated ATF2 is unknown. This study investigates the mechanism of Ras-ERK pathway-mediated regulation of acetylated ATF2 in maintaining the characteristic phenotype of pancreatic cancer cells. Methods This study was carried out using ASPC-1 and BXPC-3 pancreatic cancer cell lines transfected with the double mutant RasG12V/T35S. The levels of phosphorylated ERK1/2 were measured to establish the activated Ras-ERK pathway. The regulation of acetylated ATF2 was examined by detecting the protein level using western blotting, and the effects on cancer cell phenotype were measured using cell viability, proliferation, migration, and apoptosis assays. Also, chromatin immunoprecipitation (ChIP) assays were used to measure the effect on respective downstream target genes. Results The results showed that RasG12V/T35S reduced the level of acetylated ATF2 in ASPC-1 and BXPC-3 cells. Compared to wild-type ATF2, the mutant ATF2K357Q (which mimics the irreversible acetylated form of ATF2) reduced the cancer cell phenotype and showed decreased enrichment on target genes upon transfection with Ras. Moreover, the level of acetylated ATF2 was regulated by the degradation of p300 through E3 ubiquitin ligase mouse double minute 2 homolog (MDM2). Conclusions Activation of the Ras-ERK pathway regulates acetylated ATF2 through degradation of p300 via a proteasome-dependent pathway, which alters the transcription of downstream target genes responsible for the cancer cell phenotype.
Collapse
Affiliation(s)
- Mu Li
- Department of General Surgery, Shengjing Hospital of China Medical University, Shenyang, China
| | - Shao-Wei Song
- Department of General Surgery, the First Affiliated Hospital of China Medical University, Shenyang, China
| | - Yang Ge
- Department of General Surgery, Shengjing Hospital of China Medical University, Shenyang, China
| | - Jun-Yi Jin
- Department of General Surgery, Shengjing Hospital of China Medical University, Shenyang, China
| | - Xiao-Ying Li
- Department of General Surgery, the First Affiliated Hospital of China Medical University, Shenyang, China
| | - Xiao-Dong Tan
- Department of General Surgery, Shengjing Hospital of China Medical University, Shenyang, China
| |
Collapse
|
22
|
Reischmann N, Andrieux G, Griffin R, Reinheckel T, Boerries M, Brummer T. BRAF V600E drives dedifferentiation in small intestinal and colonic organoids and cooperates with mutant p53 and Apc loss in transformation. Oncogene 2020; 39:6053-6070. [PMID: 32792685 PMCID: PMC7498370 DOI: 10.1038/s41388-020-01414-9] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2020] [Revised: 07/19/2020] [Accepted: 07/29/2020] [Indexed: 12/19/2022]
Abstract
BRAFV600E confers poor prognosis and is associated with a distinct subtype of colorectal cancer (CRC). Little is known, however, about the genetic events driving the initiation and progression of BRAFV600E mutant CRCs. Recent genetic analyses of CRCs indicate that BRAFV600E often coexists with alterations in the WNT- and p53 pathways, but their cooperation remains ill-defined. Therefore, we systematically compared small and large intestinal organoids from mice harboring conditional BraffloxV600E, Trp53LSL-R172H, and/or Apcflox/flox alleles. Using these isogenic models, we observe tissue-specific differences toward sudden BRAFV600E expression, which can be attributed to different ERK-pathway ground states in small and large intestinal crypts. BRAFV600E alone causes transient proliferation and suppresses epithelial organization, followed by organoid disintegration. Moreover, BRAFV600E induces a fetal-like dedifferentiation transcriptional program in colonic organoids, which resembles human BRAFV600E-driven CRC. Co-expression of p53R172H delays organoid disintegration, confers anchorage-independent growth, and induces invasive properties. Interestingly, p53R172H cooperates with BRAFV600E to modulate the abundance of transcripts linked to carcinogenesis, in particular within colonic organoids. Remarkably, WNT-pathway activation by Apc deletion fully protects organoids against BRAFV600E-induced disintegration and confers growth/niche factor independence. Still, Apc-deficient BRAFV600E-mutant organoids remain sensitive toward the MEK inhibitor trametinib, albeit p53R172H confers partial resistance against this clinically relevant compound. In summary, our systematic comparison of the response of small and large intestinal organoids to oncogenic alterations suggests colonic organoids to be better suited to model the human situation. In addition, our work on BRAF-, p53-, and WNT-pathway mutations provides new insights into their cooperation and for the design of targeted therapies.
Collapse
Affiliation(s)
- Nadine Reischmann
- Institute of Molecular Medicine and Cell Research (IMMZ), University of Freiburg, Freiburg, Germany.,Spemann Graduate School of Biology and Medicine (SGBM), University of Freiburg, Freiburg, Germany.,Faculty of Biology, University of Freiburg, Freiburg, Germany
| | - Geoffroy Andrieux
- Institute of Medical Bioinformatics and Systems Medicine, Medical Center-University of Freiburg, Freiburg, Germany.,German Cancer Consortium (DKTK), partner site Freiburg; and German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Ricarda Griffin
- Institute of Molecular Medicine and Cell Research (IMMZ), University of Freiburg, Freiburg, Germany
| | - Thomas Reinheckel
- Institute of Molecular Medicine and Cell Research (IMMZ), University of Freiburg, Freiburg, Germany.,German Cancer Consortium (DKTK), partner site Freiburg; and German Cancer Research Center (DKFZ), Heidelberg, Germany.,Comprehensive Cancer Centre Freiburg (CCCF), University Medical Center Freiburg, University of Freiburg, Freiburg, Germany.,Centre for Biological Signalling Studies BIOSS, University of Freiburg, Freiburg, Germany
| | - Melanie Boerries
- Institute of Medical Bioinformatics and Systems Medicine, Medical Center-University of Freiburg, Freiburg, Germany.,German Cancer Consortium (DKTK), partner site Freiburg; and German Cancer Research Center (DKFZ), Heidelberg, Germany.,Comprehensive Cancer Centre Freiburg (CCCF), University Medical Center Freiburg, University of Freiburg, Freiburg, Germany
| | - Tilman Brummer
- Institute of Molecular Medicine and Cell Research (IMMZ), University of Freiburg, Freiburg, Germany. .,German Cancer Consortium (DKTK), partner site Freiburg; and German Cancer Research Center (DKFZ), Heidelberg, Germany. .,Comprehensive Cancer Centre Freiburg (CCCF), University Medical Center Freiburg, University of Freiburg, Freiburg, Germany. .,Centre for Biological Signalling Studies BIOSS, University of Freiburg, Freiburg, Germany.
| |
Collapse
|
23
|
The Ras-ERK1/2 signaling pathway regulates H3K9ac through PCAF to promote the development of pancreatic cancer. Life Sci 2020; 256:117936. [PMID: 32531376 DOI: 10.1016/j.lfs.2020.117936] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2019] [Revised: 05/21/2020] [Accepted: 06/07/2020] [Indexed: 12/11/2022]
Abstract
AIMS The regulation of the Ras-ERK pathway is the crucial point in pancreatic carcinogenesis, and the Ras kinase is an essential regulatory upstream signal molecule of the ERK1/2 pathway. H3K9ac is a vital histone modification, but its specific role in pancreatic cancer remains unclear. This research aims to study whether the modification level of H3K9ac can regulate the characteristic phenotype of the pancreatic cancer cells by affecting the downstream expression, proliferation, migration, and other related genes. MAIN METHODS The RasG12V/T35S were used to transfect pancreatic cancer cells, and the levels of phosphorylated ERK1/2 and H3K9ac were detected by western blotting. The colony formation assay, transwell assay, and chromatin immunoprecipitation assay were used to study cell viability, migration, and the downstream genes of the ERK1/2 pathway. KEY FINDINGS The results showed that Ras ERK1/2 reduced H3K9ac expression in ASPC-1 cells, and H3K9ac significantly repressed the viability of cells, colony formation, and ASPC-1 cell movement induced by Ras ERK1/2. Besides, HDAC1 silencing increased H3K9ac expression, and changed the effect of Ras ERK1/2 on ASPC-1 cells proliferation, its movement, and mRNAs of ERK1/2 downstream genes. Moreover, Ras ERK1/2 inhibited H3K9ac expression by the degradation of PCAF via MDM2. SIGNIFICANCE Ras ERK1/2 promotes pancreatic carcinogenesis cell movement, through down-regulating H3K9ac via MDM2 mediated PCAF degradation.
Collapse
|
24
|
Xu M, Sun J, Yu Y, Pang Q, Lin X, Barakat M, Lei R, Xu J. TM4SF1 involves in miR-1-3p/miR-214-5p-mediated inhibition of the migration and proliferation in keloid by regulating AKT/ERK signaling. Life Sci 2020; 254:117746. [PMID: 32376266 DOI: 10.1016/j.lfs.2020.117746] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2019] [Revised: 04/17/2020] [Accepted: 04/30/2020] [Indexed: 02/06/2023]
Abstract
AIMS Transmembrane 4 L six family member 1 (TM4SF1) is a small plasma membrane glycoprotein that is highly expressed in cancers. However, the role of TM4SF1 that plays in keloids remains unknown. We investigated the expression, function and the microRNA (miRNA) regulatory network of TM4SF1 in keloids. MAIN METHODS Small interfering RNAs and lentivirus were used to alter the expression of TM4SF1 in fibroblasts. Dual-luciferase reporter assays were applied to determine the miRNA targets. Immunohistochemistry, western blotting, qRT-PCR, wound healing assays, Transwell assays, cell count kit-8 assays and flow cytometry were also employed in this study. KEY FINDINGS TM4SF1 was frequently upregulated in human keloid fibroblasts (HKFs) compared with human normal skin fibroblasts (HSFs). The downregulation of TM4SF1 significantly inhibited proliferation and migration, and induced apoptosis in HKFs. Furthermore, si-TM4SF1 inhibited the AKT/ERK signaling. Meanwhile, the upregulation of TM4SF1 promoted proliferation, migration and the activation of AKT/ERK signaling in human foreskin fibroblasts (HFF-1). Moreover, TM4SF1 can be regulated by miRNAs, which have been validated to play important roles in keloids by posttranscriptional regulation of gene expression. After screening, we found miR-1-3p and miR-214-5p targeted TM4SF1, inhibited TM4SF1 expression, cell proliferation, migration, and induced apoptosis in HKFs. And the level of miR-1-3p and miR-214-5p were found lower in HKFs than in HSFs. SIGNIFICANCE Our study demonstrates a novel regulatory mechanism by which miR-1-3p, miR-214-5p, and TM4SF1 are involved in proliferation, cell motility, and apoptosis, suggesting that they may be potential targets in therapies for keloids.
Collapse
Affiliation(s)
- Mingyuan Xu
- Department of Plastic Surgery, the First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, China
| | - Jiaqi Sun
- Department of Plastic Surgery, the First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, China
| | - Yijia Yu
- Department of Plastic Surgery, the First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, China
| | - Qianqian Pang
- Department of Plastic Surgery, the First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, China
| | - Xiaohu Lin
- Department of Plastic Surgery, the First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, China
| | - May Barakat
- Center for Wound Healing and Tissue Regeneration, College of Dentistry, University of Illinois at Chicago, Chicago, IL, United States
| | - Rui Lei
- Department of Plastic Surgery, the First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, China
| | - Jinghong Xu
- Department of Plastic Surgery, the First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, China.
| |
Collapse
|
25
|
Brummer T, McInnes C. RAF kinase dimerization: implications for drug discovery and clinical outcomes. Oncogene 2020; 39:4155-4169. [PMID: 32269299 DOI: 10.1038/s41388-020-1263-y] [Citation(s) in RCA: 32] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2019] [Revised: 03/04/2020] [Accepted: 03/06/2020] [Indexed: 12/19/2022]
Abstract
The RAF kinases activated by RAS GTPases regulate cell growth and division by signal transduction through the ERK cascade and mutations leading to constitutive activity are key drivers of human tumors, as are upstream activators including RAS and receptor tyrosine kinases. The development of first-generation RAF inhibitors, including vemurafenib (VEM) and dabrafenib led to initial excitement due to high response rates and profound regression of malignant melanomas carrying BRAFV600E mutations. The excitement about these unprecedented response rates, however, was tempered by tumor unresponsiveness through both intrinsic and acquired drug-resistance mechanisms. In recent years much insight into the complexity of the RAS-RAF axis has been obtained and inactivation and signal transduction mechanisms indicate that RAF dimerization is a critical step in multiple cellular contexts and plays a key role in resistance. Both homo- and hetero-dimerization of BRAF and CRAF can modulate therapeutic response and disease progression in patients treated with ATP-competitive inhibitors and are therefore highly clinically significant. Ten years after the definition of the RAF dimer interface (DIF) by crystallography, this review focuses on the implications of RAF kinase dimerization in signal transduction and for drug development, both from a classical ATP-competitive standpoint and from the perspective of new therapeutic strategies including inhibiting dimer formation. A structural perspective of the DIF, how dimerization impacts inhibitor activation and the structure-based design of next-generation RAF kinase inhibitors with unique mechanisms of action is presented. We also discuss potential fields of application for DIF inhibitors, ranging from non-V600E oncoproteins and BRAF fusions to tumors driven by aberrant receptor tyrosine kinase or RAS signaling.
Collapse
Affiliation(s)
- Tilman Brummer
- Institute of Molecular Medicine and Cell Research, Faculty of Medicine, University of Freiburg, Stefan-Meier-Strasse 17, 79104, Freiburg im Breisgau, Germany.,German Cancer Consortium DKTK Partner Site Freiburg, German Cancer Research Center (DKFZ), Heidelberg, Germany.,Comprehensive Cancer Centre Freiburg, University of Freiburg, Freiburg im Breisgau, Germany
| | - Campbell McInnes
- Drug Discovery and Biomedical Sciences, University of South Carolina, Columbia, SC, 29208, USA.
| |
Collapse
|
26
|
Pan YJ, Wan J, Wang CB. MiR-326: Promising Biomarker for Cancer. Cancer Manag Res 2019; 11:10411-10418. [PMID: 31849530 PMCID: PMC6912009 DOI: 10.2147/cmar.s223875] [Citation(s) in RCA: 39] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2019] [Accepted: 12/02/2019] [Indexed: 02/06/2023] Open
Abstract
MicroRNAs (miRNAs) are small non-coding and highly conserved RNAs that act in biological processes including cell proliferation, invasion, apoptosis, metabolism, signal transduction, and tumorigenesis. The previously identified miRNA-326 (miR-326) has been reported to participate in cellular apoptosis, tumor growth, cell invasion, embryonic development, immunomodulation, chemotherapy resistance, and oncogenesis. This review presents a detailed overview of what is known about the effects of miR-326 on cell invasion, metastasis, drug resistance, proliferation, apoptosis, and its involvement in signaling pathways.
Collapse
Affiliation(s)
- Yao-Jie Pan
- Department of Oncology, The Affiliated Yancheng Hospital of Medicine School of Southeast University, The Third People’s Hospital of Yancheng, Yancheng224001, People’s Republic of China
| | - Jian Wan
- Department of General Surgery, Center for Difficult and Complicated Abdominal Surgery, Shanghai Tenth People’s Hospital, Tongji University School of Medicine, Shanghai200092, People’s Republic of China
| | - Chun-Bin Wang
- Department of Oncology, The Affiliated Yancheng Hospital of Medicine School of Southeast University, The Third People’s Hospital of Yancheng, Yancheng224001, People’s Republic of China
| |
Collapse
|
27
|
Shi S, Zhang J, Liu M, Dong H, Li N. Ras-ERK signalling represses H1.4 phosphorylation at serine 36 to promote non-small-cell lung carcinoma cells growth and migration. ARTIFICIAL CELLS NANOMEDICINE AND BIOTECHNOLOGY 2019; 47:2343-2351. [PMID: 31184227 DOI: 10.1080/21691401.2019.1624558] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
Recent papers suggest that oncogenic Ras participate in regulating tumour cells proliferation and metastasis. This work linked Ras with H1.4 modification in non-small-cell lung carcinoma (NSCLC), to better understand the oncogenic effects of Ras. A plasmid for expressing Ras mutated at G13D and T35S was transfected into NCI-H2126 and A549 cells. Phosphorylation of H1.4S36 was determined by immunoblotting. Effects of phosphorylation of H1.4 at serine (S) 36 (H1.4S36ph) on NCI-H2126 and A549 cells were tested by MTT assay, soft-agar colony formation assay, flow cytometry and transwell assay. Chromatin-immunoprecipitation (ChIP) and RT-qPCR were conducted to measure the effects of H1.4S36ph on Ras downstream genes. The catalyzing enzymes participate in H1.4S36 phosphorylation were further studied. We found that Ras-ERK signalling repressed the phosphorylation of H1.4 at S36. H1.4S36ph functioned as a tumour suppressor, as its overexpression repressed NCI-H2126 and A549 cells viability, colony formation, S-phase arrest, migration and invasion. H1.4S36ph was able to mediate the transcription of Ras downstream genes. Ras-ERK signalling repressed H1.4S36ph through degradation of PKA, and the degradation was mediated by MDM2. In conclusion, Ras-ERK signalling repressed H1.4 phosphorylation at S36 to participate in NSCLC cells growth, migration and invasion. Ras-ERK signalling repressed H1.4S36ph through MDM2-dependent degradation of PKA. This study provides a novel explanation for Ras-ERK's tumour-promoting function. Highlights: H1.4S36 phosphorylation is repressed by Ras-ERK activation; H1.4S36ph inhibits the phenotype of NSCLC cells; H1.4S36ph regulates the transcription of Ras downstream genes; Ras-ERK represses H1.4S36ph by MDM2-dependent degradation of PKA.
Collapse
Affiliation(s)
- Shaomin Shi
- a Department of Respiratory, China-Japan Union Hospital of Jilin University , Changchun , China
| | - Jingzhe Zhang
- b Department of Orthopedics, China-Japan Union Hospital of Jilin University , Changchun , China
| | - Meihan Liu
- c Department of Ultrasound, China-Japan Union Hospital of Jilin University , Changchun , China
| | - Hang Dong
- b Department of Orthopedics, China-Japan Union Hospital of Jilin University , Changchun , China
| | - Ning Li
- a Department of Respiratory, China-Japan Union Hospital of Jilin University , Changchun , China
| |
Collapse
|
28
|
Cdh4 Down-Regulation Impairs in Vivo Infiltration and Malignancy in Patients Derived Glioblastoma Cells. Int J Mol Sci 2019; 20:ijms20164028. [PMID: 31426573 PMCID: PMC6718984 DOI: 10.3390/ijms20164028] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2019] [Revised: 08/13/2019] [Accepted: 08/15/2019] [Indexed: 12/17/2022] Open
Abstract
The high invasive phenotype of glioblastoma is one of the main causes of therapy inefficacy and tumor relapse. Cell adhesion molecules of the cadherin family are involved in cell migration and are known as master regulators of epithelial tumor invasiveness, but their role in glioblastoma is less understood. In particular, we recently demonstrated, in the syngeneic murine model, the occurrence of a previously undescribed cadherin switch between Cdh2 and Cdh4 during gliomagenesis, which is necessary for the acquisition of the highly infiltrative and tumorigenic phenotype of these cells. In the present study, we tested the role of Cdh4 in human gliomas. Our results on patient-derived glioma cells demonstrate a positive correlation between Cdh4 expression levels and the loss of cell-cell contact inhibition of proliferation controls that allows cells to proliferate over confluence. Moreover, the silencing of Cdh4 by artificial microRNAs induced a decrease in the infiltrative ability of human glioma cells both in vitro and in vivo. More strikingly, Cdh4 silencing induced an impairment of the tumorigenic potential of these cells after orthotopic transplantation in immunodeficient mice. Overall, we conclude that in human glioblastoma, Cdh4 can also actively contribute in regulating cell invasiveness and malignancy.
Collapse
|
29
|
Ogawa F, Walters MS, Shafquat A, O'Beirne SL, Kaner RJ, Mezey JG, Zhang H, Leopold PL, Crystal RG. Role of KRAS in regulating normal human airway basal cell differentiation. Respir Res 2019; 20:181. [PMID: 31399087 PMCID: PMC6688249 DOI: 10.1186/s12931-019-1129-4] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2018] [Accepted: 07/08/2019] [Indexed: 01/25/2023] Open
Abstract
BACKGROUND KRAS is a GTPase that activates pathways involved in cell growth, differentiation and survival. In normal cells, KRAS-activity is tightly controlled, but with specific mutations, the KRAS protein is persistently activated, giving cells a growth advantage resulting in cancer. While a great deal of attention has been focused on the role of mutated KRAS as a common driver mutation for lung adenocarcinoma, little is known about the role of KRAS in regulating normal human airway differentiation. METHODS To assess the role of KRAS signaling in regulating differentiation of the human airway epithelium, primary human airway basal stem/progenitor cells (BC) from nonsmokers were cultured on air-liquid interface (ALI) cultures to mimic the airway epithelium in vitro. Modulation of KRAS signaling was achieved using siRNA-mediated knockdown of KRAS or lentivirus-mediated over-expression of wild-type KRAS or the constitutively active G12 V mutant. The impact on differentiation was quantified using TaqMan quantitative PCR, immunofluorescent and immunohistochemical staining analysis for cell type specific markers. Finally, the impact of cigarette smoke exposure on KRAS and RAS protein family activity in the airway epithelium was assessed in vitro and in vivo. RESULTS siRNA-mediated knockdown of KRAS decreased differentiation of BC into secretory and ciliated cells with a corresponding shift toward squamous cell differentiation. Conversely, activation of KRAS signaling via lentivirus mediated over-expression of the constitutively active G12 V KRAS mutant had the opposite effect, resulting in increased secretory and ciliated cell differentiation and decreased squamous cell differentiation. Exposure of BC to cigarette smoke extract increased KRAS and RAS protein family activation in vitro. Consistent with these observations, airway epithelium brushed from healthy smokers had elevated RAS activation compared to nonsmokers. CONCLUSIONS Together, these data suggest that KRAS-dependent signaling plays an important role in regulating the balance of secretory, ciliated and squamous cell differentiation of the human airway epithelium and that cigarette smoking-induced airway epithelial remodeling is mediated in part by abnormal activation of KRAS-dependent signaling mechanisms.
Collapse
Affiliation(s)
- Fumihiro Ogawa
- Department of Genetic Medicine, Weill Cornell Medical College, 1300 York Avenue, Box 164, New York, NY, 10065, USA
| | - Matthew S Walters
- Pulmonary, Critical Care & Sleep Medicine, Department of Medicine, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
| | - Afrah Shafquat
- Computational Biology, Cornell University, Ithaca, NY, USA
| | - Sarah L O'Beirne
- Department of Genetic Medicine, Weill Cornell Medical College, 1300 York Avenue, Box 164, New York, NY, 10065, USA
| | - Robert J Kaner
- Department of Genetic Medicine, Weill Cornell Medical College, 1300 York Avenue, Box 164, New York, NY, 10065, USA
| | - Jason G Mezey
- Department of Genetic Medicine, Weill Cornell Medical College, 1300 York Avenue, Box 164, New York, NY, 10065, USA.,Computational Biology, Cornell University, Ithaca, NY, USA
| | - Haijun Zhang
- Department of Genetic Medicine, Weill Cornell Medical College, 1300 York Avenue, Box 164, New York, NY, 10065, USA
| | - Philip L Leopold
- Department of Genetic Medicine, Weill Cornell Medical College, 1300 York Avenue, Box 164, New York, NY, 10065, USA
| | - Ronald G Crystal
- Department of Genetic Medicine, Weill Cornell Medical College, 1300 York Avenue, Box 164, New York, NY, 10065, USA.
| |
Collapse
|
30
|
Lu N, Malemud CJ. Extracellular Signal-Regulated Kinase: A Regulator of Cell Growth, Inflammation, Chondrocyte and Bone Cell Receptor-Mediated Gene Expression. Int J Mol Sci 2019; 20:ijms20153792. [PMID: 31382554 PMCID: PMC6696446 DOI: 10.3390/ijms20153792] [Citation(s) in RCA: 114] [Impact Index Per Article: 19.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2019] [Revised: 07/15/2019] [Accepted: 07/23/2019] [Indexed: 12/17/2022] Open
Abstract
Extracellular signal-regulated kinase (ERK) is a member of the mitogen-activated protein kinase family of signaling molecules. ERK is predominantly found in two forms, ERK1 (p44) and ERK2 (p42), respectively. There are also several atypical forms of ERK, including ERK3, ERK4, ERK5 and ERK7. The ERK1/2 signaling pathway has been implicated in many and diverse cellular events, including proliferation, growth, differentiation, cell migration, cell survival, metabolism and transcription. ERK1/2 is activated (i.e., phosphorylated) in the cytosol and subsequently translocated to the nucleus, where it activates transcription factors including, but not limited to, ETS, c-Jun, and Fos. It is not surprising that the ERK1/2 signaling cascade has been implicated in many pathological conditions, namely, cancer, arthritis, chronic inflammation, and osteoporosis. This narrative review examines many of the cellular events in which the ERK1/2 signaling cascade plays a critical role. It is anticipated that agents designed to inhibit ERK1/2 activation or p-ERK1/2 activity will be developed for the treatment of those diseases characterized by dysregulated gene expression through ERK1/2 activation.
Collapse
Affiliation(s)
- Nathan Lu
- Department of Medicine, Division of Rheumatic Diseases, Case Western Reserve University School of Medicine, University Hospitals Cleveland Medical Center, Cleveland, OH 44106, USA
| | - Charles J Malemud
- Department of Medicine, Division of Rheumatic Diseases, Case Western Reserve University School of Medicine, University Hospitals Cleveland Medical Center, Cleveland, OH 44106, USA.
| |
Collapse
|
31
|
Fan L, Wang Y, Wang W, Wei X. Carcinogenic role of K-Ras-ERK1/2 signaling in bladder cancer via inhibition of H1.2 phosphorylation at T146. J Cell Physiol 2019; 234:21135-21144. [PMID: 31032946 DOI: 10.1002/jcp.28716] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2018] [Revised: 03/29/2019] [Accepted: 04/11/2019] [Indexed: 12/17/2022]
Abstract
It has been reported that Ras-ERK signaling regulated tumor suppressive genes via epigenetic mechanisms. Herein, we set out to investigate the correlation between K-Ras-ERK1/2 signaling and H1.2 phosphorylation, to provide a better understanding of K-Ras-ERK signaling in cancer. A plasmid for expression of mutated K-Ras was transfected into human bladder carcinoma HT1197 cells. Western blot was carried out for testing the expression changes of ERK1/2 and H1.2. The 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyl tetrazolium bromide (MTT) assay, soft-agar colony formation assay, and transwell assay were used to test the effects of H1.2 phosphorylation at T146 (H1.2 T146ph ) on HT1197 cells growth and migration. Quantitative reverse transcription polymerase chain reaction (qRT-PCR) and chromatin immunoprecipitation (ChIP) were performed to test whether H1.2 T146ph regulated K-Ras-ERK1/2 downstream genes. Furthermore, how K-Ras-ERK1/2 regulated H1.2 T146ph expression was studied. We found that the ERK1/2 was activated when K-Ras was mutated, and H1.2 T146ph expression was significantly downregulated by K-Ras mutation. H1.2 T146E for mimicking H1.2 T146ph significantly attenuated K-Ras mutation induced increases in HT1197 cells viability, colony formation, and relative migration. Besides, H1.2 T146ph regulated the transcription of K-Ras-ERK1/2 downstream genes, including NT5E, GDF15, CARD16, CYR61, IGFBP3, and WNT16B. Furthermore, K-Ras-ERK1/2 signaling inhibited H1.2 phosphorylation at T146 through degradation of DNA-PK, and the degraded DNA-PK by K-Ras-ERK1/2 possibly via modulation of MDM2. In conclusion, the activation of K-Ras-ERK1/2 signaling will repress the phosphorylation of H1.2 at T146, and thereby, promoted the growth and migration of bladder cancer cells. K-Ras-ERK1/2 signaling repressed H1.2 phosphorylation possibly by MDM2-mediated degradation of DNA-PK.
Collapse
Affiliation(s)
- Li Fan
- Department of Urology, China-Japan Union Hospital of Jilin University, Changchun, China
| | - Yao Wang
- Department of Urology, China-Japan Union Hospital of Jilin University, Changchun, China
| | - Weihua Wang
- Department of Urology, China-Japan Union Hospital of Jilin University, Changchun, China
| | - Xin Wei
- Department of Urology, China-Japan Union Hospital of Jilin University, Changchun, China
| |
Collapse
|
32
|
Kiss A, Koppel AC, Murphy E, Sall M, Barlas M, Kissling G, Efimova T. Cell Type-Specific p38δ Targeting Reveals a Context-, Stage-, and Sex-Dependent Regulation of Skin Carcinogenesis. Int J Mol Sci 2019; 20:ijms20071532. [PMID: 30934690 PMCID: PMC6479675 DOI: 10.3390/ijms20071532] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2019] [Revised: 03/21/2019] [Accepted: 03/23/2019] [Indexed: 12/14/2022] Open
Abstract
Activation and/or upregulated expression of p38δ are demonstrated in human skin malignancies including cutaneous squamous cell carcinoma, suggesting a role for p38δ in skin carcinogenesis. We previously reported that mice with germline deletion of the p38δ gene are significantly protected from chemical skin carcinogenesis. Here, we investigated the effects of cell-selective targeted ablation of p38δ in keratinocytes and in immune (myeloid) cells on skin tumor development in a two-stage 7,12-dimethylbenz(a)anthracene (DMBA)/12-O-tetradecanoylphorbol-13-acetate (TPA) chemical mouse skin carcinogenesis model. Conditional keratinocyte-specific p38δ ablation (p38δ-cKO∆K) did not influence the latency, incidence, or multiplicity of chemically-induced skin tumors, but led to increased tumor volume in females during the TPA promotion stage, and reduced malignant progression in males and females relative to their wild-type counterparts. In contrast, conditional myeloid cell-specific p38δ deletion (p38δ-cKO∆M) inhibited DMBA/TPA-induced skin tumorigenesis in male but not female mice. Thus, tumor onset was delayed, and tumor incidence, multiplicity, and volume were reduced in p38δ-cKO∆M males compared with control wild-type males. Moreover, the percentage of male mice with malignant tumors was decreased in the p38δ-cKO∆M group relative to their wild-type counterparts. Collectively, these results reveal that cell-specific p38δ targeting modifies susceptibility to chemical skin carcinogenesis in a context-, stage-, and sex-specific manner.
Collapse
Affiliation(s)
- Alexi Kiss
- Department of Anatomy & Cell Biology, The George Washington University School of Medicine and Health Sciences, 2300 I Street NW, Ross Hall 550, Washington, DC 20037, USA.
- The George Washington Cancer Center, 800 22nd Street NW, Science and Engineering Hall 8160, Washington, DC 20052, USA.
| | - Aaron C Koppel
- Department of Anatomy & Cell Biology, The George Washington University School of Medicine and Health Sciences, 2300 I Street NW, Ross Hall 550, Washington, DC 20037, USA.
| | - Emily Murphy
- Department of Anatomy & Cell Biology, The George Washington University School of Medicine and Health Sciences, 2300 I Street NW, Ross Hall 550, Washington, DC 20037, USA.
- Department of Dermatology, The George Washington University School of Medicine and Health Sciences, 2150 Pennsylvania Ave NW, Suite 2B-430, Washington, DC 20037, USA.
- Georgetown University School of Medicine, 3900 Reservoir Rd NW, Washington, DC 20007, USA.
| | - Maxwell Sall
- Department of Anatomy & Cell Biology, The George Washington University School of Medicine and Health Sciences, 2300 I Street NW, Ross Hall 550, Washington, DC 20037, USA.
| | - Meral Barlas
- Department of Anatomy & Cell Biology, The George Washington University School of Medicine and Health Sciences, 2300 I Street NW, Ross Hall 550, Washington, DC 20037, USA.
| | - Grace Kissling
- Biostatistics and Computational Biology Branch, National Institute of Environmental Health Sciences, Research Triangle Park, NC 27709, USA.
| | - Tatiana Efimova
- Department of Anatomy & Cell Biology, The George Washington University School of Medicine and Health Sciences, 2300 I Street NW, Ross Hall 550, Washington, DC 20037, USA.
- The George Washington Cancer Center, 800 22nd Street NW, Science and Engineering Hall 8160, Washington, DC 20052, USA.
- Department of Dermatology, The George Washington University School of Medicine and Health Sciences, 2150 Pennsylvania Ave NW, Suite 2B-430, Washington, DC 20037, USA.
| |
Collapse
|
33
|
Tajan M, Paccoud R, Branka S, Edouard T, Yart A. The RASopathy Family: Consequences of Germline Activation of the RAS/MAPK Pathway. Endocr Rev 2018; 39:676-700. [PMID: 29924299 DOI: 10.1210/er.2017-00232] [Citation(s) in RCA: 146] [Impact Index Per Article: 20.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/24/2017] [Accepted: 06/13/2018] [Indexed: 12/13/2022]
Abstract
Noonan syndrome [NS; Mendelian Inheritance in Men (MIM) #163950] and related syndromes [Noonan syndrome with multiple lentigines (formerly called LEOPARD syndrome; MIM #151100), Noonan-like syndrome with loose anagen hair (MIM #607721), Costello syndrome (MIM #218040), cardio-facio-cutaneous syndrome (MIM #115150), type I neurofibromatosis (MIM #162200), and Legius syndrome (MIM #611431)] are a group of related genetic disorders associated with distinctive facial features, cardiopathies, growth and skeletal abnormalities, developmental delay/mental retardation, and tumor predisposition. NS was clinically described more than 50 years ago, and disease genes have been identified throughout the last 3 decades, providing a molecular basis to better understand their physiopathology and identify targets for therapeutic strategies. Most of these genes encode proteins belonging to or regulating the so-called RAS/MAPK signaling pathway, so these syndromes have been gathered under the name RASopathies. In this review, we provide a clinical overview of RASopathies and an update on their genetics. We then focus on the functional and pathophysiological effects of RASopathy-causing mutations and discuss therapeutic perspectives and future directions.
Collapse
Affiliation(s)
- Mylène Tajan
- INSERM UMR 1048, Institute of Cardiovascular and Metabolic Diseases (I2MC), University of Toulouse Paul Sabatier, Toulouse, France
| | - Romain Paccoud
- INSERM UMR 1048, Institute of Cardiovascular and Metabolic Diseases (I2MC), University of Toulouse Paul Sabatier, Toulouse, France
| | - Sophie Branka
- INSERM UMR 1048, Institute of Cardiovascular and Metabolic Diseases (I2MC), University of Toulouse Paul Sabatier, Toulouse, France
| | - Thomas Edouard
- Endocrine, Bone Diseases, and Genetics Unit, Children's Hospital, Toulouse University Hospital, Toulouse, France
| | - Armelle Yart
- INSERM UMR 1048, Institute of Cardiovascular and Metabolic Diseases (I2MC), University of Toulouse Paul Sabatier, Toulouse, France
| |
Collapse
|
34
|
Schneider T, Martinez-Martinez A, Cubillos-Rojas M, Bartrons R, Ventura F, Rosa JL. The E3 ubiquitin ligase HERC1 controls the ERK signaling pathway targeting C-RAF for degradation. Oncotarget 2018; 9:31531-31548. [PMID: 30140388 PMCID: PMC6101136 DOI: 10.18632/oncotarget.25847] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2017] [Accepted: 07/12/2018] [Indexed: 12/14/2022] Open
Abstract
The RAF/MEK/ERK cascade is a conserved intracellular signaling pathway that controls fundamental cellular processes including growth, proliferation, differentiation, survival and migration. Aberrant regulation of this signaling pathway has long been associated with human cancers. A major point of regulation of this pathway occurs at the level of the serine/threonine protein kinase C-RAF. Here, we show how the E3 ubiquitin ligase HERC1 regulates ERK signaling. HERC1 knockdown induced cellular proliferation, which is associated with an increase in ERK phosphorylation and in C-RAF protein levels. We demonstrate that overexpression of wild-type C-RAF is sufficient to increase ERK phosphorylation. Experiments with pharmacological inhibitors of RAF activity, or with interference RNA, show that the regulation of ERK phosphorylation by HERC1 is RAF-dependent. Immunoprecipitation, pull-down and confocal fluorescence microscopy experiments demonstrate an interaction between HERC1 and C-RAF proteins. Mechanistically, HERC1 controls C-RAF stability by regulating its polyubiquitylation in a lysine 48-linked chain. In vitro ubiquitylation assays indicate that C-RAF is a substrate of the E3 ubiquitin ligase HERC1. Altogether, we show how HERC1 can regulate cell proliferation through the activation of ERK signaling by a mechanism that affects C-RAF’s stability.
Collapse
Affiliation(s)
- Taiane Schneider
- Departament de Ciències Fisiològiques, IDIBELL, Campus Bellvitge, Universitat de Barcelona, L'Hospitalet de Llobregat, Barcelona, Spain
| | - Arturo Martinez-Martinez
- Departament de Ciències Fisiològiques, IDIBELL, Campus Bellvitge, Universitat de Barcelona, L'Hospitalet de Llobregat, Barcelona, Spain
| | - Monica Cubillos-Rojas
- Departament de Ciències Fisiològiques, IDIBELL, Campus Bellvitge, Universitat de Barcelona, L'Hospitalet de Llobregat, Barcelona, Spain
| | - Ramon Bartrons
- Departament de Ciències Fisiològiques, IDIBELL, Campus Bellvitge, Universitat de Barcelona, L'Hospitalet de Llobregat, Barcelona, Spain
| | - Francesc Ventura
- Departament de Ciències Fisiològiques, IDIBELL, Campus Bellvitge, Universitat de Barcelona, L'Hospitalet de Llobregat, Barcelona, Spain
| | - Jose Luis Rosa
- Departament de Ciències Fisiològiques, IDIBELL, Campus Bellvitge, Universitat de Barcelona, L'Hospitalet de Llobregat, Barcelona, Spain
| |
Collapse
|
35
|
Landim BC, de Jesus MM, Bosque BP, Zanon RG, da Silva CV, Góes RM, Ribeiro DL. Stimulating effect of palmitate and insulin on cell migration and proliferation in PNT1A and PC3 prostate cells: Counteracting role of metformin. Prostate 2018; 78:731-742. [PMID: 29635803 DOI: 10.1002/pros.23517] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/09/2017] [Accepted: 03/09/2018] [Indexed: 01/21/2023]
Abstract
BACKGROUND A potential association between obesity and prostate cancer has been proposed. Metformin, an antidiabetes drug, has antiproliferative effects being proposed for cancer treatment. However, under intense proliferative stimulation conditions such as those found in obesity, its efficacy is still uncertain. Thus, we analyzed the effects of saturated fatty acid and/or insulin under high concentrations, with or without metformin, on the proliferation and migration of prostate cells. METHODS Human prostate epithelial cell lines non-tumor (PNT1A) and tumor (PC3) were treated with control media (DMEM, C), palmitate (100 µM, HF), and/or insulin (50 µU, HI) with or without metformin (100 µM) for 24 or 48 h. RESULTS Both PNT1A and PC3 cells had greater proliferation when treated with HF, while HI treatment stimulated only PNT1A. Metformin inhibited cell proliferation caused by HF in both cell lines, but it did not block the proliferative action of HI in PNT1A cells. PNT1A increased cell migration after all treatments, while only HF influenced PC3; metformin inhibited the migration stimulated by all obese microenvironments. Both HF and HI treatments in PNT1A and HF treatment in PC3 augmented vimentin expression, resulting in a higher epithelial-mesenchymal transition (which, in turn, could influence cell migration). Metformin inhibited vimentin expression in both normal and tumor cells. Although HF treatment had increased AMPK activation, it also increased the levels of activated ERK1/2, which could be responsible for high cell proliferation in both cell lines. In contrast, HI decreased AMPK activation in both cell lines, whereas it increased ERK1/2 levels in PNT1A and decreased them in PC3 (reflecting greater cell proliferation only in non-tumor cells). Metformin maintained high activation of AMPK and decreased ERK1/2 levels after HF in both cell lines and only after HI in PNT1A, which was able to decrease the cell proliferation triggered by these treatments. CONCLUSIONS Higher concentrations of palmitate on PC3 cells and palmitate and insulin on PNT1A cells stimulate cellular activities that could favor cancer progression. Metformin inhibited most of these stimuli, showing the efficacy of this drug for cancer adjuvant therapy in obese patients (a group at increased risk for the development of prostrate cancer).
Collapse
Affiliation(s)
- Breno C Landim
- Department of Cell Biology, Histology and Embriology. Institute of Biomedical Sciences-ICBIM. Federal University of Uberlândia, Brazil
| | - Mariana M de Jesus
- Department of Structural and Functional Biology, Institute of Biology, University of Campinas-UNICAMP, Campinas, São Paulo, Brazil
| | - Beatriz P Bosque
- Department of Cell Biology, Histology and Embriology. Institute of Biomedical Sciences-ICBIM. Federal University of Uberlândia, Brazil
| | - Renata G Zanon
- Department of Anatomy, Institute of Biomedical Sciences, Federal University of Uberlândia-UFU, Brazil
| | - Claudio V da Silva
- Department of Immunology, Institute of Biomedical Sciences, Federal University of Uberlândia-UFU, Brazil
| | - Rejane M Góes
- Department of Biology. Institute of Biosciences, Humanities and Exact Sciences, State University of São Paulo-UNESP, Brazil
| | - Daniele L Ribeiro
- Department of Cell Biology, Histology and Embriology. Institute of Biomedical Sciences-ICBIM. Federal University of Uberlândia, Brazil
| |
Collapse
|
36
|
Baumgartner C, Toifl S, Farlik M, Halbritter F, Scheicher R, Fischer I, Sexl V, Bock C, Baccarini M. An ERK-Dependent Feedback Mechanism Prevents Hematopoietic Stem Cell Exhaustion. Cell Stem Cell 2018; 22:879-892.e6. [PMID: 29804890 PMCID: PMC5988582 DOI: 10.1016/j.stem.2018.05.003] [Citation(s) in RCA: 80] [Impact Index Per Article: 11.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2017] [Revised: 03/08/2018] [Accepted: 05/04/2018] [Indexed: 11/22/2022]
Abstract
Hematopoietic stem cells (HSCs) sustain hematopoiesis throughout life. HSCs exit dormancy to restore hemostasis in response to stressful events, such as acute blood loss, and must return to a quiescent state to prevent their exhaustion and resulting bone marrow failure. HSC activation is driven in part through the phosphatidylinositol 3-kinase (PI3K)/AKT/mTORC1 signaling pathway, but less is known about the cell-intrinsic pathways that control HSC dormancy. Here, we delineate an ERK-dependent, rate-limiting feedback mechanism that controls HSC fitness and their re-entry into quiescence. We show that the MEK/ERK and PI3K pathways are synchronously activated in HSCs during emergency hematopoiesis and that feedback phosphorylation of MEK1 by activated ERK counterbalances AKT/mTORC1 activation. Genetic or chemical ablation of this feedback loop tilts the balance between HSC dormancy and activation, increasing differentiated cell output and accelerating HSC exhaustion. These results suggest that MEK inhibitors developed for cancer therapy may find additional utility in controlling HSC activation.
Collapse
Affiliation(s)
- Christian Baumgartner
- Department of Microbiology, Immunobiology and Genetics, Center for Molecular Biology of the University of Vienna, Max F. Perutz Laboratories, Vienna Biocenter (VBC), 1030 Vienna, Austria
| | - Stefanie Toifl
- Department of Microbiology, Immunobiology and Genetics, Center for Molecular Biology of the University of Vienna, Max F. Perutz Laboratories, Vienna Biocenter (VBC), 1030 Vienna, Austria
| | - Matthias Farlik
- CeMM Research Center for Molecular Medicine of the Austrian Academy of Sciences, 1090 Vienna, Austria
| | - Florian Halbritter
- CeMM Research Center for Molecular Medicine of the Austrian Academy of Sciences, 1090 Vienna, Austria
| | - Ruth Scheicher
- Department for Biomedical Sciences, Institute of Pharmacology and Toxicology, University of Veterinary Medicine, 1210 Vienna, Austria
| | - Irmgard Fischer
- Department of Microbiology, Immunobiology and Genetics, Center for Molecular Biology of the University of Vienna, Max F. Perutz Laboratories, Vienna Biocenter (VBC), 1030 Vienna, Austria
| | - Veronika Sexl
- Department for Biomedical Sciences, Institute of Pharmacology and Toxicology, University of Veterinary Medicine, 1210 Vienna, Austria
| | - Christoph Bock
- CeMM Research Center for Molecular Medicine of the Austrian Academy of Sciences, 1090 Vienna, Austria; Department of Laboratory Medicine, Medical University of Vienna, Vienna, Austria; Saarland Informatics Campus, Max Planck Institute for Informatics, Saarbrücken, Germany
| | - Manuela Baccarini
- Department of Microbiology, Immunobiology and Genetics, Center for Molecular Biology of the University of Vienna, Max F. Perutz Laboratories, Vienna Biocenter (VBC), 1030 Vienna, Austria.
| |
Collapse
|
37
|
Abstract
Abnormally activated RAS proteins are the main oncogenic driver that governs the functioning of major signaling pathways involved in the initiation and development of human malignancies. Mutations in RAS genes and or its regulators, most frequent in human cancers, are the main force for incessant RAS activation and associated pathological conditions including cancer. In general, RAS is the main upstream regulator of the highly conserved signaling mechanisms associated with a plethora of important cellular activities vital for normal homeostasis. Mutated or the oncogenic RAS aberrantly activates a web of interconnected signaling pathways including RAF-MEK (mitogen-activated protein kinase kinase)-ERK (extracellular signal-regulated kinase), phosphoinositide-3 kinase (PI3K)/AKT (protein kinase B), protein kinase C (PKC) and ral guanine nucleotide dissociation stimulator (RALGDS), etc., leading to uncontrolled transcriptional expression and reprogramming in the functioning of a range of nuclear and cytosolic effectors critically associated with the hallmarks of carcinogenesis. This review highlights the recent literature on how oncogenic RAS negatively use its signaling web in deregulating the expression and functioning of various effector molecules in the pathogenesis of human malignancies.
Collapse
|
38
|
DA-Raf, a dominant-negative antagonist of the Ras–ERK pathway, is a putative tumor suppressor. Exp Cell Res 2018; 362:111-120. [DOI: 10.1016/j.yexcr.2017.11.008] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2017] [Revised: 11/07/2017] [Accepted: 11/08/2017] [Indexed: 12/30/2022]
|
39
|
Cuenda A, Lizcano JM, Lozano J. Editorial: Mitogen Activated Protein Kinases. Front Cell Dev Biol 2017; 5:80. [PMID: 28959688 PMCID: PMC5604067 DOI: 10.3389/fcell.2017.00080] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2017] [Accepted: 08/30/2017] [Indexed: 12/29/2022] Open
Affiliation(s)
- Ana Cuenda
- Department of Immunology and Oncology, Centro Nacional de Biotecnología (CSIC)Madrid, Spain
| | - José M Lizcano
- Department of Biochemistry and Molecular Biology, Institute of Neurosciences, Faculty of Medicina, Universitat Autonoma de BarcelonaBellaterra, Spain
| | - José Lozano
- Department of Molecular Biology and Biochemistry, Universidad de MálagaMálaga, Spain
| |
Collapse
|