1
|
Jo K, Liu ZY, Patel G, Yu Z, Yao L, Teague S, Johnson C, Spence J, Heemskerk I. Endogenous FGFs drive ERK-dependent cell fate patterning in 2D human gastruloids. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.07.08.602611. [PMID: 39026750 PMCID: PMC11257619 DOI: 10.1101/2024.07.08.602611] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/20/2024]
Abstract
The role of FGF is the least understood of the morphogens driving mammalian gastrulation. Here we investigated the function of FGF in a stem cell model for human gastrulation known as a 2D gastruloid. We found a ring of FGF-dependent ERK activity that closely follows the emergence of primitive streak (PS)-like cells but expands further inward. We showed that this ERK activity pattern is required for PS-like differentiation and that loss of PS-like cells upon FGF receptor inhibition can be rescued by directly activating ERK. We further demonstrated that the ERK-ring depends on localized activation of basally localized FGF receptors (FGFR) by endogenous FGF gradients. We confirm and extend previous studies in analyzing expression of FGF pathway components, showing the main receptor to be FGFR1 and the key ligands FGF2/4/17, similar to the human and monkey embryo but different from the mouse. In situ hybridization and scRNA-seq revealed that FGF4 and FGF17 expression colocalize with the PS marker TBXT but only FGF17 is maintained in nascent mesoderm and endoderm. FGF4 and FGF17 reduction both reduced ERK activity and differentiation to PS-like cells and their derivatives, indicating overlapping function. Thus, we have identified a previously unknown role for FGF-dependent ERK signaling in 2D gastruloids and possibly the human embryo, driven by a mechanism where FGF4 and FGF17 signal through basally localized FGFR1 to induce PS-like cells.
Collapse
Affiliation(s)
- Kyoung Jo
- Department of Cell and Developmental Biology, University of Michigan Medical School, Ann Arbor, Michigan
| | - Zong-Yuan Liu
- Department of Cell and Developmental Biology, University of Michigan Medical School, Ann Arbor, Michigan
| | - Gauri Patel
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, Michigan
| | - Zhiyuan Yu
- Department of Computational Medicine and Bioinformatics, University of Michigan Medical School, Ann Arbor, Michigan
| | - LiAng Yao
- Department of Cell and Developmental Biology, University of Michigan Medical School, Ann Arbor, Michigan
| | - Seth Teague
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, Michigan
| | - Craig Johnson
- Department of Cell and Developmental Biology, University of Michigan Medical School, Ann Arbor, Michigan
| | - Jason Spence
- Department of Cell and Developmental Biology, University of Michigan Medical School, Ann Arbor, Michigan
- Center for Cell Plasticity and Organ Design, University of Michigan Medical School, Ann Arbor, Michigan
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, Michigan
- Department of Internal Medicine, Gastroenterology, University of Michigan Medical School, Ann Arbor, Michigan
| | - Idse Heemskerk
- Department of Cell and Developmental Biology, University of Michigan Medical School, Ann Arbor, Michigan
- Department of Computational Medicine and Bioinformatics, University of Michigan Medical School, Ann Arbor, Michigan
- Center for Cell Plasticity and Organ Design, University of Michigan Medical School, Ann Arbor, Michigan
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, Michigan
- Department of Physics, University of Michigan, Ann Arbor, Michigan
| |
Collapse
|
2
|
Zhang Z, Wu Y, Fu J, Yu X, Su Y, Jia S, Cheng H, Shen Y, He X, Ren K, Zheng X, Guan H, Rao F, Zhao L. Proteostatic reactivation of the developmental transcription factor TBX3 drives BRAF/MAPK-mediated tumorigenesis. Nat Commun 2024; 15:4108. [PMID: 38750011 PMCID: PMC11096176 DOI: 10.1038/s41467-024-48173-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2023] [Accepted: 04/22/2024] [Indexed: 05/18/2024] Open
Abstract
MAPK pathway-driven tumorigenesis, often induced by BRAFV600E, relies on epithelial dedifferentiation. However, how lineage differentiation events are reprogrammed remains unexplored. Here, we demonstrate that proteostatic reactivation of developmental factor, TBX3, accounts for BRAF/MAPK-mediated dedifferentiation and tumorigenesis. During embryonic development, BRAF/MAPK upregulates USP15 to stabilize TBX3, which orchestrates organogenesis by restraining differentiation. The USP15-TBX3 axis is reactivated during tumorigenesis, and Usp15 knockout prohibits BRAFV600E-driven tumor development in a Tbx3-dependent manner. Deleting Tbx3 or Usp15 leads to tumor redifferentiation, which parallels their overdifferentiation tendency during development, exemplified by disrupted thyroid folliculogenesis and elevated differentiation factors such as Tpo, Nis, Tg. The clinical relevance is highlighted in that both USP15 and TBX3 highly correlates with BRAFV600E signature and poor tumor prognosis. Thus, USP15 stabilized TBX3 represents a critical proteostatic mechanism downstream of BRAF/MAPK-directed developmental homeostasis and pathological transformation, supporting that tumorigenesis largely relies on epithelial dedifferentiation achieved via embryonic regulatory program reinitiation.
Collapse
Affiliation(s)
- Zhenlei Zhang
- Department of Thyroid and Neck Oncology, Key Laboratory of Cancer Prevention and Therapy, Tianjin's Clinical Research Center for Cancer, National Clinical Research Center for Cancer, The Province and Ministry Co-sponsored Collaborative Innovation Center for Medical Epigenetics, Key Laboratory of Immune Microenvironment and Disease (Ministry of Education), Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Tianjin Medical University Cancer Institute and Hospital, Tianjin Medical University, Tianjin, China
| | - Yufan Wu
- Department of Thyroid and Neck Oncology, Key Laboratory of Cancer Prevention and Therapy, Tianjin's Clinical Research Center for Cancer, National Clinical Research Center for Cancer, The Province and Ministry Co-sponsored Collaborative Innovation Center for Medical Epigenetics, Key Laboratory of Immune Microenvironment and Disease (Ministry of Education), Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Tianjin Medical University Cancer Institute and Hospital, Tianjin Medical University, Tianjin, China
| | - Jinrong Fu
- Department of Endocrinology, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, Guangdong, China
| | - Xiujie Yu
- Department of Pathology, Tianjin Central Hospital of Gynecology and Obstetrics, Tianjin, China
| | - Yang Su
- School of Life Sciences, Southern University of Science and Technology, Shenzhen, Guangdong, China
| | - Shikai Jia
- Department of Thyroid and Neck Oncology, Key Laboratory of Cancer Prevention and Therapy, Tianjin's Clinical Research Center for Cancer, National Clinical Research Center for Cancer, The Province and Ministry Co-sponsored Collaborative Innovation Center for Medical Epigenetics, Key Laboratory of Immune Microenvironment and Disease (Ministry of Education), Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Tianjin Medical University Cancer Institute and Hospital, Tianjin Medical University, Tianjin, China
| | - Huili Cheng
- Department of Thyroid and Neck Oncology, Key Laboratory of Cancer Prevention and Therapy, Tianjin's Clinical Research Center for Cancer, National Clinical Research Center for Cancer, The Province and Ministry Co-sponsored Collaborative Innovation Center for Medical Epigenetics, Key Laboratory of Immune Microenvironment and Disease (Ministry of Education), Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Tianjin Medical University Cancer Institute and Hospital, Tianjin Medical University, Tianjin, China
| | - Yan Shen
- Department of Pathology, Tianjin Central Hospital of Gynecology and Obstetrics, Tianjin, China
| | - Xianghui He
- Department of General Surgery, Tianjin Medical University General Hospital, Tianjin Medical University, Tianjin, China
| | - Kai Ren
- Department of Radiation Oncology, Key Laboratory of Cancer Prevention and Therapy, Tianjin's Clinical Research Center for Cancer, National Clinical Research Center for Cancer, Tianjin Medical University Cancer Institute and Hospital, Tianjin, China
| | - Xiangqian Zheng
- Department of Thyroid and Neck Tumor, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin's Clinical Research Center for Cancer, Tianjin, China
| | - Haixia Guan
- Department of Endocrinology, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, Guangdong, China.
| | - Feng Rao
- School of Life Sciences, Southern University of Science and Technology, Shenzhen, Guangdong, China.
| | - Li Zhao
- Department of Thyroid and Neck Tumor, Key Laboratory of Cancer Prevention and Therapy, Tianjin's Clinical Research Center for Cancer, National Clinical Research Center for Cancer, The Province and Ministry Co-sponsored Collaborative Innovation Center for Medical Epigenetics, Key Laboratory of Immune Microenvironment and Disease (Ministry of Education), Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Tianjin Medical University Cancer Institute and Hospital, Tianjin Medical University, Tianjin, China.
| |
Collapse
|
3
|
Peng J, Zhang S, Han F, Wang Z. C1QBP is a critical component in the immune response of large yellow croaker (Larimichthys crocea) against visceral white spot disease caused by Pseudomonas plecoglossicida. FISH & SHELLFISH IMMUNOLOGY 2024; 146:109372. [PMID: 38218420 DOI: 10.1016/j.fsi.2024.109372] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/10/2023] [Revised: 12/27/2023] [Accepted: 01/10/2024] [Indexed: 01/15/2024]
Abstract
The large yellow croaker (Larimichthys crocea) stands as a cornerstone of mariculture in China due to its significant value. However, the threat of Pseudomonas plecoglossicida infection looms large, capable of triggering "visceral white spot disease" and subsequently inflicting severe economic ramifications. Through a prior genome-wide association analysis (GWAS) aimed at understanding the resistance of the large yellow croaker to this ailment, a pivotal player emerged: the complement component 1q binding protein, aptly named LcC1qbp. This protein assumes a crucial role in the activation of the complement system. This study delves deeper into the immune response by examining the expression patterns of LcC1QBP when confronted with P. plecoglossicida. The investigation into gene expression patterns reveals LcC1qbp's widespread presence, with its highest transcriptional abundance identified in the kidney tissues. Upon infection by P. plecoglossicida, the up-regulation of LcC1qbp in major immune organs manifests at both the transcriptional and translational levels. In the context of RNA interference, transcriptome analysis of C1qbp in HEK 293T cells uncovers 1327 differentially expressed genes (DEGs), featuring 41 significant immune genes. This includes pivotal components such as C1S and C3 of the complement system, along with IL11, IL12RB2, and Myd88, among others. The outcomes of enrichment analysis spotlight the prevalence of DEGs within key pathways like immune system development, myeloid leukocyte-mediated immunity, MAPK signaling, and other immune-related routes. By unveiling the immune response mechanisms of the large yellow croaker to P. plecoglossicida infection, this study bolsters our understanding. Furthermore, it lays the groundwork for pursuing effective strategies in both preventing and treating "visceral white spot disease" in the large yellow croaker.
Collapse
Affiliation(s)
- Jia Peng
- State Key Laboratory of Mariculture Breeding, Fujian Provincial Key Laboratory of Marine Fishery Resources and Eco-environment, Fisheries College, Jimei University, Xiamen, 361000, PR China
| | - Sen Zhang
- State Key Laboratory of Mariculture Breeding, Fujian Provincial Key Laboratory of Marine Fishery Resources and Eco-environment, Fisheries College, Jimei University, Xiamen, 361000, PR China
| | - Fang Han
- State Key Laboratory of Mariculture Breeding, Fujian Provincial Key Laboratory of Marine Fishery Resources and Eco-environment, Fisheries College, Jimei University, Xiamen, 361000, PR China.
| | - Zhiyong Wang
- State Key Laboratory of Mariculture Breeding, Fujian Provincial Key Laboratory of Marine Fishery Resources and Eco-environment, Fisheries College, Jimei University, Xiamen, 361000, PR China
| |
Collapse
|
4
|
Lee J, Mohammad N, Lu Y, Oshins R, Aranyos A, Brantly M. Alpha-defensins inhibit ERK/STAT3 signaling during monocyte-macrophage differentiation and impede macrophage function. Respir Res 2023; 24:309. [PMID: 38082274 PMCID: PMC10714504 DOI: 10.1186/s12931-023-02605-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2023] [Accepted: 11/13/2023] [Indexed: 12/18/2023] Open
Abstract
Alpha-1-antitrypsin deficiency (AATD) is a genetic disorder associated with a 5-tenfold decrease in lung levels of alpha-1-antitrypsin (AAT) and an increased risk for obstructive lung disease. α-defensins are cationic broad-spectrum cytotoxic and pro-inflammatory peptides found in the azurophilic granules of neutrophils. The concentration of α-defensins is less than 30 nM in the bronchoalveolar lavage fluid of healthy controls but is up to 6 μM in AATD individuals with significant lung function impairment. Alveolar macrophages are generally classified into pro-inflammatory (M1) or anti-inflammatory (M2) subsets that play distinct roles in the initiation and resolution of inflammation. Therefore, monocyte-macrophage differentiation should be tightly controlled to maintain lung integrity. In this study, we determined the effect of α-defensins on monocyte-macrophage differentiation and identified the molecular mechanism of this effect. The results of this study demonstrate that 2.5 μM of α-defensins inhibit the phosphorylation of ERK1/2 and STAT3 and suppress the expression of M2 macrophage markers, CD163 and CD206. In addition, a scratch assay shows that the high concentration of α-defensins inhibits cell movement by ~ 50%, and the phagocytosis assay using flow cytometry shows that α-defensins significantly reduce the bacterial phagocytosis rate of monocyte-derived macrophages (MDMs). To examine whether exogenous AAT is able to alleviate the inhibitory effect of α-defensins on macrophage function, we incubated MDMs with AAT prior to α-defensin treatment and demonstrate that AAT improves the migratory ability and phagocytic ability of MDMs compared with MDMs incubated only with α-defensins. Taken together, this study suggests that a high concentration of α-defensins inhibits the activation of ERK/STAT3 signaling, negatively regulates the expression of M2 macrophage markers, and impairs innate immune function of macrophages.
Collapse
Affiliation(s)
- Jungnam Lee
- Division of Pulmonary, Critical Care and Sleep Medicine, University of Florida, Gainesville, FL, USA
| | - Naweed Mohammad
- Division of Pulmonary, Critical Care and Sleep Medicine, University of Florida, Gainesville, FL, USA
| | - Yuanqing Lu
- Division of Pulmonary, Critical Care and Sleep Medicine, University of Florida, Gainesville, FL, USA
| | - Regina Oshins
- Division of Pulmonary, Critical Care and Sleep Medicine, University of Florida, Gainesville, FL, USA
| | - Alek Aranyos
- Division of Pulmonary, Critical Care and Sleep Medicine, University of Florida, Gainesville, FL, USA
| | - Mark Brantly
- Division of Pulmonary, Critical Care and Sleep Medicine, University of Florida, Gainesville, FL, USA.
| |
Collapse
|
5
|
Kim C. Extracellular Signal-Regulated Kinases Play Essential but Contrasting Roles in Osteoclast Differentiation. Int J Mol Sci 2023; 24:15342. [PMID: 37895023 PMCID: PMC10607827 DOI: 10.3390/ijms242015342] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2023] [Revised: 10/16/2023] [Accepted: 10/17/2023] [Indexed: 10/29/2023] Open
Abstract
Bone homeostasis is regulated by the balanced actions of osteoblasts that form the bone and osteoclasts (OCs) that resorb the bone. Bone-resorbing OCs are differentiated from hematopoietic monocyte/macrophage lineage cells, whereas osteoblasts are derived from mesenchymal progenitors. OC differentiation is induced by two key cytokines, macrophage colony-stimulating factor (M-CSF), a factor essential for the proliferation and survival of the OCs, and receptor activator of nuclear factor kappa-B ligand (RANKL), a factor for responsible for the differentiation of the OCs. Mitogen-activated protein kinases (MAPKs), including extracellular signal-regulated kinases (ERKs), p38, and c-Jun N-terminal kinases, play an essential role in regulating the proliferation, differentiation, and function of OCs. ERKs have been known to play a critical role in the differentiation and activation of OCs. In most cases, ERKs positively regulate OC differentiation and function. However, several reports present conflicting conclusions. Interestingly, the inhibition of OC differentiation by ERK1/2 is observed only in OCs differentiated from RAW 264.7 cells. Therefore, in this review, we summarize the current understanding of the conflicting actions of ERK1/2 in OC differentiation.
Collapse
Affiliation(s)
- Chaekyun Kim
- BK21 Program in Biomedical Science & Engineering, Laboratory for Leukocyte Signaling Research, Department of Pharmacology, College of Medicine, Inha University, Incheon 22212, Republic of Korea
| |
Collapse
|
6
|
Yoo DH, Im YS, Oh JY, Gil D, Kim YO. DUSP6 is a memory retention feedback regulator of ERK signaling for cellular resilience of human pluripotent stem cells in response to dissociation. Sci Rep 2023; 13:5683. [PMID: 37029196 PMCID: PMC10082014 DOI: 10.1038/s41598-023-32567-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2022] [Accepted: 03/29/2023] [Indexed: 04/09/2023] Open
Abstract
Cultured human pluripotent stem cells (hPSCs) grow as colonies that require breakdown into small clumps for further propagation. Although cell death mechanism by single-cell dissociation of hPSCs has been well defined, how hPSCs respond to the deadly stimulus and recover the original status remains unclear. Here we show that dissociation of hPSCs immediately activates ERK, which subsequently activates RSK and induces DUSP6, an ERK-specific phosphatase. Although the activation is transient, DUSP6 expression persists days after passaging. DUSP6 depletion using the CRISPR/Cas9 system reveals that DUSP6 suppresses the ERK activity over the long term. Elevated ERK activity by DUSP6 depletion increases both viability of hPSCs after single-cell dissociation and differentiation propensity towards mesoderm and endoderm lineages. These findings provide new insights into how hPSCs respond to dissociation in order to maintain pluripotency.
Collapse
Affiliation(s)
- Dae Hoon Yoo
- Division of Intractable Disease Research, Korea National Institute of Health, Osong, Cheongju, 28160, Republic of Korea
| | - Young Sam Im
- Division of Intractable Disease Research, Korea National Institute of Health, Osong, Cheongju, 28160, Republic of Korea
| | - Ji Young Oh
- Division of Intractable Disease Research, Korea National Institute of Health, Osong, Cheongju, 28160, Republic of Korea
| | - Dayeon Gil
- Division of Intractable Disease Research, Korea National Institute of Health, Osong, Cheongju, 28160, Republic of Korea
| | - Yong-Ou Kim
- Division of Intractable Disease Research, Korea National Institute of Health, Osong, Cheongju, 28160, Republic of Korea.
- Center for National Stem Cell and Regenerative Medicine 202, Osongsaengmyung 2-Ro, Heundeok-Gu, Cheongju, Chungcheongbuk-Do, 28160, Republic of Korea.
| |
Collapse
|
7
|
Erdenebaatar P, Gunarta IK, Suzuki R, Odongoo R, Fujii T, Fukunaga R, Kanemaki MT, Yoshioka K. Redundant roles of extra-cellular signal-regulated kinase (ERK) 1 and 2 in the G1-S transition and etoposide-induced G2/M checkpoint in HCT116 cells. Drug Discov Ther 2023; 17:10-17. [PMID: 36642508 DOI: 10.5582/ddt.2022.01120] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/15/2023]
Abstract
The extracellular signal-regulated kinase (ERK) 1 and 2 intracellular signaling pathways play key roles in a variety of cellular processes, such as proliferation and differentiation. Dysregulation of ERK1/2 signaling has been implicated in many diseases, including cancer. Although ERK1/2 signaling pathways have been extensively studied, controversy remains as to whether ERK1 and ERK2 have specific or redundant functions. In this study, we examined the functional roles of ERK1 and ERK2 in cell proliferation and cell cycle progression using an auxin-inducible degron system combined with gene knockout technology. We found that ERK1/2 double depletion, but not ERK1 or ERK2 depletion, substantially inhibited the proliferation of HCT116 cells during G1-S transition. We further demonstrated that ERK1/2-double-depleted cells were much more tolerant to etoposide-induced G2/M arrest than ERK1 or ERK2 single-knockout cells. Together, these results strongly suggest the functional redundancy of ERK1 and ERK2 in both the G1-S transition under physiological conditions and the DNA damage-induced G2/M checkpoint. Our findings substantially advance understanding of the ERK1/2 pathways, which could have strong implications for future pharmacological developments.
Collapse
Affiliation(s)
- Purev Erdenebaatar
- Division of Molecular Cell Signaling, Cancer Research Institute, Kanazawa University, Kanazawa, Japan
| | - I Ketut Gunarta
- Division of Molecular Cell Signaling, Cancer Research Institute, Kanazawa University, Kanazawa, Japan
| | - Ryusuke Suzuki
- Division of Molecular Cell Signaling, Cancer Research Institute, Kanazawa University, Kanazawa, Japan
| | - Ravdandorj Odongoo
- Division of Molecular Cell Signaling, Cancer Research Institute, Kanazawa University, Kanazawa, Japan
| | - Toshihiro Fujii
- Department of Biochemistry, Faculty of Pharmacy, Osaka Medical and Pharmaceutical University, Takatsuki, Japan
| | - Rikiro Fukunaga
- Department of Biochemistry, Faculty of Pharmacy, Osaka Medical and Pharmaceutical University, Takatsuki, Japan
| | - Masato T Kanemaki
- Department of Chromosome Science, National Institute of Genetics, Research Organization of Information and Systems (ROIS), Mishima, Japan.,Department of Genetics, The Graduate University for Advanced Studies (SOKENDAI), Mishima, Japan
| | - Katsuji Yoshioka
- Division of Molecular Cell Signaling, Cancer Research Institute, Kanazawa University, Kanazawa, Japan
| |
Collapse
|
8
|
Lin Y, Lin F, Zhang Z, Peng L, Yang W, Yang M, Luo B, Wu T, Li D, Li X, Ran B, Anuchapreeda S, Chaiwongsa R, Khamphikham P, Duangmano S, Xu J, He T, Pornprasert S. The FGFR1 Signaling Pathway Upregulates the Oncogenic Transcription Factor FOXQ1 to Promote Breast Cancer Cell Growth. Int J Biol Sci 2023; 19:744-759. [PMID: 36778115 PMCID: PMC9909991 DOI: 10.7150/ijbs.74574] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2022] [Accepted: 12/22/2022] [Indexed: 01/11/2023] Open
Abstract
FGFR1 is a receptor tyrosine kinase deregulated in certain breast cancers (BCs) with a poor prognosis. Although FGFR1-activated phosphorylation cascades have been mapped, the key genes regulated by FGFR1 in BC are largely unclear. FOXQ1 is an oncogenic transcription factor. Although we found that activation of FGFR1 robustly upregulated FOXQ1 mRNA, how FGFR1 regulates FOXQ1 gene expression and whether FOXQ1 is essential for FGFR1-stimulated cell proliferation are unknown. Herein, we confirmed that activation of FGFR1 robustly upregulated FOXQ1 mRNA and protein in BC cells. Knockdown of FOXQ1 blocked the FGFR1 signaling-stimulated BC cell proliferation, colony formation, and xenograft tumor growth. Inhibition of MEK or ERK1/2 activities, or knockout of ERK2 but not ERK1 suppressed the FGFR1 signaling-promoted FOXQ1 gene expression. Inhibition of ERK2 in ERK1 knockout cells blocked, while ectopic expression of FOXQ1 in ERK2 knockout cells rescued the FGFR1-signaling-promoted cell growth. Mechanistically, c-FOS, an early response transcription factor upregulated by the FGFR1-MEK-ERK2 pathway, bound to the FOXQ1 promoter to mediate the FGFR1 signaling-promoted FOXQ1 expression. These results indicate that the FGFR1-ERK2-c-FOS-FOXQ1 regulatory axis plays an essential role in the FGFR1 signaling-promoted BC growth. Targeting ERK2 and FOXQ1 should block BC growth caused by a deregulated FGFR1 signaling.
Collapse
Affiliation(s)
- Yan Lin
- Institute for Cancer Medicine and School of Basic Medical Sciences, Southwest Medical University, Luzhou, Sichuan, 646000, China.,Department of Medical Technology, Faculty of Associated Medical Sciences, Chiang Mai University, Chiang Mai, 50200, Thailand
| | - Fengkang Lin
- Institute for Cancer Medicine and School of Basic Medical Sciences, Southwest Medical University, Luzhou, Sichuan, 646000, China.,Department of Physiology, School of Basic Medical Sciences, Southwest Medical University, Luzhou Sichuan, 646000, China
| | - Zhuoran Zhang
- Institute for Cancer Medicine and School of Basic Medical Sciences, Southwest Medical University, Luzhou, Sichuan, 646000, China
| | - Lijia Peng
- Institute for Cancer Medicine and School of Basic Medical Sciences, Southwest Medical University, Luzhou, Sichuan, 646000, China
| | - Wenli Yang
- Institute for Cancer Medicine and School of Basic Medical Sciences, Southwest Medical University, Luzhou, Sichuan, 646000, China
| | - Mao Yang
- Institute for Cancer Medicine and School of Basic Medical Sciences, Southwest Medical University, Luzhou, Sichuan, 646000, China
| | - Bo Luo
- Institute for Cancer Medicine and School of Basic Medical Sciences, Southwest Medical University, Luzhou, Sichuan, 646000, China
| | - Ting Wu
- Institute for Cancer Medicine and School of Basic Medical Sciences, Southwest Medical University, Luzhou, Sichuan, 646000, China
| | - Dabing Li
- Institute for Cancer Medicine and School of Basic Medical Sciences, Southwest Medical University, Luzhou, Sichuan, 646000, China
| | - Xuesen Li
- Institute for Cancer Medicine and School of Basic Medical Sciences, Southwest Medical University, Luzhou, Sichuan, 646000, China
| | - Bing Ran
- Functional laboratory, School of Basic Medical Sciences, Southwest Medical University, Luzhou Sichuan, 646000, China
| | - Songyot Anuchapreeda
- Department of Medical Technology, Faculty of Associated Medical Sciences, Chiang Mai University, Chiang Mai, 50200, Thailand
| | - Rujirek Chaiwongsa
- Department of Medical Technology, Faculty of Associated Medical Sciences, Chiang Mai University, Chiang Mai, 50200, Thailand
| | - Pinyaphat Khamphikham
- Department of Medical Technology, Faculty of Associated Medical Sciences, Chiang Mai University, Chiang Mai, 50200, Thailand
| | - Suwit Duangmano
- Department of Medical Technology, Faculty of Associated Medical Sciences, Chiang Mai University, Chiang Mai, 50200, Thailand
| | - Jianming Xu
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, Texas 77030, USA
| | - Tao He
- Institute for Cancer Medicine and School of Basic Medical Sciences, Southwest Medical University, Luzhou, Sichuan, 646000, China
| | - Sakorn Pornprasert
- Department of Medical Technology, Faculty of Associated Medical Sciences, Chiang Mai University, Chiang Mai, 50200, Thailand
| |
Collapse
|
9
|
Liu T, Zhu X, Huang C, Chen J, Shu S, Chen G, Xu Y, Hu Y. ERK inhibition reduces neuronal death and ameliorates inflammatory responses in forebrain-specific Ppp2cα knockout mice. FASEB J 2022; 36:e22515. [PMID: 35997299 DOI: 10.1096/fj.202200293r] [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: 02/25/2022] [Revised: 07/24/2022] [Accepted: 08/12/2022] [Indexed: 11/11/2022]
Abstract
It has been shown that PP2A is critical for apoptosis in neural progenitor cells. However, it remains unknown whether PP2A is required for neuronal survival. To address this question, we generated forebrain-specific Ppp2cα knockout (KO) mice. We show that Ppp2cα KO mice display robust neuronal apoptosis and inflammatory responses in the postnatal cortex. Previous evidence has revealed that PD98059 is a potent ERK inhibitor and may protect the brain against cell death after cardiac arrest. To study whether PD98059 may have any effects on Ppp2cα KO mice, the latter was treated with this inhibitor. We demonstrated that the total number of cleaved caspase3 positive (+) cells in the cortex was significantly reduced in Ppp2cα KO mice treated with PD98059 compared with those without PD98059 treatment. We observed that the total number of IBA1+ cells in the cortex was significantly decreased in Ppp2cα KO mice treated with PD98059. Mechanistic analysis reveals that deletion of PP2Aca causes DNA damage, which may be attenuated by PD98059. Together, this study suggests that inhibition of ERK may be an effective strategy to reduce cell death in brain diseases with abnormal neuronal apoptosis.
Collapse
Affiliation(s)
- Tingting Liu
- Department of Neurology, Drum Tower Hospital, Medical School and The State Key Laboratory of Pharmaceutical Biotechnology, Institute of Brain Science, Nanjing University, Nanjing, China
| | - Xiaolei Zhu
- Department of Neurology, Drum Tower Hospital, Medical School and The State Key Laboratory of Pharmaceutical Biotechnology, Institute of Brain Science, Nanjing University, Nanjing, China
| | - Chaoli Huang
- State Key Laboratory of Pharmaceutical Biotechnology, MOE Key Laboratory of Model Animal for Disease Study, Model Animal Research Center, Nanjing University, Nanjing, China
| | - Jiang Chen
- Department of Neurology, Drum Tower Hospital, Medical School and The State Key Laboratory of Pharmaceutical Biotechnology, Institute of Brain Science, Nanjing University, Nanjing, China
| | - Shu Shu
- Department of Neurology, Drum Tower Hospital, Medical School and The State Key Laboratory of Pharmaceutical Biotechnology, Institute of Brain Science, Nanjing University, Nanjing, China
| | - Guiquan Chen
- State Key Laboratory of Pharmaceutical Biotechnology, MOE Key Laboratory of Model Animal for Disease Study, Model Animal Research Center, Nanjing University, Nanjing, China
| | - Yun Xu
- Department of Neurology, Drum Tower Hospital, Medical School and The State Key Laboratory of Pharmaceutical Biotechnology, Institute of Brain Science, Nanjing University, Nanjing, China
| | - Yimin Hu
- Department of Anesthesiology, The Affiliated Changzhou No. 2 People's Hospital of Nanjing Medical University, Changzhou, China
| |
Collapse
|
10
|
Zhang Y, Truong B, Fahl SP, Martinez E, Cai KQ, Al-Saleem ED, Gong Y, Liebermann DA, Soboloff J, Dunbrack R, Levine RL, Fletcher S, Kappes D, Sykes SM, Shapiro P, Wiest DL. The ERK2-DBP domain opposes pathogenesis of a mouse JAK2V617F-driven myeloproliferative neoplasm. Blood 2022; 140:359-373. [PMID: 35436326 PMCID: PMC9335498 DOI: 10.1182/blood.2021013068] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2021] [Accepted: 03/30/2022] [Indexed: 01/18/2023] Open
Abstract
Although Ras/mitogen-activated protein kinase (MAPK) signaling is activated in most human cancers, attempts to target this pathway using kinase-active site inhibitors have not typically led to durable clinical benefit. To address this shortcoming, we sought to test the feasibility of an alternative targeting strategy, focused on the ERK2 substrate binding domains, D and DEF binding pocket (DBP). Disabling the ERK2-DBP domain in mice caused baseline erythrocytosis. Consequently, we investigated the role of the ERK2-D and -DBP domains in disease, using a JAK2-dependent model of polycythemia vera (PV). Of note, inactivation of the ERK2-DBP domain promoted the progression of disease from PV to myelofibrosis, suggesting that the ERK2-DBP domain normally opposes progression. ERK2-DBP inactivation also prevented oncogenic JAK2 kinase (JAK2V617F) from promoting oncogene-induced senescence in vitro. The ERK2-DBP mutation attenuated JAK2-mediated oncogene-induced senescence by preventing the physical interaction of ERK2 with the transcription factor Egr1. Because inactivation of the ERK2-DBP created a functional ERK2 kinase limited to binding substrates through its D domain, these data suggested that the D domain substrates were responsible for promoting oncogene-induced progenitor growth and tumor progression and that pharmacologic targeting of the ERK2-D domain may attenuate cancer cell growth. Indeed, pharmacologic agents targeting the ERK2-D domain were effective in attenuating the growth of JAK2-dependent myeloproliferative neoplasm cell lines. Taken together, these data indicate that the ERK-D and -DBP domains can play distinct roles in the progression of neoplasms and that the D domain has the potential to be a potent therapeutic target in Ras/MAPK-dependent cancers.
Collapse
Affiliation(s)
- Yong Zhang
- Blood Cell Development and Function Program
| | | | | | | | | | | | - Yulan Gong
- Department of Pathology, Fox Chase Cancer Center, Philadelphia, PA
| | - Dan A Liebermann
- Fels Institute for Personalized Medicine and Molecular Biology, Lewis Katz School of Medicine, Temple University, Philadelphia, PA
| | - Jonathan Soboloff
- Fels Institute for Personalized Medicine and Molecular Biology, Lewis Katz School of Medicine, Temple University, Philadelphia, PA
| | - Roland Dunbrack
- Molecular Therapeutics Program, Fox Chase Cancer Center, Philadelphia, PA
| | - Ross L Levine
- Department of Medicine, Leukemia Service, Center for Hematologic Malignancies, Human Oncology and Pathogenesis Program, Memorial Sloan Kettering Cancer Center, New York, NY; and
| | - Steven Fletcher
- Department of Pharmaceutical Sciences, School of Pharmacy, University of Maryland, Baltimore, MD
| | | | | | - Paul Shapiro
- Department of Pharmaceutical Sciences, School of Pharmacy, University of Maryland, Baltimore, MD
| | | |
Collapse
|
11
|
McFann SE, Shvartsman SY, Toettcher JE. Putting in the Erk: Growth factor signaling and mesoderm morphogenesis. Curr Top Dev Biol 2022; 149:263-310. [PMID: 35606058 DOI: 10.1016/bs.ctdb.2022.02.007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
It has long been known that FGF signaling contributes to mesoderm formation, a germ layer found in triploblasts that is composed of highly migratory cells that give rise to muscles and to the skeletal structures of vertebrates. FGF signaling activates several pathways in the developing mesoderm, including transient activation of the Erk pathway, which triggers mesodermal fate specification through the induction of the gene brachyury and activates morphogenetic programs that allow mesodermal cells to position themselves in the embryo. In this review, we discuss what is known about the generation and interpretation of transient Erk signaling in mesodermal tissues across species. We focus specifically on mechanisms that translate the level and duration of Erk signaling into cell fate and cell movement instructions and discuss strategies for further interrogating the role that Erk signaling dynamics play in mesodermal gastrulation and morphogenesis.
Collapse
Affiliation(s)
- Sarah E McFann
- Department of Chemical and Biological Engineering, Princeton University, Princeton, NJ, United States; Lewis Sigler Institute for Integrative Genomics, Princeton University, Princeton, NJ, United States
| | - Stanislav Y Shvartsman
- Lewis Sigler Institute for Integrative Genomics, Princeton University, Princeton, NJ, United States; Department of Molecular Biology, Princeton University, Princeton, NJ, United States; Center for Computational Biology, Flatiron Institute, Simons Foundation, New York, NY, United States
| | - Jared E Toettcher
- Department of Molecular Biology, Princeton University, Princeton, NJ, United States.
| |
Collapse
|
12
|
Cingolani F, Liu Y, Shen Y, Wen J, Farris AB, Czaja MJ. Redundant Functions of ERK1 and ERK2 Maintain Mouse Liver Homeostasis Through Down-Regulation of Bile Acid Synthesis. Hepatol Commun 2022; 6:980-994. [PMID: 34936222 PMCID: PMC9035584 DOI: 10.1002/hep4.1867] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/24/2021] [Revised: 10/22/2021] [Accepted: 11/03/2021] [Indexed: 11/07/2022] Open
Abstract
Activation of extracellular signal-regulated kinase (ERK) 1/2 promotes hepatocyte proliferation in response to growth stimuli, but whether constitutive hepatocyte ERK1/2 signaling functions in liver physiology is unknown. To examine the role of ERK1/2 in hepatic homeostasis, the effects of a knockout of Erk1 and/or Erk2 in mouse liver were examined. The livers of mice with a global Erk1 knockout or a tamoxifen-inducible, hepatocyte-specific Erk2 knockout were normal. In contrast, Erk1/2 double-knockout mice developed hepatomegaly and hepatitis by serum transaminases, histology, terminal deoxynucleotide transferase-mediated deoxyuridine triphosphate nick end-labeling, and assays of hepatic inflammation. Liver injury was associated with biochemical evidence of cholestasis with increased serum and hepatic bile acids and led to hepatic fibrosis and mortality. RNA sequencing and polymerase chain reaction analysis of double-knockout mouse livers revealed that the rate-limiting bile acid synthesis gene Cyp7a1 (cholesterol 7α-hydroxylase) was up-regulated in concert with decreased expression of the transcriptional repressor short heterodimer partner. Elevated bile acids were the mechanism of liver injury, as bile acid reduction by SC-435, an inhibitor of the ileal apical sodium-dependent bile acid transporter, prevented liver injury. Conclusion: Constitutive ERK1 and ERK2 signaling has a redundant but critical physiological function in the down-regulation of hepatic bile acid synthesis to maintain normal liver homeostasis.
Collapse
Affiliation(s)
- Francesca Cingolani
- Division of Digestive DiseasesDepartment of MedicineEmory University School of MedicineAtlantaGAUSA
| | - Yunshan Liu
- Division of Digestive DiseasesDepartment of MedicineEmory University School of MedicineAtlantaGAUSA
| | - Yang Shen
- Division of Digestive DiseasesDepartment of MedicineEmory University School of MedicineAtlantaGAUSA
| | - Jing Wen
- Division of Digestive DiseasesDepartment of MedicineEmory University School of MedicineAtlantaGAUSA
| | - Alton B. Farris
- Department of Pathology & Laboratory MedicineEmory University School of MedicineAtlantaGAUSA
| | - Mark J. Czaja
- Division of Digestive DiseasesDepartment of MedicineEmory University School of MedicineAtlantaGAUSA
| |
Collapse
|
13
|
Live imaging approach of dynamic multicellular responses in ERK signaling during vertebrate tissue development. Biochem J 2022; 479:129-143. [PMID: 35050327 PMCID: PMC8883488 DOI: 10.1042/bcj20210557] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2021] [Revised: 12/28/2021] [Accepted: 01/05/2022] [Indexed: 11/17/2022]
Abstract
The chemical and mechanical responses of cells via the exchange of information during growth and development result in the formation of biological tissues. Information processing within the cells through the signaling pathways and networks inherent to the constituent cells has been well-studied. However, the cell signaling mechanisms responsible for generating dynamic multicellular responses in developing tissues remain unclear. Here, I review the dynamic multicellular response systems during the development and growth of vertebrate tissues based on the extracellular signal-regulated kinase (ERK) pathway. First, an overview of the function of the ERK signaling network in cells is provided, followed by descriptions of biosensors essential for live imaging of the quantification of ERK activity in tissues. Then adducing four examples, I highlight the contribution of live imaging techniques for studying the involvement of spatio-temporal patterns of ERK activity change in tissue development and growth. In addition, theoretical implications of ERK signaling are also discussed from the viewpoint of dynamic systems. This review might help in understanding ERK-mediated dynamic multicellular responses and tissue morphogenesis.
Collapse
|
14
|
Sumi K, Tago K, Nakazawa Y, Takahashi K, Ohe T, Mashino T, Funakoshi-Tago M. Novel Mechanism by a Bis-Pyridinium Fullerene Derivative to Induce Apoptosis by Enhancing the MEK-ERK Pathway in a Reactive Oxygen Species-Independent Manner in BCR-ABL-Positive Chronic Myeloid Leukemia-Derived K562 Cells. Int J Mol Sci 2022; 23:ijms23020749. [PMID: 35054935 PMCID: PMC8775703 DOI: 10.3390/ijms23020749] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2021] [Revised: 01/03/2022] [Accepted: 01/07/2022] [Indexed: 11/16/2022] Open
Abstract
In the treatment of breakpoint cluster region-Abelson (BCR-ABL)-positive chronic myeloid leukemia (CML) using BCR-ABL inhibitors, the appearance of a gatekeeper mutation (T315I) in BCR-ABL is a serious issue. Therefore, the development of novel drugs that overcome acquired resistance to BCR-ABL inhibitors by CML cells is required. We previously demonstrated that a bis-pyridinium fullerene derivative (BPF) induced apoptosis in human chronic myeloid leukemia (CML)-derived K562 cells partially through the generation of reactive oxygen species (ROS). We herein show that BPF enhanced the activation of the mitogen-activated protein kinase/extracellular signal-regulated kinase kinase-extracellular signal-regulated kinase (MEK-ERK) pathway in a ROS-independent manner. BPF-induced apoptosis was attenuated by trametinib, suggesting the functional involvement of the MEK-ERK pathway in apoptosis in K562 cells. In addition, the constitutive activation of the MEK-ERK pathway by the enforced expression of the BRAFV600E mutant significantly increased the sensitivity of K562 cells to BPF. These results confirmed for the first time that BPF induces apoptosis in K562 cells through dual pathways-ROS production and the activation of the MEK-ERK pathway. Furthermore, BPF induced cell death in transformed Ba/F3 cells expressing not only BCR-ABL but also T315I mutant through the activation of the MEK-ERK pathway. These results indicate that BPF is as an effective CML drug that overcomes resistance to BCR-ABL inhibitors.
Collapse
Affiliation(s)
- Kazuya Sumi
- Division of Hygienic Chemistry, Faculty of Pharmacy, Keio University, 1-5-30 Shibakoen, Minato-ku, Tokyo 105-8512, Japan; (K.S.); (Y.N.)
| | - Kenji Tago
- Division of Structural Biochemistry, Department of Biochemistry, Jichi Medical University, Shimotsuke 321-0498, Japan
- Correspondence: (K.T.); (M.F.-T.); Tel.: +81-3-5400-2689 (K.T. & M.F.-T.)
| | - Yosuke Nakazawa
- Division of Hygienic Chemistry, Faculty of Pharmacy, Keio University, 1-5-30 Shibakoen, Minato-ku, Tokyo 105-8512, Japan; (K.S.); (Y.N.)
| | - Kyoko Takahashi
- Division of Pharmaceutical Sciences, Faculty of Pharmacy, Keio University, 1-5-30 Shibakoen, Minato-ku, Tokyo 105-8512, Japan; (K.T.); (T.O.); (T.M.)
| | - Tomoyuki Ohe
- Division of Pharmaceutical Sciences, Faculty of Pharmacy, Keio University, 1-5-30 Shibakoen, Minato-ku, Tokyo 105-8512, Japan; (K.T.); (T.O.); (T.M.)
| | - Tadahiko Mashino
- Division of Pharmaceutical Sciences, Faculty of Pharmacy, Keio University, 1-5-30 Shibakoen, Minato-ku, Tokyo 105-8512, Japan; (K.T.); (T.O.); (T.M.)
| | - Megumi Funakoshi-Tago
- Division of Hygienic Chemistry, Faculty of Pharmacy, Keio University, 1-5-30 Shibakoen, Minato-ku, Tokyo 105-8512, Japan; (K.S.); (Y.N.)
- Correspondence: (K.T.); (M.F.-T.); Tel.: +81-3-5400-2689 (K.T. & M.F.-T.)
| |
Collapse
|
15
|
Maik-Rachline G, Wortzel I, Seger R. Alternative Splicing of MAPKs in the Regulation of Signaling Specificity. Cells 2021; 10:cells10123466. [PMID: 34943973 PMCID: PMC8699841 DOI: 10.3390/cells10123466] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2021] [Revised: 11/26/2021] [Accepted: 12/01/2021] [Indexed: 12/12/2022] Open
Abstract
The mitogen-activated protein kinase (MAPK) cascades transmit signals from extracellular stimuli to a variety of distinct cellular processes. The MAPKKs in each cascade specifically phosphorylate and activate their cognate MAPKs, indicating that this step funnels various signals into a seemingly linear pathway. Still, the effects of these cascades vary significantly, depending on the identity of the extracellular signals, which gives rise to proper outcomes. Therefore, it is clear that the specificity of the signals transmitted through the cascades is tightly regulated in order to secure the desired cell fate. Indeed, many regulatory components or processes that extend the specificity of the cascades have been identified. Here, we focus on a less discussed mechanism, that is, the role of distinct components in each tier of the cascade in extending the signaling specificity. We cover the role of distinct genes, and the alternatively spliced isoforms of MAPKKs and MAPKs, in the signaling specificity. The alternatively spliced MEK1b and ERK1c, which form an independent signaling route, are used as the main example. Unlike MEK1/2 and ERK1/2, this route’s functions are limited, including mainly the regulation of mitotic Golgi fragmentation. The unique roles of the alternatively spliced isoforms indicate that these components play an essential role in determining the proper cell fate in response to distinct stimulations.
Collapse
|
16
|
Sarkar M, Martufi M, Roman-Trufero M, Wang YF, Whilding C, Dormann D, Sabbattini P, Dillon N. CNOT3 interacts with the Aurora B and MAPK/ERK kinases to promote survival of differentiating mesendodermal progenitor cells. Mol Biol Cell 2021; 32:ar40. [PMID: 34613789 PMCID: PMC8694085 DOI: 10.1091/mbc.e21-02-0089] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2021] [Revised: 09/23/2021] [Accepted: 10/01/2021] [Indexed: 11/18/2022] Open
Abstract
Mesendoderm cells are key intermediate progenitors that form at the early primitive streak (PrS) and give rise to mesoderm and endoderm in the gastrulating embryo. We have identified an interaction between CNOT3 and the cell cycle kinase Aurora B that requires sequences in the NOT box domain of CNOT3 and regulates MAPK/ERK signaling during mesendoderm differentiation. Aurora B phosphorylates CNOT3 at two sites located close to a nuclear localization signal and promotes localization of CNOT3 to the nuclei of mouse embryonic stem cells (ESCs) and metastatic lung cancer cells. ESCs that have both sites mutated give rise to embryoid bodies that are largely devoid of mesoderm and endoderm and are composed mainly of cells with ectodermal characteristics. The mutant ESCs are also compromised in their ability to differentiate into mesendoderm in response to FGF2, BMP4, and Wnt3 due to reduced survival and proliferation of differentiating mesendoderm cells. We also show that the double mutation alters the balance of interaction of CNOT3 with Aurora B and with ERK and reduces phosphorylation of ERK in response to FGF2. Our results identify a potential adaptor function for CNOT3 that regulates the Ras/MEK/ERK pathway during embryogenesis.
Collapse
Affiliation(s)
- Moumita Sarkar
- Gene Regulation and Chromatin Group, Imperial College London, London W12 0NN, UK
| | - Matteo Martufi
- Gene Regulation and Chromatin Group, Imperial College London, London W12 0NN, UK
| | - Monica Roman-Trufero
- Gene Regulation and Chromatin Group, Imperial College London, London W12 0NN, UK
| | - Yi-Fang Wang
- Bioinformatics and Computing, Imperial College London, London W12 0NN, UK
| | - Chad Whilding
- Microscopy Facility, MRC London Institute of Medical Sciences, Imperial College London, London W12 0NN, UK
| | - Dirk Dormann
- Microscopy Facility, MRC London Institute of Medical Sciences, Imperial College London, London W12 0NN, UK
| | | | - Niall Dillon
- Gene Regulation and Chromatin Group, Imperial College London, London W12 0NN, UK
- Institute of Clinical Sciences, Imperial College London, London W12 0NN, UK
| |
Collapse
|
17
|
Sahu R, Upadhayay S, Mehan S. Inhibition of extracellular regulated kinase (ERK)-1/2 signaling pathway in the prevention of ALS: Target inhibitors and influences on neurological dysfunctions. Eur J Cell Biol 2021; 100:151179. [PMID: 34560374 DOI: 10.1016/j.ejcb.2021.151179] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2021] [Revised: 08/18/2021] [Accepted: 09/15/2021] [Indexed: 12/13/2022] Open
Abstract
Cell signal transduction pathways are essential modulators of several physiological and pathological processes in the brain. During overactivation, these signaling processes may lead to disease progression. Abnormal protein kinase activation is associated with several biological dysfunctions that facilitate neurodegeneration under different biological conditions. As a result, these signaling pathways are essential in understanding brain disorders' development or progression. Recent research findings indicate the crucial role of extracellular signal-regulated kinase-1/2 (ERK-1/2) signaling during the neuronal development process. ERK-1/2 is a key component of its mitogen-activated protein kinase (MAPK) group, controlling certain neurological activities by regulating metabolic pathways, cell proliferation, differentiation, and apoptosis. ERK-1/2 also influences neuronal elastic properties, nerve growth, and neurological and cognitive processing during brain injuries. The primary goal of this review is to elucidate the activation of ERK1/2 signaling, which is involved in the development of several ALS-related neuropathological dysfunctions. ALS is a rare neurological disorder category that mainly affects the nerve cells responsible for regulating voluntary muscle activity. ALS is progressive, which means that the symptoms are getting worse over time, and there is no cure for ALS and no effective treatment to avoid or reverse. Genetic abnormalities, oligodendrocyte degradation, glial overactivation, and immune deregulation are associated with ALS progression. Furthermore, the current review also identifies ERK-1/2 signaling inhibitors that can promote neuroprotection and neurotrophic effects against the clinical-pathological presentation of ALS. As a result, in the future, the potential ERK-1/2 signaling inhibitors could be used in the treatment of ALS and related neurocomplications.
Collapse
Affiliation(s)
- Rakesh Sahu
- Neuropharmacology Division, Department of Pharmacology, ISF College of Pharmacy, Moga, Punjab, India
| | - Shubham Upadhayay
- Neuropharmacology Division, Department of Pharmacology, ISF College of Pharmacy, Moga, Punjab, India
| | - Sidharth Mehan
- Neuropharmacology Division, Department of Pharmacology, ISF College of Pharmacy, Moga, Punjab, India.
| |
Collapse
|
18
|
Zhang RM, Zeyer KA, Odenthal N, Zhang Y, Reinhardt DP. The fibrillin-1 RGD motif posttranscriptionally regulates ERK1/2 signaling and fibroblast proliferation via miR-1208. FASEB J 2021; 35:e21598. [PMID: 33871068 DOI: 10.1096/fj.202100282r] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2021] [Revised: 03/28/2021] [Accepted: 03/31/2021] [Indexed: 12/17/2022]
Abstract
Fibrillin-1 is an extracellular matrix protein which contains one conserved RGD integrin-binding motif. It constitutes the backbone of microfibrils in many tissues, and mutations in fibrillin-1 cause various connective tissue disorders. Although it is well established that fibrillin-1 interacts with several RGD-dependent integrins, very little is known about the associated intracellular signaling pathways. Recent published evidence identified a subset of miRNAs regulated by fibrillin-1 RGD-cell adhesion, with miR-1208 among the most downregulated. The present study shows that the downregulated miR-1208 controls fibroblast proliferation. Inhibitor experiments revealed that fibrillin-1 RGD suppressed miR-1208 expression via c-Src kinase and the downstream JNK signaling. Bioinformatic prediction and experimental target sequence validation demonstrated four miR-1208 binding sites on the ERK2 mRNA and one on the MEK1 mRNA. ERK2 and MEK1 are critical proliferation-promoting kinases. Decreased miR-1208 levels elevated the total and phosphorylated ERK1/2 and MEK1/2 protein levels and the phosphorylated to total ERK1/2 ratio. Together, the data demonstrate a novel outside-in signaling mechanism explaining how fibrillin-1 RGD-cell binding regulates fibroblast proliferation.
Collapse
Affiliation(s)
- Rong-Mo Zhang
- Faculty of Medicine and Health Sciences, Department of Anatomy and Cell Biology, McGill University, Montreal, Canada
| | - Karina A Zeyer
- Faculty of Medicine and Health Sciences, Department of Anatomy and Cell Biology, McGill University, Montreal, Canada
| | - Nadine Odenthal
- Department of Natural Science, University of Lübeck, Lübeck, Germany
| | - Yiyun Zhang
- Faculty of Medicine and Health Sciences, Department of Anatomy and Cell Biology, McGill University, Montreal, Canada
| | - Dieter P Reinhardt
- Faculty of Medicine and Health Sciences, Department of Anatomy and Cell Biology, McGill University, Montreal, Canada.,Faculty of Dentistry, McGill University, Montreal, Canada
| |
Collapse
|
19
|
Kulesza T, Piwkowska A. The impact of type III sodium-dependent phosphate transporters (Pit 1 and Pit 2) on podocyte and kidney function. J Cell Physiol 2021; 236:7176-7185. [PMID: 33738792 DOI: 10.1002/jcp.30368] [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: 12/21/2020] [Revised: 02/26/2021] [Accepted: 03/04/2021] [Indexed: 01/07/2023]
Abstract
The sodium-dependent phosphate transporters Pit 1 and Pit 2 belong to the solute carrier 20 (SLC20) family of membrane proteins. They are ubiquitously distributed in the human body. Their crucial function is the intracellular transport of inorganic phosphate (Pi) in the form of H2 PO4 - . They are one of the main elements in maintaining physiological phosphate homeostasis. Recent data have emerged that indicate novel roles of Pit 1 and Pit 2 proteins besides the well-known function of Pi transporters. These membrane proteins are believed to be precise phosphate sensors that mediate Pi-dependent intracellular signaling. They are also involved in insulin signaling and influence cellular insulin sensitivity. In diseases that are associated with hyperphosphatemia, such as diabetes and chronic kidney disease (CKD), disturbances in the function of Pit 1 and Pit 2 are observed. Phosphate transporters from the SLC20 family participate in the calcification of soft tissues, mainly blood vessels, during the course of CKD. The glomerulus and podocytes therein can also be a target of pathological calcification that damages these structures. A few studies have demonstrated the development of Pi-dependent podocyte injury that is mediated by Pit 1 and Pit 2. This paper discusses the role of Pit 1 and Pit 2 proteins in podocyte function, mainly in the context of the development of pathological calcification that disrupts permeability of the renal filtration barrier. We also describe the mechanisms that may contribute to podocyte damage by Pit 1 and Pit 2.
Collapse
Affiliation(s)
- Tomasz Kulesza
- Laboratory of Molecular and Cellular Nephrology, Mossakowski Medical Research Institute, Polish Academy of Sciences, Gdansk, Poland
| | - Agnieszka Piwkowska
- Laboratory of Molecular and Cellular Nephrology, Mossakowski Medical Research Institute, Polish Academy of Sciences, Gdansk, Poland
| |
Collapse
|
20
|
Jafari S, Farsani FM, Ganji M, Ganjalikhany MR. The functional regulatory details of ERK2 in complex with RSK1: an in silico insight. RSC Adv 2021; 11:11048-11056. [PMID: 35423626 PMCID: PMC8695832 DOI: 10.1039/d1ra01020d] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2021] [Accepted: 03/01/2021] [Indexed: 12/12/2022] Open
Abstract
Protein kinases play a significant role in cellular activation procedures by exhibiting a vivid selection in the target, as well as recognizing and phosphorylating them. Extracellular signal-regulated kinase 2 (ERK2) is one of the main kinases in the mitogen-activated protein kinase (MAPK) signaling cascade and engages in dynamically regulating the activities of signaling proteins and physiological processes, including cell proliferation, differentiation, adhesion, migration, and survival. Predicting collective dynamic and structural motions in biological macromolecules is pivotal to obtain a better understanding of the majority of biological processes. Here, through molecular dynamic simulation and normal mode analysis, we investigated ERK2 conformations, in the forms of active (phosphorylated), inactive (unphosphorylated), and in a complex with its substrate, ribosomal protein S6 kinase alpha-1 (RSK1), to determine functional characteristics. Our finding demonstrated that ERK2 plays a switch role in the regulation of pathways. In the case that this protein kinase is in the active form, all critical regions shift to be prepared to accept the substrate and catalytic action. Meanwhile, inactive ERK2 shows contrasting results in which all motions tend to close the catalytic site and cease the phosphorylation action in the MAPK cascade. These findings are in line with those from other similar studies and provide us with novel molecular target regions and recent details on how this mechanism works. DFG, αC-helix orientation regarding the active site position and distance between K54 and Glu71 in the active and inactive states of ERK2.![]()
Collapse
Affiliation(s)
- Sepideh Jafari
- Department of Cell and Molecular Biology, Faculty of Biological Science and Technology, University of Isfahan Isfahan Iran +98-31-37932250 +98-31-37932250
| | - Farzaneh Mohamadi Farsani
- Department of Cell and Molecular Biology, Faculty of Biological Science and Technology, University of Isfahan Isfahan Iran +98-31-37932250 +98-31-37932250
| | - Maziar Ganji
- Department of Medical Genetics, School of Medicine, Shahid Beheshti University of Medical Sciences Tehran Iran
| | - Mohamad Reza Ganjalikhany
- Department of Cell and Molecular Biology, Faculty of Biological Science and Technology, University of Isfahan Isfahan Iran +98-31-37932250 +98-31-37932250
| |
Collapse
|
21
|
Chen Z, Che D, Gu X, Lin J, Deng J, Jiang P, Xu K, Xu B, Zhang T. Upregulation of PEDF Predicts a Poor Prognosis and Promotes Esophageal Squamous Cell Carcinoma Progression by Modulating the MAPK/ERK Signaling Pathway. Front Oncol 2021; 11:625612. [PMID: 33718190 PMCID: PMC7953146 DOI: 10.3389/fonc.2021.625612] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2020] [Accepted: 01/18/2021] [Indexed: 02/05/2023] Open
Abstract
Invasion and metastasis represent the primary causes of therapeutic failure in patients diagnosed with esophageal squamous cell carcinoma (ESCC). The lack of effective treatment strategies for metastatic ESCC is the major cause of the low survival rate. Therefore, it is crucial to understand the molecular mechanisms underlying ESCC metastasis and identify potential biomarkers for targeted therapy. Herein, we reported that PEDF is significantly correlated with tumor cell invasion and metastasis in ESCC. The high expression of PEDF is an independent unfavorable prognostic factor for ESCC patients’ overall survival (OS). We successfully developed and verified a nomogram to predict the preoperative OS of ESCC patients, and the actual and nomogram-predicted 1-, 3-, and 5-year survival rates had good consistency. The receiver operating characteristic (ROC) curve showed that the area under the curve (AUC) values for 1-, 3- and 5- survival were 0.764, 0.871, and 0.91, respectively. Overexpression of PEDF significantly promoted the migration and invasion of ESCC cells in vitro, while silencing PEDF yielded the opposite effects. Elevated levels of PEDF altered the expression of proteins involved in epithelial–mesenchymal transition (EMT), as indicated by the upregulation of N-cadherin and the downregulation of α-catenin and E-cadherin in ESCC cells. Mechanistically, PEDF promoted tumor cell motility and EMT by activating the MAPK/ERK signaling pathway. In conclusion, our results reveal that PEDF is involved in ESCC metastasis and could act as a prognostic factor for ESCC. Our research provides a fresh perspective into the mechanism of ESCC metastasis.
Collapse
Affiliation(s)
- Zui Chen
- Department of Laboratory Medicine, Guangzhou First People's Hospital, School of Medicine, South China University of Technology, Guangzhou, China
| | - Di Che
- Department of Clinical Biological Resource Bank, Guangzhou Institute of Pediatrics, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou, China
| | - Xiaoqiong Gu
- Department of Clinical Biological Resource Bank, Guangzhou Institute of Pediatrics, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou, China
| | - Jiamin Lin
- Department of Laboratory Medicine, Guangzhou First People's Hospital, School of Medicine, South China University of Technology, Guangzhou, China
| | - Jing Deng
- Department of Laboratory Medicine, Guangzhou First People's Hospital, School of Medicine, South China University of Technology, Guangzhou, China
| | - Ping Jiang
- Department of Laboratory Medicine, Guangzhou First People's Hospital, School of Medicine, South China University of Technology, Guangzhou, China
| | - Kaixiong Xu
- Department of Laboratory Medicine, Guangzhou First People's Hospital, School of Medicine, South China University of Technology, Guangzhou, China
| | - Banglao Xu
- Department of Laboratory Medicine, Guangzhou First People's Hospital, School of Medicine, South China University of Technology, Guangzhou, China
| | - Ting Zhang
- Department of Laboratory Medicine, Guangzhou First People's Hospital, School of Medicine, South China University of Technology, Guangzhou, China
| |
Collapse
|
22
|
Chromosomal and molecular pathway alterations in the neuroendocrine carcinoma and adenocarcinoma components of gastric mixed neuroendocrine-nonneuroendocrine neoplasm. Mod Pathol 2020; 33:2602-2613. [PMID: 32461621 DOI: 10.1038/s41379-020-0579-z] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2020] [Revised: 05/12/2020] [Accepted: 05/13/2020] [Indexed: 12/18/2022]
Abstract
Gastric mixed adenoneuroendocrine carcinoma (MANEC) is a clinically aggressive subtype of mixed neuroendocrine-nonneuroendocrine neoplasm (MiNEN) with unclear clonal origin. In this study, we analyzed high-resolution copy number (CN) profiling data using the OncoScan CNV Assay in the neuroendocrine carcinoma (NEC) and adenocarcinoma components of eight MANECs. Some common CNVs, including the gain of CCNE1 (19q12) and the loss of FAT1 (4q35.2), were frequently detected in both components; these CNVs were verified by FISH, qPCR and immunohistochemistry staining assays in samples with sufficient material. The identification of common CNVs in both components supports the likelihood of single clonal origin of morphologically heterogeneous tumor cells and suggests several novel genetic events potentially involved in the development of gastric MANEC. We also detected and validated some CNVs and alterations specific for the NEC component, such as MAPK1 loss and MAPK signaling pathway alterations, which could contribute to the neuroendocrine differentiation of gastric MANEC. In addition, we found that the NEC component presented more CNVs and greater CN loss than the adenocarcinoma component (P = 0.007 and P = 0.004, respectively); the NEC components from different cases were not clustered in the hierarchical clustering analysis, indicating the marked genetic heterogenicity of the NEC component in gastric MANEC. In summary, this study describes the cytogenetic characteristics of each component of gastric MANEC, providing some clues for further studies on the development and progression of gastric MANEC as well as providing some potential therapeutic targets.
Collapse
|
23
|
Wei G, Gao N, Chen J, Fan L, Zeng Z, Gao G, Li L, Fang G, Hu K, Pang X, Fan HY, Clevers H, Liu M, Zhang X, Li D. Erk and MAPK signaling is essential for intestinal development through Wnt pathway modulation. Development 2020; 147:dev.185678. [PMID: 32747435 DOI: 10.1242/dev.185678] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2019] [Accepted: 07/23/2020] [Indexed: 12/28/2022]
Abstract
Homeostasis of intestinal stem cells (ISCs) is maintained by the orchestration of niche factors and intrinsic signaling networks. Here, we have found that deletion of Erk1 and Erk2 (Erk1/2) in intestinal epithelial cells at embryonic stages resulted in an unexpected increase in cell proliferation and migration, expansion of ISCs, and formation of polyp-like structures, leading to postnatal death. Deficiency of epithelial Erk1/2 results in defects in secretory cell differentiation as well as impaired mesenchymal cell proliferation and maturation. Deletion of Erk1/2 strongly activated Wnt signaling through both cell-autonomous and non-autonomous mechanisms. In epithelial cells, Erk1/2 depletion resulted in loss of feedback regulation, leading to Ras/Raf cascade activation that transactivated Akt activity to stimulate the mTor and Wnt/β-catenin pathways. Moreover, Erk1/2 deficiency reduced the levels of Indian hedgehog and the expression of downstream pathway components, including mesenchymal Bmp4 - a Wnt suppressor in intestines. Inhibition of mTor signaling by rapamycin partially rescued Erk1/2 depletion-induced intestinal defects and significantly prolonged the lifespan of mutant mice. These data demonstrate that Erk/Mapk signaling functions as a key modulator of Wnt signaling through coordination of epithelial-mesenchymal interactions during intestinal development.
Collapse
Affiliation(s)
- Gaigai Wei
- Shanghai Key Laboratory of Regulatory Biology, Joint Research Center for Translational Medicine, ECNU-Fengxian Hospital, Institute of Biomedical Sciences and School of Life Sciences, East China Normal University, Shanghai 200241, China
| | - Na Gao
- Shanghai Key Laboratory of Regulatory Biology, Joint Research Center for Translational Medicine, ECNU-Fengxian Hospital, Institute of Biomedical Sciences and School of Life Sciences, East China Normal University, Shanghai 200241, China
| | - Jiwei Chen
- Shanghai Key Laboratory of Regulatory Biology, Joint Research Center for Translational Medicine, ECNU-Fengxian Hospital, Institute of Biomedical Sciences and School of Life Sciences, East China Normal University, Shanghai 200241, China
| | - Lingling Fan
- Shanghai Key Laboratory of Regulatory Biology, Joint Research Center for Translational Medicine, ECNU-Fengxian Hospital, Institute of Biomedical Sciences and School of Life Sciences, East China Normal University, Shanghai 200241, China
| | - Zhiyang Zeng
- Shanghai Key Laboratory of Regulatory Biology, Joint Research Center for Translational Medicine, ECNU-Fengxian Hospital, Institute of Biomedical Sciences and School of Life Sciences, East China Normal University, Shanghai 200241, China
| | - Ganglong Gao
- Fengxian Hospital affiliated to Southern Medical University, Shanghai 201499, China
| | - Liang Li
- Shanghai Key Laboratory of Regulatory Biology, Joint Research Center for Translational Medicine, ECNU-Fengxian Hospital, Institute of Biomedical Sciences and School of Life Sciences, East China Normal University, Shanghai 200241, China
| | - Guojiu Fang
- Fengxian Hospital affiliated to Southern Medical University, Shanghai 201499, China
| | - Kewen Hu
- Shanghai Key Laboratory of Regulatory Biology, Joint Research Center for Translational Medicine, ECNU-Fengxian Hospital, Institute of Biomedical Sciences and School of Life Sciences, East China Normal University, Shanghai 200241, China
| | - Xiufeng Pang
- Shanghai Key Laboratory of Regulatory Biology, Joint Research Center for Translational Medicine, ECNU-Fengxian Hospital, Institute of Biomedical Sciences and School of Life Sciences, East China Normal University, Shanghai 200241, China
| | - Heng-Yu Fan
- Life Sciences Institute, Zhejiang University, Hangzhou 310058, China
| | - Hans Clevers
- Hubrecht Institute, Royal Netherlands Academy of Arts and Sciences (KNAW), Uppsalalaan 8, Utrecht 3584 CT, The Netherlands
| | - Mingyao Liu
- Shanghai Key Laboratory of Regulatory Biology, Joint Research Center for Translational Medicine, ECNU-Fengxian Hospital, Institute of Biomedical Sciences and School of Life Sciences, East China Normal University, Shanghai 200241, China
| | - Xueli Zhang
- Shanghai Key Laboratory of Regulatory Biology, Joint Research Center for Translational Medicine, ECNU-Fengxian Hospital, Institute of Biomedical Sciences and School of Life Sciences, East China Normal University, Shanghai 200241, China .,Fengxian Hospital affiliated to Southern Medical University, Shanghai 201499, China
| | - Dali Li
- Shanghai Key Laboratory of Regulatory Biology, Joint Research Center for Translational Medicine, ECNU-Fengxian Hospital, Institute of Biomedical Sciences and School of Life Sciences, East China Normal University, Shanghai 200241, China
| |
Collapse
|
24
|
Parrilla I, Martinez EA, Gil MA, Cuello C, Roca J, Rodriguez-Martinez H, Martinez CA. Boar seminal plasma: current insights on its potential role for assisted reproductive technologies in swine. Anim Reprod 2020; 17:e20200022. [PMID: 33029213 PMCID: PMC7534575 DOI: 10.1590/1984-3143-ar2020-0022] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Abstract
Seminal plasma (SP) supports not only sperm function but also the ability of spermatozoa to withstand biotechnological procedures as artificial insemination, freezing or sex sorting. Moreover, evidence has been provided that SP contains identifiable molecules which can act as fertility biomarkers, and even improve the output of assisted reproductive technologies by acting as modulators of endometrial and embryonic changes of gene expression, thus affecting embryo development and fertility beyond the sperm horizon. In this overview, we discuss current knowledge of the composition of SP, mainly proteins and cytokines, and their influence on semen basic procedures, such as liquid storage or cryopreservation. The role of SP as modulator of endometrial and embryonic molecular changes that lead to successful pregnancy will also be discussed.
Collapse
Affiliation(s)
- Inmaculada Parrilla
- Departmento de Medicina y Cirugía Animal, Facultad de Veterinaria, Campus de de Excelencia International "Campus Mare Nostrum", Universidad de Murcia, Murcia, Spain.,Instituto Murciano de Investigación Biosanitaria, Campus de Ciencias de la Salud, Murcia, Spain
| | - Emilio Arsenio Martinez
- Departmento de Medicina y Cirugía Animal, Facultad de Veterinaria, Campus de de Excelencia International "Campus Mare Nostrum", Universidad de Murcia, Murcia, Spain.,Instituto Murciano de Investigación Biosanitaria, Campus de Ciencias de la Salud, Murcia, Spain
| | - Maria Antonia Gil
- Departmento de Medicina y Cirugía Animal, Facultad de Veterinaria, Campus de de Excelencia International "Campus Mare Nostrum", Universidad de Murcia, Murcia, Spain.,Instituto Murciano de Investigación Biosanitaria, Campus de Ciencias de la Salud, Murcia, Spain
| | - Cristina Cuello
- Departmento de Medicina y Cirugía Animal, Facultad de Veterinaria, Campus de de Excelencia International "Campus Mare Nostrum", Universidad de Murcia, Murcia, Spain.,Instituto Murciano de Investigación Biosanitaria, Campus de Ciencias de la Salud, Murcia, Spain
| | - Jordi Roca
- Departmento de Medicina y Cirugía Animal, Facultad de Veterinaria, Campus de de Excelencia International "Campus Mare Nostrum", Universidad de Murcia, Murcia, Spain.,Instituto Murciano de Investigación Biosanitaria, Campus de Ciencias de la Salud, Murcia, Spain
| | - Heriberto Rodriguez-Martinez
- Department of Biomedical & Clinical Sciences, BKH/Obstetrics & Gynaecology, Faculty of Medicine and Health Sciences, Linköping University, Linköping, Sweden
| | - Cristina Alicia Martinez
- Department of Biomedical & Clinical Sciences, BKH/Obstetrics & Gynaecology, Faculty of Medicine and Health Sciences, Linköping University, Linköping, Sweden
| |
Collapse
|
25
|
Kölle S, Hughes B, Steele H. Early embryo-maternal communication in the oviduct: A review. Mol Reprod Dev 2020; 87:650-662. [PMID: 32506761 DOI: 10.1002/mrd.23352] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2020] [Revised: 05/05/2020] [Accepted: 05/10/2020] [Indexed: 12/11/2022]
Abstract
An intact embryo-maternal communication is critical for the establishment of a successful pregnancy. To date, a huge number of studies have been performed describing the complex process of embryo-maternal signaling within the uterus. However, recent studies indicate that the early embryo communicates with the oviductal cells shortly after fertilizationand that this is important for the successful establishment of pregnancy. Only if the early embryo is capable to signal the mother within a precise timeframe and to garner a response, will the embryo be able to survive and reach the uterus. This review will give an overview of all the experimental designs which have investigated embryo-maternal interaction in the oviduct. In addition to that, it will provide a comprehensive analysis of the findings to date elucidating the morphological and molecular changes in the oviduct which are induced by the presence of the early embryo highlighting how the tubal responses affect embryo development and survival.
Collapse
Affiliation(s)
- Sabine Kölle
- Health Sciences Centre, School of Medicine, University College Dublin, Dublin, Ireland
| | - Barbara Hughes
- Health Sciences Centre, School of Medicine, University College Dublin, Dublin, Ireland
| | - Heather Steele
- Health Sciences Centre, School of Medicine, University College Dublin, Dublin, Ireland
| |
Collapse
|
26
|
Hall C, Yu H, Choi E. Insulin receptor endocytosis in the pathophysiology of insulin resistance. Exp Mol Med 2020; 52:911-920. [PMID: 32576931 PMCID: PMC7338473 DOI: 10.1038/s12276-020-0456-3] [Citation(s) in RCA: 53] [Impact Index Per Article: 10.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2020] [Accepted: 05/11/2020] [Indexed: 12/16/2022] Open
Abstract
Insulin signaling controls cell growth and metabolic homeostasis. Dysregulation of this pathway causes metabolic diseases such as diabetes. Insulin signaling pathways have been extensively studied. Upon insulin binding, the insulin receptor (IR) triggers downstream signaling cascades. The active IR is then internalized by clathrin-mediated endocytosis. Despite decades of studies, the mechanism and regulation of clathrin-mediated endocytosis of IR remain incompletely understood. Recent studies have revealed feedback regulation of IR endocytosis through Src homology phosphatase 2 (SHP2) and the mitogen-activated protein kinase (MAPK) pathway. Here we review the molecular mechanism of IR endocytosis and its impact on the pathophysiology of insulin resistance, and discuss the potential of SHP2 as a therapeutic target for type 2 diabetes.
Collapse
Affiliation(s)
- Catherine Hall
- Department of Pathology and Cell Biology, Vagelos College of Physicians and Surgeons, Columbia University, 630 West 168th Street, New York, NY, 10032, USA
| | - Hongtao Yu
- Laboratory of Cell Biology, School of Life Sciences, Westlake University, Hangzhou, Zhejiang, 310024, China.
- Department of Pharmacology, University of Texas Southwestern Medical Center, 6001 Forest Park Road, Dallas, TX, 75390, USA.
| | - Eunhee Choi
- Department of Pathology and Cell Biology, Vagelos College of Physicians and Surgeons, Columbia University, 630 West 168th Street, New York, NY, 10032, USA.
| |
Collapse
|
27
|
Salgado-Lucio ML, Ramírez-Ramírez D, Jorge-Cruz CY, Roa-Espitia AL, Hernández-González EO. FAK regulates actin polymerization during sperm capacitation via the ERK2/GEF-H1/RhoA signaling pathway. J Cell Sci 2020; 133:jcs239186. [PMID: 32107290 DOI: 10.1242/jcs.239186] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2019] [Accepted: 02/08/2020] [Indexed: 12/18/2022] Open
Abstract
Actin polymerization is a crucial process during sperm capacitation. We have recently described the participation of FAK during actin polymerization in guinea pig spermatozoa. However, the mechanism by which FAK mediates these processes is unknown. Our previous data have shown that MAPK1 (hereafter referred to as ERK2) is activated during the first minutes of capacitation, and inhibition of ERK2 blocked actin polymerization and the acrosome reaction. In this current study, we found that FAK is involved in ERK2 activation - as FAK was phosphorylated at tyrosine residue 925 and bound to Grb2 - and that inhibition of FAK results in a significant decrease of ERK2 activation. We also confirmed the presence of Rho guanine nucleotide exchange factor 2 (ARHGEF2, hereafter referred to as GEF-H1), which is able to associate with RhoA during capacitation. RhoA activation and its participation in actin polymerization were also analyzed. Inhibition of FAK or ERK1/2 impeded GEF-H1 phosphorylation, RhoA activation, and the association between GEF-H1 and RhoA. Finally, we observed the presence of fibronectin on the sperm surface, its role in sperm-sperm interaction as well as participation of β-integrin in the activation of ERK2. Our results show that the signaling pathway downstream of fibronectin, via integrin, FAK, Grb2, MEK1/2, ERK2, GEF-H1 and RhoA regulates the actin polymerization associated with spermatozoa capacitation.
Collapse
Affiliation(s)
- Monica L Salgado-Lucio
- Departamento de Biología Celular, Centro de Investigación y Estudios Avanzados del Instituto Politécnico Nacional, Av. Instituto Politécnico Nacional 2508, San Pedro Zacatenco, CDMX 07360, México
| | - Danelia Ramírez-Ramírez
- Departamento de Biología Celular, Centro de Investigación y Estudios Avanzados del Instituto Politécnico Nacional, Av. Instituto Politécnico Nacional 2508, San Pedro Zacatenco, CDMX 07360, México
| | - Coral Y Jorge-Cruz
- Departamento de Biología Celular, Centro de Investigación y Estudios Avanzados del Instituto Politécnico Nacional, Av. Instituto Politécnico Nacional 2508, San Pedro Zacatenco, CDMX 07360, México
| | - Ana L Roa-Espitia
- Departamento de Biología Celular, Centro de Investigación y Estudios Avanzados del Instituto Politécnico Nacional, Av. Instituto Politécnico Nacional 2508, San Pedro Zacatenco, CDMX 07360, México
| | - Enrique O Hernández-González
- Departamento de Biología Celular, Centro de Investigación y Estudios Avanzados del Instituto Politécnico Nacional, Av. Instituto Politécnico Nacional 2508, San Pedro Zacatenco, CDMX 07360, México
| |
Collapse
|
28
|
Yang M, Chen M, Liu G, Yang C, Li Z. Molecular cloning and characterization of a cDNA encoding extracellular signal-regulated kinase (ERK) from the blood clam Tegillarca granosa. DEVELOPMENTAL AND COMPARATIVE IMMUNOLOGY 2020; 105:103602. [PMID: 31918206 DOI: 10.1016/j.dci.2019.103602] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/09/2019] [Revised: 12/27/2019] [Accepted: 12/27/2019] [Indexed: 06/10/2023]
Abstract
The blood clam Tegillarca granosa is a member of the most economically important bivalve mollusk species in the Asia-Pacific region. T. granosa entirely depends on innate immunity for pathogen defense. However, there are very few reports on the immune responses of T. granosa to various pathogens. In our study, we cloned and characterized an ERK homolog from T. granosa, which was defined as TgERK. The full-length cDNA sequence of TgERK was 1644 bp in length and encoded a conserved S_TKc domain (residues 21-309) in the N terminus. The TgERK mRNA was universally expressed in all examined tissues, with the highest expression level found in hemocytes. Lipopolysaccharide (LPS) and Vibrio alginolyticus challenges strongly enhanced the expression of ERK in T. granosa, which was consistent with the results of an in vitro challenge study with cultured T. granosa hemocytes. Pathogen invasion also upregulated the expression of downstream genes in the ERK signaling pathway, such as CREB, c-Fos and SIRT1. Moreover, TgERK knockdown resulted in decreased expression of these downstream genes. Inhibition of ERK by its inhibitor U0126 decreased T. granosa hemocyte viability in a dose-dependent manner. Taken together, our results demonstrated that TgERK was a crucial regulator of the immune response to pathogen invasion, which indicated new knowledge of hemocyte immunity in T. granosa and provided a novel key molecule in immune regulation for controlling diseases in T. granosa aquaculture.
Collapse
Affiliation(s)
- Minghan Yang
- State Key Laboratory Breeding Base of Marine Genetic Resources, Third Institute of Oceanography, Ministry of Natural Resources, Xiamen, 361005, PR China
| | - Mingliang Chen
- State Key Laboratory Breeding Base of Marine Genetic Resources, Third Institute of Oceanography, Ministry of Natural Resources, Xiamen, 361005, PR China; Co-Innovation Center of Jiangsu Marine Bio-industry Technology, Jiangsu Ocean University, Lianyungang, 222005, PR China.
| | - Guosheng Liu
- State Key Laboratory Breeding Base of Marine Genetic Resources, Third Institute of Oceanography, Ministry of Natural Resources, Xiamen, 361005, PR China
| | - Chunyan Yang
- School of Life Science, Xiamen University, Xiamen, 361005, PR China.
| | - Zengpeng Li
- State Key Laboratory Breeding Base of Marine Genetic Resources, Third Institute of Oceanography, Ministry of Natural Resources, Xiamen, 361005, PR China.
| |
Collapse
|
29
|
Zhou X, Wei LJ, Li JQ, Zhang JY, Zhu SL, Zhang HT, Jia J, Yu J, Wang SS, Feng L. The Activation of Peroxisome Proliferator-activated Receptor γ Enhances Insulin Signaling Pathways Via Up-regulating Chemerin Expression in High Glucose Treated HTR-8/SVneo Cells. MATERNAL-FETAL MEDICINE 2020. [DOI: 10.1097/fm9.0000000000000044] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022] Open
|
30
|
Zhang J, Cui Y, Li X, Xiao Y, Liu L, Jia F, He J, Xie X, Parthasarathy S, Hao H, Fang N. 5F peptide promotes endothelial differentiation of bone marrow stem cells through activation of ERK1/2 signaling. Eur J Pharmacol 2020; 876:173051. [PMID: 32145325 DOI: 10.1016/j.ejphar.2020.173051] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2019] [Revised: 02/25/2020] [Accepted: 02/28/2020] [Indexed: 11/29/2022]
Abstract
Synthetic apolipoprotein A-I (apoA-I) mimetic peptide 5F exhibits anti-atherosclerotic ability with largely unknown mechanism(s). Bone marrow (BM)-derived endothelial progenitor cells (EPCs) play a critical role in vascular integrity and function. The objective of the present study was to evaluate the effect of 5F on endothelial differentiation of BM stem cells and related mechanisms. Murine BM multipotent adult progenitor cells (MAPCs) were induced to differentiate into endothelial cells in vitro with or without 5F. The expression of endothelial markers vWF, Flk-1 and CD31 was significantly increased in the cells treated with 5F with enhanced in vitro vascular tube formation and LDL uptake without significant changes on proliferation and stem cell maker Oct-4 expression. Phosphorylated ERK1/2, not Akt, was significantly increased in 5F-treated cells. Treatment of MAPCs with PD98059 or small interfering RNA against ERK2 substantially attenuated ERK1/2 phosphorylation, and effectively prevented 5F-induced enhancement of endothelial differentiation of MAPCs. In vivo studies revealed that 5F increased EPCs number in the BM in mice after acute hindlimb ischemia that was effectively prevented with PD98059 treatment. These data supported the conclusion that 5F promoted endothelial differentiation of MAPCs through activation of ERK1/2 signaling.
Collapse
Affiliation(s)
- Jia Zhang
- Department of Geriatrics, Renji Hospital, School of Medicine, Shanghai Jiao Tong University , Shanghai, 200127, China; Davis Heart & Lung Research Institute and Division of Cardiovascular Medicine, The Ohio State University Wexner Medical Center, Columbus, OH, USA
| | - Yuqi Cui
- Davis Heart & Lung Research Institute and Division of Cardiovascular Medicine, The Ohio State University Wexner Medical Center, Columbus, OH, USA
| | - Xin Li
- Davis Heart & Lung Research Institute and Division of Cardiovascular Medicine, The Ohio State University Wexner Medical Center, Columbus, OH, USA
| | - Yuan Xiao
- Davis Heart & Lung Research Institute and Division of Cardiovascular Medicine, The Ohio State University Wexner Medical Center, Columbus, OH, USA
| | - Lingjuan Liu
- Davis Heart & Lung Research Institute and Division of Cardiovascular Medicine, The Ohio State University Wexner Medical Center, Columbus, OH, USA
| | - Fengpeng Jia
- Davis Heart & Lung Research Institute and Division of Cardiovascular Medicine, The Ohio State University Wexner Medical Center, Columbus, OH, USA
| | - Jianfeng He
- Davis Heart & Lung Research Institute and Division of Cardiovascular Medicine, The Ohio State University Wexner Medical Center, Columbus, OH, USA
| | - Xiaoyun Xie
- Davis Heart & Lung Research Institute and Division of Cardiovascular Medicine, The Ohio State University Wexner Medical Center, Columbus, OH, USA
| | - Sampath Parthasarathy
- Burnett School of Biomedical Sciences, University of Central Florida College of Medicine, USA
| | - Hong Hao
- Davis Heart & Lung Research Institute and Division of Cardiovascular Medicine, The Ohio State University Wexner Medical Center, Columbus, OH, USA
| | - Ningyuan Fang
- Department of Geriatrics, Renji Hospital, School of Medicine, Shanghai Jiao Tong University , Shanghai, 200127, China.
| |
Collapse
|
31
|
Kim JY, Lee DH, Kim JK, Choi HS, Dwivedi B, Rupji M, Kowalski J, Green SJ, Song H, Park WJ, Chang JY, Kim TM, Park C. ETV2/ER71 regulates the generation of FLK1 + cells from mouse embryonic stem cells through miR-126-MAPK signaling. Stem Cell Res Ther 2019; 10:328. [PMID: 31744543 PMCID: PMC6862833 DOI: 10.1186/s13287-019-1466-8] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2019] [Revised: 09/28/2019] [Accepted: 10/22/2019] [Indexed: 11/10/2022] Open
Abstract
Previous studies including ours have demonstrated a critical function of the transcription factor ETV2 (ets variant 2; also known as ER71) in determining the fate of cardiovascular lineage development. However, the underlying mechanisms of ETV2 function remain largely unknown. In this study, we demonstrated the novel function of the miR (micro RNA)-126-MAPK (mitogen-activated protein kinase) pathway in ETV2-mediated FLK1 (fetal liver kinase 1; also known as VEGFR2)+ cell generation from the mouse embryonic stem cells (mESCs). By performing a series of experiments including miRNA sequencing and ChIP (chromatin immunoprecipitation)-PCR, we found that miR-126 is directly induced by ETV2. Further, we identified that miR-126 can positively regulate the generation of FLK1+ cells by activating the MAPK pathway through targeting SPRED1 (sprouty-related EVH1 domain containing 1). Further, we showed evidence that JUN/FOS activate the enhancer region of FLK1 through AP1 (activator protein 1) binding sequences. Our findings provide insight into the novel molecular mechanisms of ETV2 function in regulating cardiovascular lineage development from mESCs.
Collapse
Affiliation(s)
- Ju Young Kim
- Department of Pediatrics, Emory University School of Medicine, 2015 Uppergate Dr. Atlanta, Atlanta, GA, 30322, USA.,Children's Heart Research & Outcomes (HeRO) Center, Children's Healthcare of Atlanta & Emory University, Atlanta, GA, USA.,Molecular and Systems Pharmacology Program, Emory University, Atlanta, GA, USA
| | - Dong Hun Lee
- Department of Pediatrics, Emory University School of Medicine, 2015 Uppergate Dr. Atlanta, Atlanta, GA, 30322, USA.,Children's Heart Research & Outcomes (HeRO) Center, Children's Healthcare of Atlanta & Emory University, Atlanta, GA, USA
| | - Joo Kyung Kim
- Department of Pediatrics, Emory University School of Medicine, 2015 Uppergate Dr. Atlanta, Atlanta, GA, 30322, USA.,Children's Heart Research & Outcomes (HeRO) Center, Children's Healthcare of Atlanta & Emory University, Atlanta, GA, USA
| | - Hong Seo Choi
- Department of Pediatrics, Emory University School of Medicine, 2015 Uppergate Dr. Atlanta, Atlanta, GA, 30322, USA
| | - Bhakti Dwivedi
- Winship Cancer Institute, Emory University, Atlanta, GA, USA
| | - Manali Rupji
- Winship Cancer Institute, Emory University, Atlanta, GA, USA
| | - Jeanne Kowalski
- Winship Cancer Institute, Emory University, Atlanta, GA, USA.,Department of Biostatistics and Bioinformatics, Emory University, Atlanta, GA, USA.,Present Address: Department of Oncology, University of Texas at Austin, Austin, TX, 78712, USA
| | - Stefan J Green
- Sequencing Core, Research Resources Center, University of Illinois at Chicago, Chicago, IL, USA
| | - Heesang Song
- Department of Pediatrics, Emory University School of Medicine, 2015 Uppergate Dr. Atlanta, Atlanta, GA, 30322, USA.,Department of Biochemistry and Molecular Biology, Chosun University School of Medicine, Gwangju, IL, Republic of Korea
| | - Won Jong Park
- Department of Pediatrics, Emory University School of Medicine, 2015 Uppergate Dr. Atlanta, Atlanta, GA, 30322, USA
| | - Ji Young Chang
- Department of Pediatrics, Emory University School of Medicine, 2015 Uppergate Dr. Atlanta, Atlanta, GA, 30322, USA
| | - Tae Min Kim
- Graduate School of International Agricultural Technology and Institute of Green-Bio Science and Technology, Seoul National University, Pyeongchang, Gangwon-do, Republic of Korea
| | - Changwon Park
- Department of Pediatrics, Emory University School of Medicine, 2015 Uppergate Dr. Atlanta, Atlanta, GA, 30322, USA. .,Children's Heart Research & Outcomes (HeRO) Center, Children's Healthcare of Atlanta & Emory University, Atlanta, GA, USA. .,Molecular and Systems Pharmacology Program, Emory University, Atlanta, GA, USA. .,Biochemistry, Cell Biology and Developmental Biology Program, Emory University, Atlanta, GA, USA.
| |
Collapse
|
32
|
Nakanoh S, Agata K. Evolutionary view of pluripotency seen from early development of non-mammalian amniotes. Dev Biol 2019; 452:95-103. [PMID: 31029690 DOI: 10.1016/j.ydbio.2019.04.014] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2019] [Revised: 03/22/2019] [Accepted: 04/24/2019] [Indexed: 11/24/2022]
Abstract
Early embryonic cells are capable of acquiring numerous developmental fates until they become irreversibly committed to specific lineages depending on intrinsic determinants and/or regional interactions. From fertilization to gastrulation, such pluripotent cells first increase in number and then turn to undergoing differentiation. Mechanisms regulating pluripotency in each species attract great interest in developmental biology. Also, outlining the evolutionary background of pluripotency can enhance our understanding of mammalian pluripotency and provide a broader view of early development of vertebrates. Here, we introduce integrative models of pluripotent states in amniotes (mammals, birds and reptiles) to offer a comprehensive overview of widely accepted knowledge about mammalian pluripotency and our recent findings in non-mammalian amniotes, such as chicken and gecko. In particular, we describe 1) the IL6/Stat3 signaling pathway as a positive regulator of naive pluripotency, 2) Fgf/Erk signaling as a process that prepares cells for differentiation, 3) the role of the interactions between these two signaling pathways during the transition from pluripotency to differentiation, and 4) functional diversification of two transcription factors, Class V POUs and Nanog. In the last section, we also briefly discuss possible relationships of unique cell cycle properties of early embryonic cells with signaling pathways and developmental potentials in the pluripotent cell states.
Collapse
Affiliation(s)
- Shota Nakanoh
- Division of Embryology, National Institute for Basic Biology, Okazaki 444-8787, Japan; Wellcome-MRC Cambridge Stem Cell Institute, Anne McLaren Laboratory, University of Cambridge, Cambridge CB2 0SZ, UK; Department of Surgery, University of Cambridge, Cambridge CB2 0QQ, UK.
| | - Kiyokazu Agata
- Graduate Course in Life Science, Gakushuin University, Toyoshima-ku, Tokyo 171-8588, Japan.
| |
Collapse
|
33
|
Brown JL, Sones JL, Angulo CN, Abbott K, Miller AD, Boehm U, Roberson MS. Conditional loss of ERK1 and ERK2 results in abnormal placentation and delayed parturition in the mouse. Sci Rep 2019; 9:9641. [PMID: 31270345 PMCID: PMC6610138 DOI: 10.1038/s41598-019-45997-0] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2018] [Accepted: 06/21/2019] [Indexed: 11/11/2022] Open
Abstract
Extracellular-signal-regulated kinases (ERK) 1 and 2 regulate many aspects of the hypothalamic-pituitary-gonadal axis. We sought to understand the role of ERK1/2 signaling in cells expressing a Cre allele regulated by the endogenous GnRHR promoter (GRIC-ERKdko). Adult female GRIC-ERKdko mice were hypogonadotropic and anovulatory. Gonadotropin administration and mating led to pregnancy in one-third of the ERKdko females. Litters from ERKdko females and pup weights were reduced coincident with delayed parturition and 100% neonatal mortality. Based on this, we examined Cre expression in implantation sites as a potential mechanism. GnRHR mRNA levels at e10.5 and e12.5 were comparable to pituitary levels from adult female mice at proestrus and GnRHR mRNA in decidua was enriched compared to whole implantation site. In vivo studies confirmed recombination in decidua, and GRIC-ERKdko placentas showed reduced ERK2 expression. Histopathology revealed abnormalities in placental architecture in the GRIC-ERKdko animals. Regions of apoptosis at the decidual/uterine interface at e18.5 were observed in control animals but apoptotic tone in these regions was reduced in ERKdko animals. These studies support a potential model of ERK-dependent signaling within the implantation site leading to loss of placental architecture and mis-regulation of apoptotic events at parturition occurring coincident with prolonged gestation and neonatal mortality.
Collapse
Affiliation(s)
- Jessica L Brown
- Department Biomedical Science, College of Veterinary Medicine, Cornell University, Ithaca, NY, USA
| | - Jennifer L Sones
- Department Biomedical Science, College of Veterinary Medicine, Cornell University, Ithaca, NY, USA
- Department Veterinary Clinical Sciences, School of Veterinary Medicine, Louisiana State University, Baton Rouge, LA, USA
| | - Cynthia N Angulo
- Department Biomedical Science, College of Veterinary Medicine, Cornell University, Ithaca, NY, USA
| | - Keelin Abbott
- Department Biomedical Science, College of Veterinary Medicine, Cornell University, Ithaca, NY, USA
| | - Andrew D Miller
- Department Biomedical Science, College of Veterinary Medicine, Cornell University, Ithaca, NY, USA
| | - Ulrich Boehm
- Experimental Pharmacology, Center for Molecular Signaling (PZMS), Saarland University School of Medicine, Homburg, Germany
| | - Mark S Roberson
- Department Biomedical Science, College of Veterinary Medicine, Cornell University, Ithaca, NY, USA.
| |
Collapse
|
34
|
Nitration-induced ubiquitination and degradation control quality of ERK1. Biochem J 2019; 476:1911-1926. [PMID: 31196894 PMCID: PMC6604951 DOI: 10.1042/bcj20190240] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2019] [Revised: 06/12/2019] [Accepted: 06/13/2019] [Indexed: 12/19/2022]
Abstract
The mitogen-activated protein kinase ERK1/2 (ERKs, extracellular-regulated protein kinases) plays important roles in a wide spectrum of cellular processes and have been implicated in many disease states. The spatiotemporal regulation of ERK activity has been extensively studied. However, scarce information has been available regarding the quality control of the kinases to scavenge malfunctioning ERKs. Using site-specific mutagenesis and mass spectrometry, we found that the disruption of the conserved H-bond between Y210 and E237 of ERK1 through point mutation at or naturally occurring nitration on Y210 initiates a quality control program dependent on chaperon systems and CHIP (C-terminal of Hsp70-interacting protein)-mediated ubiquitination and degradation. The H-bond is also important for the quality control of ERK2, but through a distinct mechanism. These findings clearly demonstrate how malfunctioning ERKs are eliminated when cells are in certain stress conditions or unhealthy states, and could represent a general mechanism for scavenging malfunctioning kinases in stress conditions.
Collapse
|
35
|
Kurtzeborn K, Kwon HN, Kuure S. MAPK/ERK Signaling in Regulation of Renal Differentiation. Int J Mol Sci 2019; 20:E1779. [PMID: 30974877 PMCID: PMC6479953 DOI: 10.3390/ijms20071779] [Citation(s) in RCA: 52] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2019] [Revised: 04/04/2019] [Accepted: 04/08/2019] [Indexed: 12/20/2022] Open
Abstract
Congenital anomalies of the kidney and urinary tract (CAKUT) are common birth defects derived from abnormalities in renal differentiation during embryogenesis. CAKUT is the major cause of end-stage renal disease and chronic kidney diseases in children, but its genetic causes remain largely unresolved. Here we discuss advances in the understanding of how mitogen-activated protein kinase/extracellular signal-regulated kinase (MAPK/ERK) activity contributes to the regulation of ureteric bud branching morphogenesis, which dictates the final size, shape, and nephron number of the kidney. Recent studies also demonstrate that the MAPK/ERK pathway is directly involved in nephrogenesis, regulating both the maintenance and differentiation of the nephrogenic mesenchyme. Interestingly, aberrant MAPK/ERK signaling is linked to many cancers, and recent studies suggest it also plays a role in the most common pediatric renal cancer, Wilms' tumor.
Collapse
Affiliation(s)
- Kristen Kurtzeborn
- Helsinki Institute of Life Science, University of Helsinki, FIN-00014 Helsinki, Finland.
- Stem Cells and Metabolism Research Program, Faculty of Medicine, University of Helsinki, FIN-00014 Helsinki, Finland.
| | - Hyuk Nam Kwon
- Helsinki Institute of Life Science, University of Helsinki, FIN-00014 Helsinki, Finland.
- Stem Cells and Metabolism Research Program, Faculty of Medicine, University of Helsinki, FIN-00014 Helsinki, Finland.
| | - Satu Kuure
- Helsinki Institute of Life Science, University of Helsinki, FIN-00014 Helsinki, Finland.
- Stem Cells and Metabolism Research Program, Faculty of Medicine, University of Helsinki, FIN-00014 Helsinki, Finland.
- GM-unit, Laboratory Animal Center, Helsinki Institute of Life Science, University of Helsinki, FIN-00014 Helsinki, Finland.
| |
Collapse
|
36
|
Alese MO, Moodley J, Naicker TA. Signalling of ERK1/2, P38MAPK and P90RSK in HIV-associated pre-eclampsia. J OBSTET GYNAECOL 2019; 39:612-618. [PMID: 30821550 DOI: 10.1080/01443615.2018.1547695] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Due to their significance in trophoblast differentiation and survival, we evaluated the expression of the cell signalling molecules; Extracellular signal-regulated kinase 1/2 (ERK1/2), Mitogen Activated Protein Kinase 38 (MAPK38) and p90 ribosomal protein S6 kinase (p90 RSK) in buffy coat samples. Eighty pregnant women attending a large hospital in Durban, South Africa were assigned into normotensive and pre-eclamptic groups and further stratified by their HIV status. The degree of phosphorylation of the analytes was determined using the Bio-Plex ProTM Cell Signalling Immunoassay. There was a significantly lower protein concentration of the analytes in the pre-eclamptic versus the normotensive patients, irrespective of HIV status (p < .0001). Also, there was no significant difference in expression of ERK1/2 (p = .4369), p38MAPK (p = .4720) and p90 RSK (p = .0188), according to HIV status. This study demonstrates a down-regulation of ERK1/2, p38MAPK and p90RSK prosurvival markers in pre-eclampsia. This implicates the involvement of the MAPK pathway in the pathogenesis of preeclampsia. Activation of these pathways may prove useful in increasing the body of evidence on prevention of placenta dysfunction and apoptosis. Impact statement What is already known on this subject? Preeclampsia occurring in co-morbidity with HIV is a public health problem among pregnant, black South-African women. There have been conflicting theories regarding the predisposition to the development of preeclampsia as a result of compromised immune response due to HIV infection. In normal pregnancies, the MAPK pathway plays a significant role in molecular processes involved in the cells including survival and differentiation of the placental trophoblast. ERK1/2, p38MAPK and p90RSK are members of the MAPK family, which are pro-apoptotic. Inhibition in the signalling of MAPKs has been found to result in oxidative stress, a process which contributes to the defective trophoblast invasion seen in preeclampsia. What do the results of this study add? The results from this study showed that there is no relationship between HIV infection and an increased predisposition to the development of preeclampsia. In addition, this study highlights a downregulation in the expression of ERK1/2, p38 MAPK and p90RSK in preeclampsia. What are the implications of these findings for clinical practice and/or further research? These findings demonstrate the potential of these analytes as biomarkers for the diagnosis of preeclampsia. Also, this may serve as a framework for further research in the prevention of preeclampsia by elucidating more on the pathway.
Collapse
Affiliation(s)
- Margaret O Alese
- a Optics and Imaging Centre, Nelson R Mandela School of Medicine , University of KwaZulu-Natal , Durban , South Africa
| | - Jagidesa Moodley
- b Women's Health and HIV Research Group, Nelson R Mandela School of Medicine , University of KwaZulu-Natal , Durban , South Africa
| | - Thajasvarie A Naicker
- a Optics and Imaging Centre, Nelson R Mandela School of Medicine , University of KwaZulu-Natal , Durban , South Africa
| |
Collapse
|
37
|
Ma X, Peng S, Zhou X, Li S, Jin P. The amphioxus ERK2 gene is involved in innate immune response to LPS stimulation. FISH & SHELLFISH IMMUNOLOGY 2019; 86:64-69. [PMID: 30439498 DOI: 10.1016/j.fsi.2018.11.018] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/27/2018] [Revised: 11/02/2018] [Accepted: 11/05/2018] [Indexed: 06/09/2023]
Abstract
The ERK2 gene is a member of the MAPK family, which plays very important roles in responses to external environmental pressures. However, the ERK2 has yet not been identified in amphioxus to date. To further illuminate the function and evolutionary mechanism of the ERK2 gene, in this present study, we have cloned the full length of the ERK2 gene of Branchiostoma belcheri (designed as AmphiERK2), which is highly homologous to these vertebrate ERK2 genes. The AmphiERK2 protein contains the conserved S_TKc domain and the TEY motif, and its 3D structure is also highly similar to human ERK2 protein. Taken together, our results indicate that the AmphiERK2 gene belongs to a member of the ERK2 gene family. We further use qRT-PCR technology to detect an ubiquitous expression of AmphiERK2 gene in all five investigated tissues (muscle, notochord, gill, hepatic caecum and intestine), and the expression level of AmphiERK2 in both notochord and muscle is significantly higher than the other three tissues. Meanwhile our results also demonstrate that LPS stimulation can induce the up-regulation expression of AmphiERK2 gene and significantly increase the phosphorylation level of AmphiERK2 protein, which seems to imply that the AmphiERK2 may be involved in amphioxus innate immune responses. Overall, our findings provide an important insight into amphioxus innate immune function and evolution of the ERK2 gene family.
Collapse
Affiliation(s)
- Xiangyu Ma
- School of Food Science, Nanjing Xiaozhuang University, Nanjing, 211171, China; The Key Laboratory of Developmental Genes and Human Disease, Institute of Life Sciences, Southeast University, Nanjing, 210096, China
| | - Shuangli Peng
- Laboratory for Comparative Genomics and Bioinformatics & Jiangsu Key Laboratory for Biodiversity and Biotechnology, College of Life Science, Nanjing Normal University, Nanjing, 210046, China
| | - Xue Zhou
- School of Chemistry and Biological Engineering, Nanjing Normal University Taizhou College, Taizhou, 225300, China
| | - Shengjie Li
- School of Food Science, Nanjing Xiaozhuang University, Nanjing, 211171, China.
| | - Ping Jin
- Laboratory for Comparative Genomics and Bioinformatics & Jiangsu Key Laboratory for Biodiversity and Biotechnology, College of Life Science, Nanjing Normal University, Nanjing, 210046, China.
| |
Collapse
|
38
|
Tyrosine nitration of human ERK1 introduces an intra-hydrogen bond by molecular dynamics simulations. Struct Chem 2019. [DOI: 10.1007/s11224-019-01306-z] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
|
39
|
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
|
40
|
Zhang H, Ou Z, Xu M, Huang X, Liu W, Shi Y, He M. Molecular cloning and characterization of a putative mitogen-activated protein kinase (Erk1/2) gene: Involvement in mantle immunity of Pinctada fucata. FISH & SHELLFISH IMMUNOLOGY 2018; 80:63-70. [PMID: 29859309 DOI: 10.1016/j.fsi.2018.05.047] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/30/2018] [Revised: 05/21/2018] [Accepted: 05/29/2018] [Indexed: 06/08/2023]
Abstract
Extracellular signal-regulated kinases (ERKs) are conserved and related with protein-serine/threonine kinases that participate in the regulation of multiple biological processes, such as cell survival, cell differentiation, proliferation, metabolism, and inflammation. However, little is known about the roles of this kinase in the pearl oyster. In this study, we cloned and identified an ERK homolog from Pinctada fucata (PfErk). Furthermore, we have unraveled its expressional kinetics after lipopolysaccharide (LPS) and polyinosinic-epolycytidylic acid (poly I:C) immune challenge. Pferk harbored a 5' untranslated region (UTR) of 12 bp, a coding sequence of 1074 bp, and a 3' UTR of 882 bp. The putative peptide comprised a predicted molecular mass of 41.19 kDa, with a theoretical pI of 6.15. Sequence analysis showed that it possesses one STK catalytic domain and a conserved His-Arg-Asp (HRD) domain. In addition, a canonical Thr-Glu-Tyr (TEY) dual phosphorylation motif and an ATRW substrate binding site were also identified in the coding protein. Homology assessment of PfErk showed high similarity to Homo sapiens ERK. Phylogenetic analysis supported a close evolutionary relationship with molluscan orthologs. The expression patterns of Pferk were observed in seven different tissues of pearl oyster, with highest expression in the mantle and lowest expression in the digestive gland. Pferk mRNA expression levels were detected at developmental stages, with the highest expression in D-shaped larvae, followed by the 32-cell stage. The mRNA expression of Pferk was upregulated significantly in P. fucata mantle primary cells and mantle tissue after LPS and poly (I:C) treatment, and PfErk phosphorylation levels were activated by LPS and poly (I:C) challenges. Overall, our results suggested that PfErk may play important roles in pearl oyster innate immunity, and provided a new understanding of mantle immunity in the pearl oyster.
Collapse
Affiliation(s)
- Hua Zhang
- CAS Key Laboratory of Tropical Marine Bio-resources and Ecology, Guangdong Provincial Key Laboratory of Applied Marine Biology, South China Sea Institute of Oceanology, Chinese Academy of Sciences, Guangdong, Guangzhou, 510301, China; University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Zekui Ou
- CAS Key Laboratory of Tropical Marine Bio-resources and Ecology, Guangdong Provincial Key Laboratory of Applied Marine Biology, South China Sea Institute of Oceanology, Chinese Academy of Sciences, Guangdong, Guangzhou, 510301, China; University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Meng Xu
- CAS Key Laboratory of Tropical Marine Bio-resources and Ecology, Guangdong Provincial Key Laboratory of Applied Marine Biology, South China Sea Institute of Oceanology, Chinese Academy of Sciences, Guangdong, Guangzhou, 510301, China; University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Xiande Huang
- CAS Key Laboratory of Tropical Marine Bio-resources and Ecology, Guangdong Provincial Key Laboratory of Applied Marine Biology, South China Sea Institute of Oceanology, Chinese Academy of Sciences, Guangdong, Guangzhou, 510301, China
| | - Wenguang Liu
- CAS Key Laboratory of Tropical Marine Bio-resources and Ecology, Guangdong Provincial Key Laboratory of Applied Marine Biology, South China Sea Institute of Oceanology, Chinese Academy of Sciences, Guangdong, Guangzhou, 510301, China
| | - Yu Shi
- CAS Key Laboratory of Tropical Marine Bio-resources and Ecology, Guangdong Provincial Key Laboratory of Applied Marine Biology, South China Sea Institute of Oceanology, Chinese Academy of Sciences, Guangdong, Guangzhou, 510301, China
| | - Maoxian He
- CAS Key Laboratory of Tropical Marine Bio-resources and Ecology, Guangdong Provincial Key Laboratory of Applied Marine Biology, South China Sea Institute of Oceanology, Chinese Academy of Sciences, Guangdong, Guangzhou, 510301, China.
| |
Collapse
|
41
|
Thei L, Rocha-Ferreira E, Peebles D, Raivich G, Hristova M. Extracellular signal-regulated kinase 2 has duality in function between neuronal and astrocyte expression following neonatal hypoxic-ischaemic cerebral injury. J Physiol 2018; 596:6043-6062. [PMID: 29873394 PMCID: PMC6265549 DOI: 10.1113/jp275649] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2018] [Accepted: 05/29/2018] [Indexed: 01/08/2023] Open
Abstract
Key points This study identifies phosphorylated extracellular signal‐regulated kinase (ERK) to be immediately diminished followed by a rapid if transient increase for up to 4 h following hypoxic–ischaemic insult (HI) in the neonatal mouse. Phosphorylated ERK up‐regulation was prevented with systemic injection of the mitogen‐activated protein kinase kinase (MEK) inhibitor SL327. Treatment with SL327 both pre‐ and post‐HI gave a strong reduction in the number of dying cells and microgliosis. By utilising transgenic mouse mutations, we observe that neuronal ERK2 significantly contributes to tissue damage, while ERK1 and astrocytic ERK2 are neuroprotective. Compared to global inactivation, selective cell‐specific interference with ERK activity could result in stronger neuroprotection.
Abstract Hypoxia–ischaemia (HI) is a major cause of neonatal brain injury resulting in cerebral palsy, epilepsy, cognitive impairment and other neurological disabilities. The role of extracellular signal‐regulated kinase (ERK) isoforms and their mitogen‐activated protein kinase kinase (MEK)‐dependent phosphorylation in HI has previously been explored but remains unresolved at cellular level. This is pertinent given the growing awareness of the role of non‐neuronal cells in neuroprotection. Using a modified Rice–Vannucci model of HI in the neonatal mouse we observed time‐ and cell‐dependent ERK phosphorylation (pERK), with strongly up‐regulated pERK immunoreactivity first in periventricular white matter axons within 15–45 min of HI, followed by forebrain astrocytes and neurons (1–4 h post‐HI), and return to baseline by 16 h. We explored the effects of pharmacological ERK blockade through the MEK inhibitor SL327 on neonatal HI‐brain damage following HI alone (30 or 60 min) or lipopolysaccharide (LPS)‐sensitised HI insult (30 min). Global inhibition of ERK phosphorylation with systemically applied SL327 abolished forebrain pERK immunoreactivity, and significantly reduced cell death and associated microglial activation at 48 h post‐HI. We then explored the effects of cell‐specific ERK2 deletion alone or in combination with global ERK1 knockout under the same conditions of HI insult. Neuronal ERK2 deletion strongly decreased infarct size, neuronal cell death and microglial activation in grey matter following both HI alone or LPS‐sensitised HI. ERK1 deletion attenuated the protective effect of neuronal ERK2 deletion. Removal of astroglial ERK2 produced a reverse response, with a 3‐ to 4‐fold increase in microglial activation and cell death. Our data suggest a cell‐specific and time‐dependent role of ERK in neonatal HI, with a predominant, neurotoxic effect of neuronal ERK2, which is counteracted by neuroprotection by ERK1 and astrocytic ERK2. Overall, global pharmacological inhibition of ERK phosphorylation is strongly neuroprotective. This study identifies phosphorylated extracellular signal‐regulated kinase (ERK) to be immediately diminished followed by a rapid if transient increase for up to 4 h following hypoxic–ischaemic insult (HI) in the neonatal mouse. Phosphorylated ERK up‐regulation was prevented with systemic injection of the mitogen‐activated protein kinase kinase (MEK) inhibitor SL327. Treatment with SL327 both pre‐ and post‐HI gave a strong reduction in the number of dying cells and microgliosis. By utilising transgenic mouse mutations, we observe that neuronal ERK2 significantly contributes to tissue damage, while ERK1 and astrocytic ERK2 are neuroprotective. Compared to global inactivation, selective cell‐specific interference with ERK activity could result in stronger neuroprotection.
Collapse
Affiliation(s)
- Laura Thei
- UCL Institute for Women's Health, Maternal and Fetal Medicine, Perinatal Brain Repair Group, London, WC1E 6HX, UK.,School of Pharmacy, University of Reading, Reading, RG6 6UA, UK
| | - Eridan Rocha-Ferreira
- UCL Institute for Women's Health, Maternal and Fetal Medicine, Perinatal Brain Repair Group, London, WC1E 6HX, UK.,Institute of Clinical Sciences, University of Gothenburg, Gothenburg, SE 416 85, Sweden
| | - Donald Peebles
- UCL Institute for Women's Health, Maternal and Fetal Medicine, Perinatal Brain Repair Group, London, WC1E 6HX, UK
| | - Gennadij Raivich
- UCL Institute for Women's Health, Maternal and Fetal Medicine, Perinatal Brain Repair Group, London, WC1E 6HX, UK
| | - Mariya Hristova
- UCL Institute for Women's Health, Maternal and Fetal Medicine, Perinatal Brain Repair Group, London, WC1E 6HX, UK
| |
Collapse
|
42
|
Kahnamouyi S, Nouri M, Farzadi L, Darabi M, Hosseini V, Mehdizadeh A. The role of mitogen-activated protein kinase-extracellular receptor kinase pathway in female fertility outcomes: a focus on pituitary gonadotropins regulation. Ther Adv Endocrinol Metab 2018; 9:209-215. [PMID: 29977499 PMCID: PMC6022971 DOI: 10.1177/2042018818772775] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/06/2017] [Accepted: 03/30/2018] [Indexed: 11/16/2022] Open
Abstract
Mammalian reproduction systems are largely regulated by the secretion of two gonadotropins, that is, luteinizing hormone (LH) and follicle-stimulating hormone (FSH). The main action of LH and FSH on the ovary is to stimulate secretion of estradiol and progesterone, which play an important role in the ovarian function and reproductive cycle control. FSH and LH secretions are strictly controlled by the gonadotropin-releasing hormone (GnRH), which is secreted from the hypothalamus into the pituitary vascular system. Maintaining normal secretion of LH and FSH is dependent on pulsatile secretion of GnRH. Extracellular signal-regulated kinase (ERK) proteins, as the main components of mitogen-activated protein kinase (MAPK) signaling pathways, are involved in the primary regulation of GnRH-stimulated transcription of the gonadotropins' α subunit in the pituitary cells. However, GnRH-stimulated expression of the β subunit has not yet been reported. Furthermore, GnRH-mediated stimulation of ERK1 and ERK2 leads to several important events such as cell proliferation and differentiation. In this review, we briefly introduce the relationship between ERK signaling and gonadotropin secretion, and its importance in female infertility.
Collapse
Affiliation(s)
- Samira Kahnamouyi
- Stem cell Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | | | - Laya Farzadi
- Women Reproductive Health Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Masoud Darabi
- Department of Biochemistry and Clinical Laboratories, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Vahid Hosseini
- Department of Biochemistry and Clinical Laboratories, Tabriz University of Medical Sciences, Tabriz, Iran
| | | |
Collapse
|
43
|
Pfeiffer V, Götz R, Camarero G, Heinsen H, Blum R, Rapp UR. Impaired neuronal maturation of hippocampal neural progenitor cells in mice lacking CRAF. PLoS One 2018; 13:e0192067. [PMID: 29590115 PMCID: PMC5873938 DOI: 10.1371/journal.pone.0192067] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2017] [Accepted: 01/16/2018] [Indexed: 11/19/2022] Open
Abstract
RAF kinases are major constituents of the mitogen activated signaling pathway, regulating cell proliferation, differentiation and cell survival of many cell types, including neurons. In mammals, the family of RAF proteins consists of three members, ARAF, BRAF, and CRAF. Ablation of CRAF kinase in inbred mouse strains causes major developmental defects during fetal growth and embryonic or perinatal lethality. Heterozygous germline mutations in CRAF result in Noonan syndrome, which is characterized by neurocognitive impairment that may involve hippocampal physiology. The role of CRAF signaling during hippocampal development and generation of new postnatal hippocampal granule neurons has not been examined and may provide novel insight into the cause of hippocampal dysfunction in Noonan syndrome. In this study, by crossing CRAF-deficiency to CD-1 outbred mice, a CRAF mouse model was established which enabled us to investigate the interplay of neural progenitor proliferation and postmitotic differentiation during adult neurogenesis in the hippocampus. Albeit the general morphology of the hippocampus was unchanged, CRAF-deficient mice displayed smaller granule cell layer (GCL) volume at postnatal day 30 (P30). In CRAF-deficient mice a substantial number of abnormal, chromophilic, fast dividing cells were found in the subgranular zone (SGZ) and hilus of the dentate gyrus (DG), indicating that CRAF signaling contributes to hippocampal neural progenitor proliferation. CRAF-deficient neural progenitor cells showed an increased cell death rate and reduced neuronal maturation. These results indicate that CRAF function affects postmitotic neural cell differentiation and points to a critical role of CRAF-dependent growth factor signaling pathway in the postmitotic development of adult-born neurons.
Collapse
Affiliation(s)
- Verena Pfeiffer
- University of Würzburg, Institute of Anatomy and Cell Biology, Koellikerstraße 6, Würzburg, Germany
- University of Würzburg, Institute for Medical Radiation and Cell Research (MSZ), Versbacher Strasse 5, Würzburg, Germany
- * E-mail:
| | - Rudolf Götz
- University of Würzburg, Institute for Medical Radiation and Cell Research (MSZ), Versbacher Strasse 5, Würzburg, Germany
- Institute for Clinical Neurobiology, University Hospital Würzburg, Versbacher Strasse 5, Würzburg, Germany
| | - Guadelupe Camarero
- University of Würzburg, Institute for Medical Radiation and Cell Research (MSZ), Versbacher Strasse 5, Würzburg, Germany
| | - Helmut Heinsen
- University of Würzburg, Department of Psychiatry, Psychosomatics and Psychotherapy, Margarethe-Höppel-Platz 1, Würzburg, Germany
- Universidade de Sao Paulo Faculdade de Medicina, Pathology—LIM 44 Sao Paulo, SP, Brazil
| | - Robert Blum
- Institute for Clinical Neurobiology, University Hospital Würzburg, Versbacher Strasse 5, Würzburg, Germany
| | - Ulf Rüdiger Rapp
- University of Würzburg, Institute for Medical Radiation and Cell Research (MSZ), Versbacher Strasse 5, Würzburg, Germany
- Department of Lung Development and Remodeling, Max Planck Institute for Heart and Lung Research, Parkstr.1, Bad Nauheim, Germany
| |
Collapse
|
44
|
Eblen ST. Extracellular-Regulated Kinases: Signaling From Ras to ERK Substrates to Control Biological Outcomes. Adv Cancer Res 2018; 138:99-142. [PMID: 29551131 DOI: 10.1016/bs.acr.2018.02.004] [Citation(s) in RCA: 131] [Impact Index Per Article: 18.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
The extracellular-regulated kinases ERK1 and ERK2 are evolutionarily conserved, ubiquitous serine-threonine kinases that are involved in regulating cellular signaling in both normal and pathological conditions. Their expression is critical for development and their hyperactivation is a major factor in cancer development and progression. Since their discovery as one of the major signaling mediators activated by mitogens and Ras mutation, we have learned much about their regulation, including their activation, binding partners and substrates. In this review I will discuss some of what has been discovered about the members of the Ras to ERK pathway, including regulation of their activation by growth factors and cell adhesion pathways. Looking downstream of ERK activation I will also highlight some of the many ERK substrates that have been discovered, including those involved in feedback regulation, cell migration and cell cycle progression through the control of transcription, pre-mRNA splicing and protein synthesis.
Collapse
Affiliation(s)
- Scott T Eblen
- Medical University of South Carolina, Charleston, SC, United States.
| |
Collapse
|
45
|
Gunawardhana N, Jang S, Choi YH, Hong YA, Jeon YE, Kim A, Su H, Kim JH, Yoo YJ, Merrell DS, Kim J, Cha JH. Helicobacter pylori-Induced HB-EGF Upregulates Gastrin Expression via the EGF Receptor, C-Raf, Mek1, and Erk2 in the MAPK Pathway. Front Cell Infect Microbiol 2018; 7:541. [PMID: 29379775 PMCID: PMC5775237 DOI: 10.3389/fcimb.2017.00541] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2017] [Accepted: 12/26/2017] [Indexed: 12/22/2022] Open
Abstract
Helicobacter pylori is associated with hypergastrinemia, which has been linked to the development of gastric diseases. Although the molecular mechanism is not fully understood, H. pylori is known to modulate the Erk pathway for induction of gastrin expression. Herein we found that an epidermal growth factor (EGF) receptor kinase inhibitor significantly blocked H. pylori-induced gastrin promoter activity, suggesting involvement of EGF receptor ligands. Indeed, H. pylori induced mRNA expression of EGF family members such as amphiregulin, EGF, heparin-binding EGF-like growth factor (HB-EGF), and transforming growth factor-α. Of these, specific siRNA targeting of HB-EGF significantly blocked H. pylori-induced gastrin expression. Moreover, H. pylori induced HB-EGF ectodomain shedding, which we found to be a critical process for H. pylori-induced gastrin expression. Thus, we demonstrate a novel role for human mature HB-EGF in stimulating gastrin promoter activity during H. pylori infection. Further investigation using specific siRNAs targeting each isoform of Raf, Mek, and Erk elucidated that the mechanism underlying H. pylori-induced gastrin expression can be delineated as the sequential activation of HB-EGF, the EGF receptor, C-Raf, Mek1, and the Erk2 molecules in the MAPK pathway. Surprisingly, whereas Erk2 acts as a potent activator of gastrin expression, siRNA knockdown of Erk1 induced gastrin promoter activity, suggesting that Erk1 typically acts as a repressor of gastrin expression. Elucidation of the mechanism of gastrin modulation by HB-EGF-mediated EGF receptor transactivation should facilitate the development of therapeutic strategies against H. pylori-related hypergastrinemia and consequently gastric disease development, including gastric cancers.
Collapse
Affiliation(s)
- Niluka Gunawardhana
- Department of Oral Biology, Oral Science Research Center, Department of Applied Life Science, The Graduate School, BK21 Plus Project, Yonsei University College of Dentistry, Seoul, South Korea.,Department of Basic Sciences, Faculty of Dental Sciences, University of Peradeniya, Peradeniya, Sri Lanka
| | - Sungil Jang
- Department of Oral Biology, Oral Science Research Center, Department of Applied Life Science, The Graduate School, BK21 Plus Project, Yonsei University College of Dentistry, Seoul, South Korea
| | - Yun Hui Choi
- Department of Oral Biology, Oral Science Research Center, Department of Applied Life Science, The Graduate School, BK21 Plus Project, Yonsei University College of Dentistry, Seoul, South Korea
| | - Youngmin A Hong
- Department of Oral Biology, Oral Science Research Center, Department of Applied Life Science, The Graduate School, BK21 Plus Project, Yonsei University College of Dentistry, Seoul, South Korea
| | - Yeong-Eui Jeon
- Department of Oral Biology, Oral Science Research Center, Department of Applied Life Science, The Graduate School, BK21 Plus Project, Yonsei University College of Dentistry, Seoul, South Korea
| | - Aeryun Kim
- Department of Oral Biology, Oral Science Research Center, Department of Applied Life Science, The Graduate School, BK21 Plus Project, Yonsei University College of Dentistry, Seoul, South Korea
| | - Hanfu Su
- Department of Oral Biology, Oral Science Research Center, Department of Applied Life Science, The Graduate School, BK21 Plus Project, Yonsei University College of Dentistry, Seoul, South Korea.,Microbiology and Molecular Biology, Key Laboratory of Oral Medicine, Guangzhou Institute of Oral Disease, Stomatology Hospital of Guangzhou Medical University, Guangzhou, China
| | - Ji-Hye Kim
- Department of Dental Hygiene, Jeonju Kijeon College, Jeonju, South Korea
| | - Yun-Jung Yoo
- Department of Oral Biology, Oral Science Research Center, Department of Applied Life Science, The Graduate School, BK21 Plus Project, Yonsei University College of Dentistry, Seoul, South Korea
| | - D Scott Merrell
- Department of Microbiology and Immunology, Uniformed Services University of the Health Sciences, Bethesda, MD, United States
| | - Jinmoon Kim
- Department of Oral Biology, Oral Science Research Center, Department of Applied Life Science, The Graduate School, BK21 Plus Project, Yonsei University College of Dentistry, Seoul, South Korea
| | - Jeong-Heon Cha
- Department of Oral Biology, Oral Science Research Center, Department of Applied Life Science, The Graduate School, BK21 Plus Project, Yonsei University College of Dentistry, Seoul, South Korea.,Microbiology and Molecular Biology, Key Laboratory of Oral Medicine, Guangzhou Institute of Oral Disease, Stomatology Hospital of Guangzhou Medical University, Guangzhou, China
| |
Collapse
|
46
|
Yu Y, Wang X, Zhang X, Zhai Y, Lu X, Ma H, Zhu K, Zhao T, Jiao J, Zhao ZA, Li L. ERK inhibition promotes neuroectodermal precursor commitment by blocking self-renewal and primitive streak formation of the epiblast. Stem Cell Res Ther 2018; 9:2. [PMID: 29304842 PMCID: PMC5756365 DOI: 10.1186/s13287-017-0750-8] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2017] [Revised: 12/11/2017] [Accepted: 12/14/2017] [Indexed: 12/27/2022] Open
Abstract
Background Pluripotent stem cells hold great promise for regenerative medicine. However, before clinical application, reproducible protocols for pluripotent stem cell differentiation should be established. Extracellular signal-regulated protein kinase (ERK) signaling plays a central role for the self-renewal of epiblast stem cells (EpiSCs), but its role for subsequent germ layer differentiation is still ambiguous. We proposed that ERK could modulate differentiation of the epiblast. Methods PD0325901 was used to inhibit ERK activation during the differentiation of embryonic stem cells and EpiSCs. Immunofluorescence, western blot analysis, real-time PCR and flow cytometry were used to detect germ layer markers and pathway activation. Results We demonstrate that the ERK phosphorylation level is lower in neuroectoderm of mouse E7.5 embryos than that in the primitive streak. ERK inhibition results in neural lineage commitment of epiblast. Mechanistically, PD0325901 abrogates the expression of primitive streak markers by β-catenin retention in the cytoplasm, and inhibits the expression of OCT4 and NANOG during EpiSC differentiation. Thus, EpiSCs differentiate into neuroectodermal lineage efficiently under PD0325901 treatment. These results suggest that neuroectoderm differentiation does not require extrinsic signals, supporting the default differentiation of neural lineage. Conclusions We report that a single ERK inhibitor, PD0325901, can specify epiblasts and EpiSCs into neural-like cells, providing an efficient strategy for neural differentiation. Electronic supplementary material The online version of this article (doi:10.1186/s13287-017-0750-8) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Yang Yu
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, 1 Beichen West Road, Chaoyang District, Beijing, 100101, China.,University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Xiaoxiao Wang
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, 1 Beichen West Road, Chaoyang District, Beijing, 100101, China.,University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Xiaoxin Zhang
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, 1 Beichen West Road, Chaoyang District, Beijing, 100101, China
| | - Yanhua Zhai
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, 1 Beichen West Road, Chaoyang District, Beijing, 100101, China
| | - Xukun Lu
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, 1 Beichen West Road, Chaoyang District, Beijing, 100101, China.,University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Haixia Ma
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, 1 Beichen West Road, Chaoyang District, Beijing, 100101, China.,University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Kai Zhu
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, 1 Beichen West Road, Chaoyang District, Beijing, 100101, China.,University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Tongbiao Zhao
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, 1 Beichen West Road, Chaoyang District, Beijing, 100101, China
| | - Jianwei Jiao
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, 1 Beichen West Road, Chaoyang District, Beijing, 100101, China
| | - Zhen-Ao Zhao
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, 1 Beichen West Road, Chaoyang District, Beijing, 100101, China. .,Department of Cardiovascular Surgery of the First Affiliated Hospital & Institute for Cardiovascular Science, Soochow University, 708 Renmin Rd, Suzhou, Jiangsu, 215007, China.
| | - Lei Li
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, 1 Beichen West Road, Chaoyang District, Beijing, 100101, China. .,University of Chinese Academy of Sciences, Beijing, 100049, China.
| |
Collapse
|
47
|
Yin C, Fufa T, Chandrasekar G, Aeluri M, Zaky V, Abdelhady S, Rodríguez AB, Jakobsson J, Varnoosfaderani FS, Mahalingam J, Liu J, Larsson O, Hovatta O, Gaunitz F, Göndör A, Andäng M, Kitambi SS. Phenotypic Screen Identifies a Small Molecule Modulating ERK2 and Promoting Stem Cell Proliferation. Front Pharmacol 2017; 8:726. [PMID: 29114221 PMCID: PMC5660848 DOI: 10.3389/fphar.2017.00726] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2017] [Accepted: 09/27/2017] [Indexed: 11/20/2022] Open
Abstract
Stem cells display a fundamentally different mechanism of proliferation control when compared to somatic cells. Uncovering these mechanisms would maximize the impact in drug discovery with a higher translational applicability. The unbiased approach used in phenotype-based drug discovery (PDD) programs can offer a unique opportunity to identify such novel biological phenomenon. Here, we describe an integrated phenotypic screening approach, employing a combination of in vitro and in vivo PDD models to identify a small molecule increasing stem cell proliferation. We demonstrate that a combination of both in vitro and in vivo screening models improves hit identification and reproducibility of effects across various PDD models. Using cell viability and colony size phenotype measurement we characterize the structure activity relationship of the lead molecule, and identify that the small molecule inhibits phosphorylation of ERK2 and promotes stem cell proliferation. This study demonstrates a PDD approach that employs combinatorial models to identify compounds promoting stem cell proliferation.
Collapse
Affiliation(s)
- Chang Yin
- Department of Microbiology, Tumor and Cell Biology, Karolinska Institutet, Stockholm, Sweden.,Department of Oncology-Pathology, Karolinska Institutet, Stockholm, Sweden
| | - Temesgen Fufa
- Department of Microbiology, Tumor and Cell Biology, Karolinska Institutet, Stockholm, Sweden.,Klinik und Poliklinik für Neurochirurgie, Universitätsklinikum Leipzig, Leipzig, Germany
| | - Gayathri Chandrasekar
- Department of Microbiology, Tumor and Cell Biology, Karolinska Institutet, Stockholm, Sweden.,Department of Biosciences and Nutrition, Karolinska Institutet, Stockholm, Sweden
| | - Madhu Aeluri
- Department of Microbiology, Tumor and Cell Biology, Karolinska Institutet, Stockholm, Sweden.,Dr. Reddy's Institute of Life Sciences, University of Hyderabad Campus, Hyderabad, India
| | - Verina Zaky
- Department of Microbiology, Tumor and Cell Biology, Karolinska Institutet, Stockholm, Sweden
| | - Shaimaa Abdelhady
- Department of Physiology and Pharmacology, Karolinska Institutet, Stockholm, Sweden
| | - Antonio B Rodríguez
- Department of Medical Biochemistry and Biophysics, Karolinska Institutet, Stockholm, Sweden
| | - Johan Jakobsson
- Department of Medical Biochemistry and Biophysics, Karolinska Institutet, Stockholm, Sweden
| | | | | | - Jianping Liu
- Department of Medical Biochemistry and Biophysics, Karolinska Institutet, Stockholm, Sweden
| | - Olle Larsson
- Department of Oncology-Pathology, Karolinska Institutet, Stockholm, Sweden
| | - Outi Hovatta
- Division of Obstetrics and Gynecology, Department of Clinical Sciences, Intervention and Technology, Karolinska Institutet, Karolinska University Hospital, Stockholm, Sweden
| | - Frank Gaunitz
- Klinik und Poliklinik für Neurochirurgie, Universitätsklinikum Leipzig, Leipzig, Germany
| | - Anita Göndör
- Department of Oncology-Pathology, Karolinska Institutet, Stockholm, Sweden
| | - Michael Andäng
- Dr. Reddy's Institute of Life Sciences, University of Hyderabad Campus, Hyderabad, India
| | - Satish S Kitambi
- Department of Microbiology, Tumor and Cell Biology, Karolinska Institutet, Stockholm, Sweden
| |
Collapse
|
48
|
BCL2 induced by LAMTOR3/MAPK is a druggable target of chemoradioresistance in mesenchymal lung cancer. Cancer Lett 2017; 403:48-58. [DOI: 10.1016/j.canlet.2017.05.019] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2017] [Revised: 04/24/2017] [Accepted: 05/24/2017] [Indexed: 11/22/2022]
|
49
|
Wang H, Zhang Y, Yun H, Chen S, Chen Y, Liu Z. ERK expression and its correlation with STAT1 in esophageal squamous cell carcinoma. Oncotarget 2017; 8:45249-45258. [PMID: 28431406 PMCID: PMC5542183 DOI: 10.18632/oncotarget.16902] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2016] [Accepted: 03/27/2017] [Indexed: 02/05/2023] Open
Abstract
BACKGROUND Esophageal squamous cell carcinoma is one of leading causes of cancer-related deaths in Chaoshan region a high-risk region for esophageal cancer. Extracellular regulated protein kinases (ERK) usually play an important role in cell proliferation and differentiation. However, accumulating evidence has shown that the ERK was aberrantly expressed in cancers and correlated with STAT1 depression. RESULTS The activated ERK downregulates STAT1 expression in ESCC cell lines and U0126 increases expression of STAT1. Our immunohistochemistry result also confirms that the expression of ERK inversely correlated with that of STAT1 in ESCC tumors. In addition, a significantly higher expression of ERK/p-ERK was found in ESCC tissues in comparison with case-matched normal esophageal tissues (p < 0.05). Moreover, the immunohistochemical analysis demonstrated that ERK expression was paralleled with the differentiation and clinical stage. In 74 patients with follow-up data, those with ERKlow tumors survived significantly longer than those with ERKhigh tumors (p = 0.04); patients with ERKlow/STAT1high tumors had the longest survival (p = 0.001). MATERIALS AND METHODS To investigate whether ERK can mediated STAT1 expression in ESCC, we used the MEK plasmid and U0126, a MEK inhibitor, to treat the cell. To further confirm our in-vitro study, we detected the ERK, p-ERK and STAT1 expression in 131 ESCC cases and 22 case-matched normal esophageal tissues adjacent to the tumors specimens. CONCLUSIONS These findings provide pathological evidence that ERK/p-ERK is negatively correlated with STAT1 in ESCC. Our data suggests that inhibition of ERK and/or restoration of STAT1 expression maybe useful therapeutic strategies for ESCC.
Collapse
Affiliation(s)
- Hu Wang
- Department of Orthopaedics, First Affiliated Hospital of Shantou University Medical College, Shantou, Guangdong, China
| | - Ying Zhang
- Department of Pathology, Shantou University Medical College, Shantou, Guangdong Province, China
| | - Hailong Yun
- Department of Pathology, Shantou University Medical College, Shantou, Guangdong Province, China
| | - Shubiao Chen
- Department of Orthopaedics, First Affiliated Hospital of Shantou University Medical College, Shantou, Guangdong, China
| | - Yelong Chen
- Department of Orthopaedics, First Affiliated Hospital of Shantou University Medical College, Shantou, Guangdong, China
| | - Zhaoyong Liu
- Department of Orthopaedics, First Affiliated Hospital of Shantou University Medical College, Shantou, Guangdong, China
| |
Collapse
|
50
|
Ning Y, Xu M, Cao X, Chen X, Luo X. Inactivation of AKT, ERK and NF-κB by genistein derivative, 7-difluoromethoxyl-5,4′-di-n-octylygenistein, reduces ovarian carcinoma oncogenicity. Oncol Rep 2017. [DOI: 10.3892/or.2017.5709] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
|