1
|
Hu Q, Zhang T, He H, Pu F, Zhang R, Li L, Hu J, Bai L, Han C, Wang J, Liu H. Impacts of longitudinal water curtain cooling system on transcriptome-related immunity in ducks. BMC Genomics 2024; 25:333. [PMID: 38570739 PMCID: PMC10988813 DOI: 10.1186/s12864-024-10179-4] [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: 12/18/2023] [Accepted: 03/04/2024] [Indexed: 04/05/2024] Open
Abstract
BACKGROUND The closed poultry houses integrated with a longitudinal water curtain cooling system (LWCCS) are widely used in modern poultry production. This study showed the variations in environmental conditions in closed houses integrated with a longitudinal water curtain cooling system. We evaluated the influence of different environmental conditions on duck growth performance and the transcriptome changes of immune organs, including the bursa of Fabricius and the spleen. RESULT This study investigated the slaughter indicators and immune organ transcriptomes of 52-day-old Cherry Valley ducks by analyzing the LWCC at different locations (water curtain end, middle position, and fan cooling end). The results showed that the cooling effect of the LWCCS was more evident from 10:00 a.m. -14:00. And from the water curtain end to the fan cooling end, the hourly average temperature differently decreased by 0.310℃, 0.450℃, 0.480℃, 0.520℃, and 0.410℃, respectively (P < 0.05). The daily and hourly average relative humidity decreased from the water curtain end to the fan cooling end, dropping by 7.500% and 8.200%, respectively (P < 0.01). We also observed differences in production performance, such as dressing weight, half-eviscerated weight, skin fat rate, and percentage of abdominal fat (P < 0.01), which may have been caused by environmental conditions. RNA-sequencing (RNA-seq) revealed 211 and 279 differentially expressed genes (DEGs) in the ducks' bursa of Fabricius and spleen compared between the water curtain end and fan cooling end, respectively. The Gene Ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG) analysis of the two organs showed the DEGs were mainly enriched in cytokine-cytokine receptor interaction, integral component of membrane, Retinoic acid-inducible gene I (RIG-I)-like receptors (RLRs) signaling pathway, etc. Our results implied that full-closed poultry houses integrated with LWCCS could potentially alter micro-environments (water curtain vs. fan cooling), resulting in ducks experiencing various stressful situations that eventually affect their immunity and production performance. CONCLUSION In this study, our results indicated that uneven distributions of longitudinal environmental factors caused by LWCCS would affect the dressed weight, breast muscle weight, skin fat rate, and other product performance. Moreover, the expression of immune-related genes in the spleen and bursa of ducks could be affected by the LWCCS. This provides a new reference to optimize the use of LWCCS in conjunction with close duck houses in practical production.
Collapse
Affiliation(s)
- Qian Hu
- Farm Animal Genetic Resources Exploration and Innovation Key Laboratory of Sichuan Province, Sichuan Agricultural University, 611130, Chengdu, Sichuan, P.R. China
- Key Laboratory of Livestock and Poultry Multi-omics, Ministry of Agriculture and Rural Affairs, 611130, Chengdu, Wenjiang District, Sichuan, P.R. China
- National Key Laboratory for Swine and Poultry Breeding, Wuhan, P.R. China
| | - Tao Zhang
- Farm Animal Genetic Resources Exploration and Innovation Key Laboratory of Sichuan Province, Sichuan Agricultural University, 611130, Chengdu, Sichuan, P.R. China
- Key Laboratory of Livestock and Poultry Multi-omics, Ministry of Agriculture and Rural Affairs, 611130, Chengdu, Wenjiang District, Sichuan, P.R. China
- National Key Laboratory for Swine and Poultry Breeding, Wuhan, P.R. China
| | - Hua He
- Farm Animal Genetic Resources Exploration and Innovation Key Laboratory of Sichuan Province, Sichuan Agricultural University, 611130, Chengdu, Sichuan, P.R. China
- Key Laboratory of Livestock and Poultry Multi-omics, Ministry of Agriculture and Rural Affairs, 611130, Chengdu, Wenjiang District, Sichuan, P.R. China
- National Key Laboratory for Swine and Poultry Breeding, Wuhan, P.R. China
| | - Fajun Pu
- Farm Animal Genetic Resources Exploration and Innovation Key Laboratory of Sichuan Province, Sichuan Agricultural University, 611130, Chengdu, Sichuan, P.R. China
- Key Laboratory of Livestock and Poultry Multi-omics, Ministry of Agriculture and Rural Affairs, 611130, Chengdu, Wenjiang District, Sichuan, P.R. China
- National Key Laboratory for Swine and Poultry Breeding, Wuhan, P.R. China
| | - Rongping Zhang
- Farm Animal Genetic Resources Exploration and Innovation Key Laboratory of Sichuan Province, Sichuan Agricultural University, 611130, Chengdu, Sichuan, P.R. China
- Key Laboratory of Livestock and Poultry Multi-omics, Ministry of Agriculture and Rural Affairs, 611130, Chengdu, Wenjiang District, Sichuan, P.R. China
- National Key Laboratory for Swine and Poultry Breeding, Wuhan, P.R. China
| | - Liang Li
- Farm Animal Genetic Resources Exploration and Innovation Key Laboratory of Sichuan Province, Sichuan Agricultural University, 611130, Chengdu, Sichuan, P.R. China
- Key Laboratory of Livestock and Poultry Multi-omics, Ministry of Agriculture and Rural Affairs, 611130, Chengdu, Wenjiang District, Sichuan, P.R. China
- National Key Laboratory for Swine and Poultry Breeding, Wuhan, P.R. China
| | - Jiwei Hu
- Farm Animal Genetic Resources Exploration and Innovation Key Laboratory of Sichuan Province, Sichuan Agricultural University, 611130, Chengdu, Sichuan, P.R. China
- Key Laboratory of Livestock and Poultry Multi-omics, Ministry of Agriculture and Rural Affairs, 611130, Chengdu, Wenjiang District, Sichuan, P.R. China
- National Key Laboratory for Swine and Poultry Breeding, Wuhan, P.R. China
| | - Lili Bai
- Farm Animal Genetic Resources Exploration and Innovation Key Laboratory of Sichuan Province, Sichuan Agricultural University, 611130, Chengdu, Sichuan, P.R. China
- Key Laboratory of Livestock and Poultry Multi-omics, Ministry of Agriculture and Rural Affairs, 611130, Chengdu, Wenjiang District, Sichuan, P.R. China
- National Key Laboratory for Swine and Poultry Breeding, Wuhan, P.R. China
| | - Chunchun Han
- Farm Animal Genetic Resources Exploration and Innovation Key Laboratory of Sichuan Province, Sichuan Agricultural University, 611130, Chengdu, Sichuan, P.R. China
- Key Laboratory of Livestock and Poultry Multi-omics, Ministry of Agriculture and Rural Affairs, 611130, Chengdu, Wenjiang District, Sichuan, P.R. China
- National Key Laboratory for Swine and Poultry Breeding, Wuhan, P.R. China
| | - Jiwen Wang
- Farm Animal Genetic Resources Exploration and Innovation Key Laboratory of Sichuan Province, Sichuan Agricultural University, 611130, Chengdu, Sichuan, P.R. China
- Key Laboratory of Livestock and Poultry Multi-omics, Ministry of Agriculture and Rural Affairs, 611130, Chengdu, Wenjiang District, Sichuan, P.R. China
- National Key Laboratory for Swine and Poultry Breeding, Wuhan, P.R. China
| | - Hehe Liu
- Farm Animal Genetic Resources Exploration and Innovation Key Laboratory of Sichuan Province, Sichuan Agricultural University, 611130, Chengdu, Sichuan, P.R. China.
- Key Laboratory of Livestock and Poultry Multi-omics, Ministry of Agriculture and Rural Affairs, 611130, Chengdu, Wenjiang District, Sichuan, P.R. China.
- National Key Laboratory for Swine and Poultry Breeding, Wuhan, P.R. China.
| |
Collapse
|
2
|
Olomu IN, Hoang V, Madhukar BV. Low levels of nicotine and cotinine but not benzo[a]pyrene induce human trophoblast cell proliferation. Reprod Toxicol 2024; 125:108572. [PMID: 38453095 DOI: 10.1016/j.reprotox.2024.108572] [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: 09/21/2023] [Revised: 01/22/2024] [Accepted: 03/04/2024] [Indexed: 03/09/2024]
Abstract
E-cigarettes use constitutes a source of thirdhand nicotine exposure. The increasing use of electronic cigarettes in homes and public places increases the risk of exposure of pregnant women to thirdhand nicotine. The effects of exposure of pregnant women to very low levels of nicotine have not been studied in humans but detrimental in experimental animals. The objective of this study is to investigate the effect of nanomolar concentrations of nicotine and its metabolite cotinine on the proliferation of JEG-3, a human trophoblast cell line. We also studied the proliferative effect of nanomolar concentrations of benzo[a]pyrene (B[a]P), a polycyclic hydrocarbon in tobacco smoke, for comparison. We treated JEG-3 cells in culture with nanomolar concentrations of nicotine, cotinine, and B[a]P. Their effect on cell proliferation was determined, relative to untreated cells, by MTT assay. Western blotting was used to assess the mitogenic signaling pathways affected by nicotine and cotinine. In contrast to the inhibitory effects reported with higher concentrations, we showed that nanomolar concentrations of nicotine and cotinine resulted in significant JEG-3 cell proliferation and a rapid but transient increase in levels of phosphorylated ERK and AKT, but not STAT3. Biphasic, non-monotonic effect on cell growth is characteristic of endocrine disruptive chemicals like nicotine. The mitogenic effects of nicotine and cotinine potentially contribute to increased villous epithelial thickness, seen in placentas of some smoking mothers. This increases the diffusion distance for oxygen and nutrients between mother and fetus, contributing to intrauterine growth restriction in infants of smoking mothers.
Collapse
Affiliation(s)
- I Nicholas Olomu
- Department of Pediatrics & Human Development, Michigan State University, East Lansing, MI, USA; Division of Neonatology, Michigan State University, East Lansing, MI, USA.
| | - Vanessa Hoang
- Department of Pediatrics & Human Development, Michigan State University, East Lansing, MI, USA
| | - Burra V Madhukar
- Department of Pediatrics & Human Development, Michigan State University, East Lansing, MI, USA
| |
Collapse
|
3
|
Gui Y, Qian X, Ding Y, Chen Q, Fangyu Ye, Ye Y, Hou Y, Yu J, Zhao L. c-Fos regulated by TMPO/ERK axis promotes 5-FU resistance via inducing NANOG transcription in colon cancer. Cell Death Dis 2024; 15:61. [PMID: 38233377 PMCID: PMC10794174 DOI: 10.1038/s41419-024-06451-w] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2023] [Revised: 01/05/2024] [Accepted: 01/08/2024] [Indexed: 01/19/2024]
Abstract
Acquired drug resistance is one of the most common limitations for the clinical response of colon cancer to 5-Fluorouracil (5-FU)-based chemotherapy. The relevant molecular mechanisms might be diversity, but still not be elucidated clearly. In this study, we aimed to investigate the potential mechanisms of c-Fos, a subfamily of activator protein-1, in 5-FU chemoresistance. We determined that phosphorylated c-Fos promoted colon cancer cells resistance to 5-FU by facilitating the cancer stemness. Mechanically, 5-FU treatment induced autolysosome-dependent degradation of TMPO, which subsequently triggered ERK-mediated phosphorylation of c-Fos. Additionally, c-Fos was found to bind to the promoter of NANOG and phosphorylation of c-Fos at Ser 374 was required for its regulation of NANOG expression. NANOG ablation impaired c-Fos/p-c-Fos induced 5-FU resistance and stemness. Taken together, these findings revealed that TMPO-mediated phosphorylation of c-Fos conferred 5-FU resistance by regulating NANOG expression and promoting cell stemness in colon cancer cells. c-Fos could be as a therapeutic target for colon cancer.
Collapse
Affiliation(s)
- Yanping Gui
- Public Experimental Platform, China Pharmaceutical University, Nanjing, 211198, China
| | - Xiaoping Qian
- Suzhou Hospital, Affiliated Hospital of Medical School, Nanjing University, Suzhou, 215153, China
| | - Youxiang Ding
- Department of Pathology, Nanjing Drum Tower Hospital, Affiliated to Medical College of Nanjing University, Nanjing, 210008, China
| | - Qianqian Chen
- Public Experimental Platform, China Pharmaceutical University, Nanjing, 211198, China
| | - Fangyu Ye
- Public Experimental Platform, China Pharmaceutical University, Nanjing, 211198, China
| | - Yuting Ye
- Public Experimental Platform, China Pharmaceutical University, Nanjing, 211198, China
| | - Yingjian Hou
- Public Experimental Platform, China Pharmaceutical University, Nanjing, 211198, China
| | - Jun Yu
- Jiangsu Cancer Hospital, Nanjing, 210009, China
| | - Li Zhao
- Public Experimental Platform, China Pharmaceutical University, Nanjing, 211198, China.
| |
Collapse
|
4
|
Pham T, Najy AJ, Kim HRC. E3 ligase HUWE1 promotes PDGF D-mediated osteoblastic differentiation of mesenchymal stem cells by effecting polyubiquitination of β-PDGFR. J Biol Chem 2022; 298:101981. [PMID: 35472332 PMCID: PMC9133640 DOI: 10.1016/j.jbc.2022.101981] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2021] [Revised: 03/22/2022] [Accepted: 03/28/2022] [Indexed: 11/25/2022] Open
Abstract
Mesenchymal stem cells (MSCs) are adult stem cell populations and exhibit great potential in regenerative medicine and oncology. Platelet-derived growth factors (PDGFs) are well known to regulate MSC biology through their chemotactic and mitogenic properties. However, their direct roles in the regulation of MSC lineage commitment are unclear. Here, we show that PDGF D promotes the differentiation of human bone marrow mesenchymal stem cells (hBMSCs) into osteoblasts and inhibits hBMSC differentiation into adipocytes. We demonstrate that PDGF D-induced β-actin expression and polymerization are essential for mediating this differential regulation of osteoblastogenesis and adipogenesis. Interestingly, we found that PDGF D induces massive upward molecular weight shifts of its cognate receptor, PDGF receptor beta (β-PDGFR) in hBMSCs, which was not observed in fibroblasts. Proteomic analysis indicated that the E3 ubiquitin ligase HECT, UBA, and WWE domain–containing protein 1 (HUWE1) associates with the PDGF D-activated β-PDGFR signaling complex in hBMSCs, resulting in β-PDGFR polyubiquitination. In contrast to the well-known role of ubiquitin in protein degradation, we provide evidence that HUWE1-mediated β-PDGFR polyubiquitination delays β-PDGFR internalization and degradation, thereby prolonging AKT signaling. Finally, we demonstrate that HUWE1-regulated β-PDGFR signaling is essential for osteoblastic differentiation of hBMSCs, while being dispensable for PDGF D-induced hBMSC migration and proliferation as well as PDGF D-mediated inhibition of hBMSC differentiation into adipocytes. Taken together, our findings provide novel insights into the molecular mechanism by which PDGF D regulates the commitment of hBMSCs into the osteoblastic lineage.
Collapse
Affiliation(s)
- Tri Pham
- Department of Pathology, Wayne State University School of Medicine and the Barbara Ann Karmanos Cancer Institute, Detroit, MI, 48201 USA
| | - Abdo J Najy
- Department of Pathology, Wayne State University School of Medicine and the Barbara Ann Karmanos Cancer Institute, Detroit, MI, 48201 USA
| | - Hyeong-Reh C Kim
- Department of Pathology, Wayne State University School of Medicine and the Barbara Ann Karmanos Cancer Institute, Detroit, MI, 48201 USA.
| |
Collapse
|
5
|
Dai X, Murakami M, Shiraishi K, Muto J, Tohyama M, Mori H, Utsunomiya R, Sayama K. EGFR ligands synergistically increase IL-17A-induced expression of psoriasis signature genes in human keratinocytes via IκBζ and Bcl3. Eur J Immunol 2022; 52:994-1005. [PMID: 35411943 DOI: 10.1002/eji.202149706] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2021] [Revised: 04/11/2022] [Accepted: 04/11/2022] [Indexed: 11/07/2022]
Abstract
Various epidermal growth factor receptor (EGFR) ligands are highly expressed in the epidermis of psoriasis lesions, and abnormal EGFR activation appears to be involved in the pathogenesis of psoriasis. However, how EGFR signaling contributes to the development of psoriasis is unclear. Interleukin (IL)-17A, a critical effector of the IL-23/IL-17A pathway, increases the expression of psoriasis signature genes in keratinocytes and plays an essential role in the pathogenesis of psoriasis by inducing IκBζ, a critical transcriptional regulator in psoriasis. In this study, we stimulated primary human keratinocytes with IL-17A and various EGFR ligands to investigate whether EGFR ligands regulate the expression of psoriasis signature genes. In cultured normal human keratinocytes and a living skin equivalent, EGFR ligands did not induce psoriasis-related gene expression, but significantly enhanced the IL-17A-mediated induction of various psoriasis signature genes, including antimicrobial peptides, cytokines, and chemokines. This was dependent on an EGFR activation-mediated synergistic increase in IL-17A-induced IκBζ expression and was partially mediated by the EGFR-dependent upregulation of Bcl3. Therefore, EGFR ligands can act as synergistic agents of IL-17A signaling by stimulating the epidermal production of psoriasis signature genes in psoriasis lesions. This study reveals a potential mechanism by which EGFR signaling contributes to the pathogenesis of psoriasis. This article is protected by copyright. All rights reserved.
Collapse
Affiliation(s)
- Xiuju Dai
- Department of Dermatology, Ehime University Graduate School of Medicine, Ehime, Japan
| | - Masamoto Murakami
- Department of Dermatology, Ehime University Graduate School of Medicine, Ehime, Japan
| | - Ken Shiraishi
- Department of Dermatology, Ehime University Graduate School of Medicine, Ehime, Japan
| | - Jun Muto
- Department of Dermatology, Ehime University Graduate School of Medicine, Ehime, Japan
| | - Mikiko Tohyama
- Department of Dermatology, Ehime University Graduate School of Medicine, Ehime, Japan.,Department of Dermatology, National Hospital Organization Shikoku Cancer Center, Matsuyama, Ehime, Japan
| | - Hideki Mori
- Department of Dermatology, Ehime University Graduate School of Medicine, Ehime, Japan
| | - Ryo Utsunomiya
- Department of Dermatology, Ehime University Graduate School of Medicine, Ehime, Japan
| | - Koji Sayama
- Department of Dermatology, Ehime University Graduate School of Medicine, Ehime, Japan
| |
Collapse
|
6
|
Cho B, Yoo SJ, Kim SY, Lee CH, Lee YI, Lee SR, Moon C. Second-generation non-hematopoietic erythropoietin-derived peptide for neuroprotection. Redox Biol 2021; 49:102223. [PMID: 34953452 PMCID: PMC8715119 DOI: 10.1016/j.redox.2021.102223] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2021] [Accepted: 12/20/2021] [Indexed: 12/25/2022] Open
Abstract
Erythropoietin (EPO) is a well-known erythropoietic cytokine having a tissue-protective effect in various tissues against hypoxic stress, including the brain. Thus, its recombinants may function as neuroprotective compounds. However, despite considerable neuroprotective effects, the EPO-based therapeutic approach has side effects, including hyper-erythropoietic and tumorigenic effects. Therefore, some modified forms and derivatives of EPO have been proposed to minimize the side effects. In this study, we generated divergently modified new peptide analogs derived from helix C of EPO, with several amino acid replacements that interact with erythropoietin receptors (EPORs). This modification resulted in unique binding potency to EPOR. Unlike recombinant EPO, among the peptides, ML1-h3 exhibited a potent neuroprotective effect against oxidative stress without additional induction of cell-proliferation, owing to a differential activating mode of EPOR signaling. Furthermore, it inhibited neuronal death and brain injury under hypoxic stress in vitro and in an in vivo ischemic brain injury model. Therefore, the divergent modification of EPO-derivatives for affinity to EPOR could provide a basis for a more advanced and optimal neuroprotective strategy. Short peptides derived from helix C of EPO have a neuroprotective effect. Divergent modification of EPO-derived peptides has a differential affinity to EPOR. ML1 and its analogs have differential cell protective and proliferative effects. ML1-h3 protects neurons by suppressing in vitro oxidative stress. ML1-h3 mitigates brain injury in the in vivo mouse ischemic model without hematopoietic effect.
Collapse
Affiliation(s)
- Bongki Cho
- Department of Brain & Cognitive Sciences, Graduate School, DGIST, Daegu, 42988, South Korea; Convergence Research Advanced Centre for Olfaction, DGIST, Daegu, 42988, South Korea; Division of Biotechnology, DGIST, Daegu, 42988, South Korea
| | - Seung-Jun Yoo
- Department of Brain & Cognitive Sciences, Graduate School, DGIST, Daegu, 42988, South Korea; Convergence Research Advanced Centre for Olfaction, DGIST, Daegu, 42988, South Korea
| | - So Yeon Kim
- Department of Brain & Cognitive Sciences, Graduate School, DGIST, Daegu, 42988, South Korea; Convergence Research Advanced Centre for Olfaction, DGIST, Daegu, 42988, South Korea
| | - Chang-Hun Lee
- Department of New Biology, DGIST, Daegu, 42988, South Korea; New Biology Research Center, DGIST, Daegu, 42988, South Korea
| | - Yun-Il Lee
- Division of Biotechnology, DGIST, Daegu, 42988, South Korea
| | - Seong-Ryong Lee
- Department of Pharmacology and ODR Center, Brain Research Institute, School of Medicine, Keimyung University, Daegu, 42601, South Korea.
| | - Cheil Moon
- Department of Brain & Cognitive Sciences, Graduate School, DGIST, Daegu, 42988, South Korea; Convergence Research Advanced Centre for Olfaction, DGIST, Daegu, 42988, South Korea.
| |
Collapse
|
7
|
Nguyen HT, Najih M, Martin LJ. The AP-1 family of transcription factors are important regulators of gene expression within Leydig cells. Endocrine 2021; 74:498-507. [PMID: 34599696 DOI: 10.1007/s12020-021-02888-7] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/22/2021] [Accepted: 09/16/2021] [Indexed: 10/20/2022]
Abstract
PURPOSE Members of the AP-1 family of transcription factors are immediate early genes being modulated by different extracellular signals. The aim of this review is to highlight the important roles of AP-1 members in transcriptional regulation of genes important for testicular Leydig cell function and male testosterone production. METHODS A search of the relevant literature was performed in Google Scholar and NCBI Pubmed for AP-1 members and Leydig cells. Additional information was accessed from references of relevant articles. Only primary data from original peer-reviewed articles was considered for this review. RESULTS Different signaling pathways important for Leydig cells' functions are involved in the regulation of the activity of AP-1 members. These transcription factors participate in the regulation of genes related to different biological processes important for Leydig cells. CONCLUSIONS We conclude that members of the AP-1 family of transcription factors play critical roles in the regulation of Leydig cell proliferation, steroidogenesis, and cell-to-cell communication.
Collapse
Affiliation(s)
- Ha Tuyen Nguyen
- Biology Department, Université de Moncton, Moncton, NB, E1A 3E9, Canada
| | - Mustapha Najih
- Biology Department, Université de Moncton, Moncton, NB, E1A 3E9, Canada
| | - Luc J Martin
- Biology Department, Université de Moncton, Moncton, NB, E1A 3E9, Canada.
| |
Collapse
|
8
|
Rysenkova KD, Klimovich PS, Shmakova AA, Karagyaur MN, Ivanova KA, Aleksandrushkina NA, Tkachuk VA, Rubina KA, Semina EV. Urokinase receptor deficiency results in EGFR-mediated failure to transmit signals for cell survival and neurite formation in mouse neuroblastoma cells. Cell Signal 2020; 75:109741. [PMID: 32822758 DOI: 10.1016/j.cellsig.2020.109741] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2020] [Revised: 08/14/2020] [Accepted: 08/14/2020] [Indexed: 12/26/2022]
Abstract
Urokinase-type plasminogen activator uPA and its receptor (uPAR) are the central players in extracellular matrix proteolysis, which facilitates cancer invasion and metastasis. EGFR is one of the important components of uPAR interactome. uPAR/EGFR interaction controls signaling pathways that regulate cell survival, proliferation and migration. We have previously established that uPA binding to uPAR stimulates neurite elongation in neuroblastoma cells, while blocking uPA/uPAR interaction induces neurite branching and new neurite formation. Here we demonstrate that blocking the uPA binding to uPAR with anti-uPAR antibody decreases the level of pEGFR and its downstream pERK1/2, but does increase phosphorylation of Akt, p38 and c-Src Since long-term uPAR blocking results in a severe DNA damage, accompanied by PARP-1 proteolysis and Neuro2a cell death, we surmise that Akt, p38 and c-Src activation transmits a pro-apoptotic signal, rather than a survival. Serum deprivation resulting in enhanced neuritogenesis is accompanied by an upregulated uPAR mRNA expression, while EGFR mRNA remains unchanged. EGFR activation by EGF stimulates neurite growth only in uPAR-overexpressing cells but not in control or uPAR-deficient cells. In addition, AG1478-mediated inhibition of EGFR activity impedes neurite growth in control and uPAR-deficient cells, but not in uPAR-overexpressing cells. Altogether these data implicate uPAR as an important regulator of EGFR and ERK1/2 signaling, representing a novel mechanism which implicates urokinase system in neuroblastoma cell survival and differentiation.
Collapse
Affiliation(s)
- K D Rysenkova
- Laboratory of Molecular Endocrinology, Institute of Experimental Cardiology, Federal State Budgetary Organization National Cardiology Research Center Ministry of Health of the Russian Federation, Moscow, Russia; Laboratory of Gene and Cell Technologies, Faculty of Medicine, Lomonosov Moscow State University, Moscow, Russia
| | - P S Klimovich
- Laboratory of Molecular Endocrinology, Institute of Experimental Cardiology, Federal State Budgetary Organization National Cardiology Research Center Ministry of Health of the Russian Federation, Moscow, Russia
| | - A A Shmakova
- Laboratory of Molecular Endocrinology, Institute of Experimental Cardiology, Federal State Budgetary Organization National Cardiology Research Center Ministry of Health of the Russian Federation, Moscow, Russia
| | - M N Karagyaur
- Institute of Regenerative Medicine, Medical Research and Education Center, Lomonosov Moscow State University, Moscow, Russian Federation
| | - K A Ivanova
- Laboratory of Gene and Cell Technologies, Faculty of Medicine, Lomonosov Moscow State University, Moscow, Russia
| | - N A Aleksandrushkina
- Laboratory of Gene and Cell Technologies, Faculty of Medicine, Lomonosov Moscow State University, Moscow, Russia; Institute of Regenerative Medicine, Medical Research and Education Center, Lomonosov Moscow State University, Moscow, Russian Federation
| | - V A Tkachuk
- Laboratory of Molecular Endocrinology, Institute of Experimental Cardiology, Federal State Budgetary Organization National Cardiology Research Center Ministry of Health of the Russian Federation, Moscow, Russia; Laboratory of Gene and Cell Technologies, Faculty of Medicine, Lomonosov Moscow State University, Moscow, Russia
| | - K A Rubina
- Laboratory of Morphogenesis and Tissue Reparation, Faculty of Medicine, Lomonosov Moscow State University, Moscow, Russia.
| | - E V Semina
- Laboratory of Molecular Endocrinology, Institute of Experimental Cardiology, Federal State Budgetary Organization National Cardiology Research Center Ministry of Health of the Russian Federation, Moscow, Russia; Laboratory of Gene and Cell Technologies, Faculty of Medicine, Lomonosov Moscow State University, Moscow, Russia
| |
Collapse
|
9
|
Zhou H, Wang X, Wang S, Liu C, Fu Q, Qin G, Zhou J, Chen L. Inhibition of Nerve Growth Factor Signaling Alleviates Repeated Dural Stimulation-induced Hyperalgesia in Rats. Neuroscience 2019; 398:252-262. [DOI: 10.1016/j.neuroscience.2018.12.006] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2018] [Revised: 12/03/2018] [Accepted: 12/04/2018] [Indexed: 12/12/2022]
|
10
|
Da Silva MHA, De Souza DB. Current evidence for the involvement of sex steroid receptors and sex hormones in benign prostatic hyperplasia. Res Rep Urol 2019; 11:1-8. [PMID: 30662879 PMCID: PMC6327899 DOI: 10.2147/rru.s155609] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/31/2023] Open
Abstract
Benign prostatic hyperplasia (BPH) is a pathology that affects 50% of men over 50 years of age and 90% of men develop BPH in their eighth decade of life. In 2018, more than 1 billion men will be affected by this disease worldwide. However, the progression of BPH is highly complex and has been debated and studied for approximately four decades. Recent studies indicate that BPH can originate from the alteration of different hormone synthesis pathways, and that it is also linked to the function of hormone receptors. There is a close relationship between the progression of BPH and sexual hormones, such as progesterone, testosterone, dihydrotestosterone, and estrogen. The focus of this study was to characterize the interactions of these hormones and investigate the direct or indirect role of each sex hormone receptor in the progression of BPH. Although several studies have described the effects of these hormones on BPH, no conclusions have been drawn regarding their role in disease progression. Here, we present a literature review on the sexual receptors possibly involved in the progression of BPH.
Collapse
|
11
|
Madeo M, Colbert PL, Vermeer DW, Lucido CT, Cain JT, Vichaya EG, Grossberg AJ, Muirhead D, Rickel AP, Hong Z, Zhao J, Weimer JM, Spanos WC, Lee JH, Dantzer R, Vermeer PD. Cancer exosomes induce tumor innervation. Nat Commun 2018; 9:4284. [PMID: 30327461 PMCID: PMC6191452 DOI: 10.1038/s41467-018-06640-0] [Citation(s) in RCA: 170] [Impact Index Per Article: 28.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2017] [Accepted: 09/18/2018] [Indexed: 12/16/2022] Open
Abstract
Patients with densely innervated tumors suffer with increased metastasis and decreased survival as compared to those with less innervated tumors. We hypothesize that in some tumors, nerves are acquired by a tumor-induced process, called axonogenesis. Here, we use PC12 cells as an in vitro neuronal model, human tumor samples and murine in vivo models to test this hypothesis. When appropriately stimulated, PC12 cells extend processes, called neurites. We show that patient tumors release vesicles, called exosomes, which induce PC12 neurite outgrowth. Using a cancer mouse model, we show that tumors compromised in exosome release are less innervated than controls. Moreover, in vivo pharmacological blockade of exosome release similarly attenuates tumor innervation. We characterize these nerves as sensory in nature and demonstrate that axonogenesis is potentiated by the exosome-packaged axonal guidance molecule, EphrinB1. These findings indicate that tumor released exosomes induce tumor innervation and exosomes containing EphrinB1 potentiate this activity.
Collapse
Affiliation(s)
- Marianna Madeo
- Cancer Biology and Immunotherapies Group, Sanford Research, 2301 East 60th St north, Sioux Falls, SD, 57104, USA
| | - Paul L Colbert
- Cancer Biology and Immunotherapies Group, Sanford Research, 2301 East 60th St north, Sioux Falls, SD, 57104, USA
| | - Daniel W Vermeer
- Cancer Biology and Immunotherapies Group, Sanford Research, 2301 East 60th St north, Sioux Falls, SD, 57104, USA
| | - Christopher T Lucido
- Cancer Biology and Immunotherapies Group, Sanford Research, 2301 East 60th St north, Sioux Falls, SD, 57104, USA
| | - Jacob T Cain
- Pediatrics and Rare Diseases Group, Sanford Research, 2301 East 60th St north, Sioux Falls, SD, 57104, USA
| | - Elisabeth G Vichaya
- Department of Symptom Research, MD Anderson Cancer Center, 1515 Holcombe Blvd, Unit 384, Houston, TX, 77030, USA
| | - Aaron J Grossberg
- Department of Symptom Research, MD Anderson Cancer Center, 1515 Holcombe Blvd, Unit 384, Houston, TX, 77030, USA
- Department of Radiation Medicine, Cancer Early Detection Advanced Research Center, Oregon Health and Science University, 2720 SW Moody Ave KR-CEDR, Portland, OR, 97201, USA
| | - DesiRae Muirhead
- Sanford Health Pathology Clinic, Sanford Health, 1305 West 18th St, Sioux Falls, SD, 57105, USA
| | - Alex P Rickel
- Biomedical Engineering Program, University of South Dakota, 4800 North Career Ave, Sioux Falls, SD, 57107, USA
| | - Zhongkui Hong
- Biomedical Engineering Program, University of South Dakota, 4800 North Career Ave, Sioux Falls, SD, 57107, USA
| | - Jing Zhao
- Population Health Group, Sanford Research, 2301 East 60th St north, Sioux Falls, SD, 57104, USA
| | - Jill M Weimer
- Pediatrics and Rare Diseases Group, Sanford Research, 2301 East 60th St north, Sioux Falls, SD, 57104, USA
| | - William C Spanos
- Cancer Biology and Immunotherapies Group, Sanford Research, 2301 East 60th St north, Sioux Falls, SD, 57104, USA
- Sanford Ears, Nose and Throat, 1310 West 22nd St, Sioux Falls, SD, 57105, USA
| | - John H Lee
- NantKwest, 9920 Jefferson Blvd, Culver City, CA, 90232, USA
| | - Robert Dantzer
- Department of Symptom Research, MD Anderson Cancer Center, 1515 Holcombe Blvd, Unit 384, Houston, TX, 77030, USA
| | - Paola D Vermeer
- Cancer Biology and Immunotherapies Group, Sanford Research, 2301 East 60th St north, Sioux Falls, SD, 57104, USA.
| |
Collapse
|
12
|
Duan L, Hope JM, Guo S, Ong Q, François A, Kaplan L, Scherrer G, Cui B. Optical Activation of TrkA Signaling. ACS Synth Biol 2018; 7:1685-1693. [PMID: 29975841 DOI: 10.1021/acssynbio.8b00126] [Citation(s) in RCA: 33] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
Nerve growth factor/tropomyosin receptor kinase A (NGF/TrkA) signaling plays a key role in neuronal development, function, survival, and growth. The pathway is implicated in neurodegenerative disorders including Alzheimer's disease, chronic pain, inflammation, and cancer. NGF binds the extracellular domain of TrkA, leading to the activation of the receptor's intracellular kinase domain. As TrkA signaling is highly dynamic, mechanistic studies would benefit from a tool with high spatial and temporal resolution. Here we present the design and evaluation of four strategies for light-inducible activation of TrkA in the absence of NGF. Our strategies involve the light-sensitive protein Arabidopsis cryptochrome 2 and its binding partner CIB1. We demonstrate successful recapitulation of native NGF/TrkA functions by optical induction of plasma membrane recruitment and homo-interaction of the intracellular domain of TrkA. This approach activates PI3K/AKT and Raf/ERK signaling pathways, promotes neurite growth in PC12 cells, and supports survival of dorsal root ganglion neurons in the absence of NGF. This ability to activate TrkA using light bestows high spatial and temporal resolution for investigating NGF/TrkA signaling.
Collapse
Affiliation(s)
- Liting Duan
- Department of Chemistry, Stanford University, Stanford, California 94305, United States
| | - Jen M. Hope
- Department of Chemistry, Stanford University, Stanford, California 94305, United States
| | - Shunling Guo
- Department of Chemistry, Stanford University, Stanford, California 94305, United States
| | - Qunxiang Ong
- Department of Chemistry, Stanford University, Stanford, California 94305, United States
| | - Amaury François
- Department of Anesthesiology, Perioperative and Pain Medicine, Department of Molecular and Cellular Physiology, Department of Neurosurgery, Stanford Neurosciences Institute, Stanford University, Palo Alto, California 94304, United States
| | - Luke Kaplan
- Department of Chemistry, Stanford University, Stanford, California 94305, United States
| | - Grégory Scherrer
- Department of Anesthesiology, Perioperative and Pain Medicine, Department of Molecular and Cellular Physiology, Department of Neurosurgery, Stanford Neurosciences Institute, Stanford University, Palo Alto, California 94304, United States
- Robertson Investigator, New York Stem Cell Foundation, New York, New York 10019, United States
| | - Bianxiao Cui
- Department of Chemistry, Stanford University, Stanford, California 94305, United States
| |
Collapse
|
13
|
Ikpegbu E, Basta L, Clements DN, Fleming R, Vincent TL, Buttle DJ, Pitsillides AA, Staines KA, Farquharson C. FGF-2 promotes osteocyte differentiation through increased E11/podoplanin expression. J Cell Physiol 2018; 233:5334-5347. [PMID: 29215722 PMCID: PMC5900964 DOI: 10.1002/jcp.26345] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2017] [Accepted: 11/29/2017] [Indexed: 01/15/2023]
Abstract
E11/podoplanin is critical in the early stages of osteoblast‐to‐osteocyte transitions (osteocytogenesis), however, the upstream events which regulate E11 expression are unknown. The aim of this study was to examine the effects of FGF‐2 on E11‐mediated osteocytogenesis and to reveal the nature of the underlying signaling pathways regulating this process. Exposure of MC3T3 osteoblast‐like cells and murine primary osteoblasts to FGF‐2 (10 ng/ml) increased E11 mRNA and protein expression (p < 0.05) after 4, 6, and 24 hr. FGF‐2 induced changes in E11 expression were also accompanied by significant (p < 0.01) increases in Phex and Dmp1 (osteocyte markers) expression and decreases in Col1a1, Postn, Bglap, and Alpl (osteoblast markers) expression. Immunofluorescent microscopy revealed that FGF‐2 stimulated E11 expression, facilitated the translocation of E11 toward the cell membrane, and subsequently promoted the formation of osteocyte‐like dendrites in MC3T3 and primary osteoblasts. siRNA knock down of E11 expression achieved >70% reduction of basal E11 mRNA expression (p < 0.05) and effectively abrogated FGF‐2‐related changes in E11 expression and dendrite formation. FGF‐2 strongly activated the ERK signaling pathway in osteoblast‐like cells but inhibition of this pathway did not block the ability of FGF‐2 to enhance E11 expression or to promote acquisition of the osteocyte phenotype. The results of this study highlight a novel mechanism by which FGF‐2 can regulate osteoblast differentiation and osteocyte formation. Specifically, the data suggests that FGF‐2 promotes osteocytogenesis through increased E11 expression and further studies will identify if this regulatory pathway is essential for bone development and maintenance in health and disease.
Collapse
Affiliation(s)
- Ekele Ikpegbu
- Roslin Institute and R(D)SVS, The University of Edinburgh, Edinburgh, UK.,Michael Okpara University of Agriculture, Abia, Nigeria
| | - Lena Basta
- Roslin Institute and R(D)SVS, The University of Edinburgh, Edinburgh, UK
| | - Dylan N Clements
- Roslin Institute and R(D)SVS, The University of Edinburgh, Edinburgh, UK
| | - Robert Fleming
- Roslin Institute and R(D)SVS, The University of Edinburgh, Edinburgh, UK
| | - Tonia L Vincent
- Arthritis Research UK Centre for Osteoarthritis Pathogenesis, Kennedy Institute of Rheumatology, University of Oxford, Oxford, UK
| | - David J Buttle
- Department of Infection, Immunity & Cardiovascular Disease, The University of Sheffield Medical School, Sheffield, UK
| | | | | | - Colin Farquharson
- Roslin Institute and R(D)SVS, The University of Edinburgh, Edinburgh, UK
| |
Collapse
|
14
|
Golan-Lavi R, Giacomelli C, Fuks G, Zeisel A, Sonntag J, Sinha S, Köstler W, Wiemann S, Korf U, Yarden Y, Domany E. Coordinated Pulses of mRNA and of Protein Translation or Degradation Produce EGF-Induced Protein Bursts. Cell Rep 2017; 18:3129-3142. [PMID: 28355565 DOI: 10.1016/j.celrep.2017.03.014] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2014] [Revised: 01/16/2017] [Accepted: 03/01/2017] [Indexed: 11/25/2022] Open
Abstract
Protein responses to extracellular cues are governed by gene transcription, mRNA degradation and translation, and protein degradation. In order to understand how these time-dependent processes cooperate to generate dynamic responses, we analyzed the response of human mammary cells to the epidermal growth factor (EGF). Integrating time-dependent transcript and protein data into a mathematical model, we inferred for several proteins their pre-and post-stimulus translation and degradation coefficients and found that they exhibit complex, time-dependent variation. Specifically, we identified strategies of protein production and degradation acting in concert to generate rapid, transient protein bursts in response to EGF. Remarkably, for some proteins, for which the response necessitates rapidly decreased abundance, cells exhibit a transient increase in the corresponding degradation coefficient. Our model and analysis allow inference of the kinetics of mRNA translation and protein degradation, without perturbing cells, and open a way to understanding the fundamental processes governing time-dependent protein abundance profiles.
Collapse
Affiliation(s)
- Roni Golan-Lavi
- Department of Physics of Complex Systems, Weizmann Institute of Science, Rehovot 7610001, Israel; Department of Biological Regulation, Weizmann Institute of Science, Rehovot 7610001, Israel
| | - Chiara Giacomelli
- Division of Molecular Genome Analysis, German Cancer Research Center (DKFZ), Heidelberg 69120, Germany
| | - Garold Fuks
- Department of Physics of Complex Systems, Weizmann Institute of Science, Rehovot 7610001, Israel
| | - Amit Zeisel
- Department of Physics of Complex Systems, Weizmann Institute of Science, Rehovot 7610001, Israel
| | - Johanna Sonntag
- Division of Molecular Genome Analysis, German Cancer Research Center (DKFZ), Heidelberg 69120, Germany
| | - Sanchari Sinha
- Department of Biological Regulation, Weizmann Institute of Science, Rehovot 7610001, Israel
| | - Wolfgang Köstler
- Department of Biological Regulation, Weizmann Institute of Science, Rehovot 7610001, Israel
| | - Stefan Wiemann
- Division of Molecular Genome Analysis, German Cancer Research Center (DKFZ), Heidelberg 69120, Germany
| | - Ulrike Korf
- Division of Molecular Genome Analysis, German Cancer Research Center (DKFZ), Heidelberg 69120, Germany
| | - Yosef Yarden
- Department of Biological Regulation, Weizmann Institute of Science, Rehovot 7610001, Israel.
| | - Eytan Domany
- Department of Physics of Complex Systems, Weizmann Institute of Science, Rehovot 7610001, Israel.
| |
Collapse
|
15
|
Ghouili F, Martin LJ. Cooperative regulation of Gja1 expression by members of the AP-1 family cJun and cFos in TM3 Leydig and TM4 Sertoli cells. Gene 2017; 635:24-32. [DOI: 10.1016/j.gene.2017.09.017] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2017] [Revised: 08/28/2017] [Accepted: 09/08/2017] [Indexed: 12/26/2022]
|
16
|
Li Y, Dillon TJ, Takahashi M, Earley KT, Stork PJS. Protein Kinase A-independent Ras Protein Activation Cooperates with Rap1 Protein to Mediate Activation of the Extracellular Signal-regulated Kinases (ERK) by cAMP. J Biol Chem 2016; 291:21584-21595. [PMID: 27531745 DOI: 10.1074/jbc.m116.730978] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2016] [Revised: 08/03/2016] [Indexed: 11/06/2022] Open
Abstract
Cyclic adenosine monophosphate (cAMP) is an important mediator of hormonal stimulation of cell growth and differentiation through its activation of the extracellular signal-regulated kinase (ERK) cascade. Two small G proteins, Ras and Rap1, have been proposed to mediate this activation, with either Ras or Rap1 acting in distinct cell types. Using Hek293 cells, we show that both Ras and Rap1 are required for cAMP signaling to ERKs. The roles of Ras and Rap1 were distinguished by their mechanism of activation, dependence on the cAMP-dependent protein kinase (PKA), and the magnitude and kinetics of their effects on ERKs. Ras was required for the early portion of ERK activation by cAMP and was activated independently of PKA. Ras activation required the Ras/Rap guanine nucleotide exchange factor (GEF) PDZ-GEF1. Importantly, this action of PDZ-GEF1 was disrupted by mutation within its putative cyclic nucleotide-binding domain within PDZ-GEF1. Compared with Ras, Rap1 activation of ERKs was of longer duration. Rap1 activation was dependent on PKA and required Src family kinases and the Rap1 exchanger C3G. This is the first report of a mechanism for the cooperative actions of Ras and Rap1 in cAMP activation of ERKs. One physiological role for the sustained activation of ERKs is the transcription and stabilization of a range of transcription factors, including c-FOS. We show that the induction of c-FOS by cAMP required both the early and sustained phases of ERK activation, requiring Ras and Rap1, as well as for each of the Raf isoforms, B-Raf and C-Raf.
Collapse
Affiliation(s)
- Yanping Li
- From the Vollum Institute, Oregon Health and Science University, Portland, Oregon 97239-3098
| | - Tara J Dillon
- From the Vollum Institute, Oregon Health and Science University, Portland, Oregon 97239-3098
| | - Maho Takahashi
- From the Vollum Institute, Oregon Health and Science University, Portland, Oregon 97239-3098
| | - Keith T Earley
- From the Vollum Institute, Oregon Health and Science University, Portland, Oregon 97239-3098
| | - Philip J S Stork
- From the Vollum Institute, Oregon Health and Science University, Portland, Oregon 97239-3098
| |
Collapse
|
17
|
Activation of MEK/ERK Signaling by PACAP in Guinea Pig Cardiac Neurons. J Mol Neurosci 2016; 59:309-16. [PMID: 27194157 DOI: 10.1007/s12031-016-0766-z] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2016] [Accepted: 04/29/2016] [Indexed: 10/21/2022]
Abstract
Pituitary adenylate cyclase-activating polypeptide (PACAP) signaling can increase guinea pig cardiac neuron excitability in part through extracellular signal-regulated kinase (ERK) activation. The present study examined the PACAP receptors and signaling cascades that stimulate guinea pig cardiac neuron ERK signaling using confocal microscopy to quantify PACAP-induced neuronal phosphorylated ERK (pERK) immunoreactivity. PACAP and maxadilan, but not vasoactive intestinal polypeptide (VIP), increased cardiac neuron pERK, implicating primary roles for PACAP-selective PAC1 receptor (Adcyap1r1) signaling rather than VPAC receptors (Vipr1 and Vipr2) in the generation of cardiac neuron pERK. The adenylyl cyclase (AC) activator forskolin, but not the protein kinase C (PKC) activator phorbol myristate acetate (PMA), increased pERK. Also, Bim1 did not blunt PACAP activation of pERK. Together, the results suggest PAC1 receptor signal transduction via Gs/adenylyl cyclase (AC)/cAMP rather than Gq/phospholipase C (PLC) generated neuronal pERK. Activator and inhibitor studies suggested that the PACAP-mediated pERK activation was PKA-dependent rather than an exchange protein directly activated by a cAMP (EPAC), PKA-independent mechanism. The PACAP-induced pERK was inhibited by the clathrin inhibitor Pitstop2 to block receptor internalization and endosomal signaling. We propose that the PACAP-mediated MEK/ERK activation in cardiac neurons involves both AC/cAMP/PKA signaling and PAC1 receptor internalization/activation of signaling endosomes.
Collapse
|
18
|
Cerny O, Kamanova J, Masin J, Bibova I, Skopova K, Sebo P. Bordetella pertussis Adenylate Cyclase Toxin Blocks Induction of Bactericidal Nitric Oxide in Macrophages through cAMP-Dependent Activation of the SHP-1 Phosphatase. THE JOURNAL OF IMMUNOLOGY 2015; 194:4901-13. [PMID: 25876760 DOI: 10.4049/jimmunol.1402941] [Citation(s) in RCA: 41] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Subscribe] [Scholar Register] [Received: 11/24/2014] [Accepted: 03/10/2015] [Indexed: 12/23/2022]
Abstract
The adenylate cyclase toxin-hemolysin (CyaA) plays a key role in the virulence of Bordetella pertussis. CyaA penetrates complement receptor 3-expressing phagocytes and catalyzes uncontrolled conversion of cytosolic ATP to the key second messenger molecule cAMP. This paralyzes the capacity of neutrophils and macrophages to kill bacteria by complement-dependent oxidative burst and opsonophagocytic mechanisms. We show that cAMP signaling through the protein kinase A (PKA) pathway activates Src homology domain 2 containing protein tyrosine phosphatase (SHP) 1 and suppresses production of bactericidal NO in macrophage cells. Selective activation of PKA by the cell-permeable analog N(6)-benzoyladenosine-3',5'-cyclic monophosphate interfered with LPS-induced inducible NO synthase (iNOS) expression in RAW264.7 macrophages, whereas inhibition of PKA by H-89 largely restored the production of iNOS in CyaA-treated murine macrophages. CyaA/cAMP signaling induced SHP phosphatase-dependent dephosphorylation of the c-Fos subunit of the transcription factor AP-1 and thereby inhibited TLR4-triggered induction of iNOS gene expression. Selective small interfering RNA knockdown of SHP-1, but not of the SHP-2 phosphatase, rescued production of TLR-inducible NO in toxin-treated cells. Finally, inhibition of SHP phosphatase activity by NSC87877 abrogated B. pertussis survival inside murine macrophages. These results reveal that an as yet unknown cAMP-activated signaling pathway controls SHP-1 phosphatase activity and may regulate numerous receptor signaling pathways in leukocytes. Hijacking of SHP-1 by CyaA action then enables B. pertussis to evade NO-mediated killing in sentinel cells of innate immunity.
Collapse
Affiliation(s)
- Ondrej Cerny
- Laboratory of Molecular Biology of Bacterial Pathogens, Institute of Microbiology of the ASCR, v.v.i., Czech Academy of Sciences, 142 20, Prague 4, Czech Republic
| | - Jana Kamanova
- Laboratory of Molecular Biology of Bacterial Pathogens, Institute of Microbiology of the ASCR, v.v.i., Czech Academy of Sciences, 142 20, Prague 4, Czech Republic
| | - Jiri Masin
- Laboratory of Molecular Biology of Bacterial Pathogens, Institute of Microbiology of the ASCR, v.v.i., Czech Academy of Sciences, 142 20, Prague 4, Czech Republic
| | - Ilona Bibova
- Laboratory of Molecular Biology of Bacterial Pathogens, Institute of Microbiology of the ASCR, v.v.i., Czech Academy of Sciences, 142 20, Prague 4, Czech Republic
| | - Karolina Skopova
- Laboratory of Molecular Biology of Bacterial Pathogens, Institute of Microbiology of the ASCR, v.v.i., Czech Academy of Sciences, 142 20, Prague 4, Czech Republic
| | - Peter Sebo
- Laboratory of Molecular Biology of Bacterial Pathogens, Institute of Microbiology of the ASCR, v.v.i., Czech Academy of Sciences, 142 20, Prague 4, Czech Republic
| |
Collapse
|
19
|
Zhao J, Cheng YY, Fan W, Yang CB, Ye SF, Cui W, Wei W, Lao LX, Cai J, Han YF, Rong JH. Botanical drug puerarin coordinates with nerve growth factor in the regulation of neuronal survival and neuritogenesis via activating ERK1/2 and PI3K/Akt signaling pathways in the neurite extension process. CNS Neurosci Ther 2014; 21:61-70. [PMID: 25310912 DOI: 10.1111/cns.12334] [Citation(s) in RCA: 51] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2014] [Revised: 09/12/2014] [Accepted: 09/13/2014] [Indexed: 01/11/2023] Open
Abstract
AIM Nerve growth factor (NGF) regulates neuronal survival and differentiation by activating extracellular signal-regulated-kinases (ERK) 1/2 and phosphoinositide-3-kinase (PI3K)/Akt pathways in two distinct processes: latency process and neurite extension process. This study was designed to investigate whether botanical drug C-glucosylated isoflavone puerarin coordinates with NGF to regulate neuritogenesis via activating ERK1/2 and PI3K/Akt in neurite extension process. METHODS We investigated the neuroprotective and neurotrophic activities of puerarin in MPTP-lesioned mice and dopaminergic PC12 cells. The effects of puerarin on ERK1/2, Akt, Nrf2, and HO-1 were assessed by Western blotting. The neurite outgrowth was assayed by neurite outgrowth staining kit. RESULTS Puerarin protected dopaminergic cells and ameliorated the behavioral impairments in MPTP-lesioned mice. Puerarin potentiated the effect of NGF on neuritogenesis in PC12 cells by >10-fold. Mechanistic studies revealed: (1) puerarin rapidly activated ERK1/2 and Akt, leading to the activation of Nrf2/heme oxygenase-1 (HO-1) pathways; (2) ERK1/2, PI3K/Akt, and HO-1 inhibitors attenuated the neuritogenic activity of puerarin. Notably, puerarin enhanced NGF-induced neuritogenesis in a timing-dependent manner. CONCLUSION Puerarin effectively coordinated with NGF to stimulate neuritogenesis via activating ERK1/2 and PI3K/Akt pathways in neurite extension process. These results demonstrated a general mechanism supporting the therapeutic application of puerarin-related compounds in neurodegenerative diseases.
Collapse
Affiliation(s)
- Jia Zhao
- School of Chinese Medicine, University of Hong Kong, Hong Kong, China
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
20
|
Georg B, Rask L, Hannibal J, Fahrenkrug J. The Light-InducedFOSResponse in Melanopsin Expressing HEK-293 Cells is Correlated with Melanopsin Quantity and Dependent on Light Duration and Irradiance. Photochem Photobiol 2014; 90:1069-76. [DOI: 10.1111/php.12298] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2014] [Accepted: 05/29/2014] [Indexed: 01/26/2023]
Affiliation(s)
- Birgitte Georg
- Department of Clinical Biochemistry; Faculty of Health Sciences; Bispebjerg Hospital; University of Copenhagen; Copenhagen NV Denmark
| | - Lene Rask
- Department of Clinical Biochemistry; Faculty of Health Sciences; Bispebjerg Hospital; University of Copenhagen; Copenhagen NV Denmark
| | - Jens Hannibal
- Department of Clinical Biochemistry; Faculty of Health Sciences; Bispebjerg Hospital; University of Copenhagen; Copenhagen NV Denmark
| | - Jan Fahrenkrug
- Department of Clinical Biochemistry; Faculty of Health Sciences; Bispebjerg Hospital; University of Copenhagen; Copenhagen NV Denmark
| |
Collapse
|
21
|
Qin Q, Feng J, Hu C, Chen X, Qin L, Li Y. Low-Intensity Aerobic Exercise Mitigates Exercise-Induced Bronchoconstriction by Improving the Function of Adrenal Medullary Chromaffin Cells in Asthmatic Rats. TOHOKU J EXP MED 2014; 234:99-110. [DOI: 10.1620/tjem.234.99] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022]
Affiliation(s)
- Qingwu Qin
- Department of Respiratory Medicine, Xiangya Hospital, Central South University
| | - Juntao Feng
- Department of Respiratory Medicine, Xiangya Hospital, Central South University
| | - Chengping Hu
- Department of Respiratory Medicine, Xiangya Hospital, Central South University
- Bronchial Asthma Research Center of Hunan Province
| | - Xi Chen
- Department of Respiratory Medicine, Xiangya Hospital, Central South University
| | - Ling Qin
- Department of Respiratory Medicine, Xiangya Hospital, Central South University
| | - Yuanyuan Li
- Department of Respiratory Medicine, Xiangya Hospital, Central South University
| |
Collapse
|
22
|
Heberden C, Meffray E, Goustard-Langelier B, Maximin E, Lavialle M. Dexamethasone inhibits the maturation of newly formed neurons and glia supplemented with polyunsaturated fatty acids. J Steroid Biochem Mol Biol 2013; 138:395-402. [PMID: 23907015 DOI: 10.1016/j.jsbmb.2013.07.010] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/26/2013] [Revised: 06/20/2013] [Accepted: 07/22/2013] [Indexed: 01/04/2023]
Abstract
Stress bears a negative impact on adult neurogenesis. High levels of corticoids have been shown to inhibit neural stem cell proliferation, and are considered responsible for the loss of neural precursors. Their effects on the differentiation of the glial and neuronal lineages have been less studied. We examined the effect of dexamethasone (Dex), a synthetic glucocorticoid, on the differentiation of rat neural stem cells in vitro. Dex had no effect on the differentiation of cells cultured under standard conditions. Since we previously determined that NSC, when cultured under classical conditions, were deprived of polyunsaturated fatty acids (PUFA), and displayed phospholipid compositions very different from the in vivo figures [1], we examined the effect of Dex under PUFA supplementation. Dex impaired neuron and oligodendrocyte maturation in PUFA-supplemented cells, demonstrated by the reduction of neurite lengths and oligodendrocyte sizes. This effect was mediated by the glucocorticoid receptor (GR), since it was eliminated by mifepristone, a GR antagonist, and could be relayed by a reduction of ERK phosphorylation. We determined that GR was associated with PPAR β and α under basal conditions, and that this association was disrupted when PUFA were added in combination with Dex. We assumed that this effect on the receptor status enabled the effect of Dex on PUFA supplemented cells, since we determined that the binding to the glucocorticoid response element was higher in cells incubated with PUFA and Dex. In conclusion, corticoids can impair NSC differentiation, and consequently impact the entire process of neurogenesis.
Collapse
Affiliation(s)
- Christine Heberden
- INRA UR0909 Laboratoire de Nutrition et Régulations Lipidiques des Fonctions Cérébrales, CRJ 78352 Jouy-en-Josas, France.
| | | | | | | | | |
Collapse
|
23
|
ERK and RSK regulate distinct steps of a cellular program that induces transition from multicellular epithelium to single cell phenotype. Cell Signal 2013; 25:2743-51. [PMID: 24012955 DOI: 10.1016/j.cellsig.2013.08.024] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2013] [Accepted: 08/23/2013] [Indexed: 01/13/2023]
Abstract
The ERK (extracellular signal-regulated kinases) cascade has an evolutionarily conserved three tier architecture consisting of protein kinases Raf, MEK (MAPK/ERK kinase) and ERK. Following activation, ERK phosphorylates various cellular elements leading to diverse cellular responses. Downstream of ERK the family of p90 ribosomal S6 kinases (RSKs) has been proven to be an important conveyor of ERK signaling, however, little is known if ERK and RSK coordinate their functions to generate a specific biological response. Here we show that in epithelial cells conditional activation of the ERK pathway causes phenotypic conversion of epithelial cells to autonomously migrating cells. This process involves two sequential steps characterized by loss of apical-basal polarity followed by cell scattering. The activation of ERK, but not RSK, is sufficient for the execution of the first step and it requires calpain mediated remodeling of actin cytoskeleton. Conversely, RSK regulates the successive stage characterized by cell-cell contact weakening and increased cellular migration. Thus, ERK and RSK regulate different cellular subprograms and coordinated execution of these subprograms in time generates a relevant biological response. Our data also suggest that the mechanism by which the ERK pathway controls a cellular response may be distributed between ERK and RSK, rather than being elicited by a single effector kinase.
Collapse
|
24
|
|
25
|
Rodrigues AR, Almeida H, Gouveia AM. Melanocortin 5 receptor signaling and internalization: role of MAPK/ERK pathway and β-arrestins 1/2. Mol Cell Endocrinol 2012; 361:69-79. [PMID: 22871966 DOI: 10.1016/j.mce.2012.03.011] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/17/2011] [Revised: 03/19/2012] [Accepted: 03/19/2012] [Indexed: 11/25/2022]
Abstract
The Melanocortin 5 receptor (MC5R) is a G-protein coupled receptor (GPCR) that exhibits high affinity for α-MSH. Here we present evidence for MC5R-GFP internalization and subsequent recycling to cell surface, in α-MSH-stimulated HeLa cells. This melanocortin induces a biphasic activation of ERK1/2 with an early peak at 15min, a G(i)-protein driven, β-arrestins 1/2 independent process, and a late sustained activation that is regulated by β-arrestins 1/2. ERK1/2 lead to downstream phosphorylation of 90-kDa ribosomal S6 kinases (p90RSK) and mitogen- and stress-activated protein kinase 1 (MSK1). Only a small fraction (10%) of phosphorylated p90RSK and ERK1/2 translocates to the nucleus inducing c-Fos expression. α-MSH also activates CREB through cAMP/PKA pathway. In 3T3-L1 adipocytes, where MC5R is endogenously expressed, α-MSH also induces phosphorylation and cytosolic retention of the same signaling molecules. These findings provide new evidence on the signaling mechanisms underlying MC5R biological response to α-MSH.
Collapse
Affiliation(s)
- Adriana R Rodrigues
- Department of Experimental Biology, Faculty of Medicine, Universidade do Porto, Porto, Portugal
| | | | | |
Collapse
|
26
|
Basson MA. Signaling in cell differentiation and morphogenesis. Cold Spring Harb Perspect Biol 2012; 4:cshperspect.a008151. [PMID: 22570373 DOI: 10.1101/cshperspect.a008151] [Citation(s) in RCA: 98] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
All the information to make a complete, fully functional living organism is encoded within the genome of the fertilized oocyte. How is this genetic code translated into the vast array of cellular behaviors that unfold during the course of embryonic development, as the zygote slowly morphs into a new organism? Studies over the last 30 years or so have shown that many of these cellular processes are driven by secreted or membrane-bound signaling molecules. Elucidating how the genetic code is translated into instructions or signals during embryogenesis, how signals are generated at the correct time and place and at the appropriate level, and finally, how these instructions are interpreted and put into action, are some of the central questions of developmental biology. Our understanding of the causes of congenital malformations and disease has improved substantially with the rapid advances in our knowledge of signaling pathways and their regulation during development. In this article, I review some of the signaling pathways that play essential roles during embryonic development. These examples show some of the mechanisms used by cells to receive and interpret developmental signals. I also discuss how signaling pathways downstream from these signals are regulated and how they induce specific cellular responses that ultimately affect cell fate and morphogenesis.
Collapse
Affiliation(s)
- M Albert Basson
- Department of Craniofacial Development, King's College London, United Kingdom.
| |
Collapse
|
27
|
Zhang QL, Qiao LY. Regulation of IGF-1 but not TGF-β1 by NGF in the smooth muscle of the inflamed urinary bladder. ACTA ACUST UNITED AC 2012; 177:73-8. [PMID: 22579999 DOI: 10.1016/j.regpep.2012.05.088] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2011] [Revised: 09/01/2011] [Accepted: 05/02/2012] [Indexed: 10/28/2022]
Abstract
Intraperitoneal injection of cyclophosphamide (CYP) causes hemorrhagic cystitis with excess growth of muscular layer leading to bladder hypertrophy; this could be attributable to changes in the expression profiles of growth factors in the inflamed urinary bladder. The growth factors characterized in the current study include nerve growth factor (NGF), insulin-like growth factor (IGF)-1, and transforming growth factor (TGF)-β1. We found that following CYP injection for 8 h and 48 h, the mRNA levels of all three factors were increased in the inflamed bladder when compared to control. The level of NGF mRNA was mainly increased in the urothelium layer while the levels of IGF-1 mRNA and TGF-β1 mRNA were increased in the smooth muscle layer. The level of NGF high affinity receptor TrkA mRNA was also increased in both the urothelium and the smooth muscle layers during bladder inflammation. When we blocked NGF action with NGF neutralizing antibody in vivo, we found that the up-regulation of IGF-1 in the inflamed bladder was reversed while the up-regulation of TGF-β1 was not affected by NGF neutralization. The effect of NGF on regulating IGF-1 expression was further confirmed in bladder smooth muscle culture showing that exogenous NGF increased the mRNA level of IGF-1 after 30 min to 1 h stimulation. These results suggested that bladder inflammation induced region-specific changes in the expression profiles of NGF, IGF-1 and TGF-β1. The up-regulation of NGF in the urothelium may have a role in affecting bladder smooth muscle cell physiology by regulating IGF-1 expression.
Collapse
Affiliation(s)
- Qing L Zhang
- Department of Physiology and Biophysics, School of Medicine, Virginia Commonwealth University, Richmond, VA, United States
| | | |
Collapse
|
28
|
Yoo YD, Huang CT, Zhang X, Lavaute TM, Zhang SC. Fibroblast growth factor regulates human neuroectoderm specification through ERK1/2-PARP-1 pathway. Stem Cells 2012; 29:1975-82. [PMID: 21997878 DOI: 10.1002/stem.758] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
Fibroblast growth factor (FGF) signaling and PAX6 transcription are required for neuroectoderm specification of human embryonic stem cells (hESCs). In this study, we asked how FGF signaling leads to PAX6 transcription and neuroectoderm specification from hESCs. Under a chemically defined medium, FGF inhibition blocked phosphorylation of extracellular signal-regulated kinase 1/2 (ERK 1/2) with a significant reduction of PAX6-expressing neuroepithelia, indicating that FGF regulates neural induction through ERK1/2 activation. Activation of FGF-ERK1/2 pathway was necessary for the activity of poly(ADP-ribose) polymerase-1 (PARP-1), a conserved nuclear protein catalyzing polymerization of ADP-ribose units. Pharmacological inhibition and genetic ablation of PARP-1 inhibited neural induction from hESCs, suggesting that FGF-ERK1/2 signal pathway regulates neuroectoderm specification through regulating PARP-1 activity. Furthermore, FGF-ERK1/2-PARP-1 cascade regulated the expression of PAX6, a transcription determinant of human neuroectoderm. Together, we propose that FGF regulates hESC neural specification through the ERK1/2-PARP-1 signaling pathway.
Collapse
Affiliation(s)
- Young Dong Yoo
- Waisman Center, University of Wisconsin-Madison, Madison, Wisconsin 53705, USA
| | | | | | | | | |
Collapse
|
29
|
Regulation of primary response genes. Mol Cell 2011; 44:348-60. [PMID: 22055182 DOI: 10.1016/j.molcel.2011.09.014] [Citation(s) in RCA: 171] [Impact Index Per Article: 13.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2011] [Revised: 08/29/2011] [Accepted: 09/22/2011] [Indexed: 12/24/2022]
Abstract
Primary response genes (PRGs) are a set of genes that are induced in response to both cell-extrinsic and cell-intrinsic signals and do not require de novo protein synthesis for their expression. These "first responders" in the waves of transcription of signal-responsive genes play pivotal roles in a wide range of biological responses, including neuronal survival and plasticity, cardiac stress response, innate and adaptive immune responses, glucose metabolism, and oncogeneic transformation. Here we bring together recent advances and our current understanding of the signal-induced transcriptional and epigenetic regulation of PRGs.
Collapse
|
30
|
Dong W, Li Y, Gao M, Hu M, Li X, Mai S, Guo N, Yuan S, Song L. IKKα contributes to UVB-induced VEGF expression by regulating AP-1 transactivation. Nucleic Acids Res 2011; 40:2940-55. [PMID: 22169952 PMCID: PMC3326327 DOI: 10.1093/nar/gkr1216] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
Exposure to ultraviolet B (UVB) irradiation from sunlight induces the upregulation of VEGF, a potent angiogenic factor that is critical for mediating angiogenesis-associated photodamage. However, the molecular mechanisms related to UVB-induced VEGF expression have not been fully defined. Here, we demonstrate that one of the catalytic subunits of the IκB kinase complex (IKK), IKKα, plays a critical role in mediating UVB-induced VEGF expression in mouse embryonic fibroblasts (MEFs), which requires IKKα kinase activity but is independent of IKKβ, IKKγ and the transactivation of NF-κB. We further show that the transcriptional factor AP-1 functions as the downstream target of IKKα that is responsible for VEGF induction under UVB exposure. Both the accumulation of AP-1 component, c-Fos and the transactivation of AP-1 by UVB require the activated IKKα located within the nucleus. Moreover, nuclear IKKα can associate with c-Fos and recruit to the vegf promoter regions containing AP-1-responsive element and then trigger phosphorylation of the promoter-bound histone H3. Thus, our results have revealed a novel independent role for IKKα in controlling VEGF expression during the cellular UVB response by regulating the induction of the AP-1 component and phosphorylating histone H3 to facilitate AP-1 transactivation. Targeting IKKα shows promise for the prevention of UVB-induced angiogenesis and the associated photodamage.
Collapse
Affiliation(s)
- Wen Dong
- Department of Pathophysiology, Beijing Institute of Basic Medical Sciences, Beijing 100850, P R China
| | | | | | | | | | | | | | | | | |
Collapse
|
31
|
Mullenbrock S, Shah J, Cooper GM. Global expression analysis identified a preferentially nerve growth factor-induced transcriptional program regulated by sustained mitogen-activated protein kinase/extracellular signal-regulated kinase (ERK) and AP-1 protein activation during PC12 cell differentiation. J Biol Chem 2011; 286:45131-45. [PMID: 22065583 DOI: 10.1074/jbc.m111.274076] [Citation(s) in RCA: 48] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023] Open
Abstract
Neuronal differentiation of PC12 cells in response to NGF is a prototypical model in which signal duration determines a biological response. Sustained ERK activity induced by NGF, as compared with transient activity induced by EGF, is critical to the differentiation of these cells. To characterize the transcriptional program activated preferentially by NGF, we compared global gene expression profiles between cells treated with NGF and EGF for 2-4 h, when sustained ERK signaling in response to NGF is most distinct from the transient signal elicited by EGF. This analysis identified 69 genes that were preferentially up-regulated in response to NGF. As expected, up-regulation of these genes was mediated by sustained ERK signaling. In addition, they were up-regulated in response to other neuritogenic treatments (pituitary adenylate cyclase-activating polypeptide and 12-O-tetradecanoylphorbol-13-acetate plus dbcAMP) and were enriched for genes related to neuronal differentiation/function. Computational analysis and chromatin immunoprecipitation identified binding of CREB and AP-1 family members (Fos, FosB, Fra1, JunB, JunD) upstream of >30 and 50%, respectively, of the preferentially NGF-induced genes. Expression of several AP-1 family members was induced by both EGF and NGF, but their induction was more robust and sustained in response to NGF. The binding of Fos family members to their target genes was similarly sustained in response to NGF and was reduced upon MEK inhibition, suggesting that AP-1 contributes significantly to the NGF transcriptional program. Interestingly, Fra1 as well as two other NGF-induced AP-1 targets (HB-EGF and miR-21) function in positive feedback loops that may contribute to sustained AP-1 activity.
Collapse
Affiliation(s)
- Steven Mullenbrock
- Department of Biology, Boston University, Boston, Massachusetts 02215, USA
| | | | | |
Collapse
|
32
|
Vora P, Pillai PP, Zhu W, Mustapha J, Namaka MP, Frost EE. Differential effects of growth factors on oligodendrocyte progenitor migration. Eur J Cell Biol 2011; 90:649-56. [PMID: 21616555 DOI: 10.1016/j.ejcb.2011.03.006] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2010] [Revised: 03/11/2011] [Accepted: 03/22/2011] [Indexed: 10/18/2022] Open
Abstract
Oligodendrocytes are myelinating cells of the CNS that originate as progenitor cells (OP) in discrete areas of the developing brain. During brain development, OP migrate significant distances prior to proliferating and myelinating the axons of the putative white matter tracts. Growth factors play a major regulatory role in the behavior of OP. Specifically, platelet-derived growth factor A (PDGF-A) and fibroblast growth factor 2 (FGF2) are two of the most well characterized regulators of OP development. Both growth factors interact with tyrosine kinase receptors, activating various intracellular signaling pathways. The current study advances our earlier research by comparing the effects of both PDGF-A and FGF2 on OP migration. Our results show that activation of ERK is required for OP migration. These findings correlate well with our previous demonstration of the ERK pathway mediating PDGF-A induced OP migration. We also demonstrate the significance of threshold levels of growth factors and temporal regulation for OP migration. In addition, ERK activation alone is not sufficient to induce OP migration. The current research supports the involvement of the non-ERK mediated signaling pathway in OP migration.
Collapse
Affiliation(s)
- Parvez Vora
- Faculty of Pharmacy, University of Manitoba, Winnipeg, Manitoba, Canada
| | | | | | | | | | | |
Collapse
|
33
|
Iwatsuki M, Inageda K, Matsuoka M. Cadmium induces phosphorylation and stabilization of c-Fos in HK-2 renal proximal tubular cells. Toxicol Appl Pharmacol 2011; 251:209-16. [PMID: 21219922 DOI: 10.1016/j.taap.2010.12.015] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2010] [Accepted: 12/30/2010] [Indexed: 01/04/2023]
Abstract
We examined the effects of cadmium chloride (CdCl₂) exposure on the expression and phosphorylation status of members of the Fos family, components of the activator protein-1 transcription factor, in HK-2 human renal proximal tubular cells. Following the exposure to CdCl₂, the expression of c-fos, fosB, fra-1, and fra-2 increased markedly, with different magnitudes and time courses. The levels of Fos family proteins (c-Fos, FosB, Fra-1, and Fra-2) also increased in response to CdCl₂ exposure. Although the elevation of c-fos transcripts was transient, c-Fos protein levels increased progressively with lower electrophoretic mobility, suggesting stabilization of c-Fos through post-translational modifications. Consistently, we observed phosphorylation of c-Fos at Ser362 and Ser374 in HK-2 cells treated with CdCl₂. Phosphorylated forms of mitogen-activated protein kinases (MAPKs)-including extracellular signal-regulated protein kinase (ERK), c-Jun NH₂-terminal kinase, and p38-increased after CdCl₂ exposure, whereas treatment with the MAPK/ERK kinase inhibitor U0126 and the p38 inhibitor SB203580 suppressed the accumulation and phosphorylation of c-Fos. We mutated Ser362 to alanine (S362A), Ser374 to alanine (S374A), and both residues to alanines (S362A/S374A) to inhibit potential phosphorylation of c-Fos at these sites. S374A or double S362A/S374A mutations reduced c-Fos level markedly, but S362A mutation did not. On the other hand, S362A/S374A mutations induced a more pronounced reduction in c-Fos DNA-binding activity than S374A mutation. These results suggest that while Ser374 phosphorylation seems to play a role in c-Fos stabilization, phosphorylation at two C-terminal serine residues is required for the transcriptional activation of c-Fos in HK-2 cells treated with CdCl₂.
Collapse
Affiliation(s)
- Mamiko Iwatsuki
- Department of Hygiene and Public Health I, Tokyo Women's Medical University, 8-1 Kawada-cho, Shinjuku-ku, Tokyo 162-8666, Japan
| | | | | |
Collapse
|
34
|
Bakiri L, Reschke MO, Gefroh HA, Idarraga MH, Polzer K, Zenz R, Schett G, Wagner EF. Functions of Fos phosphorylation in bone homeostasis, cytokine response and tumourigenesis. Oncogene 2010; 30:1506-17. [PMID: 21119595 DOI: 10.1038/onc.2010.542] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
Mice lacking c-fos develop osteopetrosis due to a block in osteoclast differentiation. Carboxy-terminal phosphorylation of Fos on serine 374 by ERK1/2 and serine 362 by RSK1/2 regulates Fos stability and transactivation potential in vitro. To assess the physiological relevance of Fos phosphorylation in vivo, serine 362 and/or serine 374 was replaced by alanine (Fos362A, Fos374A and FosAA) or by phospho-mimetic aspartic acid (FosDD). Homozygous mutants were healthy and skeletogenesis was largely unaffected. Fos C-terminal phosphorylation, predominantly on serine 374, was found important for osteoclast differentiation in vitro and affected lipopolysaccharide (LPS)-induced cytokine response in vitro and in vivo. Importantly, skin papilloma development was delayed in FosAA, Fos362A and Rsk2-deficient mice, accelerated in FosDD mice and unaffected in Fos374A mutants. Furthermore, the related Fos protein and putative RSK2 target Fra1 failed to substitute for Fos in papilloma development. This indicates that phosphorylation of serines 362 and 374 exerts context-dependent roles in modulating Fos activity in vivo. Inhibition of Fos C-terminal phosphorylation on serine 362 by targeting RSK2 might be of therapeutic relevance for skin tumours.
Collapse
Affiliation(s)
- L Bakiri
- Genes, Development and Disease Group, F-BBVA Cancer Cell Biology programme, National Cancer Research Centre (CNIO), Madrid, Spain
| | | | | | | | | | | | | | | |
Collapse
|
35
|
Nakakuki T, Birtwistle MR, Saeki Y, Yumoto N, Ide K, Nagashima T, Brusch L, Ogunnaike BA, Okada-Hatakeyama M, Kholodenko BN. Ligand-specific c-Fos expression emerges from the spatiotemporal control of ErbB network dynamics. Cell 2010; 141:884-96. [PMID: 20493519 DOI: 10.1016/j.cell.2010.03.054] [Citation(s) in RCA: 171] [Impact Index Per Article: 12.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2008] [Revised: 11/12/2009] [Accepted: 03/11/2010] [Indexed: 10/19/2022]
Abstract
Activation of ErbB receptors by epidermal growth factor (EGF) or heregulin (HRG) determines distinct cell-fate decisions, although signals propagate through shared pathways. Using mathematical modeling and experimental approaches, we unravel how HRG and EGF generate distinct, all-or-none responses of the phosphorylated transcription factor c-Fos. In the cytosol, EGF induces transient and HRG induces sustained ERK activation. In the nucleus, however, ERK activity and c-fos mRNA expression are transient for both ligands. Knockdown of dual-specificity phosphatases extends HRG-stimulated nuclear ERK activation, but not c-fos mRNA expression, implying the existence of a HRG-induced repressor of c-fos transcription. Further experiments confirmed that this repressor is mainly induced by HRG, but not EGF, and requires new protein synthesis. We show how a spatially distributed, signaling-transcription cascade robustly discriminates between transient and sustained ERK activities at the c-Fos system level. The proposed control mechanisms are general and operate in different cell types, stimulated by various ligands.
Collapse
Affiliation(s)
- Takashi Nakakuki
- Computational Systems Biology Research Group, Advanced Computational Sciences Department, RIKEN Advanced Science Institute, 1-7-22 Tsurumi-ku, Yokohama, Kanagawa 230-0045, Japan
| | | | | | | | | | | | | | | | | | | |
Collapse
|
36
|
Leugers CJ, Lee G. Tau potentiates nerve growth factor-induced mitogen-activated protein kinase signaling and neurite initiation without a requirement for microtubule binding. J Biol Chem 2010; 285:19125-34. [PMID: 20375017 DOI: 10.1074/jbc.m110.105387] [Citation(s) in RCA: 43] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/16/2023] Open
Abstract
Microtubule-associated protein Tau is known to bind to and stabilize microtubules, thereby regulating microtubule dynamics. However, recent evidence has indicated that Tau can also interact with various components of intracellular signaling pathways, leading to the possibility that Tau might have a role in signal transduction. Here we provide evidence that during growth factor stimulation of neuronal cells, Tau has functions in advance of the neurite elongation stage. Using Tau-depleted neuronal cell lines, we demonstrate that Tau is required for neurite initiation in a manner that does not involve its microtubule binding function. In addition, we demonstrate that Tau potentiates AP-1 transcription factor activation in response to nerve growth factor (NGF). The effect of Tau on AP-1 activation is mediated through its ability to potentiate the activation of mitogen-activated protein kinase (MAPK), which occurs in response to both NGF and epidermal growth factor. Phosphorylation of Tau at Thr-231 also occurs in response to NGF and is required for Tau to impact on MAPK signaling, whereas the ability of Tau to bind to microtubules is not required. Together, these findings indicate a new functional role for Tau in early neuronal development independent of its established role in microtubule stabilization.
Collapse
Affiliation(s)
- Chad J Leugers
- Roy J and Lucille A Carver College of Medicine, University of Iowa, Iowa City, Iowa 52242, USA
| | | |
Collapse
|
37
|
Daniele S, Lecca D, Trincavelli ML, Ciampi O, Abbracchio MP, Martini C. Regulation of PC12 cell survival and differentiation by the new P2Y-like receptor GPR17. Cell Signal 2010; 22:697-706. [PMID: 20056144 DOI: 10.1016/j.cellsig.2009.12.006] [Citation(s) in RCA: 39] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2009] [Revised: 12/04/2009] [Accepted: 12/18/2009] [Indexed: 11/18/2022]
Abstract
The P2Y-like receptor GPR17 has been reported to respond to both uracil nucleotides and cysteinyl-leukotrienes (cysLTs), such as UDP-glucose and LTD(4). Our previous data suggest a potential role for GPR17 in regulation of both cell viability and differentiation state of central nervous system cells. On this basis, in the present paper we investigated the effect of GPR17 receptor ligands on PC12 cell viability, following induction of morphological differentiation by nerve growth factor (NGF). In addition, the role of GPR17 ligands, either alone or in combination with growth factors, on the degree of PC12 cell differentiation was investigated. GPR17, which was not basally expressed in undifferentiated PC12 cells, was specifically induced by a 10day-treatment with NGF, suggesting a role in the control of neuronal specification. Both UDP-glucose and LTD(4), agonists at the nucleotide and cysLT GPR17 binding sites, respectively, induced a significant pro-survival effect on PC12 cells after priming with NGF. By in vitro silencing experiments with specific small interfering RNAs and by using receptor antagonists, we confirmed that the agonist effects are indeed mediated by the selective activation of GPR17. We also demonstrated that GPR17 agonists act, both alone and synergistically with NGF, to promote neurite outgrowth in PC12 cells. In addition, GPR17 ligands were able to confer an NGF-like activity to the epidermal growth factor (EGF), that, under these experimental conditions, also promoted cell differentiation and neurite elongation. Finally, we show that GPR17 ligands activate the intracellular phosphorylation of both ERK 1/2 and p38 kinases, that have been identified as important signalling pathways for neurotrophins in PC12 cells. Our results establish GPR17 as a neurotrophic regulator for neuronal-like cells and suggest a possible interplay between endogenous uracil derivatives, cysLTs and NGF in the signalling pathways involved in neuronal survival and differentiation. They also represent the first direct demonstration, in a native system, that GPR17 can indeed be activated by uracil nucleotides and cysLTs, in line with what previously demonstrated in recombinant expression systems.
Collapse
Affiliation(s)
- Simona Daniele
- Department of Psychiatry, Neurobiology, Pharmacology and Biotechnology, University of Pisa, Via Bonanno 6, 56126 Pisa, Italy
| | | | | | | | | | | |
Collapse
|
38
|
Abstract
Cooperation among transcription factors is central for their ability to execute specific transcriptional programmes. The AP1 complex exemplifies a network of transcription factors that function in unison under normal circumstances and during the course of tumour development and progression. This Perspective summarizes our current understanding of the changes in members of the AP1 complex and the role of ATF2 as part of this complex in tumorigenesis.
Collapse
Affiliation(s)
- Pablo Lopez-Bergami
- Instituto de Biologia y Medicina Experimental, Vuelta de Obligado 2490, Buenos Aires1428, Argentina,
| | - Eric Lau
- Signal Transduction Program, Burnham Institute for Medical Research, La Jolla, CA 92037, USA,
| | - Ze'ev Ronai
- Signal Transduction Program, Burnham Institute for Medical Research, La Jolla, CA 92037, USA
| |
Collapse
|
39
|
Chung CW, Zhang QL, Qiao LY. Endogenous nerve growth factor regulates collagen expression and bladder hypertrophy through Akt and MAPK pathways during cystitis. J Biol Chem 2009; 285:4206-4212. [PMID: 19996110 DOI: 10.1074/jbc.m109.040444] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Type I collagen forms the main constituent of the extracellular matrix in visceral organs. We reported here that cyclophosphamide (CYP)-induced cystitis significantly increased the production of type I collagen in the inflamed bladder leading to increases in the bladder weight and the thickness of the bladder wall. The endogenous nerve growth factor (NGF) in the urinary bladder regulated type I collagen expression because the neutralizing NGF antibody attenuated cystitis-induced type I collagen up-regulation in the inflamed bladder. Neutralizing NGF antibody also subsequently reversed cystitis-induced increases in bladder weight. Further studies on the intermediate signaling pathways mediating NGF-induced type I collagen expression in the inflamed bladder during cystitis revealed that Akt, JNK, and ERK1/2 activities were increased in the inflamed bladder, whereas p38 MAPK remained unchanged. Suppression of endogenous NGF level with neutralizing NGF antibody significantly blocked the increased activity of Akt, JNK, and ERK1/2 in the inflamed bladder during cystitis. These results indicate that endogenous NGF plays an important role in the activation of Akt and MAPK in the urinary bladder and in bladder hypertrophy during cystitis.
Collapse
Affiliation(s)
- Chul-Won Chung
- From the Department of Physiology and Biophysics, Virginia Commonwealth University School of Medicine, Richmond, Virginia 23298-0551
| | - Qing L Zhang
- From the Department of Physiology and Biophysics, Virginia Commonwealth University School of Medicine, Richmond, Virginia 23298-0551
| | - Li-Ya Qiao
- From the Department of Physiology and Biophysics, Virginia Commonwealth University School of Medicine, Richmond, Virginia 23298-0551.
| |
Collapse
|
40
|
Park II, Zhang Q, Liu V, Kozlowski JM, Zhang J, Lee C. 17Beta-estradiol at low concentrations acts through distinct pathways in normal versus benign prostatic hyperplasia-derived prostate stromal cells. Endocrinology 2009; 150:4594-605. [PMID: 19608654 DOI: 10.1210/en.2008-1591] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
The aim of this study was to identify differential responses to low concentrations of 17beta-estradiol (E2) in primary stromal cell cultures derived from either normal organ donors or benign prostatic hyperplasia or hypertrophy (BPH) specimens. Furthermore, we sought to identify the potential mechanism of E2 action in these cell types, through either a genomic or nongenomic mechanism. We initially treated stromal cells derived from five normal prostates or five BPH specimens with low concentrations of E2 (0.001-1.0 nM) and analyzed their growth response. To determine whether genomic or nongenomic pathways were involved, we performed studies using specific estrogen receptor antagonists to confirm transcriptional activity or MAPK inhibitors to confirm the involvement of rapid signaling. Results of these studies revealed a fundamental difference in the mechanism of the response to E2. In normal cells, we found that a nongenomic, rapid E2 signaling pathway is predominantly involved, mediated by G protein-coupled receptor-30 and the subsequent activation of ERK1/2. In BPH-derived prostate stromal cells, a genomic pathway is predominantly involved because the addition of ICI 182780 was sufficient to abrogate any estrogenic effects. In conclusion, prostate stromal cells respond to far lower concentrations of E2 than previously recognized or examined, and this response is mediated through two distinct mechanisms, depending on its origin. This may provide the basis for new insights into the causes of, and possible treatments for, BPH.
Collapse
Affiliation(s)
- Irwin I Park
- Department of Urology, Northwestern University Feinberg School of Medicine, Chicago, Illinois 60611, USA
| | | | | | | | | | | |
Collapse
|
41
|
Falktoft B, Georg B, Fahrenkrug J. Signaling pathways in PACAP regulation of VIP gene expression in human neuroblastoma cells. Neuropeptides 2009; 43:387-96. [PMID: 19712974 DOI: 10.1016/j.npep.2009.08.002] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/13/2009] [Revised: 07/16/2009] [Accepted: 08/04/2009] [Indexed: 11/30/2022]
Abstract
Ganglia expressing the neuropeptide pituitary adenylate cyclase-activating polypeptide (PACAP) innervate vasoactive intestinal peptide (VIP) containing neurons suggesting a role of PACAP in regulating VIP expression. Human NB-1 neuroblastoma cells were applied to study PACAP regulated VIP gene expression aiming to identify the receptor and the signaling proteins involved. The PACAP receptor subtype PAC1 induced VIP gene expression as (i) PACAP and the PAC1 receptor agonist maxadilan were equally efficient and approximately 200-fold more potent than VIP, and (ii) PACAP6-38 and PG99-465, antagonists of PAC1 and VPAC2 receptors, respectively, abolished and did not affect the PACAP-induced VIP mRNA expression, respectively. A pivotal role of PKA was implicated in addition to partial involvement of PKC and ERK1/2 in PACAP-induced VIP gene expression as H-89, Bisindolylmaleimide I (BIS), Gö6976 and U0126 attenuated the VIP mRNA expression by 93%, 58%, 58% and 40%, respectively. PACAP modulated the phosphorylation of ERK1/2 (pERK1/2) and CREB/ATF-1 (pCREB/ATF-1) concomitant with a translocation of PKA to the nucleus. Inhibition of conventional PKC isoforms and MEK1/2 completely abolished pERK1/2 without affecting PACAP induced pCREB/ATF-1. In contrast, inhibiting PKA attenuated PACAP induced pCREB/ATF-1. PACAP also enhanced the FOS gene expression and individual presence of H-89, BIS, Gö6976 and U0126 partially attenuated the PACAP induced FOS mRNA expression. Combining the kinase inhibitors completely suppressed the PACAP induced FOS mRNA expression. Immunoblotting confirmed expression of FOS protein upon addition of PACAP, which was diminished by impairment of PKC, ERK1/2 and PKA activities. The resemblance of the signaling pathways involving concomitant activities of PKC, ERK1/2 and PKA in PACAP regulation of the FOS and VIP gene expressions suggest for the first time a role of FOS in PACAP-induced VIP gene expression in human NB-1 neuroblastoma cells.
Collapse
Affiliation(s)
- Birgitte Falktoft
- Department of Clinical Biochemistry, Bispebjerg Hospital, Bispebjerg Bakke 23, DK-2400 Copenhagen, Denmark.
| | | | | |
Collapse
|
42
|
ERK activation and cell growth require CaM kinases in MCF-7 breast cancer cells. Mol Cell Biochem 2009; 335:155-71. [DOI: 10.1007/s11010-009-0252-9] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2009] [Accepted: 09/02/2009] [Indexed: 10/20/2022]
|
43
|
Kanda N, Shibata S, Tada Y, Nashiro K, Tamaki K, Watanabe S. Prolactin enhances basal and IL-17-induced CCL20 production by human keratinocytes. Eur J Immunol 2009; 39:996-1006. [PMID: 19350575 DOI: 10.1002/eji.200838852] [Citation(s) in RCA: 37] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/26/2023]
Abstract
Psoriasis vulgaris is an autoimmune dermatosis with Th17 infiltration. Prolactin (PRL) may participate in the pathogenesis of psoriasis. The chemokine CCL20 recruits Th17 cells, and CCL20 production by epidermal keratinocytes is enhanced in psoriatic lesions. We examined the in vitro effects of PRL on CCL20 production in human keratinocytes. PRL increased basal and IL-17-induced CCL20 secretion, and mRNA expression in keratinocytes. CCL20 production by PRL was suppressed by antisense oligonucleotides against the AP-1 components c-Fos and c-Jun, whereas that by IL-17 was suppressed by antisense NF-kappaB p50 and p65. CCL20 production induced by PRL plus IL-17 was suppressed by antisense c-Fos, c-Jun, p50, and p65. PRL alone increased the transcriptional activity of AP-1, and c-Fos and c-Jun expression; moderately enhanced NF-kappaB activity and IkappaBalpha phosphorylation; and potently increased IL-17-induced NF-kappaB activity. MEK and JNK inhibitors suppressed PRL- or PRL-plus-IL-17-induced CCL20 production and AP-1 activities. MEK inhibitor suppressed PRL-induced c-Fos expression, whereas JNK inhibitor suppressed c-Jun expression. PRL induced ERK and JNK phosphorylation. These results suggest that PRL may enhance basal and IL-17-induced CCL20 production in keratinocytes by AP-1 and NF-kappaB activation, which is partially mediated via MEK/ERK and JNK. PRL may promote Th17 infiltration into psoriatic lesions via CCL20.
Collapse
Affiliation(s)
- Naoko Kanda
- Department of Dermatology, Teikyo University School of Medicine, Tokyo, Japan.
| | | | | | | | | | | |
Collapse
|
44
|
Ha S, Redmond L. ERK mediates activity dependent neuronal complexity via sustained activity and CREB-mediated signaling. Dev Neurobiol 2009; 68:1565-79. [PMID: 18837011 DOI: 10.1002/dneu.20682] [Citation(s) in RCA: 36] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Abstract
A major question in the process of dendrite development and complexity is not whether neuronal activity plays a role, but how it contributes to specific components of the mature dendrite pattern. Neurons interpret activity into the influx of calcium ions leading to activation of signaling pathways. The dynamics of calcium-activated signaling pathways after neuronal activity and the contribution to formation of dendrite complexity remain unclear. Here, we show that one calcium activated signaling pathway, extracellular signal-regulated kinase (ERK), showed differential activity in cortical neurons. In response to depolarizing stimuli, ERK was active for less than an hour in most neurons, whereas in others ERK remained active for several hours. Further, neurons in which ERK activity was sustained, displayed greater dendrite complexity than neurons that did not display sustained ERK activity. Interestingly, this difference in dendrite complexity was detected in some, but not all, morphological parameters. Pharmacological inhibition of sustained ERK activity inhibited calcium-activated dendrite complexity. Increasing the duration and degree of ERK phosphorylation, and thus activity, with dominant negative MAP kinase phosphatase-1 accentuated dendrite complexity. Neurons in which ERK activity was sustained activated downstream nuclear targets including RSK, MSK, cAMP response element binding protein (CREB), CRE-mediated gene transcription, and stabilized c-Fos. Further, the increase in dendrite complexity mediated by sustained ERK activity was inhibited by expression of a dominant negative CREB. These data indicate that ERK-mediated activity induced dendrite complexity via sustained signaling and CREB-mediated signaling.
Collapse
Affiliation(s)
- Seungshin Ha
- Department of Pharmacology and Toxicology, Medical College of Georgia, Augusta, Georgia 30912, USA
| | | |
Collapse
|
45
|
Bose C, Udupa KB. Erythropoietin enhancement of rat pancreatic tumor cell proliferation requires the activation of ERK and JNK signals. Am J Physiol Cell Physiol 2008; 295:C394-405. [DOI: 10.1152/ajpcell.00423.2007] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/17/2023]
Abstract
Erythropoietin (EPO) regulates the proliferation and differentiation of erythroid cells by binding to its specific transmembrane receptor EPOR. Recent studies, however, have shown that the EPOR is additionally present in various cancer cells and EPO induces the proliferation of these cells, suggesting a different function for EPO other than erythropoiesis. Therefore, the purpose of the present study was to examine EPOR expression and the role of EPO in the proliferation and signaling cascades involved in this process, using the rat pancreatic tumor cell line AR42J. Our results showed that AR42J cells expressed EPOR, and EPO significantly enhanced their proliferation. Cell cycle analysis of EPO-treated cells indicated an increased percentage of cells in the S phase, whereas cell numbers in G0/G1 phase were significantly reduced. Phosphorylation of extracellular regulatory kinase 1/2 (ERK1/2) and c-Jun NH2terminal kinase 1/2 (JNK1/2) was rapidly stimulated and sustained after EPO addition. Treatment of cells with mitogen-activated protein/ERK kinase (MEK) inhibitor PD98059 or JNK inhibitor SP600125 significantly inhibited EPO-enhanced proliferation and also increased the fraction of cells in G0/G1 phase. Furthermore, the inhibition of JNK using small interference RNA (siRNA) suppressed EPO-enhanced proliferation of AR42J cells. Taken together, our results indicate that AR42J cells express EPOR and that the activation of both ERK1/2 and JNK1/2 by EPO is essential in regulating proliferation and the cell cycle. Thus both appear to play a key role in EPO-enhanced proliferation and suggest that the presence of both is required for EPO-mediated proliferation of AR42J cells.
Collapse
|
46
|
Park SW, He Y, Ha SG, Loh HH, Wei LN. Epigenetic regulation of kappa opioid receptor gene in neuronal differentiation. Neuroscience 2008; 151:1034-41. [PMID: 18201839 DOI: 10.1016/j.neuroscience.2007.12.015] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2007] [Revised: 12/03/2007] [Accepted: 12/07/2007] [Indexed: 01/09/2023]
Abstract
The gene of mouse kappa opioid receptor (KOR) utilizes two promoters, P1 and P2. P1 is active in various brain areas and constitutively in P19 mouse embryonal carcinoma cells. P2 is active in limited brain stem areas of adult animals and only in late differentiated cells of P19 induced for neuronal differentiation in the presence of nerve growth factor (NGF). NGF response of P2 was found to be mediated by a specific binding site for transcription factor activation protein 2 (AP2) located in P2. Electrophoretic gel shift assay showed specific binding of this AP2 site by AP2beta, but not AP2alpha. Knockdown of endogenous AP2beta with siRNA abolished the stimulating effect of NGF on the expression of transcripts driven by P2. Binding of endogenous AP2beta on the endogenous KOR P2 chromatin region was also confirmed by chromatin immunoprecipitation. The effect of NGF was inhibited by LY2942002 (phosphatidylinositol 3-kinase, PI3K inhibitor), suggesting that PI3K was involved in signaling pathway mediating the effect of NGF stimulation on KOR P2. The chromatin of P2 in P19 was found to be specifically modified following NGF stimulation, which included demethylation at Lys9 and dimethylation at Lys4 of histone H3 and was consistent with the increased recruitment of RNA polymerase II to this promoter. This study presents the first evidence for epigenetic changes occurred on a specific KOR promoter triggered by NGF in cells undergoing neuronal differentiation. This epigenetic change is mediated by recruited AP2beta to this promoter and involves the PI3K system.
Collapse
Affiliation(s)
- S W Park
- Department of Pharmacology, University of Minnesota Medical School, 6-120 Jackson Hall, 321 Church Street Southeast, Minneapolis, MN 55455, USA
| | | | | | | | | |
Collapse
|
47
|
Kanda N, Watanabe S. Histamine enhances the production of human beta-defensin-2 in human keratinocytes. Am J Physiol Cell Physiol 2007; 293:C1916-23. [PMID: 17928537 DOI: 10.1152/ajpcell.00293.2007] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
The anti-microbial peptide human beta-defensin-2 (hBD-2), produced by epidermal keratinocytes, plays pivotal roles in anti-microbial defense, inflammatory dermatoses, and wound repair. hBD-2 induces histamine release from mast cells. We examined the in vitro effects of histamine on hBD-2 production in normal human keratinocytes. Histamine enhanced TNF-alpha- or IFN-gamma-induced hBD-2 secretion and mRNA expression. Histamine alone enhanced transcriptional activities of NF-kappaB and activator protein-1 (AP-1) and potentiated TNF-alpha-induced NF-kappaB and AP-1 activities or IFN-gamma-induced NF-kappaB and STAT1 activities. Antisense oligonucleotides against NF-kappaB components p50 and p65, AP-1 components c-Jun and c-Fos, or H1 antagonist pyrilamine suppressed hBD-2 production induced by histamine plus TNF-alpha or IFN-gamma. Antisense oligonucleotide against STAT1 only suppressed hBD-2 production induced by histamine plus IFN-gamma. Histamine induced serine phosphorylation of inhibitory NF-kappaBalpha (IkappaBalpha) alone or together with TNF-alpha or IFN-gamma. Histamine induced c-Fos mRNA expression alone or together with TNF-alpha, whereas it did not further increase c-Jun mRNA levels enhanced by TNF-alpha. Histamine induced serine phosphorylation of STAT1 alone or together with IFN-gamma, whereas it did not further enhance IFN-gamma-induced tyrosine phosphorylation of STAT1. The histamine-induced serine phosphorylation of STAT1 was suppressed by MAPKK (MEK) inhibitor PD98059. These results suggest that histamine stimulates H1 receptor and potentiates TNF-alpha- or IFN-gamma-induced hBD-2 production dependent on NF-kappaB, AP-1, or STAT1 in human keratinocytes. Histamine may potentiate anti-microbial defense, skin inflammation, and wound repair via the induction of hBD-2.
Collapse
Affiliation(s)
- Naoko Kanda
- Department of Dermatology, Teikyo University School of Medicine, Tokyo, Japan
| | | |
Collapse
|