1
|
Kim Y, Kim SH, Lim J, Kim SH. ATBS1-INTERACTING FACTOR 2 Positively Regulates Freezing Tolerance via INDUCER OF CBF EXPRESSION 1/C-REPEAT BINDING FACTOR-Induced Cold Acclimation Pathway. PLANT & CELL PHYSIOLOGY 2024; 65:1363-1376. [PMID: 38957969 DOI: 10.1093/pcp/pcae072] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/13/2023] [Accepted: 07/02/2024] [Indexed: 07/04/2024]
Abstract
The INDUCER OF CBF EXPRESSION 1/C-REPEAT BINDING FACTOR (ICE1/CBF) pathway plays a crucial role in plant responses to cold stress, impacting growth and development. Here, we demonstrated that ATBS1-INTERACTING FACTOR 2 (AIF2), a non-DNA-binding basic helix-loop-helix transcription factor, positively regulates freezing tolerance through the ICE1/CBF-induced cold tolerance pathway in Arabidopsis. Cold stress transcriptionally upregulated AIF2 expression and induced AIF2 phosphorylation, thereby stabilizing the AIF2 protein during early stages of cold acclimation. The AIF2 loss-of-function mutant, aif2-1, exhibited heightened sensitivity to freezing before and after cold acclimation. In contrast, ectopic expression of AIF2, but not the C-terminal-deleted AIF2 variant, restored freezing tolerance. AIF2 enhanced ICE1 stability during cold acclimation and promoted the transcriptional expression of CBFs and downstream cold-responsive genes, ultimately enhancing plant tolerance to freezing stress. MITOGEN-ACTIVATED PROTEIN KINASES 3 and 6 (MPK3/6), known negative regulators of freezing tolerance, interacted with and phosphorylated AIF2, subjecting it to protein degradation. Furthermore, transient co-expression of MPK3/6 with AIF2 and ICE1 downregulated AIF2/ICE1-induced transactivation of CBF2 expression. AIF2 interacted preferentially with BRASSINOSTEROID-INSENSITIVE 2 (BIN2) and MPK3/6 during the early and later stages of cold acclimation, respectively, thereby differentially regulating AIF2 activity in a cold acclimation time-dependent manner. Moreover, AIF2 acted additively in a gain-of-function mutant of BRASSINAZOLE-RESISTANT 1 (BZR1; bzr1-1D) and a triple knockout mutant of BIN2 and its homologs (bin2bil1bil2) to induce CBFs-mediated freezing tolerance. This suggests that cold-induced AIF2 coordinates freezing tolerance along with BZR1 and BIN2, key positive and negative components, respectively, of brassinosteroid signaling pathways.
Collapse
Affiliation(s)
- Yoon Kim
- Division of Biological Science and Technology, Yonsei University, 1 Yonseidae-Gil, Wonju-Si 220-710, Republic of Korea
| | - Sun-Ho Kim
- Division of Biological Science and Technology, Yonsei University, 1 Yonseidae-Gil, Wonju-Si 220-710, Republic of Korea
| | - Jun Lim
- Department of Systems Biotechnology, Konkuk University, Gwangjin-gu, Seoul 05029, Republic of Korea
| | - Soo-Hwan Kim
- Division of Biological Science and Technology, Yonsei University, 1 Yonseidae-Gil, Wonju-Si 220-710, Republic of Korea
| |
Collapse
|
2
|
Drożdż A, Duggan B, Ruddock MW, Reid CN, Kurth MJ, Watt J, Irvine A, Lamont J, Fitzgerald P, O’Rourke D, Curry D, Evans M, Boyd R, Sousa J. Stratifying risk of disease in haematuria patients using machine learning techniques to improve diagnostics. Front Oncol 2024; 14:1401071. [PMID: 38779086 PMCID: PMC11109371 DOI: 10.3389/fonc.2024.1401071] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2024] [Accepted: 04/22/2024] [Indexed: 05/25/2024] Open
Abstract
Background Detailed and invasive clinical investigations are required to identify the causes of haematuria. Highly unbalanced patient population (predominantly male) and a wide range of potential causes make the ability to correctly classify patients and identify patient-specific biomarkers a major challenge. Studies have shown that it is possible to improve the diagnosis using multi-marker analysis, even in unbalanced datasets, by applying advanced analytical methods. Here, we applied several machine learning algorithms to classify patients from the haematuria patient cohort (HaBio) by analysing multiple biomarkers and to identify the most relevant ones. Materials and methods We applied several classification and feature selection methods (k-means clustering, decision trees, random forest with LIME explainer and CACTUS algorithm) to stratify patients into two groups: healthy (with no clear cause of haematuria) or sick (with an identified cause of haematuria e.g., bladder cancer, or infection). The classification performance of the models was compared. Biomarkers identified as important by the algorithms were also analysed in relation to their involvement in the pathological processes. Results Results showed that a high unbalance in the datasets significantly affected the classification by random forest and decision trees, leading to the overestimation of the sick class and low model performance. CACTUS algorithm was more robust to the unbalance in the dataset. CACTUS obtained a balanced accuracy of 0.747 for both genders, 0.718 for females and 0.803 for males. The analysis showed that in the classification process for the whole dataset: microalbumin, male gender, and tPSA emerged as the most informative biomarkers. For males: age, microalbumin, tPSA, cystatin C, BTA, HAD and S100A4 were the most significant biomarkers while for females microalbumin, IL-8, pERK, and CXCL16. Conclusions CACTUS algorithm demonstrated improved performance compared with other methods such as decision trees and random forest. Additionally, we identified the most relevant biomarkers for the specific patient group, which could be considered in the future as novel biomarkers for diagnosis. Our results have the potential to inform future research and provide new personalised diagnostic approaches tailored directly to the needs of the individuals.
Collapse
Affiliation(s)
- Anna Drożdż
- Personal Health Data Science Group, Sano – Centre for Computational Personalised Medicine - International Research Foundation, Krakow, Poland
| | - Brian Duggan
- South Eastern Health and Social Care Trust, Ulster Hospital Dundonald, Belfast, United Kingdom
| | - Mark W. Ruddock
- Clinical Studies Group, Randox Laboratories Ltd., Co., Antrim, United Kingdom
| | - Cherith N. Reid
- Clinical Studies Group, Randox Laboratories Ltd., Co., Antrim, United Kingdom
| | - Mary Jo Kurth
- Clinical Studies Group, Randox Laboratories Ltd., Co., Antrim, United Kingdom
| | - Joanne Watt
- Clinical Studies Group, Randox Laboratories Ltd., Co., Antrim, United Kingdom
| | - Allister Irvine
- Clinical Studies Group, Randox Laboratories Ltd., Co., Antrim, United Kingdom
| | - John Lamont
- Clinical Studies Group, Randox Laboratories Ltd., Co., Antrim, United Kingdom
| | - Peter Fitzgerald
- Clinical Studies Group, Randox Laboratories Ltd., Co., Antrim, United Kingdom
| | - Declan O’Rourke
- Belfast Health and Social Care Trust, Belfast City Hospital, Belfast, United Kingdom
| | - David Curry
- Belfast Health and Social Care Trust, Belfast City Hospital, Belfast, United Kingdom
| | - Mark Evans
- Belfast Health and Social Care Trust, Belfast City Hospital, Belfast, United Kingdom
| | - Ruth Boyd
- Northern Ireland Clinical Trials Network, Belfast City Hospital, Belfast, United Kingdom
| | - Jose Sousa
- Personal Health Data Science Group, Sano – Centre for Computational Personalised Medicine - International Research Foundation, Krakow, Poland
- Centre for Public Health, Institute of Clinical Sciences, Queen’s University, Belfast, United Kingdom
| |
Collapse
|
3
|
Anastasi E, Farina A, Granato T, Colaiacovo F, Pucci B, Tartaglione S, Angeloni A. Recent Insight about HE4 Role in Ovarian Cancer Oncogenesis. Int J Mol Sci 2023; 24:10479. [PMID: 37445657 DOI: 10.3390/ijms241310479] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2023] [Revised: 06/19/2023] [Accepted: 06/20/2023] [Indexed: 07/15/2023] Open
Abstract
Currently, ovarian cancer (OC) is a target of intense biomarkers research because of its frequent late diagnosis and poor prognosis. Serum determination of Human epididymis protein 4 (HE4) is a very important early detection test. Most interestingly, HE4 plays a unique role in OC as it has been implicated not only in OC diagnosis but also in the prognosis and recurrence of this lethal neoplasm, actually acting as a clinical biomarker. There are several evidence about the predictive power of HE4 clinically, conversely less has been described concerning its role in OC oncogenesis. Based on these considerations, the main goal of this review is to clarify the role of HE4 in OC proliferation, angiogenesis, metastatization, immune response and also in the development of targeted therapy. Through a deeper understanding of its functions as a key molecule in the oncogenetic processes underlying OC, HE4 could be possibly considered as an essential resource not only for diagnosis but also for prognosis and therapy choice.
Collapse
Affiliation(s)
- Emanuela Anastasi
- Department of Experimental Medicine, Sapienza University of Rome, Viale Regina Elena 324, 00161 Rome, Italy
| | - Antonella Farina
- Department of Experimental Medicine, Sapienza University of Rome, Viale Regina Elena 324, 00161 Rome, Italy
| | - Teresa Granato
- CNR-IBPM, Institute of Molecular Biology and Pathology, Department of Molecular Medicine, Sapienza University of Rome, Viale Regina Elena 324, 00161 Rome, Italy
| | - Flavia Colaiacovo
- Department of Experimental Medicine, Sapienza University of Rome, Viale Regina Elena 324, 00161 Rome, Italy
| | - Beatrice Pucci
- Department of Experimental Medicine, Sapienza University of Rome, Viale Regina Elena 324, 00161 Rome, Italy
| | - Sara Tartaglione
- Department of Experimental Medicine, Sapienza University of Rome, Viale Regina Elena 324, 00161 Rome, Italy
| | - Antonio Angeloni
- Department of Experimental Medicine, Sapienza University of Rome, Viale Regina Elena 324, 00161 Rome, Italy
| |
Collapse
|
4
|
Fu L, Adu-Amankwaah J, Sang L, Tang Z, Gong Z, Zhang X, Li T, Sun H. Gender differences in GRK2 in cardiovascular diseases and its interactions with estrogen. Am J Physiol Cell Physiol 2023; 324:C505-C516. [PMID: 36622065 DOI: 10.1152/ajpcell.00407.2022] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023]
Abstract
G protein-coupled receptor kinase 2 (GRK2) is a multifunctional protein involved in regulating G protein-coupled receptor (GPCR) and non-GPCR signaling in the body. In the cardiovascular system, increased expression of GRK2 has been implicated in the occurrence and development of several cardiovascular diseases (CVDs). Recent studies have found gender differences in GRK2 in the cardiovascular system under physiological and pathological conditions, where GRK2's expression and activity are increased in males than in females. The incidence of CVDs in premenopausal women is lower than in men of the same age, which is related to estrogen levels. Given the shared location of GRK2 and estrogen receptors, estrogen may interact with GRK2 by modulating vital molecules such as calmodulin (CaM), caveolin, RhoA, nitrate oxide (NO), and mouse double minute 2 homolog (Mdm2), via signaling pathways mediated by estrogen's genomic (ERα and ERβ), and non-genomic (GPER) receptors, conferring cardiovascular protection in females. Highlighting the gender differences in GRK2 and understanding its interaction with estrogen in the cardiovascular system is pertinent in treating gender-related CVDs. As a result, this article explores the gender differences of GRK2 in the cardiovascular system and its relationship with estrogen during disease conditions. Estrogen's protective and therapeutic effects and its mechanism on GRK2-related cardiovascular diseases have also been discussed.
Collapse
Affiliation(s)
- Lu Fu
- Department of Physiology, Xuzhou Medical University, Xuzhou, People's Republic of China
| | - Joseph Adu-Amankwaah
- Department of Physiology, Xuzhou Medical University, Xuzhou, People's Republic of China
| | - Lili Sang
- Department of Physiology, Xuzhou Medical University, Xuzhou, People's Republic of China.,National Demonstration Center for Experimental Basic Medical Science Education, Xuzhou Medical University, Xuzhou, People's Republic of China
| | - Ziqing Tang
- Department of Physiology, Xuzhou Medical University, Xuzhou, People's Republic of China
| | - Zheng Gong
- Department of Physiology, Xuzhou Medical University, Xuzhou, People's Republic of China.,School of Public Affairs & Governance, Silliman University, Dumaguete, Philippines
| | - Xiaoyan Zhang
- Department of Physiology, Xuzhou Medical University, Xuzhou, People's Republic of China
| | - Tao Li
- Department of Physiology, Xuzhou Medical University, Xuzhou, People's Republic of China
| | - Hong Sun
- Department of Physiology, Xuzhou Medical University, Xuzhou, People's Republic of China
| |
Collapse
|
5
|
Yu Z, Ma J, Zhang M, Li X, Sun Y, Zhang M, Ding Z. Auxin promotes hypocotyl elongation by enhancing BZR1 nuclear accumulation in Arabidopsis. SCIENCE ADVANCES 2023; 9:eade2493. [PMID: 36598987 PMCID: PMC9812374 DOI: 10.1126/sciadv.ade2493] [Citation(s) in RCA: 21] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 08/04/2022] [Accepted: 11/29/2022] [Indexed: 06/17/2023]
Abstract
Auxin and brassinosteroids (BRs) are two major growth-promoting phytohormones that shape hypocotyl elongation; however, the cross-talk between auxin and BR in this process is not fully understood. In this study, we found that auxin-induced hypocotyl elongation is dependent on brassinazole-resistant 1 (BZR1), a core BR signaling component. Auxin promotes BZR1 nuclear accumulation in hypocotyl cells, a process dependent on mitogen-activated protein kinase 3 (MPK3) and MPK6, which are both activated by auxin and whose encoding genes are highly expressed in hypocotyls. We determined that MPK3/MPK6 phosphorylate and reduce the protein stability of general regulatory factor 4 (GRF4), a member of the 14-3-3 family of proteins that retain BZR1 in the cytoplasm. In summary, this study reveals the molecular mechanism by which auxin promotes hypocotyl elongation by enhancing BZR1 nuclear accumulation via MPK3/MPK6-regulated GRF4 protein stability.
Collapse
Affiliation(s)
- Zipeng Yu
- The Key Laboratory of Plant Development and Environmental Adaptation Biology, Ministry of Education, School of Life Sciences, Shandong University, Qingdao, Shandong, China
| | - Jinxin Ma
- The Key Laboratory of Plant Development and Environmental Adaptation Biology, Ministry of Education, School of Life Sciences, Shandong University, Qingdao, Shandong, China
| | - Mengyue Zhang
- The Key Laboratory of Plant Development and Environmental Adaptation Biology, Ministry of Education, School of Life Sciences, Shandong University, Qingdao, Shandong, China
| | - Xiaoxuan Li
- The Key Laboratory of Plant Development and Environmental Adaptation Biology, Ministry of Education, School of Life Sciences, Shandong University, Qingdao, Shandong, China
| | - Yi Sun
- The Key Laboratory of Plant Development and Environmental Adaptation Biology, Ministry of Education, School of Life Sciences, Shandong University, Qingdao, Shandong, China
| | - Mengxin Zhang
- The Key Laboratory of Plant Development and Environmental Adaptation Biology, Ministry of Education, School of Life Sciences, Shandong University, Qingdao, Shandong, China
| | - Zhaojun Ding
- The Key Laboratory of Plant Development and Environmental Adaptation Biology, Ministry of Education, School of Life Sciences, Shandong University, Qingdao, Shandong, China
| |
Collapse
|
6
|
Kim E, Suh JS, Jang YK, Kim H, Choi G, Kim TJ. Harmine inhibits proliferation and migration of glioblastoma via ERK signalling. Process Biochem 2022. [DOI: 10.1016/j.procbio.2022.09.014] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
|
7
|
Cheng H, Guo P, Su T, Jiang C, Zhu Z, Wei W, Zhang L, Wang Q. G protein-coupled receptor kinase type 2 and β-arrestin2: Key players in immune cell functions and inflammation. Cell Signal 2022; 95:110337. [PMID: 35461901 DOI: 10.1016/j.cellsig.2022.110337] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2022] [Revised: 04/15/2022] [Accepted: 04/15/2022] [Indexed: 02/07/2023]
|
8
|
Kuai J, Han C, Wei W. Potential Regulatory Roles of GRK2 in Endothelial Cell Activity and Pathological Angiogenesis. Front Immunol 2021; 12:698424. [PMID: 34335610 PMCID: PMC8320431 DOI: 10.3389/fimmu.2021.698424] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2021] [Accepted: 06/21/2021] [Indexed: 12/16/2022] Open
Abstract
G protein-coupled receptor (GPCR) kinase 2 (GRK2) is an integrative node in many signaling network cascades. Emerging evidence indicates that GRK2 can interact with a large number of GPCRs and non-GPCR substrates in both kinase-dependent and -independent modes. Some of these pathways are associated with endothelial cell (EC) activity. The active state of ECs is a pivotal factor in angiogenesis. The occurrence and development of some inflammation-related diseases are accompanied by pathological angiogenesis, but there remains a lack of effective targeted treatments. Alterations in the expression and/or localization of GRK2 have been identified in several types of diseases and have been demonstrated to regulate the angiogenesis process in these diseases. GRK2 as a target may be a promising candidate for anti-angiogenesis therapy.
Collapse
Affiliation(s)
| | | | - Wei Wei
- Institute of Clinical Pharmacology, Key Laboratory of Anti-Inflammatory and Immune Medicine (Anhui Medical University), Ministry of Education, Anhui Collaborative Innovation Center of Anti-inflammatory and Immune Medicine, Anhui Medical University, Hefei, China
| |
Collapse
|
9
|
Nagabushan S, Lau LMS, Barahona P, Wong M, Sherstyuk A, Marshall GM, Tyrrell V, Wegner EA, Ekert PG, Cowley MJ, Mayoh C, Trahair TN, Crowe P, Anazodo A, Ziegler DS. Efficacy of MEK inhibition in a recurrent malignant peripheral nerve sheath tumor. NPJ Precis Oncol 2021; 5:9. [PMID: 33580196 PMCID: PMC7881142 DOI: 10.1038/s41698-021-00145-8] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2020] [Accepted: 01/06/2021] [Indexed: 01/30/2023] Open
Abstract
The prognosis of recurrent malignant peripheral nerve sheath tumors (MPNST) is dismal, with surgical resection being the only definitive salvage therapy. Treatment with chemoradiation approaches has not significantly improved patient outcomes. Similarly, trials of therapies targeting MPNST genomic drivers have thus far been unsuccessful. Improved understanding of the molecular pathogenesis of MPNST indicates frequent activation of the mitogen-activated protein kinase (MAPK) cell signaling pathway. MEK inhibitors have shown activity in preclinical studies; however, their clinical efficacy has not been reported to date. We describe here a case of sustained complete response to MEK inhibition in an adolescent patient with a recurrent metastatic MPNST with multiple alterations in the MAPK pathway, guided by a precision oncology approach.
Collapse
Affiliation(s)
- Sumanth Nagabushan
- grid.414009.80000 0001 1282 788XKids Cancer Centre, Sydney Children’s Hospital, Randwick, NSW Australia ,grid.1005.40000 0004 4902 0432School of Women’s and Children’s Health, UNSW Sydney, Sydney, NSW Australia
| | - Loretta M. S. Lau
- grid.414009.80000 0001 1282 788XKids Cancer Centre, Sydney Children’s Hospital, Randwick, NSW Australia ,grid.1005.40000 0004 4902 0432School of Women’s and Children’s Health, UNSW Sydney, Sydney, NSW Australia ,grid.1005.40000 0004 4902 0432Children’s Cancer Institute, Lowy Cancer Research Centre, UNSW Sydney, Sydney, NSW Australia
| | - Paulette Barahona
- grid.1005.40000 0004 4902 0432Children’s Cancer Institute, Lowy Cancer Research Centre, UNSW Sydney, Sydney, NSW Australia
| | - Marie Wong
- grid.1005.40000 0004 4902 0432School of Women’s and Children’s Health, UNSW Sydney, Sydney, NSW Australia ,grid.1005.40000 0004 4902 0432Children’s Cancer Institute, Lowy Cancer Research Centre, UNSW Sydney, Sydney, NSW Australia
| | - Alexandra Sherstyuk
- grid.1005.40000 0004 4902 0432Children’s Cancer Institute, Lowy Cancer Research Centre, UNSW Sydney, Sydney, NSW Australia
| | - Glenn M. Marshall
- grid.414009.80000 0001 1282 788XKids Cancer Centre, Sydney Children’s Hospital, Randwick, NSW Australia ,grid.1005.40000 0004 4902 0432School of Women’s and Children’s Health, UNSW Sydney, Sydney, NSW Australia ,grid.1005.40000 0004 4902 0432Children’s Cancer Institute, Lowy Cancer Research Centre, UNSW Sydney, Sydney, NSW Australia
| | - Vanessa Tyrrell
- grid.1005.40000 0004 4902 0432Children’s Cancer Institute, Lowy Cancer Research Centre, UNSW Sydney, Sydney, NSW Australia
| | - Eva A. Wegner
- grid.415193.bDepartment of Nuclear Medicine and PET, Sydney Children’s Hospital and Prince of Wales Hospital, Randwick, NSW Australia ,grid.1005.40000 0004 4902 0432Prince of Wales Clinical School, UNSW Sydney, Sydney, NSW Australia
| | - Paul G. Ekert
- grid.1005.40000 0004 4902 0432School of Women’s and Children’s Health, UNSW Sydney, Sydney, NSW Australia ,grid.1005.40000 0004 4902 0432Children’s Cancer Institute, Lowy Cancer Research Centre, UNSW Sydney, Sydney, NSW Australia
| | - Mark J. Cowley
- grid.1005.40000 0004 4902 0432School of Women’s and Children’s Health, UNSW Sydney, Sydney, NSW Australia ,grid.1005.40000 0004 4902 0432Children’s Cancer Institute, Lowy Cancer Research Centre, UNSW Sydney, Sydney, NSW Australia
| | - Chelsea Mayoh
- grid.1005.40000 0004 4902 0432School of Women’s and Children’s Health, UNSW Sydney, Sydney, NSW Australia ,grid.1005.40000 0004 4902 0432Children’s Cancer Institute, Lowy Cancer Research Centre, UNSW Sydney, Sydney, NSW Australia
| | - Toby N. Trahair
- grid.414009.80000 0001 1282 788XKids Cancer Centre, Sydney Children’s Hospital, Randwick, NSW Australia ,grid.1005.40000 0004 4902 0432School of Women’s and Children’s Health, UNSW Sydney, Sydney, NSW Australia ,grid.1005.40000 0004 4902 0432Children’s Cancer Institute, Lowy Cancer Research Centre, UNSW Sydney, Sydney, NSW Australia
| | - Philip Crowe
- grid.1005.40000 0004 4902 0432Prince of Wales Clinical School, UNSW Sydney, Sydney, NSW Australia ,grid.415193.bNelune Comprehensive Cancer Centre, Prince of Wales Hospital, Randwick, NSW Australia ,grid.1005.40000 0004 4902 0432Sydney Sarcoma Unit, UNSW Sydney, Sydney, NSW Australia
| | - Antoinette Anazodo
- grid.414009.80000 0001 1282 788XKids Cancer Centre, Sydney Children’s Hospital, Randwick, NSW Australia ,grid.1005.40000 0004 4902 0432School of Women’s and Children’s Health, UNSW Sydney, Sydney, NSW Australia ,grid.415193.bNelune Comprehensive Cancer Centre, Prince of Wales Hospital, Randwick, NSW Australia
| | - David S. Ziegler
- grid.414009.80000 0001 1282 788XKids Cancer Centre, Sydney Children’s Hospital, Randwick, NSW Australia ,grid.1005.40000 0004 4902 0432School of Women’s and Children’s Health, UNSW Sydney, Sydney, NSW Australia ,grid.1005.40000 0004 4902 0432Children’s Cancer Institute, Lowy Cancer Research Centre, UNSW Sydney, Sydney, NSW Australia
| |
Collapse
|
10
|
Lv B, Wei K, Hu K, Tian T, Zhang F, Yu Z, Zhang D, Su Y, Sang Y, Zhang X, Ding Z. MPK14-mediated auxin signaling controls lateral root development via ERF13-regulated very-long-chain fatty acid biosynthesis. MOLECULAR PLANT 2021; 14:285-297. [PMID: 33221411 DOI: 10.1016/j.molp.2020.11.011] [Citation(s) in RCA: 57] [Impact Index Per Article: 14.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/16/2020] [Revised: 10/28/2020] [Accepted: 11/15/2020] [Indexed: 05/16/2023]
Abstract
Auxin plays a critical role in lateral root (LR) formation. The signaling module composed of auxin-response factors (ARFs) and lateral organ boundaries domain transcription factors mediates auxin signaling to control almost every stage of LR development. Here, we show that auxin-induced degradation of the APETALA2/Ethylene Responsive Factor (AP2/ERF) transcription factor ERF13, dependent on MITOGEN-ACTIVATED PROTEIN KINASE MPK14-mediated phosphorylation, plays an essential role in LR development. Overexpression of ERF13 results in restricted passage of the LR primordia through the endodermal layer, greatly reducing LR emergence, whereas the erf13 mutants showed an increase in emerged LR. ERF13 inhibits the expression of 3-ketoacyl-CoA synthase16 (KCS16), which encodes a fatty acid elongase involved in very-long-chain fatty acid (VLCFA) biosynthesis. Overexpression of KCS16 or exogenous VLCFA treatment rescues the LR emergence defects in ERF13 overexpression lines, indicating a role downstream of the auxin-MPK14-ERF13 signaling module. Collectively, our study uncovers a novel molecular mechanism by which MPK14-mediated auxin signaling modulates LR development via ERF13-regulated VLCFA biosynthesis.
Collapse
Affiliation(s)
- Bingsheng Lv
- The Key Laboratory of Plant Development and Environmental Adaptation Biology, Ministry of Education, School of Life Sciences, Shandong University, Qingdao, Shandong 266237, China; College of Horticulture, Qingdao Agricultural University, Qingdao, Shandong 266109, China
| | - Kaijing Wei
- The Key Laboratory of Plant Development and Environmental Adaptation Biology, Ministry of Education, School of Life Sciences, Shandong University, Qingdao, Shandong 266237, China
| | - Kongqin Hu
- The Key Laboratory of Plant Development and Environmental Adaptation Biology, Ministry of Education, School of Life Sciences, Shandong University, Qingdao, Shandong 266237, China
| | - Te Tian
- The Key Laboratory of Plant Development and Environmental Adaptation Biology, Ministry of Education, School of Life Sciences, Shandong University, Qingdao, Shandong 266237, China
| | - Feng Zhang
- The Key Laboratory of Plant Development and Environmental Adaptation Biology, Ministry of Education, School of Life Sciences, Shandong University, Qingdao, Shandong 266237, China
| | - Zipeng Yu
- The Key Laboratory of Plant Development and Environmental Adaptation Biology, Ministry of Education, School of Life Sciences, Shandong University, Qingdao, Shandong 266237, China
| | - Dajian Zhang
- State Key Laboratory of Crop Biology, College of Agronomy, Shandong Agricultural University, Taian, Shandong 271018, China
| | - Yinghua Su
- State Key Laboratory of Crop Biology, College of Life Sciences, Shandong Agricultural University, Taian, Shandong 271018, China
| | - Yalin Sang
- State Key Laboratory of Crop Biology, College of Forestry, Shandong Agricultural University, Taian, Shandong 271018, China
| | - Xiansheng Zhang
- State Key Laboratory of Crop Biology, College of Life Sciences, Shandong Agricultural University, Taian, Shandong 271018, China
| | - Zhaojun Ding
- The Key Laboratory of Plant Development and Environmental Adaptation Biology, Ministry of Education, School of Life Sciences, Shandong University, Qingdao, Shandong 266237, China.
| |
Collapse
|
11
|
Penela P, Ribas C, Sánchez-Madrid F, Mayor F. G protein-coupled receptor kinase 2 (GRK2) as a multifunctional signaling hub. Cell Mol Life Sci 2019; 76:4423-4446. [PMID: 31432234 PMCID: PMC6841920 DOI: 10.1007/s00018-019-03274-3] [Citation(s) in RCA: 59] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2019] [Revised: 08/06/2019] [Accepted: 08/12/2019] [Indexed: 12/18/2022]
Abstract
Accumulating evidence indicates that G protein-coupled receptor kinase 2 (GRK2) is a versatile protein that acts as a signaling hub by modulating G protein-coupled receptor (GPCR) signaling and also via phosphorylation or scaffolding interactions with an extensive number of non-GPCR cellular partners. GRK2 multifunctionality arises from its multidomain structure and from complex mechanisms of regulation of its expression levels, activity, and localization within the cell, what allows the precise spatio-temporal shaping of GRK2 targets. A better understanding of the GRK2 interactome and its modulation mechanisms is helping to identify the GRK2-interacting proteins and its substrates involved in the participation of this kinase in different cellular processes and pathophysiological contexts.
Collapse
Affiliation(s)
- Petronila Penela
- Departamento de Biología Molecular, Centro de Biología Molecular "Severo Ochoa" (UAM-CSIC), Universidad Autónoma de Madrid, C/Nicolás Cabrera 1, 28049, Madrid, Spain
- Instituto de Investigación Sanitaria La Princesa, 28006, Madrid, Spain
- CIBER de Enfermedades Cardiovasculares, ISCIII (CIBERCV), 28029, Madrid, Spain
| | - Catalina Ribas
- Departamento de Biología Molecular, Centro de Biología Molecular "Severo Ochoa" (UAM-CSIC), Universidad Autónoma de Madrid, C/Nicolás Cabrera 1, 28049, Madrid, Spain
- Instituto de Investigación Sanitaria La Princesa, 28006, Madrid, Spain
- CIBER de Enfermedades Cardiovasculares, ISCIII (CIBERCV), 28029, Madrid, Spain
| | - Francisco Sánchez-Madrid
- Instituto de Investigación Sanitaria La Princesa, 28006, Madrid, Spain
- CIBER de Enfermedades Cardiovasculares, ISCIII (CIBERCV), 28029, Madrid, Spain
- Cell-Cell Communication Laboratory, Vascular Pathophysiology Area, Centro Nacional Investigaciones Cardiovasculares (CNIC), 28029, Madrid, Spain
| | - Federico Mayor
- Departamento de Biología Molecular, Centro de Biología Molecular "Severo Ochoa" (UAM-CSIC), Universidad Autónoma de Madrid, C/Nicolás Cabrera 1, 28049, Madrid, Spain.
- Instituto de Investigación Sanitaria La Princesa, 28006, Madrid, Spain.
- CIBER de Enfermedades Cardiovasculares, ISCIII (CIBERCV), 28029, Madrid, Spain.
| |
Collapse
|
12
|
Yu S, Sun L, Jiao Y, Lee LTO. The Role of G Protein-coupled Receptor Kinases in Cancer. Int J Biol Sci 2018; 14:189-203. [PMID: 29483837 PMCID: PMC5821040 DOI: 10.7150/ijbs.22896] [Citation(s) in RCA: 41] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2017] [Accepted: 11/17/2017] [Indexed: 01/14/2023] Open
Abstract
G protein-coupled receptors (GPCRs) are the largest family of plasma membrane receptors. Emerging evidence demonstrates that signaling through GPCRs affects numerous aspects of cancer biology such as vascular remolding, invasion, and migration. Therefore, development of GPCR-targeted drugs could provide a new therapeutic strategy to treating a variety of cancers. G protein-coupled receptor kinases (GRKs) modulate GPCR signaling by interacting with the ligand-activated GPCR and phosphorylating its intracellular domain. This phosphorylation initiates receptor desensitization and internalization, which inhibits downstream signaling pathways related to cancer progression. GRKs can also regulate non-GPCR substrates, resulting in the modulation of a different set of pathophysiological pathways. In this review, we will discuss the role of GRKs in modulating cell signaling and cancer progression, as well as the therapeutic potential of targeting GRKs.
Collapse
Affiliation(s)
- Shan Yu
- Centre of Reproduction Development and Aging, Faculty of Health Sciences, University of Macau, Taipa, Macau
| | - Litao Sun
- Department of Ultrasound, The Secondary Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Yufei Jiao
- Department of Pathology, The Secondary Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Leo Tsz On Lee
- Centre of Reproduction Development and Aging, Faculty of Health Sciences, University of Macau, Taipa, Macau
| |
Collapse
|
13
|
Herbomel GG, Rojas RE, Tran DT, Ajinkya M, Beck L, Tabak LA. The GalNAc-T Activation Pathway (GALA) is not a general mechanism for regulating mucin-type O-glycosylation. PLoS One 2017; 12:e0179241. [PMID: 28719662 PMCID: PMC5515409 DOI: 10.1371/journal.pone.0179241] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2017] [Accepted: 05/25/2017] [Indexed: 12/03/2022] Open
Abstract
Mucin-type O-glycosylation is initiated by the UDP-GalNAc polypeptide:N-acetylgalactosaminyltransferase (GalNAc-T) family of enzymes. Their activity results in the GalNAc α1-O-Thr/Ser structure, termed the Tn antigen, which is further decorated with additional sugars. In neoplastic cells, the Tn antigen is often overexpressed. Because O-glycosylation is controlled by the activity of GalNAc-Ts, their regulation is of great interest. Previous reports suggest that growth factors, EGF or PDGF, induce Golgi complex-to-endoplasmic reticulum (ER) relocation of both GalNAc-Ts and Tn antigen in HeLa cells, offering a mechanism for Tn antigen overexpression termed "GALA". However, we were unable to reproduce these findings. Upon treatment of HeLa cells with either EGF or PDGF we observed no change in the co-localization of endogenous GalNAc-T1, GalNAc-T2 or Tn antigen with the Golgi complex marker TGN46. There was also no enhancement of localization with the ER marker calnexin. We conclude that growth factors do not cause redistribution of GalNAc-Ts from the Golgi complex to the ER in HeLa cells.
Collapse
Affiliation(s)
- Gaetan G. Herbomel
- Section on Biological Chemistry, National Institute of Dental and Craniofacial Research, National Institutes of Health, Bethesda, Maryland, United States of America
| | - Raul E. Rojas
- Section on Biological Chemistry, National Institute of Dental and Craniofacial Research, National Institutes of Health, Bethesda, Maryland, United States of America
| | - Duy T. Tran
- Section on Biological Chemistry, National Institute of Dental and Craniofacial Research, National Institutes of Health, Bethesda, Maryland, United States of America
| | - Monica Ajinkya
- Section on Biological Chemistry, National Institute of Dental and Craniofacial Research, National Institutes of Health, Bethesda, Maryland, United States of America
| | - Lauren Beck
- Section on Biological Chemistry, National Institute of Dental and Craniofacial Research, National Institutes of Health, Bethesda, Maryland, United States of America
| | - Lawrence A. Tabak
- Section on Biological Chemistry, National Institute of Dental and Craniofacial Research, National Institutes of Health, Bethesda, Maryland, United States of America
| |
Collapse
|
14
|
Calcium Oxalate Induces Renal Injury through Calcium-Sensing Receptor. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2016; 2016:5203801. [PMID: 27965733 PMCID: PMC5124692 DOI: 10.1155/2016/5203801] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/31/2016] [Revised: 07/29/2016] [Accepted: 10/05/2016] [Indexed: 01/24/2023]
Abstract
Objective. To investigate whether calcium-sensing receptor (CaSR) plays a role in calcium-oxalate-induced renal injury. Materials and Methods. HK-2 cells and rats were treated with calcium oxalate (CaOx) crystals with or without pretreatment with the CaSR-specific agonist gadolinium chloride (GdCl3) or the CaSR-specific antagonist NPS2390. Changes in oxidative stress (OS) in HK-2 cells and rat kidneys were assessed. In addition, CaSR, extracellular signal-regulated protein kinase (ERK), c-Jun N-terminal protein kinase (JNK), and p38 expression was determined. Further, crystal adhesion assay was performed in vitro, and the serum urea and creatinine levels and crystal deposition in the kidneys were also examined. Results. CaOx increased CaSR, ERK, JNK, and p38 protein expression and OS in vitro and in vivo. These deleterious changes were further enhanced upon pretreatment with the CaSR agonist GdCl3 but were attenuated by the specific CaSR inhibitor NPS2390 compared with CaOx treatment alone. Pretreatment with GdCl3 further increased in vitro and in vivo crystal adhesion and renal hypofunction. In contrast, pretreatment with NPS2390 decreased in vitro and in vivo crystal adhesion and renal hypofunction. Conclusions. CaOx-induced renal injury is related to CaSR-mediated OS and increased mitogen-activated protein kinase (MAPK) signaling, which subsequently leads to CaOx crystal adhesion.
Collapse
|
15
|
Zhang L, Wang X, Cao H, Chen Y, Chen X, Zhao X, Xu F, Wang Y, Woo AYH, Zhu W. Vasopressin V 1A receptor mediates cell proliferation through GRK2-EGFR-ERK 1/2 pathway in A7r5 cells. Eur J Pharmacol 2016; 792:15-25. [PMID: 27773680 DOI: 10.1016/j.ejphar.2016.10.023] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2016] [Revised: 10/13/2016] [Accepted: 10/18/2016] [Indexed: 12/23/2022]
Abstract
Abnormal proliferation and hypertrophy of vascular smooth muscle (VSMC), as the main structural component of the vasculature, is an important pathological mechanism of hypertension. Recently, increased levels of arginine vasopressin (AVP) and copeptin, the C-terminal fragment of provasopressin, have been shown to correlate with the development of preeclampsia. AVP targets on the Gq-coupled vasopressin V1A receptor and the Gs-coupled V2 receptor in VSMC and the kidneys to regulate vascular tone and water homeostasis. However, the role of the vasopressin receptor on VSM cell proliferation during vascular remodeling is unclear. Here, we studied the effects of AVP on the proliferation of the rat VSMC-derived A7r5 cells. AVP, in a time- and concentration-dependent manner, promoted A7r5 cell proliferation as indicated by the induction of proliferating cell nuclear antigen expression, methylthiazolyldiphenyl-tetrazolium reduction and incorporation of 5'-bromodeoxyuridine into cellular DNA. These effects, coupled with the phosphorylation of extracellular signal-regulated kinase 1/2 (ERK1/2), were blocked by a V1A receptor antagonist SR45059 but not by a V2 receptor antagonist lixivaptan. Although acute activation of V1A receptor induced ERK1/2 phosphorylation via a protein kinase C-dependent pathway, this effect was not involved in cell proliferation. Cell proliferation and ERK1/2 phosphorylation in response to prolonged stimulation with AVP were abolished by inhibition of G protein-coupled receptor kinase 2 (GRK2) and epidermal growth factor receptor (EGFR) using specific inhibitors or small hairpin RNA knock-down. These results suggest that activation of V1A, but not V2 receptor, produces a cell proliferative signal in A7r5 cells via a GRK2/EGFR/ERK1/2-dependent mechanism.
Collapse
Affiliation(s)
- Lingling Zhang
- Cardiovascular laboratory, Department of Pharmacology, School of Pharmacy, Nantong University, Nantong 226001, China
| | - Xiaojun Wang
- Cardiovascular laboratory, Department of Pharmacology, School of Pharmacy, Nantong University, Nantong 226001, China
| | - Hong Cao
- Cardiovascular laboratory, Department of Pharmacology, School of Pharmacy, Nantong University, Nantong 226001, China
| | - Yunxuan Chen
- Cardiovascular laboratory, Department of Pharmacology, School of Pharmacy, Nantong University, Nantong 226001, China
| | - Xianfan Chen
- Cardiovascular laboratory, Department of Pharmacology, School of Pharmacy, Nantong University, Nantong 226001, China
| | - Xi Zhao
- Cardiovascular laboratory, Department of Pharmacology, School of Pharmacy, Nantong University, Nantong 226001, China
| | - Feifei Xu
- Cardiovascular laboratory, Department of Pharmacology, School of Pharmacy, Nantong University, Nantong 226001, China
| | - Yifan Wang
- Cardiovascular laboratory, Department of Pharmacology, School of Pharmacy, Nantong University, Nantong 226001, China
| | - Anthony Yiu-Ho Woo
- Department of Pharmacology, School of Life Sciences and Biopharmaceutics, Shenyang Pharmaceutical University, Shenyang 110016, China
| | - Weizhong Zhu
- Cardiovascular laboratory, Department of Pharmacology, School of Pharmacy, Nantong University, Nantong 226001, China.
| |
Collapse
|
16
|
Something old, something new and something borrowed: emerging paradigm of insulin-like growth factor type 1 receptor (IGF-1R) signaling regulation. Cell Mol Life Sci 2013; 71:2403-27. [PMID: 24276851 PMCID: PMC4055838 DOI: 10.1007/s00018-013-1514-y] [Citation(s) in RCA: 137] [Impact Index Per Article: 11.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2013] [Revised: 10/17/2013] [Accepted: 11/07/2013] [Indexed: 12/14/2022]
Abstract
The insulin-like growth factor type 1 receptor (IGF-1R) plays a key role in the development and progression of cancer; however, therapeutics targeting it have had disappointing results in the clinic. As a receptor tyrosine kinase (RTK), IGF-1R is traditionally described as an ON/OFF system, with ligand stabilizing the ON state and exclusive kinase-dependent signaling activation. Newly added to the traditional model, ubiquitin-mediated receptor downregulation and degradation was originally described as a response to ligand/receptor interaction and thus inseparable from kinase signaling activation. Yet, the classical model has proven over-simplified and insufficient to explain experimental evidence accumulated over the last decade, including kinase-independent signaling, unbalanced signaling, or dissociation between signaling and receptor downregulation. Based on the recent findings that IGF-1R “borrows” components of G-protein coupled receptor (GPCR) signaling, including β-arrestins and G-protein-related kinases, we discuss the emerging paradigm for the IGF-1R as a functional RTK/GPCR hybrid, which integrates the kinase signaling with the IGF-1R canonical GPCR characteristics. The contradictions to the classical IGF-1R signaling concept as well as the design of anti-IGF-1R therapeutics treatment are considered in the light of this paradigm shift and we advocate recognition of IGF-1R as a valid target for cancer treatment.
Collapse
|
17
|
Robinson JD, Pitcher JA. G protein-coupled receptor kinase 2 (GRK2) is a Rho-activated scaffold protein for the ERK MAP kinase cascade. Cell Signal 2013; 25:2831-9. [PMID: 24018045 DOI: 10.1016/j.cellsig.2013.08.031] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2013] [Revised: 08/30/2013] [Accepted: 08/31/2013] [Indexed: 11/20/2022]
Abstract
The G protein-coupled receptor kinases (GRKs) are best known for their role in phosphorylating and desensitising G protein-coupled receptors (GPCRs). The GRKs also regulate signalling downstream of other families of receptors and have a number of non-receptor substrates and binding partners. Here we identify RhoAGTP and Raf1 as novel binding partners of GRK2 and report a previously unsuspected function for this kinase. GRK2 is a RhoA effector that serves as a RhoA-activated scaffold protein for the ERK MAP kinase cascade. The ability of GRK2 to bind to Raf1, MEK1 and ERK2 is dependent on RhoAGTP binding to the catalytic domain of the kinase. Exogenous GRK2 has previously been shown to increase ERK activation downstream of the epidermal growth factor receptor (EGFR). Here we find that GRK2-mediated ERK activation downstream of the EGFR is Rho-dependent and that treatment with EGF promotes RhoAGTP binding and ERK scaffolding by GRK2. Depletion of GRK2 expression by RNAi reveals that GRK2 is required for EGF-induced, Rho- and ERK-dependent thymidine incorporation in vascular smooth muscle cells (VSMCs). We therefore hypothesise that Rho-dependent ERK MAPK scaffolding by GRK2 downstream of the EGFR may have an important role in the vasculature, where increased levels of both GRK2 and RhoA have been associated with hypertension.
Collapse
Affiliation(s)
- James D Robinson
- MRC Laboratory for Molecular Cell Biology, Research Department of Neuroscience, Physiology and Pharmacology, University College London, Gower Street, London WC1E 6BT, UK
| | | |
Collapse
|
18
|
Zhao H, Wu G, Cao X. EGFR dependent subcellular communication was responsible for morphine mediated AC superactivation. Cell Signal 2012; 25:417-28. [PMID: 23142605 DOI: 10.1016/j.cellsig.2012.10.016] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2012] [Revised: 10/01/2012] [Accepted: 10/01/2012] [Indexed: 10/27/2022]
Abstract
Compensatory adenylyl cyclase (AC) superactivation has been postulated to be responsible for the development of morphine tolerance and dependence, the underlying mechanism was demonstrated to comprise c-Src-dependent upregulation of AC5 within the lipid rafts. In the present study, we demonstrated that chronic morphine treatment sensitized EGFR signaling by augmenting EGFR phosphorylation and translocation into ER, which was essential for CRT-MOR tethering within the lipid rafts and AC5 superactivation. Intriguingly, synaptic clustering of CRT-MOR was dependent on EGFR phosphorylation and presumed to implicate in alignment and organization of synaptic compartments. Taken together, our data raised the possibility that an adaptive change in MOR and EGFR signal systems might establish CRT related subcellular communication, the signaling network within brain synaptic zone was proposed to implicate in morphine tolerance and dependence.
Collapse
Affiliation(s)
- Hui Zhao
- Department of Integrative Medicine and Neurobiology, National Key lab of Medical Neurobiology, Institute of Brain Research Sciences, Shanghai Medical College, Fudan University, PR China.
| | | | | |
Collapse
|
19
|
Cyclosporin A induces apoptosis in H9c2 cardiomyoblast cells through calcium-sensing receptor-mediated activation of the ERK MAPK and p38 MAPK pathways. Mol Cell Biochem 2012; 367:227-36. [DOI: 10.1007/s11010-012-1336-5] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2012] [Accepted: 05/03/2012] [Indexed: 10/28/2022]
|
20
|
Henri P, Beaumel S, Guezennec A, Poumès C, Stoebner PE, Stasia MJ, Guesnet J, Martinez J, Meunier L. MC1R expression in HaCaT keratinocytes inhibits UVA-induced ROS production via NADPH oxidase- and cAMP-dependent mechanisms. J Cell Physiol 2012; 227:2578-85. [PMID: 21898403 DOI: 10.1002/jcp.22996] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Abstract
Ultraviolet A (UVA) radiations are responsible for deleterious effects, mainly due to reactive oxygen species (ROS) production. Alpha-melanocyte stimulating hormone (α-MSH) binds to melanocortin-1 receptor (MC1R) in melanocytes to stimulate pigmentation and modulate cutaneous inflammatory responses. MC1R may be induced in keratinocytes after UV exposure. To investigate the effect of MC1R signaling on UVA-induced ROS (UVA-ROS) production, we generated HaCaT cells that stably express human MC1R (HaCaT-MC1R) or the Arg151Cys (R(151)C) non-functional variant (HaCaT-R(151)C). We then assessed ROS production immediately after UVA exposure and found that: (1) UVA-ROS production was strongly reduced in HaCaT-MC1R but not in HaCaT-R(151)C cells compared to parental HaCaT cells; (2) this inhibitory effect was further amplified by incubation of HaCaT-MC1R cells with α-MSH before UVA exposure; (3) protein kinase A (PKA)-dependent NoxA1 phosphorylation was increased in HaCaT-MC1R compared to HaCaT and HaCaT-R(151)C cells. Inhibition of PKA in HaCaT-MC1R cells resulted in a marked increase of ROS production after UVA irradiation; (4) the ability of HaCaT-MC1R cells to produce UVA-ROS was restored by inhibiting epidermal growth factor receptor (EGFR) or extracellular signal-regulated kinases (ERK) activity before UVA exposure. Our findings suggest that constitutive activity of MC1R in keratinocytes may reduce UVA-induced oxidative stress via EGFR and cAMP-dependent mechanisms.
Collapse
Affiliation(s)
- Pauline Henri
- Institute of Biomolecules Max Mousseron (IBMM), University Montpellier I and II, UMR CNRS 5247, Montpellier Cedex 5, France
| | | | | | | | | | | | | | | | | |
Collapse
|
21
|
Woerner BM, Luo J, Brown KR, Jackson E, Dahiya SM, Mischel P, Benovic JL, Piwnica-Worms D, Rubin JB. Suppression of G-protein-coupled receptor kinase 3 expression is a feature of classical GBM that is required for maximal growth. Mol Cancer Res 2011; 10:156-66. [PMID: 22086906 DOI: 10.1158/1541-7786.mcr-11-0411] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/29/2023]
Abstract
G-protein-coupled receptor kinases (GRK) regulate the function of G-protein-coupled receptors (GPCR). Previously, we found that GPCR (CXCR4)-mediated astrocytoma growth was dependent upon abnormally sustained CXCR4 signaling and was correlated with decreased GRK-mediated receptor phosphorylation. As CXCR4 has also been implicated in the stimulation of high-grade glioma growth, we sought to determine whether dysregulation of GRK expression and/or function might also be present in high-grade gliomas. In an analysis of data from The Cancer Genome Atlas, we found that GRK3 expression is frequently decreased in glioblastoma (GBM) of the classical subtype, which possesses signature amplification or mutational activation of the epidermal growth factor (EGF) receptor. We tested the correlation between GRK3 expression and GBM subtypes, as well as the relationship between the activation of the EGF and other growth factor receptor pathways and GRK expression. In analyses of primary GBM tissue and RNA specimens, we found that GRK3 expression is correlated with established criteria for GBM subtyping including expression of EGF receptor, platelet-derived growth factor receptor (PDGFR)α, NF1, PTEN, CDKN2A, and neurofilament. We also found that established drivers of gliomagenesis, the EGF, PDGF, and TGF-β pathways, all regulate GRK expression. Coculture experiments, designed to mimic critical interactions between tumor and brain microvascular endothelial cells, showed that specifically increasing GRK3 expression reduced the trophic effect of endothelial cells on tumor cells. Together, these experiments show that GRK3 is a negative regulator of cell growth whose expression is preferentially reduced in GBM of the classical subtype as a consequence of activity in primary gliomagenic pathways.
Collapse
Affiliation(s)
- B Mark Woerner
- Department of Pediatrics, Washington University School of Medicine, St Louis, MO 63110, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
22
|
Hartl D, He CH, Koller B, Da Silva CA, Homer R, Lee CG, Elias JA. Acidic mammalian chitinase is secreted via an ADAM17/epidermal growth factor receptor-dependent pathway and stimulates chemokine production by pulmonary epithelial cells. J Biol Chem 2008; 283:33472-82. [PMID: 18824549 DOI: 10.1074/jbc.m805574200] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
Acidic mammalian chitinase (AMCase) is expressed in an exaggerated fashion in epithelial cells at sites of pulmonary T helper cell type 2 inflammation and plays important roles in the pathogenesis of anti-parasite and asthma-like responses. However, the mechanisms that control epithelial cell AMCase secretion and its effector responses have not been adequately defined. To address these issues, we used in vivo and in vitro experimental systems to define the pathways of epithelial AMCase secretion and its epithelial regulatory effects. Here we demonstrate that, in murine T helper cell type 2 modeling systems, AMCase colocalizes with the epidermal growth factor receptor (EGFR) and ADAM17 (a membrane disintegrin and metallopeptidase 17) in lung epithelial cells. In vitro cotransfection experiments in A549 cells demonstrated that AMCase and EGFR physically interact with each other. Cotransfection of AMCase and EGFR also increased, whereas EGFR inhibition decreased AMCase secretion. Interestingly, AMCase secretion was not significantly altered by treatment with EGF but was significantly decreased when the upstream EGFR transactivator ADAM17 was inhibited. AMCase secretion was also decreased when the EGFR-downstream Ras was blocked. Transfected and recombinant AMCase induced epithelial cell production of CCL2, CCL17, and CXCL8. These studies demonstrate that lung epithelial cells secrete AMCase via an EGFR-dependent pathway that is activated by ADAM17 and mediates its effects via Ras. They also demonstrate that the AMCase that is secreted feeds back in an autocrine and/or paracrine fashion to stimulate pulmonary epithelial cell chemokine production.
Collapse
Affiliation(s)
- Dominik Hartl
- Section of Pulmonary and Critical Care Medicine, Department of Internal Medicine, Yale University School of Medicine, New Haven, Connecticut 06520-8057, USA
| | | | | | | | | | | | | |
Collapse
|
23
|
Song W, Xuan H, Lin Q. Epidermal growth factor induces changes of interaction between epidermal growth factor receptor and actin in intact cells. Acta Biochim Biophys Sin (Shanghai) 2008. [DOI: 10.1111/j.1745-7270.2008.00447.x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022] Open
|
24
|
Chou CT, He S, Jan CR. Paroxetine-induced apoptosis in human osteosarcoma cells: activation of p38 MAP kinase and caspase-3 pathways without involvement of [Ca2+]i elevation. Toxicol Appl Pharmacol 2006; 218:265-73. [PMID: 17174998 DOI: 10.1016/j.taap.2006.11.012] [Citation(s) in RCA: 45] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2006] [Revised: 10/15/2006] [Accepted: 11/09/2006] [Indexed: 12/13/2022]
Abstract
Selective serotonin reuptake inhibitors (SSRIs), a group of antidepressants, are generally used for treatment of various mood and anxiety disorders. There has been much research showing the anti-tumor and cytotoxic activities of some antidepressants; but the detailed mechanisms were unclear. In cultured human osteosarcoma cells (MG63), paroxetine reduced cell viability in a concentration- and time-dependent manner. Paroxetine caused apoptosis as assessed by propidium iodide-stained cells and increased caspase-3 activation. Although immunoblotting data revealed that paroxetine could activate the phosphorylation of extracellular signal-regulated kinase (ERK), c-Jun NH(2)-terminal kinase (JNK) and p38 mitogen-activated protein kinase (p38 MAPK), only SB203580 (a p38 MAPK inhibitor) partially prevented cells from apoptosis. Paroxetine also induced [Ca(2+)](i) increases which involved the mobilization of intracellular Ca(2+) stored in the endoplasmic reticulum and Ca(2+) influx from extracellular medium. However, pretreatment with BAPTA/AM, a Ca(2+) chelator, to prevent paroxetine-induced [Ca(2+)](i) increases did not protect cells from death. The results suggest that in MG63 cells, paroxetine caused Ca(2+)-independent apoptosis via inducing p38 MAPK-associated caspase-3 activation.
Collapse
Affiliation(s)
- Chiang-Ting Chou
- Department of Medical Education and Research, Kaohsiung Veterans General Hospital, Kaohsiung, 813, Taiwan
| | | | | |
Collapse
|
25
|
Sun YH, Liu MN, Li H, Shi S, Zhao YJ, Wang R, Xu CQ. Calcium-sensing receptor induces rat neonatal ventricular cardiomyocyte apoptosis. Biochem Biophys Res Commun 2006; 350:942-8. [PMID: 17046714 DOI: 10.1016/j.bbrc.2006.09.142] [Citation(s) in RCA: 47] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2006] [Accepted: 09/25/2006] [Indexed: 11/29/2022]
Abstract
The calcium-sensing receptor (CaSR) exists in many tissues, and its expression has been identified in rat cardiac tissue. However, the physiological importance and pathophysiological involvement of CaSR in homeostatic regulation of cardiac function are unclear. To investigate the relation of CaSR and apoptosis in cardiomyocytes, we examined the role of the CaSR activator gadolinium chloride (GdCl(3)) in rat neonatal ventricular cardiomyocytes. Expression of the CaSR protein was observed by Western blot. The apoptotic ratio of rat neonatal ventricular cardiomyocytes was measured with flow cytometry and immunofluorescence techniques. A laser scan confocal microscope was used to detect the intracellular concentration of calcium ([Ca(2+)](i)) in rat neonatal ventricular cardiomyocytes using the acetoxymethyl ester of fluo-3 (fluo-3/(AM)) as a fluorescent dye. The results showed that GdCl(3) increased the phosphorylation of extracellular signal-regulated protein kinase (ERK), c-Jun NH(2)-terminal protein kinases (JNK), and p38. GdCl(3) also activated caspase 9 and increased apoptosis in myocyte by increasing [Ca(2+)](i). In conclusion, these results suggest that CaSR promotes cardiomyocyte apoptosis in rat neonatal ventricular cardiomyocytes through activation of mitogen-activated protein kinases and caspase 9 signaling pathways.
Collapse
Affiliation(s)
- Yi-hua Sun
- Department of Clinical Laboratory, The Second Affiliated Hospital of Harbin Medical University, Harbin 150086, China
| | | | | | | | | | | | | |
Collapse
|
26
|
Abstract
Receptor tyrosine kinases (RTKs) are a unique family of cell surface receptors, each containing a common intracellular domain that has tyrosine kinase activity. However, RTKs share many signaling molecules with another unique family of cell surface receptors, the seven-transmembrane receptors (7TMRs), and these receptor families can activate similar signaling cascades. In this review of RTK signaling, we describe the role of cross talk between RTKs and 7TMRs, focusing specifically on the role played in this process by beta-arrestins and by G proteins.
Collapse
Affiliation(s)
- Christopher J Hupfeld
- Department of Medicine, Division of Endocrinology and Metabolism, University of California San Diego, La Jolla, California 92093, USA.
| | | |
Collapse
|
27
|
Driver GA, Veale RB. Modulation of integrin-linked kinase (ILK) expression in human oesophageal squamous cell carcinoma cell lines by the EGF and TGFbeta1 growth factors. Cancer Cell Int 2006; 6:12. [PMID: 16643659 PMCID: PMC1559647 DOI: 10.1186/1475-2867-6-12] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2005] [Accepted: 04/27/2006] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Integrin-linked kinase (ILK) is a ubiquitously expressed protein kinase that has emerged as one of the points of convergence between integrin- and growth factor-signalling pathways. RESULTS In this study we identify the ILK isoform expressed in five human oesophageal squamous cell carcinoma cell lines of South African origin as ILK1, and demonstrate its cellular distribution. ILK expression, although similar in the majority of the cell lines, did show variation. Furthermore, the ILK expressed was shown to be catalytically functional. The effect of growth factors on ILK expression was examined. An increase in ILK expression, following EGF and TGFbeta1 exposure, was a trend across all the five oesophageal carcinoma cell lines tested. CONCLUSION These results suggest that growth factor modulation of ILK expression relies on the internalisation/recycling of growth factor receptors and stimulation of the PI3K pathway, which may have implications with regards to cell adhesion and tumourigenesis.
Collapse
Affiliation(s)
- Glenn A Driver
- School of Molecular and Cell Biology, University of the Witwatersrand, Johannesburg 2050, South Africa
| | - Robin B Veale
- School of Molecular and Cell Biology, University of the Witwatersrand, Johannesburg 2050, South Africa
| |
Collapse
|