1
|
Pan H, Xu R, Zhang Y. Role of SPRY4 in health and disease. Front Oncol 2024; 14:1376873. [PMID: 38686189 PMCID: PMC11056578 DOI: 10.3389/fonc.2024.1376873] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2024] [Accepted: 04/01/2024] [Indexed: 05/02/2024] Open
Abstract
SPRY4 is a protein encoding gene that belongs to the Spry family. It inhibits the mitogen-activated protein kinase (MAPK) signaling pathway and plays a role in various biological functions under normal and pathological conditions. The SPRY4 protein has a specific structure and interacts with other molecules to regulate cellular behavior. It serves as a negative feedback inhibitor of the receptor protein tyrosine kinases (RTK) signaling pathway and interferes with cell proliferation and migration. SPRY4 also influences inflammation, oxidative stress, and cell apoptosis. In different types of tumors, SPRY4 can act as a tumor suppressor or an oncogene. Its dysregulation is associated with the development and progression of various cancers, including colorectal cancer, glioblastoma, hepatocellular carcinoma, perihilar cholangiocarcinoma, gastric cancer, breast cancer, and lung cancer. SPRY4 is also involved in organ development and is associated with ischemic diseases. Further research is ongoing to understand the expression and function of SPRY4 in specific tumor microenvironments and its potential as a therapeutic target.
Collapse
Affiliation(s)
- Hao Pan
- Department of Hepatobiliary Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Renjie Xu
- Department of Hepatobiliary Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Yong Zhang
- Department of Hepatobiliary Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| |
Collapse
|
2
|
Tu G, Guo Y, Xiao R, Tang L, Hu M, Liao B. Effects of Exercise Training on the Phosphoproteomics of the Medial Prefrontal Cortex in Rats With Autism Spectrum Disorder Induced by Valproic Acid. Neurorehabil Neural Repair 2023; 37:94-108. [PMID: 36860155 DOI: 10.1177/15459683231152814] [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: 03/03/2023]
Abstract
BACKGROUND The key neural pathological characteristics of autism spectrum disorder (ASD) include abnormal synaptic plasticity of the medial prefrontal cortex (mPFC). Exercise therapy is widely used to rehabilitate children with ASD, but its neurobiological mechanism is unclear. METHODS To clarify whether the structural and molecular plasticity of synapses in the mPFC are related to improvement in ASD behavioral deficits after continuous exercise rehabilitation training, we applied phosphoproteomic, behavioral, morphological, and molecular biological methods to investigate the impact of exercise on the phosphoprotein expression profile and synaptic structure of the mPFC in valproic acid (VPA)-induced ASD rats. RESULTS Exercise training differentially regulated the density, morphology, and ultrastructure of synapses in mPFC subregions in the VPA-induced ASD rats. In total, 1031 phosphopeptides were upregulated and 782 phosphopeptides were downregulated in the mPFC in the ASD group. After exercise training, 323 phosphopeptides were upregulated, and 1098 phosphopeptides were downregulated in the ASDE group. Interestingly, 101 upregulated and 33 downregulated phosphoproteins in the ASD group were reversed after exercise training, and these phosphoproteins were mostly involved in synapses. Consistent with the phosphoproteomics data, the total and phosphorylated levels of the proteins MARK1 and MYH10 were upregulated in the ASD group and reversed after exercise training. CONCLUSIONS The differential structural plasticity of synapses in mPFC subregions may be the basic neural architecture of ASD behavioral abnormalities. The phosphoproteins involved in mPFC synapses, such as MARK1 and MYH10, may play important roles in the exercise rehabilitation effect on ASD-induced behavioral deficits and synaptic structural plasticity, which requires further investigation.
Collapse
Affiliation(s)
- Genghong Tu
- Department of Sports Medicine, Guangzhou Sport University, Guangzhou, Guangdong, P.R. China.,Guangdong Provincial Key Laboratory of Physical Activity and Health Promotion, Scientific Research Center, Guangzhou Sport University, Guangzhou, Guangdong, P.R. China
| | - Youli Guo
- Department of Pharmacy, Guangdong Provincial Corps Hospital of Chinese People's Armed Police Forces, Guangzhou, Guangdong, P.R. China
| | - Ruoshi Xiao
- Department of Sports Medicine, Guangzhou Sport University, Guangzhou, Guangdong, P.R. China.,Guangdong Provincial Key Laboratory of Physical Activity and Health Promotion, Scientific Research Center, Guangzhou Sport University, Guangzhou, Guangdong, P.R. China
| | - Lianying Tang
- Department of Sports Medicine, Guangzhou Sport University, Guangzhou, Guangdong, P.R. China.,Guangdong Provincial Key Laboratory of Physical Activity and Health Promotion, Scientific Research Center, Guangzhou Sport University, Guangzhou, Guangdong, P.R. China
| | - Min Hu
- Guangdong Provincial Key Laboratory of Physical Activity and Health Promotion, Scientific Research Center, Guangzhou Sport University, Guangzhou, Guangdong, P.R. China
| | - Bagen Liao
- Department of Sports Medicine, Guangzhou Sport University, Guangzhou, Guangdong, P.R. China.,Guangdong Provincial Key Laboratory of Physical Activity and Health Promotion, Scientific Research Center, Guangzhou Sport University, Guangzhou, Guangdong, P.R. China
| |
Collapse
|
3
|
Interactions between 14-3-3 Proteins and Actin Cytoskeleton and Its Regulation by microRNAs and Long Non-Coding RNAs in Cancer. ENDOCRINES 2022. [DOI: 10.3390/endocrines3040057] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
14-3-3s are a family of structurally similar proteins that bind to phosphoserine or phosphothreonine residues, forming the central signaling hub that coordinates or integrates various cellular functions, thereby controlling many pathways important in cancer, cell motility, cell death, cytoskeletal remodeling, neuro-degenerative disorders and many more. Their targets are present in all cellular compartments, and when they bind to proteins they alter their subcellular localization, stability, and molecular interactions with other proteins. Changes in environmental conditions that result in altered homeostasis trigger the interaction between 14-3-3 and other proteins to retrieve or rescue homeostasis. In circumstances where these regulatory proteins are dysregulated, it leads to pathological conditions. Therefore, deeper understanding is needed on how 14-3-3 proteins bind, and how these proteins are regulated or modified. This will help to detect disease in early stages or design inhibitors to block certain pathways. Recently, more research has been devoted to identifying the role of MicroRNAs, and long non-coding RNAs, which play an important role in regulating gene expression. Although there are many reviews on the role of 14-3-3 proteins in cancer, they do not provide a holistic view of the changes in the cell, which is the focus of this review. The unique feature of the review is that it not only focuses on how the 14-3-3 subunits associate and dissociate with their binding and regulatory proteins, but also includes the role of micro-RNAs and long non-coding RNAs and how they regulate 14-3-3 isoforms. The highlight of the review is that it focuses on the role of 14-3-3, actin, actin binding proteins and Rho GTPases in cancer, and how this complex is important for cell migration and invasion. Finally, the reader is provided with super-resolution high-clarity images of each subunit of the 14-3-3 protein family, further depicting their distribution in HeLa cells to illustrate their interactions in a cancer cell.
Collapse
|
4
|
Guo K, Zhang X, Hou Y, Liu J, Feng Q, Wang K, Xu L, Zhang Y. A novel PCV2 ORF5-interacting host factor YWHAB inhibits virus replication and alleviates PCV2-induced cellular response. Vet Microbiol 2020; 251:108893. [PMID: 33096469 PMCID: PMC7568206 DOI: 10.1016/j.vetmic.2020.108893] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2020] [Accepted: 10/11/2020] [Indexed: 11/17/2022]
Abstract
YWHAB is a PCV2 ORF5-interacting host factor. YWHAB expression is activated by PCV2 infection and ORF5 transfection. YWHAB inhibits PCV2 replication. YWHAB alleviates PCV2 infection induced ERS, autophagy, ROS production and apoptosis.
Porcine circovirus type 2 (PCV2) infection causes porcine circovirus associated diseases (PCVAD) worldwide. Identification of host factors that interact with viral proteins is a fundamental step to understand the pathogenesis of PCV2. Our previous study reported that ORF5, a newly identified PCV2 viral protein supports PCV2 replication and interacts with multiple host factors. Here, we showed that a host factor YWHAB is an ORF5-interacting protein and plays essential roles during PCV2 infection. By using protein-protein interaction assays, we confirmed that YWHAB directly interacts with PCV2-ORF5 protein. We further showed that YWHAB expression was potently induced upon ORF5 overexpression and PCV2 infection. Remarkably, we found that the YWHAB strongly inhibited PCV2 replication, suggesting its role in defending PCV2 infection. By using the ectopic overexpression and gene knockdown approaches, we revealed that YWHAB inhibits PCV2-induced endoplasmic reticulum stress (ERS), autophagy, reactive oxygen species (ROS) production and apoptosis, suggesting its vital role in alleviating PCV2-induced cellular damage. Together, this study demonstrated that an ORF5-interacting host factor YWHAB affects PCV2 infection and PCV2-induced cellular response, which expands the current understanding of YWHAB biological function and might serves as a new therapeutic target to manage PCV2 infection-associated diseases.
Collapse
Affiliation(s)
- Kangkang Guo
- College of Veterinary Medicine, Northwest A&F University, Yangling, Shaanxi 712100, China
| | - Xiuping Zhang
- College of Veterinary Medicine, Northwest A&F University, Yangling, Shaanxi 712100, China; College of Animal Science, Tarim University, Alar, Xinjiang, 843300, China
| | - Yufeng Hou
- College of Veterinary Medicine, Northwest A&F University, Yangling, Shaanxi 712100, China
| | - Jing Liu
- College of Veterinary Medicine, Northwest A&F University, Yangling, Shaanxi 712100, China
| | - Quanwen Feng
- College of Veterinary Medicine, Northwest A&F University, Yangling, Shaanxi 712100, China
| | - Kai Wang
- College of Veterinary Medicine, Northwest A&F University, Yangling, Shaanxi 712100, China
| | - Lei Xu
- College of Life Sciences, Northwest A&F University, Yangling, Shaanxi 712100, China.
| | - Yanming Zhang
- College of Veterinary Medicine, Northwest A&F University, Yangling, Shaanxi 712100, China.
| |
Collapse
|
5
|
Natarajaseenivasan K, Shanmughapriya S, Velusamy P, Sayre M, Garcia A, Gomez NM, Langford D. Inflammation-induced PINCH expression leads to actin depolymerization and mitochondrial mislocalization in neurons. Transl Neurodegener 2020; 9:32. [PMID: 32746944 PMCID: PMC7397656 DOI: 10.1186/s40035-020-00211-4] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2020] [Accepted: 07/21/2020] [Indexed: 12/11/2022] Open
Abstract
BACKGROUND Diseases and disorders with a chronic neuroinflammatory component are often linked with changes in brain metabolism. Among neurodegenerative disorders, people living with human immunodeficiency virus (HIV) and Alzheimer's disease (AD) are particularly vulnerable to metabolic disturbances, but the mechanistic connections of inflammation, neurodegeneration and bioenergetic deficits in the central nervous system (CNS) are poorly defined. The particularly interesting new cysteine histidine-rich-protein (PINCH) is nearly undetectable in healthy mature neurons, but is robustly expressed in tauopathy-associated neurodegenerative diseases including HIV infection and AD. Although robust PINCH expression has been reported in neurons in the brains of patients with HIV and AD, the molecular mechanisms and cellular consequences of increased PINCH expression in CNS disease remain largely unknown. METHODS We investigated the regulatory mechanisms responsible for PINCH protein-mediated changes in bioenergetics, mitochondrial subcellular localization and bioenergetic deficits in neurons exposed to physiological levels of TNFα or the HIV protein Tat. Changes in the PINCH-ILK-Parvin (PIP) complex association with cofilin and TESK1 were assessed to identify factors responsible for actin depolymerization and mitochondrial mislocalization. Lentiviral and pharmacological inhibition experiments were conducted to confirm PINCH specificity and to reinstate proper protein-protein complex communication. RESULTS We identified MEF2A as the PINCH transcription factor in neuroinflammation and determined the biological consequences of increased PINCH in neurons. TNFα-mediated activation of MEF2A via increased cellular calcium induced PINCH, leading to disruption of the PIP ternary complex, cofilin activation by TESK1 inactivation, and actin depolymerization. The disruption of actin led to perinuclear mislocalization of mitochondria by destabilizing the kinesin-dependent mitochondrial transport machinery, resulting in impaired neuronal metabolism. Blocking TNFα-induced PINCH expression preserved mitochondrial localization and maintained metabolic functioning. CONCLUSIONS This study reported for the first time the mechanistic and biological consequences of PINCH expression in CNS neurons in diseases with a chronic neuroinflammation component. Our findings point to the maintenance of PINCH at normal physiological levels as a potential new therapeutic target for neurodegenerative diseases with impaired metabolisms.
Collapse
Affiliation(s)
- Kalimuthusamy Natarajaseenivasan
- Department of Neurosciences and Center for Neurovirology, Lewis Katz School of Medicine at Temple University, Philadelphia, PA 19140 USA
- Department of Microbiology, Bharathidasan University, Tiruchirappalli, 620024 India
| | - Santhanam Shanmughapriya
- Heart and Vascular Institute, Department of Medicine, Department of Cellular and Molecular Physiology, Penn State College of Medicine, Hershey, PA 17033 USA
| | - Prema Velusamy
- Heart and Vascular Institute, Department of Medicine, Department of Cellular and Molecular Physiology, Penn State College of Medicine, Hershey, PA 17033 USA
| | - Matthew Sayre
- Department of Neurosciences and Center for Neurovirology, Lewis Katz School of Medicine at Temple University, Philadelphia, PA 19140 USA
| | - Alvaro Garcia
- Department of Neurosciences and Center for Neurovirology, Lewis Katz School of Medicine at Temple University, Philadelphia, PA 19140 USA
| | - Nestor Mas Gomez
- Department of Neurosciences and Center for Neurovirology, Lewis Katz School of Medicine at Temple University, Philadelphia, PA 19140 USA
| | - Dianne Langford
- Department of Neurosciences and Center for Neurovirology, Lewis Katz School of Medicine at Temple University, Philadelphia, PA 19140 USA
| |
Collapse
|
6
|
The binding of lncRNA RP11-732M18.3 with 14-3-3 β/α accelerates p21 degradation and promotes glioma growth. EBioMedicine 2019; 45:58-69. [PMID: 31202814 PMCID: PMC6642068 DOI: 10.1016/j.ebiom.2019.06.002] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2018] [Revised: 05/30/2019] [Accepted: 06/03/2019] [Indexed: 02/07/2023] Open
Abstract
Background Long noncoding RNAs (lncRNAs) have been identified as regulators of a number of developmental and tumorigenic processes. However, the functions of most lncRNAs in glioma remain unknown and the mechanisms governing the proliferation of tumor cells remain poorly defined. Methods Both in vitro and in vivo assays were performed to investigate the roles of lncRNAs in the pathophysiology of gliomas. lncRNA arrays were used to identify differentially expressed lncRNAs. Subcutaneous tumor formation and a brain orthotopic tumor model in nude mice were used to investigate the functions of lncRNAs in vivo. The in vitro functions of lncRNAs were analyzed by fluorescence-activated cell sorting, colony formation, and western blot analyses. RNA fluorescence in situ hybridization and immunoprecipitation were used to explore the underlying mechanisms. Findings Here, we describe the newly discovered noncoding RNA RP11-732M18.3, which is highly overexpressed in glioma cells and interacts with 14-3-3β/α to promote glioma growth, acting as an oncogene. Overexpression of lncRNA RP11-732 M18.3 was associated with the proliferation of glioma cells and tumor growth in vitro and in vivo. Remarkably, lncRNA RP11-732M18.3 promoted cell proliferation and G1/S cell cycle transition. lncRNA RP11-732M18.3 is predominately localized in the cytoplasm. Mechanistically, the interaction of lncRNA RP11-732M18.3 with 14-3-3β/α increases the degradation of the p21 protein. lncRNA RP11-732M18.3 promoted the recruitment of ubiquitin-conjugating enzyme E2 E1 to 14-3-3β/α and the binding of 14-3-3β/α with ubiquitin-conjugating enzyme E2 E1 (UBE2E1) promoted the degradation of p21. Interpretation Overall these data demonstrated that lncRNA RP11-732M18.3 regulates glioma growth through a newly described lncRNA-protein interaction mechanism. The inhibition of lncRNA RP11-732M18.3 could provide a novel therapeutic target for glioma treatment.
Collapse
|
7
|
|
8
|
Nagai T, Mukoyama S, Kagiwada H, Goshima N, Mizuno K. Cullin-3-KCTD10-mediated CEP97 degradation promotes primary cilium formation. J Cell Sci 2018; 131:jcs.219527. [PMID: 30404837 DOI: 10.1242/jcs.219527] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2018] [Accepted: 10/19/2018] [Indexed: 12/20/2022] Open
Abstract
Primary cilia are antenna-like sensory organelles that transmit various extracellular signals. Ciliogenesis requires the removal of CP110 and its interactor CEP97 from the mother centriole for initiating ciliary axoneme extension, but the underlying mechanism remains unknown. Here we show that, upon serum starvation, CEP97 is partially degraded by the ubiquitin-proteasome system. CEP97 was polyubiquitylated in serum-starved cells, and overexpression of a non-ubiquitylatable CEP97 mutant effectively blocked CP110 removal and ciliogenesis induced by serum-starvation. Through several screening steps, we identified the cullin-3-RBX1-KCTD10 complex as the E3 ligase that mediates CEP97 degradation and removal from the mother centriole. Depletion of each component of this E3 complex caused aberrant accumulation of CEP97 on the centrosome, suppressed the removal of CEP97 and CP110 from the mother centriole, and impaired ciliogenesis. Moreover, KCTD10 was specifically localized to the mother centriole. These results suggest that CEP97 degradation by the cullin-3-RBX1-KCTD10 complex plays a crucial role in serum-starvation-induced CP110 removal and ciliogenesis.
Collapse
Affiliation(s)
- Tomoaki Nagai
- Department of Molecular and Chemical Life Sciences, Graduate School of Life Sciences, Tohoku University, Sendai, Miyagi 980-8578, Japan
| | - Sachiho Mukoyama
- Department of Molecular and Chemical Life Sciences, Graduate School of Life Sciences, Tohoku University, Sendai, Miyagi 980-8578, Japan
| | - Harumi Kagiwada
- Functional Proteomics Team, Molecular Profiling Research Center for Drug Discovery, National Institute of Advanced Industrial Science and Technology, Tokyo 135-0064, Japan
| | - Naoki Goshima
- Functional Proteomics Team, Molecular Profiling Research Center for Drug Discovery, National Institute of Advanced Industrial Science and Technology, Tokyo 135-0064, Japan
| | - Kensaku Mizuno
- Department of Molecular and Chemical Life Sciences, Graduate School of Life Sciences, Tohoku University, Sendai, Miyagi 980-8578, Japan
| |
Collapse
|
9
|
14-3-3β and γ differentially regulate peroxisome proliferator activated receptor γ2 transactivation and hepatic lipid metabolism. BIOCHIMICA ET BIOPHYSICA ACTA-GENE REGULATORY MECHANISMS 2015; 1849:1237-47. [PMID: 26260846 DOI: 10.1016/j.bbagrm.2015.08.002] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Received: 01/02/2015] [Revised: 08/04/2015] [Accepted: 08/06/2015] [Indexed: 02/06/2023]
Abstract
Peroxisome proliferator activated receptor (PPAR) γ2 plays important roles in glucose and lipid metabolism in hepatocytes. PPARγ2 is involved in metabolic disorders, including obesity, diabetes, and fatty liver disease. Although the 14-3-3 proteins participate in a variety of cell signal pathways, the roles of the 14-3-3 proteins in regulating PPARγ2 transactivation and hepatic lipid metabolism are unknown. We identified 14-3-3β and γ as PPARγ2 transcriptional regulators. We found that 14-3-3β and γ competitively interacted with the phosphorylated Ser273 of PPARγ2, which is important for regulating glucose and lipid metabolism. 14-3-3β increased the transcriptional activity of PPARγ2 and enhanced the expression levels of PPARγ2 target genes involved in lipogenesis and lipid transport. In contrast, 14-3-3γ decreased PPARγ2 transactivation and reduced the expression levels of PPARγ2 target genes. A high concentration of free fatty acids increased PPARγ2 expression and lipid accumulation. 14-3-3β enhanced hepatic lipogenesis, which is a major symptom of non-alcoholic fatty liver disease. However, 14-3-3γ suppressed hepatic lipid accumulation in the presence of high free fatty acids. These findings indicate that 14-3-3β and γ are novel PPARγ2 regulators and are involved in hepatic lipid metabolism. 14-3-3β and γ can be therapeutic target molecules to treat non-alcoholic fatty liver disease.
Collapse
|
10
|
14-3-3 λ protein interacts with ADF1 to regulate actin cytoskeleton dynamics in Arabidopsis. SCIENCE CHINA-LIFE SCIENCES 2015; 58:1142-50. [DOI: 10.1007/s11427-015-4897-1] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/03/2015] [Accepted: 03/28/2015] [Indexed: 01/15/2023]
|
11
|
Gong F, Wang G, Ye J, Li T, Bai H, Wang W. 14-3-3β regulates the proliferation of glioma cells through the GSK3β/β-catenin signaling pathway. Oncol Rep 2013; 30:2976-82. [PMID: 24065186 DOI: 10.3892/or.2013.2740] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2013] [Accepted: 08/20/2013] [Indexed: 11/05/2022] Open
Abstract
We previously demonstrated that 14-3-3β is overexpressed in astrocytomas; however, the underlying mechanisms are poorly understood. Based on the reported multiple functions of 14-3-3β, we hypothesized that it interacts with glycogen synthase kinase 3 β (GSK3β), which regulates β-catenin-mediated oncogene expression and contributes to tumorigenesis and astrocytoma progression. To test these hypotheses, we used 14-3-3β overexpression vectors and small interfering RNA (siRNA) transfection in the human normal astrocyte cell line SVGp12 and the glioma cell line U87, respectively. The results showed that overexpression of 14-3-3β promoted the proliferation of SVGp12 cells, while knockdown of 14-3-3β inhibited the proliferation of U87 cells as analyzed by the 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide (MTT) and bromodeoxyuridine (BrdU) assays. In Flag-tagged 14-3-3β-overexpressing cells, GSK3β was co-immunoprecipitated with 14-3-3β using a Flag antibody. Knockdown of β-catenin by siRNA blocked cell proliferation induced by overexpression of 14-3-3β. Furthermore, overexpression of 14-3-3β suppressed the phosphorylation of β-catenin leading to its accumulation and nuclear translocation as revealed by western blot analysis. In addition, β-catenin nuclear translocation induced by overexpression of 14-3-3β activated the transcription of oncogenes including c-myc and cyclin D1. Collectively, these results revealed that 14-3-3β regulates the proliferation of astrocytes and glioma cells through the GSK3β/β-catenin signaling pathway. The delineated mechanism of 14-3-3β may be responsible for the tumorigenesis and progression of human astrocytomas. Thus, new therapeutic strategies or drugs aimed at 14-3-3β may have potential for the treatment of human astrocytomas.
Collapse
Affiliation(s)
- Fanghe Gong
- Department of Neurosurgery, Guangzhou General Hospital of Guangzhou Military Command, Guangzhou 510010, P.R. China
| | | | | | | | | | | |
Collapse
|
12
|
Nagai T, Ikeda M, Chiba S, Kanno SI, Mizuno K. Furry promotes acetylation of microtubules in the mitotic spindle by inhibition of SIRT2 tubulin deacetylase. J Cell Sci 2013; 126:4369-80. [PMID: 23886946 DOI: 10.1242/jcs.127209] [Citation(s) in RCA: 49] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/25/2023] Open
Abstract
The structure and function of microtubules (MTs) are regulated by post-translational modifications of tubulin subunits, such as acetylation of the Lys40 residue of α-tubulin. Regulation of the organization and dynamics of MTs is essential for the precise formation of the mitotic spindle. Spindle MTs are highly acetylated, but the mechanism regulating this acetylation is largely unknown. Furry (Fry) is an evolutionarily conserved protein that binds to MTs and colocalizes with acetylated MTs in the mitotic spindle. In this study, we examined the role of Fry in the acetylation of MTs in the mitotic spindle. Depletion of Fry significantly reduced the level of MT acetylation in the mitotic spindle. Expression of the N-terminal fragment of Fry induced hyperacetylation of MTs in both mitotic and interphase cells. These results indicate that Fry promotes MT acetylation in the mitotic spindle. We also found that Fry binds to the tubulin deacetylase SIRT2, preferentially in mitotic cells. Cell-free experiments revealed that the N-terminal region of Fry is the domain responsible for binding to and inhibiting the tubulin-deacetylase activity of SIRT2. AGK2, a specific inhibitor of SIRT2, increased the level of MT acetylation in the mitotic spindle, indicating that SIRT2 is involved in the deacetylation of spindle MTs. Furthermore, AGK2 reversed the decrease in MT acetylation induced by Fry depletion. In summary, these results suggest that Fry plays a crucial role in promoting the level of MT acetylation in the mitotic spindle by inhibiting the tubulin-deacetylase activity of SIRT2.
Collapse
Affiliation(s)
- Tomoaki Nagai
- Department of Biomolecular Sciences, Graduate School of Life Sciences, Tohoku University, Sendai, Miyagi 980-8578, Japan
| | | | | | | | | |
Collapse
|
13
|
Wu Q, Liu CZ, Tao LY, Yu L, Liu W, Chen SS, He XD. The clinicopathological and prognostic impact of 14-3-3 protein isoforms expression in human cholangiocarcinoma by immunohistochemistry. Asian Pac J Cancer Prev 2013; 13:1253-9. [PMID: 22799314 DOI: 10.7314/apjcp.2012.13.4.1253] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022] Open
Abstract
The 14-3-3 proteins are highly conserved, ubiquitous molecules involved in a variety of biologic phenomena, such as cell cycle control, and apoptosis. However, their expression in cholangiocarcinoma has not been previously characterized. In this paper, immunohistochemistry using specific anti-14-3-3 monoclonal antibodies was performed on formalin-fixed;, paraffin embedded archival tissue from 86 patients of cholangiocarcinoma. We also examined the correlation between expression and survival rate and clinicopathologic factors such as tumor location, tumor size, pathologic differentiation, lymphatic permeation, lymph node metastasis, and tumor stage. Positive 14-3-3 proteins expression was observed for 6 isoforms (β, σ, γ, θ, β, η) of these proteins in 86 patients of cholangiocarcinoma. β and σ isoform immunoreactivity was correlated with lymph node metastasis, tumor stage and patients' survival rate. In addition, δ isoform immunoreactivity showed trends with tumor location, tumor size, pathologic differentiation and tumor stage, while the θ isoform was correlated with pathologic differentiation. These results indicated that upregulated expression of some isoforms of 14-3-3 may be a common mechanism for evading apoptosis in cholangiocarcinoma, so that targeting 14-3-3 may be a novel promising strategy for the treatment of this tumor.
Collapse
Affiliation(s)
- Qiao Wu
- Department of General Surgery, Peking Union Medical College Hospital, Beijing, China
| | | | | | | | | | | | | |
Collapse
|
14
|
Signaling mechanisms and functional roles of cofilin phosphorylation and dephosphorylation. Cell Signal 2012; 25:457-69. [PMID: 23153585 DOI: 10.1016/j.cellsig.2012.11.001] [Citation(s) in RCA: 303] [Impact Index Per Article: 23.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2012] [Accepted: 11/05/2012] [Indexed: 01/12/2023]
Abstract
Cofilin and actin-depolymerizing factor (ADF) are actin-binding proteins that play an essential role in regulating actin filament dynamics and reorganization by stimulating the severance and depolymerization of actin filaments. Cofilin/ADF are inactivated by phosphorylation at the serine residue at position 3 by LIM-kinases (LIMKs) and testicular protein kinases (TESKs) and are reactivated by dephosphorylation by the slingshot (SSH) family of protein phosphatases and chronophin. This review describes recent advances in our understanding of the signaling mechanisms regulating LIMKs and SSHs and the functional roles of cofilin phospho-regulation in cell migration, tumor invasion, mitosis, neuronal development, and synaptic plasticity. Accumulating evidence demonstrates that the phospho-regulation of cofilin/ADF is a key convergence point of cell signaling networks that link extracellular stimuli to actin cytoskeletal dynamics and that spatiotemporal control of cofilin/ADF activity by LIMKs and SSHs plays a crucial role in a diverse array of cellular and physiological processes. Perturbations in the normal control of cofilin/ADF activity underlie many pathological conditions, including cancer metastasis and neurological and cardiovascular disorders.
Collapse
|
15
|
Park SG, Jung S, Ryu HH, Jung TY, Moon KS, Kim IY, Jeong YI, Pei J, Park SJ, Kang SS. Role of 14-3-3-beta in the migration and invasion in human malignant glioma cell line U87MG. Neurol Res 2012; 34:893-900. [PMID: 22925547 DOI: 10.1179/1743132812y.0000000087] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/31/2022]
Abstract
PURPOSE To assess the influence of 14-3-3-beta in modulating the migration and invasion of human glioma cells. METHODS To profile the genes associated with malignant glioma cell motility, differential display-polymerase chain reaction was performed and the findings were validated by Northern blotting in the U343MG-A, U87MG, and U87MG-10' human glioma cell lines. Antisense 14-3-3-beta cDNA plasmid was transfected into U87MG ('U87-YA-3'). To follow motility changes after transfection, simple scratch test and matrigel assay were performed. Morphological and cytoskeletal changes were documented by light and confocal microscopy. In addition, doubling times of the transfectant and endogenous 14-3-3-beta levels were determined in various glioma cell lines with different motilities. RESULTS 14-3-3-beta was highly expressed in U87MG cells. U87-YA-3 cells became small and flat, and actin was depolarized. Furthermore, U87-YA-3 cell motility was inhibited markedly versus parental U87MG cells. The doubling times of transfected and parent cells were 32 and 37 hours, respectively. Endogenous 14-3-3-beta expression in the human glioma cell lines was proportional to their migratory and invasive abilities. CONCLUSION 14-3-3-beta modulates the migration and invasion in U87MG cells, which may be useful in developing therapeutic approaches for the treatment of glioma.
Collapse
Affiliation(s)
- Sung-Geun Park
- Department of Neurosurgery, Chonnam National University Hwasun Hospital and Medical School, Gwangju, Korea
| | | | | | | | | | | | | | | | | | | |
Collapse
|
16
|
Stanton PG, Sluka P, Foo CFH, Stephens AN, Smith AI, McLachlan RI, O'Donnell L. Proteomic changes in rat spermatogenesis in response to in vivo androgen manipulation; impact on meiotic cells. PLoS One 2012; 7:e41718. [PMID: 22860010 PMCID: PMC3408499 DOI: 10.1371/journal.pone.0041718] [Citation(s) in RCA: 53] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2012] [Accepted: 06/26/2012] [Indexed: 01/11/2023] Open
Abstract
The production of mature sperm is reliant on androgen action within the testis, and it is well established that androgens act on receptors within the somatic Sertoli cells to stimulate male germ cell development. Mice lacking Sertoli cell androgen receptors (AR) show late meiotic germ cell arrest, suggesting Sertoli cells transduce the androgenic stimulus co-ordinating this essential step in spermatogenesis. This study aimed to identify germ cell proteins responsive to changes in testicular androgen levels and thereby elucidate mechanisms by which androgens regulate meiosis. Testicular androgen levels were suppressed for 9 weeks using testosterone and estradiol-filled silastic implants, followed by a short period of either further androgen suppression (via an AR antagonist) or the restoration of intratesticular testosterone levels. Comparative proteomics were performed on protein extracts from enriched meiotic cell preparations from adult rats undergoing androgen deprivation and replacement in vivo. Loss of androgenic stimulus caused changes in proteins with known roles in meiosis (including Nasp and Hsp70–2), apoptosis (including Diablo), cell signalling (including 14-3-3 isoforms), oxidative stress, DNA repair, and RNA processing. Immunostaining for oxidised DNA adducts confirmed spermatocytes undergo oxidative stress-induced DNA damage during androgen suppression. An increase in PCNA and an associated ubiquitin-conjugating enzyme (Ubc13) suggested a role for PCNA-mediated regulation of DNA repair pathways in spermatocytes. Changes in cytoplasmic SUMO1 localisation in spermatocytes were paralleled by changes in the levels of free SUMO1 and of a subunit of its activating complex, suggesting sumoylation in spermatocytes is modified by androgen action on Sertoli cells. We conclude that Sertoli cells, in response to androgens, modulate protein translation and post-translational events in spermatocytes that impact on their metabolism, survival, and completion of meiosis.
Collapse
Affiliation(s)
- Peter G Stanton
- Prince Henry's Institute, Monash Medical Centre, Clayton, Victoria, Australia.
| | | | | | | | | | | | | |
Collapse
|
17
|
Sudnitsyna MV, Seit-Nebi AS, Gusev NB. Cofilin weakly interacts with 14-3-3 and therefore can only indirectly participate in regulation of cell motility by small heat shock protein HspB6 (Hsp20). Arch Biochem Biophys 2012; 521:62-70. [PMID: 22450169 DOI: 10.1016/j.abb.2012.03.010] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2012] [Revised: 03/08/2012] [Accepted: 03/13/2012] [Indexed: 01/08/2023]
Abstract
It has been previously reported that phosphorylated cofilin interacted with 14-3-3ζ protein to generate a sub-micromolar K(d) binary complex. Here we challenge this hypothesis by analyzing the direct association of recombinant cofilin with 14-3-3ζ using different in vitro biochemical methods. Phosphorylated cofilin at high concentration binds to 14-3-3 immobilized on nitrocellulose, however no complex formation was detected by means of native gel electrophoresis or chemical crosslinking. Intact dimeric or mutant monomeric 14-3-3 was unable to form stable complexes with phosphorylated or unphosphorylated cofilin detected by size-exclusion chromatography. In co-sedimentation assay 14-3-3 did not affect interaction of cofilin with F-actin. The data of native gel electrophoresis indicate that 14-3-3 did not affect interaction of cofilin with G-actin. Thus, cofilin only weakly interacts with 14-3-3 and therefore cannot directly compete with phosphorylated small heat shock protein HspB6 for its binding to 14-3-3. It is hypothesized that phosphorylated HspB6 might affect interaction of 14-3-3 with protein phosphatases (and/or protein kinases) involved in dephosphorylation (or phosphorylation) of cofilin and by this means regulate cofilin-dependent reorganization of cytoskeleton.
Collapse
Affiliation(s)
- Maria V Sudnitsyna
- Department of Biochemistry, School of Biology, Moscow State University, Moscow 119991, Russian Federation
| | | | | |
Collapse
|
18
|
Lou D, Sun B, Wei H, Deng X, Chen H, Xu D, Li G, Xu H, Wang Y. Spatiotemporal Expression of Testicular Protein Kinase 1 After Rat Sciatic Nerve Injury. J Mol Neurosci 2012; 47:180-91. [DOI: 10.1007/s12031-012-9712-x] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2011] [Accepted: 01/20/2012] [Indexed: 11/28/2022]
|
19
|
Sluchanko NN, Gusev NB. 14-3-3 proteins and regulation of cytoskeleton. BIOCHEMISTRY (MOSCOW) 2011; 75:1528-46. [PMID: 21417993 DOI: 10.1134/s0006297910130031] [Citation(s) in RCA: 54] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
The proteins of the 14-3-3 family are universal adapters participating in multiple processes running in the cell. We describe the structure, isoform composition, and distribution of 14-3-3 proteins in different tissues. Different elements of 14-3-3 structure important for dimer formation and recognition of protein targets are analyzed in detail. Special attention is paid to analysis of posttranslational modifications playing important roles in regulation of 14-3-3 function. The data of the literature concerning participation of 14-3-3 in regulation of intercellular contacts and different elements of cytoskeleton formed by microfilaments are analyzed. We also describe participation of 14-3-3 in regulation of small G-proteins and protein kinases important for proper functioning of cytoskeleton. The data on the interaction of 14-3-3 with different components of microtubules are presented, and the probable role of 14-3-3 in developing of certain neurodegenerative diseases is discussed. The data of the literature concerning the role of 14-3-3 in formation and normal functioning of intermediate filaments are also reviewed. It is concluded that due to its adapter properties 14-3-3 plays an important role in cytoskeleton regulation. The cytoskeletal proteins that are abundant in the cell might compete with the other protein targets of 14-3-3 and therefore can indirectly regulate many intracellular processes that are dependent on 14-3-3.
Collapse
Affiliation(s)
- N N Sluchanko
- Department of Biochemistry, Faculty of Biology, Lomonosov Moscow State University, Russia
| | | |
Collapse
|
20
|
O'Donnell L, Nicholls PK, O'Bryan MK, McLachlan RI, Stanton PG. Spermiation: The process of sperm release. SPERMATOGENESIS 2011; 1:14-35. [PMID: 21866274 DOI: 10.4161/spmg.1.1.14525] [Citation(s) in RCA: 261] [Impact Index Per Article: 18.6] [Reference Citation Analysis] [Abstract] [Subscribe] [Scholar Register] [Received: 10/27/2010] [Revised: 12/16/2010] [Accepted: 12/17/2010] [Indexed: 02/06/2023]
Abstract
Spermiation is the process by which mature spermatids are released from Sertoli cells into the seminiferous tubule lumen prior to their passage to the epididymis. It takes place over several days at the apical edge of the seminiferous epithelium, and involves several discrete steps including remodelling of the spermatid head and cytoplasm, removal of specialized adhesion structures and the final disengagement of the spermatid from the Sertoli cell. Spermiation is accomplished by the co-ordinated interactions of various structures, cellular processes and adhesion complexes which make up the "spermiation machinery". This review addresses the morphological, ultrastructural and functional aspects of mammalian spermiation. The molecular composition of the spermiation machinery, its dynamic changes and regulatory factors are examined. The causes of spermiation failure and their impact on sperm morphology and function are assessed in an effort to understand how this process may contribute to sperm count suppression during contraception and to phenotypes of male infertility.
Collapse
Affiliation(s)
- Liza O'Donnell
- Prince Henry's Institute of Medical Research; Clayton, VIC Australia
| | | | | | | | | |
Collapse
|
21
|
Yang X, Cao W, Lin H, Zhang W, Lin W, Cao L, Zhen H, Huo J, Zhang X. Isoform-specific expression of 14-3-3 proteins in human astrocytoma. J Neurol Sci 2008; 276:54-9. [PMID: 18851859 DOI: 10.1016/j.jns.2008.08.040] [Citation(s) in RCA: 52] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2008] [Revised: 08/20/2008] [Accepted: 08/26/2008] [Indexed: 12/25/2022]
Abstract
BACKGROUND 14-3-3 protein plays crucial roles in tumorigenesis, including the maintenance of cell cycle and DNA repair, the prevention of apoptosis, among others. In mammalian cells, seven 14-3-3 isoforms (beta, epsilon, zeta, eta, theta, gamma and sigma) have been identified and each of these seems to have distinct tissue localizations and isoform-specific functions. In the present study, the levels of all seven 14-3-3 isoforms were examined in astrocytoma. METHODS The expression of 14-3-3 isoforms and their protein expression levels were examined in five glioma cell lines by western blotting. Then in astrocytoma tissues, we investigated expression percentages of each isoform by immunohistochemistry. The protein and mRNA expression levels of each isoform were also detected by western blotting and RT-PCR, respectively. RESULTS 14-3-3beta and eta were specifically expressed in astrocytoma, and their expression frequencies and levels increased with the increase of astrocytoma malignancy. The result from glioma cell lines was consistent with that from astrocytoma tissue. CONCLUSIONS In our study, we found two tumor-specific isoforms of 14-3-3 in astrocytoma. They might be involved in astrocytoma tumorigenesis and may be useful as targets for therapy.
Collapse
Affiliation(s)
- Xiaoliang Yang
- Institute of Neurosurgery, Xijing Hospital, Fourth Military Medical University, Xi'an, Shaanxi Province, China
| | | | | | | | | | | | | | | | | |
Collapse
|
22
|
Van Troys M, Huyck L, Leyman S, Dhaese S, Vandekerkhove J, Ampe C. Ins and outs of ADF/cofilin activity and regulation. Eur J Cell Biol 2008; 87:649-67. [PMID: 18499298 DOI: 10.1016/j.ejcb.2008.04.001] [Citation(s) in RCA: 260] [Impact Index Per Article: 15.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2008] [Revised: 03/25/2008] [Accepted: 04/02/2008] [Indexed: 12/15/2022] Open
Abstract
The actin-binding proteins of the actin-depolymerisation factor (ADF)/cofilin family were first described more than three decades ago, but research on these proteins still occupies a front role in the actin and cell migration field. Moreover, cofilin activity is implicated in the malignant, invasive properties of cancer cells. The effects of ADF/cofilins on actin dynamics are diverse and their regulation is complex. In stimulated cells, multiple signalling pathways can be initiated resulting in different activation/deactivation switches that control ADF/cofilin activity. The output of this entire regulatory system, in combination with spatial and temporal segregation of the activation mechanisms, underlies the contribution of ADF/cofilins to various cell migration/invasion phenotypes. In this framework, we describe current views on how ADF/cofilins function in migrating and invading cells.
Collapse
|
23
|
Johne C, Matenia D, Li XY, Timm T, Balusamy K, Mandelkow EM. Spred1 and TESK1--two new interaction partners of the kinase MARKK/TAO1 that link the microtubule and actin cytoskeleton. Mol Biol Cell 2008; 19:1391-403. [PMID: 18216281 DOI: 10.1091/mbc.e07-07-0730] [Citation(s) in RCA: 50] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/17/2023] Open
Abstract
The signaling from MARKK/TAO1 to the MAP/microtubule affinity-regulating kinase MARK/Par1 to phosphorylated microtubule associated proteins (MAPs) renders microtubules dynamic and plays a role in neurite outgrowth or polarity development. Because hyperphosphorylation of Tau at MARK target sites is a hallmark of Alzheimer neurodegeneration, we searched for upstream regulators by the yeast two-hybrid approach and identified two new interaction partners of MARKK, the regulatory Sprouty-related protein with EVH-1 domain1 (Spred1) and the testis-specific protein kinase (TESK1). Spred1-MARKK binding has no effect on the activity of MARKK; therefore, it does not change microtubule (MT) stability. Spred1-TESK1 binding causes inhibition of TESK1. Because TESK1 can phosphorylate cofilin and thus stabilizes F-actin stress fibers, the inhibition of TESK1 by Spred1 makes F-actin fibers dynamic. A third element in this interaction triangle is that TESK1 binds to and inhibits MARKK. Thus, in Chinese hamster ovary (CHO) cells the elevation of MARKK results in MT disruption (via activation of MARK/Par1 and phosphorylation of MAPs), but this can be blocked by TESK1. Similarly, enhanced TESK1 activity results in increased stress fibers (via phospho-cofilin), but this can be blocked by elevating Spred1. Thus, the three-way interaction between Spred1, MARKK, and TESK1 represents a pathway that links regulation of both the microtubule- and F-actin cytoskeleton.
Collapse
Affiliation(s)
- Cindy Johne
- Max-Planck-Unit for Structural Molecular Biology, D-22607 Hamburg, Germany
| | | | | | | | | | | |
Collapse
|
24
|
Cao L, Cao W, Zhang W, Lin H, Yang X, Zhen H, Cheng J, Dong W, Huo J, Zhang X. Identification of 14-3-3 protein isoforms in human astrocytoma by immunohistochemistry. Neurosci Lett 2007; 432:94-9. [PMID: 18242847 DOI: 10.1016/j.neulet.2007.11.071] [Citation(s) in RCA: 29] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2007] [Revised: 11/03/2007] [Accepted: 11/09/2007] [Indexed: 12/22/2022]
Abstract
The 14-3-3 proteins are highly conserved, ubiquitous molecules involved in a variety of biologic events, such as cell cycle control, and apoptosis. In our previous study, it has been proved that they are expressed in primary human nervous system tumors. However, the isoform-specific expression of 14-3-3 protein is still need to be identified. This study is the first detection of 14-3-3 isoforms' specific expression in human astrocytoma. In the normal brain tissues, all the seven 14-3-3 isoforms' immunoreactivity was localized mainly in the neurons, while only weak expression of epsilon, zeta and theta was found in some glial cells. However, beta, epsilon, zeta, eta and theta isoforms' immunoreactivity was seen in the majority of astrocytoma samples and its immunoreactivity score was increased markedly with an increase in the pathologic grade of human astrocytomas. These results indicate that the five isoforms may play an important role in tumorigenesis of human astrocytoma.
Collapse
Affiliation(s)
- Lei Cao
- Department of Neurosurgery, Xijing Institute of Clinical Neuroscience, Fourth Military Medical University, No. 127 Changle Western Road, Xi'an 710032, People's Republic of China.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
25
|
Rivera J, Megias D, Bravo J. Proteomics-based strategy to delineate the molecular mechanisms of the metastasis suppressor gene BRMS1. J Proteome Res 2007; 6:4006-18. [PMID: 17854218 DOI: 10.1021/pr0703167] [Citation(s) in RCA: 36] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
The breast cancer metastasis suppressor 1 (BRMS1) gene has been shown to suppress metastasis without affecting the growth of the primary tumor in mouse models. It has also been shown to suppress the metastasis of tumors derived from breast, melanoma, and, more recently, ovarian carcinoma (see ref 1). However, how BRMS1 exerts its metastasis suppressor function remains unknown. To shed light into its metastatic mechanism of action, the sensitive 2D-DIGE analysis coupled with MS has been used to identify proteins differentially expressed by either overexpressing (Mel-BRMS1) or silencing BRMS1 (sh635) in a melanoma cell line. After comparison of the protein profiles from WT, Mel-BRMS1, and sh635 cells, 79 spots were found to be differentially expressed. Mass spectrometry analysis allowed the unambiguous identification of 55 polypeptides, corresponding to 43 different proteins. Interestingly, more than 75% of the identified proteins were down-regulated in Mel-BRMS1 cells compared to WT. In contrast, all the identified proteins in sh635 cells extracts were up-regulated compared to WT. Most of the deregulated proteins are involved in cell growth/maintenance and signal transduction among other cell processes. Six differentially expressed proteins (Hsp27, Alpha1 protease inhibitor, Cofilin1, Cathepsin D, Bone morphogenetic protein receptor2, and Annexin2) were confirmed by immunoblot and functional assays. Excellent correlation was found between DIGE analysis and immunoblot results, indicating the reliability of the analysis. Available evidence on the reported functions of the identified proteins supports the emerging role of BRMS1 as negative regulator of the metastasis development. This work opens an avenue for the molecular mechanisms' characterization of metastasis suppressor genes with the aim to understand their roles.
Collapse
Affiliation(s)
- José Rivera
- Signal Transduction Group, Structural Biology and Biocomputing Programme, Centro Nacional de Investigaciones Oncológicas, Melchor Fernández Almagro 3, E-28029 Madrid, Spain.
| | | | | |
Collapse
|
26
|
Alvarez M, Altafaj X, Aranda S, de la Luna S. DYRK1A autophosphorylation on serine residue 520 modulates its kinase activity via 14-3-3 binding. Mol Biol Cell 2007; 18:1167-78. [PMID: 17229891 PMCID: PMC1838983 DOI: 10.1091/mbc.e06-08-0668] [Citation(s) in RCA: 59] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2006] [Revised: 12/05/2006] [Accepted: 01/05/2007] [Indexed: 02/06/2023] Open
Abstract
Dual-specificity tyrosine-phosphorylated and regulated kinase (DYRK) proteins are an evolutionarily conserved family of protein kinases, with members identified from yeast to humans, that participate in a variety of cellular processes. DYRKs are serine/threonine protein kinases that are activated by autophosphorylation on a tyrosine residue in the activation loop. The family member DYRK1A has been shown to phosphorylate several cytosolic proteins and a number of splicing and transcription factors, including members of the nuclear factor of activated T cells family. In the present study, we show that DYRK1A autophosphorylates, via an intramolecular mechanism, on Ser-520, in the PEST domain of the protein. We also show that phosphorylation of this residue, which we show is subjected to dynamic changes in vivo, mediates the interaction of DYRK1A with 14-3-3beta. A second 14-3-3 binding site is present within the N-terminal of the protein. In the context of the DYRK1A molecule, neither site can act independently of the other. Bacterially produced DYRK1A and the mutant DYRK1A/S520A have similar kinase activities, suggesting that Ser-520 phosphorylation does not affect the intrinsic kinase activity on its own. Instead, we demonstrate that this phosphorylation allows the binding of 14-3-3beta, which in turn stimulates the catalytic activity of DYRK1A. These findings provide evidence for a novel mechanism for the regulation of DYRK1A kinase activity.
Collapse
Affiliation(s)
- Mónica Alvarez
- *Genes and Disease Program, Centre de Regulació Genómica, Parc de Recerca Biomèdica de Barcelona, 08003 Barcelona, Spain; and
| | - Xavier Altafaj
- *Genes and Disease Program, Centre de Regulació Genómica, Parc de Recerca Biomèdica de Barcelona, 08003 Barcelona, Spain; and
| | - Sergi Aranda
- *Genes and Disease Program, Centre de Regulació Genómica, Parc de Recerca Biomèdica de Barcelona, 08003 Barcelona, Spain; and
| | - Susana de la Luna
- *Genes and Disease Program, Centre de Regulació Genómica, Parc de Recerca Biomèdica de Barcelona, 08003 Barcelona, Spain; and
- Institució Catalana de Recerca i Estudis Avançats, 08010 Barcelona, Spain
| |
Collapse
|
27
|
Sesé M, Corominas M, Stocker H, Heino TI, Hafen E, Serras F. The Cdi/TESK1 kinase is required for Sevenless signaling and epithelial organization in the Drosophila eye. J Cell Sci 2006; 119:5047-56. [PMID: 17118962 DOI: 10.1242/jcs.03294] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
How cellular behaviors such as cell-to-cell communication, epithelial organization and cell shape reorganization are coordinated during development is poorly understood. The developing Drosophila eye offers an ideal model system to study these processes. Localized actin polymerization is required to constrict the apical surface of epithelial cells of the eye imaginal disc to maintain the refined arrangement of the developing ommatidia. The identity of each photoreceptor cell within the epithelium is determined by cell-to-cell contacts involving signal transduction events. The R7 photoreceptor cell requires the activity of the Sevenless RTK to adopt a proper cell fate. We performed an EP screen for negative regulators of this inductive process, and we identified the serine/threonine kinase Center divider (cdi) as a suppressor of the phenotype caused by an activated Sevenless receptor. Cdi is homologous to the human testis-specific kinase 1 (TESK1), a member of the LIM kinases involved in cytoskeleton control through ADF/cofilin phosphorylation. We have analyzed the effects of gain- and loss-of-function of cdi and found alterations in actin organization and in the adherens junctions proteins DE-cadherin and beta-catenin, as well as in Sevenless apical localization. Interference with the function of the ADF/cofilin phosphatase Slingshot (ssh), which antagonizes Cdi, also results in a suppression of signaling triggered by the Sevenless RTK. Our results reveal a critical interplay between the localization of molecules involved in epithelial organization and signal transduction.
Collapse
Affiliation(s)
- Marta Sesé
- Departament de Genètica, Facultat de Biologia, Universitat de Barcelona, Diagonal 645, 08028 Barcelona, Spain
| | | | | | | | | | | |
Collapse
|
28
|
Tsumura Y, Toshima J, Leeksma O, Ohashi K, Mizuno K. Sprouty-4 negatively regulates cell spreading by inhibiting the kinase activity of testicular protein kinase. Biochem J 2006; 387:627-37. [PMID: 15584898 PMCID: PMC1134992 DOI: 10.1042/bj20041181] [Citation(s) in RCA: 32] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
TESK1 (testicular protein kinase 1) is a serine/threonine kinase that phosphorylates cofilin and plays a critical role in integrin-mediated actin cytoskeletal reorganization and cell spreading. We previously showed that TESK1 interacts with Sprouty-4 (referred to as Spry4), an inhibitor of growth factor-induced Ras/MAP (mitogen-activated protein) kinase signalling, but the functional role of this interaction has remained unknown. In the present study, we show that Spry4 inhibits the kinase activity of TESK1 by binding to it through the C-terminal cysteine-rich region. Expression of Spry4 in cultured cells suppressed integrin-mediated cell spreading, and TESK1 reversed the inhibitory effect of Spry4 on cell spreading. Furthermore, Spry4 suppressed integrin- and TESK1-mediated cofilin phosphorylation during the spreading of cells on laminin. These findings suggest that Spry4 suppresses cell spreading by inhibiting the kinase activity of TESK1. Although tyrosine phosphorylation is required for the inhibitory activity of Spry4 on a Ras/MAP kinase pathway, mutation of the corresponding tyrosine residue (Tyr-75 in human Spry4) to an alanine had no apparent effect on its inhibitory actions on TESK1 activity and cell spreading, which suggests a novel cellular function of Spry to regulate the actin cytoskeleton, independent of its inhibitory activity on the Ras/MAP kinase signalling.
Collapse
Affiliation(s)
- Yoshikazu Tsumura
- *Department of Biomolecular Sciences, Graduate School of Life Sciences, Tohoku University, Sendai, Miyagi 980-8578, Japan
| | - Jiro Toshima
- *Department of Biomolecular Sciences, Graduate School of Life Sciences, Tohoku University, Sendai, Miyagi 980-8578, Japan
| | - Onno C. Leeksma
- †Department of Biochemistry, Academic Medical Center, University of Amsterdam, Amsterdam, The Netherlands
| | - Kazumasa Ohashi
- *Department of Biomolecular Sciences, Graduate School of Life Sciences, Tohoku University, Sendai, Miyagi 980-8578, Japan
| | - Kensaku Mizuno
- *Department of Biomolecular Sciences, Graduate School of Life Sciences, Tohoku University, Sendai, Miyagi 980-8578, Japan
- To whom correspondence should be addressed (email )
| |
Collapse
|
29
|
Abstract
The parvins are a family of proteins involved in linking integrins and associated proteins with intracellular pathways that regulate actin cytoskeletal dynamics and cell survival. Both alpha-parvin (PARVA) and beta-parvin (PARVB) localize to focal adhesions and function in cell adhesion, spreading, motility and survival through interactions with partners, such as integrin-linked kinase (ILK), paxillin, alpha-actinin and testicular kinase 1. A complex of PARVA with ILK and the LIM protein PINCH-1 is critical for cell survival in a variety of cells, including certain cancer cells, kidney podocytes and cardiac myocytes. While PARVA inhibits the activities of Rac1 and testicular kinase 1 and cell spreading, PARVB binds alphaPIX and alpha-actinin, and can promote cell spreading. In contrast to PARVA, PARVB inhibits ILK activity and reverses some of its oncogenic effects in cancer cells. This review focuses on the structure and function of the parvins and some possible roles in human diseases.
Collapse
Affiliation(s)
- J. L. Sepulveda
- Department of Pathology, 707B Scaife Hall, University of Pittsburgh, 3550 Terrace Street, Pittsburgh, 15261 USA
| | - C. Wu
- Department of Pathology, 707B Scaife Hall, University of Pittsburgh, 3550 Terrace Street, Pittsburgh, 15261 USA
| |
Collapse
|
30
|
Abstract
Many signal transduction events are orchestrated by specific interactions of proteins mediated through discrete phosphopeptide-binding motifs. Although several phosphospecific-binding domains are now known, 14-3-3s were the first proteins recognized to specifically bind a discrete phosphoserine or phosphothreonine motif. The 14-3-3 proteins are a family of ubiquitously expressed, exclusively eukaryotic proteins with an astonishingly large number of binding partners. Consequently, 14-3-3s modulate an enormous and diverse group of cellular processes. The effects of 14-3-3 proteins on their targets can be broadly defined using three categories: (i) conformational change; (ii) physical occlusion of sequence-specific or structural protein features; and (iii) scaffolding. This review will describe the current state of knowledge on 14-3-3 proteins, highlighting several important advances, and will attempt to provide a framework by which 14-3-3 functions can be understood.
Collapse
Affiliation(s)
- Dave Bridges
- Life Sciences Institute, University of Michigan, 210 Washtenaw Avenue, Ann Arbor, MI 48109, USA
| | | |
Collapse
|
31
|
LaLonde DP, Brown MC, Bouverat BP, Turner CE. Actopaxin interacts with TESK1 to regulate cell spreading on fibronectin. J Biol Chem 2005; 280:21680-8. [PMID: 15817463 DOI: 10.1074/jbc.m500752200] [Citation(s) in RCA: 39] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
The focal adhesion protein actopaxin contributes to integrin-actin associations and is involved in cell adhesion, spreading, and motility. Herein, we identify and characterize an association between actopaxin and the serine/threonine kinase testicular protein kinase 1 (TESK1), a ubiquitously expressed protein previously reported to regulate cellular spreading and focal adhesion formation via phosphorylation of cofilin. The interaction between actopaxin and TESK1 is direct and the binding sites were mapped to the carboxyl terminus of both proteins. The association between actopaxin and TESK1 is negatively regulated by adhesion to fibronectin, and a phosphomimetic actopaxin mutant that promotes cell spreading also exhibits impaired binding to TESK1. Binding of actopaxin to TESK1 inhibits TESK1 kinase activity in vitro. Expression of the carboxyl terminus of actopaxin has previously been reported to retard cell spreading. This effect was reversed following overexpression of TESK1 and was found to be dependent on an inability of actopaxin carboxyl terminus expressing cells to promote cofilin phosphorylation upon matrix adhesion and caused by retention of TESK1 by this actopaxin mutant. Thus, the association between actopaxin and TESK1, which is likely regulated by phosphorylation of actopaxin, regulates TESK1 activity and subsequent cellular spreading on fibronectin.
Collapse
Affiliation(s)
- David P LaLonde
- Department of Cell and Developmental Biology, State University of New York Upstate Medical University, Syracuse, New York 13210, USA
| | | | | | | |
Collapse
|
32
|
Qi W, Liu X, Qiao D, Martinez JD. Isoform-specific expression of 14-3-3 proteins in human lung cancer tissues. Int J Cancer 2005; 113:359-63. [PMID: 15455356 DOI: 10.1002/ijc.20492] [Citation(s) in RCA: 145] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
Abstract
14-3-3 Proteins play important roles in a wide range of vital regulatory processes, including signal transduction, apoptosis, cell cycle progression and DNA replication. In mammalian cells, 7 14-3-3 isoforms (beta, gamma, epsilon, eta, sigma, theta and zeta) have been identified and each of these seems to have distinct tissue localizations and isoform-specific functions. Previous studies have shown that 14-3-3 protein levels are higher in human lung cancers as compared to normal tissues. It is unclear, however, which of the 14-3-3 isoform(s) are overexpressed in these cancers. In our study, the levels of all seven 14-3-3 isoforms were examined by RT-PCR and Western blotting. We show that the message for only two isoforms, 14-3-3epsilon and zeta, could be detected in normal tissues. In lung cancer biopsies, however, four isoforms, 14-3-3beta, gamma, sigma, and theta;, in addition to 14-3-3epsilon and zeta, were present in abundance. The expression frequency of 14-3-3beta, gamma, sigma and theta; isoforms was 11, 10, 13 and 8 of the 14 biopsies examined, respectively. The data from immunohistochemical staining and Western blotting were consistent with the RT-PCR results. Given the prevalence of elevated 14-3-3 expression in human lung cancers we propose that these proteins may be involved in lung cancer tumorigenesis and that specific 14-3-3 proteins may be useful as markers for lung cancer diagnosis and targets for therapy.
Collapse
Affiliation(s)
- Wenqing Qi
- Arizona Cancer Center, Department of Cell Biology and Anatomy, The University of Arizona, Tucson, AZ 85724, USA
| | | | | | | |
Collapse
|
33
|
Nagata-Ohashi K, Ohta Y, Goto K, Chiba S, Mori R, Nishita M, Ohashi K, Kousaka K, Iwamatsu A, Niwa R, Uemura T, Mizuno K. A pathway of neuregulin-induced activation of cofilin-phosphatase Slingshot and cofilin in lamellipodia. ACTA ACUST UNITED AC 2004; 165:465-71. [PMID: 15159416 PMCID: PMC2172350 DOI: 10.1083/jcb.200401136] [Citation(s) in RCA: 160] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
Cofilin mediates lamellipodium extension and polarized cell migration by stimulating actin filament dynamics at the leading edge of migrating cells. Cofilin is inactivated by phosphorylation at Ser-3 and reactivated by cofilin-phosphatase Slingshot-1L (SSH1L). Little is known of signaling mechanisms of cofilin activation and how this activation is spatially regulated. Here, we show that cofilin-phosphatase activity of SSH1L increases ∼10-fold by association with actin filaments, which indicates that actin assembly at the leading edge per se triggers local activation of SSH1L and thereby stimulates cofilin-mediated actin turnover in lamellipodia. We also provide evidence that 14-3-3 proteins inhibit SSH1L activity, dependent on the phosphorylation of Ser-937 and Ser-978 of SSH1L. Stimulation of cells with neuregulin-1β induced Ser-978 dephosphorylation, translocation of SSH1L onto F-actin–rich lamellipodia, and cofilin dephosphorylation. These findings suggest that SSH1L is locally activated by translocation to and association with F-actin in lamellipodia in response to neuregulin-1β and 14-3-3 proteins negatively regulate SSH1L activity by sequestering it in the cytoplasm.
Collapse
Affiliation(s)
- Kyoko Nagata-Ohashi
- Dept. of Biomolecular Sciences, Graduate School of Life Sciences, Tohoku University, Aoba-ku, Sendai, Miyagi 980-8578, Japan
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
34
|
Bridges D, Moorhead GBG. 14-3-3 proteins: a number of functions for a numbered protein. Sci Signal 2004; 2004:re10. [PMID: 15266103 DOI: 10.1126/stke.2422004re10] [Citation(s) in RCA: 61] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/02/2022]
Abstract
Many signal transduction events are orchestrated by specific interactions of proteins mediated through discrete phosphopeptide-binding motifs. Although several phosphospecific-binding domains are now known, 14-3-3s were the first proteins recognized to specifically bind a discrete phosphoserine or phosphothreonine motif. The 14-3-3 proteins are a family of ubiquitously expressed, exclusively eukaryotic proteins with an astonishingly large number of binding partners. Consequently, 14-3-3s modulate an enormous and diverse group of cellular processes. The effects of 14-3-3 proteins on their targets can be broadly defined using three categories: (i) conformational change; (ii) physical occlusion of sequence-specific or structural protein features; and (iii) scaffolding. This review will describe the current state of knowledge on 14-3-3 proteins, highlighting several important advances, and will attempt to provide a framework by which 14-3-3 functions can be understood.
Collapse
Affiliation(s)
- Dave Bridges
- Department of Biological Sciences, University of Calgary, 2500 University Drive N.W., Calgary, Canada AB T2N 1N4
| | | |
Collapse
|
35
|
Sarmiere PD, Bamburg JR. Regulation of the neuronal actin cytoskeleton by ADF/cofilin. ACTA ACUST UNITED AC 2004; 58:103-17. [PMID: 14598374 DOI: 10.1002/neu.10267] [Citation(s) in RCA: 162] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/15/2023]
Abstract
Actin and microtubules are major cytoskeletal elements of most cells including neurons. In order for a cell to move and change shape, its cytoskeleton must undergo rearrangements that involve breaking down and reforming filaments. Many recent reviews have focused on the signaling pathways emanating from receptors that ultimately affect axon growth and growth cone steering. This particular review will address changes in the actin cytoskeleton modulated by the family of actin dynamizing proteins known as actin depolymerizing factor (ADF)/cofilin or AC proteins. Though much is known about inactivation of AC proteins through phosphorylation at ser3 by LIM or TES kinases, new mechanisms of regulation of AC have recently emerged. A novel phosphatase, slingshot (SSH), and the 14-3-3 family of regulatory proteins have also been found to affect AC activity. The potential role of AC proteins in modulating the actin organizational changes that accompany neurite initiation, axonogenesis, growth cone guidance, and dendritic spine formation will be discussed.
Collapse
Affiliation(s)
- Patrick D Sarmiere
- Department of Biochemistry and Molecular Biology, and Molecular, Cellular and Integrative Neuroscience Program, Colorado State University, Fort Collins, Colorado 80523, USA
| | | |
Collapse
|
36
|
Cavet ME, Lehoux S, Berk BC. 14-3-3beta is a p90 ribosomal S6 kinase (RSK) isoform 1-binding protein that negatively regulates RSK kinase activity. J Biol Chem 2003; 278:18376-83. [PMID: 12618428 DOI: 10.1074/jbc.m208475200] [Citation(s) in RCA: 39] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
p90 ribosomal S6 kinase 1 (RSK1) is a serine/threonine kinase that is activated by extracellular signal-related kinases 1/2 and phosphoinositide-dependent protein kinase 1 upon mitogen stimulation. Under basal conditions, RSK1 is located in the cytosol and upon stimulation, RSK1 translocates to the plasma membrane where it is fully activated. The ability of RSK1 to bind the adapter protein 14-3-3beta was investigated because RSK1 contains several putative 14-3-3-binding motifs. We demonstrate that RSK1 specifically and directly binds 14-3-3beta. This interaction was dependent on phosphorylation of serine 154 within the motif RLSKEV of RSK1. Binding of RSK1 to 14-3-3beta was maximal under basal conditions and decreased significantly upon mitogen stimulation. After 5 min of serum stimulation, a portion of 14-3-3beta and RSK1 translocated to the membrane fraction, and immunofluorescence studies demonstrated colocalization of RSK1 and 14-3-3beta at the plasma membrane in vivo. Incubation of recombinant RSK1 with 14-3-3beta decreased RSK1 kinase activity by approximately 50%. Mutation of RSK1 serine 154 increased both basal and serum-stimulated RSK activity. In addition, the epidermal growth factor response of RSK1S154A was enhanced compared with wild type RSK. The amount of RSK1S154A was significantly increased in the membrane fraction under basal conditions. Increased phosphorylation of two sites essential for RSK1 kinase activity (Ser(380) and Ser(363)) in RSK1S154A compared with RSK1 wild type, demonstrated that 14-3-3 interferes with RSK1 phosphorylation. These data suggest that 14-3-3beta binding negatively regulates RSK1 activity to maintain signal specificity and that association/dissociation of the 14-3-3beta-RSK1 complex is likely to be important for mitogen-mediated RSK1 activation.
Collapse
MESH Headings
- 14-3-3 Proteins
- 3T3 Cells
- Amino Acid Sequence
- Animals
- Binding Sites
- COS Cells
- Cell Line
- Cell Membrane/metabolism
- Cells, Cultured
- Cricetinae
- DNA/metabolism
- Dose-Response Relationship, Drug
- Genes, Reporter
- Glutathione Transferase/metabolism
- Humans
- Immunohistochemistry
- Mice
- Microscopy, Confocal
- Microscopy, Fluorescence
- Models, Biological
- Molecular Sequence Data
- Mutagenesis, Site-Directed
- Mutation
- Phosphorylation
- Point Mutation
- Precipitin Tests
- Promoter Regions, Genetic
- Protein Binding
- Protein Biosynthesis
- Protein Isoforms
- Protein Kinases/metabolism
- Protein Transport
- Rats
- Recombinant Proteins/chemistry
- Ribosomal Protein S6 Kinases
- Ribosomal Protein S6 Kinases, 90-kDa/chemistry
- Ribosomal Protein S6 Kinases, 90-kDa/metabolism
- Serine/chemistry
- Time Factors
- Transfection
- Tyrosine 3-Monooxygenase/metabolism
- Tyrosine 3-Monooxygenase/physiology
Collapse
Affiliation(s)
- Megan E Cavet
- Center for Cardiovascular Research and Department of Medicine, University of Rochester, Rochester, New York 14642, USA
| | | | | |
Collapse
|
37
|
Brener E, Rubinstein S, Cohen G, Shternall K, Rivlin J, Breitbart H. Remodeling of the actin cytoskeleton during mammalian sperm capacitation and acrosome reaction. Biol Reprod 2003; 68:837-45. [PMID: 12604633 DOI: 10.1095/biolreprod.102.009233] [Citation(s) in RCA: 193] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/01/2022] Open
Abstract
The sperm acrosome reaction and penetration of the egg follow zona pellucida binding only if the sperm has previously undergone the poorly understood maturation process known as capacitation. We demonstrate here that in vitro capacitation of bull, ram, mouse, and human sperm was accompanied by a time-dependent increase in actin polymerization. Induction of the acrosome reaction in capacitated cells initiated fast F-actin breakdown. Incubation of sperm in media lacking BSA or methyl-beta-cyclodextrin, Ca(2+), or NaHCO(3), components that are all required for capacitation, prevented actin polymerization as well as capacitation, as assessed by the ability of the cells to undergo the acrosome reaction. Inhibition of F-actin formation by cytochalasin D blocked sperm capacitation and reduced the in vitro fertilization rate of metaphase II-arrested mouse eggs. It has been suggested that protein tyrosine phosphorylation may represent an important regulatory pathway that is associated with sperm capacitation. We show here that factors known to stimulate sperm protein tyrosine phosphorylation (i.e., NaHCO(3), cAMP, epidermal growth factor, H(2)O(2), and sodium vanadate) were able to enhance actin polymerization, whereas inhibition of tyrosine kinases prevented F-actin formation. These data suggest that actin polymerization may represent an important regulatory pathway in with sperm capacitation, whereas F-actin breakdown occurs before the acrosome reaction.
Collapse
Affiliation(s)
- Ephraim Brener
- Faculty of Life Sciences, Bar-Ilan University, Ramat-Gan 52900, Israel
| | | | | | | | | | | |
Collapse
|
38
|
Birkenfeld J, Betz H, Roth D. Identification of cofilin and LIM-domain-containing protein kinase 1 as novel interaction partners of 14-3-3 zeta. Biochem J 2003; 369:45-54. [PMID: 12323073 PMCID: PMC1223062 DOI: 10.1042/bj20021152] [Citation(s) in RCA: 71] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2002] [Accepted: 09/26/2002] [Indexed: 11/17/2022]
Abstract
Proteins of the 14-3-3 family have been implicated in various physiological processes, and are thought to function as adaptors in various signal transduction pathways. In addition, 14-3-3 proteins may contribute to the reorganization of the actin cytoskeleton by interacting with as yet unidentified actin-binding proteins. Here we show that the 14-3-3 zeta isoform interacts with both the actin-depolymerizing factor cofilin and its regulatory kinase, LIM (Lin-11/Isl-1/Mec-3)-domain-containing protein kinase 1 (LIMK1). In both yeast two-hybrid assays and glutathione S-transferase pull-down experiments, these proteins bound efficiently to 14-3-3 zeta. Deletion analysis revealed consensus 14-3-3 binding sites on both cofilin and LIMK1. Furthermore, the C-terminal region of 14-3-3 zeta inhibited the binding of cofilin to actin in co-sedimentation experiments. Upon co-transfection into COS-7 cells, 14-3-3 zeta-specific immunoreactivity was redistributed into characteristic LIMK1-induced actin aggregations. Our data are consistent with 14-3-3-protein-induced changes to the actin cytoskeleton resulting from interactions with cofilin and/or LIMK1.
Collapse
Affiliation(s)
- Jörg Birkenfeld
- Department of Neurochemistry, Max-Planck-Institute for Brain Research, Deutschordenstrasse 46, 60528 Frankfurt, Germany
| | | | | |
Collapse
|
39
|
Abstract
The functionality of the actin cytoskeleton depends on a dynamic equilibrium between filamentous and monomeric actin. Proteins of the ADF/cofilin family are essential for the high rates of actin filament turnover observed in motile cells through regulation of actin polymerization/depolymerization cycles. Rho GTPases act through p21-activated kinase-1 (Pak-1) and Rho kinase to inhibit cofilin activity via the LIM kinase (LIMK)-mediated phosphorylation of cofilin on Ser3. We report the identification of 14-3-3zeta as a novel phosphocofilin binding protein involved in the maintenance of the cellular phosphocofilin pool. A Ser3 phosphocofilin binding protein was purified from bovine brain and was identified as 14-3-3zeta by mass spectrometry. The phosphorylation-dependent interaction between cofilin and 14-3-3zeta was confirmed in pulldown and coimmunoprecipitation experiments. Both Ser3 phosphorylation and a 14-3-3 recognition motif in cofilin are necessary for 14-3-3 binding. The expression of 14-3-3zeta increases phosphocofilin levels, and the coexpression of 14-3-3zeta with LIMK further elevates phosphocofilin levels and potentiates LIMK-dependent effects on the actin cytoskeleton. This potentiation of cofilin action appears to be a result of the protection of phosphocofilin from phosphatase-mediated dephosphorylation at Ser3 by bound 14-3-3zeta. Taken together, these results suggest that 14-3-3zeta proteins may play a dynamic role in the regulation of cellular actin structures through the maintenance of phosphocofilin levels.
Collapse
Affiliation(s)
- Antje Gohla
- Department of Immunology, The Scripps Research Institute, 10550 North Torrey Pines Road, La Jolla, CA 92037, USA
| | | |
Collapse
|
40
|
Niwa R, Nagata-Ohashi K, Takeichi M, Mizuno K, Uemura T. Control of actin reorganization by Slingshot, a family of phosphatases that dephosphorylate ADF/cofilin. Cell 2002; 108:233-46. [PMID: 11832213 DOI: 10.1016/s0092-8674(01)00638-9] [Citation(s) in RCA: 540] [Impact Index Per Article: 23.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/26/2022]
Abstract
The ADF (actin-depolymerizing factor)/cofilin family is a stimulus-responsive mediator of actin dynamics. In contrast to the mechanisms of inactivation of ADF/cofilin by kinases such as LIM-kinase 1 (LIMK1), much less is known about its reactivation through dephosphorylation. Here we report Slingshot (SSH), a family of phosphatases that have the property of F actin binding. In Drosophila, loss of ssh function dramatically increased levels of both F actin and phospho-cofilin (P cofilin) and disorganized epidermal cell morphogenesis. In mammalian cells, human SSH homologs (hSSHs) suppressed LIMK1-induced actin reorganization. Furthermore, SSH and the hSSHs dephosphorylated P cofilin in cultured cells and in cell-free assays. Our results strongly suggest that the SSH family plays a pivotal role in actin dynamics by reactivating ADF/cofilin in vivo.
Collapse
Affiliation(s)
- Ryusuke Niwa
- Department of Molecular Genetics, The Institute for Virus Research, Kyoto University, Kyoto 606-8507, Japan
| | | | | | | | | |
Collapse
|