1
|
Mariño Pérez L, Ielasi FS, Lee A, Delaforge E, Juyoux P, Tengo M, Davis RJ, Palencia A, Jensen MR. Structural basis of homodimerization of the JNK scaffold protein JIP2 and its heterodimerization with JIP1. Structure 2024; 32:1394-1403.e5. [PMID: 39013462 DOI: 10.1016/j.str.2024.06.010] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2024] [Revised: 03/18/2024] [Accepted: 06/19/2024] [Indexed: 07/18/2024]
Abstract
The scaffold proteins JIP1 and JIP2 intervene in the c-Jun N-terminal kinase (JNK) pathway to mediate signaling specificity by coordinating the simultaneous assembly of multiple kinases. Using NMR, we demonstrate that JIP1 and JIP2 heterodimerize via their SH3 domains with the affinity of heterodimerization being comparable to homodimerization. We present the high-resolution crystal structure of the JIP2-SH3 homodimer and the JIP1-JIP2-SH3 heterodimeric complex. The JIP2-SH3 structure reveals how charge differences in residues at its dimer interface lead to formation of compensatory hydrogen bonds and salt bridges, distinguishing it from JIP1-SH3. In the JIP1-JIP2-SH3 complex, structural features of each homodimer are employed to stabilize the heterodimer. Building on these insights, we identify key residues crucial for stabilizing the dimer of both JIP1 and JIP2. Through targeted mutations in cellulo, we demonstrate a functional role for the dimerization of the JIP1 and JIP2 scaffold proteins in activation of the JNK signaling pathway.
Collapse
Affiliation(s)
- Laura Mariño Pérez
- University Grenoble Alpes, CEA, CNRS, IBS, Grenoble, France; Departament de Química, Universitat de les Illes Balears, Institut Universitari d'Investigació en Ciències de la Salut (IUNICS), Institut de Recerca en Ciències de la Salut (IdISBa), Palma, Spain
| | - Francesco S Ielasi
- Institute for Advanced Biosciences (IAB), Structural Biology of Novel Targets in Human Diseases, INSERM U1209, CNRS UMR5309, Université Grenoble Alpes, Grenoble, France
| | - Alexandra Lee
- Program in Molecular Medicine, University of Massachusetts Medical School, Worcester, MA 01655, USA
| | | | - Pauline Juyoux
- University Grenoble Alpes, CEA, CNRS, IBS, Grenoble, France
| | - Maud Tengo
- University Grenoble Alpes, CEA, CNRS, IBS, Grenoble, France
| | - Roger J Davis
- Program in Molecular Medicine, University of Massachusetts Medical School, Worcester, MA 01655, USA
| | - Andrés Palencia
- Institute for Advanced Biosciences (IAB), Structural Biology of Novel Targets in Human Diseases, INSERM U1209, CNRS UMR5309, Université Grenoble Alpes, Grenoble, France.
| | | |
Collapse
|
2
|
Zareifard A, Beaudry F, Ndiaye K. Janus Kinase 3 phosphorylation and the JAK/STAT pathway are positively modulated by follicle-stimulating hormone (FSH) in bovine granulosa cells. BMC Mol Cell Biol 2023; 24:21. [PMID: 37337185 DOI: 10.1186/s12860-023-00482-5] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2022] [Accepted: 05/09/2023] [Indexed: 06/21/2023] Open
Abstract
Janus kinase 3 (JAK3) is a member of the JAK family of tyrosine kinase proteins involved in cytokine receptor-mediated intracellular signal transduction through the JAK/STAT signaling pathway. JAK3 was previously shown as differentially expressed in granulosa cells (GC) of bovine pre-ovulatory follicles suggesting that JAK3 could modulate GC function and activation/inhibition of downstream targets. We used JANEX-1, a JAK3 inhibitor, and FSH treatments and analyzed proliferation markers, steroidogenic enzymes and phosphorylation of target proteins including STAT3, CDKN1B/p27Kip1 and MAPK8IP3/JIP3. Cultured GC were treated with or without FSH in the presence or not of JANEX-1. Expression of steroidogenic enzyme CYP11A1, but not CYP19A1, was upregulated in GC treated with FSH and both were significantly decreased when JAK3 was inhibited. Proliferation markers CCND2 and PCNA were reduced in JANEX-1-treated GC and upregulated by FSH. Western blots analyses showed that JANEX-1 treatment reduced pSTAT3 amounts while JAK3 overexpression increased pSTAT3. Similarly, FSH treatment increased pSTAT3 even in JANEX-1-treated GC. UHPLC-MS/MS analyses revealed phosphorylation of specific amino acid residues within JAK3 as well as CDKN1B and MAPK8IP3 suggesting possible activation or inhibition post-FSH or JANEX-1 treatments. We show that FSH activates JAK3 in GC, which could phosphorylate target proteins and likely modulate other signaling pathways involving CDKN1B and MAPK8IP3, therefore controlling GC proliferation and steroidogenic activity.
Collapse
Affiliation(s)
- Amir Zareifard
- Centre de Recherche en Reproduction Et Fertilité, Département de Biomédecine Vétérinaire, Faculté de Médecine Vétérinaire, CRRF, Université de Montréal, Saint-Hyacinthe, Québec, Canada
- Département de Biomédecine Vétérinaire, Faculté de Médecine Vétérinaire, Université de Montréal, Saint-Hyacinthe, Québec, 3200, Canada
| | - Francis Beaudry
- Département de Biomédecine Vétérinaire, Faculté de Médecine Vétérinaire, Université de Montréal, Saint-Hyacinthe, Québec, 3200, Canada
- Centre de Recherche Sur Le Cerveau Et L'apprentissage (CIRCA), Université de Montréal, Montréal, Québec, Canada
| | - Kalidou Ndiaye
- Centre de Recherche en Reproduction Et Fertilité, Département de Biomédecine Vétérinaire, Faculté de Médecine Vétérinaire, CRRF, Université de Montréal, Saint-Hyacinthe, Québec, Canada.
- Département de Biomédecine Vétérinaire, Faculté de Médecine Vétérinaire, Université de Montréal, Saint-Hyacinthe, Québec, 3200, Canada.
| |
Collapse
|
3
|
Li Y, Liang ZY, Wang HL. N6-methyl-2'-deoxyadenosine promotes self-renewal of BFU-E progenitor in erythropoiesis. iScience 2023; 26:106924. [PMID: 37283807 PMCID: PMC10239700 DOI: 10.1016/j.isci.2023.106924] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2023] [Revised: 04/08/2023] [Accepted: 05/15/2023] [Indexed: 06/08/2023] Open
Abstract
Red blood cells supply the oxygen required for all human cells and are in demand for emerging blood-loss therapy. Here we identified N6-methyl-2'-deoxyadenosine (6mdA) as an agonist that promotes the hyperproliferation of burst-forming unit erythroid (BFU-E) progenitor cells. In addition, 6mdA represses the apoptosis of erythroid progenitor cells (EPCs). Combined use of with SCF and EPO enabled cultures of isolated BFU-E to be expanded up to 5,000-fold. Transcriptome analysis showed that 6mdA upregulates the expression of the EPC-associated factors c-Kit, Myb, and Gata2 and downregulates that of the erythroid maturation-related transcription factors Gata1, Spi1, and Klf1. Mechanistic studies suggested that 6mdA enhances and prolongs the activation of erythropoiesis-associated master gene c-Kit and its downstream signaling, leading to expansion and accumulation of EPCs. Collectively, we demonstrate that 6mdA can efficiently stimulate the EPC hyperproliferation and provide a new regenerative medicine recipe to improve ex vivo generation of red blood cells.
Collapse
Affiliation(s)
- Yao Li
- State Key Laboratory of Environmental Chemistry and Ecotoxicology, Research Center for Eco-Environmental Sciences, Chinese Academy of Sciences, Beijing 100085, China
- University of Chinese Academy of Sciences, Beijing 100049, China
| | - Zi-Yu Liang
- State Key Laboratory of Environmental Chemistry and Ecotoxicology, Research Center for Eco-Environmental Sciences, Chinese Academy of Sciences, Beijing 100085, China
- University of Chinese Academy of Sciences, Beijing 100049, China
| | - Hai-Lin Wang
- State Key Laboratory of Environmental Chemistry and Ecotoxicology, Research Center for Eco-Environmental Sciences, Chinese Academy of Sciences, Beijing 100085, China
- University of Chinese Academy of Sciences, Beijing 100049, China
- Institute of Environment and Health, Jianghan University, Wuhan, Hubei 430056, China
| |
Collapse
|
4
|
Chen J, Li H, Yang C, He Y, Arai T, Huang Q, Liu X, Miao L. Citrus Naringenin Increases Neuron Survival in Optic Nerve Crush Injury Model by Inhibiting JNK-JUN Pathway. Int J Mol Sci 2021; 23:385. [PMID: 35008811 PMCID: PMC8745540 DOI: 10.3390/ijms23010385] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2021] [Revised: 12/22/2021] [Accepted: 12/27/2021] [Indexed: 11/24/2022] Open
Abstract
Traumatic nerve injury activates cell stress pathways, resulting in neuronal death and loss of vital neural functions. To date, there are no available neuroprotectants for the treatment of traumatic neural injuries. Here, we studied three important flavanones of citrus components, in vitro and in vivo, to reveal their roles in inhibiting the JNK (c-Jun N-terminal kinase)-JUN pathway and their neuroprotective effects in the optic nerve crush injury model, a kind of traumatic nerve injury in the central nervous system. Results showed that both neural injury in vivo and cell stress in vitro activated the JNK-JUN pathway and increased JUN phosphorylation. We also demonstrated that naringenin treatment completely inhibited stress-induced JUN phosphorylation in cultured cells, whereas nobiletin and hesperidin only partially inhibited JUN phosphorylation. Neuroprotection studies in optic nerve crush injury mouse models revealed that naringenin treatment increased the survival of retinal ganglion cells after traumatic optic nerve injury, while the other two components had no neuroprotective effect. The neuroprotection effect of naringenin was due to the inhibition of JUN phosphorylation in crush-injured retinal ganglion cells. Therefore, the citrus component naringenin provides neuroprotection through the inhibition of the JNK-JUN pathway by inhibiting JUN phosphorylation, indicating the potential application of citrus chemical components in the clinical therapy of traumatic optic nerve injuries.
Collapse
Affiliation(s)
- Jie Chen
- Beijing Advanced Innovation Center for Intelligent Robots and Systems, Beijing Institute of Technology, Beijing 100081, China; (J.C.); (C.Y.); (T.A.); (Q.H.)
- School of Life Sciences, Peking University, Beijing 100871, China
| | - Hui Li
- Beijing Research Institute of Chinese Medicine, Beijing University of Chinese Medicine, Beijing 100029, China; (H.L.); (Y.H.)
| | - Changming Yang
- Beijing Advanced Innovation Center for Intelligent Robots and Systems, Beijing Institute of Technology, Beijing 100081, China; (J.C.); (C.Y.); (T.A.); (Q.H.)
- School of Mechatronical Engineering, Beijing Institute of Technology, Beijing 100081, China
| | - Yinjia He
- Beijing Research Institute of Chinese Medicine, Beijing University of Chinese Medicine, Beijing 100029, China; (H.L.); (Y.H.)
| | - Tatsuo Arai
- Beijing Advanced Innovation Center for Intelligent Robots and Systems, Beijing Institute of Technology, Beijing 100081, China; (J.C.); (C.Y.); (T.A.); (Q.H.)
- School of Mechatronical Engineering, Beijing Institute of Technology, Beijing 100081, China
| | - Qiang Huang
- Beijing Advanced Innovation Center for Intelligent Robots and Systems, Beijing Institute of Technology, Beijing 100081, China; (J.C.); (C.Y.); (T.A.); (Q.H.)
- School of Mechatronical Engineering, Beijing Institute of Technology, Beijing 100081, China
| | - Xiaodong Liu
- Beijing Research Institute of Chinese Medicine, Beijing University of Chinese Medicine, Beijing 100029, China; (H.L.); (Y.H.)
| | - Linqing Miao
- Beijing Advanced Innovation Center for Intelligent Robots and Systems, Beijing Institute of Technology, Beijing 100081, China; (J.C.); (C.Y.); (T.A.); (Q.H.)
- School of Mechatronical Engineering, Beijing Institute of Technology, Beijing 100081, China
| |
Collapse
|
5
|
Singh SK, Kumar S, Viswakarma N, Principe DR, Das S, Sondarva G, Nair RS, Srivastava P, Sinha SC, Grippo PJ, Thatcher GRJ, Rana B, Rana A. MAP4K4 promotes pancreatic tumorigenesis via phosphorylation and activation of mixed lineage kinase 3. Oncogene 2021; 40:6153-6165. [PMID: 34511598 PMCID: PMC8553609 DOI: 10.1038/s41388-021-02007-w] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2021] [Revised: 08/17/2021] [Accepted: 08/27/2021] [Indexed: 11/24/2022]
Abstract
MAP4K4 is a Ste20 member and reported to play important roles in various pathologies, including in cancer. However, the mechanism by which MAP4K4 promotes pancreatic cancer is not fully understood. It is suggested that MAP4K4 might function as a cancer promoter via specific downstream target(s) in an organ-specific manner. Here we identified MLK3 as a direct downstream target of MAP4K4. The MAP4K4 and MLK3 associates with each other, and MAP4K4 phosphorylates MLK3 on Thr738 and increases MLK3 kinase activity and downstream signaling. The phosphorylation of MLK3 by MAP4K4 promotes pancreatic cancer cell proliferation, migration, and colony formation. Moreover, MAP4K4 is overexpressed in human pancreatic tumors and directly correlates with the disease progression. The MAP4K4-specific pharmacological inhibitor, GNE-495, impedes pancreatic cancer cell growth, migration, induces cell death, and arrests cell cycle progression. Additionally, the GNE-495 reduced the tumor burden and extended survival of the KPC mice with pancreatic cancer. The MAP4K4 inhibitor also reduced MAP4K4 protein expression, tumor stroma, and induced cell death in murine pancreatic tumors. These findings collectively suggest that MLK3 phosphorylation by MAP4K4 promotes pancreatic cancer, and therefore therapies targeting MAP4K4 might alleviate the pancreatic cancer tumor burden in patients.
Collapse
Affiliation(s)
- Sunil Kumar Singh
- Department of Surgery, Division of Surgical Oncology, the University of Illinois at Chicago, Chicago, IL, 60612, USA
| | - Sandeep Kumar
- Department of Surgery, Division of Surgical Oncology, the University of Illinois at Chicago, Chicago, IL, 60612, USA
| | - Navin Viswakarma
- Department of Surgery, Division of Surgical Oncology, the University of Illinois at Chicago, Chicago, IL, 60612, USA
| | - Daniel R Principe
- Department of Surgery, Division of Surgical Oncology, the University of Illinois at Chicago, Chicago, IL, 60612, USA
| | - Subhasis Das
- Department of Surgery, Division of Surgical Oncology, the University of Illinois at Chicago, Chicago, IL, 60612, USA
| | - Gautam Sondarva
- Department of Surgery, Division of Surgical Oncology, the University of Illinois at Chicago, Chicago, IL, 60612, USA
| | - Rakesh Sathish Nair
- Department of Surgery, Division of Surgical Oncology, the University of Illinois at Chicago, Chicago, IL, 60612, USA
| | - Piush Srivastava
- Department of Surgery, Division of Surgical Oncology, the University of Illinois at Chicago, Chicago, IL, 60612, USA
| | | | - Paul J Grippo
- Department of Medicine, the University of Illinois at Chicago, Chicago, IL, 60612, USA
| | - Gregory R J Thatcher
- Department of Pharmacology and Toxicology, University of Arizona, Tucson, AZ, 85721, USA
| | - Basabi Rana
- Department of Surgery, Division of Surgical Oncology, the University of Illinois at Chicago, Chicago, IL, 60612, USA
- University of Illinois Hospital & Health Sciences System Cancer Center, the University of Illinois at Chicago, Chicago, IL, 60612, USA
- Jesse Brown VA Medical Center, Chicago, IL, 60612, USA
| | - Ajay Rana
- Department of Surgery, Division of Surgical Oncology, the University of Illinois at Chicago, Chicago, IL, 60612, USA.
- University of Illinois Hospital & Health Sciences System Cancer Center, the University of Illinois at Chicago, Chicago, IL, 60612, USA.
- Jesse Brown VA Medical Center, Chicago, IL, 60612, USA.
| |
Collapse
|
6
|
González-Coronel JM, Rodríguez-Alonso G, Guevara-García ÁA. A phylogenetic study of the members of the MAPK and MEK families across Viridiplantae. PLoS One 2021; 16:e0250584. [PMID: 33891654 PMCID: PMC8064577 DOI: 10.1371/journal.pone.0250584] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2021] [Accepted: 04/09/2021] [Indexed: 11/18/2022] Open
Abstract
Protein phosphorylation is regulated by the activity of enzymes generically known as kinases. One of those kinases is Mitogen-Activated Protein Kinases (MAPK), which operate through a phosphorylation cascade conformed by members from three related protein kinase families namely MAPK kinase kinase (MEKK), MAPK kinase (MEK), and MAPK; these three acts hierarchically. Establishing the evolution of these proteins in the plant kingdom is an interesting but complicated task because the current MAPK, MAPKK, and MAPKKK subfamilies arose from duplications and subsequent sub-functionalization during the early stage of the emergence of Viridiplantae. Here, an in silico genomic analysis was performed on 18 different plant species, which resulted in the identification of 96 genes not previously annotated as components of the MAPK (70) and MEK (26) families. Interestingly, a deeper analysis of the sequences encoded by such genes revealed the existence of putative domains not previously described as signatures of MAPK and MEK kinases. Additionally, our analysis also suggests the presence of conserved activation motifs besides the canonical TEY and TDY domains, which characterize the MAPK family.
Collapse
Affiliation(s)
- José Manuel González-Coronel
- Departamento de Biología Molecular de Plantas, Instituto de Biotecnología, Universidad Nacional Autónoma de México, Cuernavaca, Morelos, México
| | - Gustavo Rodríguez-Alonso
- Departamento de Biología Molecular de Plantas, Instituto de Biotecnología, Universidad Nacional Autónoma de México, Cuernavaca, Morelos, México
- Centro de Investigación en Dinámica Celular, Instituto de Investigación en Ciencias Básicas y Aplicadas, Universidad Autónoma del Estado de Morelos, Cuernavaca, Morelos, México
| | - Ángel Arturo Guevara-García
- Departamento de Biología Molecular de Plantas, Instituto de Biotecnología, Universidad Nacional Autónoma de México, Cuernavaca, Morelos, México
| |
Collapse
|
7
|
Li Y, Jiang W, Niu Q, Sun Y, Meng C, Tan L, Song C, Qiu X, Liao Y, Ding C. eIF2α-CHOP-BCl-2/JNK and IRE1α-XBP1/JNK signaling promote apoptosis and inflammation and support the proliferation of Newcastle disease virus. Cell Death Dis 2019; 10:891. [PMID: 31767828 PMCID: PMC6877643 DOI: 10.1038/s41419-019-2128-6] [Citation(s) in RCA: 93] [Impact Index Per Article: 18.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2019] [Revised: 10/14/2019] [Accepted: 11/04/2019] [Indexed: 12/23/2022]
Abstract
Newcastle disease virus (NDV) causes severe infectious disease in poultry and selectively kills tumor cells, by inducing apoptosis and cytokines secretion. In this report, we study the mechanisms underlying NDV-induced apoptosis by investigating the unfolded protein response (UPR). We found that NDV infection activated all three branches of the UPR signaling (PERK-eIF2α, ATF6, and IRE1α) and triggered apoptosis, in avian cells (DF-1 and CEF) and in various human cancer cell types (HeLa, Cal27, HN13, A549, H1299, Huh7, and HepG2). Interestingly, the suppression of either apoptosis or UPR led to impaired NDV proliferation. Meanwhile, the inhibition of UPR by 4-PBA protected cells from NDV-induced apoptosis. Further study revealed that activation of PERK-eIF2α induced the expression of transcription factor CHOP, which subsequently promoted apoptosis by downregulating BCL-2/MCL-1, promoting JNK signaling and suppressing AKT signaling. In parallel, IRE1α mediated the splicing of XBP1 mRNA and resulted in the translation and nuclear translocation of XBP1s, thereby promoting the transcription of ER chaperones and components of ER-associated degradation (ERAD). Furthermore, IRE1α promoted apoptosis and cytokines secretion via the activation of JNK signaling. Knock down and overexpression studies showed that CHOP, IRE1α, XBP1, and JNK supported efficient virus proliferation. Our study demonstrates that the induction of eIF2α-CHOP-BCL-2/JNK and IRE1α-XBP1/JNK signaling cascades promote apoptosis and cytokines secretion, and these signaling cascades support NDV proliferation.
Collapse
Affiliation(s)
- Yanrong Li
- Department of Avian Diseases, Shanghai Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Shanghai, 200241, P. R. China
| | - Weiyu Jiang
- Department of Avian Diseases, Shanghai Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Shanghai, 200241, P. R. China
| | - Qiaona Niu
- Department of Avian Diseases, Shanghai Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Shanghai, 200241, P. R. China
| | - Yingjie Sun
- Department of Avian Diseases, Shanghai Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Shanghai, 200241, P. R. China
| | - Chunchun Meng
- Department of Avian Diseases, Shanghai Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Shanghai, 200241, P. R. China
| | - Lei Tan
- Department of Avian Diseases, Shanghai Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Shanghai, 200241, P. R. China
| | - Cuiping Song
- Department of Avian Diseases, Shanghai Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Shanghai, 200241, P. R. China
| | - Xusheng Qiu
- Department of Avian Diseases, Shanghai Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Shanghai, 200241, P. R. China
| | - Ying Liao
- Department of Avian Diseases, Shanghai Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Shanghai, 200241, P. R. China.
| | - Chan Ding
- Department of Avian Diseases, Shanghai Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Shanghai, 200241, P. R. China. .,Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou, 225009, P. R. China.
| |
Collapse
|
8
|
Park JG, Aziz N, Cho JY. MKK7, the essential regulator of JNK signaling involved in cancer cell survival: a newly emerging anticancer therapeutic target. Ther Adv Med Oncol 2019; 11:1758835919875574. [PMID: 31579105 PMCID: PMC6759727 DOI: 10.1177/1758835919875574] [Citation(s) in RCA: 33] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2019] [Accepted: 08/19/2019] [Indexed: 01/02/2023] Open
Abstract
One of the mitogen-activated protein kinases (MAPKs), c-Jun NH2-terminal protein kinase (JNK) plays an important role in regulating cell fate, such as proliferation, differentiation, development, transformation, and apoptosis. Its activity is induced through the interaction of MAPK kinase kinases (MAP3Ks), MAPK kinases (MAP2Ks), and various scaffolding proteins. Because of the importance of the JNK cascade to intracellular bioactivity, many studies have been conducted to reveal its precise intracellular functions and mechanisms, but its regulatory mechanisms remain elusive. In this review, we discuss the molecular characterization, activation process, and physiological functions of mitogen-activated protein kinase kinase 7 (MKK7), the MAP2K that most specifically controls the activity of JNK. Understanding the role of MKK7/JNK signaling in physiological conditions could spark new hypotheses for targeted anticancer therapies.
Collapse
Affiliation(s)
- Jae Gwang Park
- Department of Integrative Biotechnology, Sungkyunkwan University, Suwon, Republic of Korea
| | - Nur Aziz
- Department of Integrative Biotechnology, Sungkyunkwan University, Suwon, Republic of Korea
| | - Jae Youl Cho
- Department of Integrative Biotechnology, Sungkyunkwan University, 2066 Seobu-ro, Suwon 16419, Republic of Korea
| |
Collapse
|
9
|
Hill A, Niles B, Cuyegkeng A, Powers T. Redesigning TOR Kinase to Explore the Structural Basis for TORC1 and TORC2 Assembly. Biomolecules 2018; 8:biom8020036. [PMID: 29865216 PMCID: PMC6023025 DOI: 10.3390/biom8020036] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2018] [Revised: 05/25/2018] [Accepted: 05/25/2018] [Indexed: 12/21/2022] Open
Abstract
TOR is a serine/threonine protein kinase that assembles into distinct TOR Complexes 1 and 2 (TORC1 or TORC2) to regulate cell growth. In mammalian cells, a single mTOR incorporates stably into mTORC1 and mTORC2. By contrast, in Saccharomyces cerevisiae, two highly similar Tor1 and Tor2 proteins exist, where Tor1 assembles exclusively into TORC1 and Tor2 assembles preferentially into TORC2. To gain insight into TOR complex assembly, we used this bifurcation in yeast to identify structural elements within Tor1 and Tor2 that govern their complex specificity. We have identified a concise region of ~500 amino acids within the N-terminus of Tor2, which we term the Major Assembly Specificity (MAS) domain, that is sufficient to confer significant TORC2 activity when placed into an otherwise Tor1 protein. Consistently, introduction of the corresponding MAS domain from Tor1 into an otherwise Tor2 is sufficient to confer stable association with TORC1-specific components. Remarkably, much like mTOR, this latter chimera also retains stable interactions with TORC2 components, indicating that determinants throughout Tor1/Tor2 contribute to complex specificity. Our findings are in excellent agreement with recent ultrastructural studies of TORC1 and TORC2, where the MAS domain is involved in quaternary interactions important for complex formation and/or stability.
Collapse
Affiliation(s)
- Andrew Hill
- Department of Molecular and Cellular Biology, College of Biological Sciences, University of California Davis, Davis, CA 95616, USA.
| | - Brad Niles
- Department of Molecular and Cellular Biology, College of Biological Sciences, University of California Davis, Davis, CA 95616, USA.
| | - Andrew Cuyegkeng
- Department of Molecular and Cellular Biology, College of Biological Sciences, University of California Davis, Davis, CA 95616, USA.
| | - Ted Powers
- Department of Molecular and Cellular Biology, College of Biological Sciences, University of California Davis, Davis, CA 95616, USA.
| |
Collapse
|
10
|
Jiang M, Chu Z. Comparative analysis of plant MKK gene family reveals novel expansion mechanism of the members and sheds new light on functional conservation. BMC Genomics 2018; 19:407. [PMID: 29843611 PMCID: PMC5975520 DOI: 10.1186/s12864-018-4793-8] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2017] [Accepted: 05/14/2018] [Indexed: 12/15/2022] Open
Abstract
Background Mitogen-activated protein kinase (MAPK) cascades play critical functions in almost every aspect of plant growth and development, which regulates many physiological and biochemical processes. As a middle nodal point of the MAPK cascades, although evolutionary analysis of MKK from individual plant families had some reports, their evolutionary history in entire plants is still not clear. Results To better understand the evolution and function of plant MKKs, we performed systematical molecular evolutionary analysis of the MAPKK gene family and also surveyed their gene organizations, sequence features and expression patterns in different subfamilies. Phylogenetic analysis showed that plant MAPKK fall into five different groups (Group A–E). Majority orthology groups seemed to be a single or low-copy genes in all plant species analyzed in Group B, C and D, whereas group A MKKs undergo several duplication events, generating multiple gene copies. Further analysis showed that these duplication events were on account of whole genome duplications (WGDs) in plants and the duplicate genes maybe have undergone functional divergence. We also found that group E MKKs had mutation with one change of serine or theronine might lead to inactivity originated through the ancient tandem duplicates in monocots. Moreover, we also identified MKK3 integrated NTF2 domain that might have gradually lost the cytoplasmic-nuclear trafficking activity, which suggests that they may involve with the gene function more and more sophistication in the evolutionary process. Moreover, expression analyses indicated that plant MKK genes play probable roles in UV-B signaling. Conclusion In general, ancient gene and genome duplications are significantly conducive to the expansion of the plant MKK gene family. Our study reveals two distinct evolutionary patterns for plant MKK proteins and sheds new light on the functional evolution of this gene family. Electronic supplementary material The online version of this article (10.1186/s12864-018-4793-8) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Min Jiang
- Shanghai Key Laboratory of Plant Functional Genomics and Resources, Shanghai Chenshan Botanical Garden, Shanghai, China.,Shanghai Chenshan Plant Science Research Center, Chinese Academy of Sciences, Shanghai, China
| | - Zhaoqing Chu
- Shanghai Key Laboratory of Plant Functional Genomics and Resources, Shanghai Chenshan Botanical Garden, Shanghai, China. .,Shanghai Chenshan Plant Science Research Center, Chinese Academy of Sciences, Shanghai, China.
| |
Collapse
|
11
|
Goyal RK, Tulpan D, Chomistek N, González-Peña Fundora D, West C, Ellis BE, Frick M, Laroche A, Foroud NA. Analysis of MAPK and MAPKK gene families in wheat and related Triticeae species. BMC Genomics 2018; 19:178. [PMID: 29506469 PMCID: PMC5838963 DOI: 10.1186/s12864-018-4545-9] [Citation(s) in RCA: 33] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2017] [Accepted: 02/13/2018] [Indexed: 12/16/2022] Open
Abstract
Background The mitogen-activated protein kinase (MAPK) family is involved in signal transduction networks that underpin many different biological processes in plants, ranging from development to biotic and abiotic stress responses. To date this class of enzymes has received little attention in Triticeae species, which include important cereal crops (wheat, barley, rye and triticale) that represent over 20% of the total protein food-source worldwide. Results The work presented here focuses on two subfamilies of Triticeae MAPKs, the MAP kinases (MPKs), and the MAPK kinases (MKKs) whose members phosphorylate the MPKs. In silico analysis of multiple Triticeae sequence databases led to the identification of 152 MAPKs belonging to these two sub-families. Some previously identified MAPKs were renamed to reflect the literature consensus on MAPK nomenclature. Two novel MPKs, MPK24 and MPK25, have been identified, including the first example of a plant MPK carrying the TGY activation loop sequence common to mammalian p38 MPKs. An EF-hand calcium-binding domain was found in members of the Triticeae MPK17 clade, a feature that appears to be specific to Triticeae species. New insights into the novel MEY activation loop identified in MPK11s are offered. When the exon-intron patterns for some MPKs and MKKs of wheat, barley and ancestors of wheat were assembled based on transcript data in GenBank, they showed deviations from the same sequence predicted in Ensembl. The functional relevance of MAPKs as derived from patterns of gene expression, MPK activation and MKK-MPK interaction is discussed. Conclusions A comprehensive resource of accurately annotated and curated Triticeae MPK and MKK sequences has been created for wheat, barley, rye, triticale, and two ancestral wheat species, goat grass and red wild einkorn. The work we present here offers a central information resource that will resolve existing confusion in the literature and sustain expansion of MAPK research in the crucial Triticeae grains. Electronic supplementary material The online version of this article (10.1186/s12864-018-4545-9) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Ravinder K Goyal
- Lethbridge Research and Development Centre, Agriculture and Agri-Food Canada, 5403 - 1st Avenue South, Lethbridge, Alberta, T1J 4B1, Canada
| | - Dan Tulpan
- Information and Communication Technologies, National Research Council of Canada, 100 des Aboiteaux Street, Moncton, New Brunswick, E1A 7R1, Canada
| | - Nora Chomistek
- Lethbridge Research and Development Centre, Agriculture and Agri-Food Canada, 5403 - 1st Avenue South, Lethbridge, Alberta, T1J 4B1, Canada
| | - Dianevys González-Peña Fundora
- Lethbridge Research and Development Centre, Agriculture and Agri-Food Canada, 5403 - 1st Avenue South, Lethbridge, Alberta, T1J 4B1, Canada
| | - Connor West
- Lethbridge Research and Development Centre, Agriculture and Agri-Food Canada, 5403 - 1st Avenue South, Lethbridge, Alberta, T1J 4B1, Canada
| | - Brian E Ellis
- Michael Smith Laboratories, University of British Columbia, #301 - 2185 East Mall, Vancouver, British Columbia, V6T 1Z4, Canada
| | - Michele Frick
- Lethbridge Research and Development Centre, Agriculture and Agri-Food Canada, 5403 - 1st Avenue South, Lethbridge, Alberta, T1J 4B1, Canada
| | - André Laroche
- Lethbridge Research and Development Centre, Agriculture and Agri-Food Canada, 5403 - 1st Avenue South, Lethbridge, Alberta, T1J 4B1, Canada
| | - Nora A Foroud
- Lethbridge Research and Development Centre, Agriculture and Agri-Food Canada, 5403 - 1st Avenue South, Lethbridge, Alberta, T1J 4B1, Canada.
| |
Collapse
|
12
|
Aberrant expression of JNK-associated leucine-zipper protein, JLP, promotes accelerated growth of ovarian cancer. Oncotarget 2018; 7:72845-72859. [PMID: 27655714 PMCID: PMC5341948 DOI: 10.18632/oncotarget.12069] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2016] [Accepted: 09/10/2016] [Indexed: 01/12/2023] Open
Abstract
Ovarian cancer is the most fatal gynecologic cancer with poor prognosis. Etiological factors underlying ovarian cancer genesis and progression are poorly understood. Previously, we have shown that JNK-associated Leucine zipper Protein (JLP), promotes oncogenic signaling. Investigating the role of JLP in ovarian cancer, our present study indicates that JLP is overexpressed in ovarian cancer tissue and ovarian cancer cells. Transient overexpression of JLP promotes proliferation and invasive migration of ovarian cancer cells. In addition, ectopic expression of JLP confers long-term survival and clonogenic potential to normal fallopian tube-derived epithelial cells. Coimmunoprecipitation and colocalization analyses demonstrate the in vivo interaction of JLP and JNK, which is stimulated by lysophosphatidic acid (LPA), an oncogenic lipid growth factor in ovarian cancer. We also show that LPA stimulates the translocation of JLP-JNK complex to the perinuclear region of SKOV3-ip cells. JLP-knockdown using shRNA abrogates LPA-stimulated activation of JNK as well as LPA-stimulated proliferation and invasive migration of SKOV3-ip cells. Studies using ovarian cancer xenograft mouse model indicate that the mice bearing JLP-silenced xenografts exhibits reduced tumor volume. Analysis of the xenograft tumor tissues indicate a reduction in the levels of JLP, JNK, phosphorylated-JNK, c-Jun and phosphorylated-c-Jun in JLP-silenced xenografts, thereby correlating the attenuated JLP-JNK signaling node with suppressed tumor growth. Thus, our results identify a critical role for JLP-signaling axis in ovarian cancer and provide evidence that targeting this signaling node could provide a new avenue for therapy.
Collapse
|
13
|
Lu Z, Liu Y, Shi Y, Shi X, Wang X, Xu C, Zhao H, Dong Q. Curcumin protects cortical neurons against oxygen and glucose deprivation/reoxygenation injury through flotillin-1 and extracellular signal-regulated kinase1/2 pathway. Biochem Biophys Res Commun 2018; 496:515-522. [DOI: 10.1016/j.bbrc.2018.01.089] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2017] [Accepted: 01/13/2018] [Indexed: 12/23/2022]
|
14
|
Genistein inhibits the growth and regulates the migration and invasion abilities of melanoma cells via the FAK/paxillin and MAPK pathways. Oncotarget 2017; 8:21674-21691. [PMID: 28423510 PMCID: PMC5400615 DOI: 10.18632/oncotarget.15535] [Citation(s) in RCA: 69] [Impact Index Per Article: 9.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2016] [Accepted: 01/27/2017] [Indexed: 01/10/2023] Open
Abstract
Genistein is one of the main components of soy-based foods, which are widely known for their many benefits, including anti-cancer, anti-inflammatory, and antioxidant effects. In this study, we investigated the anti-metastasis effects of genistein on B16F10 melanoma cells. Our results showed that genistein strongly inhibited B16F10 cell proliferation and induced apoptosis in time- and concentration-dependent manners. Genistein altered the morphology of B16F10 cells to an elongated shape with slim pseudopodia-like protrusions. Moreover, genistein inhibited the invasion and migration abilities of B16F10 cells in a dose-dependent manner. On one hand, a high concentration of genistein (100 μM) significantly inhibited cell adhesion and migration, as shown by wound healing assays and transwell-migration and invasion assays. Furthermore, the expression levels of p-FAK, p-paxillin, tensin-2, vinculin, and α-actinin were decreased by genistein. As a result, genistein is believed to strongly downregulate the migration and invasion abilities of B16F10 cells via the FAK/paxillin pathway. Moreover, p-p38, p-ERK, and p-JNK levels were also dramatically decreased by treatment with genistein. Finally, genistein significantly decreased the gene expression of FAK, paxillin, vimentin, and epithelial-to-mesenchymal transition-related transcription factor Snail, as shown by real-time PCR (qPCR) analysis. On the other hand, a lower concentration of genistein (12.5 μM) significantly promoted both invasion and migration by activating the FAK/paxillin and MAPK signaling cascades. Taken together, this study showed for the first time that genistein exerts dual functional effects on melanoma cells. Our findings suggest that genistein regulates the FAK/paxillin and MAPK signaling pathways in a highly concentration-dependent manner. Patients with melanoma should therefore be cautious of consuming soy-based foods in their diets.
Collapse
|
15
|
Linden R. The Biological Function of the Prion Protein: A Cell Surface Scaffold of Signaling Modules. Front Mol Neurosci 2017; 10:77. [PMID: 28373833 PMCID: PMC5357658 DOI: 10.3389/fnmol.2017.00077] [Citation(s) in RCA: 90] [Impact Index Per Article: 12.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/26/2016] [Accepted: 03/06/2017] [Indexed: 12/18/2022] Open
Abstract
The prion glycoprotein (PrPC) is mostly located at the cell surface, tethered to the plasma membrane through a glycosyl-phosphatydil inositol (GPI) anchor. Misfolding of PrPC is associated with the transmissible spongiform encephalopathies (TSEs), whereas its normal conformer serves as a receptor for oligomers of the β-amyloid peptide, which play a major role in the pathogenesis of Alzheimer’s Disease (AD). PrPC is highly expressed in both the nervous and immune systems, as well as in other organs, but its functions are controversial. Extensive experimental work disclosed multiple physiological roles of PrPC at the molecular, cellular and systemic levels, affecting the homeostasis of copper, neuroprotection, stem cell renewal and memory mechanisms, among others. Often each such process has been heralded as the bona fide function of PrPC, despite restricted attention paid to a selected phenotypic trait, associated with either modulation of gene expression or to the engagement of PrPC with a single ligand. In contrast, the GPI-anchored prion protein was shown to bind several extracellular and transmembrane ligands, which are required to endow that protein with the ability to play various roles in transmembrane signal transduction. In addition, differing sets of those ligands are available in cell type- and context-dependent scenarios. To account for such properties, we proposed that PrPC serves as a dynamic platform for the assembly of signaling modules at the cell surface, with widespread consequences for both physiology and behavior. The current review advances the hypothesis that the biological function of the prion protein is that of a cell surface scaffold protein, based on the striking similarities of its functional properties with those of scaffold proteins involved in the organization of intracellular signal transduction pathways. Those properties are: the ability to recruit spatially restricted sets of binding molecules involved in specific signaling; mediation of the crosstalk of signaling pathways; reciprocal allosteric regulation with binding partners; compartmentalized responses; dependence of signaling properties upon posttranslational modification; and stoichiometric requirements and/or oligomerization-dependent impact on signaling. The scaffold concept may contribute to novel approaches to the development of effective treatments to hitherto incurable neurodegenerative diseases, through informed modulation of prion protein-ligand interactions.
Collapse
Affiliation(s)
- Rafael Linden
- Laboratory of Neurogenesis, Institute of Biophysics, Federal University of Rio de Janeiro Rio de Janeiro, Brazil
| |
Collapse
|
16
|
Pleinis JM, Davis CW, Cantrell CB, Qiu DY, Zhan X. Purification, auto-activation and kinetic characterization of apoptosis signal-regulating kinase I. Protein Expr Purif 2017; 132:34-43. [PMID: 28082061 DOI: 10.1016/j.pep.2017.01.002] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2016] [Revised: 12/16/2016] [Accepted: 01/04/2017] [Indexed: 01/16/2023]
Abstract
Apoptosis signal-regulating kinase I (ASK1) is a mitogen-activated protein kinase kinase kinase (MAP3K) that activates the downstream MAP kinase kinases (MKKs) from two MAP kinase cascades: c-Jun N-terminal kinase (JNK) and p38. The essential physiological functions of ASK1 have attracted extensive attention. However, our understanding of the molecular mechanisms of ASK1, including the activation mechanism of ASK1 and the catalytic mechanism of ASK1-mediated MKK phosphorylation, remain unclear. The lack of purified ASK1 protein has hindered the elucidation of ASK1-initiated signal transduction mechanisms. Here, we report a one-step chromatography method for the expression and purification of functional full-length ASK1 from Escherichia coli. The purified ASK1 demonstrates auto-phosphorylation activity. The kinase activity of auto-phosphorylated ASK1 (pASK1) was also evaluated on two MKK substrates, MKK4 and 7, from the JNK cascades. Our results show that MKK7 can be phosphorylated by pASK1 more effectively than MKK4. The steady-state kinetic analysis demonstrates that MKK7 is a better ASK1 substrate than MKK4. These observations are further confirmed by direct pull-down assays which shows ASK1 binds MKK7 significantly stronger than MKK4. Furthermore, robust phospho-tyrosine signal is observed in MKK4 phosphorylation by pASK1 in addition to the phospho-serine and phospho-threonine. This study provides novel mechanistic and kinetic insights into the ASK1-initiated MAPK signal transduction via highly controlled reconstructed protein systems.
Collapse
Affiliation(s)
- John M Pleinis
- Department of Chemistry, Tennessee Technological University, Cookeville, TN 38505, USA
| | - Cameron W Davis
- Department of Chemistry, Tennessee Technological University, Cookeville, TN 38505, USA
| | - Caleb B Cantrell
- Department of Chemistry, Tennessee Technological University, Cookeville, TN 38505, USA
| | - David Y Qiu
- Department of Chemistry, Tennessee Technological University, Cookeville, TN 38505, USA
| | - Xuanzhi Zhan
- Department of Chemistry, Tennessee Technological University, Cookeville, TN 38505, USA.
| |
Collapse
|
17
|
Oh SE, Mouradian MM. Regulation of Signal Transduction by DJ-1. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2017; 1037:97-131. [PMID: 29147906 DOI: 10.1007/978-981-10-6583-5_8] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/04/2023]
Abstract
The ability of DJ-1 to modulate signal transduction has significant effects on how the cell regulates normal processes such as growth, senescence, apoptosis, and autophagy to adapt to changing environmental stimuli and stresses. Perturbations of DJ-1 levels or function can disrupt the equilibrium of homeostatic signaling networks and set off cascades that play a role in the pathogenesis of conditions such as cancer and Parkinson's disease.DJ-1 plays a major role in various pathways. It mediates cell survival and proliferation by activating the extracellular signal-regulated kinase (ERK1/2) pathway and the phosphatidylinositol-3-kinase (PI3K)/Akt pathway. It attenuates cell death signaling by inhibiting apoptosis signal-regulating kinase 1 (ASK1) activation as well as by inhibiting mitogen-activated protein kinase kinase kinase 1 (MEKK1/MAP3K1) activation of downstream apoptotic cascades. It also modulates autophagy through the ERK, Akt, or the JNK/Beclin1 pathways. In addition, DJ-1 regulates the transcription of genes essential for male reproductive function, such as spermatogenesis, by relaying nuclear receptor androgen receptor (AR) signaling. In this chapter, we summarize the ways that DJ-1 regulates these pathways, focusing on how its role in signal transduction contributes to cellular homeostasis and the pathologic states that result from dysregulation.
Collapse
Affiliation(s)
- Stephanie E Oh
- Center for Neurodegenerative and Neuroimmunologic Diseases, Department of Neurology, Rutgers - Robert Wood Johnson Medical School, Piscataway, NJ, 08854, USA
| | - M Maral Mouradian
- Center for Neurodegenerative and Neuroimmunologic Diseases, Department of Neurology, Rutgers - Robert Wood Johnson Medical School, Piscataway, NJ, 08854, USA.
| |
Collapse
|
18
|
Janssens J, Lu D, Ni B, Chadwick W, Siddiqui S, Azmi A, Etienne H, Jushaj A, van Gastel J, Martin B, Maudsley S. Development of Precision Small-Molecule Proneurotrophic Therapies for Neurodegenerative Diseases. VITAMINS AND HORMONES 2016; 104:263-311. [PMID: 28215298 DOI: 10.1016/bs.vh.2016.10.006] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/02/2023]
Abstract
Age-related neurodegenerative diseases, such as Alzheimer's disease, will represent one of the largest future burdens on worldwide healthcare systems due to the increasing proportion of elderly in our society. As deficiencies in neurotrophins are implicated in the pathogenesis of many age-related neurodegenerative disorders, it is reasonable to consider that global neurotrophin resistance may also become a major healthcare threat. Central nervous system networks are effectively maintained through aging by neuroprotective and neuroplasticity signaling mechanisms which are predominantly controlled by neurotrophin receptor signaling. Neurotrophin receptors are single pass receptor tyrosine kinases that form dimeric structures upon ligand binding to initiate cellular signaling events that control many protective and plasticity-related pathways. Declining functionality of the neurotrophin ligand-receptor system is considered one of the hallmarks of neuropathological aging. Therefore, it is imperative to develop effective therapeutic strategies to contend with this significant issue. While the therapeutic applications of cognate ligands for neurotrophin receptors are limited, the development of nonpeptidergic, small-molecule ligands can overcome these limitations, and productively regulate this important receptor system with beneficial effects. Using our advanced knowledge of the high-dimensionality complexity of receptor systems, the future generation of precision medicines targeting these systems will be an attainable goal.
Collapse
Affiliation(s)
- J Janssens
- Translational Neurobiology Group, University of Antwerp, Antwerpen, Belgium
| | - D Lu
- Receptor Pharmacology Unit, National Institute on Aging, National Institutes of Health, Baltimore MD United States
| | - B Ni
- Receptor Pharmacology Unit, National Institute on Aging, National Institutes of Health, Baltimore MD United States
| | - W Chadwick
- Receptor Pharmacology Unit, National Institute on Aging, National Institutes of Health, Baltimore MD United States
| | - S Siddiqui
- Receptor Pharmacology Unit, National Institute on Aging, National Institutes of Health, Baltimore MD United States
| | - A Azmi
- Translational Neurobiology Group, University of Antwerp, Antwerpen, Belgium
| | - H Etienne
- Translational Neurobiology Group, University of Antwerp, Antwerpen, Belgium
| | - A Jushaj
- Translational Neurobiology Group, University of Antwerp, Antwerpen, Belgium
| | - J van Gastel
- Translational Neurobiology Group, University of Antwerp, Antwerpen, Belgium
| | - B Martin
- Metabolism Unit, National Institute on Aging, National Institutes of Health, Baltimore MD United States
| | - S Maudsley
- Translational Neurobiology Group, University of Antwerp, Antwerpen, Belgium; Receptor Pharmacology Unit, National Institute on Aging, National Institutes of Health, Baltimore MD United States.
| |
Collapse
|
19
|
Jia H, Hao L, Guo X, Liu S, Yan Y, Guo X. A Raf-like MAPKKK gene, GhRaf19, negatively regulates tolerance to drought and salt and positively regulates resistance to cold stress by modulating reactive oxygen species in cotton. PLANT SCIENCE : AN INTERNATIONAL JOURNAL OF EXPERIMENTAL PLANT BIOLOGY 2016; 252:267-281. [PMID: 27717463 DOI: 10.1016/j.plantsci.2016.07.014] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/23/2016] [Revised: 06/24/2016] [Accepted: 07/23/2016] [Indexed: 05/06/2023]
Abstract
Mitogen-activated protein kinase kinase kinases (MAPKKKs) function at the top level of MAPK cascades and play important roles in plant development and stress responses. Although MAPKKKs comprise the largest family in the MAPK cascades, very few Raf-like MAPKKKs have been functionally identified, especially in the economically important crop cotton. In this study, a Raf-like MAPKKK gene, GhRaf19, was characterized for the first time in cotton. Our data show that the expression of GhRaf19 was inhibited by PEG and NaCl and induced by cold (4°C) and H2O2. Furthermore, when GhRaf19 was silenced in cotton using virus-induced gene silencing (VIGS), tolerance to drought and salt stress were enhanced, the accumulation of reactive oxygen species (ROS) was reduced, and ROS-related gene expression was increased. Consistent with these results, in N. benthamiana, overexpressing-GhRaf19 reduced tolerance to drought and salt. However, GhRaf19-silenced plants showed lowered resistance to cold in cotton, and this effect was correlated with the accumulation of ROS. In contrast, overexpressing GhRaf19 in N. benthamiana increased resistance to cold by inducing higher levels of expression and activity of ROS-related antioxidant genes/enzymes. These results indicate that GhRaf19 negatively regulates tolerance to drought and salt and positively regulates resistance to cold stress by modulating cellular ROS in cotton.
Collapse
Affiliation(s)
- Haihong Jia
- State Key Laboratory of Crop Biology, College of Life Sciences, Shandong Agricultural University, Tai'an, Shandong, PR China
| | - Lili Hao
- State Key Laboratory of Crop Biology, College of Life Sciences, Shandong Agricultural University, Tai'an, Shandong, PR China
| | - Xulei Guo
- State Key Laboratory of Crop Biology, College of Life Sciences, Shandong Agricultural University, Tai'an, Shandong, PR China
| | - Shuchang Liu
- State Key Laboratory of Crop Biology, College of Life Sciences, Shandong Agricultural University, Tai'an, Shandong, PR China
| | - Yan Yan
- State Key Laboratory of Crop Biology, College of Life Sciences, Shandong Agricultural University, Tai'an, Shandong, PR China
| | - Xingqi Guo
- State Key Laboratory of Crop Biology, College of Life Sciences, Shandong Agricultural University, Tai'an, Shandong, PR China.
| |
Collapse
|
20
|
Cunningham CA, Cardwell LN, Guan Y, Teixeiro E, Daniels MA. POSH Regulates CD4+ T Cell Differentiation and Survival. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2016; 196:4003-13. [PMID: 27084103 PMCID: PMC4868786 DOI: 10.4049/jimmunol.1501728] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/31/2015] [Accepted: 03/14/2016] [Indexed: 12/24/2022]
Abstract
The scaffold molecule POSH is crucial for the regulation of proliferation and effector function in CD8(+) T cells. However, its role in CD4(+) T cells is not known. In this study, we found that disruption of the POSH scaffold complex established a transcriptional profile that strongly skewed differentiation toward Th2, led to decreased survival, and had no effect on cell cycle entry. This is in stark contrast to CD8(+) T cells in which POSH regulates cell cycle and does not affect survival. Disruption of POSH in CD4(+) T cells resulted in the loss of Tak1-dependent activation of JNK1/2 and Tak1-mediated survival. However, in CD8(+) T cells, POSH regulates only JNK1. Remarkably, each type of T cell had a unique composition of the POSH scaffold complex and distinct posttranslational modifications of POSH. These data indicate that the mechanism that regulates POSH function in CD4(+) T cells is different from CD8(+) T cells. All together, these data strongly suggest that POSH is essential for the integration of cell-type-specific signals that regulate the differentiation, survival, and function of T cells.
Collapse
Affiliation(s)
- Cody A Cunningham
- Department of Molecular Microbiology and Immunology, School of Medicine, University of Missouri, Columbia, MO 65212
| | - Leah N Cardwell
- Department of Molecular Microbiology and Immunology, School of Medicine, University of Missouri, Columbia, MO 65212
| | - Yue Guan
- Department of Molecular Microbiology and Immunology, School of Medicine, University of Missouri, Columbia, MO 65212
| | - Emma Teixeiro
- Department of Molecular Microbiology and Immunology, School of Medicine, University of Missouri, Columbia, MO 65212
| | - Mark A Daniels
- Department of Molecular Microbiology and Immunology, School of Medicine, University of Missouri, Columbia, MO 65212
| |
Collapse
|
21
|
Tesser-Gamba F, Lopes LJDS, Petrilli AS, Toledo SRC. MAPK7 gene controls proliferation, migration and cell invasion in osteosarcoma. Mol Carcinog 2015; 55:1700-1713. [PMID: 26460937 DOI: 10.1002/mc.22420] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2015] [Revised: 09/02/2015] [Accepted: 09/18/2015] [Indexed: 11/11/2022]
Abstract
Osteosarcomas (OS) are the most common malignant bone tumors, and the identification of useful tumor biomarkers and target proteins is required to predict the clinical outcome of patients and therapeutic response as well as to develop novel therapeutic strategies. In our previous study, MAPK7 has been identified as a candidate oncogene, and a promising prognostic marker for OS. Sequential activation of protein kinases within the mitogen-activated protein kinase (MAPK) cascades is a common mechanism of signal transduction in many cellular processes. In this study, we investigated the behavior of MAPK7 gene in OS cell lines. Technical viability, proliferation, migration, invasion, and apoptosis were used to evaluate the function of the MAPK7 gene. We evaluated the behavior of the OS cells with MAPK7 gene silenced, not silenced, and exposed to the main chemotherapy drugs used in OS treatment. We found that silenced MAPK7 gene is effective at suppressing cell proliferation, inhibiting cell migration, and invasion. Furthermore, MAPK7 is an important activator of transcription factors and is the main expression modulator of other key genes in the MAPK pathway. In summary, our study suggests that MAPK7 might be a promising therapeutic target for OS. © 2015 Wiley Periodicals, Inc.
Collapse
Affiliation(s)
- Francine Tesser-Gamba
- Department of Pediatrics, Genetics Laboratory, Pediatric Oncology Institute (IOP/GRAACC), Federal University of São Paulo, São Paulo-SP, Brazil.,Department of Morphology and Genetics, Genetics Laboratory, Pediatric Oncology Institute (IOP/GRAACC), Federal University of São Paulo, São Paulo-SP, Brazil
| | - Luana Joyce da Silva Lopes
- Department of Clinical and Experimental Oncology, Genetics Laboratory, Pediatric Oncology Institute (IOP/GRAACC), Federal University of São Paulo, São Paulo-SP, Brazil
| | - Antonio Sergio Petrilli
- Department of Pediatrics, Pediatric Oncology Institute (IOP/GRAACC), Federal University of São Paulo, São Paulo-SP, Brazil
| | - Silvia Regina Caminada Toledo
- Department of Pediatrics, Genetics Laboratory, Pediatric Oncology Institute (IOP/GRAACC), Federal University of São Paulo, São Paulo-SP, Brazil. .,Department of Morphology and Genetics, Genetics Laboratory, Pediatric Oncology Institute (IOP/GRAACC), Federal University of São Paulo, São Paulo-SP, Brazil. .,Department of Clinical and Experimental Oncology, Genetics Laboratory, Pediatric Oncology Institute (IOP/GRAACC), Federal University of São Paulo, São Paulo-SP, Brazil.
| |
Collapse
|
22
|
Dobutovic B, Sudar E, Tepavcevic S, Djordjevic J, Djordjevic A, Radojcic M, Isenovic ER. Effects of ghrelin on protein expression of antioxidative enzymes and iNOS in the rat liver. Arch Med Sci 2014; 10:806-16. [PMID: 25276168 PMCID: PMC4175782 DOI: 10.5114/aoms.2014.44872] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/11/2012] [Revised: 01/14/2013] [Accepted: 02/24/2013] [Indexed: 01/17/2023] Open
Abstract
INTRODUCTION We investigated the effects of ghrelin on protein expression of the liver antioxidant enzymes superoxide dismutases (SODs), catalase (CAT), glutathione peroxidase (GPx), and glutathione reductase (GR), nuclear factor κB (NFκB) and inducible nitric oxide synthase (iNOS). Furthermore, we aimed to investigate whether extracellular regulated protein kinase (ERK1/2) and protein kinase B (Akt) are involved in ghrelin-regulated liver antioxidant enzymes and iNOS protein expression. MATERIAL AND METHODS Male Wistar rats were treated with ghrelin (0.3 nmol/5 µl) injected into the lateral cerebral ventricle every 24 h for 5 days, and 2 h after the last treatment the animals were sacrificed and the liver excised. The Western blot method was used to determine expression of antioxidant enzymes, iNOS, phosphorylation of Akt, ERK1/2 and nuclear factor κB (NFκB) subunits 50 and 65. RESULTS There was significantly higher protein expression of CuZnSOD (p < 0.001), MnSOD (p < 0.001), CAT (p < 0.001), GPx, (p < 0.001), and GR (p < 0.01) in the liver isolated from ghrelin-treated animals compared with control animals. In contrast, ghrelin significantly (p < 0.01) reduced protein expression of iNOS. In addition, phosphorylation of NFκB subunits p65 and p50 was significantly (p < 0.001 for p65; p < 0.05 for p50) reduced by ghrelin when compared with controls. Phosphorylation of ERK1/2 and of Akt was significantly higher in ghrelin-treated than in control animals (p < 0.05 for ERK1/2; p < 0.01 for Akt). CONCLUSIONS The results show that activation of Akt and ERK1/2 is involved in ghrelin-mediated regulation of protein expression of antioxidant enzymes and iNOS in the rat liver.
Collapse
Affiliation(s)
- Branislava Dobutovic
- Laboratory for Radiobiology and Molecular Genetics, Vinca Institute of Nuclear Sciences, University of Belgrade, Belgrade, Serbia
| | - Emina Sudar
- Laboratory for Radiobiology and Molecular Genetics, Vinca Institute of Nuclear Sciences, University of Belgrade, Belgrade, Serbia
| | - Snezana Tepavcevic
- Laboratory for Molecular Biology and Endocrinology, Vinca Institute of Nuclear Sciences, University of Belgrade, Belgrade, Serbia
| | - Jelena Djordjevic
- Laboratory for Molecular Biology and Endocrinology, Vinca Institute of Nuclear Sciences, University of Belgrade, Belgrade, Serbia
| | - Ana Djordjevic
- Laboratory for Molecular Biology and Endocrinology, Vinca Institute of Nuclear Sciences, University of Belgrade, Belgrade, Serbia
| | - Marija Radojcic
- Laboratory for Molecular Biology and Endocrinology, Vinca Institute of Nuclear Sciences, University of Belgrade, Belgrade, Serbia
| | - Esma R. Isenovic
- Laboratory for Radiobiology and Molecular Genetics, Vinca Institute of Nuclear Sciences, University of Belgrade, Belgrade, Serbia
| |
Collapse
|
23
|
Yi HX, Zhang M, Wang JY, Luo RB, Jiang SY, Wang JA. Determination of protein phosphatase type 2A in monocytes from multiple trauma patients: a potential biomarker for sepsis. J Surg Res 2014; 189:89-95. [DOI: 10.1016/j.jss.2014.02.040] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2013] [Revised: 02/17/2014] [Accepted: 02/22/2014] [Indexed: 12/29/2022]
|
24
|
Nishida M, Kozakai T, Nagami K, Kanamaru Y, Yabe T. Structural alteration of cell surface heparan sulfate through the stimulation of the signaling pathway for heparan sulfate 6-O-sulfotransferase-1 in mouse fibroblast cells. Biosci Biotechnol Biochem 2014; 78:770-9. [PMID: 25035978 DOI: 10.1080/09168451.2014.905178] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Heparan sulfate (HS) is a randomly sulfated polysaccharide that is present on the cell surface and in the extracellular matrix. The sulfated structures of HS were synthesized by multiple HS sulfotransferases, thereby regulating various activities such as growth factor signaling, cell differentiation, and tumor metastasis. Therefore, if the sulfated structures of HS could be artificially controlled, those manipulations would help to understand the various functions depending on HS. However, little knowledge is currently available to realize the mechanisms controlling the expression of such enzymes. In this study, we found that the ratio of 6-O-sulfated disaccharides increased at 3 h after adrenaline stimulation in mouse fibroblast cells. Furthermore, adrenaline-induced up-regulation of HS 6-O-sulfotransferase-1 (6-OST-1) was controlled by Src-ERK1/2 signaling pathway. Finally, inhibiting the signaling pathways for 6-OST-1 intentionally suppressed the adrenaline-induced structural alteration of HS. These observations provide fundamental insights into the understanding of structural alterations in HS by extracellular cues.
Collapse
Affiliation(s)
- Mitsutaka Nishida
- a United Graduate School of Agricultural Science, Gifu University , Gifu , Japan
| | | | | | | | | |
Collapse
|
25
|
Reassembly of JIP1 scaffold complex in JNK MAP kinase pathway using heterologous protein interactions. PLoS One 2014; 9:e96797. [PMID: 24816971 PMCID: PMC4016011 DOI: 10.1371/journal.pone.0096797] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2013] [Accepted: 04/11/2014] [Indexed: 12/31/2022] Open
Abstract
Formation of signaling protein complexes is crucial for proper signal transduction. Scaffold proteins in MAP kinase pathways are thought to facilitate complex assembly, thereby promoting efficient and specific signaling. To elucidate the assembly mechanism of scaffold complexes in mammals, we attempted to rationally rewire JIP1-dependent JNK MAP kinase pathway via alternative assembly of JIP1 complex. When JIP1-JNK docking interaction in the complex was replaced with heterologous protein interaction domains, such as PDZ domains and JNK-binding domains, a functional scaffold complex was reconstituted, and JNK signaling was rescued. Reassembly of JIP1 complex using heterologous protein interactions was sufficient for restoring of JNK MAP kinase pathway to induce signaling responses, including JNK activation and cell death. These results suggest a simple yet modular mechanism for JIP1 scaffold assembly in mammals.
Collapse
|
26
|
Lu Z, Yang Q, Cui M, Liu Y, Wang T, Zhao H, Dong Q. Tissue kallikrein induces SH-SY5Y cell proliferation via epidermal growth factor receptor and extracellular signal-regulated kinase1/2 pathway. Biochem Biophys Res Commun 2014; 446:25-9. [DOI: 10.1016/j.bbrc.2014.02.027] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2014] [Accepted: 02/06/2014] [Indexed: 10/25/2022]
|
27
|
Independent pathways downstream of the Wnd/DLK MAPKKK regulate synaptic structure, axonal transport, and injury signaling. J Neurosci 2013; 33:12764-78. [PMID: 23904612 DOI: 10.1523/jneurosci.5160-12.2013] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/25/2023] Open
Abstract
Mitogen-activated protein (MAP) kinase signaling cascades orchestrate diverse cellular activities with common molecular players. To achieve specific cellular outcomes in response to specific signals, scaffolding proteins play an important role. Here we investigate the role of the scaffolding protein JNK interacting protein-1 (JIP1) in neuronal signaling by a conserved axonal MAP kinase kinase kinase, known as Wallenda (Wnd) in Drosophila and dual leucine kinase (DLK) in vertebrates and Caenorhabditis elegans. Recent studies in multiple model organisms suggest that Wnd/DLK regulates both regenerative and degenerative responses to axonal injury. Here we report a new role for Wnd in regulating synaptic structure during development, which implies that Wnd is also active in uninjured neurons. This synaptic role of Wnd can be functionally separated from the role of Wnd in axonal regeneration and injury signaling by the requirement for the JIP1 scaffold and the p38b MAP kinase. JIP1 mediates the synaptic function of Wnd via p38, which is not required for injury signaling or new axonal growth after injury. Our results indicate that Wnd regulates multiple independent pathways in Drosophila motoneurons and that JIP1 scaffolds a specific downstream cascade required for the organization of presynaptic microtubules during synaptic development.
Collapse
|
28
|
Dual leucine zipper kinase as a therapeutic target for neurodegenerative conditions. Future Med Chem 2013; 5:1923-34. [DOI: 10.4155/fmc.13.150] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/18/2023] Open
Abstract
Dual leucine zipper kinase (DLK) is a serine/threonine protein kinase that is a member of the mixed lineage kinase subfamily. Mixed lineage kinases are upstream MAP3Ks that activate the JNK pathway. DLK is primarily responsible for activating JNK and mediating the apoptotic stress response in various cell types, specifically neurons. Inhibition and knockdown of DLK has been demonstrated to have neuroprotective effects in cellular and animal models of Alzheimer’s disease, glaucoma, Parkinson’s disease and other neurodegenerative conditions. Several series of ATP-binding site inhibitors have been identified through profiling efforts providing launch points for future medicinal chemistry programs.
Collapse
|
29
|
Cunningham CA, Knudson KM, Peng BJ, Teixeiro E, Daniels MA. The POSH/JIP-1 scaffold network regulates TCR-mediated JNK1 signals and effector function in CD8+T cells. Eur J Immunol 2013; 43:3361-71. [DOI: 10.1002/eji.201343635] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2013] [Revised: 07/15/2013] [Accepted: 08/16/2013] [Indexed: 12/21/2022]
Affiliation(s)
- Cody A. Cunningham
- Department of Molecular Microbiology and Immunology & Department of Surgery; Center for Cellular and Molecular Immunology, School of Medicine, University of Missouri; Columbia MO USA
| | - Karin M. Knudson
- Department of Molecular Microbiology and Immunology & Department of Surgery; Center for Cellular and Molecular Immunology, School of Medicine, University of Missouri; Columbia MO USA
| | - Binghao J. Peng
- Department of Molecular Microbiology and Immunology & Department of Surgery; Center for Cellular and Molecular Immunology, School of Medicine, University of Missouri; Columbia MO USA
| | - Emma Teixeiro
- Department of Molecular Microbiology and Immunology & Department of Surgery; Center for Cellular and Molecular Immunology, School of Medicine, University of Missouri; Columbia MO USA
| | - Mark A. Daniels
- Department of Molecular Microbiology and Immunology & Department of Surgery; Center for Cellular and Molecular Immunology, School of Medicine, University of Missouri; Columbia MO USA
| |
Collapse
|
30
|
Sun T, Yu N, Zhai LK, Li N, Zhang C, Zhou L, Huang Z, Jiang XY, Shen Y, Chen ZY. c-Jun NH2-terminal kinase (JNK)-interacting protein-3 (JIP3) regulates neuronal axon elongation in a kinesin- and JNK-dependent manner. J Biol Chem 2013; 288:14531-14543. [PMID: 23576431 DOI: 10.1074/jbc.m113.464453] [Citation(s) in RCA: 47] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
The development of neuronal polarity is essential for the establishment of the accurate patterning of neuronal circuits in the brain. However, little is known about the underlying molecular mechanisms that control rapid axon elongation during neuronal development. Here, we report that c-Jun NH2-terminal kinase (JNK)-interacting protein-3 (JIP3) is highly expressed at axon tips during the critical period for axon development. Using gain- and loss-of-function approaches, immunofluorescence analysis, and in utero electroporation, we find that JIP3 can enhance axon elongation in primary hippocampal neurons and cortical neurons in vivo. We further demonstrate that JIP3 promotes axon elongation in a kinesin- and JNK-dependent manner using several deletion mutants of JIP3. Next, we demonstrate that the successful transportation of JIP3 to axon tips by kinesin is a prerequisite for enhancing JNK phosphorylation in this area and therefore promotes axon elongation, constituting a novel mechanism for coupling JIP3 anterograde transport with JNK signaling at the distal axons and axon elongation. Finally, our immunofluorescence data suggest that the activation of JNK at axon tips facilitates axon elongation by modulating cofilin activity and actin filament dynamics. These findings may have important implications for our understanding of neuronal axon elongation during development.
Collapse
Affiliation(s)
- Tao Sun
- Department of Neurobiology, Shandong Provincial Key Laboratory of Mental Disorders, School of Medicine, Shandong University, No. 4 Wenhua Xi Road, Jinan, Shandong 250012
| | - Nuo Yu
- Department of Neurobiology, Shandong Provincial Key Laboratory of Mental Disorders, School of Medicine, Shandong University, No. 4 Wenhua Xi Road, Jinan, Shandong 250012
| | - Lu-Kai Zhai
- Department of Neurobiology, Shandong Provincial Key Laboratory of Mental Disorders, School of Medicine, Shandong University, No. 4 Wenhua Xi Road, Jinan, Shandong 250012
| | - Na Li
- Department of Neurobiology, Shandong Provincial Key Laboratory of Mental Disorders, School of Medicine, Shandong University, No. 4 Wenhua Xi Road, Jinan, Shandong 250012
| | - Chao Zhang
- Department of Neurobiology, Shandong Provincial Key Laboratory of Mental Disorders, School of Medicine, Shandong University, No. 4 Wenhua Xi Road, Jinan, Shandong 250012
| | - Liang Zhou
- Department of Neurobiology, Key Laboratory of Medical Neurobiology of the Ministry of Health, Zhejiang Province Key Laboratory of Neurobiology, School of Medicine, Zhejiang University, Hangzhou, Zhejiang 310058
| | - Zhuo Huang
- Key Laboratory for Biological Effects of Nanomaterials and Nanosafety, National Center for Nanoscience and Technology, Chinese Academy of Sciences, Beijing 100190, China
| | - Xing-Yu Jiang
- Key Laboratory for Biological Effects of Nanomaterials and Nanosafety, National Center for Nanoscience and Technology, Chinese Academy of Sciences, Beijing 100190, China
| | - Ying Shen
- Department of Neurobiology, Key Laboratory of Medical Neurobiology of the Ministry of Health, Zhejiang Province Key Laboratory of Neurobiology, School of Medicine, Zhejiang University, Hangzhou, Zhejiang 310058
| | - Zhe-Yu Chen
- Department of Neurobiology, Shandong Provincial Key Laboratory of Mental Disorders, School of Medicine, Shandong University, No. 4 Wenhua Xi Road, Jinan, Shandong 250012.
| |
Collapse
|
31
|
Goldsmith CS, Bell-Pedersen D. Diverse roles for MAPK signaling in circadian clocks. ADVANCES IN GENETICS 2013; 84:1-39. [PMID: 24262095 DOI: 10.1016/b978-0-12-407703-4.00001-3] [Citation(s) in RCA: 70] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
The mitogen-activated protein kinase (MAPK) family of genes aids cells in sensing both extracellular and intracellular stimuli, and emerging data indicate that MAPKs have fundamental, yet diverse, roles in the circadian biological clock. In the mammalian suprachiasmatic nucleus (SCN), MAPK pathways can function as inputs allowing the endogenous clock to entrain to 24h environmental cycles. MAPKs can also interact physically and/or genetically with components of the molecular circadian oscillator, implying that MAPKs can affect the cycling of the clock. Finally, circadian rhythms in MAPK pathway activation exist in many different tissue types and in model organisms, providing a mechanism to coordinately control the expression tissue-specific target genes at the proper time of day. As such, it should probably not come as a surprise that MAPK signaling pathways and circadian clocks affect similar biological processes and defects in either pathway lead to many of the same types of human diseases, highlighting the need to better define the mechanisms that link these two fundamental pathways together.
Collapse
|
32
|
Song IS, Jun SY, Na HJ, Kim HT, Jung SY, Ha GH, Park YH, Long LZ, Yu DY, Kim JM, Kim JH, Ko JH, Kim CH, Kim NS. Inhibition of MKK7-JNK by the TOR signaling pathway regulator-like protein contributes to resistance of HCC cells to TRAIL-induced apoptosis. Gastroenterology 2012; 143:1341-1351. [PMID: 22841785 DOI: 10.1053/j.gastro.2012.07.103] [Citation(s) in RCA: 41] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/09/2011] [Revised: 06/06/2012] [Accepted: 07/19/2012] [Indexed: 12/21/2022]
Abstract
BACKGROUND & AIMS The TOR signaling pathway regulator-like (TIPRL) protein, the mammalian ortholog of yeast TIP41, was identified in an expression profiling screen for factors that regulate human liver carcinogenesis. We investigated the role of human TIPRL protein in hepatocellular carcinoma (HCC). METHODS We measured the level of TIPRL in HCC and adjacent nontumor tissues from patients. We used small interfering RNAs and zebrafish to study the function of TIPRL. We used annexin V propidium iodide staining and immunoblot analyses to measure apoptosis and activation of apoptotic signaling pathways. We used confocal microscopy, coimmunoprecipitation, and glutathione-S transferase pull-down analyses to determine interactions among mitogen-activated protein kinase kinase 7 (MKK7 or MAP2K7), TIPRL, and the protein phosphatase type 2A (PP2Ac). We studied the effects of TIPRL in tumor xenografts in mice. RESULTS Levels of TIPRL were higher in HCC tissues and cell lines than nontumor tissues and primary hepatocytes. Knockdown of tiprl expression in zebrafish led to large amounts of apoptosis throughout the embryos. Incubation of HCC cells, but not primary human hepatocytes, with small interfering RNA against TIPRL (siTIPRL) and tumor necrosis factor-related apoptosis-inducing ligand (TRAIL) caused prolonged activation (phosphorylation) of MKK7 and c-Jun N-terminal kinase (JNK) and led to apoptosis, indicated by cleavage of procaspase-8,-3 and of poly-(adenosine diphosphate-ribose) polymerase. TIPRL bound to MKK7 and PP2Ac and promoted the interaction between MKK7 and PP2Ac. In mice, injection of HCC xenograft tumors with siTIPRL and TRAIL led to tumor apoptosis and regression. CONCLUSIONS TIPRL is highly up-regulated in human HCC samples and cell lines, compared with noncancerous liver tissues. TIPRL prevents prolonged activation of MKK7 and JNK and TRAIL-induced apoptosis by mediating the interaction between MKK7 and PP2Ac.
Collapse
Affiliation(s)
- In Sung Song
- Medical Genomics Research Center, Korea Research Institute of Bioscience and Biotechnology, Daejeon, South Korea; Cardiovascular and Metabolic Disease Center, Inje University, Busan, South Korea
| | - Soo Young Jun
- Medical Genomics Research Center, Korea Research Institute of Bioscience and Biotechnology, Daejeon, South Korea; Department of Functional Genomics, University of Science and Technology, Daejeon, South Korea
| | - Hee-Jun Na
- Department of Functional Genomics, University of Science and Technology, Daejeon, South Korea; Renal Division, Brigham and Women's Hospital, Department of Medicine, Harvard Medical School, Boston, Massachusetts
| | - Hyun-Taek Kim
- Department of Biology, Chungnam National University, Daejeon, South Korea
| | - So Young Jung
- Medical Genomics Research Center, Korea Research Institute of Bioscience and Biotechnology, Daejeon, South Korea
| | - Ga Hee Ha
- Medical Genomics Research Center, Korea Research Institute of Bioscience and Biotechnology, Daejeon, South Korea
| | - Young-Ho Park
- Department of Functional Genomics, University of Science and Technology, Daejeon, South Korea; Aging Research Center, Korea Research Institute of Bioscience and Biotechnology, Daejeon, South Korea
| | - Liang Zhe Long
- Department of Pathology, School of Medicine, Chungnam National University, Daejeon, South Korea
| | - Dae-Yeul Yu
- Department of Functional Genomics, University of Science and Technology, Daejeon, South Korea; Aging Research Center, Korea Research Institute of Bioscience and Biotechnology, Daejeon, South Korea
| | - Jin-Man Kim
- Department of Pathology, School of Medicine, Chungnam National University, Daejeon, South Korea
| | - Joo Heon Kim
- Department of Pathology, Eulji University School of Medicine, Daejeon, South Korea
| | - Jeong-Heon Ko
- Daejeon-KRIBB-FHCRC Research Cooperation Center, Korea Research Institute of Bioscience and Biotechnology, Daejeon, South Korea
| | - Cheol-Hee Kim
- Department of Biology, Chungnam National University, Daejeon, South Korea.
| | - Nam-Soon Kim
- Medical Genomics Research Center, Korea Research Institute of Bioscience and Biotechnology, Daejeon, South Korea; Department of Functional Genomics, University of Science and Technology, Daejeon, South Korea.
| |
Collapse
|
33
|
Yang SH, Sharrocks AD, Whitmarsh AJ. MAP kinase signalling cascades and transcriptional regulation. Gene 2012; 513:1-13. [PMID: 23123731 DOI: 10.1016/j.gene.2012.10.033] [Citation(s) in RCA: 313] [Impact Index Per Article: 26.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2012] [Revised: 10/08/2012] [Accepted: 10/18/2012] [Indexed: 02/06/2023]
Abstract
The MAP kinase (MAPK) signalling pathways play fundamental roles in a wide range of cellular processes and are often deregulated in disease states. One major mode of action for these pathways is in controlling gene expression, in particular through regulating transcription. In this review, we discuss recent significant advances in this area. In particular we focus on the mechanisms by which MAPKs are targeted to the nucleus and chromatin, and once there, how they impact on chromatin structure and subsequent gene regulation. We also discuss how systems biology approaches have contributed to our understanding of MAPK signaling networks, and also how the MAPK pathways intersect with other regulatory pathways in the nucleus. Finally, we summarise progress in studying the physiological functions of key MAPK transcriptional targets.
Collapse
Affiliation(s)
- Shen-Hsi Yang
- Faculty of Life Sciences, University of Manchester, Michael Smith Building, Oxford Road, Manchester, M13 9PT, UK
| | | | | |
Collapse
|
34
|
p38α mitogen-activated protein kinase depletion and repression of signal transduction to translation machinery by miR-124 and -128 in neurons. Mol Cell Biol 2012; 33:127-35. [PMID: 23109423 DOI: 10.1128/mcb.00695-12] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022] Open
Abstract
The p38α to p38δ mitogen-activated protein kinases (MAPKs) are central regulatory nodes coordinating acute stress and inflammatory responses. Their activation leads to rapid adjustment of protein synthesis, for instance translational induction of proinflammatory cytokines. The only known direct link of p38 to translation machinery is the MAPK signal-integrating kinase Mnk. Only p38α and p38β transcripts are ubiquitously expressed. These mRNAs encode highly conserved proteins that equally phosphorylate recombinant Mnk1 in vitro. We discovered that expression of the p38α protein, but not the p38β isoform, is suppressed in the brain. This is due to p38α depletion by two neuron-selective microRNAs (miRNAs), miR-124 and -128. Suppression of p38α protein was reversed by miR-124/-128 antisense oligonucleotides in primary explant neuronal cultures. Targeted p38α depletion reduced Mnk1 activation, which cannot be compensated by p38β. Our research shows that p38α alone controls acute stress and cytokine signaling from p38 MAPK to translation machinery. This regulatory axis is greatly diminished in neurons, which may insulate brain physiology and function from p38α-Mnk1-mediated signaling.
Collapse
|
35
|
Abstract
Receptor internalization is a common mechanism underlying surface receptor down-regulation (and thus receptor signaling) upon its engagement with the cognate ligand. Tight regulation of surface CD40 expression is critical in regulating different functional properties of dendritic cell (DC). Engagement of CD40 on mature DC and the cognate CD40 ligand on T cell activates c-Jun N-terminal MAPK, p38 and ERK1/2 MAPK pathways in mature DC. JNK-associated leucine zipper protein (JLP) is a scaffolding protein that interacted with p38 and JNK. The molecular mechanism underlying CD40 internalization and its physiological impact on DC functions remained unclear. Here we reported that the engagement of CD40 on the LPS-activated DC down-regulated the surface expression of CD40. We examined the role of the JLP protein in DC differentiation, and in the regulation of DC function(s) in vitro. In contrast to the abundant JLP expression observed in immortal cell lines, primary immature DC expressed low levels of the JLP proteins. The induction of the JLP protein expression was observed in the LPS-mature DC that were activated by CD40 ligation, and also in the poly I:C stimulated DC. JLP-silenced DC was impaired in regulating CD40 surface expression upon LPS stimulation and CD40 induced receptor internalization. Such aberrant change in the regulation of surface CD40 expression was associated with an augmented capacity of the JLP-silenced DC in IL-12 production. Collectively, our data identified a novel role of a scaffolding protein JLP in the regulation of surface CD40 expression and fine-tuning of DC function.
Collapse
|
36
|
Nagano K, Akpan A, Warnasuriya G, Corless S, Totty N, Yang A, Stein R, Zvelebil M, Stensballe A, Burlingame A, Waterfield M, Cramer R, Timms JF, Naaby-Hansen S. Functional proteomic analysis of long-term growth factor stimulation and receptor tyrosine kinase coactivation in Swiss 3T3 fibroblasts. Mol Cell Proteomics 2012; 11:1690-708. [PMID: 22956732 DOI: 10.1074/mcp.m112.019778] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
In Swiss 3T3 fibroblasts, long-term stimulation with PDGF, but not insulin-like growth factor 1 (IGF-1) or EGF, results in the establishment of an elongated migratory phenotype, characterized by the formation of retractile dendritic protrusions and absence of actin stress fibers and focal adhesion complexes. To identify receptor tyrosine kinase-specific reorganization of the Swiss 3T3 proteome during phenotypic differentiation, we compared changes in the pattern of protein synthesis and phosphorylation during long-term exposure to PDGF, IGF-1, EGF, and their combinations using 2DE-based proteomics after (35)S- and (33)P-metabolic labeling. One hundred and five differentially regulated proteins were identified by mass spectrometry and some of these extensively validated. PDGF stimulation produced the highest overall rate of protein synthesis at any given time and induced the most sustained phospho-signaling. Simultaneous activation with two or three of the growth factors revealed both synergistic and antagonistic effects on protein synthesis and expression levels with PDGF showing dominance over both IGF-1 and EGF in generating distinct proteome compositions. Using signaling pathway inhibitors, PI3K was identified as an early site for signal diversification, with sustained activity of the PI3K/AKT pathway critical for regulating late protein synthesis and phosphorylation of target proteins and required for maintaining the PDGF-dependent motile phenotype. Several proteins were identified with novel PI3K/Akt-dependent synthesis and phosphorylations including eEF2, PRS7, RACK-1, acidic calponin, NAP1L1, Hsp73, and fascin. The data also reveal induction/suppression of key F-actin and actomyosin regulators and chaperonins that enable PDGFR to direct the assembly of a motile cytoskeleton, despite simultaneous antagonistic signaling activities. Together, the study demonstrates that long-term exposure to different growth factors results in receptor tyrosine kinase-specific regulation of relatively small subproteomes, and implies that the strength and longevity of receptor tyrosine kinase-specific signals are critical in defining the composition and functional activity of the resulting proteome.
Collapse
Affiliation(s)
- Kohji Nagano
- Discovery Research Department, Chugai Pharmaceutical Co. Ltd., Japan
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
37
|
Zalatan JG, Coyle SM, Rajan S, Sidhu SS, Lim WA. Conformational control of the Ste5 scaffold protein insulates against MAP kinase misactivation. Science 2012; 337:1218-22. [PMID: 22878499 DOI: 10.1126/science.1220683] [Citation(s) in RCA: 83] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
Cells reuse signaling proteins in multiple pathways, raising the potential for improper cross talk. Scaffold proteins are thought to insulate against such miscommunication by sequestering proteins into distinct physical complexes. We show that the scaffold protein Ste5, which organizes the yeast mating mitogen-activated protein kinase (MAPK) pathway, does not use sequestration to prevent misactivation of the mating response. Instead, Ste5 appears to use a conformation mechanism: Under basal conditions, an intramolecular interaction of the pleckstrin homology (PH) domain with the von Willebrand type A (VWA) domain blocks the ability to coactivate the mating-specific MAPK Fus3. Pheromone-induced membrane binding of Ste5 triggers release of this autoinhibition. Thus, in addition to serving as a conduit guiding kinase communication, Ste5 directly receives input information to decide if and when signal can be transmitted to mating output.
Collapse
Affiliation(s)
- Jesse G Zalatan
- Department of Cellular and Molecular Pharmacology, University of California, San Francisco, 600 16th Street, San Francisco, CA 94158, USA
| | | | | | | | | |
Collapse
|
38
|
Nithianandarajah-Jones GN, Wilm B, Goldring CEP, Müller J, Cross MJ. ERK5: structure, regulation and function. Cell Signal 2012; 24:2187-96. [PMID: 22800864 DOI: 10.1016/j.cellsig.2012.07.007] [Citation(s) in RCA: 170] [Impact Index Per Article: 14.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2012] [Accepted: 07/09/2012] [Indexed: 01/06/2023]
Abstract
Extracellular signal-regulated kinase 5 (ERK5), also termed big mitogen-activated protein kinase-1 (BMK1), is the most recently identified member of the mitogen-activated protein kinase (MAPK) family and consists of an amino-terminal kinase domain, with a relatively large carboxy-terminal of unique structure and function that makes it distinct from other MAPK members. It is ubiquitously expressed in numerous tissues and is activated by a variety of extracellular stimuli, such as cellular stresses and growth factors, to regulate processes such as cell proliferation and differentiation. Targeted deletion of Erk5 in mice has revealed that the ERK5 signalling cascade plays a critical role in cardiovascular development and vascular integrity. Recent data points to a potential role in pathological conditions such as cancer and tumour angiogenesis. This review focuses on the physiological and pathological role of ERK5, the regulation of this kinase and the recent development of small molecule inhibitors of the ERK5 signalling cascade.
Collapse
Affiliation(s)
- Gopika N Nithianandarajah-Jones
- MRC Centre for Drug Safety Science, Department of Molecular and Clinical Pharmacology, Institute of Translational Medicine, University of Liverpool, Liverpool, UK
| | | | | | | | | |
Collapse
|
39
|
Khromov AS, Momotani K, Jin L, Artamonov MV, Shannon J, Eto M, Somlyo AV. Molecular mechanism of telokin-mediated disinhibition of myosin light chain phosphatase and cAMP/cGMP-induced relaxation of gastrointestinal smooth muscle. J Biol Chem 2012; 287:20975-85. [PMID: 22544752 DOI: 10.1074/jbc.m112.341479] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Phospho-telokin is a target of elevated cyclic nucleotide concentrations that lead to relaxation of gastrointestinal and some vascular smooth muscles (SM). Here, we demonstrate that in telokin-null SM, both Ca(2+)-activated contraction and Ca(2+) sensitization of force induced by a GST-MYPT1(654-880) fragment inhibiting myosin light chain phosphatase were antagonized by the addition of recombinant S13D telokin, without changing the inhibitory phosphorylation status of endogenous MYPT1 (the regulatory subunit of myosin light chain phosphatase) at Thr-696/Thr-853 or activity of Rho kinase. Cyclic nucleotide-induced relaxation of force in telokin-null ileum muscle was reduced but not correlated with a change in MYPT1 phosphorylation. The 40% inhibited activity of phosphorylated MYPT1 in telokin-null ileum homogenates was restored to nonphosphorylated MYPT1 levels by addition of S13D telokin. Using the GST-MYPT1 fragment as a ligand and SM homogenates from WT and telokin KO mice as a source of endogenous proteins, we found that only in the presence of endogenous telokin, thiophospho-GST-MYPT1 co-precipitated with phospho-20-kDa myosin regulatory light chain 20 and PP1. Surface plasmon resonance studies showed that S13D telokin bound to full-length phospho-MYPT1. Results of a protein ligation assay also supported interaction of endogenous phosphorylated MYPT1 with telokin in SM cells. We conclude that the mechanism of action of phospho-telokin is not through modulation of the MYPT1 phosphorylation status but rather it contributes to cyclic nucleotide-induced relaxation of SM by interacting with and activating the inhibited full-length phospho-MYPT1/PP1 through facilitating its binding to phosphomyosin and thus accelerating 20-kDa myosin regulatory light chain dephosphorylation.
Collapse
Affiliation(s)
- Alexander S Khromov
- Departments of Molecular Physiology and Biological Physics, University of Virginia, Charlottesville, Virginia 22908, USA
| | | | | | | | | | | | | |
Collapse
|
40
|
Janitza P, Ullrich KK, Quint M. Toward a comprehensive phylogenetic reconstruction of the evolutionary history of mitogen-activated protein kinases in the plant kingdom. FRONTIERS IN PLANT SCIENCE 2012; 3:271. [PMID: 23230446 PMCID: PMC3515877 DOI: 10.3389/fpls.2012.00271] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/31/2012] [Accepted: 11/19/2012] [Indexed: 05/06/2023]
Abstract
The mitogen-activated protein kinase (MAPK) pathway is a three-tier signaling cascade that transmits cellular information from the plasma membrane to the cytoplasm where it triggers downstream responses. The MAPKs represent the last step in this cascade and are activated when both tyrosine and threonine residues in a conserved TxY motif are phosphorylated by MAPK kinases, which in turn are themselves activated by phosphorylation by MAPK kinase kinases. To understand the molecular evolution of MAPKs in the plant kingdom, we systematically conducted a Hidden-Markov-Model based screen to identify MAPKs in 13 completely sequenced plant genomes. In this analysis, we included green algae, bryophytes, lycophytes, and several mono- and eudicotyledonous species covering >800 million years of evolution. The phylogenetic relationships of the 204 identified MAPKs based on Bayesian inference facilitated the retraction of the sequence of emergence of the four major clades that are characterized by the presence of a TDY or TEY-A/TEY-B/TEY-C type kinase activation loop. We present evidence that after the split of TDY- and TEY-type MAPKs, initially the TEY-C clade emerged. This was followed by the TEY-B clade in early land plants until the TEY-A clade finally emerged in flowering plants. In addition to these well characterized clades, we identified another highly conserved clade of 45 MAPK-likes, members of which were previously described as Mak-homologous kinases. In agreement with their essential functions, molecular population genetic analysis of MAPK genes in Arabidopsis thaliana accessions reveal that purifying selection drove the evolution of the MAPK family, implying strong functional constraints on MAPK genes. Closely related MAPKs most likely subfunctionalized, a process in which differential transcriptional regulation of duplicates may be involved.
Collapse
Affiliation(s)
- Philipp Janitza
- Department of Molecular Signal Processing, Leibniz Institute of Plant BiochemistryHalle (Saale), Germany
| | - Kristian Karsten Ullrich
- Department of Molecular Signal Processing, Leibniz Institute of Plant BiochemistryHalle (Saale), Germany
| | - Marcel Quint
- Department of Molecular Signal Processing, Leibniz Institute of Plant BiochemistryHalle (Saale), Germany
- *Correspondence: Marcel Quint, Department of Molecular Signal Processing, Leibniz Institute of Plant Biochemistry, Weinberg 03, 06120 Halle (Saale), Germany. e-mail:
| |
Collapse
|
41
|
Drew BA, Burow ME, Beckman BS. MEK5/ERK5 pathway: the first fifteen years. Biochim Biophys Acta Rev Cancer 2011; 1825:37-48. [PMID: 22020294 DOI: 10.1016/j.bbcan.2011.10.002] [Citation(s) in RCA: 104] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2011] [Accepted: 10/07/2011] [Indexed: 12/22/2022]
Abstract
While conventional MAP kinase pathways are one of the most highly studied signal transduction molecules, less is known about the MEK5 signaling pathway. This pathway has been shown to play a role in normal cell growth cycles, survival and differentiation. The MEK5 pathway is also believed to mediate the effects of a number of oncogenes. MEK5 is the upstream activator of ERK5 in many epithelial cells. Activation of the MEK-MAPK pathway is a frequent event in malignant tumor formation and contributes to chemoresistance and anti-apoptotic signaling. This pathway may be involved in a number of more aggressive, metastatic varieties of cancer due to its role in cell survival, proliferation and EMT transitioning. Further study of this pathway may lead to new prognostic factors and new drug targets to combat more aggressive forms of cancer.
Collapse
Affiliation(s)
- Barbara A Drew
- Department of Pharmacology, Tulane University School of Medicine, New Orleans, LA 70112, USA
| | | | | |
Collapse
|
42
|
Lee SK, Kim BG, Kwon TR, Jeong MJ, Park SR, Lee JW, Byun MO, Kwon HB, Matthews BF, Hong CB, Park SC. Overexpression of the mitogen-activated protein kinase gene OsMAPK33 enhances sensitivity to salt stress in rice (Oryza sativa L.). J Biosci 2011; 36:139-51. [PMID: 21451255 DOI: 10.1007/s12038-011-9002-8] [Citation(s) in RCA: 54] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
Mitogen-activated protein kinases (MAPK) signalling cascades are activated by extracellular stimuli such as environmental stresses and pathogens in higher eukaryotic plants. To know more about MAPK signalling in plants, aMAPK cDNA clone, OsMAPK33, was isolated from rice. The gene is mainly induced by drought stress. In phylogenetic analysis, OsMAPK33 (Os02g0148100) showed approximately 47-93% identity at the amino acid level with other plant MAPKs. It was found to exhibit organ-specific expression with relatively higher expression in leaves as compared with roots or stems, and to exist as a single copy in the rice genome. To investigate the biological functions of OsMAPK33 in rice MAPK signalling, transgenic rice plants that either overexpressed or suppressed OsMAPK33 were made. Under dehydration conditions, the suppressed lines showed lower osmotic potential compared with that of wild-type plants, suggesting a role of OsMAPK33 in osmotic homeostasis. Nonetheless, the suppressed lines did not display any significant difference in drought tolerance compared with their wild-type plants. With increased salinity, there was still no difference in salt tolerance between OsMAPK33-suppressed lines and their wild-type plants. However, the overexpressing lines showed greater reduction in biomass accumulation and higher sodium uptake into cells, resulting in a lower K+/Na+ ratio inside the cell than that in the wild-type plants and OsMAPK33-suppressed lines. These results suggest that OsMAPK33 could play a negative role in salt tolerance through unfavourable ion homeostasis. Gene expression profiling of OsMAPK33 transgenic lines through rice DNA chip analysis showed that OsMAPK33 altered expression of genes involved in ion transport. Further characterization of downstream components will elucidate various biological functions of this novel rice MAPK.
Collapse
Affiliation(s)
- Seong-Kon Lee
- Department of Agricultural Biotechnology, National Academy of Agricultural Science, Rural Development Administration, Suwon 441-857, Republic of Korea
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
43
|
Wang LL, Zhao XC, Yan LF, Wang YQ, Cheng X, Fu R, Zhou LH. C-jun phosphorylation contributes to down regulation of neuronal nitric oxide synthase protein and motoneurons death in injured spinal cords following root-avulsion of the brachial plexus. Neuroscience 2011; 189:397-407. [PMID: 21596101 DOI: 10.1016/j.neuroscience.2011.04.070] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2010] [Revised: 04/11/2011] [Accepted: 04/30/2011] [Indexed: 01/11/2023]
Abstract
Previous studies have shown that c-jun and neuronal nitric oxide synthase (nNOS) are both induced in injured motoneurons, but their roles in motoneuron death remain unclear. We hypothesized that nNOS might be the downstream effector of c-jun N-terminal kinase (JNK)/c-jun in avulsion-induced motoneuron death. Here, we found that brachial root-avulsion induced a temporary increase in JNK activity and three- and four-fold increases in phospho-c-jun and c-jun, respectively; however, brachial root-avulsion caused a decrease in nNOS protein expression from 4 h to 14 days post-injury. At 14 days post-injury, almost all nNOS-positive motoneurons were co-localized with phospho-c-jun-positive motoneurons in ipsilateral ventral horns. The JNK inhibitor SP600125, applied immediately post-injury, resulted in an upregulation of nNOS protein both in injured spinal cords and motoneurons and caused a slight alleviation of motoneuron death by inhibiting c-jun phosphorylation at 14 days post-injury. Our results demonstrated that the JNK/c-jun signal transduction pathway is involved in root-avulsion. The inhibition of c-jun phosphorylation prevents nNOS levels from dropping below baseline levels in the spinal cord and partially alleviates motoneuron death following root-avulsion. Therefore, inhibiting c-jun phosphorylation or up-regulating the nNOS protein in injured spinal cords at the early stage might be used in the future as the molecular-target strategies to prevent the motoneurons degeneration in root-avulsion.
Collapse
Affiliation(s)
- L-L Wang
- Zhong Shan School of Medicine, Sun Yat-Sen University, No. 74 Zhongshan Road 2, Guangzhou 510080, PR China
| | | | | | | | | | | | | |
Collapse
|
44
|
Abstract
The opportunistic human fungal pathogen Candida albicans encounters diverse environmental stresses when it is in contact with its host. When colonizing and invading human tissues, C. albicans is exposed to ROS (reactive oxygen species) and RNIs (reactive nitrogen intermediates). ROS and RNIs are generated in the first line of host defence by phagocytic cells such as macrophages and neutrophils. In order to escape these host-induced oxidative and nitrosative stresses, C. albicans has developed various detoxification mechanisms. One such mechanism is the detoxification of NO (nitric oxide) to nitrate by the flavohaemoglobin enzyme CaYhb1. Members of the haemoglobin superfamily are highly conserved and are found in archaea, eukaryotes and bacteria. Flavohaemoglobins have a dioxygenase activity [NOD (NO dioxygenase domain)] and contain three domains: a globin domain, an FAD-binding domain and an NAD(P)-binding domain. In the present paper, we examine the nitrosative stress response in three fungal models: the pathogenic yeast C. albicans, the benign budding yeast Saccharomyces cerevisiae and the benign fission yeast Schizosaccharomyces pombe. We compare their enzymatic and non-enzymatic NO and RNI detoxification mechanisms and summarize fungal responses to nitrosative stress.
Collapse
|
45
|
Sinha AK, Jaggi M, Raghuram B, Tuteja N. Mitogen-activated protein kinase signaling in plants under abiotic stress. PLANT SIGNALING & BEHAVIOR 2011; 6:196-203. [PMID: 21512321 PMCID: PMC3121978 DOI: 10.4161/psb.6.2.14701] [Citation(s) in RCA: 265] [Impact Index Per Article: 20.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/26/2010] [Accepted: 12/30/2010] [Indexed: 05/18/2023]
Abstract
Mitogen-activated protein kinase cascade is evolutionarily conserved signal transduction module involved in transducing extracellular signals to the nucleus for appropriate cellular adjustment. This cascade consists essentially of three components, a MAPK kinase kinase (MAPKKK), a MAPK kinase (MAPKK) and a MAPK connected to each other by the event of phosphorylation. These kinases play various roles in intra- and extra-cellular signaling in plants by transferring the information from sensors to responses. Signaling through MAP kinase cascade can lead to cellular responses including cell division, differentiation as well as responses to various stresses. MAPK signaling has also been associated with hormonal responses. In plants, MAP kinases are represented by multigene families and are involved in efficient transmission of specific stimuli and also involved in the regulation of the antioxidant defense system in response to stress signaling. In the current review we summarize and investigate the participation of MAPKs as possible mediators of various abiotic stresses in plants.
Collapse
Affiliation(s)
- Alok Krishna Sinha
- National Institute of Plant Genome Research; Aruna Asaf Ali Marg; New Delhi, India
| | - Monika Jaggi
- National Institute of Plant Genome Research; Aruna Asaf Ali Marg; New Delhi, India
| | - Badmi Raghuram
- National Institute of Plant Genome Research; Aruna Asaf Ali Marg; New Delhi, India
| | - Narendra Tuteja
- International Centre for Genetic Engineering and Biotechnology; Aruna Asaf Ali Marg; New Delhi, India
| |
Collapse
|
46
|
Tang CH, Chen CF, Chen WM, Fong YC. IL-6 increases MMP-13 expression and motility in human chondrosarcoma cells. J Biol Chem 2011; 286:11056-66. [PMID: 21278254 DOI: 10.1074/jbc.m110.204081] [Citation(s) in RCA: 41] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Chondrosarcoma is a type of highly malignant tumor with a potent capacity to invade locally and cause distant metastasis. Chondrosarcoma shows a predilection for metastasis to the lungs. IL-6 is a multifunctional cytokine that is associated with the disease status and outcomes of cancers. However, the effect of IL-6 on the migration activity of human chondrosarcoma cells is mostly unknown. Here, we found that IL-6 increased the migration and expression of MMP-13 in human chondrosarcoma cells. We also found that human chondrosarcoma tissues had significant expression of IL-6, which was higher than that in normal cartilage. IL-6-mediated migration and MMP-13 up-regulation were attenuated by anti-IL-6 receptor antibody, Ras, Raf-1, and a MEK inhibitor. Activation of the Ras, Raf-1, MEK, ERK, and NF-κB signaling pathways after IL-6 treatment was demonstrated, and IL-6-induced MMP-13 expression and migration activity were inhibited by the specific inhibitor and mutant Ras, Raf-1, MEK, ERK, and NF-κB cascades. In addition, migration-prone sublines demonstrated that cells with increasing migration ability had greater expression of IL-6 and MMP-13. Taken together, these results indicate that IL-6 and IL-6 receptor interaction enhances migration of chondrosarcoma through an increase in MMP-13 production.
Collapse
Affiliation(s)
- Chih-Hsin Tang
- Department of Pharmacology, School of Medicine, China Medical University and Hospital, Taichung 404, Taiwan.
| | | | | | | |
Collapse
|
47
|
Prokudin I, Stasyk T, Rainer J, Bonn GK, Kofler R, Huber LA. Comprehensive proteomic and transcriptomic characterization of hepatic expression signatures affected in p14 liver conditional knockout mice. Proteomics 2011; 11:469-80. [PMID: 21268275 DOI: 10.1002/pmic.201000400] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2010] [Revised: 11/16/2010] [Accepted: 11/17/2010] [Indexed: 01/20/2023]
Abstract
Scaffold proteins regulate intracellular MAP kinase signaling by providing critical spatial and temporal specificities. We have shown previously that the scaffold protein MEK1 partner (MP1) is localized to late endosomes by the adaptor protein p14. Using conditional gene disruption of p14 in livers of mice (p14(Δhep) ) we analyzed protein and transcript signatures in tissue samples. Further biological network analysis predicted that the differentially expressed transcripts and proteins are involved in cell cycle progression and regulation of cellular proliferation. Although some of the here identified signatures were previously linked to phospho-ERK activity, most of them were novel targets of the late endosomal p14/MP1/MEK/ERK signaling module. Finally, the proliferation defect was confirmed in a chemically induced liver regeneration model in p14(Δhep) knockout mice.
Collapse
Affiliation(s)
- Ivan Prokudin
- Division of Cell Biology, Biocenter, Innsbruck Medical University, Innsbruck, Austria
| | | | | | | | | | | |
Collapse
|
48
|
Bilimoria PM, de la Torre-Ubieta L, Ikeuchi Y, Becker EBE, Reiner O, Bonni A. A JIP3-regulated GSK3β/DCX signaling pathway restricts axon branching. J Neurosci 2010; 30:16766-76. [PMID: 21159948 PMCID: PMC3409248 DOI: 10.1523/jneurosci.1362-10.2010] [Citation(s) in RCA: 48] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2010] [Revised: 06/21/2010] [Accepted: 06/27/2010] [Indexed: 01/26/2023] Open
Abstract
Axon branching plays a critical role in establishing the accurate patterning of neuronal circuits in the brain. However, the mechanisms that control axon branching remain poorly understood. Here we report that knockdown of the brain-enriched signaling protein JNK-interacting protein 3 (JIP3) triggers exuberant axon branching and self-contact in primary granule neurons of the rat cerebellar cortex. JIP3 knockdown in cerebellar slices and in postnatal rat pups in vivo leads to the formation of ectopic branches in granule neuron parallel fiber axons in the cerebellar cortex. We also find that JIP3 restriction of axon branching is mediated by the protein kinase glycogen synthase kinase 3β (GSK3β). JIP3 knockdown induces the downregulation of GSK3β in neurons, and GSK3β knockdown phenocopies the effect of JIP3 knockdown on axon branching and self-contact. Finally, we establish doublecortin (DCX) as a novel substrate of GSK3β in the control of axon branching and self-contact. GSK3β phosphorylates DCX at the distinct site of Ser327 and thereby contributes to DCX function in the restriction of axon branching. Together, our data define a JIP3-regulated GSK3β/DCX signaling pathway that restricts axon branching in the mammalian brain. These findings may have important implications for our understanding of neuronal circuitry during development, as well as the pathogenesis of neurodevelopmental disorders of cognition.
Collapse
Affiliation(s)
- Parizad M. Bilimoria
- Department of Pathology and
- Program in Neuroscience, Harvard Medical School, Boston, Massachusetts 02115, and
| | - Luis de la Torre-Ubieta
- Department of Pathology and
- Program in Neuroscience, Harvard Medical School, Boston, Massachusetts 02115, and
| | | | | | - Orly Reiner
- Department of Molecular Genetics, The Weizmann Institute of Science, Rehovot 76100, Israel
| | - Azad Bonni
- Department of Pathology and
- Program in Neuroscience, Harvard Medical School, Boston, Massachusetts 02115, and
| |
Collapse
|
49
|
Takawira D, Budinger GRS, Hopkinson SB, Jones JCR. A dystroglycan/plectin scaffold mediates mechanical pathway bifurcation in lung epithelial cells. J Biol Chem 2010; 286:6301-10. [PMID: 21149456 DOI: 10.1074/jbc.m110.178988] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023] Open
Abstract
In alveolar epithelial cells (AECs), the membrane-anchored proteoglycan dystroglycan (DG) is a mechanoreceptor that transmits mechanical stretch forces to activate independently the ERK1/2 and the adenosine 5'-monophosphate-activated protein kinase (AMPK) signaling cascades in a process called pathway bifurcation. We tested the hypothesis that the cytoskeleton cross-linker plectin, known to bind both DG and AMPK in muscle cells, acts as a scaffold to regulate DG-mediated mechanical stimulation and pathway bifurcation. We demonstrate that plectin and DG form a complex in AECs and that this complex interacts with ERK1/2 and AMPK. Plectin knockdown reduces DG interaction with AMPK but not with ERK1/2. Despite this, mechanoactivation of both signaling pathways is significantly attenuated in AECs deficient in plectin. Thus, DG has the dual role of mechanical receptor and scaffold for ERK1/2, whereas plectin acts as a scaffold for AMPK signaling but is also required for DG-mediated ERK1/2 activation. We conclude that the DG-plectin complex plays a central role in transmitting mechanical stress from the extracellular matrix to the cytoplasm.
Collapse
Affiliation(s)
- Desire Takawira
- Department of Cell and Molecular Biology, Feinberg School of Medicine, Northwestern University, Chicago, Illinois 60611, USA
| | | | | | | |
Collapse
|
50
|
Bandyopadhyay S, Chiang CY, Srivastava J, Gersten M, White S, Bell R, Kurschner C, Martin CH, Smoot M, Sahasrabudhe S, Barber DL, Chanda SK, Ideker T. A human MAP kinase interactome. Nat Methods 2010; 7:801-5. [PMID: 20936779 PMCID: PMC2967489 DOI: 10.1038/nmeth.1506] [Citation(s) in RCA: 165] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
Mitogen Activated Protein Kinase (MAPK) pathways form the backbone of signal transduction within the mammalian cell. Here, we apply a systematic experimental and computational approach to map 2,269 interactions between human MAPK-related proteins and other cellular machinery and to assemble these data into functional modules. A core network of 641 interactions is supported by multiple lines of evidence including conservation with yeast. Using siRNA knockdowns, we reveal that a significant number of novel interactors can modulate MAPK mediated signaling. We uncover the Na-H exchanger NHE1 as a scaffold for a novel set of MAPKs, link HSP90 chaperones to MAPK pathways, and identify MUC12 as the human analogue to the yeast signaling mucin Msb2. This study makes available a large resource of MAPK interactions along with the accompanying clone libraries. It illustrates a methodology for probing signaling networks based on functional refinement of experimentally-derived protein interaction maps.
Collapse
Affiliation(s)
- Sourav Bandyopadhyay
- Departments of Medicine and Bioengineering, University of California at San Diego, La Jolla, California, USA
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|