1
|
Zhu MM, Dai J, Dai Z, Peng Y, Zhao YY. GCN2 kinase activation mediates pulmonary vascular remodeling and pulmonary arterial hypertension. JCI Insight 2024; 9:e177926. [PMID: 39316438 PMCID: PMC11530134 DOI: 10.1172/jci.insight.177926] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2023] [Accepted: 09/04/2024] [Indexed: 09/26/2024] Open
Abstract
Pulmonary arterial hypertension (PAH) is characterized by progressive increase of pulmonary vascular resistance and remodeling that result in right heart failure. Recessive mutations of EIF2AK4 gene (encoding general control nonderepressible 2 kinase, GCN2) are linked to heritable pulmonary veno-occlusive disease (PVOD) in patients but rarely in patients with PAH. The role of GCN2 kinase activation in the pathogenesis of PAH remains unclear. Here, we show that GCN2 was hyperphosphorylated and activated in pulmonary vascular endothelial cells (ECs) of hypoxic mice, monocrotaline-treated rats, and patients with idiopathic PAH. Unexpectedly, loss of GCN2 kinase activity in Eif2ak4-/- mice with genetic disruption of the kinase domain induced neither PVOD nor pulmonary hypertension (PH) but inhibited hypoxia-induced PH. RNA-sequencing analysis suggested endothelin-1 (Edn1) as a downstream target of GCN2. GCN2 mediated hypoxia-induced Edn1 expression in human lung ECs via HIF-2α. Restored Edn1 expression in ECs of Eif2ak4-/- mice partially reversed the reduced phenotype of hypoxia-induced PH. Furthermore, GCN2 kinase inhibitor A-92 treatment attenuated PAH in monocrotaline-treated rats. These studies demonstrate that GCN2 kinase activation mediates pulmonary vascular remodeling and PAH at least partially through Edn1. Thus, targeting GCN2 kinase activation is a promising therapeutic strategy for treatment of PAH in patients without EIF2AK4 loss-of-function mutations.
Collapse
Affiliation(s)
- Maggie M. Zhu
- Program for Lung and Vascular Biology, Section for Injury Repair and Regeneration Research, Stanley Manne Children’s Research Institute, Ann & Robert H. Lurie Children’s Hospital of Chicago, Chicago, Illinois, USA
- Department of Pediatrics, Division of Critical Care, Northwestern University Feinberg School of Medicine, Chicago, Illinois, USA
| | - Jingbo Dai
- Program for Lung and Vascular Biology, Section for Injury Repair and Regeneration Research, Stanley Manne Children’s Research Institute, Ann & Robert H. Lurie Children’s Hospital of Chicago, Chicago, Illinois, USA
- Department of Pediatrics, Division of Critical Care, Northwestern University Feinberg School of Medicine, Chicago, Illinois, USA
| | - Zhiyu Dai
- Program for Lung and Vascular Biology, Section for Injury Repair and Regeneration Research, Stanley Manne Children’s Research Institute, Ann & Robert H. Lurie Children’s Hospital of Chicago, Chicago, Illinois, USA
- Department of Pediatrics, Division of Critical Care, Northwestern University Feinberg School of Medicine, Chicago, Illinois, USA
| | - Yi Peng
- Program for Lung and Vascular Biology, Section for Injury Repair and Regeneration Research, Stanley Manne Children’s Research Institute, Ann & Robert H. Lurie Children’s Hospital of Chicago, Chicago, Illinois, USA
- Department of Pediatrics, Division of Critical Care, Northwestern University Feinberg School of Medicine, Chicago, Illinois, USA
| | - You-Yang Zhao
- Program for Lung and Vascular Biology, Section for Injury Repair and Regeneration Research, Stanley Manne Children’s Research Institute, Ann & Robert H. Lurie Children’s Hospital of Chicago, Chicago, Illinois, USA
- Department of Pediatrics, Division of Critical Care, Northwestern University Feinberg School of Medicine, Chicago, Illinois, USA
- Genetic Medicine and Nanotechnology Development Center (GeneMeNDer), Stanley Manne Children’s Research Institute, Ann & Robert H. Lurie Children’s Hospital of Chicago, Chicago, Illinois, USA
- Departments of Pharmacology and Medicine and
- Feinberg Cardiovascular and Renal Research Institute, Northwestern University Feinberg School of Medicine, Chicago, Illinois, USA
| |
Collapse
|
2
|
Silva-Aguiar RP, Teixeira DE, Peruchetti DB, Peres RAS, Alves SAS, Calil PT, Arruda LB, Costa LJ, Silva PL, Schmaier AH, Rocco PRM, Pinheiro AAS, Caruso-Neves C. Toll like receptor 4 mediates the inhibitory effect of SARS-CoV-2 spike protein on proximal tubule albumin endocytosis. Biochim Biophys Acta Mol Basis Dis 2024; 1870:167155. [PMID: 38579939 DOI: 10.1016/j.bbadis.2024.167155] [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: 11/14/2023] [Revised: 03/27/2024] [Accepted: 03/29/2024] [Indexed: 04/07/2024]
Abstract
Tubular proteinuria is a common feature in COVID-19 patients, even in the absence of established acute kidney injury. SARS-CoV-2 spike protein (S protein) was shown to inhibit megalin-mediated albumin endocytosis in proximal tubule epithelial cells (PTECs). Angiotensin-converting enzyme type 2 (ACE2) was not directly involved. Since Toll-like receptor 4 (TLR4) mediates S protein effects in various cell types, we hypothesized that TLR4 could be participating in the inhibition of PTECs albumin endocytosis elicited by S protein. Two different models of PTECs were used: porcine proximal tubule cells (LLC-PK1) and human embryonic kidney cells (HEK-293). S protein reduced Akt activity by specifically inhibiting of threonine 308 (Thr308) phosphorylation, a process mediated by phosphoinositide-dependent kinase 1 (PDK1). GSK2334470, a PDK1 inhibitor, decreased albumin endocytosis and megalin expression mimicking S protein effect. S protein did not change total TLR4 expression but decreased its surface expression. LPS-RS, a TLR4 antagonist, also counteracted the effects of the S protein on Akt phosphorylation at Thr308, albumin endocytosis, and megalin expression. Conversely, these effects of the S protein were replicated by LPS, an agonist of TLR4. Incubation of PTECs with a pseudovirus containing S protein inhibited albumin endocytosis. Null or VSV-G pseudovirus, used as control, had no effect. LPS-RS prevented the inhibitory impact of pseudovirus containing the S protein on albumin endocytosis but had no influence on virus internalization. Our findings demonstrate that the inhibitory effect of the S protein on albumin endocytosis in PTECs is mediated through TLR4, resulting from a reduction in megalin expression.
Collapse
Affiliation(s)
- Rodrigo P Silva-Aguiar
- Carlos Chagas Filho Biophysics Institute, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil
| | - Douglas E Teixeira
- Carlos Chagas Filho Biophysics Institute, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil
| | - Diogo B Peruchetti
- Carlos Chagas Filho Biophysics Institute, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil
| | - Rodrigo A S Peres
- Carlos Chagas Filho Biophysics Institute, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil
| | - Sarah A S Alves
- Carlos Chagas Filho Biophysics Institute, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil
| | - Pedro T Calil
- Paulo de Góes Microbiology Institute, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil
| | - Luciana B Arruda
- Paulo de Góes Microbiology Institute, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil
| | - Luciana J Costa
- Paulo de Góes Microbiology Institute, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil
| | - Pedro L Silva
- Carlos Chagas Filho Biophysics Institute, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil; Rio de Janeiro Innovation Network in Nanosystems for Health-NanoSAÚDE/FAPERJ, Rio de Janeiro, Brazil; National Institute of Science and Technology for Regenerative Medicine, Rio de Janeiro, Brazil
| | - Alvin H Schmaier
- Department of Medicine, School of Medicine, Case Western Reserve University, Cleveland, OH, USA; University Hospitals Cleveland Medical Center, Cleveland, OH, USA
| | - Patricia R M Rocco
- Carlos Chagas Filho Biophysics Institute, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil; Rio de Janeiro Innovation Network in Nanosystems for Health-NanoSAÚDE/FAPERJ, Rio de Janeiro, Brazil; National Institute of Science and Technology for Regenerative Medicine, Rio de Janeiro, Brazil
| | - Ana Acacia S Pinheiro
- Carlos Chagas Filho Biophysics Institute, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil; Rio de Janeiro Innovation Network in Nanosystems for Health-NanoSAÚDE/FAPERJ, Rio de Janeiro, Brazil
| | - Celso Caruso-Neves
- Carlos Chagas Filho Biophysics Institute, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil; Rio de Janeiro Innovation Network in Nanosystems for Health-NanoSAÚDE/FAPERJ, Rio de Janeiro, Brazil; National Institute of Science and Technology for Regenerative Medicine, Rio de Janeiro, Brazil.
| |
Collapse
|
3
|
Hu L, Zhao X, He X, Guo Y, Cheng H, Chen S, Zhou G, Wang J, Lu Y. Gynostemma Pentaphyllum ameliorates CCl 4-induced liver injury via PDK1/Bcl-2 pathway with comprehensive analysis of network pharmacology and transcriptomics. Chin Med 2024; 19:70. [PMID: 38750545 PMCID: PMC11094861 DOI: 10.1186/s13020-024-00942-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2024] [Accepted: 05/05/2024] [Indexed: 05/19/2024] Open
Abstract
BACKGROUND Gynostemma pentaphyllum (Thunb.) Makino, commonly known as "southern ginseng", contains high amounts of ginsenoside derivatives and exhibits similar biological activities with Panax ginseng (C. A. MEY) (ginseng), which is usually used as a low-cost alternative to ginseng. G. pentaphyllum has therapeutic effects on liver diseases. However, the mechanisms underlying its hepatoprotective action have not been fully elucidated. METHODS The protective effects of the ethanolic extract of G. pentaphyllum (GPE) were evaluated using an experimental carbon tetrachloride (CCl4)-induced liver disease model. Potential targets of GPE were predicted using the "Drug-Disease" bioinformatic analysis. Furthermore, comprehensive network pharmacology and transcriptomic approaches were employed to investigate the underlying mechanisms of GPE in the treatment of liver disease. RESULTS The pathological examinations showed that GPE significantly alleviated hepatocyte necrosis and liver injury. GPE significantly downregulated Bax and cleaved-PARP expression and upregulated Bcl-2 expression during CCl4-induced hepatocyte apoptosis. We compared the effects of four typical compounds in GPE -a ginsenoside (Rb3) shared by both GPE and ginseng and three unique gypenosides in GPE. Notably, Gypenoside A (GPA), a unique saponin in GPE, markedly reduced hepatocyte apoptosis. In contrast, ginsenoside Rb3 had a weaker effect. Network pharmacology and transcriptomic analyses suggested that this anti-apoptotic effect was achieved by upregulating the PI3K/Akt signaling pathway mediated by PDK1. CONCLUSIONS These results suggested that G. pentaphyllum had a promising hepatoprotective effect, with its mechanism primarily involving the upregulation of the PDK1/Bcl-2 signaling pathway by GPA, thereby preventing cell apoptosis.
Collapse
Affiliation(s)
- Linlan Hu
- College of Pharmacy, Henan University of Chinese Medicine, Zhengzhou, 450046, China
- School of Traditional Chinese Medicine, Capital Medical University, No.10 Outside You'anmen, Fengtai District, Beijing, 100069, China
| | - Xin Zhao
- College of Pharmacy, Henan University of Chinese Medicine, Zhengzhou, 450046, China
- School of Traditional Chinese Medicine, Capital Medical University, No.10 Outside You'anmen, Fengtai District, Beijing, 100069, China
| | - Xian He
- School of Traditional Chinese Medicine, Capital Medical University, No.10 Outside You'anmen, Fengtai District, Beijing, 100069, China
| | - Yafei Guo
- School of Traditional Chinese Medicine, Capital Medical University, No.10 Outside You'anmen, Fengtai District, Beijing, 100069, China
| | - Hanxiao Cheng
- School of Traditional Chinese Medicine, Capital Medical University, No.10 Outside You'anmen, Fengtai District, Beijing, 100069, China
| | - Shaoting Chen
- School of Traditional Chinese Medicine, Capital Medical University, No.10 Outside You'anmen, Fengtai District, Beijing, 100069, China
| | - Guangde Zhou
- Centre for Clinical Pathology, Beijing You'an Hospital, Capital Medical University, No.8 Outside You'anmen, Fengtai District, Beijing, 100069, China.
| | - Jiabo Wang
- College of Pharmacy, Henan University of Chinese Medicine, Zhengzhou, 450046, China.
- School of Traditional Chinese Medicine, Capital Medical University, No.10 Outside You'anmen, Fengtai District, Beijing, 100069, China.
- State Key Laboratory for Quality Ensurance and Sustainable Use of Dao-DiHerbs, Beijing, 100700, China.
| | - Yawen Lu
- School of Traditional Chinese Medicine, Capital Medical University, No.10 Outside You'anmen, Fengtai District, Beijing, 100069, China.
| |
Collapse
|
4
|
Kailasam Natesan V, Kuppannagounder Pitchaimuthu E. Structure-based drug design and molecular dynamics studies of an allosteric modulator targeting the protein-protein interaction site of PDK1. J Mol Model 2024; 30:51. [PMID: 38277080 DOI: 10.1007/s00894-024-05842-2] [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: 07/07/2023] [Accepted: 01/09/2024] [Indexed: 01/27/2024]
Abstract
CONTEXT Protein-protein interaction interfaces play a major role in cell signaling pathways. There is always a great interest in developing protein-protein interaction (PPI) inhibitors of kinases, as they are challenging due to their hydrophobicity, flat surface, specificity, potency, etc. 3 Phosphoinositide-dependent kinase-1 (PDK1), which is involved in the PI3K/PDK1/AKT pathway, is a cancer target that has gained insight for the past two decades. PDK1 possesses a protein interaction fragment (PIF) pocket, which is a well-known PPI that targets allosteric modulators. This work focusses on energy-based pharmacophore model development which on virtual screening could yield novel scaffolds towards the drug designing objective for the kind of PDK1 modulators. A novel pyrazolo pyridine molecule was identified as an allosteric modulator that binds to the PPI site. The metadynamics simulations with free energy profiles further revealed the conformational allosteric changes stimulated on the protein structure upon ligand binding. The cytotoxic activity (IC50-20 μM) of the identified compound against five different cancer cell lines and cell cycle analysis supported the anticancer activity of the identified compound. METHODS All the computational works were carried out by the most commonly used Schrodinger Suite software. The pharmacophore was validated by Receiver Operation Characteristics (ROC) and screening against allosteric Enamine database library. The Optimized Potential Liquid Simulations (OPLS-2005) was used to minimize the structures for molecular docking calculations, and inbuilt scoring method of ranking the compounds based on docking score and Glide energy was used. Molecular dynamics simulations were conducted by Desmond implemented in Maestro. The hit compound was purchased from Enamine and tested against different cancer cell lines by MTT assay, apoptosis by western blotting technique, and by flow cytometry analysis.
Collapse
|
5
|
McColl TJ, Clarke DC. Kinetic modeling of leucine-mediated signaling and protein metabolism in human skeletal muscle. iScience 2024; 27:108634. [PMID: 38188514 PMCID: PMC10767222 DOI: 10.1016/j.isci.2023.108634] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2023] [Revised: 11/15/2023] [Accepted: 12/01/2023] [Indexed: 01/09/2024] Open
Abstract
Skeletal muscle protein levels are governed by the relative rates of muscle protein synthesis (MPS) and breakdown (MPB). The mechanisms controlling these rates are complex, and their integrated behaviors are challenging to study through experiments alone. The purpose of this study was to develop and analyze a kinetic model of leucine-mediated mTOR signaling and protein metabolism in the skeletal muscle of young adults. Our model amalgamates published cellular-level models of the IRS1-PI3K-Akt-mTORC1 signaling system and of skeletal-muscle leucine kinetics with physiological-level models of leucine digestion and transport and insulin dynamics. The model satisfactorily predicts experimental data from diverse leucine feeding protocols. Model analysis revealed that total levels of p70S6K are a primary determinant of MPS, insulin signaling substantially affects muscle net protein balance via its effects on MPB, and p70S6K-mediated feedback of mTORC1 signaling reduces MPS in a dose-dependent manner.
Collapse
Affiliation(s)
- Taylor J. McColl
- Department of Biomedical Physiology and KinesiologySimon Fraser University, Burnaby, BC V5A 1S6, Canada
- Centre for Cell Biology, Development and Disease, Simon Fraser University, Burnaby, BC V5A 1S6, Canada
| | - David C. Clarke
- Department of Biomedical Physiology and KinesiologySimon Fraser University, Burnaby, BC V5A 1S6, Canada
- Centre for Cell Biology, Development and Disease, Simon Fraser University, Burnaby, BC V5A 1S6, Canada
| |
Collapse
|
6
|
Zheng N, Wei J, Wu D, Xu Y, Guo J. Master kinase PDK1 in tumorigenesis. Biochim Biophys Acta Rev Cancer 2023; 1878:188971. [PMID: 37640147 DOI: 10.1016/j.bbcan.2023.188971] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2023] [Revised: 07/13/2023] [Accepted: 08/05/2023] [Indexed: 08/31/2023]
Abstract
3-phosphoinositide-dependent protein kinase 1 (PDK1) is considered as master kinase regulating AGC kinase family members such as AKT, SGK, PLK, S6K and RSK. Although autophosphorylation regulates PDK1 activity, accumulating evidence suggests that PDK1 is manipulated by many other mechanisms, including S6K-mediated phosphorylation, and the E3 ligase SPOP-mediated ubiquitination and degradation. Dysregulation of these upstream regulators or downstream signals involves in cancer development, as PDK1 regulating cell growth, metastasis, invasion, apoptosis and survival time. Meanwhile, overexpression of PDK1 is also exposed in a plethora of cancers, whereas inhibition of PDK1 reduces cell size and inhibits tumor growth and progression. More importantly, PDK1 also modulates the tumor microenvironments and markedly influences tumor immunotherapies. In summary, we comprehensively summarize the downstream signals, upstream regulators, mouse models, inhibitors, tumor microenvironment and clinical treatments for PDK1, and highlight PDK1 as a potential cancer therapeutic target.
Collapse
Affiliation(s)
- Nana Zheng
- National Clinical Research Center for Hematologic Diseases, Jiangsu Institute of Hematology, The First Affiliated Hospital of Soochow University, Institute of Blood and Marrow Transplantation, Collaborative Innovation Center of Hematology, Soochow University, Suzhou 215006, China
| | - Jiaqi Wei
- National Clinical Research Center for Hematologic Diseases, Jiangsu Institute of Hematology, The First Affiliated Hospital of Soochow University, Institute of Blood and Marrow Transplantation, Collaborative Innovation Center of Hematology, Soochow University, Suzhou 215006, China
| | - Depei Wu
- National Clinical Research Center for Hematologic Diseases, Jiangsu Institute of Hematology, The First Affiliated Hospital of Soochow University, Institute of Blood and Marrow Transplantation, Collaborative Innovation Center of Hematology, Soochow University, Suzhou 215006, China.
| | - Yang Xu
- National Clinical Research Center for Hematologic Diseases, Jiangsu Institute of Hematology, The First Affiliated Hospital of Soochow University, Institute of Blood and Marrow Transplantation, Collaborative Innovation Center of Hematology, Soochow University, Suzhou 215006, China.
| | - Jianping Guo
- Institute of Precision Medicine, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong 510275, China.
| |
Collapse
|
7
|
Leroux AE, Biondi RM. The choreography of protein kinase PDK1 and its diverse substrate dance partners. Biochem J 2023; 480:1503-1532. [PMID: 37792325 DOI: 10.1042/bcj20220396] [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: 06/06/2023] [Revised: 08/24/2023] [Accepted: 08/31/2023] [Indexed: 10/05/2023]
Abstract
The protein kinase PDK1 phosphorylates at least 24 distinct substrates, all of which belong to the AGC protein kinase group. Some substrates, such as conventional PKCs, undergo phosphorylation by PDK1 during their synthesis and subsequently get activated by DAG and Calcium. On the other hand, other substrates, including members of the Akt/PKB, S6K, SGK, and RSK families, undergo phosphorylation and activation downstream of PI3-kinase signaling. This review presents two accepted molecular mechanisms that determine the precise and timely phosphorylation of different substrates by PDK1. The first mechanism involves the colocalization of PDK1 with Akt/PKB in the presence of PIP3. The second mechanism involves the regulated docking interaction between the hydrophobic motif (HM) of substrates and the PIF-pocket of PDK1. This interaction, in trans, is equivalent to the molecular mechanism that governs the activity of AGC kinases through their HMs intramolecularly. PDK1 has been instrumental in illustrating the bi-directional allosteric communication between the PIF-pocket and the ATP-binding site and the potential of the system for drug discovery. PDK1's interaction with substrates is not solely regulated by the substrates themselves. Recent research indicates that full-length PDK1 can adopt various conformations based on the positioning of the PH domain relative to the catalytic domain. These distinct conformations of full-length PDK1 can influence the interaction and phosphorylation of substrates. Finally, we critically discuss recent findings proposing that PIP3 can directly regulate the activity of PDK1, which contradicts extensive in vitro and in vivo studies conducted over the years.
Collapse
Affiliation(s)
- Alejandro E Leroux
- Instituto de Investigación en Biomedicina de Buenos Aires (IBioBA) - CONICET - Partner Institute of the Max Planck Society, Buenos Aires C1425FQD, Argentina
| | - Ricardo M Biondi
- Instituto de Investigación en Biomedicina de Buenos Aires (IBioBA) - CONICET - Partner Institute of the Max Planck Society, Buenos Aires C1425FQD, Argentina
| |
Collapse
|
8
|
Sacerdoti M, Gross LZF, Riley AM, Zehnder K, Ghode A, Klinke S, Anand GS, Paris K, Winkel A, Herbrand AK, Godage HY, Cozier GE, Süß E, Schulze JO, Pastor-Flores D, Bollini M, Cappellari MV, Svergun D, Gräwert MA, Aramendia PF, Leroux AE, Potter BVL, Camacho CJ, Biondi RM. Modulation of the substrate specificity of the kinase PDK1 by distinct conformations of the full-length protein. Sci Signal 2023; 16:eadd3184. [PMID: 37311034 DOI: 10.1126/scisignal.add3184] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2022] [Accepted: 05/19/2023] [Indexed: 06/15/2023]
Abstract
The activation of at least 23 different mammalian kinases requires the phosphorylation of their hydrophobic motifs by the kinase PDK1. A linker connects the phosphoinositide-binding PH domain to the catalytic domain, which contains a docking site for substrates called the PIF pocket. Here, we used a chemical biology approach to show that PDK1 existed in equilibrium between at least three distinct conformations with differing substrate specificities. The inositol polyphosphate derivative HYG8 bound to the PH domain and disrupted PDK1 dimerization by stabilizing a monomeric conformation in which the PH domain associated with the catalytic domain and the PIF pocket was accessible. In the absence of lipids, HYG8 potently inhibited the phosphorylation of Akt (also termed PKB) but did not affect the intrinsic activity of PDK1 or the phosphorylation of SGK, which requires docking to the PIF pocket. In contrast, the small-molecule valsartan bound to the PIF pocket and stabilized a second distinct monomeric conformation. Our study reveals dynamic conformations of full-length PDK1 in which the location of the linker and the PH domain relative to the catalytic domain determines the selective phosphorylation of PDK1 substrates. The study further suggests new approaches for the design of drugs to selectively modulate signaling downstream of PDK1.
Collapse
Affiliation(s)
- Mariana Sacerdoti
- Instituto de Investigación en Biomedicina de Buenos Aires (IBioBA)-CONICET-Partner Institute of the Max Planck Society, Buenos Aires C1425FQD, Argentina
| | - Lissy Z F Gross
- Instituto de Investigación en Biomedicina de Buenos Aires (IBioBA)-CONICET-Partner Institute of the Max Planck Society, Buenos Aires C1425FQD, Argentina
| | - Andrew M Riley
- Medicinal Chemistry and Drug Discovery, Department of Pharmacology, University of Oxford, Mansfield Road, Oxford OX1 3QT, UK
| | - Karin Zehnder
- Department of Internal Medicine I, Universitätsklinikum Frankfurt, Theodor-Stern-Kai 7, 60590 Frankfurt, Germany
| | - Abhijeet Ghode
- Biological Sciences, National University of Singapore, Singapore 119077, Singapore
| | - Sebastián Klinke
- Fundación Instituto Leloir, IIBBA-CONICET, and Plataforma Argentina de Biología Estructural y Metabolómica PLABEM, Buenos Aires C1405BWE, Argentina
| | - Ganesh Srinivasan Anand
- Biological Sciences, National University of Singapore, Singapore 119077, Singapore
- Department of Chemistry, Huck Institutes of the Life Sciences, Pennsylvania State University, 104 Chemistry Building, University Park, PA 16802, USA
| | - Kristina Paris
- Department of Computational and Systems Biology, University of Pittsburgh, Pittsburgh, PA 15260, USA
- Department of Statistics, University of Pittsburgh, WWPH 1821, Pittsburgh, PA 15213, USA
| | - Angelika Winkel
- Department of Internal Medicine I, Universitätsklinikum Frankfurt, Theodor-Stern-Kai 7, 60590 Frankfurt, Germany
| | - Amanda K Herbrand
- Department of Internal Medicine I, Universitätsklinikum Frankfurt, Theodor-Stern-Kai 7, 60590 Frankfurt, Germany
| | - H Yasmin Godage
- Wolfson Laboratory of Medicinal Chemistry, Department of Life Sciences, University of Bath, Claverton Down, Bath BA2 7AY, UK
| | - Gyles E Cozier
- Wolfson Laboratory of Medicinal Chemistry, Department of Life Sciences, University of Bath, Claverton Down, Bath BA2 7AY, UK
| | - Evelyn Süß
- Department of Internal Medicine I, Universitätsklinikum Frankfurt, Theodor-Stern-Kai 7, 60590 Frankfurt, Germany
| | - Jörg O Schulze
- Department of Internal Medicine I, Universitätsklinikum Frankfurt, Theodor-Stern-Kai 7, 60590 Frankfurt, Germany
| | - Daniel Pastor-Flores
- Department of Internal Medicine I, Universitätsklinikum Frankfurt, Theodor-Stern-Kai 7, 60590 Frankfurt, Germany
- KBI Biopharma, Technologielaan 8, B-3001 Leuven, Belgium
| | - Mariela Bollini
- Centro de Investigaciones en Bionanociencias 'Elizabeth Jares-Erijman' CIBION, CONICET, Buenos Aires C1425FQD, Argentina
| | - María Victoria Cappellari
- Centro de Investigaciones en Bionanociencias 'Elizabeth Jares-Erijman' CIBION, CONICET, Buenos Aires C1425FQD, Argentina
| | - Dmitri Svergun
- European Molecular Biology Laboratory (EMBL), Hamburg Unit, 22607 Hamburg, Germany
| | - Melissa A Gräwert
- European Molecular Biology Laboratory (EMBL), Hamburg Unit, 22607 Hamburg, Germany
| | - Pedro F Aramendia
- Centro de Investigaciones en Bionanociencias 'Elizabeth Jares-Erijman' CIBION, CONICET, Buenos Aires C1425FQD, Argentina
- Departamento de Química Inorgánica, Analítica y Química Física, FCEN, Universidad de Buenos Aires, Buenos Aires C1428EHA, Argentina
| | - Alejandro E Leroux
- Instituto de Investigación en Biomedicina de Buenos Aires (IBioBA)-CONICET-Partner Institute of the Max Planck Society, Buenos Aires C1425FQD, Argentina
| | - Barry V L Potter
- Medicinal Chemistry and Drug Discovery, Department of Pharmacology, University of Oxford, Mansfield Road, Oxford OX1 3QT, UK
- Wolfson Laboratory of Medicinal Chemistry, Department of Life Sciences, University of Bath, Claverton Down, Bath BA2 7AY, UK
| | - Carlos J Camacho
- Department of Computational and Systems Biology, University of Pittsburgh, Pittsburgh, PA 15260, USA
| | - Ricardo M Biondi
- Instituto de Investigación en Biomedicina de Buenos Aires (IBioBA)-CONICET-Partner Institute of the Max Planck Society, Buenos Aires C1425FQD, Argentina
- Department of Internal Medicine I, Universitätsklinikum Frankfurt, Theodor-Stern-Kai 7, 60590 Frankfurt, Germany
- DKTK German Cancer Consortium (DKTK), Frankfurt, Germany
- German Cancer Research Center (DKFZ), D-69120 Heidelberg, Germany
| |
Collapse
|
9
|
Zhang P, Jiang Y, Ye X, Zhang C, Tang Y. PDK1 inhibition reduces autophagy and cell senescence through the PI3K/AKT signalling pathway in a cigarette smoke mouse emphysema model. Exp Ther Med 2023; 25:223. [PMID: 37123206 PMCID: PMC10133799 DOI: 10.3892/etm.2023.11922] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2022] [Accepted: 11/03/2022] [Indexed: 04/03/2023] Open
Abstract
A number of previous studies have demonstrated the pivotal role of PI3K/AKT signalling in cigarette smoke (CS)-induced emphysema, where phosphoinositide dependent protein kinase 1 (PDK1) is a critical component of this pathway. Therefore, the present study aimed to investigate the effects of a PDK1 inhibitor (GSK-2334470) on the expression levels of PI3K, AKT, cyclin-dependent kinase inhibitor 2A (p16) and LC3B in a CS + CS extract (CSE)-induced mouse emphysema model. CS exposure and intraperitoneal injections of CSE were combined for 4 weeks to establish an emphysema model. Mice (n=35) were randomly divided into the normal control, emphysema (CS), PI3K inhibitor (CS3) and PDK1 inhibitor (CS1) groups. Immunohistochemistry staining of lung tissues was used to measure the expression of the PI3K, PDK1 and AKT proteins in airway epithelial tissues. Immunofluorescence staining was also used to measure the levels of p16 and LC3BII protein expression in the airway epithelial tissues. In addition, PI3K, PDK1, AKT, p16 and LC3B protein expression was semi-quantified using western blotting. The expression of PDK1, PI3K and AKT proteins in the airway epithelial tissues was significantly increased in the CS + CSE group compared with that in the control group. The expression levels of p16 and LC3B were also increased as well in the CS + CSE group compared with those in the control group. The expression levels of PI3K, PDK1, AKT, LC3B and p16 in the airway epithelial tissues of the CS3 group were lower compared with those in the CS + CSE group. A decrease in the expression levels of PDK1, AKT, p16 and LC3B in the airway epithelial tissues of the CS1 group compared with those in the CS + CSE group was also observed. However, there were no significant differences in the expression levels of PI3K between the CS1 and the CS groups. The present study concluded that the inhibition of PDK1 can potentially reduce autophagy and cell senescence by downregulating the expression of PI3K/AKT pathway related proteins in airway epithelial cells, thereby protecting against CS + CSE-induced emphysema in mice.
Collapse
Affiliation(s)
- Peibei Zhang
- Department of Respiratory Medicine, Guizhou Provincial People's Hospital, Guiyang, Guizhou 550002, P.R. China
| | - Youjun Jiang
- Department of Respiratory Medicine, Guizhou Provincial People's Hospital, Guiyang, Guizhou 550002, P.R. China
| | - Xianwei Ye
- Department of Respiratory Medicine, Guizhou Provincial People's Hospital, Guiyang, Guizhou 550002, P.R. China
| | - Cheng Zhang
- Department of Respiratory Medicine, Guizhou Provincial People's Hospital, Guiyang, Guizhou 550002, P.R. China
| | - Yiling Tang
- Department of Respiratory Medicine, Guizhou Provincial People's Hospital, Guiyang, Guizhou 550002, P.R. China
| |
Collapse
|
10
|
Cai W, Nguyen MQ, Wilski NA, Purwin TJ, Vernon M, Tiago M, Aplin AE. A Genome-Wide Screen Identifies PDPK1 as a Target to Enhance the Efficacy of MEK1/2 Inhibitors in NRAS Mutant Melanoma. Cancer Res 2022; 82:2625-2639. [PMID: 35657206 PMCID: PMC9298960 DOI: 10.1158/0008-5472.can-21-3217] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2021] [Revised: 04/22/2022] [Accepted: 05/31/2022] [Indexed: 01/21/2023]
Abstract
Melanomas frequently harbor activating NRAS mutations. However, limited advance has been made in developing targeted therapy options for patients with NRAS mutant melanoma. MEK inhibitors (MEKi) show modest efficacy in the clinic and their actions need to be optimized. In this study, we performed a genome-wide CRISPR-Cas9-based screen and demonstrated that loss of phosphoinositide-dependent kinase-1 (PDPK1) enhances the efficacy of MEKi. The synergistic effects of PDPK1 loss and MEKi was validated in NRAS mutant melanoma cell lines using pharmacologic and molecular approaches. Combined PDPK1 inhibitors (PDPK1i) with MEKi suppressed NRAS mutant xenograft growth and induced gasdermin E-associated pyroptosis. In an immune-competent allograft model, PDPK1i+MEKi increased the ratio of intratumoral CD8+ T cells, delayed tumor growth, and prolonged survival; the combination treatment was less effective against tumors in immune-deficient mice. These data suggest PDPK1i+MEKi as an efficient immunostimulatory strategy against NRAS mutant melanoma. SIGNIFICANCE Targeting PDPK1 stimulates antitumor immunity and sensitizes NRAS mutant melanoma to MEK inhibition, providing rationale for the clinical development of a combinatorial approach for treating patients with melanoma.
Collapse
Affiliation(s)
- Weijia Cai
- Department of Cancer Biology, Thomas Jefferson University, Philadelphia, PA 19107
| | - Mai Q. Nguyen
- Department of Cancer Biology, Thomas Jefferson University, Philadelphia, PA 19107
| | - Nicole A. Wilski
- Department of Cancer Biology, Thomas Jefferson University, Philadelphia, PA 19107
| | - Timothy J. Purwin
- Department of Cancer Biology, Thomas Jefferson University, Philadelphia, PA 19107
| | - Megane Vernon
- Department of Cancer Biology, Thomas Jefferson University, Philadelphia, PA 19107
| | - Manoela Tiago
- Department of Cancer Biology, Thomas Jefferson University, Philadelphia, PA 19107
| | - Andrew E. Aplin
- Department of Cancer Biology, Thomas Jefferson University, Philadelphia, PA 19107
- Sidney Kimmel Cancer Center, Thomas Jefferson University, Philadelphia, PA 19107
| |
Collapse
|
11
|
The Landscape of PDK1 in Breast Cancer. Cancers (Basel) 2022; 14:cancers14030811. [PMID: 35159078 PMCID: PMC8834120 DOI: 10.3390/cancers14030811] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2021] [Accepted: 12/31/2021] [Indexed: 02/04/2023] Open
Abstract
Given that 3-phosphoinositide-dependent kinase 1 (PDK1) plays a crucial role in the malignant biological behaviors of a wide range of cancers, we review the influence of PDK1 in breast cancer (BC). First, we describe the power of PDK1 in cellular behaviors and characterize the interaction networks of PDK1. Then, we establish the roles of PDK1 in carcinogenesis, growth and survival, metastasis, and chemoresistance in BC cells. More importantly, we sort the current preclinical or clinical trials of PDK1-targeted therapy in BC and find that, even though no selective PDK1 inhibitor is currently available for BC therapy, the combination trials of PDK1-targeted therapy and other agents have provided some benefit. Thus, there is increasing anticipation that PDK1-targeted therapy will have its space in future therapeutic approaches related to BC, and we hope the novel approaches of targeted therapy will be conducive to ameliorating the dismal prognosis of BC patients.
Collapse
|
12
|
Hitz E, Wiedemar N, Passecker A, Graça BAS, Scheurer C, Wittlin S, Brancucci NMB, Vakonakis I, Mäser P, Voss TS. The 3-phosphoinositide-dependent protein kinase 1 is an essential upstream activator of protein kinase A in malaria parasites. PLoS Biol 2021; 19:e3001483. [PMID: 34879056 PMCID: PMC8687544 DOI: 10.1371/journal.pbio.3001483] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2021] [Revised: 12/20/2021] [Accepted: 11/12/2021] [Indexed: 01/11/2023] Open
Abstract
Cyclic adenosine monophosphate (cAMP)-dependent protein kinase A (PKA) signalling is essential for the proliferation of Plasmodium falciparum malaria blood stage parasites. The mechanisms regulating the activity of the catalytic subunit PfPKAc, however, are only partially understood, and PfPKAc function has not been investigated in gametocytes, the sexual blood stage forms that are essential for malaria transmission. By studying a conditional PfPKAc knockdown (cKD) mutant, we confirm the essential role for PfPKAc in erythrocyte invasion by merozoites and show that PfPKAc is involved in regulating gametocyte deformability. We furthermore demonstrate that overexpression of PfPKAc is lethal and kills parasites at the early phase of schizogony. Strikingly, whole genome sequencing (WGS) of parasite mutants selected to tolerate increased PfPKAc expression levels identified missense mutations exclusively in the gene encoding the parasite orthologue of 3-phosphoinositide-dependent protein kinase-1 (PfPDK1). Using targeted mutagenesis, we demonstrate that PfPDK1 is required to activate PfPKAc and that T189 in the PfPKAc activation loop is the crucial target residue in this process. In summary, our results corroborate the importance of tight regulation of PfPKA signalling for parasite survival and imply that PfPDK1 acts as a crucial upstream regulator in this pathway and potential new drug target.
Collapse
Affiliation(s)
- Eva Hitz
- Department of Medical Parasitology and Infection Biology, Swiss Tropical and Public Health Institute, Basel, Switzerland
- University of Basel, Basel, Switzerland
| | - Natalie Wiedemar
- Department of Medical Parasitology and Infection Biology, Swiss Tropical and Public Health Institute, Basel, Switzerland
- University of Basel, Basel, Switzerland
| | - Armin Passecker
- Department of Medical Parasitology and Infection Biology, Swiss Tropical and Public Health Institute, Basel, Switzerland
- University of Basel, Basel, Switzerland
| | - Beatriz A. S. Graça
- Department of Medical Parasitology and Infection Biology, Swiss Tropical and Public Health Institute, Basel, Switzerland
- University of Basel, Basel, Switzerland
| | - Christian Scheurer
- Department of Medical Parasitology and Infection Biology, Swiss Tropical and Public Health Institute, Basel, Switzerland
- University of Basel, Basel, Switzerland
| | - Sergio Wittlin
- Department of Medical Parasitology and Infection Biology, Swiss Tropical and Public Health Institute, Basel, Switzerland
- University of Basel, Basel, Switzerland
| | - Nicolas M. B. Brancucci
- Department of Medical Parasitology and Infection Biology, Swiss Tropical and Public Health Institute, Basel, Switzerland
- University of Basel, Basel, Switzerland
| | - Ioannis Vakonakis
- Department of Biochemistry, University of Oxford, Oxford, United Kingdom
| | - Pascal Mäser
- Department of Medical Parasitology and Infection Biology, Swiss Tropical and Public Health Institute, Basel, Switzerland
- University of Basel, Basel, Switzerland
| | - Till S. Voss
- Department of Medical Parasitology and Infection Biology, Swiss Tropical and Public Health Institute, Basel, Switzerland
- University of Basel, Basel, Switzerland
| |
Collapse
|
13
|
Identification of mutations that cooperate with defects in B cell transcription factors to initiate leukemia. Oncogene 2021; 40:6166-6179. [PMID: 34535769 PMCID: PMC8556320 DOI: 10.1038/s41388-021-02012-z] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2020] [Revised: 08/25/2021] [Accepted: 09/07/2021] [Indexed: 12/22/2022]
Abstract
The transcription factors PAX5, IKZF1, and EBF1 are frequently mutated in B cell acute lymphoblastic leukemia (B-ALL). We demonstrate that compound heterozygous loss of multiple genes critical for B and T cell development drives transformation, including Pax5+/-xEbf1+/-, Pax5+/-xIkzf1+/-, and Ebf1+/-xIkzf1+/- mice for B-ALL, or Tcf7+/-xIkzf1+/- mice for T-ALL. To identify genetic defects that cooperate with Pax5 and Ebf1 compound heterozygosity to initiate leukemia, we performed a Sleeping Beauty (SB) transposon screen that identified cooperating partners including gain-of-function mutations in Stat5b (~65%) and Jak1 (~68%), or loss-of-function mutations in Cblb (61%) and Myb (32%). These findings underscore the role of JAK/STAT5B signaling in B cell transformation and demonstrate roles for loss-of-function mutations in Cblb and Myb in transformation. RNA-Seq studies demonstrated upregulation of a PDK1>SGK3>MYC pathway; treatment of Pax5+/-xEbf1+/- leukemia cells with PDK1 inhibitors blocked proliferation in vitro. In addition, we identified a conserved transcriptional gene signature between human and murine leukemias characterized by upregulation of myeloid genes, most notably involving the GM-CSF pathway, that resemble a B cell/myeloid mixed-lineage leukemia. Thus, our findings identify multiple mechanisms that cooperate with defects in B cell transcription factors to generate either progenitor B cell or mixed B/myeloid-like leukemias.
Collapse
|
14
|
Cilleros-Mañé V, Just-Borràs L, Polishchuk A, Durán M, Tomàs M, Garcia N, Tomàs JM, Lanuza MA. M 1 and M 2 mAChRs activate PDK1 and regulate PKC βI and ε and the exocytotic apparatus at the NMJ. FASEB J 2021; 35:e21724. [PMID: 34133802 DOI: 10.1096/fj.202002213r] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2020] [Revised: 05/07/2021] [Accepted: 05/24/2021] [Indexed: 01/14/2023]
Abstract
Neuromuscular junctions (NMJ) regulate cholinergic exocytosis through the M1 and M2 muscarinic acetylcholine autoreceptors (mAChR), involving the crosstalk between receptors and downstream pathways. Protein kinase C (PKC) regulates neurotransmission but how it associates with the mAChRs remains unknown. Here, we investigate whether mAChRs recruit the classical PKCβI and the novel PKCε isoforms and modulate their priming by PDK1, translocation and activity on neurosecretion targets. We show that each M1 and M2 mAChR activates the master kinase PDK1 and promotes a particular priming of the presynaptic PKCβI and ε isoforms. M1 recruits both primed-PKCs to the membrane and promotes Munc18-1, SNAP-25, and MARCKS phosphorylation. In contrast, M2 downregulates PKCε through a PKA-dependent pathway, which inhibits Munc18-1 synthesis and PKC phosphorylation. In summary, our results discover a co-dependent balance between muscarinic autoreceptors which orchestrates the presynaptic PKC and their action on ACh release SNARE-SM mechanism. Altogether, this molecular signaling explains previous functional studies at the NMJ and guide toward potential therapeutic targets.
Collapse
Affiliation(s)
- V Cilleros-Mañé
- Unitat d'Histologia i Neurobiologia (UHNEUROB), Facultat de Medicina i Ciències de la Salut, Departament de Ciències Mèdiques Bàsiques, Universitat Rovira i Virgili, Reus, Spain
| | - L Just-Borràs
- Unitat d'Histologia i Neurobiologia (UHNEUROB), Facultat de Medicina i Ciències de la Salut, Departament de Ciències Mèdiques Bàsiques, Universitat Rovira i Virgili, Reus, Spain
| | - A Polishchuk
- Unitat d'Histologia i Neurobiologia (UHNEUROB), Facultat de Medicina i Ciències de la Salut, Departament de Ciències Mèdiques Bàsiques, Universitat Rovira i Virgili, Reus, Spain
| | - M Durán
- Unitat d'Histologia i Neurobiologia (UHNEUROB), Facultat de Medicina i Ciències de la Salut, Departament de Ciències Mèdiques Bàsiques, Universitat Rovira i Virgili, Reus, Spain
| | - M Tomàs
- Unitat d'Histologia i Neurobiologia (UHNEUROB), Facultat de Medicina i Ciències de la Salut, Departament de Ciències Mèdiques Bàsiques, Universitat Rovira i Virgili, Reus, Spain
| | - N Garcia
- Unitat d'Histologia i Neurobiologia (UHNEUROB), Facultat de Medicina i Ciències de la Salut, Departament de Ciències Mèdiques Bàsiques, Universitat Rovira i Virgili, Reus, Spain
| | - J M Tomàs
- Unitat d'Histologia i Neurobiologia (UHNEUROB), Facultat de Medicina i Ciències de la Salut, Departament de Ciències Mèdiques Bàsiques, Universitat Rovira i Virgili, Reus, Spain
| | - M A Lanuza
- Unitat d'Histologia i Neurobiologia (UHNEUROB), Facultat de Medicina i Ciències de la Salut, Departament de Ciències Mèdiques Bàsiques, Universitat Rovira i Virgili, Reus, Spain
| |
Collapse
|
15
|
Liang X, Wang R, Guo Y, Cheng Z, Lv D, Luo M, He A, Luo S, Xia Y. Phosphorylation of Akt at Thr308 regulates p-eNOS Ser1177 during physiological conditions. FEBS Open Bio 2021; 11:1953-1964. [PMID: 33993653 PMCID: PMC8255840 DOI: 10.1002/2211-5463.13194] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2020] [Revised: 04/28/2021] [Accepted: 05/12/2021] [Indexed: 11/30/2022] Open
Abstract
Endothelial nitric oxide synthase (eNOS)-derived nitric oxide (NO) plays a crucial role in maintaining vascular homeostasis. As a hallmark of eNOS activation, phosphorylation of eNOS at Ser1177 induced by activated protein kinase B (PKB/Akt) is pivotal for NO production. The complete activation of Akt requires its phosphorylation of both Thr308 and Ser473. However, which site plays the main role in regulating phosphorylation of eNOS Ser1177 is still controversial. The purpose of the present study is to explore the specific regulatory mechanism of phosphorylated Akt in eNOS activation. Inhibition of Akt Thr308 phosphorylation by a specific inhibitor or by siRNA in vitro led to a decrease in eNOS phosphorylation at Ser1177 and to lower NO concentration in the cell culture medium of HUVECs. However, inhibiting p-Akt Ser473 had no effect on eNOS phosphorylation at Ser1177. Next, we administered mice with inhibitors to downregulate p-Akt Ser473 or Thr308 activity. Along with the inhibition of p-Akt Thr308, vascular p-eNOS Ser1177 protein was simultaneously downregulated in parallel with a decrease in plasma NO concentration. Additionally, we cultured HUVECs at various temperature conditions (37, 22, and 4 °C). The results showed that p-Akt Ser473 was gradually decreased in line with the reduction in temperature, accompanied by increased levels of p-Akt Thr308 and p-eNOS Ser1177. Taken together, our study indicates that the phosphorylation of Akt at Thr308, but not at Ser473, plays a more significant role in regulating p-eNOS Ser1177 levels under physiological conditions.
Collapse
Affiliation(s)
- Xiao‐xue Liang
- Division of CardiologyThe First Affiliated Hospital of Chongqing Medical UniversityChina
- Institute of Life ScienceChongqing Medical UniversityChina
| | - Rui‐yu Wang
- Division of CardiologyThe First Affiliated Hospital of Chongqing Medical UniversityChina
- Institute of Life ScienceChongqing Medical UniversityChina
| | - Yong‐zheng Guo
- Division of CardiologyThe First Affiliated Hospital of Chongqing Medical UniversityChina
- Institute of Life ScienceChongqing Medical UniversityChina
| | - Zhe Cheng
- Division of CardiologyThe First Affiliated Hospital of Chongqing Medical UniversityChina
- Institute of Life ScienceChongqing Medical UniversityChina
| | - Ding‐yi Lv
- Division of CardiologyThe First Affiliated Hospital of Chongqing Medical UniversityChina
- Institute of Life ScienceChongqing Medical UniversityChina
| | - Ming‐hao Luo
- Division of CardiologyThe First Affiliated Hospital of Chongqing Medical UniversityChina
- Institute of Life ScienceChongqing Medical UniversityChina
| | - An He
- Division of CardiologyThe First Affiliated Hospital of Chongqing Medical UniversityChina
- Institute of Life ScienceChongqing Medical UniversityChina
| | - Su‐xin Luo
- Division of CardiologyThe First Affiliated Hospital of Chongqing Medical UniversityChina
| | - Yong Xia
- Division of CardiologyThe First Affiliated Hospital of Chongqing Medical UniversityChina
- Institute of Life ScienceChongqing Medical UniversityChina
| |
Collapse
|
16
|
Zhang X, Zhong S. PDK1 Inhibitor GSK-470 Exhibits Potent Anticancer Activity in a Pheochromocytoma PC12 Cell Tumor Model via Akt/mTOR Pathway. Anticancer Agents Med Chem 2021; 20:828-833. [PMID: 32188393 DOI: 10.2174/1871520620666200318100701] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2019] [Revised: 02/09/2020] [Accepted: 02/20/2020] [Indexed: 12/28/2022]
Abstract
BACKGROUND Phosphoinositide-Dependent Kinase 1 (PDK1) is now widely studied in malignant solid tumors. Researchers have previously revealed that targeting PDK1 is thought of as a promising anticancer treatment strategy. The aim of this study was designed to evaluate the anticancer activity of GSK-470, a novel and highly specific inhibitor of PDK1, in Pheochromocytoma (PCC) tumor model. METHODS PC12 cells were xenografted into nude mice to build PCC tumor model. Animals were treated with GSK-470 vs vehicle. Mean tumor volume was calculated and compared across groups. TUNEL was used to detect apoptosis. The effects of PDK1 inhibitor GSK-470 on activation of the Akt signaling and its downstream Akt/mTOR pathway in xenotransplant tumor tissues were examined by western bolt. RESULTS The mean tumor volume in GSK-470 group was significantly less than that in control group. TUNEL results found that cell apoptosis was markedly increased in GSK-470 group compared with the control group. The western bolt analysis showed that the phosphorylation of Akt at threonine 308 was significantly reduced in GSK-470 group. Also, GSK-470 strongly inhibited phosphorylation of mTOR on Ser2448, a marker for mTORC1 activity, as well as phosphorylation of p70S6K, best characterized targets of mTOR. CONCLUSION Our results showed that GSK-470 exhibited potent anticancer activity in PC12 tumor-bearing mice. Also, we found that this effect appeared to be mediated by the inhibition of the Akt/mTOR pathway. The present study once again provides new insights into the therapeutic effects of inhibiting PDK1 in the treatment of malignant PCC. Therefore, we propose that GSK-470 might be an effective therapeutic agent for the treatment of malignant PCC.
Collapse
Affiliation(s)
- Xiaohua Zhang
- Department of Urology, Huashan Hospital, Fudan University, Shanghai, China
| | - Shan Zhong
- Department of Urology, Huashan Hospital, Fudan University, Shanghai, China
| |
Collapse
|
17
|
Kim H, Lee J, Cho Y. PDK1 is a negative regulator of axon regeneration. Mol Brain 2021; 14:31. [PMID: 33579325 PMCID: PMC7881570 DOI: 10.1186/s13041-021-00748-z] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2020] [Accepted: 02/04/2021] [Indexed: 12/11/2022] Open
Abstract
Axon regeneration in the central nervous system is inefficient. However, the neurons in the peripheral nervous system display robust regeneration after injury, indicating that axonal regeneration is differentially controlled under various conditions. To identify those molecules regulating axon regeneration, comparative analysis from dorsal root ganglion neurons at embryonic or adult stages is utilized, which reveals that PDK1 is functions as a negative regulator of axon regeneration. PDK1 is downregulated in embryonic neurons after axotomy. In contrast, sciatic nerve axotomy upregulated PDK1 at protein levels from adult mice. The knockdown of PDK1 or the chemical inhibition of PDK1 promotes axon regeneration in vitro and in vivo. Here we present PDK1 as a new player to negatively regulate axon regeneration and as a potential target in the development of therapeutic applications.
Collapse
Affiliation(s)
- Hyemin Kim
- Department of Life Sciences, Korea University, Anam-ro 145, Seongbuk-gu, Seoul, 02841, Republic of Korea
| | - Jinyoung Lee
- Department of Life Sciences, Korea University, Anam-ro 145, Seongbuk-gu, Seoul, 02841, Republic of Korea
| | - Yongcheol Cho
- Department of Life Sciences, Korea University, Anam-ro 145, Seongbuk-gu, Seoul, 02841, Republic of Korea.
| |
Collapse
|
18
|
Guarnaccia AD, Rose KL, Wang J, Zhao B, Popay TM, Wang CE, Guerrazzi K, Hill S, Woodley CM, Hansen TJ, Lorey SL, Shaw JG, Payne WG, Weissmiller AM, Olejniczak ET, Fesik SW, Liu Q, Tansey WP. Impact of WIN site inhibitor on the WDR5 interactome. Cell Rep 2021; 34:108636. [PMID: 33472061 PMCID: PMC7871196 DOI: 10.1016/j.celrep.2020.108636] [Citation(s) in RCA: 35] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2020] [Revised: 11/17/2020] [Accepted: 12/21/2020] [Indexed: 01/08/2023] Open
Abstract
The chromatin-associated protein WDR5 is a promising pharmacological target in cancer, with most drug discovery efforts directed against an arginine-binding cavity in WDR5 called the WIN site. Despite a clear expectation that WIN site inhibitors will alter the repertoire of WDR5 interaction partners, their impact on the WDR5 interactome remains unknown. Here, we use quantitative proteomics to delineate how the WDR5 interactome is changed by WIN site inhibition. We show that the WIN site inhibitor alters the interaction of WDR5 with dozens of proteins, including those linked to phosphatidylinositol 3-kinase (PI3K) signaling. As proof of concept, we demonstrate that the master kinase PDPK1 is a bona fide high-affinity WIN site binding protein that engages WDR5 to modulate transcription of genes expressed in the G2 phase of the cell cycle. This dataset expands our understanding of WDR5 and serves as a resource for deciphering the action of WIN site inhibitors.
Collapse
Affiliation(s)
- Alissa D Guarnaccia
- Department of Cell and Developmental Biology, Vanderbilt University School of Medicine, Nashville, TN 37232, USA
| | - Kristie L Rose
- Department of Biochemistry, Vanderbilt University School of Medicine, Nashville, TN 37232, USA; Mass Spectrometry Research Center, Vanderbilt University School of Medicine, Nashville, TN 37232, USA
| | - Jing Wang
- Department of Biostatistics, Vanderbilt University Medical Center, Nashville, TN 37232, USA; Center for Quantitative Sciences, Vanderbilt University Medical Center, Nashville, TN, 37232, USA
| | - Bin Zhao
- Department of Biochemistry, Vanderbilt University School of Medicine, Nashville, TN 37232, USA
| | - Tessa M Popay
- Department of Cell and Developmental Biology, Vanderbilt University School of Medicine, Nashville, TN 37232, USA
| | - Christina E Wang
- Department of Cell and Developmental Biology, Vanderbilt University School of Medicine, Nashville, TN 37232, USA
| | - Kiana Guerrazzi
- Department of Cell and Developmental Biology, Vanderbilt University School of Medicine, Nashville, TN 37232, USA
| | - Salisha Hill
- Mass Spectrometry Research Center, Vanderbilt University School of Medicine, Nashville, TN 37232, USA
| | - Chase M Woodley
- Department of Cell and Developmental Biology, Vanderbilt University School of Medicine, Nashville, TN 37232, USA
| | - Tyler J Hansen
- Department of Biochemistry, Vanderbilt University School of Medicine, Nashville, TN 37232, USA
| | - Shelly L Lorey
- Department of Cell and Developmental Biology, Vanderbilt University School of Medicine, Nashville, TN 37232, USA
| | - J Grace Shaw
- Department of Biochemistry, Vanderbilt University School of Medicine, Nashville, TN 37232, USA
| | - William G Payne
- Department of Biochemistry, Vanderbilt University School of Medicine, Nashville, TN 37232, USA
| | - April M Weissmiller
- Department of Cell and Developmental Biology, Vanderbilt University School of Medicine, Nashville, TN 37232, USA
| | - Edward T Olejniczak
- Department of Biochemistry, Vanderbilt University School of Medicine, Nashville, TN 37232, USA
| | - Stephen W Fesik
- Department of Biochemistry, Vanderbilt University School of Medicine, Nashville, TN 37232, USA; Department of Pharmacology, Vanderbilt University School of Medicine, Nashville, TN 37232, USA; Department of Chemistry, Vanderbilt University, Nashville, TN 37232, USA
| | - Qi Liu
- Department of Biostatistics, Vanderbilt University Medical Center, Nashville, TN 37232, USA; Center for Quantitative Sciences, Vanderbilt University Medical Center, Nashville, TN, 37232, USA
| | - William P Tansey
- Department of Cell and Developmental Biology, Vanderbilt University School of Medicine, Nashville, TN 37232, USA; Department of Biochemistry, Vanderbilt University School of Medicine, Nashville, TN 37232, USA.
| |
Collapse
|
19
|
Wang H, Liu M, Zou G, Wang L, Duan W, He X, Ji M, Zou X, Hu Y, Yang J, Chen G. Deletion of PDK1 in oligodendrocyte lineage cells causes white matter abnormality and myelination defect in the central nervous system. Neurobiol Dis 2021; 148:105212. [PMID: 33276084 DOI: 10.1016/j.nbd.2020.105212] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2020] [Revised: 11/17/2020] [Accepted: 11/29/2020] [Indexed: 12/01/2022] Open
Abstract
PDK1 (3-Phosphoinositide dependent protein kinase-1) is a member in the PI3K (phosphatidylinositol 3 kinase) pathway and is implicated in neurodevelopmental disease with microcephaly. Although the role of PDK1 in neurogenesis has been broadly studied, it remains unknown how PDK1 may regulate oligogenesis in the central nervous system (CNS). To address this question, we generated oligodendrocyte (OL) lineage cells specific PDK1 conditional knockout (cKO) mice. We find that PDK1 cKOs display abnormal white matter (WM), massive loss of mature OLs and severe defect in myelination in the CNS. In contrast, these mutants exhibit normal neuronal development and unchanged apoptosis in the CNS. We demonstrate that deletion of PDK1 severely impairs OL differentiation. We show that genetic or pharmacological inhibition of PDK1 causes deficit in the mammalian target of rapamycin (mTor) signaling and down-regulation of Sox10. Together, these results highlight a critical role of PDK1 in OL differentiation during postnatal CNS development.
Collapse
Affiliation(s)
- He Wang
- Department of Anesthesiology, Pain and Perioperative Medicine, The First Affiliated Hospital of Zhengzhou University, No. 1 East Jianshe Road, Zhengzhou 450052, China
| | - Mengjia Liu
- State Key Laboratory of Pharmaceutical Biotechnology, MOE Key Laboratory of Model Animal for Disease Study, Model Animal Research Center, Nanjing University, 12 Xuefu Avenue, Nanjing, Jiangsu Province 210061, China
| | - Gang Zou
- Department of Anesthesiology, The Second Affiliated Hospital, Nanjing Medical University, 121 Jiangjiayuan Avenue, Nanjing, Jiangsu Province 210003, China
| | - Long Wang
- State Key Laboratory of Pharmaceutical Biotechnology, MOE Key Laboratory of Model Animal for Disease Study, Model Animal Research Center, Nanjing University, 12 Xuefu Avenue, Nanjing, Jiangsu Province 210061, China
| | - Wenbin Duan
- Department of General Surgery, The Second Clinical Medical College, Shenzhen People's Hospital, Jinan University, Shenzhen 518000, China
| | - Xue He
- Department of Anesthesiology, Pain and Perioperative Medicine, The First Affiliated Hospital of Zhengzhou University, No. 1 East Jianshe Road, Zhengzhou 450052, China
| | - Muhuo Ji
- Department of Anesthesiology, Pain and Perioperative Medicine, The First Affiliated Hospital of Zhengzhou University, No. 1 East Jianshe Road, Zhengzhou 450052, China
| | - Xiaochuan Zou
- State Key Laboratory of Pharmaceutical Biotechnology, MOE Key Laboratory of Model Animal for Disease Study, Model Animal Research Center, Nanjing University, 12 Xuefu Avenue, Nanjing, Jiangsu Province 210061, China
| | - Yimin Hu
- Department of General Surgery, The Second Clinical Medical College, Shenzhen People's Hospital, Jinan University, Shenzhen 518000, China.
| | - Jianjun Yang
- Department of Anesthesiology, Pain and Perioperative Medicine, The First Affiliated Hospital of Zhengzhou University, No. 1 East Jianshe Road, Zhengzhou 450052, China.
| | - Guiquan Chen
- State Key Laboratory of Pharmaceutical Biotechnology, MOE Key Laboratory of Model Animal for Disease Study, Model Animal Research Center, Nanjing University, 12 Xuefu Avenue, Nanjing, Jiangsu Province 210061, China.
| |
Collapse
|
20
|
Debieu S, Solier S, Colombeau L, Versini A, Sindikubwabo F, Forrester A, Müller S, Cañeque T, Rodriguez R. Small Molecule Regulators of Ferroptosis. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2021; 1301:81-121. [PMID: 34370289 DOI: 10.1007/978-3-030-62026-4_6] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Ferroptosis is a dedicated mode of cell death involving iron, reactive oxygen species and lipid peroxidation. Involved in processes such as glutathione metabolism, lysosomal iron retention or interference with lipid metabolism, leading either to activation or inhibition of ferroptosis. Given the implications of ferroptosis in diseases such as cancer, aging, Alzheimer and infectious diseases, new molecular mechanisms underlying ferroptosis and small molecules regulators that target those mechanisms have prompted a great deal of interest. Here, we discuss the current scenario of small molecules modulating ferroptosis and critically assess what is known about their mechanisms of action.
Collapse
Affiliation(s)
- Sylvain Debieu
- Institut Curie, 26 rue d'Ulm, 75248 Paris Cedex 05, France
- PSL Université Paris, Paris, France
- Chemical Biology of Cancer Laboratory, CNRS UMR 3666, INSERM U1143, Paris, France
| | - Stéphanie Solier
- Institut Curie, 26 rue d'Ulm, 75248 Paris Cedex 05, France
- PSL Université Paris, Paris, France
- Chemical Biology of Cancer Laboratory, CNRS UMR 3666, INSERM U1143, Paris, France
| | - Ludovic Colombeau
- Institut Curie, 26 rue d'Ulm, 75248 Paris Cedex 05, France
- PSL Université Paris, Paris, France
- Chemical Biology of Cancer Laboratory, CNRS UMR 3666, INSERM U1143, Paris, France
| | - Antoine Versini
- Institut Curie, 26 rue d'Ulm, 75248 Paris Cedex 05, France
- PSL Université Paris, Paris, France
- Chemical Biology of Cancer Laboratory, CNRS UMR 3666, INSERM U1143, Paris, France
| | - Fabien Sindikubwabo
- Institut Curie, 26 rue d'Ulm, 75248 Paris Cedex 05, France
- PSL Université Paris, Paris, France
- Chemical Biology of Cancer Laboratory, CNRS UMR 3666, INSERM U1143, Paris, France
| | - Alison Forrester
- Institut Curie, 26 rue d'Ulm, 75248 Paris Cedex 05, France
- PSL Université Paris, Paris, France
- Chemical Biology of Cancer Laboratory, CNRS UMR 3666, INSERM U1143, Paris, France
| | - Sebastian Müller
- Institut Curie, 26 rue d'Ulm, 75248 Paris Cedex 05, France
- PSL Université Paris, Paris, France
- Chemical Biology of Cancer Laboratory, CNRS UMR 3666, INSERM U1143, Paris, France
| | - Tatiana Cañeque
- Institut Curie, 26 rue d'Ulm, 75248 Paris Cedex 05, France
- PSL Université Paris, Paris, France
- Chemical Biology of Cancer Laboratory, CNRS UMR 3666, INSERM U1143, Paris, France
| | - Raphaël Rodriguez
- Institut Curie, 26 rue d'Ulm, 75248 Paris Cedex 05, France.
- PSL Université Paris, Paris, France.
- Chemical Biology of Cancer Laboratory, CNRS UMR 3666, INSERM U1143, Paris, France.
| |
Collapse
|
21
|
Najafov A, Luu HS, Mookhtiar AK, Mifflin L, Xia HG, Amin PP, Ordureau A, Wang H, Yuan J. RIPK1 Promotes Energy Sensing by the mTORC1 Pathway. Mol Cell 2020; 81:370-385.e7. [PMID: 33271062 DOI: 10.1016/j.molcel.2020.11.008] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2020] [Revised: 08/31/2020] [Accepted: 11/04/2020] [Indexed: 02/03/2023]
Abstract
The mechanisms of cellular energy sensing and AMPK-mediated mTORC1 inhibition are not fully delineated. Here, we discover that RIPK1 promotes mTORC1 inhibition during energetic stress. RIPK1 is involved in mediating the interaction between AMPK and TSC2 and facilitate TSC2 phosphorylation at Ser1387. RIPK1 loss results in a high basal mTORC1 activity that drives defective lysosomes in cells and mice, leading to accumulation of RIPK3 and CASP8 and sensitization to cell death. RIPK1-deficient cells are unable to cope with energetic stress and are vulnerable to low glucose levels and metformin. Inhibition of mTORC1 rescues the lysosomal defects and vulnerability to energetic stress and prolongs the survival of RIPK1-deficient neonatal mice. Thus, RIPK1 plays an important role in the cellular response to low energy levels and mediates AMPK-mTORC1 signaling. These findings shed light on the regulation of mTORC1 during energetic stress and unveil a point of crosstalk between pro-survival and pro-death pathways.
Collapse
Affiliation(s)
- Ayaz Najafov
- Department of Cell Biology, Harvard Medical School, Boston, MA 02115, USA; Ludwig Center, Harvard Medical School, Boston, MA 02115, USA.
| | - Hoang Son Luu
- Department of Cell Biology, Harvard Medical School, Boston, MA 02115, USA
| | - Adnan K Mookhtiar
- Department of Cell Biology, Harvard Medical School, Boston, MA 02115, USA
| | - Lauren Mifflin
- Department of Cell Biology, Harvard Medical School, Boston, MA 02115, USA
| | - Hong-Guang Xia
- Department of Cell Biology, Harvard Medical School, Boston, MA 02115, USA
| | - Palak P Amin
- Department of Cell Biology, Harvard Medical School, Boston, MA 02115, USA
| | - Alban Ordureau
- Department of Cell Biology, Harvard Medical School, Boston, MA 02115, USA
| | - Huibing Wang
- Department of Cell Biology, Harvard Medical School, Boston, MA 02115, USA
| | - Junying Yuan
- Department of Cell Biology, Harvard Medical School, Boston, MA 02115, USA; Ludwig Center, Harvard Medical School, Boston, MA 02115, USA.
| |
Collapse
|
22
|
Liu J, Xie X, Yan D, Wang Y, Yuan H, Cai Y, Luo J, Xu A, Huang Y, Cheung CW, Irwin MG, Xia Z. Up-regulation of FoxO1 contributes to adverse vascular remodelling in type 1 diabetic rats. J Cell Mol Med 2020; 24:13727-13738. [PMID: 33108705 PMCID: PMC7754018 DOI: 10.1111/jcmm.15935] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2020] [Revised: 09/08/2020] [Accepted: 09/10/2020] [Indexed: 02/06/2023] Open
Abstract
Vascular complications from diabetes often result in poor outcomes for patients, even after optimized interventions. Forkhead box protein O1 (FoxO1) is a key regulator of cellular metabolism and plays an important role in vessel formation and maturation. Alterations of FoxO1 occur in the cardiovascular system in diabetes, yet the role of FoxO1 in diabetic vascular complications is poorly understood. In Streptozotocin (STZ)‐induced type 1 diabetic rats, FoxO1 expression was up‐regulated in carotid arteries at 8 weeks of diabetes that was accompanied with adverse vascular remodelling characterized as increased wall thickness, carotid medial cross‐sectional area, media‐to‐lumen ratio and decreased carotid artery lumen area. This adverse vascular remodelling induced by hyperglycaemia in diabetic rats required FoxO1 activation as pharmacological inhibition of FoxO1 with 50mg/kg AS1842856 (AS) reversed vascular remodelling in type 1 diabetic rats. The adverse vascular remodelling in type 1 diabetes mellitus (T1DM) occurred concomitantly with increases in pro‐inflammatory factors, adhesion factors, apoptosis, NOD‐like receptor family protein‐3 inflammasome activation and the phenotypic switch of arterial smooth muscle cells, which were all reversed by AS. In addition, FoxO1 inhibition counteracted the down‐regulation of its upstream mediator PDK1 in T1DM. PDK1 activator reduced FoxO1 nuclear translocation, which serves as the basis for subsequent transcriptional regulation during hyperglycaemia. Taken together, our data suggest that FoxO1 is a critical trigger for type 1 diabetes‐induced vascular remodelling in rats, and inhibition of FoxO1 thus offers a potential therapeutic option for diabetes‐associated cardiovascular diseases.
Collapse
Affiliation(s)
- Jingjin Liu
- Department of Anesthesiology, University of Hong Kong, Hong Kong, China
| | - Xiang Xie
- Department of Anesthesiology, University of Hong Kong, Hong Kong, China.,Department of Anesthesiology, The Second Affiliated Hospital and Yuying Children's Hospital, Wenzhou Medical University, Wenzhou, China
| | - Dan Yan
- Department of Anesthesiology, University of Hong Kong, Hong Kong, China
| | - Yongshun Wang
- Department of Biomedical Science, University of Hong Kong, Hong Kong, China
| | - Hongbin Yuan
- Department of Anesthesiology, Changzheng Hospital, Second Military Medical University, Shanghai, China
| | - Yin Cai
- Department of Anesthesiology, University of Hong Kong, Hong Kong, China
| | - Jierong Luo
- Department of Anesthesiology, Guangzhou First People's Hospital, Guangzhou Medical University, Guangzhou, China
| | - Aimin Xu
- State Key Laboratory of Pharmaceutical Biotechnology, the University of Hong Kong, Hong Kong, China
| | - Yu Huang
- Heart and Vascular Institute and School of Biomedical Sciences, Chinese University of Hong Kong, Hong Kong SAR, China
| | - Chi Wai Cheung
- Department of Anesthesiology, University of Hong Kong, Hong Kong, China
| | - Michael G Irwin
- Department of Anesthesiology, University of Hong Kong, Hong Kong, China
| | - Zhengyuan Xia
- Department of Anesthesiology, University of Hong Kong, Hong Kong, China.,State Key Laboratory of Pharmaceutical Biotechnology, the University of Hong Kong, Hong Kong, China
| |
Collapse
|
23
|
Niederdorfer B, Touré V, Vazquez M, Thommesen L, Kuiper M, Lægreid A, Flobak Å. Strategies to Enhance Logic Modeling-Based Cell Line-Specific Drug Synergy Prediction. Front Physiol 2020; 11:862. [PMID: 32848834 PMCID: PMC7399174 DOI: 10.3389/fphys.2020.00862] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2020] [Accepted: 06/26/2020] [Indexed: 12/16/2022] Open
Abstract
Discrete dynamical modeling shows promise in prioritizing drug combinations for screening efforts by reducing the experimental workload inherent to the vast numbers of possible drug combinations. We have investigated approaches to predict combination responses across different cancer cell lines using logic models generated from one generic prior-knowledge network representing 144 nodes covering major cancer signaling pathways. Cell-line specific models were configured to agree with baseline activity data from each unperturbed cell line. Testing against experimental data demonstrated a high number of true positive and true negative predictions, including also cell-specific responses. We demonstrate the possible enhancement of predictive capability of models by curation of literature knowledge further detailing subtle biologically founded signaling mechanisms in the model topology. In silico model analysis pinpointed a subset of network nodes highly influencing model predictions. Our results indicate that the performance of logic models can be improved by focusing on high-influence node protein activity data for model configuration and that these nodes accommodate high information flow in the regulatory network.
Collapse
Affiliation(s)
- Barbara Niederdorfer
- Department of Clinical and Molecular Medicine, Norwegian University of Science and Technology, Trondheim, Norway
| | - Vasundra Touré
- Department of Biology, Norwegian University of Science and Technology, Trondheim, Norway
| | - Miguel Vazquez
- Department of Clinical and Molecular Medicine, Norwegian University of Science and Technology, Trondheim, Norway.,Barcelona Supercomputing Center, Barcelona, Spain
| | - Liv Thommesen
- Department of Biomedical Laboratory Science, Norwegian University of Science and Technology, Trondheim, Norway
| | - Martin Kuiper
- Department of Biology, Norwegian University of Science and Technology, Trondheim, Norway
| | - Astrid Lægreid
- Department of Clinical and Molecular Medicine, Norwegian University of Science and Technology, Trondheim, Norway
| | - Åsmund Flobak
- Department of Clinical and Molecular Medicine, Norwegian University of Science and Technology, Trondheim, Norway.,The Cancer Clinic, St. Olav's University Hospital, Trondheim, Norway
| |
Collapse
|
24
|
Phosphoproteomics reveals that the hVPS34 regulated SGK3 kinase specifically phosphorylates endosomal proteins including Syntaxin-7, Syntaxin-12, RFIP4 and WDR44. Biochem J 2020; 476:3081-3107. [PMID: 31665227 PMCID: PMC6824681 DOI: 10.1042/bcj20190608] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2019] [Revised: 10/02/2019] [Accepted: 10/03/2019] [Indexed: 01/04/2023]
Abstract
The serum- and glucocorticoid-regulated kinase (SGK) isoforms contribute resistance to cancer therapies targeting the PI3K pathway. SGKs are homologous to Akt and these kinases display overlapping specificity and phosphorylate several substrates at the same residues, such as TSC2 to promote tumor growth by switching on the mTORC1 pathway. The SGK3 isoform is up-regulated in breast cancer cells treated with PI3K or Akt inhibitors and recruited and activated at endosomes, through its phox homology domain binding to PtdIns(3)P. We undertook genetic and pharmacological phosphoproteomic screens to uncover novel SGK3 substrates. We identified 40 potential novel SGK3 substrates, including four endosomal proteins STX7 (Ser126) and STX12 (Ser139), RFIP4 (Ser527) and WDR44 (Ser346) that were efficiently phosphorylated in vitro by SGK3 at the sites identified in vivo, but poorly by Akt. We demonstrate that these substrates are inefficiently phosphorylated by Akt as they possess an n + 1 residue from the phosphorylation site that is unfavorable for Akt phosphorylation. Phos-tag analysis revealed that stimulation of HEK293 cells with IGF1 to activate SGK3, promoted phosphorylation of a significant fraction of endogenous STX7 and STX12, in a manner that was blocked by knock-out of SGK3 or treatment with a pan SGK inhibitor (14H). SGK3 phosphorylation of STX12 enhanced interaction with the VAMP4/VTI1A/STX6 containing the SNARE complex and promoted plasma membrane localization. Our data reveal novel substrates for SGK3 and suggest a mechanism by which STX7 and STX12 SNARE complexes are regulated by SGK3. They reveal new biomarkers for monitoring SGK3 pathway activity.
Collapse
|
25
|
An K, Qin Q, Yu S, Xue M, Wang Z, Lin Q, Ma Y, Yan G, Mo S, Chen Y, Zhang L, Zhong J, Qi Z, Xia J. Combination of N, N'-dicyclohexyl-N-arachidonic acylurea and tacrolimus prolongs cardiac allograft survival in mice. Immunol Cell Biol 2020; 98:382-396. [PMID: 32162358 DOI: 10.1111/imcb.12327] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2019] [Revised: 01/29/2020] [Accepted: 03/09/2020] [Indexed: 12/27/2022]
Abstract
Current immunosuppressive agents for organ transplantation are not ideal because of their strong toxicity and adverse effects. Hence, there is an urgent need to develop novel immunosuppressive agents. The compound N, N'-dicyclohexyl-N-arachidonic acylurea (DCAAA) is a novel highly unsaturated fatty acid from the traditional Chinese medicinal plant Radix Isatidis. In this study, we systematically investigated the toxicity, immunosuppressive effect and mechanisms underlying the activity of DCAAA. The toxicity tests showed that DCAAA treatment did not lead to red blood cell hemolysis and did not affect the liver and kidney functions in mice. The lymphocyte transformation test showed that DCAAA treatment inhibited lymphocyte proliferation in a dose-dependent manner. An in vivo cardiac allotransplantation experiment showed that DCAAA treatment could suppress the immune rejection and significantly prolong the survival of cardiac allografts in recipient mice by reducing the proportion of CD4+ T cells in the spleen and grafts, concentration of interferon-γ in the supernatant and serum and infiltration of inflammatory cells into the grafts. Moreover, a combination treatment with DCAAA and tacrolimus had a synergistic effect in preventing acute rejection of heart transplants. In vitro molecular biology experiments showed that DCAAA treatment inhibited activation of the T-cell receptor-mediated phosphoinostide 3-kinase-protein kinase B pathway, thereby arresting cell cycle transition from the G1 to the S phase, and inhibiting lymphocyte proliferation. Overall, our study reveals a novel, low-toxicity immunosuppressive agent that has the potential to reduce the toxic side effects of existing immunosuppressive agents when used in combination with them.
Collapse
Affiliation(s)
- Ke An
- Organ Transplantation Institute of Xiamen University, Fujian Provincial Key Laboratory of Organ and Tissue Regeneration, School of Medicine, Xiamen University, Xiamen, Fujian, China
| | - Qing Qin
- Department of Natural Product Chemistry, School of Pharmaceutical Sciences, Guangxi Medical University, Nanning, Guangxi, China
| | - Shengnan Yu
- Department of Obstetrics and Gynecology, Zhongshan Hospital, Xiamen University, Xiamen, Fujian, China
| | - Mengjiao Xue
- Organ Transplantation Institute of Xiamen University, Fujian Provincial Key Laboratory of Organ and Tissue Regeneration, School of Medicine, Xiamen University, Xiamen, Fujian, China
| | - Zhenzhen Wang
- Organ Transplantation Institute of Xiamen University, Fujian Provincial Key Laboratory of Organ and Tissue Regeneration, School of Medicine, Xiamen University, Xiamen, Fujian, China
| | - Qingru Lin
- Organ Transplantation Institute of Xiamen University, Fujian Provincial Key Laboratory of Organ and Tissue Regeneration, School of Medicine, Xiamen University, Xiamen, Fujian, China
| | - Yunhan Ma
- Organ Transplantation Institute of Xiamen University, Fujian Provincial Key Laboratory of Organ and Tissue Regeneration, School of Medicine, Xiamen University, Xiamen, Fujian, China
| | - Guoliang Yan
- Organ Transplantation Institute of Xiamen University, Fujian Provincial Key Laboratory of Organ and Tissue Regeneration, School of Medicine, Xiamen University, Xiamen, Fujian, China
| | - Sirui Mo
- Youjiang Medical University For Nationalities, Baise, Guangxi, China
| | - Yingyu Chen
- Organ Transplantation Institute of Xiamen University, Fujian Provincial Key Laboratory of Organ and Tissue Regeneration, School of Medicine, Xiamen University, Xiamen, Fujian, China
| | - Liyi Zhang
- Organ Transplantation Institute of Xiamen University, Fujian Provincial Key Laboratory of Organ and Tissue Regeneration, School of Medicine, Xiamen University, Xiamen, Fujian, China
| | - Jiaying Zhong
- Organ Transplantation Institute of Xiamen University, Fujian Provincial Key Laboratory of Organ and Tissue Regeneration, School of Medicine, Xiamen University, Xiamen, Fujian, China
| | - Zhongquan Qi
- School of Medicine, Guangxi University, Nanning, Guangxi, China
| | - Junjie Xia
- Organ Transplantation Institute of Xiamen University, Fujian Provincial Key Laboratory of Organ and Tissue Regeneration, School of Medicine, Xiamen University, Xiamen, Fujian, China
| |
Collapse
|
26
|
Sriskandarajah P, De Haven Brandon A, MacLeod K, Carragher NO, Kirkin V, Kaiser M, Whittaker SR. Combined targeting of MEK and the glucocorticoid receptor for the treatment of RAS-mutant multiple myeloma. BMC Cancer 2020; 20:269. [PMID: 32228485 PMCID: PMC7106683 DOI: 10.1186/s12885-020-06735-2] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2019] [Accepted: 03/11/2020] [Indexed: 12/20/2022] Open
Abstract
BACKGROUND Multiple myeloma (MM) remains incurable despite recent therapeutic advances. RAS mutations are frequently associated with relapsed/refractory disease. Efforts to target the mitogen-activated protein kinase (MAPK) pathway with the MEK inhibitor, trametinib (Tra) have been limited by toxicities and the development of resistance. Dexamethasone (Dex) is a corticosteroid commonly used in clinical practice, to enhance efficacy of anti-myeloma therapy. Therefore, we hypothesised that the combination of Tra and Dex would yield synergistic activity in RAS-mutant MM. METHODS The response of human MM cell lines to drug treatment was analysed using cell proliferation assays, Western blotting, Annexin V and propidium iodide staining by flow cytometry and reverse phase protein arrays. The efficacy of trametinib and dexamethasone treatment in the MM.1S xenograft model was assessed by measuring tumor volume over time. RESULTS The Tra/Dex combination demonstrated synergistic cytotoxicity in KRASG12A mutant lines MM.1S and RPMI-8226. The induction of apoptosis was associated with decreased MCL-1 expression and increased BIM expression. Reverse phase proteomic arrays revealed suppression of FAK, PYK2, FLT3, NDRG1 and 4EBP1 phosphorylation with the Tra/Dex combination. Notably, NDRG1 expression was associated with the synergistic response to Tra/Dex. MM cells were sensitive to PDK1 inhibition and IGF1-induced signalling partially protected from Tra/Dex treatment, highlighting the importance of this pathway. In the MM.1S tumor xenograft model, only the combination of Tra/Dex resulted in a significant inhibition of tumor growth. CONCLUSIONS Overall Tra/Dex demonstrates antiproliferative activity in RAS-mutant MM cell lines associated with suppression of pro-survival PDK1 signalling and engagement of apoptotic pathways. Our data support further investigation of this combination in RAS-mutant MM.
Collapse
Affiliation(s)
- Priya Sriskandarajah
- Division of Cancer Therapeutics, The Institute of Cancer Research, London, SW7 3RP, UK.,The Royal Marsden NHS Foundation Trust, London, UK
| | | | - Kenneth MacLeod
- Cancer Research UK Edinburgh Centre, Institute of Genetics and Molecular Medicine, The University of Edinburgh, Edinburgh, UK
| | - Neil O Carragher
- Cancer Research UK Edinburgh Centre, Institute of Genetics and Molecular Medicine, The University of Edinburgh, Edinburgh, UK
| | - Vladimir Kirkin
- Division of Cancer Therapeutics, The Institute of Cancer Research, London, SW7 3RP, UK
| | - Martin Kaiser
- The Royal Marsden NHS Foundation Trust, London, UK.,Division of Molecular Pathology, The Institute of Cancer Research, London, UK
| | - Steven R Whittaker
- Division of Cancer Therapeutics, The Institute of Cancer Research, London, SW7 3RP, UK.
| |
Collapse
|
27
|
Mansi I, Al-Sha'er MA, Mhaidat N, Taha M. Ligand Based Pharmacophore Modeling Followed by Biological Screening Lead to Discovery of Novel PDK1 Inhibitors as Anticancer Agents. Anticancer Agents Med Chem 2020; 20:476-485. [PMID: 31889497 DOI: 10.2174/1871520620666191224110600] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2019] [Revised: 08/21/2019] [Accepted: 10/22/2019] [Indexed: 11/22/2022]
Abstract
BACKGROUND Phosphoinositide-Dependent Kinase-1 (PDK1) is a serine/threonine kinase, which belongs to AGC kinase family required by cancer cells. METHODS Pharmacophoric space of 86 PDK1 inhibitors using six diverse sets of inhibitors was explored to identify high-quality pharmacophores. The best combination of pharmacophoric models and physicochemical descriptors was selected by genetic algorithm-based QSAR analysis that can elucidate the variation of bioactivity within the training inhibitors. Two successful orthogonal pharmacophores emerged in the optimum QSAR equation (r2 69 = 0.90, r2 LOO= 0.86, F= 51.92, r2 PRESS against 17 test inhibitors = 0.79). Receiver Operating Characteristic (ROC) curve analyses were used to estimate the QSAR-selected pharmacophores. RESULTS 5 out of 11 compounds tested had shown potential intracellular PDK1 inhibition with the highest inhibition percent for compounds 92 and 93 as follows; 90 and 92% PDK1 inhibition, respectively. CONCLUSION PDK1 inhibitors are potential anticancer agents that can be discovered by combination method of ligand based design with QSAR and ROC analysis.
Collapse
Affiliation(s)
- Iman Mansi
- Faculty of Pharmaceutical Sciences, The Hashemite University, P.O. Box 330127 Zarqa, 13133, Jordan
| | | | - Nizar Mhaidat
- Clinical Pharmacy Department, Faculty of Pharmacy, Jordan University of Science & Technology, Irbid, Jordan
| | - Mutasem Taha
- Drug Design Center, Faculty of Pharmacy, University of Jordan, Amman, Jordan
| |
Collapse
|
28
|
Liu J, Yang J, Hou Y, Zhu Z, He J, Zhao H, Ye X, Li D, Wu Z, Huang Z, Hao B, Yao K. Casticin inhibits nasopharyngeal carcinoma growth by targeting phosphoinositide 3-kinase. Cancer Cell Int 2019; 19:348. [PMID: 31889900 PMCID: PMC6925493 DOI: 10.1186/s12935-019-1069-6] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2019] [Accepted: 12/12/2019] [Indexed: 12/13/2022] Open
Abstract
Background Casticin, an isoflavone compound extracted from the herb Fructus Viticis, has demonstrated anti-inflammatory and anticancer activities and properties. The aim of this study was to investigate the effects and mechanisms of casticin in nasopharyngeal carcinoma (NPC) cells and to determine its potential for targeted use as a medicine. Methods NPC cells were used to perform the experiments. The CCK‑8 assay and colony formation assays were used to assess cell viability. Flow cytometry was used to measure the cell cycle and apoptosis analysis (annexin V/PI assay). A three-dimensional (3D) tumour sphere culture system was used to characterize the effect of casticin on NPC stem cells. In silico molecular docking prediction and high-throughput KINOME scan assays were used to evaluate the binding of casticin to phosphoinositide 3-kinase (PI3K), including wild-type and most of mutants variants. We also used the SelectScreen assay to detect the IC50 of ATP activity in the active site of the target kinase. Western blotting was used to evaluate the changes in key proteins involved cell cycle, apoptosis, stemness, and PI3K/protein kinase B (AKT) signalling. The effect of casticin treatment in vivo was determined by using a xenograft mouse model. Results Our results indicate that casticin is a new and novel selective PI3K inhibitor that can significantly inhibit NPC proliferation and that it induces G2/GM arrest and apoptosis by upregulating Bax/BCL2 expression. Moreover, casticin was observed to affect the self-renewal ability of the nasopharyngeal carcinoma cell lines, and a combination of casticin with BYL719 was observed to induce a decrease in the level of the phosphorylation of mTORC1 downstream targets in BYL719-insensitive NPC cell lines. Conclusion Casticin is a newly emerging selective PI3K inhibitor with potential for use as a targeted therapeutic treatment for nasopharyngeal carcinoma. Accordingly, casticin might represent a novel and effective agent against NPC and likely has high potential for combined use with pharmacological agents targeting PI3K/AKT.
Collapse
Affiliation(s)
- Jingxian Liu
- 1Guangdong Provincial Key Laboratory of Tumor Immunotherapy, Cancer Research Institute, School of Basic Medical Sciences, Southern Medical University, Guangzhou, 510515 Guangdong People's Republic of China
| | - Jinghong Yang
- 1Guangdong Provincial Key Laboratory of Tumor Immunotherapy, Cancer Research Institute, School of Basic Medical Sciences, Southern Medical University, Guangzhou, 510515 Guangdong People's Republic of China
| | - Yuhe Hou
- 1Guangdong Provincial Key Laboratory of Tumor Immunotherapy, Cancer Research Institute, School of Basic Medical Sciences, Southern Medical University, Guangzhou, 510515 Guangdong People's Republic of China
| | - Zhenwei Zhu
- 1Guangdong Provincial Key Laboratory of Tumor Immunotherapy, Cancer Research Institute, School of Basic Medical Sciences, Southern Medical University, Guangzhou, 510515 Guangdong People's Republic of China.,2Shenzhen Hospital, Southern Medical University, Shenzhen, 518000 Guangdong People's Republic of China
| | - Jie He
- 1Guangdong Provincial Key Laboratory of Tumor Immunotherapy, Cancer Research Institute, School of Basic Medical Sciences, Southern Medical University, Guangzhou, 510515 Guangdong People's Republic of China
| | - Hao Zhao
- 1Guangdong Provincial Key Laboratory of Tumor Immunotherapy, Cancer Research Institute, School of Basic Medical Sciences, Southern Medical University, Guangzhou, 510515 Guangdong People's Republic of China
| | - Xidong Ye
- 1Guangdong Provincial Key Laboratory of Tumor Immunotherapy, Cancer Research Institute, School of Basic Medical Sciences, Southern Medical University, Guangzhou, 510515 Guangdong People's Republic of China
| | - Dengke Li
- 1Guangdong Provincial Key Laboratory of Tumor Immunotherapy, Cancer Research Institute, School of Basic Medical Sciences, Southern Medical University, Guangzhou, 510515 Guangdong People's Republic of China
| | - Zhaohui Wu
- 1Guangdong Provincial Key Laboratory of Tumor Immunotherapy, Cancer Research Institute, School of Basic Medical Sciences, Southern Medical University, Guangzhou, 510515 Guangdong People's Republic of China
| | - Zhongxi Huang
- 1Guangdong Provincial Key Laboratory of Tumor Immunotherapy, Cancer Research Institute, School of Basic Medical Sciences, Southern Medical University, Guangzhou, 510515 Guangdong People's Republic of China.,2Shenzhen Hospital, Southern Medical University, Shenzhen, 518000 Guangdong People's Republic of China
| | - Bingtao Hao
- 1Guangdong Provincial Key Laboratory of Tumor Immunotherapy, Cancer Research Institute, School of Basic Medical Sciences, Southern Medical University, Guangzhou, 510515 Guangdong People's Republic of China.,3Shunde Hospital, Southern Medical University, Shunde, 528300 Guangdong People's Republic of China
| | - Kaitai Yao
- 1Guangdong Provincial Key Laboratory of Tumor Immunotherapy, Cancer Research Institute, School of Basic Medical Sciences, Southern Medical University, Guangzhou, 510515 Guangdong People's Republic of China.,2Shenzhen Hospital, Southern Medical University, Shenzhen, 518000 Guangdong People's Republic of China
| |
Collapse
|
29
|
Lee SY, Kim H, Li CM, Kang J, Najafov A, Jung M, Kang S, Wang S, Yuan J, Jung YK. Casein kinase-1γ1 and 3 stimulate tumor necrosis factor-induced necroptosis through RIPK3. Cell Death Dis 2019; 10:923. [PMID: 31801942 PMCID: PMC6892881 DOI: 10.1038/s41419-019-2146-4] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2018] [Revised: 09/13/2019] [Accepted: 10/28/2019] [Indexed: 01/23/2023]
Abstract
Upon necroptosis activation, receptor interacting serine/threonine kinase (RIPK)1 and RIPK3 form a necrosome complex with pseudokinase mixed lineage kinase-like (MLKL). Although protein phosphorylation is a key event for RIPK1 and RIPK3 activation in response to a necroptosis signal, relatively little is known about other factors that might regulate the activity of these kinases or necrosome formation. Through a gain-of-function screen with 546 kinases and 127 phosphatases, we identified casein kinase 1 gamma (CK1γ) as a candidate necroptosis-promoting factor. Here, we show that the decreased activity or amounts of CK1γ1 and CK1γ3, either by treatment with a chemical inhibitor or knockdown in cells, reduced TNFα-induced necroptosis. Conversely, ectopic expression of CK1γ1 or CK1γ3 exacerbated necroptosis, but not apoptosis. Similar to RIPK1 and RIPK3, CK1γ1 was also cleaved at Asp343 by caspase-8 during apoptosis. CK1γ1 and CK1γ3 formed a protein complex and were recruited to the necrosome harboring RIPK1, RIPK3 and MLKL. In particular, an autophosphorylated form of CK1γ3 at Ser344/345 was detected in the necrosome and was required to mediate the necroptosis. In addition, in vitro assays with purified proteins showed that CK1γ phosphorylated RIPK3, affecting its activity, and in vivo assays showed that the CK1γ-specific inhibitor Gi prevented abrupt death in mice with hypothermia in a model of TNFα-induced systemic inflammatory response syndrome. Collectively, these data suggest that CK1γ1 and CK1γ3 are required for TNFα-induced necroptosis likely by regulating RIPK3.
Collapse
Affiliation(s)
- Song-Yi Lee
- School of Biological Science, Seoul National University, 1 Gwanak-ro, Gwanak-gu, Seoul, 08826, Korea
| | - Hyunjoo Kim
- School of Biological Science, Seoul National University, 1 Gwanak-ro, Gwanak-gu, Seoul, 08826, Korea
| | - Cathena Meiling Li
- School of Biological Science, Seoul National University, 1 Gwanak-ro, Gwanak-gu, Seoul, 08826, Korea
| | - Jaemin Kang
- School of Biological Science, Seoul National University, 1 Gwanak-ro, Gwanak-gu, Seoul, 08826, Korea
| | - Ayaz Najafov
- Department of Cell Biology, Harvard Medical School, 240 Longwood Avenue, Boston, MA, 02115-5730, USA
| | - Muhah Jung
- School of Biological Science, Seoul National University, 1 Gwanak-ro, Gwanak-gu, Seoul, 08826, Korea
| | - Soosung Kang
- Department of Pharmacy, Ewha Womans University, 52 Ewhayeodae-gil, Seodaemun-gu, Seoul, 03760, Korea
| | - Shaomeng Wang
- Department of Pharmacy, University of Michigan, 210 Washtenaw Avenue, Ann Arbor, MI, 48109-2216, USA
| | - Junying Yuan
- Department of Cell Biology, Harvard Medical School, 240 Longwood Avenue, Boston, MA, 02115-5730, USA
| | - Yong-Keun Jung
- School of Biological Science, Seoul National University, 1 Gwanak-ro, Gwanak-gu, Seoul, 08826, Korea.
| |
Collapse
|
30
|
Leroux AE, Biondi RM. Renaissance of Allostery to Disrupt Protein Kinase Interactions. Trends Biochem Sci 2019; 45:27-41. [PMID: 31690482 DOI: 10.1016/j.tibs.2019.09.007] [Citation(s) in RCA: 39] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2019] [Revised: 09/18/2019] [Accepted: 09/25/2019] [Indexed: 12/11/2022]
Abstract
Protein-protein interactions often regulate the activity of protein kinases by allosterically modulating the conformation of the ATP-binding site. Bidirectional allostery implies that reverse modulation (i.e., from the ATP-binding site to the interaction and regulatory sites) must also be possible. Here, we review both the allosteric regulation of protein kinases and recent work describing how compounds binding at the ATP-binding site can promote or inhibit protein kinase interactions at regulatory sites via the reverse mechanism. Notably, the pharmaceutical industry has been developing compounds that bind to the ATP-binding site of protein kinases and potently disrupt protein-protein interactions between target protein kinases and their regulatory interacting partners. Learning to modulate allosteric processes will facilitate the development of protein-protein interaction modulators.
Collapse
Affiliation(s)
- Alejandro E Leroux
- Instituto de Investigación en Biomedicina de Buenos Aires (IBioBA), CONICET, Partner Institute of the Max Planck Society, Buenos Aires C1425FQD, Argentina
| | - Ricardo M Biondi
- Instituto de Investigación en Biomedicina de Buenos Aires (IBioBA), CONICET, Partner Institute of the Max Planck Society, Buenos Aires C1425FQD, Argentina; Department of Internal Medicine I, University Hospital, Goethe University, Theodor-Stern-Kai 7, 60590 Frankfurt, Germany; DKTK German Cancer Consortium (DKTK), Frankfurt, Germany; German Cancer Research Center (DKFZ), Heidelberg, Germany.
| |
Collapse
|
31
|
Shi X, Swanson TL, Miner NB, Eshleman AJ, Janowsky A. Activation of Trace Amine-Associated Receptor 1 Stimulates an Antiapoptotic Signal Cascade via Extracellular Signal-Regulated Kinase 1/2. Mol Pharmacol 2019; 96:493-504. [PMID: 31409621 PMCID: PMC6744391 DOI: 10.1124/mol.119.116798] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2019] [Accepted: 08/02/2019] [Indexed: 12/30/2022] Open
Abstract
Methamphetamine (MA) is highly addictive and neurotoxic, causing cell death in humans and in rodent models. MA, along with many of its analogs, is an agonist at the G protein-coupled trace amine-associated receptor 1 (TAAR1). TAAR1 activation protects against MA-induced degeneration of dopaminergic neurons, suggesting that TAAR1 plays a role in regulating MA-induced neurotoxicity. However, the mechanisms involved in TAAR1's role in neurotoxicity and cell death have not been described in detail. In this study, we investigated the apoptosis pathway in Taar1 wild-type (WT) and knockout (KO) mice and in cells expressing the recombinant receptor. Bcl-2, an antiapoptotic protein, was upregulated ∼3-fold in the midbrain area (substantial nigra and ventral tegmental area) in Taar1 KO compared with WT mice, and MA significantly increased Bcl-2 expression in WT mice but decreased Bcl-2 expression in KO mice. The proapoptotic protein Bax did not differ across genotype or in response to MA. Bcl-2 expression was significantly upregulated by the TAAR1 agonist RO5166017 ((S)-4-[(ethyl-phenyl-amino)-methyl]-4,5-dihydro-oxazol-2-ylamine) in cells expressing the recombinant mouse TAAR1. Additionally, activation of TAAR1 by RO5166017 increased phosphorylation of extracellular signal-regulated kinase (ERK) 1/2, and protein kinase B (AKT), but only inhibition of ERK1/2 phosphorylation prevented TAAR1-induced increases in Bcl-2 levels, indicating that TAAR1 activation increases Bcl-2 through an ERK1/2-dependent pathway. All changes to ERK1/2 pathway intermediates were blocked by the TAAR1 antagonist, N-(3-ethoxyphenyl)-4-(1-pyrrolidinyl)-3-(trifluoromethyl) benzamide. These findings suggest that TAAR1 activation protects against MA-induced cell apoptosis and TAAR1 may play a role in cell death in neurodegenerative diseases. SIGNIFICANCE STATEMENT: Methamphetamine stimulates TAAR1, a G protein-coupled receptor. The role and mechanisms for TAAR1 in methamphetamine-induced neurotoxicity are not known. Here, we report that, in genetic mouse models and cells expressing the recombinant receptor, TAAR1 activates the ERK1/2 pathway but not the AKT pathway to upregulate the antiapoptotic protein Bcl-2, which protects cells from drug-induced toxicity.
Collapse
Affiliation(s)
- Xiao Shi
- Research Service, Veterans Affairs Portland Health Care System, Portland, Oregon (X.S., T.L.S., N.B.M., A.J.E., A.J.); and The Methamphetamine Abuse Research Center (X.S., A.J.) and Departments of Psychiatry (X.S., T.L.S., A.J.E., A.J.) and Behavioral Neuroscience (N.B.M., A.J.E., A.J.), Oregon Health and Science University, Portland, Oregon
| | - Tracy L Swanson
- Research Service, Veterans Affairs Portland Health Care System, Portland, Oregon (X.S., T.L.S., N.B.M., A.J.E., A.J.); and The Methamphetamine Abuse Research Center (X.S., A.J.) and Departments of Psychiatry (X.S., T.L.S., A.J.E., A.J.) and Behavioral Neuroscience (N.B.M., A.J.E., A.J.), Oregon Health and Science University, Portland, Oregon
| | - Nicholas B Miner
- Research Service, Veterans Affairs Portland Health Care System, Portland, Oregon (X.S., T.L.S., N.B.M., A.J.E., A.J.); and The Methamphetamine Abuse Research Center (X.S., A.J.) and Departments of Psychiatry (X.S., T.L.S., A.J.E., A.J.) and Behavioral Neuroscience (N.B.M., A.J.E., A.J.), Oregon Health and Science University, Portland, Oregon
| | - Amy J Eshleman
- Research Service, Veterans Affairs Portland Health Care System, Portland, Oregon (X.S., T.L.S., N.B.M., A.J.E., A.J.); and The Methamphetamine Abuse Research Center (X.S., A.J.) and Departments of Psychiatry (X.S., T.L.S., A.J.E., A.J.) and Behavioral Neuroscience (N.B.M., A.J.E., A.J.), Oregon Health and Science University, Portland, Oregon
| | - Aaron Janowsky
- Research Service, Veterans Affairs Portland Health Care System, Portland, Oregon (X.S., T.L.S., N.B.M., A.J.E., A.J.); and The Methamphetamine Abuse Research Center (X.S., A.J.) and Departments of Psychiatry (X.S., T.L.S., A.J.E., A.J.) and Behavioral Neuroscience (N.B.M., A.J.E., A.J.), Oregon Health and Science University, Portland, Oregon
| |
Collapse
|
32
|
Najafov A, Mookhtiar AK, Luu HS, Ordureau A, Pan H, Amin PP, Li Y, Lu Q, Yuan J. TAM Kinases Promote Necroptosis by Regulating Oligomerization of MLKL. Mol Cell 2019; 75:457-468.e4. [DOI: 10.1016/j.molcel.2019.05.022] [Citation(s) in RCA: 63] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2017] [Revised: 04/23/2019] [Accepted: 05/13/2019] [Indexed: 01/09/2023]
|
33
|
Emmanouilidi A, Fyffe CA, Ferro R, Edling CE, Capone E, Sestito S, Rapposelli S, Lattanzio R, Iacobelli S, Sala G, Maffucci T, Falasca M. Preclinical validation of 3-phosphoinositide-dependent protein kinase 1 inhibition in pancreatic cancer. JOURNAL OF EXPERIMENTAL & CLINICAL CANCER RESEARCH : CR 2019; 38:191. [PMID: 31088502 PMCID: PMC6518649 DOI: 10.1186/s13046-019-1191-2] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/05/2018] [Accepted: 04/25/2019] [Indexed: 11/02/2022]
Abstract
BACKGROUND The very aggressive nature and low survival rate of pancreatic ductal adenocarcinoma (PDAC) dictates the necessity to find novel efficacious therapies. Recent evidence suggests that phosphoinositide 3-kinase (PI3K) and 3-phosphoinositide-dependent protein kinase 1 (PDK1) are key effectors of oncogenic KRAS in PDAC. Herein, we report the role and mechanism of action of PDK1, a protein kinase of the AGC family, in PDAC. METHODS PDAC cell lines were treated with selective PDK1 inhibitors or transfected with specific PDK1-targeting siRNAs. In vitro and in vivo assays were performed to investigate the functional role of PDK1 in PDAC. Specifically, anchorage-dependent and anchorage-independent growth was assessed in PDAC cells upon inhibition or downregulation of PDK1. Detailed investigation of the effect of PDK1 inhibition/downregulation on specific signalling pathways was also performed by Western blotting analysis. A xenograft tumour mouse model was used to determine the effect of pharmacological inhibition of PDK1 on PDAC cells growth in vivo. RESULTS Treatment with specific inhibitors of PDK1 impaired anchorage-dependent and anchorage-independent growth of pancreatic cancer cell lines, as well as pancreatic tumour growth in a xenograft model. Mechanistically, inhibition or downregulation of PDK1 resulted in reduced activation of the serum/glucocorticoid regulated kinase family member 3 and subsequent reduced phosphorylation of its target N-Myc downstream regulated 1. Additionally, we found that combination of sub-optimal concentrations of inhibitors selective for PDK1 and the class IB PI3K isoform p110γ inhibits pancreatic cancer cell growth and colonies formation more potently than each single treatment. CONCLUSIONS Our data indicate that PDK1 is a suitable target for therapeutic intervention in PDAC and support the clinical development of PDK1 inhibitors for PDAC.
Collapse
Affiliation(s)
- Aikaterini Emmanouilidi
- Metabolic Signalling Group, School of Pharmacy and Biomedical Sciences, Curtin Health Innovation Research Institute, Curtin University, Perth, Western Australia, 6102, Australia
| | - Chanse A Fyffe
- Queen Mary University of London, Barts and The London School of Medicine and Dentistry, Blizard Institute, Centre for Cell Biology and Cutaneous Research, E1 2AT, London, UK
| | - Riccardo Ferro
- Queen Mary University of London, Barts and The London School of Medicine and Dentistry, Blizard Institute, Centre for Cell Biology and Cutaneous Research, E1 2AT, London, UK
| | - Charlotte E Edling
- Queen Mary University of London, Barts and The London School of Medicine and Dentistry, Blizard Institute, Centre for Cell Biology and Cutaneous Research, E1 2AT, London, UK
| | - Emily Capone
- Dipartimento di Scienze Mediche, Orali e Biotecnologiche, University G. d'Annunzio di Chieti-Pescara, Centro Studi sull Invecchiamento, CeSI-MeT, 66100, Chieti, Italy
| | - Simona Sestito
- Department of Pharmacy, University of Pisa, Via Bonanno, 6, 56126, Pisa, Italy
| | - Simona Rapposelli
- Department of Pharmacy, University of Pisa, Via Bonanno, 6, 56126, Pisa, Italy
| | - Rossano Lattanzio
- Dipartimento di Scienze Mediche, Orali e Biotecnologiche, University G. d'Annunzio di Chieti-Pescara, Centro Studi sull Invecchiamento, CeSI-MeT, 66100, Chieti, Italy
| | - Stefano Iacobelli
- Dipartimento di Scienze Mediche, Orali e Biotecnologiche, University G. d'Annunzio di Chieti-Pescara, Centro Studi sull Invecchiamento, CeSI-MeT, 66100, Chieti, Italy.,MediaPharma Srl, Via della Colonnetta, 50/A, 66100, Chieti, Italy
| | - Gianluca Sala
- Dipartimento di Scienze Mediche, Orali e Biotecnologiche, University G. d'Annunzio di Chieti-Pescara, Centro Studi sull Invecchiamento, CeSI-MeT, 66100, Chieti, Italy
| | - Tania Maffucci
- Queen Mary University of London, Barts and The London School of Medicine and Dentistry, Blizard Institute, Centre for Cell Biology and Cutaneous Research, E1 2AT, London, UK
| | - Marco Falasca
- Metabolic Signalling Group, School of Pharmacy and Biomedical Sciences, Curtin Health Innovation Research Institute, Curtin University, Perth, Western Australia, 6102, Australia. .,Queen Mary University of London, Barts and The London School of Medicine and Dentistry, Blizard Institute, Centre for Cell Biology and Cutaneous Research, E1 2AT, London, UK.
| |
Collapse
|
34
|
Macías-García B, García-Marín LJ, Bragado MJ, González-Fernández L. The calcium-sensing receptor regulates protein tyrosine phosphorylation through PDK1 in boar spermatozoa. Mol Reprod Dev 2019; 86:751-761. [PMID: 31074040 DOI: 10.1002/mrd.23160] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2019] [Revised: 03/20/2019] [Accepted: 04/11/2019] [Indexed: 12/25/2022]
Abstract
Regulation of protein tyrosine phosphorylation is required for sperm capacitation and oocyte fertilization. The objective of the present work was to study the role of the calcium-sensing receptor (CaSR) on protein tyrosine phosphorylation in boar spermatozoa under capacitating conditions. To do this, boar spermatozoa were incubated in Tyrode's complete medium for 4 hr and the specific inhibitor of the CaSR, NPS2143, was used. Also, to study the possible mechanism(s) by which this receptor exerts its function, spermatozoa were incubated in the presence of specific inhibitors of the 3-phosphoinositide dependent protein kinase 1 (PDK1) and protein kinase A (PKA). Treatment with NPS2143, GSK2334470, an inhibitor of PDK1 and H-89, an inhibitor of PKA separately induced an increase in tyrosine phosphorylation of 18 and 32 kDa proteins, a decrease in the serine/threonine phosphorylation of the PKA substrates together with a drop in sperm motility and viability. The present work proposes a new signalling pathway of the CaSR, mediated by PDK1 and PKA in boar spermatozoa under capacitating conditions. Our results show that the inhibition of the CaSR induces the inhibition of PDK1 that blocks PKA activity resulting in a rise in tyrosine phosphorylation of p18 and p32 proteins. This novel signalling pathway has not been described before and could be crucial to understand boar sperm capacitation within the female reproductive tract.
Collapse
Affiliation(s)
- Beatriz Macías-García
- Research Institute of Biotechnology in Livestock and Cynegetic (INBIO G+C), University of Extremadura, Cáceres, Spain.,Animal Medicine Department, Faculty of Veterinary Sciences, University of Extremadura, Cáceres, Spain
| | - Luis J García-Marín
- Research Institute of Biotechnology in Livestock and Cynegetic (INBIO G+C), University of Extremadura, Cáceres, Spain.,Department of Physiology, Faculty of Veterinary Sciences, University of Extremadura, Cáceres, Spain
| | - María J Bragado
- Research Institute of Biotechnology in Livestock and Cynegetic (INBIO G+C), University of Extremadura, Cáceres, Spain.,Department of Biochemistry and Molecular Biology and Genetics, Faculty of Veterinary Sciences, University of Extremadura, Cáceres, Spain
| | - Lauro González-Fernández
- Research Institute of Biotechnology in Livestock and Cynegetic (INBIO G+C), University of Extremadura, Cáceres, Spain.,Department of Biochemistry and Molecular Biology and Genetics, Faculty of Veterinary Sciences, University of Extremadura, Cáceres, Spain
| |
Collapse
|
35
|
TLR4 counteracts BVRA signaling in human leukocytes via differential regulation of AMPK, mTORC1 and mTORC2. Sci Rep 2019; 9:7020. [PMID: 31065010 PMCID: PMC6504875 DOI: 10.1038/s41598-019-43347-8] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2018] [Accepted: 04/16/2019] [Indexed: 12/30/2022] Open
Abstract
TLR4 is implicated in diseases associated with chronic low-grade inflammation, yet homeostatic signaling mechanisms that prevent and/or are affected by chronic TLR4 activation are largely uncharacterized. We recently reported that LPS/TLR4 activates in human leukocytes signaling intermediates (SI), abbreviated TLR4-SI, which include mTORC1-specific effectors and targets, and that leukocytes of patients with T2D or after cardiopulmonary bypass (CPB) expressed similar SI. Extending these previous findings, here we show that TLR4-SI expression post-CPB was associated with low serum bilirubin and reduced preoperative expression of biliverdin reductase A (BVRA), the enzyme that converts biliverdin to bilirubin, in patient’s leukocytes. Biliverdin inhibited TLR4 signaling in leukocytes and triggered phosphorylation of mTORC2-specific targets, including Akt, PKCζ, AMPKα-LKB1-TSC1/2, and their association with BVRA. Torin, PP242, and a PKCζ inhibitory peptide, but not rapamycin, prevented these biliverdin-induced responses and TLR4 inhibition. In contrast, LPS/TLR4 triggered decreases in BVRA, AMPKα and PKCζ expression, and an increase in haptoglobin, a heme binding protein, in leukocytes in vivo and in vitro, indicating that activated TLR4 may suppress biliverdin/BVRA signaling. Significantly, compared to non-diabetics, BVRA and PKCζ expression was low and haptoglobin was high in T2D patients leukocytes. Sustained TLR4 activation may deregulate homeostatic anti-inflammatory BVRA/mTORC2 signaling and thereby contribute to chronic inflammatory diseases.
Collapse
|
36
|
AS1949490, an inhibitor of 5′-lipid phosphatase SHIP2, promotes protein kinase C-dependent stabilization of brain-derived neurotrophic factor mRNA in cultured cortical neurons. Eur J Pharmacol 2019; 851:69-79. [DOI: 10.1016/j.ejphar.2019.02.003] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2018] [Revised: 02/04/2019] [Accepted: 02/08/2019] [Indexed: 12/11/2022]
|
37
|
García-Gil A, Galán-Enríquez CS, Pérez-López A, Nava P, Alpuche-Aranda C, Ortiz-Navarrete V. SopB activates the Akt-YAP pathway to promote Salmonella survival within B cells. Virulence 2019; 9:1390-1402. [PMID: 30103648 PMCID: PMC6177241 DOI: 10.1080/21505594.2018.1509664] [Citation(s) in RCA: 31] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023] Open
Abstract
B cells are a target of Salmonella infection, allowing bacteria survival without inducing pyroptosis. This event is due to downregulation of Nlrc4 expression and lack of inflammasome complex activation, which impairs the secretion of IL-1β. YAP phosphorylation is required for downregulation of Nlrc4 in B cells during Salmonella infection; however, the microorganism’s mechanisms underlying the inhibition of the NLRC4 inflammasome in B cells are not fully understood. Our findings demonstrate that the Salmonella effector SopB triggers a signaling cascade involving PI3K, PDK1 and mTORC2 that activates Akt with consequent phosphorylation of YAP. When we deleted sopB in Salmonella, infected B cells that lack Rictor, or inhibited the signaling cascade using a pharmacological approach, we were able to restore the function of the NLRC4 inflammasome in B cells and the ability to control the infection. Furthermore, B cells from infected mice exhibited activation of Akt and YAP phosphorylation, suggesting that Salmonella also triggers this pathway in vivo. In summary, our data demonstrate that the Salmonella effector inositide phosphate phosphatase SopB triggers the PI3K-Akt-YAP pathway to inhibit the NLRC4 inflammasome in B cells. This study provides further evidence that Salmonella triggers cellular mechanisms in B lymphocytes to manipulate the host environment by turning it into a survival niche to establish a successful infection.
Collapse
Affiliation(s)
- Abraham García-Gil
- a Departamento de Biomedicina Molecular , Centro de Investigación y de Estudios Avanzados del Instituto Politécnico Nacional , Ciudad de México , México
| | - Carlos Samuel Galán-Enríquez
- a Departamento de Biomedicina Molecular , Centro de Investigación y de Estudios Avanzados del Instituto Politécnico Nacional , Ciudad de México , México
| | - Araceli Pérez-López
- b Department of Pediatrics , University of California San Diego , San Diego , CA , USA
| | - Porfirio Nava
- c Departamento de Fisiología , Biofísica y Neurociencias, Centro de Investigación y de Estudios Avanzados del Instituto Politécnico Nacional , Ciudad de México , México
| | - Celia Alpuche-Aranda
- d Centro de Investigación Sobre Enfermedades Infecciosa , Instituto Nacional de Salud Pública, SSA , Cuernavaca , México
| | - Vianney Ortiz-Navarrete
- a Departamento de Biomedicina Molecular , Centro de Investigación y de Estudios Avanzados del Instituto Politécnico Nacional , Ciudad de México , México
| |
Collapse
|
38
|
Liao B, Peng L, Zhou J, Mo H, Zhao J, Yang Z, Guo X, Zhang P, Zhang X, Zhu Z. Synthesis and Activity Evaluation of Nasopharyngeal Carcinoma Inhibitors Based on 6-(Pyrimidin-4-yl)-1H-indazole. Chem Biodivers 2019; 16:e1800598. [PMID: 30788913 DOI: 10.1002/cbdv.201800598] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2018] [Accepted: 02/18/2019] [Indexed: 11/07/2022]
Abstract
Human nasopharyngeal carcinoma is a common head and neck malignancy with high incidence in Southeast Asia and Southern China. It is necessary to develop safe, effective and inexpensive anticancer agents to improve the therapeutics of patients with nasopharyngeal carcinoma. A series of small molecular compounds based on 6-(pyrimidin-4-yl)-1H-indazole were synthesized and evaluated for antiproliferative activities against human nasopharyngeal carcinoma cell lines SUNE1. Compounds 6b, 6c, 6e and 6l showed potent antiproliferative activities similar to positive control drug cisplatin in vitro with lower nephrotoxicity than it. N-[4-(1H-Indazol-6-yl)pyrimidin-2-yl]benzene-1,3-diamine (6l) was selected for further study. It was found that 6l induced mitochondria-mediated apoptosis and G2 /M phase arrest in SUNE1 cells. Furthermore, compound 6l at 10 mg/kg can suppress the growth of an implanted SUNE1 xenograft with a TGI% (tumor growth inhibition) value of 50 % and did not cause serious side effects in BALB/c nude mice. This study suggests that 6-(pyrimidin-4-yl)-1H-indazole derivatives are a series of small molecule compounds with anti-nasopharyngeal carcinoma activities.
Collapse
Affiliation(s)
- Bohong Liao
- Integrated Hospital of Traditional Chinese Medicine, Southern Medical University, 13# Shiliugang Road, Haizhu District, Guangzhou, 510315, P. R. China
| | - Lingrong Peng
- Department of Radiology, The Third Affiliated Hospital of Sun Yat-Sen University, Guangzhou, 510630, P. R. China
| | - Jin Zhou
- Integrated Hospital of Traditional Chinese Medicine, Southern Medical University, 13# Shiliugang Road, Haizhu District, Guangzhou, 510315, P. R. China
| | - Huiting Mo
- Integrated Hospital of Traditional Chinese Medicine, Southern Medical University, 13# Shiliugang Road, Haizhu District, Guangzhou, 510315, P. R. China
| | - Jialan Zhao
- Integrated Hospital of Traditional Chinese Medicine, Southern Medical University, 13# Shiliugang Road, Haizhu District, Guangzhou, 510315, P. R. China
| | - Zike Yang
- Integrated Hospital of Traditional Chinese Medicine, Southern Medical University, 13# Shiliugang Road, Haizhu District, Guangzhou, 510315, P. R. China
| | - Xiaowen Guo
- Integrated Hospital of Traditional Chinese Medicine, Southern Medical University, 13# Shiliugang Road, Haizhu District, Guangzhou, 510315, P. R. China
| | - Peiquan Zhang
- Guangdong Provincial Key Laboratory of New Drug Screening, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou, 510515, P. R. China
| | - Xin Zhang
- Integrated Hospital of Traditional Chinese Medicine, Southern Medical University, 13# Shiliugang Road, Haizhu District, Guangzhou, 510315, P. R. China
| | - Zhibo Zhu
- Integrated Hospital of Traditional Chinese Medicine, Southern Medical University, 13# Shiliugang Road, Haizhu District, Guangzhou, 510315, P. R. China
| |
Collapse
|
39
|
Heberle AM, Razquin Navas P, Langelaar-Makkinje M, Kasack K, Sadik A, Faessler E, Hahn U, Marx-Stoelting P, Opitz CA, Sers C, Heiland I, Schäuble S, Thedieck K. The PI3K and MAPK/p38 pathways control stress granule assembly in a hierarchical manner. Life Sci Alliance 2019; 2:2/2/e201800257. [PMID: 30923191 PMCID: PMC6441495 DOI: 10.26508/lsa.201800257] [Citation(s) in RCA: 44] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2018] [Revised: 03/06/2019] [Accepted: 03/07/2019] [Indexed: 01/11/2023] Open
Abstract
PI3K and p38 act in a hierarchical manner to enhance mTORC1 activity and stress granule formation; although PI3K is the main driver, the impact of p38 gets apparent as PI3K activity declines. All cells and organisms exhibit stress-coping mechanisms to ensure survival. Cytoplasmic protein-RNA assemblies termed stress granules are increasingly recognized to promote cellular survival under stress. Thus, they might represent tumor vulnerabilities that are currently poorly explored. The translation-inhibitory eIF2α kinases are established as main drivers of stress granule assembly. Using a systems approach, we identify the translation enhancers PI3K and MAPK/p38 as pro-stress-granule-kinases. They act through the metabolic master regulator mammalian target of rapamycin complex 1 (mTORC1) to promote stress granule assembly. When highly active, PI3K is the main driver of stress granules; however, the impact of p38 becomes apparent as PI3K activity declines. PI3K and p38 thus act in a hierarchical manner to drive mTORC1 activity and stress granule assembly. Of note, this signaling hierarchy is also present in human breast cancer tissue. Importantly, only the recognition of the PI3K-p38 hierarchy under stress enabled the discovery of p38’s role in stress granule formation. In summary, we assign a new pro-survival function to the key oncogenic kinases PI3K and p38, as they hierarchically promote stress granule formation.
Collapse
Affiliation(s)
- Alexander Martin Heberle
- Laboratory of Pediatrics, Section Systems Medicine of Metabolism and Signaling, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| | - Patricia Razquin Navas
- Laboratory of Pediatrics, Section Systems Medicine of Metabolism and Signaling, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands.,Department for Neuroscience, School of Medicine and Health Sciences, Carl von Ossietzky University Oldenburg, Oldenburg, Germany
| | - Miriam Langelaar-Makkinje
- Laboratory of Pediatrics, Section Systems Medicine of Metabolism and Signaling, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| | - Katharina Kasack
- Laboratory of Molecular Tumor Pathology, Institute of Pathology, Charité Universitätsmedizin Berlin, Berlin, Germany.,German Cancer Consortium (DKTK), Partner Site Berlin, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Ahmed Sadik
- Brain Cancer Metabolism Group, German Cancer Research Center (DKFZ), German Cancer Consortium (DKTK), Heidelberg, Germany.,Faculty of Bioscience, Heidelberg University, Heidelberg, Germany
| | - Erik Faessler
- Jena University Language and Information Engineering Lab, Friedrich-Schiller-University Jena, Jena, Germany
| | - Udo Hahn
- Jena University Language and Information Engineering Lab, Friedrich-Schiller-University Jena, Jena, Germany
| | - Philip Marx-Stoelting
- German Federal Institute for Risk Assessment, Strategies for Toxicological Assessment, Experimental Toxicology and ZEBET, German Centre for the Protection of Laboratory Animals (Bf3R), Berlin, Germany
| | - Christiane A Opitz
- Brain Cancer Metabolism Group, German Cancer Research Center (DKFZ), German Cancer Consortium (DKTK), Heidelberg, Germany.,Neurology Clinic and National Center for Tumor Diseases, University Hospital of Heidelberg, Heidelberg, Germany
| | - Christine Sers
- Laboratory of Molecular Tumor Pathology, Institute of Pathology, Charité Universitätsmedizin Berlin, Berlin, Germany.,German Cancer Consortium (DKTK), Partner Site Berlin, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Ines Heiland
- Faculty of Bioscience, Fisheries and Economics, Department of Arctic and Marine Biology, UiT The Arctic University of Norway, Tromsø, Norway
| | - Sascha Schäuble
- Jena University Language and Information Engineering Lab, Friedrich-Schiller-University Jena, Jena, Germany .,Systems Biology and Bioinformatics, Leibniz Institute for Natural Product Research and Infection Biology, Hans Knöll Institute, Jena, Germany
| | - Kathrin Thedieck
- Laboratory of Pediatrics, Section Systems Medicine of Metabolism and Signaling, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands .,Department for Neuroscience, School of Medicine and Health Sciences, Carl von Ossietzky University Oldenburg, Oldenburg, Germany.,Institute of Biochemistry and Center for Molecular Biosciences Innsbruck, University of Innsbruck, Innsbruck, Austria
| |
Collapse
|
40
|
Neddylation inhibitor MLN4924 suppresses cilia formation by modulating AKT1. Protein Cell 2019; 10:726-744. [PMID: 30850948 PMCID: PMC6776484 DOI: 10.1007/s13238-019-0614-3] [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: 12/24/2018] [Accepted: 01/24/2019] [Indexed: 11/25/2022] Open
Abstract
The primary cilium is a microtubule-based sensory organelle. The molecular mechanism that regulates ciliary dynamics remains elusive. Here, we report an unexpected finding that MLN4924, a small molecule inhibitor of NEDD8-activating enzyme (NAE), blocks primary ciliary formation by inhibiting synthesis/assembly and promoting disassembly. This is mainly mediated by MLN4924-induced phosphorylation of AKT1 at Ser473 under serum-starved, ciliary-promoting conditions. Indeed, pharmaceutical inhibition (by MK2206) or genetic depletion (via siRNA) of AKT1 rescues MLN4924 effect, indicating its causal role. Interestingly, pAKT1-Ser473 activity regulates both ciliary synthesis/assembly and disassembly in a MLN4924 dependent manner, whereas pAKT-Thr308 determines the ciliary length in MLN4924-independent but VHL-dependent manner. Finally, MLN4924 inhibits mouse hair regrowth, a process requires ciliogenesis. Collectively, our study demonstrates an unexpected role of a neddylation inhibitor in regulation of ciliogenesis via AKT1, and provides a proof-of-concept for potential utility of MLN4924 in the treatment of human diseases associated with abnormal ciliogenesis.
Collapse
|
41
|
Leroux AE, Gross LZF, Sacerdoti M, Biondi RM. Allosteric Regulation of Protein Kinases Downstream of PI3-Kinase Signalling. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2019; 1163:279-311. [PMID: 31707708 DOI: 10.1007/978-981-13-8719-7_12] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Allostery is a basic principle that enables proteins to process and transmit cellular information. Protein kinases evolved allosteric mechanisms to transduce cellular signals to downstream signalling components or effector molecules. Protein kinases catalyse the transfer of the terminal phosphate from ATP to protein substrates upon specific stimuli. Protein kinases are targets for the development of small molecule inhibitors for the treatment of human diseases. Drug development has focussed on ATP-binding site, while there is increase interest in the development of drugs targeting alternative sites, i.e. allosteric sites. Here, we review the mechanism of regulation of protein kinases, which often involve the allosteric modulation of the ATP-binding site, enhancing or inhibiting activity. We exemplify the molecular mechanism of allostery in protein kinases downstream of PI3-kinase signalling with a focus on phosphoinositide-dependent protein kinase 1 (PDK1), a model kinase where small compounds can allosterically modulate the conformation of the kinase bidirectionally.
Collapse
Affiliation(s)
- Alejandro E Leroux
- Instituto de Investigación en Biomedicina de Buenos Aires (IBioBA) - CONICET - Partner Institute of the Max Planck Society, Buenos Aires, Argentina
| | - Lissy Z F Gross
- Instituto de Investigación en Biomedicina de Buenos Aires (IBioBA) - CONICET - Partner Institute of the Max Planck Society, Buenos Aires, Argentina
| | - Mariana Sacerdoti
- Instituto de Investigación en Biomedicina de Buenos Aires (IBioBA) - CONICET - Partner Institute of the Max Planck Society, Buenos Aires, Argentina
| | - Ricardo M Biondi
- Instituto de Investigación en Biomedicina de Buenos Aires (IBioBA) - CONICET - Partner Institute of the Max Planck Society, Buenos Aires, Argentina.
- Department of Internal Medicine I, Universitätsklinikum Frankfurt, Frankfurt, Germany.
- DKTK German Cancer Consortium (DKTK), Frankfurt, Germany.
- German Cancer Research Center (DKFZ), Heidelberg, Germany.
| |
Collapse
|
42
|
Ctortecka C, Palve V, Kuenzi BM, Fang B, Sumi NJ, Izumi V, Novakova S, Kinose F, Remsing Rix LL, Haura EB, Koomen JM, Rix U. Functional Proteomics and Deep Network Interrogation Reveal a Complex Mechanism of Action of Midostaurin in Lung Cancer Cells. Mol Cell Proteomics 2018; 17:2434-2447. [PMID: 30217950 PMCID: PMC6283294 DOI: 10.1074/mcp.ra118.000713] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2018] [Revised: 08/16/2018] [Indexed: 12/21/2022] Open
Abstract
Lung cancer is associated with high prevalence and mortality, and despite significant successes with targeted drugs in genomically defined subsets of lung cancer and immunotherapy, the majority of patients currently does not benefit from these therapies. Through a targeted drug screen, we found the recently approved multi-kinase inhibitor midostaurin to have potent activity in several lung cancer cells independent of its intended target, PKC, or a specific genomic marker. To determine the underlying mechanism of action we applied a layered functional proteomics approach and a new data integration method. Using chemical proteomics, we identified multiple midostaurin kinase targets in these cells. Network-based integration of these targets with quantitative tyrosine and global phosphoproteomics data using protein-protein interactions from the STRING database suggested multiple targets are relevant for the mode of action of midostaurin. Subsequent functional validation using RNA interference and selective small molecule probes showed that simultaneous inhibition of TBK1, PDPK1 and AURKA was required to elicit midostaurin's cellular effects. Immunoblot analysis of downstream signaling nodes showed that combined inhibition of these targets altered PI3K/AKT and cell cycle signaling pathways that in part converged on PLK1. Furthermore, rational combination of midostaurin with the potent PLK1 inhibitor BI2536 elicited strong synergy. Our results demonstrate that combination of complementary functional proteomics approaches and subsequent network-based data integration can reveal novel insight into the complex mode of action of multi-kinase inhibitors, actionable targets for drug discovery and cancer vulnerabilities. Finally, we illustrate how this knowledge can be used for the rational design of synergistic drug combinations with high potential for clinical translation.
Collapse
Affiliation(s)
- Claudia Ctortecka
- Department of Drug Discovery, H. Lee Moffitt Cancer Center & Research Institute, Tampa, Florida 33612
| | - Vinayak Palve
- Department of Drug Discovery, H. Lee Moffitt Cancer Center & Research Institute, Tampa, Florida 33612
| | - Brent M Kuenzi
- Department of Drug Discovery, H. Lee Moffitt Cancer Center & Research Institute, Tampa, Florida 33612; Cancer Biology PhD Program, University of South Florida, Tampa, Florida 33620
| | - Bin Fang
- Proteomics and Metabolomics Core, H. Lee Moffitt Cancer Center & Research Institute, Tampa, Florida 33612
| | - Natalia J Sumi
- Department of Drug Discovery, H. Lee Moffitt Cancer Center & Research Institute, Tampa, Florida 33612; Cancer Biology PhD Program, University of South Florida, Tampa, Florida 33620
| | - Victoria Izumi
- Proteomics and Metabolomics Core, H. Lee Moffitt Cancer Center & Research Institute, Tampa, Florida 33612
| | - Silvia Novakova
- Department of Drug Discovery, H. Lee Moffitt Cancer Center & Research Institute, Tampa, Florida 33612
| | - Fumi Kinose
- Department of Thoracic Oncology, H. Lee Moffitt Cancer Center & Research Institute, Tampa, Florida 33612
| | - Lily L Remsing Rix
- Department of Drug Discovery, H. Lee Moffitt Cancer Center & Research Institute, Tampa, Florida 33612
| | - Eric B Haura
- Department of Thoracic Oncology, H. Lee Moffitt Cancer Center & Research Institute, Tampa, Florida 33612
| | - John Matthew Koomen
- Department of Molecular Oncology, H. Lee Moffitt Cancer Center & Research Institute, Tampa, Florida 33612
| | - Uwe Rix
- Department of Drug Discovery, H. Lee Moffitt Cancer Center & Research Institute, Tampa, Florida 33612.
| |
Collapse
|
43
|
Xing T, Hass DT, Zhang SS, Barnstable CJ. The 3-Phosphoinositide-Dependent Protein Kinase 1 Inhibits Rod Photoreceptor Development. Front Cell Dev Biol 2018; 6:134. [PMID: 30364083 PMCID: PMC6191476 DOI: 10.3389/fcell.2018.00134] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2018] [Accepted: 09/24/2018] [Indexed: 01/30/2023] Open
Abstract
The transition of rod precursor cells to post-mitotic rod photoreceptors can be promoted by extrinsic factors such as insulin-like growth factor 1 (IGF-1), which regulates phosphatidylinositide concentration, and consequently the 3-phosphoinositide-dependent protein kinase-1 (PDPK-1). PDPK-1 is a 63 kDa cytoplasmic kinase that controls cell proliferation and differentiation. In the mouse retina, PDPK-1 and its phosphorylated derivative p-PDPK-1 (Ser241), showed peak expression during the first postnatal (PN) day with a substantial decline by PN7 and in the adult retina. Though initially widely distributed among cell types, PDPK-1 expression decreased first in the inner retina and later in the outer retina. When PDPK-1 is inhibited in neonatal retinal explants by BX795, there is a robust increase in rod photoreceptor numbers. The increase in rods depended on the activity of PKC, as BX795 had no effect when PKC is inhibited. Inhibition of PDPK-1-dependent kinases, such as P70-S6K, but not others, such as mTORC-1, stimulated rod development. The P70-S6K-dependent increase in rods appears to be correlated with phosphorylation of Thr252 and not at Thr389, a substrate of mTORC-1. This pathway is also inactive while PKC activity is inhibited. We also found that inhibition of the kinase mTORC-2, also stimulated by insulin activity, similarly increased rod formation, and this effect appears to be independent of PKC activity. This may represent a novel intracellular signaling pathway that also stimulates photoreceptor development. Consistent with previous studies, stimulation of STAT3 activity is sufficient to prevent any PDPK-1, P70-S6K, or mTORC2-dependent increase in rods. Together the data indicate that PDPK-1 and other intrinsic kinases downstream of IGF-1 are key regulators of rod photoreceptor formation.
Collapse
Affiliation(s)
- Tiaosi Xing
- Department of Anatomy and Cell Biology, East Carolina University, Greenville, NC, United States
| | - Daniel T Hass
- Department of Neural and Behavioral Sciences, Penn State College of Medicine, Hershey, PA, United States
| | - Samuel S Zhang
- Department of Neural and Behavioral Sciences, Penn State College of Medicine, Hershey, PA, United States
| | - Colin J Barnstable
- Department of Neural and Behavioral Sciences, Penn State College of Medicine, Hershey, PA, United States
| |
Collapse
|
44
|
Ling Y, Zhang Z, Zhang H, Huang Z. Protein Kinase Inhibitors as Therapeutic Drugs in AML: Advances and Challenges. Curr Pharm Des 2018; 23:4303-4310. [PMID: 28671056 PMCID: PMC6302345 DOI: 10.2174/1381612823666170703164114] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2017] [Revised: 05/13/2017] [Accepted: 05/18/2017] [Indexed: 12/28/2022]
Abstract
Acute myeloid leukemia (AML) is a malignant blood disorder and the cure rate has been remarkably improved over the past decade. However, recurrent or refractory leu-kemia remains the major problem of the AML and no clearly effective therapy has been es-tablished so far. Traditional treatments such as chemotherapy and hematopoietic stem cell transplantation are both far dissatisfying the patients partly for their individual variety. Be-sides, conventional treatments usually have many side effects to result in poor prognosis. Therefore, an urgent need is necessary to update therapies of AML. To date, protein kinase inhibitors as new drugs offer hope for AML treatment and many of them are on clinical tri-als. Here, this review will provide a brief summary of protein kinase inhibitors investigated in AML thus far, mainly including tyrosine protein kinase inhibitors and serine/threonine kinase inhibitors. We also presented the sketch of signal pathways involving protein kinase inhibitors, as well as discussed the clinical applications and the challenges of inhibitors in AML treatment
Collapse
Affiliation(s)
- Yuan Ling
- Guangdong Provincial Key Laboratory of Medical Molecular Diagnostics, Guangdong Medical University, Dongguan, P.R. China.,China-America Cancer Research Institute, Dongguan Key Laboratory of Epigenetics, Institute of Clinical Laboratory Medicine, Dongguan Scientific Research Center, Guangdong Medical University, Dongguan 523808, P.R. China
| | - Zikang Zhang
- Guangdong Provincial Key Laboratory of Medical Molecular Diagnostics, Guangdong Medical University, Dongguan, P.R. China.,China-America Cancer Research Institute, Dongguan Key Laboratory of Epigenetics, Institute of Clinical Laboratory Medicine, Dongguan Scientific Research Center, Guangdong Medical University, Dongguan 523808, P.R. China
| | - Hua Zhang
- Guangdong Provincial Key Laboratory of Medical Molecular Diagnostics, Guangdong Medical University, Dongguan, P.R. China.,China-America Cancer Research Institute, Dongguan Key Laboratory of Epigenetics, Institute of Clinical Laboratory Medicine, Dongguan Scientific Research Center, Guangdong Medical University, Dongguan 523808, P.R. China
| | - Zunnan Huang
- Guangdong Provincial Key Laboratory of Medical Molecular Diagnostics, Guangdong Medical University, Dongguan, P.R. China.,China-America Cancer Research Institute, Dongguan Key Laboratory of Epigenetics, Institute of Clinical Laboratory Medicine, Dongguan Scientific Research Center, Guangdong Medical University, Dongguan 523808, P.R. China
| |
Collapse
|
45
|
Yang C, Huang X, Liu H, Xiao F, Wei J, You L, Qian W. PDK1 inhibitor GSK2334470 exerts antitumor activity in multiple myeloma and forms a novel multitargeted combination with dual mTORC1/C2 inhibitor PP242. Oncotarget 2018; 8:39185-39197. [PMID: 28402933 PMCID: PMC5503605 DOI: 10.18632/oncotarget.16642] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2017] [Accepted: 03/06/2017] [Indexed: 12/17/2022] Open
Abstract
A deeper understanding of the complex pathogenesis of multiple myeloma (MM) continues to lead to novel therapeutic approaches. Prior studies suggest that 3-phosphoinositide-dependent kinase 1 (PDK1) is expressed and active, acting as a crucial regulator of molecules that are essential for myelomagenesis. In the present study, we show that GSK2334470 (GSK-470), a novel and highly specific inhibitor of PDK1, induces potent cytotoxicity in MM cell lines including Dexamethasone-resistant cell line, but not in human normal cells. Insulin-like growth factor-1 could not rescue GSK-470-induced cell death. Moreover, GSK-470 down-modulates phosphor-PDK1, thereby inhibiting downstream phosphor-AKT at Thr308 and mTOR complex 1 (mTORC1) activity. However, GSK-470 could not affect mTORC2 activity and phosphor-AKT at Ser473. RPMI 8226 and OPM-2 cells with low expression of PTEN show relative resistant to GSK-470. Knockout of PTEN by shRNA resulted in a partial reversion of GSK-470-mediated growth inhibition, whereas overexpression of PTEN enhanced myeloma cell sensitivity to GSK-470, suggesting that the sensitivity to GSK-470 is correlated with PTEN expression statue in MM cells. Combining PP242, a dual mTORC1/C2 inhibitor, with GSK-470, had greater antimyeloma activity than either one alone in vitro and in MM xenograft model established in immunodeficient mice. In particular, this combination was able to result in a complete inhibition of mTORC1/C2 and full activity of AKT. Together, these findings raise the possibility that combining PDK1 antagonist GSK-470 with mTORC1/C2 inhibitors may represent a novel strategy against MM including drug-resistant myeloma, regardless of PTEN expression status.
Collapse
Affiliation(s)
- Chunmei Yang
- Institute of Hematology, the First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, 310003, P.R. China
| | - Xianbo Huang
- Institute of Hematology, the First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, 310003, P.R. China
| | - Hui Liu
- Institute of Hematology, the First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, 310003, P.R. China
| | - Feng Xiao
- Institute of Hematology, the First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, 310003, P.R. China
| | - Jueying Wei
- Institute of Hematology, the First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, 310003, P.R. China
| | - Liangshun You
- Institute of Hematology, the First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, 310003, P.R. China
| | - Wenbin Qian
- Institute of Hematology, the First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, 310003, P.R. China
| |
Collapse
|
46
|
Manne BK, Münzer P, Badolia R, Walker-Allgaier B, Campbell RA, Middleton E, Weyrich AS, Kunapuli SP, Borst O, Rondina MT. PDK1 governs thromboxane generation and thrombosis in platelets by regulating activation of Raf1 in the MAPK pathway. J Thromb Haemost 2018; 16:1211-1225. [PMID: 29575487 PMCID: PMC5984143 DOI: 10.1111/jth.14005] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2017] [Indexed: 01/02/2023]
Abstract
Essentials Phosphoinositide 3-kinase and MAPK pathways crosstalk via PDK1. PDK1 is required for adenosine diphosphate-induced platelet activation and thromboxane generation. PDK1 regulates RAF proto-oncogene Ser/Thr kinase (Raf1) activation in the MAPK pathway. Genetic ablation of PDK1 protects against platelet-dependent thrombosis in vivo. SUMMARY Background Platelets are dynamic effector cells with functions that span hemostatic, thrombotic and inflammatory continua. Phosphoinositide-dependent protein kinase 1 (PDK1) regulates protease-activated receptor 4-induced platelet activation and thrombus formation through glycogen synthase kinase3β. However, whether PDK1 also signals through the ADP receptor and its functional importance in vivo remain unknown. Objective To establish the mechanism of PDK1 in ADP-induced platelet activation and thrombosis. Methods We assessed the role of PDK1 on 2MeSADP-induced platelet activation by measuring aggregation, thromboxane generation and phosphorylation events in the presence of BX-795, which inhibits PDK1, or by using platelet-specific PDK1 knockout mice and performing western blot analysis. PDK1 function in thrombus formation was assessed with an in vivo pulmonary embolism model. Results PDK1 inhibition with BX-795 reduced 2-methylthio-ADP (2MeSADP)-induced aggregation of human and murine platelets by abolishing thromboxane generation. Similar results were observed in pdk1-/- mice. PDK1 was also necessary for the phosphorylation of mitogen-activated protein kinase kinase 1/2 (MEK1/2), extracellular signal-regulated kinase 1/2, and cytosolic phospholipase A2, indicating that PDK1 regulates an upstream kinase in the mitogen-activated protein kinase (MAPK) pathway. We next determined that this upstream kinase is Raf-1, a serine/threonine kinase that is necessary for the phosphorylation of MEK1/2, as pharmacological inhibition and genetic ablation of PDK1 were sufficient to prevent Raf1 phosphorylation. Furthermore, in vivo inhibition or genetic ablation of PDK1 protected mice from collagen/epinephrine-induced pulmonary embolism. Conclusion PDK1 governs thromboxane generation and thrombosis in platelets that are stimulated with 2MeSADP by regulating activation of the MAPK pathway.
Collapse
Affiliation(s)
- Bhanu Kanth Manne
- Department of Internal Medicine, Molecular Medicine Program, University of Utah, Salt Lake City, UT, 84112 USA
| | - Patrick Münzer
- Department of Cardiology and Cardiovascular Medicine, University of Tübingen, Tübingen, 72076 Germany
| | - Rachit Badolia
- Sol Sherry Thrombosis Research Center, Temple University School of Medicine, Philadelphia, PA, 19140 USA
| | - Britta Walker-Allgaier
- Department of Cardiology and Cardiovascular Medicine, University of Tübingen, Tübingen, 72076 Germany
| | - Robert A Campbell
- Department of Internal Medicine, Molecular Medicine Program, University of Utah, Salt Lake City, UT, 84112 USA
| | - Elizabeth Middleton
- Department of Internal Medicine, Molecular Medicine Program, University of Utah, Salt Lake City, UT, 84112 USA
| | - Andrew S Weyrich
- Department of Internal Medicine, Molecular Medicine Program, University of Utah, Salt Lake City, UT, 84112 USA
| | - Satya P Kunapuli
- Sol Sherry Thrombosis Research Center, Temple University School of Medicine, Philadelphia, PA, 19140 USA
| | - Oliver Borst
- Department of Cardiology and Cardiovascular Medicine, University of Tübingen, Tübingen, 72076 Germany
| | - Matthew T. Rondina
- Department of Internal Medicine, Molecular Medicine Program, University of Utah, Salt Lake City, UT, 84112 USA
- Department of Internal Medicine, GRECC, George E. Wahlen VAMC, Salt Lake City, UT, 84148
| |
Collapse
|
47
|
Spatial alterations of De Novo purine biosynthetic enzymes by Akt-independent PDK1 signaling pathways. PLoS One 2018; 13:e0195989. [PMID: 29668719 PMCID: PMC5905998 DOI: 10.1371/journal.pone.0195989] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2017] [Accepted: 04/04/2018] [Indexed: 11/19/2022] Open
Abstract
A macromolecular complex of the enzymes involved in human de novo purine biosynthesis, the purinosome, has been shown to consist of a core assembly to regulate the metabolic activity of the pathway. However, it remains elusive whether the core assembly itself can be selectively controlled in the cytoplasm without promoting the purinosome. Here, we reveal that pharmacological inhibition of the cytoplasmic activity of 3-phosphoinositide-dependent protein kinase 1 (PDK1) selectively promotes the formation of the core assembly, but not the purinosome, in cancer cells. However, alternative signaling cascades that are associated with the plasma membrane-bound PDK1 activity, including Akt-mediated cascades, regulate neither the core assembly nor the purinosome in our conditions. Along with immunofluorescence microscopy and a knock-down study against PDK1 using small interfering RNAs, we reveal that cytoplasmic PDK1-associated signaling pathways regulate subcellular colocalization of three enzymes that form the core assembly of the purinosome in an Akt-independent manner. Collectively, this study reveals a new mode of compartmentalization of purine biosynthetic enzymes controlled by spatially resolved signaling pathways.
Collapse
|
48
|
Clement E, Inuzuka H, Nihira NT, Wei W, Toker A. Skp2-dependent reactivation of AKT drives resistance to PI3K inhibitors. Sci Signal 2018. [PMID: 29535262 DOI: 10.1126/scisignal.aao3810] [Citation(s) in RCA: 44] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
The PI3K-AKT kinase signaling pathway is frequently deregulated in human cancers, particularly breast cancer, where amplification and somatic mutations of PIK3CA occur with high frequency in patients. Numerous small-molecule inhibitors targeting both PI3K and AKT are under clinical evaluation, but dose-limiting toxicities and the emergence of resistance limit therapeutic efficacy. Various resistance mechanisms to PI3K inhibitors have been identified, including de novo mutations, feedback activation of AKT, or cross-talk pathways. We found a previously unknown resistance mechanism to PI3K pathway inhibition that results in AKT rebound activation. In a subset of triple-negative breast cancer cell lines, treatment with a PI3K inhibitor or depletion of PIK3CA expression ultimately promoted AKT reactivation in a manner dependent on the E3 ubiquitin ligase Skp2, the kinases IGF-1R (insulin-like growth factor 1 receptor) and PDK-1 (phosphoinositide-dependent kinase-1), and the cell growth and metabolism-regulating complex mTORC2 (mechanistic target of rapamycin complex 2), but was independent of PI3K activity or PIP3 production. Resistance to PI3K inhibitors correlated with the increased abundance of Skp2, ubiquitylation of AKT, cell proliferation in culture, and xenograft tumor growth in mice. These findings reveal a ubiquitin signaling feedback mechanism by which PI3K inhibitor resistance may emerge in aggressive breast cancer cells.
Collapse
Affiliation(s)
- Emilie Clement
- Department of Pathology, Medicine and Cancer Center, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02215, USA
| | - Hiroyuki Inuzuka
- Department of Pathology, Medicine and Cancer Center, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02215, USA
| | - Naoe T Nihira
- Department of Pathology, Medicine and Cancer Center, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02215, USA
| | - Wenyi Wei
- Department of Pathology, Medicine and Cancer Center, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02215, USA
| | - Alex Toker
- Department of Pathology, Medicine and Cancer Center, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02215, USA. .,Ludwig Center at Harvard, Harvard Medical School, Boston, MA 02215, USA
| |
Collapse
|
49
|
Maegawa S, Chinen Y, Shimura Y, Tanba K, Takimoto T, Mizuno Y, Matsumura-Kimoto Y, Kuwahara-Ota S, Tsukamoto T, Kobayashi T, Horiike S, Taniwaki M, Kuroda J. Phosphoinositide-dependent protein kinase 1 is a potential novel therapeutic target in mantle cell lymphoma. Exp Hematol 2018; 59:72-81.e2. [DOI: 10.1016/j.exphem.2017.12.006] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2017] [Revised: 12/15/2017] [Accepted: 12/17/2017] [Indexed: 10/18/2022]
|
50
|
Concomitant inhibition of receptor tyrosine kinases and downstream AKT synergistically inhibited growth of KRAS/BRAF mutant colorectal cancer cells. Oncotarget 2018; 8:5003-5015. [PMID: 28002807 PMCID: PMC5354887 DOI: 10.18632/oncotarget.14009] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2016] [Accepted: 12/07/2016] [Indexed: 12/26/2022] Open
Abstract
Receptor tyrosine kinase (RTK) signaling pathways are frequently activated in cancer cells due to mutations of RTKs and/or their downstream signaling proteins such as KRAS and BRAF. About 40% colorectal cancers (CRCs) contain KRAS or BRAF mutant genes and are resistant to treatments with individual inhibitors of RTKs, AKT, MEK, or BRAF. Therefore, an understanding of the molecular mechanisms of the drug resistance is necessary for developing effective strategies to treat the diseases. Here we report the discovery of an AKT/ERK reactivation mechanism that account for the cancer cell resistance to the AKT and MEK inhibitors treatments. The reactivations of AKT and ERK after the AKT or MEK inhibitor treatment were caused by a relief of an AKT or ERK-mediated feedback inhibition of the RTKs and/or their downstream pathways. A combination of RTK inhibitors, based on the RTK activation/phosphorylation profile, synergized with the AKT inhibitor, but not the MEK inhibitor, to completely inhibit the AKT phosphorylation and to block the growth of KRAS/BRAF mutant CRC cells. These results underscored the importance of AKT and the AKT feedback signaling to cancer cell growth and offered a novel therapeutic approach for the treatment of KRAS/BRAF mutant CRC cells.
Collapse
|