51
|
Gao X, Gao C, Liu G, Hu J. MAP4K4: an emerging therapeutic target in cancer. Cell Biosci 2016; 6:56. [PMID: 27800153 PMCID: PMC5084373 DOI: 10.1186/s13578-016-0121-7] [Citation(s) in RCA: 63] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2016] [Accepted: 10/04/2016] [Indexed: 02/08/2023] Open
Abstract
The serine/threonine kinase MAP4K4 is a member of the Ste20p (sterile 20 protein) family. MAP4K4 was initially discovered in 1995 as a key kinase in the mating pathway in Saccharomyces cerevisiae and was later found to be involved in many aspects of cell functions and many biological and pathological processes. The role of MAP4K4 in immunity, inflammation, metabolic and cardiovascular disease has been recognized. Information regarding MAP4K4 in cancers is extremely limited, but increasing evidence suggests that MAP4K4 also plays an important role in cancer and MAP4K4 may represent a novel actionable cancer therapeutic target. This review summarizes our current understanding of MAP4K4 regulation and MAP4K4 in cancer. MAP4K4-specific inhibitors have been recently developed. We hope that this review article would advocate more basic and preclinical research on MAP4K4 in cancer, which could ultimately provide biological and mechanistic justifications for preclinical and clinical test of MAP4K4 inhibitor in cancer patients.
Collapse
Affiliation(s)
- Xuan Gao
- Department of Respiratory Medicine, Southwest Hospital, Third Military Medical University, Chongqing, China ; Department of Pharmacology and Chemical Biology, University of Pittsburgh School of Medicine, Pittsburgh, USA ; University of Pittsburgh Cancer Institute, University of Pittsburgh School of Medicine, Hillman Cancer Center Research Pavilion, 2.42D, 5117 Centre Avenue, Pittsburgh, PA 15213 USA
| | - Chenxi Gao
- Department of Pharmacology and Chemical Biology, University of Pittsburgh School of Medicine, Pittsburgh, USA ; University of Pittsburgh Cancer Institute, University of Pittsburgh School of Medicine, Hillman Cancer Center Research Pavilion, 2.42D, 5117 Centre Avenue, Pittsburgh, PA 15213 USA
| | - Guoxiang Liu
- Department of Respiratory Medicine, Southwest Hospital, Third Military Medical University, Chongqing, China
| | - Jing Hu
- Department of Pharmacology and Chemical Biology, University of Pittsburgh School of Medicine, Pittsburgh, USA ; University of Pittsburgh Cancer Institute, University of Pittsburgh School of Medicine, Hillman Cancer Center Research Pavilion, 2.42D, 5117 Centre Avenue, Pittsburgh, PA 15213 USA
| |
Collapse
|
52
|
Virbasius JV, Czech MP. Map4k4 Signaling Nodes in Metabolic and Cardiovascular Diseases. Trends Endocrinol Metab 2016; 27:484-492. [PMID: 27160798 PMCID: PMC4912878 DOI: 10.1016/j.tem.2016.04.006] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/01/2016] [Revised: 04/12/2016] [Accepted: 04/12/2016] [Indexed: 12/25/2022]
Abstract
Mitogen-activated kinase kinase kinase kinase 4 (Map4k4), originally identified in small interfering (si)RNA screens and characterized by tissue-specific gene deletions, is emerging as a regulator of glucose homeostasis and cardiovascular health. Recent studies have shown that Map4k4 gene ablation or inhibition of its kinase activity attenuates hyperglycemia and plaque formation in mouse models of insulin resistance and atherosclerosis, and suggest roles for Map4k4 in multiple signaling systems, including NFκB activation, small GTPase regulation, the Hippo cascade, and regulation of cell dynamics by FERM domain proteins. This new and promising area of inquiry raises key questions that need to be addressed, such as defining which of the above or other effectors mediate Map4k4 control of metabolic and vascular functions, and identifying upstream activators of Map4k4.
Collapse
Affiliation(s)
- Joseph V Virbasius
- Program in Molecular Medicine, University of Massachusetts Medical School, Worcester, MA 01605, USA
| | - Michael P Czech
- Program in Molecular Medicine, University of Massachusetts Medical School, Worcester, MA 01605, USA.
| |
Collapse
|
53
|
Wang Q, Amato SP, Rubitski DM, Hayward MM, Kormos BL, Verhoest PR, Xu L, Brandon NJ, Ehlers MD. Identification of Phosphorylation Consensus Sequences and Endogenous Neuronal Substrates of the Psychiatric Risk Kinase TNIK. J Pharmacol Exp Ther 2016; 356:410-23. [PMID: 26645429 DOI: 10.1124/jpet.115.229880] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2015] [Accepted: 12/01/2015] [Indexed: 12/28/2022] Open
Abstract
Traf2- and Nck-interacting kinase (TNIK) is a serine/threonine kinase highly expressed in the brain and enriched in the postsynaptic density of glutamatergic synapses in the mammalian brain. Accumulating genetic evidence and functional data have implicated TNIK as a risk factor for psychiatric disorders. However, the endogenous substrates of TNIK in neurons are unknown. Here, we describe a novel selective small molecule inhibitor of the TNIK kinase family. Using this inhibitor, we report the identification of endogenous neuronal TNIK substrates by immunoprecipitation with a phosphomotif antibody followed by mass spectrometry. Phosphorylation consensus sequences were defined by phosphopeptide sequence analysis. Among the identified substrates were members of the delta-catenin family including p120-catenin, δ-catenin, and armadillo repeat gene deleted in velo-cardio-facial syndrome (ARVCF), each of which is linked to psychiatric or neurologic disorders. Using p120-catenin as a representative substrate, we show TNIK-induced p120-catenin phosphorylation in cells requires intact kinase activity and phosphorylation of TNIK at T181 and T187 in the activation loop. Addition of the small molecule TNIK inhibitor or knocking down TNIK by two shRNAs reduced endogenous p120-catenin phosphorylation in cells. Together, using a TNIK inhibitor and phosphomotif antibody, we identify endogenous substrates of TNIK in neurons, define consensus sequences for TNIK, and suggest signaling pathways by which TNIK influences synaptic development and function linked to psychiatric and neurologic disorders.
Collapse
Affiliation(s)
- Qi Wang
- Neuroscience & Pain Research Unit, BioTherapeutics Research and Development, Pfizer Inc. Cambridge, Massachusetts (Q.W., S.P.A., D.M.R., N.J.B., M.D.E.); Center of Chemistry Innovation and Excellence, Pfizer Inc., Groton, Connecticut (M.M.H.); Neuroscience Medicinal Chemistry, Pfizer Inc., Cambridge, Massachusetts (B.L.K., P.R.V.);and Program in Molecular Medicine, University of Massachusetts Medical School, Worcester, Massachusetts (L.X.)
| | - Stephen P Amato
- Neuroscience & Pain Research Unit, BioTherapeutics Research and Development, Pfizer Inc. Cambridge, Massachusetts (Q.W., S.P.A., D.M.R., N.J.B., M.D.E.); Center of Chemistry Innovation and Excellence, Pfizer Inc., Groton, Connecticut (M.M.H.); Neuroscience Medicinal Chemistry, Pfizer Inc., Cambridge, Massachusetts (B.L.K., P.R.V.);and Program in Molecular Medicine, University of Massachusetts Medical School, Worcester, Massachusetts (L.X.)
| | - David M Rubitski
- Neuroscience & Pain Research Unit, BioTherapeutics Research and Development, Pfizer Inc. Cambridge, Massachusetts (Q.W., S.P.A., D.M.R., N.J.B., M.D.E.); Center of Chemistry Innovation and Excellence, Pfizer Inc., Groton, Connecticut (M.M.H.); Neuroscience Medicinal Chemistry, Pfizer Inc., Cambridge, Massachusetts (B.L.K., P.R.V.);and Program in Molecular Medicine, University of Massachusetts Medical School, Worcester, Massachusetts (L.X.)
| | - Matthew M Hayward
- Neuroscience & Pain Research Unit, BioTherapeutics Research and Development, Pfizer Inc. Cambridge, Massachusetts (Q.W., S.P.A., D.M.R., N.J.B., M.D.E.); Center of Chemistry Innovation and Excellence, Pfizer Inc., Groton, Connecticut (M.M.H.); Neuroscience Medicinal Chemistry, Pfizer Inc., Cambridge, Massachusetts (B.L.K., P.R.V.);and Program in Molecular Medicine, University of Massachusetts Medical School, Worcester, Massachusetts (L.X.)
| | - Bethany L Kormos
- Neuroscience & Pain Research Unit, BioTherapeutics Research and Development, Pfizer Inc. Cambridge, Massachusetts (Q.W., S.P.A., D.M.R., N.J.B., M.D.E.); Center of Chemistry Innovation and Excellence, Pfizer Inc., Groton, Connecticut (M.M.H.); Neuroscience Medicinal Chemistry, Pfizer Inc., Cambridge, Massachusetts (B.L.K., P.R.V.);and Program in Molecular Medicine, University of Massachusetts Medical School, Worcester, Massachusetts (L.X.)
| | - Patrick R Verhoest
- Neuroscience & Pain Research Unit, BioTherapeutics Research and Development, Pfizer Inc. Cambridge, Massachusetts (Q.W., S.P.A., D.M.R., N.J.B., M.D.E.); Center of Chemistry Innovation and Excellence, Pfizer Inc., Groton, Connecticut (M.M.H.); Neuroscience Medicinal Chemistry, Pfizer Inc., Cambridge, Massachusetts (B.L.K., P.R.V.);and Program in Molecular Medicine, University of Massachusetts Medical School, Worcester, Massachusetts (L.X.)
| | - Lan Xu
- Neuroscience & Pain Research Unit, BioTherapeutics Research and Development, Pfizer Inc. Cambridge, Massachusetts (Q.W., S.P.A., D.M.R., N.J.B., M.D.E.); Center of Chemistry Innovation and Excellence, Pfizer Inc., Groton, Connecticut (M.M.H.); Neuroscience Medicinal Chemistry, Pfizer Inc., Cambridge, Massachusetts (B.L.K., P.R.V.);and Program in Molecular Medicine, University of Massachusetts Medical School, Worcester, Massachusetts (L.X.)
| | - Nicholas J Brandon
- Neuroscience & Pain Research Unit, BioTherapeutics Research and Development, Pfizer Inc. Cambridge, Massachusetts (Q.W., S.P.A., D.M.R., N.J.B., M.D.E.); Center of Chemistry Innovation and Excellence, Pfizer Inc., Groton, Connecticut (M.M.H.); Neuroscience Medicinal Chemistry, Pfizer Inc., Cambridge, Massachusetts (B.L.K., P.R.V.);and Program in Molecular Medicine, University of Massachusetts Medical School, Worcester, Massachusetts (L.X.)
| | - Michael D Ehlers
- Neuroscience & Pain Research Unit, BioTherapeutics Research and Development, Pfizer Inc. Cambridge, Massachusetts (Q.W., S.P.A., D.M.R., N.J.B., M.D.E.); Center of Chemistry Innovation and Excellence, Pfizer Inc., Groton, Connecticut (M.M.H.); Neuroscience Medicinal Chemistry, Pfizer Inc., Cambridge, Massachusetts (B.L.K., P.R.V.);and Program in Molecular Medicine, University of Massachusetts Medical School, Worcester, Massachusetts (L.X.)
| |
Collapse
|
54
|
|
55
|
Roth Flach RJ, Skoura A, Matevossian A, Danai LV, Zheng W, Cortes C, Bhattacharya SK, Aouadi M, Hagan N, Yawe JC, Vangala P, Menendez LG, Cooper MP, Fitzgibbons TP, Buckbinder L, Czech MP. Endothelial protein kinase MAP4K4 promotes vascular inflammation and atherosclerosis. Nat Commun 2015; 6:8995. [PMID: 26688060 PMCID: PMC4703891 DOI: 10.1038/ncomms9995] [Citation(s) in RCA: 84] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2015] [Accepted: 10/21/2015] [Indexed: 12/21/2022] Open
Abstract
Signalling pathways that control endothelial cell (EC) permeability, leukocyte adhesion and inflammation are pivotal for atherosclerosis initiation and progression. Here we demonstrate that the Sterile-20-like mitogen-activated protein kinase kinase kinase kinase 4 (MAP4K4), which has been implicated in inflammation, is abundantly expressed in ECs and in atherosclerotic plaques from mice and humans. On the basis of endothelial-specific MAP4K4 gene silencing and gene ablation experiments in Apoe(-/-) mice, we show that MAP4K4 in ECs markedly promotes Western diet-induced aortic macrophage accumulation and atherosclerotic plaque development. Treatment of Apoe(-/-) and Ldlr(-/-) mice with a selective small-molecule MAP4K4 inhibitor also markedly reduces atherosclerotic lesion area. MAP4K4 silencing in cultured ECs attenuates cell surface adhesion molecule expression while reducing nuclear localization and activity of NFκB, which is critical for promoting EC activation and atherosclerosis. Taken together, these results reveal that MAP4K4 is a key signalling node that promotes immune cell recruitment in atherosclerosis.
Collapse
Affiliation(s)
| | - Athanasia Skoura
- Cardiovascular and Metabolic Research Unit, Cambridge, Massachusetts 02139, USA
| | | | - Laura V. Danai
- Program in Molecular Medicine, Worcester, Massachusetts 01605, USA
| | - Wei Zheng
- Cardiovascular and Metabolic Research Unit, Cambridge, Massachusetts 02139, USA
| | - Christian Cortes
- Cardiovascular and Metabolic Research Unit, Cambridge, Massachusetts 02139, USA
| | | | - Myriam Aouadi
- Program in Molecular Medicine, Worcester, Massachusetts 01605, USA
| | - Nana Hagan
- Program in Molecular Medicine, Worcester, Massachusetts 01605, USA
| | - Joseph C. Yawe
- Program in Molecular Medicine, Worcester, Massachusetts 01605, USA
| | - Pranitha Vangala
- Program in Molecular Medicine, Worcester, Massachusetts 01605, USA
| | | | - Marcus P. Cooper
- Division of Cardiovascular Medicine, University of Massachusetts Medical School, Worcester, Massachusetts 01605, USA
| | - Timothy P. Fitzgibbons
- Division of Cardiovascular Medicine, University of Massachusetts Medical School, Worcester, Massachusetts 01605, USA
| | - Leonard Buckbinder
- Cardiovascular and Metabolic Research Unit, Cambridge, Massachusetts 02139, USA
| | - Michael P. Czech
- Program in Molecular Medicine, Worcester, Massachusetts 01605, USA
| |
Collapse
|
56
|
Ammirati M, Bagley SW, Bhattacharya SK, Buckbinder L, Carlo AA, Conrad R, Cortes C, Dow RL, Dowling MS, El-Kattan A, Ford K, Guimarães CRW, Hepworth D, Jiao W, LaPerle J, Liu S, Londregan A, Loria PM, Mathiowetz AM, Munchhof M, Orr STM, Petersen DN, Price DA, Skoura A, Smith AC, Wang J. Discovery of an in Vivo Tool to Establish Proof-of-Concept for MAP4K4-Based Antidiabetic Treatment. ACS Med Chem Lett 2015; 6:1128-33. [PMID: 26617966 DOI: 10.1021/acsmedchemlett.5b00215] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2015] [Accepted: 09/28/2015] [Indexed: 01/12/2023] Open
Abstract
Recent studies in adipose tissue, pancreas, muscle, and macrophages suggest that MAP4K4, a serine/threonine protein kinase may be a viable target for antidiabetic drugs. As part of the evaluation of MAP4K4 as a novel antidiabetic target, a tool compound, 16 (PF-6260933) and a lead 17 possessing excellent kinome selectivity and suitable properties were delivered to establish proof of concept in vivo. The medicinal chemistry effort that led to the discovery of these lead compounds is described herein together with in vivo pharmacokinetic properties and activity in a model of insulin resistance.
Collapse
Affiliation(s)
- Mark Ammirati
- Worldwide Medicinal Chemistry, ‡Cardiovascular and Metabolic Research Unit, §External Research Solutions, ∥Primary Pharmacology Group, and ⊥Pharmacokinetics, Dynamics and Metabolism, Pfizer Worldwide Research & Development, 610 Main Street, Cambridge, Massachusetts 02139, United States
| | - Scott W. Bagley
- Worldwide Medicinal Chemistry, ‡Cardiovascular and Metabolic Research Unit, §External Research Solutions, ∥Primary Pharmacology Group, and ⊥Pharmacokinetics, Dynamics and Metabolism, Pfizer Worldwide Research & Development, 610 Main Street, Cambridge, Massachusetts 02139, United States
| | - Samit K. Bhattacharya
- Worldwide Medicinal Chemistry, ‡Cardiovascular and Metabolic Research Unit, §External Research Solutions, ∥Primary Pharmacology Group, and ⊥Pharmacokinetics, Dynamics and Metabolism, Pfizer Worldwide Research & Development, 610 Main Street, Cambridge, Massachusetts 02139, United States
| | - Leonard Buckbinder
- Worldwide Medicinal Chemistry, ‡Cardiovascular and Metabolic Research Unit, §External Research Solutions, ∥Primary Pharmacology Group, and ⊥Pharmacokinetics, Dynamics and Metabolism, Pfizer Worldwide Research & Development, 610 Main Street, Cambridge, Massachusetts 02139, United States
| | - Anthony A. Carlo
- Worldwide Medicinal Chemistry, ‡Cardiovascular and Metabolic Research Unit, §External Research Solutions, ∥Primary Pharmacology Group, and ⊥Pharmacokinetics, Dynamics and Metabolism, Pfizer Worldwide Research & Development, 610 Main Street, Cambridge, Massachusetts 02139, United States
| | - Rebecca Conrad
- Worldwide Medicinal Chemistry, ‡Cardiovascular and Metabolic Research Unit, §External Research Solutions, ∥Primary Pharmacology Group, and ⊥Pharmacokinetics, Dynamics and Metabolism, Pfizer Worldwide Research & Development, 610 Main Street, Cambridge, Massachusetts 02139, United States
| | - Christian Cortes
- Worldwide Medicinal Chemistry, ‡Cardiovascular and Metabolic Research Unit, §External Research Solutions, ∥Primary Pharmacology Group, and ⊥Pharmacokinetics, Dynamics and Metabolism, Pfizer Worldwide Research & Development, 610 Main Street, Cambridge, Massachusetts 02139, United States
| | - Robert L. Dow
- Worldwide Medicinal Chemistry, ‡Cardiovascular and Metabolic Research Unit, §External Research Solutions, ∥Primary Pharmacology Group, and ⊥Pharmacokinetics, Dynamics and Metabolism, Pfizer Worldwide Research & Development, 610 Main Street, Cambridge, Massachusetts 02139, United States
| | - Matthew S. Dowling
- Worldwide Medicinal Chemistry, ‡Cardiovascular and Metabolic Research Unit, §External Research Solutions, ∥Primary Pharmacology Group, and ⊥Pharmacokinetics, Dynamics and Metabolism, Pfizer Worldwide Research & Development, 610 Main Street, Cambridge, Massachusetts 02139, United States
| | - Ayman El-Kattan
- Worldwide Medicinal Chemistry, ‡Cardiovascular and Metabolic Research Unit, §External Research Solutions, ∥Primary Pharmacology Group, and ⊥Pharmacokinetics, Dynamics and Metabolism, Pfizer Worldwide Research & Development, 610 Main Street, Cambridge, Massachusetts 02139, United States
| | - Kristen Ford
- Worldwide Medicinal Chemistry, ‡Cardiovascular and Metabolic Research Unit, §External Research Solutions, ∥Primary Pharmacology Group, and ⊥Pharmacokinetics, Dynamics and Metabolism, Pfizer Worldwide Research & Development, 610 Main Street, Cambridge, Massachusetts 02139, United States
| | - Cristiano R. W. Guimarães
- Worldwide Medicinal Chemistry, ‡Cardiovascular and Metabolic Research Unit, §External Research Solutions, ∥Primary Pharmacology Group, and ⊥Pharmacokinetics, Dynamics and Metabolism, Pfizer Worldwide Research & Development, 610 Main Street, Cambridge, Massachusetts 02139, United States
| | - David Hepworth
- Worldwide Medicinal Chemistry, ‡Cardiovascular and Metabolic Research Unit, §External Research Solutions, ∥Primary Pharmacology Group, and ⊥Pharmacokinetics, Dynamics and Metabolism, Pfizer Worldwide Research & Development, 610 Main Street, Cambridge, Massachusetts 02139, United States
| | - Wenhua Jiao
- Worldwide Medicinal Chemistry, ‡Cardiovascular and Metabolic Research Unit, §External Research Solutions, ∥Primary Pharmacology Group, and ⊥Pharmacokinetics, Dynamics and Metabolism, Pfizer Worldwide Research & Development, 610 Main Street, Cambridge, Massachusetts 02139, United States
| | - Jennifer LaPerle
- Worldwide Medicinal Chemistry, ‡Cardiovascular and Metabolic Research Unit, §External Research Solutions, ∥Primary Pharmacology Group, and ⊥Pharmacokinetics, Dynamics and Metabolism, Pfizer Worldwide Research & Development, 610 Main Street, Cambridge, Massachusetts 02139, United States
| | - Shenping Liu
- Worldwide Medicinal Chemistry, ‡Cardiovascular and Metabolic Research Unit, §External Research Solutions, ∥Primary Pharmacology Group, and ⊥Pharmacokinetics, Dynamics and Metabolism, Pfizer Worldwide Research & Development, 610 Main Street, Cambridge, Massachusetts 02139, United States
| | - Allyn Londregan
- Worldwide Medicinal Chemistry, ‡Cardiovascular and Metabolic Research Unit, §External Research Solutions, ∥Primary Pharmacology Group, and ⊥Pharmacokinetics, Dynamics and Metabolism, Pfizer Worldwide Research & Development, 610 Main Street, Cambridge, Massachusetts 02139, United States
| | - Paula M. Loria
- Worldwide Medicinal Chemistry, ‡Cardiovascular and Metabolic Research Unit, §External Research Solutions, ∥Primary Pharmacology Group, and ⊥Pharmacokinetics, Dynamics and Metabolism, Pfizer Worldwide Research & Development, 610 Main Street, Cambridge, Massachusetts 02139, United States
| | - Alan M. Mathiowetz
- Worldwide Medicinal Chemistry, ‡Cardiovascular and Metabolic Research Unit, §External Research Solutions, ∥Primary Pharmacology Group, and ⊥Pharmacokinetics, Dynamics and Metabolism, Pfizer Worldwide Research & Development, 610 Main Street, Cambridge, Massachusetts 02139, United States
| | - Michael Munchhof
- Worldwide Medicinal Chemistry, ‡Cardiovascular and Metabolic Research Unit, §External Research Solutions, ∥Primary Pharmacology Group, and ⊥Pharmacokinetics, Dynamics and Metabolism, Pfizer Worldwide Research & Development, 610 Main Street, Cambridge, Massachusetts 02139, United States
| | - Suvi T. M. Orr
- Worldwide Medicinal Chemistry, ‡Cardiovascular and Metabolic Research Unit, §External Research Solutions, ∥Primary Pharmacology Group, and ⊥Pharmacokinetics, Dynamics and Metabolism, Pfizer Worldwide Research & Development, 610 Main Street, Cambridge, Massachusetts 02139, United States
| | - Donna N. Petersen
- Worldwide Medicinal Chemistry, ‡Cardiovascular and Metabolic Research Unit, §External Research Solutions, ∥Primary Pharmacology Group, and ⊥Pharmacokinetics, Dynamics and Metabolism, Pfizer Worldwide Research & Development, 610 Main Street, Cambridge, Massachusetts 02139, United States
| | - David A. Price
- Worldwide Medicinal Chemistry, ‡Cardiovascular and Metabolic Research Unit, §External Research Solutions, ∥Primary Pharmacology Group, and ⊥Pharmacokinetics, Dynamics and Metabolism, Pfizer Worldwide Research & Development, 610 Main Street, Cambridge, Massachusetts 02139, United States
| | - Athanasia Skoura
- Worldwide Medicinal Chemistry, ‡Cardiovascular and Metabolic Research Unit, §External Research Solutions, ∥Primary Pharmacology Group, and ⊥Pharmacokinetics, Dynamics and Metabolism, Pfizer Worldwide Research & Development, 610 Main Street, Cambridge, Massachusetts 02139, United States
| | - Aaron C. Smith
- Worldwide Medicinal Chemistry, ‡Cardiovascular and Metabolic Research Unit, §External Research Solutions, ∥Primary Pharmacology Group, and ⊥Pharmacokinetics, Dynamics and Metabolism, Pfizer Worldwide Research & Development, 610 Main Street, Cambridge, Massachusetts 02139, United States
| | - Jian Wang
- Worldwide Medicinal Chemistry, ‡Cardiovascular and Metabolic Research Unit, §External Research Solutions, ∥Primary Pharmacology Group, and ⊥Pharmacokinetics, Dynamics and Metabolism, Pfizer Worldwide Research & Development, 610 Main Street, Cambridge, Massachusetts 02139, United States
| |
Collapse
|
57
|
Yang N, Wang Y, Hui L, Li X, Jiang X. Silencing SOX2 Expression by RNA Interference Inhibits Proliferation, Invasion and Metastasis, and Induces Apoptosis through MAP4K4/JNK Signaling Pathway in Human Laryngeal Cancer TU212 Cells. J Histochem Cytochem 2015; 63:721-33. [PMID: 26001828 DOI: 10.1369/0022155415590829] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2015] [Accepted: 05/17/2015] [Indexed: 12/19/2022] Open
Abstract
SRY (sex determining region Y)-box 2 (SOX2) plays an important role in tumor cell metastasis and apoptosis. Laryngeal squamous cell carcinoma (LSCC), responsible for 1.5% of all cancers, is one of the most common head and neck malignancies. Accumulating evidence shows that SOX2 is overexpressed in several human tumors, including lung cancer, esophageal carcinoma, pancreatic carcinoma, breast cancer, ovarian carcinoma and glioma. Our study aimed to investigate the silencing effects of SOX2 expression using RNA interference (RNAi) on various biological processes in laryngeal cancer TU212 cells, including proliferation, apoptosis, invasion and metastasis. We also studied the involvement of the MAPK/JNK signaling pathway in the biological effects of SOX2 siRNA in TU212 cells. We found that silencing SOX2 decreased the proliferation, migration, and invasion of TU212 cells, and induced apoptosis. This effect of silencing SOX2 could be reversed by silencing MAP4K4. Therefore, we consider SOX2 as a key regulator of the upstream MAP4K4/JNK signaling pathways that could be a potential therapeutic target in the treatment of patients with or prevention of laryngeal cancer.
Collapse
Affiliation(s)
- Ning Yang
- Department of Otorhinolaryngology, The First Affiliated Hospital of China Medical University, Shenyang, People’s
Republic of China (NY, YW, LH, XL, XJ)
| | - Yan Wang
- Department of Otorhinolaryngology, The First Affiliated Hospital of China Medical University, Shenyang, People’s
Republic of China (NY, YW, LH, XL, XJ)
| | - Lian Hui
- Department of Otorhinolaryngology, The First Affiliated Hospital of China Medical University, Shenyang, People’s
Republic of China (NY, YW, LH, XL, XJ)
| | - Xiaotian Li
- Department of Otorhinolaryngology, The First Affiliated Hospital of China Medical University, Shenyang, People’s
Republic of China (NY, YW, LH, XL, XJ)
| | - Xuejun Jiang
- Department of Otorhinolaryngology, The First Affiliated Hospital of China Medical University, Shenyang, People’s
Republic of China (NY, YW, LH, XL, XJ)
| |
Collapse
|
58
|
Bucchia M, Ramirez A, Parente V, Simone C, Nizzardo M, Magri F, Dametti S, Corti S. Therapeutic Development in Amyotrophic Lateral Sclerosis. Clin Ther 2015; 37:668-80. [DOI: 10.1016/j.clinthera.2014.12.020] [Citation(s) in RCA: 46] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2014] [Revised: 09/18/2014] [Accepted: 12/29/2014] [Indexed: 12/12/2022]
|
59
|
Santhana Kumar K, Tripolitsioti D, Ma M, Grählert J, Egli KB, Fiaschetti G, Shalaby T, Grotzer MA, Baumgartner M. The Ser/Thr kinase MAP4K4 drives c-Met-induced motility and invasiveness in a cell-based model of SHH medulloblastoma. SPRINGERPLUS 2015; 4:19. [PMID: 25625039 PMCID: PMC4302160 DOI: 10.1186/s40064-015-0784-2] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/22/2014] [Accepted: 01/02/2015] [Indexed: 01/20/2023]
Abstract
Medulloblastoma (MB) comprises four molecularly and genetically distinct subgroups of embryonal brain tumors that develop in the cerebellum. MB mostly affects infants and children and is difficult to treat because of frequent dissemination of tumor cells within the leptomeningeal space. A potential promoter of cell dissemination is the c-Met proto-oncogene receptor tyrosine kinase, which is aberrantly expressed in many human tumors including MB. Database analysis showed that c-Met is highly expressed in the sonic hedgehog (SHH) subgroup and in a small subset of Group 3 and Group 4 MB tumors. Using a cell-based three-dimensional cell motility assay combined with live-cell imaging, we investigated whether the c-Met ligand HGF could drive dissemination of MB cells expressing high levels of c-Met, and determined downstream effector mechanisms of this process. We detected variable c-Met expression in different established human MB cell lines, and we found that in lines expressing high c-Met levels, HGF promoted cell dissemination and invasiveness. Specifically, HGF-induced c-Met activation enhanced the capability of the individual cells to migrate in a JNK-dependent manner. Additionally, we identified the Ser/Thr kinase MAP4K4 as a novel driver of c-Met-induced invasive cell dissemination. This increased invasive motility was due to MAP4K4 control of F-actin dynamics in structures required for migration and invasion. Thus, MAP4K4 couples growth factor signaling to actin cytoskeleton regulation in tumor cells, suggesting that MAP4K4 could present a promising novel target to be evaluated for treating growth factor-induced dissemination of MB tumors of different subgroups and of other human cancers.
Collapse
Affiliation(s)
- Karthiga Santhana Kumar
- Department of Oncology, Children's Research Center, University Children's Hospital, Zurich, Switzerland
| | - Dimitra Tripolitsioti
- Department of Oncology, Children's Research Center, University Children's Hospital, Zurich, Switzerland
| | - Min Ma
- Department of Oncology, Children's Research Center, University Children's Hospital, Zurich, Switzerland
| | - Jasmin Grählert
- Department of Oncology, Children's Research Center, University Children's Hospital, Zurich, Switzerland.,Current address: Department of Biomedicine, University Hospital Basel, Basel, Switzerland
| | - Katja B Egli
- Department of Oncology, Children's Research Center, University Children's Hospital, Zurich, Switzerland
| | - Giulio Fiaschetti
- Department of Oncology, Children's Research Center, University Children's Hospital, Zurich, Switzerland
| | - Tarek Shalaby
- Department of Oncology, Children's Research Center, University Children's Hospital, Zurich, Switzerland
| | - Michael A Grotzer
- Department of Oncology, Children's Research Center, University Children's Hospital, Zurich, Switzerland
| | - Martin Baumgartner
- Department of Oncology, Children's Research Center, University Children's Hospital, Zurich, Switzerland.,University Children's Hospital Zürich, Department of Oncology, Children's Research Center, Neuro-Oncology group, August-Forel Strasse 1, CH-8008 Zürich, Switzerland
| |
Collapse
|
60
|
Wang L, Stanley M, Boggs JW, Crawford TD, Bravo BJ, Giannetti AM, Harris SF, Magnuson SR, Nonomiya J, Schmidt S, Wu P, Ye W, Gould SE, Murray LJ, Ndubaku CO, Chen H. Fragment-based identification and optimization of a class of potent pyrrolo[2,1-f][1,2,4]triazine MAP4K4 inhibitors. Bioorg Med Chem Lett 2014; 24:4546-4552. [PMID: 25139565 DOI: 10.1016/j.bmcl.2014.07.071] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2014] [Revised: 07/24/2014] [Accepted: 07/28/2014] [Indexed: 01/27/2023]
Abstract
MAP4K4 has been shown to regulate key cellular processes that are tied to disease pathogenesis. In an effort to generate small molecule MAP4K4 inhibitors, a fragment-based screen was carried out and a pyrrolotriazine fragment with excellent ligand efficiency was identified. Further modification of this fragment guided by X-ray crystal structures and molecular modeling led to the discovery of a series of promising compounds with good structural diversity and physicochemical properties. These compounds exhibited single digit nanomolar potency and compounds 35 and 44 achieved good in vivo exposure.
Collapse
Affiliation(s)
- Lan Wang
- Department of Discovery Chemistry, Genentech, Inc., 1 DNA Way, South San Francisco, CA 94080, United States
| | - Mark Stanley
- Department of Discovery Chemistry, Genentech, Inc., 1 DNA Way, South San Francisco, CA 94080, United States
| | - Jason W Boggs
- Department of Drug Metabolism and Pharmacokinetics, Genentech, Inc., 1 DNA Way, South San Francisco, CA 94080, United States
| | - Terry D Crawford
- Department of Discovery Chemistry, Genentech, Inc., 1 DNA Way, South San Francisco, CA 94080, United States
| | - Brandon J Bravo
- Department of Biochemical and Cellular Pharmacology, Genentech, Inc., 1 DNA Way, South San Francisco, CA 94080, United States
| | - Anthony M Giannetti
- Department of Biochemical and Cellular Pharmacology, Genentech, Inc., 1 DNA Way, South San Francisco, CA 94080, United States
| | - Seth F Harris
- Department of Structural Biology, Genentech, Inc., 1 DNA Way, South San Francisco, CA 94080, United States
| | - Steven R Magnuson
- Department of Discovery Chemistry, Genentech, Inc., 1 DNA Way, South San Francisco, CA 94080, United States
| | - Jim Nonomiya
- Department of Biochemical and Cellular Pharmacology, Genentech, Inc., 1 DNA Way, South San Francisco, CA 94080, United States
| | - Stephen Schmidt
- Department of Biochemical and Cellular Pharmacology, Genentech, Inc., 1 DNA Way, South San Francisco, CA 94080, United States
| | - Ping Wu
- Department of Structural Biology, Genentech, Inc., 1 DNA Way, South San Francisco, CA 94080, United States
| | - Weilan Ye
- Department of Molecular Oncology, Genentech, Inc., 1 DNA Way, South San Francisco, CA 94080, United States
| | - Stephen E Gould
- Department of Molecular Oncology, Genentech, Inc., 1 DNA Way, South San Francisco, CA 94080, United States
| | - Lesley J Murray
- Department of Drug Metabolism and Pharmacokinetics, Genentech, Inc., 1 DNA Way, South San Francisco, CA 94080, United States
| | - Chudi O Ndubaku
- Department of Discovery Chemistry, Genentech, Inc., 1 DNA Way, South San Francisco, CA 94080, United States.
| | - Huifen Chen
- Department of Discovery Chemistry, Genentech, Inc., 1 DNA Way, South San Francisco, CA 94080, United States.
| |
Collapse
|
61
|
Chuang HC, Sheu WHH, Lin YT, Tsai CY, Yang CY, Cheng YJ, Huang PY, Li JP, Chiu LL, Wang X, Xie M, Schneider MD, Tan TH. HGK/MAP4K4 deficiency induces TRAF2 stabilization and Th17 differentiation leading to insulin resistance. Nat Commun 2014; 5:4602. [PMID: 25098764 PMCID: PMC4143962 DOI: 10.1038/ncomms5602] [Citation(s) in RCA: 73] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2014] [Accepted: 07/07/2014] [Indexed: 02/06/2023] Open
Abstract
Proinflammatory cytokines play important roles in insulin resistance. Here we report that mice with a T-cell-specific conditional knockout of HGK (T-HGK cKO) develop systemic inflammation and insulin resistance. This condition is ameliorated by either IL-6 or IL-17 neutralization. HGK directly phosphorylates TRAF2, leading to its lysosomal degradation and subsequent inhibition of IL-6 production. IL-6-overproducing HGK-deficient T cells accumulate in adipose tissue and further differentiate into IL-6/IL-17 double-positive cells. Moreover, CCL20 neutralization or CCR6 deficiency reduces the Th17 population or insulin resistance in T-HGK cKO mice. In addition, leptin receptor deficiency in T cells inhibits Th17 differentiation and improves the insulin sensitivity in T-HGK cKO mice, which suggests that leptin cooperates with IL-6 to promote Th17 differentiation. Thus, HGK deficiency induces TRAF2/IL-6 upregulation, leading to IL-6/leptin-induced Th17 differentiation in adipose tissue and subsequent insulin resistance. These findings provide insight into the reciprocal regulation between the immune system and the metabolism.
Collapse
Affiliation(s)
- Huai-Chia Chuang
- Immunology Research Center, National Health Research Institutes, 35 Keyan Road, Zhunan 35053, Taiwan
| | - Wayne H. -H. Sheu
- Division of Endocrinology and Metabolism, Taichung Veterans General Hospital, 160, Sec. 3, Chung-Kang Road, Taichung 40705, Taiwan
- Faculty of Medicine, National Yang-Ming University, Taipei 11221, Taiwan
| | - Yi-Ting Lin
- Immunology Research Center, National Health Research Institutes, 35 Keyan Road, Zhunan 35053, Taiwan
| | - Ching-Yi Tsai
- Immunology Research Center, National Health Research Institutes, 35 Keyan Road, Zhunan 35053, Taiwan
| | - Chia-Yu Yang
- Immunology Research Center, National Health Research Institutes, 35 Keyan Road, Zhunan 35053, Taiwan
| | - Yu-Jhen Cheng
- Immunology Research Center, National Health Research Institutes, 35 Keyan Road, Zhunan 35053, Taiwan
| | - Pau-Yi Huang
- Immunology Research Center, National Health Research Institutes, 35 Keyan Road, Zhunan 35053, Taiwan
| | - Ju-Pi Li
- Immunology Research Center, National Health Research Institutes, 35 Keyan Road, Zhunan 35053, Taiwan
| | - Li-Li Chiu
- Division of Endocrinology and Metabolism, Taichung Veterans General Hospital, 160, Sec. 3, Chung-Kang Road, Taichung 40705, Taiwan
| | - Xiaohong Wang
- Department of Pathology and Immunology, Baylor College of Medicine, Houston, Texas 77030, USA
| | - Min Xie
- UT Southwestern Medical Center at Dallas, 5323 Harry Hines Boulevard, Dallas, Texas 75390, USA
| | - Michael D. Schneider
- Faculty of Medicine, British Heart Foundation Centre of Research Excellence, National Heart and Lung Institute, Imperial College London, South Kensington Campus, London SW7 2AZ, UK
| | - Tse-Hua Tan
- Immunology Research Center, National Health Research Institutes, 35 Keyan Road, Zhunan 35053, Taiwan
- Department of Pathology and Immunology, Baylor College of Medicine, Houston, Texas 77030, USA
| |
Collapse
|
62
|
Ma M, Baumgartner M. Intracellular Theileria annulata promote invasive cell motility through kinase regulation of the host actin cytoskeleton. PLoS Pathog 2014; 10:e1004003. [PMID: 24626571 PMCID: PMC3953445 DOI: 10.1371/journal.ppat.1004003] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2013] [Accepted: 02/01/2014] [Indexed: 11/18/2022] Open
Abstract
The intracellular, protozoan Theileria species parasites are the only eukaryotes known to transform another eukaryotic cell. One consequence of this parasite-dependent transformation is the acquisition of motile and invasive properties of parasitized cells in vitro and their metastatic dissemination in the animal, which causes East Coast Fever (T. parva) or Tropical Theileriosis (T. annulata). These motile and invasive properties of infected host cells are enabled by parasite-dependent, poorly understood F-actin dynamics that control host cell membrane protrusions. Herein, we dissected functional and structural alterations that cause acquired motility and invasiveness of T. annulata-infected cells, to understand the molecular basis driving cell dissemination in Tropical Theileriosis. We found that chronic induction of TNFα by the parasite contributes to motility and invasiveness of parasitized host cells. We show that TNFα does so by specifically targeting expression and function of the host proto-oncogenic ser/thr kinase MAP4K4. Blocking either TNFα secretion or MAP4K4 expression dampens the formation of polar, F-actin-rich invasion structures and impairs cell motility in 3D. We identified the F-actin binding ERM family proteins as MAP4K4 downstream effectors in this process because TNFα-induced ERM activation and cell invasiveness are sensitive to MAP4K4 depletion. MAP4K4 expression in infected cells is induced by TNFα-JNK signalling and maintained by the inhibition of translational repression, whereby both effects are parasite dependent. Thus, parasite-induced TNFα promotes invasive motility of infected cells through the activation of MAP4K4, an evolutionary conserved kinase that controls cytoskeleton dynamics and cell motility. Hence, MAP4K4 couples inflammatory signaling to morphodynamic processes and cell motility, a process exploited by the intracellular Theileria parasite to increase its host cell's dissemination capabilities. The protozoan parasite Theileria annulata causes the often fatal leukoproliferative disorder Tropical Theileriosis in their ruminant host animals, which is the result of widespread dissemination and proliferation of cytokine secreting, parasite-infected cells. This host cell behavior is induced by and dependent on the intracellular presence of the parasite and is reminiscent of metastatic dissemination of human cancer cells. We investigated how the intracellular parasite modulates cell motility and invasiveness, to better understand the pathogenesis of Tropical Theileriosis and to reveal conserved mechanisms of eukaryotic cell motility regulation. We found that the parasite drives host cell motility and invasiveness through the induction and activation of the host cell protein MAP4K4. We show that MAP4K4 induction is driven by the inflammatory cytokine TNFα and causes dynamic changes in the cytoskeleton of the host cell that facilitate cell motility. Thus, our findings reveal how the intracellular Theileria parasite can influence morphology and behavior of its host cell in a way that suits its propagation and highlight a novel function of chronic TNFα production for the pathogenesis of Tropical Theileriosis. Furthermore, our study revealed a novel aspect of inflammatory cytokine action, namely cell mobilization through the induction of the evolutionary conserved protein kinase MAP4K4.
Collapse
Affiliation(s)
- Min Ma
- Neuro-Oncology, Experimental Infectious Diseases and Cancer Research, University Children's Hospital Zürich, Zürich, Switzerland
- Molecular Pathobiology, University of Bern, Bern, Switzerland
- Graduate School for Cellular and Biomedical Sciences, University of Bern, Bern, Switzerland
| | - Martin Baumgartner
- Neuro-Oncology, Experimental Infectious Diseases and Cancer Research, University Children's Hospital Zürich, Zürich, Switzerland
- Molecular Pathobiology, University of Bern, Bern, Switzerland
- * E-mail:
| |
Collapse
|
63
|
Imidazopyridine-fused [1,3]-diazepinones: Synthesis and antiproliferative activity. Eur J Med Chem 2014; 75:382-90. [DOI: 10.1016/j.ejmech.2014.01.044] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2013] [Revised: 01/17/2014] [Accepted: 01/18/2014] [Indexed: 01/26/2023]
|
64
|
Fiedler LR, Maifoshie E, Schneider MD. Mouse models of heart failure: cell signaling and cell survival. Curr Top Dev Biol 2014; 109:171-247. [PMID: 24947238 DOI: 10.1016/b978-0-12-397920-9.00002-0] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
Heart failure is one of the paramount global causes of morbidity and mortality. Despite this pandemic need, the available clinical counter-measures have not altered substantially in recent decades, most notably in the context of pharmacological interventions. Cell death plays a causal role in heart failure, and its inhibition poses a promising approach that has not been thoroughly explored. In previous approaches to target discovery, clinical failures have reflected a deficiency in mechanistic understanding, and in some instances, failure to systematically translate laboratory findings toward the clinic. Here, we review diverse mouse models of heart failure, with an emphasis on those that identify potential targets for pharmacological inhibition of cell death, and on how their translation into effective therapies might be improved in the future.
Collapse
Affiliation(s)
- Lorna R Fiedler
- British Heart Foundation Centre of Research Excellence, National Heart and Lung Institute, Imperial College London, London, UK.
| | - Evie Maifoshie
- British Heart Foundation Centre of Research Excellence, National Heart and Lung Institute, Imperial College London, London, UK
| | - Michael D Schneider
- British Heart Foundation Centre of Research Excellence, National Heart and Lung Institute, Imperial College London, London, UK.
| |
Collapse
|
65
|
Wang H, Chen Y, Lin P, Li L, Zhou G, Liu G, Logsdon C, Jin J, Abbruzzese JL, Tan TH, Wang H. The CUL7/F-box and WD repeat domain containing 8 (CUL7/Fbxw8) ubiquitin ligase promotes degradation of hematopoietic progenitor kinase 1. J Biol Chem 2013; 289:4009-17. [PMID: 24362026 DOI: 10.1074/jbc.m113.520106] [Citation(s) in RCA: 33] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
HPK1, a member of mammalian Ste20-like serine/threonine kinases, is lost in >95% pancreatic cancer through proteasome-mediated degradation. However, the mechanism of HPK1 loss has not been defined. The aims of this study are to identify the ubiquitin ligase and to examine the mechanisms that targets HPK1 degradation. We found that the CUL7/Fbxw8 ubiquitin ligase targeted HPK1 for degradation via the 26 S proteasome. The ubiquitination of HPK1 required its kinase activity and autophosphorylation. Wild-type protein phosphatase 4 (PP4), but not the phosphatase-dead PP4 mutant, PP4-RL, inhibits the interaction of Fbxw8 with HPK1 and Fbxw8-mediated ubiquitination of HPK1. In addition, we showed that Thr-355 of HPK1 is a key PP4 dephosphorylation site, through which CUL7/Fbxw8 ubiquitin ligase and PP4 regulates HPK1 stability. Knockdown of Fbxw8 restores endogenous HPK1 protein expression and inhibits cell proliferation of pancreatic cancer cells. Our study demonstrated that targeted degradation of HPK1 by the CUL7/Fbxw8 ubiquitin ligase constitutes a negative-feedback loop to restrain the activity of HPK1 and that CUL7/Fbxw8 ubiquitin ligase promotes pancreatic cancer cell proliferation. CUL7/Fbxw8 ubiquitin ligase-mediated HPK1 degradation revealed a direct link and novel role of CUL7/Fbxw8 ubiquitin ligase in the MAPK pathway, which plays a critical role in cell proliferation and differentiation.
Collapse
Affiliation(s)
- Hua Wang
- From the Departments of Gastrointestinal Medical Oncology
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
66
|
Chen S, Li X, Lu D, Xu Y, Mou W, Wang L, Chen Y, Liu Y, Li X, Li LY, Liu L, Stupack D, Reisfeld RA, Xiang R, Li N. SOX2 regulates apoptosis through MAP4K4-survivin signaling pathway in human lung cancer cells. Carcinogenesis 2013; 35:613-23. [PMID: 24233838 DOI: 10.1093/carcin/bgt371] [Citation(s) in RCA: 74] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/15/2023] Open
Abstract
Previous studies have implicated cancer stem cells in tumor recurrence and revealed that the stem cell gene SOX2 plays an important role in the tumor cell resistance to apoptosis. Nonetheless, the mechanism by which SOX2 regulates apoptosis signals remained undefined. Here, we demonstrated the surprising finding that silencing of the SOX2 gene effectively induces apoptosis via the activation of death receptor and mitochondrial signaling pathways in human non-small cell lung cancer cells. Unexpectedly, reverse transcription-PCR analysis suggested that downregulation of SOX2 leads to activation of MAP4K4, previously implicated in cell survival. Evaluation of the apoptotic pathways revealed an increased expression of key inducers of apoptosis, including tumor necrosis factor-α and p53, with concurrent attenuation of Survivin. Although p53 appeared dispensable for this pathway, the loss of Survivin in SOX2-deficient cells appeared critical for the observed MAP4K4 induced cell death. Rescue experiments revealed that SOX2-silencing-mediated killing was blocked by ectopic expression of Survivin, or by reduction of MAP4K4 expression. Clinically, expressions of Survivin and SOX2 were highly correlated with each other. The results reveal a key target of SOX2 expression and highlight the unexpected context-dependent role for MAP4K4, a pluripotent activator of several mitogen-activated protein kinase pathways, in regulating tumor cell survival.
Collapse
Affiliation(s)
- Si Chen
- School of Medicine, Nankai University, 94 Weijin Road, Tianjin 300071, China
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
67
|
Haas DA, Bala K, Büsche G, Weidner-Glunde M, Santag S, Kati S, Gramolelli S, Damas M, Dittrich-Breiholz O, Kracht M, Rückert J, Varga Z, Keri G, Schulz TF. The inflammatory kinase MAP4K4 promotes reactivation of Kaposi's sarcoma herpesvirus and enhances the invasiveness of infected endothelial cells. PLoS Pathog 2013; 9:e1003737. [PMID: 24244164 PMCID: PMC3820715 DOI: 10.1371/journal.ppat.1003737] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2013] [Accepted: 09/15/2013] [Indexed: 12/15/2022] Open
Abstract
Kaposi's sarcoma (KS) is a mesenchymal tumour, which is caused by Kaposi's sarcoma herpesvirus (KSHV) and develops under inflammatory conditions. KSHV-infected endothelial spindle cells, the neoplastic cells in KS, show increased invasiveness, attributed to the elevated expression of metalloproteinases (MMPs) and cyclooxygenase-2 (COX-2). The majority of these spindle cells harbour latent KSHV genomes, while a minority undergoes lytic reactivation with subsequent production of new virions and viral or cellular chemo- and cytokines, which may promote tumour invasion and dissemination. In order to better understand KSHV pathogenesis, we investigated cellular mechanisms underlying the lytic reactivation of KSHV. Using a combination of small molecule library screening and siRNA silencing we found a STE20 kinase family member, MAP4K4, to be involved in KSHV reactivation from latency and to contribute to the invasive phenotype of KSHV-infected endothelial cells by regulating COX-2, MMP-7, and MMP-13 expression. This kinase is also highly expressed in KS spindle cells in vivo. These findings suggest that MAP4K4, a known mediator of inflammation, is involved in KS aetiology by regulating KSHV lytic reactivation, expression of MMPs and COX-2, and, thereby modulating invasiveness of KSHV-infected endothelial cells. Kaposi's sarcoma (KS) is a tumour caused by Kaposi's sarcoma herpesvirus (KSHV) and dysregulated inflammation. Both factors contribute to the high angiogenicity and invasiveness of KS. Various cellular kinases have been reported to regulate the KSHV latent-lytic switch and thereby virus pathogenicity. In this study, we have identified a STE20 kinase family member – MAP4K4 – as a modulator of KSHV lytic cycle and invasive phenotype of KSHV-infected endothelial cells. Moreover, we were able to link MAP4K4 to a known mediator of inflammation and invasiveness, cyclooxygenase-2, which also contributes to KSHV lytic replication. Finally, we could show that MAP4K4 is highly expressed in KS lesions, suggesting an important role for this kinase in tumour development and invasion.
Collapse
Affiliation(s)
- Darya A Haas
- Institute of Virology, Hannover Medical School, Hannover, Germany
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
68
|
Chen S, Xuan J, Wan L, Lin H, Couch L, Mei N, Dobrovolsky VN, Guo L. Sertraline, an antidepressant, induces apoptosis in hepatic cells through the mitogen-activated protein kinase pathway. Toxicol Sci 2013; 137:404-15. [PMID: 24194395 DOI: 10.1093/toxsci/kft254] [Citation(s) in RCA: 47] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/14/2022] Open
Abstract
Sertraline is generally used for the treatment of depression and is also approved for the treatment of panic, obsessive-compulsive, and posttraumatic stress disorders. Previously, using rat primary hepatocytes and isolated mitochondria, we demonstrated that sertraline caused hepatic cytotoxicity and mitochondrial impairment. In the current study, we investigated and characterized molecular mechanisms of sertraline toxicity in human hepatoma HepG2 cells. Sertraline decreased cell viability and induced apoptosis in a dose- and time-dependent manner. Sertraline activated the intrinsic checkpoint protein caspase-9 and caused the release of cytochrome c from mitochondria to cytosol; this process was Bcl-2 family dependent because antiapoptotic Bcl-2 family proteins were decreased. Pretreatment of the HepG2 cells with caspase-3, caspase-8, and caspase-9 inhibitors partially but significantly reduced the release of lactate dehydrogenase, indicating that sertraline-induced apoptosis is mediated by both intrinsic and extrinsic apoptotic pathways. Moreover, sertraline markedly increased the expression of tumor necrosis factor (TNF) and the phosphorylation of JNK, extracellular signal-regulated kinase (ERK1/2), and p38. In sertraline-treated cells, the induction of apoptosis and cell death was shown to be the result of activation of JNK, but not ERK1/2 or p38 in the mitogen-activated protein kinase (MAPK) pathway. Furthermore, silencing MAP4K4, the upstream kinase of JNK, attenuated both apoptosis and cell death caused by sertraline. Taken together, our findings suggest that sertraline induced apoptosis in HepG2 cells at least partially via activation of the TNF-MAP4K4-JNK cascade signaling pathway.
Collapse
Affiliation(s)
- Si Chen
- * Division of Biochemical Toxicology, National Center for Toxicological Research, U.S. FDA, Jefferson, AR 72079
| | | | | | | | | | | | | | | |
Collapse
|
69
|
Danai LV, Guilherme A, Guntur KV, Straubhaar J, Nicoloro SM, Czech MP. Map4k4 suppresses Srebp-1 and adipocyte lipogenesis independent of JNK signaling. J Lipid Res 2013; 54:2697-707. [PMID: 23924694 PMCID: PMC3770083 DOI: 10.1194/jlr.m038802] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2013] [Revised: 08/05/2013] [Indexed: 11/20/2022] Open
Abstract
Adipose tissue lipogenesis is paradoxically impaired in human obesity, promoting ectopic triglyceride (TG) deposition, lipotoxicity, and insulin resistance. We previously identified mitogen-activated protein kinase kinase kinase kinase 4 (Map4k4), a sterile 20 protein kinase reported to be upstream of c-Jun NH2-terminal kinase (JNK) signaling, as a novel negative regulator of insulin-stimulated glucose transport in adipocytes. Using full-genome microarray analysis we uncovered a novel role for Map4k4 as a suppressor of lipid synthesis. We further report here the surprising finding that Map4k4 suppresses adipocyte lipogenesis independently of JNK. Thus, while Map4k4 silencing in adipocytes enhances the expression of lipogenic enzymes, concomitant with increased conversion of (14)C-glucose and (14)C-acetate into TGs and fatty acids, JNK1 and JNK2 depletion causes the opposite effects. Furthermore, high expression of Map4k4 fails to activate endogenous JNK, while Map4k4 depletion does not attenuate JNK activation by tumor necrosis factor α. Map4k4 silencing in cultured adipocytes elevates both the total protein expression and cleavage of sterol-regulated element binding protein-1 (Srebp-1) in a rapamycin-sensitive manner, consistent with Map4k4 signaling via mechanistic target of rapamycin complex 1 (mTORC1). We show Map4k4 depletion requires Srebp-1 upregulation to increase lipogenesis and further show that Map4k4 promotes AMP-protein kinase (AMPK) signaling and the phosphorylation of mTORC1 binding partner raptor (Ser792) to inhibit mTORC1. Our results indicate that Map4k4 inhibits adipose lipogenesis by suppression of Srebp-1 in an AMPK- and mTOR-dependent but JNK-independent mechanism.
Collapse
Affiliation(s)
- Laura V. Danai
- Program in Molecular Medicine, University of Massachusetts Medical School, Worcester, MA 01605
| | - Adilson Guilherme
- Program in Molecular Medicine, University of Massachusetts Medical School, Worcester, MA 01605
| | | | - Juerg Straubhaar
- Program in Molecular Medicine, University of Massachusetts Medical School, Worcester, MA 01605
| | - Sarah M. Nicoloro
- Program in Molecular Medicine, University of Massachusetts Medical School, Worcester, MA 01605
| | - Michael P. Czech
- Program in Molecular Medicine, University of Massachusetts Medical School, Worcester, MA 01605
| |
Collapse
|
70
|
A Novel Interaction between Pyk2 and MAP4K4 Is Integrated with Glioma Cell Migration. JOURNAL OF SIGNAL TRANSDUCTION 2013; 2013:956580. [PMID: 24163766 PMCID: PMC3791834 DOI: 10.1155/2013/956580] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/17/2013] [Revised: 08/07/2013] [Accepted: 08/15/2013] [Indexed: 11/30/2022]
Abstract
Glioma cell migration correlates with Pyk2 activity, but the intrinsic mechanism that regulates the activity of Pyk2 is not fully understood. Previous studies have supported a role for the N-terminal FERM domain in the regulation of Pyk2 activity as mutations in the FERM domain inhibit Pyk2 phosphorylation. To search for novel protein-protein interactions mediated by the Pyk2 FERM domain, we utilized a yeast two-hybrid genetic selection to identify the mammalian Ste20 homolog MAP4K4 as a binding partner for the Pyk2 FERM domain. MAP4K4 coimmunoprecipitated with Pyk2 and was a substrate for Pyk2 but did not coimmunoprecipitate with the closely related focal adhesion kinase FAK. Knockdown of MAP4K4 expression inhibited glioma cell migration and effectively blocked Pyk2 stimulation of glioma cell. Increased expression of MAP4K4 stimulated glioma cell migration; however, this stimulation was blocked by knockdown of Pyk2 expression. These data support that the interaction of MAP4K4 and Pyk2 is integrated with glioma cell migration and suggest that inhibition of this interaction may represent a potential therapeutic strategy to limit glioblastoma tumor dispersion.
Collapse
|
71
|
Yang YM, Gupta SK, Kim KJ, Powers BE, Cerqueira A, Wainger BJ, Ngo HD, Rosowski KA, Schein PA, Ackeifi CA, Arvanites AC, Davidow LS, Woolf CJ, Rubin LL. A small molecule screen in stem-cell-derived motor neurons identifies a kinase inhibitor as a candidate therapeutic for ALS. Cell Stem Cell 2013; 12:713-26. [PMID: 23602540 DOI: 10.1016/j.stem.2013.04.003] [Citation(s) in RCA: 236] [Impact Index Per Article: 19.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2012] [Revised: 03/08/2013] [Accepted: 04/01/2013] [Indexed: 12/13/2022]
Abstract
Amyotrophic lateral sclerosis (ALS) is a rapidly progressing neurodegenerative disease, characterized by motor neuron (MN) death, for which there are no truly effective treatments. Here, we describe a new small molecule survival screen carried out using MNs from both wild-type and mutant SOD1 mouse embryonic stem cells. Among the hits we found, kenpaullone had a particularly impressive ability to prolong the healthy survival of both types of MNs that can be attributed to its dual inhibition of GSK-3 and HGK kinases. Furthermore, kenpaullone also strongly improved the survival of human MNs derived from ALS-patient-induced pluripotent stem cells and was more active than either of two compounds, olesoxime and dexpramipexole, that recently failed in ALS clinical trials. Our studies demonstrate the value of a stem cell approach to drug discovery and point to a new paradigm for identification and preclinical testing of future ALS therapeutics.
Collapse
Affiliation(s)
- Yin M Yang
- Department of Stem Cell and Regenerative Biology, Harvard University, Cambridge, MA 02138, USA
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
72
|
Association of common genetic variants in the MAP4K4 locus with prediabetic traits in humans. PLoS One 2012; 7:e47647. [PMID: 23094072 PMCID: PMC3475716 DOI: 10.1371/journal.pone.0047647] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2012] [Accepted: 09/14/2012] [Indexed: 12/16/2022] Open
Abstract
Mitogen-activated protein kinase kinase kinase kinase 4 (MAP4K4) is expressed in all diabetes-relevant tissues and mediates cytokine-induced insulin resistance. We investigated whether common single nucleotide polymorphisms (SNPs) in the MAP4K4 locus associate with glucose intolerance, insulin resistance, impaired insulin release, or elevated plasma cytokines. The best hit was tested for association with type 2 diabetes. Subjects (N = 1,769) were recruited from the Tübingen Family (TÜF) study for type 2 diabetes and genotyped for tagging SNPs. In a subgroup, cytokines were measured. Association with type 2 diabetes was tested in a prospective case-cohort study (N = 2,971) derived from the EPIC-Potsdam study. Three SNPs (rs6543087, rs17801985, rs1003376) revealed nominal and two SNPs (rs11674694, rs11678405) significant associations with 2-hour glucose levels. SNPs rs6543087 and rs11674694 were also nominally associated with decreased insulin sensitivity. Another two SNPs (rs2236936, rs2236935) showed associations with reduced insulin release, driven by effects in lean subjects only. Three SNPs (rs11674694, rs13003883, rs2236936) revealed nominal associations with IL-6 levels. SNP rs11674694 was significantly associated with type 2 diabetes. In conclusion, common variation in MAP4K4 is associated with insulin resistance and β-cell dysfunction, possibly via this gene’s role in inflammatory signalling. This variation’s impact on insulin sensitivity may be more important since its effect on insulin release vanishes with increasing BMI.
Collapse
|
73
|
Agonistic and antagonistic roles for TNIK and MINK in non-canonical and canonical Wnt signalling. PLoS One 2012; 7:e43330. [PMID: 22984420 PMCID: PMC3439448 DOI: 10.1371/journal.pone.0043330] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2012] [Accepted: 07/19/2012] [Indexed: 12/03/2022] Open
Abstract
Wnt signalling is a key regulatory factor in animal development and homeostasis and plays an important role in the establishment and progression of cancer. Wnt signals are predominantly transduced via the Frizzled family of serpentine receptors to two distinct pathways, the canonical ß-catenin pathway and a non-canonical pathway controlling planar cell polarity and convergent extension. Interference between these pathways is an important determinant of cellular and phenotypic responses, but is poorly understood. Here we show that TNIK (Traf2 and Nck-interacting kinase) and MINK (Misshapen/NIKs-related kinase) MAP4K signalling kinases are integral components of both canonical and non-canonical pathways in Xenopus. xTNIK and xMINK interact and are proteolytically cleaved in vivo to generate Kinase domain fragments that are active in signal transduction, and Citron-NIK-Homology (CNH) Domain fragments that are suppressive. The catalytic activity of the Kinase domain fragments of both xTNIK and xMINK mediate non-canonical signalling. However, while the Kinase domain fragments of xTNIK also mediate canonical signalling, the analogous fragments derived from xMINK strongly antagonize this signalling. Our data suggest that the proteolytic cleavage of xTNIK and xMINK determines their respective activities and is an important factor in controlling the balance between canonical and non-canonical Wnt signalling in vivo.
Collapse
|
74
|
Wang X, Li JP, Chiu LL, Lan JL, Chen DY, Boomer J, Tan TH. Attenuation of T cell receptor signaling by serine phosphorylation-mediated lysine 30 ubiquitination of SLP-76 protein. J Biol Chem 2012; 287:34091-100. [PMID: 22902619 DOI: 10.1074/jbc.m112.371062] [Citation(s) in RCA: 41] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
SLP-76 (SH2 domain-containing leukocyte protein of 76 kDa) is an adaptor protein that is essential for T cell development and T cell receptor (TCR) signaling activation. Previous studies have identified an important negative feedback regulation of SLP-76 by HPK1 (hematopoietic progenitor kinase 1; MAP4K1)-induced Ser-376 phosphorylation. Ser-376 phosphorylation of SLP-76 mediates 14-3-3 binding, resulting in the attenuation of SLP-76 activation and downstream signaling; however, the underlying mechanism of this action remains unknown. Here, we report that phosphorylated SLP-76 is ubiquitinated and targeted for proteasomal degradation during TCR signaling. SLP-76 ubiquitination is mediated by Ser-376 phosphorylation. Furthermore, Lys-30 is identified as a ubiquitination site of SLP-76. Loss of Lys-30 ubiquitination of SLP-76 results in enhanced anti-CD3 antibody-induced ERK and JNK activation. These results reveal a novel regulation mechanism of SLP-76 by ubiquitination and proteasomal degradation of activated SLP-76, which is mediated by Ser-376 phosphorylation, leading to down-regulation of TCR signaling.
Collapse
Affiliation(s)
- Xiaohong Wang
- Department of Pathology and Immunology, Baylor College of Medicine, Houston, Texas 77030, USA
| | | | | | | | | | | | | |
Collapse
|
75
|
Kyriakis JM, Avruch J. Mammalian MAPK signal transduction pathways activated by stress and inflammation: a 10-year update. Physiol Rev 2012; 92:689-737. [PMID: 22535895 DOI: 10.1152/physrev.00028.2011] [Citation(s) in RCA: 1050] [Impact Index Per Article: 80.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
The mammalian stress-activated families of mitogen-activated protein kinases (MAPKs) were first elucidated in 1994, and by 2001, substantial progress had been made in identifying the architecture of the pathways upstream of these kinases as well as in cataloguing candidate substrates. This information remains largely sound. Nevertheless, an informed understanding of the physiological and pathophysiological roles of these kinases remained to be accomplished. In the past decade, there has been an explosion of new work using RNAi in cells, as well as transgenic, knockout and conditional knockout technology in mice that has provided valuable insight into the functions of stress-activated MAPK pathways. These findings have important implications in our understanding of organ development, innate and acquired immunity, and diseases such as atherosclerosis, tumorigenesis, and type 2 diabetes. These new developments bring us within striking distance of the development and validation of novel treatment strategies. Herein we first summarize the molecular components of the mammalian stress-regulated MAPK pathways and their regulation as described thus far. We then review some of the in vivo functions of these pathways.
Collapse
Affiliation(s)
- John M Kyriakis
- Molecular Cardiology Research Institute, Tufts Medical Center, 800 Washington St., Box 8486, Boston, MA 02111, USA.
| | | |
Collapse
|
76
|
Wang X, Li JP, Kuo HK, Chiu LL, Dement GA, Lan JL, Chen DY, Yang CY, Hu H, Tan TH. Down-regulation of B cell receptor signaling by hematopoietic progenitor kinase 1 (HPK1)-mediated phosphorylation and ubiquitination of activated B cell linker protein (BLNK). J Biol Chem 2012; 287:11037-48. [PMID: 22334673 DOI: 10.1074/jbc.m111.310946] [Citation(s) in RCA: 48] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
Hematopoietic progenitor kinase 1 (HPK1) is a Ste20-like serine/threonine kinase that suppresses immune responses and autoimmunity. B cell receptor (BCR) signaling activates HPK1 by inducing BLNK/HPK1 interaction. Whether HPK1 can reciprocally regulate BLNK during BCR signaling is unknown. Here, we show that HPK1-deficient B cells display hyper-proliferation and hyper-activation of IκB kinase and MAPKs (ERK, p38, and JNK) upon the ligation of BCR. HPK1 attenuates BCR-induced cell activation via inducing BLNK threonine 152 phosphorylation, which mediates BLNK/14-3-3 binding. Furthermore, threonine 152-phosphorylated BLNK is ubiquitinated at lysine residues 37, 38, and 42, leading to attenuation of MAPK and IκB kinase activation in B cells during BCR signaling. These results reveal a novel negative feedback regulation of BCR signaling by HPK1-mediated phosphorylation, ubiquitination, and subsequent degradation of the activated BLNK.
Collapse
Affiliation(s)
- Xiaohong Wang
- Department of Pathology and Immunology, Baylor College of Medicine, Houston, Texas 77030, USA
| | | | | | | | | | | | | | | | | | | |
Collapse
|
77
|
Zoumaro-Djayoon AD, Ding V, Foong LY, Choo A, Heck AJR, Muñoz J. Investigating the role of FGF-2 in stem cell maintenance by global phosphoproteomics profiling. Proteomics 2011; 11:3962-71. [PMID: 21761559 DOI: 10.1002/pmic.201100048] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2011] [Revised: 03/02/2011] [Accepted: 04/12/2011] [Indexed: 12/26/2022]
Abstract
Human embryonic stem cells (hESCs) are of immense interest for regenerative medicine as a source of tissue replacement. Expansion of hESCs as a pluripotent population requires a balance between survival, proliferation and self-renewal signals. One of the key growth factors that maintains this balance is fibroblast growth factor-2 (FGF-2). However, the underlying molecular mechanisms are poorly understood. We recently profiled specifically tyrosine phosphorylation events that occur during FGF-2 stimulation of hESCs (Ding et al., J. Cell. Physiol. 2010, 225, 417-428). Here, we complement this phosphoproteome profiling by analyzing temporal dynamics of mostly serine and threonine protein phosphorylation events. Our multi-dimensional strategy combines strong cation exchange chromatography to reduce the sample complexity followed by titanium dioxide off-line for the enrichment of phosphopeptides and dimethylation-based stable isotope labeling for quantification. This approach allowed us to identify and quantify 3261 unique proteins from which 1064 proteins were found to be phosphorylated in one or more residues (representing 1653 unique phosphopeptides). Approximately 40% of the proteins (553 unique phosphopeptides) showed differential phosphorylation upon FGF-2 treatment. Among those are several members of the canonical pathways involved in pluripotency and self-renewal (e.g. Wnt and PI3K/AKT), hESC-associated proteins such as SOX2, RIF1, SALL4, DPPA4, DNMT3B and 53 proteins that are target genes of the pluripotency transcription factors SOX2, OCT4 and NANOG. These findings complement existing pluripotency analyses and provide new insights into how FGF-2 assists in maintaining the undifferentiated state of hESCs.
Collapse
Affiliation(s)
- Adja D Zoumaro-Djayoon
- Bijvoet Center for Biomolecular Research and Utrecht Institute for Pharmaceutical Sciences, Utrecht University, Utrecht, The Netherlands
| | | | | | | | | | | |
Collapse
|
78
|
Guimarães CRW, Rai BK, Munchhof MJ, Liu S, Wang J, Bhattacharya SK, Buckbinder L. Understanding the Impact of the P-loop Conformation on Kinase Selectivity. J Chem Inf Model 2011; 51:1199-204. [DOI: 10.1021/ci200153c] [Citation(s) in RCA: 70] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Affiliation(s)
- Cristiano R. W. Guimarães
- Worldwide Medicinal Chemistry Department, ‡Computational Sciences Center of Emphasis, §Department of Structural Biology and Biophysics, and ∥CVMED - Target Exploration, Pfizer Global Research and Development, Eastern Point Road, Groton, Connecticut 06340, United States
| | - Brajesh K. Rai
- Worldwide Medicinal Chemistry Department, ‡Computational Sciences Center of Emphasis, §Department of Structural Biology and Biophysics, and ∥CVMED - Target Exploration, Pfizer Global Research and Development, Eastern Point Road, Groton, Connecticut 06340, United States
| | - Michael J. Munchhof
- Worldwide Medicinal Chemistry Department, ‡Computational Sciences Center of Emphasis, §Department of Structural Biology and Biophysics, and ∥CVMED - Target Exploration, Pfizer Global Research and Development, Eastern Point Road, Groton, Connecticut 06340, United States
| | - Shenping Liu
- Worldwide Medicinal Chemistry Department, ‡Computational Sciences Center of Emphasis, §Department of Structural Biology and Biophysics, and ∥CVMED - Target Exploration, Pfizer Global Research and Development, Eastern Point Road, Groton, Connecticut 06340, United States
| | - Jian Wang
- Worldwide Medicinal Chemistry Department, ‡Computational Sciences Center of Emphasis, §Department of Structural Biology and Biophysics, and ∥CVMED - Target Exploration, Pfizer Global Research and Development, Eastern Point Road, Groton, Connecticut 06340, United States
| | - Samit K. Bhattacharya
- Worldwide Medicinal Chemistry Department, ‡Computational Sciences Center of Emphasis, §Department of Structural Biology and Biophysics, and ∥CVMED - Target Exploration, Pfizer Global Research and Development, Eastern Point Road, Groton, Connecticut 06340, United States
| | - Leonard Buckbinder
- Worldwide Medicinal Chemistry Department, ‡Computational Sciences Center of Emphasis, §Department of Structural Biology and Biophysics, and ∥CVMED - Target Exploration, Pfizer Global Research and Development, Eastern Point Road, Groton, Connecticut 06340, United States
| |
Collapse
|
79
|
Han SX, Zhu Q, Ma JL, Zhao J, Huang C, Jia X, Zhang D. Lowered HGK expression inhibits cell invasion and adhesion in hepatocellular carcinoma cell line HepG2. World J Gastroenterol 2010; 16:4541-8. [PMID: 20857524 PMCID: PMC2945485 DOI: 10.3748/wjg.v16.i36.4541] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
AIM: To investigate the effects of RNA interference targeting hepatocyte progenitor kinase-like kinase (HGK) in the invasion and adhesion of hepatocellular carcinoma (HCC) cell line HepG2.
METHODS: Three paired insert DNA fragments specific to HGK gene and one negative control DNA fragment were synthesized and inserted into RNAi-Ready pSIREN-RetroQ-ZsGreen vector. Western blotting assay and real-time reverse transcriptase polymerase chain reaction (RT-PCR) were used to screen the vector with a highest inhibitory rate. The vector was used to generate recombinant retrovirus specific to HGK. 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyl-2h-tetrazolium bromide (MTT) assay was used to examine cell growth; wound closure assay and cell adhesion assay were employed to investigate cell migration and adhesion respectively; and transwell assay and three-dimensional culture invasion assay were used to detect cell invasion. The expressions of matrix metalloproteinase (MMP)-2, MMP-9 and nuclear factor (NF)-κB were detected by Western blotting assay.
RESULTS: The real time RT-PCR and Western blotting assay showed that cells transfected with retrovirus mediating RNAi targeting of HGK (RV-shHGK)-1 vector had the strongest inhibition of HGK protein, with an inhibition rate of 76%, and this vector was used to generate recombinant retrovirus RV-shHGK-1. Cell adhesion assay and MTT assay found that cell adhesion and growth of the cells infected with RV-shHGK-1 were significantly lower than those of the control cells (P < 0.05). Wound closure assay, transwell assay and three-dimensional culture invasion assay showed that the cell invasiveness was significantly less in HGK knockdown cells than in the control cells (P < 0.05). The expressions of MMP-2, MMP-9 and NF-κB were inhibited in HepG2 cells infected with RV-shHGK-1.
CONCLUSION: Down-regulation of HGK can obviously inhibit the migration and invasion of HepG2 cells in vitro. HGK may be a new therapeutic target for treatment of HCC.
Collapse
|
80
|
Guntur KVP, Guilherme A, Xue L, Chawla A, Czech MP. Map4k4 negatively regulates peroxisome proliferator-activated receptor (PPAR) gamma protein translation by suppressing the mammalian target of rapamycin (mTOR) signaling pathway in cultured adipocytes. J Biol Chem 2010; 285:6595-603. [PMID: 20038583 PMCID: PMC2825455 DOI: 10.1074/jbc.m109.068502] [Citation(s) in RCA: 29] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2009] [Revised: 12/09/2009] [Indexed: 01/25/2023] Open
Abstract
The receptor peroxisome proliferator-activated receptor gamma (PPARgamma) is considered a master regulator of adipocyte differentiation and promotes glucose and lipid metabolism in mature adipocytes. We recently identified the yeast Sterile 20 (Ste20) protein kinase ortholog, Map4k4, in an RNA interference-based screen as an inhibitor of PPARgamma expression in cultured adipocytes. Here, we show that RNA interference-mediated silencing of Map4k4 elevates the levels of both PPARgamma1 and PPARgamma2 proteins in 3T3-L1 adipocytes without affecting PPARgamma mRNA levels, suggesting that Map4k4 regulates PPARgamma at a post-transcriptional step. PPARgamma degradation rates are remarkably rapid as measured in the presence of cycloheximide (t(1/2) = 2 h), but silencing Map4k4 had no effect on PPARgamma degradation. However, depletion of Map4k4 significantly enhances [(35)S]methionine/cysteine incorporation into proteins, suggesting that Map4k4 signaling decreases protein translation. We show a function of Map4k4 is to inhibit rapamycin-sensitive mammalian target of rapamycin (mTOR) activity, decreasing 4E-BP1 phosphorylation. In addition, our results show mTOR and 4E-BP1 are required for the increased PPARgamma protein expression upon Map4k4 knockdown. Consistent with this concept, adenovirus-mediated expression of Map4k4 decreased PPARgamma protein levels and mTOR phosphorylation. These data show that Map4k4 negatively regulates PPARgamma post-transcriptionally, by attenuating mTOR signaling and a 4E-BP1-dependent mechanism.
Collapse
Affiliation(s)
- Kalyani V. P. Guntur
- From the Program in Molecular Medicine, University of Massachusetts Medical School, Worcester, Massachusetts 01605
| | - Adilson Guilherme
- From the Program in Molecular Medicine, University of Massachusetts Medical School, Worcester, Massachusetts 01605
| | - Liting Xue
- From the Program in Molecular Medicine, University of Massachusetts Medical School, Worcester, Massachusetts 01605
| | - Anil Chawla
- From the Program in Molecular Medicine, University of Massachusetts Medical School, Worcester, Massachusetts 01605
| | - Michael P. Czech
- From the Program in Molecular Medicine, University of Massachusetts Medical School, Worcester, Massachusetts 01605
| |
Collapse
|
81
|
Mitomycin C-treated antigen-presenting cells as a tool for control of allograft rejection and autoimmunity: from bench to bedside. Hum Immunol 2009; 70:506-12. [PMID: 19393276 DOI: 10.1016/j.humimm.2009.04.024] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2009] [Accepted: 04/15/2009] [Indexed: 01/09/2023]
Abstract
Cells have been previously used in experimental models for tolerance induction in organ transplantation and autoimmune diseases. One problem with the therapeutic use of cells is standardization of their preparation. We discuss an immunosuppressive strategy relying on cells irreversibly transformed by a chemotherapeutic drug. Dendritic cells (DCs) of transplant donors pretreated with mitomycin C (MMC) strongly prolonged rat heart allograft survival when injected into recipients before transplantation. Likewise, MMC-DCs loaded with myelin basic protein suppressed autoreactive T cells of MS patients in vitro and prevented experimental autoimmune encephalitis in mice. Comprehensive gene microarray analysis identified genes that possibly make up the suppressive phenotype, comprising glucocorticoid leucine zipper, immunoglobulin-like transcript 3, CD80, CD83, CD86, and apoptotic genes. Based on these findings, a hypothetical model of tolerance induction by MMC-treated DCs is delineated. Finally, we describe the first clinical application of MMC-treated monocyte-enriched donor cells in an attempt to control the rejection of a haploidentical stem cell transplant in a sensitized recipient and discuss the pros and cons of using MMC-treated antigen-presenting cells for tolerance induction. Although many questions remain, MMC-treated cells are a promising clinical tool for controlling allograft rejection and deleterious immune responses in autoimmune diseases.
Collapse
|
82
|
Süveges D, Gáspári Z, Tóth G, Nyitray L. Charged single alpha-helix: a versatile protein structural motif. Proteins 2009; 74:905-16. [PMID: 18712826 DOI: 10.1002/prot.22183] [Citation(s) in RCA: 62] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
A few highly charged natural peptide sequences were recently suggested to form stable alpha-helical structures in water. In this article we show that these sequences represent a novel structural motif called "charged single alpha-helix" (CSAH). To obtain reliable candidate CSAH motifs, we developed two conceptually different computational methods capable of scanning large databases: SCAN4CSAH is based on sequence features characteristic for salt bridge stabilized single alpha-helices, whereas FT_CHARGE applies Fourier transformation to charges along sequences. Using the consensus of the two approaches, a remarkable number of proteins were found to contain putative CSAH domains. Recombinant fragments (50-60 residues) corresponding to selected hits obtained by both methods (myosin 6, Golgi resident protein GCP60, and M4K4 protein kinase) were produced and shown by circular dichroism spectroscopy to adopt largely alpha-helical structure in water. CSAH segments differ substantially both from coiled-coil and intrinsically disordered proteins, despite the fact that current prediction methods recognize them as either or both. Analysis of the proteins containing CSAH motif revealed possible functional roles of the corresponding segments. The suggested main functional features include the formation of relatively rigid spacer/connector segments between functional domains as in caldesmon, extension of the lever arm in myosin motors and mediation of transient interactions by promoting dimerization in a range of proteins.
Collapse
Affiliation(s)
- Dániel Süveges
- Department of Biochemistry, Eötvös Loránd University, Pázmány Péter s. 1/C, H1117 Budapest, Hungary
| | | | | | | |
Collapse
|
83
|
Liang JJ, Wang H, Rashid A, Tan TH, Hwang RF, Hamilton SR, Abbruzzese JL, Evans DB, Wang H. Expression of MAP4K4 is associated with worse prognosis in patients with stage II pancreatic ductal adenocarcinoma. Clin Cancer Res 2008; 14:7043-9. [PMID: 18981001 DOI: 10.1158/1078-0432.ccr-08-0381] [Citation(s) in RCA: 90] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
PURPOSE Mitogen-activated protein 4 kinase 4 (MAP4K4) is a serine/threonine kinase and belongs to the mammalian STE20/MAP4K family. Recent studies have shown that MAP4K4 is overexpressed in many types of human cancer and cancer cell lines. MAP4K4 plays an important role in transformation, invasiveness, adhesion, and cell migration. However, the expression of MAP4K4 and its significance in pancreatic ductal adenocarcinoma (PDA) has not been studied. EXPERIMENTAL DESIGN We examined the expression of MAP4K4 by immunohistochemistry using tissue microarrays consisting of 66 stage II PDA and their paired benign pancreatic tissue. The staining results were categorized as MAP4K4-H or MAP4K4-L. The results were correlated with clinicopathologic features and patient survival. RESULTS MAP4K4 was overexpressed (MAP4K4-H) in 30 of 66 (46%) PDAs and was higher than the paired benign pancreatic tissue samples (19%; P=0.002). The median overall and recurrence-free survival for patients with MAP4K4-H PDAs were 19.5 and 9.3 months, respectively, compared with 65.2 and 28.8 months for patients with MAP4K4-L tumor (P=0.02 and 0.0005, log-rank test). MAP4K4 expression was associated with poor overall and recurrence-free survival in univariate analysis (P=0.02 and 0.001). In multivariate analysis, MAP4K4 expression significantly correlated with overall and recurrence-free survival (P=0.025 and 0.004) independent of age, tumor size, differentiation, and stage. MAP4K4 expression was also associated with higher frequency of recurrence/metastasis, larger tumor size, and increased number of positive lymph nodes (P<0.05). CONCLUSION MAP4K4 was overexpressed in about half of PDAs. Overexpression of MAP4K4 was associated with worse prognosis and is a prognostic marker for stage II PDAs.
Collapse
Affiliation(s)
- John J Liang
- Department of Pathology, The University of Texas M. D. Anderson Cancer, Houston, TX 77030, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
84
|
Fu S, Zhao H, Shi J, Abzhanov A, Crawford K, Ohno-Machado L, Zhou J, Du Y, Kuo WP, Zhang J, Jiang M, Jin JG. Peripheral arterial occlusive disease: global gene expression analyses suggest a major role for immune and inflammatory responses. BMC Genomics 2008; 9:369. [PMID: 18673543 PMCID: PMC2529314 DOI: 10.1186/1471-2164-9-369] [Citation(s) in RCA: 36] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2007] [Accepted: 08/01/2008] [Indexed: 11/17/2022] Open
Abstract
Background Peripheral arterial disease (PAD), a major manifestation of atherosclerosis, is associated with significant cardiovascular morbidity, limb loss and death. However, mechanisms underlying the genesis and progression of the disease are far from clear. Genome-wide gene expression profiling of clinical samples may represent an effective approach to gain relevant information. Results After histological classification, a total of 30 femoral artery samples, including 11 intermediate lesions, 14 advanced lesions and 5 normal femoral arteries, were profiled using Affymetrix microarray platform. Following real-time RT-PCR validation, different algorithms of gene selection and clustering were applied to identify differentially expressed genes. Under a stringent cutoff, i.e., a false discovery rate (FDR) <0.5%, we found 366 genes were differentially regulated in intermediate lesions and 447 in advanced lesions. Of these, 116 genes were overlapped between intermediate and advanced lesions, including 68 up-regulated genes and 48 down-regulated ones. In these differentially regulated genes, immune/inflammatory genes were significantly up-regulated in different stages of PAD, (85/230 in intermediate lesions, 37/172 in advanced lesions). Through literature mining and pathway analysis using different databases such as Gene Ontology (GO), and the Kyoto Encyclopedia of Gene and Genomics (KEGG), genes involved in immune/inflammatory responses were significantly enriched in up-regulated genes at different stages of PAD(p < 0.05), revealing a significant correlation between immune/inflammatory responses and disease progression. Moreover, immune-related pathways such as Toll-like receptor signaling and natural killer cell mediated cytotoxicity were particularly enriched in intermediate and advanced lesions (P < 0.05), highlighting their pathogenic significance during disease progression. Conclusion Lines of evidence revealed in this study not only support previous hypotheses, primarily based on studies of animal models and other types of arterial disease, that inflammatory responses may influence the development of PAD, but also permit the recognition of a wide spectrum of immune/inflammatory genes that can serve as signatures for disease progression in PAD. Further studies of these signature molecules may eventually allow us to develop more sophisticated protocols for pharmaceutical interventions.
Collapse
Affiliation(s)
- Shijun Fu
- Institute of Health Sciences, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences and Shanghai Jiao Tong University School of Medicine, Shanghai, PR China.
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
85
|
Ehrlich M, Sanchez C, Shao C, Nishiyama R, Kehrl J, Kuick R, Kubota T, Hanash SM. ICF, an immunodeficiency syndrome: DNA methyltransferase 3B involvement, chromosome anomalies, and gene dysregulation. Autoimmunity 2008; 41:253-71. [PMID: 18432406 PMCID: PMC2430169 DOI: 10.1080/08916930802024202] [Citation(s) in RCA: 94] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2007] [Accepted: 01/02/2008] [Indexed: 02/07/2023]
Abstract
The immunodeficiency, centromeric region instability, and facial anomalies syndrome (ICF) is the only disease known to result from a mutated DNA methyltransferase gene, namely, DNMT3B. Characteristic of this recessive disease are decreases in serum immunoglobulins despite the presence of B cells and, in the juxtacentromeric heterochromatin of chromosomes 1 and 16, chromatin decondensation, distinctive rearrangements, and satellite DNA hypomethylation. Although DNMT3B is involved in specific associations with histone deacetylases, HP1, other DNMTs, chromatin remodelling proteins, condensin, and other nuclear proteins, it is probably the partial loss of catalytic activity that is responsible for the disease. In microarray experiments and real-time RT-PCR assays, we observed significant differences in RNA levels from ICF vs. control lymphoblasts for pro- and anti-apoptotic genes (BCL2L10, CASP1, and PTPN13); nitrous oxide, carbon monoxide, NF-kappaB, and TNFalpha signalling pathway genes (PRKCH, GUCY1A3, GUCY1B3, MAPK13; HMOX1, and MAP4K4); and transcription control genes (NR2F2 and SMARCA2). This gene dysregulation could contribute to the immunodeficiency and other symptoms of ICF and might result from the limited losses of DNA methylation although ICF-related promoter hypomethylation was not observed for six of the above examined genes. We propose that hypomethylation of satellite 2 at 1qh and 16qh might provoke this dysregulation gene expression by trans effects from altered sequestration of transcription factors, changes in nuclear architecture, or expression of noncoding RNAs.
Collapse
Affiliation(s)
- Melanie Ehrlich
- Hayward Human Genetics Program, Tulane Medical School, New Orleans, LA 70112, USA.
| | | | | | | | | | | | | | | |
Collapse
|
86
|
Abstract
Mitogen-activated protein kinase (MAPK) cascades are central pathways that participate in the intracellular transmission of extracellular signals. Each of the MAPK signaling cascades seems to consist of three to five tiers of protein kinases that sequentially activate each other by phosphorylation. Since the majority of MAPK cascade components are kinases, the methods used to detect their activation involve determining phosphorylation state and protein kinase activities. The primary method describes the use of immunoblotting with specific anti-phospho antibody to detect activation of MAPK components. Alternative methods described are immunoprecipitation of desired protein kinases followed by phosphorylation of specific substrates and the use of an in-gel kinase assay. These methods have proven useful in the study of the MAPK signaling cascades.
Collapse
Affiliation(s)
- Yoav Shaul
- Department of Biological Regulation, The Weizmann Institute of Science, Rehovot, Israel
| | | |
Collapse
|
87
|
Shaul Y, Seger R. The detection of MAPK signaling. CURRENT PROTOCOLS IN CELL BIOLOGY 2008; Chapter 14:Unit 14.3. [PMID: 18228462 DOI: 10.1002/0471143030.cb1403s28] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/11/2023]
Abstract
Mitogen-activated protein kinase (MAPK) cascades are central pathways that participate in the intracellular transmission of extracellular signals. Each of the MAPK signaling cascades seems to consist of three to five tiers of protein kinases that sequentially activate each other by phosphorylation. Since the majority of MAPK cascade components are kinases, the methods used to detect their activation involve determining phosphorylation state and protein kinase activities. The Basic Protocol describes the use of immunoblotting with specific anti-phospho antibody to detect activation of MAPK components. Alternative methods described are immunoprecipitation of desired protein kinases followed by phosphorylation of specific substrates and the use of an in-gel kinase assay. These methods have proven useful in the study of the MAPK signaling cascades.
Collapse
Affiliation(s)
- Yoav Shaul
- Department of Biological Regulation, The Weizmann Institute of Science, Rehovot, Israel
| | | |
Collapse
|
88
|
Puri V, Virbasius JV, Guilherme A, Czech MP. RNAi screens reveal novel metabolic regulators: RIP140, MAP4k4 and the lipid droplet associated fat specific protein (FSP) 27. Acta Physiol (Oxf) 2008; 192:103-15. [PMID: 18171433 DOI: 10.1111/j.1748-1716.2007.01786.x] [Citation(s) in RCA: 50] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Adipose tissue modulates whole body metabolism and insulin sensitivity by controlling circulating lipid levels and producing molecules that can regulate fatty acid metabolism in such tissues as muscle and liver. We have developed RNA interference (RNAi) screens to identify genes in cultured adipocytes that regulate insulin signalling and key metabolic pathways. These short interfering RNA (siRNA)-based screens identified the transcriptional corepressor receptor interacting protein 140 (RIP140) (J Clin Invest 116: 125, 2006) and the mitogen-activated protein kinase (MAP4k4) (Proc Natl Acad Sci USA 103: 2087, 2006) as negative regulators of insulin-responsive hexose uptake and oxidative metabolism. Gene expression profiling revealed that RIP140 depletion upregulates the expression of clusters of genes in the pathways of glucose uptake, glycolysis, tricarboxylic acid cycle, fatty acid oxidation, mitochondrial biogenesis and oxidative phosphorylation. RIP140-null mice resist weight gain on a high-fat diet and display enhanced glucose tolerance. MAP4k4 depletion in adipocytes increases many of the RIP140-sensitive genes, increases adipogenesis and mediates some actions of tumour necrosis factor-alpha (TNF-alpha). Remarkably, another hit in our RNAi screens was fat specific protein 27 (FSP27), a highly expressed isoform of Cidea. We discovered that FSP27 unexpectedly associates specifically with lipid droplets and regulates fat storage. We conclude that RIP140, MAP4k4 and the novel lipid droplet protein FSP27 are powerful regulators of adipose tissue metabolism and are potential therapeutic targets for controlling metabolic disease. The discovery of these novel proteins validates the power of RNAi screening for discovery of new therapeutic approaches to type 2 diabetes and obesity.
Collapse
Affiliation(s)
- V Puri
- Program in Molecular Medicine, University of Massachusetts Medical School, Worcester, MA 01605, USA
| | | | | | | |
Collapse
|
89
|
Tesz GJ, Guilherme A, Guntur KVP, Hubbard AC, Tang X, Chawla A, Czech MP. Tumor necrosis factor alpha (TNFalpha) stimulates Map4k4 expression through TNFalpha receptor 1 signaling to c-Jun and activating transcription factor 2. J Biol Chem 2007; 282:19302-12. [PMID: 17500068 DOI: 10.1074/jbc.m700665200] [Citation(s) in RCA: 71] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
Tumor necrosis factor alpha (TNFalpha) is a cytokine secreted by macrophages and adipocytes that contributes to the low grade inflammation and insulin resistance observed in obesity. TNFalpha signaling decreases peroxisome proliferator-activated receptor gamma and glucose transporter isoform 4 (GLUT4) expression in adipocytes, impairing insulin action, and this is mediated in part by the yeast Ste20 protein kinase ortholog Map4k4. Here we show that Map4k4 expression is selectively up-regulated by TNFalpha, whereas the expression of the protein kinases JNK1/2, ERK1/2, p38 stress-activated protein kinase, and mitogen-activated protein kinase kinases 4/7 shows little or no response. Furthermore, the cytokines interleukin 1beta (IL-1beta) and IL-6 as well as lipopolysaccharide fail to increase Map4k4 mRNA levels in cultured adipocytes under conditions where TNFalpha elicits a 3-fold effect. Using agonistic and antagonistic antibodies and small interfering RNA (siRNA) against TNFalpha receptor 1 (TNFR1) and TNFalpha receptor 2 (TNFR2), we show that TNFR1, but not TNFR2, mediates the increase in Map4k4 expression. TNFR1, but not TNFR2, also mediates a potent effect of TNFalpha on the phosphorylation of JNK1/2 and p38 stress-activated protein kinase and their downstream transcription factor substrates c-Jun and activating transcription factor 2 (ATF2). siRNA-based depletion of c-Jun and ATF2 attenuated TNFalpha action on Map4k4 mRNA expression. Consistent with this concept, the phosphorylation of ATF2 along with the expression and phosphorylation of c-Jun by TNFalpha signaling was more robust and prolonged compared with that of IL-1beta, which failed to modulate Map4k4. These data reveal that TNFalpha selectively stimulates the expression of a key component of its own signaling pathway, Map4k4, through a TNFR1-dependent mechanism that targets the transcription factors c-Jun and ATF2.
Collapse
Affiliation(s)
- Gregory J Tesz
- Program in Molecular Medicine, University of Massachusetts Medical School, Worcester, Massachusetts 01605, USA
| | | | | | | | | | | | | |
Collapse
|
90
|
Bouzakri K, Zierath JR. MAP4K4 gene silencing in human skeletal muscle prevents tumor necrosis factor-alpha-induced insulin resistance. J Biol Chem 2007; 282:7783-9. [PMID: 17227768 DOI: 10.1074/jbc.m608602200] [Citation(s) in RCA: 114] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023] Open
Abstract
Tumor necrosis factor-alpha (TNF-alpha) induces skeletal muscle insulin resistance by impairing insulin signaling events involved in GLUT4 translocation. We tested whether mitogenic-activated protein kinase kinase kinase kinase isoform 4 (MAP4K4) causes the TNF-alpha-induced negative regulation of extracellular signal-regulated kinase-1/2 (ERK-1/2), c-Jun NH2-terminal kinase (JNK), and the insulin receptor substrate-1 (IRS-1) on the insulin signaling pathway governing glucose metabolism. Using small interfering RNA (siRNA) to suppress the expression of MAP4K4 protein 85% in primary human skeletal muscle cells, we provide evidence that TNF-alpha-induced insulin resistance on glucose uptake was completely prevented. MAP4K4 silencing inhibited TNF-alpha-induced negative signaling inputs by preventing excessive JNK and ERK-1/2 phosphorylation, as well as IRS-1 serine phosphorylation. These results highlight the MAPK4K4/JNK/ERK/IRS module in the negative regulation of insulin signaling to glucose transport in response to TNF-alpha. Depletion of MAP4K4 also prevented TNF-alpha-induced insulin resistance on Akt and the Akt substrate 160 (AS160), providing evidence that appropriate insulin signaling inputs for glucose metabolism were rescued. Silencing of MAP2K1 and MAP2K4, signaling proteins downstream of MAP4K4, recapitulated the effect of MAP4K4 siRNA in TNF-alpha-treated cells. Thus, strategies to inhibit MAP4K4 may be efficacious in the prevention of TNF-alpha-induced inhibitory signals that cause skeletal muscle insulin resistance on glucose metabolism in humans. Moreover, in myotubes from insulin-resistant type II diabetic patients, siRNA against MAP4K4, MAP2K4, or MAP2K1 restored insulin action on glucose uptake to levels observed in healthy subjects. Collectively, our results demonstrate that MAP4K4 silencing prevents insulin resistance in human skeletal muscle and restores appropriate signaling inputs to enhance glucose uptake.
Collapse
Affiliation(s)
- Karim Bouzakri
- Department of Molecular Medicine and Surgery, Karolinska University Hospital, Karolinska Institutet, S-171 77 Stockholm, Sweden
| | | |
Collapse
|
91
|
Wang T, Lee K, Rehman A, Daoud SS. PRIMA-1 induces apoptosis by inhibiting JNK signaling but promoting the activation of Bax. Biochem Biophys Res Commun 2007; 352:203-12. [PMID: 17113036 DOI: 10.1016/j.bbrc.2006.11.006] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2006] [Accepted: 11/02/2006] [Indexed: 10/23/2022]
Abstract
The p53 protein plays a major role in the maintenance of genome stability in mammalian cells. Mutations of p53 occur in over 40% of breast cancers and are indicative of tumor resistance to chemotherapeutic agents. Recently, there has been a high degree of interest in pharmacological approaches for restoring the normal function to mutant p53. The low molecular weight compound p53 reactivation and induction of massive apoptosis (PRIMA-1) was shown to induce cytotoxic effects and apoptosis in human tumor cells with mutant p53. Here, we studied the molecular mechanisms of PRIMA-1-induced apoptosis in human breast cancer cells with p53 mutations such as MDA-231 and GI-101A as compared to MCF-7 cells. We show that PRIMA-1 selectively induces apoptosis in human breast cancer cells MDA-231 and GI-101A compared to the MCF-7. This effect was paralleled by an increase in total p53 level in the nucleus and the induction of its phosphorylation at Ser-15 site. Using the chromatin immunoprecipitation (ChIP) assays, we show that PRIMA-1 restored p53 DNA binding activity to the promoters of the proapoptotic genes such as Bax and PUMA, but inhibited the binding activity to the promoters of the MAP4K4 gene. Knockdown of p53 protein in breast cancer cells using siRNA followed by PRIMA-1 treatment resulted in decline of Bax and PUMA proteins expression. Cell incubation with either PRIMA-1 or SP600125 (c-Jun NH2-terminal kinase inhibitor) resulted in the abrogation of adriamycin-induced c-Jun NH2-terminal kinase (JNK) activation, whereas Bax activation was not inhibited. We conclude that both Bax and PUMA but not JNK signaling are involved in PRIMA-1-induced apoptosis in breast cancer cells with p53 mutation.
Collapse
Affiliation(s)
- Tao Wang
- Department of Pharmaceutical Sciences, Washington State University, Pullman, WA, USA
| | | | | | | |
Collapse
|
92
|
Mao J, Ligon KL, Rakhlin EY, Thayer SP, Bronson RT, Rowitch D, McMahon AP. A novel somatic mouse model to survey tumorigenic potential applied to the Hedgehog pathway. Cancer Res 2006; 66:10171-8. [PMID: 17047082 PMCID: PMC3806052 DOI: 10.1158/0008-5472.can-06-0657] [Citation(s) in RCA: 220] [Impact Index Per Article: 11.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
We report a novel mouse model for the generation of sporadic tumors and show the efficiency of this approach by surveying Hedgehog (Hh)-related tumors. Up-regulation of the Hh pathway is achieved by conditionally regulated expression of an activated allele of Smoothened (R26-SmoM2) using either sporadic leakage or global postnatal induction of a ubiquitously expressed inducible Cre transgene (CAGGS-CreER). Following postnatal tamoxifen induction, CAGGS-CreER; R26-SmoM2 mice developed tumors with short latency and high penetrance. All mice exhibited rhabdomyosarcoma and basal cell carcinoma; 40% also developed medulloblastoma. In addition, mice showed a novel pancreatic lesion resembling low-grade mucinous cystic neoplasms in humans. In contrast, widespread activation of SmoM2 in the postnatal prostate epithelium results in no detectable morphologic outcome in 12-month-old mice. Comparison of gene expression profiles among diverse tumors identified several signature genes, including components of platelet-derived growth factor and insulin-like growth factor pathways, which may provide a common mechanistic link to the Hh-related malignancies. This experimental model provides a robust tool for exploring the process of Hh-dependent tumorigenesis and the treatment of such tumors. More generally, this approach provides a genetic platform for identifying tumorigenic potential in putative oncogenes and tumor suppressors and for more effective modeling of sporadic cancers in mice.
Collapse
Affiliation(s)
- Junhao Mao
- Department of Molecular and Cellular Biology, Harvard University, Cambridge, Massachusetts
| | - Keith L. Ligon
- Department of Pediatric Oncology, Dana-Farber Cancer Institute and Harvard Medical Schoo, Boston, Massachusetts
- Department of Pathology, Brigham and Women’s Hospital, Boston, Massachusetts
| | - Elena Y. Rakhlin
- Department of Surgery, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts
| | - Sarah P. Thayer
- Department of Surgery, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts
| | - Roderick T. Bronson
- Department of Biomedical Sciences, Tufts University Veterinary School, North Grafton, Massachusetts
| | - David Rowitch
- Department of Pediatric Oncology, Dana-Farber Cancer Institute and Harvard Medical Schoo, Boston, Massachusetts
| | - Andrew P. McMahon
- Department of Molecular and Cellular Biology, Harvard University, Cambridge, Massachusetts
| |
Collapse
|
93
|
Shui JW, Boomer JS, Han J, Xu J, Dement GA, Zhou G, Tan TH. Hematopoietic progenitor kinase 1 negatively regulates T cell receptor signaling and T cell-mediated immune responses. Nat Immunol 2006; 8:84-91. [PMID: 17115060 DOI: 10.1038/ni1416] [Citation(s) in RCA: 151] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2006] [Accepted: 10/26/2006] [Indexed: 11/09/2022]
Abstract
HPK1 is a Ste20-related serine-threonine kinase that inducibly associates with the adaptors SLP-76 and Gads after T cell receptor (TCR) signaling. Here, HPK1 deficiency resulted in enhanced TCR-induced phosphorylation of SLP-76, phospholipase C-gamma1 and the kinase Erk, more-persistent calcium flux, and increased production of cytokines and antigen-specific antibodies. Furthermore, HPK1-deficient mice were more susceptible to experimental autoimmune encephalomyelitis. Although the interaction between SLP-76 and Gads was unaffected, the inducible association of SLP-76 with 14-3-3tau (a phosphorylated serine-binding protein and negative regulator of TCR signaling) was reduced in HPK1-deficient T cells after TCR stimulation. HPK1 phosphorylated SLP-76 and induced the interaction of SLP-76 with 14-3-3tau. Our results indicate that HPK1 negatively regulates TCR signaling and T cell-mediated immune responses.
Collapse
Affiliation(s)
- Jr-Wen Shui
- Department of Immunology, Baylor College of Medicine, Houston, Texas 77030, USA
| | | | | | | | | | | | | |
Collapse
|
94
|
Xie M, Zhang D, Dyck JRB, Li Y, Zhang H, Morishima M, Mann DL, Taffet GE, Baldini A, Khoury DS, Schneider MD. A pivotal role for endogenous TGF-beta-activated kinase-1 in the LKB1/AMP-activated protein kinase energy-sensor pathway. Proc Natl Acad Sci U S A 2006; 103:17378-83. [PMID: 17085580 PMCID: PMC1859937 DOI: 10.1073/pnas.0604708103] [Citation(s) in RCA: 282] [Impact Index Per Article: 14.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/20/2023] Open
Abstract
TGF-beta-activated kinase-1 (TAK1), also known as MAPKK kinase-7 (MAP3K7), is a candidate effector of multiple circuits in cardiac biology and disease. Here, we show that inhibition of TAK1 in mice by a cardiac-specific dominant-negative mutation evokes electrophysiological and biochemical properties reminiscent of human Wolff-Parkinson-White syndrome, arising from mutations in AMP-activated protein kinase (AMPK), most notably, accelerated atrioventricular conduction and impaired AMPK activation. To test conclusively the biochemical connection from TAK1 to AMPK suggested by this phenotype, we disrupted TAK1 in mouse embryos and embryonic fibroblasts by Cre-mediated recombination. In TAK1-null embryos, the activating phosphorylation of AMPK at T172 was blocked, accompanied by defective AMPK activity. However, loss of endogenous TAK1 causes midgestation lethality, with defective yolk sac and intraembryonic vasculature. To preclude confounding lethal defects, we acutely ablated floxed TAK1 in culture by viral delivery of Cre. In culture, endogenous TAK1 was activated by oligomycin, the antidiabetic drug metformin, 5-aminoimidazole-4-carboxamide riboside (AICAR), and ischemia, well established triggers of AMPK activity. Loss of TAK1 in culture blocked T172 phosphorylation induced by all three agents, interfered with AMPK activation, impaired phosphorylation of the endogenous AMPK substrate acetyl CoA carboxylase, and also interfered with activation of the AMPK kinase LKB1. Thus, by disrupting the endogenous TAK1 locus, we prove a pivotal role for TAK1 in the LKB1/AMPK signaling axis, an essential governor of cell metabolism.
Collapse
Affiliation(s)
- Min Xie
- *Center for Cardiovascular Development
- The DeBakey Heart Center Graduate Program in Cardiovascular Sciences, and
- Departments of Medicine
| | - Dou Zhang
- *Center for Cardiovascular Development
- Departments of Medicine
| | - Jason R. B. Dyck
- Cardiovascular Research Group, Departments of Pediatrics and Pharmacology, Faculty of Medicine, University of Alberta, Edmonton, AB, Canada T6G 2S2
| | | | - Hui Zhang
- *Center for Cardiovascular Development
- The DeBakey Heart Center Graduate Program in Cardiovascular Sciences, and
- Departments of Medicine
| | - Masae Morishima
- *Center for Cardiovascular Development
- Pediatrics, Baylor College of Medicine, Houston, TX 77030; and
| | - Douglas L. Mann
- The DeBakey Heart Center Graduate Program in Cardiovascular Sciences, and
- The Winters Center for Heart Failure Research
- Departments of Medicine
| | - George E. Taffet
- The DeBakey Heart Center Graduate Program in Cardiovascular Sciences, and
- Departments of Medicine
| | - Antonio Baldini
- *Center for Cardiovascular Development
- Pediatrics, Baylor College of Medicine, Houston, TX 77030; and
| | - Dirar S. Khoury
- The DeBakey Heart Center Graduate Program in Cardiovascular Sciences, and
- Departments of Medicine
| | - Michael D. Schneider
- *Center for Cardiovascular Development
- The DeBakey Heart Center Graduate Program in Cardiovascular Sciences, and
- Departments of Medicine
- **Molecular and Cellular Biology
- Molecular Physiology and Biophysics, and
- To whom correspondence should be addressed. E-mail:
| |
Collapse
|
95
|
Harpster MH, Bandyopadhyay S, Thomas DP, Ivanov PS, Keele JA, Pineguina N, Gao B, Amarendran V, Gomelsky M, McCormick RJ, Stayton MM. Earliest changes in the left ventricular transcriptome postmyocardial infarction. Mamm Genome 2006; 17:701-15. [PMID: 16845475 DOI: 10.1007/s00335-005-0120-1] [Citation(s) in RCA: 54] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2005] [Accepted: 02/15/2006] [Indexed: 01/06/2023]
Abstract
We report a genome-wide survey of early responses of the mouse heart transcriptome to acute myocardial infarction (AMI). For three regions of the left ventricle (LV), namely, ischemic/infarcted tissue (IF), the surviving LV free wall (FW), and the interventricular septum (IVS), 36,899 transcripts were assayed at six time points from 15 min to 48 h post-AMI in both AMI and sham surgery mice. For each transcript, temporal expression patterns were systematically compared between AMI and sham groups, which identified 515 AMI-responsive genes in IF tissue, 35 in the FW, 7 in the IVS, with three genes induced in all three regions. Using the literature, we assigned functional annotations to all 519 nonredundant AMI-induced genes and present two testable models for central signaling pathways induced early post-AMI. First, the early induction of 15 genes involved in assembly and activation of the activator protein-1 (AP-1) family of transcription factors implicates AP-1 as a dominant regulator of earliest post-ischemic molecular events. Second, dramatic increases in transcripts for arginase 1 (ARG1), the enzymes of polyamine biosynthesis, and protein inhibitor of nitric oxide synthase (NOS) activity indicate that NO production may be regulated, in part, by inhibition of NOS and coordinate depletion of the NOS substrate, L: -arginine. ARG1: was the single-most highly induced transcript in the database (121-fold in IF region) and its induction in heart has not been previously reported.
Collapse
Affiliation(s)
- Mark H Harpster
- Department of Molecular Biology, University of Wyoming, Laramie, 82071, USA
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
96
|
Abstract
Sequential activation of protein kinases within the mitogen-activated protein kinase (MAPK) cascades is a common mechanism of signal transduction in many cellular processes. Four such cascades have been elucidated thus far, and named according to their MAPK tier component as the ERK1/2, JNK, p38MAPK, and ERK5 cascades. These cascades cooperate in transmitting various extracellular signals, and thus control cellular processes such as proliferation, differentiation, development, stress response, and apoptosis. Here we describe the classic ERK1/2 cascade, and concentrate mainly on the properties of MEK1/2 and ERK1/2, including their mode of regulation and their role in various cellular processes and in oncogenesis. This cascade may serve as a prototype of the other MAPK cascades, and the study of this cascade is likely to contribute to the understanding of mitogenic and other processes in many cell lines and tissues.
Collapse
Affiliation(s)
- Hadara Rubinfeld
- Department of Biological Regulation, The Weizmann Institute of Science, Rehovot 76100, Israel
| | | |
Collapse
|
97
|
Collins CS, Hong J, Sapinoso L, Zhou Y, Liu Z, Micklash K, Schultz PG, Hampton GM. A small interfering RNA screen for modulators of tumor cell motility identifies MAP4K4 as a promigratory kinase. Proc Natl Acad Sci U S A 2006; 103:3775-80. [PMID: 16537454 PMCID: PMC1383649 DOI: 10.1073/pnas.0600040103] [Citation(s) in RCA: 156] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023] Open
Abstract
Cell motility is a complex biological process, involved in development, inflammation, homeostasis, and pathological processes such as the invasion and metastatic spread of cancer. Here, we describe a genomic screen designed to identify inhibitors of cell migration. A library of 10,996 small interfering RNAs (targeting 5,234 human genes) was screened for their ability to block the migration of a highly motile ovarian carcinoma cell line, SKOV-3, by using a 384-well wound-healing assay coupled with automated microscopy and wound quantification. Two or more small interfering RNAs against four genes, CDK7, DYRK1B, MAP4K4 (NIK/HGK) (MAP4K4, mitogen-activated protein 4 kinase 4), and SCCA-1 (SerpinB3), potently blocked the migration of SKOV-3 cells, concordant with reduced transcript levels. Further studies of the promigratory role of MAP4K4 showed that the knockdown of this transcript inhibited the migration of multiple carcinoma cell lines, indicating a broad role in cell motility and potently suppressed the invasion of SKOV-3 cells in vitro. The effect of MAP4K4 on cellular migration was found to be mediated through c-Jun N-terminal kinase, independent of AP1 activation and downstream transcription. Accordingly, small molecule inhibition of c-Jun N-terminal kinase suppressed SKOV-3 cell migration, underscoring the potential therapeutic utility of mitogen-activated protein kinase pathway inhibition in cancer progression.
Collapse
Affiliation(s)
- Cynthia S. Collins
- *Genomics Institute of the Novartis Research Foundation, 10675 John Jay Hopkins Drive, San Diego, CA 92121; and
| | - Jiyong Hong
- Department of Chemistry and The Skaggs Institute for Chemical Biology, The Scripps Research Institute, 10550 North Torrey Pines Road, SR202, La Jolla, CA 92037
| | - Lisa Sapinoso
- *Genomics Institute of the Novartis Research Foundation, 10675 John Jay Hopkins Drive, San Diego, CA 92121; and
| | - Yingyao Zhou
- *Genomics Institute of the Novartis Research Foundation, 10675 John Jay Hopkins Drive, San Diego, CA 92121; and
| | - Zheng Liu
- *Genomics Institute of the Novartis Research Foundation, 10675 John Jay Hopkins Drive, San Diego, CA 92121; and
| | - Kenneth Micklash
- *Genomics Institute of the Novartis Research Foundation, 10675 John Jay Hopkins Drive, San Diego, CA 92121; and
| | - Peter G. Schultz
- *Genomics Institute of the Novartis Research Foundation, 10675 John Jay Hopkins Drive, San Diego, CA 92121; and
- Department of Chemistry and The Skaggs Institute for Chemical Biology, The Scripps Research Institute, 10550 North Torrey Pines Road, SR202, La Jolla, CA 92037
| | - Garret M. Hampton
- *Genomics Institute of the Novartis Research Foundation, 10675 John Jay Hopkins Drive, San Diego, CA 92121; and
- To whom correspondence should be sent at the present address:
Celgene Signal Research, 4550 Towne Centre Drive, San Diego, CA 92121. E-mail:
| |
Collapse
|
98
|
Tang X, Guilherme A, Chakladar A, Powelka AM, Konda S, Virbasius JV, Nicoloro SMC, Straubhaar J, Czech MP. An RNA interference-based screen identifies MAP4K4/NIK as a negative regulator of PPARgamma, adipogenesis, and insulin-responsive hexose transport. Proc Natl Acad Sci U S A 2006; 103:2087-92. [PMID: 16461467 PMCID: PMC1413698 DOI: 10.1073/pnas.0507660103] [Citation(s) in RCA: 129] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
The insulin-regulated glucose transporter GLUT4 is a key modulator of whole body glucose homeostasis, and its selective loss in adipose tissue or skeletal muscle causes insulin resistance and diabetes. Here we report an RNA interference-based screen of protein kinases expressed in adipocytes and identify four negative regulators of insulin-responsive glucose transport: the protein kinases PCTAIRE-1 (PCTK1), PFTAIRE-1 (PFTK1), IkappaB kinase alpha, and MAP4K4/NIK. Integrin-linked protein kinase was identified as a positive regulator of this process. We characterized one of these hits, MAP4K4/NIK, and found that it is unique among mitogen-activated protein (MAP) kinases expressed in cultured adipocytes in attenuating hexose transport. Remarkably, MAP4K4/NIK suppresses expression of the adipogenic transcription factors C/EBPalpha, C/EBPbeta, and PPARgamma and of GLUT4 itself in these cells. RNA interference-mediated depletion of MAP4K4/NIK early in differentiation enhances adipogenesis and triglyceride deposition, and even in fully differentiated adipocytes its loss up-regulates GLUT4. Conversely, conditions that inhibit adipogenesis such as TNF-alpha treatment or depletion of PPARgamma markedly up-regulate MAP4K4/NIK expression in cultured adipocytes. Furthermore, TNF-alpha signaling to down-regulate GLUT4 is impaired in the absence of MAP4K4/NIK, indicating that MAP4K4 expression is required for optimal TNF-alpha action. These results reveal a MAP4K4/NIK-dependent signaling pathway that potently inhibits PPARgamma-responsive gene expression, adipogenesis, and insulin-stimulated glucose transport.
Collapse
Affiliation(s)
- Xiaoqing Tang
- Program in Molecular Medicine, University of Massachusetts Medical School, Worcester, MA 01605
| | - Adilson Guilherme
- Program in Molecular Medicine, University of Massachusetts Medical School, Worcester, MA 01605
| | - Abhijit Chakladar
- Program in Molecular Medicine, University of Massachusetts Medical School, Worcester, MA 01605
| | - Aimee M. Powelka
- Program in Molecular Medicine, University of Massachusetts Medical School, Worcester, MA 01605
| | - Silvana Konda
- Program in Molecular Medicine, University of Massachusetts Medical School, Worcester, MA 01605
| | - Joseph V. Virbasius
- Program in Molecular Medicine, University of Massachusetts Medical School, Worcester, MA 01605
| | - Sarah M. C. Nicoloro
- Program in Molecular Medicine, University of Massachusetts Medical School, Worcester, MA 01605
| | - Juerg Straubhaar
- Program in Molecular Medicine, University of Massachusetts Medical School, Worcester, MA 01605
| | - Michael P. Czech
- Program in Molecular Medicine, University of Massachusetts Medical School, Worcester, MA 01605
- To whom correspondence should be addressed. E-mail:
| |
Collapse
|
99
|
Kendall SE, Battelli C, Irwin S, Mitchell JG, Glackin CA, Verdi JM. NRAGE mediates p38 activation and neural progenitor apoptosis via the bone morphogenetic protein signaling cascade. Mol Cell Biol 2005; 25:7711-24. [PMID: 16107717 PMCID: PMC1190310 DOI: 10.1128/mcb.25.17.7711-7724.2005] [Citation(s) in RCA: 62] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022] Open
Abstract
Understanding the molecular events that govern neural progenitor lineage commitment, mitotic arrest, and differentiation into functional progeny are germane to our understanding of neocortical development. Members of the family of bone morphogenetic proteins (BMPs) play pivotal roles in regulating neural differentiation and apoptosis during neurogenesis through combined actions involving Smad and TAK1 activation. We demonstrate that BMP signaling is required for the induction of apoptosis of neural progenitors and that NRAGE is a mandatory component of the signaling cascade. NRAGE possesses the ability to bind and function with the TAK1-TAB1-XIAP complex facilitating the activation of p38. Disruption of NRAGE or any other member of the noncanonical signaling cascaded is sufficient to block p38 activation and thus the proapoptotic signals generated through BMP exposure. The function of NRAGE is independent of Smad signaling, but the introduction of a dominant-negative Smad5 also rescues neural progenitor apoptosis, suggesting that both canonical and noncanonical pathways can converge and regulate BMP-mediated apoptosis. Collectively, these results establish NRAGE as an integral component in BMP signaling and clarify its role during neural progenitor development.
Collapse
|
100
|
Miled C, Pontoglio M, Garbay S, Yaniv M, Weitzman JB. A Genomic Map of p53 Binding Sites Identifies Novel p53 Targets Involved in an Apoptotic Network. Cancer Res 2005; 65:5096-104. [PMID: 15958553 DOI: 10.1158/0008-5472.can-04-4232] [Citation(s) in RCA: 65] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
The transcriptional activity of the p53 protein is central to its role in tumor suppression. Identification of the complete repertoire of p53-regulated genes is critical for dissecting the complexity of the p53 network. Although several different approaches have been used to characterize the p53 genetic program, we still lack a comprehensive molecular understanding of how p53 prevents cancer. Using a computational approach, we generated a genome-wide map of p53 binding sites (p53BS) to identify novel p53 target genes. We show that the presence of nearby p53BS can identify new proapoptotic members of the Bcl2 family. We show that p53 binds to p53BS identified in the BCL-G/BCL2L14 gene and that induction of this gene contributes to p53-mediated apoptosis. We found that p53 activates the COL18A1 gene encoding the precursor for the antiangiogenic factor endostatin. We also show that p53 up-regulates the MAP4K4 gene and activates the c-Jun NH2-terminal kinase (JNK) pathway to drive apoptosis. Thus, unbiased mapping of the genomic landscape of p53BS provides a systematic and complementary approach to identify novel factors and connections in the p53 genetic network. Our study illustrates how systematic genomic approaches can identify binding sites that are functionally relevant for a p53 transcriptional program. The genetic link among p53, antiangiogenic factors, and the JNK signaling pathway adds new dimensions to understanding p53 function in highly connected genetic networks.
Collapse
Affiliation(s)
- Chaouki Miled
- Unit of Gene Expression and Disease CNRS FRE2850, Department of Developmental Biology, Pasteur Institute, Paris, France
| | | | | | | | | |
Collapse
|