Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 14 de 14
Filter
Add more filters










Publication year range
1.
Front Oncol ; 11: 766298, 2021.
Article in English | MEDLINE | ID: mdl-34900714

ABSTRACT

Oncoprotein expression is controlled at the level of mRNA translation and is regulated by the eukaryotic translation initiation factor 4F (eIF4F) complex. eIF4A, a component of eIF4F, catalyzes the unwinding of secondary structure in the 5'-untranslated region (5'-UTR) of mRNA to facilitate ribosome scanning and translation initiation. Zotatifin (eFT226) is a selective eIF4A inhibitor that increases the affinity between eIF4A and specific polypurine sequence motifs and has been reported to inhibit translation of driver oncogenes in models of lymphoma. Here we report the identification of zotatifin binding motifs in the 5'-UTRs of HER2 and FGFR1/2 Receptor Tyrosine Kinases (RTKs). Dysregulation of HER2 or FGFR1/2 in human cancers leads to activation of the PI3K/AKT and RAS/ERK signaling pathways, thus enhancing eIF4A activity and promoting the translation of select oncogenes that are required for tumor cell growth and survival. In solid tumor models driven by alterations in HER2 or FGFR1/2, downregulation of oncoprotein expression by zotatifin induces sustained pathway-dependent anti-tumor activity resulting in potent inhibition of cell proliferation, induction of apoptosis, and significant in vivo tumor growth inhibition or regression. Sensitivity of RTK-driven tumor models to zotatifin correlated with high basal levels of mTOR activity and elevated translational capacity highlighting the unique circuitry generated by the RTK-driven signaling pathway. This dependency identifies the potential for rational combination strategies aimed at vertical inhibition of the PI3K/AKT/eIF4F pathway. Combination of zotatifin with PI3K or AKT inhibitors was beneficial across RTK-driven cancer models by blocking RTK-driven resistance mechanisms demonstrating the clinical potential of these combination strategies.

2.
Bioorg Med Chem Lett ; 47: 128111, 2021 09 01.
Article in English | MEDLINE | ID: mdl-34353608

ABSTRACT

Flavaglines such as silvestrol (1) and rocaglamide (2) constitute an interesting class of natural products with promising anticancer activities. Their mode of action is based on inhibition of eukaryotic initiation factor 4A (eIF4A) dependent translation through formation of a stable ternary complex with eIF4A and mRNA, thus blocking ribosome scanning. Herein we describe initial SAR studies in a novel series of 1-aminomethyl substituted flavagline-inspired eIF4A inhibitors. We discovered that a variety of N-substitutions at the 1-aminomethyl group are tolerated, making this position pertinent for property and ADME profile tuning. The findings presented herein are relevant to future drug design efforts towards novel eIF4A inhibitors with drug-like properties.


Subject(s)
Antineoplastic Agents/pharmacology , Benzofurans/pharmacology , Biological Products/pharmacology , Eukaryotic Initiation Factor-4A/antagonists & inhibitors , Triterpenes/pharmacology , Antineoplastic Agents/chemical synthesis , Antineoplastic Agents/chemistry , Benzofurans/chemical synthesis , Benzofurans/chemistry , Biological Products/chemical synthesis , Biological Products/chemistry , Cell Line, Tumor , Cell Proliferation/drug effects , Dose-Response Relationship, Drug , Drug Design , Drug Screening Assays, Antitumor , Eukaryotic Initiation Factor-4A/metabolism , Humans , Molecular Structure , Structure-Activity Relationship , Triterpenes/chemical synthesis , Triterpenes/chemistry
3.
Mol Cancer Ther ; 20(1): 26-36, 2021 01.
Article in English | MEDLINE | ID: mdl-33037136

ABSTRACT

The PI3K/AKT/mTOR pathway is often activated in lymphoma through alterations in PI3K, PTEN, and B-cell receptor signaling, leading to dysregulation of eIF4A (through its regulators, eIF4B, eIF4G, and PDCD4) and the eIF4F complex. Activation of eIF4F has a direct role in tumorigenesis due to increased synthesis of oncogenes that are dependent on enhanced eIF4A RNA helicase activity for translation. eFT226, which inhibits translation of specific mRNAs by promoting eIF4A1 binding to 5'-untranslated regions (UTR) containing polypurine and/or G-quadruplex recognition motifs, shows potent antiproliferative activity and significant in vivo efficacy against a panel of diffuse large B-cell lymphoma (DLBCL), and Burkitt lymphoma models with ≤1 mg/kg/week intravenous administration. Evaluation of predictive markers of sensitivity or resistance has shown that activation of eIF4A, mediated by mTOR signaling, correlated with eFT226 sensitivity in in vivo xenograft models. Mutation of PTEN is associated with reduced apoptosis in vitro and diminished efficacy in vivo in response to eFT226. In models evaluated with PTEN loss, AKT was stimulated without a corresponding increase in mTOR activation. AKT activation leads to the degradation of PDCD4, which can alter eIF4F complex formation. The association of eFT226 activity with PTEN/PI3K/mTOR pathway regulation of mRNA translation provides a means to identify patient subsets during clinical development.


Subject(s)
Eukaryotic Initiation Factor-4A/antagonists & inhibitors , Lymphoma, B-Cell/genetics , Lymphoma, B-Cell/pathology , Oncogenes , Protein Biosynthesis/genetics , RNA, Messenger/genetics , Animals , Biomarkers, Tumor/metabolism , Cell Line, Tumor , Drug Resistance, Neoplasm/drug effects , Eukaryotic Initiation Factor-4A/metabolism , Female , Humans , Mice, Inbred NOD , Mice, SCID , PTEN Phosphohydrolase/metabolism , RNA, Messenger/metabolism , TOR Serine-Threonine Kinases/metabolism , Xenograft Model Antitumor Assays
4.
Org Lett ; 22(16): 6257-6261, 2020 08 21.
Article in English | MEDLINE | ID: mdl-32806219

ABSTRACT

Rocaglates, rocaglamides, and related flavagline natural products exert their remarkable anticancer activity through inhibition of eukaryotic initiation factor 4A (eIF4A) but generally display suboptimal drug-like properties. In our efforts to identify potent drug-like eIF4A inhibitors, we developed synthetic strategies for diastereoselectively functionalizing the C1 position of aza-rocaglamide scaffolds (cf. 14 and 18), which proceed via retention or inversion of configuration at C1 depending on the C2 substituent (cf. 15 and 21) and ultimately enabled the discovery of novel and potent eIF4A inhibitors such as 25.


Subject(s)
Benzofurans/chemistry , Eukaryotic Initiation Factor-4A/antagonists & inhibitors , Binding Sites , Biological Products , Eukaryotic Initiation Factor-4A/metabolism , Humans , Molecular Structure
5.
J Med Chem ; 63(11): 5879-5955, 2020 06 11.
Article in English | MEDLINE | ID: mdl-32470302

ABSTRACT

Dysregulation of protein translation is a key driver for the pathogenesis of many cancers. Eukaryotic initiation factor 4A (eIF4A), an ATP-dependent DEAD-box RNA helicase, is a critical component of the eIF4F complex, which regulates cap-dependent protein synthesis. The flavagline class of natural products (i.e., rocaglamide A) has been shown to inhibit protein synthesis by stabilizing a translation-incompetent complex for select messenger RNAs (mRNAs) with eIF4A. Despite showing promising anticancer phenotypes, the development of flavagline derivatives as therapeutic agents has been hampered because of poor drug-like properties as well as synthetic complexity. A focused effort was undertaken utilizing a ligand-based design strategy to identify a chemotype with optimized physicochemical properties. Also, detailed mechanistic studies were undertaken to further elucidate mRNA sequence selectivity, key regulated target genes, and the associated antitumor phenotype. This work led to the design of eFT226 (Zotatifin), a compound with excellent physicochemical properties and significant antitumor activity that supports clinical development.


Subject(s)
Benzofurans/chemistry , Drug Design , Eukaryotic Initiation Factor-4A/antagonists & inhibitors , Animals , Benzofurans/pharmacokinetics , Benzofurans/therapeutic use , Binding Sites , Breast Neoplasms/drug therapy , Breast Neoplasms/pathology , Cell Line, Tumor , Crystallography, X-Ray , Eukaryotic Initiation Factor-4A/genetics , Eukaryotic Initiation Factor-4A/metabolism , Female , Half-Life , Humans , Ligands , Mice , Mice, Nude , Molecular Dynamics Simulation , Mutagenesis, Site-Directed , Protein Structure, Tertiary , RNA, Messenger/chemistry , RNA, Messenger/metabolism , Rats , Structure-Activity Relationship
6.
J Med Chem ; 61(8): 3516-3540, 2018 04 26.
Article in English | MEDLINE | ID: mdl-29526098

ABSTRACT

Dysregulated translation of mRNA plays a major role in tumorigenesis. Mitogen-activated protein kinase interacting kinases (MNK)1/2 are key regulators of mRNA translation integrating signals from oncogenic and immune signaling pathways through phosphorylation of eIF4E and other mRNA binding proteins. Modulation of these key effector proteins regulates mRNA, which controls tumor/stromal cell signaling. Compound 23 (eFT508), an exquisitely selective, potent dual MNK1/2 inhibitor, was designed to assess the potential for control of oncogene signaling at the level of mRNA translation. The crystal structure-guided design leverages stereoelectronic interactions unique to MNK culminating in a novel pyridone-aminal structure described for the first time in the kinase literature. Compound 23 has potent in vivo antitumor activity in models of diffuse large cell B-cell lymphoma and solid tumors, suggesting that controlling dysregulated translation has real therapeutic potential. Compound 23 is currently being evaluated in Phase 2 clinical trials in solid tumors and lymphoma. Compound 23 is the first highly selective dual MNK inhibitor targeting dysregulated translation being assessed clinically.


Subject(s)
Antineoplastic Agents/therapeutic use , Intracellular Signaling Peptides and Proteins/antagonists & inhibitors , Protein Kinase Inhibitors/therapeutic use , Protein Serine-Threonine Kinases/antagonists & inhibitors , Pyridines/therapeutic use , Pyridones/therapeutic use , Pyrimidines/therapeutic use , Spiro Compounds/therapeutic use , Animals , Antineoplastic Agents/chemical synthesis , Antineoplastic Agents/chemistry , Antineoplastic Agents/pharmacology , Catalytic Domain , Cell Line, Tumor , Drug Design , Eukaryotic Initiation Factor-4E/chemistry , Eukaryotic Initiation Factor-4E/metabolism , Humans , Intracellular Signaling Peptides and Proteins/metabolism , Mice , Molecular Structure , Phosphorylation , Protein Kinase Inhibitors/chemical synthesis , Protein Kinase Inhibitors/chemistry , Protein Kinase Inhibitors/pharmacology , Protein Serine-Threonine Kinases/metabolism , Pyridines/chemical synthesis , Pyridines/chemistry , Pyridines/pharmacology , Pyridones/chemical synthesis , Pyridones/chemistry , Pyridones/pharmacology , Pyrimidines/chemical synthesis , Pyrimidines/chemistry , Pyrimidines/pharmacology , Rats , Serine/chemistry , Signal Transduction/drug effects , Spiro Compounds/chemical synthesis , Spiro Compounds/chemistry , Spiro Compounds/pharmacology , Xenograft Model Antitumor Assays
7.
J Immunol ; 191(11): 5655-68, 2013 Dec 01.
Article in English | MEDLINE | ID: mdl-24146043

ABSTRACT

The contraction phase of the T cell response is a poorly understood period after the resolution of infection when virus-specific effector cells decline in number and memory cells emerge with increased frequencies. CD8(+) T cells plummet in number and quickly reach stable levels of memory following acute lymphocytic choriomeningitis virus infection in mice. In contrast, virus-specific CD4(+) T cells gradually decrease in number and reach homeostatic levels only after many weeks. In this study, we provide evidence that MHCII-restricted viral Ag persists during the contraction phase following this prototypical acute virus infection. We evaluated whether the residual Ag affected the cell division and number of virus-specific naive and memory CD4(+) T cells and CD8(+) T cells. We found that naive CD4(+) T cells underwent cell division and accumulated in response to residual viral Ag for >2 mo after the eradication of infectious virus. Surprisingly, memory CD4(+) T cells did not undergo cell division in response to the lingering Ag, despite their heightened capacity to recognize Ag and make cytokine. In contrast to CD4(+) T cells, CD8(+) T cells did not undergo cell division in response to the residual Ag. Thus, CD8(+) T cells ceased division within days after the infection was resolved, indicating that CD8(+) T cell responses are tightly linked to endogenous processing of de novo synthesized virus protein. Our data suggest that residual viral Ag delays the contraction of CD4(+) T cell responses by recruiting new populations of CD4(+) T cells.


Subject(s)
CD4-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/immunology , Immunologic Memory , Lymphocytic Choriomeningitis/immunology , Lymphocytic choriomeningitis virus/immunology , Animals , CD4-Positive T-Lymphocytes/virology , CD8-Positive T-Lymphocytes/virology , Cell Proliferation , Cells, Cultured , Cross-Priming , Histocompatibility Antigens Class II/metabolism , Lymphocyte Activation , Mice , Mice, Inbred C57BL , Mice, Transgenic , Protein Binding
8.
Eur J Immunol ; 39(6): 1494-504, 2009 Jun.
Article in English | MEDLINE | ID: mdl-19384870

ABSTRACT

Stem cell antigen-1 (Sca1, Ly-6A/E) is a well-established marker of murine hematopoietic stem cells, and also is expressed on memory T cells. It has been suggested that the functional maintenance of T-cell memory requires the expression of Sca1 on a specialized population of memory T cells termed "memory stem cells". Here, we evaluate the requirement for Sca1 in the primary T-cell response to virus infection, and in the establishment and maintenance of T-cell memory. We find that Sca1 expression increases on almost all CD4(+) and CD8(+) T cells during virus infection, and remains high on virus-specific memory cells. However, Sca1-deficient (Sca1KO) mice generate normal primary T-cell responses to infection; the kinetics, the immunodominance hierarchy, and the absolute numbers of CD4(+) and CD8(+) T cells are essentially indistinguishable from those observed in WT mice. Furthermore, by several criteria, primary and memory T cells in Sca1-deficient mice are phenotypically and functionally normal. These data indicate that Sca1, although perhaps a useful marker of virus-specific memory T cells, is not required for the regulation of T-cell quantity or quality, or for the development of a competent pool of memory cells.


Subject(s)
Arenaviridae Infections/immunology , CD4-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/immunology , Immunologic Memory/immunology , Membrane Proteins/deficiency , Animals , Antigens, Ly/physiology , CD4-Positive T-Lymphocytes/cytology , CD4-Positive T-Lymphocytes/metabolism , CD8-Positive T-Lymphocytes/cytology , CD8-Positive T-Lymphocytes/metabolism , Cell Count , Hyaluronan Receptors/metabolism , Immunity, Cellular/immunology , Interferon-gamma/metabolism , Interleukin-2/metabolism , Kinetics , Lymphocytic choriomeningitis virus/immunology , Membrane Proteins/physiology , Mice , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , Receptors, Interferon/genetics , Spleen/cytology , Spleen/immunology , T-Lymphocyte Subsets/cytology , T-Lymphocyte Subsets/immunology , T-Lymphocyte Subsets/metabolism , T-Lymphocytes, Cytotoxic/immunology
9.
J Immunol ; 180(10): 6777-85, 2008 May 15.
Article in English | MEDLINE | ID: mdl-18453598

ABSTRACT

The precursor frequency of naive CD4(+) T cells shows an inverse relationship with the number of memory cells generated after exposure to cognate Ag. Using the lymphocytic choriomeningitis virus (LCMV) model, we show here that only when the initial number of naive virus-specific CD4(+) T cell precursors is low (< or =10(4) per spleen) do they give rise to abundant and homogeneous memory cells that are CD62L(low), IL-7R(high), and imbued with an enhanced capacity to produce cytokine, proliferate, and survive over time. Furthermore, memory cells derived from a high naive precursor number show functional deficits upon secondary exposure to virus. The negative effect of higher naive precursor frequency was not attributable to competition for limiting amounts of Ag, because LCMV-naive CD4(+) TCR-transgenic CD4 T cells were recruited into the LCMV-induced response even when their initial number was high. Instead, the T cells appear to compete for direct IFN-gamma signals as they differentiate into memory cells. These results are consistent with a model of T cell development in which the most fit effector T cells that receive sufficient direct IFN-gamma signals are selected to differentiate further into memory cells.


Subject(s)
CD4-Positive T-Lymphocytes/immunology , Immunologic Memory , Interferon-gamma/immunology , Precursor Cells, T-Lymphoid/immunology , Adoptive Transfer , Animals , CD4-Positive T-Lymphocytes/cytology , Cell Differentiation/immunology , Flow Cytometry , Interferon-gamma/metabolism , Lymphocyte Activation/immunology , Lymphocytic Choriomeningitis/immunology , Mice , Mice, Transgenic , Precursor Cells, T-Lymphoid/cytology
10.
PLoS Pathog ; 4(4): e1000041, 2008 Apr 11.
Article in English | MEDLINE | ID: mdl-18404208

ABSTRACT

T cell memory is a cornerstone of protective immunity, and is the key element in successful vaccination. Upon encountering the relevant pathogen, memory T cells are thought to initiate cell division much more rapidly than their naïve counterparts, and this is thought to confer a significant biological advantage upon an immune host. Here, we use traceable TCR-transgenic T cells to evaluate this proposed characteristic in CD4+ and CD8+ memory T cells. We find that, even in the presence of abundant antigen that was sufficient to induce in vivo IFNgamma production by memory T cells, both memory and naïve T cells show an extended, and indistinguishable, delay in the onset of proliferation. Although memory cells can detect, and respond to, virus infection within a few hours, their proliferation did not begin until approximately 3 days after infection, and occurred simultaneously in all anatomical compartments. Thereafter, cell division was extraordinarily rapid for both naïve and memory cells, with the latter showing a somewhat accelerated accumulation. We propose that, by permitting memory T cells to rapidly exert their effector functions while delaying the onset of their proliferation, evolution has provided a safeguard that balances the risk of infection against the consequences of severe T cell-mediated immunopathology.


Subject(s)
CD4-Positive T-Lymphocytes/cytology , CD8-Positive T-Lymphocytes/cytology , Immunologic Memory , Adoptive Transfer , Animals , Brefeldin A/pharmacology , CD4-Positive T-Lymphocytes/drug effects , CD4-Positive T-Lymphocytes/metabolism , CD8-Positive T-Lymphocytes/drug effects , CD8-Positive T-Lymphocytes/metabolism , Cell Division , Cell Proliferation , Disease Models, Animal , Immune Tolerance , Interferon-gamma/metabolism , Lymph Nodes/cytology , Lymph Nodes/drug effects , Lymph Nodes/immunology , Lymphocyte Activation , Lymphocytic Choriomeningitis/immunology , Lymphocytic Choriomeningitis/virology , Lymphocytic choriomeningitis virus/immunology , Mice , Mice, Inbred C57BL , Mice, Transgenic/immunology , Protein Synthesis Inhibitors/pharmacology , Receptors, Antigen, T-Cell/metabolism , Spleen/cytology , Spleen/drug effects , Spleen/immunology
11.
J Neuroimmunol ; 192(1-2): 124-33, 2007 Dec.
Article in English | MEDLINE | ID: mdl-17964666

ABSTRACT

The inoculation of MOG peptides into C57BL/6 mice induces CD4(+) and CD8(+) T cells, and recent work has shown that adoptive transfer of the latter population, after extensive in vitro stimulation, can cause EAE in naïve recipient mice. Herein, we have evaluated the incidence and severity of EAE, and the induction of CD4(+) and CD8(+) T cells, following MOG peptide inoculation of wt mice and of LMP-2KO mice that lack an intact immunoproteasome, a cytoplasmic organelle that is induced by chronic inflammation and that may be important for the presentation of MHC class I epitopes to CD8(+) T cells. We report that EAE, evaluated by both clinical and histological criteria, is similar in LMP-2KO mice and wildtype C57B/6 mice (wt) in response to immunization with MOG peptides MOG(35-55) and MOG(40-54), suggesting that the immunoproteasome does not play a key role in the development of demyelinating disease. Furthermore, and consistent with previous reports, peptide-specific CD8(+) T cells were barely detectable in the CNS of peptide-immunized mice, although peptide-specific CD4(+) T cells were abundant. Therefore, we used a new technique to look for autoreactive CD8(+) T cells in MOG peptide-immunized mice, and we report the identification of CD4(+) and CD8(+) T cells that, as late as 19 days after peptide injection, are actively producing IFNgamma in vivo, in response to in vivo antigen contact.


Subject(s)
Cysteine Endopeptidases/deficiency , Encephalomyelitis, Autoimmune, Experimental/chemically induced , Encephalomyelitis, Autoimmune, Experimental/immunology , Myelin-Associated Glycoprotein , Peptide Fragments , T-Lymphocytes/drug effects , Animals , CD4-Positive T-Lymphocytes/drug effects , CD4-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/drug effects , CD8-Positive T-Lymphocytes/immunology , Disease Models, Animal , Encephalomyelitis, Autoimmune, Experimental/pathology , Flow Cytometry , Lymphocyte Activation/drug effects , Mice , Mice, Inbred C57BL , Mice, Knockout , Myelin Proteins , Myelin-Oligodendrocyte Glycoprotein , Time Factors
12.
J Immunol ; 179(2): 1190-7, 2007 Jul 15.
Article in English | MEDLINE | ID: mdl-17617612

ABSTRACT

Studies in IFN-gamma-deficient mice suggest that the delivery of IFN-gamma to CD8(+) T cells early in virus infection programs their eventual contraction, thereby reducing the abundance of CD8(+) memory T cells. In this study, we show that such mice fail to completely eliminate virus infection and that, when evaluated without the confounding factor of persisting Ag, both CD4(+) and CD8(+) T cells undergo profound contraction when they are unable to receive IFN-gamma signals. Furthermore, the abundance of CD4(+) and CD8(+) memory cells that express the IFN-gamma receptor is approximately 100-fold higher than cells lacking this molecule. Thus, direct IFN-gamma signaling is not required for T cell contraction during virus infection, and it enhances, rather than suppresses, the development of virus-specific CD4(+) and CD8(+) T cell memory.


Subject(s)
Arenaviridae Infections/immunology , CD4-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/immunology , Immunologic Memory , Interferon-gamma/immunology , Signal Transduction/immunology , Adoptive Transfer , Animals , Flow Cytometry , Lymphocytic choriomeningitis virus/immunology , Mice , Mice, Transgenic , Receptors, Interferon/biosynthesis , Interferon gamma Receptor
13.
Appl Environ Microbiol ; 73(2): 622-9, 2007 Jan.
Article in English | MEDLINE | ID: mdl-17122394

ABSTRACT

Sponges are host to extremely diverse bacterial communities, some of which appear to be spatiotemporally stable, though how these consistent associations are assembled and maintained from one sponge generation to the next is not well understood. Here we report that a diverse group of microbes, including both bacteria and archaea, is consistently present in aggregates within embryos of the tropical sponge Corticium sp. The major taxonomic groups represented in bacterial 16S rRNA sequences amplified from the embryos are similar to those previously described in a variety of marine sponges. Three selected bacterial taxa, representing proteobacteria, actinobacteria, and a clade including recently described sponge-associated bacteria, were tested and found to be present in all adult samples tested over a 3-year period and in the embryos throughout development. Specific probes were used in fluorescence in situ hybridization to localize cells of the three types in the embryos and mesohyl. This study confirms the vertical transmission of multiple, phylogenetically diverse microorganisms in a marine sponge, and our findings lay the foundation for future work on exploring vertical transmission of specific, yet diverse, microbial assemblages in marine sponges.


Subject(s)
Archaea/physiology , Bacteria/growth & development , Embryo, Nonmammalian/microbiology , Porifera/embryology , Porifera/microbiology , Animals , Archaea/classification , Archaea/genetics , Archaea/growth & development , Bacteria/classification , Bacteria/genetics , DNA, Archaeal/analysis , DNA, Bacterial/analysis , In Situ Hybridization, Fluorescence , Molecular Sequence Data , Phylogeny , Polymerase Chain Reaction , Porifera/classification , RNA, Ribosomal, 16S/genetics , Sequence Analysis, DNA , Tropical Climate
14.
Traffic ; 6(8): 667-81, 2005 Aug.
Article in English | MEDLINE | ID: mdl-15998322

ABSTRACT

Here, we show that the Rab27a-binding protein JFC1/Slp1 (synaptotagmin-like protein) is regulated by Akt-mediated phosphorylation. Using the phosphatase and tensin homolog-null LNCaP cells and the phosphatidylinositol 3-kinase inhibitor LY294002, we show that the phosphorylation of endogenous JFC1 is dependent on the phosphatidylinositol 3-kinase/Akt pathway. JFC1 was phosphorylated in cells expressing a constitutively active Akt, confirming that it is an Akt substrate in vivo. Direct phosphorylation of JFC1 by Akt was confirmed in vitro. Using microcapillary high-performance liquid chromatography tandem mass spectrometry, we identified five Akt-phosphorylation sites in JFC1. By mutagenesis analysis and subsequent immunoprecipitation (IP), we established that Akt phosphorylates JFC1 at serine 241. JFC1 and Rab27a colocalize in the proximity of the plasma membrane in LNCaP cells. The interaction was confirmed by IP analysis and was abolished by the point mutation W83S in JFC1. Phosphorylation did not alter the ability of JFC1 to bind to Rab27a. Instead, phosphorylation by Akt dramatically decreased when JFC1 was bound to Rab27a. Finally, we show that as a consequence of in vivo phosphorylation, JFC1 dissociates from the membrane, promoting JFC1 redistribution to the cytosol. Our results suggest that Akt regulates JFC1/Slp1 function by phosphorylation and may have implications on Rab27a-containing vesicle secretion.


Subject(s)
Membrane Proteins/metabolism , Protein Serine-Threonine Kinases/physiology , Proto-Oncogene Proteins/physiology , rab GTP-Binding Proteins/metabolism , Cell Line, Tumor , Chromones/pharmacology , Enzyme Inhibitors/pharmacology , HeLa Cells , Humans , Immunoprecipitation , Interleukin-6/pharmacology , Marine Toxins , Membrane Proteins/chemistry , Membrane Proteins/genetics , Morpholines/pharmacology , Mutagenesis, Site-Directed , Oxazoles/pharmacology , Phosphoinositide-3 Kinase Inhibitors , Phosphoprotein Phosphatases/antagonists & inhibitors , Phosphorylation/drug effects , Protein Binding , Protein Serine-Threonine Kinases/pharmacology , Proto-Oncogene Proteins/pharmacology , Proto-Oncogene Proteins c-akt , Recombinant Fusion Proteins , Secretory Vesicles/physiology , Transfection , Vesicular Transport Proteins , rab27 GTP-Binding Proteins
SELECTION OF CITATIONS
SEARCH DETAIL
...