Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 11 de 11
Filter
1.
Stem Cell Reports ; 14(1): 9-20, 2020 01 14.
Article in English | MEDLINE | ID: mdl-31883920

ABSTRACT

Differentiation of human embryonic stem cells into pancreatic ß cells holds great promise for the treatment of diabetes. Recent advances have led to the production of glucose-responsive insulin-secreting cells in vitro, but resulting cells remain less mature than their adult primary ß cell counterparts. The barrier(s) to in vitro ß cell maturation are unclear. Here, we evaluated a potential role for microRNAs. MicroRNA profiling showed high expression of let-7 family microRNAs in vivo, but not in in vitro differentiated ß cells. Reduced levels of let-7 in vitro were associated with increased levels of the RNA binding protein LIN28B, a negative regulator of let-7 biogenesis. Ablation of LIN28B during human embryonic stem cell (hESC) differentiation toward ß cells led to a more mature glucose-stimulated insulin secretion profile and the suppression of juvenile-specific genes. However, let-7 overexpression had little effect. These results uncover LIN28B as a modulator of ß cell maturation in vitro.


Subject(s)
Cell Differentiation/genetics , Gene Expression Regulation, Developmental , Human Embryonic Stem Cells/cytology , Human Embryonic Stem Cells/metabolism , Insulin-Secreting Cells/cytology , Insulin-Secreting Cells/metabolism , RNA-Binding Proteins/genetics , Animals , Humans , Mice , MicroRNAs/genetics , RNA Interference , RNA-Binding Proteins/metabolism
2.
EMBO J ; 34(13): 1759-72, 2015 Jul 02.
Article in English | MEDLINE | ID: mdl-25908839

ABSTRACT

Directed differentiation of human pluripotent stem cells into functional insulin-producing beta-like cells holds great promise for cell replacement therapy for patients suffering from diabetes. This approach also offers the unique opportunity to study otherwise inaccessible aspects of human beta cell development and function in vitro. Here, we show that current pancreatic progenitor differentiation protocols promote precocious endocrine commitment, ultimately resulting in the generation of non-functional polyhormonal cells. Omission of commonly used BMP inhibitors during pancreatic specification prevents precocious endocrine formation while treatment with retinoic acid followed by combined EGF/KGF efficiently generates both PDX1(+) and subsequent PDX1(+)/NKX6.1(+) pancreatic progenitor populations, respectively. Precise temporal activation of endocrine differentiation in PDX1(+)/NKX6.1(+) progenitors produces glucose-responsive beta-like cells in vitro that exhibit key features of bona fide human beta cells, remain functional after short-term transplantation, and reduce blood glucose levels in diabetic mice. Thus, our simplified and scalable system accurately recapitulates key steps of human pancreas development and provides a fast and reproducible supply of functional human beta-like cells.


Subject(s)
Cell Culture Techniques/methods , Cell Differentiation , Embryonic Stem Cells/physiology , Insulin-Secreting Cells/physiology , Pancreas/cytology , Animals , Blood Glucose/metabolism , Cells, Cultured , Diabetes Mellitus, Experimental/blood , Diabetes Mellitus, Experimental/therapy , Embryonic Stem Cells/cytology , Glucose/pharmacology , Humans , Insulin-Secreting Cells/cytology , Insulin-Secreting Cells/drug effects , Insulin-Secreting Cells/transplantation , Mice , Mice, SCID , Mice, Transgenic , Streptozocin
3.
J Virol ; 88(22): 13135-48, 2014 Nov.
Article in English | MEDLINE | ID: mdl-25187541

ABSTRACT

UNLABELLED: Protein synthesis, the most energy-consuming process in cells, responds to changing physiologic priorities, e.g., upon mitogen- or stress-induced adaptations signaled through the mitogen-activated protein kinases (MAPKs). The prevailing status of protein synthesis machinery is a viral pathogenesis factor, particularly for plus-strand RNA viruses, where immediate translation of incoming viral RNAs shapes host-virus interactions. In this study, we unraveled signaling pathways centered on the ERK1/2 and p38α MAPK-interacting kinases MNK1/2 and their role in controlling 7-methyl-guanosine (m(7)G) "cap"-independent translation at enterovirus type 1 internal ribosomal entry sites (IRESs). Activation of Raf-MEK-ERK1/2 signals induced viral IRES-mediated translation in a manner dependent on MNK1/2. This effect was not due to MNK's known functions as eukaryotic initiation factor (eIF) 4G binding partner or eIF4E(S209) kinase. Rather, MNK catalytic activity enabled viral IRES-mediated translation/host cell cytotoxicity through negative regulation of the Ser/Arg (SR)-rich protein kinase (SRPK). Our investigations suggest that SRPK activity is a major determinant of type 1 IRES competency, host cell cytotoxicity, and viral proliferation in infected cells. IMPORTANCE: We are targeting unfettered enterovirus IRES activity in cancer with PVSRIPO, the type 1 live-attenuated poliovirus (PV) (Sabin) vaccine containing a human rhinovirus type 2 (HRV2) IRES. A phase I clinical trial of PVSRIPO with intratumoral inoculation in patients with recurrent glioblastoma (GBM) is showing early promise. Viral translation proficiency in infected GBM cells is a core requirement for the antineoplastic efficacy of PVSRIPO. Therefore, it is critically important to understand the mechanisms controlling viral cap-independent translation in infected host cells.


Subject(s)
Enterovirus/genetics , Intracellular Signaling Peptides and Proteins/metabolism , MAP Kinase Signaling System , Protein Biosynthesis , Protein Serine-Threonine Kinases/metabolism , Viral Proteins/biosynthesis , Cell Line , Gene Expression Regulation, Viral , Host-Pathogen Interactions , Humans
4.
Mol Cell Biol ; 34(3): 439-51, 2014 Feb.
Article in English | MEDLINE | ID: mdl-24248602

ABSTRACT

During mitosis, global translation is suppressed, while synthesis of proteins with vital mitotic roles must go on. Prior evidence suggests that the mitotic translation shift involves control of initiation. Yet, no signals specifically targeting translation initiation factors during mitosis have been identified. We used phosphoproteomics to investigate the central translation initiation scaffold and "ribosome adaptor," eukaryotic initiation factor 4G1 (eIF4G1) in interphase or nocodazole-arrested mitotic cells. This approach and kinase inhibition assays, in vitro phosphorylation with recombinant kinase, and kinase depletion-reconstitution experiments revealed that Ser1232 in eIF4G1 is phosphorylated by cyclin-dependent kinase 1 (Cdk1):cyclin B during mitosis. Ser1232 is located in an unstructured region of the C-terminal portion of eIF4G1 that coordinates assembly of the eIF4G/-4A/-4B helicase complex and binding of the mitogen-activated protein kinase (MAPK) signal-integrating kinase, Mnk. Intense phosphorylation of Ser1232 in mitosis strongly enhanced the interactions of eIF4A with HEAT domain 2 of eIF4G and decreased association of eIF4G/-4A with RNA. Our findings implicate phosphorylation of eIF4G1(Ser1232) by Cdk1:cyclin B and its inhibitory effects on eIF4A helicase activity in the mitotic translation initiation shift.


Subject(s)
CDC2 Protein Kinase/metabolism , Cyclin B1/metabolism , Eukaryotic Initiation Factor-4A/metabolism , Eukaryotic Initiation Factor-4G/metabolism , RNA/metabolism , Adenine/analogs & derivatives , Adenine/pharmacology , CDC2 Protein Kinase/antagonists & inhibitors , CDC2 Protein Kinase/genetics , Cell Cycle/drug effects , Cell Cycle/genetics , Cyclin B1/genetics , Eukaryotic Initiation Factor-4A/genetics , Eukaryotic Initiation Factor-4G/genetics , Flow Cytometry , HEK293 Cells , HeLa Cells , Humans , Immunoblotting , Interphase/drug effects , Interphase/genetics , Intracellular Signaling Peptides and Proteins/metabolism , Mitosis/drug effects , Mitosis/genetics , Mutation , Nocodazole/pharmacology , Phosphorylation/drug effects , Protein Binding/drug effects , Protein Biosynthesis/drug effects , Protein Serine-Threonine Kinases/metabolism , Proteomics/methods , RNA/genetics , Serine/genetics , Serine/metabolism , Thymidine/pharmacology , Tubulin Modulators/pharmacology
5.
Mol Cell Biol ; 33(1): 127-35, 2013 Jan.
Article in English | MEDLINE | ID: mdl-23109423

ABSTRACT

The p38α to p38δ mitogen-activated protein kinases (MAPKs) are central regulatory nodes coordinating acute stress and inflammatory responses. Their activation leads to rapid adjustment of protein synthesis, for instance translational induction of proinflammatory cytokines. The only known direct link of p38 to translation machinery is the MAPK signal-integrating kinase Mnk. Only p38α and p38ß transcripts are ubiquitously expressed. These mRNAs encode highly conserved proteins that equally phosphorylate recombinant Mnk1 in vitro. We discovered that expression of the p38α protein, but not the p38ß isoform, is suppressed in the brain. This is due to p38α depletion by two neuron-selective microRNAs (miRNAs), miR-124 and -128. Suppression of p38α protein was reversed by miR-124/-128 antisense oligonucleotides in primary explant neuronal cultures. Targeted p38α depletion reduced Mnk1 activation, which cannot be compensated by p38ß. Our research shows that p38α alone controls acute stress and cytokine signaling from p38 MAPK to translation machinery. This regulatory axis is greatly diminished in neurons, which may insulate brain physiology and function from p38α-Mnk1-mediated signaling.


Subject(s)
MicroRNAs/metabolism , Mitogen-Activated Protein Kinase 14/genetics , Mitogen-Activated Protein Kinase 14/metabolism , Neurons/physiology , Animals , Base Sequence , Cerebellum/cytology , Cerebellum/physiology , Eukaryotic Initiation Factor-4E/genetics , Eukaryotic Initiation Factor-4E/metabolism , Gene Expression Regulation, Enzymologic , HEK293 Cells , Humans , Mice , Mice, Inbred BALB C , MicroRNAs/genetics , Mitogen-Activated Protein Kinase 11/genetics , Mitogen-Activated Protein Kinase 11/metabolism , Molecular Sequence Data , Neurons/metabolism , Oligonucleotides, Antisense/genetics , Oligonucleotides, Antisense/metabolism , Organ Culture Techniques , Phosphorylation , Protein Biosynthesis , Protein Serine-Threonine Kinases/genetics , Protein Serine-Threonine Kinases/metabolism , Signal Transduction
6.
Future Virol ; 6(9): 1045-1058, 2011 Sep.
Article in English | MEDLINE | ID: mdl-21984883

ABSTRACT

In cancerous cells, physiologically tight regulation of protein synthesis is lost, contributing to uncontrolled growth and proliferation. We describe a novel experimental cancer therapy approach based on genetically recombinant poliovirus that targets an intriguing aberration of translation control in malignancy. This strategy is based on the confluence of several factors enabling specific and efficacious cancer cell targeting. Poliovirus naturally targets the vast majority of ectodermal/neuroectodermal cancers expressing its cellular receptor. Evidence from glioblastoma patients suggests that the poliovirus receptor is ectopically upregulated on tumor cells and may be associated with stem cell-like cancer cell populations and proliferating tumor vasculature. We exploit poliovirus' reliance on an unorthodox mechanism of protein synthesis initiation to selectively drive viral translation, propagation and cytotoxicity in glioblastoma. PVSRIPO, a prototype nonpathogenic poliovirus recombinant, is scheduled to enter clinical investigation against glioblastoma.

7.
Mol Cell Biol ; 31(14): 2947-59, 2011 Jul.
Article in English | MEDLINE | ID: mdl-21576361

ABSTRACT

Signal transduction through mitogen-activated protein kinases (MAPKs) is implicated in growth and proliferation control through translation regulation and involves posttranslational modification of translation initiation factors. For example, convergent MAPK signals to Mnk1 lead to phosphorylation of eukaryotic translation initiation factor 4E (eIF4E), which has been linked to malignant transformation. However, understanding the compound effects of mitogenic signaling on the translation apparatus and on protein synthesis control remains elusive. This is particularly true for the central scaffold of the translation initiation apparatus and ribosome adaptor eIF4G. To unravel the effects of signal transduction to eIF4G on translation, we used specific activation of protein kinase C (PKC)-Ras-Erk signaling with phorbol esters. Phospho-proteomic and mutational analyses revealed that eIF4G1 is a substrate for PKCα at Ser1186. We show that PKCα activation elicits a cascade of orchestrated phosphorylation events that may modulate eIF4G1 structure and control interaction with the eIF4E kinase, Mnk1.


Subject(s)
Eukaryotic Initiation Factor-4G/metabolism , Intracellular Signaling Peptides and Proteins/metabolism , Isoenzymes/metabolism , Protein Kinase C-alpha/metabolism , Protein Serine-Threonine Kinases/metabolism , Signal Transduction/physiology , Amino Acid Sequence , Animals , Enzyme Activation , Enzyme Inhibitors/metabolism , Eukaryotic Initiation Factor-4G/genetics , Extracellular Signal-Regulated MAP Kinases/metabolism , HEK293 Cells , Humans , Intracellular Signaling Peptides and Proteins/genetics , Isoenzymes/genetics , Molecular Sequence Data , Phorbol Esters/metabolism , Phosphorylation , Protein Binding , Protein Kinase C-alpha/genetics , Protein Serine-Threonine Kinases/genetics , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/metabolism , ras Proteins/metabolism
8.
Mol Cell Biol ; 30(21): 5160-7, 2010 Nov.
Article in English | MEDLINE | ID: mdl-20823271

ABSTRACT

The m(7)G cap binding protein eukaryotic initiation factor 4E (eIF4E) is a rate-limiting determinant of protein synthesis. Elevated eIF4E levels, commonly associated with neoplasia, promote oncogenesis, and phosphorylation of eIF4E at Ser209 is critical for its tumorigenic potential. eIF4E phosphorylation is catalyzed by mitogen-activated protein kinase (MAPK)-interacting serine/threonine kinase (Mnk), a substrate of Erk1/2 and p38 MAPKs. Interaction with the scaffolding protein eIF4G, which also binds eIF4E, brings Mnk and its substrate into physical proximity. Thus, Mnk-eIF4G interaction is important for eIF4E phosphorylation. Through coimmunoprecipitation assays, we showed that MAPK-mediated phosphorylation of the Mnk1 active site controls eIF4G binding. Utilizing a naturally occurring splice variant, we demonstrated that the C-terminal domain of Mnk1 restricts its interaction with eIF4G, preventing eIF4E phosphorylation in the absence of MAPK signaling. Furthermore, using a small-molecule Mnk1 inhibitor and kinase-dead mutant, we established that Mnk1 autoregulates its interaction with eIF4G, releasing itself from the scaffold after phosphorylation of its substrate. Our findings indicate tight control of eIF4E phosphorylation through modulation of Mnk1-eIF4G interaction.


Subject(s)
Eukaryotic Initiation Factor-4E/metabolism , Extracellular Signal-Regulated MAP Kinases/metabolism , Intracellular Signaling Peptides and Proteins/metabolism , Protein Serine-Threonine Kinases/metabolism , Animals , Base Sequence , Cell Line , DNA Primers/genetics , Eukaryotic Initiation Factor-4E/genetics , Eukaryotic Initiation Factor-4G/genetics , Eukaryotic Initiation Factor-4G/metabolism , Humans , Intracellular Signaling Peptides and Proteins/antagonists & inhibitors , Intracellular Signaling Peptides and Proteins/chemistry , Intracellular Signaling Peptides and Proteins/genetics , MAP Kinase Signaling System , Mice , Mice, Knockout , Phosphorylation , Protein Serine-Threonine Kinases/antagonists & inhibitors , Protein Serine-Threonine Kinases/chemistry , Protein Serine-Threonine Kinases/deficiency , Protein Serine-Threonine Kinases/genetics , Recombinant Proteins/genetics , Recombinant Proteins/metabolism
9.
J Virol ; 84(1): 270-9, 2010 Jan.
Article in English | MEDLINE | ID: mdl-19864386

ABSTRACT

Human pathogenic viruses manipulate host cell translation machinery to ensure efficient expression of viral genes and to thwart host cell protein synthesis. Viral strategies include cleaving translation factors, manipulating translation factor abundance and recruitment into translation initiation complexes, or expressing viral translation factor analogs. Analyzing translation factors in herpes simplex virus type 1 (HSV-1)-infected HeLa cells, we found diminished association of the polyadenylate-binding protein (PABP) with the cap-binding complex. Although total PABP levels were unchanged, HSV-1 infection prompted accumulation of cytoplasmic PABPC1, but not its physiologic binding partner PABP-interacting protein 2 (Paip2), in the nucleus. Using glutathione S-transferase-PABP pull-down and proteomic analyses, we identified several viral proteins interacting with PABPC1 including tegument protein UL47 and infected-cell protein ICP27. Transient expression of ICP27 and UL47 in HeLa cells suggested that ICP27 and UL47 jointly displace Paip2 from PABP. ICP27 expression alone was sufficient to cause PABPC1 redistribution to the nucleus. ICP27 and UL47 did not alter translation efficiency of transfected reporter RNAs but modulated transcript abundance and expression of reporter cDNAs in transfected cells. This indicates that redistribution of PABPC1 may be involved in co- and posttranscriptional regulation of mRNA processing and/or nuclear export by HSV-1 gene regulatory proteins.


Subject(s)
Herpesvirus 1, Human/physiology , Immediate-Early Proteins/metabolism , Poly(A)-Binding Protein I/metabolism , Viral Fusion Proteins/metabolism , Active Transport, Cell Nucleus , Gene Expression Regulation , HeLa Cells , Herpes Simplex , Humans , Peptide Initiation Factors/metabolism , Protein Binding , RNA, Messenger/analysis , RNA-Binding Proteins/metabolism , Repressor Proteins/metabolism
10.
RNA ; 14(10): 2170-82, 2008 Oct.
Article in English | MEDLINE | ID: mdl-18755839

ABSTRACT

Protein synthesis is tightly controlled by assembly of an intricate ribonucleoprotein complex at the m(7)GTP-cap on eukaryotic mRNAs. Ensuing linear scanning of the 5' untranslated region (UTR) is believed to transfer the preinitiation complex to the initiation codon. Eukaryotic mRNAs are characterized by significant 5' UTR heterogeneity, raising the possibility of differential control of translation initiation rate at individual mRNAs. Curiously, many mRNAs with unconventional, highly structured 5' UTRs encode proteins with central biological roles in growth control, metabolism, or stress response. The 5' UTRs of such mRNAs may influence protein synthesis rate in multiple ways, but most significantly they have been implicated in mediating alternative means of translation initiation. Cap-independent initiation bypasses strict control over the formation of initiation intermediates at the m(7)GTP cap. However, the molecular mechanisms that favor alternative means of ribosome recruitment are not understood. Here we provide evidence that eukaryotic initiation factor (eIF) 4G controls cap-independent translation initiation at the c-myc and vascular endothelial growth factor (VEGF) 5' UTRs in vivo. Cap-independent translation was investigated in tetracycline-inducible cell lines expressing either full-length eIF4G or a C-terminal fragment (Ct) lacking interaction with eIF4E and poly(A) binding protein. Expression of Ct, but not intact eIF4G, potently stimulated cap-independent initiation at the c-myc/VEGF 5' UTRs. In vitro RNA-binding assays suggest that stimulation of cap-independent translation initiation by Ct is due to direct association with the c-myc/VEGF 5' UTR, enabling 43S preinitiation complex recruitment. Our work demonstrates that variant translation initiation factors enable unconventional translation initiation at mRNA subsets with distinct structural features.


Subject(s)
5' Untranslated Regions/metabolism , Eukaryotic Initiation Factor-4G/metabolism , Peptide Chain Initiation, Translational , RNA Cap Analogs/metabolism , Cell Line , Eukaryotic Initiation Factor-4G/genetics , Humans , Polyadenylation , Proto-Oncogene Proteins c-myc/biosynthesis , Vascular Endothelial Growth Factor A/biosynthesis
11.
RNA ; 13(9): 1582-93, 2007 Sep.
Article in English | MEDLINE | ID: mdl-17652408

ABSTRACT

The 3' poly(A) tail present on the majority of mature eukaryotic mRNAs is an important regulator of protein synthesis and mRNA stability. The poly(A) tail improves the efficiency of translation initiation through recruitment of PABP, enabling its interaction with eIF4F located at the mRNA 5'-end. Recent evidence has also implicated a possible role for PABP and the poly(A) tract in translation control at steps beyond the initiation phase. Similar to conventional mRNAs, plus-strand RNA virus genomes that utilize internal ribosome entry sites (IRESes) to promote cap-independent translation are influenced by PABP and poly(A) status. However, the relative contribution of these factors to translation initiation mediated by distinct IRESes is unclear. We have investigated cis- and trans-acting effects of poly(A) and PABP, respectively, on RNAs harboring IRESes from three diverse viruses: encephalomyocarditis virus (EMCV), hepatitis C virus (HCV), and coxsackievirus B3 (CBV3). A 3' poly(A) tract enhanced translation of both capped and IRES-containing reporter RNAs. However, only CBV3 and capped transcripts were stabilized as a result of polyadenylation. Correspondingly, translation of polyadenylated CBV3 and capped RNAs displayed heightened sensitivity to the PABP inhibitor Paip2 compared with EMCV and HCV. Sucrose density gradient analyses suggested a stimulatory role for PABP and 3' poly(A) in the CBV3 initiation phase, while assembly of HCV and EMCV RNAs into ribosomal complexes was little affected by either factor. Collectively, the observed differential effects of PABP and poly(A) on translation imply mechanistic differences between viral IRES elements and suggest modulating roles for PABP and the poly(A) tail at multiple phases of translation.


Subject(s)
Poly(A)-Binding Proteins/physiology , Protein Biosynthesis/genetics , RNA, Viral/genetics , Ribosomes/physiology , Encephalomyocarditis virus/genetics , Enterovirus B, Human/genetics , Escherichia coli , HeLa Cells , Hepacivirus/genetics , Humans , Poly A/genetics , Poly(A)-Binding Proteins/chemistry , Poly(A)-Binding Proteins/genetics , RNA Stability/genetics , Ribosomes/chemistry , Ribosomes/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...