Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 26
Filter
1.
Stem Cell Reports ; 12(4): 787-800, 2019 04 09.
Article in English | MEDLINE | ID: mdl-30853374

ABSTRACT

In type 1 diabetes, a renewable source of human pancreatic ß cells, in particular from human induced pluripotent stem cell (hiPSC) origin, would greatly benefit cell therapy. Earlier work showed that pancreatic progenitors differentiated from human embryonic stem cells in vitro can further mature to become glucose responsive following macroencapsulation and transplantation in mice. Here we took a similar approach optimizing the generation of pancreatic progenitors from hiPSCs. This work demonstrates that hiPSCs differentiated to pancreatic endoderm in vitro can be efficiently and robustly generated under large-scale conditions. The hiPSC-derived pancreatic endoderm cells (HiPECs) can further differentiate into glucose-responsive islet-like cells following macroencapsulation and in vivo implantation. The HiPECs can protect mice from streptozotocin-induced hyperglycemia and maintain normal glucose homeostasis and equilibrated plasma glucose concentrations at levels similar to the human set point. These results further validate the potential use of hiPSC-derived islet cells for application in clinical settings.


Subject(s)
Diabetes Mellitus, Experimental/metabolism , Diabetes Mellitus, Experimental/therapy , Induced Pluripotent Stem Cells/cytology , Insulin-Secreting Cells/cytology , Insulin-Secreting Cells/metabolism , Stem Cell Transplantation , Animals , Biomarkers , Blood Glucose , C-Peptide/blood , Cell Differentiation , Diabetes Mellitus, Experimental/blood , Diabetes Mellitus, Experimental/etiology , Disease Models, Animal , Endoderm/cytology , Fluorescent Antibody Technique , Humans , Hyperglycemia/etiology , Hyperglycemia/metabolism , Hyperglycemia/therapy , Immunophenotyping , Insulin/biosynthesis , Mice , Models, Biological , Treatment Outcome
2.
Stem Cells Transl Med ; 4(10): 1214-22, 2015 Oct.
Article in English | MEDLINE | ID: mdl-26304037

ABSTRACT

UNLABELLED: The PEC-01 cell population, differentiated from human embryonic stem cells (hESCs), contains pancreatic progenitors (PPs) that, when loaded into macroencapsulation devices (to produce the VC-01 candidate product) and transplanted into mice, can mature into glucose-responsive insulin-secreting cells and other pancreatic endocrine cells involved in glucose metabolism. We modified the protocol for making PEC-01 cells such that 73%-80% of the cell population consisted of PDX1-positive (PDX1+) and NKX6.1+ PPs. The PPs were further differentiated to islet-like cells (ICs) that reproducibly contained 73%-89% endocrine cells, of which approximately 40%-50% expressed insulin. A large fraction of these insulin-positive cells were single hormone-positive and expressed the transcription factors PDX1 and NKX6.1. To preclude a significant contribution of progenitors to the in vivo function of ICs, we used a simple enrichment process to remove remaining PPs, yielding aggregates that contained 93%-98% endocrine cells and 1%-3% progenitors. Enriched ICs, when encapsulated and implanted into mice, functioned similarly to the VC-01 candidate product, demonstrating conclusively that in vitro-produced hESC-derived insulin-producing cells can mature and function in vivo in devices. A scaled version of our suspension culture was used, and the endocrine aggregates could be cryopreserved and retain functionality. Although ICs expressed multiple important ß cell genes, the cells contained relatively low levels of several maturity-associated markers. Correlating with this, the time to function of ICs was similar to PEC-01 cells, indicating that ICs required cell-autonomous maturation after delivery in vivo, which would occur concurrently with graft integration into the host. SIGNIFICANCE: Type 1 diabetes (T1D) affects approximately 1.25 million people in the U.S. alone and is deadly if not managed with insulin injections. This paper describes the production of insulin-producing cells in vitro and a new protocol for producing the cells, representing another potential cell source for a diabetes cell therapy. These cells can be loaded into a protective device that is implanted under the skin. The device is designed to protect the cells from immune rejection by the implant recipient. The implant can engraft and respond to glucose by secreting insulin, thus potentially replacing the ß cells lost in patients with T1D.


Subject(s)
Human Embryonic Stem Cells/cytology , Insulin-Secreting Cells/cytology , Insulin/biosynthesis , Animals , Biomarkers , Blood Glucose/analysis , Cell Differentiation , Cell Separation/methods , Cells, Cultured , Cells, Immobilized/transplantation , Cryopreservation , Gene Expression Profiling , Homeodomain Proteins/biosynthesis , Human Embryonic Stem Cells/metabolism , Humans , Insulin-Secreting Cells/metabolism , Insulin-Secreting Cells/transplantation , Mice , Proinsulin/metabolism , Protein Processing, Post-Translational , Reproducibility of Results , Trans-Activators/biosynthesis
3.
PLoS One ; 7(5): e37004, 2012.
Article in English | MEDLINE | ID: mdl-22623968

ABSTRACT

Development of a human embryonic stem cell (hESC)-based therapy for type 1 diabetes will require the translation of proof-of-principle concepts into a scalable, controlled, and regulated cell manufacturing process. We have previously demonstrated that hESC can be directed to differentiate into pancreatic progenitors that mature into functional glucose-responsive, insulin-secreting cells in vivo. In this study we describe hESC expansion and banking methods and a suspension-based differentiation system, which together underpin an integrated scalable manufacturing process for producing pancreatic progenitors. This system has been optimized for the CyT49 cell line. Accordingly, qualified large-scale single-cell master and working cGMP cell banks of CyT49 have been generated to provide a virtually unlimited starting resource for manufacturing. Upon thaw from these banks, we expanded CyT49 for two weeks in an adherent culture format that achieves 50-100 fold expansion per week. Undifferentiated CyT49 were then aggregated into clusters in dynamic rotational suspension culture, followed by differentiation en masse for two weeks with a four-stage protocol. Numerous scaled differentiation runs generated reproducible and defined population compositions highly enriched for pancreatic cell lineages, as shown by examining mRNA expression at each stage of differentiation and flow cytometry of the final population. Islet-like tissue containing glucose-responsive, insulin-secreting cells was generated upon implantation into mice. By four- to five-months post-engraftment, mature neo-pancreatic tissue was sufficient to protect against streptozotocin (STZ)-induced hyperglycemia. In summary, we have developed a tractable manufacturing process for the generation of functional pancreatic progenitors from hESC on a scale amenable to clinical entry.


Subject(s)
Batch Cell Culture Techniques/methods , Cell Differentiation/physiology , Diabetes Mellitus, Type 1/therapy , Embryonic Stem Cells/cytology , Embryonic Stem Cells/transplantation , Insulin-Secreting Cells/cytology , Analysis of Variance , Animals , Cryopreservation/methods , Embryonic Stem Cells/physiology , Flow Cytometry , Fluorescent Antibody Technique , Gene Expression Profiling , Humans , Male , Mice , Mice, SCID , Streptozocin
4.
Genes Cancer ; 1(4): 316-330, 2010 Apr 01.
Article in English | MEDLINE | ID: mdl-20740050

ABSTRACT

The transcription factor ATF2 was previously shown to be an ATM substrate. Upon phosphorylation by ATM, ATF2 exhibits a transcription-independent function in the DNA damage response through localization to DNA repair foci and control of cell cycle arrest. To assess the physiological significance of this phosphorylation, we generated ATF2 mutant mice in which the ATM phosphoacceptor sites (S472/S480) were mutated (ATF2(KI)). ATF2(KI) mice are more sensitive to ionizing radiation (IR) than wild-type (ATF2 (WT)) mice: following IR, ATF2(KI) mice exhibited higher levels of apoptosis in the intestinal crypt cells and impaired hepatic steatosis. Molecular analysis identified impaired activation of the cell cycle regulatory protein p21(Cip/Waf1) in cells and tissues of IR-treated ATF2(KI) mice, which was p53 independent. Analysis of tumor development in p53(KO) crossed with ATF2(KI) mice indicated a marked decrease in amount of time required for tumor development. Further, when subjected to two-stage skin carcinogenesis process, ATF2(KI) mice developed skin tumors faster and with higher incidence, which also progressed to the more malignant carcinomas, compared with the control mice. Using 3 mouse models, we establish the importance of ATF2 phosphorylation by ATM in the acute cellular response to DNA damage and maintenance of genomic stability.

5.
PLoS Genet ; 6(12): e1001258, 2010 Dec 23.
Article in English | MEDLINE | ID: mdl-21203491

ABSTRACT

The transcription factor ATF2 has been shown to attenuate melanoma susceptibility to apoptosis and to promote its ability to form tumors in xenograft models. To directly assess ATF2's role in melanoma development, we crossed a mouse melanoma model (Nras(Q61K)::Ink4a⁻/⁻) with mice expressing a transcriptionally inactive form of ATF2 in melanocytes. In contrast to 7/21 of the Nras(Q61K)::Ink4a⁻/⁻ mice, only 1/21 mice expressing mutant ATF2 in melanocytes developed melanoma. Gene expression profiling identified higher MITF expression in primary melanocytes expressing transcriptionally inactive ATF2. MITF downregulation by ATF2 was confirmed in the skin of Atf2⁻/⁻ mice, in primary human melanocytes, and in 50% of human melanoma cell lines. Inhibition of MITF transcription by MITF was shown to be mediated by ATF2-JunB-dependent suppression of SOX10 transcription. Remarkably, oncogenic BRAF (V600E)-dependent focus formation of melanocytes on soft agar was inhibited by ATF2 knockdown and partially rescued upon shMITF co-expression. On melanoma tissue microarrays, a high nuclear ATF2 to MITF ratio in primary specimens was associated with metastatic disease and poor prognosis. Our findings establish the importance of transcriptionally active ATF2 in melanoma development through fine-tuning of MITF expression.


Subject(s)
Activating Transcription Factor 2/metabolism , Gene Expression Regulation, Neoplastic , Melanoma/metabolism , Microphthalmia-Associated Transcription Factor/genetics , Activating Transcription Factor 2/genetics , Animals , Cell Line, Tumor , Cells, Cultured , Down-Regulation , Female , Humans , Male , Melanocytes/metabolism , Melanoma/genetics , Melanoma/pathology , Mice , Mice, Inbred C57BL , Mice, Knockout , Microphthalmia-Associated Transcription Factor/metabolism , Skin/metabolism , Skin/pathology
6.
EMBO Rep ; 10(9): 965-72, 2009 Sep.
Article in English | MEDLINE | ID: mdl-19680286

ABSTRACT

Most tumours contain a heterogeneous population of cancer cells, which harbour a range of genetic mutations and have probably undergone deregulated differentiation programmes that allow them to adapt to tumour microenvironments. Another explanation for tumour heterogeneity might be that the cells within a tumour are derived from tumour-initiating cells through diverse differentiation programmes. Tumour-initiating cells are thought to constitute one or more distinct subpopulations within a tumour and to drive tumour initiation, development and metastasis, as well as to be responsible for their recurrence after therapy. Recent studies have raised crucial questions about the nature, frequency and importance of melanoma-initiating cells. Here, we discuss our current understanding of melanoma-initiating cells and outline several approaches that the scientific community might consider to resolve the controversies surrounding these cells.


Subject(s)
Cell Transformation, Neoplastic/metabolism , Cell Transformation, Neoplastic/pathology , Melanoma/metabolism , Melanoma/pathology , Animals , Biomarkers, Tumor , Cell Proliferation , Cell Transformation, Neoplastic/genetics , Disease Models, Animal , Humans , Melanoma/genetics , Melanoma/therapy
7.
Cell Cycle ; 7(15): 2341-5, 2008 Aug.
Article in English | MEDLINE | ID: mdl-18677098

ABSTRACT

The Activating Transcription Factor (ATF2) gene encodes a transcription factor important for normal cellular development and survival. In addition to its role as a transcription factor, ATF2 functions in the DNA damage response and in control of HAT complex activity. So far genetic changes in ATF2 have not been identified in human tumors. Yet altered expression and subcellular localization of ATF2 are associated with tumor stage and prognosis. Our studies in melanoma tumor models identified oncogenic ATF2 activity correlating with development of this tumor type. Inhibiting ATF2 suffices to impede melanoma development. Yet work in other tumor models, including breast and skin tumors reveals that ATF2, in cooperation with mutated oncogenes or tumor suppressor genes, can also elicit tumor suppressor function. These findings suggest tissue- and tumor-specific function of ATF2. This review summarizes the current understanding of ATF2 regulation and function.


Subject(s)
Activating Transcription Factor 2/physiology , Genes, Tumor Suppressor/physiology , Oncogenes/physiology , Activating Transcription Factor 2/genetics , Animals , Breast Neoplasms/genetics , Breast Neoplasms/metabolism , DNA Damage/physiology , Histone Acetyltransferases/metabolism , Humans , Melanoma/genetics , Melanoma/metabolism , Models, Biological , Skin Neoplasms/genetics , Skin Neoplasms/metabolism , Transcription Factors/physiology
8.
J Immunol ; 181(3): 2056-64, 2008 Aug 01.
Article in English | MEDLINE | ID: mdl-18641343

ABSTRACT

IFN-gamma production by T cells is pivotal for defense against many pathogens, and the proximal promoter of IFN-gamma, -73 to -48 bp upstream of the transcription start site, is essential for its expression. However, transcriptional regulation mechanisms through this promoter in primary human cells remain unclear. We studied the effects of cAMP response element binding protein/activating transcription factor (CREB/ATF) and AP-1 transcription factors on the proximal promoter of IFN-gamma in human T cells stimulated with Mycobacterium tuberculosis. Using EMSA, supershift assays, and promoter pulldown assays, we demonstrated that CREB, ATF-2, and c-Jun, but not cyclic AMP response element modulator, ATF-1, or c-Fos, bind to the proximal promoter of IFN-gamma upon stimulation, and coimmunoprecipitation indicated the possibility of interaction among these transcription factors. Chromatin immunoprecipitation confirmed the recruitment of these transcription factors to the IFN-gamma proximal promoter in live Ag-activated T cells. Inhibition of ATF-2 activity in T cells with a dominant-negative ATF-2 peptide or with small interfering RNA markedly reduced the expression of IFN-gamma and decreased the expression of CREB and c-Jun. These findings suggest that CREB, ATF-2, and c-Jun are recruited to the IFN-gamma proximal promoter and that they up-regulate IFN-gamma transcription in response to microbial Ag. Additionally, ATF-2 controls expression of CREB and c-Jun during T cell activation.


Subject(s)
Activating Transcription Factor 2/metabolism , Antigens, Bacterial/immunology , Cyclic AMP Response Element-Binding Protein/metabolism , Interferon-gamma/metabolism , T-Lymphocytes/metabolism , Transcription Factor AP-1/metabolism , Activating Transcription Factor 2/genetics , Activating Transcription Factor 2/pharmacology , CD3 Complex/immunology , Cell Survival , Cells, Cultured , Electrophoretic Mobility Shift Assay , Humans , Interferon-gamma/genetics , Mycobacterium tuberculosis/immunology , Promoter Regions, Genetic/genetics , Protein Binding , RNA, Small Interfering/genetics , T-Lymphocytes/drug effects , T-Lymphocytes/immunology
9.
J Biol Chem ; 283(25): 17605-14, 2008 Jun 20.
Article in English | MEDLINE | ID: mdl-18397884

ABSTRACT

TIP60 (HTATIP) is a histone acetyltransferase (HAT) whose function is critical in regulating ataxia-telangiectasia mutated (ATM) activation, gene expression, and chromatin acetylation in DNA repair. Here we show that under non-stressed conditions, activating transcription factor-2 (ATF2) in cooperation with Cul3 ubiquitin ligase promotes degradation of TIP60, thereby attenuating its HAT activity. Inhibiting either ATF2 or Cul3 expression by small interfering RNA stabilizes the TIP60 protein. ATF2 association with TIP60 on chromatin is decreased following exposure to ionizing radiation (IR), resulting in enhanced TIP60 stability and activity. We also identified a panel of melanoma and prostate cancer cell lines whose ATF2 expression is inversely correlated with TIP60 levels and ATM activation after IR. Inhibition of ATF2 expression in these lines restored TIP60 protein levels and both basal and IR-induced levels of ATM activity. Our study provides novel insight into regulation of ATM activation by ATF2-dependent control of TIP60 stability and activity.


Subject(s)
Activating Transcription Factor 2/metabolism , Cell Cycle Proteins/metabolism , DNA-Binding Proteins/metabolism , Gene Expression Regulation, Neoplastic , Gene Expression Regulation , Histone Acetyltransferases/chemistry , Protein Serine-Threonine Kinases/metabolism , Tumor Suppressor Proteins/metabolism , Ataxia Telangiectasia Mutated Proteins , Cell Line, Tumor , Chromatin/metabolism , Cullin Proteins/metabolism , DNA Damage , HeLa Cells , Histone Acetyltransferases/metabolism , Humans , Lysine Acetyltransferase 5 , Models, Biological , Models, Genetic , Protein Binding , RNA Interference
11.
Proc Natl Acad Sci U S A ; 105(5): 1674-9, 2008 Feb 05.
Article in English | MEDLINE | ID: mdl-18227516

ABSTRACT

Activating transcription factor 2 (ATF2) regulates transcription in response to stress and growth factor stimuli. Here, we use a mouse model in which ATF2 was selectively deleted in keratinocytes. Crossing the conditionally expressed ATF2 mutant with K14-Cre mice (K14.ATF2(f/f)) resulted in selective expression of mutant ATF2 within the basal layer of the epidermis. When subjected to a two-stage skin carcinogenesis protocol [7,12-dimethylbenz[a]anthracene/phorbol 12-tetradecanoate 13-acetate (DMBA/TPA)], K14.ATF2(f/f) mice showed significant increases in both the incidence and prevalence of papilloma development compared with the WT ATF2 mice. Consistent with these findings, keratinocytes of K14.ATF2(f/f) mice exhibit greater anchorage-independent growth compared with ATF2 WT keratinocytes. Papillomas of K14.ATF2(f/f) mice exhibit reduced expression of presenilin1, which is associated with enhanced beta-catenin and cyclin D1, and reduced Notch1 expression. Significantly, a reduction of nuclear ATF2 and increased beta-catenin expression were seen in samples of squamous and basal cell carcinoma, as opposed to normal skin. Our data reveal that loss of ATF2 transcriptional activity serves to promote skin tumor formation, thereby indicating a suppressor activity of ATF2 in skin tumor formation.


Subject(s)
Activating Transcription Factor 2/physiology , Papilloma/genetics , Skin Neoplasms/genetics , Tumor Suppressor Proteins/physiology , 9,10-Dimethyl-1,2-benzanthracene/toxicity , Activating Transcription Factor 2/analysis , Activating Transcription Factor 2/genetics , Animals , Apoptosis , Carcinogens/toxicity , Cell Proliferation , Cyclin D1/metabolism , DNA/biosynthesis , Epidermis/drug effects , Epidermis/metabolism , Epidermis/pathology , Keratinocytes/drug effects , Keratinocytes/metabolism , Keratinocytes/pathology , Mice , Mice, Knockout , Papilloma/chemically induced , Papilloma/pathology , Presenilin-1/metabolism , Proto-Oncogene Proteins c-myb/metabolism , Receptor, Notch1/metabolism , Skin Neoplasms/chemically induced , Skin Neoplasms/pathology , Tetradecanoylphorbol Acetate/toxicity , Tissue Array Analysis , Tumor Suppressor Proteins/analysis , Tumor Suppressor Proteins/genetics , beta Catenin/metabolism
12.
Clin Cancer Res ; 13(22 Pt 1): 6769-78, 2007 Nov 15.
Article in English | MEDLINE | ID: mdl-18006779

ABSTRACT

PURPOSE: Sensitize melanomas to apoptosis and inhibit their growth and metastatic potential by compounds that mimic the activities of activating transcription factor 2 (ATF2)-driven peptides. EXPERIMENTAL DESIGN: Small-molecule chemical library consisting of 3,280 compounds was screened to identify compounds that elicit properties identified for ATF2 peptide, including (a) sensitization of melanoma cells to apoptosis, (b) inhibition of ATF2 transcriptional activity, (c) activation of c-Jun NH(2)-terminal kinase (JNK) and c-Jun transcriptional activity, and (d) inhibition of melanoma growth and metastasis in mouse models. RESULTS: Two compounds, celastrol (CSL) and acetyl isogambogic acid, could, within a low micromolar range, efficiently elicit cell death in melanoma cells. Both compounds efficiently inhibit ATF2 transcriptional activities, activate JNK, and increase c-Jun transcriptional activities. Similar to the ATF2 peptide, both compounds require JNK activity for their ability to inhibit melanoma cell viability. Derivatives of CSL were identified as potent inducers of cell death in mouse and human melanomas. CSL and a derivative (CA19) could also efficiently inhibit growth of human and mouse melanoma tumors and reduce the number of lung metastases in syngeneic and xenograft mouse models. CONCLUSIONS: These studies show for the first time the effect of CSL and acetyl isogambogic acid on melanoma. These compounds elicit activities that resemble the well-characterized ATF2 peptide and may therefore offer new approaches for the treatment of this tumor type.


Subject(s)
Chromones/pharmacology , Melanoma/drug therapy , Skin Neoplasms/drug therapy , Triterpenes/pharmacology , Activating Transcription Factor 2/antagonists & inhibitors , Animals , Cell Line, Tumor , Chromones/chemistry , Chromones/therapeutic use , Drug Evaluation, Preclinical , Drug Screening Assays, Antitumor , Humans , MAP Kinase Kinase 4/metabolism , Melanoma/metabolism , Melanoma/pathology , Mice , Pentacyclic Triterpenes , Proto-Oncogene Proteins c-jun/metabolism , Skin Neoplasms/metabolism , Skin Neoplasms/pathology , Small Molecule Libraries/chemistry , Transcription, Genetic , Triterpenes/chemistry , Triterpenes/therapeutic use
13.
Pigment Cell Res ; 20(6): 498-506, 2007 Dec.
Article in English | MEDLINE | ID: mdl-17935492

ABSTRACT

Signal transduction pathways play a key role in the regulation of key cellular processes, including survival and death. Growing evidence points to changes in signaling pathway that occur during skin tumor development and progression. Such changes impact the activity of downstream substrates, including transcription factors. The activating transcription factor 2 (ATF2) has been implicated in malignant and non-malignant skin tumor developments. ATF2 mediates both transcription and DNA damage control, through its phosphorylation by JNK/p38 or ATM/ATR respectively. Here, we summarize our present understanding of ATF2 regulation, function and contribution to malignant and non-malignant skin tumor development.


Subject(s)
Activating Transcription Factor 2/metabolism , DNA Damage , Activating Transcription Factor 2/genetics , Animals , Humans , Hypoxia , Melanoma/metabolism , Mice , Protein Isoforms/genetics , Protein Isoforms/metabolism , Signal Transduction/physiology , Skin Neoplasms/metabolism , Transcription, Genetic
14.
Cell Cycle ; 6(14): 1762-71, 2007 Jul 15.
Article in English | MEDLINE | ID: mdl-17637567

ABSTRACT

The c-Jun N-terminal kinases (JNKs) are ubiquitous proteins that phosphorylate their substrates, such as transcription factors, in response to physical stress, cytokines or UV radiation. This leads to changes in gene expression, ensuing either cell cycle progression or apoptosis. Active phospho JNK1 is the main in vivo kinase component of the JNK cascade, whereas JNK2 is presumed not to participate as a kinase during JNK signalling. However, there is evidence that JNK isoforms interact functionally in vivo. Also, a recent chemical genetics investigation has confirmed that JNK transient activation leads to cellular proliferation, whereas a sustained one is pro-apoptotic. Here we investigate the phosphorylation pattern of JNK2, with protein biochemistry tools and tandem mass spectrometry. We choose to focus on JNK2 because of its reported constitutive activity in glioma cells. Our results indicate that purified JNK2 from transfected nonstressed 293T cells is a mixture of the mono-sites pThr183 and pTyr185 of its activation loop and of pThr386 along its unique C-terminal region. Upon UV stimulation, its phosphorylation stoichiometry is upregulated on the activation loop, generating a mixture of mono-pTyr185 and the expected dual-pThr183/pTyr185 species, with the pThr386 specie present but unaltered respect to the basal conditions.


Subject(s)
Isoenzymes/metabolism , MAP Kinase Signaling System/physiology , Mitogen-Activated Protein Kinase 9/metabolism , Animals , Cell Line , Enzyme Activation , Humans , Isoenzymes/chemistry , Isoenzymes/genetics , Mass Spectrometry , Mice , Mitogen-Activated Protein Kinase 8/chemistry , Mitogen-Activated Protein Kinase 8/genetics , Mitogen-Activated Protein Kinase 8/metabolism , Mitogen-Activated Protein Kinase 9/chemistry , Mitogen-Activated Protein Kinase 9/genetics , Phosphopeptides/chemistry , Phosphopeptides/metabolism , Phosphorylation , Protein Conformation , Threonine/metabolism , Tyrosine/metabolism
15.
Mol Cell Biol ; 25(19): 8619-30, 2005 Oct.
Article in English | MEDLINE | ID: mdl-16166642

ABSTRACT

We report the identification and characterization of JAMP (JNK1 [Jun N-terminal kinase 1]-associated membrane protein), a predicted seven-transmembrane protein that is localized primarily within the plasma membrane and associates with JNK1 through its C-terminal domain. JAMP association with JNK1 outcompetes JNK1 association with mitogen-activated protein kinase phosphatase 5, resulting in increased and prolonged JNK1 activity following stress. Elevated expression of JAMP following UV or tunicamycin treatment results in sustained JNK activity and a higher level of JNK-dependent apoptosis. Inhibition of JAMP expression by RNA interference reduces the degree and duration of JNK activation and concomitantly the level of stress-induced apoptosis. Through its regulation of JNK1 activity, JAMP emerges as a membrane-anchored regulator of the duration of JNK1 activity in response to diverse stress stimuli.


Subject(s)
Carrier Proteins/physiology , Cell Membrane/metabolism , Gene Expression Regulation, Enzymologic , Membrane Glycoproteins/physiology , 3T3 Cells , Amino Acid Sequence , Animals , Apoptosis , Carrier Proteins/biosynthesis , Cell Line , Cell Line, Tumor , Cell Movement , DNA/metabolism , DNA, Complementary/metabolism , Dual-Specificity Phosphatases , Glycosylation , Green Fluorescent Proteins/metabolism , HeLa Cells , Humans , Immunoprecipitation , MAP Kinase Signaling System , Membrane Glycoproteins/biosynthesis , Mice , Microscopy, Confocal , Molecular Sequence Data , NIH 3T3 Cells , Phosphoprotein Phosphatases/metabolism , Protein Binding , Protein Structure, Tertiary , RNA Interference , Reverse Transcriptase Polymerase Chain Reaction , Subcellular Fractions/metabolism , Time Factors , Tissue Distribution , Transfection , Tunicamycin/pharmacology , Ultraviolet Rays
16.
Mol Cell ; 19(3): 309-20, 2005 Aug 05.
Article in English | MEDLINE | ID: mdl-16061178

ABSTRACT

Activation of the Jun-N-terminal kinase (JNK) signaling cascade by phorbol esters (TPA) or protein kinase C (PKC) is well documented, although the underlying mechanism is not known. Here, we demonstrate that the receptor for activated C kinase 1 (RACK1) serves as an adaptor for PKC-mediated JNK activation. Phosphorylation of JNK by PKC occurs on Ser129 and requires the presence of RACK1. Ser129 phosphorylation augments JNK phosphorylation by MKK4 and/or MKK7 and is required for JNK activation by TPA, TNFalpha, UV irradiation, and PKC, but not by anisomycin or MEKK1. Inhibition of RACK1 expression by siRNA attenuates JNK activation, sensitizes melanoma cells to UV-induced apoptosis, and reduces their tumorigenicity in nude mice. In finding the role of RACK1 in activation of JNK by PKC, our study also highlights the nature of crosstalk between these two signal-transduction pathways.


Subject(s)
JNK Mitogen-Activated Protein Kinases/metabolism , Neoplasm Proteins/metabolism , Protein Kinase C/metabolism , Animals , Anisomycin/pharmacology , Antibodies, Phospho-Specific/metabolism , Apoptosis , Binding Sites , Carbazoles/pharmacology , Catalysis/drug effects , Cell Line , Cell Line, Tumor , Cytoplasm/metabolism , Enzyme Inhibitors/pharmacology , GTP-Binding Proteins , Gene Deletion , Humans , Indoles/pharmacology , Isoenzymes/genetics , Isoenzymes/metabolism , JNK Mitogen-Activated Protein Kinases/genetics , MAP Kinase Kinase 4/genetics , MAP Kinase Kinase 4/metabolism , MAP Kinase Kinase 7/genetics , MAP Kinase Kinase 7/metabolism , MAP Kinase Kinase Kinase 1/genetics , MAP Kinase Kinase Kinase 1/metabolism , Mice , Mice, Nude , Mitogen-Activated Protein Kinase 8/genetics , Mitogen-Activated Protein Kinase 8/metabolism , Mitogen-Activated Protein Kinase 9/genetics , Mitogen-Activated Protein Kinase 9/metabolism , Models, Biological , Neoplasm Proteins/genetics , Phosphopeptides/analysis , Phosphorylation/drug effects , Protein Binding , Protein Kinase C/antagonists & inhibitors , Protein Kinase C/genetics , Protein Kinase C beta , RNA, Small Interfering/genetics , Receptors for Activated C Kinase , Receptors, Cell Surface , Serine/metabolism , Signal Transduction/drug effects , Signal Transduction/physiology , Tetradecanoylphorbol Acetate/pharmacology , Transfection , Tumor Necrosis Factor-alpha/pharmacology , Ultraviolet Rays
17.
Mol Cell ; 18(5): 577-87, 2005 May 27.
Article in English | MEDLINE | ID: mdl-15916964

ABSTRACT

Activating transcription factor 2 (ATF2) is regulated by JNK/p38 in response to stress. Here, we demonstrate that the protein kinase ATM phosphorylates ATF2 on serines 490 and 498 following ionizing radiation (IR). Phosphoantibodies to ATF2(490/8) reveal dose- and time-dependent phosphorylation of ATF2 by ATM that results in its rapid colocalization with gamma-H2AX and MRN components into IR-induced foci (IRIF). Inhibition of ATF2 expression decreased recruitment of Mre11 to IRIF, abrogated S phase checkpoint, reduced activation of ATM, Chk1, and Chk2, and impaired radioresistance. ATF2 requires neither JNK/p38 nor its DNA binding domain for recruitment to IRIF and the S phase checkpoint. Our findings identify a role for ATF2 in the DNA damage response that is uncoupled from its transcriptional activity.


Subject(s)
Cell Cycle Proteins/metabolism , Cyclic AMP Response Element-Binding Protein/metabolism , DNA Damage , DNA-Binding Proteins/metabolism , Protein Serine-Threonine Kinases/metabolism , Transcription Factors/metabolism , Tumor Suppressor Proteins/metabolism , Activating Transcription Factor 2 , Amino Acid Sequence , Animals , Antibodies, Phospho-Specific/metabolism , Ataxia Telangiectasia Mutated Proteins , Cell Cycle Proteins/genetics , Cell Line , Checkpoint Kinase 1 , Checkpoint Kinase 2 , Cyclic AMP Response Element-Binding Protein/genetics , DNA Repair , DNA Repair Enzymes , DNA-Binding Proteins/genetics , Fibroblasts/cytology , Fibroblasts/physiology , Histones/metabolism , Humans , MRE11 Homologue Protein , Mice , Molecular Sequence Data , Nuclear Proteins/metabolism , Phosphorylation , Protein Kinases/metabolism , Protein Serine-Threonine Kinases/genetics , RNA Interference , Radiation, Ionizing , Sequence Alignment , Serine/metabolism , Transcription Factors/genetics , Transcription, Genetic , Tumor Suppressor Proteins/genetics
18.
Cancer Res ; 64(22): 8222-30, 2004 Nov 15.
Article in English | MEDLINE | ID: mdl-15548688

ABSTRACT

The resistance of melanoma to apoptosis, as well as its growth and metastasis capabilities, can be overcome by expression of a peptide derived from amino acid (aa) 51 to 100 of ATF2. Here we show that expression of ATF2((51-100)) in human melanoma cells reduced their growth in nude mice, which was additionally inhibited upon treatment with protein kinase inhibitors UCN-01 or SB203580. Injection of a fusion protein consisting of HIV-TAT and aa 51 to 100 of ATF2 into SW1 melanomas efficiently inhibits their growth and their metastasis up to complete regression. Additionally, expression of a 10aa peptide that corresponds to aa 51 to 60 of ATF2 sensitizes melanoma cells to spontaneous apoptosis, which coincides with activation of caspase 9 and poly(ADP-ribose) polymerase cleavage, and inhibit their growth in vivo. The 10aa peptide increases the association of c-Jun NH(2)-terminal kinase with c-Jun but not with ATF2, resulting in concomitant increase in TRE-mediated transcription. Our study points to mechanisms underlying the activities of the ATF2 peptide while highlighting its possible use in drug design.


Subject(s)
Cell Division/drug effects , Cyclic AMP Response Element-Binding Protein/chemistry , Melanoma/pathology , Neoplasm Metastasis , Peptides/pharmacology , Skin Neoplasms/pathology , Transcription Factors/chemistry , Activating Transcription Factor 2 , Animals , Apoptosis , Humans , Mice , Mice, Nude , Peptides/chemistry
19.
Genes Dev ; 18(19): 2380-91, 2004 Oct 01.
Article in English | MEDLINE | ID: mdl-15466489

ABSTRACT

The small ubiquitin-like modifier (SUMO) modification alters the subcellular distribution and function of its substrates. Here we show the major role of SUMO during the development of the Caenorhabditis elegans reproductive system. smo-1 deletion mutants develop into sterile adults with abnormal somatic gonad, germ line, and vulva. SMO-1::GFP reporter is highly expressed in the somatic reproductive system. smo-1 animals lack a vulval-uterine connection as a result of impaired ventral uterine pi-cell differentiation and anchor cell fusion. Mutations in the LIN-11 LIM domain transcription factor lead to a uterine phenotype that resembles the smo-1 phenotype. LIN-11 is sumoylated, and its sumoylation is required for its activity during uterine morphogenesis. Expression of a SUMO-modified LIN-11 in the smo-1 background partially rescued pi-cell differentiation and retained LIN-11 in nuclear bodies. Thus, our results identify the reproductive system as the major SUMO target during postembryonic development and highlight LIN-11 as a physiological substrate whose sumoylation is associated with the formation of a functional vulval-uterine connection.


Subject(s)
Caenorhabditis elegans/embryology , SUMO-1 Protein/physiology , Uterus/embryology , Vulva/embryology , Animals , Base Sequence , DNA Primers , Female , Fluorescent Antibody Technique , Green Fluorescent Proteins , Luminescent Proteins/genetics , Microscopy, Confocal , Morphogenesis
20.
Cell ; 117(7): 941-52, 2004 Jun 25.
Article in English | MEDLINE | ID: mdl-15210114

ABSTRACT

Hypoxia-inducible factor-1alpha (HIF1alpha) is a central regulator of the cellular response to hypoxia. Prolyl-hydroxylation of HIF1alpha by PHD enzymes is prerequisite for HIF1alpha degradation. Here, we demonstrate that the abundance of PHD1 and PHD3 are regulated via their targeting for proteasome-dependent degradation by the E3 ubiquitin ligases Siah1a/2, under hypoxia conditions. Siah2 null fibroblasts exhibit prolonged PHD3 half-life, resulting in lower levels of HIF1alpha expression during hypoxia. Significantly, hypoxia-induced HIF1alpha expression was completely inhibited in Siah1a/2 null cells, yet could be rescued upon inhibition of PHD3 by RNAi. Siah2 targeting of PHD3 for degradation increases upon exposure to even mild hypoxic conditions, which coincides with increased Siah2 transcription. Siah2 null mice subjected to hypoxia displayed an impaired hyperpneic respiratory response and reduced levels of hemoglobin. Thus, the control of PHD1/3 by Siah1a/2 constitutes another level of complexity in the regulation of HIF1alpha during hypoxia.


Subject(s)
Cell Hypoxia , Gene Expression Regulation , Nuclear Proteins/metabolism , Procollagen-Proline Dioxygenase/genetics , Transcription Factors/metabolism , Animals , Blotting, Western , Cells, Cultured , Female , Fibroblasts/metabolism , HeLa Cells , Humans , Hypoxia-Inducible Factor 1, alpha Subunit , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Nuclear Proteins/genetics , Precipitin Tests , Procollagen-Proline Dioxygenase/metabolism , RNA Interference , Reverse Transcriptase Polymerase Chain Reaction , Spectrometry, Mass, Matrix-Assisted Laser Desorption-Ionization , Transcription, Genetic , Ubiquitin-Protein Ligases
SELECTION OF CITATIONS
SEARCH DETAIL
...