Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 15 de 15
Filter
1.
Cell ; 173(4): 864-878.e29, 2018 05 03.
Article in English | MEDLINE | ID: mdl-29681454

ABSTRACT

Diversity in the genetic lesions that cause cancer is extreme. In consequence, a pressing challenge is the development of drugs that target patient-specific disease mechanisms. To address this challenge, we employed a chemistry-first discovery paradigm for de novo identification of druggable targets linked to robust patient selection hypotheses. In particular, a 200,000 compound diversity-oriented chemical library was profiled across a heavily annotated test-bed of >100 cellular models representative of the diverse and characteristic somatic lesions for lung cancer. This approach led to the delineation of 171 chemical-genetic associations, shedding light on the targetability of mechanistic vulnerabilities corresponding to a range of oncogenotypes present in patient populations lacking effective therapy. Chemically addressable addictions to ciliogenesis in TTC21B mutants and GLUT8-dependent serine biosynthesis in KRAS/KEAP1 double mutants are prominent examples. These observations indicate a wealth of actionable opportunities within the complex molecular etiology of cancer.


Subject(s)
Carcinoma, Non-Small-Cell Lung/pathology , Cell Proliferation/drug effects , Lung Neoplasms/pathology , Small Molecule Libraries/pharmacology , Carcinoma, Non-Small-Cell Lung/metabolism , Cell Line, Tumor , Cytochrome P450 Family 4/deficiency , Cytochrome P450 Family 4/genetics , Drug Discovery , G1 Phase Cell Cycle Checkpoints/drug effects , Glucocorticoids/pharmacology , Glucose Transport Proteins, Facilitative/antagonists & inhibitors , Glucose Transport Proteins, Facilitative/genetics , Glucose Transport Proteins, Facilitative/metabolism , Humans , Kelch-Like ECH-Associated Protein 1/genetics , Kelch-Like ECH-Associated Protein 1/metabolism , Lung Neoplasms/metabolism , Microtubule-Associated Proteins/genetics , Microtubule-Associated Proteins/metabolism , Mutation , NF-E2-Related Factor 2/antagonists & inhibitors , NF-E2-Related Factor 2/genetics , NF-E2-Related Factor 2/metabolism , Proto-Oncogene Proteins p21(ras)/genetics , Proto-Oncogene Proteins p21(ras)/metabolism , RNA Interference , RNA, Small Interfering/metabolism , Receptor, Notch2/genetics , Receptor, Notch2/metabolism , Receptors, Glucocorticoid/antagonists & inhibitors , Receptors, Glucocorticoid/genetics , Receptors, Glucocorticoid/metabolism , Small Molecule Libraries/chemistry , Small Molecule Libraries/metabolism
3.
J Biol Chem ; 287(27): 22948-58, 2012 Jun 29.
Article in English | MEDLINE | ID: mdl-22577135

ABSTRACT

Previous studies suggest that up-regulation of Ras signaling in neurons promotes gliosis and astrocytoma formation in a cell nonautonomous manner. However, the underlying mechanisms remain unknown. To address this question, we generated compound mice (LSL Kras G12D/+;CamKII-Cre) that express oncogenic Kras from its endogenous locus in postmitotic neurons after birth. These mice developed progressive gliosis, which is associated with hyperactivation of Ras signaling pathways. Microarray analysis identified S100A8 and S100A9 as two secreted molecules that are significantly overexpressed in mutant cortices. In contrast to their usual predominant expression in myeloid cells, we found that overexpression of S100A8 and S100A9 in the mutant cortex is primarily in neurons. This neuronal expression pattern is associated with increased infiltration of microglia in mutant cortex. Moreover, purified S100A8-S100A9 but not S100A8 or S100A9 alone promotes growth of primary astrocytes in vitro through both TLR4 and receptor of advanced glycation end product receptors. In summary, our results identify overexpression of S100A8-S100A9 in neurons as an early step in oncogenic Kras-induced gliosis. These molecules expressed in nonhematopoietic cells may be involved in tumorigenesis at a stage much earlier than what has been reported previously.


Subject(s)
Calgranulin A/genetics , Calgranulin B/genetics , Gliosis/genetics , Neurons/physiology , Proto-Oncogene Proteins/genetics , ras Proteins/genetics , Animals , Astrocytes/cytology , Astrocytes/physiology , Brain Neoplasms/genetics , Brain Neoplasms/physiopathology , Calgranulin A/metabolism , Calgranulin B/metabolism , Cerebral Cortex/cytology , Gene Expression Regulation, Neoplastic/physiology , Glioblastoma/genetics , Glioblastoma/physiopathology , Gliosis/physiopathology , Mice , Mice, Inbred C57BL , Mice, Transgenic , Mitosis/physiology , Myeloid Cells/cytology , Myeloid Cells/physiology , Neurons/cytology , Precancerous Conditions/genetics , Precancerous Conditions/physiopathology , Primary Cell Culture , Proto-Oncogene Proteins/metabolism , Proto-Oncogene Proteins p21(ras) , Receptor for Advanced Glycation End Products , Receptors, Immunologic/metabolism , Signal Transduction/physiology , Toll-Like Receptor 4/metabolism , ras Proteins/metabolism
4.
Blood ; 118(2): 368-79, 2011 Jul 14.
Article in English | MEDLINE | ID: mdl-21586752

ABSTRACT

Both monoallelic and biallelic oncogenic NRAS mutations are identified in human leukemias, suggesting a dose-dependent role of oncogenic NRAS in leukemogenesis. Here, we use a hypomorphic oncogenic Nras allele and a normal oncogenic Nras allele (Nras G12D(hypo) and Nras G12D, respectively) to create a gene dose gradient ranging from 25% to 200% of endogenous Nras G12D/+. Mice expressing Nras G12D(hypo)/G12D(hypo) develop normally and are tumor-free, whereas early embryonic expression of Nras G12D/+ is lethal. Somatic expression of Nras G12D/G12D but not Nras G12D/+ leads to hyperactivation of ERK, excessive proliferation of myeloid progenitors, and consequently an acute myeloproliferative disease. Using a bone marrow transplant model, we previously showed that ∼ 95% of animals receiving Nras G12D/+ bone marrow cells develop chronic myelomonocytic leukemia (CMML), while ∼ 8% of recipients develop acute T-cell lymphoblastic leukemia/lymphoma [TALL] (TALL-het). Here we demonstrate that 100% of recipients transplanted with Nras G12D/G12D bone marrow cells develop TALL (TALL-homo). Although both TALL-het and -homo tumors acquire Notch1 mutations and are sensitive to a γ-secretase inhibitor, endogenous Nras G12D/+ signaling promotes TALL through distinct genetic mechanism(s) from Nras G12D/G12D. Our data indicate that the tumor transformation potential of endogenous oncogenic Nras is both dose- and cell type-dependent.


Subject(s)
Cell Transformation, Neoplastic/genetics , Gene Dosage/physiology , Genes, ras/genetics , Hematologic Neoplasms/genetics , Mutation , Amino Acid Substitution/physiology , Animals , Aspartic Acid/genetics , Bone Marrow Transplantation/physiology , Female , Genes, ras/physiology , Glutamic Acid/genetics , Hematologic Neoplasms/therapy , Humans , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic , Mutation/physiology , Oncogenes/genetics , Oncogenes/physiology , Organ Specificity/genetics
5.
Theranostics ; 13(14): 5075-5098, 2023.
Article in English | MEDLINE | ID: mdl-37771778

ABSTRACT

Background: Exploiting synthetic lethality (SL) relationships between protein pairs has emerged as an important avenue for the development of anti-cancer drugs. Nicotinamide phosphoribosyltransferase (NAMPT) is the rate-limiting enzyme of the NAD+ salvage pathway, having an SL relationship with nicotinic acid phosphoribosyltransferase (NAPRT), the key enzyme in the NAD+ Preiss-Handler pathway. NAMPT inhibitor holds clinical potential not only as a promising cancer treatment but also as a means of protection against chemotherapy-induced-peripheral-neuropathy (CIPN). However, as NAD+ is essential for normal cells, the clinical use of NAMPT inhibitors is challenging. This study aimed to identify a novel NAMPT inhibitor with enhanced selective cytotoxicity against NAPRT-deficient cancer cells as well as prominent efficacy in alleviating CIPN. Methods: We began by conducting drug derivatives screening in a panel of lung cancer cell lines to select an agent with the broadest therapeutic window between the NAPRT-negative and-positive cancer cell lines. Both in vitro and In vivo comparative analyses were conducted between A4276 and other NAMPT inhibitors to evaluate the NAPRT-negative cancer cell selectivity and the underlying distinct NAMPT inhibition mechanism of A4276. Patient-derived tumor transcriptomic data and protein levels in various cancer cell lines were analyzed to confirm the correlation between NAPRT depletion and epithelial-to-mesenchymal transition (EMT)-like features in various cancer types. Finally, the efficacy of A4276 for axonal protection and CIPN remedy was examined in vitro and in vivo. Results: The biomarker-driven phenotypic screening led to a discovery of A4276 with prominent selectivity against NAPRT-negative cancer cells compared with NAPRT-positive cancer cells and normal cells. The cytotoxic effect of A4276 on NAPRT-negative cells is achieved through its direct binding to NAMPT, inhibiting its enzymatic function at an optimal and balanced level allowing NAPRT-positive cells to survive through NAPRT-dependent NAD+ synthesis. NAPRT deficiency serves as a biomarker for the response to A4276 as well as an indicator of EMT-subtype cancer in various tumor types. Notably, A4276 protects axons from Wallerian degeneration more effectively than other NAMPT inhibitors by decreasing NMN-to-NAD+ ratio. Conclusion: This study demonstrates that A4276 selectively targets NAPRT-deficient EMT-subtype cancer cells and prevents chemotherapy-induced peripheral neuropathy, highlighting its potential as a promising anti-cancer agent for use in cancer monotherapy or combination therapy with conventional chemotherapeutics.

6.
Blood ; 116(26): 5991-6002, 2010 Dec 23.
Article in English | MEDLINE | ID: mdl-20921338

ABSTRACT

Oncogenic NRAS mutations are frequently identified in myeloid diseases involving monocyte lineage. However, its role in the genesis of these diseases remains elusive. We report a mouse bone marrow transplantation model harboring an oncogenic G12D mutation in the Nras locus. Approximately 95% of recipient mice develop a myeloproliferative disease resembling the myeloproliferative variant of chronic myelomonocytic leukemia (CMML), with a prolonged latency and acquisition of multiple genetic alterations, including uniparental disomy of oncogenic Nras allele. Based on single-cell profiling of phospho-proteins, a novel population of CMML cells is identified to display aberrant granulocyte-macrophage colony stimulating factor (GM-CSF) signaling in both the extracellular signal-regulated kinase (ERK) 1/2 and signal transducer and activator of transcription 5 (Stat5) pathways. This abnormal signaling is acquired during CMML development. Further study suggests that aberrant Ras/ERK signaling leads to expansion of granulocytic/monocytic precursors, which are highly responsive to GM-CSF. Hyperactivation of Stat5 in CMML cells is mainly through expansion of these precursors rather than up-regulation of surface expression of GM-CSF receptors. Our results provide insights into the aberrant cytokine signaling in oncogenic NRAS-associated myeloid diseases.


Subject(s)
Genes, ras/physiology , Granulocyte-Macrophage Colony-Stimulating Factor/metabolism , Granulocytes/metabolism , Leukemia, Myelomonocytic, Chronic/pathology , Monocytes/metabolism , Mutation/genetics , Animals , Blotting, Western , Bone Marrow Transplantation , Cell Proliferation , Extracellular Signal-Regulated MAP Kinases/metabolism , Granulocyte-Macrophage Colony-Stimulating Factor/genetics , Leukemia, Myelomonocytic, Chronic/genetics , Leukemia, Myelomonocytic, Chronic/metabolism , Mice , Mice, Inbred C57BL , RNA, Messenger/genetics , Receptors, Granulocyte-Macrophage Colony-Stimulating Factor/genetics , Receptors, Granulocyte-Macrophage Colony-Stimulating Factor/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Signal Transduction , Survival Rate
7.
Front Cell Dev Biol ; 9: 633661, 2021.
Article in English | MEDLINE | ID: mdl-33681212

ABSTRACT

Ras proteins control a complex intracellular signaling network. Gain-of-function mutations in RAS genes lead to RASopathy disorders in humans, including Noonan syndrome (NS). NS is the second most common syndromic cause of congenital heart disease. Although conditional expression of the NrasG12D/ + mutation in adult hematopoietic system is leukemogenic, its effects on embryonic development remain unclear. Here, we report that pan-embryonic expression of endogenous NrasG12D/ + by Mox2-Cre in mice caused embryonic lethality from embryonic day (E) 15.5 and developmental defects predominantly in the heart. At E13.5, NrasG12D/ + ; Mox2Cre/ + embryos displayed a moderate expansion of hematopoietic stem and progenitor cells without a significant impact on erythroid differentiation in the fetal liver. Importantly, the mutant embryos exhibited cardiac malformations resembling human congenital cardiac defects seen in NS patients, including ventricular septal defects, double outlet right ventricle, the hypertrabeculation/thin myocardium, and pulmonary valve stenosis. The mutant heart showed dysregulation of ERK, BMP, and Wnt pathways, crucial signaling pathways for cardiac development. Endothelial/endocardial-specific expression of NrasG12D/ + caused the cardiac morphological defects and embryonic lethality as observed in NrasG12D/ + ; Mox2Cre/ + mutants, but myocardial-specific expression of NrasG12D/ + did not. Thus, oncogenic NrasG12D mutation may not be compatible with embryonic survival.

8.
J Neurochem ; 104(5): 1260-70, 2008 Mar.
Article in English | MEDLINE | ID: mdl-17973978

ABSTRACT

The stargazer (stg) mutant mouse, having mutation in stargazin, the calcium channel gamma2 subunit, exhibited several neurological disorders including spontaneous absence seizure, cerebellar ataxia, and head tossing. To understand the molecular pathogenic mechanism of the absence seizure resulted from the loss of stargazin function, the thalamic proteomes between control mouse and stg mouse were compared. We identified 12 proteins expressed differentially (> 1.6-fold) by fluorescence two-dimensional difference gel electrophoresis and tandem mass spectrometry. Six of them are involved in basic metabolism including energy metabolism, three in stress response, two in axonal growth regulation, and one in the endoplasmic reticulum processing. All except mortalin showed decreased level of expression in stg mouse. Two stress-related proteins, mouse stress induced phosphoprotein 1 and peroxiredoxin 6 exhibited reduced levels of expression in stg mouse, while the level of another stress protein, mortalin was increased. Analysis of oxidative protein carbonylation in thalamic proteome of stg mouse showed higher level of carbonylated proteins in stg mouse than in control mouse. Interestingly, down-regulation of stress protein mouse stress induced phosphoprotein 1, metabolic enzyme isovaleryl-CoA dehydrogenase, and the two in neuronal axon growth, collapsin response mediator protein 2 and fascin homolog 1 coincides with the results of our previous study on gamma-butyrolactone-induced transient absence seizure. Our results suggest that the pathogenesis mechanism underlying absence seizure may involve the molecular events contributed by these proteins.


Subject(s)
Calcium Channels/physiology , Epilepsy, Absence/genetics , Epilepsy, Absence/metabolism , Mutant Proteins/physiology , Neurons/metabolism , Proteomics/methods , Animals , Calcium Channels/analysis , Calcium Channels/genetics , Electrophoresis, Gel, Two-Dimensional , Epilepsy, Absence/etiology , Gene Expression Regulation/physiology , Male , Mice , Mice, Inbred C57BL , Mice, Neurologic Mutants , Mutant Proteins/analysis , Mutant Proteins/genetics , Neurons/chemistry
9.
Nat Commun ; 9(1): 2165, 2018 06 04.
Article in English | MEDLINE | ID: mdl-29867097

ABSTRACT

Activation of T cell immune response is critical for the therapeutic efficacy of cancer immunotherapy. Current immunotherapies have shown remarkable clinical success against several cancers; however, significant responses remain restricted to a minority of patients. Here, we show a therapeutic strategy that combines enhancing the phagocytic activity of antigen-presenting cells with immunogenic cell death to trigger efficient antitumour immunity. Rho-kinase (ROCK) blockade increases cancer cell phagocytosis and induces antitumour immunity through enhancement of T cell priming by dendritic cells (DCs), leading to suppression of tumour growth in syngeneic tumour models. Combining ROCK blockade with immunogenic chemotherapy leads to increased DC maturation and synergistic CD8+ cytotoxic T cell priming and infiltration into tumours. This therapeutic strategy effectively suppresses tumour growth and improves overall survival in a genetic mouse mammary tumour virus/Neu tumour model. Collectively, these results suggest that boosting intrinsic cancer immunity using immunogenic killing and enhanced phagocytosis is a promising therapeutic strategy for cancer immunotherapy.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/pharmacology , Immunity/drug effects , Neoplasms, Experimental/drug therapy , Protein Kinase Inhibitors/pharmacology , rho-Associated Kinases/antagonists & inhibitors , Amides/administration & dosage , Amides/pharmacology , Animals , CD8-Positive T-Lymphocytes/drug effects , CD8-Positive T-Lymphocytes/immunology , Cell Death/drug effects , Cell Death/immunology , Cell Line, Tumor , Cells, Cultured , Cisplatin/administration & dosage , Dendritic Cells/drug effects , Dendritic Cells/immunology , Doxorubicin/administration & dosage , Humans , Immunity/immunology , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Transgenic , Neoplasms, Experimental/immunology , Neoplasms, Experimental/pathology , Protein Kinase Inhibitors/administration & dosage , Pyridines/administration & dosage , Pyridines/pharmacology , T-Lymphocytes, Cytotoxic/drug effects , T-Lymphocytes, Cytotoxic/immunology , rho-Associated Kinases/immunology , rho-Associated Kinases/metabolism
10.
Protein Eng Des Sel ; 17(4): 325-31, 2004 Apr.
Article in English | MEDLINE | ID: mdl-15126675

ABSTRACT

Unlike most globular proteins, the native form of serine protease inhibitors (serpins) is strained. Previous studies of human alpha(1)-antitrypsin, a prototype plasma serpin, revealed that various unfavorable interactions, such as overpacking of side chains, buried polar groups and cavities, are the structural basis of the strain. The local strain could be relieved by various stabilizing single amino acid substitutions, which appeared to remove these unfavorable interactions. To improve the stability of other clinically important serpin members, here we examined whether the rules found in alpha(1)-antitrypsin studies are applicable to other serpins. Amino acid substitutions were introduced at various positions in human alpha(1)-antichymotrypsin and human antithrombin III that were equivalent to the sites of stabilizing substitutions of alpha(1)-antitrypsin. Two-thirds of the substitutions increased thermostability in all serpins tested. Mutational analysis and structural examination suggest that serpins are suboptimally folded using common structural strategies at many sites, even though some structural details can vary in individual members. The results suggest that schemes discovered with alpha(1)-antitrypsin, an easily manipulative serpin, are a useful basis for engineering conformational characteristics of other clinically important serpins.


Subject(s)
Protein Engineering , Serine Proteinase Inhibitors/chemistry , alpha 1-Antitrypsin/chemistry , Amino Acid Substitution , Humans , Models, Molecular , Protein Conformation , Recombinant Proteins/chemistry
11.
J Clin Invest ; 122(10): 3692-704, 2012 Oct.
Article in English | MEDLINE | ID: mdl-22996659

ABSTRACT

Haploinsufficiency for GATA2 causes human immunodeficiency syndromes characterized by mycobacterial infection, myelodysplasia, lymphedema, or aplastic anemia that progress to myeloid leukemia. GATA2 encodes a master regulator of hematopoiesis that is also linked to endothelial biology. Though the disease-causing mutations commonly occur in the GATA-2 DNA binding domain, we identified a patient with mycobacterial infection and myelodysplasia who had an uncharacterized heterozygous deletion in a GATA2 cis-element consisting of an E-box and a GATA motif. Targeted deletion of the equivalent murine element to yield homozygous mutant mice revealed embryonic lethality later than occurred with global Gata2 knockout, hematopoietic stem/progenitor cell depletion, and impaired vascular integrity. Heterozygous mutant mice were viable, but embryos exhibited deficits in definitive, but not primitive, hematopoietic stem/progenitor activity and reduced expression of Gata2 and its target genes. Mechanistic analysis revealed disruption of the endothelial cell transcriptome and loss of vascular integrity. Thus, the composite element disrupted in a human immunodeficiency is essential for establishment of the murine hematopoietic stem/progenitor cell compartment in the fetal liver and for essential vascular processes.


Subject(s)
Blood Vessels/embryology , E-Box Elements , GATA2 Transcription Factor/deficiency , GATA2 Transcription Factor/physiology , Hematopoiesis/genetics , Immunologic Deficiency Syndromes/genetics , Myelodysplastic Syndromes/genetics , Regulatory Elements, Transcriptional , Animals , Base Sequence , Blood Vessels/pathology , Embryonic Development/genetics , Endothelium, Vascular/metabolism , GATA2 Transcription Factor/chemistry , GATA2 Transcription Factor/genetics , Genes, Lethal , Genetic Predisposition to Disease , Genotype , Hematopoiesis/physiology , Hematopoietic Stem Cells/pathology , Hemorrhage/embryology , Hemorrhage/genetics , Humans , Liver/cytology , Liver/embryology , Mice , Molecular Sequence Data , Mycobacterium Infections/etiology , Myelodysplastic Syndromes/complications , Sequence Deletion
12.
Cell Cycle ; 10(17): 2836-9, 2011 Sep 01.
Article in English | MEDLINE | ID: mdl-21857161

ABSTRACT

We previously showed that widespread expression of Nras G12D/G12D from its endogenous locus in mice leads to an acute myeloproliferative disease (MPD) with a complete penetrance, whereas bone marrow-specific expression of Nras G12D/G12D in recipient mice did not result in sustained MPD phenotypes but 100% penetrant acute T-cell lymphoblastic leukemia/lymphoma (TALL). Such a phenotypic switch also is seen in the case of endogenous oncogenic Kras. Two possibilities might account for this observation and they are not necessarily mutually exclusive. First, the MPD phenotypes in primary Nras G12D/G12D mice might be a transient phenomenon attributable to microenvironmental factors that do not necessarily imply the potential for long-term maintenance in a hematopoietic-cell autonomous manner. Second, it is likely that MPD phenotypes are maintained by genetically altered hematopoietic stem cells (HSCs). Nras G12D/G12D signaling might substantially alter HSC behaviors (e.g. induce their proliferative exhaustion) so that these HSCs no longer sustain MPD phenotypes to a lethal stage in recipient mice. Here, we show some preliminary results to support the second hypothesis. Our results suggest that different lineages of hematopoietic cells might have differential requirements of HSC activity and Nras G12D signaling during leukemogenesis.


Subject(s)
Hematopoietic Stem Cells/cytology , Myeloproliferative Disorders/pathology , Signal Transduction , ras Proteins/metabolism , Animals , Cell Transformation, Neoplastic/metabolism , Flow Cytometry , Hematopoietic Stem Cell Transplantation , Hematopoietic Stem Cells/pathology , Interferons/metabolism , Leukemia, Experimental , Mice , Mice, Transgenic , Phenotype , Precursor T-Cell Lymphoblastic Leukemia-Lymphoma/pathology , Splenomegaly/pathology
13.
Mol Cell Biochem ; 313(1-2): 91-102, 2008 Jun.
Article in English | MEDLINE | ID: mdl-18363038

ABSTRACT

Alloferon is a 13-amino acid peptide isolated from the bacteria-challenged larvae of the blow fly Calliphora vicina. The pharmaceutical value of the peptide has been well demonstrated by its capacity to stimulate NK cytotoxic activity and interferon (IFN) synthesis in animal and human models, as well as to enhance antiviral and antitumor activities in mice. Antiviral and the immunomodulatory effectiveness of alloferon have also been supported clinically proved in patients suffering with herpes simplex virus (HSV) and human papilloma virus (HPV) infections. To elucidate molecular response to alloferon treatment, we initially screened a model cell line in which alloferon enhanced IFN synthesis upon viral infection. Among the cell lines tested, Namalva was chosen for further proteomic analysis. Fluorescence difference gel electrophoresis (DIGE) revealed that the levels of a series of antioxidant proteins decreased after alloferon treatment, while at least three glycolytic enzymes and four heat-shock proteins were increased in their expression levels. Based on the result of our proteomic analysis, we speculated that alloferon may activate the NF-kappaB signaling pathway. IkappaB kinase (IKK) assay, Western blot analysis on IkappaBalpha and its phosphorylated form at Ser 32, and an NF-kappaB reporter assay verified our proteomics-driven hypothesis. Thus, our results suggest that alloferon potentiates immune cells by activating the NF-kappaB signaling pathway through regulation of redox potential. Since NF-kappaB activation is involved in IFN synthesis, our results provide further clues as to how the alloferon peptide may stimulate IFN synthesis.


Subject(s)
Antioxidants/metabolism , Down-Regulation/drug effects , I-kappa B Proteins/metabolism , NF-kappa B/metabolism , Peptides/pharmacology , Animals , Blotting, Western , Cell Line , Electrophoresis, Gel, Two-Dimensional , Humans , Interferons/biosynthesis , Mice , Models, Biological , NF-KappaB Inhibitor alpha , Oxidation-Reduction/drug effects , Phosphorylation/drug effects , Protein Processing, Post-Translational/drug effects , Proteome/analysis , Reproducibility of Results , Signal Transduction/drug effects , Virus Diseases/metabolism
14.
J Neurochem ; 102(3): 646-56, 2007 Aug.
Article in English | MEDLINE | ID: mdl-17419809

ABSTRACT

Absence seizure has been of interest because the symptom is related to sensory processing. However, the mechanism that causes the disease is not understood yet. To better understand the molecular mechanism related to the disease progress at protein level, we performed proteomic studies using the thalamus of mice for which absence seizure was induced by gamma-butyrolactone (GBL). Differential proteome expression between GBL-treated mice and control mice was examined by fluorescence 2D difference gel electrophoresis (DIGE) at three different time points (5, 10, and 30 min) after GBL-administration. We identified 16 proteins differentially expressed by >1.4-fold at any of the three time points. All proteins besides the serine protease inhibitor EIA were down-regulated in absence seizure-induced mice. The down-regulated proteins can be classified into five groups by their biological functions: cytoskeleton rearrangement, neuroprotection, neurotransmitter secretion, calcium binding, and metabolism. The maximum level of change was reached by 10 min after GBL-treatment, with the expression level returning back to the original at 30 min when mice were awakened from absence seizure thereby demonstrating the proteomic response is reversible. Our results suggest that absence seizures are associated with restricted functional sets of proteins, whose down-regulation may interfere with general function of neuronal cells.


Subject(s)
4-Butyrolactone/pharmacology , Epilepsy, Absence/metabolism , Nerve Tissue Proteins/metabolism , Neurons/metabolism , Thalamus/metabolism , Animals , Calcium-Binding Proteins/analysis , Calcium-Binding Proteins/metabolism , Convulsants/pharmacology , Cytoskeletal Proteins/analysis , Cytoskeletal Proteins/metabolism , Down-Regulation/drug effects , Down-Regulation/physiology , Electrophoresis, Gel, Two-Dimensional , Epilepsy, Absence/chemically induced , Epilepsy, Absence/physiopathology , Male , Mice , Nerve Tissue Proteins/analysis , Neurons/drug effects , Neurotransmitter Agents/biosynthesis , Neurotransmitter Agents/metabolism , Presynaptic Terminals/metabolism , Proteomics/methods , Thalamus/drug effects , Thalamus/physiopathology , Time Factors
15.
J Biol Chem ; 281(11): 7489-97, 2006 Mar 17.
Article in English | MEDLINE | ID: mdl-16407227

ABSTRACT

Pax6 is a transcriptional activator that contains two DNA binding domains and a potent transcription activation domain in the C terminus, which regulates organogenesis of the eye, nose, pancreas, and central nervous system. Homeodomain-interacting protein kinase 2 (HIPK2) interacts with transcription factors, including homeoproteins, and regulates activities of transcription factors. Here we show that HIPK2 phosphorylates the activation domain of Pax6, which augments Pax6 transactivation by enhancing its interaction with p300. Mass spectrometric analysis identified three Pax6 phosphorylation sites as threonines 281, 304, and 373. The substitutions of these threonines with alanines decreased Pax6 transactivation, whereas substitutions to glutamic acids increased transactivation in mimicry of phosphorylation. Furthermore, the knock-down of either endogenous or exogenous HIPK2 expression with HIPK2 shRNA markedly inhibited Pax6 phosphorylation and its transactivating function on proglucagon promoter in cultured cells. These results strongly indicate that HIPK2 is an upstream protein kinase for Pax6 and suggest that it modulates Pax6-mediated transcriptional regulation.


Subject(s)
Carrier Proteins/physiology , Eye Proteins/genetics , Eye Proteins/physiology , Homeodomain Proteins/genetics , Homeodomain Proteins/physiology , Paired Box Transcription Factors/genetics , Paired Box Transcription Factors/physiology , Protein Serine-Threonine Kinases/physiology , Repressor Proteins/genetics , Repressor Proteins/physiology , Alanine/chemistry , Animals , Binding Sites , Carrier Proteins/chemistry , Cell Line , Cell Line, Tumor , DNA Mutational Analysis , Eye Proteins/metabolism , Glucagon/chemistry , Glutamic Acid/chemistry , Green Fluorescent Proteins/metabolism , Homeodomain Proteins/metabolism , Humans , Immunohistochemistry , Luciferases/metabolism , Mass Spectrometry , Mice , Mutagenesis, Site-Directed , Mutation , PAX6 Transcription Factor , Paired Box Transcription Factors/metabolism , Phosphorylation , Promoter Regions, Genetic , Protein Binding , Protein Serine-Threonine Kinases/chemistry , Protein Structure, Tertiary , RNA, Small Interfering/chemistry , RNA, Small Interfering/metabolism , Repressor Proteins/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Spectrometry, Mass, Matrix-Assisted Laser Desorption-Ionization , Threonine/chemistry , Transcription, Genetic , Transcriptional Activation , Transfection , p300-CBP Transcription Factors/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL