Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 30
Filter
1.
Proc Natl Acad Sci U S A ; 119(27): e2123227119, 2022 07 05.
Article in English | MEDLINE | ID: mdl-35759659

ABSTRACT

DNA methyltransferase inhibitors (DNMTis) reexpress hypermethylated genes in cancers and leukemias and also activate endogenous retroviruses (ERVs), leading to interferon (IFN) signaling, in a process known as viral mimicry. In the present study we show that in the subset of acute myeloid leukemias (AMLs) with mutations in TP53, associated with poor prognosis, DNMTis, important drugs for treatment of AML, enable expression of ERVs and IFN and inflammasome signaling in a STING-dependent manner. We previously reported that in solid tumors poly ADP ribose polymerase inhibitors (PARPis) combined with DNMTis to induce an IFN/inflammasome response that is dependent on STING1 and is mechanistically linked to generation of a homologous recombination defect (HRD). We now show that STING1 activity is actually increased in TP53 mutant compared with wild-type (WT) TP53 AML. Moreover, in TP53 mutant AML, STING1-dependent IFN/inflammatory signaling is increased by DNMTi treatment, whereas in AMLs with WT TP53, DNMTis alone have no effect. While combining DNMTis with PARPis increases IFN/inflammatory gene expression in WT TP53 AML cells, signaling induced in TP53 mutant AML is still several-fold higher. Notably, induction of HRD in both TP53 mutant and WT AMLs follows the pattern of STING1-dependent IFN and inflammatory signaling that we have observed with drug treatments. These findings increase our understanding of the mechanisms that underlie DNMTi + PARPi treatment, and also DNMTi combinations with immune therapies, suggesting a personalized approach that statifies by TP53 status, for use of such therapies, including potential immune activation of STING1 in AML and other cancers.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols , DNA-Cytosine Methylases , Leukemia, Myeloid, Acute , Membrane Proteins , Poly(ADP-ribose) Polymerase Inhibitors , Tumor Suppressor Protein p53 , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , DNA-Cytosine Methylases/antagonists & inhibitors , Homologous Recombination/genetics , Humans , Inflammasomes/metabolism , Leukemia, Myeloid, Acute/drug therapy , Leukemia, Myeloid, Acute/genetics , Leukemia, Myeloid, Acute/immunology , Membrane Proteins/immunology , Mutation , Poly(ADP-ribose) Polymerase Inhibitors/pharmacology , Poly(ADP-ribose) Polymerase Inhibitors/therapeutic use , Signal Transduction , Tumor Suppressor Protein p53/genetics , Tumor Suppressor Protein p53/metabolism
2.
Development ; 147(1)2020 01 03.
Article in English | MEDLINE | ID: mdl-31806659

ABSTRACT

The GATA and PAX-SIX-EYA-DACH transcriptional networks (PSEDNs) are essential for proper development across taxa. Here, we demonstrate novel PSEDN roles in vivo in Drosophila hematopoiesis and in human erythropoiesis in vitro Using Drosophila genetics, we show that PSEDN members function with GATA to block lamellocyte differentiation and maintain the prohemocyte pool. Overexpression of human SIX1 stimulated erythroid differentiation of human erythroleukemia TF1 cells and primary hematopoietic stem-progenitor cells. Conversely, SIX1 knockout impaired erythropoiesis in both cell types. SIX1 stimulation of erythropoiesis required GATA1, as SIX1 overexpression failed to drive erythroid phenotypes and gene expression patterns in GATA1 knockout cells. SIX1 can associate with GATA1 and stimulate GATA1-mediated gene transcription, suggesting that SIX1-GATA1 physical interactions contribute to the observed functional interactions. In addition, both fly and human SIX proteins regulated GATA protein levels. Collectively, our findings demonstrate that SIX proteins enhance GATA function at multiple levels, and reveal evolutionarily conserved cooperation between the GATA and PSEDN networks that may regulate developmental processes beyond hematopoiesis.


Subject(s)
Drosophila Proteins/metabolism , Erythropoiesis/genetics , Gene Regulatory Networks , Hematopoiesis/genetics , Animals , Cell Line, Tumor , Drosophila , GATA Transcription Factors/metabolism , Gene Knockout Techniques , Homeodomain Proteins/metabolism , Humans , Nerve Tissue Proteins/metabolism , Nuclear Proteins/metabolism , Paired Box Transcription Factors/metabolism
3.
J Virol ; 93(16)2019 08 15.
Article in English | MEDLINE | ID: mdl-31142674

ABSTRACT

Viral proteins must intimately interact with the host cell machinery during virus replication. Here, we used the yeast Saccharomyces cerevisiae as a system to identify novel functional interactions between viral proteins and eukaryotic cells. Our work demonstrates that when the Middle East respiratory syndrome coronavirus (MERS-CoV) ORF4a accessory gene is expressed in yeast it causes a slow-growth phenotype. ORF4a has been characterized as an interferon antagonist in mammalian cells, and yet yeast lack an interferon system, suggesting further interactions between ORF4a and eukaryotic cells. Using the slow-growth phenotype as a reporter of ORF4a function, we utilized the yeast knockout library collection to perform a suppressor screen where we identified the YDL042C/SIR2 yeast gene as a suppressor of ORF4a function. The mammalian homologue of SIR2 is SIRT1, an NAD-dependent histone deacetylase. We found that when SIRT1 was inhibited by either chemical or genetic manipulation, there was reduced MERS-CoV replication, suggesting that SIRT1 is a proviral factor for MERS-CoV. Moreover, ORF4a inhibited SIRT1-mediated modulation of NF-κB signaling, demonstrating a functional link between ORF4a and SIRT1 in mammalian cells. Overall, the data presented here demonstrate the utility of yeast studies for identifying genetic interactions between viral proteins and eukaryotic cells. We also demonstrate for the first time that SIRT1 is a proviral factor for MERS-CoV replication and that ORF4a has a role in modulating its activity in cells.IMPORTANCE Middle East respiratory syndrome coronavirus (MERS-CoV) initially emerged in 2012 and has since been responsible for over 2,300 infections, with a case fatality ratio of approximately 35%. We have used the highly characterized model system of Saccharomyces cerevisiae to investigate novel functional interactions between viral proteins and eukaryotic cells that may provide new avenues for antiviral intervention. We identify a functional link between the MERS-CoV ORF4a proteins and the YDL042C/SIR2 yeast gene. The mammalian homologue of SIR2 is SIRT1, an NAD-dependent histone deacetylase. We demonstrate for the first time that SIRT1 is a proviral factor for MERS-CoV replication and that ORF4a has a role in modulating its activity in mammalian cells.


Subject(s)
Coronavirus Infections/metabolism , Coronavirus Infections/virology , Host-Pathogen Interactions , Middle East Respiratory Syndrome Coronavirus/physiology , Sirtuin 1/metabolism , Virus Replication , Cell Line , Cells, Cultured , Coronavirus Infections/genetics , Gene Silencing , Humans , Phenotype , Protein Binding , RNA Interference , Saccharomyces cerevisiae/genetics , Saccharomyces cerevisiae/growth & development , Saccharomyces cerevisiae/metabolism , Sirtuin 1/genetics , Viral Structural Proteins/genetics , Viral Structural Proteins/metabolism , Yeasts/genetics , Yeasts/metabolism
4.
Br J Haematol ; 168(4): 583-97, 2015 Feb.
Article in English | MEDLINE | ID: mdl-25312678

ABSTRACT

Expression levels of MIR144 and MIR451 increase during erythropoiesis, a pattern that is conserved from zebrafish to humans. As these two miRs are expressed from the same polycistronic transcript, we manipulated MIR144 and MIR451 in human erythroid cells individually and together to investigate their effects on human erythropoiesis. Inhibition of endogenous human MIR451 resulted in decreased numbers of erythroid (CD71(hi) CD235a(hi) CD34(-) ) cells, consistent with prior studies in zebrafish and mice. In addition, inhibition of MIR144 impaired human erythroid differentiation, unlike in zebrafish and mouse studies where the functional effect of MIR144 on erythropoiesis was minimal. In this study, we found RAB14 is a direct target of both MIR144 and MIR451. As MIR144 and MIR451 expression increased during human erythropoiesis, RAB14 protein expression decreased. Enforced RAB14 expression phenocopied the effect of MIR144 and/or MIR451 depletion, whereas shRNA-mediated RAB14 knockdown protected cells from MIR144 and/or MIR451 depletion-mediated erythropoietic inhibition. RAB14 knockdown increased the frequency and number of erythroid cells, increased ß-haemoglobin expression, and decreased CBFA2T3 expression during human erythropoiesis. In summary, we utilized MIR144 and MIR451 to identify RAB14 as a novel physiological inhibitor of human erythropoiesis.


Subject(s)
Erythropoiesis/physiology , MicroRNAs/physiology , rab GTP-Binding Proteins/physiology , Cell Line, Tumor , Erythroid Precursor Cells/cytology , Erythroid Precursor Cells/drug effects , Erythropoietin/pharmacology , Gene Expression Regulation , Genetic Vectors/genetics , Granulocyte-Macrophage Colony-Stimulating Factor/pharmacology , Humans , Lentivirus/genetics , Leukemia, Erythroblastic, Acute/pathology , MicroRNAs/antagonists & inhibitors , MicroRNAs/biosynthesis , MicroRNAs/genetics , RNA Interference , RNA, Small Interfering/pharmacology , Recombinant Proteins/pharmacology , Repressor Proteins/biosynthesis , Repressor Proteins/genetics , Transduction, Genetic , Tumor Suppressor Proteins/biosynthesis , Tumor Suppressor Proteins/genetics , rab GTP-Binding Proteins/antagonists & inhibitors , rab GTP-Binding Proteins/biosynthesis , rab GTP-Binding Proteins/genetics
5.
J Biol Chem ; 287(47): 39361-8, 2012 Nov 16.
Article in English | MEDLINE | ID: mdl-23027860

ABSTRACT

The Ca(2+)/calmodulin-stimulated protein phosphatase calcineurin is a critical component of Ca(2+) signaling cascades in eukaryotic cells. Myristoylation of the regulatory subunit of calcineurin (CNB) is conserved from yeast to humans. Here, we show that CNB myristoylation antagonizes phosphatase activation in yeast. Disruption of CNB myristoylation by mutation of the myristoylated glycine triggered constitutive expression of a calcineurin-dependent reporter gene and enhanced calcineurin-dependent phenotypes. Basal phosphatase activity was also increased in nmt1-181 yeast with reduced N-myristoyltransferase activity. Our findings are the first demonstration of a functional role for CNB myristoylation and reveal the importance of Nmt1 in modulating cellular calcineurin activation.


Subject(s)
Calcineurin/metabolism , Lipoylation/physiology , Protein Subunits/metabolism , Saccharomyces cerevisiae Proteins/metabolism , Saccharomyces cerevisiae/enzymology , Calcineurin/genetics , Enzyme Activation/physiology , Humans , Mutation , Protein Subunits/genetics , Saccharomyces cerevisiae/genetics , Saccharomyces cerevisiae Proteins/genetics
6.
iScience ; 26(2): 105963, 2023 Feb 17.
Article in English | MEDLINE | ID: mdl-36818289

ABSTRACT

Cardiac fibrosis is a hallmark in late-stage familial dilated cardiomyopathy (DCM) patients, although the underlying mechanism remains elusive. Cardiac exosomes (Exos) have been reported relating to fibrosis in ischemic cardiomyopathy. Thus, we investigated whether Exos secreted from the familial DCM cardiomyocytes could promote fibrogenesis. Using human iPSCs differentiated cardiomyocytes we isolated Exos of angiotensin II stimulation conditioned media from either DCM or control (CTL) cardiomyocytes. Of interest, cultured cardiac fibroblasts had increased fibrogenesis following exposure to DCM-Exos rather than CTL-Exos. Meanwhile, injecting DCM-Exos into mouse hearts enhanced cardiac fibrosis and impaired cardiac function. Mechanistically, we identified the upregulation of miRNA-218-5p in the DCM-Exos as a critical contributor to fibrogenesis. MiRNA-218-5p activated TGF-ß signaling via suppression of TNFAIP3, a master inflammation inhibitor. In conclusion, our results illustrate a profibrotic effect of cardiomyocytes-derived Exos that highlights an additional pathogenesis pathway for cardiac fibrosis in DCM.

7.
Transl Oncol ; 15(1): 101283, 2022 Jan.
Article in English | MEDLINE | ID: mdl-34808460

ABSTRACT

Signal transducer and activator of transcription 5 (STAT5) signaling plays a pathogenic role in both hematologic malignancies and solid tumors. In acute myeloid leukemia (AML), internal tandem duplications of fms-like tyrosine kinase 3 (FLT3-ITD) constitutively activate the FLT3 receptor, producing aberrant STAT5 signaling, driving cell survival and proliferation. Understanding STAT5 regulation may aid development of new treatment strategies in STAT5-activated cancers including FLT3-ITD AML. Poly ADP-ribose polymerase (PARP1), upregulated in FLT3-ITD AML, is primarily known as a DNA repair factor, but also regulates a diverse range of proteins through PARylation. Analysis of STAT5 protein sequence revealed putative PARylation sites and we demonstrate a novel PARP1 interaction and direct PARylation of STAT5 in FLT3-ITD AML. Moreover, PARP1 depletion and PARylation inhibition decreased STAT5 protein expression and activity via increased degradation, suggesting that PARP1 PARylation of STAT5 at least in part potentiates aberrant signaling by stabilizing STAT5 protein in FLT3-ITD AML. Importantly for translational significance, PARPis are cytotoxic in numerous STAT5-activated cancer cells and are synergistic with tyrosine kinase inhibitors (TKI) in both TKI-sensitive and TKI-resistant FLT3-ITD AML. Therefore, PARPi may have therapeutic benefit in STAT5-activated and therapy-resistant leukemias and solid tumors.

9.
Sci Rep ; 12(1): 14173, 2022 08 19.
Article in English | MEDLINE | ID: mdl-35986080

ABSTRACT

To gain insight into sialic acid biology and sialidase/neuraminidase (NEU) expression in mature human neutrophil (PMN)s, we studied NEU activity and expression in PMNs and the HL60 promyelocytic leukemic cell line, and changes that might occur in PMNs undergoing apoptosis and HL60 cells during their differentiation into PMN-like cells. Mature human PMNs contained NEU activity and expressed NEU2, but not NEU1, the NEU1 chaperone, protective protein/cathepsin A(PPCA), NEU3, and NEU4 proteins. In proapoptotic PMNs, NEU2 protein expression increased > 30.0-fold. Granulocyte colony-stimulating factor protected against NEU2 protein upregulation, PMN surface desialylation and apoptosis. In response to 3 distinct differentiating agents, dimethylformamide, dimethylsulfoxide, and retinoic acid, total NEU activity in differentiated HL60 (dHL60) cells was dramatically reduced compared to that of nondifferentiated cells. With differentiation, NEU1 protein levels decreased > 85%, PPCA and NEU2 proteins increased > 12.0-fold, and 3.0-fold, respectively, NEU3 remained unchanged, and NEU4 increased 1.7-fold by day 3, and then returned to baseline. In dHL60 cells, lectin blotting revealed decreased α2,3-linked and increased α2,6-linked sialylation. dHL60 cells displayed increased adhesion to and migration across human bone marrow-derived endothelium and increased bacterial phagocytosis. Therefore, myeloid apoptosis and differentiation provoke changes in NEU catalytic activity and protein expression, surface sialylation, and functional responsiveness.


Subject(s)
N-Acetylneuraminic Acid , Neuraminidase , Apoptosis , Cell Differentiation , Humans , N-Acetylneuraminic Acid/metabolism , Neuraminidase/metabolism , Neutrophils/metabolism
10.
iScience ; 25(8): 104656, 2022 Aug 19.
Article in English | MEDLINE | ID: mdl-35847554

ABSTRACT

Successful cell therapy requires cells to resist the hostile ischemic myocardium, be retained to continue secreting cardioprotective growth factors/exosomes, and resist immunological host responses. Clinically relevant stem/progenitor cells in a rodent model of acute myocardial infarction (MI) demonstrated that neonatal cardiac mesenchymal stromal cells (nMSCs) provide the most robust cardiac functional recovery. Transplanted nMSCs significantly increased the number of tissue reparative macrophages and regulatory T-cells and decreased monocyte-derived inflammatory macrophages and neutrophils in the host myocardium. mRNA microarray and single-cell analyses combined with targeted depletion studies established CD47 in nMSCs as a key molecule responsible for cell retention in the myocardium through an antiphagocytic mechanism regulated by miR34a-5p. Gain and loss-of-function studies demonstrated that miR34a-5p also regulated the production of exosomes and cardioprotective paracrine factors in the nMSC secretome. In conclusion, miR34a-5p and CD47 play an important role in determining the composition of nMSCs' secretome and immune evasion, respectively.

11.
PLoS One ; 16(11): e0258951, 2021.
Article in English | MEDLINE | ID: mdl-34762666

ABSTRACT

Radiation therapy plays a major role in the treatment of lung cancer patients. However, cancer cells develop resistance to radiation. Tumor radioresistance is a complex multifactorial mechanism which may be dependent on DNA damage and repair, hypoxic conditions inside tumor microenvironment, and the clonal selection of radioresistant cells from the heterogeneous tumor site, and it is a major cause of treatment failure in non-small cell lung cancer (NSCLC). In the present investigation caveolin-1 (CAV-1) has been observed to be highly expressed in radiation resistant A549 lung cancer cells. CRISPR-Cas9 knockout of CAV-1 reverted the cells to a radio sensitive phenotype. In addition, CAV-1 overexpression in parental A549 cells, led to radiation resistance. Further, gene expression analysis of A549 parental, radiation resistant, and caveolin-1 overexpressed cells, exhibited overexpression of DNA repair genes RAD51B, RAD18, SOX2 cancer stem cell marker, MMPs, mucins and cytoskeleton proteins in resistant and caveolin-1 over expressed A549 cells, as compared to parental A549 cells. Bioinformatic analysis shows upregulation of BRCA1, Nuclear Excision DNA repair, TGFB and JAK/STAT signaling pathways in radioresistant and caveolin-1 overexpressed cells, which may functionally mediate radiation resistance. Immunohistochemistry data demonstrated heterogeneous expression of CAV-1 gene in human lung cancer tissues, which was analogous to its enhanced expression in human lung cancer cell line model and mouse orthotopic xenograft lung cancer model. Also, TCGA PanCancer clinical studies have demonstrated amplification, deletions and missense mutation in CAV-1 gene in lung cancer patients, and that CAV-1 alteration has been linked to poor prognosis, and poor survival in lung cancer patients. Interestingly, we have also optimized ELISA assay to measure caveolin-1 protein in the blood of A549 radiation resistant human xenograft preclinical mouse model and discovered higher level of caveolin-1 (950 pg/ml) in tumor bearing animals treated with radiation, as compared to xenograft with radiosensitive lung cancer cells (450 pg/ml). Thus, we conclude that caveolin-1 is involved in radio-resistance and contributes to tumor aggression, and it has potential to be used as prognostic biomarker for radiation treatment response, and tumor progression for precision medicine in lung cancer patients.


Subject(s)
Biomarkers, Tumor/metabolism , Caveolin 1/metabolism , Lung Neoplasms/pathology , Radiation Tolerance , A549 Cells , Animals , Carcinoma, Squamous Cell/genetics , Carcinoma, Squamous Cell/pathology , Caveolin 1/genetics , DNA Repair/genetics , Gene Dosage , Gene Expression Profiling , Gene Expression Regulation, Neoplastic , Humans , Lung Neoplasms/genetics , Mice, Inbred BALB C , Mice, Nude , Microarray Analysis , Neoplasm Invasiveness , Neoplastic Stem Cells/metabolism , Neoplastic Stem Cells/pathology , Prognosis , Protein Interaction Maps/genetics , Up-Regulation/genetics , Xenograft Model Antitumor Assays
12.
Blood Adv ; 5(3): 711-724, 2021 02 09.
Article in English | MEDLINE | ID: mdl-33560385

ABSTRACT

Artemisinins are active against human leukemia cell lines and have low clinical toxicity in worldwide use as antimalarials. Because multiagent combination regimens are necessary to cure fully evolved leukemias, we sought to leverage our previous finding that artemisinin analogs synergize with kinase inhibitors, including sorafenib (SOR), by identifying additional synergistic antileukemic drugs with low toxicity. Screening of a targeted antineoplastic drug library revealed that B-cell lymphoma 2 (BCL2) inhibitors synergize with artemisinins, and validation assays confirmed that the selective BCL2 inhibitor, venetoclax (VEN), synergized with artemisinin analogs to inhibit growth and induce apoptotic cell death of multiple acute leukemia cell lines in vitro. An oral 3-drug "SAV" regimen (SOR plus the potent artemisinin-derived trioxane diphenylphosphate 838 dimeric analog [ART838] plus VEN) killed leukemia cell lines and primary cells in vitro. Leukemia cells cultured in ART838 had decreased induced myeloid leukemia cell differentiation protein (MCL1) levels and increased levels of DNA damage-inducible transcript 3 (DDIT3; GADD153) messenger RNA and its encoded CCATT/enhancer-binding protein homologous protein (CHOP), a key component of the integrated stress response. Thus, synergy of the SAV combination may involve combined targeting of MCL1 and BCL2 via discrete, tolerable mechanisms, and cellular levels of MCL1 and DDIT3/CHOP may serve as biomarkers for action of artemisinins and SAV. Finally, SAV treatment was tolerable and resulted in deep responses with extended survival in 2 acute myeloid leukemia (AML) cell line xenograft models, both harboring a mixed lineage leukemia gene rearrangement and an FMS-like receptor tyrosine kinase-3 internal tandem duplication, and inhibited growth in 2 AML primagraft models.


Subject(s)
Artemisinins , Bridged Bicyclo Compounds, Heterocyclic , Cell Line, Tumor , Drug Synergism , Humans , Sorafenib , Sulfonamides
13.
J Neurosci ; 29(40): 12702-10, 2009 Oct 07.
Article in English | MEDLINE | ID: mdl-19812345

ABSTRACT

cAMP can stimulate the transcription of many activity-dependent genes via activation of the transcription factor, cAMP response element-binding protein (CREB). However, in mouse cortical neuron cultures, prior to synaptogenesis, neither cAMP nor dopamine, which acts via cAMP, stimulated CREB-dependent gene transcription when NR2B-containing NMDA receptors (NMDARs) were blocked. Stimulation of transcription by cAMP was potentiated by inhibitors of excitatory amino acid uptake, suggesting a role for extracellular glutamate or aspartate in cAMP-induced transcription. Aspartate was identified as the extracellular messenger: enzymatic scavenging of l-aspartate, but not glutamate, blocked stimulation of CREB-dependent gene transcription by cAMP; moreover, cAMP induced aspartate but not glutamate release. Together, these results suggest that cAMP acts via an autocrine or paracrine pathway to release aspartate, which activates NR2B-containing NMDARs, leading to Ca(2+) entry and activation of transcription. This cAMP/aspartate/NMDAR signaling pathway may mediate the effects of transmitters such as dopamine on axon growth and synaptogenesis in developing neurons or on synaptic plasticity in mature neural networks.


Subject(s)
Aspartic Acid/metabolism , Autocrine Communication/physiology , Cyclic AMP/metabolism , Dopamine/metabolism , Neurons/physiology , Receptors, N-Methyl-D-Aspartate/metabolism , Transcription, Genetic/physiology , Amino Acid Sequence , Animals , Brain-Derived Neurotrophic Factor/genetics , Calcium/metabolism , Calcium Channels/metabolism , Cell Culture Techniques , Cerebral Cortex/embryology , Cerebral Cortex/physiology , Colforsin/metabolism , Cyclic AMP Response Element-Binding Protein/metabolism , Guanine Nucleotide Exchange Factors/genetics , Mice , Molecular Sequence Data , Neuronal Plasticity/physiology , Phosphorylation , Proto-Oncogene Proteins c-fos/genetics , Transcriptional Activation/physiology
14.
Biochem Biophys Res Commun ; 397(2): 152-6, 2010 Jun 25.
Article in English | MEDLINE | ID: mdl-20493822

ABSTRACT

The Ca(2+)/cAMP response element binding protein CREB mediates transcription of genes essential for the development and function of the central nervous system. Here we investigated the ability of caffeine to stimulate CREB-dependent gene transcription in primary cultures of developing mouse cortical neurons. Using the CREB-dependent reporter gene CRE-luciferase we show that stimulation of CREB activity by caffeine exhibits a bell-shaped dose-response curve. Maximal stimulation occurred at 10mM caffeine, which is known to release Ca(2+) from ryanodine sensitive internal stores. In our immature neuronal cultures, 10mM caffeine was more effective at stimulating CREB activity than depolarization with high extracellular KCl (50mM). Quantitative real-time PCR analysis demonstrated that transcripts derived from endogenous CREB target genes, such as the gene encoding brain-derived neurotrophic factor BDNF, are increased following caffeine treatment. The dose-response curves of CREB target genes to caffeine exhibited gene-specificity, highlighting the importance of promoter structure in shaping genomic responses to Ca(2+) signaling. In the presence of a weak depolarizing stimulus (10mM KCl), concentrations of caffeine relevant for premature infants undergoing caffeine treatment increased CRE-luciferase activity and Bdnf transcript levels. The ability of caffeine to enhance activity-dependent Bdnf expression may contribute to the neurological benefit observed in infants receiving caffeine treatment.


Subject(s)
Brain-Derived Neurotrophic Factor/genetics , Caffeine/pharmacology , Cerebral Cortex/drug effects , Cerebral Cortex/growth & development , Cyclic AMP Response Element-Binding Protein/metabolism , Gene Expression Regulation, Developmental/drug effects , Animals , Cells, Cultured , Cerebral Cortex/abnormalities , Genes, Reporter , Mice , Neurons/drug effects , Neurons/metabolism , Transcription, Genetic/drug effects
15.
Sci Rep ; 10(1): 21159, 2020 12 03.
Article in English | MEDLINE | ID: mdl-33273545

ABSTRACT

Inactivation of Ataxia-telangiectasia mutated (ATM) gene results in an increased risk to develop cancer. We show that ATM deficiency in diffuse large B-cell lymphoma (DLBCL) significantly induce mitochondrial deacetylase sirtuin-3 (SIRT3) activity, disrupted mitochondrial structure, decreased mitochondrial respiration, and compromised TCA flux compared with DLBCL cells expressing wild type (WT)-ATM. This corresponded to enrichment of glutamate receptor and glutamine pathways in ATM deficient background compared to WT-ATM DLBCL cells. ATM-/- DLBCL cells have decreased apoptosis in contrast to radiosensitive non-cancerous A-T cells. In vivo studies using gain and loss of SIRT3 expression showed that SIRT3 promotes growth of ATM CRISPR knockout DLBCL xenografts compared to wild-type ATM control xenografts. Importantly, screening of DLBCL patient samples identified SIRT3 as a putative therapeutic target, and validated an inverse relationship between ATM and SIRT3 expression. Our data predicts SIRT3 as an important therapeutic target for DLBCL patients with ATM null phenotype.


Subject(s)
Ataxia Telangiectasia Mutated Proteins/deficiency , Ataxia Telangiectasia Mutated Proteins/genetics , Lymphoma, Large B-Cell, Diffuse/genetics , Sirtuin 3/metabolism , Ataxia Telangiectasia Mutated Proteins/antagonists & inhibitors , Cell Line, Tumor , Citric Acid Cycle , Forkhead Box Protein O3/metabolism , Humans , Mitochondria/metabolism , Mitochondria/ultrastructure , Models, Biological , Oxygen Consumption , Sirtuin 1/metabolism
16.
Glia ; 57(8): 828-34, 2009 Jun.
Article in English | MEDLINE | ID: mdl-19031446

ABSTRACT

Astrocytes participate in signaling via Ca(2+) transients that spread from cell to cell across a multicellular syncytium. The effect, if any, of these Ca(2+) waves on the transcription of Ca(2+)/cAMP-regulatory element binding protein (CREB)-dependent genes is not known. We report here that, unlike neurons, increasing intracellular Ca(2+) in cultured mouse cortical astrocytes failed to activate CREB-dependent transcription, even though CREB was phosphorylated at serine 133. In contrast, both CREB phosphorylation and CREB-dependent transcription were robustly stimulated by increasing cAMP. The failure of Ca(2+)-activated transcription in astrocytes was correlated with the absence of CaMKIV, a Ca(2+)-dependent protein kinase required for Ca(2+)-stimulated gene transcription in neurons. The inability of Ca(2+) to signal via CaMKIV may insulate CREB-dependent gene transcription in astrocytes from activation by Ca(2+) waves.


Subject(s)
Astrocytes/metabolism , CREB-Binding Protein/metabolism , Calcium/metabolism , Adenosine Triphosphate/pharmacology , Animals , Animals, Newborn , Astrocytes/drug effects , CREB-Binding Protein/genetics , Calcium-Calmodulin-Dependent Protein Kinase Type 4/genetics , Calcium-Calmodulin-Dependent Protein Kinase Type 4/metabolism , Cells, Cultured , Cerebral Cortex/cytology , Chelating Agents/pharmacology , Cyclic AMP/pharmacology , Egtazic Acid/analogs & derivatives , Egtazic Acid/pharmacology , Enzyme Inhibitors/pharmacology , Ionomycin/pharmacology , Ionophores/pharmacology , Mice , Mice, Inbred C57BL , Okadaic Acid/pharmacology , Transfection/methods
17.
Wiley Interdiscip Rev RNA ; 10(5): e1537, 2019 09.
Article in English | MEDLINE | ID: mdl-31007002

ABSTRACT

Hematopoiesis is a highly-regulated development process orchestrated by lineage-specific transcription factors that direct the generation of all mature blood cells types, including red blood cells, megakaryocytes, granulocytes, monocytes, and lymphocytes. Under homeostatic conditions, the hematopoietic system of the typical adult generates over 1011 blood cells daily throughout life. In addition, hematopoiesis must be responsive to acute challenges due to blood loss or infection. MicroRNAs (miRs) cooperate with transcription factors to regulate all aspects of hematopoiesis, including stem cell maintenance, lineage selection, cell expansion, and terminal differentiation. Distinct miR expression patterns are associated with specific hematopoietic lineages and stages of differentiation and functional analyses have elucidated essential roles for miRs in regulating cell transitions, lineage selection, maturation, and function. MiRs function as downstream effectors of hematopoietic transcription factors and as upstream regulators to control transcription factor levels. Multiple miRs have been shown to play essential roles. Regulatory networks comprised of differentially expressed lineage-specific miRs and hematopoietic transcription factors are involved in controlling the quiescence and self-renewal of hematopoietic stem cells as well as proliferation and differentiation of lineage-specific progenitor cells during erythropoiesis, myelopoiesis, and lymphopoiesis. This review focuses on hematopoietic miRs that function as upstream regulators of central hematopoietic transcription factors required for normal hematopoiesis. This article is categorized under: RNA in Disease and Development > RNA in Development Regulatory RNAs/RNAi/Riboswitches > Regulatory RNAs.


Subject(s)
Hematopoiesis , MicroRNAs/metabolism , Transcription Factors/metabolism , Animals , Humans , MicroRNAs/genetics
18.
Adv Cancer Res ; 141: 1-42, 2019.
Article in English | MEDLINE | ID: mdl-30691681

ABSTRACT

The PAX-SIX-EYA-DACH network (PSEDN) is a central developmental transcriptional regulatory network from Drosophila to humans. The PSEDN is comprised of four conserved protein families; including paired box (PAX), sine oculis (SIX), eyes absent (EYA), and dachshund (DACH). Aberrant expression of PSEDN members, particularly SIX1, has been observed in multiple human cancers, where SIX1 expression correlates with increased aggressiveness and poor prognosis. In conjunction with its transcriptional activator EYA, the SIX1 transcription factor increases cancer stem cell (CSC) numbers and induces epithelial-mesenchymal transition (EMT). SIX1 promotes multiple hallmarks and enabling characteristics of cancer via regulation of cell proliferation, senescence, apoptosis, genome stability, and energy metabolism. SIX1 also influences the tumor microenvironment, enhancing recruitment of tumor-associated macrophages and stimulating angiogenesis, to promote tumor development and progression. EYA proteins are multifunctional, possessing a transcriptional activation domain and tyrosine phosphatase activity, that each contributes to cancer stem cell properties. DACH proteins function as tumor suppressors in solid cancers, opposing the actions of SIX-EYA and reducing CSC prevalence. Multiple mechanisms can lead to increased SIX1 expression, including loss of SIX1-targeting tumor suppressor microRNAs (miRs), whose expression correlates inversely with SIX1 expression in cancer patient samples. In this review, we discuss the major mechanisms by which SIX1 confers CSC and EMT features and other important cancer cell characteristics. The roles of EYA and DACH in CSCs and cancer progression are briefly highlighted. Finally, we summarize the clinical significance of SIX1 in cancer to emphasize the potential therapeutic benefits of effective strategies to disrupt PSEDN protein interactions and functions.


Subject(s)
Eye Proteins/metabolism , Homeodomain Proteins/metabolism , Intracellular Signaling Peptides and Proteins/metabolism , Neoplasms/pathology , Neoplastic Stem Cells/pathology , Nuclear Proteins/metabolism , Paired Box Transcription Factors/metabolism , Protein Tyrosine Phosphatases/metabolism , Transcription Factors/metabolism , Animals , Humans , Neoplasms/metabolism , Neoplastic Stem Cells/metabolism
19.
Dis Model Mech ; 12(10)2019 10 16.
Article in English | MEDLINE | ID: mdl-31519738

ABSTRACT

Bi-allelic GBA1 mutations cause Gaucher's disease (GD), the most common lysosomal storage disorder. Neuronopathic manifestations in GD include neurodegeneration, which can be severe and rapidly progressive. GBA1 mutations are also the most frequent genetic risk factors for Parkinson's disease. Dysfunction of the autophagy-lysosomal pathway represents a key pathogenic event in GBA1-associated neurodegeneration. Using an induced pluripotent stem cell (iPSC) model of GD, we previously demonstrated that lysosomal alterations in GD neurons are linked to dysfunction of the transcription factor EB (TFEB). TFEB controls the coordinated expression of autophagy and lysosomal genes and is negatively regulated by the mammalian target of rapamycin complex 1 (mTORC1). To further investigate the mechanism of autophagy-lysosomal pathway dysfunction in neuronopathic GD, we examined mTORC1 kinase activity in GD iPSC neuronal progenitors and differentiated neurons. We found that mTORC1 is hyperactive in GD cells as evidenced by increased phosphorylation of its downstream protein substrates. We also found that pharmacological inhibition of glucosylceramide synthase enzyme reversed mTORC1 hyperactivation, suggesting that increased mTORC1 activity is mediated by the abnormal accumulation of glycosphingolipids in the mutant cells. Treatment with the mTOR inhibitor Torin1 upregulated lysosomal biogenesis and enhanced autophagic clearance in GD neurons, confirming that lysosomal dysfunction is mediated by mTOR hyperactivation. Further analysis demonstrated that increased TFEB phosphorylation by mTORC1 results in decreased TFEB stability in GD cells. Our study uncovers a new mechanism contributing to autophagy-lysosomal pathway dysfunction in GD, and identifies the mTOR complex as a potential therapeutic target for treatment of GBA1-associated neurodegeneration.


Subject(s)
Gaucher Disease/pathology , Induced Pluripotent Stem Cells/pathology , Lysosomes/pathology , Mechanistic Target of Rapamycin Complex 1/metabolism , Neurons/metabolism , Autophagy/drug effects , Basic Helix-Loop-Helix Leucine Zipper Transcription Factors/metabolism , Biomarkers/metabolism , Cell Line , Cell Nucleus/drug effects , Cell Nucleus/metabolism , Green Fluorescent Proteins/metabolism , Humans , Induced Pluripotent Stem Cells/drug effects , Induced Pluripotent Stem Cells/metabolism , Lipids/chemistry , Lysosomes/drug effects , Lysosomes/metabolism , Naphthyridines/pharmacology , Neural Stem Cells/drug effects , Neural Stem Cells/metabolism , Protein Stability/drug effects , Up-Regulation/drug effects
20.
Tissue Eng Part A ; 24(23-24): 1715-1732, 2018 12.
Article in English | MEDLINE | ID: mdl-29845891

ABSTRACT

Stem cell cultures within perfusion bioreactors, while efficient in obtaining cell numbers, often lack the similarity to native tissues and consequently cell phenotype. We develop a three-dimensional (3D)-printed fluidic chamber for dynamic stem cell culture, with emphasis on control over flow and substrate curvature in a 3D environment, two physiologic features of native tissues. The chamber geometry, consisting of an array of vertical cylindrical pillars, facilitates actin-mediated localization of human mesenchymal stem cells (hMSCs) within ∼200 µm distance from the pillars, enabling spatial patterning of hMSCs and endothelial cells in cocultures and subsequent modulation of calcium signaling between these two essential cell types in the bone marrow microenvironment. Flow-enhanced osteogenic differentiation of hMSCs in growth media imposes spatial variations of alkaline phosphatase expression, which positively correlates with local shear stress. Proliferation of hMSCs is maintained within the chamber, exceeding the cell expansion in conventional static culture. The capability to manipulate cell spatial patterning, differentiation, and 3D tissue formation through geometry and flow demonstrates the culture chamber's relevant chemomechanical cues in stem cell microenvironments, thus providing an easy-to-implement tool to study interactions among substrate curvature, shear stress, and intracellular actin machinery in the tissue-engineered construct.


Subject(s)
Bioreactors , Cell Differentiation , Cell Proliferation , Lab-On-A-Chip Devices , Mesenchymal Stem Cells/metabolism , Coculture Techniques/instrumentation , Coculture Techniques/methods , Humans , Mesenchymal Stem Cells/cytology , Tissue Engineering/instrumentation , Tissue Engineering/methods
SELECTION OF CITATIONS
SEARCH DETAIL