Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 32
Filter
1.
Behav Genet ; 50(2): 73-83, 2020 03.
Article in English | MEDLINE | ID: mdl-31820295

ABSTRACT

The Louisville Twin Study (LTS) began in 1958 and became a premier longitudinal twin study of cognitive development. The LTS continuously collected data from twins through 2000 after which the study closed indefinitely due to lack of funding. Now that the majority of the sample is age 40 or older (61.36%, N = 1770), the LTS childhood data can be linked to midlife cognitive functioning, among other physical, biological, social, and psychiatric outcomes. We report results from two pilot studies in anticipation of beginning the midlife phase of the LTS. The first pilot study was a participant tracking study, in which we showed that approximately 90% of the Louisville families randomly sampled (N = 203) for the study could be found. The second pilot study consisted of 40 in-person interviews in which twins completed cognitive, memory, biometric, and functional ability measures. The main purpose of the second study was to correlate midlife measures of cognitive functioning to a measure of biological age, which is an alternative index to chronological age that quantifies age as a function of the breakdown of structural and functional physiological systems, and then to relate both of these measures to twins' cognitive developmental trajectories. Midlife IQ was uncorrelated with biological age (- .01) while better scores on episodic memory more strongly correlated with lower biological age (- .19 to - .31). As expected, midlife IQ positively correlated with IQ measures collected throughout childhood and adolescence. Additionally, positive linear rates of change in FSIQ scores in childhood significantly correlated with biological age (- .68), physical functioning (.71), and functional ability (- .55), suggesting that cognitive development predicts lower biological age, better physical functioning, and better functional ability. In sum, the Louisville twins can be relocated to investigate whether and how early and midlife cognitive and physical health factors contribute to cognitive aging.


Subject(s)
Aging/physiology , Cognitive Aging/physiology , Cognitive Aging/psychology , Cognition/physiology , Female , Humans , Longitudinal Studies , Male , Middle Aged , Pilot Projects , Twins/genetics , Twins/psychology , Twins, Dizygotic/genetics , Twins, Dizygotic/psychology , Twins, Monozygotic/genetics , Twins, Monozygotic/psychology
2.
Proc Natl Acad Sci U S A ; 115(18): E4245-E4254, 2018 05 01.
Article in English | MEDLINE | ID: mdl-29654145

ABSTRACT

The discovery that endoplasmic reticulum (ER) luminal chaperones such as GRP78/BiP can escape to the cell surface upon ER stress where they regulate cell signaling, proliferation, apoptosis, and immunity represents a paradigm shift. Toward deciphering the mechanisms, we report here that, upon ER stress, IRE1α binds to and triggers tyrosine kinase SRC activation, leading to ASAP1 phosphorylation and Golgi accumulation of ASAP1 and Arf1-GTP, resulting in KDEL receptor dispersion from the Golgi and suppression of retrograde transport. At the cell surface, GRP78 binds to and acts in concert with a glycosylphosphatidylinositol-anchored protein, CD109, in blocking TGF-ß signaling by promoting the routing of the TGF-ß receptor to the caveolae, thereby disrupting its binding to and activation of Smad2. Collectively, we uncover a SRC-mediated signaling cascade that leads to the relocalization of ER chaperones to the cell surface and a mechanism whereby GRP78 counteracts the tumor-suppressor effect of TGF-ß.


Subject(s)
Antigens, CD/metabolism , Endoplasmic Reticulum Stress/physiology , Heat-Shock Proteins/metabolism , Neoplasm Proteins/metabolism , Signal Transduction/physiology , Transforming Growth Factor beta/metabolism , src-Family Kinases/metabolism , ADP-Ribosylation Factor 1/genetics , ADP-Ribosylation Factor 1/metabolism , Adaptor Proteins, Signal Transducing/genetics , Adaptor Proteins, Signal Transducing/metabolism , Antigens, CD/genetics , Endoplasmic Reticulum Chaperone BiP , Enzyme Activation/physiology , GPI-Linked Proteins/genetics , GPI-Linked Proteins/metabolism , HEK293 Cells , HeLa Cells , Heat-Shock Proteins/genetics , Humans , MCF-7 Cells , Neoplasm Proteins/genetics , Protein Transport/physiology , Smad2 Protein/genetics , Smad2 Protein/metabolism , Transforming Growth Factor beta/genetics
3.
Cancer Lett ; 400: 161-174, 2017 08 01.
Article in English | MEDLINE | ID: mdl-28450161

ABSTRACT

The anticancer agent 3-bromopyruvate (3-BP) is viewed as a glycolytic inhibitor that preferentially kills glycolytic cancer cells through energy depletion. However, its cytotoxic activity is dependent on cellular drug import through transmembrane monocarboxylate transporter 1 (MCT-1), which restricts its anticancer potential to MCT-1-positive tumor cells. We created and characterized an MCT-1-independent analog of 3-BP, called NEO218. NEO218 was synthesized by covalently conjugating 3-BP to perillyl alcohol (POH), a natural monoterpene. The responses of various tumor cell lines to treatment with either compound were characterized in the presence or absence of supplemental pyruvate or antioxidants N-acetyl-cysteine (NAC) and glutathione (GSH). Drug effects on glyceraldehyde 3-phosphate dehydrogenase (GAPDH) enzyme activity were investigated by mass spectrometric analysis. The development of 3-BP resistance was investigated in MCT-1-positive HCT116 colon carcinoma cells in vitro. Our results show that NEO218: (i) pyruvylated GAPDH on all 4 of its cysteine residues and shut down enzymatic activity; (ii) severely lowered cellular ATP content below life-sustaining levels, and (iii) triggered rapid necrosis. Intriguingly, supplemental antioxidants effectively prevented cytotoxic activity of NEO218 as well as 3-BP, but supplemental pyruvate powerfully protected cells only from 3-BP, not from NEO218. Unlike 3-BP, NEO218 exerted its potent cytotoxic activity irrespective of cellular MCT-1 status. Treatment of HCT116 cells with 3-BP resulted in prompt development of resistance, based on the emergence of MCT-1-negative cells. This was not the case with NEO218, and highly 3-BP-resistant cells remained exquisitely sensitive to NEO218. Thus, our study identifies a mechanism by which tumor cells develop rapid resistance to 3-BP, and presents NEO218 as a superior agent not subject to this cellular defense. Furthermore, our results offer alternative interpretations of previously published models on the role of supplemental antioxidants: Rather than quenching reactive oxygen species (ROS), supplemental NAC or GSH directly interact with 3-BP, thereby neutralizing the drug's cytotoxic potential before it can trigger ROS production. Altogether, our study introduces new aspects of the cytotoxic mechanism of 3-BP, and characterizes NEO218 as an analog able to overcome a key cellular defense mechanism towards this drug.


Subject(s)
Antineoplastic Agents/pharmacology , Drug Resistance, Neoplasm , Monocarboxylic Acid Transporters/metabolism , Monoterpenes/pharmacology , Neoplasms/drug therapy , Pyruvates/pharmacology , Symporters/metabolism , Adenosine Triphosphate/metabolism , Alkylation , Antioxidants/pharmacology , Dose-Response Relationship, Drug , Glyceraldehyde-3-Phosphate Dehydrogenases , Glycolysis/drug effects , HCT116 Cells , Humans , MCF-7 Cells , Monocarboxylic Acid Transporters/genetics , Necrosis , Neoplasms/genetics , Neoplasms/metabolism , Neoplasms/pathology , RNA Interference , Signal Transduction/drug effects , Symporters/genetics , Transfection
4.
Immunity ; 43(2): 251-63, 2015 Aug 18.
Article in English | MEDLINE | ID: mdl-26275994

ABSTRACT

Regulatory T (Treg) cells are essential for maintenance of immune homeostasis. Here we found that hydrogen sulfide (H2S) was required for Foxp3(+) Treg cell differentiation and function and that H2S deficiency led to systemic autoimmune disease. H2S maintained expression of methylcytosine dioxygenases Tet1 and Tet2 by sulfhydrating nuclear transcription factor Y subunit beta (NFYB) to facilitate its binding to Tet1 and Tet2 promoters. Transforming growth factor-ß (TGF-ß)-activated Smad3 and interleukin-2 (IL-2)-activated Stat5 facilitated Tet1 and Tet2 binding to Foxp3. Tet1 and Tet2 catalyzed conversion of 5-methylcytosine (5mC) to 5-hydroxymethylcytosine (5hmC) in Foxp3 to establish a Treg-cell-specific hypomethylation pattern and stable Foxp3 expression. Consequently, Tet1 and Tet2 deletion led to Foxp3 hypermethylation, impaired Treg cell differentiation and function, and autoimmune disease. Thus, H2S promotes Tet1 and Tet2 expression, which are recruited to Foxp3 by TGF-ß and IL-2 signaling to maintain Foxp3 demethylation and Treg-cell-associated immune homeostasis.


Subject(s)
Colitis/immunology , DNA-Binding Proteins/metabolism , Forkhead Transcription Factors/metabolism , Hydrogen Sulfide/metabolism , Proto-Oncogene Proteins/metabolism , T-Lymphocytes, Regulatory/immunology , Adoptive Transfer , Animals , CCAAT-Binding Factor/metabolism , Cell Differentiation/genetics , Colitis/genetics , DNA Methylation/genetics , DNA-Binding Proteins/genetics , Dioxygenases , Forkhead Transcription Factors/genetics , Homeostasis/genetics , Homeostasis/immunology , Humans , Interleukin-2/immunology , Mice , Mice, Inbred C57BL , Mice, Knockout , Proto-Oncogene Proteins/genetics , STAT5 Transcription Factor/metabolism , Smad3 Protein/metabolism , T-Lymphocytes, Regulatory/transplantation , Transforming Growth Factor beta/immunology
5.
PLoS Pathog ; 11(7): e1005060, 2015 Jul.
Article in English | MEDLINE | ID: mdl-26221961

ABSTRACT

Activation of pattern recognition receptors and proper regulation of downstream signaling are crucial for host innate immune response. Upon infection, the NF-κB and interferon regulatory factors (IRF) are often simultaneously activated to defeat invading pathogens. Mechanisms concerning differential activation of NF-κB and IRF are not well understood. Here we report that a MAVS variant inhibits interferon (IFN) induction, while enabling NF-κB activation. Employing herpesviral proteins that selectively activate NF-κB signaling, we discovered that a MAVS variant of ~50 kDa, thus designated MAVS50, was produced from internal translation initiation. MAVS50 preferentially interacts with TRAF2 and TRAF6, and activates NF-κB. By contrast, MAVS50 inhibits the IRF activation and suppresses IFN induction. Biochemical analysis showed that MAVS50, exposing a degenerate TRAF-binding motif within its N-terminus, effectively competed with full-length MAVS for recruiting TRAF2 and TRAF6. Ablation of the TRAF-binding motif of MAVS50 impaired its inhibitory effect on IRF activation and IFN induction. These results collectively identify a new means by which signaling events is differentially regulated via exposing key internally embedded interaction motifs, implying a more ubiquitous regulatory role of truncated proteins arose from internal translation and other related mechanisms.


Subject(s)
Adaptor Proteins, Signal Transducing/metabolism , TNF Receptor-Associated Factor 2/metabolism , TNF Receptor-Associated Factor 6/metabolism , Amino Acid Sequence , Humans , Interferon Inducers/immunology , Interferons/metabolism , NF-kappa B/metabolism , Protein Binding/physiology
6.
Mol Cell ; 58(1): 134-46, 2015 Apr 02.
Article in English | MEDLINE | ID: mdl-25752576

ABSTRACT

RIG-I is a pattern recognition receptor that senses viral RNA and is crucial for host innate immune defense. Here, we describe a mechanism of RIG-I activation through amidotransferase-mediated deamidation. We show that viral homologs of phosphoribosylformylglycinamidine synthetase (PFAS), although lacking intrinsic enzyme activity, recruit cellular PFAS to deamidate and activate RIG-I. Accordingly, depletion and biochemical inhibition of PFAS impair RIG-I deamidation and concomitant activation. Purified PFAS and viral homolog thereof deamidate RIG-I in vitro. Ultimately, herpesvirus hijacks activated RIG-I to avoid antiviral cytokine production; loss of RIG-I or inhibition of RIG-I deamidation results in elevated cytokine production. Together, these findings demonstrate a surprising mechanism of RIG-I activation that is mediated by an enzyme.


Subject(s)
Carbon-Nitrogen Ligases with Glutamine as Amide-N-Donor/immunology , DEAD-box RNA Helicases/immunology , Gammaherpesvirinae/immunology , Immune Evasion/genetics , RNA, Viral/immunology , Viral Proteins/immunology , Amides/metabolism , Animals , Carbon-Nitrogen Ligases with Glutamine as Amide-N-Donor/genetics , Cell Line , Cytokines/antagonists & inhibitors , Cytokines/biosynthesis , DEAD Box Protein 58 , DEAD-box RNA Helicases/antagonists & inhibitors , DEAD-box RNA Helicases/genetics , DEAD-box RNA Helicases/metabolism , Enzyme Activation , Fibroblasts/enzymology , Fibroblasts/immunology , Fibroblasts/virology , Gammaherpesvirinae/genetics , Gene Expression Regulation , HEK293 Cells , Humans , Immunity, Innate , Mice , Molecular Mimicry , RNA, Small Interfering/genetics , RNA, Small Interfering/metabolism , RNA, Viral/genetics , Receptors, Immunologic , Signal Transduction , Viral Proteins/genetics
7.
PLoS One ; 10(2): e0117758, 2015.
Article in English | MEDLINE | ID: mdl-25693195

ABSTRACT

Prostate cancer (PCa) is the second-leading cause of cancer-related mortality, after lung cancer, in men from developed countries. In its early stages, primary tumor growth is dependent on androgens, thus generally can be controlled by androgen deprivation therapy (ADT). Eventually however, the disease progresses to castration-resistant prostate cancer (CRPC), a lethal form in need of more effective treatments. G-protein coupled receptors (GPCRs) comprise a large clan of cell surface proteins that have been implicated as therapeutic targets in PCa growth and progression. The findings reported here provide intriguing evidence of a role for the newly characterized glutamate family member GPR158 in PCa growth and progression. We found that GPR158 promotes PCa cell proliferation independent of androgen receptor (AR) functionality and that this requires its localization in the nucleus of the cell. This suggests that GPR158 acts by mechanisms different from other GPCRs. GPR158 expression is stimulated by androgens and GPR158 stimulates AR expression, implying a potential to sensitize tumors to low androgen conditions during ADT via a positive feedback loop. Further, we found GPR158 expression correlates with a neuroendocrine (NE) differentiation phenotype and promotes anchorage-independent colony formation implying a role for GPR158 in therapeutic progression and tumor formation. GPR158 expression was increased at the invading front of prostate tumors that formed in the genetically defined conditional Pten knockout mouse model, and co-localized with elevated AR expression in the cell nucleus. Kaplan-Meier analysis on a dataset from the Memorial Sloan Kettering cancer genome portal showed that increased GPR158 expression in tumors is associated with lower disease-free survival. Our findings strongly suggest that pharmaceuticals targeting GPR158 activities could represent a novel and innovative approach to the prevention and management of CRPC.


Subject(s)
Disease Progression , Gene Expression Regulation, Neoplastic , Prostatic Neoplasms/pathology , Receptors, G-Protein-Coupled/genetics , Receptors, G-Protein-Coupled/metabolism , Active Transport, Cell Nucleus/drug effects , Androgens/pharmacology , Animals , Cell Differentiation/drug effects , Cell Line, Tumor , Cell Nucleus/drug effects , Cell Nucleus/metabolism , Disease-Free Survival , Dose-Response Relationship, Drug , Gene Expression Regulation, Neoplastic/drug effects , Gene Knockout Techniques , Humans , Male , Mice , Neuroendocrine Cells/drug effects , Neuroendocrine Cells/pathology , PTEN Phosphohydrolase/deficiency , PTEN Phosphohydrolase/genetics , Receptors, Androgen/metabolism
8.
J Proteomics Bioinform ; 8(11): 260-265, 2015 Nov.
Article in English | MEDLINE | ID: mdl-26807013

ABSTRACT

Liquid chromatography-mass spectrometry (LC-MS) based proteomics is one of the most widely used analytical platforms for global protein discovery and quantification. One of the challenges is the difficulty of identifying low abundance biomarker proteins from limited biological samples. Extensive fractionation could expand proteomics dynamic range, however, at the cost of high sample and time consumption. Extensive fractionation would increase the sample need and the labeling cost. Also quantitative proteomics depending on high resolution MS have the limitation of spectral acquisition speed. Those practical problems hinder the in-depth quantitative proteomics analysis such as tandem mass tag (TMT) experiments. We found the joint use of hydrophilic interaction liquid chromatography (HILIC) and strong cation exchange Chromatography (SCX) prefractionation at medium level could improve MS/MS efficiency, increase proteome coverage, shorten analysis time and save valuable samples. In addition, we scripted a program, Exclusion List Convertor (ELC), which automates and streamlines data acquisition workflow using the precursor ion exclusion (PIE) method. PIE reduces redundancy of high abundance MS/MS analyses by running replicates of the sample. The precursor ions detected in the initial run(s) are excluded for MS/MS in the subsequent run. We compared PIE methods with standard data dependent acquisition (DDA) methods running replicates without PIE for their effectiveness in quantifying TMT-tagged peptides and proteins in mouse tears. We quantified a total of 845 proteins and 1401 peptides using the PIE workflow, while the DDA method only resulted in 347 proteins and 731 peptides. This represents a 144% increase of protein identifications as a result of PIE analysis.

9.
Antioxid Redox Signal ; 22(3): 259-74, 2015 Jan 20.
Article in English | MEDLINE | ID: mdl-25226451

ABSTRACT

AIMS: We showed that chronic cholestatic liver injury induced the expression of c-Myc but suppressed that of glutamate-cysteine ligase (GCL, composed of catalytic and modifier subunits GCLC and GCLM, respectively). This was associated with reduced nuclear antioxidant response element (ARE) binding by nuclear factor-erythroid 2 related factor 2 (Nrf2). Here, we examined whether c-Myc is involved in this process. RESULTS: Similar to bile duct ligation (BDL), lithocholic acid (LCA) treatment in vivo induced c-Myc but suppressed GCL subunits expression at day 14. Nrf2 expression and Nrf2 ARE binding fell markedly. However, Nrf2 heterodimerization with MafG was enhanced by LCA, which prompted us to examine whether LCA treatment in vivo altered proteins that bind to ARE using biotinylated ARE in pull-down assay followed by proteomics. LCA treatment enhanced c-Myc but lowered prohibitin 1 (PHB1) binding to ARE. This was a result of c-Myc-mediated induction of microRNA 27a/b (miR27a/b), which target both PHB1 and Nrf2 to reduce their expression. Knockdown of c-Myc or miR27a/b attenuated LCA-mediated suppression of Nrf2, PHB1, and GCL subunit expression, whereas overexpression of PHB1 protected against the fall in Nrf2 and GCL subunits. Both c-Myc and PHB1 directly interact with Nrf2 but c-Myc lowers Nrf2 binding to ARE while PHB1 enhances it. INNOVATION: This is the first work that shows how activation of this circuit in cholestatic liver injury inhibits GCL expression. CONCLUSIONS: LCA feeding and BDL activate c-Myc-miR27a/b-PHB1 circuit, with the consequence of inhibiting Nrf2 expression and ARE binding, resulting in decreased reduced glutathione synthesis and antioxidant capacity.


Subject(s)
Cholestasis, Intrahepatic/metabolism , Glutathione/biosynthesis , MicroRNAs/genetics , Proto-Oncogene Proteins c-myc/metabolism , Repressor Proteins/metabolism , Animals , Antioxidant Response Elements , Base Sequence , Binding Sites , Cell Line, Tumor , Gene Expression , Glutamate-Cysteine Ligase/metabolism , Humans , Male , Mice, Inbred C57BL , NF-E2-Related Factor 2/genetics , NF-E2-Related Factor 2/metabolism , Prohibitins , Protein Binding , RNA Interference , Repressor Proteins/genetics
10.
Cell Stem Cell ; 15(1): 66-78, 2014 Jul 03.
Article in English | MEDLINE | ID: mdl-24726192

ABSTRACT

Gaseous signaling molecules such as hydrogen sulfide (H2S) are produced endogenously and mediate effects through diverse mechanisms. H2S is one such gasotransmitters that regulates multiple signaling pathways in mammalian cells, and abnormal H2S metabolism has been linked to defects in bone homeostasis. Here, we demonstrate that bone marrow mesenchymal stem cells (BMMSCs) produce H2S in order to regulate their self-renewal and osteogenic differentiation, and H2S deficiency results in defects in BMMSC differentiation. H2S deficiency causes aberrant intracellular Ca(2+) influx because of reduced sulfhydration of cysteine residues on multiple Ca(2+) TRP channels. This decreased Ca(2+) flux downregulates PKC/Erk-mediated Wnt/ß-catenin signaling which controls osteogenic differentiation of BMMSCs. Consistently, H2S-deficient mice display an osteoporotic phenotype that can be rescued by small molecules that release H2S. These results demonstrate that H2S regulates BMMSCs and that restoring H2S levels via nontoxic donors may provide treatments for diseases such as osteoporosis that can arise from H2S deficiencies.


Subject(s)
Bone Marrow Cells/physiology , Calcium Channels, T-Type/metabolism , Hydrogen Sulfide/metabolism , Mesenchymal Stem Cells/physiology , Osteoporosis/metabolism , Animals , Calcium Channels, T-Type/chemistry , Calcium Signaling/genetics , Cell Differentiation/genetics , Cells, Cultured , Cystathionine beta-Synthase/genetics , Homeostasis , Humans , Mice , Mice, Knockout , Osteogenesis/genetics , Osteoporosis/pathology , Protein Kinase C/metabolism , RNA, Small Interfering/genetics , Sulfurtransferases/genetics , Transaminases/genetics , beta Catenin/metabolism
11.
Mol Cancer Res ; 12(4): 607-21, 2014 Apr.
Article in English | MEDLINE | ID: mdl-24464914

ABSTRACT

UNLABELLED: Annexin A1 (AnxA1), a phospholipid-binding protein and regulator of glucocorticoid-induced inflammatory signaling, has implications in cancer. Here, a role for AnxA1 in prostate adenocarcinoma was determined using primary cultures and a tumor cell line (cE1), all derived from the conditional Pten deletion mouse model of prostate cancer. AnxA1 secretion by prostate-derived cancer-associated fibroblasts (CAF) was significantly higher than by normal prostate fibroblasts (NPF). Prostate tumor cells were sorted to enrich for epithelial subpopulations based on nonhematopoietic lineage, high SCA-1, and high or medium levels of CD49f. Compared with controls, AnxA1 enhanced stem cell-like properties in high- and medium-expression subpopulations of sorted cE1 and primary cells, in vitro, through formation of greater number of spheroids with increased complexity, and in vivo, through generation of more, larger, and histologically complex glandular structures, along with increased expression of p63, a basal/progenitor marker. The differentiated medium-expression subpopulations from cE1 and primary cells were most susceptible to gain stem cell-like properties as shown by increased spheroid and glandular formation. Further supporting this increased plasticity, AnxA1 was shown to regulate epithelial-to-mesenchymal transition in cE1 cells. These results suggest that CAF-secreted AnxA1 contributes to tumor stem cell dynamics via two separate but complementary pathways: induction of a dedifferentiation process leading to generation of stem-like cells from a subpopulation of cancer epithelial cells and stimulation of proliferation and differentiation of the cancer stem-like cells. IMPLICATIONS: AnxA1 participates in a paradigm in which malignant prostate epithelial cells that are not cancer stem cells are induced to gain cancer stem cell-like properties.


Subject(s)
Annexin A1/metabolism , Neoplastic Stem Cells/pathology , Prostatic Neoplasms, Castration-Resistant/pathology , Animals , Cell Differentiation/physiology , Cell Line, Tumor , Disease Models, Animal , Fibroblasts/metabolism , Fibroblasts/pathology , Humans , Male , Mice , Mice, Inbred NOD , Mice, SCID , Mice, Transgenic , Prostatic Neoplasms, Castration-Resistant/metabolism , Signal Transduction
12.
Small ; 9(3): 421-9, 2013 Feb 11.
Article in English | MEDLINE | ID: mdl-23038676

ABSTRACT

As a consequence of their well-defined nanostructure and intrinsic bioactive functionality, virus-based nanoparticles have shown promise for mediating gene delivery. Adeno-associated virus (AAV) nanoparticles, which possess an excellent safety profile and therapeutic potential, hold potential for use in human gene therapy. However, because of their native tropisms, the applicability of AAV nanoparticles is often limited to restricted ranges of cells or tissues. Thus, retargeting AAV particles to the desired cell populations has continued to be a major research focus in many gene therapy applications. In this study, a general strategy is reported for nanoparticle targeting. This involves the site-specific modification of AAV type 2 (AAV2) by genetically incorporating a short peptide, in this case an aldehyde tag, in the viral capsid. Such a tag can be exploited for site-specific attachment of targeting molecules and allows for further introduction of targeting antibodies or ligands. It is shown that this modification neither affects the level of infectious viral titer nor intracellular trafficking properties. Furthermore, the site-specific conjugation of targeting antibodies could significantly enhance viral transduction to those target cells that have otherwise exhibited very low permissiveness to AAV2 infection. This method also allows the functional incorporation of RGD peptides onto AAV2 for enhanced delivery with implications for cancer gene therapy.


Subject(s)
Dependovirus/chemistry , Dependovirus/genetics , Genetic Engineering/methods , Cell Line , Dependovirus/physiology , HeLa Cells , Hep G2 Cells , Humans , Oligopeptides/chemistry
13.
mBio ; 3(6): e00411-12, 2012 Nov 06.
Article in English | MEDLINE | ID: mdl-23131831

ABSTRACT

Central to NF-κB signaling pathways is IKKγ/NEMO, a regulatory subunit of the cytoplasmic IκB kinase (IKK) complex, which undergoes various posttranslational modifications, specifically phosphorylation, to regulate its function. Furthermore, Kaposi's sarcoma-associated herpesvirus (KSHV) FADD-like interleukin-1ß (IL-1ß) converting enzyme (FLICE) inhibitory protein (vFLIP) activates the NF-κB signaling pathway by directly interacting with IKKγ/NEMO. However, the exact functions of IKKγ/NEMO phosphorylation and its KvFLIP interaction in NF-κB activation remain elusive. Here, we report two novel phosphorylation sites of IKKγ/NEMO and their negative effect on the IKKγ/NEMO-mediated NF-κB signaling pathway. First, the Src family protein tyrosine kinases (SF-PTKs), including Src, Fyn, Lyn, and Fgr, interact with and phosphorylate tyrosine residue 374 (Y374) of IKKγ/NEMO. Mutation of the Y374 residue to phenylalanine (Y374F) specifically abolished SF-PTK-mediated tyrosine phosphorylation, leading to increased tumor necrosis factor alpha (TNF-α)-induced NF-κB activity. Moreover, our mass spectrometry analysis found that the serine 377 residue (S377) of IKKγ/NEMO underwent robust phosphorylation upon KvFLIP expression. Replacement of the IKKγ/NEMO S377 residue by alanine (S377A) or glutamic acid (S377E) resulted in a significant increase or decrease of NF-κB activity and TNF-α-mediated IL-6 cytokine production, respectively. Our study thus demonstrates that the Y374 or S377 residue located at the C-terminal proline-rich domain of human IKKγ/NEMO undergoes phosphorylation upon TNF-α treatment or KvFLIP expression, respectively, resulting in the suppression of IKKγ/NEMO activity to induce NF-κB activation. This study suggests the potential phosphorylation-mediated feedback negative regulation of IKKγ/NEMO activity in the NF-κB signaling pathway. IMPORTANCE Since unchecked regulation of NF-κB has been linked to uncontrolled proliferation and cell death, the downregulation of the NF-κB signaling pathway is as important as its activation. Specifically, the phosphorylation-mediated modification of IKKγ/NEMO is a critical regulatory mechanism of NF-κB activity. Here, we report two novel phosphorylations of IKKγ/NEMO and their negative effects on the NF-κB signaling pathway. First, the Src family protein tyrosine kinase interacts with and phosphorylates tyrosine residue 374 of IKKγ/NEMO, suppressing tumor necrosis factor alpha (TNF-α)-induced NF-κB activity. Additionally, Kaposi's sarcoma-associated herpesvirus (KSHV) FADD-like interleukin-1ß (IL-1ß) converting enzyme (FLICE) inhibitory protein (KvFLIP) expression induces a robust phosphorylation of the serine 377 residue of IKKγ/NEMO, resulting in a significant decrease of NF-κB activity. Our study thus demonstrates that the Y374 or S377 residue of IKKγ/NEMO undergoes phosphorylation upon TNF-α treatment or KvFLIP expression, respectively, resulting in the suppression of IKKγ/NEMO activity to induce NF-κB activation. This also suggests the potential phosphorylation-mediated feedback negative regulation of IKKγ/NEMO activity in the NF-κB signaling pathway.


Subject(s)
I-kappa B Kinase/metabolism , Protein Processing, Post-Translational , Amino Acid Substitution , Herpesvirus 8, Human/pathogenicity , Humans , NF-kappa B/metabolism , Phosphorylation , Serine/genetics , Serine/metabolism , Signal Transduction , Tumor Necrosis Factor-alpha/metabolism , Tyrosine/genetics , Tyrosine/metabolism
14.
Proc Natl Acad Sci U S A ; 109(40): 16348-53, 2012 Oct 02.
Article in English | MEDLINE | ID: mdl-22988091

ABSTRACT

Protein disulfide isomerase (PDI), an endoplasmic reticulum chaperone protein, catalyzes disulfide bond breakage, formation, and rearrangement. The effect of PDI inhibition on ovarian cancer progression is not yet clear, and there is a need for potent, selective, and safe small-molecule inhibitors of PDI. Here, we report a class of propynoic acid carbamoyl methyl amides (PACMAs) that are active against a panel of human ovarian cancer cell lines. Using fluorescent derivatives, 2D gel electrophoresis, and MS, we established that PACMA 31, one of the most active analogs, acts as an irreversible small-molecule inhibitor of PDI, forming a covalent bond with the active site cysteines of PDI. We also showed that PDI activity is essential for the survival and proliferation of human ovarian cancer cells. In vivo, PACMA 31 showed tumor targeting ability and significantly suppressed ovarian tumor growth without causing toxicity to normal tissues. These irreversible small-molecule PDI inhibitors represent an important approach for the development of targeted anticancer agents for ovarian cancer therapy, and they can also serve as useful probes for investigating the biology of PDI-implicated pathways.


Subject(s)
Dipeptides/chemistry , Dipeptides/pharmacology , Ovarian Neoplasms/drug therapy , Protein Disulfide-Isomerases/antagonists & inhibitors , Thiophenes/chemistry , Thiophenes/pharmacology , Alkynes/chemistry , Amino Acid Sequence , Animals , Binding Sites/genetics , Blotting, Western , Cell Line, Tumor , Cell Survival/drug effects , Chromatography, Liquid , Cysteine/metabolism , Dipeptides/metabolism , Drug Discovery , Electrophoresis, Gel, Two-Dimensional , Female , Histological Techniques , Humans , Immunoprecipitation , Mice , Mice, Nude , Microscopy, Fluorescence , Molecular Sequence Data , Molecular Structure , Propionates/chemistry , Protein Disulfide-Isomerases/genetics , Tandem Mass Spectrometry , Thiophenes/metabolism
15.
Cell Host Microbe ; 11(3): 264-76, 2012 Mar 15.
Article in English | MEDLINE | ID: mdl-22423966

ABSTRACT

Phagocytosis and autophagy are two important and related arms of the host's first-line defense against microbial invasion. Rubicon is a RUN domain containing cysteine-rich protein that functions as part of a Beclin-1-Vps34-containing autophagy complex. We report that Rubicon is also an essential, positive regulator of the NADPH oxidase complex. Upon microbial infection or Toll-like-receptor 2 (TLR2) activation, Rubicon interacts with the p22phox subunit of the NADPH oxidase complex, facilitating its phagosomal trafficking to induce a burst of reactive oxygen species (ROS) and inflammatory cytokines. Consequently, ectopic expression or depletion of Rubicon profoundly affected ROS, inflammatory cytokine production, and subsequent antimicrobial activity. Rubicon's actions in autophagy and in the NADPH oxidase complex are functionally and genetically separable, indicating that Rubicon functions in two ancient innate immune machineries, autophagy and phagocytosis, depending on the environmental stimulus. Rubicon may thus be pivotal to generating an optimal intracellular immune response against microbial infection.


Subject(s)
Autophagy , Gram-Positive Bacterial Infections/enzymology , Intracellular Signaling Peptides and Proteins/physiology , NADPH Oxidases/metabolism , Toll-Like Receptors/physiology , Animals , Autophagy-Related Proteins , Cells, Cultured , Cytokines/metabolism , Enzyme Activation , Enzyme Stability , Female , Gram-Positive Bacterial Infections/immunology , Gram-Positive Bacterial Infections/metabolism , Humans , Immunity, Innate , Intracellular Signaling Peptides and Proteins/genetics , Intracellular Signaling Peptides and Proteins/metabolism , Listeria monocytogenes , Membrane Glycoproteins/metabolism , Mice , Mice, Inbred C57BL , Mice, Transgenic , Microbial Viability , Mycobacterium bovis , NADPH Oxidase 2 , Phagocytosis , Phagosomes/enzymology , Protein Binding , Protein Transport , Reactive Oxygen Species/metabolism , Toll-Like Receptors/metabolism
16.
J Med Chem ; 54(8): 2902-14, 2011 Apr 28.
Article in English | MEDLINE | ID: mdl-21443194

ABSTRACT

Herein, we discovered a series of propynoic acid carbamoyl methyl-amides (PACMAs) with potent cytotoxicity against a panel of cancer cell lines. These compounds interrupted cell cycle progression at low micromolar concentrations and induced early and late stage apoptosis. A representative compound suppressed tumor growth without apparent toxicity in an MDA-MB-435 mouse xenograft model. We used a Kinexus 628-antibody microarray and the Ingenuity Pathway Analysis (IPA) bioinformatics tools to better understand their mechanisms. The IPA analysis revealed the initiation of Nrf2-mediated oxidative stress through modulating the expression of SOD1 and STIP1 by compound 1. The involvement of the oxidative stress pathway was further validated by measuring the levels of the PACMA-induced mitochondrial superoxide species. To our knowledge, this is the first report on the discovery and biological evaluations of PACMAs as anticancer agents. Their broad-spectrum in vitro cytotoxicity, possibly through an oxidative stress-mediated pathway, and in vivo efficacy warrant further preclinical investigations.


Subject(s)
Alkynes/pharmacology , Amides/chemistry , Antineoplastic Agents/pharmacology , Drug Discovery , Propionates/pharmacology , Alkynes/chemistry , Alkynes/pharmacokinetics , Animals , Antineoplastic Agents/chemistry , Antineoplastic Agents/pharmacokinetics , Apoptosis/drug effects , Caspase 9/drug effects , Cell Division/drug effects , Cell Line, Tumor , Drug Evaluation, Preclinical , Humans , Magnetic Resonance Spectroscopy , Mice , Oxidative Stress , Propionates/chemistry , Propionates/pharmacokinetics , Tumor Suppressor Protein p53/drug effects , Xenograft Model Antitumor Assays
17.
J Biol Chem ; 285(26): 20015-21, 2010 Jun 25.
Article in English | MEDLINE | ID: mdl-20421296

ABSTRACT

Human methionine adenosyltransferase 2beta (MAT2beta) encodes for two major splicing variants, V1 and V2, which are differentially expressed in normal tissues. Both variants are induced in human liver cancer and positively regulate growth. The aim of this work was to identify interacting proteins of V1 and V2. His-tagged V1 and V2 were overexpressed in Rosetta pLysS cells, purified, and used in a pulldown assay to identify interacting proteins from human colon cancer cell line RKO cell lysates. The eluted lysates were subjected to Western blot and in solution proteomic analyses. HuR, an mRNA-binding protein known to stabilize the mRNA of several cyclins, was identified to interact with V1 and V2. Immunoprecipitation and Western blotting confirmed their interaction in both liver and colon cancer cells. These variant proteins are located in both nucleus and cytoplasm in liver and colon cancer cells and, when overexpressed, increased the cytoplasmic HuR content. This led to increased expression of cyclin D1 and cyclin A, known targets of HuR. When endogenous expression of V1 or V2 is reduced by small interference RNA, cytoplasmic HuR content fell and the expression of these HuR target genes also decreased. Knockdown of cyclin D1 or cyclin A blunted, whereas knockdown of HuR largely prevented, the ability of V1 or V2 overexpression to induce growth. In conclusion, MAT2beta variants reside mostly in the nucleus and regulate HuR subcellular content to affect cell proliferation.


Subject(s)
Antigens, Surface/metabolism , Intracellular Space/enzymology , Methionine Adenosyltransferase/metabolism , RNA-Binding Proteins/metabolism , Alternative Splicing , Antigens, Surface/genetics , Blotting, Western , Cell Line, Tumor , Cell Nucleus/enzymology , Cell Proliferation , Cyclin A/genetics , Cyclin D1/genetics , Cytoplasm/enzymology , ELAV Proteins , ELAV-Like Protein 1 , Gene Expression Regulation, Neoplastic , Hep G2 Cells , Humans , Isoenzymes/genetics , Isoenzymes/metabolism , Methionine Adenosyltransferase/genetics , Microscopy, Confocal , Protein Binding , RNA Interference , RNA-Binding Proteins/genetics
18.
DNA Repair (Amst) ; 9(4): 429-37, 2010 Apr 04.
Article in English | MEDLINE | ID: mdl-20117966

ABSTRACT

Human nuclease Artemis belongs to the metallo-beta-lactamase protein family. It acquires double-stranded DNA endonuclease activity in the presence of DNA-PKcs. This double-stranded DNA endonuclease activity is critical for opening DNA hairpins in V(D)J recombination and is thought to be important for processing overhangs during the nonhomologous DNA end joining (NHEJ) process. Here we show that purified human Artemis exhibits single-stranded DNA endonuclease activity. This activity is proportional to the amount of highly purified Artemis from a gel filtration column. The activity is stimulated by DNA-PKcs and modulated by purified antibodies raised against Artemis. Moreover, the divalent cation-dependence and sequence-dependence of this single-stranded endonuclease activity is the same as the double-stranded DNA endonuclease activity of Artemis:DNA-PKcs. These findings further expand the range of DNA substrates upon which Artemis and Artemis:DNA-PKcs can act. The findings are discussed in the context of NHEJ.


Subject(s)
Calcium-Binding Proteins/metabolism , DNA Ligases/metabolism , DNA Repair , Deoxyribonuclease I/metabolism , Nuclear Proteins/metabolism , Animals , Cell Line , DNA Ligase ATP , DNA-Binding Proteins , Endonucleases , Humans , Nuclear Proteins/genetics
19.
J Biol Chem ; 283(51): 35354-67, 2008 Dec 19.
Article in English | MEDLINE | ID: mdl-18957422

ABSTRACT

IkappaB kinase (IKK) complex is a key regulator of NF-kappaB pathways. Signal-induced interaction of the IKKgamma (NEMO) subunit with the C-terminal IKKgamma/NEMO-binding domain (gammaBD) of IKKbeta is an essential interaction for IKK regulation. Underlying regulatory mechanism(s) of this interaction are not known. Phosphorylation of gammaBD has been suggested to play a regulatory role for IKK activation. However, a kinase that phosphorylates gammaBD has not been identified. In this study, we used a C-terminal fragment of IKKbeta as substrate and purified Polo-like kinase 1 (Plk1) from HeLa cell extracts by standard chromatography as a gammaBD kinase. Plk1 phosphorylates serines 733, 740, and 750 in the gammaBD of IKKbeta in vitro. Phosphorylating gammaBD with Plk1 decreased its affinity for IKKgamma in pulldown assay. We generated phosphoantibodies against serine 740 and showed that gammaBD is phosphorylated in vivo. Expressing a constitutively active Plk1 in mammalian cells reduced tumor necrosis factor (TNF)-induced IKK activation, resulting in decreased phosphorylation of endogenous IkappaBalpha and reduced NF-kappaB activation. To activate endogenous Plk1, cells were treated with nocodazole, which reduced TNF-induced IKK activation, and increased the phosphorylation of gammaBD. Knocking down Plk1 in mammalian cells restored TNF-induced IKK activation in nocodazole-treated cells. Activation of Plk1 inhibited TNF-induced expression of cyclin D1. In cells in which Plk1 was knocked down, TNFalpha increased expression of cyclin D1 and the proportion of cells in the S phase of the cell cycle. Taken together, this study shows that phosphorylation regulates the interaction of gammaBD of IKKbeta with IKKgamma and therefore plays a critical role for IKK activation. Moreover, we identify Plk1 as a gammaBD kinase, which negatively regulates TNF-induced IKK activation and cyclin D1 expression, thereby affecting cell cycle regulation. Untimely activation of cyclin D1 by TNFalpha can provide a potential mechanism for an involvement of TNFalpha in inflammation-induced cancer.


Subject(s)
Cell Cycle Proteins/metabolism , Cyclin D1/metabolism , I-kappa B Kinase/metabolism , NF-kappa B/metabolism , Protein Serine-Threonine Kinases/metabolism , Proto-Oncogene Proteins/metabolism , S Phase/physiology , Animals , COS Cells , Cell Cycle Proteins/genetics , Chlorocebus aethiops , Cyclin D1/genetics , HeLa Cells , Humans , I-kappa B Kinase/genetics , I-kappa B Proteins/genetics , I-kappa B Proteins/metabolism , NF-KappaB Inhibitor alpha , NF-kappa B/genetics , Phosphorylation/physiology , Protein Serine-Threonine Kinases/genetics , Protein Structure, Tertiary/physiology , Proto-Oncogene Proteins/genetics , S Phase/drug effects , Tumor Necrosis Factor-alpha/pharmacology , Polo-Like Kinase 1
20.
J Biol Chem ; 283(27): 18573-81, 2008 Jul 04.
Article in English | MEDLINE | ID: mdl-18450743

ABSTRACT

We have previously demonstrated that neuroblastoma cells increase the expression of interleukin-6 by bone marrow stromal cells and that stimulation does not require cell-cell contact. In this study we report the purification and identification of a protein secreted by neuroblastoma cells that stimulates interleukin-6 production by stromal cells. Using a series of chromatographic purification steps including heparin-affinity, ion exchange, and molecular sieve chromatography followed by trypsin digestion and liquid chromatography tandem mass spectrometry, we identified in serum-free conditioned medium of neuroblastoma cells several secreted peptides including galectin-3-binding protein, also known as 90-kDa Mac-2-binding protein. We demonstrated the presence of the galectin-3-binding protein in the conditioned medium of several neuroblastoma cell lines and in chromatographic fractions with interleukin-6 stimulatory activity. Consistently, bone marrow stromal cells express galectin-3, the receptor for galectin-3-binding protein. Supporting a role for galectin-3-binding protein in stimulating interleukin-6 expression in bone marrow stromal cells, we observed that recombinant galectin-3-binding protein stimulated interleukin-6 expression in these cells and that interleukin-6 stimulation by neuroblastoma-conditioned medium was inhibited in the presence of lactose or a neutralizing anti-galectin-3 antibody. Down-regulation of galectin-3-binding protein expression in neuroblastoma cells also decreased the interleukin-6 stimulatory activity of the conditioned medium on bone marrow stromal cells. We also provide evidence that stimulation of interleukin-6 by galectin-3-binding protein involves activation of the Erk1/2 pathway. The data, thus, identifies galectin-3-binding protein as a factor secreted by neuroblastoma cells that stimulates the expression of interleukin-6 in bone marrow stromal cells and provides a novel function for this protein in cancer progression.


Subject(s)
Bone Marrow Cells/metabolism , Carrier Proteins/biosynthesis , Glycoproteins/biosynthesis , Interleukin-6/biosynthesis , MAP Kinase Signaling System , Neoplasm Proteins/biosynthesis , Neuroblastoma/metabolism , Antigens, Neoplasm , Biomarkers, Tumor , Bone Marrow Cells/pathology , Carrier Proteins/genetics , Carrier Proteins/pharmacology , Cell Line, Tumor , Culture Media, Conditioned/pharmacology , Down-Regulation/drug effects , Galectin 3/genetics , Galectin 3/metabolism , Galectin 3/pharmacology , Gene Expression Regulation, Neoplastic/drug effects , Glycoproteins/genetics , Glycoproteins/pharmacology , Humans , Interleukin-6/genetics , Interleukin-6/pharmacology , MAP Kinase Signaling System/drug effects , MAP Kinase Signaling System/genetics , Neoplasm Proteins/genetics , Neoplasm Proteins/pharmacology , Neuroblastoma/genetics , Recombinant Proteins/biosynthesis , Recombinant Proteins/genetics , Recombinant Proteins/pharmacology , Stromal Cells/metabolism , Stromal Cells/pathology
SELECTION OF CITATIONS
SEARCH DETAIL
...