Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 22
Filter
Add more filters










Publication year range
2.
J Biol Chem ; 291(42): 21984-22000, 2016 Oct 14.
Article in English | MEDLINE | ID: mdl-27573247

ABSTRACT

Although multidrug approaches to cancer therapy are common, few strategies are based on rigorous scientific principles. Rather, drug combinations are largely dictated by empirical or clinical parameters. In the present study we developed a strategy for rational design of a regimen that selectively targets human acute myelogenous leukemia (AML) stem cells. As a starting point, we used parthenolide, an agent shown to target critical mechanisms of redox balance in primary AML cells. Next, using proteomic, genomic, and metabolomic methods, we determined that treatment with parthenolide leads to induction of compensatory mechanisms that include up-regulated NADPH production via the pentose phosphate pathway as well as activation of the Nrf2-mediated oxidative stress response pathway. Using this knowledge we identified 2-deoxyglucose and temsirolimus as agents that can be added to a parthenolide regimen as a means to inhibit such compensatory events and thereby further enhance eradication of AML cells. We demonstrate that the parthenolide, 2-deoxyglucose, temsirolimus (termed PDT) regimen is a potent means of targeting AML stem cells but has little to no effect on normal stem cells. Taken together our findings illustrate a comprehensive approach to designing combination anticancer drug regimens.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/pharmacology , Gene Expression Regulation, Leukemic/drug effects , Leukemia, Myeloid, Acute/drug therapy , Leukemia, Myeloid, Acute/metabolism , NF-E2-Related Factor 2/metabolism , Neoplasm Proteins/metabolism , Neoplastic Stem Cells/metabolism , Deoxyglucose/pharmacology , Female , Humans , Leukemia, Myeloid, Acute/pathology , Male , NADP/biosynthesis , Neoplastic Stem Cells/pathology , Sesquiterpenes/pharmacology , Sirolimus/analogs & derivatives , Sirolimus/pharmacology , Up-Regulation/drug effects
3.
Oncotarget ; 5(17): 7303-15, 2014 Sep 15.
Article in English | MEDLINE | ID: mdl-25229978

ABSTRACT

In our prior publications we characterized a conserved acetylation motif (K(R)xxKK) of evolutionarily related nuclear receptors. Recent reports showed that peroxisome proliferator activated receptor gamma (PPARγ) deacetylation by SIRT1 is involved in delaying cellular senescence and maintaining the brown remodeling of white adipose tissue. However, it still remains unknown whether lysyl residues 154 and 155 (K154/155) of the conserved acetylation motif (RIHKK) in Pparγ1 are acetylated. Herein, we demonstrate that Pparγ1 is acetylated and regulated by both endogenous TSA-sensitive and NAD-dependent deacetylases. Acetylation of lysine 154 was identified by mass spectrometry (MS) while deacetylation of lysine 155 by SIRT1 was confirmed by in vitro deacetylation assay. An in vivo labeling assay revealed K154/K155 as bona fide acetylation sites. The conserved acetylation sites of Pparγ1 and the catalytic domain of SIRT1 are both required for the interaction between Pparγ1 and SIRT1. Sirt1 and Pparγ1 converge to govern lipid metabolism in vivo. Acetylation-defective mutants of Pparγ1 were associated with reduced lipid synthesis in ErbB2 overexpressing breast cancer cells. Together, these results suggest that the conserved lysyl residues K154/K155 of Pparγ1 are acetylated and play an important role in lipid synthesis in ErbB2-positive breast cancer cells.


Subject(s)
Breast Neoplasms/metabolism , Cellular Senescence/physiology , Lipid Metabolism/physiology , PPAR gamma/metabolism , Acetylation , Amino Acid Motifs/physiology , Animals , Blotting, Western , Cell Line, Tumor , Conserved Sequence , Humans , Immunoprecipitation , Mass Spectrometry , Mice , Mutation , Oligonucleotide Array Sequence Analysis , Protein Structure, Tertiary/physiology , Sirtuin 1/metabolism , Transfection
4.
Antiviral Res ; 110: 10-9, 2014 Oct.
Article in English | MEDLINE | ID: mdl-25051026

ABSTRACT

The alphaherpesvirus varicella-zoster virus (VZV) causes chickenpox and shingles. Current treatments are acyclovir (ACV) and its derivatives, foscarnet and brivudine (BVdU). Additional antiviral compounds with increased potency and specificity are needed to treat VZV, especially to treat post-herpetic neuralgia. We evaluated ß-l-1-[5-(E-2-bromovinyl)-2-(hydroxymethyl)-1,3-(dioxolan-4-yl)] uracil (l-BHDU, 1) and 5'-O-valyl-l-BHDU (2) in three models of VZV replication: primary human foreskin fibroblasts (HFFs), skin organ culture (SOC) and in SCID-Hu mice with skin xenografts. The efficacy of l-BHDU in vivo and its drug-drug interactions were previously not known. In HFFs, 200µM l-BHDU was noncytotoxic over 3days, and l-BHDU treatment reduced VZV genome copy number and cell to cell spread. The EC50 in HFFs for l-BHDU and valyl-l-BHDU were 0.22 and 0.03µM, respectively. However, l-BHDU antagonized the activity of ACV, BVdU and foscarnet in cultured cells. Given its similar structure to BVdU, we asked if l-BHDU, like BVdU, inhibits 5-fluorouracil catabolism. BALB/c mice were treated with 5-FU alone or in combination with l-BHDU or BVdU. l-BHDU did not interfere with 5-FU catabolism. In SCID-Hu mice implanted with human skin xenografts, l-BHDU and valyl-l-BHDU were superior to ACV and valacyclovir. The maximum concentration (Cmax) levels of l-BHDU were determined in mouse and human tissues at 2h after dosing, and comparison of concentration ratios of tissue to plasma indicated saturation of uptake at the highest dose. For the first time, an l-nucleoside analog, l-BHDU, was found to be effective and well tolerated in mice.


Subject(s)
Dioxolanes/pharmacology , Fluorouracil/metabolism , Herpesvirus 3, Human/drug effects , Nucleosides/pharmacology , Uracil/analogs & derivatives , Virus Replication/drug effects , Acyclovir/antagonists & inhibitors , Acyclovir/pharmacology , Animals , Antiviral Agents/antagonists & inhibitors , Antiviral Agents/pharmacology , Bromodeoxyuridine/analogs & derivatives , Bromodeoxyuridine/antagonists & inhibitors , Bromodeoxyuridine/pharmacology , Cell Line , Chickenpox/drug therapy , Dioxolanes/adverse effects , Drug Therapy, Combination , Foscarnet/antagonists & inhibitors , Foscarnet/pharmacology , Herpes Zoster/drug therapy , Humans , Mice , Mice, Inbred BALB C , Mice, SCID , Organ Culture Techniques , Skin/virology , Uracil/adverse effects , Uracil/pharmacology
5.
J Bacteriol ; 196(15): 2789-97, 2014 Aug.
Article in English | MEDLINE | ID: mdl-24837294

ABSTRACT

Expression of the surface protein Cnm has been directly implicated in the ability of certain strains of Streptococcus mutans to bind to collagen and to invade human coronary artery endothelial cells (HCAEC) and in the killing of Galleria mellonella. Sequencing analysis of Cnm(+) strains revealed that cnm is located between the core genes SMU.2067 and SMU.2069. Reverse transcription-PCR (RT-PCR) analysis showed that cnm is cotranscribed with SMU.2067, encoding a putative glycosyltransferase referred to here as PgfS (protein glycosyltransferase of streptococci). Notably, Cnm contains a threonine-rich domain predicted to undergo O-linked glycosylation. The previously shown abnormal migration pattern of Cnm, the presence of the threonine-rich domain, and the molecular linkage of cnm with pgfS lead us to hypothesize that PgfS modifies Cnm. A ΔpgfS strain showed defects in several traits associated with Cnm expression, including collagen binding, HCAEC invasion, and killing of G. mellonella. Western blot analysis revealed that Cnm from the ΔpgfS mutant migrated at a lower molecular weight than that from the parent strain. In addition, Cnm produced by ΔpgfS was highly susceptible to proteinase K degradation, in contrast to the high-molecular-weight Cnm version found in the parent strain. Lectin-binding analyses confirmed the glycosylated nature of Cnm and strongly suggested the presence of N-acetylglucosamine residues attached to Cnm. Based on these findings, the phenotypes observed in ΔpgfS are most likely associated with defects in Cnm glycosylation that affects protein function, stability, or both. In conclusion, this study demonstrates that Cnm is a glycoprotein and that posttranslational modification mediated by PgfS contributes to the virulence-associated phenotypes linked to Cnm.


Subject(s)
Adhesins, Bacterial/metabolism , Carrier Proteins/metabolism , Glycosyltransferases/metabolism , Protein Processing, Post-Translational , Streptococcus mutans/metabolism , Adhesins, Bacterial/genetics , Adhesins, Bacterial/isolation & purification , Animals , Bacterial Adhesion/physiology , Bacterial Proteins/genetics , Bacterial Proteins/metabolism , Carrier Proteins/genetics , Carrier Proteins/isolation & purification , Collagen/metabolism , Coronary Vessels , Endothelial Cells/microbiology , Genetic Linkage , Glycosylation , Glycosyltransferases/genetics , Humans , Lectins/metabolism , Moths , Phenotype , Protein Binding , Protein Stability , Proteolysis , Sequence Deletion , Streptococcal Infections/microbiology , Streptococcus mutans/genetics , Streptococcus mutans/pathogenicity , Virulence
6.
J Urol ; 192(2): 583-92, 2014 Aug.
Article in English | MEDLINE | ID: mdl-24530986

ABSTRACT

PURPOSE: High grade bladder cancer is an extremely aggressive malignancy associated with high rates of morbidity and mortality. Understanding how exosomes may affect bladder cancer progression could reveal novel therapeutic targets. MATERIALS AND METHODS: Exosomes derived from human bladder cancer cell lines and the urine of patients with high grade bladder cancer were assessed for the ability to promote cancer progression in standard assays. Exosomes purified from the high grade bladder cancer cell line TCC-SUP and the nonmalignant urothelial cell line SV-HUC were submitted for mass spectrometry analysis. EDIL-3 was identified and selected for further analysis. Western blot was done to determine EDIL-3 levels in urinary exosomes from patients with high grade bladder cancer. shRNA gene knockdown and recombinant EDIL-3 were applied to study EDIL-3 function. RESULTS: Exosomes isolated from high grade bladder cancer cells and the urine of patients with high grade bladder cancer promoted angiogenesis and migration of bladder cancer cells and endothelial cells. We silenced EDIL-3 expression and found that shEDIL-3 exosomes did not facilitate angiogenesis, and urothelial and endothelial cell migration. Moreover, exosomes purified from the urine of patients with high grade bladder cancer contained significantly higher EDIL-3 levels than exosomes from the urine of healthy controls. EDIL-3 activated epidermal growth factor receptor signaling while blockade of epidermal growth factor receptor signaling abrogated this EDIL-3 induced bladder cell migration. CONCLUSIONS: Exosomes derived from the urine of patients with bladder cancer contains bioactive molecules such as EDIL-3. Identifying these components and their associated oncogenic pathways could lead to novel therapeutic targets and treatment strategies.


Subject(s)
Carrier Proteins/physiology , Exosomes/physiology , Urinary Bladder Neoplasms/genetics , Urinary Bladder Neoplasms/pathology , Aged , Aged, 80 and over , Calcium-Binding Proteins , Carrier Proteins/analysis , Cell Adhesion Molecules , Disease Progression , Exosomes/chemistry , Humans , Middle Aged , Tumor Cells, Cultured
7.
Br J Pharmacol ; 171(8): 2091-8, 2014 Apr.
Article in English | MEDLINE | ID: mdl-24102583

ABSTRACT

BACKGROUND AND PURPOSE: Cardiac ischaemia-reperfusion (IR) injury remains a significant clinical problem with limited treatment options available. We previously showed that cardioprotection against IR injury by nitro-fatty acids, such as nitro-linoleate (LNO2 ), involves covalent modification of mitochondrial adenine nucleotide translocase 1 (ANT1). Thus, it was hypothesized that conjugation of LNO2 to the mitochondriotropic triphenylphosphonium (TPP(+) ) moiety would enhance its protective properties. EXPERIMENTAL APPROACH: TPP(+) -LNO2 was synthesized from aminopropyl-TPP(+) and LNO2 , and characterized by direct infusion MS/MS. Its effects were assayed in primary cultures of cardiomyocytes from adult C57BL/6 mice and in mitochondria from these cells, exposed to simulated IR (SIR) conditions (oxygen and metabolite deprivation for 1h followed by normal conditions for 1h) by measuring viability by LDH release and exclusion of Trypan blue. Nitro-alkylated mitochondrial proteins were also measured by Western blots, using antibodies to TPP(+) . KEY RESULTS: TPP(+) -LNO2 protected cardiomyocytes from SIR injury more potently than the parent compound LNO2 . In addition, TPP(+) -LNO2 modified mitochondrial proteins, including ANT1, in a manner sensitive to the mitochondrial uncoupler carbonylcyanide-p-trifluoromethoxyphenylhydrazone (FCCP) and the ANT1 inhibitor carboxyatractyloside. Similar protein nitro-alkylation was obtained in cells and in isolated mitochondria, indicating the cell membrane was not a significant barrier to TPP(+) -LNO2 . CONCLUSIONS AND IMPLICATIONS: Together, these results emphasize the importance of ANT1 as a target for the protective effects of LNO2 , and suggest that TPP(+) -conjugated electrophilic lipid compounds may yield novel tools for the investigation of cardioprotection.


Subject(s)
Cardiotonic Agents/therapeutic use , Linoleic Acids/therapeutic use , Mitochondria/drug effects , Nitro Compounds/therapeutic use , Organophosphorus Compounds/therapeutic use , Animals , Cardiotonic Agents/pharmacology , Cell Survival/drug effects , Cells, Cultured , Linoleic Acid/chemistry , Linoleic Acids/chemical synthesis , Linoleic Acids/pharmacology , Male , Membrane Potential, Mitochondrial/drug effects , Membrane Potential, Mitochondrial/physiology , Mice , Mitochondria/metabolism , Mitochondria/physiology , Mitochondrial Membrane Transport Proteins/drug effects , Mitochondrial Permeability Transition Pore , Myocardial Reperfusion Injury/drug therapy , Myocytes, Cardiac/drug effects , Myocytes, Cardiac/metabolism , Nitro Compounds/chemical synthesis , Nitro Compounds/chemistry , Nitro Compounds/pharmacology , Organophosphorus Compounds/chemical synthesis , Organophosphorus Compounds/pharmacology
8.
J Biol Chem ; 288(47): 33542-33558, 2013 Nov 22.
Article in English | MEDLINE | ID: mdl-24089526

ABSTRACT

The development of strategies to eradicate primary human acute myelogenous leukemia (AML) cells is a major challenge to the leukemia research field. In particular, primitive leukemia cells, often termed leukemia stem cells, are typically refractory to many forms of therapy. To investigate improved strategies for targeting of human AML cells we compared the molecular mechanisms regulating oxidative state in primitive (CD34(+)) leukemic versus normal specimens. Our data indicate that CD34(+) AML cells have elevated expression of multiple glutathione pathway regulatory proteins, presumably as a mechanism to compensate for increased oxidative stress in leukemic cells. Consistent with this observation, CD34(+) AML cells have lower levels of reduced glutathione and increased levels of oxidized glutathione compared with normal CD34(+) cells. These findings led us to hypothesize that AML cells will be hypersensitive to inhibition of glutathione metabolism. To test this premise, we identified compounds such as parthenolide (PTL) or piperlongumine that induce almost complete glutathione depletion and severe cell death in CD34(+) AML cells. Importantly, these compounds only induce limited and transient glutathione depletion as well as significantly less toxicity in normal CD34(+) cells. We further determined that PTL perturbs glutathione homeostasis by a multifactorial mechanism, which includes inhibiting key glutathione metabolic enzymes (GCLC and GPX1), as well as direct depletion of glutathione. These findings demonstrate that primitive leukemia cells are uniquely sensitive to agents that target aberrant glutathione metabolism, an intrinsic property of primary human AML cells.


Subject(s)
Anti-Inflammatory Agents, Non-Steroidal/pharmacology , Dioxolanes/pharmacology , Glutathione/metabolism , Leukemia, Myeloid, Acute/drug therapy , Oxidative Stress/drug effects , Sesquiterpenes/pharmacology , Antigens, CD34 , Female , Glutamate-Cysteine Ligase/antagonists & inhibitors , Glutamate-Cysteine Ligase/metabolism , Glutathione/antagonists & inhibitors , Glutathione Peroxidase/antagonists & inhibitors , Glutathione Peroxidase/metabolism , Humans , Leukemia, Myeloid, Acute/metabolism , Leukemia, Myeloid, Acute/pathology , Male , Oxidation-Reduction/drug effects , Tumor Cells, Cultured , Glutathione Peroxidase GPX1
9.
J Biol Chem ; 287(29): 24698-712, 2012 Jul 13.
Article in English | MEDLINE | ID: mdl-22584576

ABSTRACT

PKCδ, a Ser/Thr kinase, promotes cell growth, tumorigenesis, and apoptosis. Human biliverdin reductase (hBVR), a Ser/Thr/Tyr kinase, inhibits apoptosis by reducing biliverdin-IX to antioxidant bilirubin. The enzymes are activated by similar stimuli. Reportedly, hBVR is a kinase-independent activator of PKCδ and is transactivated by the PKC (Gibbs, P. E., Miralem, T., Lerner-Marmarosh, N., Tudor, C., and Maines, M. D. (2012) J. Biol. Chem. 287, 1066-1079). Presently, we examined interactions between the two proteins in the context of regulation of their activities and defining targets of hBVR phosphorylation by PKCδ. LC-MS/MS analysis of PKCδ-activated intact hBVR identified phosphorylated serine positions 21, 33, 230, and 237, corresponding to the hBVR Src homology-2 domain motif (Ser(230) and Ser(237)), flanking the ATP-binding motif (Ser(21)) and in PHPS sequence (Ser(33)) as targets of PKCδ. Ser(21) and Ser(230) were also phosphorylated in hBVR-based peptides. The Ser(230)-containing peptide was a high affinity substrate for PKCδ in vitro and in cells; the relative affinity was PKCδ > PKCßII > PKCζ. Two overlapping peptides spanning this substrate, KRNRYLSF and SFHFKSGSL, were effective inhibitors of PKCδ kinase activity and PKCδ-supported activation of transcription factors Elk1 and NF-κB. Only SFHFKSGSL, in PKCδ-transfected phorbol 12-myristate 13-acetate-stimulated cells, caused membrane blebbing and cell loss. Biliverdin noncovalently inhibited PKCδ, whereas PKCδ potentiated hBVR reductase activity and accelerated the rate of bilirubin formation. This study, together with previous findings, reveals an unexpected regulatory interplay between PKCδ and hBVR in modulating cell death/survival in response to various activating stimuli. In addition, this study has identified novel substrates for and inhibitors of PKCδ. We suggest that hBVR-based technology may have utility to modulate PKCδ-mediated functions in the cell.


Subject(s)
Oxidoreductases Acting on CH-CH Group Donors/metabolism , Oxidoreductases Acting on CH-CH Group Donors/pharmacology , Peptides/chemistry , Protein Kinase C-delta/metabolism , Protein Kinase C/metabolism , Biliverdine/pharmacology , Cell Line , HEK293 Cells , HeLa Cells , Humans , Mass Spectrometry , Microscopy, Confocal , Oxidoreductases Acting on CH-CH Group Donors/chemistry , Phosphorylation/drug effects , Signal Transduction/drug effects
10.
Methods Mol Biol ; 836: 23-34, 2012.
Article in English | MEDLINE | ID: mdl-22252625

ABSTRACT

Identification of proteoglycan chain modification sites cannot yet be reliably predicted from primary amino acid sequence data. A number of studies have shown that serine is the predominant amino acid that is modified and it is frequently flanked by a C-terminal glycine and proximal N-terminal acidic amino acids; however, not all simple Ser-Gly motifs constitute a modification site. Here we present a rapid method for cloning small, defined segments of putative proteoglycan attachment sites and expressing them as a mini-reporter protein in an insect tissue culture system that is expandable to high throughput analysis. Reporter proteins with attached proteoglycans can be readily discerned from their unmodified form, by a simple gel-shift assay and Western blot detection for an epitope tag engineered into the reporter. Unmodified proteins are generated as a reference standard by treating cells with dsRNA to knock down the endogenous polypeptide xylose transferase, which is responsible for initiating proteoglycan site attachment. Examination of proteoglycan attachment by different metazoan organisms can be studied in the same cell line by cotransfecting a polypeptide xylose transferase expression plasmid and reporter construct from human, mouse, frog, or worm, for example. Reporter proteins engineer with point mutations can be rapidly generated with this system to pinpoint the exact residue that is glycosylated, to verify the mapping data.


Subject(s)
Mutagenesis, Site-Directed , Protein Interaction Mapping , Proteoglycans/genetics , Proteoglycans/metabolism , Amino Acid Sequence , Animals , Base Sequence , Cloning, Molecular , Electrophoretic Mobility Shift Assay , Genes, Reporter , Humans , Molecular Sequence Data , Proteoglycans/chemistry
11.
J Biol Chem ; 287(1): 114-122, 2012 Jan 02.
Article in English | MEDLINE | ID: mdl-22039052

ABSTRACT

Recently, we discovered a novel non-angiotensin type 1 (non-AT1), non-AT2 angiotensin binding site in rodent and human brain membranes, which is distinctly different from angiotensin receptors and key proteases processing angiotensins. It is hypothesized to be a new member of the renin-angiotensin system. This study was designed to isolate and identify this novel angiotensin binding site. An angiotensin analog, photoaffinity probe 125I-SBpa-Ang II, was used to specifically label the non-AT1, non-AT2 angiotensin binding site in mouse forebrain membranes, followed by a two-step purification procedure based on the molecular size and isoelectric point of the photoradiolabeled binding protein. Purified samples were subjected to two-dimensional gel electrophoresis followed by mass spectrometry identification of proteins in the two-dimensional gel sections containing radioactivity. LC-MS/MS analysis revealed eight protein candidates, of which the four most abundant were immunoprecipitated after photoradiolabeling. Immunoprecipitation studies indicated that the angiotensin binding site might be the membrane-bound variant of metalloendopeptidase neurolysin (EC 3.4.24.16). To verify these observations, radioligand binding and photoradiolabeling experiments were conducted in membrane preparations of HEK293 cells overexpressing mouse neurolysin or thimet oligopeptidase (EC 3.4.24.15), a closely related metalloendopeptidase of the same family. These experiments also identified neurolysin as the non-AT1, non-AT2 angiotensin binding site. Finally, brain membranes of mice lacking neurolysin were nearly devoid of the non-AT1, non-AT2 angiotensin binding site, further establishing membrane-bound neurolysin as the binding site. Future studies will focus on the functional significance of this highly specific, high affinity interaction between neurolysin and angiotensins.


Subject(s)
Cell Membrane/metabolism , Metalloendopeptidases/metabolism , Angiotensins/metabolism , Animals , Binding Sites , Electrophoresis, Gel, Two-Dimensional , Female , Gene Knockout Techniques , HEK293 Cells , Humans , Mass Spectrometry , Metalloendopeptidases/chemistry , Metalloendopeptidases/genetics , Metalloendopeptidases/isolation & purification , Mice , Pregnancy , Prosencephalon/cytology , Protein Binding
12.
Int J Oncol ; 39(5): 1273-83, 2011 Nov.
Article in English | MEDLINE | ID: mdl-21743961

ABSTRACT

Epidemiological and clinical observations provide consistent evidence that regular intake of aspirin may effectively inhibit the occurrence of epithelial tumors; however, the molecular mechanisms are not completely understood. In the present study, we determined the ability of aspirin to acetylate and post-translationally modify cellular proteins in HCT-116 human colon cancer cells to understand the potential mechanisms by which it may exerts anti-cancer effects. Using anti-acetyl lysine antibodies, here we demonstrate that aspirin causes the acetylation of multiple proteins whose molecular weight ranged from 20 to 200 kDa. The identity of these proteins was determined, using immuno-affinity purification, mass spectrometry and immuno-blotting. A total of 33 cellular proteins were potential targets of aspirin-mediated acetylation, while 16 were identified as common to both the control and aspirin-treated samples. These include enzymes of glycolytic pathway, cytoskeleton proteins, histones, ribosomal and mitochondrial proteins. The glycolytic enzymes which were identified include aldolase, glyceraldehyde-3-phosphate dehydrogenase, enolase, pyruvate kinase M2, and lactate dehydrogenase A and B chains. Immunoblotting experiment showed that aspirin also acetylated glucose-6-phosphate dehydrogenase and transketolase, both enzymes of pentose phosphate pathway involved in ribonucleotide biosynthesis. In vitro assays of these enzymes revealed that aspirin did not affect pyruvate kinase and lactate dehydrogenase activity; however, it decreased glucose 6 phosphate dehydrogenase activity. Similar results were also observed in HT-29 human colon cancer cells. Selective inhibition of glucose-6-phosphate dehydrogenase may represent an important mechanism by which aspirin may exert its anti-cancer effects through inhibition of ribonucleotide synthesis.


Subject(s)
Aspirin/pharmacology , Colonic Neoplasms/metabolism , Proteins/metabolism , Acetylation/drug effects , Enzyme Activation/drug effects , Glucosephosphate Dehydrogenase/metabolism , Glycosylation/drug effects , HCT116 Cells , HT29 Cells , Humans , Lactate Dehydrogenases/metabolism , Mass Spectrometry , Metabolic Networks and Pathways/drug effects , Pentose Phosphate Pathway/drug effects , Phosphorylation/drug effects , Proteins/analysis , Pyruvate Kinase/metabolism
13.
Bioorg Med Chem ; 19(4): 1515-9, 2011 Feb 15.
Article in English | MEDLINE | ID: mdl-21273084

ABSTRACT

Melampomagnolide B has been identified as a new antileukemic sesquiterpene. A biotin-conjugated derivative of melampomagnolide B was designed and synthesized in order to elucidate its mechanism of action. A study of the biochemical interactions of the biotin probe suggests that melampomagnolide B derives its remarkable selectivity for leukemic cells over normal hematopoietic cells from its unique ability to exploit biochemical differences between the two cell types.


Subject(s)
Antineoplastic Agents/chemical synthesis , Antineoplastic Agents/pharmacology , Leukemia/drug therapy , Sesquiterpenes/chemical synthesis , Sesquiterpenes/pharmacology , Cell Survival/drug effects , Humans , Molecular Structure , Tumor Cells, Cultured
14.
J Proteome Res ; 9(12): 6605-14, 2010 Dec 03.
Article in English | MEDLINE | ID: mdl-20858015

ABSTRACT

The saliva proteome includes host defense factors and specific bacterial-binding proteins that modulate microbial growth and colonization of the tooth surface in the oral cavity. A multidimensional mass spectrometry approach identified the major host-derived salivary proteins that interacted with Streptococcus mutans (strain UA159), the primary microorganism associated with the pathogenesis of dental caries. Two abundant host proteins were found to tightly bind to S. mutans cells, common salivary protein-1 (CSP-1) and deleted in malignant brain tumor 1 (DMBT1, also known as salivary agglutinin or gp340). In contrast to gp340, limited functional information is available on CSP-1. The sequence of CSP-1 shares 38.1% similarity with rat CSP-1. Recombinant CSP-1 (rCSP-1) protein did not cause aggregation of S. mutans cells and was devoid of any significant biocidal activity (2.5 to 10 µg/mL). However, S. mutans cells exposed to rCSP-1 (10 µg/mL) in saliva displayed enhanced adherence to experimental salivary pellicle and to glucans in the pellicle formed on hydroxyapatite surfaces. Thus, our data demonstrate that the host salivary protein CSP-1 binds to S. mutans cells and may influence the initial colonization of this pathogenic bacterium onto the tooth surface.


Subject(s)
Dental Pellicle/metabolism , Durapatite/metabolism , Glucans/metabolism , Salivary Proteins and Peptides/metabolism , Streptococcus mutans/metabolism , Amino Acid Sequence , Animals , Bacterial Adhesion/drug effects , Calcium-Binding Proteins , Cell Line , DNA-Binding Proteins , Dental Pellicle/drug effects , Dental Pellicle/microbiology , Electrophoresis, Polyacrylamide Gel , Humans , Intercellular Signaling Peptides and Proteins , Molecular Sequence Data , Protein Binding , Proteins/genetics , Proteins/metabolism , Proteins/pharmacology , Receptors, Cell Surface/metabolism , Recombinant Proteins/metabolism , Recombinant Proteins/pharmacology , Saliva/metabolism , Saliva/microbiology , Salivary Proteins and Peptides/genetics , Salivary Proteins and Peptides/pharmacology , Sequence Homology, Amino Acid , Streptococcus mutans/drug effects , Streptococcus mutans/growth & development , Tumor Suppressor Proteins
15.
Proteomics Clin Appl ; 3(1): 116-134, 2009 Jan 01.
Article in English | MEDLINE | ID: mdl-19898684

ABSTRACT

The proteome of human salivary fluid has the potential to open new doors for disease biomarker discovery. A recent study to comprehensively identify and catalog the human ductal salivary proteome led to the compilation of 1166 proteins. The protein complexity of both saliva and plasma is large, suggesting that a comparison of these two proteomes will provide valuable insight into their physiological significance and an understanding of the unique and overlapping disease diagnostic potential that each fluid provides. To create a more comprehensive catalog of human salivary proteins, we have first compiled an extensive list of proteins from whole saliva (WS) identified through MS experiments. The WS list is thereafter combined with the proteins identified from the ductal parotid, and submandibular and sublingual (parotid/SMSL) salivas. In parallel, a core dataset of the human plasma proteome with 3020 protein identifications was recently released. A total of 1939 nonredundant salivary proteins were compiled from a total of 19 474 unique peptide sequences identified from whole and ductal salivas; 740 out of the total 1939 salivary proteins were identified in both whole and ductal saliva. A total of 597 of the salivary proteins have been observed in plasma. Gene ontology (GO) analysis showed similarities in the distributions of the saliva and plasma proteomes with regard to cellular localization, biological processes, and molecular function, but revealed differences which may be related to the different physiological functions of saliva and plasma. The comprehensive catalog of the salivary proteome and its comparison to the plasma proteome provides insights useful for future study, such as exploration of potential biomarkers for disease diagnostics.

16.
J Proteome Res ; 8(11): 5093-102, 2009 Nov.
Article in English | MEDLINE | ID: mdl-19764810

ABSTRACT

Human saliva is a protein-rich, easily accessible source of potential local and systemic biomarkers to monitor changes that occur under pathological conditions; however, little is known about the changes in abundance associated with normal aging. In this study, we performed a comprehensive proteomic profiling of pooled saliva collected from the parotid glands of healthy female subjects, divided into two age groups 1 and 2 (20-30 and 55-65 years old, respectively). Hydrophobic charge interaction chromatography was used to separate high- from low-abundance proteins prior to characterization of the parotid saliva using multidimensional protein identification technology (MudPIT). Collectively, 532 proteins were identified in the two age groups. Of these proteins, 266 were identified exclusively in one age group, while 266 proteins were common to both groups. The majority of the proteins identified in the two age groups belonged to the defense and immune response category. Of note, several defense related proteins (e.g., lysozyme, lactoferrin and histatin-1) were significantly more abundant in group 2 as determined by G-test. Selected representative mass spectrometric findings were validated by Western blot analysis. Our study reports the first quantitative analysis of differentially regulated proteins in ductal saliva collected from young and older female subjects. This study supports the use of high-throughput proteomics as a robust discovery tool. Such results provide a foundation for future studies to identify specific salivary proteins which may be linked to age-related diseases specific to women.


Subject(s)
Aging/physiology , Parotid Gland/chemistry , Salivary Proteins and Peptides/analysis , Adult , Aged , Biomarkers/analysis , Blotting, Western , Electrophoresis, Polyacrylamide Gel , Female , Humans , Middle Aged , Proteomics/methods , Reproducibility of Results , Young Adult
17.
J Proteome Res ; 8(3): 1304-14, 2009 Mar.
Article in English | MEDLINE | ID: mdl-19199708

ABSTRACT

Human ductal saliva contributes over a thousand unique proteins to whole oral fluids. The mechanism by which most of these proteins are secreted by salivary glands remains to be determined. The present study used a mass spectrometry-based, shotgun proteomics approach to explore the possibility that a subset of the proteins found in saliva are derived from exosomes, membrane-bound vesicles of endosomal origin within multivesicular endosomes. Using MudPIT (multidimensional protein identification technology) mass spectrometry, we catalogued 491 proteins in the exosome fraction of human parotid saliva. Many of these proteins were previously observed in ductal saliva from parotid glands (265 proteins). Furthermore, 72 of the proteins in parotid exosomes overlap with those previously identified as urinary exosome proteins, proteins which are also frequently associated with exosomes from other tissues and cell types. Gene Ontology (GO) and KEGG pathway analyses found that cytosolic proteins comprise the largest category of proteins in parotid exosomes (43%), involved in such processes as phosphatidylinositol signaling system, calcium signaling pathway, inositol metabolism, protein export, and signal transduction, among others; whereas the integral plasma membrane proteins and associated/peripheral plasma membrane proteins (26%) were associated with extracellular matrix-receptor interaction, epithelial cell signaling, T-cell and B-cell receptor signaling, cytokine receptor interaction, and antigen processing and presentation, among other biological functions. In addition, these putative saliva exosomal proteins were linked to specific diseases (e.g., neurodegenerative disorders, prion disease, cancers, type I and II diabetes). Consequently, parotid glands secrete exosomes that reflect the metabolic and functional status of the gland and may also carry informative protein markers useful in the diagnosis and treatment of systemic diseases.


Subject(s)
Exosomes/metabolism , Parotid Gland/metabolism , Proteome/metabolism , Saliva/metabolism , Endosomes/metabolism , Humans
18.
J Proteome Res ; 7(5): 1994-2006, 2008 May.
Article in English | MEDLINE | ID: mdl-18361515

ABSTRACT

Saliva is a body fluid with important functions in oral and general health. A consortium of three research groups catalogued the proteins in human saliva collected as the ductal secretions: 1166 identifications--914 in parotid and 917 in submandibular/sublingual saliva--were made. The results showed that a high proportion of proteins that are found in plasma and/or tears are also present in saliva along with unique components. The proteins identified are involved in numerous molecular processes ranging from structural functions to enzymatic/catalytic activities. As expected, the majority mapped to the extracellular and secretory compartments. An immunoblot approach was used to validate the presence in saliva of a subset of the proteins identified by mass spectrometric approaches. These experiments focused on novel constituents and proteins for which the peptide evidence was relatively weak. Ultimately, information derived from the work reported here and related published studies can be used to translate blood-based clinical laboratory tests into a format that utilizes saliva. Additionally, a catalogue of the salivary proteome of healthy individuals allows future analyses of salivary samples from individuals with oral and systemic diseases, with the goal of identifying biomarkers with diagnostic and/or prognostic value for these conditions; another possibility is the discovery of therapeutic targets.


Subject(s)
Parotid Gland/chemistry , Proteome/analysis , Saliva/chemistry , Salivary Proteins and Peptides/analysis , Sublingual Gland/chemistry , Submandibular Gland/chemistry , Adult , Blood Proteins/analysis , Female , Humans , Male , Middle Aged , Molecular Sequence Data , Protein Array Analysis , Tears/chemistry
19.
Mol Cell Biol ; 27(24): 8783-96, 2007 Dec.
Article in English | MEDLINE | ID: mdl-17923703

ABSTRACT

Core-type protein O glycosylation is initiated by polypeptide N-acetylgalactosamine (GalNAc) transferase (ppGalNAcT) activity and produces the covalent linkage of serine and threonine residues of proteins. More than a dozen ppGalNAcTs operate within multicellular organisms, and they differ with respect to expression patterns and substrate selectivity. These distinctive features imply that each ppGalNAcT may differentially modulate regulatory processes in animal development, physiology, and perhaps disease. We found that ppGalNAcT-1 plays key roles in cell and glycoprotein selective functions that modulate the hematopoietic system. Loss of ppGalNAcT-1 activity in the mouse results in a bleeding disorder which tracks with reduced plasma levels of blood coagulation factors V, VII, VIII, IX, X, and XII. ppGalNAcT-1 further supports leukocyte trafficking and residency in normal homeostatic physiology as well as during inflammatory responses, in part by providing a scaffold for the synthesis of selectin ligands expressed by neutrophils and endothelial cells of peripheral lymph nodes. Animals lacking ppGalNAcT-1 are also markedly impaired in immunoglobulin G production, coincident with increased germinal center B-cell apoptosis and reduced levels of plasma B cells. These findings reveal that the initiation of protein O glycosylation by ppGalNAcT-1 provides a distinctive repertoire of advantageous functions that support vascular responses and humoral immunity.


Subject(s)
Antibody Formation/immunology , Blood Physiological Phenomena , N-Acetylgalactosaminyltransferases/metabolism , Animals , Apoptosis , B-Lymphocytes/cytology , B-Lymphocytes/enzymology , Germ Cells/enzymology , Germinal Center/enzymology , Glycosylation , Hemorrhage , Hemostasis , Humans , Immunization , Immunoglobulin G/biosynthesis , Inflammation/enzymology , Leukocytes/enzymology , Ligands , Lymphocytes/enzymology , Mice , Mutagenesis , N-Acetylgalactosaminyltransferases/deficiency , Neutrophil Infiltration/immunology , Protein Transport , Selectins/metabolism , Polypeptide N-acetylgalactosaminyltransferase
20.
J Biol Chem ; 282(19): 14586-97, 2007 May 11.
Article in English | MEDLINE | ID: mdl-17369258

ABSTRACT

Proteoglycan modification is essential for development and early cell division in Caenorhabditis elegans. The specification of proteoglycan attachment sites is defined by the Golgi enzyme polypeptide xylosyltransferase. Here we evaluate the substrate specificity of this xylosyltransferase for its downstream targets by using reporter proteins containing proteoglycan modification sites from C. elegans syndecan/SDN-1. The N terminus of the SDN-1 contains a Ser-Gly proteoglycan site at Ser(71), flanked by potential mucin and N-glycosylation sites. However, Ser(71) was exclusively used as a proteoglycan site in vivo, based on mapping studies with a Ser(71) reporter protein, glycosyltransferase RNA interference, and co-expression of worm polypeptide xylosyltransferase. To elucidate the substrate requirements of this enzyme, a library of 42 point mutants of the Ser(71) reporter was expressed in tissue culture. The nematode proteoglycan modification site in SDN-1 required serine (not threonine), two flanking glycine residues (positions -1 and +1), and either one proximal acidic N-terminal amino acid (positions -4, -3, and -2) or a pair of distal N-terminal acidic amino acids (positions -6 and -5). C-terminal acidic amino acids, although present in many proteoglycan modification sites, had minimal impact on xylosylation at Ser(71). Proline inhibited glycosylation when present at -1, +1, or +2. The position of glycine, proline, and acidic amino acids allows the glycosylation machinery to discriminate between mucin and proteoglycan modification sites. The key residues that define proteoglycan modification sites also function with the Drosophila polypeptide xylosyltransferase, indicating that the specificity in the glycosylation process is evolutionarily conserved. Using a neural network method, a preliminary proteoglycan predictor has been developed.


Subject(s)
Caenorhabditis elegans/genetics , Pentosyltransferases/genetics , Proteoglycans/genetics , Syndecans/genetics , Amino Acid Sequence , Animals , Caenorhabditis elegans/enzymology , Caenorhabditis elegans/growth & development , Caenorhabditis elegans Proteins/genetics , Caenorhabditis elegans Proteins/metabolism , Cells, Cultured , Drosophila melanogaster/enzymology , Drosophila melanogaster/genetics , Drosophila melanogaster/growth & development , Glycine/chemistry , Glycosylation , Golgi Apparatus/enzymology , Molecular Sequence Data , Mucins/metabolism , Mutagenesis, Site-Directed , Pentosyltransferases/metabolism , Proline/chemistry , Proteoglycans/metabolism , Recombinant Proteins , Sequence Homology, Amino Acid , Serine/chemistry , Substrate Specificity , Syndecans/metabolism , UDP Xylose-Protein Xylosyltransferase
SELECTION OF CITATIONS
SEARCH DETAIL
...