Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 16 de 16
Filter
1.
J Am Chem Soc ; 146(27): 18427-18439, 2024 Jul 10.
Article in English | MEDLINE | ID: mdl-38946080

ABSTRACT

Pseudomonas aeruginosa bacteria are becoming increasingly resistant against multiple antibiotics. Therefore, the development of vaccines to prevent infections with these bacteria is an urgent medical need. While the immunological activity of lipopolysaccharide O-antigens in P. aeruginosa is well-known, the specific protective epitopes remain unidentified. Herein, we present the first chemical synthesis of highly functionalized aminoglycoside trisaccharide 1 and its acetamido derivative 2 found in the P. aeruginosa serotype O5 O-antigen. The synthesis of the trisaccharide targets is based on balancing the reactivity of disaccharide acceptors and monosaccharide donors. Glycosylations were analyzed by quantifying the reactivity of the hydroxyl group of the disaccharide acceptor using the orbital-weighted Fukui function and dual descriptor. The stereoselective formation of 1,2-cis-α-fucosylamine linkages was achieved through a combination of remote acyl participation and reagent modulation. The simultaneous SN2 substitution of azide groups at C2' and C2″ enabled the efficient synthesis of 1,2-cis-ß-linkages for both 2,3-diamino-D-mannuronic acids. Through a strategic orthogonal modification, the five amino groups on target trisaccharide 1 were equipped with a rare acetamidino (Am) and four acetyl (Ac) groups. Glycan microarray analyses of sera from patients infected with P. aeruginosa indicated that trisaccharides 1 and 2 are key antigenic epitopes of the serotype O5 O-antigen. The acetamidino group is not an essential determinant of antibody binding. The ß-D-ManpNAc3NAcA residue is a key motif for the antigenicity of serotype O5 O-antigen. These findings serve as a foundation for the development of glycoconjugate vaccines targeting P. aeruginosa serotype O5.


Subject(s)
Aminoglycosides , O Antigens , Pseudomonas aeruginosa , Trisaccharides , Pseudomonas aeruginosa/immunology , O Antigens/chemistry , O Antigens/immunology , Trisaccharides/chemistry , Trisaccharides/immunology , Trisaccharides/chemical synthesis , Aminoglycosides/chemistry , Aminoglycosides/chemical synthesis , Aminoglycosides/immunology
2.
Semin Cancer Biol ; 76: 301-309, 2021 11.
Article in English | MEDLINE | ID: mdl-33812985

ABSTRACT

Ras proteins are small GTPases that participate in multiple signal cascades, regulating crucial cellular processes including cell survival, proliferation, and differentiation. Mutations or deregulated activities of Ras are frequently the driving force for oncogenic transformation and tumorigenesis. Posttranslational modifications play a crucial role in mediating the stability, activity, or subcellular localization/trafficking of numerous cellular regulators including Ras proteins. A series of recent studies reveal that Ras proteins are also regulated by sumoylation. All three Ras protein isoforms (HRas, KRas, and NRas) are modified by SUMO3. The conserved lysine42 appears to be the primary site for mediating sumoylation. Expression of KRasV12/R42 mutants compromised the activation of the Raf/MEK/ERK signaling axis, leading to a reduced rate of cell migration and invasion in vitro in multiple cell lines. Moreover, treatment of transformed pancreatic cells with a SUMO E2 inhibitor blocks cell migration in a concentration-dependent manner, which is associated with a reduced level of both KRas sumoylation and expression of mesenchymal cell markers. Furthermore, mouse xenograft experiments reveal that expression of a SUMO-resistant mutant appears to suppress tumor development in vivo. Combined, these studies indicate that sumoylation functions as an important mechanism in mediating the roles of Ras in cell proliferation, differentiation, and malignant transformation and that the SUMO-modification system of Ras oncoproteins can be explored as a new druggable target for various human malignancies.


Subject(s)
Cell Transformation, Neoplastic/metabolism , Signal Transduction/physiology , Sumoylation/physiology , ras Proteins/metabolism , Animals , Humans
3.
J Cell Biochem ; 113(6): 1998-2008, 2012 Jun.
Article in English | MEDLINE | ID: mdl-22253131

ABSTRACT

Arachidonate 5-lipoxygenase (ALOX5) expression and activity has been implicated in tumor pathogenesis, yet its role in papillary thyroid carcinoma (PTC) has not been characterized. ALOX5 protein and mRNA were upregulated in PTC compared to matched, normal thyroid tissue, and ALOX5 expression correlated with invasive tumor histopathology. Evidence suggests that PTC invasion is mediated through the induction of matrix metalloproteinases (MMPs) that can degrade and remodel the extracellular matrix (ECM). A correlation between MMP-9 and ALOX5 protein expression was established by immunohistochemical analysis of PTC and normal thyroid tissues using a tissue array. Transfection of ALOX5 into a PTC cell line (BCPAP) increased MMP-9 secretion and cell invasion across an ECM barrier. The ALOX5 product, 5(S)-hydroxyeicosatetraenoic acid also increased MMP-9 protein expression by BCPAP in a dose-dependent manner. Inhibitors of MMP-9 and ALOX5 reversed ALOX5-enhanced invasion. Here we describe a new role for ALOX5 as a mediator of invasion via MMP-9 induction; this ALOX5/MMP9 pathway represents a new avenue in the search for functional biomarkers and/or potential therapeutic targets for aggressive PTC.


Subject(s)
Arachidonate 5-Lipoxygenase/metabolism , Hydroxyeicosatetraenoic Acids/metabolism , Matrix Metalloproteinase 9/metabolism , Neoplasm Invasiveness , Thyroid Neoplasms/metabolism , Adult , Aged , Arachidonate 5-Lipoxygenase/genetics , Biomarkers, Tumor , Carcinoma , Carcinoma, Papillary , Cell Cycle , Cell Line, Tumor , Cell Movement , Cell Proliferation , Extracellular Matrix/metabolism , Female , Humans , Hydroxyeicosatetraenoic Acids/pharmacology , Lipoxygenase Inhibitors/pharmacology , Male , Matrix Metalloproteinase Inhibitors , Middle Aged , RNA, Messenger/genetics , RNA, Messenger/metabolism , Thyroid Cancer, Papillary , Thyroid Neoplasms/pathology , Young Adult
4.
Proc Natl Acad Sci U S A ; 106(8): 2812-7, 2009 Feb 24.
Article in English | MEDLINE | ID: mdl-19196975

ABSTRACT

Medulloblastomas (MBs) are the most common brain tumors in children. Some are thought to originate from cerebellar granule neuron progenitors (GNPs) that fail to undergo normal cell cycle exit and differentiation. Because microRNAs regulate numerous aspects of cellular physiology and development, we reasoned that alterations in miRNA expression might contribute to MB. We tested this hypothesis using 2 spontaneous mouse MB models with specific initiating mutations, Ink4c-/-; Ptch1+/- and Ink4c-/-; p53-/-. We found that 26 miRNAs showed increased expression and 24 miRNAs showed decreased expression in proliferating mouse GNPs and MBs relative to mature mouse cerebellum, regardless of genotype. Among the 26 overexpressed miRNAs, 9 were encoded by the miR-17 approximately 92 cluster family, a group of microRNAs implicated as oncogenes in several tumor types. Analysis of human MBs demonstrated that 3 miR-17 approximately 92 cluster miRNAs (miR-92, miR-19a, and miR-20) were also overexpressed in human MBs with a constitutively activated Sonic Hedgehog (SHH) signaling pathway, but not in other forms of the disease. To test whether the miR-17 approximately 92 cluster could promote MB formation, we enforced expression of these miRNAs in GNPs isolated from cerebella of postnatal (P) day P6 Ink4c-/-; Ptch1+/- mice. These, but not similarly engineered cells from Ink4c-/-; p53-/- mice, formed MBs in orthotopic transplants with complete penetrance. Interestingly, orthotopic mouse tumors ectopically expressing miR-17 approximately 92 lost expression of the wild-type Ptch1 allele. Our findings suggest a functional collaboration between the miR-17 approximately 92 cluster and the SHH signaling pathway in the development of MBs in mouse and man.


Subject(s)
Hedgehog Proteins/metabolism , Medulloblastoma/metabolism , MicroRNAs/genetics , Multigene Family , Animals , Cerebellum/growth & development , Cerebellum/metabolism , Humans , Medulloblastoma/genetics , Medulloblastoma/pathology , Mice , Reverse Transcriptase Polymerase Chain Reaction , Signal Transduction
5.
Mol Cell Biol ; 22(10): 3450-9, 2002 May.
Article in English | MEDLINE | ID: mdl-11971976

ABSTRACT

Human Polo-like kinase 3 (Plk3, previously termed Prk or Fnk) is involved in regulation of cell cycle progression through the M phase (B. Ouyang, H. Pan, L. Lu, J. Li, P. Stambrook, B. Li, and W. Dai, J. Biol. Chem. 272:28646-28651, 1997). Here we report that in most interphase cells endogenous Plk3 was predominantly localized around the nuclear membrane. Double labeling with Plk3 and gamma-tubulin, the latter a major component of pericentriole materials, revealed that Plk3 was closely associated with centrosomes and that its localization to centrosomes was dependent on the integrity of microtubules. Throughout mitosis, Plk3 appeared to be localized to mitotic apparatus such as spindle poles and mitotic spindles. During telophase, a significant amount of Plk3 was also detected in the midbody. Ectopic expression of Plk3 mutants dramatically changed cell morphology primarily due to their effects on microtubule dynamics. Expression of a constitutively active Plk3 (Plk3-A) resulted in rapid cell shrinkage, which led to formation of cells with an elongated, unsevered, and taxol-sensitive midbody. In contrast, cells expressing a kinase-defective Plk3 (Plk3(K52R)) mutant exhibited extended, deformed cytoplasmic structures, the phenotype of which was somewhat refractory to taxol treatment. Expression of both Plk3-A and Plk3(K52R) induced apparent G(2)/M arrest followed by apoptosis, although the kinase-defective mutant was less effective. Taken together, our studies strongly suggest that Plk3 plays an important role in the regulation of microtubule dynamics and centrosomal function in the cell and that deregulated expression of Plk3 results in cell cycle arrest and apoptosis.


Subject(s)
Apoptosis/physiology , Cell Cycle Proteins/metabolism , Cell Cycle/physiology , Microtubules/metabolism , Protein Serine-Threonine Kinases/metabolism , Cell Cycle Proteins/genetics , Cell Fractionation , Cell Size , Flow Cytometry , Humans , Immunohistochemistry , Nuclear Envelope/metabolism , Protein Kinases/metabolism , Protein Serine-Threonine Kinases/genetics , Proteins/metabolism , Proto-Oncogene Proteins , Spindle Apparatus/metabolism , Transfection , Tumor Cells, Cultured , Tumor Suppressor Proteins , Polo-Like Kinase 1
6.
Oncogene ; 24(2): 277-86, 2005 Jan 10.
Article in English | MEDLINE | ID: mdl-15640843

ABSTRACT

Protein kinases play a pivotal role in execution of cell division. Polo and Polo-like kinases have emerged as major regulators for various cell cycle checkpoints. Early genetic studies have demonstrated that CDC5, a budding yeast counterpart of vertebrate Plks, is essential for successful mitotic progression. Mammalian Plks localize primarily to the centrosome during interphase and the mitotic apparatus during mitosis. Many key cell cycle regulators such as p53, Cdc25C, cyclin B, components of the anaphase-promoting complex, and mitotic motor proteins are directly targeted by Plks. Although the exact mechanism of action of these protein kinases in vivo remains to be elucidated, Plks are important mediators for various cell cycle checkpoints that monitor centrosome duplication, DNA replication, formation of bipolar mitotic spindle, segregation of chromosomes, and mitotic exit, thus protecting cells against genetic instability during cell division.


Subject(s)
Cell Cycle/physiology , Protein Serine-Threonine Kinases/physiology , Animals , Cytokinesis/physiology , DNA Damage/physiology , DNA Repair/physiology , Humans , S Phase/physiology , Spindle Apparatus/physiology , Yeasts/physiology
7.
Cancer Res ; 64(2): 440-5, 2004 Jan 15.
Article in English | MEDLINE | ID: mdl-14744753

ABSTRACT

A compromised spindle checkpoint is thought to play a key role in genetic instability that predisposes cells to malignant transformation. Loss of function mutations of BubR1, an important component of the spindle checkpoint, have been detected in human cancers. Here we show that BubR1(+/-) mouse embryonic fibroblasts are defective in spindle checkpoint activation, contain a significantly reduced amount of securin and Cdc20, and exhibit a greater level of micronuclei than do wild-type cells. RNA interference-mediated down-regulation of BubR1 also greatly reduced securin level. Moreover, compared with wild-type littermates, BubR1(+/-) mice rapidly develop lung as well as intestinal adenocarcinomas in response to challenge with carcinogen. BubR1 is thus essential for spindle checkpoint activation and tumor suppression.


Subject(s)
Mitosis , Protein Kinases/physiology , Adenoma/pathology , Animals , Cell Cycle Proteins/genetics , Cell Transformation, Neoplastic , Female , Fibroblasts/cytology , Fibroblasts/physiology , Gene Expression Regulation , HeLa Cells , Humans , Lung Neoplasms/pathology , Mice , Mice, Knockout , Protein Kinases/deficiency , Protein Kinases/genetics , Protein Serine-Threonine Kinases , RNA, Small Interfering/genetics , Spindle Apparatus/physiology
8.
Oncogene ; 23(21): 3822-9, 2004 May 06.
Article in English | MEDLINE | ID: mdl-15021912

ABSTRACT

MEK1, a gene product that regulates cell growth and differentiation, also plays an important role in Golgi breakdown during the cell cycle. We have recently shown that polo-like kinase (Plk3) is Golgi localized and involved in Golgi dynamics during the cell cycle. To study the mode of action of Plk3 in the Golgi fragmentation cascade, we examined functional as well as physical interactions between Plk3 and MEK1/ERKs. In HeLa cells, although a significant amount of Plk3 signals dispersed in a manner similar to those of Golgi during mitosis concentrated Plk3 was detected at spindle poles, which colocalized with phospho-MEKs and phospho-ERKs. Pull-down assays showed that Plk3 physically interacted with MEK1 and ERK2. Nocodazole activated Plk3 and its activation was blocked by MEK-specific inhibitors (PD98059 or U0126). Moreover, transfection of activated MEK1 resulted in an enhanced kinase activity of Plk3; Plk3-induced fragmentation of Golgi stacks was significantly reduced after treatment with MEK inhibitors. Consistently, ectopic expression of activated MEK1, but not kinase-dead MEK1(K97R), stimulated Plk3 to induce Golgi breakdown and the stimulation was not observed in cells expressing Plk3(K52R). Furthermore, PLK3(-/-) murine embryonic fibroblast cells exhibited a significantly less fragmentation of the Golgi complex than that in wild-type cells after exposed to nocodazole. Thus, our studies strongly suggest that Plk3 may be a key protein kinase mediating MEK1 function in the Golgi fragmentation pathway during cell division.


Subject(s)
Golgi Apparatus/physiology , Mitogen-Activated Protein Kinase Kinases/physiology , Protein Serine-Threonine Kinases/physiology , Cell Cycle , HeLa Cells , Humans , MAP Kinase Kinase 1 , Mitogen-Activated Protein Kinase 1/physiology , Tumor Suppressor Proteins
9.
Cell Cycle ; 1(6): 424-9, 2002.
Article in English | MEDLINE | ID: mdl-12548019

ABSTRACT

Polo-like kinase 3 (Plk3, alternatively termed Prk) is involved in the regulation of DNA damage checkpoint as well as in M-phase function. Plk3 physically interacts with p53 and phosphorylates this tumor suppressor protein on serine-20, suggesting that the role of Plk3 in cell cycle progression is mediated, at least in part, through direct regulation of p53. Here we show that Plk3 is rapidly activated by reactive oxygen species in normal diploid fibroblast cells (WI-38), correlating with a subsequent increase in p53 protein level. Plk3 physically interacts with Chk2 and the interaction is enhanced upon DNA damage. In addition, Chk2 immunoprecipitated from cell lysates of Daudi (which expressed little Plk3) is capable of stimulating the kinase activity of purified recombinant Plk3 in vitro, and this stimulation is more pronounced when Plk3 is supplemented with Chk2 immunoprecipitated from Daudi after DNA damage. Furthermore, ectopic expression Chk2 activates cellular Plk3. Together, our studies suggest Chk2 may mediate direct activation of Plk3 in response to genotoxic stresses.


Subject(s)
Cell Cycle Proteins/metabolism , Cell Cycle/genetics , Cell Transformation, Neoplastic/genetics , DNA Damage/genetics , Neoplasms/genetics , Protein Kinases/metabolism , Protein Serine-Threonine Kinases/metabolism , Cell Cycle/drug effects , Cell Cycle Proteins/drug effects , Cell Cycle Proteins/genetics , Cell Transformation, Neoplastic/metabolism , Checkpoint Kinase 2 , DNA Damage/drug effects , Humans , Neoplasms/metabolism , Oxidative Stress/drug effects , Oxidative Stress/genetics , Protein Kinases/genetics , Protein Serine-Threonine Kinases/drug effects , Protein Serine-Threonine Kinases/genetics , Reactive Oxygen Species/pharmacology , Tumor Cells, Cultured , Tumor Suppressor Protein p53/metabolism , Tumor Suppressor Proteins , Up-Regulation/drug effects , Up-Regulation/genetics
10.
J Interferon Cytokine Res ; 22(9): 957-63, 2002 Sep.
Article in English | MEDLINE | ID: mdl-12396717

ABSTRACT

After hydrogen peroxide (H(2)O(2)) treatment, the p21 (p21(Waf1/Cip1)) protein level in GM00637 fibroblast cells was rapidly decreased, reaching its nadir around 3 h. However, it rebounded within 5 hours to a level higher than that before treatment. Fluorescence microscopic analyses revealed that nuclear p21 was downregulated during the initial oxidative stress. H(2)O(2)-induced downregulation of p21 protein was accompanied by a gradual increase in p21 mRNA levels. Other inducers of genotoxic stress, such as treatment with adriamycin, a DNA damage compound, did not cause a significant decrease in p21 protein levels. Pretreatment of GM00637 cells with the proteasome inhibitors, lactacystin or MG132, completely blocked H(2)O(2)-induced p21 downregulation, suggesting that H(2)O(2) treatment accelerated p21 degradation. Conversely, cotreatment of cells with a protein synthesis inhibitor, cycloheximide, and H(2)O(2) drastically shortened the half-life of p21. Moreover, p21 mRNA levels were not downregulated by treatment with proteasome or protein synthesis inhibitors. Taken together, our studies indicate that oxidative stress induces rapid, but reversible, downregulation of functional p21 by accelerating its protein turnover.


Subject(s)
Acetylcysteine/analogs & derivatives , Cyclins/metabolism , Cysteine Endopeptidases/physiology , Hydrogen Peroxide/pharmacology , Multienzyme Complexes/physiology , Oxidative Stress/physiology , Acetylcysteine/pharmacology , Cell Line/drug effects , Cell Line/metabolism , Cyclin-Dependent Kinase Inhibitor p21 , Cycloheximide/pharmacology , Doxorubicin/pharmacology , Fibroblasts/drug effects , Fibroblasts/metabolism , HeLa Cells/drug effects , HeLa Cells/metabolism , Humans , Leupeptins/pharmacology , Neoplasm Proteins/metabolism , Protease Inhibitors/pharmacology , Proteasome Endopeptidase Complex , Protein Synthesis Inhibitors/pharmacology , Reactive Oxygen Species/pharmacology
11.
Proc Natl Acad Sci U S A ; 103(31): 11579-83, 2006 Aug 01.
Article in English | MEDLINE | ID: mdl-16864777

ABSTRACT

Conditional N-Myc deletion limits the proliferation of granule neuron progenitors (GNPs), perturbs foliation, and leads to reduced cerebellar mass. We show that c-Myc mRNA levels increase in N-Myc-null GNPs and that simultaneous deletion of both c- and N-Myc exacerbates defective cerebellar development. Moreover, N-Myc loss has been shown to trigger the precocious expression of two cyclin-dependent kinase inhibitors, Kip1 and Ink4c, in the cerebellar primordium. We now further demonstrate that the engineered disruption of the Kip1 and Ink4c genes in N-Myc-null cerebella partially rescues GNP cell proliferation and cerebellar foliation. These results provide definitive genetic evidence that expression of N-Myc and concomitant down-regulation of Ink4c and Kip1 contribute to the proper development of the cerebellum.


Subject(s)
Cerebellum/embryology , Cyclin-Dependent Kinase Inhibitor p18/metabolism , Cyclin-Dependent Kinase Inhibitor p27/metabolism , Proto-Oncogene Proteins c-myc/metabolism , Animals , Cell Proliferation , Cerebellum/anatomy & histology , Cerebellum/growth & development , Cyclin-Dependent Kinase Inhibitor p18/genetics , Cyclin-Dependent Kinase Inhibitor p27/genetics , Gene Expression Regulation, Developmental , Mice , Mice, Knockout , Mice, Transgenic , Proto-Oncogene Proteins c-myc/genetics
12.
J Biol Chem ; 280(23): 22375-84, 2005 Jun 10.
Article in English | MEDLINE | ID: mdl-15811854

ABSTRACT

Polymerase delta interacting protein 38 (PDIP38) was identified as a human DNA polymerase (pol) delta interacting protein through a direct interaction with p50, the small subunit of human pol delta. PDIP38 was also found to interact with proliferating cell nuclear antigen, which suggested that it might play a role in vivo in the processes of DNA replication and DNA repair in the nucleus. In order to characterize further this novel protein, we have examined its subcellular localization by the use of immunochemical and cellular fractionation techniques. These studies show that PDIP38 is a novel mitochondrial protein and is localized mainly to the mitochondria. PDIP38 was shown to possess a functional mitochondrial targeting sequence that is located within the first 35 N-terminal amino acid residues. The mature PDIP38 protein is about 50 amino acid residues smaller than the full-length precursor PDIP38 protein, consistent with it being processed by cleavage of the mitochondrial targeting sequence during entry into the mitochondria. His-tagged mature PDIP38 inhibited pol delta activity in vitro and interacted with human papillomavirus 16 E7 oncoprotein, suggesting that PDIP38 might play a role in the pol delta-mediated viral DNA replication. Although the localization of PDIP38 to the mitochondria suggests that it serves functions within the mitochondria, we cannot eliminate the possibility that it may be involved in pol delta-mediated DNA replication or DNA repair under certain conditions such as viral infection.


Subject(s)
DNA Polymerase III/chemistry , Nuclear Proteins/physiology , Cell Line , Cell Line, Tumor , Cell Nucleus/metabolism , Cytoplasm/metabolism , DNA Polymerase III/metabolism , DNA Repair , DNA Replication , DNA-Directed DNA Polymerase/metabolism , Dose-Response Relationship, Drug , Electrophoresis, Polyacrylamide Gel , Glutathione Transferase/metabolism , HeLa Cells , Humans , Immunohistochemistry , Microscopy, Fluorescence , Mitochondria/metabolism , Nuclear Proteins/metabolism , Oncogene Proteins, Viral/metabolism , Papillomavirus E7 Proteins , Peptides/chemistry , Protein Biosynthesis , Protein Structure, Tertiary , Recombinant Fusion Proteins/metabolism , Subcellular Fractions/metabolism , Transfection
13.
Genes Dev ; 19(22): 2656-67, 2005 Nov 15.
Article in English | MEDLINE | ID: mdl-16260494

ABSTRACT

Recurrent genetic alterations in human medulloblastoma (MB) include mutations in the sonic hedgehog (SHH) signaling pathway and TP53 inactivation (approximately 25% and 10% of cases, respectively). However, mouse models of MB, regardless of their initiating lesions, generally depend upon p53 inactivation for rapid onset and high penetrance. The gene encoding the cyclin-dependent kinase inhibitor p18(Ink4c) is transiently expressed in mouse cerebellar granule neuronal precursor cells (GNPs) as they exit the cell division cycle and differentiate. Coinactivation of Ink4c and p53 provided cultured GNPs with an additive proliferative advantage, either in the presence or absence of Shh, and induced MB with low penetrance but with greatly increased incidence following postnatal irradiation. In contrast, mice lacking one or two functional Ink4c alleles and one copy of Patched (Ptc1) encoding the Shh receptor rapidly developed MBs that retained wild-type p53. In tumor cells purified from double heterozygotes, the wild-type Ptc1 allele, but not Ink4c, was inactivated. Therefore, when combined with Ptc1 mutation, Ink4c is haploinsufficient for tumor suppression. Methylation of INK4C (CDKN2C) was observed in four of 23 human MBs, and p18(INK4C) protein expression was extinguished in 14 of 73 cases. Hence, p18(INK4C) loss may contribute to MB formation in children.


Subject(s)
Cyclin-Dependent Kinase Inhibitor p18/physiology , Medulloblastoma/metabolism , Receptors, Cell Surface/physiology , Tumor Suppressor Protein p53/physiology , Animals , Cell Proliferation , Cells, Cultured , Cerebellum/metabolism , Cyclin-Dependent Kinase Inhibitor p18/deficiency , Cyclin-Dependent Kinase Inhibitor p18/genetics , Hedgehog Proteins , Humans , Medulloblastoma/etiology , Medulloblastoma/genetics , Medulloblastoma/pathology , Mice , Mice, Knockout , Patched Receptors , Patched-1 Receptor , Signal Transduction/physiology , Trans-Activators/physiology
14.
Med Sci Monit ; 8(8): BR293-300, 2002 Aug.
Article in English | MEDLINE | ID: mdl-12165733

ABSTRACT

BACKGROUND: Hypersecretion of the precursor of the lysosomal protease cathepsin D (procathepsin D) has been implicated in the invasive phenotype of human breast cancer. However, the mechanism of the abnormal secretion of procathepsin D remains unclear. Since the mannose 6-phosphate/insulin-like growth factor II receptor (M6P/IGF2R) plays a central role in the intracellular transport and endocytosis of M6P-containing lysosomal enzymes, a deficiency in functional M6P/IGF2R may underlie the hypersecretion of procathepsin D in invasive tumors. MATERIAL/METHODS: In the present study, we compared the profiles of cathepsin D and the M6P/IGF2R between the highly invasive MDA-MB-231 and the non-invasive MCF-7 cell lines. RESULTS: MDA-MB-231cells were confirmed to secrete a much larger proportion of procathepsin D into the medium than MCF-7 cells. Addition of M6P to the culture medium significantly altered the secretion of procathepsin D by MCF-7 cells, but had little effect on cathepsin D distribution in MDA-MB-231cells. Both the M6P-binding capacity and the endocytosis of exogenous M6P-bearing proteins in MDA-MB-231 cells were far less than those in MCF-7 cells. mRNA analysis indicated that the levels of the M6P/IGF2R mRNA in MDA-MB-231 cells were not lower but were even higher than that in MCF-7 cells. Sequence analysis indicated a difference in the 3'-untranslated region of M6P/IGF2R between the two cell lines, but no mutation in the M6P-binding domain of the receptor. CONCLUSIONS: The results suggest that a potential defect in a post-transcriptional process (e.g., translation) may exist during synthesis of the M6P/IGF2R in MDA-MB-231cells, leading to failure to express sufficient functional M6P/IGF2R and thereby resulting in the hypersecretion of procathepsin D.


Subject(s)
Breast Neoplasms/metabolism , Breast Neoplasms/pathology , Cathepsin D/metabolism , Enzyme Precursors/metabolism , Receptor, IGF Type 2/metabolism , 3' Untranslated Regions/genetics , Female , Humans , Mutation , RNA, Messenger/genetics , RNA, Messenger/metabolism , Receptor, IGF Type 2/genetics , Sequence Analysis, DNA , Tumor Cells, Cultured
15.
Exp Cell Res ; 294(1): 51-9, 2004 Mar 10.
Article in English | MEDLINE | ID: mdl-14980500

ABSTRACT

The Golgi apparatus undergoes extensive fragmentation during mitosis in animal cells. Protein kinases play a critical role during fragmentation of the Golgi apparatus. We reported here that Polo-like kinase 3 (Plk3) may be an important mediator during Golgi breakdown. Specifically, Plk3 was concentrated at the Golgi apparatus in HeLa and A549 cells during interphase. At the onset of mitosis, Plk3 signals disintegrated and redistributed in a manner similar to those of Golgi stacks. Nocodazole activated Plk3 kinase activity, correlating with redistribution of Plk3 signals and Golgi fragmentation. In addition, treatment with brefeldin A (BFA), a Golgi-specific poison, also resulted in disappearance of concentrated Plk3 signals. Plk3 interacted with giantin, a Golgi-specific protein. Expression of Plk3, but not the kinase-defective Plk3 (Plk3(K52R)), resulted in significant Golgi breakdown. Given its role in cell cycle progression, Plk3 may be a protein kinase involved in regulation of Golgi fragmentation during the cell cycle.


Subject(s)
Cell Cycle Proteins/analysis , Cell Cycle Proteins/physiology , Cell Cycle , Golgi Apparatus/enzymology , Protein Serine-Threonine Kinases/analysis , Protein Serine-Threonine Kinases/physiology , Brefeldin A/pharmacology , Cell Line , Golgi Apparatus/drug effects , Golgi Apparatus/ultrastructure , Golgi Matrix Proteins , HeLa Cells , Humans , Membrane Proteins/metabolism , Nocodazole/pharmacology , Tumor Suppressor Proteins
16.
Blood ; 103(4): 1278-85, 2004 Feb 15.
Article in English | MEDLINE | ID: mdl-14576056

ABSTRACT

The physiologic function of BUBR1, a key component of the spindle checkpoint, was examined by generating BUBR1-mutant mice. BUBR1(-/-) embryos failed to survive beyond day 8.5 in utero as a result of extensive apoptosis. Whereas BUBR1(+/-) blastocysts grew relatively normally in vitro, BUBR1(-/-) blastocysts exhibited impaired proliferation and atrophied. Adult BUBR1(+/-) mice manifested splenomegaly and abnormal megakaryopoiesis. BUBR1 haploinsufficiency resulted in an increase in the number of splenic megakaryocytes, which was correlated with an increase in megakaryocytic, but a decrease in erythroid, progenitors in bone marrow cells. RNA interference-mediated down-regulation of BUBR1 also caused an increase in polyploidy formation in murine embryonic fibroblast cells and enhanced megakaryopoiesis in bone marrow progenitor cells. However, enhanced megakaryopoiesis in BUBR1(+/-) mice was not correlated with a significant increase in platelets in peripheral blood, which was at least partly due to a defect in the formation of proplatelet-producing megakaryocytes. Together, these results indicate that BUBR1 is essential for early embryonic development and normal hematopoiesis.


Subject(s)
Erythropoiesis/physiology , Hematopoiesis, Extramedullary/physiology , Megakaryocytes/cytology , Megakaryocytes/physiology , Protein Kinases/genetics , Animals , Bone Marrow/embryology , Cell Cycle Proteins , Cells, Cultured , Female , Genotype , Hematopoietic Stem Cells/cytology , Hematopoietic Stem Cells/physiology , Mice , Mice, Knockout , Platelet Count , Pregnancy , Protein Kinases/metabolism , Protein Serine-Threonine Kinases , Splenomegaly/physiopathology
SELECTION OF CITATIONS
SEARCH DETAIL