Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 32
Filter
Add more filters










Publication year range
1.
Curr Oncol ; 21(2): 91-6, 2014 Apr.
Article in English | MEDLINE | ID: mdl-24764698

ABSTRACT

UNLABELLED: Repair of radiation-induced dna double-strand breaks is a key mechanism in cancer cell radio-resistance. The synthesized compound NU7026 specifically inhibits dna-dependent protein kinase (dna-pk) within the non-homologous end-joining repair mechanism. Earlier studies demonstrated increased radiosensitivity in dna-pk deficient cells compared with wild-type cells. In chronic leukemia cells, NU7026 appears to enhance the cytotoxic effect of chlorambucil. The radio-modifying effects of NU7026 on cell survival, cell cycle, apoptosis, and dna double-strand break repair have yet to be studied in gastric cancer cells. METHODS: The gastric cancer cell line N87 was treated with 0 Gy or 4 Gy in the presence of NU7026 at a dose range of 0-20 µmol/L. Clonogenic assays were used to assess cell survival after treatment. Cell-cycle distribution was analyzed using propidium iodide with fluorescence-activated cell sorting. Apoptosis was detected using annexin-V and propidium iodide with fluorescence-activated cell sorting. The γH2AX assay was used to measure dna double-strand breaks. RESULTS: Statistically significant increases in G2/M arrest were observed in N87 cells treated with radiation and NU7026 compared with those treated with radiation alone (p = 0.0004). Combined treatment also led to an increase in apoptosis (p = 0.01). At 24 hours, the γH2AX analysis revealed more dna double-strand breaks in N87 cells treated with radiation and NU7026 than in those treated with radiation alone (p = 0.04). Clonogenic assays demonstrated declining cell survival as both the radiation and the NU7026 dose increased. The dose enhancement factor at 0.1 survival fraction was 1.28 when N87 cells were treated with 4 Gy radiation and 5 µmol/L NU7026. CONCLUSIONS: In gastric cancer cells, NU7026 appears to enhance the cytotoxic effect of irradiation as assessed by clonogenic assays. This increased cytotoxicity might be the result of an increase in dna double-strand breaks resulting in G2/M cell arrest and possibly higher levels of apoptosis.

3.
Br J Cancer ; 105(9): 1342-5, 2011 Oct 25.
Article in English | MEDLINE | ID: mdl-21970880

ABSTRACT

BACKGROUND: Immunohistological assessment of Ki 67 expression is less expensive than Oncotype Dx, which is currently used to identify patients with lymph node-negative breast cancer, who will benefit from adjuvant chemotherapy. METHODS: The relationship of immunohistologically measured Ki 67 to Oncotype DX recurrence score (RS) was examined in 53 cases of T1-2 N0 M0 (oestrogen receptor-positive, HER2/neu negative) breast cancer. RESULTS: There was a strong linear correlation between Ki 67 value and the Oncotype Dx RS. All patients in the low Ki 67 group (Ki 67 of ≤ 10%) had Oncotype Dx RSs of low or intermediate risk. The vast majority of patients (93.8%) in the high-Ki 67 group (Ki 67 ≥ 25%) had oncotype RSs of high or intermediate risk. CONCLUSION: Ki 67 proliferation value is a major, but not the sole determinant of Oncotype Dx score.


Subject(s)
Breast Neoplasms/genetics , Breast Neoplasms/metabolism , Gene Expression Profiling , Ki-67 Antigen/metabolism , Recurrence , Biomarkers, Tumor/metabolism , Breast Neoplasms/drug therapy , Chemotherapy, Adjuvant , Female , Humans , Predictive Value of Tests , Prognosis , Real-Time Polymerase Chain Reaction/methods
5.
J Chemother ; 23(1): 32-5, 2011 Feb.
Article in English | MEDLINE | ID: mdl-21482492

ABSTRACT

The combination of vinorelbine and trastuzumab (VH) is highly active and well tolerated in patients with metastatic HER2+ breast cancer. We assessed the efficacy and tolerability of VH as an alternative adjuvant treatment for patients with localized breast cancer refusing or ineligible for standard adjuvant trastuzumab-based chemotherapy. Twenty-eight patients with stage I-III breast cancer were treated only with VH as preoperative or postoperative chemotherapy. Fourteen patients received VH as adjuvant treatment for pT1a-b pN0 or eR+ pT1c pN0 cancers. VH was well tolerated, the only grade 3-4 toxicity being neutropenia with 2 cases of febrile neutropenia. At a median follow-up of 39 months, no breast cancer relapses were documented; moreover, overall and disease-free survival was 96.4%. In summary, our results indicate that VH is effective and well tolerated. VH should be prospectively tested as adjuvant treatment for pN0 pT1a-b breast cancer patients for which no standard treatment is well defined.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/administration & dosage , Breast Neoplasms/drug therapy , Receptor, ErbB-2/biosynthesis , Antibodies, Monoclonal/administration & dosage , Antibodies, Monoclonal/adverse effects , Antibodies, Monoclonal, Humanized , Antineoplastic Combined Chemotherapy Protocols/adverse effects , Breast Neoplasms/blood , Breast Neoplasms/enzymology , Breast Neoplasms/pathology , Chemotherapy, Adjuvant , Disease-Free Survival , Drug Tolerance , Female , Humans , Middle Aged , Neoplasm Staging , Neutropenia/chemically induced , Retrospective Studies , Trastuzumab , Vinblastine/administration & dosage , Vinblastine/adverse effects , Vinblastine/analogs & derivatives , Vinorelbine
7.
Oncogene ; 25(3): 349-58, 2006 Jan 19.
Article in English | MEDLINE | ID: mdl-16186807

ABSTRACT

Treatments for hematological malignancies have improved considerably over the past decade, but the growing therapeutic arsenal has not benefited adult T-cell leukemia (ATL) patients. Oncolytic viruses such as vesicular stomatitis virus (VSV) have recently emerged as a potential treatment of solid tumors and leukemias in vitro and in vivo. In the current study, we investigated the ability of VSV to lyse primary human T-lymphotropic virus type 1 (HTLV-1)-infected T-lymphocytes from patients with ATL. Ex vivo primary ATL cells were permissive for VSV and underwent rapid oncolysis in a time-dependent manner. Importantly, VSV infection showed neither viral replication nor oncolysis in HTLV-1-infected, nonleukemic cells from patients with HTLV-1-associated myelopathy/tropical spastic paraparesis (HAM/TSP), and in naive CD4(+) T-lymphocytes from normal individuals or in ex vivo cell samples from patients with chronic lymphocytic leukemia (CLL). Interestingly, activation of primary CD4(+) T-lymphocytes with anti-CD3/CD28 monoclonal antibody, and specifically with anti-CD3, was sufficient to induce limited viral replication and oncolysis. However, at a similar level of T-cell activation, VSV replication was increased fourfold in ATL cells compared to activated CD4(+) T-lymphocytes, emphasizing the concept that VSV targets genetic defects unique to tumor cells to facilitate its replication. In conclusion, our findings provide the first essential information for the development of a VSV-based treatment for ATL.


Subject(s)
Leukemia, T-Cell/therapy , Leukemia, T-Cell/virology , Vesicular stomatitis Indiana virus/physiology , Animals , CD4-Positive T-Lymphocytes/virology , Cell Death , Cell Line , Cell Line, Tumor , Cricetinae , Humans , Lymphocyte Activation , Virus Replication
8.
Leukemia ; 18(3): 409-14, 2004 Mar.
Article in English | MEDLINE | ID: mdl-14712290

ABSTRACT

The effect of imatinib on chlorambucil (CLB) cytotoxicity in chronic lymphocytic leukemia (CLL) lymphocytes was examined in vitro. Imatinib sensitizes the WSU and I83 human CLL cell lines, 10- and two-fold, respectively, to CLB. Furthermore, in primary cultures of malignant B-lymphocytes obtained from 12 patients with CLL (seven patients were untreated and five treated with CLB), imatinib synergistically sensitized these lymphocytes from two- to 20-fold to CLB. This synergistic effect was observed at concentrations of imatinib (

Subject(s)
Antineoplastic Combined Chemotherapy Protocols/pharmacology , Chlorambucil/pharmacology , Drug Resistance, Neoplasm , Enzyme Inhibitors/pharmacology , Leukemia, Lymphocytic, Chronic, B-Cell/drug therapy , Piperazines/pharmacology , Pyrimidines/pharmacology , Apoptosis/drug effects , Benzamides , Cell Cycle/drug effects , DNA-Binding Proteins/metabolism , Drug Synergism , Humans , Imatinib Mesylate , Leukemia, Lymphocytic, Chronic, B-Cell/enzymology , Leukemia, Lymphocytic, Chronic, B-Cell/pathology , Phosphorylation , Protein-Tyrosine Kinases/antagonists & inhibitors , Proto-Oncogene Proteins c-abl/metabolism , Rad51 Recombinase , Tumor Cells, Cultured/drug effects , Tumor Cells, Cultured/metabolism , Tumor Cells, Cultured/pathology
9.
J Natl Cancer Inst ; 93(19): 1473-8, 2001 Oct 03.
Article in English | MEDLINE | ID: mdl-11584063

ABSTRACT

BACKGROUND: The generation of DNA interstrand cross-links is thought to be important in the cytotoxicity of nitrogen mustard alkylating agents, such as melphalan, which have antitumor activity. Cell lines with mutations in recombinational repair pathways are hypersensitive to nitrogen mustards. Thus, resistance to melphalan may require accelerated DNA repair by either recombinational repair mechanisms involving Rad51-related proteins (including x-ray repair cross-complementing proteins Xrcc2, Xrcc3, and Rad52) or by nonhomologous endjoining involving DNA-dependent protein kinase (DNA-PK) and Ku proteins. We investigated the role of DNA repair in melphalan resistance in epithelial tumor cell lines. METHODS: Melphalan cytotoxicity was determined in 14 epithelial tumor cell lines by use of the sulforhodamine assay. Homologous recombinational repair involving Rad51-related proteins was investigated by determining the levels of Rad51, Rad52, and Xrcc3 proteins and the density of nuclear melphalan-induced Rad51 foci, which represent sites of homologous recombinational repair. Nonhomologous endjoining was investigated by determining the levels of Ku70 and Ku86 proteins and DNA-PK activity. Linear regression analysis was used to analyze correlations between the various protein levels, DNA-PK activity, or Rad51 foci formation and melphalan cytotoxicity. All statistical tests were two-sided. RESULTS: Melphalan resistance was correlated with Xrcc3 levels (r =.587; P =.027) and the density of melphalan-induced Rad51 foci (r =.848; P =.008). We found no correlation between melphalan resistance and Rad51, Rad52, or Ku protein levels or DNA-PK activity. CONCLUSION: Correlations of melphalan resistance in epithelial tumor cell lines with Xrcc3 protein levels and melphalan-induced Rad51 foci density suggest that homologous recombinational repair is involved in resistance to this nitrogen mustard.


Subject(s)
Antigens, Nuclear , Antineoplastic Agents, Alkylating/pharmacology , Cross-Linking Reagents/pharmacology , DNA Helicases , DNA Repair , DNA, Neoplasm/drug effects , Drug Resistance, Neoplasm/genetics , Melphalan/pharmacology , Neoplasm Proteins/physiology , Recombination, Genetic , Blotting, Western , DNA, Neoplasm/metabolism , DNA-Activated Protein Kinase , DNA-Binding Proteins/genetics , DNA-Binding Proteins/physiology , Fluorescent Antibody Technique, Indirect , Humans , Ku Autoantigen , Microscopy, Confocal , Neoplasm Proteins/genetics , Nuclear Proteins/genetics , Nuclear Proteins/physiology , Protein Serine-Threonine Kinases/analysis , Rad51 Recombinase , Sequence Homology, Nucleic Acid , Tumor Cells, Cultured/drug effects
10.
Clin Cancer Res ; 7(3): 454-61, 2001 Mar.
Article in English | MEDLINE | ID: mdl-11297233

ABSTRACT

Various mechanisms have been implicated in nitrogen mustard drug resistance. The role of these mechanisms in the development of chlorambucil drug resistance in chronic lymphocytic leukemia (CLL) is discussed. We review these mechanisms with emphasis on the emerging role of DNA repair, and specifically, recombinational repair. Inhibition of these repair processes may lead to new therapies, not only in CLL, but in other malignancies as well.


Subject(s)
Antineoplastic Agents, Alkylating/pharmacology , Chlorambucil/pharmacology , DNA Repair , Drug Resistance, Neoplasm , Leukemia, Lymphocytic, Chronic, B-Cell/drug therapy , Leukemia, Lymphocytic, Chronic, B-Cell/metabolism , Mechlorethamine/pharmacology , Apoptosis , Biological Transport , Cross-Linking Reagents/pharmacology , Genes, p53/genetics , Glutathione/genetics , Glutathione Transferase/genetics , Models, Genetic , Mutation , Recombination, Genetic
11.
J Cell Biol ; 155(7): 1275-85, 2001 Dec 24.
Article in English | MEDLINE | ID: mdl-11756477

ABSTRACT

Developmental sympathetic neuron death is determined by functional interactions between the TrkA/NGF receptor and the p75 neurotrophin receptor (p75NTR). A key question is whether p75NTR promotes apoptosis by directly inhibiting or modulating TrkA activity, or by stimulating cell death independently of TrkA. Here we provide evidence for the latter model. Specifically, experiments presented here demonstrate that the presence or absence of p75NTR does not alter Trk activity or NGF- and NT-3-mediated downstream survival signaling in primary neurons. Crosses of p75NTR-/- and TrkA-/- mice indicate that the coincident absence of p75NTR substantially rescues TrkA-/- sympathetic neurons from developmental death in vivo. Thus, p75NTR induces death regardless of the presence or absence of TrkA expression. These data therefore support a model where developing sympathetic neurons are "destined to die" by an ongoing p75NTR-mediated apoptotic signal, and one of the major ways that TrkA promotes neuronal survival is by silencing this ongoing death signal.


Subject(s)
Cell Survival/physiology , Neurons/cytology , Oncogene Proteins/metabolism , Receptors, Nerve Growth Factor/metabolism , Animals , Cells, Cultured , Mice , Mice, Inbred BALB C , Neurons/metabolism , Receptor, Nerve Growth Factor , Signal Transduction
12.
J Neurosci ; 20(20): 7648-56, 2000 Oct 15.
Article in English | MEDLINE | ID: mdl-11027225

ABSTRACT

The retinoblastoma tumor suppressor protein (pRb) family is essential for cortical progenitors to exit the cell cycle and survive. In this report, we test the hypothesis that pRb collaborates with basic helix-loop-helix (bHLH) transcription factors to regulate cortical neurogenesis, taking advantage of the naturally occurring dominant-inhibitory HLH protein Id2. Overexpression of Id2 in cortical progenitors completely inhibited the induction of neuron-specific genes and led to apoptosis, presumably as a consequence of conflicting differentiation signals. Both of these phenotypes were rescued by coexpression of a constitutively activated pRb mutant. In contrast, Id2 overexpression in postmitotic cortical neurons affected neither neuronal gene expression nor survival. Thus, pRb collaborates with HLHs to ensure the coordinate induction of terminal mitosis and neuronal gene expression as cortical progenitors become neurons.


Subject(s)
Cerebral Cortex/metabolism , Helix-Loop-Helix Motifs/physiology , Neurons/metabolism , Repressor Proteins , Retinoblastoma Protein/metabolism , Transcription Factors/metabolism , Adenoviridae/genetics , Animals , Apoptosis , Cell Differentiation/physiology , Cell Survival/physiology , Cells, Cultured , Cerebral Cortex/cytology , Cerebral Cortex/embryology , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , DNA-Binding Proteins/pharmacology , Gene Expression/drug effects , Genetic Vectors/genetics , Genetic Vectors/metabolism , In Situ Nick-End Labeling , Inhibitor of Differentiation Protein 2 , Mice , Neurons/cytology , Stem Cells/cytology , Stem Cells/metabolism , Transfection , Tubulin/metabolism
13.
J Neurosci ; 19(22): 9716-27, 1999 Nov 15.
Article in English | MEDLINE | ID: mdl-10559381

ABSTRACT

In this report, we examine how the Ras protein regulates neuronal survival, focusing on sympathetic neurons. Adenovirus-expressed constitutively activated Ras (RasV12) enhanced survival and the phosphorylation of Akt (protein kinase B) and MAP kinase (MAPK), two targets of Ras activity. Functional inhibition of endogenous Ras by adenovirus-expressed dominant-inhibitory Ras (N17Ras) decreased nerve growth factor (NGF)-dependent survival and both Akt and MAPK phosphorylation as well. To determine the signaling pathways through which Ras mediates survival, we used Ras effector mutants and pharmacological inhibitors that selectively suppress phosphatidylinositol 3-kinase (PI3-K)/Akt or MAP kinase kinase (MEK)/MAPK pathways. The Ras effector mutant Ras(V12)Y40C, which selectively stimulates PI3-K and Akt, rescued survival in the absence of NGF, and the PI3-K inhibitor LY 294002 inhibited both Ras- and NGF-dependent survival. Ras(V12)T(35)S, which activates MEK/MAPK but not PI3-K/Akt, was less effective at rescuing survival, whereas the MEK inhibitor PD 098059 also partially suppressed Ras-dependent survival. To investigate the mechanisms by which Ras suppresses neuronal death, we examined whether Ras functions by inhibiting the proapoptotic p53 pathway (Jun-N-terminal kinase/p53/BAX) that is necessary for neuronal death after NGF withdrawal and p75NTR activation. We found that RasV12 suppressed c-jun, BAX, and p53 levels, whereas inhibition of NGF-induced Ras-survival activity via N17Ras increased the levels of these proteins. Furthermore, the E1B55K protein, which suppresses p53 activity, blocked N17Ras-induced neuronal death. Together, these results indicate that Ras is, in part, both necessary and sufficient for survival of sympathetic neurons and that this effect is mediated by activation of both the PI3-K- and MEK-signaling cascades, which in turn suppress a proapoptotic p53 pathway.


Subject(s)
Apoptosis/physiology , Cell Survival/physiology , Neurons/cytology , Neurons/physiology , Protein Serine-Threonine Kinases , Signal Transduction/physiology , Superior Cervical Ganglion/cytology , Tumor Suppressor Protein p53/metabolism , ras Proteins/metabolism , Adenoviridae , Animals , Animals, Newborn , Cells, Cultured , Mitogen-Activated Protein Kinase Kinases/metabolism , Mitogen-Activated Protein Kinases/metabolism , Nerve Growth Factors/pharmacology , Neurons/drug effects , Phosphatidylinositol 3-Kinases/metabolism , Phosphorylation , Proto-Oncogene Proteins/metabolism , Proto-Oncogene Proteins c-akt , Rats , Recombinant Proteins/metabolism , Signal Transduction/drug effects , Superior Cervical Ganglion/physiology , Transfection
14.
Learn Mem ; 6(3): 216-31, 1999.
Article in English | MEDLINE | ID: mdl-10492004

ABSTRACT

In this paper we have investigated the hypothesis that neural activity causes rapid activation of TrkB neurotrophin receptors in the adult mammalian CNS. These studies demonstrate that kainic acid-induced seizures led to a rapid and transient activation of TrkB receptors in the cortex. Subcellular fractionation demonstrated that these activated Trk receptors were preferentially enriched in the synaptosomal membrane fraction that also contained postsynaptic glutamate receptors. The fast activation of synaptic TrkB receptors could be duplicated in isolated cortical synaptosomes with KCl, presumably as a consequence of depolarization-induced BDNF release. Importantly, TrkB activation was also observed following pharmacological activation of brain-stem noradrenergic neurons, which synthesize and anterogradely transport BDNF; treatment with yohimbine led to activation of cortical TrkB receptors within 30 min. Pharmacological blockade of the postsynaptic alpha1-adrenergic receptors with prazosin only partially inhibited this effect, suggesting that the TrkB activation was partially due to a direct effect on postsynaptic cortical neurons. Together, these data support the hypothesis that activity causes release of BDNF from presynaptic terminals, resulting in a rapid activation of postsynaptic TrkB receptors. This activity-dependent TrkB activation could play a major role in morphological growth and remodelling in both the developing and mature nervous systems.


Subject(s)
Central Nervous System/physiology , Receptor Protein-Tyrosine Kinases/physiology , Receptors, Nerve Growth Factor/physiology , Animals , Blotting, Western , Brain Stem/cytology , Brain Stem/physiology , Brain-Derived Neurotrophic Factor/physiology , Central Nervous System/cytology , Central Nervous System/ultrastructure , Cerebral Cortex/physiology , Excitatory Amino Acid Agonists/pharmacology , Excitatory Postsynaptic Potentials/physiology , Female , Kainic Acid/pharmacology , Neurons/physiology , Neurons/ultrastructure , Phosphorylation , Precipitin Tests , Rats , Rats, Sprague-Dawley , Receptor Protein-Tyrosine Kinases/drug effects , Receptor, Ciliary Neurotrophic Factor , Receptors, Nerve Growth Factor/drug effects , Seizures/chemically induced , Seizures/physiopathology , Subcellular Fractions/metabolism , Synapses/physiology , Synaptosomes/physiology , Synaptosomes/ultrastructure
15.
J Neurosci ; 19(13): 5393-408, 1999 Jul 01.
Article in English | MEDLINE | ID: mdl-10377349

ABSTRACT

In this report, we provide evidence that NGF and BDNF have functionally antagonistic actions on sympathetic neuron growth and target innervation, with NGF acting via TrkA to promote growth and BDNF via p75NTR to inhibit growth. Specifically, in cultured sympathetic neurons that themselves synthesize BDNF, exogenous BDNF inhibits and function-blocking BDNF antibodies enhance process outgrowth. Both exogenous and autocrine BDNF mediate this effect via p75NTR because (1) BDNF does not inhibit growth of neurons lacking p75NTR, (2) function-blocking p75NTR antibodies enhance NGF-mediated growth, and (3) p75NTR-/- sympathetic neurons grow more robustly in response to NGF than do their wild-type counterparts. To determine the physiological relevance of this functional antagonism, we examined the pineal gland, a well defined sympathetic target organ. BDNF is present in the pineal gland during target innervation, and incoming sympathetic axons are p75NTR positive. Moreover, the pineal glands of BDNF+/- and BDNF-/- mice are hyperinnervated with sympathetic fibers, and tyrosine hydroxylase (TH) levels are elevated. Increased tyrosine hydroxylase is also observed in the BDNF+/- carotid artery, another sympathetic neuron target. Thus, BDNF, made by sympathetic neurons and/or their target organs, acts via p75NTR to antagonize NGF-mediated growth and target innervation, suggesting that sympathetic target innervation is determined by the balance of positively and negatively acting neurotrophins present in developing and potentially mature targets.


Subject(s)
Neurons/cytology , Pineal Gland/innervation , Receptor, trkA/metabolism , Receptors, Nerve Growth Factor/metabolism , Sympathetic Nervous System/cytology , Animals , Antibodies/pharmacology , Autocrine Communication , Axons/drug effects , Axons/metabolism , Brain-Derived Neurotrophic Factor/antagonists & inhibitors , Brain-Derived Neurotrophic Factor/genetics , Brain-Derived Neurotrophic Factor/metabolism , Brain-Derived Neurotrophic Factor/pharmacology , Carotid Arteries/innervation , Carotid Arteries/metabolism , Cell Division/drug effects , Cell Survival/drug effects , Cells, Cultured , Gene Deletion , Genotype , Mice , Mice, Knockout , Nerve Growth Factors/antagonists & inhibitors , Nerve Growth Factors/pharmacology , Neurites/drug effects , Neurons/drug effects , Neurons/metabolism , Pineal Gland/metabolism , Receptor, Nerve Growth Factor , Receptors, Nerve Growth Factor/genetics , Receptors, Nerve Growth Factor/immunology , Signal Transduction/drug effects , Tyrosine 3-Monooxygenase/analysis
16.
J Cell Biol ; 143(6): 1691-703, 1998 Dec 14.
Article in English | MEDLINE | ID: mdl-9852160

ABSTRACT

Naturally occurring sympathetic neuron death is the result of two apoptotic signaling events: one normally suppressed by NGF/TrkA survival signals, and a second activated by the p75 neurotrophin receptor. Here we demonstrate that the p53 tumor suppressor protein, likely as induced by the MEKK-JNK pathway, is an essential component of both of these apoptotic signaling cascades. In cultured neonatal sympathetic neurons, p53 protein levels are elevated in response to both NGF withdrawal and p75NTR activation. NGF withdrawal also results in elevation of a known p53 target, the apoptotic protein Bax. Functional ablation of p53 using the adenovirus E1B55K protein inhibits neuronal apoptosis as induced by either NGF withdrawal or p75 activation. Direct stimulation of the MEKK-JNK pathway using activated MEKK1 has similar effects; p53 and Bax are increased and the subsequent neuronal apoptosis can be rescued by E1B55K. Expression of p53 in sympathetic neurons indicates that p53 functions downstream of JNK and upstream of Bax. Finally, when p53 levels are reduced or absent in p53+/- or p53-/- mice, naturally occurring sympathetic neuron death is inhibited. Thus, p53 is an essential common component of two receptor-mediated signal transduction cascades that converge on the MEKK-JNK pathway to regulate the developmental death of sympathetic neurons.


Subject(s)
Apoptosis/physiology , MAP Kinase Kinase Kinase 1 , Mitogen-Activated Protein Kinases , Neurons/cytology , Neurons/physiology , Proto-Oncogene Proteins c-bcl-2 , Proto-Oncogene Proteins/physiology , Receptor Protein-Tyrosine Kinases/physiology , Receptors, Nerve Growth Factor/physiology , Superior Cervical Ganglion/cytology , Tumor Suppressor Protein p53/physiology , Animals , Apoptosis/drug effects , Calcium-Calmodulin-Dependent Protein Kinases/metabolism , Cell Death , Cells, Cultured , JNK Mitogen-Activated Protein Kinases , Mice , Mice, Knockout , Models, Neurological , Nerve Growth Factors/pharmacology , Nerve Growth Factors/physiology , Neurons/drug effects , Protein Serine-Threonine Kinases/metabolism , Proto-Oncogene Proteins/metabolism , Rats , Receptor, Nerve Growth Factor , Receptor, trkA , Signal Transduction , Superior Cervical Ganglion/physiology , Tumor Suppressor Protein p53/deficiency , Tumor Suppressor Protein p53/genetics , bcl-2-Associated X Protein
17.
Endocrine ; 8(3): 231-40, 1998 Jun.
Article in English | MEDLINE | ID: mdl-9741827

ABSTRACT

The biosynthesis of met-enkephalin in human pituitary and human pituitary adenomas is still not well known. In this work, we studied the processing of proenkephalin-derived peptides in postmortem human pituitary (PMHP), ACTH-producing adenomas (ACTH-PA), nonfunctioning adenomas (NFA), and GH-producing adenomas (GH-PA). ACTH-PA contained at least 10 times more proenkephalin-derived peptides than PMHP, NFA,and GH-PA. Proenkephalin processing was different in the four tested tissues. In ACTH-PA, proenkephalin was processed to high-, intermediate-, and low-mol-wt products. The highest met-enkephalin-containing peptides levels corresponded to intermediate and low-mol-wt materials, although met-enkephalinArg-Phe and synenkephalin immunoreactivity appeared only in high-mol-wt peptides. In PMHP and NFA, met-enkephalin-Arg-Phe immunoreactivity was detected in intermediate- and low-mol-wt materials, and it was absent in GH-PA. Immunoblotting of ACTH-PA showed that met-enkephalin-Arg-Phe immunoreactivity corresponded to peptides of 44, 32-30, 27, and 17 kDa. The 32-30 and 17-kDa molecules were localized in the nuclear fraction where they were extracted after enzymatic digestion with DNase I. Plasmatic met-enkephalin levels did not increase in patients with Cushing's disease, suggesting that the pentapeptide stored in ACTH-PA was not released to the general circulation. In conclusion, we demonstrated that only ACTH-PA contained high levels of proenkephalin peptides, which were stored in cytoplasm organelles and in the nucleus, probably bound to chromatin. These results suggest an adenoma-specific physiological role of proenkephalin products.


Subject(s)
Adenoma/metabolism , Adrenocorticotropic Hormone/metabolism , Enkephalins/metabolism , Pituitary Neoplasms/metabolism , Protein Precursors/metabolism , Adult , Aged , Carboxypeptidase B , Carboxypeptidases/metabolism , Chromatography, Gel , Chromatography, High Pressure Liquid , Deoxyribonuclease I/metabolism , Enkephalin, Methionine/metabolism , Female , Human Growth Hormone/metabolism , Humans , In Vitro Techniques , Male , Middle Aged , Molecular Weight , Pituitary Gland/metabolism , Protein Processing, Post-Translational , Trypsin/metabolism
18.
J Neurosci ; 18(8): 2808-21, 1998 Apr 15.
Article in English | MEDLINE | ID: mdl-9525998

ABSTRACT

In this report, we have tested the hypothesis that brain-derived neurotrophic factor (BDNF) is an anterograde neurotrophic factor in the CNS and have focused on central noradrenergic neurons that synthesize BDNF. Double-label immunocytochemistry for BDNF and dopamine-beta-hydroxylase (DBH), a marker for noradrenergic neurons, demonstrated that BDNF is partially localized to noradrenergic nerve fibers and terminals in the adult rat brain. To test the functional importance of this anterograde BDNF, we analyzed transgenic mice carrying a DBH-BDNF minigene. Increased synthesis of BDNF in noradrenergic neurons of DBH-BDNF mice caused elevated TrkB tyrosine kinase activation throughout postnatal life in the neocortex, a noradrenergic target region. This afferently regulated increase in TrkB receptor activity led to long-lasting alterations in cortical morphology. To determine whether noradrenergic neuron-expressed BDNF also anterogradely regulated neuronal survival, we examined a second noradrenergic target, neonatal facial motoneurons. One week after axotomy, 72% of facial motoneurons were lost in control animals, whereas only 30-35% were lost in DBH-BDNF transgenic mice. Altogether, these results indicate that BDNF is anterogradely transported to fibers and terminals of noradrenergic neurons, that anterogradely secreted BDNF causes activation of TrkB in target regions, and that this secretion has functional consequences for target neuron survival and differentiation. This presynaptic secretion of BDNF may provide a cellular mechanism for modulating neural circuitry, in either the developing or mature nervous system.


Subject(s)
Brain-Derived Neurotrophic Factor/genetics , Brain-Derived Neurotrophic Factor/metabolism , Cerebral Cortex/growth & development , Neurons, Afferent/cytology , Age Factors , Animals , Animals, Newborn , Axotomy , Cell Death/physiology , Cell Division/physiology , Cell Survival/physiology , Cerebral Cortex/cytology , Dopamine beta-Hydroxylase/metabolism , Facial Nerve/cytology , Female , Gene Expression Regulation, Developmental/physiology , Heterozygote , Male , Mice , Mice, Transgenic , Motor Neurons/cytology , Nerve Regeneration/physiology , Neurons, Afferent/enzymology , Neurons, Afferent/ultrastructure , Norepinephrine/physiology , Presynaptic Terminals/chemistry , Presynaptic Terminals/enzymology
19.
J Cell Biol ; 140(4): 911-23, 1998 Feb 23.
Article in English | MEDLINE | ID: mdl-9472042

ABSTRACT

To determine whether the p75 neurotrophin receptor (p75NTR) plays a role in naturally occurring neuronal death, we examined neonatal sympathetic neurons that express both the TrkA tyrosine kinase receptor and p75NTR. When sympathetic neuron survival is maintained with low quantities of NGF or KCl, the neurotrophin brain-derived neurotrophic factor (BDNF), which does not activate Trk receptors on sympathetic neurons, causes neuronal apoptosis and increased phosphorylation of c-jun. Function-blocking antibody studies indicate that this apoptosis is due to BDNF-mediated activation of p75NTR. To determine the physiological relevance of these culture findings, we examined sympathetic neurons in BDNF-/- and p75NTR-/- mice. In BDNF-/- mice, sympathetic neuron number is increased relative to BDNF+/+ littermates, and in p75NTR-/- mice, the normal period of sympathetic neuron death does not occur, with neuronal attrition occurring later in life. This deficit in apoptosis is intrinsic to sympathetic neurons, since cultured p75NTR-/- neurons die more slowly than do their wild-type counterparts. Together, these data indicate that p75NTR can signal to mediate apoptosis, and that this mechanism is essential for naturally occurring sympathetic neuron death.


Subject(s)
Apoptosis/physiology , Receptors, Nerve Growth Factor/physiology , Sympathetic Nervous System/physiology , Animals , Brain-Derived Neurotrophic Factor/physiology , Cell Count , Cell Death/drug effects , Cell Death/physiology , Cell Size , Cell Survival/drug effects , Cell Survival/physiology , Mice , Mice, Inbred BALB C , Mice, Inbred Strains , Mice, Mutant Strains , Nerve Growth Factors/pharmacology , Nerve Tissue Proteins/metabolism , Neurons/physiology , Phosphorylation , Protein Binding , Proto-Oncogene Proteins c-jun/metabolism , Rats , Receptor, Nerve Growth Factor , Receptor, trkA/metabolism , Sympathetic Nervous System/cytology , Time Factors
20.
Neuropeptides ; 32(4): 351-9, 1998 Aug.
Article in English | MEDLINE | ID: mdl-10102680

ABSTRACT

Proenkephalin peptides produced by endocrine and nervous tissues are involved in stress-induced immunosuppression. However, the role of peptides produced by immune cells remains unknown. The present study examines the effect of acute and chronic foot-shock stress on proenkephalin peptide content in bone marrow (BMMC), thymus (TMC), and spleen (SMC) rat mononuclear cells. Proenkephalin was not processed to met-enkephalin in BMMC, while in TMC and SMC met-enkephalin represented 10% and 26% of total met-enkephalin-containing peptides, respectively. Naive rats receiving a stress stimulus showed a significant decrease of proenkephalin derived peptides in BMMC, TMC and SMC. However, in chronically stressed rats that already showed basal low peptide levels, a new stress stimulus produced a differential response in each immune tissue. That is, in BMMC peptide levels reached control rats values; in TMC remained unmodified; and in SMC, although precursors content increased, met-enkephalin levels were even lower than those observed in acutely stressed rats. Free synenkephalin content paralleled met-enkephalin changes in SMC of acutely and chronically stressed rats. The in vitro release of met-enkephalin and free synenkephalin increased in SMC of stressed rats. Met-enkephalin produced in SMC and partially processed proenkephalin peptides detected in BMMC, were only found in macrophages. However, met-enkephalin only appeared in bone marrow macrophages after at least 4 h of cell culture. Altogether, these results suggest that a stress stimulus induced proenkephalin peptide release from immune tissue macrophages. The differential response observed in chronically stressed rats suggest an alternative activation of heterogeneous proenkephalin-storing macrophage subpopulations.


Subject(s)
Bone Marrow Cells/metabolism , Enkephalins/metabolism , Leukocytes, Mononuclear/metabolism , Protein Precursors/metabolism , Spleen/metabolism , Stress, Physiological/metabolism , Thymus Gland/metabolism , Animals , Electroshock , Enkephalin, Methionine/metabolism , Macrophages/metabolism , Male , Rats , Rats, Wistar , Spleen/cytology , Thymus Gland/cytology
SELECTION OF CITATIONS
SEARCH DETAIL
...