Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 33
Filter
1.
Int J Obes (Lond) ; 42(3): 353-362, 2018 03.
Article in English | MEDLINE | ID: mdl-29151592

ABSTRACT

BACKGROUND/OBJECTIVES: Inter-individual variability in weight loss during obesity treatment is complex and poorly understood. Here we use whole body and tissue approaches to investigate fuel oxidation characteristics in skeletal muscle fibers, cells and distinct circulating protein biomarkers before and after a high fat meal (HFM) challenge in those who lost the most (obese diet-sensitive; ODS) vs the least (obese diet-resistant; ODR) amount of weight in a highly controlled weight management program. SUBJECTS/METHODS: In 20 weight stable-matched ODS and ODR women who previously completed a standardized clinical weight loss program, we analyzed whole-body energetics and metabolic parameters in vastus lateralis biopsies and plasma samples that were obtained in the fasting state and 6 h after a defined HFM, equivalent to 35% of total daily energy requirements. RESULTS: At baseline (fasting) and post-HFM, muscle fatty acid oxidation and maximal oxidative phosphorylation were significantly greater in ODS vs ODR, as was reactive oxygen species emission. Plasma proteomics of 1130 proteins pre and 1, 2, 5 and 6 h after the HFM demonstrated distinct group and interaction differences. Group differences identified S-formyl glutathione hydratase, heat shock 70 kDA protein 1A/B (HSP72), and eukaryotic translation initiation factor 5 (eIF5) to be higher in ODS vs ODR. Group-time differences included aryl hydrocarbon interacting protein (AIP), peptidylpropyl isomerase D (PPID) and tyrosine protein-kinase Fgr, which increased in ODR vs ODS over time. HSP72 levels correlated with muscle oxidation and citrate synthase activity. These proteins circulate in exosomes; exosomes isolated from ODS plasma increased resting, leak and maximal respiration rates in C2C12 myotubes by 58%, 21% and 51%, respectively, vs those isolated from ODR plasma. CONCLUSIONS: Findings demonstrate distinct muscle metabolism and plasma proteomics in fasting and post-HFM states corresponding in diet-sensitive vs diet-resistant obese women.


Subject(s)
Blood Proteins/metabolism , Muscle Fibers, Skeletal/metabolism , Obesity , Proteome/metabolism , Biomarkers/blood , Blood Proteins/analysis , Case-Control Studies , Diet , Exosomes/metabolism , Female , Humans , Middle Aged , Muscle, Skeletal/metabolism , Muscle, Skeletal/physiopathology , Obesity/blood , Obesity/diet therapy , Obesity/epidemiology , Obesity/metabolism , Proteome/analysis , Treatment Failure
2.
Dev Biol ; 384(1): 101-13, 2013 Dec 01.
Article in English | MEDLINE | ID: mdl-24056077

ABSTRACT

Pocket proteins (pRb, p107 and p130) are well studied in their role of regulating cell cycle progression. Increasing evidence suggests that these proteins also control early differentiation and even later stages of cell maturation, such as migration. However, pocket proteins also regulate apoptosis, and many of the developmental defects in knock out models have been attributed to increased cell death. Here, we eliminate ectopic apoptosis in the developing brain through the deletion of Bax, and show that pocket proteins are required for radial migration independent of their role in cell death regulation. Following loss of pRb and p107, a population of cortical neurons fails to pass through the intermediate zone into the cortical plate. Importantly, these neurons are born at the appropriate time and this migration defect cannot be rescued by eliminating ectopic cell death. In addition, we show that pRb and p107 regulate radial migration through a cell autonomous mechanism since pRb/p107 deficient neurons fail to migrate to the correct cortical layer within a wild type brain. These results define a novel role of pocket proteins in regulating cortical lamination through a cell autonomous mechanism independent of their role in apoptosis.


Subject(s)
Apoptosis , Retinoblastoma Protein/genetics , Retinoblastoma-Like Protein p107/genetics , Animals , Cell Death , Cell Differentiation , Female , Mice, Knockout , Neurons/metabolism
3.
Hum Mol Genet ; 19(19): 3734-46, 2010 Oct 01.
Article in English | MEDLINE | ID: mdl-20639397

ABSTRACT

Growing evidence highlights a role for mitochondrial dysfunction and oxidative stress as underlying contributors to Parkinson's disease (PD) pathogenesis. DJ-1 (PARK7) is a recently identified recessive familial PD gene. Its loss leads to increased susceptibility of neurons to oxidative stress and death. However, its mechanism of action is not fully understood. Presently, we report that DJ-1 deficiency in cell lines, cultured neurons, mouse brain and lymphoblast cells derived from DJ-1 patients display aberrant mitochondrial morphology. We also show that these DJ-1-dependent mitochondrial defects contribute to oxidative stress-induced sensitivity to cell death since reversal of this fragmented mitochondrial phenotype abrogates neuronal cell death. Reactive oxygen species (ROS) appear to play a critical role in the observed defects, as ROS scavengers rescue the phenotype and mitochondria isolated from DJ-1 deficient animals produce more ROS compared with control. Importantly, the aberrant mitochondrial phenotype can be rescued by the expression of Pink1 and Parkin, two PD-linked genes involved in regulating mitochondrial dynamics and quality control. Finally, we show that DJ-1 deficiency leads to altered autophagy in murine and human cells. Our findings define a mechanism by which the DJ-1-dependent mitochondrial defects contribute to the increased sensitivity to oxidative stress-induced cell death that has been previously reported.


Subject(s)
Intracellular Signaling Peptides and Proteins/deficiency , Intracellular Signaling Peptides and Proteins/genetics , Mitochondria/genetics , Mitochondria/pathology , Oncogene Proteins/deficiency , Oncogene Proteins/genetics , Parkinson Disease/genetics , Acetylcysteine/pharmacology , Animals , Autophagy/drug effects , Brain/metabolism , Brain/pathology , Cell Death/drug effects , Cell Line , Humans , Mice , Mitochondria/drug effects , Mitochondria/ultrastructure , Mutant Proteins/metabolism , Neostriatum/drug effects , Neostriatum/metabolism , Neostriatum/pathology , Neostriatum/ultrastructure , Neurons/drug effects , Neurons/enzymology , Neurons/pathology , Neurons/ultrastructure , Parkinson Disease/pathology , Peroxiredoxins , Phenotype , Protein Deglycase DJ-1 , Protein Kinases/metabolism , Reactive Oxygen Species/metabolism , Ubiquitin-Protein Ligases/metabolism
4.
Tissue Antigens ; 74(1): 50-6, 2009 Jul.
Article in English | MEDLINE | ID: mdl-19392797

ABSTRACT

Cytokine single nucleotide polymorphisms and consequent production levels have been associated with acute graft-vs-host disease (aGVHD) development. The aim of this pilot study was to determine whether polymorphisms in tumor necrosis factor (TNF), lymphotoxin alpha (LTA) and transforming growth factor beta 1 (TGFB1) showed any association with aGVHD severity. Novel alleles and polymorphisms were identified for each cytokine locus. Genotype distributions were examined in 38 recipient-donor pairs (all chronic myelogenous leukemia in the first chronic phase) with either low-grade (grades 0-I) or high-grade (grades III-IV) aGVHD. Although no significant differences were found, some trends were noted in genotype distributions among aGVHD-grade groups. Power calculations determined that substantially more pairs would be required to show significant associations in distributions among aGVHD-grade groups.


Subject(s)
Gene Frequency/genetics , Graft vs Host Disease/genetics , Lymphotoxin-alpha/genetics , Transforming Growth Factor beta1/genetics , Tumor Necrosis Factor-alpha/genetics , Adolescent , Adult , Female , Genetic Predisposition to Disease , Genotype , Hematopoietic Stem Cell Transplantation , Humans , Male , Middle Aged , Pilot Projects , Polymorphism, Single Nucleotide/genetics , Promoter Regions, Genetic , Young Adult
5.
J Cell Biol ; 140(6): 1497-509, 1998 Mar 23.
Article in English | MEDLINE | ID: mdl-9508781

ABSTRACT

In this report, we have examined the requirement for the retinoblastoma (Rb) gene family in neuronal determination with a focus on the developing neocortex. To determine whether pRb is required for neuronal determination in vivo, we crossed the Rb-/- mice with transgenic mice expressing beta-galactosidase from the early, panneuronal Talpha1 alpha-tubulin promoter (Talpha1:nlacZ). In E12.5 Rb-/- embryos, the Talpha1:nlacZ transgene was robustly expressed throughout the developing nervous system. However, by E14. 5, there were perturbations in Talpha1:nlacZ expression throughout the nervous system, including deficits in the forebrain and retina. To more precisely define the temporal requirement for pRb in neuronal determination, we functionally ablated the pRb family in wild-type cortical progenitor cells that undergo the transition to postmitotic neurons in vitro by expression of a mutant adenovirus E1A protein. These studies revealed that induction of Talpha1:nlacZ did not require proteins of the pRb family. However, in their absence, determined, Talpha1:nlacZ-positive cortical neurons underwent apoptosis, presumably as a consequence of "mixed signals" deriving from their inability to undergo terminal mitosis. In contrast, when the pRb family was ablated in postmitotic cortical neurons, there was no effect on neuronal survival, nor did it cause the postmitotic neurons to reenter the cell cycle. Together, these studies define a critical temporal window of requirement for the pRb family; these proteins are not required for induction of neuronal gene expression or for the maintenance of postmitotic neurons, but are essential for determined neurons to exit the cell cycle and survive.


Subject(s)
Gene Expression Regulation, Developmental , Neurons/chemistry , Neurons/cytology , Retinoblastoma Protein/genetics , Animals , Biotin , Cell Differentiation/physiology , Cell Survival/genetics , Cerebral Cortex/cytology , Cerebral Cortex/embryology , DNA Fragmentation , Deoxyuracil Nucleotides , Lac Operon , Mice , Mice, Transgenic , Mitosis/physiology , Spinal Cord/cytology , Spinal Cord/embryology , Staining and Labeling , Stem Cells/chemistry , Stem Cells/cytology , Time Factors , Tubulin/genetics
6.
J Cell Biol ; 129(3): 779-88, 1995 May.
Article in English | MEDLINE | ID: mdl-7730411

ABSTRACT

The retinoblastoma (RB) protein is present at low levels in early mouse embryos and in pluripotent P19 embryonal carcinoma cells; however, the levels of RB rise dramatically in neuroectoderm formed both in embryos and in differentiating cultures of P19 cells. To investigate the effect of inactivating RB and related proteins p107 and p130, we transfected P19 cells with genes encoding mutated versions of the adenovirus E1A protein that bind RB and related proteins. When these E1A-expressing P19 cells were induced to differentiate into neuroectoderm, there was a striking rise in the expression of c-fos and extensive cell death. The ultrastructural and biochemical characteristics of the dying cells were indicative of apoptosis. The dying cells were those committed to the neural lineages because neurons and astrocytes were lost from differentiating cultures. Cell death was dependent on the ability of the E1A protein to bind RB and related proteins. Our results suggest that proteins of the RB family are essential for the development of the neural lineages and that the absence of functional RB activity triggers apoptosis of differentiating neuroectodermal cells.


Subject(s)
Adenovirus E1A Proteins/metabolism , Apoptosis , Nervous System/embryology , Phosphoproteins , Retinoblastoma Protein/metabolism , Adenovirus E1A Proteins/genetics , Animals , Astrocytes/pathology , Carcinoma, Embryonal , Cell Differentiation , DNA Damage , Ectoderm , Gene Expression Regulation/drug effects , Genes, Viral/genetics , Mice , Muscles/embryology , Nervous System/drug effects , Nervous System/pathology , Nervous System/ultrastructure , Neurons/pathology , Nuclear Proteins/metabolism , Protein Binding , Proteins/metabolism , Proto-Oncogene Proteins c-fos/biosynthesis , Retinoblastoma-Like Protein p107 , Retinoblastoma-Like Protein p130 , Sequence Deletion , Stem Cells/physiology , Tretinoin/pharmacology , Tumor Cells, Cultured
7.
J Cell Biol ; 155(2): 207-16, 2001 Oct 15.
Article in English | MEDLINE | ID: mdl-11591730

ABSTRACT

p53 is a transcriptional activator which has been implicated as a key regulator of neuronal cell death after acute injury. We have shown previously that p53-mediated neuronal cell death involves a Bax-dependent activation of caspase 3; however, the transcriptional targets involved in the regulation of this process have not been identified. In the present study, we demonstrate that p53 directly upregulates Apaf1 transcription as a critical step in the induction of neuronal cell death. Using DNA microarray analysis of total RNA isolated from neurons undergoing p53-induced apoptosis a 5-6-fold upregulation of Apaf1 mRNA was detected. Induction of neuronal cell death by camptothecin, a DNA-damaging agent that functions through a p53-dependent mechanism, resulted in increased Apaf1 mRNA in p53-positive, but not p53-deficient neurons. In both in vitro and in vivo neuronal cell death processes of p53-induced cell death, Apaf1 protein levels were increased. We addressed whether p53 directly regulates Apaf1 transcription via the two p53 consensus binding sites in the Apaf1 promoter. Electrophoretic mobility shift assays demonstrated p53-DNA binding activity at both p53 consensus binding sequences in extracts obtained from neurons undergoing p53-induced cell death, but not in healthy control cultures or when p53 or the p53 binding sites were inactivated by mutation. In transient transfections in a neuronal cell line with p53 and Apaf1 promoter-luciferase constructs, p53 directly activated the Apaf1 promoter via both p53 sites. The importance of Apaf1 as a p53 target gene in neuronal cell death was evaluated by examining p53-induced apoptotic pathways in primary cultures of Apaf1-deficient neurons. Neurons treated with camptothecin were significantly protected in the absence of Apaf1 relative to those derived from wild-type littermates. Together, these results demonstrate that Apaf1 is a key transcriptional target for p53 that plays a pivotal role in the regulation of apoptosis after neuronal injury.


Subject(s)
Apoptosis , Neurons/metabolism , Proteins/genetics , Transcriptional Activation , Tumor Suppressor Protein p53/physiology , Animals , Apoptotic Protease-Activating Factor 1 , Base Sequence , Brain Ischemia/metabolism , Brain Ischemia/pathology , Camptothecin/pharmacology , Cell Line , Cells, Cultured , Mice , Mice, Transgenic , Neurons/pathology , Promoter Regions, Genetic , Protein Biosynthesis , Proteins/physiology , RNA, Messenger/biosynthesis
8.
J Cell Biol ; 135(4): 1085-96, 1996 Nov.
Article in English | MEDLINE | ID: mdl-8922388

ABSTRACT

Programmed cell death is an ongoing process in both the developing and the mature nervous system. The tumor suppressor gene, p53, can induce apoptosis in a number of different cell types. Recently, the enhanced expression of p53 has been observed during acute neurological disease. To determine whether p53 overexpression could influence neuronal survival, we used a recombinant adenovirus vector carrying wild type p53 to transduce postmitotic neurons. A control consisting of the same adenovirus vector background but carrying the lacZ reporter expression cassette was used to establish working parameters for the effective genetic manipulation of sympathetic neurons. We have found that recombinant adenovirus can be used at titers sufficiently high (10 to 50 multiplicity of infection) to transduce the majority of the neuronal population without perturbing survival, electrophysiological function, or cytoarchitecture. Moreover, we demonstrate that overexpression of wild type p53 is sufficient to induce programmed cell death in neurons. The observation that p53 is capable of inducing apoptosis in postmitotic neurons has major implications for the mechanisms of cell death in the traumatized mature nervous system.


Subject(s)
Adenoviridae , Apoptosis/physiology , Gene Transfer Techniques , Neurons/cytology , Tumor Suppressor Protein p53/genetics , Animals , Biotin , Cell Survival/genetics , Cellular Senescence/physiology , DNA Fragmentation , Deoxyuracil Nucleotides , Electrophysiology , Gene Expression/physiology , Microscopy, Electron , Mitosis/physiology , Neurons/physiology , Neurons/ultrastructure , Rats , Rats, Sprague-Dawley , Recombinant Proteins/genetics , Simplexvirus , Staining and Labeling , Superior Cervical Ganglion/cytology , Sympathetic Nervous System/cytology , Transduction, Genetic
9.
Mol Cell Biol ; 14(12): 8451-9, 1994 Dec.
Article in English | MEDLINE | ID: mdl-7969178

ABSTRACT

When introduced into P19 embryonal carcinoma cells, recombinant genes encoding MyoD converted only a small percentage (< 3%) of the transfected cells into skeletal muscle. We isolated stably transfected cells that expressed the MyoD transcript. These P19[MyoD] cells continued to express markers characteristic of undifferentiated stem cells but also expressed myf-5 and the myotonic dystrophy kinase, transcripts normally present in myoblasts but absent from P19 cells. Aggregation of P19[MyoD] cells induced the expression of myogenin, desmin, and the retinoblastoma protein and resulted in the rapid and abundant development of skeletal muscle. Both the embryonic and the slow isoforms of myosin heavy chain were present in this muscle, indicating that it resembled skeletal muscle formed from primary myoblasts. Since aggregation of P19 cells normally results in inefficient differentiation and the development of only low levels of cardiac muscle but no skeletal muscle, we conclude that MyoD imposes the skeletal muscle program on P19 cells and that the differentiation of these cells requires inductive events provided by cell aggregation.


Subject(s)
Muscles/cytology , MyoD Protein/physiology , Animals , Carcinoma, Embryonal , Cell Aggregation , Cell Differentiation , In Vitro Techniques , Mice , Muscle Proteins/metabolism , Tumor Cells, Cultured
10.
Curr Opin Neurobiol ; 6(5): 576-83, 1996 Oct.
Article in English | MEDLINE | ID: mdl-8937820

ABSTRACT

The inability to reliably express foreign proteins in postmitotic neurons has hampered numerous studies in the field of neurobiology. Within the past several years, a number of viral vectors that overcome this problem under controlled conditions in vitro have been developed. In particular, recombinant adenoviruses have proved to be efficient, non-cytotoxic vectors for manipulating neurons in dissociated and organotypic cultures, when used at low viral titres. In contrast, vectors derived from herpes simplex virus 1 still suffer from concerns regarding cellular cytotoxicity, in spite of several generations of vector development; however, recent advances in amplicon technology may solve this problem. Finally, several new-generation vectors, including those generated from adeno-associated virus, show great promise as neuronal gene-transfer vectors in vivo.


Subject(s)
Gene Transfer Techniques , Genetic Vectors , Neurons/cytology , Transfection/methods , Viruses , Adenoviridae/genetics , Animals , Cells, Cultured , Dependovirus/genetics , Herpesvirus 1, Human/genetics , Humans , Organ Culture Techniques
11.
Cell Death Differ ; 23(4): 565-75, 2016 Apr.
Article in English | MEDLINE | ID: mdl-25909886

ABSTRACT

Cell cycle proteins are important regulators of diverse cell fate decisions, and in this capacity have pivotal roles in neurogenesis and brain development. The mechanisms by which cell cycle regulation is integrated with cell fate control in the brain and other tissues are poorly understood, and an outstanding question is whether the cell cycle machinery regulates fate decisions directly or instead as a secondary consequence of proliferative control. Identification of the genes targeted by E2 promoter binding factor (E2f) transcription factors, effectors of the pRb/E2f cell cycle pathway, will provide essential insights into these mechanisms. We identified the promoter regions bound by three neurogenic E2f factors in neural precursor cells in a genome-wide manner. Through bioinformatic analyses and integration of published genomic data sets we uncovered hundreds of transcriptionally active E2f-bound promoters corresponding to genes that control cell fate processes, including key transcriptional regulators and members of the Notch, fibroblast growth factor, Wnt and Tgf-ß signaling pathways. We also demonstrate a striking enrichment of the CCCTC binding factor transcription factor (Ctcf) at E2f3-bound nervous system-related genes, suggesting a potential regulatory co-factor for E2f3 in controlling differentiation. Finally, we provide the first demonstration of extensive tissue specificity among E2f target genes in mammalian cells, whereby E2f3 promoter binding is well conserved between neural and muscle precursors at genes associated with cell cycle processes, but is tissue-specific at differentiation-associated genes. Our findings implicate the cell cycle pathway as a widespread regulator of cell fate genes, and suggest that E2f3 proteins control cell type-specific differentiation programs by regulating unique sets of target genes. This work significantly enhances our understanding of how the cell cycle machinery impacts cell fate and differentiation, and will importantly drive further discovery regarding the mechanisms of cell fate control and transcriptional regulation in the brain, as well as in other tissues.


Subject(s)
E2F Transcription Factors/metabolism , Gene Expression Regulation , Repressor Proteins/metabolism , Response Elements , Transcription, Genetic , Animals , CCCTC-Binding Factor , E2F Transcription Factors/genetics , Mice , Mice, Mutant Strains , Organ Specificity/genetics , Repressor Proteins/genetics , Retinoblastoma/genetics , Retinoblastoma/metabolism
12.
Oncogene ; 10(1): 19-25, 1995 Jan 05.
Article in English | MEDLINE | ID: mdl-7824272

ABSTRACT

We transfected P19 embryonal carcinoma (EC) cells with genes encoding the adenovirus 5 E1A products. Expression of either the 12S or 13S transcripts yielded P19 cells either incapable of proliferating or able to proliferate but having lost the characteristics of the EC cell parent. The proliferating clones of E1A expressing P19 cells were incapable of differentiating in response to retinoic acid or dimethyl sulfoxide, no longer expressed the SSEA-1 surface antigen characteristic of EC cells, and did express cytokeratin 55, a marker of epithelial tissues. We used a number of 12S E1A constructs carrying deletions in the first exon and found that the effects on P19 cell growth and differentiated properties were lost with alterations affecting either the N terminal 25 amino acids or the CR1 region of the E1A protein. Both regions are required to bind the cellular p300 protein that we showed is present in P19 cells. We conclude that binding of E1A to the p300 protein in P19 cells results in the loss of EC cell characteristics.


Subject(s)
Adenovirus E1A Proteins/physiology , Carcinoma, Embryonal/pathology , Nuclear Proteins/metabolism , Trans-Activators , Transcription Factors/metabolism , Cell Differentiation , Endoderm/metabolism , Mutation , Protein Binding , Sequence Deletion , Tumor Cells, Cultured
13.
Oncogene ; 8(6): 1585-91, 1993 Jun.
Article in English | MEDLINE | ID: mdl-8502481

ABSTRACT

The expression of the retinoblastoma susceptibility (RB) gene was investigated in P19 embryonal carcinoma cells and in these cells induced to differentiate with retinoic acid (RA) or with dimethyl sulfoxide (DMSO). In undifferentiated cells very low levels of RB mRNA and protein were present. DMSO-treated P19 cell cultures develop into mesodermal and endodermal cells and RB expression increased only slightly in these differentiating cells. RA-treated P19 cells develop into neuroectoderm and this differentiation was accompanied by a marked increase in RB expression with mRNA levels increasing 15 fold by 4-6 days following initiation of RA treatment. No such increase occurred in mutant cells that fail to respond to RA. The RB promoter did not appear to be directly activated by RA. Nevertheless, the increase in RB expression in RA-treated cells appeared to be due to enhanced initiation of transcription because cells transfected with a reporter gene driven by the RB promoter expressed the reporter gene with kinetics similar to that of the RB gene. Thus the activation of the RB gene appears to be achieved indirectly by RA-induced factor(s) in differentiating neuroectodermal cells. The post-mitotic neurons that developed in RA-treated cultures contained only the hypophosphorylated form of the RB protein. Recent studies (Clarke et al., 1992; Jacks et al., 1992; Lee et al., 1992) have shown that mice lacking the RB gene have abnormalities in early brain development suggesting that the rapid rise in RB expression and the hypophosphorylation of the protein are essential for neuronal cell differentiation.


Subject(s)
Gene Expression Regulation, Neoplastic , Genes, Retinoblastoma , Teratoma/pathology , Animals , Blotting, Western , Cell Differentiation , Chloramphenicol O-Acetyltransferase/genetics , Chloramphenicol O-Acetyltransferase/metabolism , Dimethyl Sulfoxide/pharmacology , Humans , Mice , Neurons/cytology , Neurons/drug effects , Neurons/metabolism , Promoter Regions, Genetic , RNA, Messenger/metabolism , Retinoblastoma Protein/analysis , Retinoblastoma Protein/biosynthesis , Retinoblastoma Protein/genetics , Teratoma/genetics , Transfection , Tretinoin/pharmacology , Tumor Cells, Cultured
14.
J Neurosci ; 21(14): 5017-26, 2001 Jul 15.
Article in English | MEDLINE | ID: mdl-11438577

ABSTRACT

DNA damage has been implicated as one important initiator of cell death in neuropathological conditions such as stroke. Accordingly, it is important to understand the signaling processes that control neuronal death induced by this stimulus. Previous evidence has shown that the death of embryonic cortical neurons treated with the DNA-damaging agent camptothecin is dependent on the tumor suppressor p53 and cyclin-dependent kinase (CDK) activity and that the inhibition of either pathway alone leads to enhanced and prolonged survival. We presently show that p53 and CDKs are activated independently on parallel pathways. An increase in p53 protein levels, nuclear localization, and DNA binding that result from DNA damage are not affected by the inhibition of CDK activity. Conversely, no decrease in retinoblastoma protein (pRb) phosphorylation was observed in p53-deficient neurons that were treated with camptothecin. However, either p53 deficiency or the inhibition of CDK activity alone inhibited Bax translocation, cytochrome c release, and caspase-3-like activation. Taken together, our results indicate that p53 and CDK are activated independently and then act in concert to control Bax-mediated apoptosis.


Subject(s)
Cyclin-Dependent Kinases/metabolism , DNA Damage/physiology , Neurons/metabolism , Proto-Oncogene Proteins c-bcl-2 , Proto-Oncogene Proteins/metabolism , Tumor Suppressor Protein p53/metabolism , Active Transport, Cell Nucleus/drug effects , Active Transport, Cell Nucleus/physiology , Animals , Apoptosis/physiology , Camptothecin/pharmacology , Caspase 3 , Caspases/metabolism , Cell Survival , Cells, Cultured , Cyclin-Dependent Kinases/antagonists & inhibitors , Cytochrome c Group/metabolism , Enzyme Inhibitors/pharmacology , Mice , Mice, Knockout , Neurons/cytology , Neurons/drug effects , Phosphorylation/drug effects , Proto-Oncogene Proteins/deficiency , Proto-Oncogene Proteins/genetics , Retinoblastoma Protein/metabolism , Signal Transduction/drug effects , Signal Transduction/physiology , Tumor Suppressor Protein p53/deficiency , bcl-2-Associated X Protein
15.
J Neurosci ; 19(18): 7860-9, 1999 Sep 15.
Article in English | MEDLINE | ID: mdl-10479688

ABSTRACT

p53 is a pivotal molecule regulating the death of neurons both after acute injury and during development. The molecular mechanisms by which p53 induces apoptosis in neuronal cells, however, are not well understood. We have shown previously that adenovirus-mediated p53 gene delivery to neurons was sufficient to induce apoptosis. In the present study we have examined the molecular mechanism by which p53 evokes neuronal cell death. Adenovirus-mediated delivery of p53 to cerebellar granule neurons resulted in caspase-3 (CPP32) activation followed by terminal deoxynucleotidyl transferase-mediated biotinylated UTP nick end labeling (TUNEL) staining and loss of viability as determined by an MTT survival assay. To determine whether Bax is essential for caspase-3 activation, p53 was expressed in Bax-deficient cells. Bax null neurons did not exhibit caspase-3 activation in response to p53 and were protected from apoptosis. To determine whether Bax-dependent caspase-3 activation was required in p53-mediated neuronal cell death, caspase-3-deficient neurons were examined. Our results indicate that caspase-3-deficient neurons exhibit a remarkable delay in apoptosis and a dramatic decrease in TUNEL-positive cells. These studies demonstrate that p53-induced cell death in postmitotic neurons involves a Bax-dependent caspase-3 activation, suggesting that these molecules are important determinants in neuronal cell death after injury.


Subject(s)
Apoptosis , Brain/cytology , Caspases/metabolism , Neurons/cytology , Neurons/physiology , Proto-Oncogene Proteins c-bcl-2 , Proto-Oncogene Proteins/physiology , Tumor Suppressor Protein p53/metabolism , Adenoviridae , Animals , Animals, Newborn , Brain/physiology , Caspase 3 , Caspases/genetics , Cells, Cultured , Cerebellum/cytology , Cerebellum/physiology , Genes, p53 , Kinetics , Mice , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , Proto-Oncogene Proteins/genetics , Tumor Suppressor Protein p53/genetics , bcl-2-Associated X Protein
16.
J Neurosci ; 21(18): 7089-98, 2001 Sep 15.
Article in English | MEDLINE | ID: mdl-11549719

ABSTRACT

The retinoblastoma tumor suppressor protein, pRb, is a key regulator of cell cycle and has been implicated in the terminal differentiation of neuronal cells. Mice nullizygous for pRb die by embryonic day 14.5 from hematopoietic and neurological defects attributed to failed differentiation (Clarke et al., 1992; Jacks et al., 1992; Lee et al., 1992). Previous studies by MacLeod et al. (1996) have demonstrated that the loss of p53 protects Rb-deficient CNS neurons but not peripheral nervous system (PNS) neurons from cell death. Thus, the mechanisms by which PNS neurons undergo apoptosis in response to Rb deficiency remain unknown. In view of the pivotal role of caspase 3 in the regulation of neuronal apoptosis during development, we examined its function in the execution of the wide-spread neuronal cell death induced by Rb deficiency. Our results support a number of conclusions. First, we show that caspase 3 becomes activated in all neuronal populations undergoing apoptosis. Second, caspase 3 deficiency does not extend the life span of Rb null embryos, because double null mutants exhibit high rates of liver apoptosis resulting in erythropoietic failure. Third, Rb/caspase 3 double-mutant neurons of the CNS exhibit widespread apoptosis similar to that seen in Rb mutants alone; thus caspase 3 deficiency does not protect this population from apoptosis. Finally, in contrast to the CNS, neurons of the PNS including those comprising the trigeminal ganglia and the dorsal root ganglia are protected from apoptosis in Rb/caspase 3 double-mutant embryos. Examination of the mechanistic differences between these two cell types suggest that CNS neurons may invoke other caspases to facilitate apoptosis in the absence of caspase 3. These findings suggest that PNS neurons are dependent on caspase 3 for the execution of apoptosis and that caspase 3 may serve as a key therapeutic target for neuroprotection after injury of this cell type.


Subject(s)
Caspases/deficiency , Peripheral Nervous System/physiopathology , Retinoblastoma Protein/deficiency , Amyloid beta-Protein Precursor/metabolism , Animals , Apoptosis , Caspase 3 , Caspases/biosynthesis , Caspases/genetics , Central Nervous System/cytology , Central Nervous System/embryology , Central Nervous System/metabolism , Crosses, Genetic , Enzyme Induction/physiology , Fluorescent Dyes , Ganglia, Spinal/cytology , Ganglia, Spinal/embryology , Ganglia, Spinal/metabolism , Gene Expression Regulation, Developmental/physiology , Genotype , In Situ Nick-End Labeling , Mice , Mice, Knockout , Neurons/classification , Neurons/metabolism , Neurons/pathology , Organ Specificity , Peripheral Nervous System/embryology , Peripheral Nervous System/pathology , Retinoblastoma Protein/biosynthesis , Retinoblastoma Protein/genetics
17.
Biochim Biophys Acta ; 1053(1): 89-96, 1990 Jun 12.
Article in English | MEDLINE | ID: mdl-2114183

ABSTRACT

Studies on the involvement of protein kinase C in retinoic acid-induced differentiation of human neuroblastoma were carried out with two variants of the SK-N-SH cell line namely the SH-F subline, which differentiates to give a fibroblast-like phenotype, and the SH-N subline, which develops into the typical neuronal phenotype. In SH-F, a substantial increase in protein kinase C activity accompanied morphological differentiation. Accordingly, after 7 days of retinoic acid treatment, EDTA-extracted, cytosolic protein kinase C activity increased by slightly more than 2-fold over vehicle-treated controls. Again, detergent-extracted activity, representing membrane-bound or total protein kinase C, showed a similar 2.6- to 5.1-fold increase in treated cells. A time-course study revealed an earliest increase in total activity after two days of retinoic acid treatment which continued linearly for the first 6 to 8 days, and then levelled off. A study of the effect of retinoic acid on the protein kinase C in vitro with SH-F cell extracts showed only a slight increase in activity (of 25%) at the relatively high concentration of 10(-4) M; however, no significant differences were observed at lower concentrations. In contrast, the SH-N cell line responded to retinoic acid by a 45% decrease in EDTA-extractable, and a 63% decrease in detergent-extractable protein kinase C activity. Added to SH-F cell cultures, 15 nM staurosporine was found to inhibit protein kinase C in vivo and to a lesser extent, the protein kinase A. Present together with retinoic acid, staurosporine not only prevented the augmentation but caused a marked decrease of protein kinase C activity in this cell line. Morphological studies indicated that when SH-N cells are treated with staurosporine, or staurosporine and retinoic acid together, a neuronal phenotype similar to that produced by retinoic acid alone is observed. In contrast, when the SH-F cell line is treated with staurosporine or staurosporine and retinoic acid together, the flattened fibroblast-like cell type normally induced by retinoic acids is not observed. Instead, these cells display much smaller cell bodies and elaborate extensions resembling the neuronal phenotype produced by retinoic acid induced differentiation of the SH-N variant. These results suggest that changes in the protein kinase C activity may be involved in regulating the expression of the phenotype during cell differentiation.


Subject(s)
Alkaloids/pharmacology , Neuroblastoma/enzymology , Protein Kinase C/metabolism , Tretinoin/pharmacology , Cell Differentiation/drug effects , Cytosol/enzymology , Edetic Acid , Fibroblasts/pathology , Humans , Neuroblastoma/pathology , Neurons/pathology , Staurosporine , Tumor Cells, Cultured
18.
Neuroreport ; 12(9): A55-62, 2001 Jul 03.
Article in English | MEDLINE | ID: mdl-11435948

ABSTRACT

Cell division during embryogenesis plays a crucial role in the formation of the nervous system. During this developmental process, proliferating neural precursor cells commit to a neuronal fate and, as a consequence, undergo terminal mitosis and adopt a neuronal phenotype. A key cell cycle regulator, the tumor suppressor protein, retinoblastoma (Rb), is involved in both terminal mitosis and neuronal differentiation. Neural development is a complex process involving cell proliferation, cell fate determination and differentiation, as well as programmed cell death. In this review, we will examine each of these processes in turn, focussing on the role of the Rb family proteins to examine their many influences on these events.


Subject(s)
Cell Cycle/genetics , Cell Differentiation/genetics , Nervous System/embryology , Neurons/metabolism , Retinoblastoma Protein/metabolism , Animals , Apoptosis/genetics , Humans , Nervous System/cytology , Nervous System/metabolism , Neurons/cytology , Retinoblastoma Protein/genetics , Stem Cells/cytology , Stem Cells/metabolism
19.
Clin Chest Med ; 15(4): 739-49, 1994 Dec.
Article in English | MEDLINE | ID: mdl-7867288

ABSTRACT

Pulmonary dysfunction is a common complication of head trauma and spinal cord injury. Abnormal breathing patterns reflect the influence of altered neural integration. Early arterial hypoxemia can result from ventilation-perfusion mismatching, microatelectasis, aspiration, fat embolism, or the development of the adult respiratory distress syndrome. Significant changes in lung volumes, ventilation, and gas exchange can occur in spinal cord injury as a result of the loss of diaphramatic or intercostal muscle function. Recruitment of accessory respiratory muscles plays an important role in stabilizing the rib cage and improving expiratory function. Strength training improves expiratory muscle function in quadriplegics and should be continued indefinitely. Most importantly, survival of patients with CNS injuries improves with meticulous and vigorous pulmonary hygiene. The pulmonary hygiene program should include regular changes in the patient's position, assisted coughing and deep breathing exercises, incentive spirometer, bronchodilators, fiberoptic bronchoscopy when indicated, and frequent monitoring of pulmonary mechanics. Long-term survival of the patient with head trauma or spinal cord injury is correlated to successful weaning from mechanical ventilation. Various forms of mechanical ventilator support can be adopted for the patient's ventilatory needs, and many patients will achieve some degree of freedom from mechanical ventilation. Newer ventilatory assist devices that do not require tracheostomy should be considered.


Subject(s)
Brain Injuries/complications , Respiratory Insufficiency/physiopathology , Spinal Cord Injuries/complications , Brain Injuries/physiopathology , Humans , Respiratory Insufficiency/etiology , Respiratory Insufficiency/therapy , Respiratory Mechanics/physiology , Respiratory Therapy , Spinal Cord Injuries/physiopathology
20.
Dev Genet ; 18(1): 81-91, 1996.
Article in English | MEDLINE | ID: mdl-8742837

ABSTRACT

The retinoblastoma gene (Rb) was the first tumor suppressor gene to be cloned [Dryja et al., 1986; Friend et al., 1986; Lee et al., 1987], and, as a consequence, has been studied intensively within the context of cell cycle regulation and oncogenesis. However, a number of recent findings indicate that the retinoblastoma gene product (pRb) likely plays an essential role not only in controlling entry into the cell cycle, but also in the terminal differentiation of a number of different cell types [Lee et al., 1994; Gu et al., 1993]. In particular, the phenotype of the Rb nullizygous mice, created by a number of groups using homologous recombination [Jacks et al., 1992: Clarke et al., 1992; Lee et al., 1992], indicates that pRb is essential for normal development of the nervous and hematopoietic systems and may even function to regulate apoptosis [Haas-Kogan et al., 1995]. Although this paper briefly reviews the traditional role of pRB in regulation of cellular proliferation, we focus on the role of pRB in neuronal development and apoptosis. Recent reviews have been published on the role of pRb in cell cycle and transcriptional regulation [Hamel et al., 1992; Cobrinik et al., 1992; Kouzarides, 1993; Hollingsworth et al., 1993; Helin and Harlow, 1993; Sherr, 1994], as well as the relationship between pRb and p53 [Picksley and Lane, 1994; White, 1994].


Subject(s)
Genes, Retinoblastoma , Mice/embryology , Mice/genetics , Nervous System/embryology , Neurons/physiology , Animals , Apoptosis , Cell Cycle , Cell Differentiation , Cell Division , Nervous System/cytology , Neurons/cytology , Retinoblastoma Protein/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL