Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 10 de 10
Filter
Add more filters










Publication year range
1.
Sci Rep ; 9(1): 12587, 2019 08 29.
Article in English | MEDLINE | ID: mdl-31467368

ABSTRACT

Cerebrospinal fluid (CSF) is produced in the cerebral ventricles and circulates within the subarachnoid space (SAS) of the brain and spinal cord, where it exchanges with interstitial fluid of the parenchyma. The access of CSF to the entire central nervous system (CNS) makes it an attractive medium for drug delivery. However, few intrathecal (IT) therapies have reached the clinic due, in part, to limited distribution and rapid clearance. Given the success of nanoparticle (NP) carriers in prolonging circulation and improving delivery of systemically administered agents, we sought to evaluate the distribution of IT injected NPs within the CNS. We administered fluorescent, 100 nm PEGylated-NPs into the cisterna magna of healthy mice and studied their distribution along the brain and spinal cord. Our data demonstrate that NPs are capable of distributing rapidly through the SAS along the entire neuraxis with reproducible, anatomically defined patterns of delivery. NPs were well retained within the leptomeninges for over 3 weeks, showing preference for ventral surfaces and minimal penetration into the CNS parenchyma. Clearance of NPs occurred across the cribriform plate into the nasal mucosa, with a small fraction of NPs localizing with nerve roots exiting the spinal column. Larger 10 µm particles were also capable of moving through the SAS but did not achieve as widespread distribution. These studies demonstrate the ability of NPs to achieve widespread delivery along the neuraxis and highlight IT administration as a potentially significant route of administration for delivery of nanomedicine to the subarachnoid space.


Subject(s)
Brain/metabolism , Nanoparticles , Polystyrenes/chemistry , Polystyrenes/metabolism , Spinal Cord/metabolism , Animals , Injections, Spinal , Mice , Polyethylene Glycols/chemistry , Polystyrenes/administration & dosage
2.
Oncogene ; 34(29): 3770-9, 2015 Jul.
Article in English | MEDLINE | ID: mdl-25241898

ABSTRACT

Medulloblastoma (MB) is a highly malignant brain tumor that occurs primarily in children. Although surgery, radiation and high-dose chemotherapy have led to increased survival, many MB patients still die from their disease, and patients who survive suffer severe long-term side effects as a consequence of treatment. Thus, more effective and less toxic therapies for MB are critically important. Development of such therapies depends in part on identification of genes that are necessary for growth and survival of tumor cells. Survivin is an inhibitor of apoptosis protein that regulates cell cycle progression and resistance to apoptosis, is frequently expressed in human MB and when expressed at high levels predicts poor clinical outcome. Therefore, we hypothesized that Survivin may have a critical role in growth and survival of MB cells and that targeting it may enhance MB therapy. Here we show that Survivin is overexpressed in tumors from patched (Ptch) mutant mice, a model of Sonic hedgehog (SHH)-driven MB. Genetic deletion of survivin in Ptch mutant tumor cells significantly inhibits proliferation and causes cell cycle arrest. Treatment with small-molecule antagonists of Survivin impairs proliferation and survival of both murine and human MB cells. Finally, Survivin antagonists impede growth of MB cells in vivo. These studies highlight the importance of Survivin in SHH-driven MB, and suggest that it may represent a novel therapeutic target in patients with this disease.


Subject(s)
Cerebellar Neoplasms/metabolism , Hedgehog Proteins/metabolism , Inhibitor of Apoptosis Proteins/deficiency , Medulloblastoma/metabolism , Repressor Proteins/deficiency , Animals , Apoptosis/drug effects , Apoptosis/radiation effects , Biphenyl Compounds/pharmacology , Blotting, Western , Cell Cycle/drug effects , Cell Cycle/radiation effects , Cell Proliferation/drug effects , Cell Proliferation/radiation effects , Cerebellar Neoplasms/drug therapy , Cerebellar Neoplasms/genetics , Chemoradiotherapy , Child , Hedgehog Proteins/antagonists & inhibitors , Humans , Imidazoles/pharmacology , Inhibitor of Apoptosis Proteins/antagonists & inhibitors , Inhibitor of Apoptosis Proteins/genetics , Interleukin Receptor Common gamma Subunit/deficiency , Interleukin Receptor Common gamma Subunit/genetics , Ki-67 Antigen/metabolism , Medulloblastoma/drug therapy , Medulloblastoma/genetics , Mice, Inbred C57BL , Mice, Inbred NOD , Mice, Knockout , Mice, Nude , Mice, SCID , Microscopy, Confocal , Naphthoquinones/pharmacology , Pyridines/pharmacology , Repressor Proteins/antagonists & inhibitors , Repressor Proteins/genetics , Survivin , Tumor Cells, Cultured , Xenograft Model Antitumor Assays
3.
Oncogene ; 32(35): 4181-8, 2013 Aug 29.
Article in English | MEDLINE | ID: mdl-23045271

ABSTRACT

Cerebellar granule neurons are the most abundant neurons in the brain, and a critical element of the circuitry that controls motor coordination and learning. In addition, granule neuron precursors (GNPs) are thought to represent cells of origin for medulloblastoma, the most common malignant brain tumor in children. Thus, understanding the signals that control the growth and differentiation of these cells has important implications for neurobiology and neurooncology. Our previous studies have shown that proliferation of GNPs is regulated by Sonic hedgehog (Shh), and that aberrant activation of the Shh pathway can lead to medulloblastoma. Moreover, we have demonstrated that Shh-dependent proliferation of GNPs and medulloblastoma cells can be blocked by basic fibroblast growth factor (bFGF). But while the mitogenic effects of Shh signaling have been confirmed in vivo, the inhibitory effects of bFGF have primarily been studied in culture. Here, we demonstrate that mice lacking FGF signaling in GNPs exhibit no discernable changes in GNP proliferation or differentiation. In contrast, activation of FGF signaling has a potent effect on tumor growth: treatment of medulloblastoma cells with bFGF prevents them from forming tumors following transplantation, and inoculation of tumor-bearing mice with bFGF markedly inhibits tumor growth in vivo. These results suggest that activators of FGF signaling may be useful for targeting medulloblastoma and other Shh-dependent tumors.


Subject(s)
Cerebellar Neoplasms/pathology , Cerebellum/growth & development , Fibroblast Growth Factor 2/physiology , Medulloblastoma/pathology , Signal Transduction/physiology , Animals , Cell Cycle , Cell Differentiation , Cerebellar Neoplasms/etiology , Hedgehog Proteins/physiology , Medulloblastoma/etiology , Mice , Mice, Inbred C57BL , Neurons/cytology , Receptor, Fibroblast Growth Factor, Type 1/physiology , Stem Cells/cytology
4.
Neuropathol Appl Neurobiol ; 38(3): 228-40, 2012 Jun.
Article in English | MEDLINE | ID: mdl-22070547

ABSTRACT

Medulloblastoma, the most common malignant paediatric brain tumour, is thought to arise from mutations in progenitors or stem cells in the cerebellum. Recent molecular analyses have highlighted the heterogeneity of these tumours, and demonstrated that they can be classified into at least four major subtypes that differ in terms of gene expression, genomic gains and losses, epidemiology and patient outcome. Along with analysis of human tumours, a variety of animal models of medulloblastoma have been developed using transgenic and knockout technology as well as somatic gene delivery. These models have provided valuable insight into the origins of the disease and the signalling pathways that control tumour growth. But the degree to which current models recapitulate the heterogeneity of the human disease remains unclear. Here we review the recent literature on the genomics of medulloblastoma and discuss the relationship of mouse models to the subtypes of the disease. Judicious use of existing models, and generation of additional models for poorly studied subtypes of medulloblastoma, will increase our understanding of tumour biology and allow evaluation of novel approaches to treatment of the disease.


Subject(s)
Brain Neoplasms/genetics , Brain Neoplasms/pathology , Disease Models, Animal , Medulloblastoma/genetics , Medulloblastoma/pathology , Animals , Humans , Mice , Mutation/genetics , Signal Transduction , Stem Cells/pathology
5.
Trends Neurosci ; 24(12): 680-2, 2001 Dec.
Article in English | MEDLINE | ID: mdl-11718852

ABSTRACT

Cerebellar granule cells are the most abundant neurons in the brain and are crucial to the circuitry that controls motor coordination. The proliferation of granule cell precursors (GCPs) is controlled by the secreted signaling molecule Sonic hedgehog (Shh), but the factors that regulate GCP differentiation remain a mystery. A recent study suggests that the extracellular matrix protein vitronectin might tell GCPs when to stop dividing and begin differentiation.


Subject(s)
Neurons/cytology , Neurons/physiology , Vitronectin/physiology , Animals , Cell Differentiation/physiology , Cerebellum/cytology , Extracellular Matrix Proteins/physiology
6.
Neuron ; 22(1): 103-14, 1999 Jan.
Article in English | MEDLINE | ID: mdl-10027293

ABSTRACT

Cerebellar granule cells are the most abundant type of neuron in the brain, but the molecular mechanisms that control their generation are incompletely understood. We show that Sonic hedgehog (Shh), which is made by Purkinje cells, regulates the division of granule cell precursors (GCPs). Treatment of GCPs with Shh prevents differentiation and induces a potent, long-lasting proliferative response. This response can be inhibited by basic fibroblast growth factor or by activation of protein kinase A. Blocking Shh function in vivo dramatically reduces GCP proliferation. These findings provide insight into the mechanisms of normal growth and tumorigenesis in the cerebellum.


Subject(s)
Cerebellum/cytology , Neurons/cytology , Proteins/physiology , Stem Cells/cytology , Trans-Activators , Animals , Animals, Newborn/growth & development , Animals, Newborn/metabolism , Cell Division/physiology , Cells, Cultured , Cerebellum/growth & development , Cerebellum/metabolism , Growth Substances/pharmacology , Hedgehog Proteins , Membrane Proteins/metabolism , Mice , Mice, Inbred Strains , Mitogens/pharmacology , Mitogens/physiology , Oncogene Proteins/metabolism , Patched Receptors , Proteins/metabolism , Receptors, Cell Surface , Transcription Factors/metabolism , Zinc Finger Protein GLI1
7.
Mol Cell Biol ; 18(1): 566-75, 1998 Jan.
Article in English | MEDLINE | ID: mdl-9418903

ABSTRACT

Bin1 is a Myc-interacting protein with features of a tumor suppressor. The high level of Bin1 expression in skeletal muscle prompted us to investigate its role in muscle differentiation. Significant levels of Bin1 were observed in undifferentiated C2C12 myoblasts, a murine in vitro model system. Induction of differentiation by growth factor withdrawal led to an upregulation of Bin1 mRNA and to the generation of higher-molecular-weight forms of Bin1 protein by alternate splicing. While Bin1 in undifferentiated cells was localized exclusively in the nucleus, differentiation-associated isoforms of Bin1 were found in the cytoplasm as well. To examine the function of Bin1 during differentiation, we generated stable cell lines that express exogenous human Bin1 cDNA in the sense or antisense orientation. Cells overexpressing Bin1 grew more slowly than control cells and differentiated more rapidly when deprived of growth factors. In contrast, C2C12 cells expressing antisense Bin1 showed an impaired ability to undergo differentiation. Taken together, the results indicated that Bin1 expression, structure, and localization are tightly regulated during muscle differentiation and suggested that Bin1 plays a functional role in the differentiation process.


Subject(s)
Adaptor Proteins, Signal Transducing , Carrier Proteins/genetics , Genes, Tumor Suppressor , Muscle, Skeletal/cytology , Nerve Tissue Proteins , Nuclear Proteins/genetics , Tumor Suppressor Proteins , Animals , Carrier Proteins/metabolism , Cell Differentiation/genetics , Cell Line , Gene Expression Regulation , Humans , Mice , Muscle, Skeletal/metabolism , Nuclear Proteins/metabolism
9.
Immunology ; 89(3): 356-62, 1996 Nov.
Article in English | MEDLINE | ID: mdl-8958047

ABSTRACT

Unlike mature B cells, immature B cells are not activated in response to antigen receptor cross-linking. To examine the mechanisms underlying this unresponsiveness, we have studied the effects of reagents that have been shown to alter the responses of immature B cells to antigen receptor stimulation. Bacterial lipopolysaccharide (LPS) is a polyclonal B-cell activator, and has been shown to interfere with B-cell tolerance induction in vivo and in vitro. Here we show that LPS can also overcome the unresponsiveness of immature B cells to stimulation with anti-receptor (anti-mu) antibodies. LPS synergizes with anti-mu to induce a proliferative response that exceeds the response of immature B cells to LPS alone. Moreover, pretreatment of immature cells with LPS allows them to proliferate in response to subsequent stimulation with anti-mu antibodies. This induction of responsiveness to anti-mu requires exposure to LPS for at least 8 hr. Although the mechanisms of induction are not fully understood, one component of the LPS effect appears to involve enhancement of immature B-cell survival in culture. Neonatal splenic B cells undergo spontaneous apoptosis at a much higher rate than mature B cells, but we have found that LPS causes a dramatic inhibition of apoptosis, even when it is present for only the first 8 hr of culture. The ability of LPS to promote survival of immature B cells and allow them to proliferate in response to antigen receptor stimulation provides a system for investigation of the biochemical mechanisms of unresponsiveness and tolerance susceptibility.


Subject(s)
Apoptosis/immunology , B-Lymphocytes/immunology , Immune Tolerance , Lipopolysaccharides/immunology , Receptors, Antigen, B-Cell/immunology , Animals , Animals, Newborn , B-Lymphocyte Subsets/immunology , Cell Culture Techniques , Cell Differentiation/immunology , Cell Division/immunology , Cell Survival/immunology , Dose-Response Relationship, Immunologic , Mice , Mice, Inbred BALB C , Time Factors
10.
J Immunol ; 156(12): 4562-9, 1996 Jun 15.
Article in English | MEDLINE | ID: mdl-8648097

ABSTRACT

In contrast to mature B cells, immature stage B cells do not proliferate following Ag receptor cross-linking with anti-Ig Abs. To determine where in the cell cycle immature B cells arrest, we have examined the expression of specific G, cell cycle regulators. Following surface IgM (sIgM) cross-linking on mature B cells, we observed increased expression of the early G1 kinase, cyclin-dependent kinase 4 (cdk4), and one of its regulatory subunits, cyclin D2. Mature B cells also showed increased expression of components required for G1/S transition, including cyclin E and cdk2. Whereas immature stage B cells increased expression of cyclin D2 and cdk4 after anti-IgM stimulation, unlike mature stage B cells they failed to express cyclin E and cdk2. Expression of cyclin D2 and cdk4 indicates that these cells can exit G0 and enter the initial G1 phase following sIgM ligation. Interestingly, IL-4, which by itself does not stimulate proliferation of immature B cells, induced expression of cyclin E and cdk2. These latter results suggest that IL-4 complements sIgM, signaling for proliferation by increasing the basal levels of late G1 cell cycle regulators. Consistent with this idea, IL-4 synergizes with anti-Ig Abs to promote cell cycle progression and proliferation of immature B cells. Finally, c-myc, a transcriptional regulator of some members of the cell cycle machinery, is not induced following sIgM cross-linking of immature cells. This lack of inducible expression contrasts with that seen in mature stage B cells, and in immature stage cells stimulated to proliferate with LPS. These results suggest that c-myc may be a component of the signaling pathway that induces cyclin E and cdk2 expression.


Subject(s)
B-Lymphocytes/cytology , CDC2-CDC28 Kinases , Cell Cycle , Lymphocyte Activation , Proto-Oncogene Proteins , Receptors, Antigen, B-Cell/physiology , Animals , Animals, Newborn , Cell Differentiation , Cyclin D2 , Cyclin-Dependent Kinase 2 , Cyclin-Dependent Kinase 4 , Cyclin-Dependent Kinases/metabolism , Cyclins/metabolism , Gene Expression , Genes, myc , Immunoglobulin mu-Chains/physiology , Interleukin-4/pharmacology , Mice , Mice, Inbred BALB C , Protein Serine-Threonine Kinases/metabolism , RNA, Messenger/genetics , Spleen/cytology , Up-Regulation
SELECTION OF CITATIONS
SEARCH DETAIL
...