Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 14 de 14
Filter
Add more filters










Publication year range
1.
Cell Transplant ; 26(11): 1811-1821, 2017 11.
Article in English | MEDLINE | ID: mdl-29338380

ABSTRACT

The use of regenerative medicine to treat nervous system disorders like ataxia has been proposed to either replace or support degenerating neurons. In this study, we assessed the ability of human neural progenitor cells (hNPCs) to repair and restore the function of dying neurons within the spastic Han-Wistar rat (sHW), a model of ataxia. The sHW rat suffers from neurodegeneration of specific neurons, including cerebellar Purkinje cells and hippocampal CA3 pyramidal cells leading to the observed symptoms of forelimb tremor, hind-leg rigidity, gait abnormality, motor incoordination, and a shortened life span. To alleviate the symptoms of neurodegeneration and to replace or augment dying neurons, neuronal human progenitor cells were implanted into the sHW rats. At 30 d of age, male sHW mutant rats underwent subcutaneous implantation of an Alzet osmotic pump that infused cyclosporine (15 mg/kg/d) used to suppress the rat's immune system. At 40 d, sHW rats received bilateral injections (500,000 cells in 5 µL media) of live hNPCs, dead hNPCs, live human embryonic kidney cells, or growth media either into the cerebellar cortex or into the hippocampus. To monitor results, motor activity scores (open-field testing) and weights of the animals were recorded weekly. The sHW rats that received hNPC transplantation into the cerebellum, at 60 d of age, displayed significantly higher motor activity scores and sustained greater weights and longevities than control-treated sHW rats or any hippocampal treatment group. In addition, cerebellar histology revealed that the transplanted hNPCs displayed signs of migration and signs of neuronal development in the degenerated Purkinje cell layer. This study revealed that implanted human progenitor cells reduced the ataxic symptoms in the sHW rat, identifying a future clinical use of these progenitor cells against ataxia and associated neurodegenerative diseases.


Subject(s)
Ataxia/therapy , Neural Stem Cells/cytology , Purkinje Cells/cytology , Stem Cell Transplantation/methods , Animals , Cerebellum/cytology , Disease Models, Animal , Hippocampus/cytology , Male , Neural Stem Cells/physiology , Purkinje Cells/physiology , Rats , Rats, Wistar
2.
Cell Mol Neurobiol ; 36(6): 955-964, 2016 Aug.
Article in English | MEDLINE | ID: mdl-26456684

ABSTRACT

Slit2 is a neural axon guidance and chemorepellent protein that stimulates motility in a variety of cell types. The role of Slit2 in neural development and neoplastic growth and migration has been well established, while the genetic mechanisms underlying regulation of the Slit2 gene have not. We identified the core and proximal promoter of Slit2 by mapping multiple transcriptional start sites, analyzing transcriptional activity, and confirming sequence homology for the Slit2 proximal promoter among a number of species. Deletion series and transient transfection identified the Slit2 proximal promoter as within 399 base pairs upstream of the start of transcription. A crucial region for full expression of the Slit2 proximal promoter lies between 399 base pairs and 296 base pairs upstream of the start of transcription. Computer modeling identified three transcription factor-binding consensus sites within this region, of which only site-directed mutagenesis of one of the two identified Sp1 consensus sites inhibited transcriptional activity of the Slit2 proximal promoter (-399 to +253). Bioinformatics analysis of the Slit2 proximal promoter -399 base pair to -296 base pair region shows high sequence conservation over twenty-two species, and that this region follows an expected pattern of sequence divergence through evolution.


Subject(s)
Intercellular Signaling Peptides and Proteins/genetics , Nerve Tissue Proteins/genetics , Promoter Regions, Genetic/genetics , Sp1 Transcription Factor/genetics , Sp1 Transcription Factor/metabolism , Transcription, Genetic/genetics , Transcriptional Activation/physiology , Base Sequence/genetics , Cell Line , Humans , Mutagenesis, Site-Directed/methods , Sequence Deletion/genetics , Transcriptional Activation/genetics , Transfection/methods
3.
Stem Cell Reports ; 3(5): 743-57, 2014 Nov 11.
Article in English | MEDLINE | ID: mdl-25418722

ABSTRACT

Hypoxia augments human embryonic stem cell (hESC) self-renewal via hypoxia-inducible factor 2α-activated OCT4 transcription. Hypoxia also increases the efficiency of reprogramming differentiated cells to a pluripotent-like state. Combined, these findings suggest that low O2 tension would impair the purposeful differentiation of pluripotent stem cells. Here, we show that low O2 tension and hypoxia-inducible factor (HIF) activity instead promote appropriate hESC differentiation. Through gain- and loss-of-function studies, we implicate O2 tension as a modifier of a key cell fate decision, namely whether neural progenitors differentiate toward neurons or glia. Furthermore, our data show that even transient changes in O2 concentration can affect cell fate through HIF by regulating the activity of MYC, a regulator of LIN28/let-7 that is critical for fate decisions in the neural lineage. We also identify key small molecules that can take advantage of this pathway to quickly and efficiently promote the development of mature cell types.


Subject(s)
Cell Differentiation/drug effects , Cell Proliferation/drug effects , Neural Stem Cells/drug effects , Oxygen/pharmacology , Aryl Hydrocarbon Receptor Nuclear Translocator/genetics , Aryl Hydrocarbon Receptor Nuclear Translocator/metabolism , Basic Helix-Loop-Helix Transcription Factors/genetics , Basic Helix-Loop-Helix Transcription Factors/metabolism , Cell Differentiation/genetics , Cell Hypoxia , Cell Line , Cell Proliferation/genetics , Cells, Cultured , Humans , Hypoxia-Inducible Factor 1, alpha Subunit/genetics , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , MicroRNAs/genetics , MicroRNAs/metabolism , Neural Stem Cells/cytology , Neural Stem Cells/metabolism , Neurogenesis/drug effects , Neurogenesis/genetics , Oligonucleotide Array Sequence Analysis , Oxygen/metabolism , Proto-Oncogene Proteins c-myc/genetics , Proto-Oncogene Proteins c-myc/metabolism , RNA Interference , RNA-Binding Proteins/genetics , RNA-Binding Proteins/metabolism , Signal Transduction/drug effects , Signal Transduction/genetics , Transcriptome/drug effects , Transcriptome/genetics
4.
Nat Protoc ; 7(6): 1068-85, 2012 May 10.
Article in English | MEDLINE | ID: mdl-22576106

ABSTRACT

Measurements of glycolysis and mitochondrial function are required to quantify energy metabolism in a wide variety of cellular contexts. In human pluripotent stem cells (hPSCs) and their differentiated progeny, this analysis can be challenging because of the unique cell properties, growth conditions and expense required to maintain these cell types. Here we provide protocols for analyzing energy metabolism in hPSCs and their early differentiated progenies that are generally applicable to mature cell types as well. Our approach has revealed distinct energy metabolism profiles used by hPSCs, differentiated cells, a variety of cancer cells and Rho-null cells. The protocols measure or estimate glycolysis on the basis of the extracellular acidification rate, and they measure or estimate oxidative phosphorylation on the basis of the oxygen consumption rate. Assays typically require 3 h after overnight sample preparation. Companion methods are also discussed and provided to aid researchers in developing more sophisticated experimental regimens for extended analyses of cellular bioenergetics.


Subject(s)
Energy Metabolism , Molecular Biology/methods , Pluripotent Stem Cells/metabolism , Cell Differentiation , Cells, Cultured , Humans , Oxygen/metabolism , Pluripotent Stem Cells/cytology
5.
Mol Biol Rep ; 39(3): 3185-96, 2012 Mar.
Article in English | MEDLINE | ID: mdl-21688146

ABSTRACT

The B-lymphocyte accessory molecule Ig-alpha (Ig-α) is encoded by the mouse B cell-specific gene (mb-1), and along with the Ig-beta (Ig-ß) molecule and a membrane bound immunoglobulin (mIg) makes up the B-cell receptor (BCR). Ig-α and Ig-ß form a heterodimer structure that upon antigen binding and receptor clustering primarily initiates and controls BCR intracellular signaling via a phosphorylation cascade, ultimately triggering an effector response. The signaling capacity of Ig-α is contained within its immunoreceptor tyrosine-based activation motif (ITAM), which is also a key component for intracellular signaling initiation in other immune cell-specific receptors. Although numerous studies have been devoted to the mb-1 gene product, Ig-α, and its signaling mechanism, an evolutionary analysis of the mb-1 gene has been lacking until now. In this study, mb-1 coding sequences from 19 species were compared using Bayesian inference. Analysis revealed a gene phylogeny consistent with an expected species divergence pattern, clustering species from the primate order separate from lower mammals and other species. In addition, an overall comparison of non-synonymous and synonymous nucleotide mutational changes suggests that the mb-1 gene has undergone purifying selection throughout its evolution.


Subject(s)
CD79 Antigens/genetics , Evolution, Molecular , Phylogeny , Receptors, Antigen, B-Cell/genetics , Selection, Genetic , Signal Transduction/physiology , Amino Acid Sequence , Animals , Base Sequence , Bayes Theorem , CD79 Antigens/metabolism , Computational Biology , Conserved Sequence/genetics , Mice , Models, Genetic , Molecular Sequence Data , Phosphorylation , Sequence Alignment , Species Specificity
6.
Acta Histochem ; 113(7): 729-42, 2011 Nov.
Article in English | MEDLINE | ID: mdl-21145576

ABSTRACT

Mantle cell lymphoma and small lymphocytic lymphoma are lymphocyte cancers that have similar morphologies and a common age of onset. Mantle cell lymphoma is generally an aggressive B cell lymphoma with a short median survival time, whereas small lymphocytic lymphoma is typically an indolent B cell lymphoma with a prolonged median survival time. Using primary tumor samples in bi-directional suppression subtractive hybridization, we identified genes with differential expression in an aggressive mantle cell lymphoma versus an indolent small lymphocytic lymphoma. "Virtual" Northern blot analyses of multiple lymphoma samples confirmed that a set of genes was preferentially expressed in aggressive mantle cell lymphoma compared to indolent small lymphocytic lymphoma. These analyses identified mantle cell lymphoma-specific genes that may be involved in the aggressive behavior of mantle cell lymphoma and possibly other aggressive human lymphomas. Interestingly, most of these differentially expressed genes have not been identified using other techniques, highlighting the unique ability of suppression subtractive hybridization to identify potentially rare or low expression genes.


Subject(s)
Lymphoma, Mantle-Cell/genetics , Lymphoma, Mantle-Cell/pathology , DNA, Complementary/genetics , Humans , Leukemia, Lymphocytic, Chronic, B-Cell/genetics , Leukemia, Lymphocytic, Chronic, B-Cell/pathology , Phenotype , Sequence Analysis, DNA
7.
Am J Pathol ; 175(4): 1653-61, 2009 Oct.
Article in English | MEDLINE | ID: mdl-19717643

ABSTRACT

B-cell lymphomas, the most frequent human immune system malignancies, often contain dysregulated TCL1 oncogene expression. TCL1 transgenic (TCL1-tg) mice develop a spectrum of B-cell malignancies, supporting an oncogenic role for TCL1 in B cells. Our prior global survey of DNA methylation patterns in TCL1-tg B-cell lymphomas identified many lymphoma-specific candidate hypermethylated genes, including Stk39. The Stk39 encoded protein, sterile 20-like-related proline-alanine-rich kinase (SPAK), regulates cell stress responses, and microarray studies identified reduced SPAK expression in metastatic prostate and treatment-resistant breast cancers, suggesting that its loss may have a role in cancer progression. Here we identified DNA hypermethylation and SPAK silencing in TCL1-tg B-cell lymphomas and SPAK silencing without DNA methylation in multiple subtypes of human B-cell lymphomas. SPAK knockdown by shRNA protected B cells from caspase-dependent apoptosis induced by DNA double-strand breaks but not apoptosis in response to osmotic or oxidative cell stressors. Caspase 3 activation by cleavage was impaired with SPAK repression in DNA damaged B cells. Interestingly, c-Jun NH(2)-terminal kinase is potentially activated by SPAK and pharmacological inhibition of c-Jun NH(2)-terminal kinase in SPAK-expressing B cells recapitulated the cell-protective phenotype of SPAK knockdown. Taken together, these data indicate that SPAK loss in B-cell lymphomas promotes increased cell survival with DNA damage and provides a potential mechanism for increased resistance to genotoxic stress in cancer.


Subject(s)
Apoptosis , DNA Damage , Gene Silencing , Lymphoma, B-Cell/enzymology , Lymphoma, B-Cell/pathology , Protein Serine-Threonine Kinases/metabolism , Animals , Caspase 3/metabolism , DNA Breaks, Double-Stranded , DNA Methylation , Enzyme Activation , Humans , Lymphoma, B-Cell/genetics , Mice , Mice, Transgenic , Proto-Oncogene Proteins/metabolism
8.
J Mol Biol ; 362(2): 173-83, 2006 Sep 15.
Article in English | MEDLINE | ID: mdl-16920149

ABSTRACT

B cell-specific B29 (Igbeta, CD79b) genes in rat, mouse, and human are situated between the 5' growth hormone (GH) locus control region and the 3' GH gene cluster. The entire GH genomic region is DNase 1 hypersensitive in GH-expressing pituitary cells, which predicts an "open" chromatin configuration, and yet B29 is not expressed. The B29 promoter and enhancers exhibit histone deacetylation in pituitary cells, but histone deacetylase inhibition failed to activate B29 expression. The B29 promoter and a 3' enhancer showed local dense DNA methylation in both pituitary and non-lymphoid cells consistent with gene silencing. However, DNA methyltransferase inhibition did not activate B29 expression either. B29 promoter constructs were minimally activated in transfected pituitary cells. Co-transfection of the B cell-specific octamer transcriptional co-activator Bob1 with the B29 promoter construct resulted in high level promoter activity in pituitary cells comparable to B29 promoter activity in transfected B cells. Unexpectedly, inclusion of the B29 3' enhancer in B29 promoter constructs strongly inhibited B29 transcriptional activity even when pituitary cells were co-transfected with Bob1. Both Oct-1 and Pit-1 bind the B29 3' enhancer in in vitro electrophoretic mobility shift assay and in in vivo chromatin immunoprecipitation analyses. These data indicate that the GH locus-embedded, tissue-specific B29 gene is silenced in GH-expressing pituitary cells by epigenetic mechanisms, the lack of a B cell-specific transcription factor, and likely by the B29 3' enhancer acting as a powerful silencer in a context and tissue-specific manner.


Subject(s)
CD79 Antigens/genetics , Enhancer Elements, Genetic , Gene Expression Regulation , Gene Silencing , Immunologic Factors/genetics , Pituitary Gland/cytology , Animals , Azacitidine/metabolism , CD79 Antigens/metabolism , Cell Line , Enzyme Inhibitors/metabolism , Histone Deacetylase Inhibitors , Humans , Hydroxamic Acids/metabolism , Immunologic Factors/metabolism , Mice , Octamer Transcription Factor-1/metabolism , Pituitary Gland/immunology , Promoter Regions, Genetic , Rats , Trans-Activators/genetics , Trans-Activators/metabolism , Transcription Factor Pit-1/metabolism
9.
J Mol Biol ; 350(4): 631-40, 2005 Jul 22.
Article in English | MEDLINE | ID: mdl-15967459

ABSTRACT

Hodgkin and Reed-Sternberg (HRS) cells of classical Hodgkin lymphoma (cHL) and primary effusion lymphoma (PEL) are derived from germinal center (GC) and post-GC B cells, respectively. Neither express many of the B cell genes or surface markers typically expressed by other GC-derived B cell lymphomas or normal B cells. This loss of B cell gene expression is not due to a lack of essential transcription factors, as studies have shown that the ectopic expression of missing transcription factors failed to reactivate endogenous target genes. These results implicate epigenetic mechanisms extinguishing B cell gene expression. Silenced endogenous B cell genes representing a surface receptor, B29 (Igbeta, CD79b), a signaling molecule, TCL1, and a transcription factor, Bob1 (OCA-B, OBF-1), were reactivated by 5-aza-2'-deoxycytidine, indicating that gene silencing in HRS and PEL cells is due to DNA methylation. Genomic bisulfite sequencing corroborated this prediction and revealed three distinct patterns of methylation for the silenced B29 and TCL1 promoters. These distinct patterns consisted of 5' promoter CpG methylation alone, 5' and 3' promoter CpG methylation sparing sites in the central cores, and complete CpG methylation throughout the promoter regions. The silenced Bob1 promoter showed one pattern of dense CpG methylation at essentially all sites. These consistent patterns predict that, although gene silencing in many HRS and PEL cells mimics appropriate gene silencing, in some cases of complete CpG methylation throughout entire promoters both the activation and targeting of methylation is abnormal.


Subject(s)
B-Lymphocytes/metabolism , DNA Methylation , Gene Silencing , Hodgkin Disease/metabolism , Lymphoma, B-Cell/metabolism , Promoter Regions, Genetic , Base Sequence , Cell Line, Tumor , Hodgkin Disease/genetics , Humans , Molecular Sequence Data , Sequence Analysis, DNA
10.
Exp Mol Pathol ; 74(2): 129-39, 2003 Apr.
Article in English | MEDLINE | ID: mdl-12710944

ABSTRACT

Diffuse large B-cell lymphoma (DLBCL) is more prevalent and more often fatal in AIDS patients compared to immune-competent individuals. Potential explanations for these differences include distinct tumorigenic mechanisms and/or altered cellular microenvironments. We previously discovered that the TCL1 (T-cell leukemia-1) proto-oncogene is expressed in a high proportion of AIDS-DLBCL compared to DLBCL cases and that aberrant TCL1 expression causes DLBCL in a new transgenic mouse model. Here, we continue to search for other genes that may contribute to the differential pathogenesis of DLBCL in AIDS. Gene subtraction yielded over 1800 potential AIDS-DLBCL candidates, of which about 50% were unknown and not further considered. The remaining 50% of genes were annotated and, when combined with miniarray screening from multiple patient samples, were reduced to 18 candidate genes for extended analysis. These 18 genes showed distinct patterns of expression in both AIDS-DLBCL and DLBCL samples. However, unlike TCL1, none of these genes was preferentially associated with either AIDS-DLBCL or DLBCL. Our data suggest that the increased incidence and severity of AIDS-DLBCL compared to DLBCL is likely due to crippled immune surveillance rather than to markedly different gene expression profiles.


Subject(s)
Gene Expression Regulation, Neoplastic , Lymphoma, AIDS-Related/metabolism , DNA, Complementary/genetics , Gene Expression Profiling , Genetic Heterogeneity , Humans , Lymphoma, AIDS-Related/genetics , Lymphoma, Large B-Cell, Diffuse/metabolism , Mutation , Nucleic Acid Hybridization , Oligonucleotide Array Sequence Analysis , Proto-Oncogene Mas , RNA, Messenger/genetics , RNA, Messenger/metabolism , Tumor Cells, Cultured
11.
J Biol Chem ; 278(2): 948-55, 2003 Jan 10.
Article in English | MEDLINE | ID: mdl-12421830

ABSTRACT

Cis-regions and trans-factors controlling TCL1 oncogene expression are not known. We identified the functional TCL1 promoter by mapping four transcriptional start sites 24-30 bp downstream of a TATA box. A 424-bp fragment upstream of the major start site showed robust promoter activity comparable with SV40 in both TCL1 expressing and non-expressing cell lines. Additional constructs spanning 10 kb upstream and 20 kb downstream of the start site showed only modest increases in reporter activity indicating that TCL1 expression is primarily controlled by the promoter. Ten putative Sp1-binding sites were identified within 300 bp of the start site, and three of these specifically bound Sp1. A dose-dependent transactivation of the TCL1 promoter with Sp1 addition in Sp1-negative Drosophila SL2 cells was observed, and mutation of the three identified Sp1-binding sites significantly repressed reporter gene expression in 293T cells, confirming a key role for Sp1 in activating the TCL1 promoter in vivo. In TCL1 silent cell lines, CpG DNA methylation was rarely observed at functional Sp1 sites, and methylation of a previously reported NotI restriction site was associated with dense CpG methylation rather than endogenous TCL1 gene silencing. Together, these results indicate that Sp1 mediates transactivation of the TCL1 core promoter and that TCL1 gene silencing is not dependent on mechanisms involving Sp1 and NotI site methylation.


Subject(s)
Proto-Oncogene Proteins/genetics , Proto-Oncogenes , Sp1 Transcription Factor/physiology , Transcriptional Activation , Base Sequence , Cell Line , CpG Islands , DNA Methylation , Humans , Molecular Sequence Data , Promoter Regions, Genetic
12.
Proc Natl Acad Sci U S A ; 99(22): 14392-7, 2002 Oct 29.
Article in English | MEDLINE | ID: mdl-12381789

ABSTRACT

The TCL1 protooncogene is overexpressed in many mature B cell lymphomas, especially from AIDS patients. To determine whether aberrant expression promotes B cell transformation, we generated a murine model in which a TCL1 transgene was overexpressed at similar levels in both B and T cells. Strikingly, transgenic mice developed Burkitt-like lymphoma (BLL) and diffuse large B cell lymphoma (DLBCL) with attendant Bcl-6 expression and mutated J(H) gene segments at a very high penetrance beginning at 4 months of age. In contrast, only one mouse developed a T cell malignancy at 15 months, consistent with a longer latency for transformation of T cells by TCL1. Activation of premalignant splenic B cells by means of B cell antigen receptor (BCR) engagement resulted in significantly increased proliferation and augmented AKT-dependent signaling, including increased S6 ribosomal protein phosphorylation. Transgenic spleen cells also survived longer than wild-type spleen cells in long-term culture. Together these data demonstrate that TCL1 is a powerful oncogene that, when overexpressed in both B and T cells, predominantly yields mature B cell lymphomas.


Subject(s)
Burkitt Lymphoma/metabolism , Lymphoma, Large B-Cell, Diffuse/metabolism , Protein Serine-Threonine Kinases , Proto-Oncogene Proteins/metabolism , Animals , Apoptosis , B-Lymphocytes/cytology , B-Lymphocytes/metabolism , Cell Division , Female , Gene Expression , Lymphocytosis/metabolism , Lymphoproliferative Disorders/metabolism , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic , Proto-Oncogene Proteins/genetics , Proto-Oncogene Proteins c-akt , Ribosomal Protein S6/metabolism , Spleen/cytology , T-Lymphocytes/metabolism , Time Factors , Transformation, Genetic
14.
Biochemistry ; 41(20): 6376-82, 2002 May 21.
Article in English | MEDLINE | ID: mdl-12009899

ABSTRACT

AKT has a critical role in relaying cell survival and proliferation signals initiated by ligand binding to surface receptors in mammalian cells. Induction of AKT serine/threonine kinase activity is augmented by the T-cell leukemia-1 (TCL1) oncoprotein through a physical association requiring the AKT pleckstrin homology domain. Here, we used molecular modeling and identified an exposed hydrophobic patch composed of two discontinuous amino acid stretches near one end of the TCL1 beta-barrel that was required for a TCL1-AKT association. Site-directed mutations of this region did not affect TCL1 secondary structure, yet they disrupted interactions with AKT. This region was found in other members of the TCL1 oncoprotein family, such as TCL1b and MTCP1, and suggested a conserved, novel AKT binding domain. Interestingly, TCL1 and AKT co-localize in multiple cell compartments, but only extracts from the plasma membrane stimulate optimal complex formation in vitro. Identification of an AKT binding domain on TCL1 is an important step in deciphering the complex interactions that regulate AKT kinase activity in lymphocyte development and neoplasia within the immune system.


Subject(s)
Cytoplasm/metabolism , DNA-Binding Proteins/chemistry , DNA-Binding Proteins/metabolism , Intracellular Membranes/metabolism , Models, Molecular , Oncogene Proteins/chemistry , Oncogene Proteins/metabolism , Proto-Oncogene Proteins/metabolism , Transcription Factors/chemistry , Transcription Factors/metabolism , Animals , Cell Line , Cell Membrane/metabolism , Cell Nucleus/metabolism , Cytoplasm/genetics , DNA-Binding Proteins/genetics , Group II Phospholipases A2 , Humans , Hydrophobic and Hydrophilic Interactions , Jurkat Cells , Leukemia, T-Cell/metabolism , Mice , Oncogene Proteins/genetics , Phospholipases A/metabolism , Protein Biosynthesis , Protein Serine-Threonine Kinases/metabolism , Protein Structure, Tertiary , Proto-Oncogene Proteins/genetics , Proto-Oncogene Proteins c-akt , Transcription Factors/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...