Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 11 de 11
Filter
Add more filters










Publication year range
1.
J Stroke Cerebrovasc Dis ; 28(3): 595-603, 2019 Mar.
Article in English | MEDLINE | ID: mdl-30482485

ABSTRACT

BACKGROUND: Survival following cardiac arrest (CA) and subsequent cardiopulmonary resuscitation (CPR), to a great extent, depends on brain damage. Adipose-derived stem cells (ADSCs), as a source of paracrine growth factors and the capacity of neural differentiation may reduce this brain damage. OBJECTIVE: The purpose of this study is to evaluate the protection of ADSCs to brain damage following CPR. METHODS: Rats were divided into 3 groups, sham, CA, and ADSCs group. Rats in sham group went through sham surgery. Rats in CA group went through CA, CPR, and injection PBS (phosphate buffer saline). Rats in ADSCs group went through CA, CPR, and intravenous injection of ADSCs. Rats in sham group were sacrificed immediately after operation. At 24, 72, and 168 hours after return of spontaneous circulation operation, rats in CA and ADSCs group were randomly selected and sacrificed. Brain damage was evaluated by using Neurological Deficit Scale (NDS) score, hippocampal pathology, serum level of S100ß, and apoptosis ratio of hippocampal neurons. Protein of brain derived neurotrophic factor (BDNF) and IL-6 (interleukin-6) in the hippocampus were detected. RESULTS: Compared with sham group, CA and ADSCs group showed a decrease in NDS score, an increased apoptosis ratio of hippocampal nerve cells, increased serum level of S100-ß, and a significant increase in neuroprotective IL-6 and BDNF. In comparison to CA group, ADSCs group had a mild degree of brain damage and higher expression of IL-6 and BDNF. CONCLUSIONS: In the acute stage of cerebral injury following CA, ADSCs might improve the prognosis of brain damage by stimulating the expression of neuroprotective IL-6 and BDNF.


Subject(s)
Adipose Tissue/transplantation , Heart Arrest/complications , Hippocampus/pathology , Hypoxia, Brain/prevention & control , Neurons/pathology , Stem Cell Transplantation , Adipose Tissue/cytology , Adipose Tissue/metabolism , Animals , Apoptosis , Behavior, Animal , Biomarkers/blood , Brain-Derived Neurotrophic Factor/metabolism , Cardiopulmonary Resuscitation , Cells, Cultured , Disease Models, Animal , Heart Arrest/physiopathology , Heart Arrest/therapy , Hippocampus/metabolism , Hippocampus/physiopathology , Humans , Hypoxia, Brain/etiology , Hypoxia, Brain/metabolism , Hypoxia, Brain/pathology , Interleukin-6/metabolism , Male , Neurons/metabolism , Paracrine Communication , Rats, Sprague-Dawley , S100 Calcium Binding Protein beta Subunit/blood , Signal Transduction
2.
Oncotarget ; 9(32): 22631-22641, 2018 Apr 27.
Article in English | MEDLINE | ID: mdl-29854303

ABSTRACT

INTRODUCTION: Clear-cell renal cell carcinoma (ccRCC) is the sixth most common malignancy in men in North America. Since ccRCC is a malignancy dependent on neovascularization, current first line systemic therapies like sunitinib, target the formation of new vessels allowing nutrient deprivation and cell death. However, recent studies have shown that patients develop resistance after approximately 1 year of treatment and show disease progression while on therapy. Therefore, we propose to identify the protein(s) responsible for increased migration with the aim of developing a new therapy that will target the identified protein and potentially slow down the progression of the disease. MATERIAL AND METHODS: Human renal cancer cell lines (Caki-1, Caki-2, ACHN) were treated with increasing doses of sunitinib to develop a sunitinib-conditioned renal cell carcinoma cell line. mRNA microarray and qPCR were performed to compare the differences in gene expression between Caki-1 sunitinib-conditioned and non-conditioned cells. NTN1 was assessed in our in vivo sunitinib-conditioned mouse model using immunostaining. xCELLigence and scratch assays were used to evaluate migration and MTS was used to evaluate cell viability. RESULTS: Human renal cell carcinoma sunitinib-conditioned cell lines showed upregulation of netrin-1 in microarray and q-PCR. Increased migration was demonstrated in Caki-1 sunitinib-conditioned cells when compared to the non-treated ones as well as, increased endothelial cell migration. Silencing of netrin-1 in sunitinib-conditioned Caki-1 cells did not demonstrate a significant reduction in cell migration. CONCLUSION: Netrin-1 is highly upregulated in renal cell carcinoma treated with sunitinib, but has no influence on cell viability or cell migration in metastatic RCC.

3.
Blood ; 117(19): 5057-66, 2011 May 12.
Article in English | MEDLINE | ID: mdl-21343612

ABSTRACT

Fetal liver and adult bone marrow hematopoietic stem cells (HSCs) renew or differentiate into committed progenitors to generate all blood cells. PRDM16 is involved in human leukemic translocations and is expressed highly in some karyotypically normal acute myeloblastic leukemias. As many genes involved in leukemogenic fusions play a role in normal hematopoiesis, we analyzed the role of Prdm16 in the biology of HSCs using Prdm16-deficient mice. We show here that, within the hematopoietic system, Prdm16 is expressed very selectively in the earliest stem and progenitor compartments, and, consistent with this expression pattern, is critical for the establishment and maintenance of the HSC pool during development and after transplantation. Prdm16 deletion enhances apoptosis and cycling of HSCs. Expression analysis revealed that Prdm16 regulates a remarkable number of genes that, based on knockout models, both enhance and suppress HSC function, and affect quiescence, cell cycling, renewal, differentiation, and apoptosis to various extents. These data suggest that Prdm16 may be a critical node in a network that contains negative and positive feedback loops and integrates HSC renewal, quiescence, apoptosis, and differentiation.


Subject(s)
Cell Differentiation/physiology , DNA-Binding Proteins/metabolism , Hematopoiesis/physiology , Hematopoietic Stem Cells/cytology , Transcription Factors/metabolism , Animals , Apoptosis/physiology , Cell Separation , DNA-Binding Proteins/genetics , Flow Cytometry , Gene Expression , Gene Expression Profiling , Genotype , Hematopoietic Stem Cells/physiology , Mice , Mice, Inbred C57BL , Mice, Knockout , Reverse Transcriptase Polymerase Chain Reaction , Transcription Factors/genetics
4.
Glia ; 58(13): 1640-8, 2010 Oct.
Article in English | MEDLINE | ID: mdl-20578042

ABSTRACT

HIV-1 Tat protein is an important pathogenic factor in HIV-1-associated neurological diseases. One hallmark of HIV-1 infection of the central nervous system (CNS) is astrocytosis, which is characterized by elevated glial fibrillary acidic protein (GFAP) expression in astrocytes. We have shown that Tat activates GFAP expression in astrocytes [Zhou et al., (2004) Mol Cell Neurosci 27:296-305] and that GFAP is an important regulator of Tat neurotoxicity [Zou et al., (2007) Am J Pathol 171:1293-1935]. However, the underlying mechanisms for Tat-mediated GFAP up-regulation are not understood. In this study, we reported concurrent up-regulation of adenovirus E1a-associated 300 kDa protein p300 and GFAP in Tat-expressing human astrocytoma cells and primary astrocytes. We showed that p300 was indeed induced by Tat expression and HIV-1 infection and that the induction occurred at the transcriptional level through the cis-acting elements of early growth response 1 (egr-1) within its promoter. Using siRNA, we further showed that p300 regulated both constitutive and Tat-mediated GFAP expression. Moreover, we showed that ectopic expression of p300 potentiated Tat transactivation activity and increased proliferation of HIV-1-infected astrocytes, but had little effect on HIV-1 replication in these cells. Taken together, these results demonstrate for the first time that Tat is a positive regulator of p300 expression, which in turn regulates GFAP expression, and suggest that the Tat-Egr-1-p300-GFAP axis likely contributes to Tat neurotoxicity and predisposes astrocytes to be an HIV-1 sanctuary in the CNS.


Subject(s)
Astrocytes/metabolism , E1A-Associated p300 Protein/metabolism , Gene Expression Regulation/physiology , Glial Fibrillary Acidic Protein/metabolism , Up-Regulation/physiology , tat Gene Products, Human Immunodeficiency Virus/adverse effects , Astrocytes/drug effects , Cell Line, Tumor , Cell Proliferation/drug effects , Early Growth Response Protein 1/metabolism , Gene Expression Regulation/drug effects , Glial Fibrillary Acidic Protein/genetics , Green Fluorescent Proteins/genetics , HIV Infections/metabolism , Humans , RNA, Small Interfering/pharmacology , Tetrazolium Salts , Thiazoles , Thymidine/metabolism , Transcriptional Activation , Transfection , Tritium/metabolism , Up-Regulation/drug effects , beta-Galactosidase/metabolism
5.
Nano Lett ; 8(10): 3126-30, 2008 Oct.
Article in English | MEDLINE | ID: mdl-18754690

ABSTRACT

We develop biodegradable polymeric nanoparticles to facilitate nonviral gene transfer to human embryonic stem cells (hESCs). Small (approximately 200 nm), positively charged (approximately 10 mV) particles are formed by the self assembly of cationic, hydrolytically degradable poly(beta-amino esters) and plasmid DNA. By varying the end group of the polymer, we can tune the biophysical properties of the resulting nanoparticles and their gene-delivery efficacy. We created an OCT4-driven GFP hES cell line to allow the rapid identification of nanoparticles that facilitate gene transfer while maintaining an hESC undifferentiated state. Using this cell system, we synthesized nanoparticles that have gene delivery efficacy that is up to 4 times higher than that of the leading commercially available transfection agent, Lipofectamine 2000. Importantly, these materials have minimal toxicity and do not adversely affect hESC colony morphology or cause nonspecific differentiation.


Subject(s)
Embryonic Stem Cells/cytology , Gene Transfer Techniques , Genetic Vectors/chemistry , Animals , Biocompatible Materials/chemistry , Cations , Cell Differentiation , Flow Cytometry , Genetic Techniques , Green Fluorescent Proteins/metabolism , Hydrolysis , Mice , Nanotechnology/methods , Octamer Transcription Factor-3/metabolism , Polymers/chemistry
6.
Am J Pathol ; 171(6): 1923-35, 2007 Dec.
Article in English | MEDLINE | ID: mdl-18055541

ABSTRACT

Human immunodeficiency virus (HIV)-1 Tat protein is an important pathogenic factor in HIV-associated neuropathogenesis. Despite recent progress, the molecular mechanisms underlying Tat neurotoxicity are still not completely understood. However, few therapeutics have been developed to specifically target HIV infection in the brain. Recent development of an inducible brain-specific Tat transgenic mouse model has made it possible to define the mechanisms of Tat neurotoxicity and evaluate anti-neuroAIDS therapeutic candidates in the context of a whole organism. Herein, we demonstrate that administration of EGb 761, a standardized formulation of Ginkgo biloba extract, markedly protected Tat transgenic mice from Tat-induced developmental retardation, inflammation, death, astrocytosis, and neuron loss. EGb 761 directly down-regulated glial fibrillary acidic protein (GFAP) expression at both protein and mRNA levels. This down-regulation was, at least in part, attributable to direct effects of EGb 761 on the interactions of the AP1 and NF-kappaB transcription factors with the GFAP promoter. Most strikingly, Tat-induced neuropathological phenotypes including macrophage/microglia activation, central nervous system infiltration of T lymphocytes, and oxidative stress were significantly alleviated in GFAP-null/Tat transgenic mice. Taken together, these results provide the first evidence to support the potential for clinical use of EGb 761 to treat HIV-associated neurological diseases. Moreover, these findings suggest for the first time that GFAP activation is directly involved in Tat neurotoxicity, supporting the notion that astrocyte activation or astrocytosis may directly contribute to HIV-associated neurological disorders.


Subject(s)
Glial Fibrillary Acidic Protein/antagonists & inhibitors , HIV-1 , Neurotoxicity Syndromes/drug therapy , Plant Extracts/therapeutic use , tat Gene Products, Human Immunodeficiency Virus/antagonists & inhibitors , tat Gene Products, Human Immunodeficiency Virus/toxicity , Animals , Apoptosis/drug effects , Astrocytes/drug effects , Astrocytes/metabolism , Astrocytes/virology , Cell Line, Tumor , Ginkgo biloba , Glial Fibrillary Acidic Protein/genetics , Glial Fibrillary Acidic Protein/metabolism , Humans , Mice , Mice, Transgenic , Neurotoxicity Syndromes/pathology , Neurotoxicity Syndromes/virology , Plant Extracts/pharmacology , Transcription, Genetic/drug effects , tat Gene Products, Human Immunodeficiency Virus/genetics
7.
Cell Res ; 17(1): 62-72, 2007 Jan.
Article in English | MEDLINE | ID: mdl-17211448

ABSTRACT

We previously showed that Wnt3a could stimulate human embryonic stem (hES) cell proliferation and affect cell fate determination. In the absence of feeder cell-derived factors, hES cells cultured under a feeder-free condition survived and proliferated poorly. Adding recombinant Wnt3a in the absence of feeder cell derived-factors stimulated hES cell proliferation but also differentiation. In the present study, we further extended our analysis to other Wnt ligands such as Wnt1 and Wnt5a. While Wnt1 displayed a similar effect on hES cells as Wnt3a, Wnt5a had little effect in this system. Wnt3a and Wnt1 enhanced proliferation of undifferentiated hES cells when feeder-derived self-renewal factors and bFGF are also present. To explore the possibility to promote the proliferation of undifferentiated hES cells by activating the Wnt signaling, we overexpressed Wnt3a or Wnt1 gene in immortalized human adult fibroblast (HAFi) cells that are superior in supporting long-term growth of undifferentiated hES cells than primary mouse embryonic fibroblasts. HAFi cells with or without a Wnt transgene can be propagated indefinitely. Over-expression of the Wnt3a gene significantly enhanced the ability of HAFi feeder cells to support the undifferentiated growth of 3 different hES cell lines we tested. Co-expression of three commonly-used drug selection genes in Wnt3a-overpressing HAFi cells further enabled us to select rare hES clones after stable transfection or transduction. These immortalized engineered feeder cells (W3R) that co-express growth-promoting genes such as Wnt3a and three drug selection genes should empower us to efficiently make genetic modified hES cell lines for basic and translational research.


Subject(s)
Cell Differentiation , Embryonic Stem Cells/cytology , Embryonic Stem Cells/metabolism , Signal Transduction/physiology , Wnt Proteins/physiology , Cell Culture Techniques/methods , Cell Differentiation/physiology , Cell Division/physiology , Cell Line, Transformed , Cell Proliferation , Humans , Proto-Oncogene Proteins/genetics , Wnt Proteins/genetics , Wnt-5a Protein , Wnt1 Protein/genetics , Wnt3 Protein , Wnt3A Protein
8.
Stem Cells ; 25(3): 779-89, 2007 Mar.
Article in English | MEDLINE | ID: mdl-17158240

ABSTRACT

We report here a lentiviral vector system for regulated transgene expression. We used the tetracycline repressor fused with a transcriptional suppression domain (tTS) to specifically suppress transgene expression. Human cells were first transduced with a tTS-expressing vector and subsequently transduced with a second lentiviral vector-containing transgene controlled by a regular promoter adjacent to a high-affinity tTS-binding site (tetO). After optimizing the location of the tetO site in the latter vector, we achieved a better inducible transgene expression than the previous lentiviral vectors using the tetracycline repressor systems. In this new system, the transgene transcription from a cellular promoter such as EF1alpha or ubiquitin-C promoter is suppressed by the tTS bound to the nearby tetO site. In the presence of the tetracycline analog doxycycline (Dox), however, the tTS binding is released from the transgene vector and transcription from the promoter is restored. Thus, this system simply adds an extra level of regulation, suitable for any types of promoters (ubiquitous or cell-specific). We tested this tTS-suppressive, Dox-inducible system in 293T cells, human multipotent hematopoietic progenitor cells, and three human embryonic stem cell lines, using a dual-gene vector containing the green fluorescent protein reporter or a cellular gene. We observed a tight suppression in the uninduced state. However, the suppression is reversible, and transgene expression was restored at 5 ng/ml Dox. The lentiviral vectors containing the tTS-suppressive, Dox-inducible system offer a universal, inducible, and reversible transgene expression system in essentially any mammalian cell types, including human embryonic stem cells.


Subject(s)
Gene Expression Regulation, Neoplastic , Gene Transfer Techniques , Cell Line, Tumor , Flow Cytometry , Genetic Vectors , Green Fluorescent Proteins/genetics , Humans , Lentivirus/genetics , Promoter Regions, Genetic , Transcription, Genetic
9.
Mol Cell Neurosci ; 27(3): 296-305, 2004 Nov.
Article in English | MEDLINE | ID: mdl-15519244

ABSTRACT

Human immunodeficiency virus type 1 (HIV-1) Tat protein plays an important role in HIV-associated neuropathogenesis. Astrocytosis and neuron death are two hallmarks of HIV-1 infection of the central nervous system (CNS). However, whether there is a direct link between Tat expression, astrocytosis and subsequent neuron death is not known. In this study, we expressed Tat in astrocytes and examined Tat effects on astrocyte function and subsequent neuronal survival. The results showed that Tat expression resulted in a significant increase in glial fibrillary acidic protein (GFAP) expression, a cellular marker of astrocyte activation or astrocytosis. The GFAP promoter-driven reporter gene assay showed that Tat transactivated GFAP expression at the transcriptional level. Furthermore, Tat expression markedly impaired glutamate uptake by astrocytes. Importantly, cell culture supernatants from Tat-expressing astrocytes induced dramatic neuron death. Taken together, these data provide evidence for the first time to directly link Tat expression in astrocytes to astrocytosis, astrocyte dysfunction, and subsequent neuron death. In addition, these data suggest that astrocyte dysfunction contributes, at least in part, to Tat neurotoxicity and subsequently HIV-associated neuropathogenesis.


Subject(s)
Astrocytes/metabolism , Gene Expression Regulation, Viral/physiology , Gene Products, tat/biosynthesis , Gene Products, tat/physiology , HIV-1/metabolism , Neurons/cytology , Neurons/metabolism , Animals , Astrocytes/virology , Cell Death/physiology , Cell Line, Tumor , Cells, Cultured , Gene Products, tat/genetics , Humans , Mice , Mice, Transgenic , Neurons/virology , tat Gene Products, Human Immunodeficiency Virus
10.
Neurosci Lett ; 359(3): 155-8, 2004 Apr 15.
Article in English | MEDLINE | ID: mdl-15050687

ABSTRACT

Human immunodeficiency virus type 1 Tat protein is one of the soluble neurotoxins. Most studies have to date focused on Tat as an extracellular molecule and its role in neuronal apoptosis, as recombinant Tat protein is often used in these studies. In this study, we expressed Tat protein in astrocytes and neurons, and examined its effects on these cells. We found that Tat expression resulted in growth inhibition of astrocytes, neurons, as well as non-glial cells 293T. We further showed that Tat interacted with a number of cell cycle-related proteins including cyclin A, cyclin B, cyclin D3, Cdk2, Cdk4, Cdk1/Cdc2, cdc6, p27, p53, p63, hdlg, and PCNA. These data demonstrate that Tat inhibited cell proliferation when expressed intracellularly, and suggest that Tat interactions with multiple cell cycle regulators may account for this anti-proliferative effect. These results support the notion that Tat-induced neuropathogenesis is mediated by multiple mechanisms involving both intracellular and extracellular Tat protein.


Subject(s)
Astrocytes/metabolism , Cell Cycle Proteins/metabolism , Cell Cycle , Growth Inhibitors/metabolism , Intracellular Fluid/metabolism , Kidney/metabolism , Neurons/metabolism , Trans-Activators/metabolism , Animals , Astrocytes/pathology , Cell Division , Cell Line , Cell Line, Tumor , Cell Survival , Humans , Kidney/embryology , Kidney/pathology , Neuroglia/metabolism , Neuroglia/pathology , Neurons/pathology , Rats , Recombinant Proteins/metabolism , Trans-Activators/genetics
11.
J Immunol ; 172(3): 1888-95, 2004 Feb 01.
Article in English | MEDLINE | ID: mdl-14734774

ABSTRACT

HIV-1 Tat has been proposed as a key agent in many AIDS-related disorders, including HIV-1-associated neurological diseases. We have recently shown that Tat expression induces a significant increase in T lymphocytes in the brains of Tat transgenic mice. The CNS infiltration of T lymphocytes has been noted in AIDS patients. In the present study using this unique genetic system we attempted to understand the underlying mechanisms of Tat expression-induced infiltration of T lymphocytes by examining chemokine expression. RNase protection assay revealed that in addition to CCL2 (monocyte chemoattractant protein-1), CCL3 (macrophage inflammatory protein-1alpha (MIP-1alpha)), CCL4 (MIP-1beta), CCL5 (RANTES), CXCL2 (MIP-2), and CXCL10 (inducing protein-10), XCL1 (lymphotactin/single C motif-1alpha/activation-induced, T cell-derived and chemokine-related cytokine) was identified to be up-regulated by Tat expression. XCL1 is a C chemokine and plays a specific and important role in tissue-specific recruitment of T lymphocytes. Thus, we further determined the relationship between Tat and XCL1 expression. Tat-induced XCL1 expression was further confirmed by XCL1-specific RT-PCR and ELISA. Combined in situ hybridization and immunohistochemical staining identified astrocytes, monocytes, and macrophages/microglia as XCL1-producing cells in vivo. Using human astrocytes, U87.MG cells, as an in vitro model, activation of XCL1 expression was positively correlated with Tat expression. Moreover, the XCL1 promoter-driven reporter gene assay showed that Tat-induced XCL1 expression occurred at the transcriptional level. Taken together, these results demonstrate that Tat directly trans-activated XCL1 expression and suggest potential roles of Tat-induced XCL1 expression in the CNS infiltration of T lymphocytes during HIV-1 infection and subsequent HIV-1-induced neurological diseases.


Subject(s)
Chemokines, C/biosynthesis , Gene Products, tat/physiology , HIV-1/physiology , T-Lymphocytes/immunology , T-Lymphocytes/metabolism , Amino Acid Motifs , Animals , Astrocytes/immunology , Astrocytes/metabolism , Brain/cytology , Brain/immunology , Brain/metabolism , Brain/virology , Cell Line, Tumor , Cells, Cultured , Chemokines, C/chemistry , Chemokines, C/genetics , Gene Expression Regulation , Gene Products, tat/biosynthesis , Gene Products, tat/genetics , HIV-1/genetics , Humans , Lymphocyte Activation , Lymphokines/chemistry , Macrophages/immunology , Macrophages/metabolism , Mice , Mice, Transgenic , Microglia/immunology , Microglia/metabolism , Monocytes/immunology , Monocytes/metabolism , Promoter Regions, Genetic , RNA, Messenger/biosynthesis , Sialoglycoproteins/chemistry , Up-Regulation/genetics , tat Gene Products, Human Immunodeficiency Virus
SELECTION OF CITATIONS
SEARCH DETAIL
...