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










Publication year range
1.
Cell Signal ; 109: 110763, 2023 09.
Article in English | MEDLINE | ID: mdl-37315752

ABSTRACT

Reelin and its receptor, ApoER2, play important roles in prenatal brain development and postnatally in synaptic plasticity, learning, and memory. Previous reports suggest that reelin's central fragment binds to ApoER2 and receptor clustering is involved in subsequent intracellular signaling. However, limitations of currently available assays have not established cellular evidence of ApoER2 clustering upon binding of the central reelin fragment. In the present study, we developed a novel, cell-based assay of ApoER2 dimerization using a "split-luciferase" approach. Specifically, cells were co-transfected with one recombinant ApoER2 receptor fused to the N-terminus of luciferase and one ApoER2 receptor fused to the C-terminus of luciferase. Using this assay, we directly observed basal ApoER2 dimerization/clustering in transfected HEK293T cells and, significantly, an increase in ApoER2 clustering in response to that central fragment of reelin. Furthermore, the central fragment of reelin activated intracellular signal transduction of ApoER2, indicated by increased levels of phosphorylation of Dab1, ERK1/2, and Akt in primary cortical neurons. Functionally, we were able to demonstrate that injection of the central fragment of reelin rescued phenotypic deficits observed in the heterozygous reeler mouse. These data are the first to test the hypothesis that the central fragment of reelin contributes to facilitating the reelin intracellular signaling pathway through receptor clustering.


Subject(s)
Extracellular Matrix Proteins , Serine Endopeptidases , Mice , Animals , Humans , Serine Endopeptidases/genetics , Serine Endopeptidases/metabolism , Extracellular Matrix Proteins/metabolism , Cell Adhesion Molecules, Neuronal/genetics , Cell Adhesion Molecules, Neuronal/metabolism , HEK293 Cells , Nerve Tissue Proteins/metabolism , Signal Transduction/physiology , Disease Models, Animal , Luciferases/metabolism , Cognition , Receptors, LDL/metabolism
2.
J Vis Exp ; (176)2021 10 14.
Article in English | MEDLINE | ID: mdl-34723945

ABSTRACT

Cutaneous T-cell lymphomas (CTCL) are derived from the transformation and uncontrolled proliferation of mature skin-homing T cells, and mycosis fungoides (MF) and Sézary syndrome (SS) represent the most common subtypes. Despite a number of studies on characterizing gene expression, genetic alterations, and epigenetic abnormalities of CTCL, the molecular pathogenesis of MF/SS remains unclear. MF refers to the more common CTCL with a skin-predominance, and is usually limited to skin, whereas SS is an aggressive leukemic variant of CTCL with widespread skin involvement and is characterized by neoplastic distribution mainly involving blood, skin, and lymph node. Focusing on clinical practice, the identification of gene expression biomarkers has enormous potential to improve diagnosis and treatment of MF/SS. Indeed, recent transcriptomic studies have identified potential diagnostic biomarkers from differences in gene expression between normal and malignant T cells, which may improve our understanding of SS biology, and reveal potential therapeutic targets. This manuscript describes a detailed reproducible protocol for the isolation of peripheral blood mononuclear cells from fresh whole blood from patients diagnosed with SS, selection of CD4+ memory T cells (CD4+CD45RO+ T cells), chemical stimulation, and preparation of RNA suitable for transcriptomic profiling to discover novel prognostic molecular markers to gain additional insight in disease etiology. The stimulation using chemical agonist to activate nuclear regulation provides more specific assessment for pathways important in the dynamic transcription regulation and gene expression and eliminates confounding defects that may arise from upstream signaling defects arising from TCR antigen loss at the cell membrane. The data obtained from comparison of transcriptome of unstimulated to stimulated SS T cells unmasks functional regulatory gene expression defects not evident from analysis of quiescent unstimulated cells. Furthermore, the method outlined from this approach can be adapted for studying T cell gene expression defects in other T cell immune diseases.


Subject(s)
Sezary Syndrome , Skin Neoplasms , CD4-Positive T-Lymphocytes , Humans , Leukocytes, Mononuclear/pathology , Sezary Syndrome/diagnosis , Sezary Syndrome/genetics , Skin Neoplasms/pathology , Transcriptome
3.
Oncotarget ; 10(49): 5052-5069, 2019 Aug 20.
Article in English | MEDLINE | ID: mdl-31489115

ABSTRACT

Sézary syndrome (SS) is an aggressive cutaneous T cell lymphoma with pruritic skin inflammation and immune dysfunction, driven by neoplastic, clonal memory T cells in both peripheral blood and skin. To gain insight into abnormal gene expression promoting T cell dysfunction, lymphoproliferation and transformation in SS, we first compared functional transcriptomic profiles of both resting and activated CD4+CD45RO+ T cells from SS patients and normal donors to identified differential expressed genes. Next, a meta-analysis was performed to compare our SS data to public microarray data from a novel benign disease control, lymphocytic-variant hypereosinophilic syndrome (L-HES). L-HES is a rare, clonal lymphoproliferation of abnormal memory T cells that produces similar clinical symptoms as SS, including severe pruritus and eosinophilia. Comparison revealed gene sets specific for either SS (370 genes) or L-HES (519 genes), and a subset of 163 genes that were dysregulated in both SS and L-HES T cells compared to normal donor T cells. Genes confirmed by RT-qPCR included elevated expression of PLS3, TWIST1 and TOX only in SS, while IL17RB mRNA was increased only in L-HES. CDCA7 was increased in both diseases. In an L-HES patient who progressed to peripheral T cell lymphoma, the malignant transformation identified increases in the expression of CDCA7, TIGIT, and TOX, which are highly expressed in SS, suggesting that these genes contribute to neoplastic transformation. In summary, we have identified gene expression biomarkers that implicate a common transformative mechanism and others that are unique to differentiate SS from L-HES.

4.
Neurobiol Aging ; 49: 145-153, 2017 01.
Article in English | MEDLINE | ID: mdl-27810638

ABSTRACT

Presenilin (PS)-1 is an intramembrane protease serving as the catalytic component of γ-secretase. Mutations in the PS1 gene are the most common cause of familial Alzheimer's disease (FAD). The low-density lipoprotein (LDL)-receptor family member apoER2 is a γ-secretase substrate that has been associated with AD in several ways, including acting as a receptor for apolipoprotein E (ApoE). ApoER2 is processed by γ-secretase into a C-terminal fragment (γ-CTF) that appears to regulate gene expression. FAD PS1 mutations were tested for effects on apoER2. PS1 mutation R278I showed impaired γ-secretase activity for apoER2 in the basal state or after exposure to Reelin. PS1 M146V mutation permitted accumulation of apoER2 CTFs after Reelin treatment, whereas no difference was seen between wild-type (WT) and M146V in the basal state. PS1 L282V mutation, combined with the γ-secretase inhibitor N-(N-[3,5-Difluorophenacetyl]-L-alanyl)-S-phenylglycine t-butyl ester, greatly reduced the cell-surface levels of apoER2 without affecting total apoER2 levels, suggesting a defect in receptor trafficking. These findings indicate that impaired processing or localization of apoER2 may contribute to the pathogenic effects of FAD mutations in PS1.


Subject(s)
LDL-Receptor Related Proteins/metabolism , Mutation , Presenilin-1/genetics , Presenilin-1/physiology , Alzheimer Disease/genetics , Amyloid Precursor Protein Secretases/physiology , Animals , Cell Adhesion Molecules, Neuronal , Cells, Cultured , Extracellular Matrix Proteins , Gene Expression , LDL-Receptor Related Proteins/genetics , Mice, Transgenic , Nerve Tissue Proteins , Reelin Protein , Serine Endopeptidases
5.
Cancer Immunol Immunother ; 65(5): 563-73, 2016 May.
Article in English | MEDLINE | ID: mdl-26980480

ABSTRACT

In the dose-escalation phase of a Phase I clinical trial in which six subjects each were vaccinated with PepCan at the 50, 100, 250, and 500 µg per peptide dose, the 50 µg dose showed the best histological regression rate. Ten additional subjects were vaccinated at this dose in the final dose phase. As with the dose-escalation phase, no dose-limiting toxicities were observed. Overall, the histological regression rates were 50% at the 50 µg dose (7 of 14) and 100 µg dose (3 of 6), and 45 % overall (14 of 31). Of subjects in whom HPV type 16 (HPV 16) was detected at entry, it became undetectable in three subjects after vaccination, and the viral loads significantly decreased in nine subjects in whom HPV 16 infection was detected at entry and exit (p = 0.008). Immune profiling revealed increased T-helper type 1 cells after vaccinations (p = 0.02 and 0.0004 after 2 and 4 vaccinations, respectively). T-helper type 2 cells initially increased after two vaccinations (p = 0.01), but decreased below the baseline level after four vaccinations although not significantly. Pre-vaccination regulatory T cell levels were significantly lower in histological responders compared to non-responders (p = 0.03). Feasibility of testing plasma for multiplex cytokine/chemokine analysis and of performing proteomic analysis of PBMCs was examined for potentially identifying biomarkers in the future. While these analyses are feasible to perform, attention needs to be given to how soon the blood samples would be processed after phlebotomy. As sufficient safety of PepCan has been demonstrated, enrollment for the Phase II clinical trial has been opened.


Subject(s)
Human papillomavirus 16/immunology , Papillomavirus Infections/immunology , Uterine Cervical Neoplasms/immunology , Viral Load/immunology , Adult , Chromatography, Liquid , Cytokines/blood , Cytokines/immunology , Dose-Response Relationship, Drug , Female , Host-Pathogen Interactions/drug effects , Host-Pathogen Interactions/immunology , Human papillomavirus 16/drug effects , Human papillomavirus 16/physiology , Humans , Leukocytes, Mononuclear/drug effects , Leukocytes, Mononuclear/immunology , Leukocytes, Mononuclear/metabolism , Middle Aged , Papillomavirus Infections/drug therapy , Papillomavirus Infections/virology , Proteome/immunology , Proteome/metabolism , Proteomics/methods , T-Lymphocytes, Regulatory/drug effects , T-Lymphocytes, Regulatory/immunology , Tandem Mass Spectrometry , Uterine Cervical Neoplasms/drug therapy , Uterine Cervical Neoplasms/virology , Vaccination/methods , Vaccines, Subunit/immunology , Vaccines, Subunit/therapeutic use , Viral Load/drug effects , Young Adult
6.
J Neurochem ; 137(2): 154-63, 2016 Apr.
Article in English | MEDLINE | ID: mdl-26778773

ABSTRACT

The control of NFκB in CNS neurons appears to differ from that in other cell types. Studies have reported induction of NFκB in neuronal cultures and immunostaining in vivo, but others have consistently detected little or no transcriptional activation by NFκB in brain neurons. To test if neurons lack some component of the signal transduction system for NFκB activation, we transfected cortical neurons with several members of this signaling system along with a luciferase-based NFκB-reporter plasmid; RelA was cotransfected in some conditions. No component of the NFκB pathway was permissive for endogenous NFκB activity, and none stimulated the activity of exogenous RelA. Surprisingly, however, the latter was inhibited by cotransfection of NFκB-inducing kinase (NIK). Fluorescence imaging of RelA indicated that co-expression of NIK sequestered RelA in the cytoplasm, similar to the effect of IκBα. NIK-knockout mice showed elevated expression of an NFκB-reporter construct in neurons in vivo. Cortical neurons cultured from NIK-knockout mice showed elevated expression of an NFκB-reporter transgene. Consistent with data from other cell types, a C-terminal fragment of NIK suppressed RelA activity in astrocytes as well as neurons. Therefore, the inhibitory ability of the NIK C-terminus was unbiased with regard to cell type. However, inhibition of NFκB by full-length NIK is a novel outcome that appears to be specific to CNS neurons. This has implications for unique aspects of transcription in the CNS, perhaps relevant to aspects of development, neuroplasticity, and neuroinflammation. Full-length NIK was found to inhibit (down arrow) transcriptional activation of NFκB in neurons, while it elevated (up arrow) activity in astrocytes. Deletion constructs corresponding to the N-terminus or C-terminus also inhibited NFκB in neurons, while only the C-terminus did so in astrocytes. One possible explanation is that the inhibition in neurons occurs via two different mechanisms, including the potential for a neuron-specific protein (e.g., one of the 14-3-3 class) to create a novel complex in neurons, whereas the C-terminus may interact directly with NFκB. [Structure of NIK is based on Liu J., Sudom A., Min X., Cao Z., Gao X., Ayres M., Lee F., Cao P., Johnstone S., Plotnikova O., Walker N., Chen G., and Wang Z. (2012) Structure of the nuclear factor κB-inducing kinase (NIK) kinase domain reveals a constitutively active conformation. J Biol Chem. 287, 27326-27334); N-terminal lobe is oriented at top].


Subject(s)
Central Nervous System/cytology , Gene Expression Regulation/genetics , NF-kappa B/metabolism , Neurons/metabolism , Protein Serine-Threonine Kinases/metabolism , Animals , Cells, Cultured , Embryo, Mammalian , Glycoside Hydrolases/genetics , Glycoside Hydrolases/metabolism , I-kappa B Kinase/genetics , I-kappa B Kinase/metabolism , Mice , Mice, Inbred C57BL , Mice, Transgenic , Protein Serine-Threonine Kinases/genetics , RNA, Small Interfering/genetics , RNA, Small Interfering/metabolism , Rats , Rats, Sprague-Dawley , Signal Transduction/physiology , TNF Receptor-Associated Factor 2/genetics , TNF Receptor-Associated Factor 2/metabolism , TNF Receptor-Associated Factor 6/genetics , TNF Receptor-Associated Factor 6/metabolism , Transcription Factor RelA/genetics , Transcription Factor RelA/metabolism , NF-kappaB-Inducing Kinase
7.
Oncoimmunology ; 4(10): e1031439, 2015 Oct.
Article in English | MEDLINE | ID: mdl-26451301

ABSTRACT

PURPOSE: Non-surgical treatments for cervical intraepithelial neoplasia 2/3 (CIN2/3) are needed as surgical treatments have been shown to double preterm delivery rate. The goal of this study was to demonstrate safety of a human papillomavirus (HPV) therapeutic vaccine called PepCan, which consists of four current good-manufacturing production-grade peptides covering the HPV type 16 E6 protein and Candida skin test reagent as a novel adjuvant. PATIENTS AND METHODS: The study was a single-arm, single-institution, dose-escalation phase I clinical trial, and the patients (n = 24) were women with biopsy-proven CIN2/3. Four injections were administered intradermally every 3 weeks in limbs. Loop electrical excision procedure (LEEP) was performed 12 weeks after the last injection for treatment and histological analysis. Six subjects each were enrolled (50, 100, 250, and 500 µg per peptide). RESULTS: The most common adverse events (AEs) were injection site reactions, and none of the patients experienced dose-limiting toxicities. The best histological response was seen at the 50 µg dose level with a regression rate of 83% (n = 6), and the overall rate was 52% (n = 23). Vaccine-induced immune responses to E6 were detected in 65% of recipients (significantly in 43%). Systemic T-helper type 1 (Th1) cells were significantly increased after four vaccinations (P = 0.02). CONCLUSION: This study demonstrated that PepCan is safe. A significantly increased systemic level of Th1 cells suggests that Candida, which induces interleukin-12 (IL-12) in vitro, may have a Th1 promoting effect. A phase II clinical trial to assess the full effect of this vaccine is warranted.

8.
Life Sci ; 90(25-26): 975-9, 2012 Jun 27.
Article in English | MEDLINE | ID: mdl-22634325

ABSTRACT

AIMS: Previous genetic studies have shown that a C/T polymorphism at position -889 of the IL1A promoter, specifically allele 2 (-889T), increases the risk for development of several inflammation-related disorders, such as periodontitis, osteomyelitis, toxoplasmic retinochoroiditis, contact dermatitis, as well as neurodegenerative conditions such as Alzheimer's disease. We sought to determine the differential abilities of C- and T- containing versions of the -889 sequence to bind nuclear proteins from microglia. MAIN METHODS: Microglial cells were subjected to inflammatory activation prior to the harvest of nuclear proteins. Electrophoretic mobility shift assays (EMSA) were performed using oligonucleotide probes representing 25 base pairs surrounding the IL1A -889 polymorphism. Antibodies reactive against transcription factors were used to identify the specific proteins involved in complexes with DNA. KEY FINDINGS: EMSA revealed multiple differences in DNA-binding profiles when microglial nuclear extracts were incubated with the polymorphic probes. The allele-2 probe formed specific complexes that were not detected with the allele-1 (-889C) probe, and vice versa. Formation of allele-2 nucleoprotein complexes was increased in activated microglia. Antibody supershift analysis indicated that multiple Jun-family members but not Fos-family proteins contributed to the LPS-activated allele-2 EMSA complexes. LPS-activation of allele-2 EMSA complexes could be blocked by the specific c-Jun N-terminal kinase (JNK) inhibitor SP600125. SIGNIFICANCE: These results suggest that the -889 polymorphism creates differential interactions with transcription factors that could lead to differential expression rates under proinflammatory conditions.


Subject(s)
Interleukin-1alpha/genetics , Polymorphism, Genetic/genetics , Promoter Regions, Genetic/genetics , Transcription Factor AP-1/genetics , Animals , Animals, Newborn , Cells, Cultured , Electrophoretic Mobility Shift Assay/methods , Interleukin-1alpha/metabolism , Microglia/metabolism , Protein Binding/genetics , Rats , Rats, Sprague-Dawley , Transcription Factor AP-1/metabolism
9.
J Neuroinflammation ; 6: 16, 2009 May 18.
Article in English | MEDLINE | ID: mdl-19450264

ABSTRACT

The unique physiology and function of neurons create differences in their cellular physiology, including their regulation of gene expression. We began several years ago exploring the relationships between the NFkappaB transcription factor, neuronal survival, and glutamate receptor activation in telencephalic neurons. These studies led us to conclude that this population of cells is nearly incapable of activating the NFkappaB that is nonetheless expressed at reasonable levels. A subset of the kappaB cis elements are instead bound by members of the Sp1 family in neurons. Also surprising was our discovery that Sp1 itself, typically described as ubiquitous, is severely restricted in expression within forebrain neurons; Sp4 seems to be substituted during neuronal differentiation. These findings and their implications for neuronal differentiation--as well as potential dedifferentiation during degenerative processes--are discussed here.


Subject(s)
Gene Expression Regulation , NF-kappa B/metabolism , Neurons/physiology , Sp1 Transcription Factor/metabolism , Transcription, Genetic , Animals , Apolipoproteins E/genetics , Apolipoproteins E/metabolism , Glutamic Acid/metabolism , Humans , I-kappa B Proteins/genetics , I-kappa B Proteins/metabolism , NF-kappa B/genetics , Neurogenesis/genetics , Neurogenic Inflammation/genetics , Neurogenic Inflammation/physiopathology , Neurons/cytology , Prosencephalon/cytology , Prosencephalon/physiology , Signal Transduction , Sp1 Transcription Factor/genetics , Sp4 Transcription Factor/genetics , Sp4 Transcription Factor/metabolism , Superoxide Dismutase/genetics , Superoxide Dismutase/metabolism , Transcription Factor RelA/genetics , Transcription Factor RelA/metabolism
10.
J Biol Chem ; 281(47): 35863-72, 2006 Nov 24.
Article in English | MEDLINE | ID: mdl-17023425

ABSTRACT

In addition to their conventional G-C/T target sequences, Sp1 family transcription factors (Sp-factors) can interact with a subset of the target sequences for NFkappaB. Due to the low level of bona fide NFkappaB activity in most resting cells, this interaction between Sp-factors and kappaB-sites could play important roles in cell function. Here we used mutagenesis of a canonical kappaB element from the immunoglobulin and HIV promoters to identify the GC-rich sequences at each end required for Sp-factor targeting. Through screening of multiple kappaB elements, a sequence element located in the second intron of superoxide dismutase-2 (SOD2) was identified as a good candidate for both NFkappaB and Sp-factor binding. In neurons, the prominent proteins interacting with this site were Sp3 and Sp4, whereas Sp1, Sp3, and NFkappaB were associated with this site in astroglia. The neuronal Sp-factors repressed transcriptional activity through this kappaB-site. In contrast, astroglial Sp-factors activated promoter activity through the same element. NFkappaB contributed to control of the SOD2 kappaB element only in astrocytes. These findings imply that cell-type specificity of transcription in the central nervous system, particularly with regard to kappaB elements, may include two unique aspects of neurons: 1) a recalcitrant NFkappaB and 2) the substitution of Sp4 for Sp1.


Subject(s)
Astrocytes/metabolism , Gene Expression Regulation, Developmental , NF-kappa B/genetics , Neurons/metabolism , Superoxide Dismutase/genetics , Transcription, Genetic , Animals , Brain/embryology , Cell Nucleus/metabolism , Central Nervous System/metabolism , HIV/genetics , Immunoglobulins/genetics , NF-kappa B/chemistry , Protein Binding , Rats , Sp Transcription Factors/metabolism , Superoxide Dismutase/chemistry
SELECTION OF CITATIONS
SEARCH DETAIL
...