Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 6 de 6
Filter
Add more filters










Database
Language
Publication year range
1.
Psychiatry Res ; 315: 114700, 2022 09.
Article in English | MEDLINE | ID: mdl-35803168

ABSTRACT

This study compares the psychological profile of Internally Displaced Persons (IDPs) and individuals living in host communities in the war-affected setting. We conducted a cross-sectional survey from October-November 2019. Subjects were recruited from six IDPs camps and the surrounding host communities within the metropolis of Maiduguri, Nigeria by convenience sampling. Data were collected using the Hausa version of Depression Anxiety Stress Scale-21, and analysed by logistic regression using adjusted odds ratio (AOR) at 95% Confidence Interval (CI). A total of 562 subjects were recruited. Living in IDP camp was the most significantly predictor of depression, anxiety, and stress. The common predictors were living in an IDP camp, and marital status (separated). Aged 18-29years was a protective factor compared to those ≥50years. Living in IDP camps, separated from partners, lack of education and pre-conflict employment were significant predictors of depression, anxiety and stress.


Subject(s)
Anxiety , Depression , Refugees , Humans , Anxiety/epidemiology , Armed Conflicts , Cross-Sectional Studies , Depression/epidemiology , Nigeria , Refugees/psychology
2.
Proc Natl Acad Sci U S A ; 97(11): 5924-9, 2000 May 23.
Article in English | MEDLINE | ID: mdl-10811875

ABSTRACT

The c-ski protooncogene encodes a transcription factor that binds DNA only in association with other proteins. To identify co-binding proteins, we performed a yeast two-hybrid screen. The results of the screen and subsequent co-immunoprecipitation studies identified Smad2 and Smad3, two transcriptional activators that mediate the type beta transforming growth factor (TGF-beta) response, as Ski-interacting proteins. In Ski-transformed cells, all of the Ski protein was found in Smad3-containing complexes that accumulated in the nucleus in the absence of added TGF-beta. DNA binding assays showed that Ski, Smad2, Smad3, and Smad4 form a complex with the Smad/Ski binding element GTCTAGAC (SBE). Ski repressed TGF-beta-induced expression of 3TP-Lux, the natural plasminogen activator inhibitor 1 promoter and of reporter genes driven by the SBE and the related CAGA element. In addition, Ski repressed a TGF-beta-inducible promoter containing AP-1 (TRE) elements activated by a combination of Smads, Fos, and/or Jun proteins. Ski also repressed synergistic activation of promoters by combinations of Smad proteins but failed to repress in the absence of Smad4. Thus, Ski acts in opposition to TGF-beta-induced transcriptional activation by functioning as a Smad-dependent co-repressor. The biological relevance of this transcriptional repression was established by showing that overexpression of Ski abolished TGF-beta-mediated growth inhibition in a prostate-derived epithelial cell line.


Subject(s)
DNA-Binding Proteins/physiology , Gene Expression Regulation/drug effects , Neoplasm Proteins/physiology , Proto-Oncogene Proteins/physiology , Repressor Proteins/physiology , Trans-Activators/physiology , Transforming Growth Factor beta/pharmacology , Cell Nucleus/chemistry , DNA-Binding Proteins/antagonists & inhibitors , Female , Gene Expression Regulation, Neoplastic/drug effects , Genes, Reporter , Humans , Luciferases/biosynthesis , Luciferases/genetics , Macromolecular Substances , Melanoma/pathology , Promoter Regions, Genetic , Recombinant Fusion Proteins/biosynthesis , Smad2 Protein , Smad3 Protein , Trans-Activators/antagonists & inhibitors , Transcription Factors/physiology , Transcription, Genetic/drug effects , Transforming Growth Factor beta/antagonists & inhibitors , Two-Hybrid System Techniques
3.
Exp Cell Res ; 255(2): 135-43, 2000 Mar 15.
Article in English | MEDLINE | ID: mdl-10694430

ABSTRACT

The basic helix-loop-helix/leucine zipper (bHLH/ZIP) microphthalmia-associated transcription factor (MITF) regulates transcription of genes encoding enzymes essential for melanin biosynthesis in melanocytes and retinal pigmented epithelial cells. To determine how MITF activity is regulated, we used the yeast two-hybrid system to identify proteins expressed by human melanoma cells that interact with MITF. The majority of clones that showed positive interaction with a 158-amino-acid region of MITF containing the bHLH/ZIP domain (aa 168-325) encoded the ubiquitin conjugating enzyme hUBC9. The association of MITF with hUBC9 was further confirmed by an in vitro GST pull-down assay. Although hUBC9 is known to interact preferentially with SENTRIN/SUMO1, in vitro transcription/translation analysis demonstrated greater association of MITF with ubiquitin than with SENTRIN. Importantly, cotransfection of MITF and hUBC9 expression vectors resulted in MITF protein degradation. MITF protein was stabilized by the proteasome inhibitor MG132, indicating the role of the ubiquitin-proteasome system in MITF degradation. Serine 73, which is located in a region rich in proline, glutamic acid, serine, and threonine (PEST), regulates MITF protein stability, since a serine to alanine mutation prevented hUBC9-mediated MITF (S73A) degradation. Furthermore, we identified lysine 201 as a potential ubiquitination site. A lysine to arginine mutation abolished MITF (K201R) degradation by hUBC9 in vivo. Our experiments indicate that by targeting MITF for proteasome degradation, hUBC9 is a critical regulator of melanocyte differentiation.


Subject(s)
DNA-Binding Proteins/physiology , Gene Expression Regulation , Ligases/physiology , Melanocytes/physiology , Pigment Epithelium of Eye/physiology , Ubiquitin-Conjugating Enzymes , Helix-Loop-Helix Motifs/physiology , Humans , Microphthalmia-Associated Transcription Factor , Transcription Factors/physiology , Tumor Cells, Cultured , Ubiquitins/physiology
4.
Exp Cell Res ; 253(2): 561-72, 1999 Dec 15.
Article in English | MEDLINE | ID: mdl-10585280

ABSTRACT

There is strong evidence that the senescent phenotype, whether induced by telomere shortening, oxidative damage, or oncogenic stimuli, is an important tumor suppressive mechanism. The melanocyte is a cell of neural crest origin that produces the pigment melanin and can develop into malignant melanomas. To understand how malignant cells escape senescence, it is first crucial to define what genes control senescence in the normal cell. Prolonged exposure to high levels of cAMP results in accumulation of melanin and terminal differentiation of human melanocytes. Here we present evidence that activation of a cAMP pathway correlates with multiple cellular changes in these cells: (1) increased expression of the transcription factor microphthalmia; (2) increased melanogenesis; (3) increased association of the cyclin-dependent kinase inhibitors (CDK-Is) p27(KIP1) and p16(INK4) with CDK2 and CDK4, respectively; (4) failure to phosphorylate the retinoblastoma protein (pRB); (5) decreased expression of E2F1, E2F2, and E2F4 proteins; (6) loss of E2F DNA-binding activity; and (7) phenotypic changes characteristic of senescent cells. Senescent melanocytes have potent E2F inhibitory activity, because extracts from these cells completely abolished E2F DNA-binding activity that was present in extracts from the early proliferative phase. We propose that increased activity of the CDK-Is p27 and p16 and loss of E2F activity in human melanocytes characterize a senescence program activated by the cAMP pathway. Disruption of cAMP-mediated and melanogenesis-induced senescence may cause immortalization of human melanocytes, an early step in the development of melanomas.


Subject(s)
Carrier Proteins , Cell Cycle Proteins , Cellular Senescence/physiology , Cyclin-Dependent Kinase Inhibitor p16/metabolism , DNA-Binding Proteins , Melanocytes/cytology , Microtubule-Associated Proteins/metabolism , Proto-Oncogene Proteins , Transcription Factors/metabolism , Tumor Suppressor Proteins , Cells, Cultured , Cyclic AMP/physiology , Cyclin-Dependent Kinase 4 , Cyclin-Dependent Kinase Inhibitor p16/genetics , Cyclin-Dependent Kinase Inhibitor p27 , Cyclin-Dependent Kinases/metabolism , E2F Transcription Factors , E2F1 Transcription Factor , E2F2 Transcription Factor , E2F4 Transcription Factor , Gene Expression Regulation, Enzymologic , Gene Expression Regulation, Neoplastic , Humans , Melanins/biosynthesis , Melanocytes/enzymology , Melanoma/enzymology , Melanoma/genetics , Melanoma/pathology , Microphthalmos/genetics , Microtubule-Associated Proteins/genetics , Phosphorylation , Retinoblastoma Protein/metabolism , Retinoblastoma-Binding Protein 1 , S Phase/physiology , Signal Transduction/physiology , Skin Neoplasms/enzymology , Skin Neoplasms/genetics , Skin Neoplasms/pathology , Transcription Factor DP1 , Transcription Factors/genetics
5.
J Investig Dermatol Symp Proc ; 3(1): 36-40, 1998 Aug.
Article in English | MEDLINE | ID: mdl-9732055

ABSTRACT

With aging, melanocytes become unevenly distributed in the epidermis. In light skin individuals, hypopigmentation is found in association with focal hyperpigmentation (lentigo senilis). Apparently this results from progressive loss of active melanocytes and focal increase in melanocyte proliferation and/or aggregation. This paper summarizes the present knowledge on aging of melanocytes in vivo and in vitro, with a focus on the role of melanin as a determinant for proliferation and terminal differentiation. We describe that excessive melanin deposition by cyclic AMP-inducing agents results in increased expression of the cyclin-dependent kinase inhibitors p27Kp-1 and p21SDI-1/Waf-1, increased binding of p16 to CDK4, and terminal differentiation. Importantly, the efficiency with which the melanocytes exit the cell cycle depends on the melanin background of the donor's cells. Melanocytes from skin types IV-VI that accumulate large amounts of brown black melanin (eumelanin), lose expression of the transcription factors E2F1 and E2F2, two key regulatory proteins, and withdraw from the cell cycle more rapidly than melanocytes from skin types I and II that accumulate red/yellow melanin (pheomelanin). Thus, we propose that terminal differentiation is a tumor suppressor mechanism that becomes less efficient under imperfect eumelanization.


Subject(s)
Melanocytes/pathology , Skin Aging , Animals , Cell Cycle/genetics , Gene Expression Regulation , Humans , Melanins/genetics , Melanins/metabolism , Melanocytes/metabolism
6.
Cell Motil Cytoskeleton ; 26(1): 49-65, 1993.
Article in English | MEDLINE | ID: mdl-8221907

ABSTRACT

The fatty acid 12(S)-HETE may be a new second messenger capable of activating PKC. In tumor cells 12(S)-HETE stimulates cytoskeleton-dependent cellular responses such as adhesion and spreading. Analysis of 12(S)-HETE effects on B16a melanoma cell cytoskeleton revealed reversible rearrangement of microtubules, microfilaments, the actin-binding proteins, vinculin, myosin heavy (MHC) and light chains (MLC), as well as bundling of vimentin intermediate filaments. The alterations in microfilaments and intermediate filaments occurred very rapidly, i.e., 5 min after exposure of tumor cells to 12(S)-HETE. The 12(S)-HETE-induced cytoskeletal alterations were accompanied by centrifugal organelle-translocation. Interestingly, MLC exhibited clear association with the cytoplasmic organelles. Biochemical analysis of the 12(S)-HETE effect indicated a PKC-mediated reversible hyperphosphorylation of MLC, vimentin, and a 130 kD cytoskeletal-associated protein. Optimal effects were obtained after 5 min treatment with 12(S)-HETE at 0.1 microM concentration. 12(S)-HETE pretreatment induced tumor cell spreading on a fibronectin matrix which required the intactness of all three major cytoskeletal components. The spreading process was dependent upon the activity of PKC. Our data suggest that 12(S)-HETE is a physiological stimulant of PKC. Further, it induces rearrangement of the cytoskeleton of tumor cells in interphase resulting in the stimulation of cytoskeleton-dependent cell activity such as spreading.


Subject(s)
Calcium/pharmacology , Cytoskeleton/ultrastructure , Hydroxyeicosatetraenoic Acids/pharmacology , Melanoma, Experimental/pathology , Phospholipids/pharmacology , Protein Kinases/pharmacology , Skin Neoplasms/pathology , 12-Hydroxy-5,8,10,14-eicosatetraenoic Acid , Actin Cytoskeleton/chemistry , Actin Cytoskeleton/drug effects , Actin Cytoskeleton/ultrastructure , Actins/analysis , Animals , Cell Movement/physiology , Cytoskeletal Proteins/metabolism , Cytoskeletal Proteins/physiology , Cytoskeleton/chemistry , Cytoskeleton/drug effects , Dose-Response Relationship, Drug , Fibronectins/analysis , Fluorescent Antibody Technique , Immunohistochemistry , Intermediate Filaments/chemistry , Intermediate Filaments/drug effects , Intermediate Filaments/ultrastructure , Melanoma, Experimental/chemistry , Melanoma, Experimental/ultrastructure , Mice , Myosins/analysis , Organelles/physiology , Organelles/ultrastructure , Phosphorylation , Protein Kinases/physiology , Skin Neoplasms/chemistry , Skin Neoplasms/ultrastructure , Tumor Cells, Cultured , Vimentin/analysis , Vinculin/analysis
SELECTION OF CITATIONS
SEARCH DETAIL
...