Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 14 de 14
Filter
1.
Sci Rep ; 9(1): 8980, 2019 06 20.
Article in English | MEDLINE | ID: mdl-31222072

ABSTRACT

Hydrosalpinx, the blockage of fallopian tubes, can result from pelvic inflammatory disease. Hydrosalpinx is a cause of infertility and negatively impacts in vitro fertilization. To better understand the pathobiology of hydrosalpinx, we compared the proteome of lavages from disease vs. healthy fallopian tubes. Results indicate a disruption of redox homeostasis and activation of the complement system, immune cell infiltration, and phagocytosis; pathways that may drive tubal injury. To our surprise among the most prominent proteins with hydrosalpinx was mesothelin (MSLN), which until now has only been associated with epithelial malignancies. Analogous to mesothelioma and ovarian carcinoma, a significant increase of MSLN was detected in plasma from patients with hydrosalpinx. This finding suggests MSLN may provide clinical diagnosis in lieu of the current approaches that require invasive imaging. Importantly, these findings implicate MSLN in a benign disease, indicating that the activation and role of MSLN is not restricted to cancer.


Subject(s)
Fallopian Tube Diseases/metabolism , Fallopian Tubes/metabolism , Proteome , Chromatography, Liquid , Disease Susceptibility , Fallopian Tube Diseases/etiology , Fallopian Tube Diseases/pathology , Female , Fertility , GPI-Linked Proteins/blood , Humans , Immunohistochemistry , Mesothelin , Proteomics/methods , Tandem Mass Spectrometry , Therapeutic Irrigation
2.
Sci Rep ; 7(1): 17883, 2017 12 20.
Article in English | MEDLINE | ID: mdl-29263436

ABSTRACT

Preterm infants are at significantly increased risk for lifelong neurodevelopmental disability with male offspring disproportionately affected. Corticosteroids (such as betamethasone) and magnesium sulphate (MgSO4) are administered to women in preterm labor to reduce neurologic morbidity. Despite widespread use of MgSO4 in clinical practice, its effects on adult offspring are not well known nor have sex-specific differences in therapeutic response been explored. The objective of our study was to examine the long-term effects of perinatal neuroinflammation and the effectiveness of prenatal MgSO4/betamethasone treatments between males and females in a murine model via histologic and expression analyses. Our results demonstrate that male but not female offspring exposed to intrauterine inflammation demonstrated impaired performance in neurodevelopmental testing in early life assessed via negative geotaxis, while those exposed to injury plus treatment fared better. Histologic analysis of adult male brains identified a significant reduction in hippocampal neural density in the injured group compared to controls. Evaluation of key neural markers via qRT-PCR demonstrated more profound differences in gene expression in adult males exposed to injury and treatment compared to female offspring, which largely showed resistance to injury. Prenatal treatment with MgSO4/betamethasone confers long-term benefits beyond cerebral palsy prevention with sex-specific differences in response.


Subject(s)
Betamethasone/pharmacology , Inflammation/drug therapy , Magnesium Sulfate/pharmacology , Animals , Biomarkers/metabolism , Cerebral Palsy/drug therapy , Cerebral Palsy/metabolism , Disease Models, Animal , Female , Gene Expression/drug effects , Hippocampus/drug effects , Hippocampus/metabolism , Humans , Inflammation/metabolism , Male , Mice , Pregnancy , Prenatal Care , Sex Characteristics
3.
Reprod Sci ; 21(2): 204-14, 2014 Feb.
Article in English | MEDLINE | ID: mdl-23757314

ABSTRACT

Glycosylation of plasma proteins increases during pregnancy. Our objectives were to investigate an anti-inflammatory role of these proteins in normal pregnancies and determine whether aberrant protein glycosylation promotes monocyte adhesion in preeclampsia. Plasma was prospectively collected from nonpregnant controls and nulliparous patients in all 3 trimesters. Patients were divided into cohorts based on the applicable postpartum diagnosis. U937 monocytes were preconditioned with enzymatically deglycosylated plasma, and monocyte adhesion to endothelial cell monolayers was quantified by spectrophotometry. Plasma from nonpregnant controls, first trimester normotensives, and first trimester patients with mild preeclampsia inhibited monocyte-endothelial cell adhesion (P < .05), but plasma from first trimester patients with severe preeclampsia and second and third trimester normotensives did not. Deglycosylating plasma proteins significantly increased adhesion in all the cohorts. These results support a role of plasma glycoprotein interaction in monocyte-endothelial cell adhesion and could suggest a novel therapeutic target for severe preeclampsia.


Subject(s)
Blood Proteins/metabolism , Monocytes/metabolism , Pre-Eclampsia/blood , Pre-Eclampsia/diagnosis , Severity of Illness Index , Adult , Amino Acid Sequence , Blood Proteins/genetics , Cell Adhesion/physiology , Cohort Studies , Female , Glycosylation , Human Umbilical Vein Endothelial Cells , Humans , Molecular Sequence Data , Pre-Eclampsia/genetics , Pregnancy , U937 Cells , Young Adult
4.
Fertil Steril ; 99(1): 199-205, 2013 Jan.
Article in English | MEDLINE | ID: mdl-23009827

ABSTRACT

OBJECTIVE: To investigate the effect of cigarette smoke exposure on ciliation and ciliogenesis in human oviductal epithelium. DESIGN: Molecular analysis using human tubal segments. SETTING: Academic medical center. PATIENT(S): Twenty women undergoing elective tubal sterilization procedure. INTERVENTION(S): Expression of ciliated cell-specific markers was compared in tubal segments from smokers and nonsmokers using quantitative immunohistochemistry and Western blot analysis. The expression of transcription factors in the motile ciliogenesis program was compared using quantitative polymerase chain reaction and quantitative immunohistochemistry. MAIN OUTCOME MEASURE(S): Oviductal ciliation and expression of transcription factors involved in ciliogenesis. RESULT(S): No significant differences were detected in density of ciliation between groups. Neither number of years of smoking nor pack-year history correlated with density of ciliation. Expression of ciliogenic transcription factors FOXJ1, RFX2, and RFX3 was consistent between groups. CONCLUSION(S): Few studies have evaluated the relationship between smoking and ciliated epithelium in human oviducts. Cigarette smoking does not seem to result in quantitative differences in the density of ciliation nor expression of ciliogenesis factors. Our findings suggest that pathophysiologic mechanisms other than ciliation account for the increased risk of ectopic pregnancy in women who smoke.


Subject(s)
Cilia/pathology , Cilia/physiology , Fallopian Tubes/physiology , Smoking/adverse effects , Adult , Biopsy , DNA-Binding Proteins/physiology , Epithelium/pathology , Epithelium/physiology , Fallopian Tubes/pathology , Female , Forkhead Transcription Factors/physiology , Humans , Incidence , Pregnancy , Pregnancy, Ectopic/epidemiology , Prospective Studies , Regulatory Factor X Transcription Factors , Transcription Factors/physiology
6.
Curr Colorectal Cancer Rep ; 7(2): 121-127, 2011 Jun.
Article in English | MEDLINE | ID: mdl-21980310

ABSTRACT

The Notch receptor signaling pathway regulates expression of the basic helix-loop-helix transcription factor ATOH1 (Math1/Hath1) to determine cell fate in the intestine. In differentiating intestinal stem cells, high levels of Notch activity specify absorptive enterocyte/colonocyte differentiation, whereas high ATOH1 activity specifies secretory (goblet, enteroendocrine, and Paneth) cell differentiation. In colorectal cancer, ATOH1 is a tumor suppressor that is silenced in most tumors, while Notch is oncogenic and often highly active in human tumors. In other gastrointestinal malignancies with features of intestinal metaplasia, such as esophageal and gastric cancers, the Notch-ATOH1 pathway becomes activated. In cancers and preneoplastic tissues that retain the ability to activate ATOH1, therapeutic targeting of this pathway can be achieved by inhibiting Notch activity (with Notch-targeting antibodies or small-molecule inhibitors of γ-secretase). Thus, targeting the Notch-ATOH1 pathway represents a novel approach to differentiation therapy in gastrointestinal cancers.

7.
Gastroenterology ; 139(3): 918-28, 928.e1-6, 2010 Sep.
Article in English | MEDLINE | ID: mdl-20621629

ABSTRACT

BACKGROUND & AIMS: The atonal homolog 1 (Atoh1) transcription factor is required for intestinal secretory (goblet, Paneth, enteroendocrine) cell differentiation. Notch/gamma-secretase inhibitors (GSIs) block proliferation and induce secretory cell differentiation in the intestine. We used genetic analyses of mice to determine whether Atoh1 mediates the effects of GSIs in normal and cancerous intestinal epithelia. METHODS: We studied mice with intestine-specific disruption of Atoh1 (Atoh1(Deltaintestine)), the adenomatosis polyposis coli (APC)(min) mutation, both mutations (Atoh1(Deltaintestine); APC(min)), or littermate controls; mice were given GSI or vehicle. Colorectal cancer (CRC) cell lines were treated with GSI or vehicle and with small hairpin RNAs to reduce ATOH1. Differentiation and homeostasis were assessed by protein, RNA, and histologic analyses. RESULTS: GSIs failed to induce secretory cell differentiation or apoptosis or decrease proliferation of Atoh1-null progenitor cells, compared with wild-type cells. Exposure of APC(min) adenomas to GSIs decreased proliferation and increased secretory cell numbers in an Atoh1-dependent manner. In CRC cells treated with GSI, ATOH1 levels were correlated inversely with proliferation. ATOH1 was required for secretory cell gene expression in cell lines and in mice. CONCLUSIONS: ATOH1 is required for all effects of GSIs in intestinal crypts and adenomas; Notch has no unique function in intestinal progenitors and cancer cells other than to regulate ATOH1 expression. Reducing ATOH1 activity might mitigate intestinal toxicity from systemic GSI therapy for nonintestinal diseases. Among gastrointestinal malignancies, ATOH1 mediates the effects of GSIs, so ATOH1 expression levels might predict responses to these inhibitors. We propose that only the subset of CRCs that retain ATOH1 expression will respond to GSIs.


Subject(s)
Amyloid Precursor Protein Secretases/antagonists & inhibitors , Antineoplastic Agents/pharmacology , Basic Helix-Loop-Helix Transcription Factors/metabolism , Cell Differentiation/drug effects , Cell Proliferation/drug effects , Colorectal Neoplasms/metabolism , Epithelial Cells/drug effects , Intestinal Mucosa/drug effects , Receptors, Notch/antagonists & inhibitors , Amyloid Precursor Protein Secretases/metabolism , Animals , Apoptosis/drug effects , Basic Helix-Loop-Helix Transcription Factors/genetics , Colorectal Neoplasms/genetics , Colorectal Neoplasms/pathology , Epithelial Cells/metabolism , Epithelial Cells/pathology , Gene Expression Regulation, Neoplastic , Genes, APC , HCT116 Cells , HT29 Cells , Humans , Intestinal Mucosa/metabolism , Intestinal Mucosa/pathology , Mice , Mice, Transgenic , RNA Interference , RNA, Messenger/metabolism , Receptors, Notch/metabolism , Time Factors
8.
Exp Cell Res ; 316(3): 452-65, 2010 Feb 01.
Article in English | MEDLINE | ID: mdl-19786015

ABSTRACT

BACKGROUND AND AIMS: SPDEF (also termed PDEF or PSE) is an ETS family transcription factor that regulates gene expression in the prostate and goblet cell hyperplasia in the lung. Spdef has been reported to be expressed in the intestine. In this paper, we identify an important role for Spdef in regulating intestinal epithelial cell homeostasis and differentiation. METHODS: SPDEF expression was inhibited in colon cancer cells to determine its ability to control goblet cell gene activation. The effects of transgenic expression of Spdef on intestinal differentiation and homeostasis were determined. RESULTS: In LS174T colon cancer cells treated with Notch/gamma-secretase inhibitor to activate goblet cell gene expression, shRNAs that inhibited SPDEF also repressed expression of goblet cell genes AGR2, MUC2, RETLNB, and SPINK4. Transgenic expression of Spdef caused the expansion of intestinal goblet cells and corresponding reduction in Paneth, enteroendocrine, and absorptive enterocytes. Spdef inhibited proliferation of intestinal crypt cells without induction of apoptosis. Prolonged expression of the Spdef transgene caused a progressive reduction in the number of crypts that expressed Spdef, consistent with its inhibitory effects on cell proliferation. CONCLUSIONS: Spdef was sufficient to inhibit proliferation of intestinal progenitors and induce differentiation into goblet cells; SPDEF was required for activation of goblet cell associated genes in vitro. These data support a model in which Spdef promotes terminal differentiation into goblet cells of a common goblet/Paneth progenitor.


Subject(s)
Cell Differentiation , Goblet Cells/cytology , Goblet Cells/metabolism , Proto-Oncogene Proteins c-ets/metabolism , Animals , Basic Helix-Loop-Helix Transcription Factors/metabolism , Cell Count , Cell Differentiation/drug effects , Cell Line, Tumor , Cell Proliferation/drug effects , Colonic Neoplasms/genetics , Colonic Neoplasms/pathology , DNA-Binding Proteins/metabolism , Doxycycline/pharmacology , Epithelial Cells/cytology , Epithelial Cells/drug effects , Epithelial Cells/metabolism , Gene Expression Regulation, Neoplastic/drug effects , Goblet Cells/drug effects , Humans , Intestinal Mucosa/drug effects , Intestinal Mucosa/metabolism , Intestinal Mucosa/pathology , Intestinal Mucosa/ultrastructure , Mice , Organ Specificity/drug effects , Protein Transport/drug effects , Proto-Oncogene Proteins c-ets/genetics , Stem Cells/cytology , Stem Cells/drug effects , Stem Cells/metabolism , Transcription Factors/metabolism , Transgenes/genetics
9.
PLoS Biol ; 7(2): e39, 2009 Feb 24.
Article in English | MEDLINE | ID: mdl-19243219

ABSTRACT

Colon cancer accounts for more than 10% of all cancer deaths annually. Our genetic evidence from Drosophila and previous in vitro studies of mammalian Atonal homolog 1 (Atoh1, also called Math1 or Hath1) suggest an anti-oncogenic function for the Atonal group of proneural basic helix-loop-helix transcription factors. We asked whether mouse Atoh1 and human ATOH1 act as tumor suppressor genes in vivo. Genetic knockouts in mouse and molecular analyses in the mouse and in human cancer cell lines support a tumor suppressor function for ATOH1. ATOH1 antagonizes tumor formation and growth by regulating proliferation and apoptosis, likely via activation of the Jun N-terminal kinase signaling pathway. Furthermore, colorectal cancer and Merkel cell carcinoma patients show genetic and epigenetic ATOH1 loss-of-function mutations. Our data indicate that ATOH1 may be an early target for oncogenic mutations in tissues where it instructs cellular differentiation.


Subject(s)
Basic Helix-Loop-Helix Transcription Factors/genetics , Carcinoma, Merkel Cell/genetics , Colorectal Neoplasms/genetics , Genes, Tumor Suppressor/physiology , Skin Neoplasms/genetics , Animals , Apoptosis/genetics , Basic Helix-Loop-Helix Transcription Factors/metabolism , Carcinoma, Merkel Cell/metabolism , Carcinoma, Merkel Cell/pathology , Cell Line, Tumor , Cell Proliferation , Colorectal Neoplasms/metabolism , Colorectal Neoplasms/pathology , DNA Mutational Analysis , Gene Expression Regulation, Neoplastic , Humans , Intestinal Mucosa/metabolism , Intestinal Mucosa/pathology , JNK Mitogen-Activated Protein Kinases , Male , Mice , Mice, Knockout , Mutation , Signal Transduction , Skin Neoplasms/metabolism , Skin Neoplasms/pathology
10.
J Biol Chem ; 283(46): 32056-65, 2008 Nov 14.
Article in English | MEDLINE | ID: mdl-18805794

ABSTRACT

Growth factor independent-1 (Gfi1) is a zinc finger protein with a SNAG-transcriptional repressor domain. Ajuba is a LIM domain protein that shuttles between the cytoplasm and the nucleus. Ajuba functions as a co-repressor for synthetic Gfi1 SNAG-repressor domain-containing constructs, but a role for Ajuba co-repression of the cognate DNA bound Gfi1 protein has not been defined. Co-immunoprecipitation of synthetic and endogenous proteins and co-elution with gel filtration suggest that an endogenous Ajuba.Gfi1.HDAC multiprotein complex is possible. Active histone deacetylase activity co-immunoprecipitates with Ajuba or Gfi1, and both proteins depend upon histone deacetylases for full transcriptional repression activity. Ajuba LIM domains directly bind to Gfi1, but the association is not SNAG domain-dependent. ChIP analysis and reciprocal knockdown experiments suggest that Ajuba selectively functions as a co-repressor for Gfi1 autoregulation. The data suggest that Ajuba is utilized as a corepressor selectively on Gfi1 target genes.


Subject(s)
DNA-Binding Proteins/metabolism , Down-Regulation , Histone Deacetylases/metabolism , Homeodomain Proteins/metabolism , Transcription Factors/metabolism , Cell Line , Cell Survival , DNA-Binding Proteins/genetics , Homeodomain Proteins/genetics , Humans , LIM Domain Proteins , Protein Binding , Transcription Factors/genetics
11.
Lab Invest ; 87(4): 336-44, 2007 Apr.
Article in English | MEDLINE | ID: mdl-17377622

ABSTRACT

Naphthalene exposure kills lung airway epithelial (Clara) cells, but is rapidly followed by Clara cell reconstitution coincident with proliferation of pulmonary neuroendocrine cells (PNEC). Although a role for mature PNEC in the reconstitution process has been excluded, the reconstituting progenitor cells have been suggested to enter a transient neuroendocrine (NE) differentiation phase before differentiating to Clara cells. Furthermore, these progenitors were suggested to be the target population for transformation to a NE tumor; small cell lung cancer (SCLC). Although the NE phenotype is central to SCLC oncogenesis, the relevance of NE differentiation to post naphthalene reconstitution remains to be determined. The Growth factor independent-1 (Gfi1) transcription factor is expressed in SCLC and is required for the NE differentiation of PNEC. Gfi1(-/-) mice display a 70% reduction in airway cells that express NE markers, and cells that stain for NE markers show weak expression of some markers. Therefore, to determine the relevance of the NE phenotype to post-naphthalene reconstitution, we examined post-naphthalene reconstitution in Gfi1(-/-) mice. Our analyses indicate that the post-naphthalene regeneration process includes both airway epithelial proliferation and apoptosis. Gfi1 deletion lowered both airway epithelial proliferation and apoptosis; however, the post-naphthalene rate of increase in growth and apoptosis was not significantly different between Gfi1(-/-) mice and wild-type littermates. Moreover, the timing and extent of CC10+ cell regeneration was unaffected by Gfi1 deletion. These data suggest that neither Gfi1 nor the NE phenotype play a dominant role in the regeneration process. However, the few Gfi1(-/-) cells capable of NE differentiation show a significant reduction in post-naphthalene proliferation. The modest proliferation seen in Gfi1(-/-) NE cells is consistent with the previously proposed role for Gfi1 in controlling neuroendocrine cancer growth.


Subject(s)
Carcinoma, Small Cell/pathology , Cell Proliferation/drug effects , DNA-Binding Proteins/physiology , Environmental Pollutants/toxicity , Lung Neoplasms/pathology , Lung/pathology , Naphthalenes/toxicity , Neurosecretory Systems/pathology , Transcription Factors/physiology , Animals , Apoptosis , Carcinoma, Small Cell/chemically induced , Cell Differentiation , DNA-Binding Proteins/genetics , Lung Neoplasms/chemically induced , Mice , Mice, Knockout , Phenotype , Regeneration , Respiratory Mucosa/pathology , Stem Cells/cytology , Transcription Factors/genetics
12.
Crit Rev Oncol Hematol ; 59(2): 85-97, 2006 Aug.
Article in English | MEDLINE | ID: mdl-16716599

ABSTRACT

The growth factor independence-1 (Gfi1) transcription factor is required for proper development of neuroendocrine cells, sensory neurons, and blood. Patients with mutations in Gfi1 exhibit severe congenital neutropenia (SCN) or non-immune chronic idiopathic neutropenia of adults. Gfi1 was initially described as an oncoprotein that mediates tumor progression in a mouse model of leukemia; however, recent data suggest that Gfi1 may act as either an oncogene or an anti-proliferative tumor suppressor gene depending on the cell type. Here we review the latest literature on Gfi1, and emphasize its role in the hematopoietic, sensory and neuroendocrine systems.


Subject(s)
DNA-Binding Proteins/metabolism , Oncogene Proteins/metabolism , Transcription Factors/metabolism , Tumor Suppressor Proteins/metabolism , Animals , DNA-Binding Proteins/genetics , Hematopoiesis/genetics , Humans , Mice , Neurons, Afferent/metabolism , Neurosecretory Systems/metabolism , Neutropenia/genetics , Neutropenia/metabolism , Oncogene Proteins/genetics , Organ Specificity , Transcription Factors/genetics , Tumor Suppressor Proteins/genetics
13.
Cancer Res ; 64(19): 6874-82, 2004 Oct 01.
Article in English | MEDLINE | ID: mdl-15466176

ABSTRACT

Human small cell lung cancers might be derived from pulmonary cells with a neuroendocrine phenotype. They are driven to proliferate by autocrine and paracrine neuropeptide growth factor stimulation. The molecular basis of the neuroendocrine phenotype of lung carcinomas is relatively unknown. The Achaete-Scute Homologue-1 (ASH1) transcription factor is critically required for the formation of pulmonary neuroendocrine cells and is a marker for human small cell lung cancers. The Drosophila orthologues of ASH1 (Achaete and Scute) and the growth factor independence-1 (GFI1) oncoprotein (Senseless) genetically interact to inhibit Notch signaling and specify fly sensory organ development. Here, we show that GFI1, as with ASH1, is expressed in neuroendocrine lung cancer cell lines and that GFI1 in lung cancer cell lines functions as a DNA-binding transcriptional repressor protein. Forced expression of GFI1 potentiates tumor formation of small-cell lung carcinoma cells. In primary human lung cancer specimens, GFI1 expression strongly correlates with expression of ASH1, the neuroendocrine growth factor gastrin-releasing peptide, and neuroendocrine markers synaptophysin and chromogranin A (P < 0.0000001). GFI1 colocalizes with chromogranin A and calcitonin-gene-related peptide in embryonic and adult murine pulmonary neuroendocrine cells. In addition, mice with a mutation in GFI1 display abnormal development of pulmonary neuroendocrine cells, indicating that GFI1 is important for neuroendocrine differentiation.


Subject(s)
Carcinoma, Neuroendocrine/metabolism , Carcinoma, Small Cell/metabolism , DNA-Binding Proteins/biosynthesis , Lung Neoplasms/metabolism , Lung/cytology , Neurosecretory Systems/cytology , Transcription Factors/biosynthesis , Animals , Carcinoma, Neuroendocrine/genetics , Carcinoma, Small Cell/genetics , Cell Differentiation , Cell Extracts/pharmacology , Cell Line, Tumor , DNA-Binding Proteins/genetics , Female , Humans , Lung/drug effects , Lung Neoplasms/genetics , Mice , Neoplasm Transplantation , Neurosecretory Systems/drug effects , Pregnancy , Transcription Factors/genetics , Transfection , Transplantation, Heterologous
14.
J Cell Biochem ; 89(5): 1005-18, 2003 Aug 01.
Article in English | MEDLINE | ID: mdl-12874834

ABSTRACT

Gfi-1 and Gfi-1B can repress transcription and play important roles in hematopoietic cell survival and differentiation. Although these proteins are known to bind DNA through a C-terminal zinc-finger domain and may require an N-terminal SNAG domain (SNAIL/Gfi-1) to repress transcription, the mechanism by which Gfi-1 and Gfi-1B act is unknown. A first step towards understanding the mechanism by which these proteins repress transcription is to identify interacting proteins that could contribute to transcriptional repression. ETO (also termed MTG8), was first identified through its involvement in the (8;21) translocation associated with acute myelogenous leukemia. It attaches to the nuclear matrix and associates with histone deacetylases and the co-repressors N-CoR, SMRT, and mSin3A, and may act as a co-repressor for site-specific transcriptions factors. In this report we demonstrate that Gfi-1 interacts with ETO and related proteins both in vitro and in vivo and with histone deacetylase proteins in vivo. We observed that a portion of Gfi-1 and Gfi-1B associated with the nuclear matrix, as is the case with ETO. Moreover, Gfi-1 and ETO co-localize to punctate subnuclear structures. When co-expressed in mammalian cells, Gfi-1 associates with histone deacetylse-1 (HDAC-1), HDAC-2, and HDAC-3. These data identify ETO as a partner for Gfi-1 and Gfi-1B, and suggest that Gfi-1 proteins repress transcription through recruitment of histone deacetylase-containing complexes.


Subject(s)
DNA-Binding Proteins/metabolism , Histone Deacetylases/metabolism , Hydroxamic Acids/metabolism , Nuclear Matrix/metabolism , Repressor Proteins/metabolism , Transcription Factors/metabolism , Cells, Cultured , DNA-Binding Proteins/chemistry , Enzyme Inhibitors/metabolism , Enzyme Inhibitors/pharmacology , Genes, Reporter/genetics , Histone Deacetylase Inhibitors , Humans , Hydroxamic Acids/pharmacology , Intermediate Filaments/metabolism , Luminescent Proteins/metabolism , Microscopy, Fluorescence , Proto-Oncogene Proteins/metabolism , RUNX1 Translocation Partner 1 Protein , Repressor Proteins/chemistry , Transcription, Genetic/physiology , Transfection
SELECTION OF CITATIONS
SEARCH DETAIL
...