Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 36
Filter
Add more filters

Publication year range
1.
Cell ; 155(5): 1022-33, 2013 Nov 21.
Article in English | MEDLINE | ID: mdl-24267888

ABSTRACT

Sequence polymorphisms linked to human diseases and phenotypes in genome-wide association studies often affect noncoding regions. A SNP within an intron of the gene encoding Interferon Regulatory Factor 4 (IRF4), a transcription factor with no known role in melanocyte biology, is strongly associated with sensitivity of skin to sun exposure, freckles, blue eyes, and brown hair color. Here, we demonstrate that this SNP lies within an enhancer of IRF4 transcription in melanocytes. The allele associated with this pigmentation phenotype impairs binding of the TFAP2A transcription factor that, together with the melanocyte master regulator MITF, regulates activity of the enhancer. Assays in zebrafish and mice reveal that IRF4 cooperates with MITF to activate expression of Tyrosinase (TYR), an essential enzyme in melanin synthesis. Our findings provide a clear example of a noncoding polymorphism that affects a phenotype by modulating a developmental gene regulatory network.


Subject(s)
Interferon Regulatory Factors/metabolism , Polymorphism, Single Nucleotide , Animals , Base Sequence , Enhancer Elements, Genetic , Humans , Interferon Regulatory Factors/chemistry , Interferon Regulatory Factors/genetics , Melanocytes/metabolism , Mice , Molecular Sequence Data , Pigmentation , Signal Transduction , Transcription Factor AP-2/chemistry , Transcription Factor AP-2/metabolism , Zebrafish
2.
FASEB J ; 33(5): 6412-6430, 2019 05.
Article in English | MEDLINE | ID: mdl-30779881

ABSTRACT

Growth hormone (GH) has an important function as an insulin antagonist with elevated insulin sensitivity evident in humans and mice lacking a functional GH receptor (GHR). We sought the molecular basis for this sensitivity by utilizing a panel of mice possessing specific deletions of GHR signaling pathways. Metabolic clamps and glucose homeostasis tests were undertaken in these obese adult C57BL/6 male mice, which indicated impaired hepatic gluconeogenesis. Insulin sensitivity and glucose disappearance rate were enhanced in muscle and adipose of mice lacking the ability to activate the signal transducer and activator of transcription (STAT)5 via the GHR (Ghr-391-/-) as for GHR-null (GHR-/-) mice. These changes were associated with a striking inhibition of hepatic glucose output associated with altered glycogen metabolism and elevated hepatic glycogen content during unfed state. The enhanced hepatic insulin sensitivity was associated with increased insulin receptor ß and insulin receptor substrate 1 activation along with activated downstream protein kinase B signaling cascades. Although phosphoenolpyruvate carboxykinase (Pck)-1 expression was unchanged, its inhibitory acetylation was elevated because of decreased sirtuin-2 expression, thereby promoting loss of PCK1. Loss of STAT5 signaling to defined chromatin immunoprecipitation targets would further increase lipogenesis, supporting hepatosteatosis while lowering glucose output. Finally, up-regulation of IL-15 expression in muscle, with increased secretion of adiponectin and fibroblast growth factor 1 from adipose tissue, is expected to promote insulin sensitivity.-Chhabra, Y., Nelson, C. N., Plescher, M., Barclay, J. L., Smith, A. G., Andrikopoulos, S., Mangiafico, S., Waxman, D. J., Brooks, A. J., Waters, M. J. Loss of growth hormone-mediated signal transducer and activator of transcription 5 (STAT5) signaling in mice results in insulin sensitivity with obesity.


Subject(s)
Carrier Proteins , Fatty Liver , Insulin Resistance/genetics , Liver , Obesity , STAT5 Transcription Factor/deficiency , Signal Transduction/genetics , Animals , Carrier Proteins/genetics , Carrier Proteins/metabolism , Fatty Liver/genetics , Fatty Liver/metabolism , Fatty Liver/pathology , Glucose/genetics , Glucose/metabolism , Glycogen/genetics , Glycogen/metabolism , Insulin Receptor Substrate Proteins/genetics , Insulin Receptor Substrate Proteins/metabolism , Intracellular Signaling Peptides and Proteins/genetics , Intracellular Signaling Peptides and Proteins/metabolism , Liver/metabolism , Liver/pathology , Male , Mice , Mice, Knockout , Obesity/genetics , Obesity/metabolism , Obesity/pathology , Phosphoenolpyruvate Carboxykinase (GTP)/genetics , Phosphoenolpyruvate Carboxykinase (GTP)/metabolism , Receptor, Insulin/genetics , Receptor, Insulin/metabolism , STAT5 Transcription Factor/metabolism
3.
Int J Cancer ; 140(12): 2634-2641, 2017 06 15.
Article in English | MEDLINE | ID: mdl-28076901

ABSTRACT

Tumour heterogeneity poses a distinct obstacle to therapeutic intervention. While the initiation of tumours across various physiological systems is frequently associated with signature mutations in genes that drive proliferation and bypass senescence, increasing evidence suggests that tumour progression and clonal diversity is driven at an epigenetic level. The tumour microenvironment plays a key role in driving diversity as cells adapt to demands imposed during tumour growth, and is thought to drive certain subpopulations back to a stem cell-like state. This stem cell-like phenotype primes tumour cells to react to external cues via the use of developmental pathways that facilitate changes in proliferation, migration and invasion. Because the dynamism of this stem cell-like state requires constant chromatin remodelling and rapid alterations at regulatory elements, it is of great therapeutic interest to identify the cell-intrinsic factors that confer these epigenetic changes that drive tumour progression. The nuclear factor one (NFI) family are transcription factors that play an important role in the development of many mammalian organ systems. While all four family members have been shown to act as either oncogenes or tumour suppressors across various cancer models, evidence has emerged implicating them as key epigenetic regulators during development and within tumours. Notably, NFIs have also been shown to regulate chromatin accessibility at distal regulatory elements that drive tumour cell dissemination and metastasis. Here we summarize the role of the NFIs in cancer, focusing largely on the potential mechanisms associated with chromatin remodelling and epigenetic modulation of gene expression.


Subject(s)
Epigenesis, Genetic , Gene Expression Regulation, Neoplastic , NFI Transcription Factors/genetics , Neoplasms/genetics , Disease Progression , Genome-Wide Association Study/methods , Humans , NFI Transcription Factors/metabolism , Neoplasms/metabolism , Neoplasms/pathology , Neoplastic Stem Cells/metabolism , Protein Isoforms/genetics , Protein Isoforms/metabolism , Tumor Microenvironment/genetics
4.
Cell Mol Life Sci ; 73(20): 3789-800, 2016 10.
Article in English | MEDLINE | ID: mdl-27544210

ABSTRACT

The skin forms a vital barrier between an organism's external environment, providing protection from pathogens and numerous physical and chemical threats. Moreover, the intact barrier is essential to prevent water and electrolyte loss without which terrestrial life could not be maintained. Accordingly, acute disruption of the skin through physical or chemical trauma needs to be repaired timely and efficiently as sustained skin pathologies ranging from mild irritations and inflammation through to malignancy impact considerably on morbidity and mortality. The Nuclear Hormone Receptor Family of transcriptional regulators has proven to be highly valuable targets for addressing a range of pathologies, including metabolic syndrome and cancer. Indeed members of the classic endocrine sub-group, such as the glucocorticoid, retinoid, and Vitamin D receptors, represent mainstay treatment strategies for numerous inflammatory skin disorders, though side effects from prolonged use are common. Emerging evidence has now highlighted important functional roles for nuclear receptors belonging to the adopted and orphan subgroups in skin physiology and patho-physiology. This review will focus on these subgroups and explore the current evidence that suggests these nuclear receptor hold great promise as future stand-alone or complementary drug targets in treating common skin diseases and maintaining skin homeostasis.


Subject(s)
Health , Molecular Targeted Therapy , Receptors, Cytoplasmic and Nuclear/metabolism , Skin Diseases/metabolism , Animals , Humans
5.
J Neurosci ; 34(8): 2921-30, 2014 Feb 19.
Article in English | MEDLINE | ID: mdl-24553933

ABSTRACT

Epigenetic mechanisms are essential in regulating neural progenitor cell self-renewal, with the chromatin-modifying protein Enhancer of zeste homolog 2 (EZH2) emerging as a central player in promoting progenitor cell self-renewal during cortical development. Despite this, how Ezh2 is itself regulated remains unclear. Here, we demonstrate that the transcription factor nuclear factor IB (NFIB) plays a key role in this process. Nfib(-/-) mice exhibit an increased number of proliferative ventricular zone cells that express progenitor cell markers and upregulation of EZH2 expression within the neocortex and hippocampus. NFIB binds to the Ezh2 promoter and overexpression of NFIB represses Ezh2 transcription. Finally, key downstream targets of EZH2-mediated epigenetic repression are misregulated in Nfib(-/-) mice. Collectively, these results suggest that the downregulation of Ezh2 transcription by NFIB is an important component of the process of neural progenitor cell differentiation during cortical development.


Subject(s)
Cerebral Cortex/growth & development , Epigenesis, Genetic/physiology , NFI Transcription Factors/genetics , NFI Transcription Factors/physiology , Polycomb Repressive Complex 2/genetics , Polycomb Repressive Complex 2/physiology , Animals , Cell Count , Cerebral Cortex/cytology , Cerebral Cortex/physiology , Electrophoretic Mobility Shift Assay , Enhancer of Zeste Homolog 2 Protein , Female , Hippocampus/cytology , Hippocampus/growth & development , Immunohistochemistry , Male , Mice , Mice, Knockout , Microarray Analysis , Mutation/genetics , Mutation/physiology , Neural Stem Cells/physiology , Primary Cell Culture , Promoter Regions, Genetic/genetics , Real-Time Polymerase Chain Reaction
6.
Cereb Cortex ; 24(1): 261-79, 2014 Jan.
Article in English | MEDLINE | ID: mdl-23042739

ABSTRACT

Neural progenitor cells have the ability to give rise to neurons and glia in the embryonic, postnatal and adult brain. During development, the program regulating whether these cells divide and self-renew or exit the cell cycle and differentiate is tightly controlled, and imbalances to the normal trajectory of this process can lead to severe functional consequences. However, our understanding of the molecular regulation of these fundamental events remains limited. Moreover, processes underpinning development of the postnatal neurogenic niches within the cortex remain poorly defined. Here, we demonstrate that Nuclear factor one X (NFIX) is expressed by neural progenitor cells within the embryonic hippocampus, and that progenitor cell differentiation is delayed within Nfix(-/-) mice. Moreover, we reveal that the morphology of the dentate gyrus in postnatal Nfix(-/-) mice is abnormal, with fewer subgranular zone neural progenitor cells being generated in the absence of this transcription factor. Mechanistically, we demonstrate that the progenitor cell maintenance factor Sry-related HMG box 9 (SOX9) is upregulated in the hippocampus of Nfix(-/-) mice and demonstrate that NFIX can repress Sox9 promoter-driven transcription. Collectively, our findings demonstrate that NFIX plays a central role in hippocampal morphogenesis, regulating the formation of neuronal and glial populations within this structure.


Subject(s)
Cell Differentiation/physiology , Hippocampus/embryology , NFI Transcription Factors/physiology , Neural Stem Cells/physiology , Animals , Cell Count , Coloring Agents , Computational Biology , Dentate Gyrus/embryology , Dentate Gyrus/growth & development , Dentate Gyrus/physiology , Electrophoretic Mobility Shift Assay , Electroporation , Female , Hematoxylin , Hippocampus/cytology , Hippocampus/metabolism , Immunohistochemistry , In Situ Hybridization , Luciferases/genetics , Mice , Mice, Inbred C57BL , Mice, Knockout , Microarray Analysis , NFI Transcription Factors/genetics , Neural Stem Cells/metabolism , Paraffin Embedding , Pregnancy , Promoter Regions, Genetic/genetics , Real-Time Polymerase Chain Reaction
7.
Exp Dermatol ; 23(7): 449-52, 2014 Jul.
Article in English | MEDLINE | ID: mdl-24758341

ABSTRACT

Ultraviolet radiation (UVR) is the most common mutagen that melanocytes are exposed to. UVR causes a diverse range of DNA photolesions contributing to genome instability and promotes melanoma and non-melanoma development. Melanocytes are pigment-producing cells that synthesise the photoprotective melanins when the melanocortin-1 receptor (MC1R) is activated. MC1R is a G-protein-coupled receptor expressed predominantly in melanocytes. Its signalling pathway has been directly linked to melanogenesis, enhanced cytoprotection against UV damage and augmented DNA repair response. Interestingly, previous studies have revealed that MC1R signalling induces the transcription of the NR4A subfamily of orphan nuclear receptors in response to UV. In line with this, studies have also observed that NR4A receptors are recruited to distinct nuclear foci in response to cellular stress, independent of their transcriptional roles. Here, we review the regulated expression of NR4A2 and its potential roles upon cellular stress conditions. Current work in developing synthetic NR4A2 agonists further provides exciting avenues for exploring the potential role of NR4A2 as an antiskin cancer drug target.


Subject(s)
DNA Repair , Gene Expression Regulation , Melanocytes/cytology , Melanocytes/radiation effects , Nuclear Receptor Subfamily 4, Group A, Member 2/metabolism , Receptor, Melanocortin, Type 1/metabolism , DNA/radiation effects , DNA Damage , Genetic Predisposition to Disease , Humans , Light , Receptors, G-Protein-Coupled/metabolism , Signal Transduction , Skin Neoplasms/drug therapy , Skin Neoplasms/genetics , Skin Neoplasms/metabolism , Ultraviolet Rays
8.
Traffic ; 12(5): 627-43, 2011 May.
Article in English | MEDLINE | ID: mdl-21291502

ABSTRACT

Rab GTPases including Rab27a, Rab38 and Rab32 function in melanosome maturation or trafficking in melanocytes. A screen to identify additional Rabs involved in these processes revealed the localization of GFP-Rab17 on recycling endosomes (REs) and melanosomes in melanocytic cells. Rab17 mRNA expression is regulated by microphthalmia transcription factor (MITF), a characteristic of known pigmentation genes. Rab17 siRNA knockdown in melanoma cells quantitatively increased melanosome concentration at the cell periphery. Rab17 knockdown did not inhibit melanosome maturation nor movement, but it caused accumulation of melanin inside cells. Double knockdown of Rab17 and Rab27a indicated that Rab17 acts on melanosomes downstream of Rab27a. Filopodia are known to play a role in melanosome transfer, and in Rab17 knockdown cells filopodia formation was inhibited. Furthermore, we show that stimulation of melanoma cells with α-melanocyte-stimulating hormone induces filopodia formation, supporting a role for filopodia in melanosome release. Cell stimulation also caused redistribution of REs to the periphery, and knockdown of additional RE-associated Rabs 11a and 11b produced a similar accumulation of melanosomes and melanin to that seen after loss of Rab17. Our findings reveal new functions for RE and Rab17 in pigmentation through a distal step in the process of melanosome release via filopodia.


Subject(s)
Endosomes/metabolism , Melanocytes/cytology , Melanocytes/metabolism , Melanosomes/metabolism , Pseudopodia/metabolism , rab GTP-Binding Proteins/metabolism , Animals , Biomarkers/metabolism , Cell Line, Tumor , Gene Knockdown Techniques , Humans , Melanins/metabolism , Melanocytes/drug effects , Mice , Microphthalmia-Associated Transcription Factor/genetics , Microphthalmia-Associated Transcription Factor/metabolism , Pseudopodia/ultrastructure , RNA, Small Interfering/metabolism , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/metabolism , alpha-MSH/pharmacology , rab GTP-Binding Proteins/genetics
9.
Cell Mol Neurobiol ; 33(7): 867-73, 2013 Oct.
Article in English | MEDLINE | ID: mdl-23852417

ABSTRACT

The transcription factor nuclear factor one X (NFIX) plays a central role during the development of the neocortex and hippocampus, through the activation of astrocyte-specific gene expression and the repression of progenitor-specific pathways. However, our understanding of transcriptional targets of NFIX during cortical development remains limited. Here, we identify the transcription factor Bobby sox (Bbx) as a target for NFI-mediated transcriptional control. BBX is expressed within ventricular zone progenitor cells within the developing neocortex and hippocampus, and its expression is upregulated in Nfix (-/-) mice. Moreover, we reveal that NFIX can repress Bbx promoter-driven expression. Collectively, these data suggest that Bbx is a downstream target of NFIX during development of the forebrain.


Subject(s)
NFI Transcription Factors/metabolism , Prosencephalon/embryology , Prosencephalon/metabolism , Trans-Activators/metabolism , Animals , Base Sequence , Hippocampus/cytology , Hippocampus/metabolism , Mice , Mice, Inbred C57BL , Molecular Sequence Data , Promoter Regions, Genetic/genetics , Prosencephalon/cytology , Transcription, Genetic , Up-Regulation
10.
Microsc Microanal ; 19(3): 507-12, 2013 Jun.
Article in English | MEDLINE | ID: mdl-23578755

ABSTRACT

Replication of chloroplast in plant cells is an essential process that requires co-assembly of the tubulin-like plastid division proteins FtsZ1 and FtsZ2 at mid-chloroplast to form a ring structure called the Z-ring. The Z-ring is stabilized via its interaction with the transmembrane protein ARC6 on the inner envelope membrane of chloroplasts. Plants lacking ARC6 are defective in plastid division and contain only one or two enlarged chloroplasts per cell with abnormal localization of FtsZ: instead of a single Z-ring, many short FtsZ filaments are distributed throughout the chloroplast. ARC6 is thought to be the anchoring point for FtsZ assemblies. To investigate the role of ARC6 in FtsZ anchoring, the mobility of green fluorescent protein-tagged FtsZ assemblies was assessed by single particle tracking in mutant plants lacking the ARC6 protein. Mean square displacement analysis showed that the mobility of FtsZ assemblies is to a large extent characterized by anomalous diffusion behavior (indicative of intermittent binding) and restricted diffusion suggesting that besides ARC6-mediated anchoring, an additional FtsZ-anchoring mechanism is present in chloroplasts.


Subject(s)
Arabidopsis Proteins/metabolism , Chloroplast Proteins/metabolism , Chloroplasts/physiology , DNA Replication , Arabidopsis , Genes, Reporter , Green Fluorescent Proteins/analysis , Green Fluorescent Proteins/genetics , Microscopy, Fluorescence , Protein Binding , Protein Interaction Mapping , Staining and Labeling/methods
11.
J Invest Dermatol ; 142(7): 1845-1857, 2022 07.
Article in English | MEDLINE | ID: mdl-34958806

ABSTRACT

Phenotypic plasticity drives cancer progression, impacts treatment response, and is a major driver of therapeutic resistance. In melanoma, a regulatory axis between the MITF and BRN2 transcription factors has been reported to promote tumor heterogeneity by mediating switching between proliferative and invasive phenotypes, respectively. Despite strong evidence that subpopulations of cells that exhibit a BRN2high/MITFlow expression profile switch to a predominantly invasive phenotype, the mechanisms by which this switch is propagated and promotes invasion remain poorly defined. We have found that a reciprocal relationship between BRN2 and NOTCH1/2 signaling exists in melanoma cells in vitro, within patient datasets, and in in vivo primary and metastatic human tumors that bolsters acquisition of invasiveness. Working through the epigenetic modulator EZH2, the BRN2‒NOTCH1/2 axis is potentially a key mechanism by which the invasive phenotype is maintained. Given the emergence of agents targeting both EZH2 and NOTCH, understanding the mechanism through which BRN2 promotes heterogeneity may provide crucial biomarkers to predict treatment response to prevent metastasis.


Subject(s)
Homeodomain Proteins , Melanoma , POU Domain Factors , Receptor, Notch1 , Receptor, Notch2 , Cell Line, Tumor , Cell Movement , Gene Expression Regulation, Neoplastic , Homeodomain Proteins/genetics , Humans , Melanoma/pathology , Microphthalmia-Associated Transcription Factor/genetics , Neoplasm Invasiveness/genetics , POU Domain Factors/genetics , Receptor, Notch1/genetics , Receptor, Notch2/genetics
12.
J Neurosci ; 30(27): 9127-39, 2010 Jul 07.
Article in English | MEDLINE | ID: mdl-20610746

ABSTRACT

The balance between self-renewal and differentiation of neural progenitor cells is an absolute requirement for the correct formation of the nervous system. Much is known about both the pathways involved in progenitor cell self-renewal, such as Notch signaling, and the expression of genes that initiate progenitor differentiation. However, whether these fundamental processes are mechanistically linked, and specifically how repression of progenitor self-renewal pathways occurs, is poorly understood. Nuclear factor I A (Nfia), a gene known to regulate spinal cord and neocortical development, has recently been implicated as acting downstream of Notch to initiate the expression of astrocyte-specific genes within the cortex. Here we demonstrate that, in addition to activating the expression of astrocyte-specific genes, Nfia also downregulates the activity of the Notch signaling pathway via repression of the key Notch effector Hes1. These data provide a significant conceptual advance in our understanding of neural progenitor differentiation, revealing that a single transcription factor can control both the activation of differentiation genes and the repression of the self-renewal genes, thereby acting as a pivotal regulator of the balance between progenitor and differentiated cell states.


Subject(s)
Basic Helix-Loop-Helix Transcription Factors/metabolism , Cell Differentiation/genetics , Gene Expression Regulation, Developmental/physiology , Homeodomain Proteins/metabolism , NFI Transcription Factors/physiology , Stem Cells/physiology , Telencephalon/cytology , Age Factors , Analysis of Variance , Animals , Basic Helix-Loop-Helix Transcription Factors/genetics , Bromodeoxyuridine/metabolism , Cell Count/methods , Cerebral Ventricles/cytology , Cerebral Ventricles/embryology , Chromatin Immunoprecipitation/methods , Electrophoretic Mobility Shift Assay/methods , Embryo, Mammalian , Gene Expression Regulation, Developmental/genetics , Hippocampus/cytology , Hippocampus/growth & development , Homeodomain Proteins/genetics , Mice , Mice, Inbred C57BL , Mice, Knockout , Microarray Analysis/methods , Mutation/genetics , NFI Transcription Factors/deficiency , Nerve Tissue Proteins/genetics , Nerve Tissue Proteins/metabolism , Octamer Transcription Factor-6/genetics , Octamer Transcription Factor-6/metabolism , Promoter Regions, Genetic/physiology , Receptors, Kainic Acid/genetics , Receptors, Kainic Acid/metabolism , Telencephalon/embryology , Transcription Factor HES-1 , Tumor Suppressor Proteins/genetics , Tumor Suppressor Proteins/metabolism
13.
EMBO J ; 26(24): 5048-60, 2007 Dec 12.
Article in English | MEDLINE | ID: mdl-18007594

ABSTRACT

The Tetrahymena thermophila ribosomal DNA (rDNA) replicon contains dispersed cis-acting replication determinants, including reiterated type I elements that associate with sequence-specific, single-stranded binding factors, TIF1 through TIF4. Here, we show that TIF4, previously implicated in cell cycle-controlled DNA replication and rDNA gene amplification, is the T. thermophila origin recognition complex (TtORC). We further demonstrate that TtORC contains an integral RNA subunit that participates in rDNA origin recognition. Remarkably, this RNA, designated 26T, spans the terminal 282 nts of 26S ribosomal RNA. 26T RNA exhibits extensive complementarity to the type I element T-rich strand and binds the rDNA origin in vivo. Mutations that disrupt predicted interactions between 26T RNA and its complementary rDNA target change the in vitro binding specificity of ORC and diminish in vivo rDNA origin utilization. These findings reveal a role for ribosomal RNA in chromosome biology and define a new mechanism for targeting ORC to replication initiation sites.


Subject(s)
DNA, Ribosomal/metabolism , Origin Recognition Complex/genetics , RNA, Ribosomal/metabolism , Replication Origin , Tetrahymena thermophila/genetics , Animals , Base Sequence , Cell Cycle/physiology , DNA Replication , DNA, Ribosomal/genetics , Humans , Molecular Sequence Data , Mutation , Origin Recognition Complex/metabolism , RNA, Ribosomal/genetics , Ribonucleoproteins/genetics , Ribonucleoproteins/metabolism , Sequence Alignment , Tetrahymena thermophila/metabolism , Transgenes
14.
Arch Biochem Biophys ; 513(2): 94-101, 2011 Sep 15.
Article in English | MEDLINE | ID: mdl-21781955

ABSTRACT

FtsZ was identified in bacteria as the first protein to localize mid-cell prior to division and homologs have been found in many plant species. Bacterial studies demonstrated that FtsZ forms a ring structure that is dynamically exchanged with a soluble pool of FtsZ. Our previous work established that Arabidopsis FtsZ1 and FtsZ2-1 are capable of in vitro self-assembly into two distinct filament types, termed type-I and type-II and noted the presence of filament precursor molecules which prompted this investigation. Using a combination of electron microscopy, gel chromatography and native PAGE revealed that (i) prior to FtsZ assembly initiation the pool consists solely of dimers and (ii) during assembly of the Arabidopsis FtsZ type-II filaments the most common intermediate between the dimer and filament state is a tetramer. Three-dimensional reconstructions of the observed dimer and tetramer suggest these oligomeric forms may represent consecutive steps in type-II filament assembly and a mechanism is proposed, which is expanded to include FtsZ assembly into type-I filaments. Finally, the results permit a discussion of the oligomeric nature of the soluble pool in plants.


Subject(s)
Arabidopsis Proteins/chemistry , Arabidopsis/chemistry , Arabidopsis/ultrastructure , Arabidopsis Proteins/ultrastructure , Dimerization , Imaging, Three-Dimensional , Microscopy, Electron, Transmission , Models, Molecular , Plastids/chemistry , Protein Multimerization , Protein Structure, Quaternary
15.
J Biomed Opt ; 26(10)2021 10.
Article in English | MEDLINE | ID: mdl-34689443

ABSTRACT

SIGNIFICANCE: Peripheral pitting edema is a clinician-administered measure for grading edema. Peripheral edema is graded 0, 1 + , 2 + , 3 + , or 4 + , but subjectivity is a major limitation of this technique. A pilot clinical study for short-wave infrared (SWIR) molecular chemical imaging (MCI) effectiveness as an objective, non-contact quantitative peripheral edema measure is underway. AIM: We explore if SWIR MCI can differentiate populations with and without peripheral edema. Further, we evaluate the technology for correctly stratifying subjects with peripheral edema. APPROACH: SWIR MCI of shins from healthy subjects and heart failure (HF) patients was performed. Partial least squares discriminant analysis (PLS-DA) was used to discriminate the two populations. PLS regression (PLSR) was applied to assess the ability of MCI to grade edema. RESULTS: Average spectra from edema exhibited higher water absorption than non-edema spectra. SWIR MCI differentiated healthy volunteers from a population representing all pitting edema grades with 97.1% accuracy (N = 103 shins). Additionally, SWIR MCI correctly classified shin pitting edema levels in patients with 81.6% accuracy. CONCLUSIONS: Our study successfully achieved the two primary endpoints. Application of SWIR MCI to monitor patients while actively receiving HF treatment is necessary to validate SWIR MCI as an HF monitoring technology.


Subject(s)
Heart Failure , Molecular Imaging , Discriminant Analysis , Edema/diagnostic imaging , Heart Failure/diagnostic imaging , Humans , Least-Squares Analysis
16.
Methods Mol Biol ; 536: 533-43, 2009.
Article in English | MEDLINE | ID: mdl-19378090

ABSTRACT

Protein screening/detection is an essential tool in many laboratories. Owing to the relatively large time investments that are required by standard protocols, the development of methods with higher throughput while maintaining an at least comparable signal-to-noise ratio would be highly beneficial to many researchers. This chapter describes how cold microwave technology can be used to enhance the rate of molecular interactions and provides protocols for dot blots, western blots, and ELISA procedures permitting a completion of all incubation steps (blocking and antibody steps) within 45 min.


Subject(s)
Blotting, Western/methods , Microwaves , Proteins/analysis , Staining and Labeling/methods , Animals , Cells, Cultured , Cold Temperature , Electrophoresis, Polyacrylamide Gel/methods
17.
Pigment Cell Melanoma Res ; 32(1): 9-24, 2019 01.
Article in English | MEDLINE | ID: mdl-29781575

ABSTRACT

The POU domain family of transcription factors play a central role in embryogenesis and are highly expressed in neural crest cells and the developing brain. BRN2 is a class III POU domain protein that is a key mediator of neuroendocrine and melanocytic development and differentiation. While BRN2 is a central regulator in numerous developmental programs, it has also emerged as a major player in the biology of tumourigenesis. In melanoma, BRN2 has been implicated as one of the master regulators of the acquisition of invasive behaviour within the phenotype switching model of progression. As a mediator of melanoma cell phenotype switching, it coordinates the transition to a dedifferentiated, slow cycling and highly motile cell type. Its inverse expression relationship with MITF is believed to mediate tumour progression and metastasis within this model. Recent evidence has now outlined a potential epigenetic switching mechanism in melanoma cells driven by BRN2 expression that induces melanoma cell invasion. We summarize the role of BRN2 in tumour cell dissemination and metastasis in melanoma, while also examining it as a potential metastatic regulator in other tumour models.


Subject(s)
Melanoma/pathology , POU Domain Factors/metabolism , Animals , Humans , Models, Biological , Neoplasm Invasiveness , Neoplasm Metastasis , Phenotype
18.
Pigment Cell Melanoma Res ; 31(1): 51-63, 2018 01.
Article in English | MEDLINE | ID: mdl-28755520

ABSTRACT

A SNP within intron4 of the interferon regulatory factor4 (IRF4) gene, rs12203592*C/T, has been independently associated with pigmentation and age-specific effects on naevus count in European-derived populations. We have characterized the cis-regulatory activity of this intronic region and using human foreskin-derived melanoblast strains, we have explored the correlation between IRF4 rs12203592 homozygous C/C and T/T genotypes with TYR enzyme activity, supporting its association with pigmentation traits. Further, higher IRF4 protein levels directed by the rs12203592*C allele were associated with increased basal proliferation but decreased cell viability following UVR, an etiological factor in melanoma development. Since UVR, and accompanying IFNγ-mediated inflammatory response, is associated with melanomagenesis, we evaluated its effects in the context of IRF4 status. Manipulation of IRF4 levels followed by IFNγ treatment revealed a subset of chemokines and immuno-evasive molecules that are sensitive to IRF4 expression level and genotype including CTLA4 and PD-L1.


Subject(s)
Interferon Regulatory Factors/genetics , Interferon Regulatory Factors/metabolism , Interferon-gamma/pharmacology , Melanocytes/pathology , Melanoma/pathology , Monophenol Monooxygenase/metabolism , Polymorphism, Single Nucleotide , Antiviral Agents/pharmacology , Cell Proliferation , Cell Survival , Cells, Cultured , Gene Expression Regulation , Genetic Predisposition to Disease , Genotype , Humans , Melanocytes/drug effects , Melanocytes/metabolism , Melanoma/drug therapy , Melanoma/genetics , Melanoma/metabolism , Ultraviolet Rays
19.
Mol Cancer Res ; 15(9): 1184-1196, 2017 09.
Article in English | MEDLINE | ID: mdl-28607006

ABSTRACT

Exposure of melanocytes to ultraviolet radiation (UVR) induces the formation of UV lesions that can produce deleterious effects in genomic DNA. Encounters of replication forks with unrepaired UV lesions can lead to several complex phenomena, such as the formation of DNA double-strand breaks (DSBs). The NR4A family of nuclear receptors are transcription factors that have been associated with mediating DNA repair functions downstream of the MC1R signaling pathway in melanocytes. In particular, emerging evidence shows that upon DNA damage, the NR4A2 receptor can translocate to sites of UV lesion by mechanisms requiring post-translational modifications within the N-terminal domain and at a serine residue in the DNA-binding domain at position 337. Following this, NR4A2 aids in DNA repair by facilitating chromatin relaxation, allowing accessibility for DNA repair machinery. Using A2058 and HT144 melanoma cells engineered to stably express wild-type or mutant forms of the NR4A2 proteins, we reveal that the expression of functional NR4A2 is associated with elevated cytoprotection against UVR. Conversely, knockdown of NR4A2 expression by siRNA results in a significant loss of cell viability after UV insult. By analyzing the kinetics of the ensuing 53BP1 and RAD51 foci following UV irradiation, we also reveal that the expression of mutant NR4A2 isoforms, lacking the ability to translocate, transactivate, or undergo phosphorylation, display compromised repair capacity.Implications: These data expand the understanding of the mechanism by which the NR4A2 nuclear receptor can facilitate DNA DSB repair. Mol Cancer Res; 15(9); 1184-96. ©2017 AACR.


Subject(s)
DNA Breaks, Double-Stranded , DNA Repair , DNA, Neoplasm/radiation effects , Melanoma/genetics , Nuclear Receptor Subfamily 4, Group A, Member 2/genetics , Cell Death/radiation effects , Cell Line, Tumor , DNA, Neoplasm/genetics , Humans , Melanocytes/metabolism , Melanocytes/radiation effects , Melanoma/metabolism , Melanoma/pathology , Melanoma/radiotherapy , Nuclear Receptor Subfamily 4, Group A, Member 2/biosynthesis , Nuclear Receptor Subfamily 4, Group A, Member 2/metabolism , RNA, Messenger/genetics , RNA, Messenger/metabolism , Signal Transduction , Transfection , Ultraviolet Rays
20.
EBioMedicine ; 16: 63-75, 2017 Feb.
Article in English | MEDLINE | ID: mdl-28119061

ABSTRACT

While invasion and metastasis of tumour cells are the principle factor responsible for cancer related deaths, the mechanisms governing the process remain poorly defined. Moreover, phenotypic divergence of sub-populations of tumour cells is known to underpin alternative behaviors linked to tumour progression such as proliferation, survival and invasion. In the context of melanoma, heterogeneity between two transcription factors, BRN2 and MITF, has been associated with phenotypic switching between predominantly invasive and proliferative behaviors respectively. Epigenetic changes, in response to external cues, have been proposed to underpin this process, however the mechanism by which the phenotypic switch occurs is unclear. Here we report the identification of the NFIB transcription factor as a novel downstream effector of BRN2 function in melanoma cells linked to the migratory and invasive characteristics of these cells. Furthermore, the function of NFIB appears to drive an invasive phenotype through an epigenetic mechanism achieved via the upregulation of the polycomb group protein EZH2. A notable target of NFIB mediated up-regulation of EZH2 is decreased MITF expression, which further promotes a less proliferative, more invasive phenotype. Together our data reveal that NFIB has the ability to promote dynamic changes in the chromatin state of melanoma cells to facilitate migration, invasion and metastasis.


Subject(s)
Cell Movement/genetics , Enhancer of Zeste Homolog 2 Protein/genetics , Homeodomain Proteins/genetics , Melanoma/genetics , Microphthalmia-Associated Transcription Factor/genetics , NFI Transcription Factors/genetics , POU Domain Factors/genetics , Animals , Blotting, Western , Cell Line, Tumor , Enhancer of Zeste Homolog 2 Protein/metabolism , Gene Expression Regulation, Neoplastic , Homeodomain Proteins/metabolism , Humans , Male , Melanoma/metabolism , Melanoma/pathology , Mice, Inbred BALB C , Mice, Knockout , Microphthalmia-Associated Transcription Factor/metabolism , Microscopy, Fluorescence , NFI Transcription Factors/metabolism , Neoplasm Invasiveness , POU Domain Factors/metabolism , Protein Binding , Reverse Transcriptase Polymerase Chain Reaction , Transplantation, Heterologous
SELECTION OF CITATIONS
SEARCH DETAIL