Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 27
Filter
Add more filters










Publication year range
1.
Neurogenetics ; 21(3): 205-215, 2020 07.
Article in English | MEDLINE | ID: mdl-32377986

ABSTRACT

Chronic postsurgical pain (CPSP) is a debilitating chronic pain condition that has a substantial effect on quality of life. CPSP shows considerable clinical overlap with different chronic peripheral pain syndromes, suggesting a shared aetiology. This study aims to assess the genetic overlap between different chronic pain syndromes and CPSP, providing relevant biological context for potential chronic pain markers of CPSP. To analyse the genetic overlap between CPSP and chronic peripheral pain syndromes, recent GWAS studies were combined for polygenic risk scores (PRS) analysis, using a cohort of CPSP patients as starting point. Biological contextualisation of genetic marker, overlap between CPSP and chronic pain syndromes, was assessed through Gene Ontology (GO), using Pathway Scoring Algorithm (PASCAL) and REVIGO. PRS analyses suggest a significant genetic overlap between CPSP and 3 chronic pain disorders: chronic widespread pain (CWP, p value threshold = 0.003, R2 0.06, p = 0.003), rheumatoid arthritis (RA, p value threshold = 0.0177, R2 = 0.04, p = 0.017) and possibly sciatica (p value threshold = 0.00025, R2 = 0.03, p = 0.045). Whereas no significant genetic overlap was found with cluster headache and migraine, the outcome for osteoarthritis (OA) was inconsistent between the cohorts. This is likely related to cohort composition, as repeated random reallocation of patients' nullified CPSP/OA outcome variation between the discovery and replication cohorts. GO analyses suggested an aetiological involvement of genetic markers that control neurological signalling (specifically sodium channels) and inflammatory response. The current study reaffirms the impact of sample size, cohort composition and open data accessibility on the unbiased identification of genetic overlap across disorders. In conclusion, this study is the first to report genetic overlap between regulatory processes implicated in CPSP and chronic peripheral pain syndromes. Interaction between neurological signalling and inflammatory response may explain the genetic overlap between CPSP, CWP and RA. Enhanced understanding of mechanisms underlying chronification of pain will aid the development of new therapeutic strategies for CPSP with sodium channel biochemistry as a potential candidate.


Subject(s)
Chronic Pain/genetics , Osteoarthritis/genetics , Pain, Postoperative/genetics , Algorithms , Cluster Headache/genetics , Cohort Studies , Genome-Wide Association Study , Humans , Inflammation , Migraine Disorders/genetics , Pain Management , Pain, Postoperative/etiology , Phenotype , Quality of Life , Risk Factors , Signal Transduction
2.
Front Immunol ; 11: 378, 2020.
Article in English | MEDLINE | ID: mdl-32231664

ABSTRACT

In the last decade, the microbiota, i.e., combined populations of microorganisms living inside and on the surface of the human body, has increasingly attracted attention of researchers in the medical field. Indeed, since the completion of the Human Microbiome Project, insight and interest in the role of microbiota in health and disease, also through study of its combined genomes, the microbiome, has been steadily expanding. One less explored field of microbiome research has been the female reproductive tract. Research mainly from the past decade suggests that microbial communities residing in the reproductive tract represent a large proportion of the female microbial network and appear to be involved in reproductive failure and pregnancy complications. Microbiome research is facing technological and methodological challenges, as detection techniques and analysis methods are far from being standardized. A further hurdle is understanding the complex host-microbiota interaction and the confounding effect of a multitude of constitutional and environmental factors. A key regulator of this interaction is the maternal immune system that, during the peri-conceptional stage and even more so during pregnancy, undergoes considerable modulation. This review aims to summarize the current literature on reproductive tract microbiota describing the composition of microbiota in different anatomical locations (vagina, cervix, endometrium, and placenta). We also discuss putative mechanisms of interaction between such microbial communities and various aspects of the immune system, with a focus on the characteristic immunological changes during normal pregnancy. Furthermore, we discuss how abnormal microbiota composition, "dysbiosis," is linked to a spectrum of clinical disorders related to the female reproductive system and how the maternal immune system is involved. Finally, based on the data presented in this review, the future perspectives in diagnostic approaches, research directions and therapeutic opportunities are explored.


Subject(s)
Genitalia, Female/immunology , Genitalia, Female/microbiology , Host Microbial Interactions/immunology , Microbiota , Pregnancy/immunology , Dysbiosis/immunology , Female , Humans , Pregnancy Complications/immunology , Pregnancy Complications/microbiology
3.
Cartilage ; 11(2): 203-220, 2020 04.
Article in English | MEDLINE | ID: mdl-29629573

ABSTRACT

OBJECTIVE: Lack of specific marker-sets prohibits definition and functional distinction of cellular subtypes in the intervertebral disc (IVD), such as those from the annulus fibrosus (AF) and the nucleus pulposus (NP). DESIGN: We recently generated immortalized cell lines from human NP and AF tissues; these comprise a set of functionally distinct clonal subtypes. Whole transcriptome analyses were performed of 12 phenotypically distinct clonal cell lines (4× NP-Responder, 4× NP-nonResponder, 2× AF-Sheet forming, and 2× AF-nonSheet forming). Data sets were filtered for membrane-associated marker genes and compared to literature. RESULTS: Comparison of our immortal cell lines to published primary NP, AF, and articular chondrocytes (AC) transcriptome datasets revealed preservation of AF and NP phenotypes. NP-specific membrane-associated genes were defined by comparison to AF cells in both the primary dataset (46 genes) and immortal cell-lines (161 genes). Definition of AF-specific membrane-associated genes yielded 125 primary AF cell and 92 immortal cell-line markers. Overlap between primary and immortal NP cells yielded high-confidence NP-specific marker genes for NP-R (CLDN11, TMEFF2, CA12, ANXA2, CD44) and NP-nR (EFNA1, NETO2, SLC2A1). Overlap between AF and immortal AF subtypes yielded specific markers for AF-S (COLEC12, LPAR1) and AF-nS (CHIC1). CONCLUSIONS: The current study provides a reference platform for preclinical evaluation of novel membrane-associated cell type-specific markers in the IVD. Future research will focus on their biological relevance for IVD function in development, homeostasis, and degenerate conditions.


Subject(s)
Annulus Fibrosus/metabolism , Intervertebral Disc/cytology , Nucleus Pulposus/metabolism , Cell Line , Chondrocytes/metabolism , Gene Expression Profiling , Genetic Markers/genetics , Humans , Phenotype
4.
Sci Rep ; 9(1): 5407, 2019 04 01.
Article in English | MEDLINE | ID: mdl-30932005

ABSTRACT

Expression of the non-classical human leukocyte antigen-G (HLA-G) promotes cancer progression in various malignancies including epithelial ovarian cancer (EOC). As single nucleotide polymorphisms (SNPs) in the HLA-G 3' untranslated region (UTR) regulate HLA-G expression, we investigated HLA-G 3'UTR haplotypes arranged by SNPs in healthy controls (n = 75) and primary EOC patients (n = 79) and determined soluble HLA-G (sHLA-G) levels. Results were related to the clinical status and outcome. Although haplotype frequencies were similar in patients and controls, (i) sHLA-G levels were increased in EOC independent of the haplotype, (ii) homozygosity for UTR-1 or UTR-2 genotypes were significantly associated with metastases formation and presence of circulating tumor cells before therapy, whereas (iii) the UTR-5 and UTR-7 haplotypes were significantly associated with a beneficial clinical outcome regarding negative nodal status, early FIGO staging, and improved overall survival. Lastly, (iv) the ambivalent impact on clinical EOC aspects could be deduced to specific SNPs in the HLA-G 3'UTR: +3187G, +3196G and +3035T alleles. Our results give evidence that even if the genetic background of the HLA-G 3'UTR is identical between patients and controls, certain SNPs have the potential to contribute to diametrical clinical status/outcome in EOC.


Subject(s)
3' Untranslated Regions/genetics , Carcinoma, Ovarian Epithelial/genetics , Genetic Predisposition to Disease/genetics , HLA-G Antigens/genetics , Ovarian Neoplasms/genetics , Polymorphism, Single Nucleotide , Adult , Aged , Aged, 80 and over , Alleles , Carcinoma, Ovarian Epithelial/metabolism , Carcinoma, Ovarian Epithelial/pathology , Female , Gene Frequency , Genotype , HLA-G Antigens/metabolism , Haplotypes , Humans , Kaplan-Meier Estimate , Middle Aged , Neoplasm Staging , Ovarian Neoplasms/metabolism , Ovarian Neoplasms/pathology
5.
Article in English | MEDLINE | ID: mdl-27822313

ABSTRACT

BACKGROUND: A comprehensive assessment of the epigenetic dynamics in cancer cells is the key to understanding the molecular mechanisms underlying cancer and to improving cancer diagnostics, prognostics and treatment. By combining genome-wide ChIP-seq epigenomics and microarray transcriptomics, we studied the effects of oxygen deprivation and subsequent reoxygenation on histone 3 trimethylation of lysine 4 (H3K4me3) and lysine 27 (H3K27me3) in a breast cancer cell line, serving as a model for abnormal oxygenation in solid tumors. A priori, epigenetic markings and gene expression levels not only are expected to vary greatly between hypoxic and normoxic conditions, but also display a large degree of heterogeneity across the cell population. Where traditionally ChIP-seq data are often treated as dichotomous data, the model and experiment here necessitate a quantitative, data-driven analysis of both datasets. RESULTS: We first identified genomic regions with sustained epigenetic markings, which provided a sample-specific reference enabling quantitative ChIP-seq data analysis. Sustained H3K27me3 marking was located around centromeres and intergenic regions, while sustained H3K4me3 marking is associated with genes involved in RNA binding, translation and protein transport and localization. Dynamic marking with both H3K4me3 and H3K27me3 (hypoxia-induced bivalency) was found in CpG-rich regions at loci encoding factors that control developmental processes, congruent with observations in embryonic stem cells. CONCLUSIONS: In silico-identified epigenetically sustained and dynamic genomic regions were confirmed through ChIP-PCR in vitro, and obtained results are corroborated by published data and current insights regarding epigenetic regulation.


Subject(s)
Cell Hypoxia , Histones/metabolism , Cell Line, Tumor , Chromatin Immunoprecipitation , CpG Islands , Cyclin A2/genetics , Cyclin A2/metabolism , Epigenomics , High-Throughput Nucleotide Sequencing , Histones/genetics , Humans , MCF-7 Cells , Methylation , Oligonucleotide Array Sequence Analysis , Sarcoplasmic Reticulum Calcium-Transporting ATPases/genetics , Sarcoplasmic Reticulum Calcium-Transporting ATPases/metabolism , Sequence Analysis, RNA
6.
Article in English | MEDLINE | ID: mdl-27800026

ABSTRACT

BACKGROUND: Trimethylation at histone H3 lysine 4 (H3K4me3) and lysine 27 (H3K27me3) controls gene activity during development and differentiation. Whether H3K4me3 and H3K27me3 changes dynamically in response to altered microenvironmental conditions, including low-oxygen conditions commonly present in solid tumors, is relatively unknown. Demethylation of H3K4me3 and H3K27me3 is mediated by oxygen and 2-oxoglutarate dioxygenases enzymes, suggesting that oxygen deprivation (hypoxia) may influence histone trimethylation. Using the MCF7 breast epithelial adenocarcinoma cell model, we have determined the relationship between epigenomic and transcriptomic reprogramming as a function of fluctuating oxygen tension. RESULTS: We find that in MCF7, H3K4me3 and H3K27me3 marks rapidly increase at specific locations throughout the genome and are largely reversed upon reoxygenation. Whereas dynamic changes are relatively highest for H3K27me3 marking under hypoxic conditions, H3K4me3 occupation is identified as the defining epigenetic marker of transcriptional control. In agreement with the global increase of H3K27 trimethylation, we provide direct evidence that the histone H3K27me3 demethylase KDM6B/JMJD3 is inactivated by limited oxygen. In situ immunohistochemical analysis confirms a marked rise of histone trimethylation in hypoxic tumor areas. Acquisition of H3K27me3 at H3K4me3-marked loci results in a striking increase in "bivalent" epigenetic marking. Hypoxia-induced bivalency substantially overlaps with embryonal stem cell-associated genic bivalency and is retained at numerous loci upon reoxygenation. Transcriptional activity is selectively and progressively dampened at bivalently marked loci upon repeated exposure to hypoxia, indicating that this subset of genes uniquely maintains the potential for epigenetic regulation by KDM activity. CONCLUSIONS: These data suggest that dynamic regulation of the epigenetic state within the tumor environment may have important consequences for tumor plasticity and biology.


Subject(s)
Cell Hypoxia , Epigenesis, Genetic , Histones/metabolism , Chromatin Immunoprecipitation , Genome , High-Throughput Nucleotide Sequencing , Histones/genetics , Humans , Jumonji Domain-Containing Histone Demethylases/metabolism , MCF-7 Cells , Methylation , Sequence Analysis, DNA
7.
BMC Musculoskelet Disord ; 17: 124, 2016 Mar 14.
Article in English | MEDLINE | ID: mdl-26975996

ABSTRACT

BACKGROUND: Immediate early genes (IEGs) encode transcription factors which serve as first line response modules to altered conditions and mediate appropriate cell responses. The immediate early response gene EGR1 is involved in physiological adaptation of numerous different cell types. We have previously shown a role for EGR1 in controlling processes supporting chondrogenic differentiation. We recently established a unique set of phenotypically distinct cell lines from the human nucleus pulposus (NP). Extensive characterization showed that these NP cellular subtypes represented progenitor-like cell types and more functionally mature cells. METHODS: To further understanding of cellular heterogeneity in the NP, we analyzed the response of these cell subtypes to anabolic and catabolic factors. Here, we test the hypothesis that physiological responses of distinct NP cell types are mediated by EGR1 and reflect specification of cell function using an RNA interference-based experimental approach. RESULTS: We show that distinct NP cell types rapidly induce EGR1 exposure to either growth factors or inflammatory cytokines. In addition, we show that mRNA profiles induced in response to anabolic or catabolic conditions are cell type specific: the more mature NP cell type produced a strong and more specialized transcriptional response to IL-1ß than the NP progenitor cells and aspects of this response were controlled by EGR1. CONCLUSIONS: Our current findings provide important substantiation of differential functionality among NP cellular subtypes. Additionally, the data shows that early transcriptional programming initiated by EGR1 is essentially restrained by the cells' epigenome as it was determined during development and differentiation. These studies begin to define functional distinctions among cells of the NP and will ultimately contribute to defining functional phenotypes within the adult intervertebral disc.


Subject(s)
Early Growth Response Protein 1/metabolism , Intervertebral Disc/metabolism , Cell Differentiation , Cell Line , Early Growth Response Protein 1/genetics , Gene Expression Regulation , Humans , Intercellular Signaling Peptides and Proteins/pharmacology , Interleukin-1beta/pharmacology , Intervertebral Disc/cytology , Intervertebral Disc/drug effects , Phenotype , RNA Interference , Time Factors , Transcription, Genetic , Transfection
8.
PLoS One ; 11(1): e0144497, 2016.
Article in English | MEDLINE | ID: mdl-26794306

ABSTRACT

INTRODUCTION: Loss of annulus fibrosus (AF) integrity predisposes to disc herniation and is associated with IVD degeneration. Successful implementation of biomedical intervention therapy requires in-depth knowledge of IVD cell biology. We recently generated unique clonal human nucleus pulposus (NP) cell lines. Recurring functional cellular phenotypes from independent donors provided pivotal evidence for cell heterogeneity in the mature human NP. In this study we aimed to generate and characterize immortal cell lines for the human AF from matched donors. METHODS: Non-degenerate healthy disc material was obtained as surplus surgical material. AF cells were immortalized by simian virus Large T antigen (SV40LTAg) and human telomerase (hTERT) expression. Early passage cells and immortalized cell clones were characterized based on marker gene expression under standardized culturing and in the presence of Transforming Growth factor ß (TGFß). RESULTS: The AF-specific expression signature included COL1A1, COL5A1, COL12A1, SFRP2 and was largely maintained in immortal AF cell lines. Remarkably, TGFß induced rapid 3D sheet formation in a subgroup of AF clones. This phenotype was associated with inherent differences in Procollagen type I processing and maturation, and correlated with differential mRNA expression of Prolyl 4-hydroxylase alpha polypeptide 1 and 3 (P4HA1,3) and Lysyl oxidase (LOX) between clones and differential P4HA3 protein expression between AF cells in histological sections. CONCLUSION: We report for the first time the generation of representative human AF cell lines. Gene expression profile analysis and functional comparison of AF clones revealed variation between immortalized cells and suggests phenotypic heterogeneity in the human AF. Future characterization of AF cellular (sub-)populations aims to combine identification of additional specific AF marker genes and their biological relevance. Ultimately this knowledge will contribute to clinical application of cell-based technology in IVD repair.


Subject(s)
Intervertebral Disc/cytology , Intervertebral Disc/physiology , ADAM Proteins/metabolism , ADAMTS Proteins , Adolescent , Biomarkers/metabolism , Cartilage Oligomeric Matrix Protein/metabolism , Cell Line, Transformed/drug effects , Cell Line, Transformed/metabolism , Collagen Type I/metabolism , Collagen Type I, alpha 1 Chain , Collagen Type II/genetics , Collagen Type II/metabolism , Collagen Type V/genetics , Collagen Type V/metabolism , Female , Humans , Male , Membrane Proteins/genetics , Membrane Proteins/metabolism , Procollagen-Proline Dioxygenase/genetics , Procollagen-Proline Dioxygenase/metabolism , Transforming Growth Factor beta/pharmacology , Transforming Growth Factor beta3/pharmacology
9.
PLoS One ; 10(4): e0118840, 2015.
Article in English | MEDLINE | ID: mdl-25853770

ABSTRACT

Although the MK3 gene was originally found deleted in some cancers, it is highly expressed in others. The relevance of MK3 for oncogenesis is currently not clear. We recently reported that MK3 controls ERK activity via a negative feedback mechanism. This prompted us to investigate a potential role for MK3 in cell proliferation. We here show that overexpression of MK3 induces a proliferative arrest in normal diploid human fibroblasts, characterized by enhanced expression of replication stress- and senescence-associated markers. Surprisingly, MK3 depletion evokes similar senescence characteristics in the fibroblast model. We previously identified MK3 as a binding partner of Polycomb Repressive Complex 1 (PRC1) proteins. In the current study we show that MK3 overexpression results in reduced cellular EZH2 levels and concomitant loss of epigenetic H3K27me3-marking and PRC1/chromatin-occupation at the CDKN2A/INK4A locus. In agreement with this, the PRC1 oncoprotein BMI1, but not the PCR2 protein EZH2, bypasses MK3-induced senescence in fibroblasts and suppresses P16INK4A expression. In contrast, BMI1 does not rescue the MK3 loss-of-function phenotype, suggesting the involvement of multiple different checkpoints in gain and loss of MK3 function. Notably, MK3 ablation enhances proliferation in two different cancer cells. Finally, the fibroblast model was used to evaluate the effect of potential tumorigenic MK3 driver-mutations on cell proliferation and M/SAPK signaling imbalance. Taken together, our findings support a role for MK3 in control of proliferation and replicative life-span, in part through concerted action with BMI1, and suggest that the effect of MK3 modulation or mutation on M/SAPK signaling and, ultimately, proliferation, is cell context-dependent.


Subject(s)
Cell Cycle Checkpoints , Intracellular Signaling Peptides and Proteins/metabolism , Mitogen-Activated Protein Kinase 7/metabolism , Protein Serine-Threonine Kinases/metabolism , Cell Line, Tumor , Cell Proliferation , Cellular Senescence , Fibroblasts/cytology , Fibroblasts/metabolism , Humans , Intracellular Signaling Peptides and Proteins/genetics , MAP Kinase Signaling System , Mutation , Polycomb-Group Proteins/metabolism , Protein Serine-Threonine Kinases/genetics
10.
Nat Commun ; 5: 5203, 2014 Oct 29.
Article in English | MEDLINE | ID: mdl-25351418

ABSTRACT

MicroRNAs are small regulatory RNAs that post transcriptionally control gene expression. Reduced expression of DICER, the enzyme involved in microRNA processing, is frequently observed in cancer and is associated with poor clinical outcome in various malignancies. Yet, the underlying mechanisms are not well understood. Here we identify tumour hypoxia as a regulator of DICER expression in large cohorts of breast cancer patients. We show that DICER expression is suppressed by hypoxia through an epigenetic mechanism that involves inhibition of oxygen-dependent H3K27me3 demethylases KDM6A/B and results in silencing of the DICER promoter. Subsequently, reduced miRNA processing leads to derepression of the miR-200 target ZEB1, stimulates the epithelial to mesenchymal transition and ultimately results in the acquisition of stem cell phenotypes in human mammary epithelial cells. Our study uncovers a previously unknown relationship between oxygen-sensitive epigenetic regulators, miRNA biogenesis and tumour stem cell phenotypes that may underlie poor outcome in breast cancer.


Subject(s)
DEAD-box RNA Helicases/genetics , Epigenesis, Genetic , Neoplastic Stem Cells/metabolism , Neoplastic Stem Cells/pathology , Ribonuclease III/genetics , Breast Neoplasms/genetics , Breast Neoplasms/pathology , Cell Hypoxia/genetics , DEAD-box RNA Helicases/metabolism , Epithelial-Mesenchymal Transition/genetics , Female , Gene Expression Regulation, Neoplastic , Gene Silencing , Humans , Phenotype , Prognosis , Promoter Regions, Genetic/genetics , RNA Processing, Post-Transcriptional/genetics , Ribonuclease III/metabolism
11.
Arthritis Res Ther ; 16(3): R135, 2014 Jun 27.
Article in English | MEDLINE | ID: mdl-24972717

ABSTRACT

INTRODUCTION: Relatively little is known about cellular subpopulations in the mature nucleus pulposus (NP). Detailed understanding of the ontogenetic, cellular and molecular characteristics of functional intervertebral disc (IVD) cell populations is pivotal to the successful development of cell replacement therapies and IVD regeneration. In this study, we aimed to investigate whether phenotypically distinct clonal cell lines representing different subpopulations in the human NP could be generated using immortalization strategies. METHODS: Nondegenerate healthy disc material (age range, 8 to 15 years) was obtained as surplus surgical material. Early passage NP monolayer cell cultures were initially characterized using a recently established NP marker set. NP cells were immortalized by simian virus 40 large T antigen (SV40LTag) and human telomerase reverse transcriptase expression. Immortalized cells were clonally expanded and characterized based on collagen type I, collagen type II, α1 (COL2A1), and SRY-box 9 (SOX9) protein expression profiles, as well as on expression of a subset of established in vivo NP cell lineage markers. RESULTS: A total of 54 immortal clones were generated. Profiling of a set of novel NP markers (CD24, CA12, PAX1, PTN, FOXF1 and KRT19 mRNA) in a representative set of subclones substantiated successful immortalization of multiple cellular subpopulations from primary isolates and confirmed their NP origin and/or phenotype. We were able to identify two predominant clonal NP subtypes based on their morphological characteristics and their ability to induce SOX9 and COL2A1 under conventional differentiation conditions. In addition, cluster of differentiation 24 (CD24)-negative NP responder clones formed spheroid structures in various culture systems, suggesting the preservation of a more immature phenotype compared to CD24-positive nonresponder clones. CONCLUSIONS: Here we report the generation of clonal NP cell lines from nondegenerate human IVD tissue and present a detailed characterization of NP cellular subpopulations. Differential cell surface marker expression and divergent responses to differentiation conditions suggest that the NP subtypes may correspond to distinct maturation stages and represent distinct NP cell subpopulations. Hence, we provide evidence that the immortalization strategy that we applied is capable of detecting cell heterogeneity in the NP. Our cell lines yield novel insights into NP biology and provide promising new tools for studies of IVD development, cell function and disease.


Subject(s)
Cell Proliferation , Gene Expression Profiling/methods , Intervertebral Disc/cytology , Intervertebral Disc/metabolism , Adolescent , Aggrecans/metabolism , CD24 Antigen/genetics , CD24 Antigen/metabolism , Carrier Proteins/genetics , Carrier Proteins/metabolism , Cell Line, Transformed , Cells, Cultured , Child , Clone Cells/cytology , Clone Cells/metabolism , Collagen Type I/metabolism , Collagen Type I, alpha 1 Chain , Collagen Type II/genetics , Collagen Type II/metabolism , Cytokines/genetics , Cytokines/metabolism , Female , Flow Cytometry , Forkhead Transcription Factors/genetics , Forkhead Transcription Factors/metabolism , Humans , Immunoblotting , Immunophenotyping , Male , Paired Box Transcription Factors/genetics , Paired Box Transcription Factors/metabolism , Reverse Transcriptase Polymerase Chain Reaction , SOX9 Transcription Factor/genetics , SOX9 Transcription Factor/metabolism
12.
PLoS One ; 8(3): e58083, 2013.
Article in English | MEDLINE | ID: mdl-23483971

ABSTRACT

Initiation of and progression through chondrogenesis is driven by changes in the cellular microenvironment. At the onset of chondrogenesis, resting mesenchymal stem cells are mobilized in vivo and a complex, step-wise chondrogenic differentiation program is initiated. Differentiation requires coordinated transcriptomic reprogramming and increased progenitor proliferation; both processes require chromatin remodeling. The nature of early molecular responses that relay differentiation signals to chromatin is poorly understood. We here show that immediate early genes are rapidly and transiently induced in response to differentiation stimuli in vitro. Functional ablation of the immediate early factor EGR1 severely deregulates expression of key chondrogenic control genes at the onset of differentiation. In addition, differentiating cells accumulate DNA damage, activate a DNA damage response and undergo a cell cycle arrest and prevent differentiation associated hyper-proliferation. Failed differentiation in the absence of EGR1 affects global acetylation and terminates in overall histone hypermethylation. We report novel molecular connections between EGR1 and Polycomb Group function: Polycomb associated histone H3 lysine27 trimethylation (H3K27me3) blocks chromatin access of EGR1. In addition, EGR1 ablation results in abnormal Ezh2 and Bmi1 expression. Consistent with this functional interaction, we identify a number of co-regulated targets genes in a chondrogenic gene network. We here describe an important role for EGR1 in early chondrogenic epigenetic programming to accommodate early gene-environment interactions in chondrogenesis.


Subject(s)
Chondrogenesis/genetics , Early Growth Response Protein 1/metabolism , Epigenesis, Genetic , Genes, Immediate-Early/genetics , Polycomb-Group Proteins/metabolism , Animals , Cell Cycle , Cell Proliferation , Cells, Cultured , Chondrocytes/cytology , Chondrocytes/metabolism , Core Binding Factor Alpha 1 Subunit/metabolism , DNA Damage/genetics , DNA Replication/genetics , Gene Regulatory Networks/genetics , Histones/metabolism , Mice , SOX9 Transcription Factor/metabolism , Stress, Physiological/genetics
13.
Epigenetics Chromatin ; 5(1): 12, 2012 Aug 07.
Article in English | MEDLINE | ID: mdl-22870894

ABSTRACT

BACKGROUND: Gene-environment interactions are mediated by epigenetic mechanisms. Polycomb Group proteins constitute part of an epigenetic cellular transcriptional memory system that is subject to dynamic modulation during differentiation. Molecular insight in processes that control dynamic chromatin association and dissociation of Polycomb repressive complexes during and beyond development is limited. We recently showed that MK3 interacts with Polycomb repressive complex 1 (PRC1). The functional relevance of this interaction, however, remained poorly understood. MK3 is activated downstream of mitogen- and stress-activated protein kinases (M/SAPKs), all of which fulfill crucial roles during development. We here use activation of the immediate-early response gene ATF3, a bona fide PRC1 target gene, as a model to study how MK3 and its effector kinases MAPK/ERK and SAPK/P38 are involved in regulation of PRC1-dependent ATF3 transcription. RESULTS: Our current data show that mitogenic signaling through ERK, P38 and MK3 regulates ATF3 expression by PRC1/chromatin dissociation and epigenetic modulation. Mitogenic stimulation results in transient P38-dependent H3S28 phosphorylation and ERK-driven PRC1/chromatin dissociation at PRC1 targets. H3S28 phosphorylation by itself appears not sufficient to induce PRC1/chromatin dissociation, nor ATF3 transcription, as inhibition of MEK/ERK signaling blocks BMI1/chromatin dissociation and ATF3 expression, despite induced H3S28 phosphorylation. In addition, we establish that concomitant loss of local H3K27me3 promoter marking is not required for ATF3 activation. We identify pERK as a novel signaling-induced binding partner of PRC1, and provide evidence that MK3 controls ATF3 expression in cultured cells via negative regulatory feedback on M/SAPKs. Dramatically increased ectopic wing vein formation in the absence of Drosophila MK in a Drosophila ERK gain-of-function wing vein patterning model, supports the existence of MK-mediated negative feedback regulation on pERK. CONCLUSION: We here identify and characterize important actors in a PRC1-dependent epigenetic signal/response mechanism, some of which appear to be nonspecific global responses, whereas others provide modular specificity. Our findings provide novel insight into a Polycomb-mediated epigenetic mechanism that dynamically controls gene transcription and support a direct link between PRC1 and cellular responses to changes in the microenvironment.

14.
Chromosoma ; 121(5): 447-64, 2012 Oct.
Article in English | MEDLINE | ID: mdl-22610065

ABSTRACT

Mutations in the A-type lamins A and C, two major components of the nuclear lamina, cause a large group of phenotypically diverse diseases collectively referred to as laminopathies. These conditions often involve defects in chromatin organization. However, it is unclear whether A-type lamins interact with chromatin in vivo and whether aberrant chromatin-lamin interactions contribute to disease. Here, we have used an unbiased approach to comparatively map genome-wide interactions of gene promoters with lamin A and progerin, the mutated lamin A isoform responsible for the premature aging disorder Hutchinson-Gilford progeria syndrome (HGPS) in mouse cardiac myoytes and embryonic fibroblasts. We find that lamin A-associated genes are predominantly transcriptionally silent and that loss of lamin association leads to the relocation of peripherally localized genes, but not necessarily to their activation. We demonstrate that progerin induces global changes in chromatin organization by enhancing interactions with a specific subset of genes in addition to the identified lamin A-associated genes. These observations demonstrate disease-related changes in higher order genome organization in HGPS and provide novel insights into the role of lamin-chromatin interactions in chromatin organization.


Subject(s)
Lamin Type A/metabolism , Nuclear Proteins/metabolism , Progeria/metabolism , Protein Precursors/metabolism , Animals , Cell Line , Chromosome Mapping , Fibroblasts/metabolism , Humans , Lamin Type A/genetics , Mice , Muscle Cells/metabolism , Nuclear Proteins/genetics , Progeria/genetics , Protein Binding , Protein Precursors/genetics
15.
PLoS One ; 7(3): e33467, 2012.
Article in English | MEDLINE | ID: mdl-22428055

ABSTRACT

BACKGROUND: NF-κB/p65 has been reported to be involved in regulation of chondrogenic differentiation. However, its function in relation to key chondrogenic factor Sox9 and onset of chondrogenesis during endochondral ossification is poorly understood. We hypothesized that the early onset of chondrogenic differentiation is initiated by transient NF-κB/p65 signaling. METHODOLOGY/PRINCIPAL FINDINGS: The role of NF-κB/p65 in early chondrogenesis was investigated in different in vitro, ex vivo and in vivo endochondral models: ATDC5 cells, hBMSCs, chicken periosteal explants and growth plates of 6 weeks old mice. NF-κB/p65 activation was manipulated using pharmacological inhibitors, RNAi and activating agents. Gene expression and protein expression analysis, and (immuno)histochemical stainings were employed to determine the role of NF-κB/p65 in the chondrogenic phase of endochondral development. Our data show that chondrogenic differentiation is facilitated by early transient activation of NF-κB/p65. NF-κB/p65-mediated signaling determines early expression of Sox9 and facilitates the subsequent chondrogenic differentiation programming by signaling through key chondrogenic pathways. CONCLUSIONS/SIGNIFICANCE: The presented data demonstrate that NF-κB/p65 signaling, as well as its intensity and timing, represents one of the transcriptional regulatory mechanisms of the chondrogenic developmental program of chondroprogenitor cells during endochondral ossification. Importantly, these results provide novel possibilities to improve the success of cartilage and bone regenerative techniques.


Subject(s)
Chondrogenesis/physiology , Osteogenesis/physiology , Signal Transduction/physiology , Transcription Factor RelA/metabolism , Animals , Bone Morphogenetic Protein 2/metabolism , Cell Line , Cell Proliferation , Chick Embryo , DNA Primers/genetics , Gene Expression Profiling , Immunoblotting , Immunohistochemistry , Mice , Periosteum/cytology , RNA Interference , Real-Time Polymerase Chain Reaction
16.
Nucleus ; 2(3): 195-207, 2011.
Article in English | MEDLINE | ID: mdl-21818413

ABSTRACT

A-type lamins are a major component of the nuclear lamina. Mutations in the LMNA gene, which encodes the A-type lamins A and C, cause a set of phenotypically diverse diseases collectively called laminopathies. While adult LMNA null mice show various symptoms typically associated with laminopathies, the effect of loss of lamin A/C on early post-natal development is poorly understood. Here we developed a novel LMNA null mouse (LMNA(GT-/-)) based on genetrap technology and analyzed its early post-natal development. We detect LMNA transcripts in heart, the outflow tract, dorsal aorta, liver and somites during early embryonic development. Loss of A-type lamins results in severe growth retardation and developmental defects of the heart, including impaired myocyte hypertrophy, skeletal muscle hypotrophy, decreased amounts of subcutaneous adipose tissue and impaired ex vivo adipogenic differentiation. These defects cause death at 2 to 3 weeks post partum associated with muscle weakness and metabolic complications, but without the occurrence of dilated cardiomyopathy or an obvious progeroid phenotype. Our results indicate that defective early post-natal development critically contributes to the disease phenotypes in adult laminopathies.


Subject(s)
Adipogenesis/genetics , Gene Deletion , Lamin Type A/deficiency , Lamin Type A/genetics , Metabolic Diseases/metabolism , Metabolic Diseases/pathology , Muscle Development/genetics , Animals , Embryonic Development/genetics , Fibroblasts/cytology , Fibroblasts/metabolism , Genes, Reporter/genetics , Hypertrophy/genetics , Lamin Type A/metabolism , Male , Metabolic Diseases/genetics , Metabolic Diseases/physiopathology , Mice , Muscle, Skeletal/metabolism , Muscle, Skeletal/pathology , Myocytes, Cardiac/metabolism , Myocytes, Cardiac/pathology , Phenotype , Promoter Regions, Genetic/genetics , Subcutaneous Fat/metabolism , Subcutaneous Fat/pathology , Transcriptome
17.
Methods Mol Biol ; 693: 11-36, 2011.
Article in English | MEDLINE | ID: mdl-21080272

ABSTRACT

Introduction of germ line mutations in mice via genetic engineering involves alterations of the structure and characteristics of genes. These alterations are mostly introduced via molecular genetic technology either in embryonal stem cells or in one-cell stage embryos. This chapter describes classic biotechnological methods used to generate mice from modified pre-implantation embryos.


Subject(s)
Blastocyst/cytology , Microinjections/methods , Animals , Embryo Culture Techniques/methods , Embryo, Mammalian/cytology , Embryonic Stem Cells/cytology , Female , Humans , Male , Mice , Pregnancy
18.
Methods Mol Biol ; 693: 89-101, 2011.
Article in English | MEDLINE | ID: mdl-21080276

ABSTRACT

Transgenics are powerful mouse models to understand the biological functions of genes. This chapter gives a short overview of the requirements and considerations in designing a transgene. In addition, potential important choices that have to be made in advance for the successful designing and generating a transgenic mouse model are discussed. Methods for DNA purification for microinjection are also provided in this chapter.


Subject(s)
Transgenes/genetics , Animals , Mice , Mice, Transgenic , Microinjections/methods , Models, Genetic , Promoter Regions, Genetic/genetics
19.
Nucleus ; 1(6): 460-71, 2010.
Article in English | MEDLINE | ID: mdl-21327087

ABSTRACT

The nuclear envelope and the lamina define the nuclear periphery and are implicated in many nuclear processes including chromatin organization, transcription and DNA replication. Mutations in lamin A proteins, major components of the lamina, interfere with these functions and cause a set of phenotypically diverse diseases referred to as laminopathies. The phenotypic diversity of laminopathies is thought to be the result of alterations in specific protein- and chromatin interactions due to lamin A mutations. Systematic identification of lamin A-protein and -chromatin interactions will be critical to uncover the molecular etiology of laminopathies. Here we summarize and critically discuss recent technology to analyze lamina-protein and-chromatin interactions.


Subject(s)
Chromatin/metabolism , Nuclear Lamina/metabolism , Nuclear Proteins/metabolism , Protein Interaction Mapping , Chromatin Immunoprecipitation , Humans , Lamin Type A/metabolism , Micrococcal Nuclease/chemistry , Micrococcal Nuclease/metabolism , Site-Specific DNA-Methyltransferase (Adenine-Specific)/metabolism
20.
Nucleus ; 1(6): 513-25, 2010.
Article in English | MEDLINE | ID: mdl-21327095

ABSTRACT

The nuclear lamina is an interconnected meshwork of intermediate filament proteins underlying the nuclear envelope. The lamina is an important regulator of nuclear structural integrity as well as nuclear processes, including transcription, DNA replication and chromatin remodeling. The major components of the lamina are A- and B-type lamins. Mutations in lamins impair lamina functions and cause a set of highly tissue-specific diseases collectively referred to as laminopathies. The phenotypic diversity amongst laminopathies is hypothesized to be caused by mutations affecting specific protein interactions, possibly in a tissue-specific manner. Current technologies to identify interaction partners of lamin A and its mutants are hampered by the insoluble nature of lamina components. To overcome the limitations of current technologies, we developed and applied a novel, unbiased approach to identify lamin A-interacting proteins. This approach involves expression of the high-affinity OneSTrEP-tag, precipitation of lamin-protein complexes after reversible protein cross-linking and subsequent protein identification by mass spectrometry. We used this approach to identify in mouse embryonic fibroblasts and cardiac myocyte NklTAg cell lines proteins that interact with lamin A and its mutant isoform progerin, which causes the premature aging disorder Hutchinson-Gilford progeria syndrome (HGPS). We identified a total of 313 lamina-interacting proteins, including several novel lamin A interactors, and we characterize a set of 35 proteins which preferentially interact with lamin A or progerin.


Subject(s)
Lamin Type A/metabolism , Nuclear Proteins/metabolism , Protein Precursors/metabolism , Animals , Cell Line , Fibroblasts/metabolism , Gene Knockout Techniques , Humans , Lamin Type A/chemistry , Lamin Type A/genetics , Mass Spectrometry , Mice , Mutation , Myocytes, Cardiac/metabolism , Nuclear Proteins/chemistry , Nuclear Proteins/genetics , Progeria/metabolism , Progeria/pathology , Protein Isoforms/genetics , Protein Isoforms/metabolism , Protein Precursors/chemistry , Protein Precursors/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...