Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 13 de 13
Filter
1.
Front Immunol ; 12: 744358, 2021.
Article in English | MEDLINE | ID: mdl-34804025

ABSTRACT

Our previous work identified human immunodeficiency virus type I enhancer binding protein 1 (HIVEP1) as a putative driver of LPS-induced NF-κB signaling in humans in vivo. While HIVEP1 is known to interact with NF-ĸB binding DNA motifs, its function in mammalian cells is unknown. We report increased HIVEP1 mRNA expression in monocytes from patients with sepsis and monocytes stimulated by Toll-like receptor agonists and bacteria. In complementary overexpression and gene deletion experiments HIVEP1 was shown to inhibit NF-ĸB activity and induction of NF-ĸB responsive genes. RNA sequencing demonstrated profound transcriptomic changes in HIVEP1 deficient monocytic cells and transcription factor binding site analysis showed enrichment for κB site regions. HIVEP1 bound to the promoter regions of NF-ĸB responsive genes. Inhibition of cytokine production by HIVEP1 was confirmed in LPS-stimulated murine Hivep1-/- macrophages and HIVEP1 knockdown zebrafish exposed to the common sepsis pathogen Streptococcus pneumoniae. These results identify HIVEP1 as a negative regulator of NF-κB in monocytes/macrophages that inhibits proinflammatory reactions in response to bacterial agonists in vitro and in vivo.


Subject(s)
DNA-Binding Proteins/immunology , Inflammation/immunology , Macrophages/immunology , NF-kappa B/immunology , Sepsis/immunology , Transcription Factors/immunology , Animals , DNA-Binding Proteins/metabolism , Humans , Inflammation/metabolism , Macrophages/metabolism , Mice , NF-kappa B/metabolism , Sepsis/metabolism , Transcription Factors/metabolism , Zebrafish
2.
J Am Heart Assoc ; 9(22): e018089, 2020 11 17.
Article in English | MEDLINE | ID: mdl-33167754

ABSTRACT

Background There is still uncertainty about the nature and relative impact of early determinants on childhood blood pressure. This study explored determinants of blood pressure at the age of 6 years in 2 Dutch birth cohorts. Methods and Results Results of hierarchical multiple linear regression analyses in GECKO (Groningen Expert Center for Kids With Obesity) Drenthe study (n=1613) were replicated in ABCD (Amsterdam Born Children and Their Development) study (n=2052). All analyses were adjusted for child's age, sex, height, and body mass index (BMI), and maternal education and subsequently performed in the combined sample. No associations were found between maternal smoking during pregnancy and childhood blood pressure. In the total sample, maternal prepregnancy BMI was positively associated with systolic blood pressure (SBP) (ß [95% CI], 0.09 [0.02-0.16] mm Hg) and diastolic blood pressure (ß [95% CI], 0.11 [0.04-0.17] mm Hg). Children of women with hypertension had higher SBP (ß [95% CI], 0.98 [0.17-1.79] mm Hg). Birth weight standardized for gestational age was inversely associated with SBP (ß [95% CI], -6.93 [-9.25 to -4.61] mm Hg) and diastolic blood pressure (ß [95% CI], -3.65 [-5.70 to -1.61] mm Hg). Longer gestational age was associated with lower SBP (ß [95% CI] per week, -0.25 [-0.42 to -0.08] mm Hg). Breastfeeding for 1 to 3 months was associated with lower SBP (ß [95% CI], -0.96 [-1.82 to -0.09] mm Hg) compared with no or <1 month of breastfeeding. Early BMI gain from the age of 2 to 6 years was positively associated with SBP (ß [95% CI], 0.41 [0.08-0.74] mm Hg) and diastolic blood pressure (ß [95% CI], 0.37 [0.07-0.66] mm Hg), but no effect modification by birth weight was found. Conclusions Higher maternal prepregnancy BMI, maternal hypertension, a relatively lower birth weight for gestational age, shorter gestational age, limited duration of breastfeeding, and more rapid early BMI gain contribute to higher childhood blood pressure at the age of 6 years.


Subject(s)
Birth Weight , Blood Pressure/physiology , Age Factors , Body Mass Index , Breast Feeding , Child , Child, Preschool , Cohort Studies , Female , Humans , Linear Models , Male , Netherlands
3.
Intensive Care Med ; 46(8): 1576-1589, 2020 08.
Article in English | MEDLINE | ID: mdl-32514599

ABSTRACT

PURPOSE: Sepsis is the most frequent cause of acute kidney injury (AKI). The "Acute Disease Quality Initiative Workgroup" recently proposed new definitions for AKI, classifying it as transient or persistent. We investigated the incidence, mortality, and host response aberrations associated with transient and persistent AKI in sepsis patients. METHODS: A total of 1545 patients admitted with sepsis to 2 intensive care units in the Netherlands were stratified according to the presence (defined by any urine or creatinine RIFLE criterion within the first 48 h) and evolution of AKI (with persistent defined as remaining > 48 h). We determined 30-day mortality by logistic regression adjusting for confounding variables and analyzed 16 plasma biomarkers reflecting pathways involved in sepsis pathogenesis (n = 866) and blood leukocyte transcriptomes (n = 392). RESULTS: AKI occurred in 37.7% of patients, of which 18.4% was transient and 81.6% persistent. On admission, patients with persistent AKI had higher disease severity scores and more frequently had severe (injury or failure) RIFLE AKI stages than transient AKI patients. Persistent AKI, but not transient AKI, was associated with increased mortality by day 30 and up to 1 year. Persistent AKI was associated with enhanced and sustained inflammatory and procoagulant responses during the first 4 days, and a more severe loss of vascular integrity compared with transient AKI. Baseline blood gene expression showed minimal differences with respect to the presence or evolution of AKI. CONCLUSION: Persistent AKI is independently associated with sepsis mortality, as well as with sustained inflammatory and procoagulant responses, and loss of vascular integrity as compared with transient AKI.


Subject(s)
Acute Kidney Injury , Sepsis , Acute Kidney Injury/etiology , Critical Illness , Humans , Intensive Care Units , Netherlands/epidemiology , Prospective Studies , Sepsis/complications
5.
Brain ; 140(12): 3252-3268, 2017 Dec 01.
Article in English | MEDLINE | ID: mdl-29155979

ABSTRACT

The Dlg4 gene encodes for post-synaptic density protein 95 (PSD95), a major synaptic protein that clusters glutamate receptors and is critical for plasticity. PSD95 levels are diminished in ageing and neurodegenerative disorders, including Alzheimer's disease and Huntington's disease. The epigenetic mechanisms that (dys)regulate transcription of Dlg4/PSD95, or other plasticity genes, are largely unknown, limiting the development of targeted epigenome therapy. We analysed the Dlg4/PSD95 epigenetic landscape in hippocampal tissue and designed a Dlg4/PSD95 gene-targeting strategy: a Dlg4/PSD95 zinc finger DNA-binding domain was engineered and fused to effector domains to either repress (G9a, Suvdel76, SKD) or activate (VP64) transcription, generating artificial transcription factors or epigenetic editors (methylating H3K9). These epi-editors altered critical histone marks and subsequently Dlg4/PSD95 expression, which, importantly, impacted several hippocampal neuron plasticity processes. Intriguingly, transduction of the artificial transcription factor PSD95-VP64 rescued memory deficits in aged and Alzheimer's disease mice. Conclusively, this work validates PSD95 as a key player in memory and establishes epigenetic editing as a potential therapy to treat human neurological disorders.


Subject(s)
Alzheimer Disease/genetics , Behavior, Animal , Cognition , Disks Large Homolog 4 Protein/genetics , Epigenetic Repression , Hippocampus/metabolism , Memory , Transcriptional Activation , Alzheimer Disease/pathology , Alzheimer Disease/physiopathology , Alzheimer Disease/psychology , Amyloid beta-Protein Precursor/genetics , Animals , Disease Models, Animal , Epigenesis, Genetic , Histone Code , Humans , Mice , Mice, Transgenic , Rats , Zinc Fingers
6.
Hum Mol Genet ; 26(20): 4067-4085, 2017 10 15.
Article in English | MEDLINE | ID: mdl-29016858

ABSTRACT

Pre-pregnancy maternal obesity is associated with adverse offspring outcomes at birth and later in life. Individual studies have shown that epigenetic modifications such as DNA methylation could contribute. Within the Pregnancy and Childhood Epigenetics (PACE) Consortium, we meta-analysed the association between pre-pregnancy maternal BMI and methylation at over 450,000 sites in newborn blood DNA, across 19 cohorts (9,340 mother-newborn pairs). We attempted to infer causality by comparing the effects of maternal versus paternal BMI and incorporating genetic variation. In four additional cohorts (1,817 mother-child pairs), we meta-analysed the association between maternal BMI at the start of pregnancy and blood methylation in adolescents. In newborns, maternal BMI was associated with small (<0.2% per BMI unit (1 kg/m2), P < 1.06 × 10-7) methylation variation at 9,044 sites throughout the genome. Adjustment for estimated cell proportions greatly attenuated the number of significant CpGs to 104, including 86 sites common to the unadjusted model. At 72/86 sites, the direction of the association was the same in newborns and adolescents, suggesting persistence of signals. However, we found evidence for acausal intrauterine effect of maternal BMI on newborn methylation at just 8/86 sites. In conclusion, this well-powered analysis identified robust associations between maternal adiposity and variations in newborn blood DNA methylation, but these small effects may be better explained by genetic or lifestyle factors than a causal intrauterine mechanism. This highlights the need for large-scale collaborative approaches and the application of causal inference techniques in epigenetic epidemiology.


Subject(s)
Maternal Inheritance/genetics , Obesity/complications , Pregnancy Outcome/genetics , Adult , Body Mass Index , Cohort Studies , DNA Methylation/genetics , Epigenesis, Genetic/genetics , Epigenomics/methods , Female , Humans , Infant, Newborn , Male , Maternal Inheritance/physiology , Mothers , Pregnancy/physiology , Pregnancy Outcome/epidemiology , Prenatal Exposure Delayed Effects/genetics , Prenatal Exposure Delayed Effects/metabolism
7.
Nat Genet ; 48(12): 1462-1472, 2016 12.
Article in English | MEDLINE | ID: mdl-27798627

ABSTRACT

The genetic architecture of human reproductive behavior-age at first birth (AFB) and number of children ever born (NEB)-has a strong relationship with fitness, human development, infertility and risk of neuropsychiatric disorders. However, very few genetic loci have been identified, and the underlying mechanisms of AFB and NEB are poorly understood. We report a large genome-wide association study of both sexes including 251,151 individuals for AFB and 343,072 individuals for NEB. We identified 12 independent loci that are significantly associated with AFB and/or NEB in a SNP-based genome-wide association study and 4 additional loci associated in a gene-based effort. These loci harbor genes that are likely to have a role, either directly or by affecting non-local gene expression, in human reproduction and infertility, thereby increasing understanding of these complex traits.


Subject(s)
Birth Order , Genome-Wide Association Study , Parity/genetics , Quantitative Trait Loci , Reproduction/genetics , Reproductive Behavior/physiology , Female , Fertility/genetics , Humans , Maternal Age , Phenotype , Polymorphism, Single Nucleotide/genetics , Pregnancy
8.
J Breast Cancer ; 19(1): 45-52, 2016 Mar.
Article in English | MEDLINE | ID: mdl-27066095

ABSTRACT

PURPOSE: MicroRNAs (miRNAs) have received much attention owing to their aberrant expression in various stages of cancer. In many biological processes, miRNAs negatively regulate gene expression, and may be useful in therapeutic strategies. The present study evaluated the effects of silibinin (silybin), a natural flavonoid, on miRNA expression and attempted to elucidate therapeutic targets in MCF-7 breast cancer cells. METHODS: The rates of cell proliferation and apoptosis were determined in silibinin-treated and untreated MCF-7 cells. Furthermore, the expression levels of miR-21 and miR-155 were measured in MCF-7 cells after incubation with silibinin (100 µg/mL), and the putative targets of the miRNAs within the apoptotic pathways were predicted using bioinformatic approaches. The expression levels of some of these targets were evaluated by quantitative reverse transcription polymerase chain reaction (qRT-PCR). RESULTS: Silibinin induced apoptosis in MCF-7 cells in a dose- and time-dependent manner. qRT-PCR analysis revealed a decrease in miR-21 and miR-155 expression levels in silibinin-treated cells relative to the levels in the untreated cells. Potential miR-21 and miR-155 targets within the apoptotic pathways, such as CASP-9, BID, APAF-1, CASP-3, CASP-8, and PDCD4, were predicted by in silico analysis. qRT-PCR analysis showed upregulation of some of these potential targets including caspase-9 (CASP-9) and BID after silibinin treatment for 48 hours. CONCLUSION: Our results suggest a correlation between the expression of miR-21 and miR-155, and MCF-7 cell proliferation. The antiproliferative activity of silibinin may partly be attributable to the downregulation of miR-21 and miR-155, and the upregulation of their apoptotic targets. Furthermore, the upregulation of CASP-9 and BID indicates that silibinin induces apoptosis through both the extrinsic and intrinsic pathways.

9.
Epigenetics ; 10(5): 384-96, 2015.
Article in English | MEDLINE | ID: mdl-25830725

ABSTRACT

Epigenetic silencing of tumor suppressor genes (TSGs) is considered a significant event in the progression of cancer. For example, EPB41L3, a potential biomarker in cervical cancer, is often silenced by cancer-specific promoter methylation. Artificial transcription factors (ATFs) are unique tools to re-express such silenced TSGs to functional levels; however, the induced effects are considered transient. Here, we aimed to improve the efficiency and sustainability of gene re-expression using engineered zinc fingers fused to VP64 (ZF-ATFs) or DNA methylation modifiers (ZF-Tet2 or ZF-TDG) and/or by co-treatment with epigenetic drugs [5-aza-2'-deoxycytidine or Trichostatin A (TSA)]. The EPB41L3-ZF effectively bound its methylated endogenous locus, as also confirmed by ChIP-seq. ZF-ATFs reactivated the epigenetically silenced target gene EPB41L3 (∼ 10-fold) in breast, ovarian, and cervical cancer cell lines. Prolonged high levels of EPB41L3 (∼ 150-fold) induction could be achieved by short-term co-treatment with epigenetic drugs. Interestingly, for otherwise ineffective ZF-Tet2 or ZF-TDG treatments, TSA facilitated re-expression of EPB41L3 up to twofold. ATF-mediated re-expression demonstrated a tumor suppressive role for EPB41L3 in cervical cancer cell lines. In conclusion, epigenetic reprogramming provides a novel way to improve sustainability of re-expression of epigenetically silenced promoters.


Subject(s)
Antineoplastic Agents/pharmacology , DNA Methylation/drug effects , Epigenesis, Genetic , Gene Expression Regulation, Neoplastic/drug effects , Microfilament Proteins/metabolism , Transcription Factors/metabolism , Azacitidine/analogs & derivatives , Azacitidine/pharmacology , Cell Line, Tumor , Decitabine , Histone Code/drug effects , Histone Deacetylases/metabolism , Humans , Hydroxamic Acids/pharmacology , RNA, Messenger/metabolism , Transcription Factors/chemistry
10.
Front Oncol ; 5: 22, 2015.
Article in English | MEDLINE | ID: mdl-25705610

ABSTRACT

Epigenetic modifications such as histone post-transcriptional modifications, DNA methylation, and non-protein-coding RNAs organize the DNA in the nucleus of eukaryotic cells and are critical for the spatio-temporal regulation of gene expression. These epigenetic modifications are reversible and precisely regulated by epigenetic enzymes. In addition to genetic mutations, epigenetic modifications are highly disrupted in cancer relative to normal tissues. Many epigenetic alterations (epi-mutations) are associated with aberrations in the expression and/or activity of epigenetic enzymes. Thus, epigenetic regulators have emerged as prime targets for cancer therapy. Currently, several inhibitors of epigenetic enzymes (epi-drugs) have been approved for use in the clinic to treat cancer patients with hematological malignancies. However, one potential disadvantage of epi-drugs is their lack of locus-selective specificity, which may result in the over-expression of undesirable parts of the genome. The emerging and rapidly growing field of epigenome engineering has opened new grounds for improving epigenetic therapy in view of reducing the genome-wide "off-target" effects of the treatment. In the current review, we will first describe the language of epigenetic modifications and their involvement in cancer. Next, we will overview the current strategies for engineering of artificial DNA-binding domains in order to manipulate and ultimately normalize the aberrant landscape of the cancer epigenome (epigenome engineering). Lastly, the potential clinical applications of these emerging genome-engineering approaches will be discussed.

11.
Breast Cancer Res ; 16(4): 412, 2014 Jul 29.
Article in English | MEDLINE | ID: mdl-25410383

ABSTRACT

DNA methylation and histone modifications are important epigenetic modifications associated with gene (dys)regulation. The epigenetic modifications are balanced by epigenetic enzymes, so-called writers and erasers, such as DNA (de)methylases and histone (de)acetylases. Aberrant epigenetic alterations have been associated with various diseases, including breast cancer. Since aberrant epigenetic modifications are potentially reversible, they might represent targets for breast cancer therapy. Indeed, several drugs have been designed to inhibit epigenetic enzymes (epi-drugs), thereby reversing epigenetic modifications. US Food and Drug Administration approval has been obtained for some epi-drugs for hematological malignancies. However, these drugs have had very modest anti-tumor efficacy in phase I and II clinical trials in breast cancer patients as monotherapy. Therefore, current clinical trials focus on the combination of epi-drugs with other therapies to enhance or restore the sensitivity to such therapies. This approach has yielded some promising results in early phase II trials. The disadvantage of epi-drugs, however, is genome-wide effects, which may cause unwanted upregulation of, for example, pro-metastatic genes. Development of gene-targeted epigenetic modifications (epigenetic editing) in breast cancer can provide a novel approach to prevent such unwanted events. In this context, identification of crucial epigenetic modifications regulating key genes in breast cancer is of critical importance. In this review, we first describe aberrant DNA methylation and histone modifications as two important classes of epigenetic mutations in breast cancer. Then we focus on the preclinical and clinical epigenetic-based therapies currently being explored for breast cancer. Finally, we describe epigenetic editing as a promising new approach for possible applications towards more targeted breast cancer treatment.


Subject(s)
Breast Neoplasms/genetics , Epigenesis, Genetic , Epigenomics , Animals , Antineoplastic Agents/pharmacology , Antineoplastic Agents/therapeutic use , Breast Neoplasms/drug therapy , Breast Neoplasms/metabolism , Clinical Trials as Topic , DNA (Cytosine-5-)-Methyltransferases/antagonists & inhibitors , DNA Methylation/drug effects , Drug Evaluation, Preclinical , Epigenesis, Genetic/drug effects , Female , Gene Expression Regulation, Neoplastic/drug effects , Histone Deacetylase Inhibitors/pharmacology , Histone Deacetylase Inhibitors/therapeutic use , Histones/metabolism , Humans , Treatment Outcome
12.
Mol Cancer Res ; 11(9): 1029-39, 2013 Sep.
Article in English | MEDLINE | ID: mdl-23814024

ABSTRACT

UNLABELLED: The human epidermal growth factor receptor-2 (HER2/neu/ERBB2) is overexpressed in several cancer types. Although therapies targeting the HER2/neu protein result in inhibition of cell proliferation, the anticancer effect might be further optimized by limiting HER2/neu expression at the DNA level. Towards this aim, epigenetic editing was performed to suppress HER2/neu expression by inducing epigenetic silencing marks on the HER2/neu promoter.HER2/neu expression and HER2/neu promoter epigenetic modification status were determined in a panel of ovarian and breast cancer cell lines. HER2/neu-overexpressing cancer cells were transduced to express a zinc finger protein (ZFP), targeting the HER2/neugene, fused to histone methyltransferases (G9a, SUV39-H1)/super KRAB domain (SKD). Epigenetic assessment of the HER2/neu promoter showed that HER2/neu-ZFP fused to G9a efficiently induced the intended silencing histone methylation mark (H3K9me2). Importantly, H3K9me2 induction was associated with a dramatic downregulation of HER2/neu expression in HER2/neu- overexpressing cells. Downregulation by SKD, traditionally considered transient in nature, was associated with removal of the histone acetylation mark (H3ac). The downregulation of HER2/neu by induced H3K9 methylation and/or reduced H3 acetylation was sufficient to effectively inhibit cellular metabolic activity and clonogenicity. Furthermore, genome-wide analysis indicated preferential binding of the ZFP to its target sequence. These results not only show that H3K9 methylation can be induced but also that this epigenetic mark was instructive in promoting downregulation of HER2/neu expression. IMPLICATIONS: Epigenetic editing provides a novel (synergistic) approach to modulate expression of oncogenes.


Subject(s)
Breast Neoplasms/genetics , Epigenesis, Genetic , Gene Silencing , Ovarian Neoplasms/genetics , Receptor, ErbB-2/genetics , Receptor, ErbB-2/metabolism , Breast Neoplasms/metabolism , Breast Neoplasms/pathology , Cell Line, Tumor , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , Down-Regulation , Female , Gene Expression Regulation, Neoplastic , HEK293 Cells , Histone Methyltransferases , Histone-Lysine N-Methyltransferase/metabolism , Humans , MCF-7 Cells , Methylation , Ovarian Neoplasms/metabolism , Ovarian Neoplasms/pathology , Promoter Regions, Genetic , Transcription Factors/genetics , Transcription Factors/metabolism
13.
Iran Biomed J ; 13(3): 153-60, 2009 Jul.
Article in English | MEDLINE | ID: mdl-19688021

ABSTRACT

BACKGROUND: While articular chondrocytes are among those appropriate candidates for cartilage regeneration, the cell dedifferentiation during monolayer culture has limited their application. Several investigations have indicated the usefulness of alginate, but the topic of proliferation and differentiation of chondrocytes in alginate culture has still remained controversial. METHODS: Rat articular chondrocytes were released by enzymatic digestion, plated at 5 x 104 cells/cm2 and culture-expanded. Passaged-5 cells were then cultivated in alginate as 2-mm beads for a period of two months during which the expansion rate and the level of cell differentiation were determined by [3-(A, 5- dimethylthiazolyl- 2-yl)-1, 5-diphenyl tetrazolium bromide] assay and real-time PCR analysis respectively and compared with those of chondrocytes in monolayer culture. RESULTS: Average population doubling time in alginate cultures (10.04 +/- 0.9 days) tended to be significantly (P<0.05) higher than that in monolayer cultures (2.94 +/- 0.3 days). The period of alginate culture could be subdivided into expansion phase (Days 0-40); during which proliferation appeared to be high and differentiation phase (Days 40-60) during which the expression of cartilage-specific genes including collagen II and aggrecan tended to be upregulated. During both the proliferation and differentiation phases, the expression of collagen I was low. At chondrocytes monolayer cultures, the proliferating cells appeared to have a very low expression level of cartilage-specific genes and a high expression level of collagen I gene during the entire culture period (P<0.05). CONCLUSION: It seems that alginate provides conditions in which rat articular chondrocytes are able to undergo proliferation and differentiation in certain time point of cultivation period.


Subject(s)
Alginates/pharmacology , Cartilage, Articular/cytology , Cell Differentiation/drug effects , Cell Proliferation/drug effects , Chondrocytes/cytology , Chondrocytes/physiology , Animals , Cell Count/methods , Cell Culture Techniques/instrumentation , Cell Culture Techniques/methods , Cell Differentiation/physiology , Chondrocytes/drug effects , Chondrocytes/ultrastructure , Gels/pharmacology , Glucuronic Acid/pharmacology , Hexuronic Acids/pharmacology , Male , Microscopy, Electron , Microscopy, Electron, Scanning , Rats , Rats, Wistar , Time Factors , Tissue Engineering/instrumentation , Tissue Engineering/methods
SELECTION OF CITATIONS
SEARCH DETAIL
...