26 research outputs found

    Supplementary Appendix. All-trans retinoic acid works synergistically with the Îł- secretase inhibitor crenigacestat to augment BCMA on multiple myeloma and the efficacy of BCMA-CAR T cells

    Get PDF
    Supplement Figure 1: ATRA treatment does not affect the viability of myeloma cell lines. MM.1S, OPM-2 and NCI-H929 cells were treated with ATRA for up to 72 hours. Cell viability was measured by flow cytometry and 7AAD staining (n=6). Bar diagrams show mean values +SD.Supplement Figure 2: ATRA plus crenigacestat treatment enhance BCMA expression on myeloma cell lines. Bar diagram shows BCMA expression on OPM-2 cells (n=3) after treatment with 100 nM ATRA and/or 10 nM GSI crenigacestat for 72 hours. Bar diagram shows mean values +SD. P-values between indicated groups were calculated using unpaired t-test. *p<0.05, **p<0.01.Supplement Figure 3: ATRA treatment leads to increased BCMA transcripts in OPM-2 myeloma cells. BCMA RNA levels in OPM-2 were analyzed by quantitative reverse transcription PCR (qRT-PCR) assay after incubation with increasing doses of ATRA for 48 hours (n=3). Bar diagram shows mean values +SD. P-values between indicated groups were calculated using unpaired t-test. *p<0.05.Supplement Figure 4: ATRA treatment leads to enhanced BCMA expression on primary myeloma cells. Representative flow cytometric analysis of BCMA expression on primary myeloma cells that had been cultured in the absence or presence of ATRA at different concentrations for 72 hours. 7-AAD was used to exclude dead cells from analysis.Supplement Figure 5: ATRA treatment does not impair viability of primary myeloma cells. Viability of primary myeloma cells with or without 72 hours of ATRA treatment was analyzed by flow cytometry and 7-AAD staining (n=5 biological replicates). Bar diagram shows mean values +SD.Supplement Figure 6: sBCMA does not impair BCMA CAR T cell functionality. CD8+ BCMA-CAR T-cells were co-cultured with MM.1S target cells in absence or presence of 150 ng/ml of soluble BCMA. After 4 hours, cytotoxicity was evaluated by bioluminescence- based assay. Diagram shows mean values +/-SD.Supplement Figure 7: ATRA treatment does not increase shedding of sBCMA. sBCMA concentration in the supernatant of OPM-2 and NCI-H929 after incubation with increasing doses of ATRA was analyzed by ELISA. Cell lines were cultured at 1x106/well (n=3 technical replicates). Bar diagrams show mean values +SD, P-values between indicated groups were calculated using 2way ANOVA. n.s. = not significant, *p<0.05, **p<0.01.Supplement Figure 8: BCMA-CAR T-cells confer enhanced cytotoxicity against ATRA plus crenigacestat-treated OPM-2 cells in vitro. OPM-2 cells were incubated with 100 nM ATRA and/or 10 nM GSI for 72 hours or were left untreated. Cytolytic activity of CD8+ BCMA- CAR T-cells was determined in a bioluminescence-based assay after 4h of co-incubation with target cells. Assay was performed in triplicate wells with 5,000 target cells per well. Data are presented as mean values +SD (n=4 biological replicates). P-values between indicated groups were calculated using unpaired t-test. n.s. = not significant, *p<0.05.Supplement Figure 9: Patient-derived BCMA-CAR T-cells confer enhanced cytotoxicity against ATRA-treated MM.1S cells. MM.1S cells were incubated with 50 nM ATRA for 72 hours or were left untreated. Cytolytic activity of MM patient-derived CD8+ BCMA-CAR T-cells was determined in a bioluminescence-based assay after 4h of co-incubation with target cells. Data are presented as mean values +SD of triplicate wells. P-values between indicated groups were calculated using unpaired t-test. *p<0.05, **p<0.01.Peer reviewe

    Gestational Diabetes Mellitus and fetal programming: Epigenetic investigations with different Next Generation Sequencing Techniques

    No full text
    Eine intrauterine Gestationsdiabetes (GDM) Exposition induziert in den betroffenen Nachkommen eine lebenslang erhöhte PrĂ€disposition fĂŒr metabolische und komplexe Erkrankungen. Die KrankheitssuszeptibilitĂ€t wird dabei durch epigenetische VerĂ€nderungen vermittelt, die sich ĂŒber die Regulation der GenaktivitĂ€t auch auf das Expressionsniveau und den PhĂ€notypen auswirken. Um neue Gene zu finden, die eine Rolle in der fetalen Programmierung spielen, wurden in dieser Arbeit genomweite Methylierungsmuster von Nabelschnurbluten (FCBs) aus GDM-Schwangerschaften und Kontrollen miteinander verglichen. Mit Illumina Infinium HumanMethylation 450K Arrays konnten signifikante Gruppenunterschiede fĂŒr insgesamt 65 CpG-Stellen (52 davon genassoziiert) festgestellt werden, die multiplem Testen standhielten. Mittels Pyrosequenzierung wurden vier dieser Kandidaten-Loci (ATP5A1, MFAP4, PRKCH, SLC17A4), sowie ein Gen aus der Literatur (HIF3A) genauer untersucht und die Effekte erfolgreich validiert. FĂŒr das zugrundeliegende multivariate Regressionsmodell wurden die potenziellen Störfaktoren Gestationsalter, kindliches Geschlecht und mĂŒtterlicher BMI berĂŒcksichtigt. Der GDM-Effekt zeigte sich stĂ€rker in der insulinbehandelten Subgruppe (I-GDM) als in der diĂ€tisch behandelten (D GDM) und scheint insgesamt multifaktoriell bedingt zu sein, da viele Gene betroffen waren, jedoch alle mit einer vergleichsweise niedrigen Effekt-GrĂ¶ĂŸe. ZusĂ€tzlich konnten fĂŒr den MEG3 Promotor, MEST und PEG3, drei von vier geprĂ€gten Genen, die mittels Deep Bisulfite Sequencings (DBS) analysiert wurden, ebenfalls signifikante Methylierungs-unterschiede zwischen der GDM- und Kontroll-Gruppe detektiert werden. Die identifizierten Gene stellen labile Zielregionen fĂŒr die GDM-induzierte intrauterine Programmierung dar und können zukĂŒnftig nĂŒtzliche Biomarker fĂŒr Krankheitsdiagnosen und Prognosen sein. Mittels DBS können darĂŒber hinaus EinzelmolekĂŒl-Analysen durchgefĂŒhrt werden, fĂŒr die in differentiell methylierten Regionen (DMRs) anhand eines informativen SNPs die parentale Allel-Herkunft bestimmt und bei der Berechnung von Epimutationsraten einbezogen werden kann. Epimutationen wurde als solche gewertet, wenn sie ein > 50 % abnormal (de)methyliertes Methylierungsprofil aufwiesen. Die DBS-Daten wurden mit zwei verschiedenen Sequenzierplattformen generiert (Roche GS Junior und Illumina MiSeq). FĂŒr Zweitere wurde ein eigenes, unabhĂ€ngiges Library-PrĂ€parations-Protokoll entwickelt. In Nabelschnurblut, adultem Blut und Viszeralfett wurden fĂŒr die paternal exprimierte MEST Promotor DMR und die maternal exprimierte MEG3 intergenic (IG) DMR hohe Epimutationsraten fĂŒr das jeweils unmethylierte Allel detektiert. Die geprĂ€gten (methylierten) Allele wiesen dagegen nur niedrige Epimutationsraten auf. Da MEST und MEG3 invers geprĂ€gte Gene sind, war die Hypermethylierung des nicht geprĂ€gten Allels (HNA) demnach unabhĂ€ngig von der parentalen Allel-Herkunft. Die HNA scheint außerdem erst nach der Fertilisation aufzutreten, da in Spermien nur sehr wenige Epimutationen gefunden wurden. FĂŒr die sekundĂ€re MEG3 Promotor DMR (deren PrĂ€gung von der primĂ€ren MEG3 IG-DMR reguliert wird) wurde ein deutlich schwĂ€cherer, wenngleich signifikanter HNA-Effekt im FCB gemessen, fĂŒr die paternal exprimierte PEG3 Promotor DMR konnte dagegen kein signifikanter Unterschied zwischen den beiden parentalen Epimutationsraten festgestellt werden. Der HNA-Effekt fĂŒr die MEST DMR, MEG3 IG-DMR und MEG3 Promotor DMR war weder mit GDM noch mit Adipositas assoziiert und zeigte allgemein eine große interindividuelle Varianz. Die Aufrechterhaltung differenzieller Methylierungsmuster in Imprinting Kontrollregionen (ICRs) scheint in manchen Entwicklungs-Zeitspannen von großer Bedeutung und damit streng kontrolliert zu sein, spĂ€ter jedoch redundant zu werden, was sich in der Anreicherung von stochastischen sowie umweltinduzierten Fehlern auf dem nicht geprĂ€gten Allel Ă€ußern kann. HNA-suszeptible geprĂ€gte Gene Ă€hneln in mancherlei Hinsicht metastabilen Epiallelen. Diese Studie zeigt, dass sowohl stochastische Faktoren als auch Umweltstimuli wĂ€hrend der frĂŒhen embryonalen Entwicklung u.a. ĂŒber HNA-Effekte geprĂ€gte Gen-Netzwerke programmieren, die in Wachstumsprozesse involviert sind. Um tiefere Einblicke in allelspezifische PrĂ€gungsprofile zu erhalten, wĂ€ren umfangreiche DBS HNA-LĂ€ngsschnittstudien aller 50-100 human geprĂ€gten Gene in unterschiedlichen Gewebetypen und Differenzierungsstadien wĂŒnschenswert.  Intrauterine exposure to gestational diabetes mellitus (GDM) induces lifelong increased predisposition for metabolic and complex diseases in the exposed progeny. The elevated disease susceptibility is transmitted via epigenetic alterations that influence gene expression levels and phenotypes through regulation of gene activity. Genome-wide methylation profiles of fetal cord bloods (FCBs) were investigated in GDM and control pregnancies in order to identify new genes susceptible to fetal programming. After multiple testing correction, we found 65 significantly differentially methylated CpG sites between GDM and control groups (52 of which were gene associated) within the Illumina Infinium HumanMethylation 450K array data. Using pyrosequencing, we successfully confirmed the observed results in four of these candidate loci (ATP5A1, MFAP4, PRKCH, SLC17A4) and one gene from the literature (HIF3A). A multivariate regression model was adjusted for the confounding factors gestational age, fetal sex and maternal BMI. The GDM effect was stronger within the insulin treated subgroup (I-GDM) compared to the dietary subgroup (D GDM), suggesting that GDM is a multifactorial disease evidenced by changes of small effect size in multiple genes. Significant mean methylation differences were detected between the GDM group and controls in three out of four imprinted genes (MEG3 promoter, MEST and PEG3) that were analyzed with Deep Bisulfite Sequencing (DBS). The identified genes represent labile target regions for GDM-induced intrauterine programming and could represent future biomarkers for disease diagnosis and prognosis. Furthermore, DBS enables sequencing at a single allele resolution and the calculation of allele specific epimutation rates by differentiating the parental origin of alleles via an informative SNP within differentially methylated regions (DMRs). Epimutations were characterized as alleles showing > 50 % aberrantly (de)methylated CpG sites. DBS data were generated using two different sequencing platforms (Roche GS Junior and Illumina MiSeq). An independent library preparation protocol was established for Illumina MiSeq sequencing. The paternally expressed MEST promoter DMR and the maternally expressed MEG3 intergenic (IG) DMR showed high epimutation rates for the unmethylated alleles in FCB, as well as adult blood and visceral adipose tissue. On the contrary, only minor epimutation rates were displayed by the imprinted (methylated) alleles. Thus, hypermethylation of the non-imprinted allele (HNA) was independent of parental origin, as MEST and MEG3 are opposingly imprinted genes. Very low epimutation rates in sperm indicate that the HNA effect arises after fertilization. A weak but significant HNA was also found for the secondary MEG3 promoter DMR (which is known to be regulated by the MEG3 IG-DMR). The paternally expressed PEG3 promoter DMR showed no HNA and no difference in parental epimutation rates. The observed HNA effect (for the MEST DMR, the MEG3 IG-DMR and the MEG3 promoter DMR) was neither associated with GDM nor obesity and exhibited a large interindividual variance. Maintenance of differential methylation profiles in imprinting control regions (ICRs) seems to be of great importance during some developmental periods and is therefore strictly controlled in germ cells. Later on, it might become redundant manifested in the accumulation of stochastic as well as environmentally-induced errors on the non-imprinted allele. There is evidence that HNA-susceptible imprinted genes resemble metastable epialleles in many aspects. Therefore, we suggest that stochastic as well as environmental stimuli program imprinted gene networks that are important for growth related processes during early development using HNA. Further longitudinal studies of all 50 – 100 imprinted genes would benefit in a deeper insight in allele-specific imprinting patterns of various human tissues

    Epigenetic signatures of gestational diabetes mellitus on cord blood methylation

    No full text
    Background: Intrauterine exposure to gestational diabetes mellitus (GDM) confers a lifelong increased risk for metabolic and other complex disorders to the offspring. GDM-induced epigenetic modifications modulating gene regulation and persisting into later life are generally assumed to mediate these elevated disease susceptibilities. To identify candidate genes for fetal programming, we compared genome-wide methylation patterns of fetal cord bloods (FCBs) from GDM and control pregnancies. Methods and results: Using Illumina’s 450K methylation arrays and following correction for multiple testing, 65 CpG sites (52 associated with genes) displayed significant methylation differences between GDM and control samples. Four candidate genes, ATP5A1, MFAP4, PRKCH, and SLC17A4, from our methylation screen and one, HIF3A, from the literature were validated by bisulfite pyrosequencing. The effects remained significant after adjustment for the confounding factors maternal BMI, gestational week, and fetal sex in a multivariate regression model. In general, GDM effects on FCB methylation were more pronounced in women with insulin-dependent GDM who had a more severe metabolic phenotype than women with dietetically treated GDM. Conclusions: Our study supports an association between maternal GDM and the epigenetic status of the exposed offspring. Consistent with a multifactorial disease model, the observed FCB methylation changes are of small effect size but affect multiple genes/loci. The identified genes are primary candidates for transmitting GDM effects to the next generation. They also may provide useful biomarkers for the diagnosis, prognosis, and treatment of adverse prenatal exposures

    The link between cytogenetics/genomics and imaging patterns of relapse and progression in patients with relapsed/refractory multiple myeloma: a pilot study utilizing 18F-FDG PET/CT

    Get PDF
    Utilizing 18F-fluorodeoxyglucose (18F-FDG) positron emission tomography (PET)/computed tomography (CT), we performed this pilot study to evaluate the link between cytogenetic/genomic markers and imaging patterns in relapsed/refractory (RR) multiple myeloma (MM). We retrospectively analyzed data of 24 patients with RRMM who were treated at our institution between November 2018 and February 2020. At the last relapse/progression, patients had been treated with a median of three (range 1–10) lines of therapy. Six (25%) patients showed FDG avid extramedullary disease without adjacency to bone. We observed significantly higher maximum standardized uptake values (SUVmax) in patients harboring del(17p) compared with those without del(17p) (p = 0.025). Moreover, a high SUVmax of >15 indicated significantly shortened progression-free survival (PFS) (p = 0.01) and overall survival (OS) (p = 0.0002). One female patient exhibited biallelic TP53 alteration, i.e., deletion and mutation, in whom an extremely high SUVmax of 37.88 was observed. In summary, this pilot study suggested a link between del(17p)/TP53 alteration and high SUVmax on 18F-FDG PET/CT in RRMM patients. Further investigations are highly warranted at this point

    Single CpG hypermethylation, allele methylation errors, and decreased expression of multiple tumor suppressor genes in normal body cells of mutation‐negative early‐onset and high‐risk breast cancer patients

    No full text
    To evaluate the role of constitutive epigenetic changes in normal body cells of BRCA1/BRCA2‐mutation negative patients, we have developed a deep bisulfite sequencing assay targeting the promoter regions of 8 tumor suppressor (TS) genes (BRCA1, BRCA2, RAD51C, ATM, PTEN, TP53, MLH1, RB1) and the estrogene receptor gene (ESR1), which plays a role in tumor progression. We analyzed blood samples of two breast cancer (BC) cohorts with early onset (EO) and high risk (HR) for a heterozygous mutation, respectively, along with age‐matched controls. Methylation analysis of up to 50,000 individual DNA molecules per gene and sample allowed quantification of epimutations (alleles with >50% methylated CpGs), which are associated with epigenetic silencing. Compared to ESR1, which is representative for an average promoter, TS genes were characterized by a very low (< 1%) average methylation level and a very low mean epimutation rate (EMR; < 0.0001% to 0.1%). With exception of BRCA1, which showed an increased EMR in BC (0.31% vs. 0.06%), there was no significant difference between patients and controls. One of 36 HR BC patients exhibited a dramatically increased EMR (14.7%) in BRCA1, consistent with a disease‐causing epimutation. Approximately one third (15 of 44) EO BC patients exhibited increased rates of single CpG methylation errors in multiple TS genes. Both EO and HR BC patients exhibited global underexpression of blood TS genes. We propose that epigenetic abnormalities in normal body cells are indicative of disturbed mechanisms for maintaining low methylation and appropriate expression levels and may be associated with an increased BC risk

    Single CpG hypermethylation, allele methylation errors, and decreased expression of multiple tumor suppressor genes in normal body cells of mutation‐negative early‐onset and high‐risk breast cancer patients

    No full text
    To evaluate the role of constitutive epigenetic changes in normal body cells of BRCA1/BRCA2‐mutation negative patients, we have developed a deep bisulfite sequencing assay targeting the promoter regions of 8 tumor suppressor (TS) genes (BRCA1, BRCA2, RAD51C, ATM, PTEN, TP53, MLH1, RB1) and the estrogene receptor gene (ESR1), which plays a role in tumor progression. We analyzed blood samples of two breast cancer (BC) cohorts with early onset (EO) and high risk (HR) for a heterozygous mutation, respectively, along with age‐matched controls. Methylation analysis of up to 50,000 individual DNA molecules per gene and sample allowed quantification of epimutations (alleles with >50% methylated CpGs), which are associated with epigenetic silencing. Compared to ESR1, which is representative for an average promoter, TS genes were characterized by a very low (< 1%) average methylation level and a very low mean epimutation rate (EMR; < 0.0001% to 0.1%). With exception of BRCA1, which showed an increased EMR in BC (0.31% vs. 0.06%), there was no significant difference between patients and controls. One of 36 HR BC patients exhibited a dramatically increased EMR (14.7%) in BRCA1, consistent with a disease‐causing epimutation. Approximately one third (15 of 44) EO BC patients exhibited increased rates of single CpG methylation errors in multiple TS genes. Both EO and HR BC patients exhibited global underexpression of blood TS genes. We propose that epigenetic abnormalities in normal body cells are indicative of disturbed mechanisms for maintaining low methylation and appropriate expression levels and may be associated with an increased BC risk

    Additional file 2: Figure S1. of Epigenetic signatures of gestational diabetes mellitus on cord blood methylation

    No full text
    Estimation of blood cell composition based on 450K methylation array profiles. Blue box plots show the distribution of cell types in GDM cord blood and red box plots in control samples. The median is represented by a horizontal line. The bottom of the box indicates the 25th percentile and the top the 75th percentile. Outliers are shown as circles. (DOC 146 kb

    Additional file 4: Table S3. of Epigenetic signatures of gestational diabetes mellitus on cord blood methylation

    No full text
    Multivariate analyses (adjusting for maternal BMI, gestational age, and fetal sex): CpG methylation of candidate genes in GDM versus control FCB samples. (DOC 68 kb
    corecore