scholarly journals The impact of infertility diagnosis on embryo-endometrial dialogue

Reproduction ◽  
2018 ◽  
Vol 155 (6) ◽  
pp. 543-552 ◽  
Author(s):  
Jason C Parks ◽  
Blair R McCallie ◽  
Alyssa L Patton ◽  
Zain A Al-Safi ◽  
Alex J Polotsky ◽  
...  

Initial stages of implantation involve bi-directional molecular crosstalk between the blastocyst and endometrium. This study investigated an association between infertility etiologies, specifically advanced maternal age (AMA) and endometriosis, on the embryo-endometrial molecular dialogue prior to implantation. Co-culture experiments were performed with endometrial epithelial cells (EEC) and cryopreserved day 5 blastocysts (n = 41 ≥ Grade 3BB) donated from patients presenting with AMA or endometriosis, compared to fertile donor oocyte controls. Extracellular vesicles isolated from co-culture supernatant were analyzed for miRNA expression and revealed significant alterations correlating to AMA or endometriosis. Specifically, AMA resulted in 16 miRNAs with increased expression (P ≤ 0.05) and strong evidence for negative regulation toward 206 target genes. VEGFA, a known activator of cell adhesion, displayed decreased expression (P ≤ 0.05), validating negative regulation by 4 of these increased miRNAs: miR-126; 150; 29a; 29b (P ≤ 0.05). In endometriosis patients, a total of 10 significantly altered miRNAs displayed increased expression compared to controls (miR-7b; 9; 24; 34b; 106a; 191; 200b; 200c; 342-3p; 484) (P ≤ 0.05), targeting 1014 strong evidence-based genes. Three target genes of miR-106a (CDKN1A, E2F1 and RUNX1) were independently validated. Functional annotation analysis of miRNA-target genes revealed enriched pathways for both infertility etiologies, including disrupted cell cycle regulation and proliferation (P ≤ 0.05). These extracellular vesicle-bound secreted miRNAs are key transcriptional regulators in embryo-endometrial dialogue and may be prospective biomarkers of implantation success. One of the limitations of this study is that it was a stimulated, in vitro model and therefore may not accurately reflect the in-vivo environment.

2021 ◽  
Author(s):  
Rui Yang ◽  
Wenzhe Wang ◽  
Meichen Dong ◽  
Kristen Roso ◽  
Paula Greer ◽  
...  

Myc plays a central role in tumorigenesis by orchestrating the expression of genes essential to numerous cellular processes1-4. While it is well established that Myc functions by binding to its target genes to regulate their transcription5, the distribution of the transcriptional output across the human genome in Myc-amplified cancer cells, and the susceptibility of such transcriptional outputs to therapeutic interferences remain to be fully elucidated. Here, we analyze the distribution of transcriptional outputs in Myc-amplified medulloblastoma (MB) cells by profiling nascent total RNAs within a temporal context. This profiling reveals that a major portion of transcriptional action in these cells was directed at the genes fundamental to cellular infrastructure, including rRNAs and particularly those in the mitochondrial genome (mtDNA). Notably, even when Myc protein was depleted by as much as 80%, the impact on transcriptional outputs across the genome was limited, with notable reduction mostly only in genes involved in ribosomal biosynthesis, genes residing in mtDNA or encoding mitochondria-localized proteins, and those encoding histones. In contrast to the limited direct impact of Myc depletion, we found that the global transcriptional outputs were highly dependent on the activity of Inosine Monophosphate Dehydrogenases (IMPDHs), rate limiting enzymes for de novo guanine nucleotide synthesis and whose expression in tumor cells was positively correlated with Myc expression. Blockage of IMPDHs attenuated the global transcriptional outputs with a particularly strong inhibitory effect on infrastructure genes, which was accompanied by the abrogation of MB cells proliferation in vitro and in vivo. Together, our findings reveal a real time action of Myc as a transcriptional factor in tumor cells, provide new insight into the pathogenic mechanism underlying Myc-driven tumorigenesis, and support IMPDHs as a therapeutic vulnerability in cancer cells empowered by a high level of Myc oncoprotein.


2016 ◽  
Vol 36 (7) ◽  
pp. 1180-1193 ◽  
Author(s):  
Nathan L. Price ◽  
Brandon Holtrup ◽  
Stephanie L. Kwei ◽  
Martin Wabitsch ◽  
Matthew Rodeheffer ◽  
...  

White adipose tissue (WAT) is essential for maintaining metabolic function, especially during obesity. The intronic microRNAs miR-33a and miR-33b, located within the genes encoding sterol regulatory element-binding protein 2 (SREBP-2) and SREBP-1, respectively, are transcribed in concert with their host genes and function alongside them to regulate cholesterol, fatty acid, and glucose metabolism. SREBP-1 is highly expressed in mature WAT and plays a critical role in promotingin vitroadipocyte differentiation. It is unknown whether miR-33b is induced during or involved in adipogenesis. This is in part due to loss of miR-33b in rodents, precludingin vivoassessment of the impact of miR-33b using standard mouse models. This work demonstrates that miR-33b is highly induced upon differentiation of human preadipocytes, along withSREBP-1. We further report that miR-33b is an important regulator of adipogenesis, as inhibition of miR-33b enhanced lipid droplet accumulation. Conversely, overexpression of miR-33b impaired preadipocyte proliferation and reduced lipid droplet formation and the induction of peroxisome proliferator-activated receptor γ (PPARγ) target genes during differentiation. These effects may be mediated by targeting of HMGA2, cyclin-dependent kinase 6 (CDK6), and other predicted miR-33b targets. Together, these findings demonstrate a novel role of miR-33b in the regulation of adipocyte differentiation, with important implications for the development of obesity and metabolic disease.


Author(s):  
Ning Jiang ◽  
Yihao Liao ◽  
Miaomiao Wang ◽  
Youzhi Wang ◽  
Keke Wang ◽  
...  

Abstract Background The incidence of bladder urothelial carcinoma (UC), a common malignancy of the urinary tract, is approximately three times higher in men than in women. High expression of the mitotic kinase BUB1 is associated with the occurrence and development of several cancers, although the relationship between BUB1 and bladder tumorigenesis remains unclear. Methods Using a microarray approach, we found increased BUB1 expression in human BCa. The association between BUB1 and STAT3 phosphorylation was determined through molecular and cell biological methods. We evaluated the impact of pharmacologic inhibition of BUB1 kinase activity on proliferation and BCa progression in vitro and in vivo. Results In this study, we found that BUB1 expression was increased in human bladder cancer (BCa). We further identified through a series of molecular and cell biological approaches that BUB1 interacted directly with STAT3 and mediated the phosphorylation of STAT3 at Ser727. In addition, the findings that pharmacologic inhibition of BUB1 kinase activity significantly suppressed BCa cell proliferation and the progression of bladder cancer in vitro and in vivo were further verified. Finally, we found that the BUB1/STAT3 complex promoted the transcription of STAT3 target genes and that depletion of BUB1 and mutation of the BUB1 kinase domain abrogated this transcriptional activity, further highlighting the critical role of kinase activity in the activation of STAT3 target genes. A pharmacological inhibitor of BUB1 (2OH-BNPP1) was able to significantly inhibit the growth of BCa cell xenografts. Conclusion This study showed that the BUB1 kinase drives the progression and proliferation of BCa by regulating the transcriptional activation of STAT3 signaling and may be an attractive candidate for therapeutic targeting in BCa.


2021 ◽  
Vol 23 (Supplement_1) ◽  
pp. i5-i5
Author(s):  
Rui Yang ◽  
Wenzhe Wang ◽  
Meichen Dong ◽  
Kristen Roso ◽  
Xuhui Bao ◽  
...  

Abstract Myc plays a central role in tumorigenesis by orchestrating the expression of genes essential to numerous cellular processes. While it is well established that Myc functions by binding to its target genes to regulate their transcription, the distribution of the transcriptional output across human genome in Myc-amplified cancer cells, and the susceptibility of such transcriptional outputs to therapeutic interferences remain to be fully elucidated. Here, we analyze the distribution of transcriptional outputs in Myc-amplified medulloblastoma (MB) cells by profiling nascent total RNAs within a temporal context. This profiling reveals a major portion of transcriptional action in these cells was directed at the genes fundamental to cellular infrastructures, including rRNAs and particularly those in the mitochondrial genome (mtDNA). Notably, even when Myc protein was depleted by as much as 80%, the impact on transcriptional outputs across the genome was limited, with notable reduction mostly in genes of involved in ribosomal biosynthesis, genes residing in mtDNA or encoding mitochondria-localized proteins, and those encoding histones. In contrast to the limited direct impact of Myc depletion, we found that the global transcriptional outputs were highly dependent on the activity of Inosine Monophosphate Dehydrogenases (IMPDHs), rate limiting enzymes for de novo guanine nucleotide synthesis and whose expression in tumor cells was positively correlated with Myc’s expression. Blockage of IMPDHs attenuated the global transcriptional outputs with a particularly strong inhibitory effect on the aforementioned infrastructure genes, which was accompanied by the abrogation of MB cell’s proliferation in vitro and in vivo. Together, our findings reveal a real time action of Myc as a transcriptional factor in tumor cells, gain new insight into the pathogenic mechanism underlying Myc-driven tumorigenesis, and support IMPDHs as a therapeutic vulnerability in MB cells empowered by a high level of Myc oncoprotein.


2019 ◽  
Vol 10 (12) ◽  
Author(s):  
Kai Zhao ◽  
Xiaoteng Cui ◽  
Qixue Wang ◽  
Chuan Fang ◽  
Yanli Tan ◽  
...  

AbstractRunt-Related Transcription Factor 1 (RUNX1) is highly expressed in the Mesenchymal (Mes) subtype of glioblastoma (GBM). However, the specific molecular mechanism of RUNX1 in Mes GBM remains largely elusive. In this study, cell and tumor tissue typing were performed by RNA-sequencing. Co-immunoprecipitation (co-IP) and immunofluorescence (IF) were employed to identify members of the RUNX1 transcriptional protein complex. Bioinformatics analysis, chromatin immunoprecipitation (ChIP), and luciferase reporter experiments were utilized to verify target genes. Analyses of The Cancer Genome Atlas (TCGA) and Chinese Glioma Genome Atlas (CGGA) verified the expression levels and prognoses associated with RUNX1/p-SMAD3/SUV39H1 target genes. In vivo patient-derived xenograft (PDX) studies and in vitro functional studies verified the impact of RUNX1 on the occurrence and development of GBM. The results showed that RUNX1 was upregulated in Mes GBM cell lines, tissues and patients and promoted proliferation and invasion in GBM in a TGFβ pathway-dependent manner in vivo and in vitro. We found and verified that BCL3 and MGP are transcriptionally activated by p-SMAD3 /RUNX1, while MXI1 is transcriptionally suppressed by the RUNX1/SUV39H1-H3K9me3 axis. This finding offers a theoretical rationale for using molecular markers and choosing therapeutic targets for the Mes type of GBM.


Nutrients ◽  
2021 ◽  
Vol 13 (9) ◽  
pp. 3000
Author(s):  
Marta Cykowiak ◽  
Violetta Krajka-Kuźniak ◽  
Robert Kleszcz ◽  
Małgorzata Kucińska ◽  
Hanna Szaefer ◽  
...  

Background: Increasing evidence suggests that combinations of phytochemicals are more efficient than single components in the modulation of signaling pathways involved in cancer development. In this study, the impact of phenethyl isothiocyanate (PEITC), indole-3-carbinol (I3C), xanthohumol, (X), and resveratrol (RES) and their combinations on the activation and expression of Nrf2 and NF-κB in human hepatocytes and HCC cells were evaluated. Methods: THLE-2 and HepG2 cells were exposed to single phytochemicals and their combinations for 24 h. The activation of Nrf2 and NF-κB, expression of their target genes, and effect on cells survival were assessed. The tumor burden was evaluated in mice carrying xenografts. Results: All phytochemicals enhanced the activation and expression of Nrf2 and its target genes SOD and NQO1 in HepG2 cells. The increased expression of NQO1 (~90%) was associated with increased ROS generation. X + PEITC downregulated NF-κB activation reducing binding of its active subunits to DNA resulting in diminished COX-2 expression. In contrast to single phytochemicals, X + PEITC induced apoptosis. Moderate reduction of tumor burden in mice carrying xenografts following X and PEITC or their combination was observed. Conclusions: Since Nrf2 is overexpressed in HCC its reduced activation together with diminished level of NF-κB by X + PEITC may be considered as a strategy to support conventional HCC therapy.


2021 ◽  
Author(s):  
Lichun Yang ◽  
Xiaorui Liu ◽  
Lei Zhang ◽  
Danni Li ◽  
Guili Li ◽  
...  

Abstract Background: Endometrial epithelial cells proliferation and secretion of various cytokines have a strong impact on the formation of receptive endometrium, which is known as a physiological status that allows an activated embryo to attach to the endometrium for a limited time. Circular RNAs and miRNAs can be involved in the dynamic physiological changes of endometrium by regulating relevant functional target genes in the uterus. Our work presented here with the ultimate purpose of revealing the latent molecular mechanism of FBXO18/circRNA211/miR-431/CSF1 axis in the establishment of endometrial receptivity of dairy goats.Results: In vitro, we found a regulatory network of FBXO18/circRNA211/miR-431/CSF1 in goat endometrial epithelial cells that circRNA211 severed as a sponge for miR-431, resulting in weakening the inhibition of miR-431 on target genes CSF1 and FBXO18. FBXO18/circRNA211/miR-431/CSF1 axis promoted the proliferation through regulating the key proteins of Ras, Raf, MEK, ERK in MAPK pathway via CCK-8, EdU, flow cytometry and Western blot assays. Furthermore, FBXO18/circRNA211/miR-431/CSF1 axis activated the phosphorylation of key proteins PI3K, AKT and mTOR in PI3K-mTOR pathway by CSF1R, thereby promoting the establishment of endometrial receptivity. In vivo models, mice injected with miR-431 agomir showed that the endometrial thickness and the number of pinopodes were significantly decreased by HE staining and scanning electron microscope. Immunohistochemistry results showed that VEGF and OPN proteins were down-regulated and MUC1 protein was up-regulated under the treatment of miR-431 agomir. Further study demonstrated that miR-431 inhibited embryo implantation by impeding the establishment of endometrial receptivity.Conclusion: Ultimately, our study revealed a regulatory mechanism of FBXO18/circRNA211/miR-431/CSF1 axis in goat endometrial epithelial cells. This circRNA/miRNA/mRNA regulatory network presented here in vitro and in vivo models may provide a novel insight into the potentially regulating endometrium biological functions and promoting the formation of endometrium receptivity.


Blood ◽  
2012 ◽  
Vol 120 (21) ◽  
pp. 3439-3439
Author(s):  
Amy E Campbell ◽  
Lorna Wilkinson-White ◽  
Joel P Mackay ◽  
Jacqueline M Matthews ◽  
Gerd A Blobel

Abstract Abstract 3439 Missense mutations in the gene encoding hematopoietic transcription factor GATA1 cause congenital anemias and/or thrombocytopenias. All seven reported mutations give rise to amino acid substitutions within the amino-terminal zinc finger (NF), but produce a range of phenotypes. The clinical severity depends on the site and type of substitution, and different substitutions of the same residue can produce disparate phenotypes. We combined structural, biochemical, in vivo conditional rescue approaches, and genomic analyses to systematically characterize all known GATA1 mutations with the goal of determining how they alter GATA1 function to result in disease. Introducing mutant forms of GATA1 into GATA1-null erythroid or bipotential erythromegakaryocytic cell lines essentially recapitulated patient phenotypes. The V205M, G208S, G208R, and D218Y mutations severely impaired both erythroid and megakaryocyte maturation, while the R216Q, R216W, and D218G mutations had only a mild effect on the maturation of these lineages. Global differentiation defects were reproduced at the level of individual GATA1 target genes. Moreover, the former mutants greatly impaired both the transcriptional activation and repression functions of GATA1, while the latter moderately impaired gene activation but had no effect on repression. It had been suggested previously that GATA1 mutations could be categorized into two classes, those that impair binding of the NF to the essential GATA1 cofactor FOG1 (V205M, G208S, G208R) and those that diminish binding of the NF to DNA (R216Q and R216W). The impact of the final two mutations (D218G and D218Y) remained uncertain, as this residue is not part of any known interaction face. Our work led to the following novel conclusions: Binding studies using isothermal titration calorimetry (ITC) and chromatin immunoprecipitation (ChIP) produced concurrent results showing that the V205M, G208S, G208R, and D218Y mutations diminish the GATA1-FOG1 interaction in vitro and FOG1 recruitment to GATA1 target genes in vivo. Interestingly, in contrast to D218Y, D218G did not affect FOG1 binding in vitro or in vivo. Furthermore, G208S had a less pronounced impact on FOG1 binding than the other three mutations, thus correlating the severity of the clinical presentation with the degree of FOG1 disruption. This confirms and extends previous work linking impaired FOG1 binding to the disease phenotypes associated with this class of mutations.ITC showed that R216Q and R216W disrupt DNA binding in vitro, consistent with previous in vitro studies. However, remarkably, ChIP assays revealed that neither mutation impaired in vivo GATA1 target site occupancy at any examined simple or palindromic GATA elements, suggesting that failure to bind DNA does not account for the associated clinical phenotypes.Notably, the R216Q and D218G mutations selectively diminished recruitment of Tal1/SCL without affecting the interaction with FOG1 or DNA. This implicates for the first time the Tal1/SCL complex in the pathogenesis of disorders caused by GATA1 mutations. Since the Tal1/SCL complex functions mostly during GATA1 gene activation, this also explains the observation that these GATA1 mutants largely retain their ability to repress transcription. Moreover, changes in the gene expression profiles of R216Q and D218G expressing cells are highly correlated with each other but clearly distinct from the gene expression changes associated with different substitutions at the same residues (R216W or D218Y), revealing a specific subset of genes that are most sensitive to disruption of the GATA1-Tal1/SCL interaction.An unexpected finding from our studies is that different substitutions of the same residue can disrupt binding to distinct cofactors (e.g. D218G impairs Tal1/SCL binding while D218Y impairs FOG1 binding), thus accounting for variable disease presentation. In concert, our work on GATA1 mutations in their native environment reveals critical new insights not obtainable from in vitro studies. This highlights the usefulness of gene complementation studies in the relevant lineages for the dissection of transcription pathways to better understand and ultimately diagnose and treat hematologic disease. Disclosures: No relevant conflicts of interest to declare.


Blood ◽  
2009 ◽  
Vol 114 (22) ◽  
pp. 732-732
Author(s):  
Erin K Hertlein ◽  
Thomas S Lin ◽  
Wagner J Amy ◽  
Towns H William ◽  
Virginia M Goettl ◽  
...  

Abstract Abstract 732 One attractive therapeutic target currently being explored in CLL is HSP90, a chaperone which stabilizes various client proteins (AKT, Raf, ZAP-70) which are important for survival of CLL cells. Interfering with HSP90 protein binding to these client proteins leads to their rapid degradation. Our group and others have demonstrated that 17-allylamino-17-demethoxy-geldanamycin (17-AAG) depletes only select chaperone proteins and promotes modest cytotoxicity in CLL patient cells. 17-dimethylaminoethylamino-17-demethoxy-geldanamycin (17-DMAG) is a novel HSP90 inhibitor with improved solubility, bioavailability and cytotoxicity in cancer cell lines as compared to 17-AAG. We demonstrate that 17-DMAG more potently induces caspase-dependent apoptosis of primary CLL cells compared to 17-AAG. This enhanced cytotoxicity is tumor cell selective as it spares normal T-cells and NK-cells, and only modestly affects normal B cells. In addition to the broad range of Hsp90 client proteins which are regulated by 17-AAG (AKT, CDK9, ZAP-70), we found that 17-DMAG was also able to deplete both IKKαa and IKKβ, the activating kinases of the NF-κB family of transcription factors. The impact of 17-DMAG on both IKKαa and β is very relevant, as there have been several studies investigating the effect of IKK inhibitors on in vitro apoptosis in CLL, however there is still no clear therapeutic option for targeting these kinases in the clinic. Most IKK inhibitors including 17-AAG are specific for the IKKβ subunit that mediates classical NF-κB signaling, leaving IKKαa and the alternative NF-κB pathway intact. This is particularly important given recent evidence by Lam et al. (Clin Can Res 2005 Jan 1;11(1):28-40) demonstrating that IKKαa is able to compensate for the loss of IKKβ in DLBCL cells lines. Therefore the ability of 17-DMAG to target both subunits of the IKK complex potentially makes it a very potent and effective NF-κB inhibitor in CLL. To validate the downstream significance of this NF-κB regulation, we show that 17-DMAG effectively reduces NF-κB nuclear localization DNA binding in CLL patient cells resulting in decreased transcript and protein levels of NF-κB targets MCL1 and BCL2, known to be major factors in CLL cell survival and drug resistance. The decrease in MCL1 and BCL2 following 17-DMAG treatment was not prevented by treatment with the caspase inhibitor zVAD-fmk even though viability was rescued, suggesting that the decrease in these survival proteins precedes cell death and is not simply a consequence of the apoptotic process. Finally, we determined the in vivo significance of 17-DMAG treatment using a TCL1-SCID transplant model. We found that NF-κB targets genes (A20, BCL2, cIAP, MCL1 and XIAP) were decreased in vivo following treatment with 17-DMAG. In addition to typical genes which are activated by NF-κB, we also determined the in vivo effect of 17-DMAG on NF-κB mediated transcriptional repression. Our group has recently determined that the transcription factor FOXD3 is transcriptionally silenced by an NF-κB repressor complex very early in the disease progression of the TCL1 mouse model, an event which facilitates subsequent global DNA methylation and gene silencing in CLL. We found that treatment with 17-DMAG leads to re-expression of FOXD3 suggesting that 17-DMAG is an effective therapeutic tool to target NF-κB mediated gene repression as well as NF-κB mediated expression of survival genes. Furthermore, 17-DMAG treatment significantly prolonged the survival of these mice (75 days vs. 66 days, p=0.027, n=10/group). Together, our data demonstrate that the HSP90 inhibitor 17-DMAG represents a novel multi-subunit IKK inhibitor leading to a decrease in anti-apoptosis genes relevant to CLL survival while increasing the expression of genes silenced during CLL disease progression. Given its oral formulation, which allows administration of 17-DMAG by continuous dosing and uninterrupted inhibition of HSP90, initiation of phase II clinical trials in CLL that include detailed pharmacodynamic studies monitoring NF-κB target genes are indicated. Disclosures: No relevant conflicts of interest to declare.


2013 ◽  
Vol 150 (3) ◽  
pp. 1024-1031 ◽  
Author(s):  
Mohammad Hossein Boskabady ◽  
Sakine Shahmohammadi Mehrjardi ◽  
Abadorrahim Rezaee ◽  
Houshang Rafatpanah ◽  
Sediqeh Jalali

Sign in / Sign up

Export Citation Format

Share Document