SwePub
Sök i SwePub databas

  Utökad sökning

Träfflista för sökning "WFRF:(Hases Linnea) ;conttype:(scientificother)"

Sökning: WFRF:(Hases Linnea) > Övrigt vetenskapligt/konstnärligt

  • Resultat 1-5 av 5
Sortera/gruppera träfflistan
   
NumreringReferensOmslagsbildHitta
1.
  • Birgersson, Madeleine, et al. (författare)
  • Intestinal estrogen receptor beta modulates the tumor immune microenvironment in a mouse model of colitis-associated cancer
  • Annan publikation (övrigt vetenskapligt/konstnärligt)abstract
    • Chronic inflammation promotes the development of colorectal cancer (CRC), as evidenced by patients with inflammatory bowel disease (IBD), and sex disparities are evident in CRC. The tumor microenvironment (TME) is composed of stromal cells and infiltrating immune cells that directly affect processes including antitumor immunity. We have previously shown that intestinal estrogen receptor beta (ERβ) protects against colitis and colitis-induced cancer (CAC) by modulating inflammatory signaling and that males are more sensitive to the induction of colitis and cancer. However, sex differences between tumors and the impact of ERβ the tumor immune microenvironment have not been investigated. In this study, we have analyzed colon samples from AOM/DSS-treated wild-type and ERβKOVil mice (that lack intestinal ERβ) and profiled the differences in the transcriptome and immune response to CAC on the basis of sex and ERβ expression. RNA-sequencing revealed differences in gene expression and enriched biological processes depending on sex and genotype, and the immune response to cancer appears altered between tumors from female WT and ERβKOVil mice. Immunostaining subsequently showed that tumors from ERβKOVil mice display significantly increased CD68+ macrophage infiltration, decreased CD3+ T cell infiltration, and, strikingly, impaired NK cell infiltration. Here, for the first time, we show that intestinal ERβ modulates the tumor immune microenvironment during CAC and that lack of intestinal ERβ appears to promote the formation of an immunosuppressive TME. Our findings indicate that activation of ERβ could be used to treat CRC, possibly together with immunotherapies, and provide a foundation for future studies investigating ERβ and immunity. 
  •  
2.
  • Hases, Linnea, et al. (författare)
  • ERβ and Inflammation
  • 2022
  • Ingår i: Advances in Experimental Medicine and Biology. - Cham : Springer Nature. ; 1390, s. 213-225
  • Bokkapitel (övrigt vetenskapligt/konstnärligt)abstract
    • Estrogen, through the regulation of cytokine production, can act both as pro-inflammatory and anti-inflammatory signals dependent on the tissue context. In breast cancer cells, ERα is known to modulate inflammatory signaling through interaction with NFκB. Whether ERβ has a role in inflammation is less explored. Low levels of ERβ have been corroborated in several immune-related organs and, for example, in colonic epithelial cells. Specifically, an impact of ERβ on colitis and colitis-associated colorectal cancer (CRC) is experimentally supported, using ERβ-selective agonists, full-body ERβ knockout mice and, most recently, intestinal epithelial-specific knockout mice. An intricate crosstalk between ERβ and TNFα/NFκB signaling in the colon is supported, and ERβ activation appears to reduce macrophage infiltration also during high fat diet (HFD)-induced colon inflammation. Finally, the gut microbiota plays a fundamental role in the pathogenesis of colitis and ERβ has been indicated to modulate the microbiota diversity during colitis and colitis-induced CRC. ERβ is thus proposed to protect against colitis, by modulating NFκB signaling, immune cell infiltration, and/or microbiota composition. Selective activation of ERβ may therefore constitute a suitable preventative approach for the treatment of for example colitis-associated CRC. 
  •  
3.
  • Hases, Linnea (författare)
  • Estrogen signaling in colon inflammation and colorectal cancer
  • 2020
  • Doktorsavhandling (övrigt vetenskapligt/konstnärligt)abstract
    • Colorectal cancer (CRC) is the third most deadly form of cancer in the Western world. Although screening efforts have reduced the overall mortality, the incidence is increasing among young adults. The frequency of inflammatory bowel disease (IBD) and obesity are increasing in parallel, which suggest a common underlying environmental link between the conditions. This increase is thought to correlate to an increased intake of high fat diets, and obesity is a major risk factor for CRC. Chronic inflammation, which is a hallmark for CRC promotion, is a well-known underlying factor in both obesity and IBD. The gut microbiota is another hallmark, and an impaired relationship between the host and gut microbes can contribute to obesity, IBD and CRC. The risk-benefit balance of current CRC-preventative treatments is poor, and there is a need for safer and better preventatives in order to reduce the CRC mortality. Both obesity and IBD place men at a significant higher risk of CRC compared to women. This indicates a protective role for estrogen. The use of full Estrogen receptor (ER) β knockout mice has demonstrated ERβ protective effects against experimentally induced CRC. However, it is unknown through which cells these protective effects are mediated. There are only low mRNA levels of ERβ in the colon, unclear if adequate for a functional role, and ERβ may also be expressed in intestinal immune cells. Understanding the CRC-preventative effects of intestinal epithelial ERβ in both sexes is important and may provide the background for a novel CRC chemopreventive approach.The overall aim of the thesis is the functional characterization of intestinal epithelial ERβ during colon inflammation and colitis-induced CRC and identification of potential sex differences, which can ultimately provide novel opportunities for chemopreventive exploitation (Figure 1). In paper I we utilized intestinal epithelial ERβ knockout mice (ERβKOVil) of both sexes and induced colitis and colitis associated CRC (CA-CRC). We found that intestinal epithelial ERβ is protective against colitis and CA-CRC in both sexes, but in a sex-dependent manner. The underlying mechanism includes an intricate crosstalk with TNFα-induced NFκB signaling.In paper II we identify that both sex and intestinal epithelial ERβ impact the microbiota composition. This may contribute to the exacerbated colitis and colitis-induced tumor formation observed in ERβKOVil mice.In paper III we induced colon inflammation by feeding the mice a high-fat diet (HFD, 60%) for 13 weeks and explored treatment with estrogen receptor-selective ligands. We identified that estrogen signaling, via ERβ, modulated the HFD-induced changes in the colon microenvironment. This included sex-dependent effects on epithelial cell proliferation, macrophage infiltration, and regulation of core circadian clock gene expression.In paper IV we utilized paired-normal and CRC clinical samples and identified sex differences in the transcriptome of both normal colon and CRC. By applying data-driven feature selection and machine learning on sex-separated TCGA data, we proposed sex-specific diagnostic biomarkers and prognostic biomarkers using survival analysis. In summary, this thesis characterizes intestinal epithelial ERβ as a novel chemopreventative target for CA-CRC in both sexes, and identifies related biological pathways, including crosstalk with nuclear factor κB (NFκB) signaling and modulation of circadian clock genes. ERβ activity in intestinal epithelial cells is manifested by altered microbiota composition, cell proliferation and immune cell infiltration. The identification of several significant sex differences provides evidence for the need to take sex into account in colitis and CRC research to improve health interventions.
  •  
4.
  •  
5.
  • Hases, Linnea, et al. (författare)
  • The importance of sex in colorectal cancer biomarker discovery
  • Annan publikation (övrigt vetenskapligt/konstnärligt)abstract
    • Colorectal cancer (CRC) is the third leading cause for cancer deaths, indicating the needfor new diagnostic and prognostic biomarkers. The advances in omics technologies andbioinformatics can speed up and improve current biomarker discovery strategies.Machine learning has been integrated for analysis of transcriptomic data for severalcancers. However, in addition to improved data-analysis, there is a need to investigate theimpact of sex in the biomarker discovery since there are several sex-differences in theincidence, mortality, prognosis and tumor characteristics of CRC. First we investigated ifthere are any sex-differences in the transcriptome of normal colon and CRC andinvestigated if there are any sex-differences in the differentially expressed genes betweenpaired-normal and CRC. In an attempt to study sex-specific biomarkers we used TCGAdata and performed feature selection with Vita, Boruta and MRMR in combination withmachine learning to identify top CRC biomarkers. Interestingly, we found stronger sexdifferencesin the normal colon compared to in CRC. Although the sex-differences werestronger in normal colon, sex showed to have a significant impact of the prognostic valueof the biomarkers. 13 of the selected features showed a sex-specific prognostic value. Thepreviously proposed prognostic biomarkers ESM1 and GUCA2A showed a male-specificprognostic value whereas CLDN1 was specific for females. Additionally, we found somenovel prognostic biomarkers including TSPAN7 (females), SLC25A23 (females) andC2orf88 (males). In conclusion, our data show the importance of sex in the discovery ofCRC biomarkers and proposes 13 sex-specific CRC prognostic biomarkers.
  •  
Skapa referenser, mejla, bekava och länka
  • Resultat 1-5 av 5

Kungliga biblioteket hanterar dina personuppgifter i enlighet med EU:s dataskyddsförordning (2018), GDPR. Läs mer om hur det funkar här.
Så här hanterar KB dina uppgifter vid användning av denna tjänst.

 
pil uppåt Stäng

Kopiera och spara länken för att återkomma till aktuell vy