SwePub
Sök i SwePub databas

  Extended search

Träfflista för sökning "WFRF:(Bellamkonda Kishan) "

Search: WFRF:(Bellamkonda Kishan)

  • Result 1-7 of 7
Sort/group result
   
EnumerationReferenceCoverFind
1.
  • Bellamkonda, Kishan, et al. (author)
  • Montelukast, a CysLT1 receptor antagonist, reduces colon cancer stemness and tumor burden in a mouse xenograft model of human colon cancer
  • 2018
  • In: Cancer Letters. - : Elsevier BV. - 0304-3835. ; 437, s. 13-24
  • Journal article (peer-reviewed)abstract
    • Inflammation is implicated in the etiology of sporadic colon cancer (CC), which is one of the leading causes of cancer-related deaths worldwide. Here, we report that inhibition of the inflammatory receptor CysLT1 through its antagonist, montelukast, is beneficial in minimizing stemness in CC and thereby minimizing tumor growth in a mouse xenograft model of human colon cancer. Upon treatment with montelukast, colonospheres derived from HT-29 and SW-480 human colon cancer cells exhibited a significant phenotypic change coupled with the downregulation of mRNA and protein expression of cancer stem cell (CSC) markers ALDH1 and DCLK1. Moreover, montelukast reduced the size of HT-29 cell-derived tumors in mice. The reduction in tumor size was associated with decreased levels of ALDH1A1, DCLK1, BCL2 mRNA and macrophage infiltration into the tumor tissue. Interestingly, this treatment elevated levels of the tumor suppressor 15-PGDH while reducing COX-2 expression. Our data highlight the association of CysLT1R with CSCs and demonstrate that inhibition of CysLT1R could prove beneficial in minimizing CSC-induced tumor growth. This work advances the notion that targeting CSCs is a promising approach to improve outcomes in those afflicted with colon cancer.
  •  
2.
  • Bellamkonda, Kishan, et al. (author)
  • The eicosanoids leukotriene D4 and prostaglandin E2 promote the tumorigenicity of colon cancer-initiating cells in a xenograft mouse model
  • 2016
  • In: BMC Cancer. - : Springer Science and Business Media LLC. - 1471-2407. ; 16:1
  • Journal article (peer-reviewed)abstract
    • Background: Colorectal cancer is one of the most common types of cancers worldwide. Recent studies have identified cancer-initiating cells (CICs) as a subgroup of replication-competent cells in the development of colorectal cancer. Although it is understood that an inflammation-rich tumor microenvironment presumably supports CIC functions, the contributory factors are not very well defined. The present study advances our understanding of the role of the eicosanoids leukotriene D4 (LTD4) and prostaglandin E2 (PGE2) in the tumorigenic ability of CICs and investigates the consequential changes occurring in the tumor environment that might support tumor growth. Methods: In this study we used human HCT-116 colon cancer ALDH+ cells in a nude mouse xenograft model. Protein expression and immune cell was determined in tumor-dispersed cells by flow cytometry and in tumor sections by immunohistochemistry. mRNA expressions were quantified using RT-q-PCR and plasma cytokine levels by Multiplex ELISA. Results: We observed that LTD4 and PGE2 treatment augmented CIC-induced tumor growth. LTD4-and PGE2-treated xenograft tumors revealed a robust increase in ALDH and Dclk1 protein expression, coupled with activated β-catenin signaling and COX-2 up-regulation. Furthermore, LTD4 or PGE2 accentuated the accumulation of CD45 expressing cells within xenograft tumors. Further analysis revealed that these infiltrating immune cells consisted of neutrophils (LY6G) and M2 type macrophages (CD206+). In addition, LTD4 and PGE2 treatment significantly elevated the plasma levels of cysteinyl leukotrienes and PGE2, as well as levels of IL-1β, IL-2, IL-6, TNF-α and CXCL1/KC/GRO. In addition, increased mRNA expression of IL-1β, IL-6 and IL-10 were detected in tumors from mice that had been treated with LTD4 or PGE2. Conclusion: Our data suggest that both LTD4 and PGE2 promote CICs in initiating tumor growth by allowing modifications in the tumor environment. Our data indicate that new therapeutic strategies targeting eicosanoids, specifically LTD4 and PGE2, could be tested for better therapeutic management of colon cancer.
  •  
3.
  • Bellamkonda, Kishan, et al. (author)
  • The impact of inflammatory lipid mediators on colon cancer-initiating cells.
  • 2015
  • In: Molecular Carcinogenesis. - : Wiley. - 1098-2744 .- 0899-1987. ; 54:11, s. 1315-1327
  • Journal article (peer-reviewed)abstract
    • The role of inflammatory lipid-mediators in tumor progression is well recognized in colorectal cancer; however, if this includes promotion of cancer-initiating cells remains unclear. We show that the inflammatory lipid-mediators leukotriene D4 and prostaglandin E2 increased the Aldehyde dehydrogenase (ALDH(+) ) population, the colony formation capacity, and tumor growth in a xenograft model of colon cancer. The ALDH(+) cells showed significant resistance to irradiation and 5-fluorouracil treatment that could be further augmented by these lipid-mediators, occurring in parallel with increased target gene expression. Our data emphasize a role for tumor microenvironment derived inflammatory lipid-mediators to favor cancer stem cells-like characteristics and thus promote tumor progression. © 2014 Wiley Periodicals, Inc.
  •  
4.
  • Bellamkonda, Kishan (author)
  • The Role of Inflammatory Lipid Mediators on Colon Cancer Initiating Cells (CICs)
  • 2015
  • Doctoral thesis (other academic/artistic)abstract
    • Colorectal cancer (CRC) is one of the major causes of cancer globally. Recent studies proposed a role for cancer initiating cells (CICs), a small subset of replication-competent cells, in colon carcinogenesis. Although the role of inflammatory lipid-mediators in CRC progression is well known, their role in the promotion of cancer-initiating cells remains to be elucidated. For this thesis, we investigated the role of eicosanoids – leukotriene D4 (LTD4) or prostaglandin E2 (PGE2) – on CIC properties and changes occurring in the tumor environment that could possibly support CIC-induced tumor growth. To this end, we identified the CICs on the basis of ALDH expression and evaluated their in vitro characteristics like colony formation, radio or chemoresistance and in vivo tumorigenic properties in the presence of LTD4 or PGE2. We showed that LTD4 and PGE2 enriched the ALDH+ cell population and augmented the colonies formation and tumor progression in xenograft mice model. The ALDH+ cells were also resistant to 5-fluorouracil and radiation that is additionally augmented by both the lipid-mediators. Moreover the impact of lipid inflammatory mediators on the stemness properties of CICs was evident by increased expression of genes that confer survival and self-renewal ability to CICs. In immunodeficient mice, LTD4 or PGE2 treatment amplified CIC-induced tumor growth. Furthermore, LTD4 and PGE2 increased cell proliferation activated β-catenin signaling and up-regulated COX-2. Additionally, LTD4 or PGE2 drive massive inflammatory responses identified as CD45+ enrichment, particularly of macrophages within tumors. The ability of ALDH+ cells to form tumors in immunodeficient mice could not be challenged by radiation therapy. In a separate series of experiments, we investigated the contribution of CICs in the development of sensitivity against montelukast, a CysLT1R antagonist. In this context we report that sensitivity of tumors against montelukast could depend on the variation in CICs content, activation of prosurvival factors such as BCL-2 and β-catenin signaling. Collectively, our data showed that LTD4 and PGE2 exacerbate CIC characteristics and promote tumor growth by allowing modifications in the tumor environment. New therapeutic strategies could aim to resolve not only cancer associated inflammation, but also to target CICs in order to achieve better remission and cure advanced colon cancer stages.
  •  
5.
  • Osman, Janina, et al. (author)
  • Cysteinyl leukotriene receptor 1 facilitates tumorigenesis in a mouse model of colitis-associated colon cancer
  • 2017
  • In: Oncotarget. - : Impact Journals, LLC. - 1949-2553. ; 8:21, s. 34773-34786
  • Journal article (peer-reviewed)abstract
    • Cysteinyl leukotriene receptor 1 (CysLT1R) has been shown to be up-regulated in the adenocarcinomas of colorectal cancer patients, which is associated with a poor prognosis. In a spontaneous model of colon cancer, CysLT1R disruption was associated with a reduced tumor burden in double-mutant female mice (ApcMin/+/Cysltr1-/-) compared to ApcMin/+ littermates. In the current study, we utilized a genetic approach to investigate the effect of CysLT1R in the induced azoxymethane/dextran sulfate sodium (AOM/DSS) model of colitis-associated colon cancer. We found that AOM/DSS female mice with a global disruption of the Cysltr1 gene (Cysltr1-/-) had a higher relative body weight, a more normal weight/length colon ratio and smaller-sized colonic polyps compared to AOM/DSS wild-type counterparts. The Cysltr1-/- colonic polyps exhibited low-grade dysplasia, while wild-type polyps had an adenoma-like phenotype. The Cysltr1-/- colonic polyps exhibited significant decreases in nuclear β-catenin and COX-2 protein expression, while the normal crypts surrounding the polyps exhibited increased Mucin 2 expression. Furthermore, Cysltr1-/- mice exhibited an overall reduction in inflammation, with a significant decrease in proinflammatory cytokines, polyp 5-LOX expression and infiltration of CD45 leukocytes and F4/80 macrophages. In conclusion, the present genetic approach in an AOM/DSS model further supports an important role for CysLT1R in colon tumorigenesis.
  •  
6.
  • Osman, Janina, et al. (author)
  • The WNT5A Agonist Foxy5 Reduces the Number of Colonic Cancer Stem Cells in a Xenograft Mouse Model of Human Colonic Cancer
  • 2019
  • In: Anticancer research. - : Anticancer Research USA Inc.. - 1791-7530 .- 0250-7005. ; 39:4, s. 1719-1728
  • Journal article (peer-reviewed)abstract
    • BACKGROUND: The wingless-type mammary tumour virus integration site 5A (WNT5A) agonist Foxy5 was shown in vitro to affect intracellular signalling implicated in the regulation of colonic cancer stem cells (CSCs). MATERIALS AND METHODS: In order to study whether Foxy5 can modulate CSCs, either HT-29 or Caco-2 human colonic cancer cells, both lacking endogenous WNT5A expression, were inoculated subcutaneously into nude mice. RESULTS: Foxy5 reduced the expression of the stem-cell marker aldehyde dehydrogenase and, interestingly, the specific colon CSC marker double cortin-like kinase 1. Foxy5 also reduced active β-catenin and the expression of its downstream target Achaete Scute complex homolog 2, a CSC-preserving transcription factor. Foxy5 also reduced cyclo-oxygenase 2 expression, responsible for the formation of the CSC-promoting prostaglandin E2 (PGE2), but increased that of 15-hydroxyprostaglandin dehydrogenase expression, a PGE2-degrading enzyme. Accordingly, Foxy5 impairs both β-catenin and PGE2 signalling, both of which have been implicated in promoting the niche of colonic CSCs. CONCLUSION: Our data suggest that Foxy5 can complement the traditional adjuvant chemotherapeutic treatment to which CSCs are resistant.
  •  
7.
  • Rodriguez-Zabala, Maria, et al. (author)
  • Combined GLUT1 and OXPHOS inhibition eliminates acute myeloid leukemia cells by restraining their metabolic plasticity
  • 2023
  • In: Blood Advances. - 2473-9529. ; 7:18, s. 5382-5395
  • Journal article (peer-reviewed)abstract
    • Acute myeloid leukemia (AML) is initiated and propagated by leukemia stem cells (LSCs), a self-renewing population of leukemia cells responsible for therapy resistance. Hence, there is an urgent need to identify new therapeutic opportunities targeting LSCs. Here, we performed an in vivo CRISPR knockout screen to identify potential therapeutic targets by interrogating cell surface dependencies of LSCs. The facilitated glucose transporter type 1 (GLUT1) emerged as a critical in vivo metabolic dependency for LSCs in a murine MLL::AF9–driven model of AML. GLUT1 disruption by genetic ablation or pharmacological inhibition led to suppression of leukemia progression and improved survival of mice that received transplantation with LSCs. Metabolic profiling revealed that Glut1 inhibition suppressed glycolysis, decreased levels of tricarboxylic acid cycle intermediates and increased the levels of amino acids. This metabolic reprogramming was accompanied by an increase in autophagic activity and apoptosis. Moreover, Glut1 disruption caused transcriptional, morphological, and immunophenotypic changes, consistent with differentiation of AML cells. Notably, dual inhibition of GLUT1 and oxidative phosphorylation (OXPHOS) exhibited synergistic antileukemic effects in the majority of tested primary AML patient samples through restraining of their metabolic plasticity. In particular, RUNX1-mutated primary leukemia cells displayed striking sensitivity to the combination treatment compared with normal CD34+ bone marrow and cord blood cells. Collectively, our study reveals a GLUT1 dependency of murine LSCs in the bone marrow microenvironment and demonstrates that dual inhibition of GLUT1 and OXPHOS is a promising therapeutic approach for AML.
  •  
Skapa referenser, mejla, bekava och länka
  • Result 1-7 of 7

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 Close

Copy and save the link in order to return to this view