Journal articles on the topic 'TGFbeta pathway'

To see the other types of publications on this topic, follow the link: TGFbeta pathway.

Create a spot-on reference in APA, MLA, Chicago, Harvard, and other styles

Select a source type:

Consult the top 50 journal articles for your research on the topic 'TGFbeta pathway.'

Next to every source in the list of references, there is an 'Add to bibliography' button. Press on it, and we will generate automatically the bibliographic reference to the chosen work in the citation style you need: APA, MLA, Harvard, Chicago, Vancouver, etc.

You can also download the full text of the academic publication as pdf and read online its abstract whenever available in the metadata.

Browse journal articles on a wide variety of disciplines and organise your bibliography correctly.

1

Verma, Amit. "Presentation name: TGFbeta pathway targeting." Leukemia Research 108 (September 2021): 106682.8. http://dx.doi.org/10.1016/j.leukres.2021.106682.8.

Full text
APA, Harvard, Vancouver, ISO, and other styles
2

Weiss, Alexander, and Liliana Attisano. "The TGFbeta Superfamily Signaling Pathway." Wiley Interdisciplinary Reviews: Developmental Biology 2, no. 1 (October 5, 2012): 47–63. http://dx.doi.org/10.1002/wdev.86.

Full text
APA, Harvard, Vancouver, ISO, and other styles
3

Krishna, S., L. L. Maduzia, and R. W. Padgett. "Specificity of TGFbeta signaling is conferred by distinct type I receptors and their associated SMAD proteins in Caenorhabditis elegans." Development 126, no. 2 (January 15, 1999): 251–60. http://dx.doi.org/10.1242/dev.126.2.251.

Full text
Abstract:
In C. elegans, the TGFbeta-like type II receptor daf-4 is required for two distinct signaling pathways. In association with the type I receptor daf-1, it functions in the dauer pathway. In addition, it is also required for body size determination and male tail patterning, roles which do not require daf-1. In an effort to determine how two different signals are transmitted through daf-4, we looked for other potential signaling partners for DAF-4. We have cloned and characterized a novel type I receptor and show that it is encoded by sma-6. Mutations in sma-6 generate the reduced body size (Sma) and abnormal mail tail (Mab) phenotypes identical to those observed in daf-4 and sma-2, sma-3, sma-4 mutants (C. elegans Smads), indicating that they function in a common signaling pathway. However, mutations in sma-6, sma-2, sma-3, or sma-4 do not produce constitutive dauers, which demonstrates that the unique biological functions of daf-4 are mediated by distinct type I receptors functioning in parallel pathways. We propose that the C. elegans model for TGFbeta-like signaling, in which distinct type I receptors determine specificity, may be a general mechanism of achieving specificity in other organisms. These findings distinguish between the manner in which signaling specificity is achieved in TGFbeta-like pathways and receptor tyrosine-kinase (RTK) pathways.
APA, Harvard, Vancouver, ISO, and other styles
4

Tran, Dat Q., Ellen Regalado, and Dianna Milewicz. "Immune Perturbation In Patients With Tgfbeta Pathway Defects." Journal of Allergy and Clinical Immunology 133, no. 2 (February 2014): AB248. http://dx.doi.org/10.1016/j.jaci.2013.12.881.

Full text
APA, Harvard, Vancouver, ISO, and other styles
5

Wang, ZacK Z., Hao Bai, Melanie Arzigian, Yong-Xing Gao, and Wen-Shu Wu. "BMP4 and TGFbeta Differentially Regulate CD34+ Progenitor Development in Human Embryonic Stem Cells through SMAD-Dependent Pathway." Blood 112, no. 11 (November 16, 2008): 889. http://dx.doi.org/10.1182/blood.v112.11.889.889.

Full text
Abstract:
Abstract Pluripotent stem cells derived from patients, including embryonic stem (ES) cells and “induced pluripotent stem” (iPS) cells, are a promising area of regenerative medical research. A major roadblock toward human clinical therapies using ES cells or iPS cells is to define the factors that direct ES cell differentiation into lineage specific cells. We previously established a simple and efficient human embryonic stem cell (hESC) differentiation system to generate CD34+/CD31+ progenitor cells that gave rise to hematopoietic and endothelial cells (Nat Biotech.25:317, 2007). To advance potential clinical application and to define the effects of growth factors on hematopoietic and vascular differentiation, we assessed hESC differentiation on human feeder cells in serum-free condition without intermediate embryoid body (EB) formation. We investigated the roles of BMPs, TGFbeta, VEGF, and FGF2 in directing hESC differentiation. Growth factors were added into culture at different time points to test their stage specific roles. Our study demonstrated that BMP proteins, including BMP2, BMP4, and BMP7, but not BMP9, had synergic effects to VEGF and FGF-2 on hESC differentiation to CD34+/CD31+ progenitor cells. BMP4 was essential to initial CD34+/CD31+ cell development, whereas VEGF and FGF2 promoted the differentiation in later stage, suggesting the sequential roles of BMP4, VEGF and FGF2 in directing hESC differentiation to CD34+/CD31+ progenitor cells. TGFbeta or activin promoted hESC differentiation into CD34+/CD31− cells that were unable to give rise to hematopoietic, endothelial, and smooth muscle cells. Furthermore, TGFbeta or activin activated Smad2/3 signaling, and suppressed BMP4-induced CD34+/CD31+ cells. Microarray analysis revealed that BMP4-induced CD34+ cells expressed hematopoietic, endothelial and smooth muscle genes, including GATA2, gamma globins, VE-Cad, KDR, CD31, Tie2, and aortic smooth muscle actin, whereas TGFbeta-induced CD34+ cells expressed pluripotent markers and endoderm markers, including Oct3/4, Sox2, and Nanog, HHEX, GATA6, and FoxA2. Both canonical BMP signaling (Smad1/5/8-dependent) and non-canonical BMP signaling (p38 MAPK and p42 ERK pathway) were activated by BMP4 in hESCs. Dorsomorphin specifically inhibited BMP4-mediated phosphorylation of Smad1/5/8, and blocked hESC differentiation into CD34+/CD31+ cells. In summary, BMPs and TGFbeta regulate distinct populations of CD34+ cells in hESCs. BMP-Smad1/5/8 pathway is critical for hematopoietic and vascular progenitor development.
APA, Harvard, Vancouver, ISO, and other styles
6

French, Deborah, Francesca Belleudi, Maria Mauro, Francesca Mazzetta, Salvatore Raffa, Vincenza Fabiano, Antonio Frega, and Maria Torrisi. "Expression of HPV16 E5 down-modulates the TGFbeta signaling pathway." Molecular Cancer 12, no. 1 (2013): 38. http://dx.doi.org/10.1186/1476-4598-12-38.

Full text
APA, Harvard, Vancouver, ISO, and other styles
7

Tran, Dat, Ellen Regalado, and Dianna Milewicz. "Immune perturbation in patients with TGFbeta pathway defects (LYM7P.729)." Journal of Immunology 192, no. 1_Supplement (May 1, 2014): 193.17. http://dx.doi.org/10.4049/jimmunol.192.supp.193.17.

Full text
Abstract:
Abstract Intro: Mutations in TGFbR1, TGFbR2 and SMAD3 are associated with familial thoracic aortic aneurysms and aortic dissections (TAAD). These patients offer an opportunity to study their immune development. Methods: PBMC from TAAD (n=9) and controls (CT, n=8) were analyzed by FACS. Th1 (IFNg) and Th17 (IL17A) were determined with intracellular cytokine staining. Foxp3+ Tregs were detected with anti-Foxp3. CD19+ were analyzed for naive (IgD+CD27-), unswitched (IgD+CD27+) and switched memory (IgD-CD27+). Plasmacytoid (CD303+pDC) and myeloid (CD1c+mDC) were defined within lineage-1 negative population. Results: %CD3-CD16+NK, CD3+CD16+NKT, CD4+, CD8+ and CD4+CD45RA+ in TAAD were similar to HC. Average %CD19+ (20.8vs7.3, p=0.006) and naive B cells (81.3vs66.6, p=0.004) were higher in TAADvsCT. The unswitched were similar but the switched B cells were lower (8.6vs15.5, p=0.01). While the %Tregs were similar, there was a remarkable reduction (1/2-3 folds, p<0.05) in Foxp3 concentration based on median fluorescence intensity of Foxp3 in TAAD. There was a significant reduction of %Th17 (0.14vs0.61, p=0.01), while the Th1 were similar. %pDC (9.4vs24.1, p=0.009) and %mDC (11.4vs17.9, p=0.01) were also lower in TAAD. Conclusions: These results demonstrate the involvement of TGFbeta signaling in B cells, DCs, Th17 and Treg development. Further studies and monitoring of the clinical effects of these immunological perturbations are needed to appreciate the impact of their underlying disease.
APA, Harvard, Vancouver, ISO, and other styles
8

Tervonen, Topi A., Denis Belitškin, Pauliina Munne, Shishir M. Pant, Ilida Suleymanova, Kati Belitškina, Jeroen Pouwels, and Juha Klefström. "Abstract 834: Serine protease hepsin regulates tumor growth via TGFbeta-EGFR signaling axis." Cancer Research 82, no. 12_Supplement (June 15, 2022): 834. http://dx.doi.org/10.1158/1538-7445.am2022-834.

Full text
Abstract:
Abstract Hepsin (encoded by HPN gene) is a type II transmembrane serine protease, which is commonly overexpressed in carcinomas of prostate and breast. Hepsin protein is known to be stabilized by Ras-MAPK pathway signaling, and downstream, this protease regulates the degradation of extracellular matrix (ECM) components and activates growth factor pathways, including hepatocyte growth factor (HGF) and transforming growth factor beta (TGF beta) pathway. However, the impact of the hepsin-dependent signaling on cell proliferation and tumor growth is not well-understood. Therefore, we sought to clarify the role of hepsin during these events using engineered breast cancer cell lines, Hpn CRISPR knockout mouse model crossed with Wap-Myc breast cancer mouse model, and patient-derived explant cultures (PDEC)s from human breast tumors. We isolated Wap-Myc; Hpn-/- mammary tumor cells and made orthotopic transplantation into syngeneic wild-type recipient mice. The resulting Wap-Myc tumors that lack hepsin had reduced size both in primary and metastatic site compared to tumors derived from Wap-Myc; Hpn+/+ tumor cells. This decrease in growth was accompanied by downregulation in TGF beta and EGFR signaling as well as substantial reduction in total EGFR protein level. We further demonstrated that in 3D culture conditions overexpression of hepsin induced cell proliferation, which was TGF beta 1 and EGFR signaling-dependent, while PDECs treated with hepsin inhibitory antibodies and small molecules had decreased EGFR and TGF beta signaling activity and reduction in proliferation marker expression. Taken together, this study demonstrates a role for hepsin as a regulator of cell proliferation and tumor growth through TGF beta and EGFR pathways and may provide an interesting druggable upstream target to inhibit TGF beta and EGFR pathways in breast cancer. Citation Format: Topi A. Tervonen, Denis Belitškin, Pauliina Munne, Shishir M. Pant, Ilida Suleymanova, Kati Belitškina, Jeroen Pouwels, Juha Klefström. Serine protease hepsin regulates tumor growth via TGFbeta-EGFR signaling axis [abstract]. In: Proceedings of the American Association for Cancer Research Annual Meeting 2022; 2022 Apr 8-13. Philadelphia (PA): AACR; Cancer Res 2022;82(12_Suppl):Abstract nr 834.
APA, Harvard, Vancouver, ISO, and other styles
9

Tang, S. J., P. A. Hoodless, Z. Lu, M. L. Breitman, R. R. McInnes, J. L. Wrana, and M. Buchwald. "The Tlx-2 homeobox gene is a downstream target of BMP signalling and is required for mouse mesoderm development." Development 125, no. 10 (May 15, 1998): 1877–87. http://dx.doi.org/10.1242/dev.125.10.1877.

Full text
Abstract:
TGFbeta-related factors are critical regulators of vertebrate mesoderm development. However, the signalling cascades required for their function during this developmental process are poorly defined. Tlx-2 is a homeobox gene expressed in the primitive streak of mouse embryos. Exogenous BMP-2 rapidly activates Tlx-2 expression in the epiblast of E6.5 embryos. A Tlx-2 promoter element responds to BMP-2 signals in P19 cells, and this response is mediated by BMP type I receptors and Smad1. These results suggest that Tlx-2 is a downstream target gene for BMP signalling in the primitive streak where BMP-4 and other TGFbeta-related factors are expressed. Furthermore, disruption of Tlx-2 function leads to early embryonic lethality. Similar to BMP4 and ALK3 mutants, the mutant embryos display severe defects in primitive streak and mesoderm formation. These experiments thus define a BMP/Tlx-2 signalling pathway that is required during early mammalian gastrulation.
APA, Harvard, Vancouver, ISO, and other styles
10

Thatcher, J. D., C. Haun, and P. G. Okkema. "The DAF-3 Smad binds DNA and represses gene expression in the Caenorhabditis elegans pharynx." Development 126, no. 1 (January 1, 1999): 97–107. http://dx.doi.org/10.1242/dev.126.1.97.

Full text
Abstract:
Gene expression in the pharyngeal muscles of Caenorhabditis elegans is controlled in part by organ-specific signals, which in the myo-2 gene target a short DNA sequence termed the C subelement. To identify genes contributing to these signals, we performed a yeast one-hybrid screen for cDNAs encoding factors that bind the C subelement. One clone recovered was from daf-3, which encodes a Smad most closely related to vertebrate Smad4. We demonstrated that DAF-3 binds C subelement DNA directly and specifically using gel mobility shift and DNase1 protection assays. Mutation of any base in the sequence GTCTG interfered with binding in the gel mobility shift assay, demonstrating that this pentanucleotide is a core recognition sequence for DAF-3 binding. daf-3 is known to promote formation of dauer larvae and this activity is negatively regulated by TGFbeta-like signaling. To determine how daf-3 affects C subelement enhancer activity in vivo, we examined expression a gfp reporter controlled by a concatenated C subelement oligonucleotide in daf-3 mutants and other mutants affecting the TGFbeta-like signaling pathway controlling dauer formation. Our results demonstrate that wild-type daf-3 can repress C subelement enhancer activity during larval development and, like its dauer-promoting activity, daf-3's repressor activity is negatively regulated by TGFbeta-like signaling. We have examined expression of this gfp reporter in dauer larvae and have observed no daf-3-dependent repression of C activity. These results suggest daf-3 directly regulates pharyngeal gene expression during non-dauer development.
APA, Harvard, Vancouver, ISO, and other styles
11

Vargesson, Neil. "Vascularization of the developing chick limb bud: role of the TGFbeta signalling pathway." Journal of Anatomy 202, no. 1 (January 2003): 93–103. http://dx.doi.org/10.1046/j.1469-7580.2003.00133.x.

Full text
APA, Harvard, Vancouver, ISO, and other styles
12

Koromila, T., Z. Dailiana, S. Samara, C. Chassanidis, V. Aleporou - Marinou, and P. Kollia. "TGFbeta-BMP signaling pathway gene expression in osteoporotic postmenopausal women by DNA microarrays." Bone 48 (May 2011): S159. http://dx.doi.org/10.1016/j.bone.2011.03.350.

Full text
APA, Harvard, Vancouver, ISO, and other styles
13

Yang, Guo-Xiang, Zhe-Xiong Lian, Ya-Hui Chuang, Yuki Moritoki, Aftab A. Ansari, Richard A. Flavell, William M. Ridgway, and M. Eric Gershwin. "CD8 T cells Play a Critical Role in Primary Biliary Cirrhosis of dnTGFbetaRII Mice (130.28)." Journal of Immunology 178, no. 1_Supplement (April 1, 2007): S233. http://dx.doi.org/10.4049/jimmunol.178.supp.130.28.

Full text
Abstract:
Abstract Human primary biliary cirrhosis (PBC) is characterized serologically by antimitochondrial antibodies (AMA) and histologically by an intense T cell portal infiltrate that destroys bile ducts. We have taken advantage of the PBC-like disease exhibited by dnTGFbetaRII mice and focused our attention on the liver T cell infiltrate. One major advantage to this animal model is the ability to perform cell transfer. Therefore to address the issue of T cell effector mechanisms, we isolated dnTGFbetaRII splenic CD4+ or CD8+ T cells and transferred these populations into Rag1 k/o mice. Importantly, the CD8+ T cells transfer group demonstrated a significant expansion of T cells and the presence of portal tract infiltrates in recipient mice. In contrast, although CD4+ T cells did expand in the recipient group, they did not home or focus within the portal tracts. Our results demonstrate that the impaired TGFbeta signaling pathway in these mice leads to a CD8 cytotoxic T cell population that plays a critical role in biliary cell damage. These data have implications not only for understanding TGFbeta signaling and autoimmunity in these mice, but also in developing appropriate focused immunotherapy to prevent biliary damage.
APA, Harvard, Vancouver, ISO, and other styles
14

Postigo, A. A. "Opposing functions of ZEB proteins in the regulation of the TGFbeta/BMP signaling pathway." EMBO Journal 22, no. 10 (May 15, 2003): 2443–52. http://dx.doi.org/10.1093/emboj/cdg225.

Full text
APA, Harvard, Vancouver, ISO, and other styles
15

Moubarak, Rana, Ana De Pablos-Aragoneses, Vanessa Ortiz-Barahona, Richard Von Itter, Yixiao Gong, Michael Gowen, Sebastian Stefan, et al. "Abstract B30: The histone demethylase PHF8 epigenetically regulates the TGFbeta pathway to promote melanoma metastasis." Cancer Research 80, no. 19_Supplement (October 1, 2020): B30. http://dx.doi.org/10.1158/1538-7445.mel2019-b30.

Full text
Abstract:
Abstract The specific contribution of epigenetic dysregulation to the metastatic process remains understudied. Through a meta-analysis of gene expression datasets followed by a functional mini-screen, we identified PHF8, a histone demethylase of the Jumonji C proteins family, as a novel prometastatic factor in melanoma. Loss- and gain-of-function approaches demonstrate that PHF8 promotes cell invasion without affecting proliferation in vitro, and increases dissemination but not tumor growth in vivo, thus strengthening its specific contribution to the acquisition of metastatic potential. PHF8 demethylase activity is required for its proinvasive role. Transcriptomic and epigenomic analyses revealed that PHF8 orchestrates molecular changes that directly control the TGFβ signaling pathway and, as a direct consequence, melanoma invasion. Citation Format: Rana Moubarak, Ana De Pablos-Aragoneses, Vanessa Ortiz-Barahona, Richard Von Itter, Yixiao Gong, Michael Gowen, Sebastian Stefan, Diana Argibay, Eleazar De Miera-Vega, Igor Dolgalev, Elena Sokolova, Farbod Darvishian, Aristotelis Tsirigos, Iman Osman, Eva Hernando. The histone demethylase PHF8 epigenetically regulates the TGFbeta pathway to promote melanoma metastasis [abstract]. In: Proceedings of the AACR Special Conference on Melanoma: From Biology to Target; 2019 Jan 15-18; Houston, TX. Philadelphia (PA): AACR; Cancer Res 2020;80(19 Suppl):Abstract nr B30.
APA, Harvard, Vancouver, ISO, and other styles
16

Mintzer, K. A., M. A. Lee, G. Runke, J. Trout, M. Whitman, and M. C. Mullins. "Lost-a-fin encodes a type I BMP receptor, Alk8, acting maternally and zygotically in dorsoventral pattern formation." Development 128, no. 6 (March 15, 2001): 859–69. http://dx.doi.org/10.1242/dev.128.6.859.

Full text
Abstract:
TGFbeta signaling pathways of the bone morphogenetic protein (BMP) subclass are essential for dorsoventral pattern formation of both vertebrate and invertebrate embryos. Here we determine by chromosomal mapping, linkage analysis, cDNA sequencing and mRNA rescue that the dorsalized zebrafish mutant lost-a-fin (laf) is defective in the gene activin receptor-like kinase 8 (alk8), which encodes a novel type I TGFbeta receptor. The alk8 mRNA is expressed both maternally and zygotically. Embyros that lack zygotic, but retain maternal Laf/Alk8 activity, display a weak dorsalization restricted to the tail and die by 3 days postfertilization. We rescued the laf dorsalized mutant phenotype by alk8 mRNA injection and generated homozygous laf/alk8 mothers to investigate the maternal role of Laf/Alk8 activity. Adult fish lacking Laf/Alk8 activity are fertile, exhibit a growth defect and are significantly smaller than their siblings. Embryos derived from homozygous females, which lack both maternal and zygotic Laf/Alk8 activity, display a strongly dorsalized mutant phenotype, no longer limited to the tail. These mutant embryos lack almost all gastrula ventral cell fates, with a concomitant expansion of dorsal cell types. During later stages, most of the somitic mesoderm and neural tissue circumscribe the dorsoventral axis of the embryo. Zygotic laf/alk8 mutants can be rescued by overexpression of the BMP signal transducer Smad5, but not the Bmp2b or Bmp7 ligands, consistent with the Laf/Alk8 receptor acting within a BMP signaling pathway, downstream of a Bmp2b/Bmp7 signal. Antibodies specific for the phosphorylated, activated form of Smad1/5, show that BMP signaling is nearly absent in gastrula lacking both maternal and zygotic Laf/Alk8 activity, providing further evidence that Laf/Alk8 transduces a BMP signal. In total, our work strongly supports the role of Laf/Alk8 as a type I BMP receptor required for the specification of ventral cell fates.
APA, Harvard, Vancouver, ISO, and other styles
17

Yokouchi, Y., J. Sakiyama, T. Kameda, H. Iba, A. Suzuki, N. Ueno, and A. Kuroiwa. "BMP-2/-4 mediate programmed cell death in chicken limb buds." Development 122, no. 12 (December 1, 1996): 3725–34. http://dx.doi.org/10.1242/dev.122.12.3725.

Full text
Abstract:
During limb development, the mesenchymal cells in restricted areas of limb bud, anterior necrotic zone, posterior necrotic zone, opaque zone and interdigital necrotic zones, are eliminated by programmed cell death. The transcripts of bone morphogenetic protein (Bmp)-2 and −4 were first detected in the areas where cell death was observed, then showed overlapping expression with the programmed cell death zones except the opaque zone. To investigate the function of BMP-2 and BMP-4 during limb pattern formation, the dominant negative form of BMP receptor was overexpressed in chick leg bud via a replication-competent retrovirus to block the endogenous BMP-2/-4 signaling pathway. This resulted in excess web formation at the anterior and posterior regions of limb buds in addition to marked suppression of the regression of webbing at the interdigital regions. Significant reductions in the number of apoptotic cells in these three necrotic zones were found in the limb buds which received the virus carrying dominant negative BMP receptor. This indicates that extra tissue formation is due to suppression of programmed cell death in the three necrotic zones. Moreover, BMP-2/-4 protein induced apoptosis of mesenchymal cells isolated from the interdigital region in vitro. Other TGFbeta family proteins as TGFbeta1 and Activin did not show this effect. These results suggest that BMP-2 and BMP-4 are the apoptotic signal molecules of the programmed cell death process in the chick limb buds.
APA, Harvard, Vancouver, ISO, and other styles
18

Wu, Xiao-Ling, Wei-Zheng Zeng, Ming-De Jiang, Jian-Ping Qin, and Hui Xu. "Effect of Oxymatrine on the TGFbeta-Smad signaling pathway in rats with CCl4-induced hepatic fibrosis." World Journal of Gastroenterology 14, no. 13 (2008): 2100. http://dx.doi.org/10.3748/wjg.14.2100.

Full text
APA, Harvard, Vancouver, ISO, and other styles
19

Fazio, Grazia, Chiara Palmi, Greta Giordano Attianese, Andrea Biondi, Antonius Rolink, and Giovanni Cazzaniga. "PAX5/TEL Causes down Regulation of CD19 and TGFbeta Resistance in PreBI Cells." Blood 108, no. 11 (November 16, 2006): 1416. http://dx.doi.org/10.1182/blood.v108.11.1416.1416.

Full text
Abstract:
Abstract The PAX5/TEL chimeric gene was cloned from the translocation t(9;12)(q11;p13) in an ALL patient. Recent data indicate that the PAX5/TEL fusion defines the cytogenetic entity dic(9;12)(p13;p13), which accounts for about 1% of childhood ALL, almost exclusively B-progenitor ALL. PAX5/TEL is likely to be an aberrant transcription factor, resulting from joining the 5′ region of PAX5 (a transcription factor essential for B cell development) to the 3′ region of TEL/ETV6, containing the Ets-family DNA binding domain. We have cloned the FLAG-full length chimeric PAX5/TEL cDNA in the retroviral vector pMSCV-IRES-GFP (MigR1) to transduce target cells. We have demonstrated a specific nuclear localization of the chimeric protein in NIH3T3 by immunofluorescence analysis. Moreover, we observed a PAX5/TEL dependent decrease of the cellular growth rate in IL-3 dependent murine proB Ba/F3 cells. We further investigated the function of the PAX5/TEL chimeric protein as a potential oncoprotein in murine preBI cells, as a more physiological model. Murine PAX5 −/− preBI cells and wild type preBI cells were purified as B220+/c-KIT+ cells from mouse fetal liver and they were cultured on OP9 and DL1-OP9 stroma cells in presence of IL-7. The OP9 stroma supports B cell proliferation and survival; the DL1-OP9 stroma expresses Delta-like1, one of the Notch ligands, and it’s important to support T cell development. Both PAX5 −/− preBI cells and wild type preBI cells were transduced with the retroviral construct pMSCV-PAX5/TEL-IRES-GFP to analyze cell proliferation, differentiation and growth-dependence on IL-7. Wild type preBI cells expressing PAX5/TEL showed down modulation of CD19 when cultured on OP9 stroma in presence of IL-7; an inverse correlation was observed between the levels of expression of GFP and of CD19. The down modulation of CD19 can be involved in driving the preBI cell into differentiation block. A possible explanation of CD19 repression can rely on a potential competition between PAX5/TEL and endogenous PAX5 to bind PAX5 consensus region on DNA. On OP9 stroma, PAX5/TEL preBI cells are resistant to TGFbeta anti-proliferative and apoptotic effects, with a three-fold increased growth rate than control cells. Although the specific mechanism of PAX5/TEL disruption of TGFbeta signalling pathway remains to be investigated, we propose the TGFbeta resistance by PAX5/TEL as a way to evade the immunosurveillance. PAX5/TEL-preBI cells cultured on DL1-OP9 showed a different phenotype, with up-regulation of c-KIT and down-regulation of CD44. PAX5−/− preBI cells infected with PAX5TEL and grown on OP9 were CD19 negative even in the presence of PAX5TEL. On DL1-OP9 stroma, PAX5TEL cells were able to differentiate maintaining the developmental plasticity of PAX5 −/− preBI cells. These preliminary results indicate a role of PAX5/TEL as a transcription factor, potentially with a suppressor function, down regulating CD19 expression, thus suggesting a function on B cell differentiation. The chimera is able to interfere with TGFbeta pathway, inducing resistance and conferring an advantage in cell survival, evading the immunosurveillance. PAX5TEL do not replace PAX5 functions in PAX5−/− cells, it cannot activate PAX5 target genes as CD19, important for restoring B cell differentiation. Further analyeis are needed to better evaluate the role of PAX5/TEL protein, both in vivo and in vitro models.
APA, Harvard, Vancouver, ISO, and other styles
20

Zhao, JY, and B. Chen. "ID: 118: UBIQUITIN E3 LIGASE FIEL1 REGUALTES TGFBETA SIGNALING IN LUNG FIBROBLAST." Journal of Investigative Medicine 64, no. 4 (March 22, 2016): 967.2–967. http://dx.doi.org/10.1136/jim-2016-000120.116.

Full text
Abstract:
Tumor growth factor β (TGFb) signaling plays a key role in the pathogenesis of tissue fibrosis, specifically Idiopathic Pulmonary Fibrosis (IPF). IPF is the deadliest of interstitial lung diseases with a median survival of merely three years. Currently IPF affects more than 35,000 patients each year, but despite extensive research, there is still no effective treatment. Thus, the discovery of new models that regulate TGFb signaling and reveal “authentic” drug targets could serve as a springboard for the introduction of a new generation of anti-fibrotic agents to be used in the clinical arena. There is a fundamental, unmet scientific and clinical need to discover new mechanistic models that control overactive TGFb signaling and, when left unchecked, can produce severe tissue fibrosis.PIAS4 is a pivotal protein in controlling TGFβ effects. Here we discovered a new protein isoform encoded by KIAA0317, termed FIEL1 (Fibrosis Inducing E3 Ligase 1), that potently stimulates TGFβ signaling pathway through the site-specific ubiquitination and destabilization of PIAS4. FIEL1 protein is highly expressed in lung tissues from patients with Idiopathic Pulmonary Fibrosis (IPF), whereas PIAS4 protein levels are significantly reduced. FIEL1 overexpression significantly activates TGFβ signaling. Further, we developed a novel small molecule inhibitor towards FIEL1 that is highly effective in ameliorating TGFβ signaling. This study provides a basis for IPF therapeutic intervention by modulating PIAS4 protein abundance.
APA, Harvard, Vancouver, ISO, and other styles
21

Lints, R., and S. W. Emmons. "Patterning of dopaminergic neurotransmitter identity among Caenorhabditis elegans ray sensory neurons by a TGFbeta family signaling pathway and a Hox gene." Development 126, no. 24 (December 15, 1999): 5819–31. http://dx.doi.org/10.1242/dev.126.24.5819.

Full text
Abstract:
We have investigated the mechanism that patterns dopamine expression among Caenorhabditis elegans male ray sensory neurons. Dopamine is expressed by the A-type sensory neurons in three out of the nine pairs of rays. We used expression of a tyrosine hydroxylase reporter transgene as well as direct assays for dopamine to study the genetic requirements for adoption of the dopaminergic cell fate. In loss-of-function mutants affecting a TGFbeta family signaling pathway, the DBL-1 pathway, dopaminergic identity is adopted irregularly by a wider subset of the rays. Ectopic expression of the pathway ligand, DBL-1, from a heat-shock-driven transgene results in adoption of dopaminergic identity by rays 3–9; rays 1 and 2 are refractory. The rays are therefore prepatterned with respect to their competence to be induced by a DBL-1 pathway signal. Temperature-shift experiments with a temperature-sensitive type II receptor mutant, as well as heat-shock induction experiments, show that the DBL-1 pathway acts during an interval that extends from two to one cell generation before ray neurons are born and begin to differentiate. In a mutant of the AbdominalB class Hox gene egl-5, rays that normally express EGL-5 do not adopt dopaminergic fate and cannot be induced to express DA when DBL-1 is provided by a heat-shock-driven dbl-1 transgene. Therefore, egl-5 is required for making a subset of rays capable of adopting dopaminergic identity, while the function of the DBL-1 pathway signal is to pattern the realization of this capability.
APA, Harvard, Vancouver, ISO, and other styles
22

Chen, Junfeng, Xiaojun Guo, Guangjun Zeng, and Jianhua Liu. "Effect of miR-34a on Postmenopausal Osteoporosis in Rats Through TGFβ/Smad Signaling Pathway." Journal of Biomaterials and Tissue Engineering 10, no. 4 (April 1, 2020): 487–92. http://dx.doi.org/10.1166/jbt.2020.2281.

Full text
Abstract:
30 healthy SD rats of three months were chosen and divided randomly into shamed group, model group, miR-34a mimic transfection group, each of 10 cases. Then to detect serum miR-34a level after three months, TGF-β , Smad4 proteins levels by western blot, serum Ca level, bone mineral density (BMD), bone metabolic markers total type I collagen amino-terminal elongation peptide (TPINP), unique sequence of beta-collagen (beta-CTX), Histopathological Observation and Morphometric Detection of Bone after executed. Results The levels of miR-34a, TGF-β, Smad4 in transfection group were significantly higher than the model group, shamed group the least (P < 0 05), while serum Ca level and BMD value were lowest (P < 0 05). What's more, the TPINP and -CTX levels in transfection group were significantly higher than the model group, shamed group the least (P < 0 05). It proved that fractured trabeculae widened spacing and disordered structure, reduction of trabecular area percentage and mean trabecular thickness in model group and transfection group. It may influence postmenopausal osteoporosis in rats via activation of TGFbeta/Smad signalling pathway by higher expression of miR-34a.
APA, Harvard, Vancouver, ISO, and other styles
23

Malik, M., J. Britten-Webb, G. Levy, M. Gilden, and W. H. Catherino. "Tgfbeta mediated fibrosis in leiomyoma cells works through the smad dependent signaling and not through MAPK/ERK pathway." Fertility and Sterility 98, no. 3 (September 2012): S72. http://dx.doi.org/10.1016/j.fertnstert.2012.07.260.

Full text
APA, Harvard, Vancouver, ISO, and other styles
24

Baron, Margaret H., Stephen Willey, Kenneth Sahr, Hailan Zhang, Kevin Balbi, Michael A. Dyer, and Matthew Adlam. "Induction and Patterning of Pre-Hemato-Vascular Mesoderm by the Mouse Mix Homeodomain Protein." Blood 104, no. 11 (November 16, 2004): 565. http://dx.doi.org/10.1182/blood.v104.11.565.565.

Full text
Abstract:
Abstract Members of the Xenopus and zebrafish Mix/Bix family of paired class homeodomain proteins play determining roles in both mesoderm and endoderm development and are induced by members of the TGFbeta/BMP family of signaling molecules. A single Mix gene has been identified in mouse, humans and chick. Prior to gastrulation, the mouse Mix (mMix) gene is expressed in the visceral endoderm and later in the primitive streak and nascent mesoderm, where it overlaps, in part, with T. Mix expression in ES-derived embryoid bodies is early and transient, overlapping partially with Flk1 activation around the time of formation of hemangioblasts. Both mMix mRNA and protein are found in a FACS-sorted population of T+Flk1+ cells from ES cell-derived embryoid bodies (EBs) which contains hemangioblasts. A complex embryonic lethal phenotype has been reported for Mix deficient embryos, including defects in allantoic (vascular) and cardiogenesis. Mesoderm forms in these embryos but is not patterned properly. Embryonic lethality occurs around E10.5–11, presumably as a result of the cardiovascular defects. We have generated inducible ES cell lines in which expression of Mix protein is responsive to doxycycline. Ectopic expression of Mix in EBs results in premature, enhanced expression of hemangioblast, angioblast and hematopoietic stem cell markers (mRNA and FACS analyses) and increased formation of stem/progenitor cells in clonogenic assays in methylcellulose. Together, the expression analyses, knockout phenotype, and gain-of-function studies in ES cells suggest that mMix functions early in induction and patterning of mesoderm, including formation of hematopoietic and endothelial lineages. Potential mMix target genes are being identified by microarray analyses of the inducible Mix ES lines. To examine mMix activities in vivo, we have generated null and conditional mMix knockout mice from several independently targeted ES cell lines. Analysis of these animals is in progress. Like Xenopus Mix.1, mouse Mix may represent an important connection between the TGFbeta/BMP pathway and hematopoietic/vascular development in the embryo.
APA, Harvard, Vancouver, ISO, and other styles
25

Nagamatsu, Yasuko, and Yasumi Ohshima. "Mechanisms for the control of body size by a G-kinase and a downstream TGFbeta signal pathway in Caenorhabditis elegans." Genes to Cells 9, no. 1 (January 2004): 39–47. http://dx.doi.org/10.1111/j.1356-9597.2004.00700.x.

Full text
APA, Harvard, Vancouver, ISO, and other styles
26

Bakker, W., CKE Dingenouts, K. Lodder, MR De Vries, HJ Mager, RJ Snijder, CJ Westerman, P. Dijke, PH Quax, and MJTH Goumans. "P348Impaired macrophage polarization in endoglin haplo-insufficiency leading to defective tissue repair is recovered by counter balance the TGFbeta pathway." Cardiovascular Research 103, suppl 1 (June 27, 2014): S63.4—S63. http://dx.doi.org/10.1093/cvr/cvu091.34.

Full text
APA, Harvard, Vancouver, ISO, and other styles
27

Wang, S. X., J. Wang, E. Ozhegov, R. Srinivasan, O. O. Olowokure, A. X. Qu, B. Aronow, and V. Bogdanov. "Identification of unique gene expression signatures in colon cancer stroma using microarray technology." Journal of Clinical Oncology 29, no. 4_suppl (February 1, 2011): 447. http://dx.doi.org/10.1200/jco.2011.29.4_suppl.447.

Full text
Abstract:
447 Background: Microarrays are widely used to study gene expression patterns in cancer. In colorectal cancer, it has proven useful to predict recurrence. The majority of expression profiles are generated from the cancer itself. Given the increasing evidence of importance of the microenvironment for tumor invasion, progression and metastasis, we explored tumor stroma gene signature using microarrays. Methods: Four formalin-fixed paraffin embedded (FFPE) colon cancer specimen carrying a pathological stage of T3-4/N0-2 were retrieved. Tumor stroma and normal stroma were separated using microdissection technology. Random sampling was used to minimize sampling bias. Total RNA was extracted, amplified, and labeled using Nugene FFPE kit, with array analysis using Affymetrix Human Gene 1.0. Eight samples, four normal stroma and four tumor stroma, were analyzed and compared. Array data were balanced and analyzed using standard software. To identify gene signature differences in tumor vs normal stroma, ComparativeMarker analysis and unsupervised cluster analysis were carried out. Results: We identified a 969 Affymetrix probe set as a signature that is highly expressed in tumor stroma. The top 117 genes were further analyzed to carry out a pathway analysis. We found a strong signature evident in tumor stroma, and much of this signature comprised the genes of the extracellular matrix. The pathway analysis revealed evidence of the generalized IGF1/TGFbeta/CTGF/activin mediated effect on the stroma, raising the possibility that some of this derives from, or is accompanied by, angiotensin receptor signaling. Through literature search, we found that several upregulated genes (e.g., FAP) were reported to be associated with stroma activation in vitro and in vivo. Conclusions: In this study, we successfully applied microarrays to study reactive colon tumor stroma in FFPE samples. We identified a specific gene signature reflecting stromal reaction to tumor invasion. We further identified the potential pathway that was activated in the reactive tumor stroma. We provide evidence that microarrays are useful in stroma analysis and may help identify new stromal pathways with potential diagnostic and therapeutic value. No significant financial relationships to disclose.
APA, Harvard, Vancouver, ISO, and other styles
28

Hess, Julia, Kristian Unger, Cornelius Maihoefer, Lars Schüttrumpf, Peter Weber, Sebastian Marschner, Ludmila Wintergerst, et al. "Integration of p16/HPV DNA Status with a 24-miRNA-Defined Molecular Phenotype Improves Clinically Relevant Stratification of Head and Neck Cancer Patients." Cancers 14, no. 15 (July 31, 2022): 3745. http://dx.doi.org/10.3390/cancers14153745.

Full text
Abstract:
Human papillomavirus (HPV)-driven head and neck squamous cell carcinomas (HNSCC) generally have a more favourable prognosis. We hypothesized that HPV-associated HNSCC may be identified by an miRNA-signature according to their specific molecular pathogenesis, and be characterized by a unique transcriptome compared to HPV-negative HNSCC. We performed miRNA expression profiling of two p16/HPV DNA characterized HNSCC cohorts of patients treated by adjuvant radio(chemo)therapy (multicentre DKTK-ROG n = 128, single-centre LMU-KKG n = 101). A linear model predicting HPV status built in DKTK-ROG using lasso-regression was tested in LMU-KKG. LMU-KKG tumours (n = 30) were transcriptome profiled for differential gene expression and miRNA-integration. A 24-miRNA signature predicted HPV-status with 94.53% accuracy (AUC: 0.99) in DKTK-ROG, and 86.14% (AUC: 0.86) in LMU-KKG. The prognostic values of 24-miRNA- and p16/HPV DNA status were comparable. Combining p16/HPV DNA and 24-miRNA status allowed patient sub-stratification and identification of an HPV-associated patient subgroup with impaired overall survival. HPV-positive tumours showed downregulated MAPK, Estrogen, EGFR, TGFbeta, WNT signaling activity. miRNA-mRNA integration revealed HPV-specific signaling pathway regulation, including PD−L1 expression/PD−1 checkpoint pathway in cancer in HPV-associated HNSCC. Integration of clinically established p16/HPV DNA with 24-miRNA signature status improved clinically relevant risk stratification, which might be considered for future clinical decision-making with respect to treatment de-escalation in HPV-associated HNSCC.
APA, Harvard, Vancouver, ISO, and other styles
29

Zhang, Boxi, Ali Elmabsout, Hazem Khalaf, Vladimir T. Basic, Kartheyaene Jayaprakash, Robert Kruse, Torbjörn Bengtsson, and Allan Sirsjö. "The periodontal pathogen Porphyromonas gingivalis changes the gene expression in vascular smooth muscle cells involving the TGFbeta/Notch signalling pathway and increased cell proliferation." BMC Genomics 14, no. 1 (2013): 770. http://dx.doi.org/10.1186/1471-2164-14-770.

Full text
APA, Harvard, Vancouver, ISO, and other styles
30

Krause, Daniela S., Keertik Fulzele, Andre Catic, Michael Hurley, Sanon Lezeau, Robert Hasserjian, Joy Wu, et al. "Parathyroid Hormone-Induced Modulation of the Bone Marrow Microenvironment Reduces Leukemic Stem Cells in Murine Chronic Myelogenous-Leukemia-Like Disease Via a TGFbeta-Dependent Pathway." Blood 118, no. 21 (November 18, 2011): 1670. http://dx.doi.org/10.1182/blood.v118.21.1670.1670.

Full text
Abstract:
Abstract Abstract 1670 It is known that osteoblastic cells regulate the normal hematopoietic stem cell (HSC) niche and control its size. Parathyroid hormone (PTH) is an important regulator of osteoblasts and osteoclasts maintaining calcium homeostasis and, additionally, increasing HSC number in transplant recipients and protecting HSCs from repeated exposure to cytotoxic chemotherapy. We, therefore, hypothesized, that PTH-treatment may allow normal HSCs to outcompete leukemic stem cells (LSCs) in a murine model of chronic myelogenous leukemia. Mice with osteoblastic cell-specific constitutive activation of the receptor for PTH and PTH-related protein (Col1-caPPR mice) are characterized by activation of osteoblastic cells and increases in osteoclast and osteoblast number, trabecular bone, bone turnover and cortical porosity. Col1-caPPR mice have significantly prolonged survival and reduced leukemic mortality compared to wildtype (wt) littermates in a murine retroviral transduction/transplantation model of BCR-ABL1-induced CML-like disease (p=0.002) and B-cell acute lymphoblastic leukemia (B-ALL) (p=0.0004). However, a leukemogenic fusion transcription factor, MLL-AF9, known to cause acute myeloid leukemia in this model, led to more rapid death in the Col1-caPPR recipients compared with their wt counterparts (p<0.0001), indicating that the increased survival of Col1-caPPR recipients is specific for BCR-ABL1-induced leukemia. Continuous infusion of human PTH(1–34) into wt mice with BCR-ABL1-induced CML led to a statistically significant decrease in spleen weights and decreased bone marrow infiltration by BCR-ABL+ cells. Limiting dilution secondary transplantation of BM cells from saline- or PTH-treated primary animals with fully established CML into wt recipients revealed a 15-fold reduction of LSCs in a PTH-treated environment. Secondary mice who received BM from saline-treated donors had an overall survival that was 1/4 that of recipients of marrow from a PTH-treated BM microenvironment. Transforming growth factor beta-1 (TGFβ-1), whose largest and most concentrated tissue source is bone, was increased in the bones of Col1-caPPR mice. TGFβ-1 significantly decreased the in-vitro growth of the BCR-ABL+ cell line K562, but not the MLL-AF9+ cell line THP-1 suggesting that TGFβ-1, increased in the bone marrow microenvironment of Col1-caPPR mice, may be actively suppressing the growth of the BCR-ABL+ diseases, but not of MLL-AF9+ AML. Conversely, blockade of TGFβ-1, -2, and -3 by anti- TGFβ antibody treatment increased the incidence of CML in Col1-caPPR mice from 50% to 75%. Knockdown of TGF Receptor I in transplanted BCR-ABL+ BM in the CML model increased the percentage of BCR-ABL+ myeloid cells in peripheral blood in wt and, more strikingly, in Col1-caPPR recipient mice and increased the overall incidence of CML in Col1-caPPR mice. These results argue that reduction in TGFβ-1 signaling may rescue the CML phenotype in Col1-caPPR mice. In conclusion, these studies suggest that modulation of the bone marrow microenvironment by PTH reduces the frequency of LSCs in CML, possibly by suppression of LSCs via TGFβ-1. Consequently, a clinical trial on the combined use of imatinib and PTH in patients with CML has been initiated at our institution. Disclosures: No relevant conflicts of interest to declare.
APA, Harvard, Vancouver, ISO, and other styles
31

Ellenrieder, Volker, Sandra F. Hendler, Claudia Ruland, Thomas Seufferlein, Wolfgang Boeck, Guido Adler, and Thomas M. Gress. "Evidence for a cross-talk between the TGFbeta-signalling cascade and the RAS/MEK/MAPK-pathway in mediating transdifferentiation and invasion of pancreatic cancer cells." Gastroenterology 118, no. 4 (April 2000): A531. http://dx.doi.org/10.1016/s0016-5085(00)84255-9.

Full text
APA, Harvard, Vancouver, ISO, and other styles
32

Chang, Seon Hee, Heon Park, and Chen Dong. "Act1 adaptor protein is an immediate and essential signaling component of interleukin-17 receptor (B52)." Journal of Immunology 178, no. 1_Supplement (April 1, 2007): LB11. http://dx.doi.org/10.4049/jimmunol.178.supp.b52.

Full text
Abstract:
Abstract Interleukin (IL)-17, the founding member of the IL-17 cytokine family, is the hallmark of a novel subset of CD4+ T cells that is regulated by TGFbeta, IL-6, and IL-23. IL-17 plays an important role in promoting tissue inflammation in host defense against infection and in autoimmune diseases. Although IL-17 has been reported to regulate the expression of proinflammatory cytokines, chemokines, and matrix metalloproteinases, the signaling mechanism of IL-17 receptor has not been understood. An earlier study found that IL-17 activates NF-kappaB and MAPK pathways and requires TRAF6 to induce IL-6. However, it is unknown what molecule(s) directly associates with IL-17 receptor to initiate the signaling. We demonstrate here that IL-17 receptor family shares sequence homology in their intracellular region with Toll-IL-1 receptor (TIR) domains and with Act1, a novel adaptor previously reported as an NF-kappaB activator. MyD88 and IRAK4, downstream signaling components of TIR, are not required for IL-17 signaling. On the other hand, Act1 and IL-17 receptor directly associate likely via homotypic interaction. Deficiency of Act1 in fibroblast abrogates IL-17-induced cytokine and chemokine expression, as well as the induction of C/EBPbeta, C/EBPdelta, and IkappaBzeta. Also, absence of Act1 results in a selective defect in IL-17-induced activation of NF-kappaB pathway. These results thus indicate Act1 as a membrane-proximal adaptor of IL-17 receptor with an essential role in induction of inflammatory genes. Our study not only for the first time reveals an immediate signaling mechanism downstream of an IL-17 family receptor but also has implications in therapeutic treatment of various immune diseases
APA, Harvard, Vancouver, ISO, and other styles
33

Carniti, Cristiana, Silvia Gimondi, Davide Lucini, Jacopo Mariotti, Anisa Bermema, Antonio Vendramin, Mara Morelli, Chiara Formica, and Paolo Corradini. "Noninvasive Prediction of Acute Graft-Verus-Host Disease Following Allogeneic Hematopoietic Stem Cell Transplantation by Circulating miRNA Profiling." Blood 118, no. 21 (November 18, 2011): 311. http://dx.doi.org/10.1182/blood.v118.21.311.311.

Full text
Abstract:
Abstract Abstract 311 Background: Acute graft-versus-host disease (aGVHD) results in significant morbidity and mortality and remains the main complication of alloHSCT. Noninvasive, diagnostic and prognostic tests for aGVHD are currently lacking but essential to predict GVHD and to improve the safety and accessibility of alloHSCT. We hypothesized that the prospective analysis of miRNA expression profile in the plasma of allografted patients could allow for the detection of specific miRNAs with predictive role for aGVHD. Methods: After informed consent, we collected plasma samples from 10 healthy donors and 22 patients (median age: 59 and 41 years) who received unmanipulated alloHSCT (18 from Matched Unrelated Donors and 4 from HLA-matched siblings). Blood samples were collected weekly after HSCT and patients were monitored to assess aGVHD onset. MicroRNAs were isolated from the plasma and the miRNA expression profile examined using a quantitative PCR-method (TaqMan® Human microRNA Cards, Applied Biosystems). The results obtained were subsequently validated with specific miRNA Single Assays (Applied Biosystems). To verify whether the miRNAs emerged from the human studies represent markers of aGVHD and provide information regarding the involvement of specific target organs, a major histocompatibility complex (MHC) mismatched HSCT mouse model was used. Recipient BALB/c mice were lethally irradiated and treated either with spleen and bone marrow (BM) cells from C57BL/6 (B6) donors (GVHD cohort, n=22) or with BM cells only (negative control, n=18). Syngeneic transplants (B6àB6, n=6 were also included. Mice were characterized for GVHD onset by monitoring overall survival and weight loss. Recipient mice were sacrificed and tissues harvested on day 9, 14 and 18 post transplant and GVHD confirmed by histology and scored according to Foley et al, 2008. MiRNAs expression profile have been characterized in the plasma, skin, liver, colon and lymphocytes of GVHD and non-GVHD control cohorts Results: Three of 22 patients developed intestinal GVHD (grade 2) and 9 of 22 patients developed cutaneous GVHD (grade 2–3). By comparing the circulating miRNAs expression profiles of GVHD patients and non GVHD patients, we identified a group of 8 differentially expressed miRNAs (miR-136, 194, 203, 367, 148b, 196b, 26a, 340) (p<0.05). On day 14 post transplant mice in the GVHD cohort developed GVHD as confirmed by organ evaluation (GVHD score: 14–15 out of 20) and weight loss while the control group had no GVHD (GVHD score 0–1 out of 20). The analysis of circulating miRNAs in mice supported the results in humans: the 8 circulating microRNA signature could clearly predict those subjects developing GVHD (area under the ROC curve ≥ 0.75). Of note, pathway enrichment analysis performed using DIANA-mirPath software on the gene targets predicted by microT-4.0, indicate that these miRNAs regulate critical pathways of GVHD pathogenesis (TGFbeta and cytokine signaling, T and B cell differentiation and proliferation, immune response). The analysis of the miRNAs expression profiles of the lymphocytes and of histologically confirmed skin, liver and colon biopsies of GVHD mice highlighted the presence of several deregulated miRNAs when compared to control samples. Pathway enrichment analysis indicated that the differentially expressed miRNAs mainly target genes involved in the TGFbeta and Wnt signaling pathways as well as in the cytoskeleton rearrangements. Of interest, when analysing the expression of the 8 circulating miRNAs able to discriminate GVHD samples from controls, in the organs of mice developing GVHD, miR-203 is also abundant in the skin and colon, miR-367 in the colon and miR-136 in the lymphocytes. These findings strengthen a role for these miRNAs in the modulation of aGVHD and suggest that the presence of plasma miRNAs is linked to the specific organs target of this pathological process. Conclusions: Considering the noninvasive characteristics of plasma sampling and the reproducible and easy detection of miRNAs, our results indicate that circulating miRNAs might represent a promising tool for the early diagnosis of aGVHD thus enhancing therapeutic success and increasing life expectancy of allografted patients. In addition the miRNA profiling of the target organs and lymphocytes of GVHD mice, allowed the identification of several deregulated genes that might play a role in the modulation of aGVHD and warrant further investigations. Disclosures: No relevant conflicts of interest to declare.
APA, Harvard, Vancouver, ISO, and other styles
34

Tabe, Yoko, Linhua Jin, Yasuhito Hatanaka, Hiromichi Matsushita, Saiko Kazuno, Tsutomu Fujimura, Takashi Ueno, et al. "Integrative Genomic and Proteomic Analysis Of Low-Dose ionizing Irradiation Effects On Bone Marrow Stromal Microenvironment and On Survival Of Pre-Leukemic Cells." Blood 122, no. 21 (November 15, 2013): 3775. http://dx.doi.org/10.1182/blood.v122.21.3775.3775.

Full text
Abstract:
Abstract Ionizing radiation is known to induce remodeling of stromal microenvironment and enhance cancer progression. In this study, we investigated the molecular alterations of low-dose ionizing radiation (LDIR) induced non-targeted/bystander responses which affect a complex interplay of stromal cells and pre-leukemic cells in the bone marrow (BM) microenvironment. As a model of BM stromal cells and pre-leukemic cells, we utilized primary BM-derived stromal cells (MSCs) and the Epstein- Barr virus (EBV) infected and immortalized pre-leukemic B-lymphocyte cell line (EBV-B). LDIR (100 mGy, 4MV X ray from a LINAC) caused cell growth inhibition and moderate apoptosis induction in MSCs (viable cells % of control; 75.8 ± 2.4, specific SubG1 % 7.1 ± 0.8 at 24 h) but not in EBV-B cells. We further observed persistent upregulation of p21 mRNA (p< 0.001, RQ-PCR) after acute low-dose irradiation in MSCs but not in EBV-B cells, suggesting radiation induced senescence-like changes in MSCs. In EBV-B cells co-cultured with MSCs, low-dose irradiation induced moderate cell growth inhibition (viable cells % of control; 81.3 ± 6.5) without significant apoptosis induction. To gain insights into the molecular changes induced by LDIR in both, MSC and EBV-B cells we utilized genomic and proteomic analyses. To exclude possible contamination of MSCs, we confirmed negative expression of CD90 mRNA in the tested EBV-B samples. We first screened up to 28,869 genes by cDNA microarray (Affymetrix) and performed functional network analysis by MetaCore (GeneGo). LDIR induced upregulation of 48 genes and the downregulation of 45 (i.e., > 1.3-fold regulation) with prominent stimulation of cell adhesion pathways in MSCs. Of note, 31 of 48 up-regulated genes were small nucleolar RNAs. In EBV-B cells, LDIR upregulated 69 genes/downregulated 130 genes with significant stimulation of the TGFbeta dependent induction of epithelial-mesenchymal transition (EMT) pathways. In EBV-B cells co-cultured with MSCs, LDIR induced immune response signaling along with integrin-mediated cell-matrix adhesion pathway with 42 genes upregulation / 34 genes downregulation. Up-regulation of inflammatory IL8 mRNA (2.0±0.03 fold, p<0.001) by LDIR were further detected in EBV-B cells co-cultured with MSCs. Since ionizing radiation is known to change levels of specific microRNA depending on cell type, we investigated the changes of four microRNA, let-7a, miR-16, miR-19b and miR-21 by RT-PCR. After acute LDIR, all of the tested miRNAs were upegulated in EBV-B cells cultured alone, but downregulated in EBV-B cells co-cultured with MSCs. These changes were accompanied by the coordinate modulation of their common target GPAM mRNA, a key enzyme of phospholipids, and ribozyme CPEB3 mRNA. Indeed, proteomic analysis by isobaric tags for relative and absolute quantitation (iTRAQ, Applied Biosystems) detected the upregulation of Protein kinase C delta (p = 0.04) which is strictly dependent on the presence of phospholipids. iTRAQ detected more than 1500 proteins in MSCs and EBV-B cells. In MSCs LDIR resulted in 32 upregulated proteins and 1 downregulated protein (p < 0.05) with the activation of focal adhesion (6 proteins, p < 0.001) and apoptosis/senescence pathways (three proteins, p = 0.02), concordant with cDNA microarray data. In EBV-B cells, LDIR induced upregulation of 47 proteins including 19 ribosome related proteins (40%) and downregulation of 19 proteins including 6 apoptosis/senescence related proteins, suggesting an activation of molecular repair mechanisms from LDIR-triggeredstress. In contrast, under MSC co-culture conditions LDIR downregulated 25 of ribosomal proteins (74% of the 34 downregulated proteins) in EBV-B cells, indicating suppression of the ribosomal biogenesis and translational activity, which might cause the observed cell quiescence with cell cycle arrest of EBV-B cells mediated by the low-dose irradiated neighboring MSCs. In summary, we have demonstrated the LDIR effects on BM-derived stromal cells and pre-leukemic cells under MSC co-culture conditions mimicking the BM microenvironment. We conclude that LDIR may support pre-malignant cells survival via direct activation of TGFbeta dependent EMT pathways, which contribute to antiapoptosis, and via interactions with irradiated neighboring BM stromal cells which promote quiescence and survival of pre-leukemic cells. Disclosures: No relevant conflicts of interest to declare.
APA, Harvard, Vancouver, ISO, and other styles
35

Daley, Jessica D., Elina Mukherjee, A. Carolina Tufino, Riyue Bao, and Kelly M. Bailey. "Abstract 2641: Transcriptional reprogramming of Ewing tumors: gains and losses noted when utilizing immunocompetent in vivo models." Cancer Research 84, no. 6_Supplement (March 22, 2024): 2641. http://dx.doi.org/10.1158/1538-7445.am2024-2641.

Full text
Abstract:
Abstract Objective: Ewing sarcoma is an aggressive cancer most frequently diagnosed in adolescents and young adults. Currently, there is a limited understanding of the immunobiology of Ewing sarcoma, including limited pre-clinical studies of immunotherapeutic interventions. This, in part, is due to the historical lack of an immuno-competent murine model. Here, we utilize an immunocompetent, humanized mouse model to investigate the Ewing tumor immune microenvironment. Specifically, we sought to determine the transcriptional differences that occur when comparing human Ewing tumors established in a humanized backdrop versus those developed in an NSG (immuno-deficient) model. Methods: Human Ewing cell lines (A673, TC32, TC71) and PDX (PSaRC219) were injected orthotopically into humanized or NSG mice. After ~3 weeks, bulk RNA sequencing was performed on tumors utilizing an Illumina Next Generation Sequencer. Tumor single-cell suspensions underwent immunophenotyping by flow cytometry (CyTek Aurora) for the following human markers: CD45, CD3, CD4, CD8, CD56, CD14, CD16, etc.. Peripheral blood was collected, PBMCs isolated, and immunophenotyping was again performed to determine peripheral blood immune cell populations. Results: Ewing tumors established in humanized mice demonstrate infiltration of human CD45+ cells as well as immune cell sub-populations (CD3+, CD14+, CD56+ cells). Bulk RNA sequencing analysis utilizing unsupervised hierarchal clustering revealed that the transcriptional profile of Ewing tumors generated in humanized vs NSG mice cluster distinctly. Pathway analysis of differentially expressed genes (p &lt;0.05) in Ewing tumors developed in humanized versus NSG mice demonstrates upregulation of pathways including extracellular matrix remodeling, collagen biosynthesis, and Wnt signaling. Notably, transcripts known to be specific to TGFBeta-dependent signaling in Ewing tumors are upregulated in the immune competent model, including TGFB1, FBLN5, and COLEC12. Conclusion: This work demonstrates the Ewing tumor transcriptional remodeling that occurs when tumors are developed in an immunocompetent in vivo backdrop. Upregulation of several pro-metastatic pathways were noted. Ongoing work focuses on testing drug response differences between Ewing tumors developed in humanized versus NSG backdrops and pursuing immunocompetent Ewing models as a component of preclinical agent testing. Citation Format: Jessica D. Daley, Elina Mukherjee, A Carolina Tufino, Riyue Bao, Kelly M. Bailey. Transcriptional reprogramming of Ewing tumors: gains and losses noted when utilizing immunocompetent in vivo models [abstract]. In: Proceedings of the American Association for Cancer Research Annual Meeting 2024; Part 1 (Regular Abstracts); 2024 Apr 5-10; San Diego, CA. Philadelphia (PA): AACR; Cancer Res 2024;84(6_Suppl):Abstract nr 2641.
APA, Harvard, Vancouver, ISO, and other styles
36

Shapira, I., R. Huffman, E. Neculiseanu, A. Banavali, K. Guddati, A. Shih, C. Mason, and A. Lee. "P2: SNP ANALYSIS IN BRCA POSITIVE AND BRCA NEGATIVE SUBJECTS WITH AND WITHOUT BREAST CANCER (BRCA) REVEAL CENTRAL ROLE OF ALK SNPS AND TGFBETA SUPERFAMILY IN MALIGNANT TRANSFORMATION." Journal of Investigative Medicine 64, no. 3 (February 25, 2016): 817.3–818. http://dx.doi.org/10.1136/jim-2016-000080.42.

Full text
Abstract:
Purpose of StudyOver 240,000 individuals are diagnosed with breast cancer each year in the USA. Outcomes depend on DNA deregulations in tumors. Carriers of deleterious BRCA1 and BRCA2 mutations are predisposed to 30 fold higher lifetime risks of breast and ovarian cancer.Aims:1. To check for differences in SNPs of genomic DNA obtained in BRCA+/− with and without BrCa.2. Analyze correlates of molecular mechanisms occurring in BRCA mutant patients.Methods UsedWe analyzed 94 subjects (41 BRCA positive) with or without BrCa to detect SNPs whose expression is significantly differentially expressed between breast cancer and controls. DNA samples were extracted from PBMCs. Samples were measured for DNA concentration using an Invitrogen QuBit Fluorometer, and diluted to 50 ng/µL.All samples were collected between 2010 and 2014 and survival data was known in all cancer patients. Processed samples were sequenced using an Illumina MiSeq Sequencer with a 300 cycle kit to detect SNPs. Variant Call Files were analyzed in Microsoft Excel using Fisher's Exact Test.Summary of ResultsALK SNPs were commonly found in cancer relative to control. Significant associations of ALK SNPs were seen in BRCA mutation subjects. ALK protein was overexpressed in 47% of BRCA mutations cases, which was significantly higher than in non-BRCA cases. Our results show that the ALK signaling pathway possibly is more common in early onset of breast cancer as seen with BRCA mutations. Coremine analysis showed SNPs identified in cancer were most commonly associated with deregulation of Transforming Growth Factor-Beta Superfamily protein synthesis and binding function.ConclusionsDifferences in the associations of the modifying polymorphisms with BrCarisk for BRCA1 and BRCA2 mutation carriers are likely to reflect differences in the biology of tumor development in these two groups of women at high risk of breast cancer. The identification of modifying polymorphisms could therefore lead to a better understanding of the etiology of tumors in mutation carriers and also to the development of effective and more specific therapies for BrCa in mutation carriers.
APA, Harvard, Vancouver, ISO, and other styles
37

Nagel, Stefan, Letizia Venturini, Grzegorz K. Przybylski, Piotr Grabarczyk, Corinna Meyer, Maren Kaufmann, Karin Battmer, et al. "NK-Like Homeodomain Proteins Regulate NOTCH3-Signaling in T-Cells." Blood 112, no. 11 (November 16, 2008): 1194. http://dx.doi.org/10.1182/blood.v112.11.1194.1194.

Full text
Abstract:
Abstract Three NK-like (NKL) homeobox genes, TLX1/HOX11, TLX3/HOX11L2 and NKX2- 5/CSX, have been implicated in T-cell acute lymphoblastic leukemia (T-ALL). Here we screened further NKL genes in 24 T-ALL cell lines by RT-PCR and identified common expression of MSX2, highlighting this homeobox gene as a potential physiological family member in T-cells. Subsequent quantification of MSX2 confirmed expression in primary hematopoietic cells demonstrating higher levels in CD34+ stem cells when compared to peripheral blood cells or mature CD3+ T-cells. Analysis of core thymic factors in T-ALL cell lines, including IL7, BMP4, TGFbeta, NOTCH and T-cell receptor signaling, suggests their involvement in MSX2 regulation during T-cell differentiation. Chromosomal and genomic analysis of the MSX2 locus (at 5q35) uncovered deletion in t(5;14)(q35;q32) positive T-ALL cell lines associated with low expression levels of MSX2 and ectopic activation of TLX3 or NKX2-5, respectively. For functional analysis we lentivirally transduced T-ALL cells for overexpression of either MSX2 or oncogenic TLX1 and NKX2-5. These cells displayed transcriptional activation of NOTCH3-signaling, as indicated by expression array profiling and real-time PCR analysis of NOTCH3, HES1 and HEY1. The sensitivities to gamma-secretase inhibitor analyzed by MTT-assay of cells overexpressing MSX2, TLX1 or NKX2-5, respectively, were consistently decreased. Furthermore, in addition to MSX2, both TLX1 and NKX2-5 proteins interacted with repressor proteins of the NOTCH-pathway, SPEN/MINT and TLE1/GRG1, as shown by co-immunoprecipitation, probably representing one mechanism of (de)regulation. Elevated expression of NOTCH3 and HEY1 mRNA was detected in TLX1/3 positive T-ALL patients, confirming data obtained from cell lines. In conclusion, we have defined expression patterns, regulation and targets of MSX2 in hematopoietic cells, to reveal a novel modulatory activity in T-cell differentiation operating via NOTCH-signaling, and in leukemogenesis when replaced or supplemented by oncogenic NKL homeodomain proteins.
APA, Harvard, Vancouver, ISO, and other styles
38

Labbe, E., A. Letamendia, and L. Attisano. "Cooperative signalling by TGFbeta and Wnt signalling pathways." Biochemical Society Transactions 28, no. 5 (October 1, 2000): A272. http://dx.doi.org/10.1042/bst028a272c.

Full text
APA, Harvard, Vancouver, ISO, and other styles
39

Hope, Chelsea, Ellen Hebron, Jaehyup Kim, Jeff Jensen, Claire Flanagan, Neehar Bhatia, Catherine Leith, et al. "Molecular Pathways That Determine the Activation State of Macrophages within the Myeloma Niche." Blood 120, no. 21 (November 16, 2012): 443. http://dx.doi.org/10.1182/blood.v120.21.443.443.

Full text
Abstract:
Abstract Abstract 443 Benefit from cytotoxic therapy in myeloma may be limited by persistence of residual tumor cells nested within favorable niches. However, the contribution of macrophages to the regulation of myeloma niches is still incompletely understood. We have previously shown that macrophages provide growth and anti-apoptotic signals to myeloma cells when grown in co-culture. These results prompted us to investigate the regulation of primary monocytes/macrophages that reside within myeloma niches in the bone marrow. Unmanipulated CD14+ monocytic cells, freshly explanted from myeloma bone marrows, displayed a pre-dominantly pro-inflammatory transcriptomic profile when compared to normal monocytes. We found enhanced transcription of genes encoding pro-inflammatory mediators, known to support growth and survival of myeloma cells, such as TNFalpha, IL-1beta, IL-6, IL-8 and TACI. Downregulation of TGFbeta was also consistent with a pro-inflammatory (M1) signature. Interestingly, we also found concurrent transcription of some genes characteristic of “alternative macrophage activation” (M2 phenotype) such as IL-10 and IL1-receptor antagonist (IL-1RN). These results suggest that myeloma-associated macrophages, while being predominantly pro-inflammatory, display significant plasticity between the M1-M2 phenotypic extremes. To obtain insights into the underlying mechanisms, we examined the role of TPL2 (Cot, MAP3K8), a serine/threonine kinase with central and non-redundant roles in regulating innate immune responses in macrophages following activation by Toll receptor (TLR) ligands and members of the TNF ligand superfamily. In myeloma-associated macrophages, we found constitutive activation of a TPL2 kinase-dependent, ERK-mediated pathway that promotes synthesis and processing of pro-inflammatory cytokines, including TNFalpha and IL-1beta. We also discovered constitutive activation of AKT at Ser473, a site dependent for its phosphorylation on TPL2 activity in macrophages responding to TLR signaling. Notably, the Akt/mTOR pathway limits the magnitude and duration of macrophage activation, in part through synthesis of the anti-inflammatory cytokine IL-10. These events involve signaling through STAT3. Accordingly, we discovered constitutive phosphorylation of STAT3 at a site regulated by TPL2 in activated macrophages. In addition to non-tumor cell autonomous roles in regulating myeloma through macrophages, we showed a tumor cell-autonomous, growth-regulatory role of TPL2 kinase. Treatment of myeloma cells with a TPL2 small molecule inhibitor resulted in apoptosis that was not rescued by the presence of patient-derived stromal cells. We postulate that TPL2 inhibition interferes with growth signaling in myeloma cells because TPL2 has been shown to substitute for RAF proteins in growth signal transduction. Interestingly, we found that TPL2 was activated by phosphorylation as cells entered G2/M. Treatment with nocodazole increased the proportion of cells that co-expressed phosphorylated TPL2 and phosphorylated histone H3. Moreover, we found that TPL2 activity was required for MAPK pathway signal transduction in response to TNF receptor stimulation in myeloma cells. Taken together, our results provide important novel insights into the regulation of macrophages within primary myeloma niches in the bone marrow. Plasticity between M1 and M2 phenotypes may correlate tightly with the actions of TPL2 kinase. In the myeloma niche, TPL2 activity helps to fine-tune macrophage activation by promoting synthesis and release of pro-inflammatory cytokines required by the myeloma tumor cell while engaging counter-regulatory mechanisms to prevent tissue destruction mediated by activated macrophages. Additionally, we described a growth regulatory role of TPL2 in the tumor cell itself. Thus, TPL2 blockade may disrupt crucial macrophage-tumor cell interactions within myeloma niches. Disclosures: No relevant conflicts of interest to declare.
APA, Harvard, Vancouver, ISO, and other styles
40

Faure, S., M. A. Lee, T. Keller, P. ten Dijke, and M. Whitman. "Endogenous patterns of TGFbeta superfamily signaling during early Xenopus development." Development 127, no. 13 (July 1, 2000): 2917–31. http://dx.doi.org/10.1242/dev.127.13.2917.

Full text
Abstract:
Transforming growth factor beta (TGFbeta) superfamily signaling has been implicated in patterning of the early Xenopus embryo. Upon ligand stimulation, TGFbeta receptors phosphorylate Smad proteins at carboxy-terminal SS(V/M)S consensus motifs. Smads 1/5/8, activated by bone morphogenetic protein (BMP) signaling, induce ventral mesoderm whereas Smad2, activated by activin-like ligands, induces dorsal mesoderm. Although ectopic expression studies are consistent with roles for TGFbeta signals in early Xenopus embryogenesis, when and where BMP and activin-like signaling pathways are active endogenously has not been directly examined. In this study, we investigate the temporal and spatial activation of TGFbeta superfamily signaling in early Xenopus development by using antibodies specific for the type I receptor-phosphorylated forms of Smad1/5/8 and Smad2. We find that Smad1/5/8 and two distinct isoforms of Smad2, full-length Smad2 and Smad2(delta)exon3, are phosphorylated in early embryos. Both Smad1/5/8 and Smad2/Smad2(delta)exon3 are activated after, but not before, the mid-blastula transition (MBT). Endogenous activation of Smad2/Smad2(delta)exon3 requires zygotic transcription, while Smad1/5/8 activation at MBT appears to involve transcription-independent regulation. We also find that the competence of embryonic cells to respond to TGF(delta) superfamily ligands is temporally regulated and may be a determinant of early patterning. Levels of phospho-Smad1/5/8 and of phospho-Smad2/Smad2(delta)exon3 are asymmetrically distributed across both the animal-vegetal and dorsoventral axes. The timing of the development of these asymmetries differs for phospho-Smad1/5/8 and for phospho-Smad2/Smad2(delta)exon3, and the spatial distribution of phosphorylation of each Smad changes dramatically as gastrulation begins. We discuss the implications of our results for endogenous functions of BMP and activin-like signals as candidate morphogens regulating primary germ layer formation and dorsoventral patterning of the early Xenopus embryo.
APA, Harvard, Vancouver, ISO, and other styles
41

Price, Colles O., Ping Chen, Zejuan Li, Yuanyuan Li, Anissa Wiley, Chunjiang He, Hao Huang, et al. "MLL-Rearrangements Result in Upregulation of Mir-9 and Subsequent Inhibition of the Tumor Suppressor TGFBI." Blood 118, no. 21 (November 18, 2011): 2453. http://dx.doi.org/10.1182/blood.v118.21.2453.2453.

Full text
Abstract:
Abstract Abstract 2453 Acute Leukemia represents one of the most deadly cancers in the United States. Clinical treatments in leukemia have progressed significantly through the use of therapies targeted specifically to chromosomal translocations. The success of these therapies has provided a model for future treatment in various cancers. However, there are various subtypes of leukemia where five-year survival and relapse rates have poor clinical outcome, indicating that new therapies are needed. A particular leukemia subtype, namely mixed lineage leukemia (MLL)-rearranged leukemia that is a result of chromosomal rearrangements leading to fusions between MLL and partner genes, is associated with a dismal outcome. Therapeutic targeting of MLL rearrangements has proven challenging as there have been dozens of described rearrangements. We have performed both messenger RNA (mRNA) and microRNA (a class of small non-coding RNA) microarray analyses on over 100 leukemia patient samples and have identified that microRNA-9 (miR-9) is highly upregulated in MLL-associated leukemias. Through correlating expression of miR-9 and those of its predicted target genes, we identified TGFbeta-induced protein (TGFBI) as a potential target of miR-9 that exhibited a significantly (P<0.05) inverse correlation of expression with miR-9 in acute leukemia. Our further luciferase reporter/mutagenesis assays show that TGFBI is a direct target of miR-9. TGFBI has been described as a tumor suppressor in breast, lung and ovarian tumors through two mechanisms, AKT inhibition and microtubule stabilization, but its involvement in leukemia has never been reported. We found that TGFBI expression was decreased while miR-9 was upregulated in MLL-rearrangement acute myeloid leukemias. Upon further testing we discovered that exogenous expression of miR-9 increased proliferation and enhanced the colony-forming ability of progenitor cells infected with MLL-AF9 fusions. Expression of TGFBI was sufficient to inhibit proliferation and significantly reduced the colony-forming ability of MLL-AF9. This reduction in colony-forming ability was also observed when MLL-AF9 progenitor cells were infected with TGFBI and miR-9 together, showing that TGFBI expression is sufficient to block the effects of miR-9 overexpression. We suspect that TGBFI expression modulates AKT in MLL-rearranged leukemias, thus making it susceptible to combination of standard leukemia chemotherapies with AKT-inhibitors. We will test the effects on NVP-BEZ235, a PI3K/mTOR inhibitor in phase II clinical trials, alone and in combination with other chemotherapies against MLL-rearrangement leukemias. We expect that the specific targeting of this pathway in these leukemias will be highly efficacious. Thus these studies could identify a novel therapeutic strategy for the treatment of MLL-rearrangement leukemias that is more effective and specific than the current therapies. Disclosures: No relevant conflicts of interest to declare.
APA, Harvard, Vancouver, ISO, and other styles
42

Salvi, Amrita, Laura R. Hardy, Kimberly N. Heath, Samantha Watry, Melissa R. Pergande, Stephanie M. Cologna, and Joanna E. Burdette. "Abstract B030: PAX8 modulates the tumor microenvironment of high grade serous ovarian cancer through changes in the secretome." Cancer Research 83, no. 2_Supplement_2 (January 15, 2023): B030. http://dx.doi.org/10.1158/1538-7445.metastasis22-b030.

Full text
Abstract:
Abstract High grade serous ovarian cancer (HGSC), the most common and lethal form of ovarian cancer, is a highly heterogeneous disease. HGSC is rarely detected early, and likely arises from the fimbriated end of the fallopian tube epithelium (FTE), and in some cases, the ovarian surface epithelium (OSE). PAX8 is a commonly used biomarker for ovarian serous tumors and is expressed in ~90% of HGSC. Although the OSE does not express PAX8, murine models of HGSC derived from the OSE acquire PAX8, suggesting that it is not only a marker of Müllerian origin, but also an essential part of cancer progression, potentially from both the OSE and FTE. Our data shows that PAX8 loss by CRISPR and shRNA in HGSC cell lines causes tumor cell death and reduces cell migration and invasion. Additionally, loss of PAX8 significantly reduced tumor burden in a xenograft model of HGSC. Herein, secretome analysis was performed on PAX8 deleted cells, and we identified a reduction of the extracellular matrix (ECM) components, collagen and fibronectin. Immunoblotting and immunofluorescence in PAX8 deleted OVCAR8 HGSC cells further validated the results from the secretome analysis. PAX8 loss reduced the amount of secreted TGFbeta, a cytokine that plays a crucial role in remodeling of the tumor microenvironment. Furthermore, PAX8 loss reduced the integrity of 3D spheroids and caused a reduction of ECM proteins in 3D cultures: fibronectin and collagen. Due to the ubiquitous expression of PAX8 in HGSC, regardless of cell origin, and evidence that reducing PAX8 protein levels inhibits tumor growth, a PAX8 inhibitor could be a promising drug lead against HGSC. To accomplish this, we generated a murine oviductal epithelial (MOE) cell line stably expressing the PAX8 promoter driving luciferase reporter protein. Using this cell line, we performed a screening assay with a library of FDA-approved drugs (Prestwick Library) and quantitatively assessed these compounds for their inhibition of PAX8-luciferase. We identified two hits: losartan and captropril, both inhibitors of the renin-angiotensin pathway that inhibit PAX8 expression and function. We are currently working to monitor if these compounds reduce tumor burden via PAX8 reduction. Further, if PAX8 reduction in vivo diminishes collagen and fibronectin, this may impact immune cell infiltration via changes in the tumor microenvironment. Overall, this study validates PAX8 as a regulator of ECM deposition in the tumor microenvironment. Citation Format: Amrita Salvi, Laura R. Hardy, Kimberly N. Heath, Samantha Watry, Melissa R. Pergande, Stephanie M. Cologna, Joanna E. Burdette. PAX8 modulates the tumor microenvironment of high grade serous ovarian cancer through changes in the secretome [abstract]. In: Proceedings of the AACR Special Conference: Cancer Metastasis; 2022 Nov 14-17; Portland, OR. Philadelphia (PA): AACR; Cancer Res 2022;83(2 Suppl_2):Abstract nr B030.
APA, Harvard, Vancouver, ISO, and other styles
43

Papadas, Athanasios, Gauri Deb, Adam Officer, Chelsea Hope, Philip Emmerich, Alexander Cicala, Joshua Wiesner, et al. "936 Stromal remodeling regulates dendritic cell abundance in the tumor microenvironment." Journal for ImmunoTherapy of Cancer 9, Suppl 2 (November 2021): A982. http://dx.doi.org/10.1136/jitc-2021-sitc2021.936.

Full text
Abstract:
BackgroundStimulatory dendritic cells (SDC), enriched within the Batf3-DC lineage (also known as conventional type 1 DC, cDC1), engage in productive interactions with CD8+ effectors along tumor-stroma boundaries. This puzzling pattern of T-cell-DC localization has been interpreted as ”tumor-exclusion”, limiting anti-tumor immunity. To understand this paradox, we hypothesized that dynamic matrix remodeling at the invasive margin generates unique activation and cell-fate cues critical for Batf3-DC homeostasis.MethodsWe studied immunocompetent tumor models of lung carcinoma, breast carcinoma, melanoma and multiple myeloma. For mechanistic experiments, we generated novel Vcan-targeted models through CRISPR-Cas9 targeting. We delineated DC subsets through multi-parametric flow cytometry and tumor immune contexture through mass cytometry. Batf3-DC cellular models included MutuDC1940 immortalized DC and iCD103 primary cells. TCGA data were mined for human validation.ResultsWe find that CD8+ T cells massively infiltrate tumor matrices undergoing robust matrix proteoglycan versican (VCAN) proteolysis, an essential organ-sculpting modification in development and adult tissue-plane forging. Across 7591 samples from 20 TCGA cancer types, a significant-positive correlation between VCAN substrate expression and Batf3-DC score was observed, suggesting that the VCAN pathway may regulate Batf3-DC across several cancer types. Experimental Vcan depletion in the tumor microenvironment was detrimental for Batf3-DC. Batf3-DC abundance was restored through the VCAN N-terminal fragment (matrikine) versikine, physiologically generated through ADAMTS protease activity in remodeled stroma. In addition to Batf3-DC expansion, versikine resulted in G-MDSC contraction as well as the emergence of an atypical innate lymphoid (NK/ILC1) subset expressing cytotoxicity receptors, low IFNgamma and robust pro-survival GM-CSF. Despite broad intratumoral IRF8 induction (10-100-fold), adoptive transfer of pre-DC into versikine-replete microenvironments did not influence their differentiation choice between Batf3-DC and cDC2. Instead, versikine delivered a distinct Batf3-DC activation signal characterized by non-TLR maturation as well as downregulation of TGFbeta and Wnt signaling. In vivo, versikine promoted Batf3-DC abundance through NK cells but independently of stromal TLR2 or CD44. Versikine sensitized immune-evasive tumors to STING agonist immunotherapy in a Batf3-DC dependent manner and promoted antigen-specific CD8+ responses. Versikine-DC signatures correlated with CD8+ T cell scores in human lung cancers.ConclusionsWe demonstrate that dynamic extracellular matrix remodeling controls Batf3-DC abundance in the tumor microenvironment. N-terminal proteolysis of the matrix proteoglycan versican (VCAN), releases a bioactive fragment (matrikine), versikine, that is remarkably necessary and sufficient for Batf3-DC accumulation. Versikine orchestrates a multi-lineage network that regulates Batf3-DC activation and survival at matrix-remodeling interfaces. Therapeutic harnessing of matrix-Batf3-DC cross-talk sensitizes immune-evasive tumors to immunotherapy.AcknowledgementsWe acknowledge support by the National Cancer Institute (R01CA252937 and U01CA196406), the American Cancer Society (127508-RSG-15-045-01-LIB), the Leukemia and Lymphoma Society (6551–18), the UW Trillium Myeloma Fund and the Robert J. Shillman Foundation.Ethics ApprovalLaboratory animal work was performed under IACUC-approved protocols #M5476 and #S19109 in the University of Wisconsin-Madison and University of California, San Diego respectively.
APA, Harvard, Vancouver, ISO, and other styles
44

Kelley, Todd W., and Olga Efimova. "The Antioxidant n-Acetylcysteine Blocks Direct Regulatory T Cell Mediated Suppression of CD4+ Effector T Cells In Vitro." Blood 116, no. 21 (November 19, 2010): 2765. http://dx.doi.org/10.1182/blood.v116.21.2765.2765.

Full text
Abstract:
Abstract Abstract 2765 Background: CD4+CD25+FOXP3+ regulatory T cells (Tregs) employ a range of suppressive strategies including factors that are cytotoxic to target CD4+CD25−FOXP3− effector T cells (Teff) such as perforin and granzyme B and those that suppress proliferation, differentiation and/or cytokine production including TGF-beta1, IL-10 and CTLA-4. The relative contribution of each of these mechanisms to Treg function is unclear but from the available data their importance appears context specific. Because T cells are very sensitive to the redox state of the microenvironment, and display a pattern of impaired activation under conditions of oxidative stress, we investigated the potential contribution of an oxidant dependent suppressive pathway on direct Treg mediated suppression of Teff in vitro using the antioxidant n-acetylcysteine (NAC) to block reactive oxidants. Methods: Tregs and Teff were derived from the spleens of 2–4 month old C57BL/6 mice maintained in pathogen free conditions. T cell subsets were isolated using magnetic bead based techniques. Purity was assessed by flow cytometry. For suppression assays, Tregs were co-cultured with CFSE-labeled Teff at the indicated ratios and stimulated with anti-CD3/CD28 coated beads for 3 days. Proliferation was measured by flow cytometric evaluation of CFSE dilutional staining. Suppression was calculated by comparing proliferation of Teff cultured alone to those co-cultured with Tregs (% suppression= 1 − Tregs:Teff/Teff alone). In other experiments, CFSE-labeled Teff were cultured alone in the indicated conditions with TGF-beta +/− NAC and proliferation was assessed by CFSE staining. Intracellular reactive oxygen species (ROS) were measured by flow cytometry using the redox sensitive cell permeable indicator dye 5-(and-6)-chloromethyl-2',7'-dichlorodihydrofluorescein diacetate, acetyl ester (DCFDA). Results: The presence of NAC prevented Treg suppression of Teff in a dose dependent fashion at Treg:Teff ratios of 1:2, 1:1 and 2:1 (see figure). Suppression was significantly decreased at 0.1mM NAC (p<0.05 at 1:1; p<0.01 at 2:1) and essentially absent at 1mM NAC. Proliferation of Teff was markedly higher in the setting of 1mM NAC compared to conditions without NAC, even during co-culture with Tregs at a 2:1 ratio of Tregs:Teff. Treg suppression of Teff proliferation was dependent on TGF-beta as neutralizing antibodies reversed the effect (p<0.001). The presence of NAC was sufficient to overcome the suppressive effects of exogenous TGF-beta on CD3/CD28 stimulated Teff proliferation. Treatment of CD3/CD28 stimulated Teff with TGFbeta resulted in a significant dose dependent increase in the levels of intracellular ROS (p<0.0001 at 10ng/mL TGF-beta) that inversely correlated with the degree of proliferation. Conclusion: NAC blocks Treg mediated TGF-beta dependent suppression in vitro. This suggests that TGF-beta may function to suppress proliferation of Teff via a ROS dependent mechanism and raises the possibility that targeted delivery of antioxidants may have clinical utility for modulating the effects of Tregs in vivo. Disclosures: No relevant conflicts of interest to declare.
APA, Harvard, Vancouver, ISO, and other styles
45

Bordonaro, Michael, Shruti Tewari, Wafa Atamna, and Darina L. Lazarova. "The Notch ligand Delta-like 1 integrates inputs from TGFbeta/Activin and Wnt pathways." Experimental Cell Research 317, no. 10 (June 2011): 1368–81. http://dx.doi.org/10.1016/j.yexcr.2011.03.019.

Full text
APA, Harvard, Vancouver, ISO, and other styles
46

Jia, Haixia, Suhua Hao, Meiting Cao, Lifang Wang, Hua Bai, Wen Shui, and Xiaotang Yang. "m6A-Related lncRNAs Are Potential Prognostic Biomarkers of Cervical Cancer and Affect Immune Infiltration." Disease Markers 2022 (April 11, 2022): 1–22. http://dx.doi.org/10.1155/2022/8700372.

Full text
Abstract:
The correlation of m6A-related lncRNAs with the prognosis and immune microenvironment of cervical cancer is not yet clear. In this study, we identified 7 m6A-related prognostic lncRNAs by Pearson correlation and univariate Cox regression analyses based on TCGA-cervical cancer dataset. Then, patients were divided into two clusters by consensus clustering based on the 7 m6A-related prognostic lncRNA expression. Cluster 1 was characterized by survival and stage disadvantage, enrichment of immunosuppressive and carcinogenic activation pathways. Besides, cluster 1 had higher immunosuppressive factor TGFbeta and lower immune cell infiltration compared with cluster 2. According to the expression of 7 m6A-related lncRNA, a 6-m6A-related lncRNA risk score model was established in the training set by LASSO regression analysis. The high-risk group had worse overall survival than the low-risk group. No matter in the training or validation sets, the m6A-related lncRNA risk score was an independent prognostic factor for overall survival. Meanwhile, we validated the independent prognostic value of risk score in the disease-specific survival and progression-free survival by multivariate Cox analysis. The high-risk group was characterized by higher TGFbeta and regulatory T cell and was rich in malignant pathways. Additionally, we also detected and compared the expression levels of four m6A-related prognostic lncRNA in 9 tumor samples and 9 normal tissues using quantitative real-time polymerase chain reaction assay. In conclusion, the novel m6A-related lncRNA risk score is a potential prognostic predictor of cervical cancer patients. These 6 m6A-related lncRNAs might serve as key mediators of the immune microenvironment and represent promising therapeutic targets for improving cervical cancer prognosis.
APA, Harvard, Vancouver, ISO, and other styles
47

Tremblay, K. D., P. A. Hoodless, E. K. Bikoff, and E. J. Robertson. "Formation of the definitive endoderm in mouse is a Smad2-dependent process." Development 127, no. 14 (July 15, 2000): 3079–90. http://dx.doi.org/10.1242/dev.127.14.3079.

Full text
Abstract:
TGFbeta growth factors specify cell fate and establish the body plan during early vertebrate development. Diverse cellular responses are elicited via interactions with specific cell surface receptor kinases that in turn activate Smad effector proteins. Smad2-dependent signals arising in the extraembryonic tissues of early mouse embryos serve to restrict the site of primitive streak formation and establish anteroposterior identity in the epiblast. Here we have generated chimeric embryos using lacZ-marked Smad2-deficient ES cells. Smad2 mutant cells extensively colonize ectodermal and mesodermal populations without disturbing normal development, but are not recruited into the definitive endoderm lineage during gastrulation. These experiments provide the first evidence that TGFbeta signaling pathways are required for specification of the definitive endoderm lineage in mammals and identify Smad2 as a key mediator that directs epiblast derivatives towards an endodermal as opposed to a mesodermal fate. In largely Smad2-deficient chimeras, asymmetric nodal gene expression is maintained and expression of pitx2, a nodal target, is also unaffected. These results strongly suggest that other Smad(s) act downstream of Nodal signals in mesodermal populations. We found Smad2 and Smad3 transcripts both broadly expressed in derivatives of the epiblast. However, Smad2 and not Smad3 mRNA is expressed in the visceral endoderm, potentially explaining why the primary defect in Smad2 mutant embryos originates in this cell population.
APA, Harvard, Vancouver, ISO, and other styles
48

Maxeiner, Hagen, Yaser Abdallah, Christoph Rüdiger Wolfram Kuhlmann, Klaus-Dieter Schlüter, and Sibylle Wenzel. "Effects of cerivastatin on adrenergic pathways, hypertrophic growth and TGFbeta expression in adult ventricular cardiomyocytes." European Journal of Cell Biology 91, no. 5 (May 2012): 367–74. http://dx.doi.org/10.1016/j.ejcb.2011.12.006.

Full text
APA, Harvard, Vancouver, ISO, and other styles
49

Stoika, Rostyslav, Mariya Yakymovych, Serhiy Souchelnytskyi, and Ihor Yakymovych. "Potential role of transforming growth factor beta1 in drug resistance of tumor cells." Acta Biochimica Polonica 50, no. 2 (June 30, 2003): 497–508. http://dx.doi.org/10.18388/abp.2003_3702.

Full text
Abstract:
Acquired drug resistance of tumor cells is frequently observed in cancer patients undergoing chemotherapy. We studied murine leukemia L1210 cells sensitive and resistant to the cytotoxic action of cisplatin and showed that cisplatin-resistant leukemia cells were also refractory to TGF beta1-dependent growth inhibition and apoptosis. Addressing the question about the mechanisms responsible for the cross-resistance to cisplatin and TGF beta1, we found that cisplatin- and TGF beta1-resistant L1210 cells possessed a decreased expression of type I TGF beta1 receptor, while the expression of type II TGF beta1 receptor was not affected. Western blot analysis of Smad proteins 2, 3, 4, 6, and 7, which participate in signal transduction pathway down-stream of the TGF beta1 receptors, revealed an increased expression of Smad 6, inhibiting TGF beta1 action, only in cisplatin- and TGF beta1-resistant L1210 cells. TGF beta1 and especially the cytotoxic mistletoe agglutinin increased Smad 6 expression in TGF beta1-sensitive but not in TGF beta1-resistant L1210 cells. TGF beta1-resistant L1210 cells also differed from TGF beta1-sensitive cells by the lack of expression of the pro-apoptotic p53 protein and higher level of expression of the anti-apoptotic Bcl-2 protein. Thus, the described co-expression of tumor cell refractoriness to an anti-cancer drug and to the inhibitory cytokine TGF beta1 is accompanied by multiple changes in the TGF beta1 signal transduction pathway and in other regulatory systems of the target cells. Besides, we found that various anti-tumor drugs and cytotoxic plant lectins increased the level of TGF beta1 expression in both TGFbeta1-sensitive and -resistant L1210 cells. A hypothesis is proposed that TGFbeta1 can at least partly mediate the effect of cell-stressing agents and, thus, the development of TGF beta1 resistance may be responsible for the appearance of tumor cell refractoriness to the action of some anti-cancer drugs.
APA, Harvard, Vancouver, ISO, and other styles
50

Wharton, Natalia, Alyson Parris, Amy Reynolds, Esther M. Mitchell, Anastasia Sobolewski, Ahmed El Hadi, Michael P. Lewis, et al. "Su1734 Canonical Wnt Signals Combined With Suppressed TGFbeta/BMP Pathways Promote Renewal of the Native Human Colonic Epithelium." Gastroenterology 144, no. 5 (May 2013): S—463. http://dx.doi.org/10.1016/s0016-5085(13)61712-6.

Full text
APA, Harvard, Vancouver, ISO, and other styles
We offer discounts on all premium plans for authors whose works are included in thematic literature selections. Contact us to get a unique promo code!

To the bibliography