Academic literature on the topic 'TNFα-mediated cytotoxicity'

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

Select a source type:

Consult the lists of relevant articles, books, theses, conference reports, and other scholarly sources on the topic 'TNFα-mediated cytotoxicity.'

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.

Journal articles on the topic "TNFα-mediated cytotoxicity"

1

Tyrinova, Tamara, Olga Leplina, Sergey Mishinov, Marina Tikhonova, Evgeniya Dolgova, Anastasiya Proskurina, Vyacheslav Stupack, Sergey Bogachev, Alexander Ostanin, and Elena Chernykh. "Defective Regulation of Membrane TNFα Expression in Dendritic Cells of Glioblastoma Patients Leads to the Impairment of Cytotoxic Activity against Autologous Tumor Cells." International Journal of Molecular Sciences 21, no. 8 (April 21, 2020): 2898. http://dx.doi.org/10.3390/ijms21082898.

Full text
Abstract:
Besides an antigen-presenting function and ability to induce antitumor immune responses, dendritic cells (DCs) possess a direct tumoricidal activity. We previously reported that monocyte-derived IFNα-induced DCs (IFN-DCs) of glioblastoma multiforme patients express low levels of membrane TNFα molecule (mTNFα) and have impaired TNFα/TNF-R1-mediated cytotoxicity against immortalized tumor cell line HEp-2. However, whether the observed defect could affect killer activity of glioma patient DCs against autologous tumor cells remained unclear. Here, we show that donor IFN-DCs possess cytotoxic activity against glioblastoma cell lines derived from a primary tumor culture. Granule-mediated and TNFα/TNF-R1-dependent pathways were established as the main mechanisms underlying cytotoxic activity of IFN-DCs. Glioblastoma patient IFN-DCs showed lower cytotoxicity against autologous glioblastoma cells sensitive to TNFα/TNFR1-mediated lysis, which was associated with low TNFα mRNA expression and high TACE/ADAM-17 enzyme activity. Recombinant IL-2 (rIL-2) and human double-stranded DNA (dsDNA) increased 1.5-fold cytotoxic activity of patient IFN-DCs against autologous glioblastoma cells. dsDNA, but not rIL-2, enhanced the expression of TNFα mRNA and decreased expression and activity of TACE/ADAM-17 enzyme. In addition, dsDNA and rIL-2 stimulated the expression of perforin and granzyme B (in the presence of dsDNA), suggesting the possibility of enhancing DC cytotoxicity against autologous glioblastoma cells via various mechanisms.
APA, Harvard, Vancouver, ISO, and other styles
2

Laurent, Guy, Fabien Guilloton, Aurelie de Thonel, Christine Jean, Cecile Demur, Veronique De Mas, and Anne Quillet-Mary. "TNFα Stimulates NKG2D-Mediated Lytic Activity of Acute Myeloid Leukemic Cells." Blood 106, no. 11 (November 16, 2005): 4411. http://dx.doi.org/10.1182/blood.v106.11.4411.4411.

Full text
Abstract:
Abstract The mechanism by which leukemic cells interfere with normal hematopoiesis remains unclear. We show that, whereas the leukemic KG1a cells are naturally devoid from cellular cytotoxicity, once activated by TNFα, they display cytolytic activity towards various cellular targets including CFU-GM. This mechanism is dependent on stimulation of Granzyme B/Perforin system. In addition, KG1a cells expressed the NKG2D receptor and its signal-transducing adaptator DAP10, which were functional as confirmed by redirected lysis experiments. Interestingly, flow cytometry analysis of 20 samples of patient with acute myeloid leukemia (AML) (FAB M0 to M5) revealed the expression of NKG2D (40%) and other natural cytotoxicity receptors (40% for NKp30, 74% for NKp44, 39% for NKp46) by pool >15% of leukemic cells. Futhermore, CD34+ hematopoietic progenitors undergoing granulo-monocytic differentiation expressed NKG2D ligands. Altogether, we propose a model in which upon stimulation by TNFα leukemic cells may exert cytotoxicity against myeloid progenitors. This finding may have important clinical implications in the context of diseases characterized by TNFα accumulation, such as AML or myelodisplasic syndromes.
APA, Harvard, Vancouver, ISO, and other styles
3

Chen, Chih-Chiun, Jennifer L. Young, Ricardo I. Monzon, Ningyu Chen, Viktor Todorović, and Lester F. Lau. "Cytotoxicity of TNFα is regulated by integrin-mediated matrix signaling." EMBO Journal 26, no. 5 (February 22, 2007): 1257–67. http://dx.doi.org/10.1038/sj.emboj.7601596.

Full text
APA, Harvard, Vancouver, ISO, and other styles
4

Miyaji, Chikako, Hisami Watanabe, Ryoko Miyakawa, Hisashi Yokoyama, Chika Tsukada, Yuiko Ishimoto, Shinobu Miyazawa, and Toru Abo. "Identification of effector cells for TNFα-mediated cytotoxicity against WEHI164S cells." Cellular Immunology 216, no. 1-2 (March 2002): 43–49. http://dx.doi.org/10.1016/s0008-8749(02)00525-7.

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

Li, Y., T. D. Nguyen, A. C. Stechschulte, D. J. Stechschulte, and K. N. Dileepan. "Effect of mast cell granules on the gene expression of nitric oxide synthase and tumour necrosis factor-α in macrophages." Mediators of Inflammation 7, no. 5 (1998): 355–61. http://dx.doi.org/10.1080/09629359890884.

Full text
Abstract:
Previous studies have shown that mast cell granules (MCG) inhibit numerous macrophage functions including tumour cytotoxicity, superoxide and nitric oxide (NO) production, and FCγ2a receptor-mediated phagocytosis. In this study, the effect of MCG on macrophage TNFα and nitric oxide synthase (iNOS) mRNA expression, and the production and fate of TNFα were examined. Upon activation with LPS+IFNγ, macrophages expressed both TNFα and iNOS mRNA and produced both TNFα and NO. Co-incubation of LPS+IFNγ-activated macrophages with MCG resulted in dose-dependent inhibition of iNOS mRNA expression. TNFα production in the activated macrophages was decreased by MCG, which was associated with a reduction in TNFα mRNA expression. MCG were also capable of degrading both macrophage-generated and recombinant TNFα. The direct effect of MCG on TNFα was partially reversed by a mixture of protease inhibitors. These results demonstrate that MCG decrease the production of NO and TNFα by inhibiting macrophage iNOS and TNFα gene expression. Furthermore, MCG post-transcriptionally alter TNFα levels via proteolytic degradation.
APA, Harvard, Vancouver, ISO, and other styles
6

Hu, Zhengbo, Ramya Viswanathan, Hui Cheng, Jianghong Chen, Xinping Yang, Angel Huynh, Paul Clavijo, et al. "Inhibiting WEE1 and IKK-RELA Crosstalk Overcomes TNFα Resistance in Head and Neck Cancers." Molecular Cancer Research 20, no. 6 (February 17, 2022): 867–82. http://dx.doi.org/10.1158/1541-7786.mcr-21-0624.

Full text
Abstract:
Abstract TNFα is a key mediator of immune and radiotherapy-induced cytotoxicity, but many cancers, including head and neck squamous cell carcinomas (HNSCC), display TNF resistance due to activation of the canonical IKK–NF-κB/RELA pro-survival pathway. However, toxicities associated with direct targeting of the canonical pathway point to the need to identify mechanism(s) contributing to TNFα resistance and synthetic lethal targets to overcome such resistance in cancer cells. Here, RNAi screening for modulators of TNFα–NF-κB reporter activity and cell survival unexpectedly implicated the WEE1 and CDC2 G2–M checkpoint kinases. The IKKα/β-RELA and WEE1-CDC2 signaling pathways are activated by TNFα and form a complex in cell lines derived from both human papillomavirus (−) and (+) subtypes of HNSCC. WEE1 inhibitor AZD1775 reduced IKK/RELA phosphorylation and the expression of NF-κB–dependent pro-survival proteins Cyclin D1 and BCL2. Combination of TNFα and AZD1775 enhanced caspase-mediated apoptosis in vitro, and combination treatment with radiotherapy and AZD1775 potentiated inhibition of HNSCC tumor xenograft growth in vivo, which could be significantly attenuated by TNFα depletion. These data offer new insight into the interplay between NF-κB signaling and WEE1-mediated regulation of the G2–M cell-cycle checkpoint in HNSCC. Implications: Inhibiting WEE1 and IKK-RELA crosstalk could potentially enhance the effects of therapies mediated by TNFα with less systemic immune suppression and toxicity than observed with direct interruption of IKK-NF-κB/RELA signaling.
APA, Harvard, Vancouver, ISO, and other styles
7

Almishri, Wagdi, Tania Santodomingo-Garzon, Tyson Le, Danuta Stack, Christopher H. Mody, and Mark G. Swain. "TNFα Augments Cytokine-Induced NK Cell IFNγ Production through TNFR2." Journal of Innate Immunity 8, no. 6 (2016): 617–29. http://dx.doi.org/10.1159/000448077.

Full text
Abstract:
NK cells play a central role in innate immunity, acting directly through cell-mediated cytotoxicity and by secreting cytokines. TNFα activation of TNFR2 enhances NK cell cytotoxicity, but its effects on the other essential function of NK cells - cytokine production, for which IFNγ is paramount - are poorly defined. We identify the expression of both TNFα receptors on human peripheral blood NK cells (TNFR2 > TNFR1) and show that TNFα significantly augments IFNγ production from IL-2-/IL-12-treated NK cells in vitro, an effect mimicked by a TNFR2 agonistic antibody. TNFα also enhanced murine NK cell IFNγ production via TNFR2 in vitro. In a mouse model characterized by the hepatic recruitment and activation of NK cells, TNFR2 also regulated NK cell IFNγ production in vivo. Specifically, in this model, after activation of an innate immune response, hepatic numbers of TNFR2-expressing and IFNγ-producing NK cells were both significantly increased; however, the frequency of IFNγ-producing hepatic NK cells was significantly reduced in TNFR2-deficient mice. We delineate an important role for TNFα, acting through TNFR2, in augmenting cytokine-induced NK cell IFNγ production in vivo and in vitro, an effect with significant potential implications for the regulation of innate and adaptive immune responses.
APA, Harvard, Vancouver, ISO, and other styles
8

Toni, Tiffany, Ramya Viswanathan, Yvette Robbins, Sreenivasulu Gunti, Xinping Yang, Angel Huynh, Hui Cheng, et al. "Combined Inhibition of IAPs and WEE1 Enhances TNFα- and Radiation-Induced Cell Death in Head and Neck Squamous Carcinoma." Cancers 15, no. 4 (February 6, 2023): 1029. http://dx.doi.org/10.3390/cancers15041029.

Full text
Abstract:
Head and neck squamous cell carcinoma (HNSCC) remains a prevalent diagnosis with current treatment options that include radiotherapy and immune-mediated therapies, in which tumor necrosis factor-α (TNFα) is a key mediator of cytotoxicity. However, HNSCC and other cancers often display TNFα resistance due to activation of the canonical IKK–NFκB/RELA pathway, which is activated by, and induces expression of, cellular inhibitors of apoptosis proteins (cIAPs). Our previous studies have demonstrated that the IAP inhibitor birinapant sensitized HNSCC to TNFα-dependent cell death in vitro and radiotherapy in vivo. Furthermore, we recently demonstrated that the inhibition of the G2/M checkpoint kinase WEE1 also sensitized HNSCC cells to TNFα-dependent cell death, due to the inhibition of the pro-survival IKK-NFκB/RELA complex. Given these observations, we hypothesized that dual-antagonist therapy targeting both IAP and WEE1 proteins may have the potential to synergistically sensitize HNSCC to TNFα-dependent cell death. Using the IAP inhibitor birinapant and the WEE1 inhibitor AZD1775, we show that combination treatment reduced cell viability, proliferation and survival when compared with individual treatment. Furthermore, combination treatment enhanced the sensitivity of HNSCC cells to TNFα-induced cytotoxicity via the induction of apoptosis and DNA damage. Additionally, birinapant and AZD1775 combination treatment decreased cell proliferation and survival in combination with radiotherapy, a critical source of TNFα. These results support further investigation of IAP and WEE1 inhibitor combinations in preclinical and clinical studies in HNSCC.
APA, Harvard, Vancouver, ISO, and other styles
9

Ariana, Ardeshir, Norah A. Alturki, Stephanie Hajjar, Deborah J. Stumpo, Christopher Tiedje, Emad S. Alnemri, Matthias Gaestel, Perry J. Blackshear, and Subash Sad. "Tristetraprolin regulates necroptosis during tonic Toll-like receptor 4 (TLR4) signaling in murine macrophages." Journal of Biological Chemistry 295, no. 14 (February 24, 2020): 4661–72. http://dx.doi.org/10.1074/jbc.ra119.011633.

Full text
Abstract:
The necrosome is a protein complex required for signaling in cells that results in necroptosis, which is also dependent on tumor necrosis factor receptor (TNF-R) signaling. TNFα promotes necroptosis, and its expression is facilitated by mitogen-activated protein (MAP) kinase–activated protein kinase 2 (MK2) but is inhibited by the RNA-binding protein tristetraprolin (TTP, encoded by the Zfp36 gene). We have stimulated murine macrophages from WT, MyD88−/−, Trif−/−, MyD88−/−Trif−/−, MK2−/−, and Zfp36−/− mice with graded doses of lipopolysaccharide (LPS) and various inhibitors to evaluate the role of various genes in Toll-like receptor 4 (TLR4)–induced necroptosis. Necrosome signaling, cytokine production, and cell death were evaluated by immunoblotting, ELISA, and cell death assays, respectively. We observed that during TLR4 signaling, necrosome activation is mediated through the adaptor proteins MyD88 and TRIF, and this is inhibited by MK2. In the absence of MK2-mediated necrosome activation, lipopolysaccharide-induced TNFα expression was drastically reduced, but MK2-deficient cells became highly sensitive to necroptosis even at low TNFα levels. In contrast, during tonic TLR4 signaling, WT cells did not undergo necroptosis, even when MK2 was disabled. Of note, necroptosis occurred only in the absence of TTP and was mediated by the expression of TNFα and activation of JUN N-terminal kinase (JNK). These results reveal that TTP plays an important role in inhibiting TNFα/JNK-induced necrosome signaling and resultant cytotoxicity.
APA, Harvard, Vancouver, ISO, and other styles
10

Souders, Christopher L., Juan J. Aristizabal-Henao, Sarah J. Patuel, John A. Bowden, Jasenka Zubcevic, and Christopher J. Martyniuk. "Interaction between Butyrate and Tumor Necrosis Factor α in Primary Rat Colonocytes." Biomolecules 13, no. 2 (January 30, 2023): 258. http://dx.doi.org/10.3390/biom13020258.

Full text
Abstract:
Butyrate, a short-chain fatty acid, is utilized by the gut epithelium as energy and it improves the gut epithelial barrier. More recently, it has been associated with beneficial effects on immune and cardiovascular homeostasis. Conversely, tumor necrosis factor alpha (TNFα) is a pro-inflammatory and pro-hypertensive cytokine. While butyrate and TNFα are both linked with hypertension, studies have not yet addressed their interaction in the colon. Here, we investigated the capacity of butyrate to modulate a host of effects of TNFα in primary rodent colonic cells in vitro. We measured ATP levels, cell viability, mitochondrial membrane potential (MMP), reactive oxygen species (ROS), mitochondrial oxidative phosphorylation, and glycolytic activity in colonocytes following exposure to either butyrate or TNFα, or both. To address the potential mechanisms, transcripts related to oxidative stress, cell fate, and cell metabolism (Pdk1, Pdk2, Pdk4, Spr, Slc16a1, Slc16a3, Ppargc1a, Cs, Lgr5, Casp3, Tnfr2, Bax, Bcl2, Sod1, Sod2, and Cat) were measured, and untargeted liquid chromatography–tandem mass spectrometry (LC-MS/MS) was employed to profile the metabolic responses of colonocytes following exposure to butyrate and TNFα. We found that both butyrate and TNFα lowered cellular ATP levels towards a quiescent cell energy phenotype, characterized by decreased oxygen consumption and extracellular acidification. Co-treatment with butyrate ameliorated TNFα-induced cytotoxicity and the reduction in cell viability. Butyrate also opposed the TNFα-mediated decrease in MMP and mitochondrial-to-intracellular calcium ratios, suggesting that butyrate may protect colonocytes against TNFα-induced cytotoxicity by decreasing mitochondrial calcium flux. The relative expression levels of pyruvate dehydrogenase kinase 4 (Pdk4) were increased via co-treatment of butyrate and TNFα, suggesting the synergistic inhibition of glycolysis. TNFα alone reduced the expression of monocarboxylate transporters slc16a1 and slc16a3, suggesting effects of TNFα on butyrate uptake into colonocytes. Of the 185 metabolites that were detected with LC-MS, the TNFα-induced increase in biopterin produced the only significant change, suggesting an alteration in mitochondrial biogenesis in colonocytes. Considering the reports of elevated colonic TNFα and reduced butyrate metabolism in many conditions, including in hypertension, the present work sheds light on cellular interactions between TNFα and butyrate in colonocytes that may be important in understanding conditions of the colon.
APA, Harvard, Vancouver, ISO, and other styles

Dissertations / Theses on the topic "TNFα-mediated cytotoxicity"

1

Jameei, Aida. "Development and Characterization of a potent Tumor Necrosis Factor-alpha blocking agent." Thesis, 2018. https://etd.iisc.ac.in/handle/2005/5483.

Full text
Abstract:
Tumor necrosis factor-α (TNFα), one of the major pro-inflammatory cytokines, plays a pivotal role in an effective inflammation process and immune response. TNFα interacts with two different receptors, TNFR1 and TNFR2, which are differentially expressed on cells to initiate both, distinct and overlapping signal transduction pathways. The diverse signalling pathways lead to different cellular responses, including cell death, survival, differentiation, proliferation and cell migration. Vascular endothelial cells respond to TNFα by undergoing several pro-inflammatory changes which increase leukocyte adhesion, endothelial translocation and vascular leak to promote thrombosis. The central role of TNFα in inflammation has been demonstrated by the ability of agents that block the action of TNFα to treat a range of inflammatory conditions, including Rheumatoid arthritis, Psoriasis, Psoriatic arthritis, Crohn’s disease, Ulcerative colitis, Ankylosing spondylitis etc. Inhibition of TNFα by pharmacological inhibitors or antibodies has proven to be effective in palliative treatment to some extent. Despite having beneficial effects of anti-TNFα agents in inflammation associated diseases, there are some unexpected, aberrant adverse events with all approved TNFα inhibitors, such as opportunistic infections, tuberculosis, demyelinating disease and drug-induced lupus erythematosus. Antibodies have different applications, including, a) medicine (diagnosis, therapy), b) biomedical research, (Western blotting, immunosorbent assays, immunohistochemistry, immunoprecipitation, and flow cytometry). Due to the cardinal role of antibodies in therapies, such as antibody drug conjugates and tumour therapy, development of therapeutic antibodies has become one of the most challenging area of medical biology. Pharmaceutical industries and medical biologist have been trying to use the specificity of antibodies for drug development against a range of diseases. The present study focuses on development of a neutralizing anti-TNFα antibody which may be used as a therapeutic option to inhibit TNFα-mediated cytotoxicity and cell death. To achieve this goal, we have generated and characterized a library of 13 hybridoma clones secreting monoclonal antibodies (mAbs) against human-TNFα as described in chapter 2. Four of the mAbs rescued L929 fibroblast cells from TNFα triggered cell death as examined by MTT assay, out of 5 which mAb C8 was found to have the highest affinity to human-TNFα and importantly, inhibited TNFα toxicity at very low concentrations. In order to gain insights in to the mechanism by which C8 antibody inhibits human TNFα-mediated toxicity, we attempted to delineate the epitope corresponding to the mAb C8 as described in chapter 3. Thus, towards the objective, truncation and mutational analysis of TNFα revealed that the epitope corresponding to mAb C8 lies in the stretch of amino acids 99-120, and that the amino acids 102-104(QRE) form the core epitope. The Antigen-Antibody complex model also confirmed the result. The data presented in chapter 4 describes studies carried out to compare the binding affinities of mAb C8 with the commercially available Infliximab and also their effectiveness as TNFα inhibitors. Recent progress in the field of antibody engineering opened a new window for generating single-chain variable fragments (scFv). To generate C8 scFv, the variable regions of the heavy (VH) and light chains (VL) were amplified from the hybridoma mRNA and linked together. ELISA results revealed that immobilized scFv binds specifically to hTNFα. However, the scFv did not bind to the antigen when in solution phase, suggesting that further studies are required to stabilize its conformation. The data are presented in chapter 5. In brief, the thesis is presented in five chapters as described above: Chapter 1- ‘Introduction’, provides a brief overview on the discovery of Tumor necrosis factor-α, its’ structure, expression and signalling, TNFα role in physiological condition as well as during inflammation, TNFα associated diseases and available inhibitors of therapeutic activity. Chapter 2- describes the characterization and screening of monoclonal antibodies against human-TNFα and the identification of the highest affinity mAb C8. Chapter 3- presents the identification of the epitope corresponding to the neutralizing mAb C8. Chapter 4- discusses the results obtained from comparative study of mAb C8 and Infliximab (anti-TNFα mAb). And Chapter 5- describes the generation of single-chain variable fragment (scFv) from mAb C8 secreting hybridoma and investigating its binding. In summary, we have obtained a monoclonal antibody to hTNFα that can effectively neutralize TNFα-mediated cytotoxicity and maybe explored for use in anti-inflammation therapy.
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