Abrogation Of Gram Negative Bacteria Induced Inflammation

Main Article Content

Manojit Bysack
Rajen Dey

Abstract

Lipopolysaccharide (LPS) is a prototypical exogenous endotoxicpyrogen as it binds with TLR4, and CD14 receptors of especially expressed on monocytes, macrophages, dendritic cells thereby secreting pro-inflammatory cytokines, nitric oxide, eicosanoids superoxide etc. As a result, LPS generates a variety of mediators, such as TNF-α, which triggers NF-κB, which results in cellular damage and fatal tissue injury, such as multiple organ dysfunction syndrome (MODS) and systemic inflammatory response syndrome (SIRS). The TNF-α being soluble diffuses through the membrane and interacts with the trimeric mTNFR1. This in turn causes the activation of STAT1 and STAT5 resulting in production of NF-κB induced P65-P50 heterodimer that transcribe mRNA. This results in the production of pro-inflammatory cytokines such as IL-1, IL-6, and IL-12, which in turn regulate inflammation. When the cell is at rest, the PIP2 on its surface facilitates the recruitment of an adaptor called MyD88-adapter-like (MAL/TIRAP). When MAL and MyD88 are recruited during the initial phases of Microbes-associated molecular pattern (MAMP) recognition, TLR4 activation occurs after stimulation. causes the MAP kinases JNK, p38, and NF-κβ to become activated as a result, which in turn causes the pro-inflammatory cytokine TNF-α to be produced. When cells release TNF-α, they bind with TNFR1, which releases the transcription factor domain death (SODD). Additionally, they recruit receptor interacting protein-1 (RIP-1), which in turn recruits mitogen-activated protein kinase kinasekinase 3, transforming growth factor β-activated kinase (TAK-1), and finally, they activate inhibitor of NF-κβ kinase (IKK) complex. The IKK complex then phosphorylates IκBα and thereby releasing NF-κB subunit, bound to unstimulatedIκBα by ubiquitination and degradation of IκBα. This soluble NF-κBtranslocate into the nucleus and thereafter evaluate gene transcription that is a central mediator of pro-inflammatory effects. The signalling takes place via activation of STAT1 and triggering the expression of IL-12, NOS-2 and Suppressor of cytokine signalling (SOCS), characterized by the expression of TNF-α as a major and iNOS as well as high nitric oxide and intermediate ROS production. By convention blocking of production of pro-inflammatory cytokines may be resulting in production of anti-inflammatory causes the regulation of LPS induced sepsis by anti-inflammatory mediators. This may be an alternative regulating option to diminish the septic shock via activation and translocation of STAT6 by JAK1 and JAK3 signalling. In addition to that IL-10 is produced by these cytokines causes activation of STAT3 via up-regulating the expression of SOCS3, which mediates the suppression of pro-inflammatory cytokine signalling pathways. Thus TNFRSF1A blocking will surely help society to find out future treatment strategies of acute and chronic diseases such as sepsis, disorientation of vital body functions, certain antibiotic resistant, Capillary leak syndrome, disseminated Intravascular Coagulation, purpura, ecchymoses, gangrene, multiple sclerosis etc. via the wound healing sub one secrete IL-4 and remodelling of the degrading tissues.

Downloads

Download data is not yet available.

Article Details

How to Cite
Manojit Bysack, & Rajen Dey. (2024). Abrogation Of Gram Negative Bacteria Induced Inflammation. Journal of Advanced Zoology, 45(2), 1564–1573. https://doi.org/10.53555/jaz.v45i2.4453
Section
Articles
Author Biographies

Manojit Bysack

Department of Medical Laboratory Technology, School of Allied Health Sciences, Swami Vivekananda University, Telinipara, Barasat-Barrackpore Rd, Bara Kanthalia, West Bengal-700121, India

Rajen Dey

Department of Medical Laboratory Technology, School of Allied Health Sciences, Swami Vivekananda University, Telinipara, Barasat-Barrackpore Rd, Bara Kanthalia, West Bengal-700121, India

References

Huber Mayer, U. RamadasBhat, Hussein Masoud, Joanna Radziejewska-Lebrecht, Christiane Widemann and Jurgen H. Krauss, Bacterial lipopolysaccharides, Pure & Appl. Chem. (1989), Vol. 61, No. 7, pp. 1271-1282.

Raetz, C. R. (1990). Biochemistry of endotoxins. Annual review of biochemistry, 59(1), 129-170.

Solov'eva, T., Davydova, V., Krasikova, I., &Yermak, I. (2013). Marine compounds with therapeutic potential in gram-negative sepsis. Marine drugs, 11(6), 2216-2229.

Zhu, C. B., Blakely, R. D., & Hewlett, W. A. (2006). The proinflammatory cytokines interleukin-1beta and tumor necrosis factor-alpha activate serotonin transporters. Neuropsychopharmacology, 31(10), 2121-2131.

Blüml, S., Scheinecker, C., Smolen, J. S., &Redlich, K. (2012). Targeting TNF receptors in rheumatoid arthritis. International immunology, 24(5), 275-281.

Rogero, M. M., & Calder, P. C. (2018). Obesity, inflammation, toll-like receptor 4 and fatty acids. Nutrients, 10(4), 432.

Liang, H., Yin, B., Zhang, H., Zhang, S., Zeng, Q., Wang, J., ...& Li, Z. (2008). Blockade of tumor necrosis factor (TNF) receptor type 1-mediated TNF-α signaling protected Wistar rats from diet-induced obesity and insulin resistance. Endocrinology, 149(6), 2943-2951..

Williams, S. K., Maier, O., Fischer, R., Fairless, R., Hochmeister, S., Stojic, A., ...& Diem, R. (2014). Antibody-mediated inhibition of TNFR1 attenuates disease in a mouse model of multiple sclerosis. PloS one, 9(2), e90117.

Martinez, F. O., & Gordon, S. (2014). The M1 and M2 paradigm of macrophage activation: time for reassessment. F1000prime reports, 6.

Jakobi, V., &Petry, F. (2008). Humoral immune response in IL‐12 and IFN‐γ deficient mice after infection with Cryptosporidium parvum. Parasite immunology, 30(3), 151-161.

Doyle, A. G., Herbein, G., Montaner, L. J., Minty, A. J., Caput, D., Ferrara, P., & Gordon, S. (1994). Interleukin‐13 alters the activation state of murine macrophages in vitro: Comparison with interleukin‐4 and interferon‐γ. European journal of immunology, 24(6), 1441-1445.

Mills, C. D., Kincaid, K., Alt, J. M., Heilman, M. J., & Hill, A. M. (2000). M-1/M-2 macrophages and the Th1/Th2 paradigm. The Journal of immunology, 164(12), 6166-6173.

Jenkins, S. J., & Allen, J. E. (2010). Similarity and diversity in macrophage activation by nematodes, trematodes, and cestodes. Journal of Biomedicine and Biotechnology, 2010.

Hotchkiss, R. S., & Karl, I. E. (2003). The pathophysiology and treatment of sepsis. New England journal of medicine, 348(2), 138-150.

Vassalli, P. (1992). The pathophysiology of tumor necrosis factors. Annual review of immunology, 10(1), 411-452.

Ruuls, S. R., Hoek, R. M., Ngo, V. N., McNeil, T., Lucian, L. A., Janatpour, M. J., ...& Sedgwick, J. D. (2001). Membrane-bound TNF supports secondary lymphoid organ structure but is subservient to secreted TNF in driving autoimmune inflammation. Immunity, 15(4), 533-543.

Flynn, J. L., Goldstein, M. M., Chan, J., Triebold, K. J., Pfeffer, K., Lowenstein, C. J., ... & Bloom, B. R. (1995). Tumor necrosis factor-α is required in the protective immune response against Mycobacterium tuberculosis in mice. Immunity, 2(6), 561-572.

Garcia, I., Miyazaki, Y., Araki, K., Araki, M., Lucas, R., Grau, G. E., ...&Vassalli, P. (1995). Transgenic mice expressing high levels of soluble tnf‐r1 fusion protein are protected from lethal septic shock and cerebral malaria, and are highly sensitive to Listeria monocytogenes and Leishmania major infections. European journal of immunology, 25(8), 2401-2407.

Zantl, N., Uebe, A., Neumann, B., Wagner, H., Siewert, J. R., Holzmann, B., ...&Pfeffer, K. (1998). Essential role of gamma interferon in survival of colon ascendens stent peritonitis, a novel murine model of abdominal sepsis. Infection and immunity, 66(5), 2300-2309.

Secher, T., Vasseur, V., Poisson, D. M., Mitchell, J. A., Cunha, F. Q., Alves-Filho, J. C., &Ryffel, B. (2009). Crucial role of TNF receptors 1 and 2 in the control of polymicrobial sepsis. The Journal of Immunology, 182(12), 7855-7864.

Waterston, A. M., Salway, F., Andreakos, E., Butler, D. M., Feldmann, M., & Coombes, R. C. (2004). TNF autovaccination induces self anti-TNF antibodies and inhibits metastasis in a murine melanoma model. British journal of cancer, 90(6), 1279-1284.

Maitra, U., Deng, H., Glaros, T., Baker, B., Capelluto, D. G., Li, Z., & Li, L. (2012). Molecular mechanisms responsible for the selective and low-grade induction of proinflammatory mediators in murine macrophages by lipopolysaccharide. The Journal of Immunology, 189(2), 1014-1023.

Lowry, O., Rosebrough, N., Farr, A. L., & Randall, R. (1951). Protein measurement with the Folin phenol reagent. Journal of biological chemistry, 193(1), 265-275.

Ghasemi, A., Hedayati, M., &Biabani, H. (2007). Protein precipitation methods evaluated for determination of serum nitric oxide end products by the Griess assay. Jmsr, 2(15), 29-32.

Tracey, K. J., Beutler, B., Lowry, S. F., Merryweather, J., Wolpe, S., Milsark, I. W., ...&Cerami, A. (1986). Shock and tissue injury induced by recombinant human cachectin. Science, 234(4775), 470-474.

Arteaga Figueroa, L., Abarca-Vargas, R., García Alanis, C., &Petricevich, V. L. (2017). Comparison between peritoneal macrophage activation by Bougainvillea xbuttiana extract and LPS and/or interleukins. BioMed Research International, 2017.

Itoh, K., Udagawa, N., Kobayashi, K., Suda, K., Li, X., Takami, M., ...& Takahashi, N. (2003). Lipopolysaccharide promotes the survival of osteoclasts via Toll-like receptor 4, but cytokine production of osteoclasts in response to lipopolysaccharide is different from that of macrophages. The Journal of Immunology, 170(7), 3688-3695.

Zakharova, M., & Ziegler, H. K. (2005). Paradoxical anti-inflammatory actions of TNF-α: inhibition of IL-12 and IL-23 via TNF receptor 1 in macrophages and dendritic cells. The Journal of Immunology, 175(8), 5024-5033.

Wang, L. X., Zhang, S. X., Wu, H. J., Rong, X. L., &Guo, J. (2019). M2b macrophage polarization and its roles in diseases. Journal of leukocyte biology, 106(2), 345-358.

Tan, H. Y., Wang, N., Li, S., Hong, M., Wang, X., &Feng, Y. (2016). The reactive oxygen species in macrophage polarization: reflecting its dual role in progression and treatment of human diseases. Oxidative medicine and cellular longevity, 2016.

Elizur, A., Adair-Kirk, T. L., Kelley, D. G., Griffin, G. L., Demello, D. E., & Senior, R. M. (2008). Tumor necrosis factor-α from macrophages enhances LPS-induced Clara cell expression of keratinocyte-derived chemokine. American journal of respiratory cell and molecular biology, 38(1), 8-15.

Sedger, L. M., & McDermott, M. F. (2014). TNF and TNF-receptors: From mediators of cell death and inflammation to therapeutic giants–past, present and future. Cytokine & growth factor reviews, 25(4), 453-472.

Fischer, R., Kontermann, R. E., & Maier, O. (2015). Targeting sTNF/TNFR1 signaling as a new therapeutic strategy. Antibodies, 4(1), 48-70.

Gane, J. M., Stockley, R. A., &Sapey, E. (2016). TNF-α autocrine feedback loops in human monocytes: the pro-and anti-inflammatory roles of the TNF-α receptors support the concept of selective TNFR1 blockade in vivo. Journal of immunology research, 2016.

Tamura, K., Noyama, T., Ishizawa, Y. H., Takamatsu, N., Shiba, T., & Ito, M. (2004). Xenopus death receptor-M1 and-M2, new members of the tumor necrosis factor receptor superfamily, trigger apoptotic signaling by differential mechanisms. Journal of Biological Chemistry, 279(9), 7629-7635.

Kratochvill, F., Neale, G., Haverkamp, J. M., Van de Velde, L. A., Smith, A. M., Kawauchi, D., ...& Murray, P. J. (2015). TNF counterbalances the emergence of M2 tumor macrophages. Cell reports, 12(11), 1902-1914.

Chang, Y. T. (1964). Long-term cultivation of mouse peritoneal macrophages. Journal of the National Cancer Institute, 32(1), 19-35.

Guo, M., Xiao, J., Sheng, X., Zhang, X., Tie, Y., Wang, L., ...&Ji, X. (2018). Ginsenoside Rg3 mitigates atherosclerosis progression in diabetic apoE–/–mice by skewing macrophages to the M2 phenotype. Frontiers in Pharmacology, 9, 464.

Ghasemi, A., Hedayati, M., &Biabani, H. (2007). Protein precipitation methods evaluated for determination of serum nitric oxide end products by the Griess assay. Jmsr, 2(15), 29-32.

Nyberg, P., &Klockars, M. (1990). Measurement of reactive oxygen metabolites produced by human monocyte-derived macrophages exposed to mineral dusts. International Journal of Experimental Pathology, 71(4), 537.

Sawoo, R., Dey, R., Ghosh, R., &Bishayi, B. (2021). TLR4 and TNFR1 blockade dampen M1 macrophage activation and shifts them towards an M2 phenotype. Immunologic Research, 69, 334-351.

Most read articles by the same author(s)