Less Severe Polymicrobial Sepsis in Conditional mgmt-Deleted Mice Using LysM-Cre System, Impacts of DNA Methylation and MGMT Inhibitor in Sepsis
Abstract
:1. Introduction
2. Results
2.1. Proteomic Analysis of Lipopolysaccharide (LPS)-Activated Wild-Type Macrophages and Impacts of mgmt Null and MGMT Inhibitor on LPS Stimulation
2.2. The Mgmt Null Mice Demonstrated Less Severe Cecal Ligation and Puncture (CLP) Sepsis than the Littermate Control
3. Discussion
3.1. Possible LPS-Induced Macrophage Injury from the Proteomic Analysis in the Wild-Type Cells
3.2. Impact of mgmt and DNA Repair in Sepsis, In Vitro and In Vivo
3.3. Clinical Aspect and Future Experiments
4. Materials and Methods
4.1. The Proteomic Analysis
4.2. The In Vitro Experiments
4.3. Animal and Animal Model
4.4. Mouse Sample Analysis
4.5. Statistical Analysis
5. Conclusions
Supplementary Materials
Author Contributions
Funding
Institutional Review Board Statement
Data Availability Statement
Conflicts of Interest
References
- Brady, J.; Horie, S.; Laffey, J.G. Role of the adaptive immune response in sepsis. Intensive Care Med. Exp. 2020, 8, 20. [Google Scholar] [CrossRef] [PubMed]
- Sae-Khow, K.; Charoensappakit, A.; Chiewchengchol, D.; Leelahavanichkul, A. High-Dose Intravenous Ascorbate in Sepsis, a Pro-Oxidant Enhanced Microbicidal Activity and the Effect on Neutrophil Functions. Biomedicines 2022, 11, 51. [Google Scholar] [CrossRef] [PubMed]
- Amornphimoltham, P.; Yuen, P.S.T.; Star, R.A.; Leelahavanichkul, A. Gut Leakage of Fungal-Derived Inflammatory Mediators: Part of a Gut-Liver-Kidney Axis in Bacterial Sepsis. Dig. Dis. Sci. 2019, 64, 2416–2428. [Google Scholar] [CrossRef] [PubMed]
- Perner, A.; Rhodes, A.; Venkatesh, B.; Angus, D.C.; Martin-Loeches, I.; Preiser, J.C.; Vincent, J.L.; Marshall, J.; Reinhart, K.; Joannidis, M.; et al. Sepsis: Frontiers in supportive care, organisation and research. Intensive Care Med. 2017, 43, 496–508. [Google Scholar] [CrossRef]
- Nemeth, K.; Leelahavanichkul, A.; Yuen, P.S.; Mayer, B.; Parmelee, A.; Doi, K.; Robey, P.G.; Leelahavanichkul, K.; Koller, B.H.; Brown, J.M.; et al. Bone marrow stromal cells attenuate sepsis via prostaglandin E(2)-dependent reprogramming of host macrophages to increase their interleukin-10 production. Nat. Med. 2009, 15, 42–49. [Google Scholar] [CrossRef] [Green Version]
- Leelahavanichkul, A.; Yasuda, H.; Doi, K.; Hu, X.; Zhou, H.; Yuen, P.S.; Star, R.A. Methyl-2-acetamidoacrylate, an ethyl pyruvate analog, decreases sepsis-induced acute kidney injury in mice. Am. J. Physiol. Physiol. 2008, 295, F1825–F1835. [Google Scholar] [CrossRef]
- Taratummarat, S.; Sangphech, N.; Vu, C.T.B.; Palaga, T.; Ondee, T.; Surawut, S.; Sereemaspun, A.; Ritprajak, P.; Leelahavanichkul, A. Gold nanoparticles attenuates bacterial sepsis in cecal ligation and puncture mouse model through the induction of M2 macrophage polarization. BMC Microbiol. 2018, 18, 85. [Google Scholar] [CrossRef]
- Panpetch, W.; Chancharoenthana, W.; Bootdee, K.; Nilgate, S.; Finkelman, M.; Tumwasorn, S.; Leelahavanichkul, A. Lactobacillus rhamnosus L34 Attenuates Gut Translocation-Induced Bacterial Sepsis in Murine Models of Leaky Gut. Infect. Immun. 2018, 86, e00700-17. [Google Scholar] [CrossRef] [Green Version]
- Issara-Amphorn, J.; Chancharoenthana, W.; Visitchanakun, P.; Leelahavanichkul, A. Syk Inhibitor Attenuates Polymicrobial Sepsis in FcgRIIb-Deficient Lupus Mouse Model, the Impact of Lupus Characteristics in Sepsis. J. Innate Immun. 2020, 12, 461–479. [Google Scholar] [CrossRef]
- Dang, C.P.; Leelahavanichkul, A. Over-expression of miR-223 induces M2 macrophage through glycolysis alteration and attenuates LPS-induced sepsis mouse model, the cell-based therapy in sepsis. PLoS ONE 2020, 15, e0236038. [Google Scholar] [CrossRef]
- Chancharoenthana, W.; Udompronpitak, K.; Manochantr, Y.; Kantagowit, P.; Kaewkanha, P.; Issara-Amphorn, J.; Leelahavanichkul, A. Repurposing of High-Dose Erythropoietin as a Potential Drug Attenuates Sepsis in Preconditioning Renal Injury. Cells 2021, 10, 3133. [Google Scholar] [CrossRef]
- Dang, C.P.; Issara-Amphorn, J.; Charoensappakit, A.; Udompornpitak, K.; Bhunyakarnjanarat, T.; Saisorn, W.; Sae-Khow, K.; Leelahavanichkul, A. BAM15, a Mitochondrial Uncoupling Agent, Attenuates Inflammation in the LPS Injection Mouse Model: An Adjunctive Anti-Inflammation on Macrophages and Hepatocytes. J. Innate Immun. 2021, 13, 359–375. [Google Scholar] [CrossRef]
- Mithal, L.B.; Arshad, M.; Swigart, L.R.; Khanolkar, A.; Ahmed, A.; Coates, B.M. Mechanisms and modulation of sepsis-induced immune dysfunction in children. Pediatr. Res. 2022, 91, 447–453. [Google Scholar] [CrossRef]
- Schrijver, I.T.; Theroude, C.; Roger, T. Myeloid-Derived Suppressor Cells in Sepsis. Front. Immunol. 2019, 10, 327. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Cao, C.; Ma, T.; Chai, Y.F.; Shou, S.T. The role of regulatory T cells in immune dysfunction during sepsis. World J. Emerg. Med. 2015, 6, 5–9. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Vergadi, E.; Vaporidi, K.; Tsatsanis, C. Regulation of Endotoxin Tolerance and Compensatory Anti-inflammatory Response Syndrome by Non-coding RNAs. Front. Immunol. 2018, 9, 2705. [Google Scholar] [CrossRef] [Green Version]
- Chancharoenthana, W.; Sutnu, N.; Visitchanakun, P.; Sawaswong, V.; Chitcharoen, S.; Payungporn, S.; Schuetz, A.; Schultz, M.J.; Leelahavanichkul, A. Critical roles of sepsis-reshaped fecal virota in attenuating sepsis severity. Front. Immunol. 2022, 13, 940935. [Google Scholar] [CrossRef] [PubMed]
- Chancharoenthana, W.; Kamolratanakul, S.; Ariyanon, W.; Thanachartwet, V.; Phumratanaprapin, W.; Wilairatana, P.; Leelahavanichkul, A. Abnormal Blood Bacteriome, Gut Dysbiosis, and Progression to Severe Dengue Disease. Front. Cell. Infect. Microbiol. 2022, 12, 890817. [Google Scholar] [CrossRef]
- Hiengrach, P.; Visitchanakun, P.; Tongchairawewat, P.; Tangsirisatian, P.; Jungteerapanich, T.; Ritprajak, P.; Wannigama, D.L.; Tangtanatakul, P.; Leelahavanichkul, A. Sepsis Encephalopathy Is Partly Mediated by miR370-3p-Induced Mitochondrial Injury but Attenuated by BAM15 in Cecal Ligation and Puncture Sepsis Male Mice. Int. J. Mol. Sci. 2022, 23, 5445. [Google Scholar] [CrossRef]
- Thim-Uam, A.; Makjaroen, J.; Issara-Amphorn, J.; Saisorn, W.; Wannigama, D.L.; Chancharoenthana, W.; Leelahavanichkul, A. Enhanced Bacteremia in Dextran Sulfate-Induced Colitis in Splenectomy Mice Correlates with Gut Dysbiosis and LPS Tolerance. Int. J. Mol. Sci. 2022, 23, 1676. [Google Scholar] [CrossRef]
- Ondee, T.; Surawut, S.; Taratummarat, S.; Hirankarn, N.; Palaga, T.; Pisitkun, P.; Pisitkun, T.; Leelahavanichkul, A. Fc Gamma Receptor IIB Deficient Mice: A Lupus Model with Increased Endotoxin Tolerance-Related Sepsis Susceptibility. Shock 2017, 47, 743–752. [Google Scholar] [CrossRef] [PubMed]
- Seeley, J.J.; Ghosh, S. Molecular mechanisms of innate memory and tolerance to LPS. J. Leukoc. Biol. 2017, 101, 107–119. [Google Scholar] [CrossRef] [PubMed]
- Gillen, J.; Ondee, T.; Gurusamy, D.; Issara-Amphorn, J.; Manes, N.P.; Yoon, S.H.; Leelahavanichkul, A.; Nita-Lazar, A. LPS Tolerance Inhibits Cellular Respiration and Induces Global Changes in the Macrophage Secretome. Biomolecules 2021, 11, 164. [Google Scholar] [CrossRef]
- Lopez-Collazo, E.; del Fresno, C. Pathophysiology of endotoxin tolerance: Mechanisms and clinical consequences. Crit. Care 2013, 17, 242. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Naler, L.B.; Hsieh, Y.P.; Geng, S.; Zhou, Z.; Li, L.; Lu, C. Epigenomic and transcriptomic analyses reveal differences between low-grade inflammation and severe exhaustion in LPS-challenged murine monocytes. Commun. Biol. 2022, 5, 102. [Google Scholar] [CrossRef]
- Koos, B.; Moderegger, E.L.; Rump, K.; Nowak, H.; Willemsen, K.; Holtkamp, C.; Thon, P.; Adamzik, M.; Rahmel, T. LPS-Induced Endotoxemia Evokes Epigenetic Alterations in Mitochondrial DNA That Impacts Inflammatory Response. Cells 2020, 9, 2282. [Google Scholar] [CrossRef]
- Lacal, I.; Ventura, R. Epigenetic Inheritance: Concepts, Mechanisms and Perspectives. Front. Mol. Neurosci. 2018, 11, 292. [Google Scholar] [CrossRef] [Green Version]
- Han, M.; Jia, L.; Lv, W.; Wang, L.; Cui, W. Epigenetic Enzyme Mutations: Role in Tumorigenesis and Molecular Inhibitors. Front. Oncol. 2019, 9, 194. [Google Scholar] [CrossRef]
- Casorelli, I.; Russo, M.T.; Bignami, M. Role of mismatch repair and MGMT in response to anticancer therapies. Anti-Cancer Agents Med. Chem. 2008, 8, 368–380. [Google Scholar] [CrossRef]
- Sharma, S.; Salehi, F.; Scheithauer, B.W.; Rotondo, F.; Syro, L.V.; Kovacs, K. Role of MGMT in tumor development, progression, diagnosis, treatment and prognosis. Anticancer Res. 2009, 29, 3759–3768. [Google Scholar]
- Rye, P.T.; Delaney, J.C.; Netirojjanakul, C.; Sun, D.X.; Liu, J.Z.; Essigmann, J.M. Mismatch repair proteins collaborate with methyltransferases in the repair of O(6)-methylguanine. DNA Repair 2008, 7, 170–176. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Drablos, F.; Feyzi, E.; Aas, P.A.; Vaagbo, C.B.; Kavli, B.; Bratlie, M.S.; Pena-Diaz, J.; Otterlei, M.; Slupphaug, G.; Krokan, H.E. Alkylation damage in DNA and RNA--repair mechanisms and medical significance. DNA Repair 2004, 3, 1389–1407. [Google Scholar] [CrossRef] [PubMed]
- Klapacz, J.; Pottenger, L.H.; Engelward, B.P.; Heinen, C.D.; Johnson, G.E.; Clewell, R.A.; Carmichael, P.L.; Adeleye, Y.; Andersen, M.E. Contributions of DNA repair and damage response pathways to the non-linear genotoxic responses of alkylating agents. Mutat. Res. Mol. Mech. Mutagen. 2016, 767, 77–91. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Warren, J.J.; Forsberg, L.J.; Beese, L.S. The structural basis for the mutagenicity of O(6)-methyl-guanine lesions. Proc. Natl. Acad. Sci. USA 2006, 103, 19701–19706. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Kay, J.; Thadhani, E.; Samson, L.; Engelward, B. Inflammation-induced DNA damage, mutations and cancer. DNA Repair 2019, 83, 102673. [Google Scholar] [CrossRef]
- Ezerskyte, M.; Paredes, J.A.; Malvezzi, S.; Burns, J.A.; Margison, G.P.; Olsson, M.; Scicchitano, D.A.; Dreij, K. O(6)-methylguanine-induced transcriptional mutagenesis reduces p53 tumor-suppressor function. Proc. Natl. Acad. Sci. USA 2018, 115, 4731–4736. [Google Scholar] [CrossRef] [Green Version]
- Christmann, M.; Verbeek, B.; Roos, W.P.; Kaina, B. O(6)-Methylguanine-DNA methyltransferase (MGMT) in normal tissues and tumors: Enzyme activity, promoter methylation and immunohistochemistry. Biochim. Biophys. Acta 2011, 1816, 179–190. [Google Scholar] [CrossRef]
- Kirkham, P. Oxidative stress and macrophage function: A failure to resolve the inflammatory response. Biochem. Soc. Trans. 2007, 35, 284–287. [Google Scholar] [CrossRef] [Green Version]
- Tang, C.H.; Wei, W.; Liu, L. Regulation of DNA repair by S-nitrosylation. Biochim. Biophys. Acta 2012, 1820, 730–735. [Google Scholar] [CrossRef] [Green Version]
- Colonna, M. DNA damage response impacts macrophage functions. Blood 2015, 126, 2440–2442. [Google Scholar] [CrossRef] [Green Version]
- Cao, L.; Zhu, T.; Lang, X.; Jia, S.; Yang, Y.; Zhu, C.; Wang, Y.; Feng, S.; Wang, C.; Zhang, P.; et al. Inhibiting DNA Methylation Improves Survival in Severe Sepsis by Regulating NF-kappaB Pathway. Front. Immunol. 2020, 11, 1360. [Google Scholar] [CrossRef]
- Wang, X.; Cao, Q.; Yu, L.; Shi, H.; Xue, B.; Shi, H. Epigenetic regulation of macrophage polarization and inflammation by DNA methylation in obesity. JCI Insight 2016, 1, e87748. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Khan, O.A.; Ranson, M.; Michael, M.; Olver, I.; Levitt, N.C.; Mortimer, P.; Watson, A.J.; Margison, G.P.; Midgley, R.; Middleton, M.R. A phase II trial of lomeguatrib and temozolomide in metastatic colorectal cancer. Br. J. Cancer 2008, 98, 1614–1618. [Google Scholar] [CrossRef] [PubMed]
- Rumienczyk, I.; Kulecka, M.; Statkiewicz, M.; Ostrowski, J.; Mikula, M. Oncology Drug Repurposing for Sepsis Treatment. Biomedicines 2022, 10, 921. [Google Scholar] [CrossRef]
- Benjaskulluecha, S.; Boonmee, A.; Pattarakankul, T.; Wongprom, B.; Klomsing, J.; Palaga, T. Screening of compounds to identify novel epigenetic regulatory factors that affect innate immune memory in macrophages. Sci. Rep. 2022, 12, 1912. [Google Scholar] [CrossRef]
- Pegg, A.E. Multifaceted roles of alkyltransferase and related proteins in DNA repair, DNA damage, resistance to chemotherapy, and research tools. Chem. Res. Toxicol. 2011, 24, 618–639. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Cheng, Y.; He, C.; Wang, M.; Ma, X.; Mo, F.; Yang, S.; Han, J.; Wei, X. Targeting epigenetic regulators for cancer therapy: Mechanisms and advances in clinical trials. Signal Transduct. Target. Ther. 2019, 4, 62. [Google Scholar] [CrossRef] [Green Version]
- Fernando, L.; Nguyen, V.; Hansen, T.; Golden, A.; Allen, A. Loss of proteasome subunit RPN-12 causes an increased mean lifespan at a higher temperature in C. elegans. MicroPubl. Biol. 2020, 2020. [Google Scholar] [CrossRef]
- Murata, S.; Udono, H.; Tanahashi, N.; Hamada, N.; Watanabe, K.; Adachi, K.; Yamano, T.; Yui, K.; Kobayashi, N.; Kasahara, M.; et al. Immunoproteasome assembly and antigen presentation in mice lacking both PA28alpha and PA28beta. EMBO J. 2001, 20, 5898–5907. [Google Scholar] [CrossRef] [Green Version]
- Flotynska, J.; Klause, D.; Kulecki, M.; Cieluch, A.; Chomicka-Pawlak, R.; Zozulinska-Ziolkiewicz, D.; Uruska, A. Higher NADH Dehydrogenase [Ubiquinone] Iron-Sulfur Protein 8 (NDUFS8) Serum Levels Correlate with Better Insulin Sensitivity in Type 1 Diabetes. Curr. Issues Mol. Biol. 2022, 44, 3872–3883. [Google Scholar] [CrossRef]
- Yu, J.; Hederstedt, L.; Piggot, P.J. The cytochrome bc complex (menaquinone:cytochrome c reductase) in Bacillus subtilis has a nontraditional subunit organization. J. Bacteriol. 1995, 177, 6751–6760. [Google Scholar] [CrossRef] [Green Version]
- Zhao, Y.; Sun, X.; Nie, X.; Sun, L.; Tang, T.S.; Chen, D.; Sun, Q. COX5B regulates MAVS-mediated antiviral signaling through interaction with ATG5 and repressing ROS production. PLoS Pathog. 2012, 8, e1003086. [Google Scholar] [CrossRef] [PubMed]
- Antoniel, M.; Jones, K.; Antonucci, S.; Spolaore, B.; Fogolari, F.; Petronilli, V.; Giorgio, V.; Carraro, M.; Di Lisa, F.; Forte, M.; et al. The unique histidine in OSCP subunit of F-ATP synthase mediates inhibition of the permeability transition pore by acidic pH. EMBO Rep. 2018, 19, 257–268. [Google Scholar] [CrossRef] [PubMed]
- Sweder, K.; Madura, K. Regulation of repair by the 26S proteasome. J. Biomed. Biotechnol. 2002, 2, 94–105. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Sakai, W.; Yuasa-Sunagawa, M.; Kusakabe, M.; Kishimoto, A.; Matsui, T.; Kaneko, Y.; Akagi, J.I.; Huyghe, N.; Ikura, M.; Ikura, T.; et al. Functional impacts of the ubiquitin-proteasome system on DNA damage recognition in global genome nucleotide excision repair. Sci. Rep. 2020, 10, 19704. [Google Scholar] [CrossRef]
- Lopes, A.F.C. Mitochondrial metabolism and DNA methylation: A review of the interaction between two genomes. Clin. Epigenet. 2020, 12, 182. [Google Scholar] [CrossRef] [PubMed]
- Chen, J.; Wu, Y.; Sun, Y.; Dong, X.; Wang, Z.; Zhang, Z.; Xiao, Y.; Dong, G. Bacterial Lipopolysaccharide Induced Alterations of Genome-Wide DNA Methylation and Promoter Methylation of Lactation-Related Genes in Bovine Mammary Epithelial Cells. Toxins 2019, 11, 298. [Google Scholar] [CrossRef] [Green Version]
- Angrisano, T.; Pero, R.; Brancaccio, M.; Coretti, L.; Florio, E.; Pezone, A.; Calabro, V.; Falco, G.; Keller, S.; Lembo, F.; et al. Cyclical DNA Methylation and Histone Changes Are Induced by LPS to Activate COX-2 in Human Intestinal Epithelial Cells. PLoS ONE 2016, 11, e0156671. [Google Scholar] [CrossRef] [PubMed]
- Podhorecka, M.; Skladanowski, A.; Bozko, P. H2AX Phosphorylation: Its Role in DNA Damage Response and Cancer Therapy. J. Nucleic Acids 2010, 2010, 920161. [Google Scholar] [CrossRef] [Green Version]
- Seminotti, B.; Amaral, A.U.; Grings, M.; Ribeiro, C.A.J.; Leipnitz, G.; Wajner, M. Lipopolysaccharide-Elicited Systemic Inflammation Induces Selective Vulnerability of Cerebral Cortex and Striatum of Developing Glutaryl-CoA Dehydrogenase Deficient (Gcdh(-/-)) Mice to Oxidative Stress. Neurotox. Res. 2020, 38, 1024–1036. [Google Scholar] [CrossRef]
- Joo, Y.M.; Chae, M.K.; Hwang, S.Y.; Jin, S.C.; Lee, T.R.; Cha, W.C.; Jo, I.J.; Sim, M.S.; Song, K.J.; Jeong, Y.K.; et al. Impact of timely antibiotic administration on outcomes in patients with severe sepsis and septic shock in the emergency department. Clin. Exp. Emerg. Med. 2014, 1, 35–40. [Google Scholar] [CrossRef]
- Leentjens, J.; Kox, M.; van der Hoeven, J.G.; Netea, M.G.; Pickkers, P. Immunotherapy for the adjunctive treatment of sepsis: From immunosuppression to immunostimulation. Time for a paradigm change? Am. J. Respir. Crit. Care Med. 2013, 187, 1287–1293. [Google Scholar] [CrossRef] [PubMed]
- Tungsanga, S.; Panpetch, W.; Bhunyakarnjanarat, T.; Udompornpitak, K.; Katavetin, P.; Chancharoenthana, W.; Chatthanathon, P.; Somboonna, N.; Tungsanga, K.; Tumwasorn, S.; et al. Uremia-Induced Gut Barrier Defect in 5/6 Nephrectomized Mice Is Worsened by Candida Administration through a Synergy of Uremic Toxin, Lipopolysaccharide, and (1➔3)-β-D-Glucan, but Is Attenuated by Lacticaseibacillus rhamnosus L34. Int. J. Mol. Sci. 2022, 23, 2511. [Google Scholar] [CrossRef] [PubMed]
- Visitchanakun, P.; Panpetch, W.; Saisorn, W.; Chatthanathon, P.; Wannigama, D.L.; Thim-Uam, A.; Svasti, S.; Fucharoen, S.; Somboonna, N.; Leelahavanichkul, A. Increased susceptibility to dextran sulfate-induced mucositis of iron-overload beta-thalassemia mice, another endogenous cause of septicemia in thalassemia. Clin. Sci. 2021, 135, 1467–1486. [Google Scholar] [CrossRef] [PubMed]
- Boonhai, S.; Bootdee, K.; Saisorn, W.; Takkavatakarn, K.; Sitticharoenchai, P.; Tungsanga, S.; Tiranathanagul, K.; Leelahavanichkul, A. TMAO reductase, a biomarker for gut permeability defect induced inflammation, in mouse model of chronic kidney disease and dextran sulfate solution-induced mucositis. Asian Pac. J. Allergy Immunol. 2021. Epub ahead of print. [Google Scholar] [CrossRef]
- Hiengrach, P.; Panpetch, W.; Chindamporn, A.; Leelahavanichkul, A. Macrophage depletion alters bacterial gut microbiota partly through fungal overgrowth in feces that worsens cecal ligation and puncture sepsis mice. Sci. Rep. 2022, 12, 9345. [Google Scholar] [CrossRef] [PubMed]
- Doi, K.; Leelahavanichkul, A.; Yuen, P.S.; Star, R.A. Animal models of sepsis and sepsis-induced kidney injury. J. Clin. Investig. 2009, 119, 2868–2878. [Google Scholar] [CrossRef] [Green Version]
- Binmama, S.; Dang, C.P.; Visitchanakun, P.; Hiengrach, P.; Somboonna, N.; Cheibchalard, T.; Pisitkun, P.; Chindamporn, A.; Leelahavanichkul, A. Beta-Glucan from S. cerevisiae Protected AOM-Induced Colon Cancer in cGAS-Deficient Mice Partly through Dectin-1-Manipulated Macrophage Cell Energy. Int. J. Mol. Sci. 2022, 23, 10951. [Google Scholar] [CrossRef]
- Charoensappakit, A.; Sae-Khow, K.; Leelahavanichkul, A. Gut Barrier Damage and Gut Translocation of Pathogen Molecules in Lupus, an Impact of Innate Immunity (Macrophages and Neutrophils) in Autoimmune Disease. Int. J. Mol. Sci. 2022, 23, 8223. [Google Scholar] [CrossRef]
- Marshall, R.S.; Vierstra, R.D. Dynamic Regulation of the 26S Proteasome: From Synthesis to Degradation. Front. Mol. Biosci. 2019, 6, 40. [Google Scholar] [CrossRef] [Green Version]
- Rousseau, A.; Bertolotti, A. Regulation of proteasome assembly and activity in health and disease. Nat. Rev. Mol. Cell Biol. 2018, 19, 697–712. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Silswal, N.; Reis, J.; Qureshi, A.A.; Papasian, C.; Qureshi, N. of Mice and Men: Proteasome’s Role in LPS-Induced Inflammation and Tolerance. Shock 2017, 47, 445–454. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Reis, J.; Guan, X.Q.; Kisselev, A.F.; Papasian, C.J.; Qureshi, A.A.; Morrison, D.C.; Van Way, C.W., 3rd; Vogel, S.N.; Qureshi, N. LPS-induced formation of immunoproteasomes: TNF-alpha and nitric oxide production are regulated by altered composition of proteasome-active sites. Cell Biochem. Biophys. 2011, 60, 77–88. [Google Scholar] [CrossRef] [Green Version]
- Hansen, M.E.; Simmons, K.J.; Tippetts, T.S.; Thatcher, M.O.; Saito, R.R.; Hubbard, S.T.; Trumbull, A.M.; Parker, B.A.; Taylor, O.J.; Bikman, B.T. Lipopolysaccharide Disrupts Mitochondrial Physiology in Skeletal Muscle via Disparate Effects on Sphingolipid Metabolism. Shock 2015, 44, 585–592. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Strichman-Almashanu, L.Z.; Lee, R.S.; Onyango, P.O.; Perlman, E.; Flam, F.; Frieman, M.B.; Feinberg, A.P. A genome-wide screen for normally methylated human CpG islands that can identify novel imprinted genes. Genome Res. 2002, 12, 543–554. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Saxonov, S.; Berg, P.; Brutlag, D.L. A genome-wide analysis of CpG dinucleotides in the human genome distinguishes two distinct classes of promoters. Proc. Natl. Acad. Sci. USA 2006, 103, 1412–1417. [Google Scholar] [CrossRef] [Green Version]
- Ezponda, T.; Licht, J.D. Molecular pathways: Deregulation of histone h3 lysine 27 methylation in cancer-different paths, same destination. Clin. Cancer Res. 2014, 20, 5001–5008. [Google Scholar] [CrossRef] [Green Version]
- Nichol, J.N.; Dupere-Richer, D.; Ezponda, T.; Licht, J.D.; Miller, W.H., Jr. H3K27 Methylation: A Focal Point of Epigenetic Deregulation in Cancer. Adv. Cancer Res. 2016, 131, 59–95. [Google Scholar] [CrossRef] [Green Version]
- Yue, D.; Wang, Z.; Yang, Y.; Hu, Z.; Luo, G.; Wang, F. EZH2 inhibitor GSK343 inhibits sepsis-induced intestinal disorders. Exp. Ther. Med. 2021, 21, 437. [Google Scholar] [CrossRef]
- Jin, B.; Li, Y.; Robertson, K.D. DNA methylation: Superior or subordinate in the epigenetic hierarchy? Genes Cancer 2011, 2, 607–617. [Google Scholar] [CrossRef] [Green Version]
- Du, Q.; Luu, P.L.; Stirzaker, C.; Clark, S.J. Methyl-CpG-binding domain proteins: Readers of the epigenome. Epigenomics 2015, 7, 1051–1073. [Google Scholar] [CrossRef] [PubMed]
- Moore, L.D.; Le, T.; Fan, G. DNA methylation and its basic function. Neuropsychopharmacology 2013, 38, 23–38. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Thomas, B.; Matson, S.; Chopra, V.; Sun, L.; Sharma, S.; Hersch, S.; Rosas, H.D.; Scherzer, C.; Ferrante, R.; Matson, W. A novel method for detecting 7-methyl guanine reveals aberrant methylation levels in Huntington disease. Anal. Biochem. 2013, 436, 112–120. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Fan, C.H.; Liu, W.L.; Cao, H.; Wen, C.; Chen, L.; Jiang, G. O6-methylguanine DNA methyltransferase as a promising target for the treatment of temozolomide-resistant gliomas. Cell Death Dis. 2013, 4, e876. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Yu, W.; Zhang, L.; Wei, Q.; Shao, A. O(6)-Methylguanine-DNA Methyltransferase (MGMT): Challenges and New Opportunities in Glioma Chemotherapy. Front. Oncol. 2019, 9, 1547. [Google Scholar] [CrossRef] [Green Version]
- Gopisetty, G.; Ramachandran, K.; Singal, R. DNA methylation and apoptosis. Mol. Immunol. 2006, 43, 1729–1740. [Google Scholar] [CrossRef]
- Kunanopparat, A.; Leelahavanichkul, A.; Visitchanakun, P.; Kueanjinda, P.; Phuengmaung, P.; Sae-Khow, K.; Boonmee, A.; Benjaskulluecha, S.; Palaga, T.; Hirankarn, N. The Regulatory Roles of Ezh2 in Response to Lipopolysaccharide (LPS) in Macrophages and Mice with Conditional Ezh2 Deletion with LysM-Cre System. Int. J. Mol. Sci. 2023, 24, 5363. [Google Scholar] [CrossRef]
- Zhang, Q.; Sun, H.; Zhuang, S.; Liu, N.; Bao, X.; Liu, X.; Ren, H.; Lv, D.; Li, Z.; Bai, J.; et al. Novel pharmacological inhibition of EZH2 attenuates septic shock by altering innate inflammatory responses to sepsis. Int. Immunopharmacol. 2019, 76, 105899. [Google Scholar] [CrossRef]
- Zhao, D.; Li, Z.; Liu, X.; Liu, N.; Bao, X.; Sun, H.; Meng, Q.; Ren, H.; Bai, J.; Zhou, X.; et al. Lymphocyte expression of EZH2 is associated with mortality and secondary infectious complications in sepsis. Int. Immunopharmacol. 2020, 89, 107042. [Google Scholar] [CrossRef]
- Das, A.; Henderson, F.C., Jr.; Alshareef, M.; Porto, G.B.F.; Kanginakudru, I.; Infinger, L.K.; Vandergrift, W.A., 3rd; Lindhorst, S.M.; Varma, A.K.; Patel, S.J.; et al. MGMT-inhibitor in combination with TGF-betaRI inhibitor or CDK 4/6 inhibitor increases temozolomide sensitivity in temozolomide-resistant glioblastoma cells. Clin. Transl. Oncol. 2021, 23, 612–619. [Google Scholar] [CrossRef]
- Davies, R.; O’Dea, K.; Gordon, A. Immune therapy in sepsis: Are we ready to try again? J. Intensive Care Soc. 2018, 19, 326–344. [Google Scholar] [CrossRef] [Green Version]
- Ruenjaiman, V.; Butta, P.; Leu, Y.W.; Pongpanich, M.; Leelahavanichkul, A.; Kueanjinda, P.; Palaga, T. Profile of Histone H3 Lysine 4 Trimethylation and the Effect of Lipopolysaccharide/Immune Complex-Activated Macrophages on Endotoxemia. Front. Immunol. 2019, 10, 2956. [Google Scholar] [CrossRef] [PubMed]
- Leelahavanichkul, A.; Somparn, P.; Bootprapan, T.; Tu, H.; Tangtanatakul, P.; Nuengjumnong, R.; Worasilchai, N.; Tiranathanagul, K.; Eiam-ong, S.; Levine, M.; et al. High-dose ascorbate with low-dose amphotericin B attenuates severity of disease in a model of the reappearance of candidemia during sepsis in the mouse. Am. J. Physiol. Regul. Integr. Comp. Physiol. 2015, 309, R223–R234. [Google Scholar] [CrossRef] [Green Version]
- Vu, C.T.B.; Thammahong, A.; Leelahavanichkul, A.; Ritprajak, P. Alteration of macrophage immune phenotype in a murine sepsis model is associated with susceptibility to secondary fungal infection. Asian Pac. J. Allergy Immunol. 2022, 40, 162–171. [Google Scholar] [CrossRef]
- Vu, C.T.B.; Thammahong, A.; Yagita, H.; Azuma, M.; Hirankarn, N.; Ritprajak, P.; Leelahavanichkul, A. Blockade of PD-1 Attenuated Postsepsis Aspergillosis via the Activation of IFN-gamma and the Dampening of IL-10. Shock 2020, 53, 514–524. [Google Scholar] [CrossRef] [PubMed]
- Aleksandrov, R.; Hristova, R.; Stoynov, S.; Gospodinov, A. The Chromatin Response to Double-Strand DNA Breaks and Their Repair. Cells 2020, 9, 1853. [Google Scholar] [CrossRef]
- White, R.R.; Vijg, J. Do DNA Double-Strand Breaks Drive Aging? Mol. Cell 2016, 63, 729–738. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Cannan, W.J.; Pederson, D.S. Mechanisms and Consequences of Double-Strand DNA Break Formation in Chromatin. J. Cell. Physiol. 2016, 231, 3–14. [Google Scholar] [CrossRef] [Green Version]
- Glukhov, I.L.; Sirota, N.P.; Kuznetsova, E.A. DNA damage in human mononuclear cells induced by bacterial endotoxin. Bull. Exp. Biol. Med. 2008, 146, 301–303. [Google Scholar] [CrossRef]
- Kietzmann, T.; Petry, A.; Shvetsova, A.; Gerhold, J.M.; Gorlach, A. The epigenetic landscape related to reactive oxygen species formation in the cardiovascular system. Br. J. Pharmacol. 2017, 174, 1533–1554. [Google Scholar] [CrossRef] [Green Version]
- Alhmoud, J.F.; Woolley, J.F.; Al Moustafa, A.E.; Malki, M.I. DNA Damage/Repair Management in Cancers. Cancers 2020, 12, 10502. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Wyatt, M.D.; Pittman, D.L. Methylating agents and DNA repair responses: Methylated bases and sources of strand breaks. Chem. Res. Toxicol. 2006, 19, 1580–1594. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Griffin, S.; Branch, P.; Xu, Y.Z.; Karran, P. DNA mismatch binding and incision at modified guanine bases by extracts of mammalian cells: Implications for tolerance to DNA methylation damage. Biochemistry 1994, 33, 4787–4793. [Google Scholar] [CrossRef] [PubMed]
- Xaus, J.; Comalada, M.; Valledor, A.F.; Lloberas, J.; Lopez-Soriano, F.; Argiles, J.M.; Bogdan, C.; Celada, A. LPS induces apoptosis in macrophages mostly through the autocrine production of TNF-alpha. Blood 2000, 95, 3823–3831. [Google Scholar] [CrossRef]
- Canton, M.; Sanchez-Rodriguez, R.; Spera, I.; Venegas, F.C.; Favia, M.; Viola, A.; Castegna, A. Reactive Oxygen Species in Macrophages: Sources and Targets. Front. Immunol. 2021, 12, 734229. [Google Scholar] [CrossRef]
- Luo, M.; Bao, Z.; Xu, F.; Wang, X.; Li, F.; Li, W.; Chen, Z.; Ying, S.; Shen, H. Unrepaired DNA damage in macrophages causes elevation of particulate matter- induced airway inflammatory response. Aging 2018, 10, 549–560. [Google Scholar] [CrossRef] [Green Version]
- Kraus, A.; McKeague, M.; Seiwert, N.; Nagel, G.; Geisen, S.M.; Ziegler, N.; Trantakis, I.A.; Kaina, B.; Thomas, A.D.; Sturla, S.J.; et al. Immunological and mass spectrometry-based approaches to determine thresholds of the mutagenic DNA adduct O(6)-methylguanine in vivo. Arch. Toxicol. 2019, 93, 559–572. [Google Scholar] [CrossRef]
- Aguilera, A.; Gomez-Gonzalez, B. Genome instability: A mechanistic view of its causes and consequences. Nat. Rev. Genet. 2008, 9, 204–217. [Google Scholar] [CrossRef]
- Snezhkina, A.V.; Kudryavtseva, A.V.; Kardymon, O.L.; Savvateeva, M.V.; Melnikova, N.V.; Krasnov, G.S.; Dmitriev, A.A. ROS Generation and Antioxidant Defense Systems in Normal and Malignant Cells. Oxidative Med. Cell. Longev. 2019, 2019, 6175804. [Google Scholar] [CrossRef] [Green Version]
- Ranson, M.; Hersey, P.; Thompson, D.; Beith, J.; McArthur, G.A.; Haydon, A.; Davis, I.D.; Kefford, R.F.; Mortimer, P.; Harris, P.A.; et al. Randomized trial of the combination of lomeguatrib and temozolomide compared with temozolomide alone in chemotherapy naive patients with metastatic cutaneous melanoma. J. Clin. Oncol. 2007, 25, 2540–2545. [Google Scholar] [CrossRef] [Green Version]
- Chancharoenthana, W.; Kamolratanakul, S.; Schultz, M.J.; Leelahavanichkul, A. The leaky gut and the gut microbiome in sepsis—Targets in research and treatment. Clin. Sci. 2023, 137, 645–662. [Google Scholar] [CrossRef]
- Arango Duque, G.; Descoteaux, A. Macrophage cytokines: Involvement in immunity and infectious diseases. Front. Immunol. 2014, 5, 491. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Yu, H.; Yang, Z.; Li, F.; Xu, L.; Sun, Y. Cell-mediated targeting drugs delivery systems. Drug Deliv. 2020, 27, 1425–1437. [Google Scholar] [CrossRef] [PubMed]
- Greco, M.; Mazzei, A.; Suppressa, S.; Palumbo, C.; Verri, T.; Lobreglio, G. Human Leukocyte Antigen-DR Isotype Expression in Monocytes and T Cells Interferon-Gamma Release Assay in Septic Patients and Correlation with Clinical Outcome. J. Clin. Med. Res. 2021, 13, 293–303. [Google Scholar] [CrossRef] [PubMed]
- Mansfield, S.; Griessl, M.; Gutknecht, M.; Cook, C.H. Sepsis and cytomegalovirus: Foes or conspirators? Med. Microbiol. Immunol. 2015, 204, 431–437. [Google Scholar] [CrossRef]
- Aloisi, C.M.N.; Sturla, S.J.; Gahlon, H.L. A gene-targeted polymerase-mediated strategy to identify O(6)-methylguanine damage. Chem. Commun. 2019, 55, 3895–3898. [Google Scholar] [CrossRef] [Green Version]
- Song, J.; Park, D.W.; Moon, S.; Cho, H.J.; Park, J.H.; Seok, H.; Choi, W.S. Diagnostic and prognostic value of interleukin-6, pentraxin 3, and procalcitonin levels among sepsis and septic shock patients: A prospective controlled study according to the Sepsis-3 definitions. BMC Infect. Dis. 2019, 19, 968. [Google Scholar] [CrossRef] [Green Version]
- Wang, L.; Zhao, H.; Wang, D. Inflammatory cytokine expression in patients with sepsis at an intensive care unit. Exp. Ther. Med. 2018, 16, 2126–2131. [Google Scholar] [CrossRef] [Green Version]
- Hiengrach, P.; Visitchanakun, P.; Finkelman, M.A.; Chancharoenthana, W.; Leelahavanichkul, A. More Prominent Inflammatory Response to Pachyman than to Whole-Glucan Particle and Oat-beta-Glucans in Dextran Sulfate-Induced Mucositis Mice and Mouse Injection through Proinflammatory Macrophages. Int. J. Mol. Sci. 2022, 23, 4026. [Google Scholar] [CrossRef]
- Udompornpitak, K.; Charoensappakit, A.; Sae-Khow, K.; Bhunyakarnjanarat, T.; Dang, C.P.; Saisorn, W.; Visitchanakun, P.; Phuengmaung, P.; Palaga, T.; Ritprajak, P.; et al. Obesity Exacerbates Lupus Activity in Fc Gamma Receptor IIb Deficient Lupus Mice Partly through Saturated Fatty Acid-Induced Gut Barrier Defect and Systemic Inflammation. J. Innate Immun. 2022, 15, 240–261. [Google Scholar] [CrossRef]
- Singkham-In, U.; Phuengmaung, P.; Makjaroen, J.; Saisorn, W.; Bhunyakarnjanarat, T.; Chatsuwan, T.; Chirathaworn, C.; Chancharoenthana, W.; Leelahavanichkul, A. Chlorhexidine Promotes Psl Expression in Pseudomonas aeruginosa That Enhances Cell Aggregation with Preserved Pathogenicity Demonstrates an Adaptation against Antiseptic. Int. J. Mol. Sci. 2022, 23, 8308. [Google Scholar] [CrossRef] [PubMed]
- Kirstein, A.; Schilling, D.; Combs, S.E.; Schmid, T.E. Lomeguatrib Increases the Radiosensitivity of MGMT Unmethylated Human Glioblastoma Multiforme Cell Lines. Int. J. Mol. Sci. 2021, 22, 6781. [Google Scholar] [CrossRef] [PubMed]
- Saisorn, W.; Saithong, S.; Phuengmaung, P.; Udompornpitak, K.; Bhunyakarnjanarat, T.; Visitchanakun, P.; Chareonsappakit, A.; Pisitkun, P.; Chiewchengchol, D.; Leelahavanichkul, A. Acute Kidney Injury Induced Lupus Exacerbation through the Enhanced Neutrophil Extracellular Traps (and Apoptosis) in Fcgr2b Deficient Lupus Mice with Renal Ischemia Reperfusion Injury. Front. Immunol. 2021, 12, 669162. [Google Scholar] [CrossRef] [PubMed]
- Tzafrir, T.; Wainstock, T.; Sheiner, E.; Miodownik, S.; Pariente, G. Fetal Growth Restriction and Long-Term Cardiovascular Morbidity of Offspring in Dichorionic-Diamniotic Twin Pregnancies. J. Clin. Med. 2023, 12, 1628. [Google Scholar] [CrossRef]
- Visitchanakun, P.; Tangtanatakul, P.; Trithiphen, O.; Soonthornchai, W.; Wongphoom, J.; Tachaboon, S.; Srisawat, N.; Leelahavanichkul, A. Plasma miR-370-3P as a Biomarker of Sepsis-Associated Encephalopathy, the Transcriptomic Profiling Analysis of Microrna-Arrays from Mouse Brains. Shock 2020, 54, 347–357. [Google Scholar] [CrossRef] [PubMed]
- Issara-Amphorn, J.; Surawut, S.; Worasilchai, N.; Thim-Uam, A.; Finkelman, M.; Chindamporn, A.; Palaga, T.; Hirankarn, N.; Pisitkun, P.; Leelahavanichkul, A. The Synergy of Endotoxin and (1-->3)-beta-D-Glucan, from Gut Translocation, Worsens Sepsis Severity in a Lupus Model of Fc Gamma Receptor IIb-Deficient Mice. J. Innate Immun. 2018, 10, 189–201. [Google Scholar] [CrossRef]
- Panpetch, W.; Somboonna, N.; Bulan, D.E.; Issara-Amphorn, J.; Finkelman, M.; Worasilchai, N.; Chindamporn, A.; Palaga, T.; Tumwasorn, S.; Leelahavanichkul, A. Oral administration of live- or heat-killed Candida albicans worsened cecal ligation and puncture sepsis in a murine model possibly due to an increased serum (1-->3)-beta-D-glucan. PLoS ONE 2017, 12, e0181439. [Google Scholar] [CrossRef] [Green Version]
- Ranson, M.; Middleton, M.R.; Bridgewater, J.; Lee, S.M.; Dawson, M.; Jowle, D.; Halbert, G.; Waller, S.; McGrath, H.; Gumbrell, L.; et al. Lomeguatrib, a potent inhibitor of O6-alkylguanine-DNA-alkyltransferase: Phase I safety, pharmacodynamic, and pharmacokinetic trial and evaluation in combination with temozolomide in patients with advanced solid tumors. Clin. Cancer Res. 2006, 12, 1577–1584. [Google Scholar] [CrossRef] [Green Version]
- Panpetch, W.; Phuengmaung, P.; Hiengrach, P.; Issara-Amphorn, J.; Cheibchalard, T.; Somboonna, N.; Tumwasorn, S.; Leelahavanichkul, A. Candida Worsens Klebsiella pneumoniae Induced-Sepsis in a Mouse Model with Low Dose Dextran Sulfate Solution through Gut Dysbiosis and Enhanced Inflammation. Int. J. Mol. Sci. 2022, 23, 7050. [Google Scholar] [CrossRef]
- Panpetch, W.; Kullapanich, C.; Dang, C.P.; Visitchanakun, P.; Saisorn, W.; Wongphoom, J.; Wannigama, D.L.; Thim-Uam, A.; Patarakul, K.; Somboonna, N.; et al. Candida Administration Worsens Uremia-Induced Gut Leakage in Bilateral Nephrectomy Mice, an Impact of Gut Fungi and Organismal Molecules in Uremia. mSystems 2021, 6, e01187-20. [Google Scholar] [CrossRef]
- Makjaroen, J.; Thim-Uam, A.; Dang, C.P.; Pisitkun, T.; Somparn, P.; Leelahavanichkul, A. A Comparison Between 1 Day versus 7 Days of Sepsis in Mice with the Experiments on LPS-Activated Macrophages Support the Use of Intravenous Immunoglobulin for Sepsis Attenuation. J. Inflamm. Res. 2021, 14, 7243–7263. [Google Scholar] [CrossRef] [PubMed]
Name | Forward | Reverse |
---|---|---|
Inducible nitric oxide synthase (iNOS) | 5′-ACCCACATCTGGCAGAATGAG-3′ | 5′-AGCCATGACCTTTCGCATTAG-3′ |
Interleukin-1β (IL-1β) | 5′-GAAATGCCACCTTTTGACAGTG-3′ | 5′-TGGATGCTCTCATCAGGACAG-3′ |
Arginase-1 (Arg-1) | 5′-CTTGGCTTGCTTCGGAACTC-3′ | 5′-GGAGAAGGCGTTTGCTTAGTT-3′ |
Resistin-like molecule-α1 (Fizz-1) | 5′-GCCAGGTCCTGGAACCTTTC-3′ | 5′-GGAGCAGGGAGATGCAGATGA-3′ |
Transforming growth factor-β (TGF-β) | 5′-CAGAGCTGCGCTTGCAGAG-3′ | 5′-GTCAGCAGCCGGTTACCAAG-3′ |
Beta-actin (β-actin) | 5′-CGGTTCCGATGCCCTGAGGCTCTT-3′ | 5′-CGTCACACTTCATGATGGAATTGA-3′ |
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content. |
© 2023 by the authors. Licensee MDPI, Basel, Switzerland. This article is an open access article distributed under the terms and conditions of the Creative Commons Attribution (CC BY) license (https://creativecommons.org/licenses/by/4.0/).
Share and Cite
Sae-khow, K.; Phuengmaung, P.; Issara-Amphorn, J.; Makjaroen, J.; Visitchanakun, P.; Boonmee, A.; Benjaskulluecha, S.; Palaga, T.; Leelahavanichkul, A. Less Severe Polymicrobial Sepsis in Conditional mgmt-Deleted Mice Using LysM-Cre System, Impacts of DNA Methylation and MGMT Inhibitor in Sepsis. Int. J. Mol. Sci. 2023, 24, 10175. https://doi.org/10.3390/ijms241210175
Sae-khow K, Phuengmaung P, Issara-Amphorn J, Makjaroen J, Visitchanakun P, Boonmee A, Benjaskulluecha S, Palaga T, Leelahavanichkul A. Less Severe Polymicrobial Sepsis in Conditional mgmt-Deleted Mice Using LysM-Cre System, Impacts of DNA Methylation and MGMT Inhibitor in Sepsis. International Journal of Molecular Sciences. 2023; 24(12):10175. https://doi.org/10.3390/ijms241210175
Chicago/Turabian StyleSae-khow, Kritsanawan, Pornpimol Phuengmaung, Jiraphorn Issara-Amphorn, Jiradej Makjaroen, Peerapat Visitchanakun, Atsadang Boonmee, Salisa Benjaskulluecha, Tanapat Palaga, and Asada Leelahavanichkul. 2023. "Less Severe Polymicrobial Sepsis in Conditional mgmt-Deleted Mice Using LysM-Cre System, Impacts of DNA Methylation and MGMT Inhibitor in Sepsis" International Journal of Molecular Sciences 24, no. 12: 10175. https://doi.org/10.3390/ijms241210175
APA StyleSae-khow, K., Phuengmaung, P., Issara-Amphorn, J., Makjaroen, J., Visitchanakun, P., Boonmee, A., Benjaskulluecha, S., Palaga, T., & Leelahavanichkul, A. (2023). Less Severe Polymicrobial Sepsis in Conditional mgmt-Deleted Mice Using LysM-Cre System, Impacts of DNA Methylation and MGMT Inhibitor in Sepsis. International Journal of Molecular Sciences, 24(12), 10175. https://doi.org/10.3390/ijms241210175