1
|
Kang Z, Di L, Liu Y, Qu W, Zeng Y, Lu X, Wang L. The Correlation Between the Expression Levels of Serum miR-19b and SOCS-1 mRNA and Clinical Symptoms in Patients with Allergic Rhinitis. J Asthma Allergy 2025; 18:619-627. [PMID: 40292344 PMCID: PMC12032965 DOI: 10.2147/jaa.s518177] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2025] [Accepted: 04/07/2025] [Indexed: 04/30/2025] Open
Abstract
Objective To investigate the correlation between the expression levels of microRNA-19b (miR-19b) and suppressor of cytokine signaling 1 messenger RNA (SOCS-1 mRNA) in the serum of patients with allergic rhinitis (AR) and clinical symptoms. Methods This prospective study included a total of 86 patients with allergic rhinitis who were admitted to the People's Hospital Affiliated to Hubei University of Medicine from January 2022 to January 2023. Seventy healthy individuals were included in the control group. The case group was further divided into a mild group (n=45) and a moderate to severe group (n=41) according to the severity of AR. The expression levels of miR-19b and SOCS-1 mRNA in serum samples of the two groups were detected and compared using real-time fluorescence quantitative polymerase chain reaction (qRT-PCR) technique. The receiver operating characteristic (ROC) curve and the area under the curve (AUC) were utilized to evaluate the predictive value of miR-19b and SOCS-1 levels for the severity of AR. Results The expression level of miR-19b in the serum of the case group was 1.52±0.36, significantly higher than that of the control group (1.02±0.24, P<0.05). Logistic regression analysis indicated that low levels of serum miR-19b and SOCS-1 mRNA were independent risk factors for moderate to severe AR, with an odds ratio (OR) of 3.575 (95% CI: 1.572-8.133, P<0.001) for miR-19b and 3.725 (95% CI: 1.637-8.473, P<0.001) for SOCS-1 mRNA. ROC curve analysis demonstrated that the levels of serum miR-19b and SOCS-1 mRNA had high accuracy in predicting moderate to severe AR, with AUC values of 0.879 (95% CI: 0.791-0.940) and 0.795 (95% CI: 0.694-0.875), respectively. When combined for prediction, the efficacy was significantly higher than that of individual detection, with an AUC value of 0.923 (95% CI: 0.856-0.967), Z=3.261, P<0.01. Conclusion The expression levels of serum miR-19b and SOCS-1 mRNA are closely related to the severity of clinical symptoms in patients with AR and may serve as new biomarkers for evaluating the condition of AR and guiding treatment.
Collapse
Affiliation(s)
- Zhaopeng Kang
- Department of Andrology, Renmin Hospital, Hubei University of Medicine, Shiyan, Hubei, 442000, People’s Republic of China
| | - Lingling Di
- Department of Otolaryngology Head and Neck Surgery, General Hospital of Ningxia Medical University, Yinchuan, Ningxia Hui Autonomous Region, 756099, People’s Republic of China
| | - Yi Liu
- Department of Otolaryngology Head and Neck Surgery, Renmin Hospital, Hubei University of Medicine, Shiyan, Hubei, 442000, People’s Republic of China
| | - Wei Qu
- Department of Otolaryngology Head and Neck Surgery, Renmin Hospital, Hubei University of Medicine, Shiyan, Hubei, 442000, People’s Republic of China
| | - Yi Zeng
- Department of Otolaryngology Head and Neck Surgery, Renmin Hospital, Hubei University of Medicine, Shiyan, Hubei, 442000, People’s Republic of China
| | - Xue Lu
- Department of Otolaryngology Head and Neck Surgery, Renmin Hospital, Hubei University of Medicine, Shiyan, Hubei, 442000, People’s Republic of China
| | - Lixin Wang
- Department of Otolaryngology Head and Neck Surgery, Renmin Hospital, Hubei University of Medicine, Shiyan, Hubei, 442000, People’s Republic of China
| |
Collapse
|
2
|
Chen Y, Zhong W, Xie Z, Li B, Li H, Gao K, Ning Z. Suppressor of cytokine signaling 1 (SOCS1) inhibits antiviral responses to facilitate Senecavirus A infection by regulating the NF-κB signaling pathway. Virus Res 2022; 313:198748. [PMID: 35304133 DOI: 10.1016/j.virusres.2022.198748] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2022] [Revised: 03/12/2022] [Accepted: 03/14/2022] [Indexed: 11/19/2022]
Abstract
Senecavirus A (SVA) is a new virus inducing porcine idiopathic vesicular disease that causes significant economic losses. Although some progress has been made in etiological research, the role of host factors in SVA infection remains unclear. This study investigated the role of the host factor, suppressor of cytokine signaling 1 (SOCS1), in SVA infection. The expression of SOCS1 was significantly upregulated with infection of SVA in a dose-dependent manner, and SOCS1 inhibited the expression of type I interferons (IFN-α, IFN-β) and the production of interferon stimulating genes (ISGs) (ISG56, ISG54, PKR), thereby facilitating viral replication. Further results showed that inhibition of antiviral responses of SOCS1 was achieved by regulating the NF-κB signaling pathway, which attenuates the production of IFNs and pro-inflammatory cytokines. These findings provide a new perspective of SVA pathogenesis and may partially explain the persistence of this infection. Moreover, the data indicate that targeting SOCS1 can help in developing new agents against SVA infection.
Collapse
Affiliation(s)
- Yongjie Chen
- College of Veterinary Medicine, South China Agricultural University, Guangzhou 510642, China
| | - Wenxia Zhong
- College of Veterinary Medicine, South China Agricultural University, Guangzhou 510642, China
| | - Zhenxin Xie
- College of Veterinary Medicine, South China Agricultural University, Guangzhou 510642, China
| | - Baojian Li
- College of Veterinary Medicine, South China Agricultural University, Guangzhou 510642, China
| | - Huizi Li
- College of Veterinary Medicine, South China Agricultural University, Guangzhou 510642, China
| | - Kuipeng Gao
- College of Veterinary Medicine, South China Agricultural University, Guangzhou 510642, China
| | - Zhangyong Ning
- College of Veterinary Medicine, South China Agricultural University, Guangzhou 510642, China; Guangdong Laboratory for Lingnan Modern Agriculture, Maoming Branch, Maoming 525000, China.
| |
Collapse
|
3
|
Lee HJ, Hong WG, Woo Y, Ahn JH, Ko HJ, Kim H, Moon S, Hahn TW, Jung YM, Song DK, Jung YJ. Lysophosphatidylcholine Enhances Bactericidal Activity by Promoting Phagosome Maturation via the Activation of the NF-κB Pathway during Salmonella Infection in Mouse Macrophages. Mol Cells 2020; 43:989-1001. [PMID: 33250450 PMCID: PMC7772511 DOI: 10.14348/molcells.2020.0030] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2020] [Revised: 10/29/2020] [Accepted: 11/03/2020] [Indexed: 12/13/2022] Open
Abstract
Salmonella enterica serovar Typhimurium (S. Typhimurium) is a facultative intracellular pathogen that causes salmonellosis and mortality worldwide. S. Typhimurium infects macrophages and survives within phagosomes by avoiding the phagosome-lysosome fusion system. Phagosomes sequentially acquire different Rab GTPases during maturation and eventually fuse with acidic lysosomes. Lysophosphatidylcholine (LPC) is a bioactive lipid that is associated with the generation of chemoattractants and reactive oxygen species (ROS). In our previous study, LPC controlled the intracellular growth of Mycobacterium tuberculosis by promoting phagosome maturation. In this study, to verify whether LPC enhances phagosome maturation and regulates the intracellular growth of S. Typhimurium, macrophages were infected with S. Typhimurium. LPC decreased the intracellular bacterial burden, but it did not induce cytotoxicity in S. Typhimuriuminfected cells. In addition, combined administration of LPC and antibiotic significantly reduced the bacterial burden in the spleen and the liver. The ratios of the colocalization of intracellular S. Typhimurium with phagosome maturation markers, such as early endosome antigen 1 (EEA1) and lysosome-associated membrane protein 1 (LAMP-1), were significantly increased in LPC-treated cells. The expression level of cleaved cathepsin D was rapidly increased in LPCtreated cells during S. Typhimurium infection. Treatment with LPC enhanced ROS production, but it did not affect nitric oxide production in S. Typhimurium-infected cells. LPC also rapidly triggered the phosphorylation of IκBα during S. Typhimurium infection. These results suggest that LPC can improve phagosome maturation via ROS-induced activation of NF-κB pathway and thus may be developed as a therapeutic agent to control S. Typhimurium growth.
Collapse
Affiliation(s)
- Hyo-Ji Lee
- Department of Biological Sciences and Institute of Life Sciences, Kangwon National University, Chuncheon 2434, Korea
- Kangwon Radiation Convergence Research Support Center, Kangwon National University, Chuncheon 24341, Korea
| | - Wan-Gi Hong
- BIT Medical Convergence Graduate Program, Kangwon National University, Chuncheon 4341, Korea
| | - Yunseo Woo
- Department of Biological Sciences and Institute of Life Sciences, Kangwon National University, Chuncheon 2434, Korea
- Kangwon Radiation Convergence Research Support Center, Kangwon National University, Chuncheon 24341, Korea
| | - Jae-Hee Ahn
- Department of Pharmacy, Kangwon National University, Chuncheon 2441, Korea
| | - Hyun-Jeong Ko
- Department of Pharmacy, Kangwon National University, Chuncheon 2441, Korea
- Kangwon Radiation Convergence Research Support Center, Kangwon National University, Chuncheon 24341, Korea
| | - Hyeran Kim
- Department of Biological Sciences and Institute of Life Sciences, Kangwon National University, Chuncheon 2434, Korea
| | - Sungjin Moon
- Department of Biological Sciences and Institute of Life Sciences, Kangwon National University, Chuncheon 2434, Korea
- Kangwon Radiation Convergence Research Support Center, Kangwon National University, Chuncheon 24341, Korea
| | - Tae-Wook Hahn
- Department of Veterinary Medicine, Kangwon National University, Chuncheon 231, Korea
| | - Young Mee Jung
- Department of Chemistry, Kangwon National University, Chuncheon 24341, Korea
- Kangwon Radiation Convergence Research Support Center, Kangwon National University, Chuncheon 24341, Korea
| | - Dong-Keun Song
- Department of Pharmacology, College of Medicine, Hallym University, Chuncheon 24252, Korea
| | - Yu-Jin Jung
- Department of Biological Sciences and Institute of Life Sciences, Kangwon National University, Chuncheon 2434, Korea
- BIT Medical Convergence Graduate Program, Kangwon National University, Chuncheon 4341, Korea
- Kangwon Radiation Convergence Research Support Center, Kangwon National University, Chuncheon 24341, Korea
| |
Collapse
|
4
|
Vreman S, Stockhofe-Zurwieden N, Popma-de Graaf DJ, Savelkoul HFJ, Barnier-Quer C, Collin N, Collins D, McDaid D, Moore AC, Rebel JMJ. Immune responses induced by inactivated porcine reproductive and respiratory syndrome virus (PRRSV) vaccine in neonatal pigs using different adjuvants. Vet Immunol Immunopathol 2020; 232:110170. [PMID: 33383553 DOI: 10.1016/j.vetimm.2020.110170] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2019] [Revised: 12/12/2020] [Accepted: 12/13/2020] [Indexed: 12/20/2022]
Abstract
Vaccination of neonatal pigs could be supportive to prevent porcine reproductive and respiratory syndrome virus (PRRSV), which is an important porcine pathogen causing worldwide welfare and health problems in pigs of different age classes. However, neonatal immunity substantially differs to adults, thus different vaccines may be required in neonateal pigs. We examined if the immunogenicity and efficacy of inactivated PRRSV (iPRRSV) vaccines in neonatal pigs could be improved with adjuvants containing oil-in water (O/W) emulsions with or without Toll-like receptor (TLR) agonists and by altering the delivery route from intramuscular (i.m.) to the skin. Three-day-old PRRSV-naïve piglets (n = 54, divided in 6 groups) received a prime vaccination and a booster vaccination four weeks later. The vaccine formulations consisted of different O/W emulsions (Montanide™ ISA28RVG (ISA28)), a squalene in water emulsion (SWE) for i.m. or a Stable Emulsion (SE) with squalene for skin vaccination) and/or a mixture of TLR1/2, 7/8 and 9 agonists (TLRa) combined with iPRRSV strain 07V063. These vaccines were delivered either i.m. (ISA28, SWE, TLRa or SWE + TLRa) or into the skin (skiSE + TLRa) with dissolving microneedle (DMN)-patches. All animals received a challenge with homologous PRRSV three weeks after booster vaccination. Specific antibodies, IFN-γ production and viremia were measured at several time-points after vaccination and/or challenge, while lung pathology was studied at necropsy. After booster vaccination, only ISA28 induced a specific antibody response while a specific T-cell IFN-γ response was generated in the SWE group, that was lower for ISA28, and absent in the other groups. This suggests that prime vaccination in neonates induced a specific immune response after booster vaccination, dependent on the emulsion formulation, but not dependent on the presence of the TLRa or delivery route. Despite the measured immune responses none of the vaccines showed any efficacy. Further research focused on the early immune response in draining lymph nodes is needed to elucidate the potential of TLR agonists in vaccines for neonatal pigs.
Collapse
Affiliation(s)
- Sandra Vreman
- Wageningen Bioveterinary Research, Wageningen University & Research, P.O. Box 29703, 2502 LS, The Hague, the Netherlands.
| | - Norbert Stockhofe-Zurwieden
- Wageningen Bioveterinary Research, Wageningen University & Research, P.O. Box 29703, 2502 LS, The Hague, the Netherlands
| | - Ditta J Popma-de Graaf
- Wageningen Bioveterinary Research, Wageningen University & Research, P.O. Box 29703, 2502 LS, The Hague, the Netherlands
| | - Huub F J Savelkoul
- Cell Biology & Immunology Group, Wageningen University & Research P.O. Box 338, 6700 HA, Wageningen, the Netherlands
| | - C Barnier-Quer
- Vaccine Formulation Laboratory, University of Lausanne, Epalinges, Switzerland
| | - N Collin
- Vaccine Formulation Laboratory, University of Lausanne, Epalinges, Switzerland
| | | | | | - Anne C Moore
- School of Biochemistry and Cell Biology, School of Pharmacy, University College Cork, Cork, Ireland
| | - Johanna M J Rebel
- Wageningen Livestock Research, Wageningen University & Research, the Netherlands
| |
Collapse
|
5
|
Lee HJ, Kang SJ, Woo Y, Hahn TW, Ko HJ, Jung YJ. TLR7 Stimulation With Imiquimod Induces Selective Autophagy and Controls Mycobacterium tuberculosis Growth in Mouse Macrophages. Front Microbiol 2020; 11:1684. [PMID: 32765474 PMCID: PMC7380068 DOI: 10.3389/fmicb.2020.01684] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2019] [Accepted: 06/29/2020] [Indexed: 12/16/2022] Open
Abstract
Autophagy is a lysosomal self-digestion pathway that maintains internal homeostasis inside cells and critical process by which the innate immune system eliminates intracellular bacteria. In this study, we showed that stimulation of toll-like receptor 7 (TLR7) with imiquimod (IMQ) triggered autophagic cell death in macrophages by enhancing the generation of reactive oxygen species (ROS) via the p38- or MEK/ERK1/2-mediated signaling pathway in the early phase. IMQ significantly increased mitochondrial ROS and targeted autophagosomes to the mitochondria. Stimulation of TLR7 with IMQ enhanced the expression of BNIP3, which was localized to mitochondria and interacted with beclin-1, leading to mitophagy. In addition, IMQ substantially induced NO production through the GSK-3β-mediated signaling pathway, which led to autophagy in the late stage. We further examined whether the induction of autophagy by IMQ effectively eliminated intracellular microbes. Macrophages were infected with a virulent Mycobacterium tuberculosis (Mtb) strain, H37Rv, and then treated with IMQ. IMQ suppressed intracellular Mtb growth by inducing autophagy in a dose-dependent manner and increased NO production. Inhibition of autophagy using 3-methyladenine (3-MA) prevented autophagosome formation and control of intracellular Mtb growth in macrophages. These findings revealed a novel mechanism by which IMQ induces selective autophagy to promote intracellular killing machinery against Mtb infection in macrophages.
Collapse
Affiliation(s)
- Hyo-Ji Lee
- Department of Biological Sciences, Kangwon National University, Chuncheon, South Korea.,Institute of Life Sciences, Kangwon National University, Chuncheon, South Korea
| | - Su-Jin Kang
- Department of Biological Sciences, Kangwon National University, Chuncheon, South Korea
| | - Yunseo Woo
- Department of Biological Sciences, Kangwon National University, Chuncheon, South Korea.,Institute of Life Sciences, Kangwon National University, Chuncheon, South Korea
| | - Tae-Wook Hahn
- College of Veterinary Medicine, Kangwon National University, Chuncheon, South Korea
| | - Hyun-Jeong Ko
- College of Pharmacy, Kangwon National University, Chuncheon, South Korea
| | - Yu-Jin Jung
- Department of Biological Sciences, Kangwon National University, Chuncheon, South Korea.,Institute of Life Sciences, Kangwon National University, Chuncheon, South Korea
| |
Collapse
|
6
|
Duncan SA, Sahu R, Dixit S, Singh SR, Dennis VA. Suppressors of Cytokine Signaling (SOCS)1 and SOCS3 Proteins Are Mediators of Interleukin-10 Modulation of Inflammatory Responses Induced by Chlamydia muridarum and Its Major Outer Membrane Protein (MOMP) in Mouse J774 Macrophages. Mediators Inflamm 2020; 2020:7461742. [PMID: 32684836 PMCID: PMC7333066 DOI: 10.1155/2020/7461742] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2020] [Accepted: 04/10/2020] [Indexed: 12/26/2022] Open
Abstract
The immunopathology of chlamydial diseases is exacerbated by a broad-spectrum of inflammatory mediators, which we reported are inhibited by IL-10 in macrophages. However, the chlamydial protein moiety that induces the inflammatory mediators and the mechanisms by which IL-10 inhibits them are unknown. We hypothesized that Chlamydia major outer membrane protein (MOMP) mediates its disease pathogenesis, and the suppressor of cytokine signaling (SOCS)1 and SOCS3 proteins are mediators of the IL-10 inhibitory actions. Our hypothesis was tested by exposing mouse J774 macrophages to chlamydial stimulants (live Chlamydia muridarum and MOMP) with and without IL-10. MOMP significantly induced several inflammatory mediators (IL-6, IL-12p40, CCL5, CXCL10), which were dose-dependently inhibited by IL-10. Chlamydial stimulants induced the mRNA gene transcripts and protein expression of SOCS1 and SOCS3, with more SOCS3 expression. Notably, IL-10 reciprocally regulated their expression by reducing SOCS1 and increasing SOCS3. Specific inhibitions of MAPK pathways revealed that p38, JNK, and MEK1/2 are required for inducing inflammatory mediators as well as SOCS1 and SOCS3. Chlamydial stimulants triggered an M1 pro-inflammatory phenotype evidently by an enhanced nos2 (M1 marker) expression, which was skewed by IL-10 towards a more M2 anti-inflammatory phenotype by the increased expression of mrc1 and arg1 (M2 markers) and the reduced SOCS1/SOCS3 ratios. Neutralization of endogenously produced IL-10 augmented the secretion of inflammatory mediators, reduced SOCS3 expression, and skewed the chlamydial M1 to an M2 phenotype. Inhibition of proteasome degradation increased TNF but decreased IL-10, CCL5, and CXCL10 secretion by suppressing SOCS1 and SOCS3 expressions and dysregulating their STAT1 and STAT3 transcription factors. Our data show that SOCS1 and SOCS3 are regulators of IL-10 inhibitory actions, and underscore SOCS proteins as therapeutic targets for IL-10 control of inflammation for Chlamydia and other bacterial inflammatory diseases.
Collapse
Affiliation(s)
- Skyla A. Duncan
- Center for NanoBiotechnology Research (CNBR), Department of Biological Sciences, Alabama State University, 1627 Harris Way, Montgomery, AL 36104, USA
| | - Rajnish Sahu
- Center for NanoBiotechnology Research (CNBR), Department of Biological Sciences, Alabama State University, 1627 Harris Way, Montgomery, AL 36104, USA
| | - Saurabh Dixit
- Center for NanoBiotechnology Research (CNBR), Department of Biological Sciences, Alabama State University, 1627 Harris Way, Montgomery, AL 36104, USA
| | - Shree R. Singh
- Center for NanoBiotechnology Research (CNBR), Department of Biological Sciences, Alabama State University, 1627 Harris Way, Montgomery, AL 36104, USA
| | - Vida A. Dennis
- Center for NanoBiotechnology Research (CNBR), Department of Biological Sciences, Alabama State University, 1627 Harris Way, Montgomery, AL 36104, USA
| |
Collapse
|
7
|
Nigar S, Shimosato T. Cooperation of Oligodeoxynucleotides and Synthetic Molecules as Enhanced Immune Modulators. Front Nutr 2019; 6:140. [PMID: 31508424 PMCID: PMC6718720 DOI: 10.3389/fnut.2019.00140] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2019] [Accepted: 08/13/2019] [Indexed: 12/18/2022] Open
Abstract
Unmethylated cytosine–guanine dinucleotide (CpG) motifs are potent stimulators of the host immune response. Cellular recognition of CpG motifs occurs via Toll-like receptor 9 (TLR9), which normally activates immune responses to pathogen-associated molecular patterns (PAMPs) indicative of infection. Oligodeoxynucleotides (ODNs) containing unmethylated CpGs mimic the immunostimulatory activity of viral/microbial DNA. Synthetic ODNs harboring CpG motifs resembling those identified in viral/microbial DNA trigger an identical response, such that these immunomodulatory ODNs have therapeutic potential. CpG DNA has been investigated as an agent for the management of malignancy, asthma, allergy, and contagious diseases, and as an adjuvant in immunotherapy. In this review, we discuss the potential synergy between synthetic ODNs and other synthetic molecules and their immunomodulatory effects. We also summarize the different synthetic molecules that function as immune modulators and outline the phenomenon of TLR-mediated immune responses. We previously reported a novel synthetic ODN that acts synergistically with other synthetic molecules (including CpG ODNs, the synthetic triacylated lipopeptide Pam3CSK4, lipopolysaccharide, and zymosan) that could serve as an immune therapy. Additionally, several clinical trials have evaluated the use of CpG ODNs with other immune factors such as granulocyte-macrophage colony-stimulating factor, cytokines, and both endosomal and cell-surface TLR ligands as adjuvants for the augmentation of vaccine activity. Furthermore, we discuss the structural recognition of ODNs by TLRs and the mechanism of functional modulation of TLRs in the context of the potential application of ODNs as wide-spectrum therapeutic agents.
Collapse
Affiliation(s)
- Shireen Nigar
- Department of Nutrition and Food Technology, Jashore University of Science and Technology, Jashore, Bangladesh
| | - Takeshi Shimosato
- Department of Biomolecular Innovation, Institute for Biomedical Sciences, Shinshu University, Nagano, Japan
| |
Collapse
|
8
|
Lee HJ, Ko HJ, Kim SH, Jung YJ. Pasakbumin A controls the growth of Mycobacterium tuberculosis by enhancing the autophagy and production of antibacterial mediators in mouse macrophages. PLoS One 2019; 14:e0199799. [PMID: 30865638 PMCID: PMC6415846 DOI: 10.1371/journal.pone.0199799] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2018] [Accepted: 02/10/2019] [Indexed: 12/12/2022] Open
Abstract
Tuberculosis (TB) is a chronic infectious disease caused by Mycobacterium tuberculosis (Mtb) and remains a major health problem worldwide. Thus, identification of new and more effective drugs to treat emerging multidrug-resistant TB (MDR-TB) and to reduce the side effects of anti-TB drugs, such as liver toxicity and other detrimental changes, is urgently needed. In this study, to develop a novel candidate drug for effective TB treatment with few side effects in the host, we selected pasakbumin A isolated from Eurycoma longifolia (E. longifolia) Jack, which protected host cells against Mtb infection-induced death. Pasakbumin A significantly inhibited intracellular Mtb growth by inducing the autophagy via the ERK1/2-mediated signaling pathway in Mtb-infected macrophages. We further investigated whether pasakbumin A could be used as a potential adjuvant for TB treatment. Treatment with pasakbumin A and anti-TB drug rifampicin (RMP) potently suppressed intracellular Mtb killing by promoting autophagy as well as TNF-α production via the ERK1/2- and NF-κB-mediated signaling pathways in Mtb-infected cells. Our results suggest that pasakbumin A could be developed as a novel anti-TB drug or host-directed therapeutic (HDT) strategy to protect against host cell death and improve host defense mechanisms against Mtb infection in macrophages.
Collapse
Affiliation(s)
- Hyo-Ji Lee
- Department of Biological Sciences and Institute of Life Sciences, Kangwon National University, Chuncheon, Republic of Korea
| | - Hyun-Jeong Ko
- College of Pharmacy, Kangwon National University, Chuncheon, Republic of Korea
| | - Seung Hyun Kim
- College of Pharmacy, Yonsei University, Incheon, Republic of Korea
| | - Yu-Jin Jung
- Department of Biological Sciences and Institute of Life Sciences, Kangwon National University, Chuncheon, Republic of Korea
- * E-mail:
| |
Collapse
|
9
|
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. [PMID: 30515175 PMCID: PMC6255943 DOI: 10.3389/fimmu.2018.02705] [Citation(s) in RCA: 89] [Impact Index Per Article: 12.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2018] [Accepted: 11/01/2018] [Indexed: 12/17/2022] Open
Abstract
The onset and the termination of innate immune response must be tightly regulated to maintain homeostasis and prevent excessive inflammation, which can be detrimental to the organism, particularly in the context of sepsis. Endotoxin tolerance and compensatory anti-inflammatory response syndrome (CARS) describe a state of hypo-responsiveness characterized by reduced capacity of myeloid cells to respond to inflammatory stimuli, particularly those initiated by bacterial lipopolysaccharide (LPS). To achieve endotoxin tolerance, extensive reprogramming otherwise termed as “innate immune training”, is required that leads to both modifications of the intracellular components of TLR signaling and also to alterations in extracellular soluble mediators. Non-coding RNAs (ncRNAs) have been recognized as critical regulators of TLR signaling. Specifically, several microRNAs (miR-146, miR-125b, miR-98, miR-579, miR-132, let-7e and others) are induced upon TLR activation and reciprocally promote endotoxin tolerance and/or cross tolerance. Many other miRNAs have been also shown to negatively regulate TLR signaling. The long non-coding (lnc)RNAs (Mirt2, THRIL, MALAT1, lincRNA-21 and others) are also altered upon TLR activation and negatively regulate TLR signaling. Furthermore, the promotion or termination of myeloid cell tolerance is not only regulated by intracellular mediators but is also affected by other TLR-independent soluble signals that often achieve their effect via modulation of intracellular ncRNAs. In this article, we review recent evidence on the role of different ncRNAs in the context of innate immune cell tolerance and trained immunity, and evaluate their impact on immune system homeostasis.
Collapse
Affiliation(s)
- Eleni Vergadi
- Department of Paediatrics, Medical School, University of Crete, Heraklion, Greece.,Department of Clinical Chemistry, Medical School, University of Crete, Heraklion, Greece
| | - Katerina Vaporidi
- Department of Intensive Care Medicine, Medical School, University of Crete, Heraklion, Greece
| | - Christos Tsatsanis
- Department of Clinical Chemistry, Medical School, University of Crete, Heraklion, Greece
| |
Collapse
|
10
|
Sun L, Li Y, Luo H, Wang K, Li L, Zeng Y. Effects of Toll‑like receptor 9 and CpG oligodeoxynucleotides 1826 on sodium taurocholate‑induced acute pancreatitis rats. Mol Med Rep 2018; 18:3818-3824. [PMID: 30106134 PMCID: PMC6131499 DOI: 10.3892/mmr.2018.9346] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2016] [Accepted: 05/25/2017] [Indexed: 01/06/2023] Open
Abstract
The aim of the present study was to investigate the effects of Toll-like receptor (TLR) 9 and CpG oligodeoxynucleotide (CpG-ODN)1826 on sodium taurocholate-induced acute pancreatitis (AP) rats at different time points. Pathological examination indicated that the severity of pancreatic tissue damage following AP increased with time. Additionally, TLR9 protein levels were upregulated after AP, and were higher at 6 h compared with at 3 h. Subsequently, the TLR9 protein levels were downregulated at 12 h, but remained higher than the control group. In rats subjected to AP, tumor necrosis factor (TNF-α protein expression levels and serum amylase (AMS) in the serum were increased until 12 h. The expression level of TNF-α protein in the AP 12 h group was higher than that in the AP 3 and 6 h groups. In addition, following CpG-ODN1826 administration, the morphology of pancreatic tissue appeared worse compared with that in the AP only groups. Furthermore, CpG-ODN1826 administration induced an increase in TLR9 expression levels compared with the AP alone group at 0, 3, 6 and 12 h. TNF-α in the CpG + AP 12 h group was upregulated compared with that in the CpG + AP 3 and 6 h groups; however, no change was observed between 3 and 6 h. Thus, these data indicate that CpG-ODN1826 aggravates sodium taurocholate-induced pancreas damage in rats.
Collapse
Affiliation(s)
- Liang Sun
- Department of Gastrointestinal Surgery, The First Affiliated Hospital of Kunming Medical University, Kunming, Yunnan 650031, P.R. China
| | - Yuanhua Li
- Department of Anesthesiology, The First Affiliated Hospital of Kunming Medical University, Kunming, Yunnan 650031, P.R. China
| | - Huayou Luo
- Department of Gastrointestinal Surgery, The First Affiliated Hospital of Kunming Medical University, Kunming, Yunnan 650031, P.R. China
| | - Kunhua Wang
- Department of Gastrointestinal Surgery, The First Affiliated Hospital of Kunming Medical University, Kunming, Yunnan 650031, P.R. China
| | - Li Li
- Department of Hepatobiliary Surgery, The Affiliated Ganmei Hospital of Kunming Medical University, Kunming, Yunnan 650011, P.R. China
| | - Yujian Zeng
- Department of Gastrointestinal Surgery, The First Affiliated Hospital of Kunming Medical University, Kunming, Yunnan 650031, P.R. China
| |
Collapse
|
11
|
Lee HJ, Ko HJ, Song DK, Jung YJ. Lysophosphatidylcholine Promotes Phagosome Maturation and Regulates Inflammatory Mediator Production Through the Protein Kinase A-Phosphatidylinositol 3 Kinase-p38 Mitogen-Activated Protein Kinase Signaling Pathway During Mycobacterium tuberculosis Infection in Mouse Macrophages. Front Immunol 2018; 9:920. [PMID: 29755479 PMCID: PMC5934435 DOI: 10.3389/fimmu.2018.00920] [Citation(s) in RCA: 43] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2018] [Accepted: 04/13/2018] [Indexed: 11/13/2022] Open
Abstract
Tuberculosis is caused by the infectious agent Mycobacterium tuberculosis (Mtb). Mtb has various survival strategies, including blockade of phagosome maturation and inhibition of antigen presentation. Lysophosphatidylcholine (LPC) is a major phospholipid component of oxidized low-density lipoprotein and is involved in various cellular responses, such as activation of second messengers and bactericidal activity in neutrophils. In this study, macrophages were infected with a low infectious dose of Mtb and treated with LPC to investigate the bactericidal activity of LPC against Mtb. In macrophages infected with Mtb strain, H37Ra or H37Rv, LPC suppressed bacterial growth; however, this effect was suppressed in bone marrow-derived macrophages (BMDMs) isolated from G2A (a G protein-coupled receptor involved in some LPC actions) knockout mice. LPC also promoted phagosome maturation via phosphatidylinositol 3 kinase (PI3K)–p38 mitogen-activated protein kinase (MAPK)-mediated reactive oxygen species production and intracellular Ca2+ release during Mtb infection. In addition, LPC induced increased levels of intracellular cyclic adenosine monophosphate (cAMP) and phosphorylated glycogen synthase kinase 3 beta (GSK3β) in Mtb-infected macrophages. Protein kinase A (PKA)-induced phosphorylation of GSK3β suppressed activation of NF-κB in LPC-treated macrophages during Mtb infection, leading to decreased secretion of pro-inflammatory cytokines and increased secretion of anti-inflammatory cytokines. These results suggest that LPC can effectively control Mtb growth by promoting phagosome maturation via cAMP-induced activation of the PKA–PI3K–p38 MAPK pathway. Moreover, LPC can regulate excessive production of pro-inflammatory cytokines associated with bacterial infection of macrophages.
Collapse
Affiliation(s)
- Hyo-Ji Lee
- Department of Biological Sciences, Kangwon National University, Chuncheon, South Korea.,Institute of Life Sciences, Kangwon National University, Chuncheon, South Korea
| | - Hyun-Jeong Ko
- College of Pharmacy, Kangwon National University, Chuncheon, South Korea
| | - Dong-Kun Song
- Department of Pharmacology, College of Medicine, Hallym University, Chuncheon, South Korea
| | - Yu-Jin Jung
- Department of Biological Sciences, Kangwon National University, Chuncheon, South Korea
| |
Collapse
|
12
|
Cho JH, Lee HJ, Ko HJ, Yoon BI, Choe J, Kim KC, Hahn TW, Han JA, Choi SS, Jung YM, Lee KH, Lee YS, Jung YJ. The TLR7 agonist imiquimod induces anti-cancer effects via autophagic cell death and enhances anti-tumoral and systemic immunity during radiotherapy for melanoma. Oncotarget 2018; 8:24932-24948. [PMID: 28212561 PMCID: PMC5421900 DOI: 10.18632/oncotarget.15326] [Citation(s) in RCA: 74] [Impact Index Per Article: 10.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2016] [Accepted: 01/23/2017] [Indexed: 12/31/2022] Open
Abstract
Toll-like receptor (TLR) ligands are strongly considered immune-adjuvants for cancer immunotherapy and have been shown to exert direct anti-cancer effects. This study was performed to evaluate the synergistic anti-cancer and anti-metastatic effects of the TLR7 agonist imiquimod (IMQ) during radiotherapy for melanoma. The pretreatment of B16F10 or B16F1 cells with IMQ combined with γ-ionizing radiation (IR) led to enhanced cell death via autophagy, as demonstrated by increased expression levels of autophagy-related genes, and an increased number of autophagosomes in both cell lines. The results also confirmed that the autophagy process was accelerated via the reactive oxygen species (ROS)-mediated MAPK and NF-κB signaling pathway in the cells pretreated with IMQ combined with IR. Mice subcutaneously injected with melanoma cells showed a reduced tumor growth rate after treatment with IMQ and IR. Treatment with 3-methyladenine (3-MA), ameliorated the anti-cancer effect of IMQ combined with IR. Additionally, the combination therapy enhanced anti-cancer immunity, as demonstrated by an increased number of CD8+ T cells and decreased numbers of regulatory T cells (Treg) and myeloid-derived suppressor cells (MDSCs) in the tumor lesions. Moreover, the combination therapy decreased the number of metastatic nodules in the lungs of mice that were injected with B16F10 cells via the tail vein. In addition, the combination therapy enhanced systemic anti-cancer immunity by increasing the abundances of T cell populations expressing IFN-γ and TNF-α. Therefore, these findings suggest that IMQ could serve as a radiosensitizer and immune booster during radiotherapy for melanoma patients.
Collapse
Affiliation(s)
- Jeong Hyun Cho
- Department of Biological Sciences, Kangwon National University, Chuncheon, Republic of Korea
| | - Hyo-Ji Lee
- Department of Biological Sciences, Kangwon National University, Chuncheon, Republic of Korea
| | - Hyun-Jeong Ko
- College of Pharmacy, Kangwon National University, Chuncheon, Republic of Korea
| | - Byung-Il Yoon
- Department of Veterinary Medicine, Kangwon National University, Chuncheon, Republic of Korea
| | - Jongseon Choe
- Department of Microbiology, School of Medicine, Kangwon National University, Chuncheon, Republic of Korea
| | - Keun-Cheol Kim
- Department of Biological Sciences, Kangwon National University, Chuncheon, Republic of Korea
| | - Tae-Wook Hahn
- Department of Veterinary Medicine, Kangwon National University, Chuncheon, Republic of Korea
| | - Jeong A Han
- Department of Biochemistry and Molecular Biology, School of Medicine, Kangwon National University, Chuncheon, Republic of Korea
| | - Sun Shim Choi
- Department of Medical Biotechnology, College of Biomedical Science, Kangwon National University, Chuncheon, Republic of Korea
| | - Young Mee Jung
- Department of Chemistry, Kangwon National University, Chuncheon, Republic of Korea
| | - Kee-Ho Lee
- Division of Radiation Cancer Research, Korea Institute of Radiological and Medical Sciences, Nowon-gu, Seoul, Republic of Korea
| | - Yun-Sil Lee
- Graduate School of Pharmaceutical Sciences, Ewha Womans University, Seodaemun-gu, Seoul, Republic of Korea
| | - Yu-Jin Jung
- Department of Biological Sciences, Kangwon National University, Chuncheon, Republic of Korea
| |
Collapse
|
13
|
Petes C, Odoardi N, Gee K. The Toll for Trafficking: Toll-Like Receptor 7 Delivery to the Endosome. Front Immunol 2017; 8:1075. [PMID: 28928743 PMCID: PMC5591332 DOI: 10.3389/fimmu.2017.01075] [Citation(s) in RCA: 120] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2017] [Accepted: 08/18/2017] [Indexed: 01/12/2023] Open
Abstract
Toll-like receptor (TLR)-7 is an endosomal innate immune sensor capable of detecting single-stranded ribonucleic acid. TLR7-mediated induction of type I interferon and other inflammatory cytokine production is important in antiviral immune responses. Furthermore, altered TLR7 expression levels are implicated in various autoimmune disorders, indicating a key role for this receptor in modulating inflammation. This review is focused on the regulation of TLR7 expression and localization compared to that of the other endosomal TLRs: TLR3, 8, and 9. Endosomal TLR localization is a tightly controlled and intricate process with some shared components among various TLRs. However, TLR-specific mechanisms must also be in place in order to regulate the induction of pathogen- and cell-specific responses. It is known that TLR7 is shuttled from the endoplasmic reticulum to the endosome via vesicles from the Golgi. Several chaperone proteins are required for this process, most notably uncoordinated 93 homolog B1 (Caenorhabditis elegans), recently identified to also be involved in the localization of the other endosomal TLRs. Acidification of the endosome and proteolytic cleavage of TLR7 are essential for TLR7 signaling in response to ligand binding. Cleavage of TLR7 has been demonstrated to be accomplished by furin peptidases in addition to cathepsins and asparagine endopeptidases. Moreover, triggering receptor expressed on myeloid cells like 4, a protein associated with antigen presentation and apoptosis in immune cells, has been implicated in the amplification of TLR7 signaling. Understanding these and other molecular mechanisms controlling TLR7 expression and trafficking will give insight into the specific control of TLR7 activity compared to the other endosomal TLRs.
Collapse
Affiliation(s)
- Carlene Petes
- Department of Biomedical and Molecular Sciences, Queen's University, Kingston, ON, Canada
| | - Natalya Odoardi
- Department of Biomedical and Molecular Sciences, Queen's University, Kingston, ON, Canada
| | - Katrina Gee
- Department of Biomedical and Molecular Sciences, Queen's University, Kingston, ON, Canada
| |
Collapse
|
14
|
Messer JS. The cellular autophagy/apoptosis checkpoint during inflammation. Cell Mol Life Sci 2017; 74:1281-1296. [PMID: 27837217 PMCID: PMC11107496 DOI: 10.1007/s00018-016-2403-y] [Citation(s) in RCA: 61] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2015] [Revised: 10/17/2016] [Accepted: 10/27/2016] [Indexed: 12/22/2022]
Abstract
Cell death is a major determinant of inflammatory disease severity. Whether cells live or die during inflammation largely depends on the relative success of the pro-survival process of autophagy versus the pro-death process of apoptosis. These processes interact and influence each other during inflammation and there is a checkpoint at which cells irrevocably commit to either one pathway or another. This review will discuss the concept of the autophagy/apoptosis checkpoint and its importance during inflammation, the mechanisms of inflammation leading up to the checkpoint, and how the checkpoint is regulated. Understanding these concepts is important since manipulation of the autophagy/apoptosis checkpoint represents a novel opportunity for treatment of inflammatory diseases caused by too much or too little cell death.
Collapse
Affiliation(s)
- Jeannette S Messer
- Department of Medicine, Knapp Center for Biomedical Discovery, University of Chicago, 900 E. 57th Street, 9th Floor, Chicago, IL, 60637, USA.
| |
Collapse
|
15
|
Lee HJ, Ko HJ, Jung YJ. Insufficient Generation of Mycobactericidal Mediators and Inadequate Level of Phagosomal Maturation Are Related with Susceptibility to Virulent Mycobacterium tuberculosis Infection in Mouse Macrophages. Front Microbiol 2016; 7:541. [PMID: 27148227 PMCID: PMC4834433 DOI: 10.3389/fmicb.2016.00541] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2016] [Accepted: 04/04/2016] [Indexed: 12/13/2022] Open
Abstract
Tuberculosis is caused by Mycobacterium tuberculosis infection, and it remains major life-threatening infectious diseases worldwide. Although, M. tuberculosis has infected one-third of the present human population, only 5–10% of immunocompetent individuals are genetically susceptible to tuberculosis. All inbred strains of mice are susceptible to tuberculosis; however, some mouse strains are much more susceptible than others. In a previous report, we showed that Th1-mediated immunity was not responsible for the differential susceptibility between mouse models. To examine whether these susceptibility differences between inbred mouse strains are due to the insufficient production of effector molecules in the early stage of innate immunity, we investigated mycobacteriostatic function of bone marrow-derived macrophages (BMDMs) in resistant (BALB/c and C57BL/6) and susceptible strains (DBA/2) that were infected with virulent M. tuberculosis (H37Rv) or attenuated M. tuberculosis (H37Ra). The growth rate of virulent M. tuberculosis in infected cells was significantly higher in DBA/2 BMDMs, whereas the growth of the attenuated strain was similar in the three inbred mouse BMDM strains. In addition, the death rate of M. tuberculosis-infected cells increased with the infectious dose when DBA/2 BMDMs were infected with H37Rv. The intracellular reactive oxygen species level was lower in DBA/2 BMDMs that were infected with virulent M. tuberculosis at an early post-infection time point. The expression levels of phagosomal maturation markers, including early endosomal antigen-1 (EEA1) and lysosome-associated membrane protein-1 (LAMP-1), were significantly decreased in DBA/2 BMDM that were infected with virulent M. tuberculosis, whereas IFNγ-treatment restored the phagosomal maturation activity. The nitric oxide (NO) production levels were also significantly lower in DBA/2 BMDMs that were infected with virulent H37Rv at late post-infection points; however, this was not observed with the attenuated H37Ra strain. Furthermore, IFNγ-treatment rescued the low NO production level and insufficient M. tuberculosis growth control of DBA/2 BMDMs to the same level as of both resistant strains. The secreted TNF-α and IL-10 level were not significantly different between strains. Therefore, our findings suggest that DBA/2 BMDMs may have defects in the phagosomal maturation process and in inflammatory mediator production, as they showed innate immune defects when infected with the virulent, but not attenuated M. tuberculosis strain.
Collapse
Affiliation(s)
- Hyo-Ji Lee
- Department of Biological Sciences and BIT Medical Convergence Graduate Program, Kangwon National University Chuncheon, South Korea
| | - Hyun-Jeong Ko
- College of Pharmacy, Kangwon National University Chuncheon, South Korea
| | - Yu-Jin Jung
- Department of Biological Sciences and BIT Medical Convergence Graduate Program, Kangwon National University Chuncheon, South Korea
| |
Collapse
|
16
|
Dehghan-Manshadi M, Hadinedoushan H, Amirbaigy MK, Zare F, Eslami G, Mirghanizade-Bafghi SA, Akhondi-Meybodi M. Relative Expression of Toll-Like Receptors 2 and 7 mRNA in Peripheral Blood of Patients With Hepatitis C. HEPATITIS MONTHLY 2015; 15:e30427. [PMID: 26834788 PMCID: PMC4719121 DOI: 10.5812/hepatmon.30427] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/31/2015] [Revised: 09/13/2015] [Accepted: 09/26/2015] [Indexed: 12/11/2022]
Abstract
BACKGROUND Hepatitis C virus (HCV) is an important human pathogen affecting an estimated 120 - 170 million individuals in the world. Toll-Like receptors (TLRs) are pattern-recognition receptors that recognize pathogen-associated molecular patterns, and stimulate immune responses. OBJECTIVES The aim of this study was to determine the mRNA expression level of TLR2 and TLR7 in HCV-infected patients in comparison with normal controls. PATIENTS AND METHODS Nineteen consecutive patients with HCV infection and nineteen sex and age-matched healthy controls were studied in a case-controlled research. RESULTS Our results showed that the expressions of TLR7 in HCV infected samples were significantly increased in comparison those of the controls (P = 0.02), while the expression of TLR2 was similar between the case and the control group (P = 0.8). There were no associations between the expression levels of TLR2 and TLR7 with HCV viral load and HCV genotypes. Also, there was no association between viral load and genotypes of the virus. CONCLUSIONS Our findings showed that HCV infection could lead to increased expression level of TLR7 mRNA in peripheral blood cells of HCV infected samples. The viral load and genotypes of HCV did not affect the mRNA expression levels of TLR2 and TLR7.
Collapse
Affiliation(s)
- Mahdi Dehghan-Manshadi
- Immunology Department, Faculty of Medicine, Shahid Sadoughi University of Medical Sciences, Yazd, IR Iran
| | - Hossein Hadinedoushan
- Immunology Department, Faculty of Medicine, Shahid Sadoughi University of Medical Sciences, Yazd, IR Iran
- Immunology Department, Reproductive Immunology Research Center, Shahid Sadoughi University of Medical Sciences, Yazd, IR Iran
- Corresponding Author: Hossein Hadinedoushan, Immunology Department, Reproductive Immunology Research Center, Shahid Sadoughi University of Medical Sciences, Yazd, IR Iran. Tel: +98-3536285406; +98-9133576620, Fax: +98-3536238561, E-mail:
| | - Mohammad Kazem Amirbaigy
- Gastrology Department, Faculty of Medicine, Shahid Sadoughi Hospital, Shahid Sadoughi University of Medical Sciences, Yazd, IR Iran
| | - Fateme Zare
- Immunology Department, Faculty of Medicine, Shahid Sadoughi University of Medical Sciences, Yazd, IR Iran
| | - Gilda Eslami
- Parasitology and Mycology Department, Shahid Sadoughi University of Medical Sciences, Yazd, IR Iran
| | | | - Mohsen Akhondi-Meybodi
- Gastrology Department, Faculty of Medicine, Shahid Sadoughi Hospital, Shahid Sadoughi University of Medical Sciences, Yazd, IR Iran
| |
Collapse
|
17
|
MicroRNA-381 Negatively Regulates TLR4 Signaling in A549 Cells in Response to LPS Stimulation. BIOMED RESEARCH INTERNATIONAL 2015; 2015:849475. [PMID: 26688820 PMCID: PMC4672107 DOI: 10.1155/2015/849475] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/03/2015] [Revised: 11/05/2015] [Accepted: 11/08/2015] [Indexed: 01/11/2023]
Abstract
It is widely reported that miR-381 is dysregulated in various tumors. However, the specific role of miR-381 in respiratory infections has not been reported. To probe this role, A549 cells were pretreated with 1 μg/mL LPS for 24 h. The level of miR-381 was detected using RT-qPCR. The expression of proinflammatory cytokines was determined using an ELISA kit and western blotting. Bioinformatics analysis was used to predict the target genes of miR-381, and a luciferase reporter assay was used to validate the expression of the target genes. miR-381 expression was increased in A549 cells treated with LPS, which is a ligand of TLRs. Further study revealed that the overexpression of miR-381 increased the activity of NF-κB signaling, thereby increasing the expression of IL-6, TNFα, and COX-2. Further study revealed that IκBα was a target gene of miR-381. The upregulation of miR-381 under LPS stimulation contributes to respiratory infections mainly by targeting IκBα.
Collapse
|