• 제목/요약/키워드: mouse model of sepsis

검색결과 10건 처리시간 0.031초

패혈증 마우스 모델에서 가와사키병의 관상동맥 병변 관찰: 예비연구와 문헌고찰 (Coronary Arterial Lesions of Kawasaki Disease Observed in a Mouse Model of Sepsis: A Pilot Study and a Review of the Literature)

  • 김주현;김효진;신정하;최의윤;이수영;한지환
    • Pediatric Infection and Vaccine
    • /
    • 제24권2호
    • /
    • pp.102-107
    • /
    • 2017
  • 목적: 가와사키병의 환아뿐만 아니라, 다른 전신 염증 질병의 환아들에 관상동맥 병변(coronary arterial lesion [CAL])의 발생이 보고되었다. 본 연구에서는 전형적인 전신염증반응증후군(systemic inflammatory response syndrome [SIRS])의 소견을 보이는 패혈증 마우스 모델에서 가와사키병 환아에게 관찰되는 CAL이 발생하는지 확인하고자 하였다. 방법: 생후 6주 C57BL/6 마우스에 내독소를 복강내주사하여 SIRS를 보이는 패혈증 모델을 유도하였다. 대조군과 패혈증군의 주요 장기의 조직학적 소견을 비교하였고 패혈증 마우스에서 CAL을 찾기 위한 시도를 하였다. 결과: 대조군과 비교하여, 염증세포의 침윤이 패혈증 마우스의 심장, 간, 신장에 상대적으로 증가하였다. 패혈증 마우스의 심장에서 심근(심근염)과 심장 주위 연조직에 림프구 침윤을 확인하였다. 또한, 패혈증 마우스의 관상동맥을 관찰하였지만, CAL을 확인할 수는 없었다. 결론: 본 연구에서 패혈증 마우스 모델에서 CAL의 존재를 확인하는 것은 실패하였다. 하지만, SIRS를 유발하는 많은 종류의 원인 질병에서 CAL이 발생한다는 것은 잘 알려져 있다. 가와사키병을 포함한 다양한 전신 염증 질병에 나타나는 CAL의 임상적 의미에 대한 연구가 필요할 것이다.

Renal protective effects of zingerone in a mouse model of sepsis

  • Lee, Bong-Seon;Lee, Changhun;Yang, Sumin;Ku, Sae-Kwang;Bae, Jong-Sup
    • BMB Reports
    • /
    • 제52권4호
    • /
    • pp.271-276
    • /
    • 2019
  • Zingerone (ZGR), a phenolic alkanone isolated from ginger, has been reported to possess pharmacological activities such as anti-inflammatory and anti-apoptotic effects. This study was initiated to determine whether ZGR could modulate renal functional damage in a mouse model of sepsis and to elucidate the underlying mechanisms. The potential of ZGR treatment to reduce renal damage induced by cecal ligation and puncture (CLP) surgery in mice was measured by assessment of serum creatinine, blood urea nitrogen (BUN), lipid peroxidation, total glutathione, glutathione peroxidase activity, catalase activity, and superoxide dismutase activity. Treatment with ZGR resulted in elevated plasma levels of BUN and creatinine, and of protein in urine in mice with CLP-induced renal damage. Moreover, ZGR inhibited nuclear $factor-{\kappa}B$ activation and reduced the induction of nitric oxide synthase and excessive production of nitric acid. ZGR treatment also reduced the plasma levels of interleukin-6 and tumor necrosis $factor-{\alpha}$, reduced lethality due to CLP-induced sepsis, increased lipid peroxidation, and markedly enhanced the antioxidant defense system by restoring the levels of superoxide dismutase, glutathione peroxidase, and catalase in kidney tissues. Our study showed renal suppressive effects of zingerone in a mouse model of sepsis, suggesting that ZGR protects mice against sepsis-triggered renal injury.

Anti-septic activity of α-cubebenoate isolated from Schisandra chinensis

  • Kook, Minsoo;Lee, Sung Kyun;Kim, Sang Doo;Lee, Ha Young;Hwang, Jae Sam;Choi, Young Whan;Bae, Yoe-Sik
    • BMB Reports
    • /
    • 제48권6호
    • /
    • pp.336-341
    • /
    • 2015
  • Sepsis is a life-threatening, infectious, systemic inflammatory disease. In this study, we investigated the therapeutic effect of α-cubebenoate, a novel compound isolated from Schisandra chinensis against polymicrobial sepsis in a cecal ligation and puncture (CLP) experimental model. Administration of α-cubebenoate strongly enhanced survival in the CLP model. α-cubebenoate administration also markedly blocked CLP-induced lung inflammation and increased bactericidal activity by enhancing phagocytic activity and hydrogen peroxide generation in mouse bone marrow-derived macrophages and neutrophils. Expression of two important inflammatory cytokines, IL-1 and IL-6, was strongly increased in the CLP model, and this was dramatically blocked by α-cubebenoate. Lymphocyte apoptosis and caspase-3 activation, which are associated with immune paralysis during sepsis, were markedly attenuated by α-cubebenoate. Taken together, our findings indicate that α-cubebenoate, a natural compound isolated from Schisandra chinensis, is a powerful potential anti-septic agent. [BMB Reports 2015; 48(6): 336-341]

Contributory Role of BLT2 in the Production of Proinflammatory Cytokines in Cecal Ligation and Puncture-Induced Sepsis

  • Park, Donghwan;Ro, MyungJa;Lee, A-Jin;Kwak, Dong-Wook;Chung, Yunro;Kim, Jae-Hong
    • Molecules and Cells
    • /
    • 제44권12호
    • /
    • pp.893-899
    • /
    • 2021
  • BLT2 is a low-affinity receptor for leukotriene B4, a potent lipid mediator of inflammation generated from arachidonic acid via the 5-lipoxygenase pathway. The aim of this study was to investigate whether BLT2 plays any role in sepsis, a systemic inflammatory response syndrome caused by infection. A murine model of cecal ligation and puncture (CLP)-induced sepsis was used to evaluate the role of BLT2 in septic inflammation. In the present study, we observed that the levels of ligands for BLT2 (LTB4 [leukotriene B4] and 12(S)-HETE [12(S)-hydroxyeicosatetraenoic acid]) were significantly increased in the peritoneal lavage fluid and serum from mice with CLP-induced sepsis. We also observed that the levels of BLT2 as well as 5-lipoxygenase (5-LO) and 12-LO, which are synthesizing enzymes for LTB4 and 12(S)-HETE, were significantly increased in lung and liver tissues in the CLP mouse model. Blockade of BLT2 markedly suppressed the production of sepsis-associated cytokines (IL-6 [interleukin-6], TNF-α [tumor necrosis factor alpha], and IL-1β [interleukin-β] as well as IL-17 [interleukin-17]) and alleviated lung inflammation in the CLP group. Taken together, our results suggest that BLT2 cascade contributes to lung inflammation in CLP-induced sepsis by mediating the production of inflammatory cytokines. These findings suggest that BLT2 may be a potential therapeutic target for sepsis patients.

패혈증 동물 모델에서 Peroxiredoxin 및 Thioredoxin의 발현 변화 (Altered Expression of Peroxiredoxin and Thioredoxin in Septic Animal Model)

  • 김형중;채호준;안철민;김성규;이원영
    • Tuberculosis and Respiratory Diseases
    • /
    • 제47권4호
    • /
    • pp.451-459
    • /
    • 1999
  • 배 경 : 패혈증에서 과도한 활성산소종의 생성은 급성 폐손상의 병리 기전에 중요한 역할을 한다. Catalase 및 MnSOD 등의 항산화 단백은 패혈증 환자의 혈청내 증가하며 급성호흡곤란증후군의 발생 예측 인자 및 패혈증의 예후 인자로 알려져 있다. Peroxiredoxin(Prx) 는 최근 독특하고 중요한 세포내 항산화 단백으로 알려져 있다. 본 연구는 대식세포인 mouse monocyte-macrophages(RAW 264.7) 세포에 산화 스트레스 및 내독소 처리후 Prx I 및 Prx II의 발현 평가하고 패혈증 동물 모델에서 복강세척액 및 기관지폐포세척액내 Prx I, Prx II 및 Trx의 양을 측정하였다. 방 법 : Prx I, Prx II 및 Trx에 대한 특이 항체를 이용하여 immunoblot 분석으로 호중구, 대식세포 및 적혈구에 이들의 분포를 평가하였다. Mouse monocyte-macrophages 세포에 $5\;{\mu}M$ menadione 및 1 mg/ml lipopolysaccharide(LPS) 을 처치하여 Prx I 및 Prx II의 발현을 평가하였으며 6 mg/Kg LPS를 복강내 투여하여 유발한 복강내 패혈증 동물의 복강세척액내 Prx I, Prx II 및 Trx의 양을 측정하였으며 복강내 패혈증 동물 및 5 mg/Kg LPS를 정맥내 투여하여 유발한 정맥내 패혈증 동물에서 기관지폐포세척액내 Prx I, Prx II 및 Trx을 측정하였으며 폐장의 염증 정도와 비교하였다. 결 과 : Prx I 및 Prx II의 분포는 호중구, 폐포대식세포 및 적혈구에서 서로 다른 양상을 보였다. Mouse monocyte-macrophages 세포에 $5\;{\mu}M$ menadione 및 $1\;{\mu}g/ml$ lipopolysaccharide을 처치하였을 때 Prx I 발현은 증가하였으나 Prx II 발현은 변화하지 않았다. 복강내 패혈증 동물에서 복강세척액내 Prx I, Prx II 및 Trx의 양은 증가하였으나 복강내 패혈증 및 정맥내 패혈증 동물에서 기관지폐포세척액내 폐장 염증 정도와 관계없이 Prx I, Prx II 및 Trx의 양은 증가하지 않았다. 결 론 : 세포내 항산화 단백으로서 mouse monocyte-macrophages 세포에서 Prx I의 발현은 산화 스트레스 및 내독소 처치후 증가한다. Prx I, Prx II 및 Trx양은 패혈증 동물 모델에서 국소 염증 부위에서 증가하나 손상된 폐장에서는 증가하지 않는다.

  • PDF

Aurantio-obtusin exerts an anti-inflammatory effect on acute kidney injury by inhibiting NF-κB pathway

  • Haiyan Xiang;Yun Zhang;Yan Wu;Yaling Xu;Yuanhao Hong
    • The Korean Journal of Physiology and Pharmacology
    • /
    • 제28권1호
    • /
    • pp.11-19
    • /
    • 2024
  • Acute kidney injury (AKI) is one of the major complications of sepsis. Aurantio-obtusin (AO) is an anthraquinone compound with antioxidant and anti-inflammatory activities. This study was developed to concentrate on the role and mechanism of AO in sepsis-induced AKI. Lipopolysaccharide (LPS)-stimulated human renal proximal tubular epithelial cells (HK-2) and BALB/c mice receiving cecal ligation and puncture (CLP) surgery were used to establish in vitro cell model and in vivo mouse model. HK-2 cell viability was measured using MTT assays. Histological alterations of mouse renal tissues were analyzed via hematoxylin and eosin staining. Renal function of mice was assessed by measuring the levels of serum creatinine (SCr) and blood urea nitrogen (BUN). The concentrations of pro-inflammatory cytokines in HK-2 cells and serum samples of mice were detected using corresponding ELISA kits. Protein levels of factors associated with nuclear factor kappa-B (NF-κB) pathway were measured in HK-2 cells and renal tissues by Western blotting. AO exerted no cytotoxic effect on HK-2 cells and AO dose-dependently rescued LPS-induced decrease in HK-2 cell viability. The concentrations of pro-inflammatory cytokines were increased in response to LPS or CLP treatment, and the alterations were reversed by AO treatment. For in vivo experiments, AO markedly ameliorated renal injury and reduced high levels of SCr and BUN in mice underwent CLP operation. In addition, AO administration inhibited the activation of NF-κB signaling pathway in vitro and in vivo. In conclusion, AO alleviates septic AKI by suppressing inflammatory responses through inhibiting the NF-κB pathway.

Interleukin-$32{\gamma}$ Transgenic Mice Resist LPS-Mediated Septic Shock

  • Kim, Sun Jong;Lee, Siyoung;Kwak, Areum;Kim, Eunsom;Jo, Seunghyun;Bae, Suyoung;Lee, Youngmin;Ryoo, Soyoon;Choi, Jida;Kim, Soohyun
    • Journal of Microbiology and Biotechnology
    • /
    • 제24권8호
    • /
    • pp.1133-1142
    • /
    • 2014
  • Interleukin-32 (IL-32) is a cytokine and inducer of various proinflammatory cytokines such as $TNF{\alpha}$, IL-$1{\beta}$, and IL-6 as well as chemokines. There are five splicing variants (${\alpha}$, ${\beta}$, ${\gamma}$, ${\delta}$, and ${\varepsilon}$) and IL-$32{\gamma}$ is the most active isoform. We generated human IL-$32{\gamma}$ transgenic (IL-$32{\gamma}$ TG) mice to express high level of IL-$32{\gamma}$ in various tissues, including immune cells. The pathology of sepsis is based on the systemic inflammatory response that is characterized by upregulating inflammatory cytokines in whole body, particularly in response to gram-negative bacteria. We investigated the role of IL-$32{\gamma}$ in a mouse model of experimental sepsis by using lipopolysaccharides (LPS). We found that IL-$32{\gamma}TG$ mice resisted LPS-induced lethal endotoxemia. IL-$32{\gamma}$ reduced systemic cytokines release after LPS administration but not the local immune response. IL-$32{\gamma}TG$ increased neutrophil influx into the initial foci of the primary injected site, and prolonged local cytokines and chemokines production. These results suggest that neutrophil recruitment in IL-$32{\gamma}TG$ occurred as a result of the local induction of chemokines but not the systemic inflammatory cytokine circulation. Together, our results suggest that IL-$32{\gamma}$ enhances an innate immune response against local infection but inhibits the spread of immune responses, leading to systemic immune disorder.

Effects of C-Terminal Residues of 12-Mer Peptides on Antibacterial Efficacy and Mechanism

  • Son, Kkabi;Kim, Jieun;Jang, Mihee;Chauhan, Anil Kumar;Kim, Yangmee
    • Journal of Microbiology and Biotechnology
    • /
    • 제29권11호
    • /
    • pp.1707-1716
    • /
    • 2019
  • The development of new antimicrobial agents is essential for the effective treatment of diseases such as sepsis. We previously developed a new short peptide, Pap12-6, using the 12 N-terminal residues of papiliocin, which showed potent and effective antimicrobial activity against multidrug-resistant Gram-negative bacteria. Here, we investigated the antimicrobial mechanism of Pap12-6 and a newly designed peptide, Pap12-7, in which the 12th Trp residue of Pap12-6 was replaced with Val to develop a potent peptide with high bacterial selectivity and a different antibacterial mechanism. Both peptides showed high antimicrobial activity against Gram-negative bacteria, including multidrug-resistant Gram-negative bacteria. In addition, the two peptides showed similar anti-inflammatory activity against lipopolysaccharide-stimulated RAW 264.7 cells, but Pap12-7 showed very low toxicities against sheep red blood cells and mammalian cells compared to that showed by Pap12-6. A calcein dye leakage assay, membrane depolarization, and confocal microscopy observations revealed that the two peptides with one single amino acid change have different mechanisms of antibacterial action: Pap12-6 directly targets the bacterial cell membrane, whereas Pap12-7 appears to penetrate the bacterial cell membrane and exert its activities in the cell. The therapeutic efficacy of Pap12-7 was further examined in a mouse model of sepsis, which increased the survival rate of septic mice. For the first time, we showed that both peptides showed anti-septic activity by reducing the infiltration of neutrophils and the production of inflammatory factors. Overall, these results indicate Pap12-7 as a novel non-toxic peptide with potent antibacterial and anti-septic activities via penetrating the cell membrane.

RAW 264.7 대식세포와 염증유도 동물모델에서 산겨릅나무 추출물의 항산화 및 항염증 효과 (In Vitro and In Vivo Anti-Oxidative and Anti-Inflammatory Activities of Acer tegmentosum Maxim Extracts)

  • 이초은;정현희;조진아;이선영
    • 한국식품영양과학회지
    • /
    • 제46권1호
    • /
    • pp.1-9
    • /
    • 2017
  • 산겨릅나무 $80^{\circ}C$ 열수 및 95% 에탄올 추출물을 제조하여 항산화 및 항염증 효과를 확인한 결과 추출물은 우수한 항산화 효과를 나타내었고 RAW 264.7 세포에서도 항염증 효과를 보였으며, 그중 95% 에탄올 추출물의 효능이 더 큰 것으로 나타났다. 따라서 효능이 더 뛰어난 산겨릅나무 에탄올 추출물을 이용하여 in vivo 실험에서 추출물의 세포 보호효과 및 항염증 효능을 재확인하였다. ICR mouse에 일주일간 100 mg/kg B.W 산겨릅나무 에탄올 추출물을 경구투여한 후 LPS를 처리하여 염증반응 및 산화적 스트레스를 유도시켜 Comet assay, serum tumor necrosis $factor(TNF)-{\alpha}$$interferon-{\gamma}$, 장점막세포에서의 $TNF-{\alpha}$와 interleukin-6에 대한 생성능을 측정한 결과 산겨릅나무 에탄올 추출물은 DNA 손상을 억제하였으며 사이토카인 생성을 유의적으로 억제하여 항산화와 항염증 효과를 보였다. 따라서 본 연구에서 사용한 산겨릅나무의 에탄올 추출물은 산화적 스트레스 및 염증을 억제하는 효과를 나타내는 세포보호 기능적 소재로서 가능성이 있는 것으로 생각한다.

Lipopolysaccharide 유도된 Raw264.7 세포주에서 전사조절인자 NF-κB와 IRF-1의 공동작용에 의해 조절되는 PD-1 발현연구 (PD-1 Expression in LPS-Induced Raw264.7 Cells Is Regulated via Co-activation of Transcription Factor NF-κB and IRF-1)

  • 최은경;이수운;이수웅
    • 미생물학회지
    • /
    • 제49권4호
    • /
    • pp.301-308
    • /
    • 2013
  • Programmed Death-1 (PD-1)은 중요한 면역조절분자들 중 하나로 다양한 면역활성인자에 자극된 T 세포, B 세포, NKT 세포 및 대식세포에서 발현된다. Lipopolysaccaride (LPS)는 그람음성세균의 세포벽구성물질로 PD-1 발현을 유도하는 중요 면역원들 중 하나로 알려져 있다. 그러나 선천면역세포에서 PD-1 발현기전에 관한 연구는 미비한 실정이다. 본 연구에서는 LPS에 의해 자극된 Raw264.7 세포주를 대상으로 PD-1 발현 및 발현조전기전을 RT-PCR, Western Blot, 유세포분석기, ChIP assay 및 co-immunoprecipitation 방법으로 조사하였다. Raw264.7 세포주가 LPS로 자극되었을 때 PI3K 및 p38 신호전달경로를 경유하여 PD-1 발현이 크게 증가되었다. 또한 LPS 주사된 생쥐의 비장유래 대식세포에서도 PD-1 발현이 증가됨을 확인 하였다. PD-1 유전자의 프로모터 분석을 통해서 NF-${\kappa}B$ 및 IRF-1 결합부위가 PD-1 발현에 중요함을 알 수 있었다. 또한 PD-1 발현을 극대화하기 위하여 전사조절인자 NF-${\kappa}B$ 및 IRF-1의 공동활성이 필수적임을 확인하였다. 본 연구결과는 LPS 유도 생쥐패혈증모델에서 선천면역세포에 발현된 PD-1분자의 제어를 통한 질병 연구에 유용한 자료로 이용될 수 있을 것으로 사료된다.