335 research outputs found

    RIG-I, MDA5 and TLR3 Synergistically Play an Important Role in Restriction of Dengue Virus Infection

    Get PDF
    Dengue virus (DV) infection is one of the most common mosquito-borne viral diseases in the world. The innate immune system is important for the early detection of virus and for mounting a cascade of defense measures which include the production of type 1 interferon (IFN). Hence, a thorough understanding of the innate immune response during DV infection would be essential for our understanding of the DV pathogenesis. A recent application of the microarray to dengue virus type 1 (DV1) infected lung carcinoma cells revealed the increased expression of both extracellular and cytoplasmic pattern recognition receptors; retinoic acid inducible gene-I (RIG-I), melanoma differentiation associated gene-5 (MDA-5) and Toll-like receptor-3 (TLR3). These intracellular RNA sensors were previously reported to sense DV infection in different cells. In this study, we show that they are collectively involved in initiating an effective IFN production against DV. Cells silenced for these genes were highly susceptible to DV infection. RIG-I and MDA5 knockdown HUH-7 cells and TLR3 knockout macrophages were highly susceptible to DV infection. When cells were silenced for only RIG-I and MDA5 (but not TLR3), substantial production of IFN-ฮฒ was observed upon virus infection and vice versa. High susceptibility to virus infection led to ER-stress induced apoptosis in HUH-7 cells. Collectively, our studies demonstrate that the intracellular RNA virus sensors (RIG-I, MDA5 and TLR3) are activated upon DV infection and are essential for host defense against the virus

    The Mannose Receptor Mediates Dengue Virus Infection of Macrophages

    Get PDF
    Macrophages (Mร˜) and mononuclear phagocytes are major targets of infection by dengue virus (DV), a mosquito-borne flavivirus that can cause haemorrhagic fever in humans. To our knowledge, we show for the first time that the Mร˜ mannose receptor (MR) binds to all four serotypes of DV and specifically to the envelope glycoprotein. Glycan analysis, ELISA, and blot overlay assays demonstrate that MR binds via its carbohydrate recognition domains to mosquito and human cellโ€“produced DV antigen. This binding is abrogated by deglycosylation of the DV envelope glycoprotein. Surface expression of recombinant MR on NIH3T3 cells confers DV binding. Furthermore, DV infection of primary human Mร˜ can be blocked by anti-MR antibodies. MR is a prototypic marker of alternatively activated Mร˜, and pre-treatment of human monocytes or Mร˜ with type 2 cytokines (IL-4 or IL-13) enhances their susceptibility to productive DV infection. Our findings indicate a new functional role for the MR in DV infection

    Effect of pretreatment with chromium picolinate on haematological parameters during dengue virus infection in mice

    Get PDF
    Background & objectives: Dengue virus (DV) has caused severe epidemics of dengue fever (DF) and dengue haemorrhagic fever (DHF) and is endemic all over India. We have earlier reported that exposure of mice to hexavalent chromium [Cr(VI)] compounds increased the severity of dengue virus infection. Trivalent chromium picolinate (CrP) is used worldwide as micronutrient and nutritional supplement. The present study was therefore, carried out to investigate the effects of CrP on various haematological parameters during DV infection of mice. Methods: The Swiss Albino smice were inoculated with dengue virus (1000 LD50, intracerebrally) and fed with chromium picolinate (CrP) in drinking water (100 and 250 mg/l) for 24 wk. Peripheral blood leucocytes and other haematological parameters, and spleens were studied on days 4 and 8 after virus inoculations and the findings were compared with those given only CrP and the normal control age matched mice. Results: CrP in drinking water for 24 wk had no significant effects on peripheral blood cells of mice. On the other hand, there was significant decrease in different haematological parameters following inoculation of normal mice with DV. In CrP fed mice the effects of DV infection were abolished on most of the haematological parameters. Interpretation & conclusions: The findings of present study showed that the adverse effects of DV infection, specially on platelets and leucocytes, were abrogated by pretreatment of mice with CrP. The therapeutic utility of CrP in viral infections including dengue needs to be studied in depth

    Distinct Behaviors of Infected and Bystander Dendritic Cells Following Exposure to Dengue Virus: A Dissertation

    Get PDF
    Dengue viruses (DV) are re-emerging mosquito-borne pathogens for which four distinct lineages, grouped based on serology and referred to as serotypes 1-4 (DIV-D4V), have been described. Epidemiological data imply that re-infection with a heterologous serotype, i.e, one other than that to which the individual was originally exposed, enhances the risk for development of severe disease, dengue hemorrhagic fever (DHF). The hallmark of DHF is a transient capillary leakage syndrome of rapid onset, temporally associated with the resolution of fever and viremia. In its most grave form, the vascular permeability phenomenon in DHF may progress to dengue shock syndrome (DSS), which is often fatal in the absence of appropriate medical care. Despite the fulminant nature of vascular leakage during DHF/DSS, this phenomenon does not appear to be due to direct cytopathic effects of DV. Rather, inappropriate reactivation and/or regulation of dengue-specific memory are the prevailing theorized (immunopathological) etiologies. Traditional vaccine development techniques have proven insufficient for DV, since any vaccine must offer complete protection against all four serotypes to avoid enhanced pathology on natural viral challenge. Understanding the underlying mechanisms that contribute to dengue disease, particularly the development of dengue-specific memory, is therefore of critical importance. Dengue immunopathology and the specific aspects of immunological memory that determine disease severity are heatedly debated. Previous research in our lab has suggested that T cell responses contribute to the severity of dengue illness. Clinical data indicate enhanced immune activation in more grave cases of DV infection, and serotype cross-reactive T cells from multiple individuals are present after both primary and secondary dengue infections. However, little is known about the conditions under which T cells are primed and dengue-specific memory is generated. Dendritic cells (DCs) are bone marrow-derived cells that play a central role in directing activity within the immune system. DCs shape quantitative and qualitative aspects of adaptive immunity, and therefore the intrinsic characteristics of host memory to a pathogen. DCs are essential in generating primary immune responses, due to their particular effectiveness in stimulating naรฏve T cells. DCs also play important roles in the reactivation of memory to an infectious agent, and as reservoirs for the dissemination of invading microorganisms. Exposure to pathogens or their products initiates a series of phenotypic and functional changes in DCs, termed maturation. DC maturation involves a coordinated response of immunomodulatory surface molecule elaboration and cytokine production, culminating in antigen presentation to, and co-stimulation of, T cells specific for the invading agent. The DC response is ostensibly tailored to facilitate effective elimination by regulating effective downstream interactions of the DC with T cells. A number of viruses have evolved to infect DCs and alter their functional behavior, facilitating their own survival within the host, and the herd. DV readily infects DCs both in primary cell cultures and in vivo. However, reports on the effects of DV infection on DC maturation vary both with regard to some of the cytokines produced, and the phenotypes of infected versus bystander cells. Although DCs appear to be activated following DV exposure, responses on the single-cell level appear to depend on the infection state of the cell, hypothetically driven by intracellular virus-mediated effects. Therefore, downstream responses to these divergent populations - i.e., actively infected cells versus uninfected bystander cells - are likely to be the consequence of at least two modes of DC behavior. Because DCs play a pivotal role in adaptive immune development, and because the resulting memory response appears to be critical in affecting disease pathology after heterologous DV re-infection, I sought to explore the phenomena of DC maturation in response to dengue exposure, and to begin to answer the question of how active infection alters the functional capabilities of DCs. Notably, primary dengue infection is generally well-controlled with minimal pathology. Therefore, this thesis addresses the hypothesis that DV infection of DCs results in cellular activation and stimulation of antiviral immunity, despite virus-mediated alteration of DC maturation. In order to address this hypothesis, I examined both DV infection-dependent and independent effects on DC functional responses including surface molecule regulation secretory activity, and CD4 T cell allostimulatory priming. DCs derived from human peripheral blood monocytes were readily infected with multiple strains of DV. DV infection of DCs derived from separate donors was dose-dependent, with substantial variability in DC susceptibility to infection. Exposure to live DV activated surface molecule expression in DCs, similar to the effects of defined maturation stimuli including a combination of TNF-ฮฑ and IFN-ฮฑ, or LPS. In addition, UV-inactivated DV induced expression of cell surface molecules, albeit to a lesser extent than did live virus demonstrating inherent stimulatory properties of DV particles. Using intracellular staining for DV envelope (E) protein, I detected increased surface molecule expression on both infected DCs and uninfected bystander DCs from the same culture, as compared to mock-infected DCs. These data indicate that activation was not prevented in cells undergoing active viral replication. However, the degree of surface molecule induction depended on the infection state of the cell. Infected DCs had enhanced PD-L2 and MHC II expression relative to uninfected bystander cells, while PD-L1, CD80, CD86, and MHC I expression were suppressed with active infection. Therefore, intracellular DV replication altered the process of cell surface molecule regulation within these cells. DV infection of DCs also resulted in the secretion of a broad array of cytokines and chernokines. These included the antiviral cytokine IFN-ฮฑ, inflammatory cytokines TNF-ฮฑ, IL-6, and IL-1ฮฑ, and inflammatory chemokines IP10, MCP-1, MIP-1ฮฑ, and RANTES. DV infection did not induce DC production of the IL-12 p70 heterodimer, and secretion of the immunosuppressive cytokine IL-10 was low in most experiments. Similar to the results seen with surface molecule induction, UV inactivation of DV reduced, but did not eliminate, cytokine and chemokine responses. At the single-cell level, TNF-ฮฑ and IP10 production profiles of infected DCs and uninfected bystander DCs were distinct. DV infection in DCs reduced production of IP10, but stimulated TNF-ฮฑ as compared to uninfected bystander cells in the same culture. Blocking experiments demonstrated that IFN-ฮฑ/ฮฒ produced by DCs in response to infection actively inhibited viral protein expression and drove IP10, but not TNF-ฮฑ, production. DV infection of DCs did not consistently suppress DC stimulation of allogeneic CD4 T cell proliferation. In cases where infection enhanced DC stimulatory function, T cell proliferation was less pronounced than that induced by DCs activated with exogenous TNF-ฮฑ plus IFN-ฮฑ. Increasing multiplicity of infection (MOI) of DCs with DV resulted in increasing DC infection rates, but a statistically significant trend at the highest MOIs for decreased T cell alloproliferation, suggesting that direct infection of DCs reduces their CD4 T cell priming function. MOI-dependent reduction in DC stimulatory function depended on replication-competent virus. Increased MOIs during DV infection of DCs did not cause an elevation in detectable IL-10 in supernatants derived from T-DC co-cultures. In addition, increased DV MOI of DCs was not associated with increased levels of either IL-13 or IFN-ฮณ in supernatants from T-DC co-culture, suggesting that actively infected DC do not skew CD4 T cells towards a specific Th phenotype. These data demonstrate that DV infection induces functional maturation of DCs that is modified by the presence of virus through both IFN-dependent and independent mechanisms. However, the allostimulatory phenotype of DCs was not universally enhanced, nor was it skewed towards antiviral (Th1)-type responses. These data suggest a model whereby dengue infection during primary illness results in controlled immune stimulation through activation of bystander DCs, and the generation of mixed Th-type responses. Direct DV infection of DCs appears to attenuate activation of, and potentially clearance by, antiviral mechanisms. During secondary infection, reduced IP10 production and enhanced TNF-ฮฑ secretion by infected cells coupled with MHC I downregulation and enhanced PD-L2 expression, would subvert both Th1 CD4 T cell recruitment and result in CD8 T cell suppression and death. Furthermore, DV-specific effects on DCs would allow for continued viral replication in the absence of effective clearance. These DV-mediated effects would modify T cell memory responses to infected DC, and potentially facilitate the expansion of pathologic T cell subsets. Contributing to this pathological cascade, antibody-dependent enhancement of infection in monocytic cells and macrophages would shift antigen presentation and cytokine production paradigms, increasing the risk of DHF

    Vascular endothelium: the battlefield of dengue viruses

    Get PDF
    Increased vascular permeability without morphological damage to the capillary endothelium is the cardinal feature of dengue haemorrhagic fever (DHF)/dengue shock syndrome (DSS). Extensive plasma leakage in various tissue spaces and serous cavities of the body, including the pleural, pericardial and peritoneal cavities in patients with DHF, may result in profound shock. Among various mechanisms that have been considered include immune complex disease, T-cell-mediated, antibodies cross-reacting with vascular endothelium, enhancing antibodies, complement and its products, various soluble mediators including cytokines, selection of virulent strains and virus virulence, but the most favoured are enhancing antibodies and memory T cells in a secondary infection resulting in cytokine tsunami. Whatever the mechanism, it ultimately targets vascular endothelium (making it a battlefield) leading to severe dengue disease. Extensive recent work has been done in vitro on endothelial cell monolayer models to understand the pathophysiology of vascular endothelium during dengue virus (DV) infection that may be translated to help understand the pathogenesis of DHF/DSS. The present review provides a broad overview of the effects of DV infection and the associated host responses contributing towards alterations in vascular endothelial cell physiology and damage that may be responsible for the DHF/DSS

    Dengue Vaccines: Strongly Sought but Not a Reality Just Yet

    Get PDF
    abstract: Dengue virus (DV) infections cause undisputedly the most important arthropod-borne viral disease in terms of worldwide prevalence, human suffering, and cost. Worldwide DV infection prevalence in 2010 was between 284 to 528 million cases. Approximately 84% of these cases come from Asia and the Americas, where the cost for emerging economies can be as high as 580 million dollars per year. Thus, the need for an efficient vaccine against DV is extreme.The article is published at http://journals.plos.org/plospathogens/article?id=10.1371/journal.ppat.100355

    Lethal Antibody Enhancement of Dengue Disease in Mice Is Prevented by Fc Modification

    Get PDF
    Immunity to one of the four dengue virus (DV) serotypes can increase disease severity in humans upon subsequent infection with another DV serotype. Serotype cross-reactive antibodies facilitate DV infection of myeloid cells in vitro by promoting virus entry via Fcฮณ receptors (FcฮณR), a process known as antibody-dependent enhancement (ADE). However, despite decades of investigation, no in vivo model for antibody enhancement of dengue disease severity has been described. Analogous to human infants who receive anti-DV antibodies by transplacental transfer and develop severe dengue disease during primary infection, we show here that passive administration of anti-DV antibodies is sufficient to enhance DV infection and disease in mice using both mouse-adapted and clinical DV isolates. Antibody-enhanced lethal disease featured many of the hallmarks of severe dengue disease in humans, including thrombocytopenia, vascular leakage, elevated serum cytokine levels, and increased systemic viral burden in serum and tissue phagocytes. Passive transfer of a high dose of serotype-specific antibodies eliminated viremia, but lower doses of these antibodies or cross-reactive polyclonal or monoclonal antibodies all enhanced disease in vivo even when antibody levels were neutralizing in vitro. In contrast, a genetically engineered antibody variant (E60-N297Q) that cannot bind FcฮณR exhibited prophylactic and therapeutic efficacy against ADE-induced lethal challenge. These observations provide insight into the pathogenesis of antibody-enhanced dengue disease and identify a novel strategy for the design of therapeutic antibodies against dengue

    Invariant NKT Cell Response to Dengue Virus Infection in Human

    Get PDF
    BACKGROUND:Dengue viral infection is a global health threat without vaccine or specific treatment. The clinical outcome varies from asymptomatic, mild dengue fever (DF) to severe dengue hemorrhagic fever (DHF). While adaptive immune responses were found to be detrimental in the dengue pathogenesis, the roles of earlier innate events remain largely uninvestigated. Invariant natural killer T (iNKT) cells represent innate-like T cells that could dictate subsequent adaptive response but their role in human dengue virus infection is not known. We hypothesized that iNKT cells play a role in human dengue infection. METHODS:Blood samples from a well-characterized cohort of children with DF, DHF, in comparison to non-dengue febrile illness (OFI) and healthy controls at various time points were studied. iNKT cells activation were analyzed by the expression of CD69 by flow cytometry. Their cytokine production was then analyzed after ฮฑ-GalCer stimulation. Further, the CD1d expression on monocytes, and CD69 expression on conventional T cells were measured. RESULTS:iNKT cells were activated during acute dengue infection. The level of iNKT cell activation associates with the disease severity. Furthermore, these iNKT cells had altered functional response to subsequent ex vivo stimulation with ฮฑ-GalCer. Moreover, during acute dengue infection, monocytic CD1d expression was also upregulated and conventional T cells also became activated. CONCLUSION:iNKT cells might play an early and critical role in the pathogenesis of severe dengue viral infection in human. Targeting iNKT cells and CD1d serve as a potential therapeutic strategy for severe dengue infection in the future

    DC-SIGN (CD209) mediates dengue virus infection of human dendritic cells

    Get PDF
    Dengue virus is a single-stranded, enveloped RNA virus that productively infects human dendritic cells (DCs) primarily at the immature stage of their differentiation. We now find that all four serotypes of dengue use DC-SIGN (CD209), a C-type lectin, to infect dendritic cells. THP-1 cells become susceptible to dengue infection after transfection of DC-specific ICAM-3 grabbing nonintegrin (DC-SIGN), or its homologue L-SIGN, whereas the infection of dendritic cells is blocked by anti-DC-SIGN antibodies and not by antibodies to other molecules on these cells. Viruses produced by dendritic cells are infectious for DC-SIGN- and L-SIGN-bearing THP-1 cells and other permissive cell lines. Therefore, DC-SIGN may be considered as a new target for designing therapies that block dengue infection

    ๋งˆํฌ๋กœํŒŒ์ง€ ๋งค๊ฐœ ์‚ฌ์ดํ† ์นด์ธ ์กฐ์ ˆ์„ ํ†ตํ•œ ๋ฐ”์ด๋Ÿฌ์Šค ๋ฉด์—ญ ์น˜๋ฃŒ์ œ ๊ฐœ๋ฐœ ์—ฐ๊ตฌ

    Get PDF
    ํ•™์œ„๋…ผ๋ฌธ(๋ฐ•์‚ฌ) -- ์„œ์šธ๋Œ€ํ•™๊ต๋Œ€ํ•™์› : ์˜๊ณผ๋Œ€ํ•™ ์˜ํ•™๊ณผ, 2023. 2. ์„์Šนํ˜.Macrophages are an essential component of innate cellular immunity with flexible functions prominently involved in host defense and immunity against foreign microorganisms, including bacteria, viruses, and fungi. Many viruses target macrophages, and activated macrophages lead to phagocytosis and the release of pro-inflammatory cytokines and chemokines. However, excessive secretion of pro-inflammatory cytokines by macrophages contributes to local tissue damage and a dangerous systemic inflammatory response. Since monocytes and macrophages are the central cells that secrete pro-inflammatory cytokines, the efficient control of these cells can be used as a therapeutic target to regulate cytokines. In this study, I aimed to regulate influenza virus-mediated hyper-inflammation by targeting macrophages and then examined the immune mechanism regulating interleukin (IL)-12 in macrophages during dengue virus infection. First, in this study, I developed liposomes that are selectively delivered to macrophages. Also, I found that liposomal dexamethasone (DEX/lipo) significantly reduced the protein level of tumor necrosis factor-alpha (TNF-ฮฑ), IL-1ฮฒ, IL-6, and the C-X-C motif chemokine ligand 2 (CXCL2) as well as the number of infiltrated immune cells in the bronchoalveolar lavage fluids as compared to the control and free dexamethasone (DEX) in influenza virus-infected mice. Moreover, the intranasal delivery of DEX/lipo during disease progression reduced the death rate by 20%. Therefore, the intranasal delivery of DEX/lipo may serve as a novel promising therapeutic strategy for treating influenza virus-induced pneumonia (Chapter 1). Next, I revealed the cause of decreased IL-12 after severe dengue virus infection in macrophages through dengue virus binding receptor-mediated signaling. As a result, in type I interferon receptor knockout (IFNAR KO) mice infected with severe dengue virus, down-regulated IL-12 decreased vessel tight junction through increasing matrix metalloproteinase-9 (MMP-9). In addition, I found that mice treated with recombinant IL-12 rapidly regained body weight and attenuated dengue hemorrhagic fever. Based on a reliable model, I have developed a potential therapeutic strategy to attenuate dengue hemorrhagic fever (Chapter 2). Together, my results provide valuable insights into the development of immunotherapies for viral disease via regulating cytokines by targeting macrophages.๋งˆํฌ๋กœํŒŒ์ง€๋Š” ์ˆ™์ฃผ์˜ ๋ฐฉ์–ด์™€ ์„ธ๊ท , ๋ฐ”์ด๋Ÿฌ์Šค, ์ง„๊ท ์„ ํฌํ•จํ•œ ์™ธ๋ถ€ ๋ฏธ์ƒ๋ฌผ์— ๋Œ€ํ•œ ๋ฉด์—ญ์— ํ˜„์ €ํ•˜๊ฒŒ ๊ด€์—ฌํ•˜๋Š” ์—ฌ๋Ÿฌ ๊ธฐ๋Šฅ์„ ๊ฐ€์ง„ ํƒ€๊ณ ๋‚œ ์„ธํฌ ๋ฉด์—ญ์˜ ์ค‘์š”ํ•œ ๊ตฌ์„ฑ ์š”์†Œ์ด๋‹ค. ๋งŽ์€ ๋ฐ”์ด๋Ÿฌ์Šค๊ฐ€ ๋งˆํฌ๋กœํŒŒ์ง€๋ฅผ ํ‘œ์ ์œผ๋กœ ํ•˜๊ณ , ํ™œ์„ฑํ™”๋œ ๋งˆํฌ๋กœํŒŒ์ง€๋Š” ์‹์„ธํฌ์ž‘์šฉ์„ ์ผ์œผํ‚ค๋ฉฐ ์—ผ์ฆ์„ฑ ์‚ฌ์ดํ† ์นด์ธ๊ณผ ์ผ€๋ชจ์นด์ธ์„ ๋ถ„๋น„ํ•œ๋‹ค. ๊ทธ๋Ÿฌ๋‚˜ ๋งˆํฌ๋กœํŒŒ์ง€์— ์˜ํ•œ ์—ผ์ฆ ์œ ๋ฐœ ์‚ฌ์ดํ† ์นด์ธ์˜ ๊ณผ๋„ํ•œ ๋ถ„๋น„๋Š” ๊ตญ์†Œ ์กฐ์ง ์†์ƒ๊ณผ ์œ„ํ—˜ํ•œ ์ „์‹  ์—ผ์ฆ ๋ฐ˜์‘์— ๊ธฐ์—ฌํ•œ๋‹ค. ๋‹จํ•ต๊ตฌ์™€ ๋งˆํฌ๋กœํŒŒ์ง€๋Š” ์—ผ์ฆ์„ฑ ์‚ฌ์ดํ† ์นด์ธ์„ ๋ถ„๋น„ํ•˜๋Š” ์ฃผ์š” ์„ธํฌ์ด๊ธฐ ๋•Œ๋ฌธ์— ์ด๋Ÿฌํ•œ ์„ธํฌ์˜ ํšจ์œจ์ ์ธ ์ œ์–ด๋Š” ์‚ฌ์ดํ† ์นด์ธ์„ ์กฐ์ ˆํ•˜๋Š” ์น˜๋ฃŒ ํ‘œ์ ์œผ๋กœ ์‚ฌ์šฉ๋  ์ˆ˜ ์žˆ๋‹ค. ๋ณธ ์—ฐ๊ตฌ์—์„œ๋Š” ๋งˆํฌ๋กœํŒŒ์ง€๋ฅผ ํ‘œ์ ์œผ๋กœ ํ•˜์—ฌ ์ธํ”Œ๋ฃจ์—”์ž ๋ฐ”์ด๋Ÿฌ์Šค ๋งค๊ฐœ ์—ผ์ฆ ๋ฐ˜์‘์„ ์กฐ์ ˆํ•˜๊ณ , ์ดํ›„ ๋Ž…๊ธฐ ๋ฐ”์ด๋Ÿฌ์Šค ๊ฐ์—ผ ์‹œ ๋งˆํฌ๋กœํŒŒ์ง€์—์„œ ํŠน์ด์ ์œผ๋กœ IL-12๋ฅผ ์กฐ์ ˆํ•˜๋Š” ๋ฉด์—ญ ๊ธฐ์ „์„ ์กฐ์‚ฌํ•˜๊ณ ์ž ํ•˜์˜€๋‹ค. ์ฒซ๋ฒˆ์งธ๋กœ, ๋ณธ ์—ฐ๊ตฌ๋Š” ๋งˆํฌ๋กœํŒŒ์ง€์— ์„ ํƒ์ ์œผ๋กœ ์ „๋‹ฌ์ด ๋˜๋Š” ๋ฆฌํฌ์†œ์„ ๊ฐœ๋ฐœํ•˜์˜€๋‹ค. ์ดํ›„ ์ธํ”Œ๋ฃจ์—”์ž ๋ฐ”์ด๋Ÿฌ์Šค ๊ฐ์—ผ ๋งˆ์šฐ์Šค ๋ชจ๋ธ์—์„œ ๋ฑ์‚ฌ๋ฉ”ํƒ€์† ๋ฆฌํฌ์†œ์„ ์ „๋‹ฌ ํ–ˆ์„ ๋•Œ TNF-ฮฑ, IL-1ฮฒ, IL-6, CXCL2์™€ ๊ฐ™์€ ์—ผ์ฆ์„ฑ ์‚ฌ์ดํ† ์นด์ธ, ์ผ€๋ชจ์นด์ธ์ด ๊ฐ์†Œํ•˜๊ณ , ์นจ์œค๋œ ์—ผ์ฆ์„ฑ ์„ธํฌ๋ฅผ ๊ฐ์†Œ๋œ๋‹ค๋Š” ๊ฒƒ์„ ๋ฐํ˜”๋‹ค. ์ด๋กœ ์ธํ•ด ์งˆ๋ณ‘์ด ์ง„ํ–‰๋˜๋Š” ๋™์•ˆ ๋ฑ์‚ฌ๋ฉ”ํƒ€์† ๋ฆฌํฌ์†œ ์น˜๋ฃŒ๋Š” ์‚ฌ๋ง๋ฅ ์„ 20%๊นŒ์ง€ ๊ฐ์†Œ์‹œ์ผฐ๋‹ค. ๊ฒฐ๊ณผ์ ์œผ๋กœ ๋ฑ์‚ฌ๋ฉ”ํƒ€์† ๋ฆฌํฌ์†œ์˜ ๋น„๊ฐ• ๋‚ด ์ „๋‹ฌ์€ ์ธํ”Œ๋ฃจ์—”์ž ๋ฐ”์ด๋Ÿฌ์Šค ๋งค๊ฐœ ํ๋ ด์˜ ์น˜๋ฃŒ๋ฅผ ์œ„ํ•œ ์œ ๋งํ•œ ์น˜๋ฃŒ ์ „๋žต์œผ๋กœ ์ž‘์šฉํ•  ์ˆ˜ ์žˆ์Œ์„ ์‹œ์‚ฌํ•œ๋‹ค (Chapter 1). ๋‹ค์Œ์œผ๋กœ ๋งˆํฌ๋กœํŒŒ์ง€์— ์‹ฌ๊ฐํ•œ ๋Ž…๊ธฐ ๋ฐ”์ด๋Ÿฌ์Šค ๊ฐ์—ผ ํ›„ IL-12๊ฐ€ ๊ฐ์†Œํ•˜๋Š” ์›์ธ์„ ๋Ž…๊ธฐ ๋ฐ”์ด๋Ÿฌ์Šค ๊ฒฐํ•ฉ ์ˆ˜์šฉ์ฒด ๋งค๊ฐœ ์‹ ํ˜ธ์ „๋‹ฌ์„ ํ†ตํ•ด ๋ฐํ˜€๋ƒˆ๋‹ค. ๊ทธ ๊ฒฐ๊ณผ, IFNAR KO ๋งˆ์šฐ์Šค๊ฐ€ ์ค‘์ฆ ๋Ž…๊ธฐ ๋ฐ”์ด๋Ÿฌ์Šค์— ๊ฐ์—ผ๋œ ๊ฒฝ์šฐ, ๊ฐ์†Œ๋œ IL-12๊ฐ€ MMP-9๋ฅผ ์ฆ๊ฐ€์‹œ์ผœ ํ˜ˆ๊ด€ ํˆฌ๊ณผ์„ฑ์„ ์ฆ๊ฐ€์‹œํ‚จ๋‹ค๋Š” ๊ฒƒ์„ ๋ฐํ˜”๋‹ค. ๋˜ํ•œ, ์žฌ์กฐํ•ฉ IL-12๋กœ ์น˜๋ฃŒํ•œ ๋งˆ์šฐ์Šค๊ฐ€ ์ฒด์ค‘์„ ๋น ๋ฅด๊ฒŒ ํšŒ๋ณตํ•˜๊ณ  ๋Ž…๊ธฐ์ถœํ˜ˆ์—ด์„ ์™„ํ™”์‹œํ‚ค๋Š” ๊ฒƒ์„ ๋ฐํ˜”๋‹ค. ๋ณธ ์—ฐ๊ตฌ์—์„œ๋Š” ๋Ž…๊ธฐ์ถœํ˜ˆ์—ด์„ ์–ต์ œํ•  ์ˆ˜ ์žˆ๋Š” ์ž ์žฌ์ ์ธ ์น˜๋ฃŒ ์ „๋žต์„ ์ œ์‹œํ•˜์˜€๋‹ค (Chapter 2). ๊ฒฐ๋ก ์ ์œผ๋กœ ๋ณธ ์—ฐ๊ตฌ๋Š” ๋งˆํฌ๋กœํŒŒ์ง€๋ฅผ ํ‘œ์ ์œผ๋กœ ํ•˜์—ฌ ์‚ฌ์ดํ† ์นด์ธ์„ ์กฐ์ ˆํ•จ์œผ๋กœ์จ ๋ฐ”์ด๋Ÿฌ์Šค์— ๋Œ€ํ•œ ๋ฉด์—ญ ์š”๋ฒ•์˜ ์น˜๋ฃŒ์  ์ ‘๊ทผ์— ์ค‘์š”ํ•œ ๊ทผ๊ฑฐ ์ž๋ฃŒ๋กœ ์‚ฌ์šฉํ•  ์ˆ˜ ์žˆ์„ ๊ฒƒ์œผ๋กœ ๊ธฐ๋Œ€๋œ๋‹ค.List of figures 1 List of abbreviations 3 Chapter 1 5 Introduction 6 Materials and Methods 9 Results 15 Discussions 27 Chapter 2 32 Introduction 33 Materials and Methods 36 Results 47 Discussions 73 References 79 Abstract in Korean 86๋ฐ•
    • โ€ฆ
    corecore