内质网应激在高迁移率族蛋白B1诱导树突状细胞成熟分化中的作用
详细信息    本馆镜像全文|  推荐本文 |  |   获取CNKI官网全文
摘要
目的:内质网(ER)是真核细胞中非常重要的细胞器,非常敏感,各种应激刺激均可导致内质网功能失调,即内质网应激(ERS)。ERS持续时间及应激水平对于应激细胞的功能与生存均具有重要影响,与多种人类疾病关系密切。我们前期实验结果表明,高迁移率族蛋白B1(HMGB1)对树突状细胞(DC)成熟分化具有重要免疫调节作用。本实验拟对ERS是否参与HMGB1对DC免疫调节机制进行探讨,旨在从新的角度认识ERS在HMGB1介导DC免疫功能障碍中的地位与调节机制,为探索严重烧伤后脓毒症的免疫调控途径提供新思路。
     方法:采用雄性BALB/c小鼠分离培养脾脏DC,以含10%胎牛血清RPMI-1640调整细胞浓度1-2×106/ml,5%CO237℃培养过夜后随机分组进行如下实验:1.给予HMGB1(0.1-10μg/ml)或衣霉素(TM,ERS特异性诱导剂,0.1.10μg/ml)刺激24-72 h后,应用real time-PCR技术及Western blot分析检测ERS相关的关键分子(GRP78、SEPS1及XBP-1)表达及活化水平的改变。2.给予TM(0.1-10μg/ml)刺激24-72 h后,流式细胞术检测DC表面标志物及共刺激分子的表达情况;应用ELISA和real time-PCR技术检测TNF-α、IL-12的分泌水平。另外,采用MTT试剂盒检测其对T淋巴细胞增殖活性的影响。3.通过RNA干扰技术诱导DC中ERS关键调节分子XBP-1基因沉默后,给予HMGB1刺激(1μg/ml,48h),采用Western blot分析、流式细胞术、ELISA、real time-PCR等技术方法检测抑制ERS反应对HMGB1调节DC成熟分化效应的影响。4.给予TM刺激(1μg/ml,48h)正常DC,或HMGB1(1μg/ml,48h)刺激XBP-1基因沉默型DC后,流式细胞术检测DC表面晚期糖基化终末产物受体(RAGE)表达水平。
     结果:1.ERS在HMGB1诱导DC成熟分化过程中具有重要作用。HMGB1刺激可明显诱导DC中ERS反应的发生,与正常对照组相比,0.1μg/ml、1μg/ml、10μg/ml的HMGB1作用24~72h后,DC中ERS相关分子GRP78基因及蛋白水平均明显升高,XBP-1表达及活化水平亦有显著上调,其中尤以1μg/ml HMGB1刺激24 h作用效果显著(P<0.05)。但与ERS特异性诱导剂TM作用不同的是,除0.1μg/ml HMGB1作用48 h诱导SEPS1的蛋白表达水平升高外(P<0.05),本实验所观察的其他刺激浓度和作用时间未能引起SEPS1表达的明显上调。2.TM诱导药物性ERS反应对DC功能状态有一定的影响。0.1μg/ml、1μg/ml、10μg/ml的TM作用24~72 h可导致TNF-α、IL-12的分泌增加(P<0.05),但DC表面共刺激分子表达改变不尽一致,CD80表达在0.1μg/ml TM刺激48h、MHC-Ⅱ表达在0.1μg/ml和1μg/ml TM刺激24-48 h可见明显增强,而CD86在本实验观察的TM各刺激浓度和时间点无显著升高。与HMGB1预孵育DC增强T淋巴细胞增殖反应不同,TM预孵育的DC对T细胞增殖活性具有明显抑制效应(P<0.05)。3.小鼠脾脏DC经XBP-1基因沉默后给予HMGB1刺激(1μg/ml,48 h),与非基因沉默细胞(未转染或转染无效序列)相比,其表面标志物及共刺激分子表达上调均明显受抑,对T淋巴细胞增殖反应及Thl功能极化的促进作用亦明显降低(P<0.05)。4.与非基因沉默细胞(未转染或转染无效序列)相比,XBP-1基因沉默阻抑HMGB1诱导的DC表面RAGE受体表达的上调(P<0.05)。TM诱导的药物性ERS反应对DC表面受体RAGE表达具有明显的上调作用(P<0.05)。
     结论:HMGB1刺激可诱导小鼠脾脏DC发生明显的ERS反应,应用RNA干扰技术抑制ERS反应中关键分子XBP-1的活化可阻抑HMGB1介导的DC成熟分化和表面受体RAGE的表达上调。TM诱导的ERS反应与HMGB1所致ERS反应呈现一定的差异,TM刺激可增加DC细胞因子TNF-α及IL-12的分泌,但对其表型的成熟无明显的诱导效应,TM预孵育DC对T淋巴细胞的增殖反应呈抑制作用。ERS在HMGB1诱导的DC成熟分化中具有重要意义。
Background:Endoplasmic reticulum (ER), the most important organelle in eukaryotic cells, is exceedingly sensitive to alterations in homeostasis. Physiological states that increase the demand for protein folding, or stimuli that disrupt proteins folding reactions, create an imbalance between the protein-folding load and the capacity of the ER, causing unfolded or misfolded proteins to accumulate in the ER lumen, a condition referred to as ER stress (ERS). Dendritic cells (DCs) are pivotal regulators of immune reactivity and immune tolerance. The status of DC maturation and function undoubtedly has important influence on the development of septic complications. High mobility group box-1 protein (HMGB1), was recently described as an important late inflammatory cytokine. Our previous study had demonstrated the contribution of HMGB1 to the maturation and differentiation of DCs. Because the ERS response has the function to regulate the balance between homeostasis and apoptosis, we ventured to speculate that the ERS response might contribute to DC maturation and activation. The present study was performed to clarify the involvement of ERS during maturation of mouse splenic DCs induced by HMGB1.
     Methods:DCs isolated from the spleens of male BALB/c mice were suspended in RPMI-1640 with 10% FCS at 1-2×106 cells/ml on cell culture plates. Cells were then cultured at 37℃in 5% CO2 in humidified air overnight for recovery before subjected to treatments in the following listed experiments.1. DCs were stimulated with HMGB1 or tunicamycin (TM, pharmacological ERS inducer) at increasing dosages (0.1,1,10μg/ml) for different durations (24,48 or 72 hours). The expression levels of two key ER chaperones, glucose regulated protein (GRP) 78 and selenoprotein S (SEPS1), as well as the activation of key molecule of ERS response, X-box binding protein 1 (XBP-1) were determined by real-time RT-PCR and Western blot analysis.2. DCs were stimulated with TM in different concentrations (0.1,1, or 10μg/ml) for different duration (24,48, or 72 hours). Phenotype and cytokine secretion of DCs were analyzed by flow cytometry, real-time PCR and ELISA, respectively. Modified mixed lymphocyte reaction assay was performed to assess the effect of TM-treated DC on the proliferation rate of T cells.3. DCs were transfected with RNAi lentiviral vector to induce gene silence of XBP-1. Using this cell model, we further investigated the involvement of ERS in immune regulation of HMGB1 on DCs maturation and function.4. XBP-1 silenced DCs were treated with 1μg/ml HMGB1 for 48 hours. The influence of ERS on the expression of the receptor for advanced glycation end products (RAGE) on surface of DCs was detected by flow cytometry.
     Results:1. Treatment with HMGB1 induced significant up-regulation of ERS-related molecules in mouse splenic DCs. Protein and mRNA levels of GRP 78 were increased after HMGB1 stimulation in a time-and dose-dependent manner. The expression and activity of XBP-1 were also significantly elevated. 2. Pharmacological ERS induced by TM resulted in elevated cytokine release, but failed to induce up-regualation of costimulatory molecules including CD80 and CD86 on the surface of DCs. T cell proliferation in response to Con A was inhibited after co-culture with TM-treated DCs.3. Gene silence of XBP-1 in mouse splenic DCs decreased the levels of CD80, CD86 as well as MHC-II expression and cytokine secretion after HMGB1 treatment, when compared with untransfected or NTi-transfected DC (P<0.05). Further more, XBP-1 silenced DC showed immunosuppressive or tolerogenic DC and failed to stimulate notable proliferation response of T cell, even after treatment with HMGB1.4. Gene silence of XBP-1 resulted in down-regulation of RAGE expression on the surface of mouse splenic DCs after HMGB1 stimulation. The conventionally used pharmacological ERS inducer TM could induce up-regulation of RAGE on the surface of DC in a dose-and time-dependent manner.
     Conclusion:HMGB1 stimulation induced significant ERS response in mouse splenic DCs. Gene silence of XBP-1 resulted in suppression in activation of DCs and also the up-regulation of RAGE induced by HMGB1. Pharmacological ERS induced by TM demonstrated difference with that induced by HMGB1 in extent of reaction and features and did not effectively arouse DC maturation and activation. Our findings support the notion that ERS response plays an important role in regulation of DC maturation as well as activation induced by HMGB1.
     INTRODUCTION
     Sepsis is denoted as a complex clinical syndrome that results from a serious infection followed by an amplified and dysregulated inflammatory response. Despite continuing progress in the development of antibiotics and other supportive care therapies, sepsis remains a leading cause of the high morbidity and mortality in the intensive care unit. Severe complications like multiple organ dysfunctions syndrome (MODS) with high mortality and the lack of knowledge concerning the underlying pathogenetic mechanisms continue to hamper the development of effective therapeutic strategy for septic complications.
     High mobility group box-1 protein (HMGB1), an evolutionarily ancient non-histone DNA-binding protein within the nucleus, was recently described as a late inflammatory cytokine.Unlike cytokines, such as tumor necrosis factor (TNF)-αand interleukin (IL)-1β, which are released early in the development of a systemic inflammatory response, HMGB1 was released with a delay of 9-16 hours, sustaining for several days after endotoxin exposure by activated macrophages, NK cells, and dendritic cells (DCs) . It was found that patients and animals with sepsis or endotoxemia presented high levels of systemic HMGB1. Numerous evidences indicate that HMGB1 is a necessary late mediator of severe sepsis, and therefore, the delayed kinetic action of HMGB1 provides a wider time window for clinical intervention of sepsis .
     Extracellular HMGB1 has been shown to be able to provoke inflammation, to regulate the migration of monocytes , and also to contribute to the activation of DCs , which is the pivotal regulator of immune reactivity and immune tolerance. Since its discovery more than 30 years ago, DCs have emerged as the most important potent antigen-presenting cells (APCs), which induce and coordinate host immune response. Interacted with agents including foreign pathogens, such as lipopolysaccharide (LPS), or endogenous signals of cellular injury or damage, DCs are stimulated to mature and play roles in engendering the differentiation of different clones of T helper (Th) cells . The maturation of DC has been implicated as the bridge plank between the innate and adaptive immune systems , and it plays a significant role in the pathogenesis of infection, thus providing a protective mechanism in prevention of infection . Our previous studies had demonstrated HMGB1 to be a potential immunostimulatory signal that induced maturation and differentiation of DCs via the receptor for advanced glycation end products (RAGE), and regulated T cell-mediated immunity . However, the endogenous changes and mechanisms involved in the control of maturation and differentiation of DCs after HMGB1 stimuli are poorly known.
     The status of DC maturation and function undoubtedly has important influence on the prognosis of sepsis. Recent study revealed that endoplasmic reticulum (ER) stress response led to hepatocyte apoptosis and might mediate the long-term adaptive and deleterious hepatic dysfunction after severe thermal injury in mice, thus providing intensive insights into endogenous sources of cellular stress as the focus of investigation in immune regulatory mechanisms.
     ER, a membranous network of branching tubules and flattened sacs that extends throughout the cytoplasm of the cell, being contiguous with the nuclear membrane, is one of the most important organelles in eukaryotic cells. The ER is mainly recognized as a protein-folding factory, responsible for the biosynthesis, folding, assembly and modification of numerous soluble proteins and membrane proteins. The ER also functions as a dynamic calcium store, which responds to growth factors, hormones, and stimuli that perturb cellular energy levels, nutrient availability or redox status. Given the importance of this organelle, it is extremely sensitive to alterations in homeostasis. Physiological states that increase the demand for protein folding, or stimuli that disrupt the reactions by which proteins fold, create an imbalance between the protein-folding load and the capacity of the ER, causing unfolded or misfolded proteins to accumulate in the ER lumen, a condition referred to as ER stress (ERS). In response to ERS, mammalian cells trigger unfolded protein response (UPR) signaling pathways to cope with stressful conditions and to monitor the protein folding capacity of the ER and transmit that information to mechanisms that can modulate the ER environment, regulate various aspects of cellular metabolism, and even influence cell fate.
     Because the ERS response functions to regulate the balance between homeostasis and apoptosis, it was our attempt to determine whether the ERS response and UPR pathway, as induced by HMGB1 might contribute to DC maturation and activation. In the present study, we assessed the expression of two ER resident chaperones, namely glucose-regulated protein (GRP) 78 (BIP) and SEPS1 (selenoprotein S, VIMP, Tanis or SelS), as well as the activity of X-box binding protein 1 (XBP-1), which is the key regulatory molecule of UPR. We demonstrated that treatment of mouse splenic DCs with HMGB1 induced ERS response. The expression of GRP 78 and SEPS1, the two markers of ERS, and the activity of XBP-1 were significantly up-regulated. Surprisingly, pharmacological ERS induced by the use of tunicamycin (TM), an inhibitor of N-linked glycosylation and pharmacological agent that disrupted protein folding and assembly in the ER, failed to activate DC completely, and resulted in tolerance of DC. Furthermore, we found that the reduction of XBP-1 translation by RNA interference (RNAi) in mouse splenic DCs with small hairpin RNA (shRNA) led to a decrease in HMGB1-mediated regulatory effects on DCs.
引文
1. Pinsky MR. Dysregulation of the immune response in severe sepsis. Am J Med Sci,2004,328:220-229.
    2. Ulloa L, Tracey KJ. The "cytokine profile":a code for sepsis. Trends Mol Med,2005,11:56-63.
    3. Ulloa L. The vagus nerve and the nicotinic anti-inflammatory pathway. Nat Rev Drug Discov,2005,4:673-684.
    4. Lotze MT, Tracey KJ. High-mobility group box 1 protein (HMGB1):nuclear weapon in the immune arsenal. Nat Rev Immunol,2005,5:331-342.
    5. Ulloa L, Batliwalla FM, Andersson U, Gregersen PK, Tracey KJ. High mobility group box chromosomal protein 1 as a nuclear protein, cytokine, and potential therapeutic target in arthritis. Arthritis Rheum,2003,48:876-881.
    6. Wang H, Bloom O, Zhang M, Vishnubhakat JM, Ombrellino M, Che J, Frazier A, Yang H, Ivanova S, Borovikova L, Manogue KR, Faist E, Abraham E, Andersson J, Andersson U, Molina PE, Abumrad NN, Sama A, Tracey KJ. HMG-1 as a late mediator of endotoxin lethality in mice. Science,1999, 285:248-251.
    7. Wang H, Yang H, Czura CJ, Sama AE, Tracey KJ. HMGB1 as a late mediator of lethal systemic inflammation. Am J Respir Crit Care Med,2001,164: 1768-1773.
    8. Chen G, Ward MF, Sama AE, Wang H. Extracellular HMGB1 as a proinflammatory cytokine. J Interf Cytok Res,2004,24:329-333.
    9. Yang H, Ochani M, Li J, Qiang X, Tanovic M, Harris HE, Susarla SM, Ulloa L, Wang H, DiRaimo R, Czura CJ, Wang H, Roth J, Warren HS, Fink MP, Fenton MJ, Andersson U, Tracey KJ. Reversing established sepsis with antagonists of endogenous high-mobility group box 1. Proc Natl Acad Sci USA,2004,101:296-301.
    10. Qin S, Wang H, Yuan R, Li H, Ochani M, Ochani K, Rosas-Ballina M, Czura CJ, Huston JM, Miller E, Lin X, Sherry B, Kumar A, Larosa G, Newman W, Tracey KJ, Yang H. Role of HMGB1 in apoptosis-mediated sepsis lethality. J Exp Med,2006,203:1637-1642.
    11. Rouhiainen A, Kuja-Panula J, Wilkman E, Pakkanen J, Stenfors J, Tuominen RK, Lepantalo M, Carpen O, Parkkinen J, Rauvala H. Regulation of monocyte migration by amphoterin(HMGB1). Blood,2004,104:1174-1182.
    12. Dumitriu IE, Baruah P, Bianchi ME, Manfredi AA, Rovere-Querini P. Requirement of HMGB1 and RAGE for the maturation of human plasmacytoid dendritic cells. Eur J Immunol,2005,35:2184-2190.
    13. Messmer D, Yang H, Telusma G, Knoll F, Li J, Messmer B, Tracey KJ, Chiorazzi N. High mobility group box protein 1:an endogenous signal for dendritic cell maturation and Thl polarization. J Immunol,2004,173: 307-313.
    14. Dumitriu IE, Baruah P, Valentinis B, Voll RE, Herrmann M, Nawroth PP, Arnold B, Bianchi ME, Manfredi AA, Rovere-Querini P. Release of high mobility group box 1 by dendritic cells controls T cell activation via the receptor for advanced glycation end products. J Immunol,2005,174: 7506-7515.
    15. Yang D, Chen Q, Yang H, Tracey KJ, Bustin M, Oppenheim JJ. High mobility group box-1 protein induces the migration and activation of human dendritic cells and acts as an alarmin. J Leukoc Biol,2007,81:59-66.
    16. Lipscomb MF, Masten BJ. Dendritic cells:immune regulators in health and disease. Physiol Rev,2002,82:97-130.
    17. Zitvogel L. Dendritic and natural killer cells cooperate in the control/switch of innate immunity. J Exp Med,2002,195:F9-F14.
    18. Banchereau J, Steinman RM. Dendritic cells and the control of immunity. Nature,1998,392:245-252.
    19. Steinman RM, Banchereau J. Taking dendritic cells into medicine. Nature, 2007,449:419-426.
    20. Zhang LT, Yao YM, Dong N, Dong YQ, Yu Y, Sheng ZY Relationship between high-mobility group box 1 protein release and T-cell suppression in rats after thermal injury. Shock,2008,30:449-455.
    21. Zhu XM, Yao YM, Liang HP, Liu F, Dong N, Yu Y, Sheng ZY. Effect of high mobility group box-1 protein on apoptosis of peritoneal macrophages. Arch Biochem Biophys,2009,492:54-61.
    22. Song J, Finnerty CC, Herndon DN, Boehning D, Jeschke MG. Severe burn-induced endoplasmic reticulum stress and hepatic damage in mice. Mol Med,2009,15:316-320.
    23. Aridor M, Balch WE. Integration of endoplasmic reticulum signaling in health and disease. Nat Med,1999,5:745-751.
    24. Xue X, Piao JH, Nakajima A, Sakon-Komazawa S, Kojima Y, Mori K, Yagita H, Okumura K, Harding H, Nakano H. Tumor necrosis factor alpha (TNF alpha) induces the unfolded protein response (UPR) in a reactive oxygen species (ROS)-dependent fashion, and the UPR counteracts ROS accumulation by TNFalpha. J Biol Chem,2005,280:33917-33925.
    25. Wang XZ, Harding HP, Zhang Y, Jolicoeur EM, Kuroda M, Ron D. Cloning of mammalian Irel reveals diversity in the ER stress responses. EMBO J, 1998,17:5708-5717.
    26. Haze K, Yoshida H, Yanagi H, Yura T, Mori K. Mammalian transcription factor ATF6 is synthesized as a transmembrane protein and activated by proteolysis in response to endoplasmic reticulum stress. Mol Biol Cell,1999, 10:3787-3799.
    27. Curran JE, Jowett JB, Elliott KS, Gao Y, Gluschenko K, Wang J, Abel Azim DM, Cai G, Mahaney MC, Comuzzie AG, Dyer TD, Walder KR, Zimmet P, MacCluer JW, Collier GR, Kissebah AH, Blangero J. Genetic variation in selenoprotein S influences inflammatory response. Nat Genet,2005,37: 1234-1241.
    28. Zhang KZ, Kaufman RJ. From endoplasmic-reticulum stress to the inflammatory response. Nature,2008,454:455-462.
    29. Gass JN, Gifford NM, Brewer JW. Activation of an unfolded protein response during differentiation of antibody-secreting b cells. J Biol Chem,2002,277: 49047-49054
    30. Iwakoshi NN, Pypaert M, Glimcher LH. The transcription factor XBP-1 is essential for the development and survival of dendritic cells. J Exp Med,2007, 204:2267-2275.
    31. Julius MH, Simpson E, Herzenberg LA. A rapid method for the isolation of functional thymus-derived murine lymphocytes. Eur J Immunol,1973,3: 645-649.
    32. Li M, Qian H, Ichim TE, Ge WW, Popov IA, Rycerz K, Neu J, White D, Zhong R, Min WP. Induction of RNA interference in dendritic cells. Immunol
    Res,2004,30:215-230.
    33. Reimold AM, Ponath PD, Li YS, Hardy RR, David CS, Strominger JL, Glimcher LH. Transcription factor B cell lineagespecific activator protein regulates the gene for human X-box binding protein 1. J Exp Med,1996, 183:393-401.
    34. Kamimura D, Bevan MJ. Endoplasmic reticulum stress regulator XBP-1 contributes to effector CD8+T cell differentiation during acute infection. J Immunol,2008,181:5433-5441.
    35. Wheeler MC, Rizzi M, Sasik R, Almanza G, Hardiman G, Zanetti M. KDEL-retained antigen in B lymphocytes induces a proinflammatory response:a possible role for endoplasmic reticulum stress in adaptive T cell immunity. J Immunol,2008,181:256-264.
    36. Kokkola R, Andersson A, Mullins G, Ostberg T, Treutiger CJ, Arnold B, Nawroth P, Andersson U, Harris RA, Harris HE. RAGE is the major receptor for the proinflammatory activity of HMGB1 in rodent macrophages. Scand J Immunol,2005,61:1-9.
    37. Lechler R, Ng WF, Steinman RM. Dendritic cells in transplantation:friend or foe? Immunity,2001,14:357-368.
    38. Steinman RM. Dendritic cells:understanding immunogenicity. Eur J Immunol,2007,37:S53-S60.
    39. Steinman RM. Lasker basic medical research award, dendritic cells:versatile controllers of the immune system. Nat Med,2007,13:1155-1159.
    40. Pichlmair A, Reis e Sousa C. Innate recognition of viruses. Immunity,2007, 27:370-383.
    41. Steinman RM, Hawiger D, Nussenzweig MC. Tolerogenic dendritic cells. Annu Rev Immunol,2003,21:685-711.
    42. Pierre P. Immunity and the regulation of protein synthesis:surprising connections. Curr Opin Immunol,2009,21:70-77.
    43. Salter RD, Watkins SC. Dendritic cell altered states:what role for calcium? Immunol Rev,2009,231:278-88.
    44. Lee AS. The glucose-regulated proteins:stress induction and clinical applications. Trends Biochem Sci,2001,26:504-510.
    45. Bertolotti A, Zhang Y, Hendershot LM, Harding HP, Ron D. Dynamic interaction of BIP and ER stress transducers in the unfolded-protein response. Nat Cell Biol,2000,2:326-332.
    46. Gao Y, Hannan NR, Wanyonyi S, Konstantopolous N, Pagnon J, Feng HC, Jowett JB, Kim KH, Walder K, Collier GR. Activation of the selenoprotein SEPS1 gene expression by pro-inflammatory cytokines in HepG2 cells. Cytokine,2006,33:246-251.
    47. Kim KH, Gao Y, Walder K, Collier GR, Skelton J, Kissebah AH. SEPS1 protects RAW264.7 cells from pharmacological ER stress agent-induced apoptosis. Biochem Biophy Res Commun,2007,354:127-132.
    48. Chambers I, Frampton J, Goldfarb P, Affara N, McBain W, Harrison PR. The structure of the mouse glutathione peroxidase gene:the selenocysteine in the active site is encoded by the'termination'codon, TGA. EMBO J,1986, 5:1221-1227.
    49. Castellano S, Lobanov AV, Chapple C, Novoselov SV, Albrecht M, Hua D, Lescure A, Lengauer T, Krol A, Gladyshev VN, Guigo R. Diversity and functional plasticity of eukaryotic selenoproteins:identification and characterization of the SelJ family. Proc Natl Acad Sci USA,2005,102: 16188-16193.
    50. Papp LV, Lu J, Holmgren A, Khanna KK. From selenium to selenoproteins: synthesis, identity, and their role in human health. Antioxid Redox Signal, 2007,9:775-806.
    51. Kelly E, Greene CM, Carroll TP, McElvaney NG, O'Neill SJ. Selenoprotein S/SEPS1 modifies ER stress in Z variant alpha-1 antitrypsin deficiency. J Biol Chem, paper in press. Jbc.M 109.006288.
    52. Harding HP, Zhang Y, Bertolotti A, Zeng H, Ron D. Perk is essential for translational regulation and cell survival during the unfolded protein response. Mol Cell,2000,5:897-904.
    53. Harding HP, Zhang Y, Ron D. Protein translation and folding are coupled by an endoplasmic-reticulum-resident kinase. Nature,1999,397:271-274.
    54. Glimcher LH. XBP1:the last two decades. Ann Rheum Dis,2010,69:i67-i71.
    55. Liou HC, Boothby MR, Glimcher LH. Distinct cloned class Ⅱ MHC DNA-binding proteins recognize the X box transcription element. Science, 1988,242:69-71.
    56. Liou HC, Polla BS, Aragnol D, Leserman LD, Griffith IJ, Glimcher LH. A tissue-specific DNase Ⅰ-hypersensitive site in a class II A alpha gene is under trans-regulatory control. Proc Natl Acad Sci USA,1988,85:2738-2742.
    57. Shen X, Ellis RE, Lee K, Liu CY, Yang K, Solomon A, Yoshida H, Morimoto R, Kurnit DM, Mori K, Kaufman RJ. Complementary signaling pathways regulate the unfolded protein response and are required for C. elegans development. Cell,2001,107:893-903.
    58. Yoshida H, Matsui T, Yamamoto A, Okada T, Mori K. XBP1 mRNA is induced by ATF6 and spliced by IRE1 in response to ER stress to produce a highly active transcription factor. Cell,2001,107:881-891.
    59. Calfon M, Zeng H, Urano FI, Till JH, Hubbard SR, Harding HP, Clark SG, Ron D. IRE1 couples endoplasmic reticulum load to secretory capacity by processing the XBP-1 mRNA. Nature,2002,415:92-96.
    60. Todd DJ, Lee AH, Glimcher LH. The endoplasmic reticulum stress response in immunity and autoimmunity. Nat Rev Immunol,2008,8:663-674.
    1. Aridor M, Balch WE. Integration of endoplasmic reticulum signaling in health and disease [J]. Nat Med,1999,5(7):745-751.
    2. Kaufman RJ. Stress signling from the lumen of the endoplasmic reticolum: coordination of gene transcriptional and translational controls [J]. Gene Dev, 1999,13(10):1211-1233.
    3. Lee AH, Iwakoshi NN, Glimcher LH. XBP-1 regulates a subset of endoplasmic reticulum resident chaperone genes in the unfolded protein response. Mol Cell Biol,2003,23(21):7448-7459.
    4. Nakagawa T, Zhu H, Morishima N, et al. Caspase-12 mediates endoplasmic-reticulum-specific apoptosis and cytotoxicity by amyloid-beta [J]. Nature, 2000,403(6765):98-103.
    5. NakagawaT,Yuan JY. Cross-talk between two cysteine protease families. activation of caspase-12 by calpain in apoptosis[J]. J Cell Biol,2000,150(4): 887-894.
    6. Rao RV, Peel A, Logvinova A, et al.Coupling endoplasmic reticulum stress to the cell death program:role of the ER chaperone GRP78 [J]. FEBS Lett,2002, 514(2-3):122-128.
    7. Morishima N, Nakanishi K, Takenouchi H, et al. An endoplasmic reticulum stress_specific caspase cascade in apoptosis. Cytochrome c-independent activation of caspase-9 by caspase-12 [J]. J Biol Chem,2002,277(37):34287-34294
    8. Oyadomari S, Mori M. Roles of CHOP/GADD153 in endoplasmic reticulum stress [J]. Cell Death Differ,2004,11(4):381-389.
    9. Gotoh T, Oyadomari S, Mori K, et al. Nitric oxide-induced apoptosis in RAW 264.7 macrophages is mediated by endoplasmic reticulum stress pathway involving ATF6 and CHOP [J]. J Biol Chem,2002,277(14):12343-12350.
    10. Tajiri S, Oyadomari S, Yano S, et al. Ischemia-induced neuronal cell death is mediated by the endoplasmic reticulum stress pathway involving CHOP [J]. Cell Death Differ,2004,11(4):403-415.
    11. McCullough KD, Martindale JL, Klotz LO, et al. Gadd153 sensitizes cells to endoplasmic reticulum stress by downregulating Bcl2 and perturbing the cellular redox state [J]. Mol Cell Biol,2001,21(4):1249-1259.
    12. Marciniak SJ, Yun CY, Oyadomari S, et al. CHOP induces death by promoting protein synthesis and oxidation in the stressed endoplasmic reticulum [J]. Genes Dev,2004,18(24):3066-3077.
    13. Gotoh T, Terada K, Oyadomari S, et al. hsp70-DnaJ chaperone pair prevents nitric oxide-and CHOP-induced apoptosis by inhibiting translocation of Bax to mitochondria [J]. Cell Death Differ,2004,11(4):390-402.
    14. Urano F, Wang X, Bertolotti A, et al. Coupling of stress in the ER to activation of JNK protein kinases by transmembrane protein kinase IRE1 [J]. Science, 2000,287(5453):664-666.
    15.Nishitoh H, Matsuzawa A, Tobiume K, et al. ASK1 is essential for endoplasmic reticulum stress-induced neuronal cell death triggered by expanded polyglutamine repeats [J]. Genes Dev,2002,16(11):1345-1355.
    16. Suyama K, Ohmuraya M, Hirota M, et al. C/EBP homologous protein is crucial for the acceleration of experimental pancreatitis. Biochem Biophys Res Commun,2008,367(1):176-182.
    17. Dixit G, Mikoryak C, Hayslett T, et al. Cholera toxin up-regulates endoplasmic reticulum proteins that correlate with sensitivity to the toxin. Exp Biol Med (Maywood),2008,233(2):163-175.
    18. Majors AK, Austin RC, de la Motte CA, et al. Endoplasmic Reticulum Stress Induces Hyaluronan Deposition and Leukocyte Adhesion [J]. J Biol Chem, 278(47):47223-47231
    19.祝筱梅,刘秀华,蔡莉蓉,等.p38丝裂素活化蛋白激酶介导缺氧预处理诱导的内质网应激相关的心肌细胞保护.生理学报,2006,58(5):463-470.
    20.祝筱梅,刘秀华,蔡莉蓉.缺氧后处理对缺氧/复氧心肌细胞保护作用及其机理研究.中国微循环,2007,11(4):223-230.
    21. Dear JW, Leelahavanichkul A, Aponte A, et al. Liver proteomics for therapeutic drug discovery:inhibition of the cyclophilin receptor CD 147 attenuates sepsis-induced acute renal failure. Crit Care Med,2007,35(10): 2319-2328.
    22. Saleh M, Vaillancourt JP, Graham RK, et al. Differential modulation of endotoxin responsiveness by human caspase-12 polymorphisms. Nature,2004, 429(6987):75-79.
    23. Saleh M, Mathison JC, Wolinski MK., et al. Enhanced bacterial clearance and sepsis resistance in caspase-12-deficient mice. Nature,2006,440(7087): 1064-1068.
    24. Reimold AM, Iwakoshi NN, Manis J, et al. Plasma cell differentiation requires the transcription factor XBP-1 [J]. Nature,2001,412 (6844):300-307.
    25. Wiest DL, Burkhardt JK, Hester S, et al. Membrane biogenesis during B cell differentiation:most endoplasmic reticulum proteins are expressed coordinately [J]. J Cell Biol,1990,110(5):1501-1511.
    26. Lewis MJ, Mazzarella RA, Green M. Structure and assembly of the endoplasmic reticulum. The synthesis of three major endoplasmic reticulum proteins during lipopolysaccharide-induced differentiation of murine lymphocytes [J]. J Biol Chem,1985,260(5):3050-3057.
    27. Iwakoshi NN, Lee AH, Glimcher LH. The X-box binding protein-1 transcription factor is required for plasma cell differentiation and the unfolded protein response [J]. Immunol Rev,2003,194(1):29-38.
    28. Gass JN, Gifford NM, Brewer JW. Activation of an unfolded protein response during differentiation of antibody-secreting B cells [J]. J Biol Chem,2002, 277(50):49047-49054.
    29. Doody GM, Stephenson S, Tooze RM. BLIMP-1 is a target of cellular stress and downstream of the unfolded protein response [J]. Eur J Immunol,2006, 36(6):1572-1582.
    30. Yoshimura FK, Luo X. Induction of endoplasmic reticulum stress in thymic lymphocytes by the envelope precursor polyprotein of a murine leukemia virus during the preleukemic period. J Virol,2007,81(8):4374-4377.
    31. Kim KH, Gao Y, Walder K, et al. SEPS1 protects RAW264.7 cells from pharmacological ERS agent-induced apoptosis [J]. Biochem Biophys Res Commun,2007,354(1):127-32.
    32. Liu YJ. Dendritic cell subsets and lineages, and their functions in innate and adaptive immunity. Cell,2001,106(3):259-262.
    33. Steinman R M, Banchereau J. Taking dendritic cells into medicine. Nature, 2007,449(27):419-426.
    34. Iwakoshi NN, Pypaert M, Glimcher LH. The transcription factor XBP-1 is essential for the development and survival of dendritic cells. J Exp Med,2007, 204(10):2267-2275.
    1. Mariathasan S, Monack DM. Inflammasome adaptors and sensors: intracellular regulators of infection and inflammation. Nat Rev Immunol, 2007,7(1):31-40.
    2. Watanabe H, Gehrke S, Contassot E, et al. Danger signaling through the inflammasome acts as a master switch between tolerance and sensitization. J Immunol,2008,180(9):5826-5832.
    3. Feldmann J, Prieur AM, Quartier P, et al. Chronic infantile neurological cutaneous and articular syndrome is caused by mutations in CIAS1, a gene highly expressed in polymorphonuclear cells and chondrocytes. Am J Hum Genet,2002,71(1):198-203.
    4. Kanneganti TD, Ozoren N, Body-Malapel M, et al. Bacterial RNA and small antiviral compounds activate caspase-1 through cryopyrin/Nalp3. Nature, 2006,440(7081):233-236.
    5. Martinon F, Petrilli V, Mayor A, et al. Gout-associated uric acid crystals activate the NALP3 inflammasome. Nature,2006,440(7081):237-241.
    6. Mariathasan S, Weiss DS, Newton K, et al. Cryopyrin activates the inflammasome in response to toxins and ATP. Nature,2006, 440(7081):228-232.
    7. Martinon F, Burns K, Tschopp J. The inflammasome:a molecular platform triggering activation of inflammatory caspases and processing of proIL-beta. Mol Cell,2002,10(2):417-426.
    8. Tschopp J, Martinon F, Burns K. NALPs:a novel p rotein family involved in inflammation. Nat RevMol Cell Biol,2003,4(2):95-104.
    9. Franchi L, McDonald C, Kanneganti TD, et al. Nucleotidebinding oligomerization domain-like receptors:intracellular pattern recognition molecules for pathogen detection and host defense. J Immunol,2006,177(6): 3507-3513.
    10. Mariathasan S, Newton K, Monack D M, et al. Differential activation of the inflammasome by caspase-1 adaptors ASC and Ipaf. Nature,2004, 430(6996):213-218.
    11. Poyet JL, Srinivasula SM, Tnani M, et al. Identification of Ipaf, a human caspase-1-activating protein related to Apaf-1. J Biol Chem,2001,276(30): 28309-28313.
    12. Boyden ED, Dietrich WF. Nalplb controls mouse macrophage susceptibility to anthrax lethal toxin. Nat Genet,2006,38(2):240-244.
    13. Fink SL, Bergsbaken T, Cookson BT. Anthrax lethal toxin and Salmonella elicit the common cell death pathway of caspase-1-dependent pyroptosis via distinct mechanisms. Proc Natl Acad Sci U S A.2008,105(11):4312-4317.
    14. Faustin B, Lartigue L, Bruey JM, et al. Reconstituted NALP1 inflammasome reveals two-step mechanism of caspase-1 activation. Mol Cell,2007, 25(5):713-724.
    15. Gonzalez-Benitez JF, Juarez-Verdayes MA, Rodriguez-Martinez S, et al. The NALP3/Cryopyrin-inflammasome complex is expressed in LPS-induced ocular inflammation. Mediators Inflamm,2008,2008:614345.
    16. Muruve DA, Petrilli V, Zaiss AK, et al. The inflammasome recognizes cytosolic microbial and host DNA and triggers an innate immune response. Nature,2008,452(7183):103-107.
    17. Pelegrin P, Surprenant A. Pannexin-1 mediates large pore formation and interleukin-1 release by the ATP-gated P2X7 receptor. EMBO J,2006, 25(21):5071-5082.
    18. Kanneganti TD, Lamkanfi M, Kim YG, et al. Pannexin-1-mediated recognition of bacterial molecules activates the cryopyrin inflammasome independent of Toll-like receptor signaling. Immunity,2007,26(4):433-443.
    19. Giamarellos-Bourboulis E J, Mouktaroudi M, Bodar E, et al. Crystals of monosodium urate monohydrate enhance lipopolysaccharide-induced release of interleukin 1 beta by mononuclear cells through a caspase 1-mediated process. Ann Rheum Dis,2009,68(2):273-278.
    20. Beck C, Morbach H, Richl P, et al. How can calcium pyrophosphate crystals induce inflammation in hypophosphatasia or chronic inflammatory joint diseases? Rheumatol Int,2008,29(3):229-238.
    21. Dostert C, Petrilli V, Van Bruggen R, et al. Innate immune activation through Nalp3 inflammasome sensing of asbestos and silica. Science,2008, 320(5876):674-677.
    22. Amer A, Franchi L, Kanneganti TD, et al. Regulation of Legionella phagosome maturation and infection through flagellin and host Ipaf. J Biol Chem,2006,281(46):35217-35223.
    23. Franchi L, Amer A, Body-Malapel M, et al. Cytosolic flagellin requires Ipaf for activation of caspase-1 and interleukin 1(3 in salmonella-infected macrophages. Nat Immunol,2006,7(6):576-582.
    24. Miao EA, Alpuche-Aranda CM, Dors M, et al. Cytoplasmic flagellin activates caspase-1 and secretion of interleukin 1 via Ipaf. Nat Immunol,2006, 7(6):569-575.
    25. Sutterwala FS, Mijares LA, Li L, et al. Immune recognition of Pseudomonas aeruginosa mediated by the IPAF/NLRC4 inflammasome. J Exp Med,2007, 204(13):3235-3245.
    26. Bruey JM, Bruey-Sedano N, Newman R, et al. PAN1/NALP2/PYPAF2, an inducible inflammatory mediator that regulates NF-κB and caspase-1 activation in macrophages. J Biol Chem,2004,279(50):51897-51907.
    27. Martinon F, Tschopp J. Inflammatory caspases and inflammasomes:master switches of inflammation. Cell Death Differ,2007,14(1):10-22.
    28. Master SS, Rampini SK, Davis AS, et al. Mycobacterium tuberculosis prevents inflammasome activation. Cell Host Microbe,2008,3(4):224-232.
    29. Ikejima T, Okusawa S, Ghezzi P, et al. Interleukin-1 induces tumor necrosis factor (TNF) in human peripheral blood mononuclear cells in vitro and a circulating TNF-like activity in rabbits. J Infect Dis,1990,162(1):215-223.
    30. Shalaby MR, Waage A, Aarden L, et al. Endotoxin, tumor necrosis factor-alpha and interleukin 1 induce interleukin 6 production in vivo. Clin Immunol Immunopathol,1989,53(3):488-498.
    31. Fantuzzi G, Dinarello CA. Interleukin-18 and interleukin-1 beta:two cytokine substrates for ICE(caspase-1). J Clin Immunol,1999,19(1):1-11.
    32. Dinarello CA. Interleukin-1 beta, interleukin-18, and the interleukin-1 beta converting enzyme. Ann N Y Acad Sci,1998,856(1):1-11.
    33. Fahy RJ, Exline MC, Gavrilin MA, et al. Inflammasome mRNA expression in human monocytes during early septic shock. Am J Respir Crit Care Med, 2008,177(9):983-988.
    34. Gotsch F, Romero R, Chaiworapongsa T, et al. Evidence of the involvement of caspase-1 under physiologic and pathologic cellular stress during human pregnancy:a link between the inflammasome and parturition. J Matern Fetal Neonatal Med,2008,21(9):605-616.
    35. Granell M, Urbano-Ispizua A, Pons A, et al. Common variants in NLRP2 and NLRP3 genes are strong prognostic factors for the outcome of HLA-identical sibling allogeneic stem cell transplantation. Blood,2008,112(10):4337-4342.
    36. Kool M, Petrilli V, De Smedt T, et al. Cutting edge:alum adjuvant stimulates inflammatory dendritic cells through activation of the NALP3 inflammasome. J Immunol,2008,181(6):3755-3759.
    37. Keller M, Ruegg A, Werner S, et al. Active caspase-1 is a regulator of unconventional protein secretion. Cell,2008,132(5):818-831.

© 2004-2018 中国地质图书馆版权所有 京ICP备05064691号 京公网安备11010802017129号

地址:北京市海淀区学院路29号 邮编:100083

电话:办公室:(+86 10)66554848;文献借阅、咨询服务、科技查新:66554700