表达免疫刺激因子的重组狂犬疫苗对狂犬病的暴露前及暴露后预防研究
详细信息    本馆镜像全文|  推荐本文 |  |   获取CNKI官网全文
摘要
狂犬病(Rabies)是由RNA病毒引起、通过患狂犬病动物咬、抓伤传播的严重威胁人类健康的人兽共患传染病。每年,全世界大约有55000人死于狂犬病感染,且多于1000万的人接受狂犬病暴露后免疫治疗。大多数人类狂犬病主要发生在亚洲、非洲等发展中国家。在发达国家,由于对宠物的常规免疫,已基本消除了人类狂犬病。当前用于人狂犬病免疫的疫苗主要为灭活疫苗,此种疫苗具有安全、有效等优点,但狂犬病暴露人群通常需要在较长时间(14天)内进行多次免疫注射(至少4次)。此外,此种疫苗价格昂贵,进一步阻止了其在发展中国家的使用。弱毒疫苗或重组活疫苗也被批准用于动物,尤其是野生动物的免疫。这些疫苗已在欧洲及北美洲很大范围内消除了野生动物的狂犬病,但通常会引起副反应,且对犬及臭鼬没有良好的保护效果。因此,狂犬病的控制需要更加廉价、有效的疫苗,尤其是在广大发展中国家。目前,狂犬病仍没有有效的治疗手段,一旦出现临床症状,几乎所有的方法都无能无力。截止今天,全世界共有9人在出现狂犬病症状后依然存活,然而他们中的大多数或产生严重的神经系统后遗症或死于持续的疾病。因此,探讨新的狂犬病治疗方法对于控制狂犬病具有重要意义。
     近来研究表明,先天性免疫应答的激活,尤其是趋化因子及干扰素,是狂犬病致弱的重要分子机制。为进一步研究趋化因子在狂犬病感染中的作用,通过反向遗传技术,将巨噬细胞炎性蛋白(Macrophage inflammatory protein 1α, MIP-1α)克隆入HEP-Flury致弱狂犬病毒株,拯救获得重组病毒并命名为rHEP-MIP1α。rHEP-MIP1α与母本病毒在NA细胞的生长特性相似,这表明重组病毒的体外增殖不受外源基因表达的影响。ELISA检测病毒感染细胞后MIP-1α的表达,发现rHEP-MIP1α以剂量依赖形式表达MIP-1α。将重组病毒脑内注射小鼠,检测MIP-1α表达对重组病毒致病性的影响。与母本病毒相比,rHEP-MIP1α可在中枢神经系统(Central nervous system, CNS)诱导短暂的趋化因子表达及炎性细胞浸润,且其对小鼠的致病性降低。结果说明rHEP-MIP1α可通过诱导短暂的CNS先天性免疫应答进一步降低病毒的致病性。
     为进一步研究rHEP-MIP1α的免疫原性,将重组病毒肌肉免疫小鼠后,以快速荧光灶抑制试验(RFFIT)测定病毒诱导机体产生的中和抗体水平。与母本病毒相比,rHEP-MIP1α肌肉注射后可诱导机体产生显著高的狂犬病中和抗体。以CVS-24强毒进行攻击后,显著多的rHEP-MIP1α免疫小鼠可抵抗强病毒感染。通过荧光定量PCR分析免疫局部的病毒增殖、趋化因子表达及先天性免疫应答分子聚集,结果发现不同病毒在免疫局部的增殖相似,这说明rHEP-MIP1α诱导的适应性免疫应答与病毒在免疫局部的增殖无关。与母本病毒相比,rHEP-MIP1α可在免疫局部诱导表达更高水平的MIP-1α、IL-4、CD19及CD11c。此外,流式细胞术分析发现rHEP-MIP1α可在淋巴结及外周血聚集并激活更多的树突状细胞(Dendritic cells, DCs)及B淋巴细胞。这些数据说明,rHEP-MIP1α可在免疫局部表达高水平的MIP-1α,进而在淋巴结及外周血聚集并激活更多的DCs及B细胞,从而产生高水平的中和抗体。因此,DCs的聚集及激活在增强病毒的保护性免疫应答过程中起重要作用。进一步分析了表达粒细胞-巨噬细胞集落刺激因子(Granulocyte-macrophage colony-stimulating factor, GMCSF )、巨噬细胞来源趋化因子( Macrophage derived chemokines, MDC)及MIP-1α等DCs刺激分子的重组病毒的免疫原性。结果表明,重组病毒表达这些分子后可显著增强DCs的聚集及激活,进而增强病毒的保护性免疫应答。
     为研究表达免疫刺激因子的重组狂犬病毒对狂犬病的暴露后预防作用,在小鼠肌肉感染高致病性狂犬病街毒株后不同时间,用不同重组病毒以脑内(i.c.)、肌注(i.m.)、皮下(i.d.)及滴鼻(i.n.)等途径进行治疗。结果发现,即使在街毒感染后5天开始治疗,表达MIP-1α、GM-CSF、MDC及IP-10的重组病毒仍具有明显的保护作用。尽管紫外线灭活的重组病毒诱导机体产生了较高的中和抗体,但对街毒感染没有保护作用。测定荧光素钠(Sodium fluorescein, NaF)由血液循环进入CNS的量,发现表达免疫刺激因子的重组病毒可显著提高大脑及小脑的血脑屏障(Blood brain barrier, BBB)通透性。通过荧光定量PCR、多重ELISA(Multiplex ELISA)及流式细胞术分析CNS的趋化因子、细胞因子表达及炎性细胞浸润。结果表明,与紫外线灭活病毒相比,表达免疫刺激因子的重组病毒脑内注射后在CNS及外周血引起更高水平的趋化因子及细胞因子表达,炎性细胞浸润。更为重要的是,可增强血脑屏障通透性的趋化因子MCP-1(Chemoattractant protein 1, MCP-1)显著增加了灭活病毒的保护效果。这些数据说明,表达免疫刺激因子的重组病毒可通过在CNS诱导炎性因子表达,增强炎性细胞浸润及提高血脑屏障的通透性,允许更多的炎性细胞或免疫效应因子进入CNS,进而加速病毒的清除及阻止狂犬病感染的发生。
     本研究成功构建了表达免疫刺激因子的重组狂犬病毒,证实了外源基因在病毒基因组的表达对病毒的体外生长没有影响,且重组病毒可通过诱导短暂的先天性免疫应答分子表达进一步降低病毒的致病性,DCs的聚集及激活在增强病毒的保护性免疫应答中具有重要作用,筛选获得的重组疫苗不仅可用于狂犬病的暴露前及暴露后预防免疫,而且还可用于狂犬病的临床治疗。此研究结果为研究狂犬病的致病机理及筛选新型、低毒、高效狂犬病疫苗提供了重要理论依据,并为狂犬病的治疗提供了新的手段及工具。
Rabies is a life-threatening disease caused by an RNA virus that is usually transmitted to human through bites from rabid animals. Each year, approximately 55000 individuals worldwide die of rabies and millions more undergo post-exposure prophylaxis (PEP). Most of the human cases occur in the developing nations of Asia and Africa where dog rabies remains to be the main source for human exposure. In the developed countries, human rabies has dramatically declined as a direct consequence of routine vaccination of pet animals. Current rabies vaccines used for human are inactivated virus vaccine. Although these vaccines are safe and efficacious, multiple doses (at least 4) must be administered over an extended period of time (14 days) to people who are exposed to rabies or suspected rabid animals. In addition, the high cost associated with these inactivated rabies virus (RABV) vaccines prevents their effective use in developing countries where the vaccines are needed most. Live attenuated RABV vaccines or recombinant live vaccines have been licensed, particularly for wild animals. These vaccines have been used for large scale elimination of rabies in Europe as well as in North America. However, they usually induce intensive skin inflammation and not worked well for dogs and skunks. Furthermore, those vaccines are not so effective in oral vaccination for dogs and skunks. Therefore, more efficacious and affordable RABV vaccines are needed, particularly in the developing nations. Once the clinic manifestations of rabies developed, however, treatment options for rabies are limited. To date, only nine individuals have survived rabies virus infection. Unfortunately, most of these patients died either after prolonged illness or developed severe neurogical sequel. Therefore, more effective therapeutics is needed in management of clinical rabies.
     It was found previously that induction of innate immunity, particularly chemokines and type I interferons, is an important mechanism of rabies virus attenuation. To evaluated the effect of overexpression of chemokines on RABV infection. Macrophage inflammatory protein 1α(MIP-1α, CCL3) was cloned into the genome of attenuated RABV strain HEP-Flury, the recombinant rabies virus (rRABV) was rescued and designated as rHEP-MIP1α. To characterize the rRABV in vitro, virus growth kinetics was examined in NA cells. No significant difference was found between the rRABV and parental virus, indicating that the viral growth was not affected by the insertion of MIP-1α. The ability of rHEP-MIP1αto produce MIP-1αwas determined by measuring MIP-1αin virus-infected cells with ELISA kits. It was found that MIP-1αwas expressed by rHEP-MIP1αin a dose-dependent manner. To determine the effect of chemokine expression on RABV infection, mice were infected with the rRABV by intracerebral route. A transient MIP-1αexpression and inflammatory cell infiltration was induced in the central nervous system (CNS) of mice infected with rHEP-MIP1α, compared with those infected with parental virus. The pathogenicity of rHEP-MIP1αwas more decreased than parental virus. It indicates that overexpression of MIP-1αfurther decreased RABV pathogenicity by inducing a transient innate immune response in the CNS.
     To investigate the immunogenicity of rHEP-MIP1α, mice were immunized with rRABV by the intramuscular (i.c.) route. The virus neutralizing antibody titer (VNA) in serum was measured by the rapid fluorescent focus inhibiton test (RFFIT). It was found that the level of VNA was significantly higher in mice immunized with rHEP-MIP1αthan that induced by immunization with parent virus. After challenge with virulent CVS-24, more mice immunized with rHEP-MIP1αsurvived than mice immunized with the parental virus. Virus replication, expression of chemokines, and/or recruitment of immune cells at local sites was determined by quantitative real-time PCR (qRT-PCR). There was no difference in virus replication in mice infected with each virus, indicating the induction of adaptive immunity by rHEP-MIP1αis not due to the rate of virus replication at the local site. However, significantly more MIP-1α, CD19, CD11c and IL-4 mRNA was detected in the muscle tissue of mice infected with rHEP-MIP1αthan in mice infected with the parent virus. CD11c dendritic cells (DCs) and B cells were analyzed by flow cytometry. It was found that the recruitment of DCs and B cells in lymph node and peripheral blood was significantly increased in mice immunized with rHEP-MIP1α, than those immunized with parent virus. All the data demonstrate that expression of MIP-1αenhances the immunogenicity by recruiting DCs and B cells to the site of immunization, lymph nodes, and the blood. Therefore, recruitment and/or activation of DCs play an important role in enhancing the protective immune response against RABV. To address the importance of DCs activation for RABV vaccine efficacy, several DCs stimulatory molecules, e.g. granulocyte-macrophage colony-stimulating factor (GM-CSF), macrophage derived chemokines (MDC), and MIP-1α, were cloned into the genome of RABV strain SAD L16. The rRABVs were rescued and designated as rLBNSE-GM-CSF, rLBNSE-MDC, and rLBNSE-MIP1α, respectively. It was found that the rRABVs expressed these molecules activate/recruit DCs and enhanced protective immune response.
     To investigate if the rRABVs expressing the immunostimulatory molecules have the ability to prevent animals from developing rabies, mice were infected with lethal doses of street RABV and then treated with rRABV at different time points after infection by intracerebral (i.c.), intramuscular (i.m.), intradermal (i.d.) or intranasal (i.n.) route. It was found that significantly more mice intracerebrally treated with rRABV expressing MIP-1α, GM-CSF, macrophage-derived chemokine (MDC), or IP-10 as late as day 5 after infection with virulent RABV were protected from developing rabies than sham-treated mice. On the other hand, treatment with UV-inactivated rRABV did not provide protection despite the fact that virus VNA were induced in the periphery. The leakage of fluorescein sodium (NaF) from the circulation into CNS tissues was measured in cerebrum, cerebellum or spinal cord. It was found that BBB permeability to NaF was significantly elevated in the cerebrum and cerebellum of mice treated with rRABVs than sham-treated mice. Expression of chemokines/cytokines, and infiltration of inflammatory cells were measured by qRT-PCR, multiplex ELISA and flow cytometry. Intracerebral treatment of mice with live rRABVs induced significantly higher levels of chemokine/cytokine expression in the CNS and in the periphery, and more infiltration of inflammatory and immune cells into the CNS, than sham-treated mice or mice treated with UV-inactivated rRABV. Most importantly, treatment with a chemokine (chemoattractant protein-1, MCP-1, also termed CCL2) with doses known to enhance BBB permeability increased the protective efficacy of UV-inactivated rRABV. In sum, these studies confirm that chemokines can enhance the BBB permeability, allow infiltration of inflammatory cells and other immune effectors enter into the CNS to clear the virus and to prevent the development of rabies.
     In the present study, rRABVs expressing the immunostimulatory molecules were constructed. It was found that the viral growth was not affected by the insertion of foreign genes. Overexpression of these molecules further decreased RABV pathogenicity by inducing a transient innate immune response. Recruitment and activation of DCs is important in enhancing the protective immune responses against rabies. These rRBAVs expressing immunostimulatory molecules could have the potential to be used not only for pre- and post-exposure immunization but also for therapy in clinical rabies. The data from this study provides an important theoretical basis for study on pathogenesis of RABV, and selection of more viable and affordable RABV vaccines. It may also provide a new therapeutic of management of clinical rabies.
引文
[1] Hemachudha T, Laothamatas J, Rupprecht CE. Human rabies: a disease of complex neuropathogenetic mechanisms and diagnostic challenges [J]. Lancet Neurol, 2002, 1: 101-109.
    [2] Pulmanausahakul R, Li J, Schnell MJ, et al. The glycoprotein and the matrix protein of rabies virus affect pathogenicity by regulating viral replication and facilitating cell-to-cell spread [J]. J Virol, 2008, 82: 2330-2338.
    [3] Menager P, Roux P, Megret F, et al. Toll-like receptor 3 (TLR3) plays a major role in the formation of rabies virus Negri Bodies [J]. PLoS Pathog, 2009, 5: e1000315.
    [4] Mebatsion T, Weiland F, Conzelmann KK. Matrix protein of rabies virus is responsible for the assembly and budding of bullet-shaped particles and interacts with the transmembrane spike glycoprotein G [J]. J Virol, 1999, 73: 242-250.
    [5] Yang J, Hooper DC, Wunner WH, et al. The specificity of rabies virus RNA encapsidation by nucleoprotein [J]. Virology, 1998, 242: 107-117.
    [6] Yang J, Koprowski H, Dietzschold B, et al. Phosphorylation of rabies virus nucleoprotein regulates viral RNA transcription and replication by modulating leader RNA encapsidation [J]. J Virol, 1999, 73: 1661-1664.
    [7] Chenik M, Chebli K, Blondel D. Translation initiation at alternate in-frame AUG codons in the rabies virus phosphoprotein mRNA is mediated by a ribosomal leaky scanning mechanism [J]. J Virol 1995, 69: 707-712.
    [8] Gigant B, Iseni F, Gaudin Y, et al. Neither phosphorylation nor the amino-terminal part of rabies virus phosphoprotein is required for its oligomerization [J]. J Gen Virol, 2000, 81: 1757-1761.
    [9] Shoji Y, Inoue S, Nakamichi K, et al. Generation and characterization of P gene-deficient rabies virus [J]. Virology, 2004, 318: 295-305.
    [10] Raux H, Flamand A, Blondel D. Interaction of the rabies virus P protein with the LC8 dynein light chain [J]. J Virol ,2000, 74: 10212-10216.
    [11] Tan GS, Preuss MA, Williams JC, et al. The dynein light chain 8 binding motif of rabies virus phosphoprotein promotes efficient viral transcription [J]. Proc Natl Acad Sci U S A, 2007, 104: 7229-7234.
    [12] Brzozka K, Finke S, Conzelmann KK. Identification of the rabies virus alpha/beta interferon antagonist: phosphoprotein P interferes with phosphorylation of interferon regulatory factor 3 [J]. J Virol, 2005, 79: 7673-7681.
    [13] Brzozka K, Finke S, Conzelmann KK. Inhibition of interferon signaling by rabies virus phosphoprotein P: activation-dependent binding of STAT1 and STAT2 [J]. J Virol, 2006, 80: 2675-2683.
    [14] Vidy A, Chelbi-Alix M, Blondel D. Rabies virus P protein interacts with STAT1 and inhibits interferon signal transduction pathways [J]. J Virol, 2005, 79: 14411-14420.
    [15] Vidy A, El Bougrini J, Chelbi-Alix MK, et al. The nucleocytoplasmic rabies virus P proteincounteracts interferon signaling by inhibiting both nuclear accumulation and DNA binding of STAT1[J]. J Virol ,2007, 81: 4255-4263.
    [16] Komarova AV, Real E, Borman AM, et al. Rabies virus matrix protein interplay with eIF3, new insights into rabies virus pathogenesis[J]. Nucleic Acids Res, 2007, 35: 1522-1532.
    [17] Flamand A, Raux H, Gaudin Y, et al. Mechanisms of rabies virus neutralization [J].Virology ,1993,194: 302-313.
    [18] Schnell MJ, Tan GS, Dietzschold B.The application of reverse genetics technology in the study of rabies virus (RV) pathogenesis and for the development of novel RV vaccines [J]. J Neurovirol , 2005,11: 76-81.
    [19] Mazarakis ND, Azzouz M, Rohll JB, et al. Rabies virus glycoprotein pseudotyping of lentiviral vectors enables retrograde axonal transport and access to the nervous system after peripheral delivery [J]. Hum Mol Genet, 2001, 10: 2109-2121.
    [20] Faber M, Pulmanausahakul R, Hodawadekar SS, et al. Overexpression of the rabies virus glycoprotein results in enhancement of apoptosis and antiviral immune response[J]. J Virol, 2002, 76: 3374-3381.
    [21] Morimoto K, Hooper DC, Spitsin S,et al. Pathogenicity of different rabies virus variants inversely correlates with apoptosis and rabies virus glycoprotein expression in infected primary neuron cultures[J]. J Virol , 1999, 73: 510-518.
    [22] Prehaud C, Lay S, Dietzschold B, et al.Glycoprotein of nonpathogenic rabies viruses is a key determinant of human cell apoptosis [J]. J Virol, 2003, 77: 10537-10547.
    [23] Sarmento L, Li XQ, Howerth E, J et al. Glycoprotein-mediated induction of apoptosis limits the spread of attenuated rabies viruses in the central nervous system of mice [J]. J Neurovirol, 2005, 11: 571-581.
    [24] Yan X, Prosniak M, Curtis MT, et al. Silver-haired bat rabies virus variant does not induce apoptosis in the brain of experimentally infected mice[J]. J Neuroviro, 2001, 7: 518-527.
    [25] Yan X, Mohankumar PS, Dietzschold B, et al.The rabies virus glycoprotein determines the distribution of different rabies virus strains in the brain [J]. J Neurovirol, 2002, 8: 345-352.
    [26] Schnell MJ, Conzelmann KK. Polymerase activity of in vitro mutated rabies virus L protein [J]. Virology ,1995, 214: 522-530.
    [27] Finke S, Conzelmann KK. Replication strategies of rabies virus[J]. Virus Res, 2005, 111: 120-131.
    [28] Finke S, Conzelmann KK. Dissociation of rabies virus matrix protein functions in regulation of viral RNA synthesis and virus assembly [J]. J Virol, 2003, 77: 12074-12082.
    [29] Finke S, Mueller-Waldeck R, Conzelmann KK. Rabies virus matrix protein regulates the balance of virus transcription and replication [J]. J Gen Virol, 2003, 84: 1613-1621.
    [30] Rupprecht CE, Gibbons RV. Clinical practice. Prophylaxis against rabies[J]. N Engl J Med, 2004, 351: 2626-2635.
    [31] Messenger SL, Smith JS, Rupprecht CE. Emerging epidemiology of bat-associated cryptic cases of rabies in humans in the United States[J]. Clin Infect Dis,2002, 35: 738-747.
    [32] Srinivasan A, Burton EC, Kuehnert MJ, et al. Transmission of rabies virus from an organ donor to four transplant recipients [J]. N Engl J Med, 2005, 352: 1103-1111.
    [33] Leung AK, Davies HD, Hon KL. Rabies: epidemiology, pathogenesis, and prophylaxis[J]. Adv Ther ,2007, 24: 1340-1347.
    [34] Jackson AC. Recovery from rabies[J]. N Engl J Med, 2005, 352: 2549-2550.
    [35] Wiktor TJ, Fernandes MV, Koprowski H Cultivation of Rabies Virus in Human Diploid Cell Strain Wi-38[J]. J Immunol, 1964, 93: 353-366.
    [36] Fu ZF Rabies and rabies research: past, present and future[J]. Vaccine, 1997, 15 Suppl: S20-24.
    [37] Wandeler AI. Oral immunization against rabies: afterthoughts and foresight[J].Schweiz Arch Tierheilkd, 2000, 142: 455-462.
    [38] Tolson ND, Charlton KM, Stewart RB, et al. Immune response in skunks to a vaccinia virus recombinant expressing the rabies virus glycoprotein[J]. Can J Vet Res,1987, 51: 363-366.
    [39] Rupprecht CE, Smith JS, Fekadu M, et al, The ascension of wildlife rabies: a cause for public health concern or intervention? [J] Emerg Infect Dis, 1995, 1: 107-114.
    [40] Hanlon CA, Niezgoda M, Morrill P, Rupprecht CE , 2002, Oral efficacy of an attenuated rabies virus vaccine in skunks and raccoons[J]. J Wildl Dis 38: 420-427.
    [41] Schnell MJ, Mebatsion T, Conzelmann KK. Infectious rabies viruses from cloned cDNA[J]. EMBO J, 1994, 13: 4195-4203.
    [42] McGettigan JP, Foley HD, Belyakov IM, et al. Rabies virus-based vectors expressing human immunodeficiency virus type 1 (HIV-1) envelope protein induce a strong, cross-reactive cytotoxic T-lymphocyte response against envelope proteins from different HIV-1 isolates[J]. J Virol, 2001, 75: 4430-4434.
    [43] Siler CA, McGettigan JP, Dietzschold B, et al. Live and killed rhabdovirus-based vectors as potential hepatitis C vaccines [J]. Virology, 2002, 292: 24-34.
    [44] Faber M, Lamirande EW, Roberts A, et al. A single immunization with a rhabdovirus-based vector expressing severe acute respiratory syndrome coronavirus (SARS-CoV) S protein results in the production of high levels of SARS-CoV-neutralizing antibodies[J]. J Gen Virol, 2005, 86: 1435-1440.
    [45] Pulmanausahakul R, Faber M, Morimoto K, et al. Overexpression of cytochrome C by a recombinant rabies virus attenuates pathogenicity and enhances antiviral immunity[J]. J Virol, 2001, 75: 10800-10807.
    [46] Faber M, Bette M, Preuss MA, et al. Overexpression of tumor necrosis factor alpha by a recombinant rabies virus attenuates replication in neurons and prevents lethal infection in mice[J]. J Virol, 2005, 79: 15405-15416.
    [47] McGettigan JP, Koser ML, McKenna PM, et al. Enhanced humoral HIV-1-specific immune responses generated from recombinant rhabdoviral-based vaccine vectors co-expressing HIV-1 proteins and IL-2 [J]. Virology ,2006, 344: 363-377.
    [48] Faber M, Li J, Kean RB, et al. Effective preexposure and postexposure prophylaxis of rabies with a highly attenuated recombinant rabies virus[J]. Proc Natl Acad Sci U S A , 2009,106: 11300-11305.
    [49] Mebatsion T. Extensive attenuation of rabies virus by simultaneously modifying the dynein light chain binding site in the P protein and replacing Arg333 in the G protein [J]. J Virol, 2001, 75:11496-11502.
    [50] Wang ZW, Sarmento L, Wang Y, et al. Attenuated rabies virus activates, while pathogenic rabies virus evades, the host innate immune responses in the central nervous system [J]. J Virol, 2005, 79: 12554-12565.
    [51] Manning SE, Rupprecht CE, Fishbein D, et al. Human rabies prevention--United States, 2008: recommendations of the Advisory Committee on Immunization Practices[J]. MMWR Recomm Rep, 2008, 57: 1-28.
    [52] McDermid RC, Saxinger L, Lee B, et al. Human rabies encephalitis following bat exposure: failure of therapeutic coma[J]. CMAJ, 2008, 178: 557-561.
    [53] Rupprecht CE BD, Brown CM, Franka R, et al. Use of a reduced (4-dose) vaccine schedule for postexposure prophylaxis to prevent human rabies: recommendations of the advisory committee on immunization practices[J]. MMWR Recomm Rep 2010 Mar 19;59(RR-2):1-9.
    [54] Howdieshell TR, Heffernan D, Dipiro JT. Surgical infection society guidelines for vaccination after traumatic injury[J]. Surg Infect (Larchmt), 2006, 7: 275-303.
    [55] CDC , 2010, Presumptive abortive human rabies - Texas, 2009. MMWR Morb Mortal Wkly Rep 59: 185-190.
    [56] Jackson AC, Warrell MJ, Rupprecht CE, et al. Management of rabies in humans[J]. Clin Infect Dis, 2003, 36: 60-63.
    [57] Hattwick MA, Weis TT, Stechschulte CJ, et al. Recovery from rabies. A case report. Ann Intern Med, 1972, 76: 931-942.
    [58] Wilde H, Hemachudha T, Jackson AC. Viewpoint: Management of human rabies[J]. Trans R Soc Trop Med Hyg , 2008, 102: 979-982.
    [59] Willoughby RE, Jr., Tieves KS, Hoffman GM, et al. Survival after treatment of rabies with induction of coma[J]. N Engl J Med, 2005, 352: 2508-2514.
    [60] Schmiedel S, Panning M, Lohse A, et al. Case report on fatal human rabies infection in Hamburg, Germany[J], March 2007. Euro Surveill 12: E070531 070535.
    [61] Rubin J, David D, Willoughby RE, et al. Applying the Milwaukee protocol to treat canine rabies in Equatorial Guinea [J]. Scand J Infect Dis, 2009, 41: 372-375.
    [62] Hunter M, Johnson N, Hedderwick S, et al. Immunovirological correlates in human rabies treated with therapeutic coma[J]. J Med Virol , 2010, 82: 1255-1265.
    [63] Hemachudha T, Sunsaneewitayakul B, Desudchit T, et al. Failure of therapeutic coma and ketamine for therapy of human rabies[J]. J Neurovirol, 2006, 12: 407-409.
    [64] Noah DL, Drenzek CL, Smith JS, et al. Epidemiology of human rabies in the United States, 1980 to 1996. Ann Intern Med, 1998, 128: 922-930.
    [65] Superti F, Seganti L, Pana A, et al. Effect of amantadine on rhabdovirus infection[J]. Drugs Exp Clin Res ,1985, 11: 69-74.
    [66] Dietzschold B, Li J, Faber M, et al. Concepts in the pathogenesis of rabies [J]. Future Virol, 2008, 3: 481-490.
    [67] Dietzschold B, Wang HH, Rupprecht CE, et al. Induction of protective immunity against rabies by immunization with rabies virus ribonucleoprotein[J]. Proc Natl Acad Sci U S A, 1987, 84: 9165-9169.
    [68] Tollis M, Dietzschold B, Volia CB, et al. Immunization of monkeys with rabies ribonucleoprotein (RNP) confers protective immunity against rabies[J]. Vaccine, 1991, 9: 134-136.
    [69] Anderson LJ, Williams LP, Jr., Layde JB, et al. Nosocomial rabies: investigation of contacts of human rabies cases associated with a corneal transplant[J]. Am J Public Health, 1984, 74: 370-372.
    [70] CDC. Rabies in laboratory wirker - Texas. Morb Mortal Wkly Rep, 1972: 1113-1124.
    [71] CDC. Rabies in laboratory worker - New York. Morb Mortal Wkly Rep, 1977: 183-184.
    [72] Constantine DG. Rabies transmission by nonbite route. Public Health Rep, 1962, 77: 287-289.
    [73] Charlton KM, Casey GA. Experimental rabies in skunks: immunofluorescence light and electron microscopic studies [J]. Lab Invest ,1979, 41: 36-44.
    [74] Murphy FA, Bauer SP. Early street rabies virus infection in striated muscle and later progression to the central nervous system[J]. Intervirology, 1974, 3: 256-268.
    [75] Shankar V, Dietzschold B, Koprowski H. Direct entry of rabies virus into the central nervous system without prior local replication [J]. J Virol, 1991,65: 2736-2738.
    [76] Klingen Y, Conzelmann KK, Finke S. Double-labeled rabies virus: live tracking of enveloped virus transport[J]. J Virol , 2008, 82: 237-245.
    [77] Tsiang H. Evidence for an intraaxonal transport of fixed and street rabies virus[J]. J Neuropathol Exp Neurol, 1979, 38: 286-299.
    [78] Murphy FA. Rabies pathogenesis[J]. Arch Virol, 1977, 54: 279-297.
    [79] Charlton KM, Casey GA, Campbell JB. Experimental rabies in skunks: mechanisms of infection of the salivary glands[J]. Can J Comp Med, 1983, 47: 363-369.
    [80] Jackson AC, Rossiter JP. Apoptosis plays an important role in experimental rabies virus infection[J]. J Virol , 1997, 71: 5603-5607.
    [81] Fu ZF, Weihe E, Zheng YM, et al. Differential effects of rabies and borna disease viruses on immediate-early- and late-response gene expression in brain tissues[J]. J Virol , 1993, 67: 6674-6681.
    [82] Tsiang H Neuronal function impairment in rabies-infected rat brain[J]. J Gen Virol, 1982, 61 (Pt 2): 277-281.
    [83] Ceccaldi PE, Fillion MP, Ermine A. Rabies virus selectively alters 5-HT1 receptor subtypes in rat brain[J]. Eur J Pharmacol , 1993, 245: 129-138.
    [84] Bouzamondo E, Ladogana A, Tsiang H. Alteration of potassium-evoked 5-HT release from virus-infected rat cortical synaptosomes[J]. Neuroreport, 1993, 4: 555-558.
    [85] Iwata M, Komori S, Unno T. Modification of membrane currents in mouse neuroblastoma cells following infection with rabies virus[J]. Br J Pharmacol , 1999,126: 1691-1698.
    [86] Hooper DC, Morimoto K, Bette M. et al. Collaboration of antibody and inflammation in clearance of rabies virus from the central nervous system[J]. J Virol, 1998, 72: 3711-3719.
    [87] Dietzschold B, Faber M, Schnell MJ. New approaches to the prevention and eradication of rabies[J]. Expert Rev Vaccines, 2003, 2: 399-406.
    [88] Wiktor TJ, Doherty PC, Koprowski H. Suppression of cell-mediated immunity by street rabiesvirus[J]. J Exp Med, 1977, 145: 1617-1622.
    [89] Lafon M. Subversive neuroinvasive strategy of rabies virus[J]. Arch Virol Suppl, 2004,: 149-159.
    [90] Dietzschold B, Schnell M, Koprowski H. Pathogenesis of rabies[J]. Curr Top Microbiol Immunol, 2005, 292: 45-56.
    [91] TL Lentz TB, AL Smith, J Crick,et al. Is the acetylcholine receptor a rabies virus receptor? [J]. Science, 1982, 215: 182-184.
    [92] Thoulouze MI, Lafage M, Schachner M, et al. The neural cell adhesion molecule is a receptor for rabies virus [J]. J Virol, 1998, 72: 7181-7190.
    [93] Tuffereau C, Benejean J, Blondel D, et al. Low-affinity nerve-growth factor receptor (P75NTR) can serve as a receptor for rabies virus[J]. EMBO J, 1998, 17: 7250-7259.
    [94] Tuffereau C, Schmidt K, Langevin C, et al. The rabies virus glycoprotein receptor p75NTR is not essential for rabies virus infection[J]. J Virol , 2007, 81: 13622-13630.
    [95] Lewis P LT. Rabies virus entry into cultured rat hippocampal neurons[J]. J Neurocytol , 1998, 27: 559-573.
    [96] Marsh M HA. Virus entry into animal cells[J]. Adv Virus Res, 1989, 36: 107-151.
    [97] Gaudin Y, Tuffereau C, Segretain D, et al. Reversible conformational changes and fusion activity of rabies virus glycoprotein[J]. J Virol, 1991, 65: 4853-4859.
    [98] Dietzschold B, Wunner WH, Wiktor TJ, et al. Characterization of an antigenic determinant of the glycoprotein that correlates with pathogenicity of rabies virus[J]. Proc Natl Acad Sci U S A, 1983, 80: 70-74.
    [99] Faber M, Faber ML, Papaneri A, et al. A single amino acid change in rabies virus glycoprotein increases virus spread and enhances virus pathogenicity[J]. J Virol, 2005, 79: 14141-14148.
    [100] Faber M, Pulmanausahakul R, Nagao K, et al. Identification of viral genomic elements responsible for rabies virus neuroinvasiveness[J]. Proc Natl Acad Sci U S A , 2004, 101: 16328-16332.
    [101] Dietzschold B, Wiktor TJ, Trojanowski JQ, et al. Differences in cell-to-cell spread of pathogenic and apathogenic rabies virus in vivo and in vitro[J]. J Virol, 1985, 56: 12-18.
    [102] Morimoto K, Ni YJ, Kawai A. Syncytium formation is induced in the murine neuroblastoma cell cultures which produce pathogenic type G proteins of the rabies virus[J]. Virology, 1992, 189: 203-216.
    [103] Coulon P, Derbin C, Kucera P, et al. Invasion of the peripheral nervous systems of adult mice by the CVS strain of rabies virus and its avirulent derivative AvO1[J]. J Virol ,1989, 63: 3550-3554.
    [104] Kucera P, Dolivo M, Coulon P, et al. Pathways of the early propagation of virulent and avirulent rabies strains from the eye to the brain[J]. J Virol, 1985, 55: 158-162.
    [105] Fredericksen BL, Whitt MA. Vesicular stomatitis virus glycoprotein mutations that affect membrane fusion activity and abolish virus infectivity[J]. J Virol, 1995, 69: 1435-1443.
    [106] Tang Y, Rampin O, Giuliano F, et al. Spinal and brain circuits to motoneurons of the bulbospongiosus muscle: retrograde transneuronal tracing with rabies virus[J]. J Comp Neurol, 1999, 414: 167-192.
    [107] Kelly RM SP. Rabies as a transneuronal tracer of circuits in the central nervous system [J]. JNeurosci Methods , 2000, 103: 63-71.
    [108] Etessami R, Conzelmann KK, Fadai-Ghotbi B, et al. Spread and pathogenic characteristics of a G-deficient rabies virus recombinant: an in vitro and in vivo study[J]. J Gen Virol, 2000, 81: 2147-2153.
    [109] Jacob Y, Badrane H, Ceccaldi PE, et al. Cytoplasmic dynein LC8 interacts with lyssavirus phosphoprotein[J]. J Virol, 2000, 74: 10217-10222.
    [110] Faber M, Faber ML, Li J, et al. Dominance of a nonpathogenic glycoprotein gene over a pathogenic glycoprotein gene in rabies virus[J]. J Virol , 2007, 81: 7041-7047.
    [111] Otsuka M, Jing Q, Georgel P, et al. Hypersusceptibility to vesicular stomatitis virus infection in Dicer1-deficient mice is due to impaired miR24 and miR93 expression[J]. Immunity, 2007, 27: 123-134.
    [112] Jopling CL, Norman KL, Sarnow P. Positive and negative modulation of viral and cellular mRNAs by liver-specific microRNA miR-122[J]. Cold Spring Harb Symp Quant Biol , 2006, 71: 369-376.
    [113] Randall G, Panis M, Cooper JD, et al. , 2007, Cellular cofactors affecting hepatitis C virus infection and replication[J]. Proc Natl Acad Sci U S A 104: 12884-12889.
    [114] Uller L, Andersson M, Greiff L, et al. Occurrence of apoptosis, secondary necrosis, and cytolysis in eosinophilic nasal polyps[J]. Am J Respir Crit Care Med, 2004, 170: 742-747.
    [115] Ting JP WS, Bergstralh DT. NLRs at the intersection of cell death and immunity[J]. Nat Rev Immunol, 2008, 8: 372-379.
    [116] Kono H RK. How dying cells alert the immune system to danger[J]. Nat Rev Immunol, 2008, 8: 279-289.
    [117] Dietzschold B, Morimoto K, Hooper DC, et al. Genotypic and phenotypic diversity of rabies virus variants involved in human rabies: implications for postexposure prophylaxis[J]. J Hum Virol , 2000, 3: 50-57.
    [118] Li J, McGettigan JP, Faber M, et al. Infection of monocytes or immature dendritic cells (DCs) with an attenuated rabies virus results in DC maturation and a strong activation of the NFkappaB signaling pathway[J]. Vaccine, 2008, 26: 419-426.
    [119] Zhao L, Toriumi H, Kuang Y, et al. The roles of chemokines in rabies virus infection: overexpression may not always be beneficial[J]. J Virol , 2009, 83: 11808-11818.
    [120] Banchereau J, Briere F, Caux C, et al. Immunobiology of dendritic cells[J]. Annu Rev Immunol, 2000, 18: 767-811.
    [121] Banchereau J, Steinman RM. Dendritic cells and the control of immunity[J]. Nature, 1998, 392: 245-252.
    [122] Blondel D, Regad T, Poisson N, et al. Rabies virus P and small P products interact directly with PML and reorganize PML nuclear bodies[J]. Oncogene, 2002, 21: 7957-7970.
    [123] Hooper DC. The role of immune responses in the pathogenesis of rabies[J]. J Neurovirol, 2005, 11: 88-92.
    [124] Irwin DJ, Wunner WH, Ertl HC, et al. Basis of rabies virus neurovirulence in mice: expression of major histocompatibility complex class I and class II mRNAs[J]. J Neurovirol, 1999, 5: 485-494.
    [125] Hirai K, Kawano H, Mifune K, et al. Suppression of cell-mediated immunity by street rabies virus infection[J]. Microbiol Immunol, 1992, 36: 1277-1290.
    [126] Camelo S, Lafage M, Galelli A,et al. Selective role for the p55 Kd TNF-alpha receptor in immune unresponsiveness induced by an acute viral encephalitis[J]. J Neuroimmunol, 2001, 113: 95-108.
    [127] Camelo S, Lafage M, Lafon M. Absence of the p55 Kd TNF-alpha receptor promotes survival in rabies virus acute encephalitis[J]. J Neurovirol , 2000, 6: 507-518.
    [128] Baloul L, Lafon M. Apoptosis and rabies virus neuroinvasion[J]. Biochimie, 2003, 85: 777-788.
    [129] Sugamata M, Miyazawa M, Mori S, et al. Paralysis of street rabies virus-infected mice is dependent on T lymphocytes[J]. J Virol, 1992, 66: 1252-1260.
    [130] Weiland F, Cox JH, Meyer S, et al. Rabies virus neuritic paralysis: immunopathogenesis of nonfatal paralytic rabies[J]. J Virol, 1992, 66: 5096-5099.
    [131] Hooper DC, Ohnishi ST, Kean R, et al. Local nitric oxide production in viral and autoimmune diseases of the central nervous system[J]. Proc Natl Acad Sci U S A, 1995, 92: 5312-5316.
    [132] Astoul E, Lafage M, Lafon M. Rabies superantigen as a Vbeta T-dependent adjuvant[J]. J Exp Med, 1996, 183: 1623-1631.
    [133] Prosniak M, Hooper DC, Dietzschold B, et al. Effect of rabies virus infection on gene expression in mouse brain[J]. Proc Natl Acad Sci U S A, 2001, 98: 2758-2763.
    [134] Ray NB, Ewalt LC, Lodmell DL. Rabies virus replication in primary murine bone marrow macrophages and in human and murine macrophage-like cell lines: implications for viral persistence[J]. J Virol, 1995, 69: 764-772.
    [135] Morimoto K, Patel M, Corisdeo S, et al. Characterization of a unique variant of bat rabies virus responsible for newly emerging human cases in North America[J].Proc Natl Acad Sci U S A, 1996, 93: 5653-5658.
    [136] Thoulouze MI, Lafage M, Montano-Hirose JA, et al. Rabies virus infects mouse and human lymphocytes and induces apoptosis[J]. J Virol, 1997, 71: 7372-7380.
    [137] CDC. Investigation of rabies infections in organ donor and transplant recipients--Alabama, Arkansas, Oklahoma, and Texas[J], 2004. MMWR Morb Mortal Wkly Rep 53: 586-589.
    [138] Baer GM, Shaddock JH, Moore SA, et al. Successful prophylaxis against rabies in mice and Rhesus monkeys: the interferon system and vaccine[J]. J Infect Dis, 1977, 136: 286-291.
    [139] Leroy M, Pire G, Baise E, et al. Expression of the interferon-alpha/beta-inducible bovine Mx1 dynamin interferes with replication of rabies virus[J]. Neurobiol Dis, 2006, 21: 515-521.
    [140] Janeway CA, Jr. Approaching the asymptote? Evolution and revolution in immunology[J]. Cold Spring Harb Symp Quant Biol,1989, 54 Pt 1: 1-13.
    [141] West AP, Koblansky AA, Ghosh S. Recognition and signaling by toll-like receptors[J]. Annu Rev Cell Dev Biol, 2006, 22: 409-437.
    [142] Guidotti LG, Chisari FV. Cytokine-mediated control of viral infections[J]. Virology, 2000, 273: 221-227.
    [143] Tough ALBaDF. Links between innate and adaptive immunity via type I interferon[J]. Current Opinion in Immunology, 2002, 14: 432-436.
    [144] Nakamichi K, Inoue S, Takasaki T, et al. Rabies virus stimulates nitric oxide production and CXC chemokine ligand 10 expression in macrophages through activation of extracellular signal-regulated kinases 1 and 2[J]. J Virol, 2004, 78: 9376-9388.
    [145] Mendonca RZ, Pereira CA. Relationship of interferon synthesis and the resistance of mice infected with street rabies virus[J]. Braz J Med Biol Res, 1994, 27: 691-695.
    [146] Zlotnik A YO. Chemokines: a new classification system and their role in immunity [J]. Immunity, 2000, 12: 121-127.
    [147] Dorner BG SA, Rolph MS, Huser MB, et al. MIP-1alpha, MIP-1beta, RANTES, and ATAC/lymphotactin function together with IFN-gamma as type 1 cytokines[J]. Proc Natl Acad Sci U S A, 2002, 99: 6181-6186.
    [148] Chen BP, Kuziel WA, Lane TE. Lack of CCR2 results in increased mortality and impaired leukocyte activation and trafficking following infection of the central nervous system with a neurotropic coronavirus[J]. J Immunol, 2001, 167: 4585-4592.
    [149] Trumpfheller C, Tenner-Racz K, Racz P, et al. Expression of macrophage inflammatory protein (MIP)-1alpha, MIP-1beta, and RANTES genes in lymph nodes from HIV+ individuals: correlation with a Th1-type cytokine response[J]. Clin Exp Immunol, 1998, 112: 92-99.
    [150] Takeda K, Akira S. Toll-like receptors in innate immunity[J]. Int Immunol, 2005, 17: 1-14.
    [151] Takeshita F, Tanaka T, Matsuda T, et al. Toll-like receptor adaptor molecules enhance DNA-raised adaptive immune responses against influenza and tumors through activation of innate immunity[J]. J Virol, 2006, 80: 6218-6224.
    [152] Messenger SL, Smith JS, Orciari LA, et al. Emerging pattern of rabies deaths and increased viral infectivity[J]. Emerg Infect Dis, 2003, 9: 151-154.
    [153] CDC. Human rabies prevention--United States. Recommendations of the Advisory Committee on Immunization Practices (ACIP). MMWR Recomm Rep, 1999, 48: 1-21.
    [154] Phares TW, Kean RB, Mikheeva T, et al. Regional differences in blood-brain barrier permeability changes and inflammation in the apathogenic clearance of virus from the central nervous system[J]. J Immunol , 2006, 176: 7666-7675.
    [155] Roy A, Phares TW, Koprowski H, et al. Failure to open the blood-brain barrier and deliver immune effectors to central nervous system tissues leads to the lethal outcome of silver-haired bat rabies virus infection[J]. J Virol, 2007, 81: 1110-1118.
    [156] Roy A, Hooper DC. Lethal silver-haired bat rabies virus infection can be prevented by opening the blood-brain barrier[J]. J Virol, 2007, 81: 7993-7998.
    [157] Jackson AC, Fenton MB. Human rabies and bat bites[J]. Lancet, 2001, 357: 1714.
    [158] CDC. Human rabies--Indiana and California. MMWR Morb Mortal Wkly Rep, 2007, 56: 361-365.
    [159] Roy A, Hooper DC. Immune evasion by rabies viruses through the maintenance of blood-brain barrier integrity[J]. J Neurovirol, 2008, 14: 401-411.
    [160] Hosking MP, Lane TE. The role of chemokines during viral infection of the CNS[J]. PLoS Pathog, 2010, 6: e1000937.
    [161] Fenza Cocchi ALD, Alfredo Garzino-Demo, Suresh K.et al. Identification of RANTES, MIP-1α,and MIP-1βas the Major HIV-Suppressive Factors Produced by CD8+ T Cells[J]. Science, 1995, 270: 1811-1815.
    [162] Lane TE, Asensio VC, Yu N, et al. Dynamic regulation of alpha- and beta-chemokine expression in the central nervous system during mouse hepatitis virus-induced demyelinating disease[J]. J Immunol, 1998, 160: 970-978.
    [163] Christensen JE, Simonsen S, Fenger C, et al. Fulminant lymphocytic choriomeningitis virus-induced inflammation of the CNS involves a cytokine-chemokine-cytokine-chemokine cascade[J]. J Immunol, 2009, 182: 1079-1087.
    [164] So EY, Kim BS. Theiler's virus infection induces TLR3-dependent upregulation of TLR2 critical for proinflammatory cytokine production[J]. Glia, 2009, 57: 1216-1226.
    [165] Aravalli RN, Hu S, Rowen TN, et al. Cutting edge: TLR2-mediated proinflammatory cytokine and chemokine production by microglial cells in response to herpes simplex virus[J]. J Immunol, 2005, 175: 4189-4193.
    [166] van Marle G, Henry S, Todoruk T, et al. Human immunodeficiency virus type 1 Nef protein mediates neural cell death: a neurotoxic role for IP-10[J]. Virology, 2004, 329: 302-318.
    [167] Klein RS, Lin E, Zhang B, et al. Neuronal CXCL10 directs CD8+ T-cell recruitment and control of West Nile virus encephalitis[J]. J Virol, 2005, 79: 11457-11466.
    [168] Sasseville VG, Smith MM, Mackay CR,et al. Chemokine expression in simian immunodeficiency virus-induced AIDS encephalitis[J]. Am J Pathol, 1996, 149: 1459-1467.
    [169] Prehaud C, Megret F, Lafage M, et al. Virus infection switches TLR-3-positive human neurons to become strong producers of beta interferon[J]. J Virol, 2005, 79: 12893-12904.
    [170] Wuest T, Austin BA, Uematsu S, et al. Intact TRL 9 and type I interferon signaling pathways are required to augment HSV-1 induced corneal CXCL9 and CXCL10[J]. J Neuroimmunol, 2006, 179: 46-52.
    [171] Ireland DD, Stohlman SA, Hinton DR,et al. Type I interferons are essential in controlling neurotropic coronavirus infection irrespective of functional CD8 T cells [J]. J Virol, 2008, 82: 300-310.
    [172] Scott EP, Branigan PJ, Del Vecchio AM, et al. Chemokine expression during mouse-hepatitis-virus-induced encephalitis: contributions of the spike and background genes[J]. J Neurovirol, 2008, 14: 5-16.
    [173] Zhang B, Chan YK, Lu B, et al. CXCR3 mediates region-specific antiviral T cell trafficking within the central nervous system during West Nile virus encephalitis[J]. J Immunol, 2008, 180: 2641-2649.
    [174] Yan XT, Tumpey TM, Kunkel SL, et al. Role of MIP-2 in neutrophil migration and tissue injury in the herpes simplex virus-1-infected cornea[J]. Invest Ophthalmol Vis Sci, 1998, 39: 1854-1862.
    [175] Hosking MP, Liu L, Ransohoff RM, et al. A protective role for ELR+ chemokines during acute viral encephalomyelitis[J]. PLoS Pathog , 2009, 5: e1000648.
    [176] Kim JV, Kang SS, Dustin ML, et al. Myelomonocytic cell recruitment causes fatal CNS vascular injury during acute viral meningitis[J]. Nature , 2009, 457: 191-195.
    [177] McCandless EE, Zhang B, Diamond MS, et al. CXCR4 antagonism increases T cell trafficking inthe central nervous system and improves survival from West Nile virus encephalitis[J]. Proc Natl Acad Sci U S A , 2008, 105: 11270-11275.
    [178] Asensio VC, Campbell IL. Chemokine gene expression in the brains of mice with lymphocytic choriomeningitis[J]. J Virol, 1997, 71: 7832-7840.
    [179] Liu MT, Chen BP, Oertel P, et al. The T cell chemoattractant IFN-inducible protein 10 is essential in host defense against viral-induced neurologic disease[J]. J Immunol, 2000, 165: 2327-2330.
    [180] Wuest TR, Carr DJ. Dysregulation of CXCR3 signaling due to CXCL10 deficiency impairs the antiviral response to herpes simplex virus 1 infection[J]. J Immunol, 2008, 181: 7985-7993.
    [181] Trifilo MJ, Montalto-Morrison C, Stiles LN, et al. CXC chemokine ligand 10 controls viral infection in the central nervous system: evidence for a role in innate immune response through recruitment and activation of natural killer cells[J]. J Virol, 2004, 78: 585-594.
    [182] Muse M, Kane JA, Carr DJ, et al. Insertion of the CXC chemokine ligand 9 (CXCL9) into the mouse hepatitis virus genome results in protection from viral-induced encephalitis and hepatitis[J]. Virology, 2008, 382: 132-144.
    [183] Glass WG, Lim JK, Cholera R, et al. Chemokine receptor CCR5 promotes leukocyte trafficking to the brain and survival in West Nile virus infection[J]. J Exp Med, 2005, 202: 1087-1098.
    [184] Glass WG, Liu MT, Kuziel WA, et al. Reduced macrophage infiltration and demyelination in mice lacking the chemokine receptor CCR5 following infection with a neurotropic coronavirus[J]. Virology, 2001, 288: 8-17.
    [185] Lim JK, Louie CY, Glaser C, et al. Genetic deficiency of chemokine receptor CCR5 is a strong risk factor for symptomatic West Nile virus infection: a meta-analysis of 4 cohorts in the US epidemic[J]. J Infect Dis, 2008, 197: 262-265.
    [186] Fung-Leung WP, Kundig TM, Zinkernagel RM, et al. Immune response against lymphocytic choriomeningitis virus infection in mice without CD8 expression[J]. J Exp Med, 1991, 174: 1425-1429.
    [187] Christensen JE, de Lemos C, Moos T, et al. CXCL10 is the key ligand for CXCR3 on CD8+ effector T cells involved in immune surveillance of the lymphocytic choriomeningitis virus-infected central nervous system[J]. J Immunol , 2006, 176: 4235-4243.
    [188] Christensen JE, Nansen A, Moos T, et al. Efficient T-cell surveillance of the CNS requires expression of the CXC chemokine receptor 3[J]. J Neurosci, 2004, 24: 4849-4858.
    [189] Hofer MJ, Carter SL, Muller M, et al. Unaltered neurological disease and mortality in CXCR3-deficient mice infected intracranially with lymphocytic choriomeningitis virus-Armstrong[J]. Viral Immunol , 2008, 21: 425-433.
    [190] Liu MT, Keirstead HS, Lane TE. Neutralization of the chemokine CXCL10 reduces inflammatory cell invasion and demyelination and improves neurological function in a viral model of multiple sclerosis[J]. J Immunol , 2001, 167: 4091-4097.
    [191] Glass WG, Hickey MJ, Hardison JL, et al. Antibody targeting of the CC chemokine ligand 5 results in diminished leukocyte infiltration into the central nervous system and reduced neurologic disease in a viral model of multiple sclerosis[J]. J Immunol , 2004, 172: 4018-4025.
    [192] Vergote D, Butler GS, Ooms M, et al. Proteolytic processing of SDF-1alpha reveals a change inreceptor specificity mediating HIV-associated neurodegeneration[J]. Proc Natl Acad Sci U S A, 2006, 103: 19182-19187.
    [193] Kolodziej A, Schulz S, Guyon A, et al. Tonic activation of CXC chemokine receptor 4 in immature granule cells supports neurogenesis in the adult dentate gyru[J]. J Neurosci, 2008, 28: 4488-4500.
    [194] Jackson AC. Rabies pathogenesi[J]. J Neurovirol , 2002, 8: 267-269.
    [195] Martinez L. Global infectious disease surveillance[J]. Int J Infect Dis, 2000, 4: 222-228.
    [196] Abelseth MK. An Attenuated Rabies Vaccine for Domestic Animals Produced in Tissue Culture[J]. Can Vet,1964, J 5: 279-286.
    [197] Fenje P. Propagation of rabies virus in cultures of hamster kidney cells[J]. Can J Microbiol,1960, 6: 479-484.
    [198] Johnson N, McKimmie CS, Mansfield KL, et al. Lyssavirus infection activates interferon gene expression in the brain[J]. J Gen Virol, 2006, 87: 2663-2667.
    [199] Kuang Y, Lackay SN, Zhao L, et al. Role of chemokines in the enhancement of BBB permeability and inflammatory infiltration after rabies virus infection[J]. Virus Res, 2009, 144: 18-26.
    [200] Ubogu EE, Cossoy MB, Ransohoff RM. The expression and function of chemokines involved in CNS inflammation[J]. Trends Pharmacol Sci , 2006, 27: 48-55.
    [201] Glabinski AR, Tani M, Aras S, et al. Regulation and function of central nervous system chemokines[J]. Int J Dev Neurosci, 1995, 13: 153-165.
    [202] Glass WG, Rosenberg HF, Murphy PM. Chemokine regulation of inflammation during acute viral infection[J]. Curr Opin Allergy Clin Immunol , 2003, 3: 467-473.
    [203] Melchjorsen J, Sorensen LN, Paludan SR. Expression and function of chemokines during viral infections: from molecular mechanisms to in vivo function[J]. J Leukoc Biol, 2003, 74: 331-343.
    [204] Nakayama T, Shirane J, Hieshima K, et al. Novel antiviral activity of chemokines[J]. Virology, 2006, 350: 484-492.
    [205] Fife BT, Kennedy KJ, Paniagua MC, et al. CXCL10 (IFN-gamma-inducible protein-10) control of encephalitogenic CD4+ T cell accumulation in the central nervous system during experimental autoimmune encephalomyelitis[J]. J Immunol , 2001, 166: 7617-7624.
    [206] Inoue K, Shoji Y, Kurane I, et al. An improved method for recovering rabies virus from cloned cDNA[J]. J Virol Methods , 2003, 107: 229-236.
    [207] Takayama-Ito M, Inoue K, Shoji Y, et al. A highly attenuated rabies virus HEP-Flury strain reverts to virulent by single amino acid substitution to arginine at position 333 in glycoprotein[J]. Virus Res , 2006, 119: 208-215.
    [208] Koprowski H, Black J. Studies on chick-embryo-adapted rabies virus. VII. Immunological responses of animals to vaccination with high egg passage Flury strain[J]. J Immunol , 1954, 72: 503-510.
    [209] Foley HD, McGettigan JP, Siler CA, et al. A recombinant rabies virus expressing vesicular stomatitis virus glycoprotein fails to protect against rabies virus infection[J]. Proc Natl Acad Sci U S A , 2000, 97: 14680-14685.
    [210] Huffnagle GB, McNeil LK. Dissemination of C. neoformans to the central nervous system: role of chemokines, Th1 immunity and leukocyte recruitment[J]. J Neurovirol, 1999, 5: 76-81.
    [211] Fahey TJ, 3rd, Tracey KJ, Tekamp-Olson P, et al. Macrophage inflammatory protein 1 modulates macrophage function[J]. J Immunol, 1992, 148: 2764-2769.
    [212] Cook DN, Beck MA, Coffman TM, et al. Requirement of MIP-1 alpha for an inflammatory response to viral infection[J]. Science, 1995, 269: 1583-1585.
    [213] Domachowske JB, Bonville CA, Gao JL, et al. The chemokine macrophage-inflammatory protein-1 alpha and its receptor CCR1 control pulmonary inflammation and antiviral host defense in paramyxovirus infection[J]. J Immunol, 2000, 165: 2677-2682.
    [214] Salazar-Mather TP, Orange JS, Biron CA. Early murine cytomegalovirus (MCMV) infection induces liver natural killer (NK) cell inflammation and protection through macrophage inflammatory protein 1alpha (MIP-1alpha)-dependent pathways[J]. J Exp Med , 1998, 187: 1-14.
    [215] Trifilo MJ, Bergmann CC, Kuziel WA, et al. CC chemokine ligand 3 (CCL3) regulates CD8(+)-T-cell effector function and migration following viral infection[J]. J Virol , 2003, 77: 4004-4014.
    [216] Tumpey TM, Cheng H, Cook DN, et al. Absence of macrophage inflammatory protein-1alpha prevents the development of blinding herpes stromal keratitis[J]. J Virol, 1998, 72: 3705-3710.
    [217] Haeberle HA, Kuziel WA, Dieterich HJ, et al. Inducible expression of inflammatory chemokines in respiratory syncytial virus-infected mice: role of MIP-1alpha in lung pathology[J]. J Virol, 2001, 75: 878-890.
    [218] Bahloul C, Ahmed SB, B'Chir B I, et al. Post-exposure therapy in mice against experimental rabies: a single injection of DNA vaccine is as effective as five injections of cell culture-derived vaccine[J]. Vaccine , 2003, 22: 177-184.
    [219] Maurer M, von Stebut E. Macrophage inflammatory protein-1[J]. Int J Biochem Cell Biol, 2004, 36: 1882-1886.
    [220] Rock RB, Gekker G, Hu S, et al. Role of microglia in central nervous system infections[J]. Clin Microbiol Rev , 2004, 17: 942-964, table of contents.
    [221] Frazatti-Gallina NM, Mourao-Fuches RM, Paoli RL, et al. Vero-cell rabies vaccine produced using serum-free medium[J]. Vaccine , 2004, 23: 511-517.
    [222] Meltzer MI. Assessing the costs and benefits of an oral vaccine for raccoon rabies: a possible model[J]. Emerg Infect Dis, 1996, 2: 343-349.
    [223] Shim E, Hampson K, Cleaveland S, et al. Evaluating the cost-effectiveness of rabies post-exposure prophylaxis: a case study in Tanzania[J]. Vaccine , 2009, 27: 7167-7172.
    [224] Blancou J, Artois M, Brochier B, et al. [Safety and efficacy of an antirabies vaccine consisting of recombinant vaccinia-rabies virus administered orally to the fox, dog and cat] [J]. Ann Rech Vet, 1989, 20: 195-204.
    [225] Brochier B, Kieny MP, Costy F, et al. Large-scale eradication of rabies using recombinant vaccinia-rabies vaccine[J]. Nature, 1991, 354: 520-522.
    [226] Hanlon CA, Niezgoda M, Hamir AN, et al. First North American field release of a vaccinia-rabies glycoprotein recombinant virus[J]. J Wildl Dis, 1998, 34: 228-239.
    [227] Flamand A, Coulon P, Lafay F. Avirulent mutants of rabies virus and their use as live vaccine[J].Trends Microbiol , 1993, 1: 317-320.
    [228] CDC. Human vaccinia infection after contact with a raccoon rabies vaccine bait - Pennsylvania[J]. 2009. MMWR Morb Mortal Wkly Rep 58: 1204-1207.
    [229] Rupprecht CE, Blass L, Smith K, et al. Human infection due to recombinant vaccinia-rabies glycoprotein virus[J]. N Engl J Med, 2001, 345: 582-586.
    [230] Zhao L, Toriumi H, Wang H, et al. Expression of MIP-1{alpha} (CCL3) by a Recombinant Rabies Virus Enhances Its Immunogenicity by Inducing Innate Immunity and Recruiting Dendritic Cells and B Cells[J]. J Virol , 2010, 84: 9642-9648.
    [231] Becker Y. Immunological and regulatory functions of uninfected and virus infected immature and mature subtypes of dendritic cells--a review[J]. Virus Genes, 2003, 26: 119-130.
    [232] Brilot F, Strowig T, Munz C. NK cells interactions with dendritic cells shape innate and adaptive immunity[J]. Front Biosci , 2008, 13: 6443-6454.
    [233] Rasalingam P, Rossiter JP, Mebatsion T, et al. Comparative pathogenesis of the SAD-L16 strain of rabies virus and a mutant modifying the dynein light chain binding site of the rabies virus phosphoprotein in young mice[J]. Virus Res, 2005, 111: 55-60.
    [234] Smith JS, Yager, P.A., Baer, G.M. A rapid fluorescence focus inhibition test (RFFIT) for determining rabies virus-neutralisng antibody. In: Meslin, F-X, Kaplan, M,M, Koprowski, H (Eds), Laboratory Techniques in Rabies, 4th ed pp, 1996, 181-192.
    [235] Reed LJ, and H. Muench. A simple method of estimating fifty percent end points[J]. Am J Hyg, 1938: 493-497.
    [236] Barouch DH, McKay PF, Sumida SM, et al. Plasmid chemokines and colony-stimulating factors enhance the immunogenicity of DNA priming-viral vector boosting human immunodeficiency virus type 1 vaccines[J]. J Virol , 2003, 77: 8729-8735.
    [237] McKay PF, Barouch DH, Santra S, et al. Recruitment of different subsets of antigen-presenting cells selectively modulates DNA vaccine-elicited CD4+ and CD8+ T lymphocyte responses[J]. Eur J Immunol , 2004, 34: 1011-1020.
    [238] Geissmann F, Jung S, Littman DR. Blood monocytes consist of two principal subsets with distinct migratory properties[J]. Immunity, 2003, 19: 71-82.
    [239] Kurts C. CD11c: not merely a murine DC marker, but also a useful vaccination target[J]. Eur J Immunol , 2008, 38: 2072-2075.
    [240] Yong M, Mitchell D, Caudron A, et al. Expression of maturation markers on murine dendritic cells in response to group A streptococcal lipopeptide vaccines[J]. Vaccine, 2009, 27: 3313-3318.
    [241] Clark EA . Regulation of B lymphocytes by dendritic cells[J]. J Exp Med, 1997, 185: 801-803.
    [242] Dubois B, Bridon JM, Fayette J, et al. Dendritic cells directly modulate B cell growth and differentiation[J]. J Leukoc Biol, 1999, 66: 224-230.
    [243] Wykes M, Pombo A, Jenkins C, et al. Dendritic cells interact directly with naive B lymphocytes to transfer antigen and initiate class switching in a primary T-dependent response[J]. J Immunol , 1998, 161: 1313-1319.
    [244] Wykes M, MacPherson G. Dendritic cell-B-cell interaction: dendritic cells provide B cells with CD40-independent proliferation signals and CD40-dependent survival signals[J]. Immunology,2000, 100: 1-3.
    [245] Kozmik Z, Wang S, Dorfler P, et al. The promoter of the CD19 gene is a target for the B-cell-specific transcription factor BSAP[J]. Mol Cell Biol, 1992, 12: 2662-2672.
    [246] Castigli E, Young F, Carossino AM, et al. CD40 expression and function in murine B cell ontogeny[J]. Int Immunol, 1996, 8: 405-411.
    [247] Corcione A, Tortolina G, Bonecchi R, et al. Chemotaxis of human tonsil B lymphocytes to CC chemokine receptor (CCR) 1, CCR2 and CCR4 ligands is restricted to non-germinal center cells[J]. Int Immunol, 2002, 14: 883-892.
    [248] Schall TJ, Bacon K, Camp RD, et al. Human macrophage inflammatory protein alpha (MIP-1 alpha) and MIP-1 beta chemokines attract distinct populations of lymphocytes[J]. J Exp Med, 1993, 177: 1821-1826.
    [249] Hamilton JA, Anderson GP . GM-CSF Biology[J]. Growth Factors , 2004, 22: 225-231.
    [250] Metcalf D. Hematopoietic cytokines[J]. Blood, 2008, 111: 485-491.
    [251] Iellem A, Mariani M, Lang R, et al. Unique chemotactic response profile and specific expression of chemokine receptors CCR4 and CCR8 by CD4(+)CD25(+) regulatory T cells[J]. J Exp Med, 2001, 194: 847-853.
    [252] Imai T, Nagira M, Takagi S, et al. Selective recruitment of CCR4-bearing Th2 cells toward antigen-presenting cells by the CC chemokines thymus and activation-regulated chemokine and macrophage-derived chemokine[J]. Int Immunol, 1999, 11: 81-88.
    [253] Yoshie O, Imai T, Nomiyama H. Chemokines in immunity[J]. Adv Immunol , 2001, 78: 57-110.
    [254] Chantry D, Romagnani P, Raport CJ, et al. Macrophage-derived chemokine is localized to thymic medullary epithelial cells and is a chemoattractant for CD3(+), CD4(+), CD8(low) thymocytes[J]. Blood, 1999, 94: 1890-1898.
    [255] Godiska R, Chantry D, Raport CJ, et al. Human macrophage-derived chemokine (MDC), a novel chemoattractant for monocytes, monocyte-derived dendritic cells, and natural killer cells[J]. J Exp Med, 1997, 185: 1595-1604.
    [256] Wu M, Fang H, Hwang ST. Cutting edge: CCR4 mediates antigen-primed T cell binding to activated dendritic cells[J]. J Immunol , 2001, 167: 4791-4795.
    [257] Schaniel C, Pardali E, Sallusto F, et al. Activated murine B lymphocytes and dendritic cells produce a novel CC chemokine which acts selectively on activated T cells[J]. J Exp Med, 1998, 188: 451-463.
    [258] Disis ML, Bernhard H, Shiota FM, et al. Granulocyte-macrophage colony-stimulating factor: an effective adjuvant for protein and peptide-based vaccines[J]. Blood, 1996, 88: 202-210.
    [259] Haddad D, Ramprakash J, Sedegah M, et al. Plasmid vaccine expressing granulocyte-macrophage colony-stimulating factor attracts infiltrates including immature dendritic cells into injected muscles[J]. J Immunol, 2000, 165: 3772-3781.
    [260] Morrissey PJ, Bressler L, Park LS, et al. Granulocyte-macrophage colony-stimulating factor augments the primary antibody response by enhancing the function of antigen-presenting cells[J].J Immunol ,1987,139: 1113-1119.
    [261] Tarr PE. Granulocyte-macrophage colony-stimulating factor and the immune system[J]. MedOncol, 1996, 13: 133-140.
    [262] Biragyn A, Belyakov IM, Chow YH, et al. DNA vaccines encoding human immunodeficiency virus-1 glycoprotein 120 fusions with proinflammatory chemoattractants induce systemic and mucosal immune responses[J]. Blood , 2002, 100: 1153-1159.
    [263] WHO Expert Committee on Rabies. World Health Organ Tech Rep Ser, 1999, 824: 1-84.
    [264] Lackay SN, Kuang Y, Fu ZF. Rabies in small animals. Vet Clin North Am Small Anim Pract, 2008, 38: 851-861.
    [265] Nigg AJ, Walker PL. Overview, prevention, and treatment of rabies[J]. Pharmacotherapy, 2009, 29: 1182-1195.
    [266] Bakker AB, Python C, Kissling CJ, et al. First administration to humans of a monoclonal antibody cocktail against rabies virus: safety, tolerability, and neutralizing activity[J]. Vaccine, 2008, 26: 5922-5927.
    [267] Bechmann I, Galea I, Perry VH. What is the blood-brain barrier (not) [J]? Trends Immunol, 2007, 28: 5-11.
    [268] Rupprecht CE, Hanlon CA, Hemachudha T. Rabies re-examined[J]. Lancet Infect Dis, 2002, 2: 327-343.
    [269] Pachter JS, de Vries HE, Fabry Z. The blood-brain barrier and its role in immune privilege in the central nervous system[J]. J Neuropathol Exp Neurol , 2003, 62: 593-604.
    [270] Kean RB, Spitsin SV, Mikheeva T, et al. The peroxynitrite scavenger uric acid prevents inflammatory cell invasion into the central nervous system in experimental allergic encephalomyelitis through maintenance of blood-central nervous system barrier integrity[J].J Immunol, 2000,165: 6511-6518.
    [271] Hooper DC, Kean RB, Scott GS, et al. The central nervous system inflammatory response to neurotropic virus infection is peroxynitrite dependent[J]. J Immunol, 2001, 167: 3470-3477.
    [272] Phares TW, Fabis MJ, Brimer CM, et al. A peroxynitrite-dependent pathway is responsible for blood-brain barrier permeability changes during a central nervous system inflammatory response: TNF-alpha is neither necessary nor sufficient[J]. J Immunol, 2007, 178: 7334-7343.
    [273] Stanimirovic D, Satoh K. Inflammatory mediators of cerebral endothelium: a role in ischemic brain inflammation[J]. Brain Pathol , 2000, 10: 113-126.
    [274] Petty MA, Lo EH. Junctional complexes of the blood-brain barrier: permeability changes in neuroinflammation[J]. Prog Neurobiol , 2002, 68: 311-323.
    [275] Merrill JE, Murphy SP. Inflammatory events at the blood brain barrier: regulation of adhesion molecules, cytokines, and chemokines by reactive nitrogen and oxygen species[J]. Brain Behav Immun, 1997, 11: 245-263.
    [276] Stamatovic SM, Shakui P, Keep RF, et al. Monocyte chemoattractant protein-1 regulation of blood-brain barrier permeability[J]. J Cereb Blood Flow Metab, 2005, 25: 593-606.
    [277] Luster AD. Chemokines--chemotactic cytokines that mediate inflammation[J]. N Engl J Med, 1998, 338: 436-445.
    [278] Baggiolini M. Chemokines and leukocyte traffic[J]. Nature, 1998, 392: 565-568.
    [279] Hooper DC, Phares TW, Fabis MJ, et al. The production of antibody by invading B cells isrequired for the clearance of rabies virus from the central nervous system[J]. PLoS Negl Trop Dis, 2009, 3: e535.
    [280] Barker CF, Billingham RE. Immunologically privileged sites. Adv Immunol,1977, 25: 1-54.
    [281] Wen Y, Wang H, Wu H, Yang F, Tripp RA, et al. Rabies virus expressing dendritic cell-activating molecules enhances the innate and adaptive immune response to vaccination. J Virol. 2010.
    [282] Hemachudha T, Phanuphak P, Sriwanthana B, Manutsathit S, Phanthumchinda K, et al. Immunologic study of human encephalitic and paralytic rabies. Preliminary report of 16 patients. Am J Med, 1988, 84: 673-677.

© 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