首页 | 本学科首页   官方微博 | 高级检索  
相似文献
 共查询到20条相似文献,搜索用时 15 毫秒
1.
Studies conducted some 50 years ago showed that serial intracerebral passage of dengue viruses in mice selected for neurovirulent mutants that also exhibited significant attenuation for humans. We investigated the genetic basis of mouse neurovirulence of dengue virus because it might be directly or indirectly associated with attenuation for humans. Analysis of the sequence in the C-PreM-E-NS1 region of the parental dengue type 2 virus (DEN2) New Guinea C (NGC) strain and its mouse-adapted, neurovirulent mutant revealed that 10 nucleotide changes occurred during serial passage in mice. Seven of these changes resulted in amino acid substitutions, i.e., Leu55-Phe and Arg57-Lys in PreM, Glu71-Asp, Glu126-Lys, Phe402-Ile, and Thr454-Ile in E, and Arg105-Gln in NS1. The sequence of C was fully conserved between the parental and mutant DEN2. We constructed intertypic chimeric dengue viruses that contained the PreM-E genes or only the NS1 gene of neurovirulent DEN2 NGC substituting for the corresponding genes of DEN4. The DEN2 (PreM-E)/DEN4 chimera was neurovirulent for mice, whereas DEN2 (NS1)/DEN4 was not. The mutations present in the neurovirulent DEN2 PreM-E genes were then substituted singly or in combination into the sequence of the nonneurovirulent, parental DEN2. Intracerebral titration of the various mutant chimeras so produced identified two amino acid changes, namely, Glu71-Asp and Glu126-Lys, in DEN2 E as being responsible for mouse neurovirulence. The conservative amino acid change of Glu71-Asp probably had a minor effect, if any. The Glu126-Lys substitution in DEN2 E, representing a change from a negatively charged amino acid to a positively charged amino acid, most likely plays an important role in conferring mouse neurovirulence.  相似文献   

2.
Small plaque mutants of Western equine encephalitis virus were obtained from the surviving fractions of wild-type virus which was irradiated with gamma rays. The frequency with which small plaque mutants appeared in the surviving fraction increased with the radiation dose. These mutants were not more resistant to radiation than wild-type virus. The growth rate of a mutant, S127, was lower than that of wild-type. Clonally purified mutant virions presented two peaks in a velocity sedimentation profile; peak 1 corresponded to the peak of wild type and peak 2 moved faster than peak 1. Virions of both peaks were infectious and consistently formed small plaques in chicken embryo cells. Virions reisolated from either peak and grown in chicken embryo cells also revealed two peaks in sedimentation analysis. In the electron microscope examination peak 2 proved to consist of giant form particles, each of which contained more than one nucleoid surrounded with a common envelope. Despite this remarkable morphological difference, densities of the wild-type and S127 mutant virions were similar in cesium chloride gradients. The RNAs and proteins of mutant virions could not be distinguished from those of wild type on the basis of size or charge.  相似文献   

3.
我国分离的XJ-90260病毒鉴定为西方马脑炎病毒   总被引:7,自引:0,他引:7  
XJ-90260病毒是从新疆乌苏县境内采集的赫坎按蚊中分离到的一株病毒,病毒的鉴定结果显示:XJ-90260病毒可引起BHK-21细胞病变,表现为圆缩,脱落;可引起Vero细胞病变,表现为圆缩,破碎,脱落;可以在C6/36细胞中增殖,但不引起细胞病变。对3日龄小白鼠2-3天致死,对3周龄小白鼠3-4天致死。该病毒株对酸、乙醚敏感,抵抗5-氟脱氧尿苷。病毒与甲病毒组特异性免疫腹水起反应,与乙型脑炎病毒及布尼亚病毒组特异性免疫腹水不反应。进一步的分子生物学鉴定表明,该毒株基因组3′非编码区(ntranslated region,UTR)核苷酸序列具有典型的西方马脑炎病毒特征,与标准西方马脑炎病毒的首次报导,有重要的流行病学意义。我国9省区,886份血清的流行病学调查显示,该病毒抗体阳性血清24份,阳性率为2.71%。其中新疆(8/157),河南(6/76)、甘肃(5/94)三省区抗体阳性数较多,占总阳性数的79.2%(19/24)。  相似文献   

4.
During 1971, an epizootic of Venezuelan equine encephalitis (VEE) reached the United States. Laboratory tests were performed on a large number of sick, healthy, unvaccinated, and vaccinated horses. Neutralization (N) tests in cell cultures revealed that 153 of 193 (79.3%) equines outside the state of Texas and 175 of 204 (85.8%) within Texas (82.6% overall) had detectable N antibody to VEE virus a week or more after vaccination. Twenty-six of 40 (65%) non-Texas equines and 18 of 29 (62%) Texas equines which had no detectable antibody against VEE virus a week or more after vaccination had N antibody against Eastern equine encephalitis (EEE) or Western equine encephalitis (WEE) virus or both, whereas only 50 of 153 (32.7%) non-Texas equines and 82 of 175 (46.9%) Texas equines with demonstrable N antibody against VEE also had N antibody against EEE and/or WEE virus. In vaccinated equines, significant negative correlations were found between the occurrence of antibody to VEE and antibody to EEE and/or WEE virus. These findings support the hypothesis that pre-existing antibody to EEE and/or WEE virus may modify or interfere with infection by VEE virus. The epizoologic significance of this possibility is discussed briefly.  相似文献   

5.
6.
The within-host diversity of virus populations can be drastically limited during between-host transmission, with primary infection of hosts representing a major constraint to diversity maintenance. However, there is an extreme paucity of quantitative data on the demographic changes experienced by virus populations during primary infection. Here, the multiplicity of cellular infection (MOI) and population bottlenecks were quantified during primary mosquito infection by Venezuelan equine encephalitis virus, an arbovirus causing neurological disease in humans and equids.  相似文献   

7.
Two plaque mutants were isolated from tissue cultures infected persistently with Western equine encephalitis virus. A large plaque mutant proved to be markedly avirulent for mice.  相似文献   

8.
Western equine encephalitis virus (WEEV; Togaviridae, Alphavirus) is an enveloped RNA virus that is typically transmitted to vertebrate hosts by infected mosquitoes. WEEV is an important cause of viral encephalitis in humans and horses in the Americas, and infection results in a range of disease, from mild flu-like illnesses to encephalitis, coma, and death. In addition to spreading via mosquito vectors, human WEEV infections can potentially occur directly via aerosol transmission. Due to its aerosol infectivity and virulence, WEEV is thus classified as a biological safety level 3 (BSL-3) agent. Because of its highly infectious nature and containment requirements, it has not been possible to investigate WEEV''s structure or assembly mechanism using standard structural biology techniques. Thus, to image WEEV and other BSL-3 agents, we have constructed a first-of-its-kind BSL-3 cryoelectron microscopy (cryoEM) containment facility. cryoEM images of WEEV were used to determine the first three-dimensional structure of this important human pathogen. The overall organization of WEEV is similar to those of other alphaviruses, consistent with the high sequence similarity among alphavirus structural proteins. Surprisingly, the nucleocapsid of WEEV, a New World virus, is more similar to the Old World alphavirus Sindbis virus than to other New World alphaviruses.The alphaviruses comprise a genus of single-stranded, plus-sense, enveloped RNA viruses that, together with rubella virus, comprise the family Togaviridae. The current classification of the genus Alphavirus includes 29 different species, with multiple subtypes and/or varieties represented within some species (30). These species can be grouped into 8 different complexes based on antigenic and/or genetic similarities (20). Most viruses from the New World are found in the Eastern, Venezuelan, and Western equine encephalitis (EEE, VEE, and WEE, respectively) complexes and cause encephalitis in humans and a variety of domesticated animals. Old World alphaviruses, on the other hand, typically cause only an arthralgia and rash syndrome that is rarely life threatening (5, 24). Among the New World alphaviruses, EEE, VEE, and WEE viruses (EEEV, VEEV, and WEEV, respectively) are potential biological weapons as well as naturally emerging pathogens and are therefore included on the category B Priority Pathogens list of the National Institute of Allergy and Infectious Diseases of the National Institutes of Health (http://www.niaid.nih.gov/topics/biodefenserelated/biodefense/research/pages/cata.aspx).Alphaviruses replicate in the cytoplasm of infected cells after entry via receptor-mediated endocytosis (8). Following internalization, fusion of the viral envelope with the endocytic membrane is mediated by a low-pH-induced conformational change that exposes a fusion peptide found in the E1 envelope glycoprotein. The nucleocapsid then disassembles upon interactions with ribosomes, and an open reading frame (ORF) found in the 5′ two-thirds of the genome is translated. The resultant polyprotein is cleaved into 4 nonstructural proteins (nsP1 to -4) that mediate viral RNA replication, RNA capping, and polyprotein processing (Fig. (Fig.1).1). The structural proteins, including the two envelope glycoproteins E2 and E1 as well as the capsid protein, are encoded in a second ORF that is translated from a subgenomic message often referred to as 26S RNA. Following auto-cleavage of the capsid protein in the cytoplasm, the remaining polyprotein is inserted into the endoplasmic reticulum, where it is cleaved by host cell proteases and then processed through the secretory pathway, where the glycosylation of E2 and E1 occurs. Virion maturation occurs after E2/E1 heterodimers are inserted into the plasma membrane and 240 copies of the capsid protein interact with one copy of the genomic RNA to form nucleocapsids. These nucleocapsids then interact with a cytoplasmic domain of the E2 protein to initiate budding. The mature virion thus includes 240 copies of the capsid protein and 240 E2/E1 heterodimers arranged as trimeric spikes on the surface of the virus (8).Open in a separate windowFIG. 1.Diagram of the alphavirus genome, showing the 5′ cap, 5′ untranslated region, nonstructural polyprotein open reading frame, and major functions of the individual proteins, subgenomic promoter, structural polyprotein open reading frame, 3′ untranslated region, and poly(A) tail.The structures of several different alphaviruses, including Sindbis virus (SINV) (13), Ross River virus (RRV) (3, 35), Semliki Forest virus (SFV), (11), and VEEV (16), have been solved to subnanometer resolution using cryoelectron microscopy (cryoEM), and the X-ray crystallographic structure of the E1 protein from Semliki Forest virus has been determined to atomic resolution (9). The alphaviruses are ca. 700 Å in diameter, with 80 trimeric spikes on their surfaces. By fitting the E1 crystal structure into cryoEM reconstruction maps of whole viruses, the orientations of both envelope proteins within the spikes have been estimated (36). The E1 and E2 proteins are similar in shape, and the E2 proteins extend to the tips of the spikes, where most glycosylation and antibody-binding sites have been mapped (13). The underlying T=4 icosahedral capsid is constructed from regularly ordered capsomers arranged as hexons and pentons. These pentons and hexons consist of capsid protein monomers that apparently represent only the C-terminal half of the protein. Crystal structures of alphavirus capsid proteins also indicate that only the C terminus, including the protease domain, is ordered (25). cryoEM reconstructions of VEEV nucleocapsids isolated from virions have a less ordered structure, with density redistributed from the 3-fold to the 5-fold axis, suggesting that the envelope and/or the envelope glycoproteins constrain and stabilize the nucleocapsid in a compressed structure (15). Additionally, the VEEV nucleocapsids within viruses differ from those of Old World alphaviruses, with a counterclockwise rotation of the pentameric and hexameric capsomers in VEEV (16). Similar differences were observed in the capsid of Aura virus (AURAV), another New World alphavirus (34).In addition to being an important human and equine pathogen, WEEV is one of three alphaviruses that descended from a recombinant ancestor (6, 31). This ancestor derived its nonstructural and capsid protein genes from an ancestral EEEV strain, whereas its envelope glycoprotein genes were provided from an ancestral SINV. The recombination event was apparently followed by compensatory mutations in the cytoplasmic domain of the E2 protein that restored efficient interactions with the EEEV-like capsid protein (6). If this interpretation of the WEEV ancestral recombination event is correct, its nucleocapsids, constructed from capsid proteins derived from the New World EEEV ancestor, would be expected be more similar to those of the New World VEEV than to those of the Old World SINV, RRV, and SFV. To test this hypothesis and to investigate other structural features of interest related to its recombinant history and pathogenicity, we determined the structure of WEEV to a 13-Å resolution using cryoEM image reconstruction.  相似文献   

9.
In mice infected with the Venezuelan equine encephalomyelitis (VEE) virus and exposed to high intensity light (2500 lux) with a 12 h light: 12 h dark photoperiod, a significant increase in the levels of melatonin in the olfactory bulb was observed. The significance of these findings deserves further studies to understand the mechanisms involved in this effect since the olfactory bulbs have been proposed as first portal for VEE virus entry into the CNS. The increase in melatonin content could represent one of the mechanisms of defense against the viral attack.  相似文献   

10.
Many viruses have been implicated in utilizing or modulating the Ubiquitin Proteasome System (UPS) to enhance viral multiplication and/or to sustain a persistent infection. The mosquito-borne Venezuelan equine encephalitis virus (VEEV) belongs to the Togaviridae family and is an important biodefense pathogen and select agent. There are currently no approved vaccines or therapies for VEEV infections; therefore, it is imperative to identify novel targets for therapeutic development. We hypothesized that a functional UPS is required for efficient VEEV multiplication. We have shown that at non-toxic concentrations Bortezomib, a FDA-approved inhibitor of the proteasome, proved to be a potent inhibitor of VEEV multiplication in the human astrocytoma cell line U87MG. Bortezomib inhibited the virulent Trinidad donkey (TrD) strain and the attenuated TC-83 strain of VEEV. Additional studies with virulent strains of Eastern equine encephalitis virus (EEEV) and Western equine encephalitis virus (WEEV) demonstrated that Bortezomib is a broad spectrum inhibitor of the New World alphaviruses. Time-of-addition assays showed that Bortezomib was an effective inhibitor of viral multiplication even when the drug was introduced many hours post exposure to the virus. Mass spectrometry analyses indicated that the VEEV capsid protein is ubiquitinated in infected cells, which was validated by confocal microscopy and immunoprecipitation assays. Subsequent studies revealed that capsid is ubiquitinated on K48 during early stages of infection which was affected by Bortezomib treatment. This study will aid future investigations in identifying host proteins as potential broad spectrum therapeutic targets for treating alphavirus infections.  相似文献   

11.
Phospholipid analyses of Venezuelan equine encephalitis virus showed that virus propagated in L-cell monolayers had a higher sphingomyelin content and a lower phosphatidylcholine content than virus grown in chick fibroblast monolayers. Virus of L-cell origin also was found to possess greater thermal stability than virus derived from the chick fibroblast cell.  相似文献   

12.
13.
The problem of evaluating the epidemic potential of western equine encephalitis in the northeastern United States is presented and possible reasons are discussed for the present lack of human and horse cases of this disease even though increased numbers of isolations of the virus have been obtained in the East during recent years. Epidemiologic factors of vector bionomics and virus strain variations are considered. It is concluded that while this virus strain can no longer be regarded as uncommon in the Northeast, the evidence indicates there is little potential for epidemic expression of this agent in the human and horse population. This appears to be due to differences in the bionomics of the mosquito Culiseta melanura, which serves as the primary enzootic vector in the northeastern United States and in the bionomics of Culex tarsalis that is the vector in the western region of the United States. Other limiting factors in the epidemic potential may be variations between virus strains located in the East and West.  相似文献   

14.
Mouse Potency Assay for Western Equine Encephalomyelitis Vaccines   总被引:2,自引:0,他引:2       下载免费PDF全文
A potency assay for Western equine encephalomyelitis vaccine was developed which utilized mice as the test animal instead of guinea pigs or hamsters. By immunizing several groups of mice with dilutions of the vaccine and challenging them intracerebrally with virulent virus, it was possible to determine mathematically a dose of vaccine capable of protecting 50% of the animals (ED(50)). When log dilutions of virulent virus were used to challenge mice which were immunized with dilutions of the vaccine, there was no difference among the ED(50) values for the dilutions of challenge virus. In a direct comparison of ED(50) values determined from the immunization of mice and those determined from the immunization of guinea pigs, there were no differences in the rankings of the vaccines.  相似文献   

15.
16.
The host innate immune response provides a critical first line of defense against invading pathogens, inducing an antiviral state to impede the spread of infection. While numerous studies have documented antiviral responses within actively infected tissues, few have described the earliest innate response induced systemically by infection. Here, utilizing Venezuelan equine encephalitis virus (VEE) replicon particles (VRP) to limit infection to the initially infected cells in vivo, a rapid activation of the antiviral response was demonstrated not only within the murine draining lymph node, where replication was confined, but also within distal tissues. In the liver and brain, expression of interferon-stimulated genes was detected by 1 to 3 h following VRP footpad inoculation, reaching peak expression of >100-fold over that in mock-infected animals. Moreover, mice receiving a VRP footpad inoculation 6, 12, or 24 h prior to an otherwise lethal VEE footpad challenge were completely protected from death, including a drastic reduction in challenge virus titers. VRP pretreatment also provided protection from intranasal VEE challenge and extended the average survival time following intracranial challenge. Signaling through the interferon receptor was necessary for antiviral gene induction and protection from VEE challenge. However, VRP pretreatment failed to protect mice from a heterologous, lethal challenge with vesicular stomatitis virus, yet conferred protection following challenge with influenza virus. Collectively, these results document a rapid modulation of the host innate response within hours of infection, capable of rapidly alerting the entire animal to pathogen invasion and leading to protection from viral disease.Venezuelan equine encephalitis virus (VEE) is an arthropod-borne, single-stranded, message-sense RNA virus belonging to the Alphavirus genus and Togaviridae family. Associated with periodic epidemics and equine epizootics in the Western Hemisphere, VEE also serves as a leading model for the study of alphavirus pathogenesis in vivo. In the murine model, which closely mimics infection of horses in nature, VEE causes a two-phase disease: an initial, acute lymphotropic phase characterized by a high serum viremia, followed by invasion of the central nervous system during a neurotropic phase that leads to fatal encephalitis (22, 27). Using the infectious molecular clone of VEE and an extensive panel of mutants blocked at various stages of infection, the course of infection and disease in the mouse model has been well characterized (3, 14, 15, 17, 27).Studies examining the molecular aspects of VEE pathogenesis have underscored the critical role of virus genetics and the subsequent host response in dictating the course and outcome of infection (6, 12, 23, 27, 35, 60, 64, 73). However, many details of the earliest host-pathogen interactions during VEE infection remain largely unknown. A tool paramount to studying early events in infection are VEE replicon particles (VRP). VRP are propagation-defective particles that undergo only one round of infection, as the structural genes which normally drive the assembly of progeny virions are deleted from the replicon genome (51). Infection of cells by VRP results in amplification of replicon viral RNA, but there is no packaging of new progeny and thus no further spread to other cells. As such, VRP infection is limited to the first round of targeted cells, allowing examination of the earliest interactions between virus and host.VRP infection of mice facilitated the identification of the draining lymph node (DLN) as the initial site of VEE viral amplification in vivo (44). Following footpad inoculation of mice with VRP, resident dendritic cells in the skin serve as the cellular target for infection. These infected dendritic cells then rapidly migrate from the site of inoculation in the skin to the local DLN (44). In the case of VRP infection, while no new viral progeny are packaged or released, the replicon genome continues to be replicated within these initially infected skin dendritic cells that have migrated and reside in the DLN. However, during infection with VEE virus, new viral progeny are eventually released into the DLN environment and infection spreads to adjacent cells.Based on these observations, we hypothesize that the earliest host-pathogen interactions within the DLN set the stage for the specific course of events that define VEE-induced pathogenesis. The innate immune response, including interferon (IFN) signaling, has been extensively documented as a critical component of controlling viral infection and spread (45, 47, 62, 66). In fact, utilizing a VRP-based mRNP-tagging system in vivo, we recently reported the robust activation of the host innate antiviral response directly within the infected cells of the DLN, as well as in surrounding uninfected bystander cells, at early times postinoculation (39). A consequence of this early, robust innate immune response at the initial site of replication is likely a contemporaneous induction of an antiviral state in tissues distal to the primary infection.We postulated that if early viral replication in the DLN induces the production of soluble immune mediators, such as IFN-α/β, then the induction of innate immune responses may be rapidly transmitted downstream from this primary site to distal tissues. Utilizing VRP to limit viral spread, we examined the host antiviral response within the DLN and tissues remote from the site of replication at early times following infection. In the liver and brain, the robust expression of a panel of IFN-stimulated genes, a hallmark of the antiviral state, was detected by 1 to 3 h following VRP footpad inoculation and peaked at expression levels >100-fold over mock animals. These results suggest that the early innate response to VRP infection is capable of rapidly inducing a systemically active antiviral state within the entire infected animal. Moreover, we found that mice pretreated by footpad inoculation with VRP for 6, 12, or 24 h were protected from an otherwise lethal VEE footpad or intranasal challenge, and the average survival time of mice challenged intracranially with VEE was significantly extended.Protection from VEE infection has typically been associated with the presence of neutralizing antibody (11, 24, 29, 49, 55). However, nonspecific protection against VEE has been suggested, including the involvement of the innate immune response (10, 26, 28, 33, 61, 73). In one instance, mice “vaccinated” with an attenuated clone of VEE were protected against lethal VEE challenge administered just 24 h after vaccination (26). In separate studies, the complete attenuation of a VEE mutant harboring a single noncoding nucleotide change was attributed to a heightened sensitivity of the virus to the host antiviral state (73). Additionally, mice with severe combined immunodeficiency survive longer than immunocompetent mice (9 days as opposed to 6 days) following infection with virulent VEE (12). These findings firmly indicate that the nonspecific host response to VEE is a critical component of controlling the earliest stages of infection.While IFN and the IFN-induced antiviral state are undoubtedly key mediators of the initial response to VRP infection in vivo, they may not solely be responsible for a rapidly induced protective state. In the challenge model presented here, VRP pretreatment was unable to protect mice from death following heterologous challenge with another IFN-sensitive virus, vesicular stomatitis virus (VSV). However, VRP pretreatment successfully protected mice from lethal challenge with influenza virus. Collectively, our results raise at least three important implications. First, the innate host response is rapidly mobilized following infection with VRP/VEE, at areas both proximal and distal to the site of active replication. Second, there exist components of the innate immune response to VEE that remain uncharacterized. Third, viruses are specifically and differentially sensitive to unique innate immune response profiles. These data provide new insight into the rapid mobilization of the host response to viral infection and present an effective pretreatment/challenge model to further investigate specific components of the innate response critical to protection against infectious pathogens.  相似文献   

17.
18.
19.
Venezuelan equine encephalitis (VEE) virus was purified and concentrated by chromatography of tissue culture supernatant fluids on diethylaminoethyl-cellulose columns. Stepwise gradient elution studies indicated a broad elution pattern for the virus, with recovery occurring from 0.05 to 0.7 m NaCl. Optical density, infectivity, hemagglutination (HA), and complement fixation (CF) assays indicated that complete recovery of input virus in highly purified form was possible. Single-step elution with 0.7 m tris(hydroxymethyl)aminomethane-succinate-salt buffer resulted in a virus volume decrease of 85% with a concomitant increase in infectivity and antigenicity. Recoveries consistently equaled or exceeded 100% of the input preparations. Additional purification of column-recovered virus was obtained by sedimentation of pooled virus eluates on 50% sucrose cushions. Exposure of borate saline and 0.5% histidine suspensions of purified VEE virus preparations to 6 x 10(6) r of gamma radiation resulted in a loss of infectivity for tissue culture and a loss of lethality for weanling and suckling mice. Inactivation was an exponential function of the dosage. In contrast to infectivity, antigencity (HA and CF) of both saline and histidine preparations was retained after irradiation with doses as high as 6 x 10(6) r. Purified and irradiated VEE virus preparations have been successfully used for routine serological tests and are being evaluated as vaccines.  相似文献   

20.
In contrast to Eastern equine encephalitis virus, the replication of Venezuelan equine encephalitis (VEE) virus was strongly inhibited at 44 C in chick embryo cells. The inhibited steps were analyzed by shifting the incubating temperatures up or down, and by determining during the shifts the rate and extent of infectious ribonucleic acid (RNA) synthesis, intact virus synthesis, and formation of complement-fixing antigen or of antigen detectable by a direct fluorescent-antibody technique. The inhibition appeared to be due to two temperature-sensitive steps involved in the synthesis of VEE virus in chick embryo cells. The first step of inhibition at 44 C occurred early in virus replication and could be completely reversed simply by transferring cultures to 37 C. The inhibition appeared to take place at some point between the time when the virus entered the cell and was uncoated and the beginning of viral RNA synthesis. The second temperature-sensitive step in VEE virus synthesis was irreversible; it occurred at a point after the synthesis of viral RNA, and before the formation of virus protein measured as complement-fixing antigen or as antigen that could be stained with fluorescent antibody.  相似文献   

设为首页 | 免责声明 | 关于勤云 | 加入收藏

Copyright©北京勤云科技发展有限公司  京ICP备09084417号