更全的杂志信息网

Hepatitis B virus-persistent infection and innate immunity defect: Cell-related or virus-related?

更新时间:2016-07-05

INTRODUCTION

Hepatitis B virus (HBV) infection remains a global health concern, as about 257 million people worldwide are chronically infected with HBV currently[1]. Although the HBV infection rate has been partly controlled by various prophylaxis strategies, researchers have yet to discover a cure for chronic HBV infection. Chronic hepatitis B (CHB)is typically asymptomatic, but harbors the potential for development of life-threatening complications[2]. To overcome this threat, the mechanisms of HBV infection that underlie progression to chronicity need to first be fully elucidated.

HBV, a member of hepadnavirus family, acts as a“stealth” virus, not inducing any obvious innate immune responses in the early stage of infection[3]. Furthermore,the target cells (hepatocytes) do not recognize HBV efficiently through known signaling pathways, indicating the possibility of an HBV immune evasion mechanism[4].HBV also has the ability to suppress functions of innate immune cells[5-7]. HBV interaction with innate immunity would suggest that HBV persistence is related to a multitude of host and viral factors.

Herein, we summarize the recent knowledge regarding HBV persistence and the evidenced and theorized relations with intrahepatocyte signaling pathways and innate immune cells. Collation of such information will provide a useful overview of the field today, possibly providing new insights into novel therapeutic treatments for CHB.

Fundamentally, the innate immunity system responds to viral infection in three phases. In the first phase, various sensors in the cytoplasm recognize pathogen-associated molecular patterns, such as foreign DNA or RNA, and send a warning message to initiate downstream signals. The second phase involves the proteins of the downstream signaling pathways transmitting the danger message to the nucleus,activating effector elements. In the last phase, the consequently up-regulated effectors [i.e., inflammatory factors or interferon (IFN)-stimulating genes] degrade the exogenous viral elements. Defect or suppression of the involved sensors and signaling pathways tends to result in persistent existence of HBV in the host, since,under such circumstances, HBV cannot be recognized and eliminated in a timely manner. We will begin this review by discussing several intra-hepatocelluar pathways that are closely related to HBV recognition and which exert an anti-HBV effect.

《青苗法》每年获利息300万贯。单以熙宁六年为例,青苗利息就达 292万贯;《免役法》每年获利1872万9千3百,而支出只用所入的三分之一;《市易法》每年息钱和市利钱多达达133万2千缗之多。不仅如此,各州县岁收也大大增加。据统计,“诸路常平、免役、坊场、河渡、户绝庄乡之钱粟积于州县者无虑数十百巨万”,如作为地方政府经费,“可以支24年用”。 以上四组材料,充分说明王安石变法确实达到了“富国”的目的。

HEPATOCYTES LACK IMMUNE RESPONSIVENESS TO HBV

Pattern recognition receptors

Pattern recognition receptors (PRRs) are the major sensors of exogenous pathogens, and they include the Toll-like receptors (TLRs), retinoic acid-inducible gene I(RIG-I) and melanoma differentiation-associated gene 5(MAD5)[8]. These molecules are able to recognize diverse pathogen-associated common loci of specific pathogens,subsequently activating downstream signaling pathways that induce IFNs and inflammatory factors to control virus replication[9]. However, a recent study of liver tissues from CHB patients found that hepatocytes do not respond to HBV, even though they were able to produce IFNs and induce expression of IFN-stimulating genes when stimulated by other infection-related factors,such as those related to TLR3 binding and Sendai virus infection. This finding suggested that HBV is invisible to PRRs on hepatocytes[10].

Stimulator of IFN genes

Natural killer (NK) cells constitute 30%-40% of the intrahepatic lymphocytes, and are believed to play an indispensable role in HBV clearance. The NK cells exert their direct antiviral effects via secretion of IFN-γ,tumor necrosis factor (TNF)-α, granulocyte-macrophage stimulating factor, interleukin (IL)-10 and transforming growth factor-β to inhibit HBV replication or stimulate killing of infected cells[14,15]. However, studies of the functions of NK cells in early HBV infection have produced controversial results.

In vivo and in vitro experiments have shown that STING is NOT expressed in human hepatocytes, which might explain the dysfunction of DNA sensing[4]. Hence,HBV DNA may have adapted to this particular “biological niche”, whereby it can escape immune detection. More interestingly, c-GAMP is able to induce a robust cytokine response in a HBV-infected C3AhNTCP hepatoma cell line if added in cell culture, suggesting its capacities of inhibiting HBV replication[13].

INNATE IMMUNE CELLS HAVE WEAK RESPONSIVENESS TO HBV

Natural killer cells

Stimulator of IFN genes (STING) serves as the adaptor protein of multiple cytoplasmic DNA receptors that recognize exogenous pathogens[11]. It has been reported that cytoplasmic DNA-activated cyclic GMP-AMP synthase (commonly known as cGAS) binds to STING,thereby inducing secretion of type Ⅰ IFN and other cytokines[12]. This pathway is of great importance for eukaryotes (eukaryotic cells) to defend against bacterial,viral and other eukaryotic pathogens[11].

In the woodchuck model of acute hepatitis B, NK cells are activated within 48-72 h after infection, consequently leading to a transient reduction of HBV replication; but,this activation is unable to induce a timely adaptive T cell response[16]. Further, in chimpanzee models with selflimited HBV infection, NK cells do not become activated until the major histocompatibility complex-restricted α/β T cells enter the liver and recognize antigen; the T-specific cells, for the most part, carry out the clearance of HBV[3].Since these studies were carried out in animal models,it remains unclear to what extent the results mirror the human molecular processes.

In fact, the function of NK cells in early human HBV infection has proven very difficult to determine because the time frame from infection to diagnosis is typically more than 10-12 wk[17]. Regardless of how the NK cells function in this process, T-specific cells are the primary cell-types responsible for HBV clearance. Thus, the inaction of NK cells, along with immature or coincidently impaired functions of T cells, may explain how HBV infection progresses to chronicity.

A study group found that the pDC-NK cell crosstalk was suppressed by HBV[5], which might explain why the reaction of NK cells in early HBV infection is dull.Activation of NK cells is strongly dependent on IFN-α produced by pDCs[52]. HBsAg and HBeAg were also found to impair NK cell function, through the downregulation of IFN-γ expression[7,53]. In CHB patients,IFN-γ production is declined, and restoration of the IFN-γ-producing capacity is achievable after viral load reduction[54]. Consequently, the antiviral capacity of NK cells is compromised in chronic HBV infection[55-58]. In addition, HBV can stimulate generation of suppressive monocytes to initiate regulatory NK cell differentiation,resulting in T cell inhibition[59].

In addition, in human CHB, immune responses of NK cells are known to be altered. These alterations include impaired cytolytic activity[18] and up-regulation of antiviral T cell death receptors, the latter of which renders the cell susceptible to targeting for destruction by host mechanisms[19]. Given that several studies have demonstrated that activated NK cells and T cells can cause liver damage[20-23], the defective NK cells’ functions and the NK cell-mediated disarming of T-specific cells might be protective for hepatocytes while simultaneously contributing to HBV persistence.

Kupffer cells

Kupffer cells (KCs) are macrophages residing in the liver sinusoids. Programmed to screen and clear pathogens that they engage, the KCs serve as scavengers.They produce pro- and anti-inflammatory cytokines, as well as other molecules known to have antipathogen activities. In the early phase of HBV infection, KCs directly interact with HBV and produce proinflammatory cytokines (i.e., IL-6, TNF-α and IFN-α) to elicit their antiviral effects[24,25]. Subsequently, NK cells and HBV-specific immunity are also activated to aid in eradication of the virus. While the interaction between KCs and hepatitis B surface antigen (HBsAg) is mostly dependent upon the TLRs[25,26], expression of TLR3 is found to be significantly decreased in CHB patients (compared to that in healthy controls); this aberrant expression may contribute to HBV chronicity.

Moreover, if HBV infection progresses to chronicity,the role of KCs will change, just as that of NK cells. The major cytokines secreted by KCs shift towards an antiinflammatory profile (i.e., increased IL-10, which is known to contribute to the persistent existence of HBV in the host). In virus-persistent mouse models, stimulation of TLR2 by the hepatitis B core antigen leads to upregulation of IL-10 secretion by KCs, with the ultimate detrimental exhaustion of anti-HBV CD8+ T cells[27]. IL-10 suppresses not only T cell-mediated specific immunity but also humoral immunity[28]. As such, KC depletion or IL-10 deficiency will lead to restoration of CD8+ T cell function, breaking the cycle of humoral immune tolerance and allowing for clearance of the HBV[29]. Thus,the collective results in the literature have revealed that KC secretes IL-10 in chronic HBV infection to disarm humoral and cellular immunity, which leads to HBV persistence.

Dendritic cells

Dendritic cells (DCs) represent the most efficient professional antigen-presenting cell type. As such,although they reside universally throughout the body,their frequency accounts for less than 1% of the total peripheral blood mononuclear cells. Classified by expression of specific surface markers, the DCs are divided into several types, with those most frequently studied in chronic HBV infection being the plasmacytoid(p)DCs and the myeloid (m)DCs.

Besides antigen processing and presenting, the pDCs produce appreciable amounts of IFN-α, which serves to inhibit viral replication via the TLR7/9 signaling pathways, when stimulated. However, some studies have demonstrated that the frequency of pDCs is reduced in CHB patients (as compared with normal controls)[30-32],indicating that HBV persistence is associated with pDC reduction. This hypothesis, however, has been doubted following the finding of pDC frequency in CHB patients being similar to that in healthy individuals[33-36].Considering the rarity of pDCs in blood, it is likely that different experimental protocols account for the inconsistencies among these findings.

Regardless of the frequencies, the functions of pDCs are indeed hampered in chronic HBV infection. Most studies have reported a significant reduction of IFN-α secretion from pDCs[6,30-34]. Notably, the level of IFN-α has been found to be negatively correlated with alanine aminotransferase levels[33]. This finding could reflect a mechanism of HBV immune evasion and persistence,and an immune regulatory role for pDCs in this infection.Nevertheless, findings from an exceptional study led the authors to argue that pDC function was not impaired in chronic HBV infection[35]. The overall question remains unanswered.

mDCs are primarily responsible for inducing T cell differentiation and producing TNF-α. Unlike the pDCs,most studies on this cell type have found no difference in mDC frequency between CHB patients and healthy people[34-36]. Similar to the pDCs, however, it has been observed that the capacity of TNF-α production is impaired in mDCs when stimulated by HBV[34]. The other primary ability of mDCs, that of inducing T cell differentiation and proliferation, has not yet been defined in CHB patients.

HBV VIRUS-MEDIATED DISRUPTION OF THE HOST IMMUNOLOGICAL RESPONSE

RIG-I

据韩联社报道,韩国青瓦台2018年9月20日表示,北京时间当天早上6点27分,韩国总统文在寅夫妇从平壤机场乘坐飞机,于7点20分抵达白头山(长白山)附近的朝鲜三池渊机场。

TLRs

MAVS is a downstream signaling pathway protein of RIG-Ⅰ and MAD5. Localized in mitochondria[47], mitochondriaassociated endoplasmic reticular membranes[48] and peroxisomes[49], the protein contains an N-terminal CARD-like domain and a C-terminal transmembrane domain, anchoring to the mitochondrial membrane[47].RIG-I and MAD5 detect exogenous RNA in lymphocytes,subsequently communicating with the mitochondrial membrane and interacting with MAVS. Consequently,NF-κB and IRF3 become activated and induce IFN. Thus,MAVS is regarded as the central hub of the RIG-IFN axis.In the absence of MAVS, cells do not produce type Ⅰor Ⅲ IFN or proinflammatory cytokines upon infection with RNA virus[47]. This result was recapitulated in MAVS knockout mice[50]. In addition, the HBV X protein mediates the degradation of MAVS by utilizing Lys(136)ubiquitin (directly on the MAVS protein), thereby suppressing the induction of IFN-γ[51] (Figure 1).

As mentioned above, KCs’ ability to combat HBV infection mainly depends on expression of TLRs, but HBV itself can develop the ability to suppress TLR-mediated innate immune responses in the liver[26,60].Accordingly, HBV replication will be controlled when TLRs are stimulated by agonists; the effective TLRs that potently inhibit HBV replication have been identified as TLR3 and TLR4[41]. In addition, the expression of TLR3 displays restoration after antiviral therapy[61] and hepatic HBV infection triggers a TLR3-dependent immune responses in the absence of HBsAg[62]. Thus, HBsAg might be at the center of the yet-to-be-defined HBV evasion mechanism.

温度和通风是一对调节因素,需适当的配合使用。培养期间结合温度、湿度情况进行通风,每天2~3次,温度高、湿度大时,应增加通风次数和延长时间。

MAVS

In rat models, TLR3 activation has been shown to result in the production of type Ⅰ IFN to control HBV replication[41]. In mammalian cells, TLR2 and TLR4 share the MyD88-dependent signaling pathway, so that they mediate activation of the same downstream signaling pathways, including the NF-κB, MAPK and PI-3k/Akt pathways. Ultimately, the production of proinflammatory cytokines (i.e., TNF-α and IL-8) are up-regulated in hepatocytes, to inhibit the HBV replication[42].

HBV-MEDIATED DYSFUNCTION OF INNATE IMMUNE CELLS

HBV alters NK cell function

通过以上研究分析,集成管块式融雪剂喷洒装置安装城市快速路中央隔离带处,能够满足道路建筑限界的要求,可以保证车辆行驶安全,不会对道路使用带来严重不良影响。因此,为更好地提升城市冬季除雪应急保障能力,可逐步推广该装置在城市快速路上的安装使用。

HBV-induced type Ⅲ IFN expression depends on RIG-I[37]. A recent study has suggested that overexpressed RIG-I could dramatically reduce the levels of HBV mRNA and DNA in vitro[9]. More interestingly,RIG-I can directly exert antivirus function by preventing the HBV P protein from binding with the HBV pregenomic RNA 5’ stem-loop region[37]. MAD5 and RIG-I can recognize many viral RNAs, subsequently initiating downstream signaling pathways by up-regulating the adapter protein mitochondria-antiviral signaling protein[MAVS, also known as virus-induced signaling adaptor(VISA) and interferon promoter stimulator-1 (IPS-1)].In turn, the IFN regulatory transcription factor 3 (IRF3)and nuclear factor-kappa B (NF-κB), two of the most important proinflammatory transcription factors, become activated[38]. It has been reported, however, that the quantity of MDA5 is obviously decreased in CHB patients (compared to healthy controls)[39]. Moreover,a recent study revealed that HBV-induced miR146a attenuated innate immunity through targeting of RIG-I and RIG-G[40].

HBV suppresses the function of TLRs in KCs

Intriguingly, lower expression of TLR2 has been observed in hepatocytes, KCs and peripheral monocytes of hepatitis B e antigen (HBeAg)-positive CHB patients(compared with that in HBeAg-negative CHB and controls)[43]. Further research found that this down-regulated expression correlated with the levels of plasma HBsAg[44] and that HBsAg was able to selectively inhibit TLR2-induced IL-12 production from human monocytes/macrophages in a dose-dependent manner[45]. HBeAg was also shown to specifically inhibit the TLR-mediated activation of NF-κB and IFN-β[46].

广州古代皇家园林遗址,具有非常高的文化价值,应重视其遗址的保护工作。一是保护南越国皇家园林遗产,重点保护南越王宫以及东部宫苑,拓展其作为研究岭南地区秦汉时代园林艺术的宝贵文化价值。二是保护和拓展南汉国皇家园林遗产资源,重点保护现存的药洲遗址,恢复古时文人酬唱之盛景。通过开发和利用,拓展广州古代皇家园林遗产文化的内容。通过构建广州古代皇家园林遗址公园,增加古代园林的文化内容与景观,从而丰实广州古代的文化内容。通过建设广州皇家园林遗址公园,丰富广州的遗址性公园景观,增强岭南园林的景观资源,使其成为旅游资源的重要组成部分,从而推动社会经济的发展,为广州城市的经济注入源源不断的来源。

她叫林岚,或许叫凌岚,凌宇生的另一私生女。但是她的真实身份已经不重要了,从此以后,她就是凌安安,就是那个继承凌宇生大笔遗产让众人羡慕的私生女。

Figure 1 Relationships between hepatitis B virus and innate immunity system. NK cell: Nature killer cell; IFN-α: Interferon-α; IFNR: Interferon receptor; HBV:Hepatitis B virus; cGAS: Cyclic GMP-AMP synthase; TLRs: Toll-like receptors; pDC: Plasmacytoid dendritic cells; MyD88: Myeloid differentiation primary response 88; MAPK: Mitogen-activated protein kinase; PI3K: Phosphatidylinositol-3 kinase; NF-κB: Nuclear factor-kappa B; IRF3: IFN regulatory transcription factor 3; ER:Endoplasmic reticulum; TNF-α: Tumor necrosis factor alpha; IL8: Interleukin 8; RIG1: Retinoic acid-inducible gene Ⅰ.

HBV hampers secretion of antiviral cytokines from DCs

HBV lacks the capacity to activate pDCs, and many studies have also revealed that HBV could be able to inhibit IFN-α secretion from these cells[6,30-34]. Similarly,HBV can also reduce TNF-α production by mDCs[34].Thus, HBV-mediated impairment of functions of both pDCs and mDCs suggests a potential mechanism of HBV persistence; but, the mechanism by which HBV induces such dysfunction remains elusive.

NOVEL THERAPEUTIC STRATEGIES THAT BOOST INNATE IMMUNITY

Currently, the most frequently applied strategy of CHB treatment is PEG-IFN-α/nucleoside analogs (NA)combination which works quite well. However, if altered NK cell functions are corrected, the outcome of the drug combination could possibly be improved. As described above, the functions and number of innate immune cells such as NK cells, KCs and DCs can be impaired in chronic HBV infection. Thus, replacing, restoring or boosting the functions of innate immune cells can be potential therapeutic targets for viral load reduction and further, HBV clearance. And in return, once viral load reduction is achieved, the antiviral capacities of innate immune cells often restore.

对学生与数据分析观念相关的调查研究主要是从三个方面进行研究:一是就某一些统计概念的理解展开调查,如曲元海对初中生统计量理解的调查研究[12].二是单独对学生的数据分析观念进行调查,如李红梅对七年级学生数据分析观念的现状调查[4].三是在调查数学核心素养中包含数据分析,如张淑梅对高中数学核心素养统计分析过程中发现,数学建模与数据分析的相关性较大[13].

IFN-γ is able to inhibit the HBV replication within hepatocytes in the absence of immune cells[63]. Given that IFN-γ-producing capacity of NK cells restores after viral load reduction, it seems to be a positive-feedback when IFN-γ is used in HBV infection. What’s more,compared with IFN-α, IFN-γ serves as efficient as IFN-α but causes less side effects[64]. However, how IFN-γ combat HBV independent from immune cells remains unclear.

Agents enhancing the recognition of HBV could be used to wipe out HBV in the early stages of infection.Some known PRR agonists and TLR agonists could trigger a series of reaction in infected hepatocytes via IRF or NFκB pathways; since PRRs and TLRs are abundantly expressed in innate immune cells, such agonists are capable of rendering immune cells more powerful in recognizing HBV, thus contributing to early HBV clearance[65,66]. Vesatolimod (GS-9620), an oral TLR7 agonist, which was already involved in the 2nd phase clinical trial, was surprisingly found to be able to cause dose-dependent pharmacodynamic induction of ISG15 and a significant increase of serum cytokines[67].

(2) 词+动词 (动 名 词 ):power-brake 静 力 刹 车 器 ;machine-building机器制造。

Stimulation of certain innate immune cells to produce direct antiviral cytokines is another theoretical way to fight against human CHB. But in CHB patients,there is a balance between antiviral activity and antiinflammatory activity. It is unknown whether the agents would break the balance and what consequences they would bring (Table 1).

Table 1 Cytokines and factors implicated in causing inflammation in atherosclerosis

Cytokines/factors Abbreviations Hepatitis B virus HBV Chronic hepatitis B CHB Interferon IFN Pattern recognition receptors PRRs Toll-like receptors TLRs Melanoma differentiation-associated gene 5 MAD5 Retinoic acid-inducible gene I RIG I Stimulator of IFN genes STING Cyclic GMP-AMP synthase cGAS Human sodium taurocholate cotransporting polypeptide hNTCP Natural killer cell NK cell Tumor necrosis factor TNF Interleukin IL Kupffer cells KCs Dendritic cells DCs Plasmacytoid dendritic cells pDC Myeloid dendritic cells mDC Mitochondria-antiviral signaling protein MAVS Virus-induced signaling adaptor VISA Nucleoside analogs NA

CONCLUSION

The crosstalk between innate immunity and HBV persistence has been a controversial topic for a number of years. The collective findings we present in this review demonstrate that HBV is able to hamper innate immunity in many ways, and even to alter the functions of innate immune cells in order to suppress specific immune responses. This partly explains why HBV manages to progress to chronicity and to exist persistently. We hope the field will build upon these insights, leading to a deeper and more comprehensive understanding of HBV persistence and the role of innate immunity. Such knowledge will serve as a foundation for future development of effective immunomodulation treatment for chronic HBV infection. Indeed, therapeutic strategies that aim to restore innate immune responses may represent remarkably potent tools for reducing HBV chronicity and, further, for eradicating HBV infection.

REFERENCES

1 Seeger CWHO. Hepatitis B. 2018. Available from: URL: http://www.who.int/mediacentre/factsheets/fs204/en/

2 Seeger C, Mason WS. Molecular biology of hepatitis B virus infection. Virology 2015; 479-480: 672-686 [PMID: 25759099 DOI: 10.1016/j.virol.2015.02.031]

3 Wieland S, Thimme R, Purcell RH, Chisari FV. Genomic analysis of the host response to hepatitis B virus infection. Proc Natl Acad Sci USA 2004; 101: 6669-6674 [PMID: 15100412 DOI: 10.1073/pnas.0401771101]

4 Thomsen MK, Nandakumar R, Stadler D, Malo A, Valls RM,Wang F, Reinert LS, Dagnaes-Hansen F, Hollensen AK, Mikkelsen JG, Protzer U, Paludan SR. Lack of immunological DNA sensing in hepatocytes facilitates hepatitis B virus infection. Hepatology 2016; 64: 746-759 [PMID: 27312012 DOI: 10.1002/hep.28685]

5 Shi CC, Tjwa ET, Biesta PJ, Boonstra A, Xie Q, Janssen HL,Woltman AM. Hepatitis B virus suppresses the functional interaction between natural killer cells and plasmacytoid dendritic cells. J Viral Hepat 2012; 19: e26-e33 [PMID: 22239523 DOI:10.1111/j.1365-2893.2011.01496.x]

6 Martinet J, Dufeu-Duchesne T, Bruder Costa J, Larrat S, Marlu A, Leroy V, Plumas J, Aspord C. Altered functions of plasmacytoid dendritic cells and reduced cytolytic activity of natural killer cells in patients with chronic HBV infection. Gastroenterology 2012; 143: 1586-1596.e8 [PMID: 22960656 DOI: 10.1053/j.gastro.2012.08.046]

7 Yang Y, Han Q, Zhang C, Xiao M, Zhang J. Hepatitis B virus antigens impair NK cell function. Int Immunopharmacol 2016; 38:291-297 [PMID: 27341035 DOI: 10.1016/j.intimp.2016.06.015]

8 Pichlmair A, Reis e Sousa C. Innate recognition of viruses.Immunity 2007; 27: 370-383 [PMID: 17892846 DOI: 10.1016/j.immuni.2007.08.012]

9 Guo H, Jiang D, Ma D, Chang J, Dougherty AM, Cuconati A,Block TM, Guo JT. Activation of pattern recognition receptormediated innate immunity inhibits the replication of hepatitis B virus in human hepatocyte-derived cells. J Virol 2009; 83: 847-858[PMID: 18971270 DOI: 10.1128/JVI.02008-08]

10 Suslov A, Boldanova T, Wang X, Wieland S, Heim MH. Hepatitis B Virus Does Not Interfere With Innate Immune Responses in the Human Liver. Gastroenterology 2018; 154: 1778-1790 [PMID:29408639 DOI: 10.1053/j.gastro.2018.01.034]

11 Burdette DL, Vance RE. STING and the innate immune response to nucleic acids in the cytosol. Nat Immunol 2013; 14: 19-26[PMID: 23238760 DOI: 10.1038/ni.2491]

12 Gao D, Wu J, Wu YT, Du F, Aroh C, Yan N, Sun L, Chen ZJ.Cyclic GMP-AMP synthase is an innate immune sensor of HIV and other retroviruses. Science 2013; 341: 903-906 [PMID:23929945 DOI: 10.1126/science.1240933]

13 Guo F, Tang L, Shu S, Sehgal M, Sheraz M, Liu B, Zhao Q, Cheng J, Zhao X, Zhou T, Chang J, Guo JT. Activation of Stimulator of Interferon Genes in Hepatocytes Suppresses the Replication of Hepatitis B Virus. Antimicrob Agents Chemother 2017; 61: pii:e00771-17 [PMID: 28717041 DOI: 10.1128/AAC.00771-17]

14 Fisicaro P, Valdatta C, Boni C, Massari M, Mori C, Zerbini A,Orlandini A, Sacchelli L, Missale G, Ferrari C. Early kinetics of innate and adaptive immune responses during hepatitis B virus infection. Gut 2009; 58: 974-982 [PMID: 19201769 DOI: 10.1136/gut.2008.163600]

15 Li J, Han Y, Jin K, Wan Y, Wang S, Liu B, Liu Y, Lu S, Huang Z. Dynamic changes of cytotoxic T lymphocytes (CTLs), natural killer (NK) cells, and natural killer T (NKT) cells in patients with acute hepatitis B infection. Virol J 2011; 8: 199 [PMID: 21535873 DOI: 10.1186/1743-422X-8-199]

16 Guy CS, Mulrooney-Cousins PM, Churchill ND, Michalak TI. Intrahepatic expression of genes affiliated with innate and adaptive immune responses immediately after invasion and during acute infection with woodchuck hepadnavirus. J Virol 2008; 82:8579-8591 [PMID: 18596101 DOI: 10.1128/JVI.01022-08]

17 Webster GJ, Reignat S, Maini MK, Whalley SA, Ogg GS, King A, Brown D, Amlot PL, Williams R, Vergani D, Dusheiko GM,Bertoletti A. Incubation phase of acute hepatitis B in man: dynamic of cellular immune mechanisms. Hepatology 2000; 32: 1117-1124[PMID: 11050064 DOI: 10.1053/jhep.2000.19324]

18 Yang Y, Han Q, Hou Z, Zhang C, Tian Z, Zhang J. Exosomes mediate hepatitis B virus (HBV) transmission and NK-cell dysfunction. Cell Mol Immunol 2017; 14: 465-475 [PMID:27238466 DOI: 10.1038/cmi.2016.24]

19 Peppa D, Gill US, Reynolds G, Easom NJ, Pallett LJ, Schurich A,Micco L, Nebbia G, Singh HD, Adams DH, Kennedy PT, Maini MK. Up-regulation of a death receptor renders antiviral T cells susceptible to NK cell-mediated deletion. J Exp Med 2013; 210:99-114 [PMID: 23254287 DOI: 10.1084/jem.20121172]

20 Moriyama T, Guilhot S, Klopchin K, Moss B, Pinkert CA,Palmiter RD, Brinster RL, Kanagawa O, Chisari FV. Immunobiology and pathogenesis of hepatocellular injury in hepatitis B virus transgenic mice. Science 1990; 248: 361-364 [PMID:1691527 DOI: 10.1126/science.1691527]

21 Ando K, Moriyama T, Guidotti LG, Wirth S, Schreiber RD,Schlicht HJ, Huang SN, Chisari FV. Mechanisms of class I restricted immunopathology. A transgenic mouse model of fulminant hepatitis. J Exp Med 1993; 178: 1541-1554 [PMID:8228807 DOI: 10.1084/jem.178.5.1541]

22 Maini MK, Boni C, Lee CK, Larrubia JR, Reignat S, Ogg GS,King AS, Herberg J, Gilson R, Alisa A, Williams R, Vergani D,Naoumov NV, Ferrari C, Bertoletti A. The role of virus-specific CD8(+) cells in liver damage and viral control during persistent hepatitis B virus infection. J Exp Med 2000; 191: 1269-1280[PMID: 10770795 DOI: 10.1016/S0168-8278(00)80508-5]

23 Zheng Q, Zhu YY, Chen J, Ye YB, Li JY, Liu YR, Hu ML, Zheng YC, Jiang JJ. Activated natural killer cells accelerate liver damage in patients with chronic hepatitis B virus infection. Clin Exp Immunol 2015; 180: 499-508 [PMID: 25639451 DOI: 10.1111/cei.12597]

24 Hösel M, Quasdorff M, Wiegmann K, Webb D, Zedler U,Broxtermann M, Tedjokusumo R, Esser K, Arzberger S, Kirschning CJ, Langenkamp A, Falk C, Büning H, Rose-John S, Protzer U.Not interferon, but interleukin-6 controls early gene expression in hepatitis B virus infection. Hepatology 2009; 50: 1773-1782[PMID: 19937696 DOI: 10.1002/hep.23226]

25 Boltjes A, van Montfoort N, Biesta PJ, Op den Brouw ML,Kwekkeboom J, van der Laan LJ, Janssen HL, Boonstra A,Woltman AM. Kupffer cells interact with hepatitis B surface antigen in vivo and in vitro, leading to proinflammatory cytokine production and natural killer cell function. J Infect Dis 2015; 211:1268-1278 [PMID: 25362194 DOI: 10.1093/infdis/jiu599]

26 Wu J, Meng Z, Jiang M, Pei R, Trippler M, Broering R, Bucchi A, Sowa JP, Dittmer U, Yang D, Roggendorf M, Gerken G, Lu M, Schlaak JF. Hepatitis B virus suppresses toll-like receptormediated innate immune responses in murine parenchymal and nonparenchymal liver cells. Hepatology 2009; 49: 1132-1140[PMID: 19140219 DOI: 10.1002/hep.22751]

27 Li M, Sun R, Xu L, Yin W, Chen Y, Zheng X, Lian Z, Wei H, Tian Z. Kupffer Cells Support Hepatitis B Virus-Mediated CD8+ T Cell Exhaustion via Hepatitis B Core Antigen-TLR2 Interactions in Mice. J Immunol 2015; 195: 3100-3109 [PMID: 26304988 DOI:10.4049/jimmunol.1500839]

28 Xu L, Yin W, Sun R, Wei H, Tian Z. Kupffer cell-derived IL-10 plays a key role in maintaining humoral immune tolerance in hepatitis B virus-persistent mice. Hepatology 2014; 59: 443-452[PMID: 23929689 DOI: 10.1002/hep.26668]

29 Tian Y, Kuo CF, Akbari O, Ou JH. Maternal-Derived Hepatitis B Virus e Antigen Alters Macrophage Function in Offspring to Drive Viral Persistence after Vertical Transmission. Immunity 2016; 44: 1204-1214 [PMID: 27156385 DOI: 10.1016/j.immuni.2016.04.008]

30 Beckebaum S, Cicinnati VR, Dworacki G, Müller-Berghaus J,Stolz D, Harnaha J, Whiteside TL, Thomson AW, Lu L, Fung JJ,Bonham CA. Reduction in the circulating pDC1/pDC2 ratio and impaired function of ex vivo-generated DC1 in chronic hepatitis B infection. Clin Immunol 2002; 104: 138-150 [PMID: 12165275 DOI: 10.1006/clim.2002.5245]

31 Duan XZ, Wang M, Li HW, Zhuang H, Xu D, Wang FS. Decreased frequency and function of circulating plasmocytoid dendritic cells(pDC) in hepatitis B virus infected humans. J Clin Immunol 2004;24: 637-646 [PMID: 15622448 DOI: 10.1007/s10875-004-6249-y]

32 Shi B, Ren G, Hu Y, Wang S, Zhang Z, Yuan Z. HBsAg inhibits IFN-α production in plasmacytoid dendritic cells through TNF-α and IL-10 induction in monocytes. PLoS One 2012; 7: e44900[PMID: 23024774 DOI: 10.1371/journal.pone.0044900]

33 Woltman AM, Op den Brouw ML, Biesta PJ, Shi CC, Janssen HL.Hepatitis B virus lacks immune activating capacity, but actively inhibits plasmacytoid dendritic cell function. PLoS One 2011; 6:e15324 [PMID: 21246041 DOI: 10.1371/journal.pone.0015324]

34 van der Molen RG, Sprengers D, Binda RS, de Jong EC,Niesters HG, Kusters JG, Kwekkeboom J, Janssen HL. Functional impairment of myeloid and plasmacytoid dendritic cells of patients with chronic hepatitis B. Hepatology 2004; 40: 738-746 [PMID:15349914 DOI: 10.1002/hep.20366]

35 Tavakoli S, Mederacke I, Herzog-Hauff S, Glebe D, Grün S, Strand D, Urban S, Gehring A, Galle PR, Böcher WO. Peripheral blood dendritic cells are phenotypically and functionally intact in chronic hepatitis B virus (HBV) infection. Clin Exp Immunol 2008; 151:61-70 [PMID: 18031557 DOI: 10.1111/j.1365-2249.2007.03547.x]

36 Gehring AJ, Haniffa M, Kennedy PT, Ho ZZ, Boni C, Shin A,Banu N, Chia A, Lim SG, Ferrari C, Ginhoux F, Bertoletti A.Mobilizing monocytes to cross-present circulating viral antigen in chronic infection. J Clin Invest 2013; 123: 3766-3776 [PMID:23908113 DOI: 10.1172/JCI66043]

37 Sato S, Li K, Kameyama T, Hayashi T, Ishida Y, Murakami S,Watanabe T, Iijima S, Sakurai Y, Watashi K, Tsutsumi S, Sato Y,Akita H, Wakita T, Rice CM, Harashima H, Kohara M, Tanaka Y, Takaoka A. The RNA sensor RIG-I dually functions as an innate sensor and direct antiviral factor for hepatitis B virus.Immunity 2015; 42: 123-132 [PMID: 25557055 DOI: 10.1016/j.immuni.2014.12.016]

38 Cao X, Ding Q, Lu J, Tao W, Huang B, Zhao Y, Niu J, Liu YJ,Zhong J. MDA5 plays a critical role in interferon response during hepatitis C virus infection. J Hepatol 2015; 62: 771-778 [PMID:25463548 DOI: 10.1016/j.jhep.2014.11.007]

39 Ebrahim M, Mirzaei V, Bidaki R, Shabani Z, Daneshvar H,Karimi-Googheri M, Khaleghinia M, Afrooz MR, Yousefpoor Y,Arababadi MK. Are RIG-1 and MDA5 Expressions Associated with Chronic HBV Infection? Viral Immunol 2015; 28: 504-508[PMID: 26485346 DOI: 10.1089/vim.2015.0056]

40 Hou Z, Zhang J, Han Q, Su C, Qu J, Xu D, Zhang C, Tian Z.Hepatitis B virus inhibits intrinsic RIG-I and RIG-G immune signaling via inducing miR146a. Sci Rep 2016; 6: 26150 [PMID:27210312 DOI: 10.1038/srep26150]

41 Wu J, Lu M, Meng Z, Trippler M, Broering R, Szczeponek A,Krux F, Dittmer U, Roggendorf M, Gerken G, Schlaak JF. Toll-like receptor-mediated control of HBV replication by nonparenchymal liver cells in mice. Hepatology 2007; 46: 1769-1778 [PMID:17929296 DOI: 10.1002/hep.21897]

42 Thompson AJ, Colledge D, Rodgers S, Wilson R, Revill P,Desmond P, Mansell A, Visvanathan K, Locarnini S. Stimulation of the interleukin-1 receptor and Toll-like receptor 2 inhibits hepatitis B virus replication in hepatoma cell lines in vitro. Antivir Ther 2009; 14: 797-808 [PMID: 19812442 DOI: 10.3851/IMP1294]

43 Visvanathan K, Skinner NA, Thompson AJ, Riordan SM, Sozzi V, Edwards R, Rodgers S, Kurtovic J, Chang J, Lewin S, Desmond P, Locarnini S. Regulation of Toll-like receptor-2 expression in chronic hepatitis B by the precore protein. Hepatology 2007; 45:102-110 [PMID: 17187404 DOI: 10.1002/hep.21482]

44 Chen Z, Cheng Y, Xu Y, Liao J, Zhang X, Hu Y, Zhang Q, Wang J, Zhang Z, Shen F, Yuan Z. Expression profiles and function of Toll-like receptors 2 and 4 in peripheral blood mononuclear cells of chronic hepatitis B patients. Clin Immunol 2008; 128: 400-408[PMID: 18565796 DOI: 10.1016/j.clim.2008.04.006]

45 Wang S, Chen Z, Hu C, Qian F, Cheng Y, Wu M, Shi B, Chen J,Hu Y, Yuan Z. Hepatitis B virus surface antigen selectively inhibits TLR2 ligand-induced IL-12 production in monocytes/macrophages by interfering with JNK activation. J Immunol 2013; 190:5142-5151 [PMID: 23585678 DOI: 10.4049/jimmunol.1201625]

46 Lang T, Lo C, Skinner N, Locarnini S, Visvanathan K, Mansell A.The hepatitis B e antigen (HBeAg) targets and suppresses activation of the toll-like receptor signaling pathway. J Hepatol 2011; 55:762-769 [PMID: 21334391 DOI: 10.1016/j.jhep.2010.12.042]

47 Seth RB, Sun L, Ea CK, Chen ZJ. Identification and characterization of MAVS, a mitochondrial antiviral signaling protein that activates NF-kappaB and IRF 3. Cell 2005; 122: 669-682[PMID: 16125763 DOI: 10.1016/j.cell.2005.08.012]

48 Horner SM, Liu HM, Park HS, Briley J, Gale M Jr. Mitochondrialassociated endoplasmic reticulum membranes (MAM) form innate immune synapses and are targeted by hepatitis C virus. Proc Natl Acad Sci USA 2011; 108: 14590-14595 [PMID: 21844353 DOI:10.1073/pnas.1110133108]

49 Dixit E, Boulant S, Zhang Y, Lee AS, Odendall C, Shum B,Hacohen N, Chen ZJ, Whelan SP, Fransen M, Nibert ML, Superti-Furga G, Kagan JC. Peroxisomes are signaling platforms for antiviral innate immunity. Cell 2010; 141: 668-681 [PMID:20451243 DOI: 10.1016/j.cell.2010.04.018]

50 Sun Q, Sun L, Liu HH, Chen X, Seth RB, Forman J, Chen ZJ. The specific and essential role of MAVS in antiviral innate immune responses. Immunity 2006; 24: 633-642 [PMID: 16713980 DOI:10.1016/j.immuni.2006.04.004]

51 Wei C, Ni C, Song T, Liu Y, Yang X, Zheng Z, Jia Y, Yuan Y, Guan K, Xu Y, Cheng X, Zhang Y, Yang X, Wang Y, Wen C, Wu Q, Shi W,Zhong H. The hepatitis B virus X protein disrupts innate immunity by downregulating mitochondrial antiviral signaling protein. J Immunol 2010; 185: 1158-1168 [PMID: 20554965 DOI: 10.4049/jimmunol.0903874]

52 Gerosa F, Gobbi A, Zorzi P, Burg S, Briere F, Carra G, Trinchieri G. The reciprocal interaction of NK cells with plasmacytoid or myeloid dendritic cells profoundly affects innate resistance functions. J Immunol 2005; 174: 727-734 [PMID: 15634892 DOI:10.4049/jimmunol.174.2.727]

53 Jegaskanda S, Ahn SH, Skinner N, Thompson AJ, Ngyuen T, Holmes J, De Rose R, Navis M, Winnall WR, Kramski M,Bernardi G, Bayliss J, Colledge D, Sozzi V, Visvanathan K,Locarnini SA, Kent SJ, Revill PA. Downregulation of interleukin-18-mediated cell signaling and interferon gamma expression by the hepatitis B virus e antigen. J Virol 2014; 88: 10412-10420 [PMID:24872585 DOI: 10.1128/JVI.00111-14]

54 Tjwa ET, van Oord GW, Hegmans JP, Janssen HL, Woltman AM.Viral load reduction improves activation and function of natural killer cells in patients with chronic hepatitis B. J Hepatol 2011; 54:209-218 [PMID: 21095036 DOI: 10.1016/j.jhep.2010.07.009]

55 Oliviero B, Varchetta S, Paudice E, Michelone G, Zaramella M, Mavilio D, De Filippi F, Bruno S, Mondelli MU. Natural killer cell functional dichotomy in chronic hepatitis B and chronic hepatitis C virus infections. Gastroenterology 2009; 137:1151-1160, 1160.e1-1160.e7 [PMID: 19470388 DOI: 10.1053/j.gastro.2009.05.047]

56 Lunemann S, Malone DF, Hengst J, Port K, Grabowski J,Deterding K, Markova A, Bremer B, Schlaphoff V, Cornberg M, Manns MP, Sandberg JK, Ljunggren HG, Björkström NK,Wedemeyer H. Compromised function of natural killer cells in acute and chronic viral hepatitis. J Infect Dis 2014; 209: 1362-1373[PMID: 24154737 DOI: 10.1093/infdis/jit561]

57 Heiberg IL, Pallett LJ, Winther TN, Høgh B, Maini MK, Peppa D. Defective natural killer cell anti-viral capacity in paediatric HBV infection. Clin Exp Immunol 2015; 179: 466-476 [PMID:25311087 DOI: 10.1111/cei.12470]

58 Ghosh S, Nandi M, Pal S, Mukhopadhyay D, Chakraborty BC,Khatun M, Bhowmick D, Mondal RK, Das S, Das K, Ghosh R,Banerjee S, Santra A, Chatterjee M, Chowdhury A, Datta S. Natural killer cells contribute to hepatic injury and help in viral persistence during progression of hepatitis B e-antigen-negative chronic hepatitis B virus infection. Clin Microbiol Infect 2016; 22: 733.e9-733.e19 [PMID: 27208430 DOI: 10.1016/j.cmi.2016.05.009]

59 Li H, Zhai N, Wang Z, Song H, Yang Y, Cui A, Li T, Wang G, Niu J, Crispe IN, Su L, Tu Z. Regulatory NK cells mediated between immunosuppressive monocytes and dysfunctional T cells in chronic HBV infection. Gut 2017; pii: gutjnl-2017-314098 [PMID:28899983 DOI: 10.1136/gutjnl-2017-314098]

60 Jiang M, Broering R, Trippler M, Poggenpohl L, Fiedler M,Gerken G, Lu M, Schlaak JF. Toll-like receptor-mediated immune responses are attenuated in the presence of high levels of hepatitis B virus surface antigen. J Viral Hepat 2014; 21: 860-872 [PMID:24498958 DOI: 10.1111/jvh.12216]

61 Huang YW, Lin SC, Wei SC, Hu JT, Chang HY, Huang SH,Chen DS, Chen PJ, Hsu PN, Yang SS, Kao JH. Reduced Tolllike receptor 3 expression in chronic hepatitis B patients and its restoration by interferon therapy. Antivir Ther 2013; 18: 877-884[PMID: 23744559 DOI: 10.3851/IMP2630]

62 Real CI, Lu M, Liu J, Huang X, Trippler M, Hossbach M, Deckert J, Jahn-Hofmann K, Ickenstein LM, John MJ, Gibbert K, Dittmer U, Vornlocher HP, Schirmbeck R, Gerken G, Schlaak JF, Broering R. Hepatitis B virus genome replication triggers toll-like receptor 3-dependent interferon responses in the absence of hepatitis B surface antigen. Sci Rep 2016; 6: 24865 [PMID: 27121087 DOI:10.1038/srep24865]

63 Xia Y, Stadler D, Lucifora J, Reisinger F, Webb D, Hösel M, Michler T, Wisskirchen K, Cheng X, Zhang K, Chou WM, Wettengel JM, Malo A, Bohne F, Hoffmann D, Eyer F,Thimme R, Falk CS, Thasler WE, Heikenwalder M, Protzer U.Interferon-γ and Tumor Necrosis Factor-α Produced by T Cells Reduce the HBV Persistence Form, cccDNA, Without Cytolysis.Gastroenterology 2016; 150: 194-205 [PMID: 26416327 DOI:10.1053/j.gastro.2015.09.026]

64 Chan HLY, Ahn SH, Chang TT, Peng CY, Wong D, Coffin CS,Lim SG, Chen PJ, Janssen HLA, Marcellin P, Serfaty L, Zeuzem S, Cohen D, Critelli L, Xu D, Wind-Rotolo M, Cooney E; LIRA-B Study Team. Peginterferon lambda for the treatment of HBeAgpositive chronic hepatitis B: A randomized phase 2b study(LIRA-B). J Hepatol 2016; 64: 1011-1019 [PMID: 26739688 DOI:10.1016/j.jhep.2015.12.018]

65 Isogawa M, Robek MD, Furuichi Y, Chisari FV. Toll-like receptor signaling inhibits hepatitis B virus replication in vivo. J Virol 2005;79: 7269-7272 [PMID: 15890966 DOI: 10.1128/JVI.79.11.7269-7 272.2005]

66 Lucifora J, Maadadi S, Floriot O, Daffis S, Fletcher S, Zoulim F, Durantel D. P0535: Direct antiviral effects of various pattern recognition receptor (PRR) agonists in HBV-replicating hepatocytes. J Hepatol 2015; 62: S515-S516 [DOI: 10.1016/S0168-8278(15)30742-X]

67 Janssen HLA, Brunetto MR, Kim YJ, Ferrari C, Massetto B,Nguyen AH, Joshi A, Woo J, Lau AH, Gaggar A, Subramanian GM,Yoshida EM, Ahn SH, Tsai NCS, Fung S, Gane EJ. Safety, efficacy and pharmacodynamics of vesatolimod (GS-9620) in virally suppressed patients with chronic hepatitis B. J Hepatol 2018; 68:431-440 [PMID: 29104121 DOI: 10.1016/j.jhep.2017.10.027]

-Y,-J,-Z
《World Journal of Clinical Cases》2018年第9期文献

服务严谨可靠 7×14小时在线支持 支持宝特邀商家 不满意退款

本站非杂志社官网,上千家国家级期刊、省级期刊、北大核心、南大核心、专业的职称论文发表网站。
职称论文发表、杂志论文发表、期刊征稿、期刊投稿,论文发表指导正规机构。是您首选最可靠,最快速的期刊论文发表网站。
免责声明:本网站部分资源、信息来源于网络,完全免费共享,仅供学习和研究使用,版权和著作权归原作者所有
如有不愿意被转载的情况,请通知我们删除已转载的信息