Regulatory B cells that produce IL-10 A breath of fresh air in allergic airway disease

合集下载

调节性B淋巴细胞在自身免疫性肝炎发生发展中的作用机制 童聪

调节性B淋巴细胞在自身免疫性肝炎发生发展中的作用机制 童聪

淋巴细胞可导致小鼠脾脏中巨噬细胞数量增多,提示Breg 对巨 等抑炎因子分泌减少可能与Bregs 数量及功能降低相关。
噬细胞存在负性调节作用。
2. 2 Breg 的功能 目前,B 淋巴细胞在AIH 中发挥的作用逐
Breg 也可以通过膜表面分子FasL、PDL - 1 和GITRL 调节 渐引起学者的关注。Béland 等[4]的研究表明,在小鼠AIH 模型
表型
主要特征
CD19 + CD24hi CD27 + CD19 + CD24hi CD38hi CD19 + CD38 + CD27int CD25hi CD71hi CD73low CD19 + CD25 + CD1dhi IgMhi CD5 - CD23 - TIM - 1 - CD5CD24 - / + CD27hi CD38dim
发挥其负性调节作用,还可以通过多种膜表面分子如人凋亡相 来越多的人把目光放到了Breg 上,对Breg 的了解也在不断
关因子配体(FasL)、细胞程序性死亡- 配体1(programmed cell 加深。
death - ligand ,1 PD - L1)和糖皮质激素诱导的肿瘤坏死因子受 1. 2 Breg 的分化及亚群 Breg 的生成受一些因素影响,Toll 样
体配体( 受体对 的分化起着重要的作用, 样受体能够与非甲 glucocorticoid - induced tumor necrosis factor receptor lig
Breg
Toll

and,GITRL)作用于其他免疫细胞发挥调节免疫作用[2]。目前 基化CpG 基序结合,刺激Breg 产生IL - 10。Mizoguchi 等[7]通
作用于 淋巴细胞,诱导 细胞群来抑制炎症。 进行检测, 组 、 T

双虎清肝颗粒对慢性乙型肝炎患者IL-10和TGF-β的影响以及临床疗效研究

双虎清肝颗粒对慢性乙型肝炎患者IL-10和TGF-β的影响以及临床疗效研究

双虎清肝颗粒对慢性乙型肝炎患者IL-10和TGF-β的影响以及临床疗效研究祝丽超;毕夏;陈晓杨【摘要】目的:观察双虎清肝颗粒对慢性乙型肝炎患者临床疗效及白细胞介素-10(IL-10)和转化生长因子-β(TGF-β)表达的影响.方法:符合纳入标准的慢性乙型肝炎患者134例,使用信封法随机分为对照组(阿德福韦酯)和观察组(双虎清肝颗粒联合阿德福韦酯),每组67例.采用ELISA法检测IL-10和TGF-β表达情况.结果:治疗前两组患者丙氨酸氨基转移酶(ALT)、天门冬氨酸氨基转移酶(AST)、总胆红素(TBiL)、透明质酸酶(HA)、层黏连蛋白(LN)、Ⅲ型前胶原(Ⅲ-PC)、Ⅳ型胶原(Ⅳ-C)、IL-10和TGF-β无显著性差异(P>0.05).治疗后观察组对上诉指标的减低幅度显著高于对照组(P<0.05).治疗结束后观察组转阴率为68.6%(46/67),显著高于对照组的50.7% (34/67)(P<0.05).观察组不良反应发生率为10.1% (7/67),对照组不良反应发生率为9.0% (6/67),两组不良反应无显著性差异(P>0.05).结论:双虎清肝颗粒可以减低慢性乙型肝炎患者IL-10和TGF-β因子表达,有利于肝功能的恢复并发挥抗肝纤维化的作用,临床使用安全、有效.%Objective:To observe the effectsof Shuanghu Qinggan granules on IL-10 and TGF-β in patients with chronic hepatitis B and its curative efficacy.Methods:134 patients were randomly divided into the control group (n =67) and the observation group (n=67);the control group was treated with Adefovir Dipivoxil tablets,on which basis,the observation group was also given Shuanghu Qinggan granules.The levels of IL-10 and TGF-β were detected by ELISAmethod.Results:There was no significant difference in terms ofALT,AST,TBiL,HA,L N,Ⅲ-PC,ⅣV-C,IL-10,and TGF-β between the two groupsbefore treatment(P > 0.05).After treatment,they all became significantly lower in the observation group than those in the control group(P <0.05);the negative conversion rate in the observation group was 68.6%(46/67),which was significantly higher than 50.7% (34/67) in the control group(P <0.05);the incidence of adverse reactions was 10.1% (7/67) in the observation group and 9% (6/67) in the control group,but there was no significant difference between the two groups (P > 0.05).Conclusion:The therapy of Shuanghu Qinggan granules can reduce the expressions of IL-10 and TGF-β in patients with chronic hepatitis B,which was conducive to the recovery of liver function and it plays a role in anti-fibrosis,with its safety and effectiveness in clinic.【期刊名称】《中医药信息》【年(卷),期】2017(034)006【总页数】4页(P74-77)【关键词】双虎清肝颗粒;慢性乙型肝炎;白细胞介素-10;转化生长因子-β【作者】祝丽超;毕夏;陈晓杨【作者单位】辽宁中医药大学附属第三医院,辽宁沈阳110000;辽宁中医药大学附属第三医院,辽宁沈阳110000;辽宁中医药大学附属第三医院,辽宁沈阳110000【正文语种】中文【中图分类】R285.6乙型肝炎病毒感染是全世界范围内主要的健康问题,我国60岁以下人群的乙型肝炎病毒表面抗原携带率为7.2%,大约有20%~40%的携带者会发展为慢性乙型肝炎,成为我国肝硬化的主要原因[1]。

IL-10-producing regulatory B cells develop exacerbated disease

IL-10-producing regulatory B cells develop exacerbated disease

J Immunol. 2011 May 15;186(10):5569-79. Epub 2011 Apr 4.Mice lacking endogenousIL-10-producing regulatory B cells develop exacerbated disease and present with an increased frequency of Th1/Th17 but a decrease in regulatory T cells.Carter NA, Vasconcellos R, Rosser EC, Tulone C, Muñoz-Suano A, Kamanaka M, Ehrenstein MR, Flavell RA, Mauri C.大鼠缺乏产内生性IL-10的调节性B细胞加重病情,且Th1/Th17的表达增加而在调节性T 细胞中减少SourceDivision of Medicine, Centre for Rheumatology Research, University College London, London W1T 4JF, United Kingdom.AbstractIL-10-producing B cells, also known as regulatory B cells (Bregs), play a key role in controlling autoimmunity.In this study, we report that chimeric mice specifically lackingIL-10-producing B cells (IL-10(-/-)B cell) developed an exacerbated arthritis compared with chimeric wild-type (WT) B cell mice.相对于嵌合的野生型B细胞小鼠组,嵌合体小鼠缺乏产IL-10的B细胞(IL-10(-/-)B cell)明显地加重了关节炎。

IBD疾病中的细胞因子-cytokines in inflammatory bowel disease

IBD疾病中的细胞因子-cytokines in inflammatory bowel disease

Cytokines in inflammatory bowel diseaseAL.C. MOLDOVEANU1,3, M. DICULESCU2,3, CARMEN FIERBINŢEANU BRATICEVICI1,3 1Internal Medicine II and Gastroenterology Clinic, University Hospital Bucharest2Department of Gastroenterology, Fundeni Clinical Institute, Bucharest3“Carol Davila” University of Medicine and Pharmacy, Bucharest Inflammatory bowel diseases are chronic afflictions, characterized by active and remission periods. Inflammation is the most common type of response that the human body uses as a defense mechanism against aggressors from the environment. The frequency and degree of inflammation depends on the size of the affected tissues. The gastrointestinal tract is, by far, the most susceptible tissue to inflammatory responses, because of its constant exposure to various antigenic, mutagenic and toxic factors.In inflammatory bowel diseases there is a loss of immune tolerance to intestinal flora that is mediated by various substances, including cytokines.Cytokines represent a key signal in the intestinal immune response. Activated dendritic cells and macrophages secrete cytokines that actively intervene in inflammation regulation, in both Crohn’s disease and ulcerative colitis. After their secretion by antigen presented cells, cytokines activate and differentiate T cells, stirring up the adaptive immune response.Cytokines have an important role in the pathogenesis of inflammatory bowel diseases. The identification of new cytokines, as well as the changing of the pathogenesis paradigms in inflammatory bowel diseases has been done on animal tests and clinical studies. Thus, there is promising evidence basis for future therapy research based on cytokines, and anti-cytokine antibodies.Keywords: inflammatory bowel disease, immune response, cytokines.Inflammatory bowel diseases (IBD) are chronic afflictions, characterized by active and remission periods. The IBD entity includes three forms of disease: Crohn’s Disease (CD), Ulcerative Colitis (UC) and Indeterminate Colitis. As of this moment, the pathogenesis of this group of diseases is not fully understood.Inflammation is the most common type of response that the human body uses as a defense mechanism against aggressors from the environment. The frequency and degree of inflammation depends on the size of the affected tissues. The gastro-intestinal tract is, by far, the most susceptible tissue to inflammatory responses, because of its constant exposure to various antigenic, mutagenic and toxic factors. Even in normal conditions, the intestinal mucosa is in a state of “physiologic inflammation”, manifested by the abundance of leukocytes in the intraepithelial and subepithelial compartments [1][2]. This is the consequence of an immune response to bacterial antigens and antigens in food. This concept has led to the idea that the bowel represents the largest lymphoid organ in the human body [1][3].There is, also, a real inflammatory response (what could be called “pathological inflammation”), dominated by cells of the immune system from the intestinalmucosa. According to this theory, theactive immune cells, represented by the neutrophils,macrophages and cytotoxic T-cells act as aggressorsthat attack and destroy the cells in the vicinity,either by direct contact or indirectly by releasingsoluble factors like reactive oxygen species ornitric metabolites, cytotoxic proteins, lytic enzymesor cytokines [1][4][5].There are two response models for the non-immune cells of the intestinal mucosa:1. A unidirectional model, where all non-immune cells of the mucosa are passive, waiting tobe damaged and eventually die as a result of theactions of effector immune cells [1] (Fig. 1).2. A bidirectional or multidirectional model,where all the cell types that populate the mucosahave an active role in the intestinal immunity andinflammation. Epithelial, endothelial, mesenchymal,nervous cells have effector and regulatory roles,including immune-like functions. Even cellularcomponents appear to play an important active rolein the immuno-regulation activity of the extra-cellular matrix, in normal conditions as well as inROM. J. INTERN. MED., 2015, 53, 2, 118–127DOI: 10.1515/rjim-2015-00162 Cytokines in inflammatory bowel disease119the presence of inflammation [1][6][7]. The multi-directional model of interference between effector cells and target cells is probably the most realistic and can best explain the mechanism of chronic inflammation of the bowel in IBDs [1] (Figure 2). Thus, the reactive behavior of non-immune cells can be explained. This causes symptoms and clinical modifications like pain, changes in intestinal motility, fibrous structures, fistulas, intestinal occlusion or neoplastic transformation.In IBDs there is a loss of immune tolerance to intestinal flora that is mediated by various substances, including cytokines.Cytokines represent a key signal in the intestinal immune response. Cytokines are small peptides produced by immune cells that have the role of transmitting intercellular signals, the stimulation of cell proliferation for the antigen specific effector cells and the autocrine, paracrine and endocrine mediation of local and systemic inflammation. Activated dendritic cells and macrophages secrete cytokines that actively intervene in inflammation regulation, in both CD and UC. After their secretion by antigen presented cells (APCs), cytokines activate and differentiate T cells, activating the adaptiveimmune response.Fig. 1. Unidirectional model of cell-cell interactions in intestinal inflammation.Fig. 2. The bidirectional or multidirectional model.Al.C. Moldoveanu et al .3120 T helper cells (Th) can be differentiated in either Th1 or Th2 cells that can produce various types of cytokines. In IBDs, the elimination of over-activated and auto-reactive T cells is perturbed and a disequilibrium of activated Treg/Th1, Th2 and Th17 cells occurs. The lack of regulation by T cells or an overproduction of effector T cells participates in the development and exacerbation of IBDs [8][9].Together, APCs, Th1, Th2, regulatory T cells and Th17 (recently discovered) and cytokines produced by them have a complex role in the development of IBDs [9][10]. The pathway by which the activation of cytokines influences the immune response in IBDs can be very different from one form of the disease to another [9] [11].Cellular interactions can be modulated by the action of traditional cytokines like Tumor Necrosis Factor α (TNF-α), Gamma Interferon (INF-γ), Interleukins (IL-1, IL-6, IL-4, IL-5, IL-10), Trans-forming Growth Factor β (TGF-β) – or of other, more recently described cytokines like IL-13, IL-12, IL-18, IL-23, that can have a pro-inflammatory effect as well as an anti-inflammatory effect [9].It appears that CD is mediated by the Th1/ Th17 cytokines and characterized by the increase in production of INF-γ and TNF-α (Figure 3). IL-12 and IL-23 control the differentiation of Th1 cells that, together with IL-15, IL-18 and IL-21, produce a stabilization of Th1 cells.UC is characterized by the production of IL-13 by the natural killer T (NKT) CD1 cells, and by the production of cytokines by Th2 cells [9]. It appears that UC is a disease mediated by Th2 cytokines andis characterized by an increase in production of IL-5 and a normal production of INF-γ [12-14]. In contrast, the production of IL-4 by Th2 cells was not increased in UC, which leads to the conclusion that it is a “Th-2 like” disease, rather than a true Th2 disease. NKT cells that produce IL-13 and IL-5 are the effectors of the main response [14] (Figure 4).PRO-INFLAMMATORY CYTOKINESTUMOR NECROSIS FACTOR αLymphocytes and APCs are responsible for the inflammation in IBDs, especially by increasing the production of the Tumor Necrosis Factor α (TNF-α).TNF-α is produced by macrophages, mono-cytes and differentiated T cells. It has a pro-inflammatory effect through an increased production of IL-1β and IL-6, the expression of adhesion molecules, the proliferation of fibroblasts and pro-coagulant factors and the initiation of the acute response, cytotoxic and apoptotic phases, but also the inhibition of apoptosis [15][16]. The expression of TNF-α has been identified in the colonic tissues and macrophages in both CD and UC [17]. The serum levels of TNF-α correlates with the presence of clinical and biological elements that indicate the activity of the intestinal disease [18]. Clinical studies have shown an important improvement in patients with CD, after the administration of anti-TNF-α therapy, like infliximab, adalimumab orcertolizumab pergola [19].Fig. 3. Th1/Th17 Response in Crohn’s Disease.4 Cytokines in inflammatory bowel disease121Fig. 4. Th2-Like Response in Ulcerative Colitis.The signaling pathway of TNF-α uses two serum soluble receptors, TNF receptor type I and II (STNF-RI, STNF-RII). The levels of these receptors correlate well with disease activity in IBD patients. More precisely, sTNF-RI up-regulates in the serum of IBD patients, compared to the healthy controls, and could potentially be used as a marker for disease activity [20]. The levels of sTNF-RII are significantly higher in patients with CD compared to patients with UC and could be used as an extra parameter for the differentiation between the two diseases [9][19].However, the activation of the STNF-RI based immune response was recently shown on animal models to reduce the mortality associated with intestinal lesions [21].TNF-LIKE LIGAND 1ATNF-like 1A (TL1A) is a cytokine, whose participation to intestinal inflammation has been relatively recently demonstrated. It is secreted by APCs, T cells as well as endothelial cells. Their signaling pathway involves binding of the Death Domain-Containing Receptor 3(DR3).TL1A appears to stimulate the secretion of INF-γ through the stimulation of Th1 cells. A larger percentage of lymphocytes express the DR3 receptor for TL1A, in biopsies taken from afflicted bowel areas in CD and UC patients, and an increased synthesis of INF-γ has correlated with disease severity in IBD patients [22].The studies performed on mice suggest that TL1A is a co-stimulating cytokine that optimizes the Th1 and Th17 responses, inducing inflame-mation [14]. Its pro-inflammatory effect affects both Th1/Th17 and Th2 cells. This dichotomy augments both types of T cell response. Studies performed on animals regarding TL1A are consistent with what is found in IBDs. Increased levels of TL1A have been shown in both CD and UC [22-25]. CD14+ macrophages in CD produce large quan-tities of TL1A, which will determine an increase in INF-γ and IL-17.It appears that TL1A produces an augmentation of the inflammatory effect more than the induction of an inflammatory effect as its primary mechanism. The polymorphism of TL1A genes is associated with an increased risk for CD [26].INTERLEUKIN 1Interleukin 1 (IL-1) along with TNF-α is important in the pathogenesis of IBD due to its up-regulatory and pro-inflammatory activity. The IL-1 system is formed mainly by IL-1α and IL-1β, both of which induce the production of type 2 cyclo-oxygenase, phospholipase A and inducible nitric oxide synthase (iNOS) [27]. The tissular level of IL-1 was found to be significantly increased in UC patients compared to control [28].The IL-1 system also includes antagonists of the IL-1 receptors (IL-1Ra), as a control mechanism. In UC, IL-1, IL-1Ra and the Transforming GrowthAl.C. Moldoveanu et al. 5 122factor β1(TGFβ1) influence the size and time length of the inflammatory process. The IL-1Ra/ IL-1 ratio has frequently been used as a predictor of inflammation of the mucosa, however the level of IL-1, rather than the level of IL-1Ra or the ratio IL-1Ra/IL-1 correlates best with the degree of severity of inflammation [28].IL-1Ra inhibits IL-1 only in severely inflamed tissues of IBD patients while TGFβ1’s controlling effect on inflammation has been more obvious in mild cases of UC [28].INTERLEUKIN 6Interleukin 6 (IL-6) performs its pro-inflam-matory action by means of the IL-6 soluble receptor (sIL-6R). The combination of the soluble receptor IL-6 (sIL-6R) and IL-6 plays an important role in the multiple immunological reactions that appear in IBD and the levels of IL-6 and of sIL-6R can correlate with multiple clinical manifestations that appears in CD and UC, including fibrogenesis [9, 29-32].The peripheral immune cells, colonic epithelial cells and cells from the lamina propria along with an active form of the IL-6/STAT3 system could be responsible for the strong correlation with the degree of mucosa inflammation [33].STAT-3 induces anti-apoptotic factors like Bcl-2 and Bcl-xL, resulting in increased T cell resistance to apoptosis. This cycle of T cell accu-mulation, mediated by the resistance to apoptosis finally leads to chronic inflammation [34]. Blocking the IL-6 signaling pathway leads to the apoptosis of T cells, which shows that the IL-6 – sIL-6R system mediates the resistance of T cells to apoptosis in CD [32, 34]. Anti sIL-6R antibodies have been shown to reduce the levels of INF-γ, TNF-α and IL-1β mRNA and have suppressed the expression of several intercellular adhesion molecules in the colonic vascular endothelium [35][36]. The IL-6/ STAT3 signaling in the activation of T mucosal cells has been suggested as a therapeutic target for the future [37].INTERLEUKIN 18Interleukin 18 (IL-18) is produced by intestinal epithelial cells and has been initially identified as an IFN-γ producing factor. It has several similarities with the IL-1 family, including structure, action, receptor, signal transmission model and anti-inflammatory action [38].The equilibrium between IL-18 and its natural inhibitor, Interleukin 18 Binding Protein (IL-18BP), can have a role in IBD pathogenesis [39]. An increase in the local production of IL-18 and IL18-BP has been shown in chronic lesions compared to control in CD [40], but the increase in IL18-BP is insufficient to control inflammation [39].The IL-12 cytokine can act synergic with IL-18 to promote the production of INF-γ, causing severe inflammation of the intestine [41]. The development of Th1 CD4+ cells in the intestinal mucosa is induced by IL-12 produced by activated macrophages and IL-18 produced by activated macrophages and colonic epithelial cells. The synergic effect is caused by up-regulation mechanism of the receptor for IL-18 by IL-12 [42].IL-18 may play a key role in Th1 mediated disorders, including CD, and thus, there could be a rationale for an anti-IL-18 based therapy [40].INTERLEUKIN 17Recently, a new type of T helper cell has been identified, called “Th17” and it is charac-terized by the production of Interleukin 17(IL-17) and has been identified as having an important role in inflammatory response [43]. Five other members of the IL-17 family have been identified, named IL-17B to IL-17F. IL-17A is exclusively produced in Th17 cells [44].IL-17 favorizes the production of multiple pro-inflammatory factors, including TNF-α, IL-6 and IL-1β, which has led to the conclusion that IL-17 has a very important role in the amplification and localization of inflammation [45-47], as well as the process of fibrosis in CD [48][49]. TNF-α and IL-6, both of which are produced by Th17 cells, not only sustain the development of Th17 cells, but also have a synergic action with IL-17 to increase the production of pro-inflammatory mediators [45].IL-17 as well as Th17 cells have been identified as having an increased serum and intestinal tissue level in IBD patients [50]. IL-17 has not been identified in the tissues of inactive patients or other types of colitis [51].INTERLEUKIN 12 AND INTERLEUKIN 23Interleukin 12 (IL-12) and Interleukin 23 (IL-23) belong to the IL-12 family of cytokines. They are produced mainly by activated APCs and other cells like dendritic cells and phagocytes [52]. IL-12 induces the production of INF-γ and the differentiation of Th0 cells into Th1 cells. It represents a link between innate resistance and adaptive resistance.6 Cytokines in inflammatory bowel disease123ANTI-INFLAMMATORY ANDIMMUNOMODULATOR CYTOKINESINTERLEUKIN 4Interleukin 4 (IL-4) induces the differentiation of naive T helper cells (Th0 cells) to Th2 cells. A study on animal models has shown that treatment with anti IL-4 antibodies leads to increased INF-γ production and to the induction of a shift of Th2 cells to the Th1 type [53].Another study has shown that the administration of IL-4 determines a significant reduction in the increase of Vascular Endothelial Growth Factor (VEGF) that normally appears in the peripheral blood in CD and UC patients, suggesting that a defective immunosuppressive effect of IL-4 may contribute to the pathologic mechanism of IBD [54].INTERLEUKIN 10Interleukin 10 (IL-10) is a cytokine with anti-inflammatory effects that inhibits both the antigen presentation and the subsequent release of pro-inflammatory cytokines. An inactivation of IL-10 in mice has led to the increased production of IL-12 and INF-γ [55][56]. In CD with active inflame-mation and granulomas, the IL-10 levels were found to be decreased.Regulatory B cell subtype (Bregs cells) participate in the production of IL-10, and thus contribute to the anti-inflammatory effect and have a protective role [57]. It has been shown on animal models that transferring Breg cells in IL-10 deficient mice blocked the development of colitis [58].TRANSFORMATION GROWTH FACTOR βTGF-β as an inhibitor cytokine acts as a regulatory key for the inflammatory response. The decrease in TGF-β activity is considered responsible for the development of several autoimmune disorders, including IBDs [59]. Some evidence suggests that TGF-β acts by protecting the host tissues from the luminal aggressions and facilitates the repair of the mucosal lesions in IBD [60][61].OTHER CYTOKINES WITH ROLES IN IBDSOther cytokines, like IL-21 and IL-22, can play a role in the induction of IBDs as well as in other autoimmune diseases.INTERLEUKIN 21Interleukin 21 (IL-21) acts on the intestinal epithelium, maintaining the inflammation induced by Th1 by induction of INF-γ production [62][63]. It has been demonstrated that IL-21 increases the number of NK cells on a large scale [64]. IL-21 is expressed by T cells, B cells and non-immune cells, like fibroblasts. IL-21 contributes to the dif-ferentiation of Th17 cells [65], with an over expression in both CD and UC [50]. The highest levels were identified in CD [66]. IL-21 also induces IL-22 synthesis in CD4+ T cells [67].INTERLEUKIN 22Interleukin 22 (IL-22) acts on subepithelial myofibroblasts and increases the production of pro-inflammatory cytokines and matrix-degrading molecules [68]. IL-22 has a pro-inflammatory role and is increased in both CD and UC [50][69][70].Surprisingly, it was shown that IL-22 has a protective role in IBDs, as it attenuates the intestinal inflammation, inducing the production of a mucine membrane by goblet cells [69-71]. Administration of anti-IL-22 antibodies and IL-22 knockout mice has shown delayed healing of colonic injuries [72], while administration of IL-22 has led to a more rapid recovery in Th2-mediated colitis [70]. This makes IL-22 a promising future target for therapy.Gene mutations involved in encoding IL-22 and the IL-10R2 subunit of the IL-22R complex have been found to be associated with IBD [73][74]. Another recent paper showed that IL23R genotypes affect IL-22 serum concentrations, linking genetic CD susceptibility to Th17 function [69].CONCLUSIONSCytokines have an important role in the pathogenesis of IBDs. The identification of new cytokines, as well as the changing of the patho-genesis paradigms in IBDs has been done on animal tests and clinical studies. Thus, there is promising evidence basis for future therapy research based on cytokines and anti-cytokine antibodies. Non-Standard Abbreviations:CD – Crohn’s disease, UC – ulcerative colitis, IFN – interferon, IL – interleukin, NKT – natural killer T cell, TNF – tumor necrosis factor, DC – dendrite cells, Treg – regulatoryAl.C. Moldoveanu et al. 7 124T cells, Th – T helper cells, TGF-β – transforming growth factor-β; TL1A – TNF-like 1A, APC – antigen presenting cell, STNF-RI – TNF receptor type I, STNF-RII – TNF receptor type II, iNOS – inducible nitric oxide synthase, VEGF – vascular endothelial growth factor, DR3 – death domain-containing receptor 3.Bolile inflamatorii intestinale sunt afecţiuni cronice, caracterizate prin perioade de activitate şi perioade de remisiune. Inflamaţia este cel mai frecvent tipde răspuns al corpului uman ca mecanism de apărare împotriva agresorilor dinmediu. Frecvenţa şi gradul de inflamaţie depinde de aria ţesutului afectat. Tractulgastrointestinal este, de departe, cel mai susceptibil ţesut la răspuns inflamatordatorită expunerii constante la diverşi factori antigenici, mutagenici sau toxici.În bolile inflamatorii intestinale există o pierdere a toleranţei imune la flora intestinală care este mediată de diverse substanţe, inclusiv citokinele. Citokinelereprezintă un semnal cheie în răspunsul imun intestinal. Celulele dendriticeactivate şi macrofagele secretă citokine care intervin activ in reglarea inflamaţiei,atât în boala Crohn cât şi în colita ulcerativă. După secreţia lor de către celuleleprezentatoare de antigen, citokinele se activează şi diferenţiază celulele T,activând răspunsul imun dobândit.Citokinele au un rol important în patogeneza bolilor inflamatorii intestinale.Identificarea unor citokine noi precum şi schimbarea modelelor de patogeneză abolilor inflamatorii intestinale a fost făcută cu ajutorul testelor pe animale şi astudiilor clinice. Există dovezi promiţătoare pentru cercetarea în viitor a uneiterapii bazate pe citokine şi anticorpi anti-citokine.Corresponding author: Prof. Dr. Carmen Fierbinţeanu Braticevici,UMF “Carol Davila”E-mail: cfierbinteanu@The authors declare no conflict of interest.REFERENCES1. FIOCCHI C. Intestinal inflammation: a complex interplay of immune and nonimmune cell interactions. The American journal ofphysiology. 1997; 273(4 Pt 1):G769-75.2. FIOCCHI C. Immunity and inflammation: separate or unified? In: MacDermott RP, editor. Clinical Immunology inGastroenterology and Hepatology: from Bench to Bedside. New Orleans: American Gastroenterological Association; 1994; 182-8.3. BRANDTZAEG P, HALSTENSEN TS, KETT K, KRAJCI P, KVALE D, ROGNUM TO, et al. Immunobiology andimmunopathology of human gut mucosa: humoral immunity and intraepithelial lymphocytes. Gastroenterology. 1989;97(6):1562-84.4. GRISHAM MB, YAMADA T. Neutrophils, nitrogen oxides, and inflammatory bowel disease.Annals of the New YorkAcademy of Sciences. 1992; 664:103-15.5. FIOCCHI CB, DG. KATZ, JA. Cytokine production in the human gastrointestinal tract during inflammation. Current Opinionin Gastroenterology. 1994; 10(6):639-44.6. RAGHOW R. T he role of extracellular matrix in postinflammatory wound healing and fibrosis. FASEB journal: officialpublication of the Federation of American Societies for Experimental Biology. 1994; 8(11):823-31.7. SHIMIZU Y, SHAW S. Lymphocyte interactions with extracellular matrix.FASEB journal: official publication of theFederation of American Societies for Experimental Biology. 1991; 5(9):2292-9.8. LEON F, SMYTHIES LE, SMITH PD, KELSALL BL. Involvement of dendritic cells in the pathogenesis of inflammatorybowel disease. Advances in experimental medicine and biology. 2006; 579:117-32.9. SANCHEZ-MUNOZ F, DOMINGUEZ-LOPEZ A, YAMAMOTO-FURUSHO JK. Role of cytokines in inflammatory boweldisease. World journal of gastroenterology : WJG. 2008; 14(27):4280-8.10. XAVIER RJ, PODOLSKY DK. Unravelling the pathogenesis of inflammatory bowel disease. Nature. 2007; 448(7152):427-34.11. INCE MN, ELLIOTT DE. Immunologic and molecular mechanisms in inflammatory bowel disease. The Surgical clinics ofNorth America. 2007; 87(3):681-96.12. BREESE E, BRAEGGER CP, CORRIGAN CJ, WALKER-SMITH JA, MACDONALD TT. Interleukin-2- and interferon-gamma-secreting T cells in normal and diseased human intestinal mucosa. Immunology. 1993; 78(1):127-31.8 Cytokines in inflammatory bowel disease125 13. FUSS IJ, NEURATH M, BOIRIVANT M, KLEIN JS, DE LA MOTTE C, STRONG SA, et al. Disparate CD4+ lamina propria(LP) lymphokine secretion profiles in inflammatory bowel disease. Crohn's disease LP cells manifest increased secretion of IFN-gamma, whereas ulcerative colitis LP cells manifest increased secretion of IL-5. Journal of immunology. 1996;157(3):1261-70.14. STROBER W, FUSS IJ. Proinflammatory cytokines in the pathogenesis of inflammatory bowel diseases. Gastroenterology.2011; 140(6):1756-67.15. BAUMANN H, GAULDIE J. The acute phase respons e. Immunology today. 1994; 15(2):74-80.16. BEGUE B, WAJANT H, BAMBOU JC, DUBUQUOY L, SIEGMUND D, BEAULIEU JF, et al. Implication of TNF-relatedapoptosis-inducing ligand in inflammatory intestinal epithelial lesions. Gastroenterology. 2006;130(7):1962-74.17. STUCCHI A, REED K, O'BRIEN M, CERDA S, ANDREWS C, GOWER A, et al.A new transcription factor that regulatesTNF-alpha gene expression, LITAF, is increased in intestinal tissues from patients with CD and UC. Inflammatory bowel diseases. 2006; 12(7):581-7.18. REIMUND JM, WITTERSHEIM C, DUMONT S, MULLER CD, BAUMANN R, POINDRON P, et al. Mucosal inflammatorycytokine production by intestinal biopsies in patients with ulcerative colitis and Crohn's disease. Journal of clinical immunology. 1996; 16(3):144-50.19. YAMAMOTO-FURUSHO JK. Innovative therapeutics for inflammatory bowel disease. World journal of gastroenterology:WJG. 2007; 13(13):1893-6.20. SPOETTL T, HAUSMANN M, KLEBL F, DIRMEIER A, KLUMP B, HOFFMANN J, et al.Serum soluble TNF receptor I andII levels correlate with disease activity in IBD patients. Inflammatory bowel diseases. 2007; 13(6):727-32.21. MIZOGUCHI E, HACHIYA Y, KAWADA M, NAGATANI K, OGAWA A, SUGIMOTO K, et al. TNF receptor typeI-dependent activation of innate responses to reduce intestinal damage-associated mortality. Gastroenterology. 2008; 134(2):470-80. 22. BAMIAS G, MARTIN C, 3RD, MARINI M, HOANG S, MISHINA M, ROSS WG, et al. Expression, localization, andfunctional activity of TL1A, a novel Th1-polarizing cytokine in inflammatory bowel disease. Journal of immunology. 2003;171(9):4868-74.23. PREHN JL, MEHDIZADEH S, LANDERS CJ, LUO X, CHA SC, WEI P, et al.Potential role for TL1A, the new TNF-familymember and potent costimulator of IFN-gamma, in mucosal inflammation. Clinical immunology. 2004;112(1):66-77.24. KAMADA N, HISAMATSU T, HONDA H, KOBAYASHI T, CHINEN H, TAKAYAMA T, et al. TL1A produced by laminapropria macrophages induces Th1 and Th17 immune responses in cooperation with IL-23 in patients with Crohn's disease.Inflammatory bowel diseases. 2010; 16(4):568-75.25. PREHN JL, THOMAS LS, LANDERS CJ, YU QT, MICHELSEN KS, TARGAN SR. The T cell costimulator TL1A is inducedby FcgammaR signaling in human monocytes and dendritic cells. Journal of immunology. 2007; 178(7):4033-8.26. MICHELSEN KS, THOMAS LS, TAYLOR KD, YU QT, MEI L, LANDERS CJ, et al. IBD-associated TL1A gene (TNFSF15)haplotypes determine increased expression of TL1A protein. PloS one. 2009; 4(3):e4719.27. DINARELLO CA. The IL-1 family and inflammatory diseases. Clinical and experimental rheumatology. 2002; 20 (5 Suppl 27):S1-13.28. ASHWOOD P, HARVEY R, VERJEE T, WOLSTENCROFT R, THOMPSON RP, POWELL JJ.Functional interactionsbetween mucosal IL-1, IL-ra and TGF-beta 1 in ulcerative colitis.Inflammation research : official journal of the European Histamine Research Society [et a.l]. 2004; 53(2):53-9.29. MITSUYAMA K, TOYONAGA A, SASAKI E, ISHIDA O, IKEDA H, TSURUTA O, et al. Soluble interleukin-6 receptors ininflammatory bowel disease: relation to circulating interleukin-6. Gut. 1995; 36(1):45-9.30. REINISCH W, GASCHE C, TILLINGER W, WYATT J, LICHTENBERGER C, WILLHEIM M, et al. Clinical relevance ofserum interleukin-6 in Crohn's disease: single point measurements, therapy monitoring, and prediction of clinical relapse. The American journal of gastroenterology. 1999; 94(8):2156-64.31. SUZUKI A, HANADA T, MITSUYAMA K, YOSHIDA T, KAMIZONO S, HOSHINO T, et al. CIS3/SOCS3/SSI3 plays anegative regulatory role in STAT3 activation and intestinal inflammation.The Journal of experimental medicine. 2001;193(4):471-81.32. VAN KEMSEKE C, BELAICHE J, LOUIS E. Frequently relapsing Crohn's disease is characterized by persistent elevation ininterleukin-6 and soluble interleukin-2 receptor serum levels during remission. International journal of colorectal disease. 2000;15(4):206-10.33. ATREYA R, MUDTER J, FINOTTO S, MULLBERG J, JOSTOCK T, WIRTZ S, et al. Blockade of interleukin 6 transsignaling suppresses T-cell resistance against apoptosis in chronic intestinal inflammation: evidence in Crohn disease and experimental colitis in vivo. Nature medicine. 2000; 6(5):583-8.34. MUDTER J, NEURATH MF. Apoptosis of T cells and the control of inflammatory bowel disease: therapeutic implications.Gut. 2007; 56(2):293-303.35. KALLEN KJ. The role of transsignalling via the agonistic soluble IL-6 receptor in human diseases. Biochimica et biophysicaacta. 2002; 1592(3):323-43.36. YAMAMOTO M, YOSHIZAKI K, KISHIMOTO T, ITO H. IL-6 is required for the development of Th1 cell-mediated murinecolitis. Journal of immunology. 2000; 164(9):4878-82.37. CAREY R, JURICKOVA I, BALLARD E, BONKOWSKI E, HAN X, XU H, et al.Activation of an IL-6:STAT3-dependenttranscriptome in pediatric-onset inflammatory bowel disease. Inflammatory bowel diseases. 2008; 14(4):446-57.38. LEBEL-BINAY S, BERGER A, ZINZINDOHOUE F, CUGNENC P, THIOUNN N, FRIDMAN WH, et al. Interleukin-18:biological properties and clinical implications. European cytokine network. 2000; 11(1):15-26.39. LEACH ST, MESSINA I, LEMBERG DA, NOVICK D, RUBENSTEIN M, DAY AS. Local and systemic interleukin-18 andinterleukin-18-binding protein in children with inflammatory bowel disease. Inflammatory bowel diseases. 2008; 14(1):68-74.。

结直肠癌组织Tregs_占比和Foxp3、IL-10、TGF-β1_mRNA_表达变化及其意义

结直肠癌组织Tregs_占比和Foxp3、IL-10、TGF-β1_mRNA_表达变化及其意义

结直肠癌组织Tregs 占比和Foxp3、IL -10、TGF -β1 mRNA 表达变化及其意义马虎林1,孙晓霞2,陈良全1,张浩1,党利敏31 内蒙古自治区人民医院腹部肿瘤外科,呼和浩特 010010;2 内蒙古自治区人民医院外科门诊;3 内蒙古医科大学内蒙古临床学院摘要:目的 观察结直肠癌(CRC )组织中调节性T 细胞(Tregs )占比及叉头转录因子(Foxp3)、白细胞介素10(IL -10)、转化生长因子β1(TGF -β1)表达情况,并分析Tregs 占比与CRC 临床病理参数的关系。

方法 选取手术切除的CRC 组织及正常结肠组织各24例份,采用流式细胞术测算两组织中Tregs 占比,实时定量PCR 法检测两组织中Foxp3、IL -10、TGF -β1 mRNA 相对表达量,Spearman 相关分析法分析Tregs 占比与CRC 临床病理参数的关系。

结果 与正常结肠组织比较,CRC 组织Tregs 占比增加(P <0.05);与正常结肠组织比较,CRC 组织Foxp3、IL -10、TGF -β1 mRNA 相对表达量增加(P 均<0.05);Tregs 占比与CRC 肿瘤分期相关(P <0.05)。

结论 CRC 组织中Tregs 占比增加,Foxp3、IL -10、TGF -β1表达升高,Tregs 占比与患者肿瘤分期有关,Tregs 可能通过调节Foxp3、IL -10、TGF -β1发挥免疫逃逸作用。

关键词:结直肠癌;调节性T 细胞;叉头状转录因子;白细胞介素10;转化生长因子β1doi :10.3969/j.issn.1002-266X.2023.24.004中图分类号:R735.3 文献标志码:A 文章编号:1002-266X (2023)24-0017-04Proportion of Tregs in colorectal cancer and expression changes of Foxp3, IL -10 and TGF -β1 mRNA and their significanceMA Hulin 1, SUN Xiaoxia , CHEN Liangquan , ZHANG Hao , DANG Limin1 Abdominal Tumor Surgery Department , Inner Mongolia People's Hospital , Hohhot 010010, ChinaAbstract : Objective To explore the proportion of regulatory T cells (Tregs ) in colorectal cancer (CRC ) tissuesand the expression of Foxp3, interleukin 10 (IL -10), and transforming growth factor β1 (TGF -β1), and to analyze the rela‑tionships between the proportion of Tregs and the clinicopathological parameters of CRC. Methods Twenty -four CRC tis‑sues and 24 normal colon tissues were selected from surgical resection. Flow cytometry was used to detect the proportion of Tregs , and real -time quantitative PCR was used to detect the expression levels of Foxp3, IL -10 and TGF -β1 mRNA in these two groups. Spearman correlation analysis was used to analyze the relationships between the proportion of Tregs andthe clinicopathological parameters of CRC. Results Compared with the normal colon tissues , the proportion of Tregs inCRC tissues increased (P <0.05). Compared with the normal colon tissues , the relative expression of Foxp3, IL -10 and TGF -β1 mRNA in CRC tissue increased (all P <0.05). Tregs proportion was correlated with the stage of CRC (P <0.05).Conclusions The proportion of Tregs in CRC tissues increased , and the expression of Foxp3, IL -10 and TGF -β1 in‑creased. The proportion of Tregs was related to the tumor stage of patients , and Tregs played an immune escape role proba‑bly by regulating Foxp3, IL -10 and TGF -β1.Key words : colorectal carcinoma ; regulatory T cells ; Foxp3; interleukin -10; transforming growth factor -β1结直肠癌(CRC )是世界范围内最常见的恶性肿瘤之一,也是癌症相关死亡的主要原因之一[1]。

药学英语第五版原文翻译

药学英语第五版原文翻译

Introduction to PhysiologyIntroductionPhysiology is the study of the functions of living matter. It is concerned with how an organism performs its varied activities: how it feeds, how it moves, how it adapts to changing circumstances, how it spawns new generations. The subject is vast and embraces the whole of life. The success of physiology in explaining how organisms perform their daily tasks is based on the notion that they are intricate and exquisite machines whose operation is governed by the laws of physics and chemistry.Although some processes are similar across the whole spectrum of biology—the replication of the genetic code for or example—many are specific to particular groups of organisms. For this reason it is necessary to divide the subject into various parts such as bacterial physiology, plant physiology, and animal physiology.To study how an animal works it is first necessary to know how it is built. A full appreciation of the physiology of an organism must therefore be based on a sound knowledge of its anatomy. Experiments can then be carried out to establish how particular parts perform their functions. Although there have been many important physiological investigations on human volunteers, the need for precise control over the experimental conditions has meant that much of our present physiological knowledge has been derived from studies on other animals such as frogs, rabbits, cats, and dogs. When it is clear that a specific physiological process has a common basis in a wide variety of animal species, it is reasonable to assume that the same principles will apply to humans. The knowledge gained from this approach has given us a great insight into human physiology and endowed us with a solid foundation for the effective treatment of many diseases.The building blocks of the body are the cells, which are grouped together to form tissues. The principal types of tissue are epithelial, connective, nervous, and muscular, each with its own characteristics. Many connective tissues have relatively few cells but have an extensive extracellular matrix. In contrast, smooth muscle consists of densely packed layers of muscle cells linked together via specific cell junctions. Organs such as the brain, the heart, the lungs, the intestines, and the liver are formed by the aggregation of different kinds of tissues. The organs are themselves parts of distinct physiological systems. The heart and blood vessels form the cardiovascular system; the lungs, trachea, and bronchi together with the chest wall and diaphragm form the respiratory system; the skeleton and skeletal muscles form the musculoskeletal system; the brain, spinal cord, autonomic nerves and ganglia, and peripheral somatic nerves form the nervous system, and so on.Cells differ widely in form and function but they all have certain生理学简介介绍生理学是研究生物体功能的科学。

A case for regulatory B cells.

A case for regulatory B cells.

J Immunol. 2006 Jan 15;176(2):705-10.A case for regulatoryB cells. Mizoguchi A, Bhan AK.SourceImmunopathology Unit, Department of Pathology, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02114, USA.AbstractB cells are typically characterized by their ability to produce Abs, including autoantibodies.B细胞能产生抗体,包括自身抗体,为其显著特征。

However, B cells possess additional immune functions, including the production of cytokines and the ability to function as a secondary APC.然而,B细胞表现另一免疫功能,包括细胞因子的功能,以及作为次级抗原提呈细胞的功能。

As with T cells, the B cell population contains functionally distinct subsets capable of performing both pathogenic and regulatory functions.与T细胞一样,B细胞家族包含某些从功能上有区别的因子,这些因子表现出致病及调节功能。

Recent studies indicate that regulatory B cells develop in several murine models of chronic inflammation, including inflammatory bowel disease, rheumatoid arthritis, and experimental autoimmune encephalomyelitis.最近的研究发现调节性B细胞在一些慢性炎症的大鼠模型中形成,包括肠炎症疾病,风湿性关节炎,以及实验性自身免疫性脑脊髓炎。

肺癌患者血清中IL-12、IL-10水平测定及其临床意义

肺癌患者血清中IL-12、IL-10水平测定及其临床意义

肺癌患者血清中IL -12、IL -10水平测定及其临床意义陈婺徐美青梅新宇(安徽医科大学附属省立医院胸外科,安徽合肥230000)〔关键词〕肺癌;IL-12;IL-10〔中图分类号〕R734.2〔文献标识码〕A〔文章编号〕1005-9202(2012)09-1944-02;doi :10.3969/j.issn.1005-9202.2012.09.083通讯作者:徐美青(1962-),男,主任医师,硕士生导师,主要从事肺癌研究。

第一作者:陈婺(1981-),男,硕士,医师,主要从事肺癌分子生物学研究。

辅助性T (Th )细胞在机体免疫应答过程中具有重要调节作用。

细胞因子谱与细胞免疫功能有重要关系,正常情况下,细胞因子Th1和Th2处于相互制约、转化的平衡状态,当Th1/Th2细胞群发生亚状态,机体免疫力就会遭到破坏,发生病理反应,严重者会产生肿瘤〔1,2〕。

大多数肿瘤患者体内Th2细胞呈现优势漂移〔3,4〕。

在机体抗肿瘤免疫中,Th1类细胞因子起重要作用,肿瘤组织分泌Th2类细胞因子,成为肿瘤免疫逃逸机制之一〔5〕。

IL-12和IL-10是两种与Th 细胞功能密切相关的细胞因子,某些肿瘤患者血清IL-12和IL-10均下降〔6〕,这些细胞因子在原发性肺癌患者中的改变尚不完全清楚。

本研究拟检测原发性肺癌患者血清中IL-12和IL-10水平变化,以探讨其水平变化与免疫功能的关系及其临床意义。

1资料与方法1.1研究对象2011年5 8月我院住院原发性肺癌患者30例,男17例,女13例,年龄(51.3ʃ7.5)岁;其中鳞癌12例,腺癌18例。

健康体检者10例,均为健康体检者,男8例,女2例,年龄(47.1ʃ3.9)岁。

两组年龄及性别构成无显著差异,具有可比性。

1.2IL-12和IL-10的测定清晨空腹采患者肘静脉血5ml ,留取上层血清并分装,置-20ħ保存待测。

采用ELISA 法检测IL-12和IL-10水平,试剂盒购自武汉华美生物试剂公司,操作按说明书。

从Raji细胞克隆IL-10基因

从Raji细胞克隆IL-10基因
a oI t es q e c fh ma L 1 e ead tecd d c c 0d o t e u n eo u nI -0g n n o e DNA fh ma L 1 e ewa r lid h h o u n I-0 gn sa ̄ ie t ,ll 1 ] e协州 丘群
t r o n c swe cn e  ̄dⅥi MI1一 etra dtasome nocmp tn c lDI5 fEsh f hac it o sma tel ̄l- e h t p )8 T vco n rn fr dit o ee t e - ao e e e i oloe t 1 i n h ' on o
关 键词 I广0; ̄i克 隆 ;序 列 分析 L1 Sj;
中图分类号
Q a T5
文 献标识码

文章编号
0 1 — 6 120 ) ~1 6 — 2 5 7 6 1 ( 7 3 1 4 0 0 6 7
∞ l a0 嚷 0 1 f 0Gel . ll a R CI a ̄ m t mln
陈 劳 2 李 明 (河 农 大 ,南 州 其新 , 风学 , 1 南 业 学河 郑 , ;北 市 药 械 校北 01 2 京 医器 学 , 0) . 京1 2 o
摘要 [ 目的] 索一种 简便 可行的 白细胞介 素一 (L 0基 因的克隆 方法。[ 法]以人 Brt s 巴瘤 细胞 系 mj细胞 为材料 , 取 探 1 I 1) 0 广 方 ukt 淋 i’ i 提 培 养的 Ri细胞 中的 总 R A a i N 。根据 G nak中 Ⅱ 1基 因序 列设计 1 eBn 人 ,0 对特 异性 引物 , R- R方法扩 增人 I- 基 因 的编 码 eN 。 用 TP C L1 0 D A 回收 目的片段 , 将其 与 I 1-  ̄D 8 T载体 连接并转化到 大肠杆 菌感 受态细胞 D 5 , H a 构建该基 因的重组质粒。 重组质 粒经 和双酶切鉴 定 后 进行测序 。[ 结果]以骨髓细胞组织 总 R A为模板进 行 R -C , N TP R 可从 mj细胞 中获得 了预期 的约 50 p i 5 特异性条 带。成 功构建 了卫 b l 基 因的重 组质 粒 , E 0 P R扩增和 双酶切 的鉴定 结果说明该插入 片段 可能 为 I- N 。从获得 的阳性克隆 中挑选 1 茵落来分析 重组 LI e A OD 个 质 粒的插入 D A片段序 列。序列分析 结果证 实其与报道 的 I I基 因的序 列一 致。[ N LO - 结论 ] 该研 究为 I- 的重组表达及其 生物学活性 L1 0 分析奠定 了基础 。

间充质干细胞产品及其外泌体在炎症性肠病治疗中的研究进展

间充质干细胞产品及其外泌体在炎症性肠病治疗中的研究进展

间充质干细胞产品及其外泌体在炎症性肠病治疗中的研究进展杨婧雯1,陈 芊1,单云龙1,刘嘉莉1,尉 宁1,2,王 婧2,王广基1*,周 芳1**(1中国药科大学药物代谢动力学重点实验室, 南京 210009;2江苏睿源生物技术有限公司, 南京211103)摘 要 炎症性肠病(inflammatory bowel disease, IBD )发病机制不明,特征为进行性和终身复发性消化道炎症反应。

尽管现阶段新的治疗药物和策略不断涌现,但治疗作用局限于单一的抗炎功能,在复杂黏膜免疫环境下易出现耐药导致治疗失败。

间充质干细胞(mesenchymal stem cells, MSCs )能定向归巢到结肠炎症部位,具有强大的免疫调节能力,可重塑肠道免疫环境和修复上皮屏障,为药物难治性患者的治疗提供了极具潜力的替代方案。

本文对MSCs 产品及其衍生的外泌体在临床上的应用、作用机制和工程化进行综述,以期为MSCs 及其外泌体产品用于IBD 的治疗提供参考。

关键词 炎症性肠病;间充质干细胞;外泌体;工程化中图分类号 R574 文献标志码 A文章编号 1000−5048(2024)01−0103−12doi :10.11665/j.issn.1000−5048.2023113001引用本文 杨婧雯,陈芊,单云龙,等. 间充质干细胞产品及其外泌体在炎症性肠病治疗中的研究进展[J]. 中国药科大学学报,2024,55(1):103 − 114.Cite this article as: YANG Jingwen, CHEN Qian, SHAN Yunlong, et al . Research progress on mesenchymal stem cell products and their exosomes in the treatment of inflammatory bowel disease[J]. J China Pharm Univ , 2024, 55(1): 103 − 114.Research progress on mesenchymal stem cell products and their exosomes in the treatment of inflammatory bowel diseaseYANG Jingwen 1, CHEN Qian 1, SHAN Yunlong 1, LIU Jiali 1, WEI Ning 1,2, WANG Jing 2, WANG Guangji 1*,ZHOU Fang 1**1Key Laboratory of Drug Metabolism and Pharmacokinetics, China Pharmaceutical University, Nanjing 210009;2Renocell Biotechnology Co., Ltd., Nanjing 211103, ChinaAbstract Inflammatory bowel disease (IBD), whose pathogenesis remains elusive, is a group of autoimmune diseases characterized by chronic, progressive, and lifelong inflammation of the digestive tract. The pathogenesis of IBD remains elusive. Although a number of drugs have been developed to treat IBD, their effects are merely anti-inflammatory. In addition, current treatments for IBD are easily susceptible to resistance in clinical practice.Mesenchymal stem cells (MSCs) have been reported to have the ability to migrate to the site of inflammation,with potent immunoregulatory effects, and to rebalance the immune microenvironment and restore the integrity of the epithelial barrier with significant value of application, particularly for patients who are refractory to classic medicines. In this paper, we reviewed the clinical applications, mechanisms and engineerable properties of MSC products and their exosomes to provide some reference for the use of MSCs and their exosomes in the treatment of IBD.Key words inflammatory bowel disease; mesenchymal stem cells; exosomes; engineering收稿日期 2023-11-30 通信作者 *Tel :************ E-mail :*************************Tel :************ E-mail :**************基金项目 国家自然科学基金项目(No. 82073928);南京市生命健康科技专项(No.202110006);细胞生态海河实验室创新基金项目(22HHXBSS00005);江苏省南京市联合资助项目(SBK2023070039)学报 2024, 55(1): 103 − 114103This study was supported by the National Natural Science Foundation of China (No. 82073928); the Nanjing Life and Health Science and Technology Program (No.202110006); the Haihe Laboratory of Cell Ecosystem Innovation Fund (22HHXBSS00005); and the Co-funded Programs in Nanjing, Jiangsu Province (SBK2023070039)炎症性肠病(inflammatory bowel disease, IBD)包括溃疡性结肠炎(ulcerative colitis, UC)和克罗恩病(Crohn's disease, CD),是由环境、免疫系统、肠道微生物组和个体遗传等因素的复杂互作引起的一类自身免疫性疾病,其一线治疗药物包括糖皮质激素、免疫抑制剂、抗生素和抗肿瘤坏死因子-α(tumor necrosis factor, TNF-α)疗法。

IL-10生物学作用

IL-10生物学作用

IL-10生物学作用1、简介在1989年,mosmannand及他们的同事描述了一种新的免疫介质,由th2细胞克隆分泌,能够抑制th1细胞克隆il-2和ifnr的合成。

早期被命名为细胞因子合成抑制因子(csif),这种因子后来被命名为il-10,在这被发现的21年期间,很多研究深入剖析了这个细胞因子的生物学特性2、il-10基因和蛋白人类il-10基因位于1号染色体上,包括总共5.1kb的序列包含5个外显子,il-10基因的启动子中有很多的snp位点,一些证据表明这些基因多态性在体外会影响il-10的表达,il-10基因由178个氨基酸组成蛋白质,分泌时会被切去18个氨基酸的信号肽,人类和鼠类的il-10约有75%的氨基酸序列是一致的,wlodawer和walter小组用x射线解决了il-10的晶体结构,有趣的是il-10的结构与ifnr的结构类似,人il-10是一个35kd的二聚体由两个单体通过非共价键形式结合,二聚体有两个v型的结构域,每个结构域包含六个螺旋结构,其中a-d属于一个单体,另外两个(e0和f0)属于另外一个单体。

在单体内有两个二硫键(分别是c30-c126和c80-c132)来维持因子结构和生物学活性。

除了各种不同的哺乳动物il-10相关分子外,还有四种病毒il-10类似物,由eb病毒,马疱疹病毒2型,口疮病毒和巨细胞病毒产生,除了cmv病毒的il-10,其它病毒与细胞分泌的的il-10氨基酸序列结构相似,比如,eb 病毒的il-10氨基酸序列与人il-10序列有83%相同,除了一些微小的变异外主要的不一致集中在n末端,因此导致两者结构非常类似。

病毒il-10的表达似乎是在病毒感染的细胞裂解期,病毒因子似乎也通过相同的il-10受体来作用,和人类il-10相比,多数病毒il-10的作用只有其效力的1/1000,但不幸的是,多数的抗人il-10抗体和elisa试剂不能区别两者,最近,认识到新的一些类似于il-10人分子结构,这些细胞因子被统称为il-10家族,包括il-10,il-19,il-20,il-22,il-24,和il-26。

一种白细胞介素10的突变体及其制备方法和应用

一种白细胞介素10的突变体及其制备方法和应用

一种白细胞介素10的突变体及其制备方法和应用白细胞介素10(IL-10)是一种抗炎性细胞因子,在调节免疫反应和维持免疫平衡方面发挥重要作用。

然而,IL-10的生物活性和稳定性受到很多限制,包括短半衰期和低活性。

因此,寻找新型的IL-10突变体,以提高其生物活性和稳定性,对于开发更有效的免疫治疗药物具有重要意义。

最近的研究表明,通过突变IL-10的氨基酸序列,可以获得具有更强生物活性和更长半衰期的IL-10突变体。

这些突变体可能通过增加受体结合亲和力、提高信号转导效率或改善蛋白稳定性等方式来增强其生物活性。

本文将介绍一种IL-10的突变体及其制备方法和应用。

一、IL-10突变体的设计在设计IL-10的突变体时,可以考虑以下几个方面:1.增加受体结合亲和力:通过突变IL-10的氨基酸序列,可以增加其与受体的结合亲和力,进而增强其生物活性。

例如,可以通过引入一些关键氨基酸突变,增强IL-10与受体之间的相互作用,提高其受体结合亲和力。

2.提高信号转导效率:IL-10通过其受体激活JAK-STAT信号通路,抑制炎症因子的产生。

可以设计突变体,增强其激活JAK-STAT信号通路的效率,从而增强其免疫调节功能。

3.改善蛋白稳定性:IL-10在体内易被降解,导致其生物活性较低。

可以通过改善其蛋白稳定性,延长其半衰期,提高其生物活性。

通过对IL-10突变体的设计,可以获得具有更强生物活性和更长半衰期的IL-10突变体,从而开发更有效的免疫治疗药物。

二、IL-10突变体的制备方法IL-10突变体的制备方法通常包括以下几个步骤:1.基因工程:通过基因工程技术,将设计好的IL-10突变体的氨基酸序列插入表达载体中。

可以利用大肠杆菌等表达宿主,进行大规模的突变体蛋白生产。

2.表达和纯化:将表达载体转染至表达宿主细胞中,利用不同的表达条件和纯化方法,获得突变体蛋白。

3.结构分析:通过质谱、核磁共振等技术,对蛋白的结构和功能进行分析,验证IL-10突变体的设计效果。

白细胞介素-10基因敲除小鼠血液生理生化指标分析

白细胞介素-10基因敲除小鼠血液生理生化指标分析

1 . 1 实 验 动 物 纯 合 型 I L一1 0 K O小鼠( 1 2 9一 I L l 0 / J ) 及对照组 I L一1 0 WT小 鼠 ( C 5 7 B L / 6 ) , 雄 性, 均 购 自美 国 J a c k s o n L a b o r a t o r y公 司。小 鼠的饲 养 和繁殖按照无特 定病原体 ( s p e c i i f c p a t h o g e n f r e e ) 级 动
K O小 鼠和 6只 I L一1 0 wT小 鼠 , 空腹 1 2 h , 用 摘 除眼 球法采 血 , 每 只取血 0 . 8 m L , 装入E p p e n d o f管 中, 室温 静置 3 0 m i n , 3 0 0 0 r / m i n离心 1 0 m i n , 分离 血清 , 用 日 立A U 5 4 0 0全 自动生化 分析 仪 , 美 国罗 氏诊 断产 品有
分析 白细胞介素 一1 0 ( I L一1 0 ) 基 因敲 除小鼠和野生型 小鼠血液生理 生化指标差异 。方 法
与野生型小鼠比较 , I L一1 0基 因敲 除小鼠血液生理指标 中红细胞参数 明显 降低 , 血小
用摘除眼球法采集 I L一1 0基 因敲除小鼠和野生型小鼠血液 , 用全 自动 生化分析仪及血 细胞分析仪检 测常 用血液
影响 小鼠部分的血液 、 血清生化指标。
I L一1 0基 因缺失 可
【 关键 词】 白细胞介素 一 1 0 ; 基 因敲除 ; 血液学 ; 生化标记物
白细胞介素 一1 0 ( i n t e r l e u k i n一 1 0 , I L一1 0 ) 是一 种 分子量相 当于 1 8~2 1 k D的细胞 因子 , 属于 T辅 助 细 胞 2类因子 ( T h 2 ) , 由各种 淋 巴细 胞产生 , 能抑 制多 种 炎性 细胞产生各 种炎性 细胞 因子 , 在下调 炎症反应 中 发挥关键作用… , 是 目前非常受关注 的抗炎介质 之一 。 建立 I L一1 0基因敲除 ( k n o c k o u t , K O) 动物 模型对深 入 阐明 I L一1 0的功 能和 致病 机 制 有 十分 重 要 的意 义 。 目前该小 鼠在 国外 的报道 主要是集 中于它们 在动物模 型中的应 用 , 以及这 些 基 因在 机体 中各 种 功 能 的研 究 , 而它们 的生理生化 等部分 生物 学特性 未见相 关 报 道。2 0 1 2年 3 —1 2月 , 本 实验选 取 I L一1 0 K O小 鼠 作 为实验组 , 以I L一1 0野生 ( I L一1 0 w i d e t y p e , I L一1 0 wT ) 小 鼠作为对照组 , 对其 血液生 理指标 和血 清生化 指标进行检测分析 , 为研究 I L一1 0相关疾病 提供部分

B细胞及其新型免疫调节功能研究

B细胞及其新型免疫调节功能研究

Is neonatal Breg activity associated with a particular subset ?
20.7
79.3
CD5
0.26
CD10
Adu B
CD5
Is neonatal Breg activity associated with a particular subset ?
2005
Newborn mice SLE in PN mice
2007
CHS in CD19-/- mice CIA in DBA/1 mice CHS in CD19-/- and hCD19Tg mice SLE in MRL/lpr mice EAE in C57BL/6 mice Allergy and EAE in BALB/c et C57BL/6 mice Allergy in BALB/c and C57BL/6 mice EAE in C57BL/6 mice SLE in NZB/W mice Ulcerative colitis in C57BL/6 mice AIA dans des souris µMT/B6 mice
CD40 ligation
? ?
IL-10
B2
T2-MZP
T2-MZP
IL-10
Zhang, CMI, 2013
Breg classification
“Innate” Bregs
TLRLs/microbes
Innate-like B cellderived Bregs
IgM
ILBs
ILBs
IgM
“Adaptive” Bregs
IL-10
T CD4+

小鼠IL-10基因的克隆

小鼠IL-10基因的克隆

质粒 pT-IL-10
原核表 达载体 pET28a
双酶切、回收片段、连接
BL21宿主菌
转 化
pETIL-10
பைடு நூலகம்PTG诱导剂量为1.5μL/mL和 2.0μL/mL的pETIL-10
IPTG诱导剂量为 1μL/mL的pETIL-10
BL21菌 未诱导
转染重组pETIL10诱导的BL21的 免疫印记
根据总 RNA 提取试剂盒说明
P1:5′GGATCCATGCCTGGCTCAGCACTGCTATGCTGCCTGCTCTT3′ 原核表达载体 pET2IL210 的构建 P2:5′AAGCTTTTAGCTTTTCATTTTGATCATCATGTATGC3′
IL210 在大肠杆菌中的表达及其检测 酶切位点:BamH Ⅰ HindⅢ
10mg/L 刀豆素 ConA 以及 1% 双抗(终浓度为 100 μ g/mL 青霉素、链霉素 )进行培养。
pMD18-T
质粒 pT-IL-10
大肠杆菌JM109感受态细胞
含Amp 100 g/mL 的LB 培养基 37 ℃振荡培养 16h 提取质粒 用限制性内切酶(BamHⅠ、Hind Ⅲ) 酶切和 PCR 方法鉴定 获得重组质粒 命名为pT-IL-10
技术路线
脾细胞的制备 总 RNA 的提取 引物的设计与合成
根据 GenBank 中报道的小鼠 (登录号NM-010548 ) IL210mRNA 序列设计特异性引物 P1 和 P2,预期扩增长度为 PCR 产物凝胶回收克隆及序列测定 于37 ℃下用MEM培养液( 8%的犊牛血清, 537bp。
IL210 基因的 RT-PCR 扩增
M
原核表达 载体 pET28a

免疫抑制因子名词解释

免疫抑制因子名词解释

免疫抑制因子名词解释英文回答:Immunosuppressive factors refer to substances or mechanisms that inhibit or suppress the immune response in the body. These factors can be endogenous (produced by the body itself) or exogenous (introduced from external sources). They play a crucial role in regulating the immune system and preventing excessive immune responses that can lead to autoimmune diseases or tissue damage.There are various types of immunosuppressive factors, including cytokines, chemokines, regulatory T cells, and immunosuppressive drugs. Cytokines are small proteins secreted by immune cells that regulate immune responses. Some cytokines, such as interleukin-10 (IL-10) and transforming growth factor-beta (TGF-β), have immunosuppressive effects by inhibiting the activation and function of immune cells.Chemokines are another type of immunosuppressive factor that regulate the migration and recruitment of immune cells. Some chemokines, such as CCL17 and CCL22, attractregulatory T cells (Tregs) to the site of inflammation, where they exert immunosuppressive effects by suppressing the activation of other immune cells.Regulatory T cells, also known as Tregs, are a specialized subset of T cells that play a crucial role in maintaining immune homeostasis and preventing excessive immune responses. They suppress the activation and function of other immune cells, such as T helper cells and cytotoxic T cells, through various mechanisms, including thesecretion of immunosuppressive cytokines like IL-10 andTGF-β.Immunosuppressive drugs are pharmacological agents used to suppress the immune response in various medical conditions, such as organ transplantation and autoimmune diseases. Examples of immunosuppressive drugs include corticosteroids, calcineurin inhibitors (e.g., cyclosporine), and biological agents (e.g., monoclonalantibodies targeting specific immune cells or cytokines).In summary, immunosuppressive factors are substances or mechanisms that inhibit or suppress the immune response. They play a crucial role in regulating the immune system and preventing excessive immune responses. These factors include cytokines, chemokines, regulatory T cells, and immunosuppressive drugs.中文回答:免疫抑制因子是指抑制或压制机体免疫反应的物质或机制。

调节性B细胞对肿瘤发展促进作用的研究进展

调节性B细胞对肿瘤发展促进作用的研究进展

调节性B细胞对肿瘤发展促进作用的研究进展欧洁;吴红艳【摘要】调节性B细胞(Bregs)是新近发现的B细胞子集,其在抑制肿瘤免疫和促进肿瘤细胞的增殖和转移中起重要作用.Bregs可通过分泌IL-10、IL-35和TGF-β等细胞因子抑制多种细胞亚型,从而抑制抗肿瘤免疫应答.【期刊名称】《基础医学与临床》【年(卷),期】2019(039)001【总页数】5页(P110-114)【关键词】调节性B细胞;肿瘤;细胞因子【作者】欧洁;吴红艳【作者单位】三峡大学医学院免疫系,湖北宜昌443002;三峡大学医学院免疫系,湖北宜昌443002【正文语种】中文【中图分类】R730.3以往B细胞被认为是产生特异性抗体和呈递抗原而促进免疫效应的成分,近年的研究发现部分B细胞子集可以起到免疫负调节作用,该类细胞被称为调节性B细胞(regulatory B cells,Bregs)。

关于该类细胞的研究涉及寄生虫感染、免疫性疾病和肿瘤等方面[1-2]。

近年来,Bregs在促进肿瘤逃逸和生长的作用日益受到关注。

它能通过分泌白介素10(interleukin 10,IL-10)、白介素35(interleukin 35,IL-35)和转化生长因子-β(transforming growth factor-β,TGF-β)等,以及调节骨髓源性抑制细胞(myeloid-derived suppressor cells,MDSC)和中性粒细胞等的功能,抑制抗肿瘤免疫效应细胞、促进肿瘤细胞增殖和转移。

本文简要综述Bregs促进肿瘤发展的机制。

1 与肿瘤相关的Bregs的基本特征与肿瘤相关的Bregs主要来源于肿瘤微环境诱导B细胞转化。

肿瘤微环境通过多种途径引起B细胞获得负调节功能和扩增。

其部分机制包括肝细胞癌(hepatocellular carcinoma, HCC)中肿瘤诱生的单核细胞作用于树突细胞(dendritic cells, DCs),以CD95L依赖的途径诱导B细胞FcγRII(也称为CD32)表达降低(不同于正常肝脏的FcγRII相对高表达),而获得、激活和维持IL-10-Bregs的生存;Toll样受体(toll-like receptors, TLR)激动剂Pam3 CysSK4、脂多糖(lipopolysaccharide)、透明质酸(hyaluronan)片段和CpG寡脱氧核苷酸(CpG oligodeoxynucleotide, CpG ODN)等激活PI3K/AKT、MAPK和NFκB通路,阻碍STAT6(signal transducers and activators of transcription 6,STAT6)的激活,进一步增加B细胞中的BCL6(B-cell lymphoma 6, BCL6)的表达而增加PD-1(program-med death 1,PD-1)的表达;另外,聚集于肿瘤的巨噬细胞或者单核细胞通过激活CD40通路并以细胞接触的方式呈递IL-15于B细胞和黑色素瘤分泌IL-35对B细胞作用等[3-7]途径促进Bregs的增殖分化。

B10细胞在器官移植术后免疫耐受中的研究现状和前景

B10细胞在器官移植术后免疫耐受中的研究现状和前景

B10细胞在器官移植术后免疫耐受中的研究现状和前景吕少诚;白纯;李先亮【摘要】器官移植术后的理想目标是达到移植物的免疫耐受状态,相关研究显示调节性B细胞(regulatory B cells,Bregs)在诱导器官移植术后免疫耐受中有积极作用,但起主导作用的细胞表型和具体机制尚无共识.近年来B10细胞的发现进一步揭示了调节性B细胞诱导器官移植术后免疫耐受的相关机制,并有望最终诱导免疫耐受状态.本文就B10细胞的研究现状及其在器官移植免疫耐受中的作用、前景进行综述.【期刊名称】《解放军医学院学报》【年(卷),期】2016(037)011【总页数】3页(P1183-1185)【关键词】B10细胞;器官移植;免疫耐受【作者】吕少诚;白纯;李先亮【作者单位】首都医科大学附属北京朝阳医院肝胆外科,北京100020;首都医科大学附属北京朝阳医院肝胆外科,北京100020;首都医科大学附属北京朝阳医院肝胆外科,北京100020【正文语种】中文【中图分类】R617B淋巴细胞起源于机体的骨髓造血干细胞,主要参与机体的体液免疫,并产生特异性的抗原抗体,同时还可以表达共刺激分子、产生细胞因子或作为抗原提呈细胞而调节T细胞对内源性或外源性抗原的免疫应答。

过去的研究认为,B淋巴细胞是机体体液免疫的起源,主要起促进机体炎症反应的作用[1]。

而近年来越来越多的研究表明,在众多不同表型的B淋巴细胞中同时存在一类对免疫应答具有负向调节作用的B淋巴细胞,被称为调节性B细胞[2]。

B10细胞就是众多调节性B细胞中的一种,其在机体的自身免疫性疾病、炎症性疾病、肿瘤疾病和器官移植免疫耐受等方面是潜在的治疗靶点[3-4]。

本文就B10细胞在器官移植免疫耐受中可能作用的研究现状做一综述。

随着新型免疫抑制药物的出现和移植技术的不断进步,器官移植术后手术死亡率和急性排斥反应发生率都已大大降低,目前慢性排斥反应导致的移植物失活成为影响器官移植术后长期效果的首要原因[5]。

调节性B细胞与肿瘤免疫

调节性B细胞与肿瘤免疫

调节性B细胞与肿瘤免疫杨绍臻;杜国盛【摘要】调节性B细胞(Breg)是继调节性T细胞后免疫领域研究的新热点,Breg是通过白细胞介素(IL)-10、IL-35,转化生长因子(TGF)-β产生免疫抑制作用的一类细胞。

Breg通过阻止致病性T细胞和其他促炎性淋巴细胞扩增,从而发挥抑制免疫病理损伤作用。

研究已证实,Breg在炎症、自身免疫性疾病以及癌症进展、转归和预后中发挥一定作用。

本文就Breg与癌症的关系及其研究进展作一综述。

【期刊名称】《癌症进展》【年(卷),期】2016(014)004【总页数】4页(P296-299)【关键词】癌症;B细胞;调节性B细胞;肿瘤免疫【作者】杨绍臻;杜国盛【作者单位】解放军第三〇九医院器官移植研究所肝胆外科,北京1000910;解放军第三〇九医院器官移植研究所肝胆外科,北京1000910【正文语种】中文【中图分类】R730.3B细胞作为产生免疫球蛋白浆细胞的祖细胞,可以将抗原呈递给T细胞、iNKT细胞,并且产生Th1和Th2细胞因子以促进其他淋巴细胞的激活和分化。

研究表明,在某些环境中,有一类特殊的具有调节功能的B细胞亚群对控制炎症进展发挥一定作用,这一类B细胞被称作调节性B细胞(regulatory B cell,Breg)[1-2]。

Breg可通过产生白细胞介素(IL)-10和(或)IL-35,转化生长因子(TGF)-β等细胞因子,进而产生免疫抑制作用[3-4]。

将产生IL-10的Breg称为Br1细胞,产生TGF-1的Breg称为Br3细胞[5-6]。

早在1970年Katz等[7]提出假说,认为B细胞可以通过产生抗体来阻止超敏反应发生。

随后在20世纪80年代中期,有研究报道B细胞与抗原特异性抑制性T细胞产生有关。

1996年,Wolf等[8]在B细胞缺陷小鼠(μMT)和野生型小鼠(WT)中分别诱导实验性自身免疫性脑脊髓炎(experimental autoimmune encephalomyelitis,EAE)以建立其小鼠模型时发现,前者会产生疾病进展倾向,而后者有部分自愈倾向。

  1. 1、下载文档前请自行甄别文档内容的完整性,平台不提供额外的编辑、内容补充、找答案等附加服务。
  2. 2、"仅部分预览"的文档,不可在线预览部分如存在完整性等问题,可反馈申请退款(可完整预览的文档不适用该条件!)。
  3. 3、如文档侵犯您的权益,请联系客服反馈,我们会尽快为您处理(人工客服工作时间:9:00-18:30)。

EditorialRegulatory B cells that produce IL-10:A breath of fresh air in allergic airway diseaseThomas F.Tedder,PhD,and Takashi Matsushita,MD,PhD Durham,NCKey words:Asthma,IL-10,regulatory B cellIn this issue of the Journal,Amu et al1demonstrate a significant role for IL-10–producing regulatory B(Breg)cells during murine models of allergic airway inflammation.The authors identified a Breg cell subpopulation that expands in vivo and in vitro in re-sponse to parasitic Schistosoma mansoni worm infection.The adoptive transfer of these Breg cells into allergen-sensitized mice suppresses anaphylaxis and allergen-induced airway hyper-responsiveness through IL-10–dependent mechanisms.1,2These importantfindings expand the clinical significance of studies showing that IL-10–competent Breg cells dramatically regulate inflammation and autoimmunity in murine models of contact hy-persensitivity,3experimental autoimmune encephalomyelitis,4,5 collagen-induced arthritis,6and inflammatory bowel disease.7 These studies focus on a relatively rare IL-10–competent murine B-cell subset that represents only1%to2%of splenic B cells in naive wild-type mice.8We call these cells‘‘B10cells’’because IL-10secretion is universally recognized as their mechanism of regu-latory function,they only produce IL-10transcripts,3,9and multiple other B-cell subsets with regulatory properties are likely to exist. B10cells are predominantly contained within a phenotypically unique CD1d hi CD51CD19hi B-cell subpopulation that normally represents only2%to7%of splenic B cells.3B10cells appear to be functionally mature because they can be identified based on cy-toplasmic IL-10expression after only5hours of in vitro stimulation with phorbol12-myristate13-acetate and ionomycin.B10progen-itor(B10pro)cells have also been functionally identified within the splenic CD1d hi CD51B-cell subpopulation,but these cells require 48hours of in vitro stimulation through CD40or with LPS before they acquire the ability to express cytoplasmic IL-10after5hours of phorbol12-myristate13-acetate and ionomycin stimulation.9,10 Therefore purifying splenic CD1d hi or CD1d hi CD51B cells enriches for functionally potent B10and B10pro cells that can be adoptively transferred into recipient mice to shift the normal balance of regulatory networks toward a more immunosuppressive phenotype.Parasitic infections with S mansoni worms in the current study drives both B-cell and B10-cell expansion in mice(Fig1).1Transferring splenic CD1d hi B cells from worm-infected mice into ovalbumin(allergen)–challenged recipients inhibits both acute and established airway inflammation.Most likely,B10cells expand more than other B cells in vivo because they proliferate more vigorously in response to polyclonal mitogens when com-pared with non-B10cells.9Because antigen-specific B10cells are required to inhibit contact hypersensitivity and autoimmu-nity,3,5it is unlikely that worm antigen-specific B10cells would inhibit ovalbumin-driven disease.Thus it will be important to de-termine whether S mansoni worms are driving polyclonal, antigen-specific,or cross-reactive B10-and B10pro-cell expan-sion/maturation.It will also be important to determine whether worm-driven B10-cell expansion can regulate contact hypersensi-tivity and autoimmunity.Regardless,helminth-driven B10-cell expansion supports the‘‘hygiene hypothesis,’’whereby a de-crease in helminth infections within a population is proposed to increase allergic disease incidence.11Thus B10-cell function and relative frequencies might also be important factors contrib-uting to human allergic diseases.The authors propose to have identified a distinct IL-101CD1d hi CD21hi CD231IgD1IgM hi CD191splenic Breg cell subpopulation.However,there are currently no cell-surface markers that uniquely delineate all IL-10–competent B10cells or B10pro cells.Rather,the ability of B10cells to produce IL-10is the single functional marker that unifies most current studies and identifies a population of cells with a fairly homogenous cell-surface phenotype.3Isolating B cells based on IL-10expression alone is technically problematic because this selects for either IL-10–secreting cells or cytoplasmic IL-101cells that must be permeabilized,whereas functionally important B10pro cells are lost with these methods.Moreover,IL-10competence is most frequently measured after phorbol12-myristate13-acetate and ion-ophore stimulation in vitro.As shown by Amu et al,1single markers,such as CD1d hi,CD21hi,CD231,IgD low,or IgM hi,could be used to enrich for B10cells,but they also exclude a substantial proportion of functionally competent B10cells that are then diluted within the remaining nonselected B-cell population.IL-10–compe-tent B cells found within other murine tissues also differently express some of these cell-surface markers.9Despite these techni-cal issues,most studies within the evolving Breg cellfield are likely to be examining the same rare and functionally unique B10-and B10pro-cell subset that regulates immune responses through the production of IL-10.B cells contribute to asthma pathogenesis by producing IgE.12 However,Amu et al1also identified B-cell subsets that either ex-acerbated or regulated allergic airway inflammation(Fig1).Al-though most B cells express CD1d,a splenic CD1d low B-cell subset was expanded in helminth-infected mice.Asthma was ex-acerbated when these CD1d low B cells were adoptively trans-ferred into allergen-sensitized mice.Although it was not determined whether these B cells contributed to IgE production,From the Department of Immunology,Duke University Medical Center.Supported by National Institutes of Health grants(AI057157and AI56363).Disclosure of potential conflict of interest:T.F.Tedder has consultant arrangements withMedImmune and has consultant arrangements with and is a shareholder in AngelicaTherapeutics.T.Matsushita declares no conflicts of interest.Received for publication March22,2010;accepted for publication March24,2010.Reprint requests:Thomas F.Tedder,PhD,Box3010,Department of Immunology,Room353Jones Bldg,Research Dr,Duke University Medical Center,Durham,NC27710.E-mail:thomas.tedder@.J Allergy Clin Immunol2010;125:1125-7.0091-6749/$36.00Ó2010American Academy of Allergy,Asthma&Immunologydoi:10.1016/j.jaci.2010.03.0241125the future characterization of these cells might reveal a novel B-cell subset that preferentially contributes to disease pathogenesis through unknown mechanisms.Distinct B-cell subsets with op-posing pathogenic and negative regulatory functions have also been observed during experimental autoimmune encephalomye-litis pathogenesis in mice.5Mature B-cell depletion with CD20mAb before experimental autoimmune encephalomyelitis induc-tion exacerbates subsequent disease,whereas B-cell depletion during experimental autoimmune encephalomyelitis progression dramatically reduces disease symptoms.Exacerbated autoim-mune disease results from B10-cell depletion before disease ini-tiation,which is ameliorated by the adoptive transfer of splenic CD1d hi CD51B cells.B10cells and other B cells have also been found to have opposing protective and pathogenic functions during murine models of systemic lupus erythematosus,respec-tively.13,14Therefore different B-cell subsets might display op-posing protective and pathogenic functions during human asthma because B-cell depletion might improve atopic eczema.15Thus future murine and patient studies are needed to further un-cover the likely complexities of B-cell function during different stages of airway immunopathology.Amu et al 1demonstrate that IL-10production by CD1d hi B cells is required to observe their regulatory effects,and they show that S mansoni –infected CD1d-deficient mice are highly susceptible to allergic airway inflammation.However,it remains essential to determine whether B-cell CD1d expression is re-quired for B10-or B10pro-cell function because CD1d expression is not required for B10-cell development.9Multiple leukocytelineages and subsets produce IL-10,and the mechanisms by which IL-10can inhibit or augment immune responses are equally complex.However,Amu et al 1propose that adoptively transferred CD1d hi B cells suppress airway inflammation by inducing natural forkhead box protein 3–positve CD41regulatory T (Treg)cell recruitment into the lungs,where they suppress lung inflammation (Fig 1).Whether B10cells must enter the lung to induce these changes or whether they can exert these effects distally remains unknown.It is also unknown whether B10cells actually control Treg cell migration or whether enhanced Treg cell emigration into the lung is an indirect consequence of reduced inflammation.Nonetheless,Treg cell numbers are significantly decreased in CD19-deficient NZB/W mice that have few B10cells,whereas wild-type CD1d hi CD51B cells transferred into CD192/2NZB/W mice induce Treg cell expansion.14These independent studies suggest a potential link between B10-cell function and Treg cell frequencies that needs to be explored.Multiple laboratories have demonstrated that Breg cells are functionally significant in diverse diseases.It will be important to determine whether other parasites and infectious agents also drive B10-cell expansion as a potential mechanism for reducing host immune responses.This will further open the door for identifying B10cell–directed therapies.Turning these laboratory observations into therapeutic targets for modulating immune responses and pathology will be a significant but important challenge for the future.We thank Drs Cynthia Magro,David DiLillo,Michelle Schweitzer,Jonathan Poe,and Susan Smith for theircomments.FIG 1.IL-10–producing regulatory B10cells inhibit allergic airway disease.Splenic B10pro cells mature into antigen-specific CD1d hi CD51regulatory B10cells that are competent to produce and secrete IL-10in response to allergen (antigen)challenge.Amu et al 1show that splenic B10pro/B10cells are induced to mature/expand in response to S mansoni worm infection.The subsequent adoptive transfer of CD1d hi B cells purified from infected mice into allergen-sensitized recipients suppressed the induction of acute and allergic airway inflammation,which is proposed to result from the recruitment of Treg cells into the lungs of challenged mice.From our studies,B10cells are also known to inhibit CD41T-cell production of IFN-g and TNF-a ,to reduce the antigen-presenting capacity of dendritic cells,and to reduce inflammatory responses through the production of IL-10.Therefore B10cells are likely to inhibit lung inflammation through multiple IL-10–dependent mechanisms of negative regulation.TLR ,Toll-like receptor.J ALLERGY CLIN IMMUNOLMAY 20101126TEDDER AND MATSUSHITAREFERENCES1.Amu S,Saunders SP,Kronenberg M,Mangan NE,Atzberger A,Fallon PG.Regulatory B cells prevent and reverse allergic airway inflammation via FoxP3-positive T regulatory cells in a murine model.J Allergy Clin Immunol2010;1114-24.2.Mangan NE,Fallon RE,Smith P,van Rooijen N,McKenzie AN,Fallon PG.Hel-minth infection protects mice from anaphylaxis via IL-10-producing B cells.J Immunol2004;173:6346-56.3.Yanaba K,Bouaziz J-D,Haas KM,Poe JC,Fujimoto M,Tedder TF.A regulatoryB cell subset with a unique CD1dhiCD51phenotype controls T cell-dependent in-flammatory responses.Immunity2008;28:639-50.4.Fillatreau S,Sweenie CH,McGeachy MJ,Gray D,Anderton SM.B cells regulateautoimmunity by provision of IL-10.Nat Immunol2002;3:944-50.5.Matsushita T,Yanaba K,Bouaziz J-D,Fujimoto M,Tedder TF.Regulatory B cellsinhibit EAE initiation in mice while other B cells promote disease progression.J Clin Invest2008;118:3420-30.6.Mauri C,Gray D,Mushtaq N,Londei M.Prevention of arthritis by interleukin10-producing B cells.J Exp Med2003;197:489-501.7.Mizoguchi A,Mizoguchi E,Takedatsu H,Blumberg RS,Bhan AK.Chronic intes-tinal inflammatory condition generates IL-10-producing regulatory B cell subset characterized by CD1d upregulation.Immunity2002;16:219-30.8.DiLillo DJ,Matsushita T,Tedder TF.B10cells and regulatory B cells balance im-mune responses during inflammation,autoimmunity,and cancer.Ann N Y Acad Sci2010;1183:38-57.9.Yanaba K,Bouaziz J-D,Matsushita T,Tasubata T,Tedder TF.The developmentand function of regulatory B cells expressing IL-10(B10cells)requires antigen receptor diversity and TLR signals.J Immunol2009;182:7459-72.10.Yanaba K,Bouaziz JD,Matsushita T,Magro CM,St Clair EW,Tedder TF.B-lym-phocyte contributions to human autoimmune disease.Immunol Rev2008;223: 284-99.11.Yazdanbakhsh M,Kremsner PG,van Ree R.Allergy,parasites,and the hygienehypothesis.Science2002;296:490-4.12.Boyce JA,Broide D,Matsumoto K,Bochner BS.Advances in mechanisms ofasthma,allergy,and immunology in2008.J Allergy Clin Immunol2009;123:569-74.13.Haas KM,Watanabe R,Matsushita T,Nakashima H,Ishiura N,Okochi H,et al.Protective and pathogenic roles for B cells during systemic autoimmunity in NZB/W F1mice.J Immunol2010;In press.14.Watanabe R,Ishiura N,Nakashima H,Kuwano Y,Okochi H,Tamaki K,et al.Reg-ulatory B cells(B10cells)have a suppressive role in murine lupus:CD19and B10 cell deficiency exacerbates systemic autoimmunity.J Immunol2010;In press. 15.Simon D,Ho¨sli S,Kostylina G,Yawalkar N,Simon HU.Anti-CD20(rituximab)treatment improves atopic eczema.J Allergy Clin Immunol2008;121:122-8.J ALLERGY CLIN IMMUNOLVOLUME125,NUMBER5TEDDER AND MATSUSHITA1127。

相关文档
最新文档