首页 | 本学科首页   官方微博 | 高级检索  
相似文献
 共查询到20条相似文献,搜索用时 152 毫秒
1.
雌激素、免疫细胞因子和骨组织的代谢三者之间具有密切而又复杂的联系。雌激素除了可直接与成骨细胞和破骨细胞上的雌激素受体结合产生生物学效应外,另一方面还可以影响骨细胞和成纤维细胞等对某些细胞因子的表达,如激活OPG/RANK/RANKL系统、增加TGF-β、IGF-1等的分泌;减少IL-1、IL-6、TNF-α等抑制性细胞因子的表达等。各种细胞因子之间相互作用、联系,组成复杂的调控网络,与细胞外基质一起形成骨组织发育所特需的骨微环境,它们作用于成骨细胞和破骨细胞,介导骨细胞的分化及成熟,参与正常的骨组织的代谢。因此绝经后由于雌激素的丢失,使细胞因子的表达发生改变,其在骨组织的代谢及代谢性骨病的发病中起到重要作用。  相似文献   

2.
IL-17在类风湿性关节炎中的作用   总被引:1,自引:0,他引:1  
类风湿性关节炎的发病与多种细胞因子如TNF-α、IL-1、IL-17等有关.IL-17是一种前炎症细胞因子,在活动性类风湿性关节炎患者血清及关节液中表达均增高.IL-17主要由活化的T细胞产生,并能诱导活化T细胞,促进滑膜细胞分泌多种细胞因子,抑制软骨细胞合成基质,增强破骨细胞活性,最终导致骨侵蚀.IL-17能与其他细胞因子相互作用,导致类风湿性关节炎进一步发展.IL-17在类风湿性关节炎进程中起重要的调节作用,因此抗IL-17抗体生物治疗有望成为治疗类风湿性关节炎的新方法.  相似文献   

3.
绝经后骨质疏松是因雌激素水平下降引起骨免疫功能增强,导致骨重建失衡所致。成骨细胞和破骨细胞相互作用是调控骨重建的核心因素,而RANKL/RANK/OPG系统是连接骨与免疫系统之间的分子桥梁,在调节骨吸收和骨形成耦联中发挥关键作用。骨细胞和免疫细胞通过共同的细胞因子及受体相互作用,调节骨代谢平衡,以共同的骨代谢转录因子和信号分子调控骨重建过程。雌激素作为骨细胞与免疫细胞的共同交叉机制,通过经典性激素受体途径调控T细胞活化,介导免疫因子的产生和表达,影响免疫细胞、成骨细胞、破骨细胞的活性和功能,既有正性调节作用,又有负性调节作用,决定骨重建方向。因此,骨免疫在调控绝经后骨质疏松症骨重建失衡中发挥重要作用。补肾法是中医药治疗绝经后骨质疏松症的根本治则,长期临床应用疗效显著,其作用机制可能是通过干预骨免疫而调节骨重建失衡,为治疗绝经后骨质疏松症提供新的切入点。  相似文献   

4.
雌激素缺乏是绝经后骨质疏松症发生的主要原因,雌激素或雌激素受体调节剂与雌激素受体结合后通过多种分子生物学机制,抑制破骨细胞骨吸收,促进成骨细胞骨形成、提高骨密度,达到治疗绝经后骨质疏松症的目的。本文综述了雌激素、选择性雌激素受体调节剂与雌激素受体相互作用对骨代谢的调节机制、雌激素治疗骨质疏松症的动物实验研究以及雌激素治疗骨质疏松症的临床研究进展,旨在为临床骨质疏松治疗策略提供科学依据。  相似文献   

5.
破骨细胞分化成熟因子及其信号转导通路   总被引:3,自引:0,他引:3       下载免费PDF全文
破骨细胞从起源发育至成熟,再经活化发挥吸收作用是一个复杂的多级调控过程,始终都受到一系列细胞因子的影响。有些细胞因子对破骨细胞的成熟分化起促进作用,如:RANKL、TNF-α、IL-1、IL-6、1,25-(OH)2D3、PTH、M-CSF等,其中RANKL和M-CSF是破骨细胞形成和分化过程中的两个必需的因子;有些因子起抑制作用,如:OPG、IL-4、IL-10、雌激素、降钙素、TGF-β等。OPG/RANK/RANKL系统在破骨细胞分化成熟过程中起着枢纽作用,大部分细胞因子都直接或间接地通过OPG/RANK/RANKL系统来发挥作用,其中还涉及到成骨细胞、破骨细胞、基质细胞等复杂的相互作用。介导破骨细胞分化成熟的各种细胞因子反应的信号传导路径主要包括MAPK、NF-kappaB、CN/NFAT等通路,全面地了解破骨细胞因子及其信号传导通路,将有助于临床更好地分析各种骨代谢性疾病的病因及发病机制,进而为治疗提供理论依据。  相似文献   

6.
目的:通过观察雌激素对骨髓来源的成骨细胞所产生的细胞因子的调节作用,探讨雌激素抑制破骨细胞功能的机制。方法:在诱导小鼠骨髓细胞分化为成骨细胞后,于成骨细胞培养中加入雌激素,应用抗体荧光免疫标记法检测成骨细胞培养液中IL-1、IL-6和TNF-α水平。结果:与对照组比较,经0.01-1nM雌激素处理后, 成骨细胞所产生的IL-1和IL-6水平呈浓度依赖性下降,TNF-α水平则无明显变化。结论:雌激素具有调节成骨细胞分泌细胞因子功能,提示雌激素抑制破骨细胞作用机制源自调控成骨细胞旁分泌。  相似文献   

7.
绝经后骨质疏松症的骨微环境改变探析   总被引:2,自引:1,他引:1  
董万涛  宋敏 《颈腰痛杂志》2007,28(6):524-526
绝经后雌激素低落引起的成骨细胞、破骨细胞、骨细胞异常凋亡,导致骨形成、骨吸收脱偶联,是绝经后骨质疏松症发生的重要原因,由于雌激素的缺乏破坏了其对骨局部细胞因子、生长因子、体液因子的调控,骨微环境改变,刺激骨吸收大于骨形成,本文就绝经后骨质疏松症的骨微环境改变作一理论探析,为临床治疗拓展新途径.  相似文献   

8.
原发性骨质疏松症是以骨质减少,骨的微观结构退化为特征的,致使骨的脆性增加以及易于发生骨折的骨骼疾病。其发病机制与多种因素相关,除了传统的内分泌机制之外,一种新的骨骼免疫机制已逐渐被深入研究:通过免疫细胞T淋巴细胞和B淋巴细胞、树突状细胞等,分泌多种细胞因子,并与多种细胞因子相互作用,通过信号通路的正负反馈调控,精细调节成骨细胞和破骨细胞的分化与增殖平衡,从而影响原发性骨质疏松症的发生。与破骨形成相关的T细胞,Th17细胞通过双重机制调控骨质吸收,Th1和Th2细胞亚群分别分泌IFN-γ和IL-4,通过抑制破骨细胞前体细胞发育成成熟的破骨细胞,从而抑制骨质吸收。Treg细胞通过表达CTLA-4,促进破骨细胞前体细胞的凋亡,抑制骨质吸收。B淋巴细胞通过调控RANKL和OPG的比例参与骨质代谢。树突状细胞既可以与CD4+T细胞结合,启动经典的RANKL/RANK破骨细胞形成的信号通路,参与骨质疏松的形成;也可以作为破骨细胞前体细胞的方式,在M-CSF等炎性因子的刺激下,直接分化为破骨细胞。现就这种免疫细胞与细胞因子精细调节骨质生成与骨质吸收平衡作用机制的最新研究进展进行阐述。  相似文献   

9.
骨的重建是骨组织在成骨与破骨间一个连续、协调的平衡过程.成骨细胞与破骨细胞参与此过程,破骨细胞主要通过酸化作用和释放溶酶体酶使骨吸收;成骨细胞则具有两个主要功能,一是合成细胞外基质成分(主要是Ⅰ型胶原)参与骨形成,二是产生细胞因子调控破骨细胞的形成或活性[1-4].  相似文献   

10.
破骨细胞数量和骨吸收活性的改变是引起骨溶解疾病的重要机制,如何调节破骨细胞的形成和功能,抑制骨吸收活性是治疗骨质疏松、骨髓瘤等骨溶解疾病的关键.研究发现破骨细胞分化、形成及作用的发挥受多种细胞因子调节,如破骨细胞标志性基因之间的相互作用可调节破骨细胞分化,某些转录因子可调节破骨细胞形成,其他相关因子可对破骨细胞产生影响等.该文就相关因子对破骨细胞调节的研究进展作一简要综述.  相似文献   

11.
Nagata N  Kitaura H  Yoshida N  Nakayama K 《BONE》2003,33(4):721-732
IL-12 was shown to have the potential to inhibit osteoclast formation in mouse bone marrow cells treated with macrophage colony-stimulating factor (M-CSF) and receptor activator of NF-kappaB ligand (RANKL). When bone marrow macrophages (BMM) were used as osteoclast precursors, IL-12 failed to inhibit M-CSF/RANKL-induced osteoclast formation from BMM. In coculture experiments using transwells, IL-12 did inhibit osteoclast formation from BMM cocultured with whole bone marrow cells. These results indicated that IL-12 indirectly affected M-CSF/RANKL-induced osteoclastogenesis in bone marrow cells and that the inhibition of IL-12 on osteoclast formation was caused by a humoral factor from bone marrow cells treated with IL-12. Experiments with anti-interferon (IFN)-gamma antibody and bone marrow cells from IFN-gamma receptor knockout mice revealed that IFN-gamma might be involved in the inhibition of osteoclast formation in this system. The expression of osteoprotegerin mRNA in bone marrow cells was not affected by treatment with IL-12. The inhibitory effect of IL-12 on osteoclast formation was also seen in the T cell-depleted bone marrow cells of normal mice and the whole bone marrow cells of athymic nude mice, while the inhibitory effect of IL-12 was partially suppressed in the B cell-depleted bone marrow cells. The inhibitory effect of IL-12 on M-CSF/RANKL-induced osteoclastogenesis was not accompanied with cell death, in contrast with our previous finding that the inhibitory effect of IL-12 on M-CSF/TNF-alpha-induced osteoclastogenesis is attributable to Fas and FasL-mediated apoptosis.  相似文献   

12.
Osteoclastogenic cytokines produced by T and B lineage cells and interleukin (IL)-7-induced expansion of the pool size of osteoclast precursors have been suggested to play an important role in acceleration of osteoclastogenesis induced by estrogen deficiency. However, the contribution of increased RANKL produced by osteoblasts/stromal cells to increase osteoclastogenesis in a mouse model of estrogen-deficient osteoporosis and in vitro effects of IL-7 on osteoclast precursor generation remain controversial. Thus, we investigated the effect of ovariectomy (OVX) of mice on production of RANKL, osteoprotegerin (OPG), and IL-7 in bone and the effect of IL-7 on osteoclast precursor generation in vitro. OVX did not significantly stimulate mRNA expressions of RANKL and OPG in whole femurs. Because the epiphysis, but not the femoral shaft (diaphysis) or bone marrow, is the main site of osteoclastogenesis, it is important to specifically analyze mRNA expression by osteoblasts/stromal cells at these parts of the femur. Therefore, we isolated RNA from bone marrow cell-free epiphysis, diaphysis, and flushed-out bone marrow and examined mRNA expression. The results showed no significant changes of RANKL and OPG mRNA expression in any part of the femur. In addition, OVX did not significantly affect RANKL and OPG mRNA expression by the adherent stromal cells isolated from flushed-out bone marrow cells but did stimulate RANKL mRNA expression by B220+ cells in the nonadherent cell fraction. On the other hand, OVX increased IL-7 mRNA expression in the femur as well as IL-7 concentrations in bone fluid. In cultures of unfractionated bone cells isolated by vigorous agitation of minced whole long bones to release the cells tightly attached to the bone surfaces, but not in cocultures of clonal osteoblasts/stromal cells and flushed-out bone marrow cells, IL-7 stimulated generations of osteoclasts as well as osteoclast precursors. These data suggest that increased RANKL production by osteoblasts/stromal cells is unlikely to play a central role in acceleration of osteoclastogenesis in estrogen deficiency of mice and that IL-7 stimulates osteoclast precursor generation, presumably through an action of IL-7 on the cells attached to bone rather than on cells contained in the bone marrow cell population.  相似文献   

13.
IL-4 is an important cytokine that can influence bone. We identified two distinct actions of IL-4 to inhibit osteoclast formation: one direct on osteoclast progenitors and the second through the production of a novel T-cell surface-associated molecule(s). These data show a new link between the immune system and bone. The Th2 cytokine interleukin (IL)-4 inhibits osteoclast formation in vitro but also acts on other cell types found in bone, including T-cells and macrophages. Because some osteoclastogenesis inhibitors (e.g., IL-12) act indirectly through T-cells, we investigated IL-4 action on osteoclastogenesis in the presence of T-cells. Osteoclast formation from murine spleen cells treated with RANKL and macrophage colony-stimulating factor (M-CSF) was blocked by IL-4 even when spleen cells were depleted of T-cells (Thy 1.2+) and/or B-cells (B220+). Also, IL-4 inhibited osteoclastogenesis in RANKL/M-CSF-stimulated adherent spleen cells, Rag1 -/- (lymphocyte-deficient) spleen cells, and bone marrow macrophages, indicating an action on myelomonocytic cells to block osteoclastogenesis. In contrast, IL-4 did not inhibit osteoclastogenesis in cells from IL-4 receptor null mice (IL-4R -/-). However, when wildtype T-cells were added to IL-4R -/- spleen cell cultures, IL-4 inhibited osteoclast formation, indicating a T-cell-dependent action. Osteoclast formation in RANKL-stimulated RAW 264.7 cells was not inhibited by IL-4 unless T-cells were added to the culture. Separation of RAW 264.7 cells and T-cells by semipermeable membrane ablated this action of IL-4, suggesting the induction of a membrane-associated osteoclastogenesis inhibitor. However, membrane-bound inhibitors thymic shared antigen-1 (TSA-1) and osteoclast inhibitory lectin (OCIL) were not regulated by IL-4. In summary, at least two mechanisms of IL-4 -mediated osteoclastogenesis inhibition exist, including a direct action on myelomonocytic progenitors (from which osteoclasts derive) and an indirect action through T-cells that may involve novel anti-osteoclastic factors.  相似文献   

14.
Rheumatoid arthritis (RA) is an autoimmune disease characterized by a heavy lymphocytic infiltration into the synovial cavity, resulting in the secretion of a variety of cytokines which ultimately leads to destruction of joint tissue. Among the infiltrating cells are activated T cells which produce specific cytokines capable of osteoclast progenitor cell expansion, fusion, and activation. Cultures of activated human T cells and human osteoblasts (hOBs) were used to study the possibility that lymphokines may act on osteoblasts to produce the osteoclastogenic factor interleukin-6 (IL-6). Purified T cells were activated with a combination of anti-CD3 and anti-CD28 antibodies, cocultured with hOBs in direct physical contact or separated by a transwell system, and conditioned media (CM) were assayed for IL-6 production. After a 72 h incubation period, activated T cell-hOB interaction resulted in a 100-fold increase of IL-6 production over basal levels. The immunosuppressant cyclosporine A (CsA) inhibited T cell tumor necrosis factor alpha and IL-6 production but did not inhibit the T cell induction of IL-6 from hOB. Assay of activated T-cell CM on hOB revealed that a soluble factor, not cell-cell contact, was the major inducer of IL-6. The induction of IL-6 mRNA by both activated T cell CM and CsA-treated activated T cell CM was confirmed by Northern blot analysis. Neutralizing antibodies to IL-13 and IL-17 did not affect IL-6 production. These findings suggest that activated T cells produce a novel, potent, IL-6 inducing factor that may be responsible for the bone loss observed in RA patients.  相似文献   

15.
Macrophage-colony stimulating factor (M-CSF) is an essential requirement for human osteoclast formation, but its effect on the proliferation and differentiation of circulating osteoclast precursor cells is unknown. Other growth factors and cytokines are also known to support/stimulate osteoclast formation from mouse marrow precursors, but it is not certain whether these factors similarly influence human osteoclast formation. In this study, human monocytes were cocultured with osteoblast-like UMR-106 cells on coverslips and dentine slices for up to 21 days in the presence of 1,25 dihydroxyvitamin D(3) (10(-7) mol/L), dexamethasone (10(-8) mol/L), and various concentrations of either M-CSF or other humoral factors (interleukin [IL]-1beta, IL-3, IL-6, and IL-11; tumor necrosis factor-alpha [TNF-alpha]; and granulocyte macrophage [GM]-CSF). The effect on osteoclast formation was assessed by tartrate-resistant acid phosphatase (TRAP) and vitronectin receptor staining and lacunar bone resorption. The results of time-course and proliferation studies showed that M-CSF stimulated both the proliferative and differentiation stages of human osteoclast formation from circulating osteoclast precursors in a dose-dependent manner. A high concentration of M-CSF (100 ng/mL) did not inhibit osteoclast formation. IL-3 and GM-CSF were also capable of stimulating human osteoclast formation, although these growth factors were much less potent than M-CSF. IL-3- and GM-CSF-stimulated osteoclast formation was inhibited by an antibody specific for human M-CSF. Osteoclast formation and lacunar resorption was not seen when either TNF-alpha, IL-1beta, IL-6 (+ soluble IL-6 receptor), or IL-11 was substituted for M-CSF during coculture. These results confirm that M-CSF is essential for human osteoclast formation from circulating mononuclear precursors, and also shows that IL-3 and GM-CSF may support osteoclast differentiation via the stimulation of M-CSF production by human monocytes.  相似文献   

16.
Compelling evidences suggest that increased production of osteoclastogenic cytokines by activated T cells plays a relevant role in the bone loss induced by estrogen deficiency in the mouse. However, little information is available on the role of T cells in post-menopausal bone loss in humans. To investigate this issue we have assessed the production of cytokines involved in osteoclastogenesis (RANKL, TNFalpha and OPG), in vitro osteoclast (OC) formation in pre and post-menopausal women, the latter with or without osteoporosis. We evaluated also OC precursors in peripheral blood and the ability of peripheral blood mononuclear cells to produce TNFalpha in both basal and stimulated condition by flow cytometry in these subjects. Our data demonstrate that estrogen deficiency enhances the production of the pro-osteoclastogenetic cytokines TNFalpha and RANKL and increases the number of circulating OC precursors. Furthermore, we show that T cells and monocytes from women with osteoporosis exhibit a higher production of TNFalpha than those from the other two groups. Our findings suggest that estrogen deficiency stimulates OC formation both by increasing the production of TNFalpha and RANKL and increasing the number of OC precursors. Women with post-menopausal osteoporosis have a higher T cell activity than healthy post-menopausal subjects; T cells thus contribute to the bone loss induced by estrogen deficiency in humans as they do in the mouse.  相似文献   

17.
Excessive production of tumor necrosis factor (TNF) and interleukin-1 (IL-1) secondary to estrogen deficiency have been implicated as the cause of osteoporosis in postmenopausal woman. These cytokines appear to stimulate osteoclast precursor proliferation and activate mature osteoclast formation directly and possibly indirectly via osteoblasts. To investigate the other possible roles that these cytokines may play in stimulating the bone resorption process, we examined the effect of TNF-alpha and IL-1beta on cell-cell adhesion molecules, cadherins, in osteoblastic MC3T3-E1 cells. In this study, we investigated cadherin expression and the effect of TNF-alpha, IL-1beta, and parathyroid hormone (PTH) on the expression of cadherins in MC3T3-E1 cells. Confluent cultures of MC3T3-E1 cells were challenged with recombinant human TNF-alpha (1-100 U/ml), recombinant human IL-1beta (1-100 ng/ml) and human PTH(1-34) (1-100 ng/ml), respectively. The results show that MC3T3-E1 cells express functional cadherin molecules, N-cadherin and OB-cadherin. TNF-alpha (10-100 U/ml) and IL-1beta (10-100 ng/ml) suppressed N-cadherin without changing OB-cadherin expression, while PTH (1-100 ng/ml) had no effect on cadherin expression. These results raise the possibility that TNF-alpha and IL-1beta may compromise the cell-cell adhesion of osteoblasts which cover the bone surface. The ensuing compromised cell-cell adhesion of osteoblasts may in turn facilitate the direct adhesion of osteoclasts on the calcified bone matrix surface. These results implicate an indirect role for osteoblasts in the promotion of bone resorption by TNF-alpha and IL-1beta.  相似文献   

18.
There is accumulating evidence that T cells may be involved in osteoclastogenesis in a variety of murine systems. However, the precise role of human T cells in the regulation of osteoclast generation is still unclear. To address this issue, we investigated the effect of resting peripheral T cells on receptor activator of NF-kappaB ligand (RANKL)-induced osteoclast generation from human peripheral monocytes. Although osteoclasts were not generated in the culture of human peripheral blood mononuclear cells (PBMC) in the presence of RANKL and macrophage colony-stimulating factor (M-CSF), the addition of cyclosporine A (CsA), a potent inhibitor of T-cell function, resulted in the formation of an increasing number of lacunae resorption on dentine, suggesting T cells may inhibit osteoclast formation. In a coculture of T cells and monocytes, which were isolated from PBMC, T cells inhibited the osteoclast generation from monocytes, as determined by tartrate-resistant acid phosphatase (TRAP) staining and a pit assay using dentine. This inhibition of osteoclast generation by T cells was also observed in a culture of the parathyroid hormone-stimulated SaOS4/3 osteoblast cell line and monocytes. The culture in Transwell plates revealed that the cell-to-cell interaction was not required for the inhibition, suggesting that T-cell cytokines may be responsible for the inhibition. Among inhibitory T-cell cytokines on osteoclastogenesis, granulocyte-macrophage colony-stimulating factor (GM-CSF) and interferon-gamma (IFN-gamma) were actively produced by CD4 T cells but not CD8 T cells in the coculture of T cells with monocytes, and the neutralizing antibodies to these cytokines partially rescued the T-cell-induced inhibition of osteoclast formation. Although CsA did not affect RANKL-induced osteoclast generation in the culture of monocytes alone, it completely rescued the T-cell-induced inhibition of osteoclast formation and strongly inhibited the production of GM-CSF and IFN-gamma. Thus, we demonstrate that resting T cells negatively regulate the osteoclast generation via production of GM-CSF and IFN-gamma by CD4 T cells and that CsA stimulates the osteoclast generation through the inhibition of the production of these cytokines. These findings provide new insight into therapeutic strategies for immunosuppression-induced bone loss in transplant and other diseases.  相似文献   

19.
Role for osteoprotegerin in rheumatoid inflammation   总被引:8,自引:0,他引:8  
Osteoprotegerin (OPG), a member of the TNF-receptor family expressed by osteoblasts, has documented effects on the regulation of bone metabolism. OPG inhibits bone resorption and binds with strong affinity to its ligand RANKL, thereby preventing RANKL from binding to its receptor RANK. This system is regulated by calcium-modifying hormones. OPG may also be pivotal in modulating the immune system. RANKL-deficient mice exhibit both severe immunological abnormalities and osteopetrosis, and activated T cells express RANKL mRNA. RANKL secretion by activated T cells may induce osteoclastogenesis via a mechanism enhanced by several cytokines (TNF-alpha, IL-1, and IL-17) that promote both inflammation and bone resorption. Conversely, this mechanism is inhibited by OPG, IL-4, and IL-10, which have antiinflammatory effects and inhibit osteoclast formation. Activated T cells in the rheumatoid synovium express RANKL. Synoviocytes can differentiate to osteoclast-like cells under specific conditions, particularly when they are cultured with M-CSF and RANKL. Thus, the bony erosions seen in RA may result from RANKL/RANK system activation by activated T cells. This raises the possibility that OPG therapy to block this mechanism might prove beneficial in patients with RA.  相似文献   

20.
It has been reported that osteoclastogenesis is induced by tumor necrosis factor (TNF)-α. Interleukin (IL)-4 is the most important cytokine involved in humoral immunity. However, no studies have investigated the effect of IL-4 on TNF-α-mediated osteoclast formation in vivo. In this study, we investigated the effect of IL-4 on TNF-α-mediated osteoclast formation in vivo. TNF-α was administered with and without IL-4 into the supracalvariae of mice. The number of osteoclasts and the levels of mRNA for cathepsin K and tartrate-resistant acid phosphate, both osteoclast markers, in mice administered TNF-α and IL-4 were lower than those in mice administered TNF-α alone. The level of tartrate-resistant acid phosphatase form 5b (TRACP5b) as a marker of bone resorption in mice administered both TNF-α and IL-4 was also lower. We showed that IL-4 inhibited TNF-α-mediated osteoclast formation in osteoclast precursors in vitro. Expression of receptor activator of NF-κB ligand (RANKL) in TNF-α-activated stromal cells was also inhibited. Furthermore, we investigated whether IL-4 had effects on both stromal cells and osteoclast precursors in TNF-α-mediated osteoclast formation in vivo. Using mice whose stromal cells and osteoclast precursors were chimeric for the presence of TNF receptors, IL-4 inhibited TNF-α-mediated osteoclast formation in the presence of TNF-α-responsive stromal cells, and TNF-α-responsive osteoclast precursors in vivo. IL-4 also inhibited TNF-α-induced RANKL expression in the presence of TNF-α-responsive stromal cells in vivo. This event is dependent on p38 inhibition in vitro. Additionally, IL-4 inhibited TNF-α-mediated osteoclast formation in T cell-depleted mice. In summary, we conclude that IL-4 inhibited TNF-α-mediated osteoclast formation by inhibiting expression of RANKL in TNF-α-activated stromal cells, and directly inhibited TNF-α-activated osteoclast precursors in vivo via a T cell-independent mechanism.  相似文献   

设为首页 | 免责声明 | 关于勤云 | 加入收藏

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