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 109 毫秒
1.
Smad3基因剔除导致小鼠骨关节炎。为了进一步深入研究Smad3基因缺失导致骨关节炎形成的分子机制,寻找骨关节炎发生早期的分子变化。用双向电泳技术结合肽质量指纹谱技术对Smad3基因剔除小鼠和野生型小鼠血清蛋白质组进行了初步分析。对其中7个表达差异蛋白质进行了鉴定,并探讨了所鉴定蛋白质与骨关节炎发生的关系。为揭示SMAD3介导的TGF-β信号在骨骼发育中的重要作用提供了线索。   相似文献   

2.
王浩  张继帅  孙强  杨晓 《遗传学报》2007,34(8):698-708
此前发现 Smad3 基因敲除小鼠(Smad3ex8/ex8)的关节软骨细胞异常肥大分化,出现类似于人类骨关节炎的表型。为了进一步明确转化生长因子-β(TGF-β)/Smad3 信号通过调节哪些靶基因的表达来抑制关节软骨细胞的肥大分化,以及研究骨关节炎发病的分子机制,利用寡核苷酸芯片技术分析了 5 日龄 Smad3 基因敲除小鼠与野生型对照小鼠关节软骨细胞基因表达谱的改变。通过对差异表达基因的分析,发现在 Smad3 基因敲除小鼠软骨细胞中骨形态发生蛋白(BMP)与 TGF-β/细胞分裂周期基因 42(Cdc42)信号通路活性增强。此外,还发现其他信号通路,如生长激素(growth hormone)/胰岛素样生长因子 1(Igf1)以及成纤维细胞生长因子(Fgf)信号通路相关基因表达的改变。值得注意的是,还发现了 Smad3 基因敲除小鼠软骨细胞中蛋白合成与电子传递链相关基因的表达水平普遍上调,这意味着蛋白质合成速率的加快与细胞有氧呼吸的增强可能与关节软骨细胞的肥大分化和骨关节炎的发生相关。  相似文献   

3.
Smad3基因剔除小鼠的繁殖与基因型鉴定   总被引:5,自引:1,他引:4  
目的为进一步深入研究Smad3基因在脊椎动物发育中的重要作用,对Smad3基因剔除小鼠进行保种和繁育研究.方法采用基因剔除杂合子小鼠进行保种,通过PCR和Southern杂交对杂合子小鼠交配所产生的后代进行基因型鉴定,纯合子小鼠和野生型小鼠用于表型分析,杂合子小鼠用于留种和繁殖生产.结果采用PCR方法对278只子代小鼠进行了基因型鉴定,83只为野生型,133只为杂合子,62只为纯合子.结论Smad3基因剔除突变能稳定遗传.采用杂合子小鼠保种,子代小鼠三种基因型比例符合孟德尔遗传定律.  相似文献   

4.
为研究TGF β1 SMAD3信号对小鼠软骨细胞增殖和分化的影响 ,分离了野生型与Smad3基因剔除 (Smad3ex8 ex8)突变纯合子小鼠肋骨软骨细胞并进行了体外培养 .通过3 H TdR参入实验检测了体外培养软骨细胞的增殖能力 .TGF β1可以刺激野生型软骨细胞的增殖 ,Smad3基因缺失导致小鼠软骨细胞丧失对TGF β1刺激生长作用的应答 .Northern杂交显示 ,TGF β1促进野生型小鼠软骨细胞表达Ⅱ型胶原 ,而Smad3基因缺失突变纯合子软骨细胞大量表达肥大性软骨细胞的分子标记物X型胶原 .结果表明 ,SMAD3介导转化生长因子TGF β1刺激软骨细胞增殖并抑制软骨细胞的肥大性分化  相似文献   

5.
陈静  沈红  赵勇 《中国实验动物学报》2010,18(1):9-12,I0003
目的通过小鼠骨髓细胞剔除Smad3基因,观察小鼠病理变化以及免疫T细胞状态。方法将Smad3基因剔除Smad3-/-)的小鼠骨髓细胞和野生型(Smad3+/+)小鼠骨髓细胞分别移植给60Co射线照射GFP小鼠。观察骨髓移植后GFP小鼠体征变化,第6周处死小鼠,取肠道固定,HE染色观察其病理变化,流式细胞技术检测淋巴结中T细胞变化。结果移植Smad3-/-骨髓细胞的GFP小鼠逐渐消瘦,大肠出现炎症;淋巴结中活化型的CD4+CD62LloT细胞增多。结论骨髓细胞TGF-β信号受阻,可导致小鼠患炎症疾病,引起免疫T细胞活化。  相似文献   

6.
Smad5双等位基因剔除ES细胞的建立及其初步研究   总被引:3,自引:1,他引:3  
Smad5是TGF-信号的细胞质内信号转导分子. Smad5的定位剔除导致小鼠胚胎期死亡. 为了进一步研究Smad5在组织器官发育中的功能, 利用同源重组技术获得了Smad5双等位基因剔除的胚胎干(ES)细胞. 首先利用Cre-LoxP系统删除了Smad5中靶ES细胞的抗性基因, 再用相同的打靶载体进行转染, 经Southern杂交筛选获得了Smad5双等位基因剔除的ES细胞. 嵌合体研究的结果表明, Smad5在心脏和神经管的正常发育中可能起重要的作用. Smad5双等位基因剔除ES细胞在裸鼠皮下可以形成畸胎瘤, 并可分化成包括神经细胞、肌肉细胞、软骨细胞、内皮细胞等在内的多种细胞, 因而Smad5双等位基因剔除的ES细胞可用于研究Smad5介导的TGF-信号在多种组织器官发育和ES细胞体外定向分化中的作用.  相似文献   

7.
基于Cre/LoxP系统的Smad2条件基因打靶小鼠的建立   总被引:9,自引:0,他引:9  
Smads家族是转化生长因子TGF-b信号转导途径中一个重要的新基因家族.SMAD2属于受体激活的SMADs.Smad2基因在多种肿瘤中发生突变,是一种可能的肿瘤抑制基因. Smad2基因完全剔除导致小鼠在胚胎期6.5d死亡.为了研究Smad2在脊椎动物成体各组织器官及肿瘤发生中的可能作用,利用Cre/LoxP系统构建了Smad2条件基因打靶载体,将两个方向相同的LoxP序列置于编码SMAD2蛋白C末端功能域序列两侧的内含子中,并在组成型表达Cre重组酶的大肠杆菌中检测了LoxP位点的功能;用打靶载体电击转染小鼠胚胎干细胞,经Southern杂交筛选到3株发生正确同源重组的中靶ES细胞克隆;通过囊胚显微注射将中靶ES细胞导入受体小鼠囊胚腔,获得了ES细胞种系嵌合体;对高嵌合度小鼠与C57BL/6J的子代小鼠进行基因型鉴定,成功地获得了Smad2条件基因打靶小鼠.  相似文献   

8.
Smads家族是最新发现的TGF-β信号转导途径中一个重要的新基因家族,SMAD2属于受体激活的SMADs。Smad2在某些肿瘤中发生突变,是一种可能的肿瘤抑制基因。Smad2基因完全剔除小鼠在胚胎期E6.5天死亡,为了研究Smad2在成体各组织器官及肿瘤发生中的可能作用,构建了Smad2条件基因剔除载体,将LoxP置于Smad2基因组序列C末端功能域两侧,并在组成型表达Cre重组酶的大肠杆菌中检测了LoxP位点的功能,该载体的构建为进行Smad2组织特异性基因剔除研究了奠定了基础。  相似文献   

9.
目的 探讨Smad3基因对剔除小鼠血清酶活性的影响。方法 采用荷兰半自动生化分析仪对 35日龄、70日龄、6月龄的三种不同基因型的小鼠的ALP、AST、ALT、CK、LDH L进行测定。结果 纯合型小鼠各项指标较野生型高 ,且表现出ALP随着年龄的增长而降低 ;AST、ALT、CK、LDH L随着年龄的增长而增高。结论 Smad3基因的剔除对小鼠的血清酶活性有一定的影响 ,为该小鼠的进一步研究提供基础。  相似文献   

10.
小鼠Smad3基因的克隆及其在小鼠组织中的表达   总被引:4,自引:0,他引:4  
采用PCR获得的Smad3cDNA片段作为探针筛选小鼠脑cDNA文库 .克隆了小鼠全长的Smad3基因 .对小鼠Smad3基因的全编码区进行了序列测定 .结果表明 ,小鼠SMAD3与人SMAD3氨基酸同源性高达 99% .与小鼠Smad2基因相比 ,碱基同源性高达 91 8% .Northern杂交显示 ,Smad3基因在小鼠胚胎发育和各成体器官中普遍表达 .原位杂交显示 ,Smad3基因表达在小鼠胚胎期E16 5d的软骨、骨髓和皮肤角质细胞中  相似文献   

11.
Smad3 mediates TGF-beta1 induction of VEGF production in lung fibroblasts   总被引:5,自引:0,他引:5  
Transforming growth factor-beta1 (TGF-beta1) is a key factor in a variety of physiological and pathological processes. Vascular endothelial growth factor (VEGF) is a key angiogenic factor, and vascular change is one of the features of airway remodeling. We examined the effect of TGF-beta1 on VEGF production by fibroblasts from mice lacking expression of Smad2 or Smad3 as well as human lung fibroblasts treated with or without Smad2 or Smad3 siRNA. TGF-beta1 stimulated VEGF production by fibroblasts from Smad2 deficient animals and wildtype animals. In contrast, TGF-beta1 did not affect VEGF production by fibroblasts from Samd3 deficient mice. Similarly, TGF-beta1 failed to stimulate VEGF production by HFL-1 cells treated with Samd3 siRNA but significantly increased VEGF production by the cells treated with Smad2 siRNA. These result suggest that TGF-beta1 stimulation of VEGF production by fibroblasts is regulated by Smad3 but not by Smad2 signaling.  相似文献   

12.
13.
Smad3 protein is a prominent member of the Tgfb receptor signaling pathway. Smad3(-/-) mice display decreased radiation-induced skin fibrosis, suggesting a defect in both Tgfb-mediated fibroblast proliferation and migration. We established bone marrow stromal cell lines from Smad3(-/-) mice and homozygous littermate(+/+) mice. Smad3(-/-) cells displayed a significant increase in radiation resistance with a D(0)=2.25+/- 0.14 Gy compared to Smad3(+/+) cells with a D(0)=1.75+/- 0.03 (P=0.023). Radioresistance was abrogated by reinsertion of the human SMAD3 transgene, resulting in a D(0)=1.49 0.10 (P=0.028) for Smad3(-/-)(3) cells. More Smad3(-/-) cells than Smad3(+/+) cells were in the G(2)/M phase; Smad3(-/-)(3) cells were similar to Smad3(+/+) cells. Smad3(+/+) cells exhibited increased apoptosis 24 h after 5 Gy (15%) or 8 Gy (43%) compared to less than 1% in Smad3(-/-) cells exposed to either dose. The movement of Smad3(-/-) cells, measured in an automated cell tracking system, was slower than that of Smad3(+/+) cells. Smad3(-/-)(3) cells resembled Smad3(+/+) cells. These studies establish concordance of a defective Tgfb signal transduction pathway, an increased proportion of G(2)/M cells, and radioresistance. The decreased migratory capacity of Smad3(-/-) cells in vitro correlates with decreased radiation fibrosis in vivo in mice deficient in Tgfb signaling.  相似文献   

14.
Transforming growth factor-beta (TGF-beta) signaling plays an important regulatory role during lung fibrogenesis. Smad3 was identified in the pathway for transducing TGF-beta signals from the cell membrane to the nucleus. Using mice without Smad3 gene expression, we investigated whether Smad3 could regulate bleomycin-induced pulmonary fibrosis in vivo. Mice deficient in Smad3 demonstrated suppressed type I procollagen mRNA expression and reduced hydroxyproline content in the lungs compared with wild-type mice treated with bleomycin. Furthermore, loss of Smad3 greatly attenuated morphological fibrotic responses to bleomycin in the mouse lungs, suggesting that Smad3 is implicated in the pathogenesis of pulmonary fibrosis. These results show that Smad3 contributes to bleomycin-induced lung injury and that Smad3 may serve as a novel target for potential therapeutic treatment of lung fibrosis.  相似文献   

15.
Smad signalling in the ovary   总被引:1,自引:0,他引:1  
It has now been a decade since the first discovery of the intracellular Smad proteins, the downstream signalling molecules of one of the most important growth factor families in the animal kingdom, the transforming growth factor beta (TGF-beta) superfamily. In the ovary, several TGF-beta superfamily members are expressed by the oocyte, granulosa and thecal cells at different stages of folliculogenesis, and they signal mainly through two different Smad pathways in an autocrine/paracrine manner. Defects in the upstream signalling cascade molecules, the ligands and receptors, are known to have adverse effects on ovarian organogenesis and folliculogenesis, but the role of the individual Smad proteins in the proper function of the ovary is just beginning to be understood for example through the use of Smad knockout models. Although most of the different Smad knockouts are embryonic lethal, it is known, however, that in Smad1 and Smad5 knockout mice primordial germ cell development is impaired and that Smad3 deficient mice harbouring a deletion in exon 8 exhibit impaired folliculogenesis and reduced fertility. In this minireview we discuss the role of Smad structure and function in the ovarian context.  相似文献   

16.
17.
Zhu L  Wang L  Wang X  Luo X  Yang L  Zhang R  Yin H  Xie D  Pan Y  Chen Y 《PloS one》2011,6(2):e17415

Background

TGF-β has been known to play an important role in various liver diseases including fibrosis and alcohol-induced fatty liver. Smad7 is an intracellular negative regulator of TGF-β signaling. It is currently unclear whether endogenous Smad7 has an effect on liver function and alcoholic liver damage.

Methodology/Principal Findings

We used Cre/loxP system by crossing Alb-Cre mice with Smad7loxP/loxP mice to generate liver-specific deletion of Smad7 with loss of the indispensable MH2 domain. Alcoholic liver injury was achieved by feeding mice with a liquid diet containing 5% ethanol for 6 weeks, followed by a single dose of ethanol gavage. Deletion of Smad7 in the liver was associated with increased Smad2/3 phosphorylation in the liver or upon TGF-β treatment in primary hepatocytes. The majority of mice with liver specific deletion of Smad7 (Smad7liver-KO) were viable and phenotypically normal, accompanied by only slight or no reduction of Smad7 expression in the liver. However, about 30% of Smad7liver-KO mice with high efficiency of Smad7 deletion had spontaneous liver dysfunction, demonstrated as low body weight, overall deterioration, and increased serum levels of AST and ALT. Degeneration and elevated apoptosis of liver cells were observed with these mice. TGF-β-induced epithelial to mesenchymal transition (EMT) was accelerated in Smad7-deleted primary hepatocytes. In addition, alcohol-induced liver injury and steatosis were profoundly aggravated in Smad7 deficient mice, associated with upregulation of critical genes involved in lipogenesis and inflammation. Furthermore, alcohol-induced ADH1 expression was significantly abrogated by Smad7 deletion in hepatocytes.

Conclusion/Significance

In this study, we provided in vivo evidence revealing that endogenous Smad7 plays an important role in liver function and alcohol-induced liver injury.  相似文献   

18.

Background

New neurons are continuously being generated in the adult hippocampus, a phenomenon that is regulated by external stimuli, such as learning, memory, exercise, environment or stress. However, the molecular mechanisms underlying neuron production and how they are integrated into existing circuits under such physiological conditions remain unclear. Indeed, the intracellular modulators that transduce the extracellular signals are not yet fully understood.

Results

We show that Smad3, an intracellular molecule involved in the transforming growth factor (TGF)-β signaling cascade, is strongly expressed by granule cells in the dentate gyrus (DG) of adult mice, although the loss of Smad3 in null mutant mice does not affect their survival. Smad3 is also expressed by adult progenitor cells in the subgranular zone (SGZ) and more specifically, it is first expressed by Type 2 cells (intermediate progenitor cells). Its expression persists through the distinct cell stages towards that of the mature neuron. Interestingly, proliferative intermediate progenitor cells die in Smad3 deficiency, which is associated with a large decrease in the production of newborn neurons in Smad3 deficient mice. Smad3 signaling appears to influence adult neurogenesis fulfilling distinct roles in the rostral and mid-caudal regions of the DG. In rostral areas, Smad3 deficiency increases proliferation and promotes the cell cycle exit of undifferentiated progenitor cells. By contrast, Smad3 deficiency impairs the survival of newborn neurons in the mid-caudal region of the DG at early proliferative stages, activating apoptosis of intermediate progenitor cells. Furthermore, long-term potentiation (LTP) after high frequency stimulation (HFS) to the medial perforant path (MPP) was abolished in the DG of Smad3-deficient mice.

Conclusions

These data show that endogenous Smad3 signaling is central to neurogenesis and LTP induction in the adult DG, these being two forms of hippocampal brain plasticity related to learning and memory that decline with aging and as a result of neurological disorders.
  相似文献   

19.
Protein phosphatase magnesium-dependent 1A (PPM1A), a protein serine/threonine phosphatase, controls several signal pathways through cleavage of phosphate from its substrates. However, the in vivo function of Ppm1a in mammals remains unknown. Here we reported that mice lacking Ppm1a developed normally but were impaired in re-epithelialization process during cutaneous wound healing. Specifically, complete or keratinocyte-specific deletion of Ppm1a led to delayed re-epithelialization with reduced keratinocyte migration upon wounding. We showed that this effect was the result of an increase in Smad2/3 phosphorylation in keratinocytes. Keratinocyte-specific Smad2 deficient mice displayed accelerated re-epithelialization with enhanced keratinocyte migration. Importantly, Smad2 and Ppm1a double mutant mice also exhibited accelerated re-epithelialization, demonstrating that the effect of Ppm1a on promoting re-epithelialization is mediated by Smad2 signaling. Furthermore, the decreased expression of specific integrins and matrix metalloproteinases (MMPs) may contribute to the retarded re-epithelialization in Ppm1a mutant mice. These data indicate that Ppm1a, through suppressing Smad2 signaling, plays a critical role in re-epithelialization during wound healing.  相似文献   

20.
Transforming growth factor-beta1 is essential to maintain T cell homeostasis, as illustrated by multiorgan inflammation in mice deficient in TGF-beta1 signaling. Despite the physiological importance, the mechanisms that TGF-beta1 uses to regulate T cell expansion remain poorly understood. TGF-beta1 signals through transmembrane receptor serine/threonine kinases to activate multiple intracellular effector molecules, including the cytosolic signaling transducers of the Smad protein family. We used Smad3(-/-) mice to investigate a role for Smad3 in IL-2 production and proliferation in T cells. Targeted disruption of Smad3 abrogated TGF-beta1-mediated inhibition of anti-CD3 plus anti-CD28-induced steady state IL-2 mRNA and IL-2 protein production. CFSE labeling demonstrated that TGF-beta1 inhibited entry of wild-type anti-CD3 plus anti-CD28-stimulated cells into cycle cell, and this inhibition was greatly attenuated in Smad3(-/-) T cells. In contrast, disruption of Smad3 did not affect TGF-beta1-mediated inhibition of IL-2-induced proliferation. These results demonstrate that TGF-beta1 signals through Smad3-dependent and -independent pathways to inhibit T cell proliferation. The inability of TGF-beta1 to inhibit TCR-induced proliferation of Smad3(-/-) T cells suggests that IL-2 is not the primary stimulus driving expansion of anti-CD3 plus anti-CD28-stimulated T cells. Thus, we establish that TGF-beta1 signals through multiple pathways to suppress T cell proliferation.  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