首页 | 本学科首页   官方微博 | 高级检索  
相似文献
 共查询到20条相似文献,搜索用时 15 毫秒
1.
Aberrant activity of the phosphatidylinositol 3-kinase (PI3K) pathway supports growth of many tumors including those of breast, lung, and prostate. Resistance of breast cancer cells to targeted chemotherapies including tyrosine kinase inhibitors (TKI) has been linked to persistent PI3K activity, which may in part be due to increased membrane expression of epidermal growth factor (EGF) receptors (HER2 and HER3). Recently we found that proteins of the RGS (regulator of G protein signaling) family suppress PI3K activity downstream of the receptor by sequestering its p85α subunit from signaling complexes. Because a substantial percentage of breast tumors have RGS16 mutations and reduced RGS16 protein expression, we investigated the link between regulation of PI3K activity by RGS16 and breast cancer cell growth. RGS16 overexpression in MCF7 breast cancer cells inhibited EGF-induced proliferation and Akt phosphorylation, whereas shRNA-mediated extinction of RGS16 augmented cell growth and resistance to TKI treatment. Exposure to TKI also reduced RGS16 expression in MCF7 and BT474 cell lines. RGS16 bound the amino-terminal SH2 and inter-SH2 domains of p85α and inhibited its interaction with the EGF receptor-associated adapter protein Gab1. These results suggest that the loss of RGS16 in some breast tumors enhances PI3K signaling elicited by growth factors and thereby promotes proliferation and TKI evasion downstream of HER activation.The role of the PI3K3 pathway in cell proliferation and survival, adhesion, metabolism, migration, drug resistance, and cytoskeletal rearrangement is well documented (13). Mutations in PI3K and dysregulation of the PI3K pathway have been implicated in many human cancers including lymphoma, multiple myeloma, and melanoma (48). Because the PI3K signal is a gatekeeper for tumor growth, an understanding of its regulation is critical for the therapeutic intervention of cancer.PI3K, which catalyzes the production of phosphatidylinositol 3,4,5-trisphosphate from phosphatidylinositol 3,4-bisphosphate (9, 10), is activated by extracellular receptor tyrosine kinases including the EGF receptor (EGFR or HER) family, platelet-derived growth factor receptor, and the insulin growth factor receptor. HER stimulation activates Class IA PI3Ks consisting of dimers of p85α or β and either p110α, β, and δ catalytic subunits (11). Tyrosine phosphorylation of the adapter protein Grb2-associated binder 1 (Gab1) recruits p85 to the EGFR complex through a Src homology 2 (SH2) domain in p85 (12), which co-localizes the catalytic p110 subunit and membrane phospholipid substrates at the plasma membrane. Phosphatidylinositol 3,4,5-trisphosphate generated by PI3K activity recruits phosphoinositide-dependent kinase 1 through its pleckstrin homology domain, which in turn phosphorylates the mitogenic and antiapoptotic kinase Akt. Substrates of Akt include mTOR, BAD, IKK, FOXO, p27, MDM2, and GSK3β, all of which are signaling molecules with vital functions in cell cycle regulation and growth (3). Overexpression of Akt has been shown in several tumors such as ovarian and breast carcinoma and may lead directly to transformation of malignant melanoma (5).Proteins of the RGS (regulator of G protein signaling) family mediate cellular desensitization to G protein-coupled receptor stimulation. RGS proteins act as GTPase-accelerating proteins to reduce the life span of activated (GTP-bound) Gα subunits of the G protein-coupled receptor signal-transducing heterotrimeric G protein (13). The R4 subfamily of RGSs (RGS1, 2, 4, 5, 8, 13, 16, 18, and 21) are the smallest members of the family, containing few residues outside of the ∼120-amino acid RGS domain that mediates binding to Gα proteins and GTPase-accelerating protein activity. We found recently that several R4 RGS proteins interacted with the phosphorylated p85α subunit of PI3K (14). In mast cells, RGS13 inhibited PI3K activation induced by high affinity IgE receptor (FcϵRI) cross-linking by antigen. FcϵRI stimulates PI3K by recruiting its catalytic p110δ subunit through p85 binding to a multi-protein complex that includes Gab2 and Grb2 at the plasma membrane (15). PI3K has an essential function in allergic responses (16). As a result of increased PI3K activation, mice deficient in RGS13 had more IgE-mediated mast cell degranulation and anaphylaxis (14).RGS16, an R4 RGS protein homologous to RGS13, was identified originally as a p53 target gene in breast and colon cancer cells (17, 18). Recent analysis of 222 primary breast cancers found a high rate (50%) of genomic instability at the RGS16 locus (19). Because RGS16 associates with both EGFR (20) and p85α (14), we investigated how it affected the growth and survival of breast cancer cells. We found that RGS16 directly bound the amino-terminal SH2 and inter-SH2 domains of phosphorylated p85α, which mediate p110 and adapter binding and membrane localization (21). RGS16 overexpression in MCF7 breast cancer cells suppressed proliferation and EGF-induced Akt phosphorylation, whereas extinction of RGS16 expression increased cell growth and resistance to TKI treatment. Thus, through regulation of PI3K activity, RGS16 may limit proliferation of mammary cells and render cancer cells more susceptible to TKIs or other therapeutic compounds.  相似文献   

2.
本室以前已经报道G蛋白偶联受体APJ的内源性配体多肽apelin-13促进单核细胞-血管内皮细胞黏附,本文研究PI3K信号途径是否参与apelin-13促进单核细胞-血管内皮细胞黏附,探讨apelin/APJ系统的细胞信号转导机制.MPO方法检测细胞黏附;Western blot方法检测PI3K、VCAM-1的表达.Western blot方法结果显示,apelin-13(0、0.5、1、2、4μmol/L)浓度依赖性刺激血管内皮细胞PI3K磷酸化,以1μmol/L最为明显;1μmol/L apelin-13时间依赖性促进血管内皮细胞PI3K磷酸化,在30 min增加最为显著;PI3K抑制剂LY294002明显抑制apelin-13诱导的VCAM-1表达和单核细胞-血管内皮细胞黏附.上述结果表明,PI3K信号途径介导apelin-13促进单核细胞-血管内皮细胞黏附.  相似文献   

3.
CD5 acts as a coreceptor on T lymphocytes and plays an important role in T-cell signaling and T-cell–B-cell interactions. Costimulation of T lymphocytes with anti-CD5 antibodies results in an increase of the intracellular Ca2+ levels, and subsequently in the activation of Ca2+/calmodulin-dependent (CaM) kinase type IV. In the present study, we have characterized the initial signaling pathway induced by anti-CD5 costimulation. The activation of phosphatidylinositol (PI) 3-kinase through tyrosine phosphorylation of its p85 subunit is a proximal event in the CD5-signaling pathway and leads to the activation of the lipid kinase activity of the p110 subunit. The PI 3-kinase inhibitors wortmannin and LY294002 inhibit the CD5-induced response as assessed in interleukin-2 (IL-2) secretion experiments. The expression of an inactivated Rac1 mutant (Rac1 · N17) in T lymphocytes transfected with an IL-2 promoter-driven reporter construct also abrogates the response to CD5 costimulation, while the expression of a constitutively active Rac1 mutant (Rac1-V12) completely replaces the CD5 costimulatory signal. The Rac1-specific guanine nucleotide exchange factor Vav is heavily phosphorylated on tyrosine residues upon CD5 costimulation, which is a prerequisite for its activation. A role for Vav in the CD5-induced signaling pathway is further supported by the findings that the expression of a dominant negative Vav mutant (Vav-C) completely abolishes the response to CD5 costimulation while the expression of a constitutively active Vav mutant [Vav(Δ1–65)] makes the CD5 costimulation signal superfluous. Wortmannin is unable to block the Vav(Δ1–65)- or Rac1 · V12-induced signals, indicating that both Vav and Rac1 function downstream from PI 3-kinase. Vav and Rac1 both act upstream from the CD5-induced activation of CaM kinase IV, since KN-62, an inhibitor of CaM kinases, and a dominant negative CaM kinase IV mutant block the Vav(Δ1–65)-and Rac1 · V12-mediated signals. We propose a model for the CD5-induced signaling pathway in which the PI 3-kinase lipid products, together with tyrosine phosphorylation, activate Vav, resulting in the activation of Rac1 by the Vav-mediated exchange of GDP for GTP.  相似文献   

4.
1. A high expression of angiotensin II receptors and of angiotensin-converting enzyme (ACE) activity was detected in confluent NIH 3T3 fibroblasts.2. Characterization with selective ligands, dithiothreitol, and GTPS, indicated that only the AT2 subtype was expressed.3. AT2 receptors and ACE expression were strictly dependent on the cell density and growth phase of the cells, with AT2 receptors being expressed earlier than ACE. In contrast, high expression of AT2 receptors irrespective of their growth state was observed in NIH 3T3 cells lacking contact inhibition upon neoplastic transformation with ras.4. Our results imply a possible relation of AT2 receptors to cell growth and cell–cell contact.  相似文献   

5.
6.
7.
8.
The growth arrest-specific gene, Gas-1, is preferentially expressed in quiescent NIH3T3 cells and inhibits DNA synthesis, suggesting that Gas-1 may be a tumor suppressor gene. When GAS1 cDNA, under the control of the strong constitutive CMV promoter, was transfected into NIH3T3 cells, no stable transfectant cell lines were produced, confirming that high levels of expression of GAS1 mRNA inhibit proliferation. GAS1, under the control of a dexamethasone-inducible promoter, was also transfected into NIH3T3 cells, resulting in normal numbers of transfectant clones. When expression of GAS1 mRNA was induced with dexamethasone, the growth rate was greatly inhibited. Morphological changes characteristic of growth arrest were also observed. To determine if antisense inhibition of expression of Gas-1 will transform normal fibroblasts, GAS1 cDNA, cloned in the antisense orientation, was transfected into NIH3T3 cells and expression of endogenous Gas-1 mRNA was inhibited. The GAS1-antisense cells had altered morphology and grew to a much higher saturation density than control cell lines with a loss of contact inhibition. However, there was no change in requirements for serum or any development of anchorage-independence. Antisense inhibition of expression of GAS1 is therefore insufficient to transform the cells, suggesting that additional genetic events are required for a fully malignant phenotype.  相似文献   

9.
Benzo[a]pyrene-7,8-diol-9,10-epoxide (B[a]PDE), the major metabolite of B[a]P, has been well recognized as one ubiquitous carcinogen, but the molecular mechanism involved in its carcinogenic effect remains obscure. In the present study, we found that bronchial epithelial cells (Beas-2B) and hepatocytes treated with B[a]PDE presented a significant increase of cyclin D1 expression. Moreover, Akt, p70s6k, and MAPKs including JNK, Erks, and p38 were notably activated in B[a]PDE-treated Beas-2B cells, whereas NF-κB, NFAT, and Egr-1 were not. Our results demonstrated that JNK and Erks were required in B[a]PDE-induced cyclin D1 expression because the inhibition of JNK or Erks by a selective chemical inhibitor or dominant negative mutant robustly impaired the cyclin D1 induction by B[a]PDE. Furthermore, we found that overexpression of the dominant negative mutant of p85 (regulatory subunit of phosphatidylinositol 3-kinase) or Akt dramatically suppressed B[a]PDE-induced JNK and Erk activation as well as cyclin D1 expression, suggesting that cyclin D1 induction by B[a]PDE is via the phosphatidylinositol 3-kinase/Akt/MAPK-dependent pathway. In addition, we clarified that p70s6k is also involved in B[a]PDE-induced cyclin D1 expression because rampamycin pretreatment dramatically reduced cyclin D1 induction by B[a]PDE. More importantly, we demonstrated that up-regulated cyclin D1 by B[a]PDE plays a critical role in oncogenic transformation and tumorigenesis of Beas-2B cells. These results not only broaden our knowledge of the molecular mechanism of B[a]PDE carcinogenicity but also lead to the further study of chemoprevention of B[a]PDE-associated human cancers.  相似文献   

10.
11.

Background

Connective tissue growth factor (CTGF; also known as CCN2) is an inflammatory mediator, and shows elevated levels in regions of severe injury and inflammatory diseases. CTGF is abundantly expressed in osteoarthritis (OA). However, the relationship between CTGF and IL-6 in OA synovial fibroblasts (OASFs) is mostly unknown.

Methodology/Principal Findings

OASFs showed significant expression of CTGF, and expression was higher than in normal SFs. OASFs stimulation with CTGF induced concentration-dependent increases in IL-6 expression. CTGF mediated IL-6 production was attenuated by αvβ5 integrin neutralized antibody and apoptosis signal-regulating kinase 1 (ASK1) shRNA. Pretreatment with p38 inhibitor (SB203580), JNK inhibitor (SP600125), AP-1 inhibitors (Curcumin and Tanshinone IIA), and NF-κB inhibitors (PDTC and TPCK) also inhibited the potentiating action of CTGF. CTGF-mediated increase of NF-κB and AP-1 luciferase activity was inhibited by SB203580 and SP600125 or ASK1 shRNA or p38 and JNK mutant.

Conclusions/Significance

Our results suggest that CTGF increased IL-6 production in OASFs via the αvβ5 integrin, ASK1, p38/JNK, and AP-1/NF-κB signaling pathways.  相似文献   

12.
Myoblast fusion is an essential step during myoblast differentiation that remains poorly understood. M-cadherin–dependent pathways that signal through Rac1 GTPase activation via the Rho-guanine nucleotide exchange factor (GEF) Trio are important for myoblast fusion. The ADP-ribosylation factor (ARF)6 GTPase has been shown to bind to Trio and to regulate Rac1 activity. Moreover, Loner/GEP100/BRAG2, a GEF of ARF6, has been involved in mammalian and Drosophila myoblast fusion, but the specific role of ARF6 has been not fully analyzed. Here, we show that ARF6 activity is increased at the time of myoblast fusion and is required for its implementation in mouse C2C12 myoblasts. Specifically, at the onset of myoblast fusion, ARF6 is associated with the multiproteic complex that contains M-cadherin, Trio, and Rac1 and accumulates at sites of myoblast fusion. ARF6 silencing inhibits the association of Trio and Rac1 with M-cadherin. Moreover, we demonstrate that ARF6 regulates myoblast fusion through phospholipase D (PLD) activation and phosphatidylinositol 4,5-bis-phosphate production. Together, these data indicate that ARF6 is a critical regulator of C2C12 myoblast fusion and participates in the regulation of PLD activities that trigger both phospholipids production and actin cytoskeleton reorganization at fusion sites.  相似文献   

13.
Novel techniques were used to determine when in the cell cycle of proliferating NIH 3T3 cells cellular Ras and cyclin D1 are required. For comparison, in quiescent cells, all four of the inhibitors of cell cycle progression tested (anti-Ras, anti-cyclin D1, serum removal, and cycloheximide) became ineffective at essentially the same point in G1 phase, approximately 4 h prior to the beginning of DNA synthesis. To extend these studies to cycling cells, a time-lapse approach was used to determine the approximate cell cycle position of individual cells in an asynchronous culture at the time of inhibitor treatment and then to determine the effects of the inhibitor upon recipient cells. With this approach, anti-Ras antibody efficiently inhibited entry into S phase only when introduced into cells prior to the preceding mitosis, several hours before the beginning of S phase. Anti-cyclin D1, on the other hand, was an efficient inhibitor when introduced up until just before the initiation of DNA synthesis. Cycloheximide treatment, like anti-cyclin D1 microinjection, was inhibitory throughout G1 phase (which lasts a total of 4 to 5 h in these cells). Finally, serum removal blocked entry into S phase only during the first hour following mitosis. Kinetic analysis and a novel dual-labeling technique were used to confirm the differences in cell cycle requirements for Ras, cyclin D1, and cycloheximide. These studies demonstrate a fundamental difference in mitogenic signal transduction between quiescent and cycling NIH 3T3 cells and reveal a sequence of signaling events required for cell cycle progression in proliferating NIH 3T3 cells.  相似文献   

14.
Type-I plasminogen activator inhibitor (PAI-1) is the primary inhibitor of both tissue- and urokinase-type plasminogen activators (t-PA, u-PA) and is thus a primary regulator of plasminogen activation and possibly of extracellular proteolysis. In anchorage-dependent cells, the PAI-1 gene was regulated by cell adhesion. PAI-1 gene expression was induced more evidently in cells adhered to the culture plate than in nonadherent cells. In this study, we investigated the signal pathway of the PAI-1 gene expression regulated by cell adhesion. We found the induction of both PAI-1 mRNA and protein, when cells adhered to culture dish, was inhibited by the PI-3 kinase specific inhibitors (Ly294002 and wortmannin). The cells seeded on collagen-1 coated plate with low serum further demonstrated that the PAI-1 gene expression was prolonged by the cell adhesion. The above-mentioned PI-3 kinase specific inhibitors also blocked the PAI-1 maintenance when cell adhered to collagen-1 coated plate. In addition, we found that both PI-3 kinase and its downstream molecule, Akt, were activated more evidently in adherent cells than in nonadherent cells. Furthermore, we transfected antisense oligodeoxynucleotides of Akt (AS-ODN-Akt) into cells to block the expression of Akt and found that the induction of PAI-1 mRNA was also inhibited. Hence, we conclude that the induction of PAI-1 gene expression is cell adhesion dependent and is through PI-3 kinase and Akt activation.  相似文献   

15.
PI3K/Akt signaling plays an important role in the regulation of cardiomyocyte death machinery, which can cause stress-induced cardiac dysfunction. Here, we report that apoptosis regulator through modulating IAP expression (ARIA), a recently identified transmembrane protein, regulates the cardiac PI3K/Akt signaling and thus modifies the progression of doxorubicin (DOX)-induced cardiomyopathy. ARIA is highly expressed in the mouse heart relative to other tissues, and it is also expressed in isolated rat cardiomyocytes. The stable expression of ARIA in H9c2 cardiac muscle cells increased the levels of membrane-associated PTEN and subsequently reduced the PI3K/Akt signaling and the downstream phosphorylation of Bad, a proapoptotic BH3-only protein. When challenged with DOX, ARIA-expressing H9c2 cells exhibited enhanced apoptosis, which was reversed by the siRNA-mediated silencing of Bad. ARIA-deficient mice exhibited normal heart morphology and function. However, DOX-induced cardiac dysfunction was significantly ameliorated in conjunction with reduced cardiomyocyte death and cardiac fibrosis in ARIA-deficient mice. Phosphorylation of Akt and Bad was substantially enhanced in the heart of ARIA-deficient mice even after treatment with DOX. Moreover, repressing the PI3K by cardiomyocyte-specific expression of dominant-negative PI3K (p110α) abolished the cardioprotective effects of ARIA deletion. Notably, targeted activation of ARIA in cardiomyocytes but not in endothelial cells reduced the cardiac PI3K/Akt signaling and exacerbated the DOX-induced cardiac dysfunction. These studies, therefore, revealed a previously undescribed mode of manipulating cardiac PI3K/Akt signaling by ARIA, thus identifying ARIA as an attractive new target for the prevention of stress-induced myocardial dysfunction.  相似文献   

16.
17.
Dendritic cells (DC) play a critical role in modulating antigen-specific immune responses elicited by T cells via engagement of the prototypic T cell costimulatory receptor CD28 by the cognate ligands CD80/CD86, expressed on DC. Although CD28 signaling in T cell activation has been well characterized, it has only recently been shown that CD80/CD86, which have no demonstrated binding domains for signaling proteins in their cytoplasmic tails, nonetheless also transduce signals to the DC. Functionally, CD80/CD86 engagement results in DC production of the pro-inflammatory cytokine IL-6, which is necessary for full T cell activation. However, ligation of CD80/CD86 by CTLA4 also induces DC production of the immunosuppressive enzyme indoleamine 2,3-dioxygenase (IDO), which depletes local pools of the essential amino acid tryptophan, resulting in blockade of T cell activation. Despite the significant role of CD80/CD86 in immunological processes and the seemingly opposing roles they play by producing IL-6 and IDO upon their activation, how CD80/CD86 signal remains poorly understood. We have now found that cross-linking CD80/CD86 in human DC activates the PI3K/AKT pathway. This results in phosphorylation/inactivation of its downstream target, FOXO3A, and alleviates FOXO3A-mediated suppression of IL-6 expression. A second event downstream of AKT phosphorylation is activation of the canonical NF-κB pathway, which induces IL-6 expression. In addition to these downstream pathways, we unexpectedly found that CD80/CD86-induced PI3K signaling is regulated by previously unrecognized cross-talk with NOTCH1 signaling. This cross-talk is facilitated by NOTCH-mediated up-regulation of the expression of prolyl isomerase PIN1, which in turn increases enzyme activity of casein kinase II. Subsequently, phosphatase and tensin homolog (which suppresses PI3K activity) is inactivated via phosphorylation by casein kinase II. This results in full activation of PI3K signaling upon cross-linking CD80/CD86. Similar to IL-6, we have found that CD80/CD86-induced IDO production by DC at late time points is also dependent upon the PI3K → AKT → NF-κB pathway and requires cross-talk with NOTCH signaling. These data further suggest that the same signaling pathways downstream of DC CD80/CD86 cross-linking induce early IL-6 production to enhance T cell activation, followed by later IDO production to self-limit this activation. In addition to characterizing the pathways downstream of CD80/CD86 in IL-6 and IDO production, identification of a novel cross-talk between NOTCH1 and PI3K signaling may provide new insights in other biological processes where PI3K signaling plays a major role.  相似文献   

18.
汉坦病毒是引起肾综合征出血热(HFRS)和汉坦病毒型肺炎综合征(HPS)的主要病原体.由S基因编码的核蛋白(NP)主要与机体的细胞免疫有关,并调节病毒的复制及诱导细胞程序性死亡.构建了汉坦病毒Z10株核蛋白cDNA与含有pac基因的反转录病毒鼠干细胞病毒(MSCV)重组体MSCV-FlagNP,通过磷酸钙转录法导入产病毒的包装细胞系BOSC23中,产生完整的重组MSCV-FlagNP病毒.然后以重组病毒感染NIH 3T3细胞,利用Puromycin的选择特性(pac基因)对感染细胞进行连续压力筛选,获得了转核蛋白抗性细胞.利用Southern blot和PCR方法分别对核蛋白基因在抗性细胞染色体整合情况及其完整性进行了鉴定.并且用Western blot在抗性细胞中可检测到核蛋白的表达.进一步以Flag单克隆抗体介导的免疫荧光染色联合共聚焦激光扫描荧光显微镜,分析了内源性Flag融合核蛋白在抗性细胞内分布,发现核蛋白主要分布于胞浆及胞核周围区,并且部分核蛋白可聚集形成胞浆包涵体.转核蛋白基因细胞模型的建立,对进一步研究汉坦病毒核蛋白功能以及病毒复制机制有重要意义.  相似文献   

19.
Abstract: Glial cell line-derived neurotrophic factor (GDNF) is a potent survival factor for midbrain dopaminergic neurons. To begin to understand the intracellular signaling pathways used by GDNF, we investigated the role of phosphatidylinositol 3-kinase activity in GDNF-stimulated cellular function and differentiation of dopaminergic neurons. We found that treatment of dopaminergic neuron cultures with 10 ng/ml GDNF induced maximal levels of Ret phosphorylation and produced a profound increase in phosphatidylinositol 3-kinase activity, as measured by western blot analysis and lipid kinase assays. Treatment with 1 µ M 2-(4-morpholinyl)-8-phenylchromone (LY294002) or 100 n M wortmannin, two distinct and potent inhibitors of phosphatidylinositol 3-kinase activity, completely inhibited GDNF-induced phosphatidylinositol 3-kinase activation, but did not affect Ret phosphorylation. Furthermore, we examined specific biological functions of dopaminergic neurons: dopamine uptake activity and morphological differentiation of tyrosine hydroxylase-immunoreactive neurons. GDNF significantly increased dopamine uptake activity and promoted robust morphological differentiation. Treatment with LY294002 completely abolished the GDNF-induced increases of dopamine uptake and morphological differentiation of tyrosine hydroxylase-immunoreactive neurons. Our findings show that GDNF-induced differentiation of dopaminergic neurons requires phosphatidylinositol 3-kinase activation.  相似文献   

20.
Using a procedure to stain cells simultaneously for cyclin B1 protein and DNA, we have examined cyclin B1 expression by flow cytometry in human cells under a variety of perturbing and nonperturbing conditions. The method described is useful for measuring relative differences in cyclin B level (immunochemically detectable epitope) as a function of cell cycle position on an individual cell basis and thus to examine cell cycle-related changes in cyclin B expression without prior cell synchronization. We show that in HeLa S3 cells, cyclin B1 accumulates in cells only after they become 4C and have resided in G2 for a short period of time. During colcemid-induced mitotic arrest cyclin B1 continues to accumulate in HeLa S3 cells, and under specific conditions of aphidicolin-induced unbalanced cell growth induced, cyclin B accumulates to supranormal levels prior to mitosis. Flow cytometric analysis of cyclin B expression and DNA content permits detailed examination of the effects of cell cycle perturbations on cyclin B expression under a variety of conditions.  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