Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 18 de 18
Filter
Add more filters











Publication year range
2.
Cell Death Dis ; 11(12): 1061, 2020 12 12.
Article in English | MEDLINE | ID: mdl-33311453

ABSTRACT

Although the survival rate of patients with cancer have increased due to the use of current chemotherapeutic agents, adverse effects of cancer therapy remain a concern. The reversal of drug resistance, reduction in harmful side effects and accelerated increase in efficiency have often been addressed in the development of combination therapeutics. Tazemetostat (EPZ-6438), a histone methyltransferase EZH2 selective inhibitor, was approved by the FDA for the treatment of advanced epithelioid sarcoma. However, the effect of tazemetostat on colorectal cancer (CRC) and 5-FU sensitivity remains unclear. In this study, the enhancement of tazemetostat on 5-FU sensitivity was examined in CRC cells. Our findings demonstrated that tazemetostat combined with 5-FU exhibits synergistic antitumor function in vitro and in vivo in CRC cells. In addition, tazemetostat promotes PUMA induction through the ROS/ER stress/CHOP axis. PUMA depletion attenuates the antitumor effect of the combination therapy. Therefore, tazemetostat may be a novel treatment to improve the sensitivity of tumors to 5-FU in CRC therapy. In conclusion, the combination of 5-FU and tazemetostat shows high therapeutic possibility with reduced unfavorable effects.


Subject(s)
Apoptosis Regulatory Proteins/genetics , Colorectal Neoplasms/drug therapy , Colorectal Neoplasms/genetics , Enhancer of Zeste Homolog 2 Protein/antagonists & inhibitors , Fluorouracil/therapeutic use , Gene Expression Regulation, Neoplastic , Proto-Oncogene Proteins/genetics , Up-Regulation/genetics , Animals , Apoptosis/drug effects , Apoptosis Regulatory Proteins/metabolism , Benzamides/pharmacology , Benzamides/therapeutic use , Biphenyl Compounds/pharmacology , Biphenyl Compounds/therapeutic use , Cell Line, Tumor , Colorectal Neoplasms/pathology , Endoplasmic Reticulum Stress/drug effects , Enhancer of Zeste Homolog 2 Protein/metabolism , Fluorouracil/pharmacology , Humans , Mice, Inbred BALB C , Mice, Nude , Morpholines/pharmacology , Morpholines/therapeutic use , Phenylurea Compounds/pharmacology , Phenylurea Compounds/therapeutic use , Proto-Oncogene Proteins/metabolism , Pyridines/pharmacology , Pyridines/therapeutic use , Pyridones/pharmacology , Pyridones/therapeutic use , Reactive Oxygen Species/metabolism , Transcription Factor CHOP/metabolism , Tumor Suppressor Protein p53/metabolism , Up-Regulation/drug effects , Xenograft Model Antitumor Assays
3.
Cell Death Dis ; 11(10): 902, 2020 10 23.
Article in English | MEDLINE | ID: mdl-33097691

ABSTRACT

HCC (hepatocellular carcinoma) is a major health threat for the Chinese population and has poor prognosis because of strong resistance to chemotherapy in patients. For instance, a considerable challenge for the treatment of HCC is sorafenib resistance. The aberrant glucose metabolism in cancer cells aerobic glycolysis is associated with resistance to chemotherapeutic agents. Drug-resistance cells and tumors were exposed to sorafenib to establish sorafenib-resistance cell lines and tumors. Western blotting and real-time PCR or IHC staining were used to analyze the level of CLCF1 in the sorafenib resistance cell lines or tumors. The aerobic glycolysis was analyzed by ECAR assay. The mechanism mediating the high expression of CLCF1 in sorafenib-resistant cells and its relationships with miR-130-5p was determined by bioinformatic analysis, dual luciferase reporter assays, real-time PCR, and western blotting. The in vivo effect was evaluated by xenografted with nude mice. The relation of CLCF1 and miR-30a-5p was determined in patients' samples. In this study, we report the relationship between sorafenib resistance and increased glycolysis in HCC cells. We also show the vital role of CLCF1 in promoting glycolysis by activating PI3K/AKT signaling and its downstream genes, thus participating in glycolysis in sorafenib-resistant HCC cells. Furthermore, we also show that miR-30a-5p directly targets CLCF1 and that sorafenib-mediated suppression of miR-30a-5p results in the upregulation of CLCF1 in HCC cells resistant to sorafenib. We also found that when a cholesterol modified agomiR-30a-5p was delivered systemically to mice harboring sorafenib-resistant HCC tumors, tumor growth decreased significantly. There is an uncharacterized mechanism of biochemical resistance to hormone therapies orchestrated by the miR-30a-5p/CLCF1 axis to mediate sorafenib resistance and aerobic glycolysis in HCC. Therefore, this study indicates that targeting the miR-30a-5p/CLCF1 axis may hold promise for therapeutic intervention in HCC sorafenib resistance patients.


Subject(s)
Carcinoma, Hepatocellular/genetics , Drug Resistance, Neoplasm/genetics , Liver Neoplasms/genetics , MicroRNAs/genetics , Sorafenib , Animals , Antineoplastic Agents/pharmacology , Carcinoma, Hepatocellular/pathology , Cell Line, Tumor , Cell Movement/genetics , Cell Proliferation/drug effects , Cell Proliferation/genetics , Drug Resistance, Neoplasm/drug effects , Gene Expression Regulation, Neoplastic/drug effects , Gene Expression Regulation, Neoplastic/genetics , Humans , Liver Neoplasms/pathology , Mice , Signal Transduction/drug effects , Sorafenib/metabolism , Sorafenib/pharmacology
4.
J Cell Mol Med ; 24(18): 10830-10841, 2020 09.
Article in English | MEDLINE | ID: mdl-32735377

ABSTRACT

Overexpression of P2X7R has been observed in several tumours and is related to cancer advancement and metastasis. However, the role of P2X7R in colorectal cancer (CRC) patients is not well understood. In the current study, overexpression of P2X7R and the effects at the molecular and functional levels in CRC were assessed in a mouse orthotopic model. Functional assays, such as the CCK-8 assay, wound healing and transwell assay, were used to determine the biological role of P2X7R in CRC cells. CSC-related genes and properties were detected via sphere formation and real-time PCR assays. The underlying mechanisms were explored by Western blotting, real-time PCR and Flow cytometry. In this study, we found that overexpression of P2X7R increases in the in vivo growth of tumours. P2X7R overexpression also increased CD31, VEGF and concurrent angiogenesis. P2X7R up-regulates aldehyde dehydrogenase-1 (ALDH1) and CSC characteristics. Transplanted tumour cells with P2X7R overexpression stimulated cytokines to recruit tumour-associated macrophage (TAMs) to increase the growth of tumours. We also found that the NF-κB signalling pathway is involved in P2X7R-induced cytokine up-regulation. P2X7R promotes NF-κB-dependent cytokine induction, which leads to TAM recruitment to control tumour growth and advancement and remodelling of the stroma. Our findings demonstrate that P2X7R plays a key role in TAM recruitment, which may be a therapeutic target for CRC patients.


Subject(s)
Adenocarcinoma/physiopathology , Colorectal Neoplasms/physiopathology , NF-kappa B/metabolism , Neoplasm Proteins/physiology , Neovascularization, Pathologic/physiopathology , Receptors, Purinergic P2X7/physiology , Tumor-Associated Macrophages/physiology , Adenocarcinoma/immunology , Adenocarcinoma/metabolism , Adenocarcinoma/pathology , Animals , Cell Line, Tumor , Cell Movement , Colorectal Neoplasms/immunology , Colorectal Neoplasms/metabolism , Colorectal Neoplasms/pathology , Cytokines/metabolism , Gene Knockdown Techniques , Humans , Mice , Mice, Inbred BALB C , Neoplasm Invasiveness , Receptors, Purinergic P2X7/biosynthesis , Receptors, Purinergic P2X7/genetics , Recombinant Proteins/metabolism , Signal Transduction/physiology
5.
J Cell Mol Med ; 24(13): 7600-7608, 2020 07.
Article in English | MEDLINE | ID: mdl-32410348

ABSTRACT

MicroRNAs (miRNAs) have been found to play a key role in drug resistance. In the current study, we aimed to explore the potential role of miR-126 in trastuzumab resistance in breast cancer cells. We found that the trastuzumab-resistant cell lines SKBR3/TR and BT474/TR had low expression of miR-126 and increased ability to migrate and invade. The resistance, invasion and mobilization abilities of the cells resistant to trastuzumab were reduced by ectopic expression of miR-126 mimics. In comparison, inhibition of miR-126 in SKBR3 parental cells had the opposite effect of an increased resistance to trastuzumab as well as invasion and migration. It was also found that miR-126 directly targets PIK3R2 in breast cancer cells. PIK3R2-knockdown cells showed decreased resistance to trastuzumab, while overexpression of PIK3R2 increased trastuzumab resistance. In addition, our finding showed that overexpression of miR-126 reduced resistance to trastuzumab in the trastuzumab-resistant cells and that inhibition of the PIK3R2/PI3K/AKT/mTOR signalling pathway was involved in this effect. SKBR3/TR cells also showed increased sensitivity to trastuzumab mediated by miR-126 in vivo. In conclusion, the above findings demonstrated that overexpression of miR-126 or down-regulation of its target gene may be a potential approach to overcome trastuzumab resistance in breast cancer cells.


Subject(s)
Breast Neoplasms/drug therapy , Breast Neoplasms/genetics , Drug Resistance, Neoplasm/genetics , MicroRNAs/metabolism , Phosphatidylinositol 3-Kinases/metabolism , Proto-Oncogene Proteins c-akt/metabolism , TOR Serine-Threonine Kinases/metabolism , Trastuzumab/therapeutic use , Animals , Base Sequence , Cell Line, Tumor , Cell Movement/drug effects , Cell Movement/genetics , Down-Regulation/drug effects , Down-Regulation/genetics , Female , Gene Expression Regulation, Neoplastic/drug effects , Humans , Mice, Nude , MicroRNAs/genetics , Neoplasm Invasiveness , Phosphatidylinositol 3-Kinases/genetics , Signal Transduction/drug effects , Trastuzumab/pharmacology
6.
Oncotarget ; 6(38): 41324-38, 2015 Dec 01.
Article in English | MEDLINE | ID: mdl-26510914

ABSTRACT

The role of death receptor 5 (DR5), a well-known cell surface pro-apoptotic protein, in the negative regulation of invasion and metastasis of human cancer cells and the underlying mechanisms are largely unknown and were hence the focus of this study. In this report, we have demonstrated that DR5 functions to suppress invasion and metastasis of human cancer cells, as evidenced by enhanced cancer cell invasion and metastasis upon genetic suppression of DR5 either by gene knockdown or knockout. When DR5 is suppressed, FADD and caspase-8 may recruit and stabilize TRAF2 to form a metastasis and invasion signaling complex, resulting in activation of ERK and JNK/AP-1 signaling that mediate the elevation and activation of matrix metalloproteinase-1 (MMP1) and eventual promotion of cancer invasion and metastasis. Our findings thus highlight a novel non-apoptotic function of DR5 as a suppressor of human cancer cell invasion and metastasis and suggest a basic working model elucidating the underlying biology.


Subject(s)
Caspase 8/metabolism , MAP Kinase Signaling System , Neoplasms/metabolism , Receptors, TNF-Related Apoptosis-Inducing Ligand/metabolism , TNF Receptor-Associated Factor 2/metabolism , Animals , Blotting, Western , Cell Line, Tumor , Enzyme Activation , Fas-Associated Death Domain Protein/metabolism , Female , HCT116 Cells , HEK293 Cells , Humans , Matrix Metalloproteinase 1/metabolism , Mice, Nude , Neoplasm Invasiveness , Neoplasm Metastasis , Neoplasms/genetics , Neoplasms/therapy , RNA Interference , RNAi Therapeutics/methods , Receptors, TNF-Related Apoptosis-Inducing Ligand/genetics , Xenograft Model Antitumor Assays/methods
7.
Oncotarget ; 6(19): 17532-42, 2015 Jul 10.
Article in English | MEDLINE | ID: mdl-26009898

ABSTRACT

Carfilzomib (CFZ) is a second generation proteasome inhibitor approved for the treatment of patients with multiple myeloma. It induces apoptosis in human cancer cells; but the underlying mechanisms remain undefined. In the present study, we show that CFZ decreases the survival of several human cancer cell lines and induces apoptosis. Induction of apoptosis by CFZ occurs, at least in part, due to activation of the extrinsic apoptotic pathway, since FADD deficiency protected cancer cells from undergoing apoptosis. CFZ increased total and cell surface levels of DR5 in different cancer cell lines; accordingly it enhanced TRAIL-induced apoptosis. DR5 deficiency protected cancer cells from induction of apoptosis by CFZ either alone or in combination with TRAIL. These data together convincingly demonstrate that DR5 upregulation is a critical mechanism accounting for CFZ-induced apoptosis and enhancement of TRAIL-induced apoptosis. CFZ inhibited the degradation of DR5, suggesting that DR5 stabilization contributes to CFZ-induced DR5 upregulation. In summary, the present study highlights the important role of DR5 upregulation in CFZ-induced apoptosis and enhancement of TRAIL-induced apoptosis in human cancer cells.


Subject(s)
Antineoplastic Agents/pharmacology , Apoptosis/drug effects , Oligopeptides/pharmacology , Proteasome Inhibitors/pharmacology , Receptors, TNF-Related Apoptosis-Inducing Ligand/metabolism , Blotting, Western , Cell Line, Tumor , Humans , Polymerase Chain Reaction , RNA, Small Interfering , Transfection , Up-Regulation
8.
Reprod Sci ; 18(10): 983-9, 2011 Oct.
Article in English | MEDLINE | ID: mdl-21693773

ABSTRACT

Dynein light chain, Tctex-type 3 (DYNLT3), is a member of the cytoplasmic dynein DYNLT light chain family and has been reported to have a potential role in chromosome congression in human mitosis. However, its role in mammalian meiosis is unclear. In this study, we examined its localization, expression, and functions in mouse oocyte meiosis. Immunofluorescent staining showed that DYNLT3 was restricted to the germinal vesicle and associated with kinetochores at the germinal vesicle breakdown stage, metaphase I and metaphase II. The expression level of DYNLT3 was similar at all meiotic stages. Depletion of DYNLT3 by antibody injection resulted in chromosome misalignment and decrease of the polar body extrusion rate. We further found that DYNLT3-depleted oocytes displayed kinetochore-microtubule detachments. Chromosome-spread experiments showed that depletion of DYNLT3 inhibited the metaphase-anaphase transition by preventing homologous chromosome segregation in meiosis I. Our data suggest that DYNLT3 is required for chromosome alignment and homologous chromosome segregation during mouse oocyte meiosis.


Subject(s)
Chromosome Segregation/physiology , Dyneins/genetics , Kinetochores/physiology , Meiosis/physiology , Oocytes/cytology , Animals , Blotting, Western , Female , Mice , Mice, Inbred ICR , Microscopy, Confocal , Oocytes/ultrastructure , RNA, Messenger/chemistry , RNA, Messenger/genetics , Reverse Transcriptase Polymerase Chain Reaction
9.
Cell Cycle ; 10(11): 1861-70, 2011 Jun 01.
Article in English | MEDLINE | ID: mdl-21552007

ABSTRACT

GM130, a cis-Golgi protein, plays key roles in various mitotic events, but its function in mammalian oocyte meiosis remains unknown. In this study, we found that GM130 was localized to the spindle poles at both metaphase I and metaphase II stages and associated with the midbody at telophase I stage. The association of GM130 with spindle poles was further confirmed by its colocalization with the centrosome-associated proteins, MEK1/2. By nocodazole treatment, we clarified that GM130 localization was consistently dependent on spindle assembly. Then we investigated the possible function of GM130 by specific morpholino microinjection. This treatment caused abnormal spindle formation, and decreased first polar body extrusion. Our results showed that knockdown of GM130 impaired the localization of MTOCs proteins γ-tubulin and Plk1. Using live cell imaging we observed that depletion of GM130 affected spindle migration and resulted in elongated spindle and large polar body extrusion. We further found that depletion of GM130 blocked p-MEK1/2 accumulation at the spindle poles. And, it was shown that GM130 detached from the spindle poles in oocytes treated with MEK specific inhibitor U0126. Taken together, our results suggested that GM130 regulates microtubule organization and might cooperate with the MAPK pathway to play roles in spindle organization, migration and asymmetric division during mouse oocyte maturation.


Subject(s)
Autoantigens/physiology , Cell Division , Cytoskeleton/metabolism , Meiosis , Membrane Proteins/physiology , Oocytes/cytology , Spindle Apparatus , Animals , Mice , Microtubules , Mitogen-Activated Protein Kinase 1 , Mitogen-Activated Protein Kinases
10.
PLoS One ; 6(3): e16781, 2011 Mar 08.
Article in English | MEDLINE | ID: mdl-21408143

ABSTRACT

Icaritin, a compound from Epimedium Genus, has selective estrogen receptor (ER) modulating activities, and possess anti-tumor activity. Here, we examined icaritin effect on cell growth of human endometrial cancer Hec1A cells and found that icaritin potently inhibited proliferation of Hec1A cells. Icaritin-inhibited cell growth was associated with increased levels of p21 and p27 expression and reduced cyclinD1 and cdk 4 expression. Icaritin also induced cell apoptosis accompanied by activation of caspases as evidenced by the cleavage of endogenous substrate Poly (ADP-ribose) polymerase (PARP) and cytochrome c release, which was abrogated by pretreatment with the pan-caspase inhibitor z-VAD-fmk. Icaritin treatment also induced expression of pro-apoptotic protein Bax with a concomitant decrease of Bcl-2 expression. Furthermore, icaritin induced sustained phosphorylation of extracellular signal-regulated kinase1/2 (the MAPK/ ERK1/2) in Hec1A cells and U0126, a specific MAP kinase kinase (MEK1/2) inhibitor, blocked the ERK1/2 activation by icaritin and abolished the icaritin-induced growth inhibition and apoptosis. Our results demonstrated that icaritin induced sustained ERK 1/2 activation and inhibited growth of endometrial cancer Hec1A cells, and provided a rational for preclinical and clinical evaluation of icaritin for endometrial cancer therapy.


Subject(s)
Apoptosis/drug effects , Endometrial Neoplasms/enzymology , Endometrial Neoplasms/pathology , Flavonoids/pharmacology , Mitogen-Activated Protein Kinase 1/metabolism , Mitogen-Activated Protein Kinase 3/metabolism , Caspases/biosynthesis , Cell Cycle/drug effects , Cell Line, Tumor , Cell Proliferation/drug effects , Endometrial Neoplasms/drug therapy , Enzyme Activation/drug effects , Enzyme Induction/drug effects , Female , Flavonoids/chemistry , Flavonoids/therapeutic use , Humans , MAP Kinase Signaling System/drug effects , Proto-Oncogene Proteins c-bcl-2/metabolism
11.
Microsc Microanal ; 17(2): 197-205, 2011 Apr.
Article in English | MEDLINE | ID: mdl-21281539

ABSTRACT

It is well known that c-Jun N-terminal kinase (JNK) plays pivotal roles in various mitotic events, but its function in mammalian oocyte meiosis remains unknown. In this study, we found that no specific JNK2 signal was detected in germinal vesicle stage. JNK2 was associated with the spindles especially the spindle poles and cytoplasmic microtubule organizing centers at prometaphase I, metaphase I, and metaphase II stages. JNK2 became diffusely distributed and associated with the midbody at telophase I stage. Injection of myc-tagged JNK2α1 mRNA into oocytes also revealed its localization on spindle poles. The association of JNK2 with spindle poles was further confirmed by colocalization with the centrosomal proteins, γ-tubulin and Plk1. Nocodazole treatment showed that JNK2 may interact with Plk1 to regulate the spindle assembly. Then we investigated the possible function of JNK2 by JNK2 antibody microinjection and JNK specific inhibitor SP600125 treatment. These two manipulations caused abnormal spindle formation and decreased the rate of first polar body (PB1) extrusion. In addition, inhibition of JNK2 resulted in impaired localization of Plk1. Taken together, our results suggest that JNK2 plays an important role in spindle assembly and PB1 extrusion during mouse oocyte meiotic maturation.


Subject(s)
Meiosis , Mitogen-Activated Protein Kinase 9/metabolism , Oocytes/cytology , Oocytes/enzymology , Oogenesis , Spindle Apparatus/enzymology , Animals , Cell Cycle Proteins/genetics , Cell Cycle Proteins/metabolism , Centrosome/enzymology , Centrosome/metabolism , Female , Mice , Mice, Inbred ICR , Mitogen-Activated Protein Kinase 9/genetics , Protein Binding , Protein Serine-Threonine Kinases/genetics , Protein Serine-Threonine Kinases/metabolism , Protein Transport , Proto-Oncogene Proteins/genetics , Proto-Oncogene Proteins/metabolism , Spindle Apparatus/genetics , Polo-Like Kinase 1
12.
PLoS One ; 5(12): e14242, 2010 Dec 07.
Article in English | MEDLINE | ID: mdl-21151922

ABSTRACT

The objective of this study was to evaluate the feasibility of preserving porcine oocytes without freezing. To optimize preservation conditions, porcine cumulus-oocyte complexes (COCs) were preserved in TCM-199, porcine follicular fluid (pFF) and FCS at different temperatures (4°C, 20°C, 25°C, 27.5°C, 30°C and 38.5°C) for 1 day, 2 days or 3 days. After preservation, oocyte morphology, germinal vesicle (GV) rate, actin cytoskeleton organization, cortical granule distribution, mitochondrial translocation and intracellular glutathione level were evaluated. Oocyte maturation was indicated by first polar body emission and spindle morphology after in vitro culture. Strikingly, when COCs were stored at 27.5°C for 3 days in pFF or FCS, more than 60% oocytes were still arrested at the GV stage and more than 50% oocytes matured into MII stages after culture. Almost 80% oocytes showed normal actin organization and cortical granule relocation to the cortex, and approximately 50% oocytes showed diffused mitochondria distribution patterns and normal spindle configurations. While stored in TCM-199, all these criteria decreased significantly. Glutathione (GSH) level in the pFF or FCS group was higher than in the TCM-199 group, but lower than in the non-preserved control group. The preserved oocytes could be fertilized and developed to blastocysts (about 10%) with normal cell number, which is clear evidence for their retaining the developmental potentiality after 3d preservation. Thus, we have developed a simple method for preserving immature pig oocytes at an ambient temperature for several days without evident damage of cytoplasm and keeping oocyte developmental competence.


Subject(s)
Cumulus Cells/cytology , Oocytes/cytology , Preservation, Biological/methods , Actins/chemistry , Animals , Blastocyst/cytology , Culture Media/metabolism , Female , Follicular Fluid/metabolism , Glutathione/metabolism , Mitochondria/metabolism , Oocytes/metabolism , Organ Preservation Solutions , Swine , Temperature
13.
PLoS One ; 5(11): e15408, 2010 Nov 04.
Article in English | MEDLINE | ID: mdl-21079811

ABSTRACT

BACKGROUND: Recently, a variant of ER-α, ER-α36 was identified and cloned. ER-α36 lacks intrinsic transcription activity and mainly mediates non-genomic estrogen signaling. The purpose of this study was to investigate the function and the underlying mechanisms of ER-α36 in growth regulation of endometrial Ishikawa cancer cells. METHODS: The cellular localization of ER-α36 and ER-α66 were determined by immunofluorescence in the Ishikawa cells. Ishikawa endometrial cancer control cells transfected with an empty expression vector, Ishikawa cells with shRNA knockdown of ER-α36 (Ishikawa/RNAiER36) and Ishikawa cells with shRNA knockdown of ER-α66 (Ishikawa/RNAiER66) were treated with E2 and E2-conjugated to bovine serum albumin (E2-BSA, membrane impermeable) in the absence and presence of different kinase inhibitors HBDDE, bisindolylmaleimide, rottlerin, H89 and U0126. The phosphorylation levels of signaling molecules and cyclin D1/cdk4 expression were examined with Western blot analysis and cell growth was monitored with the MTT assay. RESULTS: Immunofluorescence staining of Ishikawa cells demonstrated that ER-α36 was expressed mainly on the plasma membrane and in the cytoplasm, while ER-α66 was predominantly localized in the cell nucleus. Both E2 and E2-BSA rapidly activated PKCδ not PKCα in Ishikawa cells, which could be abrogated by ER-α36 shRNA expression. E2-and E2-BSA-induced ERK phosphorylation required ER-α36 and PKCδ. However, only E2 was able to induce Camp-dependent protein kinase A (PKA) phosphorylation. Furthermore, E2 enhances cyclin D1/cdk4 expression via ER-α36. CONCLUSION: E2 activates the PKCδ/ERK pathway and enhances cyclin D1/cdk4 expression via the membrane-initiated signaling pathways mediated by ER-α36, suggesting a possible involvement of ER-α36 in E2-dependent growth-promoting effects in endometrial cancer cells.


Subject(s)
Cell Proliferation/drug effects , Estrogen Receptor alpha/metabolism , Estrogens/pharmacology , Extracellular Signal-Regulated MAP Kinases/metabolism , Protein Kinase C-delta/metabolism , Blotting, Western , Cell Line, Tumor , Cyclic AMP-Dependent Protein Kinases/metabolism , Cyclin D1/metabolism , Cyclin-Dependent Kinase 4/metabolism , Endometrial Neoplasms/genetics , Endometrial Neoplasms/metabolism , Endometrial Neoplasms/pathology , Enzyme Activation/drug effects , Estrogen Receptor alpha/genetics , Female , Humans , Phosphorylation/drug effects , Protein Isoforms/genetics , Protein Isoforms/metabolism , RNA Interference , Serum Albumin, Bovine/pharmacology , Signal Transduction/drug effects
14.
PLoS One ; 5(9)2010 Sep 29.
Article in English | MEDLINE | ID: mdl-20927325

ABSTRACT

ERK3 (extracellular signal-regulated kinase 3) is an atypical member of the mitogen-activated protein (MAP) kinase family of serine/threonine kinases. Little is known about its function in mitosis, and even less about its roles in mammalian oocyte meiosis. In the present study, we examined the localization, expression and functions of ERK3 during mouse oocyte meiotic maturation. Immunofluorescent analysis showed that ERK3 localized to the spindles from the pre-MI stage to the MII stage. ERK3 co-localized with α-tubulin on the spindle fibers and asters in oocytes after taxol treatment. Deletion of ERK3 by microinjection of ERK3 morpholino (ERK3 MO) resulted in oocyte arrest at the MI stage with severely impaired spindles and misaligned chromosomes. Most importantly, the spindle assembly checkpoint protein BubR1 could be detected on kinetochores even in oocytes cultured for 10 h. Low temperature treatment experiments indicated that ERK3 deletion disrupted kinetochore-microtubule (K-MT) attachments. Chromosome spreading experiments showed that knock-down of ERK3 prevented the segregation of homologous chromosomes. Our data suggest that ERK3 is crucial for spindle stability and required for the metaphase-anaphase transition in mouse oocyte maturation.


Subject(s)
Anaphase , Meiosis , Metaphase , Mitogen-Activated Protein Kinase 6/metabolism , Oocytes/cytology , Oocytes/enzymology , Animals , Female , Mice , Mitogen-Activated Protein Kinase 6/genetics , Protein Transport
15.
PLoS One ; 5(6): e11247, 2010 Jun 28.
Article in English | MEDLINE | ID: mdl-20596525

ABSTRACT

MAPK-activated protein kinase 2 (MK2), a direct substrate of p38 MAPK, plays key roles in multiple physiological functions in mitosis. Here, we show for the first time the unique distribution pattern of MK2 in meiosis. Phospho-MK2 was localized on bipolar spindle minus ends and along the interstitial axes of homologous chromosomes extending over centromere regions and arm regions at metaphase of first meiosis (MI stage) in mouse oocytes. At metaphase of second meiosis (MII stage), p-MK2 was localized on the bipolar spindle minus ends and at the inner centromere region of sister chromatids as dots. Knockdown or inhibition of MK2 resulted in spindle defects. Spindles were surrounded by irregular nondisjunction chromosomes, which were arranged in an amphitelic or syntelic/monotelic manner, or chromosomes detached from the spindles. Kinetochore-microtubule attachments were impaired in MK2-deficient oocytes because spindle microtubules became unstable in response to cold treatment. In addition, homologous chromosome segregation and meiosis progression were inhibited in these oocytes. Our data suggest that MK2 may be essential for functional meiotic bipolar spindle formation, chromosome segregation and proper kinetochore-microtubule attachments.


Subject(s)
Kinetochores/metabolism , MAP Kinase Kinase 2/metabolism , Meiosis , Microtubules/metabolism , Spindle Apparatus , Animals , Mice , Nocodazole/pharmacology , Nondisjunction, Genetic , Oocytes/cytology , Oocytes/drug effects , Paclitaxel/pharmacology , Subcellular Fractions/enzymology
16.
Cell Cycle ; 9(13): 2640-6, 2010 Jul 01.
Article in English | MEDLINE | ID: mdl-20543581

ABSTRACT

Sumoylation is an important post-translational modification in which SUMO (small ubiquitin-related modifier) proteins are bonded covalently to their substrates. Studies on the roles of sumoylation in cell cycle regulation have been emerging in both mitosis from yeast to mammals and meiosis in budding yeast, but the functions of sumoylation in mammalian meiosis, especially in oocyte meiotic maturation are not well known. Here, we examined the localization and expression of SUMO-1 and SUMO-2/3, the two basic proteins in the sumoylation pathway and investigated their roles through over-expression of Senp2 during mouse oocyte maturation. Immunofluorescent staining revealed differential patterns of SUMO-1 and SUMO-2/3 localization: SUMO-1 was localized to the spindle poles in prometaphase I, MI and MII stages, around the separating homologues in anaphase I and telophase I stages of first meiosis, while SUMO-2/3 was mainly concentrated near centromeres during mouse oocyte maturation. Immunoblot analysis uncovered the different expression profiles of SUMO-1 and SUMO-2/3 modified proteins during mouse oocyte maturation. Over-expression of Senp2, a SUMO-specific isopeptidase, caused changes of SUMO-modified proteins and led to defects in MII spindle organization in mature eggs. These results suggest that the SUMO pathway may play an indispensable role during mouse oocyte meiotic maturation.


Subject(s)
Cell Differentiation , Oocytes/cytology , Oocytes/metabolism , Signal Transduction , Small Ubiquitin-Related Modifier Proteins/metabolism , Animals , Cysteine Endopeptidases , Female , Gene Expression Profiling , Meiosis , Mice , Multienzyme Complexes/metabolism , Protein Transport , Spindle Apparatus/metabolism , Subcellular Fractions/metabolism , Sumoylation , Time Factors
17.
Cell Cycle ; 9(11): 2230-6, 2010 Jun 01.
Article in English | MEDLINE | ID: mdl-20505367

ABSTRACT

Spindly was first identified in Drosophila; its homologues are termed SPDL-1 in Caenorhabditis elegans and Hs Spindly/hSpindly in humans. In all species, Spindly and its homologues function by recruiting dynein to kinetochores and silencing SAC in mitosis of somatic cells. Depletion of Spindly causes an extensive metaphase arrest during somatic mitoses in Drosophila, C. elegans and humans. In Drosophila, Spindly is required for shedding of Rod and Mad2 from the kinetochores in metaphase; in C. elegans, SPDL-1 presides over the recruitment of dynein and MDF-1 to the kinetochores; in humans, Hs Spindly is required for recruiting both dynein and dynactin to kinetochores but it is dispensable for removal of checkpoint proteins from kinetochores. The present study was designed to investigate the localization and function of the Spindly homologue (mSpindly) during mouse oocyte meiotic maturation by immunofluorescent analysis, and by overexpression and knockdown of mSpindly. We found that mSpindly was typically localized to kinetochores when chromatin condensed into chromosomes after GVBD. In metaphase of both first meiosis and second meiosis, mSpindly was localized not only to kinetochores but also to the spindle poles. Overexpression of mSpindly did not affect meiotic progression, but its depletion resulted in an arrest of the pro-MI/MI stage, failure of anaphase entry and subsequent polar body emission, and in abnormal spindle morphology and misaligned chromosomes. Our data suggest that mSpindly participates in SAC silencing and in spindle formation as a recruiter and/or a transporter of kinetochore proteins in mouse oocytes, but that it needs to cooperate with other factors to fulfill its function.


Subject(s)
Cell Cycle Proteins/analysis , Meiosis , Microtubule-Associated Proteins/analysis , Oocytes/cytology , Animals , Cell Cycle Proteins/metabolism , Cell Cycle Proteins/physiology , Chromosome Painting , Kinetochores/metabolism , Metaphase , Mice , Microtubule-Associated Proteins/metabolism , Microtubule-Associated Proteins/physiology , Oocytes/metabolism , Proto-Oncogene Proteins c-myc/genetics , Proto-Oncogene Proteins c-myc/metabolism , Spindle Apparatus/metabolism , Spindle Apparatus/physiology
18.
Xi Bao Yu Fen Zi Mian Yi Xue Za Zhi ; 23(12): 1144-6, 2007 Dec.
Article in Chinese | MEDLINE | ID: mdl-18062888

ABSTRACT

AIM: To construct the vector and express anti-HIV-1 envelope glycoprotein single chain Fv fragment in Pichia pastoris. METHODS: The target gene was digested from plasmid pET28-scFv and cloned into pichia pastoris vector via gene engineering and DNA recombination techniques. The recombinant plasmid was linearized and transferred into Pichia pastoris strains GS115 by electroporation. After positive recombinant was selected and expression was induced by methanol, the target protein was analyzed by RT-PCR, SDS-PAGE and double-antibody sandwich ELISA. RESULTS: High copies of transformant were obtained by phenotype determining and PCR amplification. RT-PCR and SDS-PAGE demonstrated the target protein was successfully expressed. And the yield account for about 18 percent of the total cell proteins. Double-antibody sandwich ELISA analysis proved that the recombinant scFv had good biological activities since it could be recognized and induce special immune respond with gp120 antigen. CONCLUSION: The scFv was expressed successfully. This research will lay the foundation for AIDS target therapy and further study of anti-HIV activities.


Subject(s)
HIV-1/immunology , Immunoglobulin Variable Region/genetics , Immunoglobulin Variable Region/immunology , Pichia/genetics , env Gene Products, Human Immunodeficiency Virus/immunology , Electrophoresis, Polyacrylamide Gel , Enzyme-Linked Immunosorbent Assay , Gene Expression , Immunoglobulin Variable Region/analysis , Immunoglobulin Variable Region/biosynthesis , Reverse Transcriptase Polymerase Chain Reaction
SELECTION OF CITATIONS
SEARCH DETAIL