Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 11 de 11
Filter
Add more filters










Publication year range
1.
Int J Biol Macromol ; 139: 298-306, 2019 Oct 15.
Article in English | MEDLINE | ID: mdl-31386870

ABSTRACT

In this study, two novel lipases, rice bran lipase 1 (RBL1) and rice bran lipase 2 (RBL2), were first identified in rice (Oryza sativa) bran. Through the purification by ammonium sulfate precipitation, ion-exchange chromatography and size-exclusion chromatography, RBL1 and RBL2 were purified to 36- and 339-fold with the final specific activity of 4.73 and 44.06 U/mg, respectively. The purified RBL1 and RBL2 had the molecular weight of 18.8 and 35.5 kDa, respectively. The Km values of RBL1 and RBL2 were 0.766 and 0.432 mM while catalytic efficiency (kcat/Km) values of RBL1 and RBL2 were 102.4 and 2559.3 s-1/mM, respectively. RBL1 and RBL2 both showed esterification activity, but had no transesterification activity. In a solvent-free system, RBL1 and RBL2 exhibited degree of esterification (ED) of 4.4% and 6.5%, respectively. These two novel lipases exerted great properties for their potentials in industrial applications. First, RBL1 and RBL2 showed both mild reaction pH of 7.0 and temperature of 35 °C and 50 °C, respectively. Secondly, they showed great tolerance to several organic solvents and detergents while RBL1 exhibited great pH stability across a very broad range of pH (pH 3-12). Lastly and most importantly, RBL1 and RBL2 both exhibited esterification activity better than a commercial lipase, Candida rugosa lipase (CRL), in a solvent-free system. In conclusion, two novel lipases, RBL1 and RBL2, are different from published native lipases in rice bran and may be alternative potential candidates of biocatalysts contributing to the development of diverse industrial application fields.


Subject(s)
Lipase/chemistry , Lipase/isolation & purification , Oryza/enzymology , Chromatography, Gel , Enzyme Activation , Esterification , Hydrogen-Ion Concentration , Kinetics , Lipolysis , Metals , Molecular Weight , Retinoblastoma-Like Protein p107/chemistry , Retinoblastoma-Like Protein p130/chemistry , Solvents , Substrate Specificity , Temperature
2.
Eur Rev Med Pharmacol Sci ; 22(8): 2343-2350, 2018 04.
Article in English | MEDLINE | ID: mdl-29762838

ABSTRACT

OBJECTIVE: Glioma is a tumor of the brain. Although the clinical regimens and surgical techniques for glioma have improved, therapies of advanced glioma remain challenging, carrying dismal overall survival and therapeutic success rates. Evidence has shown that miRNAs played important roles in glioma development. The current study aimed at investigating the function of a novel cancerogenic miRNA, miR-93, in glioma progression by investigating the expression and mechanism of it. PATIENTS AND METHODS: qRT-PCR was conducted to assess the miR-93 expression and the mRNA expression of target gene in glioma tissues and cells. The invasion and migration abilities of the glioma cells were determined by transwell assays. Luciferase reporter assay was performed to confirm the target of miR-93. RESULTS: The results indicated that miR-93 expression in glioma tissues and cells was increased significantly than that in normal brain tissues and cells. Furthermore, miR-93 promoted glioma cell migration and invasion. RBL2 was recognized as a direct target of miR-93 in glioma cells, and overexpression of RBL2 could reverse the stimulative effect of miR-93 in glioma cell. CONCLUSIONS: The above findings suggested that miR-93 together with RBL2 could be diagnostic targets and novel prognostic markers for glioma.


Subject(s)
MicroRNAs/metabolism , Retinoblastoma-Like Protein p130/metabolism , 3' Untranslated Regions , Antagomirs/metabolism , Base Sequence , Brain Neoplasms/genetics , Brain Neoplasms/pathology , Cell Line, Tumor , Cell Movement , Disease Progression , Female , Gene Expression Regulation, Neoplastic , Glioma/genetics , Glioma/pathology , Humans , MicroRNAs/antagonists & inhibitors , MicroRNAs/genetics , Mutagenesis , Retinoblastoma-Like Protein p130/chemistry , Retinoblastoma-Like Protein p130/genetics , Sequence Alignment
3.
Virol J ; 13: 2, 2016 Jan 04.
Article in English | MEDLINE | ID: mdl-26728921

ABSTRACT

BACKGROUND: Retinoblastoma like protein 2 (RBL2) or p130 is a member of the pocket protein family, which is infrequently mutated in human tumours. Its expression is posttranscriptionally regulated and largely G0 restricted. We have previously shown that E6/E7 oncoproteins encoded by human papillomavirus (HPV) type 16, which is a high-risk type for cervical cancer development, must target p130 to promote the host cell to exit from quiescence (G0) state and enter S phase of the cell cycle. P130 is associated with the DREAM (DP, RB-like, E2F and MuvB) complex in G0/G1, which prevents S phase progression by repressing transcription of E2F-regulated genes. E7 proteins could potentially disrupt the p130-DREAM complex through two known mechanisms: direct interaction with p130 or induction of cyclin dependent kinase 2 (CDK2) phosphorylation by interacting with its inhibitor, p21(CIP1). METHODS: In this study we have used p130 mutants deficient in binding the E7 LXCXE domain (p130mE7), unphosphorylatable by CDK2 (p130PM22) or a combination of both (p130PM22/mE7) to investigate these mechanisms used by E7 proteins to disrupt the p130-DREAM complex and promote cell cycle progression. RESULTS: We found that HPV16 E7 binding to p130 through its LXCXE domain was absolutely required to disrupt p130-DREAM to promote S phase of the cell cycle, as HPV16 E7 was unable to suppress p130mE7 but could suppress p130PM22. In contrast, the E7 protein encoded by a cutaneous HPV type that lacks a functional LXCXE domain, HPV 48 E7, was also able to disrupt p130-DREAM to promote cell cycling, but through the alternative mechanism. Thus, HPV48 E7 could suppress a cell cycle block imposed by p130mE7, but was unable to suppress p130PM22. CONCLUSIONS: Overall, these results indicate that suppression of p130 is required for HPV-induced cell cycling, and that different HPV E7 proteins can use alternative mechanisms to achieve this.


Subject(s)
Alphapapillomavirus/classification , Alphapapillomavirus/metabolism , Cell Cycle Checkpoints , Papillomavirus E7 Proteins/metabolism , Retinoblastoma-Like Protein p130/metabolism , Amino Acid Motifs , Amino Acid Sequence , Humans , Mutation , Papillomavirus Infections/metabolism , Papillomavirus Infections/virology , Protein Binding , Protein Interaction Domains and Motifs , Retinoblastoma-Like Protein p130/chemistry , Retinoblastoma-Like Protein p130/genetics
4.
Genes Dev ; 29(9): 961-74, 2015 May 01.
Article in English | MEDLINE | ID: mdl-25917549

ABSTRACT

The DREAM complex represses cell cycle genes during quiescence through scaffolding MuvB proteins with E2F4/5 and the Rb tumor suppressor paralog p107 or p130. Upon cell cycle entry, MuvB dissociates from p107/p130 and recruits B-Myb and FoxM1 for up-regulating mitotic gene expression. To understand the biochemical mechanisms underpinning DREAM function and regulation, we investigated the structural basis for DREAM assembly. We identified a sequence in the MuvB component LIN52 that binds directly to the pocket domains of p107 and p130 when phosphorylated on the DYRK1A kinase site S28. A crystal structure of the LIN52-p107 complex reveals that LIN52 uses a suboptimal LxSxExL sequence together with the phosphate at nearby S28 to bind the LxCxE cleft of the pocket domain with high affinity. The structure explains the specificity for p107/p130 over Rb in the DREAM complex and how the complex is disrupted by viral oncoproteins. Based on insights from the structure, we addressed how DREAM is disassembled upon cell cycle entry. We found that p130 and B-Myb can both bind the core MuvB complex simultaneously but that cyclin-dependent kinase phosphorylation of p130 weakens its association. Together, our data inform a novel target interface for studying MuvB and p130 function and the design of inhibitors that prevent tumor escape in quiescence.


Subject(s)
Cell Cycle/genetics , Gene Expression Regulation , Multiprotein Complexes/chemistry , Multiprotein Complexes/metabolism , Cell Cycle Proteins/metabolism , Crystallization , Humans , Phosphorylation , Protein Binding , Protein Structure, Tertiary , Retinoblastoma-Like Protein p107/chemistry , Retinoblastoma-Like Protein p107/metabolism , Retinoblastoma-Like Protein p130/chemistry , Retinoblastoma-Like Protein p130/metabolism , Sequence Alignment , Trans-Activators/metabolism
5.
Genes Cells ; 18(10): 839-49, 2013 Oct.
Article in English | MEDLINE | ID: mdl-23890198

ABSTRACT

Inorganic arsenic exerts toxic effect on multiple systems including the immune system. We previously showed in a study on mouse thymocytes and B-cell lymphoma A20 cells that arsenite induces cell cycle arrest at G0/G1 by suppressing expression of E2F-target genes. In this study, we furthermore investigated the involvement of retinoblastoma (RB) family proteins in E2F-dependent cell cycle arrest by arsenite. Arsenite exposure of A20 cells was showed to increase the protein level of p130, a RB family member, without changing the mRNA level. Suppression of arsenite-induced p130 by siRNA reduced the G0/G1 phase, indicating that p130 accumulation is responsible for arsenite-induced G0/G1 arrest. The accumulated p130 was shown to increase the p130 complex with E2F4, a transcription-suppressing E2F. Comparison by Western blotting of arsenite-induced p130 and p130 accumulated by a proteasome inhibitor suggested that arsenite-induced p130 is hypophosphorylated and hypoubiquitinated and refractory to proteasome-dependent degradation. We also showed that arsenite increases mRNA and protein of p16(INK4a), an inhibitor of CDK4/6 that phosphorylates p130. Down-regulation of arsenite-induced p16(INK4a) by siRNA suppressed the p130 accumulation. We propose a novel mechanism in which arsenite inhibits phosphorylation/ubiquitin-dependent proteasome degradation of p130 by inducing p16(INK4a) and the accumulated p130 causes cell cycle arrest with E2F4.


Subject(s)
Arsenites/pharmacology , Cell Cycle Checkpoints , Cyclin-Dependent Kinase Inhibitor p16/metabolism , E2F4 Transcription Factor/metabolism , Retinoblastoma-Like Protein p130/metabolism , Ubiquitin/metabolism , Animals , Cell Cycle Checkpoints/genetics , Cell Cycle Proteins/genetics , Cell Cycle Proteins/metabolism , Cell Line, Tumor , Cyclin-Dependent Kinase Inhibitor p16/genetics , Cyclin-Dependent Kinase Inhibitor p21/metabolism , E2F4 Transcription Factor/chemistry , Gene Expression Regulation , Lymphoma, B-Cell/metabolism , Mice , Phosphorylation , RNA, Small Interfering , Retinoblastoma-Like Protein p130/chemistry , Retinoblastoma-Like Protein p130/genetics , Tumor Suppressor Protein p53/metabolism
6.
J Cell Physiol ; 226(4): 1027-34, 2011 Apr.
Article in English | MEDLINE | ID: mdl-20857408

ABSTRACT

One of the mechanisms by which all-trans retinoic acid (ATRA) has been shown to suppress the growth of CAOV3 ovarian carcinoma cells involves an increase in the accumulation of Rb2/p130 protein, a member of the retinoblastoma family of tumor suppressors. This increase in accumulation of RB2/p130 by ATRA results from increased stability of Rb2/p130 protein as a result of an increase in dephosphorylation of the protein by the serine/threonine phosphatase PP2A. We show that upon ATRA treatment, PP2A interacts with the Rb2/p130 C-terminus and specifically dephosphorylates two residues (S1080 and T1097) adjacent to NLS1 and NLS2 of Rb2/p130. Moreover, co-immunoprecipitation studies reveal that Rb2/p130 can form a complex with the nuclear transport proteins, importin α and importin ß, binding to the same dephosphorylated NLS1 and NLS2 sites. Finally, mutation of S1080 and T1097 results in retension of Rb2/p130 in the cytoplasm. Our studies suggest that one mechanism by which ATRA treatment of CAOV3 cells induces G0/G1 arrest involves the recruitment of PP2A to the C-terminus of Rb2/p130, resulting in the dephosphorylation of the S1080 and T1097 adjacent to the NLS and the subsequent interaction of Rb2/p130 with importins leading to transport of the Rb2/p130 to the nucleus where it inhibits cell-cycle progression.


Subject(s)
Cell Nucleus/drug effects , Cell Nucleus/metabolism , Ovarian Neoplasms/enzymology , Ovarian Neoplasms/pathology , Protein Phosphatase 2/metabolism , Retinoblastoma-Like Protein p130/metabolism , Tretinoin/pharmacology , Amino Acid Sequence , Catalytic Domain , Cell Line, Tumor , Female , Humans , Karyopherins/metabolism , Models, Biological , Molecular Sequence Data , Nuclear Localization Signals/chemistry , Peptides/metabolism , Phosphorylation/drug effects , Protein Binding/drug effects , Protein Transport/drug effects , Retinoblastoma-Like Protein p130/chemistry
7.
J Virol ; 82(10): 4862-73, 2008 May.
Article in English | MEDLINE | ID: mdl-18321970

ABSTRACT

The E7 proteins of human papillomaviruses (HPVs) promote S-phase reentry in differentiated keratinocytes of the squamous epithelia to support viral DNA amplification. In this study, we showed that nuclear p130 was present in the differentiated strata of several native squamous epithelia susceptible to HPV infection. In contrast, p130 was below the level of detection in HPV-infected patient specimens. In submerged and organotypic cultures of primary human keratinocytes, the E7 proteins of the high-risk mucosotrophic HPV-18, the benign cutaneous HPV-1, and, to a lesser extent, the low-risk mucosotropic HPV-11 destabilized p130. This E7 activity depends on an intact pocket protein binding domain and a casein kinase II (CKII) phosphorylation motif. Coimmunoprecipitation experiments showed that both E7 domains were important for binding to p130 in extracts of organotypic cultures. Metabolic labeling in vivo demonstrated that E7 proteins were indeed phosphorylated in a CKII motif-dependent manner. Moreover, the efficiencies of the E7 proteins of various HPV types or mutations to induce S-phase reentry in spinous cells correlated with their relative abilities to bind and to destabilize p130. Collectively, these data support the notion that p130 controls the homeostasis of the differentiated keratinocytes and is therefore targeted by E7 for degradation to establish conditions permissive for viral DNA amplification.


Subject(s)
Cell Cycle , Keratinocytes/chemistry , Keratinocytes/virology , Papillomaviridae/physiology , Papillomavirus E7 Proteins/metabolism , Retinoblastoma-Like Protein p130/metabolism , Amino Acid Sequence , Cells, Cultured , Epithelium/chemistry , Epithelium/virology , Human papillomavirus 11/physiology , Human papillomavirus 18/physiology , Humans , Molecular Sequence Data , Papillomavirus E7 Proteins/chemistry , Phosphorylation , Protein Binding , Protein Interaction Mapping , Protein Structure, Tertiary , Retinoblastoma-Like Protein p130/chemistry
8.
Cell Cycle ; 6(21): 2591-3, 2007 Nov 01.
Article in English | MEDLINE | ID: mdl-17726381

ABSTRACT

Retinoblastoma (RB) family proteins pRb, p107 and pRb2/p130 are important cellular factors which play a well-recognized role as tumor and growth suppressors. These proteins are actively involved in the negative control of the cell cycle and their function is modulated via complex homeostatic processes, most of them involving post-translational regulation of their phosphorylation status. Interestingly, the family members p107 and pRb2/p130 share the ability to physically interact and inhibit the kinase activity of the Cdk2/Cyclin A and Cdk2/Cyclin E complexes. Regarding pRb2/p130, its inhibitory effect on Cdk2/Cyclin A activity has been attributed to the "spacer" region. Recently, a 39 aa-long pRb2/p130 spacer-derived peptide (Spa310, aa 641-679) was selected as the sequence responsible for Cdk2/Cyclin A inhibition. Following the identification of this active sequence, here we propose a computer-generated three-dimensional model of the interaction between the Cdk2/Cyclin A complex and the N-terminal nine-amino acid sequence of the Spa310 peptide. We believe this model to be useful for the rational development of peptide or peptidomimetic kinase inhibitors for negative cell cycle modulation in cancer cells.


Subject(s)
Carrier Proteins/metabolism , Cyclin A/metabolism , Cyclin-Dependent Kinase 2/metabolism , DNA, Intergenic/metabolism , Models, Molecular , Retinoblastoma-Like Protein p130/metabolism , Amino Acid Sequence , Binding Sites/genetics , Carrier Proteins/chemistry , Computer Simulation , Cyclin A/chemistry , Cyclin-Dependent Kinase 2/chemistry , DNA, Intergenic/chemistry , Humans , Molecular Sequence Data , Protein Structure, Tertiary/genetics , Retinoblastoma-Binding Protein 2 , Retinoblastoma-Like Protein p130/chemistry
9.
J Cell Physiol ; 213(2): 403-6, 2007 Nov.
Article in English | MEDLINE | ID: mdl-17708530

ABSTRACT

Tumor suppressor pRb2/p130 gene belongs to the retinoblastoma (Rb) gene family, which also includes pRb/p105 and pRb/p107. The members of the Rb gene family have attracted a great deal of interest because of their essential role in regulating cell cycle and, consequently, cell proliferation. This mini review discusses the potential therapeutic applications both of pRb2/p130 and its derived product Spa310 spacer domain in cancer treatment.


Subject(s)
Genes, Tumor Suppressor , Neoplasms , Retinoblastoma-Like Protein p130/chemistry , Retinoblastoma-Like Protein p130/metabolism , Animals , Cell Cycle/physiology , Cell Proliferation , Humans , Multigene Family , Neoplasms/genetics , Neoplasms/metabolism , Neoplasms/therapy , Protein Structure, Tertiary , Retinoblastoma-Like Protein p130/genetics
10.
Oncogene ; 26(13): 1829-39, 2007 Mar 22.
Article in English | MEDLINE | ID: mdl-17043661

ABSTRACT

One strategy in the development of anticancer therapeutics has been to arrest malignant proliferation through inhibition of the enzymatic activity of cyclin-dependent kinases (cdks), which are key regulatory molecules of the cell cycle. Over the past few years, numerous compounds with remarkable cdk inhibitory activity have been studied in cancer therapy, although it is very difficult to point out the best cdk to target. An excellent candidate appears to be cdk2, whose alteration is a pathogenic hallmark of tumorigenesis. The small molecule described in our study showed an inhibitory effect on the kinase activity of cdk2, a significant growth arrest observed in a colony formation assay and a reduction in the size of the tumor in nude mice, thus suggesting its potential role as a promising new type of mechanism-based antitumor drug, also for the treatment of hyperproliferative disorders.


Subject(s)
Cell Cycle , Cyclin-Dependent Kinase 2/antagonists & inhibitors , Neoplasms, Experimental/pathology , Peptides/pharmacology , Retinoblastoma-Like Protein p130/physiology , 3T3 Cells , Amino Acid Sequence , Animals , Cell Line, Tumor , Flow Cytometry , Fluorescent Antibody Technique , Mice , Molecular Sequence Data , Retinoblastoma-Like Protein p130/chemistry
11.
J Cell Physiol ; 206(2): 495-502, 2006 Feb.
Article in English | MEDLINE | ID: mdl-16206244

ABSTRACT

All-trans retinoic acid (ATRA) treatment causes CAOV3 ovarian carcinoma cells to growth arrest in the G0/G1 phase and to elevate the level of Rb2/p130 protein. PP2A, a serine/threonine phosphatase, binds and dephosphorylates Rb2/p130, thereby increasing the half-life of Rb2/p130 in the cell. In order to further characterize the interaction between Rb2/p130 and PP2A upon ATRA treatment, we examined the posttranslational modification of PP2A. ATRA treatment leads to hypophosphorylation of PP2A catalytic subunit (PP2Ac) that correlates with increased PP2A activity. In addition, the N-terminus of PP2Ac binds directly to NLS sequences located in the C-terminus of Rb2/p130. Furthermore, CAOV3 cells transfected with a truncated Rb2/p130 construct consisting of only the wt C-terminus grew more aggressively and were less sensitive to ATRA treatment when compared to parental CAOV3 cells. In contrast, CAOV3 cells transfected with a truncated Rb2/p130 construct consisting of only the C-terminus in which the NLS sites were mutated and which could not interact with PP2A, were as sensitive to ATRA treatment as parental CAOV3 cells. These studies suggest that ATRA treatment suppresses the growth of CAOV3 cells via a novel posttranscriptional mechanism involving PP2A.


Subject(s)
Ovarian Neoplasms/metabolism , Phosphoprotein Phosphatases/metabolism , Retinoblastoma-Like Protein p130/pharmacology , Retinoblastoma-Like Protein p130/physiology , Tretinoin/pharmacology , Catalytic Domain , Cell Line, Tumor , Cell Proliferation/drug effects , Female , Humans , Ovarian Neoplasms/pathology , Phosphoprotein Phosphatases/chemistry , Phosphorylation , Protein Processing, Post-Translational , Retinoblastoma-Like Protein p130/chemistry , Retinoblastoma-Like Protein p130/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...