Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 31
Filter
1.
Protein Expr Purif ; 149: 31-36, 2018 09.
Article in English | MEDLINE | ID: mdl-29674116

ABSTRACT

Proteins and polypeptides represent nature's most complex and versatile polymer. They provide complicated shapes, diverse chemical functionalities, and tightly regulated and controlled sizes. Several disease states are related to the misfolding or overproduction of polypeptides and yet polypeptides are present in several therapeutic molecules. In addition to biological roles; short chain polypeptides have been shown to interact with and drive the bio-inspired synthesis or modification of inorganic materials. This paper outlines the development of a versatile cloning vector which allows for the expression of a short polypeptide by controlling the incorporation of a desired DNA coding insert. As a demonstration of the efficacy of the expression system, a solid binding polypeptide identified from M13 phage display was expressed and purified. The solid binding polypeptide was expressed as a soluble 6xHis-SUMO tagged construct. Expression was performed in E. coli using auto-induction followed by Ni-NTA affinity chromatography and ULP1 protease cleavage. Methodology demonstrates the production of greater than 8 mg of purified polypeptide per liter of E. coli culture. Isotopic labeling of the peptide is also demonstrated. The versatility of the designed cloning vector, use of the 6xHis-SUMO solubility partner, bacterial expression in auto-inducing media and the purification methodology make this expressionun vector a readily scalable and user-friendly system for the creation of desired peptide domains.


Subject(s)
Genetic Vectors/genetics , Peptides/genetics , Bacteriophage M13 , Cell Surface Display Techniques , DNA/genetics , Escherichia coli , Gene Expression , Recombinant Fusion Proteins/biosynthesis , Recombinant Fusion Proteins/genetics , Small Ubiquitin-Related Modifier Proteins/biosynthesis , Small Ubiquitin-Related Modifier Proteins/genetics , Solubility
2.
J Biol Chem ; 292(25): 10574-10585, 2017 06 23.
Article in English | MEDLINE | ID: mdl-28465351

ABSTRACT

Hepatocyte nuclear factor 4α (HNF4α) controls the expression of liver-specific protein-coding genes. However, some microRNAs are also modulated by HNF4α, and it is not known whether they are direct targets of HNF4α and whether they influence hepatic function. In this study, we found that HNF4α regulates microRNAs, indicated by marked down-regulation of miR-194 and miR-192 (miR-194/192) in liver-specific Hnf4a-null (Hnf4aΔH) mice. Transactivation of the shared miR-194/192 promoter was dependent on HNF4α expression, indicating that miR-194/192 is a target gene of HNF4α. Screening of potential mRNAs targeted by miR-194/192 revealed that expression of genes involved in glucose metabolism (glycogenin 1 (Gyg1)), cell adhesion and migration (activated leukocyte cell adhesion molecule (Alcam)), tumorigenesis and tumor progression (Rap2b and epiregulin (Ereg)), protein SUMOylation (Sumo2), epigenetic regulation (Setd5 and Cullin 4B (Cln4b)), and the epithelial-mesenchymal transition (moesin (Msn)) was up-regulated in Hnf4aΔH mice. Moreover, we also found that miR-194/192 binds the 3'-UTR of these mRNAs. siRNA knockdown of HNF4α suppressed miR-194/192 expression in human hepatocellular carcinoma (HCC) cells and resulted in up-regulation of their mRNA targets. Inhibition and overexpression experiments with miR-194/192 revealed that Gyg1, Setd5, Sumo2, Cln4b, and Rap2b are miR-194 targets, whereas Ereg, Alcam, and Msn are miR-192 targets. These findings reveal a novel HNF4α network controlled by miR-194/192 that may play a critical role in maintaining the hepatocyte-differentiated state by inhibiting expression of genes involved in dedifferentiation and tumorigenesis. These insights may contribute to the development of diagnostic markers for early HCC detection, and targeting of the miR-194/192 pathway could be useful for managing HCC.


Subject(s)
Gene Expression Regulation/physiology , Hepatocyte Nuclear Factor 4/metabolism , Hepatocytes/metabolism , MicroRNAs/metabolism , Signal Transduction/physiology , 3' Untranslated Regions/physiology , Activated-Leukocyte Cell Adhesion Molecule/biosynthesis , Activated-Leukocyte Cell Adhesion Molecule/genetics , Animals , Cell Transformation, Neoplastic/genetics , Cell Transformation, Neoplastic/metabolism , Epiregulin/biosynthesis , Epiregulin/genetics , Glucosyltransferases/biosynthesis , Glucosyltransferases/genetics , Glycoproteins/biosynthesis , Glycoproteins/genetics , Hepatocyte Nuclear Factor 4/genetics , Liver Neoplasms/genetics , Liver Neoplasms/metabolism , Mice , Mice, Mutant Strains , MicroRNAs/genetics , Microfilament Proteins/biosynthesis , Microfilament Proteins/genetics , Small Ubiquitin-Related Modifier Proteins/biosynthesis , Small Ubiquitin-Related Modifier Proteins/genetics
3.
Nat Methods ; 11(11): 1161-9, 2014 Nov.
Article in English | MEDLINE | ID: mdl-25262208

ABSTRACT

Cancer stem cells (CSCs) are thought to drive tumor growth, metastasis and chemoresistance. Although surface markers such as CD133 and CD44 have been successfully used to isolate CSCs, their expression is not exclusively linked to the CSC phenotype and is prone to environmental alteration. We identified cells with an autofluorescent subcellular compartment that exclusively showed CSC features across different human tumor types. Primary tumor-derived autofluorescent cells did not overlap with side-population (SP) cells, were enriched in sphere culture and during chemotherapy, strongly expressed pluripotency-associated genes, were highly metastatic and showed long-term in vivo tumorigenicity, even at the single-cell level. Autofluorescence was due to riboflavin accumulation in membrane-bounded cytoplasmic structures bearing ATP-dependent ABCG2 transporters. In summary, we identified and characterized an intrinsic autofluorescent phenotype in CSCs of diverse epithelial cancers and used this marker to isolate and characterize these cells.


Subject(s)
Biomarkers, Tumor/metabolism , Cell Separation/methods , Neoplastic Stem Cells/metabolism , Neoplastic Stem Cells/pathology , Optical Imaging/methods , Riboflavin/metabolism , ATP Binding Cassette Transporter, Subfamily G, Member 2 , ATP-Binding Cassette Transporters/metabolism , Animals , Autophagy , Autophagy-Related Protein 12 , Carcinoma, Hepatocellular/pathology , Carcinoma, Non-Small-Cell Lung/pathology , Carcinoma, Pancreatic Ductal/pathology , Colorectal Neoplasms/pathology , Female , Humans , Liver Neoplasms/pathology , Lung Neoplasms/pathology , Mice , Mice, Nude , Microtubule-Associated Proteins/biosynthesis , Neoplasm Proteins/metabolism , Pancreatic Neoplasms/pathology , Small Ubiquitin-Related Modifier Proteins/biosynthesis , Tumor Cells, Cultured
4.
Sheng Wu Gong Cheng Xue Bao ; 30(4): 658-68, 2014 Apr.
Article in Chinese | MEDLINE | ID: mdl-25195255

ABSTRACT

Fibroblast growth factor -21 (FGF-21) is a recently discovered metabolic regulation factor, regulating glucose and lipid metabolism and increasing insulin sensitivity. FGF-21 is expected to be a potential anti-diabetic drug. Expression of FGF-21 as inclusion bodies has advantages for high yield and purity, but the bioactivity of the protein is almost totally lost after denature and renature. That is why FGF-21 is currently expressed in soluble form. As a result, the yield is considerably low. In this study, we used SUMO vector to express SUMO-human FGF-21 (SUMO-hFGF-21) in form of inclusion body. We optimized the culture conditions to increase the yield of the bioactive human fibroblast growth factor-21. We applied the hollow fiber membrane filtration column to enrich the bacteria, wash, denature and renature inclusion bodies. After affinity and gel filtration chromatography, we examined the hypoglycemic activity of FGF-21 by the glucose uptake assay in HepG2 cells. We also detected the blood glucose concentration of type 2 diabetic db/db model mice after short or long-term treatment. The results show that the yield of ihFGF-21 was 4 times higher than that of shFGF-21. The yield was 20 mg/L for ihFGF-21 vs. 6 mg/L for shFGF-21. The purity of ihFGF-21 was above 95%, while shFGF-21 was 90%. Compared with the traditional method of extracting inclusion bodies, the production cycle was about three times shortened by application of hollow fiber membrane filtration column technology, but the bioactivity did not significantly differ. This method provides an efficient and cost-effective strategy to the pilot and industrial production of hFGF-21.


Subject(s)
Fibroblast Growth Factors/biosynthesis , Hypoglycemic Agents/isolation & purification , Inclusion Bodies/metabolism , Animals , Bacteria/metabolism , Diabetes Mellitus, Experimental/drug therapy , Disease Models, Animal , Genetic Vectors , Glucose/metabolism , Hep G2 Cells , Humans , Mice , Recombinant Fusion Proteins/biosynthesis , Small Ubiquitin-Related Modifier Proteins/biosynthesis
5.
Int J Mol Med ; 34(2): 545-52, 2014 Aug.
Article in English | MEDLINE | ID: mdl-24899049

ABSTRACT

We have previously reported that Tougu Xiaotong capsule (TXC) inhibits tidemark replication and cartilage degradation by regulating chondrocyte autophagy in vivo. Autophagy, a cell protective mechanism for maintaining cellular homeostasis, has been shown to be a constitutively active and protective process for chondrocyte survival. However, it remains unclear whether TXC promotes chondrocyte autophagy by regulating the autophagy-related (Atg)12/microtubule-associated protein 1 light chain 3 (LC3) conjugation systems. Thus, in the present study, we investigated the effects of TXC on primary chondrocytes treated with cobalt chloride (CoCl2). We found that CoCl2 induced a decrease in chondrocyte viability and the autophagosome formation of chondrocytes, indicating that CoCl2 induced autophagic death in a dose- and time-dependent manner. To determine the effects of TXC on CoCl2-exposed chondrocytes, we assessed cell viability by MTT assay. Our results revealed that TXC enhanced the viability of CoCl2-exposed chondrocytes. To gain insight into the mechanisms responsible for the enhancing effects of TXC on CoCl2-exposed chondrocytes, the expression of Atg genes was assessed in chondrocytes exposed to CoCl2 and treated with or without TXC. The results revealed that the expression of beclin 1, Atg3, Atg5, Atg7, Atg10, Atg12 and LC3 II/LC3 I in the chondrocytes treated with TXC increased, compared to that in the untreated chondrocytes. In addition, ultrastructural analysis indicated that treated chondrocytes contained more autophagosomes than the untreated cells, suggesting that TXC increased the formation of autophagosomes in the chondrocytes to clear the CoCl2-induced autophagic death. Therefore, these data suggest that TXC is a potential therapeutic agent for the reduction of cartilage degradation that occurs in osteoarthritis.


Subject(s)
Autophagy/drug effects , Drugs, Chinese Herbal/administration & dosage , Microtubule-Associated Proteins/biosynthesis , Osteoarthritis/drug therapy , Small Ubiquitin-Related Modifier Proteins/biosynthesis , Apoptosis/drug effects , Autophagy-Related Protein 12 , Cartilage/drug effects , Cartilage/metabolism , Cell Line , Chondrocytes/drug effects , Chondrocytes/metabolism , Cobalt/toxicity , Humans , Microtubule-Associated Proteins/metabolism , Osteoarthritis/metabolism , Osteoarthritis/pathology , Phagosomes/drug effects , Small Ubiquitin-Related Modifier Proteins/genetics
6.
J Immunol Methods ; 403(1-2): 37-51, 2014 Jan 31.
Article in English | MEDLINE | ID: mdl-24291344

ABSTRACT

This is a first report of recombinant production of human prepro-Urocortin 2 in Escherichia coli by N-terminal fusion with a triple His6-SUMO-eXact tag and its subsequent use as an antigen for the production and screening of very high affinity monoclonal antibodies. The rationale for this combinatorial construct is that the His tag allows first step protein purification of insoluble and soluble proteins, the SUMO tag enhances protein expression level and solubility, while the eXact tag facilitates anion-triggered on-column cleavage of the triple tag to recover pure native proteins in a simple two-step protein purification procedure. Compared with an eXact fusion alone, the presence of the SUMO moiety enhanced overall expression levels by 4 to 10 fold but not the solubility of the highly basic prepro-Urocortin 2. Insoluble SUMO-eXact-preproUCN2 was purified in milligram quantities by denaturing IMAC and solubilized in native phosphate buffer by on-column refolding or step-wise dialysis. Only a small fraction of this solubilized protein was able to bind onto the eXact™ affinity column and cleaved by NaF treatment. To test whether binding and cleavage failure was due to improperly refolded SUMO-eXact-preproUCN2 or to the presence of N- and C-terminal sequences flanking the eXact moiety, we created a SUMO-eXact-thioredoxin construct which was overexpressed mainly in the soluble form. This protein bound to and was cleaved efficiently on the eXact™ column to yield native thioredoxin. Solubilized SUMO-eXact-preproUCN2 was used successfully to generate two high affinity mouse monoclonal antibodies (KD~10⁻¹° and 10⁻¹¹ M) specific to the pro-region of Urocortin 2.


Subject(s)
Antibodies, Monoclonal/biosynthesis , Cloning, Molecular/methods , Corticotropin-Releasing Hormone/biosynthesis , Escherichia coli/metabolism , Histidine/biosynthesis , Oligopeptides/biosynthesis , Protein Precursors/biosynthesis , Saccharomyces cerevisiae Proteins/biosynthesis , Small Ubiquitin-Related Modifier Proteins/biosynthesis , Urocortins/biosynthesis , Amino Acid Sequence , Animals , Antibody Specificity , Chromatography, Affinity , Corticotropin-Releasing Hormone/administration & dosage , Corticotropin-Releasing Hormone/genetics , Corticotropin-Releasing Hormone/immunology , Escherichia coli/genetics , Histidine/genetics , Humans , Immunization , Injections , Mice , Mice, Inbred BALB C , Molecular Sequence Data , Mutation , Oligopeptides/genetics , Protein Binding , Protein Denaturation , Protein Precursors/administration & dosage , Protein Precursors/genetics , Protein Precursors/immunology , Recombinant Fusion Proteins/biosynthesis , Recombinant Fusion Proteins/immunology , Saccharomyces cerevisiae Proteins/genetics , Small Ubiquitin-Related Modifier Proteins/genetics , Solubility , Subtilisin/genetics , Subtilisin/metabolism , Urocortins/administration & dosage , Urocortins/genetics , Urocortins/immunology
7.
Microb Cell Fact ; 13: 178, 2014 Dec 31.
Article in English | MEDLINE | ID: mdl-25551787

ABSTRACT

BACKGROUND: Amphiphilic peptides are important building blocks to generate nanostructured biomaterials for drug delivery and tissue engineering applications. We have shown that the self-assembling peptide SA2 (Ac-AAVVLLLWEE) can be recombinantly produced in E. coli when fused to the small ubiquitin-like modifier (SUMO) protein. Although this system yielded peptides of high purity with no residual amino acids after cleavage of the SUMO fusion protein, the yield after purification was generally low (~1 mg/L bacterial culture) as compared to other peptides and proteins produced with the same method and under the same conditions. RESULTS: The aim of this study is to understand the underlying mechanisms causing the low yield of this recombinant peptide in E. coli and to optimize both production and purification of recombinant SA2 peptides. It was demonstrated that by simply changing the medium to a well-balanced auto-induction medium the yield of recombinant production was augmented (~4 fold). Moreover, it was demonstrated that self-assembly of SUMO-SA2 fusion proteins caused the low peptide yields after purification. By replacing the second IMAC purification step with a selective precipitation step, peptide yields could be increased approx. 3 fold. With these optimizations in place the overall yield of purified SA2 peptide increased with 12-fold. CONCLUSION: Premature self-assembly of the SUMO-SA2 fusion construct interfered with proper purification of the SA2 peptide, resulting in low yields of purified peptide and this could be prevented by changing the mode of purification. These findings are important when setting up purification schemes for other self-assembling peptides with the use of a SUMO fusion construct.


Subject(s)
Escherichia coli , Peptides , Small Ubiquitin-Related Modifier Proteins , Humans , Peptides/chemistry , Peptides/genetics , Peptides/isolation & purification , Peptides/metabolism , Recombinant Proteins/biosynthesis , Recombinant Proteins/chemistry , Recombinant Proteins/genetics , Recombinant Proteins/isolation & purification , Small Ubiquitin-Related Modifier Proteins/biosynthesis , Small Ubiquitin-Related Modifier Proteins/chemistry , Small Ubiquitin-Related Modifier Proteins/genetics , Small Ubiquitin-Related Modifier Proteins/isolation & purification
8.
Oncol Rep ; 31(2): 649-56, 2014 Feb.
Article in English | MEDLINE | ID: mdl-24297300

ABSTRACT

AZD8055 is a potent inhibitor of mTORC1 and mTOR2 and shows inhibitory effects in several types of cancer cells in vitro and in vivo. However, the effect of AZD8055 on hepatocellular carcinoma (HCC) cells has not been studied. We report that AZD8055 inhibits cell proliferation and colony formation of Hep3B and Huh7 cells but does not cause PARP cleavage, or caspase activation, suggesting that classical apoptosis is not its main mechanism of cell death. By contrast, AZD8055-induced cell death was associated with several characteristics of autophagy, including an increase in acidic vesicular organelle content, conversion of cytosolic LC3-I to membrane-bound LC3-II and elevation of the levels of Atg-5/12, BECN1 and LC3-II. Inhibition of autophagy by 3-methyladenine (3-MA) partially inhibited AZD8055-induced cell death. Furthermore, AZD8055 caused the activation of AMPK and co-treatment with the AMPK inhibitor dorsomorphin also caused a partial but significant reduction of AZD8055-induced cell death. In conclusion, AZD8055-induced HCC cell death is associated with induction of autophagy and activation of AMPK.


Subject(s)
AMP-Activated Protein Kinases/metabolism , Autophagy/drug effects , Carcinoma, Hepatocellular/drug therapy , Liver Neoplasms/drug therapy , Morpholines/pharmacology , Adenine/analogs & derivatives , Adenine/pharmacology , Apoptosis/drug effects , Apoptosis Regulatory Proteins/biosynthesis , Autophagy-Related Protein 12 , Autophagy-Related Protein 5 , Beclin-1 , Caspases/metabolism , Cell Line, Tumor , Cell Proliferation/drug effects , Cell Survival/drug effects , Enzyme Activation/drug effects , Humans , Mechanistic Target of Rapamycin Complex 1 , Mechanistic Target of Rapamycin Complex 2 , Membrane Proteins/biosynthesis , Microtubule-Associated Proteins/biosynthesis , Microtubule-Associated Proteins/metabolism , Multiprotein Complexes/antagonists & inhibitors , Phosphorylation/drug effects , Poly (ADP-Ribose) Polymerase-1 , Poly(ADP-ribose) Polymerases/metabolism , Signal Transduction/drug effects , Small Ubiquitin-Related Modifier Proteins/biosynthesis , TOR Serine-Threonine Kinases/antagonists & inhibitors
10.
Int J Oncol ; 42(3): 993-1000, 2013 Mar.
Article in English | MEDLINE | ID: mdl-23354080

ABSTRACT

Baicalin is one of the major compounds in the traditional Chinese medicinal herb from Scutellaria baicalensis Georgi. We investigated the molecular mechanisms of cell autophagy induced by baicalin in human bladder cancer T24 cells. Baicalin inhibited cell survival as shown by MTT assay and increased cell death by trypan blue exclusion assay in a concentration-dependent manner. Baicalin did not induce apoptotic cell death in T24 cells by TUNEL and caspase-3 activity assay. Baicalin induced the acidic vesicular organelle cell autophagy marker, manifested by acridine orange (AO) and monodansylcadaverine (MDC) staining and cleavage of microtubule-associated protein 1 light chain 3 (LC3). The protein expression levels of the Atg 5, Atg 7, Atg 12, Beclin-1 and LC3-II were upregulated in T24 cells after baicalin treatment. Inhibition of autophagy by 3-methyl-adenine (an inhibitor of class III phosphatidylinositol-3 kinase; 3-MA) reduced the cleavage of LC3 in T24 cells after baicalin treatment. Furthermore, protein expression levels of phospho-AKT (Ser473) and enzyme activity of AKT were downregulated in T24 cells after baicalin treatment. In conclusion, baicalin triggered cell autophagy through the AKT signaling pathway in T24 cells.


Subject(s)
Autophagy/drug effects , Enzyme Inhibitors/pharmacology , Flavonoids/pharmacology , Proto-Oncogene Proteins c-akt/metabolism , Urinary Bladder Neoplasms/drug therapy , Acridine Orange , Adenine/analogs & derivatives , Adenine/pharmacology , Apoptosis Regulatory Proteins/biosynthesis , Autophagy-Related Protein 12 , Autophagy-Related Protein 5 , Autophagy-Related Protein 7 , Beclin-1 , Cadaverine/analogs & derivatives , Caspase 3 , Cell Line, Tumor , Cell Survival/drug effects , Humans , Medicine, Chinese Traditional , Membrane Proteins/biosynthesis , Microtubule-Associated Proteins/biosynthesis , Microtubule-Associated Proteins/metabolism , Phosphoinositide-3 Kinase Inhibitors , Proto-Oncogene Proteins c-akt/biosynthesis , Signal Transduction/drug effects , Small Ubiquitin-Related Modifier Proteins/biosynthesis , Transglutaminases/antagonists & inhibitors , Ubiquitin-Activating Enzymes/biosynthesis
11.
Diabetes Metab Res Rev ; 28(2): 196-202, 2012 Feb.
Article in English | MEDLINE | ID: mdl-22423385

ABSTRACT

BACKGROUND: Type 1 diabetes is a multi-factorial autoimmune disease that results from the destruction of insulin-producing ß cells of the pancreas; both genetic and environmental factors are thought to contribute to its development. Recently, a novel gene encoding small ubiquitin-like modifier protein 4 (SUMO4) was cloned and a single nucleotide substitution (M55V) was found to be strongly associated with type 1 diabetes. SUMO4 was shown to interact with IκBα and inhibit NFκB transcriptional activity. The M55V substitution of SUMO4 may affect its ability to modify IκBα by sumoylation, and so lead to activation of NFκB and transcription of genes implicated in the development of type 1 diabetes. However, the effects of sumoylation on immune cells are poorly understood. METHODS: Human SUMO1, 2, 3, 4 and mouse SUMO2 (mSUMO2) were cloned and overexpressed in T and B cells using retroviral transduction. We then investigated whether SUMO overexpression affected their functions in vitro. To study the function of mSUMO2 in vivo, we made transgenic mice overexpressing mSUMO2 in T cells and pancreatic ß cells and compared them with transgenic mice expressing a super-repressor of NFκB (a dominant negative form of NFκB, IκBαΔN) in T cells. Diabetes was induced in the two groups of mice by i.p. injection of streptozotocin. RESULTS: Human SUMO1, 2, 3, 4 and mSUMO2 were all found to negatively regulate the transcriptional activity of T and B cells. Supporting this idea, mSUMO2 overexpression in T cells suppressed the production of both Th1 and Th2 cytokines unlike T cells from the IκBαΔN mice. However, transgenic mice overexpressing mSUMO2 had the same susceptibility to diabetes as wild type whereas the mice overexpressing IκBαΔN Tg were completely protected against diabetes. CONCLUSION: These results indicate that at least in T cells, whereas NFκB has pro-apoptotic activity, mSUMO2 plays a more complex role in the development of autoimmune diabetes. The relative influence of NFκB and sumoylation on the development of autoimmune diabetes in vivo may vary depending on the developmental stage and cell type.


Subject(s)
Diabetes Mellitus, Type 1/genetics , Small Ubiquitin-Related Modifier Proteins/physiology , Animals , Dendritic Cells/drug effects , Dendritic Cells/metabolism , Humans , I-kappa B Proteins/metabolism , Interleukin-12/biosynthesis , Mice , Mice, Transgenic , NF-KappaB Inhibitor alpha , NF-kappa B/metabolism , SUMO-1 Protein/physiology , Small Ubiquitin-Related Modifier Proteins/biosynthesis , Small Ubiquitin-Related Modifier Proteins/genetics , Sumoylation , T-Lymphocytes/drug effects , T-Lymphocytes/metabolism
12.
Sheng Wu Gong Cheng Xue Bao ; 27(6): 952-62, 2011 Jun.
Article in Chinese | MEDLINE | ID: mdl-22034825

ABSTRACT

Nowadays, SUMO fusion system is important for recombinant protein production in Escherichia coli, yet a few aspects remain to be improved, including the efficacy for vector construction and protein solubility. In this study, we found the SUMO gene Smt3 (Sm) of Saccharomyces cerevisiae conferred an unexpected activity of constitutive prokaryotic promoter during its PCR cloning, and the gene coding regions of SUMOs in most species had a sigma70-dependent prokaryotic promoter embedded, through the prediction via the BPROM program developed by Softberry. By combining the characters of Sm promoter activity and the Stu I site (added at the 3'-terminal of Sm), and introducing a His-tag and a hyper-acidic solubility-enhancing tag, we further constructed a set of versatile vectors for gene cloning and expression on the basis of Sm'-LacZa fusion gene. Experimentally started from these vectors, several target genes were subcloned and expressed through blue-white screening and SDS-PAGE analysis. The results manifest a few of expectable advantages such as rapid vector construction, highly soluble protein expression and feasible co-expression of correlated proteins. Conclusively, our optimized SUMO fusion technology herein could confer a large potential in E. coli protein expression system, and the simultaneously established co-expression vector systems could also be very useful in studying the protein-protein interactions in vivo.


Subject(s)
Genetic Vectors/genetics , Promoter Regions, Genetic/genetics , Recombinant Fusion Proteins/biosynthesis , Saccharomyces cerevisiae Proteins/genetics , Small Ubiquitin-Related Modifier Proteins/genetics , Sumoylation , Amino Acid Sequence , Base Sequence , Escherichia coli/genetics , Escherichia coli/metabolism , Lac Operon/genetics , Molecular Sequence Data , Recombinant Fusion Proteins/genetics , Saccharomyces cerevisiae Proteins/biosynthesis , Small Ubiquitin-Related Modifier Proteins/biosynthesis
13.
Diabetes Metab Res Rev ; 27(8): 854-61, 2011 Nov.
Article in English | MEDLINE | ID: mdl-22069273

ABSTRACT

BACKGROUND: Type 1 diabetes is a multi-factorial autoimmune disease that results from the destruction of insulin-producing ß cells of the pancreas; both genetic and environmental factors are thought to contribute to its development. Recently, a novel gene encoding small ubiquitin-like modifier protein 4 (SUMO4) was cloned and a single nucleotide substitution (M55V) was found to be strongly associated with type 1 diabetes. SUMO4 was shown to interact with IκBα and inhibit NFκB transcriptional activity. The M55V substitution of SUMO4 may affect its ability to modify IκBα by sumoylation, and so lead to activation of NFκB and transcription of genes implicated in the development of type 1 diabetes. However, the effects of sumoylation on immune cells are poorly understood. METHODS: Human SUMO1, 2, 3, 4 and mouse SUMO2 (mSUMO2) were cloned and overexpressed in dendritic, T and B cells using retroviral transduction. We then investigated whether SUMO overexpression affected their functions in vitro. To study the function of mSUMO2 in vivo, we made transgenic mice overexpressing mSUMO2 in T cells and pancreatic ß cells and compared them with transgenic mice expressing a super-repressor of NFκB (a dominant negative form of NFκB, IκBαΔN) in T cells. Diabetes was induced in the two groups of mice by i.p. injection of streptozotocin. RESULTS: Human SUMO1, 2, 3, 4 and mSUMO2 were all found to negatively regulate the transcriptional activity of T, B and dendritic cells. Although mSUMO2 overexpression in dendritic cells did not alter the expression of major histocompatibility complex class II proteins or B7, IL-1, IL-6 and IL-7, IL-12 expression decreased, switching Th1-directed immune responses into Th2 responses. Unlike T cells from the IκBαΔN mice, mSUMO2 overexpression in T cells suppressed the production of both Th1 and Th2 cytokines. Whereas the mice overexpressing IκBαΔN were completely protected against diabetes, those expressing mSUMO2 had the same susceptibility to diabetes as wild type. CONCLUSION: These results indicate that at least in T cells, whereas NFκB has pro-apoptotic activity, mSUMO2 plays a more complex role in the development of autoimmune diabetes. The relative influence of NFκB and sumoylation on the development of autoimmune diabetes in vivo may vary depending on the developmental stage and cell type.


Subject(s)
Diabetes Mellitus, Type 1/genetics , Small Ubiquitin-Related Modifier Proteins/physiology , Animals , Dendritic Cells/drug effects , Dendritic Cells/metabolism , Humans , I-kappa B Proteins/metabolism , Interleukin-12/biosynthesis , Mice , Mice, Transgenic , NF-KappaB Inhibitor alpha , NF-kappa B/metabolism , SUMO-1 Protein , Small Ubiquitin-Related Modifier Proteins/biosynthesis , Small Ubiquitin-Related Modifier Proteins/genetics , Sumoylation , T-Lymphocytes/drug effects , T-Lymphocytes/metabolism
14.
Mol Cell Biol ; 31(11): 2299-310, 2011 Jun.
Article in English | MEDLINE | ID: mdl-21444718

ABSTRACT

Global sumoylation, SUMO chain formation, and genome stabilization are all outputs generated by a limited repertoire of enzymes. Mechanisms driving selectivity for each of these processes are largely uncharacterized. Here, through crystallographic analyses we show that the SUMO E2 Ubc9 forms a noncovalent complex with a SUMO-like domain of Rad60 (SLD2). Ubc9:SLD2 and Ubc9:SUMO noncovalent complexes are structurally analogous, suggesting that differential recruitment of Ubc9 by SUMO or Rad60 provides a novel means for such selectivity. Indeed, deconvoluting Ubc9 function by disrupting either the Ubc9:SLD2 or Ubc9:SUMO noncovalent complex reveals distinct roles in facilitating sumoylation. Ubc9:SLD2 acts in the Nse2 SUMO E3 ligase-dependent pathway for DNA repair, whereas Ubc9:SUMO instead promotes global sumoylation and chain formation, via the Pli1 E3 SUMO ligase. Moreover, this Pli1-dependent SUMO chain formation causes the genome instability phenotypes of SUMO-targeted ubiquitin ligase (STUbL) mutants. Overall, we determine that, unexpectedly, Ubc9 noncovalent partner choice dictates the role of sumoylation in distinct cellular pathways.


Subject(s)
Chromosomal Proteins, Non-Histone/metabolism , DNA Repair , Schizosaccharomyces pombe Proteins/metabolism , Small Ubiquitin-Related Modifier Proteins/metabolism , Ubiquitin-Conjugating Enzymes/metabolism , Blotting, Western , Carrier Proteins/chemistry , Chromosomal Proteins, Non-Histone/chemistry , Crystallography, X-Ray , Humans , Mass Spectrometry , Models, Molecular , Mutation , Protein Structure, Quaternary , Schizosaccharomyces , Schizosaccharomyces pombe Proteins/chemistry , Small Ubiquitin-Related Modifier Proteins/biosynthesis , Sumoylation , Ubiquitin-Conjugating Enzymes/chemistry
15.
Mol Biol Cell ; 22(2): 165-78, 2011 Jan 15.
Article in English | MEDLINE | ID: mdl-21119005

ABSTRACT

The protumorigenic functions for autophagy are largely attributed to its ability to promote cancer cell survival in response to diverse stresses. Here we demonstrate an unexpected connection between autophagy and glucose metabolism that facilitates adhesion-independent transformation driven by a strong oncogenic insult-mutationally active Ras. In cells ectopically expressing oncogenic H-Ras as well as human cancer cell lines harboring endogenous K-Ras mutations, autophagy is induced following extracellular matrix detachment. Inhibiting autophagy due to the genetic deletion or RNA interference-mediated depletion of multiple autophagy regulators attenuates Ras-mediated adhesion-independent transformation and proliferation as well as reduces glycolytic capacity. Furthermore, in contrast to autophagy-competent cells, both proliferation and transformation in autophagy-deficient cells expressing oncogenic Ras are insensitive to reductions in glucose availability. Overall, increased glycolysis in autophagy-competent cells facilitates Ras-mediated adhesion-independent transformation, suggesting a unique mechanism by which autophagy may promote Ras-driven tumor growth in specific metabolic contexts.


Subject(s)
Autophagy , Cell Transformation, Neoplastic/metabolism , ras Proteins/physiology , Animals , Anoikis , Autophagy-Related Protein 12 , Autophagy-Related Protein 7 , Cell Adhesion , Cell Line, Transformed , Cell Proliferation , Cell Transformation, Neoplastic/pathology , Female , Glycolysis , Humans , Mice , Proto-Oncogene Proteins c-bcl-2/biosynthesis , RNA Interference , Small Ubiquitin-Related Modifier Proteins/biosynthesis , Small Ubiquitin-Related Modifier Proteins/genetics , Tumor Cells, Cultured , Ubiquitin-Activating Enzymes/biosynthesis , Ubiquitin-Activating Enzymes/genetics , ras Proteins/biosynthesis , ras Proteins/genetics
16.
Eur J Immunol ; 40(5): 1461-72, 2010 May.
Article in English | MEDLINE | ID: mdl-20162553

ABSTRACT

The induction of the autophagy machinery, a process for the catabolism of cytosolic proteins and organelles, constitutes a crucial mechanism in innate immunity. However, the involvement of autophagy in human neutrophils and the possible inducers of this process have not been completely elucidated. In this study, the induction of autophagy was examined in human neutrophils treated with various activators and detected by the formation of acidified autophagosomes through monodansylcadaverine staining and via LC-3B conversion screened by immunoblotting and immunofluorescence confocal microscopy. In addition, the expression of the ATG genes was assessed by real-time RT-PCR. We provide evidence that autophagy is implicated in human neutrophils in both a phagocytosis-independent (rapamycin, TLR agonists, PMA) and phagocytosis (Escherichia coli)-dependent initiation manner. ROS activation is a positive mechanism for autophagy induction in the case of PMA, TLR activation and phagocytosis. Furthermore, LC3B gene expression was uniformly upregulated, indicating a transcriptional level of regulation for the autophagic machinery. This study provides a stepping stone toward further investigation of autophagy in neutrophil-driven inflammatory disorders.


Subject(s)
Autophagy/physiology , Neutrophils/physiology , Adenine/analogs & derivatives , Adenine/pharmacology , Autophagy/drug effects , Autophagy/genetics , Cadaverine/analogs & derivatives , Cadaverine/analysis , Chromones/pharmacology , Coloring Agents/analysis , Escherichia coli , Guanosine/analogs & derivatives , Guanosine/pharmacology , Humans , Hydrogen-Ion Concentration , Inflammation/immunology , Microscopy, Confocal , Morpholines/pharmacology , Neutrophils/cytology , Neutrophils/drug effects , Phagosomes/physiology , Phagosomes/ultrastructure , Poly I-C/pharmacology , Reactive Oxygen Species/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Signal Transduction/drug effects , Sirolimus/pharmacology , Small Ubiquitin-Related Modifier Proteins/biosynthesis , Small Ubiquitin-Related Modifier Proteins/genetics , Tetradecanoylphorbol Acetate/pharmacology , Toll-Like Receptors/drug effects , Toll-Like Receptors/physiology , Transcription, Genetic , Vacuoles/physiology
17.
Blood ; 115(14): 2827-34, 2010 Apr 08.
Article in English | MEDLINE | ID: mdl-19965618

ABSTRACT

Multiple myeloma (MM) is a plasma cell neoplasm that proceeds through a premalignant state of monoclonal gammopathy of unknown significance; however, the molecular events responsible for myelomagenesis remain uncharacterized. To identify cellular pathways deregulated in MM, we addressed that sumoylation is homologous to ubiquitination and results in the attachment of the ubiquitin-like protein Sumo onto target proteins. Sumoylation was markedly enhanced in MM patient lysates compared with normal plasma cells and expression profiling indicated a relative induction of sumoylation pathway genes. The Sumo-conjugating enzyme Ube2I, the Sumo-ligase PIAS1, and the Sumo-inducer ARF were elevated in MM patient samples and cell lines. Survival correlated with expression because 80% of patients with low UBE2I and PIAS1 were living 6 years after transplantation, whereas only 45% of patients with high expression survived 6 years. UBE2I encodes the sole Sumo-conjugating enzyme in mammalian cells and cells transfected with a dominant-negative sumoylation-deficient UBE2I mutant exhibited decreased survival after radiation exposure, impaired adhesion to bone marrow stroma cell and decreased bone marrow stroma cell-induced proliferation. UBE2I confers cells with multiple advantages to promote tumorigenesis and predicts decreased survival when combined with PIAS1. The sumoylation pathway is a novel therapeutic target with implications for existing proteasomal-based treatment strategies.


Subject(s)
Multiple Myeloma/metabolism , Plasma Cells/metabolism , Protein Processing, Post-Translational , SUMO-1 Protein/metabolism , Bone Marrow Cells/metabolism , Cell Adhesion/genetics , Cell Line, Tumor , Cell Survival/genetics , Disease-Free Survival , Female , Gene Expression Regulation, Neoplastic/genetics , Humans , Male , Multiple Myeloma/genetics , Multiple Myeloma/mortality , Multiple Myeloma/therapy , Mutation , Proteasome Endopeptidase Complex/genetics , Proteasome Endopeptidase Complex/metabolism , Protein Inhibitors of Activated STAT/biosynthesis , SUMO-1 Protein/genetics , Small Ubiquitin-Related Modifier Proteins/biosynthesis , Stem Cell Transplantation , Stromal Cells/metabolism , Transplantation, Homologous , Ubiquitin-Conjugating Enzymes/biosynthesis
18.
Biotechnol Appl Biochem ; 54(1): 1-9, 2009 Jul 06.
Article in English | MEDLINE | ID: mdl-19575694

ABSTRACT

Antimicrobial peptides are an essential component of innate immunity and play an important role in host defence against microbial pathogens. They have received increasing attention recently as potential novel pharmaceutical agents. To meet the requirement for necessary basic science studies and clinical trials, large quantities of these peptides are needed. In general, isolation from natural sources and chemical synthesis are not cost-effective. The relatively low cost and easy scale-up of the recombinant approach renders it the most attractive means for large-scale production of antimicrobial peptides. Among the many systems available for protein expression, Escherichia coli remains the most widely used host. Antimicrobial peptides produced in E. coli are often expressed as fusion proteins, which effectively masks these peptides' potential lethal effect towards the bacterial host and protects the peptides from proteolytic degradation. Although some carriers confer peptide solubility, others promote the formation of inclusion bodies. The present minireview considers the most commonly used carrier proteins for fusion expression of antimicrobial peptides in E. coli. The favourable properties of SUMO (small ubiquitin-related modifier) as a novel fusion partner are also discussed.


Subject(s)
Antimicrobial Cationic Peptides/biosynthesis , Escherichia coli/metabolism , Protein Engineering/methods , Recombinant Fusion Proteins/biosynthesis , Recombinant Fusion Proteins/chemistry , Amidophosphoribosyltransferase/biosynthesis , Amidophosphoribosyltransferase/chemistry , Amidophosphoribosyltransferase/genetics , Antimicrobial Cationic Peptides/chemistry , Antimicrobial Cationic Peptides/genetics , Escherichia coli/genetics , Glutathione Transferase/biosynthesis , Glutathione Transferase/chemistry , Glutathione Transferase/genetics , Recombinant Fusion Proteins/genetics , Small Ubiquitin-Related Modifier Proteins/biosynthesis , Small Ubiquitin-Related Modifier Proteins/chemistry , Small Ubiquitin-Related Modifier Proteins/genetics , Thioredoxins/biosynthesis , Thioredoxins/chemistry , Thioredoxins/genetics
20.
J Cancer Res Clin Oncol ; 135(9): 1287-91, 2009 Sep.
Article in English | MEDLINE | ID: mdl-19288270

ABSTRACT

PURPOSE: Protein inhibitors of activated STATs (PIAS) regulate the interferon-gamma (IFN-gamma) signaling pathway, which has numerous effects on tumor development and tumor cell biology. PIAS's also regulate STAT family members not directly involved in IFN-gamma signaling. This project was designed to assess PIAS1 expression in colon cancer. METHODS: To determine whether PIAS1, one of the PIAS family members, or IFN-gamma signaling pathway components could be used to stratify colon tumors, we stained tissue microarrays for PIAS1, interferon regulatory factor-1 (IRF-1) and STAT1alpha. RESULTS: PIAS1 staining of the colon cancer tissue microarrays indicated a strong correlation of normal colon cells, and adenomas, with high expression of both PIAS1 and IRF-1. CONCLUSION: The PIAS1 results in particular may represent a basis for new approaches for efficiently distinguishing adenomas from colon cancer.


Subject(s)
Colonic Neoplasms/metabolism , Colonic Neoplasms/pathology , Disease Progression , Down-Regulation , Protein Inhibitors of Activated STAT/biosynthesis , Protein Inhibitors of Activated STAT/deficiency , Small Ubiquitin-Related Modifier Proteins/biosynthesis , Small Ubiquitin-Related Modifier Proteins/deficiency , Adult , Aged , Aged, 80 and over , Colonic Neoplasms/diagnosis , Female , Humans , Immunohistochemistry , Interferon Regulatory Factor-1/biosynthesis , Interferon Regulatory Factor-1/metabolism , Male , Middle Aged , Protein Inhibitors of Activated STAT/metabolism , Small Ubiquitin-Related Modifier Proteins/metabolism , Tissue Array Analysis
SELECTION OF CITATIONS
SEARCH DETAIL
...