Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 13 de 13
Filter
Add more filters










Publication year range
1.
Curr Mol Med ; 23(10): 991-1006, 2023.
Article in English | MEDLINE | ID: mdl-36239722

ABSTRACT

Aging is an inevitable risk factor for many diseases, including cardiovascular diseases, neurodegenerative diseases, cancer, and diabetes. Investigation into the molecular mechanisms involved in aging and longevity will benefit the treatment of age-dependent diseases and the development of preventative medicine for agingrelated diseases. Current evidence has revealed that FoxO3, encoding the transcription factor (FoxO)3, a key transcription factor that integrates different stimuli in the intrinsic and extrinsic pathways and is involved in cell differentiation, protein homeostasis, stress resistance and stem cell status, plays a regulatory role in longevity and in age-related diseases. However, the precise mechanisms by which the FoxO3 transcription factor modulates aging and promotes longevity have been unclear until now. Here, we provide a brief overview of the mechanisms by which FoxO3 mediates signaling in pathways involved in aging and aging-related diseases, as well as the current knowledge on the role of the FoxO3 transcription factor in the human lifespan and its clinical prospects. Ultimately, we conclude that FoxO3 signaling pathways, including upstream and downstream molecules, may be underlying therapeutic targets in aging and age-related diseases.


Subject(s)
Aging , Forkhead Box Protein O3 , Longevity , Humans , Aging/genetics , Forkhead Box Protein O3/genetics , Neoplasms/genetics
2.
Am J Physiol Heart Circ Physiol ; 319(6): H1302-H1312, 2020 12 01.
Article in English | MEDLINE | ID: mdl-33095057

ABSTRACT

Hyperphosphatemia is the primary risk factor for vascular calcification, which is closely associated with cardiovascular morbidity and mortality. Recent evidence showed that oxidative stress by high inorganic phosphate (Pi) mediates calcific changes in vascular smooth muscle cells (VSMCs). However, intracellular signaling responsible for Pi-induced oxidative stress remains unclear. Here, we investigated molecular mechanisms of Pi-induced oxidative stress related with intracellular Ca2+ ([Ca2+]i) disturbance, which is critical for calcification of VSMCs. VSMCs isolated from rat thoracic aorta or A7r5 cells were incubated with high Pi-containing medium. Extracellular signal-regulated kinase (ERK) and mammalian target of rapamycin were activated by high Pi that was required for vascular calcification. High Pi upregulated expressions of type III sodium-phosphate cotransporters PiT-1 and -2 and stimulated their trafficking to the plasma membrane. Interestingly, high Pi increased [Ca2+]i exclusively dependent on extracellular Na+ and Ca2+ as well as PiT-1/2 abundance. Furthermore, high-Pi induced plasma membrane depolarization mediated by PiT-1/2. Pretreatment with verapamil, as a voltage-gated Ca2+ channel (VGCC) blocker, inhibited Pi-induced [Ca2+]i elevation, oxidative stress, ERK activation, and osteogenic differentiation. These protective effects were reiterated by extracellular Ca2+-free condition, intracellular Ca2+ chelation, or suppression of oxidative stress. Mitochondrial superoxide scavenger also effectively abrogated ERK activation and osteogenic differentiation of VSMCs by high Pi. Taking all these together, we suggest that high Pi activates depolarization-triggered Ca2+ influx via VGCC, and subsequent [Ca2+]i increase elicits oxidative stress and osteogenic differentiation. PiT-1/2 mediates Pi-induced [Ca2+]i overload and oxidative stress but in turn, PiT-1/2 is upregulated by consequences of these alterations.NEW & NOTEWORTHY The novel findings of this study are type III sodium-phosphate cotransporters PiT-1 and -2-dependent depolarization by high Pi, leading to Ca2+ entry via voltage-gated Ca2+ channels in vascular smooth muscle cells. Cytosolic Ca2+ increase and subsequent oxidative stress are indispensable for osteogenic differentiation and calcification. In addition, plasmalemmal abundance of PiT-1/2 relies on Ca2+ overload and oxidative stress, establishing a positive feedback loop. Identification of mechanistic components of a vicious cycle could provide novel therapeutic strategies against vascular calcification in hyperphosphatemic patients.


Subject(s)
Calcium Signaling/drug effects , Calcium/metabolism , Hyperphosphatemia/chemically induced , Muscle, Smooth, Vascular/drug effects , Myocytes, Smooth Muscle/drug effects , Osteogenesis/drug effects , Oxidative Stress/drug effects , Phosphates/toxicity , Vascular Calcification/chemically induced , Animals , Calcium Channels/metabolism , Cell Line , Hyperphosphatemia/metabolism , Hyperphosphatemia/pathology , Male , Muscle, Smooth, Vascular/metabolism , Muscle, Smooth, Vascular/pathology , Myocytes, Smooth Muscle/metabolism , Myocytes, Smooth Muscle/pathology , Rats, Sprague-Dawley , Sodium-Phosphate Cotransporter Proteins, Type III/metabolism , Vascular Calcification/metabolism , Vascular Calcification/pathology
3.
Aging Cell ; 18(5): e12990, 2019 10.
Article in English | MEDLINE | ID: mdl-31264342

ABSTRACT

Paraquat (PQ) promotes cell senescence in brain tissue, which contributes to Parkinson's disease. Furthermore, PQ induces heart failure and oxidative damage, but it remains unknown whether and how PQ induces cardiac aging. Here, we demonstrate that PQ induces phenotypes associated with senescence of cardiomyocyte cell lines and results in cardiac aging-associated phenotypes including cardiac remodeling and dysfunction in vivo. Moreover, PQ inhibits the activation of Forkhead box O3 (FoxO3), an important longevity factor, both in vitro and in vivo. We found that PQ-induced senescence phenotypes, including proliferation inhibition, apoptosis, senescence-associated ß-galactosidase activity, and p16INK4a expression, were significantly enhanced by FoxO3 deficiency in cardiomyocytes. Notably, PQ-induced cardiac remolding, apoptosis, oxidative damage, and p16INK4a expression in hearts were exacerbated by FoxO3 deficiency. In addition, both in vitro deficiency and in vivo deficiency of FoxO3 greatly suppressed the activation of antioxidant enzymes including catalase (CAT) and superoxide dismutase 2 (SOD2) in the presence of PQ, which was accompanied by attenuation in cardiac function. The direct in vivo binding of FoxO3 to the promoters of the Cat and Sod2 genes in the heart was verified by chromatin immunoprecipitation (ChIP). Functionally, overexpression of Cat or Sod2 alleviated the PQ-induced senescence phenotypes in FoxO3-deficient cardiomyocyte cell lines. Overexpression of FoxO3 and CAT in hearts greatly suppressed the PQ-induced heart injury and phenotypes associated with aging. Collectively, these results suggest that FoxO3 protects the heart against an aging-associated decline in cardiac function in mice exposed to PQ, at least in part by upregulating the expression of antioxidant enzymes and suppressing oxidative stress.


Subject(s)
Aging/metabolism , Antioxidants/metabolism , Forkhead Box Protein O3/metabolism , Paraquat/antagonists & inhibitors , Protective Agents/metabolism , Up-Regulation , Aging/drug effects , Animals , Catalase/genetics , Catalase/metabolism , Heart/drug effects , Mice , Mice, Knockout , Paraquat/pharmacology , Phenotype , Superoxide Dismutase/genetics , Superoxide Dismutase/metabolism , Up-Regulation/drug effects
4.
Math Biosci Eng ; 16(3): 1376-1391, 2019 02 20.
Article in English | MEDLINE | ID: mdl-30947425

ABSTRACT

For discovery of new usage of drugs, the function type of their target genes plays an important role, and the hypothesis of "Antagonist-GOF" and "Agonist-LOF" has laid a solid foundation for supporting drug repurposing. In this research, an active gene annotation corpus was used as training data to predict the gain-of-function or loss-of-function or unknown character of each human gene after variation events. Unlike the design of(entity, predicate, entity) triples in a traditional three way tensor, a four way and a five way tensor, GMFD-/GMAFD-tensor, were designed to represent higher order links among or among part of these entities: genes(G), mutations(M), functions(F), diseases( D) and annotation labels(A). A tensor decomposition algorithm, CP decomposition, was applied to the higher order tensor and to unveil the correlation among entities. Meanwhile, a state-of-the-art baseline tensor decomposition algorithm, RESCAL, was carried on the three way tensor as a comparing method. The result showed that CP decomposition on higher order tensor performed better than RESCAL on traditional three way tensor in recovering masked data and making predictions. In addition, The four way tensor was proved to be the best format for our issue. At the end, a case study reproducing two disease-gene-drug links(Myelodysplatic Syndromes-IL2RA-Aldesleukin, Lymphoma- IL2RA-Aldesleukin) presented the feasibility of our prediction model for drug repurposing.


Subject(s)
Drug Repositioning/economics , Drug Repositioning/methods , Genetic Variation , Machine Learning , Mutation , Algorithms , Cost-Benefit Analysis , Genetic Diseases, Inborn/genetics , Humans , Interleukin-2/analogs & derivatives , Interleukin-2/therapeutic use , Interleukin-2 Receptor alpha Subunit/genetics , Lymphoma/genetics , Models, Genetic , Molecular Sequence Annotation , Myelodysplastic Syndromes/genetics , Recombinant Proteins/therapeutic use , Software
5.
FASEB J ; : fj201800093, 2018 Jun 13.
Article in English | MEDLINE | ID: mdl-29897811

ABSTRACT

The clustered regularly interspaced short palindromic repeats (CRISPR)/CRISPR-associated (Cas) 9 system has emerged as a powerful tool for knock-in of DNA fragments via donor plasmid and homology-independent DNA repair mechanism; however, conventional integration includes unnecessary plasmid backbone and may result in the unfaithful expression of the modified endogenous genes. Here, we report an efficient and precise CRISPR/Cas9-mediated integration strategy using a donor plasmid that harbors 2 of the same cleavage sites that flank the cassette at both sides. After the delivery of donor plasmid, together with Cas9 mRNA and guide RNA, into cells or fertilized eggs, concurrent cleavages at both sides of the exogenous cassette and the desired chromosomal site result in precise targeted integration without plasmid backbone. We successfully used this approach to precisely integrate the EGFP reporter gene into the myh6 locus or the GAPDH locus in Xenopus tropicalis or human cells, respectively. Furthermore, we demonstrate that replacing conventional terminators with the endogenous 3UTR of target genes in the cassette greatly improves the expression of reporter gene after integration. Our efficient and precise method will be useful for a variety of targeted genome modifications, not only in X. tropicalis, but also in mammalian cells, and can be readily adapted to many other organisms.-Mao, C.-Z., Zheng, L., Zhou, Y.-M., Wu, H.-Y., Xia, J.-B., Liang, C.-Q., Guo, X.-F., Peng, W.-T., Zhao, H., Cai, W.-B., Kim, S.-K., Park, K.-S., Cai, D.-Q., Qi, X.-F. CRISPR/Cas9-mediated efficient and precise targeted integration of donor DNA harboring double cleavage sites in Xenopus tropicalis.

6.
J Cell Physiol ; 233(5): 4245-4257, 2018 05.
Article in English | MEDLINE | ID: mdl-29057537

ABSTRACT

Unmethylated CpG oligodeoxynucleotide (CpG-ODN), a Toll-like receptor 9 (TLR9) ligand, has been shown to protect against myocardial ischemia/reperfusion injury. However, the potential effects of CpG-ODN on myocardial infarction (MI) induced by persistent ischemia remains unclear. Here, we investigated whether and how CpG-ODN preconditioning protects against MI in mice. C57BL/6 mice were treated with CpG-ODN by i.p. injection 2 hr prior to MI induction, and cardiac function, and histology were analyzed 2 weeks after MI. Both 1826-CpG and KSK-CpG preconditioning significantly improved the left ventricular (LV) ejection fraction (LVEF) and LV fractional shortening (LVFS) when compared with non-CpG controls. Histological analysis further confirmed the cardioprotection of CpG-ODN preconditioning. In vitro studies further demonstrated that CpG-ODN preconditioning increases cardiomyocyte survival under hypoxic/ischemic conditions by enhancing stress tolerance through TLR9-mediated inhibition of the SERCA2/ATP and activation of AMPK pathways. Moreover, CpG-ODN preconditioning significantly increased angiogenesis in the infarcted myocardium compared with non-CpG. However, persistent TLR9 activation mediated by lentiviral infection failed to improve cardiac function after MI. Although CpG-ODN preconditioning increased angiogenesis in vitro, both the persistent stimulation of CpG-ODN and stable overexpression of TLR9 suppressed the tube formation of cardiac microvascular endothelial cells. CpG-ODN preconditioning significantly protects cardiac function against MI by suppressing the energy metabolism of cardiomyocytes and promoting angiogenesis. Our data also indicate that CpG-ODN preconditioning may be useful in MI therapy.


Subject(s)
Myocardial Infarction/drug therapy , Neovascularization, Pathologic/drug therapy , Oligodeoxyribonucleotides/administration & dosage , Ventricular Function, Left/drug effects , Animals , Disease Models, Animal , Energy Metabolism/drug effects , Humans , Ischemic Preconditioning, Myocardial/methods , Mice , Myocardial Infarction/genetics , Myocardial Infarction/pathology , Neovascularization, Pathologic/genetics , Neovascularization, Pathologic/pathology , Sarcoplasmic Reticulum Calcium-Transporting ATPases/genetics , Toll-Like Receptor 9/genetics
7.
Sci Rep ; 7(1): 13273, 2017 10 16.
Article in English | MEDLINE | ID: mdl-29038511

ABSTRACT

Vascular endothelial growth factor (VEGF) plays important roles in improvement of cardiac function following myocardial infarction (MI). However, the lack of a steerable delivery system of VEGF targeting the infarcted myocardium reduces the therapeutic efficacy and safety. Here, we constructed a series of lentiviral vector systems which could express a fusion protein consisted of a collagen-binding domain (CBD) and hVEGF (CBDhVEGF), under the control of 5HRE-hCMVmp (5HRE), the hypoxia-inducible promoter consists of five copies of the hypoxia-responsive element (HRE) and a human cytomegalovirus minimal promoter (hCMVmp). We demonstrated that 5HRE has the comparable ability to strongly drive CBDhVEGF under hypoxic condition as the ubiquitous CMV promoter, but it can hardly drive target gene under normoxic condition. 5HRE-drived CBDhVEGF specifically bound to type I collagen and significantly promoted the viability of HUVEC cells. Moreover, after injection of lentivirus into heart of mouse with MI, CBDhVEGF was mainly retained in infarcted myocardium where containing rich collagen and significantly improved angiogenesis and cardiac function when compared with hVEGF. Moreover, CBDhVEGF mediated by lentivirus has little leakage from infarcted zone into blood than hVEGF. Taken together, our results indicate that 5HRE-CBDhVEGF lentiviral vector system could improve cardiac function in the collagen-targeting and hypoxia-inducible manners.


Subject(s)
Collagen/genetics , Gene Transfer Techniques , Hypoxia/genetics , Myocardial Infarction/metabolism , Myocardial Infarction/physiopathology , Vascular Endothelial Growth Factors/genetics , Animals , Cell Line , Collagen/metabolism , Disease Models, Animal , Echocardiography , Gene Expression , Genetic Therapy , Genetic Vectors/genetics , Human Umbilical Vein Endothelial Cells , Humans , Hypoxia/metabolism , Lentivirus/genetics , Mice , Myocardial Infarction/etiology , Myocardial Infarction/therapy , Promoter Regions, Genetic , Response Elements , Vascular Endothelial Growth Factors/metabolism
8.
Gene Expr Patterns ; 23-24: 1-6, 2017 01.
Article in English | MEDLINE | ID: mdl-28034797

ABSTRACT

Recent studies suggest that ribosome-binding protein 1 (RRBP1) is involved in multiple diseases such as tumorigenesis and cardiomyopathies. However, its function during embryonic development remains largely unknown. We searched Xenopus laevis database with human RRBP1 protein sequence and identified two cDNA sequences encoding Xenopus orthologs of RRBP1 including rrbp1a (NM_001089623) and rrbp1b (NM_001092468). Both genes were firstly detected at blastula stage 8 with weak signals in animal hemisphere by whole mount in situ hybridization. Evident expression of rrbp1 was mainly detected in cement gland and notochord at neurula and tailbud stages. Heart expression of rrbp1 was detected at stage 36. RT-PCR results indicated that very weak expression of rrbp1a was firstly detected in oocytes, followed by increasing expression until stage 39. Differently, very weak expression of rrbp1b was firstly observed at stage 2, and then maintained at a lower level to stage 17 followed by an intense expression from stages 19-39. Moreover, both expression profiles were also different in adult tissues. This study reports Xenopus rrbp1 expression during early embryonic development and in adult tissues. Our study will facilitate the functional analysis of Rrbp1 family during embryonic development.


Subject(s)
Carrier Proteins/genetics , Xenopus Proteins/genetics , Xenopus laevis/embryology , Xenopus laevis/metabolism , Animals , Carrier Proteins/chemistry , Carrier Proteins/metabolism , Embryo, Nonmammalian/metabolism , Embryonic Development , Endoplasmic Reticulum/metabolism , Gene Expression Regulation, Developmental , Myocardium/metabolism , Transcriptome , Xenopus Proteins/chemistry , Xenopus Proteins/metabolism , Xenopus laevis/genetics
9.
Cytokine ; 81: 63-70, 2016 May.
Article in English | MEDLINE | ID: mdl-26891076

ABSTRACT

CXCL10, the chemokine with potent chemotactic activity on immune cells and other non-immune cells expressing its receptor CXCR3, has been demonstrated to involve in myocardial infarction, which was resulted from hypoxia/ischemia. The cardiac microvascular endothelial cells (CMECs) are the first cell type which is implicated by hypoxia/ischemia. However, the potential molecular mechanism by which hypoxia/ischemia regulates the expression of CXCL10 in CMECs remains unclear. In the present study, the expression of CXCL10 was firstly examined by real-time PCR and ELISA analysis. Several potential binding sites (BS) for transcription factors including NF-kappaB (NFkB), HIF1 alpha (HIF1α) and FoxO3a were identified in the promoter region of CXCL10 gene from -2000 bp to -1 bp using bioinformatics software. Luciferase reporter gene vectors for CXCL10 promoter and for activation of above transcription factors were constructed. The activation of NFkB, hypoxia-inducible transcription factor-1 alpha (HIF-1α) and FoxO3a was also analyzed by Western blotting. It was shown that the production of CXCL10 in CMECs was significantly increased by hypoxia/ischemia treatment, in parallel with the activation of CXCL10 promoter examined by reporter gene vector system. Furthermore, transcription factors including NFkB, HIF1α and FoxO3a were activated by hypoxia/ischemia in CMECs. However, over-expression of NFkB, but not that of HIF1α or FoxO3a, significantly promoted the activation of CXCL10 promoter reporter gene. These findings indicated that CXCL10 production in CMECs was significantly increased by hypoxia/ischemia, at least in part, through activation of NFkB pathway and subsequently binding to CXCL10 promoter, finally promoted the transcription of CXCL10 gene.


Subject(s)
Chemokine CXCL10/metabolism , Coronary Vessels/cytology , Endothelial Cells/metabolism , NF-kappa B/metabolism , Animals , Base Sequence , Binding Sites/genetics , Blotting, Western , Cell Hypoxia , Cells, Cultured , Chemokine CXCL10/genetics , Enzyme-Linked Immunosorbent Assay , Forkhead Box Protein O3/genetics , Forkhead Box Protein O3/metabolism , Gene Expression , Hypoxia-Inducible Factor 1, alpha Subunit/genetics , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Ischemia , NF-kappa B/genetics , Promoter Regions, Genetic/genetics , Protein Binding , Rats, Sprague-Dawley , Reverse Transcriptase Polymerase Chain Reaction
10.
Exp Mol Pathol ; 100(2): 257-65, 2016 Apr.
Article in English | MEDLINE | ID: mdl-26835911

ABSTRACT

CXCL10 is a chemokine with potent chemotactic activity for immune and non-immune cells expressing its receptor CXCR3. Previous studies have demonstrated that CXCL10 is involved in myocardial infarction. However, the role of CXCL10 in cardiac microvascular endothelial cell (CMEC) regulation and related mechanisms remains unclear. In this study, we investigated the effects of CXCL10 on the CMEC migration and explored its potential molecular mechanism by wound healing, cell proliferation and viability analysis. Furthermore, migration-related signaling pathways, including FAK, Erk, p38 and Smad, were examined by Western blotting. We found that CXCL10 significantly promotes CMEC migration under normal conditions and during hypoxia/ischemia. However, no significant differences in CMEC proliferation and viability were observed with or without CXCL10 treatment. CXCL10-mediated CMEC migration was greatly blocked by treatment with an anti-CXCR3 antibody. Although CXCL10 treatment promoted phosphorylation and activation of the FAK, Erk, and p38 pathways during hypoxia/ischemia, CXCL10-mediated CMEC migration was significantly blocked by p38 and FAK inhibitors, but not by an Erk inhibitor. Furthermore, CXCL10-mediated FAK activation was suppressed by the p38 inhibitor. These findings indicated that the CXCL10/CXCR3 pathway promotes the migration of CMECs under normal conditions and during hypoxia/ischemia in a proliferation-independent manner, at least in part, through regulation of the p38/FAK pathways.


Subject(s)
Cell Movement/drug effects , Cell Proliferation/drug effects , Chemokine CXCL10/pharmacology , Endothelial Cells/metabolism , Focal Adhesion Kinase 1/metabolism , Receptors, CXCR3/metabolism , p38 Mitogen-Activated Protein Kinases/metabolism , Animals , Blotting, Western , Cell Hypoxia , Cells, Cultured , Coronary Vessels/cytology , Enzyme Activation/drug effects , Enzyme Inhibitors/pharmacology , Focal Adhesion Kinase 1/antagonists & inhibitors , Gene Expression/drug effects , Models, Biological , Phosphorylation/drug effects , Rats, Sprague-Dawley , Receptors, CXCR3/genetics , Reverse Transcriptase Polymerase Chain Reaction , Signal Transduction/drug effects , p38 Mitogen-Activated Protein Kinases/antagonists & inhibitors
11.
J Mol Cell Cardiol ; 81: 114-26, 2015 Apr.
Article in English | MEDLINE | ID: mdl-25655933

ABSTRACT

FoxO3a plays an important role in the aging process and decreases with age. However, the potential regulatory roles of FoxO3a in processes involved in cardiac microvascular endothelial cell (CMEC) senescence, and its underlying molecular mechanisms have not been elucidated. This study demonstrates that FoxO3a is deactivated in senescent CMECs together with the inhibition of proliferation and tube formation. Furthermore, the activation of the antioxidant enzymes catalase and SOD, downstream FoxO3a targets, was significantly decreased, thereby leading to cell cycle arrest in G1-phase by increased ROS generation and subsequently the activation of the p27(Kip1) pathway. However, FoxO3a overexpression in primary low-passage CMECs not only significantly suppressed the senescence process by increasing the activation of catalase and SOD but also markedly inhibited ROS generation and p27(Kip1) activation, although it failed to reverse cellular senescence. Moreover, both cell viability and tube formation were greatly increased by FoxO3a overexpression in primary CMECs during continuous passage. In addition, FoxO3a, deficiency in low-passage CMECs, accelerated the senescence process. Collectively, our data suggest that FoxO3a suppresses the senescence process in CMECs by regulating the antioxidant/ROS/p27(Kip1) pathways, although it fails to reverse the cellular senescent phenotype.


Subject(s)
Cyclin-Dependent Kinase Inhibitor p27/genetics , Endothelial Cells/metabolism , Forkhead Transcription Factors/genetics , Myocardium/metabolism , Reactive Oxygen Species/metabolism , Animals , Base Sequence , Catalase/genetics , Catalase/metabolism , Cell Survival , Cellular Senescence , Cyclin-Dependent Kinase Inhibitor p27/metabolism , Endothelial Cells/pathology , Forkhead Box Protein O3 , Forkhead Transcription Factors/metabolism , G1 Phase Cell Cycle Checkpoints/genetics , Gene Expression Regulation , Genes, Reporter , Lentivirus/genetics , Luciferases/genetics , Luciferases/metabolism , Molecular Sequence Data , Myocardium/pathology , Primary Cell Culture , Rats , Rats, Sprague-Dawley , Signal Transduction , Superoxide Dismutase/genetics , Superoxide Dismutase/metabolism
12.
J Theor Biol ; 343: 186-92, 2014 Feb 21.
Article in English | MEDLINE | ID: mdl-24189096

ABSTRACT

DNA-binding proteins play a vitally important role in many biological processes. Prediction of DNA-binding proteins from amino acid sequence is a significant but not fairly resolved scientific problem. Chaos game representation (CGR) investigates the patterns hidden in protein sequences, and visually reveals previously unknown structure. Fractal dimensions (FD) are good tools to measure sizes of complex, highly irregular geometric objects. In order to extract the intrinsic correlation with DNA-binding property from protein sequences, CGR algorithm, fractal dimension and amino acid composition are applied to formulate the numerical features of protein samples in this paper. Seven groups of features are extracted, which can be computed directly from the primary sequence, and each group is evaluated by the 10-fold cross-validation test and Jackknife test. Comparing the results of numerical experiments, the group of amino acid composition and fractal dimension (21-dimension vector) gets the best result, the average accuracy is 81.82% and average Matthew's correlation coefficient (MCC) is 0.6017. This resulting predictor is also compared with existing method DNA-Prot and shows better performances.


Subject(s)
Computational Biology/methods , DNA-Binding Proteins/metabolism , Fractals , Support Vector Machine , DNA-Binding Proteins/chemistry , Databases, Protein , Models, Molecular , Nonlinear Dynamics , Protein Structure, Tertiary , Regression Analysis , Reproducibility of Results
13.
Protein Pept Lett ; 19(9): 940-8, 2012 Sep.
Article in English | MEDLINE | ID: mdl-22486614

ABSTRACT

Obtaining soluble proteins in sufficient concentrations is a major obstacle in various experimental studies. How to predict the propensity of targets in large-scale proteomics projects to be soluble is a significant but not fairly resolved scientific problem. Chaos game representation (CGR) can investigate the patterns hiding in protein sequences, and can visually reveal previously unknown structure. Fractal dimensions are good tools to measure sizes of complex, highly irregular geometric objects. In this paper, we convert each protein sequence into a high-dimensional vector by CGR algorithm and fractal dimension, and then predict protein solubility by these fractal features together with Chou's pseudo amino acid composition features and support vector machine (SVM). We extract and study six groups of features computed directly from the primary sequence, and each group is evaluated by the 10-fold cross-validation test. As the results of comparisons, the group of 445-dimensional vector gets the best results, the average accuracy is 0.8741 and average MCC is 0.7358. The resulting predictor is also compared with existing methods and shows significant improvement.


Subject(s)
Fractals , Nonlinear Dynamics , Proteins/chemistry , Amino Acids/chemistry , Models, Chemical , Solubility , Support Vector Machine
SELECTION OF CITATIONS
SEARCH DETAIL
...