Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 13 de 13
Filter
1.
J Bionic Eng ; : 1-19, 2023 Mar 01.
Article in English | MEDLINE | ID: mdl-37361682

ABSTRACT

Nowadays, meta-heuristic algorithms are attracting widespread interest in solving high-dimensional nonlinear optimization problems. In this paper, a COVID-19 prevention-inspired bionic optimization algorithm, named Coronavirus Mask Protection Algorithm (CMPA), is proposed based on the virus transmission of COVID-19. The main inspiration for the CMPA originated from human self-protection behavior against COVID-19. In CMPA, the process of infection and immunity consists of three phases, including the infection stage, diffusion stage, and immune stage. Notably, wearing masks correctly and safe social distancing are two essential factors for humans to protect themselves, which are similar to the exploration and exploitation in optimization algorithms. This study simulates the self-protection behavior mathematically and offers an optimization algorithm. The performance of the proposed CMPA is evaluated and compared to other state-of-the-art metaheuristic optimizers using benchmark functions, CEC2020 suite problems, and three truss design problems. The statistical results demonstrate that the CMPA is more competitive among these state-of-the-art algorithms. Further, the CMPA is performed to identify the parameters of the main girder of a gantry crane. Results show that the mass and deflection of the main girder can be improved by 16.44% and 7.49%, respectively.

2.
Opt Lett ; 45(15): 4224-4227, 2020 Aug 01.
Article in English | MEDLINE | ID: mdl-32735264

ABSTRACT

We experimentally demonstrate a real-time quantum random number generator by using a room-temperature single-photon emitter from the defect in a commercial gallium nitride wafer. Due to the brightness of our single-photon emitter, the raw bit generation rate is about 1.8 MHz, and the unbiased bit generation rate is about 420 kHz after the von Neumann's randomness extraction procedure. Our results show that the commercial gallium nitride wafer has great potential for the development of integrated high-speed quantum random number generator devices.

3.
Materials (Basel) ; 11(5)2018 Apr 27.
Article in English | MEDLINE | ID: mdl-29702610

ABSTRACT

A flake is a crack that is induced by trapped hydrogen within steel. To study its formation mechanism, previous studies mostly focused on the formation process and magnitude of hydrogen pressure in hydrogen traps such as cavities and cracks. However, according to recent studies, the hydrogen leads to the decline of the mechanical properties of steel, which is known as hydrogen embrittlement, is another reason for flake formation. In addition, the phenomenon of stress induced hydrogen uphill diffusion should not be neglected. All of the three behaviors are at work simultaneously. In order to further explore the formation mechanism of flakes in steel, the process of flake initiation and growth were studied with the following three coupling factors: trap hydrogen pressure, hydrogen embrittlement, and stress induced hydrogen re-distribution. The analysis model was established using the finite element method, and a crack whose radius is 0.5 mm was set in its center. The cohesive method and Bilinear Traction Separate Law (BTSL) were used to address the coupling effect. The results show that trap hydrogen pressure is the main driving force for flake formation. After the high hydrogen pressure was generated around the trap, a stress field formed. In addition, the trap is the center of stress concentration. Then, hydrogen is concentrated in a distribution around this trap, and most of the steel mechanical properties are reduced. The trap size is a key factor for defining the critical hydrogen content for flake formation and propagation. However, when the trap size exceeds the specified value, the critical hydrogen content does not change any more. As for the crack whose radius is 0.5 mm, the critical hydrogen content of Cr5VMo steel is 2.2 ppm, which is much closer to the maximum safe hydrogen concentration of 2.0 ppm used in China. The work presented in this article increases our understanding of flake formation and propagation mechanisms in steel.

4.
PLoS Genet ; 12(5): e1006054, 2016 05.
Article in English | MEDLINE | ID: mdl-27195754

ABSTRACT

Hedgehog (Hh) signaling regulates multiple aspects of metazoan development and tissue homeostasis, and is constitutively active in numerous cancers. We identified Ubr3, an E3 ubiquitin ligase, as a novel, positive regulator of Hh signaling in Drosophila and vertebrates. Hh signaling regulates the Ubr3-mediated poly-ubiquitination and degradation of Cos2, a central component of Hh signaling. In developing Drosophila eye discs, loss of ubr3 leads to a delayed differentiation of photoreceptors and a reduction in Hh signaling. In zebrafish, loss of Ubr3 causes a decrease in Shh signaling in the developing eyes, somites, and sensory neurons. However, not all tissues that require Hh signaling are affected in zebrafish. Mouse UBR3 poly-ubiquitinates Kif7, the mammalian homologue of Cos2. Finally, loss of UBR3 up-regulates Kif7 protein levels and decreases Hh signaling in cultured cells. In summary, our work identifies Ubr3 as a novel, evolutionarily conserved modulator of Hh signaling that boosts Hh in some tissues.


Subject(s)
Drosophila Proteins/genetics , Eye/metabolism , Kinesins/genetics , Ubiquitin-Protein Ligases/genetics , Animals , Drosophila Proteins/metabolism , Drosophila melanogaster/genetics , Drosophila melanogaster/growth & development , Eye/growth & development , Hedgehog Proteins/genetics , Kinesins/metabolism , Mice , Photoreceptor Cells/metabolism , Polyubiquitin , Proteolysis , RNA, Small Interfering , Signal Transduction , Ubiquitin-Protein Ligases/metabolism , Ubiquitination , Zebrafish/genetics
5.
PLoS Biol ; 14(2): e1002375, 2016 Feb.
Article in English | MEDLINE | ID: mdl-26863604

ABSTRACT

In Hedgehog (Hh) signaling, binding of Hh to the Patched-Interference Hh (Ptc-Ihog) receptor complex relieves Ptc inhibition on Smoothened (Smo). A longstanding question is how Ptc inhibits Smo and how such inhibition is relieved by Hh stimulation. In this study, we found that Hh elevates production of phosphatidylinositol 4-phosphate (PI(4)P). Increased levels of PI(4)P promote, whereas decreased levels of PI(4)P inhibit, Hh signaling activity. We further found that PI(4)P directly binds Smo through an arginine motif, which then triggers Smo phosphorylation and activation. Moreover, we identified the pleckstrin homology (PH) domain of G protein-coupled receptor kinase 2 (Gprk2) as an essential component for enriching PI(4)P and facilitating Smo activation. PI(4)P also binds mouse Smo (mSmo) and promotes its phosphorylation and ciliary accumulation. Finally, Hh treatment increases the interaction between Smo and PI(4)P but decreases the interaction between Ptc and PI(4)P, indicating that, in addition to promoting PI(4)P production, Hh regulates the pool of PI(4)P associated with Ptc and Smo.


Subject(s)
Drosophila Proteins/metabolism , G-Protein-Coupled Receptor Kinase 2/metabolism , Hedgehog Proteins/metabolism , Phosphatidylinositol Phosphates/metabolism , Receptors, G-Protein-Coupled/metabolism , Animals , Cilia/metabolism , Drosophila , Mice , NIH 3T3 Cells , Patched Receptors , Patched-1 Receptor , Phosphorylation , Receptors, Cell Surface/metabolism , Smoothened Receptor
6.
Proc Natl Acad Sci U S A ; 111(45): E4842-50, 2014 Nov 11.
Article in English | MEDLINE | ID: mdl-25349414

ABSTRACT

Smoothened (Smo) is essential for transduction of the Hedgehog (Hh) signal in both insects and vertebrates. Cell surface/cilium accumulation of Smo is thought to play an important role in Hh signaling, but how the localization of Smo is controlled remains poorly understood. In this study, we demonstrate that atypical PKC (aPKC) regulates Smo phosphorylation and basolateral accumulation in Drosophila wings. Inactivation of aPKC by either RNAi or a mutation inhibits Smo basolateral accumulation and attenuates Hh target gene expression. In contrast, expression of constitutively active aPKC elevates basolateral accumulation of Smo and promotes Hh signaling. The aPKC-mediated phosphorylation of Smo at Ser680 promotes Ser683 phosphorylation by casein kinase 1 (CK1), and these phosphorylation events elevate Smo activity in vivo. Moreover, aPKC has an additional positive role in Hh signaling by regulating the activity of Cubitus interruptus (Ci) through phosphorylation of the Zn finger DNA-binding domain. Finally, the expression of aPKC is up-regulated by Hh signaling in a Ci-dependent manner. Our findings indicate a direct involvement of aPKC in Hh signaling beyond its role in cell polarity.


Subject(s)
DNA-Binding Proteins/metabolism , Drosophila Proteins/metabolism , Gene Expression Regulation, Enzymologic/physiology , Hedgehog Proteins/metabolism , Protein Kinase C/biosynthesis , Receptors, G-Protein-Coupled/metabolism , Signal Transduction/physiology , Transcription Factors/metabolism , Animals , Casein Kinase I/genetics , Casein Kinase I/metabolism , DNA-Binding Proteins/genetics , Drosophila Proteins/genetics , Drosophila melanogaster , Hedgehog Proteins/genetics , Phosphorylation/physiology , Protein Kinase C/genetics , Protein Structure, Tertiary , Receptors, G-Protein-Coupled/genetics , Smoothened Receptor , Transcription Factors/genetics , Up-Regulation/physiology , Wings, Animal/metabolism
7.
PLoS One ; 8(11): e79021, 2013.
Article in English | MEDLINE | ID: mdl-24244405

ABSTRACT

In Hedgehog (Hh) signaling, the seven-transmembrane protein Smoothened (Smo) acts as a signal transducer that is regulated by phosphorylation, ubiquitination, and cell surface accumulation. However, it is not clear how Smo cell surface accumulation and intracellular trafficking are regulated. Here, we demonstrate that inactivation of Hrs by deletion or RNAi accumulates Smo in the late endosome that is marked by late endosome markers. Inactivation of Hrs enhances the wing defects caused by dominant-negative Smo. We show that Hrs promotes Smo ubiquitination, deleting the ubiquitin-interacting-motif (UIM) in Hrs abolishes the ability of Hrs to regulate Smo ubiquitination. However, the UIM domain neither recognizes the ubiquitinated Smo nor directly interacts with Smo. Hrs lacking UIM domain still downregulates Smo activity even though to a less extent. We have characterized that the N-terminus of Hrs directly interacts with the PKA/CK1 phosphorylation clusters to prevent Smo phosphorylation and activation, indicating an ubiquitin-independent regulation of Smo by Hrs. Finally, we found that knockdown of Tsg101 accumulates Smo that is co-localized with Hrs and other late endosome markers. Taken together, our data indicate that Hrs mediates Smo trafficking in the late endosome by not only promoting Smo ubiquitination but also blocking Smo phosphorylation.


Subject(s)
Drosophila Proteins/metabolism , Endosomal Sorting Complexes Required for Transport/metabolism , Endosomes/metabolism , Hedgehog Proteins/metabolism , Phosphoproteins/metabolism , Receptors, G-Protein-Coupled/metabolism , Signal Transduction/physiology , Ubiquitination/physiology , Animals , Drosophila Proteins/genetics , Drosophila melanogaster , Endosomal Sorting Complexes Required for Transport/genetics , Endosomes/genetics , Hedgehog Proteins/genetics , Phosphoproteins/genetics , Protein Transport/physiology , Receptors, G-Protein-Coupled/genetics , Smoothened Receptor
8.
Dev Biol ; 366(2): 172-84, 2012 Jun 15.
Article in English | MEDLINE | ID: mdl-22537496

ABSTRACT

The activation of Smoothened (Smo) requires phosphorylation at three clusters of Serine residues in Drosophila Hedgehog (Hh) signaling. However, the mechanism by which phosphorylation promotes Smo conformational change and subsequently activates Smo in response to Hh gradient remains unclear. Here, we show that the conformational states of Smo are determined by not only the amount but also the position of the negative charges provided by phosphorylation. By using a Smo phospho-specific antibody, we demonstrate that Smo is differentially phosphorylated at three clusters of serine residues in response to levels of Hh activity. Mutating the first cluster, compared to mutating the other clusters, impairs Smo activity more severely, whereas mutating the last cluster prohibits C-terminus dimerization. In addition, phosphorylation of the membrane proximal cluster promotes phosphorylation of the distal cluster. We propose a zipper-lock model in which the gradual phosphorylation at these clusters induces a gradual conformational change in the Smo cytoplasmic tail, which promotes the interaction between Smo and Costal2 (Cos2). Moreover, we show that Hh regulates both PKA and CK1 phosphorylation of Smo. Thus, the differential phosphorylation of Smo mediates the thresholds of Hh activity.


Subject(s)
Drosophila Proteins/metabolism , Drosophila/metabolism , Hedgehog Proteins/metabolism , Receptors, G-Protein-Coupled/metabolism , Animals , Drosophila Proteins/chemistry , Models, Molecular , Phosphorylation , Protein Conformation , Protein Structure, Tertiary , Receptors, G-Protein-Coupled/chemistry , Signal Transduction , Smoothened Receptor
9.
PLoS One ; 7(4): e35153, 2012.
Article in English | MEDLINE | ID: mdl-22509396

ABSTRACT

Prostate cancer is a major health problem for men in Western societies. Here we report a Prostate Cancer-Specific Targeting Gene-Viro-Therapy (CTGVT-PCa), in which PTEN was inserted into a DD3-controlled oncolytic viral vector (OV) to form Ad.DD3.E1A.E1B(Δ55)-(PTEN) or, briefly, Ad.DD3.D55-PTEN. The woodchuck post-transcriptional element (WPRE) was also introduced at the downstream of the E1A coding sequence, resulting in much higher expression of the E1A gene. DD3 is one of the most prostate cancer-specific genes and has been used as a clinical bio-diagnostic marker. PTEN is frequently inactivated in primary prostate cancers, which is crucial for prostate cancer progression. Therefore, the Ad.DD3.D55-PTEN has prostate cancer specific and potent antitumor effect. The tumor growth rate was almost completely inhibited with the final tumor volume after Ad.DD3.D55-PTEN treatment less than the initial volume at the beginning of Ad.DD3.D55-PTEN treatment, which shows the powerful antitumor effect of Ad.DD3.D55-PTEN on prostate cancer tumor growth. The CTGVT-PCa construct reported here killed all of the prostate cancer cell lines tested, such as DU145, 22RV1 and CL1, but had a reduced or no killing effect on all the non-prostate cancer cell lines tested. The mechanism of action of Ad.DD3.D55-PTEN was due to the induction of apoptosis, as detected by TUNEL assays and flow cytometry. The apoptosis was mediated by mitochondria-dependent and -independent pathways, as determined by caspase assays and mitochondrial membrane potential.


Subject(s)
Antigens, Neoplasm/genetics , Genetic Therapy/methods , Oncolytic Virotherapy/methods , PTEN Phosphohydrolase/genetics , Prostatic Neoplasms/therapy , Adenovirus E1A Proteins , Animals , Antigens, Neoplasm/metabolism , Apoptosis/genetics , Cell Line, Tumor , Genetic Vectors , Humans , Male , Mice , Mice, Nude , Oncolytic Viruses/genetics , Prostatic Neoplasms/genetics
10.
PLoS Biol ; 10(1): e1001238, 2012 Jan.
Article in English | MEDLINE | ID: mdl-22253573

ABSTRACT

The seven transmembrane protein Smoothened (Smo) is a critical component of the Hedgehog (Hh) signaling pathway and is regulated by phosphorylation, dimerization, and cell-surface accumulation upon Hh stimulation. However, it is not clear how Hh regulates Smo accumulation on the cell surface or how Hh regulates the intracellular trafficking of Smo. In addition, little is known about whether ubiquitination is involved in Smo regulation. In this study, we demonstrate that Smo is multi-monoubiquitinated and that Smo ubiquitination is inhibited by Hh and by phosphorylation. Using an in vivo RNAi screen, we identified ubiquitin-specific protease 8 (USP8) as a deubiquitinase that down-regulates Smo ubiquitination. Inactivation of USP8 increases Smo ubiquitination and attenuates Hh-induced Smo accumulation, leading to decreased Hh signaling activity. Moreover, overexpression of USP8 prevents Smo ubiquitination and elevates Smo accumulation, leading to increased Hh signaling activity. Mechanistically, we show that Hh promotes the interaction of USP8 with Smo aa625-753, which covers the three PKA and CK1 phosphorylation clusters. Finally, USP8 promotes the accumulation of Smo at the cell surface and prevents localization to the early endosomes, presumably by deubiquitinating Smo. Our studies identify USP8 as a positive regulator in Hh signaling by down-regulating Smo ubiquitination and thereby mediating Smo intracellular trafficking.


Subject(s)
Endopeptidases/physiology , Endosomal Sorting Complexes Required for Transport/physiology , Receptors, G-Protein-Coupled/metabolism , Ubiquitin Thiolesterase/physiology , Animals , Drosophila , Endopeptidases/genetics , Endosomal Sorting Complexes Required for Transport/genetics , HEK293 Cells , Humans , Mice , Models, Biological , NIH 3T3 Cells , Protein Transport/genetics , Receptors, G-Protein-Coupled/genetics , Signal Transduction/genetics , Signal Transduction/physiology , Smoothened Receptor , Subcellular Fractions/metabolism , Tissue Distribution , Transfection , Ubiquitin Thiolesterase/genetics , Ubiquitination/genetics
11.
J Mol Med (Berl) ; 84(12): 1077-86, 2006 Dec.
Article in English | MEDLINE | ID: mdl-17077982

ABSTRACT

A major goal in cancer gene therapy is to develop efficient gene transfer protocols that allow tissue-specific and tightly regulated expression of therapeutic genes. The ideal vector should efficiently transduce cancer cells with minimal toxicity on normal tissues and persistently express foreign genes. One of the most promising regulatory systems is the mifepristone/RU486-regulated system, which has much lower basal transcriptional activity and high inducibility. In this work, we modified this system by incorporating a cancer-specific promoter, the human telomerase reverse transcriptase (hTERT) promoter. By utilizing hTERT promoter to control the regulator, RU486 could specifically induce the expression of foreign genes in cancer cells but not in normal cells. In the context of this system, a dominant negative mutant of survivin (surDN) was controllably expressed in colorectal tumor cells. The surDN expression induced by RU486 showed a dosage- and time-dependent pattern. Regulated expression of surDN caused caspase-dependent apoptosis in colorectal tumor cells but had little effect on normal cells. Analysis of cell viability showed that RU486-induced expression of surDN suppressed colorectal tumor cell growth and had synergic effect in combination with chemotherapeutic agents. The potential of this system in cancer therapy was evaluated in experimental animals. Tumor xenograft models were established in nude mice with colorectal tumor cells, and RU486 was intraperitoneally administered. The results showed that conditional expression of surDN efficiently inhibited tumor growth in vivo and prolonged the life of tumor-burdened mice. Synergized with the chemotherapeutic drug cisplatin, regulated surDN expression completely suppressed tumor growth. These results indicated that this modified RU486-regulated system could be useful in cancer-targeting therapy.


Subject(s)
Colorectal Neoplasms/genetics , Microtubule-Associated Proteins/physiology , Mifepristone/therapeutic use , Neoplasm Proteins/physiology , Promoter Regions, Genetic , Telomerase/genetics , Animals , Antimetabolites, Antineoplastic/therapeutic use , Antineoplastic Agents/therapeutic use , Apoptosis , Caspases/metabolism , Cell Line , Cell Line, Tumor , Cell Survival/drug effects , Cisplatin/therapeutic use , Colorectal Neoplasms/drug therapy , Colorectal Neoplasms/metabolism , Colorectal Neoplasms/pathology , Dose-Response Relationship, Drug , Drug Therapy, Combination , Fluorouracil/therapeutic use , Gene Expression Regulation, Neoplastic/drug effects , Gene Transfer Techniques , Genes, Dominant , Genes, Reporter , HeLa Cells , Humans , Inhibitor of Apoptosis Proteins , Luciferases/metabolism , Mice , Mice, Inbred BALB C , Mice, Nude , Microtubule-Associated Proteins/metabolism , Neoplasm Proteins/metabolism , Random Allocation , Survivin , Telomerase/metabolism , Time Factors , Transfection , Xenograft Model Antitumor Assays
12.
J Gene Med ; 8(10): 1232-42, 2006 Oct.
Article in English | MEDLINE | ID: mdl-16900558

ABSTRACT

BACKGROUND: Conditionally replicating adenoviruses or oncolytic adenoviruses, which can replicate selectively in tumor cells and kill them, represent an innovative class of promising cancer therapeutics. Survivin is the smallest member of the inhibitor of apoptosis (IAP) family, which is transcriptionally upregulated exclusively in most malignant tissues but not in normal tissues. It has been reported that activity of the survivin promoter is tumor-specific, which makes the survivin promoter a good candidate to construct oncolytic viral vectors. METHODS: A luciferase reporter assay was used to determine the activity of the survivin promoter in tumor and normal cells. An oncolytic adenovirus (Ad.SP/E1A) was generated by homologous recombination. The oncolytic efficacy of Ad.SP/E1A was evaluated in cell lines and in a human lung xenograft tumor mouse model. RESULTS: Survivin expression was highly upregulated in tumor cells both at the protein and mRNA level. The luciferase reporter assay showed that survivin promoter activity is tumor-specific. Ad.SP/E1A expressed E1A selectively in tumor cells and induced cytotoxicity, but not in normal cells. Moreover, in animal experiments, intratumoral administration of Ad.SP/E1A significantly suppressed the growth of xenograft tumors. Further investigation showed that Ad.SP/E1A induced cell death by an apoptosis-independent pathway. CONCLUSIONS: Ad.SP/E1A could be a potent therapeutic agent for cancer gene therapy. The investigation of the mechanisms of oncolytic virus-induced cell death in this work will shed light on the construction of more powerful vectors for cancer therapy.


Subject(s)
Adenoviridae/physiology , Carcinoma/therapy , Genetic Therapy/methods , Lung Neoplasms/therapy , Microtubule-Associated Proteins/genetics , Neoplasm Proteins/genetics , Oncolytic Viruses/physiology , Adenoviridae/genetics , Adenovirus E1A Proteins/genetics , Animals , Antineoplastic Agents/therapeutic use , Antineoplastic Agents/toxicity , Carcinoma/mortality , Cell Death , Cells, Cultured , Cloning, Molecular , Female , Gene Expression Regulation, Neoplastic , Genes, Transgenic, Suicide , Genetic Vectors/chemical synthesis , HeLa Cells , Humans , Inhibitor of Apoptosis Proteins , Lung Neoplasms/mortality , Mice , Mice, Inbred BALB C , Mice, Nude , Microtubule-Associated Proteins/metabolism , Neoplasm Proteins/metabolism , Oncolytic Viruses/genetics , Promoter Regions, Genetic , Survivin , Treatment Outcome , Tumor Cells, Cultured , Xenograft Model Antitumor Assays
13.
Cell Signal ; 17(2): 217-29, 2005 Feb.
Article in English | MEDLINE | ID: mdl-15494213

ABSTRACT

The immediate early proteins ICP0 and BICP0 from Herpes virus are promiscuous activators of both viral and cellular genes and play a critical role in virus life cycle. Here we report that ICP0 and BICP0 could induce NF-kappaB translocation from cytoplasm into nucleus and strongly activate NF-kappaB responsive genes specifically. This process was dependent on the RING domain of both proteins. In addition, ICP0 interacted specifically with IkappaBalpha and its activating effect was attenuated by Ubch5A(C85A) and MG132, but not by IkappaBalpha(S32A/S36A). Remarkably, IkappaBalpha was poly-ubiquitinated by both ICP0 and BICP0, in vitro and in vivo. These data indicate that ICP0 and BICP0, functioning as ubiquitin ligases, are bona fide activators of NF-kappaB signaling pathway. Our study identifies a new way ICP0 and BICP0 explore to regulate gene expression.


Subject(s)
I-kappa B Proteins/metabolism , Immediate-Early Proteins/physiology , NF-kappa B/metabolism , Trans-Activators/physiology , Ubiquitin-Protein Ligases/metabolism , Viral Proteins/physiology , Animals , Cattle , Cell Line , Cell Line, Tumor , Cysteine Proteinase Inhibitors/pharmacology , Electrophoretic Mobility Shift Assay , Gene Expression Regulation, Viral , Humans , I-kappa B Kinase , I-kappa B Proteins/genetics , Immediate-Early Proteins/genetics , Immediate-Early Proteins/metabolism , Iron-Binding Proteins/genetics , Iron-Binding Proteins/metabolism , Leupeptins/pharmacology , Mutation , NF-KappaB Inhibitor alpha , Protein Serine-Threonine Kinases/metabolism , Protein Transport/physiology , Simplexvirus , Terminal Repeat Sequences/genetics , Trans-Activators/genetics , Trans-Activators/metabolism , Transcription Factor RelA , Transcription, Genetic/drug effects , Transcription, Genetic/genetics , Transfection , Ubiquitin/metabolism , Ubiquitin-Conjugating Enzymes/genetics , Ubiquitin-Conjugating Enzymes/metabolism , Viral Proteins/genetics , Viral Proteins/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...