Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 15 de 15
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
J Comput Biol ; 30(1): 70-81, 2023 01.
Artículo en Inglés | MEDLINE | ID: mdl-35727687

RESUMEN

Codon optimization of a DNA sequence can significantly increase efficiency of protein expression, reducing the cost to manufacture biologic pharmaceuticals. Although directed methods based on such factors as codon usage bias and GC nucleotide content are often used to optimize protein expression, undirected optimization using machine learning could further improve the process by capitalizing on undiscovered patterns that exist within real DNA sequences. To explore this hypothesis, Chinese hamster DNA sequences were used to train a recurrent neural network (RNN) model of codon optimization. The model was used to generate optimized DNA sequence based on an input amino acid sequence for the example receptor programmed death-ligand 1 and for an example monoclonal antibody. When RNN-optimized sequences were transfected transiently or stably into Chinese hamster ovary cells, the resulting protein expression was as high or higher than that produced by DNA sequences optimized by conventional algorithms.


Asunto(s)
Algoritmos , Redes Neurales de la Computación , Cricetinae , Animales , Células CHO , Cricetulus , Codón/genética
2.
Antiviral Res ; 199: 105271, 2022 03.
Artículo en Inglés | MEDLINE | ID: mdl-35240221

RESUMEN

COVID-19, an infectious disease caused by the SARS-CoV-2 virus, emerged globally in early 2020 and has remained a serious public health issue. To date, although several preventative vaccines have been approved by FDA and EMA, vaccinated individuals increasingly suffer from breakthrough infections. Therapeutic antibodies may provide an alternative strategy to neutralize viral infection and treat serious cases; however, the clinical data and our experiments show that some FDA-approved monoclonal antibodies lose function against COVID-19 variants such as Omicron. Therefore, in this study, we present a novel therapeutic agent, SI-F019, an ACE2-Fc fusion protein whose neutralization efficiency is not compromised, but actually strengthened, by the mutations of dominant variants including Omicron. Comprehensive biophysical analyses revealed the mechanism of increased inhibition to be enhanced interaction of SI-F019 with all the tested spike variants, in contrast to monoclonal antibodies which tended to show weaker binding to some variants. The results imply that SI-F019 may be a broadly useful agent for treatment of COVID-19.


Asunto(s)
Tratamiento Farmacológico de COVID-19 , SARS-CoV-2 , Enzima Convertidora de Angiotensina 2 , Anticuerpos Neutralizantes , Anticuerpos Antivirales/uso terapéutico , Humanos , SARS-CoV-2/genética , Glicoproteína de la Espiga del Coronavirus
3.
Antibodies (Basel) ; 11(1)2022 Jan 13.
Artículo en Inglés | MEDLINE | ID: mdl-35076451

RESUMEN

The epidermal growth factor receptor (EGFR) is a receptor tyrosine kinase whose proliferative effects can contribute to the development of many types of solid tumors when overexpressed. For this reason, EGFR inhibitors such as cetuximab can play an important role in treating cancers such as colorectal cancer and head and neck cancer. Cetuximab is a chimeric monoclonal antibody containing mouse variable regions that bind to EGFR and prevent it from signaling. Although cetuximab has been used clinically since 2004 to successfully control solid tumors, advances in protein engineering have created the opportunity to address some of its shortcomings. In particular, the presence of mouse sequences could contribute to immunogenicity in the form of anti-cetuximab antibodies, and an occupied glycosylation site in FR3 can contribute to hypersensitivity reactions and product heterogeneity. Using simple framework graft or sequence-/structure-guided approaches, cetuximab was humanized onto 11 new frameworks. In addition to increasing humanness and removing the VH glycosylation site, dynamic light scattering revealed increases in stability, and bio-layer interferometry confirmed minimal changes in binding affinity, with patterns emerging across the humanization method. This work demonstrates the potential to improve the biophysical and clinical properties of first-generation protein therapeutics and highlights the advantages of computationally guided engineering.

4.
Molecules ; 21(11)2016 Nov 21.
Artículo en Inglés | MEDLINE | ID: mdl-27879643

RESUMEN

Ribosome-inactivating proteins (RIPs) including ricin, Shiga toxin, and trichosanthin, are RNA N-glycosidases that depurinate a specific adenine residue (A-4324 in rat 28S ribosomal RNA, rRNA) in the conserved α-sarcin/ricin loop (α-SRL) of rRNA. RIPs are grouped into three types according to the number of subunits and the organization of the precursor sequences. RIPs are two-domain proteins, with the active site located in the cleft between the N- and C-terminal domains. It has been found that the basic surface residues of the RIPs promote rapid and specific targeting to the ribosome and a number of RIPs have been shown to interact with the C-terminal regions of the P proteins of the ribosome. At present, the structural basis for the interaction of trichosanthin and ricin-A chain toward P2 peptide is known. This review surveys the structural features of the representative RIPs and discusses how they approach and interact with the ribosome.


Asunto(s)
Modelos Moleculares , Estructura Molecular , Proteínas Inactivadoras de Ribosomas/química , Ribosomas/química , Dominio Catalítico , Unión Proteica , Dominios y Motivos de Interacción de Proteínas , Proteínas Inactivadoras de Ribosomas/clasificación , Proteínas Inactivadoras de Ribosomas/metabolismo , Subunidades Ribosómicas/genética , Subunidades Ribosómicas/metabolismo , Relación Estructura-Actividad , Especificidad por Sustrato
5.
Toxins (Basel) ; 7(1): 156-69, 2015 Jan 19.
Artículo en Inglés | MEDLINE | ID: mdl-25606813

RESUMEN

Ribosome inactivating proteins (RIPs) inhibit protein synthesis by depurinating the large ribosomal RNA and some are found to possess anti-human immunodeficiency virus (HIV) activity. Maize ribosome inactivating protein (RIP) has an internal inactivation loop which is proteolytically removed for full catalytic activity. Here, we showed that the recombinant active maize RIP protected chimeric simian-human immunodeficiency virus (SHIV) 89.6-infected macaque peripheral blood mononuclear cells from lysis ex vivo and transiently reduced plasma viral load in SHIV89.6-infected rhesus macaque model. No evidence of immune dysregulation and other obvious side-effects was found in the treated macaques. Our work demonstrates the potential development of maize RIP as an anti-HIV agent without impeding systemic immune functions.


Asunto(s)
Antirretrovirales/uso terapéutico , Proteínas Inactivadoras de Ribosomas/uso terapéutico , Síndrome de Inmunodeficiencia Adquirida del Simio/tratamiento farmacológico , Virus de la Inmunodeficiencia de los Simios , Carga Viral/efectos de los fármacos , Zea mays , Animales , Leucocitos Mononucleares/efectos de los fármacos , Leucocitos Mononucleares/virología , Macaca mulatta , Masculino , Proteínas Recombinantes/uso terapéutico , Síndrome de Inmunodeficiencia Adquirida del Simio/sangre , Síndrome de Inmunodeficiencia Adquirida del Simio/virología
6.
Curr Opin Struct Biol ; 23(1): 93-9, 2013 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-23265998

RESUMEN

TAL effectors are proteins secreted by bacterial pathogens into plant cells, where they enter the nucleus and activate expression of individual genes. TAL effectors display a modular architecture that includes a central DNA-binding region comprising a tandem array of nearly identical repeats that are almost all 34 residues long. Residue number 13 in each TAL repeat (one of two consecutive polymorphic amino acids that are termed 'repeat variable diresidues', or 'RVDs') specifies the identity of a single base; collectively the sequential repeats and their RVDs dictate the recognition of sequential bases along one of the two DNA strands. The modular architecture of TAL effectors has facilitated their extremely rapid development and application as artificial gene targeting reagents, particularly in the form of site-specific nucleases. Recent crystallographic and biochemical analyses of TAL effectors have established the structural basis of their DNA recognition properties and provide clear directions for future research.


Asunto(s)
Proteínas Bacterianas/química , Proteínas Bacterianas/metabolismo , ADN de Plantas/química , ADN de Plantas/metabolismo , Sitios de Unión , Unión Proteica , Dominios y Motivos de Interacción de Proteínas
7.
PLoS One ; 7(12): e49608, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-23251345

RESUMEN

Ribosome-inactivating proteins (RIPs) inactivate prokaryotic or eukaryotic ribosomes by removing a single adenine in the large ribosomal RNA. Here we show maize RIP (MOD), an atypical RIP with an internal inactivation loop, interacts with the ribosomal stalk protein P2 via Lys158-Lys161, which is located in the N-terminal domain and at the base of its internal loop. Due to subtle differences in the structure of maize RIP, hydrophobic interaction with the 'FGLFD' motif of P2 is not as evidenced in MOD-P2 interaction. As a result, interaction of P2 with MOD was weaker than those with trichosanthin and shiga toxin A as reflected by the dissociation constants (K(D)) of their interaction, which are 1037.50 ± 65.75 µM, 611.70 ± 28.13 µM and 194.84 ± 9.47 µM respectively.Despite MOD and TCS target at the same ribosomal protein P2, MOD was found 48 and 10 folds less potent than trichosanthin in ribosome depurination and cytotoxicity to 293T cells respectively, implicating the strength of interaction between RIPs and ribosomal proteins is important for the biological activity of RIPs. Our work illustrates the flexibility on the docking of RIPs on ribosomal proteins for targeting the sarcin-ricin loop and the importance of protein-protein interaction for ribosome-inactivating activity.


Asunto(s)
Fosfoproteínas/metabolismo , Proteínas Ribosómicas/metabolismo , Proteínas Inactivadoras de Ribosomas/metabolismo , Ribosomas/metabolismo , Zea mays/metabolismo , Animales , Hígado/efectos de los fármacos , Hígado/metabolismo , Fosfoproteínas/genética , ARN Ribosómico/metabolismo , Ratas , Proteínas Ribosómicas/genética , Proteínas Inactivadoras de Ribosomas/genética , Proteínas Inactivadoras de Ribosomas/farmacología , Ribosomas/efectos de los fármacos , Toxina Shiga/genética , Toxina Shiga/metabolismo , Tricosantina/genética , Tricosantina/metabolismo , Zea mays/genética
8.
Science ; 335(6069): 716-9, 2012 Feb 10.
Artículo en Inglés | MEDLINE | ID: mdl-22223736

RESUMEN

DNA recognition by TAL effectors is mediated by tandem repeats, each 33 to 35 residues in length, that specify nucleotides via unique repeat-variable diresidues (RVDs). The crystal structure of PthXo1 bound to its DNA target was determined by high-throughput computational structure prediction and validated by heavy-atom derivatization. Each repeat forms a left-handed, two-helix bundle that presents an RVD-containing loop to the DNA. The repeats self-associate to form a right-handed superhelix wrapped around the DNA major groove. The first RVD residue forms a stabilizing contact with the protein backbone, while the second makes a base-specific contact to the DNA sense strand. Two degenerate amino-terminal repeats also interact with the DNA. Containing several RVDs and noncanonical associations, the structure illustrates the basis of TAL effector-DNA recognition.


Asunto(s)
Proteínas Bacterianas/química , Proteínas Bacterianas/metabolismo , ADN de Plantas/química , ADN de Plantas/metabolismo , Factores de Virulencia/química , Factores de Virulencia/metabolismo , Xanthomonas/química , Secuencia de Aminoácidos , Sitios de Unión , Fenómenos Químicos , Cristalografía por Rayos X , Proteínas de Unión al ADN/química , Proteínas de Unión al ADN/metabolismo , Ensayos Analíticos de Alto Rendimiento , Enlace de Hidrógeno , Modelos Moleculares , Datos de Secuencia Molecular , Unión Proteica , Conformación Proteica , Pliegue de Proteína , Estructura Secundaria de Proteína , Secuencias Repetitivas de Aminoácido , Xanthomonas/patogenicidad
9.
Proc Natl Acad Sci U S A ; 108(32): 13077-82, 2011 Aug 09.
Artículo en Inglés | MEDLINE | ID: mdl-21784983

RESUMEN

Homing endonucleases mobilize their own genes by generating double-strand breaks at individual target sites within potential host DNA. Because of their high specificity, these proteins are used for "genome editing" in higher eukaryotes. However, alteration of homing endonuclease specificity is quite challenging. Here we describe the identification and phylogenetic analysis of over 200 naturally occurring LAGLIDADG homing endonucleases (LHEs). Biochemical and structural characterization of endonucleases from one clade within the phylogenetic tree demonstrates strong conservation of protein structure contrasted against highly diverged DNA target sites and indicates that a significant fraction of these proteins are sufficiently stable and active to serve as engineering scaffolds. This information was exploited to create a targeting enzyme to disrupt the endogenous monoamine oxidase B gene in human cells. The ubiquitous presence and diversity of LHEs described in this study may facilitate the creation of many tailored nucleases for genome editing.


Asunto(s)
Endonucleasas/metabolismo , Marcación de Gen , Secuencia de Aminoácidos , Secuencia de Bases , Endonucleasas/química , Humanos , Modelos Moleculares , Datos de Secuencia Molecular , Monoaminooxidasa/genética , Mutagénesis/genética , Unión Proteica , Ingeniería de Proteínas , Especificidad por Sustrato
10.
Structure ; 18(10): 1321-31, 2010 Oct 13.
Artículo en Inglés | MEDLINE | ID: mdl-20800503

RESUMEN

The GIY-YIG endonuclease family comprises hundreds of diverse proteins and a multitude of functions; none have been visualized bound to DNA. The structure of the GIY-YIG restriction endonuclease R.Eco29kI has been solved both alone and bound to its target site. The protein displays a domain-swapped homodimeric structure with several extended surface loops encircling the DNA. Only three side chains from each protein subunit contact DNA bases, two directly and one via a bridging solvent molecule. Both tyrosine residues within the GIY-YIG motif are positioned in the catalytic center near a putative nucleophilic water; the remainder of the active site resembles the HNH endonuclease family. The structure illustrates how the GIY-YIG scaffold has been adapted for the highly specific recognition of a DNA restriction site, in contrast to nonspecific DNA cleavage by GIY-YIG domains in homing endonucleases or structure-specific cleavage by DNA repair enzymes such as UvrC.


Asunto(s)
ADN/química , Desoxirribonucleasas de Localización Especificada Tipo II/química , Pliegue de Proteína , Estructura Terciaria de Proteína , Secuencia de Aminoácidos , Secuencia de Bases , Sitios de Unión/genética , Cristalización , Cristalografía por Rayos X , ADN/metabolismo , Desoxirribonucleasas de Localización Especificada Tipo II/genética , Desoxirribonucleasas de Localización Especificada Tipo II/metabolismo , Modelos Moleculares , Datos de Secuencia Molecular , Mutación Missense , Unión Proteica , Multimerización de Proteína , Homología de Secuencia de Aminoácido
11.
Nucleic Acids Res ; 38(19): 6803-12, 2010 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-20558598

RESUMEN

Maize ribosome-inactivating protein (RIP) is a plant toxin that inactivates eukaryotic ribosomes by depurinating a specific adenine residue at the α-sarcin/ricin loop of 28S rRNA. Maize RIP is first produced as a proenzyme with a 25-amino acid internal inactivation region on the protein surface. During germination, proteolytic removal of this internal inactivation region generates the active heterodimeric maize RIP with full N-glycosidase activity. This naturally occurring switch-on mechanism provides an opportunity for targeting the cytotoxin to pathogen-infected cells. Here, we report the addition of HIV-1 protease recognition sequences to the internal inactivation region and the activation of the maize RIP variants by HIV-1 protease in vitro and in HIV-infected cells. Among the variants generated, two were cleaved efficiently by HIV-1 protease. The HIV-1 protease-activated variants showed enhanced N-glycosidase activity in vivo as compared to their un-activated counterparts. They also possessed potent inhibitory effect on p24 antigen production in human T cells infected by two HIV-1 strains. This switch-on strategy for activating the enzymatic activity of maize RIP in target cells provides a platform for combating pathogens with a specific protease.


Asunto(s)
Fármacos Anti-VIH/química , Proteínas Inactivadoras de Ribosomas/genética , Proteínas Inactivadoras de Ribosomas/farmacología , Secuencia de Aminoácidos , Fármacos Anti-VIH/metabolismo , Fármacos Anti-VIH/farmacología , Línea Celular , Activación Enzimática , Proteasa del VIH/metabolismo , VIH-1/fisiología , Humanos , Datos de Secuencia Molecular , Proteínas Recombinantes de Fusión/química , Proteínas Recombinantes de Fusión/metabolismo , Proteínas Inactivadoras de Ribosomas/metabolismo , Linfocitos T/virología , Zea mays/enzimología , Productos del Gen tat del Virus de la Inmunodeficiencia Humana/genética
12.
J Mol Biol ; 395(5): 897-907, 2010 Feb 05.
Artículo en Inglés | MEDLINE | ID: mdl-19900464

RESUMEN

Ribosome-inactivating proteins (RIPs) are N-glycosidases that depurinate a specific adenine residue in the conserved sarcin/ricin loop of ribosomal RNA. This modification renders the ribosome unable to bind the elongation factors, thereby inhibiting the protein synthesis. Maize RIP, a type III RIP, is unique compared to the other type I and type II RIPs because it is synthesized as a precursor with a 25-residue internal inactivation region, which is removed in order to activate the protein. In this study, we describe the first solution structure of this type of RIP, a 28-kDa active mutant of maize RIP (MOD). The overall protein structure of MOD is comparable to those of the other type I RIPs and the A-chain of type II RIPs but shows significant differences in specific regions, including (1) shorter beta6 and alphaB segments, probably for accommodating easier substrate binding, and (2) an alpha-helix instead of an antiparallel beta-sheet in the C-terminal domain, which has been reported to be involved in binding ribosomal protein P2 in some RIPs. Furthermore, NMR chemical shift perturbation experiments revealed that the P2 binding site on MOD is located at the N-terminal domain near the internal inactivation region. This relocation of the P2 binding site can be rationalized by concerted changes in the electrostatic surface potential and 3D structures on the MOD protein and provides vital clues about the underlying molecular mechanism of this unique type of RIP.


Asunto(s)
Fosfoproteínas/química , Proteínas de Plantas/química , Proteínas Ribosómicas/química , Proteínas Inactivadoras de Ribosomas/química , Zea mays/química , Secuencia de Aminoácidos , Sitios de Unión , Cristalografía por Rayos X , Modelos Moleculares , Datos de Secuencia Molecular , Mutación , Resonancia Magnética Nuclear Biomolecular , Fosfoproteínas/genética , Fosfoproteínas/metabolismo , Proteínas de Plantas/genética , Proteínas de Plantas/metabolismo , Conformación Proteica , Dominios y Motivos de Interacción de Proteínas , Proteínas Recombinantes/química , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Proteínas Ribosómicas/genética , Proteínas Ribosómicas/metabolismo , Proteínas Inactivadoras de Ribosomas/genética , Proteínas Inactivadoras de Ribosomas/metabolismo , Homología de Secuencia de Aminoácido , Electricidad Estática , Zea mays/genética , Zea mays/metabolismo
13.
Nucleic Acids Res ; 37(2): 602-10, 2009 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-19073700

RESUMEN

Ribosome-inactivating proteins (RIPs) inhibit protein synthesis by enzymatically depurinating a specific adenine residue at the sarcin-ricin loop of the 28S rRNA, which thereby prevents the binding of elongation factors to the GTPase activation centre of the ribosome. Here, we present the 2.2 A crystal structure of trichosanthin (TCS) complexed to the peptide SDDDMGFGLFD, which corresponds to the conserved C-terminal elongation factor binding domain of the ribosomal P protein. The N-terminal region of this peptide interacts with Lys173, Arg174 and Lys177 in TCS, while the C-terminal region is inserted into a hydrophobic pocket. The interaction with the P protein contributes to the ribosome-inactivating activity of TCS. This 11-mer C-terminal P peptide can be docked with selected important plant and bacterial RIPs, indicating that a similar interaction may also occur with other RIPs.


Asunto(s)
Fosfoproteínas/química , Proteínas Ribosómicas/química , Tricosantina/química , Secuencia de Aminoácidos , Modelos Moleculares , Datos de Secuencia Molecular , Péptidos/química , Estructura Terciaria de Proteína , Homología de Secuencia de Aminoácido
14.
Nucleic Acids Res ; 35(18): 6259-67, 2007.
Artículo en Inglés | MEDLINE | ID: mdl-17855394

RESUMEN

Maize ribosome-inactivating protein is classified as a class III or an atypical RNA N-glycosidase. It is synthesized as an inactive precursor with a 25-amino acid internal inactivation region, which is removed in the active form. As the first structural example of this class of proteins, crystals of the precursor and the active form were diffracted to 2.4 and 2.5 A, respectively. The two proteins are similar, with main chain root mean square deviation (RMSD) of 0.519. In the precursor, the inactivation region is found on the protein surface and consists of a flexible loop followed by a long alpha-helix. This region diminished both the interaction with ribosome and cytotoxicity, but not cellular uptake. Like bacterial ribosome-inactivating proteins, maize ribosome-inactivating protein does not have a back-up glutamate in the active site, which helps the protein to retain some activity if the catalytic glutamate is mutated. The structure reveals that the active site is too small to accommodate two glutamate residues. Our structure suggests that maize ribosome-inactivating protein may represent an intermediate product in the evolution of ribosome-inactivating proteins.


Asunto(s)
Ácido Glutámico/química , Proteínas de Plantas/química , Inhibidores de la Síntesis de la Proteína/química , Proteínas Inactivadoras de Ribosomas/química , Animales , Sitios de Unión , Línea Celular Tumoral , Cristalografía por Rayos X , Ácido Glutámico/genética , Humanos , Modelos Moleculares , Mutación , Proteínas de Plantas/genética , Proteínas de Plantas/farmacología , Inhibidores de la Síntesis de la Proteína/metabolismo , Inhibidores de la Síntesis de la Proteína/farmacología , Ratas , Proteínas Ribosómicas/metabolismo , Proteínas Inactivadoras de Ribosomas/genética , Proteínas Inactivadoras de Ribosomas/farmacología , Ribosomas/química , Relación Estructura-Actividad
15.
Biomol NMR Assign ; 1(2): 187-9, 2007 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-19636861

RESUMEN

We report the full resonance assignments of MOD, which is an active mutant of maize ribosome-inactivating protein (mRIP). mRIP is a unique RIP which is synthesized as an inactive precursor and processed by removal of an internal inactivation region to yield an active form.


Asunto(s)
Espectroscopía de Resonancia Magnética/métodos , Proteínas Inactivadoras de Ribosomas/química , Zea mays/genética , Zea mays/metabolismo , Secuencia de Aminoácidos , Isótopos de Carbono/química , Peso Molecular , Mutación , Isótopos de Nitrógeno/química , Estructura Terciaria de Proteína , Protones
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...