Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 378
Filtrar
1.
ACS Infect Dis ; 10(5): 1839-1855, 2024 May 10.
Artículo en Inglés | MEDLINE | ID: mdl-38725407

RESUMEN

Multidrug resistance against conventional antibiotics has dramatically increased the difficulty of treatment and accelerated the need for novel antibacterial agents. The peptide Tat (47-57) is derived from the transactivating transcriptional activator of human immunodeficiency virus 1, which is well-known as a cell-penetrating peptide in mammalian cells. However, it is also reported that the Tat peptide (47-57) has antifungal activity. In this study, a series of membrane-active hydrocarbon-stapled α-helical amphiphilic peptides were synthesized and evaluated as antibacterial agents against Gram-positive and Gram-negative bacteria, including multidrug-resistant strains. The impact of hydrocarbon staple, the position of aromatic amino acid residue in the hydrophobic face, the various types of aromatic amino acids, and the hydrophobicity on bioactivity were also investigated and discussed in this study. Among those synthesized peptides, analogues P3 and P10 bearing a l-2-naphthylalanine (Φ) residue at the first position and a Tyr residue at the eighth position demonstrated the highest antimicrobial activity and negligible hemolytic toxicity. Notably, P3 and P10 showed obviously enhanced antimicrobial activity against multidrug-resistant bacteria, low drug resistance, high cell selectivity, extended half-life in plasma, and excellent performance against biofilm. The antibacterial mechanisms of P3 and P10 were also preliminarily investigated in this effort. In conclusion, P3 and P10 are promising antimicrobial alternatives for the treatment of the antimicrobial-resistance crisis.


Asunto(s)
Antibacterianos , Pruebas de Sensibilidad Microbiana , Antibacterianos/farmacología , Antibacterianos/química , Humanos , Productos del Gen tat del Virus de la Inmunodeficiencia Humana/química , Bacterias Gramnegativas/efectos de los fármacos , Farmacorresistencia Bacteriana Múltiple/efectos de los fármacos , Bacterias Grampositivas/efectos de los fármacos , Interacciones Hidrofóbicas e Hidrofílicas , Hidrocarburos/química , Hidrocarburos/farmacología , Hemólisis/efectos de los fármacos , Conformación Proteica en Hélice alfa
2.
Int J Mol Sci ; 24(11)2023 May 23.
Artículo en Inglés | MEDLINE | ID: mdl-37298089

RESUMEN

Human immunodeficiency virus-1 (HIV-1) transactivator (Tat)-mediated transcription is essential for HIV-1 replication. It is determined by the interaction between Tat and transactivation response (TAR) RNA, a highly conserved process representing a prominent therapeutic target against HIV-1 replication. However, owing to the limitations of current high-throughput screening (HTS) assays, no drug that disrupts the Tat-TAR RNA interaction has been uncovered yet. We designed a homogenous (mix-and-read) time-resolved fluorescence resonance energy transfer (TR-FRET) assay using europium cryptate as a fluorescence donor. It was optimized by evaluating different probing systems for Tat-derived peptides or TAR RNA. The specificity of the optimal assay was validated by mutants of the Tat-derived peptides and TAR RNA fragment, individually and by competitive inhibition with known TAR RNA-binding peptides. The assay generated a constant Tat-TAR RNA interaction signal, discriminating the compounds that disrupted the interaction. Combined with a functional assay, the TR-FRET assay identified two small molecules (460-G06 and 463-H08) capable of inhibiting Tat activity and HIV-1 infection from a large-scale compound library. The simplicity, ease of operation, and rapidity of our assay render it suitable for HTS to identify Tat-TAR RNA interaction inhibitors. The identified compounds may also act as potent molecular scaffolds for developing a new HIV-1 drug class.


Asunto(s)
VIH-1 , Productos del Gen tat del Virus de la Inmunodeficiencia Humana , Humanos , Productos del Gen tat del Virus de la Inmunodeficiencia Humana/genética , Productos del Gen tat del Virus de la Inmunodeficiencia Humana/química , VIH-1/fisiología , Transferencia Resonante de Energía de Fluorescencia , Transactivadores , ARN Viral/genética
3.
ACS Nano ; 17(13): 12247-12265, 2023 07 11.
Artículo en Inglés | MEDLINE | ID: mdl-37350353

RESUMEN

Cells penetrating molecules in living systems hold promise of capturing and eliminating threats and damage that can plan intracellular fate promptly. However, it remains challenging to construct cell penetration systems that are physiologically stable with predictable self-assembly behavior and well-defined mechanisms. In this study, we develop a core-shell nanoparticle using a hyaluronic acid (HA)-coated protein transduction domain (PTD) derived from the human immunodeficiency virus (HIV). This nanoparticle can encapsulate pathogens, transporting the PTD into macrophages via lipid rafts. PTD forms hydrogen bonds with the components of the membrane through TAT, which has a high density of positive charges and reduces the degree of membrane order through Tryptophan (Trp)-zipper binding to the acyl tails of phospholipid molecules. HA-encapsulated PTD increases the resistance to trypsin and proteinase K, thereby penetrating macrophages and eliminating intracellular infections. Interestingly, the nonagglutination mechanism of PTD against pathogens ensures the safe operation of the cellular system. Importantly, PTD can activate the critical pathway of antiferroptosis in macrophages against pathogen infection. The nanoparticles developed in this study demonstrate safety and efficacy against Gram-negative and Gram-positive pathogens in three animal models. Overall, this work highlights the effectiveness of the PTD nanoparticle in encapsulating pathogens and provides a paradigm for transduction systems-anti-intracellular infection therapy.


Asunto(s)
Ferroptosis , Productos del Gen tat del Virus de la Inmunodeficiencia Humana , Animales , Humanos , Productos del Gen tat del Virus de la Inmunodeficiencia Humana/química , Productos del Gen tat del Virus de la Inmunodeficiencia Humana/genética , Productos del Gen tat del Virus de la Inmunodeficiencia Humana/metabolismo , Triptófano , Transporte Biológico , Macrófagos/metabolismo , Transducción Genética , Proteínas Recombinantes de Fusión/metabolismo
4.
Nature ; 617(7962): 835-841, 2023 05.
Artículo en Inglés | MEDLINE | ID: mdl-37198487

RESUMEN

Cellular processes are the product of interactions between biomolecules, which associate to form biologically active complexes1. These interactions are mediated by intermolecular contacts, which if disrupted, lead to alterations in cell physiology. Nevertheless, the formation of intermolecular contacts nearly universally requires changes in the conformations of the interacting biomolecules. As a result, binding affinity and cellular activity crucially depend both on the strength of the contacts and on the inherent propensities to form binding-competent conformational states2,3. Thus, conformational penalties are ubiquitous in biology and must be known in order to quantitatively model binding energetics for protein and nucleic acid interactions4,5. However, conceptual and technological limitations have hindered our ability to dissect and quantitatively measure how conformational propensities affect cellular activity. Here we systematically altered and determined the propensities for forming the protein-bound conformation of HIV-1 TAR RNA. These propensities quantitatively predicted the binding affinities of TAR to the RNA-binding region of the Tat protein and predicted the extent of HIV-1 Tat-dependent transactivation in cells. Our results establish the role of ensemble-based conformational propensities in cellular activity and reveal an example of a cellular process driven by an exceptionally rare and short-lived RNA conformational state.


Asunto(s)
Duplicado del Terminal Largo de VIH , VIH-1 , Conformación de Ácido Nucleico , ARN Viral , Activación Transcripcional , Productos del Gen tat del Virus de la Inmunodeficiencia Humana , Duplicado del Terminal Largo de VIH/genética , ARN Viral/química , ARN Viral/genética , ARN Viral/metabolismo , Productos del Gen tat del Virus de la Inmunodeficiencia Humana/química , Productos del Gen tat del Virus de la Inmunodeficiencia Humana/metabolismo , VIH-1/genética , VIH-1/metabolismo
5.
J Mol Biol ; 435(2): 167880, 2023 01 30.
Artículo en Inglés | MEDLINE | ID: mdl-36370804

RESUMEN

Human immunodeficiency virus type 1 (HIV-1) trans-activator of transcription (Tat) is a small, intrinsically disordered basic protein that plays diverse roles in the HIV-1 replication cycle, including promotion of efficient viral RNA transcription. Tat is released by infected cells and subsequently absorbed by healthy cells, thereby contributing to HIV-1 pathogenesis including HIV-associated neurocognitive disorder. It has been shown that, in HIV-1-infected primary CD4 T-cells, Tat accumulates at the plasma membrane (PM) for secretion, a mechanism mediated by phosphatidylinositol 4,5-bisphosphate (PI(4,5)P2). However, the structural basis for Tat interaction with the PM and thereby secretion is lacking. Herein, we employed NMR and biophysical methods to characterize Tat86 (86 amino acids) interactions with PI(4,5)P2 and lipid nanodiscs (NDs). Our data revealed that Arg49, Lys50 and Lys51 (RKK motif) constitute the PI(4,5)P2 binding site, that Tat86 interaction with lipid NDs is dependent on PI(4,5)P2 and phosphatidylserine (PS), and that the arginine-rich motif (RRQRRR) preferentially interacts with PS. Furthermore, we show that Trp11, previously implicated in Tat secretion, penetrates deeply in the membrane; substitution of Trp11 severely reduced Tat86 interaction with membranes. Deletion of the entire highly basic region and Trp11 completely abolished Tat86 binding to lipid NDs. Our data support a mechanism by which HIV-1 Tat secretion from the PM is mediated by a tripartite signal consisting of binding of the RKK motif to PI(4,5)P2, arginine-rich motif to PS, and penetration of Trp11 in the membrane. Altogether, these findings provide new insights into the molecular requirements for Tat binding to membranes during secretion.


Asunto(s)
Infecciones por VIH , VIH-1 , Productos del Gen tat del Virus de la Inmunodeficiencia Humana , Humanos , Arginina/metabolismo , Membrana Celular/metabolismo , Infecciones por VIH/metabolismo , VIH-1/genética , VIH-1/metabolismo , Lípidos , Unión Proteica , ARN Viral/metabolismo , Productos del Gen tat del Virus de la Inmunodeficiencia Humana/química , Productos del Gen tat del Virus de la Inmunodeficiencia Humana/metabolismo , Transporte de Proteínas
6.
Fluids Barriers CNS ; 19(1): 22, 2022 Mar 17.
Artículo en Inglés | MEDLINE | ID: mdl-35300705

RESUMEN

BACKGROUND: Neurodegenerative diseases (NDs) are an accelerating global health problem. Nevertheless, the stronghold of the brain- the blood-brain barrier (BBB) prevents drug penetrance and dwindles effective treatments. Therefore, it is crucial to identify Trojan horse-like drug carriers that can effectively cross the blood-brain barrier and reach the brain tissue. We have previously developed polyunsaturated fatty acids (PUFA)-based nanostructured lipid carriers (NLC), namely DHAH-NLC. These carriers are modulated with BBB-permeating compounds such as chitosan (CS) and trans-activating transcriptional activator (TAT) from HIV-1 that can entrap neurotrophic factors (NTF) serving as nanocarriers for NDs treatment. Moreover, microglia are suggested as a key causative factor of the undergoing neuroinflammation of NDs. In this work, we used in vitro models to investigate whether DHAH-NLCs can enter the brain via the BBB and investigate the therapeutic effect of NTF-containing DHAH-NLC and DHAH-NLC itself on lipopolysaccharide-challenged microglia. METHODS: We employed human induced pluripotent stem cell-derived brain microvascular endothelial cells (BMECs) to capitalize on the in vivo-like TEER of this BBB model and quantitatively assessed the permeability of DHAH-NLCs. We also used the HMC3 microglia cell line to assess the therapeutic effect of NTF-containing DHAH-NLC upon LPS challenge. RESULTS: TAT-functionalized DHAH-NLCs successfully crossed the in vitro BBB model, which exhibited high transendothelial electrical resistance (TEER) values (≈3000 Ω*cm2). Specifically, the TAT-functionalized DHAH-NLCs showed a permeability of up to 0.4% of the dose. Furthermore, using human microglia (HMC3), we demonstrate that DHAH-NLCs successfully counteracted the inflammatory response in our cultures after LPS challenge. Moreover, the encapsulation of glial cell-derived neurotrophic factor (GNDF)-containing DHAH-NLCs (DHAH-NLC-GNDF) activated the Nrf2/HO-1 pathway, suggesting the triggering of the endogenous anti-oxidative system present in microglia. CONCLUSIONS: Overall, this work shows that the TAT-functionalized DHAH-NLCs can cross the BBB, modulate immune responses, and serve as cargo carriers for growth factors; thus, constituting an attractive and promising novel drug delivery approach for the transport of therapeutics through the BBB into the brain.


Asunto(s)
Barrera Hematoencefálica , Nanopartículas , Factores de Crecimiento Nervioso , Enfermedades Neurodegenerativas , Productos del Gen tat del Virus de la Inmunodeficiencia Humana , Barrera Hematoencefálica/metabolismo , Ácidos Docosahexaenoicos/química , Humanos , Células Madre Pluripotentes Inducidas/metabolismo , Liposomas , Microglía/metabolismo , Factores de Crecimiento Nervioso/administración & dosificación , Enfermedades Neurodegenerativas/tratamiento farmacológico , Productos del Gen tat del Virus de la Inmunodeficiencia Humana/química
7.
J Virol ; 96(1): e0150521, 2022 01 12.
Artículo en Inglés | MEDLINE | ID: mdl-34613791

RESUMEN

During evolution, viruses had to adapt to an increasingly complex environment of eukaryotic cells. Viral proteins that need to enter the cell nucleus or associate with nucleoli possess nuclear localization signals (NLSs) and nucleolar localization signals (NoLSs) for nuclear and nucleolar accumulation, respectively. As viral proteins are relatively small, acquisition of novel sequences seems to be a more complicated task for viruses than for eukaryotes. Here, we carried out a comprehensive analysis of the basic domain (BD) of HIV-1 Tat to show how viral proteins might evolve with NLSs and NoLSs without an increase in protein size. The HIV-1 Tat BD is involved in several functions, the most important being the transactivation of viral transcription. The BD also functions as an NLS, although it is substantially longer than a typical NLS. It seems that different regions in the BD could function as NLSs due to its enrichment with positively charged amino acids. Additionally, the high positive net charge inevitably causes the BD to function as an NoLS through a charge-specific mechanism. The integration of NLSs and NoLSs into functional domains enriched with positively charged amino acids might be a mechanism that allows the condensation of different functional sequences in small protein regions and, as a result, reduces protein size, influencing the origin and evolution of NLSs and NoLSs in viruses. IMPORTANCE Here, we investigated the molecular mechanism of nuclear localization signal (NLS) and nucleolar localization signal (NoLS) integration into the basic domain of HIV-1 Tat (49RKKRRQRRR57) and found that these two supplementary functions (i.e., function of NLS and function of NoLS) are embedded in the basic domain amino acid sequence. The integration of NLSs and NoLSs into functional domains of viral proteins enriched with positively charged amino acids is a mechanism that allows the concentration of different functions within small protein regions. Integration of NLS and NoLS into functional protein domains might have influenced the viral evolution, as this could prevent an increase in the protein size.


Asunto(s)
Regulación Viral de la Expresión Génica , Infecciones por VIH/virología , VIH-1/fisiología , Señales de Localización Nuclear , Dominios y Motivos de Interacción de Proteínas , Productos del Gen tat del Virus de la Inmunodeficiencia Humana/química , Productos del Gen tat del Virus de la Inmunodeficiencia Humana/metabolismo , Secuencia de Aminoácidos , Sitios de Unión , Nucléolo Celular/metabolismo , Núcleo Celular/metabolismo , Secuencia de Consenso , Evolución Molecular , Interacciones Huésped-Patógeno , Modelos Moleculares , Unión Proteica , Transporte de Proteínas , Relación Estructura-Actividad , Proteínas Virales/metabolismo , Productos del Gen tat del Virus de la Inmunodeficiencia Humana/genética
8.
Molecules ; 26(24)2021 Dec 10.
Artículo en Inglés | MEDLINE | ID: mdl-34946571

RESUMEN

HIV-1 transactivating factor Tat is released by infected cells. Extracellular Tat homodimerizes and engages several receptors, including integrins, vascular endothelial growth factor receptor 2 (VEGFR2) and heparan sulfate proteoglycan (HSPG) syndecan-1 expressed on various cells. By means of experimental cell models recapitulating the processes of lymphocyte trans-endothelial migration, here, we demonstrate that upon association with syndecan-1 expressed on lymphocytes, Tat triggers simultaneously the in cis activation of lymphocytes themselves and the in trans activation of endothelial cells (ECs). This "two-way" activation eventually induces lymphocyte adhesion and spreading onto the substrate and vascular endothelial (VE)-cadherin reorganization at the EC junctions, with consequent endothelial permeabilization, leading to an increased extravasation of Tat-presenting lymphocytes. By means of a panel of biochemical activation assays and specific synthetic inhibitors, we demonstrate that during the above-mentioned processes, syndecan-1, integrins, FAK, src and ERK1/2 engagement and activation are needed in the lymphocytes, while VEGFR2, integrin, src and ERK1/2 are needed in the endothelium. In conclusion, the Tat/syndecan-1 complex plays a central role in orchestrating the setup of the various in cis and in trans multimeric complexes at the EC/lymphocyte interface. Thus, by means of computational molecular modelling, docking and dynamics, we also provide a characterization at an atomic level of the binding modes of the Tat/heparin interaction, with heparin herein used as a structural analogue of the heparan sulfate chains of syndecan-1.


Asunto(s)
Endotelio/metabolismo , Proteoglicanos de Heparán Sulfato/metabolismo , Linfocitos/metabolismo , Productos del Gen tat del Virus de la Inmunodeficiencia Humana/metabolismo , Adhesión Celular , Movimiento Celular , Endotelio/química , Proteoglicanos de Heparán Sulfato/química , Humanos , Linfocitos/química , Modelos Moleculares , Estructura Molecular , Estereoisomerismo , Células Tumorales Cultivadas , Productos del Gen tat del Virus de la Inmunodeficiencia Humana/química
9.
Viruses ; 13(10)2021 10 11.
Artículo en Inglés | MEDLINE | ID: mdl-34696473

RESUMEN

Mother-to-child transmission (MTCT) of HIV-1 may occur during pregnancy, labor, and breastfeeding; however, the molecular mechanism of MTCT of virus remains poorly understood. Infant tonsil mucosal epithelium may sequester HIV-1, serving as a transient reservoir, and may play a critical role in MTCT. Innate immune proteins human beta-defensins 2 (hBD-2) and -3 may inactivate intravesicular virions. To establish delivery of hBD-2 and -3 into vesicles containing HIV-1, we tagged hBDs with the protein transduction domain (PTD) of HIV-1 Tat, which facilitates an efficient translocation of proteins across cell membranes. Our new findings showed that hBD-2 and -3 proteins tagged with PTD efficiently penetrated polarized tonsil epithelial cells by endocytosis and direct penetration. PTD-initiated internalization of hBD-2 and -3 proteins into epithelial cells led to their subsequent penetration of multivesicular bodies (MVB) and vacuoles containing HIV-1. Furthermore, PTD played a role in the fusion of vesicles containing HIV-1 with lysosomes, where virus was inactivated. PTD-initiated internalization of hBD-2 and -3 proteins into ex vivo tonsil tissue explants reduced the spread of virus from epithelial cells to CD4+ T lymphocytes, CD68+ macrophages, and CD1c+ dendritic cells, suggesting that this approach may serve as an antiviral strategy for inactivating intraepithelial HIV-1 and reducing viral MTCT.


Asunto(s)
Polaridad Celular/fisiología , Células Epiteliales/virología , VIH-1/fisiología , Tonsila Palatina/virología , beta-Defensinas/metabolismo , Productos del Gen tat del Virus de la Inmunodeficiencia Humana/metabolismo , Linfocitos T CD4-Positivos , Endocitosis , Epitelio , Infecciones por VIH , Humanos , Transmisión Vertical de Enfermedad Infecciosa , Macrófagos/virología , Membrana Mucosa/virología , Dominios Proteicos , beta-Defensinas/genética , Productos del Gen tat del Virus de la Inmunodeficiencia Humana/química
10.
PLoS One ; 16(9): e0254468, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34473728

RESUMEN

Cell-penetrating peptides (CPPs) are capable of transporting molecules to which they are tethered across cellular membranes. Unsurprisingly, CPPs have attracted attention for their potential drug delivery applications, but several technical hurdles remain to be overcome. Chief among them is the so-called 'endosomal escape problem,' i.e. the propensity of CPP-cargo molecules to be endocytosed but remain entrapped in endosomes rather than reaching the cytosol. Previously, a CPP fused to calmodulin that bound calmodulin binding site-containing cargos was shown to efficiently deliver cargos to the cytoplasm, effectively overcoming the endosomal escape problem. The CPP-adaptor, "TAT-CaM," evinces delivery at nM concentrations and more rapidly than we had previously been able to measure. To better understand the kinetics and mechanism of CPP-adaptor-mediated cargo delivery, a real-time cell penetrating assay was developed in which a flow chamber containing cultured cells was installed on the stage of a confocal microscope to allow for observation ab initio. Also examined in this study was an improved CPP-adaptor that utilizes naked mole rat (Heterocephalus glaber) calmodulin in place of human and results in superior internalization, likely due to its lesser net negative charge. Adaptor-cargo complexes were delivered into the flow chamber and fluorescence intensity in the midpoint of baby hamster kidney cells was measured as a function of time. Delivery of 400 nM cargo was observed within seven minutes and fluorescence continued to increase linearly as a function of time. Cargo-only control experiments showed that the minimal uptake which occurred independently of the CPP-adaptor resulted in punctate localization consistent with endosomal entrapment. A distance analysis was performed for cell-penetration experiments in which CPP-adaptor-delivered cargo showing wider dispersions throughout cells as compared to an analogous covalently-bound CPP-cargo. Small molecule endocytosis inhibitors did not have significant effects upon delivery. The real-time assay is an improvement upon static endpoint assays and should be informative in a broad array of applications.


Asunto(s)
Calmodulina/metabolismo , Péptidos de Penetración Celular/química , Sistemas de Liberación de Medicamentos/métodos , Endosomas/metabolismo , Proteínas de Unión a Maltosa/metabolismo , Bibliotecas de Moléculas Pequeñas/administración & dosificación , Productos del Gen tat del Virus de la Inmunodeficiencia Humana/metabolismo , Animales , Bioensayo/métodos , Calmodulina/química , Línea Celular , Cricetinae , Citosol/metabolismo , Sistemas de Liberación de Medicamentos/instrumentación , Endosomas/efectos de los fármacos , Humanos , Microscopía Fluorescente/métodos , Ratas , Bibliotecas de Moléculas Pequeñas/química , Productos del Gen tat del Virus de la Inmunodeficiencia Humana/química
11.
Front Immunol ; 12: 715644, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34421920

RESUMEN

Tat/Rev Induced Limiting Dilution Assay (TILDA) is instrumental in estimating the size of latent reservoirs of HIV-1. Here, we report an optimized TILDA containing a broader detection range compared to the reported methods and high sensitivity. Giving priority to sequence conservation, we positioned the two forward primers and the probe in exon-1 of HIV-1. The reverse primers are positioned in highly conserved regions of exon-7. The optimized TILDA detected eight molecular clones belonging to five major genetic subtypes of HIV-1 with a comparable detection sensitivity. Using the optimized assay, we show that only a minor proportion of CD4+ T cells of primary clinical samples can spontaneously generate multiply spliced viral transcripts. A significantly larger proportion of the cells produced viral transcripts following activation. The optimized TILDA is suitable to characterize HIV-1 latent reservoirs and the therapeutic strategies intended to target the reservoir size.


Asunto(s)
Infecciones por VIH/diagnóstico , Infecciones por VIH/virología , VIH-1/fisiología , Técnicas de Amplificación de Ácido Nucleico , Carga Viral , Latencia del Virus , Productos del Gen rev del Virus de la Inmunodeficiencia Humana/genética , Productos del Gen tat del Virus de la Inmunodeficiencia Humana/genética , Línea Celular , Secuencia Conservada , Variación Genética , Infecciones por VIH/tratamiento farmacológico , Humanos , Técnicas de Amplificación de Ácido Nucleico/métodos , Técnicas de Amplificación de Ácido Nucleico/normas , Reacción en Cadena de la Polimerasa , ARN Viral , Reproducibilidad de los Resultados , Sensibilidad y Especificidad , Productos del Gen rev del Virus de la Inmunodeficiencia Humana/química , Productos del Gen tat del Virus de la Inmunodeficiencia Humana/química
12.
Int J Biol Macromol ; 186: 849-863, 2021 Sep 01.
Artículo en Inglés | MEDLINE | ID: mdl-34245737

RESUMEN

Chemotherapy drugs are still one of the first treatment options used in many cancers; however, problems such as cytotoxic side effects on normal cells after systemic administration and resistance to treatment have reduced the use of chemotherapeutics day by day. Targeted delivery of these drugs to the tumor site and sensitization of cancer cells to death induced by chemotherapy drugs are ways that can overcome the limitations of the use of these drugs. In this study, we designed and generated a novel nanocarrier composed of chitosan lactate nanoparticles (NPs) functionalized by HIV-1 derived TAT peptide (Transactivating transcriptional activator) and hyaluronate (HA) to deliver CD73 siRNA and doxorubicin to 4T1 and CT26 cancer cells, both in vivo and in vitro, as a novel combinatorial treatment strategy. The CD73 molecule plays a key role in many cancer cell behaviors such as proliferation, angiogenesis, metastasis, imunosuppression, and resistance to chemotherapy. Therefore, we decided to reduce the side effects of DOX by simultaneously transmitting CD73 siRNA and DOX by CL-TAT-HA NPs, increase the susceptibility of cancer cells to DOX-induced cell death, and stimulate anti-tumor immune responses, for the first time. These results indicated that simultaneous transfer of CD73 siRNA and DOX to cancer cells (4 T1 and CT26) increased cell death and inhibited the prolifration and spread of cancer cells. Also, the preferential aggregation of NPs in the tumor microenvironment reduced tumor growh, promoted the survival of tumor-bearing mice, and induced anti-tumor immune responses. These findings indicate that CL-TAT-HA NPs are a good candidate for targeted siRNA/drug delivery to cancer cells and the simultaneous transfer of CD73 siRNA and DOX to cancer cells using this nanocarrier can be used to treat cancer.


Asunto(s)
5'-Nucleotidasa/genética , Antibióticos Antineoplásicos/farmacología , Neoplasias de la Mama/terapia , Quitosano/química , Neoplasias Colorrectales/terapia , Doxorrubicina/farmacología , Ácido Hialurónico/química , Lactatos/química , ARN Interferente Pequeño/genética , Tratamiento con ARN de Interferencia , Productos del Gen tat del Virus de la Inmunodeficiencia Humana/química , 5'-Nucleotidasa/metabolismo , Animales , Antibióticos Antineoplásicos/química , Antibióticos Antineoplásicos/toxicidad , Apoptosis/efectos de los fármacos , Neoplasias de la Mama/genética , Neoplasias de la Mama/metabolismo , Neoplasias de la Mama/patología , Línea Celular Tumoral , Movimiento Celular/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Quitosano/análogos & derivados , Neoplasias Colorrectales/genética , Neoplasias Colorrectales/metabolismo , Neoplasias Colorrectales/patología , Doxorrubicina/química , Doxorrubicina/toxicidad , Composición de Medicamentos , Femenino , Proteínas Ligadas a GPI/genética , Proteínas Ligadas a GPI/metabolismo , Ratones , Ratones Endogámicos BALB C , Nanopartículas , Nanotecnología , Invasividad Neoplásica , Neovascularización Patológica , ARN Interferente Pequeño/química , ARN Interferente Pequeño/metabolismo
13.
Phys Chem Chem Phys ; 23(19): 11211-11223, 2021 May 19.
Artículo en Inglés | MEDLINE | ID: mdl-34010381

RESUMEN

Ribonucleic acid (RNA) molecules are known to undergo conformational changes in response to various environmental stimuli including temperature, pH, and ligands. In particular, viral RNA molecules are a key example of conformationally adapting molecules that have evolved to switch between many functional conformations. The transactivation response element (TAR) RNA from the type-1 human immunodeficiency virus (HIV-1) is a viral RNA molecule that is being increasingly explored as a potential therapeutic target due to its role in the viral replication process. In this work, we have studied the dynamics in TAR RNA in apo and liganded states by performing explicit-solvent molecular dynamics (MD) simulations initiated with 27 distinct structures. We determined that the TAR RNA structure is significantly stabilized on ligand binding with especially decreased fluctuations in its two helices. This rigidity is further coupled with the decreased flipping of bulge nucleotides, which were observed to flip more frequently in the absence of ligands. We found that initially-distinct structures of TAR RNA converged to similar conformations on removing ligands. We also report that conformational dynamics in unliganded TAR structures leads to the formation of binding pockets capable of accommodating ligands of various sizes.


Asunto(s)
VIH-1/química , ARN Viral/química , Productos del Gen tat del Virus de la Inmunodeficiencia Humana/química , Ligandos , Simulación de Dinámica Molecular , Conformación de Ácido Nucleico
14.
Commun Biol ; 4(1): 214, 2021 02 16.
Artículo en Inglés | MEDLINE | ID: mdl-33594275

RESUMEN

Graphene quantum dots (GQDs) are nano-sized graphene slices. With their small size, lamellar and aromatic-ring structure, GQDs tend to enter into the cell nucleus and interfere with DNA activity. Thus, GQD alone is expected to be an anticancer reagent. Herein, we developed GQDs that suppress the growth of tumor by selectively damaging the DNA of cancer cells. The amine-functionalized GQDs were modified with nucleus targeting TAT peptides (TAT-NGs) and further grafted with cancer-cell-targeting folic acid (FA) modified PEG via disulfide linkage (FAPEG-TNGs). The resulting FAPEG-TNGs exhibited good biocompatibility, nucleus uptake, and cancer cell targeting. They adsorb on DNA via the π-π and electrostatic interactions, which induce the DNA damage, the upregulation of the cell apoptosis related proteins, and the suppression of cancer cell growth, ultimately. This work presents a rational design of GQDs that induce the DNA damage to realize high therapeutic performance, leading to a distinct chemotherapy strategy for targeted tumor therapy.


Asunto(s)
Antineoplásicos/farmacología , Materiales Biocompatibles , Núcleo Celular/efectos de los fármacos , Daño del ADN , Grafito/farmacología , Puntos Cuánticos , Neoplasias del Cuello Uterino/tratamiento farmacológico , Animales , Antineoplásicos/química , Antineoplásicos/metabolismo , Muerte Celular/efectos de los fármacos , Núcleo Celular/metabolismo , Núcleo Celular/patología , Composición de Medicamentos , Femenino , Ácido Fólico/química , Ácido Fólico/metabolismo , Grafito/química , Grafito/metabolismo , Células HeLa , Humanos , Ratones Endogámicos BALB C , Ratones Desnudos , Nanomedicina , Fragmentos de Péptidos/química , Fragmentos de Péptidos/metabolismo , Polietilenglicoles/química , Neoplasias del Cuello Uterino/metabolismo , Neoplasias del Cuello Uterino/patología , Ensayos Antitumor por Modelo de Xenoinjerto , Productos del Gen tat del Virus de la Inmunodeficiencia Humana/química , Productos del Gen tat del Virus de la Inmunodeficiencia Humana/metabolismo
15.
Protein Expr Purif ; 182: 105844, 2021 06.
Artículo en Inglés | MEDLINE | ID: mdl-33592251

RESUMEN

The human autophagy-related protein ATG7 (hATG7), an E1-like ubiquitin enzyme, activates two ubiquitin-like proteins, LC3 (Atg8) and Atg12, and promotes autophagosome formation. While hATG7 plays an essential role for the autophagy conjugation system, the production of full-length functional hATG7 in bacterial systems remains challenging. Previous studies have demonstrated that the HIV-1 virus-encoded Tat peptide ('GRKKRRQRRR') can increase the yield and solubility of heterologous proteins. Here, functional full-length hATG7 was expressed using the pET28b-Tat expression vector in the Escherichia coli BL21 (DE3) strain. Recombinant hATG7 protein aggregated as inclusion bodies while expressed with widely used prokaryotic expression plasmids. In contrast, the solubility of Tat-tagged hATG7 increased significantly with prolonged time compared to Tat-free hATG7. The recombinant proteins were purified to >90% homogeneity under native conditions with a single step of affinity chromatography purification. The results of in vitro pull-down and LC3B-I lipidation assays showed that Tat-tagged hATG7 directly interacted with LC3B-I and promoted LC3B-I lipidation, suggesting that Tat-tagged hATG7 has significant catalytic activity. Overall, this study provides a novel method for improving the functional expression of full-length hATG7 in bacterial systems by fusion with the Tat peptide, a process which may be applied in future studies of hATG7 structure and function.


Asunto(s)
Proteína 7 Relacionada con la Autofagia , Escherichia coli , Expresión Génica , VIH-1/genética , Proteínas Recombinantes de Fusión , Productos del Gen tat del Virus de la Inmunodeficiencia Humana , Proteína 7 Relacionada con la Autofagia/biosíntesis , Proteína 7 Relacionada con la Autofagia/química , Proteína 7 Relacionada con la Autofagia/genética , Proteína 7 Relacionada con la Autofagia/aislamiento & purificación , Escherichia coli/genética , Escherichia coli/metabolismo , Humanos , Proteínas Recombinantes de Fusión/biosíntesis , Proteínas Recombinantes de Fusión/química , Proteínas Recombinantes de Fusión/genética , Proteínas Recombinantes de Fusión/aislamiento & purificación , Productos del Gen tat del Virus de la Inmunodeficiencia Humana/biosíntesis , Productos del Gen tat del Virus de la Inmunodeficiencia Humana/química , Productos del Gen tat del Virus de la Inmunodeficiencia Humana/genética , Productos del Gen tat del Virus de la Inmunodeficiencia Humana/aislamiento & purificación
16.
PLoS One ; 16(2): e0247396, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-33606823

RESUMEN

Among various delivery systems for vaccine and drug delivery, cell-penetrating peptides (CPPs) have been known as a potent delivery system because of their capability to penetrate cell membranes and deliver some types of cargoes into cells. Several CPPs were found in the proteome of viruses such as Tat originated from human immunodeficiency virus-1 (HIV-1), and VP22 derived from herpes simplex virus-1 (HSV-1). In the current study, a wide-range of CPPs was identified in the proteome of SARS-CoV-2, a new member of coronaviruses family, using in silico analyses. These CPPs may play a main role for high penetration of virus into cells and infection of host. At first, we submitted the proteome of SARS-CoV-2 to CellPPD web server that resulted in a huge number of CPPs with ten residues in length. Afterward, we submitted the predicted CPPs to C2Pred web server for evaluation of the probability of each peptide. Then, the uptake efficiency of each peptide was investigated using CPPred-RF and MLCPP web servers. Next, the physicochemical properties of the predicted CPPs including net charge, theoretical isoelectric point (pI), amphipathicity, molecular weight, and water solubility were calculated using protparam and pepcalc tools. In addition, the probability of membrane binding potential and cellular localization of each CPP were estimated by Boman index using APD3 web server, D factor, and TMHMM web server. On the other hand, the immunogenicity, toxicity, allergenicity, hemolytic potency, and half-life of CPPs were predicted using various web servers. Finally, the tertiary structure and the helical wheel projection of some CPPs were predicted by PEP-FOLD3 and Heliquest web servers, respectively. These CPPs were divided into: a) CPP containing tumor homing motif (RGD) and/or tumor penetrating motif (RXXR); b) CPP with the highest Boman index; c) CPP with high half-life (~100 hour) in mammalian cells, and d) CPP with +5.00 net charge. Based on the results, we found a large number of novel CPPs with various features. Some of these CPPs possess tumor-specific motifs which can be evaluated in cancer therapy. Furthermore, the novel and potent CPPs derived from SARS-CoV-2 may be used alone or conjugated to some sequences such as nuclear localization sequence (NLS) for vaccine and drug delivery.


Asunto(s)
Vacunas contra la COVID-19/química , Péptidos de Penetración Celular/química , Biología Computacional , Simulación por Computador , Sistemas de Liberación de Medicamentos , Proteoma , SARS-CoV-2/química , Animales , COVID-19 , Vacunas contra la COVID-19/genética , Vacunas contra la COVID-19/metabolismo , Péptidos de Penetración Celular/genética , Péptidos de Penetración Celular/metabolismo , VIH-1/química , VIH-1/genética , Herpesvirus Humano 1/química , Herpesvirus Humano 1/genética , Humanos , SARS-CoV-2/genética , SARS-CoV-2/metabolismo , Proteínas Estructurales Virales/química , Proteínas Estructurales Virales/genética , Productos del Gen tat del Virus de la Inmunodeficiencia Humana/química , Productos del Gen tat del Virus de la Inmunodeficiencia Humana/genética
17.
J Nanobiotechnology ; 19(1): 28, 2021 Jan 21.
Artículo en Inglés | MEDLINE | ID: mdl-33478501

RESUMEN

BACKGROUND: Spinal Cord injury (SCI) is a kind of severe traumatic disease. The inflammatory response is a significant feature after SCI. Tetramethylpyrazine (TMP), a perennial herb of umbelliferae, is an alkaloid extracted from ligustici. TMP can inhibit the production of nitric oxide and reduce the inflammatory response in peripheral tissues. It can be seen that the therapeutic effect of TMP on SCI is worthy of affirmation. TMP has defects such as short half-life and poor water-solubility. In addition, the commonly used dosage forms of TMP include tablets, dropping pills, injections, etc., and its tissue and organ targeting is still a difficult problem to solve. To improve the solubility and targeting of TMP, here, we developed a nanotechnology-based drug delivery system, TMP-loaded nanoparticles modified with HIV trans-activator of transcription (TAT-TMP-NPs). RESULTS: The nanoparticles prepared in this study has integrated structure. The hemolysis rate of each group is less than 5%, indicating that the target drug delivery system has good safety. The results of in vivo pharmacokinetic studies show that TAT-TMP-NPs improves the bioavailability of TMP. The quantitative results of drug distribution in vivo show that TAT-TMP-NPs is more distributed in spinal cord tissue and had higher tissue targeting ability compared with other treatment groups. CONCLUSIONS: The target drug delivery system can overcome the defect of low solubility of TMP, achieve the targeting ability, and show the further clinical application prospect.


Asunto(s)
Preparaciones de Acción Retardada/química , Pirazinas/administración & dosificación , Albúmina Sérica/química , Traumatismos de la Médula Espinal/tratamiento farmacológico , Vasodilatadores/administración & dosificación , Productos del Gen tat del Virus de la Inmunodeficiencia Humana/química , Animales , Línea Celular , Sistemas de Liberación de Medicamentos , Humanos , Masculino , Ratones , Nanopartículas/química , Pirazinas/farmacocinética , Pirazinas/uso terapéutico , Ratas Sprague-Dawley , Traumatismos de la Médula Espinal/patología , Vasodilatadores/farmacocinética , Vasodilatadores/uso terapéutico
18.
Biochim Biophys Acta Mol Cell Res ; 1868(1): 118887, 2021 01.
Artículo en Inglés | MEDLINE | ID: mdl-33075383

RESUMEN

The G protein-coupled receptor (GPCR) dimer interface plays an important role in the formation and stabilization of the dimer. Therefore, identifying the potential receptor-receptor interface is an important part of studying GPCRs. Various strategies have been employed to study the GPCR dimer interface and explore its functional significance, but experimental methods lack robustness and calculations are laborious. Herein, we report a combined optimized experimental and calculation approach for identifying and structurally characterizing GPCR dimer interfaces, and constructing atomic resolution models. Using a transmembrane domain (TM) peptide containing a human immunodeficiency virus trans-acting transcriptional activator (HIV-TAT) protein transduction motif, matrix-assisted laser desorption tandem time-of-flight mass spectrometry (MALDITOF-MS), and bioluminescence resonance energy transfer (BRET), we successfully identified Apelin receptor (APJ)/Nociceptin receptor 1 (ORL1) and APJ/Vasopressin receptor 2 (V2R) heterodimer interfaces, and two key sites mediating dimerization. This method can identify dimer interfaces of GPCR homodimers and heterodimers.


Asunto(s)
Dimerización , Conformación Proteica , Receptores Acoplados a Proteínas G/ultraestructura , Proteínas Recombinantes de Fusión/genética , Survivin/genética , Productos del Gen tat del Virus de la Inmunodeficiencia Humana/genética , Secuencias de Aminoácidos/genética , Membrana Celular/química , Proteínas de Unión al GTP/química , Células HEK293 , Humanos , Modelos Moleculares , Dominios Proteicos/genética , Multimerización de Proteína/genética , Receptores Acoplados a Proteínas G/química , Receptores Acoplados a Proteínas G/genética , Proteínas Recombinantes de Fusión/química , Espectrometría de Masa por Láser de Matriz Asistida de Ionización Desorción , Survivin/química , Productos del Gen tat del Virus de la Inmunodeficiencia Humana/química
19.
Int J Nanomedicine ; 15: 8383-8400, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-33149582

RESUMEN

PURPOSE: Cancer tissue-specific and nuclei-targeted drug delivery is ideal for the delivery of chemotherapy. However, it has only been achieved in in vitro studies mainly due to low efficiency in vivo. In this study, we aimed to establish an efficient dual-targeted system that targets liver cancer tissue as well as the nuclei of cancer cells in vivo. METHODS: We first synthesized TAT peptide (TATp)-mesoporous silica nanoparticle (MSN) complex (TATp-MSN) and generated liposomes that carried liver cancer-specific aptamer TLS11a (TLS11a-LB). We then generated the drug TLS11a-LB@TATp-MSN/doxorubicin (DOX) by mixing TLS11a-LB and DOX-loaded TATp-MSN. After physical and chemical characterization of the nanoparticles, DOX release from these formulations was evaluated at pH 5.0 and 7.4. Furthermore, we also evaluated nuclear localization and cytotoxicity of the drug in H22 cells in vitro and investigated the liver cancer targeting and antitumor activities of the nano-drug in vivo using a H22 tumor-bearing mice model. RESULTS: TLS11a-LB@TATp-MSN/DOX and its controls were confirmed as nano-drugs (<100 nm) using transmission electron microscopy (TEM). The DOX release rate of TLS11a-LB@TATp-MSN/DOX was significantly faster at pH 5.0 than at pH 7.4. TLS11a-LB@TATp-MSN/DOX effectively targeted the nuclei of H22 cells and released DOX with a higher efficiency than that of the control groups. In addition, TLS11a-LB@TATp-MSN/DOX exhibited slight cytotoxicity, but not significantly more than controls. In vivo studies showed that TLS11a-LB@TATp-MSN accumulated in subcutaneous H22 tumors in the right axilla of BALB/c mice, reaching peak levels at 48 h after intravenous injection, respectively, and demonstrated that TLS11a-LB@TATp-MSN/DOX group enhanced tumor treatment efficacy while reducing systemic side effects. CONCLUSION: TLS11a-LB@TATp-MSN/DOX can efficiently deliver DOX to the nuclei of liver cancer cells by dual targeting liver cancer tissue and the nuclei of the cancer cells in mice. Thus, it is a promising nano-drug for the treatment of liver cancer.


Asunto(s)
Núcleo Celular/patología , Portadores de Fármacos/química , Neoplasias Hepáticas/patología , Nanopartículas/química , Dióxido de Silicio/química , Animales , Antineoplásicos/uso terapéutico , Aptámeros de Péptidos/química , Carcinoma Hepatocelular/tratamiento farmacológico , Línea Celular Tumoral , Doxorrubicina/administración & dosificación , Doxorrubicina/uso terapéutico , Sistemas de Liberación de Medicamentos , Humanos , Concentración de Iones de Hidrógeno , Neoplasias Hepáticas/tratamiento farmacológico , Ratones Endogámicos BALB C , Nanopartículas/ultraestructura , Porosidad , Productos del Gen tat del Virus de la Inmunodeficiencia Humana/química
20.
Int J Mol Sci ; 21(19)2020 Oct 08.
Artículo en Inglés | MEDLINE | ID: mdl-33050051

RESUMEN

Phosphoglycerate mutase 1 (PGAM1) is a glycolytic enzyme that increases glycolytic flux in the brain. In the present study, we examined the effects of PGAM1 in conditions of oxidative stress and ischemic damage in motor neuron-like (NSC34) cells and the rabbit spinal cord. A Tat-PGAM1 fusion protein was prepared to allow easy crossing of the blood-brain barrier, and Control-PGAM1 was synthesized without the Tat peptide protein transduction domain. Intracellular delivery of Tat-PGAM1, not Control-PGAM1, was achieved in a time- and concentration-dependent manner. Immunofluorescent staining confirmed the intracellular expression of Tat-PGAM1 in NSC34 cells. Tat-PGAM1, but not Control-PGAM1, significantly alleviated H2O2-induced oxidative stress, neuronal death, mitogen-activated protein kinase, and apoptosis-inducing factor expression in NSC34 cells. After ischemia induction in the spinal cord, Tat-PGAM1 treatment significantly improved ischemia-induced neurological impairments and ameliorated neuronal cell death in the ventral horn of the spinal cord 72 h after ischemia. Tat-PGAM1 treatment significantly mitigated the ischemia-induced increase in malondialdehyde and 8-iso-prostaglandin F2α production in the spinal cord. In addition, Tat-PGAM1, but not Control-PGAM1, significantly decreased microglial activation and secretion of pro-inflammatory cytokines, such as interleukin (IL)-1ß, IL-6, and tumor necrosis factor (TNF)-α induced by ischemia in the ventral horn of the spinal cord. These results suggest that Tat-PGAM1 can be used as a therapeutic agent to reduce spinal cord ischemia-induced neuronal damage by lowering the oxidative stress, microglial activation, and secretion of pro-inflammatory cytokines, such as IL-1ß, IL-6, and TNF-α.


Asunto(s)
Muerte Celular/efectos de los fármacos , Sistemas de Liberación de Medicamentos/métodos , Neuronas Motoras/metabolismo , Mielitis/tratamiento farmacológico , Fármacos Neuroprotectores/administración & dosificación , Fosfoglicerato Mutasa/administración & dosificación , Isquemia de la Médula Espinal/tratamiento farmacológico , Animales , Citocinas/metabolismo , Modelos Animales de Enfermedad , Células Híbridas , Peróxido de Hidrógeno/farmacología , Masculino , Ratones , Neuronas Motoras/efectos de los fármacos , Estrés Oxidativo/efectos de los fármacos , Conejos , Transducción de Señal/efectos de los fármacos , Productos del Gen tat del Virus de la Inmunodeficiencia Humana/química
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...