Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 394
Filtrar
1.
Bioorg Med Chem ; 112: 117883, 2024 Oct 01.
Artigo em Inglês | MEDLINE | ID: mdl-39180861

RESUMO

Tagging of cell permeable nuclear localization sequence (NLS) with receptor targeting peptide vectors is an attractive strategy for selectively targeted translocation of therapeutic cargoes. The present study aimed at grafting nuclear localization sequence (NLS) onto breast cancer targeting rL-A9 peptide. Molecular docking analysis revealed higher binding affinity of the peptide, DOTA-NLS-rL-A9 (-26.1 kJ/mol) towards HER2 receptor in comparison to DOTA-rL-A9 peptide (-22.2 kJ/mol). Confocal microscopy data suggested significantly enhanced cellular internalization of NLS-tagged peptide. The engineered HER2-selective, DOTA-NLS-rL-A9 peptide scaffold was radiolabeled with Lu-177 for intracellular delivery of the theranostic radionuclide into tumor cells. [177Lu]Lu-DOTA-NLS-rL-A9 exhibited significantly enhanced binding affinity (4.58 ± 1.77 nM) towards human breast carcinoma SKBR3 cells and cellular internalization (85 % at 24 h) compared to its original analog, [177Lu]Lu-DOTA-rL-A9. In vivo biodistribution studies showed consistent retention of [177Lu]Lu-DOTA-NLS-rL-A9 in the tumor with negligible washout of radioactivity (∼4.1 % ID/g at 48 h). Prolonged tumor activity with rapid off-target tissue clearance resulted in significantly high tumor-to-background ratios. The radiopeptide, [177Lu]Lu-DOTA-NLS-rL-A9 thus, being precisely confined into HER2-expressing tumor cells and exhibiting favourable pharmacokinetic features is an efficient candidate for further screening.


Assuntos
Lutécio , Sinais de Localização Nuclear , Radioisótopos , Receptor ErbB-2 , Humanos , Lutécio/química , Receptor ErbB-2/metabolismo , Animais , Sinais de Localização Nuclear/química , Radioisótopos/química , Camundongos , Feminino , Peptídeos/química , Peptídeos/síntese química , Linhagem Celular Tumoral , Neoplasias da Mama/patologia , Simulação de Acoplamento Molecular , Compostos Radiofarmacêuticos/química , Compostos Radiofarmacêuticos/síntese química , Compostos Radiofarmacêuticos/farmacologia , Distribuição Tecidual
2.
Nano Lett ; 24(34): 10605-10613, 2024 Aug 28.
Artigo em Inglês | MEDLINE | ID: mdl-39145462

RESUMO

A metal-organic frameworks (MOFs)-in-MOF nanovehicle (160 nm), which was constructed with newly prepared ultrasmall Cu(I)Cu(II)-BTC MOFs (UCMs, 2.95 nm) loaded with doxorubicin (DOX) and a nuclear localization signal (NLS) peptide as multicores (UCMDNs) and ZIF-8 as the shell MOF, was proposed to cross layers of biological barriers with adaptive size evolution capacity for achieving efficient nucleus-targeted drug delivery. It first enhanced tumor tissue penetration through its larger nanosize effect. Then the acidic tumor environment made the ZIF-8 shell degrade, releasing small-sized UCMDNs to enter into the cell and into the nucleus under the guidance of NLS. Furthermore, due to the distinct surface structural characteristics of UCMs, UCMDNs remained stable in the cytoplasm and collapsed in the nucleus due to the DOX-DNA interaction to deliver DOX precisely. It showed superior performance in the nucleus-directed delivery of DOX (delivery efficiency up to 56.7%) and a high tumor growth inhibition rate (96.4%), offering promising prospects in tumor chemotherapy.


Assuntos
Núcleo Celular , Doxorrubicina , Estruturas Metalorgânicas , Doxorrubicina/química , Doxorrubicina/farmacologia , Doxorrubicina/administração & dosagem , Estruturas Metalorgânicas/química , Humanos , Núcleo Celular/metabolismo , Núcleo Celular/efeitos dos fármacos , Animais , Camundongos , Portadores de Fármacos/química , Neoplasias/tratamento farmacológico , Neoplasias/patologia , Linhagem Celular Tumoral , Sistemas de Liberação de Medicamentos , Sinais de Localização Nuclear/química , Antineoplásicos/química , Antineoplásicos/farmacologia , Antineoplásicos/administração & dosagem , Tamanho da Partícula , Nanopartículas/química , Cobre/química
3.
Int J Pharm ; 662: 124492, 2024 Sep 05.
Artigo em Inglês | MEDLINE | ID: mdl-39038720

RESUMO

PURPOSES: The objective of this study is to develop a versatile gene carrier based on lipopeptides capable of delivering genetic material into target cells with minimal cytotoxicity. METHODS: Two lipopeptide molecules, palmitoyl-CKKHH and palmitoyl-CKKHH-YGRKKRRQRRR-PKKKRKV, were synthesized using solid phase peptide synthesis and evaluated as transfection agents. Physicochemical characterization of the lipopeptides included a DNA shift mobility assay, particle size measurement, and transmission electron microscopy (TEM) analysis. Cytotoxicity was assessed in CHO-K1 and HepG2 cells using the MTT assay, while transfection efficiency was determined by evaluating the expression of the green fluorescent protein-encoding gene. RESULTS: Our findings demonstrate that the lipopeptides can bind, condense, and shield DNA from DNase degradation. The inclusion of the YGRKKRRQRRR sequence, a transcription trans activator, and the PKKKRKV sequence, a nuclear localization signal, imparts desirable properties. Lipopeptide-based TAT-NLS/DNA nanoparticles exhibited stability for up to 20 days when stored at 6-8 °C, displaying uniformity with a compact size of approximately 120 nm. Furthermore, the lipopeptides exhibited lower cytotoxicity compared to the poly-L-lysine. Transfection experiments revealed that protein expression mediated by the lipopeptide occurred at a charge ratio ranging from 4.0 to 8.0. CONCLUSION: These results indicate that the lipopeptide, composed of a palmitoyl alkyl chain and TAT and NLS sequences, can efficiently condense and protect DNA, form stable and uniform nanoparticles, and exhibit promising characteristics as a potential gene carrier with minimal cytotoxicity.


Assuntos
Cricetulus , DNA , Técnicas de Transferência de Genes , Lipopeptídeos , Nanopartículas , Humanos , Lipopeptídeos/química , Lipopeptídeos/administração & dosagem , Nanopartículas/química , Células Hep G2 , Animais , DNA/administração & dosagem , Células CHO , Sinais de Localização Nuclear/química , Transfecção/métodos , Sobrevivência Celular/efeitos dos fármacos , Tamanho da Partícula , Proteínas de Fluorescência Verde/genética
4.
J Chem Inf Model ; 64(13): 5262-5272, 2024 Jul 08.
Artigo em Inglês | MEDLINE | ID: mdl-38869471

RESUMO

Venezuelan equine encephalitis virus (VEEV) is a highly virulent pathogen whose nuclear localization signal (NLS) sequence from capsid protein binds to the host importin-α transport protein and blocks nuclear import. We studied the molecular mechanisms by which two small ligands, termed I1 and I2, interfere with the binding of VEEV's NLS peptide to importin-α protein. To this end, we performed all-atom replica exchange molecular dynamics simulations probing the competitive binding of the VEEV coreNLS peptide and I1 or I2 ligand to the importin-α major NLS binding site. As a reference, we used our previous simulations, which examined noncompetitive binding of the coreNLS peptide or the inhibitors to importin-α. We found that both inhibitors completely abrogate the native binding of the coreNLS peptide, forcing it to adopt a manifold of nonnative loosely bound poses within the importin-α major NLS binding site. Both inhibitors primarily destabilize the native coreNLS binding by masking its amino acids rather than competing with it for binding to importin-α. Because I2, in contrast to I1, binds off-site localizing on the edge of the major NLS binding site, it inhibits fewer coreNLS native binding interactions than I1. Structural analysis is supported by computations of the free energies of the coreNLS peptide binding to importin-α with or without competition from the inhibitors. Specifically, both inhibitors reduce the free energy gain from coreNLS binding, with I1 causing significantly larger loss than I2. To test our simulations, we performed AlphaScreen experiments measuring IC50 values for both inhibitors. Consistent with in silico results, the IC50 value for I1 was found to be lower than that for I2. We hypothesize that the inhibitory action of I1 and I2 ligands might be specific to the NLS from VEEV's capsid protein.


Assuntos
Ligação Competitiva , Simulação de Dinâmica Molecular , Sinais de Localização Nuclear , alfa Carioferinas , alfa Carioferinas/metabolismo , alfa Carioferinas/química , alfa Carioferinas/antagonistas & inibidores , Ligantes , Sinais de Localização Nuclear/química , Vírus da Encefalite Equina Venezuelana/metabolismo , Vírus da Encefalite Equina Venezuelana/química , Ligação Proteica , Peptídeos/química , Peptídeos/metabolismo , Peptídeos/farmacologia , Sequência de Aminoácidos
5.
FEBS Lett ; 598(7): 801-817, 2024 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-38369616

RESUMO

Secretory proteins of Plasmodium exhibit differential spatial and functional activity within the host cell nucleus. However, the nuclear localization signals (NLSs) for these proteins remain largely uncharacterized. In this study, we have identified and characterized two NLSs in the circumsporozoite protein of Plasmodium falciparum (Pf-CSP). Both NLSs in the Pf-CSP contain clusters of lysine and arginine residues essential for specific interactions with the conserved tryptophan and asparagine residues of importin-α, facilitating nuclear translocation of Pf-CSP. While the two NLSs of Pf-CSP function independently and are both crucial for nuclear localization, a single NLS of Pf-CSP leads to weak nuclear localization. These findings shed light on the mechanism of nuclear penetrability of secretory proteins of Plasmodium proteins.


Assuntos
Sinais de Localização Nuclear , Plasmodium falciparum , Sinais de Localização Nuclear/genética , Sinais de Localização Nuclear/química , Sinais de Localização Nuclear/metabolismo , alfa Carioferinas/metabolismo , Transporte Ativo do Núcleo Celular , Proteínas de Protozoários/genética , Proteínas de Protozoários/metabolismo , Núcleo Celular/metabolismo
6.
J Mol Biol ; 436(3): 168433, 2024 02 01.
Artigo em Inglês | MEDLINE | ID: mdl-38182104

RESUMO

The ligand-regulated PAS domains are one of the most diverse signal-integrating domains found in proteins from prokaryotes to humans. By biochemically connecting cellular processes with their environment, PAS domains facilitate an appropriate cellular response. PAS domain-containing Kinase (PASK) is an evolutionarily conserved protein kinase that plays important signaling roles in mammalian stem cells to establish stem cell fate. We have shown that the nuclear translocation of PASK is stimulated by differentiation signaling cues in muscle stem cells. However, the mechanistic basis of the regulation of PASK nucleo-cytoplasmic translocation remains unknown. Here, we show that the PAS-A domain of PASK contains a putative monopartite nuclear localization sequence (NLS) motif. This NLS is inhibited in cells through intramolecular association with a short linear motif, termed the PAS Interacting Motif (PIM), found upstream of the kinase domain. This interaction serves to retain PASK in the cytosol in the absence of signaling cues. Consistent with that, we show that metabolic inputs induce PASK nuclear import, likely by disrupting this association. We suggest that a route for such linkage may occur through the PAS-A ligand binding cavity. We show that PIM recruitment and artificial ligand binding to the PAS-A domain occur at neighboring locations that could facilitate metabolic control of the PAS-PIM interaction. Thus, the intramolecular interaction in PASK integrates metabolic signaling cues for nuclear translocation and could be targeted to control the balance between self-renewal and differentiation in stem cells.


Assuntos
Sinais de Localização Nuclear , Proteínas Serina-Treonina Quinases , Animais , Humanos , Transporte Ativo do Núcleo Celular , Diferenciação Celular , Ligantes , Fosforilação , Transdução de Sinais , Sinais de Localização Nuclear/química , Domínios Proteicos , Proteínas Serina-Treonina Quinases/química
7.
Protein Sci ; 33(2): e4876, 2024 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-38108201

RESUMO

Nucleocytoplasmic transport regulates the passage of proteins between the nucleus and cytoplasm. In the best characterized pathway, importin (IMP) α bridges cargoes bearing basic, classical nuclear localization signals (cNLSs) to IMPß1, which mediates transport through the nuclear pore complex. IMPα recognizes three types of cNLSs via two binding sites: the major binding site accommodates monopartite cNLSs, the minor binding site recognizes atypical cNLSs, while bipartite cNLSs simultaneously interact with both major and minor sites. Despite the growing knowledge regarding IMPα-cNLS interactions, our understanding of the evolution of cNLSs is limited. We combined bioinformatic, biochemical, functional, and structural approaches to study this phenomenon, using polyomaviruses (PyVs) large tumor antigens (LTAs) as a model. We characterized functional cNLSs from all human (H)PyV LTAs, located between the LXCXE motif and origin binding domain. Surprisingly, the prototypical SV40 monopartite NLS is not well conserved; HPyV LTA NLSs are extremely heterogenous in terms of structural organization, IMPα isoform binding, and nuclear targeting abilities, thus influencing the nuclear accumulation properties of full-length proteins. While several LTAs possess bipartite cNLSs, merkel cell PyV contains a hybrid bipartite cNLS whose upstream stretch of basic amino acids can function as an atypical cNLS, specifically binding to the IMPα minor site upon deletion of the downstream amino acids after viral integration in the host genome. Therefore, duplication of a monopartite cNLS and subsequent accumulation of point mutations, optimizing interaction with distinct IMPα binding sites, led to the evolution of bipartite and atypical NLSs binding at the minor site.


Assuntos
Antígenos de Neoplasias , Sinais de Localização Nuclear , alfa Carioferinas , Humanos , Transporte Ativo do Núcleo Celular/fisiologia , alfa Carioferinas/genética , alfa Carioferinas/química , alfa Carioferinas/metabolismo , Sequência de Aminoácidos , Antígenos de Neoplasias/metabolismo , Núcleo Celular/metabolismo , Sinais de Localização Nuclear/química , Sinais de Localização Nuclear/genética , Sinais de Localização Nuclear/metabolismo
8.
FEBS Lett ; 598(2): 199-209, 2024 01.
Artigo em Inglês | MEDLINE | ID: mdl-38158756

RESUMO

Human cytomegalovirus DNA polymerase processivity factor UL44 is transported into the nucleus by importin (IMP) α/ß through a classical nuclear localization signal (NLS), and this region is susceptible to cdc2-mediated phosphorylation at position T427. Whilst phosphorylation within and close to the UL44 NLS regulates nuclear transport, the details remain elusive, due to the paucity of structural information regarding the role of negatively charged cargo phosphate groups. We addressed this issue by studying the effect of UL44 T427 phosphorylation on interaction with several IMPα isoforms by biochemical and structural approaches. Phosphorylation decreased UL44/IMPα affinity 10-fold, and a comparative structural analysis of UL44 NLS phosphorylated and non-phosphorylated peptides complexed with mouse IMPα2 revealed the structural rearrangements responsible for phosphorylation-dependent inhibition of UL44 nuclear import.


Assuntos
Núcleo Celular , Citomegalovirus , Animais , Humanos , Camundongos , Transporte Ativo do Núcleo Celular , Núcleo Celular/metabolismo , Citomegalovirus/genética , Citomegalovirus/metabolismo , DNA Polimerase Dirigida por DNA/metabolismo , Sinais de Localização Nuclear/química , Sinais de Localização Nuclear/genética , Sinais de Localização Nuclear/metabolismo , Fosforilação
9.
Int J Pharm ; 644: 123299, 2023 Sep 25.
Artigo em Inglês | MEDLINE | ID: mdl-37558147

RESUMO

Polyamidoamine (PAMAM) dendrimers have been explored as an alternative to polyethylenimine (PEI) as a gene delivery carrier because of their relatively low cytotoxicity and excellent biocompatibility. The transfection efficiency of PAMAM dendrimers can be improved by the addition of nuclear localization signal (NLS), a positively charged peptide sequence recognized by cargo proteins in the cytoplasm for nuclear transport. However, increased positive charges from NLS can cause damage to the cytoplasmic and mitochondrial membranes and lead to reactive oxygen species (ROS)-induced cytotoxicity. This negative effect of NLS can be negated without a significant reduction in transfection efficiency by adding histidine, an essential amino acid known as a natural antioxidant, to NLS. However, little is known about the exact mechanism by which histidine reduces cytotoxicity of NLS-modified dendrimers. In this study, we selected cystamine core PAMAM dendrimer generation 2 (cPG2) and conjugated it with NLS derived from Merkel cell polyomavirus large T antigen and histidine (n = 0-3) to improve transfection efficiency and reduce cytoxicity. NLS-modified cPG2 derivatives showed similar or higher transfection efficiency than PEI 25 kDa in NIH3T3 and human mesenchymal stem cells (hMSC). The cytotoxicity of NLS-modified cPG2 derivatives was substantially lower than PEI 25 kDa and was further reduced as the number of histidine in NLS increased. To understand the mechanism of cytoprotective effect of histidine-conjugated NLS, we examined ROS scavenging, hydroxyl radical generation and mitochondrial membrane potential as a function of the number of histidine in NLS. As the number of hisidine increased, cPG2 scavenged ROS more effectively as evidenced by the hydroxyl radical antioxidant capacity (HORAC) assay. This was consistent with the reduced intracellular hydroxyl radical concentration measured by 2',7'-dichlorodihydrofluorescein diacetate (DCFDA) assay in NIH3T3. Finally, fluorescence imaging with JC-1 confirmed that the mitochondrial membranes of NIH 3T3 were well-protected during the transfection when NLS contained histidine. These experimental results confirm the hypothesis that histidine residues scavenge ROS that is generated during the transfection process, preventing the excessive damage to mitochondrial membranes, leading to reduced cytotoxicity.


Assuntos
Dendrímeros , Sinais de Localização Nuclear , Animais , Camundongos , Humanos , Sinais de Localização Nuclear/química , Sinais de Localização Nuclear/genética , Sinais de Localização Nuclear/metabolismo , Histidina , Dendrímeros/química , Células NIH 3T3 , Antioxidantes/farmacologia , Radical Hidroxila , Espécies Reativas de Oxigênio , DNA/química , Técnicas de Transferência de Genes , Transfecção , Sobrevivência Celular
10.
Structure ; 31(8): 891-892, 2023 08 03.
Artigo em Inglês | MEDLINE | ID: mdl-37541190

RESUMO

In this issue of Structure, Gonzalez et al. present the cryo-EM structure of Karyopherin-ß2 bound to the proline-tyrosine nuclear localization signal (PY-NLS) of heterogeneous nuclear ribonucleoprotein H2 (HNRNPH2). The structure advances our understanding of not only the diversity of PY-NLSs but also the pathogenic mechanisms arising from HNRNPH2 variants.


Assuntos
Transtornos do Neurodesenvolvimento , Sinais de Localização Nuclear , Humanos , Sinais de Localização Nuclear/química , Sinais de Localização Nuclear/metabolismo , beta Carioferinas/química , beta Carioferinas/metabolismo , Carioferinas/metabolismo , Tirosina/metabolismo , Transtornos do Neurodesenvolvimento/metabolismo , Núcleo Celular/metabolismo
11.
Biochim Biophys Acta Mol Cell Res ; 1870(7): 119502, 2023 10.
Artigo em Inglês | MEDLINE | ID: mdl-37268023

RESUMO

Our study maps the classic nuclear localization signal (cNLS) domain within WRNIP that directs the protein's nuclear positioning.


Assuntos
Sinais de Localização Nuclear , Sinais de Localização Nuclear/química , Sinais de Localização Nuclear/metabolismo , Humanos
12.
Bioorg Chem ; 135: 106483, 2023 06.
Artigo em Inglês | MEDLINE | ID: mdl-37062230

RESUMO

Nitric oxide (NO) is an important gaseous signaling molecule in various physiological processes, which functions through interactions with its acceptor molecules located in organelles. NO has an extremely short half-life, making it challenging to experimentally achieve effective NO levels in organelles to study these interactions. Here we developed an organelle-specific, peptide-based NO delivery material that targets the nucleus. NO was attached to the SH group of a cysteine residue inserted into the N-terminus of a cell-penetrating peptide (CPP) conjugated to varying repeats of the nuclear localization signal (NLS), which we denoted NO-CysCPP-NLS, through S-nitrosylation. NO-CysCPP-NLS strongly induced osteogenic differentiation of mesenchymal stem cells. This delivery concept can be extended to cells other than stem cells to elucidate the effects of NO release in the nucleus. Furthermore, conjugation of NO to CysCPP fused to mitochondria- or lysosome-targeting signals can be used to deliver NO to other organelles such as mitochondria and lysosomes, respectively.


Assuntos
Peptídeos Penetradores de Células , Células-Tronco Mesenquimais , Humanos , Osteogênese , Óxido Nítrico/farmacologia , Diferenciação Celular , Peptídeos Penetradores de Células/farmacologia , Peptídeos Penetradores de Células/química , Sinais de Localização Nuclear/química , Sinais de Localização Nuclear/farmacologia , Fatores Imunológicos/farmacologia
13.
J Phys Chem B ; 127(14): 3175-3186, 2023 04 13.
Artigo em Inglês | MEDLINE | ID: mdl-37001021

RESUMO

Although Venezuelan equine encephalitis virus (VEEV) is a life-threatening pathogen with a capacity for epidemic outbreaks, there are no FDA-approved VEEV antivirals for humans. VEEV cytotoxicity is partially attributed to the formation of a tetrameric complex between the VEEV capsid protein, the nuclear import proteins importin-α and importin-ß, and the nuclear export protein CRM1, which together block trafficking through the nuclear pore complex. Experimental studies have identified small molecules from the CL6662 scaffold as potential inhibitors of the viral nuclear localization signal (NLS) sequence binding to importin-α. However, little is known about the molecular mechanism of CL6662 inhibition. To address this issue, we employed all-atom replica exchange molecular dynamics simulations to probe, in atomistic detail, the binding mechanism of CL6662 ligands to importin-α. Three ligands, including G281-1485 and two congeners with varying hydrophobicities, were considered. We investigated the distribution of ligand binding poses, their locations, and ligand specificities measured by the strength of binding interactions. We found that G281-1485 binds nonspecifically without forming well-defined binding poses throughout the NLS binding site. Binding of the less hydrophobic congener becomes strongly on-target with respect to the NLS binding site but remains nonspecific. However, a more hydrophobic congener is a strongly specific binder and the only ligand out of three to form a well-defined binding pose, while partially overlapping with the NLS binding site. On the basis of free energy estimates, we argue that all three ligands weakly compete with the viral NLS sequence for binding to importin-α in an apparent compromise to preserve host NLS binding. We further show that all-atom replica exchange binding simulations are a viable tool for studying ligands binding nonspecifically without forming well-defined binding poses.


Assuntos
Vírus da Encefalite Equina Venezuelana , alfa Carioferinas , Animais , Cavalos , Humanos , alfa Carioferinas/química , alfa Carioferinas/metabolismo , Vírus da Encefalite Equina Venezuelana/metabolismo , Simulação de Dinâmica Molecular , Ligantes , Sinais de Localização Nuclear/química , Sinais de Localização Nuclear/metabolismo , Núcleo Celular/metabolismo , Sítios de Ligação , Ligação Proteica
14.
J Biomol Struct Dyn ; 41(10): 4641-4649, 2023 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-35510584

RESUMO

TULP3 is involved in cell regulation pathways including transcription and signal transduction. In some pathological states like in cancers, increased level of TULP3 has been observed so it can serve as a potential target to hamper the activation of those pathways. We propose a novel idea of inhibiting nuclear localization signal (NLS) to interrupt nuclear translocation of TULP3 so that the downstream activations of pathways are blocked. In current in silico study, 3D structure of TULP3 was modeled using 8 different tools including I-TASSER, CABS-FOLD, Phyre2, PSIPRED, RaptorX, Robetta, Rosetta and Prime by Schrödinger. Best structure was selected after quality evaluation by SAVES and implied for the investigation of NLS sequence. Mapped NLS sequence was further used to dock with natural ligand importin-α as control docking to validate the NLS sequence as binding site. After docking and molecular dynamics (MD) simulation validation, these residues were used as binding side for subsequent docking studies. 70 alkaloids were selected after intensive literature survey and were virtually docked with NLS sequence where natural ligand importin-α is supposed to be bound. This study demonstrates the virtual inhibition of NLS sequence so that it paves a way for future in-vivo studies to use NLS as a new drug target for cancer therapeutics.Communicated by Ramaswamy H. Sarma.


Assuntos
Sinais de Localização Nuclear , alfa Carioferinas , Sinais de Localização Nuclear/química , alfa Carioferinas/química , Ligantes , Ligação Proteica , Núcleo Celular/metabolismo , Transporte Ativo do Núcleo Celular
15.
Biomater Sci ; 10(15): 4309-4323, 2022 Jul 26.
Artigo em Inglês | MEDLINE | ID: mdl-35771211

RESUMO

The design of non-viral vectors that efficiently deliver genetic materials into cells, in particular to the nucleus, remains a major challenge in gene therapy and vaccine development. To tackle the problems associated with cellular uptake and nuclear targeting, here we introduce a delivery platform based on the self-assembly of an amphiphilic peptide carrying an N-terminal KRKR sequence that functions as a nuclear localization signal (NLS). By means of a single-step self-assembly process, the amphiphilic peptides afford the generation of NLS-functionalized multicompartment micellar nanostructures that can embed various oligonucleotides between their individual compartments. Detailed physicochemical, cellular and ultrastructural analyses demonstrated that integrating an NLS in the hydrophilic domain of the peptide along with tuning its hydrophobic domain led to self-assembled DNA-loaded multicompartment micelles (MCMs) with enhanced cellular uptake and nuclear translocation. We showed that the nuclear targeting ensued via the NLS interaction with the nuclear transport receptors of the karyopherin family. Importantly, we observed that the treatment of MCF-7 cells with NLS-MCMs loaded with anti-BCL2 antisense oligonucleotides resulted in up to 86% knockdown of BCL2, an inhibitor of apoptosis that is overexpressed in more than half of all human cancers. We envision that this platform can be used to efficiently entrap and deliver diverse genetic payloads to the nucleus and find applications in basic research and biomedicine.


Assuntos
Sinais de Localização Nuclear , Oligonucleotídeos , Transporte Ativo do Núcleo Celular/genética , Núcleo Celular/metabolismo , Humanos , Micelas , Sinais de Localização Nuclear/química , Sinais de Localização Nuclear/genética , Sinais de Localização Nuclear/metabolismo , Oligonucleotídeos/metabolismo , Peptídeos/química
16.
Molecules ; 27(12)2022 Jun 13.
Artigo em Inglês | MEDLINE | ID: mdl-35744930

RESUMO

Compromised activity is a common impediment for biologics requiring endosome trafficking into target cells. In cancer cells, antibody-drug conjugates (ADCs) are trapped in endosomes or subsequently pumped extracellularly, leading to a reduction in intracellular accumulation. In subsets of dendritic cells (DCs), endosome-engulfed antigens face non-specific proteolysis and collateral damage to epitope immunogenicity before proteasomal processing and subsequent surface presentation. To bypass these shortcomings, we devised Accum™, a conjugable biotechnology harboring cholic acid (ChAc) and a nuclear localization signal (NLS) sequence for endosome escape and prompt nuclear targeting. Combined, these mechanisms culminate in enhanced intracellular accumulation and functionalization of coupled biologics. As proof-of-principle, we have biochemically characterized Accum, demonstrating its adaptability to ADCs or antigens in different cancer settings. Additionally, we have validated that endosome escape and nuclear routing are indispensable for effective intracellular accumulation and guaranteed target cell selectivity. Importantly, we have demonstrated that the unique mechanism of action of Accum translates into enhanced tumor cytotoxicity when coupled to ADCs, and durable therapeutic and prophylactic anti-cancer immunogenicity when coupled to tumor antigens. As more pre-clinical evidence accumulates, the adaptability, unique mechanism of action, and high therapeutic potency of Accum signal a promising transition into clinical investigations in the context of onco-immunotherapy.


Assuntos
Produtos Biológicos , Imunoconjugados , Antígenos de Neoplasias , Produtos Biológicos/farmacologia , Endossomos , Imunoconjugados/farmacologia , Imunoconjugados/uso terapêutico , Imunoterapia , Sinais de Localização Nuclear/química
17.
J Biomol Struct Dyn ; 40(17): 8100-8111, 2022 10.
Artigo em Inglês | MEDLINE | ID: mdl-33950784

RESUMO

The novel corona virus (Covid-19) has become a great challenge worldwide since 2019, as no drug has been reported yet. Different clinical trials are still under way. Among them is Ivermectin (IVM), an FDA approved drug which was recently reported as a successful candidate to reduce SARS-CoV-2 viral load by inhibiting Importin-α1 (IMP-α1) protein which subsequently affects nuclear transport of viral proteins but its basic binding mode and inhibitory mechanism is unknown. Therefore, we aimed to explore the inhibitory mechanism and binding mode of IVM with IMP-α1 via different computational methods. Initially, comparative docking of IVM was performed against two different binding sites (Nuclear Localization Signal (NLS) major and minor sites) of IMP-α1 to predict the probable binding mode of IVM. Then, classical MD simulation was performed (IVM/NLS-Major site and IVM/NLS-Minor site), to predict its comparative stability dynamics and probable inhibitory mechanism. The stability dynamics and biophysical analysis of both sites highlighted the stable binding of IVM within NLS-Minor site by establishing and maintaining more hydrophobic contacts with crucial residues, required for IMP-α1 inhibition which were not observed in NLS-major site. Altogether, these results recommended the worth of IVM as a possible drug to limit the SARS-CoV-2 viral load and consequently reduces its progression.Communicated by Ramaswamy H. Sarma.


Assuntos
Tratamento Farmacológico da COVID-19 , Sinais de Localização Nuclear , Humanos , Ivermectina/farmacologia , Simulação de Acoplamento Molecular , Simulação de Dinâmica Molecular , Sinais de Localização Nuclear/química , SARS-CoV-2 , Proteínas Virais
18.
J Biol Chem ; 297(2): 100831, 2021 08.
Artigo em Inglês | MEDLINE | ID: mdl-34174284

RESUMO

The ATR pathway is one of the major DNA damage checkpoints, and Rad17 is a DNA-binding protein that is phosphorylated upon DNA damage by ATR kinase. Rad17 recruits the 9-1-1 complex that mediates the checkpoint activation, and proteasomal degradation of Rad17 is important for recovery from the ATR pathway. Here, we identified several Rad17 mutants deficient in nuclear localization and resistant to proteasomal degradation. The nuclear localization signal was identified in the central basic domain of Rad17. Rad17 Δ230-270 and R240A/L243A mutants that were previously postulated to lack the destruction box, a sequence that is recognized by the ubiquitin ligase/anaphase-promoting complex that mediates degradation of Rad17, also showed cytoplasmic localization. Our data indicate that the nuclear translocation of Rad17 is functionally linked to the proteasomal degradation. The ATP-binding activity of Rad17, but not hydrolysis, is essential for the nuclear translocation, and the ATPase domain orchestrates the nuclear translocation, the proteasomal degradation, as well as the interaction with the 9-1-1 complex. The Rad17 mutant that lacked a nuclear localization signal was proficient in the interaction with the 9-1-1 complex, suggesting cytosolic association of Rad17 and the 9-1-1 complex. Finally, we identified two tandem canonical and noncanonical destruction boxes in the N-terminus of Rad17 as the bona fide destruction box, supporting the role of anaphase-promoting complex in the degradation of Rad17. We propose a model in which Rad17 is activated in the cytoplasm for translocation into the nucleus and continuously degraded in the nucleus even in the absence of exogenous DNA damage.


Assuntos
Ciclossomo-Complexo Promotor de Anáfase/metabolismo , Proteínas de Ciclo Celular/metabolismo , Núcleo Celular/metabolismo , Dano ao DNA , Sinais de Localização Nuclear/metabolismo , Complexo de Endopeptidases do Proteassoma/metabolismo , Animais , Pontos de Checagem do Ciclo Celular , Células Cultivadas , Chlorocebus aethiops , Humanos , Sinais de Localização Nuclear/química , Fosforilação , Proteólise
19.
Clin Exp Pharmacol Physiol ; 48(10): 1346-1357, 2021 10.
Artigo em Inglês | MEDLINE | ID: mdl-34060659

RESUMO

Rheumatoid arthritis (RA) is a chronic inflammatory autoimmune disease that causes functional disability due to bone destruction and severe joint pain. Current anti-rheumatic treatments develop severe complications and do not provide complete remission. Gold nanoparticles (AuNPs) have garnered attention because of their unique physical and chemical properties. In this study, we have evaluated the therapeutic effects of gold nanospheres (AuNSs) with two different ligands (targeted-nanoparticles) against collagen-induced arthritis (CIA) and compared the outcomes with conventional methotrexate (MTX) and biological (infliximab) treatments. Clinical evaluation was performed by radiographic and histological examinations. The bioaccumulation of AuNSs in vital organs was assessed. The mechanistic studies targeting pro-inflammatory/anti-inflammatory and angiogenic mediators' expressions were performed. Radiographic examination showed that the targeted AuNSs reduced joint space narrowing and bone erosion. Moreover, histopathological examination of rat ankle joints demonstrated that targeted AuNSs reduce bone and cartilage degeneration/inflammation. Gold nanospheres-conjugated with nucleus localized peptide (nuclear membrane-targeted) (AuNSs@NLS) has resolved bone destruction and inflammation compared to gold nanospheres-conjugated at polyethylene glycol (AuNSs@PEG). Although the AuNSs accumulated in different organs in both cases, they did not induce any toxicity or tissue damage. The two different targeted AuNSs significantly suppress inflammatory and angiogenic mediators' expression and induced anti-inflammatory cytokine production, but the AuNSs@NLS had superior therapeutic efficacy. In conclusion, these results suggested that nuclear membrane-targeted AuNSs effectively attenuated arthritis progression without systemic side effects.


Assuntos
Artrite Experimental/tratamento farmacológico , Ouro/administração & dosagem , Nanopartículas Metálicas/uso terapêutico , Nanosferas/administração & dosagem , Animais , Artrite Experimental/induzido quimicamente , Artrite Experimental/patologia , Modelos Animais de Doenças , Feminino , Ouro/química , Nanopartículas Metálicas/química , Nanosferas/química , Sinais de Localização Nuclear/química , Polietilenoglicóis/química , Ratos , Ratos Wistar , Distribuição Tecidual
20.
RNA ; 27(2): 234-241, 2021 02.
Artigo em Inglês | MEDLINE | ID: mdl-33148600

RESUMO

Correct cellular localization is essential for the function of many eukaryotic proteins and hence cell physiology. Here, we present a synthetic genetic device that allows the control of nuclear and cytosolic localization based on controlled alternative splicing in human cells. The device is based on the fact that an alternative 3' splice site is located within a TetR aptamer that in turn is positioned between the branch point and the canonical splice site. The novel splice site is only recognized when the TetR repressor is bound. Addition of doxycycline prevents TetR aptamer binding and leads to recognition of the canonical 3' splice site. It is thus possible to produce two independent splice isoforms. Since the terminal loop of the aptamer may be replaced with any sequence of choice, one of the two isoforms may be extended by the respective sequence of choice depending on the presence of doxycycline. In a proof-of-concept study, we fused a nuclear localization sequence to a cytosolic target protein, thus directing the protein into the nucleus. However, the system is not limited to the control of nuclear localization. In principle, any target sequence can be integrated into the aptamer, allowing not only the production of a variety of different isoforms on demand, but also to study the function of mislocalized proteins. Moreover, it also provides a valuable tool for investigating the mechanism of alternative splicing in human cells.


Assuntos
Processamento Alternativo , Aptâmeros de Nucleotídeos/metabolismo , Sinais de Localização Nuclear/metabolismo , Sítios de Splice de RNA , Proteínas Repressoras/metabolismo , Transporte Ativo do Núcleo Celular/efeitos dos fármacos , Processamento Alternativo/efeitos dos fármacos , Aptâmeros de Nucleotídeos/síntese química , Aptâmeros de Nucleotídeos/genética , Pareamento de Bases , Sequência de Bases , Sítios de Ligação , Núcleo Celular/química , Núcleo Celular/metabolismo , Citosol/química , Citosol/metabolismo , Doxiciclina/farmacologia , Éxons , Células HeLa , Humanos , Íntrons , Modelos Moleculares , Sinais de Localização Nuclear/química , Sinais de Localização Nuclear/genética , Ligação Proteica , Conformação Proteica em alfa-Hélice , Conformação Proteica em Folha beta , Domínios e Motivos de Interação entre Proteínas , Proteínas Repressoras/química , Proteínas Repressoras/genética
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA