Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 163
Filtrar
Más filtros

Tipo del documento
Intervalo de año de publicación
1.
Cell ; 174(6): 1465-1476.e13, 2018 09 06.
Artículo en Inglés | MEDLINE | ID: mdl-30122350

RESUMEN

Cell-penetrating peptides (CPPs) are short protein segments that can transport cargos into cells. Although CPPs are widely studied as potential drug delivery tools, their role in normal cell physiology is poorly understood. Early during infection, the L2 capsid protein of human papillomaviruses binds retromer, a cytoplasmic trafficking factor required for delivery of the incoming non-enveloped virus into the retrograde transport pathway. Here, we show that the C terminus of HPV L2 proteins contains a conserved cationic CPP that drives passage of a segment of the L2 protein through the endosomal membrane into the cytoplasm, where it binds retromer, thereby sorting the virus into the retrograde pathway for transport to the trans-Golgi network. These experiments define the cell-autonomous biological role of a CPP in its natural context and reveal how a luminal viral protein engages an essential cytoplasmic entry factor.


Asunto(s)
Proteínas de la Cápside/metabolismo , Péptidos de Penetración Celular/metabolismo , Proteínas Oncogénicas Virales/metabolismo , Secuencia de Aminoácidos , Proteínas de la Cápside/química , Proteínas de la Cápside/genética , Péptidos de Penetración Celular/química , Péptidos de Penetración Celular/genética , Endosomas/metabolismo , Aparato de Golgi/virología , Proteínas Fluorescentes Verdes/genética , Proteínas Fluorescentes Verdes/metabolismo , Células HEK293 , Células HeLa , Papillomavirus Humano 16/genética , Papillomavirus Humano 16/fisiología , Humanos , Mutagénesis , Proteínas Oncogénicas Virales/química , Proteínas Oncogénicas Virales/genética , Transporte de Proteínas , Proteínas Recombinantes de Fusión/genética , Proteínas Recombinantes de Fusión/metabolismo , Alineación de Secuencia , Acoplamiento Viral , Internalización del Virus
2.
Curr Issues Mol Biol ; 45(11): 9181-9214, 2023 Nov 16.
Artículo en Inglés | MEDLINE | ID: mdl-37998753

RESUMEN

Over 100 innovative in vitro transcribed (IVT)-mRNAs are presently undergoing clinical trials, with a projected substantial impact on the pharmaceutical market in the near future. Τhe idea behind this is that after the successful cellular internalization of IVT-mRNAs, they are subsequently translated into proteins with therapeutic or prophylactic relevance. Simultaneously, cancer immunotherapy employs diverse strategies to mobilize the immune system in the battle against cancer. Therefore, in this review, the fundamental principles of IVT-mRNA to its recruitment in cancer immunotherapy, are discussed and analyzed. More specifically, this review paper focuses on the development of mRNA vaccines, the exploitation of neoantigens, as well as Chimeric Antigen Receptor (CAR) T-Cells, showcasing their clinical applications and the ongoing trials for the development of next-generation immunotherapeutics. Furthermore, this study investigates the synergistic potential of combining the CAR immunotherapy and the IVT-mRNAs by introducing our research group novel, patented delivery method that utilizes the Protein Transduction Domain (PTD) technology to transduce the IVT-mRNAs encoding the CAR of interest into the Natural Killer (NK)-92 cells, highlighting the potential for enhancing the CAR NK cell potency, efficiency, and bioenergetics. While IVT-mRNA technology brings exciting progress to cancer immunotherapy, several challenges and limitations must be acknowledged, such as safety, toxicity, and delivery issues. This comprehensive exploration of IVT-mRNA technology, in line with its applications in cancer therapeutics, offers valuable insights into the opportunities and challenges in the evolving landscape of cancer immunotherapy, setting the stage for future advancements in the field.

3.
Biochem Biophys Res Commun ; 640: 32-39, 2023 01 15.
Artículo en Inglés | MEDLINE | ID: mdl-36502629

RESUMEN

Although the T helper 2 (Th2) subset is a critical player in the humoral immune response to extracellular parasites and suppression of Th1-mediated inflammation, Th2 cells have been implicated in allergic inflammatory diseases such as asthma, allergic rhinitis, and atopic dermatitis. GATA binding protein 3 (GATA3) is a primary transcription factor that mediates Th2 differentiation and secretion of Th2 cytokines, including IL-4, IL-5, and IL-13. Here, a nucleus-deliverable form of GATA3-transcription modulation domain (TMD) (ndG3-TMD) was generated using Hph-1 human protein transduction domain (PTD) to modulate the transcriptional function of endogenous GATA3 without genetic manipulation. ndG3-TMD was shown to be efficiently delivered into the cell nucleus quickly without affecting cell viability or intracellular signaling events for T cell activation. ndG3-TMD exhibited a specific inhibitory function for the endogenous GATA3-mediated transcription, such as Th2 cell differentiation and Th2-type cytokine production. Intranasal administration of ndG3-TMD significantly alleviated airway hyperresponsiveness, infiltration of immune cells, and serum IgE level in an OVA-induced mouse model of asthma. Also, Th2 cytokine secretion by the splenocytes isolated from the ndG3-TMD-treated mice substantially decreased. Our results suggest that ndG3-TMD can be a new therapeutic reagent to suppress Th2-mediated allergic diseases through intranasal delivery.


Asunto(s)
Asma , Factor de Transcripción GATA3 , Hipersensibilidad Respiratoria , Animales , Humanos , Ratones , Administración Intranasal , Asma/terapia , Núcleo Celular/metabolismo , Citocinas/metabolismo , Modelos Animales de Enfermedad , Factor de Transcripción GATA3/administración & dosificación , Factor de Transcripción GATA3/química , Ratones Endogámicos BALB C , Ovalbúmina , Hipersensibilidad Respiratoria/terapia , Células Th2
4.
Mol Ther ; 30(2): 855-867, 2022 02 02.
Artículo en Inglés | MEDLINE | ID: mdl-34547467

RESUMEN

Cell-penetrating peptides (CPPs) hold great promise for intracellular delivery of therapeutic proteins. However, endosomal entrapment of transduced cargo is a major bottleneck hampering their successful application. While developing a transducible zinc finger protein-based artificial transcription factor targeting the expression of endothelin receptor A, we identified interaction between the CPP and the endosomal membrane or endosomal entanglement as a main culprit for endosomal entrapment. To achieve endosomal disentanglement, we utilized endosome-resident proteases to sever the artificial transcription factor from its CPP upon arrival inside the endosome. Using this approach, we greatly enhanced the correct subcellular localization of the disentangled artificial transcription factor, significantly increasing its biological activity and distribution in vivo. With rational engineering of proteolytic sensitivity, we propose a new design principle for transducible therapeutic proteins, helping CPPs attain their full potential as delivery vectors for therapeutic proteins.


Asunto(s)
Péptidos de Penetración Celular , Receptores de Endotelina , Péptidos de Penetración Celular/metabolismo , Endosomas/metabolismo , Receptores de Endotelina/metabolismo , Factores de Transcripción/metabolismo
5.
Biochem J ; 479(3): 259-272, 2022 02 11.
Artículo en Inglés | MEDLINE | ID: mdl-35015082

RESUMEN

Murine cytomegalovirus protein M45 contains a RIP homotypic interaction motif (RHIM) that is sufficient to confer protection of infected cells against necroptotic cell death. Mechanistically, the N-terminal region of M45 drives rapid self-assembly into homo-oligomeric amyloid fibrils, and interacts with the endogenous RHIM domains of receptor-interacting serine/threonine protein kinases (RIPK) 1, RIPK3, Z-DNA-binding protein 1, and Toll/interleukin-1 receptor domain-containing adaptor-inducing interferon-ß. Remarkably, all four aforementioned mammalian proteins harbouring such a RHIM domain are key components of inflammatory signalling and regulated cell death (RCD) processes. Immunogenic cell death by regulated necrosis causes extensive tissue damage in a wide range of diseases, including ischaemia reperfusion injury, myocardial infarction, sepsis, stroke, and solid organ transplantation. To harness the cell death suppression properties of M45 protein in a therapeutically usable manner, we developed a synthetic peptide encompassing only the RHIM domain of M45. To trigger delivery of RHIM into target cells, we fused the transactivator protein transduction domain of human immunodeficiency virus 1 to the N-terminus of the peptide. The fused peptide could efficiently penetrate eukaryotic cells, but unexpectedly it eradicated or destroyed all tested cancer cell lines and primary cells irrespective of species without further stimulus through a necrosis-like cell death. Typical inhibitors of different forms of RCD cannot impede this process, which appears to involve a direct disruption of biomembranes. Nevertheless, our finding has potential clinical relevance; reliable induction of a necrotic form of cell death distinct from all known forms of RCD may offer a novel therapeutic approach to combat resistant tumour cells.


Asunto(s)
Productos del Gen tat/química , Productos del Gen tat/metabolismo , Dominios Proteicos , Proteínas Recombinantes de Fusión/metabolismo , Ribonucleótido Reductasas/química , Ribonucleótido Reductasas/metabolismo , Transducción de Señal/genética , Proteínas Virales/química , Proteínas Virales/metabolismo , Secuencia de Aminoácidos , Amiloide/metabolismo , Animales , Productos del Gen tat/genética , VIH-1/química , Células HT29 , Humanos , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Células 3T3 NIH , Necroptosis/genética , Proteína Serina-Treonina Quinasas de Interacción con Receptores/metabolismo , Ribonucleótido Reductasas/genética , Células U937 , Proteínas Virales/genética
6.
Proc Natl Acad Sci U S A ; 117(11): 6121-6128, 2020 03 17.
Artículo en Inglés | MEDLINE | ID: mdl-32123072

RESUMEN

Virus replication requires critical interactions between viral proteins and cellular proteins that mediate many aspects of infection, including the transport of viral genomes to the site of replication. In human papillomavirus (HPV) infection, the cellular protein complex known as retromer binds to the L2 capsid protein and sorts incoming virions into the retrograde transport pathway for trafficking to the nucleus. Here, we show that short synthetic peptides containing the HPV16 L2 retromer-binding site and a cell-penetrating sequence enter cells, sequester retromer from the incoming HPV pseudovirus, and inhibit HPV exit from the endosome, resulting in loss of viral components from cells and in a profound, dose-dependent block to infection. The peptide also inhibits cervicovaginal HPV16 pseudovirus infection in a mouse model. These results confirm the retromer-mediated model of retrograde HPV entry and validate intracellular virus trafficking as an antiviral target. More generally, inhibiting virus replication with agents that can enter cells and disrupt essential protein-protein interactions may be applicable in broad outline to many viruses.


Asunto(s)
Proteínas de la Cápside/metabolismo , Péptidos de Penetración Celular/farmacología , Papillomavirus Humano 16/efectos de los fármacos , Proteínas Oncogénicas Virales/metabolismo , Infecciones por Papillomavirus/tratamiento farmacológico , Internalización del Virus/efectos de los fármacos , Animales , Péptidos de Penetración Celular/uso terapéutico , Cuello del Útero/virología , Modelos Animales de Enfermedad , Femenino , Células HEK293 , Células HeLa , Papillomavirus Humano 16/fisiología , Humanos , Ratones , Infecciones por Papillomavirus/virología , Unión Proteica/efectos de los fármacos , Mapas de Interacción de Proteínas/efectos de los fármacos , Vagina/virología
7.
Int J Mol Sci ; 24(8)2023 Apr 16.
Artículo en Inglés | MEDLINE | ID: mdl-37108504

RESUMEN

Thioredoxin (Trx) plays a critical role in maintaining redox balance in various cells and exhibits anti-oxidative, anti-apoptotic, and anti-inflammatory effects. However, whether exogenous Trx can inhibit intracellular oxidative damage has not been investigated. In previous study, we have identified a novel Trx from the jellyfish Cyanea capillata, named CcTrx1, and confirmed its antioxidant activities in vitro. Here, we obtained a recombinant protein, PTD-CcTrx1, which is a fusion of CcTrx1 and protein transduction domain (PTD) of HIV TAT protein. The transmembrane ability and antioxidant activities of PTD-CcTrx1, and its protective effects against H2O2-induced oxidative damage in HaCaT cells were also detected. Our results revealed that PTD-CcTrx1 exhibited specific transmembrane ability and antioxidant activities, and it could significantly attenuate the intracellular oxidative stress, inhibit H2O2-induced apoptosis, and protect HaCaT cells from oxidative damage. The present study provides critical evidence for application of PTD-CcTrx1 as a novel antioxidant to treat skin oxidative damage in the future.


Asunto(s)
Péptidos de Penetración Celular , Escifozoos , Animales , Productos del Gen tat/metabolismo , Peróxido de Hidrógeno/farmacología , Peróxido de Hidrógeno/metabolismo , Péptidos de Penetración Celular/farmacología , Péptidos de Penetración Celular/metabolismo , Antioxidantes/farmacología , Antioxidantes/metabolismo , Proteínas Recombinantes de Fusión/genética , Proteínas Recombinantes de Fusión/farmacología , Proteínas Recombinantes de Fusión/metabolismo , Estrés Oxidativo , Escifozoos/metabolismo , Tiorredoxinas/genética , Tiorredoxinas/farmacología , Tiorredoxinas/química
8.
Biochem Biophys Res Commun ; 598: 26-31, 2022 04 02.
Artículo en Inglés | MEDLINE | ID: mdl-35151200

RESUMEN

Globins are heme proteins such as hemoglobin (Hb), myoglobin (Mb) and neuroglobin (Ngb), playing important roles in biological system. In addition to normal functions, zebrafish Ngb was able to penetrate cell membranes, whereas less was known for other globin members. In this study, to improve the cell-membrane-penetrating activity of globins, we used sperm whale Mb as a model protein and constructed a quadruple mutant of G5K/Q8K/A19K/V21K Mb (termed 4K Mb), by introduction of four positive charges on the protein surface, which was designed according to the amino acid alignment with that of zebrafish Ngb. Spectroscopic and crystallographic studies showed that the four positively charged Lys residues did not affect the protein structure. Cell-membrane-penetrating essay further showed that 4K Mb exhibited enhanced activity compared to that of native Mb. This study provides valuable information for the effect of distribution of charged residues on the protein structure and the cell-membrane-penetrating activity of globins. Therefore, it will guide the design of protein-based biomaterials for biological applications.


Asunto(s)
Membrana Celular/metabolismo , Mioglobina/química , Mioglobina/metabolismo , Animales , Membrana Celular/efectos de los fármacos , Supervivencia Celular/efectos de los fármacos , Dicroismo Circular , Cristalografía por Rayos X , Fluoresceína-5-Isotiocianato/química , Humanos , Lisina/química , Células MCF-7 , Mutación , Mioglobina/genética , Mioglobina/farmacocinética , Espectrofotometría Ultravioleta , Cachalote
9.
Mol Pharm ; 19(11): 3869-3876, 2022 11 07.
Artículo en Inglés | MEDLINE | ID: mdl-36036888

RESUMEN

The carboxyl groups of a protein can be esterified by reaction with a diazo compound, 2-diazo-2-(p-methylphenyl)-N,N-dimethylacetamide. This esterification enables the entry of the protein into the cytosol of a mammalian cell, where the nascent ester groups are hydrolyzed by endogenous esterases. The low aqueous solubility of the ensuing esterified protein is, however, a major practical challenge. Solubility screening revealed that ß-cyclodextrin (ß-CD) is an optimal solubilizing agent for esterified green fluorescent protein (est-GFP). Its addition can increase the recovery of est-GFP by 10-fold. α-CD, γ-CD, and cucurbit-7-uril are less effective excipients. 1H NMR titration experiments revealed that ß-CD encapsulates the hydrophobic tolyl group of ester conjugates with Ka = 321 M-1. Combining l-arginine and sucrose with ß-CD enables the nearly quantitative recovery of est-GFP. Thus, the insolubility of esterified proteins can be overcome with excipients.


Asunto(s)
Ciclodextrinas , beta-Ciclodextrinas , Animales , Solubilidad , Excipientes/química , beta-Ciclodextrinas/química , Ésteres/química , Esterificación , Ciclodextrinas/química , Mamíferos
10.
Mol Ther ; 28(8): 1858-1875, 2020 08 05.
Artículo en Inglés | MEDLINE | ID: mdl-32504545

RESUMEN

Lentivirus vectors (LVs) are efficient tools for gene transfer, but the non-specific nature of transgene integration by the viral integration machinery carries an inherent risk for genotoxicity. We modified the integration machinery of LVs and harnessed the cellular DNA double-strand break repair machinery to integrate transgenes into ribosomal DNA, a promising genomic safe-harbor site for transgenes. LVs carrying modified I-PpoI-derived homing endonuclease proteins were characterized in detail, and we found that at least 21% of all integration sites localized to ribosomal DNA when LV transduction was coupled to target DNA cleavage. In addition to the primary sequence recognized by the endonuclease, integration was also enriched in chromatin domains topologically associated with nucleoli, which contain the targeted ribosome RNA genes. Targeting of this highly repetitive region for integration was not associated with detectable DNA deletions or negative impacts on cell health in transduced primary human T cells. The modified LVs characterized here have an overall lower risk for insertional mutagenesis than regular LVs and can thus improve the safety of gene and cellular therapy.


Asunto(s)
ADN Ribosómico/genética , Endonucleasas/metabolismo , Vectores Genéticos/genética , Lentivirus/genética , Sitios de Carácter Cuantitativo , Integración Viral/genética , Secuencia de Aminoácidos , Biología Computacional/métodos , Ontología de Genes , Genes de ARNr , Ingeniería Genética , Genoma Viral , VIH-1/genética , Humanos , Mutagénesis Insercional , ARN de Transferencia/genética , Secuencias Repetitivas de Ácidos Nucleicos , Transgenes
11.
Int J Mol Sci ; 22(12)2021 Jun 17.
Artículo en Inglés | MEDLINE | ID: mdl-34204592

RESUMEN

NADH dehydrogenase (ubiquinone) Fe-S protein 8 (NDUFS8) is a nuclear-encoded core subunit of human mitochondrial complex I. Defects in NDUFS8 are associated with Leigh syndrome and encephalomyopathy. Cell-penetrating peptide derived from the HIV-1 transactivator of transcription protein (TAT) has been successfully applied as a carrier to bring fusion proteins into cells without compromising the biological function of the cargoes. In this study, we developed a TAT-mediated protein transduction system to rescue complex I deficiency caused by NDUFS8 defects. Two fusion proteins (TAT-NDUFS8 and NDUFS8-TAT) were exogenously expressed and purified from Escherichia coli for transduction of human cells. In addition, similar constructs were generated and used in transfection studies for comparison. The results showed that both exogenous TAT-NDUFS8 and NDUFS8-TAT were delivered into mitochondria and correctly processed. Interestingly, the mitochondrial import of TAT-containing NDUFS8 was independent of mitochondrial membrane potential. Treatment with TAT-NDUFS8 not only significantly improved the assembly of complex I in an NDUFS8-deficient cell line, but also partially rescued complex I functions both in the in-gel activity assay and the oxygen consumption assay. Our current findings suggest the considerable potential of applying the TAT-mediated protein transduction system for treatment of complex I deficiency.


Asunto(s)
Complejo I de Transporte de Electrón/deficiencia , Potencial de la Membrana Mitocondrial , Mitocondrias/genética , Mitocondrias/metabolismo , NADH Deshidrogenasa/metabolismo , Proteínas Recombinantes de Fusión/metabolismo , Productos del Gen tat del Virus de la Inmunodeficiencia Humana/metabolismo , Secuencia de Aminoácidos , Línea Celular , Supervivencia Celular , Células Cultivadas , Proteínas del Complejo de Cadena de Transporte de Electrón/genética , Proteínas del Complejo de Cadena de Transporte de Electrón/metabolismo , Humanos , Mitocondrias/efectos de los fármacos , NADH Deshidrogenasa/genética , Transporte de Proteínas , Interferencia de ARN , ARN Mensajero/genética , ARN Interferente Pequeño/genética , Proteínas Recombinantes de Fusión/química , Proteínas Recombinantes de Fusión/genética , Proteínas Recombinantes de Fusión/farmacología , Productos del Gen tat del Virus de la Inmunodeficiencia Humana/genética
12.
J Cell Mol Med ; 24(22): 13507-13522, 2020 11.
Artículo en Inglés | MEDLINE | ID: mdl-33079436

RESUMEN

While the bone morphogenetic protein-7 (BMP-7) is a well-known therapeutic growth factor reverting many fibrotic diseases, including peritoneal fibrosis by peritoneal dialysis (PD), soluble growth factors are largely limited in clinical applications owing to their short half-life in clinical settings. Recently, we developed a novel drug delivery model using protein transduction domains (PTD) overcoming limitation of soluble recombinant proteins, including bone morphogenetic protein-7 (BMP-7). This study aims at evaluating the therapeutic effects of PTD-BMP-7 consisted of PTD and full-length BMP-7 on epithelial-mesenchymal transition (EMT)-related fibrosis. Human peritoneal mesothelial cells (HPMCs) were then treated with TGF-ß1 or TGF-ß1 + PTD-BMP-7. Peritoneal dialysis (PD) catheters were inserted into Sprague-Dawley rats, and these rats were infused intra-peritoneally with saline, peritoneal dialysis fluid (PDF) or PDF + PTD-BMP-7. In vitro, TGF-ß1 treatment significantly increased fibronectin, type I collagen, α-SMA and Snail expression, while reducing E-cadherin expression in HPMCs (P < .001). PTD-BMP-7 treatment ameliorated TGF-ß1-induced fibronectin, type I collagen, α-SMA and Snail expression, and restored E-cadherin expression in HPMCs (P < .001). In vivo, the expressions of EMT-related molecules and the thickness of the sub-mesothelial layer were significantly increased in the peritoneum of rats treated with PDF, and these changes were significantly abrogated by the intra-peritoneal administration of PTD-BMP-7. PTD-BMP-7 treatment significantly inhibited the progression of established PD fibrosis. These findings suggest that PTD-BMP-7, as a prodrug of BMP-7, can be an effective therapeutic agent for peritoneal fibrosis in PD patients.


Asunto(s)
Proteína Morfogenética Ósea 7/administración & dosificación , Sistemas de Liberación de Medicamentos , Fibrosis Peritoneal/tratamiento farmacológico , Animales , Biomarcadores , Proteína Morfogenética Ósea 7/química , Modelos Animales de Enfermedad , Diseño de Fármacos , Transición Epitelial-Mesenquimal/efectos de los fármacos , Femenino , Humanos , Inmunohistoquímica , Microscopía Intravital , Masculino , Ratones , Fibrosis Peritoneal/etiología , Fibrosis Peritoneal/metabolismo , Fibrosis Peritoneal/patología , Ratas , Proteínas Recombinantes , Resultado del Tratamiento
13.
Artículo en Inglés | MEDLINE | ID: mdl-32070492

RESUMEN

Oligodendrocyte precursor cells (OPCs) are ideal therapeutic cells for treatment of spinal cord injuries and diseases that affect myelin. However, it is necessary to generate a cell population with a low risk of teratoma formation and oncogenesis from a patient's somatic cells. In this study, we investigated the direct reprogramming of fibroblasts to oligodendrocyte-like cells in one step with a safe non-genetic delivery method that used protein transduction. Cell morphology and the lineage-specific marker expression profile indicated that human foreskin fibroblasts (HFFs) were converted into oligodendrocyte-like cells by the application of pluripotency factors and the use of a permissible induction medium. Our data demonstrated that SOX2 was sufficient to directly drive OPC fate conversion from HFF by a genetic-free approach. Therefore, this work has provided a strategy to OPC reprogramming by a non-integrating approach for future use in disease modeling and may ultimately provide applications for patient-specific cell-based regenerative medicine.

14.
Rev Med Virol ; 29(2): e2031, 2019 03.
Artículo en Inglés | MEDLINE | ID: mdl-30609200

RESUMEN

Tat (transactivator of transcription) regulates transcription from the HIV provirus. It plays a crucial role in disease progression, supporting efficient replication of the viral genome. Tat also modulates many functions in the host genome via its interaction with chromatin and proteins. Many of the functions of Tat are associated with its basic domain rich in arginine and lysine residues. It is still unknown why the basic domain exhibits so many diverse functions. However, the highly charged basic domain, coupled with the overall structural flexibility of Tat protein itself, makes the basic domain a key player in binding to or associating with cellular and viral components. In addition, the basic domain undergoes diverse posttranslational modifications, which further expand and modulate its functions. Here, we review the current knowledge of Tat basic domain and its versatile role in the interaction between the virus and the host cell.


Asunto(s)
Infecciones por VIH/virología , VIH-1/crecimiento & desarrollo , Provirus/crecimiento & desarrollo , Productos del Gen tat del Virus de la Inmunodeficiencia Humana/metabolismo , Interacciones Huésped-Patógeno , Humanos , Dominios Proteicos , Procesamiento Proteico-Postraduccional
15.
Angew Chem Int Ed Engl ; 59(47): 21007-21015, 2020 11 16.
Artículo en Inglés | MEDLINE | ID: mdl-32777124

RESUMEN

Protein semi-synthesis inside live cells from exogenous and endogenous parts offers unique possibilities for studying proteins in their native context. Split-intein-mediated protein trans-splicing is predestined for such endeavors and has seen some successes, but a much larger variety of established split inteins and associated protocols is urgently needed. We characterized the association and splicing parameters of the Gp41-1 split intein, which favorably revealed a nanomolar affinity between the intein fragments combined with the exceptionally fast splicing rate. Following bead-loading of a chemically modified intein fragment precursor into live mammalian cells, we fluorescently labeled target proteins on their N- and C-termini with short peptide tags, thus ensuring minimal perturbation of their structure and function. In combination with a nuclear-entrapment strategy to minimize cytosolic fluorescence background, we applied our technique for super-resolution imaging and single-particle tracking of the outer mitochondrial protein Tom20 in HeLa cells.


Asunto(s)
Proteínas de Transporte de Membrana/biosíntesis , Receptores de Superficie Celular/biosíntesis , Células HeLa , Humanos , Inteínas , Proteínas de Transporte de Membrana/química , Microscopía Fluorescente , Proteínas del Complejo de Importación de Proteínas Precursoras Mitocondriales , Imagen Óptica , Biosíntesis de Proteínas , Empalme de Proteína , Receptores de Superficie Celular/química
16.
Mol Ther ; 26(1): 184-198, 2018 01 03.
Artículo en Inglés | MEDLINE | ID: mdl-28988715

RESUMEN

Type 1 diabetes (T1D) is characterized by massive destruction of insulin-producing ß cells by autoreactive T lymphocytes, arising via defective immune tolerance. Therefore, effective anti-T1D therapeutics should combine autoimmunity-preventing and insulin production-restoring properties. We constructed a cell-permeable PDX1-FOXP3-TAT fusion protein (FP) composed of two transcription factors: forkhead box P3 (FOXP3), the master regulator of differentiation and functioning of self-tolerance-promoting Tregs, and pancreatic duodenal homeobox-1 (PDX1), the crucial factor supporting ß cell development and maintenance. The FP was tested in vitro and in a non-obese diabetic mouse T1D model. In vitro, FP converted naive CD4+ T cells into a functional "Treg-like" subset, which suppressed cytokine secretion, downregulated antigen-specific responses, and curbed viability of diabetogenic effector cells. In hepatic stem-like cells, FP potentiated endocrine transdifferentiation, inducing expression of Insulin2 and other ß lineage-specific genes. In vivo, FP administration to chronically diabetic mice triggered (1) a significant elevation of insulin and C-peptide levels, (2) the formation of insulin-containing cell clusters in livers, and (3) a systemic anti-inflammatory shift (higher Foxp3+CD4+CD25+ T cell frequencies, elevated rates of IL-10-producing cells, and reduced rates of IFN-γ-secreting cells). Overall, in accordance with its design, PDX1-FOXP3-TAT FP delivered both Treg-stabilizing anti-autoimmune and de novo insulin-producing effects, proving its anti-T1D therapeutic potential.


Asunto(s)
Autoinmunidad , Diabetes Mellitus Tipo 1/inmunología , Diabetes Mellitus Tipo 1/metabolismo , Factores de Transcripción Forkhead/metabolismo , Proteínas de Homeodominio/metabolismo , Secreción de Insulina , Linfocitos T Reguladores/inmunología , Linfocitos T Reguladores/metabolismo , Transactivadores/metabolismo , Animales , Microambiente Celular/inmunología , Modelos Animales de Enfermedad , Femenino , Factores de Transcripción Forkhead/genética , Hepatocitos/metabolismo , Proteínas de Homeodominio/genética , Células Secretoras de Insulina/inmunología , Células Secretoras de Insulina/metabolismo , Ratones , Ratones Endogámicos NOD , Ratones Transgénicos , Fenotipo , Unión Proteica , Proteínas Recombinantes de Fusión , Subgrupos de Linfocitos T/inmunología , Subgrupos de Linfocitos T/metabolismo , Transactivadores/genética
17.
Molecules ; 24(5)2019 Mar 07.
Artículo en Inglés | MEDLINE | ID: mdl-30866424

RESUMEN

Cell-penetrating-peptides (CPPs) are small amino-acid sequences characterized by their ability to cross cellular membranes. They can transport various bioactive cargos inside cells including nucleic acids, large proteins, and other chemical compounds. Since 1988, natural and synthetic CPPs have been developed for applications ranging from fundamental to applied biology (cell imaging, gene editing, therapeutics delivery). In recent years, a great number of studies reported the potential of CPPs as carriers for the treatment of various diseases. Apart from a good efficacy due to a rapid and potent delivery, a crucial advantage of CPP-based therapies is the peptides low toxicity compared to most drug carriers. On the other hand, they are quite unstable and lack specificity. Higher specificity can be obtained using a cell-specific CPP to transport the therapeutic agent or using a non-specific CPP to transport a cargo with a targeted activity. CPP-cargo complexes can also be conjugated to another moiety that brings cell- or tissue-specificity. Studies based on all these approaches are showing promising results. Here, we focus on recent advances in the potential usage of CPPs in the context of cancer therapy, with a particular interest in CPP-mediated delivery of anti-tumoral proteins.


Asunto(s)
Péptidos de Penetración Celular/química , Portadores de Fármacos/química , Neoplasias/tratamiento farmacológico , Animales , Antineoplásicos/química , Antineoplásicos/uso terapéutico , Péptidos de Penetración Celular/efectos adversos , Portadores de Fármacos/efectos adversos , Humanos , Especificidad de Órganos
18.
Molecules ; 24(4)2019 Feb 20.
Artículo en Inglés | MEDLINE | ID: mdl-30791543

RESUMEN

Patients with uncontrolled diabetes are susceptible to implant failure due to impaired bone metabolism. Hypoxia-inducible factor 1α (HIF-1α), a transcription factor that is up-regulated in response to reduced oxygen during bone repair, is known to mediate angiogenesis and osteogenesis. However, its function is inhibited under hyperglycemic conditions in diabetic patients. This study thus evaluates the effects of exogenous HIF-1α on bone formation around implants by applying HIF-1α to diabetic mice and normal mice via a protein transduction domain (PTD)-mediated DNA delivery system. Implants were placed in the both femurs of diabetic and normal mice. HIF-1α and placebo gels were injected to implant sites of the right and left femurs, respectively. We found that bone-to-implant contact (BIC) and bone volume (BV) were significantly greater in the HIF-1α treated group than placebo in diabetic mice (p < 0.05). Bioinformatic analysis showed that diabetic mice had 216 differentially expressed genes (DEGs) and 21 target genes. Among the target genes, NOS2, GPNMB, CCL2, CCL5, CXCL16, and TRIM63 were found to be associated with bone formation. Based on these results, we conclude that local administration of HIF-1α via PTD may boost bone formation around the implant and induce gene expression more favorable to bone formation in diabetic mice.


Asunto(s)
Proteínas de Unión al ADN/metabolismo , Implantes Dentales , Diabetes Mellitus Experimental , Subunidad alfa del Factor 1 Inducible por Hipoxia/farmacología , Osteogénesis/efectos de los fármacos , Animales , Huesos/efectos de los fármacos , Huesos/metabolismo , Huesos/patología , Biología Computacional/métodos , Perfilación de la Expresión Génica , Regulación de la Expresión Génica/efectos de los fármacos , Inmunohistoquímica , Ratones , Neovascularización Fisiológica/efectos de los fármacos , Neovascularización Fisiológica/genética , Osteogénesis/genética , Permeabilidad
19.
J Cell Sci ; 129(5): 893-7, 2016 Mar 01.
Artículo en Inglés | MEDLINE | ID: mdl-26801086

RESUMEN

The use of cell-penetrating peptides (CPPs) as biomolecular delivery vehicles holds great promise for therapeutic and other applications, but development has been stymied by poor delivery and lack of endosomal escape. We have developed a CPP-adaptor system capable of efficient intracellular delivery and endosomal escape of user-defined protein cargos. The cell-penetrating sequence of HIV transactivator of transcription was fused to calmodulin, which binds with subnanomolar affinity to proteins containing a calmodulin binding site. Our strategy has tremendous advantage over prior CPP technologies because it utilizes high-affinity non-covalent, but reversible coupling between CPP and cargo. Three different cargo proteins fused to a calmodulin binding sequence were delivered to the cytoplasm of eukaryotic cells and released, demonstrating the feasibility of numerous applications in living cells including alteration of signaling pathways and gene expression.


Asunto(s)
Péptidos de Penetración Celular/metabolismo , Endosomas/metabolismo , Mioglobina/metabolismo , Proteínas Recombinantes de Fusión/metabolismo , Calmodulina/química , Péptidos de Penetración Celular/química , Productos del Gen tat/química , Células HEK293 , Humanos , Transporte de Proteínas , Proteínas Recombinantes de Fusión/química
20.
J Cell Sci ; 129(5): 912-20, 2016 Mar 01.
Artículo en Inglés | MEDLINE | ID: mdl-26795560

RESUMEN

A comprehensive analysis of the molecular network of cellular factors establishing and maintaining pluripotency as well as self renewal of pluripotent stem cells is key for further progress in understanding basic stem cell biology. Nanog is necessary for the natural induction of pluripotency in early mammalian development but dispensable for both its maintenance and its artificial induction. To gain further insight into the molecular activity of Nanog, we analyzed the outcomes of Nanog gain-of-function in various cell models employing a recently developed biologically active recombinant cell-permeant protein, Nanog-TAT. We found that Nanog enhances the proliferation of both NIH 3T3 and primary fibroblast cells. Nanog transduction into primary fibroblasts results in suppression of senescence-associated ß-galactosidase activity. Investigation of cell cycle factors revealed that transient activation of Nanog correlates with consistent downregulation of the cell cycle inhibitor p27(KIP1) (also known as CDKN1B). By performing chromatin immunoprecipitation analysis, we confirmed bona fide Nanog-binding sites upstream of the p27(KIP1) gene, establishing a direct link between physical occupancy and functional regulation. Our data demonstrates that Nanog enhances proliferation of fibroblasts through transcriptional regulation of cell cycle inhibitor p27 gene.


Asunto(s)
Senescencia Celular , Inhibidor p27 de las Quinasas Dependientes de la Ciclina/genética , Proteína Homeótica Nanog/fisiología , Animales , Sitios de Unión , Adhesión Celular , Proliferación Celular , Inhibidor p27 de las Quinasas Dependientes de la Ciclina/metabolismo , Regulación hacia Abajo , Fibroblastos/fisiología , Expresión Génica , Silenciador del Gen , Sitios Genéticos , Humanos , Ratones , Células 3T3 NIH
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA