Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 16 de 16
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Biomacromolecules ; 24(8): 3666-3679, 2023 08 14.
Artículo en Inglés | MEDLINE | ID: mdl-37507377

RESUMEN

Survivin, a well-known member of the inhibitor of apoptosis protein family, is upregulated in many cancer cells, which is associated with resistance to chemotherapy. To circumvent this, inhibitors are currently being developed to interfere with the nuclear export of survivin by targeting its protein-protein interaction (PPI) with the export receptor CRM1. Here, we combine for the first time a supramolecular tweezer motif, sequence-defined macromolecular scaffolds, and ultrasmall Au nanoparticles (us-AuNPs) to tailor a high avidity inhibitor targeting the survivin-CRM1 interaction. A series of biophysical and biochemical experiments, including surface plasmon resonance measurements and their multivalent evaluation by EVILFIT, reveal that for divalent macromolecular constructs with increasing linker distance, the longest linkers show superior affinity, slower dissociation, as well as more efficient PPI inhibition. As a drawback, these macromolecular tweezer conjugates do not enter cells, a critical feature for potential applications. The problem is solved by immobilizing the tweezer conjugates onto us-AuNPs, which enables efficient transport into HeLa cells. On the nanoparticles, the tweezer valency rises from 2 to 16 and produces a 100-fold avidity increase. The hierarchical combination of different scaffolds and controlled multivalent presentation of supramolecular binders was the key to the development of highly efficient survivin-CRM1 competitors. This concept may also be useful for other PPIs.


Asunto(s)
Oro , Nanopartículas del Metal , Humanos , Survivin , Células HeLa , Proteínas Inhibidoras de la Apoptosis/metabolismo , Sustancias Macromoleculares/metabolismo , Transporte Activo de Núcleo Celular , Núcleo Celular/metabolismo
2.
Small ; 19(33): e2300871, 2023 08.
Artículo en Inglés | MEDLINE | ID: mdl-37035950

RESUMEN

Nanobodies are highly affine binders, often used to track disease-relevant proteins inside cells. However, they often fail to interfere with pathobiological functions, required for their clinical exploitation. Here, a nanobody targeting the disease-relevant apoptosis inhibitor and mitosis regulator Survivin (SuN) is utilized. Survivin's multifaceted functions are regulated by an interplay of dynamic cellular localization, dimerization, and protein-protein interactions. However, as Survivin harbors no classical "druggable" binding pocket, one must aim at blocking extended protein surface areas. Comprehensive experimental evidence demonstrates that intracellular expression of SuN allows to track Survivin at low nanomolar concentrations but failed to inhibit its biological functions. Small angle X-ray scattering of the Survivin-SuN complex locates the proposed interaction interface between the C-terminus and the globular domain, as such not blocking any pivotal interaction. By clicking multiple SuN to ultrasmall (2 nm) gold nanoparticles (SuN-N), not only intracellular uptake is enabled, but additionally, Survivin crosslinking and interference with mitotic progression in living cells are also enabled. In sum, it is demonstrated that coupling of nanobodies to nanosized scaffolds can be universally applicable to improve their function and therapeutic applicability.


Asunto(s)
Nanopartículas del Metal , Anticuerpos de Dominio Único , Survivin , Oro , Proteínas Asociadas a Microtúbulos/metabolismo , Proteínas Inhibidoras de la Apoptosis/metabolismo , Proteínas de Neoplasias/metabolismo , Apoptosis
3.
Nanoscale Adv ; 4(21): 4502-4516, 2022 Oct 25.
Artículo en Inglés | MEDLINE | ID: mdl-36341304

RESUMEN

Azide-terminated ultrasmall gold nanoparticles (2 nm gold core) were covalently functionalized with alkyne-terminated small-interfering siRNA duplexes by copper-catalyzed azide-alkyne cycloaddition (CuAAC; click chemistry). The nanoparticle core was visualized by transmission electron microscopy. The number of attached siRNA molecules per nanoparticle was determined by a combination of atomic absorption spectroscopy (AAS; for gold) and UV-Vis spectroscopy (for siRNA). Each nanoparticle carried between 6 and 10 siRNA duplex molecules which corresponds to a weight ratio of siRNA to gold of about 2.2 : 1. Different kinds of siRNA were conjugated to the nanoparticles, depending on the gene to be silenced. In general, the nanoparticles were readily taken up by cells and highly efficient in gene silencing, in contrast to free siRNA. This was demonstrated in HeLa-eGFP cells (silencing of eGFP) and in LPS-stimulated macrophages (silencing of NF-κB). Furthermore, we demonstrated that nanoparticles carrying antiviral siRNA potently inhibited the replication of Herpes simplex virus 2 (HSV-2) in vitro. This highlights the strong potential of siRNA-functionalized ultrasmall gold nanoparticles in a broad spectrum of applications, including gene silencing and treatment of viral infections, combined with a minimal dose of gold.

4.
Small ; 18(31): e2201167, 2022 08.
Artículo en Inglés | MEDLINE | ID: mdl-35712760

RESUMEN

Ultrasmall gold nanoparticles (2 nm) easily penetrate the membranes of intestinal murine epithelial cells (MODE-K) and colorectal cancer cells (CT-26). They are also taken up by 3D spheroids (400 µm) of these cell types and primary gut organoids (500 µm). In contrast, dissolved dyes are not taken up by any of these cells or 3D structures. The distribution of fluorescent ultrasmall gold nanoparticles inside cells, spheroids, and gut organoids is examined by confocal laser scanning microscopy. Nanoparticles conjugated with the cytostatic drug doxorubicin and a fluorescent dye exhibit significantly greater cytotoxicity toward CT-26 tumor spheroids than equally concentrated dissolved doxorubicin, probably because they enter the interior of a spheroid much more easily than dissolved doxorubicin. Comprehensive analyses show that the cellular uptake of ultrasmall gold nanoparticles occurs by different endocytosis pathways.


Asunto(s)
Nanopartículas del Metal , Neoplasias , Animales , Doxorrubicina/química , Doxorrubicina/farmacología , Oro , Humanos , Ratones , Esferoides Celulares
5.
Front Immunol ; 13: 830938, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-35251021

RESUMEN

Calcium phosphate nanoparticles (CaP-NPs) are biodegradable carriers that can be functionalized with biologically active molecules. As such, they are potential candidates for delivery of therapeutic molecules in cancer therapies. In this context, it is important to explore whether CaP-NPs impair the natural or therapy-induced immune cell activity against cancer cells. Therefore, in this study, we have investigated the effects of different CaP-NPs on the anti-tumor activity of natural killer (NK) cells using different ovarian cancer (OC) cell line models. We explored these interactions in coculture systems consisting of NK cells, OC cells, CaP-NPs, and therapeutic Cetuximab antibodies (anti-EGFR, ADCC-inducing antibody). Our experiments revealed that aggregated CaP-NPs can serve as artificial targets, which activate NK cell degranulation and impair ADCC directed against tumor targets. However, when CaP-NPs were properly dissolved by sonication, they did not cause substantial activation. CaP-NPs with SiO2-SH-shell induced some activation of NK cells that was not observed with polyethyleneimine-coated CaP-NPs. Addition of CaP-NPs to NK killing assays did not impair conjugation of NK with OC and subsequent tumor cytolytic NK degranulation. Therapeutic antibody coupled to functionalized CaP-NPs maintained substantial levels of antibody-dependent cellular cytotoxic activity. Our study provides a cell biological basis for the application of functionalized CaP-NPs in immunologic anti-cancer therapies.


Asunto(s)
Nanopartículas , Neoplasias Ováricas , Citotoxicidad Celular Dependiente de Anticuerpos , Fosfatos de Calcio/farmacología , Carcinoma Epitelial de Ovario/metabolismo , Línea Celular Tumoral , Receptores ErbB/metabolismo , Femenino , Humanos , Células Asesinas Naturales , Dióxido de Silicio/farmacología
6.
Molecules ; 27(6)2022 Mar 09.
Artículo en Inglés | MEDLINE | ID: mdl-35335152

RESUMEN

Three different alkyne-terminated aggregation-induced emission molecules based on a para-substituted di-thioether were attached to the surface of ultrasmall gold nanoparticles (2 nm) by copper-catalyzed azide-alkyne cycloaddition (click chemistry). They showed a strong fluorescence and were well water-dispersible, in contrast to the dissolved AIE molecules. The AIE-loaded nanoparticles were not cytotoxic and easily penetrated the membrane of HeLa cells, paving the way for an intracellular application of AIE molecules, e.g., for imaging.


Asunto(s)
Oro , Nanopartículas del Metal , Azidas/química , Química Clic , Oro/química , Células HeLa , Humanos , Nanopartículas del Metal/química
7.
mBio ; 12(1)2021 02 02.
Artículo en Inglés | MEDLINE | ID: mdl-33531395

RESUMEN

PD-1-targeted therapies have shown modest antiviral effects in preclinical models of chronic viral infection. Thus, novel therapy protocols are necessary to enhance T cell immunity and viral control to overcome T cell dysfunction and immunosuppression. Here, we demonstrate that nanoparticle-based therapeutic vaccination improved PD-1-targeted therapy during chronic infection with Friend retrovirus (FV). Prevention of inhibitory signals by blocking PD-L1 in combination with therapeutic vaccination with nanoparticles containing the microbial compound CpG and a CD8+ T cell Gag epitope peptide synergistically enhanced functional virus-specific CD8+ T cell responses and improved viral clearance. We characterized the CD8+ T cell populations that were affected by this combination therapy, demonstrating that new effector cells were generated and that exhausted CD8+ T cells were reactivated at the same time. While CD8+ T cells with high PD-1 (PD-1hi) expression turned into a large population of granzyme B-expressing CD8+ T cells after combination therapy, CXCR5-expressing follicular cytotoxic CD8+ T cells also expanded to a high degree. Thus, our study describes a very efficient approach to enhance virus control and may help us to understand the mechanisms of combination immunotherapy reactivating CD8+ T cell immunity. A better understanding of CD8+ T cell immunity during combination therapy will be important for developing efficient checkpoint therapies against chronic viral infections and cancer.IMPORTANCE Despite significant efforts, vaccines are not yet available for every infectious pathogen, and the search for a protective approach to prevent the establishment of chronic infections, i.e., with HIV, continues. Immune checkpoint therapies targeting inhibitory receptors, such as PD-1, have shown impressive results against solid tumors. However, immune checkpoint therapies have not yet been licensed to treat chronic viral infections, since a blockade of inhibitory receptors alone provides only limited benefit, as demonstrated in preclinical models of chronic viral infection. Thus, there is a high interest in the development of potent combination immunotherapies. Here, we tested whether the combination of a PD-L1 blockade and therapeutic vaccination with functionalized nanoparticles is a potent therapy during chronic Friend retrovirus infection. We demonstrate that the combination therapy induced a synergistic reinvigoration of the exhausted virus-specific CD8+ T cell immunity. Taken together, our results provide further information on how to improve PD-1-targeted therapies during chronic viral infection and cancer.


Asunto(s)
Linfocitos T CD8-positivos/inmunología , Virus de la Leucemia Murina de Friend/inmunología , Inhibidores de Puntos de Control Inmunológico/uso terapéutico , Activación de Linfocitos , Infecciones por Retroviridae/terapia , Vacunación , Animales , Células Cultivadas , Femenino , Ratones , Ratones Endogámicos C57BL , Receptores CXCR5/análisis , Infecciones por Retroviridae/inmunología
8.
Acta Biomater ; 111: 406-417, 2020 07 15.
Artículo en Inglés | MEDLINE | ID: mdl-32439614

RESUMEN

Calcium phosphate nanoparticles (100 nm) were fluorescently labelled with poly(ethyleneimine) (PEIATTO490LS; red fluorescence). They were loaded with a Tandem fusion protein consisting of mRFP1-eGFP (red and green fluorescence in the same molecule)that acts as smart biological pH sensor to trace nanoparticles inside cells. Its fluorescence is also coupled to the structural integrity of the protein, i.e. it is also a label for a successful delivery of a functional protein into the cell. At pH 7.4, the fluorescence of both proteins (red and green) is detectable. At a pH of 4.5-5 inside the lysosomes, the green fluorescence is quenched due to the protonation of the eGFP chromophore, but the pH-independent red fluorescence of mRFP1 remains. The nanoparticles were taken up by cells (cell lines: HeLa, Caco-2 and A549) via endocytic pathways and then directed to lysosomes. Time-resolved confocal laser scanning microscopy confirmed mRFP1 and nanoparticles co-localizing with lysosomes. The fluorescence of eGFP was only detectable outside lysosomes, i.e. most likely inside early endosomes or at the cell membrane during the uptake, indicating the neutral pH at these locations. The Tandem fusion protein provides a versatile platform to follow the intracellular pathway of bioactive nanocarriers, e.g. therapeutic proteins. The transfection with a Tandem-encoding plasmid by calcium phosphate nanoparticles led to an even intracellular protein distribution in cytosol and nucleoplasm, i.e. very different from direct protein uptake. Neither dissolved protein nor dissolved plasmid DNA were taken up by the cells, underscoring the necessity for a suitable carrier like a nanoparticle. STATEMENT OF SIGNIFICANCE: A pH-sensitive protein ("tandem") was used to follow the pathway of calcium phosphate nanoparticles. This protein consists of a pH-sensitive fluorophore (eGFP; green) and a pH-independent fluorophore (mRFP1; red). This permits to follow the pathway of a nanoparticle inside a cell. At a low pH inside an endolysosome, the green fluorescence vanishes but the red fluorescence persists. This is also a very useful model for the delivery of therapeutic proteins into cells. The delivery by nanoparticles was compared with the protein expression after cell transfection with plasmid DNA encoding for the tandem protein. High-resolution image analysis gave quantitative data on the intracellular protein distribution.


Asunto(s)
Nanopartículas , Células CACO-2 , Fosfatos de Calcio , Proteínas Fluorescentes Verdes/genética , Humanos , Concentración de Iones de Hidrógeno , Transfección
9.
Acta Biomater ; 109: 244-253, 2020 06.
Artículo en Inglés | MEDLINE | ID: mdl-32251787

RESUMEN

Calcium phosphate nanoparticles were covalently surface-functionalized with the ligand DOTA and loaded with the radioisotope 68Ga. The biodistribution of such 68Ga-labelled nanoparticles was followed in vivo in mice by positron emission tomography in combination with computer tomography (PET-CT). The biodistribution of 68Ga-labelled nanoparticles was compared for different application routes: intravenous, intramuscular, intratumoral, and into soft tissue. The particle distribution was measured in vivo by PET-CT after 5 min, 15 min, 30 min, 1 h, 2 h, and 4 h, and ex vivo after 5 h. After intravenous injection (tail vein), the nanoparticles rapidly entered the lungs with later redistribution into liver and spleen. The nanoparticles remained mostly at the injection site following intramuscular, intratumoral, or soft tissue application, with less than 10 percent being mobilized into the blood stream. STATEMENT OF SIGNIFICANCE: The in vivo biodistribution of DOTA-terminated calcium phosphate nanoparticles was followed by PET/CT. To our knowledge, this is the first study of this kind. Four different application routes of clinical relevance were pursued: Intravascular, intramuscular, intratumoral, and into soft tissue. Given the high importance of calcium phosphate as biomaterial and for nanoparticular drug delivery and immunization, this is most important to assess the biofate of calcium phosphate nanoparticles for therapeutic application and also judge biodistribution of nanoscopic calcium phosphate ceramics, including debris from endoprostheses and related implants.


Asunto(s)
Fosfatos de Calcio/farmacocinética , Nanopartículas/química , Neoplasias/metabolismo , Animales , Fosfatos de Calcio/administración & dosificación , Fosfatos de Calcio/química , Línea Celular Tumoral , Radioisótopos de Galio/administración & dosificación , Radioisótopos de Galio/química , Radioisótopos de Galio/farmacocinética , Compuestos Heterocíclicos con 1 Anillo/administración & dosificación , Compuestos Heterocíclicos con 1 Anillo/química , Compuestos Heterocíclicos con 1 Anillo/farmacocinética , Humanos , Inyecciones Intramusculares , Ratones Endogámicos BALB C , Nanopartículas/administración & dosificación , Neoplasias/diagnóstico por imagen , Tomografía Computarizada por Tomografía de Emisión de Positrones
10.
Vaccines (Basel) ; 8(1)2020 Mar 01.
Artículo en Inglés | MEDLINE | ID: mdl-32121590

RESUMEN

The ability of vaccines to induce T cell responses is crucial for preventing diseases caused by viruses. Nanoparticles (NPs) are considered to be efficient tools for the initiation of potent immune responses. Calcium phosphate (CaP) NPs are a class of biodegradable nanocarriers that are able to deliver immune activating molecules across physiological barriers. Therefore, the aim of this study was to assess whether Toll-like receptor (TLR) ligand and viral antigen functionalized CaP NPs are capable of inducing efficient maturation of human antigen presenting cells (APC). To achieve this, we generated primary human dendritic cells (DCs) and stimulated them with CpG or poly(I:C) functionalized CaP NPs. DCs were profoundly stronger when activated upon NP stimulation compared to treatment with soluble TLR ligands. This is indicated by increased levels of costimulatory molecules and the secretion of proinflammatory cytokines. Consequently, coculture of NP-stimulated APCs with CD8+ T cells resulted in a significant expansion of virus-specific T cells. In summary, our data suggest that functionalized CaP NPs are a suitable tool for activating human virus-specific CD8+ T cells and may represent an excellent vaccine delivery system.

11.
Mol Cancer Ther ; 18(6): 1069-1080, 2019 06.
Artículo en Inglés | MEDLINE | ID: mdl-30962317

RESUMEN

Recognition of immunoactive oligonucleotides by the immune system, such as Toll-like receptor ligand CpG, leads to increased antibody and T-cell responses. Systemic application often results in unwanted generalized nonantigen-specific activation of the immune system. Nanoparticles are ideal carriers for small and large molecules. Recently, we have demonstrated that calcium phosphate (CaP) nanoparticles functionalized with CpG, and viral antigens are able to induce specific T-cell immunity that protects mice against viral infection and efficiently reactivates the exhausted CD8+ T-cell compartment during chronic retroviral infection. Therefore, CaP nanoparticles are promising vaccine vehicles for therapeutic applications. In this study, we investigated the therapeutic potential use of these nanoparticles in a murine xenograft colorectal cancer model. Therapeutic vaccination with CaP nanoparticles functionalized with CpG and tumor model antigens increased the frequencies of cytotoxic CD8+ T cells in the tumor in a type I interferon-dependent manner. This was accompanied with significantly repressed tumor growth in contrast to the systemic administration of soluble CpG and antigens. Combination therapy of CaP nanoparticles and immune checkpoint blocker against PD-L1 further enhanced the cytotoxic CD8+ T-cell response and eradicated the tumors. Strikingly, vaccination with CaP nanoparticles functionalized with CpG and a primary tumor cell lysate was also sufficient to control the tumor growth. In conclusion, our results represent a translational approach for the use of CaP nanoparticles as a potent cancer vaccine vehicle.


Asunto(s)
Traslado Adoptivo/métodos , Antígenos de Neoplasias/química , Vacunas contra el Cáncer/uso terapéutico , Neoplasias del Colon/terapia , Sistemas de Liberación de Medicamentos/métodos , Nanopartículas/química , Péptidos/química , Aloinjertos , Animales , Anticuerpos Monoclonales/farmacología , Antígenos Virales/genética , Antígeno B7-H1/antagonistas & inhibidores , Antígeno B7-H1/inmunología , Linfocitos T CD8-positivos/inmunología , Fosfatos de Calcio/química , Línea Celular Tumoral , Neoplasias del Colon/patología , Islas de CpG , Modelos Animales de Enfermedad , Hemaglutininas/genética , Interferón Tipo I/metabolismo , Activación de Linfocitos/efectos de los fármacos , Ratones , Ratones Endogámicos BALB C , Ratones Transgénicos , Transfección
12.
Front Immunol ; 9: 614, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-29740425

RESUMEN

T cell dysfunction and immunosuppression are characteristic for chronic viral infections and contribute to viral persistence. Overcoming these burdens is the goal of new therapeutic strategies to cure chronic infectious diseases. We recently described that therapeutic vaccination of chronic retrovirus infected mice with a calcium phosphate (CaP) nanoparticle (NP)-based vaccine carrier, functionalized with CpG and viral peptides is able to efficiently reactivate the CD8+ T cell response and improve the eradication of virus infected cells. However, the mechanisms underlying this effect were largely unclear. While type I interferons (IFNs I) are considered to drive T cell exhaustion by persistent immune activation during chronic viral infection, we here describe an indispensable role of IFN I induced by therapeutic vaccination to efficiently reinforce cytotoxic CD8+ T cells (CTL) and improve control of chronic retroviral infection. The induction of IFN I is CpG dependent and leads to significant IFN signaling indicated by upregulation of IFN stimulated genes. By vaccinating chronically retrovirus-infected mice lacking the IFN I receptor (IFNAR-/-) or by blocking IFN I signaling in vivo during therapeutic vaccination, we demonstrate that IFN I signaling is necessary to drive full reactivation of CTLs. Surprisingly, we also identified an impaired suppressive capability of regulatory T cells in the presence of IFNα, which implicates an important role for vaccine-induced IFNα in the regulation of the T cell response during chronic retroviral infection. Our data suggest that inducing IFN I signaling in conjunction with the presentation of viral antigens can reactivate immune functions and reduce viral loads in chronic infections. Therefore, we propose CaP NPs as potential therapeutic tool to treat chronic infections.


Asunto(s)
Linfocitos T CD8-positivos/inmunología , Interferón Tipo I/metabolismo , Nanopartículas/administración & dosificación , Infecciones por Retroviridae/inmunología , Retroviridae/fisiología , Vacunas Virales/inmunología , Animales , Fosfatos de Calcio/química , Línea Celular , Citotoxicidad Inmunológica , Humanos , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Nanopartículas/química , Receptor de Interferón alfa y beta/genética , Transducción de Señal , Vacunación
13.
ACS Chem Biol ; 12(12): 2958-2964, 2017 12 15.
Artículo en Inglés | MEDLINE | ID: mdl-29058870

RESUMEN

RNAs are involved in interaction networks with other biomolecules and are crucial for proper cell function. Yet their biochemical analysis remains challenging. For Förster Resonance Energy Transfer (FRET), a common tool to study such interaction networks, two interacting molecules have to be fluorescently labeled. "Spinach" is a genetically encodable RNA aptamer that starts to fluoresce upon binding of an organic molecule. Therefore, it is a biological fluorophore tag for RNAs. However, spinach has never been used in a FRET assembly before. Here, we describe how spinach is quenched when close to acceptors. We used RNA-DNA hybridization to bring quenchers or red organic dyes in close proximity to spinach. Furthermore, we investigate RNA-protein interactions quantitatively on the example of Pseudomonas aeruginosa phage coat protein 7 (PP7) and its interacting pp7-RNA. We utilize spinach quenching as a fully genetically encodable system even under lysate conditions. Therefore, this work represents a direct method to analyze RNA-protein interactions by quenching the spinach aptamer.


Asunto(s)
Proteínas de Plantas/metabolismo , ARN de Planta/metabolismo , Spinacia oleracea/metabolismo , Proteínas de la Cápside/química , Proteínas de la Cápside/metabolismo , Fluorescencia , Transferencia Resonante de Energía de Fluorescencia , Regulación de la Expresión Génica de las Plantas/fisiología , Modelos Moleculares , Proteínas de Plantas/genética , Unión Proteica , Conformación Proteica , Fagos Pseudomonas/metabolismo , ARN de Planta/química
14.
PLoS Pathog ; 13(9): e1006649, 2017 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-28938014

RESUMEN

Inflammatory bowel diseases (IBD) are chronic inflammatory disorders of the gastrointestinal tract, strongly associated with an increased risk of colorectal cancer development. Parasitic infections caused by helminths have been shown to modulate the host's immune response by releasing immunomodulatory molecules and inducing regulatory T cells (Tregs). This immunosuppressive state provoked in the host has been considered as a novel and promising approach to treat IBD patients and alleviate acute intestinal inflammation. On the contrary, specific parasite infections are well known to be directly linked to carcinogenesis. Whether a helminth infection interferes with the development of colitis-associated colon cancer (CAC) is not yet known. In the present study, we demonstrate that the treatment of mice with the intestinal helminth Heligmosomoides polygyrus at the onset of tumor progression in a mouse model of CAC does not alter tumor growth and distribution. In contrast, H. polygyrus infection in the early inflammatory phase of CAC strengthens the inflammatory response and significantly boosts tumor development. Here, H. polygyrus infection was accompanied by long-lasting alterations in the colonic immune cell compartment, with reduced frequencies of colonic CD8+ effector T cells. Moreover, H. polygyrus infection in the course of dextran sulfate sodium (DSS) mediated colitis significantly exacerbates intestinal inflammation by amplifying the release of colonic IL-6 and CXCL1. Thus, our findings indicate that the therapeutic application of helminths during CAC might have tumor-promoting effects and therefore should be well-considered.


Asunto(s)
Colitis/complicaciones , Neoplasias del Colon/etiología , Helmintiasis/complicaciones , Parasitosis Intestinales/complicaciones , Infecciones por Strongylida/complicaciones , Animales , Carcinogénesis/inmunología , Modelos Animales de Enfermedad , Femenino , Citometría de Flujo , Helmintiasis/inmunología , Parasitosis Intestinales/inmunología , Ratones , Ratones Endogámicos BALB C , Nematospiroides dubius , Infecciones por Strongylida/inmunología
15.
Nanomedicine ; 13(8): 2395-2403, 2017 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-28800875

RESUMEN

The local interference of cytokine signaling mediated by siRNA-loaded nanoparticles might be a promising new therapeutic approach to dampen inflammation during pulmonary diseases. For the local therapeutic treatment of pulmonary inflammation, we produced multi-shell nanoparticles consisting of a calcium phosphate core, coated with siRNAs directed against pro-inflammatory mediators, encapsulated into poly(lactic-co-glycolic acid), and coated with a final outer layer of polyethylenimine. Nasal instillation of nanoparticles loaded with a mixture of siRNAs directed against different cytokines to mice suffering from TH1 cell-mediated lung inflammation, or of siRNA directed against NS-1 in an influenza infection model led to a significant reduction of target gene expression which was accompanied by distinct amelioration of lung inflammation in both models. Thus, this study provides strong evidence that the specific and local modulation of the inflammatory response by CaP/PLGA nanoparticle-mediated siRNA delivery could be a promising approach for the treatment of inflammatory disorders of the lung.


Asunto(s)
Fosfatos de Calcio/química , Nanopartículas/química , Neumonía/terapia , ARN Interferente Pequeño/uso terapéutico , Tratamiento con ARN de Interferencia/métodos , Animales , Células Cultivadas , Citocinas/genética , Humanos , Ácido Láctico/química , Ratones Endogámicos BALB C , Neumonía/genética , Neumonía/patología , Polietileneimina/química , Ácido Poliglicólico/química , Copolímero de Ácido Poliláctico-Ácido Poliglicólico , ARN Interferente Pequeño/administración & dosificación , ARN Interferente Pequeño/genética
16.
Acc Chem Res ; 50(6): 1383-1390, 2017 06 20.
Artículo en Inglés | MEDLINE | ID: mdl-28480714

RESUMEN

Research on nanoparticles has evolved into a major topic in chemistry. Concerning biomedical research, nanoparticles have decisively entered the field, creating the area of nanomedicine where nanoparticles are used for drug delivery, imaging, and tumor targeting. Besides these functions, scientists have addressed the specific ways in which nanoparticles interact with biomolecules, with proteins being the most prominent example. Depending on their size, shape, charge, and surface functionality, specifically designed nanoparticles can interact with proteins in a defined way. Proteins have typical dimensions of 5-20 nm. Ultrasmall nanoparticles (size about 1-2 nm) can address specific epitopes on the surface of a protein, for example, an active center of an enzyme. Medium-sized nanoparticles (size about 5 nm) can interact with proteins on a 1:1 basis. Large nanoparticles (above 20 nm) are big in comparison to many proteins and therefore are at the borderline to a two-dimensional surface onto which a protein will adsorb. This can still lead to irreversible structural changes in a protein and a subsequent loss of function. However, as most cells readily take up nanoparticles of almost any size, it is easily possible to use nanoparticles as transporters for proteins into a cell, for example, to address an internal receptor. Much work has been dedicated to this approach, but it is constrained by two processes that can only be observed in living cells or organisms. First, nanoparticles are usually taken up by endocytosis and are delivered into an intracellular endosome. After fusion with a lysosome, a degradation or denaturation of the protein cargo by the acidic environment or by proteases may occur before it can enter the cytoplasm. Second, nanoparticles are rapidly coated with proteins upon contact with biological media like blood. This so-called protein corona influences the contact with other proteins, cells, or tissue and may prevent the desired interaction. Essentially, these effects cannot be understood in purely chemical approaches but require biological environments and systems because the underlying processes are simply too complicated to be modeled in nonbiological systems. The area of nanoparticle-protein interactions strongly relies on different approaches: Synthetic chemistry is involved to prepare, stabilize, and functionalize nanoparticles. High-end analytical chemistry is required to understand the nature of a nanoparticle surface and the steps of its interaction with proteins. Concepts from supramolecular chemistry help to understand the complex noncovalent interactions between the surfaces of proteins and nanoparticles. Protein chemistry and biophysical chemistry are required to understand the behavior of a protein in contact with a nanoparticle. Finally, all chemical concepts must live up to the "biological reality", first in cell culture experiments in vitro and finally in animal or human experiments in vivo, to open new therapies in the 21st century. This interdisciplinary approach makes the field highly exciting but also highly demanding for chemists who, however, have to learn to understand the language of other areas.


Asunto(s)
Nanopartículas/química , Proteínas/química , Animales , Humanos , Lisosomas/química , Sustancias Macromoleculares/química , Tamaño de la Partícula
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...