Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 7 de 7
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Bio Protoc ; 13(14): e4724, 2023 Jul 20.
Artículo en Inglés | MEDLINE | ID: mdl-37497456

RESUMEN

The immune-inhibitory molecule programmed cell death ligand 1 (PD-L1) has been shown to play a role in pathologies such as autoimmunity, infections, and cancer. The expression of PD-L1 not only on cancer cells but also on non-transformed host cells is known to be associated with cancer progression. Generation of PD-L1 deficiency in the murine system enables us to specifically study the role of PD-L1 in physiological processes and diseases. One of the most versatile and easy to use site-specific gene editing tools is the CRISPR/Cas9 system, which is based on an RNA-guided nuclease system. Similar to its predecessors, the Zinc finger nucleases or transcription activator-like effector nucleases (TALENs), CRISPR/Cas9 catalyzes double-strand DNA breaks, which can result in frameshift mutations due to random nucleotide insertions or deletions via non-homologous end joining (NHEJ). Furthermore, although less frequently, CRISPR/Cas9 can lead to insertion of defined sequences due to homology-directed repair (HDR) in the presence of a suitable template. Here, we describe a protocol for the knockout of PD-L1 in the murine C57BL/6 background using CRISPR/Cas9. Targeting of exon 3 coupled with the insertion of a HindIII restriction site leads to a premature stop codon and a loss-of-function phenotype. We describe the targeting strategy as well as founder screening, genotyping, and phenotyping. In comparison to NHEJ-based strategy, the presented approach results in a defined stop codon with comparable efficiency and timelines as NHEJ, generates convenient founder screening and genotyping options, and can be swiftly adapted to other targets.

2.
Small Methods ; 7(2): e2201061, 2023 02.
Artículo en Inglés | MEDLINE | ID: mdl-36572638

RESUMEN

Imaging of iron-based nanoparticles (NPs) remains challenging because of the presence of endogenous iron in tissues that is difficult to distinguish from exogenous iron originating from the NPs. Here, an analytical cascade for characterizing the biodistribution of biomedically relevant iron-based NPs from the organ scale to the cellular and subcellular scales is introduced. The biodistribution on an organ level is assessed by elemental analysis and quantification of magnetic iron by electron paramagnetic resonance, which allowed differentiation of exogenous and endogenous iron. Complementary to these bulk analysis techniques, correlative whole-slide optical and electron microscopy provided spatially resolved insight into the biodistribution of endo- and exogenous iron accumulation in macrophages, with single-cell and single-particle resolution, revealing coaccumulation of iron NPs with endogenous iron in splenic macrophages. Subsequent transmission electron microscopy revealed two types of morphologically distinct iron-containing structures (exogenous nanoparticles and endogenous ferritin) within membrane-bound vesicles in the cytoplasm, hinting at an attempt of splenic macrophages to extract and recycle iron from exogenous nanoparticles. Overall, this strategy enables the distinction of endo- and exogenous iron across scales (from cm to nm, based on the analysis of thousands of cells) and illustrates distribution on organ, cell, and organelle levels.


Asunto(s)
Hierro , Macrófagos , Distribución Tisular , Microscopía Electrónica , Microscopía Electrónica de Transmisión
3.
NMR Biomed ; 35(6): e4690, 2022 06.
Artículo en Inglés | MEDLINE | ID: mdl-34994020

RESUMEN

Microscopic magnetic field inhomogeneities caused by iron deposition or tissue-air interfaces may result in rapid decay of transverse magnetization in MRI. The aim of this study is to detect and quantify the distribution of iron-based nanoparticles in mouse models by applying ultrashort-echo-time (UTE) sequences in tissues exhibiting extremely fast transverse relaxation. In 24 C57BL/6 mice (two controls), suspensions containing either non-oxidic Fe or AuFeOx nanoparticles were injected into the tail vein at two doses (200 µg and 600 µg per mouse). Mice underwent MRI using a UTE sequence at 4.7 T field strength with five different echo times between 100 µs and 5000 µs. Transverse relaxation times T2 * were computed for the lung, liver, and spleen by mono-exponential fitting. In UTE imaging, the MRI signal could reliably be detected even in liver parenchyma exhibiting the highest deposition of nanoparticles. In animals treated with Fe nanoparticles (600 µg per mouse), the relaxation time substantially decreased in the liver (3418 ± 1534 µs (control) versus 228 ± 67 µs), the spleen (2170 ± 728 µs versus 299 ± 97 µs), and the lungs (663 ± 101 µs versus 413 ± 99 µs). The change in transverse relaxation was dependent on the number and composition of the nanoparticles. By pixel-wise curve fitting, T2 * maps were calculated showing nanoparticle distribution. In conclusion, UTE sequences may be used to assess and quantify nanoparticle distribution in tissues exhibiting ultrafast signal decay in MRI.


Asunto(s)
Hierro , Nanopartículas , Animales , Modelos Animales de Enfermedad , Imagen por Resonancia Magnética/métodos , Ratones , Ratones Endogámicos C57BL
4.
Sci Transl Med ; 13(611): eabc8188, 2021 Sep 15.
Artículo en Inglés | MEDLINE | ID: mdl-34524861

RESUMEN

Platelet-derived peripheral serotonin has pleiotropic effects on coagulation, metabolism, tissue regeneration, and cancer growth; however, the effect of serotonin on the tumor microenvironment remains understudied. Peripheral serotonin­deficient (Tph1−/−) mice displayed reduced growth of subcutaneous and orthotopically injected syngeneic murine pancreatic and colorectal cancers with enhanced accumulation of functional CD8+ T cells compared to control C57BL/6 mice, resulting in extended overall survival. Subcutaneous and orthotopic syngeneic tumors from Tph1−/− mice expressed less programmed cell death 1 ligand 1 (PD-L1), suggesting serotonin-mediated regulation. Serotonin enhanced expression of PD-L1 on mouse and human cancer cells in vitro via serotonylation, which is the formation of covalent bonds between glutamine residues and serotonin, resulting in activation of small G proteins. Serotonin concentrations in metastases of patients with abdominal tumors negatively correlated to the number of CD8+ tumor-infiltrating T cells. Depletion of serotonin cargo or inhibition of serotonin release from thrombocytes decreased growth of syngeneic pancreatic and colorectal tumors in wild-type mice, increased CD8+ T cell influx, and decreased PD-L1 expression. Pharmacological serotonin depletion with oral fluoxetine or intraperitoneal injection of the TPH1 inhibitor telotristat augmented the effects of programmed cell death protein 1 (PD-1) checkpoint blockade and triggered long-term tumor control in mice subcutaneously inoculated with syngeneic colorectal and pancreatic tumors. Overall, peripheral serotonin weakens effector functions of CD8+ T cells within tumors. Clinically approved serotonin targeting agents alone or in combination with PD-1 blockade provided long-term control of established tumors in murine models, warranting further investigation of the clinical translatability of these findings.


Asunto(s)
Inhibidores de Puntos de Control Inmunológico , Neoplasias , Animales , Ratones , Neoplasias/tratamiento farmacológico , Serotonina
5.
Proc Natl Acad Sci U S A ; 118(27)2021 07 06.
Artículo en Inglés | MEDLINE | ID: mdl-34183415

RESUMEN

The liver is a major metastatic target organ, and little is known about the role of immunity in controlling hepatic metastases. Here, we discovered that the concerted and nonredundant action of two innate lymphocyte subpopulations, conventional natural killer cells (cNKs) and tissue-resident type I innate lymphoid cells (trILC1s), is essential for antimetastatic defense. Using different preclinical models for liver metastasis, we found that trILC1 controls metastatic seeding, whereas cNKs restrain outgrowth. Whereas the killing capacity of trILC1s was not affected by the metastatic microenvironment, the phenotype and function of cNK cells were affected in a cancer type-specific fashion. Thus, individual cancer cell lines orchestrate the emergence of unique cNK subsets, which respond differently to tumor-derived factors. Our findings will contribute to the development of therapies for liver metastasis involving hepatic innate cells.


Asunto(s)
Inmunidad Innata/inmunología , Células Asesinas Naturales/inmunología , Neoplasias Hepáticas/inmunología , Neoplasias Hepáticas/secundario , Linfocitos/inmunología , Animales , Femenino , Regulación Neoplásica de la Expresión Génica , Integrina alfa1/metabolismo , Interleucina-15/metabolismo , Hígado/inmunología , Hígado/patología , Neoplasias Hepáticas/genética , Ratones , Ratones Endogámicos C57BL , RNA-Seq , Análisis de la Célula Individual , Transcriptoma/genética , Factor de Crecimiento Transformador beta/metabolismo , Microambiente Tumoral/genética , Microambiente Tumoral/inmunología
6.
Cell Host Microbe ; 27(6): 922-936.e6, 2020 06 10.
Artículo en Inglés | MEDLINE | ID: mdl-32416061

RESUMEN

Initial enteropathogen growth in the microbiota-colonized gut is poorly understood. Salmonella Typhimurium is metabolically adaptable and can harvest energy by anaerobic respiration using microbiota-derived hydrogen (H2) as an electron donor and fumarate as an electron acceptor. As fumarate is scarce in the gut, the source of this electron acceptor is unclear. Here, transposon sequencing analysis along the colonization trajectory of S. Typhimurium implicates the C4-dicarboxylate antiporter DcuABC in early murine gut colonization. In competitive colonization assays, DcuABC and enzymes that convert the C4-dicarboxylates aspartate and malate into fumarate (AspA, FumABC), are required for fumarate/H2-dependent initial growth. Thus, S. Typhimurium obtains fumarate by DcuABC-mediated import and conversion of L-malate and L-aspartate. Fumarate reduction yields succinate, which is exported by DcuABC in exchange for L-aspartate and L-malate. This cycle allows S. Typhimurium to harvest energy by H2/fumarate respiration in the microbiota-colonized gut. This strategy may also be relevant for commensal E. coli diminishing the S. Typhimurium infection.


Asunto(s)
Ácido Aspártico/metabolismo , Fumaratos/metabolismo , Microbioma Gastrointestinal/fisiología , Malatos/metabolismo , Salmonella/metabolismo , Administración Oral , Animales , Ácido Aspártico/administración & dosificación , Proteínas Bacterianas/metabolismo , Ciclo del Ácido Cítrico , Modelos Animales de Enfermedad , Escherichia coli/metabolismo , Heces/microbiología , Femenino , Microbioma Gastrointestinal/genética , Intestinos/microbiología , Malatos/administración & dosificación , Masculino , Ratones , Ratones Endogámicos C57BL , Mutagénesis , ARN Ribosómico 16S/genética , Salmonella/genética , Salmonella/crecimiento & desarrollo , Salmonella typhimurium , Análisis de Secuencia de ADN , Ácido Succínico
7.
Cell Rep ; 29(5): 1336-1350.e4, 2019 Oct 29.
Artículo en Inglés | MEDLINE | ID: mdl-31665644

RESUMEN

Bacteriophages provide excellent tools for diagnostics, remediation, and targeted microbiome manipulation, yet isolating viruses with suitable host specificity remains challenging. Using Listeria phage PSA, we present a synthetic biology blueprint for host-range engineering through targeted modification of serovar-specific receptor binding proteins (RBPs). We identify Gp15 as the PSA RBP and construct a synthetic phage library featuring sequence-randomized RBPs, from which host range mutants are isolated and subsequently integrated into a synthetic, polyvalent phage with extended host range. To enable rational design of chimeric RBPs, we determine the crystal structure of the Gp15 receptor-binding carboxyl terminus at 1.7-Å resolution and employ bioinformatics to identify compatible, prophage-encoded RBPs targeting different Listeria serovars. Structure-guided design enables exchange of heterologous RBP head, neck, or shoulder domains to generate chimeric phages with predictable and extended host ranges. These strategies will facilitate the development of phage biologics based on standardized virus scaffolds with tunable host specificities.


Asunto(s)
Bacteriófagos/metabolismo , Especificidad del Huésped , Listeria monocytogenes/virología , Receptores Virales/metabolismo , Pared Celular/metabolismo , Secuencia Conservada , Cristalografía por Rayos X , Galactosa/metabolismo , Mutación/genética , Unión Proteica , Dominios Proteicos , Receptores Virales/química , Homología Estructural de Proteína , Ácidos Teicoicos/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...