Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 24
Filtrar
Más filtros












Base de datos
Intervalo de año de publicación
1.
Cell Rep ; 38(6): 110348, 2022 02 08.
Artículo en Inglés | MEDLINE | ID: mdl-35114110

RESUMEN

The increasing prevalence of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) variants with the ability to escape existing humoral protection conferred by previous infection and/or immunization necessitates the discovery of broadly reactive neutralizing antibodies (nAbs). Utilizing mRNA display, we identify a set of antibodies against SARS-CoV-2 spike (S) proteins and characterize the structures of nAbs that recognize epitopes in the S1 subunit of the S glycoprotein. These structural studies reveal distinct binding modes for several antibodies, including the targeting of rare cryptic epitopes in the receptor-binding domain (RBD) of S that interact with angiotensin-converting enzyme 2 (ACE2) to initiate infection, as well as the S1 subdomain 1. Further, we engineer a potent ACE2-blocking nAb to sustain binding to S RBD with the E484K and L452R substitutions found in multiple SARS-CoV-2 variants. We demonstrate that mRNA display is an approach for the rapid identification of nAbs that can be used in combination to combat emerging SARS-CoV-2 variants.

2.
Front Immunol ; 12: 729837, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34603305

RESUMEN

We have developed a dual-antigen COVID-19 vaccine incorporating genes for a modified SARS-CoV-2 spike protein (S-Fusion) and the viral nucleocapsid (N) protein with an Enhanced T-cell Stimulation Domain (N-ETSD) to increase the potential for MHC class II responses. The vaccine antigens are delivered by a human adenovirus serotype 5 platform, hAd5 [E1-, E2b-, E3-], previously demonstrated to be effective in the presence of Ad immunity. Vaccination of rhesus macaques with the hAd5 S-Fusion + N-ETSD vaccine by subcutaneous prime injection followed by two oral boosts elicited neutralizing anti-S IgG and T helper cell 1-biased T-cell responses to both S and N that protected the upper and lower respiratory tracts from high titer (1 x 106 TCID50) SARS-CoV-2 challenge. Notably, viral replication was inhibited within 24 hours of challenge in both lung and nasal passages, becoming undetectable within 7 days post-challenge.


Asunto(s)
Vacunas contra la COVID-19/inmunología , COVID-19/prevención & control , Proteínas de la Nucleocápside de Coronavirus/inmunología , SARS-CoV-2/inmunología , Glicoproteína de la Espiga del Coronavirus/inmunología , Adenovirus Humanos/genética , Adenovirus Humanos/inmunología , Adenovirus Humanos/metabolismo , Administración Oral , Animales , Anticuerpos Neutralizantes/sangre , Anticuerpos Antivirales/sangre , Vacunas contra la COVID-19/administración & dosificación , Citocinas/sangre , Inmunización Secundaria/métodos , Inmunoglobulina G/sangre , Pulmón/virología , Macaca mulatta , Nariz/virología , Fosfoproteínas/inmunología , Dominios Proteicos/inmunología , Linfocitos T Colaboradores-Inductores/inmunología , Vacunación , Replicación Viral/inmunología
3.
bioRxiv ; 2021 Sep 14.
Artículo en Inglés | MEDLINE | ID: mdl-34545362

RESUMEN

The increasing prevalence of SARS-CoV-2 variants with the ability to escape existing humoral protection conferred by previous infection and/or immunization necessitates the discovery of broadly-reactive neutralizing antibodies (nAbs). Utilizing mRNA display, we identified a set of antibodies against SARS-CoV-2 spike (S) proteins and characterized the structures of nAbs that recognized epitopes in the S1 subunit of the S glycoprotein. These structural studies revealed distinct binding modes for several antibodies, including targeting of rare cryptic epitopes in the receptor-binding domain (RBD) of S that interacts with angiotensin- converting enzyme 2 (ACE2) to initiate infection, as well as the S1 subdomain 1. A potent ACE2-blocking nAb was further engineered to sustain binding to S RBD with the E484K and L452R substitutions found in multiple SARS-CoV-2 variants. We demonstrate that mRNA display is a promising approach for the rapid identification of nAbs that can be used in combination to combat emerging SARS-CoV-2 variants.

4.
Sci Rep ; 11(1): 14917, 2021 07 21.
Artículo en Inglés | MEDLINE | ID: mdl-34290317

RESUMEN

We have developed a COVID-19 vaccine, hAd5 S-Fusion + N-ETSD, that expresses SARS-CoV-2 spike (S) and nucleocapsid (N) proteins with modifications to increase immune responses delivered using a human adenovirus serotype 5 (hAd5) platform. Here, we demonstrate subcutaneous (SC) prime and SC boost vaccination of CD-1 mice with this dual-antigen vaccine elicits T-helper cell 1 (Th1) biased T-cell and humoral responses to both S and N that are greater than those seen with hAd5 S wild type delivering only unmodified S. We then compared SC to intranasal (IN) prime vaccination with SC or IN boosts and show that an IN prime with an IN boost is as effective at generating Th1 biased humoral responses as the other combinations tested, but an SC prime with an IN or SC boost elicits greater T cell responses. Finally, we used a combined SC plus IN (SC + IN) prime with or without a boost and found the SC + IN prime alone to be as effective in generating humoral and T-cell responses as the SC + IN prime with a boost. The finding that SC + IN prime-only delivery has the potential to provide broad immunity-including mucosal immunity-against SARS-CoV-2 supports further testing of this vaccine and delivery approach in animal models of viral challenge.


Asunto(s)
Vacunas contra la COVID-19/administración & dosificación , COVID-19/inmunología , COVID-19/prevención & control , SARS-CoV-2/inmunología , Adenoviridae/genética , Administración Intranasal , Animales , Anticuerpos Neutralizantes , Anticuerpos Antivirales/biosíntesis , Anticuerpos Antivirales/inmunología , Vacunas contra la COVID-19/inmunología , Femenino , Vectores Genéticos , Hipodermoclisis , Inmunidad Celular/inmunología , Inmunidad Mucosa/inmunología , Inmunización Secundaria , Ratones , Ratones Endogámicos , Vacunación/métodos
5.
J Immunother Cancer ; 9(6)2021 06.
Artículo en Inglés | MEDLINE | ID: mdl-34172517

RESUMEN

BACKGROUND: Therapeutic regimens designed to augment the immunological response of a patient with breast cancer (BC) to tumor tissue are critically informed by tumor mutational burden and the antigenicity of expressed neoepitopes. Herein we describe a neoepitope and cognate neoepitope-reactive T-cell identification and validation program that supports the development of next-generation immunotherapies. METHODS: Using GPS Cancer, NantOmics research, and The Cancer Genome Atlas databases, we developed a novel bioinformatic-based approach which assesses mutational load, neoepitope expression, human leukocyte antigen (HLA)-binding prediction, and in vitro confirmation of T-cell recognition to preferentially identify targetable neoepitopes. This program was validated by application to a BC cell line and confirmed using tumor biopsies from two patients with BC enrolled in the Tumor-Infiltrating Lymphocytes and Genomics (TILGen) study. RESULTS: The antigenicity and HLA-A2 restriction of the BC cell line predicted neoepitopes were determined by reactivity of T cells from HLA-A2-expressing healthy donors. For the TILGen subjects, tumor-infiltrating lymphocytes (TILs) recognized the predicted neoepitopes both as peptides and on retroviral expression in HLA-matched Epstein-Barr virus-lymphoblastoid cell line and BC cell line MCF-7 cells; PCR clonotyping revealed the presence of T cells in the periphery with T-cell receptors for the predicted neoepitopes. These high-avidity immune responses were polyclonal, mutation-specific and restricted to either HLA class I or II. Interestingly, we observed the persistence and expansion of polyclonal T-cell responses following neoadjuvant chemotherapy. CONCLUSIONS: We demonstrate our neoepitope prediction program allows for the successful identification of neoepitopes targeted by TILs in patients with BC, providing a means to identify tumor-specific immunogenic targets for individualized treatment, including vaccines or adoptively transferred cellular therapies.


Asunto(s)
Antígenos de Neoplasias/genética , Neoplasias de la Mama/genética , Antígenos de Histocompatibilidad Clase I/inmunología , Inmunoterapia/métodos , Femenino , Humanos
6.
Sci Rep ; 11(1): 12740, 2021 06 17.
Artículo en Inglés | MEDLINE | ID: mdl-34140558

RESUMEN

The SARS-CoV-2 variants replacing the first wave strain pose an increased threat by their potential ability to escape pre-existing humoral protection. An angiotensin converting enzyme 2 (ACE2) decoy that competes with endogenous ACE2 for binding of the SARS-CoV-2 spike receptor binding domain (S RBD) and inhibits infection may offer a therapeutic option with sustained efficacy against variants. Here, we used Molecular Dynamics (MD) simulation to predict ACE2 sequence substitutions that might increase its affinity for S RBD and screened candidate ACE2 decoys in vitro. The lead ACE2(T27Y/H34A)-IgG1FC fusion protein with enhanced S RBD affinity shows greater live SARS-CoV-2 virus neutralization capability than wild type ACE2. MD simulation was used to predict the effects of S RBD variant mutations on decoy affinity that was then confirmed by testing of an ACE2 Triple Decoy that included an additional enzyme activity-deactivating H374N substitution against mutated S RBD. The ACE2 Triple Decoy maintains high affinity for mutated S RBD, displays enhanced affinity for S RBD N501Y or L452R, and has the highest affinity for S RBD with both E484K and N501Y mutations, making it a viable therapeutic option for the prevention or treatment of SARS-CoV-2 infection with a high likelihood of efficacy against variants.


Asunto(s)
Sustitución de Aminoácidos , Enzima Convertidora de Angiotensina 2/química , Enzima Convertidora de Angiotensina 2/metabolismo , Antivirales/farmacología , COVID-19/metabolismo , Descubrimiento de Drogas/métodos , Simulación de Dinámica Molecular , SARS-CoV-2/metabolismo , Transducción de Señal/efectos de los fármacos , Secuencia de Aminoácidos , COVID-19/virología , Humanos , Mutación , Unión Proteica/efectos de los fármacos , Dominios Proteicos/genética , Glicoproteína de la Espiga del Coronavirus/metabolismo , Internalización del Virus/efectos de los fármacos
7.
Elife ; 102021 01 13.
Artículo en Inglés | MEDLINE | ID: mdl-33438576

RESUMEN

Generating mammalian cells with specific mitochondrial DNA (mtDNA)-nuclear DNA (nDNA) combinations is desirable but difficult to achieve and would be enabling for studies of mitochondrial-nuclear communication and coordination in controlling cell fates and functions. We developed 'MitoPunch', a pressure-driven mitochondrial transfer device, to deliver isolated mitochondria into numerous target mammalian cells simultaneously. MitoPunch and MitoCeption, a previously described force-based mitochondrial transfer approach, both yield stable isolated mitochondrial recipient (SIMR) cells that permanently retain exogenous mtDNA, whereas coincubation of mitochondria with cells does not yield SIMR cells. Although a typical MitoPunch or MitoCeption delivery results in dozens of immortalized SIMR clones with restored oxidative phosphorylation, only MitoPunch can produce replication-limited, non-immortal human SIMR clones. The MitoPunch device is versatile, inexpensive to assemble, and easy to use for engineering mtDNA-nDNA combinations to enable fundamental studies and potential translational applications.


Mitochondria are specialized structures within cells that generate vital energy and biological building blocks. Mitochondria have a double membrane and contain many copies of their own circular DNA (mitochondrial DNA), which include the blueprints to create just thirteen essential mitochondrial proteins. Like all genetic material, mitochondrial DNA can become damaged or mutated, and these changes can be passed on to offspring. Some of these alterations are linked to severe and debilitating diseases. Both the double membrane of the mitochondria and their high number of DNA copies make treating such diseases difficult. A successful therapy must be capable of correcting almost every copy of mitochondrial DNA. However, the multiple copies of mitochondrial DNA create a problem for genetic research as current techniques are unable to reliably introduce particular mitochondrial mutations to all types of human cells to investigate how they may alter cell function. Sercel, Patananan et al. have developed a method to deliver new mitochondria into thousands of cells at the same time. This technique, called MitoPunch, uses a pressure-driven device to propel mitochondria taken from donor cells into recipient cells without mitochondrial DNA to reestablish their function. Using human cancer cells and healthy skin cells that lack mitochondrial DNA, Sercel, Patananan et al. showed that cells that received mitochondria retained the new mitochondrial DNA. The technique uses readily accessible parts, meaning it can be performed quickly and inexpensively in any laboratory. It further only requires a small amount of donor starting material, meaning that even precious samples with limited material could be used as mitochondrial donors. This new technique has several important potential applications for mitochondrial DNA research. It could be used in the lab to create large numbers of cell lineswith known mutations in the mitochondrial DNA to establish new systems that test drugs or probe the interaction between mitochondrial and nuclear DNA. It could be used to study a broad spectrum of biological questions since mitochondrial function is essential for several processes required for life. Critically, it could also be used as a starting point to develop next-generation therapies capable of treating inherited mitochondrial genetic diseases in severely affected patients.


Asunto(s)
Diferenciación Celular , Núcleo Celular/metabolismo , ADN Mitocondrial/genética , Mitocondrias/metabolismo , Animales , Línea Celular , Células HEK293 , Humanos , Ratones
8.
Lab Chip ; 21(6): 1049-1060, 2021 03 21.
Artículo en Inglés | MEDLINE | ID: mdl-33313615

RESUMEN

In molecular and cellular biological research, cell isolation and sorting are required for accurate investigation of cell populations of specific physical or biological characteristics. By employing unique cell properties to distinguish between heterogeneous cell populations, rapid and accurate sorting with high efficiency is possible. Dielectrophoresis-based cell manipulation has significant promise for separation of cells based on their physical properties and is used in diverse areas ranging from cellular diagnostics to therapeutic applications. In this study, we present a microfluidic device that can achieve label-free and size-based cell separation with high size differential resolution from a mono-cellular population or complex sample matrices. It was realized by using the tunnel dielectrophoresis (TDEP) technique to manipulate the spatial position of individual cells three dimensionally with high resolution. Cells were processed in high speed flows in high ionic strength buffers. A mixture of different sizes of polystyrene micro-particles with a size difference as small as 1 µm can be separated with high purity (>90%). For the first time, high-pass, low-pass, and band-pass filtering within a mono-cellular mammalian cell population were demonstrated with a tunable bandwidth as small as 3 µm. In addition, leukocyte subtype separation was demonstrated by sorting monocytes out of peripheral blood mononuclear cells (PBMCs) from whole blood with high purity (>85%). Its ability to deliver real-time adjustable cut-off threshold size-based cell sorting and its capability to provide an arbitrary cell size pick-up band could potentially enable many research and clinical applications.


Asunto(s)
Dispositivos Laboratorio en un Chip , Leucocitos Mononucleares , Animales , Separación Celular , Monocitos , Poliestirenos
9.
Cell Rep ; 33(13): 108562, 2020 12 29.
Artículo en Inglés | MEDLINE | ID: mdl-33378680

RESUMEN

Generating mammalian cells with desired mitochondrial DNA (mtDNA) sequences is enabling for studies of mitochondria, disease modeling, and potential regenerative therapies. MitoPunch, a high-throughput mitochondrial transfer device, produces cells with specific mtDNA-nuclear DNA (nDNA) combinations by transferring isolated mitochondria from mouse or human cells into primary or immortal mtDNA-deficient (ρ0) cells. Stable isolated mitochondrial recipient (SIMR) cells isolated in restrictive media permanently retain donor mtDNA and reacquire respiration. However, SIMR fibroblasts maintain a ρ0-like cell metabolome and transcriptome despite growth in restrictive media. We reprogrammed non-immortal SIMR fibroblasts into induced pluripotent stem cells (iPSCs) with subsequent differentiation into diverse functional cell types, including mesenchymal stem cells (MSCs), adipocytes, osteoblasts, and chondrocytes. Remarkably, after reprogramming and differentiation, SIMR fibroblasts molecularly and phenotypically resemble unmanipulated control fibroblasts carried through the same protocol. Thus, our MitoPunch "pipeline" enables the production of SIMR cells with unique mtDNA-nDNA combinations for additional studies and applications in multiple cell types.


Asunto(s)
Reprogramación Celular , Fibroblastos/metabolismo , Técnicas de Transferencia de Gen , Ensayos Analíticos de Alto Rendimiento/métodos , Mitocondrias/genética , Mitocondrias/metabolismo , Mitocondrias/trasplante , Animales , Diferenciación Celular , Línea Celular , ADN Mitocondrial/metabolismo , Células HEK293 , Humanos , Células Madre Pluripotentes Inducidas/metabolismo , Metaboloma , Ratones , Ratones Endogámicos C57BL , Transcriptoma
10.
Sci Rep ; 9(1): 11088, 2019 07 31.
Artículo en Inglés | MEDLINE | ID: mdl-31366998

RESUMEN

Deep learning has achieved spectacular performance in image and speech recognition and synthesis. It outperforms other machine learning algorithms in problems where large amounts of data are available. In the area of measurement technology, instruments based on the photonic time stretch have established record real-time measurement throughput in spectroscopy, optical coherence tomography, and imaging flow cytometry. These extreme-throughput instruments generate approximately 1 Tbit/s of continuous measurement data and have led to the discovery of rare phenomena in nonlinear and complex systems as well as new types of biomedical instruments. Owing to the abundance of data they generate, time-stretch instruments are a natural fit to deep learning classification. Previously we had shown that high-throughput label-free cell classification with high accuracy can be achieved through a combination of time-stretch microscopy, image processing and feature extraction, followed by deep learning for finding cancer cells in the blood. Such a technology holds promise for early detection of primary cancer or metastasis. Here we describe a new deep learning pipeline, which entirely avoids the slow and computationally costly signal processing and feature extraction steps by a convolutional neural network that directly operates on the measured signals. The improvement in computational efficiency enables low-latency inference and makes this pipeline suitable for cell sorting via deep learning. Our neural network takes less than a few milliseconds to classify the cells, fast enough to provide a decision to a cell sorter for real-time separation of individual target cells. We demonstrate the applicability of our new method in the classification of OT-II white blood cells and SW-480 epithelial cancer cells with more than 95% accuracy in a label-free fashion.


Asunto(s)
Separación Celular/métodos , Citometría de Flujo/métodos , Algoritmos , Células Cultivadas , Aprendizaje Profundo , Humanos , Procesamiento de Imagen Asistido por Computador/métodos , Aprendizaje Automático , Microscopía/métodos , Procesamiento de Señales Asistido por Computador
11.
Cancer Immunol Res ; 7(8): 1359-1370, 2019 08.
Artículo en Inglés | MEDLINE | ID: mdl-31292145

RESUMEN

Progressive tumor growth is associated with deficits in the immunity generated against tumor antigens. Vaccines targeting tumor neoepitopes have the potential to address qualitative defects; however, additional mechanisms of immune failure may underlie tumor progression. In such cases, patients would benefit from additional immune-oncology agents targeting potential mechanisms of immune failure. This study explores the identification of neoepitopes in the MC38 colon carcinoma model by comparison of tumor to normal DNA and tumor RNA sequencing technology, as well as neoepitope delivery by both peptide- and adenovirus-based vaccination strategies. To improve antitumor efficacies, we combined the vaccine with a group of rationally selected immune-oncology agents. We utilized an IL15 superagonist to enhance the development of antigen-specific immunity initiated by the neoepitope vaccine, PD-L1 blockade to reduce tumor immunosuppression, and a tumor-targeted IL12 molecule to facilitate T-cell function within the tumor microenvironment. Analysis of tumor-infiltrating leukocytes demonstrated this multifaceted treatment regimen was required to promote the influx of CD8+ T cells and enhance the expression of transcripts relating to T-cell activation/effector function. Tumor-targeted IL12 resulted in a marked increase in clonality of T-cell repertoire infiltrating the tumor, which when sculpted with the addition of either a peptide or adenoviral neoepitope vaccine promoted efficient tumor clearance. In addition, the neoepitope vaccine induced the spread of immunity to neoepitopes expressed by the tumor but not contained within the vaccine. These results demonstrate the importance of combining neoepitope-targeting vaccines with a multifaceted treatment regimen to generate effective antitumor immunity.


Asunto(s)
Antígenos de Neoplasias/inmunología , Vacunas contra el Cáncer/inmunología , Epítopos/inmunología , Neoplasias/inmunología , Animales , Linfocitos T CD8-positivos/inmunología , Linfocitos T CD8-positivos/metabolismo , Linfocitos T CD8-positivos/patología , Línea Celular Tumoral , Terapia Combinada , Modelos Animales de Enfermedad , Femenino , Perfilación de la Expresión Génica , Humanos , Inmunomodulación , Inmunoterapia/métodos , Linfocitos Infiltrantes de Tumor/inmunología , Linfocitos Infiltrantes de Tumor/metabolismo , Ratones , Neoplasias/genética , Neoplasias/patología , Neoplasias/terapia , Resultado del Tratamiento , Carga Tumoral , Vacunación
12.
Sci Rep ; 9(1): 8457, 2019 06 11.
Artículo en Inglés | MEDLINE | ID: mdl-31186484

RESUMEN

The biology of tumor-derived exosomes (TEX) is only partially understood and much remains to be studied in order to define the effect that the tumor microenvironment or the activation of tumor cells exerts on their composition and functions. Increased expression and activity of toll-like receptor 4 (TLR4) in chronic infectious and inflammatory conditions is related with cancer progression: its activation induces an inflammatory signaling that increases the tumorigenic potential of cancer cells promoting their immune evasion. We investigated the immune modulatory properties of TEX released upon cell TLR4 activation, and we found that, although differences were observed depending on the type of the tumor, the treatment influences TEX composition and boosts their immunosuppressive ability. Our results suggest that the activation of TLR4 supports tumor progression by stimulating the release of more effective immunosuppressive exosomes, which allow tumor cells to escape immune surveillance and probably even play a role in the metastatic process.


Asunto(s)
Exosomas/genética , Neoplasias/genética , Receptor Toll-Like 4/genética , Microambiente Tumoral/inmunología , Línea Celular Tumoral , Exosomas/inmunología , Regulación Neoplásica de la Expresión Génica/inmunología , Humanos , Inmunosupresores/inmunología , Inmunosupresores/metabolismo , MicroARNs/genética , Neoplasias/clasificación , Neoplasias/inmunología , Neoplasias/patología , Transducción de Señal , Receptor Toll-Like 4/inmunología , Microambiente Tumoral/genética
13.
Sci Rep ; 8(1): 13325, 2018 09 06.
Artículo en Inglés | MEDLINE | ID: mdl-30190615

RESUMEN

The predominant mechanism by which adipose mesenchymal stem cells (AMSCs) participate to tissue repair is through a paracrine activity and their communication with the inflammatory microenvironment is essential part of this process. This hypothesis has been strengthened by the recent discovery that stem cells release not only soluble factors but also extracellular vesicles, which elicit similar biological activity to the stem cells themselves. We demonstrated that the treatment with inflammatory cytokines increases the immunosuppressive and anti-inflammatory potential of AMSCs-derived exosomes, which acquire the ability to shift macrophages from M1 to M2 phenotype by shuttling miRNA regulating macrophages polarization. This suggests that the immunomodulatory properties of AMSCs-derived exosomes may be not constitutive, but are instead induced by the inflammatory microenvironment.


Asunto(s)
Tejido Adiposo/inmunología , Microambiente Celular/inmunología , Exosomas/inmunología , Tolerancia Inmunológica , Macrófagos/inmunología , Células Madre Mesenquimatosas/inmunología , Humanos , Inflamación/inmunología
14.
NPJ Genom Med ; 3: 14, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-29928512

RESUMEN

Immune heterogeneity within the tumor microenvironment undoubtedly adds several layers of complexity to our understanding of drug sensitivity and patient prognosis across various cancer types. Within the tumor microenvironment, immunogenicity is a favorable clinical feature in part driven by the antitumor activity of CD8+ T cells. However, tumors often inhibit this antitumor activity by exploiting the suppressive function of regulatory T cells (Tregs), thus suppressing the adaptive immune response. Despite the seemingly intuitive immunosuppressive biology of Tregs, prognostic studies have produced contradictory results regarding the relationship between Treg enrichment and survival. We therefore analyzed RNA-seq data of Treg-enriched tumor samples to derive a pan-cancer gene signature able to help reconcile the inconsistent results of Treg studies, by better understanding the variable clinical association of Tregs across alternative tumor contexts. We show that increased expression of a 32-gene signature in Treg-enriched tumor samples (n = 135) is able to distinguish a cohort of patients associated with chemosensitivity and overall survival. This cohort is also enriched for CD8+ T cell abundance, as well as the antitumor M1 macrophage subtype. With a subsequent validation in a larger TCGA pool of Treg-enriched patients (n = 626), our results reveal a gene signature able to produce unsupervised clusters of Treg-enriched patients, with one cluster of patients uniquely representative of an immunogenic tumor microenvironment. Ultimately, these results support the proposed gene signature as a putative biomarker to identify certain Treg-enriched patients with immunogenic tumors that are more likely to be associated with features of favorable clinical outcome.

15.
Mediators Inflamm ; 2017: 4814987, 2017.
Artículo en Inglés | MEDLINE | ID: mdl-28634420

RESUMEN

The purpose of this study is to characterize synovial fluid- (SF-) derived exosomes of patients with gonarthrosis comparing two methods of isolation and to investigate their immune regulatory properties. Extracellular vesicles (EVs) have been isolated from inflamed SF by polymer precipitation method and quantified by Exocet kit and by nanoparticle tracking analysis. Vesicles expressed all the specific exosomal markers by immunoblot and FACS. After isolation with Exoquick, a relevant contamination by immune complexes was detected, which required further magnetic bead-based purification to remove. SF-derived exosomes significantly stimulated the release of several inflammatory cytokines and chemokines and metalloproteinases by M1 macrophages but did not influence the expression of CD80 and CD86 costimulatory molecules. In conclusion, we characterized purified exosomes isolated from inflamed SF and demonstrate that purified exosomes are functionally active in their ability to stimulate the release of proinflammatory factors from M1 macrophages. Our data indicate that SF-derived exosomes from gonarthrosis patients play a role in disease progression.


Asunto(s)
Exosomas/metabolismo , Osteoartritis/metabolismo , Líquido Sinovial/metabolismo , Anciano , Anciano de 80 o más Años , Células Cultivadas , Quimiocinas/metabolismo , Citocinas/metabolismo , Exosomas/química , Vesículas Extracelulares/química , Vesículas Extracelulares/metabolismo , Femenino , Humanos , Leucocitos Mononucleares/metabolismo , Macrófagos/metabolismo , Masculino , Persona de Mediana Edad
16.
Cell Metab ; 23(5): 921-9, 2016 05 10.
Artículo en Inglés | MEDLINE | ID: mdl-27166949

RESUMEN

mtDNA sequence alterations are challenging to generate but desirable for basic studies and potential correction of mtDNA diseases. Here, we report a new method for transferring isolated mitochondria into somatic mammalian cells using a photothermal nanoblade, which bypasses endocytosis and cell fusion. The nanoblade rescued the pyrimidine auxotroph phenotype and respiration of ρ0 cells that lack mtDNA. Three stable isogenic nanoblade-rescued clones grown in uridine-free medium showed distinct bioenergetics profiles. Rescue lines 1 and 3 reestablished nucleus-encoded anapleurotic and catapleurotic enzyme gene expression patterns and had metabolite profiles similar to the parent cells from which the ρ0 recipient cells were derived. By contrast, rescue line 2 retained a ρ0 cell metabolic phenotype despite growth in uridine-free selection. The known influence of metabolite levels on cellular processes, including epigenome modifications and gene expression, suggests metabolite profiling can help assess the quality and function of mtDNA-modified cells.


Asunto(s)
Luz , Mamíferos/metabolismo , Metaboloma , Mitocondrias/metabolismo , Nanopartículas/química , Temperatura , Animales , Secuencia de Bases , Línea Celular Tumoral , Células Clonales , ADN Mitocondrial/genética , Metabolismo Energético , Regulación de la Expresión Génica , Humanos , Metaboloma/genética , Metabolómica , Reproducibilidad de los Resultados
17.
Sci Rep ; 6: 21471, 2016 Mar 15.
Artículo en Inglés | MEDLINE | ID: mdl-26975219

RESUMEN

Label-free cell analysis is essential to personalized genomics, cancer diagnostics, and drug development as it avoids adverse effects of staining reagents on cellular viability and cell signaling. However, currently available label-free cell assays mostly rely only on a single feature and lack sufficient differentiation. Also, the sample size analyzed by these assays is limited due to their low throughput. Here, we integrate feature extraction and deep learning with high-throughput quantitative imaging enabled by photonic time stretch, achieving record high accuracy in label-free cell classification. Our system captures quantitative optical phase and intensity images and extracts multiple biophysical features of individual cells. These biophysical measurements form a hyperdimensional feature space in which supervised learning is performed for cell classification. We compare various learning algorithms including artificial neural network, support vector machine, logistic regression, and a novel deep learning pipeline, which adopts global optimization of receiver operating characteristics. As a validation of the enhanced sensitivity and specificity of our system, we show classification of white blood T-cells against colon cancer cells, as well as lipid accumulating algal strains for biofuel production. This system opens up a new path to data-driven phenotypic diagnosis and better understanding of the heterogeneous gene expressions in cells.


Asunto(s)
Algoritmos , Biología Computacional/métodos , Validación de Programas de Computación , Inteligencia Artificial , Línea Celular Tumoral/clasificación , Línea Celular Tumoral/patología , Chlamydomonas reinhardtii/clasificación , Chlamydomonas reinhardtii/citología , Chlamydomonas reinhardtii/metabolismo , Humanos , Redes Neurales de la Computación , Reconocimiento de Normas Patrones Automatizadas/métodos , Reproducibilidad de los Resultados , Máquina de Vectores de Soporte , Linfocitos T/clasificación , Linfocitos T/citología
18.
Org Lett ; 17(14): 3608-11, 2015 Jul 17.
Artículo en Inglés | MEDLINE | ID: mdl-26144210

RESUMEN

An analogue 2 of Brasilicardin A, 1 (BraA), a potent immunosuppressive and cytotoxic agent, was synthesized in which the natural tricyclic skeleton was replaced with a synthetically more accessible substituted tetrahydronaphthalene core. BraA, this analogue (BraL), and cyclosporine A were tested for their ability to inhibit the proliferation of human T cells upon CD3/CD28 activation. Although BraL did not impact T cell activation over the dose range tested, this study shows the inhibitory activity of BraA on human T cells for the first time.


Asunto(s)
Aminoglicósidos/síntesis química , Aminoglicósidos/farmacología , Antineoplásicos/síntesis química , Antineoplásicos/farmacología , Ciclosporina/química , Inmunosupresores/farmacología , Naftalenos/química , Linfocitos T/efectos de los fármacos , Aminoglicósidos/química , Antineoplásicos/química , Antígenos CD28 , Complejo CD3 , Humanos , Inmunosupresores/química , Estructura Molecular
19.
Biomed Opt Express ; 4(9): 1618-25, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-24049682

RESUMEN

Flow cytometry is a powerful tool for cell counting and biomarker detection in biotechnology and medicine especially with regards to blood analysis. Standard flow cytometers perform cell type classification both by estimating size and granularity of cells using forward- and side-scattered light signals and through the collection of emission spectra of fluorescently-labeled cells. However, cell surface labeling as a means of marking cells is often undesirable as many reagents negatively impact cellular viability or provide activating/inhibitory signals, which can alter the behavior of the desired cellular subtypes for downstream applications or analysis. To eliminate the need for labeling, we introduce a label-free imaging-based flow cytometer that measures size and cell protein concentration simultaneously either as a stand-alone instrument or as an add-on to conventional flow cytometers. Cell protein concentration adds a parameter to cell classification, which improves the specificity and sensitivity of flow cytometers without the requirement of cell labeling. This system uses coherent dispersive Fourier transform to perform phase imaging at flow speeds as high as a few meters per second.

20.
J Neurosci Methods ; 212(2): 190-4, 2013 Jan 30.
Artículo en Inglés | MEDLINE | ID: mdl-23022695

RESUMEN

Studies from multiple laboratories have identified the roles of several ER stress-induced cell death modulators and effectors. Earlier, we described the role of p23 a small co-chaperone protein in preventing ER stress-induced cell death. p23 is cleaved by caspases at D142 to yield p19 (a 19 kDa product) during ER stress-induced cell death. Mutation of the caspase cleavage site not only blocks formation of the 19 kDa product but also attenuates the cell death process triggered by various ER stressors. Thus, uncleavable p23 (p23D142N) emerges as a reasonable candidate to test for potential inhibition of neurodegenerative disease phenotype that features misfolded proteins and ER stress. In the present work we report the generation of transgenic mouse lines that overexpress wild-type p23 or uncleavable p23 under the control of a ROSA promoter. These mice should prove useful for studying the role of p23 and/or uncleavable p23 in cellular stress-induced cell death.


Asunto(s)
Encéfalo/fisiología , Oxidorreductasas Intramoleculares/genética , Ratones Transgénicos , Animales , Western Blotting , Inmunohistoquímica , Ratones , Ratones Endogámicos C57BL , Reacción en Cadena de la Polimerasa , Regiones Promotoras Genéticas , Prostaglandina-E Sintasas , Transgenes
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...