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.
J Am Soc Mass Spectrom ; 34(12): 2857-2863, 2023 Dec 06.
Artículo en Inglés | MEDLINE | ID: mdl-37874901

RESUMEN

Liquid chromatography-mass spectrometry (LC-MS) metabolomics studies produce high-dimensional data that must be processed by a complex network of informatics tools to generate analysis-ready data sets. As the first computational step in metabolomics, data processing is increasingly becoming a challenge for researchers to develop customized computational workflows that are applicable for LC-MS metabolomics analysis. Ontology-based automated workflow composition (AWC) systems provide a feasible approach for developing computational workflows that consume high-dimensional molecular data. We used the Automated Pipeline Explorer (APE) to create an AWC for LC-MS metabolomics data processing across three use cases. Our results show that APE predicted 145 data processing workflows across all the three use cases. We identified six traditional workflows and six novel workflows. Through manual review, we found that one-third of novel workflows were executable whereby the data processing function could be completed without obtaining an error. When selecting the top six workflows from each use case, the computational viable rate of our predicted workflows reached 45%. Collectively, our study demonstrates the feasibility of developing an AWC system for LC-MS metabolomics data processing.


Asunto(s)
Hominidae , Programas Informáticos , Animales , Flujo de Trabajo , Metabolómica/métodos , Espectrometría de Masas , Cromatografía Liquida/métodos
2.
Metabolomics ; 19(2): 11, 2023 02 06.
Artículo en Inglés | MEDLINE | ID: mdl-36745241

RESUMEN

BACKGROUND: Liquid chromatography-high resolution mass spectrometry (LC-HRMS) is a popular approach for metabolomics data acquisition and requires many data processing software tools. The FAIR Principles - Findability, Accessibility, Interoperability, and Reusability - were proposed to promote open science and reusable data management, and to maximize the benefit obtained from contemporary and formal scholarly digital publishing. More recently, the FAIR principles were extended to include Research Software (FAIR4RS). AIM OF REVIEW: This study facilitates open science in metabolomics by providing an implementation solution for adopting FAIR4RS in the LC-HRMS metabolomics data processing software. We believe our evaluation guidelines and results can help improve the FAIRness of research software. KEY SCIENTIFIC CONCEPTS OF REVIEW: We evaluated 124 LC-HRMS metabolomics data processing software obtained from a systematic review and selected 61 software for detailed evaluation using FAIR4RS-related criteria, which were extracted from the literature along with internal discussions. We assigned each criterion one or more FAIR4RS categories through discussion. The minimum, median, and maximum percentages of criteria fulfillment of software were 21.6%, 47.7%, and 71.8%. Statistical analysis revealed no significant improvement in FAIRness over time. We identified four criteria covering multiple FAIR4RS categories but had a low %fulfillment: (1) No software had semantic annotation of key information; (2) only 6.3% of evaluated software were registered to Zenodo and received DOIs; (3) only 14.5% of selected software had official software containerization or virtual machine; (4) only 16.7% of evaluated software had a fully documented functions in code. According to the results, we discussed improvement strategies and future directions.


Asunto(s)
Metabolómica , Programas Informáticos , Metabolómica/métodos , Cromatografía Liquida/métodos , Espectrometría de Masas/métodos , Manejo de Datos
3.
Curr Opin Clin Nutr Metab Care ; 25(5): 292-297, 2022 09 01.
Artículo en Inglés | MEDLINE | ID: mdl-35838294

RESUMEN

PURPOSE OF REVIEW: Precision health provides an unprecedented opportunity to improve the assessment of infant nutrition and health outcomes. Breastfeeding is positively associated with infant health outcomes, yet only 58.3% of children born in 2017 were still breastfeeding at 6 months. There is an urgent need to examine the application of precision health tools that support the development of public health interventions focused on improving breastfeeding outcomes. RECENT FINDINGS: In this review, we discussed the novel and highly sensitive techniques that can provide a vast amount of omics data and clinical information just by evaluating small volumes of milk samples, such as RNA sequencing, cytometry by time-of-flight, and human milk analyzer for clinical implementation. These advanced techniques can run multiple samples in a short period of time making them ideal for the routine clinical evaluation of milk samples. SUMMARY: Precision health tools are increasingly used in clinical research studies focused on infant nutrition. The integration of routinely collected multiomics human milk data within the electronic health records has the potential to identify molecular biomarkers associated with infant health outcomes.


Asunto(s)
Leche Humana , Medicina de Precisión , Lactancia Materna , Niño , Preescolar , Femenino , Humanos , Lactante , Fenómenos Fisiológicos Nutricionales del Lactante
4.
Neurospine ; 19(1): 13-29, 2022 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-35130421

RESUMEN

Spinal cord gliomas are rare entities that often have limited surgical options. Immunotherapy has shown promise in intracranial gliomas with some research suggesting benefit for spinal cord gliomas. A focused review of immunotherapies that have been investigated in spinal cord gliomas was performed. The primary methods of immunotherapy investigated in spinal cord gliomas include immune checkpoint inhibitors, adoptive T-cell therapies, and vaccine strategies. There are innumerable challenges that must be overcome to effectively apply immunotherapeutic strategies to the spinal cord gliomas including low incidence, few antigenic targets, the blood spinal cord barrier, the immunosuppressive tumor microenvironment and neurotoxic treatment effects. Nonetheless, research has suggested ways to overcome these challenges and treatments have been effective in case reports for metastatic non-small cell lung cancer, melanoma, midline glioma and glioblastoma. Current therapies for spinal cord gliomas are markedly limited. Further research is needed to determine if the success of immunotherapy for intracranial gliomas can be effectively applied to these unique tumors.

5.
Neuromolecular Med ; 24(1): 8-12, 2022 03.
Artículo en Inglés | MEDLINE | ID: mdl-34101090

RESUMEN

Glioblastoma (GBM) is the most common primary brain tumor in adults and is universally lethal with a median survival of less than two years with standard therapy. RNA-based immunotherapies have significant potential to establish a durable treatment response for malignant brain tumors including GBM. RNA offers clear advantages over antigen-focused approaches but cannot often be directly administered due to biological instability. This review will focus on utilization of RNA dendritic cell vaccines and RNA nanoparticle therapies in the treatment of GBM. RNA-pulsed dendritic cell vaccines have been shown to be safe in a small phase I clinical trial and RNA-loaded nanoparticle vaccines will soon be underway in GBM patients (NCT04573140).


Asunto(s)
Neoplasias Encefálicas , Vacunas contra el Cáncer , Glioblastoma , Adulto , Neoplasias Encefálicas/genética , Neoplasias Encefálicas/terapia , Vacunas contra el Cáncer/uso terapéutico , Ensayos Clínicos Fase I como Asunto , Células Dendríticas/patología , Glioblastoma/genética , Glioblastoma/terapia , Humanos , Inmunoterapia , ARN/genética , ARN/uso terapéutico
6.
Curr Treat Options Oncol ; 22(12): 108, 2021 10 23.
Artículo en Inglés | MEDLINE | ID: mdl-34687357

RESUMEN

OPINION STATEMENT: Laser interstitial thermal therapy (LITT) is a minimally invasive treatment option for brain tumors including glioblastoma, other primary central nervous system (CNS) neoplasms, metastases, and radiation necrosis. LITT employs a fiber optic coupled laser delivery probe stabilized via stereotaxis to deliver thermal energy that induces coagulative necrosis in tumors to achieve effective cytoreduction. LITT complements surgical resection, radiation treatment, tumor treating fields, and systemic therapy, especially in patients who are high risk for surgical resection due to tumor location in eloquent regions or poor functional status. These factors must be balanced with the increased rate of cerebral edema post LITT compared to surgical resection. LITT has also been shown to induce transient disruption of the blood-brain barrier (BBB), especially in the peritumoral region, which allows for enhanced CNS delivery of anti-neoplastic agents, thus greatly expanding the armamentarium against brain tumors to include highly effective anti-neoplastic agents that have poor BBB penetration. In addition, hyperthermia-induced immunogenic cell death is another secondary side effect of LITT that opens up immunotherapy as an attractive adjuvant treatment for brain tumors. Numerous large studies have demonstrated the safety and efficacy of LITT against various CNS tumors and as the literature continues to grow on this novel technique so will its indications.


Asunto(s)
Neoplasias Encefálicas/cirugía , Glioblastoma/cirugía , Terapia por Láser/métodos , Traumatismos por Radiación/cirugía , Antineoplásicos/uso terapéutico , Barrera Hematoencefálica/metabolismo , Neoplasias Encefálicas/patología , Neoplasias Encefálicas/secundario , Quimioterapia Adyuvante , Glioblastoma/patología , Humanos , Técnicas Estereotáxicas
7.
Front Immunol ; 12: 705580, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34421912

RESUMEN

Evading T cell surveillance is a hallmark of cancer. Patients with solid tissue malignancy, such as glioblastoma (GBM), have multiple forms of immune dysfunction, including defective T cell function. T cell dysfunction is exacerbated by standard treatment strategies such as steroids, chemotherapy, and radiation. Reinvigoration of T cell responses can be achieved by utilizing adoptively transferred T cells, including CAR T cells. However, these cells are at risk for depletion and dysfunction as well. This review will discuss adoptive T cell transfer strategies and methods to avoid T cell dysfunction for the treatment of brain cancer.


Asunto(s)
Neoplasias Encefálicas , Glioblastoma , Inmunoterapia Adoptiva , Receptores Quiméricos de Antígenos , Neoplasias Encefálicas/inmunología , Neoplasias Encefálicas/terapia , Glioblastoma/inmunología , Glioblastoma/terapia , Humanos , Receptores Quiméricos de Antígenos/inmunología , Receptores Quiméricos de Antígenos/uso terapéutico
8.
Front Oncol ; 11: 691246, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34150663

RESUMEN

Immunotherapy has revolutionized care for many solid tissue malignancies, and is being investigated for efficacy in the treatment of malignant brain tumors. Identifying a non-invasive monitoring technique such as metabolomics monitoring to predict patient response to immunotherapy has the potential to simplify treatment decision-making and to ensure therapy is tailored based on early patient response. Metabolomic analysis of peripheral immune response is feasible due to large metabolic shifts that immune cells undergo when activated. The utility of this approach is under investigation. In this review, we discuss the metabolic changes induced during activation of an immune response, and the role of metabolic profiling to monitor immune responses in the context of immunotherapy for malignant brain tumors. This review provides original insights into how metabolomics monitoring could have an important impact in the field of tumor immunotherapy if achievable.

9.
Neuro Oncol ; 22(9): 1249-1261, 2020 09 29.
Artículo en Inglés | MEDLINE | ID: mdl-32391559

RESUMEN

In oncology, "immunotherapy" is a broad term encompassing multiple means of utilizing the patient's immune system to combat malignancy. Prominent among these are immune checkpoint inhibitors, cellular therapies including chimeric antigen receptor T-cell therapy, vaccines, and oncolytic viruses. Immunotherapy for glioblastoma (GBM) has had mixed results in early trials. In this context, the past, present, and future of immune oncology for the treatment of GBM was discussed by clinical, research, and thought leaders as well as patient advocates at the first annual Remission Summit in 2019. The goal was to use current knowledge (published and unpublished) to identify possible causes of treatment failures and the best strategies to advance immunotherapy as a treatment modality for patients with GBM. The discussion focuses on past failures, current limitations, failure analyses, and proposed best practices moving forward.


Asunto(s)
Neoplasias Encefálicas , Glioblastoma , Virus Oncolíticos , Adulto , Neoplasias Encefálicas/terapia , Glioblastoma/terapia , Humanos , Inmunoterapia
10.
J Immunother Cancer ; 7(1): 321, 2019 11 21.
Artículo en Inglés | MEDLINE | ID: mdl-31753028

RESUMEN

BACKGROUND: Dendritic cell (DC) vaccine efficacy is directly related to the efficiency of DC migration to the lymph node after delivery to the patient. We discovered that a naturally occurring metabolite, sarcosine, increases DC migration in human and murine cells resulting in significantly improved anti-tumor efficacy. We hypothesized that sarcosine induced cell migration was due to chemokine signaling. METHODS: DCs were harvested from the bone marrow of wild type C57BL/6 mice and electroporated with tumor messenger RNA (mRNA). Human DCs were isolated from peripheral blood mononuclear cells (PBMCs). DCs were treated with 20 mM of sarcosine. Antigen specific T cells were isolated from transgenic mice and injected intravenously into tumor bearing mice. DC vaccines were delivered via intradermal injection. In vivo migration was evaluated by flow cytometry and immunofluorescence microscopy. Gene expression in RNA was investigated in DCs via RT-PCR and Nanostring. RESULTS: Sarcosine significantly increased human and murine DC migration in vitro. In vivo sarcosine-treated DCs had significantly increased migration to both the lymph nodes and spleens after intradermal delivery in mice. Sarcosine-treated DC vaccines resulted in significantly improved tumor control in a B16F10-OVA tumor flank model and improved survival in an intracranial GL261-gp100 glioma model. Gene expression demonstrated an upregulation of CXCR2, CXCL3 and CXCL1 in sarcosine- treated DCs. Further metabolic analysis demonstrated the up-regulation of cyclooxygenase-1 and Pik3cg. Sarcosine induced migration was abrogated by adding the CXCR2 neutralizing antibody in both human and murine DCs. CXCR2 neutralizing antibody also removed the survival benefit of sarcosine-treated DCs in the tumor models. CONCLUSION: Sarcosine increases the migration of murine and human DCs via the CXC chemokine pathway. This platform can be utilized to improve existing DC vaccine strategies.


Asunto(s)
Vacunas contra el Cáncer/inmunología , Movimiento Celular/efectos de los fármacos , Movimiento Celular/inmunología , Células Dendríticas/efectos de los fármacos , Células Dendríticas/fisiología , Receptores CXCR/metabolismo , Sarcosina/farmacología , Transducción de Señal/efectos de los fármacos , Traslado Adoptivo , Animales , Biomarcadores , Modelos Animales de Enfermedad , Humanos , Inmunoterapia , Ratones , Neoplasias/inmunología , Neoplasias/metabolismo , Neoplasias/patología , Neoplasias/terapia , Estrés Oxidativo , Receptores CXCR/genética
11.
Neuro Oncol ; 21(6): 730-741, 2019 06 10.
Artículo en Inglés | MEDLINE | ID: mdl-30668768

RESUMEN

BACKGROUND: The changes induced in host immunity and the tumor microenvironment by chemotherapy have been shown to impact immunotherapy response in both a positive and a negative fashion. Temozolomide is the most common chemotherapy used to treat glioblastoma (GBM) and has been shown to have variable effects on immune response to immunotherapy. Therefore, we aimed to determine the immune modulatory effects of temozolomide that would impact response to immune checkpoint inhibition in the treatment of experimental GBM. METHODS: Immune function and antitumor efficacy of immune checkpoint inhibition were tested after treatment with metronomic dose (MD) temozolomide (25 mg/kg × 10 days) or standard dose (SD) temozolomide (50 mg/kg × 5 days) in the GL261 and KR158 murine glioma models. RESULTS: SD temozolomide treatment resulted in an upregulation of markers of T-cell exhaustion such as LAG-3 and TIM-3 in lymphocytes which was not seen with MD temozolomide. When temozolomide treatment was combined with programmed cell death 1 (PD-1) antibody therapy, the MD temozolomide/PD-1 antibody group demonstrated a decrease in exhaustion markers in tumor infiltrating lymphocytes that was not observed in the SD temozolomide/PD-1 antibody group. Also, the survival advantage of PD-1 antibody therapy in a murine syngeneic intracranial glioma model was abrogated by adding SD temozolomide to treatment. However, when MD temozolomide was added to PD-1 inhibition, it preserved the survival benefit that was seen by PD-1 antibody therapy alone. CONCLUSION: The peripheral and intratumoral immune microenvironments are distinctively affected by dose modulation of temozolomide.


Asunto(s)
Anticuerpos Monoclonales/farmacología , Neoplasias Encefálicas/inmunología , Glioblastoma/inmunología , Linfocitos Infiltrantes de Tumor/inmunología , Receptor de Muerte Celular Programada 1/inmunología , Linfocitos T/inmunología , Temozolomida/farmacología , Animales , Antineoplásicos Alquilantes/farmacología , Neoplasias Encefálicas/tratamiento farmacológico , Neoplasias Encefálicas/patología , Relación Dosis-Respuesta a Droga , Quimioterapia Combinada , Glioblastoma/tratamiento farmacológico , Glioblastoma/patología , Humanos , Inmunoterapia , Linfocitos Infiltrantes de Tumor/efectos de los fármacos , Linfocitos Infiltrantes de Tumor/patología , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Linfocitos T/efectos de los fármacos , Linfocitos T/patología , Células Tumorales Cultivadas , Microambiente Tumoral
12.
Oncoimmunology ; 8(1): e1514921, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-30546954

RESUMEN

Controversy surrounds the role of cytomegalovirus (CMV) in glioblastoma (GBM). However, several studies have shown that CMV nucleic acids and proteins are present within GBM tumor tissue. CMV has been implicated in GBM pathogenesis by affecting tumor stem cell factors, angiogenesis and immune pathways. Anti-viral therapy has not been found to definitively improve outcomes for patients with GBM. Several studies have leveraged CMV by targeting CMV antigens using ex-vivo expanded T cells or dendritic cell vaccines. The initial results from these studies are promising and larger studies are underway.

13.
Neuro Oncol ; 20(12): 1566-1572, 2018 11 12.
Artículo en Inglés | MEDLINE | ID: mdl-29733389

RESUMEN

Temozolomide is the most widely used chemotherapy for patients with glioblastoma (GBM) despite the fact that approximately half of treated patients have temozolomide resistance and all patients eventually fail therapy. Due to the limited efficacy of existing therapies, immunotherapy is being widely investigated for patients with GBM. However, initial immunotherapy trials in GBM patients have had disappointing results as monotherapy. Therefore, combinatorial treatment strategies are being investigated. Temozolomide has several effects on the immune system that are dependent on mode of delivery and the dosing strategy, which may have unpredicted effects on immunotherapy. Here we summarize the immune modulating role of temozolomide alone and in combination with immunotherapies such as dendritic cell vaccines, T-cell therapy, and immune checkpoint inhibitors for patients with GBM.


Asunto(s)
Antineoplásicos Alquilantes/uso terapéutico , Neoplasias Encefálicas/tratamiento farmacológico , Glioblastoma/tratamiento farmacológico , Inmunomodulación/efectos de los fármacos , Temozolomida/uso terapéutico , Neoplasias Encefálicas/inmunología , Glioblastoma/inmunología , Humanos , Pronóstico
14.
J Immunol ; 200(8): 2894-2904, 2018 04 15.
Artículo en Inglés | MEDLINE | ID: mdl-29514949

RESUMEN

The transition of effector T cells or memory precursors into distinct long-lived memory T cell subsets is not well understood. Although many molecules made by APCs can contribute to clonal expansion and effector cell differentiation, it is not clear if clonal contraction and memory development is passive or active. Using respiratory virus infection, we found that CD8 T cells that cannot express the TNF family molecule lymphotoxin-like, exhibits inducible expression, competes with HSV glycoprotein D for herpes virus entry mediator, a receptor expressed by T lymphocytes (LIGHT) are unimpaired in their initial response and clonally expand to form effector cell pools. Thereafter, LIGHT-deficient CD8 T cells undergo strikingly enhanced clonal contraction with resultant compromised accumulation of both circulating and tissue-resident memory cells. LIGHT expression at the peak of the effector response regulates the balance of several pro- and antiapoptotic genes, including Akt, and has a preferential impact on the development of the peripheral memory population. These results underscore the importance of LIGHT activity in programming memory CD8 T cell development, and suggest that CD8 effector T cells can dictate their own fate into becoming memory cells by expressing LIGHT.


Asunto(s)
Linfocitos T CD8-positivos/inmunología , Memoria Inmunológica/inmunología , Infecciones del Sistema Respiratorio/inmunología , Miembro 14 de la Superfamilia de Ligandos de Factores de Necrosis Tumoral/inmunología , Virosis/inmunología , Animales , Diferenciación Celular/inmunología , Femenino , Ratones , Ratones Endogámicos C57BL , Infecciones del Sistema Respiratorio/virología
15.
Immunol Cell Biol ; 95(2): 121-128, 2017 02.
Artículo en Inglés | MEDLINE | ID: mdl-27725666

RESUMEN

PTPN22 (protein tyrosine phosphatase non receptor 22) encodes a tyrosine phosphatase that functions as a key regulator of immune homeostasis. In particular, PTPN22 inhibits T-cell receptor signaling and selectively promotes type I interferon responses in myeloid cells. To date, there is little information on the CD8 T-cell-intrinsic role of PTPN22 in response to a viral pathogen. We unexpectedly found that PTPN22-deficient virus-specific CD8 T cells failed to accumulate in wild-type hosts after lymphocytic choriomeningitis virus infection. Lack of PTPN22 expression altered CD8 T-cell activation and antiviral cytokine production, but did not significantly affect the composition of effector and memory cell precursors. Most significantly, in vivo, PTPN22-deficient CD8 T cells showed a profound defect in upregulating STAT-1 after lymphocytic choriomeningitis virus infection and considerably less phosphorylation of STAT-1 in response to IFN-α treatment in vitro compared with their wild-type counterparts. In stark contrast, following transfer into lymphopenic mice, CD8 T-cell expansion and central-like phenotype, was considerably increased in the absence of PTPN22. Collectively, our results suggest that PTPN22 has dual roles in T-cell clonal expansion and effector function; whereas it promotes antigen-driven responses during acute infection by positively regulating interferon signaling in T cells, PTPN22 inhibits homeostatic-driven proliferation.


Asunto(s)
Linfocitos T CD8-positivos/inmunología , Homeostasis , Virus de la Coriomeningitis Linfocítica/fisiología , Proteína Tirosina Fosfatasa no Receptora Tipo 22/metabolismo , Animales , Biomarcadores/metabolismo , Proliferación Celular , Citocinas/metabolismo , Interferón Tipo I/metabolismo , Activación de Linfocitos/inmunología , Coriomeningitis Linfocítica/inmunología , Coriomeningitis Linfocítica/virología , Ratones Endogámicos C57BL , Fosforilación , Proteína Tirosina Fosfatasa no Receptora Tipo 22/deficiencia , Factor de Transcripción STAT1/metabolismo , Transducción de Señal , Regulación hacia Arriba
16.
J Exp Med ; 213(13): 2897-2911, 2016 12 12.
Artículo en Inglés | MEDLINE | ID: mdl-27879287

RESUMEN

How tissue-specific anatomical distribution and phenotypic specialization are linked to protective efficacy of memory T cells against reinfection is unclear. Here, we show that lung environmental cues program recently recruited central-like memory cells with migratory potentials for their tissue-specific functions during lethal respiratory virus infection. After entering the lung, some central-like cells retain their original CD27hiCXCR3hi phenotype, enabling them to localize near the infected bronchiolar epithelium and airway lumen to function as the first line of defense against pathogen encounter. Others, in response to local cytokine triggers, undergo a secondary program of differentiation that leads to the loss of CXCR3, migration arrest, and clustering within peribronchoarterial areas and in interalveolar septa. Here, the immune system adapts its response to prevent systemic viral dissemination and mortality. These results reveal the striking and unexpected spatial organization of central- versus effector-like memory cells within the lung and how cooperation between these two subsets contributes to host defense.


Asunto(s)
Linfocitos T CD8-positivos/inmunología , Memoria Inmunológica , Alveolos Pulmonares/inmunología , Infecciones del Sistema Respiratorio/inmunología , Infecciones por Respirovirus/inmunología , Respirovirus/inmunología , Animales , Linfocitos T CD8-positivos/patología , Femenino , Ratones , Ratones Transgénicos , Alveolos Pulmonares/patología , Infecciones del Sistema Respiratorio/genética , Infecciones del Sistema Respiratorio/patología , Infecciones del Sistema Respiratorio/virología , Infecciones por Respirovirus/genética , Infecciones por Respirovirus/patología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...