Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 23
Filtrar
Más filtros












Base de datos
Intervalo de año de publicación
1.
Sci Rep ; 14(1): 5758, 2024 03 08.
Artículo en Inglés | MEDLINE | ID: mdl-38459035

RESUMEN

Two types of immunity, humoral and cellular, offer protection against COVID. Humoral protection, contributed by circulating neutralizing antibodies, can provide immediate protection but decays more quickly than cellular immunity and can lose effectiveness in the face of mutation and drift in the SARS-CoV-2 spike protein. Therefore, population-level seroprevalence surveys used to estimate population-level immunity may underestimate the degree to which a population is protected against COVID. In early 2021, before India began its vaccination campaign, we tested for humoral and cellular immunity to SARS-Cov-2 in representative samples of slum and non-slum populations in Bangalore, India. We found that 29.7% of samples (unweighted) had IgG antibodies to the spike protein and 15.5% had neutralizing antibodies, but at up to 46% showed evidence of cellular immunity. We also find that prevalence of cellular immunity is significantly higher in slums than in non-slums. These findings suggest (1) that a significantly larger proportion of the population in Bangalore, India, had cellular immunity to SARS-CoV-2 than had humoral immunity, as measured by serological surveys, and (2) that low socio-economic status communities display higher frequency of cellular immunity, likely because of greater exposure to infection due to population density.


Asunto(s)
COVID-19 , SARS-CoV-2 , Glicoproteína de la Espiga del Coronavirus , Humanos , India/epidemiología , COVID-19/epidemiología , Estudios Seroepidemiológicos , Inmunidad Celular , Anticuerpos Neutralizantes , Inmunidad Humoral , Anticuerpos Antivirales , Vacunación
2.
Rev Cardiovasc Med ; 23(11): 381, 2022 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-39076184

RESUMEN

Cardiovascular disease (CVD), a broad-spectrum term comprising coronary artery disease, stroke, hypertension, and heart failure, presents as one of the most significant strains on global healthcare systems. Coronary artery disease, caused by atherosclerosis, has various modifiable risk factors such as dietary changes and exercise. Since these risk factors are found to be linked to oxidative stress and inflammations, the dietary supplementation with vitamins' role in treating and preventing the diseases has been of much debate. With various vitamins having anti-inflammatory and antioxidative properties, studies have explored their correlation with cardiovascular health. Therefore, this narrative review explores and evaluates the benefits and risks of all vitamin supplementations in patients with CVD and provides future recommendations.

3.
Sci Adv ; 7(40): eabh3243, 2021 10.
Artículo en Inglés | MEDLINE | ID: mdl-34586841

RESUMEN

Mutant isocitrate-dehydrogenase 1 (mIDH1) synthesizes the oncometabolite 2-hydroxyglutarate (2HG), which elicits epigenetic reprogramming of the glioma cells' transcriptome by inhibiting DNA and histone demethylases. We show that the efficacy of immune-stimulatory gene therapy (TK/Flt3L) is enhanced in mIDH1 gliomas, due to the reprogramming of the myeloid cells' compartment infiltrating the tumor microenvironment (TME). We uncovered that the immature myeloid cells infiltrating the mIDH1 TME are mainly nonsuppressive neutrophils and preneutrophils. Myeloid cell reprogramming was triggered by granulocyte colony-stimulating factor (G-CSF) secreted by mIDH1 glioma stem/progenitor-like cells. Blocking G-CSF in mIDH1 glioma­bearing mice restores the inhibitory potential of the tumor-infiltrating myeloid cells, accelerating tumor progression. We demonstrate that G-CSF reprograms bone marrow granulopoiesis, resulting in noninhibitory myeloid cells within mIDH1 glioma TME and enhancing the efficacy of immune-stimulatory gene therapy.

4.
Methods Enzymol ; 632: 215-228, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-32000897

RESUMEN

Despite advances in uncovering the molecular mechanisms that mediate glioma progression and the implementation of novel therapeutic modalities, patients' prognosis remains dismal. This is due to both systemic and local tumor induced immune suppression. We are particularly interested in the role played by infiltrating immunosuppressive myeloid derived suppressor cells (MDSCs) in the glioma tumor microenvironment (TME). This immunosuppressive TME also interferes with the effectiveness of immunotherapies against glioma. Development of multipronged treatment approaches is imperative when aiming to generate a robust anti-glioma immune response. Evaluating the inhibitory potential of MDSCs within the TME is an important aspect for developing effective treatments for glioma. Herein, we discuss methodology to assess the inhibitory effects of MDSCs isolated from the TME using a mouse glioma model.


Asunto(s)
Neoplasias Encefálicas/inmunología , Glioma/inmunología , Células Supresoras de Origen Mieloide/inmunología , Linfocitos T/inmunología , Microambiente Tumoral , Animales , Neoplasias Encefálicas/patología , Proliferación Celular , Separación Celular/métodos , Células Cultivadas , Modelos Animales de Enfermedad , Femenino , Glioma/patología , Tolerancia Inmunológica , Ratones , Ratones Endogámicos C57BL , Células Supresoras de Origen Mieloide/patología , Linfocitos T/patología
5.
Methods Enzymol ; 631: 91-106, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-31948569

RESUMEN

Diffuse Gliomas represent 80% of brain tumors with an average survival of the most aggressive form glioblastoma (GBM) 15-22 months from the time of diagnosis. The current standard of care includes tumor resection, chemotherapy and radiation, nevertheless, the incidence of recurrence remains high and there is a critical need for developing new therapeutic strategies. T-cell mediated immunotherapy that triggers an anti-tumor T cell-mediated memory response is a promising approach since it will not only attack the primary tumor but also prevent recurrence. Multiple immunotherapeutic strategies against glioma are currently being tested in clinical trials. We have developed an immune-mediated gene therapy (Thymidine kinase plus Fms-like tyrosine kinase 3 ligand: TK/Flt3L) which induces a robust anti-tumor T cell response leading to tumor regression, long-term survival and immunological memory in GBM models. Efficacy of the anti-glioma T cell therapy is determined by anti-tumor specific effector T cells. Therefore, assessing effector T cell activation status and function are critical readouts for determining the effectiveness of the therapy. Here, we detail methodologies to evaluate tumor specific T-cell responses using a genetically engineered Sleeping Beauty transposase-mediated glioma model. We first describe the glioma model and the generation of neurospheres (NS) that express the surrogate antigen cOVA. Then, we describe functional assays to determine anti-tumor T-cell response.


Asunto(s)
Antígenos de Neoplasias , Neoplasias Encefálicas/terapia , Pruebas Inmunológicas de Citotoxicidad/métodos , Terapia Genética/métodos , Glioma/terapia , Linfocitos Infiltrantes de Tumor/inmunología , Microambiente Tumoral/inmunología , Animales , Neoplasias Encefálicas/inmunología , Glioma/inmunología , Memoria Inmunológica , Inmunoterapia/métodos , Proteínas de la Membrana , Ratones , Linfocitos T/metabolismo , Timidina Quinasa
6.
ACS Nano ; 13(2): 1365-1384, 2019 02 26.
Artículo en Inglés | MEDLINE | ID: mdl-30721028

RESUMEN

Glioblastoma multiforme (GBM) is an aggressive primary brain tumor, for which there is no cure. Treatment effectiveness for GBM has been limited due to tumor heterogeneity, an immunosuppressive tumor microenvironment (TME), and the presence of the blood-brain barrier, which hampers the transport of chemotherapeutic compounds to the central nervous system (CNS). High-density lipoprotein (HDL)-mimicking nanodiscs hold considerable promise to achieve delivery of bioactive compounds into tumors. Herein, we tested the ability of synthetic HDL nanodiscs to deliver chemotherapeutic agents to the GBM microenvironment and elicit tumor regression. To this end, we developed chemo-immunotherapy delivery vehicles based on sHDL nanodiscs loaded with CpG, a Toll-like receptor 9 (TLR9) agonist, together with docetaxel (DTX), a chemotherapeutic agent, for targeting GBM. Our data show that delivery of DTX-sHDL-CpG nanodiscs into the tumor mass elicited tumor regression and antitumor CD8+ T cell responses in the brain TME. We did not observe any overt off-target side effects. Furthermore, the combination of DTX-sHDL-CpG treatment with radiation (IR), which is the standard of care for GBM, resulted in tumor regression and long-term survival in 80% of GBM-bearing animals. Mice remained tumor-free upon tumor cell rechallenge in the contralateral hemisphere, indicating the development of anti-GBM immunological memory. Collectively, these data indicate that sHDL nanodiscs constitute an effective drug delivery platform for the treatment of GBM, resulting in tumor regression, long-term survival, and immunological memory when used in combination with IR. The proposed delivery platform has significant potential for clinical translation.


Asunto(s)
Glioblastoma/tratamiento farmacológico , Glioblastoma/terapia , Inmunoterapia/métodos , Lipoproteínas HDL/química , Lipoproteínas HDL/uso terapéutico , Animales , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Docetaxel/química , Docetaxel/uso terapéutico , Sistemas de Liberación de Medicamentos/métodos , Femenino , Citometría de Flujo , Humanos , Inmunohistoquímica , Lomustina/química , Lomustina/uso terapéutico , Ratones , Modelos Biológicos , Paclitaxel/química , Paclitaxel/uso terapéutico , Ratas , Linfocitos T/efectos de los fármacos , Linfocitos T/metabolismo
7.
Sci Transl Med ; 11(479)2019 02 13.
Artículo en Inglés | MEDLINE | ID: mdl-30760578

RESUMEN

Patients with glioma whose tumors carry a mutation in isocitrate dehydrogenase 1 (IDH1R132H) are younger at diagnosis and live longer. IDH1 mutations co-occur with other molecular lesions, such as 1p/19q codeletion, inactivating mutations in the tumor suppressor protein 53 (TP53) gene, and loss-of-function mutations in alpha thalassemia/mental retardation syndrome X-linked gene (ATRX). All adult low-grade gliomas (LGGs) harboring ATRX loss also express the IDH1R132H mutation. The current molecular classification of LGGs is based, partly, on the distribution of these mutations. We developed a genetically engineered mouse model harboring IDH1R132H, TP53 and ATRX inactivating mutations, and activated NRAS G12V. Previously, we established that ATRX deficiency, in the context of wild-type IDH1, induces genomic instability, impairs nonhomologous end-joining DNA repair, and increases sensitivity to DNA-damaging therapies. In this study, using our mouse model and primary patient-derived glioma cultures with IDH1 mutations, we investigated the function of IDH1R132H in the context of TP53 and ATRX loss. We discovered that IDH1R132H expression in the genetic context of ATRX and TP53 gene inactivation (i) increases median survival in the absence of treatment, (ii) enhances DNA damage response (DDR) via epigenetic up-regulation of the ataxia-telangiectasia-mutated (ATM) signaling pathway, and (iii) elicits tumor radioresistance. Accordingly, pharmacological inhibition of ATM or checkpoint kinases 1 and 2, essential kinases in the DDR, restored the tumors' radiosensitivity. Translation of these findings to patients with IDH1132H glioma harboring TP53 and ATRX loss could improve the therapeutic efficacy of radiotherapy and, consequently, patient survival.


Asunto(s)
Daño del ADN/genética , Epigénesis Genética , Glioma/genética , Isocitrato Deshidrogenasa/genética , Mutación/genética , Proteínas Supresoras de Tumor/genética , Regulación hacia Arriba/genética , Animales , Proteínas de la Ataxia Telangiectasia Mutada/metabolismo , Neoplasias Encefálicas/genética , Neoplasias Encefálicas/patología , Diferenciación Celular , Metilación de ADN/genética , Reparación del ADN/genética , Modelos Animales de Enfermedad , Ontología de Genes , Genoma , Glioma/patología , Histonas/metabolismo , Humanos , Ratones , Oligodendroglía/patología , Tolerancia a Radiación , Transducción de Señal , Análisis de Supervivencia
8.
Oncoimmunology ; 7(3): e1408750, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-29399415

RESUMEN

Tumors are associated with expansion of immunosuppressive cells such as tumor associated macrophages (TAMs), regulatory T cells (Tregs) and myeloid derived suppressor cells (MDSCs). These cells promote tumor growth, angiogenesis, metastasis and immune escape. Cancer patients frequently present symptoms such as anemia, leukocytosis and/or cytopenia; associated with poor prognosis. To uncover tumor-mediated hematopoietic abnormalities and identify novel targets that can be harnessed to improve tumor-specific immune responses, we investigated the hematopoietic stem and progenitor cell compartment in melanoma bearing mice. We show that melanoma growth results in expansion of myeloid lineages such as MDSCs, macrophages and DCs along with a reduction in mature RBCs and platelets. Mature B lymphocytes in the blood and BM of melanoma mice were also reduced. Mice bearing melanoma showed extramedullary hematopoiesis in the spleen. Increased expansion of myeloid lineages occurred directly at the level of stem and progenitor cells. The reduction in mature B lymphocytes resulted from a block at the Pro-B cell stage in the bone marrow. Addition of recombinant IL-3 to bone marrow cells resulted in the expansion of committed myeloid progenitors including common myeloid precursors, granulocyte-monocyte precursors and megakaryocyte-erythrocyte precursors. In vivo, IL-3 receptor stimulation in melanoma bearing mice using an IL-3 antibody also resulted in a robust expansion of committed myeloid progenitors and hematopoietic stem cells. Collectively our findings demonstrate that tumor growth plays a pivotal role in reprogramming the host immune system by impacting hematopoiesis directly at the level of stem cell compartment.

9.
Immunotherapy ; 10(4): 317-339, 2018 02 01.
Artículo en Inglés | MEDLINE | ID: mdl-29421984

RESUMEN

There is a large unmet need for effective therapeutic approaches for glioma, the most malignant brain tumor. Clinical and preclinical studies have enormously expanded our knowledge about the molecular aspects of this deadly disease and its interaction with the host immune system. In this review we highlight the wide array of immunotherapeutic interventions that are currently being tested in glioma patients. Given the molecular heterogeneity, tumor immunoediting and the profound immunosuppression that characterize glioma, it has become clear that combinatorial approaches targeting multiple pathways tailored to the genetic signature of the tumor will be required in order to achieve optimal therapeutic efficacy.


Asunto(s)
Neoplasias Encefálicas/terapia , Glioma/terapia , Inmunoterapia/métodos , Encéfalo/inmunología , Neoplasias Encefálicas/inmunología , Glioma/inmunología , Humanos , Inmunoterapia/tendencias
10.
Clin Immunol ; 189: 34-42, 2018 04.
Artículo en Inglés | MEDLINE | ID: mdl-27777083

RESUMEN

Various preclinical studies have demonstrated that the success of immunotherapeutic strategies in inhibiting tumor progression in animal models of Glioblastoma multiforme (GBM). It is also evident that tumor-induced immune suppression drastically impacts the efficacy of immune based therapies. Among the mechanisms employed by GBM to induce immunosuppression is the accumulation of regulatory T cells (Tregs) and Myeloid derived suppressor cells (MDSCs). Advancing our understanding about the pathways regulating the expansion, accumulation and activity of MDSCs will allow for the development of therapies aimed at abolishing the inhibitory effect of these cells on immunotherapeutic approaches. In this review, we have focused on the origin, expansion and immunosuppressive mechanisms of MDSCs in animal models and human cancer, in particular GBM.


Asunto(s)
Inmunoterapia/métodos , Células Mieloides/inmunología , Células Supresoras de Origen Mieloide/inmunología , Neoplasias/terapia , Microambiente Tumoral/inmunología , Animales , Neoplasias Encefálicas/inmunología , Neoplasias Encefálicas/patología , Neoplasias Encefálicas/terapia , Glioblastoma/inmunología , Glioblastoma/patología , Glioblastoma/terapia , Humanos , Neoplasias/inmunología , Neoplasias/patología , Linfocitos T Reguladores/inmunología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...