Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 4 de 4
Filtrar
Más filtros











Intervalo de año de publicación
1.
Biomark Res ; 11(1): 99, 2023 Nov 17.
Artículo en Inglés | MEDLINE | ID: mdl-37978566

RESUMEN

While the field of precision oncology is rapidly expanding and more targeted options are revolutionizing cancer treatment paradigms, therapeutic resistance particularly to immunotherapy remains a pressing challenge. This can be largely attributed to the dynamic tumor-stroma interactions that continuously alter the microenvironment. While to date most advancements have been made through examining the clinical utility of tissue-based biomarkers, their invasive nature and lack of a holistic representation of the evolving disease in a real-time manner could result in suboptimal treatment decisions. Thus, using minimally-invasive approaches to identify biomarkers that predict and monitor treatment response as well as alert to the emergence of recurrences is of a critical need. Currently, research efforts are shifting towards developing liquid biopsy-based biomarkers obtained from patients over the course of disease. Liquid biopsy represents a unique opportunity to monitor intercellular communication within the tumor microenvironment which could occur through the exchange of extracellular vesicles (EVs). EVs are lipid bilayer membrane nanoscale vesicles which transfer a plethora of biomolecules that mediate intercellular crosstalk, shape the tumor microenvironment, and modify drug response. The capture of EVs using innovative approaches, such as microfluidics, magnetic beads, and aptamers, allow their analysis via high throughput multi-omics techniques and facilitate their use for biomarker discovery. Artificial intelligence, using machine and deep learning algorithms, is advancing multi-omics analyses to uncover candidate biomarkers and predictive signatures that are key for translation into clinical trials. With the increasing recognition of the role of EVs in mediating immune evasion and as a valuable biomarker source, these real-time snapshots of cellular communication are promising to become an important tool in the field of precision oncology and spur the recognition of strategies to block resistance to immunotherapy. In this review, we discuss the emerging role of EVs in biomarker research describing current advances in their isolation and analysis techniques as well as their function as mediators in the tumor microenvironment. We also highlight recent lung cancer and melanoma studies that point towards their application as predictive biomarkers for immunotherapy and their potential clinical use in precision immuno-oncology.

2.
Cell Physiol Biochem ; 54(4): 517-537, 2020 May 20.
Artículo en Inglés | MEDLINE | ID: mdl-32428391

RESUMEN

BACKGROUND/AIMS: Src kinase family members, including c-Src, are involved in numerous signaling pathways and have been observed inside different cellular compartments. Notably, c-Src modulates carbohydrate and fatty acid metabolism and is involved in the metabolic rewiring of cancer cells. This kinase is found within mitochondria where it targets different proteins to impact on the organelle functions and overall metabolism. Surprisingly, no global metabolic characterization of Src has been performed although c-Src knock-out mice have been available for 30 years. Considering that c-Src is sensitive to various metabolites, c-Src might represent a crucial player in metabolic adjustments induced by nutrient stress. The aim of this work was to characterize the impact of c-Src on mitochondrial activity and overall metabolism using multi-omic characterization. METHODS: Src+/+ and Src-/- mice were fed ad libitum or fasted during 24h and were then analyzed using multi-omics. RESULTS: We observed that deletion of c-Src is linked to lower phosphorylation of Y412-NDUFA8, inhibition of oxygen consumption and accumulation of metabolites involved in glycolysis, TCA cycle and amino acid metabolism in mice fed ad libitum. Finally, metabolomics and (phosphotyrosine) proteomics are differently impacted by Src according to nutrient availability. CONCLUSION: The findings presented here highlight that c-Src reduces mitochondrial metabolism and impacts the metabolic adjustment induced by nutrient stress.


Asunto(s)
Mitocondrias/metabolismo , Fosfotirosina/metabolismo , Proteoma/metabolismo , Familia-src Quinasas/metabolismo , Animales , Encéfalo/metabolismo , Cromatografía Liquida , Ciclo del Ácido Cítrico/genética , Cromatografía de Gases y Espectrometría de Masas , Glucólisis/genética , Riñón/metabolismo , Ratones , Ratones Noqueados , Mitocondrias/enzimología , Mitocondrias/genética , Mitocondrias Hepáticas/genética , Mitocondrias Hepáticas/metabolismo , Nutrientes/metabolismo , Fosforilación , Fosfotirosina/genética , Proteómica , Espectrometría de Masas en Tándem , Familia-src Quinasas/genética
3.
Cell Death Dis ; 10(12): 940, 2019 12 09.
Artículo en Inglés | MEDLINE | ID: mdl-31819039

RESUMEN

High levels and activity of Src kinase are common among breast cancer subtypes, and several inhibitors of the kinase are currently tested in clinical trials. Alterations in mitochondrial activity is also observed among the different types of breast cancer. Src kinase is localized in several subcellular compartments, including mitochondria where it targets several proteins to modulate the activity of the organelle. Although the subcellular localization of other oncogenes modulates the potency of known treatments, nothing is known about the specific role of intra-mitochondrial Src (mtSrc) in breast cancer. The aim of this work was to determine whether mtSrc kinase has specific impact on breast cancer cells. We first observed that activity of mtSrc is higher in breast cancer cells of the triple negative subtype. Over-expression of Src specifically targeted to mitochondria reduced mtDNA levels, mitochondrial membrane potential and cellular respiration. These alterations of mitochondrial functions led to lower cellular viability, shorter cell cycle and increased invasive capacity. Proteomic analyses revealed that mtSrc targets the mitochondrial single-stranded DNA-binding protein, a regulator of mtDNA replication. Our findings suggest that mtSrc promotes aggressiveness of breast cancer cells via phosphorylation of mitochondrial single-stranded DNA-binding protein leading to reduced mtDNA levels and mitochondrial activity. This study highlights the importance of considering the subcellular localization of Src kinase in the development of potent therapy for breast cancer.


Asunto(s)
Neoplasias de la Mama/metabolismo , Mitocondrias/metabolismo , Familia-src Quinasas/metabolismo , Adenosina Trifosfato/biosíntesis , Apoptosis/genética , Neoplasias de la Mama/patología , Movimiento Celular/genética , Proliferación Celular/genética , Respiración de la Célula/genética , ADN Mitocondrial/metabolismo , Proteínas de Unión al ADN/metabolismo , Femenino , Humanos , Células MCF-7 , Potencial de la Membrana Mitocondrial/genética , Fosforilación/genética , Especies Reactivas de Oxígeno/metabolismo , Transfección , Familia-src Quinasas/genética
4.
Bioessays ; 39(12)2017 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-29058339

RESUMEN

CB1 receptors are functionally present within brain mitochondria (mtCB1), although they are usually considered specifically targeted to plasma membrane. Acute activation of mtCB1 alters mitochondrial ATP generation, synaptic transmission, and memory performance. However, the detailed mechanism linking disrupted mitochondrial metabolism and synaptic transmission is still uncharacterized. CB1 receptors are among the most abundant G protein-coupled receptors in the brain and impact on several processes, including fear coping, anxiety, stress, learning, and memory. Mitochondria perform several key physiological processes for neuronal homeostasis, including production of ATP and reactive oxygen species, calcium buffering, metabolism of neurotransmitters, and apoptosis. It is therefore possible that acute activation of mtCB1 impacts on these different mitochondrial functions to modulate synaptic transmission. In reviewing and integrating across the literature in this area, we describe the possible mechanisms involved in the regulation of brain physiology by mtCB1 receptors.


Asunto(s)
Encéfalo/metabolismo , Mitocondrias/genética , Neuronas/metabolismo , Receptor Cannabinoide CB1/metabolismo , Transmisión Sináptica/fisiología , Adenosina Trifosfato/biosíntesis , Animales , Ansiedad/genética , Ansiedad/metabolismo , Ansiedad/fisiopatología , Encéfalo/citología , Calcio/metabolismo , Miedo/fisiología , Humanos , Memoria/fisiología , Mitocondrias/metabolismo , Plasticidad Neuronal/fisiología , Neuronas/citología , Especies Reactivas de Oxígeno/metabolismo , Receptor Cannabinoide CB1/genética , Estrés Psicológico/genética , Estrés Psicológico/metabolismo , Estrés Psicológico/fisiopatología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA