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.
Heliyon ; 10(1): e23682, 2024 Jan 15.
Artículo en Inglés | MEDLINE | ID: mdl-38187312

RESUMEN

Cardiovascular diseases are the leading cause of death worldwide, with heart failure being a complex condition that affects millions of individuals. Single-nucleus RNA sequencing has recently emerged as a powerful tool for unraveling the molecular mechanisms behind cardiovascular diseases. This cutting-edge technology enables the identification of molecular signatures, intracellular networks, and spatial relationships among cardiac cells, including cardiomyocytes, mast cells, lymphocytes, macrophages, lymphatic endothelial cells, endocardial cells, endothelial cells, epicardial cells, adipocytes, fibroblasts, neuronal cells, pericytes, and vascular smooth muscle cells. Despite these advancements, the discovery of essential therapeutic targets and drugs for precision cardiology remains a challenge. To bridge this gap, we conducted comprehensive in silico analyses of single-nucleus RNA sequencing data, functional enrichment, protein interactome network, and identification of the shortest pathways to physiological phenotypes. This integrated multi-omics analysis generated CardiOmics signatures, which allowed us to pinpoint three therapeutically actionable targets (ADRA1A1, PPARG, and ROCK2) and 15 effective drugs, including adrenergic receptor agonists, adrenergic receptor antagonists, norepinephrine precursors, PPAR receptor agonists, and Rho-associated kinase inhibitors, involved in late-stage cardiovascular disease clinical trials.

2.
Sci Rep ; 12(1): 11100, 2022 06 30.
Artículo en Inglés | MEDLINE | ID: mdl-35773405

RESUMEN

Many primary-tumor subregions exhibit low levels of molecular oxygen and restricted access to nutrients due to poor vascularization in the tissue, phenomenon known as hypoxia. Hypoxic tumors are able to regulate the expression of certain genes and signaling molecules in the microenvironment that shift it towards a more aggressive phenotype. The transcriptional landscape of the tumor favors malignant transformation of neighboring cells and their migration to distant sites. Herein, we focused on identifying key proteins that participate in the signaling crossroads between hypoxic environment and metastasis progression that remain poorly defined. To shed light on these mechanisms, we performed an integrated multi-omics analysis encompassing genomic/transcriptomic alterations of hypoxia-related genes and Buffa hypoxia scores across 17 pancarcinomas taken from the PanCancer Atlas project from The Cancer Genome Atlas consortium, protein-protein interactome network, shortest paths from hypoxia-related proteins to metastatic and angiogenic phenotypes, and drugs involved in current clinical trials to treat the metastatic disease. As results, we identified 30 hypoxia-related proteins highly involved in metastasis and angiogenesis. This set of proteins, validated with the MSK-MET Project, could represent key targets for developing therapies. The upregulation of mRNA was the most prevalent alteration in all cancer types. The highest frequencies of genomic/transcriptomic alterations and hypoxia score belonged to tumor stage 4 and positive metastatic status in all pancarcinomas. The most significantly associated signaling pathways were HIF-1, PI3K-Akt, thyroid hormone, ErbB, FoxO, mTOR, insulin, MAPK, Ras, AMPK, and VEGF. The interactome network revealed high-confidence interactions among hypoxic and metastatic proteins. The analysis of shortest paths revealed several ways to spread metastasis and angiogenesis from hypoxic proteins. Lastly, we identified 23 drugs enrolled in clinical trials focused on metastatic disease treatment. Six of them were involved in advanced-stage clinical trials: aflibercept, bevacizumab, cetuximab, erlotinib, ipatasertib, and panitumumab.


Asunto(s)
Neoplasias , Fosfatidilinositol 3-Quinasas , Hipoxia de la Célula/genética , Línea Celular Tumoral , Humanos , Hipoxia/genética , Hipoxia/metabolismo , Subunidad alfa del Factor 1 Inducible por Hipoxia/metabolismo , Metástasis de la Neoplasia , Neoplasias/patología , Neovascularización Patológica/tratamiento farmacológico , Neovascularización Patológica/genética , Fosfatidilinositol 3-Quinasas/metabolismo , Transducción de Señal , Microambiente Tumoral
3.
Front Pharmacol ; 13: 833174, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-35422702

RESUMEN

Background: It is imperative to identify drugs that allow treating symptoms of severe COVID-19. Respiratory failure is the main cause of death in severe COVID-19 patients, and the host inflammatory response at the lungs remains poorly understood. Methods: Therefore, we retrieved data from post-mortem lungs from COVID-19 patients and performed in-depth in silico analyses of single-nucleus RNA sequencing data, inflammatory protein interactome network, and shortest pathways to physiological phenotypes to reveal potential therapeutic targets and drugs in advanced-stage COVID-19 clinical trials. Results: Herein, we analyzed transcriptomics data of 719 inflammatory response genes across 19 cell types (116,313 nuclei) from lung autopsies. The functional enrichment analysis of the 233 significantly expressed genes showed that the most relevant biological annotations were inflammatory response, innate immune response, cytokine production, interferon production, macrophage activation, blood coagulation, NLRP3 inflammasome complex, and the TLR, JAK-STAT, NF-κB, TNF, oncostatin M signaling pathways. Subsequently, we identified 34 essential inflammatory proteins with both high-confidence protein interactions and shortest pathways to inflammation, cell death, glycolysis, and angiogenesis. Conclusion: We propose three small molecules (baricitinib, eritoran, and montelukast) that can be considered for treating severe COVID-19 symptoms after being thoroughly evaluated in COVID-19 clinical trials.

4.
Med. interna Méx ; 35(5): 802-806, sep.-oct. 2019. tab, graf
Artículo en Español | LILACS-Express | LILACS | ID: biblio-1250274

RESUMEN

Resumen: La hemoglobinuria paroxística nocturna es un trastorno adquirido de las células madre hematopoyéticas que se caracteriza por episodios de hemólisis intravascular. Aunque es una enfermedad poco frecuente, afecta en su mayor parte a adultos jóvenes, sin distinción de sexo. Comunicamos el caso de un paciente de 32 años de edad, que acudió a consulta con cuadro clínico de palidez, ictericia, hemoglobinuria y dolor en el hipocondrio derecho. El estudio de citometría de flujo de médula ósea reportó la ausencia de marcadores CD55 y CD59, indicativos del diagnóstico de hemoglobinuria paroxística nocturna, además de una imagen tomográfica hipodensa en el hígado compatible con absceso. En la bibliografía médica éste es el primer caso en el que se describe la coexistencia de estas dos afecciones.


Abstract: Paroxysmal nocturnal hemoglobinuria is an acquired disorder of hematopoietic stem cells characterized by episodes of intravascular hemolysis. Although it is a rare disease, it mostly affects young adults, regardless of sex. We present the case of a 32-year-old man with acute symptoms of paleness, jaundice, hemoglobinuria and pain in the right hypochondrium. The study of flow cytometry of bone marrow reported the absence of CD55 and CD59 markers, diagnostic indicators of nocturnal paroxysmal hemoglobinuria in addition to a hypodense tomographic image in the liver compatible with abscess. In the medical literature, this is the first case in which the coexistence of these two medical conditions is described.

SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA