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.
Folia Med (Plovdiv) ; 66(2): 282-286, 2024 Apr 30.
Artículo en Inglés | MEDLINE | ID: mdl-38690826

RESUMEN

The diagnosis of intrathoracic non-tuberculous mycobacteriosis (NTM) is challenging. We report a case of a pediatric pulmonary NTM with endobronchial lesion and lymphadenitis in a child with HIV infection diagnosed by bronchoscopic biopsy, EBUS-TBNA and probe-based confocal laser endomicroscopy (pCLE). The pCLE showed a large number of highly fluorescent cells and zones of density and disorganized elastin fibers at alveolar areas. A combination of diagnostic endoscopic procedures is required to establish the diagnosis of NTM.


Asunto(s)
Broncoscopía , Biopsia por Aspiración con Aguja Fina Guiada por Ultrasonido Endoscópico , Infecciones por VIH , Microscopía Confocal , Infecciones por Mycobacterium no Tuberculosas , Humanos , Broncoscopía/métodos , Niño , Microscopía Confocal/métodos , Infecciones por Mycobacterium no Tuberculosas/diagnóstico , Infecciones por Mycobacterium no Tuberculosas/patología , Masculino , Infecciones por VIH/complicaciones , Infecciones por VIH/patología , Biopsia/métodos
2.
Artículo en Inglés | MEDLINE | ID: mdl-37675917

RESUMEN

Sjögren's syndrome is systemic autoimmune disease characterized by lymphocytic infiltration of various organs with wide frequency of pulmonary involvement. Diffuse cystic lung disease in Sjögren's syndrome is a rare condition and requires differential diagnosis with other cystic pathologies such as lymphangioleyomiomatosis or Langerhans cell histiocytosis. Probe-based confocal laser endomicroscopy (pCLE) is a method of in vivo investigation of airways and lung tissue on microscopic level during bronchoscopy. We used this method in diffuse cystic lung disease caused by Sjögren's syndrome. The pCLE image showed a large number of fluorescent cells presumably lymphocytes in bronchioles, dilated alveolar spaces with fluid and thin alveolar walls. We think that the presence of the bronchiolar cells pattern can be used to differentiate between the pulmonary manifestations of Sjögren's disease and other cystic lung diseases.

3.
Diagnostics (Basel) ; 13(6)2023 Mar 14.
Artículo en Inglés | MEDLINE | ID: mdl-36980405

RESUMEN

Patients with COVID-19 demonstrate higher rates of cardiovascular complications, including thromboses and thromboembolism. One may suppose that the action of SARS-CoV-2 transforms stable atherosclerotic plaques into unstable status. Cardiovascular complications in COVID-19 may be caused by progressive viral alteration of the blood vessels, including Vasa vasorum. A lethal case of ischemic brain disease caused by cerebral atherosclerosis and exacerbated by a stroke during COVID-19 infection is briefly described. The results of the autopsy showed perivascular lymphocytic infiltration and signs of Vasa vasorum vasculitis with thrombi of adventitial microvasculature. The data discussed in the article are interpreted in the context of the concept giving the important role in atherogenesis to Vasa vasorum.

4.
Am J Physiol Heart Circ Physiol ; 323(6): H1244-H1261, 2022 12 01.
Artículo en Inglés | MEDLINE | ID: mdl-36240436

RESUMEN

Extracellular purine nucleotides and nucleosides released from activated or injured cells influence multiple aspects of cardiac physiology and pathophysiology. Ectonucleoside triphosphate diphosphohydrolase-1 (ENTPD1; CD39) hydrolyzes released nucleotides and thereby regulates the magnitude and duration of purinergic signaling. However, the impact of CD39 activity on post-myocardial infarction (MI) remodeling is incompletely understood. We measured the levels and activity of ectonucleotidases in human left ventricular samples from control and ischemic cardiomyopathy (ICM) hearts and examined the impact of ablation of Cd39 expression on post-myocardial infarction remodeling in mice. We found that human CD39 levels and activity are significantly decreased in ICM hearts (n = 5) compared with control hearts (n = 5). In mice null for Cd39, cardiac function and remodeling are significantly compromised in Cd39-/- mice following myocardial infarction. Fibrotic markers including plasminogen activator inhibitor-1 (PAI-1) expression, fibrin deposition, α-smooth muscle actin (αSMA), and collagen expression are increased in Cd39-/- hearts. Importantly, we found that transforming growth factor ß1 (TGF-ß1) stimulates ATP release and induces Cd39 expression and activity on cardiac fibroblasts, constituting an autocrine regulatory pathway not previously appreciated. Absence of CD39 activity on cardiac fibroblasts exacerbates TGF-ß1 profibrotic responses. Treatment with exogenous ectonucleotidase rescues this profibrotic response in Cd39-/- fibroblasts. Together, these data demonstrate that CD39 has important interactions with TGF-ß1-stimulated autocrine purinergic signaling in cardiac fibroblasts and dictates outcomes of cardiac remodeling following myocardial infarction. Our results reveal that ENTPD1 (CD39) regulates TGF-ß1-mediated fibroblast activation and limits adverse cardiac remodeling following myocardial infarction.NEW & NOTEWORTHY We show that CD39 is a critical modulator of TGF-ß1-mediated fibroblast activation and cardiac remodeling following myocardial infarction via modulation of nucleotide signaling. TGF-ß1-induced CD39 expression generates a negative feedback loop that attenuates cardiac fibroblast activation. In the absence of CD39 activity, collagen deposition is increased, elastin expression is decreased, and diastolic dysfunction is worsened. Treatment with ecto-apyrase attenuates the TGF-ß1-induced profibrotic cardiac fibroblast phenotype, revealing a novel approach to combat post-myocardial infarction cardiac fibrosis.


Asunto(s)
Infarto del Miocardio , Factor de Crecimiento Transformador beta1 , Humanos , Ratones , Animales , Factor de Crecimiento Transformador beta1/metabolismo , Remodelación Ventricular , Miocardio/metabolismo , Fibrosis , Fibroblastos/metabolismo , Colágeno/metabolismo
5.
Sci Rep ; 11(1): 14424, 2021 07 13.
Artículo en Inglés | MEDLINE | ID: mdl-34257356

RESUMEN

Lung adenocarcinoma (ADC) is a heterogeneous group of tumors associated with different survival rates, even when detected at an early stage. Here, we aim to investigate whether CyTOF identifies cellular and molecular predictors of tumor behavior. We developed and validated a CyTOF panel of 34 antibodies in four ADC cell lines and PBMC. We tested our panel in a set of 10 ADCs, classified into long- (LPS) (n = 4) and short-predicted survival (SPS) (n = 6) based on radiomics features. We identified cellular subpopulations of epithelial cancer cells (ECC) and their microenvironment and validated our results by multiplex immunofluorescence (mIF) applied to a tissue microarray (TMA) of LPS and SPS ADCs. The antibody panel captured the phenotypical differences in ADC cell lines and PBMC. LPS ADCs had a higher proportion of immune cells. ECC clusters (ECCc) were identified and uncovered two ADC groups. ECCc with high HLA-DR expression were correlated with CD4+ and CD8+ T cells, with LPS samples being enriched for those clusters. We confirmed a positive correlation between HLA-DR expression on ECC and T cell number by mIF staining on TMA slides. Spatial analysis demonstrated shorter distances from T cells to the nearest ECC in LPS. Our results demonstrate a distinctive cellular profile of ECC and their microenvironment in ADC. We showed that HLA-DR expression in ECC is correlated with T cell infiltration, and that a set of ADCs with high abundance of HLA-DR+ ECCc and T cells is enriched in LPS samples. This suggests new insights into the role of antigen presenting tumor cells in tumorigenesis.


Asunto(s)
Adenocarcinoma del Pulmón , Antígenos HLA-DR , Leucocitos Mononucleares , Linfocitos T CD4-Positivos , Linfocitos T CD8-positivos , Humanos
6.
PLoS One ; 16(6): e0252424, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34101732

RESUMEN

Cancer associated fibroblasts (CAF) play a key role in cancer progression and metastasis. Diminished TGFß response on CAF correlates with poor outcome and recurrence in cancer patients. Mechanisms behind lost TGFß signaling on CAF are poorly understood, but, utilizing MMTV-PyMT mouse model, we have previously demonstrated that in tumor microenvironment myeloid cells, producing adenosine, contribute to downregulated TGFß signaling on CAFs. In the current work, we performed serial in vitro studies to investigate the role of adenosine/TGFß axis in mouse mammary fibroblast functions, i.e., proliferation, protein expression, migration, and contractility. We found that adenosine analog NECA diminished TGFß-induced CCL5 and MMP9 expression. Additionally, we discovered that NECA completely inhibited effect of TGFß to upregulate αSMA, key protein of cytoskeletal rearrangements, necessary for migration and contractility of fibroblasts. Our results show that TGFß increases contractility of mouse mammary fibroblasts and human fibroblast cell lines, and NECA attenuates theses effects. Using pharmacological approach and genetically modified animals, we determined that NECA effects on TGFß pathway occur via A2A/A2B adenosine receptor-AC-PKA dependent manner. Using isolated CD11b+ cells from tumor tissue of CD73-KO and CD39-KO animals in co-culture experiments with ATP and AMP, we confirmed that myeloid cells can affect functions of mammary fibroblasts through adenosine signaling. Our data suggest a novel mechanism of interaction between adenosine and TGFß signaling pathways that can impact phenotype of fibroblasts in a tumor microenvironment.


Asunto(s)
Adenosina/metabolismo , Movimiento Celular/fisiología , Factor de Crecimiento Transformador beta/metabolismo , Adenilil Ciclasas/metabolismo , Animales , Western Blotting , Línea Celular , Movimiento Celular/genética , Células Cultivadas , Fibroblastos/metabolismo , Fibroblastos/fisiología , Citometría de Flujo , Ratones , Reacción en Cadena en Tiempo Real de la Polimerasa , Transducción de Señal/fisiología , Microambiente Tumoral/fisiología
7.
Front Bioinform ; 12021 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-35813245

RESUMEN

Modern technologies designed for tissue structure visualization like brightfield microscopy, fluorescent microscopy, mass cytometry imaging (MCI) and mass spectrometry imaging (MSI) provide large amounts of quantitative and spatial information about cells and tissue structures like vessels, bronchioles etc. Many published reports have demonstrated that the structural features of cells and extracellular matrix (ECM) and their interactions strongly predict disease development and progression. Computational image analysis methods in combination with spatial analysis and machine learning can reveal novel structural patterns in normal and diseased tissue. Here, we have developed a Python package designed for integrated analysis of cells and ECM in a spatially dependent manner. The package performs segmentation, labeling and feature analysis of ECM fibers, combines this information with pre-generated single-cell based datasets and realizes cell-cell and cell-fiber spatial analysis. To demonstrate performance and compatibility of our computational tool, we integrated it with a pipeline designed for cell segmentation, classification, and feature analysis in the KNIME analytical platform. For validation, we used a set of mouse mammary gland tumors and human lung adenocarcinoma tissue samples stained for multiple cellular markers and collagen as the main ECM protein. The developed package provides sufficient performance and precision to be used as a novel method to investigate cell-ECM relationships in the tissue, as well as detect structural patterns correlated with specific disease outcomes.

8.
Front Comput Sci ; 22020 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-32905440

RESUMEN

Open-source software tools are often used for analysis of scientific image data due to their flexibility and transparency in dealing with rapidly evolving imaging technologies. The complex nature of image analysis problems frequently requires many tools to be used in conjunction, including image processing and analysis, data processing, machine learning and deep learning, statistical analysis of the results, visualization, correlation to heterogeneous but related data, and more. However, the development, and therefore application, of these computational tools is impeded by a lack of integration across platforms. Integration of tools goes beyond convenience, as it is impractical for one tool to anticipate and accommodate the current and future needs of every user. This problem is emphasized in the field of bioimage analysis, where various rapidly emerging methods are quickly being adopted by researchers. ImageJ is a popular open-source image analysis platform, with contributions from a global community resulting in hundreds of specialized routines for a wide array of scientific tasks. ImageJ's strength lies in its accessibility and extensibility, allowing researchers to easily improve the software to solve their image analysis tasks. However, ImageJ is not designed for development of complex end-to-end image analysis workflows. Scientists are often forced to create highly specialized and hard-to-reproduce scripts to orchestrate individual software fragments and cover the entire life-cycle of an analysis of an image dataset. KNIME Analytics Platform, a user-friendly data integration, analysis, and exploration workflow system, was designed to handle huge amounts of heterogeneous data in a platform-agnostic, computing environment and has been successful in meeting complex end-to-end demands in several communities, such as cheminformatics and mass spectrometry. Similar needs within the bioimage analysis community led to the creation of the KNIME Image Processing extension which integrates ImageJ into KNIME Analytics Platform, enabling researchers to develop reproducible and scalable workflows, integrating a diverse range of analysis tools. Here we present how users and developers alike can leverage the ImageJ ecosystem via the KNIME Image Processing extension to provide robust and extensible image analysis within KNIME workflows. We illustrate the benefits of this integration with examples, as well as representative scientific use cases.

9.
Cancer Res ; 80(12): 2628-2638, 2020 06 15.
Artículo en Inglés | MEDLINE | ID: mdl-32312837

RESUMEN

TGFß plays a crucial role in the tumor microenvironment by regulating cell-cell and cell-stroma interactions. We previously demonstrated that TGFß signaling on myeloid cells regulates expression of CD73, a key enzyme for production of adenosine, a protumorigenic metabolite implicated in regulation of tumor cell behaviors, immune response, and angiogenesis. Here, using an MMTV-PyMT mouse mammary tumor model, we discovered that deletion of TGFß signaling on myeloid cells (PyMT/TGFßRIILysM) affects extracellular matrix (ECM) formation in tumor tissue, specifically increasing collagen and decreasing fibronectin deposition. These changes were associated with mitigated tumor growth and reduced metastases. Reduced TGFß signaling on fibroblasts was associated with their proximity to CD73+ myeloid cells in tumor tissue. Consistent with these findings, adenosine significantly downregulated TGFß signaling on fibroblasts, an effect regulated by A2A and A2B adenosine receptors. METABRIC dataset analysis revealed that patients with triple-negative breast cancer and basal type harbored a similar signature of adenosine and ECM profiles; high expression of A2B adenosine receptors correlated with decreased expression of Col1 and was associated with poor outcome. Taken together, our studies reveal a new role for TGFß signaling on myeloid cells in tumorigenesis. This discovered cross-talk between TGFß/CD73 on myeloid cells and TGFß signaling on fibroblasts can contribute to ECM remodeling and protumorigenic actions of cancer-associated fibroblasts. SIGNIFICANCE: TGFß signaling on fibroblasts is decreased in breast cancer, correlates with poor prognosis, and appears to be driven by adenosine that accelerates tumor progression and metastasis via ECM remodeling.


Asunto(s)
Adenosina/metabolismo , Matriz Extracelular/patología , Neoplasias Mamarias Experimentales/patología , Células Mieloides/metabolismo , Factor de Crecimiento Transformador beta/metabolismo , Neoplasias de la Mama Triple Negativas/patología , 5'-Nucleotidasa/metabolismo , Adulto , Anciano , Animales , Mama/patología , Fibroblastos Asociados al Cáncer/metabolismo , Carcinogénesis , Conjuntos de Datos como Asunto , Femenino , Perfilación de la Expresión Génica , Humanos , Estimación de Kaplan-Meier , Glándulas Mamarias Animales/patología , Ratones , Ratones Transgénicos , Persona de Mediana Edad , Receptor de Adenosina A2B/metabolismo , Transducción de Señal , Neoplasias de la Mama Triple Negativas/mortalidad
10.
ILAR J ; 59(1): 51-65, 2018 12 01.
Artículo en Inglés | MEDLINE | ID: mdl-30462242

RESUMEN

For decades, histopathology with routine hematoxylin and eosin staining has been and remains the gold standard for reaching a morphologic diagnosis in tissue samples from humans and veterinary species. However, within the past decade, there has been exponential growth in advanced techniques for in situ tissue biomarker imaging that bridge the divide between anatomic and molecular pathology. It is now possible to simultaneously observe localization and expression magnitude of multiple protein, nucleic acid, and molecular targets in tissue sections and apply machine learning to synthesize vast, image-derived datasets. As these technologies become more sophisticated and widely available, a team-science approach involving subspecialists with medical, engineering, and physics backgrounds is critical to upholding quality and validity in studies generating these data. The purpose of this manuscript is to detail the scientific premise, tools and training, quality control, and data collection and analysis considerations needed for the most prominent advanced imaging technologies currently applied in tissue sections: immunofluorescence, in situ hybridization, laser capture microdissection, matrix-assisted laser desorption ionization imaging mass spectrometry, and spectroscopic/optical methods. We conclude with a brief overview of future directions for ex vivo and in vivo imaging techniques.


Asunto(s)
Biomarcadores , Animales , Humanos , Inmunohistoquímica , Hibridación in Situ , Microscopía Fluorescente , Control de Calidad , Espectrometría de Masa por Láser de Matriz Asistida de Ionización Desorción
11.
Cancer Res ; 78(21): 6086-6097, 2018 11 01.
Artículo en Inglés | MEDLINE | ID: mdl-30154147

RESUMEN

Abnormalities in nuclear shape are a well-known feature of cancer, but their contribution to malignant progression remains poorly understood. Here, we show that depletion of the cytoskeletal regulator, Diaphanous-related formin 3 (DIAPH3), or the nuclear membrane-associated proteins, lamin A/C, in prostate and breast cancer cells, induces nuclear shape instability, with a corresponding gain in malignant properties, including secretion of extracellular vesicles that contain genomic material. This transformation is characterized by a reduction and/or mislocalization of the inner nuclear membrane protein, emerin. Consistent with this, depletion of emerin evokes nuclear shape instability and promotes metastasis. By visualizing emerin localization, evidence for nuclear shape instability was observed in cultured tumor cells, in experimental models of prostate cancer, in human prostate cancer tissues, and in circulating tumor cells from patients with metastatic disease. Quantitation of emerin mislocalization discriminated cancer from benign tissue and correlated with disease progression in a prostate cancer cohort. Taken together, these results identify emerin as a mediator of nuclear shape stability in cancer and show that destabilization of emerin can promote metastasis.Significance: This study identifies a novel mechanism integrating the control of nuclear structure with the metastatic phenotype, and our inclusion of two types of human specimens (cancer tissues and circulating tumor cells) demonstrates direct relevance to human cancer.Graphical Abstract: http://cancerres.aacrjournals.org/content/canres/78/21/6086/F1.large.jpg Cancer Res; 78(21); 6086-97. ©2018 AACR.


Asunto(s)
Núcleo Celular/metabolismo , Regulación Neoplásica de la Expresión Génica , Proteínas de la Membrana/metabolismo , Metástasis de la Neoplasia , Proteínas Nucleares/metabolismo , Neoplasias de la Próstata/metabolismo , Neoplasias de la Próstata/patología , Animales , Apoptosis , Biomarcadores de Tumor , Línea Celular Tumoral , Movimiento Celular , Progresión de la Enfermedad , Humanos , Masculino , Ratones , Ratones SCID , Invasividad Neoplásica , Células Neoplásicas Circulantes , Membrana Nuclear
12.
Circ Heart Fail ; 10(2)2017 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-28209764

RESUMEN

BACKGROUND: Despite increased secondary cardiovascular events in patients with ischemic cardiomyopathy (ICM), the expression of innate cardiac protective molecules in the hearts of patients with ICM is incompletely characterized. Therefore, we used a nonbiased RNAseq approach to determine whether differences in cardiac protective molecules occur with ICM. METHODS AND RESULTS: RNAseq analysis of human control and ICM left ventricular samples demonstrated a significant decrease in KCNJ11 expression with ICM. KCNJ11 encodes the Kir6.2 subunit of the cardioprotective KATP channel. Using wild-type mice and kcnj11-deficient (kcnj11-null) mice, we examined the effect of kcnj11 expression on cardiac function during ischemia-reperfusion injury. Reactive oxygen species generation increased in kcnj11-null hearts above that found in wild-type mice hearts after ischemia-reperfusion injury. Continuous left ventricular pressure measurement during ischemia and reperfusion demonstrated a more compromised diastolic function in kcnj11-null compared with wild-type mice during reperfusion. Analysis of key calcium-regulating proteins revealed significant differences in kcnj11-null mice. Despite impaired relaxation, kcnj11-null hearts increased phospholamban Ser16 phosphorylation, a modification that results in the dissociation of phospholamban from sarcoendoplasmic reticulum Ca2+, thereby increasing sarcoendoplasmic reticulum Ca2+-mediated calcium reuptake. However, kcnj11-null mice also had increased 3-nitrotyrosine modification of the sarcoendoplasmic reticulum Ca2+-ATPase, a modification that irreversibly impairs sarcoendoplasmic reticulum Ca2+ function, thereby contributing to diastolic dysfunction. CONCLUSIONS: KCNJ11 expression is decreased in human ICM. Lack of kcnj11 expression increases peroxynitrite-mediated modification of the key calcium-handling protein sarcoendoplasmic reticulum Ca2+-ATPase after myocardial ischemia-reperfusion injury, contributing to impaired diastolic function. These data suggest a mechanism for ischemia-induced diastolic dysfunction in patients with ICM.


Asunto(s)
Cardiomiopatías/metabolismo , Infarto del Miocardio/metabolismo , Daño por Reperfusión Miocárdica/metabolismo , Miocardio/metabolismo , Estrés Oxidativo , Canales de Potasio de Rectificación Interna/deficiencia , Especies de Nitrógeno Reactivo/metabolismo , Especies Reactivas de Oxígeno/metabolismo , Adulto , Animales , Canales de Calcio Tipo L/metabolismo , Señalización del Calcio , Proteínas de Unión al Calcio/metabolismo , Cardiomiopatías/genética , Cardiomiopatías/fisiopatología , Estudios de Casos y Controles , Modelos Animales de Enfermedad , Femenino , Predisposición Genética a la Enfermedad , Humanos , Masculino , Ratones Endogámicos C57BL , Ratones Noqueados , Persona de Mediana Edad , Infarto del Miocardio/genética , Infarto del Miocardio/fisiopatología , Daño por Reperfusión Miocárdica/genética , Daño por Reperfusión Miocárdica/fisiopatología , Fenotipo , Canales de Potasio de Rectificación Interna/genética , ATPasas Transportadoras de Calcio del Retículo Sarcoplásmico/metabolismo , Tirosina/análogos & derivados , Tirosina/metabolismo , Disfunción Ventricular Izquierda/metabolismo , Disfunción Ventricular Izquierda/fisiopatología , Función Ventricular Izquierda , Presión Ventricular
13.
Life Sci ; 179: 54-59, 2017 Jun 15.
Artículo en Inglés | MEDLINE | ID: mdl-27756600

RESUMEN

AIMS: Prior work suggests that ischemic preconditioning increases the level of CD39 in the heart and contributes to cardiac protection. Therefore, we examined if targeted cardiac expression of CD39 protects against myocardial injury. MAIN METHODS: Mice with cardiac-specific expression of human CD39 (αMHC/hCD39-Tg) were generated, characterized and subjected to left coronary artery ischemia-reperfusion injury and infarct size at 24h following injury quantified. KEY FINDINGS: αMHC/hCD39-Tg mice have increased in cardiac ATPase and ADPase activity compared to WT littermates. The increased activity in αMHC/hCD39-mice was inhibited by the CD39 antagonist sodium polyoxotungstate (POM-1). Measurement of basal cardiac function by echocardiography revealed that αMHC/hCD39-Tg mice have a lower resting heart rate and increased stroke volume. In response to myocardial ischemia, systolic and diastolic function was better preserved in αMHC/hCD39-Tg compared to WT mice. Comparison of Tau also revealed preserved cardiac relaxation during ischemia in αMHC/hCD39-Tg hearts. Assessment of myocardial infarct size in response to 60min of ischemia and 24h of reperfusion demonstrated a significant reduction in infarct size in αMHC/hCD39-Tg hearts. Analysis of isolated cardiomyocytes revealed no basal difference in calcium transients between WT and αMHC/hCD39-Tg cardiomyocytes. However, in response to isoproterenol stimulation, there was a trend toward lower calcium transients in αMHC/hCD39 cardiomyocytes suggesting less calcium accumulation in response to metabolic stress. SIGNIFICANCE: Cardiac-specific expression of CD39 reduces myocardial dysfunction and infarct size following ischemia-reperfusion injury. Increasing nucleotidase expression in the heart may be a novel approach to protect the heart from ischemic injury.


Asunto(s)
Antígenos CD/genética , Apirasa/genética , Expresión Génica , Infarto del Miocardio/patología , Daño por Reperfusión Miocárdica/complicaciones , Animales , Calcio/metabolismo , Modelos Animales de Enfermedad , Ecocardiografía , Frecuencia Cardíaca/genética , Humanos , Isoproterenol/farmacología , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Infarto del Miocardio/genética , Daño por Reperfusión Miocárdica/genética , Miocitos Cardíacos/metabolismo , Estrés Fisiológico/genética , Volumen Sistólico/genética
15.
Arterioscler Thromb Vasc Biol ; 36(9): 1809-20, 2016 09.
Artículo en Inglés | MEDLINE | ID: mdl-27417582

RESUMEN

OBJECTIVE: Circulating blood cells and endothelial cells express ectonucleoside triphosphate diphosphohydrolase-1 (CD39) and ecto-5'-nucleotidase (CD73). CD39 hydrolyzes extracellular ATP or ADP to AMP. CD73 hydrolyzes AMP to adenosine. The goal of this study was to examine the interplay between CD39 and CD73 cascade in arterial thrombosis. APPROACH AND RESULTS: To determine how CD73 activity influences in vivo thrombosis, the time to ferric chloride-induced arterial thrombosis was measured in CD73-null mice. In response to 5% FeCl3, but not to 10% FeCl3, there was a significant decrease in the time to thrombosis in CD73-null mice compared with wild-type mice. In mice overexpressing CD39, ablation of CD73 did not inhibit the prolongation in the time to thrombosis conveyed by CD39 overexpression. However, the CD73 inhibitor α-ß-methylene-ADP nullified the prolongation in the time to thrombosis in human CD39 transgenic (hC39-Tg)/CD73-null mice. To determine whether hematopoietic-derived cells or endothelial cell CD39 activity regulates in vivo arterial thrombus, bone marrow transplant studies were conducted. FeCl3-induced arterial thrombosis in chimeric mice revealed a significant prolongation in the time to thrombosis in hCD39-Tg reconstituted wild-type mice, but not on wild-type reconstituted hCD39-Tg mice. Monocyte depletion with clodronate-loaded liposomes normalized the time to thrombosis in hCD39-Tg mice compared with hCD39-Tg mice treated with control liposomes, demonstrating that increased CD39 expression on monocytes protects against thrombosis. CONCLUSIONS: These data demonstrate that ablation of CD73 minimally effects in vivo thrombosis, but increased CD39 expression on hematopoietic-derived cells, especially monocytes, attenuates in vivo arterial thrombosis.


Asunto(s)
5'-Nucleotidasa/metabolismo , Antígenos CD/metabolismo , Apirasa/metabolismo , Arteriopatías Oclusivas/enzimología , Coagulación Sanguínea , Trombosis/enzimología , 5'-Nucleotidasa/deficiencia , 5'-Nucleotidasa/genética , Adenosina/metabolismo , Adenosina Difosfato/metabolismo , Adenosina Monofosfato/metabolismo , Adenosina Trifosfato/metabolismo , Animales , Antígenos CD/genética , Apirasa/genética , Arteriopatías Oclusivas/sangre , Arteriopatías Oclusivas/inducido químicamente , Arteriopatías Oclusivas/genética , Trasplante de Médula Ósea , Cloruros , Modelos Animales de Enfermedad , Células Endoteliales/enzimología , Compuestos Férricos , Predisposición Genética a la Enfermedad , Células HEK293 , Humanos , Hidrólisis , Ratones Endogámicos C57BL , Ratones Noqueados , Ratones Transgénicos , Monocitos/enzimología , Fenotipo , Activación Plaquetaria , Trombosis/sangre , Trombosis/inducido químicamente , Trombosis/genética , Factores de Tiempo , Transfección
16.
J Mol Cell Cardiol ; 93: 47-56, 2016 04.
Artículo en Inglés | MEDLINE | ID: mdl-26891859

RESUMEN

Following myocardial infarction, purinergic nucleotides and nucleosides are released via non-specific and specific mechanisms in response to cellular activation, stress, or injury. These extracellular nucleotides are potent mediators of physiologic and pathologic responses, contributing to the inflammatory and fibrotic milieu within the injured myocardium. Via autocrine or paracrine signaling, cell-specific effects occur through differentially expressed purinergic receptors of the P2X, P2Y, and P1 families. Nucleotide activation of the ionotropic (ligand-gated) purine receptors (P2X) and several of the metabotropic (G-protein-coupled) purine receptors (P2Y) or adenosine activation of the P1 receptors can have profound effects on inflammatory cell function, fibroblast function, and cardiomyocyte function. Extracellular nucleotidases that hydrolyze released nucleotides regulate the magnitude and duration of purinergic signaling. While there are numerous studies on the role of the purinergic signaling pathway in cardiovascular disease, the extent to which the purinergic signaling pathway modulates cardiac fibrosis is incompletely understood. Here we provide an overview of the current understanding of how the purinergic signaling pathway modulates cardiac fibroblast function and myocardial fibrosis.


Asunto(s)
Miocardio/metabolismo , Miocardio/patología , Nucleótidos/metabolismo , Transducción de Señal , Adenosina/metabolismo , Animales , Espacio Extracelular/metabolismo , Fibrosis , Humanos , Hidrólisis , Receptores Purinérgicos/metabolismo
17.
Am J Physiol Renal Physiol ; 310(8): F705-F716, 2016 04 15.
Artículo en Inglés | MEDLINE | ID: mdl-26661656

RESUMEN

No therapies have been shown to accelerate recovery or prevent fibrosis after acute kidney injury (AKI). In part, this is because most therapeutic candidates have to be given at the time of injury and the diagnosis of AKI is usually made too late for drugs to be efficacious. Strategies to enhance post-AKI repair represent an attractive approach to address this. Using a phenotypic screen in zebrafish, we identified 4-(phenylthio)butanoic acid (PTBA), which promotes proliferation of embryonic kidney progenitor cells (EKPCs), and the PTBA methyl ester UPHD25, which also increases postinjury repair in ischemia-reperfusion and aristolochic acid-induced AKI in mice. In these studies, a new panel of PTBA analogs was evaluated. Initial screening was performed in zebrafish EKPC assays followed by survival assays in a gentamicin-induced AKI larvae zebrafish model. Using this approach, we identified UPHD186, which in contrast to UPHD25, accelerates recovery and reduces fibrosis when administered several days after ischemia-reperfusion AKI and reduces fibrosis after unilateral ureteric obstruction in mice. UPHD25 and 186 are efficiently metabolized to the active analog PTBA in liver and kidney microsome assays, indicating both compounds may act as PTBA prodrugs in vivo. UPHD186 persists longer in the circulation than UPHD25, suggesting that sustained levels of UPHD186 may increase efficacy by acting as a reservoir for renal metabolism to PTBA. These findings validate use of zebrafish EKPC and AKI assays as a drug discovery strategy for molecules that reduce fibrosis in multiple AKI models and can be administered days after initiation of injury.


Asunto(s)
Lesión Renal Aguda/tratamiento farmacológico , Butiratos/uso terapéutico , Riñón/efectos de los fármacos , Sulfuros/uso terapéutico , Lesión Renal Aguda/patología , Animales , Butiratos/farmacología , Proliferación Celular/efectos de los fármacos , Supervivencia Celular/efectos de los fármacos , Modelos Animales de Enfermedad , Fibrosis/tratamiento farmacológico , Fibrosis/patología , Riñón/patología , Masculino , Ratones , Sulfuros/farmacología , Pez Cebra
18.
Am J Physiol Renal Physiol ; 306(5): F496-504, 2014 Mar 01.
Artículo en Inglés | MEDLINE | ID: mdl-24370591

RESUMEN

Phenylthiobutanoic acids (PTBAs) are a new class of histone deacetylase (HDAC) inhibitors that accelerate recovery and reduce postinjury fibrosis after ischemia-reperfusion-induced acute kidney injury. However, unlike the more common scenario in which patients present with protracted and less clearly defined onset of renal injury, this model of acute kidney injury gives rise to a clearly defined injury that begins to resolve over a short period of time. In these studies, we show for the first time that treatment with the PTBA analog methyl-4-(phenylthio)butanoate (M4PTB) accelerates recovery and reduces postinjury fibrosis in a progressive model of acute kidney injury and renal fibrosis that occurs after aristolochic acid injection in mice. These effects are apparent when M4PTB treatment is delayed 4 days after the initiating injury and are associated with increased proliferation and decreased G2/M arrest of regenerating renal tubular epithelial cells. In addition, there is reduced peritubular macrophage infiltration and decreased expression of the macrophage chemokines CX3Cl1 and CCL2. Since macrophage infiltration plays a role in promoting kidney injury, and since renal tubular epithelial cells show defective repair and a marked increase in maladaptive G2/M arrest after aristolochic acid injury, these findings suggest M4PTB may be particularly beneficial in reducing injury and enhancing intrinsic cellular repair even when administered days after aristolochic acid ingestion.


Asunto(s)
Lesión Renal Aguda/tratamiento farmacológico , Butiratos/farmacología , Sulfuros/farmacología , Lesión Renal Aguda/inducido químicamente , Animales , Ácidos Aristolóquicos/farmacología , Butiratos/análisis , Modelos Animales de Enfermedad , Fibrosis/tratamiento farmacológico , Fibrosis/prevención & control , Inhibidores de Histona Desacetilasas/farmacología , Túbulos Renales/efectos de los fármacos , Túbulos Renales/metabolismo , Ratones , Ratones Biozzi , Daño por Reperfusión/inducido químicamente , Daño por Reperfusión/tratamiento farmacológico , Daño por Reperfusión/metabolismo , Sulfuros/análisis
19.
J Am Soc Nephrol ; 24(6): 943-53, 2013 May.
Artículo en Inglés | MEDLINE | ID: mdl-23620402

RESUMEN

At present, there are no effective therapies to ameliorate injury, accelerate recovery, or prevent postinjury fibrosis after AKI. Here, we sought to identify candidate compounds that accelerate recovery after AKI by screening for small molecules that increase proliferation of renal progenitor cells in zebrafish embryos. One compound identified from this screen was the histone deacetylase inhibitor methyl-4-(phenylthio)butanoate, which we subsequently administered to zebrafish larvae and mice 24-48 hours after inducing AKI. In zebrafish, treatment with the compound increased larval survival and proliferation of renal tubular epithelial cells. In mice, treatment accelerated recovery, reduced postinjury tubular atrophy and interstitial fibrosis, and increased the regenerative capacity of actively cycling renal tubular cells by decreasing the number of cells in G2/M arrest. These data suggest that accelerating recovery may be a viable approach to treating AKI and provide proof of concept that a screen in zebrafish embryos can identify therapeutic candidates for kidney injury.


Asunto(s)
Lesión Renal Aguda/tratamiento farmacológico , Lesión Renal Aguda/enzimología , Histona Desacetilasa 1/antagonistas & inhibidores , Inhibidores de Histona Desacetilasas/farmacología , Fenilbutiratos/farmacología , Proteínas de Pez Cebra/antagonistas & inhibidores , Lesión Renal Aguda/patología , Animales , Modelos Animales de Enfermedad , Fibrosis , Gentamicinas/toxicidad , Histona Desacetilasa 1/metabolismo , Isquemia/tratamiento farmacológico , Isquemia/enzimología , Isquemia/patología , Riñón/efectos de los fármacos , Riñón/enzimología , Riñón/patología , Masculino , Ratones , Ratones Endogámicos BALB C , Inhibidores de la Síntesis de la Proteína/toxicidad , Recuperación de la Función/efectos de los fármacos , Pez Cebra , Proteínas de Pez Cebra/metabolismo
20.
PLoS One ; 8(2): e56062, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-23409123

RESUMEN

PURPOSE: Pax3cre-mediated deletion of fibroblast growth factor receptor 2 (Fgfr2) broadly in renal and urinary tract mesenchyme led to ureteric bud (UB) induction defects and vesicoureteral reflux (VUR), although the mechanisms were unclear. Here, we investigated whether Fgfr2 acts specifically in peri-Wolffian duct stroma (ST) to regulate UB induction and development of VUR and the mechanisms of Fgfr2 activity. METHODS: We conditionally deleted Fgfr2 in ST (Fgfr2(ST-/-)) using Tbx18cre mice. To look for ureteric bud induction defects in young embryos, we assessed length and apoptosis of common nephric ducts (CNDs). We performed 3D reconstructions and histological analyses of urinary tracts of embryos and postnatal mice and cystograms in postnatal mice to test for VUR. We performed in situ hybridization and real-time PCR in young embryos to determine mechanisms underlying UB induction defects. RESULTS: We confirmed that Fgfr2 is expressed in ST and that Fgfr2 was efficiently deleted in this tissue in Fgfr2(ST-/-) mice at embryonic day (E) 10.5. E11.5 Fgfr2(ST-/-) mice had randomized UB induction sites with approximately 1/3 arising too high and 1/3 too low from the Wolffian duct; however, apoptosis was unaltered in E12.5 mutant CNDs. While ureters were histologically normal, E15.5 Fgfr2(ST-/-) mice exhibit improper ureteral insertion sites into the bladder, consistent with the ureteric induction defects. While ureter and bladder histology appeared normal, postnatal day (P) 1 mutants had high rates of VUR versus controls (75% versus 3%, p = 0.001) and occasionally other defects including renal hypoplasia and duplex systems. P1 mutant mice also had improper ureteral bladder insertion sites and shortened intravesicular tunnel lengths that correlated with VUR. E10.5 Fgfr2(ST-/-) mice had decreases in Bmp4 mRNA in stromal tissues, suggesting a mechanism underlying the ureteric induction and VUR phenotypes. CONCLUSION: Mutations in FGFR2 could possibly cause VUR in humans.


Asunto(s)
Eliminación de Gen , Receptor Tipo 2 de Factor de Crecimiento de Fibroblastos/genética , Células del Estroma/metabolismo , Uréter/anomalías , Uréter/embriología , Reflujo Vesicoureteral/genética , Conductos Mesonéfricos/metabolismo , Animales , Proteína Morfogenética Ósea 4/genética , Proteína Morfogenética Ósea 4/metabolismo , Regulación del Desarrollo de la Expresión Génica , Ratones , Ratones Noqueados , Proteínas de Dominio T Box/genética , Proteínas de Dominio T Box/metabolismo , Vejiga Urinaria/anomalías , Vejiga Urinaria/embriología , Anomalías Urogenitales/embriología , Anomalías Urogenitales/genética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...