Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 22
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
STAR Protoc ; 5(1): 102754, 2024 Mar 15.
Artículo en Inglés | MEDLINE | ID: mdl-38096060

RESUMEN

Characterization of isolated extracellular vesicles and particles (EVPs) is crucial for determining functions and biomarker potential. Here, we present a protocol to analyze size, number, morphology, and EVP protein cargo and to validate EVP proteins in both humans and mice. We describe steps for nanoparticle tracking analysis, transmission electron microscopy, single-EVP immunodetection, EVP proteomic mass spectrometry and bioinformatic analysis, and EVP protein validation by ExoELISA and western blot analysis. This allows for EVP cross-validation across different platforms. For complete details on the use and execution of this protocol, please refer to Hoshino et al.1.


Asunto(s)
Vesículas Extracelulares , Proteómica , Humanos , Animales , Ratones , Western Blotting , Biología Computacional , Espectrometría de Masas
2.
EMBO Rep ; 24(12): e57339, 2023 Dec 06.
Artículo en Inglés | MEDLINE | ID: mdl-37929643

RESUMEN

Breast adipose tissue is an important contributor to the obesity-breast cancer link. Extracellular vesicles (EVs) are nanosized particles containing selective cargo, such as miRNAs, that act locally or circulate to distant sites to modulate target cell functions. Here, we find that long-term education of breast cancer cells with EVs obtained from breast adipose tissue of women who are overweight or obese (O-EVs) results in increased proliferation. RNA-seq analysis of O-EV-educated cells demonstrates increased expression of genes involved in oxidative phosphorylation, such as ATP synthase and NADH: ubiquinone oxidoreductase. O-EVs increase respiratory complex protein expression, mitochondrial density, and mitochondrial respiration in tumor cells. The mitochondrial complex I inhibitor metformin reverses O-EV-induced cell proliferation. Several miRNAs-miR-155-5p, miR-10a-3p, and miR-30a-3p-which promote mitochondrial respiration and proliferation, are enriched in O-EVs relative to EVs from lean women. O-EV-induced proliferation and mitochondrial activity are associated with stimulation of the Akt/mTOR/P70S6K pathway, and are reversed upon silencing of P70S6K. This study reveals a new facet of the obesity-breast cancer link with human breast adipose tissue-derived EVs causing metabolic reprogramming of breast cancer cells.


Asunto(s)
Neoplasias de la Mama , Vesículas Extracelulares , MicroARNs , Humanos , Femenino , Proteínas Quinasas S6 Ribosómicas 70-kDa/metabolismo , Tejido Adiposo/metabolismo , MicroARNs/genética , MicroARNs/metabolismo , Obesidad/metabolismo , Neoplasias de la Mama/metabolismo , Proteínas/metabolismo , Vesículas Extracelulares/metabolismo
3.
Sci Immunol ; 8(86): eadg0878, 2023 08 04.
Artículo en Inglés | MEDLINE | ID: mdl-37624910

RESUMEN

During persistent antigen stimulation, such as in chronic infections and cancer, CD8 T cells differentiate into a hypofunctional programmed death protein 1-positive (PD-1+) exhausted state. Exhausted CD8 T cell responses are maintained by precursors (Tpex) that express the transcription factor T cell factor 1 (TCF-1) and high levels of the costimulatory molecule CD28. Here, we demonstrate that sustained CD28 costimulation is required for maintenance of antiviral T cells during chronic infection. Low-level CD28 engagement preserved mitochondrial fitness and self-renewal of Tpex, whereas stronger CD28 signaling enhanced glycolysis and promoted Tpex differentiation into TCF-1neg exhausted CD8 T cells (Tex). Furthermore, enhanced differentiation by CD28 engagement did not reduce the Tpex pool. Together, these findings demonstrate that continuous CD28 engagement is needed to sustain PD-1+ CD8 T cells and suggest that increasing CD28 signaling promotes Tpex differentiation into more functional effector-like Tex, possibly without compromising long-term responses.


Asunto(s)
Antígenos CD28 , Factor 1 de Transcripción de Linfocitos T , Factor 1 de Transcripción de Linfocitos T/genética , Receptor de Muerte Celular Programada 1 , Linfocitos T CD8-positivos , Diferenciación Celular , Factores de Transcripción
4.
bioRxiv ; 2023 Feb 09.
Artículo en Inglés | MEDLINE | ID: mdl-36798307

RESUMEN

Breast adipose tissue is an important contributor to the obesity-breast cancer link. Dysregulated cell metabolism is now an accepted hallmark of cancer. Extracellular vesicles (EVs) are nanosized particles containing selective cargo, such as miRNAs, that act locally or circulate to distant sites to modulate target cell functions. Here, we found that long-term education of breast cancer cells (MCF7, T47D) with EVs from breast adipose tissue of women who are overweight or obese (O-EVs) leads to sustained increased proliferative potential. RNA-Seq of O-EV-educated cells demonstrates increased expression of genes, such as ATP synthase and NADH: ubiquinone oxidoreductase, involved in oxidative phosphorylation. O-EVs increase respiratory complex protein expression, mitochondrial density, and mitochondrial respiration in tumor cells. Mitochondrial complex I inhibitor, metformin, reverses O-EV-induced cell proliferation. Several miRNAs, miR-155-5p, miR-10a-3p, and miR-30a-3p, which promote mitochondrial respiration and proliferation, are enriched in O-EVs relative to EVs from lean women. O-EV-induced proliferation and mitochondrial activity are associated with stimulation of the Akt/mTOR/P70S6K pathway, and are reversed upon silencing of P70S6K. This study reveals a new facet of the obesity-breast cancer link with human breast adipose tissue-derived EVs causing the metabolic reprogramming of ER+ breast cancer cells.

6.
Cell Rep ; 26(12): 3203-3211.e5, 2019 03 19.
Artículo en Inglés | MEDLINE | ID: mdl-30893594

RESUMEN

The prognosis of patients with glioblastoma (GBM) remains dismal, with a median survival of approximately 15 months. Current preclinical GBM models are limited by the lack of a "normal" human microenvironment and the inability of many tumor cell lines to accurately reproduce GBM biology. To address these limitations, we have established a model system whereby we can retro-engineer patient-specific GBMs using patient-derived glioma stem cells (GSCs) and human embryonic stem cell (hESC)-derived cerebral organoids. Our cerebral organoid glioma (GLICO) model shows that GSCs home toward the human cerebral organoid and deeply invade and proliferate within the host tissue, forming tumors that closely phenocopy patient GBMs. Furthermore, cerebral organoid tumors form rapidly and are supported by an interconnected network of tumor microtubes that aids in the invasion of normal host tissue. Our GLICO model provides a system for modeling primary human GBM ex vivo and for high-throughput drug screening.


Asunto(s)
Neoplasias Encefálicas/metabolismo , Glioblastoma/metabolismo , Modelos Biológicos , Células Madre Neoplásicas/metabolismo , Organoides/metabolismo , Neoplasias Encefálicas/patología , Glioblastoma/patología , Humanos , Invasividad Neoplásica , Células Madre Neoplásicas/patología , Organoides/patología
7.
Nat Commun ; 9(1): 3533, 2018 08 30.
Artículo en Inglés | MEDLINE | ID: mdl-30166553

RESUMEN

Granular cell tumors (GCTs) are rare tumors that can arise in multiple anatomical locations, and are characterized by abundant intracytoplasmic granules. The genetic drivers of GCTs are currently unknown. Here, we apply whole-exome sequencing and targeted sequencing analysis to reveal mutually exclusive, clonal, inactivating somatic mutations in the endosomal pH regulators ATP6AP1 or ATP6AP2 in 72% of GCTs. Silencing of these genes in vitro results in impaired vesicle acidification, redistribution of endosomal compartments, and accumulation of intracytoplasmic granules, recapitulating the cardinal phenotypic characteristics of GCTs and providing a novel genotypic-phenotypic correlation. In addition, depletion of ATP6AP1 or ATP6AP2 results in the acquisition of oncogenic properties. Our results demonstrate that inactivating mutations of ATP6AP1 and ATP6AP2 are likely oncogenic drivers of GCTs and underpin the genesis of the intracytoplasmic granules that characterize them, providing a genetic link between endosomal pH regulation and tumorigenesis.


Asunto(s)
Tumor de Células Granulares/genética , Mutación/genética , Receptores de Superficie Celular/genética , ATPasas de Translocación de Protón Vacuolares/genética , Proliferación Celular/genética , Proliferación Celular/fisiología , Exoma , Femenino , Citometría de Flujo , Estudios de Asociación Genética , Células HEK293 , Humanos , Masculino
8.
Nat Cell Biol ; 20(3): 332-343, 2018 03.
Artículo en Inglés | MEDLINE | ID: mdl-29459780

RESUMEN

The heterogeneity of exosomal populations has hindered our understanding of their biogenesis, molecular composition, biodistribution and functions. By employing asymmetric flow field-flow fractionation (AF4), we identified two exosome subpopulations (large exosome vesicles, Exo-L, 90-120 nm; small exosome vesicles, Exo-S, 60-80 nm) and discovered an abundant population of non-membranous nanoparticles termed 'exomeres' (~35 nm). Exomere proteomic profiling revealed an enrichment in metabolic enzymes and hypoxia, microtubule and coagulation proteins as well as specific pathways, such as glycolysis and mTOR signalling. Exo-S and Exo-L contained proteins involved in endosomal function and secretion pathways, and mitotic spindle and IL-2/STAT5 signalling pathways, respectively. Exo-S, Exo-L and exomeres each had unique N-glycosylation, protein, lipid, DNA and RNA profiles and biophysical properties. These three nanoparticle subsets demonstrated diverse organ biodistribution patterns, suggesting distinct biological functions. This study demonstrates that AF4 can serve as an improved analytical tool for isolating extracellular vesicles and addressing the complexities of heterogeneous nanoparticle subpopulations.


Asunto(s)
Fraccionamiento Celular/métodos , Exosomas/metabolismo , Nanopartículas , Neoplasias/metabolismo , Proteínas/metabolismo , Animales , Biomarcadores/metabolismo , ADN/genética , ADN/metabolismo , Metabolismo Energético , Exosomas/clasificación , Exosomas/genética , Exosomas/patología , Femenino , Glicómica , Glicosilación , Células HCT116 , Humanos , Melanoma Experimental/genética , Melanoma Experimental/metabolismo , Melanoma Experimental/patología , Metabolómica , Ratones , Ratones Endogámicos C57BL , Células 3T3 NIH , Neoplasias/genética , Neoplasias/patología , Células PC-3 , Fenotipo , Proteómica , ARN/genética , ARN/metabolismo , Transducción de Señal , Distribución Tisular
9.
Dev Biol ; 435(1): 41-55, 2018 03 01.
Artículo en Inglés | MEDLINE | ID: mdl-29331499

RESUMEN

Small heat shock proteins are chaperones with variable mechanisms of action. The function of cardiac family member Hspb7 is unknown, despite being identified through GWAS as a potential cardiomyopathy risk gene. We discovered that zebrafish hspb7 mutants display mild focal cardiac fibrosis and sarcomeric abnormalities. Significant mortality was observed in adult hspb7 mutants subjected to exercise stress, demonstrating a genetic and environmental interaction that determines disease outcome. We identified large sarcomeric proteins FilaminC and Titin as Hspb7 binding partners in cardiac cells. Damaged FilaminC undergoes autophagic processing to maintain sarcomeric homeostasis. Loss of Hspb7 in zebrafish or human cardiomyocytes stimulated autophagic pathways and expression of the sister gene encoding Hspb5. Inhibiting autophagy caused FilaminC aggregation in HSPB7 mutant human cardiomyocytes and developmental cardiomyopathy in hspb7 mutant zebrafish embryos. These studies highlight the importance of damage-processing networks in cardiomyocytes, and a previously unrecognized role in this context for Hspb7.


Asunto(s)
Cardiomiopatías/embriología , Proteínas de Choque Térmico HSP27/metabolismo , Proteostasis , Sarcómeros/metabolismo , Proteínas de Pez Cebra/metabolismo , Pez Cebra/embriología , Animales , Autofagia/genética , Cardiomiopatías/genética , Cardiomiopatías/patología , Filaminas/genética , Filaminas/metabolismo , Proteínas de Choque Térmico HSP27/genética , Humanos , Miocitos Cardíacos/metabolismo , Miocitos Cardíacos/patología , Sarcómeros/genética , Pez Cebra/genética , Proteínas de Pez Cebra/genética
10.
J Am Soc Nephrol ; 28(5): 1437-1449, 2017 May.
Artículo en Inglés | MEDLINE | ID: mdl-27881606

RESUMEN

The innate immune system has been implicated in both AKI and CKD. Damaged mitochondria release danger molecules, such as reactive oxygen species, DNA, and cardiolipin, which can cause NLRP3 inflammasome activation and upregulation of IL-18 and IL-1ß It is not known if mitochondrial damage persists long after ischemia to sustain chronic inflammasome activation. We conducted a 9-month study in Sprague-Dawley rats after 45 minutes of bilateral renal ischemia. We detected glomerular and peritubular capillary rarefaction, macrophage infiltration, and fibrosis at 1 month. Transmission electron microscopy revealed mitochondrial degeneration, mitophagy, and deformed foot processes in podocytes. These changes progressed over the study period, with a persistent increase in renal cortical expression of IL-18, IL-1ß, and TGF-ß, despite a gradual decline in TNF-α expression and macrophage infiltration. Treatment with a mitoprotective agent (SS-31; elamipretide) for 6 weeks, starting 1 month after ischemia, preserved mitochondrial integrity, ameliorated expression levels of all inflammatory markers, restored glomerular capillaries and podocyte structure, and arrested glomerulosclerosis and interstitial fibrosis. Further, helium ion microscopy vividly demonstrated the restoration of podocyte structure by SS-31. The protection by SS-31 was sustained for ≥6 months after treatment ended, with normalization of IL-18 and IL-1ß expression. These results support a role for mitochondrial damage in inflammasome activation and CKD and suggest mitochondrial protection as a novel therapeutic approach that can arrest the progression of CKD. Notably, SS-31 is effective when given long after AKI and provides persistent protection after termination of drug treatment.


Asunto(s)
Interleucina-18/fisiología , Interleucina-1beta/fisiología , Isquemia/complicaciones , Riñón/irrigación sanguínea , Mitocondrias/efectos de los fármacos , Mitocondrias/fisiología , Oligopéptidos/farmacología , Oligopéptidos/uso terapéutico , Insuficiencia Renal Crónica/etiología , Insuficiencia Renal Crónica/prevención & control , Regulación hacia Arriba/efectos de los fármacos , Enfermedad Aguda , Animales , Masculino , Podocitos/efectos de los fármacos , Ratas , Ratas Sprague-Dawley , Factores de Tiempo
11.
Nat Biotechnol ; 34(8): 845-51, 2016 08.
Artículo en Inglés | MEDLINE | ID: mdl-27398792

RESUMEN

Refined cancer models are needed to bridge the gaps between cell line, animal and clinical research. Here we describe the engineering of an organotypic colon cancer model by recellularization of a native human matrix that contains cell-populated mucosa and an intact muscularis mucosa layer. This ex vivo system recapitulates the pathophysiological progression from APC-mutant neoplasia to submucosal invasive tumor. We used it to perform a Sleeping Beauty transposon mutagenesis screen to identify genes that cooperate with mutant APC in driving invasive neoplasia. We identified 38 candidate invasion-driver genes, 17 of which, including TCF7L2, TWIST2, MSH2, DCC, EPHB1 and EPHB2 have been previously implicated in colorectal cancer progression. Six invasion-driver genes that have not, to our knowledge, been previously described were validated in vitro using cell proliferation, migration and invasion assays and ex vivo using recellularized human colon. These results demonstrate the utility of our organoid model for studying cancer biology.


Asunto(s)
Proteína de la Poliposis Adenomatosa del Colon/genética , Colon/metabolismo , Neoplasias del Colon/metabolismo , Neoplasias del Colon/patología , Perfilación de la Expresión Génica/métodos , Proteínas de Neoplasias/metabolismo , Carcinogénesis/genética , Sistema Libre de Células/metabolismo , Células Cultivadas , Colon/patología , Genes Relacionados con las Neoplasias/genética , Humanos , Organogénesis , Ingeniería de Tejidos/métodos
12.
J Exp Med ; 209(12): 2291-305, 2012 Nov 19.
Artículo en Inglés | MEDLINE | ID: mdl-23091165

RESUMEN

Treatment of acute cardiac ischemia focuses on reestablishment of blood flow in coronary arteries. However, impaired microvascular perfusion damages peri-infarct tissue, despite arterial patency. Identification of cytokines that induce microvascular dysfunction would provide new targets to limit microvascular damage. Pro-nerve growth factor (NGF), the precursor of NGF, is a well characterized cytokine in the brain induced by injury. ProNGF activates p75 neurotrophin receptor (p75(NTR)) and sortilin receptors to mediate proapoptotic responses. We describe induction of proNGF by cardiomyocytes, and p75(NTR) in human arterioles after fatal myocardial infarction, but not with unrelated pathologies. After mouse cardiac ischemia-reperfusion (I-R) injury, rapid up-regulation of proNGF by cardiomyocytes and p75(NTR) by microvascular pericytes is observed. To identify proNGF actions, we generated a mouse expressing a mutant Ngf allele with impaired processing of proNGF to mature NGF. The proNGF-expressing mouse exhibits cardiac microvascular endothelial activation, a decrease in pericyte process length, and increased vascular permeability, leading to lethal cardiomyopathy in adulthood. Deletion of p75(NTR) in proNGF-expressing mice rescues the phenotype, confirming the importance of p75(NTR)-expressing pericytes in the development of microvascular injury. Furthermore, deficiency in p75(NTR) limits infarct size after I-R. These studies identify novel, nonneuronal actions for proNGF and suggest that proNGF represents a new target to limit microvascular dysfunction.


Asunto(s)
Encéfalo/metabolismo , Microvasos/patología , Infarto del Miocardio/metabolismo , Factor de Crecimiento Nervioso/metabolismo , Pericitos/metabolismo , Precursores de Proteínas/metabolismo , Daño por Reperfusión/metabolismo , Animales , Western Blotting , Cartilla de ADN/genética , Ecocardiografía , Ensayo de Inmunoadsorción Enzimática , Técnicas de Sustitución del Gen , Humanos , Inmunohistoquímica , Ratones , Microscopía Electrónica , Microscopía Fluorescente , Microvasos/metabolismo , Mutagénesis Sitio-Dirigida , Infarto del Miocardio/patología , Proteínas del Tejido Nervioso/deficiencia , Proteínas del Tejido Nervioso/metabolismo , Receptores de Factor de Crecimiento Nervioso/deficiencia , Receptores de Factor de Crecimiento Nervioso/metabolismo , Daño por Reperfusión/patología
13.
Nat Immunol ; 10(9): 1008-17, 2009 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-19648924

RESUMEN

Contact-dependent communication between immune cells generates protection but also facilitates viral spread. Here we found that macrophages formed long-range actin-propelled conduits in response to negative factor (Nef), a human immunodeficiency virus type 1 (HIV-1) protein with immunosuppressive functions. Conduits attenuated immunoglobulin G2 (IgG2) and IgA class switching in systemic and intestinal lymphoid follicles by shuttling Nef from infected macrophages to B cells through a guanine-exchange factor-dependent pathway involving the amino-terminal anchor, central core and carboxy-terminal flexible loop of Nef. By showing stronger virus-specific IgG2 and IgA responses in patients with Nef-deficient virions, our data suggest that HIV-1 exploits intercellular 'highways' as a 'Trojan horse' to deliver Nef to B cells and evade humoral immunity systemically and at mucosal sites of entry.


Asunto(s)
Linfocitos B/metabolismo , Comunicación Celular , Anticuerpos Anti-VIH/inmunología , VIH-1/inmunología , Inmunoglobulina A/inmunología , Inmunoglobulina G/inmunología , Productos del Gen nef del Virus de la Inmunodeficiencia Humana/fisiología , Actinas/metabolismo , Antígenos CD40/fisiología , Centro Germinal/fisiología , Proteína p24 del Núcleo del VIH/fisiología , Humanos , Cambio de Clase de Inmunoglobulina , Macrófagos/virología , Células U937
14.
J Am Soc Nephrol ; 19(12): 2321-30, 2008 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-18715993

RESUMEN

Recruitment of various stem and progenitor cells is crucial for the regeneration of an injured organ. Levels of uric acid, one of the prototypical "alarm signals," surge after ischemia-reperfusion injury. Exogenous uric acid rapidly mobilizes endothelial progenitor cells and hematopoietic stem cells and protects the kidney from ischemia. The relatively fast responses to uric acid suggest that preformed second messengers may be released from a storage pool. Here, it is reported that monosodium urate (MSU) results in exocytosis of Weibel-Palade bodies in vitro and in vivo, leading to the release of IL-8, von Willebrand factor, and angiopoietin 2 in the culture medium or circulation. Confocal and immunoelectron microscopy confirmed depletion of von Willebrand factor in MSU-treated aortic endothelial cells. Angiopoietin 2 alone induced exocytosis of Weibel-Palade bodies, mobilized hematopoietic stem cells and depleted splenic endothelial progenitor cells, partially reproducing the actions of MSU. In addition, pretreatment with angiopoietin 2 protected the kidneys from an ischemic insult, suggesting that the previously reported renoprotection conferred by MSU likely results from exocytosis of Weibel-Palade bodies. Furthermore, experiments with toll-like receptor 4 (TLR-4)-and TLR-2-deficient mice demonstrated that uric acid-induced exocytosis of Weibel-Palade bodies is mediated by TLR-4 and that uric acid-induced release of IL-8 requires both TLR-2 and TLR-4. In summary, these results suggest that exocytosis of Weibel-Palade bodies links postischemic repair with inflammation and mobilization of stem cells.


Asunto(s)
Exocitosis/fisiología , Isquemia , Células Madre/citología , Cuerpos de Weibel-Palade/metabolismo , Angiopoyetina 2/metabolismo , Células Cultivadas , Células Madre Hematopoyéticas/citología , Humanos , Interleucina-8/metabolismo , Daño por Reperfusión , Receptor Toll-Like 2/metabolismo , Receptor Toll-Like 4/metabolismo , Ácido Úrico/metabolismo , Factor de von Willebrand/metabolismo
15.
Development ; 135(15): 2627-36, 2008 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-18599511

RESUMEN

The mechanisms that mediate the establishment of totipotency during the egg-to-embryo transition in mammals remain poorly understood. However, it is clear that unique factors stored in the oocyte cytoplasm are crucial for orchestrating this complex cellular transition. The oocyte cytoplasmic lattices (CPLs) have long been predicted to function as a storage form for the maternal contribution of ribosomes to the early embryo. We recently demonstrated that the CPLs cannot be visualized in Padi6-/- oocytes and that Padi6-/- embryos arrest at the two-cell stage. Here, we present evidence further supporting the association of ribosomes with the CPLs by demonstrating that the sedimentation properties of the small ribosomal subunit protein, S6, are dramatically altered in Padi6-/- oocytes. We also show that the abundance and localization of ribosomal components is dramatically affected in Padi6-/- two-cell embryos and that de novo protein synthesis is also dysregulated in these embryos. Finally, we demonstrate that embryonic genome activation (EGA) is defective in Padi6-/- two-cell embryos. These results suggest that, in mammals, ribosomal components are stored in the oocyte CPLs and are required for protein translation during early development.


Asunto(s)
Citoplasma/metabolismo , Embrión de Mamíferos/embriología , Embrión de Mamíferos/metabolismo , Hidrolasas/metabolismo , Oocitos/metabolismo , Ribosomas/metabolismo , Animales , Citoplasma/ultraestructura , Embrión de Mamíferos/ultraestructura , Femenino , Regulación del Desarrollo de la Expresión Génica , Genoma/genética , Hidrolasas/deficiencia , Hidrolasas/genética , Ratones , Ratones Noqueados , Microscopía Electrónica , Oocitos/ultraestructura , Biosíntesis de Proteínas/genética , Arginina Deiminasa Proteína-Tipo 6 , Desiminasas de la Arginina Proteica , Ribosomas/ultraestructura , Solubilidad
16.
J Sex Med ; 5(6): 1334-44, 2008 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-18331272

RESUMEN

INTRODUCTION: The immunophilin-ligand FK506 has been shown to ameliorate erectile function and preserve cavernous nerve (CN) architecture in short-term-studies using rat models of CN injury. AIM: The aim of this series was to ascertain the optimal dose and timing of FK506 administration in this animal model. METHODS: Rats underwent bilateral CN crush and were treated with FK506 at different time points. There were control (C) and sham groups for each time point. Based on preliminary experiments, the CN-crush rats had no treatment (C) or either FK506 1 mg/kg (BL) or 3.2 mg/kg (BH) for 3 days prior to and the day of CN crush (PRE), on the day of and for 3 days following CN crush (POST) and for 3 days pre-, on the day of, and 3 days post-CN crush (PP). MAIN OUTCOME MEASUREMENTS: All animals had measurement of intracavernosal pressure/mean arterial blood pressure (ICP/MAP) ratios at 28 days post-CN crush. Structural analysis was conducted in the POST groups. Penile tissue was assessed for apoptosis with terminal deoxynucleotidyl transferase biotin-dUTP nick end labeling assay and immunohistochemically for neural factors (growth associated protein 43 [GAP43], nerve growth factor [NGF], and neural nitric oxide synthase [nNOS]). The CN architecture was examined by transmission electron microscopy (TEM). RESULTS: Sham animals had an ICP/MAP ratio of 70%. Only the BH-POST group revealed an improved ICP/MAP ratio compared with C (50 +/- 9% vs. 32 +/- 8%, P < 0.01). nNOS staining was significantly restored reaching sham levels in BL-POST and BH-POST groups vs. C (P < 0.05). NGF and GAP43 staining displayed no significant differences between C and treatment groups (P < 0.05). Apoptosis was significantly reduced in BL-POST and BH-POST groups compared with C (16 +/- 4%, 21 +/- 9%, and 63 +/- 7%, P < 0.001). TEM exhibited preservation of CN architecture for BH-POST compared with C. CONCLUSION: These results suggest that short-term treatment with doses of FK506 higher than previously utilized preserves erectile function in the rat CN-injury model. Pretreatment appears to offer no advantage. However, FK506 administration just prior to CN injury and for a short-time post-injury achieves the best functional and structural preservation outcomes.


Asunto(s)
Fármacos Neuroprotectores/administración & dosificación , Erección Peniana/efectos de los fármacos , Pene/lesiones , Pene/inervación , Tacrolimus/administración & dosificación , Animales , Apoptosis , Presión Sanguínea , Relación Dosis-Respuesta a Droga , Esquema de Medicación , Proteína GAP-43/metabolismo , Inmunohistoquímica , Masculino , Microscopía Electrónica de Transmisión , Modelos Animales , Factor de Crecimiento Nervioso/metabolismo , Óxido Nítrico Sintasa/metabolismo , Pene/metabolismo , Pene/patología , Ratas , Ratas Sprague-Dawley
17.
J Sex Med ; 5(5): 1126-1136, 2008 May.
Artículo en Inglés | MEDLINE | ID: mdl-18331274

RESUMEN

INTRODUCTION: Radical prostatectomy (RP) is associated with erectile dysfunction (ED). A single, placebo-controlled, human study has assessed the effects of regular sildenafil use after RP and demonstrated an increased chance of preservation of preoperative erectile function. Aim. This study was undertaken to define the effects of such a regimen in an animal model. METHODS: Using the cavernous nerve (CN) crush injury model, animals were divided into a number of groups: no CN injury (sham), bilateral CN injury exposed to either no sildenafil (control) or sildenafil at two doses (10 and 20 mg/kg) subcutaneously daily for three different durations (3, 10, 28 days). MAIN OUTCOME MEASURES: At these time points, CN electrical stimulation was used to assess erectile function by mean intracavernosal pressure (ICP)/mean arterial pressure (MAP) ratio. For the structural analyses, whole rat penes were harvested. Staining for Masson's trichrome was utilized to calculate the smooth muscle-collagen ratio. Immunohistochemical antibody staining was performed for endothelial (CD31 and eNOS) and neural (GAP43, NGF, and nNOS) factors and immunoblotting was performed to analyze the AKT/eNOS pathway. Terminal deoxynucleotidyl transferase biotin-dUTP nick end labeling (TUNEL) assay was used for the assessment of apoptotic indices and the CN architecture was evaluated by transmission electron microscopy (TEM). RESULTS: Erectile function was improved with sildenafil in a time- and dose-dependent fashion with maximization of erectile function recovery occurring with daily 20 mg/kg at the 28-day time point. Sildenafil use resulted in smooth muscle-collagen ratio protection and CD31 and eNOS expression preservation. Sildenafil reduced apoptotic indices significantly compared with control. Animals exposed to sildenafil had increased phosphorylation of akt and eNOS. Tem demonstrated distinct differences in architecture between control and sildenafil groups toward an increased amount of myelinized nerve fibers. CONCLUSIONS: Sildenafil use in the CN crush injury model preserves erectile function that appears to be mediated predominantly through preservation of smooth muscle content and endothelial function as well as through reduction in apoptosis.


Asunto(s)
Disfunción Eréctil/tratamiento farmacológico , Pene/lesiones , Pene/inervación , Piperazinas/administración & dosificación , Sulfonas/administración & dosificación , Vasodilatadores/administración & dosificación , Animales , Apoptosis/efectos de los fármacos , Colágeno/metabolismo , Relación Dosis-Respuesta a Droga , Esquema de Medicación , Estimulación Eléctrica , Disfunción Eréctil/etiología , Masculino , Modelos Animales , Músculo Liso Vascular/metabolismo , Compresión Nerviosa , Fibras Nerviosas Mielínicas/patología , Óxido Nítrico Sintasa de Tipo III/metabolismo , Pene/patología , Fosforilación , Molécula-1 de Adhesión Celular Endotelial de Plaqueta/metabolismo , Proteínas Proto-Oncogénicas c-akt/metabolismo , Purinas/administración & dosificación , Ratas , Ratas Sprague-Dawley , Citrato de Sildenafil
18.
Virus Res ; 132(1-2): 76-85, 2008 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-18054406

RESUMEN

Bovine Viral Diarrhea Virus (BVDV) is a positive sense, single-stranded RNA virus which exhibits two biotypes in standard cell culture systems. The cytopathic strains of this virus (cpBVDV) induce dramatic cytoplasmic vacuolization in cell cultures, while infection with the non-cytopathic (NCP-BVDV) strains produces no overt changes in the host cells. Our results show that extensive cytoplasmic vacuolization is the earliest morphological change in response to cpBVDV infection in MDBK cells. Cells with extensive vacuolization showed no co-existing chromatin condensation, caspase activation, or loss of membrane integrity. In addition, the caspase inhibitor (zVAD-fmk), although improving cell viability of infected cells from 6.7+/-2.2% to 18.8+/-2.2%, did not prevent vacuolization. On the ultrastructural level, the virus-induced cytoplasmic vacuoles are single membrane structures containing organelles and cellular debris, which appear capable of fusing with other vacuoles and engulfing surrounding cytoplasmic materials. LysoTracker Red which marks lysosomes did not stain the virus-induced cytoplasmic vacuoles. In addition, this lysosomal dye could be observed in the cytoplasm of vacuolized cells, suggesting a lysosomal abnormality. Our data demonstrate that cpBVDV induced a novel cell death pathway in MDBK cells that is primarily associated with lysosomal dysfunction and the formation of phagocytic cytoplasmic vacuoles, and this mode of cell death is different from apoptosis and necrosis.


Asunto(s)
Virus de la Diarrea Viral Bovina/fisiología , Vacuolas/virología , Clorometilcetonas de Aminoácidos/farmacología , Animales , Inhibidores de Caspasas , Caspasas/metabolismo , Bovinos , Supervivencia Celular/efectos de los fármacos , Células Cultivadas , Inhibidores de Cisteína Proteinasa/farmacología , Efecto Citopatogénico Viral/efectos de los fármacos , Lisosomas/fisiología , Lisosomas/virología , Microscopía Electrónica , Vacuolas/fisiología , Vacuolas/ultraestructura , Proteínas no Estructurales Virales/análisis , Proteínas no Estructurales Virales/metabolismo
19.
Am J Physiol Heart Circ Physiol ; 292(1): H285-94, 2007 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-16963618

RESUMEN

Chronic kidney diseases are accompanied by the accumulation of substances like asymmetric dimethylarginine, phenylacetic acid, homocysteine, and advanced glycation end products, known to either inhibit endothelial nitric oxide synthase (eNOS) or uncouple it, consequently limiting the amount of available nitric oxide (NO). Reduced bioavailability of NO induces endothelial dysfunction. An early loss of peritubular capillaries in tubulointerstitial fibrotic areas and injury to endothelial cells have been linked to progressive renal disease. Screening endothelial genes in cells treated with NOS inhibitors showed upregulation of collagen XVIII, a precursor of a potent antiangiogenic substance, endostatin. This finding was confirmed at the level of mRNA and protein expression. Tie-2 promoter-driven green fluorescent protein mice treated with nonhypertensinogenic doses of a NOS inhibitor exhibited upregulation of collagen XVIII/endostatin and rarefaction of capillary profiles. This was accompanied by the increased expression of transforming growth factor-beta and connective tissue growth factor in the kidney. Occasional endothelial cells expressed both the marker of endothelial lineage (green fluorescent protein) and mesenchymal marker (alpha-smooth muscle actin or calponin). In vitro studies of endothelial cells treated with asymmetric dimethylarginine showed decreased expression of eNOS and Flk-1 and enhanced expression of calponin and fibronectin, additional markers of smooth muscle and mesenchymal cells. These cells overexpressed transforming growth factor-beta and connective tissue growth factor, as well as endostatin. In conclusion, data presented here 1) ascribe to NO deficiency in endothelial cells the function of a profibrotic stimulus associated with the expression of an antiangiogenic fragment of collagen XVIII (endostatin) and 2) provide evidence of endothelial-mesenchymal transdifferentiation in the course of inhibition of NOS by a pathophysiologically important antagonist, asymmetric dimethylarginine. Both mechanisms may account for microvascular rarefaction.


Asunto(s)
Células Endoteliales/citología , Células Endoteliales/metabolismo , Inhibidores Enzimáticos/farmacología , Células Madre Mesenquimatosas/citología , Células Madre Mesenquimatosas/metabolismo , Óxido Nítrico Sintasa/antagonistas & inhibidores , Factores de Crecimiento Transformadores/metabolismo , Animales , Diferenciación Celular/efectos de los fármacos , Células Cultivadas , Células Endoteliales/efectos de los fármacos , Factores de Crecimiento Endotelial/metabolismo , Humanos , Células Madre Mesenquimatosas/efectos de los fármacos , Ratones , Ratones Noqueados , Óxido Nítrico/metabolismo
20.
Am J Physiol Cell Physiol ; 290(3): C832-43, 2006 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-16251472

RESUMEN

Caveolae and their associated structural proteins, the caveolins, are specialized plasmalemmal microdomains involved in endocytosis and compartmentalization of cell signaling. We examined the expression and distribution of caveolae and caveolins in retinal pigment epithelium (RPE), which plays key roles in retinal support, visual cycle, and acts as the main barrier between blood and retina. Electron microscopic observation of rat RPE, in situ primary cultures of rat and human RPE and a rat RPE cell line (RPE-J) demonstrated in all cases the presence of caveolae in both apical and basolateral domains of the plasma membrane. Caveolae were rare in RPE in situ but were frequent in primary RPE cultures and in RPE-J cells, which correlated with increased levels in the expression of caveolin-1 and -2. The bipolar distribution of caveolae in RPE is striking, as all other epithelial cells examined to date (liver, kidney, thyroid, and intestinal) assemble caveolae only at the basolateral side. This might be related to the nonpolar distribution of both caveolin-1 and 2 in RPE because caveolin-2 is basolateral and caveolin-1 nonpolar in other epithelial cells. The bipolar localization of plasmalemmal caveolae in RPE cells may reflect specialized roles in signaling and trafficking important for visual function.


Asunto(s)
Caveolas/metabolismo , Epitelio Pigmentado Ocular/citología , Epitelio Pigmentado Ocular/metabolismo , Animales , Caveolina 1/genética , Caveolina 1/metabolismo , Caveolina 2/genética , Caveolina 2/metabolismo , Línea Celular , Perros , Regulación de la Expresión Génica , Humanos , Epitelio Pigmentado Ocular/ultraestructura , Ratas
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...