Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 31
Filtrar
1.
Sci Adv ; 10(11): eadg9278, 2024 Mar 15.
Artículo en Inglés | MEDLINE | ID: mdl-38478616

RESUMEN

Canonical Wnt and sphingosine-1-phosphate (S1P) signaling pathways are highly conserved systems that contribute to normal vertebrate development, with key consequences for immune, nervous, and cardiovascular system function; despite these functional overlaps, little is known about Wnt/ß-catenin-S1P cross-talk. In the vascular system, both Wnt/ß-catenin and S1P signals affect vessel maturation, stability, and barrier function, but information regarding their potential coordination is scant. We report an instance of functional interaction between the two pathways, including evidence that S1P receptor 1 (S1PR1) is a transcriptional target of ß-catenin. By studying vascular smooth muscle cells and arterial injury response, we find a specific requirement for the ß-catenin carboxyl terminus, which acts to induce S1PR1, and show that this interaction is essential for vascular remodeling. We also report that pharmacological inhibition of the ß-catenin carboxyl terminus reduces S1PR1 expression, neointima formation, and atherosclerosis. These findings provide mechanistic understanding of how Wnt/ß-catenin and S1P systems collaborate during vascular remodeling and inform strategies for therapeutic manipulation.


Asunto(s)
Aterosclerosis , Cateninas , Lisofosfolípidos , Esfingosina/análogos & derivados , Humanos , Cateninas/metabolismo , beta Catenina/metabolismo , Remodelación Vascular , Transducción de Señal
2.
Front Neurosci ; 17: 1227144, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-37811322

RESUMEN

Xanthogranulomas are considered rare tumors, with their sellar and non-sellar frequency ranging from 1.6 to 7% among intracranial lesions, and described as a separate entity by the World Health Organization in 2000. The diagnosis of sellar xanthogranulomas is challenging, given their uncertain origin and clinical course. In addition, the limited reporting of sellar xanthogranuloma cases and the absence of characteristic images make these entities difficult to distinguish from other cystic lesions of the sellar region, such as adamantinomatous craniopharyngiomas, Rathke's cleft cysts, pituitary tumors, arachnoid cysts, epidermoid cysts, and dermoid cysts. Here, we describe the clinical presentation, radiological findings, immunohistochemical/histopathological analysis, and the ultrastructural examination by transmission electron microscopy of five sellar xanthogranulomas cases reported in two care centers in Cordoba, Argentina. Two males and three females between 37 and 73 years of age (average 51.8 years) presented with persistent headaches, generalized endocrine defects, and visual problems. MRI revealed cystic formations in the sellar region, which usually projected into adjacent tissues such as the suprasellar region or cavernous sinuses, and compressed other structures such as the optic chiasm, pituitary gland, and cranial nerves. All patients underwent surgical intervention to remove the tumor tissue. The histopathological analysis of the samples showed cellular tissue with a xanthogranulomatous appearance, inflammatory cellular infiltrate (mainly lymphocytes and macrophages), fibroblasts, abundant collagen fibers, and hemorrhages. An ultrastructural analysis helped to identify cellular infiltrates and granules resulting from tumor cell activity. The data support the hypothesis that sellar xanthogranulomas could occur as an inflammatory reaction secondary to the rupture and hemorrhage of a previous cystic process, thereby generating an expansion of the tumor body toward adjacent tissues. The information obtained from these cases contributes to the current knowledge about this disease's origin and clinical and histological evolution. However, the scarcity of patients and the observed phenotypic heterogeneity make its diagnosis still challenging. Undoubtedly, more investigations are needed to provide additional information in order to be able to achieve a more accurate diagnosis and effective treatment of this rare disease.

4.
Exp Gerontol ; 148: 111304, 2021 06.
Artículo en Inglés | MEDLINE | ID: mdl-33676974

RESUMEN

The human prostate is an androgen-dependent gland where an imbalance in cell proliferation can lead to benign prostatic hyperplasia (BPH), which results in voiding lower urinary tract symptoms in the elderly. In the last decades, novel evidence has suggested that BPH might represent an element into the wide spectrum of disorders conforming the Metabolic Syndrome (MS). The dyslipidemic state and the other atherogenic factors of the MS have been shown to induce, maintain and/or aggravate the pathological growth of different organs, with data regarding the prostate being still limited. We here review the available epidemiological and experimental studies about the association of BPH with dyslipidemias. In particular, we have focused on Oxidized Low-Density Lipoproteins (OxLDL) as a potential trigger for vascular disease and cellular proliferation in atherogenic contexts, analyzing their putative molecular mechanisms, including the induction of specific extracellular vesicles (EVs)-derived miRNAs. In addition to the epidemiological evidence, OxLDL is proposed to play a fundamental role in the upregulation of prostatic cell proliferation by activating the Rho/Akt/p27Kip1 pathway in atherogenic contexts. miR-21, miR-141, miR-143, miR-145, miR-155, and miR-221 would be involved in the transcription of genes related to the proliferative process. Although much remains to be investigated regarding the impact of OxLDL, its receptors, and molecular mechanisms on the prostate, it is clear that EVs and miRNAs represent a promising target for proliferative pathologies of the prostate gland.


Asunto(s)
Aterosclerosis , Vesículas Extracelulares , MicroARNs , Hiperplasia Prostática , Anciano , Humanos , Masculino , MicroARNs/genética
5.
Int J Pharm ; 591: 119985, 2020 Dec 15.
Artículo en Inglés | MEDLINE | ID: mdl-33069891

RESUMEN

Androgens play a central role in homeostatic and pathological processes of the prostate gland. At the cellular level, testosterone activates both the genomic signaling pathway, through the intracellular androgen receptor (AR), and membrane-initiated androgen signaling (MIAS), by plasma membrane receptors. We have previously shown that the activation of MIAS induces uncontrolled proliferation and fails to stimulate the beneficial immunomodulatory effects of testosterone in prostatic cells, becoming necessary to investigate if genomic signaling mediates homeostatic effects of testosterone. However, the lack of specific modulators for genomic androgen signaling has delayed the understanding of this mechanism. In this article, we demonstrate that monosialoganglioside (GM1) micelles are capable of delivering testosterone into the cytoplasm to specifically activate genomic signaling. Stimulation with testosterone-loaded GM1 micelles led to the activation of androgen response element (ARE)-regulated genes in vitro as well as to the recovery of normal prostate size and histology after castration in mice. In addition, these micelles avoided MIAS, as demonstrated by the absence of rapid signaling pathway activation and the inability to induce uncontrolled cell proliferation. In conclusion, our results validate a novel tool for the specific activation of genomic androgen signaling and demonstrate the importance of selective pathway activation in androgen-mediated proliferation.


Asunto(s)
Neoplasias de la Próstata , Receptores Androgénicos , Andrógenos , Animales , Gangliósido G(M1) , Genómica , Humanos , Masculino , Ratones , Micelas , Receptores Androgénicos/genética , Transducción de Señal , Testosterona
6.
J Reprod Immunol ; 142: 103190, 2020 11.
Artículo en Inglés | MEDLINE | ID: mdl-32853844

RESUMEN

Mucosal immunity defines the relationship of surfaces in contact with the environment and integrates diverse tissues such as epidermis, gum, nose, gut, uterus and prostate with the immune system. Although considered part of a system, each mucosa presents specific immune features beyond the barrier and secretory functions. Information regarding the mucosal immunology of the male reproductive tract and the prostate gland in particular is scarce. In this review, we approach the prostate as an epithelial barrier and as part of the mucosal immune system. Finally, we also raise a series of questions that will improve the understanding of this gland, its role in reproduction and its sensitivity/resistance to disease.


Asunto(s)
Inmunidad Mucosa , Próstata/inmunología , Infecciones del Sistema Genital/inmunología , Semen/inmunología , Transducción de Señal/inmunología , Animales , Modelos Animales de Enfermedad , Interacciones Huésped-Patógeno/inmunología , Humanos , Inmunoglobulina A/metabolismo , Inmunoglobulina G/metabolismo , Masculino , Proteínas NLR/metabolismo , Próstata/metabolismo , Próstata/microbiología , Infecciones del Sistema Genital/microbiología , Semen/metabolismo , Receptores Toll-Like/metabolismo
7.
PLoS One ; 15(5): e0226233, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-32379832

RESUMEN

Allergic asthma is the most common phenotype of the pathology, having an early-onset in childhood and producing a Th2-driven airways remodeling process that leads to symptoms and pathophysiological changes. The avoidance of aeroallergen exposure in early life has been shown to prevent asthma, but without repeated success and with the underlying preventive mechanisms at the beginning of asthma far to be fully recognized. In the present study, we aimed to evaluate if neonatal LPS-induced boost in epithelial host defenses contribute to prevent OVA-induced asthma in adult mice. To this, we focused on the response of bronchiolar club cells (CC), which are highly specialized in maintaining the epithelial homeostasis in the lung. In these cells, neonatal LPS administration increased the expression of TLR4 and TNFα, as well as the immunodulatory/antiallergic proteins: club cell secretory protein (CCSP) and surfactant protein D (SP-D). LPS also prevented mucous metaplasia of club cells and reduced the epidermal growth factor receptor (EGFR)-dependent mucin overproduction, with mice displaying normal breathing patterns after OVA challenge. Furthermore, the overexpression of the epithelial Th2-related molecule TSLP was blunted, and normal TSLP and IL-4 levels were found in the bronchoalveolar lavage. A lower eosinophilia was detected in LPS-pretreated mice, along with an increase in phagocytes and regulatory cells (CD4+CD25+FOXP3+ and CD4+IL-10+), together with higher levels of IL-12 and TNFα. In conclusion, our study demonstrates stable asthma-preventive epithelial effects promoted by neonatal LPS stimulation, leading to the presence of regulatory cells in the lung. These anti-allergic dynamic mechanisms would be overlaid in the epithelium, favored by an adequate epidemiological environment, during the development of asthma.


Asunto(s)
Asma/inmunología , Bronquiolos/efectos de los fármacos , Bronquiolos/inmunología , Citocinas/metabolismo , Epitelio/inmunología , Inmunidad Innata , Lipopolisacáridos/inmunología , Lipopolisacáridos/farmacología , Alérgenos/inmunología , Animales , Animales Recién Nacidos , Asma/prevención & control , Modelos Animales de Enfermedad , Epitelio/efectos de los fármacos , Ratones , Ratones Endogámicos BALB C , Ovalbúmina/inmunología , Ovalbúmina/farmacología , Linfocitos T Reguladores/efectos de los fármacos , Linfocitos T Reguladores/inmunología
8.
Front Immunol ; 9: 1980, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-30233581

RESUMEN

Neutrophils are major effectors of acute inflammation against infection and tissue damage, with ability to adapt their phenotype according to the microenvironment. Although sex hormones regulate adaptive immune cells, which explains sex differences in immunity and infection, little information is available about the effects of androgens on neutrophils. We therefore aimed to examine neutrophil recruitment and plasticity in androgen-dependent and -independent sites under androgen manipulation. By using a bacterial model of prostate inflammation, we showed that neutrophil recruitment was higher in testosterone-treated rats, with neutrophil accumulation being positively correlated to serum levels of testosterone and associated to stronger inflammatory signs and tissue damage. Testosterone also promoted LPS-induced neutrophil recruitment to the prostate, peritoneum, and liver sinusoids, as revealed by histopathology, flow cytometry, and intravital microscopy. Strikingly, neutrophils in presence of testosterone exhibited an impaired bactericidal ability and a reduced myeloperoxidase activity. This inefficient cellular profile was accompanied by high expression of the anti-inflammatory cytokines IL10 and TGFß1, which is compatible with the "N2-like" neutrophil phenotype previously reported in the tumor microenvironment. These data reveal an intriguing role for testosterone promoting inefficient, anti-inflammatory neutrophils that prolong bacterial inflammation, generating a pathogenic environment for several conditions. However, these immunomodulatory properties might be beneficially exploited in autoimmune and other non-bacterial diseases.


Asunto(s)
Andrógenos/metabolismo , Infecciones por Escherichia coli/inmunología , Neutrófilos/inmunología , Prostatitis/inmunología , Testosterona/metabolismo , Infecciones Urinarias/inmunología , Escherichia coli Uropatógena/fisiología , Andrógenos/administración & dosificación , Animales , Células Cultivadas , Modelos Animales de Enfermedad , Humanos , Interleucina-10/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Infiltración Neutrófila , Ratas , Ratas Wistar , Testosterona/administración & dosificación , Factor de Crecimiento Transformador beta/metabolismo , Microambiente Tumoral
9.
Front Immunol ; 9: 379, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-29545799

RESUMEN

Galectins, a family of animal lectins characterized by their affinity for N-acetyllactosamine-enriched glycoconjugates, modulate several immune cell processes shaping the course of innate and adaptive immune responses. Through interaction with a wide range of glycosylated receptors bearing complex branched N-glycans and core 2-O-glycans, these endogenous lectins trigger distinct signaling programs thereby controling immune cell activation, differentiation, recruitment and survival. Given the unique features of mucosal inflammation and the differential expression of galectins throughout the gastrointestinal tract, we discuss here key findings on the role of galectins in intestinal inflammation, particularly Crohn's disease, ulcerative colitis, and celiac disease (CeD) patients, as well as in murine models resembling these inflammatory conditions. In addition, we present new data highlighting the regulated expression of galectin-1 (Gal-1), a proto-type member of the galectin family, during intestinal inflammation in untreated and treated CeD patients. Our results unveil a substantial upregulation of Gal-1 accompanying the anti-inflammatory and tolerogenic response associated with gluten-free diet in CeD patients, suggesting a major role of this lectin in favoring resolution of inflammation and restoration of mucosal homeostasis. Thus, a coordinated network of galectins and their glycosylated ligands, exerting either anti-inflammatory or proinflammatory responses, may influence the interplay between intestinal epithelial cells and the highly specialized gut immune system in physiologic and pathologic settings.


Asunto(s)
Enfermedad Celíaca/inmunología , Galectina 1/metabolismo , Inflamación/inmunología , Mucosa Intestinal/inmunología , Intestinos/inmunología , Animales , Diferenciación Celular , Galectina 1/genética , Homeostasis , Humanos , Tolerancia Inmunológica , Ratones , Ratones Noqueados
10.
Mol Cancer Res ; 16(5): 833-845, 2018 05.
Artículo en Inglés | MEDLINE | ID: mdl-29523762

RESUMEN

Emerging evidence suggests that unregulated Toll-like receptor (TLR) signaling promotes tumor survival signals, thus favoring tumor progression. Here, the mechanism underlying TLR4 overexpression in papillary thyroid carcinomas (PTC) mainly harboring the BRAFV600E mutation was studied. TLR4 was overexpressed in PTC compared with nonneoplastic thyroid tissue. Moreover, paired clinical specimens of primary PTC and its lymph node metastasis showed a significant upregulation of TLR4 levels in the metastatic tissues. In agreement, conditional BRAFV600E expression in normal rat thyroid cells and mouse thyroid tissue upregulated TLR4 expression levels. Furthermore, functional TLR4 expression was demonstrated in PTC cells by increased NF-κB transcriptional activity in response to the exogenous TLR4-agonist lipopolysaccharide. Of note, The Cancer Genome Atlas data analysis revealed that BRAFV600E-positive tumors with high TLR4 expression were associated with shorter disease-free survival. Transcriptomic data analysis indicated a positive correlation between TLR4 expression levels and MAPK/ERK signaling activation. Consistently, chemical blockade of MAPK/ERK signaling abrogated BRAFV600E-induced TLR4 expression. A detailed study of the TLR4 promoter revealed a critical MAPK/ERK-sensitive Ets-binding site involved in BRAFV600E responsiveness. Subsequent investigation revealed that the Ets-binding factor ETS1 is critical for BRAFV600E-induced MAPK/ERK signaling-dependent TLR4 gene expression. Together, these data indicate that functional TLR4 overexpression in PTCs is a consequence of thyroid tumor-oncogenic driver dysregulation of MAPK/ERK/ETS1 signaling.Implications: Considering the participation of aberrant NF-κB signaling activation in the promotion of thyroid tumor growth and the association of high TLR4 expression with more aggressive tumors, this study suggests a prooncogenic potential of TLR4 downstream signaling in thyroid tumorigenesis. Mol Cancer Res; 16(5); 833-45. ©2018 AACR.


Asunto(s)
Proteínas Quinasas Activadas por Mitógenos/metabolismo , Proteína Proto-Oncogénica c-ets-1/genética , Cáncer Papilar Tiroideo/genética , Cáncer Papilar Tiroideo/metabolismo , Receptor Toll-Like 4/metabolismo , Animales , Femenino , Humanos , Masculino , Ratones , Ratones Transgénicos , Proteínas Quinasas Activadas por Mitógenos/genética , Proteína Proto-Oncogénica c-ets-1/metabolismo , Ratas , Ratas Endogámicas F344 , Transducción de Señal , Cáncer Papilar Tiroideo/patología , Receptor Toll-Like 4/genética , Transfección
11.
Endocrinology ; 159(2): 945-956, 2018 02 01.
Artículo en Inglés | MEDLINE | ID: mdl-29194490

RESUMEN

Androgen signaling in prostate smooth muscle cells (pSMCs) is critical for the maintenance of prostate homeostasis, the alterations of which are a central aspect in the development of pathological conditions. Testosterone can act through the classic androgen receptor (AR) in the cytoplasm, eliciting genomic signaling, or through different types of receptors located at the plasma membrane for nongenomic signaling. We aimed to find evidence of nongenomic testosterone-signaling mechanisms in pSMCs and their participation in cell proliferation, differentiation, and the modulation of the response to lipopolysaccharide. We demonstrated that pSMCs can respond to testosterone by a rapid activation of ERK1/2 and Akt. Furthermore, a pool of ARs localized at the cell surface of pSMCs is responsible for a nongenomic testosterone-induced increase in cell proliferation. Through membrane receptor stimulation, testosterone favors a muscle phenotype, indicated by an increase in smooth muscle markers. We also showed that the anti-inflammatory effects of testosterone, capable of attenuating lipopolysaccharide-induced proinflammatory actions, are promoted only by receptors located inside the cell. We postulate that testosterone might perform prohomeostatic effects through intracellular-initiated mechanisms by modulating cell proliferation and inflammation, whereas some pathological, hyperproliferative actions would be induced by membrane-initiated nongenomic signaling in pSMCs.


Asunto(s)
Músculo Liso/efectos de los fármacos , Miocitos del Músculo Liso/efectos de los fármacos , Próstata/efectos de los fármacos , Receptores Androgénicos/metabolismo , Testosterona/farmacología , Animales , Células Cultivadas , Masculino , Músculo Liso/metabolismo , Miocitos del Músculo Liso/metabolismo , Próstata/citología , Próstata/metabolismo , Ratas Wistar , Transducción de Señal/efectos de los fármacos , Distribución Tisular
12.
Cell Biol Int ; 41(11): 1234-1238, 2017 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-28493535

RESUMEN

One of the recognized issues in prostate cancer research is the lack of animal models allowing the research of pathological, biochemical, and genetic factors in immunocompetent animals. Our research group has successfully employed the gerbil in several studies for prostate diseases. In the present work, we aimed to analyze the effect of chronic bacterial inflammation on N-methyl-N-nitrosourea (MNU)-induced prostate carcinogenesis in gerbils. Histopathological assessment of the prostatic complex revealed that treatment combinations with MNU plus testosterone or bacterial infection resulted in a promotion of prostate cancer, with bacterial inflammation being more effective in increasing premalignant and malignant tissular alterations than testosterone in the prostate. Furthermore, chronic bacterial inflammation itself induced premalignant lesions in the ventral lobe and increased their frequency in the dorsolateral lobe as well as malignant lesions in the ventral prostate. These animals showed a rich inflammatory microenvironment, characterized as intraluminal and periductal foci. These data indicate that chronic inflammation induced by Escherichia coli acts as a potent tumor promoter, in the early stages of carcinogenesis in the gerbil, in line with the hypothesis of inflammation supporting several steps of tumor development in the prostate gland.


Asunto(s)
Modelos Animales de Enfermedad , Gerbillinae , Neoplasias de la Próstata/metabolismo , Neoplasias de la Próstata/patología , Animales , Carcinogénesis , Inflamación/inducido químicamente , Inflamación/metabolismo , Inflamación/patología , Masculino , Metilnitrosourea , Neoplasias de la Próstata/inducido químicamente
13.
Cell Biol Int ; 41(11): 1223-1233, 2017 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-28244686

RESUMEN

The prostate gland is a strictly androgen-dependent organ which is also the main target of infectious and inflammatory diseases in the male reproductive tract. Host defenses and immunity of the gland have unique features to maintain a constant balance between response and tolerance to diverse antigens. In this context, the effects of reproductive hormones on the male tract are thus complex and have just started to be defined. From the classical description of "the prostatic antibacterial factor," many host defense proteins with potent microbicidal and anti-tumoral activities have been described in the organ. Indeed, it has been proposed a central role for resident cells, that is, epithelial and smooth muscle cells, in the prostatic response against injuries. However, these cells also represent the target of the inflammatory damage, leading to the development of a Proliferative Inflammatory Atrophy-like process in the epithelium and a myofibroblastic-like reactive stroma. Available data on androgen regulation of inflammation led to a model of the complex control, in which the final effect will depend on the tissue microenvironment, the cause of inflammation, and the levels of androgens among other factors. In this paper, we review the current scientific literature about the inflammatory process in the gland, the modulation of host defense proteins, and the influence of testosterone on the resolution of prostatitis.


Asunto(s)
Andrógenos/inmunología , Próstata/inmunología , Andrógenos/metabolismo , Andrógenos/fisiología , Animales , Humanos , Inflamación/inmunología , Inflamación/metabolismo , Masculino , Próstata/metabolismo , Receptores Androgénicos/inmunología , Receptores Androgénicos/metabolismo , Testosterona/inmunología , Testosterona/metabolismo
14.
Artículo en Inglés | MEDLINE | ID: mdl-28352169

RESUMEN

In spite of the numerous studies on chronic obstructive pulmonary disease (COPD), the cellular and molecular basis of the disease's development remain unclear. Neutrophils and eosinophils are known to be key players in COPD. Recently, neutrophil extracellular trap cell death (NETosis), a mechanism due to decondensation and extrusion of chromatin to form extracellular traps, has been demonstrated in COPD. However, there is limited knowledge about eosinophil extracellular trap cell death (EETosis) and its role in the pathogenesis of COPD. The aim of this study was to evaluate EETosis in stable COPD. Induced sputum obtained from healthy smokers and low exacerbation risk COPD A or B group patients or high exacerbation risk COPD C or D group patients were included. Samples were examined using electron microscopy and immunofluorescence. Healthy smokers (n=10) and COPD A (n=19) group exhibited neutrophilic or paucigranulocytic phenotypes, with NETosis being absent in these patients. In contrast, COPD B (n=29), with eosinophilic or mixed phenotypes, showed EETosis and incipient NETosis. COPD C (n=18) and COPD D groups (n=13) were differentiated from low exacerbation rate-COPD group by the abundant cellular debris, with COPD C group having an eosinophilic pattern and numerous cells undergoing EETosis. A hallmark of this group was the abundant released membranes that often appeared phagocytosed by neutrophils, which coincidentally exhibited early NETosis changes. The COPD D group included patients with a neutrophilic or mixed pattern, with abundant neutrophil extracellular trap-derived material. This study is the first to demonstrate EETosis at different stages of stable COPD. The results suggest a role for eosinophils in COPD pathophysiology, especially at the beginning and during the persistence of the disease, regardless of whether the patient quit smoking, with EETosis debris probably triggering uncontrolled NETosis. The main target of these findings should be young smokers with the potential to develop COPD.


Asunto(s)
Eosinófilos/ultraestructura , Trampas Extracelulares/metabolismo , Pulmón/ultraestructura , Neutrófilos/ultraestructura , Enfermedad Pulmonar Obstructiva Crónica/patología , Estudios de Casos y Controles , Muerte Celular , Estudios Transversales , Eosinófilos/metabolismo , Femenino , Técnica del Anticuerpo Fluorescente , Volumen Espiratorio Forzado , Humanos , Pulmón/metabolismo , Pulmón/fisiopatología , Masculino , Microscopía Confocal , Microscopía Electrónica , Persona de Mediana Edad , Neutrófilos/metabolismo , Enfermedad Pulmonar Obstructiva Crónica/metabolismo , Enfermedad Pulmonar Obstructiva Crónica/fisiopatología , Fumar/efectos adversos , Cese del Hábito de Fumar , Prevención del Hábito de Fumar , Esputo/citología , Esputo/metabolismo , Capacidad Vital
15.
Circ Res ; 120(11): 1789-1799, 2017 May 26.
Artículo en Inglés | MEDLINE | ID: mdl-28302649

RESUMEN

RATIONALE: Nonclassical mouse monocyte (CX3CR1high, Ly-6Clow) patrolling along the vessels of the microcirculation is critical for endothelial homeostasis and inflammation. Because of technical challenges, it is currently not established how patrolling occurs in large arteries. OBJECTIVE: This study was undertaken to elucidate the molecular, migratory, and functional phenotypes of patrolling monocytes in the high shear and pulsatile environment of large arteries in healthy, hyperlipidemic, and atherosclerotic conditions. METHODS AND RESULTS: Applying a new method for stable, long-term 2-photon intravital microscopy of unrestrained large arteries in live CX3CR1-GFP (green fluorescent protein) mice, we show that nonclassical monocytes patrol inside healthy carotid arteries at a velocity of 36 µm/min, 3× faster than in microvessels. The tracks are less straight but lead preferentially downstream. The number of patrolling monocytes is increased 9-fold by feeding wild-type mice a Western diet or by applying topical TLR7/8 (Toll-like receptor) agonists. A similar increase is seen in CX3CR1+/GFP/apoE-/- mice on chow diet, with a further 2- to 3-fold increase on Western diet (22-fold over healthy). In plaque conditions, monocytes are readily captured onto the endothelium from free flow. Stable patrolling is unaffected in CX3CR1-deficient mice and involves the contribution of LFA-1 (lymphocyte-associated antigen 1) and α4 integrins. The endothelial damage in atherosclerotic carotid arteries was assessed by electron microscopy and correlates with the number of intraluminal patrollers. Abolishing patrolling monocytes in Nr4a1-/- apoE-/- mice leads to pronounced endothelial apoptosis. CONCLUSIONS: Arterial patrolling is a prominent new feature of nonclassical monocytes with unique molecular and kinetic properties. It is highly upregulated in hyperlipidemia and atherosclerosis in a CX3CR1-independent fashion and plays a potential role in endothelial protection.


Asunto(s)
Arterias/metabolismo , Aterosclerosis/metabolismo , Dieta Occidental/efectos adversos , Endotelio Vascular/metabolismo , Monocitos/metabolismo , Receptores de Quimiocina/deficiencia , Animales , Arterias/patología , Aterosclerosis/patología , Receptor 1 de Quimiocinas CX3C , Endotelio Vascular/patología , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados
16.
J Cell Physiol ; 232(10): 2806-2817, 2017 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-27861881

RESUMEN

Prostatic smooth muscle cells (pSMCs) differentiation is a key factor for prostatic homeostasis, with androgens exerting multiple effects on these cells. Here, we demonstrated that the myodifferentiator complex Srf/Myocd is up-regulated by testosterone in a dose-dependent manner in primary cultures of rat pSMCs, which was associated to the increase in Acta2, Cnn1, and Lmod1 expressions. Blocking Srf or Myocd by siRNAs inhibited the myodifferentiator effect of testosterone. While LPS led to a dedifferentiated phenotype in pSMCs, characterized by down-regulation of Srf/Myocd and smooth muscle cell (SMC)-restricted genes, endotoxin treatment on Myocd-overexpressing cells did not result in phenotypic alterations. Testosterone at a physiological dose was able to restore the muscular phenotype by normalizing Srf/Myocd expression in inflammation-induced dedifferentiated pSMCs. Moreover, the androgen reestablished the proliferation rate and IL-6 secretion increased by LPS. These results provide novel evidence regarding the myodifferentiating role of testosterone on SMCs by modulating Srf/Myocd. Thus, androgens preserve prostatic SMC phenotype, which is essential to maintain the normal structure and function of the prostate. J. Cell. Physiol. 232: 2806-2817, 2017. © 2016 Wiley Periodicals, Inc.


Asunto(s)
Desdiferenciación Celular/efectos de los fármacos , Miocitos del Músculo Liso/efectos de los fármacos , Proteínas Nucleares/metabolismo , Testosterona/farmacología , Transactivadores/metabolismo , Factores de Transcripción/metabolismo , Actinas/metabolismo , Animales , Proteínas de Unión al Calcio/metabolismo , Proliferación Celular/efectos de los fármacos , Células Cultivadas , Relación Dosis-Respuesta a Droga , Interleucina-6/metabolismo , Lipopolisacáridos/farmacología , Masculino , Proteínas de Microfilamentos/metabolismo , Miocitos del Músculo Liso/metabolismo , Proteínas Nucleares/genética , Fenotipo , Próstata , Interferencia de ARN , Ratas Wistar , Transducción de Señal/efectos de los fármacos , Transactivadores/genética , Factores de Transcripción/genética , Transfección , Calponinas
18.
Exp Biol Med (Maywood) ; 240(7): 904-16, 2015 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-25504013

RESUMEN

Atopic asthma is a chronic allergic disease that involves T-helper type 2 (Th2)-inflammation and airway remodeling. Bronchiolar club cells (CC) and alveolar macrophages (AM) are sentinel cells of airway barrier against inhaled injuries, where allergy induces mucous metaplasia of CC and the alternative activation of AM, which compromise host defense mechanisms and amplify Th2-inflammation. As there is evidence that high levels of environmental endotoxin modulates asthma, the goal of this study was to evaluate if the activation of local host defenses by Lipopolysaccharide (LPS) previous to allergy development can contribute to preserving CC and AM protective phenotypes. Endotoxin stimulus before allergen exposition reduced hallmarks of allergic inflammation including eosinophil influx, Interleukin-4 and airway hyperreactivity, while the T-helper type 1 related cytokines IL-12 and Interferon-γ were enhanced. This response was accompanied by the preservation of the normal CC phenotype and the anti-allergic proteins Club Cell Secretory Protein (CCSP) and Surfactant-D, thereby leading to lower levels of CC metaplasia and preventing the increase of the pro-Th2 cytokine Thymic stromal lymphopoietin. In addition, classically activated alveolar macrophages expressing nitric oxide were promoted over the alternatively activated ones that expressed arginase-1. We verified that LPS induced a long-term overexpression of CCSP and the innate immune markers Toll-like receptor 4, and Tumor Necrosis Factor-α, changes that were preserved in spite of the allergen challenge. These results demonstrate that LPS pre-exposition modifies the local bronchioalveolar microenvironment by inducing natural anti-allergic mechanisms while reducing local factors that drive Th2 type responses, thus modulating allergic inflammation.


Asunto(s)
Asma/inmunología , Macrófagos Alveolares/inmunología , Mucosa Respiratoria/citología , Mucosa Respiratoria/inmunología , Animales , Western Blotting , Modelos Animales de Enfermedad , Endotoxinas/inmunología , Endotoxinas/toxicidad , Ensayo de Inmunoadsorción Enzimática , Femenino , Técnica del Anticuerpo Fluorescente , Inmunohistoquímica , Ratones , Ratones Endogámicos BALB C , Fenotipo , Uteroglobina/metabolismo
19.
Int J Exp Pathol ; 94(6): 399-411, 2013 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-23998365

RESUMEN

Bronchiolar Clara cells play a critical role in lung homoeostasis. The main goal of this study was to evaluate the effects of chronic allergy on these cells and the efficacy of budesonide (BUD) and montelukast (MK) in restoring their typical phenotypes after ovalbumin-induced chronic allergy in mice. Chronic allergy induced extensive bronchiolar Alcian blue-periodic acid-Schiff (AB/PAS)-positive metaplasia. In addition, cells accumulated numerous big electron-lucent granules negative for Clara cell main secretory protein (CC16), and consequently, CC16 was significantly reduced in bronchoalveolar lavage. A concomitant reduction in SP-D and CYP2E1 content was observed. The phenotypic changes induced by allergy were pharmacologically reversed by both treatments; MK was more efficient than BUD in doing so. MK decreased AB/PAS reactivity to control levels whereas they remained persistently elevated after BUD. Moreover, most non-ciliated cells recovered their normal morphology after MK, whereas for BUD normal cells coexisted with 'transitional' cells that contained remnant mucous granules and stained strongly for CC16 and SP-D. Glucocorticoids were also less able to reduce inflammatory infiltration and maintained higher percentage of neutrophils, which may have contributed to prolonged mucin expression. These results show that chronic allergy-induced mucous metaplasia of Clara cells affects their defensive mechanisms. However, anti-inflammatory treatments were able to re-establish the normal phenotype of Clara cell, with MK being more efficient at restoring a normal profile than BUD. This study highlights the role of epithelial cells in lung injuries and their contribution to anti-inflammatory therapies.


Asunto(s)
Acetatos/uso terapéutico , Asma/tratamiento farmacológico , Asma/patología , Bronquios/patología , Budesonida/uso terapéutico , Fenotipo , Quinolinas/uso terapéutico , Acetatos/farmacología , Animales , Antiasmáticos/farmacología , Antiasmáticos/uso terapéutico , Antiinflamatorios/farmacología , Antiinflamatorios/uso terapéutico , Asma/inducido químicamente , Bronquios/efectos de los fármacos , Bronquios/metabolismo , Budesonida/farmacología , Enfermedad Crónica , Ciclopropanos , Citocromo P-450 CYP2E1/metabolismo , Modelos Animales de Enfermedad , Epitelio/efectos de los fármacos , Epitelio/metabolismo , Epitelio/patología , Femenino , Ratones , Ratones Endogámicos BALB C , Ovalbúmina/efectos adversos , Proteína D Asociada a Surfactante Pulmonar/metabolismo , Quinolinas/farmacología , Sulfuros , Uteroglobina/metabolismo
20.
J Cell Physiol ; 228(7): 1551-60, 2013 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-23280522

RESUMEN

Prostate smooth muscle cells (pSMCs) are capable of responding to inflammatory stimuli by secreting proinflammatory products, which causes pSMCs to undergo dedifferentiation. Although it has been proposed that androgens decrease proinflammatory molecules in many cells and under various conditions, the role of testosterone in the prostate inflammatory microenvironment is still unclear. Therefore, our aim was to evaluate if testosterone was able to modulate the pSMCs response to bacterial LPS by stimulating primary pSMC cultures, containing testosterone or vehicle, with LPS (1 or 10 µg/ml) for 24-48 h. The LPS challenge induced pSMCs dedifferentiation as evidenced by a decrease of calponin and alpha smooth muscle actin along with an increase of vimentin in a dose-dependent manner, whereas testosterone abrogated these alterations. Additionally, an ultrastructural analysis showed that pSMCs acquired a secretory profile after LPS and developed proteinopoietic organelles, while pSMCs preincubated with testosterone maintained a more differentiated phenotype. Testosterone downregulated the expression of surface TLR4 in control cells and inhibited any increase after LPS treatment. Moreover, testosterone prevented IκB-α degradation and the LPS-induced NF-κB nuclear translocation. Testosterone also decreased TNF-α and IL6 production by pSMCs after LPS as quantified by ELISA. Finally, we observed that testosterone inhibited the induction of pSMCs proliferation incited by LPS. Taken together, these results indicate that testosterone reduced the proinflammatory pSMCs response to LPS, with these cells being less reactive in the presence of androgens. In this context, testosterone might have a homeostatic role by contributing to preserve a contractile phenotype on pSMCs under inflammatory conditions.


Asunto(s)
Miocitos del Músculo Liso/citología , Miocitos del Músculo Liso/metabolismo , Próstata/citología , Próstata/metabolismo , Testosterona/metabolismo , Receptor Toll-Like 4/metabolismo , Animales , Diferenciación Celular/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Citocinas/biosíntesis , Mediadores de Inflamación/metabolismo , Lipopolisacáridos/farmacología , Masculino , Modelos Biológicos , Miocitos del Músculo Liso/efectos de los fármacos , FN-kappa B/metabolismo , Fenotipo , Próstata/efectos de los fármacos , Ratas , Ratas Wistar , Testosterona/farmacología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA