Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 33
Filtrar
1.
J Neurochem ; 157(4): 1182-1195, 2021 05.
Artículo en Inglés | MEDLINE | ID: mdl-33030215

RESUMEN

The Nucleus Basalis of Meynert (NBM) is the main source of cholinergic neurons in the basal forebrain to be crucially involved in cognitive functions and whose degeneration correlates with cognitive decline in major degenerative pathologies as Alzheimer's and Parkinson's diseases. However, knowledge concerning NBM neurons derived from human brain is very limited to date. We recently characterized a primary culture of proliferating neuroblasts isolated from the human fetal NBM (hfNBM) as immature cholinergic neurons expressing the machinery to synthetize and release acetylcholine. Here we studied in detail electrophysiological features and cholinergic effects in this cell culture by patch-clamp recordings. Our data demonstrate that atropine-blocked muscarinic receptor activation by acetylcholine or carbachol enhanced IK and reduced INa currents by stimulating Gi -coupled M2 or phospholipase C-coupled M3 receptors, respectively. Inhibition of acetylcholine esterase activity by neostigmine unveiled a spontaneous acetylcholine release from hfNBM neuroblasts that might account for an autocrine/paracrine signaling during human brain development. Present data provide the first description of cholinergic effects in human NBM neurons and point to a role of acetylcholine as an autocrine/paracrine modulator of voltage-dependent channels. Our research could be of relevance in understanding the mechanisms of cholinergic system development and functions in the human brain, either in health or disease.


Asunto(s)
Acetilcolina/metabolismo , Potenciales de Acción/fisiología , Prosencéfalo Basal/metabolismo , Neuronas Colinérgicas/metabolismo , Células-Madre Neurales/metabolismo , Núcleo Basal de Meynert/metabolismo , Células Cultivadas , Feto , Humanos , Transducción de Señal/fisiología
2.
J Neuroendocrinol ; 32(3): e12837, 2020 03.
Artículo en Inglés | MEDLINE | ID: mdl-32077170

RESUMEN

It has been well established, particularly in animal models, that oestrogens exert neuroprotective effects in brain areas linked to cognitive processes. A key protective role could reside in the capacity of oestrogen to modulate the inflammatory response. However, the direct neuroprotective actions of oestrogens on neurones are complex and remain to be fully clarified. In the present study, we took advantage of a previously characterised primary culture of human cholinergic neurones (hfNBM) from the foetal nucleus basalis of Meynert, which is known to regulate hippocampal and neocortical learning and memory circuits, aiming to investigate the direct effects of oestrogens under inflammatory conditions. Exposure of cells to tumour necrosis factor (TNF)α (10 ng mL-1 ) determined the activation of an inflammatory response, as demonstrated by nuclear factor-kappa B p65 nuclear translocation and cyclooxygenase-2 mRNA expression. These effects were inhibited by treatment with either 17ß-oestradiol (E2 ) (10 nmol L-1 ) or G1 (100 nmol L-1 ), the selective agonist of the G protein-coupled oestrogen receptor (GPER1). Interestingly, the GPER1 antagonist G15 abolished the effects of E2 in TNFα-treated cells, whereas the ERα/ERß inhibitor tamoxifen did not. Electrophysiological measurements in hfNBMs revealed a depolarising effect caused by E2 that was specifically blocked by tamoxifen and not by G15. Conversely, G1 specifically hyperpolarised the cell membrane and also increased both inward and outward currents elicited by a depolarising stimulus, suggesting a modulatory action on hfNBM excitability by GPER1 activation. Interestingly, pretreating cells with TNFα completely blocked the effects of G1 on membrane properties and also significantly reduced GPER1 mRNA expression. In addition, we found a peculiar subcellular localisation of GPER1 to focal adhesion sites that implicates new possible mechanisms of action of GPER1 in the neuronal perception of mechanical stimuli. The results obtained in the present study indicate a modulatory functional role of GPER1 with respect to mediating the oestrogen neuroprotective effect against inflammation in brain cholinergic neurones and, accordingly, may help to identify protective strategies for preventing cognitive impairments.


Asunto(s)
Antiinflamatorios/farmacología , Núcleo Basal de Meynert/efectos de los fármacos , Neuronas Colinérgicas/efectos de los fármacos , Ciclopentanos/farmacología , Estradiol/farmacología , Estrógenos/farmacología , Quinolinas/farmacología , Receptores de Estrógenos/metabolismo , Receptores Acoplados a Proteínas G/metabolismo , Núcleo Basal de Meynert/metabolismo , Neuronas Colinérgicas/metabolismo , Humanos , Inflamación/inducido químicamente , Inflamación/metabolismo , Transducción de Señal/efectos de los fármacos , Factor de Necrosis Tumoral alfa
4.
Front Cell Neurosci ; 11: 339, 2017.
Artículo en Inglés | MEDLINE | ID: mdl-29163051

RESUMEN

The degeneration of cholinergic neurons of the nucleus basalis of Meynert (NBM) in the basal forebrain (BF) is associated to the cognitive decline of Alzheimer's disease (AD) patients. To date no resolutive therapies exist. Cell-based replacement therapy is a strategy currently under consideration, although the mechanisms underlying the generation of stem cell-derived NBM cholinergic neurons able of functional integration remain to be clarified. Since fetal brain is an optimal source of neuronal cells committed towards a specific phenotype, this study is aimed at isolating cholinergic neurons from the human fetal NBM (hfNBMs) in order to study their phenotypic, maturational and functional properties. Extensive characterization confirmed the cholinergic identity of hfNBMs, including positivity for specific markers (such as choline acetyltransferase) and acetylcholine (Ach) release. Electrophysiological measurements provided the functional validation of hfNBM cells, which exhibited the activation of peculiar sodium (INa) and potassium (IK) currents, as well as the presence of functional cholinergic receptors. Accordingly, hfNBMs express both nicotinic and muscarinic receptors, which were activated by Ach. The hfNBMs cholinergic phenotype was regulated by the nerve growth factor (NGF), through the activation of the high-affinity NGF receptor TrkA, as well as by 17-ß-estradiol through a peculiar recruitment of its own receptors. When intravenously administered in NBM-lesioned rats, hfNBMs determined a significant improvement in memory functions. Histological examination of brain sections showed that hfNBMs (labeled with PKH26 fluorescent dye prior to administration) reached the damaged brain areas. The study provides a useful model to study the ontogenetic mechanisms regulating the development and maintenance of the human brain cholinergic system and to assess new lines of research, including disease modeling, drug discovery and cell-based therapy for AD.

5.
J Clin Endocrinol Metab ; 102(1): 46-56, 2017 01 01.
Artículo en Inglés | MEDLINE | ID: mdl-27736314

RESUMEN

Context: Inflammatory pathways may impair central regulatory networks involving gonadotropin-releasing hormone (GnRH) neuron activity. Studies in humans are limited by the lack of human GnRH neuron cell lines. Objective: To establish an in vitro model of human GnRH neurons and analyze the effects of proinflammatory cytokines. Design: The primary human fetal hypothalamic cells (hfHypo) were isolated from 12-week-old fetuses. Responsiveness to kisspeptin, the main GnRH neurons' physiological regulator, was evaluated for biological characterization. Tumor necrosis factor alpha (TNF-α) was used as a proinflammatory stimulus. Main Outcome Measures: Expression of specific GnRH neuron markers by quantitative reverse transcription-polymerase chain reaction, flow cytometry, and immunocytochemistry analyses; and GnRH-releasing ability and electrophysiological changes in response to kisspeptin. Results: The primary hfHypo showed a high percentage of GnRH-positive cells (80%), expressing a functional kisspeptin receptor (KISS1R) and able to release GnRH in response to kisspeptin. TNF-α exposure determined a specific inflammatory intracellular signaling and reduced GnRH secretion, KISS1R expression, and kisspeptin-induced depolarizing effect. Moreover, hfHypo possessed a primary cilium, whose assembly was inhibited by TNF-α. Conclusion: The hfHypo cells represent a novel tool for investigations on human GnRH neuron biology. TNF-α directly affects GnRH neuron function by interfering with KISS1R expression and ciliogenesis, thereby impairing kisspeptin signaling.


Asunto(s)
Feto/metabolismo , Regulación de la Expresión Génica/efectos de los fármacos , Hormona Liberadora de Gonadotropina/metabolismo , Kisspeptinas/metabolismo , Neuronas/metabolismo , Receptores Acoplados a Proteínas G/metabolismo , Factor de Necrosis Tumoral alfa/farmacología , Células Cultivadas , Femenino , Feto/citología , Feto/efectos de los fármacos , Humanos , Kisspeptinas/genética , Masculino , Neuronas/citología , Neuronas/efectos de los fármacos , Receptores Acoplados a Proteínas G/genética , Receptores de Kisspeptina-1 , Transducción de Señal/efectos de los fármacos
6.
Mol Cell Neurosci ; 75: 50-62, 2016 09.
Artículo en Inglés | MEDLINE | ID: mdl-27370937

RESUMEN

Over the past decades, studies in both Huntington's disease animal models and pilot clinical trials have demonstrated that replacement of degenerated striatum and repair of circuitries by grafting fetal striatal primordium is feasible, safe and may counteract disease progression. However, a better comprehension of striatal ontogenesis is required to assess the fetal graft regenerative potential. During neuronal development, neurotrophins exert pleiotropic actions in regulating cell fate and synaptic plasticity. In this regard, brain-derived neurotrophic factor (BDNF) and fibroblast growth factor 2 (FGF2) are crucially implicated in the control of fate choice of striatal progenitor cells. In this study, we intended to refine the functional features of human striatal precursor (HSP) cells isolated from ganglionic eminence of 9-12week old human fetuses, by studying with electrophysiological methods the effect of BDNF and FGF2 on the membrane biophysical properties and the voltage-dependent Ca(2+) currents. These features are particularly relevant to evaluate neuronal cell functioning and can be considered reliable markers of the developmental phenotype of human striatal primordium. Our results have demonstrated that BDNF and FGF2 induced membrane hyperpolarization, increased the membrane capacitance and reduced the resting total and specific conductance values, suggesting a more efficient control of resting ionic fluxes. Moreover, the treatment with both neurotrophins enhanced N-type Ca(2+) current amplitude and reduced L- and T-type ones. Overall, our data indicate that BDNF and FGF2 may help HSP cells to attain a more functionally mature phenotype.


Asunto(s)
Potenciales de Acción , Factor Neurotrófico Derivado del Encéfalo/farmacología , Canales de Calcio/metabolismo , Cuerpo Estriado/fisiología , Factor 2 de Crecimiento de Fibroblastos/farmacología , Células-Madre Neurales/fisiología , Neurogénesis , Células Cultivadas , Cuerpo Estriado/citología , Cuerpo Estriado/embriología , Humanos , Células-Madre Neurales/efectos de los fármacos
7.
Inflammation ; 39(3): 1238-52, 2016 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-27165639

RESUMEN

T helper 1 (Th1) type cytokines and chemokines are bioactive mediators in inflammation underling several diseases and co-morbid conditions, such as cardiovascular and metabolic disorders. Th1 chemokine CXCL10 participates in heart damage initiation/progression; cardioprotection has been recently associated with sildenafil, a type 5 phosphodiesterase inhibitor. We aimed to evaluate the effect of sildenafil on CXCL10 in inflammatory conditions associated with diabetic cardiomyopathy. We analyzed: CXCL10 gene and protein in human cardiac, endothelial, and immune cells challenged by pro-inflammatory stimuli with and without sildenafil; serum CXCL10 in diabetic subjects at cardiomyopathy onset, before and after 3 months of treatment with sildenafil vs. placebo. Sildenafil significantly decreased CXCL10 protein secretion (IC50 = 2.6 × 10(-7)) and gene expression in human cardiomyocytes and significantly decreased circulating CXCL10 in subjects with chemokine basal level ≥ 930 pg/ml, the cut-off value as assessed by ROC analysis. In conclusion, sildenafil could be a pharmacologic tool to control CXCL10-associated inflammation in diabetic cardiomyopathy.


Asunto(s)
Quimiocina CXCL10/efectos de los fármacos , Cardiomiopatías Diabéticas/patología , Miocitos Cardíacos/metabolismo , Inhibidores de Fosfodiesterasa 5/farmacología , Citrato de Sildenafil/farmacología , Células Cultivadas , Quimiocina CXCL10/análisis , Quimiocina CXCL10/genética , Cardiomiopatías Diabéticas/tratamiento farmacológico , Expresión Génica/efectos de los fármacos , Humanos , Inflamación/tratamiento farmacológico
9.
Cell Transplant ; 24(5): 811-7, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-24380491

RESUMEN

Fetal grafting in a human diseased brain was thought to be less immunogenic than other solid organ transplants, hence the minor impact on the efficacy of the transplant. How much prophylactic immune protection is required for neural allotransplantation is also debated. High-sensitive anti-HLA antibody screening in this field has never been reported. Sixteen patients with Huntington's disease underwent human fetal striatal transplantation in the frame of an open-label observational trial, which is being carried out at Florence University. All patients had both brain hemispheres grafted in two separate robotic-stereotactic procedures. The trial started in February 2006 with the first graft to the first patient (R1). R16 was given his second graft on March 2011. All patients received triple immunosuppressive treatment. Pre- and posttransplant sera were analyzed for the presence of anti-HLA antibodies using the multiplexed microsphere-based suspension array Luminex xMAP technology. Median follow-up was 38.5 months (range 13-85). Six patients developed anti-HLA antibodies, which turned out to be donor specific. Alloimmunization occurred in a time window of 0-49 months after the first neurosurgical procedure. The immunogenic determinants were non-self-epitopes from mismatched HLA antigens. These determinants were both public epitopes shared by two or more HLA molecules and private epitopes unique to individual HLA molecules. One patient had non-donor-specific anti-HLA antibodies in her pretransplant serum sample, possibly due to previous sensitization events. Although the clinical significance of donor-specific antibodies is far from being established, particularly in the setting of neuronal transplantation, these findings underline the need of careful pre- and posttransplant immunogenetic evaluation of patients with intracerebral grafts.


Asunto(s)
Cuerpo Estriado/trasplante , Antígenos HLA , Enfermedad de Huntington/sangre , Enfermedad de Huntington/cirugía , Isoanticuerpos/sangre , Aloinjertos , Femenino , Feto , Humanos , Masculino , Factores de Tiempo
10.
Stereotact Funct Neurosurg ; 92(4): 211-7, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-25096235

RESUMEN

BACKGROUND: Restoration of functions in Huntington's disease (HD) by neurotransplantation stems from the formation of a striatum-like structure capable of establishing host connections as a result of grafted striatal neuroblast maturation. For the first time, we demonstrated some developmental steps accomplished by progenitor cells in the brain of an HD patient and analysed the molecular asset of the human primordium. CASE REPORT: Surgery involved bilateral (two sessions) stereotactic, caudate-putaminal transplantation of whole ganglionic eminence fragments from single legally aborted fetuses. MRI showed that the tissue deposits of the left hemisphere grew and joined to constitute a single tissue mass that remodelled basal ganglia anatomy and remained stable in size over time. No evidence of graft growth was observed contralaterally. PET demonstrated increased striatal and stable cortical metabolism. Unified Huntington's Disease Rating Scale assessments demonstrated improvement of motor performances, which faded over the 36-month follow-up. Cognitive performance tended to decrease at a lower rate than before transplantation. CONCLUSION: The striatal primordium grew into the host brain and this process was associated with metabolic change and some clinical benefit. The study suggests the plasticity and reparative potential of un-manipulated primordium in an era where promising cell-based therapies are still in their infancy.


Asunto(s)
Trasplante de Tejido Encefálico , Cuerpo Estriado/patología , Trasplante de Tejido Fetal , Enfermedad de Huntington/cirugía , Plasticidad Neuronal , Telencéfalo/trasplante , Adulto , Trasplante de Tejido Encefálico/métodos , Fármacos del Sistema Nervioso Central/uso terapéutico , Trastornos del Conocimiento/etiología , Terapia Combinada , Cuerpo Estriado/diagnóstico por imagen , Trasplante de Tejido Fetal/métodos , Estudios de Seguimiento , Perfilación de la Expresión Génica , Supervivencia de Injerto , Humanos , Enfermedad de Huntington/tratamiento farmacológico , Enfermedad de Huntington/patología , Enfermedad de Huntington/psicología , Italia , Imagen por Resonancia Magnética , Masculino , Neuroimagen , Tomografía de Emisión de Positrones , Robótica , Índice de Severidad de la Enfermedad , Técnicas Estereotáxicas , Telencéfalo/embriología , Telencéfalo/metabolismo
11.
Exp Neurol ; 257: 130-47, 2014 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-24792640

RESUMEN

Grafting fetal striatal cells into the brain of Huntington's disease (HD) patients has raised certain expectations in the past decade as an effective cell-based-therapy for this devastating condition. We argue that the first requirement for successful transplantation is defining the window of plasticity for the striatum during development when the progenitor cells, isolated from their environment, are able to maintain regional-specific-identity and to respond appropriately to cues. The primary cell culture from human fetal striatal primordium described here consists of a mixed population of neural stem cells, neuronal-restricted progenitors and striatal neurons. These cells express trophic factors, such as BDNF and FGF2. We show that these neurotrophins maintain cell plasticity, inducing the expression of neuronal precursor markers and cell adhesion molecules, as well as promoting neurogenesis, migration and survival. We propose that BDNF and FGF2 play an important autocrine-paracrine role during early striatum development in vivo and that their release by fetal striatal grafts may be relevant in the setting of HD cell therapy.


Asunto(s)
Factor Neurotrófico Derivado del Encéfalo/farmacología , Moléculas de Adhesión Celular/metabolismo , Cuerpo Estriado/citología , Factor 2 de Crecimiento de Fibroblastos/farmacología , Células-Madre Neurales/efectos de los fármacos , Antígenos/genética , Antígenos/metabolismo , Antígenos CD/genética , Antígenos CD/metabolismo , Factor Neurotrófico Derivado del Encéfalo/metabolismo , Moléculas de Adhesión Celular/genética , Diferenciación Celular/efectos de los fármacos , Movimiento Celular/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Células Cultivadas , Feto , Factor 2 de Crecimiento de Fibroblastos/metabolismo , Regulación de la Expresión Génica/efectos de los fármacos , Humanos , Proteínas del Tejido Nervioso/genética , Proteínas del Tejido Nervioso/metabolismo , Neuritas/efectos de los fármacos , Neuroglía/efectos de los fármacos , Neuronas/efectos de los fármacos , Antígenos O/genética , Antígenos O/metabolismo , Proteoglicanos/genética , Proteoglicanos/metabolismo
12.
Mol Cell Endocrinol ; 382(1): 107-119, 2014 Jan 25.
Artículo en Inglés | MEDLINE | ID: mdl-24064031

RESUMEN

Rabbits with high fat diet (HFD)-induced metabolic syndrome (MetS) developed hypogonadotropic hypogonadism (HH) and showed a reduced gonadotropin-releasing hormone (GnRH) immunopositivity in the hypothalamus. This study investigated the relationship between MetS and hypothalamic alterations in HFD-rabbits. Gonadotropin levels decreased as a function of MetS severity, hypothalamic gene expression of glucose transporter 4 (GLUT4) and interleukin-6 (IL-6). HFD determined a low-grade inflammation in the hypothalamus, significantly inducing microglial activation, expression and immunopositivity of IL-6, as well as GLUT4 and reduced immunopositivity for KISS1 receptor, whose mRNA expression was negatively correlated to glucose intolerance. Correcting glucose metabolism with obetcholic acid improved hypothalamic alterations, reducing GLUT4 and IL-6 immunopositivity and significantly increasing GnRH mRNA, without, however, preventing HFD-related HH. No significant effects at the hypothalamic level were observed after systemic anti-inflammatory treatment (infliximab). Our results suggest that HFD-induced metabolic derangements negatively affect GnRH neuron function through an inflammatory injury at the hypothalamic level.


Asunto(s)
Hormona Liberadora de Gonadotropina/metabolismo , Inflamación/etiología , Síndrome Metabólico/complicaciones , Neuronas/patología , Área Preóptica/metabolismo , Área Preóptica/patología , Animales , Anticuerpos Monoclonales/farmacología , Biomarcadores/metabolismo , Ácido Quenodesoxicólico/análogos & derivados , Ácido Quenodesoxicólico/farmacología , Dieta Alta en Grasa , Regulación de la Expresión Génica/efectos de los fármacos , Transportador de Glucosa de Tipo 4/genética , Transportador de Glucosa de Tipo 4/metabolismo , Hormona Liberadora de Gonadotropina/sangre , Hormona Liberadora de Gonadotropina/genética , Inmunohistoquímica , Inflamación/sangre , Infliximab , Masculino , Síndrome Metabólico/sangre , Neuronas/efectos de los fármacos , Neuronas/metabolismo , Área Preóptica/efectos de los fármacos , ARN Mensajero/genética , ARN Mensajero/metabolismo , Conejos , Receptores Acoplados a Proteínas G/metabolismo
13.
Prostate ; 73(4): 428-41, 2013 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-22996758

RESUMEN

BACKGROUND: Phosphodiesterase type 5 (PDE5) inhibitors improve benign prostatic hyperplasia (BPH)-related lower urinary tract symptoms (LUTS), often associated with metabolic syndrome (MetS). This study investigated the effects of PDE5 inhibition in the prostate of rabbits fed a high fat diet (HFD) for 12 weeks. HFD-rabbits develop the most important features of human MetS (glucose intolerance, dyslipidemia, increased abdominal adiposity, and hypertension), along with hypogonadism and LUT abnormalities (prostate and bladder inflammation/tissue remodeling). METHODS: Gene expression was evaluated by quantitative RT-PCR. Prostate morphological changes and oxygenation were evaluated by immunohistochemistry. RESULTS: HFD prostates showed increased PDE5 expression, suggesting a peculiar sensitivity of prostate to the action of PDE5 inhibitors during MetS. Accordingly, prostate PDE5 mRNA was negatively associated to plasma testosterone/estradiol ratio, whose reduction characterizes MetS, and positively with the expression in prostate of several genes exploring pathogenetic processes for BPH/LUTS, such as inflammation, leukocyte infiltration, and fibrosis/myofibroblast activation. Most of these genes was up-regulated by HFD, and significantly reduced by PDE5 inhibition, through either chronic (12 weeks) or, at a lower extent, acute (1-week) tadalafil dosing. Tadalafil was also able to reduce blood pressure and visceral fat in HFD rabbits, without changing any other MetS parameter. Interestingly, 1-week tadalafil administration to HFD rabbits, significantly blunted prostate inflammation (increased CD45 immunopositivity), fibrosis (reduced muscle/fiber ratio) and hypo-oxygenation, thus suggesting a potential curative effect of PDE5 inhibition on MetS-related prostate alterations. CONCLUSIONS: Our data provide the experimental evidences to support the multiple potentiality of PDE5 inhibitors as a useful therapeutic tool in LUTS.


Asunto(s)
Fosfodiesterasas de Nucleótidos Cíclicos Tipo 5/metabolismo , Modelos Animales de Enfermedad , Síndrome Metabólico/tratamiento farmacológico , Síndrome Metabólico/enzimología , Inhibidores de Fosfodiesterasa 5/farmacología , Próstata/efectos de los fármacos , Próstata/enzimología , Animales , Dieta Alta en Grasa/efectos adversos , Masculino , Síndrome Metabólico/patología , Inhibidores de Fosfodiesterasa 5/uso terapéutico , Próstata/patología , Hiperplasia Prostática/tratamiento farmacológico , Hiperplasia Prostática/enzimología , Hiperplasia Prostática/patología , Conejos , Regulación hacia Arriba/efectos de los fármacos , Regulación hacia Arriba/fisiología
14.
Int J Endocrinol ; 2013: 684659, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-24489542

RESUMEN

Metabolic disorders are often associated with male hypogonadotropic hypogonadism, suggesting that hypothalamic defects involving GnRH neurons may impair the reproductive function. Among metabolic factors hyperglycemia has been implicated in the control of the reproductive axis at central level, both in humans and in animal models. To date, little is known about the direct effects of pathological high glucose concentrations on human GnRH neurons. In this study, we investigated the high glucose effects in the human GnRH-secreting FNC-B4 cells. Gene expression profiling by qRT-PCR, confirmed that FNC-B4 cells express GnRH and several genes relevant for GnRH neuron function (KISS1R, KISS1, sex steroid and leptin receptors, FGFR1, neuropilin 2, and semaphorins), along with glucose transporters (GLUT1, GLUT3, and GLUT4). High glucose exposure (22 mM; 40 mM) significantly reduced gene and protein expression of GnRH, KISS1R, KISS1, and leptin receptor, as compared to normal glucose (5 mM). Consistent with previous studies, leptin treatment significantly induced GnRH mRNA expression at 5 mM glucose, but not in the presence of high glucose concentrations. In conclusion, our findings demonstrate a deleterious direct contribution of high glucose on human GnRH neurons, thus providing new insights into pathogenic mechanisms linking metabolic disorders to reproductive dysfunctions.

15.
J Endocrinol ; 215(3): 347-62, 2012 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-23045189

RESUMEN

We recently demonstrated that testosterone dosing ameliorated the metabolic profile and reduced visceral adipose tissue (VAT) in a high-fat diet (HFD)-induced rabbit model of metabolic syndrome (MetS). We studied the effects of HFD and in vivo testosterone dosing on VAT function and the adipogenic capacity of rabbit preadipocytes isolated from VAT of regular diet (RD), HFD, and testosterone-treated HFD rabbits. VAT was studied by immunohistochemistry, western blot, and RT-PCR. Isolated rPADs were exposed to adipocyte differentiating mixture (DIM) to evaluate adipogenic potential. Adipocyte size was significantly increased in HFD VAT compared with RD, indicating adipocyte dysfunction, which was normalized by testosterone dosing. Accordingly, perilipin, an anti-lipolytic protein, was significantly increased in HFD VAT, when compared with other groups. HFD VAT was hypoxic, while testosterone dosing normalized VAT oxygenation. In VAT, androgen receptor expression was positively associated with mRNA expression of GLUT4 (SLC2A4) (insulin-regulated glucose transporter) and STAMP2 (STEAP4) (androgen-dependent gene required for insulin signaling). In testosterone-treated HFD VAT, STAMP2 mRNA was significantly increased when compared with the other groups. Moreover, GLUT4 membrane translocation was significantly reduced in HFD VAT, compared with RD, and increased by testosterone. In DIM-exposed preadipocytes from HFD, triglyceride accumulation, adipocyte-specific genes, insulin-stimulated triglyceride synthesis, glucose uptake, and GLUT4 membrane translocation were reduced compared with preadipocytes from RD and normalized by in vivo testosterone dosing. In conclusion, testosterone dosing in a MetS animal model positively affects VAT functions. This could reflect the ability of testosterone in restoring insulin sensitivity in VAT, thus counteracting metabolic alterations.


Asunto(s)
Tejido Adiposo/efectos de los fármacos , Tejido Adiposo/patología , Síndrome Metabólico/tratamiento farmacológico , Testosterona/uso terapéutico , Adipocitos/citología , Adipocitos/efectos de los fármacos , Tejido Adiposo/metabolismo , Animales , Diferenciación Celular/efectos de los fármacos , Diferenciación Celular/genética , Células Cultivadas , Citometría de Flujo , Inmunohistoquímica , Grasa Intraabdominal/efectos de los fármacos , Grasa Intraabdominal/metabolismo , Grasa Intraabdominal/patología , Masculino , Síndrome Metabólico/patología , Conejos , Receptores Androgénicos/metabolismo
16.
J Steroid Biochem Mol Biol ; 132(1-2): 80-92, 2012 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-22406511

RESUMEN

In the male, metabolic syndrome (MetS) is associated to an increased risk of benign prostatic hyperplasia (BPH) and lower urinary tract symptoms (LUTS). A recently established rabbit model of high fat diet (HFD)-induced MetS showed hypogonadism and the presence of prostate gland alterations, including inflammation, hypoxia and fibrosis. The present study investigated whether HFD-induced MetS might also alter bladder structure and function. Testosterone and the farnesoid X receptor (FXR) agonist INT-747, were evaluated for possible effects on HFD bladder. MetS rabbits develop bladder alterations, including fibrosis (reduced muscle/fiber ratio), hypoxia [2-fold increase as compared to regular diet (RD) group], low-grade inflammation (increased leukocyte infiltration and inflammatory markers) and RhoA/ROCK hyperactivity. Bladder strips from HFD rabbits, pre-contracted with carbachol, showed an overactive response to the selective ROCK inhibitor Y-27632. All these HFD-induced bladder alterations were partially blunted by testosterone and almost completely reverted by INT-747. Both treatments prevented some MetS features (glucose intolerance and visceral fat increase), thus suggesting that their effects on bladder could be ascribed to an improvement of the metabolic and/or hypogonadal state. However, a pathogenetic role for hypogonadism has been ruled out as GnRH analog-induced hypogonadal rabbits, fed a regular diet, did not show any detectable bladder alterations. In addition, INT-747 did not revert the MetS-induced hypogonadal state. FXR mRNA was highly expressed in rabbit bladder and positively associated with visceral fat increase. A direct effect of INT-747 on bladder smooth muscle was further suggested by inhibition of RhoA/ROCK-mediated activity by in vitro experiments on isolated cells. In conclusion, HFD-related MetS features are associated to bladder derangements, which are ameliorated by testosterone or INT-747 administration. INT-747 showed the most marked effects in counteracting MetS-related RhoA/ROCK overactivity, thus opening novel therapeutic opportunities for this drug.


Asunto(s)
Andrógenos/uso terapéutico , Ácido Quenodesoxicólico/análogos & derivados , Hipogonadismo/tratamiento farmacológico , Síndrome Metabólico/tratamiento farmacológico , Receptores Citoplasmáticos y Nucleares/agonistas , Testosterona/uso terapéutico , Animales , Glucemia/análisis , Movimiento Celular , Ácido Quenodesoxicólico/uso terapéutico , Colesterol/sangre , Dieta Alta en Grasa , Modelos Animales de Enfermedad , Hipogonadismo/inducido químicamente , Hipogonadismo/patología , Hipogonadismo/fisiopatología , Masculino , Síndrome Metabólico/etiología , Síndrome Metabólico/patología , Síndrome Metabólico/fisiopatología , Contracción Muscular/efectos de los fármacos , Miocitos del Músculo Liso/efectos de los fármacos , Miocitos del Músculo Liso/patología , Miocitos del Músculo Liso/fisiología , Próstata/efectos de los fármacos , Próstata/metabolismo , Próstata/patología , ARN Mensajero/metabolismo , Conejos , Receptores Androgénicos/genética , Receptores Citoplasmáticos y Nucleares/genética , Triglicéridos/sangre , Pamoato de Triptorelina , Vejiga Urinaria/efectos de los fármacos , Vejiga Urinaria/patología , Vejiga Urinaria/fisiología
17.
Eur J Cell Biol ; 91(2): 139-49, 2012 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-22176919

RESUMEN

Inflammatory myopathies (IMs) are systemic diseases characterized by a T helper (Th) 1 type inflammatory response and cell infiltrates within skeletal muscles. The mainstay of treatment is drugs aimed at suppressing the immune system - corticosteroids and immunosuppressants. About 25% of patients are non-responders. Skeletal muscle cells seem actively involved in the immune-inflammatory response and not only a target; understanding the molecular bases of IMs might help drug development strategies. Within muscles the interaction between the chemokine interferon (IFN)γ inducible 10 kDa protein, CXCL10 or IP-10, and its specific receptor CXCR3, present on Th1 type infiltrating cells, likely plays a pivotal role, potentially offering the opportunity for therapeutic intervention. We aimed to clarify the involvement of human skeletal muscle cells in inflammatory processes in terms of CXCL10 secretion, to elucidate the engaged molecular mechanism(s) and, finally, to evaluate muscular cell responses, if any, to some immunosuppressants routinely used in IM treatment, such as methylprednisolone, methotrexate, cyclosporin A and Infliximab. We first isolated and characterized human fetal skeletal muscle cells (Hfsmc), which expressed the specific lineage markers and showed the competence to react in the context of an in vitro alloresponse. CXCL10 protein secretion by Hfsmc was similarly induced by the inflammatory cytokines interferon (IFN)γ and tumor necrosis factor (TNF)α, above undetectable control levels, through the activation of Stat1 and NF-kB pathways, respectively; CXCL10 secretion was significantly magnified by cytokine combination, and this synergy was associated to a significant up-regulation of TNFαRII; cytokine-induced CXCL10 secretion was considerably affected only by Infliximab. Our data suggested that human skeletal muscle cells might actively self-promote muscular inflammation by eliciting CXCL10 secretion, which is known to amplify Th1 cell tissue infiltration in vivo. In conclusion, we sustain that pharmacological targeting of CXCL10 within muscular cells might contribute to keep in control pro-Th1 polarization of the immune/inflammatory response.


Asunto(s)
Quimiocina CXCL10/metabolismo , Células Musculares/metabolismo , Músculo Esquelético/metabolismo , Miositis/metabolismo , Receptores CXCR3/metabolismo , Anticuerpos Monoclonales/farmacología , Células Cultivadas , Quimiocina CXCL10/inmunología , Ciclosporina/farmacología , Feto , Humanos , Inmunosupresores/farmacología , Infliximab , Interferón gamma/farmacología , Activación de Linfocitos , Metilprednisolona/farmacología , Células Musculares/efectos de los fármacos , Células Musculares/inmunología , Músculo Esquelético/efectos de los fármacos , Músculo Esquelético/inmunología , Miositis/inmunología , FN-kappa B/metabolismo , Receptores CXCR3/inmunología , Factor de Transcripción STAT1/metabolismo , Transducción de Señal , Células TH1/inmunología , Factor de Necrosis Tumoral alfa/farmacología
18.
Biometals ; 25(1): 63-74, 2012 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-21796402

RESUMEN

The effects of cadmium on the central nervous system are still relatively poorly understood and its role in neurodegenerative diseases has been debated. In our research, cultured explants from 25 human foetal spinal cords (10-11 weeks gestational age) were incubated with 10 and 100 µM cadmium chloride (CdCl(2)) for 24 h. After treatment, an immunohistochemical study [for Sglial fibrillary acidic protein (GFAP) and choline acetyltransferase (ChAT)], a Western blot analysis (for GFAP, ß-Tubulin III, nerve growth factor receptor, Caspase 8 and poly (ADP-ribose) polymerase), and a terminal deoxynucleotidyl transferase biotin-dUTP nick end labelling (TUNEL) assay (for detection of apoptotic bodies) were performed. The treatment with CdCl(2) induced a significant and dose-dependent change in the ratio motor neurons/glial cells in the ventral horns of human foetal spinal cord. The decrease of the choline acetyltransferase-positive cells (motor neurons) and the reduction of ß Tubulin III indicate that CdCl(2) specifically affects motor neurons of the ventral horns. While the number of motor neurons decreased for the activation of apoptotic pathways (as shown by the increased expression of Caspase 8, nerve growth factor receptor, and poly (ADP-ribose) polymerase), glial cells, both in the subependymal zone and in the gray matter of the ventral horns, increased (as shown by the increase of GFAP expression). These results provide the evidence that during human spinal cord development, CdCl(2) may affect the fate of neural and glial cells thus, being potentially involved in the etiopathogenesis of neurodegenerative diseases.


Asunto(s)
Cloruro de Cadmio/farmacología , Morfogénesis/efectos de los fármacos , Morfogénesis/fisiología , Médula Espinal/efectos de los fármacos , Médula Espinal/embriología , Médula Espinal/crecimiento & desarrollo , Animales , Niño , Femenino , Feto/anatomía & histología , Feto/fisiología , Edad Gestacional , Humanos , Etiquetado Corte-Fin in Situ
19.
J Endocrinol ; 212(1): 71-84, 2012 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-22010203

RESUMEN

Metabolic syndrome (MetS) and benign prostatic hyperplasia (BPH)/lower urinary tract symptoms (LUTS) are often associated. One of their common denominators is hypogonadism. However, testosterone supplementation is limited by concerns for potential prostatic side effects. The objective was to determine whether MetS-associated prostate alterations are prevented by testosterone supplementation. We used a previously described animal model of MetS, obtained by feeding male rabbits a high-fat diet (HFD) for 12 weeks. Subsets of HFD rabbits were treated with testosterone or with the farnesoid X receptor agonist INT-747. Rabbits fed a standard diet were used as controls. HFD-animals develop hypogonadism and all the MetS features: hyperglycemia, glucose intolerance, dyslipidemia, hypertension, and visceral obesity. In addition, HFD-animals show a prostate inflammation. Immunohistochemical analysis demonstrated that HFD-induced prostate fibrosis, hypoxia, and inflammation. The mRNA expression of several proinflammatory (IL8, IL6, IL1ß, and TNFα), T lymphocyte (CD4, CD8, Tbet, Gata3, and ROR γt), macrophage (TLR2, TLR4, and STAMP2), neutrophil (lactoferrin), inflammation (COX2 and RAGE), and fibrosis/myofibroblast activation (TGFß, SM22α, αSMA, RhoA, and ROCK1/ROCK2) markers was significantly increased in HFD prostate. Testosterone, as well as INT-747, treatment prevented some MetS features, although only testosterone normalized all the HFD-induced prostate alterations. Interestingly, the ratio between testosterone and estradiol plasma level retains a significant, negative, association with all the fibrosis and the majority of inflammatory markers analyzed. These data highlight that testosterone protects rabbit prostate from MetS-induced prostatic hypoxia, fibrosis, and inflammation, which can play a role toward the development/progression of BPH/LUTS.


Asunto(s)
Andrógenos/uso terapéutico , Ácido Quenodesoxicólico/análogos & derivados , Síndrome Metabólico/complicaciones , Prostatitis/prevención & control , Testosterona/uso terapéutico , Animales , Biomarcadores/metabolismo , Ácido Quenodesoxicólico/uso terapéutico , Grasas de la Dieta/efectos adversos , Modelos Animales de Enfermedad , Evaluación Preclínica de Medicamentos , Estradiol/sangre , Fibrosis/metabolismo , Masculino , Próstata/metabolismo , Próstata/patología , Prostatitis/etiología , Prostatitis/metabolismo , Prostatitis/patología , ARN Mensajero/metabolismo , Conejos , Receptores Citoplasmáticos y Nucleares/agonistas , Receptores Citoplasmáticos y Nucleares/metabolismo , Receptores de Esteroides/metabolismo , Testosterona/sangre
20.
J Sex Med ; 8(10): 2746-60, 2011 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-21812935

RESUMEN

INTRODUCTION: In humans, prostate phosphodiesterase type 5 inhibitors (PDE5) expression was prominently localized in the endothelial and smooth muscle cells of the vascular bed, suggesting a possible action of PDE5 inhibitors (PDE5i) on prostate blood flow. AIM: To investigate PDE5 expression in human and rat lower urinary tract (LUT) tissues, including vasculature, and determine the effects of PDE5 inhibition with tadalafil on prostatic blood perfusion. MAIN OUTCOME MEASURES: Human vesicular-deferential arteries (which originate from the inferior vesical artery, the main arterial source of blood supply to the bladder and prostate) were analyzed for PDE5 expression and activity. The effects of tadalafil on prostate oxygenation were studied in spontaneously hypertensive rats (SHR), characterized by ischemia/hypoxia of the genitourinary tract. METHODS: PDE5 expression was evaluated by quantitative reverse transcription-polymerase chain reaction and immunohistochemistry. SHR were treated with tadalafil (2 mg/kg/day) for 1, 7, or 28 days and compared with untreated SHR and the unaffected counterpart Wistar-Kyoto (WKY) rats. Prostate oxygenation was detected by Hypoxyprobe-1 and hypoxia markers (hypoxia-inducible factor-1α[HIF-1α] and endothelin-1 type B [ETB]) immunostaining. RESULTS: Human vesicular-deferential artery expressed high levels of PDE5, similar to corpora cavernosa, immunolocalized in the endothelial and smooth muscle layer. In these arteries, tadalafil inhibited cyclic guanosine monophosphate breakdown (half maximal inhibitory concentration (IC(50) ) in the low nanomolar range, as in corpora cavernosa) and increased the relaxant response to sodium nitroprusside. SHR prostate resulted markedly hypoxic (hypoxyprobe immunopositivity) and positive for HIF-1α and ETB, while tadalafil treatment restored oxygenation to WKY level at each time point. The mRNA expression of the HIF-1α target gene, BCL2/adenovirus E1B 19 kDa interacting protein 3, was significantly increased in SHR prostate and partially restored to WKY level by tadalafil. CONCLUSION: Human vesicular-deferential artery is characterized by a high expression and activity of PDE5, which was inhibited by tadalafil in vitro. In SHR, tadalafil increases prostate tissue oxygenation, thus suggesting a possible mechanism through which PDE5i exert beneficial effects on LUT symptoms.


Asunto(s)
Carbolinas/farmacología , Fosfodiesterasas de Nucleótidos Cíclicos Tipo 5/metabolismo , Inhibidores de Fosfodiesterasa 5/farmacología , Próstata/efectos de los fármacos , Sistema Urinario/enzimología , Animales , Endotelina-1/metabolismo , Humanos , Subunidad alfa del Factor 1 Inducible por Hipoxia/metabolismo , Masculino , Músculo Liso Vascular/efectos de los fármacos , Músculo Liso Vascular/enzimología , Músculo Liso Vascular/metabolismo , Nitroprusiato/farmacología , Oxígeno/metabolismo , Próstata/metabolismo , Ratas , Ratas Endogámicas SHR , Ratas Endogámicas WKY , Reacción en Cadena en Tiempo Real de la Polimerasa , Tadalafilo , Vejiga Urinaria/irrigación sanguínea , Vejiga Urinaria/enzimología , Vejiga Urinaria/metabolismo , Sistema Urinario/irrigación sanguínea , Sistema Urinario/efectos de los fármacos , Sistema Urinario/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...