Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 16 de 16
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
F1000Res ; 11: 1086, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-36176547

RESUMEN

The COVID-19 pandemic profoundly changed how scientific conferences are organized, fostering virtual meetings. These online events have allowed students and researchers to overcome geographical, administrative and economic barriers to attend and organize high-quality international symposiums. Moreover, these virtual conferences have contributed to the creation of inclusive activities that strengthen scientific communities. Here, we summarize the main activities and learnings from the 1st Ecuadorian-Venezuelan Symposium of Young Bioinformatics Researchers (1SEVJIB), organized by the Ecuadorian and Venezuelan ISCB-SC Regional Student Groups (RSGs). This symposium aimed to provide an opportunity for undergraduate and postgraduate students from Ecuador, Venezuela, and other Latin American countries to share their Bioinformatics research. The 1SEVJIB was the first bi-national conference organized by two RSGs from Latin America (LatAm). This symposium was a two-day virtual meeting with five activities: 1) oral student presentations, 2) poster session, 3) keynote lectures, 4) workshop, and 5) round table. This conference promoted the scientific exchange and cooperation networks between young Bioinformatics researchers and students from Ecuador, Venezuela, and LatAm, giving them opportunities to boost their scientific careers.


Asunto(s)
Biología Computacional , Congresos como Asunto , COVID-19 , Biología Computacional/educación , Ecuador , Humanos , Pandemias , Investigadores , Sociedades Científicas , Estudiantes , Venezuela
2.
Am J Physiol Renal Physiol ; 317(4): F874-F880, 2019 10 01.
Artículo en Inglés | MEDLINE | ID: mdl-31390230

RESUMEN

Parathyroid hormone-related protein (PTHrP) and its receptor are abundantly expressed throughout the renal parenchyma, where PTHrP exerts a modulatory action on renal function. PTHrP upregulation is a common event associated with the mechanism of renal injury and repair. However, no study has yet explored the putative excretion of PTHrP in urine, including its potential relationship with renal function. In the present study, we tested this hypothesis by studying the well-known rat model of acute renal injury induced by the chemotherapeutic agent cisplatin. Using Western blot analysis, we could detect a single protein band, corresponding to intact PTHrP, in the urine of both control and cisplatin-injected rats, whose levels were significantly higher in the latter group. PTHrP was detected in rat urine by dot blot, and its quantification with two specific ELISA kits showed that, compared with control rats, those treated with cisplatin displayed a significant increase in urinary PTHrP (expressed as the PTHrP-to-creatinine ratio or 24-h excretion). In addition, a positive correlation between urinary PTHrP excretion and serum creatinine was found in these animals. In conclusion, our data demonstrate that PTHrP is excreted in rat urine and that this excretion is higher with the decrease of renal function. This suggests that urinary PTHrP levels might be a renal function marker.


Asunto(s)
Lesión Renal Aguda/inducido químicamente , Lesión Renal Aguda/orina , Antineoplásicos/toxicidad , Cisplatino/toxicidad , Proteína Relacionada con la Hormona Paratiroidea/orina , Lesión Renal Aguda/patología , Animales , Biomarcadores/orina , Creatinina/orina , Riñón/patología , Pruebas de Función Renal , Masculino , Ratas , Ratas Wistar
3.
J Diabetes Res ; 2013: 162846, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-23984429

RESUMEN

Parathyroid hormone-related protein (PTHrP) and its receptor type 1 (PTH1R) are extensively expressed in the kidney, where they are able to modulate renal function. Renal PTHrP is known to be overexpressed in acute renal injury. Recently, we hypothesized that PTHrP involvement in the mechanisms of renal injury might not be limited to conditions with predominant damage of the renal tubulointerstitium and might be extended to glomerular diseases, such as diabetic nephropathy (DN). In experimental DN, the overexpression of both PTHrP and the PTH1R contributes to the development of renal hypertrophy as well as proteinuria. More recent data have shown, for the first time, that PTHrP is upregulated in the kidney from patients with DN. Collectively, animal and human studies have shown that PTHrP acts as an important mediator of diabetic renal cell hypertrophy by a mechanism which involves the modulation of cell cycle regulatory proteins and TGF- ß 1. Furthermore, angiotensin II (Ang II), a critical factor in the progression of renal injury, appears to be responsible for PTHrP upregulation in these conditions. These findings provide novel insights into the well-known protective effects of Ang II antagonists in renal diseases, paving the way for new therapeutic approaches.


Asunto(s)
Diabetes Mellitus Experimental/metabolismo , Diabetes Mellitus Tipo 1/metabolismo , Nefropatías Diabéticas/etiología , Proteína Relacionada con la Hormona Paratiroidea/metabolismo , Animales , Nefropatías Diabéticas/metabolismo , Humanos , Riñón/metabolismo , Receptor de Hormona Paratiroídea Tipo 1/metabolismo
4.
J Cell Physiol ; 227(5): 1980-7, 2012 May.
Artículo en Inglés | MEDLINE | ID: mdl-21732369

RESUMEN

Hypertrophy of human mesangial cells (HMC) is among the earliest characteristics in patients with diabetic nephropathy (DN). Recently, we observed the upregulation of parathyroid hormone (PTH)-related protein (PTHrP) in experimental DN, associated with renal hypertrophy. Herein, we first examined whether PTHrP was overexpressed in human DN, and next assessed the putative role of this protein on high glucose (HG)-induced HMC hypertrophy. As previously found in mice, kidneys from diabetic patients showed an increased tubular and glomerular immunostaining for PTHrP. In HMC, HG medium increased PTHrP protein expression associated with the development of hypertrophy as assessed by cell protein content. This effect was also induced by PTHrP(1-36). HG and PTHrP(1-36)-induced hypertrophy were associated with an increase in cyclin D1 and p27Kip1 protein expression, a decreased cyclin E expression, and the prevention of cyclin E/cdk2 complex activation. Both PTHrP neutralizing antiserum (α-PTHrP) and the PTH/PTHrP receptor antagonist (JB4250) were able to abolish HG induction of hypertrophy, the aforementioned changes in cell cycle proteins, and also TGF-ß1 up-regulation. Moreover, the capability of both HG and PTHrP(1-36) to induce HMC hypertrophy was abolished by α-TGFß1. These data show for the first time that PTHrP is upregulated in the kidney of patients with DN. Our findings also demonstrate that PTHrP acts as an important mediator of HG-induced HMC hypertrophy by modulating cell cycle regulatory proteins and TGF-ß1.


Asunto(s)
Nefropatías Diabéticas/metabolismo , Nefropatías Diabéticas/patología , Riñón/metabolismo , Riñón/patología , Células Mesangiales/metabolismo , Células Mesangiales/patología , Proteína Relacionada con la Hormona Paratiroidea/metabolismo , Animales , Proliferación Celular , Células Cultivadas , Ciclina D1/genética , Ciclina D1/metabolismo , Ciclina E/genética , Ciclina E/metabolismo , Quinasa 2 Dependiente de la Ciclina/genética , Quinasa 2 Dependiente de la Ciclina/metabolismo , Inhibidor p27 de las Quinasas Dependientes de la Ciclina/genética , Inhibidor p27 de las Quinasas Dependientes de la Ciclina/metabolismo , Femenino , Humanos , Hipertrofia/metabolismo , Hipertrofia/patología , Riñón/citología , Masculino , Células Mesangiales/citología , Ratones , Persona de Mediana Edad , Proteína Relacionada con la Hormona Paratiroidea/genética , Factor de Crecimiento Transformador beta1/metabolismo
5.
Horm Res Paediatr ; 75(3): 225-30, 2011.
Artículo en Inglés | MEDLINE | ID: mdl-21196695

RESUMEN

BACKGROUND: FSH-secreting pituitary adenomas are extremely rare in children and are seldom associated with clinical manifestations of high serum gonadotrophin levels. Thus, most patients have a late presentation, usually as macroadenomas. CASE REPORTS: Two different clinical forms of presentation of FSH-secreting pituitary adenomas are reported: one in a 12-year-old boy with macroorchidism due to a pituitary microadenoma, probably FSH-secreting, and the other in a 15-year-old boy with panhypopituitarism due to an FSH-producing macroadenoma. Both patients presented slightly high or high FSH with low LH and high inhibin B levels. In the first case, the microadenoma was treated medically with cabergoline, which failed to reduce FSH and inhibin B levels. No radiological progression has been observed despite increasing testicular volume. In the second case, surgery was performed on the macroadenoma, leading to a decrease in FSH and inhibin B levels. The patient developed severe hypothalamic obesity and is currently under treatment with somatostatin. CONCLUSIONS: FSH-secreting pituitary tumors have an extremely variable clinical expression. The discrepancy between normal or slightly increased FSH and low LH values, together with high inhibin B levels, strongly suggests FSH hypersecretion which should be studied.


Asunto(s)
Adenoma/diagnóstico , Hormona Folículo Estimulante/metabolismo , Hipopituitarismo/etiología , Neoplasias Hipofisarias/diagnóstico , Enfermedades Testiculares/etiología , Adenoma/sangre , Adenoma/patología , Adenoma/fisiopatología , Adolescente , Niño , Hormona Folículo Estimulante/sangre , Humanos , Hiperplasia/etiología , Hiperplasia/patología , Subunidades beta de Inhibinas/sangre , Hormona Luteinizante/sangre , Masculino , Neoplasias Hipofisarias/sangre , Neoplasias Hipofisarias/patología , Neoplasias Hipofisarias/fisiopatología , Enfermedades Testiculares/patología , Testículo/patología
6.
J Biomed Biotechnol ; 2011: 290874, 2011.
Artículo en Inglés | MEDLINE | ID: mdl-21052497

RESUMEN

Parathyroid hormone- (PTH-) related protein (PTHrP) and its receptor, the PTH1 receptor (PTH1R), are widely expressed in the kidney, where PTHrP exerts a modulatory action on renal function. PTHrP is known to be upregulated in several experimental nephropathies such as acute renal failure (ARF), obstructive nephropathy (ON) as well as diabetic nephropathy (DN). In this paper, we will discuss the functional consequences of chronic PTHrP overexpression in the damaged kidney using a transgenic mouse strain overexpressing PTHrP in the renal proximal tubule. In both ARF and ON, PTHrP displays proinflammatory and profibrogenic actions including the induction of epithelia to mesenquima transition. Moreover, PTHrP participates in the mechanisms of renal hypertrophy as well as proteinuria in experimental DN. Angiotensin II (Ang II), a critical factor in the progression of renal injury, appears to be, at least in part, responsible for endogenous PTHrP upregulation in these pathophysiological settings. These findings provide novel insights into the well-known protective effects of Ang II antagonists in renal diseases, paving the way for new therapeutic approaches.


Asunto(s)
Modelos Animales de Enfermedad , Enfermedades Renales/metabolismo , Ratones , Proteína Relacionada con la Hormona Paratiroidea/fisiología , Lesión Renal Aguda/tratamiento farmacológico , Lesión Renal Aguda/metabolismo , Angiotensina II/antagonistas & inhibidores , Animales , Humanos , Hipertrofia/tratamiento farmacológico , Hipertrofia/metabolismo , Enfermedades Renales/tratamiento farmacológico , Ratones Transgénicos , Proteína Relacionada con la Hormona Paratiroidea/genética , Proteinuria/tratamiento farmacológico , Proteinuria/metabolismo
7.
Nephrol Dial Transplant ; 25(8): 2447-57, 2010 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-20200004

RESUMEN

BACKGROUND: Hypertrophy of podocytes is characteristic in diabetic nephropathy (DN). Previously, we observed the upregulation of parathyroid hormone-related protein (PTHrP) and its receptor PTH1R, in experimental DN, associated with renal hypertrophy. Herein, we test the hypothesis that PTHrP participates in the mechanism of high glucose (HG)-induced podocyte hypertrophy. METHODS: On mouse podocytes, hypertrophy was assessed by protein content/cell and [H(3)]leucine incorporation. Podocytes were stimulated with HG (25 mM), PTHrP(1-36) (100 nM), angiotensin II (AngII) (100 nM) or TGF-beta(1) (5 ng/mL) in the presence or absence of PTHrP-neutralizing antibodies (alpha-PTHrP), the PTH1R antagonist JB4250 (10 microM), PTHrP silencer RNA (siRNA) or TGF-beta(1) siRNA. Protein expression was analysed by western blot and immunohistochemistry. RESULTS: HG-induced hypertrophy was abolished in the presence of either alpha-PTHrP or PTHrP siRNA. This effect was associated with an inhibition of the upregulation of TGF-beta(1) and p27(Kip1). JB4250 also inhibited HG-induced p27(Kip1) upregulation. Interestingly, whilst HG and AngII were unable to stimulate the expression of p27(Kip1) on PTHrP siRNA-transfected podocytes, TGF-beta(1) was still able to upregulate p27(Kip1) in these cells. Moreover, HG and PTHrP-induced hypertrophy as well as p27(Kip1) upregulation were abolished on TGF-beta(1) siRNA-transfected podocytes. Furthermore, the glomeruli of transgenic PTHrP-overexpressing mice showed a constitutive overexpression of TGF-beta(1) and p27(Kip1) to a degree similar to that of diabetic animals. CONCLUSIONS: PTHrP seems to participate in the hypertrophic signalling triggered by HG. In this condition, AngII induces the upregulation of PTHrP, which might induce the expression of TGF-beta(1) and p27(Kip1). These findings provide new insights into the protective effects of AngII antagonists in DN, opening new paths for intervention.


Asunto(s)
Inhibidor p27 de las Quinasas Dependientes de la Ciclina/metabolismo , Nefropatías Diabéticas/metabolismo , Proteína Relacionada con la Hormona Paratiroidea/metabolismo , Podocitos/metabolismo , Podocitos/patología , Factor de Crecimiento Transformador beta1/metabolismo , Angiotensina II/antagonistas & inhibidores , Angiotensina II/farmacología , Animales , Células Cultivadas , Nefropatías Diabéticas/patología , Modelos Animales de Enfermedad , Glucosa/efectos adversos , Hipertrofia/metabolismo , Hipertrofia/patología , Ratones , Proteína Relacionada con la Hormona Paratiroidea/farmacología , Fragmentos de Péptidos/farmacología , Podocitos/efectos de los fármacos , Transducción de Señal/fisiología
8.
J Neurol Sci ; 277(1-2): 167-71, 2009 Feb 15.
Artículo en Inglés | MEDLINE | ID: mdl-19027924

RESUMEN

Myotilinopathies are a group of muscle disorders caused by mutations in the MYOT gene. It was first described in two families suffering from limb girdle muscle dystrophy type 1 (LGMD 1A), and later identified in a subset of dominant or sporadic patients suffering from myofibrillar myopathy, as well as in a family with spheroid body myopathy. Disease phenotypes associated with MYOT mutations are clinically heterogeneous and include pure LGMD forms as well as late-onset distal myopathies. We report here on a 53-year-old male suffering from a unique clinical profile characterized by generalized symmetrical increase in muscle bulk leading to a Herculean appearance. Muscle weakness and stiffness in the lower extremities were the patient's main complaints. Muscle MRI showed extensive fatty infiltration in the thigh and leg muscles and a muscle biopsy showed a myofibrillar myopathy with prominent protein aggregates. Gene sequencing revealed a Ser55Phe missense mutation in the myotilin gene. The mutation was identified in his older brother, who presented a mild hypertrophic appearance and had a myopathic pattern in EMG, despite not presenting any of the complaints of the proband and having normal muscle strength. This finding, and his deceased father and paternal aunt's similar gait disorders, suggest that this is in fact a new autosomal dominant kindred. The present observations further expand the spectrum of clinical manifestations associated with mutations in the myotilin gene.


Asunto(s)
Proteínas del Citoesqueleto/genética , Proteínas Musculares/genética , Rigidez Muscular/genética , Rigidez Muscular/patología , Enfermedades Musculares/genética , Enfermedades Musculares/patología , Biopsia , Conectina , Salud de la Familia , Humanos , Hipertrofia , Imagen por Resonancia Magnética , Masculino , Proteínas de Microfilamentos , Persona de Mediana Edad , Debilidad Muscular/genética , Debilidad Muscular/patología , Fenotipo , Mutación Puntual
9.
Clin Sci (Lond) ; 113(7): 319-27, 2007 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-17501718

RESUMEN

Evidence suggests that PTHrP [PTH (parathyroid hormone)-related protein] can act as an inflammatory mediator in several pathological settings including cardiovascular disease. The aim of the present study was to determine whether PTHrP might be involved in human platelet activation. We used a turbidimetric method to determine platelet aggregation. The expression of PTH1R (PTH type 1 receptor) in human platelets was analysed by Western blot and flow cytometry analyses. PTHrP-(1-36) (10(-7) mol/l) by itself failed to modify the activation of platelets. However, it significantly enhanced ADP-induced platelet activation, and also increased the ability of other agonists (thrombin, collagen and arachidonic acid) to induce platelet aggregation. H89 (10(-6) mol/l) and 25 x 10(-6) mol/l Rp-cAMPS (adenosine 3',5'-cyclic monophosphorothioate Rp-isomer), two protein kinase A inhibitors, and 25 x 10(-9) mol/l bisindolylmaleimide I, a protein kinase C inhibitor, partially decreased the enhancing effect of PTHrP-(1-36) on ADP-induced platelet activation. Meanwhile, 10(-6) mol/l PTHrP-(7-34), a PTH1R antagonist, as well as 10(-5) mol/l PD098059, a MAPK (mitogen-activated protein kinase) inhibitor, or a farnesyltransferase inhibitor abolished this effect of PTHrP-(1-36). Moreover, 10(-7) mol/l PTHrP-(1-36) increased (2-fold over control) MAPK activation in human platelets. PTH1R was detected in platelets, and the number of platelets expressing it on their surface in patients during AMI (acute myocardial infarction) was not different from that in a group of patients with similar cardiovascular risk factors without AMI. Western blot analysis showed that total PTH1R protein levels were markedly higher in platelets from control than those from AMI patients. PTH1R was found in plasma, where its levels were increased in AMI patients compared with controls. In conclusion, human platelets express the PTH1R. PTHrP can interact with this receptor to enhance human platelet activation induced by several agonists through a MAPK-dependent mechanism.


Asunto(s)
Infarto del Miocardio/fisiopatología , Proteína Relacionada con la Hormona Paratiroidea/farmacología , Activación Plaquetaria/efectos de los fármacos , Anciano , Anciano de 80 o más Años , Plaquetas/metabolismo , Western Blotting , Citometría de Flujo , Humanos , Indoles/metabolismo , Maleimidas/metabolismo , Proteínas Quinasas Activadas por Mitógenos/metabolismo , Proteína Relacionada con la Hormona Paratiroidea/fisiología , Agregación Plaquetaria/efectos de los fármacos
10.
J Am Soc Nephrol ; 17(6): 1594-603, 2006 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-16672315

RESUMEN

Parathyroid hormone-related protein (PTHrP) is shortly upregulated in acute renal injury, but its pathophysiologic role is unclear. Investigated was whether PTHrP might act as a profibrogenic factor in mice that do or do not overexpress PTHrP in the proximal tubule after folic acid (FA) nephrotoxicity, a model of acute renal damage followed by partial regeneration and patchy tubulointerstitial fibrosis. It was found that constitutive PTHrP overexpression in these animals conveyed a significant increase in tubulointerstitial fibrosis, associated with both fibroblast activation (as alpha-smooth muscle actin staining) and macrophage influx, compared with control littermates at 2 to 3 wk after FA damage. Cell proliferation and survival was higher (P<0.01) in the renal interstitium of PTHrP-overexpressing mice than in control littermates within this period after injury. Moreover, the former mice had a constitutive Bcl-XL protein overexpression. In vitro studies in renal tubulointerstitial and fibroblastic cells strongly suggest that PTHrP (1-36) (100 nM) reduced FA-induced apoptosis through a dual mechanism involving Bcl-XL upregulation and Akt and Bad phosphorylation. PTHrP (1-36) also stimulated monocyte chemoattractant protein-1 expression in tubuloepithelial cells, as well as type-1 procollagen gene expression and fibronectin (mRNA levels and protein secretion) in these cells and renal fibroblastic cells. Our findings indicate that this peptide, by interaction with the PTH1 receptor, can increase tubulointerstitial cell survival and seems to act as a proinflammatory and profibrogenic factor in the FA-damaged kidney.


Asunto(s)
Apoptosis , Ácido Fólico/toxicidad , Túbulos Renales/efectos de los fármacos , Riñón/efectos de los fármacos , Proteína Relacionada con la Hormona Paratiroidea/fisiología , Actinas/metabolismo , Animales , Proliferación Celular , Matriz Extracelular/metabolismo , Fibrosis , Riñón/lesiones , Túbulos Renales/lesiones , Macrófagos/metabolismo , Ratones , Ratones Transgénicos , Músculo Liso/metabolismo , Proteína Relacionada con la Hormona Paratiroidea/metabolismo , Ratas
11.
J Am Soc Nephrol ; 16(4): 939-49, 2005 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-15728788

RESUMEN

Parathyroid hormone-related protein (PTHrP), a mitogenic factor for renal cells, is overexpressed in acute renal failure (ARF). Recent data support an association between PTHrP and the renin-angiotensin system in the damaged kidney. The effects of angiotensin II (Ang II) inhibitors (quinapril, enalapril, and/or losartan) on PTHrP and the PTH1 receptor (PTH1R) expression in rats with either folic acid (FA)- or gentamicin-induced ARF were analyzed. The decreased renal function and the PTHrP upregulation and PTH1R downregulation induced by the nephrotoxins were inhibited by the Ang II blockers. In tubuloepithelial cells NRK-52E, the rapid (10 min) increase in PTHrP mRNA by FA, associated with a perinuclear relocalization of Ang II/AT1 receptor, was inhibited by losartan but not candesartan, which traps Ang II receptors at the cell surface. Maximal PTHrP protein overexpression by FA (at 24 to 72 h)-or by exogenous Ang II-was abolished by both Ang II antagonists. PTHrP upregulation by FA was preceded by increased extracellular signal-regulated kinase (ERK) phosphorylation and inhibited by the ERK inhibitor PD098059. FA also activated cAMP response element-binding (CREB) protein, and this was prevented by losartan in these cells. Moreover, PTHrP mRNA overexpression by either FA or Ang II occurred in NRK 52E that were transfected with a CREB construct but not the dominant-negative CREB133 construct. These findings demonstrate that the decreased renal function and PTHrP overexpression in nephrotoxin-damaged kidney depends on renin-angiotensin system. In this setting, intracellular Ang II/AT1 receptor recycling seems to be related to PTHrP induction through ERK and CREB activation in tubuloepithelial cells.


Asunto(s)
Lesión Renal Aguda/metabolismo , Proteína Relacionada con la Hormona Paratiroidea/metabolismo , Sistema Renina-Angiotensina , Lesión Renal Aguda/inducido químicamente , Lesión Renal Aguda/patología , Lesión Renal Aguda/fisiopatología , Angiotensina II/antagonistas & inhibidores , Angiotensina II/metabolismo , Bloqueadores del Receptor Tipo 1 de Angiotensina II/farmacología , Animales , Línea Celular , Enalapril/farmacología , Ácido Fólico/farmacología , Gentamicinas , Riñón/efectos de los fármacos , Riñón/fisiopatología , Túbulos Renales/efectos de los fármacos , Túbulos Renales/metabolismo , Túbulos Renales/patología , Losartán/farmacología , Masculino , Quinapril , Ratas , Ratas Wistar , Receptor de Hormona Paratiroídea Tipo 1/metabolismo , Tetrahidroisoquinolinas/farmacología
12.
AJNR Am J Neuroradiol ; 25(7): 1274-82, 2004 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-15313724

RESUMEN

BACKGROUND AND PURPOSE: Sonographic brain studies are classically performed through the anterior fontanelle, but visualization of posterior supratentorial and infratentorial structures is poor with this approach. Posterior fontanelle sonography is recommended for better assessment of these structures. Our purpose was 1) to determine whether sonography of the brain through the posterior fontanelle (PF) improves visualization of brain lesions when added to the routine anterior fontanelle (AF) approach and 2) to describe standardized PF coronal and sagittal sections. METHODS: In this prospective study (conducted from February 1999 to January 2001), PF sonography was added to AF sonography in 165 consecutive premature neonates with a birth weight of < 2000 g. Sonograms were recorded in digital format for re-evaluation at the end of the study. Lesions were grouped as congenital, infectious, hemorrhagic, or hypoxic-ischemic. The chi2 test for paired data and the kappa coefficient were used to compare diagnoses with AF alone and diagnoses with AF plus PF. RESULTS: PF sonography was performed in 164 of 165 patients. Results were normal in 86 and abnormal in 78. The single posterior fossa malformation detected in this series was best delineated with the PF approach. PF sonography increased the diagnostic rate of grade II hemorrhage by 32%. Cerebellar hemorrhage (two patients) and cerebellar abscesses (one patient) were diagnosed by using the PF approach. PF sonography did not contribute to the diagnosis of periventricular leukomalacia. CONCLUSION: Study of the neonatal brain with the addition of PF sonography afforded greater accuracy in detecting intraventricular hemorrhage compared with AF sonography alone, especially when the ventricle was not dilated. The PF approach better defines posterior fossa malformations.


Asunto(s)
Daño Encefálico Crónico/congénito , Daño Encefálico Crónico/diagnóstico por imagen , Tronco Encefálico/anomalías , Tronco Encefálico/diagnóstico por imagen , Enfermedades Cerebelosas/congénito , Enfermedades Cerebelosas/diagnóstico por imagen , Ecoencefalografía/instrumentación , Enfermedades del Prematuro/diagnóstico por imagen , Absceso Encefálico/congénito , Absceso Encefálico/diagnóstico por imagen , Fosa Craneal Posterior/anomalías , Fosa Craneal Posterior/diagnóstico por imagen , Suturas Craneales/diagnóstico por imagen , Diagnóstico Diferencial , Femenino , Humanos , Hipoxia Encefálica/congénito , Hipoxia Encefálica/diagnóstico por imagen , Recién Nacido de Bajo Peso , Recién Nacido , Hemorragias Intracraneales/congénito , Hemorragias Intracraneales/diagnóstico por imagen , Leucomalacia Periventricular/diagnóstico por imagen , Masculino , Meningitis Bacterianas/congénito , Meningitis Bacterianas/diagnóstico por imagen , Tamizaje Neonatal/instrumentación , Estudios Prospectivos , Sensibilidad y Especificidad , Transductores
13.
Free Radic Biol Med ; 36(10): 1241-7, 2004 May 15.
Artículo en Inglés | MEDLINE | ID: mdl-15110389

RESUMEN

Oxidative stress has been implicated in the pathophysiology of Alzheimer's disease (AD) and diabetes mellitus (DM). N epsilon-carboxymethyllysine (CML) is an advanced glycation end product (AGE) recently found to be associated with oxidative stress mechanisms. Using immunocytochemical methods we examined the distribution of CML in brain tissue from AD and DM subjects and aging controls. CML reactivity was present in the cytoplasm of neurons, but there were marked differences in the intensity of expression, number of cells, and topographical distribution. CML expression was higher in hippocampus than in frontal and temporal cortex. In the hippocampus, neuronal and, to an extent, glial expression was more marked in CA3 and CA4 than in CA1 and CA2. In AD, CML was found to be coexpressed with tau protein, showing the similar neurofibrillary tangle shape, as well as in neuritic plaques but not in the core of amyloid plaques. We noted an increasing degree of CML expression such that the highest reactivity was evident in those with both AD and DM, followed by AD, DM, and aging controls. There was an inverse relationship between Braak staging and topography of CML expression. Although DM cases did not show Abeta deposition or neurofibrillary tangles, these findings suggest increased CML expression is not limited to AD. Nonetheless, high CML expression in AD with coexistent DM suggests there are additive effects compared with AD alone. It is plausible that the microangiopathy also associated with DM could worsen AD pathogenesis.


Asunto(s)
Envejecimiento/efectos de los fármacos , Enfermedad de Alzheimer/metabolismo , Encéfalo/efectos de los fármacos , Diabetes Mellitus/metabolismo , Lisina/análogos & derivados , Lisina/farmacología , Neuronas/efectos de los fármacos , Anciano , Anciano de 80 o más Años , Enfermedad de Alzheimer/patología , Citoplasma/efectos de los fármacos , Citoplasma/metabolismo , Diabetes Mellitus/patología , Femenino , Productos Finales de Glicación Avanzada , Humanos , Masculino , Persona de Mediana Edad , Ovillos Neurofibrilares/metabolismo , Ovillos Neurofibrilares/patología , Neuronas/citología , Neuronas/metabolismo , Placa Amiloide/efectos de los fármacos , Proteínas tau/metabolismo
14.
J Am Soc Nephrol ; 15(1): 112-25, 2004 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-14694163

RESUMEN

Treatment of acute renal failure (ARF) would be enhanced by identification of factors that accelerate renal recovery from injury. Parathyroid hormone-related protein (PTHrP) and hepatocyte growth factor (HGF) have been shown to stimulate proliferation in proximal nephron-derived cells. For studying the pathophysiologic roles and therapeutic potential of these two factors in ARF, transgenic mice overexpressing PTHrP or HGF in the proximal tubule under the direction of the gamma-glutamyl transpeptidase-I promoter were developed. These mice display (1) abundant expression of the respective transgenes in the kidney; (2) similar PTH type I receptor and HGF receptor (c-met) expression levels in the proximal tubule compared with control littermates; and (3) normal renal morphology, function, and tubule cell proliferation under basal conditions. However, in contrast to control mice, when acute ischemic renal injury was induced, renal function rapidly and dramatically recovered in HGF-overexpressing mice. In addition, 48 h after ischemia, HGF-overexpressing transgenic mice displayed a fourfold increase in tubule cell proliferation and a threefold decrease in apoptotic tubule cell death compared with control mice. In contrast, PTHrP-overexpressing mice responded to either ischemic or folic acid-induced renal damage similarly to control mice. These studies demonstrate that overexpression of PTHrP in the proximal nephron of mice does not seem to provide protection against acute renal injury. In marked contrast, HGF overexpression results in dramatic protection from ischemia-induced ARF, without inducing any apparent alteration in the physiology of the kidney under normal conditions. These studies suggest that HGF, when targeted specifically to the proximal tubule, may have therapeutic potential in providing protection against ischemia-induced renal failure.


Asunto(s)
Lesión Renal Aguda/prevención & control , Factor de Crecimiento de Hepatocito/administración & dosificación , Riñón/irrigación sanguínea , Proteína Relacionada con la Hormona Paratiroidea/administración & dosificación , Daño por Reperfusión/prevención & control , Animales , Regulación de la Expresión Génica , Factor de Crecimiento de Hepatocito/biosíntesis , Túbulos Renales Proximales/metabolismo , Ratones , Ratones Transgénicos , Proteína Relacionada con la Hormona Paratiroidea/biosíntesis
15.
Acta Neuropathol ; 105(1): 69-75, 2003 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-12471464

RESUMEN

We report an unusual case of encephalo-entero-myopathy associated with the A8344G mutation in the tRNA(Lys) gene of mitochondrial DNA (mtDNA). This patient had mitochondrial myopathy, multiple lipomatosis, mild hearing loss, stroke-like episodes, and paralytic ileus, but she lacked the canonical clinical features of MERRF, myoclonus, epilepsy, or ataxia. We conducted genetic, biochemical, histochemical, and immunohistochemical studies in skeletal muscle, brain, intestine, and lipoma tissue. The mutation was abundant in all tissues, and cytochrome c oxidase (COX) activity was selectively decreased in brain and small intestine. COX deficiency was also documented histochemically and immunohistochemically in the small intestine, suggesting that mitochondrial dysfunction played a role in the pathogenesis of paralytic ileus. This case illustrates an unusual and dramatic clinical phenotype of the A8344G mutation, characterized by stroke-like episodes and acute ileus.


Asunto(s)
ADN Mitocondrial/genética , Enfermedades Gastrointestinales/genética , Síndrome MERRF/genética , Mutación/genética , ARN de Transferencia de Lisina/genética , Accidente Cerebrovascular/genética , Adulto , Encéfalo/enzimología , Deficiencia de Citocromo-c Oxidasa/complicaciones , Complejo IV de Transporte de Electrones/metabolismo , Femenino , Enfermedades Gastrointestinales/enzimología , Enfermedades Gastrointestinales/patología , Histocitoquímica , Humanos , Inmunohistoquímica , Intestino Delgado/enzimología , Accidente Cerebrovascular/complicaciones , Accidente Cerebrovascular/enzimología , Accidente Cerebrovascular/patología
16.
J Am Soc Nephrol ; 13(6): 1595-607, 2002 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-12039989

RESUMEN

Angiotensin II (AngII) participates in the pathogenesis of kidney damage. Parathyroid hormone (PTH)-related protein (PTHrP), a vasodilator and mitogenic agent, is upregulated during renal injury. The aim of this study was to investigate the potential relation between AngII and PTHrP system in the kidney. Different methods were used to find that both rat mesangial and mouse tubuloepithelial cells express PTHrP and the type 1 PTH/PTHrP receptor (PTH1R). In these cells, AngII increased PTHrP mRNA and protein production. In contrast, PTH1R mRNA was increased in mesangial cells and downregulated in tubular cells, but its protein levels were unmodified in both cells. AT(1) antagonist, but not AT(2), abolished AngII effects on PTHrP/PTH1R. The in vivo effect of AngII was further investigated by systemic infusion (a low dose of 50 ng/kg per min) into normal rats. In controls, PTHrP immunostaining was mainly detected in renal tubules. In AngII-infused rats, PTHrP staining increased in renal tubules and appeared in the glomerulus and the renal vessels. After AngII infusion, PTHR1 staining was markedly increased in all these renal structures at day 3 but remained elevated only in tubules at day 7. The AT(1) antagonist, but not the AT(2), significantly diminished AngII-induced PTHrP and PTHR1 overexpression in the renal tissue, associated with a decrease in tubular damage and fibrosis. The results indicate that AngII regulates renal PTHrP/PTH1R system via AT(1) receptors. These findings demonstrate that PTHrP upregulation occurs in association with the mechanisms of AngII-induced kidney injury.


Asunto(s)
Angiotensina II/toxicidad , Riñón/efectos de los fármacos , Hormonas Peptídicas/biosíntesis , Receptores de Hormona Paratiroidea/biosíntesis , Animales , Aorta/metabolismo , Células Cultivadas , Femenino , Mesangio Glomerular/efectos de los fármacos , Mesangio Glomerular/metabolismo , Riñón/metabolismo , Proteína Relacionada con la Hormona Paratiroidea , Hormonas Peptídicas/análisis , Ratas , Ratas Wistar , Receptor de Angiotensina Tipo 1 , Receptor de Hormona Paratiroídea Tipo 1 , Receptores de Angiotensina/fisiología , Receptores de Hormona Paratiroidea/análisis , Regulación hacia Arriba
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...