Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 17 de 17
Filter
1.
Transl Stroke Res ; 2024 Jan 17.
Article in English | MEDLINE | ID: mdl-38231413

ABSTRACT

Advances in neonatology have significantly reduced mortality rates due to prematurity. However, complications of prematurity have barely changed in recent decades. Germinal matrix-intraventricular hemorrhage (GM-IVH) is one of the most severe complications of prematurity, and these children are prone to suffer short- and long-term sequelae, including cerebral palsy, cognitive and motor impairments, or neuropsychiatric disorders. Nevertheless, GM-IVH has no successful treatment. VP3.15 is a small, heterocyclic molecule of the 5-imino-1,2,4-thiadiazole family with a dual action as a phosphodiesterase 7 and glycogen synthase kinase-3ß (GSK-3ß) inhibitor. VP3.15 reduces neuroinflammation and neuronal loss in other neurodegenerative disorders and might ameliorate complications associated with GM-IVH. We administered VP3.15 to a mouse model of GM-IVH. VP3.15 reduces the presence of hemorrhages and microglia in the short (P14) and long (P110) term. It ameliorates brain atrophy and ventricle enlargement while limiting tau hyperphosphorylation and neuronal and myelin basic protein loss. VP3.15 also improves proliferation and neurogenesis as well as cognition after the insult. Interestingly, plasma gelsolin levels, a feasible biomarker of brain damage, improved after VP3.15 treatment. Altogether, our data support the beneficial effects of VP3.15 in GM-IVH by ameliorating brain neuroinflammatory, vascular and white matter damage, ultimately improving cognitive impairment associated with GM-IVH.

2.
Diabetologia ; 66(8): 1557-1575, 2023 08.
Article in English | MEDLINE | ID: mdl-37351595

ABSTRACT

AIMS/HYPOTHESIS: Type 2 diabetes is associated with increased risk of cognitive decline although the pathogenic basis for this remains obscure. Deciphering diabetes-linked molecular mechanisms in cells of the cerebral cortex could uncover novel therapeutic targets. METHODS: Single-cell transcriptomic sequencing (scRNA-seq) was conducted on the cerebral cortex in a mouse model of type 2 diabetes (db/db mice) and in non-diabetic control mice in order to identify gene expression changes in distinct cell subpopulations and alterations in cell type composition. Immunohistochemistry and metabolic assessment were used to validate the findings from scRNA-seq and to investigate whether these cell-specific dysfunctions impact the neurovascular unit (NVU). Furthermore, the behavioural and cognitive alterations related to these dysfunctions in db/db mice were assessed via Morris water maze and novel object discrimination tests. Finally, results were validated in post-mortem sections and protein isolates from individuals with type 2 diabetes. RESULTS: Compared with non-diabetic control mice, the db/db mice demonstrated disrupted brain function as revealed by losses in episodic and spatial memory and this occurred concomitantly with dysfunctional NVU, neuronal circuitry and cerebral atrophy. scRNA-seq of db/db mouse cerebral cortex revealed cell population changes in neurons, glia and microglia linked to functional regulatory disruption including neuronal maturation and altered metabolism. These changes were validated through immunohistochemistry and protein expression analysis not just in the db/db mouse cerebral cortex but also in post-mortem sections and protein isolates from individuals with type 2 diabetes (74.3 ± 5.5 years) compared with non-diabetic control individuals (87.0 ± 8.5 years). Furthermore, metabolic and synaptic gene disruptions were evident in cortical NVU cell populations and associated with a decrease in vascular density. CONCLUSIONS/INTERPRETATION: Taken together, our data reveal disruption in the cellular and molecular architecture of the cerebral cortex induced by diabetes, which can explain, at least in part, the basis for progressive cognitive decline in individuals with type 2 diabetes. DATA AVAILABILITY: The single-cell sequencing data that supports this study are available at GEO accession GSE217665 ( https://www.ncbi.nlm.nih.gov/geo/query/acc.cgi?acc=GSE217665 ).


Subject(s)
Cognitive Dysfunction , Diabetes Mellitus, Type 2 , Mice , Animals , Diabetes Mellitus, Type 2/complications , Cognitive Dysfunction/drug therapy , Cerebral Cortex/metabolism , Disease Models, Animal
3.
Fluids Barriers CNS ; 19(1): 88, 2022 Nov 07.
Article in English | MEDLINE | ID: mdl-36345028

ABSTRACT

BACKGROUND: While aging is the main risk factor for Alzheimer´s disease (AD), emerging evidence suggests that metabolic alterations such as type 2 diabetes (T2D) are also major contributors. Indeed, several studies have described a close relationship between AD and T2D with clinical evidence showing that both diseases coexist. A hallmark pathological event in AD is amyloid-ß (Aß) deposition in the brain as either amyloid plaques or around leptomeningeal and cortical arterioles, thus constituting cerebral amyloid angiopathy (CAA). CAA is observed in 85-95% of autopsy cases with AD and it contributes to AD pathology by limiting perivascular drainage of Aß. METHODS: To further explore these alterations when AD and T2D coexist, we have used in vivo multiphoton microscopy to analyze over time the Aß deposition in the form of plaques and CAA in a relevant model of AD (APPswe/PS1dE9) combined with T2D (db/db). We have simultaneously assessed the effects of high-fat diet-induced prediabetes in AD mice. Since both plaques and CAA are implicated in oxidative-stress mediated vascular damage in the brain, as well as in the activation of matrix metalloproteinases (MMP), we have also analyzed oxidative stress by Amplex Red oxidation, MMP activity by DQ™ Gelatin, and vascular functionality. RESULTS: We found that prediabetes accelerates amyloid plaque and CAA deposition, suggesting that initial metabolic alterations may directly affect AD pathology. T2D significantly affects vascular pathology and CAA deposition, which is increased in AD-T2D mice, suggesting that T2D favors vascular accumulation of Aß. Moreover, T2D synergistically contributes to increase CAA mediated oxidative stress and MMP activation, affecting red blood cell velocity. CONCLUSIONS: Our data support the cross-talk between metabolic disease and Aß deposition that affects vascular integrity, ultimately contributing to AD pathology and related functional changes in the brain microvasculature.


Subject(s)
Alzheimer Disease , Cerebral Amyloid Angiopathy , Diabetes Mellitus, Type 2 , Prediabetic State , Animals , Mice , Alzheimer Disease/metabolism , Disease Models, Animal , Diabetes Mellitus, Type 2/complications , Diabetes Mellitus, Type 2/metabolism , Diabetes Mellitus, Type 2/pathology , Prediabetic State/complications , Prediabetic State/metabolism , Prediabetic State/pathology , Cerebral Amyloid Angiopathy/metabolism , Amyloid beta-Peptides/metabolism , Plaque, Amyloid/complications , Plaque, Amyloid/metabolism , Plaque, Amyloid/pathology , Brain/metabolism , Matrix Metalloproteinases
4.
Front Cell Dev Biol ; 10: 908045, 2022.
Article in English | MEDLINE | ID: mdl-36035990

ABSTRACT

Germinal matrix-intraventricular hemorrhage (GM-IVH) is the most frequent intracranial hemorrhage in the preterm infant (PT). Long-term GM-IVH-associated sequelae include cerebral palsy, sensory and motor impairment, learning disabilities, or neuropsychiatric disorders. The societal and health burden associated with GM-IVH is worsened by the fact that there is no successful treatment to limit or reduce brain damage and neurodevelopment disabilities. Caffeine (Caf) is a methylxanthine that binds to adenosine receptors, regularly used to treat the apnea of prematurity. While previous studies support the beneficial effects at the brain level of Caf in PT, there are no studies that specifically focus on the role of Caf in GM-IVH. Therefore, to further understand the role of Caf in GM-IVH, we have analyzed two doses of Caf (10 and 20 mg/kg) in a murine model of the disease. We have analyzed the short (P14) and long (P70) effects of the treatment on brain atrophy and neuron wellbeing, including density, curvature, and phospho-tau/total tau ratio. We have analyzed proliferation and neurogenesis, as well as microglia and hemorrhage burdens. We have also assessed the long-term effects of Caf treatment at cognitive level. To induce GM-IVH, we have administered intraventricular collagenase to P7 CD1 mice and have analyzed these animals in the short (P14) and long (P70) term. Caf showed a general neuroprotective effect in our model of GM-IVH of the PT. In our study, Caf administration diminishes brain atrophy and ventricle enlargement. Likewise, Caf limits neuronal damage, including neurite curvature and tau phosphorylation. It also contributes to maintaining neurogenesis in the subventricular zone, a neurogenic niche that is severely affected after GM-IVH. Furthermore, Caf ameliorates small vessel bleeding and inflammation in both the cortex and the subventricular zone. Observed mitigation of brain pathological features commonly associated with GM-IVH also results in a significant improvement of learning and memory abilities in the long term. Altogether, our data support the promising effects of Caf to reduce central nervous system complications associated with GM-IVH.

5.
Trends Endocrinol Metab ; 33(1): 50-71, 2022 01.
Article in English | MEDLINE | ID: mdl-34794851

ABSTRACT

Type 2 diabetes (T2D) is associated with multiple comorbidities, including diabetic retinopathy (DR) and cognitive decline, and T2D patients have a significantly higher risk of developing Alzheimer's disease (AD). Both DR and AD are characterized by a number of pathological mechanisms that coalesce around the neurovascular unit, including neuroinflammation and degeneration, vascular degeneration, and glial activation. Chronic hyperglycemia and insulin resistance also play a significant role, leading to activation of pathological mechanisms such as increased oxidative stress and the accumulation of advanced glycation end-products (AGEs). Understanding these common pathways and the degree to which they occur simultaneously in the brain and retina during diabetes will provide avenues to identify T2D patients at risk of cognitive decline.


Subject(s)
Alzheimer Disease , Cognitive Dysfunction , Diabetes Mellitus, Type 2 , Diabetic Retinopathy , Alzheimer Disease/metabolism , Cognitive Dysfunction/etiology , Diabetes Mellitus, Type 2/complications , Diabetic Retinopathy/metabolism , Diabetic Retinopathy/pathology , Glycation End Products, Advanced/metabolism , Humans
6.
Alzheimers Res Ther ; 13(1): 112, 2021 06 12.
Article in English | MEDLINE | ID: mdl-34118986

ABSTRACT

BACKGROUND: The described relationship between Alzheimer's disease (AD) and type 2 diabetes (T2D) and the fact that AD has no succesful treatment has led to the study of antidiabetic drugs that may limit or slow down AD pathology. MAIN BODY: Although T2D treatment has evident limitations, options are increasing including glucagon-like peptide 1 analogs. Among these, liraglutide (LRGT) is commonly used by T2D patients to improve ß cell function and suppress glucagon to restore normoglycaemia. Interestingly, LRGT also counterbalances altered brain metabolism and has anti-inflammatory properties. Previous studies have reported its capacity to reduce AD pathology, including amyloid production and deposition, tau hyperphosphorylation, or neuronal and synaptic loss in animal models of AD, accompanied by cognitive improvement. Given the beneficial effects of LRGT at central level, studies in patients have been carried out, showing modest beneficial effects. At present, the ELAD trial (Evaluating Liraglutide in Alzheimer's Disease NCT01843075) is an ongoing phase IIb study in patients with mild AD. In this minireview, we resume the outcomes of LRGT treatment in preclinical models of AD as well as the available results in patients up to date. CONCLUSION: The effects of LRGT on animal models show significant benefits in AD pathology and cognitive impairment. While studies in patients are limited, ongoing clinical trials will probably provide more definitive conclusions on the role of LRGT in AD patients.


Subject(s)
Alzheimer Disease , Diabetes Mellitus, Type 2 , Alzheimer Disease/drug therapy , Amyloid beta-Peptides , Animals , Diabetes Mellitus, Type 2/complications , Diabetes Mellitus, Type 2/drug therapy , Disease Models, Animal , Humans , Hypoglycemic Agents/therapeutic use , Liraglutide/therapeutic use , tau Proteins
7.
Biomolecules ; 11(2)2021 02 10.
Article in English | MEDLINE | ID: mdl-33578998

ABSTRACT

Alzheimer's disease (AD) is the most common cause of dementia. Epidemiological studies show the association between AD and type 2 diabetes (T2DM), although the mechanisms are not fully understood. Dietary habits and lifestyle, that are risk factors in both diseases, strongly modulate gut microbiota composition. Also, the brain-gut axis plays a relevant role in AD, diabetes and inflammation, through products of bacterial metabolism, like short-chain fatty acids. We provide a comprehensive review of current literature on the relation between dysbiosis, altered inflammatory cytokines profile and microglia in preclinical models of AD, T2DM and models that reproduce both diseases as commonly observed in the clinic. Increased proinflammatory cytokines, such as IL-1ß and TNF-α, are widely detected. Microbiome analysis shows alterations in Actinobacteria, Bacteroidetes or Firmicutes phyla, among others. Altered α- and ß-diversity is observed in mice depending on genotype, gender and age; therefore, alterations in bacteria taxa highly depend on the models and approaches. We also review the use of pre- and probiotic supplements, that by favoring a healthy microbiome ameliorate AD and T2DM pathologies. Whereas extensive studies have been carried out, further research would be necessary to fully understand the relation between diet, microbiome and inflammation in AD and T2DM.


Subject(s)
Alzheimer Disease/complications , Diabetes Complications/metabolism , Diet , Microbiota , Actinobacteria/metabolism , Alzheimer Disease/metabolism , Alzheimer Disease/microbiology , Animals , Bacteroidetes/metabolism , Diabetes Mellitus/metabolism , Diabetes Mellitus/microbiology , Disease Models, Animal , Firmicutes/metabolism , Gastrointestinal Microbiome , Humans , Inflammation , Life Style , Mice , Prediabetic State/metabolism , Prediabetic State/microbiology , Probiotics , Risk Factors
8.
Front Aging Neurosci ; 13: 741923, 2021.
Article in English | MEDLINE | ID: mdl-34975451

ABSTRACT

Alzheimer's disease is the most common form of dementia, and epidemiological studies support that type 2 diabetes (T2D) is a major contributor. The relationship between both diseases and the fact that Alzheimer's disease (AD) does not have a successful treatment support the study on antidiabetic drugs limiting or slowing down brain complications in AD. Among these, liraglutide (LRGT), a glucagon-like peptide-1 agonist, is currently being tested in patients with AD in the Evaluating Liraglutide in Alzheimer's Disease (ELAD) clinical trial. However, the effects of LRGT on brain pathology when AD and T2D coexist have not been assessed. We have administered LRGT (500 µg/kg/day) to a mixed murine model of AD and T2D (APP/PS1xdb/db mice) for 20 weeks. We have evaluated metabolic parameters as well as the effects of LRGT on learning and memory. Postmortem analysis included assessment of brain amyloid-ß and tau pathologies, microglia activation, spontaneous bleeding and neuronal loss, as well as insulin and insulin-like growth factor 1 receptors. LRGT treatment reduced glucose levels in diabetic mice (db/db and APP/PS1xdb/db) after 4 weeks of treatment. LRGT also helped to maintain insulin levels after 8 weeks of treatment. While we did not detect any effects on cortical insulin or insulin-like growth factor 1 receptor m-RNA levels, LRGT significantly reduced brain atrophy in the db/db and APP/PS1xdb/db mice. LRGT treatment also rescued neuron density in the APP/PS1xdb/db mice in the proximity (p = 0.008) far from amyloid plaques (p < 0.001). LRGT reduced amyloid plaque burden in the APP/PS1 animals (p < 0.001), as well as Aß aggregates levels (p = 0.046), and tau hyperphosphorylation (p = 0.009) in the APP/PS1xdb/db mice. Spontaneous bleeding was also ameliorated in the APP/PS1xdb/db animals (p = 0.012), and microglia burden was reduced in the proximity of amyloid plaques in the APP/PS1 and APP/PS1xdb/db mice (p < 0.001), while microglia was reduced in areas far from amyloid plaques in the db/db and APP/PS1xdb/db mice (p < 0.001). This overall improvement helped to rescue cognitive impairment in AD-T2D mice in the new object discrimination test (p < 0.001) and Morris water maze (p < 0.001). Altogether, our data support the role of LRGT in reduction of associated brain complications when T2D and AD occur simultaneously, as regularly observed in the clinical arena.

9.
Front Cell Dev Biol ; 8: 571258, 2020.
Article in English | MEDLINE | ID: mdl-33043002

ABSTRACT

The germinal matrix-intraventricular hemorrhage (GM-IVH) is one of the most devastating complications of prematurity. The short- and long-term neurodevelopmental consequences after severe GM-IVH are a major concern for neonatologists. These kids are at high risk of psychomotor alterations and cerebral palsy; however, therapeutic approaches are limited. Erythropoietin (EPO) has been previously used to treat several central nervous system complications due to its role in angiogenesis, neurogenesis and as growth factor. In addition, EPO is regularly used to reduce the number of transfusions in the preterm infant. Moreover, EPO crosses the blood-brain barrier and EPO receptors are expressed in the human brain throughout development. To analyze the role of EPO in the GM-IVH, we have administered intraventricular collagenase (Col) to P7 mice, as a model of GM-IVH of the preterm infant. After EPO treatment, we have characterized our animals in the short (14 days) and the long (70 days) term. In our hands, EPO treatment significantly limited brain atrophy and ventricle enlargement. EPO also restored neuronal density and ameliorated dendritic spine loss. Likewise, inflammation and small vessel bleeding were also reduced, resulting in the preservation of learning and memory abilities. Moreover, plasma gelsolin levels, as a feasible peripheral marker of GM-IVH-induced damage, recovered after EPO treatment. Altogether, our data support the positive effect of EPO treatment in our preclinical model of GM-IVH, both in the short and the long term.

10.
Alzheimers Res Ther ; 12(1): 40, 2020 04 07.
Article in English | MEDLINE | ID: mdl-32264944

ABSTRACT

BACKGROUND: Both Alzheimer's disease (AD) and type 2 diabetes (T2D) share common pathological features including inflammation, insulin signaling alterations, or vascular damage. AD has no successful treatment, and the close relationship between both diseases supports the study of antidiabetic drugs to limit or slow down brain pathology in AD. Empagliflozin (EMP) is a sodium-glucose co-transporter 2 inhibitor, the newest class of antidiabetic agents. EMP controls hyperglycemia and reduces cardiovascular comorbidities and deaths associated to T2D. Therefore, we have analyzed the role of EMP at the central level in a complex mouse model of AD-T2D. METHODS: We have treated AD-T2D mice (APP/PS1xdb/db mice) with EMP 10 mg/kg for 22 weeks. Glucose, insulin, and body weight were monthly assessed. We analyzed learning and memory in the Morris water maze and the new object discrimination test. Postmortem brain assessment was conducted to measure brain atrophy, senile plaques, and amyloid-ß levels. Tau phosphorylation, hemorrhage burden, and microglia were also measured in the brain after EMP treatment. RESULTS: EMP treatment helped to maintain insulin levels in diabetic mice. At the central level, EMP limited cortical thinning and reduced neuronal loss in treated mice. Hemorrhage and microglia burdens were also reduced in EMP-treated mice. Senile plaque burden was lower, and these effects were accompanied by an amelioration of cognitive deficits in APP/PS1xdb/db mice. CONCLUSIONS: Altogether, our data support a feasible role for EMP to reduce brain complications associated to AD and T2D, including classical pathological features and vascular disease, and supporting further assessment of EMP at the central level.


Subject(s)
Alzheimer Disease , Benzhydryl Compounds , Cognitive Dysfunction , Diabetes Mellitus, Experimental , Diabetes Mellitus, Type 2 , Glucosides , Alzheimer Disease/complications , Alzheimer Disease/drug therapy , Amyloid beta-Peptides/metabolism , Amyloid beta-Protein Precursor/metabolism , Animals , Benzhydryl Compounds/therapeutic use , Brain/metabolism , Cognitive Dysfunction/drug therapy , Diabetes Mellitus, Type 2/complications , Diabetes Mellitus, Type 2/drug therapy , Disease Models, Animal , Female , Glucosides/therapeutic use , Male , Mice , Mice, Transgenic
11.
J Neurochem ; 154(6): 673-692, 2020 09.
Article in English | MEDLINE | ID: mdl-32068886

ABSTRACT

The classic neuropathological features of Alzheimer's disease (AD) are accompanied by other complications, including alterations in adult cell proliferation and neurogenesis. Moreover recent studies have shown that traditional markers of the neurogenic process, such as doublecortin (DCX), may also be expressed in CD8+ T cells and ionized calcium-binding adaptor molecule 1 (Iba1+ ) microglia, in the close proximity to senile plaques, increasing the complexity of the condition. Altered glucose tolerance, observed in metabolic alteratioins, may accelerate the neurodegenerative process and interfere with normal adult cell proliferation and neurogenesis. To further explore the role of metabolic disease in AD, we analyzed cell proliferation and neurogenesis using 5'-bromo-2'-deoxyuridine and DCX immunohistochemistry in three different mouse models of AD and metabolic alterations: APP/PS1xdb/db mice, APP/PS1 mice on a long-term high-fat diet, and APP/PS1 mice treated with streptozotozin. As reported previously, an overall reduction in cell proliferation and neurogenesis was observed after streptozotocin administration. In contrast, an increase in cell proliferation and neurogenesis was detected in neurogenic niches in 14- and 26-week-old APP/PS1xdb/db mice, accompanied by a slight increase in cortical cell proliferation. While a similar trend was observed in animals on a high-fat diet, differences were not statistically significant. We observed very few DCX+ /CD8+ cells and no DCX+ /Iba1+ cells were observed in the close proximity to senile plaques in any of the groups. Interestingly, metabolic parameters such as body weight and glucose and insulin levels were identified as reliable predictors of cell proliferation and neurogenesis in APP/PS1xdb/db mice. Furthermore, metabolic parameters were also associated with altered Aß levels in the cortex and hippocampus of APP/PS1xdb/db mice. Altogether, our data suggest that metabolic disease may also interfere with central complications in AD.


Subject(s)
Alzheimer Disease/pathology , Cell Proliferation , Diabetes Mellitus, Type 2/complications , Diabetes Mellitus, Type 2/pathology , Neurogenesis , Alzheimer Disease/genetics , Amyloid beta-Peptides/metabolism , Amyloid beta-Protein Precursor/genetics , Animals , Bromodeoxyuridine/pharmacology , CD8 Antigens/genetics , CD8 Antigens/metabolism , Calcium-Binding Proteins/genetics , Calcium-Binding Proteins/metabolism , Diabetes Mellitus, Experimental/metabolism , Diet, High-Fat , Doublecortin Domain Proteins , Doublecortin Protein , Female , Male , Mice , Mice, Transgenic , Microfilament Proteins/genetics , Microfilament Proteins/metabolism , Microtubule-Associated Proteins/metabolism , Neuropeptides/metabolism , Oligopeptides/genetics , Peptide Fragments/metabolism
12.
Neurobiol Aging ; 36(2): 1057-71, 2015 Feb.
Article in English | MEDLINE | ID: mdl-25534236

ABSTRACT

Imbalances between excitatory and inhibitory transmissions in the brain anticipate the neuronal damage and death that occur in the neurodegenerative diseases like Alzheimer's disease (AD). We previously showed that amyloid-ß (Aß), a natural peptide involved in the onset and development of AD, counteracts the neurotrophic activity of the nerve growth factor (NGF) by dampening the γ-aminobutyric acid (GABA)ergic connectivity of cultured hippocampal neurons. Neuronal plasticity is partly controlled by the NGF-promoted expression of the homologue of enhancer-of-split 1 (Hes1), a transcription factor that regulates the formation of GABAergic synapses. We now show that Hes1 controls the expression of cerebellin 4 (Cbln4), a member of a small family of secreted synaptic proteins, and we present the evidence that Cbln4 plays an essential role in the formation and maintenance of inhibitory GABAergic connections. Cbln4 immunoreactivity was found in the hippocampus, mostly in the dendrites and somata of pyramidal neurons. In the CA1, the hippocampal region where the first neurons degenerate in AD, Cbln4 immunoreactivity was associated with GABAergic synapses (detected by vesicular inhibitory amino acid transporter [VGAT] immunostaining), which appear to surround and embrace the somata of CA1 pyramidal neurons (basket cells). Moreover, significant decreases of Hes1, Cbln4, and VGAT immunoreactivities and messenger RNA expression were found in the hippocampus of a mouse model of AD. We also found that either the overexpression of Cbln4 in cultured hippocampal neurons or the application of recombinant Cbln4 to the cultures increased the number of GABAergic varicosities, rescuing neurons from Aß-induced death. In contrast, knockdown of Cbln4 gene in cultured neurons was followed by a large reduction of GABAergic connections. Such an effect was reverted by exogenously added Cbln4. These findings suggest a therapeutic potential for Cbln4 in the treatment of AD.


Subject(s)
Alzheimer Disease/genetics , Amyloid beta-Peptides/toxicity , GABAergic Neurons/pathology , Nerve Tissue Proteins/physiology , Protein Precursors/physiology , Alzheimer Disease/therapy , Animals , Basic Helix-Loop-Helix Transcription Factors/physiology , CA1 Region, Hippocampal/cytology , Cells, Cultured , GABAergic Neurons/physiology , Gene Expression Regulation/genetics , Homeodomain Proteins/physiology , Humans , Mice, Inbred C57BL , Mice, Transgenic , Molecular Targeted Therapy , Neuronal Plasticity/genetics , Transcription Factor HES-1
13.
Endocrinol Nutr ; 58(2): 84-9, 2011 Feb.
Article in Spanish | MEDLINE | ID: mdl-21339088

ABSTRACT

The rare hereditary syndrome, multiple endocrine neoplasia type1 (MEN-1), is known to predispose affected individuals to endocrine neoplasms in a variety of tissues such as the parathyroid glands, the pituitary gland and the gastrointestinal tract. We describe the case of a man with traditionally-described manifestations (hyperparathyroidism and gastrinoma) and with other tumoral lesions arising from endocrine cells (insulinoma, gastric carcinoid, adrenal adenoma and pancreatic non-functioning neuroendocrine tumors) and non-endocrine cells (lipoma and collagenoma). Frequent recurrences in susceptible tissues that are not totally removed (as occurs in hyperparathyroidism and duodenal gastrinoma) and their unknown clinical significance have aroused current controversies in the therapeutic management of these entities, which is briefly reviewed.


Subject(s)
Multiple Endocrine Neoplasia Type 1/pathology , Adenoma/genetics , Adrenal Gland Neoplasms/genetics , Adult , Carcinoid Tumor/genetics , Combined Modality Therapy , Duodenal Neoplasms/drug therapy , Duodenal Neoplasms/genetics , Duodenal Neoplasms/surgery , Fibroma/genetics , Gastrinoma/drug therapy , Gastrinoma/genetics , Gastrinoma/surgery , Humans , Hyperparathyroidism/drug therapy , Hyperparathyroidism/genetics , Insulinoma/genetics , Lipoma/genetics , Lymphatic Metastasis , Male , Multiple Endocrine Neoplasia Type 1/drug therapy , Neuroendocrine Tumors/drug therapy , Neuroendocrine Tumors/genetics , Octreotide/therapeutic use , Omeprazole/therapeutic use , Pancreatic Neoplasms/genetics , Pancreatic Neoplasms/surgery , Skin Neoplasms/genetics , Stomach Neoplasms/genetics
14.
Endocrinol. nutr. (Ed. impr.) ; 58(2): 84-89, feb. 2011. tab
Article in Spanish | IBECS | ID: ibc-89539

ABSTRACT

La neoplasia endocrina múltiple de tipo 1 (MEN1) es un síndrome hereditario raro conocido por la predisposición a la aparición de neoplasias endocrinas en distintos tejidos como paratiroides, hipófisis y tracto gastrointestinal. Se presenta el caso de un varón en el que además de manifestaciones tradicionalmente descritas (hipeparatiroidismo y gastrinoma) se objetivan otras lesiones tumorales procedentes de células de estirpe endocrinológica (insulinoma, carcinoide gástrico, adenoma suprarrenal, tumores neuroendocrino no funcionantes del páncreas) y no endocrinológica (lipoma y colagenoma). La frecuente recurrencia de las lesiones sobre los tejidos susceptibles no resecados en su totalidad (como en el caso del hiperparatiroidismo y del gastrinoma duodenal) y las dudas sobre su significado clínico en el MEN1 suscitan cierta controversia en la actualidad sobre las recomendaciones en el manejo terapéutico de dichas lesiones que se revisa brevemente (AU)


The rare hereditary syndrome, multiple endocrine neoplasia type1 (MEN-1), is known to predispose affected individuals to endocrine neoplasms in a variety of tissues such as the parathyroid glands, the pituitary gland and the gastrointestinal tract. We describe the case of a man with traditionally-described manifestations (hyperparathyroidism and gastrinoma)and with other tumoral lesions arising from endocrine cells (insulinoma, gastric carcinoid,adrenal adenoma and pancreatic non-functioning neuroendocrine tumors) and non-endocrinecells (lipoma and collagenoma). Frequent recurrences in susceptible tissues that are not totally removed (as occurs in hyperparathyroidism and duodenal gastrinoma) and their unknown clinical significance have aroused current controversies in the therapeutic management of these entities, which is briefly reviewed (AU)


Subject(s)
Humans , Male , Adult , Multiple Endocrine Neoplasia Type 1/pathology , Adenoma , Combined Modality Therapy , Fibroma/genetics , Gastrinoma/drug therapy , Gastrinoma/genetics , Gastrinoma/surgery , Hyperparathyroidism/drug therapy , Hyperparathyroidism/genetics , Insulinoma/genetics , Lipoma/genetics , Lymphatic Metastasis , Multiple Endocrine Neoplasia Type 1/drug therapy , Octreotide/therapeutic use , Omeprazole/therapeutic use
15.
Endocrinol Nutr ; 56(7): 361-8, 2009.
Article in Spanish | MEDLINE | ID: mdl-19883896

ABSTRACT

OBJECTIVE: To identify possible risk factors associated with persistent disease 5 years after total or near-total thyroidectomy in patients with differentiated thyroid cancer (DTC). PATIENTS AND METHOD: Retrospective study evaluating data from 63 patients 5 years after they were first diagnosed of DTC. At this time of the study, 46 subjects were considered disease-free (F group) whereas 17 had evidence of persistent disease or had died from DTC (P group). We compared both groups of patients regarding the following variables: a) variables at diagnosis related to the patient (age, gender) and the tumor (histological type, size, extrathyroidal involvement, vascular invasion, multifocality, lymph node and distant metastases), and b) variables recorded during follow-up: percentage of subjects showing serum stimulated thyroglobulin > or = 10 ng/ml few weeks postoperatively (Tg0) and 6 to 12 months later (Tg1). RESULTS: Male gender, extrathyroidal involvement and lymph node metastases were more frequent in P group than in F group (41 vs. 11%, 60 vs. 18% and 50 vs. 5.5%; p < 0.05). During the follow-up the percentage of patients showing Tg > or = 10 ng/ml was higher in P group compared to F group, both at a few weeks postoperatively and 6 to 12 months later (Tg0, 75 vs. 13%; Tg1, 69% vs. 0; p < 0,05). CONCLUSIONS: In our patients, male gender, extrathyroidal involvement, and lymph node metastases at diagnosis were associated with persistent disease 5 years later. Serum stimulated thyroglobulin had a very high predictive value both just after surgery and in the next 6 to 12 months and could help identifying subjects who need a closer follow-up.


Subject(s)
Neoplasm Recurrence, Local/epidemiology , Thyroid Neoplasms/epidemiology , Thyroid Neoplasms/surgery , Thyroidectomy , Adult , Female , Humans , Male , Retrospective Studies , Risk Factors , Thyroid Neoplasms/diagnosis , Time Factors
16.
Endocrinol. nutr. (Ed. impr.) ; 56(7): 361-368, ago.-sept. 2009. tab
Article in Spanish | IBECS | ID: ibc-76922

ABSTRACT

Objetivo: Identificar en el cáncer diferenciado de tiroides (CDT) factores asociados a la persistencia de enfermedad a los 5 años de la tiroidectomía total o casi total. Pacientes y método: Estudio retrospectivo en 63 pacientes con CDT, de los que se consideró libres de enfermedad a los 5 años a 46 (grupo L) y 17 presentaban enfermedad persistente al quinto año o habían fallecido por CDT (grupo P). Se compararon las siguientes variables entre estos grupos: a) al diagnóstico, relacionadas con el paciente (edad, sexo) y con el tumor (tipo histológico, tamaño, extensión extratiroidea, invasión vascular, multifocalidad, metástasis ganglionares y a distancia), y b) recogidas durante el seguimiento: proporción de pacientes con tiroglobulina (Tg) sérica estimulada ≥ 10 ng/ml tras la cirugía (Tg0) y entre los 6 y los 12 meses (Tg1). Resultados: Al diagnóstico, el sexo masculino, la extensión extratiroidea y las metástasis ganglionares resultaron más frecuentes en el grupo P (el 41 frente al 11%, el 60 frente al 18% y el 50 frente al 5,5% respectivamente; p < 0,05). Durante el seguimiento, el porcentaje de sujetos con Tg ≥ 10 ng/ml fue mayor en el grupo P (Tg0, el 75 frente al 13%; Tg1, el 69% frente a 0; p < 0,05). Conclusiones: En nuestro medio, el sexo masculino, la extensión extratiroidea y las metástasis ganglionares son factores asociados a la enfermedad persistente. El elevado valor predictivo de la Tg estimulada tras la cirugía y a los 6-12 meses ayuda a identificar a los pacientes que precisan de un seguimiento más estrecho (AU)


Objective: To identify possible risk factors associated with persistent disease 5 years after total or near-total thyroidectomy in patients with differentiated thyroid cancer (DTC). Patients and method: Retrospective study evaluating data from 63 patients 5 years after they were first diagnosed of DTC. At this time of the study, 46 subjects were considered disease-free (F group) whereas 17 had evidence of persistent disease or had died from DTC (P group). We compared both groups of patients regarding the following variables: a) variables at diagnosis related to the patient (age, gender) and the tumor (histological type, size, extrathyroidal involvement, vascular invasion, multifocality, lymph node and distant metastases), and b) variables recorded during follow-up: percentage of subjects showing serum stimulated thyroglobulin ≥ 10 ng/ml few weeks postoperatively (Tg0) and 6 to 12 months later (Tg1). Results: Male gender, extrathyroidal involvement and lymph node metastases were more frequent in P group than in F group (41 vs. 11%, 60 vs. 18% and 50 vs. 5.5%; p < 0.05). During the follow-up the percentage of patients showing Tg ≥ 10 ng/ml was higher in P group compared to F group, both at a few weeks postoperatively and 6 to 12 months later (Tg0, 75 vs. 13%; Tg1, 69% vs. 0; p < 0,05). Conclusions: In our patients, male gender, extrathyroidal involvement, and lymph node metastases at diagnosis were associated with persistent disease 5 years later. Serum stimulated thyroglobulin had a very high predictive value both just after surgery and in the next 6 to 12 months and could help identifying subjects who need a closer follow-up (AU)


Subject(s)
Humans , Male , Female , Adult , Thyroidectomy , Neoplasm Recurrence, Local/epidemiology , Thyroid Neoplasms/epidemiology , Retrospective Studies , Risk Factors , Thyroid Neoplasms/diagnosis , Time Factors
17.
Rev. diagn. biol ; 53(2): 55-60, abr. 2004. tab, graf
Article in Es | IBECS | ID: ibc-35268

ABSTRACT

Objetivos: Evaluar las alteraciones biológicas en pacientes diagnosticados de obesidad mórbida (OM) y la potencial reversibilidad de las mismas tras la pérdida de peso posterior a la cirugía bariátrica. Material y Métodos: Estudio retrospectivo de 97 pacientes OM intervenidos en nuestro centro. Se evalúan datos antropométricos y analíticos previo al by-pass y con seguimiento de 6,12 y 24 meses. Resultados: Previo a cirugía encontramos hiperglucemia, hiperinsulinemia, dislipemia, elevación de los niveles de PCR, TSH y cortisol. A los 6 meses tras el by-pass los niveles descienden y se mantienen durante el seguimiento. Discusión: La disminución de los niveles de insulina tras el bypass, coincide con la disminución de las alteraciones asociadas al síndrome metabólico, lo que nos indica que nuestros pacientes lo padecen. Conclusiones: El descenso del IMC, la consiguiente reducción de peso y la mejoría de parámetros bioquímicos, disminuye las comorbilidades y reduce la tasa de mortalidad de OM (AU)


Subject(s)
Adult , Female , Male , Middle Aged , Humans , Obesity, Morbid/surgery , Gastric Bypass , Metabolic Syndrome/physiopathology , Comorbidity/trends , Postoperative Period
SELECTION OF CITATIONS
SEARCH DETAIL
...