Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 89
Filtrar
1.
Mol Psychiatry ; 28(8): 3220-3230, 2023 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-37353586

RESUMEN

After decades of research in the neurobiology of IGF-I, its role as a prototypical neurotrophic factor is undisputed. However, many of its actions in the adult brain indicate that this growth factor is not only involved in brain development or in the response to injury. Following a three-layer assessment of its role in the central nervous system, we consider that at the cellular level, IGF-I is indeed a bona fide neurotrophic factor, modulating along ontogeny the generation and function of all the major types of brain cells, contributing to sculpt brain architecture and adaptive responses to damage. At the circuit level, IGF-I modulates neuronal excitability and synaptic plasticity at multiple sites, whereas at the system level, IGF-I intervenes in energy allocation, proteostasis, circadian cycles, mood, and cognition. Local and peripheral sources of brain IGF-I input contribute to a spatially restricted, compartmentalized, and timed modulation of brain activity. To better define these variety of actions, we consider IGF-I a modulator of brain states. This definition aims to reconcile all aspects of IGF-I neurobiology, and may provide a new conceptual framework in the design of future research on the actions of this multitasking neuromodulator in the brain.


Asunto(s)
Factor I del Crecimiento Similar a la Insulina , Neuroprotección , Humanos , Adulto , Factor I del Crecimiento Similar a la Insulina/metabolismo , Encéfalo/metabolismo , Sistema Nervioso Central/metabolismo , Factores de Crecimiento Nervioso/metabolismo
2.
Growth Horm IGF Res ; 45: 25-30, 2019 04.
Artículo en Inglés | MEDLINE | ID: mdl-30825797

RESUMEN

Insulin (IR) and insulin-like growth factor I (IGF-IR) receptors share structural homology and can form hybrid heterodimers. While different observations suggest that hybrid receptors are important in physiology and pathology, little is known about their function in the brain. To gain further insight into the role of IR/IGF-IR hybrids in this organ, we analyzed their cellular distribution in the mouse brain. We combined proximity ligation assays (PLA) for IR and IGF-IR, a technique that detects close protein-protein interactions, with immunocytochemistry for brain cell markers to identify IR/IGF-IR hybrids in the major types of brain cells. Intriguingly, while all the types of brain cells analyzed co-express both receptors, only neurons, astroglia, and microglia show readily detectable IR/IGF-IR hybrids. Hybrid PLA signal was negligible in brain endothelial cells and was absent in oligodendrocytes. Hybrids were comparatively more abundant in neurons and peaked after brain development was completed. Cell-specific expression and greater abundance in the adult brain suggests specialized actions of IR/IGF-IR hybrids in this organ, particularly in neurons.


Asunto(s)
Astrocitos/metabolismo , Encéfalo/metabolismo , Microglía/metabolismo , Neuronas/metabolismo , Receptor IGF Tipo 1/metabolismo , Receptor de Insulina/metabolismo , Animales , Astrocitos/citología , Encéfalo/citología , Células Cultivadas , Masculino , Ratones , Ratones Endogámicos C57BL , Microglía/citología , Neuronas/citología , Multimerización de Proteína , Receptor IGF Tipo 1/genética , Receptor de Insulina/genética
3.
Brain Plast ; 4(1): 3-15, 2018 Dec 12.
Artículo en Inglés | MEDLINE | ID: mdl-30564544

RESUMEN

The search for the cause of Alzheimer's disease (AD), that affects millions of people worldwide, is currently one of the most important scientific endeavors from a clinical perspective. There are so many mechanisms proposed, and so disparate changes observed, that it is becoming a challenging task to provide a comprehensive view of possible pathogenic processes in AD. Tauopathy (intracellular neurofibrillary tangles) and amyloidosis (extracellular amyloid plaques) are the anatomical hallmarks of the disease, and the formation of these proteinaceous aggregates in specific brain areas is widely held as the ultimate pathogenic mechanism. However, the triggers of this dysproteostasis process remain unknown. Further, neurofibrillary tangles and plaques may only constitute the last stages of a process of still uncertain origin. Thus, without an established knowledge of its etiology, and no cure in the horizon, prevention - or merely delaying its development, has become a last-resort goal in AD research. As with other success stories in preventive medicine, epidemiological studies have provided basic knowledge of risk factors in AD that may contribute to understand its etiology. Disregarding old age, gender, and ApoE4 genotype as non preventable risk factors, there are diverse life-style traits - many of them closely related to cardiovascular health, that have been associated to AD risk. Most prominent among them are diet, physical and mental activity, exposure to stress, and sleep/wake patterns. We argue that all these life-style factors engage insulinergic pathways that affect brain function, providing a potentially unifying thread for life-style and AD risk. Although further studies are needed to firmly establish a link between faulty insulinergic function and AD, we herein summarize the evidence that this link should be thoroughly considered.

4.
Cereb Cortex ; 28(6): 2007-2014, 2018 06 01.
Artículo en Inglés | MEDLINE | ID: mdl-28449086

RESUMEN

In response to injury, the brain produces different neuroprotective molecules, such as insulin-like growth factor I (IGF-I). However, IGF-I is also taken up by the brain from the circulation in response to physiological stimuli. Herein, we analyzed in mice the relative contribution of circulating and locally produced IGF-I to increased brain IGF-I levels after insult. Traumatic brain injury (TBI) induced by a controlled impact resulted in increased IGF-I levels in the vicinity of the lesion, but mice with low serum IGF-I showed significantly lower increases. Indeed, in normal mice, peripheral IGF-I accumulated at the lesion site after injury, and at the same time serum IGF-I levels decreased. Collectively, these data suggest that serum IGF-I enter into the brain after TBI and contributes to increased brain IGF-I levels at the injury site. This connection between central and circulating IGF-I provides an amenable route for treatment, as subcutaneous administration of IGF-I to TBI mice led to functional recovery. These latter results add further support to the use of systemic IGF-I or its mimetics for treatment of brain injuries.


Asunto(s)
Lesiones Traumáticas del Encéfalo/metabolismo , Factor I del Crecimiento Similar a la Insulina/metabolismo , Animales , Masculino , Ratones , Ratones Endogámicos C57BL
5.
Mol Cell Neurosci ; 80: 100-110, 2017 04.
Artículo en Inglés | MEDLINE | ID: mdl-28286293

RESUMEN

Inherited neurodegenerative diseases such as Friedreich's ataxia (FRDA), produced by deficiency of the mitochondrial chaperone frataxin (Fxn), shows specific neurological deficits involving different subset of neurons even though deficiency of Fxn is ubiquitous. Because astrocytes are involved in neurodegeneration, we analyzed whether they are also affected by frataxin deficiency and contribute to the disease. We also tested whether insulin-like growth factor I (IGF-I), that has proven effective in increasing frataxin levels both in neurons and in astrocytes, also exerts in vivo protective actions. Using the GFAP promoter expressed by multipotential stem cells during development and mostly by astrocytes in the adult, we ablated Fxn in a time-dependent manner in mice (FGKO mice) and found severe ataxia and early death when Fxn was eliminated during development, but not when deleted in the adult. Analysis of underlying mechanisms revealed that Fxn deficiency elicited growth and survival impairments in developing cerebellar astrocytes, whereas forebrain astrocytes grew normally. A similar time-dependent effect of frataxin deficiency in astrocytes was observed in a fly model. In addition, treatment of FGKO mice with IGF-I improved their motor performance, reduced cerebellar atrophy, and increased survival. These observations indicate that a greater vulnerability of developing cerebellar astrocytes to Fxn deficiency may contribute to cerebellar deficits in this inherited disease. Our data also confirm a therapeutic benefit of IGF-I in early FRDA deficiency.


Asunto(s)
Astrocitos/efectos de los fármacos , Astrocitos/fisiología , Cerebelo/citología , Ataxia de Friedreich/tratamiento farmacológico , Factor I del Crecimiento Similar a la Insulina/uso terapéutico , Proteínas de Unión a Hierro/metabolismo , Animales , Animales Recién Nacidos , Peso Corporal/efectos de los fármacos , Peso Corporal/genética , Calbindinas/metabolismo , Modelos Animales de Enfermedad , Drosophila , Proteínas de Drosophila/genética , Proteínas de Drosophila/metabolismo , Ataxia de Friedreich/complicaciones , Ataxia de Friedreich/genética , Ataxia de Friedreich/patología , Proteína Ácida Fibrilar de la Glía/metabolismo , Humanos , Factor I del Crecimiento Similar a la Insulina/farmacología , Proteínas de Unión a Hierro/genética , Ratones , Ratones Endogámicos C57BL , Fosfopiruvato Hidratasa/metabolismo , Prosencéfalo/citología , Trastornos Psicomotores/etiología , Trastornos Psicomotores/prevención & control , Especies Reactivas de Oxígeno/metabolismo , Receptor IGF Tipo 1/genética , Receptor IGF Tipo 1/metabolismo , Frataxina
6.
Prog Brain Res ; 225: 243-68, 2016.
Artículo en Inglés | MEDLINE | ID: mdl-27130419

RESUMEN

Aging impairs cerebrovascular plasticity and subsequently leads cerebral hypoperfusion, which synergistically accelerates aging-associated cognitive dysfunction and neurodegenerative diseases associated with impaired neuronal plasticity. On the other hand, over two decades of researches have successfully demonstrated that exercise, or higher level of physical activity, is a powerful and nonpharmacological approach to improve brain function. Most of the studies have focused on the neuronal aspects and found that exercise triggers improvements in neuronal plasticity, such as neurogenesis; however, exercise can improve cerebrovascular plasticity as well. In this chapter, to understand these beneficial effects of exercise on the cerebral vasculature, we first discuss the issue of changes in cerebral blood flow and its regulation during acute bouts of exercise. Then, how regular exercise improves cerebrovascular plasticity will be discussed. In addition, to shed light on the importance of understanding interactions between the neuron and cerebral vasculature, we describe neuronal activity-driven uptake of circulating IGF-I into the brain.


Asunto(s)
Circulación Cerebrovascular/fisiología , Ejercicio Físico/fisiología , Neovascularización Fisiológica/fisiología , Plasticidad Neuronal/fisiología , Envejecimiento/fisiología , Animales , Encéfalo/citología , Humanos , Factor I del Crecimiento Similar a la Insulina/metabolismo
7.
Oncogene ; 33(32): 4114-22, 2014 Aug 07.
Artículo en Inglés | MEDLINE | ID: mdl-24056966

RESUMEN

Loss-of-function mutations in the phosphatase PTEN (phosphatase and tensin homolog deleted on chromosome10) contribute to aberrant cell growth in part through upregulation of the mitogenic IGF-1/PI3K/Akt pathway. In turn, this pathway exerts a homeostatic feedback over PTEN. Using mutagenesis analysis to explore a possible impact of this mutual control on astrocyte growth, we found that truncation of the C-terminal region of PTEN (Δ51) associates with a marked increase in NFκB activity, a transcription factor overactivated in astrocyte tumors. Whereas mutations of PTEN are considered to lead to a loss-of-function, PTENΔ51, a truncation that comprises a region frequently mutated in human gliomas, displayed a neomorphic (gain-of-function) activity that was independent of its phosphatase activity. This gain-of-function of PTENΔ51 includes stimulation of IGF-1 synthesis through protein kinase A activation of the IGF-1 promoter. Increased IGF-1 originates an autocrine loop that activates Akt and NFκB. Constitutive activation of NFκB in PTENΔ51-expressing astrocytes leads to aberrant cell growth; astrocytes expressing this mutant PTEN generate colonies in vitro and tumors in vivo. Mutations converting a tumor suppressor such as PTEN into a tumor promoter through a gain-of-function involving IGF-1 production may further our understanding of the role played by this growth factor in glioma growth and help us define druggable targets for personalized therapy.


Asunto(s)
Astrocitos/citología , Regulación Neoplásica de la Expresión Génica , Factor I del Crecimiento Similar a la Insulina/metabolismo , Mutación , Fosfohidrolasa PTEN/genética , Fosfohidrolasa PTEN/fisiología , Animales , Línea Celular Tumoral , Proliferación Celular , Proteínas Quinasas Dependientes de AMP Cíclico/metabolismo , Glioma/metabolismo , Humanos , Ratones , Ratones SCID , FN-kappa B/metabolismo , Trasplante de Neoplasias , Estructura Terciaria de Proteína , Especies Reactivas de Oxígeno/metabolismo
8.
Transl Psychiatry ; 3: e330, 2013 Dec 03.
Artículo en Inglés | MEDLINE | ID: mdl-24301648

RESUMEN

Circulating insulin-like growth factor I (IGF-I) enters the brain and promotes clearance of amyloid peptides known to accumulate in Alzheimer's disease (AD) brains. Both patients and mouse models of AD show decreased level of circulating IGF-I enter the brain as evidenced by a lower ratio of cerebrospinal fluid/plasma IGF-I. Importantly, in presymptomatic AD mice this reduction is already manifested as a decreased brain input of serum IGF-I in response to environmental enrichment. To explore a potential diagnostic use of this early loss of IGF-I input, we monitored electrocorticogram (ECG) responses to systemic IGF-I in mice. Whereas control mice showed enhanced ECG activity after IGF-I, presymptomatic AD mice showed blunted ECG responses. Because nonhuman primates showed identically enhanced electroencephalogram (EEG) activity in response to systemic IGF-I, loss of the EEG signature of serum IGF-I may be exploited as a disease biomarker in AD patients.


Asunto(s)
Enfermedad de Alzheimer/diagnóstico , Encéfalo/efectos de los fármacos , Electroencefalografía/efectos de los fármacos , Factor I del Crecimiento Similar a la Insulina/farmacología , Enfermedad de Alzheimer/sangre , Enfermedad de Alzheimer/líquido cefalorraquídeo , Animales , Encéfalo/metabolismo , Estudios de Casos y Controles , Modelos Animales de Enfermedad , Diagnóstico Precoz , Humanos , Factor I del Crecimiento Similar a la Insulina/líquido cefalorraquídeo , Factor I del Crecimiento Similar a la Insulina/metabolismo , Macaca , Ratones
9.
Neurosci Lett ; 509(2): 101-4, 2012 Feb 16.
Artículo en Inglés | MEDLINE | ID: mdl-22230888

RESUMEN

The influence of insulin-like growth factor I (IGF-I) on the progression of Alzheimer's disease (AD) is discussed controversially. To help clarify the role of this circulating neurotrophic factor in brain amyloidosis, the major pathological trait in AD, we analyzed plaque formation in a mouse model of AD transgenic for human APP and PS1 mutations with reduced serum IGF-I levels (LIDAD mice). We found that brain amyloidosis in LIDAD mice appeared earlier than in AD mice, at 2 months of age, while attained comparable levels at 6 months. In parallel, early microgliosis was observed in LIDAD mice also at 2 months and remained exacerbated at 6 months. Collectively, these observations suggest a role of serum IGF-I in delaying early brain amyloidosis.


Asunto(s)
Precursor de Proteína beta-Amiloide/metabolismo , Amiloidosis/metabolismo , Encéfalo/metabolismo , Encéfalo/patología , Factor I del Crecimiento Similar a la Insulina/deficiencia , Presenilina-1/metabolismo , Edad de Inicio , Envejecimiento/metabolismo , Enfermedad de Alzheimer/sangre , Enfermedad de Alzheimer/genética , Enfermedad de Alzheimer/metabolismo , Precursor de Proteína beta-Amiloide/deficiencia , Precursor de Proteína beta-Amiloide/genética , Amiloidosis/sangre , Animales , Femenino , Gliosis/sangre , Gliosis/metabolismo , Humanos , Inflamación/sangre , Inflamación/metabolismo , Factor I del Crecimiento Similar a la Insulina/análisis , Masculino , Ratones , Ratones Transgénicos , Mutación , Presenilina-1/deficiencia , Presenilina-1/genética , Factores de Tiempo
10.
Mol Psychiatry ; 17(7): 705-18, 2012 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-22005929

RESUMEN

Whether insulin-like growth factor I (IGF-I) signaling in Alzheimer's disease (AD) is beneficial or detrimental remains controversial. We now show that a competitive regulation by IGF-I of the phosphatase calcineurin in reactive, but not in quiescent astrocytes drives Alzheimer's pathology. Calcineurin de-phosphorylates the transcription factor Foxo3 in response to tumor necrosis factor-α (TNFα), an inflammatory cytokine increased in AD, activating nuclear factor-κB (NFκB) inflammatory signaling in astrocytes. In turn, IGF-I inactivates and displaces Foxo3 from calcineurin in TNFα-stimulated astrocytes by recruiting the transcription factor peroxisome proliferator-activated receptor-γ, and NFκB signaling is inhibited. This antagonistic mechanism reversibly drives the course of the disease in AD mice, even at advanced stages. As hallmarks of this calcineurin/Foxo3/NFκB pathway are present in human AD brains, treatment with IGF-I may be beneficial by antagonizing it.


Asunto(s)
Enfermedad de Alzheimer/enzimología , Enfermedad de Alzheimer/patología , Astrocitos/patología , Calcineurina/fisiología , Factor I del Crecimiento Similar a la Insulina/fisiología , Placa Amiloide/patología , Transducción de Señal/fisiología , Enfermedad de Alzheimer/metabolismo , Animales , Astrocitos/efectos de los fármacos , Astrocitos/metabolismo , Encéfalo/efectos de los fármacos , Encéfalo/enzimología , Encéfalo/metabolismo , Encéfalo/patología , Inhibidores de la Calcineurina , Células Cultivadas , Modelos Animales de Enfermedad , Proteína Forkhead Box O3 , Factores de Transcripción Forkhead/metabolismo , Factor I del Crecimiento Similar a la Insulina/farmacología , Aprendizaje por Laberinto/fisiología , Ratones , Ratones Transgénicos , FN-kappa B/antagonistas & inhibidores , FN-kappa B/metabolismo , Receptores Activados del Proliferador del Peroxisoma/metabolismo , Fosforilación , Reconocimiento en Psicología/fisiología , Transducción de Señal/efectos de los fármacos , Factor de Necrosis Tumoral alfa/farmacología , Factor de Necrosis Tumoral alfa/fisiología
11.
Exp Gerontol ; 46(2-3): 96-9, 2011.
Artículo en Inglés | MEDLINE | ID: mdl-20863877

RESUMEN

Insulin-like growth factors (IGFs) are important modulators of organismal life-span all along phylogeny. These growth factors are widely viewed as detrimental for long life by reducing tissue resistance to oxidative stress. However, IGF-I has been consistently shown to be a potent neuroprotective factor in mammals, and as such, a deterrent of brain aging. Conversely, recent data suggest that IGF-I may contribute to amyloid neurodegeneration underlying Alzheimer's disease. These opposing observations underline an incomplete understanding of the significance of this ancestral hormone pathway in relation to brain aging. It is possible that these opposite results are the consequence of using different experimental approaches. Thus, brain amyloid injury is reduced in mutant mice partially defective in IGF-I receptor function, whereas IGF-I is neuroprotective when administered to animal models of neurodegenerative disease or normal brain aging. This approach-dependent effect of IGF-I highlights a fundamental gap in our knowledge of the relationship between peripheral and brain IGF-I function and the actual biological impact of experimental modulation of brain IGF-I function. We suggest to directly address brain IGF-I function in the varying experimental approaches used to confirm that changes have taken place in the desired way.


Asunto(s)
Envejecimiento/fisiología , Encéfalo/fisiología , Factor I del Crecimiento Similar a la Insulina/fisiología , Enfermedad de Alzheimer/metabolismo , Animales , Humanos
12.
Neuroscience ; 160(2): 402-11, 2009 May 05.
Artículo en Inglés | MEDLINE | ID: mdl-19264110

RESUMEN

Peripheral and central diabetic neuropathies were studied in streptozotocin-diabetic rats, using behavioral, biochemical and electrophysiological techniques. Diabetic rats showed thermal hypoalgesia and decreasing motor nerve conduction velocity at 4 and 8 weeks of diabetes. In addition, amplitude of the evoked potential recorded in primary somatosensory cortex after stimulation of the sciatic nerve was markedly reduced at 8 weeks of diabetes. This decrease was accompanied by decreases in GluR2/3 AMPA receptor subunits. These changes seem to be specific to the somatosensory system and to originate in higher centers since they were not present in the hippocampus and were not observed at the level of gracilis nucleus. Insulin-like growth factor I (IGF-I) treatment reversed the reduced thermal sensitivity and peripheral nerve conduction velocity but did not reverse changes in the CNS, suggesting that once initiated, both anomalies may develop independently in this model of diabetic neuropathy. In conclusion, the results indicate that diabetes induces a wide spectrum of alterations in the central somatosensory system that are independent of the decreases in peripheral sensory transmission that could be responsible for the disturbances in somatosensory perception observed in diabetes.


Asunto(s)
Diabetes Mellitus Experimental/fisiopatología , Neuropatías Diabéticas/fisiopatología , Conducción Nerviosa/fisiología , Umbral del Dolor/fisiología , Corteza Somatosensorial/fisiopatología , Trastornos Somatosensoriales/fisiopatología , Animales , Neuropatías Diabéticas/complicaciones , Modelos Animales de Enfermedad , Potenciales Evocados Somatosensoriales/fisiología , Factor I del Crecimiento Similar a la Insulina/fisiología , Masculino , Neuronas Motoras/fisiología , Sistema Nervioso Periférico/fisiopatología , Ratas , Ratas Wistar , Receptores AMPA/fisiología , Trastornos Somatosensoriales/complicaciones
13.
Endocrinology ; 149(12): 5952-7, 2008 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-18818286

RESUMEN

It is lay knowledge now that Alzheimer's dementia (AD) is one of the most devastating diseases afflicting our societies. A major thrust in search for a cure has relied in the development of animal models of the disease. Thanks to progress in the genetics of the rare inherited forms of AD, various transgenic mouse models harboring human mutated proteins were developed, yielding very significant advancements in the understanding of pathological pathways. Although these models led to testing many different new therapies, none of the preclinical successes have translated yet into much needed therapeutic improvements. Further insight into the metabolic disturbances that are probably associated with the onset of the disease may also rely on new animal models of AD involving insulin/IGF-I signaling that could mimic the far most common sporadic forms of AD associated with old age. Combination of models of familial AD that develop severe amyloidosis with those displaying defects in insulin/IGF-I signaling may help clarify the link between putative initial metabolic disturbances and mechanisms of pathological progression.


Asunto(s)
Enfermedad de Alzheimer/metabolismo , Demencia/metabolismo , Modelos Animales de Enfermedad , Envejecimiento/fisiología , Enfermedad de Alzheimer/genética , Enfermedad de Alzheimer/fisiopatología , Amiloidosis/metabolismo , Amiloidosis/fisiopatología , Animales , Demencia/genética , Demencia/fisiopatología , Humanos , Insulina/metabolismo , Factor I del Crecimiento Similar a la Insulina/metabolismo , Modelos Biológicos
14.
Neuromolecular Med ; 10(2): 99-107, 2008.
Artículo en Inglés | MEDLINE | ID: mdl-18286390

RESUMEN

Physical exercise has long been recognized as highly beneficial for brain and body health. The molecular mechanisms responsible for translation of exercise stimuli in the brain have claimed attention due to mounting evidence for the neuroprotective actions of the exercise and its positive effects in preventing both ageing and neurodegenerative disease. These molecular mediators are currently under investigation with new tools able to yield deep insights into the neurobiology of exercise. In the present work we focus on the evidence pertaining to the mediation of exercise effects by insulin-like growth factor 1 (IGF1), as recent reports suggest that this growth factor shows brain area-specific, temporal rank-sensitive, and behavioural task-dependent features in response to exercise.


Asunto(s)
Encéfalo/metabolismo , Péptidos y Proteínas de Señalización Intercelular/metabolismo , Condicionamiento Físico Animal/fisiología , Esfuerzo Físico/fisiología , Animales , Hipocampo/metabolismo , Humanos , Factor I del Crecimiento Similar a la Insulina/metabolismo , Aprendizaje/fisiología , Plasticidad Neuronal/fisiología , Células Madre/metabolismo
15.
Mol Cell Neurosci ; 37(2): 402-11, 2008 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-18086533

RESUMEN

Knowledge about the effects of physical exercise on brain is accumulating although the mechanisms through which exercise exerts these actions remain largely unknown. A possible involvement of adult hippocampal neurogenesis (AHN) in the effects of exercise is debated while the physiological and pathological significance of AHN is under intense scrutiny. Recently, both neurogenesis-dependent and independent mechanisms have been shown to mediate the effects of physical exercise on spatial learning and anxiety-like behaviors. Taking advantage that the stimulating effects of exercise on AHN depend among others, on serum insulin-like growth factor I (IGF-I), we now examined whether the behavioral effects of running exercise are related to variations in hippocampal neurogenesis, by either increasing or decreasing it according to serum IGF-I levels. Mutant mice with low levels of serum IGF-I (LID mice) had reduced AHN together with impaired spatial learning. These deficits were not improved by running. However, administration of exogenous IGF-I ameliorated the cognitive deficit and restored AHN in LID mice. We also examined the effect of exercise in LID mice in the novelty-suppressed feeding test, a measure of anxiety-like behavior in laboratory animals. Normal mice, but not LID mice, showed reduced anxiety after exercise in this test. However, after exercise, LID mice did show improvement in the forced swim test, a measure of behavioral despair. Thus, many, but not all of the beneficial effects of exercise on brain function depend on circulating levels of IGF-I and are associated to increased hippocampal neurogenesis, including improved cognition and reduced anxiety.


Asunto(s)
Trastornos de Ansiedad/genética , Hipocampo/metabolismo , Factor I del Crecimiento Similar a la Insulina/metabolismo , Trastornos de la Memoria/genética , Plasticidad Neuronal/fisiología , Condicionamiento Físico Animal , Animales , Trastornos de Ansiedad/tratamiento farmacológico , Trastornos de Ansiedad/fisiopatología , Proliferación Celular , Cognición/efectos de los fármacos , Cognición/fisiología , Trastorno Depresivo/tratamiento farmacológico , Trastorno Depresivo/genética , Trastorno Depresivo/fisiopatología , Terapia por Ejercicio , Factor I del Crecimiento Similar a la Insulina/genética , Factor I del Crecimiento Similar a la Insulina/farmacología , Aprendizaje/efectos de los fármacos , Aprendizaje/fisiología , Memoria/efectos de los fármacos , Memoria/fisiología , Trastornos de la Memoria/tratamiento farmacológico , Trastornos de la Memoria/fisiopatología , Ratones , Ratones Endogámicos C57BL , Ratones Mutantes , Neuronas/metabolismo , Células Madre/metabolismo , Natación/fisiología , Natación/psicología
16.
Mol Psychiatry ; 12(12): 1118-28, 2007 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-17848918

RESUMEN

Increasing evidence indicates that circulating insulin-like growth factor I (IGF-I) acts as a peripheral neuroactive signal participating not only in protection against injury but also in normal brain function. Epidemiological studies in humans as well as recent evidence in experimental animals suggest that blood-borne IGF-I may be involved in cognitive performance. In agreement with observations in humans, we found that mice with low-serum IGF-I levels due to liver-specific targeted disruption of the IGF-I gene presented cognitive deficits, as evidenced by impaired performance in a hippocampal-dependent spatial-recognition task. Mice with serum IGF-I deficiency also have disrupted long-term potentiation (LTP) in the hippocampus, but not in cortex. Impaired hippocampal LTP was associated with a reduction in the density of glutamatergic boutons that led to an imbalance in the glutamatergic/GABAergic synapse ratio in this brain area. Behavioral and synaptic deficits were ameliorated in serum IGF-I-deficient mice by prolonged systemic administration of IGF-I that normalized the density of glutamatergic boutons in the hippocampus. Altogether these results indicate that liver-derived circulating IGF-I affects crucial aspects of mature brain function; that is, learning and synaptic plasticity, through its trophic effects on central glutamatergic synapses. Declining levels of serum IGF-I during aging may therefore contribute to age-associated cognitive loss.


Asunto(s)
Encéfalo/metabolismo , Trastornos del Conocimiento/metabolismo , Trastornos del Conocimiento/fisiopatología , Factor I del Crecimiento Similar a la Insulina/metabolismo , Extractos Hepáticos/química , Factores de Edad , Animales , Conducta Animal , Encéfalo/anatomía & histología , Factor Neurotrófico Derivado del Encéfalo/metabolismo , Trastornos del Conocimiento/tratamiento farmacológico , Trastornos del Conocimiento/genética , Glutamato Descarboxilasa/metabolismo , Humanos , Factor I del Crecimiento Similar a la Insulina/genética , Factor I del Crecimiento Similar a la Insulina/uso terapéutico , Aprendizaje por Laberinto/fisiología , Ratones , Ratones Transgénicos , Proteína 1 de Transporte Vesicular de Glutamato/metabolismo
17.
Neuroscience ; 147(3): 631-8, 2007 Jul 13.
Artículo en Inglés | MEDLINE | ID: mdl-17570601

RESUMEN

A direct relation between the rate of adult hippocampal neurogenesis in mice and the immobility time in a forced swim test after living in an enriched environment has been suggested previously. In the present work, young adult mice living in an enriched environment for 2 months developed considerably more immature differentiating neurons (doublecortin-positive, DCX(+)) than control, non-enriched animals. Furthermore, we found that the more DCX(+) cells they possessed, the lower the immobility time they scored in the forced swim test. This DCX(+) subpopulation is composed of mostly differentiating dentate neurons independently of the birthdates of every individual cell. However, variations found in this subpopulation were not the result of a general effect on the survival of any newborn neuron in the granule cell layer, as 5-bromo-2-deoxyuridine (BrdU)-labeled cells born during a narrow time window included in the longer lifetime period of DCX(+) cells, were not significantly modified after enrichment. In contrast, the survival of the mature population of neurons in the granule cell layer of the dentate gyrus in enriched animals increased, although this did not influence their performance in the Porsolt test, nor did it influence the dentate gyrus volume or granule neuronal nuclei size. These results indicate that the population of immature, differentiating neurons in the adult hippocampus is one factor directly related to the protective effect of an enriched environment against a highly stressful event.


Asunto(s)
Giro Dentado/citología , Ambiente , Pérdida de Tono Postural/fisiología , Neuronas/fisiología , Natación , Animales , Conducta Animal , Bromodesoxiuridina/metabolismo , Recuento de Células/métodos , Proteínas de Dominio Doblecortina , Proteína Doblecortina , Femenino , Ratones , Ratones Endogámicos C57BL , Proteínas Asociadas a Microtúbulos/metabolismo , Neuropéptidos/metabolismo , Estadísticas no Paramétricas , Factores de Tiempo
18.
Growth Horm IGF Res ; 17(2): 89-95, 2007 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-17317256

RESUMEN

All tissues in the body are under the influence of insulin-like growth factor-I (IGF-I). Together with insulin, IGF-I is a key regulator of cell metabolism and growth. IGF-I also acts in the central nervous system, where it affects many different cell populations. In this brief review, we discuss the many roles of IGF-I in the adult brain, and present the idea that diseases affecting the brain will perturb IGF-I activity, although more refined studies at the molecular and cellular level are needed before we can firmly established this possibility. We also suggest that under normal physiological conditions IGF-I may play a significant role in higher brain functions underlying cognition, and may serve a homeostatic role during brain aging. Among newly emerging issues, the effects of IGF-I on non-neuronal cells within the nervous system and their impact in brain physiology and pathology are becoming very important in understanding the biology of this peptide in the brain.


Asunto(s)
Astrocitos/fisiología , Encéfalo/irrigación sanguínea , Encéfalo/fisiología , Factor I del Crecimiento Similar a la Insulina/fisiología , Adulto , Enfermedad de Alzheimer/metabolismo , Animales , Astrocitos/citología , Encéfalo/citología , Proliferación Celular , Endotelio Vascular/metabolismo , Humanos , Factor I del Crecimiento Similar a la Insulina/metabolismo , Ratones
19.
Hippocampus ; 16(5): 480-90, 2006.
Artículo en Inglés | MEDLINE | ID: mdl-16596582

RESUMEN

In the adult hippocampus, neurogenesis is influenced both by external stimuli, such as physical exercise, and by intrinsic conditions like age and disease. However, the way in which many of these external and internal cues interact in this process remains poorly understood. We have used a new, more precise, stereological cell counting method that involves confocal microscopy to analyze the effects of exercise on adult neurogenesis in the mouse. We found that treadmill exercise increases the number of differentiating neurons (doublecortin/calretinin cells) in the granule cell layer of the mouse hippocampus in a manner that is directly related to the size of the mature granule cell population. More immature neurons were found after exercise in animals that had a larger dentate gyrus (DG), while no changes were observed in those with a smaller DG. This differential response to physical exercise suggests that the pre-existing neuronal population regulates the neurogenic response in the DG to external stimuli. These data raise the possibility of anticipating an individuals' response to therapeutic interventions (like exercise) aimed at augmenting dentate neurogenesis and alleviating or preventing cognitive decline.


Asunto(s)
Hipocampo/citología , Hipocampo/fisiología , Neuronas/fisiología , Esfuerzo Físico/fisiología , Algoritmos , Animales , Recuento de Células , Gránulos Citoplasmáticos/fisiología , Giro Dentado/citología , Giro Dentado/fisiología , Hipocampo/crecimiento & desarrollo , Inmunohistoquímica , Masculino , Ratones , Ratones Endogámicos C57BL , Microscopía Confocal
20.
Neurobiol Aging ; 27(9): 1250-7, 2006 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-16183170

RESUMEN

Transgenic mice expressing mutant forms of both amyloid-beta (Abeta) precursor protein (APP) and presenilin (PS) 2 develop severe brain amyloidosis and cognitive deficits, two pathological hallmarks of Alzheimer's disease (AD). One-year-old APP/PS2 mice with high brain levels of Abeta and abundant Abeta plaques show disturbances in spatial learning and memory. Treatment of these deteriorated mice with a systemic slow-release formulation of insulin-like growth factor I (IGF-I) significantly ameliorated AD-like disturbances. Thus, IGF-I enhanced cognitive performance, decreased brain Abeta load, increased the levels of synaptic proteins, and reduced astrogliosis associated to Abeta plaques. The beneficial effects of IGF-I were associated to a significant increase in brain Abeta complexed to protein carriers such as albumin, apolipoprotein J or transthyretin. Since levels of APP were not modified after IGF-I therapy, and in vitro data showed that IGF-I increases the transport of Abeta/carrier protein complexes through the choroid plexus barrier, it seems that IGF-I favors elimination of Abeta from the brain, supporting a therapeutic use of this growth factor in AD.


Asunto(s)
Precursor de Proteína beta-Amiloide/genética , Amiloidosis/tratamiento farmacológico , Encefalopatías/tratamiento farmacológico , Factor I del Crecimiento Similar a la Insulina/uso terapéutico , Péptidos beta-Amiloides/metabolismo , Amiloidosis/complicaciones , Amiloidosis/fisiopatología , Análisis de Varianza , Animales , Conducta Animal/efectos de los fármacos , Barrera Hematoencefálica/efectos de los fármacos , Barrera Hematoencefálica/fisiopatología , Química Encefálica/efectos de los fármacos , Encefalopatías/complicaciones , Encefalopatías/fisiopatología , Trastornos del Conocimiento/tratamiento farmacológico , Trastornos del Conocimiento/etiología , Modelos Animales de Enfermedad , Inmunohistoquímica/métodos , Aprendizaje por Laberinto/efectos de los fármacos , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Fragmentos de Péptidos/metabolismo , Conducta Espacial/efectos de los fármacos
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...