Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 34
Filtrar
1.
Proc Natl Acad Sci U S A ; 120(29): e2302892120, 2023 07 18.
Artigo em Inglês | MEDLINE | ID: mdl-37428916

RESUMO

Although microglia possess the unique ability to migrate, whether mobility is evident in all microglia, is sex dependent, and what molecular mechanisms drive this, is not well understood in the adult brain. Using longitudinal in vivo two-photon imaging of sparsely labeled microglia, we find a relatively small population of microglia (~5%) are mobile under normal conditions. Following injury (microbleed), the fraction of mobile microglia increased in a sex-dependent manner, with male microglia migrating significantly greater distances toward the microbleed relative to their female counterparts. To understand the signaling pathways involved, we interrogated the role of interferon gamma (IFNγ). Our data show that in male mice, stimulating microglia with IFNγ promotes migration whereas inhibiting IFNγ receptor 1 signaling inhibits them. By contrast, female microglia were generally unaffected by these manipulations. These findings highlight the diversity of microglia migratory responses to injury, its dependence on sex and the signaling mechanisms that modulate this behavior.


Assuntos
Interferon gama , Microglia , Animais , Masculino , Feminino , Camundongos , Microglia/metabolismo , Interferon gama/metabolismo , Transdução de Sinais , Encéfalo/metabolismo , Hemorragia Cerebral/metabolismo
2.
J Neurosci ; 38(40): 8707-8722, 2018 10 03.
Artigo em Inglês | MEDLINE | ID: mdl-30201775

RESUMO

Microcirculatory damage is a common complication for those with vascular risk factors, such as diabetes. To resolve vascular insults, the brain's immune cells (microglia) must rapidly envelop the site of injury. Currently, it is unknown whether Type 1 diabetes, a condition associated with chronic immune system dysfunction, alters microglial responses to damage and what mechanisms are responsible. Using in vivo two-photon microscopy in adult male mice, we show that microglial envelopment of laser-induced cerebral microbleeds is diminished in a hyperglycemic mouse model of Type 1 diabetes, which could not be fully rescued with chronic insulin treatment. Microglia were important for vessel repair because reduced microglial accumulation in diabetic mice or near-complete depletion in healthy controls was associated with greater secondary leakage of the damaged vessel. Broadly suppressing inflammation with dexamethasone in diabetic mice but not healthy controls, significantly enhanced microglial responses to microbleeds and attenuated secondary vessel leakage. These enhancements were associated with changes in IFN-γ signaling because dexamethasone suppressed abnormally high levels of IFN-γ protein levels in brain and blood serum of diabetic mice. Further, blocking IFN-γ in diabetic mice with neutralizing antibodies restored normal microglial chemotaxic responses and purinoceptor P2ry12 gene expression, as well as mitigated secondary leakage. These results suggest that abnormal IFN-γ signaling disrupts microglial function in the diabetic brain, and that immunotherapies targeting IFN-γ can stimulate microglial repair of damaged vessels.SIGNIFICANCE STATEMENT Although Type 1 diabetes is an established risk factor for vascular complications, such as microbleeds, and is known to hinder wound healing in the body, no study has examined how diabetes impacts the brain's innate immune reparative response (involving cells called microglia) to vascular injury. Here we show that microglial responses to brain microbleeds were diminished in diabetic animals, which also exacerbated secondary leakage from damaged vessels. These impairments were related to abnormally high levels of the proinflammatory cytokine IFN-γ because reducing IFN-γ with immunosuppressant drugs or blocking antibodies helped restore normal microglial responses and repair of damaged vessels. These data highlight the use of IFN-γ modulating therapeutics to enhance vascular repair in at-risk populations.


Assuntos
Córtex Cerebral/imunologia , Hemorragia Cerebral/imunologia , Diabetes Mellitus Tipo 1/imunologia , Interferon gama/imunologia , Microglia/imunologia , Animais , Córtex Cerebral/irrigação sanguínea , Hemorragia Cerebral/complicações , Diabetes Mellitus Experimental/sangue , Diabetes Mellitus Experimental/complicações , Diabetes Mellitus Experimental/imunologia , Diabetes Mellitus Tipo 1/sangue , Diabetes Mellitus Tipo 1/complicações , Modelos Animais de Doenças , Encefalite/sangue , Encefalite/complicações , Encefalite/imunologia , Masculino , Camundongos Endogâmicos C57BL , Imagem Óptica
4.
J Neurosci ; 35(13): 5128-43, 2015 Apr 01.
Artigo em Inglês | MEDLINE | ID: mdl-25834040

RESUMO

Diabetes is a common comorbidity in stroke patients and a strong predictor of poor functional outcome. To provide a more mechanistic understanding of this clinically relevant problem, we focused on how diabetes affects blood-brain barrier (BBB) function after stroke. Because the BBB can be compromised for days after stroke and thus further exacerbate ischemic injury, manipulating its function presents a unique opportunity for enhancing stroke recovery long after the window for thrombolytics has passed. Using a mouse model of Type 1 diabetes, we discovered that ischemic stroke leads to an abnormal and persistent increase in vascular endothelial growth factor receptor 2 (VEGF-R2) expression in peri-infarct vascular networks. Correlating with this, BBB permeability was markedly increased in diabetic mice, which could not be prevented with insulin treatment after stroke. Imaging of capillary ultrastructure revealed that BBB permeability was associated with an increase in endothelial transcytosis rather than a loss of tight junctions. Pharmacological inhibition (initiated 2.5 d after stroke) or vascular-specific knockdown of VEGF-R2 after stroke attenuated BBB permeability, loss of synaptic structure in peri-infarct regions, and improved recovery of forepaw function. However, the beneficial effects of VEGF-R2 inhibition on stroke recovery were restricted to diabetic mice and appeared to worsen BBB permeability in nondiabetic mice. Collectively, these results suggest that aberrant VEGF signaling and BBB dysfunction after stroke plays a crucial role in limiting functional recovery in an experimental model of diabetes. Furthermore, our data highlight the need to develop more personalized stroke treatments for a heterogeneous clinical population.


Assuntos
Barreira Hematoencefálica/fisiopatologia , Diabetes Mellitus Experimental/metabolismo , Recuperação de Função Fisiológica/efeitos dos fármacos , Transdução de Sinais/fisiologia , Acidente Vascular Cerebral/metabolismo , Receptor 2 de Fatores de Crescimento do Endotélio Vascular/antagonistas & inibidores , Receptor 2 de Fatores de Crescimento do Endotélio Vascular/metabolismo , Animais , Barreira Hematoencefálica/efeitos dos fármacos , Barreira Hematoencefálica/patologia , Encéfalo/irrigação sanguínea , Encéfalo/efeitos dos fármacos , Encéfalo/patologia , Encéfalo/fisiopatologia , Encéfalo/ultraestrutura , Capilares/patologia , Capilares/ultraestrutura , Espinhas Dendríticas/patologia , Diabetes Mellitus Experimental/complicações , Diabetes Mellitus Experimental/tratamento farmacológico , Modelos Animais de Doenças , Células Endoteliais/metabolismo , Células Endoteliais/ultraestrutura , Expressão Gênica , Indóis/farmacologia , Infarto/complicações , Infarto/patologia , Insulina/uso terapêutico , Camundongos , Permeabilidade/efeitos dos fármacos , Pirróis/farmacologia , Recuperação de Função Fisiológica/fisiologia , Acidente Vascular Cerebral/complicações , Acidente Vascular Cerebral/patologia , Acidente Vascular Cerebral/fisiopatologia , Sinapses/patologia , Transcitose , Receptor 2 de Fatores de Crescimento do Endotélio Vascular/efeitos dos fármacos
5.
Neurobiol Dis ; 78: 1-11, 2015 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-25829228

RESUMO

Clinical and experimental studies have shown a clear link between diabetes, vascular dysfunction and cognitive impairment. However, the molecular underpinnings of this association remain unclear. Since vascular endothelial growth factor (VEGF) signaling is important for maintaining vascular integrity and function, we hypothesized that vascular and cognitive impairment in the diabetic brain could be related to a deficiency in VEGF signaling. Here we show that chronic hyperglycemia (~8weeks) in a mouse model of type 1 diabetes leads to a selective reduction in the expression of VEGF and its cognate receptor (VEGF-R2) in the hippocampus. Correlating with this, diabetic mice showed selective deficits in spatial memory in the Morris water maze, increased vessel area, width and permeability in the dentate gyrus/CA1 region of the hippocampus and reduced spine densities in CA1 neurons. Chronic low dose infusion of VEGF in diabetic mice was sufficient to restore VEGF signaling, protect them from memory deficits, as well as vascular and synaptic abnormalities in the hippocampus. These findings suggest that a hippocampal specific reduction in VEGF signaling and resultant vascular/neuronal defects may underlie early manifestations of cognitive impairment commonly associated with diabetes. Furthermore, restoring VEGF signaling may be a useful strategy for preserving hippocampal-related brain circuitry in degenerative vascular diseases.


Assuntos
Diabetes Mellitus Tipo 1/metabolismo , Hipocampo/metabolismo , Fator A de Crescimento do Endotélio Vascular/administração & dosagem , Fator A de Crescimento do Endotélio Vascular/metabolismo , Animais , Barreira Hematoencefálica/efeitos dos fármacos , Barreira Hematoencefálica/metabolismo , Espinhas Dendríticas/efeitos dos fármacos , Espinhas Dendríticas/patologia , Diabetes Mellitus Tipo 1/prevenção & controle , Diabetes Mellitus Tipo 1/psicologia , Modelos Animais de Doenças , Hipocampo/irrigação sanguínea , Hipocampo/efeitos dos fármacos , Infusões Intraventriculares , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Memória Espacial/efeitos dos fármacos , Receptor 2 de Fatores de Crescimento do Endotélio Vascular/metabolismo
6.
J Neurosci ; 33(49): 19194-204, 2013 Dec 04.
Artigo em Inglês | MEDLINE | ID: mdl-24305815

RESUMO

Stroke usually affects people with underlying medical conditions. In particular, diabetics are significantly more likely to have a stroke and the prognosis for recovery is poor. Because diabetes is associated with degenerative changes in the vasculature of many organs, we sought to determine how hyperglycemia affects blood flow dynamics after an ischemic stroke. Longitudinal in vivo two-photon imaging was used to track microvessels before and after photothrombotic stroke in a diabetic mouse model. Chronic hyperglycemia exacerbated acute (3-7 d) ischemia-induced increases in blood flow velocity, vessel lumen diameter, and red blood cell flux in peri-infarct regions. These changes in blood flow dynamics were most evident in superficial blood vessels within 500 µm from the infarct, rather than deeper or more distant cortical regions. Long-term imaging of diabetic mice not subjected to stroke indicated that these acute stroke-related changes in vascular function could not be attributed to complications from hyperglycemia alone. Treating diabetic mice with insulin immediately after stroke resulted in less severe alterations in blood flow within the first 7 d of recovery, but had more variable results at later time points. Analysis of microvessel branching patterns revealed that stroke led to a pruning of microvessels in peri-infarct cortex, with very few instances of sprouting. These results indicate that chronic hyperglycemia significantly affects the vascular response to ischemic stroke and that insulin only partially mitigates these changes. The combination of these acute and chronic alterations in blood flow dynamics could underlie diabetes-related deficits in cortical plasticity and stroke recovery.


Assuntos
Diabetes Mellitus/fisiopatologia , Microcirculação/fisiologia , Acidente Vascular Cerebral/fisiopatologia , Animais , Velocidade do Fluxo Sanguíneo/fisiologia , Glicemia/metabolismo , Peso Corporal/fisiologia , Infarto Cerebral/patologia , Circulação Cerebrovascular/fisiologia , Eritrócitos/fisiologia , Hiperglicemia/fisiopatologia , Trombose Intracraniana/fisiopatologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Neuroimagem , Recuperação de Função Fisiológica , Mecânica Respiratória/fisiologia
7.
Neuron ; 112(9): 1378-1380, 2024 May 01.
Artigo em Inglês | MEDLINE | ID: mdl-38697020

RESUMO

Adequate reperfusion after ischemic stroke is a major determinant of functional outcome yet remains unpredictable and insufficient for most survivors. In this issue of Neuron, Binder et al.1 identify leptomeningeal collaterals (LMCs) in mice and human patients as a key factor in regulating reperfusion and hemorrhagic transformation following stroke.


Assuntos
Circulação Colateral , Reperfusão , Acidente Vascular Cerebral , Humanos , Animais , Acidente Vascular Cerebral/fisiopatologia , Circulação Colateral/fisiologia , Camundongos , AVC Isquêmico/fisiopatologia , Circulação Cerebrovascular/fisiologia , Meninges/irrigação sanguínea , Isquemia Encefálica/fisiopatologia
8.
J Neurosci ; 32(4): 1207-19, 2012 Jan 25.
Artigo em Inglês | MEDLINE | ID: mdl-22279206

RESUMO

The molecular mechanisms that mediate experience-based changes in the function of the cerebral cortex, particularly in the adult animal, are poorly understood. Here we show using in vivo voltage-sensitive dye imaging, that whisker trimming leads to depression of whisker-evoked sensory responses in primary, secondary and associative somatosensory cortical regions. Given the importance of cholinergic neurotransmission in cognitive and sensory functions, we examined whether α4-containing (α4*) nicotinic acetylcholine receptors (nAChRs) mediate cortical depression. Using knock-in mice that express YFP-tagged α4 nAChRs subunits, we show that whisker trimming selectively increased the number α4*-YFP nAChRs in layer 4 of deprived barrel columns within 24 h, which persisted until whiskers regrew. Confocal and electron microscopy revealed that these receptors were preferentially increased on the cell bodies of GABAergic neurons. To directly link these receptors with functional cortical depression, we show that depression could be induced in normal mice by topical application or micro-injection of α4* nAChR agonist in the somatosensory cortex. Furthermore, cortical depression could be blocked after whisker trimming with chronic infusions of an α4* nAChR antagonist. Collectively, these results uncover a new role for α4* nAChRs in regulating rapid changes in the functional responsiveness of the adult somatosensory cortex.


Assuntos
Depressão Alastrante da Atividade Elétrica Cortical/genética , Receptores Nicotínicos/fisiologia , Córtex Somatossensorial/fisiologia , Vibrissas/inervação , Fatores Etários , Animais , Depressão Alastrante da Atividade Elétrica Cortical/fisiologia , Potenciais Somatossensoriais Evocados/efeitos dos fármacos , Potenciais Somatossensoriais Evocados/genética , Técnicas de Introdução de Genes , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Agonistas Nicotínicos/farmacologia , Antagonistas Nicotínicos/farmacologia , Receptores Nicotínicos/biossíntese , Receptores Nicotínicos/genética , Córtex Somatossensorial/efeitos dos fármacos , Vibrissas/efeitos dos fármacos
9.
J Neurosci ; 32(15): 5132-43, 2012 Apr 11.
Artigo em Inglês | MEDLINE | ID: mdl-22496559

RESUMO

Diabetics are at greater risk of having a stroke and are less likely to recover from it. To understand this clinically relevant problem, we induced an ischemic stroke in the primary forelimb somatosensory (FLS1) cortex of diabetic mice and then examined sensory-evoked changes in cortical membrane potentials and behavioral recovery of forelimb sensory-motor function. Consistent with previous studies, focal stroke in non-diabetic mice was associated with acute deficits in forelimb sensorimotor function and a loss of forelimb evoked cortical depolarizations in peri-infarct cortex that gradually recovered over several weeks time. In addition, we discovered that damage to FLS1 cortex led to an enhancement of forelimb evoked depolarizations in secondary forelimb somatosensory (FLS2) cortex. Enhanced FLS2 cortical responses appeared to play a role in stroke recovery given that silencing this region was sufficient to reinstate forelimb impairments. By contrast, the functional reorganization of FLS1 and FLS2 cortex was largely absent in diabetic mice and could not be explained by more severe cortical infarctions. Diabetic mice also showed persistent behavioral deficits in sensorimotor function of the forepaw, which could not be rescued by chronic insulin therapy after stroke. Collectively these results indicate that diabetes has a profound effect on brain plasticity, especially when challenged, as is often the case, by an ischemic event. Further, our data suggest that secondary cortical regions play an important role in the restoration of sensorimotor function when primary cortical regions are damaged.


Assuntos
Isquemia Encefálica/fisiopatologia , Complicações do Diabetes/fisiopatologia , Plasticidade Neuronal/fisiologia , Recuperação de Função Fisiológica , Córtex Somatossensorial/fisiopatologia , Acidente Vascular Cerebral/fisiopatologia , Animais , Glicemia/metabolismo , Isquemia Encefálica/complicações , Infarto Cerebral/patologia , Diabetes Mellitus Experimental/complicações , Diabetes Mellitus Experimental/fisiopatologia , Vias Eferentes/fisiopatologia , Membro Anterior/inervação , Membro Anterior/fisiologia , Lateralidade Funcional/fisiologia , Hipoglicemiantes/uso terapêutico , Insulina/uso terapêutico , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Movimento/fisiologia , Sensação/fisiologia , Acidente Vascular Cerebral/complicações , Trombose/complicações
10.
Cell Rep Methods ; 3(6): 100489, 2023 06 26.
Artigo em Inglês | MEDLINE | ID: mdl-37426748

RESUMO

Adeno-associated viruses (AAVs) are used in a wide array of experimental situations for driving expression of biosensors, recombinases, and opto-/chemo-genetic actuators in the brain. However, conventional approaches for minimally invasive, spatially precise, and ultra-sparse AAV-mediated transduction of cells during imaging experiments have remained a significant challenge. Here, we show that intravenous injection of commercially available AAVs at different doses, combined with laser-based perforation of cortical capillaries through a cranial widow, allows for ultra-sparse, titratable, and micron-level precision for delivery of viral vectors with relatively little inflammation or tissue damage. Further, we show the utility of this approach for eliciting sparse expression of GCaMP6, channelrhodopsin, or fluorescent reporters in neurons and astrocytes within specific functional domains in normal and stroke-damaged cortex. This technique represents a facile approach for targeted delivery of viral vectors that should assist in the study of cell types and circuits in the cortex.


Assuntos
Barreira Hematoencefálica , Neurônios , Camundongos , Animais , Neurônios/metabolismo , Encéfalo , Astrócitos/metabolismo , Crânio
11.
Pharmacol Ther ; 229: 107929, 2022 01.
Artigo em Inglês | MEDLINE | ID: mdl-34171341

RESUMO

The complex computations of the brain require a constant supply of blood flow to meet its immense metabolic needs. Perturbations in blood supply, even in the smallest vascular networks, can have a profound effect on neuronal function and cognition. Type 1 diabetes is a prevalent and insidious metabolic disorder that progressively and heterogeneously disrupts vascular signalling and function in the brain. As a result, it is associated with an array of adverse vascular changes such as impaired regulation of vascular tone, pathological neovascularization and vasoregression, capillary plugging and blood brain barrier disruption. In this review, we highlight the link between microvascular dysfunction and cognitive impairment that is commonly associated with type 1 diabetes, with the aim of synthesizing current knowledge in this field.


Assuntos
Disfunção Cognitiva , Diabetes Mellitus Tipo 1 , Encéfalo/irrigação sanguínea , Cognição , Disfunção Cognitiva/etiologia , Diabetes Mellitus Tipo 1/complicações , Humanos
12.
Front Cell Neurosci ; 16: 876746, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-35722620

RESUMO

The capillaries of the brain, owing to their small diameter and low perfusion pressure, are vulnerable to interruptions in blood flow. These tiny occlusions can have outsized consequences on angioarchitecture and brain function; especially when exacerbated by disease states or accumulate with aging. A distinctive feature of the brain's microvasculature is the ability for active neurons to recruit local blood flow. The coupling of neural activity to blood flow could play an important role in recanalizing obstructed capillaries. To investigate this idea, we experimentally induced capillary obstructions in mice by injecting fluorescent microspheres and then manipulated neural activity levels though behavioral or pharmacologic approaches. We show that engaging adult and aged mice with 12 h exposure to an enriched environment (group housing, novel objects, exercise wheels) was sufficient to significantly reduce the density of obstructed capillaries throughout the forebrain. In order to more directly manipulate neural activity, we pharmacologically suppressed or increased neuronal activity in the somatosensory cortex. When we suppressed cortical activity, recanalization was impaired given the density of obstructed capillaries was significantly increased. Conversely, increasing cortical activity improved capillary recanalization. Since systemic cardiovascular factors (changes in heart rate, blood pressure) could explain these effects on recanalization, we demonstrate that unilateral manipulations of neural activity through whisker trimming or injection of muscimol, still had significant and hemisphere specific effects on recanalization, even in mice exposed to enrichment where cardiovascular effects would be evident in both hemispheres. In summary, our studies reveal that neural activity bi-directionally regulates the recanalization of obstructed capillaries. Further, we show that stimulating brain activity through behavioral engagement (i.e., environmental enrichment) can promote vascular health throughout the lifespan.

13.
Sci Adv ; 7(34)2021 08.
Artigo em Inglês | MEDLINE | ID: mdl-34407943

RESUMO

The cellular events that dictate the repair of damaged vessels in the brain, especially in those with vascular risk factors such as diabetes, is poorly understood. Here, we dissected the role of resident microglia and infiltrative macrophages in determining the repair of ruptured cerebral microvessels. Using in vivo time-lapse imaging, gene expression analysis, and immunohistochemistry, we identified a unique population of phagocytic Galectin 3 (Gal3) expressing macrophages, distinct from resident microglia, which infiltrated and aggregated at the site of injury in diabetic mice and were associated with the elimination of microvessels. Depletion of these infiltrative macrophages in diabetic mice attenuated phagocytic activity and prevented the loss of blood vessels after injury. These findings highlight a previously unknown role for infiltrative Gal3 expressing macrophages in promoting vessel elimination after brain injury and provide impetus for future studies to determine whether depleting these cells can facilitate vascular repair in at risk populations.


Assuntos
Diabetes Mellitus Experimental , Galectina 3 , Animais , Encéfalo/metabolismo , Diabetes Mellitus Experimental/genética , Galectina 3/genética , Galectina 3/metabolismo , Macrófagos/metabolismo , Camundongos , Microglia/metabolismo
14.
Nat Commun ; 12(1): 6112, 2021 10 20.
Artigo em Inglês | MEDLINE | ID: mdl-34671051

RESUMO

Stroke profoundly disrupts cortical excitability which impedes recovery, but how it affects the function of specific inhibitory interneurons, or subpopulations therein, is poorly understood. Interneurons expressing vasoactive intestinal peptide (VIP) represent an intriguing stroke target because they can regulate cortical excitability through disinhibition. Here we chemogenetically augmented VIP interneuron excitability in a murine model of photothrombotic stroke and show that it enhances somatosensory responses and improves recovery of paw function. Using longitudinal calcium imaging, we discovered that stroke primarily disrupts the fidelity (fraction of responsive trials) and predictability of sensory responses within a subset of highly active VIP neurons. Partial recovery of responses occurred largely within these active neurons and was not accompanied by the recruitment of minimally active neurons. Importantly, chemogenetic stimulation preserved sensory response fidelity and predictability in highly active neurons. These findings provide a new depth of understanding into how stroke and prospective therapies (chemogenetics), can influence subpopulations of inhibitory interneurons.


Assuntos
Interneurônios/fisiologia , Acidente Vascular Cerebral/terapia , Peptídeo Intestinal Vasoativo/metabolismo , Animais , Clozapina/análogos & derivados , Clozapina/uso terapêutico , Humanos , Interneurônios/efeitos dos fármacos , Interneurônios/metabolismo , Camundongos , Inibição Neural/efeitos dos fármacos , Receptor Muscarínico M3/genética , Receptor Muscarínico M3/metabolismo , Recuperação de Função Fisiológica , Córtex Somatossensorial/citologia , Córtex Somatossensorial/efeitos dos fármacos , Córtex Somatossensorial/fisiologia , Acidente Vascular Cerebral/metabolismo , Acidente Vascular Cerebral/fisiopatologia
15.
J Neurosci ; 29(6): 1719-34, 2009 Feb 11.
Artigo em Inglês | MEDLINE | ID: mdl-19211879

RESUMO

After brain damage such as stroke, topographically organized sensory and motor cortical representations remap onto adjacent surviving tissues. It is conceivable that cortical remapping is accomplished by changes in the temporal precision of sensory processing and regional connectivity in the cortex. To understand how the adult cortex remaps and processes sensory signals during stroke recovery, we performed in vivo imaging of sensory-evoked changes in membrane potential, as well as multiphoton imaging of dendrite structure and tract tracing. In control mice, forelimb stimulation evoked a brief depolarization in forelimb cortex that quickly propagated to, and dissipated within, adjacent motor/hindlimb areas (<100 ms). One week after forelimb cortex stroke, the cortex was virtually unresponsive to tactile forelimb stimulation. After 8 weeks recovery, forelimb-evoked depolarizations reemerged with a characteristic pattern in which responses began within surviving portions of forelimb cortex (<20 ms after stimulation) and then spread horizontally into neighboring peri-infarct motor/hindlimb areas in which depolarization persisted 300-400% longer than controls. These uncharacteristically prolonged responses were not limited to the remapped peri-infarct zone and included distant posteromedial retrosplenial cortex, millimeters from the stroke. Structurally, the remapped peri-infarct area selectively exhibited high levels of dendritic spine turnover, shared more connections with retrosplenial cortex and striatum, and lost inputs from lateral somatosensory cortical regions. Our findings demonstrate that sensory remapping during stroke recovery is accompanied by the development of prolonged sensory responses and new structural circuits in both the peri-infarct zone as well as more distant sites.


Assuntos
Mapeamento Encefálico , Infarto Cerebral/diagnóstico , Corantes Fluorescentes , Rede Nervosa/fisiologia , Plasticidade Neuronal/fisiologia , Rosa Bengala , Córtex Somatossensorial/anatomia & histologia , Córtex Somatossensorial/fisiologia , Animais , Mapeamento Encefálico/métodos , Infarto Cerebral/fisiopatologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Rede Nervosa/química , Rede Nervosa/fisiopatologia , Estimulação Luminosa/métodos , Córtex Somatossensorial/química , Fatores de Tempo
16.
Hippocampus ; 20(2): 305-22, 2010 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-19437420

RESUMO

Trans-synaptic cell-adhesion molecules have been implicated in regulating CNS synaptogenesis. Among these, the Neuroligin (NL) family (NLs 1-4) of postsynaptic adhesion proteins has been shown to promote the development and specification of excitatory versus inhibitory synapses. NLs form a heterophilic complex with the presynaptic transmembrane protein Neurexin (NRX). A differential association of NLs with postsynaptic scaffolding proteins and NRX isoforms has been suggested to regulate the ratio of excitatory to inhibitory synapses (E/I ratio). Using transgenic mice, we have tested this hypothesis by overexpressing NL1 in vivo to determine whether the relative levels of these cell adhesion molecules may influence synapse maturation, long-term potentiation (LTP), and/or learning. We found that NL1-overexpressing mice show significant deficits in memory acquisition, but not in memory retrieval. Golgi and electron microscopy analysis revealed changes in synapse morphology indicative of increased maturation of excitatory synapses. In parallel, electrophysiological examination indicated a shift in the synaptic activity toward increased excitation as well as impairment in LTP induction. Our results demonstrate that altered balance in the expression of molecules necessary for synapse specification and development (such as NL1) can lead to defects in memory formation and synaptic plasticity and outline the importance of rigidly controlled synaptic maturation processes.


Assuntos
Hipocampo/fisiopatologia , Deficiências da Aprendizagem/fisiopatologia , Moléculas de Adesão de Célula Nervosa/metabolismo , Plasticidade Neuronal/fisiologia , Sinapses/fisiologia , Transmissão Sináptica/fisiologia , Animais , Encéfalo/fisiopatologia , Encéfalo/ultraestrutura , Moléculas de Adesão Celular Neuronais , Espinhas Dendríticas/fisiologia , Espinhas Dendríticas/ultraestrutura , Hipocampo/ultraestrutura , Técnicas In Vitro , Deficiências da Aprendizagem/patologia , Potenciação de Longa Duração/fisiologia , Potenciais da Membrana/fisiologia , Memória/fisiologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Moléculas de Adesão de Célula Nervosa/genética , Inibição Neural/fisiologia , Sinapses/ultraestrutura
17.
J Cereb Blood Flow Metab ; 40(12): 2475-2490, 2020 12.
Artigo em Inglês | MEDLINE | ID: mdl-31903837

RESUMO

Vessel loss in the aging brain is commonly reported, yet important questions remain concerning whether there are regional vulnerabilities and what mechanisms could account for these regional differences, if they exist. Here we imaged and quantified vessel length, tortuosity and width in 15 brain regions in young adult and aged mice. Our data indicate that vessel loss was most pronounced in white matter followed by cortical, then subcortical grey matter regions, while some regions (visual cortex, amygdala, thalamus) showed no decline with aging. Regions supplied by the anterior cerebral artery were more vulnerable to loss than those supplied by middle or posterior cerebral arteries. Vessel width and tortuosity generally increased with age but neither reliably predicted regional vessel loss. Since capillaries are naturally prone to plugging and prolonged obstructions often lead to vessel pruning, we hypothesized that regional susceptibilities to plugging could help predict vessel loss. By mapping the distribution of microsphere-induced capillary obstructions, we discovered that regions with a higher density of persistent obstructions were more likely to show vessel loss with aging and vice versa. These findings indicate that age-related vessel loss is region specific and can be explained, at least partially, by regional susceptibilities to capillary plugging.


Assuntos
Isquemia Encefálica/patologia , Encéfalo/irrigação sanguínea , Capilares/patologia , Córtex Cerebral/patologia , Demência Vascular/patologia , Rarefação Microvascular/fisiopatologia , Envelhecimento/fisiologia , Animais , Encéfalo/patologia , Isquemia Encefálica/fisiopatologia , Capilares/crescimento & desenvolvimento , Capilares/fisiopatologia , Córtex Cerebral/irrigação sanguínea , Córtex Cerebral/fisiopatologia , Circulação Cerebrovascular/fisiologia , Demência Vascular/etiologia , Demência Vascular/fisiopatologia , Feminino , Substância Cinzenta/irrigação sanguínea , Substância Cinzenta/patologia , Substância Cinzenta/fisiopatologia , Humanos , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Valor Preditivo dos Testes , Substância Branca/irrigação sanguínea , Substância Branca/patologia , Substância Branca/fisiopatologia
18.
eNeuro ; 6(3)2019.
Artigo em Inglês | MEDLINE | ID: mdl-31118206

RESUMO

Dendritic spines are the postsynaptic targets of excitatory synaptic inputs that undergo extensive proliferation and maturation during the first postnatal month in mice. However, our understanding of the molecular mechanisms that regulate spines during this critical period is limited. Previous work has shown that pannexin 1 (Panx1) regulates neurite growth and synaptic plasticity. We therefore investigated the impact of global Panx1 KO on spontaneous cortical neuron activity using Ca2+ imaging and in silico network analysis. Panx1 KO increased both the number and size of spontaneous co-active cortical neuron network ensembles. To understand the basis for these findings, we investigated Panx1 expression in postnatal synaptosome preparations from early postnatal mouse cortex. Between 2 and 4 postnatal weeks, we observed a precipitous drop in cortical synaptosome protein levels of Panx1, suggesting it regulates synapse proliferation and/or maturation. At the same time points, we observed significant enrichment of the excitatory postsynaptic density proteins PSD-95, GluA1, and GluN2a in cortical synaptosomes from global Panx1 knock-out mice. Ex vivo analysis of pyramidal neuron structure in somatosensory cortex revealed a consistent increase in dendritic spine densities in both male and female Panx1 KO mice. Similar findings were observed in an excitatory neuron-specific Panx1 KO line (Emx1-Cre driven; Panx1 cKOE) and in primary Panx1 KO cortical neurons cultured in vitro. Altogether, our study suggests that Panx1 negatively regulates cortical dendritic spine development.


Assuntos
Córtex Cerebral/crescimento & desenvolvimento , Conexinas/fisiologia , Espinhas Dendríticas/fisiologia , Proteínas do Tecido Nervoso/fisiologia , Animais , Sinalização do Cálcio , Córtex Cerebral/metabolismo , Conexinas/genética , Conexinas/metabolismo , Espinhas Dendríticas/metabolismo , Proteína 4 Homóloga a Disks-Large/metabolismo , Feminino , Masculino , Camundongos Endogâmicos C57BL , Camundongos Knockout , Proteínas do Tecido Nervoso/genética , Proteínas do Tecido Nervoso/metabolismo , Vias Neurais/crescimento & desenvolvimento , Vias Neurais/metabolismo , Imagem Óptica , Sinaptossomos/metabolismo , Sinaptossomos/fisiologia
19.
J Neurosci ; 27(15): 4101-9, 2007 Apr 11.
Artigo em Inglês | MEDLINE | ID: mdl-17428988

RESUMO

Recovery of function after stroke is thought to be dependent on the reorganization of adjacent, surviving areas of the brain. Macroscopic imaging studies (functional magnetic resonance imaging, optical imaging) have shown that peri-infarct regions adopt new functional roles to compensate for damage caused by stroke. To better understand the process by which these regions reorganize, we used in vivo two-photon imaging to examine changes in dendritic and vascular structure in cortical regions recovering from stroke. In adult control mice, dendritic arbors were relatively stable with very low levels of spine turnover (<0.5% turnover over 6 h). After stroke, however, the organization of dendritic arbors in peri-infarct cortex was fundamentally altered with both apical dendrites and blood vessels radiating in parallel from the lesion. On a finer scale, peri-infarct dendrites were exceptionally plastic, manifested by a dramatic increase in the rate of spine formation that was maximal at 1-2 weeks (5-8-fold increase), and still evident 6 weeks after stroke. These changes were selective given that turnover rates were not significantly altered in ipsilateral cortical regions more distant to the lesion (>1.5 mm). These data provide a structural framework for understanding functional and behavioral changes that accompany brain injury and suggest new targets that could be exploited by future therapies to rebuild and rewire neuronal circuits lost to stroke.


Assuntos
Córtex Cerebral/ultraestrutura , Espinhas Dendríticas/ultraestrutura , Plasticidade Neuronal , Recuperação de Função Fisiológica , Acidente Vascular Cerebral/patologia , Animais , Córtex Cerebral/patologia , Córtex Cerebral/fisiologia , Infarto Cerebral/patologia , Infarto Cerebral/fisiopatologia , Espinhas Dendríticas/patologia , Espinhas Dendríticas/fisiologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Plasticidade Neuronal/fisiologia , Recuperação de Função Fisiológica/fisiologia , Acidente Vascular Cerebral/fisiopatologia
20.
Stroke ; 39(4): 1286-91, 2008 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-18323506

RESUMO

BACKGROUND AND PURPOSE: Focal stroke is associated with cell death, abnormal synaptic activity, and neurologic impairments. Given that many of these neuropathologic processes can be attributed to events that occur shortly after injury, it is necessary to understand how stroke affects the structure of neurons in surviving peri-infarct regions, particularly at the level of the dendritic spines, which transmit normal and potentially abnormal and injurious synaptic signaling. Recently, we described ischemia-induced changes in the structure of layer 1 dendritic tufts of transgenic mice expressing YFP in layer 5 cortical neurons. However, these in vivo imaging experiments could not address ischemia-related phenomena that occur in deeper cortical structures/layers, other cortical regions, or submicron changes in dendritic spine structure. METHODS: Focal stroke was induced in the forelimb sensorimotor cortex by the photothrombotic method. Two, 6, and 24 hours after stroke, brains were processed for Golgi-Cox staining to permit a detailed analysis of primary apical dendritic spine structure from layer 2/3 and 5 cortical pyramidal neurons. RESULTS: Photothrombotic stroke caused a rapid deterioration of neurons, as revealed by Golgi-Cox labeling, in the infarct core that could be readily distinguished from surviving peri-infarct regions. Analysis of >15,000 dendritic spines revealed that although many spines were lost in the peri-infarct cortex during the first 24 hours after stroke (approximately 38% lost), spines that remained were significantly longer (approximately 25% at 6 hours). Furthermore, these effects were found in both layer 2/3 and 5 neurons and were restricted primarily to peri-infarct regions (<200 mum from the infarct border). CONCLUSIONS: These rapid changes in dendritic spine number and length may reflect an early adaptive response of potentially vulnerable peri-infarct neurons coping with postischemic spreading depression-like depolarizations and the loss of presynaptic contacts.


Assuntos
Isquemia Encefálica/fisiopatologia , Infarto Cerebral/fisiopatologia , Espinhas Dendríticas/fisiologia , Plasticidade Neuronal/fisiologia , Acidente Vascular Cerebral/fisiopatologia , Animais , Isquemia Encefálica/patologia , Infarto Cerebral/patologia , Espinhas Dendríticas/patologia , Modelos Animais de Doenças , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Córtex Motor/patologia , Córtex Motor/fisiopatologia , Células Piramidais/patologia , Células Piramidais/fisiologia , Células Piramidais/ultraestrutura , Recuperação de Função Fisiológica/fisiologia , Coloração pela Prata , Córtex Somatossensorial/patologia , Córtex Somatossensorial/fisiopatologia , Acidente Vascular Cerebral/patologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA