Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 9 de 9
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
2.
Nat Commun ; 13(1): 600, 2022 02 09.
Artigo em Inglês | MEDLINE | ID: mdl-35140203

RESUMO

Ultrasound has been used to non-invasively manipulate neuronal functions in humans and other animals. However, this approach is limited as it has been challenging to target specific cells within the brain or body. Here, we identify human Transient Receptor Potential A1 (hsTRPA1) as a candidate that confers ultrasound sensitivity to mammalian cells. Ultrasound-evoked gating of hsTRPA1 specifically requires its N-terminal tip region and cholesterol interactions; and target cells with an intact actin cytoskeleton, revealing elements of the sonogenetic mechanism. Next, we use calcium imaging and electrophysiology to show that hsTRPA1 potentiates ultrasound-evoked responses in primary neurons. Furthermore, unilateral expression of hsTRPA1 in mouse layer V motor cortical neurons leads to c-fos expression and contralateral limb responses in response to ultrasound delivered through an intact skull. Collectively, we demonstrate that hsTRPA1-based sonogenetics can effectively manipulate neurons within the intact mammalian brain, a method that could be used across species.


Assuntos
Canal de Cátion TRPA1/genética , Canal de Cátion TRPA1/metabolismo , Canais de Potencial de Receptor Transitório/genética , Canais de Potencial de Receptor Transitório/metabolismo , Animais , Encéfalo/metabolismo , Cálcio/metabolismo , Feminino , Humanos , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Neurônios Motores/metabolismo
3.
Adv Sci (Weinh) ; 9(2): e2101950, 2022 01.
Artigo em Inglês | MEDLINE | ID: mdl-34747144

RESUMO

Ultrasound has been used to manipulate cells in both humans and animal models. While intramembrane cavitation and lipid clustering have been suggested as likely mechanisms, they lack experimental evidence. Here, high-speed digital holographic microscopy (kiloHertz order) is used to visualize the cellular membrane dynamics. It is shown that neuronal and fibroblast membranes deflect about 150 nm upon ultrasound stimulation. Next, a biomechanical model that predicts changes in membrane voltage after ultrasound exposure is developed. Finally, the model predictions are validated using whole-cell patch clamp electrophysiology on primary neurons. Collectively, it is shown that ultrasound stimulation directly defects the neuronal membrane leading to a change in membrane voltage and subsequent depolarization. The model is consistent with existing data and provides a mechanism for both ultrasound-evoked neurostimulation and sonogenetic control.


Assuntos
Modelos Neurológicos , Neurônios/fisiologia , Ondas Ultrassônicas , Animais , Membrana Celular , Células Cultivadas , Humanos , Microscopia , Modelos Animais , Técnicas de Patch-Clamp , Ratos
4.
Neuron ; 93(3): 574-586.e8, 2017 Feb 08.
Artigo em Inglês | MEDLINE | ID: mdl-28111081

RESUMO

Microglia are the intrinsic immune sentinels of the central nervous system. Their activation restricts tissue injury and pathogen spread, but in some settings, including viral infection, this response can contribute to cell death and disease. Identifying mechanisms that control microglial responses is therefore an important objective. Using replication-incompetent adenovirus 5 (Ad5)-based vectors as a model, we investigated the mechanisms through which microglia recognize and respond to viral uptake. Transgenic, immunohistochemical, molecular-genetic, and fluorescence imaging approaches revealed that phosphatidylserine (PtdSer) exposure on the outer leaflet of transduced cells triggers their engulfment by microglia through TAM receptor-dependent mechanisms. We show that inhibition of phospholipid scramblase 1 (PLSCR1) activity reduces intracellular calcium dysregulation, prevents PtdSer externalization, and enables months-long protection of vector-transduced, transgene-expressing cells from microglial phagocytosis. Our study identifies PLSCR1 as a potent target through which the innate immune response to viral vectors, and potentially other stimuli, may be controlled.


Assuntos
Infecções por Adenoviridae/imunologia , Adenoviridae/imunologia , Vetores Genéticos/imunologia , Imunidade Inata/imunologia , Microglia/imunologia , Neurônios/imunologia , Fagocitose/imunologia , Fosfatidilserinas/imunologia , Proteínas de Transferência de Fosfolipídeos/imunologia , Animais , Técnicas de Silenciamento de Genes , Imuno-Histoquímica , Camundongos Transgênicos , Neurônios/virologia , Imagem Óptica , Proteínas de Transferência de Fosfolipídeos/genética
5.
Nature ; 532(7598): 240-244, 2016 Apr 14.
Artigo em Inglês | MEDLINE | ID: mdl-27049947

RESUMO

Microglia are damage sensors for the central nervous system (CNS), and the phagocytes responsible for routine non-inflammatory clearance of dead brain cells. Here we show that the TAM receptor tyrosine kinases Mer and Axl regulate these microglial functions. We find that adult mice deficient in microglial Mer and Axl exhibit a marked accumulation of apoptotic cells specifically in neurogenic regions of the CNS, and that microglial phagocytosis of the apoptotic cells generated during adult neurogenesis is normally driven by both TAM receptor ligands Gas6 and protein S. Using live two-photon imaging, we demonstrate that the microglial response to brain damage is also TAM-regulated, as TAM-deficient microglia display reduced process motility and delayed convergence to sites of injury. Finally, we show that microglial expression of Axl is prominently upregulated in the inflammatory environment that develops in a mouse model of Parkinson's disease. Together, these results establish TAM receptors as both controllers of microglial physiology and potential targets for therapeutic intervention in CNS disease.


Assuntos
Encéfalo/metabolismo , Microglia/fisiologia , Proteínas Proto-Oncogênicas/metabolismo , Receptores Proteína Tirosina Quinases/metabolismo , Animais , Apoptose , Encéfalo/irrigação sanguínea , Encéfalo/citologia , Encéfalo/patologia , Lesões Encefálicas/metabolismo , Lesões Encefálicas/patologia , Modelos Animais de Doenças , Feminino , Inflamação/metabolismo , Peptídeos e Proteínas de Sinalização Intercelular/metabolismo , Ligantes , Masculino , Camundongos , Neurogênese , Doença de Parkinson/metabolismo , Fagocitose , Proteína S/metabolismo , Proteínas Proto-Oncogênicas/deficiência , Receptores Proteína Tirosina Quinases/deficiência , Transdução de Sinais , Nicho de Células-Tronco , Regulação para Cima , c-Mer Tirosina Quinase , Receptor Tirosina Quinase Axl
6.
Nat Protoc ; 6(9): 1453-70, 2011 Sep 01.
Artigo em Inglês | MEDLINE | ID: mdl-21886108

RESUMO

Brain stimulation methods are indispensable to the study of brain function. They have also proven effective for treating some neurological disorders. Historically used for medical imaging, ultrasound (US) has recently been shown to be capable of noninvasively stimulating brain activity. Here we provide a general protocol for the stimulation of intact mouse brain circuits using transcranial US, and, using a traditional mouse model of epilepsy, we describe how to use transcranial US to disrupt electrographic seizure activity. The advantages of US for brain stimulation are that it does not necessitate surgery or genetic alteration, but it confers spatial resolutions superior to other noninvasive methods such as transcranial magnetic stimulation. With a basic working knowledge of electrophysiology, and after an initial setup, ultrasonic neuromodulation (UNMOD) can be implemented in less than 1 h. Using the general protocol that we describe, UNMOD can be readily adapted to support a broad range of studies on brain circuit function and dysfunction.


Assuntos
Mapeamento Encefálico/métodos , Ultrassonografia Doppler Transcraniana/métodos , Animais , Encéfalo/fisiologia , Eletrofisiologia , Feminino , Masculino , Camundongos , Camundongos Endogâmicos C57BL
7.
Neuron ; 66(5): 681-94, 2010 Jun 10.
Artigo em Inglês | MEDLINE | ID: mdl-20547127

RESUMO

Electromagnetic-based methods of stimulating brain activity require invasive procedures or have other limitations. Deep-brain stimulation requires surgically implanted electrodes. Transcranial magnetic stimulation does not require surgery, but suffers from low spatial resolution. Optogenetic-based approaches have unrivaled spatial precision, but require genetic manipulation. In search of a potential solution to these limitations, we began investigating the influence of transcranial pulsed ultrasound on neuronal activity in the intact mouse brain. In motor cortex, ultrasound-stimulated neuronal activity was sufficient to evoke motor behaviors. Deeper in subcortical circuits, we used targeted transcranial ultrasound to stimulate neuronal activity and synchronous oscillations in the intact hippocampus. We found that ultrasound triggers TTX-sensitive neuronal activity in the absence of a rise in brain temperature (<0.01 degrees C). Here, we also report that transcranial pulsed ultrasound for intact brain circuit stimulation has a lateral spatial resolution of approximately 2 mm and does not require exogenous factors or surgical invasion.


Assuntos
Rede Nervosa/fisiologia , Ultrassom , Animais , Encéfalo/fisiologia , Hipocampo/fisiologia , Camundongos , Neurônios/fisiologia , Ultrassonografia Doppler Transcraniana/métodos
9.
PLoS One ; 3(10): e3511, 2008.
Artigo em Inglês | MEDLINE | ID: mdl-18958151

RESUMO

Possessing the ability to noninvasively elicit brain circuit activity yields immense experimental and therapeutic power. Most currently employed neurostimulation methods rely on the somewhat invasive use of stimulating electrodes or photon-emitting devices. Due to its ability to noninvasively propagate through bone and other tissues in a focused manner, the implementation of ultrasound (US) represents a compelling alternative approach to current neuromodulation strategies. Here, we investigated the influence of low-intensity, low-frequency ultrasound (LILFU) on neuronal activity. By transmitting US waveforms through hippocampal slice cultures and ex vivo mouse brains, we determined LILFU is capable of remotely and noninvasively exciting neurons and network activity. Our results illustrate that LILFU can stimulate electrical activity in neurons by activating voltage-gated sodium channels, as well as voltage-gated calcium channels. The LILFU-induced changes in neuronal activity were sufficient to trigger SNARE-mediated exocytosis and synaptic transmission in hippocampal circuits. Because LILFU can stimulate electrical activity and calcium signaling in neurons as well as central synaptic transmission we conclude US provides a powerful tool for remotely modulating brain circuit activity.


Assuntos
Rede Nervosa/fisiologia , Técnicas de Patch-Clamp/métodos , Ultrassom , Animais , Canais de Cálcio/fisiologia , Células Cultivadas , Estimulação Elétrica/instrumentação , Estimulação Elétrica/métodos , Exocitose/fisiologia , Camundongos , Modelos Biológicos , Rede Nervosa/diagnóstico por imagem , Neurônios/metabolismo , Neurônios/fisiologia , Técnicas de Patch-Clamp/instrumentação , Proteínas SNARE/fisiologia , Canais de Sódio/metabolismo , Canais de Sódio/fisiologia , Ultrassonografia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...