Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 28
Filtrar
1.
Mol Psychiatry ; 26(9): 4864-4883, 2021 09.
Artigo em Inglês | MEDLINE | ID: mdl-32661257

RESUMO

Abnormalities of or reductions in GABAergic interneurons are implicated in the pathology of severe neuropsychiatric disorders, for which effective treatments are still elusive. Transplantation of human stem cell-derived interneurons is a promising cell-based therapy for treatment of these disorders. In mouse xenograft studies, human stem cell-derived-interneuron precursors could differentiate in vivo, but required a prolonged time of four to seven months to migrate from the graft site and integrate with the host tissue. This poses a serious roadblock for clinical translation of this approach. For transplantation to be effective, grafted neurons should migrate to affected areas at a faster rate. We have previously shown that endothelial cells of the periventricular vascular network are the natural substrates for GABAergic interneurons in the developing mouse forebrain, and provide valuable guidance cues for their long-distance migration. In addition, periventricular endothelial cells house a GABA signaling pathway with direct implications for psychiatric disease origin. In this study we translated this discovery into human, with significant therapeutic implications. We generated human periventricular endothelial cells, using human pluripotent stem cell technology, and extensively characterized its molecular, cellular, and functional properties. Co-culture of human periventricular endothelial cells with human interneurons significantly accelerated interneuron migration in vitro and led to faster migration and wider distribution of grafted interneurons in vivo, compared to neuron-only transplants. Furthermore, the co-transplantation strategy was able to rescue abnormal behavioral symptoms in a pre-clinical model of psychiatric disorder, within 1 month after transplantation. We anticipate this strategy to open new doors and facilitate exciting advances in angiogenesis-mediated treatment of psychiatric disorders.


Assuntos
Neurônios GABAérgicos , Transtornos Mentais , Animais , Movimento Celular , Células Endoteliais , Humanos , Interneurônios , Transtornos Mentais/terapia , Camundongos , Prosencéfalo
2.
J Allergy Clin Immunol ; 142(6): 1894-1908.e7, 2018 12.
Artigo em Inglês | MEDLINE | ID: mdl-29470999

RESUMO

BACKGROUND: Given their unique capacity for antigen uptake, processing, and presentation, antigen-presenting cells (APCs) are critical for initiating and regulating innate and adaptive immune responses. We have previously shown the role of nicotinamide adenine dinucleotide (NAD+) in T-cell differentiation independently of the cytokine milieu, whereas the precise mechanisms remained unknown. OBJECTIVE: The objective of this study is to further dissect the mechanism of actions of NAD+ and determine the effect of APCs on NAD+-mediated T-cell activation. METHODS: Isolated dendritic cells and bone marrow-derived mast cells (MCs) were used to characterize the mechanisms of action of NAD+ on CD4+ T-cell fate in vitro. Furthermore, NAD+-mediated CD4+ T-cell differentiation was investigated in vivo by using wild-type C57BL/6, MC-/-, MHC class II-/-, Wiskott-Aldrich syndrome protein (WASP)-/-, 5C.C7 recombination-activating gene 2 (Rag2)-/-, and CD11b-DTR transgenic mice. Finally, we tested the physiologic effect of NAD+ on the systemic immune response in the context of Listeria monocytogenes infection. RESULTS: Our in vivo and in vitro findings indicate that after NAD+ administration, MCs exclusively promote CD4+ T-cell differentiation, both in the absence of antigen and independently of major APCs. Moreover, we found that MCs mediated CD4+ T-cell differentiation independently of MHC II and T-cell receptor signaling machinery. More importantly, although treatment with NAD+ resulted in decreased MHC II expression on CD11c+ cells, MC-mediated CD4+ T-cell differentiation rendered mice resistant to administration of lethal doses of L monocytogenes. CONCLUSIONS: Collectively, our study unravels a novel cellular and molecular pathway that regulates innate and adaptive immunity through MCs exclusively and underscores the therapeutic potential of NAD+ in the context of primary immunodeficiencies and antimicrobial resistance.


Assuntos
Linfócitos T CD4-Positivos/efeitos dos fármacos , Mastócitos/efeitos dos fármacos , NAD/farmacologia , Adulto , Animais , Apresentação de Antígeno , Linfócitos T CD4-Positivos/imunologia , Diferenciação Celular/efeitos dos fármacos , Linhagem Celular , Humanos , Listeria monocytogenes , Listeriose/tratamento farmacológico , Listeriose/imunologia , Mastócitos/imunologia , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , NAD/uso terapêutico
3.
Development ; 139(17): 3136-41, 2012 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-22872083

RESUMO

Neuronal migration, a key event during brain development, remains largely unexplored in the mesencephalon, where dopaminergic (DA) and GABA neurons constitute two major neuronal populations. Here we study the migrational trajectories of DA and GABA neurons and show that they occupy ventral mesencephalic territory in a temporally and spatially specific manner. Our results from the Pitx3-deficient aphakia mouse suggest that pre-existing DA neurons modulate GABA neuronal migration to their final destination, providing novel insights and fresh perspectives concerning neuronal migration and connectivity in the mesencephalon in normal as well as diseased brains.


Assuntos
Movimento Celular/fisiologia , Neurônios Dopaminérgicos/metabolismo , Neurônios GABAérgicos/fisiologia , Mesencéfalo/embriologia , Animais , Afacia/genética , Bromodesoxiuridina , Proteínas de Homeodomínio , Imuno-Histoquímica , Camundongos , Camundongos Mutantes , Fatores de Transcrição/deficiência
4.
Stem Cells ; 32(7): 1789-804, 2014 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-24648391

RESUMO

GABAergic interneurons regulate cortical neural networks by providing inhibitory inputs, and their malfunction, resulting in failure to intricately regulate neural circuit balance, is implicated in brain diseases such as Schizophrenia, Autism, and Epilepsy. During early development, GABAergic interneuron progenitors arise from the ventral telencephalic area such as medial ganglionic eminence (MGE) and caudal ganglionic eminence (CGE) by the actions of secreted signaling molecules from nearby organizers, and migrate to their target sites where they form local synaptic connections. In this study, using combinatorial and temporal modulation of developmentally relevant dorsoventral and rostrocaudal signaling pathways (SHH, Wnt, and FGF8), we efficiently generated MGE cells from multiple human pluripotent stem cells. Most importantly, modulation of FGF8/FGF19 signaling efficiently directed MGE versus CGE differentiation. Human MGE cells spontaneously differentiated into Lhx6-expressing GABAergic interneurons and showed migratory properties. These human MGE-derived neurons generated GABA, fired action potentials, and displayed robust GABAergic postsynaptic activity. Transplantation into rodent brains results in well-contained neural grafts enriched with GABAergic interneurons that migrate in the host and mature to express somatostatin or parvalbumin. Thus, we propose that signaling modulation recapitulating normal developmental patterns efficiently generate human GABAergic interneurons. This strategy represents a novel tool in regenerative medicine, developmental studies, disease modeling, bioassay, and drug screening.


Assuntos
Encéfalo/citologia , Interneurônios/fisiologia , Células-Tronco Pluripotentes/fisiologia , Animais , Padronização Corporal , Encéfalo/embriologia , Linhagem Celular , Fatores de Crescimento de Fibroblastos/fisiologia , Neurônios GABAérgicos/fisiologia , Proteínas Hedgehog/metabolismo , Humanos , Interneurônios/transplante , Camundongos , Camundongos Endogâmicos NOD , Camundongos SCID , Células-Tronco Neurais/fisiologia , Transdução de Sinais
5.
J Neurosci ; 33(37): 14809-15, 2013 Sep 11.
Artigo em Inglês | MEDLINE | ID: mdl-24027281

RESUMO

Current models of brain development support the view that VEGF, a signaling protein secreted by neuronal cells, regulates angiogenesis and neuronal development. Here we demonstrate an autonomous and pivotal role for endothelial cell-derived VEGF that has far-reaching consequences for mouse brain development. Selective deletion of Vegf from endothelial cells resulted in impaired angiogenesis and marked perturbation of cortical cytoarchitecture. Abnormal cell clusters or heterotopias were detected in the marginal zone, and disorganization of cortical cells induced several malformations, including aberrant cortical lamination. Critical events during brain development-neuronal proliferation, differentiation, and migration were significantly affected. In addition, axonal tracts in the telencephalon were severely defective in the absence of endothelial VEGF. The unique roles of endothelial VEGF cannot be compensated by neuronal VEGF and underscores the high functional significance of endothelial VEGF for cerebral cortex development and from disease perspectives.


Assuntos
Córtex Cerebral , Células Endoteliais/metabolismo , Regulação da Expressão Gênica no Desenvolvimento/fisiologia , Neurônios/metabolismo , Fator A de Crescimento do Endotélio Vascular/metabolismo , Fatores Etários , Animais , Bromodesoxiuridina/metabolismo , Diferenciação Celular/genética , Proliferação de Células , Córtex Cerebral/citologia , Córtex Cerebral/embriologia , Córtex Cerebral/metabolismo , Embrião de Mamíferos , Feminino , Regulação da Expressão Gênica no Desenvolvimento/genética , Antígeno Ki-67/metabolismo , Laminina/metabolismo , Lectinas/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Proteínas Associadas aos Microtúbulos/metabolismo , Mutação/genética , Neovascularização Fisiológica , Molécula-1 de Adesão Celular Endotelial a Plaquetas/metabolismo , Fator A de Crescimento do Endotélio Vascular/genética
6.
Elife ; 122024 Feb 19.
Artigo em Inglês | MEDLINE | ID: mdl-38372712

RESUMO

Septic shock is characterized by an excessive inflammatory response depicted in a cytokine storm that results from invasive bacterial, fungi, protozoa, and viral infections. Non-canonical inflammasome activation is crucial in the development of septic shock promoting pyroptosis and proinflammatory cytokine production via caspase-11 and gasdermin D (GSDMD). Here, we show that NAD+ treatment protected mice toward bacterial and lipopolysaccharide (LPS)-induced endotoxic shock by blocking the non-canonical inflammasome specifically. NAD+ administration impeded systemic IL-1ß and IL-18 production and GSDMD-mediated pyroptosis of macrophages via the IFN-ß/STAT-1 signaling machinery. More importantly, NAD+ administration not only improved casp-11 KO (knockout) survival but rendered wild type (WT) mice completely resistant to septic shock via the IL-10 signaling pathway that was independent from the non-canonical inflammasome. Here, we delineated a two-sided effect of NAD+ blocking septic shock through a specific inhibition of the non-canonical inflammasome and promoting immune homeostasis via IL-10, underscoring its unique therapeutic potential.


Assuntos
Citocinas , Choque Séptico , Animais , Camundongos , Interleucina-10 , Inflamassomos , NAD , Choque Séptico/prevenção & controle , Macrófagos
7.
Sci Rep ; 13(1): 9595, 2023 06 13.
Artigo em Inglês | MEDLINE | ID: mdl-37311798

RESUMO

The proper development and function of telencephalic GABAergic interneurons is critical for maintaining the excitation and inhibition (E/I) balance in cortical circuits. Glutamate contributes to cortical interneuron (CIN) development via N-methyl-D-aspartate receptors (NMDARs). NMDAR activation requires the binding of a co-agonist, either glycine or D-serine. D-serine (co-agonist at many mature forebrain synapses) is racemized by the neuronal enzyme serine racemase (SR) from L-serine. We utilized constitutive SR knockout (SR-/-) mice to investigate the effect of D-serine availability on the development of CINs and inhibitory synapses in the prelimbic cortex (PrL). We found that most immature Lhx6 + CINs expressed SR and the obligatory NMDAR subunit NR1. At embryonic day 15, SR-/- mice had an accumulation of GABA and increased mitotic proliferation in the ganglionic eminence and fewer Gad1 + (glutamic acid decarboxylase 67 kDa; GAD67) cells in the E18 neocortex. Lhx6 + cells develop into parvalbumin (PV+) and somatostatin (Sst+) CINs. In the PrL of postnatal day (PND) 16 SR-/- mice, there was a significant decrease in GAD67+ and PV+, but not SST + CIN density, which was associated with reduced inhibitory postsynaptic potentials in layer 2/3 pyramidal neurons. These results demonstrate that D-serine availability is essential for prenatal CIN development and postnatal cortical circuit maturation.


Assuntos
Traumatismos Craniocerebrais , Neocórtex , Feminino , Gravidez , Animais , Camundongos , Interneurônios , Córtex Pré-Frontal , Ácido Glutâmico
8.
Nat Neurosci ; 11(4): 429-39, 2008 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-18344991

RESUMO

Prevailing notions of cerebral vascularization imply that blood vessels sprout passively into the brain parenchyma from pial vascular plexuses to meet metabolic needs of growing neuronal populations. Endothelial cells, building blocks of blood vessels, are thought to be homogeneous in the brain with respect to their origins, gene expression patterns and developmental mechanisms. These current notions that cerebral angiogenesis is regulated by local environmental signals contrast with current models of cell-autonomous regulation of neuronal development. Here we demonstrate that telencephalic angiogenesis in mice progresses in an orderly, ventral-to-dorsal gradient regulated by compartment-specific homeobox transcription factors. Our data offer new perspectives on intrinsic regulation of angiogenesis in the embryonic telencephalon, call for a revision of the current models of telencephalic angiogenesis and support novel roles for endothelial cells in brain development.


Assuntos
Endotélio Vascular/embriologia , Regulação da Expressão Gênica no Desenvolvimento/fisiologia , Proteínas de Homeodomínio/metabolismo , Telencéfalo/irrigação sanguínea , Telencéfalo/embriologia , Animais , Padronização Corporal/fisiologia , Circulação Cerebrovascular/fisiologia , Embrião de Mamíferos , Células Endoteliais/citologia , Células Endoteliais/metabolismo , Endotélio Vascular/metabolismo , Proteínas do Olho/genética , Proteínas do Olho/metabolismo , Feminino , Proteínas de Homeodomínio/genética , Técnicas In Vitro , Camundongos , Camundongos Knockout , Camundongos Mutantes , Neovascularização Fisiológica/fisiologia , Proteínas Nucleares/genética , Proteínas Nucleares/metabolismo , Organogênese/fisiologia , Fator de Transcrição PAX6 , Fatores de Transcrição Box Pareados/genética , Fatores de Transcrição Box Pareados/metabolismo , Gravidez , Receptor TIE-2/genética , Receptor TIE-2/metabolismo , Proteínas Repressoras/genética , Proteínas Repressoras/metabolismo , Fator Nuclear 1 de Tireoide , Fatores de Transcrição/genética , Fatores de Transcrição/metabolismo
9.
Sci Rep ; 12(1): 4922, 2022 03 22.
Artigo em Inglês | MEDLINE | ID: mdl-35318369

RESUMO

Our recent studies uncovered a novel GABA signaling pathway in embryonic forebrain endothelial cells that works independently from neuronal GABA signaling and revealed that disruptions in endothelial GABAA receptor-GABA signaling from early embryonic stages can directly contribute to the origin of psychiatric disorders. In the GABAA receptor ß3 subunit endothelial cell conditional knockout (Gabrb3ECKO) mice, the ß3 subunit is deleted selectively from endothelial cells, therefore endothelial GABAA receptors become inactivated and dysfunctional. There is a reduction in vessel densities and increased vessel morphology in the Gabrb3ECKO telencephalon that persists in the adult neocortex. Gabrb3ECKO mice show behavioral deficits such as impaired reciprocal social interactions, communication deficits, heightened anxiety, and depression. Here, we characterize the functional changes in Gabrb3ECKO mice by evaluating cortical blood flow, examine the consequences of loss of endothelial Gabrb3 on cardiac tissue, and define more in-depth altered behaviors. Red blood cell velocity and blood flow were increased in the cortical microcirculation of the Gabrb3ECKO mice. The Gabrb3ECKO mice had a reduction in vessel densities in the heart, similar to the brain; exhibited wavy, myocardial fibers, with elongated 'worm-like' nuclei in their cardiac histology, and developed hypertension. Additional alterations in behavioral function were observed in the Gabrb3ECKO mice such as increased spontaneous exploratory activity and rearing in an open field, reduced short term memory, decreased ambulatory activity in CLAMS testing, and altered prepulse inhibition to startle, an important biomarker of psychiatric diseases such as schizophrenia. Our results imply that vascular Gabrb3 is a key player in the brain as well as the heart, and its loss in both organs can lead to concurrent development of psychiatric and cardiac dysfunction.


Assuntos
Hipertensão , Receptores de GABA-A , Animais , Células Endoteliais/metabolismo , Hipertensão/genética , Hipertensão/metabolismo , Camundongos , Camundongos Knockout , Receptores de GABA-A/metabolismo , Ácido gama-Aminobutírico/metabolismo
10.
Hippocampus ; 20(5): 608-20, 2010 May.
Artigo em Inglês | MEDLINE | ID: mdl-19530222

RESUMO

The basement membrane (BM) is a specialized form of extracellular matrix (ECM) underlying epithelia and endothelia and surrounding many types of mesenchymal cells. Nidogen, along with collagen IV and laminin, is a major component of BMs. Although certain ECM proteins such as laminin or reelin influence neuronal function via interactions with cell-surface receptors such as integrins, behavioral neurological impairments due to deficits of BM components have been recognized only recently. Here, alterations in neuronal network function underlying these behavioral changes are revealed. Using nidogen-1 knockout mice, with or without additional heterozygous nidogen-2 knockout (NID1(-/-)/NID2(+/+) or NID1(-/-)/NID2(+/-)), we demonstrate that nidogen is essential for normal neuronal network excitability and plasticity. In nidogen-1 knockouts, seizurelike behavior occurs, and epileptiform spiking was seen in hippocampal in vivo EEG recordings. In vitro, hippocampal field potential recordings revealed that lack of nidogen-1, while not causing conspicuous morphological changes, led to the appearance of spontaneous and evoked epileptiform activity, significant increase of the input/output ratio of synaptically evoked responses in CA1 and dentate gyrus, as well as of paired pulse accentuation, and loss of perforant-path long-term synaptic potentiation. Nidogen-1 is thus essential for normal network excitability and plasticity.


Assuntos
Potenciais Evocados/fisiologia , Hipocampo/citologia , Glicoproteínas de Membrana/fisiologia , Plasticidade Neuronal/fisiologia , Sinapses/fisiologia , Fatores Etários , Análise de Variância , Animais , Biofísica , Proteínas de Ligação ao Cálcio , Moléculas de Adesão Celular , Estimulação Elétrica/métodos , Eletroencefalografia/métodos , Potenciais Evocados/efeitos dos fármacos , Potenciais Evocados/genética , Antagonistas de Aminoácidos Excitatórios/farmacologia , Proteína Glial Fibrilar Ácida/metabolismo , Hipocampo/fisiologia , Técnicas In Vitro , Laminina/metabolismo , Glicoproteínas de Membrana/deficiência , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Modelos Neurológicos , Movimento , Plasticidade Neuronal/genética , Propionatos/farmacologia , Proteína Reelina , Sinapses/efeitos dos fármacos
11.
J Vis Exp ; (155)2020 01 23.
Artigo em Inglês | MEDLINE | ID: mdl-32065131

RESUMO

Role of brain vasculature in nervous system development and etiology of brain disorders is increasingly gaining attention. Our recent studies have identified a special population of vascular cells, the periventricular endothelial cells, that play a critical role in the migration and distribution of forebrain GABAergic interneurons during embryonic development. This, coupled with their cell-autonomous functions, alludes to novel roles of periventricular endothelial cells in the pathology of neuropsychiatric disorders like schizophrenia, epilepsy, and autism. Here, we have described three different in vitro assays that collectively evaluate the functions of periventricular endothelial cells and their interaction with GABAergic interneurons. Use of these assays, particularly in a human context, will allow us to identify the link between periventricular endothelial cells and brain disorders. These assays are simple, low cost, and reproducible, and can be easily adapted to any adherent cell type.


Assuntos
Técnicas de Cocultura/métodos , Células Progenitoras Endoteliais/metabolismo , Neurônios GABAérgicos/metabolismo , Células-Tronco/fisiologia , Movimento Celular , Feminino , Humanos , Masculino
12.
Sci Adv ; 6(41)2020 10.
Artigo em Inglês | MEDLINE | ID: mdl-33036972

RESUMO

Intrinsic defects within blood vessels from the earliest developmental time points can directly contribute to psychiatric disease origin. Here, we show that nicotinamide adenine dinucleotide (NAD+), administered during a critical window of prenatal development, in a mouse model with dysfunctional endothelial γ-aminobutyric acid type A (GABAA) receptors (Gabrb3 endothelial cell knockout mice), results in a synergistic repair of impaired angiogenesis and normalization of brain development, thus preventing the acquisition of abnormal behavioral symptoms. The prenatal NAD+ treatment stimulated extensive cellular and molecular changes in endothelial cells and restored blood vessel formation, GABAergic neuronal development, and forebrain morphology by recruiting an alternate pathway for cellular repair, via previously unknown transcriptional mechanisms and purinergic receptor signaling. Our findings illustrate a novel and powerful role for NAD+ in sculpting prenatal brain development that has profound implications for rescuing brain blood flow in a permanent and irreversible manner, with long-lasting consequences for mental health outcome.


Assuntos
Células Endoteliais , NAD , Animais , Células Endoteliais/metabolismo , Feminino , Neurônios GABAérgicos/metabolismo , Camundongos , Camundongos Knockout , NAD/metabolismo , Gravidez , Prosencéfalo/metabolismo , Receptores de GABA-A/metabolismo
13.
Sci Rep ; 9(1): 16256, 2019 11 07.
Artigo em Inglês | MEDLINE | ID: mdl-31700116

RESUMO

The developing cerebral cortex uses a complex developmental plan involving angiogenesis, neurogenesis and neuronal migration. Our recent studies have highlighted the importance of endothelial cell secreted GABA signaling in the embryonic forebrain and established novel autonomous links between blood vessels and the origin of neuropsychiatric diseases. A GABA pathway operates in both endothelial cells and GABAergic neurons of the embryonic telencephalon; however, while the neuronal GABA pathway has been extensively studied, little is known about the endothelial GABA pathway. Our recently generated Vgat endothelial cell knockout mouse model that blocks GABA release from endothelial cells, serves as a new tool to study how endothelial GABA signaling shapes angiogenesis and neurovascular interactions during prenatal development. Quantitative gene expression profiling reveals that the endothelial GABA signaling pathway influences genes connected to specific processes like endothelial cell proliferation, differentiation, migration, tight junction formation, vascular sprouting and integrity. It also shows how components of the neuronal GABA pathway, for instance receptor mediated signaling, cell cycle related components and transcription factors are affected in the absence of endothelial GABA release. Taken together, our findings delineate the close relationship between vascular and nervous systems that begin early in embryogenesis establishing their future interactions and interdependence.


Assuntos
Comunicação Autócrina , Células Endoteliais/metabolismo , Comunicação Parácrina , Prosencéfalo/metabolismo , Receptores de GABA/metabolismo , Transdução de Sinais , Ácido gama-Aminobutírico/metabolismo , Animais , Diferenciação Celular , Biologia Computacional/métodos , Desenvolvimento Embrionário , Perfilação da Expressão Gênica , Regulação da Expressão Gênica no Desenvolvimento , Masculino , Camundongos , Camundongos Knockout , Modelos Biológicos , Neurogênese , Prosencéfalo/embriologia , Transcriptoma
14.
Open Neurol J ; 13: 1-9, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-30984305

RESUMO

The mammalian brain receives the lion's share of the body's blood supply and is a highly vascularized organ. The vascular and nervous systems arise at two distinct time points of embryogenesis; however, their functions tend to overlap or complement each other in the growth promoting milieu of the embryonic Central Nervous System (CNS). The pre-existing idea that mental disorders are a direct result from defects solely in neuronal populations and networks is gradually changing. Several studies have implicated blood vessel pathologies and blood flow changes in mental health disorders. Our own studies provide new perspectives as to how intrinsic defects in periventricular endothelial cells, from the earliest developmental time points can lead to the origin of mental health disorders such as schizophrenia, autism spectrum disorders (ASD), anxiety, and depression, thereby establishing direct links. In this article, we provide an overview of how the endothelial cell compartment in the brain is now gaining attention in the context of mental health disorders.

15.
Front Physiol ; 10: 1583, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-32038280

RESUMO

Our understanding of the etiological mechanisms leading up to epilepsy has undergone radical changes over time due to more insights into the complexity of the disease. The traditional hypothesis emphasized network hyperexcitability and an imbalance of inhibition and excitation, eventually leading to seizures. In this context, the contribution of the vascular system, and particularly the interactions between blood vessels and neuronal tissue, came into focus only recently. Thus, one highly exciting causative or contributing factor of epileptogenesis is the disruption of the blood-brain barrier (BBB) in the context of not only posttraumatic epilepsy, but also other etiologies. This hypothesis is now recognized as a synergistic mechanism that can give rise to epilepsy, and BBB repair for restoration of cerebrovascular integrity is considered a therapeutic alternative. Endothelial cells lining the inner surface of blood vessels are an integral component of the BBB system. Sealed by tight junctions, they are crucial in maintaining homeostatic activities of the brain, as well as acting as an interface in the neurovascular unit. Additional potential vascular mechanisms such as inflammation, altered neurovascular coupling, or changes in blood flow that can modulate neuronal circuit activity have been implicated in epilepsy. Our own work has shown how intrinsic defects within endothelial cells from the earliest developmental time points, which preclude neuronal changes, can lead to vascular abnormalities and autonomously support the development of hyperexcitability and epileptiform activity. In this article, we review the importance of vascular integrity and signaling for network excitability and epilepsy by highlighting complementary basic and clinical research studies and by outlining possible novel therapeutic strategies.

16.
Cell Res ; 28(2): 221-248, 2018 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-29086765

RESUMO

The cerebral cortex is essential for integration and processing of information that is required for most behaviors. The exquisitely precise laminar organization of the cerebral cortex arises during embryonic development when neurons migrate successively from ventricular zones to coalesce into specific cortical layers. While radial glia act as guide rails for projection neuron migration, pre-formed vascular networks provide support and guidance cues for GABAergic interneuron migration. This study provides novel conceptual and mechanistic insights into this paradigm of vascular-neuronal interactions, revealing new mechanisms of GABA and its receptor-mediated signaling via embryonic forebrain endothelial cells. With the use of two new endothelial cell specific conditional mouse models of the GABA pathway (Gabrb3ΔTie2-Cre and VgatΔTie2-Cre), we show that partial or complete loss of GABA release from endothelial cells during embryogenesis results in vascular defects and impairs long-distance migration and positioning of cortical interneurons. The downstream effects of perturbed endothelial cell-derived GABA signaling are critical, leading to lasting changes to cortical circuits and persistent behavioral deficits. Furthermore, we illustrate new mechanisms of activation of GABA signaling in forebrain endothelial cells that promotes their migration, angiogenesis and acquisition of blood-brain barrier properties. Our findings uncover and elucidate a novel endothelial GABA signaling pathway in the CNS that is distinct from the classical neuronal GABA signaling pathway and shed new light on the etiology and pathophysiology of neuropsychiatric diseases, such as autism spectrum disorders, epilepsy, anxiety, depression and schizophrenia.


Assuntos
Córtex Cerebral/embriologia , Células Endoteliais/metabolismo , Neurônios GABAérgicos/fisiologia , Interneurônios/fisiologia , Ácido gama-Aminobutírico/metabolismo , Animais , Comportamento Animal , Movimento Celular , Córtex Cerebral/irrigação sanguínea , Córtex Cerebral/citologia , Feminino , Perfilação da Expressão Gênica , Masculino , Camundongos , Camundongos Knockout , Camundongos Transgênicos , Modelos Animais , Transtornos do Neurodesenvolvimento/etiologia , Transtornos do Neurodesenvolvimento/fisiopatologia , Neurogênese/fisiologia , Fenótipo , Gravidez , RNA/genética , Receptores de GABA-A/fisiologia , Proteínas Vesiculares de Transporte de Aminoácidos Inibidores/metabolismo
17.
Int J Tryptophan Res ; 10: 1178646917713491, 2017.
Artigo em Inglês | MEDLINE | ID: mdl-28659716

RESUMO

Increasing evidence underscores the interesting ability of tryptophan to regulate immune responses. However, the exact mechanisms of tryptophan's immune regulation remain to be determined. Tryptophan catabolism via the kynurenine pathway is known to play an important role in tryptophan's involvement in immune responses. Interestingly, quinolinic acid, which is a neurotoxic catabolite of the kynurenine pathway, is the major pathway for the de novo synthesis of nicotinamide adenine dinucleotide (NAD+). Recent studies have shown that NAD+, a natural coenzyme found in all living cells, regulates immune responses and creates homeostasis via a novel signaling pathway. More importantly, the immunoregulatory properties of NAD+ are strongly related to the overexpression of tryptophan hydroxylase 1 (Tph1). This review provides recent knowledge of tryptophan and NAD+ and their specific and intriguing roles in the immune system. Furthermore, it focuses on the mechanisms by which tryptophan regulates NAD+ synthesis as well as innate and adaptive immune responses.

18.
Brain Res ; 1073-1074: 139-45, 2006 Feb 16.
Artigo em Inglês | MEDLINE | ID: mdl-16458269

RESUMO

Early onset torsion dystonia is characterized by involuntary movements and distorted postures and is usually caused by a 3-bp (GAG) deletion in the DYT1 (TOR1A) gene. DYT1 codes for torsinA, a member of the AAA+ family of proteins, implicated in membrane recycling and chaperone functions. A close relative, torsinB may be involved in similar cellular functions. We investigated torsinA and torsinB message and protein levels in the developing mouse brain. TorsinA expression was highest during prenatal and early postnatal development (until postnatal day 14; P14), whereas torsinB expression was highest during late postnatal periods (from P14 onwards) and in the adult. In addition, significant regional variation in the expression of the two torsins was seen within the developing brain. Thus, torsinA expression was highest in the cerebral cortex from embryonic day 15 (E15)-E17 and in the striatum from E17-P7, while torsinB was highest in the cerebral cortex between P7-P14 and in the striatum from P7-P30. TorsinA was also highly expressed in the thalamus from P0-P7 and in the cerebellum from P7-P14. Although functional significance of the patterns of torsinA and B expression in the developing brain remains to be established, our findings provide a basis for investigating the role of torsins in specific processes such as neurogenesis, neuronal migration, axon/dendrite development, and synaptogenesis.


Assuntos
Encéfalo/metabolismo , Proteínas de Transporte/metabolismo , Regulação da Expressão Gênica no Desenvolvimento/fisiologia , Chaperonas Moleculares/metabolismo , Fatores Etários , Animais , Northern Blotting/métodos , Western Blotting/métodos , Encéfalo/embriologia , Encéfalo/crescimento & desenvolvimento , Proteínas de Transporte/genética , Feminino , Camundongos , Chaperonas Moleculares/genética , Peso Molecular , Gravidez
19.
Biochem Pharmacol ; 64(2): 339-49, 2002 Jul 15.
Artigo em Inglês | MEDLINE | ID: mdl-12123755

RESUMO

Nonsteroidal anti-inflammatory drugs (NSAIDs) are known to cause small intestinal damage but the pathogenesis of this toxicity is not well established. Intestinal epithelial cells are thought to be affected by these drugs in the course of their absorption. These cells are of different types, viz. villus, middle and crypt cells. There is little information on which of these cells, if any, are particularly vulnerable to the effects of NSAIDs. This paper aimed to study the effects of indomethacin, an NSAID commonly used in toxicity studies, on different populations of enterocytes. Effects of the drug were assessed in terms of oxidative damage, mitotic activity, mitochondrial function and lipid composition in enterocytes isolated from the small intestine of rats that had been orally administered indomethacin. In addition, the effects of arginine and zinc in protecting against such changes were assessed. Cell viability, tetrazolium dye (MTT) reduction and oxygen uptake were significantly reduced in villus tip cells from rats dosed with the drug. Thymidine uptake was higher in the crypt cell fraction from these rats. Similarly, products of lipid peroxidation were elevated in the villus tip cells with a corresponding decrease in the level of the anti-oxidant, alpha-tocopherol. In isolated mitochondrial preparations from various enterocyte fractions, significant functional impairment and altered lipid composition were seen mainly in mitochondria from villus cells. Arginine and zinc pre-treatment were found to protect against these effects. These results suggest for the first time that the villus tip cells are more vulnerable to the damaging effects of indomethacin and that oxidative stress is possibly involved in this damage.


Assuntos
Anti-Inflamatórios não Esteroides/farmacologia , Enterócitos/efeitos dos fármacos , Indometacina/farmacologia , Mitocôndrias/efeitos dos fármacos , Estresse Oxidativo/fisiologia , Animais , Arginina/farmacologia , Interações Medicamentosas , Enterócitos/enzimologia , Enterócitos/metabolismo , Feminino , Lipídeos/química , Masculino , Mitocôndrias/fisiologia , Peroxidase/metabolismo , Ratos , Zinco/farmacologia
20.
Biomol Concepts ; 5(5): 371-82, 2014 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-25367618

RESUMO

Midbrain GABA neurons, endowed with multiple morphological, physiological and molecular characteristics as well as projection patterns are key players interacting with diverse regions of the brain and capable of modulating several aspects of behavior. The diversity of these GABA neuronal populations based on their location and function in the dorsal, medial or ventral midbrain has challenged efforts to rapidly uncover their developmental regulation. Here we review recent developments that are beginning to illuminate transcriptional control of GABA neurons in the embryonic midbrain (mesencephalon) and discuss its implications for understanding and treatment of neurological and psychiatric illnesses.


Assuntos
Neurônios GABAérgicos/fisiologia , Mesencéfalo/embriologia , Ácido gama-Aminobutírico/metabolismo , Animais , Neurônios GABAérgicos/patologia , Regulação da Expressão Gênica no Desenvolvimento , Humanos , Transtornos Mentais/patologia , Mesencéfalo/citologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA