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

Base de dados
Tipo de documento
País de afiliação
Intervalo de ano de publicação
1.
Development ; 149(20)2022 10 15.
Artigo em Inglês | MEDLINE | ID: mdl-35748297

RESUMO

Oligodendrocytes, the myelinating cells of the central nervous system, possess great potential for disease modeling and cell transplantation-based therapies for leukodystrophies. However, caveats to oligodendrocyte differentiation protocols ( Ehrlich et al., 2017; Wang et al., 2013; Douvaras and Fossati, 2015) from human embryonic stem and induced pluripotent stem cells (iPSCs), which include slow and inefficient differentiation, and tumorigenic potential of contaminating undifferentiated pluripotent cells, are major bottlenecks towards their translational utility. Here, we report the rapid generation of human oligodendrocytes by direct lineage conversion of human dermal fibroblasts (HDFs). We show that the combination of the four transcription factors OLIG2, SOX10, ASCL1 and NKX2.2 is sufficient to convert HDFs to induced oligodendrocyte precursor cells (iOPCs). iOPCs resemble human primary and iPSC-derived OPCs based on morphology and transcriptomic analysis. Importantly, iOPCs can differentiate into mature myelinating oligodendrocytes in vitro and in vivo. Finally, iOPCs derived from patients with Pelizaeus Merzbacher disease, a hypomyelinating leukodystrophy caused by mutations in the proteolipid protein 1 (PLP1) gene, showed increased cell death compared with iOPCs from healthy donors. Thus, human iOPCs generated by direct lineage conversion represent an attractive new source for human cell-based disease models and potentially myelinating cell grafts.


Assuntos
Células-Tronco Pluripotentes Induzidas , Doença de Pelizaeus-Merzbacher , Diferenciação Celular/fisiologia , Fibroblastos , Humanos , Células-Tronco Pluripotentes Induzidas/metabolismo , Oligodendroglia/metabolismo , Doença de Pelizaeus-Merzbacher/genética , Doença de Pelizaeus-Merzbacher/metabolismo , Doença de Pelizaeus-Merzbacher/terapia
2.
Lancet ; 390(10111): 2481-2489, 2017 Dec 02.
Artigo em Inglês | MEDLINE | ID: mdl-29029896

RESUMO

BACKGROUND: Multiple sclerosis is a degenerative inflammatory disease of the CNS characterised by immune-mediated destruction of myelin and progressive neuroaxonal loss. Myelin in the CNS is a specialised extension of the oligodendrocyte plasma membrane and clemastine fumarate can stimulate differentiation of oligodendrocyte precursor cells in vitro, in animal models, and in human cells. We aimed to analyse the efficacy and safety of clemastine fumarate as a treatment for patients with multiple sclerosis. METHODS: We did this single-centre, 150-day, double-blind, randomised, placebo-controlled, crossover trial (ReBUILD) in patients with relapsing multiple sclerosis with chronic demyelinating optic neuropathy on stable immunomodulatory therapy. Patients who fulfilled international panel criteria for diagnosis with disease duration of less than 15 years were eligible. Patients were randomly assigned (1:1) via block randomisation using a random number generator to receive either clemastine fumarate (5·36 mg orally twice daily) for 90 days followed by placebo for 60 days (group 1), or placebo for 90 days followed by clemastine fumarate (5·36 mg orally twice daily) for 60 days (group 2). The primary outcome was shortening of P100 latency delay on full-field, pattern-reversal, visual-evoked potentials. We analysed by intention to treat. The trial is registered with ClinicalTrials.gov, number NCT02040298. FINDINGS: Between Jan 1, 2014, and April 11, 2015, we randomly assigned 50 patients to group 1 (n=25) or group 2 (n=25). All patients completed the study. The primary efficacy endpoint was met with clemastine fumarate treatment, which reduced the latency delay by 1·7 ms/eye (95% CI 0·5-2·9; p=0·0048) when analysing the trial as a crossover. Clemastine fumarate treatment was associated with fatigue, but no serious adverse events were reported. INTERPRETATION: To our knowledge, this is the first randomised controlled trial to document efficacy of a remyelinating drug for the treatment of chronic demyelinating injury in multiple sclerosis. Our findings suggest that myelin repair can be achieved even following prolonged damage. FUNDING: University of California, San Francisco and the Rachleff Family.


Assuntos
Clemastina/uso terapêutico , Esclerose Múltipla/tratamento farmacológico , Adulto , Estudos Cross-Over , Método Duplo-Cego , Potenciais Evocados Visuais/efeitos dos fármacos , Feminino , Humanos , Masculino , Remielinização/efeitos dos fármacos , Tomografia de Coerência Óptica , Resultado do Tratamento
3.
J Neurosci ; 36(30): 7925-35, 2016 07 27.
Artigo em Inglês | MEDLINE | ID: mdl-27466337

RESUMO

UNLABELLED: Remyelinating therapies seek to promote restoration of function and normal cellular architecture following demyelination in diseases, such as multiple sclerosis (MS). Functional screening for small molecules or novel targets for remyelination is a major hurdle to the identification and development of rational therapeutics for MS. Recent findings and technical advances provide us with a unique opportunity to provide insight into the cell autonomous mechanisms for remyelination and address this unmet need. Upon screening a G-protein-coupled receptor small-molecule library, we report the identification of a cluster of κ-opioid receptor (KOR) agonists that significantly promotes oligodendrocyte differentiation and myelination. KOR agonists were validated in purified rat oligodendroglial cultures, and the (±)U-50488 compound proved to be most effective for differentiation. (±)U-50488 treatment significantly enhances differentiation and myelination in purified oligodendroglial cocultures and greatly accelerates the kinetics of remyelination in vivo after focal demyelination with lysolecithin. The effect of (±)U-50488 is attenuated by KOR antagonists and completely abolished in KOR-null oligodendroglia. Conditional deletion of KOR in murine oligodendrocyte precursor cells (OPCs) greatly inhibits remyelination after focal demyelination lacking any response to (±)U-50488 treatment. To determine whether agonism of KOR represents a feasible therapeutic approach, human induced pluripotent stem cell-derived OPCs were treated with (±)U-50488. Consistent with findings, differentiation of human OPCs into mature oligodendrocytes was significantly enhanced. Together, KOR is a therapeutic target to consider for future remyelination therapy. SIGNIFICANCE STATEMENT: Remyelination represents a promising strategy to achieve functional recovery in demyelinating diseases, like MS. Thus, identification of potent compounds and targets that promote remyelination represents a critically unmet need. This study reports a cluster of compounds that are highly effective in enhancing remyelination and identifies κ-opioid receptor (KOR) as a positive regulator for oligodendroglial differentiation, implicating KOR agonism as a potential strategy to accelerate remyelination.


Assuntos
Doenças Desmielinizantes/fisiopatologia , Bainha de Mielina/fisiologia , Regeneração Nervosa/fisiologia , Oligodendroglia/citologia , Oligodendroglia/fisiologia , Receptores Opioides kappa/metabolismo , Animais , Diferenciação Celular/fisiologia , Proliferação de Células/fisiologia , Células Cultivadas , Doenças Desmielinizantes/tratamento farmacológico , Sistemas de Liberação de Medicamentos/métodos , Feminino , Masculino , Camundongos , Neurogênese/fisiologia , Receptores Opioides kappa/agonistas
4.
Glia ; 65(12): 2024-2037, 2017 12.
Artigo em Inglês | MEDLINE | ID: mdl-28856805

RESUMO

Inflammation is a major risk factor for neonatal white matter injury (NWMI), which is associated with later development of cerebral palsy. Although recent studies have demonstrated maturation arrest of oligodendrocyte progenitor cells (OPCs) in NWMI, the identity of inflammatory mediators with direct effects on OPCs has been unclear. Here, we investigated downstream effects of pro-inflammatory IL-1ß to induce cyclooxygenase-2 (COX2) and prostaglandin E2 (PGE2) production in white matter. First, we assessed COX2 expression in human fetal brain and term neonatal brain affected by hypoxic-ischemic encephalopathy (HIE). In the developing human brain, COX2 was expressed in radial glia, microglia, and endothelial cells. In human term neonatal HIE cases with subcortical WMI, COX2 was strongly induced in reactive astrocytes with "A2" reactivity. Next, we show that OPCs express the EP1 receptor for PGE2, and PGE2 acts directly on OPCs to block maturation in vitro. Pharmacologic blockade with EP1-specific inhibitors (ONO-8711, SC-51089), or genetic deficiency of EP1 attenuated effects of PGE2. In an IL-1ß-induced model of NWMI, astrocytes also exhibit "A2" reactivity and induce COX2. Furthermore, in vivo inhibition of COX2 with Nimesulide rescues hypomyelination and behavioral impairment. These findings suggest that neonatal white matter astrocytes can develop "A2" reactivity that contributes to OPC maturation arrest in NWMI through induction of COX2-PGE2 signaling, a pathway that can be targeted for neonatal neuroprotection.


Assuntos
Ciclo-Oxigenase 2/metabolismo , Dinoprostona/metabolismo , Oligodendroglia/metabolismo , Substância Branca/citologia , Substância Branca/efeitos dos fármacos , Animais , Animais Recém-Nascidos , Astrócitos/efeitos dos fármacos , Células Cultivadas , Feminino , Feto/citologia , Regulação da Expressão Gênica no Desenvolvimento/efeitos dos fármacos , Regulação da Expressão Gênica no Desenvolvimento/fisiologia , Proteína Glial Fibrilar Ácida/metabolismo , Humanos , Interleucina-1beta/farmacologia , L-Lactato Desidrogenase/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Oligodendroglia/efeitos dos fármacos , Ratos , Transdução de Sinais/efeitos dos fármacos , Transdução de Sinais/fisiologia , Memória Espacial/efeitos dos fármacos , Memória Espacial/fisiologia , Substância Branca/crescimento & desenvolvimento , Substância Branca/metabolismo
5.
Acta Neuropathol ; 130(2): 171-83, 2015 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-25975378

RESUMO

Human congenital central hypoventilation syndrome (CCHS), resulting from mutations in transcription factor PHOX2B, manifests with impaired responses to hypoxemia and hypercapnia especially during sleep. To identify brainstem structures developmentally affected in CCHS, we analyzed two postmortem neonatal-lethal cases with confirmed polyalanine repeat expansion (PARM) or Non-PARM (PHOX2B∆8) mutation of PHOX2B. Both human cases showed neuronal losses within the locus coeruleus (LC), which is important for central noradrenergic signaling. Using a conditionally active transgenic mouse model of the PHOX2B∆8 mutation, we found that early embryonic expression (

Assuntos
Hipoventilação/congênito , Locus Cerúleo/crescimento & desenvolvimento , Locus Cerúleo/patologia , Apneia do Sono Tipo Central/patologia , Apneia do Sono Tipo Central/fisiopatologia , Idade de Início , Animais , Modelos Animais de Doenças , Proteínas de Homeodomínio/genética , Proteínas de Homeodomínio/metabolismo , Humanos , Hipoventilação/genética , Hipoventilação/patologia , Hipoventilação/fisiopatologia , Recém-Nascido , Recém-Nascido Prematuro , Locus Cerúleo/fisiopatologia , Masculino , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Mutação , Neurogênese/fisiologia , Neurônios/patologia , Neurônios/fisiologia , Respiração , Apneia do Sono Tipo Central/genética , Técnicas de Cultura de Tecidos , Fatores de Transcrição/genética , Fatores de Transcrição/metabolismo
6.
Ann Neurol ; 72(5): 750-65, 2012 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-22941903

RESUMO

OBJECTIVE: Pathological findings in neonatal brain injury associated with preterm birth include focal and/or diffuse white matter injury (WMI). Despite the heterogeneous nature of this condition, reactive astrogliosis and microgliosis are frequently observed. Thus, molecular mechanisms by which glia activation contribute to WMI were investigated. METHODS: Postmortem brains of neonatal brain injury were investigated to identify molecular features of reactive astrocytes. The contribution of astrogliosis to WMI was further tested in a mouse model in genetically engineered mice. RESULTS: Activated STAT3 signaling in reactive astrocytes was found to be a common feature in postmortem brains of neonatal brain injury. In a mouse model of neonatal WMI, conditional deletion of STAT3 in astrocytes resulted in exacerbated WMI, which was associated with delayed maturation of oligodendrocytes. Mechanistically, the delay occurred in association with overexpression of transforming growth factor (TGF)ß-1 in microglia, which in healthy controls decreased with myelin maturation in an age-dependent manner. TGFß-1 directly and dose-dependently inhibited the maturation of purified oligodendrocyte progenitors, and pharmacological inhibition of TGFß-1 signaling in vivo reversed the delay in myelin development. Factors secreted from STAT3-deficient astrocytes promoted elevated TGFß-1 production in cultured microglia compared to wild-type astrocytes. INTERPRETATION: These results suggest that myelin development is regulated by a mechanism involving crosstalk between microglia and oligodendrocyte progenitors. Reactive astrocytes may modify this signaling in a STAT3-dependent manner, preventing the pathological expression of TGFß-1 in microglia and the impairment of oligodendrocyte maturation.


Assuntos
Astrócitos/metabolismo , Lesões Encefálicas/complicações , Lesões Encefálicas/patologia , Gliose/etiologia , Bainha de Mielina/metabolismo , Fator de Transcrição STAT3/metabolismo , Fatores Etários , Animais , Animais Recém-Nascidos , Astrócitos/química , Astrócitos/efeitos dos fármacos , Benzamidas/farmacologia , Benzamidas/uso terapêutico , Diferenciação Celular/efeitos dos fármacos , Diferenciação Celular/genética , Proliferação de Células/efeitos dos fármacos , Células Cultivadas , Meios de Cultivo Condicionados/farmacologia , Dioxóis/farmacologia , Dioxóis/uso terapêutico , Modelos Animais de Doenças , Ensaio de Imunoadsorção Enzimática , Feminino , Regulação da Expressão Gênica no Desenvolvimento/efeitos dos fármacos , Proteína Glial Fibrilar Ácida/metabolismo , Gliose/tratamento farmacológico , Humanos , Lactente , Recém-Nascido , Lipopolissacarídeos/toxicidade , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Mudanças Depois da Morte , Receptores de Fatores de Crescimento Transformadores beta/antagonistas & inibidores , Fator de Transcrição STAT3/deficiência , Transdução de Sinais/efeitos dos fármacos , Transdução de Sinais/genética , Proteína Smad2/metabolismo , Células-Tronco/efeitos dos fármacos , Fator de Crescimento Transformador beta1/metabolismo
7.
Proc Natl Acad Sci U S A ; 107(45): 19555-60, 2010 Nov 09.
Artigo em Inglês | MEDLINE | ID: mdl-20978211

RESUMO

The neuropeptide vasoactive intestinal peptide (VIP) has been shown to inhibit macrophage proinflammatory actions, promote a positive Th2/Th1 balance, and stimulate regulatory T-cell production. The fact that this peptide is highly efficacious in animal models of inflammatory diseases such as collagen-induced arthritis and experimental autoimmune encephalomyelitis (EAE) suggests that the endogenous peptide might normally provide protection against such pathologies. We thus studied the response of VIP-deficient (i.e., VIP KO) mice to myelin oligodendrocyte protein-induced EAE. Surprisingly, VIP KO mice were almost completely resistant to EAE, with delayed onset and mild or absent clinical profile. Despite this, flow cytometric analyses and antigen-rechallenge experiments indicated that myelin oligodendrocyte protein-treated VIP KO mice exhibited robust Th1/Th17 cell inductions and antigen-specific proliferation and cytokine responses. Moreover, adoptive transfer of lymphocytes from immunized VIP KO mice to WT recipients resulted in full-blown EAE, supporting their encephalitogenic potential. In contrast, transfer of encephalitogenic WT cells to VIP KO hosts did not produce EAE, suggesting that loss of VIP specifically affected the effector phase of the disease. Histological analyses indicated that CD4 T cells entered the meningeal and perivascular areas of VIP-deficient mice, but that parenchymal infiltration was strongly impaired. Finally, VIP pretreatment of VIP KO mice before immunization was able to restore their sensitivity to EAE. These results indicate that VIP plays an unanticipated permissive and/or proinflammatory role in the propagation of the inflammatory response in the CNS, a finding with potential therapeutic relevance in autoimmune neuroinflammatory diseases such as multiple sclerosis.


Assuntos
Movimento Celular/imunologia , Sistema Nervoso Central/imunologia , Encefalomielite Autoimune Experimental/imunologia , Linfócitos T/imunologia , Peptídeo Intestinal Vasoativo/deficiência , Peptídeo Intestinal Vasoativo/imunologia , Animais , Doenças Autoimunes/etiologia , Citocinas/biossíntese , Encefalomielite Autoimune Experimental/induzido quimicamente , Encefalomielite Autoimune Experimental/etiologia , Inflamação/etiologia , Ativação Linfocitária , Camundongos , Camundongos Knockout , Glicoproteína Associada a Mielina/farmacologia , Células Th1 , Células Th17
8.
Neuron ; 107(3): 436-453.e12, 2020 08 05.
Artigo em Inglês | MEDLINE | ID: mdl-32485136

RESUMO

New methods for investigating human astrocytes are urgently needed, given their critical role in the central nervous system. Here we show that CD49f is a novel marker for human astrocytes, expressed in fetal and adult brains from healthy and diseased individuals. CD49f can be used to purify fetal astrocytes and human induced pluripotent stem cell (hiPSC)-derived astrocytes. We provide single-cell and bulk transcriptome analyses of CD49f+ hiPSC-astrocytes and demonstrate that they perform key astrocytic functions in vitro, including trophic support of neurons, glutamate uptake, and phagocytosis. Notably, CD49f+ hiPSC-astrocytes respond to inflammatory stimuli, acquiring an A1-like reactive state, in which they display impaired phagocytosis and glutamate uptake and fail to support neuronal maturation. Most importantly, we show that conditioned medium from human reactive A1-like astrocytes is toxic to human and rodent neurons. CD49f+ hiPSC-astrocytes are thus a valuable resource for investigating human astrocyte function and dysfunction in health and disease.


Assuntos
Astrócitos/metabolismo , Células-Tronco Pluripotentes Induzidas/metabolismo , Integrina alfa6/metabolismo , Doença de Alzheimer/metabolismo , Animais , Astrócitos/fisiologia , Biomarcadores/metabolismo , Citometria de Fluxo , Perfilação da Expressão Gênica , Ácido Glutâmico/metabolismo , Humanos , Inflamação/metabolismo , Inflamação/fisiopatologia , Camundongos , Técnicas de Patch-Clamp , Fagocitose/fisiologia , RNA-Seq , Análise de Célula Única
9.
Dev Biol ; 313(1): 359-70, 2008 Jan 01.
Artigo em Inglês | MEDLINE | ID: mdl-18036580

RESUMO

Hedgehog (Hh) proteins and cAMP-dependent protein kinase A (PKA) generally play opposing roles in developmental patterning events. Humans and mice heterozygous for mutations in the sonic hedgehog (Shh) receptor gene patched-1 (ptc1) have an increased incidence of certain types of cancer, including medulloblastoma (MB), a highly aggressive tumor of the cerebellum. Despite the importance of PKA in Hh signaling, little is known about how PKA activity is regulated in the context of Hh signaling, or the consequences of improper regulation. One molecule that can influence PKA activity is pituitary adenylyl cyclase-activating peptide (PACAP), which has been shown to regulate cerebellar granule precursor proliferation in vitro, a cell population thought to give rise to MB. To test for a PACAP/Hh interaction in the initiation or propagation of these tumors, we introduced a PACAP mutation into ptc1 mutant mice. Deletion of a single copy of PACAP increased MB incidence approximate 2.5-fold, to 66%, thereby demonstrating that PACAP exerts a powerful inhibitory action on the induction, growth or survival of these tumors. Tumors from PACAP/ptc1 mutant mice retained PACAP receptor gene expression, and exhibited superinduction of Hh target genes compared to those from ptc1+/- mice. Moreover, PACAP inhibited proliferation of cell lines derived from tumors in a PKA-dependent manner, and inhibited expression of the Hh target gene gli1. The results provide genetic evidence that PACAP acts as a physiological factor that regulates the pathogenesis of Hh pathway-associated MB tumors.


Assuntos
Meduloblastoma/genética , Polipeptídeo Hipofisário Ativador de Adenilato Ciclase/genética , Receptores de Superfície Celular/genética , Animais , Linhagem Celular Tumoral , Proliferação de Células , Células Cultivadas , Cerebelo/metabolismo , Expressão Gênica , Proteínas Hedgehog/metabolismo , Meduloblastoma/metabolismo , Camundongos , Receptores Patched , Receptor Patched-1
10.
Behav Neurosci ; 123(3): 694-700, 2009 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-19485576

RESUMO

Theories of cerebellar learning propose that alterations in synaptic plasticity resulting in decreases in cerebellar cortical inhibition and increases in sensory activation of interpositus nuclei underlie the development of adaptively timed conditioned motor responses. The authors found that with concurrent pharmacological disconnection of the cerebellar cortex and intense sensory stimulation in the untrained rabbit, eyeblink responses were generated. Neither sensory stimulation nor disconnection alone generated significant eyeblink responses. These results are consistent with dual plasticity models of cerebellar learning and strongly support the general hypothesis that conditioned responses are the result of strengthening of preexisting connections in the nervous system.


Assuntos
Piscadela/fisiologia , Córtex Cerebelar/fisiologia , Inibição Neural/fisiologia , Estimulação Acústica , Animais , Aprendizagem por Associação/fisiologia , Piscadela/efeitos dos fármacos , Cateterismo , Córtex Cerebelar/efeitos dos fármacos , Antagonistas GABAérgicos/farmacologia , Modelos Neurológicos , Inibição Neural/efeitos dos fármacos , Estimulação Luminosa , Picrotoxina/farmacologia , Coelhos
11.
J Neurosci Methods ; 313: 1-5, 2019 02 01.
Artigo em Inglês | MEDLINE | ID: mdl-30389488

RESUMO

BACKGROUND: Animal models of optic nerve injury are often used to study central nervous system (CNS) degeneration and regeneration, and targeting the optic nerve is a powerful approach for axon-protective or remyelination therapy. However, the experimental delivery of drugs or cells to the optic nerve is rarely performed because injections into this structure are difficult in small animals, especially in mice. NEW METHOD: We investigated and developed methods to deliver drugs or cells to the mouse optic nerve through 3 different routes: a) intraorbital, b) through the optic foramen and c) transcranial. RESULTS: The methods targeted different parts of the mouse optic nerve: intraorbital proximal (intraorbital), intracranial middle (optic-foramen) or intracranial distal (transcranial) portion. COMPARISON WITH EXISTING METHODS: Most existing methods target the optic nerve indirectly. For instance, intravitreally delivered cells often cannot cross the inner limiting membrane to reach retinal neurons and optic nerve axons. Systemic delivery, eye drops and intraventricular injections do not always successfully target the optic nerve. Intraorbital and transcranial injections into the optic nerve or chiasm have been performed but these methods have not been well described. We approached the optic nerve with more selective and precise targeting than existing methods. CONCLUSIONS: We successfully targeted the murine optic nerve intraorbitally, through the optic foramen, and transcranially. Of all methods, the injection through the optic foramen is likely the most innovative and fastest. These methods offer additional approaches for therapeutic intervention to be used by those studying white matter damage and axonal regeneration in the CNS.


Assuntos
Modelos Animais de Doenças , Injeções/métodos , Nervo Óptico/efeitos dos fármacos , Órbita , Base do Crânio , Animais , Camundongos , Camundongos Endogâmicos C57BL
12.
Cell Stem Cell ; 25(4): 531-541.e6, 2019 Oct 03.
Artigo em Inglês | MEDLINE | ID: mdl-31585094

RESUMO

Pelizaeus-Merzbacher disease (PMD) is an X-linked leukodystrophy caused by mutations in Proteolipid Protein 1 (PLP1), encoding a major myelin protein, resulting in profound developmental delay and early lethality. Previous work showed involvement of unfolded protein response (UPR) and endoplasmic reticulum (ER) stress pathways, but poor PLP1 genotype-phenotype associations suggest additional pathogenetic mechanisms. Using induced pluripotent stem cell (iPSC) and gene-correction, we show that patient-derived oligodendrocytes can develop to the pre-myelinating stage, but subsequently undergo cell death. Mutant oligodendrocytes demonstrated key hallmarks of ferroptosis including lipid peroxidation, abnormal iron metabolism, and hypersensitivity to free iron. Iron chelation rescued mutant oligodendrocyte apoptosis, survival, and differentiationin vitro, and post-transplantation in vivo. Finally, systemic treatment of Plp1 mutant Jimpy mice with deferiprone, a small molecule iron chelator, reduced oligodendrocyte apoptosis and enabled myelin formation. Thus, oligodendrocyte iron-induced cell death and myelination is rescued by iron chelation in PMD pre-clinical models.


Assuntos
Deferiprona/uso terapêutico , Células-Tronco Pluripotentes Induzidas/fisiologia , Quelantes de Ferro/uso terapêutico , Ferro/metabolismo , Proteína Proteolipídica de Mielina/metabolismo , Oligodendroglia/fisiologia , Doença de Pelizaeus-Merzbacher/terapia , Animais , Diferenciação Celular , Células Cultivadas , Ferroptose , Humanos , Células-Tronco Pluripotentes Induzidas/efeitos dos fármacos , Células-Tronco Pluripotentes Induzidas/transplante , Peroxidação de Lipídeos , Camundongos , Camundongos Mutantes , Mutação/genética , Proteína Proteolipídica de Mielina/genética , Oligodendroglia/efeitos dos fármacos , Oligodendroglia/transplante , Doença de Pelizaeus-Merzbacher/genética , Doença de Pelizaeus-Merzbacher/patologia , Transplante de Células-Tronco , Reparo Gênico Alvo-Dirigido
13.
Int J Cancer ; 122(8): 1803-9, 2008 Apr 15.
Artigo em Inglês | MEDLINE | ID: mdl-18098289

RESUMO

Pituitary adenylyl cyclase activating peptide (PACAP) is expressed in central, sensory, autonomic, and enteric neurons. Although it classically acts as a neurotransmitter/neuromodulator, recent studies indicate that PACAP can also regulate immune function. To this effect, PACAP has been shown to reduce clinical symptoms and inflammation in mouse models of human immune-based diseases such as rheumatoid arthritis, Crohn's Disease, septic shock and multiple sclerosis. Despite these findings, the role of the endogenous peptide in regulating immune function is unknown. To determine if endogenous PACAP plays a protective role in inflammatory bowel disease (IBD) and IBD-associated colorectal cancer in mice, PACAP-deficient (KO) mice were subjected to 3 cycles of dextran sulfate sodium (DSS) in drinking water over 2 months, an established mouse model for colitis. Compared to wild type (WT) controls, PACAP KO mice exhibited more severe clinical symptoms of colitis and had significantly higher colonic inflammation on pathological examination. Moreover, 60% of the PACAP KO mice developed colorectal tumors with an aggressive-appearing pathology. Consistent with published data, DSS-treated WT mice did not develop such tumors. The results demonstrate a new mouse model which rapidly develops inflammation-associated colorectal cancer in the absence of a carcinogen.


Assuntos
Colite/patologia , Colite/prevenção & controle , Neoplasias Colorretais/patologia , Neoplasias Colorretais/prevenção & controle , Polipeptídeo Hipofisário Ativador de Adenilato Ciclase/deficiência , Polipeptídeo Hipofisário Ativador de Adenilato Ciclase/imunologia , Animais , Colite/induzido quimicamente , Colite/imunologia , Neoplasias Colorretais/imunologia , Sulfato de Dextrana , Regulação da Expressão Gênica , Doenças Inflamatórias Intestinais/patologia , Doenças Inflamatórias Intestinais/prevenção & controle , Interleucina-1beta/metabolismo , Interleucina-6/metabolismo , Camundongos , Distribuição Aleatória , Índice de Gravidade de Doença , Fator de Necrose Tumoral alfa/metabolismo
14.
Cell Stem Cell ; 23(4): 464-465, 2018 10 04.
Artigo em Inglês | MEDLINE | ID: mdl-30290175

RESUMO

While myelinating oligodendrocytes are attractive candidates for cell-based regenerative therapies, producing them in adequate quantities and regulation of progenitor differentiation pathways has proven limiting. Recently, Hubler et al. (2018) and Madhavan et al. (2018) generated cerebral organoids with myelinating oligodendrocytes and manipulated sterol pathway small molecules to promote myelin synthesis.


Assuntos
Bainha de Mielina , Oligodendroglia , Diferenciação Celular , Humanos
15.
Neuropsychopharmacology ; 32(3): 710-8, 2007 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-16794568

RESUMO

Mecamylamine, a noncompetitive nicotinic cholinergic antagonist, inhibits nicotine self-administration in animals and may attenuate tobacco smoking in humans trying to quit. Our preliminary data suggested that this agent, at a dose of 2 mg/kg (subcutaneous (s.c.)), also attenuates cue-induced relapse to nicotine-seeking behavior in rats. This study determined whether mecamylamine-induced attenuation can be obtained at doses lower than the high 2 mg/kg dose used in the first study, and whether it is specific to nicotine-associated cues. Male Sprague-Dawley rats were trained to intravenously self-administer nicotine (0.03 mg/kg/infusion) on a fixed-ratio 5 schedule. Each infusion was accompanied by a visual cue (1 s onset of a lever light followed by offset of a house light for 20 s during which time no infusions could be obtained). After the nicotine-maintained responding was extinguished by withholding the delivery of nicotine (saline substitution) and its associated cue, reinstatement tests were conducted. Response-contingent re-presentation of the cue without further availability of nicotine significantly reinstated extinguished responding at the previously nicotine-reinforced lever. Pretreatment with mecamylamine (0.5, 1, and 2 mg/kg, s.c.) dose-dependently attenuated the cue-induced reinstatement of lever responding. Mecamylamine did not change food-taking and -seeking responses, whereas the highest dose (2 mg/kg) decreased nicotine self-administration behavior. The results confirm previous findings that stimuli conditioned to nicotine self-administration effectively elicit reinstatement of nicotine-seeking behavior after extinction and demonstrate that mecamylamine, besides suppressing self-administration of nicotine, effectively attenuates cue-induced nicotine-seeking behavior. These findings suggest that the response-reinstatement procedures used in this study may be useful for studying neurobiological mechanisms of nicotine-seeking behavior and that mecamylamine-like drugs may be potential candidates for pharmacological treatment and prevention of relapse to tobacco smoking in abstinent smokers.


Assuntos
Sinais (Psicologia) , Mecamilamina/farmacologia , Nicotina/administração & dosagem , Agonistas Nicotínicos/administração & dosagem , Antagonistas Nicotínicos/farmacologia , Reforço Psicológico , Análise de Variância , Animais , Comportamento Animal/efeitos dos fármacos , Condicionamento Operante/efeitos dos fármacos , Relação Dose-Resposta a Droga , Extinção Psicológica/efeitos dos fármacos , Masculino , Ratos , Ratos Sprague-Dawley , Autoadministração
16.
Psychopharmacology (Berl) ; 184(3-4): 417-25, 2006 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-16163522

RESUMO

RATIONALE: Smoking-related environmental stimuli have been implicated as an important factor in triggering relapse in abstinent tobacco smokers, and recent evidence indicates that drug-associated stimuli can reinstate nicotine-seeking in rats. However, there is little investigation on the factors that contribute to the latter effect. OBJECTIVE: This study examined whether a nicotine-associated visual stimulus (VS) can reinstate nicotine-seeking after extinction in a response-reinstatement model of relapse, and whether the behavioral effects of the VS are sensitive to pharmacological blockade of nicotinic neurotransmission. It also determined whether active lever reassignment after food training influences nicotine self-administration and the VS-induced reinstatement. METHODS: Male Sprague-Dawley rats were trained to self-administer nicotine (0.03 mg/kg/infusion, IV) and associate a VS with each nicotine infusion in 30 daily 1-h sessions. Half of the animals received nicotine infusions for responding at the same lever that previously delivered food; for the other half, infusions resulted from pressing the previously inactive lever during food training. Then, the nicotine-maintained response was extinguished by saline substitution and withholding the VS. One day after rats reached extinction criterion, the reinstatement tests were conducted where the VS was response-contingent represented without further delivery of nicotine. In pharmacological tests, a nicotinic antagonist, mecamylamine, was subcutaneously administered 30 min before reinstatement sessions. RESULTS: Presentation of the nicotine-associated VS significantly reinstated responding at the previously drug-reinforced lever and pretreatment with mecamylamine effectively attenuated the response-reinstating effect of the VS. Additionally, animals showed similar profiles of nicotine-taking and nicotine-seeking behavior regardless of reassignment of the active lever after food training. CONCLUSIONS: Nicotine self-administration and the VS-induced reinstatement of nicotine-seeking do not result from a lever bias due to prior experience for food reinforcement. Significantly, these results suggest that environmental stimuli associated with nicotine self-administration can effectively elicit nicotine-seeking behavior in abstinent subjects, that this effect is blocked by nicotine antagonism, and that the present procedures may be useful for studying neurobiological mechanisms of nicotine-seeking behavior and relapse.


Assuntos
Aprendizagem por Associação , Sinais (Psicologia) , Extinção Psicológica , Motivação , Nicotina/administração & dosagem , Meio Social , Tabagismo/psicologia , Animais , Modelos Animais de Doenças , Masculino , Mecamilamina/farmacologia , Antagonistas Nicotínicos/farmacologia , Ratos , Ratos Sprague-Dawley , Recidiva , Esquema de Reforço , Autoadministração
17.
J Leukoc Biol ; 95(2): 357-67, 2014 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-24068730

RESUMO

Although the immune system may provide early protection against cancer, tumors may exploit the healing arm of the immune system to enhance their growth and metastasis. For example, myeloid derived suppressor cells (MDSCs) are thought to promote tumor growth by several mechanisms, including the suppression of T cell activity. It has been suggested that STAT3 activation in myeloid cells modulates multiple aspects of MDSC physiology, including their expansion and activity. Whereas most animal studies investigating tumor immunology have used tumor implants, we used transgenic mice (Smo*) that spontaneously develop medulloblastoma brain tumors to investigate the temporal accumulation of MDSCs within tumors and how myeloid STAT3 disruption affects MDSC and other immune cell types. We found distinct populations of MDSC in medulloblastoma tumors, with a high prevalence of CD11b(+)Ly6G(+)Ly6C(low/-) cells, described previously by others as G-MDSCs. These were found early in tumor development, in premalignant lesions located on the surface of the cerebellum of 28-day-old mice. In fully developed tumors, pSTAT3 was found in the majority of these cells. Conditional STAT3 gene disruption in myeloid cells resulted in an enhanced proinflammatory phenotype of macrophages in Smo* mice. Moreover, a significant reduction in the abundance of G-MDSCs and Tregs was observed within tumors along with an increased presence of CD4(+) and CD8(+) cells. Despite these alterations in immune cells induced by myeloid STAT3 disruption, we found no effect on tumor incidence in Smo* mice with this deletion.


Assuntos
Deleção de Genes , Marcação de Genes , Tolerância Imunológica , Meduloblastoma/imunologia , Meduloblastoma/patologia , Células Mieloides/metabolismo , Fator de Transcrição STAT3/metabolismo , Animais , Modelos Animais de Doenças , Feminino , Inflamação/imunologia , Inflamação/patologia , Masculino , Meduloblastoma/irrigação sanguínea , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Células Mieloides/patologia , Neovascularização Patológica/patologia , Receptores Acoplados a Proteínas G/metabolismo , Transdução de Sinais/imunologia , Receptor Smoothened , Linfócitos T Reguladores/imunologia
18.
Neuron ; 81(3): 574-87, 2014 Feb 05.
Artigo em Inglês | MEDLINE | ID: mdl-24507192

RESUMO

Abnormal GABAergic interneuron density, and imbalance of excitatory versus inhibitory tone, is thought to result in epilepsy, neurodevelopmental disorders, and psychiatric disease. Recent studies indicate that interneuron cortical density is determined primarily by the size of the precursor pool in the embryonic telencephalon. However, factors essential for regulating interneuron allocation from telencephalic multipotent precursors are poorly understood. Here we report that Olig1 represses production of GABAergic interneurons throughout the mouse brain. Olig1 deletion in mutant mice results in ectopic expression and upregulation of Dlx1/2 genes in the ventral medial ganglionic eminences and adjacent regions of the septum, resulting in an ∼30% increase in adult cortical interneuron numbers. We show that Olig1 directly represses the Dlx1/2 I12b intergenic enhancer and that Dlx1/2 functions genetically downstream of Olig1. These findings establish Olig1 as an essential repressor of Dlx1/2 and interneuron production in developing mammalian brain.


Assuntos
Fatores de Transcrição Hélice-Alça-Hélice Básicos/metabolismo , Encéfalo/citologia , Regulação da Expressão Gênica no Desenvolvimento/fisiologia , Proteínas de Homeodomínio/metabolismo , Interneurônios/fisiologia , Fatores de Transcrição/metabolismo , Potenciais de Ação/genética , Potenciais de Ação/fisiologia , Fatores Etários , Animais , Fatores de Transcrição Hélice-Alça-Hélice Básicos/genética , Encéfalo/embriologia , Encéfalo/crescimento & desenvolvimento , Contagem de Células , Proteínas de Ligação a DNA/genética , Proteínas de Ligação a DNA/metabolismo , Embrião de Mamíferos , Regulação da Expressão Gênica no Desenvolvimento/genética , Glutamato Descarboxilase/metabolismo , Proteínas de Homeodomínio/genética , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Mutação/genética , Proteínas do Tecido Nervoso/metabolismo , Neuropeptídeos/metabolismo , Técnicas de Cultura de Órgãos , Técnicas de Patch-Clamp , Sinapses/fisiologia , Fatores de Transcrição/genética
19.
PLoS One ; 7(5): e36922, 2012.
Artigo em Inglês | MEDLINE | ID: mdl-22615845

RESUMO

Vasoactive intestinal peptide (VIP) is a pleiotropic neuropeptide with immunomodulatory properties. The administration of this peptide has been shown to have beneficial effects in murine models of inflammatory diseases including septic shock, rheumatoid arthritis, multiple sclerosis (MS) and Crohn's disease. However, the role of the endogenous peptide in inflammatory disease remains obscure because VIP-deficient mice were recently found to exhibit profound resistance in a model of MS. In the present study, we analyzed the response of female VIP deficient (KO) mice to intraperitoneal lipopolysaccharide (LPS) administration. We observed significant resistance to LPS in VIP KO mice, as evidenced by lower mortality and reduced tissue damage. The increased survival was associated with decreased levels of proinflammatory cytokines (TNFα, IL-6 and IL-12) in sera and peritoneal suspensions of these mice. Moreover, the expression of TNFα and IL-6 mRNA was reduced in peritoneal cells, spleens and lungs from LPS-treated VIP KO vs. WT mice, suggesting that the resistance might be mediated by an intrinsic defect in the responsiveness of immune cells to endotoxin. In agreement with this hypothesis, peritoneal cells isolated from VIP KO naive mice produced lower levels of proinflammatory cytokines in response to LPS in vitro. Finally, decreased NF-κB pathway activity in peritoneal cells was observed both in vivo and in vitro, as determined by assay of phosphorylated I-κB. The results demonstrate that female VIP KO mice exhibit resistance to LPS-induced shock, explainable in part by the presence of an intrinsic defect in the responsiveness of inflammatory cells to endotoxin.


Assuntos
Endotoxemia/imunologia , Mediadores da Inflamação/imunologia , Inflamação/imunologia , Lipopolissacarídeos/farmacologia , Neuropeptídeos/imunologia , Peptídeo Intestinal Vasoativo/deficiência , Peptídeo Intestinal Vasoativo/imunologia , Animais , Endotoxemia/induzido quimicamente , Endotoxinas/administração & dosagem , Endotoxinas/imunologia , Feminino , Inflamação/sangue , Inflamação/genética , Inflamação/metabolismo , Interleucina-12/sangue , Interleucina-12/imunologia , Interleucina-6/sangue , Interleucina-6/imunologia , Lipopolissacarídeos/imunologia , Pulmão/imunologia , Pulmão/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , NF-kappa B/imunologia , NF-kappa B/metabolismo , Neuropeptídeos/genética , RNA Mensageiro/genética , RNA Mensageiro/imunologia , Baço/imunologia , Baço/metabolismo , Fator de Necrose Tumoral alfa/sangue , Fator de Necrose Tumoral alfa/imunologia , Peptídeo Intestinal Vasoativo/genética
20.
ASN Neuro ; 3(4)2011 Nov 17.
Artigo em Inglês | MEDLINE | ID: mdl-22007738

RESUMO

Studies in humans and animal models link maternal infection and imbalanced levels of inflammatory mediators in the foetal brain to the aetiology of neuropsychiatric disorders. In a number of animal models, it was shown that exposure to viral or bacterial agents during a period that corresponds to the second trimester in human gestation triggers brain and behavioural abnormalities in the offspring. However, little is known about the early cellular and molecular events elicited by inflammation in the foetal brain shortly after maternal infection has occurred. In this study, maternal infection was mimicked by two consecutive intraperitoneal injections of 200 µg of LPS (lipopolysaccharide)/kg to timed-pregnant rats at GD15 (gestational day 15) and GD16. Increased thickness of the CP (cortical plate) and hippocampus together with abnormal distribution of immature neuronal markers and decreased expression of markers for neural progenitors were observed in the LPS-exposed foetal forebrains at GD18. Such effects were accompanied by decreased levels of reelin and the radial glial marker GLAST (glial glutamate transporter), and elevated levels of pro-inflammatory cytokines in maternal serum and foetal forebrains. Foetal inflammation elicited by maternal injections of LPS has discrete detrimental effects on brain development. The early biochemical and morphological changes described in this work begin to explain the sequelae of early events that underlie the neurobehavioural deficits reported in humans and animals exposed to prenatal insults.


Assuntos
Encefalite/induzido quimicamente , Encefalite/patologia , Lipopolissacarídeos/toxicidade , Efeitos Tardios da Exposição Pré-Natal/induzido quimicamente , Efeitos Tardios da Exposição Pré-Natal/fisiopatologia , Prosencéfalo , Fatores Etários , Animais , Animais Recém-Nascidos , Movimento Celular/efeitos dos fármacos , Citocinas/metabolismo , Proteínas do Citoesqueleto/metabolismo , Embrião de Mamíferos , Feminino , Regulação da Expressão Gênica no Desenvolvimento/efeitos dos fármacos , Masculino , Proteínas do Tecido Nervoso/metabolismo , Gravidez , Prosencéfalo/efeitos dos fármacos , Prosencéfalo/embriologia , Prosencéfalo/patologia , Ratos , Proteína Reelina , Fatores de Tempo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA