Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 123
Filtrar
Mais filtros

Base de dados
Tipo de documento
Intervalo de ano de publicação
1.
Nature ; 566(7745): 543-547, 2019 02.
Artigo em Inglês | MEDLINE | ID: mdl-30747918

RESUMO

Oligodendrocyte pathology is increasingly implicated in neurodegenerative diseases as oligodendrocytes both myelinate and provide metabolic support to axons. In multiple sclerosis (MS), demyelination in the central nervous system thus leads to neurodegeneration, but the severity of MS between patients is very variable. Disability does not correlate well with the extent of demyelination1, which suggests that other factors contribute to this variability. One such factor may be oligodendrocyte heterogeneity. Not all oligodendrocytes are the same-those from the mouse spinal cord inherently produce longer myelin sheaths than those from the cortex2, and single-cell analysis of the mouse central nervous system identified further differences3,4. However, the extent of human oligodendrocyte heterogeneity and its possible contribution to MS pathology remain unknown. Here we performed single-nucleus RNA sequencing from white matter areas of post-mortem human brain from patients with MS and from unaffected controls. We identified subclusters of oligodendroglia in control human white matter, some with similarities to mouse, and defined new markers for these cell states. Notably, some subclusters were underrepresented in MS tissue, whereas others were more prevalent. These differences in mature oligodendrocyte subclusters may indicate different functional states of oligodendrocytes in MS lesions. We found similar changes in normal-appearing white matter, showing that MS is a more diffuse disease than its focal demyelination suggests. Our findings of an altered oligodendroglial heterogeneity in MS may be important for understanding disease progression and developing therapeutic approaches.


Assuntos
Encéfalo/metabolismo , Encéfalo/patologia , Esclerose Múltipla/patologia , Oligodendroglia/patologia , Adulto , Idoso , Idoso de 80 Anos ou mais , Animais , Autopsia , Biomarcadores , Estudos de Casos e Controles , Progressão da Doença , Feminino , Perfilação da Expressão Gênica , Regulação da Expressão Gênica/genética , Humanos , Masculino , Camundongos , Pessoa de Meia-Idade , Esclerose Múltipla/genética , Bainha de Mielina/genética , Bainha de Mielina/metabolismo , Bainha de Mielina/patologia , Oligodendroglia/metabolismo , Remielinização/genética , Análise de Sequência de RNA , Transcrição Gênica , Substância Branca/citologia , Substância Branca/metabolismo , Substância Branca/patologia
3.
Brain Behav Immun ; 109: 1-22, 2023 03.
Artigo em Inglês | MEDLINE | ID: mdl-36584795

RESUMO

Multiple sclerosis (MS) is a chronic autoimmune disease of the central nervous system (CNS) characterized by focal inflammatory lesions and prominent demyelination. Even though the currently available therapies are effective in treating the initial stages of disease, they are unable to halt or reverse disease progression into the chronic progressive stage. Thus far, no repair-inducing treatments are available for progressive MS patients. Hence, there is an urgent need for the development of new therapeutic strategies either targeting the destructive immunological demyelination or boosting endogenous repair mechanisms. Using in vitro, ex vivo, and in vivo models, we demonstrate that selective inhibition of phosphodiesterase 4 (PDE4), a family of enzymes that hydrolyzes and inactivates cyclic adenosine monophosphate (cAMP), reduces inflammation and promotes myelin repair. More specifically, we segregated the myelination-promoting and anti-inflammatory effects into a PDE4D- and PDE4B-dependent process respectively. We show that inhibition of PDE4D boosts oligodendrocyte progenitor cells (OPC) differentiation and enhances (re)myelination of both murine OPCs and human iPSC-derived OPCs. In addition, PDE4D inhibition promotes in vivo remyelination in the cuprizone model, which is accompanied by improved spatial memory and reduced visual evoked potential latency times. We further identified that PDE4B-specific inhibition exerts anti-inflammatory effects since it lowers in vitro monocytic nitric oxide (NO) production and improves in vivo neurological scores during the early phase of experimental autoimmune encephalomyelitis (EAE). In contrast to the pan PDE4 inhibitor roflumilast, the therapeutic dose of both the PDE4B-specific inhibitor A33 and the PDE4D-specific inhibitor Gebr32a did not trigger emesis-like side effects in rodents. Finally, we report distinct PDE4D isoform expression patterns in human area postrema neurons and human oligodendroglia lineage cells. Using the CRISPR-Cas9 system, we confirmed that pde4d1/2 and pde4d6 are the key targets to induce OPC differentiation. Collectively, these data demonstrate that gene specific PDE4 inhibitors have potential as novel therapeutic agents for targeting the distinct disease processes of MS.


Assuntos
Encefalomielite Autoimune Experimental , Esclerose Múltipla , Inibidores da Fosfodiesterase 4 , Humanos , Camundongos , Animais , Bainha de Mielina/metabolismo , Esclerose Múltipla/metabolismo , Nucleotídeo Cíclico Fosfodiesterase do Tipo 4/metabolismo , Nucleotídeo Cíclico Fosfodiesterase do Tipo 4/farmacologia , Nucleotídeo Cíclico Fosfodiesterase do Tipo 4/uso terapêutico , Potenciais Evocados Visuais , Oligodendroglia/metabolismo , Encefalomielite Autoimune Experimental/metabolismo , Diferenciação Celular , Inibidores da Fosfodiesterase 4/farmacologia , Inibidores da Fosfodiesterase 4/uso terapêutico , Anti-Inflamatórios/farmacologia , Camundongos Endogâmicos C57BL
4.
Nature ; 547(7663): 350-354, 2017 07 20.
Artigo em Inglês | MEDLINE | ID: mdl-28700576

RESUMO

After liver injury, regeneration occurs through self-replication of hepatocytes. In severe liver injury, hepatocyte proliferation is impaired-a feature of human chronic liver disease. It is unclear whether other liver cell types can regenerate hepatocytes. Here we use two independent systems to impair hepatocyte proliferation during liver injury to evaluate the contribution of non-hepatocytes to parenchymal regeneration. First, loss of ß1-integrin in hepatocytes with liver injury triggered a ductular reaction of cholangiocyte origin, with approximately 25% of hepatocytes being derived from a non-hepatocyte origin. Second, cholangiocytes were lineage traced with concurrent inhibition of hepatocyte proliferation by ß1-integrin knockdown or p21 overexpression, resulting in the significant emergence of cholangiocyte-derived hepatocytes. We describe a model of combined liver injury and inhibition of hepatocyte proliferation that causes physiologically significant levels of regeneration of functional hepatocytes from biliary cells.


Assuntos
Ductos Biliares Intra-Hepáticos/citologia , Hepatócitos/patologia , Regeneração Hepática , Fígado/citologia , Fígado/patologia , Células-Tronco/citologia , Animais , Linhagem da Célula , Proliferação de Células , Feminino , Integrina beta1/genética , Fígado/lesões , Hepatopatias/patologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL
5.
J Neurosci ; 41(38): 7954-7964, 2021 09 22.
Artigo em Inglês | MEDLINE | ID: mdl-34341156

RESUMO

Oligodendrocytes generate myelin sheaths vital for the formation, health, and function of the CNS. Myelin sheath length is a key property that determines axonal conduction velocity and is known to be variable across the CNS. Myelin sheath length can be modified by neuronal activity, suggesting that dynamic regulation of sheath length might contribute to the functional plasticity of neural circuits. Although the mechanisms that establish and refine myelin sheath length are important determinants of brain function, our understanding of these remains limited. In recent years, the membranes of myelin sheaths have been increasingly recognized to contain ion channels and transporters that are associated with specific important oligodendrocyte functions, including metabolic support of axons and the regulation of ion homeostasis, but none have been shown to influence sheath architecture. In this study, we determined that hyperpolarization-activated, cyclic nucleotide-gated (HCN) ion channels, typically associated with neuronal and cardiac excitability, regulate myelin sheath length. Using both in vivo and in vitro approaches, we show that oligodendrocytes abundantly express functional, predominantly HCN2 subunit-containing ion channels. These HCN ion channels retain key pharmacological and biophysical features and regulate the resting membrane potential of myelinating oligodendrocytes. Further, reduction of their function via pharmacological blockade or generation of transgenic mice with two independent oligodendrocyte-specific HCN2 knock-out strategies reduced myelin sheath length. We conclude that HCN2 ion channels are key determinants of myelin sheath length in the CNS.SIGNIFICANCE STATEMENT Myelin sheath length is a critical determinant of axonal conduction velocity, but the signaling mechanisms responsible for determining sheath length are poorly understood. Here we find that oligodendrocytes express functional hyperpolarization-activated, cyclic nucleotide-gated 2 (HCN2) ion channels that regulate the length of myelin sheaths formed by oligodendrocytes in myelinating cultures and in the mouse brain and spinal cord. These results suggest that the regulation of HCN2 channel activity is well placed to refine sheath length and conduction along myelinated axons, providing a potential mechanism for alterations in conduction velocity and circuit function in response to axonal signals such as those generated by increased activity.


Assuntos
Canais Disparados por Nucleotídeos Cíclicos Ativados por Hiperpolarização/metabolismo , Bainha de Mielina/metabolismo , Oligodendroglia/metabolismo , Córtex Pré-Frontal/metabolismo , Animais , Axônios/fisiologia , Canais Disparados por Nucleotídeos Cíclicos Ativados por Hiperpolarização/genética , Camundongos , Camundongos Transgênicos , Condução Nervosa/fisiologia , Neurônios/metabolismo
6.
Development ; 146(16)2019 08 29.
Artigo em Inglês | MEDLINE | ID: mdl-31371375

RESUMO

Development of the central nervous system requires coordination of the proliferation and differentiation of neural stem cells. Here, we show that laminin alpha 2 (lm-α2) is a component of the midbrain dopaminergic neuron (mDA) progenitor niche in the ventral midbrain (VM) and identify a concentration-dependent role for laminin α2ß1γ1 (lm211) in regulating mDA progenitor proliferation and survival via a distinct set of receptors. At high concentrations, lm211-rich environments maintain mDA progenitors in a proliferative state via integrins α6ß1 and α7ß1, whereas low concentrations of lm211 support mDA lineage survival via dystroglycan receptors. We confirmed our findings in vivo, demonstrating that the VM was smaller in the absence of lm-α2, with increased apoptosis; furthermore, the progenitor pool was depleted through premature differentiation, resulting in fewer mDA neurons. Examination of mDA neuron subtype composition showed a reduction in later-born mDA neurons of the ventral tegmental area, which control a range of cognitive behaviours. Our results identify a novel role for laminin in neural development and provide a possible mechanism for autism-like behaviours and the brainstem hypoplasia seen in some individuals with mutations of LAMA2.


Assuntos
Neurônios Dopaminérgicos/fisiologia , Laminina/fisiologia , Mesencéfalo/embriologia , Neurogênese , Animais , Linhagem Celular , Proliferação de Células , Sobrevivência Celular , Humanos , Integrinas/metabolismo , Laminina/genética , Mesencéfalo/citologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Neurogênese/genética
7.
Nat Rev Neurosci ; 18(12): 753-769, 2017 11 16.
Artigo em Inglês | MEDLINE | ID: mdl-29142295

RESUMO

Although the core concept of remyelination - based on the activation, migration, proliferation and differentiation of CNS progenitors - has not changed over the past 20 years, our understanding of the detailed mechanisms that underlie this process has developed considerably. We can now decorate the central events of remyelination with a host of pathways, molecules, mediators and cells, revealing a complex and precisely orchestrated process. These advances have led to recent drug-based and cell-based clinical trials for myelin diseases and have opened up hitherto unrecognized opportunities for drug-based approaches to therapeutically enhance remyelination.


Assuntos
Doenças Desmielinizantes/fisiopatologia , Bainha de Mielina/metabolismo , Regeneração Nervosa/fisiologia , Oligodendroglia/metabolismo , Animais , Diferenciação Celular/fisiologia , Sistema Nervoso Central/metabolismo , Humanos
8.
Glia ; 68(9): 1840-1858, 2020 09.
Artigo em Inglês | MEDLINE | ID: mdl-32125730

RESUMO

During axonal ensheathment, noncompact myelin channels formed at lateral edges of the myelinating process become arranged into tight paranodal spirals that resemble loops when cut in cross section. These adhere to the axon, concentrating voltage-dependent sodium channels at nodes of Ranvier and patterning the surrounding axon into distinct molecular domains. The signals responsible for forming and maintaining the complex structure of paranodal myelin are poorly understood. Here, we test the hypothesis that the planar cell polarity determinant Vangl2 organizes paranodal myelin. We show that Vangl2 is concentrated at paranodes and that, following conditional knockout of Vangl2 in oligodendrocytes, the paranodal spiral loosens, accompanied by disruption to the microtubule cytoskeleton and mislocalization of autotypic adhesion molecules between loops within the spiral. Adhesion of the spiral to the axon is unaffected. This results in disruptions to axonal patterning at nodes of Ranvier, paranodal axon diameter and conduction velocity. When taken together with our previous work showing that loss of the apico-basal polarity protein Scribble has the opposite phenotype-loss of axonal adhesion but no effect on loop-loop autotypic adhesion-our results identify a novel mechanism by which polarity proteins control the shape of nodes of Ranvier and regulate conduction in the CNS.


Assuntos
Bainha de Mielina , Nós Neurofibrosos , Axônios , Polaridade Celular , Oligodendroglia
9.
Small ; 16(37): e2003656, 2020 09.
Artigo em Inglês | MEDLINE | ID: mdl-32790058

RESUMO

A key hallmark of many diseases, especially those in the central nervous system (CNS), is the change in tissue stiffness due to inflammation and scarring. However, how such changes in microenvironment affect the regenerative process remains poorly understood. Here, a biomimicking fiber platform that provides independent variation of fiber structural and intrinsic stiffness is reported. To demonstrate the functionality of these constructs as a mechanotransduction study platform, these substrates are utilized as artificial axons and the effects of axon structural versus intrinsic stiffness on CNS myelination are independently analyzed. While studies have shown that substrate stiffness affects oligodendrocyte differentiation, the effects of mechanical stiffness on the final functional state of oligodendrocyte (i.e., myelination) has not been shown prior to this. Here, it is demonstrated that a stiff mechanical microenvironment impedes oligodendrocyte myelination, independently and distinctively from oligodendrocyte differentiation. Yes-associated protein is identified to be involved in influencing oligodendrocyte myelination through mechanotransduction. The opposing effects on oligodendrocyte differentiation and myelination provide important implications for current work screening for promyelinating drugs, since these efforts have focused mainly on promoting oligodendrocyte differentiation. Thus, the platform may have considerable utility as part of a drug discovery program in identifying molecules that promote both differentiation and myelination.


Assuntos
Mecanotransdução Celular , Bainha de Mielina , Axônios , Diferenciação Celular , Oligodendroglia
10.
Mol Psychiatry ; 24(11): 1641-1654, 2019 11.
Artigo em Inglês | MEDLINE | ID: mdl-31481758

RESUMO

Although the underlying neurobiology of major mental illness (MMI) remains unknown, emerging evidence implicates a role for oligodendrocyte-myelin abnormalities. Here, we took advantage of a large family carrying a balanced t(1;11) translocation, which substantially increases risk of MMI, to undertake both diffusion tensor imaging and cellular studies to evaluate the consequences of the t(1;11) translocation on white matter structural integrity and oligodendrocyte-myelin biology. This translocation disrupts among others the DISC1 gene which plays a crucial role in brain development. We show that translocation-carrying patients display significant disruption of  white matter integrity compared with familial controls. At a cellular level, we observe dysregulation of key pathways controlling oligodendrocyte development and morphogenesis in induced pluripotent stem cell (iPSC) derived case oligodendrocytes. This is associated with reduced proliferation and a stunted morphology in vitro. Further, myelin internodes in a humanized mouse model that recapitulates the human translocation as well as after transplantation of t(1;11) oligodendrocyte progenitors were significantly reduced when  compared with controls. Thus we provide evidence that the t(1;11) translocation has biological effects at both the systems and cellular level that together suggest oligodendrocyte-myelin dysfunction.


Assuntos
Bainha de Mielina/metabolismo , Oligodendroglia/metabolismo , Translocação Genética/genética , Adulto , Animais , Cromossomos Humanos Par 1/genética , Cromossomos Humanos Par 11/genética , Imagem de Tensor de Difusão/métodos , Feminino , Humanos , Células-Tronco Pluripotentes Induzidas/metabolismo , Masculino , Transtornos Mentais/genética , Camundongos , Pessoa de Meia-Idade , Proteínas do Tecido Nervoso/genética , Proteínas do Tecido Nervoso/metabolismo , Substância Branca/metabolismo , Substância Branca/fisiologia
11.
Mol Ther ; 27(2): 411-423, 2019 02 06.
Artigo em Inglês | MEDLINE | ID: mdl-30611662

RESUMO

The loss of oligodendrocytes (OLs) and subsequently myelin sheaths following injuries or pathologies in the CNS leads to debilitating functional deficits. Unfortunately, effective methods of remyelination remain limited. Here, we present a scaffolding system that enables sustained non-viral delivery of microRNAs (miRs) to direct OL differentiation, maturation, and myelination. We show that miR-219/miR-338 promoted primary rat OL differentiation and myelination in vitro. Using spinal cord injury as a proof-of-concept, we further demonstrate that miR-219/miR-338 could also be delivered non-virally in vivo using an aligned fiber-hydrogel scaffold to enhance remyelination after a hemi-incision injury at C5 level of Sprague-Dawley rats. Specifically, miR-219/miR-338 mimics were incorporated as complexes with the carrier, TransIT-TKO (TKO), together with neurotrophin-3 (NT-3) within hybrid scaffolds that comprised poly(caprolactone-co-ethyl ethylene phosphate) (PCLEEP)-aligned fibers and collagen hydrogel. After 1, 2, and 4 weeks post-treatment, animals that received NT-3 and miR-219/miR-338 treatment preserved a higher number of Olig2+ oligodendroglial lineage cells as compared with those treated with NT-3 and negative scrambled miRs (Neg miRs; p < 0.001). Additionally, miR-219/miR-338 increased the rate and extent of differentiation of OLs. At the host-implant interface, more compact myelin sheaths were observed when animals received miR-219/miR-338. Similarly within the scaffolds, miR-219/miR-338 samples contained significantly more myelin basic protein (MBP) signals (p < 0.01) and higher myelination index (p < 0.05) than Neg miR samples. These findings highlight the potential of this platform to promote remyelination within the CNS.


Assuntos
Sistema Nervoso Central/metabolismo , Portadores de Fármacos/química , MicroRNAs/metabolismo , Remielinização/fisiologia , Animais , Feminino , Hidrogéis/química , Imuno-Histoquímica , MicroRNAs/química , MicroRNAs/genética , Microscopia Eletrônica de Varredura , Fatores de Crescimento Neural/metabolismo , Ratos , Ratos Sprague-Dawley , Remielinização/genética
12.
Glia ; 67(2): 376-392, 2019 02.
Artigo em Inglês | MEDLINE | ID: mdl-30450647

RESUMO

The tightly controlled processes of myelination and remyelination require the participation of the cytoskeleton. The reorganization of the cytoskeleton is controlled by small GTPases of the RhoA family. Here, we report that Vav3, a Rho GTPase regulating guanine nucleotide exchange factor (GEF) is involved in oligodendrocyte maturation, myelination and remyelination. When Vav3 was eliminated by genetic recombination, oligodendrocyte precursor cell (OPC) differentiation toward mature oligodendrocytes was accelerated. In contrast, Vav3-deficient oligodendrocytes displayed a reduced capacity to myelinate synthetic microfibers in vitro. Furthermore, remyelination was impaired in Vav3 knockout cerebellar slice cultures that were demyelinated by the addition of lysolecithin. In agreement with these observations, remyelination was compromised when the cuprizone model of myelin lesion was performed in Vav3-deficient mice. When Vav3-deficient oligodendrocytes were examined with Förster resonance energy transfer (FRET)-based biosensors, an altered activation profile of RhoA GTPases was revealed on the cellular level, which could be responsible for an impaired remyelination. Taken together, this study highlights Vav3 as a novel regulator of oligodendrocyte maturation and remyelination, suggesting that manipulation of the Vav3-dependent signaling pathway could help to improve myelin repair.


Assuntos
Diferenciação Celular/genética , Leucoencefalopatias/patologia , Células Precursoras de Oligodendrócitos/fisiologia , Proteínas Proto-Oncogênicas c-vav/metabolismo , Remielinização/genética , Animais , Caspase 3/metabolismo , Diferenciação Celular/efeitos dos fármacos , Células Cultivadas , Quelantes/toxicidade , Cuprizona/toxicidade , Modelos Animais de Doenças , GTP Fosfo-Hidrolases/metabolismo , Antígeno Ki-67/metabolismo , Leucoencefalopatias/induzido quimicamente , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Proteína Básica da Mielina/metabolismo , Fibras Nervosas Mielinizadas/patologia , Células Precursoras de Oligodendrócitos/patologia , Técnicas de Cultura de Órgãos , Proteínas Proto-Oncogênicas c-vav/genética , Remielinização/efeitos dos fármacos , Proteína rhoA de Ligação ao GTP/metabolismo
13.
Proc Natl Acad Sci U S A ; 113(48): E7828-E7836, 2016 11 29.
Artigo em Inglês | MEDLINE | ID: mdl-27849577

RESUMO

Several factors are known to enhance adult hippocampal neurogenesis but a factor capable of inducing a long-lasting neurogenic enhancement that attenuates age-related neurogenic decay has not been described. Here, we studied hippocampal neurogenesis following conditional VEGF induction in the adult brain and showed that a short episode of VEGF exposure withdrawn shortly after the generation of durable new vessels (but not under conditions where newly made vessels failed to persist) is sufficient for neurogenesis to proceed at a markedly elevated level for many months later. Continual neurogenic increase over several months was not accompanied by accelerated exhaustion of the neuronal stem cell (NSC) reserve, thereby allowing neurogenesis to proceed at a markedly elevated rate also in old mice. Neurogenic enhancement by VEGF preconditioning was, in part, attributed to rescue of age-related NSC quiescence. Remarkably, VEGF caused extensive NSC remodelling manifested in transition of the enigmatic NSC terminal arbor onto long cytoplasmic processes engaging with and spreading over even remote blood vessels, a configuration reminiscent of early postnatal "juvenile" NSCs. Together, these findings suggest that VEGF preconditioning might be harnessed for long-term neurogenic enhancement despite continued exposure to an "aged" systemic milieu.


Assuntos
Envelhecimento , Giro Denteado/citologia , Células-Tronco Neurais/fisiologia , Neurogênese , Fator A de Crescimento do Endotélio Vascular/fisiologia , Animais , Forma Celular , Circulação Cerebrovascular , Giro Denteado/irrigação sanguínea , Giro Denteado/fisiologia , Feminino , Expressão Gênica , Masculino , Camundongos Transgênicos , Nicho de Células-Tronco
14.
PLoS Biol ; 13(3): e1002107, 2015 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-25807062

RESUMO

The development and regeneration of myelin by oligodendrocytes, the myelin-forming cells of the central nervous system (CNS), requires profound changes in cell shape that lead to myelin sheath initiation and formation. Here, we demonstrate a requirement for the basal polarity complex protein Scribble in CNS myelination and remyelination. Scribble is expressed throughout oligodendroglial development and is up-regulated in mature oligodendrocytes where it is localised to both developing and mature CNS myelin sheaths. Knockdown of Scribble expression in cultured oligodendroglia results in disrupted morphology and myelination initiation. When Scribble expression is conditionally eliminated in the myelinating glia of transgenic mice, myelin initiation in CNS is disrupted, both during development and following focal demyelination, and longitudinal extension of the myelin sheath is disrupted. At later stages of myelination, Scribble acts to negatively regulate myelin thickness whilst suppressing the extracellular signal-related kinase (ERK)/mitogen-activated protein kinase (MAP) kinase pathway, and localises to non-compact myelin flanking the node of Ranvier where it is required for paranodal axo-glial adhesion. These findings demonstrate an essential role for the evolutionarily-conserved regulators of intracellular polarity in myelination and remyelination.


Assuntos
Sistema Nervoso Central/metabolismo , Peptídeos e Proteínas de Sinalização Intracelular/genética , Oligodendroglia/metabolismo , Animais , Polaridade Celular , Sistema Nervoso Central/ultraestrutura , MAP Quinases Reguladas por Sinal Extracelular/genética , MAP Quinases Reguladas por Sinal Extracelular/metabolismo , Regulação da Expressão Gênica , Peptídeos e Proteínas de Sinalização Intracelular/antagonistas & inibidores , Peptídeos e Proteínas de Sinalização Intracelular/metabolismo , Camundongos , Camundongos Transgênicos , Oligodendroglia/ultraestrutura , RNA Interferente Pequeno/genética , RNA Interferente Pequeno/metabolismo , Transdução de Sinais
15.
Lancet ; 385 Suppl 1: S69, 2015 Feb 26.
Artigo em Inglês | MEDLINE | ID: mdl-26312891

RESUMO

BACKGROUND: Retinitis pigmentosa, which affects one in 3000 people, causes blindness and has no treatment. Mutations in the retinitis pigmentosa GTPase regulator (RPGR) gene cause 20% of all cases. Recent work suggests that RPGR, localised to the photoreceptor connecting cilium, regulates rhodopsin transport to the outer segment through its effect on the turnover of actin. We set out to establish a novel model for RPGR disease to test the hypothesis that RPGR mutations lead to retinal degeneration due to a dysregulation of the actin cytoskeleton. METHODS: Patients with RPGR mutations and their unaffected relatives were recruited and skin biopsy samples taken. Fibroblast lines were established and reprogrammed to generate induced pluripotent stem cell (iPSC) lines. A three-dimensional organogenesis protocol was optimised whereby embryoid bodies were formed and patterned towards an eye field fate in a 100-day retinal differentiation protocol, allowing three-dimensional optic cups to form. RPGR-mutated cultures were compared with their healthy controls. FINDINGS: Mutant and wild-type iPSC lines were generated and characterised. Differentiation of all lines resulted in the generation of optic cups in a self-organising manner after 100 days in culture. These cultures contained mature photoreceptors, as evidenced by morphology and both RNA and protein expression. Photoreceptor cultures from RGPR-mutated iPSCs had increased actin polymerisation compared with controls (mean confocal pixel intensity count 59·02 [SD 16·24] vs 23·70 [8·20], p=0·0081). This finding was confirmed by assessment of F-actin with western blot. Pathways regulating actin turnover were explored; western blot analysis showed a reduction in both Src and ERK phosphorylation in RGPR-mutated photoreceptor cultures. An unbiased protein array confirmed this reduction in ERK and Src activation. Several other pathways were also shown to be dysregulated in the RGPR-mutated photoreceptor cultures. INTERPRETATION: This study supports the hypothesis that RPGR mutations lead to actin dysregulation. We have identified several pathways that are interrupted in RPGR-mutant photoreceptor cultures and could be contributing to disease. This study is the first use, to our knowledge, of human iPSCs with retinitis pigmentosa-causing mutations to look at pathophysiology of disease. FUNDING: Wellcome Trust.

16.
Glia ; 63(9): 1621-35, 2015 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-25847153

RESUMO

Oligodendrocytes, the myelin forming cells of the CNS, are characterized by their numerous membranous extensions, which enwrap neuronal axons and form myelin sheaths. During differentiation oligodendrocytes pass different morphological stages, downregulate the expression of the proteoglycan NG2, and acquire major myelin specific proteins, such as myelin basic proteins (MBP) and proteolipid protein. MBP mRNA is transported in RNA granules along the microtubules (MTs) to the periphery and translated locally. MTs participate in the elaboration and stabilization of the myelin forming extensions and are essential for cellular sorting processes. Their dynamic properties are regulated by microtubule associated proteins (MAPs). The MAP tau is present in oligodendrocytes and involved in the regulation and stabilization of the MT network. To further elucidate the functional significance of tau in oligodendrocytes, we have downregulated tau by siRNA technology and studied the effects on cell differentiation and neuron-glia contact formation. The data show that tau knockdown impairs process outgrowth and leads to a decrease in MBP expression. Furthermore, MBP mRNA transport to distant cellular extensions is impaired and cells remain in the NG2 stage. In myelinating cocultures with dorsal root ganglion neurons, oligodendrocyte precursor cells after tau miR RNA lentiviral knockdown develop into NG2 positive cells with very long and thin processes, contacting axons loosely, but fail to form internodes. This demonstrates that tau is important for MBP mRNA transport and involved in process formation. The disturbance of the balance of tau leads to abnormalities in oligodendrocyte differentiation, neuron-glia contact formation and the early myelination process.


Assuntos
Proteína Básica da Mielina/metabolismo , Oligodendroglia/metabolismo , Transporte de RNA/fisiologia , RNA Mensageiro/metabolismo , Proteínas tau/metabolismo , Animais , Encéfalo/metabolismo , Linhagem Celular , Células Cultivadas , Técnicas de Cocultura , Regulação para Baixo , Gânglios Espinais/metabolismo , Humanos , Microtúbulos/metabolismo , Bainha de Mielina/metabolismo , Neurônios/metabolismo , Proteínas Proto-Oncogênicas c-fyn/metabolismo , Ratos Wistar , Tubulina (Proteína)/metabolismo , Proteínas tau/genética
17.
Ann Neurol ; 76(1): 5-19, 2014 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-24916848

RESUMO

Hypomyelinating leukodystrophies represent a genetically heterogeneous but clinically overlapping group of heritable disorders. Current management approaches in the care of the patient with a hypomyelinating leukodystrophy include use of serial magnetic resonance imaging (MRI) to establish and monitor hypomyelination, molecular diagnostics to determine a specific etiology, and equally importantly, careful attention to neurologic complications over time. Emerging research in oligodendrocyte biology and neuroradiology with bedside applications may result in the possibility of clinical trials in the near term, yet there are significant gaps in knowledge in disease classification, characterization, and outcome measures in this group of disorders. Here we review the biological background of myelination, the clinical and genetic variability in hypomyelinating leukodystrophies, and the insights that can be obtained from current MRI techniques. In addition, we discuss ongoing research approaches to define potential outcome markers for future clinical trials.


Assuntos
Doenças Desmielinizantes Hereditárias do Sistema Nervoso Central/diagnóstico , Imageamento por Ressonância Magnética , Pesquisa Translacional Biomédica , Doenças Desmielinizantes Hereditárias do Sistema Nervoso Central/terapia , Humanos
18.
Development ; 138(20): 4443-50, 2011 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-21880787

RESUMO

The majority of axons in the central nervous system (CNS) are eventually myelinated by oligodendrocytes, but whether the timing and extent of myelination in vivo reflect intrinsic properties of oligodendrocytes, or are regulated by axons, remains undetermined. Here, we use zebrafish to study CNS myelination at single-cell resolution in vivo. We show that the large caliber Mauthner axon is the first to be myelinated (shortly before axons of smaller caliber) and that the presence of supernumerary large caliber Mauthner axons can profoundly affect myelination by single oligodendrocytes. Oligodendrocytes that typically myelinate just one Mauthner axon in wild type can myelinate multiple supernumerary Mauthner axons. Furthermore, oligodendrocytes that exclusively myelinate numerous smaller caliber axons in wild type can readily myelinate small caliber axons in addition to the much larger caliber supernumerary Mauthner axons. These data indicate that single oligodendrocytes can myelinate diverse axons and that their myelinating potential is actively regulated by individual axons.


Assuntos
Axônios/fisiologia , Bainha de Mielina/fisiologia , Oligodendroglia/fisiologia , Peixe-Zebra/fisiologia , Animais , Animais Geneticamente Modificados , Axônios/ultraestrutura , Sequência de Bases , Primers do DNA/genética , Microscopia Eletrônica de Transmissão , Proteína Básica da Mielina/genética , Proteína Básica da Mielina/metabolismo , Bainha de Mielina/ultraestrutura , Oligodendroglia/ultraestrutura , Peixe-Zebra/genética , Peixe-Zebra/crescimento & desenvolvimento , Proteínas de Peixe-Zebra/genética , Proteínas de Peixe-Zebra/metabolismo
19.
Development ; 138(24): 5321-31, 2011 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-22071102

RESUMO

The generation of astrocytes during the development of the mammalian spinal cord is poorly understood. Here, we demonstrate for the first time that the extracellular matrix glycoprotein tenascin C regulates the expression of key patterning genes during late embryonic spinal cord development, leading to a timely maturation of gliogenic neural precursor cells. We first show that tenascin C is expressed by gliogenic neural precursor cells during late embryonic development. The loss of tenascin C leads to a sustained generation and delayed migration of Fgfr3-expressing immature astrocytes in vivo. Consistent with an increased generation of astroglial cells, we documented an increased number of GFAP-positive astrocytes at later stages. Mechanistically, we could demonstrate an upregulation and domain shift of the patterning genes Nkx6.1 and Nkx2.2 in vivo. In addition, sulfatase 1, a known downstream target of Nkx2.2 in the ventral spinal cord, was also upregulated. Sulfatase 1 regulates growth factor signalling by cleaving sulphate residues from heparan sulphate proteoglycans. Consistent with this function, we observed changes in both FGF2 and EGF responsiveness of spinal cord neural precursor cells. Taken together, our data implicate Tnc in the regulation of proliferation and lineage progression of astroglial progenitors in specific domains of the developing spinal cord.


Assuntos
Astrócitos/citologia , Padronização Corporal/genética , Regulação da Expressão Gênica no Desenvolvimento , Neurogênese/genética , Medula Espinal/crescimento & desenvolvimento , Tenascina/metabolismo , Animais , Movimento Celular , Células Cultivadas , Feminino , Fator 2 de Crescimento de Fibroblastos/metabolismo , Proteoglicanas de Heparan Sulfato/metabolismo , Proteína Homeobox Nkx-2.2 , Proteínas de Homeodomínio/metabolismo , Camundongos , Camundongos Knockout , Células-Tronco Neurais/metabolismo , Receptor Tipo 3 de Fator de Crescimento de Fibroblastos/metabolismo , Sulfotransferases/metabolismo , Fatores de Transcrição/metabolismo , Regulação para Cima , Proteínas de Peixe-Zebra
20.
Brain ; 136(Pt 1): 116-31, 2013 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-23365094

RESUMO

Remyelination following central nervous system demyelination is essential to prevent axon degeneration. However, remyelination ultimately fails in demyelinating diseases such as multiple sclerosis. This failure of remyelination is likely mediated by many factors, including changes in the extracellular signalling environment. Here, we examined the expression of the extracellular matrix molecule fibronectin on demyelinating injury and how this affects remyelination by oligodendrocytes progenitors. In toxin-induced lesions undergoing efficient remyelination, fibronectin expression was transiently increased within demyelinated areas and declined as remyelination proceeded. Fibronectin levels increased both by leakage from the blood circulation and by production from central nervous system resident cells. In chronically demyelinated multiple sclerosis lesions, fibronectin expression persisted in the form of aggregates, which may render fibronectin resistant to degradation. Aggregation of fibronectin was similarly observed at the relapse phase of chronic experimental autoimmune encephalitis, but not on toxin-induced demyelination, suggesting that fibronectin aggregation is mediated by inflammation-induced demyelination. Indeed, the inflammatory mediator lipopolysaccharide induced fibronectin aggregation by astrocytes. Most intriguingly, injection of astrocyte-derived fibronectin aggregates in toxin-induced demyelinated lesions inhibited oligodendrocyte differentiation and remyelination, and fibronectin aggregates are barely expressed in remyelinated multiple sclerosis lesions. Therefore, these findings suggest that fibronectin aggregates within multiple sclerosis lesions contribute to remyelination failure. Hence, the inhibitory signals induced by fibronectin aggregates or factors that affect fibronectin aggregation could be potential therapeutic targets for promoting remyelination.


Assuntos
Doenças Desmielinizantes/metabolismo , Encefalomielite Autoimune Experimental/metabolismo , Fibronectinas/metabolismo , Esclerose Múltipla/metabolismo , Bainha de Mielina/metabolismo , Animais , Astrócitos/metabolismo , Astrócitos/patologia , Axônios/metabolismo , Axônios/patologia , Encéfalo/metabolismo , Encéfalo/patologia , Encéfalo/fisiopatologia , Células Cultivadas , Doenças Desmielinizantes/patologia , Doenças Desmielinizantes/fisiopatologia , Encefalomielite Autoimune Experimental/patologia , Encefalomielite Autoimune Experimental/fisiopatologia , Feminino , Humanos , Esclerose Múltipla/patologia , Esclerose Múltipla/fisiopatologia , Bainha de Mielina/patologia , Regeneração Nervosa/fisiologia , Oligodendroglia/metabolismo , Oligodendroglia/patologia , Ratos , Ratos Sprague-Dawley , Ratos Wistar , Medula Espinal/patologia , Medula Espinal/fisiopatologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA