Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 11 de 11
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
J Neurosci ; 42(34): 6506-6517, 2022 08 24.
Artigo em Inglês | MEDLINE | ID: mdl-35906072

RESUMO

Schwann cells play a critical role after peripheral nerve injury by clearing myelin debris, forming axon-guiding bands of Büngner, and remyelinating regenerating axons. Schwann cells undergo epigenomic remodeling to differentiate into a repair state that expresses unique genes, some of which are not expressed at other stages of Schwann cell development. We previously identified a set of enhancers that are activated in Schwann cells after nerve injury, and we determined whether these enhancers are preprogrammed into the Schwann cell epigenome as poised enhancers before injury. Poised enhancers share many attributes of active enhancers, such as open chromatin, but are marked by repressive histone H3 lysine 27 (H3K27) trimethylation rather than H3K27 acetylation. We find that most injury-induced enhancers are not marked as poised enhancers before injury indicating that injury-induced enhancers are not preprogrammed in the Schwann cell epigenome. Injury-induced enhancers are enriched with AP-1 binding motifs, and the c-JUN subunit of AP-1 had been shown to be critical to drive the transcriptional response of Schwann cells after injury. Using in vivo chromatin immunoprecipitation sequencing analysis in rat, we find that c-JUN binds to a subset of injury-induced enhancers. To test the role of specific injury-induced enhancers, we focused on c-JUN-binding enhancers upstream of the Sonic hedgehog (Shh) gene, which is only upregulated in repair Schwann cells compared with other stages of Schwann cell development. We used targeted deletions in male/female mice to show that the enhancers are required for robust induction of the Shh gene after injury.SIGNIFICANCE STATEMENT The proregenerative actions of Schwann cells after nerve injury depends on profound reprogramming of the epigenome. The repair state is directed by injury-induced transcription factors, like JUN, which is uniquely required after nerve injury. In this study, we test whether the injury program is preprogrammed into the epigenome as poised enhancers and define which enhancers bind JUN. Finally, we test the roles of these enhancers by performing clustered regularly interspaced short palindromic repeat (CRISPR)-mediated deletion of JUN-bound injury enhancers in the Sonic hedgehog gene. Although many long-range enhancers drive expression of Sonic hedgehog at different developmental stages of specific tissues, these studies identify an entirely new set of enhancers that are required for Sonic hedgehog induction in Schwann cells after injury.


Assuntos
Proteínas Hedgehog , Traumatismos dos Nervos Periféricos , Proteínas Proto-Oncogênicas c-jun , Animais , Feminino , Proteínas Hedgehog/metabolismo , Masculino , Camundongos , Bainha de Mielina/metabolismo , Traumatismos dos Nervos Periféricos/genética , Traumatismos dos Nervos Periféricos/metabolismo , Proteínas Proto-Oncogênicas c-jun/metabolismo , Ratos , Células de Schwann/metabolismo , Fator de Transcrição AP-1/metabolismo
2.
J Biol Chem ; 297(1): 100852, 2021 07.
Artigo em Inglês | MEDLINE | ID: mdl-34090875

RESUMO

The induction of nerve injury response genes in Schwann cells depends on both transcriptional and epigenomic reprogramming. The nerve injury response program is regulated by the repressive histone mark H3K27 trimethylation (H3K27me3), deposited by Polycomb repressive complex 2 (PRC2). Loss of PRC2 function leads to early and augmented induction of the injury response gene network in peripheral nerves, suggesting H3K27 demethylases are required for derepression of Polycomb-regulated nerve injury genes. To determine the function of H3K27 demethylases in nerve injury, we generated Schwann cell-specific knockouts of H3K27 demethylase Kdm6b and double knockouts of Kdm6b/Kdm6a (encoding JMJD3 and UTX). We found that H3K27 demethylases are largely dispensable for Schwann cell development and myelination. In testing the function of H3K27 demethylases after injury, we found early induction of some nerve injury genes was diminished compared with control, but most injury genes were largely unaffected at 1 and 7 days post injury. Although it was proposed that H3K27 demethylases are required to activate expression of the cyclin-dependent kinase inhibitor Cdkn2a in response to injury, Schwann cell-specific deletion of H3K27 demethylases affected neither the expression of this gene nor Schwann cell proliferation after nerve injury. To further characterize the regulation of nerve injury response genes, we found that injury genes are associated with repressive histone H2AK119 ubiquitination catalyzed by PRC1, which declines after injury. Overall, our results indicate H3K27 demethylation is not required for induction of injury response genes and that other mechanisms likely are involved in activating Polycomb-repressed injury genes in peripheral nerve.


Assuntos
Inibidor p16 de Quinase Dependente de Ciclina/genética , Histona Desmetilases/genética , Histona Desmetilases com o Domínio Jumonji/genética , Traumatismos dos Nervos Periféricos/genética , Animais , Regulação da Expressão Gênica no Desenvolvimento/genética , Código das Histonas/genética , Histonas/genética , Humanos , Metilação , Camundongos , Traumatismos dos Nervos Periféricos/patologia , Complexo Repressor Polycomb 2/genética , Células de Schwann/metabolismo , Células de Schwann/patologia , Ubiquitinação/genética
3.
Chem Res Toxicol ; 34(6): 1530-1541, 2021 06 21.
Artigo em Inglês | MEDLINE | ID: mdl-33914522

RESUMO

Smoke inhalation from a structure fire is a common route of cyanide poisoning in the U.S. Cyanide inhibits cellular respiration, often leading to death. Its rapid distribution throughout the body can result in injuries to multiple organs, and cyanide victims were reported to experience myocardial infarction and other cardiac complications. However, molecular mechanisms of such complications are yet to be elucidated. While FDA-approved CN antidotes such as sodium thiosulfate and hydroxocobalamin are clinically used, they have foreseeable limitations during mass casualty situations because they require intravenous administration. To facilitate the development of better antidotes and therapeutic treatments, a global view of molecular changes induced by cyanide exposure is necessary. As an exploratory pursuit, we performed oligonucleotide microarrays to establish cardiac transcriptomes of an animal model of nose-only inhalation exposure to hydrogen cyanide (HCN), which is relevant to smoke inhalation. We also profiled cardiac transcriptomes after subcutaneous injection of potassium cyanide (KCN). Although the KCN injection model has often been used to evaluate medical countermeasures, this study demonstrated that cardiac transcriptomes are largely different from that of the HCN inhalation model at multiple time points within 24 h after exposure. Pathway analysis identified that HCN-induced transcriptomes were enriched with genes encoding mediators of pathways critical in modulation of cardiac complications and that a large number of such genes were significantly decreased in expression. We utilized the upstream regulatory analysis to propose drugs that can be potentially employed to treat cyanide-induced cardiac complications.


Assuntos
Traumatismos Cardíacos/complicações , Cianeto de Hidrogênio/intoxicação , Cianeto de Potássio/intoxicação , Animais , Cianeto de Hidrogênio/administração & dosagem , Exposição por Inalação , Injeções Subcutâneas , Masculino , Camundongos , Cianeto de Potássio/administração & dosagem
4.
Elife ; 102021 01 21.
Artigo em Inglês | MEDLINE | ID: mdl-33475496

RESUMO

After nerve injury, myelin and Remak Schwann cells reprogram to repair cells specialized for regeneration. Normally providing strong regenerative support, these cells fail in aging animals, and during chronic denervation that results from slow axon growth. This impairs axonal regeneration and causes significant clinical problems. In mice, we find that repair cells express reduced c-Jun protein as regenerative support provided by these cells declines during aging and chronic denervation. In both cases, genetically restoring Schwann cell c-Jun levels restores regeneration to control levels. We identify potential gene candidates mediating this effect and implicate Shh in the control of Schwann cell c-Jun levels. This establishes that a common mechanism, reduced c-Jun in Schwann cells, regulates success and failure of nerve repair both during aging and chronic denervation. This provides a molecular framework for addressing important clinical problems, suggesting molecular pathways that can be targeted to promote repair in the PNS.


Assuntos
Envelhecimento , Regeneração Nervosa , Proteínas Proto-Oncogênicas c-jun/genética , Células de Schwann/metabolismo , Animais , Feminino , Masculino , Camundongos , Proteínas Proto-Oncogênicas c-jun/metabolismo
5.
Glia ; 66(11): 2487-2502, 2018 11.
Artigo em Inglês | MEDLINE | ID: mdl-30306639

RESUMO

The transition of differentiated Schwann cells to support of nerve repair after injury is accompanied by remodeling of the Schwann cell epigenome. The EED-containing polycomb repressive complex 2 (PRC2) catalyzes histone H3K27 methylation and represses key nerve repair genes such as Shh, Gdnf, and Bdnf, and their activation is accompanied by loss of H3K27 methylation. Analysis of nerve injury in mice with a Schwann cell-specific loss of EED showed the reversal of polycomb repression is required and a rate limiting step in the increased transcription of Neuregulin 1 (type I), which is required for efficient remyelination. However, mouse nerves with EED-deficient Schwann cells display slow axonal regeneration with significantly low expression of axon guidance genes, including Sema4f and Cntf. Finally, EED loss causes impaired Schwann cell proliferation after injury with significant induction of the Cdkn2a cell cycle inhibitor gene. Interestingly, PRC2 subunits and CDKN2A are commonly co-mutated in the transition from benign neurofibromas to malignant peripheral nerve sheath tumors (MPNST's). RNA-seq analysis of EED-deficient mice identified PRC2-regulated molecular pathways that may contribute to the transition to malignancy in neurofibromatosis.


Assuntos
Proliferação de Células/fisiologia , Regulação da Expressão Gênica/genética , Regeneração Nervosa/efeitos dos fármacos , Complexo Repressor Polycomb 2/metabolismo , Células de Schwann/fisiologia , Neuropatia Ciática/fisiopatologia , Animais , Proliferação de Células/efeitos dos fármacos , Imunoprecipitação da Cromatina , Modelos Animais de Doenças , Regulação da Expressão Gênica/efeitos dos fármacos , Histonas/metabolismo , Masculino , Camundongos , Camundongos Transgênicos , Microscopia Eletrônica , Regeneração Nervosa/genética , Proteínas do Tecido Nervoso/genética , Proteínas do Tecido Nervoso/metabolismo , Neurregulinas/metabolismo , Proteína Oncogênica v-akt/metabolismo , Complexo Repressor Polycomb 2/genética , RNA Mensageiro/metabolismo , Ratos , Ratos Sprague-Dawley , Células de Schwann/efeitos dos fármacos , Células de Schwann/ultraestrutura , Transdução de Sinais/efeitos dos fármacos , Transdução de Sinais/genética
6.
Neuroscientist ; 24(6): 627-638, 2018 12.
Artigo em Inglês | MEDLINE | ID: mdl-29307265

RESUMO

The journey of Schwann cells from their origin in the neural crest to their ensheathment and myelination of peripheral nerves is a remarkable one. Their apparent static function in enabling saltatory conduction of mature nerve is not only vital for long-term health of peripheral nerve but also belies an innate capacity of terminally differentiated Schwann cells to radically alter their differentiation status in the face of nerve injury. The transition from migrating neural crest cells to nerve ensheathment, and then myelination of large diameter axons has been characterized extensively and several of the transcriptional networks have been identified. However, transcription factors must also modify chromatin structure during Schwann cell maturation and this review will focus on chromatin modification machinery that is involved in promoting the transition to, and maintenance of, myelinating Schwann cells. In addition, Schwann cells are known to play important regenerative roles after peripheral nerve injury, and information on epigenomic reprogramming of the Schwann cell genome has emerged. Characterization of epigenomic requirements for myelin maintenance and Schwann cell responses to injury will be vital in understanding how the various Schwann cell functions can be optimized to maintain and repair peripheral nerve function.


Assuntos
Epigênese Genética , Células de Schwann/metabolismo , Animais , Cromatina/metabolismo , Humanos
7.
J Neurochem ; 140(3): 368-382, 2017 02.
Artigo em Inglês | MEDLINE | ID: mdl-27891578

RESUMO

Schwann cells and oligodendrocytes are the myelinating cells of the peripheral and central nervous system, respectively. Despite having different myelin components and different transcription factors driving their terminal differentiation there are shared molecular mechanisms between the two. Sox10 is one common transcription factor required for several steps in development of myelinating glia. However, other factors are divergent as Schwann cells need the transcription factor early growth response 2/Krox20 and oligodendrocytes require Myrf. Likewise, some signaling pathways, like the Erk1/2 kinases, are necessary in both cell types for proper myelination. Nonetheless, the molecular mechanisms that control this shared signaling pathway in myelinating cells remain only partially characterized. The hypothesis of this study is that signaling pathways that are similarly regulated in both Schwann cells and oligodendrocytes play central roles in coordinating the differentiation of myelinating glia. To address this hypothesis, we have used genome-wide binding data to identify a relatively small set of genes that are similarly regulated by Sox10 in myelinating glia. We chose one such gene encoding Dual specificity phosphatase 15 (Dusp15) for further analysis in Schwann cell signaling. RNA interference and gene deletion by genome editing in cultured RT4 and primary Schwann cells showed Dusp15 is necessary for full activation of Erk1/2 phosphorylation. In addition, we show that Dusp15 represses expression of several myelin genes, including myelin basic protein. The data shown here support a mechanism by which early growth response 2 activates myelin genes, but also induces a negative feedback loop through Dusp15 to limit over-expression of myelin genes.


Assuntos
Fosfatases de Especificidade Dupla/fisiologia , Sistema de Sinalização das MAP Quinases/fisiologia , Bainha de Mielina/enzimologia , Células de Schwann/enzimologia , Animais , Linhagem Celular , Ativação Enzimática/fisiologia , Feminino , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Bainha de Mielina/genética , Ratos
8.
J Neurosci ; 36(35): 9135-47, 2016 08 31.
Artigo em Inglês | MEDLINE | ID: mdl-27581455

RESUMO

UNLABELLED: The rapid and dynamic transcriptional changes of Schwann cells in response to injury are critical to peripheral nerve repair, yet the epigenomic reprograming that leads to the induction of injury-activated genes has not been characterized. Polycomb Repressive Complex 2 (PRC2) catalyzes the trimethylation of lysine 27 of histone H3 (H3K27me3), which produces a transcriptionally repressive chromatin environment. We find that many promoters and/or gene bodies of injury-activated genes of mature rat nerves are occupied with H3K27me3. In contrast, the majority of distal enhancers that gain H3K27 acetylation after injury are not repressed by H3K27 methylation before injury, which is normally observed in developmentally poised enhancers. Injury induces demethylation of H3K27 in many genes, such as Sonic hedgehog (Shh), which is silenced throughout Schwann cell development before injury. In addition, experiments using a Schwann cell-specific mouse knock-out of the Eed subunit of PRC2 indicate that demethylation is a rate-limiting step in the activation of such genes. We also show that some transcription start sites of H3K27me3-repressed injury genes of uninjured nerves are bound with a mark of active promoters H3K4me3, for example, Shh and Gdnf, and the reduction of H3K27me3 results in increased trimethylation of H3K4. Our findings identify reversal of polycomb repression as a key step in gene activation after injury. SIGNIFICANCE STATEMENT: Peripheral nerve regeneration after injury is dependent upon implementation of a novel genetic program in Schwann cells that supports axonal survival and regeneration. Identifying means to enhance Schwann cell reprogramming after nerve injury could be used to foster effective remyelination in the treatment of demyelinating disorders and in identifying pathways involved in regenerative process of myelination. Although recent progress has identified transcriptional determinants of successful reprogramming of the Schwann cell transcriptome after nerve injury, our results have highlighted a novel epigenomic pathway in which reversal of the Polycomb pathway of repressive histone methylation is required for activation of a significant number of injury-induced genes.


Assuntos
Reprogramação Celular/fisiologia , Epigenômica , Regulação da Expressão Gênica/fisiologia , Células de Schwann/metabolismo , Neuropatia Ciática/metabolismo , Neuropatia Ciática/fisiopatologia , Animais , Benzazepinas/farmacologia , Reprogramação Celular/genética , Imunoprecipitação da Cromatina , Biologia Computacional , Modelos Animais de Doenças , Inibidores Enzimáticos/farmacologia , Histonas/genética , Histonas/metabolismo , Técnicas In Vitro , Histona Desmetilases com o Domínio Jumonji/genética , Histona Desmetilases com o Domínio Jumonji/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Complexo Repressor Polycomb 2/genética , Complexo Repressor Polycomb 2/metabolismo , Pirimidinas/farmacologia , RNA Mensageiro/metabolismo , Ratos , Ratos Sprague-Dawley , Regeneração/genética , Células de Schwann/efeitos dos fármacos , Células de Schwann/fisiologia , Transdução de Sinais/genética
9.
Hum Mol Genet ; 25(18): 3925-3936, 2016 09 15.
Artigo em Inglês | MEDLINE | ID: mdl-27466180

RESUMO

Schwann cells are the myelinating glia of the peripheral nervous system and dysfunction of these cells causes motor and sensory peripheral neuropathy. The transcription factor SOX10 is critical for Schwann cell development and maintenance, and many SOX10 target genes encode proteins required for Schwann cell function. Loss-of-function mutations in the gene encoding myotubularin-related protein 2 (MTMR2) cause Charcot-Marie-Tooth disease type 4B1 (CMT4B1), a severe demyelinating peripheral neuropathy characterized by myelin outfoldings along peripheral nerves. Previous reports indicate that MTMR2 is ubiquitously expressed making it unclear how loss of this gene causes a Schwann cell-specific phenotype. To address this, we performed computational and functional analyses at MTMR2 to identify transcriptional regulatory elements important for Schwann cell expression. Through these efforts, we identified an alternative, SOX10-responsive promoter at MTMR2 that displays strong regulatory activity in immortalized rat Schwann (S16) cells. This promoter directs transcription of a previously unidentified MTMR2 transcript that is enriched in mouse Schwann cells compared to immortalized mouse motor neurons (MN-1), and is predicted to encode an N-terminally truncated protein isoform. The expression of the endogenous transcript is induced in a heterologous cell line by ectopically expressing SOX10, and is nearly ablated in Schwann cells by impairing SOX10 function. Intriguingly, overexpressing the two MTMR2 protein isoforms in HeLa cells revealed that both localize to nuclear puncta and the shorter isoform displays higher nuclear localization compared to the longer isoform. Combined, our data warrant further investigation of the truncated MTMR2 protein isoform in Schwann cells and in CMT4B1 pathogenesis.


Assuntos
Doença de Charcot-Marie-Tooth/genética , Proteínas Tirosina Fosfatases não Receptoras/biossíntese , Elementos Reguladores de Transcrição/genética , Fatores de Transcrição SOXE/genética , Animais , Doença de Charcot-Marie-Tooth/fisiopatologia , Regulação da Expressão Gênica , Células HeLa , Humanos , Camundongos , Neurônios Motores/metabolismo , Neurônios Motores/patologia , Mutação , Bainha de Mielina/genética , Nervos Periféricos/crescimento & desenvolvimento , Nervos Periféricos/metabolismo , Nervos Periféricos/patologia , Regiões Promotoras Genéticas , Proteínas Tirosina Fosfatases não Receptoras/genética , Ratos , Células de Schwann/metabolismo , Células de Schwann/patologia
10.
Neural Regen Res ; 11(12): 1930-1931, 2016 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-28197188
11.
J Neurosci ; 35(22): 8640-52, 2015 Jun 03.
Artigo em Inglês | MEDLINE | ID: mdl-26041929

RESUMO

Myelination of peripheral nerves by Schwann cells requires coordinate regulation of gene repression as well as gene activation. Several chromatin remodeling pathways critical for peripheral nerve myelination have been identified, but the functions of histone methylation in the peripheral nerve have not been elucidated. To determine the role of histone H3 Lys27 methylation, we have generated mice with a Schwann cell-specific knock-out of Eed, which is an essential subunit of the polycomb repressive complex 2 (PRC2) that catalyzes methylation of histone H3 Lys27. Analysis of this mutant revealed no significant effects on early postnatal development of myelin. However, its loss eventually causes progressive hypermyelination of small-diameter axons and apparent fragmentation of Remak bundles. These data identify the PRC2 complex as an epigenomic modulator of mature myelin thickness, which is associated with changes in Akt phosphorylation. Interestingly, we found that Eed inactivation causes derepression of several genes, e.g., Sonic hedgehog (Shh) and Insulin-like growth factor-binding protein 2 (Igfbp2), that become activated after nerve injury, but without activation of a primary regulator of the injury program, c-Jun. Analysis of the activated genes in cultured Schwann cells showed that Igfbp2 regulates Akt activation. Our results identify an epigenomic pathway required for establishing thickness of mature myelin and repressing genes that respond to nerve injury.


Assuntos
Regulação da Expressão Gênica/fisiologia , Bainha de Mielina/metabolismo , Complexo Repressor Polycomb 2/metabolismo , Células de Schwann/metabolismo , Nervo Isquiático/citologia , Animais , Animais Recém-Nascidos , Células Cultivadas , Imunoprecipitação da Cromatina , Proteínas Hedgehog/metabolismo , Proteína 2 de Ligação a Fator de Crescimento Semelhante à Insulina/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Microscopia Eletrônica , Proteína P0 da Mielina/genética , Proteína Oncogênica v-akt/metabolismo , Complexo Repressor Polycomb 2/genética , Ratos , Nervo Isquiático/ultraestrutura , Transdução de Sinais/fisiologia , Transfecção
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...