Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 13 de 13
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
J Neurosci ; 41(42): 8710-8724, 2021 10 20.
Artigo em Inglês | MEDLINE | ID: mdl-34507952

RESUMO

We report that the neurotrophin receptor p75 contributes to sensory neuron survival through the regulation of cholesterol metabolism in Schwann cells. Selective deletion of p75 in mouse Schwann cells of either sex resulted in a 30% loss of dorsal root ganglia (DRG) neurons and diminished thermal sensitivity. P75 regulates Schwann cell cholesterol biosynthesis in response to BDNF, forming a co-receptor complex with ErbB2 and activating ErbB2-mediated stimulation of sterol regulatory element binding protein 2 (SREBP2), a master regulator of cholesterol synthesis. Schwann cells lacking p75 exhibited decreased activation of SREBP2 and a reduction in 7-dehydrocholesterol (7-DHC) reductase (DHCR7) expression, resulting in accumulation of the neurotoxic intermediate, 7-dehyrocholesterol in the sciatic nerve. Restoration of DHCR7 in p75 null Schwann cells in mice significantly attenuated DRG neuron loss. Together, these results reveal a mechanism by which the disruption of lipid metabolism in glial cells negatively influences sensory neuron survival, which has implications for a wide range of peripheral neuropathies.SIGNIFICANCE STATEMENT Although expressed in Schwann cells, the role of p75 in myelination has remained unresolved in part because of its dual expression in sensory neurons that Schwann cells myelinate. When p75 was deleted selectively among Schwann cells, myelination was minimally affected, while sensory neuron survival was reduced by 30%. The phenotype is mainly due to dysregulation of cholesterol biosynthesis in p75-deficient Schwann cells, leading to an accumulation of neurotoxic cholesterol precursor, 7-dehydrocholesterol (7-DHC). Mechanism-wise, we discovered that in response to BDNF, p75 recruits and activates ErbB2 independently of ErbB3, thereby stimulating the master regulator, sterol regulatory element binding protein 2 (SREBP2). These results together highlight a novel role of p75 in Schwann cells in regulating DRG neuron survival by orchestrating proper cholesterol metabolism.


Assuntos
Receptores de Fator de Crescimento Neural/deficiência , Receptores de Fator de Crescimento Neural/genética , Células de Schwann/metabolismo , Células Receptoras Sensoriais/metabolismo , Animais , Sobrevivência Celular/fisiologia , Células Cultivadas , Feminino , Células HEK293 , Humanos , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Ratos , Células de Schwann/ultraestrutura , Células Receptoras Sensoriais/ultraestrutura
2.
J Biol Chem ; 289(45): 31458-72, 2014 Nov 07.
Artigo em Inglês | MEDLINE | ID: mdl-25253694

RESUMO

p75 is expressed among Purkinje cells in the adult cerebellum, but its function has remained obscure. Here we report that p75 is involved in maintaining the frequency and regularity of spontaneous firing of Purkinje cells. The overall spontaneous firing activity of Purkinje cells was increased in p75(-/-) mice during the phasic firing period due to a longer firing period and accompanying reduction in silence period than in the wild type. We attribute these effects to a reduction in small conductance Ca(2+)-activated potassium (SK) channel activity in Purkinje cells from p75(-/-) mice compared with the wild type littermates. The mechanism by which p75 regulates SK channel activity appears to involve its ability to activate Rac1. In organotypic cultures of cerebellar slices, brain-derived neurotrophic factor increased RacGTP levels by activating p75 but not TrkB. These results correlate with a reduction in RacGTP levels in synaptosome fractions from the p75(-/-) cerebellum, but not in that from the cortex of the same animals, compared with wild type littermates. More importantly, we demonstrate that Rac1 modulates SK channel activity and firing patterns of Purkinje cells. Along with the finding that spine density was reduced in p75(-/-) cerebellum, these data suggest that p75 plays a role in maintaining normalcy of Purkinje cell firing in the cerebellum in part by activating Rac1 in synaptic compartments and modulating SK channels.


Assuntos
Cerebelo/metabolismo , Neuropeptídeos/metabolismo , Células de Purkinje/metabolismo , Receptores de Fator de Crescimento Neural/metabolismo , Proteínas rac1 de Ligação ao GTP/metabolismo , Animais , Eletrofisiologia , Complexo de Golgi/metabolismo , Glicoproteínas de Membrana/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Microscopia Eletrônica , Técnicas de Patch-Clamp , Canais de Potássio/metabolismo , Proteínas Tirosina Quinases/metabolismo , Transdução de Sinais , Canais de Potássio Ativados por Cálcio de Condutância Baixa/metabolismo , Sinaptossomos/metabolismo , Tetraetilamônio/química , Proteínas rac de Ligação ao GTP/metabolismo
3.
J Neurosci ; 33(2): 397-410, 2013 Jan 09.
Artigo em Inglês | MEDLINE | ID: mdl-23303920

RESUMO

The lack of effective therapies for spinal cord injury points to the need for identifying novel targets for therapeutic intervention. Here we report that a small molecule, LM11A-31, developed to block proNGF-p75 interaction and p75-mediated cell death crosses the blood-brain barrier efficiently when delivered orally. Administered starting 4 h postinjury, LM11A-31 promotes functional recovery without causing any toxicity or increased pain in a mouse model of spinal contusion injury. In both weight-bearing open-field tests and nonweight-bearing swim tests, LM11A-31 was effective in improving motor function and coordination. Such functional improvement correlated with a >50% increase in the number of surviving oligodendrocytes and myelinated axons. We also demonstrate that LM11A-31 indeed inhibits proNGF-p75 interaction in vivo, thereby curtailing the JNK3-mediated apoptotic cascade. These results thus highlight p75 as a novel therapeutic target for an orally delivered treatment for spinal cord injury.


Assuntos
Isoleucina/análogos & derivados , Morfolinas/uso terapêutico , Bainha de Mielina/metabolismo , Fator de Crescimento Neural/metabolismo , Precursores de Proteínas/metabolismo , Receptor de Fator de Crescimento Neural/efeitos dos fármacos , Receptor de Fator de Crescimento Neural/metabolismo , Traumatismos da Medula Espinal/tratamento farmacológico , Animais , Western Blotting , DNA/genética , Relação Dose-Resposta a Droga , Membro Anterior/fisiologia , Membro Posterior/fisiologia , Hiperalgesia/tratamento farmacológico , Imuno-Histoquímica , Isoleucina/uso terapêutico , Locomoção/efeitos dos fármacos , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Mitocôndrias/efeitos dos fármacos , Mitocôndrias/metabolismo , Proteína Quinase 10 Ativada por Mitógeno/metabolismo , Reação em Cadeia da Polimerase , Traumatismos da Medula Espinal/patologia , Natação/fisiologia
4.
Cerebellum ; 12(3): 300-3, 2013 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-23307658

RESUMO

The p75 neurotrophin receptor is highly expressed in the developing nervous system and is required for neuronal survival, growth, and synaptic transmission. In young mice, p75 is present in both granular cells and Purkinje cells of the cerebellum. Although p75 has been implicated in modulation of neuronal excitability in several neuronal types, whether and how it affects the excitability of cerebellar Purkinje neurons remained unclear. Using extracellular recordings of spontaneous firing of Purkinje neurons in cerebellar slices prepared from wild type and p75 knockout mice, we measured intrinsic firing properties in the presence of fast synaptic blockers of more than 200 Purkinje cells, each for a period of 5 min, for each genotype. We detected a significant increase in the mean firing frequency in p75(-/-) neurons comparing to the wild type littermates. Upon separating tonically firing from phasically firing cells, i.e., cells with firing pauses of longer than 300 ms, we observed that the change mainly arose from phasic firing cells and can be explained by an increase in the firing/silence ratio and a decrease in the number of long pauses during the 5-min recording period. We conclude that p75 plays an important role in regulating the firing-to-silence transition during the phasic firing period of the spontaneous firing of Purkinje cells. Thus, p75 exerts a modulatory function on Purkinje cell firing patterns, through which it may act as a key player in motor coordination and other cerebellum-regulated activities since Purkinje cells represent the sole neuronal output of the cerebellar cortex.


Assuntos
Potenciais de Ação/genética , Cerebelo/citologia , Células de Purkinje/fisiologia , Receptores de Fator de Crescimento Neural/deficiência , Animais , Animais Recém-Nascidos , Técnicas In Vitro , Camundongos , Camundongos Knockout
5.
Neuron ; 75(5): 824-37, 2012 Sep 06.
Artigo em Inglês | MEDLINE | ID: mdl-22958823

RESUMO

Although Aß peptides are causative agents in Alzheimer's disease (AD), the underlying mechanisms are still elusive. We report that Aß42 induces a translational block by activating AMPK, thereby inhibiting the mTOR pathway. This translational block leads to widespread ER stress, which activates JNK3. JNK3 in turn phosphorylates APP at T668, thereby facilitating its endocytosis and subsequent processing. In support, pharmacologically blocking translation results in a significant increase in Aß42 in a JNK3-dependent manner. Thus, JNK3 activation, which is increased in human AD cases and a familial AD (FAD) mouse model, is integral to perpetuating Aß42 production. Concomitantly, deletion of JNK3 from FAD mice results in a dramatic reduction in Aß42 levels and overall plaque loads and increased neuronal number and improved cognition. This reveals AD as a metabolic disease that is under tight control by JNK3.


Assuntos
Doença de Alzheimer/metabolismo , Peptídeos beta-Amiloides/metabolismo , Proteína Quinase 10 Ativada por Mitógeno/metabolismo , Fragmentos de Peptídeos/metabolismo , Estresse Fisiológico/fisiologia , Doença de Alzheimer/patologia , Peptídeos beta-Amiloides/biossíntese , Peptídeos beta-Amiloides/toxicidade , Animais , Modelos Animais de Doenças , Humanos , Camundongos , Camundongos Endogâmicos , Camundongos Knockout , Proteína Quinase 10 Ativada por Mitógeno/deficiência , Proteína Quinase 10 Ativada por Mitógeno/genética , Técnicas de Cultura de Órgãos , Fragmentos de Peptídeos/biossíntese , Fragmentos de Peptídeos/toxicidade , Cultura Primária de Células , Ratos
6.
J Biol Chem ; 287(2): 1600-8, 2012 Jan 06.
Artigo em Inglês | MEDLINE | ID: mdl-22128191

RESUMO

Brain-derived neurotrophic factor (BDNF) was shown to play a role in Schwann cell myelination by recruiting Par3 to the axon-glial interface, but the underlying mechanism has remained unclear. Here we report that Par3 regulates Rac1 activation by BDNF but not by NRG1-Type III in Schwann cells, although both ligands activate Rac1 in vivo. During development, active Rac1 signaling is localized to the axon-glial interface in Schwann cells by a Par3-dependent polarization mechanism. Knockdown of p75 and Par3 individually inhibits Rac1 activation, whereas constitutive activation of Rac1 disturbs the polarized activation of Rac1 in vivo. Polarized Rac1 activation is necessary for myelination as Par3 knockdown attenuates myelination in mouse sciatic nerves as well as in zebrafish. Specifically, Par3 knockdown in zebrafish disrupts proper alignment between the axon and Schwann cells without perturbing Schwann cell migration, suggesting that localized Rac1 activation at the axon-glial interface helps identify the initial wrapping sites. We therefore conclude that polarization of Rac1 activation is critical for myelination.


Assuntos
Fator Neurotrófico Derivado do Encéfalo/metabolismo , Proteínas de Transporte/metabolismo , Moléculas de Adesão Celular/metabolismo , Bainha de Mielina/metabolismo , Proteínas do Tecido Nervoso/metabolismo , Neuropeptídeos/metabolismo , Células de Schwann/metabolismo , Transdução de Sinais/fisiologia , Proteínas de Peixe-Zebra/metabolismo , Peixe-Zebra/embriologia , Proteínas rac de Ligação ao GTP/metabolismo , Proteínas rac1 de Ligação ao GTP/metabolismo , Proteínas Adaptadoras de Transdução de Sinal , Animais , Axônios/metabolismo , Fator Neurotrófico Derivado do Encéfalo/genética , Proteínas de Transporte/genética , Moléculas de Adesão Celular/genética , Proteínas de Ciclo Celular , Movimento Celular/fisiologia , Células Cultivadas , Camundongos , Proteínas do Tecido Nervoso/genética , Neuroglia/citologia , Neuroglia/metabolismo , Neuropeptídeos/genética , Ratos , Células de Schwann/citologia , Peixe-Zebra/genética , Proteínas de Peixe-Zebra/genética , Proteínas rac de Ligação ao GTP/genética , Proteínas rac1 de Ligação ao GTP/genética
7.
J Neurosci ; 31(41): 14481-7, 2011 Oct 12.
Artigo em Inglês | MEDLINE | ID: mdl-21994364

RESUMO

Axonal mRNA transport is robust in cultured neurons but there has been limited evidence for this in vivo. We have used a genetic approach to test for in vivo axonal transport of reporter mRNAs. We show that ß-actin's 3'-UTR can drive axonal localization of GFP mRNA in mature DRG neurons, but mice with γ-actin's 3'-UTR show no axonal GFP mRNA. Peripheral axotomy triggers transport of the ß-actin 3'-UTR containing transgene mRNA into axons. This GFP-3'-ß-actin mRNA accumulates in injured PNS axons before activation of the transgene promoter peaks in the DRG. Spinal cord injury also increases axonal GFP signals in mice carrying this transgene without any increase in transgene expression in the DRGs. These data show for the first time that the ß-actin 3'-UTR is sufficient for axonal localization in both PNS and CNS neurons in vivo.


Assuntos
Axônios/metabolismo , Gânglios Espinais/citologia , Neurônios/citologia , RNA Mensageiro/metabolismo , Medula Espinal/citologia , Regiões 3' não Traduzidas/genética , Actinas/genética , Actinas/metabolismo , Análise de Variância , Animais , Proteína Quinase Tipo 2 Dependente de Cálcio-Calmodulina/genética , Células Cultivadas , Dendritos/metabolismo , Regulação da Expressão Gênica/genética , Proteínas de Fluorescência Verde/genética , Camundongos , Camundongos Transgênicos , Microscopia Confocal/métodos , Proteínas do Tecido Nervoso/genética , Proteínas do Tecido Nervoso/metabolismo , RNA Mensageiro/genética , RNA Ribossômico 18S/genética , RNA Ribossômico 18S/metabolismo , Células de Schwann/metabolismo , Neuropatia Ciática/metabolismo , Neuropatia Ciática/patologia , Traumatismos da Medula Espinal/metabolismo , Traumatismos da Medula Espinal/patologia
8.
EMBO J ; 30(22): 4665-77, 2011 Sep 30.
Artigo em Inglês | MEDLINE | ID: mdl-21964071

RESUMO

Subcellular localization of mRNAs is regulated by RNA-protein interactions. Here, we show that introduction of a reporter mRNA with the 3'UTR of ß-actin mRNA competes with endogenous mRNAs for binding to ZBP1 in adult sensory neurons. ZBP1 is needed for axonal localization of ß-actin mRNA, and introducing GFP with the 3'UTR of ß-actin mRNA depletes axons of endogenous ß-actin and GAP-43 mRNAs and attenuates both in vitro and in vivo regrowth of severed axons. Consistent with limited levels of ZBP1 protein in adult neurons, mice heterozygous for the ZBP1 gene are haploinsufficient for axonal transport of ß-actin and GAP-43 mRNAs and for regeneration of peripheral nerve. Exogenous ZBP1 can rescue the RNA transport deficits, but the axonal growth deficit is only rescued if the transported mRNAs are locally translated. These data support a direct role for ZBP1 in transport and translation of mRNA cargos in axonal regeneration in vitro and in vivo.


Assuntos
Actinas/genética , Axônios/fisiologia , Glicoproteínas/metabolismo , Regeneração Nervosa/fisiologia , RNA Mensageiro/metabolismo , Regiões 3' não Traduzidas/genética , Actinas/metabolismo , Animais , Transporte Axonal/genética , Proliferação de Células , Células Cultivadas , Proteína GAP-43/deficiência , Proteína GAP-43/genética , Proteína GAP-43/metabolismo , Genes Reporter/genética , Proteínas de Fluorescência Verde/genética , Cones de Crescimento/fisiologia , Camundongos , Camundongos Endogâmicos C57BL , Transporte de RNA/genética , Proteínas de Ligação a RNA/genética , Proteínas de Ligação a RNA/metabolismo , Ratos , Ratos Sprague-Dawley , Células Receptoras Sensoriais/metabolismo
9.
J Biol Chem ; 285(26): 20358-68, 2010 Jun 25.
Artigo em Inglês | MEDLINE | ID: mdl-20421303

RESUMO

During the development of the sympathetic nervous system, the p75 neurotrophin receptor (p75NTR) has a dual function: promoting survival together with TrkA in response to NGF, but inducing cell death upon binding pro or mature brain-derived neurotrophic factor (BDNF). Apoptotic signaling through p75NTR requires activation of the stress kinase, JNK. However, the receptor also undergoes regulated proteolysis, first by a metalloprotease, and then by gamma-secretase, in response to pro-apoptotic ligands and this is necessary for receptor mediated neuronal death (Kenchappa, R. S., Zampieri, N., Chao, M. V., Barker, P. A., Teng, H. K., Hempstead, B. L., and Carter, B. D. (2006) Neuron 50, 219-232). Hence, the relationship between JNK activation and receptor proteolysis remains to be defined. Here, we report that JNK3 activation is necessary for p75NTR cleavage; however, following release of the intracellular domain, there is a secondary activation of JNK3 that is cleavage dependent. Receptor proteolysis and apoptosis were prevented in sympathetic neurons from jnk3(-/-) mice, while activation of JNK by ectopic expression of MEKK1 induced p75NTR cleavage and cell death. Proteolysis of the receptor was not detected until 6 h after BDNF treatment, suggesting that JNK3 promotes cleavage through a transcriptional mechanism. In support of this hypothesis, BDNF up-regulated tumor necrosis factor-alpha-converting enzyme (TACE)/ADAM17 mRNA and protein in wild-type, but not jnk3(-/-) sympathetic neurons. Down-regulation of TACE by RNA interference blocked BDNF-induced p75NTR cleavage and apoptosis, indicating that this metalloprotease is responsible for the initial processing of the receptor. Together, these results demonstrate that p75NTR-mediated activation of JNK3 is required for up-regulation of TACE, which promotes receptor proteolysis, leading to prolonged activation of JNK3 and subsequent apoptosis in sympathetic neurons.


Assuntos
Proteínas ADAM/metabolismo , Apoptose , Proteína Quinase 10 Ativada por Mitógeno/metabolismo , Neurônios/metabolismo , Receptores de Fator de Crescimento Neural/metabolismo , Proteínas ADAM/genética , Proteína ADAM17 , Animais , Antracenos/farmacologia , Western Blotting , Fator Neurotrófico Derivado do Encéfalo/farmacologia , Linhagem Celular , Células Cultivadas , Ativação Enzimática/efeitos dos fármacos , Humanos , Cinética , MAP Quinase Quinase Quinase 1/genética , MAP Quinase Quinase Quinase 1/metabolismo , Camundongos , Camundongos Knockout , Proteína Quinase 10 Ativada por Mitógeno/antagonistas & inibidores , Proteína Quinase 10 Ativada por Mitógeno/genética , Fator de Crescimento Neural/farmacologia , Neurônios/citologia , Neurônios/efeitos dos fármacos , Interferência de RNA , Ratos , Ratos Sprague-Dawley , Receptores de Fator de Crescimento Neural/genética , Gânglio Cervical Superior/citologia , Regulação para Cima
10.
Neoplasia ; 10(11): 1213-21, 2008 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-18953430

RESUMO

Signaling events leading to Schwann cell tumor initiation have been extensively characterized in the context of neurofibromatosis (NF). Similar tumors are also observed in patients with the endocrine neoplasia syndrome Carney complex, which results from inactivating mutations in PRKAR1A. Loss of PRKAR1A causes enhanced protein kinase A activity, although the pathways leading to tumorigenesis are not well characterized. Tissue-specific ablation of Prkar1a in neural crest precursor cells (TEC3KO mice) causes schwannomas with nearly 80% penetrance by 10 months. These heterogeneous neoplasms were clinically characterized as genetically engineered mouse schwannomas, grades II and III. At the molecular level, analysis of the tumors revealed almost complete loss of both NF proteins, despite the fact that transcript levels were increased, implying posttranscriptional regulation. Although Erk and Akt signaling are typically enhanced in NF-associated tumors, we observed no activation of either of these pathways in TEC3KO tumors. Furthermore, the small G proteins Ras, Rac1, and RhoA are all known to be involved with NF signaling. In TEC3KO tumors, all three molecules showed modest increases in total protein, but only Rac1 showed significant activation. These data suggest that dysregulated protein kinase A activation causes tumorigenesis through pathways that overlap but are distinct from those described in NF tumorigenesis.


Assuntos
Subunidade RIalfa da Proteína Quinase Dependente de AMP Cíclico/metabolismo , Neurilemoma/metabolismo , Neurofibromina 1/metabolismo , Neurofibromina 2/metabolismo , Animais , Western Blotting , Subunidade RIalfa da Proteína Quinase Dependente de AMP Cíclico/genética , Imunofluorescência , Regulação Neoplásica da Expressão Gênica , Genes da Neurofibromatose 1 , Genes da Neurofibromatose 2 , Humanos , Imuno-Histoquímica , Camundongos , Camundongos Knockout , Proteínas Monoméricas de Ligação ao GTP/metabolismo , Neurilemoma/genética , Reação em Cadeia da Polimerase , Células de Schwann/metabolismo , Transdução de Sinais
11.
J Biol Chem ; 283(36): 24690-7, 2008 Sep 05.
Artigo em Inglês | MEDLINE | ID: mdl-18625710

RESUMO

p75 and the Nogo receptor form a signaling unit for myelin inhibitory molecules, with p75 being responsible for RhoA activation. Because p75 lacks the GDP/GTP exchange factor domain, it has remained unclear how p75 activates RhoA. Here, we report that Kalirin9, a dual RhoGEF, binds p75 directly and regulates p75-Nogo receptor-dependent RhoA activation and neurite inhibition in response to myelin-associated glycoprotein. The region of p75 that Kalirin9 binds includes its mastoparan-like fifth helix, which was shown to recruit RhoGDI-RhoA. As predicted from the presence of a shared binding site, we found that Kalirin9 competes with RhoGDI for p75 binding in a dose-dependent manner in vitro. In line with these data, myelin-associated glycoprotein addition to cerebellar granule neurons resulted in a reduction in the association of Kalirin9 with p75, and a simultaneous increase in the binding of RhoGDI to p75. These results reveal a mechanism by which the fifth helix of p75 regulates RhoA activation.


Assuntos
Cerebelo/metabolismo , Fatores de Troca do Nucleotídeo Guanina/metabolismo , Neuritos/metabolismo , Receptores de Fator de Crescimento Neural/metabolismo , Receptores de Peptídeos/metabolismo , Proteína rhoA de Ligação ao GTP/metabolismo , Animais , Linhagem Celular , Cerebelo/citologia , Proteínas Ligadas por GPI , Inibidores de Dissociação do Nucleotídeo Guanina/metabolismo , Proteínas da Mielina , Glicoproteína Associada a Mielina/metabolismo , Proteínas do Tecido Nervoso , Receptor Nogo 1 , Estrutura Secundária de Proteína/fisiologia , Estrutura Terciária de Proteína/fisiologia , Ratos , Receptores de Superfície Celular , Receptores de Fatores de Crescimento , Fatores de Troca de Nucleotídeo Guanina Rho , Inibidores da Dissociação do Nucleotídeo Guanina rho-Específico
12.
J Neurosci ; 27(31): 8395-404, 2007 Aug 01.
Artigo em Inglês | MEDLINE | ID: mdl-17670986

RESUMO

Although oligodendrocytes undergo apoptosis after spinal cord injury, molecular mechanisms responsible for their death have been unknown. We report that oligodendrocyte apoptosis is regulated oppositely by c-Jun N-terminal kinase 3 (JNK3) and protein interacting with the mitotic kinase, never in mitosis A I (Pin1), the actions of which converge on myeloid cell leukemia sequence-1 (Mcl-1). Activated after injury, JNK3 induces cytochrome c release by facilitating the degradation of Mcl-1, the stability of which is maintained in part by Pin1. Pin1 binds Mcl-1 at its constitutively phosphorylated site, Thr163Pro, and stabilizes it by inhibiting ubiquitination. After injury JNK3 phosphorylates Mcl-1 at Ser121Pro, facilitating the dissociation of Pin1 from Mcl-1. JNK3 thus induces Mcl-1 degradation by counteracting the protective binding of Pin1. These results are confirmed by the opposing phenotypes observed between JNK3-/- and Pin1-/- mice: oligodendrocyte apoptosis and cytochrome c release are reduced in JNK3-/- but elevated in Pin1-/- mice. This report thus unveils a mechanism by which cytochrome c release is under the opposite control of JNK3 and Pin1, regulators for which the activities are intricately coupled.


Assuntos
Apoptose/fisiologia , Proteína Quinase 10 Ativada por Mitógeno/fisiologia , Oligodendroglia/enzimologia , Peptidilprolil Isomerase/fisiologia , Traumatismos da Medula Espinal/enzimologia , Animais , Apoptose/genética , Camundongos , Camundongos Knockout , Camundongos Transgênicos , Proteína Quinase 10 Ativada por Mitógeno/deficiência , Proteína Quinase 10 Ativada por Mitógeno/genética , Peptidilprolil Isomerase de Interação com NIMA , Oligodendroglia/citologia , Oligodendroglia/patologia , Peptidilprolil Isomerase/deficiência , Peptidilprolil Isomerase/genética , Traumatismos da Medula Espinal/genética , Traumatismos da Medula Espinal/patologia , Ubiquitina/antagonistas & inibidores , Ubiquitina/metabolismo
13.
Biochemistry ; 43(30): 9888-900, 2004 Aug 03.
Artigo em Inglês | MEDLINE | ID: mdl-15274643

RESUMO

A large family of uridine 5'-diphosphate (UDP)-alpha-N-acetylgalactosamine (GalNAc):polypeptide N-acetylgalactosaminyl transferases (ppGalNAc Ts) initiates mucin-type O-glycan biosynthesis at serine and threonine. The peptide substrate specificities of individual family members are not well characterized or understood, leaving an inability to rationally predict or comprehend sites of O-glycosylation. Recently, a kinetic modeling approach demonstrated neighboring residue glycosylation as a major factor modulating the O-glycosylation of the porcine submaxillary gland mucin 81 residue tandem repeat by ppGalNAc T1 and T2 [Gerken et al. (2002) J. Biol. Chem. 277, 49850-49862]. To confirm the general applicability of this model and its parameters, the ppGalNAc T1 and T2 glycosylation kinetics of the 80+ residue tandem repeat from the canine submaxillary gland mucin was obtained and characterized. To reproduce the glycosylation patterns of both mucins (comprising 50+ serine/threonine residues), specific effects of neighboring peptide sequence, in addition to the previously described effects of neighboring residue glycosylation, were required of the model. Differences in specificity of the two transferases were defined by their sensitivities to neighboring proline and nonglycosylated hydroxyamino acid residues, from which a ppGalNAc T2 motif was identified. Importantly, the model can approximate the previously reported ppGalNAc T2 glycosylation kinetics of the IgA1 hinge domain peptide [Iwasaki, et al. (2003) J. Biol. Chem. 278, 5613-5621], further validating both the approach and the ppGalNAc T2 positional weighting parameters. The characterization of ppGalNAc transferase specificity by this approach may prove useful for the search for isoform-specific substrates, the creation of isoform-specific inhibitors, and the prediction of mucin-type O-glycosylation sites.


Assuntos
Modelos Químicos , Mucinas/química , N-Acetilgalactosaminiltransferases/química , Fragmentos de Peptídeos/química , Glândula Submandibular/enzimologia , Sequências de Repetição em Tandem , Sequência de Aminoácidos , Animais , Apoproteínas/química , Apoproteínas/metabolismo , Bovinos , Cães , Glicosilação , Humanos , Imunoglobulina A/química , Imunoglobulina A/metabolismo , Isoenzimas/química , Isoenzimas/metabolismo , Cinética , Dados de Sequência Molecular , Mucinas/metabolismo , N-Acetilgalactosaminiltransferases/isolamento & purificação , N-Acetilgalactosaminiltransferases/metabolismo , Fragmentos de Peptídeos/metabolismo , Estrutura Terciária de Proteína , Análise de Sequência de Proteína , Especificidade por Substrato , Suínos
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...