Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 20
Filtrar
1.
J Am Soc Nephrol ; 27(7): 2069-81, 2016 07.
Artigo em Inglês | MEDLINE | ID: mdl-26574047

RESUMO

A positional isomer of 3',5'-cAMP, 2',3'-cAMP, is produced by kidneys in response to energy depletion, and renal 2',3'-cyclic nucleotide 3'-phosphodiesterase (CNPase) metabolizes 2',3'-cAMP to 2'-AMP; 2',3'-cAMP is a potent opener of mitochondrial permeability transition pores (mPTPs), which can stimulate autophagy. Because autophagy protects against AKI, it is conceivable that inhibition of CNPase protects against ischemia-reperfusion (IR) -induced AKI. Therefore, we investigated renal outcomes, mitochondrial function, number, area, and autophagy in CNPase-knockout (CNPase(-/-)) versus wild-type (WT) mice using a unique two-kidney, hanging-weight model of renal bilateral IR (20 minutes of ischemia followed by 48 hours of reperfusion). Analysis of urinary purines showed attenuated metabolism of 2',3'-cAMP to 2'-AMP in CNPase(-/-) mice. Neither genotype nor IR affected BP, heart rate, urine volume, or albumin excretion. In WT mice, renal IR reduced (14)C-inulin clearance (index of GFR) and increased renal vascular resistance (measured by transit time nanoprobes) and urinary excretion of kidney injury molecule-1 and neutrophil gelatinase-associated lipocalin. IR did not affect these parameters in CNPase(-/-) mice. Histologic analysis revealed that IR induced severe damage in kidneys from WT mice, whereas histologic changes were minimal after IR in CNPase(-/-) mice. Measurements of renal cardiolipin levels, citrate synthase activity, rotenone-sensitive NADH oxidase activity, and proximal tubular mitochondrial and autophagosome area and number (by transmission electron microscopy) indicted accelerated autophagy/mitophagy in injured CNPase(-/-) mice. We conclude that CNPase deletion attenuates IR-induced AKI, in part by accelerating autophagy with targeted removal of damaged mitochondria.


Assuntos
2',3'-Nucleotídeo Cíclico 3'-Fosfodiesterase/fisiologia , Injúria Renal Aguda/enzimologia , Injúria Renal Aguda/etiologia , Animais , Feminino , Rim/irrigação sanguínea , Masculino , Camundongos , Camundongos Knockout , Traumatismo por Reperfusão/complicações , Índice de Gravidade de Doença
2.
J Pharmacol Exp Ther ; 354(2): 175-83, 2015 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-25998049

RESUMO

The 3',5'-cAMP-adenosine pathway (3',5'-cAMP→5'-AMP→adenosine) and the 2',3'-cAMP-adenosine pathway (2',3'-cAMP→2'-AMP/3'-AMP→adenosine) are active in the brain. Oligodendrocytes participate in the brain 2',3'-cAMP-adenosine pathway via their robust expression of 2',3'-cyclic nucleotide 3'-phosphodiesterase (CNPase; converts 2',3'-cAMP to 2'-AMP). Because Schwann cells also express CNPase, it is conceivable that the 2',3'-cAMP-adenosine pathway exists in the peripheral nervous system. To test this and to compare the 2',3'-cAMP-adenosine pathway to the 3',5'-cAMP-adenosine pathway in Schwann cells, we examined the metabolism of 2',3'-cAMP, 2'-AMP, 3'-AMP, 3',5'-cAMP, and 5'-AMP in primary rat Schwann cells in culture. Addition of 2',3'-cAMP (3, 10, and 30 µM) to Schwann cells increased levels of 2'-AMP in the medium from 0.006 ± 0.002 to 21 ± 2, 70 ± 3, and 187 ± 10 nM/µg protein, respectively; in contrast, Schwann cells had little ability to convert 2',3'-cAMP to 3'-AMP or 3',5'-cAMP to either 3'-AMP or 5'-AMP. Although Schwann cells slightly converted 2',3'-cAMP and 2'-AMP to adenosine, they did so at very modest rates (e.g., 5- and 3-fold, respectively, more slowly compared with our previously reported studies in oligodendrocytes). Using transected myelinated rat sciatic nerves in culture medium, we observed a time-related increase in endogenous intracellular 2',3'-cAMP and extracellular 2'-AMP. These findings indicate that Schwann cells do not have a robust 3',5'-cAMP-adenosine pathway but do have a 2',3'-cAMP-adenosine pathway; however, because the pathway mostly involves 2'-AMP formation rather than 3'-AMP, and because the conversion of 2'-AMP to adenosine is slow, metabolism of 2',3'-cAMP mostly results in the accumulation of 2'-AMP. Accumulation of 2'-AMP in peripheral nerves postinjury could have pathophysiological consequences.


Assuntos
Nucleotídeos de Adenina/metabolismo , Monofosfato de Adenosina/metabolismo , Líquido Extracelular/metabolismo , Células de Schwann/metabolismo , Animais , Animais Recém-Nascidos , Células Cultivadas , Ratos
3.
Am J Physiol Renal Physiol ; 307(6): F680-5, 2014 Sep 15.
Artigo em Inglês | MEDLINE | ID: mdl-24990899

RESUMO

CD73 metabolizes extracellular 5'-AMP to adenosine; yet recent experiments in brain tissue suggest that CD73 is not required for the metabolism of 5'-AMP to adenosine because of tissue nonspecific alkaline phosphatase (TNAP), which like CD73 is a GPI-anchored ecto-enyzme with 5'-nucleotidase activity. Because adenosine importantly regulates renovascular function, we investigated whether both TNAP and CD73 are involved in the renovascular metabolism of 5'-AMP. To test this, we examined in isolated, perfused mouse kidneys the metabolism of 5'-AMP (applied to the lumen of the renal vasculature via intrarenal artery administration) to adenosine by measuring renal venous levels of 5'-AMP, adenosine, and inosine (adenosine metabolite) by mass spectrometry. In one study, we compared 5'-AMP metabolism in naive CD73+/+ (wild-type, n = 16) vs. CD73-/- (knockout, n = 16) kidneys; and in a second study, we compared 5'-AMP metabolism in CD73+/+ (n = 9) vs. CD73-/- (n = 8) kidneys pretreated with levamisole (1 mmol/l; TNAP inhibitor). In naive kidneys, 5'-AMP increased renal venous 5'-AMP, adenosine, and inosine, and these responses were similar in CD73+/+ vs. CD73-/- kidneys. Levamisole per se did not inhibit renovascular 5'-AMP metabolism; however, in the presence of levamisole, 5'-AMP increased renal venous 5'-AMP threefold more in CD73-/- vs. CD73+/+ kidneys and knockout of CD73 inhibited 5'-induced adenosine and inosine by 81 and 86%, respectively. TNAP mRNA, protein, and activity were similar in CD73+/+ vs. CD73-/- kidneys. In conclusion, CD73 and TNAP play interactive roles to metabolize luminally applied 5'-AMP in the renal vasculature such that inhibition of both is required to inhibit the production of adenosine.


Assuntos
5'-Nucleotidase/metabolismo , Monofosfato de Adenosina/metabolismo , Fosfatase Alcalina/metabolismo , Vasos Sanguíneos/metabolismo , Rim/metabolismo , Animais , Feminino , Rim/irrigação sanguínea , Masculino , Camundongos , Camundongos Knockout
4.
Am J Physiol Cell Physiol ; 304(5): C406-21, 2013 Mar 01.
Artigo em Inglês | MEDLINE | ID: mdl-23242185

RESUMO

The aim of this investigation was to test the hypothesis that extracellular guanosine regulates extracellular adenosine levels. Rat preglomerular vascular smooth muscle cells were incubated with adenosine, guanosine, or both. Guanosine (30 µmol/l) per se had little effect on extracellular adenosine levels. Extracellular adenosine levels 1 h after addition of adenosine (3 µmol/l) were 0.125 ± 0.020 µmol/l, indicating rapid disposition of extracellular adenosine. Extracellular adenosine levels 1 h after addition of adenosine (3 µmol/l) plus guanosine (30 µmol/l) were 1.173 ± 0.061 µmol/l, indicating slow disposition of extracellular adenosine. Cell injury increased extracellular levels of endogenous adenosine and guanosine, and the effects of cell injury on endogenous extracellular adenosine were modulated by altering the levels of endogenous extracellular guanosine with exogenous purine nucleoside phosphorylase (converts guanosine to guanine) or 8-aminoguanosine (inhibits purine nucleoside phosphorylase). Extracellular guanosine also slowed the disposition of extracellular adenosine in rat preglomerular vascular endothelial cells, mesangial cells, cardiac fibroblasts, and kidney epithelial cells and in human aortic and coronary artery vascular smooth muscle cells and coronary artery endothelial cells. The effects of guanosine on adenosine levels were not mimicked or attenuated by 5-iodotubericidin (adenosine kinase inhibitor), erythro-9-(2-hydroxy-3-nonyl)-adenine (adenosine deaminase inhibitor), 5-aminoimidazole-4-carboxamide (guanine deaminase inhibitor), aristeromycin (S-adenosylhomocysteine hydrolase inhibitor), low sodium (inhibits concentrative nucleoside transporters), S-(4-nitrobenzyl)-6-thioinosine [inhibits equilibrative nucleoside transporter (ENT) type 1], zidovudine (inhibits ENT type 2), or acadesine (known modulator of adenosine levels). Guanosine also increases extracellular inosine, uridine, thymidine, and cytidine, yet decreases extracellular uric acid. In conclusion, extracellular guanosine regulates extracellular adenosine levels.


Assuntos
Adenosina/metabolismo , Guanosina/metabolismo , Músculo Liso Vascular/metabolismo , Miócitos de Músculo Liso/metabolismo , Adenosina/farmacologia , Animais , Aorta/metabolismo , Células Cultivadas , Vasos Coronários/metabolismo , Células Endoteliais/efeitos dos fármacos , Células Endoteliais/metabolismo , Fibroblastos/metabolismo , Guanosina/análogos & derivados , Guanosina/farmacologia , Humanos , Rim/irrigação sanguínea , Rim/citologia , Rim/metabolismo , Masculino , Células Mesangiais/metabolismo , Músculo Liso Vascular/efeitos dos fármacos , Miocárdio/citologia , Miócitos de Músculo Liso/efeitos dos fármacos , Purina-Núcleosídeo Fosforilase/metabolismo , Ratos , Ratos Endogâmicos WKY
5.
Glia ; 61(10): 1595-606, 2013 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-23922219

RESUMO

Extracellular adenosine 3',5'-cyclic monophosphate (3',5'-cAMP) is an endogenous source of localized adenosine production in many organs. Recent studies suggest that extracellular 2',3'-cAMP (positional isomer of 3',5'-cAMP) is also a source of adenosine, particularly in the brain in vivo post-injury. Moreover, in vitro studies show that both microglia and astrocytes can convert extracellular 2',3'-cAMP to adenosine. Here, we examined the ability of primary mouse oligodendrocytes and neurons to metabolize extracellular 2',3'-cAMP and their respective adenosine monophosphates (2'-AMP and 3'-AMP). Cells were also isolated from mice deficient in 2',3'-cyclic nucleotide-3'-phosphodiesterase (CNPase). Oligodendrocytes metabolized 2',3'-cAMP to 2'-AMP with 10-fold greater efficiency than did neurons (and also more than previously examined microglia and astrocytes); whereas, the production of 3'-AMP was minimal in both oligodendrocytes and neurons. The production of 2'-AMP from 2',3'-cAMP was reduced by 65% in CNPase -/- versus CNPase +/+ oligodendrocytes. Oligodendrocytes also converted 2'-AMP to adenosine, and this was also attenuated in CNPase -/- oligodendrocytes. Inhibition of classic 3',5'-cAMP-3'-phosphodiesterases with 3-isobutyl-1-methylxanthine did not block metabolism of 2',3'-cAMP to 2'-AMP and inhibition of classic ecto-5'-nucleotidase (CD73) with α,ß-methylene-adenosine-5'-diphosphate did not attenuate the conversion of 2'-AMP to adenosine. These studies demonstrate that oligodendrocytes express the extracellular 2',3'-cAMP-adenosine pathway (2',3'-cAMP → 2'-AMP → adenosine). This pathway is more robustly expressed in oligodendrocytes than in all other CNS cell types because CNPase is the predominant enzyme that metabolizes 2',3'-cAMP to 2-AMP in CNS cells. By reducing levels of 2',3'-cAMP (a mitochondrial toxin) and increasing levels of adenosine (a neuroprotectant), oligodendrocytes may protect axons from injury.


Assuntos
2',3'-Nucleotídeo Cíclico Fosfodiesterases/metabolismo , Adenosina/metabolismo , Líquido Extracelular/metabolismo , Oligodendroglia/metabolismo , Transdução de Sinais/fisiologia , 1-Metil-3-Isobutilxantina/farmacologia , 2',3'-Nucleotídeo Cíclico Fosfodiesterases/deficiência , Nucleotídeos de Adenina/metabolismo , Monofosfato de Adenosina/metabolismo , Trifosfato de Adenosina/análogos & derivados , Trifosfato de Adenosina/farmacologia , Animais , Células Cultivadas , Embrião de Mamíferos , Camundongos , Camundongos Transgênicos , Neurônios/efeitos dos fármacos , Neurônios/fisiologia , Oligodendroglia/efeitos dos fármacos , Inibidores de Fosfodiesterase/farmacologia , Transdução de Sinais/genética
6.
J Pharmacol Exp Ther ; 347(2): 516-28, 2013 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-24023368

RESUMO

Pleckstrin homology domain and leucine-rich repeat protein phosphatase 1 (PHLPP1) inhibits protein kinase B (AKT) survival signaling in neurons. Small molecule pan-PHLPP inhibitors (selective for PHLPP1 and PHLPP2) may offer a translatable method to induce AKT neuroprotection. We tested several recently discovered PHLPP inhibitors (NSC117079 and NSC45586; benzoic acid, 5-[2-[4-[2-(2,4-diamino-5-methylphenyl)diazenyl]phenyl]diazenyl]-2-hydroxy-,sodium salt.) in rat cortical neurons and astrocytes and compared the biochemical response of these agents with short hairpin RNA (shRNA)-mediated PHLPP1 knockdown (KD). In neurons, both PHLPP1 KD and experimental PHLPP inhibitors activated AKT and ameliorated staurosporine (STS)-induced cell death. Unexpectedly, in astrocytes, both inhibitors blocked AKT activation, and NSC117079 reduced viability. Only PHLPP2 KD mimicked PHLPP inhibitors on astrocyte biochemistry. This suggests that these inhibitors could have possible detrimental effects on astrocytes by blocking novel PHLPP2-mediated prosurvival signaling mechanisms. Finally, because PHLPP1 levels are reportedly high in the hippocampus (a region prone to ischemic death), we characterized hippocampal changes in PHLPP and several AKT targeting prodeath phosphatases after cardiac arrest (CA)-induced brain injury. PHLPP1 levels increased in rat brains subjected to CA. None of the other AKT inhibitory phosphatases increased after global ischemia (i.e., PHLPP2, PTEN, PP2A, and PP1). Selective PHLPP1 inhibition (such as by shRNA KD) activates AKT survival signaling in neurons and astrocytes. Nonspecific PHLPP inhibition (by NSC117079 and NSC45586) only activates AKT in neurons. Taken together, these results suggest that selective PHLPP1 inhibitors should be developed and may yield optimal strategies to protect injured hippocampal neurons and astrocytes-namely from global brain ischemia.


Assuntos
Antraquinonas/farmacologia , Astrócitos/efeitos dos fármacos , Compostos Azo/farmacologia , Fármacos Neuroprotetores/farmacologia , Proteínas Nucleares/antagonistas & inibidores , Fenilenodiaminas/farmacologia , Proteínas Proto-Oncogênicas c-akt/metabolismo , Sulfonamidas/farmacologia , Animais , Antraquinonas/química , Astrócitos/metabolismo , Astrócitos/patologia , Compostos Azo/química , Isquemia Encefálica/etiologia , Isquemia Encefálica/metabolismo , Isquemia Encefálica/patologia , Isquemia Encefálica/prevenção & controle , Técnicas de Cultura de Células , Sobrevivência Celular/efeitos dos fármacos , Relação Dose-Resposta a Droga , Células HEK293 , Parada Cardíaca/complicações , Parada Cardíaca/metabolismo , Parada Cardíaca/patologia , Humanos , Estrutura Molecular , Neurônios/efeitos dos fármacos , Neurônios/metabolismo , Neurônios/patologia , Fármacos Neuroprotetores/química , Proteínas Nucleares/genética , Fenilenodiaminas/química , Ratos , Ratos Sprague-Dawley , Transdução de Sinais/efeitos dos fármacos , Sulfonamidas/química
7.
Am J Physiol Renal Physiol ; 303(7): F1000-5, 2012 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-22874760

RESUMO

A(1) receptors may participate in renal sympathetic neurotransmission by enhancing the postjunctional effects of norepinephrine. The purpose of this study was to test this concept using A(1) receptor knockout (A(1)AR-/-) mice. In isolated kidneys from nontransgenic mice perfused with Tyrode's solution at a constant rate, renal sympathetic nerve stimulation (RSNS) increased (P < 0.0001) renal venous perfusate levels of inosine (adenosine metabolite) from 23.9 ± 3.7 to 32.7 ± 5.1, 68.2 ± 12.4, and 94.0 ± 14.3 ng/ml at 3, 5, and 7 Hz, respectively (n = 28), suggesting frequency-dependent production of adenosine. Conversely, RSNS decreased (P < 0.0001) renal venous perfusate levels of 5'-AMP (adenosine precursor) from 1.4 ± 0.3 to 1.1 ± 0.3, 0.80 ± 0.2, and 0.6 ± 0.2 ng/ml at 3, 5, and 7 Hz, respectively (n = 28), suggesting frequency-dependent increased metabolism of 5'-AMP. In kidneys from nontransgenic mice, blockade of adenosine receptors with 1,3-dipropyl-8-p-sulfophenylxanthine attenuated (P = 0.0130) vasoconstrictor responses to RSNS at 3, 5, and 7 Hz [control (n = 29): 22 ± 4, 34 ± 6, 42 ± 6 mmHg, respectively; 1,3-dipropyl-8-p-sulfophenylxanthine-treated (n = 11): 6 ± 1, 12 ± 3, 15 ± 3 mmHg, respectively]. In A(1)AR-/- kidneys (n = 10), vasoconstrictor responses to RSNS at 3, 5, and 7 Hz were 7 ± 3, 20 ± 5, and 36 ± 9 mmHg, respectively. In kidneys from wild-type littermates (n = 9), responses were 27 ± 9, 58 ± 14, and 59 ± 11 mmHg, respectively (effect of genotype: P = 0.0363). In kidneys from nontransgenic mice, 2-chloro-N(6)-cyclopentyladenosine (CCPA; highly selective A(1) receptor agonist) increased renal vasoconstriction induced by norepinephrine (P = 0.0008; n = 28). In kidneys from A(1)AR-/- the response to norepinephrine was attenuated and the ability of CCPA to enhance responses to norepinephrine was abolished. In conclusion, adenosine formed during RSNS enhances the postjunctional effects of released norepinephrine by activating A(1) receptors.


Assuntos
Rim/metabolismo , Receptor A1 de Adenosina/metabolismo , Sistema Nervoso Simpático/metabolismo , Transmissão Sináptica/fisiologia , Antagonistas do Receptor A1 de Adenosina/farmacologia , Animais , Rim/efeitos dos fármacos , Rim/inervação , Masculino , Camundongos , Camundongos Knockout , Norepinefrina/farmacologia , Receptor A1 de Adenosina/genética , Sistema Nervoso Simpático/efeitos dos fármacos , Transmissão Sináptica/efeitos dos fármacos , Vasoconstrição/efeitos dos fármacos , Vasoconstrição/fisiologia , Xantinas/farmacologia
8.
J Neurochem ; 122(1): 115-25, 2012 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-22360621

RESUMO

Although multiple biochemical pathways produce adenosine, studies suggest that the 2',3'-cAMP-adenosine pathway (2',3'-cAMP→2'-AMP/3'-AMP→adenosine) contributes to adenosine production in some cells/tissues/organs. To determine whether the 2',3'-cAMP-adenosine pathway exists in vivo in the brain, we delivered to the brain (gray matter and white matter separately) via the inflow perfusate of a microdialysis probe either 2',3'-cAMP, 3',5'-cAMP, 2'-AMP, 3'-AMP, or 5'-AMP and measured the recovered metabolites in the microdialysis outflow perfusate with mass spectrometry. In both gray and white matter, 2',3'-cAMP increased 2'-AMP, 3'-AMP and adenosine, and 3',5'-cAMP increased 5'-AMP and adenosine. In both brain regions, 2'-AMP, 3-AMP and 5'-AMP were converted to adenosine. Microdialysis experiments in 2',3'-cyclic nucleotide-3'-phosphodiesterase (CNPase) wild-type mice demonstrated that traumatic brain injury (controlled cortical impact model) activated the brain 2',3'-cAMP-adenosine pathway; similar experiments in CNPase knockout mice indicated that CNPase was involved in the metabolism of endogenous 2',3'-cAMP to 2'-AMP and to adenosine. In CSF from traumatic brain injury patients, 2',3'-cAMP was significantly increased in the initial 12 h after injury and strongly correlated with CSF levels of 2'-AMP, 3'-AMP, adenosine and inosine. We conclude that in vivo, 2',3'-cAMP is converted to 2'-AMP/3'-AMP, and these AMPs are metabolized to adenosine. This pathway exists endogenously in both mice and humans.


Assuntos
Nucleotídeos de Adenina/metabolismo , Adenosina/metabolismo , Encéfalo/metabolismo , Transdução de Sinais/fisiologia , 2',3'-Nucleotídeo Cíclico Fosfodiesterases/deficiência , Monofosfato de Adenosina/metabolismo , Adulto , Animais , Encéfalo/citologia , Lesões Encefálicas/líquido cefalorraquidiano , Lesões Encefálicas/patologia , AMP Cíclico/metabolismo , Modelos Animais de Doenças , Líquido Extracelular/metabolismo , Feminino , Regulação da Expressão Gênica/genética , Escala de Coma de Glasgow , Humanos , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Microdiálise , Transdução de Sinais/genética , Adulto Jovem
9.
J Neurosci ; 30(34): 11388-97, 2010 Aug 25.
Artigo em Inglês | MEDLINE | ID: mdl-20739560

RESUMO

Misexpression and cytosolic retention of peripheral myelin protein 22 (PMP22) within Schwann cells (SCs) is associated with a genetically heterogeneous group of demyelinating peripheral neuropathies. PMP22 overproducer C22 and spontaneous mutant Trembler J (TrJ) mice display neuropathic phenotypes and affected nerves contain abnormally localized PMP22. Nutrient deprivation-induced autophagy is able to suppress the formation of PMP22 aggregates in a toxin-induced cellular model, and improve locomotor performance and myelination in TrJ mice. As a step toward therapies, we assessed whether pharmacological activation of autophagy by rapamycin (RM) could facilitate the processing of PMP22 within neuropathic SCs and enhance their capacity to myelinate peripheral axons. Exposure of mouse SCs to RM induced autophagy in a dose- and time-dependent manner and decreased the accumulation of poly-ubiquitinated substrates. The treatment of myelinating dorsal root ganglion (DRG) explant cultures from neuropathic mice with RM (25 nm) improved the processing of PMP22 and increased the abundance and length of myelin internodes, as well as the expression of myelin proteins. Notably, RM is similarly effective in both the C22 and TrJ model, signifying that the benefit overlaps among distinct genetic models of PMP22 neuropathies. Furthermore, lentivirus-mediated shRNA knockdown of the autophagy-related gene 12 (Atg12) abolished the activation of autophagy and the increase in myelin proteins, demonstrating that autophagy is critical for the observed improvement. Together, these results support the potential use of RM and other autophagy-enhancing compounds as therapeutic agents for PMP22-associated demyelinating neuropathies.


Assuntos
Autofagia/efeitos dos fármacos , Doenças Desmielinizantes/tratamento farmacológico , Fibras Nervosas Mielinizadas/efeitos dos fármacos , Neuralgia/tratamento farmacológico , Sirolimo/farmacologia , Sirolimo/uso terapêutico , Animais , Autofagia/fisiologia , Doenças Desmielinizantes/patologia , Feminino , Técnicas de Silenciamento de Genes , Masculino , Camundongos , Camundongos Mutantes Neurológicos , Bainha de Mielina/efeitos dos fármacos , Bainha de Mielina/patologia , Fibras Nervosas Mielinizadas/patologia , Neuralgia/patologia , Técnicas de Cultura de Órgãos
10.
J Neurochem ; 118(6): 979-87, 2011 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-21777245

RESUMO

Many organs express the extracellular 3',5'-cAMP-adenosine pathway (conversion of extracellular 3',5'-cAMP to 5'-AMP and 5'-AMP to adenosine). Some organs release 2',3'-cAMP (isomer of 3',5'-cAMP) and convert extracellular 2',3'-cAMP to 2'- and 3'-AMP and convert these AMPs to adenosine (extracellular 2',3'-cAMP-adenosine pathway). As astrocytes and microglia are important participants in the response to brain injury and adenosine is an endogenous neuroprotectant, we investigated whether these extracellular cAMP-adenosine pathways exist in these cell types. 2',3'-, 3',5'-cAMP, 5'-, 3'-, and 2'-AMP were incubated with mouse primary astrocytes or primary microglia for 1 h and purine metabolites were measured in the medium by mass spectrometry. There was little evidence of a 3',5'-cAMP-adenosine pathway in either astrocytes or microglia. In contrast, both cell types converted 2',3'-cAMP to 2'- and 3'-AMP (with 2'-AMP being the predominant product). Although both cell types converted 2'- and 3'-AMP to adenosine, microglia were five- and sevenfold, respectively, more efficient than astrocytes in this regard. Inhibitor studies indicated that the conversion of 2',3'-cAMP to 2'-AMP was mediated by a different ecto-enzyme than that involved in the metabolism of 2',3'-cAMP to 3'-AMP and that although CD73 mediates the conversion of 5'-AMP to adenosine, an alternative ecto-enzyme metabolizes 2'- or 3'-AMP to adenosine.


Assuntos
Nucleotídeos de Adenina/metabolismo , Adenosina/metabolismo , Astrócitos/metabolismo , Microglia/metabolismo , Transdução de Sinais/fisiologia , 1-Metil-3-Isobutilxantina/farmacologia , 5'-Nucleotidase/metabolismo , Animais , Astrócitos/efeitos dos fármacos , Separação Celular , Cromatografia Líquida de Alta Pressão , AMP Cíclico/metabolismo , Feminino , Imunofluorescência , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Microglia/efeitos dos fármacos , Inibidores de Fosfodiesterase/farmacologia , Espectrometria de Massas por Ionização por Electrospray , Espectrometria de Massas em Tandem , Xantinas/farmacologia
11.
J Neurochem ; 115(4): 941-55, 2010 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-20819118

RESUMO

Pleckstrin homology and leucine rich repeat protein phosphatases (PHLPPs) are a novel class of potent protein kinase B (AKT) inhibitors that have been intensely investigated in relation to AKT activity in cancer. Currently, our understanding of the role of PHLPP1α in the central nervous system is limited. In this study, we characterized PHLPP protein expression and target kinases in the adult hippocampus. We directly verify PHLPP1α inhibits AKT in hippocampal neurons and demonstrate a novel role for PHLPP1ß/SCOP, to promote AKT activation. PHLPP1α expression changes dramatically in the hippocampus during development, constituting the most abundant PHLPP protein in adult neurons. Further, while all PHLPP proteins could be observed in the cytosolic fraction, only PHLPP1α could be localized to the nucleus. The results provide unique evidence for a divergence in the function of PHLPP1α and PHLPP1ß/SCOP, and suggest that PHLPP1α plays a major role in regulating AKT signaling in neurons.


Assuntos
Hipocampo/fisiologia , Neurônios/fisiologia , Proteínas Nucleares/genética , Fosfoproteínas Fosfatases/genética , Proteína Quinase C-alfa/fisiologia , Proteínas Proto-Oncogênicas c-akt/fisiologia , Splicing de RNA , Transdução de Sinais/genética , Envelhecimento/genética , Envelhecimento/fisiologia , Animais , Linhagem Celular , Células Cultivadas , Feminino , Hipocampo/embriologia , Hipocampo/enzimologia , Humanos , Isoenzimas/genética , Isoenzimas/fisiologia , Masculino , Neurônios/enzimologia , Neurônios/metabolismo , Proteínas Nucleares/fisiologia , Fosfoproteínas Fosfatases/fisiologia , Proteína Quinase C-alfa/metabolismo , Proteínas Proto-Oncogênicas c-akt/antagonistas & inibidores , Proteínas Proto-Oncogênicas c-akt/metabolismo , Splicing de RNA/fisiologia , Ratos , Ratos Endogâmicos BN , Ratos Endogâmicos F344
12.
J Neurosci Res ; 88(12): 2558-68, 2010 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-20648646

RESUMO

The process of Schwann cell myelination requires precisely coordinated gene expression. At the onset of myelination, there is an increase in the expression of differentiation-promoting transcription factors that regulate key Schwann cell genes. Further control of myelin gene expression occurs at the posttranscriptional level and, in part, is mediated by RNA binding proteins and micro-RNAs (miRNAs). miRNAs are small, endogenously derived RNA molecules that repress gene expression by specifically binding to their mRNA targets. In the experiments described here, we tested whether miRNAs were essential in controlling myelination by reducing the levels of Dicer, an essential endoribonuclease in miRNA biogenesis. We decreased the expression of Dicer by about 60% within Schwann cells using a lentiviral vector expressing an shRNA against Dicer. The reduced levels of Dicer led to a decrease in the steady-state expression of selected miRNAs and of the transcription factors Oct6 and Egr2/Krox20, both of which are critical for Schwann cells differentiation and myelination. In contrast, the levels of c-jun and Sox2 were up-regulated by the reduction in Dicer and were associated with an increase in Schwann cell proliferation. In dorsal root ganglion cocultures, Schwann cells transduced with Dicer shRNA synthesized less myelin, which was accompanied by significant reductions in the levels of myelin basic protein and protein zero. These findings support a critical role for Dicer and miRNAs in Schwann cell differentiation and myelination.


Assuntos
Diferenciação Celular/genética , MicroRNAs/genética , Bainha de Mielina/metabolismo , Ribonuclease III/genética , Ribonuclease III/metabolismo , Células de Schwann/metabolismo , Animais , Animais Recém-Nascidos , Células Cultivadas , Técnicas de Cocultura , Regulação para Baixo/fisiologia , Gânglios Espinais/citologia , Gânglios Espinais/metabolismo , Bainha de Mielina/fisiologia , Interferência de RNA/fisiologia , Ratos , Células de Schwann/citologia , Células Receptoras Sensoriais/citologia , Células Receptoras Sensoriais/metabolismo , Células-Tronco/metabolismo , Células-Tronco/fisiologia , Regulação para Cima/fisiologia
13.
J Neurosci ; 28(45): 11720-30, 2008 Nov 05.
Artigo em Inglês | MEDLINE | ID: mdl-18987208

RESUMO

MicroRNAs (miRNAs) play important roles in modulating gene expression at the posttranscriptional level. In postnatal oligodendrocyte lineage cells, the miRNA expression profile ("microRNAome") contains 43 miRNAs whose expression dynamically changes during the transition from A2B5(+) oligodendrocyte progenitor cells to premyelinating GalC(+) cells. The combination of microRNAome profiling with analyses of the oligodendrocyte transcriptome reveals a target bias for a class of miRNAs which includes miR-9. We show that miR-9 is downregulated during oligodendrocyte differentiation. In addition, miR-9 expression level inversely correlates with the expression of its predicted targets, among which is the peripheral myelin protein PMP22. We found that PMP22 mRNA but not protein is detectable in oligodendrocytes, whereas Schwann cells producing PMP22 protein lack miR-9. We demonstrate that miR-9 interacts with the 3' untranslated region of PMP22 and downregulates its expression. Our results support models in which miRNAs can act as guardians of the transcriptome.


Assuntos
Regulação da Expressão Gênica no Desenvolvimento/fisiologia , MicroRNAs/metabolismo , Dinâmica não Linear , Oligodendroglia/fisiologia , RNA Mensageiro/metabolismo , Animais , Animais Recém-Nascidos , Antígenos CD/metabolismo , Encéfalo/citologia , Células Cultivadas , Biologia Computacional , Galactosilceramidas/metabolismo , Gangliosídeos/genética , Gangliosídeos/metabolismo , Proteínas de Fluorescência Verde/biossíntese , Análise em Microsséries/métodos , Proteínas da Mielina/metabolismo , Ratos , Ratos Sprague-Dawley , Nervo Isquiático/citologia , Transfecção/métodos
14.
Glia ; 57(12): 1265-79, 2009 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-19170179

RESUMO

Peripheral myelin protein 22 (PMP22) is a dose-sensitive, disease-associated protein primarily expressed in myelinating Schwann cells. Either reduction or overproduction of PMP22 can result in hereditary neuropathy, suggesting a requirement for correct protein expression for peripheral nerve biology. PMP22 is post-transcriptionally regulated and the 3'untranslated region (3'UTR) of the gene exerts a negative effect on translation. MicroRNAs (miRNAs) are small regulatory molecules that function at a post-transcriptional level by targeting the 3'UTR in a reverse complementary manner. We used cultured Schwann cells to demonstrate that alterations in the miRNA biogenesis pathway affect PMP22 levels, and endogenous PMP22 is subjected to miRNA regulation. GW-body formation, the proposed cytoplasmic site for miRNA-mediated repression, and Dicer expression, an RNase III family ribonuclease involved in miRNA biogenesis, are co-regulated with the differentiation state of Schwann cells. Furthermore, the levels of Dicer inversely correlate with PMP22, while the inhibition of Dicer leads to elevated PMP22. Microarray analysis of actively proliferating and differentiated Schwann cells, in conjunction with bioinformatics programs, identified several candidate PMP22-targeting miRNAs. Here we demonstrate that miR-29a binds and inhibits PMP22 reporter expression through a specific miRNA seed binding region. Over-expression of miR-29a enhances the association of PMP22 RNA with Argonaute 2, a protein involved in miRNA function, and reduces the steady-state levels of PMP22. In contrast, inhibition of endogenous miR-29a relieves the miRNA-mediated repression of PMP22. Correlation analyses of miR-29 and PMP22 in sciatic nerves reveal an inverse relationship, both developmentally and in post-crush injury. These results identify PMP22 as a target of miRNAs and suggest that myelin gene expression by Schwann cells is regulated by miRNAs.


Assuntos
Regulação da Expressão Gênica , MicroRNAs/metabolismo , Proteínas da Mielina/genética , Proteínas da Mielina/metabolismo , Células de Schwann/fisiologia , Animais , Proteínas Argonautas , Western Blotting , Proliferação de Células , Células Cultivadas , Fator de Iniciação 2 em Eucariotos/metabolismo , Imunoprecipitação , Proteínas de Membrana/metabolismo , Compressão Nervosa , Análise de Sequência com Séries de Oligonucleotídeos , RNA Interferente Pequeno/metabolismo , Ratos , Ribonuclease III/antagonistas & inibidores , Ribonuclease III/genética , Ribonuclease III/metabolismo , Nervo Isquiático/crescimento & desenvolvimento , Nervo Isquiático/lesões , Nervo Isquiático/fisiologia , Transdução de Sinais/fisiologia , Fatores de Tempo , Transfecção
15.
Neurobiol Dis ; 34(1): 146-54, 2009 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-19320048

RESUMO

Charcot-Marie-Tooth type 1A (CMT1A) neuropathies linked to the misexpression of peripheral myelin protein 22 (PMP22) are progressive demyelinating disorders of the peripheral nervous system. In this study we asked whether dietary restriction by intermittent fasting (IF) could alleviate the neuropathic phenotype in the Trembler J (TrJ) mouse model of CMT1A. Our results show that neuropathic mice kept on a five month long IF regimen had improved locomotor performance compared to ad libitum (AL) fed littermates. The functional benefits of this dietary intervention are associated with an increased expression of myelin proteins combined with a thicker myelin sheath, less redundant basal lamina, and a reduction in aberrant Schwann cell proliferation. These morphological improvements are accompanied by a decrease in PMP22 protein aggregates, and enhanced expression of cytosolic chaperones and constituents of the autophagy-lysosomal pathway. These results indicate that dietary restriction is beneficial for peripheral nerve function in TrJ neuropathic mice, as it promotes the maintenance of locomotor performance.


Assuntos
Doença de Charcot-Marie-Tooth/dietoterapia , Jejum , Análise de Variância , Animais , Membrana Basal/fisiopatologia , Proliferação de Células , Doença de Charcot-Marie-Tooth/patologia , Doença de Charcot-Marie-Tooth/fisiopatologia , Modelos Animais de Doenças , Locomoção , Masculino , Camundongos , Camundongos Mutantes , Proteínas da Mielina/metabolismo , Proteínas da Mielina/fisiologia , Bainha de Mielina/fisiologia , Bainha de Mielina/ultraestrutura , Células de Schwann/fisiologia , Nervo Isquiático/patologia , Nervo Isquiático/fisiopatologia
16.
Brain Res ; 1594: 27-35, 2015 Jan 12.
Artigo em Inglês | MEDLINE | ID: mdl-25451117

RESUMO

BACKGROUND: Some cells, tissues and organs release 2',3'-cAMP (a positional isomer of 3',5'-cAMP) and convert extracellular 2',3'-cAMP to 2'-AMP plus 3'-AMP and convert these AMPs to adenosine (called the extracellular 2',3'-cAMP-adenosine pathway). Recent studies show that microglia have an extracellular 2',3'-cAMP-adenosine pathway. The goal of the present study was to investigate whether the extracellular 2',3'-cAMP-adenosine pathway could have functional consequences on the production of cytokines/chemokines by activated microglia. METHODS: Experiments were conducted in cultures of primary murine microglia. In the first experiment, the effect of 2',3'-cAMP, 3'-AMP, 2'-AMP and adenosine on LPS-induced TNF-α and CXCL10 production was determined. In the next experiment, the first protocol was replicated but with the addition of 1,3-dipropyl-8-p-sulfophenylxanthine (DPSPX) (0.1 µM; antagonist of adenosine receptors). The last experiment compared the ability of 2-chloro-N(6)-cyclopentyladenosine (CCPA) (10 µM; selective A1 agonist), 5'-N-ethylcarboxamide adenosine (NECA) (10 µM; agonist for all adenosine receptor subtypes) and CGS21680 (10 µM; selective A2A agonist) to inhibit LPS-induced TNF-α and CXCL10 production. RESULTS: (1) 2',3'-cAMP, 3'-AMP, 2'-AMP and adenosine similarly inhibited LPS-induced TNF-α and CXCL10 production; (2) DPSPX nearly eliminated the inhibitory effects of 2',3'-cAMP, 3'-AMP, 2'-AMP and adenosine on LPS-induced TNF-α and CXCL10 production; (3) CCPA did not affect LPS-induced TNF-α and CXCL10; (4) NECA and CGS21680 similarly inhibited LPS-induced TNF-α and CXCL10 production. CONCLUSIONS: 2',3'-cAMP and its metabolites (3'-AMP, 2'-AMP and adenosine) inhibit LPS-induced TNF-α and CXCL10 production via A2A-receptor activation. Adenosine and its precursors, via A2A receptors, likely suppress TNF-α and CXCL10 production by activated microglia in brain diseases.


Assuntos
Nucleotídeos de Adenina/metabolismo , Quimiocina CXCL10/biossíntese , Microglia/metabolismo , Receptor A2A de Adenosina/metabolismo , Fator de Necrose Tumoral alfa/biossíntese , Adenosina/metabolismo , Monofosfato de Adenosina/metabolismo , Animais , Células Cultivadas , Camundongos , Camundongos Endogâmicos C57BL
17.
Resuscitation ; 85(2): 284-91, 2014 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-24513126

RESUMO

Neuro-cognitive disabilities are a well-recognized complication of hypothermic circulatory arrest. We and others have reported that prolonged cardiac arrest (CA) produces neuronal death and microglial proliferation and activation that are only partially mitigated by hypothermia. Microglia, and possibly other cells, are suggested to elaborate tumor necrosis factor alpha (TNF-α), which can trigger neuronal death cascades and exacerbate edema after CNS insults. Minocycline is neuroprotective in some brain ischemia models in part by blunting the microglial response. We tested the hypothesis that minocycline would attenuate neuroinflammation as reflected by brain tissue levels of TNF-α after hypothermic CA in rats. Rats were subjected to rapid exsanguination, followed by a 6 min normothermic CA. Hypothermia (30 °C) was then induced by an aortic saline flush. After a total of 20 min CA, resuscitation was achieved via cardiopulmonary bypass (CPB). After 5 min reperfusion, minocycline (90 mg kg−1; n = 6) or vehicle (PBS; n = 6) was given. Hypothermia (34 °C) was maintained for 6 h. Rats were sacrificed at 6 or 24 h. TNF-α was quantified (ELISA) in four brain regions (cerebellum, CEREB; cortex, CTX; hippocampus, HIP; striatum, STRI). Naïve rats (n = 6) and rats subjected to the same anesthesia and CPB but no CA served as controls (n = 6). Immunocytochemistry was used to localize TNF-α. Naïve rats and CPB controls had no detectable TNF-α in any brain region. CA markedly increased brain TNF-α. Regional differences were seen, with the highest TNF-α levels in striatum in CA groups (10-fold higher, P < 0.05 vs. all other brain regions). TNF-α was undetectable at 24 h. Minocycline attenuated TNF-α levels in CTX, HIP and STRI (P < 0.05). TNF-α showed unique co-localization with neurons. In conclusion, we report region-dependent early increases in brain TNF-α levels after prolonged hypothermic CA, with maximal increases in striatum. Surprisingly, TNF-α co-localized in neurons and not microglia. Minocycline attenuated TNF-α by approximately 50% but did not totally ablate its production. That minocycline decreased brain TNF-α levels suggests that it may represent a therapeutic adjunct to hypothermia in CA neuroprotection. University of Pittsburgh IACUC 0809278B-3.


Assuntos
Encéfalo/metabolismo , Parada Cardíaca/patologia , Minociclina/farmacologia , Neurônios/efeitos dos fármacos , Neurônios/patologia , Fármacos Neuroprotetores/farmacologia , Fator de Necrose Tumoral alfa/metabolismo , Animais , Reanimação Cardiopulmonar , Hipotermia Induzida , Imuno-Histoquímica , Masculino , Microglia/efeitos dos fármacos , Microglia/patologia , Monitorização Fisiológica , Ratos Sprague-Dawley , Taxa de Sobrevida
18.
Physiol Rep ; 1(3)2013 Aug 20.
Artigo em Inglês | MEDLINE | ID: mdl-24066228

RESUMO

Adenosine formed during renal sympathetic nerve stimulation (RSNS) enhances, by activating A1 receptors, the postjunctional effects of released norepinephrine and participates in renal sympathetic neurotransmission. Because in many cell types CD73 (ecto-5'-nucleotidase) is important for the conversion of 5'-AMP to adenosine, we investigated whether CD73 is necessary for normal renal sympathetic neurotransmission. In isolated kidneys from CD73 wild-type mice (CD73 +/+; n=17) perfused at a constant rate with Tyrode's solution, RSNS increased perfusion pressure by 17±4, 36±8 and 44±10 mm Hg at 3, 5 and 7 Hz, respectively. Similar responses were elicited from kidneys isolated from CD73 knockout mice (CD73 -/-; n=13; 28±11, 43±10 and 44±10 mm Hg at 3, 5 and 7 Hz, respectively); and a high concentration (100 µmol/L) of α,ß-methyleneadenosine 5'-diphosphate (CD73 inhibitor) did not alter responses to RSNS in C57BL/6 mouse kidneys (n=5; 21±5, 36±8 and 43±9 at 3, 5 and 7 Hz, respectively). Measurements of renal venous adenosine and inosine (adenosine metabolite) by liquid chromatography-tandem mass spectrometry demonstrated that the metabolism of exogenous 5'-AMP to adenosine and inosine was similar in CD73 -/- versus CD73 +/+ kidneys. A1 receptor mRNA expression was increased in CD73 -/- kidneys, and 2-chloro-N6-cyclopentyladenosine (0.1 µmol/L; A1 receptor agonist) enhanced renovascular responses to norepinephrine more in CD73 -/- versus CD73 +/+ kidneys. We conclude that CD73 is not essential for renal sympathetic neurotransmission because in the absence of renal CD73 other enzymes metabolize 5'-AMP to adenosine and because of compensatory upregulation of postjunctional coincident signaling between norepinephrine and adenosine.

19.
PLoS One ; 6(5): e20553, 2011.
Artigo em Inglês | MEDLINE | ID: mdl-21647387

RESUMO

The disease processes underlying inherited retinal disease are complex and are not completely understood. Many of the corrective gene therapies designed to treat diseases linked to mutations in genes specifically expressed in photoreceptor cells restore function to these cells but fail to stop progression of the disease. There is growing consensus that effective treatments for these diseases will require delivery of multiple therapeutic proteins that will be selected to treat specific aspects of the disease process. The purpose of this study was to design a lentiviral transgene that reliably expresses all of the proteins it encodes and does so in a consistent manner among infected cells. We show, using both in vitro and in vivo analyses, that bicistronic lentiviral transgenes encoding two fluorescent proteins fused to a viral 2A-like cleavage peptide meet these expression criteria. To determine if this transgene design is suitable for therapeutic applications, we replaced one of the fluorescent protein genes with the gene encoding guanylate cyclase-1 (GC1) and delivered lentivirus carrying this transgene to the retinas of the GUCY1*B avian model of Leber congenital amaurosis-1 (LCA1). GUCY1*B chickens carry a null mutation in the GC1 gene that disrupts photoreceptor function and causes blindness at hatching, a phenotype that closely matches that observed in humans with LCA1. We found that treatment of these animals with the 2A lentivector encoding GC1 restored vision to these animals as evidenced by the presence of optokinetic reflexes. We conclude that 2A-like peptides, with proper optimization, can be successfully incorporated into therapeutic vectors designed to deliver multiple proteins to neural retinal. These results highlight the potential of this vector design to serve as a platform for the development of combination therapies designed to enhance or prolong the benefits of corrective gene therapies.


Assuntos
Genes Virais/genética , Vetores Genéticos/genética , Amaurose Congênita de Leber/fisiopatologia , Lentivirus/genética , Proteínas Luminescentes/genética , Recuperação de Função Fisiológica/genética , Visão Ocular/fisiologia , Animais , Embrião de Galinha , Modelos Animais de Doenças , Expressão Gênica , Guanilato Ciclase/genética , Células HEK293 , Humanos , Amaurose Congênita de Leber/genética , Amaurose Congênita de Leber/terapia , Regiões Promotoras Genéticas/genética , Receptores de Superfície Celular/genética , Retina/metabolismo , Retina/fisiopatologia , Transfecção , Transgenes/genética
20.
Neurobiol Dis ; 25(2): 252-65, 2007 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-17174099

RESUMO

The accumulation of misfolded proteins is associated with various neurodegenerative conditions. Peripheral myelin protein 22 (PMP22) is a hereditary neuropathy-linked, short-lived molecule that forms aggresomes when the proteasome is inhibited or the protein is mutated. We previously showed that the removal of pre-existing PMP22 aggregates is assisted by autophagy. Here we examined whether the accumulation of such aggregates could be suppressed by experimental induction of autophagy and/or chaperones. Enhancement of autophagy during proteasome inhibition hinders protein aggregate formation and correlates with a reduction in accumulated proteasome substrates. Conversely, simultaneous inhibition of autophagy and the proteasome augments the formation of aggregates. An increase of heat shock protein levels by geldanamycin treatment or heat shock preconditioning similarly hampers aggresome formation. The beneficial effects of autophagy and chaperones in preventing the accumulation of misfolded PMP22 are additive and provide a potential avenue for therapeutic approaches in hereditary neuropathies linked to PMP22 mutations.


Assuntos
Transtornos Heredodegenerativos do Sistema Nervoso/metabolismo , Chaperonas Moleculares/metabolismo , Proteínas da Mielina/metabolismo , Fibras Nervosas Mielinizadas/metabolismo , Nervos Periféricos/metabolismo , Doenças do Sistema Nervoso Periférico/metabolismo , Animais , Animais Recém-Nascidos , Autofagia/fisiologia , Células Cultivadas , Citoplasma/metabolismo , Citoplasma/patologia , Proteínas de Choque Térmico/metabolismo , Transtornos Heredodegenerativos do Sistema Nervoso/genética , Transtornos Heredodegenerativos do Sistema Nervoso/fisiopatologia , Corpos de Inclusão/metabolismo , Corpos de Inclusão/ultraestrutura , Camundongos , Camundongos Mutantes Neurológicos , Microscopia Eletrônica de Transmissão , Fibras Nervosas Mielinizadas/patologia , Nervos Periféricos/patologia , Nervos Periféricos/fisiopatologia , Doenças do Sistema Nervoso Periférico/genética , Doenças do Sistema Nervoso Periférico/fisiopatologia , Fagossomos/metabolismo , Fagossomos/ultraestrutura , Complexo de Endopeptidases do Proteassoma/metabolismo , Dobramento de Proteína , Ratos
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA