RESUMO
Pyruvate serves as a key metabolite in energy production and as an anti-oxidant. In our previous study, exogenous pyruvate starvation under high-glucose conditions induced IMS32 Schwann cell death because of the reduced glycolysis-tricarboxylic acid (TCA) cycle flux and adenosine triphosphate (ATP) production. Thus, this study focused on poly-(ADP-ribose) polymerase (PARP) to investigate the detailed molecular mechanism of cell death. Rucaparib, a PARP inhibitor, protected Schwann cells against cell death and decreased glycolysis but not against an impaired TCA cycle under high-glucose conditions in the absence of pyruvate. Under such conditions, reduced pyruvate dehydrogenase (PDH) activity and glycolytic and mitochondrial ATP production were observed but not oxidative phosphorylation or the electric transfer chain. In addition, rucaparib supplementation restored glycolytic ATP production but not PDH activity and mitochondrial ATP production. No differences in the increased activity of caspase 3/7 and the localization of apoptosis-inducing factor were found among the experimental conditions. These results indicate that Schwann cells undergo necrosis rather than apoptosis or parthanatos under the aforementioned conditions. Exogenous pyruvate plays a pivotal role in maintaining the flux in PARP-dependent glycolysis and the PARP-independent TCA cycle in Schwann cells under high-glucose conditions.
Assuntos
Trifosfato de Adenosina , Ciclo do Ácido Cítrico , Glucose , Glicólise , Poli(ADP-Ribose) Polimerases , Ácido Pirúvico , Células de Schwann , Glicólise/efeitos dos fármacos , Ciclo do Ácido Cítrico/efeitos dos fármacos , Trifosfato de Adenosina/metabolismo , Glucose/metabolismo , Ácido Pirúvico/metabolismo , Animais , Células de Schwann/metabolismo , Células de Schwann/efeitos dos fármacos , Poli(ADP-Ribose) Polimerases/metabolismo , Mitocôndrias/metabolismo , Mitocôndrias/efeitos dos fármacos , Ratos , Inibidores de Poli(ADP-Ribose) Polimerases/farmacologia , Indóis/farmacologia , Linhagem Celular , Apoptose/efeitos dos fármacosRESUMO
Hereditary transthyretin (TTR) amyloidosis (ATTRv) is characterized by TTR amyloid deposition in the peripheral nervous system. It remains unknown why variant TTR preferentially deposits in the peripheral nerves and dorsal root ganglia. We previously detected low levels of TTR expression in Schwann cells and established an immortalized Schwann cell line, TgS1, derived from a mouse model of ATTRv amyloidosis expressing the variant TTR gene. In the present study, the expression of TTR and Schwann cell marker genes was investigated in TgS1 cells by quantitative RT-PCR. TTR gene expression was markedly upregulated in TgS1 cells incubated in non-growth medium-Dulbecco's modified Eagle's medium supplemented with 10% fetal bovine serum. The expression levels of c-Jun, Gdnf and Sox2 were increased, while Mpz was downregulated, suggesting that TgS1 cells exhibit a repair Schwann cell-like phenotype in the non-growth medium. Western blot analysis revealed that TTR protein was produced and secreted by the TgS1 cells. Furthermore, downregulation of Hsf1 with siRNA induced TTR aggregates in the TgS1 cells. These findings indicate that TTR expression is markedly increased in repair Schwann cells, likely to promote axonal regeneration. Therefore, aged dysfunctional repair Schwann cells may cause the deposition of variant TTR aggregates in the nerves of patients with ATTRv.
Assuntos
Neuropatias Amiloides Familiares , Pré-Albumina , Camundongos , Animais , Humanos , Idoso , Pré-Albumina/genética , Pré-Albumina/metabolismo , Neuropatias Amiloides Familiares/genética , Células de Schwann/metabolismo , Gânglios Espinais/metabolismo , Expressão GênicaRESUMO
One of the major functions of peroxisomes in mammals is oxidation of very long-chain fatty acids (VLCFAs). Genetic defects in peroxisomal ß-oxidation result in the accumulation of VLCFAs and lead to a variety of health problems, such as demyelination of nervous tissues. However, the mechanisms by which VLCFAs cause tissue degeneration have not been fully elucidated. Recently, we found that the addition of small amounts of isopropanol can enhance the solubility of saturated VLCFAs in an aqueous medium. In this study, we characterized the biological effect of extracellular VLCFAs in peroxisome-deficient Chinese hamster ovary (CHO) cells, neural crest-derived pheochromocytoma cells (PC12), and immortalized adult Fischer rat Schwann cells (IFRS1) using this solubilizing technique. C20:0 FA was the most toxic of the C16-C26 FAs tested in all cells. The basis of the toxicity of C20:0 FA was apoptosis and was observed at 5 µM and 30 µM in peroxisome-deficient and wild-type CHO cells, respectively. The sensitivity of wild-type CHO cells to cytotoxic C20:0 FA was enhanced in the presence of a peroxisomal ß-oxidation inhibitor. Further, a positive correlation was evident between cell toxicity and the extent of intracellular accumulation of toxic FA. These results suggest that peroxisomes are pivotal in the detoxification of apoptotic VLCFAs by preventing their accumulation.
Assuntos
Ácidos Graxos , Peroxissomos , Cricetinae , Animais , Peroxissomos/metabolismo , Ácidos Graxos/metabolismo , Células CHO , Cricetulus , OxirreduçãoRESUMO
It is suggested that activation of receptor for advanced glycation end products (RAGE) induces proinflammatory response in diabetic nerve tissues. Macrophage infiltration is invoked in the pathogenesis of diabetic polyneuropathy (DPN), while the association between macrophage and RAGE activation and the downstream effects of macrophages remain to be fully clarified in DPN. This study explored the role of RAGE in the pathogenesis of DPN through the modified macrophages. Infiltrating proinflammatory macrophages impaired insulin sensitivity, atrophied the neurons in dorsal root ganglion, and slowed retrograde axonal transport (RAT) in the sciatic nerve of type 1 diabetic mice. RAGE-null mice showed an increase in the population of antiinflammatory macrophages, accompanied by intact insulin sensitivity, normalized ganglion cells, and RAT. BM transplantation from RAGE-null mice to diabetic mice protected the peripheral nerve deficits, suggesting that RAGE is a major determinant for the polarity of macrophages in DPN. In vitro coculture analyses revealed proinflammatory macrophage-elicited insulin resistance in the primary neuronal cells isolated from dorsal root ganglia. Applying time-lapse recording disclosed a direct impact of proinflammatory macrophage and insulin resistance on the RAT deficits in primary neuronal cultures. These results provide a potentially novel insight into the development of RAGE-related DPN.
Assuntos
Diabetes Mellitus Experimental , Neuropatias Diabéticas , Resistência à Insulina , Camundongos , Animais , Receptor para Produtos Finais de Glicação Avançada/genética , Diabetes Mellitus Experimental/complicações , MacrófagosRESUMO
Oxaliplatin (OHP) is a platinum-based agent that can cause peripheral neuropathy, an adverse effect in which the dorsal root ganglion (DRG) neurons are targeted. Zonisamide has exhibited neuroprotective activities toward adult rat DRG neurons in vitro and therefore, we aimed to assess its potential efficacy against OHP-induced neurotoxicity. Pretreatment with zonisamide (100 µM) alleviated the DRG neuronal death caused by OHP (75 µM) and the protective effects were attenuated by a co-incubation with 25 µM of the mitogen-activated protein kinase (MAPK; MEK/ERK) inhibitor, U0126, or the phosphatidyl inositol-3'-phosphate-kinase (PI3K) inhibitor, LY294002. Pretreatment with zonisamide also suppressed the OHP-induced p38 MAPK phosphorylation in lined DRG neurons, ND7/23, while the OHP-induced DRG neuronal death was alleviated by pretreatment with the p38 MAPK inhibitor, SB239063 (25 µM). Although zonisamide failed to protect the immortalized rat Schwann cells IFRS1 from OHP-induced cell death, it prevented neurite degeneration and demyelination-like changes, as well as the reduction of the serine/threonine-specific protein kinase (AKT) phosphorylation in DRG neuron-IFRS1 co-cultures exposed to OHP. Zonisamide's neuroprotection against the OHP-induced peripheral sensory neuropathy is possibly mediated by a stimulation of the MEK/ERK and PI3K/AKT signaling pathways and suppression of the p38 MAPK pathway in DRG neurons. Future studies will allow us to solidify zonisamide as a promising remedy against the neurotoxic adverse effects of OHP.
Assuntos
Gânglios Espinais , Doenças do Sistema Nervoso Periférico , Animais , Células Cultivadas , Técnicas de Cocultura , Gânglios Espinais/metabolismo , Quinases de Proteína Quinase Ativadas por Mitógeno/metabolismo , Neurônios/metabolismo , Oxaliplatina/efeitos adversos , Doenças do Sistema Nervoso Periférico/induzido quimicamente , Fosfatidilinositol 3-Quinases/metabolismo , Proteínas Proto-Oncogênicas c-akt/metabolismo , Ratos , Células de Schwann/metabolismo , Zonisamida/efeitos adversos , Zonisamida/metabolismo , Proteínas Quinases p38 Ativadas por Mitógeno/metabolismoRESUMO
Pyruvate functions as a key molecule in energy production and as an antioxidant. The efficacy of pyruvate supplementation in diabetic retinopathy and nephropathy has been shown in animal models; however, its significance in the functional maintenance of neurons and Schwann cells under diabetic conditions remains unknown. We observed rapid and extensive cell death under high-glucose (> 10 mM) and pyruvate-starved conditions. Exposure of Schwann cells to these conditions led to a significant decrease in glycolytic flux, mitochondrial respiration and ATP production, accompanied by enhanced collateral glycolysis pathways (e.g., polyol pathway). Cell death could be prevented by supplementation with 2-oxoglutarate (a TCA cycle intermediate), benfotiamine (the vitamin B1 derivative that suppresses the collateral pathways), or the poly (ADP-ribose) polymerase (PARP) inhibitor, rucaparib. Our findings suggest that exogenous pyruvate plays a pivotal role in maintaining glycolysis-TCA cycle flux and ATP production under high-glucose conditions by suppressing PARP activity.
Assuntos
Nefropatias Diabéticas/patologia , Glucose/metabolismo , Hiperglicemia/complicações , Ácido Pirúvico/metabolismo , Trifosfato de Adenosina/metabolismo , Animais , Linhagem Celular , Sobrevivência Celular/efeitos dos fármacos , Ciclo do Ácido Cítrico/efeitos dos fármacos , Nefropatias Diabéticas/prevenção & controle , Modelos Animais de Doenças , Feminino , Glicólise/efeitos dos fármacos , Humanos , Hiperglicemia/sangue , Hiperglicemia/metabolismo , Indóis/farmacologia , Indóis/uso terapêutico , Camundongos , Neurônios/efeitos dos fármacos , Neurônios/metabolismo , Neurônios/patologia , Poli(ADP-Ribose) Polimerases/metabolismo , Cultura Primária de Células , Ratos , Células de Schwann/efeitos dos fármacos , Células de Schwann/metabolismo , Células de Schwann/patologia , Tiamina/análogos & derivados , Tiamina/farmacologia , Tiamina/uso terapêuticoRESUMO
Schwann cells play an important role in peripheral nerve function, and their dysfunction has been implicated in the pathogenesis of diabetic neuropathy and other demyelinating diseases. The physiological functions of insulin in Schwann cells remain unclear and therefore define the aim of this study. By using immortalized adult Fischer rat Schwann cells (IFRS1), we investigated the mechanism of the stimulating effects of insulin on the cell proliferation and expression of myelin proteins (myelin protein zero (MPZ) and myelin basic protein (MBP). The application of insulin to IFRS1 cells increased the proliferative activity and induced phosphorylation of Akt and ERK, but not P38-MAPK. The proliferative potential of insulin-stimulated IFRS1 was significantly suppressed by the addition of LY294002, a PI3 kinase inhibitor. The insulin-stimulated increase in MPZ expression was significantly suppressed by the addition of PD98059, a MEK inhibitor. Furthermore, insulin-increased MBP expression was significantly suppressed by the addition of LY294002. These findings suggest that both PI3-K/Akt and ERK/MEK pathways are involved in insulin-induced cell growth and upregulation of MPZ and MBP in IFRS1 Schwann cells.
Assuntos
Insulina/farmacologia , Células de Schwann/efeitos dos fármacos , Animais , Divisão Celular/efeitos dos fármacos , Linhagem Celular Transformada , Cromonas/farmacologia , Neuropatias Diabéticas/metabolismo , MAP Quinases Reguladas por Sinal Extracelular/metabolismo , Flavonoides/farmacologia , Regulação da Expressão Gênica/efeitos dos fármacos , Quinases de Proteína Quinase Ativadas por Mitógeno/antagonistas & inibidores , Morfolinas/farmacologia , Proteínas da Mielina/biossíntese , Proteínas da Mielina/genética , Fosfatidilinositol 3-Quinases/efeitos dos fármacos , Fosforilação , Processamento de Proteína Pós-Traducional/efeitos dos fármacos , Proteínas Proto-Oncogênicas c-akt/metabolismo , Ratos , Ratos Endogâmicos F344 , Receptor de Insulina/biossíntese , Receptor de Insulina/genética , Transdução de Sinais/efeitos dos fármacosRESUMO
Besides its insulinotropic actions on pancreatic ß cells, neuroprotective activities of glucagon-like peptide-1 (GLP-1) have attracted attention. The efficacy of a GLP-1 receptor (GLP-1R) agonist exendin-4 (Ex-4) for functional repair after sciatic nerve injury and amelioration of diabetic peripheral neuropathy (DPN) has been reported; however, the underlying mechanisms remain unclear. In this study, the bioactivities of Ex-4 on immortalized adult rat Schwann cells IFRS1 and adult rat dorsal root ganglion (DRG) neuron-IFRS1 co-culture system were investigated. Localization of GLP-1R in both DRG neurons and IFRS1 cells were confirmed using knockout-validated monoclonal Mab7F38 antibody. Treatment with 100 nM Ex-4 significantly enhanced survival/proliferation and migration of IFRS1 cells, as well as stimulated the movement of IFRS1 cells toward neurites emerging from DRG neuron cell bodies in the co-culture with the upregulation of myelin protein 22 and myelin protein zero. Because Ex-4 induced phosphorylation of serine/threonine-specific protein kinase AKT in these cells and its effects on DRG neurons and IFRS1 cells were attenuated by phosphatidyl inositol-3'-phosphate-kinase (PI3K) inhibitor LY294002, Ex-4 might act on both cells to activate PI3K/AKT signaling pathway, thereby promoting myelination in the co-culture. These findings imply the potential efficacy of Ex-4 toward DPN and other peripheral nerve lesions.
Assuntos
Neuropatias Diabéticas/tratamento farmacológico , Exenatida/farmacologia , Peptídeo 1 Semelhante ao Glucagon/genética , Receptor do Peptídeo Semelhante ao Glucagon 1/genética , Animais , Movimento Celular/genética , Sobrevivência Celular/genética , Cromonas/farmacologia , Técnicas de Cocultura , Neuropatias Diabéticas/genética , Neuropatias Diabéticas/patologia , Exenatida/genética , Gânglios Espinais/citologia , Gânglios Espinais/efeitos dos fármacos , Gânglios Espinais/patologia , Receptor do Peptídeo Semelhante ao Glucagon 1/agonistas , Humanos , Células Secretoras de Insulina/efeitos dos fármacos , Células Secretoras de Insulina/metabolismo , Células Secretoras de Insulina/patologia , Morfolinas/farmacologia , Bainha de Mielina/genética , Bainha de Mielina/metabolismo , Neurônios/efeitos dos fármacos , Neurônios/metabolismo , Fosfatidilinositol 3-Quinases/genética , Proteínas Proto-Oncogênicas c-akt/genética , Ratos , Células de Schwann/citologia , Células de Schwann/efeitos dos fármacos , Células de Schwann/metabolismo , Nervo Isquiático/lesõesRESUMO
Aldose reductase (AR) is a member of the reduced nicotinamide adenosine dinucleotide phosphate (NADPH)-dependent aldo-keto reductase superfamily. It is also the rate-limiting enzyme of the polyol pathway, catalyzing the conversion of glucose to sorbitol, which is subsequently converted to fructose by sorbitol dehydrogenase. AR is highly expressed by Schwann cells in the peripheral nervous system (PNS). The excess glucose flux through AR of the polyol pathway under hyperglycemic conditions has been suggested to play a critical role in the development and progression of diabetic peripheral neuropathy (DPN). Despite the intensive basic and clinical studies over the past four decades, the significance of AR over-activation as the pathogenic mechanism of DPN remains to be elucidated. Moreover, the expected efficacy of some AR inhibitors in patients with DPN has been unsatisfactory, which prompted us to further investigate and review the understanding of the physiological and pathological roles of AR in the PNS. Particularly, the investigation of AR and the polyol pathway using immortalized Schwann cells established from normal and AR-deficient mice could shed light on the causal relationship between the metabolic abnormalities of Schwann cells and discordance of axon-Schwann cell interplay in DPN, and led to the development of better therapeutic strategies against DPN.
Assuntos
Aldeído Redutase/metabolismo , Redes e Vias Metabólicas , Polímeros/metabolismo , Células de Schwann/metabolismo , Aldeído Redutase/antagonistas & inibidores , Aldeído Redutase/genética , Animais , Diabetes Mellitus/metabolismo , Inibidores Enzimáticos/química , Inibidores Enzimáticos/farmacologia , Glucose/metabolismo , Humanos , Oxirredução , Sorbitol/metabolismoRESUMO
Patients with transthyretin (TTR)-type familial amyloid polyneuropathy (FAP) typically exhibit sensory dominant polyneuropathy and autonomic neuropathy. However, the molecular pathogenesis of the neuropathy remains unclear. In this study, we characterize the features of FAP TTR the substitution of lysine for glutamic acid at position 61 (E61K). This FAP was late-onset, with sensory dominant polyneuropathy, autonomic neuropathy, and cardiac amyloidosis. Interestingly, no amyloid deposits were found in the endoneurium of the four nerve specimens examined. Therefore, we examined the amyloidogenic properties of E61K TTR in vitro. Recombinant wild-type TTR, the substitution of methionine for valine at position 30 (V30M) TTR, and E61K TTR proteins were incubated at 37°C for 72 hr, and amyloid fibril formation was assessed using the thioflavin-T binding assay. Amyloid fibril formation by E61K TTR was less than that by V30M TTR, and similar to that by wild-type TTR. E61K TTR did not have an inhibitory effect on neurite outgrowth from adult rat dorsal root ganglion (DRG) neurons, but V30M TTR did. Furthermore, we studied the sural nerve of our patient by terminal deoxynucleotidyl transferase dUTP nick end labeling and electron microscopy. A number of apoptotic cells were observed in the endoneurium of the nerve by transferase dUTP nick end labeling. Chromatin condensation was confirmed in the nucleus of non-myelinating Schwann cells by electron microscopy. These findings suggest that E61K TTR is low amyloidogenic, in vitro and in vivo. However, TTR aggregates and amyloid fibrils in the DRG may cause sensory impairments in FAP because the DRG has no blood-nerve barrier. Moreover, Schwann cell apoptosis may contribute to the neurodegeneration.
Assuntos
Neuropatias Amiloides Familiares/genética , Amiloide/biossíntese , Pré-Albumina/genética , Substituição de Aminoácidos , Amiloide/genética , Amiloidose/patologia , Animais , Apoptose , Cristalografia por Raios X , Humanos , Mutação , Nervos Periféricos/patologia , Placa Amiloide/patologia , Pré-Albumina/química , Ratos , Ratos Wistar , Proteínas Recombinantes/farmacologia , Células de Schwann/metabolismo , Nervo Sural/patologiaRESUMO
Glycolaldehyde (GA) is a highly reactive hydroxyaldehyde and one of the glycolytic metabolites producing advanced glycation endproducts (AGEs), but its toxicity toward neurons and Schwann cells remains unclear. In the present study, we found that GA exhibited more potent toxicity than other AGE precursors (glyceraldehyde, glyoxal, methylglyoxal and 3-deoxyglucosone) against immortalized IFRS1 adult rat Schwann cells and ND7/23 neuroblastoma × neonatal rat dorsal root ganglion (DRG) neuron hybrid cells. GA affected adult rat DRG neurons and ND7/23 cells more severely than GA-derived AGEs, and exhibited concentration- and time-dependent toxicity toward ND7/23 cells (10 < 100 < 250 < 500 µM; 6 h < 24 h). Treatment with 500 µM GA significantly up-regulated the phosphorylation of c-jun N-terminal kinase (JNK) and p-38 mitogen-activated kinase (p-38 MAPK) in ND7/23 cells. Furthermore, GA-induced ND7/23 cell death was significantly inhibited due to co-treatment with 10 µM of the JNK inhibitor SP600125 or the p-38 MAPK inhibitor SB239063. These findings suggest the involvement of JNK and p-38 MAPK-signaling pathways in GA-induced neuronal cell death and that enhanced GA production under diabetic conditions might be involved in the pathogenesis of diabetic neuropathy.
Assuntos
Acetaldeído/análogos & derivados , Proteínas Quinases JNK Ativadas por Mitógeno/metabolismo , Células Receptoras Sensoriais/efeitos dos fármacos , Transdução de Sinais/efeitos dos fármacos , Proteínas Quinases p38 Ativadas por Mitógeno/metabolismo , Acetaldeído/farmacologia , Animais , Morte Celular/efeitos dos fármacos , Células Cultivadas , Ativação Enzimática/efeitos dos fármacos , Feminino , Ratos , Ratos Wistar , Células Receptoras Sensoriais/metabolismoRESUMO
Zonisamide, an anti-epileptic and anti-Parkinson's disease drug, displays neurotrophic activity on cultured motor neurons and facilitates axonal regeneration after peripheral nerve injury in mice, but its underlying mechanisms remain unclear. In this study, zonisamide enhanced neurite outgrowth from cultured adult rat dorsal root ganglion (DRG) neurons in a concentration-dependent manner (1 µM < 10 µM < 100 µM), and its activity was significantly attenuated by co-treatment with a phosphatidyl inositol-3'-phosphate-kinase (PI3K) inhibitor LY294002 or a mitogen-activated protein kinase (MAPK) inhibitor U0126. In agreement with these findings, 100 µM zonisamide for 1 h induced phosphorylation of AKT and ERK1/2, key molecules of PI3K and MAPK signaling pathways, respectively in mouse neuroblastoma × rat DRG neuron hybrid cells ND7/23. In contrast, zonisamide failed to promote proliferation or migration of immortalized Fischer rat Schwann cells 1 (IFRS1). These findings suggest that the beneficial effects of zonisamide on peripheral nerve regeneration may be attributable to its direct actions on neurons through PI3K and MAPK pathways, rather than the stimulation of Schwann cells.
Assuntos
Anticonvulsivantes/farmacologia , Gânglios Espinais/efeitos dos fármacos , Neuritos/efeitos dos fármacos , Crescimento Neuronal/efeitos dos fármacos , Neurônios/efeitos dos fármacos , Zonisamida/farmacologia , Animais , Movimento Celular , Proliferação de Células , Células Cultivadas , Relação Dose-Resposta a Droga , Feminino , Gânglios Espinais/metabolismo , Neuritos/metabolismo , Neurônios/metabolismo , Ratos , Ratos Wistar , Células de Schwann/citologia , Células de Schwann/metabolismo , Relação Estrutura-AtividadeRESUMO
A large variety of drugs have been reported to cause peripheral neuropathies as dose-limiting adverse effects; however, most of them primarily affect axons and/or neuronal cell bodies rather than Schwann cells and/or myelin sheaths. In this chapter, we focus on the drugs that seem to elicit the neuropathies with schwannopathy and/or myelinopathy-predominant phenotypes, such as amiodarone, dichloroacetate, and tumor necrosis factor-α antagonists. Although the pathogenesis of demyelination induced by these drugs remain largely obscure, the recent in vivo and in vitro studies have implicated the involvement of metabolic abnormalities and impaired autophagy in Schwann cells and immune system disorders in the disruption of neuron-Schwann cell contact and interactions.
Assuntos
Doenças Desmielinizantes/induzido quimicamente , Efeitos Colaterais e Reações Adversas Relacionados a Medicamentos , Bainha de Mielina/patologia , Doenças do Sistema Nervoso Periférico/induzido quimicamente , Células de Schwann/patologia , Amiodarona/efeitos adversos , Axônios , Ácido Dicloroacético/efeitos adversos , Humanos , Bainha de Mielina/efeitos dos fármacos , Células de Schwann/efeitos dos fármacos , Fator de Necrose Tumoral alfa/antagonistas & inibidoresRESUMO
AIMS/INTRODUCTION: Dental pulp stem cells (DPSCs) can be easily obtained from teeth for general orthodontic reasons. We have previously reported the therapeutic effects of DPSC transplantation for diabetic polyneuropathy. As abundant secretomes from DPSCs are considered to play a central role in the improvement of diabetic polyneuropathy, we investigated whether direct injection of DPSC-conditioned media (DPSC-CM) into hindlimb skeletal muscles ameliorates diabetic polyneuropathy in diabetic rats. MATERIALS AND METHODS: DPSCs were isolated from the dental pulp of Sprague-Dawley rats. Eight weeks after the induction of diabetes, DPSC-CM was injected into the unilateral hindlimb skeletal muscles in both normal and diabetic rats. The effects of DPSC-CM on diabetic polyneuropathy were assessed 4 weeks after DPSC-CM injection. To confirm the angiogenic effect of DPSC-CM, the effect of DPSC-CM on cultured human umbilical vascular endothelial cell proliferation was investigated. RESULTS: The administration of DPSC-CM into the hindlimb skeletal muscles significantly ameliorated sciatic motor/sensory nerve conduction velocity, sciatic nerve blood flow and intraepidermal nerve fiber density in the footpads of diabetic rats. We also showed that DPSC-CM injection significantly increased the capillary density of the skeletal muscles, and suppressed pro-inflammatory reactions in the sciatic nerves of diabetic rats. Furthermore, an in vitro study showed that DPSC-CM significantly increased the proliferation of umbilical vascular endothelial cells. CONCLUSIONS: We showed that DPSC-CM injection into hindlimb skeletal muscles has a therapeutic effect on diabetic polyneuropathy through neuroprotective, angiogenic and anti-inflammatory actions. DPSC-CM could be a novel cell-free regenerative medicine treatment for diabetic polyneuropathy.
Assuntos
Indutores da Angiogênese/farmacologia , Anti-Inflamatórios/farmacologia , Meios de Cultivo Condicionados/farmacologia , Polpa Dentária/citologia , Neuropatias Diabéticas/prevenção & controle , Fármacos Neuroprotetores/farmacologia , Medicina Regenerativa , Células-Tronco/citologia , Animais , Proliferação de Células/efeitos dos fármacos , Diabetes Mellitus Experimental/complicações , Neuropatias Diabéticas/etiologia , Neuropatias Diabéticas/patologia , Células Endoteliais da Veia Umbilical Humana , Humanos , Masculino , Músculo Esquelético/irrigação sanguínea , Músculo Esquelético/efeitos dos fármacos , Condução Nervosa , Ratos , Ratos Sprague-Dawley , Nervo Isquiático/irrigação sanguínea , Nervo Isquiático/efeitos dos fármacosRESUMO
AIMS/INTRODUCTION: Recent studies advocate that omega-3 polyunsaturated fatty acids (ω-3 PUFAs) have direct anti-oxidative and anti-inflammatory effects in the vasculature; however, the role of ω-3 PUFAs in Schwann cells remains undetermined. MATERIALS AND METHODS: Immortalized mouse Schwann (IMS32) cells were incubated with the ω-3 PUFAs docosahexaenoic acid (DHA) and eicosapentaenoic acid (EPA). The messenger ribonucleic acid levels of several anti-oxidant enzymes (heme oxygenase-1 [Ho-1], nicotinamide adenine dinucleotide [phosphate] H quinone oxidoreductase 1, catalase, superoxide dismutase and glutathione peroxidase) were identified using real-time reverse transcription polymerase chain reaction. Ho-1 and nicotinamide adenine dinucleotide [phosphate] H quinone oxidoreductase 1 protein levels were evaluated using Western blotting. Nuclear factor (erythroid-derived 2)-related factor 2 (Nrf2) of the nuclear fraction was also quantified using western blotting. Catalase activity and glutathione content were determined by colorimetric assay kits. Nrf2 promoter-luciferase activity was evaluated by a dual luciferase assay system. RESULTS: Treatment with tert-butyl hydroperoxide decreased cell viability dose-dependently. DHA or EPA pretreatment significantly alleviated tert-butyl hydroperoxide-induced cytotoxicity. DHA or EPA increased the messenger ribonucleic acid levels of Ho-1, nicotinamide adenine dinucleotide (phosphate) H quinone oxidoreductase 1 and catalase dose-dependently. Ho-1 protein level, catalase activity, Nrf2 promoter-luciferase activity and intracellular glutathione content were significantly increased by DHA and EPA. CONCLUSIONS: These findings show that DHA and EPA can induce Ho-1 and catalase through Nrf2, thus protecting Schwann cells against oxidative stress. ω-3 PUFAs appear to exert their neuroprotective effect by increasing defense mechanisms against oxidative stress in diabetic neuropathies.
Assuntos
Antioxidantes/metabolismo , Ácidos Graxos Ômega-3/farmacologia , Regulação Enzimológica da Expressão Gênica/efeitos dos fármacos , Fator 2 Relacionado a NF-E2/metabolismo , Estresse Oxidativo/efeitos dos fármacos , Células de Schwann/efeitos dos fármacos , Animais , Glutationa/metabolismo , Camundongos , Oxirredução , Células de Schwann/metabolismoRESUMO
Hypoglycemia and fluctuating high or low glucose conditions are under-appreciated sources of oxidative stress contributing to diabetic neuropathy. We investigated the effects of recurrent short-term hypoglycemia and hyperglycemia, on apoptosis and oxidative stress in Schwann cells. Immortalized adult mouse Schwann (IMS32) cells were exposed to five different glucose treatments over 3 days: 1) normal glucose (NG), 2) constant low glucose (LG), 3) constant high glucose (HG), 4) intermittent low glucose (ILG; 1 h three times per day), 5) intermittent high glucose (IHG; 1 h three times per day). Cell viability was decreased by all treatment variants, in comparison to NG. Thiobarbituric acid reactive substance (TBARS) levels were increased by HG, LG, IHG, and ILG. High glucose (HG and IHG) and low glucose (LG and ILG) increased the expression of cleaved caspase-3 and reduced that of Bcl-2. In addition, endoplasmic reticulum (ER) stress-responsive transcription factor C/EBP homologous protein (CHOP) expression was increased under low and high glucose conditions. Cell death and oxidative stress induced by HG, LG, IHG, and ILG were significantly reduced by 4-phenyl butyric acid (4-PBA), an ER stress inhibitor. These findings indicate that recurrent short-term hypoglycemia and hyperglycemia induce apoptosis and oxidative stress via the ER stress response in Schwann cells.
Assuntos
Apoptose/efeitos dos fármacos , Glucose/farmacologia , Hiperglicemia/metabolismo , Hipoglicemia/metabolismo , Estresse Oxidativo/efeitos dos fármacos , Animais , Linhagem Celular , Sobrevivência Celular/efeitos dos fármacos , Estresse do Retículo Endoplasmático , Interleucina-6 , Malondialdeído/metabolismo , Camundongos , Fenilbutiratos/farmacologia , Células de Schwann , Fator de Necrose Tumoral alfaRESUMO
Small integral membrane protein of the lysosome/late endosome (SIMPLE) is a 161-amino acid cellular protein that contains a characteristic C-terminal domain known as the SIMPLE-like domain (SLD), which is well conserved among species. Several studies have demonstrated that SIMPLE localizes to the trans-Golgi network (TGN), early endosomes, lysosomes, multivesicular bodies, aggresomes and the plasma membrane. However, the amino acid regions responsible for its subcellular localization have not yet been identified. The SLD resembles the FYVE domain, which binds phosphatidylinositol (3)-phosphate (PI3P) and determines the subcellular localization of FYVE domain-containing proteins. In the present study, we have found that SIMPLE binds specifically to PI4P through its SLD. SIMPLE co-localized with PI4P and Rab11, a marker for recycling endosomes (REs, organelles enriched in PI4P) in both the IMS32 mouse Schwann cell line and Hela cells. Sucrose density-gradient centrifugation revealed that SIMPLE co-fractionated with syntaxin-6 (a TGN marker) and Rab11. We have also found that SIMPLE knockdown impeded recycling of transferrin and of transferrin receptor. Our overall results indicate that SIMPLE may regulate protein trafficking physiologically by localizing to the TGN and/or REs by binding PI4P.
Assuntos
Endossomos/metabolismo , Proteínas Nucleares/metabolismo , Fosfatos de Fosfatidilinositol/metabolismo , Células de Schwann/metabolismo , Fatores de Transcrição/metabolismo , Rede trans-Golgi/metabolismo , Animais , Linhagem Celular Transformada , Proteínas de Ligação a DNA , Endossomos/genética , Células HeLa , Humanos , Camundongos , Proteínas Nucleares/genética , Fosfatos de Fosfatidilinositol/genética , Domínios Proteicos , Fatores de Transcrição/genética , Proteínas rab de Ligação ao GTP/genética , Proteínas rab de Ligação ao GTP/metabolismo , Rede trans-Golgi/genéticaRESUMO
BACKGROUND: Although previous reports have revealed the therapeutic potential of stem cell transplantation in diabetic polyneuropathy, the effects of cell transplantation on long-term diabetic polyneuropathy have not been investigated. In this study, we investigated whether the transplantation of dental pulp stem cells (DPSCs) ameliorated long-term diabetic polyneuropathy in streptozotocin (STZ)-induced diabetic rats. METHODS: Forty-eight weeks after STZ injection, we transplanted DPSCs into the unilateral hindlimb skeletal muscles. Four weeks after DPSC transplantation (i.e., 52 weeks after STZ injection) the effects of DPSC transplantation on diabetic polyneuropathy were assessed. RESULTS: STZ-induced diabetic rats showed significant reductions in the sciatic motor/sensory nerve conduction velocity, increases in the current perception threshold, and decreases in capillary density in skeletal muscles and intra-epidermal nerve fiber density compared with normal rats, all of which were ameliorated by DPSC transplantation. Furthermore, sural nerve morphometrical analysis revealed that the transplantation of DPSCs significantly increased the myelin thickness and area. DPSC-conditioned media promoted the neurite outgrowth of dorsal root ganglion neurons and increased the viability and myelin-related protein expression of Schwann cells. CONCLUSIONS: These results indicated that the transplantation of DPSCs contributed to the neurophysiological and neuropathological recovery from a long duration of diabetic polyneuropathy.
Assuntos
Polpa Dentária/citologia , Diabetes Mellitus Experimental/terapia , Neuropatias Diabéticas/terapia , Nervo Isquiático/patologia , Transplante de Células-Tronco/métodos , Células-Tronco/citologia , Animais , Polpa Dentária/fisiologia , Diabetes Mellitus Experimental/induzido quimicamente , Diabetes Mellitus Experimental/patologia , Neuropatias Diabéticas/induzido quimicamente , Neuropatias Diabéticas/patologia , Gânglios Espinais/patologia , Gânglios Espinais/fisiopatologia , Incisivo/citologia , Incisivo/fisiologia , Masculino , Músculo Esquelético/patologia , Músculo Esquelético/fisiopatologia , Fibras Nervosas Mielinizadas/patologia , Condução Nervosa/fisiologia , Ratos , Ratos Sprague-Dawley , Células de Schwann/patologia , Nervo Isquiático/fisiopatologia , Células-Tronco/fisiologia , EstreptozocinaRESUMO
An increase in the rates of morbidity and mortality associated with diabetic complications is a global concern. Glycemic control is important to prevent the development and progression of diabetic complications. Various classes of anti-diabetic agents are currently available, and their pleiotropic effects on diabetic complications have been investigated. Incretin-based therapies such as dipeptidyl peptidase (DPP)-4 inhibitors and glucagon-like peptide-1 receptor agonists (GLP-1RA) are now widely used in the treatment of patients with type 2 diabetes. A series of experimental studies showed that incretin-based therapies have beneficial effects on diabetic complications, independent of their glucose-lowering abilities, which are mediated by anti-inflammatory and anti-oxidative stress properties. Based on these findings, clinical studies to assess the effects of DPP-4 inhibitors and GLP-1RA on diabetic microvascular and macrovascular complications have been performed. Several but not all studies have provided evidence to support the beneficial effects of incretin-based therapies on diabetic complications in patients with type 2 diabetes. We herein discuss the experimental and clinical evidence of incretin-based therapy for diabetic complications.
Assuntos
Complicações do Diabetes/tratamento farmacológico , Inibidores da Dipeptidil Peptidase IV/uso terapêutico , Receptor do Peptídeo Semelhante ao Glucagon 1/agonistas , Hipoglicemiantes/uso terapêutico , Incretinas/uso terapêutico , HumanosRESUMO
Amiodarone hydrochloride (AMD), an anti-arrhythmic agent, has been shown to cause peripheral neuropathy; however, its pathogenesis remains unknown. We examined the toxic effects of AMD on an immortalized adult rat Schwann cell line, IFRS1, and cocultures of IFRS1 cells and adult rat dorsal root ganglion neurons or nerve growth factor-primed PC12 cells. Treatment with AMD (1, 5, and 10 µm) induced time- and dose-dependent cell death, accumulation of phospholipids and neutral lipids, upregulation of the expression of gangliosides, and oxidative stress (increased nuclear factor E2-related factor in nuclear extracts and reduced GSH/GSSG ratios) in IFRS1 cells. It also induced the upregulation of LC3-II and p62 expression, with phosphorylation of p62, suggesting that deficient autolysosomal degradation is involved in AMD-induced IFRS1 cell death. Furthermore, treatment of the cocultures with AMD induced detachment of IFRS1 cells from neurite networks in a time- and dose-dependent manner. These findings suggest that AMD-induced lysosomal storage accompanied by enhanced oxidative stress and impaired lysosomal degradation in Schwann cells might be a cause of demyelination in the peripheral nervous system.