Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 87
Filtrar
1.
Genes Cells ; 29(3): 207-216, 2024 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-38163647

RESUMO

α-Synuclein (α-Syn)-positive intracellular fibrillar protein deposits, known as Lewy bodies, are thought to be involved in the pathogenesis of Parkinson's disease (PD). Although recent lines of evidence suggested that extracellular α-Syn secreted from pathogenic neurons contributes to the propagation of PD pathology, the precise mechanism of action remains unclear. We have reported that extracellular α-Syn caused sphingosine 1-phosphate (S1P) receptor type 1 (S1PR1) uncoupled from Gi and inhibited downstream G-protein signaling in SH-SY5Y cells, although its patho/physiological role remains to be clarified. Here we show that extracellular α-Syn caused S1P receptor type 3 (S1PR3) uncoupled from G protein in HeLa cells. Further studies indicated that α-Syn treatment reduced cathepsin D activity while enhancing the secretion of immature pro-cathepsin D into cell culture medium, suggesting that lysosomal delivery of cathepsin D was disturbed. Actually, extracellular α-Syn attenuated the retrograde trafficking of insulin-like growth factor-II/mannose 6-phosphate (IGF-II/M6P) receptor, which is under the regulation of S1PR3. These findings shed light on the understanding of dissemination of the PD pathology, that is, the mechanism underlying how extracellular α-Syn secreted from pathogenic cells causes lysosomal dysfunction of the neighboring healthy cells, leading to propagation of the disease.


Assuntos
Neuroblastoma , Doença de Parkinson , Humanos , alfa-Sinucleína/metabolismo , Catepsina D/metabolismo , Células HeLa , Lisossomos/metabolismo , Neuroblastoma/metabolismo , Doença de Parkinson/patologia , Receptores de Esfingosina-1-Fosfato/metabolismo
2.
Nephrol Dial Transplant ; 37(2): 262-270, 2022 01 25.
Artigo em Inglês | MEDLINE | ID: mdl-34586410

RESUMO

BACKGROUND: Although Lowe syndrome and Dent disease-2 are caused by Oculocerebrorenal syndrome of Lowe (OCRL) mutations, their clinical severities differ substantially and their molecular mechanisms remain unclear. Truncating mutations in OCRL exons 1-7 lead to Dent disease-2, whereas those in exons 8-24 lead to Lowe syndrome. Herein we identified the mechanism underlying the action of novel OCRL protein isoforms. METHODS: Messenger RNA samples extracted from cultured urine-derived cells from a healthy control and a Dent disease-2 patient were examined to detect the 5' end of the OCRL isoform. For protein expression and functional analysis, vectors containing the full-length OCRL transcripts, the isoform transcripts and transcripts with truncating mutations detected in Lowe syndrome and Dent disease-2 patients were transfected into HeLa cells. RESULTS: We successfully cloned the novel isoform transcripts from OCRL exons 6-24, including the translation-initiation codons present in exon 8. In vitro protein-expression analysis detected proteins of two different sizes (105 and 80 kDa) translated from full-length OCRL, whereas only one protein (80 kDa) was found from the isoform and Dent disease-2 variants. No protein expression was observed for the Lowe syndrome variants. The isoform enzyme activity was equivalent to that of full-length OCRL; the Dent disease-2 variants retained >50% enzyme activity, whereas the Lowe syndrome variants retained <20% activity. CONCLUSIONS: We elucidated the molecular mechanism underlying the two different phenotypes in OCRL-related diseases; the functional OCRL isoform translated starting at exon 8 was associated with this mechanism.


Assuntos
Doença de Dent , Síndrome Oculocerebrorrenal , Monoéster Fosfórico Hidrolases , Doença de Dent/diagnóstico , Doença de Dent/genética , Células HeLa , Humanos , Mutação/genética , Síndrome Oculocerebrorrenal/diagnóstico , Síndrome Oculocerebrorrenal/genética , Fenótipo , Monoéster Fosfórico Hidrolases/genética , Isoformas de Proteínas/genética
3.
iScience ; 24(11): 103351, 2021 Nov 19.
Artigo em Inglês | MEDLINE | ID: mdl-34805799

RESUMO

The importance of the G-protein ßγ subunits in the regulation of cargo transport from the trans-Golgi network (TGN) to the plasma membrane (PM) is well accepted; however, the molecular mechanism underlying the G-protein activation at the TGN remains unclear. We show here that sphingosine 1-phosphate (S1P) receptors at the PM were trafficked to the TGN in response to a surface transport cargo, temperature-sensitive vesicular stomatitis virus glycoprotein tagged with green fluorescent protein accumulation in the Golgi. The receptor internalization occurred in an S1P-independent manner but required phosphorylation by G-protein receptor kinase 2 and ß-arrestin association before internalization. Continuously activated S1P receptors in a manner dependent on S1P at the TGN kept transmitting G-protein signals including the ßγ subunits supply necessary for transport carrier formation at the TGN destined for the PM.

4.
Kobe J Med Sci ; 66(3): E94-E101, 2020 Nov 17.
Artigo em Inglês | MEDLINE | ID: mdl-33431782

RESUMO

Macropinocytosis is a highly conserved cellular process of endocytosis by which extracellular fluid and nutrients are taken up into cells through large, heterogeneous vesicles known as macropinosomes. Growth factors such as epidermal growth factor (EGF) can induce macropinocytosis in many types of cells, although precise mechanism underlying EGF-induced macropinocytosis remains unclear. In the present studies we have shown the involvement of S1P signaling in EGF-induced macropinocytosis in COS7 cells. First, EGF-induced macropinocytosis was strongly impaired in sphingosine kinase isozymes, SphK1 or SphK2-depleted cells, which was completely rescued by the expression of the corresponding wild-type isozyme but not the catalytically inactive one, suggesting the involvement of sphingosine 1-phosphate (S1P) in this phenomenon. Next, we observed that EGF-induced macropinocytosis was strongly inhibited in S1P type 1 receptor (S1P1R)-knockdown cells, implying involvement of S1P1R in this event. Furthermore, we could successfully demonstrate EGF-induced trans-activation of S1P1R using one-molecular fluorescence resonance energy transfer (FRET) technique. Moreover, for EGF-induced Rac1 activation, a step essential to F-actin formation and subsequent macropinocytosis, S1P signaling is required for its full activation, as judged by FRET analysis. These findings indicate that growth factors such as EGF utilize receptor-mediated S1P signaling for the regulation of macropinocytosis to fulfil vital cell activity.


Assuntos
Fator de Crescimento Epidérmico/metabolismo , Lisofosfolipídeos/metabolismo , Pinocitose/fisiologia , Receptores de Esfingosina-1-Fosfato/metabolismo , Esfingosina/análogos & derivados , Animais , Células COS , Chlorocebus aethiops , Transferência Ressonante de Energia de Fluorescência , Reação em Cadeia da Polimerase em Tempo Real , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Transdução de Sinais , Esfingosina/metabolismo
5.
Sci Signal ; 12(562)2019 01 01.
Artigo em Inglês | MEDLINE | ID: mdl-30600259

RESUMO

Atypical protein kinase C (aPKC) isozymes are unique in the PKC superfamily in that they are not regulated by the lipid second messenger diacylglycerol, which has led to speculation about whether a different second messenger acutely controls their function. Here, using a genetically encoded reporter that we designed, aPKC-specific C kinase activity reporter (aCKAR), we found that the lipid mediator sphingosine 1-phosphate (S1P) promoted the cellular activity of aPKC. Intracellular S1P directly bound to the purified kinase domain of aPKC and relieved autoinhibitory constraints, thereby activating the kinase. In silico studies identified potential binding sites on the kinase domain, one of which was validated biochemically. In HeLa cells, S1P-dependent activation of aPKC suppressed apoptosis. Together, our findings identify a previously undescribed molecular mechanism of aPKC regulation, a molecular target for S1P in cell survival regulation, and a tool to further explore the biochemical and biological functions of aPKC.


Assuntos
Proteínas Luminescentes/metabolismo , Lisofosfolipídeos/metabolismo , Proteína Quinase C/metabolismo , Transdução de Sinais , Esfingosina/análogos & derivados , Animais , Apoptose , Células COS , Linhagem Celular Tumoral , Chlorocebus aethiops , Ativação Enzimática , Células HeLa , Células Hep G2 , Humanos , Isoenzimas/genética , Isoenzimas/metabolismo , Proteínas Luminescentes/genética , Células MCF-7 , Microscopia de Fluorescência , Simulação de Acoplamento Molecular , Ligação Proteica , Proteína Quinase C/genética , Esfingosina/metabolismo
6.
Kobe J Med Sci ; 63(4): E123-E129, 2018 May 25.
Artigo em Inglês | MEDLINE | ID: mdl-29955024

RESUMO

Sphingosine 1-phosphate (S1P) is a bioactive phosphorylated product of sphingosine catalyzed by sphingosine kinase (SphK) and implicated in diverse cellular functions including vesicular trafficking. In the present study we have shown the importance of one of the subtypes of SphK, SphK2, in the regulation of cargo content in exosomes released from human myeloid leukemia K562 cells. First, SphK2 has been shown to localize with N-Rh-PE-positive late endosomes in the cells. Next, siRNA-mediated knockdown of Sphk2 but not SphK1 resulted in a reduction of cargo content in purified exosomes. The involvement of SphK2 in this phenomenon was further investigated by pharmacological approaches. When cells were treated with N,N-dimethylsphingosine (DMS), one of the most frequently used inhibitors for SphK, cargo contents in purified exosomes were enhanced unexpectedly. Finally, it has been shown that DMS has a potency to stimulate SphK2 activity depending on the substrate sphingosine- and the inhibitor-doses as estimated by in vitro assay systems using a purified SphK2. These findings suggest that SphK2/S1P signaling plays an important role in the regulation of cargo content in exosomes in K562 cells.


Assuntos
Exossomos/metabolismo , Fosfotransferases (Aceptor do Grupo Álcool)/metabolismo , Células HEK293 , Humanos , Células K562 , Lisofosfolipídeos/metabolismo , Corpos Multivesiculares/metabolismo , Esfingosina/análogos & derivados , Esfingosina/metabolismo
7.
J Biol Chem ; 293(21): 8208-8216, 2018 05 25.
Artigo em Inglês | MEDLINE | ID: mdl-29632069

RESUMO

α-Synuclein (α-Syn)-positive intracytoplasmic inclusions, known as Lewy bodies, are thought to be involved in the pathogenesis of Lewy body diseases, such as Parkinson's disease (PD). Although growing evidence suggests that cell-to-cell transmission of α-Syn is associated with the progression of PD and that extracellular α-Syn promotes formation of inclusion bodies, its precise mechanism of action in the extracellular space remains unclear. Here, as indicated by both conventional fractionation techniques and FRET-based protein-protein interaction analysis, we demonstrate that extracellular α-Syn causes expulsion of sphingosine 1-phosphate receptor subtype 1 (S1P1R) from the lipid raft fractions. S1P1R regulates vesicular trafficking, and its expulsion involved α-Syn binding to membrane-surface gangliosides. Consequently, the S1P1R became refractory to S1P stimulation required for activating inhibitory G-protein (Gi) in the plasma membranes. Moreover, the extracellular α-Syn also induced uncoupling of the S1P1R on internal vesicles, resulting in the reduced amount of CD63 molecule (CD63) in the lumen of multivesicular endosomes, together with a decrease in CD63 in the released exosomes from α-Syn-treated cells. Furthermore, cholesterol-depleting agent-induced S1P1R expulsion from the rafts also resulted in S1P1R uncoupling. Taken together, these results suggest that extracellular α-Syn-induced expulsion of S1P1R from lipid rafts promotes the uncoupling of S1P1R from Gi, thereby blocking subsequent Gi signals, such as inhibition of cargo sorting into exosomal vesicles in multivesicular endosomes. These findings help shed additional light on PD pathogenesis.


Assuntos
Subunidades alfa Gi-Go de Proteínas de Ligação ao GTP/metabolismo , Microdomínios da Membrana/metabolismo , Corpos Multivesiculares/metabolismo , Neuroblastoma/patologia , Receptores de Lisoesfingolipídeo/metabolismo , alfa-Sinucleína/metabolismo , Movimento Celular , Subunidades alfa Gi-Go de Proteínas de Ligação ao GTP/genética , Humanos , Neuroblastoma/genética , Neuroblastoma/metabolismo , Transporte Proteico , Receptores de Lisoesfingolipídeo/genética , Transdução de Sinais , Células Tumorais Cultivadas , alfa-Sinucleína/genética
10.
J Biol Chem ; 293(1): 245-253, 2018 01 05.
Artigo em Inglês | MEDLINE | ID: mdl-29133526

RESUMO

Exosomes play a critical role in cell-to-cell communication by delivering cargo molecules to recipient cells. However, the mechanism underlying the generation of the exosomal multivesicular endosome (MVE) is one of the mysteries in the field of endosome research. Although sphingolipid metabolites such as ceramide and sphingosine 1-phosphate (S1P) are known to play important roles in MVE formation and maturation, the detailed molecular mechanisms are still unclear. Here, we show that Rho family GTPases, including Cdc42 and Rac1, are constitutively activated on exosomal MVEs and are regulated by S1P signaling as measured by fluorescence resonance energy transfer (FRET)-based conformational changes. Moreover, we detected S1P signaling-induced filamentous actin (F-actin) formation. A selective inhibitor of Gßγ subunits, M119, strongly inhibited both F-actin formation on MVEs and cargo sorting into exosomal intralumenal vesicles of MVEs, both of which were fully rescued by the simultaneous expression of constitutively active Cdc42 and Rac1. Our results shed light on the mechanism underlying exosomal MVE maturation and inform the understanding of the physiological relevance of continuous activation of the S1P receptor and subsequent downstream G protein signaling to Gßγ subunits/Rho family GTPases-regulated F-actin formation on MVEs for cargo sorting into exosomal intralumenal vesicles.


Assuntos
Actinas/metabolismo , Subunidades alfa Gi-Go de Proteínas de Ligação ao GTP/metabolismo , Subunidades beta da Proteína de Ligação ao GTP/metabolismo , Subunidades gama da Proteína de Ligação ao GTP/metabolismo , Receptores de Lisoesfingolipídeo/metabolismo , Citoesqueleto de Actina/metabolismo , Movimento Celular/fisiologia , Endossomos/metabolismo , Exossomos/metabolismo , Transferência Ressonante de Energia de Fluorescência/métodos , Células HeLa , Humanos , Lisofosfolipídeos/metabolismo , Corpos Multivesiculares/metabolismo , Transporte Proteico , Transdução de Sinais , Esfingosina/análogos & derivados , Esfingosina/metabolismo , Proteína cdc42 de Ligação ao GTP/metabolismo , Proteínas rac1 de Ligação ao GTP/metabolismo , Proteínas rho de Ligação ao GTP/metabolismo
11.
Drug Metab Dispos ; 46(1): 11-19, 2018 01.
Artigo em Inglês | MEDLINE | ID: mdl-29051147

RESUMO

The influence of transporters on the pharmacokinetics of drugs is being increasingly recognized, and DDIs via transporters may be a risk factor for adverse events. Cyclosporine A, a strong OATP inhibitor, has been reported to increase the systemic exposure of rosuvastatin, an OATP substrate, by 7.1-fold in clinical studies. PXB mice are chimeric mice with humanized livers that are highly repopulated with human hepatocytes and have been widely used for drug discovery in drug metabolism and pharmacokinetics studies. In the present study, we examined in vivo and in vitro DDIs between rosuvastatin and cyclosporine A in PXB mice and fresh human hepatocytes (PXB cells) obtained from PXB mice. We initially investigated the active transport of rosuvastatin into PXB cells, and found concentration-dependent uptake with a Michaelis-Menten constant value of 4.0 µmol/l and a Vmax value of 4.63 pmol/min per 106 cells. Cyclosporine A inhibited the uptake of rosuvastatin with an IC50 value of 0.21 µmol/l. We then examined in vivo DDIs, and the exposure of orally administered rosuvastatin increased by 3.3-fold and 11-fold in PXB mice pretreated with 10 and 50 mg/kg cyclosporine A, whereas it increased by 2.5-fold and 6.2-fold when rosuvastatin was administered intravenously, in studies that were conducted for considering gastrointestinal DDIs. The liver-to-blood concentration ratio of rosuvastatin was dose-dependently decreased by pretreatment with cyclosporine A in PXB mice and SCID mice. Observed DDIs in vivo were considered to be reasonable based on the estimated concentrations of cyclosporine A at the inlet to the liver and in the liver tissues of both mice. In conclusion, our results indicate that PXB mice might be a useful tool for predicting human OATP-mediated DDIs in drug discovery, and its limitation due to the differences of gastrointestinal condition from human should also be considered.


Assuntos
Ciclosporina/farmacologia , Inibidores de Hidroximetilglutaril-CoA Redutases/farmacologia , Transportadores de Ânions Orgânicos/metabolismo , Rosuvastatina Cálcica/farmacologia , Quimeras de Transplante/metabolismo , Animais , Interações Medicamentosas , Células HEK293 , Hepatócitos/metabolismo , Hepatócitos/transplante , Humanos , Fígado/citologia , Fígado/metabolismo , Masculino , Camundongos , Camundongos SCID , Modelos Animais , Transportadores de Ânions Orgânicos/antagonistas & inibidores
12.
Sci Rep ; 7(1): 16552, 2017 11 29.
Artigo em Inglês | MEDLINE | ID: mdl-29185452

RESUMO

Sphingosine 1-phosphate (S1P) is a pleiotropic lipid mediator involved in the regulation of immune cell trafficking and vascular permeability acting mainly through G-protein-coupled S1P receptors (S1PRs). However, mechanism underlying how S1PRs are coupled with G-proteins remains unknown. Here we have uncovered that palmitoylation of a prototypical subtype S1P1R is prerequisite for subsequent inhibitory G-protein (Gi) coupling. We have identified DHHC5 as an enzyme for palmitoylation of S1P1R. Under basal conditions, S1P1R was functionally associated with DHHC5 in the plasma membranes (PM) and was fully palmitoylated, enabling Gi coupling. Upon stimulation, the receptor underwent internalisation leaving DHHC5 in PM, resulting in depalmitoylation of S1P1R. We also revealed that while physiological agonist S1P-induced endocytosed S1P1R readily recycled back to PM, pharmacological FTY720-P-induced endocytosed S1P1R-positive vesicles became associated with DHHC5 in the later phase, persistently transmitting Gi signals there. This indicates that FTY720-P switches off the S1P signal in PM, while switching on its signal continuously inside the cells. We propose that DHHC5-mediated palmitoylation of S1P1R determines Gi coupling and its signalling in a spatio/temporal manner.


Assuntos
Aciltransferases/metabolismo , Receptores de Lisoesfingolipídeo/metabolismo , Linhagem Celular , Membrana Celular/efeitos dos fármacos , Membrana Celular/metabolismo , Endocitose/efeitos dos fármacos , Humanos , Lipoilação , Lisofosfolipídeos/metabolismo , Organofosfatos/farmacologia , Transdução de Sinais/efeitos dos fármacos , Esfingosina/análogos & derivados , Esfingosina/metabolismo , Esfingosina/farmacologia
13.
Kobe J Med Sci ; 62(6): E162-E167, 2017 May 09.
Artigo em Inglês | MEDLINE | ID: mdl-28490713

RESUMO

α-Synuclein (α-Syn) is implicated in several neurodegenerative disorders, including Parkinson's disease, known collectively as the synucleinopathies. α-Syn is known to be secreted from the cells and may contribute to the progression of the disease. Although extracellular α-Syn is shown to impair platelet-derived growth factor-induced chemotaxis, molecular mechanism of α-Syn-induced motility failure remains elusive. Here we have aimed at phospholipase D (PLD) as a potential target for α-Syn and examined the involvement of this enzyme in α-Syn action. Indeed, extracellular α-Syn caused inhibition of agonist-induced PLD activation. However, inhibition of hydrolytic activity of PLD by 1-butanol treatment showed little or no effect on agonist-induced chemotaxis. These results suggest that some signaling pathways other than PLD may be involved in α-Syn-induced inhibition of chemotaxis.


Assuntos
Quimiotaxia , Fosfolipase D/metabolismo , alfa-Sinucleína/fisiologia , Animais , Células COS , Chlorocebus aethiops
14.
Sci Rep ; 7: 44248, 2017 03 16.
Artigo em Inglês | MEDLINE | ID: mdl-28300069

RESUMO

Parkinson's disease (PD) is the second most common neurodegenerative disorder. The presence of α-synuclein (α-Syn)-positive intracytoplasmic inclusions, known as Lewy bodies, is the cytopathological hallmark of PD. Increasing bodies of evidence suggest that cell-to-cell transmission of α-Syn plays a role in the progression of PD. Although extracellular α-Syn is known to cause abnormal cell motility, the precise mechanism remains elusive. Here we show that impairment of platelet-derived growth factor-induced cell motility caused by extracellular α-Syn is mainly attributed to selective inhibition of sphingosine 1-phosphate (S1P) signalling. Treatment of human neuroblastoma cells with recombinant α-Syn caused S1P type 1 (S1P1) receptor-selective uncoupling from inhibitory G-protein (Gi) as determined by both functional and fluorescence resonance energy transfer (FRET)-based structural analyses. By contrast, α-Syn caused little or no effect on S1P2 receptor-mediated signalling. Both wild-type and α-Syn(A53T), a mutant found in familiar PD, caused uncoupling of S1P1 receptor, although α-Syn(A53T) showed stronger potency in uncoupling. Moreover, S1P1 receptor-mediated ß-arrestin signal was unaltered by α-Syn(A53T). These results suggest that exogenous α-Syn modulates S1P1 receptor-mediated signalling from both Gi and ß-arrestin signals into ß-arrestin-biased signal. These findings uncovered a novel function of exogenous α-Syn in the cells.


Assuntos
Subunidades alfa Gi-Go de Proteínas de Ligação ao GTP/genética , Lisofosfolipídeos/metabolismo , Receptores de Lisoesfingolipídeo/genética , Proteínas Recombinantes/farmacologia , Esfingosina/análogos & derivados , alfa-Sinucleína/farmacologia , beta-Arrestinas/genética , Linhagem Celular Tumoral , Movimento Celular/efeitos dos fármacos , Transferência Ressonante de Energia de Fluorescência , Subunidades alfa Gi-Go de Proteínas de Ligação ao GTP/metabolismo , Regulação da Expressão Gênica , Humanos , Mutação , Neurônios/citologia , Neurônios/efeitos dos fármacos , Neurônios/metabolismo , Fator de Crescimento Derivado de Plaquetas/farmacologia , Receptores de Lisoesfingolipídeo/metabolismo , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Transdução de Sinais , Esfingosina/metabolismo , Receptores de Esfingosina-1-Fosfato , alfa-Sinucleína/genética , alfa-Sinucleína/metabolismo , beta-Arrestinas/metabolismo
15.
J Tissue Eng Regen Med ; 11(4): 977-988, 2017 04.
Artigo em Inglês | MEDLINE | ID: mdl-25689375

RESUMO

We developed a nude rat model for determining the capacity of trichogenous cells to restore in vivo-damaged hair follicles (HFs). A surgical scalpel was inserted into the rat's dermis to generate the in vivo-damaged pelage HFs, the HFs whose lower parts were lost, but the upper parts containing sebaceous and bulge regions remained intact. Dermal papilla cells (DPCs) and dermal sheath cells (DSCs) from EGFP transgenic rat vibrissae were propagated in culture, and each alone (DPC or DSC) or a mixture (DPC/DSC) was transplanted into the intradermal path made by a scalpel. It was found that the in vivo-damaged HFs had hair self-restoration ability, and the transplanted trichogenic dermal cells prominently enhanced this ability, DPC/DSC transplants being more effective in enhancement than DPC or DSC alone. The restored bulbs contained EGFP-positive cells, shed their original straight shafts, generated new shafts, and further developed into hairs with a sebaceous gland and bulge structures by ~6 weeks post-transplantation. Compared to the preceding animal models, this model is less invasive, requires fewer donor cells and allows repeated operations with higher reproducibility and accuracy. The present study suggests that conditions causing in situ-damaged HFs, such as androgenic alopecia, in which HFs are damaged and miniaturized, can be restored by functional trichogenous dermal cell transplantation therapy. Copyright © 2015 John Wiley & Sons, Ltd.


Assuntos
Transplante de Células , Derme/citologia , Derme/transplante , Cabelo/fisiologia , Regeneração/fisiologia , Animais , Biomarcadores/metabolismo , Feminino , Cabelo/crescimento & desenvolvimento , Folículo Piloso/patologia , Ratos Nus
16.
Sci Rep ; 6: 37810, 2016 11 25.
Artigo em Inglês | MEDLINE | ID: mdl-27886249

RESUMO

Parkinson's disease (PD) is characterized by α-synuclein (α-Syn)-positive intracytoplasmic inclusions, known as Lewy bodies. Although it is known that extracellular α-Syn is detected in the plasma and cerebrospinal fluid, its physiological significance remains unclear. Here, we show that extracellular α-Syn suppresses platelet-derived growth factor (PDGF)-induced chemotaxis in human neuroblastoma SH-SY5Y cells. The inhibitory effect was stronger in the mutant α-Syn(A53T), found in hereditary PD, and the degree of inhibition was time-dependent, presumably because of the oligomerization of α-Syn. PDGF-induced activation of Akt or Erk was not influenced by α-Syn(A53T). Further studies revealed that α-Syn(A53T) inhibited PDGF-induced Rac1 activation, whereas Cdc42 activation remained unaffected, resulting in unbalanced actin filament remodeling. These results shed light on the understanding of pathological as well as physiological functions of extracellular α-Syn in neuronal cells.


Assuntos
Quimiotaxia/fisiologia , Fator de Crescimento Derivado de Plaquetas/fisiologia , alfa-Sinucleína/fisiologia , Proteínas rac1 de Ligação ao GTP/antagonistas & inibidores , Linhagem Celular Tumoral , Humanos , Fator de Crescimento Derivado de Plaquetas/metabolismo , Transdução de Sinais
17.
Pediatr Nephrol ; 31(9): 1459-67, 2016 09.
Artigo em Inglês | MEDLINE | ID: mdl-27056061

RESUMO

BACKGROUND: Glomerulopathy with fibronectin deposits (GFND) is a rare autosomal dominant disease characterized by massive fibronectin deposits, leading to end-stage renal failure. Although mutations within the heparin-binding domains of the fibronectin 1 gene (FN1) have been associated with GFND, no mutations have been reported within the integrin-binding domains. METHODS: In this study, FN1 mutational analysis was conducted in 12 families with GFND. Biochemical and functional features of mutated proteins were examined using recombinant fibronectin fragments encompassing both the integrin- and heparin-binding domains. RESULTS: We report six FN1 mutations from 12 families with GFND, including five that are novel (p.Pro969Leu, p.Pro1472del, p.Trp1925Cys, p.Lys1953_Ile1961del, and p.Leu1974Pro). p.Pro1472del is localized in the integrin-binding domain of fibronectin, while the others are in heparin-binding domains. We detected p.Tyr973Cys, p.Pro1472del, and p.Leu1974Pro mutations in multiple families, and haplotype analysis implied that p.Pro1472del and p.Leu1974Pro are founder mutations. The protein encoded by the novel integrin-binding domain mutation p.Pro1472del showed decreased cell binding ability via the integrin-binding site. Most affected patients developed urine abnormalities during the first or second decade of life, and some mutation carriers were completely asymptomatic. CONCLUSIONS: This is the second large-scale analysis of GFND families and the first report of an integrin-binding domain mutation. These findings may help determine the pathogenesis of GFND.


Assuntos
Citocinas/genética , Glomerulonefrite Membranoproliferativa/genética , Mutação , Adolescente , Adulto , Idoso , Criança , Feminino , Fibronectinas , Glomerulonefrite Membranoproliferativa/complicações , Heparina , Humanos , Falência Renal Crônica/etiologia , Masculino , Pessoa de Meia-Idade
18.
Genes Cells ; 21(2): 185-99, 2016 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-26791143

RESUMO

Ultraviolet (UV) B is a major factor in melanomagenesis. This fact is linked to the resistance of melanocytes to UVB-induced apoptosis. In this study, we characterized the involvement of Mcl-1L in the regulation of UVB-induced apoptosis in melanocytes and in melanoma cells. In melanocytes, apoptosis was not evident at 24 h after UVB irradiation. The Mcl-1L expression increased after UVB irradiation, and the high Mcl-1L expression continued for at least 24 h. This UVB-dependent increase in Mcl-1L was mediated by the MEK-ERK-pS-STAT3 (STAT3 phosphorylated at Ser727) pathway. The Ser727 phosphorylation facilitated nuclear localization of STAT3. In melanoma cells, the expression levels of Mcl-1L varied depending on the cell line. WM39 melanoma cells expressed high levels of Mcl-1L via the MEK-ERK-pS-STAT3 pathway and were resistant to UVB-induced apoptosis without up-regulation of Mcl-1L. In melanocytes and in WM39 cells, transfection with Mcl-1 siRNA promoted UVB-induced apoptosis. Immunohistochemical studies showed that melanoma cells in in situ lesions expressed high amounts of Mcl-1L. These results indicate that the high expression of Mcl-1L mediated by the MEK-ERK-pS-STAT3 pathway protects melanocytes and melanoma cells from UVB-induced apoptosis.


Assuntos
Sistema de Sinalização das MAP Quinases , Melanócitos/citologia , Melanoma/metabolismo , Proteína de Sequência 1 de Leucemia de Células Mieloides/metabolismo , Raios Ultravioleta/efeitos adversos , Apoptose , Linhagem Celular Tumoral , Núcleo Celular/metabolismo , Regulação Neoplásica da Expressão Gênica/efeitos dos fármacos , Humanos , Melanócitos/metabolismo , Melanoma/genética , Proteína de Sequência 1 de Leucemia de Células Mieloides/genética , Metástase Neoplásica , Fosforilação/efeitos dos fármacos , Fator de Transcrição STAT3/metabolismo , Regulação para Cima
19.
Exp Dermatol ; 24(3): 225-7, 2015 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-25557326

RESUMO

No model is available for examining whether in vivo-damaged human hair follicles (hu-HFs) are rescued by transplanting cultured hu-HF dermal cells (dermal papilla and dermal sheath cells). Such a model might be valuable for examining whether in vivo-damaged hu-HFs such as miniaturized hu-HFs in androgenic alopecia are improvable by auto-transplanting hu-HF dermal cells. In this study, we first developed mice with humanized skin composed of hu-keratinocytes and hu-dermal fibroblasts. Then, a 'humanized scalp model mouse' was generated by transplanting hu-scalp HFs into the humanized skin. To demonstrate the usability of the model, the lower halves of the hu-HFs in the model were amputated in situ, and cultured hu-HF dermal cells were injected around the amputated area. The results demonstrated that the transplanted cells contributed to the restoration of the damaged HFs. This model could be used to explore clinically effective technologies for hair restoration therapy by autologous cell transplantation.


Assuntos
Modelos Animais de Doenças , Fibroblastos/transplante , Folículo Piloso/lesões , Folículo Piloso/fisiologia , Queratinócitos/transplante , Regeneração , Animais , Células Cultivadas , Cabelo/crescimento & desenvolvimento , Folículo Piloso/citologia , Folículo Piloso/transplante , Humanos , Camundongos , Couro Cabeludo , Fatores de Tempo
20.
Folia Neuropathol ; 52(1): 70-8, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-24729344

RESUMO

Sphingosine kinases (SphK 1&2) are involved in the regulation of cell survival, differentiation and neurotransmitter secretion. Current data suggest potential links between sphingolipid signalling, α-synuclein (ASN) and Alzheimer's disease (AD). Our aim was to investigate the possible role of SphKs and ASN in the regulation of the production and secretion of the amyloid ß precursor protein (APP). We have previously shown that ASN intensified the secretion and toxicity of amyloid ß (Aß) to the point where it caused cell death. Our current results show that APP, the precursor protein for Aß, is also influenced by ASN. The stable overexpression of wtASN in SH-SY5Y cells caused a three-fold, significant increase of the cellular APP level. This suggests that the influence of ASN on Aß metabolism may actually occur at the level of APP protein rather than only through the changes of its cleavage into Aß. To elucidate the mechanisms of APP modulation the cells were exposed to S1P and an SphK inhibitor (SKI). 72 h S1P treatment at 5 µM caused a nearly 50% reduction of the cellular APP signal. S1P also caused a tendency towards higher APP secretion, though the results were insignificant. The inhibition of SphKs decreased medium APP levels in a dose-dependent manner, reaching significance at 5 µM SKI with a correspondingly elevated intracellular level. Thus, it is reasonable to expect that in fact the influence of SphK activity on APP might be pro-secretory. This would also be in agreement with numerous articles on SphK-dependent secretion in the literature. The chronic nature of AD further suggests that subtle alterations in APP metabolism could have the potential to drive important changes in brain condition.


Assuntos
Precursor de Proteína beta-Amiloide/metabolismo , Lisofosfolipídeos/metabolismo , Neurônios/enzimologia , Neurônios/metabolismo , Esfingosina/análogos & derivados , alfa-Sinucleína/metabolismo , Doença de Alzheimer/metabolismo , Western Blotting , Linhagem Celular Tumoral , Humanos , Reação em Cadeia da Polimerase , Esfingosina/metabolismo , Transfecção
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...