Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 13 de 13
Filtrar
1.
EMBO Mol Med ; 16(3): 616-640, 2024 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-38383802

RESUMO

Haplo-insufficiency of the gene encoding the myelin protein PMP22 leads to focal myelin overgrowth in the peripheral nervous system and hereditary neuropathy with liability to pressure palsies (HNPP). Conversely, duplication of PMP22 causes Charcot-Marie-Tooth disease type 1A (CMT1A), characterized by hypomyelination of medium to large caliber axons. The molecular mechanisms of abnormal myelin growth regulation by PMP22 have remained obscure. Here, we show in rodent models of HNPP and CMT1A that the PI3K/Akt/mTOR-pathway inhibiting phosphatase PTEN is correlated in abundance with PMP22 in peripheral nerves, without evidence for direct protein interactions. Indeed, treating DRG neuron/Schwann cell co-cultures from HNPP mice with PI3K/Akt/mTOR pathway inhibitors reduced focal hypermyelination. When we treated HNPP mice in vivo with the mTOR inhibitor Rapamycin, motor functions were improved, compound muscle amplitudes were increased and pathological tomacula in sciatic nerves were reduced. In contrast, we found Schwann cell dedifferentiation in CMT1A uncoupled from PI3K/Akt/mTOR, leaving partial PTEN ablation insufficient for disease amelioration. For HNPP, the development of PI3K/Akt/mTOR pathway inhibitors may be considered as the first treatment option for pressure palsies.


Assuntos
Artrogripose , Doença de Charcot-Marie-Tooth , Neuropatia Hereditária Motora e Sensorial , Fosfatidilinositol 3-Quinases , Camundongos , Animais , Proteínas Proto-Oncogênicas c-akt , Roedores/metabolismo , Doença de Charcot-Marie-Tooth/genética , Doença de Charcot-Marie-Tooth/patologia , Proteínas da Mielina/genética , Proteínas da Mielina/metabolismo , Serina-Treonina Quinases TOR
2.
Cell Metab ; 35(12): 2136-2152.e9, 2023 Dec 05.
Artigo em Inglês | MEDLINE | ID: mdl-37989315

RESUMO

The peripheral nervous system harbors a remarkable potential to regenerate after acute nerve trauma. Full functional recovery, however, is rare and critically depends on peripheral nerve Schwann cells that orchestrate breakdown and resynthesis of myelin and, at the same time, support axonal regrowth. How Schwann cells meet the high metabolic demand required for nerve repair remains poorly understood. We here report that nerve injury induces adipocyte to glial signaling and identify the adipokine leptin as an upstream regulator of glial metabolic adaptation in regeneration. Signal integration by leptin receptors in Schwann cells ensures efficient peripheral nerve repair by adjusting injury-specific catabolic processes in regenerating nerves, including myelin autophagy and mitochondrial respiration. Our findings propose a model according to which acute nerve injury triggers a therapeutically targetable intercellular crosstalk that modulates glial metabolism to provide sufficient energy for successful nerve repair.


Assuntos
Bainha de Mielina , Nervos Periféricos , Bainha de Mielina/metabolismo , Neuroglia , Células de Schwann/metabolismo , Regeneração Nervosa/fisiologia
3.
Nat Neurosci ; 24(1): 47-60, 2021 01.
Artigo em Inglês | MEDLINE | ID: mdl-33349711

RESUMO

The repair of inflamed, demyelinated lesions as in multiple sclerosis (MS) necessitates the clearance of cholesterol-rich myelin debris by microglia/macrophages and the switch from a pro-inflammatory to an anti-inflammatory lesion environment. Subsequently, oligodendrocytes increase cholesterol levels as a prerequisite for synthesizing new myelin membranes. We hypothesized that lesion resolution is regulated by the fate of cholesterol from damaged myelin and oligodendroglial sterol synthesis. By integrating gene expression profiling, genetics and comprehensive phenotyping, we found that, paradoxically, sterol synthesis in myelin-phagocytosing microglia/macrophages determines the repair of acutely demyelinated lesions. Rather than producing cholesterol, microglia/macrophages synthesized desmosterol, the immediate cholesterol precursor. Desmosterol activated liver X receptor (LXR) signaling to resolve inflammation, creating a permissive environment for oligodendrocyte differentiation. Moreover, LXR target gene products facilitated the efflux of lipid and cholesterol from lipid-laden microglia/macrophages to support remyelination by oligodendrocytes. Consequently, pharmacological stimulation of sterol synthesis boosted the repair of demyelinated lesions, suggesting novel therapeutic strategies for myelin repair in MS.


Assuntos
Doenças Desmielinizantes/patologia , Microglia/fisiologia , Esteróis/biossíntese , Animais , Colesterol/metabolismo , Desmosterol/metabolismo , Encefalomielite Autoimune Experimental , Feminino , Perfilação da Expressão Gênica , Humanos , Inflamação/metabolismo , Inflamação/patologia , Metabolismo dos Lipídeos , Receptores X do Fígado/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Pessoa de Meia-Idade , Esclerose Múltipla , Oligodendroglia/metabolismo , Fagocitose , Esqualeno/metabolismo
4.
J Neurosci Res ; 98(10): 1933-1952, 2020 10.
Artigo em Inglês | MEDLINE | ID: mdl-32588471

RESUMO

Charcot-Marie-Tooth disease 1 A (CMT1A) is caused by an intrachromosomal duplication of the gene encoding for PMP22 leading to peripheral nerve dysmyelination, axonal loss, and progressive muscle weakness. No therapy is available. PXT3003 is a low-dose combination of baclofen, naltrexone, and sorbitol which has been shown to improve disease symptoms in Pmp22 transgenic rats, a bona fide model of CMT1A disease. However, the superiority of PXT3003 over its single components or dual combinations have not been tested. Here, we show that in a dorsal root ganglion (DRG) co-culture system derived from transgenic rats, PXT3003 induced myelination when compared to its single and dual components. Applying a clinically relevant ("translational") study design in adult male CMT1A rats for 3 months, PXT3003, but not its dual components, resulted in improved performance in behavioral motor and sensory endpoints when compared to placebo. Unexpectedly, we observed only a marginally increased number of myelinated axons in nerves from PXT3003-treated CMT1A rats. However, in electrophysiology, motor latencies correlated with increased grip strength indicating a possible effect of PXT3003 on neuromuscular junctions (NMJs) and muscle fiber pathology. Indeed, PXT3003-treated CMT1A rats displayed an increased perimeter of individual NMJs and a larger number of functional NMJs. Moreover, muscles of PXT3003 CMT1A rats displayed less neurogenic atrophy and a shift toward fast contracting muscle fibers. We suggest that ameliorated motor function in PXT3003-treated CMT1A rats result from restored NMJ function and muscle innervation, independent from myelination.


Assuntos
Baclofeno/administração & dosagem , Doença de Charcot-Marie-Tooth/tratamento farmacológico , Doenças Desmielinizantes/tratamento farmacológico , Naltrexona/administração & dosagem , Junção Neuromuscular/efeitos dos fármacos , Sorbitol/administração & dosagem , Animais , Doença de Charcot-Marie-Tooth/genética , Doença de Charcot-Marie-Tooth/fisiopatologia , Técnicas de Cocultura , Doenças Desmielinizantes/genética , Doenças Desmielinizantes/fisiopatologia , Sinergismo Farmacológico , Quimioterapia Combinada , Feminino , Masculino , Proteínas da Mielina/genética , Condução Nervosa/efeitos dos fármacos , Condução Nervosa/fisiologia , Junção Neuromuscular/fisiologia , Ratos , Ratos Sprague-Dawley , Ratos Transgênicos
5.
EMBO J ; 38(19): e101468, 2019 10 01.
Artigo em Inglês | MEDLINE | ID: mdl-31506973

RESUMO

Excitatory amino acid transporters (EAATs) mediate glial and neuronal glutamate uptake to terminate synaptic transmission and to ensure low resting glutamate concentrations. Effective glutamate uptake is achieved by cotransport with 3 Na+ and 1 H+ , in exchange with 1 K+ . The underlying principles of this complex transport stoichiometry remain poorly understood. We use molecular dynamics simulations and electrophysiological experiments to elucidate how mammalian EAATs harness K+ gradients, unlike their K+ -independent prokaryotic homologues. Glutamate transport is achieved via elevator-like translocation of the transport domain. In EAATs, glutamate-free re-translocation is prevented by an external gate remaining open until K+  binding closes and locks the gate. Prokaryotic GltPh contains the same K+ -binding site, but the gate can close without K+ . Our study provides a comprehensive description of K+ -dependent glutamate transport and reveals a hitherto unknown allosteric coupling mechanism that permits adaptions of the transport stoichiometry without affecting ion or substrate binding.


Assuntos
Proteínas de Transporte de Glutamato da Membrana Plasmática/química , Proteínas de Transporte de Glutamato da Membrana Plasmática/metabolismo , Potássio/metabolismo , Regulação Alostérica , Transporte Biológico , Células HEK293 , Humanos , Modelos Moleculares , Simulação de Dinâmica Molecular , Conformação Proteica , Transmissão Sináptica
6.
PLoS One ; 14(1): e0209752, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-30650121

RESUMO

The most common type of Charcot-Marie-Tooth disease is caused by a duplication of PMP22 leading to dysmyelination, axonal loss and progressive muscle weakness (CMT1A). Currently, no approved therapy is available for CMT1A patients. A novel polytherapeutic proof-of-principle approach using PXT3003, a low-dose combination of baclofen, naltrexone and sorbitol, slowed disease progression after long-term dosing in adult Pmp22 transgenic rats, a known animal model of CMT1A. Here, we report an early postnatal, short-term treatment with PXT3003 in CMT1A rats that delays disease onset into adulthood. CMT1A rats were treated from postnatal day 6 to 18 with PXT3003. Behavioural, electrophysiological, histological and molecular analyses were performed until 12 weeks of age. Daily oral treatment for approximately 2 weeks ameliorated motor deficits of CMT1A rats reaching wildtype levels. Histologically, PXT3003 corrected the disturbed axon calibre distribution with a shift towards large motor axons. Despite dramatic clinical amelioration, only distal motor latencies were improved and correlated with phenotype performance. On the molecular level, PXT3003 reduced Pmp22 mRNA overexpression and improved the misbalanced downstream PI3K-AKT / MEK-ERK signalling pathway. The improved differentiation status of Schwann cells may have enabled better long-term axonal support function. We conclude that short-term treatment with PXT3003 during early development may partially prevent the clinical and molecular manifestations of CMT1A. Since PXT3003 has a strong safety profile and is currently undergoing a phase III trial in CMT1A patients, our results suggest that PXT3003 therapy may be a bona fide translatable therapy option for children and young adolescent patients suffering from CMT1A.


Assuntos
Baclofeno/farmacologia , Doença de Charcot-Marie-Tooth/tratamento farmacológico , Naltrexona/farmacologia , Sorbitol/farmacologia , Animais , Axônios/metabolismo , Doença de Charcot-Marie-Tooth/genética , Doença de Charcot-Marie-Tooth/metabolismo , Doenças Desmielinizantes/patologia , Modelos Animais de Doenças , Combinação de Medicamentos , MAP Quinase Quinase Quinases/metabolismo , Sistema de Sinalização das MAP Quinases/efeitos dos fármacos , Masculino , Debilidade Muscular/metabolismo , Proteínas da Mielina/efeitos dos fármacos , Proteínas da Mielina/genética , Proteínas da Mielina/metabolismo , Condução Nervosa , Fosfatidilinositol 3-Quinases/metabolismo , Estudo de Prova de Conceito , Proteínas Proto-Oncogênicas c-akt/metabolismo , Ratos , Ratos Sprague-Dawley , Ratos Transgênicos , Células de Schwann/efeitos dos fármacos , Transdução de Sinais/efeitos dos fármacos
7.
J Biol Chem ; 290(48): 28988-96, 2015 Nov 27.
Artigo em Inglês | MEDLINE | ID: mdl-26475859

RESUMO

Crystal structures of the archaeal homologue GltPh have provided important insights into the molecular mechanism of transport of the excitatory neurotransmitter glutamate. Whereas mammalian glutamate transporters can translocate both glutamate and aspartate, GltPh is only one capable of aspartate transport. Most of the amino acid residues that surround the aspartate substrate in the binding pocket of GltPh are highly conserved. However, in the brain transporters, Thr-352 and Met-362 of the reentrant hairpin loop 2 are replaced by the smaller Ala and Thr, respectively. Therefore, we have studied the effects of T352A and M362T on binding and transport of aspartate and glutamate by GltPh. Substrate-dependent intrinsic fluorescence changes were monitored in transporter constructs containing the L130W mutation. GltPh-L130W/T352A exhibited an ~15-fold higher apparent affinity for l-glutamate than the wild type transporter, and the M362T mutation resulted in an increased affinity of ~40-fold. An even larger increase of the apparent affinity for l-glutamate, around 130-fold higher than that of wild type, was observed with the T352A/M362T double mutant. Radioactive uptake experiments show that GltPh-T352A not only transports aspartate but also l-glutamate. Remarkably, GltPh-M362T exhibited l-aspartate but not l-glutamate transport. The double mutant retained the ability to transport l-glutamate, but its kinetic parameters were very similar to those of GltPh-T352A alone. The differential impact of mutation on binding and transport of glutamate suggests that hairpin loop 2 not only plays a role in the selection of the substrate but also in its translocation.


Assuntos
Ácido Aspártico/química , Proteínas de Transporte de Glutamato da Membrana Plasmática/química , Ácido Glutâmico/química , Mutação de Sentido Incorreto , Proteínas do Tecido Nervoso/química , Substituição de Aminoácidos , Ácido Aspártico/genética , Ácido Aspártico/metabolismo , Encéfalo/metabolismo , Proteínas de Transporte de Glutamato da Membrana Plasmática/genética , Proteínas de Transporte de Glutamato da Membrana Plasmática/metabolismo , Ácido Glutâmico/genética , Ácido Glutâmico/metabolismo , Humanos , Transporte de Íons/genética , Proteínas do Tecido Nervoso/genética , Proteínas do Tecido Nervoso/metabolismo , Estrutura Secundária de Proteína , Especificidade por Substrato/genética
8.
Cell ; 160(3): 542-53, 2015 Jan 29.
Artigo em Inglês | MEDLINE | ID: mdl-25635461

RESUMO

Excitatory amino acid transporters (EAATs) are essential for terminating glutamatergic synaptic transmission. They are not only coupled glutamate/Na(+)/H(+)/K(+) transporters but also function as anion-selective channels. EAAT anion channels regulate neuronal excitability, and gain-of-function mutations in these proteins result in ataxia and epilepsy. We have combined molecular dynamics simulations with fluorescence spectroscopy of the prokaryotic homolog GltPh and patch-clamp recordings of mammalian EAATs to determine how these transporters conduct anions. Whereas outward- and inward-facing GltPh conformations are nonconductive, lateral movement of the glutamate transport domain from intermediate transporter conformations results in formation of an anion-selective conduction pathway. Fluorescence quenching of inserted tryptophan residues indicated the entry of anions into this pathway, and mutations of homologous pore-forming residues had analogous effects on GltPh simulations and EAAT2/EAAT4 measurements of single-channel currents and anion/cation selectivities. These findings provide a mechanistic framework of how neurotransmitter transporters can operate as anion-selective and ligand-gated ion channels.


Assuntos
Sistema X-AG de Transporte de Aminoácidos/química , Ânions/metabolismo , Proteínas Arqueais/química , Proteínas de Transporte de Glutamato da Membrana Plasmática/química , Simulação de Dinâmica Molecular , Pyrococcus horikoshii/química , Sequência de Aminoácidos , Sistema X-AG de Transporte de Aminoácidos/metabolismo , Animais , Proteínas Arqueais/metabolismo , Proteínas de Transporte de Glutamato da Membrana Plasmática/genética , Proteínas de Transporte de Glutamato da Membrana Plasmática/metabolismo , Humanos , Dados de Sequência Molecular , Mutação , Técnicas de Patch-Clamp , Ratos , Alinhamento de Sequência
9.
J Biol Chem ; 289(15): 10387-10398, 2014 Apr 11.
Artigo em Inglês | MEDLINE | ID: mdl-24519939

RESUMO

The ß-subunit associates with the α1 pore-forming subunit of high voltage-activated calcium channels and modulates several aspects of ion conduction. Four ß-subunits are encoded by four different genes with multiple splice variants. Only two members of this family, ß2a and ß2e, associate with the plasma membrane in the absence of the α1-subunit. Palmitoylation on a di-cysteine motif located at the N terminus of ß2a promotes membrane targeting and correlates with the unique ability of this protein to slow down inactivation. In contrast, the mechanism by which ß2e anchors to the plasma membrane remains elusive. Here, we identified an N-terminal segment in ß2e encompassing a cluster of positively charged residues, which is strictly required for membrane anchoring, and when transferred to the cytoplasmic ß1b isoform it confers membrane localization to the latter. In the presence of negatively charged phospholipid vesicles, this segment binds to acidic liposomes dependently on the ionic strength, and the intrinsic fluorescence emission maxima of its single tryptophan blue shifts considerably. Simultaneous substitution of more than two basic residues impairs membrane targeting. Coexpression of the fast inactivating R-type calcium channels with wild-type ß2e, but not with a ß2e membrane association-deficient mutant, slows down inactivation. We propose that a predicted α-helix within this domain orienting parallel to the membrane tethers the ß2e-subunit to the lipid bilayer via electrostatic interactions. Penetration of the tryptophan side chain into the lipidic core stabilizes the membrane-bound conformation. This constitutes a new mechanism for membrane anchoring among the ß-subunit family that also sustains slowed inactivation.


Assuntos
Canais de Cálcio Tipo L/química , Canais de Cálcio Tipo L/metabolismo , Membrana Celular/química , Lipídeos/química , Sequência de Aminoácidos , Animais , Eletrofisiologia , Lipossomos/química , Microscopia Confocal , Dados de Sequência Molecular , Fenótipo , Mutação Puntual , Ligação Proteica , Conformação Proteica , Estrutura Terciária de Proteína , Ratos , Homologia de Sequência de Aminoácidos , Eletricidade Estática , Triptofano/química
10.
Proc Natl Acad Sci U S A ; 110(30): 12486-91, 2013 Jul 23.
Artigo em Inglês | MEDLINE | ID: mdl-23840066

RESUMO

Excitatory amino acid transporters (EAATs) are a class of glutamate transporters that terminate glutamatergic synaptic transmission in the mammalian CNS. GltPh, an archeal EAAT homolog from Pyrococcus horikoshii, is currently the only member with a known 3D structure. Here, we studied the kinetics of substrate binding of a single tryptophan mutant (L130W) GltPh in detergent micelles. At low millimolar [Na(+)], the addition of L-aspartate resulted in complex time courses of W130 fluorescence changes over tens of seconds. With increasing [Na(+)], the kinetics were dominated by a fast component [k(obs,fast); K(D) (Na(+)) = 22 ± 3 mM, n(Hill )= 1.7 ± 0.3] with values of k(obs,fast) rising in a saturable manner to ≈ 500 s(-1) (at 6 °C) with increasing [L-aspartate]. The binding kinetics of L-aspartate differed from the binding kinetics of two alternative substrates: L-cysteine sulfinic acid and d-aspartate. L-cysteine sulfinic acid bound with higher affinity than L-aspartate but involved lower saturating rates, whereas the saturating rates after D-aspartate binding were higher. Thus, after the association of two Na(+) to the empty transporter, GltPh binds amino acids by induced fit. Cross-linking and proteolysis experiments suggest that the induced fit results from the closure of helical hairpin 2. This conformational change is faster for GltPh than for most mammalian homologues, whereas the amino acid association rates are similar. Our data reveal the importance of induced fit for substrate selection in EAATs and illustrate how high-affinity binding and the efficient transport of glutamate can be accomplished simultaneously by this class of transporters.


Assuntos
Sistema X-AG de Transporte de Aminoácidos/metabolismo , Archaea/metabolismo , Sistema X-AG de Transporte de Aminoácidos/química , Isomerismo , Modelos Moleculares , Espectrometria de Fluorescência , Especificidade por Substrato
11.
Biochim Biophys Acta ; 1813(3): 377-89, 2011 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-21215280

RESUMO

Adaptations in the oxidative capacity of skeletal muscle cells can occur under several physiological or pathological conditions. We investigated the effect of increasing extracellular glucose concentration on the expression of markers of energy metabolism in primary skeletal muscle cells and the C2C12 muscle cell line. Growth of myotubes in 25mM glucose (high glucose, HG) compared with 5.55mM led to increases in the expression and activity of glyceraldehyde-3-phosphate dehydrogenase (GAPDH), a marker of glycolytic energy metabolism, while oxidative markers peroxisome proliferator-activated receptor γ coactivator 1α and citrate synthase decreased. HG induced metabolic adaptations as are seen during a slow-to-fast fiber transformation. Furthermore, HG increased fast myosin heavy chain (MHC) IId/x but did not change slow MHCI/ß expression. Protein phosphatase 2A (PP2A) was shown to mediate the effects of HG on GAPDH and MHCIId/x. Carbohydrate response element-binding protein (ChREBP), a glucose-dependent transcription factor downstream of PP2A, partially mediated the effects of glucose on metabolic markers. The glucose-induced increase in PP2A activity was associated with an increase in p38 mitogen-activated protein kinase activity, which presumably mediates the increase in MHCIId/x promoter activity. Liver X receptor, another possible mediator of glucose effects, induced only an incomplete metabolic shift, mainly increasing the expression of the glycolytic marker. Taken together, HG induces a partial slow-to-fast transformation comprising metabolic enzymes together with an increased expression of MHCIId/x. This work demonstrates a functional role for ChREBP in determining the metabolic type of muscle fibers and highlights the importance of glucose as a signaling molecule in muscle.


Assuntos
Metabolismo Energético , Regulação da Expressão Gênica , Glucose/metabolismo , Fibras Musculares Esqueléticas/metabolismo , Proteínas Nucleares/metabolismo , Fatores de Transcrição/metabolismo , Animais , Fatores de Transcrição de Zíper de Leucina e Hélice-Alça-Hélix Básicos , Linhagem Celular , Células Cultivadas , Gliceraldeído-3-Fosfato Desidrogenases/genética , Receptores X do Fígado , Camundongos , Cadeias Pesadas de Miosina/genética , Receptores Nucleares Órfãos/metabolismo , Regiões Promotoras Genéticas , Proteína Fosfatase 2/metabolismo , Coelhos
12.
J Biol Chem ; 285(31): 23676-86, 2010 Jul 30.
Artigo em Inglês | MEDLINE | ID: mdl-20519505

RESUMO

Excitatory amino acid transporter (EAAT) glutamate transporters function not only as secondary active glutamate transporters but also as anion channels. Recently, a conserved aspartic acid (Asp(112)) within the intracellular loop near to the end of transmembrane domain 2 was proposed as a major determinant of substrate-dependent gating of the anion channel associated with the glial glutamate transporter EAAT1. We studied the corresponding mutation (D117A) in another EAAT isoform, EAAT4, using heterologous expression in mammalian cells, whole cell patch clamp, and noise analysis. In EAAT4, D117A modifies unitary conductances, relative anion permeabilities, as well as gating of associated anion channels. EAAT4 anion channel gating is characterized by two voltage-dependent gating processes with inverse voltage dependence. In wild type EAAT4, external l-glutamate modifies the voltage dependence as well as the minimum open probabilities of both gates, resulting in concentration-dependent changes of the number of open channels. Not only transport substrates but also anions affect wild type EAAT4 channel gating. External anions increase the open probability and slow down relaxation constants of one gating process that is activated by depolarization. D117A abolishes the anion and glutamate dependence of EAAT4 anion currents and shifts the voltage dependence of EAAT4 anion channel activation by more than 200 mV to more positive potentials. D117A is the first reported mutation that changes the unitary conductance of an EAAT anion channel. The finding that mutating a pore-forming residue modifies gating illustrates the close linkage between pore conformation and voltage- and substrate-dependent gating in EAAT4 anion channels.


Assuntos
Ácido Aspártico/química , Transportador 4 de Aminoácido Excitatório/química , Ânions/química , Biofísica/métodos , Membrana Celular/metabolismo , Relação Dose-Resposta a Droga , Eletrofisiologia/métodos , Ácido Glutâmico/química , Humanos , Cinética , Neurotransmissores/metabolismo , Técnicas de Patch-Clamp , Permeabilidade , Isoformas de Proteínas , Estrutura Terciária de Proteína
13.
Biophys J ; 91(4): 1471-9, 2006 Aug 15.
Artigo em Inglês | MEDLINE | ID: mdl-16731558

RESUMO

An opsin-encoding cDNA was cloned from the marine alga Acetabularia acetabulum. The cDNA was expressed in Xenopus oocytes into functional Acetabularia rhodopsin (AR) mediating H+ carried outward photocurrents of up to 1.2 microA with an action spectrum maximum at 518 nm (AR518). AR is the first ion-pumping rhodopsin found in a plant organism. Steady-state photocurrents of AR are always positive and rise sigmoidally from negative to positive transmembrane voltages. Numerous kinetic details (amplitudes and time constants), including voltage-dependent recovery of the dark state after light-off, are documented with respect to their sensitivities to light, internal and external pH, and the transmembrane voltage. The results are analyzed by enzyme kinetic formalisms using a simplified version of the known photocycle of bacteriorhodopsin (BR). Blue-light causes a shunt of the photocycle under H+ reuptake from the extracellular side. Similarities and differences of AR with BR are pointed out. This detailed electrophysiological characterization highlights voltage dependencies in catalytic membrane processes of this eukaryotic, H+ -pumping rhodopsin and of microbial-type rhodopsins in general.


Assuntos
Acetabularia/fisiologia , Potenciais da Membrana/fisiologia , Bombas de Próton/fisiologia , Rodopsina/fisiologia , Acetabularia/efeitos da radiação , Relação Dose-Resposta à Radiação , Luz , Biologia Marinha , Potenciais da Membrana/efeitos da radiação , Bombas de Próton/efeitos da radiação , Doses de Radiação , Rodopsina/efeitos da radiação , Água do Mar/microbiologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA