Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 7 de 7
Filtrar
Mais filtros

Base de dados
Tipo de documento
Intervalo de ano de publicação
1.
ACS Chem Neurosci ; 10(1): 599-609, 2019 01 16.
Artigo em Inglês | MEDLINE | ID: mdl-30272946

RESUMO

Manganese (Mn) is an essential metal that can be neurotoxic when elevated exposition occurs leading to parkinsonian-like syndromes. Mutations in the Slc30a10 gene have been identified in new forms of familial parkinsonism. SLC30A10 is a cell surface protein involved in the efflux of Mn and protects the cell against Mn toxicity. Disease-causing mutations block the efflux activity of SLC30A10, resulting in Mn accumulation. Determining the intracellular localization of Mn when disease-causing SLC30A10 mutants are expressed is essential to elucidate the mechanisms of Mn neurotoxicity. Here, using organelle fluorescence microscopy and synchrotron X-ray fluorescence (SXRF) imaging, we found that Mn accumulates in the Golgi apparatus of human cells transfected with the disease-causing SLC30A10-Δ105-107 mutant under physiological conditions and after exposure to Mn. In cells expressing the wild-type SLC30A10 protein, cellular Mn content was low after all exposure conditions, confirming efficient Mn efflux. In nontransfected cells that do not express endogenous SLC30A10 and in mock transfected cells, Mn was located in the Golgi apparatus, similarly to its distribution in cells expressing the mutant protein, confirming deficient Mn efflux. The newly developed SXRF cryogenic nanoimaging (<50 nm resolution) indicated that Mn was trapped in single vesicles within the Golgi apparatus. Our results confirm the role of SLC30A10 in Mn efflux and the accumulation of Mn in cells expressing the disease-causing SLC30A10-Δ105-107 mutation. Moreover, we identified suborganelle Golgi nanovesicles as the main compartment of Mn accumulation in SLC30A10 mutants, suggesting interactions with the vesicular trafficking machinery as a cause of the disease.


Assuntos
Proteínas de Transporte de Cátions/genética , Complexo de Golgi/metabolismo , Mutação/genética , Transtornos Parkinsonianos/genética , Proteínas de Transporte de Cátions/metabolismo , Complexo de Golgi/genética , Humanos , Manganês/metabolismo , Síndromes Neurotóxicas/genética , Síndromes Neurotóxicas/metabolismo , Transtornos Parkinsonianos/metabolismo , Transporte Proteico/genética , Transporte Proteico/fisiologia
2.
Metallomics ; 10(8): 1053-1064, 2018 08 15.
Artigo em Inglês | MEDLINE | ID: mdl-29989630

RESUMO

SLC30 proteins belong to the cation diffusion facilitator (CDF) superfamily of metal transporters. SLC30A10 mediates manganese efflux, while other SLC30 members transport zinc. Metal specificity of CDFs may be conferred by amino acids that form a transmembrane metal binding site (Site A). Site A of zinc-transporting CDFs, such as SLC30A1/ZnT1, have a HXXXD motif, but manganese transporters, such as SLC30A10, harbor a NXXXD motif. This critical histidine-to-asparagine substitution, at residue 43, was proposed to underlie manganese transport specificity of SLC30A10. However, we recently discovered that asparagine-43 was dispensable for manganese efflux in HeLa cells; instead, glutamate-25, aspartate-40, asparagine-127, and aspartate-248 were required. In contrast, another group reported that asparagine-43 was required in a chicken cell line. The goal of this study was to resolve the divergent results about the requirement of the crucial asparagine-43 residue. For this, we compared the manganese efflux activity of four cell types that stably over-expressed SLC30A10wild-type (WT), SLC30A10N43A or SLC30A10E25A: physiologically-relevant hepatic HepG2 and neuronal AF5 cells, HEK cells, and embryonic fibroblasts from Slc30a10-/- mice. In all cell types, manganese efflux activity of SLC30A10N43A was comparable to WT, while SLC30A10E25A lacked activity. Importantly, unlike SLC30A10, the histidine residue of the HXXXD motif of SLC30A1/ZnT1 was required for zinc transport. These results imply that the mechanisms of ion coordination within the transmembrane domain of SLC30A10 substantially differ from previously-studied CDFs, suggest that factors beyond Site A residues may confer metal specificity to CDFs, and improve understanding of the pathobiology of manganese toxicity due to mutations in SLC30A10.


Assuntos
Proteínas de Transporte de Cátions/metabolismo , Embrião de Mamíferos/metabolismo , Fibroblastos/metabolismo , Manganês/metabolismo , Zinco/metabolismo , Sequência de Aminoácidos , Animais , Sítios de Ligação , Proteínas de Transporte de Cátions/química , Proteínas de Transporte de Cátions/genética , Células Cultivadas , Embrião de Mamíferos/citologia , Fibroblastos/citologia , Células HEK293 , Células Hep G2 , Humanos , Transporte de Íons , Camundongos , Camundongos Knockout , Mutação , Conformação Proteica , Homologia de Sequência
3.
Adv Neurobiol ; 18: 35-49, 2017.
Artigo em Inglês | MEDLINE | ID: mdl-28889262

RESUMO

While the neurotoxic effects of manganese were recognized in 1837, the first genetic disorder of manganese metabolism was described only in 2012 when homozygous mutations in SLC30A10 were reported to cause manganese-induced neurotoxicity. Two other genetic disorders of manganese metabolism have now been described - mutations in SLC39A14 cause manganese toxicity, while mutations in SLC39A8 cause manganese and zinc deficiency. Study of rare genetic disorders often provides unique insights into disease pathobiology, and the discoveries of these three inherited disorders of manganese metabolism are already transforming our understanding of manganese homeostasis, detoxification, and neurotoxicity. Here, we review the mechanisms by which mutations in SLC30A10, SLC39A14, and SLC39A8 impact manganese homeostasis to cause human disease.


Assuntos
Deficiências Nutricionais/metabolismo , Intoxicação por Manganês/metabolismo , Manganês/metabolismo , Erros Inatos do Metabolismo dos Metais/metabolismo , Proteínas de Transporte de Cátions/genética , Deficiências Nutricionais/genética , Deficiências Nutricionais/psicologia , Humanos , Manganês/deficiência , Intoxicação por Manganês/genética , Intoxicação por Manganês/psicologia , Erros Inatos do Metabolismo dos Metais/genética , Erros Inatos do Metabolismo dos Metais/psicologia , Zinco/deficiência , Transportador 8 de Zinco/genética
4.
J Biol Chem ; 291(31): 15940-57, 2016 07 29.
Artigo em Inglês | MEDLINE | ID: mdl-27307044

RESUMO

Homozygous mutations in SLC30A10 lead to the development of familial manganese-induced parkinsonism. We previously demonstrated that SLC30A10 is a cell surface-localized manganese efflux transporter, and parkinsonism-causing mutations block its trafficking and efflux activity. Interestingly, other transporters in the SLC30 family mediate zinc efflux. Determining the mechanisms that allow SLC30A10 to transport manganese, which are unclear, is essential to understand its role in parkinsonism. Here, we generated a predicted structure of SLC30A10, based on the structure of the bacterial zinc transporter YiiP, and performed functional studies. In YiiP, side chains of residues Asp-45 and Asp-49 in the second and His-153 and Asp-157 in the fifth transmembrane segments coordinate zinc and are required for transport. In SLC30A10, the corresponding residues are Asn-43 and Asp-47 in the second and His-244 and Asp-248 in the fifth transmembrane segments. Surprisingly, although alanine substitution of Asp-248 abolished manganese efflux, that of Asn-43 and Asp-47 did not. Instead, side chains of charged or polar residues adjacent to Asp-248 in the first (Glu-25) or fourth (Asn-127) transmembrane segments were required. Further analyses revealed that residues His-333 and His-350 in the cytoplasmic C-terminal domain were required for full activity. However, the C-terminal domain failed to transfer manganese transport capability to a related zinc transporter. Overall, our results indicate that residues in the transmembrane and C-terminal domains together confer optimal manganese transport capability to SLC30A10 and suggest that the mechanism of ion coordination in the transmembrane domain of SLC30A10 may be substantially different from that in YiiP/other SLC30 proteins.


Assuntos
Proteínas de Transporte de Cátions , Manganês , Proteínas de Transporte de Cátions/química , Proteínas de Transporte de Cátions/genética , Proteínas de Transporte de Cátions/metabolismo , Proteínas de Escherichia coli/química , Proteínas de Escherichia coli/genética , Proteínas de Escherichia coli/metabolismo , Células HeLa , Humanos , Transporte de Íons/fisiologia , Manganês/química , Manganês/metabolismo , Proteínas de Membrana Transportadoras/química , Proteínas de Membrana Transportadoras/genética , Proteínas de Membrana Transportadoras/metabolismo , Domínios Proteicos , Homologia Estrutural de Proteína , Transportador 8 de Zinco
5.
J Struct Biol ; 193(3): 196-205, 2016 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-26724270

RESUMO

Polyketides such as the clinically-valuable antibacterial agent mupirocin are constructed by architecturally-sophisticated assembly lines known as trans-acyltransferase polyketide synthases. Organelle-sized megacomplexes composed of several copies of trans-acyltransferase polyketide synthase assembly lines have been observed by others through transmission electron microscopy to be located at the Bacillus subtilis plasma membrane, where the synthesis and export of the antibacterial polyketide bacillaene takes place. In this work we analyze ten crystal structures of trans-acyltransferase polyketide synthases ketosynthase domains, seven of which are reported here for the first time, to characterize a motif capable of zippering assembly lines into a megacomplex. While each of the three-helix LINKS (Laterally-INteracting Ketosynthase Sequence) motifs is observed to similarly dock with a spatially-reversed copy of itself through hydrophobic and ionic interactions, the amino acid sequences of this motif are not conserved. Such a code is appropriate for mediating homotypic contacts between assembly lines to ensure the ordered self-assembly of a noncovalent, yet tightly-knit, enzymatic network. LINKS-mediated lateral interactions would also have the effect of bolstering the vertical association of the polypeptides that comprise a polyketide synthase assembly line.


Assuntos
Motivos de Aminoácidos/genética , Complexos Multiproteicos/química , Polienos/química , Policetídeo Sintases/química , Bacillus subtilis/química , Bacillus subtilis/enzimologia , Cristalografia por Raios X , Modelos Moleculares , Complexos Multiproteicos/genética , Polienos/metabolismo , Policetídeo Sintases/genética , Policetídeo Sintases/ultraestrutura , Estrutura Terciária de Proteína
6.
J Neurosci ; 34(42): 14079-95, 2014 Oct 15.
Artigo em Inglês | MEDLINE | ID: mdl-25319704

RESUMO

Manganese (Mn) is an essential metal, but elevated cellular levels are toxic and may lead to the development of an irreversible parkinsonian-like syndrome that has no treatment. Mn-induced parkinsonism generally occurs as a result of exposure to elevated Mn levels in occupational or environmental settings. Additionally, patients with compromised liver function attributable to diseases, such as cirrhosis, fail to excrete Mn and may develop Mn-induced parkinsonism in the absence of exposure to elevated Mn. Recently, a new form of familial parkinsonism was reported to occur as a result of mutations in SLC30A10. The cellular function of SLC30A10 and the mechanisms by which mutations in this protein cause parkinsonism are unclear. Here, using a combination of mechanistic and functional studies in cell culture, Caenorhabditis elegans, and primary midbrain neurons, we show that SLC30A10 is a cell surface-localized Mn efflux transporter that reduces cellular Mn levels and protects against Mn-induced toxicity. Importantly, mutations in SLC30A10 that cause familial parkinsonism blocked the ability of the transporter to traffic to the cell surface and to mediate Mn efflux. Although expression of disease-causing SLC30A10 mutations were not deleterious by themselves, neurons and worms expressing these mutants exhibited enhanced sensitivity to Mn toxicity. Our results provide novel insights into the mechanisms involved in the onset of a familial form of parkinsonism and highlight the possibility of using enhanced Mn efflux as a therapeutic strategy for the potential management of Mn-induced parkinsonism, including that occurring as a result of mutations in SLC30A10.


Assuntos
Proteínas de Transporte de Cátions/genética , Proteínas de Transporte de Cátions/metabolismo , Manganês/metabolismo , Mutação/genética , Transtornos Parkinsonianos/genética , Transtornos Parkinsonianos/metabolismo , Animais , Caenorhabditis elegans , Membrana Celular/genética , Membrana Celular/metabolismo , Células Cultivadas , Feminino , Células HeLa , Humanos , Líquido Intracelular/metabolismo , Masculino , Camundongos Endogâmicos C57BL , Transporte Proteico/fisiologia , Transportador 8 de Zinco
7.
Biochim Biophys Acta ; 1837(11): 1892-1903, 2014 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-25091280

RESUMO

From the crystal structures of reaction centers (RCs) from purple photosynthetic bacteria, two pathways for electron transfer (ET) are apparent but only one pathway (the A side) operates in the native protein-cofactor complex. Partial activation of the B-side pathway has unveiled the true inefficiencies of ET processes on that side in comparison to analogous reactions on the A side. Of significance are the relative rate constants for forward ET and the competing charge recombination reactions. On the B side, these rate constants are nearly equal for the secondary charge-separation step (ET from bacteriopheophytin to quinone), relegating the yield of this process to <50%. Herein we report efforts to optimize this step. In surveying all possible residues at position 131 in the M subunit, we discovered that when glutamic acid replaces the native valine the efficiency of the secondary ET is nearly two-fold higher than in the wild-type RC. The positive effect of M131 Glu is likely due to formation of a hydrogen bond with the ring V keto group of the B-side bacteriopheophytin leading to stabilization of the charge-separated state involving this cofactor. This change slows charge recombination by roughly a factor of two and affords the improved yield of the desired forward ET to the B-side quinone terminal acceptor.

SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA