Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 11 de 11
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Zootaxa ; 4802(3): zootaxa.4802.3.4, 2020 Jun 24.
Artigo em Inglês | MEDLINE | ID: mdl-33056044

RESUMO

Up to the present, the genus Archiboreoiulus included only two species: A. sollaudi from France and A. pallidus with a wide European distribution range. Here we describe a previously unknown species of the genus Archiboreoiulus, A. serbansarbui, collected from the mesothermal sulfurous Movile Cave, Mangalia, Romania, harboring a rich and diverse troglobitic community.


Assuntos
Artrópodes , Cavernas , Animais , Filogenia , Romênia
2.
Sci Rep ; 6: 36937, 2016 11 17.
Artigo em Inglês | MEDLINE | ID: mdl-27853235

RESUMO

Nuclear receptor PPARγ has been proven to affect metabolism in multiple tissues, and has received considerable attention for its involvement in colon cancer and inflammatory disease. However, its role in intestinal metabolism has been largely ignored. To investigate this potential aspect of PPARγ function, we submitted intestinal epithelium-specific PPARγ knockout mice (iePPARγKO) to a two-week period of 25% caloric restriction (CR), following which iePPARγKO mice retained more fat than their wild type littermates. In attempting to explain this discrepancy, we analysed the liver, skeletal muscle, intestinal lipid trafficking, and the microbiome, none of which appeared to contribute to the adiposity phenotype. Interestingly, under conditions of CR, iePPARγKO mice failed to activate their sympathetic nervous system (SNS) and increase CR-specific locomotor activity. These KO mice also manifested a defective control of their body temperature, which was overly reduced. Furthermore, the white adipose tissue of iePPARγKO CR mice showed lower levels of both hormone-sensitive lipase, and its phosphorylated form. This would result from impaired SNS signalling and possibly cause reduced lipolysis. We conclude that intestinal epithelium PPARγ plays an essential role in increasing SNS activity under CR conditions, thereby contributing to energy mobilization during metabolically stressful episodes.


Assuntos
PPAR gama/metabolismo , Sistema Nervoso Simpático/metabolismo , Tecido Adiposo Branco/metabolismo , Adiposidade/fisiologia , Animais , Restrição Calórica/métodos , Mucosa Intestinal/metabolismo , Lipólise/fisiologia , Fígado/metabolismo , Locomoção/fisiologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Músculo Esquelético/metabolismo
3.
Pflugers Arch ; 468(4): 563-72, 2016 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-26555760

RESUMO

L-arginine is a semi-essential amino acid that serves as precursor for the production of urea, nitric oxide (NO), polyamines, and other biologically important metabolites. Hence, a fast and reliable assessment of its intracellular concentration changes is highly desirable. Here, we report on a genetically encoded Förster resonance energy transfer (FRET)-based arginine nanosensor that employs the arginine repressor/activator ahrC gene from Bacillus subtilis. This new nanosensor was expressed in HEK293T cells, and experiments with cell lysate showed that it binds L-arginine with high specificity and with a K d of ∼177 µM. Live imaging experiments showed that the nanosensor was expressed throughout the cytoplasm and displayed a half maximal FRET increase at an extracellular L-arginine concentration of ∼22 µM. By expressing the nanosensor together with SLC7A1, SLC7A2B, or SLC7A3 cationic amino acid transporters (CAT1-3), it was shown that L-arginine was imported at a similar rate via SLC7A1 and SLC7A2B and slower via SLC7A3. In contrast, upon withdrawal of extracellular L-arginine, intracellular levels decreased as fast in SLC7A3-expressing cells compared with SLC7A1, but the efflux was slower via SLC7A2B. SLC7A4 (CAT4) could not be convincingly shown to transport L-arginine. We also demonstrated the impact of membrane potential on L-arginine transport and showed that physiological concentrations of symmetrical and asymmetrical dimethylarginine do not significantly interfere with L-arginine transport through SLC7A1. Our results demonstrate that the FRET nanosensor can be used to assess L-arginine transport through plasma membrane in real time.


Assuntos
Proteínas de Bactérias/metabolismo , Técnicas Biossensoriais/métodos , Transportador 1 de Aminoácidos Catiônicos/metabolismo , Transferência Ressonante de Energia de Fluorescência/métodos , Proteínas Repressoras/metabolismo , Transativadores/metabolismo , Arginina/metabolismo , Proteínas de Bactérias/química , Células HEK293 , Humanos , Potenciais da Membrana , Proteínas Repressoras/química , Transativadores/química
4.
Nat Commun ; 6: 7250, 2015 May 22.
Artigo em Inglês | MEDLINE | ID: mdl-25998567

RESUMO

Mammalian target of rapamycin 1 (mTORC1), a master regulator of cellular growth, is activated downstream of growth factors, energy signalling and intracellular essential amino acids (EAAs) such as Leu. mTORC1 activation occurs at the lysosomal membrane, and involves V-ATPase stimulation by intra-lysosomal EAA (inside-out activation), leading to activation of the Ragulator, RagA/B-GTP and mTORC1 via Rheb-GTP. How Leu enters the lysosomes is unknown. Here we identified the lysosomal protein LAPTM4b as a binding partner for the Leu transporter, LAT1-4F2hc (SLC7A5-SLAC3A2). We show that LAPTM4b recruits LAT1-4F2hc to lysosomes, leading to uptake of Leu into lysosomes, and is required for mTORC1 activation via V-ATPase following EAA or Leu stimulation. These results demonstrate a functional Leu transporter at the lysosome, and help explain the inside-out lysosomal activation of mTORC1 by Leu/EAA.


Assuntos
Transportador 1 de Aminoácidos Neutros Grandes/metabolismo , Leucina/metabolismo , Lisossomos/metabolismo , Proteínas de Membrana/metabolismo , Complexos Multiproteicos/metabolismo , Proteínas Oncogênicas/metabolismo , Serina-Treonina Quinases TOR/metabolismo , Células HeLa , Humanos , Alvo Mecanístico do Complexo 1 de Rapamicina , ATPases Vacuolares Próton-Translocadoras/metabolismo
5.
PLoS One ; 9(2): e89270, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-24586648

RESUMO

The immune system and iron availability are intimately linked as appropriate iron supply is needed for cell proliferation, while excess iron, as observed in hemochromatosis, may reduce subsets of lymphocytes. We have tested the effects of a ferritin H gene deletion on lymphocytes. Mx-Cre mediated conditional deletion of ferritin H in bone marrow reduced the number of mature B cells and peripheral T cells in all lymphoid organs. FACS analysis showed an increase in the labile iron pool, enhanced reactive oxygen species formation and mitochondrial depolarization. The findings were confirmed by a B-cell specific deletion using Fth(lox/lox) ; CD19-Cre mice. Mature B cells were strongly under-represented in bone marrow and spleen of the deleted mice, whereas pre-B and immature B cells were not affected. Bone marrow B cells showed increased proliferation as judged by the number of cells in S and G2/M phase as well as BrdU incorporation. Upon in vitro culture with B-cell activating factor of the tumor necrosis factor family (BAFF), ferritin H-deleted spleen B cells showed lower survival rates than wild type cells. This was partially reversed with iron-chelator deferiprone. The loss of T cells was also confirmed by a T cell-specific deletion in Fth(lox/lox) ;CD4-Cre mice. Our data show that ferritin H is required for B and T cell survival by actively reducing the labile iron pool. They further suggest that natural B and T cell maturation is influenced by intracellular iron levels and possibly deregulated in iron excess or deprivation.


Assuntos
Apoferritinas/genética , Linfócitos B/citologia , Proliferação de Células/genética , Ferro/metabolismo , Linfócitos T/citologia , Animais , Linfócitos B/metabolismo , Contagem de Células , Diferenciação Celular/genética , Camundongos , Camundongos Knockout , Espécies Reativas de Oxigênio/metabolismo , Baço/citologia , Baço/metabolismo , Linfócitos T/metabolismo
6.
Exp Hematol ; 42(1): 59-69, 2014 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-24141093

RESUMO

The objective was to explore how ferritin-H deletion influences (59)Fe-distribution and excretion-kinetics in mice. Kinetics of (59)Fe-release from organs, whole-body excretion, and distribution-kinetics of intravenously injected (59)Fe trace amounts were compared in iron-deficient and iron-replete mice with (Fth(Δ/Δ)) and without (Fth(lox/lox)) conditional Mx-Cre-induced ferritin-H deletion. (59)Fe was released from spleen and liver beginning on day 2 and day 5 after ferritin-H deletion, respectively, but was not excreted from the body. Plasma-(59)Fe was cleared significantly faster in iron-deficient Fth(Δ/Δ)-mice than in iron-adequate Fth(lox/lox)-controls. (59)Fe-distribution showed a transient peak (e.g., in heart, kidney, muscle) in Fth(lox/lox) control mice, but not in ferritin-H-deleted Fth(Δ/Δ) mice 24 hours after (59)Fe injection. (59)Fe uptake into the liver and spleen was significantly lower in iron-deficient Fth(Δ/Δ) than in Fth(lox/lox) mice 24 hours and 7 days after injection, respectively, and rapidly appeared in circulating erythrocytes instead. The rate of (59)Fe release after ferritin-H deletion supports earlier data on ferritin turnover in mammals; released (59)Fe is not excreted from the body. Instead, (59)Fe is channeled into erythropoiesis and circulating erythrocytes significantly more extensively and faster. Along with a lack of transient interim (59)Fe storage (e.g., in the heart and kidney), this finding is evidence for ferritin-related iron storage-capacity affecting rate and extent of iron utilization.


Assuntos
Apoferritinas/fisiologia , Radioisótopos de Ferro/farmacocinética , Animais , Ferro/metabolismo , Camundongos , Camundongos Endogâmicos C57BL
7.
Cell Host Microbe ; 12(5): 693-704, 2012 Nov 15.
Artigo em Inglês | MEDLINE | ID: mdl-23159058

RESUMO

Disease tolerance is a defense strategy that limits the fitness costs of infection irrespectively of pathogen burden. While restricting iron (Fe) availability to pathogens is perceived as a host defense strategy, the resulting tissue Fe overload can be cytotoxic and promote tissue damage to exacerbate disease severity. Examining this interplay during malaria, the disease caused by Plasmodium infection, we find that expression of the Fe sequestering protein ferritin H chain (FtH) in mice, and ferritin in humans, is associated with reduced tissue damage irrespectively of pathogen burden. FtH protection relies on its ferroxidase activity, which prevents labile Fe from sustaining proapoptotic c-Jun N-terminal kinase (JNK) activation. FtH expression is inhibited by JNK activation, promoting tissue Fe overload, tissue damage, and malaria severity. Mimicking FtH's antioxidant effect or inhibiting JNK activation pharmacologically confers therapeutic tolerance to malaria in mice. Thus, FtH provides metabolic adaptation to tissue Fe overload, conferring tolerance to malaria.


Assuntos
Apoferritinas/metabolismo , Ferritinas/metabolismo , Sobrecarga de Ferro/metabolismo , Malária/metabolismo , Malária/parasitologia , Plasmodium chabaudi/imunologia , Plasmodium chabaudi/fisiologia , Animais , Antioxidantes/metabolismo , Células Cultivadas , Ceruloplasmina/metabolismo , Citoproteção , Ativação Enzimática , Hepatócitos/metabolismo , Interações Hospedeiro-Parasita , Humanos , Ferro/metabolismo , Proteínas Quinases JNK Ativadas por Mitógeno/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Plasmodium berghei/fisiologia , Plasmodium vivax/fisiologia
8.
Cell Metab ; 12(3): 273-82, 2010 Sep 08.
Artigo em Inglês | MEDLINE | ID: mdl-20816093

RESUMO

To maintain appropriate body iron levels, iron absorption by the proximal duodenum is thought to be controlled by hepcidin, a polypeptide secreted by hepatocytes in response to high serum iron. Hepcidin limits basolateral iron efflux from the duodenal epithelium by binding and downregulating the intestinal iron exporter ferroportin. Here, we found that mice with an intestinal ferritin H gene deletion show increased body iron stores and transferrin saturation. As expected for iron-loaded animals, the ferritin H-deleted mice showed induced liver hepcidin mRNA levels and reduced duodenal expression of DMT1 and DcytB mRNA. In spite of these feedback controls, intestinal ferroportin protein and (59)Fe absorption were increased more than 2-fold in the deleted mice. Our results demonstrate that hepcidin-mediated regulation alone is insufficient to restrict iron absorption and that intestinal ferritin H is also required to limit iron efflux from intestinal cells.


Assuntos
Apoferritinas/metabolismo , Absorção Intestinal/fisiologia , Mucosa Intestinal/metabolismo , Ferro/metabolismo , Animais , Peptídeos Catiônicos Antimicrobianos/genética , Peptídeos Catiônicos Antimicrobianos/metabolismo , Apoferritinas/genética , Proteínas de Transporte de Cátions/genética , Proteínas de Transporte de Cátions/metabolismo , Feminino , Hepcidinas , Intestinos/anatomia & histologia , Proteínas Reguladoras de Ferro/genética , Proteínas Reguladoras de Ferro/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Receptores da Transferrina/genética , Receptores da Transferrina/metabolismo
9.
Hepatology ; 50(3): 852-60, 2009 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-19492434

RESUMO

UNLABELLED: Ferritin plays a central role in iron metabolism by acting both as iron storage and a detoxifying protein. We generated a ferritin H allele with loxP sites and studied the conditional ferritin H deletion in adult mice. Ten days after Mx-Cre induced deletion, ferritin H messenger RNA (mRNA) was below 5% in the liver, spleen, and bone marrow of deleted mice compared to control littermates. Mice lost their cellular iron stores indicating the requirement of ferritin H in iron deposition. Serum iron and transferrin saturation were slightly increased and correlated with a two-fold increased liver hepcidin 1 mRNA and a reduced duodenal DcytB mRNA level. Under a normal iron regimen, deleted mice survived for 2 years without visible disadvantage. Mice fed on a high iron diet prior to ferritin H deletion suffered from severe liver damage. Similarly, ferritin H deleted mouse embryonic fibroblasts showed rapid cell death after exposure to iron salt in the medium. This was reversed by wild-type ferritin H but not by a ferritin H mutant lacking ferroxidase activity. Cell death was preceded by an increase in cytoplasmic free iron, reactive oxygen species, and mitochondrial depolarization. CONCLUSION: Our results provide evidence that the iron storage function of ferritin plays a major role in preventing iron-mediated cell and tissue damage.


Assuntos
Apoferritinas/genética , Ferro/metabolismo , Hepatopatias/etiologia , Animais , Peptídeos Catiônicos Antimicrobianos/biossíntese , Apoferritinas/deficiência , Morte Celular/efeitos dos fármacos , Embrião de Mamíferos/efeitos dos fármacos , Hepcidinas , Ferro/toxicidade , Fígado , Masculino , Camundongos , RNA Mensageiro/metabolismo
10.
Genes Cells ; 10(12): 1203-10, 2005 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-16324156

RESUMO

Thioredoxin, an oxidoreductase, is a multifunction protein. The thioredoxin system is composed of NADPH, thioredoxin reductase and thioredoxin. This enzyme is highly conserved from bacteria to humans. We have characterized TRX-1, a thioredoxin homolog in C. elegans, which has about 36% identity in amino acid sequence with human thioredoxin. By gfp reporter system, trx-1 has been shown to be restrictedly expressed in ASI and ASJ neurons and in intestine. Immunostaining confirmed the intestinal expression. Full-length cDNA of trx-1 has been isolated by cDNA library PCR and subsequently cloned and sequenced. We have shown that the encoded protein functions as a reductase in the insulin reducing assay. Moreover, we have isolated a deletion mutant by PCR-based TMP-UV mutagenesis method. Mutant animals have reduced life span and are sensitive to oxidative stress. Reintroduction of trx-1 into mutant worms fully restored the wild-type phenotype. Our results suggest that trx-1 has important functions in life span regulation and oxidative stress response in C. elegans.


Assuntos
Proteínas de Caenorhabditis elegans/metabolismo , Caenorhabditis elegans/fisiologia , Expectativa de Vida , Estresse Oxidativo/fisiologia , Tiorredoxinas/genética , Sequência de Aminoácidos , Animais , Caenorhabditis elegans/enzimologia , Caenorhabditis elegans/genética , Proteínas de Caenorhabditis elegans/química , Proteínas de Caenorhabditis elegans/genética , Humanos , Dados de Sequência Molecular , Estresse Oxidativo/genética , Oxirredutases/metabolismo , Proteínas Recombinantes de Fusão/genética , Proteínas Recombinantes de Fusão/metabolismo , Alinhamento de Sequência , Tiorredoxinas/química , Tiorredoxinas/metabolismo
11.
J Mol Biol ; 352(2): 313-8, 2005 Sep 16.
Artigo em Inglês | MEDLINE | ID: mdl-16084527

RESUMO

The enteric muscle contraction (EMC) is the last step of the defecation behavior which occurs every 50 s in Caenorhabditis elegans. This EMC is regulated by intestinal and anal depressor muscles, which are innervated by GABA motor neurons. Our data show that calcineurin (tax-6) is expressed in intestinal muscle and anal depressor muscle, and the gain-of-function mutant of calcineurin, tax-6(jh107), shows defects in enteric muscle contractions. In addition, the intracellular region of EXP-1, an excitatory GABA receptor, specifically binds to calcineurin A. This interaction between TAX-6 and EXP-1 appears to be independent of both calcium and CNB, which is the calcium-binding regulatory subunit. Genetic evidence of epistasis between cnb-1(jh103) and exp-1(sa6) suggests that calcineurin functions as a negative regulator of excitatory GABA receptor in GABA signaling in C.elegans.


Assuntos
Proteínas de Caenorhabditis elegans/fisiologia , Caenorhabditis elegans/fisiologia , Calcineurina/fisiologia , Receptores de GABA/fisiologia , Sequência de Aminoácidos , Animais , Caenorhabditis elegans/genética , Proteínas de Caenorhabditis elegans/genética , Sistema Digestório/inervação , Fenômenos Fisiológicos do Sistema Digestório , Dados de Sequência Molecular , Neurônios Motores/fisiologia , Contração Muscular/fisiologia , Músculo Liso/inervação , Músculo Liso/fisiologia , Mutação , Receptores de GABA/genética , Transdução de Sinais
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...