Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 807
Filtrar
1.
Kaohsiung J Med Sci ; 38(2): 149-156, 2022 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-34741409

RESUMO

Previous studies have demonstrated that the levels of asymmetric dimethylarginine (ADMA), an endogenous inhibitor of nitric oxide (NO) synthesis, are strongly associated with hypertension, diabetes, and cardiovascular diseases. Profilin-1, an actin-binding protein, has been documented to be involved in endothelial injury and in the proliferation of vascular smooth muscle cells resulting from hypertension. However, the role of profilin-1 in ADMA-induced vascular injury in hypertension remains largely unknown. Forty healthy subjects and forty-two matched patients with essential hypertension were enrolled, and the related indexes of vascular injury in plasma were detected. Rat aortic smooth muscle cells (RASMCs) were treated with different concentrations of ADMA for different periods of time and transfected with profilin-1 small hairpin RNA to interrupt the expression of profilin-1. To determine the role of the Janus kinase 2/signal transducer and activator of transcription 3 (JAK2/STAT3) pathway, RASMCs were pretreated with AG490 or rapamycin. The expression of profilin-1 was tested using real-time polymerase chain reaction (PCR) and western blot analysis. Cell proliferation was measured by flow cytometry and 3-(4,5-dimethylthiazol-2yl)-2,5-diphenyltetrazoliumbromide assays. Compared with healthy subjects, the levels of ADMA and profilin-1 were markedly elevated in hypertensive individuals, while the levels of NO were significantly decreased (p < 0.05). In vitro, studies showed ADMA-induced profilin-1 expression in a concentration- and time-dependent manner in RASMCs (p < 0.05), concomitantly with promoting the proliferation of RASMCs. Furthermore, ADMA-mediated proliferation of RASMCs and upregulation expression of profilin-1 were inhibited by blockade of the JAK2/STAT3 pathway or knockdown of profilin-1. Profilin-1 implicated in the ADMA-mediated vascular lesions in hypertension.


Assuntos
Arginina/análogos & derivados , Endotélio Vascular/efeitos dos fármacos , Hipertensão/etiologia , Miócitos de Músculo Liso/efeitos dos fármacos , Profilinas/fisiologia , Animais , Arginina/farmacologia , Arginina/fisiologia , Proliferação de Células , Endotélio Vascular/patologia , Humanos , Miócitos de Músculo Liso/patologia , Ratos
2.
Bull Exp Biol Med ; 172(2): 270-275, 2021 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-34855076

RESUMO

Incubation of primary culture of pulmonary fibroblasts with non-opiate analogue of leuenkephalin (NALE; Phe-D-Ala-Gly-Phe-Leu-Arg, 0.1 µM) reduced generation of superoxide anion-radical (by 20.7%) and decreased the number of p53+ cells (by 40.2%) induced by exposure to H2O2 (60 µM). The cytoprotective effect of NALE was potentiated by NO synthase inhibitor L-NAME (1 mM): the number of p53+ cells decreased by 65.3% and morphometric parameters of the cell nuclei and nucleoli were improved. Incubation of pulmonary fibroblasts culture with peptide G (Phe-D-Ala-Gly-Phe-Leu-Gly, 0.1 µM) also significantly reduced the damaging effect of H2O2: the number of p53+ cells decreased by 73.5%, the area of cell nuclei returned to normal, and generation of superoxide anion-radical decreased by 18.4%. These results indicate that C-terminal amino acid Arg and activation of NO synthase are not involved in the direct cytoprotective effect of NALE.


Assuntos
Arginina/fisiologia , Encefalina Leucina/farmacologia , Óxido Nítrico/fisiologia , Animais , Arginina/farmacologia , Células Cultivadas , Citoproteção/efeitos dos fármacos , Encefalina Leucina/análogos & derivados , Fibroblastos/efeitos dos fármacos , Fibroblastos/fisiologia , Peróxido de Hidrogênio/farmacologia , Pulmão/citologia , NG-Nitroarginina Metil Éster/farmacologia , Óxido Nítrico/farmacologia , Estresse Oxidativo/efeitos dos fármacos , Substâncias Protetoras/farmacologia , Ratos , Ratos Wistar
3.
Cells ; 10(8)2021 08 17.
Artigo em Inglês | MEDLINE | ID: mdl-34440879

RESUMO

Massive platelet activation and thrombotic events characterize severe COVID-19, highlighting their critical role in SARS-CoV-2-induced immunopathology. Since there is a well-described expansion of myeloid-derived suppressor cells (MDSC) in severe COVID-19, we evaluated their possible role in platelet activation during SARS-CoV-2 infection. During COVID-19, a lower plasmatic L-arginine level was observed compared to healthy donors, which correlated with MDSC frequency. Additionally, activated GPIIb/IIIa complex (PAC-1) expression was higher on platelets from severe COVID-19 patients compared to healthy controls and inversely correlated with L-arginine plasmatic concentration. Notably, MDSC were able to induce PAC-1 expression in vitro by reducing L-arginine concentration, indicating a direct role of PMN-MDSC in platelet activation. Accordingly, we found a positive correlation between ex vivo platelet PAC-1 expression and PMN-MDSC frequency. Overall, our data demonstrate the involvement of PMN-MDSC in triggering platelet activation during COVID-19, highlighting a novel role of MDSC in driving COVID-19 pathogenesis.


Assuntos
Arginina/imunologia , COVID-19/imunologia , Células Supressoras Mieloides/imunologia , Ativação Plaquetária , Trombose/etiologia , Adulto , Idoso , Idoso de 80 Anos ou mais , Arginina/fisiologia , COVID-19/complicações , COVID-19/fisiopatologia , Feminino , Humanos , Masculino , Pessoa de Meia-Idade , Células Supressoras Mieloides/fisiologia , Adulto Jovem
4.
Am J Physiol Endocrinol Metab ; 320(1): E160-E168, 2021 01 01.
Artigo em Inglês | MEDLINE | ID: mdl-33225718

RESUMO

Mutation of arginine 264 in ERα has been shown to abrogate rapid membrane ERα-mediated endothelial effects. Our novel finding that mutation of R264 is dispensable for ERα-mediated skeletal effects supports the concept that R264 determines tissue specificity of ERα. Estrogen protects against bone loss but is not a suitable treatment due to adverse effects in other tissues. Therefore, increased knowledge regarding estrogen signaling in estrogen-responsive tissues is warranted to aid the development of bone-specific estrogen treatments. Estrogen receptor-α (ERα), the main mediator of estrogenic effects in bone, is widely subjected to posttranslational modifications (PTMs). In vitro studies have shown that methylation at site R260 in the human ERα affects receptor localization and intracellular signaling. The corresponding amino acid R264 in murine ERα has been shown to have a functional role in endothelium in vivo, although the methylation of R264 in the murine gene is yet to be empirically demonstrated. The aim of this study was to investigate whether R264 in ERα is involved in the regulation of the skeleton in vivo. Dual-energy X-ray absorptiometry (DEXA) analysis at 3, 6, 9, and 12 mo of age showed no differences in total body areal bone mineral density (BMD) between R264A and wild type (WT) in either female or male mice. Furthermore, analyses using computed tomography (CT) demonstrated that trabecular bone mass in tibia and vertebra and cortical thickness in tibia were similar between R264A and WT mice. In addition, R264A females displayed a normal estrogen treatment response in trabecular bone mass as well as in cortical thickness. Furthermore, uterus, thymus, and adipose tissue responded similarly in R264A and WT female mice after estrogen treatment. In conclusion, our novel finding that mutation of R264 in ERα does not affect the regulation of the skeleton, together with the known role of R264 for ERα-mediated endothelial effects, supports the concept that R264 determines tissue specificity of ERα.NEW & NOTEWORTHY Mutation of arginine 264 in ERα has been shown to abrogate rapid membrane ERα-mediated endothelial effects. Our novel finding that mutation of R264 is dispensable for ERα-mediated skeletal effects supports the concept that R264 determines tissue specificity of ERα.


Assuntos
Arginina/genética , Arginina/fisiologia , Osso e Ossos/fisiologia , Receptor alfa de Estrogênio/genética , Absorciometria de Fóton , Envelhecimento/fisiologia , Animais , Densidade Óssea , Osso e Ossos/diagnóstico por imagem , Endotélio/metabolismo , Estrogênios/farmacologia , Feminino , Metilação , Camundongos , Tamanho do Órgão/genética , Ovariectomia , Coluna Vertebral/química , Coluna Vertebral/metabolismo , Tíbia/química , Tíbia/metabolismo , Tomografia Computadorizada por Raios X
5.
FEBS Open Bio ; 10(10): 2003-2009, 2020 10.
Artigo em Inglês | MEDLINE | ID: mdl-32794631

RESUMO

Chronic kidney disease (CKD) is a worldwide public health problem that is caused by repeated injuries to the glomerulus or renal tubules. Renal fibrosis commonly accompanies CKD, and it is histologically characterized by excessive deposition of extracellular matrix proteins, such as fibronectin and collagen I, in interstitial areas. Indirect in vivo experimental data suggest that renal asymmetric dimethylarginine (ADMA) exerts antifibrotic activity in CKD. In this study, we aimed to demonstrate that renal ADMA has a direct effect on fibrosis in vivo. Normal saline, ADMA, nonsense control siRNA, Ddah1 siRNA or Ddah2 siRNA was administered into the kidney through the left ureter in a mouse model of unilateral ureteral obstruction (UUO). UUO kidneys were harvested at day 1 or 7. Western blotting was performed to assess the expression of ADMA, DDAH1 and DDAH2 and the expression of fibrotic markers, such as fibronectin, collagen I, α-smooth muscle actin, phosphorylation of Smad3 and connective tissue growth factor. Masson's trichrome staining was used to further evaluate renal fibrosis. We observed that intrarenal administration of ADMA increased the renal accumulation of ADMA and attenuated renal fibrosis at days 1 and 7. Knockdown of Ddah1 or Ddah2 increased the amount of ADMA in UUO kidneys and inhibited the expression of fibrotic proteins at days 1 and 7, which was further confirmed by Masson's staining. Thus, our in vivo data suggest that renal ADMA exerts direct antifibrotic effects in a mouse model of UUO.


Assuntos
Arginina/análogos & derivados , Fibrose/fisiopatologia , Amidoidrolases/metabolismo , Amidoidrolases/fisiologia , Animais , Arginina/metabolismo , Arginina/fisiologia , Modelos Animais de Doenças , Fibronectinas/metabolismo , Fibronectinas/farmacologia , Rim/patologia , Nefropatias/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Insuficiência Renal Crônica , Obstrução Ureteral/complicações , Obstrução Ureteral/metabolismo , Obstrução Ureteral/patologia
6.
Cell Death Dis ; 11(8): 662, 2020 08 20.
Artigo em Inglês | MEDLINE | ID: mdl-32814773

RESUMO

Arginine auxotrophy due to the silencing of argininosuccinate synthetase 1 (ASS1) occurs in many carcinomas and in the majority of sarcomas. Arginine deiminase (ADI-PEG20) therapy exploits this metabolic vulnerability by depleting extracellular arginine, causing arginine starvation. ASS1-negative cells develop resistance to ADI-PEG20 through a metabolic adaptation that includes re-expressing ASS1. As arginine-based multiagent therapies are being developed, further characterization of the changes induced by arginine starvation is needed. In order to develop a systems-level understanding of these changes, activity-based proteomic profiling (ABPP) and phosphoproteomic profiling were performed before and after ADI-PEG20 treatment in ADI-PEG20-sensitive and resistant sarcoma cells. When integrated with metabolomic profiling, this multi-omic analysis reveals that cellular response to arginine starvation is mediated by adaptive ERK signaling and activation of the Myc-Max transcriptional network. Concomitantly, these data elucidate proteomic changes that facilitate oxaloacetate production by enhancing glutamine and pyruvate anaplerosis and altering lipid metabolism to recycle citrate for oxidative glutaminolysis. Based on the complexity of metabolic and cellular signaling interactions, these multi-omic approaches could provide valuable tools for evaluating response to metabolically targeted therapies.


Assuntos
Arginina/metabolismo , Sistema de Sinalização das MAP Quinases/fisiologia , Proteínas Proto-Oncogênicas c-myc/metabolismo , Arginina/fisiologia , Argininossuccinato Sintase/genética , Argininossuccinato Sintase/metabolismo , Linhagem Celular Tumoral , Regulação Neoplásica da Expressão Gênica/fisiologia , Glutamina/metabolismo , Humanos , Hidrolases/metabolismo , Hidrolases/farmacologia , Sistema de Sinalização das MAP Quinases/genética , Metabolômica/métodos , Fosfoproteínas/metabolismo , Polietilenoglicóis/farmacologia , Proteômica/métodos , Proteínas Proto-Oncogênicas c-myc/fisiologia , Sarcoma/metabolismo , Transdução de Sinais/fisiologia
7.
Zhongguo Yi Xue Ke Xue Yuan Xue Bao ; 42(6): 815-819, 2020 Dec 30.
Artigo em Chinês | MEDLINE | ID: mdl-33423731

RESUMO

Kidney is one of the important organs of the body.With both excretory and endocrine functions,it plays a vital role in regulating the normal physiological state.As a precursor of the nitric oxide(NO)synthesis in vivo,L-arginine is involved in intracellular and intercellular signaling via NO,a vasoactive factor,thus plays a key role in maintaining the normal physiological functions of the kidney.Alpha1-adrenergic receptor(α1-AR)mediates sympathetic nerves to regulate the heart,blood vessels,and nervous system of the body.The α1-AR distributed in vascular smooth muscle mainly mediates vasoconstriction.The responsiveness of α1-AR to adrenergic agonists decreases in rat models of kidney failure,diabetes,hypertension,and left ventricular hypertrophy,which affects the hemodynamic state and vascular tone of the kidney.Here we analyze the ways via which L-arginine improves the responsiveness of α1-AR to its agonists by ellucidating the action mode of NO/α1-AR and their effects on renal functions.


Assuntos
Arginina/fisiologia , Rim/fisiologia , Receptores Adrenérgicos alfa 1/fisiologia , Animais , Músculo Liso Vascular , Óxido Nítrico/fisiologia , Ratos , Insuficiência Renal/fisiopatologia , Transdução de Sinais , Vasoconstrição
8.
Artigo em Chinês | WPRIM (Pacífico Ocidental) | ID: wpr-878683

RESUMO

Kidney is one of the important organs of the body.With both excretory and endocrine functions,it plays a vital role in regulating the normal physiological state.As a precursor of the nitric oxide(NO)synthesis


Assuntos
Animais , Ratos , Arginina/fisiologia , Rim/fisiologia , Músculo Liso Vascular , Óxido Nítrico/fisiologia , Receptores Adrenérgicos alfa 1/fisiologia , Insuficiência Renal/fisiopatologia , Transdução de Sinais , Vasoconstrição
9.
Sci Rep ; 9(1): 17176, 2019 11 20.
Artigo em Inglês | MEDLINE | ID: mdl-31748670

RESUMO

In recent years methicillin-resistant Staphylococcus aureus has posed a challenge in treating skin and soft tissue infections. Finding new antimicrobial agents has therefore become imperative. We evaluated the in vitro antimicrobial activity of a synthetic peptide, P6, against multidrug resistant clinical strains of Staphylococcus aureus isolated from skin and soft tissue infections. The P6 antimicrobial effect was evaluated in vitro by determining MIC/MBC, the ratio of live/dead cells and the effects induced at membrane level. The therapeutic efficiency was determined against human skin cells. P6 inhibited growth for all strains between 8 and 16 mg/L and killed all bacterial strains at 16 mg/L. The therapeutic potential was found to be 30 and 15 in the presence of BSA. We showed that P6 localizes at membrane level, where it acts slowly, by depolarizing it and affecting its integrity. P6 can be considered a good candidate for use as an antimicrobial agent in topical applications.


Assuntos
Arginina/fisiologia , Staphylococcus aureus Resistente à Meticilina/efeitos dos fármacos , Peptídeos/farmacologia , Infecções dos Tecidos Moles/tratamento farmacológico , Infecções Estafilocócicas/tratamento farmacológico , Infecções Cutâneas Estafilocócicas/tratamento farmacológico , Triptofano/farmacologia , Antibacterianos/farmacologia , Humanos , Testes de Sensibilidade Microbiana/métodos
10.
Clin Sci (Lond) ; 133(20): 2061-2067, 2019 10 30.
Artigo em Inglês | MEDLINE | ID: mdl-31654065

RESUMO

The precise mechanisms underlying resistant hypertension remain elusive. Reduced nitric oxide (NO) bioavailability is frequently documented in chronic kidney disease, obesity, diabetes and advanced age, all of which are risk factors for resistant hypertension. Sympathetic overactivity and chronic activation of the renin-angiotensin system are salient features of resistant hypertension. Interestingly, recent data indicate that renal sympathetic overactivity can reduce the expression of neuronal nitric oxide synthase in the paraventricular nucleus. Reduced NO levels in the paraventricular nucleus can increase sympathetic outflow and this can create a vicious cycle contributing to resistant hypertension. Angiotensin II can reduce l-arginine transport and hence NO production. Reduced NO levels may reduce the formation of angiotensin 1-7 dampening the cardio-protective effects of the renin-angiotensin system contributing to resistant hypertension. In addition, interleukin-6 (IL-6) is demonstrated to be independently associated with resistant hypertension, and IL-6 can reduce NO synthesis. Despite this, NO levels have not been quantified in resistant hypertension. Findings from a small proof of concept study indicate that NO donors can reduce blood pressure in patients with resistant hypertension but more studies are required to validate these preliminary findings. In the present paper, we put forward the hypothesis that reduced NO bioavailability contributes substantially to the development of resistant hypertension.


Assuntos
Arginina/fisiologia , Hipertensão/fisiopatologia , Óxido Nítrico/fisiologia , Disponibilidade Biológica , Endotélio Vascular/fisiopatologia , Humanos , Hipertensão/etiologia , Hipertensão/terapia , Inflamação/complicações , Óxido Nítrico/deficiência , Óxido Nítrico/farmacocinética , Sistema Renina-Angiotensina/fisiologia , Transdução de Sinais/fisiologia , Sistema Nervoso Simpático/fisiopatologia , Falha de Tratamento , Rigidez Vascular/fisiologia
11.
Protein J ; 38(4): 377-388, 2019 08.
Artigo em Inglês | MEDLINE | ID: mdl-31401776

RESUMO

The twin-arginine protein translocation (Tat) system has been characterized in bacteria, archaea and the chloroplast thylakoidal membrane. This system is distinct from other protein transport systems with respect to two key features. Firstly, it accepts cargo proteins with an N-terminal signal peptide that carries the canonical twin-arginine motif, which is essential for transport. Second, the Tat system only accepts and translocates fully folded cargo proteins across the respective membrane. Here, we review the core essential features of folded protein transport via the bacterial Tat system, using the three-component TatABC system of Escherichia coli and the two-component TatAC systems of Bacillus subtilis as the main examples. In particular, we address features of twin-arginine signal peptides, the essential Tat components and how they assemble into different complexes, mechanistic features and energetics of Tat-dependent protein translocation, cytoplasmic chaperoning of Tat cargo proteins, and the remarkable proofreading capabilities of the Tat system. In doing so, we present the current state of our understanding of Tat-dependent protein translocation across biological membranes, which may serve as a lead for future investigations.


Assuntos
Proteínas de Escherichia coli , Proteínas de Membrana Transportadoras , Transporte Proteico/fisiologia , Sistema de Translocação de Argininas Geminadas , Arginina/fisiologia , Bacillus subtilis , Membrana Celular/metabolismo , Escherichia coli , Proteínas de Escherichia coli/química , Proteínas de Escherichia coli/fisiologia , Proteínas de Membrana Transportadoras/química , Proteínas de Membrana Transportadoras/fisiologia , Dobramento de Proteína , Sinais Direcionadores de Proteínas/fisiologia , Canais de Translocação SEC/química , Canais de Translocação SEC/fisiologia , Sistema de Translocação de Argininas Geminadas/química , Sistema de Translocação de Argininas Geminadas/fisiologia
12.
Sex Med Rev ; 7(4): 661-668, 2019 10.
Artigo em Inglês | MEDLINE | ID: mdl-30987932

RESUMO

INTRODUCTION: Testosterone deficiency is known to induce endothelial dysfunction, which can lead to erectile dysfunction and/or vascular dysfunction. In some basic and clinical reports, testosterone has been shown to regulate the nitric oxide (NO)/cyclic guanosine monophosphate (cGMP) pathway and thereby influence endothelial function and endothelial progenitor cells (EPCs), which are key for the endothelial repair system. AIM: To review the association between testosterone and endothelial dysfunction focusing on NO and EPCs. METHODS: A review of relevant literature up to September 2018 was performed via PubMed. MAIN OUTCOME MEASURES: We reviewed the association between testosterone and endothelial dysfunction focusing on NO derived from endothelial NO synthase, phosphodiesterase type 5 (PDE-5), asymmetric dimethylarginine (ADMA), inflammation, and EPCs. RESULTS: Numerous articles describing the association between testosterone deficiency and endothelial dysfunction have been published. Some reports have shown that testosterone deficiency decreases NO production by altering the expression and activity of NO synthase and by regulating ADMA expression. Testosterone also regulates the expression of phosphodiesterase type 5. In addition, some basic and clinical studies have shown that testosterone affects the function and number of EPCs. However, some inconsistencies among these reports have been noted. CONCLUSION: Testosterone deficiency might cause endothelial dysfunction by decreasing NO levels through regulating the expression and activity of NO synthase and increasing ADMA expression. In addition, testosterone might affect the endothelial repair system by regulating the proliferation and migration of EPCs. Testosterone replacement therapy might be useful for treating endothelial dysfunction, considering that some reports have shown that this therapy improved NO bioavailability and EPC function. Hotta Y, Kataoka T, Kimura K. Testosterone Deficiency and Endothelial Dysfunction: Nitric Oxide, Asymmetric Dimethylarginine, and Endothelial Progenitor Cells. Sex Med Rev 2019;7:661-668.


Assuntos
Arginina/análogos & derivados , Células Progenitoras Endoteliais/fisiologia , Endotélio Vascular/fisiologia , Óxido Nítrico/fisiologia , Testosterona/deficiência , Arginina/fisiologia , Nucleotídeo Cíclico Fosfodiesterase do Tipo 5/fisiologia , Humanos , Masculino , Óxido Nítrico Sintase Tipo III/fisiologia , Testosterona/uso terapêutico , Vasculite/fisiopatologia
13.
J Anim Sci ; 96(12): 5035-5051, 2018 Dec 03.
Artigo em Inglês | MEDLINE | ID: mdl-30445424

RESUMO

Arginine (Arg) has traditionally not been considered as a deficient nutrient in diets for gestating or lactating swine due to the assumption that these animals can synthesize sufficient amounts of Arg to meet their physiological needs. The lack of full knowledge about Arg nutrition has contributed to suboptimal efficiency of pork production. Over the past 25 yr, there has been growing interest in Arg metabolism in the pig, which is an agriculturally important species and a useful model for studying human biology. Arginine is a highly abundant amino acid in tissues of pigs, a major amino acid in allantoic fluid, and a key regulator of gene expression, cell signaling, and antioxidative reactions. Emerging evidence suggests that dietary supplementation with 0.5% to 1% Arg maintains gut health and prevents intestinal dysfunction in weanling piglets, while enhancing their growth performance and survival. Also, the inclusion of 1% Arg in diets is required to maximize skeletal muscle accretion and feed efficiency in growing pigs, whereas dietary supplementation with 1% Arg reduces muscle loss in endotoxin-challenged pigs. Furthermore, supplementing 0.83% Arg to corn- and soybean meal-based diets promotes embryonic/fetal survival in swine and milk production by lactating sows. Thus, an adequate amount of dietary Arg as a quantitatively major nutrient is necessary to support maximum growth, lactation, and reproduction performance of swine. These results also have important implications for improving the nutrition and health of humans and other animals.


Assuntos
Fenômenos Fisiológicos da Nutrição Animal , Arginina/administração & dosagem , Lactação/fisiologia , Prenhez , Suínos/fisiologia , Animais , Arginina/fisiologia , Feminino , Gravidez
14.
Proc Natl Acad Sci U S A ; 115(47): 11923-11928, 2018 11 20.
Artigo em Inglês | MEDLINE | ID: mdl-30397112

RESUMO

Arginine-rich cell-penetrating peptides do not enter cells by directly passing through a lipid membrane; they instead passively enter vesicles and live cells by inducing membrane multilamellarity and fusion. The molecular picture of this penetration mode, which differs qualitatively from the previously proposed direct mechanism, is provided by molecular dynamics simulations. The kinetics of vesicle agglomeration and fusion by an iconic cell-penetrating peptide-nonaarginine-are documented via real-time fluorescence techniques, while the induction of multilamellar phases in vesicles and live cells is demonstrated by a combination of electron and fluorescence microscopies. This concert of experiments and simulations reveals that the identified passive cell penetration mechanism bears analogy to vesicle fusion induced by calcium ions, indicating that the two processes may share a common mechanistic origin.


Assuntos
Peptídeos Penetradores de Células/química , Peptídeos Penetradores de Células/metabolismo , Fusão de Membrana/fisiologia , Arginina/metabolismo , Arginina/fisiologia , Transporte Biológico , Membrana Celular/metabolismo , Cinética , Bicamadas Lipídicas/química , Fusão de Membrana/efeitos dos fármacos , Membranas/metabolismo , Simulação de Dinâmica Molecular , Peptídeos/química , Peptídeos/fisiologia , Pseudópodes/metabolismo , Pseudópodes/fisiologia
15.
Exp Eye Res ; 174: 185-195, 2018 09.
Artigo em Inglês | MEDLINE | ID: mdl-29782825

RESUMO

Several mutations associated with congenital cataracts in human beings target conserved arginine residues in αA-crystallin. The N-terminal region of αA-crystallin is a "mutational hotspot," with multiple cataract-related mutations reported in this region. Two mutations at arginine 21 in the N-terminal domain of αA-crystallin - αA-R21L and αA-R21W have been associated with congenital cataract. A third mutant of R21, αA-R21Q, was recently identified to be associated with congenital cataract in a South Australian family. The point mutation was reported to compromise the quaternary structure of αA-crystallin by preventing its assembly into higher ordered oligomers. To assess the effect of the αA-R21Q mutation on αA-crystallin function, recombinant αA-R21Q was expressed, purified and characterized in vitro. Compared to wild-type αA-crystallin, the recombinant αA-R21Q exhibits enhanced chaperone-like activity, increased surface hydrophobicity, lesser stability in urea and increased susceptibility to digestion by trypsin. αA-R21Q demonstrated increased binding affinity towards unfolding ADH and bovine lens fiber cell membranes. αA-R21Q homo-oligomers and hetero-oligomers also prevented H2O2-induced apoptosis in ARPE-19 cells. Taken together, αA-R21Q exhibited a gain of function despite subtle structural differences as compared to wild-type αA-crystallin. This study further validates the involvement of arginine 21 in regulating αA-crystallin structure and function.


Assuntos
Catarata , Cristalino/química , Cadeia A de alfa-Cristalina , Animais , Apoptose/fisiologia , Arginina/genética , Arginina/fisiologia , Catarata/genética , Catarata/metabolismo , Bovinos , Humanos , Chaperonas Moleculares/fisiologia , Mutação Puntual , Ligação Proteica/fisiologia , Dobramento de Proteína , Ureia/metabolismo , Cadeia A de alfa-Cristalina/química , Cadeia A de alfa-Cristalina/genética , Cadeia A de alfa-Cristalina/fisiologia
16.
Sci Rep ; 8(1): 3066, 2018 02 15.
Artigo em Inglês | MEDLINE | ID: mdl-29449648

RESUMO

Mutations in the sodium bicarbonate cotransporter NBCe1 (SLC4A4) cause proximal renal tubular acidosis (pRTA). We recently described a novel pRTA mutation p.Gln913Arg (Q913R), inherited in compound heterozygous form with p.Arg510His (R510H). Q913R causes intracellular retention of NBCe1 and a 'gain of function' Cl- leak. To learn more about the importance of glutamine at position 913, we substituted a variety of alternative amino-acid residues (Cys, Glu, Lys, Leu, Ser) at position 913. Studying cRNA-injected Xenopus oocytes by voltage clamp, we find that most de novo mutants exhibit close-to-normal NBCe1 activity; only Q913K expresses a Cl- leak. Studying transiently-transfected, polarised kidney cells by fluorescence microscopy we find that most de novo mutants (except Q913E) are intracellularly retained. A 3D homology model predicts that Gln913 is located in the gating domain of NBCe1 and neighbours the 3D space occupied by another pRTA-associated residue (Arg881), highlighting an important and conformationally-sensitive region of NBCe1. We conclude that the intracellular retention of Q913R is caused by the loss of Gln at position 913, but that the manifestation of the Cl- leak is related to the introduction of Arg at position 913. Our findings will inform future studies to elucidate the nature and the consequences of the leak.


Assuntos
Arginina , Mutação com Ganho de Função/genética , Glutamina , Espaço Intracelular/metabolismo , Domínios Proteicos/genética , Simportadores de Sódio-Bicarbonato , Acidose Tubular Renal/genética , Acidose Tubular Renal/patologia , Animais , Arginina/genética , Arginina/fisiologia , Bicarbonatos/metabolismo , Membrana Celular/metabolismo , Cães , Glutamina/genética , Glutamina/fisiologia , Humanos , Células Madin Darby de Rim Canino , Oócitos , Sódio/metabolismo , Simportadores de Sódio-Bicarbonato/química , Simportadores de Sódio-Bicarbonato/genética , Xenopus laevis
17.
J Pharm Pharmacol ; 70(4): 507-515, 2018 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-29380385

RESUMO

OBJECTIVES: The main purpose of this study was to assess the role of l-arginine/SNAP/NO/cGMP/KATP channel pathway in analgesic effects of α-terpineol in mice. METHODS: Male NMRI mice were pretreated intraperitoneally with NO precursor (l-arginine, 100 mg/kg), NO synthase inhibitor (l-NAME, 30 mg/kg), NO donor (SNAP, 1 mg/kg), guanylyl cyclase inhibitor (methylene blue, 20 mg/kg), PDE inhibitor (sildenafil, 0.5 mg/kg), KATP channel blocker (glibenclamide, 10 mg/kg) and naloxone (2 mg/kg) 20 min before the administration of α-terpineol. The formalin test was performed 20 min after the administration of α-terpineol, and nociceptive responses of mice were recorded during 30 min. KEY FINDINGS: A significant and dose-dependent antinociception was produced by α-terpineol (40 and 80 mg/kg) in both the phases of formalin test. The antinociceptive effect of α-terpineol was significantly potentiated by l-arginine in the second phase while significantly antagonized by l-NAME in both phases of formalin test. Also, SNAP and sildenafil non-significantly enhanced-while methylene blue significantly diminished-the antinociceptive effect of α-terpineol in both phases of formalin test. Glibenclamide significantly reversed the α-terpineol-induced antinociception, indicating the involvement of KATP channels in antinociceptive effect of α-terpineol. CONCLUSIONS: These results indicate that the antinociceptive effect of α-terpineol is mediated through l-arginine/SNAP/NO/cGMP/KATP channel pathway.


Assuntos
Arginina/fisiologia , GMP Cíclico/fisiologia , Cicloexenos/uso terapêutico , Canais KATP/fisiologia , Monoterpenos/uso terapêutico , Óxido Nítrico/fisiologia , Medição da Dor/efeitos dos fármacos , S-Nitroso-N-Acetilpenicilamina/farmacologia , Analgésicos/farmacologia , Analgésicos/uso terapêutico , Animais , Monoterpenos Cicloexânicos , Cicloexenos/farmacologia , Relação Dose-Resposta a Droga , Masculino , Camundongos , Monoterpenos/farmacologia , NG-Nitroarginina Metil Éster/farmacologia , Óxido Nítrico/antagonistas & inibidores , Dor/tratamento farmacológico , Medição da Dor/métodos , Transdução de Sinais/efeitos dos fármacos , Transdução de Sinais/fisiologia , Resultado do Tratamento
18.
Nihon Saikingaku Zasshi ; 72(4): 219-227, 2017.
Artigo em Japonês | MEDLINE | ID: mdl-29109335

RESUMO

Many bacteria symbiotic and parasitic in humans are included in the genera Bacteroides, Prevotella, Porphyromonas and others, which belong to the phylum Bacteroidetes. We have been studying gingipain, a major secretory protease of Porphyromonas gingivalis which is a periodontopathogenic bacterium belonging to the genus Porphyromonas, and pili which contribute to host colonization in the bacterium. In the process, it was found that gingipain was secreted by a system not reported previously. Furthermore, this secretion system was found to exist widely in the Bacteroidetes phylum bacteria and closely related to the gliding motility of bacteroidete bacteria, and it was named the Por secretion system (later renamed the type IX secretion system). Regarding P. gingivalis pili, it was found that the pilus protein is transported as a lipoprotein to the cell surface, and the pilus formation occurs due to degradation by arginine-gingipain. Pili with this novel formation mechanism was found to be widely present in bacteria belonging to the class Bacteroidia in the phylum Bacteroidetes and was named the type V pili.


Assuntos
Adesinas Bacterianas/fisiologia , Sistemas de Secreção Bacterianos , Bacteroidetes/metabolismo , Bacteroidetes/patogenicidade , Cisteína Endopeptidases/metabolismo , Cisteína Endopeptidases/fisiologia , Fímbrias Bacterianas/fisiologia , Animais , Arginina/fisiologia , Aderência Bacteriana , Proteínas de Bactérias/metabolismo , Bacteroidetes/genética , Bacteroidetes/fisiologia , Membrana Celular/metabolismo , Fímbrias Bacterianas/metabolismo , Genoma Bacteriano , Cisteína Endopeptidases Gingipaínas , Humanos , Lipoproteínas/metabolismo , Camundongos , Periodontite/microbiologia , Porphyromonas gingivalis , Transporte Proteico
19.
Am J Epidemiol ; 186(5): 603-611, 2017 Sep 01.
Artigo em Inglês | MEDLINE | ID: mdl-28911008

RESUMO

The amino acid arginine is a physiological precursor to nitric oxide, which is a key mediator of embryonic survival, fetal growth, and pregnancy maintenance. We evaluated the association between consumption of the amino acid arginine and the rate of adverse birth outcomes using data from a double-blind, randomized, placebo-controlled micronutrient supplementation trial among pregnant women in Dar es Salaam, Tanzania (2001-2004). Dietary intakes of arginine were assessed using repeated 24-hour recalls that were administered throughout pregnancy. Participants (n = 7,591) were monitored by research midwives throughout follow-up to assess pregnancy outcomes. Cubic-restricted splines and multivariable log-Poisson regression with empirical standard errors were used to estimate the continuous and categorical associations between arginine intake and adverse birth outcomes. Compared with women within the lowest quintile of arginine intake, those within the highest quintile had 0.79 times the risk of preterm birth before 37 weeks (95% confidence interval: 0.63, 1.00; P = 0.03). The continuous associations of arginine intake with preterm birth before 37 weeks and with preterm birth before 34 weeks were characterized by an initial rapid decrease in risk with increasing intake (P for nonlinearity < 0.01). Arginine intake was not associated with fetal loss or giving birth to infants who were born small for their gestational ages. This data suggest that the association between dietary arginine intake and preterm birth warrants further investigation.


Assuntos
Arginina/fisiologia , Dieta , Resultado da Gravidez/epidemiologia , Nascimento Prematuro/epidemiologia , Arginina/administração & dosagem , Suplementos Nutricionais/estatística & dados numéricos , Feminino , Humanos , Recém-Nascido , Micronutrientes/administração & dosagem , Micronutrientes/fisiologia , Distribuição de Poisson , Gravidez , Nascimento Prematuro/prevenção & controle , Estudos Prospectivos , Ensaios Clínicos Controlados Aleatórios como Assunto , Tanzânia/epidemiologia
20.
Toxins (Basel) ; 9(3)2017 03 06.
Artigo em Inglês | MEDLINE | ID: mdl-28272322

RESUMO

Asymmetric and symmetric dimethylarginine (ADMA and SDMA, respectively) are toxic, non-proteinogenic amino acids formed by post-translational modification and are uremic toxins that inhibit nitric oxide (NO) production and play multifunctional roles in many human diseases. Both ADMA and SDMA have emerged as strong predictors of cardiovascular events and death in a range of illnesses. Major progress has been made in research on ADMA-lowering therapies in animal studies; however, further studies are required to fill the translational gap between animal models and clinical trials in order to treat human diseases related to elevated ADMA/SDMA levels. Here, we review the reported impacts of ADMA and SDMA on human health and disease, focusing on the synthesis and metabolism of ADMA and SDMA; the pathophysiological roles of these dimethylarginines; clinical conditions and animal models associated with elevated ADMA and SDMA levels; and potential therapies against ADMA and SDMA. There is currently no specific pharmacological therapy for lowering the levels and counteracting the deleterious effects of ADMA and SDMA. A better understanding of the mechanisms underlying the impact of ADMA and SDMA on a wide range of human diseases is essential to the development of specific therapies against diseases related to ADMA and SDMA.


Assuntos
Arginina/análogos & derivados , Toxinas Biológicas , Animais , Arginina/metabolismo , Arginina/fisiologia , Humanos , Toxinas Biológicas/metabolismo , Toxinas Biológicas/fisiologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...