Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 267
Filtrar
1.
Ann Indian Acad Neurol ; 24(4): 518-523, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-34728944

RESUMO

BACKGROUND: Establishing an association between gait variability and direction specific balance indices may help in identifying the risk of falls in patients with spinocerebellar ataxia (SCA) which may help in developing an appropriate intervention. This study is intended to identify the association between balance and gait parameters especially gait variability in these patients. METHODS: Patients with genetically confirmed SCA (n = 24) as well as controls (n = 24) who met the study criteria were recruited. Gait was assessed using the GAITRite system and balance was assessed using dynamic posturography (Biodex) to record direction-specific dynamic balance indices. Disease severity was assessed using international cooperative ataxia rating scale (ICARS). RESULTS: The mean age of the SCA group (38.83 ± 13.03 years) and the control group (36.38 ± 9.09 years) were comparable. The age of onset of illness was 32 ± 10.62 years and duration of 5.67 ± 3.62 years. The mean ICARS was 45.10 ± 16.75. There was a significant difference in the overall balance index (OBI), anterior-posterior index (API), medial/lateral index (MLI) between SCA patients (4.56 ± 2.09, 3.49 ± 1.88, 2.94 ± 1.32) and the controls (2.72 ± 1.25, 2.08 ± 0.85, 1.85 ± 0.97). However, correlation was observed only between gait stability and balance parameters in API direction. CONCLUSIONS: There was an increased anteroposterior oriented balance deficit in patients with SCA, which was significantly correlating with the gait parameters. The balance training intervention may focus on improving anteroposterior direction to prevent falls and improving walking efficiency.

3.
Colloids Surf B Biointerfaces ; 171: 75-84, 2018 Nov 01.
Artigo em Inglês | MEDLINE | ID: mdl-30015141

RESUMO

In this work, phytoprotein functionalized platinum nanoparticles (PtNCs) were synthesized using the proteins from fresh green spinach leaves. Transmission electron microscopy showed that PtNCs were spherical shape with size ∼5 nm, which self assembled into spherical platinum nanoclustures (PtNCs) with size within the range of 100-250 nm. The presence of elemental platinum was confirmed by EDX analysis. FTIR studies confirm that the PtNCs were stabilized by the protein. As prepared PtNCs inhibits the growth of the food borne pathogen, Salmonella typhi with minimum inhibitory concentration (MIC) of 12.5 µM. Light microscopy evidenced that the PtNCs can damage the established biofilms. Antibacterial mechanistic study revealed that PtNCs damages the S. typhi membranes, which was confirmed by scanning electron microscopy and further by fluorescence microscopy using acridine orange/propidium iodide dual staining assay. Besides membrane damage, PtNCs also triggered the intracellular ROS-mediated oxidative damage over the antioxidant defense and kills S. typhi. The hemolytic test showed low cytotoxicity of PtNCs at 100 µM (four times higher the MIC). Finally, the therapeutic efficacy of PtNCs was validated in S. typhi infected zebrafish animal model and the obtained results are discussed.


Assuntos
Antibacterianos/síntese química , Antibacterianos/farmacologia , Biofilmes/efeitos dos fármacos , Nanopartículas Metálicas/química , Platina/farmacologia , Platina/uso terapêutico , Salmonella typhi/efeitos dos fármacos , Febre Tifoide/tratamento farmacológico , Animais , Antibacterianos/química , Modelos Animais de Doenças , Testes de Sensibilidade Microbiana , Microscopia Eletrônica de Varredura , Microscopia de Fluorescência , Tamanho da Partícula , Platina/química , Salmonella typhi/crescimento & desenvolvimento , Propriedades de Superfície , Peixe-Zebra
4.
Br J Pharmacol ; 173(23): 3292-3306, 2016 12.
Artigo em Inglês | MEDLINE | ID: mdl-27747870

RESUMO

BACKGROUND AND PURPOSE: Pancreatic cancer is a solid tumour that is often fatal. Hence, there is an urgent need to identify new drug targets for this disease. Highly proliferating cancer cells have an increased demand for nutrients and, therefore, need to up-regulate selective amino acid transporters. Here, we investigated which amino acid transporters are up-regulated in pancreatic cancer and whether any of these transporters has potential as a drug target for this fatal disease. EXPERIMENTAL APPROACH: The expression of amino acid transporters in pancreatic cancer was analysed using publicly available microarray datasets, and the findings with the transporter SLC6A14 were validated by mRNA and protein analysis. The potential of SLC6A14 as a drug target was evaluated using a pharmacological blocker in vitro and in vivo. KEY RESULTS: SLC6A14 was up-regulated several fold in patient-derived xenografts, primary tumour tissues and pancreatic cancer cells lines compared to normal pancreatic tissue or normal pancreatic epithelial cells. The magnitude of the up-regulation of SLC6A14 was the highest among the amino acid transporters examined. A pharmacological blocker of SLC6A14, α-methyltryptophan, induced amino acid starvation in pancreatic cancer cells and reduced the growth and proliferation of these cells, both in vitro and in vivo. CONCLUSION AND IMPLICATIONS: The salient features of this study are that SLC6A14 is markedly up-regulated in pancreatic cancer and that pharmacological blockade of this transporter interferes with amino acid nutrition and reduces growth and proliferation of pancreatic cancer cells. These findings identify SLC6A14 as a novel druggable target for pancreatic cancer.


Assuntos
Sistemas de Transporte de Aminoácidos Neutros/genética , Terapia de Alvo Molecular , Neoplasias Pancreáticas/tratamento farmacológico , Triptofano/análogos & derivados , Sistemas de Transporte de Aminoácidos , Sistemas de Transporte de Aminoácidos Neutros/antagonistas & inibidores , Animais , Antineoplásicos/farmacologia , Linhagem Celular Tumoral , Feminino , Regulação Neoplásica da Expressão Gênica , Humanos , Camundongos , Camundongos Nus , Neoplasias Pancreáticas/genética , Neoplasias Pancreáticas/patologia , RNA Mensageiro/metabolismo , Triptofano/farmacologia , Regulação para Cima , Ensaios Antitumorais Modelo de Xenoenxerto
5.
Oncogenesis ; 5(6): e238, 2016 Jun 27.
Artigo em Inglês | MEDLINE | ID: mdl-27348268

RESUMO

Composition of the gut microbiota has profound effects on intestinal carcinogenesis. Diet and host genetics play critical roles in shaping the composition of gut microbiota. Whether diet and host genes interact with each other to bring specific changes in gut microbiota that affect intestinal carcinogenesis is unknown. Ability of dietary fibre to specifically increase beneficial gut microbiota at the expense of pathogenic bacteria in vivo via unknown mechanism is an important process that suppresses intestinal inflammation and carcinogenesis. Free fatty acid receptor 2 (FFAR2 or GPR43) is a receptor for short-chain fatty acids (acetate, propionate and butyrate), metabolites of dietary fibre fermentation by gut microbiota. Here, we show FFAR2 is down modulated in human colon cancers than matched adjacent healthy tissue. Consistent with this, Ffar2(-/-) mice are hypersusceptible to development of intestinal carcinogenesis. Dietary fibre suppressed colon carcinogenesis in an Ffar2-dependent manner. Ffar2 played an essential role in dietary fibre-mediated promotion of beneficial gut microbiota, Bifidobacterium species (spp) and suppression of Helicobacter hepaticus and Prevotellaceae. Moreover, numbers of Bifidobacterium is reduced, whereas those of Prevotellaceae are increased in human colon cancers than matched adjacent normal tissue. Administration of Bifidobacterium mitigated intestinal inflammation and carcinogenesis in Ffar2(-/-) mice. Taken together, these findings suggest that interplay between dietary fibre and Ffar2 play a key role in promoting healthy composition of gut microbiota that stimulates intestinal health.

6.
Cell Death Differ ; 22(12): 1922-34, 2015 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-25952549

RESUMO

The Ufm1 conjugation system is a novel ubiquitin-like modification system, consisting of Ufm1, Uba5 (E1), Ufc1 (E2) and poorly characterized E3 ligase(s). RCAD/Ufl1 (also known as KIAA0776, NLBP and Maxer) was reported to function as a Ufm1 E3 ligase in ufmylation (Ufm1-mediated conjugation) of DDRGK1 and ASC1 proteins. It has also been implicated in estrogen receptor signaling, unfolded protein response (UPR) and neurodegeneration, yet its physiological function remains completely unknown. In this study, we report that RCAD/Ufl1 is essential for embryonic development, hematopoietic stem cell (HSC) survival and erythroid differentiation. Both germ-line and somatic deletion of RCAD/Ufl1 impaired hematopoietic development, resulting in severe anemia, cytopenia and ultimately animal death. Depletion of RCAD/Ufl1 caused elevated endoplasmic reticulum stress and evoked UPR in bone marrow cells. In addition, loss of RCAD/Ufl1 blocked autophagic degradation, increased mitochondrial mass and reactive oxygen species, and led to DNA damage response, p53 activation and enhanced cell death of HSCs. Collectively, our study provides the first genetic evidence for the indispensable role of RCAD/Ufl1 in murine hematopoiesis and development. The finding of RCAD/Ufl1 as a key regulator of cellular stress response sheds a light into the role of a novel protein network including RCAD/Ufl1 and its associated proteins in regulating cellular homeostasis.


Assuntos
Hematopoese , Ubiquitina-Proteína Ligases/metabolismo , Anemia/etiologia , Animais , Autofagia , Células da Medula Óssea/citologia , Células da Medula Óssea/metabolismo , Células Cultivadas , Estresse do Retículo Endoplasmático , Células HEK293 , Células-Tronco Hematopoéticas/citologia , Células-Tronco Hematopoéticas/metabolismo , Humanos , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Proteínas Associadas aos Microtúbulos/genética , Proteínas Associadas aos Microtúbulos/metabolismo , Mitocôndrias/metabolismo , Células-Tronco Embrionárias Murinas/citologia , Células-Tronco Embrionárias Murinas/metabolismo , Interferência de RNA , Espécies Reativas de Oxigênio/metabolismo , Proteína Supressora de Tumor p53/metabolismo , Ubiquitina-Proteína Ligases/deficiência , Ubiquitina-Proteína Ligases/genética , Resposta a Proteínas não Dobradas
7.
Cell Death Dis ; 5: e1246, 2014 May 22.
Artigo em Inglês | MEDLINE | ID: mdl-24853424

RESUMO

Hyperactivation of the Wingless-type (Wnt)/ß-catenin pathway promotes tumor initiation, tumor growth and metastasis in various tissues. Although there is evidence for the involvement of Wnt/ß-catenin pathway activation in salivary gland tumors, the precise mechanisms are unknown. Here we report for the first time that downregulation of the Wnt inhibitory factor 1 (WIF1) is a widespread event in salivary gland carcinoma ex-pleomorphic adenoma (CaExPA). We also show that WIF1 downregulation occurs in the CaExPA precursor lesion pleomorphic adenoma (PA) and indicates a higher risk of progression from benign to malignant tumor. Our results demonstrate that diverse mechanisms including WIF1 promoter hypermethylation and loss of heterozygosity contribute to WIF1 downregulation in human salivary gland tumors. In accordance with a crucial role in suppressing salivary gland tumor progression, WIF1 re-expression in salivary gland tumor cells inhibited cell proliferation, induced more differentiated phenotype and promoted cellular senescence, possibly through upregulation of tumor-suppressor genes, such as p53 and p21. Most importantly, WIF1 significantly diminished the number of salivary gland cancer stem cells and the anchorage-independent cell growth. Consistent with this observation, WIF1 caused a reduction in the expression of pluripotency and stemness markers (OCT4 and c-MYC), as well as adult stem cell self-renewal and multi-lineage differentiation markers, such as WNT3A, TCF4, c-KIT and MYB. Furthermore, WIF1 significantly increased the expression of microRNAs pri-let-7a and pri-miR-200c, negative regulators of stemness and cancer progression. In addition, we show that WIF1 functions as a positive regulator of miR-200c, leading to downregulation of BMI1, ZEB1 and ZEB2, with a consequent increase in downstream targets such as E-cadherin. Our study emphasizes the prognostic and therapeutic potential of WIF1 in human salivary gland CaExPA. Moreover, our findings demonstrate a novel mechanism by which WIF1 regulates cancer stemness and senescence, which might have major implications in the field of cancer biology.


Assuntos
Proteínas Adaptadoras de Transdução de Sinal/metabolismo , Adenoma Pleomorfo/metabolismo , Biomarcadores Tumorais/metabolismo , Carcinoma/metabolismo , Senescência Celular , Células-Tronco Neoplásicas/metabolismo , Proteínas Repressoras/metabolismo , Neoplasias das Glândulas Salivares/metabolismo , Via de Sinalização Wnt , Proteínas Adaptadoras de Transdução de Sinal/genética , Adenoma Pleomorfo/genética , Adenoma Pleomorfo/patologia , Biomarcadores Tumorais/genética , Carcinoma/genética , Carcinoma/patologia , Pontos de Checagem do Ciclo Celular , Proteínas de Ciclo Celular/genética , Proteínas de Ciclo Celular/metabolismo , Linhagem Celular Tumoral , Proliferação de Células , Metilação de DNA , Progressão da Doença , Regulação para Baixo , Regulação Neoplásica da Expressão Gênica , Humanos , Perda de Heterozigosidade , MicroRNAs/metabolismo , Células-Tronco Neoplásicas/patologia , Regiões Promotoras Genéticas , Proteínas Repressoras/genética , Neoplasias das Glândulas Salivares/genética , Neoplasias das Glândulas Salivares/patologia , Fatores de Tempo , Transfecção
8.
Biochim Biophys Acta ; 1832(8): 1149-58, 2013 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-23579073

RESUMO

Elevated levels of homocysteine produce detrimental effects in humans but its role in preterm birth is not known. Here we used a mouse model of hyperhomocysteinemia to examine the relevance of homocysteine to preterm birth. The mouse carries a heterozygous deletion of cystathionine ß-synthase (Cbs(+/-)). Gestational period was monitored in wild type and Cbs(+/-) female mice. Mouse uterine and placental tissues, human primary trophoblast cells, and human myometrial and placental cell lines were used to determine the influence of homocysteine on expression of specific genes in vitro. The activity of BKCa channel in the myometrial cell line was monitored using the patch-clamp technique. We found that hyperhomocysteinemia had detrimental effects on pregnancy and induced preterm birth in mice. Homocysteine increased the expression of oxytocin receptor and Cox-2 as well as PGE2 production in uterus and placenta, and initiated premature uterine contraction. A Cox-2 inhibitor reversed these effects. Gpr109a, a receptor for niacin, induced Cox-2 in uterus. Homocysteine upregulated GPR109A and suppressed BKCa channel activity in human myometrial cells. Deletion of Gpr109a in Cbs(+/-) mice reversed premature birth. We conclude that hyperhomocysteinemia causes preterm birth in mice through upregulation of the Gpr109a/Cox-2/PGE2 axis and that pharmacological blockade of Gpr109a may have potential in prevention of preterm birth.


Assuntos
Homocisteína/sangue , Hiper-Homocisteinemia/fisiopatologia , Complicações na Gravidez/sangue , Nascimento Prematuro/sangue , Animais , Linhagem Celular , Ciclo-Oxigenase 2/genética , Ciclo-Oxigenase 2/metabolismo , Inibidores de Ciclo-Oxigenase 2/farmacologia , Cistationina beta-Sintase/genética , Cistationina beta-Sintase/metabolismo , Dinoprostona/genética , Dinoprostona/metabolismo , Feminino , Homocisteína/genética , Homocisteína/metabolismo , Humanos , Hiper-Homocisteinemia/genética , Hiper-Homocisteinemia/metabolismo , Subunidades alfa do Canal de Potássio Ativado por Cálcio de Condutância Alta/genética , Subunidades alfa do Canal de Potássio Ativado por Cálcio de Condutância Alta/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Contração Muscular , Miométrio/metabolismo , Miométrio/fisiopatologia , Placenta/metabolismo , Placenta/fisiopatologia , Gravidez , Complicações na Gravidez/genética , Complicações na Gravidez/fisiopatologia , Nascimento Prematuro/genética , Nascimento Prematuro/fisiopatologia , Receptores Nicotínicos/genética , Receptores Nicotínicos/metabolismo , Receptores de Ocitocina/genética , Receptores de Ocitocina/metabolismo , Trofoblastos/metabolismo , Regulação para Cima , Útero/metabolismo , Útero/patologia , Útero/fisiopatologia
9.
Oncogene ; 30(38): 4026-37, 2011 Sep 22.
Artigo em Inglês | MEDLINE | ID: mdl-21499304

RESUMO

There has been growing interest among the public and scientists in dichloroacetate (DCA) as a potential anticancer drug. Credible evidence exists for the antitumor activity of this compound, but high concentrations are needed for significant therapeutic effect. Unfortunately, these high concentrations produce detrimental side effects involving the nervous system, thereby precluding its use for cancer treatment. The mechanistic basis of the compound's antitumor activity is its ability to activate the pyruvate dehydrogenase complex through inhibition of pyruvate dehydrogenase kinase. As the compound inhibits the kinase at micromolar concentrations, it is not known why therapeutically prohibitive high doses are needed for suppression of tumor growth. We hypothesized that lack of effective mechanisms for the entry of DCA into tumor cells may underlie this phenomenon. Here we show that SLC5A8 transports DCA very effectively with high affinity. This transporter is expressed in normal cells, but expression is silenced in tumor cells by epigenetic mechanisms. The lack of the transporter makes tumor cells resistant to the antitumor activity of DCA. However, if the transporter is expressed in tumor cells ectopically, the cells become sensitive to the drug at low concentrations. This is evident in breast cancer cells, colon cancer cells and prostate cancer cells. Normal cells, which constitutively express the transporter, are however not affected by the compound, indicating tumor cell-selective therapeutic activity. The mechanism of the compound's antitumor activity still remains its ability to inhibit pyruvate dehydrogenase kinase and force mitochondrial oxidation of pyruvate. As silencing of SLC5A8 in tumors involves DNA methylation and its expression can be induced by treatment with DNA methylation inhibitors, our findings suggest that combining DCA with a DNA methylation inhibitor would offer a means to reduce the doses of DCA to avoid detrimental effects associated with high doses but without compromising antitumor activity.


Assuntos
Antineoplásicos/farmacologia , Proteínas de Transporte de Cátions/fisiologia , Ácido Dicloroacético/farmacologia , Proteínas Supressoras de Tumor/fisiologia , Animais , Apoptose/efeitos dos fármacos , Linhagem Celular Tumoral , Ácido Dicloroacético/farmacocinética , Humanos , Proteínas de Membrana Transportadoras/fisiologia , Transportadores de Ácidos Monocarboxílicos , Ácido Pirúvico/metabolismo , Sódio/metabolismo , Xenopus laevis
10.
Protein J ; 29(6): 417-26, 2010 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-20665236

RESUMO

Antibodies to the solute carrier protein, CTL2/SLC44A2, cause hearing loss in animals, are frequently found in autoimmune hearing loss patients, and are implicated in transfusion-related acute lung injury. We cloned a novel CTL2/SLC44A2 isoform (CTL2 P1) from inner ear and identified an alternate upstream promoter and exon 1a encoding a protein of 704 amino acids which differs in the first 10-12 amino acids from the known exon 1b isoform (CTL2 P2; 706 amino acids). The expression of these CTL2/SLC44A2 isoforms, their posttranslational modifications in tissues and their localization in HEK293 cells expressing rHuCTL2/SLC44A2 were assessed. P1 and P2 isoforms with differing glycosylation are variably expressed in cochlea, tongue, heart, colon, lung, kidney, liver and spleen suggesting tissue specific differences that may influence function in each tissue. Because antibodies to CTL2/SLC44A2 have serious pathologic consequences, it is important to understand its distribution and modifications. Heterologous expression in X. laevis oocytes shows that while human CTL2-P1 does not transport choline, human CTL2-P2 exhibits detectable choline transport activity.


Assuntos
Glicoproteínas de Membrana/metabolismo , Proteínas de Membrana Transportadoras/metabolismo , Animais , Antígenos CD/biossíntese , Antígenos CD/química , Antígenos CD/genética , Antígenos CD/metabolismo , Doenças Autoimunes , Linhagem Celular , Simulação por Computador , Orelha Interna/metabolismo , Glicosilação , Cobaias , Perda Auditiva , Humanos , Imuno-Histoquímica , Glicoproteínas de Membrana/biossíntese , Glicoproteínas de Membrana/química , Glicoproteínas de Membrana/genética , Proteínas de Membrana Transportadoras/biossíntese , Proteínas de Membrana Transportadoras/química , Proteínas de Membrana Transportadoras/genética , Camundongos , Microscopia de Fluorescência , Proteínas de Transporte de Cátions Orgânicos/biossíntese , Proteínas de Transporte de Cátions Orgânicos/química , Proteínas de Transporte de Cátions Orgânicos/genética , Proteínas de Transporte de Cátions Orgânicos/metabolismo , Isoformas de Proteínas , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Distribuição Tecidual
11.
Oncogene ; 29(23): 3423-34, 2010 Jun 10.
Artigo em Inglês | MEDLINE | ID: mdl-20348951

RESUMO

Activation of cGMP-dependent protein kinase (PKG) has anti-tumor effects in colon cancer cells but the mechanisms are not fully understood. This study has examined the regulation of beta-catenin/TCF signaling, as this pathway has been highlighted as central to the anti-tumor effects of PKG. We show that PKG activation in SW620 cells results in reduced beta-catenin expression and a dramatic inhibition of TCF-dependent transcription. PKG did not affect protein stability, nor did it increase phosphorylation of the amino-terminal Ser33/37/Thr41 residues that are known to target beta-catenin for degradation. However, we found that PKG potently inhibited transcription from a luciferase reporter driven by the human CTNNB1 promoter, and this corresponded to reduced beta-catenin mRNA levels. Although PKG was able to inhibit transcription from both the CTNNB1 and TCF reporters, the effect on protein levels was less consistent. Ectopic PKG had a marginal effect on beta-catenin protein levels in SW480 and HCT116 but was able to inhibit TCF-reporter activity by over 80%. Investigation of alternative mechanisms revealed that cJun-N-terminal kinase (JNK) activation was required for the PKG-dependent regulation of TCF activity. PKG activation caused beta-catenin to bind to FOXO4 in colon cancer cells, and this required JNK. Activation of PKG was also found to increase the nuclear content of FOXO4 and increase the expression of the FOXO target genes MnSOD and catalase. FOXO4 activation was required for the inhibition of TCF activity as FOXO4-specific short-interfering RNA completely blocked the inhibitory effect of PKG. These data illustrate a dual-inhibitory effect of PKG on TCF activity in colon cancer cells that involves reduced expression of beta-catenin at the transcriptional level, and also beta-catenin sequestration by FOXO4 activation.


Assuntos
Neoplasias do Colo/metabolismo , Proteínas Quinases Dependentes de GMP Cíclico/fisiologia , Transdução de Sinais/fisiologia , Fatores de Transcrição TCF/fisiologia , Fatores de Transcrição/fisiologia , beta Catenina/fisiologia , Proteínas de Ciclo Celular , Linhagem Celular Tumoral , Fatores de Transcrição Forkhead , Quinase 3 da Glicogênio Sintase/fisiologia , Glicogênio Sintase Quinase 3 beta , Humanos , Proteínas Quinases JNK Ativadas por Mitógeno/fisiologia , Fatores de Transcrição TCF/antagonistas & inibidores , Fatores de Transcrição TCF/genética , beta Catenina/antagonistas & inibidores , beta Catenina/genética
12.
Eur Neuropsychopharmacol ; 18(3): 222-9, 2008 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-17728108

RESUMO

Cholinergic neurotransmission depends on the integrity of nicotinic acetylcholine receptors (nAChRs), and impairment of both is characteristic for various neurodegenerative diseases. Visualization of specific receptor subtypes by positron emission tomography (PET) has potential to assist with diagnosis of such neurodegenerative diseases and with design of suitable therapeutic approaches. The goal of our study was to evaluate in vivo the potential of (18)F-labelled (+)- and (-)-norchloro-fluoro-homoepibatidine ([(18)F]NCFHEB) in comparison to 2-[(18)F]F-A-85380 as PET tracers. In the brains of NMRI mice, highest levels of radioactivity were detected at 20 min post-injection of (+)-[(18)F]NCFHEB, (-)-[(18)F]NCFHEB, and 2-F-[(18)F]-A-85380 (7.45, 5.60, and 3.2% ID/g tissue, respectively). No marked pharmacological adverse effects were observed at 25 mug NCFHEB/kg. Uptake studies in RBE4 cells and in situ perfusion studies suggest an interaction of epibatidine and NCFHEB with the carrier-mediated choline transport at the blood-brain barrier. The data indicate that (+)- and (-)-[(18)F]NCFHEB have potential for further development as PET tracers.


Assuntos
Benzamidas , Compostos Bicíclicos Heterocíclicos com Pontes , Compostos Radiofarmacêuticos , Receptores Nicotínicos/metabolismo , Animais , Azetidinas , Benzamidas/química , Benzamidas/farmacocinética , Transporte Biológico Ativo , Barreira Hematoencefálica/efeitos dos fármacos , Compostos Bicíclicos Heterocíclicos com Pontes/química , Compostos Bicíclicos Heterocíclicos com Pontes/farmacocinética , Colina/metabolismo , Feminino , Indicadores e Reagentes , Marcação por Isótopo , Masculino , Camundongos , Perfusão , Tomografia por Emissão de Pósitrons , Compostos Radiofarmacêuticos/química , Compostos Radiofarmacêuticos/farmacocinética , Estereoisomerismo , Distribuição Tecidual
13.
Biochim Biophys Acta ; 1768(11): 2690-7, 2007 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-17692818

RESUMO

Recently, we cloned two Na(+)-coupled lactate transporters from mouse kidney, a high-affinity transporter (SMCT1 or slc5a8) and a low-affinity transporter (SMCT2 or slc5a12). Here we report on the cloning and functional characterization of human SMCT2 (SLC5A12) and compare the immunolocalization patterns of slc5a12 and slc5a8 in mouse kidney. The human SMCT2 cDNA codes for a protein consisting of 618 amino acids. When expressed in mammalian cells or Xenopus oocytes, human SMCT2 mediates Na(+) -coupled transport of lactate, pyruvate and nicotinate. The affinities of the transporter for these substrates are lower than those reported for human SMCT1. Several non-steroidal anti-inflammatory drugs inhibit human SMCT2-mediated nicotinate transport, suggesting that NSAIDs interact with the transporter as they do with human SMCT1. Immunofluorescence microscopy of mouse kidney sections with an antibody specific for SMCT2 shows that the transporter is expressed predominantly in the cortex. Similar studies with an anti-SMCT1 antibody demonstrate that SMCT1 is also expressed mostly in the cortex. Dual-labeling of SMCT1 and SMCT2 with 4F2hc (CD98), a marker for basolateral membrane of proximal tubular cells in the S1 and S2 segments of the nephron, shows that both SMCT1 and SMCT2 are expressed in the apical membrane of the tubular cells. These studies also show that while SMCT2 is broadly expressed along the entire length of the proximal tubule (S1/S2/S3 segments), the expression of SMCT1 is mostly limited to the S3 segment. These studies suggest that the low-affinity transporter SMCT2 initiates lactate absorption in the early parts of the proximal tubule followed by the participation of the high-affinity transporter SMCT1 in the latter parts of the proximal tubule.


Assuntos
Rim/química , Transportadores de Ácidos Monocarboxílicos/análise , Transportadores de Ácidos Monocarboxílicos/genética , Sequência de Aminoácidos , Animais , Clonagem Molecular , DNA Complementar/química , Imunofluorescência , Humanos , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Dados de Sequência Molecular , Transportadores de Ácidos Monocarboxílicos/fisiologia , Simportadores , Xenopus laevis
14.
Placenta ; 28(10): 1082-90, 2007 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-17559929

RESUMO

Antenatal steroids like dexamethasone (DEX) are used to augment fetal lung maturity and there is a major concern that they impair fetal growth. If delivery is delayed after using antenatal DEX, placental function and hence fetal growth may be compromised even further. To investigate the effects of DEX on placental function, we treated 9 pregnant C57/BL6 mice with DEX and 9 pregnant mice were injected with saline to serve as controls. Placental gene expression was studied using microarrays in 3 pairs and other 6 pairs were used to confirm microarray results by semi-quantitative RT-PCR, real-time PCR, in situ hybridization, western blot analysis and Oligo ApopTaq assay. DEX-treated placentas were hydropic, friable, pale, and weighed less (80.0+/-15.1mg compared to 85.6.8+/-7.6mg, p=0.05) (n=62 placentas). Fetal weight was significantly reduced after DEX use (940+/-32mg compared to 1162+/-79mg, p=0.001) (n=62 fetuses). There was >99% similarity within and between the three gene chip data sets. DEX led to down-regulation of 1212 genes and up-regulation of 1382 genes. RT-PCR studies showed that DEX caused a decrease in expression of genes involved in cell division such as cyclins A2, B1, D2, cdk 2, cdk 4 and M-phase protein kinase along with growth-promoting genes such as EGF-R, BMP4 and IGFBP3. Oligo ApopTaq assay and western blot studies showed that DEX-treatment increased apoptosis of trophoblast cells. DEX-treatment led to up-regulation of aquaporin 5 and tryptophan hydroxylase genes as confirmed by real-time PCR, and in situ hybridization studies. Thus antenatal DEX treatment led to a reduction in placental and fetal weight, and this effect was associated with a decreased expression of several growth-promoting genes and increased apoptosis of trophoblast cells.


Assuntos
Apoptose/efeitos dos fármacos , Dexametasona/farmacologia , Regulação da Expressão Gênica no Desenvolvimento/efeitos dos fármacos , Placenta/efeitos dos fármacos , Placenta/metabolismo , Animais , Aquaporina 5/biossíntese , Caspase 1/biossíntese , Caspase 3/biossíntese , Dexametasona/toxicidade , Regulação para Baixo , Feminino , Retardo do Crescimento Fetal/induzido quimicamente , Camundongos , Camundongos Endogâmicos C57BL , Análise de Sequência com Séries de Oligonucleotídeos , Gravidez , Triptofano Hidroxilase/biossíntese , Regulação para Cima
15.
Exp Eye Res ; 84(1): 191-9, 2007 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-17094966

RESUMO

D-serine, an endogenous co-agonist of NMDA receptors in vertebrate retina, may modulate glutamate sensitivity of retinal neurons. This study determined at the functional and molecular level the transport process responsible for D-serine in retinal Müller cells. RT-PCR and immunoblotting showed that serine racemase (SR), the synthesizing enzyme for D-serine, is expressed in the rMC-1 Müller cell line and primary cultures of mouse Müller cells (1 degrees MCs). The relative contributions of different amino acid transport systems to d-serine uptake were determined based on differential substrate specificities and ion dependencies. D-serine uptake was obligatorily dependent on Na+, eliminating Na+-independent transporters (asc-1 and system L) for D-serine in Müller cells. The Na+:substrate stoichiometry for the transport process was 1:1. D-serine transport was inhibited by alanine, serine, cysteine, glutamine, and asparagine, but not anionic amino acids or cationic amino acids, suggesting that D-serine transport in Müller cells occurs via ASCT2 rather than ASCT1 or ATB0,+. The expression of mRNAs specific for ASCT1, ASCT2, and ATB0,+ was analyzed by RT-PCR confirming the expression of ASCT2 (and ASCT1) mRNA, but not ATB0,+, in Müller cells. Immunoblotting detected ASCT2 in neural retina and in 1 degrees MCs; immunohistochemistry confirmed these data in retinal sections and in cultures of 1 degrees MCs. The efflux of D-serine via ASCT2 by ASCT2 substrates was demonstrable using the Xenopus laevis oocyte heterologous expression system. These data provide the first molecular evidence for SR and ASCT2 expression in a Müller cell line and in 1 degrees MCs and suggest that D-serine, synthesized in Müller cells by SR, is effluxed via ASCT2 to regulate NMDA receptors in adjacent neurons.


Assuntos
Neurotransmissores/metabolismo , Retina/metabolismo , Serina/metabolismo , Sistema ASC de Transporte de Aminoácidos/fisiologia , Sistemas de Transporte de Aminoácidos/fisiologia , Animais , Transporte Biológico/fisiologia , Células Cultivadas , Expressão Gênica , Camundongos , Antígenos de Histocompatibilidade Menor , RNA Mensageiro/genética , Racemases e Epimerases/genética , Racemases e Epimerases/metabolismo , Ratos , Retina/citologia , Reação em Cadeia da Polimerase Via Transcriptase Reversa/métodos , Sódio/fisiologia , Especificidade por Substrato
17.
Cell Tissue Res ; 324(2): 189-202, 2006 May.
Artigo em Inglês | MEDLINE | ID: mdl-16609915

RESUMO

The cystine-glutamate exchanger, system x(c)(-), mediates the Na(+)-independent exchange of cystine into cells, coupled to the efflux of intracellular glutamate. System x(c)(-) plays a critical role in glutathione homeostasis. Early studies of brain suggested that system x(c)(-) was present primarily in astrocytes but not neurons. More recent work indicates that certain brain neurons have an active system x(c)(-). In the retina, system x(c)(-) has been demonstrated in Müller and retinal pigment epithelial cells. We have recently suggested that two protein components of system x(c)(-), xCT and 4F2hc, are present in ganglion cells of the intact retina. Here, we have used (1) molecular and immunohistochemical assays to determine whether system x(c)(-) is present in primary ganglion cells isolated from neonatal mouse retinas and (2) functional assays to determine whether its activity is regulated by oxidative stress in a retinal ganglion cell line (RGC-5). Primary mouse ganglion cells and RGC-5 cells express xCT and 4F2hc. RGC-5 cells take up [(3)H]glutamate in the absence of Na(+), and this uptake is blocked by known substrates of system x(c)(-) (glutamate, cysteine, cystine, quisqualic acid). Treatment of RGC-5 cells with NO and reactive oxygen species donors leads to increased activity of system x(c)(-) associated with an increase in the maximal velocity of the transporter with no significant change in the substrate affinity. This is the first report of system x(c)(-) in primary retinal ganglion cells and RGC-5 cells. Oxidative stress upregulates this transport system in RGC-5 cells, and the process is associated with an increase in xCT mRNA and protein but no change in 4F2hc mRNA or protein.


Assuntos
Sistema y+ de Transporte de Aminoácidos/genética , Sistema y+ de Transporte de Aminoácidos/metabolismo , Óxido Nítrico/farmacologia , Células Ganglionares da Retina/efeitos dos fármacos , Células Ganglionares da Retina/metabolismo , Animais , Antioxidantes/metabolismo , Sequência de Bases , Linhagem Celular , Células Cultivadas , Primers do DNA/genética , Cadeia Pesada da Proteína-1 Reguladora de Fusão/genética , Cadeia Pesada da Proteína-1 Reguladora de Fusão/metabolismo , Regulação da Expressão Gênica/efeitos dos fármacos , Glutationa/metabolismo , Cinética , Camundongos , Doadores de Óxido Nítrico/farmacologia , Estresse Oxidativo , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Ratos , Espécies Reativas de Oxigênio/farmacologia , S-Nitroso-N-Acetilpenicilamina/farmacologia
18.
Placenta ; 27(6-7): 550-9, 2006.
Artigo em Inglês | MEDLINE | ID: mdl-16129486

RESUMO

NaS2 is a Na+-coupled transporter for sulfate that belongs to the SLC13 gene family. This transporter was originally cloned from high endothelial venule endothelial cells, but nothing is known about the functional characteristics of this transporter except that it transports sulfate in a Na+-coupled manner. Northern blot analysis indicates that NaS2 is expressed most robustly in placenta. In the present study, we cloned NaS2 from rat placenta and characterized its transport function in detail using the Xenopus laevis oocyte expression system. Rat NaS2 consists of 629 amino acids and is highly similar to human NaS2. In situ hybridization studies with mouse placental sections show that NaS2 transcripts are expressed primarily in trophoblasts of the labyrinth zone. The expression of the transporter is confirmed in primary cultures of trophoblasts isolated from human placenta. When expressed in X. laevis oocytes, rat NaS2 mediates Na+-coupled transport of sulfate. The transport of sulfate is inhibited by oxyanions of selenium, chromium, arsenic, molybdenum, and phosphorous, suggesting that the transporter may mediate the transport of these oxyanions in addition to sulfate. The Kt for sulfate is 153+/-30 microM and the Na+:sulfate stoichiometry is 3:1. The transport process is electrogenic as evidenced from the inhibition of the uptake process by K+-induced depolarization. We conclude that NaS2 is a placenta-specific Na+-coupled, electrogenic, transporter for sulfate expressed in trophoblasts and that it is also responsible for the transport of oxyanions of the micronutrients selenium and chromium.


Assuntos
Ânions/metabolismo , Cromo/metabolismo , Transportadores de Ânions Orgânicos Dependentes de Sódio/metabolismo , Selênio/metabolismo , Sulfatos/metabolismo , Simportadores/metabolismo , Trofoblastos/metabolismo , Sequência de Aminoácidos , Animais , Biblioteca Gênica , Humanos , Hibridização In Situ , Dados de Sequência Molecular , Análise de Sequência com Séries de Oligonucleotídeos , RNA Mensageiro/metabolismo , Ratos , Homologia de Sequência de Aminoácidos , Transportadores de Sulfato , Xenopus laevis
19.
Artigo em Inglês | MEDLINE | ID: mdl-16375689

RESUMO

ATB(0,+) is a unique amino acid transporter because of its broad substrate specificity and concentrative ability. This transporter recognizes neutral as well as cationic amino acids. It is energized by Na(+) and Cl(-) gradients and membrane potential. Many of the amino acids and amino acid derivatives that are substrates for ATB(0,+) serve as therapeutic agents (e.g., D-serine, carnitine, and nitric oxide synthase inhibitors). Recent studies have shown that the potential of ATB(0,+) as a drug delivery system may be greater than previously envisaged. ATB(0,+) can transport antiviral drugs such as acyclovir and ganciclovir when they are covalently coupled to the side chain of anionic amino acids. Chemical modification of the carboxyl groups in the side chain of aspartate and glutamate with drugs converts these anionic amino acids into neutral amino acid derivatives. Therefore, the modified drugs are recognized by ATB(0,+). Interestingly, even when acyclovir and ganciclovir are coupled as esters with alpha-carboxyl group of neutral amino acids, the modified drugs are transported via ATB(0,+). Similarly, the hydroxyl group in the side chains of serine and threonine can also be used to covalently couple drugs for delivery into cells via ATB(0,+). This increases the potential for designing a wide variety of amino acid-based prodrugs that can utilize ATB(0,+) as drug delivery system. Furthermore, the transporter is expressed in the colon, lung, and eye, the tissues easily amenable for drug delivery. These findings argue strongly in support of ATB(0,+) as a potential delivery system for a wide variety of drugs and prodrugs.


Assuntos
Sistema ASC de Transporte de Aminoácidos/química , Sistemas de Liberação de Medicamentos/métodos , Preparações Farmacêuticas/administração & dosagem , Pró-Fármacos/administração & dosagem , Sistema ASC de Transporte de Aminoácidos/biossíntese , Transporte Biológico , Humanos , Antígenos de Histocompatibilidade Menor , Preparações Farmacêuticas/química , Pró-Fármacos/química , Pró-Fármacos/farmacocinética , Especificidade por Substrato
20.
Biochem Soc Trans ; 33(Pt 1): 237-40, 2005 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-15667316

RESUMO

SLC5A8 is a candidate tumour suppressor gene that is silenced in colon cancer, gastric cancer and possibly other cancers in humans. This gene codes for a transporter belonging to the Na(+)/glucose co-transporter gene family (SLC5). The cancer-associated silencing of the gene involves hypermethylation of CpG islands present in exon 1 of the gene. SLC5A8 is expressed in colon, ileum, kidney and thyroid gland. The protein coded by the gene mediates the Na(+)-coupled and electrogenic transport of a variety of monocarboxylates, including short-chain fatty acids, lactate and nicotinate. It may also transport iodide. The normal physiological function of this transporter in the intestinal tract and kidney is likely to facilitate the active absorption of short-chain fatty acids, lactate and nicotinate. One of the short-chain fatty acids that serves as a substrate for SLC5A8 is butyrate. This fatty acid is an inhibitor of histone deacetylases and is known to induce apoptosis in a variety of tumours including colonic tumour. Since butyrate is produced in the colonic lumen at high concentrations by bacterial fermentation of dietary fibre, we speculate that the ability of SLC5A8 to mediate the entry of this short-chain fatty acid into colonic epithelial cells underlies the potential tumour suppressor function of this transporter.


Assuntos
Proteínas de Transporte de Cátions/fisiologia , Genes Supressores de Tumor , Proteínas de Transporte de Cátions/genética , Inativação Gênica , Humanos , Transportadores de Ácidos Monocarboxílicos
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA