Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 13 de 13
Filtrar
Mais filtros

Base de dados
País/Região como assunto
Tipo de documento
País de afiliação
Intervalo de ano de publicação
2.
J Clin Virol ; 167: 105579, 2023 10.
Artigo em Inglês | MEDLINE | ID: mdl-37683299

RESUMO

BACKGROUND: Quantitative polymerase chain reaction (qPCR) for Epstein-Barr virus (EBV)-DNA is an important diagnostic tool for EBV-associated disease, but interpretation of its clinical significance is challenging. OBJECTIVES: We assessed the diagnostic and clinical performance of WHO-standardised qPCR for EBV-DNA (WHO EBV-qPCR) in plasma and whole blood (WB) for proven EBV disease in a prospectively accrued patient cohort. STUDY DESIGN: Central Denmark Region patients, tested with WHO EBV-qPCR from November 2017 to March 2019, were screened for EBV disease. Incidence (IR) was estimated by Poisson regression. Sensitivity, specificity, positive and negative predictive values (PPV, NPV) were calculated for EBV-qPCR in plasma and WB. Risk of diagnostic latency was compared between patients with EBV-positive and EBV-negative lymphomas. RESULTS: EBV disease was diagnosed in 95 of 1484 participants (IR: 16.3 per 1000 patientyears 95%CI; 13.3-19.9). Sensitivity and specificity of WHO EBV-qPCR in plasma was 82.4% (95% CI; 74.2-90.7%) and 87.8% (95% CI; 85.6-90%), yielding a PPV of 32.2% (95% CI; 24.9-39.5%) and NPV of 98.6% (95% CI; 97.7-99.5%) for proven EBV disease. Sensitivity and NPV were comparable in WB, while specificity and PPV decreased to 66.9% (95% CI; 60.6-73.1%) and 18.1% (95% CI; 7.5-28.7%). Risk of diagnostic latency was 2.3-fold (95% CI 1.4-4.1) higher for patients with EBV-positive compared with EBV-negative lymphomas. CONCLUSIONS: WHO EBV-qPCR in plasma and WB have a low PPV but a high NPV for proven EBV disease. Implementation of WHO EBV-qPCR could improve interpretation and facilitate EBV-positive lymphoma diagnosis.


Assuntos
Infecções por Vírus Epstein-Barr , Herpesvirus Humano 4 , Humanos , Herpesvirus Humano 4/genética , Infecções por Vírus Epstein-Barr/diagnóstico , Plasma , DNA , Relevância Clínica
4.
BMC Biochem ; 12: 21, 2011 May 17.
Artigo em Inglês | MEDLINE | ID: mdl-21586110

RESUMO

BACKGROUND: The inorganic (Pi) phosphate transporter (PiT) family comprises known and putative Na(+)- or H(+)-dependent Pi-transporting proteins with representatives from all kingdoms. The mammalian members are placed in the outer cell membranes and suggested to supply cells with Pi to maintain house-keeping functions. Alignment of protein sequences representing PiT family members from all kingdoms reveals the presence of conserved amino acids and that bacterial phosphate permeases and putative phosphate permeases from archaea lack substantial parts of the protein sequence when compared to the mammalian PiT family members. Besides being Na(+)-dependent P(i) (NaP(i)) transporters, the mammalian PiT paralogs, PiT1 and PiT2, also are receptors for gamma-retroviruses. We have here exploited the dual-function of PiT1 and PiT2 to study the structure-function relationship of PiT proteins. RESULTS: We show that the human PiT2 histidine, H(502), and the human PiT1 glutamate, E(70),--both conserved in eukaryotic PiT family members--are critical for P(i) transport function. Noticeably, human PiT2 H(502) is located in the C-terminal PiT family signature sequence, and human PiT1 E(70) is located in ProDom domains characteristic for all PiT family members.A human PiT2 truncation mutant, which consists of the predicted 10 transmembrane (TM) domain backbone without a large intracellular domain (human PiT2ΔR(254)-V(483)), was found to be a fully functional P(i) transporter. Further truncation of the human PiT2 protein by additional removal of two predicted TM domains together with the large intracellular domain created a mutant that resembles a bacterial phosphate permease and an archaeal putative phosphate permease. This human PiT2 truncation mutant (human PiT2ΔL(183)-V(483)) did also support P(i) transport albeit at very low levels. CONCLUSIONS: The results suggest that the overall structure of the P(i)-transporting unit of the PiT family proteins has remained unchanged during evolution. Moreover, in combination, our studies of the gene structure of the human PiT1 and PiT2 genes (SLC20A1 and SLC20A2, respectively) and alignment of protein sequences of PiT family members from all kingdoms, along with the studies of the dual functions of the human PiT paralogs show that these proteins are excellent as models for studying the evolution of a protein's structure-function relationship.


Assuntos
Biologia Computacional , Sequência Conservada , Fosfatos/metabolismo , Proteínas Cotransportadoras de Sódio-Fosfato Tipo III/química , Proteínas Cotransportadoras de Sódio-Fosfato Tipo III/metabolismo , Sequência de Aminoácidos , Animais , Transporte Biológico , Células CHO , Linhagem Celular Tumoral , Cricetinae , Cricetulus , Cães , Evolução Molecular , Éxons/genética , Gammaretrovirus/metabolismo , Humanos , Espaço Intracelular/metabolismo , Íntrons/genética , Camundongos , Dados de Sequência Molecular , Estrutura Terciária de Proteína , Alinhamento de Sequência , Deleção de Sequência , Proteínas Cotransportadoras de Sódio-Fosfato Tipo III/genética , Especificidade da Espécie
5.
Clin Genitourin Cancer ; 18(1): 62-68.e2, 2020 02.
Artigo em Inglês | MEDLINE | ID: mdl-31640912

RESUMO

BACKGROUND: Pazopanib can induce liver toxicity in patients with metastatic renal cell carcinoma (mRCC). We assessed the effect of a TA repeat polymorphism in the UGT1A1 (uridine diphosphate glucuronosyltransferase 1A1) gene encoding uridine diphosphate glucuronosyltransferase 1A1 on liver toxicity, dose reductions, and patient outcomes. PATIENTS AND METHODS: Patients with mRCC treated with first-line pazopanib developing liver toxicity underwent genotyping for the UGT1A1 polymorphism. Liver toxicity was assessed using the Common Terminology Criteria for Adverse Events, version 4.0. Progression-free survival and overall survival were assessed using the Kaplan-Meier and log-rank methods. RESULTS: Of 261 patients, 34 (13%) had developed liver toxicity after a median of 29 days (range, 5-155 days). Grade 4, 3, and 2 alanine aminotransferase or bilirubin had increased in 2 (6%), 17 (50%), and 8 (24%) patients, respectively. The UGT1A1 assessment demonstrated that 18 patients (53%) had TA6/TA7, 7 (21%) had TA7/TA7, and 9 (26%) had wild-type TA6/TA6. The UGT1A1 polymorphism was associated with improved median progression-free survival (TA6/TA6, 5.5 months; TA6/TA7, 34.2 months; TA7/TA7, 22.3 months; unknown UGT1A1 status, 9.2 months; UGT1A1 polymorphisms combined vs. unknown status, P = .021). UGT1A1 polymorphism was associated with improved median overall survival (TA6/TA6, 8.1 months, TA6/TA7 or TA7/TA7 not reached, unknown UGT1A1 status, 16.6 months; UGT1A1 polymorphisms combined vs. unknown status, P = .033). Patients with UGT1A1 polymorphism safely resumed pazopanib at ultra-low doses determined by the degree of liver toxicity and UGT1A1 polymorphism. CONCLUSIONS: UGT1A1 polymorphisms were associated with improved outcomes, despite pazopanib interruption and dose reductions. UGT1A1 assessment could improve the management of pazopanib-induced liver toxicity in patients with mRCC.


Assuntos
Inibidores da Angiogênese/efeitos adversos , Carcinoma de Células Renais/tratamento farmacológico , Doença Hepática Induzida por Substâncias e Drogas/epidemiologia , Glucuronosiltransferase/genética , Neoplasias Renais/tratamento farmacológico , Pirimidinas/efeitos adversos , Sulfonamidas/efeitos adversos , Adulto , Idoso , Idoso de 80 Anos ou mais , Inibidores da Angiogênese/administração & dosagem , Carcinoma de Células Renais/genética , Carcinoma de Células Renais/mortalidade , Doença Hepática Induzida por Substâncias e Drogas/diagnóstico , Doença Hepática Induzida por Substâncias e Drogas/genética , Relação Dose-Resposta a Droga , Feminino , Seguimentos , Humanos , Indazóis , Estimativa de Kaplan-Meier , Neoplasias Renais/genética , Neoplasias Renais/mortalidade , Testes de Função Hepática , Estudos Longitudinais , Masculino , Pessoa de Meia-Idade , Polimorfismo Genético , Intervalo Livre de Progressão , Estudos Prospectivos , Pirimidinas/administração & dosagem , Estudos Retrospectivos , Sulfonamidas/administração & dosagem
6.
Sci Rep ; 6: 31972, 2016 08 23.
Artigo em Inglês | MEDLINE | ID: mdl-27549929

RESUMO

The Na(+)/K(+)-ATPases maintain Na(+) and K(+) electrochemical gradients across the plasma membrane, a prerequisite for electrical excitability and secondary transport in neurons. Autosomal dominant mutations in the human ATP1A3 gene encoding the neuron-specific Na(+)/K(+)-ATPase α3 isoform cause different neurological diseases, including rapid-onset dystonia-parkinsonism (RDP) and alternating hemiplegia of childhood (AHC) with overlapping symptoms, including hemiplegia, dystonia, ataxia, hyperactivity, epileptic seizures, and cognitive deficits. Position D801 in the α3 isoform is a mutational hotspot, with the D801N, D801E and D801V mutations causing AHC and the D801Y mutation causing RDP or mild AHC. Despite intensive research, mechanisms underlying these disorders remain largely unknown. To study the genotype-to-phenotype relationship, a heterozygous knock-in mouse harboring the D801Y mutation (α3(+/D801Y)) was generated. The α3(+/D801Y) mice displayed hyperactivity, increased sensitivity to chemically induced epileptic seizures and cognitive deficits. Interestingly, no change in the excitability of CA1 pyramidal neurons in the α3(+/D801Y) mice was observed. The cognitive deficits were rescued by administration of the benzodiazepine, clonazepam, a GABA positive allosteric modulator. Our findings reveal the functional significance of the Na(+)/K(+)-ATPase α3 isoform in the control of spatial learning and memory and suggest a link to GABA transmission.


Assuntos
Transtornos Cognitivos/genética , Mutação , Convulsões/genética , ATPase Trocadora de Sódio-Potássio/genética , Animais , Região CA1 Hipocampal/metabolismo , Linhagem Celular , Transtornos Cognitivos/psicologia , Modelos Animais de Doenças , Técnicas de Introdução de Genes , Humanos , Masculino , Memória , Camundongos , Fenótipo , Convulsões/induzido quimicamente , ATPase Trocadora de Sódio-Potássio/metabolismo , Aprendizagem Espacial
7.
Sci Rep ; 6: 22047, 2016 Feb 25.
Artigo em Inglês | MEDLINE | ID: mdl-26911348

RESUMO

Migraine is a complex brain disorder, and understanding the complexity of this prevalent disease could improve quality of life for millions of people. Familial Hemiplegic Migraine type 2 (FHM2) is a subtype of migraine with aura and co-morbidities like epilepsy/seizures, cognitive impairments and psychiatric manifestations, such as obsessive-compulsive disorder (OCD). FHM2 disease-mutations locate to the ATP1A2 gene encoding the astrocyte-located α2-isoform of the sodium-potassium pump (α2Na(+)/K(+)-ATPase). We show that knock-in mice heterozygous for the FHM2-associated G301R-mutation (α2(+/G301R)) phenocopy several FHM2-relevant disease traits e.g., by mimicking mood depression and OCD. In vitro studies showed impaired glutamate uptake in hippocampal mixed astrocyte-neuron cultures from α2(G301R/G301R) E17 embryonic mice, and moreover, induction of cortical spreading depression (CSD) resulted in reduced recovery in α2(+/G301R) male mice. Moreover, NMDA-type glutamate receptor antagonists or progestin-only treatment reverted specific α2(+/G301R) behavioral phenotypes. Our findings demonstrate that studies of an in vivo relevant FHM2 disease knock-in mouse model provide a link between the female sex hormone cycle and the glutamate system and a link to co-morbid psychiatric manifestations of FHM2.


Assuntos
Ácido Glutâmico/metabolismo , Enxaqueca com Aura/genética , Enxaqueca com Aura/metabolismo , Mutação , Fenótipo , Estimulação Acústica , Animais , Comportamento Animal , Transporte Biológico , Circulação Cerebrovascular , Biologia Computacional/métodos , Depressão Alastrante da Atividade Elétrica Cortical/genética , Modelos Animais de Doenças , Feminino , Hormônios Esteroides Gonadais/metabolismo , Masculino , Camundongos , Camundongos Transgênicos , Enxaqueca com Aura/diagnóstico , Enxaqueca com Aura/tratamento farmacológico , Atividade Motora , Tempo de Reação , ATPase Trocadora de Sódio-Potássio/genética , Estresse Fisiológico
8.
FEBS J ; 272(12): 3060-74, 2005 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-15955065

RESUMO

The mammalian members of the inorganic phosphate (P(i)) transporter (PiT) family, the type III sodium-dependent phosphate (NaP(i)) transporters PiT1 and PiT2, have been assigned housekeeping P(i) transport functions and are suggested to be involved in chondroblastic and osteoblastic mineralization and ectopic calcification. The PiT family members are conserved throughout all kingdoms and use either sodium (Na+) or proton (H+) gradients to transport P(i). Sequence logo analyses revealed that independent of their cation dependency these proteins harbor conserved signature sequences in their N- and C-terminal ends with the common core consensus sequence GANDVANA. With the exception of 10 proteins from extremophiles all 109 proteins analyzed carry an aspartic acid in one or both of the signature sequences. We changed either of the highly conserved aspartates, Asp28 and Asp506, in the N- and C-terminal signature sequences, respectively, of human PiT2 to asparagine and analyzed P(i) uptake function in Xenopus laevis oocytes. Both mutant proteins were expressed at the cell surface of the oocytes but exhibited knocked out NaP(i) transport function. Human PiT2 is also a retroviral receptor and we have previously shown that this function can be exploited as a control for proper processing and folding of mutant proteins. Both mutant transporters displayed wild-type receptor functions implying that their overall architecture is undisturbed. Thus the presence of an aspartic acid in either of the PiT family signature sequences is critical for the Na+-dependent P(i) transport function of human PiT2. The conservation of the aspartates among proteins using either Na+- or H+-gradients for P(i) transport suggests that they are involved in H+-dependent P(i) transport as well. Current results favor a membrane topology model in which the N- and C-terminal PiT family signature sequences are positioned in intra- and extracellular loops, respectively, suggesting that they are involved in related functions on either side of the membrane. The present data are in agreement with a possible role of the signature sequences in translocation of cations.


Assuntos
Ácido Aspártico/genética , Simportadores/metabolismo , Sequência de Aminoácidos , Substituição de Aminoácidos , Animais , Asparagina/genética , Proteínas de Bactérias/genética , Transporte Biológico , Células CHO/virologia , Proteínas de Transporte/genética , Células Cultivadas , Sequência Conservada , Cricetinae , Cricetulus , Evolução Molecular , Feminino , Humanos , Vírus da Leucemia Murina/metabolismo , Vírus da Leucemia Murina/patogenicidade , Proteínas de Membrana/genética , Dados de Sequência Molecular , Família Multigênica , Mutação , Oócitos/metabolismo , Proteínas Cotransportadoras de Sódio-Fosfato , Proteínas Cotransportadoras de Sódio-Fosfato Tipo III , Simportadores/genética
9.
Neurosci Biobehav Rev ; 36(2): 855-71, 2012 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-22067897

RESUMO

The two autosomal dominantly inherited neurological diseases: familial hemiplegic migraine type 2 (FHM2) and familial rapid-onset of dystonia-parkinsonism (Familial RDP) are caused by in vivo mutations of specific alpha subunits of the sodium-potassium pump (Na(+)/K(+)-ATPase). Intriguingly, patients with classical FHM2 and RDP symptoms additionally suffer from other manifestations, such as epilepsy/seizures and developmental disabilities. Recent studies of FHM2 and RDP mouse models provide valuable tools for dissecting the vital roles of the Na(+)/K(+)-ATPases, and we discuss their relevance to the complex patient symptoms and manifestations. Thus, it is interesting that mouse models targeting a specific α-isoform cause different, although still comparable, phenotypes consistent with classical symptoms and other manifestations observed in FHM2 and RDP patients. This review highlights that use of mouse models have broad potentials for future research concerning migraine and dystonia-related diseases, which will contribute towards understanding the, yet unknown, pathophysiologies.


Assuntos
Distonia/genética , Transtornos de Enxaqueca/genética , Transtornos de Enxaqueca/fisiopatologia , Mutação/genética , ATPase Trocadora de Sódio-Potássio/genética , Animais , Distonia/fisiopatologia , Humanos , Camundongos
10.
J Comp Neurol ; 519(2): 376-404, 2011 Feb 01.
Artigo em Inglês | MEDLINE | ID: mdl-21165980

RESUMO

The Na(+)/K(+)-ATPase1 alpha subunit 3 (ATP1α(3)) is one of many essential components that maintain the sodium and potassium gradients across the plasma membrane in animal cells. Mutations in the ATP1A3 gene cause rapid-onset of dystonia parkinsonism (RDP), a rare movement disorder characterized by sudden onset of dystonic spasms and slowness of movement. To achieve a better understanding of the pathophysiology of the disease, we used immunohistochemical approaches to describe the regional and cellular distribution of ATP1α(3) in the adult mouse brain. Our results show that localization of ATP1α(3) is restricted to neurons, and it is expressed mostly in projections (fibers and punctuates), but cell body expression is also observed. We found high expression of ATP1α(3) in GABAergic neurons in all nuclei of the basal ganglia (striatum, globus pallidus, subthalamic nucleus, and substantia nigra), which is a key circuitry in the fine movement control. Several thalamic nuclei structures harboring connections to and from the cortex expressed high levels of the ATP1α(3) isoform. Other structures with high expression of ATP1α(3) included cerebellum, red nucleus, and several areas of the pons (reticulotegmental nucleus of pons). We also found high expression of ATP1α(3) in projections and cell bodies in hippocampus; most of these ATP1α(3)-positive cell bodies showed colocalization to GABAergic neurons. ATP1α(3) expression was not significant in the dopaminergic cells of substantia nigra. In conclusion, and based on our data, ATP1α(3) is widely expressed in neuronal populations but mainly in GABAergic neurons in areas and nuclei related to movement control, in agreement with RDP symptoms.


Assuntos
Encéfalo/anatomia & histologia , Encéfalo/metabolismo , Isoformas de Proteínas/metabolismo , ATPase Trocadora de Sódio-Potássio/metabolismo , Animais , Encéfalo/fisiopatologia , Distúrbios Distônicos/genética , Distúrbios Distônicos/metabolismo , Distúrbios Distônicos/fisiopatologia , Feminino , Humanos , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Mutação , Isoformas de Proteínas/genética , ATPase Trocadora de Sódio-Potássio/genética
11.
Am J Physiol Cell Physiol ; 291(6): C1377-87, 2006 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-16790504

RESUMO

The general phosphate need in mammalian cells is accommodated by members of the P(i) transport (PiT) family (SLC20), which use either Na(+) or H(+) to mediate inorganic phosphate (P(i)) symport. The mammalian PiT paralogs PiT1 and PiT2 are Na(+)-dependent P(i) (NaP(i)) transporters and are exploited by a group of retroviruses for cell entry. Human PiT1 and PiT2 were characterized by expression in Xenopus laevis oocytes with (32)P(i) as a traceable P(i) source. For PiT1, the Michaelis-Menten constant for P(i) was determined as 322.5 +/- 124.5 microM. PiT2 was analyzed for the first time and showed positive cooperativity in P(i) uptake with a half-maximal activity constant for P(i) of 163.5 +/- 39.8 microM. PiT1- and PiT2-mediated Na(+)-dependent P(i) uptake functions were not significantly affected by acidic and alkaline pH and displayed similar Na(+) dependency patterns. However, only PiT2 was capable of Na(+)-independent P(i) transport at acidic pH. Study of the impact of divalent cations Ca(2+) and Mg(2+) revealed that Ca(2+) was important, but not critical, for NaP(i) transport function of PiT proteins. To gain insight into the NaP(i) cotransport function, we analyzed PiT2 and a PiT2 P(i) transport knockout mutant using (22)Na(+) as a traceable Na(+) source. Na(+) was transported by PiT2 even without P(i) in the uptake medium and also when P(i) transport function was knocked out. This is the first time decoupling of P(i) from Na(+) transport has been demonstrated for a PiT family member. Moreover, the results imply that putative transmembrane amino acids E(55) and E(575) are responsible for linking P(i) import to Na(+) transport in PiT2.


Assuntos
Fosfatos/metabolismo , Proteínas Cotransportadoras de Sódio-Fosfato Tipo III/metabolismo , Sódio/metabolismo , Animais , Transporte Biológico , Cálcio/metabolismo , Enzimas de Restrição do DNA/metabolismo , Humanos , Concentração de Íons de Hidrogênio , Magnésio/metabolismo , Oócitos/citologia , Oócitos/fisiologia , Fosfatos/química , Radioisótopos de Fósforo/metabolismo , Isoformas de Proteínas/genética , Isoformas de Proteínas/metabolismo , RNA Complementar/metabolismo , Sódio/química , Radioisótopos de Sódio/metabolismo , Proteínas Cotransportadoras de Sódio-Fosfato Tipo III/genética , Xenopus laevis
12.
J Virol ; 78(17): 9564-7, 2004 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-15308749

RESUMO

The type III sodium-dependent phosphate (NaPi) cotransporter, Pit2, is a receptor for amphotropic murine leukemia virus (A-MuLV) and 10A1 MuLV. In order to determine what is sufficient for Pit2 receptor function, a deletion mutant lacking about the middle half of the protein was made. The mutant supported entry for both viruses, unequivocally narrowing down the identification of the sequence that is sufficient to specify the receptor functions of Pit2 to its N-terminal 182 amino acids and C-terminal 170 amino acids.


Assuntos
Vírus da Leucemia Murina/metabolismo , Receptores Virais/química , Receptores Virais/metabolismo , Animais , Células CHO , Cricetinae , Humanos , Vírus da Leucemia Murina/fisiologia , Receptores Virais/genética , Deleção de Sequência/genética
13.
J Biol Chem ; 277(45): 42741-7, 2002 Nov 08.
Artigo em Inglês | MEDLINE | ID: mdl-12205090

RESUMO

Type III sodium-dependent phosphate (NaP(i)) cotransporters, Pit1 and Pit2, have been assigned housekeeping P(i) transport functions and suggested involved in chondroblastic and osteoblastic mineralization and ectopic calcification. Both proteins exhibit dual function, thus, besides being transporters, they also serve as receptors for several gammaretroviruses. We here show that it is possible to uncouple transport and receptor functions of a type III NaP(i) cotransporter and thus exploit the retroviral receptor function as a control for proper processing and folding of mutant proteins. Thus exchanging two putative transmembranic glutamate residues in human Pit2, Glu(55) and Glu(575), with glutamine or with lysine severely impaired or knocked out, respectively, P(i) transport function, but left viral receptor function undisturbed. Both glutamates are conserved in type III NaP(i) cotransporters, in fungal NaP(i) cotransporters PHO-4 and Pho89, and in other known or putative phosphate permeases from a number of species and are the first residues shown to be critical for type III NaP(i) cotransport. Their putative transmembranic positions together with the presented data are consistent with Glu(55) and Glu(575) being parts of a cation liganding site or playing roles in conformational changes associated with substrate transport. Finally, the results also show that Pit2 retroviral receptor function per se is not dependent on Pit2 P(i) transport function.


Assuntos
Ácido Glutâmico , Receptores Virais/genética , Retroviridae/fisiologia , Simportadores/genética , Substituição de Aminoácidos , Animais , Biotinilação , Linhagem Celular , Sequência Conservada , Glutamina , Humanos , Lisina , Mutagênese Sítio-Dirigida , Oócitos/fisiologia , Fosfatos/metabolismo , Dobramento de Proteína , Receptores Virais/química , Receptores Virais/metabolismo , Proteínas Recombinantes/química , Proteínas Recombinantes/metabolismo , Proteínas de Saccharomyces cerevisiae , Proteínas Cotransportadoras de Sódio-Fosfato , Proteínas Cotransportadoras de Sódio-Fosfato Tipo III , Simportadores/química , Simportadores/metabolismo , Transfecção , Xenopus laevis
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA