Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 21
Filtrar
Mais filtros











Intervalo de ano de publicação
1.
J Membr Biol ; 252(1): 1-16, 2019 02.
Artigo em Inglês | MEDLINE | ID: mdl-30392010

RESUMO

The present research report describes Na+/H+ antiport by brush border membrane vesicles isolated from whole larvae of Aedes aegypti (AeBBMVw). Our hypothesis is that acid quenching of acridine orange by AeBBMVw is predominantly mediated by Na+/H+ antiport via the NHA1 component of the AeBBMVw in the absence of amino acids and ATP. AeNHA1 is a Na+/H+ antiporter that has been postulated to exchange Na+ and H+ across the apical plasma membrane in posterior midgut of A. aegypti larvae. Its principal function is to recycle the H+ and Na+ that are transported during amino acid uptake, e.g., phenylalanine. This uptake is mediated, in part, by a voltage-driven, Na+-coupled, nutrient amino acid transporter (AeNAT8). The voltage is generated by an H+ V-ATPase. All three components, V-ATPase, antiporter, and nutrient amino acid transporter (VAN), are present in brush border membrane vesicles isolated from whole larvae of A. aegypti. By omitting ATP and amino acids, Na+/H+ antiport was measured by fluorescence quenching of acridine orange (AO) caused by acidification of either the internal vesicle medium (Na+in > Na+out) or the external fluid-membrane interface (Na+in < Na+out). Vesicles with 100 micromolar Na+ inside and 10 micromolar Na+ outside or with 0.01 micromolar Na+ inside and 100 micromolar Na+ outside quenched fluorescence of AO by as much as 30%. Acidification did not occur in the absence of AeBBMVw. Preincubation of AeBBMVw with antibodies to NHA1 inhibit Na+/H+ antiport dependent fluorescence quenching, indicating that AeNHA1 has a significant role in Na+/H+ exchange.


Assuntos
Aedes/efeitos dos fármacos , Aedes/metabolismo , Anticorpos/farmacologia , Vesículas Citoplasmáticas/metabolismo , Microvilosidades/metabolismo , Trocadores de Sódio-Hidrogênio/antagonistas & inibidores , Animais , Transporte Biológico/efeitos dos fármacos , Vesículas Citoplasmáticas/ultraestrutura , Concentração de Íons de Hidrogênio , Íons/metabolismo , Cinética , Larva , Microvilosidades/ultraestrutura , Modelos Biológicos
2.
J Insect Physiol ; 58(4): 590-8, 2012 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-22410306

RESUMO

Deafness is a serious condition that affects millions of people and can also lead to dementia. Moreover, Karet and associates reported in 1999 that mutations in the gene encoding H(+) V-ATPase subunit B(1) lead to deafness. Yet ionic flows that enable humans to hear high-pitched sounds at 20,000 cycles/sec (20 kHz) are not well understood. Sound is transduced to electrical signals by stereocilia of hair cells by influx of Ca(2+) and K(+) as the "transducer channel" opens transiently and reduces the ∼90 mV (endolymph positive) endocochlear potential (EP) by ∼20 mV as the receptor potential. The EP as well as concentrations of Ca(2+), H(+) and K(+) must remain constant to produce reliable signals. Ca(2+) entry is balanced by Ca(2+) exit via a plasma membrane Ca(2+) ATPase (PMCA2a) but the Ca(2+) exit is coupled to H(+) entry. Moreover, K(+) entry is balanced by K(+) exit via a long diffusion route through several channels which is too slow to account for 20 kHz signaling. The problem is solved by a new hypothesis in which an H(+) V-ATPase generates the EP and removes the H(+) while a new K(+)/H(+) antiporter uses the voltage to drive H(+) back in and the K(+) back out. In the new model, Ca(2+), H(+) and K(+) cycle between unstirred layers on the endolymph- and cytoplasmic- borders of the stereocilial membrane through distances of ∼20 nanometers with travel time of ∼10 µs, which is fast enough to account for the 50 µs open/close time for 20 kHz signaling. Central to this model is the hypothesis that a K(+) pump which secretes K(+) into a K(+)-rich compartment is composed of a voltage producing (electrogenic) H(+) V-ATPase that is electrically coupled to a voltage-driven (electrophoretic) K(+)/nH(+) antiporter (KHA). Conversely, for an H(+) V-ATPase to secrete K(+) into a K(+) rich compartment, it must be coupled to a KHA. Richard Keynes reviewed evidence in 1969 that such a K(+) pump, which he called a Type V pump, is present in the stria vascularis of cochlea and the goblet cell apical membrane of caterpillars. Its signature is a large outside positive potential of ∼100 mV, K(+) secretion into a K(+) rich compartment and reversible inhibition by anoxia. The key role of the Type V K(+) pump in generating the EP was recognized by Sellick and Bock in 1974 and others but has disappeared from the hearing literature during the past decades. Its revival here is based on immunolocalization of KHA2 in the stereocilial membrane and Gillespie's generously shared mass spectroscopy evidence that all but one of the V(1) ATPase subunits are detected in isolated chicken stereocilia but V(o) and KHAs are not detected (implying that KHAs must be in the membrane). The new model proposed in the present paper could lead to important changes in our understanding of sensory physiology.


Assuntos
Células Ciliadas Auditivas/metabolismo , Antiportadores de Potássio-Hidrogênio/metabolismo , Estereocílios/metabolismo , ATPases Vacuolares Próton-Translocadoras/metabolismo , Animais , Trato Gastrointestinal/metabolismo , Células Caliciformes/metabolismo , Humanos , Larva/metabolismo , Lepidópteros/metabolismo , Rana catesbeiana , Estria Vascular/metabolismo
3.
J Insect Physiol ; 58(4): 580-9, 2012 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-22251673

RESUMO

Brush border membrane vesicles from whole Aedes aegypti larvae (AaBBMVw) are confirmed to be valid preparations for membrane transport studies. The Abdul-Rauf and Ellar method was used to isolate AaBBMVw that were frozen, stored for several months, transported to a distant site, thawed and used to study Na(+)-coupled, (3)H-labeled, phenylalanine (Phe) uptake. The affinity for all components of the uptake was very high with half maximal values in the sub-micromolar range. By contrast a K(0.5)(Phe) of 0.2mM and a K(0.5)(Na) of 26 mM were calculated from Phe-induced electrical currents in Xenopus oocytes that were heterologously expressing the Anopheles gambiae symporter (co-transporter), AgNAT8, in a buffer with 98 mM Na(+). What accounts for the >1000-fold discrepancy in affinity for substrates between the BBMV and oocyte experiments? Is it because Ae. aegypti were used to isolate BBMVw whereas An. gambiae were used to transfect oocytes? More likely, it is because BBMVw were exposed to [Na(+)] in the micromolar range with the transporter(s) being surrounded by native lipids. By contrast, the oocyte measurements were made at [Na(+)] 100,000 times higher with AgNAT8 surrounded by foreign frog lipids. The results show that AaBBMVw are osmotically sealed; the time-course has a Na(+)-induced overshoot, the pH optimum is ∼7 and the K(0.5) values for Phe and Na(+) are very low. The transport is virtually unchanged when Na(+) is replaced by K(+) or Li(+) but decreased by Rb(+). This approach to resolving discrepancies between electrical data on solute transporters such as AgNAT8 that are over-expressed in oocytes and flux data on corresponding transporters that are highly expressed in native membrane vesicles, may serve as a model for similar studies that add membrane biochemistry to molecular biology in efforts to identify targets for the development of new methods to control disease-vector mosquitoes.


Assuntos
Aedes/metabolismo , Microvilosidades/metabolismo , Fenilalanina/metabolismo , Vesículas Transportadoras/metabolismo , Aedes/ultraestrutura , Animais , Concentração de Íons de Hidrogênio , Cinética , Larva/metabolismo , Larva/ultraestrutura , Osmose , Sódio/metabolismo , Cloreto de Sódio/metabolismo , Trítio , Xenopus
4.
J Insect Physiol ; 58(4): 570-9, 2012 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-22206887

RESUMO

The newly identified metazoan Na(+)/H(+) antiporter (NHA) family is represented by two paralogues, AgNHA1 and AgNHA2, in the genome of the African malaria mosquito, Anopheles gambiae. Both antiporters are postulated to be electrophoretic i.e. voltage-driven. AgNHA1 was first cloned from An. gambiae larvae and immunolocalized with respect to the H(+) V-ATPase by the Harvey laboratory. Little is known about the properties of NHA1s; attempts to characterize AgNHA1 in Na(+)/H(+) exchanger (NHE)-lacking Chinese hamster ovary cells and in yeast cells or frog oocytes were unsuccessful. Even less is known about AgNHA2. It is predicted to have a relative molecular mass of ∼60 kDa and shares 30.5% amino acid identity with AgNHA1. Immunolocalization images show AgNHA2 on the apical plasma membrane of stellate cells in Malpighian tubules of An. gambiae larvae and adults. When heterologously expressed in a mutant strain of the yeast, Saccharomyces cerevisiae, which lacks endogenous cation/proton antiporters and pumps, AgNHA2 enhanced repression of growth by the alkali metal cations, Li(+), Na(+), or K(+) and enhanced Li(+) accumulation. The yeast growth studies invite the speculation that AgNHA2 is an electrophoretic antiporter with a stoichiometry of nNa(+) to 1H(+) with n > 1. Immunolocalization images provide direct evidence that H(+) V-ATPase is co-localized with AgNHA1 on the apical membrane of principal cells but it is not present in the stellate cells where AgNHA2 is localized apically. These results are consistent with the notion that the outside positive voltage that the H(+) V-ATPase generates across the apical membrane of principal cells appears with but little attenuation across the apical membrane of stellate cells. This immunolocalization pattern is consistent with the hypothesis that the voltage acts via AgNHA1 to drive nH(+) into the principal cells and Na(+) out to the lumen and acts via AgNHA2 to drive nNa(+) into the stellate cells and H(+) out to the lumen. Precious Na(+) is then retained by ejection into the blood via a basal Na(+)/K(+)-ATPase. Localizations of anion transporters and their functions in stellate and principal cells are described by Linser, Romero and associates in this volume. The role that the electrogenic H(+) V-ATPase and the electrophoretic cationic and anionic transporters play in ion homeostasis is incorporated into a model for Malpighian tubule cells of larval mosquitoes.


Assuntos
Anopheles/metabolismo , Proteínas de Insetos/metabolismo , Túbulos de Malpighi/metabolismo , Trocadores de Sódio-Hidrogênio/metabolismo , ATPases Vacuolares Próton-Translocadoras/metabolismo , Sequência de Aminoácidos , Animais , Epitélio/metabolismo , Trato Gastrointestinal/metabolismo , Homeostase , Larva/metabolismo , Dados de Sequência Molecular , Saccharomyces cerevisiae/metabolismo
5.
J Am Mosq Control Assoc ; 27(2): 165-7, 2011 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-21805853

RESUMO

The release of infected mosquitoes or other arthropods by bioterrorists, i.e., arboterrorism, to cause disease and terror is a threat to the USA. A workshop to assess mosquito control response capabilities to mount rapid and effective responses to eliminate an arboterrorism attack provided recommendations to improve capabilities in the USA. It is essential that mosquito control professionals receive training in possible responses, and it is recommended that a Council for Emergency Mosquito Control be established in each state to coordinate training, state resources, and actions for use throughout the state.


Assuntos
Bioterrorismo , Culicidae/microbiologia , Culicidae/parasitologia , Planejamento em Desastres , Transmissão de Doença Infecciosa/prevenção & controle , Controle de Mosquitos , Animais , Planejamento em Desastres/economia , Planejamento em Desastres/organização & administração , Florida , Insetos Vetores , Política Pública
6.
J Insect Physiol ; 56(10): 1377-89, 2010 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-20435040

RESUMO

Brush border membrane vesicles (BBMVs) from Whole larvae of Aedes aegypti (AeBBMVWs) contain an H(+) V-ATPase (V), a Na(+)/H(+) antiporter, NHA1 (A) and a Na(+)-coupled, nutrient amino acid transporter, NAT8 (N), VAN for short. All V-ATPase subunits are present in the Ae. aegypti genome and in the vesicles. AgNAT8 was cloned from Anopheles gambiae, localized in BBMs and characterized in Xenopus laevis oocytes. AgNHA1 was cloned and localized in BBMs but characterization in oocytes was compromised by an endogenous cation conductance. AeBBMVWs complement Xenopus oocytes for characterizing membrane proteins, can be energized by voltage from the V-ATPase and are in their natural lipid environment. BBMVs from caterpillars were used in radio-labeled solute uptake experiments but approximately 10,000 mosquito larvae are needed to equal 10 caterpillars. By contrast, functional AeBBMVWs can be prepared from 10,000 whole larvae in 4h. Na(+)-coupled (3H)phenylalanine uptake mediated by AeNAT8 in AeBBMVs can be compared to the Phe-induced inward Na(+) currents mediated by AgNAT8 in oocytes. Western blots and light micrographs of samples taken during AeBBMVW isolation are labeled with antibodies against all of the VAN components. The use of AeBBMVWs to study coupling between electrogenic V-ATPases and the electrophoretic transporters is discussed.


Assuntos
Aedes/enzimologia , Proteínas de Insetos/metabolismo , Vesículas Secretórias/enzimologia , ATPases Vacuolares Próton-Translocadoras/metabolismo , Aedes/classificação , Aedes/genética , Aedes/crescimento & desenvolvimento , Sequência de Aminoácidos , Animais , Proteínas de Insetos/genética , Larva/classificação , Larva/enzimologia , Larva/genética , Larva/crescimento & desenvolvimento , Dados de Sequência Molecular , Filogenia , Vesículas Secretórias/genética , ATPases Vacuolares Próton-Translocadoras/genética
7.
J Exp Biol ; 212(Pt 10): 1559-67, 2009 May.
Artigo em Inglês | MEDLINE | ID: mdl-19411550

RESUMO

The nutrient amino acid transporter (NAT) subfamily of the neurotransmitter sodium symporter family (NSS, also known as the solute carrier family 6, SLC6) represents transport mechanisms with putative synergistic roles in the absorption of essential and conditionally essential neutral amino acids. It includes a large paralogous expansion of insect-specific genes, with seven genes from the genome of the malaria mosquito, Anopheles gambiae. One of the An. gambiae NATs, AgNAT8, was cloned, functionally expressed and characterized in X. laevis oocytes as a cation-coupled symporter of aromatic amino acids, preferably l-phenylalanine, l-tyrosine and l-DOPA. To explore an evolutionary trend of NAT-SLC6 phenotypes, we have cloned and characterized AgNAT6, which represents a counterpart of AgNAT8 descending from a recent gene duplication (53.1% pairwise sequence identity). In contrast to AgNAT8, which preferably mediates the absorption of phenol-branched substrates, AgNAT6 mediates the absorption of indole-branched substrates with highest apparent affinity to tryptophan (K(0.5)(Trp)=1.3 micromol l(-1) vs K(0.5)(Phe)=430 micromol l(-1)) and [2 or 1 Na(+) or K(+)]:[aromatic substrate] stoichiometry. AgNAT6 is highly transcribed in absorptive and secretory regions of the alimentary canal and specific neuronal structures, including the neuropile of ventral ganglia and sensory afferents. The alignment of AgNATs and LeuT(Aa), a bacterial NAT with a resolved 3D structure, reveals three amino acid differences in the substrate-binding pocket that may be responsible for the indole- vs phenol-branch selectivity of AgNAT6 vs AgNAT8. The identification of transporters with a narrow selectivity for essential amino acids suggests that basal expansions in the SLC6 family involved duplication and retention of NATs, improving the absorption and distribution of under-represented essential amino acids and related metabolites. The identified physiological and expression profiles suggest unique roles of AgNAT6 in the active absorption of indole-branched substrates that are used in the synthesis of the neurotransmitter serotonin as well as the key circadian hormone and potent free-radical scavenger melatonin.


Assuntos
Anopheles/metabolismo , Clonagem Molecular , Sódio/metabolismo , Simportadores/metabolismo , Triptofano/metabolismo , Sequência de Aminoácidos , Animais , Anopheles/genética , Regulação da Expressão Gênica/fisiologia , Larva/metabolismo , Dados de Sequência Molecular , Filogenia , Simportadores/química , Simportadores/genética
8.
J Exp Biol ; 212(Pt 11): 1620-9, 2009 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-19448072

RESUMO

This review provides alternatives to two well established theories regarding membrane energization by H(+) V-ATPases. Firstly, we offer an alternative to the notion that the H(+) V-ATPase establishes a protonmotive force (pmf) across the membrane into which it is inserted. The term pmf, which was introduced by Peter Mitchell in 1961 in his chemiosmotic hypothesis for the synthesis of ATP by H(+) F-ATP synthases, has two parts, the electrical potential difference across the phosphorylating membrane, Deltapsi, and the pH difference between the bulk solutions on either side of the membrane, DeltapH. The DeltapH term implies three phases - a bulk fluid phase on the H(+) input side, the membrane phase and a bulk fluid phase on the H(+) output side. The Mitchell theory was applied to H(+) V-ATPases largely by analogy with H(+) F-ATP synthases operating in reverse as H(+) F-ATPases. We suggest an alternative, voltage coupling model. Our model for V-ATPases is based on Douglas B. Kell's 1979 'electrodic view' of ATP synthases in which two phases are added to the Mitchell model - an unstirred layer on the input side and another one on the output side of the membrane. In addition, we replace the notion that H(+) V-ATPases normally acidify the output bulk solution with the hypothesis, which we introduced in 1992, that the primary action of a H(+) V-ATPase is to charge the membrane capacitance and impose a Deltapsi across the membrane; the translocated hydrogen ions (H(+)s) are retained at the outer fluid-membrane interface by electrostatic attraction to the anions that were left behind. All subsequent events, including establishing pH differences in the outside bulk solution, are secondary. Using the surface of an electrode as a model, Kell's 'electrodic view' has five phases - the outer bulk fluid phase, an outer fluid-membrane interface, the membrane phase, an inner fluid-membrane interface and the inner bulk fluid phase. Light flash, H(+) releasing and binding experiments and other evidence provide convincing support for Kell's electrodic view yet Mitchell's chemiosmotic theory is the one that is accepted by most bioenergetics experts today. First we discuss the interaction between H(+) V-ATPase and the K(+)/2H(+) antiporter that forms the caterpillar K(+) pump, and use the Kell electrodic view to explain how the H(+)s at the outer fluid-membrane interface can drive two H(+) from lumen to cell and one K(+) from cell to lumen via the antiporter even though the pH in the bulk fluid of the lumen is highly alkaline. Exchange of outer bulk fluid K(+) (or Na(+)) with outer interface H(+) in conjunction with (K(+) or Na(+))/2H(+) antiport, transforms the hydrogen ion electrochemical potential difference, mu(H), to a K(+) electrochemical potential difference, mu(K) or a Na(+) electrochemical potential difference, mu(Na). The mu(K) or mu(Na) drives K(+)- or Na(+)-coupled nutrient amino acid transporters (NATs), such as KAAT1 (K(+) amino acid transporter 1), which moves Na(+) and an amino acid into the cell with no H(+)s involved. Examples in which the voltage coupling model is used to interpret ion and amino acid transport in caterpillar and larval mosquito midgut are discussed.


Assuntos
Potenciais da Membrana/fisiologia , Potássio/metabolismo , ATPases Translocadoras de Prótons/metabolismo , Sódio/metabolismo , Animais , Bactérias/metabolismo
9.
J Exp Biol ; 212(Pt 3): 347-57, 2009 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-19151209

RESUMO

Glycolysis, the citric acid cycle and other metabolic pathways of living organisms generate potentially toxic acids within all cells. One ubiquitous mechanism for ridding cells of the acids is to expel H(+) in exchange for extracellular Na(+), mediated by electroneutral transporters called Na(+)/H(+) exchangers (NHEs) that are driven by Na(+) concentration gradients. The exchange must be important because the human genome contains 10 NHEs along with two Na(+)/H(+) antiporters (NHAs). By contrast, the genomes of two principal disease vector mosquitoes, Anopheles gambiae and Aedes aegypti, contain only three NHEs along with the two NHAs. This shortfall may be explained by the presence of seven nutrient amino acid transporters (NATs) in the mosquito genomes. NATs transport Na(+) stoichiometrically linked to an amino acid into the cells by a process called symport or co-transport. Three of the mosquito NATs and two caterpillar NATs have previously been investigated after heterologous expression in Xenopus laevis oocytes and were found to be voltage driven (electrophoretic). Moreover, the NATs are present in the same membrane as the H(+) V-ATPase, which generates membrane potentials as high as 120 mV. We review evidence that the H(+) V-ATPase moves H(+) out of the cells and the resulting membrane potential (V(m)) drives Na(+) linked to an amino acid into the cells via a NAT. The H(+) efflux by the V-ATPase and Na(+) influx by the NAT comprise the same ion exchange as that mediated by an NHE; so the V and NAT working together constitute an NHE that we call NHE(VNAT). As the H(+) V-ATPase is widely distributed in mosquito epithelial cells and there are seven NATs in the mosquito genomes, there are potentially seven NHE(VNAT)s that could replace the missing NHEs. We review published evidence in support of this hypothesis and speculate about broader functions of NHE(VNAT)s.


Assuntos
Sistemas de Transporte de Aminoácidos Neutros/metabolismo , Sistemas de Transporte de Aminoácidos/metabolismo , Culicidae/metabolismo , Proteínas de Insetos/metabolismo , Modelos Moleculares , Filogenia , Trocadores de Sódio-Hidrogênio/metabolismo , Sódio/metabolismo , ATPases Vacuolares Próton-Translocadoras/metabolismo , Sequência de Aminoácidos , Sistemas de Transporte de Aminoácidos/genética , Sistemas de Transporte de Aminoácidos Neutros/genética , Animais , Proteínas de Insetos/genética , Transporte de Íons , Potenciais da Membrana , Dados de Sequência Molecular , Trocadores de Sódio-Hidrogênio/genética
10.
J Exp Biol ; 211(Pt 19): 3067-76, 2008 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-18805805

RESUMO

Mosquito larvae live in dynamic aqueous environments, which can fluctuate drastically in salinity due to environmental events such as rainfall and evaporation. Larval survival depends upon the ability to regulate hemolymph osmolarity by absorbing and excreting ions. A major organ involved in ion regulation is the rectum, the last region for modification of the primary urine before excretion. The ultrastructure and function of culicine larval recta have been studied extensively; however, very little published data exist on the recta of anopheline larvae. To gain insight into the structure and functions of this organ in anopheline species, we used immunohistochemistry to compare the localization of three proteins [carbonic anhydrase (CA9), Na+/K+ P-ATPase and H+ V-ATPase] in the recta of anopheline larvae reared in freshwater and saline water with the localization of the same proteins in culicine larvae reared under similar conditions. Based on the following key points, we concluded that anophelines differ from culicines in larval rectal structure and in regulation of protein expression: (1) despite the fact that obligate freshwater and saline-tolerant culicines have structurally distinct recta, all anophelines examined (regardless of saline-tolerance) have a structurally similar rectum consisting of distinct DAR (dorsal anterior rectal) cells and non-DAR cells; (2) anopheline larvae undergo a dramatic shift in rectal Na+/K+-ATPase localization when reared in freshwater vs saline water. This shift is not seen in any culicine larvae examined. Additionally, we use these immunohistochemical analyses to suggest possible functions for the DAR and non-DAR cells of anopheline larvae in freshwater and saline conditions.


Assuntos
Anopheles/enzimologia , Culicidae/enzimologia , Proteínas de Insetos/análise , Adaptação Fisiológica , Animais , Anopheles/anatomia & histologia , Anopheles/citologia , Anidrases Carbônicas/análise , Anidrases Carbônicas/metabolismo , Culicidae/anatomia & histologia , Culicidae/citologia , Imuno-Histoquímica , Proteínas de Insetos/metabolismo , Larva/anatomia & histologia , Larva/metabolismo , ATPases Translocadoras de Prótons/análise , ATPases Translocadoras de Prótons/metabolismo , Reto/anatomia & histologia , Reto/citologia , Reto/metabolismo , Cloreto de Sódio/química , ATPase Trocadora de Sódio-Potássio/análise , ATPase Trocadora de Sódio-Potássio/metabolismo , Água/química
11.
J Exp Biol ; 211(Pt 10): 1594-602, 2008 May.
Artigo em Inglês | MEDLINE | ID: mdl-18456887

RESUMO

The nutrient amino acid transporter (NAT) subfamily is the largest subdivision of the sodium neurotransmitter symporter family (SNF; also known as SLC6; HUGO). There are seven members of the NAT population in the African malaria mosquito Anopheles gambiae, two of which, AgNAT6 and AgNAT8, preferably transport indole- and phenyl-branched substrates, respectively. The relative expression and distribution of these aromatic NATs were examined with transporter-specific antibodies in Xenopus oocytes and mosquito larval alimentary canal, representing heterologous and tissue expression systems, respectively. NAT-specific aromatic-substrate-induced currents strongly corresponded with specific accumulation of both transporters in the plasma membrane of oocytes. Immunolabeling revealed elevated expressions of both transporters in specific regions of the larval alimentary canal, including salivary glands, cardia, gastric caeca, posterior midgut and Malpighian tubules. Differences in relative expression densities and spatial distribution of the transporters were prominent in virtually all of these regions, suggesting unique profiles of the aromatic amino acid absorption. For the first time reversal of the location of a transporter between apical and basal membranes was identified in posterior and anterior epithelial domains corresponding with secretory and absorptive epithelial functions, respectively. Both aromatic NATs formed putative homodimers in the larval gut whereas functional monomers were over-expressed heterologously in Xenopus oocytes. The results unequivocally suggest functional synergy between substrate-specific AgNAT6 and AgNAT8 in intracellular absorption of aromatic amino acids. More broadly, they suggest that the specific selectivity, regional expression and polarized membrane docking of NATs represent key adaptive traits shaping functional patterns of essential amino acid absorption in the metazoan alimentary canal and other tissues.


Assuntos
Sistemas de Transporte de Aminoácidos/metabolismo , Anopheles/metabolismo , Sistema Digestório/metabolismo , Animais , Western Blotting , Eletroforese em Gel de Poliacrilamida , Larva/metabolismo , Oligopeptídeos/genética , Oócitos/metabolismo , Xenopus laevis
12.
J Exp Biol ; 211(Pt 6): 957-68, 2008 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-18310121

RESUMO

Anopheles gambiae larvae (Diptera: Culicidae) live in freshwater with low Na(+) concentrations yet they use Na(+) for alkalinization of the alimentary canal, for electrophoretic amino acid uptake and for nerve function. The metabolic pathway by which larvae accomplish these functions has anionic and cationic components that interact and allow the larva to conserve Na(+) while excreting H(+) and HCO(3)(-). The anionic pathway consists of a metabolic CO(2) diffusion process, carbonic anhydrase and Cl(-)/HCO(3)(-) exchangers; it provides weak HCO(3)(-) and weaker CO(3)(2-) anions to the lumen. The cationic pathway consists of H(+) V-ATPases and Na(+)/H(+) antiporters (NHAs), Na(+)/K(+) P-ATPases and Na(+)/H(+) exchangers (NHEs) along with several (Na(+) or K(+)):amino acid(+/-) symporters, a.k.a. nutrient amino acid transporters (NATs). This paper considers the cationic pathway, which provides the strong Na(+) or K(+) cations that alkalinize the lumen in anterior midgut then removes them and restores a lower pH in posterior midgut. A key member of the cationic pathway is a Na(+)/H(+) antiporter, which was cloned recently from Anopheles gambiae larvae, localized strategically in plasma membranes of the alimentary canal and named AgNHA1 based upon its phylogeny. A phylogenetic comparison of all cloned NHAs and NHEs revealed that AgNHA1 is the first metazoan NHA to be cloned and localized and that it is in the same clade as electrophoretic prokaryotic NHAs that are driven by the electrogenic H(+) F-ATPase. Like prokaryotic NHAs, AgNHA1 is thought to be electrophoretic and to be driven by the electrogenic H(+) V-ATPase. Both AgNHA1 and alkalophilic bacterial NHAs face highly alkaline environments; to alkalinize the larva mosquito midgut lumen, AgNHA1, like the bacterial NHAs, would have to move nH(+) inwardly and Na(+) outwardly. Perhaps the alkaline environment that led to the evolution of electrophoretic prokaryotic NHAs also led to the evolution of an electrophoretic AgNHA1 in mosquito larvae. In support of this hypothesis, antibodies to both AgNHA1 and H(+) V-ATPase label the same membranes in An. gambiae larvae. The localization of H(+) V-ATPase together with (Na(+) or K(+)):amino acid(+/-) symporter, AgNAT8, on the same apical membrane in posterior midgut cells constitutes the functional equivalent of an NHE that lowers the pH in the posterior midgut lumen. All NATs characterized to date are Na(+) or K(+) symporters so the deduction is likely to have wide application. The deduced colocalization of H(+) V-ATPase, AgNHA1 and AgNAT8, on this membrane forms a pathway for local cycling of H(+) and Na(+) in posterior midgut. The local H(+) cycle would prevent unchecked acidification of the lumen while the local Na(+) cycle would regulate pH and support Na(+):amino acid(+/-) symport. Meanwhile, a long-range Na(+) cycle first transfers Na(+) from the blood to gastric caeca and anterior midgut lumen where it initiates alkalinization and then returns Na(+) from the rectal lumen to the blood, where it prevents loss of Na(+) during H(+) and HCO(3)(-) excretion. The localization of H(+) V-ATPase and Na(+)/K(+)-ATPase in An. gambiae larvae parallels that reported for Aedes aegypti larvae. The deduced colocalization of the two ATPases along with NHA and NAT in the alimentary canal constitutes a cationic pathway for Na(+)-conserving midgut alkalinization and de-alkalinization which has never been reported before.


Assuntos
Anopheles/metabolismo , Sequência de Aminoácidos , Sistemas de Transporte de Aminoácidos/genética , Sistemas de Transporte de Aminoácidos/metabolismo , Animais , Anopheles/genética , Anopheles/crescimento & desenvolvimento , Cátions/metabolismo , Polaridade Celular , Sistema Digestório/citologia , Sistema Digestório/metabolismo , Concentração de Íons de Hidrogênio , Imuno-Histoquímica , Proteínas de Insetos/genética , Proteínas de Insetos/metabolismo , Transporte de Íons , Larva/metabolismo , Modelos Biológicos , Sódio/metabolismo , Trocadores de Sódio-Hidrogênio/genética , Trocadores de Sódio-Hidrogênio/metabolismo , ATPase Trocadora de Sódio-Potássio/genética , ATPase Trocadora de Sódio-Potássio/metabolismo , ATPases Vacuolares Próton-Translocadoras/genética , ATPases Vacuolares Próton-Translocadoras/metabolismo
13.
J Exp Biol ; 210(Pt 21): 3848-61, 2007 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-17951426

RESUMO

We have cloned a cDNA encoding a new ion transporter from the alimentary canal of larval African malaria mosquito, Anopheles gambiae Giles sensu stricto. Phylogenetic analysis revealed that the corresponding gene is in a group that has been designated NHA, and which includes (Na+ or K+)/H+ antiporters; so the novel transporter is called AgNHA1. The annotation of current insect genomes shows that both AgNHA1 and a close relative, AgNHA2, belong to the cation proton antiporter 2 (CPA2) subfamily and cluster in an exclusive clade of genes with high identity from Aedes aegypti, Drosophila melanogaster, D. pseudoobscura, Apis mellifera and Tribolium castaneum. Although NHA genes have been identified in all phyla for which genomes are available, no NHA other than AgNHA1 has previously been cloned, nor have the encoded proteins been localized or characterized. The AgNHA1 transcript was localized in An. gambiae larvae by quantitative real-time PCR (qPCR) and in situ hybridization. AgNHA1 message was detected in gastric caeca and rectum, with much weaker transcription in other parts of the alimentary canal. Immunolabeling of whole mounts and longitudinal sections of isolated alimentary canal showed that AgNHA1 is expressed in the cardia, gastric caeca, anterior midgut, posterior midgut, proximal Malpighian tubules and rectum, as well as in the subesophageal and abdominal ganglia. A phylogenetic analysis of NHAs and KHAs indicates that they are ubiquitous. A comparative molecular analysis of these antiporters suggests that they catalyze electrophoretic alkali metal ion/hydrogen ion exchanges that are driven by the voltage from electrogenic H+ V-ATPases. The tissue localization of AgNHA1 suggests that it plays a key role in maintaining the characteristic longitudinal pH gradient in the lumen of the alimentary canal of An. gambiae larvae.


Assuntos
Anopheles/metabolismo , Proteínas de Insetos/análise , Trocadores de Sódio-Hidrogênio/análise , Sequência de Aminoácidos , Animais , Anopheles/classificação , Anopheles/genética , Western Blotting , Clonagem Molecular , Proteínas de Insetos/química , Proteínas de Insetos/genética , Larva/genética , Larva/metabolismo , Dados de Sequência Molecular , Filogenia , Estrutura Terciária de Proteína , RNA Mensageiro/metabolismo , Alinhamento de Sequência , Trocadores de Sódio-Hidrogênio/química , Trocadores de Sódio-Hidrogênio/genética
15.
J Exp Biol ; 209(Pt 16): 3183-98, 2006 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-16888066

RESUMO

Nutrient amino acid transporters (NATs, subfamily of sodium neurotransmitter symporter family SNF, a.k.a. SLC6) represent a set of phylogenetically and functionally related transport proteins, which perform intracellular absorption of neutral, predominantly essential amino acids. Functions of NATs appear to be critical for the development and survival in organisms. However, mechanisms of specific and synergetic action of various NAT members in the amino acid transport network are virtually unexplored. A new transporter, agNAT8, was cloned from the malaria vector mosquito Anopheles gambiae (SS). Upon heterologous expression in Xenopus oocytes it performs high-capacity, sodium-coupled (2:1) uptake of nutrients with a strong preference for aromatic catechol-branched substrates, especially phenylalanine and its derivatives tyrosine and L-DOPA, but not catecholamines. It represents a previously unknown SNF phenotype, and also appears to be the first sodium-dependent B(0) type transporter with a narrow selectivity for essential precursors of catecholamine synthesis pathways. It is strongly and specifically transcribed in absorptive and secretory parts of the larval alimentary canal and specific populations of central and peripheral neurons of visual-, chemo- and mechano-sensory afferents. We have identified a new SNF transporter with previously unknown phenotype and showed its important role in the accumulation and redistribution of aromatic substrates. Our results strongly suggest that agNAT8 is an important, if not the major, provider of an essential catechol group in the synthesis of catecholamines for neurochemical signaling as well as ecdysozoan melanization and sclerotization pathways, which may include cuticle hardening/coloring, wound curing, oogenesis, immune responses and melanization of pathogens.


Assuntos
Sistemas de Transporte de Aminoácidos/fisiologia , Aminoácidos Aromáticos/metabolismo , Anopheles/metabolismo , Proteínas de Insetos/fisiologia , Sequência de Aminoácidos , Sistemas de Transporte de Aminoácidos/química , Sistemas de Transporte de Aminoácidos/genética , Animais , Anopheles/anatomia & histologia , Anopheles/genética , Catecolaminas/biossíntese , Catecóis/metabolismo , Clonagem Molecular , Condutividade Elétrica , Proteínas de Insetos/química , Proteínas de Insetos/genética , Dados de Sequência Molecular , Filogenia , Proteínas da Membrana Plasmática de Transporte de Neurotransmissores/genética , Estrutura Terciária de Proteína , RNA Mensageiro/metabolismo , Alinhamento de Sequência , Transdução de Sinais/genética , Xenopus
16.
Proc Natl Acad Sci U S A ; 102(5): 1360-5, 2005 Feb 01.
Artigo em Inglês | MEDLINE | ID: mdl-15665107

RESUMO

The biosynthesis of structural and signaling molecules depends on intracellular concentrations of essential amino acids, which are maintained by a specific system of plasma membrane transporters. We identify a unique population of nutrient amino acid transporters (NATs) within the sodium-neurotransmitter symporter family and have characterized a member of the NAT subfamily from the larval midgut of the Yellow Fever vector mosquito, Aedes aegypti (aeAAT1, AAR08269), which primarily supplies phenylalanine, an essential substrate for the synthesis of neuronal and cuticular catecholamines. Further analysis suggests that NATs constitute a comprehensive transport metabolon for the epithelial uptake and redistribution of essential amino acids including precursors of several neurotransmitters. In contrast to the highly conserved subfamily of orthologous neurotransmitter transporters, lineage-specific, paralogous NATs undergo rapid gene multiplication/substitution that enables a high degree of evolutionary plasticity of nutrient amino acid uptake mechanisms and facilitates environmental and nutrient adaptations of organisms. These findings provide a unique model for understanding the molecular mechanisms, physiology, and evolution of amino acid and neurotransmitter transport systems and imply that monoamine and GABA transporters evolved by selection and conservation of earlier neuronal NATs.


Assuntos
Sistemas de Transporte de Aminoácidos/genética , Aedes/crescimento & desenvolvimento , Sequência de Aminoácidos , Sistemas de Transporte de Aminoácidos/classificação , Animais , Sequência de Bases , Clonagem Molecular , Primers do DNA , Feminino , Hibridização In Situ , Larva , Potenciais da Membrana/fisiologia , Dados de Sequência Molecular , Oócitos/fisiologia , Técnicas de Patch-Clamp , Filogenia , Mapeamento por Restrição , Alinhamento de Sequência , Homologia de Sequência de Aminoácidos , Transcrição Gênica , Xenopus laevis
17.
J Exp Biol ; 206(Pt 2): 245-54, 2003 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-12477895

RESUMO

KAAT1, a K(+)-coupled, neutral amino acid transporter from larval insect midgut, differs from other members of the Na(+):neurotransmitter transporter family (SNF) in two important ways: (1) it transports nutrient L-, alpha-amino acids, rather than neurotransmitters such as gamma-aminobutyric acid (GABA), and (2) it accepts K(+) as well as Na(+) as a co-substrate. To determine whether the restoration of KAAT1 residues to their GABA transporter GAT1 cation-binding equivalents might abolish its K(+) but not its Na(+) recognition site, we constructed a multiple mutant in which nine divergent KAAT1 residues were mutated back to the conserved form of the superfamily. To investigate the amino-acid-binding site, we constructed several single mutants that had been identified in GAT1. Wild-type (WT) or mutant cRNA was injected into Xenopus oocytes and the effects of external amino acids and ions upon labeled leucine uptake and substrate-induced currents were examined. The multiple mutant exhibited no amino-acid-induced currents, indicating that one or more of the mutated residues are crucial for function. W75L and R76E mutations in the first transmembrane helix of KAAT1 led to results equivalent to those observed in the corresponding mutants of GAT1; namely, substrate (leucine) uptake and substrate-evoked net inward current were severely curtailed. The KAAT1 A523S mutant, which corresponds to a serotonin transporter mutant that is thought to render Li(+) equivalent to Na(+) as a co-transported ion, functioned no differently to WT. The effects of mutation Y147F in the third transmembrane helix of KAAT1 were dramatically different from the equivalent mutation, Y140F, in GAT1. Although kinetic characteristics, expression levels and plasma membrane localization were all similar in Y147F and WT, the Y147F mutant exhibited a sevenfold increase in labeled leucine uptake by Xenopus oocytes in Na(+) buffer. This increase is in sharp contrast to the complete loss of uptake activity in the GAT1 Y140F mutant. KAAT1 Y147F also differed from WT in cation selectivity and substrate spectrum, as revealed by amino-acid-induced net inward currents that were measured with a two-electrode voltage clamp. Amino-acid-independent currents induced by Li(+) and Na(+) chloride salts were observed in both WT and the Y147F mutant. The Li(+)-induced current was 30% higher in Y147F than in WT, whereas no substrate-independent K(+)-induced currents above control levels were detected either in WT or Y147F. These results suggest that transport of K(+), the physiological co-substrate in insect midgut, is tightly coupled to that of amino acids in KAAT1, in contrast to the independence of cation and amino acid transport in the closely related cation amino acid transporter channel, CAATCH1.


Assuntos
Sistemas de Transporte de Aminoácidos Neutros , Proteínas de Transporte/genética , Proteínas de Transporte/metabolismo , Glicoproteínas de Membrana/genética , Glicoproteínas de Membrana/metabolismo , Sequência de Aminoácidos , Substituição de Aminoácidos , Aminoácidos/metabolismo , Animais , Sítios de Ligação , Transporte Biológico Ativo/efeitos dos fármacos , Proteínas de Transporte/química , Feminino , Técnicas In Vitro , Proteínas de Insetos/química , Proteínas de Insetos/genética , Proteínas de Insetos/metabolismo , Cinética , Leucina/metabolismo , Leucina/farmacologia , Lítio/farmacologia , Manduca/genética , Manduca/metabolismo , Glicoproteínas de Membrana/química , Dados de Sequência Molecular , Oócitos/metabolismo , Mutação Puntual , Potássio/metabolismo , Potássio/farmacologia , Estrutura Secundária de Proteína , Proteínas Recombinantes/química , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Sódio/farmacologia , Xenopus
18.
BMC Med Ethics ; 3: E3, 2002 Aug 12.
Artigo em Inglês | MEDLINE | ID: mdl-12171602

RESUMO

BACKGROUND: Intensivists must provide enough analgesia and sedation to ensure dying patients receive good palliative care. However, if it is perceived that too much is given, they risk prosecution for committing euthanasia. The goal of this study is to develop consensus guidelines on analgesia and sedation in dying intensive care unit patients that help distinguish palliative care from euthanasia. METHODS: Using the Delphi technique, panelists rated levels of agreement with statements describing how analgesics and sedatives should be given to dying ICU patients and how palliative care should be distinguished from euthanasia. Participants were drawn from 3 panels: 1) Canadian Academic Adult Intensive Care Fellowship program directors and Intensive Care division chiefs (N = 9); 2) Deputy chief provincial coroners (N = 5); 3) Validation panel of Intensivists attending the Canadian Critical Care Trials Group meeting (N = 12). RESULTS: After three Delphi rounds, consensus was achieved on 16 statements encompassing the role of palliative care in the intensive care unit, the management of pain and suffering, current areas of controversy, and ways of improving palliative care in the ICU. CONCLUSION: Consensus guidelines were developed to guide the administration of analgesics and sedatives to dying ICU patients and to help distinguish palliative care from euthanasia.


Assuntos
Analgésicos/uso terapêutico , Hipnóticos e Sedativos/uso terapêutico , Cuidados Paliativos/métodos , Cuidados Paliativos/normas , Guias de Prática Clínica como Assunto , Assistência Terminal/normas , Consenso , Eutanásia Ativa , Humanos , Unidades de Terapia Intensiva , Dor/tratamento farmacológico , Estresse Psicológico/tratamento farmacológico , Suicídio Assistido , Doente Terminal
19.
J Exp Biol ; 205(Pt 16): 2545-53, 2002 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-12124378

RESUMO

CAATCH1 functions both as an amino-acid-gated cation channel and as a cation-dependent, proline-preferring, nutrient amino acid transporter in which the two functions are thermodynamically uncoupled. This study focuses on the ionic channel aspect, in which a Tyr(147) (wild type) to Phe(147) (Y147F) site-directed mutation was investigated by steady-state electrophysiological measurements in the Xenopus laevis oocyte expression system. This tyrosine residue is conserved within the third transmembrane domain in members of the Na(+):neurotransmitter transporter family (SNF), where it plays a role in binding pharmacological ligands such as cocaine to the serotonin (SERT), dopamine (DAT) and norepinephrine (NET) transporters. Epithelial CAATCH1 is a member of the SNF family. The results show that amino acid ligand-gating selectivity and current magnitudes in Na(+)- and K(+)-containing media are differentially altered in CAATCH1 Y147F compared with the wild type. In the absence of amino acid ligands, the channel conductance of Na(+), K(+) and Li(+) that is observed in the wild type was reduced to virtually zero in Y147F. In the wild type, proline binding increased conductance strongly in Na(+)-containing medium and moderately in K(+)-containing medium, whereas in Y147F proline failed to elicit any cation currents beyond those of N-methyl-D-glucamine- or water-injected oocytes. In the wild type, methionine binding strongly inhibited inward Na(+) currents, whereas in Y147F it strongly stimulated inward currents in both Na(+) and K(+)-containing media. Indeed, in Na(+)-containing medium, the relative potency ranking for inward current inhibition in the wild type (Met>Leu>Gly>Phe>Thr) was similar to the ranking of ligand-permissive gating of large inward currents in Y147F. In Na(+)-containing medium, current/voltage relationships elicited by ligands in the wild type were complex and reversing, whereas in Y147F they were linear and inwardly rectifying. In K(+)-containing medium, current/voltage relationships remained non-linear in Y147F. Both wild-type and Y147F currents were Cl(-)-independent. Together, these data demonstrate a critical role for Tyr(147) in ligand-binding selectivity and modulation of the ionic channel conductance in CAATCH1. The results support the argument that inhibition of the CAATCH1 conductance by free methionine shares some properties in common with ligand inhibition of DAT, SERT, NET and the gamma-aminobutyric acid transporter (GAT1).


Assuntos
Proteínas de Transporte/fisiologia , Proteínas de Insetos , Ativação do Canal Iônico/fisiologia , Proteínas de Membrana/fisiologia , Tirosina/fisiologia , Sequência de Aminoácidos , Animais , Sequência de Bases , Proteínas de Transporte/química , Proteínas de Transporte/genética , Primers do DNA , Humanos , Imuno-Histoquímica , Ligantes , Manduca , Proteínas de Membrana/química , Proteínas de Membrana/genética , Dados de Sequência Molecular , Mutagênese Sítio-Dirigida , Homologia de Sequência de Aminoácidos
20.
Artigo em Inglês | MEDLINE | ID: mdl-12052727

RESUMO

Nitrites and nitrates are widely used reporters of endogenous activity of nitric oxide synthases (NOS), an important group of enzymes producing the gaseous signal molecule nitric oxide (NO). However, due to the great chemical heterogeneity of neuronal tissues, standard analytical protocols for evaluation of neuronal nitrite/nitrate concentrations are inefficient. We optimized a high-performance capillary zone electrophoresis (CZE) technique to analyze nitrite/nitrate concentrations in submicroliter samples from mammalian neuronal tissues. The measurements were made using a PrinCE 476 computerized capillary electrophoresis system with a Crystal 1000 contact conductivity detector. Isotachophoretic stacking injection of 1000- to 10000-fold diluted samples, which had been pretreated with a custom-designed solid-phase microextraction (SPME) cartridge, was employed to assay micromolar and nanomolar nitrite and nitrate levels in the presence of the high millimolar chloride concentrations characteristic of many biological samples. In the presented technique, a 10-microl volume of diluted ganglionic sample was used for chloride removal and sample cleanup. The method yields high analytical performance, including good reproducibility, resolution, and accuracy. The limits of detection relative to undiluted sample matrix were 8.9 nM (0.41 ppb) and 3.54 nM (0.22 ppb) for nitrite and nitrate, respectively. In addition, this technique resolves other anions that are present in neuronal tissues at sub-nanomolar concentrations and can be broadly applied for high-throughput anionic profiling. In rat dorsal root ganglia, endogenous levels of nitrate (231+/-29 microM; n=6) and nitrite (24-96 microM) were found. These concentrations exceeded those previously found in neuronal tissue homogenates using different techniques.


Assuntos
Eletroforese Capilar/métodos , Neurônios/química , Nitratos/análise , Nitritos/análise
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA