• No se han encontrado resultados

canales de cloro- una perspectiva molecular

N/A
N/A
Protected

Academic year: 2020

Share "canales de cloro- una perspectiva molecular"

Copied!
8
0
0

Texto completo

(1)

Chloride channels: a molecular perspective

Thomas J Jentsch

Plasma membrane Ct- channels perform a variety of functions, including control of excitability in neurons and muscle, cell volume regulation and transepithelial transport. Structurally, three classes of Cl- channels have been

identified: ligand-gated, postsynaptic Cl- channels (e.g. GABA and glycine receptors); the cystic fibrosis transmembrane conductance regulator (CFTR) Cl- channels (which belong to the traffic ATPase superfamily); and the CLC family of Cl- channels. Recent developments of note include further characterization of the expanding CLC Cl- channel family, advances in understanding the regulation of the CFTR Cl- channel and its emergent role as a regulator of other channels, clarification of issues related to swelling-activated Cl- channels, and the discovery that several co-transporter molecules are now known to induce Cl- currents in Xenopus oocytes.

Address

Centre for Molecular Neurobiology Hamburg (ZMNH),

Hamburg University, Martinistrasse 52, O-20246 Hamburg, Germany Abbreviations

CFTR cystic fibrosis transmembrane conductance regulator GABA yaminobutyric acid

NBF nucleotide-binding fold

Current Opinion in Neurobiology 1996, 6:303-310

0 Current Biology Ltd ISSN 0959-4388

Introduction

Chloride channels are found in the plasma membrane of probably all eukaryotic cells. In contrast to the intracellular concentration of ions such as Caz+ or Na+, the intracellular concentration of Cl- is close to its electrochemical equilibrium. Thus, Cl- has no established function as a second messenger, and is only rarely involved in electrical excitation, Rather, in a way similar to K+ channels, Cl- channels dampen electrical excitability. This is most evident in the nervous system, where GABA and glycine receptors generally generate inhibitory postsynaptic currents, and in skeletal muscle. Mutations in glycine receptor subunits result in hyperekplexia (startle disease) [l], which is attributable to a lack of inhibitory input in a subset of neurons. Furthermore, mutations in the skeletal muscle Cl- channel protein CLC-1 lead to muscle hyperexcitability, resulting in myotonia [2,3]. Interestingly, GABA receptors may have, as an exception, excitatory effects in certain neurons where co-transporters can raise the intracellular Cl- concentration above its electrochemical equilibrium [4].

Chloride is also important for the osmotic control of water flow. Although Cl- is accompanied by K+ when its efflux via swelling-activated Cl- channels counteracts osmotic swelling, Cl- is often joined by Na+ when it creates tiny transepithelial osmotic gradients necessary for fluid secretion or absorption. Being not far from its electrochemical equilibrium, transport of Cl- across both membranes (apical and basolateral) is energetically cheap. The importance of Cl- channels for transepithelial transport is apparent from disorders such as cholera or cystic fibrosis, which are attributable to an activation or mutational inactivation, respectively, of CAMP-regulated Cl- channels. Further, the recent discovery that mutations in a kidney Cl- channel (CLC-5) lead to kidney stones [S”] again suggests a defect in transepithelial transport.

Cl- channels also fulfil important roles in intracellular organelles, including endocytotic and synaptic vesicles. This topic has been reviewed recently f6] and will not be covered here. Instead, this review will focus primarily on Cl- channels whose molecular identity is known, without, however, considering postsynaptic GABA and glycine receptors. The emphasis is placed on the most recent developments in this field. Focusing exclusively on Cl- channels in the nervous system would ignore many important developments, and is thus avoided.

CLC chloride channel proteins

The CLC proteins form the only known large gene family of Cl- channels. They show a high degree of evolutionary conservation and homologues are present in yeast and bacteria. CLC proteins comprise about 12 transmembrane domains (Figure lb) 17,801 and function as multimers [2,9].

(2)

304 Signalling mechanisms

Figure 1

(a) GABA and glycine receptors (b) CLC Cl’ channels

D9.. ... D12 ... ...

(c) CFTR

Q 1996 Current Opinion in Neurobiology

Topology models for GABA and glycine receptors, CLC Cl- channels, and CFTR. (a) GABA and glycine receptors are typical members of the ligand-gated ion channel superfamily, which includes the nicotinic acetylcholine receptor. Members have four transmembrane domains (Ml-M4) and probably function ss (mostly hetero-) pentamers. (b) CLC Cl- channels have about 12 tran~embrane domains (01 -Dl2), the exact topology of which is not yet known [8*,29]. A previous model I1 01 had to be modified, as the loop between D8 and 09 is now known to be glycosylated [9,13]. Analysis of new CLC genes suggests that D4 is not a conserved hydrophobic domain, eliminating it as a transmembrane span [20]. Whereas D2 is thought not to cross the membrane in an alternative model [91, several algorithms predict that it is a very good candidate for a transmembrane domain. Further, deletion of this domain destroys channel activity [5**,7] and results in kidney stones [5**]. The topology in the broad hydrophobic region OS-D1 2 is obscure, and the conserved region 013 is known to be cytoplasmic [l 11. (c) CFTR has

two blocks of six putative transmembrane domains (TM1 -TM6 and TM7-TM12), which are followed by the nucleotide-binding folds NBFl and NBFP, respectively, The regulatory ‘R’ domain must be phosphorylated before regulation of channel opening by ATP (at the NBFs) can occur.

detail exciting developments regarding specific CLC Cl- mechanism by which a defect in a Cl- channel leads to

channel proteins. hypercalciuria and proteinuria is still unclear.

CLC-5 CLC-1

(3)

Dendrogram showing the degree of similarity between the known mammalian members of the CLC superfamily. Members of the fmt branch, which includes CLC-1, CLC-2, CLC-Ka and CLCKb, are those most closely related to the Torpedo channel UC-O, the founding member of this gene family [lo]. The second branch, including CLC-3, CLC-4 and CLC-5, contains those members most closely related to the yeast homologue SCCLC 1301. (Interestingly, the recent completion of the yeast genome project indicates that there is only one CLC gene in yeast, which we suggest calling ‘scCW.) Finally, a recent add&ion to the CLC sup&amity is the CLC-6 and CLG7 branch. Tissue expression patterns and proposed functions are also indicated.

Thus, it seems unlikely that all these mutations affect gate structures directly.

CLC-0

Whereas many cation channels sense the membrane voltage using charged amino acids in transmembrane domains, ‘fast’ gating of CLC-0 can be described by a fundamentally different model in which the permeating anion itself provides the ‘gating charge’ [@I. Opening rates are increased by the presence of anions close to the internal end of the pore. This model provides a simple explanation for the observed voltage and Cl--concentration dependence of CLC-0 gating, and is supported by the analysis of mutations at the very end of the transmembrane spans. These mutations modulate gating, single-channel conductance, and ion selectivity, suggesting that this region is important for permeation. Ion selectivity is however affected by mutations in other regions as well [Z], so it has not yet been possible to convincingly identify all the structures directly lining the pore region.

CLC-2

CLC-2 can be activated by cell swelling or strong hyperpolarization in Xenopus oocytes [l I]. Superficially similar currents are present in native cells, including epithelia [23-251, neurons [26,27] and osteoblasts [28], but

the osmosensitivity has often not been checked and may even be reversed [28]. Although northern analysis suggests that CLC-2 is expressed ubiquitously, in situ hybridization reveals that CLC-2 is expressed differentiaIly in brain 1271, according to a pattern that differs from that of CLC3 [15,17]. It was proposed that CLC-2 serves to prevent neuronal Cl- accumulation above equilibrium, thereby modulating the effects of postsynaptic GABA receptors [4,26]. Consistent with this hypothesis, expression was observed to be absent or low in neurons exhibiting a ‘paradoxical’ excitation by GABA, attributable to a Cl- efflux via GABA receptors in cells with a high internal Cl- concentration [27].

CLC-4, CLC-6, CLC-7 and CLC-KS

(4)

306

Signalling

mechanisms

domain, and as a similar deletion in CLC-5 eliminates

channel activity and leads to kidney stones [5"'].

T h e lack of functional expression could be explained by

complicated regulation of channel activity or by the lack

of important subunits, among others. Alternatively, these

putative channels might serve intracellular roles. T h e

iron-repressible

petite

phenotype (deficiency in respiration)

of a yeast strain deleted for the yeast C L C homologue

scCLC encoded by the

GEF-1

gene ([30]; see also

Figure 2 legend) may point in the direction of the latter

explanation.

Multiple functions of CFTR

An increasing number of reports indicate that C F T R ,

the gene product defective in cystic fibrosis, performs

regulatory roles in addition to being a cAMP-activated

C I - c h a n n e l . This additional function has been signifi-

cantly, albeit indirectly, bolstered by the discovery that

the sulfonylurea receptor (SUR) regulates a m e m b e r of

the inwardly rectifying K ÷ channel family (BIR) to yield

ATP-semitive K ÷ channels [31]. Both C F T R and the

sulfonylurea receptor belong to the superfamily of traffic

ATPases, combining two blocks of transmembrane spans

with two nucleotide-binding folds (Figure lc), although

their topologies differ in detail.

Data obtained previously indicated that C F T R regulates

an outwardly rectifying CI- channel expressed in many

cells [32]. Intriguing experiments now suggest that this

regulation is mediated by ATP, which leaves the cell

in response to C F T R stimulation [33°,34]. This exodus

might be achieved via a pathway closely regulated by

C F T R , or directly via the C F T R pore itself, as suggested

previously [35]. On the other hand, no effect of C F T R on

ATP efflux was found in a different system [36]. A direct

ATP-channel function of C F T R may be surprising for a

pore otherwise highly selective for small anions. Indeed,

several groups have reported recently that C F T R does

not conduct ATP [37]. Extracellular ATP may activate

outwardly rectifying CI- channels by interacting with

purinergic receptors [33°,38] Whether this activation is

mediated by direct interaction, intracellular Ca 2÷, and/or

other pathways is not yet clear, and the molecular identity

of the outward rectifier is unknown. Intriguingly, some

mutations associated with cystic fibrosis have been shown

to have differential effects on intrinsic C I - channel and

regulator activities [39].

In addition to a defective CI- conductance, airway

epithelia of cystic fibrosis patients show an increase in

Na÷ currents. It is now known that C F T R leads to

cAMP-stimulated inhibition of co-transfected epithelial

Na ÷ channels [40"]. Although this effect is small in

fibroblasts, the inhibition observed in epithelial cells

seems substantial. Interactions between both channels

were also observed in the

Xenopus

oocyte expression

system [41]. T h e mechanism of this interaction is still

unclear, but these observations suggest that a search for

interactions with other channels might be fruitful. Indeed,

first reports indicate that C F T R may also regulate inwardly

rectifying K ÷ channels (CM McNicholas, WB Guggino,

SC Hebert, ME Egan,

Pediatr Pulmonol

1995, 19:77), and

indirect evidence suggests that C F T R may also affect

volume-activated K ÷ channels [42]. Clearly, more research

is needed to elucidate these effects, as it would be

surprising if C F T R were involved in all these diverse

processes.

In view of these regulatory roles, the additional function

of C F T R as a C I - channel is surprising, but well

established. T h e ATP-channel model [33"], although very

controversial [36,37], reconciles both functions. As C F T R

belongs to the traffic ATPase family, several studies

have focused on the roles of its two nucleotide-binding

folds (NBFs) [43",44"°,45,46]. In contrast with 'normal'

transport ATPases, in which ATP is hydrolyzed upon each

active transport cycle, the passive pore of the C F T R

channel is thought to be regulated by ATP hydrolysis.

This regulation can only occur after C F T R has been

phosphorylated by protein kinase A at the 'R domain'.

Extending previous results [47], the new data suggest

that ATP hydrolysis at NBF1 is important for channel

opening. Binding of ATP at the second N B F (NBF2) is

thought to stabilize the open state, and ATP hydrolysis

at NBF2 facilitates channel closure [44"%46]. A previously

undetected second open state (02) with a slightly different

conductance has been exploited in an elegant series

of experiments

[44"].

T h e results suggest that ATP

hydrolysis at NBF2 leads to a transition from the first

open state (O1) to O 2, from which the channel can

close spontaneously [44"']. Although these studies only

indirectly suggest that ATP is hydrolyzed by C F T R , new

biochemical data indicate that NBF1 (in a fusion protein)

may indeed function as an ATPase [48].

T h e most frequent mutant form of C F T R , AF508, does

not reach the cell surface but instead is degraded in

a pre-Golgi compartment. It has now been shown that,

rather like other m e m b r a n e proteins, both wild-type and

mutant C F T R proteins are degraded in part by the

ubiquitin-proteasome pathway [49,50].

Although changes in ion selectivity have been corre-

lated with mutations in several transmembrane domains

([51,52]; but, see [53]), the pore of this extensively studied

molecule is not yet well understood. It is a sure bet that

C F T R still has many surprises in store.

Chloride channels and regulation of cell

volume

(5)

(MDR1) generates volume-activated C I - c h a n n e l s [54]

has stirred considerable interest and controversy over

the years [55-60]. It now seems clear that M D R 1

is not itself a CI- channel. Instead, it is thought to

confer regulation by protein kinase C onto endogenous

swelling-activated CI- channels [61]. As MDR1 is a

traffic ATPase (indeed, a typical one), this suggestion

fits very well the regulatory roles of C F T R and the

sulfonylurea receptor. (Interestingly, but also controver-

sially, M D R l - - l i k e C F T R - - h a s been suggested to be

an ATP channel [62].) T h e mechanism of interaction

between MDR1 and the volume-activated channel is

nonetheless not known, and the regulatory role has been

questioned also [63].

Osmotic swelling activates C L C - 2 in the oocyte expression

system, and structures important for this regulation have

been localized to its amino terminus [11]. CLC-2 does not

however underlie the typical volume-activated CI- channel

observed in numerous cell types. In contrast with CLC-2,

the typical volume-activated current displays an I->CI-

permselectivity, is outwardly rectifying, deactivates at

positive potentials, and depends on intracellular ATP. A

detailed study in glioma cells [64 °] has shown that this

channel has a single-channel conductance of 15-50pS

(depending on voltage), resolving a discrepancy between

single-channel [65] and noise [66,67] analysis, which

suggested a lower value. This current is blocked by

extracellular nucleotides in several cell types [68-70],

resembling the current seen upon overexpression of

plcln, a putative C I - c h a n n e l [71]. Indeed, antisense

oligonucleotides [70] or antibodies [72] directed against

plc1 n inhibit the volume-activated CI- current in native

cells. A convincing proof that plci n and the CI- channel

are one and the same is missing, however, and it is

possible that plcI n instead interacts with endogenous

CI- channels [72].

In the case of regulatory volume decrease, a loss of

intracellular organic osmolytes (taurine and

myo-inositol,

among others) may be equally important as a loss of KCI.

An increasing body of evidence suggests that this loss may

involve the same swelling-activated anion channel [73-75].

C h l o r i d e c u r r e n t s a s s o c i a t e d w i t h

c o - t r a n s p o r t e r s

Several co-transport or counter-transport molecules elicit

anion currents when expressed in

Xenopus

oocytes. For

instance, several members of an excitatory amino-acid/Na ÷

co-transporter family were shown to yield aspartate-

evoked CI- currents when expressed in

Xenopus

oocytes

[76,77]. T h e s e rather small currents were not stoichio-

metrically coupled to amino acid transport, suggesting

that these transporters may have an additional function

of a 'ligand-gated' CI- channel. This could dampen cell

excitability upon neurotransmitter re-uptake.

In addition, expression of a renal sodium-phosphate

co-transporter (NaPi-1) in

Xenopus

oocytes yielded quite

large, time-independent, slightly outwardly rectifying

anion currents with an I-->CI- selectivity [78], which

superficially resemble the currents reported, for example,

for CLC-3 [15]. This current was not directly d e p e n d e n t

on the substrates of the co-transporter (i.e. Na ÷ and

PO42-).

Finally, expression in

Xenopus

oocytes of the trout

erythrocyte anion exchanger (AE, or band 3), but not of

mouse anion exchanger AE1, induced increased taurine

fluxes and a sizeable CI-conductance that was only slightly

outwardly rectifying [75]. Analysis of chimeric transporters

has shown that an extracellular loop and carboxy-terminal

transmembrane domains are necessary for the induction of

these currents.

It has not yet been established whether these proteins pos-

sess dual transporter/channel activities, or whether their

expression in

Xenopus

oocytes activates endogenous chan-

nels. Oocytes possess a variety of endogenous CI- chan-

nels, w h i c h - - i n some cases, very r e p r o d u c i b l y - - m a y be

activated by the overexpression of foreign proteins that

are not themselves channels. Thus, expression of diverse

membrane proteins elicits hyperpolarization-activated an-

ion currents in

Xenopus

oocytes [20,79°,80]. T h e s e currents

were previously thought to be specific for phospholemman

[81]. Several criteria must be m e t before a cloned c D N A

can safely be assumed to encode a channel [29]. One

can, however, by no means exclude the possibility that

molecules have dual transporter/channel functions. This

scenario seems particularly likely for the aspartate-gated

CI- currents.

C o n c l u s i o n s

T h o u g h three gene families of CI- channels are known,

our molecular picture of CI- channels is far from complete.

Several important channels have not yet been cloned,

including the seemingly ubiquitous volume-activated

CI- channel (which may also conduct organic osmolytes),

and Ci- channels activated by intracellular CaZ+. Homol-

ogy cloning continues to uncover new m e m b e r s of the

C L C family, but many of these proteins have yet to

be assigned a function. In the absence of unambiguous

functional expression, it is even possible that some are

not even CI- channels. Many proteins overexpressed in

(6)

308 Signalling mechanisms

In addition to the well recognized roles of postsynaptic

ligand-gated CI- channels for signal transduction in

the CNS, the role of voltage-activated C I - c h a n n e l s

in controlling electrical excitability becomes increasingly

clear. This is most obvious for skeletal muscle [2], but

probably is also true for neuronal cells [26,27], and it

would not be surprising if genetic defects in brain CI-

channels turn out to be the basis for certain inherited

neurological disorders. T h e emerging complex regulations

of ion channels, as exemplified by the multiple functions

of C F T R , is likely to add an additional level of complexity

to neurobiology as well.

Acknowledgements

l thank Michael Pusch, Blanche Schwappach, and Klaus Steinmeyer for critical reading of the manuscript. Work in the author's laboratory is supported by the Deutsche Forschungsgemeinschaft, the US Muscular Dystrophy Association, and the Fonds der Chemischen lndustrie.

References and recommended reading

Papers of particular interest, published within the annual period of review, have been highlighted as:

• of special interest • • of outstanding interest

1. Rajendra S, Schofield PR: Molecular mechanisms of Inherited startle syndrome. Trends Neurosci 1995, 18:80-82.

2. Steinmeyer K, Lorenz C, Pusch M, Koch MC, Jentsch TJ: Multimeric structure of ClC-1 chloride channel revealed by mutations in dominant myotonla congenita. EMBO J 1994, 13:737-743.

3. Meyer-Kleine C, Steinmeyer K, Ricker K, Jentsch TJ, Koch MC: Spectrum of mutations In the major human skeletal muscle chloride channel gene (ClCN1) leading to myotonla. Am J Hum Genet 1995, 57:1325-1334.

4. Staley K, Soldo BL, Proctor WR: ionic mechanism of neuronal excitation by Inhibitory GABA A receptors. Science 1995, 269:977-981.

5. Lloyd SE, Pearce SHS, Fisher SE, Steinmeyer K, Schwappach B, • e Scheinman S J, Harding B, Bolino A, Devoto M, Goodyer P e t al.:

A common molecular basis for three Inherited kidney stone diseases. Nature 1996, 379:445-449.

Dent's disease, X-linked recessive nephrolithiasis, and X-linked recessive hy- pophosphatemic rickets (XLRH) are three kidney stone disorders that share the symptoms hypercalciuria and proteinuria. All three diseases are shown to be caused by inactivating mutations in the CLC-5 CI- channel that, in humans, are very kidney specific. These mutations include non-sense and mis-sense mutations, as well as an in-frame deletion of transmembrane do- main D2. Functional expression in Xenopus oocytes shows that these muta- tions specifically reduce or abolish CLC-5 currents. As proteins are mainly re-absorbed by the proximal tubule, it is important to note that CLC-5 is also expressed in that segment [19]; however, the mechanism by which CLC-5 inactivation leads to hyporcalciuria and proteinuria is unclear.

6. AI-Awqati Q: Chloride channels of Intracellular organelles. Curr Opin Cell Biol 1995, 7:504-508.

7. Jentsch TJ, Gi~nther W, Pusch M, Schwappach B: Properties of voltage-gated chloride channels of the ClC gene family.

J Physiol (Lond) 1995, 482:19S-25S.

8. Pusch M, Ludewig U, Rehfeldt A, Jentsch TJ: Gating of the • voltage-dependent chloride channel ClC-0 by the permeant

anion. Nature 1995, 373:527-531.

This paper provides evidence for a gating model that differs in a fundamental way from the intrinsic voltage sensor model for cation channels. Open proba- bility of the 'fast' gate of the Torpedo channel CLC-O is shown to depend with a slope of -50 mV to -60 mV per tenfold change of external, but not internal, e l - concentration. This effect and the nominal gating charge of 1 can be explained by a simple model in which CI- itself provides the gating charge. It senses the voltage as it crosses the membrane, and its presence at the end of the pore facilitates channel opening. Further, both conductance and gating show an anomalous mole fraction behaviour, and mutations decreasing the ion selectivity of conductance also decrease the ion selectivity of gating.

9. Middleton RE, Pheasant D J, Miller C: Purification, reconstitution, and subunlt composition of a voltage-gated chloride channel from Torpedo electroplax. Biochemistry 1994, 33:13189-13198. 10. Jentsch TJ, Steinmeyer K, Schwarz G: Primary structure of

Torpedo mermorata chloride channel Isolated by expression cloning in Xenopus oocytes. Nature 1990, 348:510-514. 11. Gr0nder S, Thiemann A, Pusch M, Jentsch TJ: Regions involved

in the opening of CLC-2 chloride channel by voltage and cell volume. Nature 1992, 360:759-762.

12. Uchida S, Sasaki S, Nitta K, Uchida K, Horita S, Nihei H, Marumo F: Locellzation and functional charactarlzation of rat kidney-specific chloride channel, ClC-KI. J C/in Invest 1995, 95:104-113.

13. Kieferle S, Fong P, Bans M, Vandewalle A, Jentsch TJ: Two highly homologous members of the ClC chloride channel family expressed in both rat and human kidney. Proc Nat/Acad Sci USA 1994, 91:6943-6947.

14. Adachi S, Uchida S, Ito H, Hata H, Hiroe M, Marumo F, Sasaki S: Two Isoforms of a chloride channel predominantly expressed In thick ascending limb of Henle's loop and collecting ducts of rat kidney. J Biol Chem 1994, 269:17677-17683.

15. Kawasaki M, Uchida S, Monkawa T, Miyawaki A, Mikoshiba K, Marumo F, Sasaki S: Cloning and expression of a protein kinase C-regulated chlodde channel abundantly expressed in rat brain neuronal cells. Neuron 1994, 12:597-604.

16. Van Slegtenhorst MA, Bassi MT, Borseni G, Wapenar MC, Ferrero GB, De Conciliis L, Rugarli El, Grillo A, Franco B, Zoghni HY, Ballabio A: A gene from the Xp22.3 region shares homology with voltage-gated chloride channels. Hum Mol Genet 1994, 3:547-552.

17 Borsani G, Rugarti El, Taglialatela M, Wong C, Ballabio A: Characterization of a human and murlne gene (CLCN3) sharing similarities to voltage-gated chloride channels and to a yeast Integral membrane protein. Genomics 1995, 27:131-141. 18. Fisher SE, Black GCM, Lloyd SE, Hatchwell E, Wrong O,

Thakker RV, Craig IW: Isolation and partial characterization of a chloride channel gene which Is expressed In kidney and is a candidate for Dent's disease (an X-linked hereditary nephrollthlasls). Hum Mol Genet 1994, 3:2053-2059. 19. Steinmeyer K, Schwappach B, Bens M, Vandewalle A, Jentsch TJ:

Cloning and functional expression of rat CLC-5, a chloride channel related to kidney disease. J Biol Chem 1995, 270:31172-31177.

20. Brandt S, Jentsch TJ: CLC-6 and CLC-7 are two novel broadly expressed members of the CLC chloride channel family. FEBS Lett 1995, 377:15-20.

21. Pusch M, Steinmeyer K, Koch MC, Jentsch TJ: Mutations in dominant human myotonia congenlta drastically alter the voltage dependence of the CIC-1 chloride channel. Neuron 1995, 15:1455-1463.

22. Fahlke C, R~idel R, Mitrovic N, Zhou M, George AL: An aspertlc acid residue Important for voltage-dependent gating of human muscle chloride channels. Neuron 1995, 15:463-472. 23. Komwatana P, Dinudom A, Young JA, Cook Dh Characterization

of the e l - conductance in the granular duct cells of mouse mandibular glands. Pflugers Arch 1994, 428:641-647 24. Fritsch J, Edelman A: Modulation of the hyperpolarlzation-

activated el- current in human intestinal T84 cells by

phosphorylation. J Physiol (L ond) 1996, 490.1:115-128. 25. Arreola J, Park K, Melvin JE, Begenisich T: Three distinct chloride

channels control anion movements in rat parotld aclnar calls.

J Physiol (Lond) 1996, 490:351-362.

26. Staley K: The role of an inwardly rectifying chloride conductance in postaynaptlc inhibition. J Neurophysiol 1994, 72:273-284.

27. Smith RL, Clayton GH, Wilcox CL, Esudero KVV, Staley K J: Differential expression of an Inwardly rectifying chloride conductance in rat brain neurons: a potential mechanism for cell-specific modulation of postsynaptic inhibition. J Neurosci

1995, 15:4057-4067.

28. Chesnoy-Marchais D, Fritsch J: Activation by hyperpolarlzatlon and atypical osmosensitivlty of a e l - current In rat osteoblestlc cells. J Membr Bio11994, 140:173-188.

(7)

30. Greene JR, Brown NH, DiDomenico BJ, Kaplan J, Eide DJ: The GEF.1 gene of Saccheromyces cerevisiee encodes an integral membrane protein; mutations in which have effects on respiration and Iron-limited growth. Mo/Gen Genet 1993, 241:542-553.

31. Inagaki N, Gonoi T, Clement JP, Namba N, Inazawa N, Gonz~lez G, Aguilar-Bryan L, Seino S, Bryan J: Reconstitutlon of IKMp: an inward rectifier subunlt plus the sulfonylurea receptor. Science 1995, 270:1166-1170.

32. Egan M, Flotte T, Afione S, Solow R, Zeitlin PL, Carter B J, Guggino WB: Defective regulation of outwardly rectifying Cl- channels by protein klnase A corrected by insertion of CFTR.

Nature 1992, 358:581-584.

33. Schwiabert EM, Egan ME, Hwang TH, Fulmer SB, Allen SS, • Cutting GR, Guggino WB: CFTR regulates outwardly rectifying

chloride channels through an autocrlne mechanism involving ATR Cell 1995, 81:1063-1073.

This work suggests that CFTR, in addition to being a linear CI- channel, also regulates an outwardly rectifying CI- channel. This regulation depends on the efflux of ATP from the cells after stimulation by protein kinase A. ATP-acavenging systems applied to the extracellular surface prevents this activation, which presumably is mediated via purinergic P2u receptors. The mechanism for this regulation (e.g. direct protein-protein interactions, or second messengers) is not yet clear. Under appropriate conditions, CFTR stimulation elicits currents carried by ATP anions, which either flow through a channel regulated by CFTR or through CFTR itself [35]. The latter model would reconcile the channel and regulator function of CFTR, but is highly controversial (see [36,37]).

34. Jovov B, Ismailov II, Berdlev BK, Fuller CM, Sorscher E J, Dedman JR, Kaetzel MA, Benos D J: Interaction between cystic fibrosis transmembrane conductance regulator and outwardly rectifying chlodde channels. J Biol Chem 1995, 270:29194-29200.

35. Reisin IL, Prat AG, Abraham EH, Amara JF, Gregory R J, Ausiallo DA, Cantiello HF: The cystic fibrosis trensmembrane conductance regulator Is a dual ATP and chloride channel.

J Biol Chem 1994, 269:20584-20591.

36. Takahashi T, Matsushita K, Welsh M J, Stokes JB: Effect of cAMP on intracellular and extracellular ATP content of Cl- secratlng epithelia and 3T3 fibroblasts. J Biol Chem 1994, 269:17853-17857.

37. Reddy MM, Quinton PM, Haws C, Wins J J, Grygorczyk R, Tabcharani JA, Hanrahan JW, Gunderson KL, Kopito RR: Failure of the cystic fibrosis transmembrane conductance regulator to conduct ATR Science 1996, 271:1876-1879.

38. Stutts MJ, Rtz JG, Paradiso AM, Boucher RC: Multiple modes of regulation of airway epithelial chloride secretion by extracelluler ATR Am J Physiol 1994, 267:C1442-C1451. 39. Fulmer SB, Schwiebert EM, Morales MM, Guggino WB,

Cutting GR: Two cystic fibrosis transmembrane conductance regulator mutations have different effects on both pulmonary phenotype and regulation of outwardly rectified chloride currents. Proc Natl Acad Sci USA 1995, 92:6832-6836. 40. Stutts M J, Canessa CM, Olsen JC, Hamrick M, Cohn JA, • Rossier BC, Boucher RC: CFTR as a cAMP-dependent regulator

of sodium channels. Science 1995, 269:847-850.

This important work investigates the effects of CFTR on the epithelial Na + channel ENaC in cells transfected with all three ENaC subunits. Using epithelial cell layers of transfected MDCK cells in CI--free solutions, it is shown that forskolin (which raises cAMP) stimulates short-circuit current in the absence of CFTR. This current is mediated in large part by ENaC in the apical membrane. In cells co-transfected with CFTR, in contrast, cAMP causes a slight inhibition. Although these effects are large, whole-cell cur- rent measurements in transfected 3T3 cells yield similar, but much less im- pressive results. Broadly similar results have been obtained in the Xenopus

oocyte expression system [41 ]. The mechanism of interaction between both channel molecules is still unclear. The results of this work are highly relevant for cystic fibrosis, as an increase in Na + re-absorption contributes to lung disease in the patients studied.

41. Mall M, Hipper A, Greger R, Kunzelmann K: Wild type but not ~F508 CFTR Inhibits Na + conductance when coexpressed in

Xenopus oocytes. FEBS Lett 1996, 381:47-52.

42. Valverde MA, O'Brien JA, Sep01veda FV, Ratctiff RA, Evans M J, Colledge WH: Impaired cell volume regulation in intestinal crypt epithelia of cystic fibrosis mice. Proc Nat/Acad Sci USA 1995, 92:9038-9041.

43. Carson MR, Travis SM, Welsh M J: The two nucleotide-blndlng • domains of cystic fibrosis transmombrane conductance

regulator (CFTR) have distinct functions in controlling channel activity. J Bio/Chem 1995, 270:1711-1717.

Using patch-clamp analysis of transfected cells, the analysis of a number of CFTR point mutants, and of ATP and non-hydrolyzable analogs indi- rectly, suggests that ATP hydrolysis at NBF1 initiates burst activity (during which the channel fluctuates many times between open and closed states), whereas hydrolysis of ATP at NBF2 terminates the burst. Non-hydrolyzable ATP analogs prolong the duration of only some burets. These conclusions are broadly similar to those presented in [44"].

44. Gunderson KL, Kopito RR: Conformational states of CFTR • . associated with channel gating: the role of ATP binding and

hydrolysis. Cell 1995, 82:231-239.

Heavy electrical filtering (10 Hz) allows the detection of a previously unknown open state (O 2) of CFTR reconstituted into lipid bilayere. 02 differs by only 1 pS from the other open state O1. Statistical analysis shows that channel gating predominantly occurs in an asymmetric cycle C->O!--~O2--)C, where C is the closed state. This indicates an input of energy, which is most likely supplied by ATP hydrolysis. Non-hydrolyzable compounds lock CFTR in the O1 state. The predominant irreversible step is the O1--)O 2 transition, which is coupled to ATP hydrolysis at NBF2. These conclusions are supported by the analysis of point mutants. Thus, this elegant study suggests that each cycle of CFTR opening is coupled to the hydmlysia of one ATP at NBF2 (and possibly one ATP at NBF1, the role of which is less clear). The appar- ent contrast with a study on intact cells [43"] - which suggests that ATP hydrolysis at NBF2 terminates a burst of activity - may be attributable, in part, to the heavy filtering employed here, which may not allow the detection of fast closures.

45. Schultz BD, Venglarik C J, Bridges R J, Frizzell RA: Regulation of CFTR Cl- channel gating by ADP and ATP analogues. J Gen Physiol 1995, 105:329-361.

46. Wilkinson D J, Mansoure MK, Watson PY, Smit LS, Collins FS, Dawson DC: CFTR: the nucleotide binding folds regulate the accessibility of the activated stste. J Gen Physiol 1996, 107:103-119.

47. Hwang TC, Nagel G, Naim AC, Gadsby DC: Regulation of the gating of cystic fibrosis transmembrane conductance regulator Cl channels by phosphorylation and ATP hydrolysis. Proc Natl Acad Sci USA 1994, 91:4698-4702.

48. Ko YH, Pedersen PL: The first nucleoflde binding fold of the cystic fibrosis tmnsmembrane conductance regulator can function as an active ATPase. J Bio/Chem 1995, 270:22093-22096.

49. Jensen TJ, Loo MA, Pind S, Williams DB, Goldberg AL, Riordan JR: Multiple proteolytic systems, Including the proteasome, contribute to CFTR processing. Ceil 1995, 83:129-135.

50. Ward LW, Omura S, Kopito RR: Degradation of CFTR by the ubiqultln-proteasome pathway. Ceil 1995, 83:121-127. 51. Anderson MP, Gregory R J, Thompson S, Souza DW, Paul S,

Mulligan RC, Smith AE, Welsh M J: Demonstration that CFTR Is a chlodde channel by alteration of its anion selectivity. Science 1991, 253:202-205.

52. McDonough S, Davidson N, Lester HA, McCarty NA: Novel pore- lining residues In CFTR that govem permeation and open- channel block. Neuron 1994, 13:623-634.

53. Carroll TP, Morales MM, Fulmer SB, Allen SS, Rotte TR,

Cutting GR, Guggino WB: Alternate translation initiation codons can create functional forms of cystic fibrosis transmembrane conductance regulator. J Biol Chem 1995, 270:11941-11946. 54. Valverde MA, Diaz M, Sep01veda FV, Gill DR, Hyde SC,

Higgins CF: Volume-regulated chloride channels associated with the human multldrug-reslstance P-glycoprotein. Nature 1992, 355:830-833.

55. Mori XK, Bond TD, Loo TW, Clarke DM, Bear CE: Failure of P-glycoprotein (MDR1) expressed in Xenopus oocytes to produce swelling-activated chlodde channel activity. J Physiol (Lond) 1995, 468.3:707-714.

56. Rasola A, Galietta I_IV, Gruanert DC, Romeo G: Volume-sensitive chloride currents in four epithelial cell lines are not directly correlated to the expression of the MDR-1 gene. J Biol Chem

1995, 269:1432-1436.

57. De Greef C, Sehrer J, Viana F, Van Acker K, Eggermont J, Martens L, Raeymaekere L Droogmans G, Nilius B: Volume-activated chloride currents are not correlated with P-glycoprotein expression. Biochem J 1995, 307:713-718. 58. Luckie DB, Krouse ME, Harper KL, Law TC, Wine J J: Selection

(8)

310 Signalling mechanisms

K + and Cl- currents in NIH/3T3 ceils. Am J Physiol 1994,

267 :C650-C658.

59. Ehring GR, Osipchuk YV, Cahalan MD: Swelling-activated chlodde channels In multidrug-sensitive and -resistant cells.

J Gen Physio11994, 104:1129-1161.

60. Kunzelmann K, Slotki IN, Koslowski T, Ausiello DA, Greger R, Cabantchik Zh Effects of P-glycoproteln expression on cyclic

AMP and volume-activated ion fluxes and conductances in HT-29 colon edenocsrcinoma cells. J Cell Physio/1994,

161:393-406.

61. Hardy SP, Goodfellow HR, Valverde MA, Gill DR, Sept~lveda IV, Higgins CF: Protein klnase C-mediated phosphorylation of the human multidrug resistance P-glycoproteln regulates cell volume-activated chloride channels. EMBO J 1995, 14:68-75. 62. Abraham EH, Pret AG, Gerweck L, Seneveretne T, Arceci RJ,

Kramer R, Guidotti G, Cantiello HF: The muItidrug resistance

(mdrl) gene product functions as an ATP channel. Proc Nat/ Aced Sci USA 1993, 90:312-316.

63. Tominaga M, Torninaga T, Miwa A, Okada Y: Volume-sensitive chloride channel activity does not depend on endogenous P-glycoprotein. J Bio/Chem 1995, 270:27887-27893. 64. Jackson PS, Strange K: Single-channel properties of a volume- • sensitive anion conductance. Current activation occurs by

abrupt switching of closed channels to an open state. J Gen Physio/1995, 105:643-660.

This detailed study resolves a controversy regarding the single-channel con- ductance of volume-sensitive anion channels. Because open probability does not increase in a graded fashion, stationary noise analysis incorrectly sug- gests a small single-channel conductance, whereas single-channel data and non-stationary noise analysis consistently reveal a 15 pS to 50 i0S channel (at 0 mV and +120 mV, respectively). Once 'activated', channel open probability is 1. It is speculated that this 'activation' may be attributable to insertion of porin-like molecules into the plasma membrane, in a model similar to that for plci n [70]. Together with its companion paper [68], this is a thorough biophysical analysis of swelling-activated CI- channels, which probably also allow the flow of organic osmolytes [73].

65. Solc CK, Wine J J: Swelling-induced and depolarization-induced Cl- channels In normal and cystic fibrosis epithelial cell. Am J Physiol 1991,261 :C658-C674.

66. Lewis RS, Ross PE, Cahalan MD: Chloride channels activated

by osmotic stress in T lymphocytes. J Gen Physiol 1993, 101:801-803.

67. Nilius B, Oike M, Zahradnik I, Droogmans G: Activation of a CI- current by hypotonic volume Increase in human endothelial cells. J Gen Physiol 1994, 103:787-805.

68. Jackson PS, Strange K: Characterization of the voltage- dependent properties of a volume-sensitive anion conductance. J Gen Physiol 1995, 105:661-677.

69. Ackerman M J, Wickman KD, Clapham DE: Hypotonlcity activates a native chloride currents in Xenopus oocytes. J Gen Physiol

1994, 103:153-179.

70. Gschwentner M, Nagl UO, W~II E, Schmarda A, Ritter M, Paulmichl M: Antlsense ollgonucleotldes suppress cell-volume-

induced activation of chloride channels. Pflugers Arch 1995, 430:464-470.

71. Paulmichl M, Li Y, Wickman K, Ackerman M, Peralta E, Clapham DE: New mammalian chloride channel identified by

expression cloning. Nature 1992, 356:238-241. 72. Krapivinsky GB, Ackerman M J, Gordon F_A, Krapivinsky LD,

Clapham DE: Molecular characterization of a swelling-induced chloride conductance regulatory protein, plcl n. Cell 1 g94, 76:439-448.

73. Strange K, Jackson PS: Swelling-activated organic osmolyte efflux: a new role for anion channels. Kidney Int 1995, 48:994-1003.

74. Roy G: Amino acid current through anion channels in cultured human glial cells. J Membr Bio11995, 147:35-44.

75. Fi6vet B, Gabillat N, Borgese F, Motels R: Expression of band 3 anion exchanger induces chloride current and taurine transport: structure-function analysis. EMBO J 1995, 14:5158-5169.

76. Fairman WA, Vandenberg RJ, Arriza JL, Kavanaugh MP, Amara SG: An excitatory amino-acid transporter with properties of e ligand-gated chloride channel. Nature 1995, 375:599-603. 77. Wadiche JI, Amara SG, Kavanaugh MP: Ion fluxes associated

with excitatory amino acid transport. Neuron 1 g95, 15:721-728.

78. Busch AE, Schuster A, Waldegger S, Wagner CA, Zempel G, Broer S, Biber J, Murer H, Lang F: Expression of s renal type I sodium/phosphate-transporter (NaPI-1) induces a conductance in Xenopus oocytes permeable for organic and inorganic anions. Proc Natl Acad Sci USA 1 g96, in press. 79. Tzounopoulos T, Maylie J, Adeiman JP: Induction of endogenous • channels by high levels of heterologous membrane proteins in

Xenopus oocytes. Biophys J 1995, 69:904-908.

This paper shows that overexpression of many membrane proteins, including those that do not function as channels, induces slowly hyperpolarization- activated currents in Xenopus oocytes. Thus, these currents, which were thought to be specific for phospholemman [81], are attributable to an acti- vation of an endogenous Xenopua oocyte channel. Similar observations have been made for non-functional CLC proteins [2,20]. The experiments strongly suggests that hyperpolarization opens a non-specific cation channel, and that the inflow of Ca 2+ activates a Ca2+-dependent CI- channel. Thus, this paper investigates one of the pitfalls encountered when studying functional expression of CI- channels.

80. Shimbo K, Brassard DL, Lamb RA, Pinto LH: Viral and cellular small integral membrane proteins can modify ion channel endogenous to Xenopus oocytes. Biophys J 1995, 69:1819-1829.

81. Moorman JR, Palmer C J, John JE, Durieux ME, Jones LR: Phospholemman expression induces a hyperpolsrlzatlon- activated chloride current in Xenopus oocytes. J Biol Chem 1992, 267:14551-14554.

Referencias

Documento similar

In the “big picture” perspective of the recent years that we have described in Brazil, Spain, Portugal and Puerto Rico there are some similarities and important differences,

The Dwellers in the Garden of Allah 109... The Dwellers in the Garden of Allah

that when looking at the formal and informal linguistic environments in language acquisition and learning it is necessary to consider the role of the type of

A January effect is present in nominal terms, but it is negative in real and dollar adjusted returns.. The January effect present in nominal terms cannot be explained by

Keywords: Metal mining conflicts, political ecology, politics of scale, environmental justice movement, social multi-criteria evaluation, consultations, Latin

Expected and observed 95% CL limits on the s-channel production cross section times branching fraction for H + → tb as a function of the charged Higgs boson mass, in the (a)

Díaz Soto has raised the point about banning religious garb in the ―public space.‖ He states, ―for example, in most Spanish public Universities, there is a Catholic chapel

In this work, we have undertaken the study of SUZ12, a Polycomb group protein and the microRNAs (miRNA) expressed by the oncogenic Epstein Barr Virus (EBV) in