Renal TRPathies

July 24, 2017 | Autor: Alexander Dietrich | Categoría: Kidney diseases, Humans, Magnesium, Clinical Sciences, Homeostasis, Podocytes, The American, Podocytes, The American
Share Embed


Descripción

BRIEF REVIEW

www.jasn.org

Renal TRPathies Alexander Dietrich,* Vladimir Chubanov,† and Thomas Gudermann† *Institute of Pharmacology and Toxicology, School of Medicine, University of Marburg, Marburg, Germany; and † Walther-Straub-Institute of Pharmacology and Toxicology, University of Munich, Munich, Germany

ABSTRACT Many ion channels and transporters are involved in the filtration, secretion, and resorption of electrolytes by the kidney. In recent years, the superfamily of transient receptor potential (TRP) ion channels have received deserved attention because mutated TRP channels are linked to human kidney diseases. This review focuses on two TRP members—TRPC6 and TRPM6 —and their functions in the kidney. Gain-of-function mutations in TRPC6 are the cause for progressive kidney failure with urinary protein loss such as FSGS. Thus, TRPC6 is an essential signaling component in a functional slit diaphragm formed by podocytes around the glomerular capillaries. Loss-of-function mutations in TRPM6 are a molecular cause of hypomagnesemia with secondary hypocalcemia, suggesting that TRPM6 is critically involved in transcellular Mg2⫹ transport in the kidney. Here, we highlight how recent studies analyzing function and expression of these channels in the kidney improve our mechanistic understanding of TRP channel function in general and pave the way to new, promising therapeutic strategies to target kidney diseases such as FSGS and hypomagnesemia with secondary hypocalcemia. J Am Soc Nephrol 21: 736 –744, 2010. doi: 10.1681/ASN.2009090948

One key function of the kidneys is the ultrafiltration of plasma by glomeruli to dispose of metabolic end products, excess electrolytes, and water. A vast array of ion channels and transporters are critical for filtration, secretion, and resorption of electrolytes to maintain homeostasis. In recent years, investigations have focused on several members of the large transient receptor potential (TRP) family of ion channels, because mutations in genes encoding members of three different TRP ion channel subfamilies have been linked to human kidney diseases.1 Mutations in PKD1 (encoding TRPP1) and PKD2 (coding for TRPP2) occur at high frequency in individuals with autosomal dominant polycystic kidney disease.2,3 The latter mutations and the pathophysiology of polycystic kidney disease are not discussed further in this overview because the topic has been covered by numerous insightful review articles.4 –7 Missense and nonsense mutations in 736

ISSN : 1046-6673/2105-736

the gene coding for TRPC6 segregate with autosomal dominant FSGS, a kidney disease that leads to progressive renal failure.8,9 Loss-of-function mutations in TRPM6 are also associated with hypomagnesemia with secondary hypocalcemia (HSH), a rare autosomal recessive disorder.10 –12 The involvement of TRP channel mutations in hereditary kidney disease has shed new light on the molecular pathogenesis of renal failure and significantly enhanced our appreciation of the physiologic roles of TRP channels on tubular function. In this review, we focus exclusively on TRPC6 and TRPM6.

TRPC6 AND PODOCYTE FUNCTION

TRPC6 is a nonselective cation channel and one of the seven members of the clas-

sical transient receptor potential (TRPC) family, which can be divided into subfamilies on the basis of amino acid similarity. Whereas TRPC1 and TRPC2 are almost unique, TRPC4 and TRPC5 share approximately 64% amino acid identity. TRPC3, TRPC6, and TRPC7 form a structural and functional subfamily displaying 65 to 78% identity at the amino acid level and share a common activator, diacylglycerol (DAG).13 DAG is produced by phospholipase C isozymes activated after agonist binding to appropriate receptors, such as the interaction of angiotensin II (AngII) with AT1 receptors. Like all TRPC family members, TRPC6 harbors an invariant sequence, the TRP box (containing the amino acid sequence EWKFAR), in its C-terminal tail as well as three ankyrin repeats in the N-terminus (Figure 1, A and B). The predicted transmembrane topology is similar to that of other TRP channels with intracellular N- and C-termini, six membrane-spanning helices (S1 through S6), and a presumed pore-forming loop (P) between S5 and S6 (Figure 1, A and B). As deduced from Northern blot analyses, TRPC6 channels are most prominently expressed in lung tissues,14 where they Published online ahead of print. Publication date available at www.jasn.org. A.D. and V.C. contributed equally to this work. Correspondence: Dr. Thomas Gudermann, Walther-Straub-Institute of Pharmacology and Toxicology, University of Munich, Goethestrasse 33, D-80336 Munich, Germany. Phone: ⫹49-89-218075700; Fax: ⫹49-89-2180-75701; E-mail: thomas. [email protected] Copyright 䊚 2010 by the American Society of Nephrology

J Am Soc Nephrol 21: 736–744, 2010

www.jasn.org

A

C Urine

Podocyte foot process

PIP2 PLCε

PM

S1 S2 S3 S4 S5

DAG

TRPC6

S6

+

+

Podocin

Ca2+

+

Nephrin GBM Fenestrated endothelium

10

S1-5

P

735

S6

CC

931

Q 8 E889K 97 K

CC

P

L

N

G

EW

431

A

80

A

9S 12 5S

A

L7

1

K8 R 74* 89 5C

T

P1 12

S2

70

Q M 1 N 32T 14 3S

B

KF AR

Blood

Figure 1. Structural features of TRPC6 and its function in podocytes. (A) Topology of TRPC6 in the plasma membrane (PM) indicating transmembrane regions (S1 through S6) and the predicted pore domain (P). Two glycosylated sites in TRPC6 are indicated by covalently bound carbohydrates (gray).105 (B) Localization of the identified mutations in patients with nephrotic syndromes. A, ankyrin repeat; L, lipid/trafficking domain; CC, predicted coiled-coil domain; EWKFAR, conserved TRP box motif. Amino acid changes are indicated by the single-letter code and a number for the exact position in the protein; *a stop codon (truncation mutant). (C) Podocyte foot process with important signaling molecules. Mechanical activation of podocin or receptor-stimulated activation of phospholipase C-␧ (PLC␧) activates TRPC6 channels, leading to actin (red line) reorganization and subsequent closure of the slit diaphragm by homophilic nephrin interactions (arrows). See text for more details. PIP2, phosphatidylinositol-3,4-bisphosphate; GBM, glomerular basement membrane.

play an important role in vascular and pulmonary smooth muscle cells.15 In the kidney, TRPC6 and TRPC3 are detected along the glomerulus and the collecting duct.16 Heteromeric complexes comprising TRPC3 and TRPC6 in podocytes co-localize with aquaporin 2 in principal cells of the collecting duct.17 In polarized cultures of M1 and IMCD-3 collecting duct cells, however, TRPC3 localizes exclusively to the apical domain, whereas TRPC6 is on both basolateral and apical membranes.18 Native TRPC3/6 heteromers are also found in MDCK cells.19 Sclerotic lesions in the subtotal of renal glomeruli characterize FSGS. In six families with hereditary forms of FSGS, two research groups independently identified gain-of-function mutations in the TRPC6 channel (Figure 1C) leading to enhanced calcium influx (P112Q8) or inJ Am Soc Nephrol 21: 736 –744, 2010

creased current amplitudes in electrophysiological recordings of expressed ion channels (R895C and E897K9). TRPC6 expression in the kidney localizes to glomeruli and tubuli8 as well as podocytes.9 Three other mutations identified by Reiser et al.20 (N143S, S270T, and K874*) do affect current amplitudes but allow channels to stay open longer than wildtype TRPC6 channels. Moreover, channel density at the plasma membrane is significantly higher in HEK293 cells expressing the P112Q mutant. Recently, a seventh mutation (Q889K) was found in 31 Chinese pedigrees with late-onset FSGS.21 HEK293 cells expressing the mutant channel protein show significantly higher Ca2⫹ influx after stimulation by a membrane-permeable analogue of DAG—an observation attributable to a significantly higher level of expression of

BRIEF REVIEW

the mutant protein. TRPC6 mutation analysis in 130 Spanish patients with FSGS identified three additional, as yet functionally uncharacterized TRPC6 missense mutations, two in the N-terminus close to the ankyrin repeats (G109S and N125S) and a third one adjacent to the TRP domain (L780P).22 Most notably, TRPC6 mutations are also described in children and adults with nonfamilial FSGS. An 11th missense mutation (M132T) that shows a mean inward calcium current 10-fold larger than that of wild-type TRPC6 was recently identified in childhood FSGS.23 In aggregate, the mutations described so far indicate a frequency of 6% of TRPC6 mutations in familial FSGS and approximately 2% in sporadic cases.22 All mutations map to the terminal domains of the TRPC6 protein (see Figure 1). Six N-terminal missense mutations (G109S, P112Q, M132T, N125S, N143S, and S270T) locate in or near ankyrin repeats and an adjacent lipid/trafficking domain. The ankyrin domains are responsible for self-association of TRPC homomers,24 whereas the lipid-binding domain binds DAG and participates in the translocation of the channel to the plasma membrane.25 Whereas the L780P mutation is located near to the EWKFAR motif conserved in all TRP channels, four additional mutations (K874*, Q889K, R895C, and E897K) map to a predicted coiled-coil domain at the C-terminus. It is tempting to speculate that gain-of-function phenotypes occur after more efficient multimerization and/or translocation to the plasma membrane or impaired ion channel endocytosis, as well as by blocking the binding of an inhibitory protein to a predicted Cterminal coiled-coil domain; however, so far, there are no hard experimental data to support any of these speculations. Other proteins mutated in patients with FSGS or related diseases include nephrin, podocin, ␣4-actinin, phospholipase C-␧, laminin ␤2, and others.26 Most of these mutations lead to earlyonset disease,27 underscoring their indispensable role in filtration at the slit diaphragm. The TRPC6 protein directly interacts with key structural and signalRenal TRPathies

737

BRIEF REVIEW

www.jasn.org

ing proteins of the podocyte, podocin and nephrin. The podocin homologue mec-2 contributes to mechanosensation in Caenorhabditis elegans by linking mechanosensitive ion channels to the cytoskeleton; therefore, podocin might detect mechanical forces exerted by the glomerular filtration process, resulting in TRPC6 activation. Accordingly, heterologously expressed podocin regulates TRPC6 activity in a cholesterol-dependent manner.28 Moreover, mutations in the podocin gene give rise to hereditary nephrotic syndromes in humans, compatible with an important role of podocin as a mechanosensor at the slit diaphragm. The recently proposed mechanosensitivity of TRPC6 channels29 is not detectable at physiologically relevant pressures of ⬍80 mmHg,30 and three additional, independent studies failed to demonstrate that TRPC6 per se is mechanosensitive.31–33 The second TRPC6-interacting protein, nephrin, is an essential constituent of the slit diaphragm contributing to actin reorganization in podocytes by engaging a phosphoinositide 3-kinase pathway34 subsequent to phosphorylation by the Src family protein tyrosine kinase Fyn.35 The functional relevance of TRPC6 phosphorylation by Fyn36 was called into question on the basis of functional analyses of TRPC6 phosphorylation site mutants.37 Disruption of the slit diaphragm architecture in nephrin-deficient mice leads to overexpression and mislocalization of TRPC6 in podocytes9 and supports the concept that TRPC6 integrates into an organized signaling complex in podocytes with nephrin, podocin, and probably AT1 AngII receptors.1 Because TRPC6 is a Ca2⫹-permeable ion channel, one may assume that Ca2⫹ influx through activated channels induces actin reorganization, leading to a more flattened cell shape and a closure of the slit diaphragm mediated by the homophilic interaction of nephrin molecules (Figure 1C). Moreover, lossof-function mutations in phospholipase C-␧, a central signaling molecule that cleaves phosphatidylinositol-3,4bisphosphate to generate DAG, a potent lipid activator of TRPC6, in re738

sponse to receptor stimulation were identified by screening patients with early-onset nephrotic syndromes such as FSGS and diffuse mesangial sclerosis.38 In a mechanistic model, elevated BP with increased levels of AngII or increased shear stress engage AT1 receptors, activating TRPC6 channels subsequently to close the slit diaphragm; however, at first glance, the lack of an overt renal phenotype in TRPC6⫺/⫺ mice does not conform to the aforementioned model. However, the closely related TRPC channel, TRPC3, is upregulated in vascular and airway smooth muscle cells.39,40 Because endogenous TRPC3 expression in podocytes has been demonstrated,17,41 compensatory upregulation of TRPC3 is an intriguing possibility to explain the lack of phenotype in mice and the absence of TRPC6 loss-of-function mutations in patients with FSGS. An essential role of TRPC channels has been invoked in cardiac hypertrophy and remodeling. Ca2⫹-mediated activation of calcineurin induces the hypertrophic response by downstream nuclear factor of activated T cells (NFAT) transcription factors, which are necessary and sufficient for the induction of cardiac hypertrophy.42 Interestingly, similar mechanisms operate in cultured podocytes heterologously expressing three TRPC6 gain-of-function mutations (P112Q, R895C, and E897K). All three channel mutants result in enhanced basal NFAT-mediated transcription in several cell lines including cultured podocytes.43 Although the genes transcribed after activation of NFAT in podocytes are still unknown, a similar scenario to the one worked out for the heart is conceivable. It should not go unnoticed, however, that increased levels of TRPC6 expression in cultured podocytes result in a disruption of the actin cytoskeleton, and in vivo delivery of cDNA encoding TRPC6 into mice induces proteinuria.44 Moreover, disruption of the ultrafiltration process at the slit diaphragm might also be induced by Ca2⫹ overload in podocytes, leading to apoptosis. Although experimental data stringently confirming these hypotheses are lacking,45 AngII induces apoptosis in rat podocytes.46

Journal of the American Society of Nephrology

In summary, little is known about the exact role of TRPC6 in podocytes or the ultrafiltration process. Nevertheless, our understanding of the molecular processes governing podocyte function is increasing and may lead to the identification of therapeutic strategies to treat FSGS and related diseases.

ROLE OF TRPM6 CHANNELS IN RENAL MAGNESIUM HOMEOSTASIS

Divalent cation-selective outwardly rectifying currents, which are induced upon removal of intracellular Mg2⫹, are a general feature of various invertebrate cells and have been identified in all mammalian cells examined so far.47–53 These currents are referred to as magnesiuminhibited currents (MIC) or magnesium nucleotide-regulated metal ion currents (MagNuM).47– 49,54,55 TRPM6 and TRPM7 (melastatin-related members of the TRP gene family) were subsequently defined as molecular correlates of MIC/MagNuM channels. The current scientific literature reflects a broad spectrum of speculations on the physiologic roles of TRPM6 and TRPM7; however, compelling initial information about the physiologic relevance of TRPM6 and TRPM7 was obtained by clinical and molecular studies on a hereditary disorder associated with renal Mg2⫹ wasting. Mutations in the human TRPM6 gene give rise to autosomal recessive HSH.10,56 Here, we describe the latest progress made toward the molecular characterization of TRPM6/7 in light of their possible role in renal magnesium handling. Other aspects of TRPM channels are addressed in a number of excellent review articles.7,12,57– 64 A few years ago, two independent groups showed that loss-of-function mutations in the human TRPM6 gene are the molecular cause of HSH.10,56 HSH is an autosomal recessive disease that manifests in early infancy with generalized convulsions or symptoms of increased neuromuscular excitability including muscle spasms and tetJ Am Soc Nephrol 21: 736 –744, 2010

www.jasn.org

P1017R

A

N

B 1

2

3

4

5

3

2

6

4

1

N

5

6

C Kinase I174R

C

4

5



M7 6

3 2

S141L

C

M6

1 6

– – ++

2 M7 3 4

5

– 5

C 6

1

N

M6

1

C

4 3

2

N

T354P

C707Y

Figure 2. Architecture of TRPM6/M7 kinase-linked channel complexes. (A) Schematic domain topology of TRPM6 channels. A hexahelical channel segment of TRPM6 (1 through 6) is fused to a C-terminal serine/threonine protein kinase domain (Kinase). Positions of five missense mutations identified in patients with HSH66,77,78 are indicated by red dots. (B) Pore-forming segment of heterotetrameric channel complexes formed by TRPM6 and TRPM7. A short stretch between transmembrane helices 5 and 6 contains a putative pore-forming loop; four subunits of the tetramer are thought to contribute to a common channel pore. Four negatively charged glutamates (⫺) form a common highaffinity biding site for permeant ions such as Mg2⫹ and Ca2⫹ (⫹⫹).73

any.10,12,56,65,66 Clinical examination at the time of manifestation typically reveals very low Mg2⫹ (0.1 to 0.3 mM) and low Ca2⫹ (1.0 to 1.6 mM) serum concentrations compared with normal (0.7 to 1.1 mM [Mg2⫹] and 2.2 to 2.9 mM [Ca2⫹]). Of note, patients with HSH exhibit a high fractional excretion of Mg2⫹ of approximately 3%, whereas healthy individuals preserve Mg2⫹ by reducing fractional excretion to ⬍1% during Mg2⫹ deficiency, thus indicating that renal Mg2⫹ handling is primarily affected in individuals with HSH. Hypocalcemia seems to be caused by diminished parathyroid hormone release as a result of profound hypomagnesemia.56,67,68 Relief of HSH symptoms is achieved by administration of high dosages of Mg2⫹. Despite this treatment, serum Mg2⫹ levels remain in the subnormal range (approximately 0.5 mM). Delay in diagnosis and therapy results in permanent neurologic deficits or even fatality.10,12,56,65,66 TRPM6 belongs to the TRPM family of cation channels whose members exhibit a marked diversity in ion selectivity and activation mechanisms.60,62,69 –74 TRPM6 displays approximately 50% amino acid sequence identity with its homologous J Am Soc Nephrol 21: 736 –744, 2010

family member, TRPM7.10,11,47,50,56 Both TRPM6 and TRPM7 encode cation channel subunits characterized by the covalent linkage to a C-terminal serine/threonine protein kinase domain resembling elongation factor 2 kinase and other ␣-kinases (Figure 2A).75 It is generally assumed that the architecture of the pore-forming segment in TRPM channels has significant homology to tetrameric voltage-gated K⫹ channel complexes: A short stretch between transmembrane helices 5 and 6 contains a hydrophobic pore helix followed by a pore loop; the loops of all four subunits of the tetramer are thought to contribute to a common selectivity filter (Figure 2B). The human TRPM6 gene comprises 39 exons and encodes multiple mRNA isoforms. For instance, alternative 5⬘ exons of the gene can be spliced to a common second exon, suggesting the TRPM6 promoter governs mRNA expression by means of alternative transcription start sites.11,76 Furthermore, several isoforms encode TRPM6 kinases because the alternatively spliced transcripts lack exons coding for the transmembrane segment of the channel.11,76 Functional consequences of the naturally occurring TRPM6 mutations include introduction of frame shifts, stop codons,

BRIEF REVIEW

or disruption of exon splice sites.66,77,78 Only five missense mutations have been identified so far (Figure 2A)66,77,78; their impact on TRPM6 function is discussed next. The pathophysiologic connection between hypomagnesemia and mutations in TRPM6 is mainly deduced from our current view on Mg2⫹ homeostasis.12,65,79 – 83 The total Mg2⫹ concentration in the majority of cells is between 10 and 20 mM. Intracellular Mg2⫹ is mostly bound to phosphonucleotides, phospholipids, and proteins. The free cellular Mg2⫹ concentration ([Mg2⫹]i) is estimated to be in the range of 0.5 to 1.0 mM, whereas the extracellular concentration of Mg2⫹ is between 0.9 and 1.0 mM, approximately 50% of which is bound to albumin and other molecules. Thus, mammalian cells lack a substantial transmembrane chemical gradient for ionized Mg2⫹. It is assumed, therefore, that the entry of Mg2⫹ into the cell is primarily driven by the electrical gradient across the plasma membrane.12,65,79 – 83 Physiologic studies identify two different transport systems for Mg2⫹ in the kidney and intestinal epithelia: An active transcellular and passive paracellular transport pathway.65,83,84 The saturable transcellular uptake consists of apical entry into epithelial cells most likely through Mg2⫹-selective ion channels and a putative basolateral extrusion step, which may couple Mg2⫹ export to Na⫹ influx (Figure 3). In the kidney, these two different transport pathways are arranged sequentially along the nephron.65,83,84 Most of the filtered Mg2⫹ is resorbed in the thick ascending limb of Henle’s loop by a passive paracellular route driven by positive transepithelial voltage. Tight junctions containing claudin-19 (CLDN19) are pivotal cellular components intimately involved in this paracellular transport.85 Only 5 to 10% of the filtered Mg2⫹ is resorbed in the distal convoluted tubule (DCT), which resorbs Mg2⫹ solely by the transcellular route. Unlike upstream nephron segments, the DCT has a negative transepithelial voltage and high epithelial resistance (Figure 3), and no Mg2⫹ resorption is detected in more disRenal TRPathies

739

BRIEF REVIEW

www.jasn.org

Lumen

Blood

TRPM7

Mg2+ Ca2+ TRPM7

Na+ Mg2+ TRPM6

Mg2+ Ca2+ TRPM7

[Mg2+]o 0.2 mM –10 mV

[Mg2+]i 0.5 mM –65 mV

[Mg2+]o 0.6 mM 0 mV

Figure 3. Schematic model depicting the role of TRPM6/7 channel complexes for transcellular Mg2⫹ transport in the DCT. Note that the DCT segment is characterized by a negative transepithelial voltage and lacks a substantial transmembrane chemical gradient for ionized Mg2⫹. For further explanation, see text.

tal nephron segments; therefore, the DCT determines the final urinary [Mg2⫹].64,65,83,86 As already alluded to, an electrical gradient is the primary driving force for Mg2⫹ along either the transcellular or the paracellular route. In line with this notion, numerous hormones, drugs, and certain pathophysiologic states that affect the membrane potential of the apical or basolateral plasma membrane of the DCT have a strong impact on net Mg2⫹ resorption.7,12,83,87,88 Against this background, it is reasonable to test whether TRPM6 is enriched in the DCT. In fact, by transcript copy number and in situ hybridization, pronounced expression of TRPM6 transcripts is present in the DCT.10,11,65 Subsequently, Schlingmann and colleagues10,65 postulated that TRPM6 represents the apical Mg2⫹-permeable ion channel responsible for Mg2⫹ uptake by DCT cells. More recently, immunohistochemistry of mouse kidney sections confirmed TRPM6 is present on the apical cell border along the DCT.89 On the contrary, mRNA encoding TRPM7 is detectable at a similar level in all nephron segments.11,65 The latter observation is not too surprising, because previous patch-clamp experiments and expression studies revealed that TRPM7 is ubiquitously present in all cell types tested so far.47–50,72,90 Thus, both ion 740

channels are coexpressed in the same nephron segment, but, in terms of function, TRPM6 is nonredundant. Conflicting findings are reported concerning functional characteristics of TRPM6 in heterologous cell systems. According to Voets et al.89 and Li et al.,90 recombinant TRPM6 expressed in HEK293 or CHO cells forms homo-oligomeric channels with biophysical characteristics similar to those of TRPM7. In particular, TRPM6 is somewhat more permeable to Mg2⫹ than to Ca2⫹ and, like TRPM7, sensitive to intracellular Mg2⫹ levels.89,90 In addition, TRPM6 channel activity is negatively regulated by receptor of activated protein kinase C 1, the repressor of estrogen receptor activity, and ATP through binding sites located in the TRPM6 kinase domain.91–93 An EGF receptor– dependent pathway regulates TRPM6 trafficking to the cell surface.94 Interestingly, EGF was identified as a magnesiotropic hormone, because a missense mutation in the EGF gene underlies isolated autosomal recessive renal hypomagnesemia (IRH).95 Groenestege et al.95 suggested that reduced TRPM6 function as a result of impaired trafficking to the plasma membrane is responsible for the IRH phenotype. It should be noted, however, that, unlike patients with HSH, patients with IRH showed normal serum concen-

Journal of the American Society of Nephrology

trations of Ca2⫹ and parathyroid hormone,95 illustrating that the pathophysiology of IRH differs from that of HSH, which is caused by mutations in TRPM6. In contrast to these studies, other studies11,77,96 provided biophysical, biochemical, and cellular evidence that TRPM6 displays features remarkably different from those of TRPM7. In particular, TRPM6 expressed in HEK293 cells, Xenopus oocytes, or DT40 lymphocytes96,97 does not efficiently form homomultimeric channel complexes in the plasma membrane and required TRPM7 for co-targeting to the cell surface. In the hetero-multimers formed, the TRPM6 kinase domain cross-phosphorylates TRPM7 but not vice versa.96 TRPM6/7 hetero-oligomeric channels are distinguishable in terms of singlechannel conductance, divalent cation permeability, and pH sensitivity from homo-multimers.90 TRPM7 is expressed ubiquitously, whereas TRPM6 expression is restricted to a few cell types, such as DCT cells.10,11,47,56 Furthermore, attempts to record native currents mediated solely by TRPM6 homo-oligomers have not been successful.90 In this vein, the specific and efficient heteromeric assembly of TRPM6 and TRPM7 may reflect the intrinsic mode of TRPM6 function as a subunit of TRPM6/7 complexes. Accordingly, both TRPM6 and TRPM7 contribute to transcellular Mg2⫹ transport in the DCT (Figure 3). Two scenarios emerge to explain the pathophysiologic consequences of lossof-function mutations in TRPM6. First, TRPM6/7 hetero-oligomers are the main mediators of MIC/MagNuM currents, and TRPM7 alone can only partially restore proper DCT function in terms of Mg2⫹ resorption. Second, a specific, still unidentified functional characteristic of heteromeric TRPM6/7 complexes is essential for DCT cell function and cannot be conferred by TRPM7 homo-multimers (Figure 3). The majority of HSH mutations in the TRPM6 gene most likely result in the complete lack of protein expression, thereby offering a straightforward explanation for the loss-of-function phenoJ Am Soc Nephrol 21: 736 –744, 2010

www.jasn.org

type. So far, only five point mutations in TRPM6 are known (Figure 2A). Four of them are located in the N-terminus of the channel protein, suggesting a critical role of this domain for channel function. In fact, the TRPM6S141L channel mutant is unable to associate with TRPM7.11 Consistently, the introduction of the homologous missense mutation into TRPM7 also affects ion channel assembly. More recently, a new missense mutation, P1017R, located in the putative pore-forming region of TRPM6 (Figure 2A) was characterized.78 This mutation affects neither expression of the TRPM6 protein nor its co-assembly with TRPM7 but impairs channel activity of TRPM6/7 complexes by dominant negative suppression. The effect exerted by TRPM6P1017R strongly supports the notion that both subunits contribute to a common channel pore in TRPM6/7 complexes (Figure 2B). Thus, one may conclude that considering the ion channel and kinase moieties of TRPM6, suppression of cation fluxes by TRPM6/7 complexes is sufficient for the development of HSH. Hence, these observations provide invaluable information about the poorly understood contribution of TRPM6 channel and kinase activities to the HSH phenotype. Overall, both of these studies suggest that TRPM7 is critically involved in transcellular Mg2⫹ transport and that genetic disruption of TRPM7 would have a much stronger impact on renal Mg2⫹ handling than inactivation of TRPM6. Thus, it would be interesting to investigate whether TRPM7⫹/⫺ mice98 display hypomagnesemia. Two genetic model organisms, mouse and zebrafish, have been used to gain deeper insight into the role of TRPM7 in vivo.98,99 Thus, a set of loss-of-function mutations in the TRPM7 gene cause complex changes in the development of zebrafish.99 Mutant larvae exhibit neural crest-derived melanophore deficiencies and temporary touch unresponsiveness and die 14 to 16 days after fertilization. Interestingly, Mg2⫹ supplementation partially rescues melanophore development, whereas Ca2⫹ partially restores J Am Soc Nephrol 21: 736 –744, 2010

both melanophore development and touch responsiveness, suggesting that mutations in TRPM7 affect the homeostasis of both cations in zebrafish. As in zebrafish, TRPM7 is essential for the embryonic development of mice: Embryos deficient in TRPM7 die at the preimplantation stage for unknown reasons.98 Conditional deletion of TRPM7 in the T cell lineage disrupted thymopoiesis, resulting in progressive depletion of thymic medullary cells. TRPM7-deficient thymocytes exhibit a dysregulated synthesis of various growth factors; however, unlike DT40 lymphocytes, thymocytes from TRPM7-deficient mice showed normal intracellular Mg2⫹ levels. Therefore, the authors conclude that TRPM7 is not essential for Mg2⫹ uptake by mouse thymocytes.98 It should be mentioned, however, that genetic interactions of TRPM7 with other proteins are still poorly understood. Thus, it is imaginable that a particular cellular phenotype of TRPM7-deficient cells may critically depend on the expression profile of other Mg2⫹-permeable ion channels and Mg2⫹ transporters such as MagT1 and TUSC3.100 In keeping with this notion, Mg2⫹ deficiency in DT40 cells with a disrupted TRPM7 gene is rescued by overexpression of the plasma membrane Mg2⫹ transporter SLC41A2 or constitutively active phosphoinositide 3-kinase.101,102 The recent description of a TRPM6deficient mouse model adds yet another level of complexity.103 The absence of TRPM6 gives rise to a different phenotype in mice than in humans. In contrast to previous expectations, the majority of TRPM6⫺/⫺ mice die at approximately day 12.5. Those that survive display massive neural tube closure defects.103 Interestingly, plasma Mg2⫹ levels are lower in TRPM6⫹/⫺ as compared with TRPM6⫹/⫹ mice. In contrast to HSH, high-Mg2⫹ diet does not prevent mortality in TRPM6-null embryos. These findings indicate a critical role of TRPM6 in mouse development. With regard to the human situation, it will be enlightening to reinvestigate HSH pedigrees for fetal wastage and developmental abnormalities.

BRIEF REVIEW

CONCLUSIONS

In the past few years, TRP channels have emerged as central players in human renal physiology, and mutations in these proteins are associated with frequently occurring kidney diseases as well as other pathologies.104 As more information on the in vivo role of TRP channels in suitable animal models and additional clinical data from patients carrying mutations in TRP channel genes become available, our knowledge of the role of TRP channels in renal pathophysiology will expand considerably. Further progress in this field will improve our mechanistic understanding of TRP channel function in general and may help in identification of novel therapeutic targets.

ACKNOWLEDGMENTS Work in the authors’ laboratories is supported by research project grants from the Deutsche Forschungsgemeinschaft (A.D., T.G., and V.C.), the Wissenschaftliches Herausgeberkollegium der Mu¨nchener Medizinischen Wochenschrift Stiftung (V.C.), and the Fritz-Thyssen-Stiftung (A.D. and T.G.).

DISCLOSURES None.

REFERENCES 1. Gudermann T: A new TRP to kidney disease. Nat Genet 37: 663– 664, 2005 2. Wu G, Somlo S: Molecular genetics and mechanism of autosomal dominant polycystic kidney disease. Mol Genet Metab 69: 1–15, 2000 3. Chang MY, Ong AC: Autosomal dominant polycystic kidney disease: Recent advances in pathogenesis and treatment. Nephron Physiol 108: 1–7, 2008 4. Wilson PD: Mouse models of polycystic kidney disease. Curr Top Dev Biol 84: 311– 350, 2008 5. Patel V, Chowdhury R, Igarashi P: Advances in the pathogenesis and treatment of polycystic kidney disease. Curr Opin Nephrol Hypertens 18: 99 –106, 2009 6. Harris PC, Torres VE: Polycystic kidney disease. Annu Rev Med 60: 321–337, 2009

Renal TRPathies

741

BRIEF REVIEW

www.jasn.org

7. Woudenberg-Vrenken TE, Bindels RJ, Hoenderop JG: The role of transient receptor potential channels in kidney disease. Nat Rev Nephrol 5: 441– 449, 2009 8. Winn MP, Conlon PJ, Lynn KL, Farrington MK, Creazzo T, Hawkins AF, Daskalakis N, Kwan SY, Ebersviller S, Burchette JL, Pericak-Vance MA, Howell DN, Vance JM, Rosenberg PB: A mutation in the TRPC6 cation channel causes familial focal segmental glomerulosclerosis. Science 308: 1801–1804, 2005 9. Reiser J, Polu KR, Moller CC, Kenlan P, Altintas MM, Wei C, Faul C, Herbert S, Villegas I, Avila-Casado C, McGee M, Sugimoto H, Brown D, Kalluri R, Mundel P, Smith PL, Clapham DE, Pollak MR: TRPC6 is a glomerular slit diaphragm-associated channel required for normal renal function. Nat Genet 37: 739 –744, 2005 10. Schlingmann KP, Weber S, Peters M, Niemann Nejsum L, Vitzthum H, Klingel K, Kratz M, Haddad E, Ristoff E, Dinour D, Syrrou M, Nielsen S, Sassen M, Waldegger S, Seyberth HW, Konrad M: Hypomagnesemia with secondary hypocalcemia is caused by mutations in TRPM6, a new member of the TRPM gene family. Nat Genet 31: 166 –170, 2002 11. Chubanov V, Waldegger S, Mederos y Schnitzler M, Vitzthum H, Sassen MC, Seyberth HW, Konrad M, Gudermann T: Disruption of TRPM6/TRPM7 complex formation by a mutation in the TRPM6 gene causes hypomagnesemia with secondary hypocalcemia. Proc Natl Acad Sci U S A 101: 2894 –2899, 2004 12. Konrad M, Schlingmann KP, Gudermann T: Insights into the molecular nature of magnesium homeostasis. Am J Physiol Renal Physiol 286: F599 –F605, 2004 13. Dietrich A, Mederos y Schnitzler M, Kalwa H, Storch U, Gudermann T: Functional characterization and physiological relevance of the TRPC3/6/7 subfamily of cation channels. Naunyn Schmiedebergs Arch Pharmacol 371: 257–265, 2005 14. Boulay G, Zhu X, Peyton M, Jiang M, Hurst R, Stefani E, Birnbaumer L: Cloning and expression of a novel mammalian homolog of Drosophila transient receptor potential (Trp) involved in calcium entry secondary to activation of receptors coupled by the Gq class of G protein. J Biol Chem 272: 29672–29680, 1997 15. Dietrich A, Chubanov V, Kalwa H, Rost BR, Gudermann T: Cation channels of the transient receptor potential superfamily: Their role in physiological and pathophysiological processes of smooth muscle cells. Pharmacol Ther 112: 744 –760, 2006 16. Hsu YJ, Hoenderop JG, Bindels RJ: TRP channels in kidney disease. Biochim Biophys Acta 1772: 928 –936, 2007 17. Goel M, Sinkins WG, Zuo CD, Estacion M, Schilling WP: Identification and localization

742

18.

19.

20.

21.

22.

23.

24.

25.

26.

of TRPC channels in the rat kidney. Am J Physiol Renal Physiol 290: F1241–F1252, 2006 Goel M, Zuo CD, Sinkins WG, Schilling WP: TRPC3 channels colocalize with Na⫹/ Ca2⫹ exchanger and Na⫹ pump in axial component of transverse-axial tubular system of rat ventricle. Am J Physiol Heart Circ Physiol 292: H874 –H883, 2007 Bandyopadhyay BC, Swaim WD, Liu X, Redman RS, Patterson RL, Ambudkar IS: Apical localization of a functional TRPC3/ TRPC6-Ca2⫹-signaling complex in polarized epithelial cells: Role in apical Ca2⫹ influx. J Biol Chem 280: 12908 –12916, 2005 Moller CC, Flesche J, Reiser J: Sensitizing the slit diaphragm with TRPC6 ion channels. J Am Soc Nephrol 20: 950 –953, 2009 Zhu B, Chen N, Wang ZH, Pan XX, Ren H, Zhang W, Wang WM: Identification and functional analysis of a novel TRPC6 mutation associated with late onset familial focal segmental glomerulosclerosis in Chinese patients. Mutat Res 664: 84 –90, 2009 Santin S, Ars E, Rossetti S, Salido E, Silva I, Garcia-Maset R, Gimenez I, Ruiz P, Mendizabal S, Nieto JL, Pena A, Camacho JA, Fraga G, Cobo MA, Bernis C, Ortiz A, de Pablos AL, Sanchez-Moreno A, Pintos G, Mirapeix E, Fernandez-Llama P, Ballarin J, Torra R, FSGS Study Group, Zamora I, Lo´pez-Hellin J, Madrid A, Ventura C, Vilalta R, Espinosa L, Garcı´a C, Melgosa M, Navarro M, Gime´nez A, Cots JV, Alexandra S, Caramelo C, Egido J, San Jose´ MD, de la Cerda F, Sala P, Raspall F, Vila A, Daza AM, Va´zquez M, Ecija JL, Espinosa M, Justa ML, Poveda R, Aparicio C, Rosell J, Muley R, Montenegro J, Gonza´lez D, Hidalgo E, de Frutos DB, Trillo E, Gracia S, de los Rı´os FJ: TRPC6 mutational analysis in a large cohort of patients with focal segmental glomerulosclerosis. Nephrol Dial Transplant 24: 3089 –3096, 2009 Heeringa SF, Moller CC, Du J, Yue L, Hinkes B, Chernin G, Vlangos CN, Hoyer PF, Reiser J, Hildebrandt F: A novel TRPC6 mutation that causes childhood FSGS. PLoS One 4: e7771, 2009 Lepage PK, Lussier MP, McDuff FO, Lavigne P, Boulay G: The self-association of two N-terminal interaction domains plays an important role in the tetramerization of TRPC4. Cell Calcium 45: 251–259, 2009 van Rossum DB, Oberdick D, Rbaibi Y, Bhardwaj G, Barrow RK, Snyder SH, Kiselyov K, Patterson RL: TRP_2: A lipid-binding/trafficking domain that mediates diacylglycerol-induced vesicle fusion. J Biol Chem 283: 34384 –34392, 2008 Lowik MM, Groenen PJ, Levtchenko EN, Monnens LA, van den Heuvel LP: Molecular genetic analysis of podocyte genes in focal segmental glomerulosclerosis: A review. Eur J Pediatr 168: 1291–1304, 2009

Journal of the American Society of Nephrology

27. Kriz W: TRPC6: A new podocyte gene involved in focal segmental glomerulosclerosis. Trends Mol Med 11: 527–530, 2005 28. Huber TB, Schermer B, Benzing T: Podocin organizes ion channel-lipid supercomplexes: Implications for mechanosensation at the slit diaphragm. Nephron Exp Nephrol 106: e27– e31, 2007 29. Spassova MA, Hewavitharana T, Xu W, Soboloff J, Gill DL: A common mechanism underlies stretch activation and receptor activation of TRPC6 channels. Proc Natl Acad Sci U S A 103: 16586 –16591, 2006 30. Mederos y Schnitzler M, Storch U, Meibers S, Nurwakagari P, Breit A, Essin K, Gollasch M, Gudermann T: Gq-coupled receptors as mechanosensors mediating myogenic vasoconstriction. EMBO J 27: 3092–3103, 2008 31. Gottlieb P, Folgering J, Maroto R, Raso A, Wood TG, Kurosky A, Bowman C, Bichet D, Patel A, Sachs F, Martinac B, Hamill OP, Honore E: Revisiting TRPC1 and TRPC6 mechanosensitivity. Pflugers Arch 455: 1097–1103, 2008 32. Inoue R, Jensen LJ, Jian Z, Shi J, Hai L, Lurie AI, Henriksen FH, Salomonsson M, Morita H, Kawarabayashi Y, Mori M, Mori Y, Ito Y: Synergistic activation of vascular TRPC6 channel by receptor and mechanical stimulation via phospholipase C/diacylglycerol and phospholipase A2/omega-hydroxylase/20-HETE pathways. Circ Res 104: 1399 –1409, 2009 33. Morita H, Honda A, Inoue R, Ito Y, Abe K, Nelson MT, Brayden JE: Membrane stretch-induced activation of a TRPM4-like nonselective cation channel in cerebral artery myocytes. J Pharmacol Sci 103: 417– 426, 2007 34. Zhu J, Sun N, Aoudjit L, Li H, Kawachi H, Lemay S, Takano T: Nephrin mediates actin reorganization via phosphoinositide 3-kinase in podocytes. Kidney Int 73: 556 –566, 2008 35. Verma R, Wharram B, Kovari I, Kunkel R, Nihalani D, Wary KK, Wiggins RC, Killen P, Holzman LB: Fyn binds to and phosphorylates the kidney slit diaphragm component Nephrin. J Biol Chem 278: 20716 –20723, 2003 36. Hisatsune C, Kuroda Y, Nakamura K, Inoue T, Nakamura T, Michikawa T, Mizutani A, Mikoshiba K: Regulation of TRPC6 channel activity by tyrosine phosphorylation. J Biol Chem 279: 18887–18894, 2004 37. Kawasaki BT, Liao Y, Birnbaumer L: Role of Src in C3 transient receptor potential channel function and evidence for a heterogeneous makeup of receptor- and store-operated Ca2⫹ entry channels. Proc Natl Acad Sci U S A 103: 335–340, 2006 38. Hinkes B, Wiggins RC, Gbadegesin R, Vlangos CN, Seelow D, Nurnberg G, Garg P, Verma R, Chaib H, Hoskins BE, Ashraf S, Becker C, Hennies HC, Goyal M, Wharram BL, Schachter AD, Mudumana S, Drum-

J Am Soc Nephrol 21: 736 –744, 2010

www.jasn.org

39.

40.

41.

42.

43.

44.

45.

46.

47.

mond I, Kerjaschki D, Waldherr R, Dietrich A, Ozaltin F, Bakkaloglu A, Cleper R, BaselVanagaite L, Pohl M, Griebel M, Tsygin AN, Soylu A, Muller D, Sorli CS, Bunney TD, Katan M, Liu J, Attanasio M, O’Toole JF, Hasselbacher K, Mucha B, Otto EA, Airik R, Kispert A, Kelley GG, Smrcka AV, Gudermann T, Holzman LB, Nurnberg P, Hildebrandt F: Positional cloning uncovers mutations in PLCE1 responsible for a nephrotic syndrome variant that may be reversible. Nat Genet 38: 1397–1405, 2006 Dietrich A, Mederos YS, Gollasch M, Gross V, Storch U, Dubrovska G, Obst M, Yildirim E, Salanova B, Kalwa H, Essin K, Pinkenburg O, Luft FC, Gudermann T, Birnbaumer L: Increased vascular smooth muscle contractility in TRPC6⫺/⫺ mice. Mol Cell Biol 25: 6980 – 6989, 2005 Sel S, Rost BR, Yildirim AO, Sel B, Kalwa H, Fehrenbach H, Renz H, Gudermann T, Dietrich A: Loss of classical transient receptor potential 6 channel reduces allergic airway response. Clin Exp Allergy 38: 1548 –1558, 2008 Kim EY, Alvarez-Baron CP, Dryer SE: Canonical transient receptor potential channel (TRPC)3 and TRPC6 associate with large-conductance Ca2⫹-activated K⫹ (BKCa) channels: Role in BKCa trafficking to the surface of cultured podocytes. Mol Pharmacol 75: 466 – 477, 2009 Dietrich A, Kalwa H, Fuchs B, Grimminger F, Weissmann N, Gudermann T: In vivo TRPC functions in the cardiopulmonary vasculature. Cell Calcium 42: 233–244, 2007 Schlondorff J, Del Camino D, Carrasquillo R, Lacey V, Pollak MR: TRPC6 mutations associated with focal segmental glomerulosclerosis cause constitutive activation of NFAT-dependent transcription. Am J Physiol Cell Physiol 296: C558 –C569, 2009 Moller CC, Wei C, Altintas MM, Li J, Greka A, Ohse T, Pippin JW, Rastaldi MP, Wawersik S, Schiavi S, Henger A, Kretzler M, Shankland SJ, Reiser J: Induction of TRPC6 channel in acquired forms of proteinuric kidney disease. J Am Soc Nephrol 18: 29 – 36, 2007 Mukerji N, Damodaran TV, Winn MP: TRPC6 and FSGS: The latest TRP channelopathy. Biochim Biophys Acta 1772: 859 – 868, 2007 Ding G, Reddy K, Kapasi AA, Franki N, Gibbons N, Kasinath BS, Singhal PC: Angiotensin II induces apoptosis in rat glomerular epithelial cells. Am J Physiol Renal Physiol 283: F173–F180, 2002 Nadler MJ, Hermosura MC, Inabe K, Perraud AL, Zhu Q, Stokes AJ, Kurosaki T, Kinet JP, Penner R, Scharenberg AM, Fleig A: LTRPC7 is a Mg.ATP-regulated divalent cation channel required for cell viability. Nature 411: 590 –595, 2001

J Am Soc Nephrol 21: 736 –744, 2010

48. Monteilh-Zoller MK, Hermosura MC, Nadler MJ, Scharenberg AM, Penner R, Fleig A: TRPM7 provides an ion channel mechanism for cellular entry of trace metal ions. J Gen Physiol 121: 49–60, 2003 49. Kozak JA, Kerschbaum HH, Cahalan MD: Distinct properties of CRAC and MIC channels in RBL cells. J Gen Physiol 120: 221– 235, 2002 50. Runnels LW, Yue L, Clapham DE: TRP-PLIK, a bifunctional protein with kinase and ion channel activities. Science 291: 1043–1047, 2001 51. Xing J, Yan X, Estevez A, Strange K: Highly Ca2⫹-selective TRPM channels regulate IP3-dependent oscillatory Ca2⫹ signaling in the C. elegans intestine. J Gen Physiol 131: 245–255, 2008 52. Teramoto T, Lambie EJ, Iwasaki K: Differential regulation of TRPM channels governs electrolyte homeostasis in the C. elegans intestine. Cell Metab 1: 343–354, 2005 53. Estevez AY, Roberts RK, Strange K: Identification of store-independent and storeoperated Ca2⫹ conductances in Caenorhabditis elegans intestinal epithelial cells. J Gen Physiol 122: 207–223, 2003 54. Kerschbaum HH, Kozak JA, Cahalan MD: Polyvalent cations as permeant probes of MIC and TRPM7 pores. Biophys J 84: 2293–2305, 2003 55. Kozak JA, Cahalan MD: MIC channels are inhibited by internal divalent cations but not ATP. Biophys J 84: 922–927, 2003 56. Walder RY, Landau D, Meyer P, Shalev H, Tsolia M, Borochowitz Z, Boettger MB, Beck GE, Englehardt RK, Carmi R, Sheffield VC: Mutation of TRPM6 causes familial hypomagnesemia with secondary hypocalcemia. Nat Genet 31: 171–174, 2002 57. Perraud AL, Knowles HM, Schmitz C: Novel aspects of signaling and ion-homeostasis regulation in immunocytes: The TRPM ion channels and their potential role in modulating the immune response. Mol Immunol 41: 657– 673, 2004 58. Touyz RM: Transient receptor potential melastatin 6 and 7 channels, magnesium transport, and vascular biology: Implications in hypertension. Am J Physiol Heart Circ Physiol 294: H1103–H1118, 2008 59. Penner R, Fleig A: The Mg2⫹ and Mg(2⫹)nucleotide-regulated channel-kinase TRPM7. Handb Exp Pharmacol 313–328, 2007 60. Nilius B, Owsianik G, Voets T, Peters JA: Transient receptor potential cation channels in disease. Physiol Rev 87: 165–217, 2007 61. Ramsey IS, Delling M, Clapham DE: An introduction to TRP channels. Annu Rev Physiol 68: 619 – 647, 2006 62. Venkatachalam K, Montell C: TRP channels. Annu Rev Biochem 76: 387– 417, 2007 63. Drennan D, Ryazanov AG: Alpha-kinases: Analysis of the family and comparison with

64.

65.

66.

67.

68.

69.

70. 71.

72.

73.

74.

75.

76.

77.

BRIEF REVIEW

conventional protein kinases. Prog Biophys Mol Biol 85: 1–32, 2004 Aarts MM, Tymianski M: TRPMs and neuronal cell death. Pflugers Arch 451: 243–249, 2005 Schlingmann KP, Gudermann T: A critical role of TRPM channel-kinase for human magnesium transport. J Physiol 566: 301– 308, 2005 Schlingmann KP, Sassen MC, Weber S, Pechmann U, Kusch K, Pelken L, Lotan D, Syrrou M, Prebble JJ, Cole DE, Metzger DL, Rahman S, Tajima T, Shu SG, Waldegger S, Seyberth HW, Konrad M: Novel TRPM6 mutations in 21 families with primary hypomagnesemia and secondary hypocalcemia. J Am Soc Nephrol 16: 3061– 3069, 2005 Suh SM, Tashjian AH Jr, Matsuo N, Parkinson DK, Fraser D: Pathogenesis of hypocalcemia in primary hypomagnesemia: Normal end-organ responsiveness to parathyroid hormone, impaired parathyroid gland function. J Clin Invest 52: 153– 160, 1973 Mennes P, Rosenbaum R, Martin K, Slatopolsky E: Hypomagnesemia and impaired parathyroid hormone secretion in chronic renal disease. Ann Intern Med 88: 206 –209, 1978 Kaske S, Krasteva G, Konig P, Kummer W, Hofmann T, Gudermann T, Chubanov V: TRPM5, a taste-signaling transient receptor potential ion-channel, is a ubiquitous signaling component in chemosensory cells. BMC Neurosci 8: 49, 2007 Damann N, Voets T, Nilius B: TRPs in our senses. Curr Biol 18: R880 –R889, 2008 Owsianik G, Talavera K, Voets T, Nilius B: Permeation and selectivity of TRP channels. Annu Rev Physiol 68: 685–717, 2006 Fleig A, Penner R: The TRPM ion channel subfamily: Molecular, biophysical and functional features. Trends Pharmacol Sci 25: 633– 639, 2004 Mederos y Schnitzler M, Waring J, Gudermann T, Chubanov V: Evolutionary determinants of divergent calcium selectivity of TRPM channels. FASEB J 22: 1540 –1551, 2008 Hofmann T, Chubanov V, Gudermann T, Montell C: TRPM5 is a voltage-modulated and Ca(2⫹)-activated monovalent selective cation channel. Curr Biol 13: 1153–1158, 2003 Ryazanov AG, Pavur KS, Dorovkov MV: Alpha-kinases: A new class of protein kinases with a novel catalytic domain. Curr Biol 9: R43–R45, 1999 Chubanov V, Mederos y Schnitzler M, Waring J, Plank A, Gudermann T: Emerging roles of TRPM6/TRPM7 channel kinase signal transduction complexes. Naunyn Schmiedebergs Arch Pharmacol 371: 334 –341, 2005 Jalkanen R, Pronicka E, Tyynismaa H,

Renal TRPathies

743

BRIEF REVIEW

78.

79. 80.

81.

82.

83.

84.

85.

86.

87.

744

www.jasn.org

Hanauer A, Walder R, Alitalo T: Genetic background of HSH in three Polish families and a patient with an X;9 translocation. Eur J Hum Genet 14: 55– 62, 2006 Chubanov V, Schlingmann KP, Waring J, Heinzinger J, Kaske S, Waldegger S, Mederos y Schnitzler M, Gudermann T: Hypomagnesemia with secondary hypocalcemia due to a missense mutation in the putative pore-forming region of TRPM6. J Biol Chem 282: 7656 –7667, 2007 Romani AM: Magnesium homeostasis in mammalian cells. Front Biosci 12: 308–331, 2007 Rubin H: Magnesium: The missing element in molecular views of cell proliferation control. Bioessays 27: 311–320, 2005 Wolf FI, Torsello A, Fasanella S, Cittadini A: Cell physiology of magnesium. Mol Aspects Med 24: 11–26, 2003 Wolf FI, Cittadini A: Chemistry and biochemistry of magnesium. Mol Aspects Med 24: 3–9, 2003 Dai LJ, Ritchie G, Kerstan D, Kang HS, Cole DE, Quamme GA: Magnesium transport in the renal distal convoluted tubule. Physiol Rev 81: 51– 84, 2001 Quamme GA: Recent developments in intestinal magnesium absorption. Curr Opin Gastroenterol 24: 230 –235, 2008 Konrad M, Schaller A, Seelow D, Pandey AV, Waldegger S, Lesslauer A, Vitzthum H, Suzuki Y, Luk JM, Becker C, Schlingmann KP, Schmid M, Rodriguez-Soriano J, Ariceta G, Cano F, Enriquez R, Juppner H, Bakkaloglu SA, Hediger MA, Gallati S, Neuhauss SC, Nurnberg P, Weber S: Mutations in the tightjunction gene claudin 19 (CLDN19) are associated with renal magnesium wasting, renal failure, and severe ocular involvement. Am J Hum Genet 79: 949–957, 2006 Quamme GA: Renal magnesium handling: New insights in understanding old problems. Kidney Int 52: 1180 –1195, 1997 Pham PC, Pham PM, Pham SV, Miller JM, Pham PT: Hypomagnesemia in patients with type 2 diabetes. Clin J Am Soc Nephrol 2: 366 –373, 2007

88. Schlingmann KP, Waldegger S, Konrad M, Chubanov V, Gudermann T: TRPM6 and TRPM7: Gatekeepers of human magnesium metabolism. Biochim Biophys Acta 1772: 813– 821, 2007 89. Voets T, Nilius B, Hoefs S, van der Kemp AW, Droogmans G, Bindels RJ, Hoenderop JG: TRPM6 forms the Mg2⫹ influx channel involved in intestinal and renal Mg2⫹ absorption. J Biol Chem 279: 19 –25, 2004 90. Li M, Jiang J, Yue L: Functional characterization of homo- and heteromeric channel kinases TRPM6 and TRPM7. J Gen Physiol 127: 525–537, 2006 91. Cao G, van der Wijst J, van der Kemp A, van Zeeland F, Bindels RJ, Hoenderop JG: Regulation of the epithelial MG2⫹ channel TRPM6 by estrogen and the associated repressor protein of estrogen receptor activity (REA). J Biol Chem 284: 14788 –14795, 2009 92. Cao G, Thebault S, van der Wijst J, van der Kemp A, Lasonder E, Bindels RJ, Hoenderop JG: RACK1 inhibits TRPM6 activity via phosphorylation of the fused alpha-kinase domain. Curr Biol 18: 168 –176, 2008 93. Thebault S, Cao G, Venselaar H, Xi Q, Bindels RJ, Hoenderop JG: Role of the alphakinase domain in transient receptor potential melastatin 6 channel and regulation by intracellular ATP. J Biol Chem 283: 19999 – 20007, 2008 94. Thebault S, Alexander RT, Tiel Groenestege WM, Hoenderop JG, Bindels RJ: EGF increases TRPM6 activity and surface expression. J Am Soc Nephrol 20: 78 – 85, 2009 95. Groenestege WM, Thebault S, van der Wijst J, van den Berg D, Janssen R, Tejpar S, van den Heuvel LP, van Cutsem E, Hoenderop JG, Knoers NV, Bindels RJ: Impaired basolateral sorting of pro-EGF causes isolated recessive renal hypomagnesemia. J Clin Invest 117: 2260 –2267, 2007 96. Schmitz C, Dorovkov MV, Zhao X, Davenport BJ, Ryazanov AG, Perraud AL: The channel kinases TRPM6 and TRPM7 are

Journal of the American Society of Nephrology

97.

98.

99.

100.

101.

102.

103.

104.

105.

functionally nonredundant. J Biol Chem 280: 37763–37771, 2005 Schmitz C, Perraud AL, Johnson CO, Inabe K, Smith MK, Penner R, Kurosaki T, Fleig A, Scharenberg AM: Regulation of vertebrate cellular Mg2⫹ homeostasis by TRPM7. Cell 114: 191–200, 2003 Jin J, Desai BN, Navarro B, Donovan A, Andrews NC, Clapham DE: Deletion of Trpm7 disrupts embryonic development and thymopoiesis without altering Mg2⫹ homeostasis. Science 322: 756 –760, 2008 Elizondo MR, Arduini BL, Paulsen J, MacDonald EL, Sabel JL, Henion PD, Cornell RA, Parichy DM: Defective skeletogenesis with kidney stone formation in dwarf zebrafish mutant for trpm7. Curr Biol 15: 667– 671, 2005 Zhou H, Clapham DE: Mammalian MagT1 and TUSC3 are required for cellular magnesium uptake and vertebrate embryonic development. Proc Natl Acad Sci U S A 106: 15750 –15755, 2009 Sahni J, Nelson B, Scharenberg AM: SLC41A2 encodes a plasma-membrane Mg2⫹ transporter. Biochem J 401: 505– 513, 2007 Sahni J, Scharenberg AM: TRPM7 ion channels are required for sustained phosphoinositide 3-kinase signaling in lymphocytes. Cell Metab 8: 84 –93, 2008 Walder RY, Yang B, Stokes JB, Kirby PA, Cao X, Shi P, Searby CC, Husted RF, Sheffield VC: Mice defective in Trpm6 show embryonic mortality and neural tube defects. Hum Mol Genet 18: 4367– 4385, 2009 Nilius B, Owsianik G: Transient receptor potential channelopathies. Pflugers Arch February 4, 2010 [epub ahead of print] Dietrich A, Mederos y Schnitzler M, Emmel J, Kalwa H, Hofmann T, Gudermann T: Nlinked protein glycosylation is a major determinant for basal TRPC3 and TRPC6 channel activity. J Biol Chem 278: 47842– 47852, 2003

J Am Soc Nephrol 21: 736 –744, 2010

Lihat lebih banyak...

Comentarios

Copyright © 2017 DATOSPDF Inc.