Ion channel transport (Homo sapiens)

From WikiPathways

Jump to: navigation, search
689832, 97, 105, 106, 1237, 81, 10293, 99, 1246, 16, 17, 23, 39...12827, 80, 91, 94, 10413, 31, 11613, 31, 1165, 37, 44, 54, 85...18, 25, 2912, 19, 21, 34, 35, 7740, 42, 10045, 655566, 1145, 37, 44, 54, 85...8, 3693, 124282, 1089, 33, 49, 50, 56...48, 7810, 2443, 47, 8458, 86, 89, 11014, 52, 11976, 1093, 11, 15, 12161, 9659, 122, 125261, 4, 69, 79, 11838641, 696474, 95887020, 22, 30, 53, 62...Golgi lumencytosollysosomal lumenmitochondrial intermembrane spacesperm flagellumendosome lumenlate endosome lumenmitochondrial matrixendoplasmic reticulum lumenplatelet dense tubular network lumensarcoplasmic reticulum lumenRPS27A(1-76) ASIC3 TRPV3 Na+Ub-SCNN channelsCu2+CLCN7 Ca2+PiC-terminus CLCAsurateTSC22D3 WWP1 CLCN7:OSTM1Cl-Ca2+WNKsPiH+SLC9B2ASIC1 SLC9B1 FXYD1 ATP2B3 CLCA1 (22-?) ATP6V1G1 ASPH ATP13A4 SLNRAF1 ATP9A Ca2+ATPATP6V0E1 UbH+ ATP1B2 Ca2+PiFXYD7 ATP1A3 SLC17A3(1-498)ATP6V1C2 MCOLN1 TCIRG1 ATPSLC9C2 cationATP2A1 ASIC2 ANO10 STOML3 CALM1 ANO1TRPV1 CLCNKA Li+ ATP2A3 TRPM4,5H+ATP4A STOML3 ATP2C2 ATP6V1G1 WNK1 H+ASIC trimers:H+PiP-type ATPases typeIVCLCNKB H+ATPUBB(153-228) PiATP6V1E1 PiADPCLCA4 (?-919) ATP6V0C ATP6V0A4 CLCA1 (?-914) TRDN CLCA3P SRIH+Na+ATP7A:PDZD11TRPV6 ATP10D ADPdivalent metalcationCl-ATP6V0C CLCA2 (?-943) ANO4 PiFXYD6 ATP7BATP6V0D2 SLC9B1/C2CLCA3P (?-262) Cl-Ca2+ATP10A PiCl-NALCN:UNC79:UNC80PS p-T357,S358-MLKL oligomer ATP1A2 ADPTRPV4 Na+ANOsATP6V1C1 ATP6V1A H+RYR3 ATP6V1F p-T287-CAMK2D amilorideSLC9C1TCIRG1 Ca2+PLN ATP6V1H ATP2A2 Mg2+ V-ATPase:ATP6AP1ATP8A2 UBC(533-608) Ca2+CUTC ATP6V1D H+Na+ATP6AP1 CLCA4 ANO2 H2OATPSCNN channelsATP6V1C2 CLCA2 TRPM5 CLCN1/2/KA/KBBSND Ca2+CLCN1 Ca2+ ANO3 Ca2+CALM1ASIC5 UBC(457-532) ATP7A ASIC trimersATP2A1-3ATPUBA52(1-76) ASIC3 SCNN1A H2OATP13A4, 5CLCN5 Cl-ATP6V0A2 ATP8B4 ATP6V1G3 ATP2A1-3PS H2OMCOLN3 ATP2A3 UNC80 H+ATP12A TRPM2 PDZD11 BESTsATP8B1 ATP8A1 STOM ATP6V1E2 SCNN1G ATP6V1B2 ATP2C1 Na+ATP2C1/2:Mg2+ATP10B ATP6V0A2 V-ATPaseH+ATP6V1B1 ATP8B3 urateCu2+ATP11A ASIC4CLCA4 (22-?) Ca2+RYR2 STOM ATP1A1 ASICtrimers:H+:STOML3,(STOM)p-S-RIPK1:p-S199,227-RIPK3:p-T357,S358-MLKL oligomerH2OOSTM1 TRPM3 TRPV2 UNC79 ASIC3 ATP6V0E1 ATP1B3 MCOLN2 Na+Mn2+ASIC1 Na+p-T287-CAMK2G SGK3 ATPCa2+Cl-ATP13A1ATPSGK2 Ca2+H2OH+PiH2OCAMK2heteromer:CALM:4xCa2+ATP1B1 ATP6V1E2 ATPATP6V0D1 CLCN4/5/6FKBP1B ATP11B ADPCLCA3P (21-?) BEST2 TTYH1-3ATP6V1C1 Cu1+CLCAsATP6V0A1 BEST1 Ca2+PiCl-UBC(153-228) TRPC6 SCNN1B BEST4 UBC(1-76) TRPM7 H+ATP2B1-4H+ATP6V1B2 ATP2A2 TPCN1 Na+WNK4 H+ATP6V0A4 BEST3 p-S-RIPK1:p-S199,227-RIPK3 oligomer CLCA1 PiATP6V0B ATP6V1F ATP9B H+ ASIC2 Na+RYRtetramer:FKBP1Btetramer:CASQpolymer:TRDN:junctinUb-SCNN1G CASQ1 polymer N-terminus CLCAsUBC(229-304) Na+CASQ2 polymer divalent metalcationamilorideTRPM4 K+H2Op-T287-CAMK2B cationAPLsSLC17A3Na+ANO1 HCO3-SGK1 ATP8B2 FXYD2 UBC(305-380) UBC(609-684) TRPM1 ATP6V0B CUTC tetramerHCO3-ANO5 TPCN1/2TRPC5 ATP6V0D2 WNK3 ATP2B4 TPCN2 Ub-SCNN1A Ca2+Na+RAF1:SGK:TSC22D3:WPPSCNN1B TTYH2 K+NALCN Na+ATP11C Mn2+ASIC2 Na+ATP2B2 ASIC5 ATPFXYD3 H+UBC(381-456) CLCN2 ATP4B ATP1A:ATP1B:FXYDCl-TRPC7 Li+ Cu2+CLCN4 Na+/Li+p-S16-PLN TRPA1 ADPCUTC:4xCu+ADPADPK+NAADPTRPM6 ANO7 TRPC4AP ATP4A/12A:ATP4BCLCA2 (32-?) ANO8 TRPV5 ATP6V0E2 TTYH2/3CLCN6 WNK2 Na+ATP6V1G2 ATP1A4 TRPM8 H+NSAIDASIC1 H+ATP6V1G3 TRPC1 ATP2A1 ANO6 STOML3, (STOM)ASIC5 Na+/Li+Cu1+ CLIC2ADPATPATP6V1G2 ATP6V1H TRPC3(1-848) ATP6V0D1 ADPUBB(1-76) PE SCNN1D H+ATP6V1B1 ATP6V0E2 p-T286-CAMK2A Na+ Na+ TRPsCa2+UBC(77-152) ATP13A2CUTC TTYH3 p-S16-PLN pentamerSCNN1D ANO9 UBB(77-152) APLsATP6V1A TRPC4 H+ATP6V0A1 H2OFXYD4 ATP6AP1ATP6V1E1 PLN pentamerATP2B1 PE CLCN3Na+ATP6V1D ATP13A5 RYR1 NEDD4L 9246, 92


Description

Ion channels mediate the flow of ions across the plasma membrane of cells. They are integral membrane proteins, typically a multimer of proteins, which, when arranged in the membrane, create a pore for the flow of ions. There are different types of ion channels. P-type ATPases undergo conformational changes to translocate ions. Ligand-gated ion channels operate like a gate, opened or closed by a chemical signal. Voltage-gated ion channels are activated by changes in electrical potential difference at the membrane (Purves, 2001; Kuhlbrandt, 2004). View original pathway at Reactome.

Comments

Reactome-Converter 
Pathway is converted from Reactome ID: 983712
Reactome-version 
Reactome version: 73
Reactome Author 
Reactome Author: Jassal, Bijay

Try the New WikiPathways

View approved pathways at the new wikipathways.org.

Quality Tags

Ontology Terms

 

Bibliography

View all...
  1. Bokkala S, el-Daher SS, Kakkar VV, Wuytack F, Authi KS.; ''Localization and identification of Ca2+ATPases in highly purified human platelet plasma and intracellular membranes. Evidence that the monoclonal antibody PL/IM 430 recognizes the SERCA 3 Ca2+ATPase in human platelets.''; PubMed Europe PMC Scholia
  2. Supek F, Supekova L, Mandiyan S, Pan YC, Nelson H, Nelson N.; ''A novel accessory subunit for vacuolar H(+)-ATPase from chromaffin granules.''; PubMed Europe PMC Scholia
  3. Staub O, Abriel H, Plant P, Ishikawa T, Kanelis V, Saleki R, Horisberger JD, Schild L, Rotin D.; ''Regulation of the epithelial Na+ channel by Nedd4 and ubiquitination.''; PubMed Europe PMC Scholia
  4. Gorski PA, Glaves JP, Vangheluwe P, Young HS.; ''Sarco(endo)plasmic reticulum calcium ATPase (SERCA) inhibition by sarcolipin is encoded in its luminal tail.''; PubMed Europe PMC Scholia
  5. Schaefer L, Sakai H, Mattei M, Lazdunski M, Lingueglia E.; ''Molecular cloning, functional expression and chromosomal localization of an amiloride-sensitive Na(+) channel from human small intestine.''; PubMed Europe PMC Scholia
  6. Nakashima Y, Nishimura S, Maeda A, Barsoumian EL, Hakamata Y, Nakai J, Allen PD, Imoto K, Kita T.; ''Molecular cloning and characterization of a human brain ryanodine receptor.''; PubMed Europe PMC Scholia
  7. Kawasaki M, Fukuma T, Yamauchi K, Sakamoto H, Marumo F, Sasaki S.; ''Identification of an acid-activated Cl(-) channel from human skeletal muscles.''; PubMed Europe PMC Scholia
  8. Lai HJ, Lin CH, Wu RM.; ''Early-onset autosomal-recessive parkinsonian-pyramidal syndrome.''; PubMed Europe PMC Scholia
  9. Koch MC, Steinmeyer K, Lorenz C, Ricker K, Wolf F, Otto M, Zoll B, Lehmann-Horn F, Grzeschik KH, Jentsch TJ.; ''The skeletal muscle chloride channel in dominant and recessive human myotonia.''; PubMed Europe PMC Scholia
  10. Verma AK, Filoteo AG, Stanford DR, Wieben ED, Penniston JT, Strehler EE, Fischer R, Heim R, Vogel G, Mathews S.; ''Complete primary structure of a human plasma membrane Ca2+ pump.''; PubMed Europe PMC Scholia
  11. Farr TJ, Coddington-Lawson SJ, Snyder PM, McDonald FJ.; ''Human Nedd4 interacts with the human epithelial Na+ channel: WW3 but not WW1 binds to Na+-channel subunits.''; PubMed Europe PMC Scholia
  12. Yurtsever Z, Sala-Rabanal M, Randolph DT, Scheaffer SM, Roswit WT, Alevy YG, Patel AC, Heier RF, Romero AG, Nichols CG, Holtzman MJ, Brett TJ.; ''Self-cleavage of human CLCA1 protein by a novel internal metalloprotease domain controls calcium-activated chloride channel activation.''; PubMed Europe PMC Scholia
  13. Scudieri P, Sondo E, Ferrera L, Galietta LJ.; ''The anoctamin family: TMEM16A and TMEM16B as calcium-activated chloride channels.''; PubMed Europe PMC Scholia
  14. Stobrawa SM, Breiderhoff T, Takamori S, Engel D, Schweizer M, Zdebik AA, Bösl MR, Ruether K, Jahn H, Draguhn A, Jahn R, Jentsch TJ.; ''Disruption of ClC-3, a chloride channel expressed on synaptic vesicles, leads to a loss of the hippocampus.''; PubMed Europe PMC Scholia
  15. Soundararajan R, Melters D, Shih IC, Wang J, Pearce D.; ''Epithelial sodium channel regulated by differential composition of a signaling complex.''; PubMed Europe PMC Scholia
  16. Kim E, Youn B, Kemper L, Campbell C, Milting H, Varsanyi M, Kang C.; ''Characterization of human cardiac calsequestrin and its deleterious mutants.''; PubMed Europe PMC Scholia
  17. Ono M, Yano M, Hino A, Suetomi T, Xu X, Susa T, Uchinoumi H, Tateishi H, Oda T, Okuda S, Doi M, Kobayashi S, Yamamoto T, Koseki N, Kyushiki H, Ikemoto N, Matsuzaki M.; ''Dissociation of calmodulin from cardiac ryanodine receptor causes aberrant Ca(2+) release in heart failure.''; PubMed Europe PMC Scholia
  18. Grishin AV, Sverdlov VE, Kostina MB, Modyanov NN.; ''Cloning and characterization of the entire cDNA encoded by ATP1AL1--a member of the human Na,K/H,K-ATPase gene family.''; PubMed Europe PMC Scholia
  19. Gibson A, Lewis AP, Affleck K, Aitken AJ, Meldrum E, Thompson N.; ''hCLCA1 and mCLCA3 are secreted non-integral membrane proteins and therefore are not ion channels.''; PubMed Europe PMC Scholia
  20. Kawakami K, Ohta T, Nojima H, Nagano K.; ''Primary structure of the alpha-subunit of human Na,K-ATPase deduced from cDNA sequence.''; PubMed Europe PMC Scholia
  21. Hamann M, Gibson A, Davies N, Jowett A, Walhin JP, Partington L, Affleck K, Trezise D, Main M.; ''Human ClCa1 modulates anionic conduction of calcium-dependent chloride currents.''; PubMed Europe PMC Scholia
  22. Malik N, Canfield VA, Beckers MC, Gros P, Levenson R.; ''Identification of the mammalian Na,K-ATPase 3 subunit.''; PubMed Europe PMC Scholia
  23. Dulhunty AF, Pouliquin P, Coggan M, Gage PW, Board PG.; ''A recently identified member of the glutathione transferase structural family modifies cardiac RyR2 substate activity, coupled gating and activation by Ca2+ and ATP.''; PubMed Europe PMC Scholia
  24. Schatzmann HJ.; ''ATP-dependent Ca++-extrusion from human red cells.''; PubMed Europe PMC Scholia
  25. Maeda M, Oshiman K, Tamura S, Futai M.; ''Human gastric (H+ + K+)-ATPase gene. Similarity to (Na+ + K+)-ATPase genes in exon/intron organization but difference in control region.''; PubMed Europe PMC Scholia
  26. Ye G, Chen C, Han D, Xiong X, Kong Y, Wan B, Yu L.; ''Cloning of a novel human NHEDC1 (Na+/H+ exchanger like domain containing 1) gene expressed specifically in testis.''; PubMed Europe PMC Scholia
  27. Leisle L, Ludwig CF, Wagner FA, Jentsch TJ, Stauber T.; ''ClC-7 is a slowly voltage-gated 2Cl(-)/1H(+)-exchanger and requires Ostm1 for transport activity.''; PubMed Europe PMC Scholia
  28. Cohen Y, Megyeri M, Chen OC, Condomitti G, Riezman I, Loizides-Mangold U, Abdul-Sada A, Rimon N, Riezman H, Platt FM, Futerman AH, Schuldiner M.; ''The yeast p5 type ATPase, spf1, regulates manganese transport into the endoplasmic reticulum.''; PubMed Europe PMC Scholia
  29. Ma JY, Song YH, Sjöstrand SE, Rask L, Mårdh S.; ''cDNA cloning of the beta-subunit of the human gastric H,K-ATPase.''; PubMed Europe PMC Scholia
  30. Lane LK, Shull MM, Whitmer KR, Lingrel JB.; ''Characterization of two genes for the human Na,K-ATPase beta subunit.''; PubMed Europe PMC Scholia
  31. Tian Y, Schreiber R, Kunzelmann K.; ''Anoctamins are a family of Ca2+-activated Cl- channels.''; PubMed Europe PMC Scholia
  32. Chelly J, Tümer Z, Tønnesen T, Petterson A, Ishikawa-Brush Y, Tommerup N, Horn N, Monaco AP.; ''Isolation of a candidate gene for Menkes disease that encodes a potential heavy metal binding protein.''; PubMed Europe PMC Scholia
  33. Fischer M, Janssen AG, Fahlke C.; ''Barttin activates ClC-K channel function by modulating gating.''; PubMed Europe PMC Scholia
  34. Toda M, Tulic MK, Levitt RC, Hamid Q.; ''A calcium-activated chloride channel (HCLCA1) is strongly related to IL-9 expression and mucus production in bronchial epithelium of patients with asthma.''; PubMed Europe PMC Scholia
  35. Hoshino M, Morita S, Iwashita H, Sagiya Y, Nagi T, Nakanishi A, Ashida Y, Nishimura O, Fujisawa Y, Fujino M.; ''Increased expression of the human Ca2+-activated Cl- channel 1 (CaCC1) gene in the asthmatic airway.''; PubMed Europe PMC Scholia
  36. Ramonet D, Podhajska A, Stafa K, Sonnay S, Trancikova A, Tsika E, Pletnikova O, Troncoso JC, Glauser L, Moore DJ.; ''PARK9-associated ATP13A2 localizes to intracellular acidic vesicles and regulates cation homeostasis and neuronal integrity.''; PubMed Europe PMC Scholia
  37. García-Añoveros J, Derfler B, Neville-Golden J, Hyman BT, Corey DP.; ''BNaC1 and BNaC2 constitute a new family of human neuronal sodium channels related to degenerins and epithelial sodium channels.''; PubMed Europe PMC Scholia
  38. Melis D, Havelaar AC, Verbeek E, Smit GP, Benedetti A, Mancini GM, Verheijen F.; ''NPT4, a new microsomal phosphate transporter: mutation analysis in glycogen storage disease type Ic.''; PubMed Europe PMC Scholia
  39. Tunwell RE, Wickenden C, Bertrand BM, Shevchenko VI, Walsh MB, Allen PD, Lai FA.; ''The human cardiac muscle ryanodine receptor-calcium release channel: identification, primary structure and topological analysis.''; PubMed Europe PMC Scholia
  40. Thomas GR, Forbes JR, Roberts EA, Walshe JM, Cox DW.; ''The Wilson disease gene: spectrum of mutations and their consequences.''; PubMed Europe PMC Scholia
  41. Smith JS, Rousseau E, Meissner G.; ''Calmodulin modulation of single sarcoplasmic reticulum Ca2+-release channels from cardiac and skeletal muscle.''; PubMed Europe PMC Scholia
  42. Yang XL, Miura N, Kawarada Y, Terada K, Petrukhin K, Gilliam T, Sugiyama T.; ''Two forms of Wilson disease protein produced by alternative splicing are localized in distinct cellular compartments.''; PubMed Europe PMC Scholia
  43. Sudbrak R, Brown J, Dobson-Stone C, Carter S, Ramser J, White J, Healy E, Dissanayake M, Larrègue M, Perrussel M, Lehrach H, Munro CS, Strachan T, Burge S, Hovnanian A, Monaco AP.; ''Hailey-Hailey disease is caused by mutations in ATP2C1 encoding a novel Ca(2+) pump.''; PubMed Europe PMC Scholia
  44. Voilley N, de Weille J, Mamet J, Lazdunski M.; ''Nonsteroid anti-inflammatory drugs inhibit both the activity and the inflammation-induced expression of acid-sensing ion channels in nociceptors.''; PubMed Europe PMC Scholia
  45. Lu B, Zhang Q, Wang H, Wang Y, Nakayama M, Ren D.; ''Extracellular calcium controls background current and neuronal excitability via an UNC79-UNC80-NALCN cation channel complex.''; PubMed Europe PMC Scholia
  46. Petrie EJ, Sandow JJ, Jacobsen AV, Smith BJ, Griffin MDW, Lucet IS, Dai W, Young SN, Tanzer MC, Wardak A, Liang LY, Cowan AD, Hildebrand JM, Kersten WJA, Lessene G, Silke J, Czabotar PE, Webb AI, Murphy JM.; ''Conformational switching of the pseudokinase domain promotes human MLKL tetramerization and cell death by necroptosis.''; PubMed Europe PMC Scholia
  47. Vanoevelen J, Dode L, Van Baelen K, Fairclough RJ, Missiaen L, Raeymaekers L, Wuytack F.; ''The secretory pathway Ca2+/Mn2+-ATPase 2 is a Golgi-localized pump with high affinity for Ca2+ ions.''; PubMed Europe PMC Scholia
  48. Kwasnicka-Crawford DA, Carson AR, Roberts W, Summers AM, Rehnström K, Järvelä I, Scherer SW.; ''Characterization of a novel cation transporter ATPase gene (ATP13A4) interrupted by 3q25-q29 inversion in an individual with language delay.''; PubMed Europe PMC Scholia
  49. Cid LP, Montrose-Rafizadeh C, Smith DI, Guggino WB, Cutting GR.; ''Cloning of a putative human voltage-gated chloride channel (CIC-2) cDNA widely expressed in human tissues.''; PubMed Europe PMC Scholia
  50. Nozu K, Inagaki T, Fu XJ, Nozu Y, Kaito H, Kanda K, Sekine T, Igarashi T, Nakanishi K, Yoshikawa N, Iijima K, Matsuo M.; ''Molecular analysis of digenic inheritance in Bartter syndrome with sensorineural deafness.''; PubMed Europe PMC Scholia
  51. Franceschini S, Ilari A, Verzili D, Zamparelli C, Antaramian A, Rueda A, Valdivia HH, Chiancone E, Colotti G.; ''Molecular basis for the impaired function of the natural F112L sorcin mutant: X-ray crystal structure, calcium affinity, and interaction with annexin VII and the ryanodine receptor.''; PubMed Europe PMC Scholia
  52. Borsani G, Rugarli EI, Taglialatela M, Wong C, Ballabio A.; ''Characterization of a human and murine gene (CLCN3) sharing similarities to voltage-gated chloride channels and to a yeast integral membrane protein.''; PubMed Europe PMC Scholia
  53. Vanmolkot KR, Kors EE, Hottenga JJ, Terwindt GM, Haan J, Hoefnagels WA, Black DF, Sandkuijl LA, Frants RR, Ferrari MD, van den Maagdenberg AM.; ''Novel mutations in the Na+, K+-ATPase pump gene ATP1A2 associated with familial hemiplegic migraine and benign familial infantile convulsions.''; PubMed Europe PMC Scholia
  54. Donier E, Rugiero F, Jacob C, Wood JN.; ''Regulation of ASIC activity by ASIC4--new insights into ASIC channel function revealed by a yeast two-hybrid assay.''; PubMed Europe PMC Scholia
  55. Qu Z, Hartzell HC.; ''Bestrophin Cl- channels are highly permeable to HCO3-.''; PubMed Europe PMC Scholia
  56. Takeuchi Y, Uchida S, Marumo F, Sasaki S.; ''Cloning, tissue distribution, and intrarenal localization of ClC chloride channels in human kidney.''; PubMed Europe PMC Scholia
  57. Birkenhäger R, Otto E, Schürmann MJ, Vollmer M, Ruf EM, Maier-Lutz I, Beekmann F, Fekete A, Omran H, Feldmann D, Milford DV, Jeck N, Konrad M, Landau D, Knoers NV, Antignac C, Sudbrak R, Kispert A, Hildebrandt F.; ''Mutation of BSND causes Bartter syndrome with sensorineural deafness and kidney failure.''; PubMed Europe PMC Scholia
  58. Sun H, Tsunenari T, Yau KW, Nathans J.; ''The vitelliform macular dystrophy protein defines a new family of chloride channels.''; PubMed Europe PMC Scholia
  59. Li J, Ji C, Chen J, Yang Z, Wang Y, Fei X, Zheng M, Gu X, Wen G, Xie Y, Mao Y.; ''Identification and characterization of a novel Cut family cDNA that encodes human copper transporter protein CutC.''; PubMed Europe PMC Scholia
  60. Waldegger S, Jentsch TJ.; ''Functional and structural analysis of ClC-K chloride channels involved in renal disease.''; PubMed Europe PMC Scholia
  61. Brailoiu E, Churamani D, Cai X, Schrlau MG, Brailoiu GC, Gao X, Hooper R, Boulware MJ, Dun NJ, Marchant JS, Patel S.; ''Essential requirement for two-pore channel 1 in NAADP-mediated calcium signaling.''; PubMed Europe PMC Scholia
  62. Ovchinnikov YuA, Monastyrskaya GS, Broude NE, Ushkaryov YuA, Melkov AM, Smirnov YuV, Malyshev IV, Allikmets RL, Kostina MB, Dulubova IE.; ''Family of human Na+, K+-ATPase genes. Structure of the gene for the catalytic subunit (alpha III-form) and its relationship with structural features of the protein.''; PubMed Europe PMC Scholia
  63. Shull MM, Pugh DG, Lingrel JB.; ''Characterization of the human Na,K-ATPase alpha 2 gene and identification of intragenic restriction fragment length polymorphisms.''; PubMed Europe PMC Scholia
  64. Paulusma CC, Oude Elferink RP.; ''The type 4 subfamily of P-type ATPases, putative aminophospholipid translocases with a role in human disease.''; PubMed Europe PMC Scholia
  65. Lu B, Su Y, Das S, Liu J, Xia J, Ren D.; ''The neuronal channel NALCN contributes resting sodium permeability and is required for normal respiratory rhythm.''; PubMed Europe PMC Scholia
  66. Prawitt D, Monteilh-Zoller MK, Brixel L, Spangenberg C, Zabel B, Fleig A, Penner R.; ''TRPM5 is a transient Ca2+-activated cation channel responding to rapid changes in [Ca2+]i.''; PubMed Europe PMC Scholia
  67. Scholl U, Hebeisen S, Janssen AG, Müller-Newen G, Alekov A, Fahlke C.; ''Barttin modulates trafficking and function of ClC-K channels.''; PubMed Europe PMC Scholia
  68. Kühlbrandt W.; ''Biology, structure and mechanism of P-type ATPases.''; PubMed Europe PMC Scholia
  69. Spamer C, Heilmann C, Gerok W.; ''Ca2+-activated ATPase in microsomes from human liver.''; PubMed Europe PMC Scholia
  70. Lapatsina L, Jira JA, Smith ES, Poole K, Kozlenkov A, Bilbao D, Lewin GR, Heppenstall PA.; ''Regulation of ASIC channels by a stomatin/STOML3 complex located in a mobile vesicle pool in sensory neurons.''; PubMed Europe PMC Scholia
  71. Marx SO, Reiken S, Hisamatsu Y, Jayaraman T, Burkhoff D, Rosemblit N, Marks AR.; ''PKA phosphorylation dissociates FKBP12.6 from the calcium release channel (ryanodine receptor): defective regulation in failing hearts.''; PubMed Europe PMC Scholia
  72. Schlingmann KP, Konrad M, Jeck N, Waldegger P, Reinalter SC, Holder M, Seyberth HW, Waldegger S.; ''Salt wasting and deafness resulting from mutations in two chloride channels.''; PubMed Europe PMC Scholia
  73. Board PG, Coggan M, Watson S, Gage PW, Dulhunty AF.; ''CLIC-2 modulates cardiac ryanodine receptor Ca2+ release channels.''; PubMed Europe PMC Scholia
  74. Xiang M, Feng M, Muend S, Rao R.; ''A human Na+/H+ antiporter sharing evolutionary origins with bacterial NhaA may be a candidate gene for essential hypertension.''; PubMed Europe PMC Scholia
  75. Steinmeyer K, Lorenz C, Pusch M, Koch MC, Jentsch TJ.; ''Multimeric structure of ClC-1 chloride channel revealed by mutations in dominant myotonia congenita (Thomsen).''; PubMed Europe PMC Scholia
  76. Wu LJ, Sweet TB, Clapham DE.; ''International Union of Basic and Clinical Pharmacology. LXXVI. Current progress in the mammalian TRP ion channel family.''; PubMed Europe PMC Scholia
  77. Kamada F, Suzuki Y, Shao C, Tamari M, Hasegawa K, Hirota T, Shimizu M, Takahashi N, Mao XQ, Doi S, Fujiwara H, Miyatake A, Fujita K, Chiba Y, Aoki Y, Kure S, Tamura G, Shirakawa T, Matsubara Y.; ''Association of the hCLCA1 gene with childhood and adult asthma.''; PubMed Europe PMC Scholia
  78. Schmidt K, Wolfe DM, Stiller B, Pearce DA.; ''Cd2+, Mn2+, Ni2+ and Se2+ toxicity to Saccharomyces cerevisiae lacking YPK9p the orthologue of human ATP13A2.''; PubMed Europe PMC Scholia
  79. Matsumoto T, Hisamatsu Y, Ohkusa T, Inoue N, Sato T, Suzuki S, Ikeda Y, Matsuzaki M.; ''Sorcin interacts with sarcoplasmic reticulum Ca(2+)-ATPase and modulates excitation-contraction coupling in the heart.''; PubMed Europe PMC Scholia
  80. Pangrazio A, Poliani PL, Megarbane A, Lefranc G, Lanino E, Di Rocco M, Rucci F, Lucchini F, Ravanini M, Facchetti F, Abinun M, Vezzoni P, Villa A, Frattini A.; ''Mutations in OSTM1 (grey lethal) define a particularly severe form of autosomal recessive osteopetrosis with neural involvement.''; PubMed Europe PMC Scholia
  81. Neagoe I, Stauber T, Fidzinski P, Bergsdorf EY, Jentsch TJ.; ''The late endosomal ClC-6 mediates proton/chloride countertransport in heterologous plasma membrane expression.''; PubMed Europe PMC Scholia
  82. Zorzato F, Fujii J, Otsu K, Phillips M, Green NM, Lai FA, Meissner G, MacLennan DH.; ''Molecular cloning of cDNA encoding human and rabbit forms of the Ca2+ release channel (ryanodine receptor) of skeletal muscle sarcoplasmic reticulum.''; PubMed Europe PMC Scholia
  83. Fujii J, Willard HF, MacLennan DH.; ''Characterization and localization to human chromosome 1 of human fast-twitch skeletal muscle calsequestrin gene.''; PubMed Europe PMC Scholia
  84. Hu Z, Bonifas JM, Beech J, Bench G, Shigihara T, Ogawa H, Ikeda S, Mauro T, Epstein EH.; ''Mutations in ATP2C1, encoding a calcium pump, cause Hailey-Hailey disease.''; PubMed Europe PMC Scholia
  85. Price MP, Snyder PM, Welsh MJ.; ''Cloning and expression of a novel human brain Na+ channel.''; PubMed Europe PMC Scholia
  86. Petrukhin K, Koisti MJ, Bakall B, Li W, Xie G, Marknell T, Sandgren O, Forsman K, Holmgren G, Andreasson S, Vujic M, Bergen AA, McGarty-Dugan V, Figueroa D, Austin CP, Metzker ML, Caskey CT, Wadelius C.; ''Identification of the gene responsible for Best macular dystrophy.''; PubMed Europe PMC Scholia
  87. Kieferle S, Fong P, Bens M, Vandewalle A, Jentsch TJ.; ''Two highly homologous members of the ClC chloride channel family in both rat and human kidney.''; PubMed Europe PMC Scholia
  88. Jutabha P, Anzai N, Kitamura K, Taniguchi A, Kaneko S, Yan K, Yamada H, Shimada H, Kimura T, Katada T, Fukutomi T, Tomita K, Urano W, Yamanaka H, Seki G, Fujita T, Moriyama Y, Yamada A, Uchida S, Wempe MF, Endou H, Sakurai H.; ''Human sodium phosphate transporter 4 (hNPT4/SLC17A3) as a common renal secretory pathway for drugs and urate.''; PubMed Europe PMC Scholia
  89. Tsunenari T, Sun H, Williams J, Cahill H, Smallwood P, Yau KW, Nathans J.; ''Structure-function analysis of the bestrophin family of anion channels.''; PubMed Europe PMC Scholia
  90. Estévez R, Boettger T, Stein V, Birkenhäger R, Otto E, Hildebrandt F, Jentsch TJ.; ''Barttin is a Cl- channel beta-subunit crucial for renal Cl- reabsorption and inner ear K+ secretion.''; PubMed Europe PMC Scholia
  91. Graves AR, Curran PK, Smith CL, Mindell JA.; ''The Cl-/H+ antiporter ClC-7 is the primary chloride permeation pathway in lysosomes.''; PubMed Europe PMC Scholia
  92. Cai Z, Jitkaew S, Zhao J, Chiang HC, Choksi S, Liu J, Ward Y, Wu LG, Liu ZG.; ''Plasma membrane translocation of trimerized MLKL protein is required for TNF-induced necroptosis.''; PubMed Europe PMC Scholia
  93. Suzuki M.; ''The Drosophila tweety family: molecular candidates for large-conductance Ca2+-activated Cl- channels.''; PubMed Europe PMC Scholia
  94. Pangrazio A, Pusch M, Caldana E, Frattini A, Lanino E, Tamhankar PM, Phadke S, Lopez AG, Orchard P, Mihci E, Abinun M, Wright M, Vettenranta K, Bariae I, Melis D, Tezcan I, Baumann C, Locatelli F, Zecca M, Horwitz E, Mansour LS, Van Roij M, Vezzoni P, Villa A, Sobacchi C.; ''Molecular and clinical heterogeneity in CLCN7-dependent osteopetrosis: report of 20 novel mutations.''; PubMed Europe PMC Scholia
  95. Taglicht D, Padan E, Schuldiner S.; ''Proton-sodium stoichiometry of NhaA, an electrogenic antiporter from Escherichia coli.''; PubMed Europe PMC Scholia
  96. Calcraft PJ, Ruas M, Pan Z, Cheng X, Arredouani A, Hao X, Tang J, Rietdorf K, Teboul L, Chuang KT, Lin P, Xiao R, Wang C, Zhu Y, Lin Y, Wyatt CN, Parrington J, Ma J, Evans AM, Galione A, Zhu MX.; ''NAADP mobilizes calcium from acidic organelles through two-pore channels.''; PubMed Europe PMC Scholia
  97. Vulpe C, Levinson B, Whitney S, Packman S, Gitschier J.; ''Isolation of a candidate gene for Menkes disease and evidence that it encodes a copper-transporting ATPase.''; PubMed Europe PMC Scholia
  98. Horisberger JD.; ''Amiloride-sensitive Na channels.''; PubMed Europe PMC Scholia
  99. Campbell HD, Kamei M, Claudianos C, Woollatt E, Sutherland GR, Suzuki Y, Hida M, Sugano S, Young IG.; ''Human and mouse homologues of the Drosophila melanogaster tweety (tty) gene: a novel gene family encoding predicted transmembrane proteins.''; PubMed Europe PMC Scholia
  100. Bull PC, Thomas GR, Rommens JM, Forbes JR, Cox DW.; ''The Wilson disease gene is a putative copper transporting P-type ATPase similar to the Menkes gene.''; PubMed Europe PMC Scholia
  101. Ruiz A, Bhat SP, Bok D.; ''Expression and synthesis of the Na,K-ATPase beta 2 subunit in human retinal pigment epithelium.''; PubMed Europe PMC Scholia
  102. Zdebik AA, Zifarelli G, Bergsdorf EY, Soliani P, Scheel O, Jentsch TJ, Pusch M.; ''Determinants of anion-proton coupling in mammalian endosomal CLC proteins.''; PubMed Europe PMC Scholia
  103. Ishibashi K, Marumo F.; ''Molecular cloning of a DEG/ENaC sodium channel cDNA from human testis.''; PubMed Europe PMC Scholia
  104. Frattini A, Pangrazio A, Susani L, Sobacchi C, Mirolo M, Abinun M, Andolina M, Flanagan A, Horwitz EM, Mihci E, Notarangelo LD, Ramenghi U, Teti A, Van Hove J, Vujic D, Young T, Albertini A, Orchard PJ, Vezzoni P, Villa A.; ''Chloride channel ClCN7 mutations are responsible for severe recessive, dominant, and intermediate osteopetrosis.''; PubMed Europe PMC Scholia
  105. Dierick HA, Adam AN, Escara-Wilke JF, Glover TW.; ''Immunocytochemical localization of the Menkes copper transport protein (ATP7A) to the trans-Golgi network.''; PubMed Europe PMC Scholia
  106. Petris MJ, Mercer JF.; ''The Menkes protein (ATP7A; MNK) cycles via the plasma membrane both in basal and elevated extracellular copper using a C-terminal di-leucine endocytic signal.''; PubMed Europe PMC Scholia
  107. Meij IC, Koenderink JB, van Bokhoven H, Assink KF, Groenestege WT, de Pont JJ, Bindels RJ, Monnens LA, van den Heuvel LP, Knoers NV.; ''Dominant isolated renal magnesium loss is caused by misrouting of the Na(+),K(+)-ATPase gamma-subunit.''; PubMed Europe PMC Scholia
  108. Qin A, Cheng TS, Lin Z, Pavlos NJ, Jiang Q, Xu J, Dai KR, Zheng MH.; ''Versatile roles of V-ATPases accessory subunit Ac45 in osteoclast formation and function.''; PubMed Europe PMC Scholia
  109. Latorre R, Zaelzer C, Brauchi S.; ''Structure-functional intimacies of transient receptor potential channels.''; PubMed Europe PMC Scholia
  110. Marquardt A, Stöhr H, Passmore LA, Krämer F, Rivera A, Weber BH.; ''Mutations in a novel gene, VMD2, encoding a protein of unknown properties cause juvenile-onset vitelliform macular dystrophy (Best's disease).''; PubMed Europe PMC Scholia
  111. de Weille JR, Bassilana F, Lazdunski M, Waldmann R.; ''Identification, functional expression and chromosomal localisation of a sustained human proton-gated cation channel.''; PubMed Europe PMC Scholia
  112. 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 myotonia.''; PubMed Europe PMC Scholia
  113. Choudhury K, McQuillin A, Puri V, Pimm J, Datta S, Thirumalai S, Krasucki R, Lawrence J, Bass NJ, Quested D, Crombie C, Fraser G, Walker N, Nadeem H, Johnson S, Curtis D, St Clair D, Gurling HM.; ''A genetic association study of chromosome 11q22-24 in two different samples implicates the FXYD6 gene, encoding phosphohippolin, in susceptibility to schizophrenia.''; PubMed Europe PMC Scholia
  114. Launay P, Fleig A, Perraud AL, Scharenberg AM, Penner R, Kinet JP.; ''TRPM4 is a Ca2+-activated nonselective cation channel mediating cell membrane depolarization.''; PubMed Europe PMC Scholia
  115. Taske NL, Eyre HJ, O'Brien RO, Sutherland GR, Denborough MA, Foster PS.; ''Molecular cloning of the cDNA encoding human skeletal muscle triadin and its localisation to chromosome 6q22-6q23.''; PubMed Europe PMC Scholia
  116. Yang YD, Cho H, Koo JY, Tak MH, Cho Y, Shim WS, Park SP, Lee J, Lee B, Kim BM, Raouf R, Shin YK, Oh U.; ''TMEM16A confers receptor-activated calcium-dependent chloride conductance.''; PubMed Europe PMC Scholia
  117. Niemeyer MI, Cid LP, Yusef YR, Briones R, Sepúlveda FV.; ''Voltage-dependent and -independent titration of specific residues accounts for complex gating of a ClC chloride channel by extracellular protons.''; PubMed Europe PMC Scholia
  118. Toyofuku T, Curotto Kurzydlowski K, Narayanan N, MacLennan DH.; ''Identification of Ser38 as the site in cardiac sarcoplasmic reticulum Ca(2+)-ATPase that is phosphorylated by Ca2+/calmodulin-dependent protein kinase.''; PubMed Europe PMC Scholia
  119. Hara-Chikuma M, Yang B, Sonawane ND, Sasaki S, Uchida S, Verkman AS.; ''ClC-3 chloride channels facilitate endosomal acidification and chloride accumulation.''; PubMed Europe PMC Scholia
  120. Keryanov S, Gardner KL.; ''Physical mapping and characterization of the human Na,K-ATPase isoform, ATP1A4.''; PubMed Europe PMC Scholia
  121. Heise CJ, Xu BE, Deaton SL, Cha SK, Cheng CJ, Earnest S, Sengupta S, Juang YC, Stippec S, Xu Y, Zhao Y, Huang CL, Cobb MH.; ''Serum and glucocorticoid-induced kinase (SGK) 1 and the epithelial sodium channel are regulated by multiple with no lysine (WNK) family members.''; PubMed Europe PMC Scholia
  122. Calafato S, Swain S, Hughes S, Kille P, Stürzenbaum SR.; ''Knock down of Caenorhabditis elegans cutc-1 exacerbates the sensitivity toward high levels of copper.''; PubMed Europe PMC Scholia
  123. Ambrosini L, Mercer JF.; ''Defective copper-induced trafficking and localization of the Menkes protein in patients with mild and copper-treated classical Menkes disease.''; PubMed Europe PMC Scholia
  124. Suzuki M, Mizuno A.; ''A novel human Cl(-) channel family related to Drosophila flightless locus.''; PubMed Europe PMC Scholia
  125. Li Y, Du J, Zhang P, Ding J.; ''Crystal structure of human copper homeostasis protein CutC reveals a potential copper-binding site.''; PubMed Europe PMC Scholia
  126. de Carvalho Aguiar P, Sweadner KJ, Penniston JT, Zaremba J, Liu L, Caton M, Linazasoro G, Borg M, Tijssen MA, Bressman SB, Dobyns WB, Brashear A, Ozelius LJ.; ''Mutations in the Na+/K+ -ATPase alpha3 gene ATP1A3 are associated with rapid-onset dystonia parkinsonism.''; PubMed Europe PMC Scholia
  127. Swoboda KJ, Kanavakis E, Xaidara A, Johnson JE, Leppert MF, Schlesinger-Massart MB, Ptacek LJ, Silver K, Youroukos S.; ''Alternating hemiplegia of childhood or familial hemiplegic migraine? A novel ATP1A2 mutation.''; PubMed Europe PMC Scholia
  128. Wang D, King SM, Quill TA, Doolittle LK, Garbers DL.; ''A new sperm-specific Na+/H+ exchanger required for sperm motility and fertility.''; PubMed Europe PMC Scholia

History

View all...
CompareRevisionActionTimeUserComment
112669view16:05, 9 October 2020ReactomeTeamReactome version 73
101586view11:45, 1 November 2018ReactomeTeamreactome version 66
101122view21:29, 31 October 2018ReactomeTeamreactome version 65
100650view20:03, 31 October 2018ReactomeTeamreactome version 64
100200view16:48, 31 October 2018ReactomeTeamreactome version 63
99751view15:14, 31 October 2018ReactomeTeamreactome version 62 (2nd attempt)
99315view12:46, 31 October 2018ReactomeTeamreactome version 62
93525view11:26, 9 August 2017ReactomeTeamNew pathway

External references

DataNodes

View all...
NameTypeDatabase referenceComment
ADPMetaboliteCHEBI:456216 (ChEBI)
ANO1 ProteinQ5XXA6 (Uniprot-TrEMBL)
ANO10 ProteinQ9NW15 (Uniprot-TrEMBL)
ANO1ProteinQ5XXA6 (Uniprot-TrEMBL)
ANO2 ProteinQ9NQ90 (Uniprot-TrEMBL)
ANO3 ProteinQ9BYT9 (Uniprot-TrEMBL)
ANO4 ProteinQ32M45 (Uniprot-TrEMBL)
ANO5 ProteinQ75V66 (Uniprot-TrEMBL)
ANO6 ProteinQ4KMQ2 (Uniprot-TrEMBL)
ANO7 ProteinQ6IWH7 (Uniprot-TrEMBL)
ANO8 ProteinQ9HCE9 (Uniprot-TrEMBL)
ANO9 ProteinA1A5B4 (Uniprot-TrEMBL)
ANOsComplexR-HSA-2684894 (Reactome)
APLsComplexR-ALL-947588 (Reactome)
APLsComplexR-ALL-947594 (Reactome)
ASIC trimers:H+:STOML3,(STOM)ComplexR-HSA-8863519 (Reactome)
ASIC trimers:H+ComplexR-HSA-2671886 (Reactome)
ASIC trimersComplexR-HSA-9650164 (Reactome)
ASIC1 ProteinP78348 (Uniprot-TrEMBL)
ASIC2 ProteinQ16515 (Uniprot-TrEMBL)
ASIC3 ProteinQ9UHC3 (Uniprot-TrEMBL)
ASIC4ProteinQ96FT7 (Uniprot-TrEMBL)
ASIC5 ProteinQ9NY37 (Uniprot-TrEMBL)
ASPH ProteinQ12797 (Uniprot-TrEMBL)
ATP10A ProteinO60312 (Uniprot-TrEMBL)
ATP10B ProteinO94823 (Uniprot-TrEMBL)
ATP10D ProteinQ9P241 (Uniprot-TrEMBL)
ATP11A ProteinP98196 (Uniprot-TrEMBL)
ATP11B ProteinQ9Y2G3 (Uniprot-TrEMBL)
ATP11C ProteinQ8NB49 (Uniprot-TrEMBL)
ATP12A ProteinP54707 (Uniprot-TrEMBL)
ATP13A1ProteinQ9HD20 (Uniprot-TrEMBL)
ATP13A2ProteinQ9NQ11 (Uniprot-TrEMBL)
ATP13A4 ProteinQ4VNC1 (Uniprot-TrEMBL)
ATP13A4, 5ComplexR-HSA-5252043 (Reactome)
ATP13A5 ProteinQ4VNC0 (Uniprot-TrEMBL)
ATP1A1 ProteinP05023 (Uniprot-TrEMBL)
ATP1A2 ProteinP50993 (Uniprot-TrEMBL)
ATP1A3 ProteinP13637 (Uniprot-TrEMBL)
ATP1A4 ProteinQ13733 (Uniprot-TrEMBL)
ATP1A:ATP1B:FXYDComplexR-HSA-936770 (Reactome)
ATP1B1 ProteinP05026 (Uniprot-TrEMBL)
ATP1B2 ProteinP14415 (Uniprot-TrEMBL)
ATP1B3 ProteinP54709 (Uniprot-TrEMBL)
ATP2A1 ProteinO14983 (Uniprot-TrEMBL)
ATP2A1-3ComplexR-HSA-418312 (Reactome)
ATP2A1-3ComplexR-HSA-427905 (Reactome)
ATP2A2 ProteinP16615 (Uniprot-TrEMBL)
ATP2A3 ProteinQ93084 (Uniprot-TrEMBL)
ATP2B1 ProteinP20020 (Uniprot-TrEMBL)
ATP2B1-4ComplexR-HSA-418306 (Reactome)
ATP2B2 ProteinQ01814 (Uniprot-TrEMBL)
ATP2B3 ProteinQ16720 (Uniprot-TrEMBL)
ATP2B4 ProteinP23634 (Uniprot-TrEMBL)
ATP2C1 ProteinP98194 (Uniprot-TrEMBL)
ATP2C1/2:Mg2+ComplexR-HSA-936921 (Reactome)
ATP2C2 ProteinO75185 (Uniprot-TrEMBL)
ATP4A ProteinP20648 (Uniprot-TrEMBL)
ATP4A/12A:ATP4BComplexR-HSA-937301 (Reactome)
ATP4B ProteinP51164 (Uniprot-TrEMBL)
ATP6AP1 ProteinQ15904 (Uniprot-TrEMBL)
ATP6AP1ProteinQ15904 (Uniprot-TrEMBL)
ATP6V0A1 ProteinQ93050 (Uniprot-TrEMBL)
ATP6V0A2 ProteinQ9Y487 (Uniprot-TrEMBL)
ATP6V0A4 ProteinQ9HBG4 (Uniprot-TrEMBL)
ATP6V0B ProteinQ99437 (Uniprot-TrEMBL)
ATP6V0C ProteinP27449 (Uniprot-TrEMBL)
ATP6V0D1 ProteinP61421 (Uniprot-TrEMBL)
ATP6V0D2 ProteinQ8N8Y2 (Uniprot-TrEMBL)
ATP6V0E1 ProteinO15342 (Uniprot-TrEMBL)
ATP6V0E2 ProteinQ8NHE4 (Uniprot-TrEMBL)
ATP6V1A ProteinP38606 (Uniprot-TrEMBL)
ATP6V1B1 ProteinP15313 (Uniprot-TrEMBL)
ATP6V1B2 ProteinP21281 (Uniprot-TrEMBL)
ATP6V1C1 ProteinP21283 (Uniprot-TrEMBL)
ATP6V1C2 ProteinQ8NEY4 (Uniprot-TrEMBL)
ATP6V1D ProteinQ9Y5K8 (Uniprot-TrEMBL)
ATP6V1E1 ProteinP36543 (Uniprot-TrEMBL)
ATP6V1E2 ProteinQ96A05 (Uniprot-TrEMBL)
ATP6V1F ProteinQ16864 (Uniprot-TrEMBL)
ATP6V1G1 ProteinO75348 (Uniprot-TrEMBL)
ATP6V1G2 ProteinO95670 (Uniprot-TrEMBL)
ATP6V1G3 ProteinQ96LB4 (Uniprot-TrEMBL)
ATP6V1H ProteinQ9UI12 (Uniprot-TrEMBL)
ATP7A ProteinQ04656 (Uniprot-TrEMBL)
ATP7A:PDZD11ComplexR-HSA-5358995 (Reactome)
ATP7BProteinP35670 (Uniprot-TrEMBL)
ATP8A1 ProteinQ9Y2Q0 (Uniprot-TrEMBL)
ATP8A2 ProteinQ9NTI2 (Uniprot-TrEMBL)
ATP8B1 ProteinO43520 (Uniprot-TrEMBL)
ATP8B2 ProteinP98198 (Uniprot-TrEMBL)
ATP8B3 ProteinO60423 (Uniprot-TrEMBL)
ATP8B4 ProteinQ8TF62 (Uniprot-TrEMBL)
ATP9A ProteinO75110 (Uniprot-TrEMBL)
ATP9B ProteinO43861 (Uniprot-TrEMBL)
ATPMetaboliteCHEBI:30616 (ChEBI)
BEST1 ProteinO76090 (Uniprot-TrEMBL)
BEST2 ProteinQ8NFU1 (Uniprot-TrEMBL)
BEST3 ProteinQ8N1M1 (Uniprot-TrEMBL)
BEST4 ProteinQ8NFU0 (Uniprot-TrEMBL)
BESTsComplexR-HSA-2744364 (Reactome)
BSND ProteinQ8WZ55 (Uniprot-TrEMBL)
C-terminus CLCAsComplexR-HSA-5333737 (Reactome)
CALM1 ProteinP0DP23 (Uniprot-TrEMBL)
CALM1ProteinP0DP23 (Uniprot-TrEMBL)
CAMK2 heteromer:CALM:4xCa2+ComplexR-HSA-444601 (Reactome)
CASQ1 polymer R-HSA-2855198 (Reactome)
CASQ2 polymer R-HSA-2855188 (Reactome)
CLCA1 (22-?) ProteinA8K7I4 (Uniprot-TrEMBL)
CLCA1 (?-914) ProteinA8K7I4 (Uniprot-TrEMBL)
CLCA1 ProteinA8K7I4 (Uniprot-TrEMBL)
CLCA2 (32-?) ProteinQ9UQC9 (Uniprot-TrEMBL)
CLCA2 (?-943) ProteinQ9UQC9 (Uniprot-TrEMBL)
CLCA2 ProteinQ9UQC9 (Uniprot-TrEMBL)
CLCA3P (21-?) ProteinQ9Y6N3 (Uniprot-TrEMBL)
CLCA3P (?-262) ProteinQ9Y6N3 (Uniprot-TrEMBL)
CLCA3P ProteinQ9Y6N3 (Uniprot-TrEMBL)
CLCA4 (22-?) ProteinQ14CN2 (Uniprot-TrEMBL)
CLCA4 (?-919) ProteinQ14CN2 (Uniprot-TrEMBL)
CLCA4 ProteinQ14CN2 (Uniprot-TrEMBL)
CLCAsComplexR-HSA-5333763 (Reactome)
CLCN1 ProteinP35523 (Uniprot-TrEMBL)
CLCN1/2/KA/KBComplexR-HSA-2744251 (Reactome)
CLCN2 ProteinP51788 (Uniprot-TrEMBL)
CLCN3ProteinP51790 (Uniprot-TrEMBL)
CLCN4 ProteinP51793 (Uniprot-TrEMBL)
CLCN4/5/6ComplexR-HSA-2730688 (Reactome)
CLCN5 ProteinP51795 (Uniprot-TrEMBL)
CLCN6 ProteinP51797 (Uniprot-TrEMBL)
CLCN7 ProteinP51798 (Uniprot-TrEMBL)
CLCN7:OSTM1ComplexR-HSA-2730964 (Reactome)
CLCNKA ProteinP51800 (Uniprot-TrEMBL)
CLCNKB ProteinP51801 (Uniprot-TrEMBL)
CLIC2ProteinO15247 (Uniprot-TrEMBL)
CUTC ProteinQ9NTM9 (Uniprot-TrEMBL)
CUTC tetramerComplexR-HSA-5334801 (Reactome)
CUTC:4xCu+ComplexR-HSA-5336194 (Reactome)
Ca2+ MetaboliteCHEBI:29108 (ChEBI)
Ca2+MetaboliteCHEBI:29108 (ChEBI)
Cl-MetaboliteCHEBI:17996 (ChEBI)
Cu1+ MetaboliteCHEBI:49552 (ChEBI)
Cu1+MetaboliteCHEBI:49552 (ChEBI)
Cu2+MetaboliteCHEBI:29036 (ChEBI)
FKBP1B ProteinP68106 (Uniprot-TrEMBL)
FXYD1 ProteinO00168 (Uniprot-TrEMBL)
FXYD2 ProteinP54710 (Uniprot-TrEMBL)
FXYD3 ProteinQ14802 (Uniprot-TrEMBL)
FXYD4 ProteinP59646 (Uniprot-TrEMBL)
FXYD6 ProteinQ9H0Q3 (Uniprot-TrEMBL)
FXYD7 ProteinP58549 (Uniprot-TrEMBL)
H+ MetaboliteCHEBI:15378 (ChEBI)
H+MetaboliteCHEBI:15378 (ChEBI)
H2OMetaboliteCHEBI:15377 (ChEBI)
HCO3-MetaboliteCHEBI:17544 (ChEBI)
K+MetaboliteCHEBI:29103 (ChEBI)
Li+ MetaboliteCHEBI:49713 (ChEBI)
MCOLN1 ProteinQ9GZU1 (Uniprot-TrEMBL)
MCOLN2 ProteinQ8IZK6 (Uniprot-TrEMBL)
MCOLN3 ProteinQ8TDD5 (Uniprot-TrEMBL)
Mg2+ MetaboliteCHEBI:18420 (ChEBI)
Mn2+MetaboliteCHEBI:29035 (ChEBI)
N-terminus CLCAsComplexR-HSA-5333722 (Reactome)
NAADPMetaboliteCHEBI:76072 (ChEBI)
NALCN ProteinQ8IZF0 (Uniprot-TrEMBL)
NALCN:UNC79:UNC80ComplexR-HSA-2730669 (Reactome)
NEDD4L ProteinQ96PU5 (Uniprot-TrEMBL)
NSAIDMetaboliteCHEBI:35475 (ChEBI)
Na+ MetaboliteCHEBI:29101 (ChEBI)
Na+/Li+ComplexR-ALL-2889062 (Reactome)
Na+/Li+ComplexR-ALL-2889081 (Reactome)
Na+MetaboliteCHEBI:29101 (ChEBI)
OSTM1 ProteinQ86WC4 (Uniprot-TrEMBL)
P-type ATPases type IVComplexR-HSA-939735 (Reactome)
PDZD11 ProteinQ5EBL8 (Uniprot-TrEMBL)
PE MetaboliteCHEBI:16038 (ChEBI)
PLN ProteinP26678 (Uniprot-TrEMBL)
PLN pentamerComplexR-HSA-5578811 (Reactome)
PS MetaboliteCHEBI:18303 (ChEBI)
PiMetaboliteCHEBI:18367 (ChEBI)
RAF1 ProteinP04049 (Uniprot-TrEMBL)
RAF1:SGK:TSC22D3:WPPComplexR-HSA-2682342 (Reactome)
RPS27A(1-76) ProteinP62979 (Uniprot-TrEMBL)
RYR

tetramer:FKBP1B tetramer:CASQ

polymer:TRDN:junctin
ComplexR-HSA-2855167 (Reactome)
RYR1 ProteinP21817 (Uniprot-TrEMBL)
RYR2 ProteinQ92736 (Uniprot-TrEMBL)
RYR3 ProteinQ15413 (Uniprot-TrEMBL)
SCNN channelsComplexR-HSA-2672342 (Reactome)
SCNN1A ProteinP37088 (Uniprot-TrEMBL)
SCNN1B ProteinP51168 (Uniprot-TrEMBL)
SCNN1D ProteinP51172 (Uniprot-TrEMBL)
SCNN1G ProteinP51170 (Uniprot-TrEMBL)
SGK1 ProteinO00141 (Uniprot-TrEMBL)
SGK2 ProteinQ9HBY8 (Uniprot-TrEMBL)
SGK3 ProteinQ96BR1 (Uniprot-TrEMBL)
SLC17A3(1-498)ProteinO00476 (Uniprot-TrEMBL)
SLC17A3ProteinO00476 (Uniprot-TrEMBL)
SLC9B1 ProteinQ4ZJI4 (Uniprot-TrEMBL)
SLC9B1/C2ComplexR-HSA-2889056 (Reactome)
SLC9B2ProteinQ86UD5 (Uniprot-TrEMBL)
SLC9C1ProteinQ4G0N8 (Uniprot-TrEMBL)
SLC9C2 ProteinQ5TAH2 (Uniprot-TrEMBL)
SLNProteinO00631 (Uniprot-TrEMBL)
SRIProteinP30626 (Uniprot-TrEMBL)
STOM ProteinP27105 (Uniprot-TrEMBL)
STOML3 ProteinQ8TAV4 (Uniprot-TrEMBL)
STOML3, (STOM)ComplexR-HSA-8863527 (Reactome)
TCIRG1 ProteinQ13488 (Uniprot-TrEMBL)
TPCN1 ProteinQ9ULQ1 (Uniprot-TrEMBL)
TPCN1/2ComplexR-HSA-2685517 (Reactome)
TPCN2 ProteinQ8NHX9 (Uniprot-TrEMBL)
TRDN ProteinQ13061 (Uniprot-TrEMBL)
TRPA1 ProteinO75762 (Uniprot-TrEMBL)
TRPC1 ProteinP48995 (Uniprot-TrEMBL)
TRPC3(1-848) ProteinQ13507 (Uniprot-TrEMBL)
TRPC4 ProteinQ9UBN4 (Uniprot-TrEMBL)
TRPC4AP ProteinQ8TEL6 (Uniprot-TrEMBL)
TRPC5 ProteinQ9UL62 (Uniprot-TrEMBL)
TRPC6 ProteinQ9Y210 (Uniprot-TrEMBL)
TRPC7 ProteinQ9HCX4 (Uniprot-TrEMBL)
TRPM1 ProteinQ7Z4N2 (Uniprot-TrEMBL)
TRPM2 ProteinO94759 (Uniprot-TrEMBL)
TRPM3 ProteinQ9HCF6 (Uniprot-TrEMBL)
TRPM4 ProteinQ8TD43 (Uniprot-TrEMBL)
TRPM4,5ComplexR-HSA-3295585 (Reactome)
TRPM5 ProteinQ9NZQ8 (Uniprot-TrEMBL)
TRPM6 ProteinQ9BX84 (Uniprot-TrEMBL)
TRPM7 ProteinQ96QT4 (Uniprot-TrEMBL)
TRPM8 ProteinQ7Z2W7 (Uniprot-TrEMBL)
TRPV1 ProteinQ8NER1 (Uniprot-TrEMBL)
TRPV2 ProteinQ9Y5S1 (Uniprot-TrEMBL)
TRPV3 ProteinQ8NET8 (Uniprot-TrEMBL)
TRPV4 ProteinQ9HBA0 (Uniprot-TrEMBL)
TRPV5 ProteinQ9NQA5 (Uniprot-TrEMBL)
TRPV6 ProteinQ9H1D0 (Uniprot-TrEMBL)
TRPsComplexR-HSA-3295582 (Reactome)
TSC22D3 ProteinQ99576 (Uniprot-TrEMBL)
TTYH1-3ProteinQ9H313-3 (Uniprot-TrEMBL)
TTYH2 ProteinQ9BSA4 (Uniprot-TrEMBL)
TTYH2/3ComplexR-HSA-2744253 (Reactome)
TTYH3 ProteinQ9C0H2 (Uniprot-TrEMBL)
UBA52(1-76) ProteinP62987 (Uniprot-TrEMBL)
UBB(1-76) ProteinP0CG47 (Uniprot-TrEMBL)
UBB(153-228) ProteinP0CG47 (Uniprot-TrEMBL)
UBB(77-152) ProteinP0CG47 (Uniprot-TrEMBL)
UBC(1-76) ProteinP0CG48 (Uniprot-TrEMBL)
UBC(153-228) ProteinP0CG48 (Uniprot-TrEMBL)
UBC(229-304) ProteinP0CG48 (Uniprot-TrEMBL)
UBC(305-380) ProteinP0CG48 (Uniprot-TrEMBL)
UBC(381-456) ProteinP0CG48 (Uniprot-TrEMBL)
UBC(457-532) ProteinP0CG48 (Uniprot-TrEMBL)
UBC(533-608) ProteinP0CG48 (Uniprot-TrEMBL)
UBC(609-684) ProteinP0CG48 (Uniprot-TrEMBL)
UBC(77-152) ProteinP0CG48 (Uniprot-TrEMBL)
UNC79 ProteinQ9P2D8 (Uniprot-TrEMBL)
UNC80 ProteinQ8N2C7 (Uniprot-TrEMBL)
Ub-SCNN channelsComplexR-HSA-2684917 (Reactome)
Ub-SCNN1A ProteinP37088 (Uniprot-TrEMBL)
Ub-SCNN1G ProteinP51170 (Uniprot-TrEMBL)
UbComplexR-HSA-113595 (Reactome)
V-ATPase:ATP6AP1ComplexR-HSA-5252081 (Reactome)
V-ATPaseComplexR-HSA-912600 (Reactome)
WNK1 ProteinQ9H4A3 (Uniprot-TrEMBL)
WNK2 ProteinQ9Y3S1 (Uniprot-TrEMBL)
WNK3 ProteinQ9BYP7 (Uniprot-TrEMBL)
WNK4 ProteinQ96J92 (Uniprot-TrEMBL)
WNKsComplexR-HSA-2684956 (Reactome)
WWP1 ProteinQ9H0M0 (Uniprot-TrEMBL)
amilorideMetaboliteCHEBI:2639 (ChEBI)
cationMetaboliteCHEBI:36916 (ChEBI)
divalent metal cationMetaboliteCHEBI:60240 (ChEBI)
p-S-RIPK1:p-S199,227-RIPK3 oligomer R-HSA-5218908 (Reactome)
p-S-RIPK1:p-S199,227-RIPK3:p-T357,S358-MLKL oligomerComplexR-HSA-5357794 (Reactome)
p-S16-PLN ProteinP26678 (Uniprot-TrEMBL)
p-S16-PLN pentamerComplexR-HSA-5578810 (Reactome)
p-T286-CAMK2A ProteinQ9UQM7 (Uniprot-TrEMBL)
p-T287-CAMK2B ProteinQ13554 (Uniprot-TrEMBL)
p-T287-CAMK2D ProteinQ13557 (Uniprot-TrEMBL)
p-T287-CAMK2G ProteinQ13555 (Uniprot-TrEMBL)
p-T357,S358-MLKL oligomer R-HSA-5357857 (Reactome)
urateMetaboliteCHEBI:17775 (ChEBI)

Annotated Interactions

View all...
SourceTargetTypeDatabase referenceComment
ADPArrowR-HSA-2682349 (Reactome)
ADPArrowR-HSA-5251989 (Reactome)
ADPArrowR-HSA-5692462 (Reactome)
ADPArrowR-HSA-5692480 (Reactome)
ADPArrowR-HSA-936802 (Reactome)
ADPArrowR-HSA-936883 (Reactome)
ADPArrowR-HSA-936895 (Reactome)
ADPArrowR-HSA-936897 (Reactome)
ADPArrowR-HSA-937311 (Reactome)
ADPArrowR-HSA-939763 (Reactome)
ADPArrowR-HSA-947591 (Reactome)
ANO1mim-catalysisR-HSA-9659568 (Reactome)
ANOsmim-catalysisR-HSA-2684901 (Reactome)
APLsArrowR-HSA-939763 (Reactome)
APLsArrowR-HSA-947591 (Reactome)
APLsR-HSA-939763 (Reactome)
APLsR-HSA-947591 (Reactome)
ASIC trimers:H+:STOML3,(STOM)ArrowR-HSA-8863494 (Reactome)
ASIC trimers:H+ArrowR-HSA-9650165 (Reactome)
ASIC trimers:H+R-HSA-8863494 (Reactome)
ASIC trimers:H+mim-catalysisR-HSA-2671885 (Reactome)
ASIC trimersR-HSA-9650165 (Reactome)
ASIC4TBarR-HSA-2671885 (Reactome)
ATP13A1mim-catalysisR-HSA-5692462 (Reactome)
ATP13A2mim-catalysisR-HSA-5692480 (Reactome)
ATP13A4, 5mim-catalysisR-HSA-5251989 (Reactome)
ATP1A:ATP1B:FXYDmim-catalysisR-HSA-936897 (Reactome)
ATP2A1-3mim-catalysisR-HSA-418365 (Reactome)
ATP2A1-3mim-catalysisR-HSA-427910 (Reactome)
ATP2B1-4mim-catalysisR-HSA-418309 (Reactome)
ATP2C1/2:Mg2+mim-catalysisR-HSA-936883 (Reactome)
ATP4A/12A:ATP4Bmim-catalysisR-HSA-937311 (Reactome)
ATP6AP1R-HSA-5252133 (Reactome)
ATP7A:PDZD11mim-catalysisR-HSA-936802 (Reactome)
ATP7Bmim-catalysisR-HSA-936895 (Reactome)
ATPArrowR-HSA-2855020 (Reactome)
ATPR-HSA-2682349 (Reactome)
ATPR-HSA-5251989 (Reactome)
ATPR-HSA-5692462 (Reactome)
ATPR-HSA-5692480 (Reactome)
ATPR-HSA-936802 (Reactome)
ATPR-HSA-936883 (Reactome)
ATPR-HSA-936895 (Reactome)
ATPR-HSA-936897 (Reactome)
ATPR-HSA-937311 (Reactome)
ATPR-HSA-939763 (Reactome)
ATPR-HSA-947591 (Reactome)
BESTsmim-catalysisR-HSA-2744361 (Reactome)
BESTsmim-catalysisR-HSA-2752067 (Reactome)
C-terminus CLCAsArrowR-HSA-5333671 (Reactome)
CALM1TBarR-HSA-2855020 (Reactome)
CAMK2 heteromer:CALM:4xCa2+ArrowR-HSA-427910 (Reactome)
CLCAsR-HSA-5333671 (Reactome)
CLCAsmim-catalysisR-HSA-5333671 (Reactome)
CLCN1/2/KA/KBmim-catalysisR-HSA-2744228 (Reactome)
CLCN3mim-catalysisR-HSA-2731002 (Reactome)
CLCN4/5/6mim-catalysisR-HSA-2730692 (Reactome)
CLCN7:OSTM1mim-catalysisR-HSA-2730959 (Reactome)
CLIC2TBarR-HSA-2855020 (Reactome)
CUTC tetramerR-HSA-5334788 (Reactome)
CUTC:4xCu+ArrowR-HSA-5334788 (Reactome)
Ca2+ArrowR-HSA-2684901 (Reactome)
Ca2+ArrowR-HSA-2685505 (Reactome)
Ca2+ArrowR-HSA-2744242 (Reactome)
Ca2+ArrowR-HSA-2744361 (Reactome)
Ca2+ArrowR-HSA-2855020 (Reactome)
Ca2+ArrowR-HSA-3295579 (Reactome)
Ca2+ArrowR-HSA-418309 (Reactome)
Ca2+ArrowR-HSA-418365 (Reactome)
Ca2+ArrowR-HSA-427910 (Reactome)
Ca2+ArrowR-HSA-5333671 (Reactome)
Ca2+ArrowR-HSA-936883 (Reactome)
Ca2+ArrowR-HSA-9659568 (Reactome)
Ca2+R-HSA-2685505 (Reactome)
Ca2+R-HSA-2855020 (Reactome)
Ca2+R-HSA-3295579 (Reactome)
Ca2+R-HSA-418309 (Reactome)
Ca2+R-HSA-418365 (Reactome)
Ca2+R-HSA-427910 (Reactome)
Ca2+R-HSA-936883 (Reactome)
Ca2+TBarR-HSA-2730664 (Reactome)
Cl-ArrowR-HSA-2684901 (Reactome)
Cl-ArrowR-HSA-2730692 (Reactome)
Cl-ArrowR-HSA-2730959 (Reactome)
Cl-ArrowR-HSA-2731002 (Reactome)
Cl-ArrowR-HSA-2744228 (Reactome)
Cl-ArrowR-HSA-2744242 (Reactome)
Cl-ArrowR-HSA-2744349 (Reactome)
Cl-ArrowR-HSA-2744361 (Reactome)
Cl-ArrowR-HSA-9659568 (Reactome)
Cl-R-HSA-2684901 (Reactome)
Cl-R-HSA-2730692 (Reactome)
Cl-R-HSA-2730959 (Reactome)
Cl-R-HSA-2731002 (Reactome)
Cl-R-HSA-2744228 (Reactome)
Cl-R-HSA-2744242 (Reactome)
Cl-R-HSA-2744349 (Reactome)
Cl-R-HSA-2744361 (Reactome)
Cl-R-HSA-9659568 (Reactome)
Cu1+R-HSA-5334788 (Reactome)
Cu2+ArrowR-HSA-936802 (Reactome)
Cu2+ArrowR-HSA-936895 (Reactome)
Cu2+R-HSA-936802 (Reactome)
Cu2+R-HSA-936895 (Reactome)
H+ArrowR-HSA-2730692 (Reactome)
H+ArrowR-HSA-2730959 (Reactome)
H+ArrowR-HSA-2731002 (Reactome)
H+ArrowR-HSA-2872444 (Reactome)
H+ArrowR-HSA-2872463 (Reactome)
H+ArrowR-HSA-2889070 (Reactome)
H+ArrowR-HSA-418365 (Reactome)
H+ArrowR-HSA-427910 (Reactome)
H+ArrowR-HSA-937311 (Reactome)
H+R-HSA-2730692 (Reactome)
H+R-HSA-2730959 (Reactome)
H+R-HSA-2731002 (Reactome)
H+R-HSA-2872444 (Reactome)
H+R-HSA-2872463 (Reactome)
H+R-HSA-2889070 (Reactome)
H+R-HSA-418365 (Reactome)
H+R-HSA-427910 (Reactome)
H+R-HSA-937311 (Reactome)
H+R-HSA-9650165 (Reactome)
H2OR-HSA-5251989 (Reactome)
H2OR-HSA-5692462 (Reactome)
H2OR-HSA-5692480 (Reactome)
H2OR-HSA-936802 (Reactome)
H2OR-HSA-936883 (Reactome)
H2OR-HSA-936895 (Reactome)
H2OR-HSA-936897 (Reactome)
H2OR-HSA-937311 (Reactome)
H2OR-HSA-939763 (Reactome)
H2OR-HSA-947591 (Reactome)
HCO3-ArrowR-HSA-2752067 (Reactome)
HCO3-R-HSA-2752067 (Reactome)
K+ArrowR-HSA-936897 (Reactome)
K+ArrowR-HSA-937311 (Reactome)
K+R-HSA-936897 (Reactome)
K+R-HSA-937311 (Reactome)
Mn2+ArrowR-HSA-5692462 (Reactome)
Mn2+R-HSA-5692462 (Reactome)
N-terminus CLCAsArrowR-HSA-2684901 (Reactome)
N-terminus CLCAsArrowR-HSA-2744242 (Reactome)
N-terminus CLCAsArrowR-HSA-2744361 (Reactome)
N-terminus CLCAsArrowR-HSA-5333671 (Reactome)
N-terminus CLCAsArrowR-HSA-9659568 (Reactome)
NAADPArrowR-HSA-2685505 (Reactome)
NALCN:UNC79:UNC80mim-catalysisR-HSA-2730664 (Reactome)
NSAIDTBarR-HSA-2671885 (Reactome)
Na+/Li+ArrowR-HSA-2889070 (Reactome)
Na+/Li+R-HSA-2889070 (Reactome)
Na+ArrowR-HSA-2671885 (Reactome)
Na+ArrowR-HSA-2672334 (Reactome)
Na+ArrowR-HSA-2730664 (Reactome)
Na+ArrowR-HSA-2872444 (Reactome)
Na+ArrowR-HSA-2872463 (Reactome)
Na+ArrowR-HSA-2872498 (Reactome)
Na+ArrowR-HSA-3295580 (Reactome)
Na+ArrowR-HSA-936897 (Reactome)
Na+R-HSA-2671885 (Reactome)
Na+R-HSA-2672334 (Reactome)
Na+R-HSA-2730664 (Reactome)
Na+R-HSA-2872444 (Reactome)
Na+R-HSA-2872463 (Reactome)
Na+R-HSA-2872498 (Reactome)
Na+R-HSA-3295580 (Reactome)
Na+R-HSA-936897 (Reactome)
P-type ATPases type IVmim-catalysisR-HSA-939763 (Reactome)
P-type ATPases type IVmim-catalysisR-HSA-947591 (Reactome)
PLN pentamerTBarR-HSA-427910 (Reactome)
PiArrowR-HSA-2682349 (Reactome)
PiArrowR-HSA-2872498 (Reactome)
PiArrowR-HSA-5251989 (Reactome)
PiArrowR-HSA-5692462 (Reactome)
PiArrowR-HSA-5692480 (Reactome)
PiArrowR-HSA-936802 (Reactome)
PiArrowR-HSA-936883 (Reactome)
PiArrowR-HSA-936895 (Reactome)
PiArrowR-HSA-936897 (Reactome)
PiArrowR-HSA-937311 (Reactome)
PiArrowR-HSA-939763 (Reactome)
PiArrowR-HSA-947591 (Reactome)
PiR-HSA-2872498 (Reactome)
R-HSA-2671885 (Reactome) Acid-sensing ion channels 1, 2, 3 and 5 (ASIC1, 2, 3 and 5, aka amiloride-sensitive cation channels) are homotrimeric, multi-pass membrane proteins which can transport sodium (Na+) when activated by extracellular protons. Members of the ASIC family are sensitive to amiloride and function in neurotransmission. The encoded proteins function in learning, pain transduction, touch sensation, and development of memory and fear. Many neuronal diseases cause acidosis, accompanied by pain and neuronal damage; ASICs can mediate the pathophysiological effects seen in acidiosis (Wang & Xu 2011, Qadri et al. 2012). The diuretic drug amiloride inhibits these channels, resulting in analgesic effects. NSAIDs (Nonsteroidal anti-inflammatory drugs) can also inhibit ASICs to produce analgesia (Voilley et al. 2001). ASICs are also partially permeable to Ca2+, Li+ and K+ (not shown here). ASIC1 and 2 are expressed mostly in brain (Garcia-Anoveros et al. 1997, Price et al. 1996), ASIC3 is strongly expressed in testis (de Weille et al. 1998, Ishibashi & Marumo 1998) and ASIC5 is found mainly in intestine (Schaefer et al. 2000). ASIC4 subunits do not form functional channels and it's activity is unknown. It could play a part in regulating other ASIC activity (Donier et al. 2008).
R-HSA-2672334 (Reactome) Amiloride-sensitive sodium channels (SCNNs, aka ENaCs, epithelial Na+ channels, non voltage-gated sodium channels) belong to the epithelial Na+ channel/degenerin (ENaC/DEG) protein family and mediate the transport of Na+ (and associated water) through the apical membrane of epithelial cells in kidney, colon and lungs. This makes SCNNs important determinants of systemic blood pressure. The physiological activator for SCNNs is unknown but as they belong in the same family as acid-sensitive ion channels (ASICs, which are mediated by protons), these may also be the activating ligands for SCNNs. SCNNs are probable heterotrimers comprising an alpha (or interchangeable delta subunit), beta and gamma subunit (Horisberger 1998).
R-HSA-2682349 (Reactome) Amiloride-sensitive sodium channels (SCNNs, aka ENaCs, epithelial Na+ channels, non voltage-gated sodium channels) comprises three subunits (alpha, beta and gamma) and plays an essential role in Na+ and fluid absorption in the kidney, colon and lung. The number of channels at the cell's surface (consequently its function) can be regulated. This is achieved by ubiquitination of SCNN via E3 ubiquitin-protein ligases (NED4L and WPP1) (Staub et al. 2000, Farr et al. 2000). NED4L/WPP1 is found in a signaling complex including Raf1 (RAF proto-oncogene serine/threonine-protein kinase), SGK (serum/glucocorticoid-regulated kinase) and GILZ (glucocorticoid-induced leucine zipper protein, TSC22D3) (Soundararajan et al. 2009). Ubiquitinated SCNN (Ub-SCNN) is targeted for degradation so a lesser number of channels are present at the cell surface, reducing the amount of Na+ absorption. Proline-rich sequences at the C-terminus of SCNNs include the PY motif containing a PPxY sequence. PY motifs bind WW domains of NED4L/WPP1. Protein kinases with no lysine K (WNKs) can activate SCNN activity by interacting non-enzymatically with the signaling complex, specifically SGK although the mechanism is unknown (Heise et al. 2010).
R-HSA-2684901 (Reactome) Calcium-activated chloride channels (CaCCs) are ubiquitously expressed and implicated in physiological processes such as sensory transduction, fertilization, epithelial secretion, and smooth muscle contraction. The anoctamin family of transmembrane proteins (ANO, TMEM16) belong to CaCCs and have been shown to transport Cl- ions when activated by intracellular Ca2+ (Galietta 2009, Huang et al. 2012). There are currently 10 members, ANO1-10, all having a similar structure, with eight putative transmembrane domains and cytosolic amino- and carboxy-termini. ANO1 and 2 possess Ca2+ activated Cl- transport activity (Yang et al. 2008, Scudieri et al. 2012) while the remaining members also show some demonstrable activity (Tian et al. 2012).
R-HSA-2685505 (Reactome) Calcium (Ca2+) can be mobilised from intracellular stores by the presence of nicotinic acid adenine dinucleotide phosphate (NAADP). Two pore calcium channel proteins 1 and 2 (TPCN1 and 2) are expressed on endosomal (not shown here) and lysosomal membranes and mediate the mobilization of Ca2+ from these organelles when activated by NAADP (Brailoiu et al. 2009, Calcraft et al. 2009).
R-HSA-2730664 (Reactome) The sodium leak channel non-selective protein NALCN, a nonselective cation channel, forms the background Na+ leak conductance and controls neuronal excitability (Lu et al. 2007, Ren 2011). Mice with mutant NALCN have a severely disrupted respiratory rhythm and die within 24 hours of birth. Calcium (Ca2+) influences neuronal excitability via the NALCN:UNC79:UNC80 complex, with high Ca2+ concentrations inhibiting transport of Na+ (Lu et al. 2010). Mutations in human NALCN lead to complex neurodevelopmental syndromes, including infantile hypotonia with psychomotor retardation and characteristic facies (IHPRF) and congenital contractures of limbs and face, hypotonia and developmental delay (CLIFAHDD) (Bouasse et al. 2019).
R-HSA-2730692 (Reactome) The H+/Cl- exchange transporters CLCN4 (Kawasaki et al. 1999, Zdebik et al. 2008), CLCN5 (Zdebik et al. 2008) and CLCN6 (Neagoe et al. 2010) mediate the exchange of endosomal Cl- for cytosolic H+ across endosomal membranes, contributing to the acidification of endosomes.
R-HSA-2730959 (Reactome) Chloride channel 7 comprises H+/Cl- exchange transporter 7 (CLCN7) and osteopetrosis-associated transmembrane protein 1 (OSTM1) (Leisle et al. 2011). This complex localises to the lysosomal membrane where it mediates the exchange of Cl- and H+ ions, perhaps playing a role in the acidification of the lysosome (Graves et al. 2008).

Defects in CLCN7 cause osteopetrosis autosomal recessive types 2 and 4 (OPTB2, MIM:166600 and OPTB4, MIM:611490) (Frattini et al. 2003, Pangrazio et al. 2010). Defects in OSTM1 cause osteopetrosis autosomal recessive type 5 (OPTB5, MIM:259720) (Pangrazio et al. 2006).
R-HSA-2731002 (Reactome) The H+/Cl- exchange transporter CLCN3 (Borsani et al. 1995) mediates the exchange of endosomal Cl- for cytosolic H+ across late endosomal membranes, contributing to the acidification of endosomes. The activity of CLCN3 is inferred from experiments in mice (Stobrawa et al. 2001, Hara-Chikuma et al. 2005).
R-HSA-2744228 (Reactome) Chloride channel proteins 1, 2, Ka and Kb (CLCN1, 2, KA, KB) can mediate Cl- influx across the plasma membrane of almost all cells. CLCN1 is expressed mainly on skeletal muscle where it is involved in the electrical stability of the muscle. CLCN1 is thought to function in a homotetrameric form (Steimeyer et al. 1994). CLCN2 is ubiquitously expressed, playing a role in the regulation of cell volume (Cid et al. 1995, Niemeyer et al. 2009). Defects in CLCN1 cause myotonia congenita, an autosomal dominant disease (MCD aka Thomsen disease, MIM:160800). It is characterized by muscle stiffness due to delayed relaxation, resulting from membrane hyperexcitability (Meyer-Kleine et al. 1995, Steimeyer et al. 1994). Defects in CLCN1 also cause autosomal recessive myotonia congenita (MCR aka Becker disease, MIM:255700) (Koch et al. 1992, Meyer-Kleine et al. 1995), a nondystrophic skeletal muscle disorder characterized by muscle stiffness and an inability of the muscle to relax after voluntary contraction. Becker disease is more common and more severe than Thomsen disease.

CLCNKA and B (Kieferle et al. 1994) are predominantly expressed in distal nephron segments of the kidney (Takeuchi et al. 1995) and the inner ear (Estevez et al. 2001, Schlingmann et al. 2004). They are tightly associated with their essential beta subunit barttin (BSND), requiring it to be fully functional channels (Fischer et al. 2010, Scholl et al. 2006). These channels bound to BSND are essential for renal Cl- reabsorption (Waldegger & Jentsch 2000) and K+ recycling in the inner ear (Estevez et al. 2001). Defects in CLCNKA and B cause Bartter syndrome type 4B (BS4B; MIM:613090) characterized by impaired salt reabsorption and sensorineural deafness (Schlingmann et al. 2004, Nozu et al. 2008). Defects in BSND cause Bartter syndrome type 4A (BS4A aka infantile Bartter syndrome with sensorineural deafness; MIM:602522) characterized by impaired salt reabsorption in the thick ascending loop of Henle and sensorineural deafness (Birkenhager et al. 2001, Nozu et al. 2008).
R-HSA-2744242 (Reactome) Human homologues 2 and 3 (TTYH2 and 3) mediate the efflux of Cl- from cells in response to the increase in intracellular Ca2+ levels (Suzuki & Mizuno 2004, Suzuki 2006).
R-HSA-2744349 (Reactome) Protein tweety homolog 1 (TTYH1) has 5 isoforms. Isoform 3 (Campbell et al. 2000) mediates the Ca+-independent efflux of Cl- across plasma membranes (Suzuki & Mizuno 2004, Suzuki 2006).
R-HSA-2744361 (Reactome) Bestrophins 1-4 (BEST1-4, aka vitelliform macular dystrophy proteins) mediate cytosolic Cl- efflux across plasma membranes. This transport is sensitive to intracellular Ca2+ concentrations (Sun et al. 2002, Tsunenari et al. 2003). Mutations in bestrophins that impair their function are implicated in macular degeneration in the eye. Defects in BEST1 cause vitelliform macular dystrophy (BVMD, Best's disease, MIM:153700), an autosomal dominant form of macular degeneration that usually begins in childhood and is characterized lesions due to abnormal accumulation of lipofuscin within and beneath retinal pigment epithelium (RPE) cells (Marquardt et al. 1998, Petrukhin et al. 1998).
R-HSA-2752067 (Reactome) Many Cl- channels such as CFTR, ClC, CaCC, and ligand-gated anion channels are permeable to bicarbonate (HCO3-) which is an important anion in the regulation of pH. Many tissues, including retinal pigment epithelium (RPE), utilize HCO3- transporters to mediate transport of HCO3-. Bestrophns 1-4 (BEST1-4, aka vitelliform macular dystrophy proteins) have high permeability to HCO3- (Hu & Hartzell 2008). Defective BEST1 may play a role in macular degeneration in the eye due to impaired HCO3- and Cl- conductance (Hu & Hartzell 2008).
R-HSA-2855020 (Reactome) Ryanodine receptors (RYRs) are located in the sarcoplasmic reticulum (SR) membrane and mediate the release of Ca2+ from intracellular stores during excitation-contraction (EC) coupling in both cardiac and skeletal muscle. RYRs are the largest known ion channels (>2MDa) and are functional in their homotetrameric forms. There are three mammalian isoforms (RYR1-3); RYR1 is prominent in skeletal muscle (Zorzato et al. 1990), RYR2 in cardiac muscle (Tunwell et al. 1996) and RYR3 is found in the brain (Nakashima et al. 1997, Lanner et al. 2010). The function of RYRs are controlled by peptidyl-prolyl cis-trans isomerase (FKBP1B), intracellular Ca2+-binding proteins calsequestrin 1 and 2 (CASQ1 and 2) and the anchoring proteins triadin (TRDN) and junctin. Together, they make up the Ca2+-release complex. CASQ1 and 2 buffer intra-SR Ca2+ stores in skeletal muscle and cardiac muscle respectively (Fujii et al. 1990, Kim et al. 2007). When Ca2+ concentrations reach 1mM, CASQs polymerise (Kim et al. 2007) and can attach to one end of RYRs, mediated by anchoring proteins TRDN and junctin (Taske et al. 1995). By sequestering Ca2+ ions, CASQs can inhibit RYRs function (Beard et al. 2004, Beard et al. 2009a, Beard et al. 2009b).

A member of the intracellular Cl- channel protein family, CLIC2, has also been determined to inhibit RYR-mediated Ca2+ transport (Board et al. 2004), potentially playing a role in the homeostasis of Ca2+ release from intracellular stores. Inhibition is thought to be via reducing activation of the channels by their primary endogenous cytoplasmic ligands, ATP and Ca2+ (Dulhunty et al. 2005). Protein kinase A (PKA) phosphorylation of RYR2 dissociates FKBP1B and results in defective channel function (Marx et al. 2000). The penta-EF hand protein sorcin (SRI) can modulate Ca2+-induced calcium-release in the heart via the interaction with several Ca2+ channels such as RYRs. A natural ligand, F112L, impairs this modulating activity (Franceschini et al. 2008). Calmodulin (CALM1) is considered a gatekeeper of RYR2. CALM1 acts directly by binding to RYR2 at residues 3583–3603, inhibiting RYR2 both at physiological and higher, pathological Ca2+ concentrations (Smith et al. 1989, Ono et al. 2010).
R-HSA-2872444 (Reactome) The sodium/hydrogen exchanger 9B1 (SLC9B1 aka Na+/H+ exchanger like domain containing 1, NHEDC1) is specifically expressed on the plasma membrane of the testis and may be implicated in infertility (Ye et al. 2006). Sodium/hydrogen exchanger 9C2 (SLC9C2), also localized to the plasma membrane, may be involved in pH regulation although this protein has not been fully characterized.
R-HSA-2872463 (Reactome) The sperm-specific Na+/H+ exchanger SLC9C1 (aka sodium/hydrogen exchanger 10, NHE10) is localized to the flagellar membrane and is involved in pH regulation of spermatozoa required for sperm motility and fertility. The activity of human SLC9C1 is inferred from experiments using mouse Slc9c1 (Wang et al. 2003).
R-HSA-2872497 (Reactome) Human serum urate levels are largely maintained by its reabsorption and secretion in the kidney. Loss of this maintenance can elevate urate levels leading to gout, hypertension, and various cardiovascular diseases. Renal urate reabsorption is controlled via two proximal tubular urate transporters; apical SLC22A12 (URAT1) and basolateral SLC2A9 (URATv1, GLUT9). On the other hand, urate secretion is mediated by the orphan sodium phosphate transporter 4 isoform 2 (SLC17A3, NPT4 isoform 2). It is mainly expressed at the apical side of renal tubules and functions as a voltage-driven urate transporter (Jutabha et al. 2010).

Genetic variations in SLC17A3 influence the variance in serum uric acid concentrations and define the serum uric acid concentration quantitative trait locus 4 (UAQTL4; MIM:612671). Excess serum urate (hyperuricemia) can lead to the development of gout, characterized by tissue deposition of monosodium urate crystals.
R-HSA-2872498 (Reactome) The microsomal Na+/(PO4)3- transporter isoform 1 (SLC17A3, NPT4 isoform 1) is a member of the anion-cation symporter family. It is expressed in liver, kidney and intestine and may function as a cotransporter of sodium (Na+) and phosphate ((PO4)3- or Pi) across the ER membrane (Melis et al. 2004).
R-HSA-2889070 (Reactome) Mitochondrial sodium/hydrogen exchanger 9B2 (SLC9B2, aka NHA2) is ubiquitously expressed and mediates the electrogenic exchange of 1 Na+ (or 1 Li+) for 2 H+ across the inner mitochondrial membrane (Xiang et al. 2007, Taglicht et al. 1993). This transport is thought to play a role in salt homeostasis and pH regulation in humans.
R-HSA-3295579 (Reactome) All functionally characterized TRP channels are permeable to the divalent cation Ca2+ except TRPM4 and 5 (Latorre et al. 2009, Wu et al. 2010).
R-HSA-3295580 (Reactome) Most members of the TRP channel family are permeable to Ca2+. Although TRPM4 and 5 can be activated by increased levels of intracellular Ca2+, they are impermeable to it. Instead, they are monovalent-selective with highest affinity for Na+ transport, of which leads to depolarization of the membrane (Launay et al. 2002, Prawitt et al. 2003). They play a central role in cardiomyocytes, neurons, endocrine pancreas cells, kidney epithelial cells and cochlea hair cells.
R-HSA-418309 (Reactome) The plasma membrane Ca-ATPases 1-4 (ATP2B1-4, PMCAs) are P-type Ca2+-ATPases regulated by calmodulin. The PMCA also counter-transports a proton. PMCA is important for Ca2+ homeostasis and function.
R-HSA-418365 (Reactome) Intracellular pools of Ca2+ serve as the source for inositol 1,4,5-trisphosphate (IP3) -induced alterations in cytoplasmic free Ca2+. In most human cells Ca2+ is stored in the lumen of the sarco/endoplastic reticulum by ATPases known as SERCAs (ATP2As). In platelets, ATP2As transport Ca2+ into the platelet dense tubular network. ATP2As are P-type ATPases, similar to the plasma membrane Na+ and Ca+-ATPases. Humans have three genes for SERCA pumps; ATP2A1-3. Studies on ATP2A1 suggest that it binds two Ca2+ ions from the cytoplasm and is subsequently phosphorylated at Asp351 before translocating Ca2+ into the SR lumen. There is a counter transport of two or possibly three protons ensuring partial charge balancing. Sarcolipin (SLN) can reversibly inhibit the activity of ATP2A1 by decreasing the apparent affinity of the ATPase for Ca2+ (Gorski et al. 2013) whereas activated Ca2+/CaM-dependent protein kinase II (CAMK2) and sorcin (SRI) can both stimulate ATP2A1-3 activity (Toyofuku et al. 1994, Matsumoto et al. 2005).
R-HSA-427910 (Reactome) Intracellular pools of Ca2+ serve as the source for inositol 1,4,5-trisphosphate (IP3) -induced alterations in cytoplasmic free Ca2+. In most human cells Ca2+ is stored in the lumen of the sarco/endoplastic reticulum by ATPases known as SERCAs (ATP2As). In platelets, ATP2As transport Ca2+ into the platelet dense tubular network. ATP2As are P-type ATPases, similar to the plasma membrane Na+ and Ca+-ATPases. Humans have three genes for SERCA pumps; ATP2A1-3. Studies on ATP2A1 suggest that it binds two Ca2+ ions from the cytoplasm and is subsequently phosphorylated at Asp351 before translocating Ca2+ into the SR lumen. There is a counter transport of two or possibly three protons ensuring partial charge balancing. Sarcolipin (SLN) can reversibly inhibit the activity of ATP2A1 by decreasing the apparent affinity of the ATPase for Ca2+ (Gorski et al. 2013) whereas activated Ca2+/CaM-dependent protein kinase II (CAMK2) and sorcin (SRI) can both stimulate ATP2A1-3 activity (Toyofuku et al. 1994, Matsumoto et al. 2005).
R-HSA-5251989 (Reactome) Probable cation-transporting ATPases 13A4 and 13A5 (ATP13A4, 5) belong to the P5 subfamily of P-tpre ATPases. This subfamily is the most poorly understood in terms of chracterisation and function. The yeast orthologue YPK9 (aka Yor291w) shows toxicity when deleted from yeast cells, suggesting a role in sequestration of divalent heavy metal ions (Schmidt et al. 2009). The ATP13A4 gene may be disrupted by a 3q25-q29 inversion of the long arm of chromosome 3, which may result in a specific language impairment (SLI) (Kwasnicka-Crawford et al. 2005).
R-HSA-5252133 (Reactome) Vacuolar-type H+-ATPases (V-ATPases) are proton pumps that acidify intracellular cargos and deliver protons across the plasma membrane of many specialised cells. V-type proton ATPase subunit S1 (ATP6AP1) is thought to function as an accessory subunit of the V0 subcomplex of V-ATPase, facilitating acidification (Supek et al. 1994). Experiments with the mouse orthologue reveals a role for Atp6ap1 in osteoclast formation and function (Qin et al. 2011).
R-HSA-5333671 (Reactome) Calcium-activated chloride channel regulators 1,2,3P and 4 (CLCA1,2,3P and 4), originally named as CLCAs based on the observation that overexpression of them all induced chloride current in response to cytosolic calcium flux. More recent evidence points to them being secreted proteins (Gibson et al. 2005) and being responsible for chloride channel modulation rather that forming chloride channels per se (Hamann et al. 2009). CLCA1 has been found to be overexpressed in aiways of patients suffering from asthma and chronic obstructive pulmonary disease (Hoshino et al. 2002, Toda et al. 2002, Kamada et al. 2004). All CLCAs contain a consensus cleavage motif which is recognised by an internal zincin metalloprotease domain within the N terminus. Self-proteolysis within the secretory pathway yields N- and C-terminal fragments, a step critical for CLCA activation of calcium-activated chloride channels (CaCCs) mediated through the N-terminal fragment (Yurtsever et al. 2012).
R-HSA-5334788 (Reactome) A potential regulator of human copper metabolism has recently been described. Copper homeostasis protein cutC homolog (CutC) is a highly conserved cytosolic copper binding protein (Li et al. 2005, Li et al. 2010). Its function in human copper metabolism remains unclear but in C. elegans, the orthologous cutc1 protein down-regulation increases sensitivity to high levels of copper (Calafato et al. 2008).
R-HSA-5692462 (Reactome) Manganese-transporting ATPase 13A1 (ATP13A1) mediates the transport of manganese (Mn2+) into the endoplasmic reticulum. ATPase activity is required for cellular manganese homeostasis (Cohen et al. 2013).
R-HSA-5692480 (Reactome) The probable cation-transporting ATPase 13A2 (ATP13A2) is thought to play a role in intracellular cation homeostasis and the maintenance of neuronal integrity (Ramonet et al. 2012). Defects in ATP13A2 can cause Kufor-Rakeb syndrome (KRS; MIM:606693), a juvenile-onset Levodopa-responsive parkinsonism (Lai et al. 2012).
R-HSA-8863494 (Reactome) Stomatin (STOM) and Stomatin-like protein 3 (STOML3) are accessory proteins for the ASIC channels (Zeng et al. 2014). STOML3 and stomatin are expressed by primary sensory neurons of the dorsal root ganglia (DRG) (Mannsfeldt et al. 1999, Wetzel et al. 2007) and regulate mechanoreceptor sensitivity in mice (Wetzel et al. 2007, Martinez-Saldago et al. 2007). STOML3 and to a lesser extent STOM can modulate the gating of ASICs (Price et al. 2004, Wetzel et al. 2007). STOML3 can bind ASIC1a, 1b, 2a, 2b, 3 and 4 (Lapatsina et al. 2012) . The function of STOML3 may be to prime the transduction complex for insertion into the plasma membrane (Lapatsina et al. 2012). Alternatively, STOML3 may control membrane mechanics by binding cholesterol to tune the sensitivity of mechano-gated channels(Qi et al. 2015).
R-HSA-936802 (Reactome) The human gene ATP7A (MNK) encodes the copper-transporting ATPase 1 (ATP7A, ATPase1, Menkes protein) which is expressed in most tissues except the liver (Vulpe et al, 1993; Chelly et al, 1993). Normally, ATP7A resides on the trans-Golgi membrane (Dierick et al, 1997). When cells are exposed to excessive copper levels, it is rapidly relocalized to the plasma membrane where it functions in copper efflux (Petris and Mercer, 1999). Defects in ATP7A are the cause of Menkes disease (MNKD), an X-linked recessive disorder of copper metabolism characterized by generalized copper deficiency (Ambrosini and Mercer, 1999).
R-HSA-936883 (Reactome) Accumulation of calcium into the Golgi apparatus is mediated by sarco(endo)plasmic reticulum calcium-ATPases (SERCAs) and by secretory pathway calcium-ATPases (SPCAs). There are two human genes which encode SPCAs; ATP2C1 and ATP2C2 which encode magnesium-dependent calcium-transporting ATPase type 2C members 1 and 2 (ATP2C1 and 2) respectively (Sudbrak et al, 2000; Vanoevelen et al, 2005). Defects in ATP2C1 are the cause of Hailey-Hailey disease (HHD), an autosomal dominant disease characterized by persistent blisters and erosions of the skin (Hu et al, 2000).
R-HSA-936895 (Reactome) The human gene ATP7B encodes the copper-transporting ATPase 2 (ATP7B, ATPase2, Wilson's protein) which is expressed mainly in the liver, brain and kidneys (Bull et al, 1993). ATP7B resides on the trans-Golgi membrane where it it thought to sequester copper from the cytosol into the golgi (Yang et al, 1997). Defects in ATP7B are the cause of Wilson disease (WD), an autosomal recessive disorder of copper metabolism characterized by the toxic accumulation of copper in a number of organs, particularly the liver and brain (Thomas et al, 1995).
R-HSA-936897 (Reactome) The sodium/potassium-transporting ATPase (ATP1A:ATP1B:FXYD) is composed of three subunits - alpha (catalytic part), beta and gamma. The trimer catalyzes the hydrolysis of ATP coupled with the exchange of sodium and potassium ions across the plasma membrane, creating the electrochemical gradient which provides energy for the active transport of various nutrients.
Four human genes encode the catalytic alpha subunits, ATP1A1-4 (Kawakami et al, 1986; Shull et al, 1989; Ovchinnikov et al, 1988; Keryanov and Gardner, 2002). Defects in ATP1A2 cause alternating hemiplegia of childhood (AHC) (Swoboda et al, 2004). Another defect in ATP1A2 causes familial hemiplegic migraine type 2 (FHM2) (Vanmolkot et al, 2003). Defects in ATP1A3 are the cause of dystonia type 12 (DYT12) (de Carvalho Aguiar et al, 2004).

Three human genes encode the non-catalytic beta subunits, ATP1B1-3. The beta subunits are thought to mediate the number of sodium pumps transported to the plasma membrane (Lane et al, 1989; Ruiz et al, 1996; Malik et al, 1996). FXYD proteins belong to a family of small membrane proteins that are auxiliary gamma subunits of Na-K-ATPase. At least six members of this family, FYD1-4, 6 and 7, have been shown to regulate Na-K-ATPase activity (Geering 2006, Choudhury et al. 2007). Defects in FXYD2 are the cause of hypomagnesemia type 2 (HOMG2) (Meij et al, 2000). ATP1A1-4 and ATP1B1-4 play a minor role during phase 2, since they begin to restore ion concentrations. The high concentration of intracellular calcium starts contraction of those cells, which is sustained in the plateau phase.
R-HSA-937311 (Reactome) The potassium-transporting ATPase heterodimer (ATP4A/12A:ATP4B) catalyzes the hydrolysis of ATP coupled with the exchange of H+ and K+ ions across the plasma membrane. It is composed of alpha and beta chains. Two human genes encode the catalytic alpha subunit, ATP4A and ATP12A (Maeda et al, 1990; Grishin et al, 1994). ATP4A is responsible for acid production in the stomach. ATP12A is responsible for potassium absorption in various tissues. One human gene encodes the beta subunit, ATP4B (Ma et al, 1991).
R-HSA-939763 (Reactome) The plasma membrane contains a broad range of lipids making up the bilayer. Aminophospholipids such as phosphatidylserine (PS) and phosphatidylethanolamine (PE) are distributed in this bilayer and their arrangement is mediated by the P-type ATPases type IV family (Paulusma and Oude Elferink, 2005).
R-HSA-947591 (Reactome) The plasma membrane contains a broad range of lipids making up the bilayer. Aminophospholipids (APLs) such as phosphatidylserine (PS) and phosphatidylethanolamine (PE) are distributed in this bilayer and their arrangement is mediated by the P-type ATPases type IV family (Paulusma and Oude Elferink, 2005).
R-HSA-9650165 (Reactome) Acid-sensing ion channels 1, 2, 3 and 5 (ASIC1, 2, 3 and 5, aka amiloride-sensitive cation channels) are homotrimeric, multi-pass membrane proteins which can transport sodium (Na+) when activated by extracellular protons. Members of the ASIC family are sensitive to amiloride and function in neurotransmission. The encoded proteins function in learning, pain transduction, touch sensation, and development of memory and fear. Many neuronal diseases cause acidosis, accompanied by pain and neuronal damage; ASICs can mediate the pathophysiological effects seen in acidiosis (Wang & Xu 2011, Qadri et al. 2012). The diuretic drug amiloride inhibits these channels, resulting in analgesic effects. NSAIDs (nonsteroidal anti-inflammatory drugs) can also inhibit ASICs to produce analgesia (Voilley et al. 2001). ASICs are also partially permeable to Ca2+, Li+ and K+ (not shown here). ASIC1 and 2 are expressed mostly in brain (Garcia-Anoveros et al. 1997, Price et al. 1996), ASIC3 is strongly expressed in testis (de Weille et al. 1998, Ishibashi & Marumo 1998) and ASIC5 is found mainly in intestine (Schaefer et al. 2000). ASIC4 subunits do not form functional channels and it's activity is unknown. It could play a part in regulating other ASIC activity (Donier et al. 2008).
R-HSA-9659568 (Reactome) Calcium-activated chloride channels (CaCCs) are ubiquitously expressed and implicated in physiological processes such as sensory transduction, fertilization, epithelial secretion, and smooth muscle contraction. The anoctamin family of transmembrane proteins (ANO, TMEM16) belong to CaCCs and have been shown to transport Cl- ions when activated by intracellular Ca2+ (Galietta 2009, Huang et al. 2012). ANO1 and 2 possess Ca2+ activated Cl- transport activity (Yang et al. 2008, Scudieri et al. 2012) while the remaining members also show some demonstrable activity (Tian et al. 2012).
RAF1:SGK:TSC22D3:WPPmim-catalysisR-HSA-2682349 (Reactome)
RYR

tetramer:FKBP1B tetramer:CASQ

polymer:TRDN:junctin
mim-catalysisR-HSA-2855020 (Reactome)
SCNN channelsR-HSA-2682349 (Reactome)
SCNN channelsmim-catalysisR-HSA-2672334 (Reactome)
SLC17A3(1-498)mim-catalysisR-HSA-2872497 (Reactome)
SLC17A3mim-catalysisR-HSA-2872498 (Reactome)
SLC9B1/C2mim-catalysisR-HSA-2872444 (Reactome)
SLC9B2mim-catalysisR-HSA-2889070 (Reactome)
SLC9C1mim-catalysisR-HSA-2872463 (Reactome)
SLNTBarR-HSA-427910 (Reactome)
SRIArrowR-HSA-2855020 (Reactome)
SRIArrowR-HSA-427910 (Reactome)
STOML3, (STOM)R-HSA-8863494 (Reactome)
TPCN1/2mim-catalysisR-HSA-2685505 (Reactome)
TRPM4,5mim-catalysisR-HSA-3295580 (Reactome)
TRPsmim-catalysisR-HSA-3295579 (Reactome)
TTYH1-3mim-catalysisR-HSA-2744349 (Reactome)
TTYH2/3mim-catalysisR-HSA-2744242 (Reactome)
Ub-SCNN channelsArrowR-HSA-2682349 (Reactome)
UbR-HSA-2682349 (Reactome)
V-ATPase:ATP6AP1ArrowR-HSA-5252133 (Reactome)
V-ATPaseR-HSA-5252133 (Reactome)
WNKsArrowR-HSA-2682349 (Reactome)
amilorideTBarR-HSA-2671885 (Reactome)
amilorideTBarR-HSA-2672334 (Reactome)
cationArrowR-HSA-5692480 (Reactome)
cationR-HSA-5692480 (Reactome)
divalent metal cationArrowR-HSA-5251989 (Reactome)
divalent metal cationR-HSA-5251989 (Reactome)
p-S-RIPK1:p-S199,227-RIPK3:p-T357,S358-MLKL oligomerArrowR-HSA-3295579 (Reactome)
p-S16-PLN pentamerArrowR-HSA-427910 (Reactome)
urateArrowR-HSA-2872497 (Reactome)
urateR-HSA-2872497 (Reactome)
Personal tools