KCNB1: Difference between revisions

Jump to navigation Jump to search
m (Robot: Automated text replacement (-{{WikiDoc Cardiology Network Infobox}} +, -<references /> +{{reflist|2}}, -{{reflist}} +{{reflist|2}}))
 
m (1 revision imported)
 
(2 intermediate revisions by 2 users not shown)
Line 1: Line 1:
<!-- The PBB_Controls template provides controls for Protein Box Bot, please see Template:PBB_Controls for details. -->
{{Infobox_gene}}
{{PBB_Controls
<!-- Deleted image removed: [[File:4jta.1 1 chimera colorbysym 1000 1000.png|thumb|Transmembrane view of Kv2.1 channel]] -->
| update_page = yes
<!-- Deleted image removed: [[File:4jta.1 0 chimera colorbysym 1000 1000.png|thumb|Top view of open Kv2.1 channel with four subunit selectivity pore conducting potassium current]] -->
| require_manual_inspection = no
'''Potassium voltage-gated channel, Shab-related subfamily, member 1''', also known as '''KCNB1''' or '''K<sub>v</sub>2.1''', is a [[protein]] that, in humans, is encoded by the ''KCNB1'' [[gene]].<ref name="entrez">{{cite web | title = Entrez Gene: KCNB1 potassium voltage-gated channel, Shab-related subfamily, member 1| url = https://www.ncbi.nlm.nih.gov/sites/entrez?Db=gene&Cmd=ShowDetailView&TermToSearch=3745| accessdate = }}</ref><ref name="Melis_1995">{{cite journal | vauthors = Melis R, Stauffer D, Zhao X, Zhu XL, Albrecht B, Pongs O, Brothman A, Leppert M | title = Physical and genetic localization of a Shab subfamily potassium channel (KCNB1) gene to chromosomal region 20q13.2 | journal = Genomics | volume = 25 | issue = 1 | pages = 285–7 | date = January 1995 | pmid = 7774931 | doi = 10.1016/0888-7543(95)80138-C }}</ref><ref name="Gutman_2005">{{cite journal | vauthors = Gutman GA, Chandy KG, Grissmer S, Lazdunski M, McKinnon D, Pardo LA, Robertson GA, Rudy B, Sanguinetti MC, Stühmer W, Wang X | title = International Union of Pharmacology. LIII. Nomenclature and molecular relationships of voltage-gated potassium channels | journal = Pharmacological Reviews | volume = 57 | issue = 4 | pages = 473–508 | date = December 2005 | pmid = 16382104 | doi = 10.1124/pr.57.4.10 }}</ref>
| update_protein_box = yes
| update_summary = yes
| update_citations = yes
}}


<!-- The GNF_Protein_box is automatically maintained by Protein Box Bot.  See Template:PBB_Controls to Stop updates. -->
Potassium voltage-gated channel subfamily B member one, or simply known as KCNB1, is a delayed rectifier and [[voltage-gated potassium channel]] found throughout the body. The channel has a diverse number of functions. However, its main function, as a delayed rectifier, is to propagate current in its respective location. It is commonly expressed in the [[central nervous system]], but may also be found in [[Pulmonary artery|pulmonary arteries]], auditory outer hair cells, [[Stem cell|stem cells]], the [[retina]], and organs such as the [[heart]] and [[pancreas]]. Modulation of K+ channel activity and expression has been found to be at the crux of many profound pathophysiological disorders in several cell types.<ref name="Shah_2017" />
{{GNF_Protein_box
| image = 
| image_source = 
| PDB =
| Name = Potassium voltage-gated channel, Shab-related subfamily, member 1
| HGNCid = 6231
| Symbol = KCNB1
| AltSymbols =; DRK1; KV2.1; h-DRK1
| OMIM = 600397
| ECnumber = 
| Homologene = 37988
| MGIid = 96666
| GeneAtlas_image1 = PBB_GE_KCNB1_211006_s_at_tn.png
| Function = {{GNF_GO|id=GO:0005251 |text = delayed rectifier potassium channel activity}} {{GNF_GO|id=GO:0005515 |text = protein binding}} {{GNF_GO|id=GO:0030955 |text = potassium ion binding}}
| Component = {{GNF_GO|id=GO:0005624 |text = membrane fraction}} {{GNF_GO|id=GO:0008076 |text = voltage-gated potassium channel complex}} {{GNF_GO|id=GO:0016020 |text = membrane}}
| Process = {{GNF_GO|id=GO:0006811 |text = ion transport}} {{GNF_GO|id=GO:0006813 |text = potassium ion transport}}
| Orthologs = {{GNF_Ortholog_box
    | Hs_EntrezGene = 3745
    | Hs_Ensembl = ENSG00000158445
    | Hs_RefseqProtein = NP_004966
    | Hs_RefseqmRNA = NM_004975
    | Hs_GenLoc_db = 
    | Hs_GenLoc_chr = 20
    | Hs_GenLoc_start = 47418353
    | Hs_GenLoc_end = 47532591
    | Hs_Uniprot = Q14721
    | Mm_EntrezGene = 16500
    | Mm_Ensembl = ENSMUSG00000050556
    | Mm_RefseqmRNA = NM_008420
    | Mm_RefseqProtein = NP_032446
    | Mm_GenLoc_db = 
    | Mm_GenLoc_chr = 2
    | Mm_GenLoc_start = 166794583
    | Mm_GenLoc_end = 166880004
    | Mm_Uniprot = Q0N2S5
  }}
}}
'''Potassium voltage-gated channel, Shab-related subfamily, member 1''', also known as '''KCNB1''' or '''K<sub>v</sub>2.1''', is a human [[gene]].<ref name="entrez">{{cite web | title = Entrez Gene: KCNB1 potassium voltage-gated channel, Shab-related subfamily, member 1| url = http://www.ncbi.nlm.nih.gov/sites/entrez?Db=gene&Cmd=ShowDetailView&TermToSearch=3745| accessdate = }}</ref>


<!-- The PBB_Summary template is automatically maintained by Protein Box Bot.  See Template:PBB_Controls to Stop updates. -->
Potassium channels are among the most diverse of all ion channels in eukaryotes. With over 100 genes coding numerous functions, many [[Protein isoform|isoforms]] of potassium channels are present in the body, but most are divided up into two main groups: inactivating transient channels and non-inactivating delayed rectifiers. Due to the multiple varied forms, potassium delayed rectifier channels open or close in response to a myriad of signals. These include: cell [[depolarization]] or [[Hyperpolarization (biology)|hyperpolarization]], increases in intracellular calcium concentrations, neurotransmitter binding, or second messenger activity such as [[G protein|G-proteins]] or [[kinase]]s.<ref name=Interpro>{{cite web | url = https://www.ebi.ac.uk/interpro/entry/IPR005400 | title = Potassium channel, voltage-dependent, beta subunit, KCNAB1 (IPR005400) | publisher = EMBL-EBI | work  = InterPro | access-date = 2017-04-04 }}</ref>
{{PBB_Summary
| section_title =
| summary_text = Voltage-gated potassium (Kv) channels represent the most complex class of voltage-gated ion channels from both functional and structural standpoints. Their diverse functions include regulating neurotransmitter release, heart rate, insulin secretion, neuronal excitability, epithelial electrolyte transport, smooth muscle contraction, and cell volume. Four sequence-related potassium channel genes - shaker, shaw, shab, and shal - have been identified in Drosophila, and each has been shown to have human homolog(s). This gene encodes a member of the potassium channel, voltage-gated, shab-related subfamily. This member is a delayed rectifier potassium channel and its activity is modulated by some other family members.<ref name="entrez">{{cite web | title = Entrez Gene: KCNB1 potassium voltage-gated channel, Shab-related subfamily, member 1| url = http://www.ncbi.nlm.nih.gov/sites/entrez?Db=gene&Cmd=ShowDetailView&TermToSearch=3745| accessdate = }}</ref>
}}


==See also==
== Structure ==
The general structure of all potassium channels contain a centered pore composed of alpha subunits with a pore loop expressed by a segment of conserved [[DNA]], T/SxxTxGxG. This general sequence comprises the selectivity of the potassium channel. Depending on the channel, the alpha subunits are constructed in either a homo- or hetero-association, creating a 4-subunit selectivity pore or a 2-subunit pore, each with accessory beta subunits attached intracellularly. Also on the cytoplasmic side are the N- and C- termini, which play a crucial role in activating and deactivating KCNB1 channels. This pore creates the main opening of the channel where potassium ions flow through.<ref name="Wray_2004">{{cite journal | vauthors = Wray D | title = The roles of intracellular regions in the activation of voltage-dependent potassium channels| journal = European Biophysics Journal | volume = 33 | issue = 3 | pages = 194–200 | date = May 2004 | pmid = 14608450 | doi = 10.1007/s00249-003-0363-2 }}</ref>
 
The type of pore domain (number of subunits) determines if the channel has the typical 6 [[Transmembrane protein|transmembrane]] (protein) spanning regions, or the less dominant [[Inward-rectifier potassium ion channel|inward rectifier]] type of only 2 regions. KCNB1 has 6TM labeled S1-S6, each with a tetrameric structure. S5 and S6 create the p-loop, while S4 is the location of the voltage sensor. S4, along with S2 and S3 create the ‘activating’ portions of the delayed rectifier channel.<ref name="Wray_2004"/> The heteromeric complexes that contain the distinct pore are electrically inactive or non-conducting, but unlike other potassium families, the pore of the KCNB1 group has numerous phosphorylation sites allowing kinase activity. Maturing KCNB1 channels develop these phosphorylation sites within the channel pore, but lack a glycosylation stage in the N-terminus.<ref name="Patel_2016" />
 
Specifically, the KCNB1 delayed rectifier channel conducts a potassium current (K+). This mediates high frequency firing due to the [[phosphorylation]] sites located within the channel via kinases and a major calcium influx typical of all neurons.<ref name="Patel_2016">{{cite journal | vauthors = Patel R, Sesti F | title = Oxidation of ion channels in the aging nervous system | journal = Brain Research | volume = 1639 | pages = 174–85 | date = May 2016 | pmid = 26947620 | doi = 10.1016/j.brainres.2016.02.046 }}</ref>
 
=== Kinetics ===
The kinetics surrounding the activation and deactivation of the KCNB1 channel is relatively unknown, and has been under considerable study. Three of the six transmembrane regions, S2, S3 and S4, contribute to the activation phase of the channel. Upon depolarization, the S4 region, which is positively charged, is moved in response to the subsequent positive charge of the depolarization. As a result of S4 movement, the negatively charged regions of S2 and S3 appear to move as well.<ref name="Wray_2004"/> The movement of these regions causes an opening of the channel gate within regions of S5 and S6.<ref name="Wray_2009">{{cite journal | vauthors = Wray D | title = Intracellular regions of potassium channels: Kv2.1 and heag| journal = European Biophysics Journal | volume = 38 | issue = 3 | pages = 285–92 | date = March 2009 | pmid = 18607586 | doi = 10.1007/s00249-008-0354-4 }}</ref> The intracellular regions of the C and N-terminus also play a crucial role in the activation kinetics of the channel. The two termini interact with one other, as the C-terminus folds around the N-terminus during channel activation. The relative movement between the N- and C- termini greatly aids in producing a conformational change of the channel necessary for channel opening. This interaction between these intracellular regions is believed to be linked with membrane-spanning regions of S1 and S6, and thus aid in the movement of S2, S3, and S4 in opening the channel.<ref name="Wray_2004"/><ref name="Wray_2009"/> Studies on selective mutations knocking out these intracellular termini have been shown to produce larger reductions in speed and probability of channel opening, which indicates their importance in channel activation.<ref name="Wray_2004"/>
 
== Function ==
Voltage-gated potassium ([[Voltage-gated potassium channel|K<sub>v</sub>]]) channels represent the most complex class of voltage-gated ion channels from both functional and structural standpoints.<ref name="entrez" /> Delayed rectifier potassium channels’ most prevalent role is in the falling phase of physiological [[action potential]]s. KCNB1 rectifiers are also important in forming the cardiac beat and rate synchronicity that exists within the heart, and the lysis of target molecules in the immune response. These channels  can also act as effectors in downstream signaling in [[G protein–coupled receptor|G-protein coupled receptor]] transduction. KCNB1’s regulation and propagation of current provides a means for regulatory control over several physiological functions.<ref name=Interpro/> Their diverse functions include regulating [[neurotransmitter]] release, [[heart rate]], [[insulin]] secretion, neuronal excitability, epithelial electrolyte transport, smooth muscle contraction, and [[apoptosis]].<ref name="entrez" />
 
Voltage-gated potassium channels are essential in regulating neuronal [[membrane potential]], and in contributing to action potential production and firing.<ref>{{cite journal | vauthors = Sesti F | title = Oxidation of K(+) Channels in Aging and Neurodegeneration | journal = [[Aging and Disease]] | volume = 7 | issue = 2 | pages = 130–5 | date = March 2016 | pmid = 27114846 | pmc = 4809605 | doi = 10.14336/AD.2015.0901 }}</ref> In mammalian CNS neurons, KCNB1 is a predominant delayed rectifier potassium current that regulates neuronal excitability, action potential duration, and tonic spiking. This is necessary when it comes to proper neurotransmitter release, as such release is dependent on membrane potential. In mouse cardiomyocytes, KCNB1 channel is the molecular substrate of major repolarization current I<sub>K-slow2</sub>. [[Genetically modified organism|Transgenic]] mice, expressing a [[dominant-negative]] isoform of KCNB1, exhibit markedly prolonged [[action potential]]s and demonstrate [[cardiac arrhythmia|arrhythmia]].<ref name="Murakoshi_1999">{{cite journal | vauthors = Murakoshi H, Trimmer JS | title = Identification of the Kv2.1 K+ channel as a major component of the delayed rectifier K+ current in rat hippocampal neurons | journal = The Journal of Neuroscience | volume = 19 | issue = 5 | pages = 1728–35 | date = March 1999 | pmid = 10024359 | url = http://www.jneurosci.org/content/jneuro/19/5/1728.full.pdf }}</ref> KCNB1 also contributes to the function and regulation of smooth muscle fibers. Human studies on pulmonary arteries have shown that normal, physiological inhibition of KCNB1 current aids [[vasoconstriction]] of arteries.<ref>{{cite journal | vauthors = Joseph BK, Thakali KM, Moore CL, Rhee SW | title = Ion channel remodeling in vascular smooth muscle during hypertension: Implications for novel therapeutic approaches | journal = Pharmacological Research | volume = 70 | issue = 1 | pages = 126–38 | date = April 2013 | pmid = 23376354 | pmc = 3607210 | doi = 10.1016/j.phrs.2013.01.008 }}</ref> In human pancreatic ß cells, KCNB1, which mediates potassium efflux, produces a downstroke of the action potential in the cell.<ref>{{cite journal | vauthors = Yang SN, Shi Y, Yang G, Li Y, Yu J, Berggren PO | title = Ionic mechanisms in pancreatic β cell signaling | journal = Cellular and Molecular Life Sciences | volume = 71 | issue = 21 | pages = 4149–77 | date = November 2014 | pmid = 25052376 | doi = 10.1007/s00018-014-1680-6 }}</ref> In effect, this behavior halts insulin secretion, as its activation decreases the Ca<sub>v</sub> channel-mediated calcium influx that is necessary for insulin exocytosis. KCNB1 has also been found to promote apoptosis within neuronal cells. It is currently believed that KCNB1-induced apoptosis occurs in response to an increase in [[reactive oxygen species]] (ROS) that results either from acute oxidation or as a consequence of other cellular stresses.<ref name="Patel_2016" />
 
== Regulation ==
KCNB1 conductance is regulated primarily by [[oligomer]]ization and [[phosphorylation]]. Additional forms of regulation include [[SUMO protein|SUMOylation]] and [[acetylation]], although the direct effect of these modifications is still under investigation. KCNB1 consensus sites in the N-terminus are not subject to [[glycosylation]].<ref name="Shah_2017" />
 
=== Phosphorylation ===
Many proteins undergo phosphorylation, or the addition of phosphate groups to [[Amino acid|amino acids]] subunits. Phosphorylation is modulated by [[Kinase|kinases]], which add phosphate groups, and [[Phosphatase|phosphatases]], which remove phosphate groups. In its phosphorylated state, KCNB1 is a poor conductor of current. There are 16 phosphorylation sites that are subject to the activity of kinases, such as [[cyclin-dependent kinase 5]] and [[AMP-activated protein kinase]]. These sites are reversibly regulated by phosphatases such as, phosphatase [[calcineurin]]. Under periods of high electrical activity, depolarization of the neuron increases calcium influx and triggers phosphatase activity. Under resting conditions, KCNB1 tends to be phosphorylated. Phosphorylation raises the threshold voltage requirement for activation and allows microdomains to bind the channel, preventing KCNB1 from entering the plasma membrane. Microdomains localize KCNB1 in dendrites in cell bodies of hippocampal and cortical neurons. Conductance associated with de-phosphorylation of this channel acts to decrease or end periods high excitability. However, this relationship is not static and is cell dependent. The role of phosphorylation can be affected by reactive oxygen species (ROS) that increase during oxidative stress. ROS act to increase the levels of zinc (Zn<sup>2+)</sup> and calcium (Ca<sup>2+)</sup> intracellularly that act with protein kinases to phosphorylate certain sites on KCNB1. This phosphorylation increases the insertion of KCNB1 into the membrane and elevates conductance. Under these conditions the interaction with [[SNARE (protein)|SNARE protein]] [[syntaxin]], is enhanced. This surge of KCNB1 current induces activation of a pro-apoptotic pathway, DNA fragmentation, and caspase activation.<ref name="Shah_2017">{{cite journal | vauthors = Shah NH, Aizenman E | title = Voltage-gated potassium channels at the crossroads of neuronal function, ischemic tolerance, and neurodegeneration | journal = Translational Stroke Research | volume = 5 | issue = 1 | pages = 38–58 | date = February 2014 | pmid = 24323720 | pmc = 3946373 | doi = 10.1007/s12975-013-0297-7 }}</ref>
 
=== Oligomerization ===
Another proposed mechanism for regulation of apoptosis is oligomerization, or the process of forming multi-protein complexes held together through [[Disulfide|disulfide bonds]]. Under oxidative stress, [[reactive oxygen species]] (ROS) form and act to regulate KCNB1 through oxidation. Increase in oxygen radicals directly causes formation of KCNB1 oligomers that then accumulate in the plasma membrane and initially decrease current flow <ref>{{cite journal |vauthors=Wu X, Hernandez-Enriquez B, Banas M, Xu R, Sesti F |title=Molecular mechanisms underlying the apoptotic effect of KCNB1 K+ channel oxidation.|journal=J Biol Chem|date=2013|volume=288|issue=6|pages=4128–4134|doi=10.1074/jbc.M112.440933|pmc=3567663}}</ref><ref>{{cite journal |vauthors=Cotella D, Hernandez B, Wu X, Li R, Pan Z, Leveille J, Link CD, Oddo S, Sesti F |title=Toxic role of K+ channel oxidation in mammalian brain|journal=J. Neurosci.|date=2012|volume=32|issue=12|pages=4133–4144|doi=10.1523/JNEUROSCI.6153-11.2012}}</ref>.  Oligomer activation of c-Src and JNK kinases induces the initial pro-apoptotic signal, which is coupled to KCNB1 current. This further promotes the apoptosis pathway <ref>{{cite journal |vauthors=Yu W, Gowda M, Singh S, Sesti F |title=Oxidation of KCNB1 potassium channels triggers apoptotic integrin signaling in the brain.|journal=Cell Death Dis.|date=2017|volume=8|issue=4|page=e2737|doi=10.1038/cddis.2017.160}}</ref>. KCNB1 oligomers have been detected in the post mortem human hippocampus <ref>{{cite journal |vauthors=Wei Y, Shih R, Sesti F |title=Oxidation of KCNB1 channels in the human brain and in mouse model of Alzheimer's disease |journal=Cell Death Dis. |date=2018 |volume=9 |issue=820 |doi=10.1038/s41419-018-0886-1}}</ref>
 
== Blockers ==
Potassium delayed rectifiers have been implicated in many pharmacological uses in the investigation of biological toxins for drug development. A main component to many of the toxins with negative effects on delayed rectifiers contain [[cystine]] inhibitors that are arranged around [[Disulfide|disulfide bond]] formations. Many of these toxins originate from species of tarantulas. ''G. spatulata'' produces the [[hanatoxin]], which was the first drug to be manipulated to interact with KCNB1 receptors by inhibiting the activation of most potassium voltage-gated channels. Other toxins, such as [[stromatoxin]], heteroscordratoxin, and [[guangxitoxin]], target the selectivity of voltage KCNB1 rectifiers, by either lowering potassium binding affinity or increasing the binding rate of potassium. This can lead to [[excitotoxicity]], or overstimulation of postsynaptic neurons. In nature, the prey of tarantula that are injected with these endogenous toxins induces this excitotoxic effect, producing paralysis for easy capture. Physiologically, these venoms work on KCNB1 rectifier affinity by altering the channels’ voltage sensor, making it more or less sensitive to extracellular potassium concentrations.<ref>{{cite journal | vauthors = Swartz KJ | title = Tarantula toxins interacting with voltage sensors in potassium channels | journal = Toxicon | volume = 49 | issue = 2 | pages = 213–30 | date = February 2007 | pmid = 17097703 | pmc = 1839852 | doi = 10.1016/j.toxicon.2006.09.024 }}</ref> KCNB1 is also susceptible to [[tetraethylammonium]] (TEA) and [[4-Aminopyridine|4-aminopyridine]] (4-AP), which completely block all channel activity. TEA also works on calcium-activated potassium channels, furthering its inhibitory effects on neurons and skeletal muscle. Some isoforms of TEA are beneficial for patients with severe [[Alzheimer's disease|Alzheimer’s]], as blocking KCNB1 channels reduces the amount of neuronal apoptosis, thereby slowing the rate of dementia.<ref>{{cite journal | vauthors = Quinn CC, Begenisich T | title = Pharmacology and surface electrostatics of the K channel outer pore vestibule | journal = The Journal of Membrane Biology | volume = 212 | issue = 1 | pages = 51–60 | date = 2017-04-12 | pmid = 17206516 | pmc = 1784061 | doi = 10.1007/s00232-006-0039-9 }}</ref> This has been attributed to the oxidative properties of the channel by ROS.<ref name="Interpro" />
 
== Physiological Role in Disease ==
 
=== Neurodegenerative Disease ===
Oxidative damage is widely considered to play a role in neurodegenerative disorders, including [[Alzheimers disease]]. Such oxidative stress alters the redox sensitivity of the Kv2.1 delayed rectifier, resulting in the modulation of the channel.<ref name="Shah_2017">{{cite journal | vauthors = Shah NH, Aizenman E | title = Voltage-gated potassium channels at the crossroads of neuronal function, ischemic tolerance, and neurodegeneration | journal = Translational Stroke Research | volume = 5 | issue = 1 | pages = 38–58 | date = February 2014 | pmid = 24323720 | pmc = 3946373 | doi = 10.1007/s12975-013-0297-7 }}</ref> ''In vitro'' studies and studies in animal models show that when KCNB1 is oxidized, it no longer conducts, leading to neurons becoming hyperpolarized and dying; oxidized KCNB1 also clusters in [[Lipid raft|lipid rafts]] and cannot be internalized, which also leads to apoptosis. These alterations disrupt normal neuronal signaling and increase the likelihood of neurological diseases. Oxidized (oligomerized) KCNB1 channels are present in the hippocampi of old (Braak stage 1-2) and Alzheimer's disease (Braak stage 5) donors of either sexes <ref>{{cite journal |vauthors=Wei Y, Shih R, Sesti F |title=Oxidation of KCNB1 channels in the human brain and in mouse model of Alzheimer's disease |journal=Cell Death Dis. |date=2018 |volume=9 |issue=820 |doi=10.1038/s41419-018-0886-1}}</ref> <ref name="Peers_2014">{{cite journal | vauthors = Peers C, Boyle JP | title = Oxidative modulation of K+ channels in the central nervous system in neurodegenerative diseases and aging | journal = Antioxidants & Redox Signaling | volume = 22 | issue = 6 | pages = 505–21 | date = February 2015 | pmid = 25333910 | doi = 10.1089/ars.2014.6007 }}</ref> 
 
Increased probability of the channel remaining open can also potentially drive neurodegeneration.  [[HIV dementia|Human immunodeficiency virus type-1 (HIV-1)-associated dementia]] (HAD) may be driven by an overabundance of [[Glutamic acid|glutamate]], which in turn can trigger increased calcium levels, which in turn can drive calcium-dependent dephosphorylation of KCNB1 channels, which increases probability of channel activation and current conductance. Enhanced KCNB1 current couples cell shrinkage associated with apoptosis and dendritic beading leading to diminished [[Long-term potentiation|long term potentiation]]. These neuronal modifications may explain the atrophy of cell layer volume and late stage cell death observed in HAD disease.<ref>{{cite journal | vauthors = Keblesh J, Hu D, Xiong H | title = Voltage-gated potassium channels in human immunodeficiency virus type-1 (HIV-1)-associated neurocognitive disorders | journal = Journal of Neuroimmune Pharmacology | volume = 4 | issue = 1 | pages = 60–70 | date = March 2009 | pmid = 18459047 | pmc = 3974578 | doi = 10.1007/s11481-008-9106-6 }}</ref>
 
=== Cancer ===
Exploitation of this channel is advantageous in cancer cell survival as they have the ability to produce [[Heme oxygenase|heme oxygenase-1]], an enzyme with the ability to generate carbon monoxide (CO). Oncogenic cells benefit from producing CO due to the antagonizing effects of the KCNB1 channel. Inhibition of KCNB1 allows cancer [[Cell proliferation|proliferation]] without the apoptotic pathway preventing tumor formation. Although potassium channels are studied as a therapeutic target for cancer, this apoptotic regulation is dependent on cancer type, potassium channel type, expression levels, intracellular localization as well as regulation by pro- or anti-apoptotic factors. <ref>{{cite journal | vauthors = Kondratskyi A, Kondratska K, Skryma R, Prevarskaya N | title = Ion channels in the regulation of apoptosis | journal = Biochimica et Biophysica Acta | volume = 1848 | issue = 10 Pt B | pages = 2532–46 | date = October 2015 | pmid = 25450339 | doi = 10.1016/j.bbamem.2014.10.030 | series = Membrane Channels and Transporters in Cancers }}</ref>
 
== Interactions ==
 
KCNB1 has been shown to [[Protein-protein interaction|interact]] with:
* [[KCNH1]],<ref name = "Ottschytsch_2002">{{cite journal | vauthors = Ottschytsch N, Raes A, Van Hoorick D, Snyders DJ | title = Obligatory heterotetramerization of three previously uncharacterized Kv channel alpha-subunits identified in the human genome | journal = Proceedings of the National Academy of Sciences of the United States of America | volume = 99 | issue = 12 | pages = 7986–91 | date = June 2002 | pmid = 12060745 | pmc = 123007 | doi = 10.1073/pnas.122617999 }}</ref> and
* [[PTPRE]].<ref name = "Peretz_2000">{{cite journal | vauthors = Peretz A, Gil-Henn H, Sobko A, Shinder V, Attali B, Elson A | title = Hypomyelination and increased activity of voltage-gated K(+) channels in mice lacking protein tyrosine phosphatase epsilon | journal = The EMBO Journal | volume = 19 | issue = 15 | pages = 4036–45 | date = August 2000 | pmid = 10921884 | pmc = 306594 | doi = 10.1093/emboj/19.15.4036 }}</ref>
 
== See also ==
* [[Voltage-gated potassium channel]]
* [[Voltage-gated potassium channel]]
* [[Guangxitoxin]]


==References==
== References ==
{{reflist|2}}
{{reflist|33em}}


==Further reading==
== Further reading ==
{{refbegin | 2}}
{{refbegin|33em}}
{{PBB_Further_reading
* {{cite journal | vauthors = Albrecht B, Lorra C, Stocker M, Pongs O | title = Cloning and characterization of a human delayed rectifier potassium channel gene | journal = Receptors & Channels | volume = 1 | issue = 2 | pages = 99–110 | year = 1994 | pmid = 8081723 | doi =  }}
| citations =
* {{cite journal | vauthors = Hugnot JP, Salinas M, Lesage F, Guillemare E, de Weille J, Heurteaux C, Mattéi MG, Lazdunski M | title = Kv8.1, a new neuronal potassium channel subunit with specific inhibitory properties towards Shab and Shaw channels | journal = The EMBO Journal | volume = 15 | issue = 13 | pages = 3322–31 | date = July 1996 | pmid = 8670833 | pmc = 451895 | doi =  }}
*{{cite journal  | author=Gutman GA, Chandy KG, Grissmer S, ''et al.'' |title=International Union of Pharmacology. LIII. Nomenclature and molecular relationships of voltage-gated potassium channels. |journal=Pharmacol. Rev. |volume=57 |issue= 4 |pages= 473-508 |year= 2006 |pmid= 16382104 |doi= 10.1124/pr.57.4.10 }}
* {{cite journal | vauthors = Post MA, Kirsch GE, Brown AM | title = Kv2.1 and electrically silent Kv6.1 potassium channel subunits combine and express a novel current | journal = FEBS Letters | volume = 399 | issue = 1–2 | pages = 177–82 | date = December 1996 | pmid = 8980147 | doi = 10.1016/S0014-5793(96)01316-6 }}
*{{cite journal  | author=Melis R, Stauffer D, Zhao X, ''et al.'' |title=Physical and genetic localization of a Shab subfamily potassium channel (KCNB1) gene to chromosomal region 20q13.2. |journal=Genomics |volume=25 |issue= 1 |pages= 285-7 |year= 1995 |pmid= 7774931 |doi=  }}
* {{cite journal | vauthors = Patel AJ, Lazdunski M, Honoré E | title = Kv2.1/Kv9.3, a novel ATP-dependent delayed-rectifier K+ channel in oxygen-sensitive pulmonary artery myocytes | journal = The EMBO Journal | volume = 16 | issue = 22 | pages = 6615–25 | date = November 1997 | pmid = 9362476 | pmc = 1170266 | doi = 10.1093/emboj/16.22.6615 }}
*{{cite journal | author=Albrecht B, Lorra C, Stocker M, Pongs O |title=Cloning and characterization of a human delayed rectifier potassium channel gene. |journal=Recept. Channels |volume=1 |issue= 2 |pages= 99-110 |year= 1994 |pmid= 8081723 |doi=  }}
* {{cite journal | vauthors = Shepard AR, Rae JL | title = Electrically silent potassium channel subunits from human lens epithelium | journal = The American Journal of Physiology | volume = 277 | issue = 3 Pt 1 | pages = C412-24 | date = September 1999 | pmid = 10484328 | doi =  }}
*{{cite journal | author=Hugnot JP, Salinas M, Lesage F, ''et al.'' |title=Kv8.1, a new neuronal potassium channel subunit with specific inhibitory properties towards Shab and Shaw channels. |journal=EMBO J. |volume=15 |issue= 13 |pages= 3322-31 |year= 1996 |pmid= 8670833 |doi=  }}
* {{cite journal | vauthors = Zhu XR, Netzer R, Böhlke K, Liu Q, Pongs O | title = Structural and functional characterization of Kv6.2 a new gamma-subunit of voltage-gated potassium channel | journal = Receptors & Channels | volume = 6 | issue = 5 | pages = 337–50 | year = 1999 | pmid = 10551266 | doi =  }}
*{{cite journal | author=Post MA, Kirsch GE, Brown AM |title=Kv2.1 and electrically silent Kv6.1 potassium channel subunits combine and express a novel current. |journal=FEBS Lett. |volume=399 |issue= 1-2 |pages= 177-82 |year= 1997 |pmid= 8980147 |doi= }}
* {{cite journal | vauthors = Sano Y, Mochizuki S, Miyake A, Kitada C, Inamura K, Yokoi H, Nozawa K, Matsushime H, Furuichi K | title = Molecular cloning and characterization of Kv6.3, a novel modulatory subunit for voltage-gated K(+) channel Kv2.1 | journal = FEBS Letters | volume = 512 | issue = 1–3 | pages = 230–4 | date = February 2002 | pmid = 11852086 | doi = 10.1016/S0014-5793(02)02267-6 }}
*{{cite journal | author=Patel AJ, Lazdunski M, Honoré E |title=Kv2.1/Kv9.3, a novel ATP-dependent delayed-rectifier K+ channel in oxygen-sensitive pulmonary artery myocytes. |journal=EMBO J. |volume=16 |issue= 22 |pages= 6615-25 |year= 1998 |pmid= 9362476 |doi= 10.1093/emboj/16.22.6615 }}
* {{cite journal | vauthors = Kurata HT, Soon GS, Eldstrom JR, Lu GW, Steele DF, Fedida D | title = Amino-terminal determinants of U-type inactivation of voltage-gated K+ channels | journal = The Journal of Biological Chemistry | volume = 277 | issue = 32 | pages = 29045–53 | date = August 2002 | pmid = 12021261 | doi = 10.1074/jbc.M111470200 }}
*{{cite journal | author=Shepard AR, Rae JL |title=Electrically silent potassium channel subunits from human lens epithelium. |journal=Am. J. Physiol. |volume=277 |issue= 3 Pt 1 |pages= C412-24 |year= 1999 |pmid= 10484328 |doi=  }}
* {{cite journal | vauthors = MacDonald PE, Wang G, Tsuk S, Dodo C, Kang Y, Tang L, Wheeler MB, Cattral MS, Lakey JR, Salapatek AM, Lotan I, Gaisano HY | title = Synaptosome-associated protein of 25 kilodaltons modulates Kv2.1 voltage-dependent K(+) channels in neuroendocrine islet beta-cells through an interaction with the channel N terminus | journal = Molecular Endocrinology | volume = 16 | issue = 11 | pages = 2452–61 | date = November 2002 | pmid = 12403834 | doi = 10.1210/me.2002-0058 }}
*{{cite journal | author=Zhu XR, Netzer R, Böhlke K, ''et al.'' |title=Structural and functional characterization of Kv6.2 a new gamma-subunit of voltage-gated potassium channel. |journal=Recept. Channels |volume=6 |issue= 5 |pages= 337-50 |year= 1999 |pmid= 10551266 |doi=  }}
* {{cite journal | vauthors = Ju M, Stevens L, Leadbitter E, Wray D | title = The Roles of N- and C-terminal determinants in the activation of the Kv2.1 potassium channel | journal = The Journal of Biological Chemistry | volume = 278 | issue = 15 | pages = 12769–78 | date = April 2003 | pmid = 12560340 | doi = 10.1074/jbc.M212973200 }}
*{{cite journal | author=Peretz A, Gil-Henn H, Sobko A, ''et al.'' |title=Hypomyelination and increased activity of voltage-gated K(+) channels in mice lacking protein tyrosine phosphatase epsilon. |journal=EMBO J. |volume=19 |issue= 15 |pages= 4036-45 |year= 2000 |pmid= 10921884 |doi= 10.1093/emboj/19.15.4036 }}
* {{cite journal | vauthors = Tiran Z, Peretz A, Attali B, Elson A | title = Phosphorylation-dependent regulation of Kv2.1 Channel activity at tyrosine 124 by Src and by protein-tyrosine phosphatase epsilon | journal = The Journal of Biological Chemistry | volume = 278 | issue = 19 | pages = 17509–14 | date = May 2003 | pmid = 12615930 | doi = 10.1074/jbc.M212766200 }}
*{{cite journal  | author=Deloukas P, Matthews LH, Ashurst J, ''et al.'' |title=The DNA sequence and comparative analysis of human chromosome 20. |journal=Nature |volume=414 |issue= 6866 |pages= 865-71 |year= 2002 |pmid= 11780052 |doi= 10.1038/414865a }}
* {{cite journal | vauthors = Consiglio JF, Korn SJ | title = Influence of permeant ions on voltage sensor function in the Kv2.1 potassium channel | journal = The Journal of General Physiology | volume = 123 | issue = 4 | pages = 387–400 | date = April 2004 | pmid = 15024041 | pmc = 2217458 | doi = 10.1085/jgp.200308976 }}
*{{cite journal  | author=Sano Y, Mochizuki S, Miyake A, ''et al.'' |title=Molecular cloning and characterization of Kv6.3, a novel modulatory subunit for voltage-gated K(+) channel Kv2.1. |journal=FEBS Lett. |volume=512 |issue= 1-3 |pages= 230-4 |year= 2002 |pmid= 11852086 |doi= }}
* {{cite journal | vauthors = Thébaud B, Michelakis ED, Wu XC, Moudgil R, Kuzyk M, Dyck JR, Harry G, Hashimoto K, Haromy A, Rebeyka I, Archer SL | title = Oxygen-sensitive Kv channel gene transfer confers oxygen responsiveness to preterm rabbit and remodeled human ductus arteriosus: implications for infants with patent ductus arteriosus | journal = Circulation | volume = 110 | issue = 11 | pages = 1372–9 | date = September 2004 | pmid = 15353504 | doi = 10.1161/01.CIR.0000141292.28616.65 }}
*{{cite journal | author=Kurata HT, Soon GS, Eldstrom JR, ''et al.'' |title=Amino-terminal determinants of U-type inactivation of voltage-gated K+ channels. |journal=J. Biol. Chem. |volume=277 |issue= 32 |pages= 29045-53 |year= 2002 |pmid= 12021261 |doi= 10.1074/jbc.M111470200 }}
* {{cite journal | vauthors = Kerschensteiner D, Soto F, Stocker M | title = Fluorescence measurements reveal stoichiometry of K+ channels formed by modulatory and delayed rectifier alpha-subunits | journal = Proceedings of the National Academy of Sciences of the United States of America | volume = 102 | issue = 17 | pages = 6160–5 | date = April 2005 | pmid = 15827117 | pmc = 1087924 | doi = 10.1073/pnas.0500468102 }}
*{{cite journal | author=Ottschytsch N, Raes A, Van Hoorick D, Snyders DJ |title=Obligatory heterotetramerization of three previously uncharacterized Kv channel alpha-subunits identified in the human genome. |journal=Proc. Natl. Acad. Sci. U.S.A. |volume=99 |issue= 12 |pages= 7986-91 |year= 2002 |pmid= 12060745 |doi= 10.1073/pnas.122617999 }}
*{{cite journal  | author=MacDonald PE, Wang G, Tsuk S, ''et al.'' |title=Synaptosome-associated protein of 25 kilodaltons modulates Kv2.1 voltage-dependent K(+) channels in neuroendocrine islet beta-cells through an interaction with the channel N terminus. |journal=Mol. Endocrinol. |volume=16 |issue= 11 |pages= 2452-61 |year= 2003 |pmid= 12403834 |doi=  }}
*{{cite journal  | author=Strausberg RL, Feingold EA, Grouse LH, ''et al.'' |title=Generation and initial analysis of more than 15,000 full-length human and mouse cDNA sequences. |journal=Proc. Natl. Acad. Sci. U.S.A. |volume=99 |issue= 26 |pages= 16899-903 |year= 2003 |pmid= 12477932 |doi= 10.1073/pnas.242603899 }}
*{{cite journal | author=Ju M, Stevens L, Leadbitter E, Wray D |title=The Roles of N- and C-terminal determinants in the activation of the Kv2.1 potassium channel. |journal=J. Biol. Chem. |volume=278 |issue= 15 |pages= 12769-78 |year= 2003 |pmid= 12560340 |doi= 10.1074/jbc.M212973200 }}
*{{cite journal | author=Tiran Z, Peretz A, Attali B, Elson A |title=Phosphorylation-dependent regulation of Kv2.1 Channel activity at tyrosine 124 by Src and by protein-tyrosine phosphatase epsilon. |journal=J. Biol. Chem. |volume=278 |issue= 19 |pages= 17509-14 |year= 2003 |pmid= 12615930 |doi= 10.1074/jbc.M212766200 }}
*{{cite journal | author=Consiglio JF, Korn SJ |title=Influence of permeant ions on voltage sensor function in the Kv2.1 potassium channel. |journal=J. Gen. Physiol. |volume=123 |issue= 4 |pages= 387-400 |year= 2004 |pmid= 15024041 |doi= 10.1085/jgp.200308976 }}
*{{cite journal | author=Thébaud B, Michelakis ED, Wu XC, ''et al.'' |title=Oxygen-sensitive Kv channel gene transfer confers oxygen responsiveness to preterm rabbit and remodeled human ductus arteriosus: implications for infants with patent ductus arteriosus. |journal=Circulation |volume=110 |issue= 11 |pages= 1372-9 |year= 2005 |pmid= 15353504 |doi= 10.1161/01.CIR.0000141292.28616.65 }}
*{{cite journal | author=Kerschensteiner D, Soto F, Stocker M |title=Fluorescence measurements reveal stoichiometry of K+ channels formed by modulatory and delayed rectifier alpha-subunits. |journal=Proc. Natl. Acad. Sci. U.S.A. |volume=102 |issue= 17 |pages= 6160-5 |year= 2005 |pmid= 15827117 |doi= 10.1073/pnas.0500468102 }}
}}
{{refend}}
{{refend}}


Line 91: Line 80:
* {{MeshName|KCNB1+protein,+human}}
* {{MeshName|KCNB1+protein,+human}}


{{membrane-protein-stub}}
{{NLM content}}
{{NLM content}}
{{Ion channels}}
{{Ion channels|g3}}
 
[[Category:Ion channels]]
[[Category:Ion channels]]
{{WikiDoc Sources}}

Latest revision as of 07:14, 10 January 2019

VALUE_ERROR (nil)
Identifiers
Aliases
External IDsGeneCards: [1]
Orthologs
SpeciesHumanMouse
Entrez
Ensembl
UniProt
RefSeq (mRNA)

n/a

n/a

RefSeq (protein)

n/a

n/a

Location (UCSC)n/an/a
PubMed searchn/an/a
Wikidata
View/Edit Human

Potassium voltage-gated channel, Shab-related subfamily, member 1, also known as KCNB1 or Kv2.1, is a protein that, in humans, is encoded by the KCNB1 gene.[1][2][3]

Potassium voltage-gated channel subfamily B member one, or simply known as KCNB1, is a delayed rectifier and voltage-gated potassium channel found throughout the body. The channel has a diverse number of functions. However, its main function, as a delayed rectifier, is to propagate current in its respective location. It is commonly expressed in the central nervous system, but may also be found in pulmonary arteries, auditory outer hair cells, stem cells, the retina, and organs such as the heart and pancreas. Modulation of K+ channel activity and expression has been found to be at the crux of many profound pathophysiological disorders in several cell types.[4]

Potassium channels are among the most diverse of all ion channels in eukaryotes. With over 100 genes coding numerous functions, many isoforms of potassium channels are present in the body, but most are divided up into two main groups: inactivating transient channels and non-inactivating delayed rectifiers. Due to the multiple varied forms, potassium delayed rectifier channels open or close in response to a myriad of signals. These include: cell depolarization or hyperpolarization, increases in intracellular calcium concentrations, neurotransmitter binding, or second messenger activity such as G-proteins or kinases.[5]

Structure

The general structure of all potassium channels contain a centered pore composed of alpha subunits with a pore loop expressed by a segment of conserved DNA, T/SxxTxGxG. This general sequence comprises the selectivity of the potassium channel. Depending on the channel, the alpha subunits are constructed in either a homo- or hetero-association, creating a 4-subunit selectivity pore or a 2-subunit pore, each with accessory beta subunits attached intracellularly. Also on the cytoplasmic side are the N- and C- termini, which play a crucial role in activating and deactivating KCNB1 channels. This pore creates the main opening of the channel where potassium ions flow through.[6]

The type of pore domain (number of subunits) determines if the channel has the typical 6 transmembrane (protein) spanning regions, or the less dominant inward rectifier type of only 2 regions. KCNB1 has 6TM labeled S1-S6, each with a tetrameric structure. S5 and S6 create the p-loop, while S4 is the location of the voltage sensor. S4, along with S2 and S3 create the ‘activating’ portions of the delayed rectifier channel.[6] The heteromeric complexes that contain the distinct pore are electrically inactive or non-conducting, but unlike other potassium families, the pore of the KCNB1 group has numerous phosphorylation sites allowing kinase activity. Maturing KCNB1 channels develop these phosphorylation sites within the channel pore, but lack a glycosylation stage in the N-terminus.[7]

Specifically, the KCNB1 delayed rectifier channel conducts a potassium current (K+). This mediates high frequency firing due to the phosphorylation sites located within the channel via kinases and a major calcium influx typical of all neurons.[7]

Kinetics

The kinetics surrounding the activation and deactivation of the KCNB1 channel is relatively unknown, and has been under considerable study. Three of the six transmembrane regions, S2, S3 and S4, contribute to the activation phase of the channel. Upon depolarization, the S4 region, which is positively charged, is moved in response to the subsequent positive charge of the depolarization. As a result of S4 movement, the negatively charged regions of S2 and S3 appear to move as well.[6] The movement of these regions causes an opening of the channel gate within regions of S5 and S6.[8] The intracellular regions of the C and N-terminus also play a crucial role in the activation kinetics of the channel. The two termini interact with one other, as the C-terminus folds around the N-terminus during channel activation. The relative movement between the N- and C- termini greatly aids in producing a conformational change of the channel necessary for channel opening. This interaction between these intracellular regions is believed to be linked with membrane-spanning regions of S1 and S6, and thus aid in the movement of S2, S3, and S4 in opening the channel.[6][8] Studies on selective mutations knocking out these intracellular termini have been shown to produce larger reductions in speed and probability of channel opening, which indicates their importance in channel activation.[6]

Function

Voltage-gated potassium (Kv) channels represent the most complex class of voltage-gated ion channels from both functional and structural standpoints.[1] Delayed rectifier potassium channels’ most prevalent role is in the falling phase of physiological action potentials. KCNB1 rectifiers are also important in forming the cardiac beat and rate synchronicity that exists within the heart, and the lysis of target molecules in the immune response. These channels can also act as effectors in downstream signaling in G-protein coupled receptor transduction. KCNB1’s regulation and propagation of current provides a means for regulatory control over several physiological functions.[5] Their diverse functions include regulating neurotransmitter release, heart rate, insulin secretion, neuronal excitability, epithelial electrolyte transport, smooth muscle contraction, and apoptosis.[1]

Voltage-gated potassium channels are essential in regulating neuronal membrane potential, and in contributing to action potential production and firing.[9] In mammalian CNS neurons, KCNB1 is a predominant delayed rectifier potassium current that regulates neuronal excitability, action potential duration, and tonic spiking. This is necessary when it comes to proper neurotransmitter release, as such release is dependent on membrane potential. In mouse cardiomyocytes, KCNB1 channel is the molecular substrate of major repolarization current IK-slow2. Transgenic mice, expressing a dominant-negative isoform of KCNB1, exhibit markedly prolonged action potentials and demonstrate arrhythmia.[10] KCNB1 also contributes to the function and regulation of smooth muscle fibers. Human studies on pulmonary arteries have shown that normal, physiological inhibition of KCNB1 current aids vasoconstriction of arteries.[11] In human pancreatic ß cells, KCNB1, which mediates potassium efflux, produces a downstroke of the action potential in the cell.[12] In effect, this behavior halts insulin secretion, as its activation decreases the Cav channel-mediated calcium influx that is necessary for insulin exocytosis. KCNB1 has also been found to promote apoptosis within neuronal cells. It is currently believed that KCNB1-induced apoptosis occurs in response to an increase in reactive oxygen species (ROS) that results either from acute oxidation or as a consequence of other cellular stresses.[7]

Regulation

KCNB1 conductance is regulated primarily by oligomerization and phosphorylation. Additional forms of regulation include SUMOylation and acetylation, although the direct effect of these modifications is still under investigation. KCNB1 consensus sites in the N-terminus are not subject to glycosylation.[4]

Phosphorylation

Many proteins undergo phosphorylation, or the addition of phosphate groups to amino acids subunits. Phosphorylation is modulated by kinases, which add phosphate groups, and phosphatases, which remove phosphate groups. In its phosphorylated state, KCNB1 is a poor conductor of current. There are 16 phosphorylation sites that are subject to the activity of kinases, such as cyclin-dependent kinase 5 and AMP-activated protein kinase. These sites are reversibly regulated by phosphatases such as, phosphatase calcineurin. Under periods of high electrical activity, depolarization of the neuron increases calcium influx and triggers phosphatase activity. Under resting conditions, KCNB1 tends to be phosphorylated. Phosphorylation raises the threshold voltage requirement for activation and allows microdomains to bind the channel, preventing KCNB1 from entering the plasma membrane. Microdomains localize KCNB1 in dendrites in cell bodies of hippocampal and cortical neurons. Conductance associated with de-phosphorylation of this channel acts to decrease or end periods high excitability. However, this relationship is not static and is cell dependent. The role of phosphorylation can be affected by reactive oxygen species (ROS) that increase during oxidative stress. ROS act to increase the levels of zinc (Zn2+) and calcium (Ca2+) intracellularly that act with protein kinases to phosphorylate certain sites on KCNB1. This phosphorylation increases the insertion of KCNB1 into the membrane and elevates conductance. Under these conditions the interaction with SNARE protein syntaxin, is enhanced. This surge of KCNB1 current induces activation of a pro-apoptotic pathway, DNA fragmentation, and caspase activation.[4]

Oligomerization

Another proposed mechanism for regulation of apoptosis is oligomerization, or the process of forming multi-protein complexes held together through disulfide bonds. Under oxidative stress, reactive oxygen species (ROS) form and act to regulate KCNB1 through oxidation. Increase in oxygen radicals directly causes formation of KCNB1 oligomers that then accumulate in the plasma membrane and initially decrease current flow [13][14]. Oligomer activation of c-Src and JNK kinases induces the initial pro-apoptotic signal, which is coupled to KCNB1 current. This further promotes the apoptosis pathway [15]. KCNB1 oligomers have been detected in the post mortem human hippocampus [16]

Blockers

Potassium delayed rectifiers have been implicated in many pharmacological uses in the investigation of biological toxins for drug development. A main component to many of the toxins with negative effects on delayed rectifiers contain cystine inhibitors that are arranged around disulfide bond formations. Many of these toxins originate from species of tarantulas. G. spatulata produces the hanatoxin, which was the first drug to be manipulated to interact with KCNB1 receptors by inhibiting the activation of most potassium voltage-gated channels. Other toxins, such as stromatoxin, heteroscordratoxin, and guangxitoxin, target the selectivity of voltage KCNB1 rectifiers, by either lowering potassium binding affinity or increasing the binding rate of potassium. This can lead to excitotoxicity, or overstimulation of postsynaptic neurons. In nature, the prey of tarantula that are injected with these endogenous toxins induces this excitotoxic effect, producing paralysis for easy capture. Physiologically, these venoms work on KCNB1 rectifier affinity by altering the channels’ voltage sensor, making it more or less sensitive to extracellular potassium concentrations.[17] KCNB1 is also susceptible to tetraethylammonium (TEA) and 4-aminopyridine (4-AP), which completely block all channel activity. TEA also works on calcium-activated potassium channels, furthering its inhibitory effects on neurons and skeletal muscle. Some isoforms of TEA are beneficial for patients with severe Alzheimer’s, as blocking KCNB1 channels reduces the amount of neuronal apoptosis, thereby slowing the rate of dementia.[18] This has been attributed to the oxidative properties of the channel by ROS.[5]

Physiological Role in Disease

Neurodegenerative Disease

Oxidative damage is widely considered to play a role in neurodegenerative disorders, including Alzheimers disease. Such oxidative stress alters the redox sensitivity of the Kv2.1 delayed rectifier, resulting in the modulation of the channel.[4] In vitro studies and studies in animal models show that when KCNB1 is oxidized, it no longer conducts, leading to neurons becoming hyperpolarized and dying; oxidized KCNB1 also clusters in lipid rafts and cannot be internalized, which also leads to apoptosis. These alterations disrupt normal neuronal signaling and increase the likelihood of neurological diseases. Oxidized (oligomerized) KCNB1 channels are present in the hippocampi of old (Braak stage 1-2) and Alzheimer's disease (Braak stage 5) donors of either sexes [19] [20]

Increased probability of the channel remaining open can also potentially drive neurodegeneration. Human immunodeficiency virus type-1 (HIV-1)-associated dementia (HAD) may be driven by an overabundance of glutamate, which in turn can trigger increased calcium levels, which in turn can drive calcium-dependent dephosphorylation of KCNB1 channels, which increases probability of channel activation and current conductance. Enhanced KCNB1 current couples cell shrinkage associated with apoptosis and dendritic beading leading to diminished long term potentiation. These neuronal modifications may explain the atrophy of cell layer volume and late stage cell death observed in HAD disease.[21]

Cancer

Exploitation of this channel is advantageous in cancer cell survival as they have the ability to produce heme oxygenase-1, an enzyme with the ability to generate carbon monoxide (CO). Oncogenic cells benefit from producing CO due to the antagonizing effects of the KCNB1 channel. Inhibition of KCNB1 allows cancer proliferation without the apoptotic pathway preventing tumor formation. Although potassium channels are studied as a therapeutic target for cancer, this apoptotic regulation is dependent on cancer type, potassium channel type, expression levels, intracellular localization as well as regulation by pro- or anti-apoptotic factors. [22]

Interactions

KCNB1 has been shown to interact with:

See also

References

  1. 1.0 1.1 1.2 "Entrez Gene: KCNB1 potassium voltage-gated channel, Shab-related subfamily, member 1".
  2. Melis R, Stauffer D, Zhao X, Zhu XL, Albrecht B, Pongs O, Brothman A, Leppert M (January 1995). "Physical and genetic localization of a Shab subfamily potassium channel (KCNB1) gene to chromosomal region 20q13.2". Genomics. 25 (1): 285–7. doi:10.1016/0888-7543(95)80138-C. PMID 7774931.
  3. Gutman GA, Chandy KG, Grissmer S, Lazdunski M, McKinnon D, Pardo LA, Robertson GA, Rudy B, Sanguinetti MC, Stühmer W, Wang X (December 2005). "International Union of Pharmacology. LIII. Nomenclature and molecular relationships of voltage-gated potassium channels". Pharmacological Reviews. 57 (4): 473–508. doi:10.1124/pr.57.4.10. PMID 16382104.
  4. 4.0 4.1 4.2 4.3 Shah NH, Aizenman E (February 2014). "Voltage-gated potassium channels at the crossroads of neuronal function, ischemic tolerance, and neurodegeneration". Translational Stroke Research. 5 (1): 38–58. doi:10.1007/s12975-013-0297-7. PMC 3946373. PMID 24323720.
  5. 5.0 5.1 5.2 "Potassium channel, voltage-dependent, beta subunit, KCNAB1 (IPR005400)". InterPro. EMBL-EBI. Retrieved 2017-04-04.
  6. 6.0 6.1 6.2 6.3 6.4 Wray D (May 2004). "The roles of intracellular regions in the activation of voltage-dependent potassium channels". European Biophysics Journal. 33 (3): 194–200. doi:10.1007/s00249-003-0363-2. PMID 14608450.
  7. 7.0 7.1 7.2 Patel R, Sesti F (May 2016). "Oxidation of ion channels in the aging nervous system". Brain Research. 1639: 174–85. doi:10.1016/j.brainres.2016.02.046. PMID 26947620.
  8. 8.0 8.1 Wray D (March 2009). "Intracellular regions of potassium channels: Kv2.1 and heag". European Biophysics Journal. 38 (3): 285–92. doi:10.1007/s00249-008-0354-4. PMID 18607586.
  9. Sesti F (March 2016). "Oxidation of K(+) Channels in Aging and Neurodegeneration". Aging and Disease. 7 (2): 130–5. doi:10.14336/AD.2015.0901. PMC 4809605. PMID 27114846.
  10. Murakoshi H, Trimmer JS (March 1999). "Identification of the Kv2.1 K+ channel as a major component of the delayed rectifier K+ current in rat hippocampal neurons" (PDF). The Journal of Neuroscience. 19 (5): 1728–35. PMID 10024359.
  11. Joseph BK, Thakali KM, Moore CL, Rhee SW (April 2013). "Ion channel remodeling in vascular smooth muscle during hypertension: Implications for novel therapeutic approaches". Pharmacological Research. 70 (1): 126–38. doi:10.1016/j.phrs.2013.01.008. PMC 3607210. PMID 23376354.
  12. Yang SN, Shi Y, Yang G, Li Y, Yu J, Berggren PO (November 2014). "Ionic mechanisms in pancreatic β cell signaling". Cellular and Molecular Life Sciences. 71 (21): 4149–77. doi:10.1007/s00018-014-1680-6. PMID 25052376.
  13. Wu X, Hernandez-Enriquez B, Banas M, Xu R, Sesti F (2013). "Molecular mechanisms underlying the apoptotic effect of KCNB1 K+ channel oxidation". J Biol Chem. 288 (6): 4128–4134. doi:10.1074/jbc.M112.440933. PMC 3567663.
  14. Cotella D, Hernandez B, Wu X, Li R, Pan Z, Leveille J, Link CD, Oddo S, Sesti F (2012). "Toxic role of K+ channel oxidation in mammalian brain". J. Neurosci. 32 (12): 4133–4144. doi:10.1523/JNEUROSCI.6153-11.2012.
  15. Yu W, Gowda M, Singh S, Sesti F (2017). "Oxidation of KCNB1 potassium channels triggers apoptotic integrin signaling in the brain". Cell Death Dis. 8 (4): e2737. doi:10.1038/cddis.2017.160.
  16. Wei Y, Shih R, Sesti F (2018). "Oxidation of KCNB1 channels in the human brain and in mouse model of Alzheimer's disease". Cell Death Dis. 9 (820). doi:10.1038/s41419-018-0886-1.
  17. Swartz KJ (February 2007). "Tarantula toxins interacting with voltage sensors in potassium channels". Toxicon. 49 (2): 213–30. doi:10.1016/j.toxicon.2006.09.024. PMC 1839852. PMID 17097703.
  18. Quinn CC, Begenisich T (2017-04-12). "Pharmacology and surface electrostatics of the K channel outer pore vestibule". The Journal of Membrane Biology. 212 (1): 51–60. doi:10.1007/s00232-006-0039-9. PMC 1784061. PMID 17206516.
  19. Wei Y, Shih R, Sesti F (2018). "Oxidation of KCNB1 channels in the human brain and in mouse model of Alzheimer's disease". Cell Death Dis. 9 (820). doi:10.1038/s41419-018-0886-1.
  20. Peers C, Boyle JP (February 2015). "Oxidative modulation of K+ channels in the central nervous system in neurodegenerative diseases and aging". Antioxidants & Redox Signaling. 22 (6): 505–21. doi:10.1089/ars.2014.6007. PMID 25333910.
  21. Keblesh J, Hu D, Xiong H (March 2009). "Voltage-gated potassium channels in human immunodeficiency virus type-1 (HIV-1)-associated neurocognitive disorders". Journal of Neuroimmune Pharmacology. 4 (1): 60–70. doi:10.1007/s11481-008-9106-6. PMC 3974578. PMID 18459047.
  22. Kondratskyi A, Kondratska K, Skryma R, Prevarskaya N (October 2015). "Ion channels in the regulation of apoptosis". Biochimica et Biophysica Acta. Membrane Channels and Transporters in Cancers. 1848 (10 Pt B): 2532–46. doi:10.1016/j.bbamem.2014.10.030. PMID 25450339.
  23. Ottschytsch N, Raes A, Van Hoorick D, Snyders DJ (June 2002). "Obligatory heterotetramerization of three previously uncharacterized Kv channel alpha-subunits identified in the human genome". Proceedings of the National Academy of Sciences of the United States of America. 99 (12): 7986–91. doi:10.1073/pnas.122617999. PMC 123007. PMID 12060745.
  24. Peretz A, Gil-Henn H, Sobko A, Shinder V, Attali B, Elson A (August 2000). "Hypomyelination and increased activity of voltage-gated K(+) channels in mice lacking protein tyrosine phosphatase epsilon". The EMBO Journal. 19 (15): 4036–45. doi:10.1093/emboj/19.15.4036. PMC 306594. PMID 10921884.

Further reading

External links

This article incorporates text from the United States National Library of Medicine, which is in the public domain.