Skip to main content
Erschienen in: Molecular Cancer 1/2013

Open Access 01.12.2013 | Short communication

BAK multimerization for apoptosis, but not bid binding, is inhibited by negatively charged residue in the BAK hydrophobic groove

verfasst von: Abul Azad, Alan Storey

Erschienen in: Molecular Cancer | Ausgabe 1/2013

download
DOWNLOAD
print
DRUCKEN
insite
SUCHEN

Abstract

Background

BCL-2 family proteins BAK and BAX orchestrate outer mitochondrial membrane permeabilization (MOMP) during apoptosis by forming pores in the membrane to release apoptogenic factors that commits a cell to death. BAK and BAX therefore function as a ‘point of no return’ in the apoptotic cascade. BAK activation is a multi-step process involving conformational changes, mediated by BH3-only proteins or p53, which lead eventually to oligomerization and pore formation. Further, recent reports show that BAK activation is also linked to and dependent upon dephosphorylation of both tyrosine and serine residues.

Findings

We hypothesized that phosphorylation of BAK at tyrosine residue 110 (Y110) was functionally important during the BAK activation process. BAK/BAX double knockout HCT116 cells expressing a phosphor-mimetic BAK mutant (BAK Y110E), showed impaired dimerization and multimerization capacity when treated with either UV irradiation or etoposide when compared to cells reconstituted to express wild-type BAK. The Y110E mutant also showed decreased release of cytochrome c from isolated mitochondria challenged with tBid protein, resulting in a failure to activate caspase 3. Interestingly, co-immunoprecipitation experiments suggest that a negative charge at this residue may be important for the recruitment of Bid to BAK, but conversely that this also impairs BAK:BAK interactions.

Conclusion

These findings implicate dephosphorylation of Y110 as having an important mechanistic role in BAK activation, and underscores how post-translational modifications are intimately linked and coupled to the protein-protein interactions required for BAK activation during apoptosis.
Hinweise

Electronic supplementary material

The online version of this article (doi:10.​1186/​1476-4598-12-65) contains supplementary material, which is available to authorized users.

Competing interests

The authors state that they have no competing interests.

Authors’ contributions

AA and AS designed the experiments; AA performed all experimental procedures; AA and AS analysed data and generated images, and prepared and approved the final version of the manuscript.

Findings

The abrogation of apoptotic responses is a common feature of almost all cancer cells. Whether or not a cell undergoes apoptosis in response to stress depends on responses following the receipt of signals generated by cellular damage. The mitochondrial apoptotic pathway is regulated by BCL-2 proteins that can be divided into different pro- and anti-apoptotic groups depending on their structure and function [1]. Pro-apoptotic BH3-only proteins, such as Bid, Bim and Noxa, are required for the activation of multi-domain BCL-2 effector proteins BAK and BAX. BAK and BAX constitute a pre-requisite gateway for mitochondrial apoptosis to proceed, indeed cells lacking both BAK and BAX fail to undergo mitochondrial apoptosis [2]. The BAK responds to a myriad of death signals and plays a key role in executing mitochondrial apoptosis, effecting MOMP through oligomerization of the protein that releases apoptogenic factors including cytochrome c, that in turn lead to downstream activation of caspase 3. BAK activation is complex, involving a number of intra-molecular conformational changes leading to dimerization followed by oligomerization [24]. An early event during BAK activation is a conformational change towards to N-terminus of the protein [5]. This is followed by exposure of the BAK BH3 domain that then inserts into a hydrophobic groove on another BAK molecule leading to dimer formation [6]. Resultant homodimers then can form higher order structures via interaction between α6:α6 helices [7]. BAK N-terminal conformational change can be brought about by one of two mechanisms: first, by the transient binding of BH3-only proteins (such as tBid) [8] to the BAK hydrophobic groove [2, 9], or alternatively by the binding of p53 to residues near the BAK N-terminus [1012]. Binding of BH3 proteins such as tBid to the BAK hydrophobic groove occurs with high affinity, but is necessarily transient as this same region of BAK is also required to nucleate BAK multimerization. Recently a model of BAK activation that tries to take into account the differing affinities of BH3 proteins for both pro- and anti-apoptotic BCL-2 proteins has been proposed [13].
In healthy, otherwise undamaged cells, BAK is present almost exclusively in a very heavily phosphorylated form. We recently demonstrated that BAK activation for apoptosis induction is closely linked to, and indeed dependent upon, specific dephosphorylation events on the protein [14]. The initial event in the BAK activation process is dephosphorylation at tyrosine 108 (Y108), an obligatory step that is required to permit conformational change by BH3 or p53 proteins [14, 15]. Further, we found that a subsequent PP2A-mediated dephosphorylation of BAK at serine 117 (S117) was required both for BH3 proteins to gain access to the BAK hydrophobic groove, and permit BAK dimerization via BAK-BH3:BAK-groove interactions [16].
During our investigations into the role of phosphorylation in regulating BAK activation, we reported in mass spectrometry analysis that BAK was also phosphorylated at residue Y110 [14]. Mutation of this residue to mimic either the dephosphorylated or phosphorylated forms of the protein (BAK mutants Y110F and Y110E respectively) did not affect the ability of BAK to undergo N-terminal conformational change [14]. However, modelling using PyMol based on BAK structure 2IMS, suggested that phosphorylation at Y110 might impinge upon the ability of BH3 proteins to bind BAK and may also inhibit BAK multimerization, as the Y110 side chain, like that of S117, may occlude access to the hydrophobic groove (Figure 1A). To test this idea, we performed multimerization assays using sub-cellular fractions enriched in mitochondria from HCT116bak−/−bax−/− cells that had been reconstituted to express either wild-type (WT) or mutant BAK proteins (HCT-BAK cells). Cross-linking reactions were performed with the sulfhydryl-to-sulfhydryl crosslinker BMH (1, 6-bismaleimidohexane, Pierce) on sub-cellular fractions enriched in mitochondria, then analysed by western blotting to detected BAK multimers, as previously described [14, 16]. Following DNA damage by UV, BAK dimers, trimers and higher-order complexes were observed with WT BAK and the Y110F mutant, but multimer formation was impaired severely by the Y110E mutation (Figure 1B, upper panel). As we have noted previously, BAK cross-linking with BMH can be problematic with the formation of a faster migrating intra-molecularly cross-linked monomeric BAK protein (Mx) and generation of non-specific dimer bands without any DNA damage. A further cross-linking experiment a different sulfhydryl crosslinker, BMOE (Bismaleimidoethane), that generates only BAK dimers and at the same time minimizes the detection of both intra-molecularly cross-linked BAK monomers and non-specific dimer forms [16], supported the previous experiments with dimers being detected only when WT or the Y110F mutant was used (Figure 1B, lower panel). When the experiment was repeated using etoposide as the apoptotic inducer in place of UV, again dimers were readily detected in cells expressing the WT and Y110F proteins, but were not generated by the Y110E mutant (Figure 1C). The stronger dimer band present generated by the Y110F mutant compared to WT we suppose may be due to the mutant forming dimers more readily if the WT protein was not fully dephosphorylated following UV damage.
The failure of the BAK Y110E mutant to dimerize or multimerize we reasoned may be due to interference with the binding of BH3 proteins, as observed previously for the S117E phosphor-mimic [16]. We therefore tested the ability of the BH3-activator protein tBid to release cytochrome c from mitochondrial preparations using a previously established method [16, 17]. When purified tBid protein was incubated with mitochondrial preparations from cells expressing either the WT or Y110F BAK proteins, cytochrome c was readily released into the supernatant (Figure 2A). Consistent with the multimerization assays, tBid was unable to release cytochrome c from mitochondrial preparations from the BAK-Y110E mutant cells compared to the HCT116-BAK or HCT116-BAKY110F cells (Figure 2A). The small quantity of cytochrome c that was detected in the supernatant fraction derived from the Y110E mutant might be due to the mutant being very inefficient at releasing cytochrome c since the substituted amino acid may not correctly mimic a phosphor-tyrosine residue, however we have noted that processing of the samples leads to a degree of leakiness of the mitochondrial preparations where mitochondria expressing BAK mutants we find to be more fragile compared to cells expressing WT BAK, as previously noted [16]. Likewise, the Y110F mutant showed amounts of cytochrome c retained in the pellet fraction decreased somewhat when mitochondrial preparations were treated with increasing amounts of tBid, but by comparison cytochrome c was readily detected in the supernatant fraction even in the absence of tBid, and did not increase to the same extent as observed for the WT BAK protein. To investigate further whether the cytochrome c release was likely to be functionally significant and bypass potential problems of mitochondrial fragility, we performed caspase 3 activation assays using a FITC-conjugated antibody that recognizes only activated caspase 3 using FACS on intact cells. This revealed that UV irradiation resulted in an increase in caspase 3 activation in BAK WT or Y110F cells, but in marked contrast no caspase 3 activation was detected in UV-irradiated Y110E cells, findings in-line with and supporting the cytochrome c release experiment (Figure 2B). We conclude that the Y110E mutation interferes with the ability of BAK to form dimers and multimers, and that this results in the failure to release cytochrome c and activate caspase 3.
In previous studies, we found that neither the BAK Y110F nor Y110E mutations had any effect on BAK being able to undergo the N-terminal conformational change associated with an early step in BAK activation, indicating that the mutations did not perturb the overall BAK structure [14]. Since either p53 or BH3 proteins bring about BAK conformational change by different routes [16], we next asked whether the BAK mutants recruited p53 to the mitochondria as efficiently as the WT protein. The p53 binding site on BAK is located near the N-terminus of the protein involving residues E24/E25 [12, 16], and p53 recruitment to the mitochondria is dependent on BAK expression [10, 18, 19]. Following DNA damage by UV irradiation, we found that p53 was readily recruited to the mitochondria as was detected by western blot in mitochondrial extracts of cells expressing the WT or either of the two mutant BAK Y110F or Y110E (Figure 3A). As the Bid BH3 sequence interacts with the BAK hydrophobic groove, we next examined whether Bid was able to Co-IP BAK following UV irradiation. As expected, Bid was able to pull down BAK protein following UV irradiation from whole cell extracts of cells expressing either the WT or Y110F mutant BAK proteins (Figure 3B). Somewhat surprisingly, Bid was able to pull down more BAK from the Y110E cells when compared to cells expressing either WT or the Y110F mutant (Figure 3B). To explore further the role of a negative charge at Y110 on the recruitment of Bid to BAK, we performed similar IP/western blot experiments using mitochondrial enriched preparations. We found that similar levels of Bid were able to be immunoprecipitated in the three BAK cell lines expressing either WT, Y110F or Y110E mutants, irrespective of whether the cells has been treated with UV or not (Figure 3C). As before, WT or Y110F BAK proteins were only co-precipitated with Bid following UV damage, and that a greater amount of the Y110E BAK mutant we again found was readily pulled down by Co-IP with Bid following UV treatment (Figure 3C). However, in these mitochondrial enriched subcellular fractions we were also now able to detect BAK that had been pulled down by the Bid antibody even in the absence of UV treatment. Together these findings provides the first evidence that a negative charge located in the BAK hydrophobic surface groove may be a factor important in the recruitment of BH3 proteins such as Bid to BAK.
Blockade of the BAK hydrophobic groove by phosphorylation of S117, or an S117E mutant, each impair binding of either BH3 peptides or proteins to BAK. Located further along the same helix as S117 but also facing the hydrophobic BAK surface pocket, we now identify a role for Y110 in the BAK activation process. Taken together, our results identify for the first time a novel and important bi-functional role for post-translational modifications at Y110. Binding of BH3 proteins to BAK has been reported to occur with high affinity yet the interaction must be transient [20], as the same hydrophobic surface interface is required to nucleate BAK dimerization. We recently reported that phosphorylation of BAK at S117 blocks access of BH3 sequences of Bid and BAK to the hydrophobic pocket on BAK. In contrast, these findings suggest that while the Y110E mutant interferes with the ability to form stable BAK-BH3:BAK-groove interactions for dimerization, a negative charge at this site may also be a positive factor that is involved in the recruitment of BH3-containing proteins, such as Bid, to BAK. This implies that dephosphorylation of S117 must occur prior to dephosphorylation of Y110 in order to permit Bid recruitment, however the exact timing of these events and whether binding of other BH3-containing BCL-2 family proteins is also affected by this modification remains to be established.
Findings presented here further underscore the importance of post-translational modifications in regulating different steps in BAK activation, and may reveal new avenues to either potentiate or inhibit BAK activity through modulation of the phosphorylation status of the protein.

Acknowledgements

This work was supported by funding from Cancer Research UK. We also thank R. Youle for the gift of the HCT116bax−/− bak−/− double knockout cells.
This article is published under license to BioMed Central Ltd. This is an Open Access article distributed under the terms of the Creative Commons Attribution License (http://​creativecommons.​org/​licenses/​by/​2.​0), which permits unrestricted use, distribution, and reproduction in any medium, provided the original work is properly cited.

Competing interests

The authors state that they have no competing interests.

Authors’ contributions

AA and AS designed the experiments; AA performed all experimental procedures; AA and AS analysed data and generated images, and prepared and approved the final version of the manuscript.
Anhänge

Authors’ original submitted files for images

Literatur
2.
Zurück zum Zitat Wei MC, Zong WX, Cheng EH, Lindsten T, Panoutsakopoulou V, Ross AJ, Roth KA, MacGregor GR, Thompson CB, Korsmeyer SJ: Proapoptotic BAX and BAK: a requisite gateway to mitochondrial dysfunction and death. Science. 2001, 292: 727-730. 10.1126/science.1059108PubMedCentralCrossRefPubMed Wei MC, Zong WX, Cheng EH, Lindsten T, Panoutsakopoulou V, Ross AJ, Roth KA, MacGregor GR, Thompson CB, Korsmeyer SJ: Proapoptotic BAX and BAK: a requisite gateway to mitochondrial dysfunction and death. Science. 2001, 292: 727-730. 10.1126/science.1059108PubMedCentralCrossRefPubMed
3.
Zurück zum Zitat van Van Delft MF, Huang DC: How the Bcl-2 family of proteins interact to regulate apoptosis. Cell Res. 2006, 16: 203-213. 10.1038/sj.cr.7310028CrossRef van Van Delft MF, Huang DC: How the Bcl-2 family of proteins interact to regulate apoptosis. Cell Res. 2006, 16: 203-213. 10.1038/sj.cr.7310028CrossRef
4.
Zurück zum Zitat Youle RJ, Strasser A: The BCL-2 protein family: opposing activities that mediate cell death. Nat Rev Mol Cell Biol. 2008, 9: 47-59. 10.1038/nrm2308CrossRefPubMed Youle RJ, Strasser A: The BCL-2 protein family: opposing activities that mediate cell death. Nat Rev Mol Cell Biol. 2008, 9: 47-59. 10.1038/nrm2308CrossRefPubMed
5.
Zurück zum Zitat Griffiths GJ, Dubrez L, Morgan CP, Jones NA, Whitehouse J, Corfe BM, Dive C, Hickman JA: Cell damage-induced conformational changes of the pro-apoptotic protein Bak in vivo precede the onset of apoptosis. J Cell Biol. 1999, 144: 903-914. 10.1083/jcb.144.5.903PubMedCentralCrossRefPubMed Griffiths GJ, Dubrez L, Morgan CP, Jones NA, Whitehouse J, Corfe BM, Dive C, Hickman JA: Cell damage-induced conformational changes of the pro-apoptotic protein Bak in vivo precede the onset of apoptosis. J Cell Biol. 1999, 144: 903-914. 10.1083/jcb.144.5.903PubMedCentralCrossRefPubMed
6.
Zurück zum Zitat Dewson G, Kratina T, Sim HW, Puthalakath H, Adams JM, Colman PM, Kluck RM: To trigger apoptosis, Bak exposes its BH3 domain and homodimerizes via BH3:groove interactions. Mol Cell. 2008, 30: 369-380. 10.1016/j.molcel.2008.04.005CrossRefPubMed Dewson G, Kratina T, Sim HW, Puthalakath H, Adams JM, Colman PM, Kluck RM: To trigger apoptosis, Bak exposes its BH3 domain and homodimerizes via BH3:groove interactions. Mol Cell. 2008, 30: 369-380. 10.1016/j.molcel.2008.04.005CrossRefPubMed
7.
Zurück zum Zitat Dewson G, Kratina T, Czabotar P, Day CL, Adams JM, Kluck RM: Bak activation for apoptosis involves oligomerization of dimers via their alpha6 helices. Mol Cell. 2009, 36: 696-703. 10.1016/j.molcel.2009.11.008CrossRefPubMed Dewson G, Kratina T, Czabotar P, Day CL, Adams JM, Kluck RM: Bak activation for apoptosis involves oligomerization of dimers via their alpha6 helices. Mol Cell. 2009, 36: 696-703. 10.1016/j.molcel.2009.11.008CrossRefPubMed
8.
Zurück zum Zitat Kim H, Rafiuddin-Shah M, Tu HC, Jeffers JR, Zambetti GP, Hsieh JJ, Cheng EH: Hierarchical regulation of mitochondrion-dependent apoptosis by BCL-2 subfamilies. Nat Cell Biol. 2006, 8: 1348-1358. 10.1038/ncb1499CrossRefPubMed Kim H, Rafiuddin-Shah M, Tu HC, Jeffers JR, Zambetti GP, Hsieh JJ, Cheng EH: Hierarchical regulation of mitochondrion-dependent apoptosis by BCL-2 subfamilies. Nat Cell Biol. 2006, 8: 1348-1358. 10.1038/ncb1499CrossRefPubMed
9.
Zurück zum Zitat Korsmeyer SJ, Wei MC, Saito M, Weiler S, Oh KJ, Schlesinger PH: Pro-apoptotic cascade activates BID, which oligomerizes BAK or BAX into pores that result in the release of cytochrome c. Cell Death Differ. 2000, 7: 1166-1173. 10.1038/sj.cdd.4400783CrossRefPubMed Korsmeyer SJ, Wei MC, Saito M, Weiler S, Oh KJ, Schlesinger PH: Pro-apoptotic cascade activates BID, which oligomerizes BAK or BAX into pores that result in the release of cytochrome c. Cell Death Differ. 2000, 7: 1166-1173. 10.1038/sj.cdd.4400783CrossRefPubMed
10.
Zurück zum Zitat Leu JI, Dumont P, Hafey M, Murphy ME, George DL: Mitochondrial p53 activates Bak and causes disruption of a Bak-Mcl1 complex. Nat Cell Biol. 2004, 6: 443-450. 10.1038/ncb1123CrossRefPubMed Leu JI, Dumont P, Hafey M, Murphy ME, George DL: Mitochondrial p53 activates Bak and causes disruption of a Bak-Mcl1 complex. Nat Cell Biol. 2004, 6: 443-450. 10.1038/ncb1123CrossRefPubMed
11.
Zurück zum Zitat Mihara M, Erster S, Zaika A, Petrenko O, Chittenden T, Pancoska P, Moll UM: p53 has a direct apoptogenic role at the mitochondria. Mol Cell. 2003, 11: 577-590. 10.1016/S1097-2765(03)00050-9CrossRefPubMed Mihara M, Erster S, Zaika A, Petrenko O, Chittenden T, Pancoska P, Moll UM: p53 has a direct apoptogenic role at the mitochondria. Mol Cell. 2003, 11: 577-590. 10.1016/S1097-2765(03)00050-9CrossRefPubMed
12.
Zurück zum Zitat Pietsch EC, Perchiniak E, Canutescu AA, Wang G, Dunbrack RL, Murphy ME: Oligomerization of BAK by p53 utilizes conserved residues of the p53 DNA binding domain. J Biol Chem. 2008, 283: 21294-21304. 10.1074/jbc.M710539200PubMedCentralCrossRefPubMed Pietsch EC, Perchiniak E, Canutescu AA, Wang G, Dunbrack RL, Murphy ME: Oligomerization of BAK by p53 utilizes conserved residues of the p53 DNA binding domain. J Biol Chem. 2008, 283: 21294-21304. 10.1074/jbc.M710539200PubMedCentralCrossRefPubMed
13.
Zurück zum Zitat Llambi F, Moldoveanu T, Tait SW, Bouchier-Hayes L, Temirov J, McCormick LL, Dillon CP, Green DR: A unified model of mammalian BCL-2 protein family interactions at the mitochondria. Mol Cell. 2011, 44: 517-531. 10.1016/j.molcel.2011.10.001PubMedCentralCrossRefPubMed Llambi F, Moldoveanu T, Tait SW, Bouchier-Hayes L, Temirov J, McCormick LL, Dillon CP, Green DR: A unified model of mammalian BCL-2 protein family interactions at the mitochondria. Mol Cell. 2011, 44: 517-531. 10.1016/j.molcel.2011.10.001PubMedCentralCrossRefPubMed
14.
Zurück zum Zitat Fox JL, Ismail F, Azad A, Ternette N, Leverrier S, Edelmann MJ, Kessler BM, Leigh IM, Jackson S, Storey A: Tyrosine dephosphorylation is required for Bak activation in apoptosis. EMBO J. 2010, 29: 3853-3868. 10.1038/emboj.2010.244PubMedCentralCrossRefPubMed Fox JL, Ismail F, Azad A, Ternette N, Leverrier S, Edelmann MJ, Kessler BM, Leigh IM, Jackson S, Storey A: Tyrosine dephosphorylation is required for Bak activation in apoptosis. EMBO J. 2010, 29: 3853-3868. 10.1038/emboj.2010.244PubMedCentralCrossRefPubMed
15.
Zurück zum Zitat Fox J, Azad A, Ismail F, Storey A: "Licensed to kill": tyrosine dephosphorylation and Bak activation. Cell Cycle. 2011, 10: 598-603. 10.4161/cc.10.4.14793PubMedCentralCrossRefPubMed Fox J, Azad A, Ismail F, Storey A: "Licensed to kill": tyrosine dephosphorylation and Bak activation. Cell Cycle. 2011, 10: 598-603. 10.4161/cc.10.4.14793PubMedCentralCrossRefPubMed
16.
Zurück zum Zitat Azad A, Fox J, Leverrier S, Storey A: Blockade of the BAK hydrophobic groove by inhibitory phosphorylation regulates commitment to apoptosis. PLoS One. 2012, 7: e49601- 10.1371/journal.pone.0049601PubMedCentralCrossRefPubMed Azad A, Fox J, Leverrier S, Storey A: Blockade of the BAK hydrophobic groove by inhibitory phosphorylation regulates commitment to apoptosis. PLoS One. 2012, 7: e49601- 10.1371/journal.pone.0049601PubMedCentralCrossRefPubMed
17.
Zurück zum Zitat Waterhouse NJ, Trapani JA: A new quantitative assay for cytochrome c release in apoptotic cells. Cell Death Differ. 2003, 10: 853-855. 10.1038/sj.cdd.4401263CrossRefPubMed Waterhouse NJ, Trapani JA: A new quantitative assay for cytochrome c release in apoptotic cells. Cell Death Differ. 2003, 10: 853-855. 10.1038/sj.cdd.4401263CrossRefPubMed
18.
Zurück zum Zitat Moll UM, Wolff S, Speidel D, Deppert W: Transcription-independent pro-apoptotic functions of p53. Curr Opin Cell Biol. 2005, 17: 631-636. 10.1016/j.ceb.2005.09.007CrossRefPubMed Moll UM, Wolff S, Speidel D, Deppert W: Transcription-independent pro-apoptotic functions of p53. Curr Opin Cell Biol. 2005, 17: 631-636. 10.1016/j.ceb.2005.09.007CrossRefPubMed
19.
Zurück zum Zitat Leverrier S, Bergamaschi D, Ghali L, Ola A, Warnes G, Akgul B, Blight K, Garcia-Escudero R, Penna A, Eddaoudi A, Storey A: Role of HPV E6 proteins in preventing UVB-induced release of pro-apoptotic factors from the mitochondria. Apoptosis. 2007, 12: 549-560. 10.1007/s10495-006-0004-1CrossRefPubMed Leverrier S, Bergamaschi D, Ghali L, Ola A, Warnes G, Akgul B, Blight K, Garcia-Escudero R, Penna A, Eddaoudi A, Storey A: Role of HPV E6 proteins in preventing UVB-induced release of pro-apoptotic factors from the mitochondria. Apoptosis. 2007, 12: 549-560. 10.1007/s10495-006-0004-1CrossRefPubMed
20.
Zurück zum Zitat Dai H, Smith A, Meng XW, Schneider PA, Pang YP, Kaufmann SH: Transient binding of an activator BH3 domain to the Bak BH3-binding groove initiates Bak oligomerization. J Cell Biol. 2011, 194: 39-48. 10.1083/jcb.201102027PubMedCentralCrossRefPubMed Dai H, Smith A, Meng XW, Schneider PA, Pang YP, Kaufmann SH: Transient binding of an activator BH3 domain to the Bak BH3-binding groove initiates Bak oligomerization. J Cell Biol. 2011, 194: 39-48. 10.1083/jcb.201102027PubMedCentralCrossRefPubMed
Metadaten
Titel
BAK multimerization for apoptosis, but not bid binding, is inhibited by negatively charged residue in the BAK hydrophobic groove
verfasst von
Abul Azad
Alan Storey
Publikationsdatum
01.12.2013
Verlag
BioMed Central
Erschienen in
Molecular Cancer / Ausgabe 1/2013
Elektronische ISSN: 1476-4598
DOI
https://doi.org/10.1186/1476-4598-12-65

Weitere Artikel der Ausgabe 1/2013

Molecular Cancer 1/2013 Zur Ausgabe

Update Onkologie

Bestellen Sie unseren Fach-Newsletter und bleiben Sie gut informiert.