ArticlePDF Available

Abstract and Figures

Mitochondria play a fundamental role in ATP synthesis within the majority of mammalian cells. Potassium channels present in the inner mitochondrial membrane are fine regulators of mitochondrial function, based on inner membrane K+ permeability. These channels are regulated by a plethora of factors and conditions in a way similar to plasma membrane potassium channels. Regulators of mitochondrial potassium channels include the membrane potential, calcium ions, free fatty acids and ATP levels within the cells. Recently, it was shown that these channels are regulated by the respiratory chain, stretching of the membrane and phosphorylation. The essential interest that has driven studies of mitochondrial potassium channels for nearly 25 years is their role in cytoprotection and in cell death. Mitochondrial potassium channels have been described in neurons, astrocytoma, cardiac and skeletal muscles, fibroblasts, keratinocytes and endothelial cells. In this overview, we summarize the current knowledge of mitochondrial potassium channels. This summary will be done with a special focus on studies performed over the last 20 years in the Laboratory of Intracellular Ion Channels at the Nencki Institute. These include studies on the electrophysiological and pharmacological properties of mitochondrial potassium channels and on their regulation by endogenous intracellular substances. Additionally, the regulation of mitochondrial potassium channels by the respiratory chain and by stretching of the inner mitochondrial membrane will be reviewed. Properties of mitochondrial potassium channels in various organisms will also be summarized.
Content may be subject to copyright.
190 www.postepybiochemii.pl
Joanna Bandorowicz-Pikuła*
Agnieszka Kinga Seliga
Laboratory of Cellular Metabolism, Nencki In-
stitute of Experimental Biology, 3 Pasteura St.,
02-093 Warsaw, Poland
*e-mail: j.bandorowicz-pikula@nencki.gov.pl
Received: September 25, 2018
Accepted: October 23, 2018
Key words: annexin A6, cholesterol metabo-
lism, membrane repair, vesicular transport
Acknowledgements: The research in Authors’
Laboratory is supported by statutory fund
from the Nencki Institute of Experimental Bio-
logy, Polish Academy of Sciences and by an
Opus grant, reg. no 2016/23/B/NZ3/03116,
from the National Science Center to JBP.
Annexin A6 as a cholesterol and nucleotide binding protein involved
in membrane repair and in controlling membrane transport
during endo- and exocytosis
ABSTRACT
Annexins, calcium- and membrane-binding proteins, have been extensively studied at the
Nencki Institute since early 1990s, in terms of their structure, potential ligands and func-
tions in the organism, with emphasis on mineralization processes in norm and pathology.
The results of recently performed studies have revealed that annexins are playing essential
roles in membrane organization. In this review we characterize the largest member of the
annexin family of proteins, annexin A6 (AnxA6), in respect to its cholesterol and nucleoti-
de binding properties, as well as intracellular pH sensing and ability to change membrane
permeability to ions. Furthermore, we discuss biological functions of AnxA6 such as partici-
pation in membrane lateral organization, cell membrane repair and regulation of vesicular
transport.
INTRODUCTION – THE ANNEXINS
The vertebrate annexin (AnxA) superfamily consists of 12 members of cal-
cium and phospholipid binding proteins which share high structural homology
[Morgan et al. 2004; 2006; Gerke et al. 2005; Bandorowicz-Pikula 2003; Bandoro-
wicz Pikula et al. 2001; 2012; Domon et al. 2012; Kodavali et al. 2014; Grewal et al.
2016]. In keeping with this hallmark feature, annexins have been implicated in
Ca2+-controlled regulation of a broad range of membrane related events, which
may suggest its potential therapeutic value, namely, the regulation of immune
response and control of tissue homeostasis [Schloer et al. 2018]. Recent highlights
concerning transgenic (knockout animals) are summarized in table 1.
In this review, we focus on one of the main subjects of our investigations,
annexin A6 (AnxA6), its properties and potential functions in addition to major
achievements made by us in this eld since 1997, we discuss recent advances
related to hypothetical functions of AnxA6, including organization of biological
membranes, or membrane repair mechanisms. The latter may be controlled in
response to a disrupted cellular hemostasis and vesicle-related cellular process-
es, such as biomineralization [Balcerzak et al. 2008; Kapustin & Shanahan 2016;
Minashima & Kirsch 2018; Bottini et al. 2018], transport and storage of cholesterol
[Enrich et al. 2011; 2017; Reverter et al. 2011].
ANNEXIN A6 AT THE NENCKI INSTITUTE
Annexins were rst introduced to the Nencki Institute as one among sever-
al experimental subjects of the Laboratory of Plasma Membrane Receptors at
early 1990s [Bandorowicz et al. 1992; 1996; Sobota et al. 1993], and since 1997
further explored at the Laboratory of Lipid Biochemistry and Laboratory of
Cellular Metabolism, but also since 1991 by the members of Laboratory of Cal-
cium Binding Proteins [Filipek et al. 1991; 1995; Filipek & Wojda 1996]. During
this time the important discoveries have been made concerning the annexin
structure, potential ligands and nally their cellular and organismal functions.
They are listed in table 2. It should be underlined that structural features, bio-
chemical and biophysical properties of AnxA6 together with its functions have
been well characterized in vitro and in cellular systems mainly on the basis of
two fundamental discoveries – nucleotide binding properties of AnxA6 [Ban-
dorowicz-Pikula & Awasthi 1997; Bandorowicz-Pikula et al. 1997a; 1997b; 1999;
Bandorowicz-Pikula 1998; Bandorowicz-Pikula & Pikula 1998; Danieluk et al.
1999] and its interaction with cholesterol and cholesterol enriched biological
membranes in a calcium- and pH-dependent manner [Sztolsztener et al. 2010;
2012; Domon et al. 2010; 2011; 2013a,b]. It should be stressed that cholesterol
binding properties of AnxA6, characterized by us on the basis of a series of
in vivo and ex vivo experiments, have been extensively studied in many other
laboratories around the world. The obtained results suggest that AnxA6 acts
as a multifunctional scaffold protein and is able to recruit vast number of sig-
Postępy Biochemii 64 (2–3) 2018 191
naling proteins, modulates cholesterol transport and its
distribution within the cell, and also regulates membrane
transport through actin dynamics. These activities suggest
that AnxA6 may contribute to the formation of specic
protein complexes and membrane domains relevant in sig-
nal transduction, cholesterol homeostasis and endo-/exo-
cytosis [Grewal et al. 2017].
MEMBRANE-RELATED FUNCTIONS OF ANNEXIN A6
MEBRANE LATERAL ORGANIZATION
Annexin A6, as a cholesterol binding and multifunc-
tional scaffold protein plays a crucial role in cell motility
[Hayes et al. 2004; Monastyrskaya et al. 2009; Grewal et al.
2017] and is implicated also in cell signaling [Koese et al.
2013; Hoque et al. 2014; Qi et al. 2015; Cornely et al. 2016;
Raouf et al. 2018]. Moreover, annexin A6 has been report-
ed to regulate a formation of multifunctional signaling
complexes at the membranes, affect membrane lateral or-
ganization, or inuence cholesterol metabolism and dis-
tribution, but also to participate in the vesicular transport
both in endo- and exocytosis [Cubells et al. 2007; 2008;
Enrich et al. 2014; Garcia-Melero et al. 2016; Cairns et al.
2017]. In addition, it has been shown that AnxA6 recruit-
ed to the plasma membrane is able to affect membrane re-
modelling, e.g. upregulated AnxA6 in the cell decreased
plasma membrane order through the regulation of cellu-
lar cholesterol homeostasis and its interaction with the
actin cytoskeleton in the living cells [Alvarez-Guaita et
al. 2015]. Strong experimental evidence has been accu-
mulated that AnxA6 due to its unique, among annexins,
structure affecting the distribution of cell specic surface
receptors, recruits the interaction partners and simultane-
ously bridges specialized membrane domains with corti-
cal actin surrounding activated receptors [Cornely et al.
2011].
MEBRANE REPAIR
The features of AnxA6 described above may be further
extended to the observations suggesting its participation
Table 1. Properties of mammalian annexins revealed on the basis of analyses of knockout animals.
Annexin Gene
encodingMW (Da) Total aa C-terminal core domain Some proposed functions*
Annexin A1 ANXA1 38,714 346 4 repeat domains
anti- or pro-inammatory responses, wound closure, epithelial
motility, cancer cell metastasis, insulin secretion, cell fusion,
vesicular transport, cell signaling, uptake of viruses
Annexin A2 ANXA2 40,41138,604 357
339 4 repeat domains cancer cell metastasis, brinolysis, pathogen
recognition, defense against bacterial infection
Annexin A3 ANXA3 36,375 323 4 repeat domains nd
Annexin A4 ANXA4 35,883 321 4 repeat domains cardiomyocyte signaling, integrity of urothelium
Annexin A5 ANXA5 35,937 320 4 repeat domains biomineralization, thrombosis, angiogenesis,
recognition of apoptotic cells
Annexin A6 ANXA6 75,873
72,423
673
641
8 repeat domains
and linker
calcium homeostasis, plasma membrane
organization, membrane repair, gluconeogenesis,
biomineralization, chondrocyte differentiation
Annexin A7 ANXA8 52,739
50,316
488
466 4 repeat domains cardiac contraction and remodeling, insulin
secretion, cell proliferation
Annexin A8 ANXA9 36,881 327 4 repeat domains nd
Annexin A9 ANXA10 38,364 345 4 repeat domains nd
Annexin A10 ANXA11 37,278 324 4 repeat domains nd
Annexin A11 ANXA12 54,390 505 4 repeat domains nd
Annexin A13 ANXA13 35,415
39,744
316
357 4 repeat domains nd
Information taken from https://www.uniprot.org, www.ncbi.nlm.nih.gov/protein/. *Some intra and extracellular
functions suggested on the basis of the experiments performed using knockout animals, reviewed in [Grewal
et al. 2016; Schloer et al. 2018]. Abbreviations: aa – amino acid residue, nd – not determined.
192 www.postepybiochemii.pl
in membrane repair mechanisms. Efcient cell membrane
repair mechanisms are essential for maintaining mem-
brane integrity and, thus, for cell life [Lauritzen et al.
2015; Demonbreun et al. 2016; Boye et al. 2017].
Initially, it has been observed that AnxA1 is involved
in the repair of plasmalemmal lesions induced by bacteri-
al toxins. Furthermore, highly Ca2+-sensitive AnxA6, that
responds faster to [Ca2+]i elevation than AnxA1, promotes
formation of lesions, and therefore is able to react to a
limited and sustained membrane injury [Potez et al. 2011].
The AnxA6 contribution to membrane repair mechanism
has been further elaborated on the basis of in vitro obser-
vations that AnxA4 and AnxA6 involved in plasma mem-
brane repair cause rapid closure of micron-size holes in
membranes. It has been demonstrated that AnxA4 binds
to membranes and generates curvature force, whereas
AnxA6 induces constriction force. In cells, plasma mem-
brane injury and concomitant Ca2+ inux result in AnxA4
recruitment to the vicinity of membrane wound edges.
Then, homo-trimerization of AnxA4 leads to membrane
curvature near the edges. Mediated by AnxA6 constric-
tion force is responsible for pulling the wound edges to-
gether for membrane fusion and nal repair [Boye et al.
2017]. In agreement are observations performed by means
of whole genome sequencing and RNA sequencing which
identied AnxA6 on the mouse model of muscular dys-
trophy associated with cardiomyopathy. Its truncated
version called ANXA6N32 was found to be responsible
for disrupting the whole AnxA6-rich cap and the associ-
ated (surrounding) repair zone at the site of sarcolemma
disruption, resulting in a membrane leak, characteristic
for muscular dystrophy [Swaggart et al. 2014].
VESICULAR TRANSPORT
AnxA6 features allowed many investigators to think
about this protein as a potential modulator of vesicular
transport events. It has been suggested that AnxA6 is impli-
cated in endocytosis, especially at the stage of fusion of au-
tophagosomes with endocytic compartment in hepatocytes
[Tebar et al. 2014; Enrich et al. 2017]. Moreover, AnxA6 high-
ly expressed in smooth muscles, hepatocytes, endothelial
cells and cardiomyocytes, has been found to affect various
stages of endocytotic route of cholesterol transport [Cubells
et al. 2007; Enrich et al. 2011; Reverter et al. 2011; Rentero et
al. 2018].
In addition, AnxA6 was found to participate in choles-
terol storage and the control of late endosomal cholesterol
levels, that modulate integrin recycling and cell migration
[Garcia-Melero et al. 2018], as well as inuenza A replica-
tion and propagation [Musiol et al. 2013]. AnxA6 has also
been linked to triglyceride storage in adipocytes [Cairns et
al. 2017].
Participation of annexins in exocytosis was rst postu-
lated almost 30 years ago [Creutz 1992]. Further studies
has revealed, that the number of observations suggesting
functioning of AnxA6 in exocytosis is limited. Investiga-
tors, however, agree that this multifunctional protein plays
a regulatory role in membrane trafcking during exocytosis
too [Enrich et al. 2017; Cairns et al. 2018].
CONCLUDING REMARKS
We have actively contributed to experiments, results
of which show that AnxA6 is an exceptional member
Table 2. Properties and potential cellular functions of annexin A6 studied in the Laboratory of Lipid Biochemistry.
Feature Description References
Cellular localization Plasma membrane, lysosomes and endosomes, matrix vesicles
[Bandorowicz et al. 1992; Balcerzak
et al. 2008; Strzelecka-Kiliszek et al.
2008; Sztolsztener et al. 2010; 2012;
Cmoch et al. 2011; Bottini et al. 2018]
Membrane binding properties
and ion channel-like activity
Calcium dependent binding to plasma
membrane phosphatidylserine
A pH and calcium dependent interaction
of AnxA6 with cholesterol, identication of
cholesterol binding domain in AnxA6
Mechanism of folding of AnxA6 in membranes at acidic pH
[Bandorowicz-Pikula et al. 1996]
[Domon et al. 2010; 2011; 2013a; 2013b]
[Golczak et al. 2001; 2004; Pikula
2003; Buzhynskyy et al. 2009]
Nucleotide binding properties
Identication of a putative consensus sequence
for the nucleotide-binding site in AnxA6
GTP-induced ion channel activity of AnxA6
[Kirilenko et al. 2002; 2006; Bandorowicz-
Pikula 2003; Bandorowicz-Pikula et al. 2003]
Intracellular functions
Lateral organization of plasma membrane – microdomains
Transport and storage of cholesterol
Catecholamine and interleukin-2 secretion
Biomineralization and matrix vesicles biogenesis
[Domon et al. 2012]
[Bandorowicz-Pikula et al. 2012]
[Podszywalow-Bartnicka et al. 2007;
2010; Strzelecka-Kiliszek et al. 2008]
Pathogenesis Lipid storage in Niemann-Pick type C disease
associated with mitochondrial dysfunction [Wos et al. 2016; 2018]
Postępy Biochemii 64 (2–3) 2018 193
of the annexin family of proteins resembling genuine
cholesterol-interacting proteins. Our studies indicate
particularly that AnxA6 intracellular localization and
membrane binding at low pH is determined by choles-
terol. Although, the overall picture of possible AnxA6
functions still requires further studies to identify/clari-
fy/completely unveil physiological and/or pathological
processes involving AnxA6 ability to change membrane
permeability to ions or mechanisms of membrane repair.
To sum up, step should be taken to elucidate the overall
importance of AnxA6 for the whole organism as it may
form specic target to identify and cure human diseases
in which AnxA6 may play a signicant role.
REFERENCES
Alvarez-Guaita A, Vilà de Muga S, Owen DM, Williamson D, Magenau
A, García-Melero A, Reverter M, Hoque M, Cairns R, Cornely R, Tebar
F, Grewal T, Gaus K, Ayala-Sanmartín J, Enrich C, Rentero C (2015)
Evidence for annexin A6-dependent plasma membrane remodelling
of lipid domains. Br J Pharmacol 172: 1677-1690
Balcerzak M, Malinowska A, Thouverey C, Sekrecka A, Dadlez M, Buchet
R, Pikula S (2008) Proteome analysis of matrix vesicles isolated from
femurs of chicken embryo. Proteomics 8: 192-205
Bandorowicz-Pikula J (2003) The nucleotide face of annexins. In: Annex-
ins: Biological Importance and Annexin-Related Pathologies (Bandor-
owicz-Pikula J, ed.), pp. 234-256, Kluwer Academic/Plenum Publish-
ers, New York, NY & Landes Bioscience, Georgetown, TX
Bandorowicz-Pikula J (1998) A nucleotide-binding domain of porcine
liver annexin VI. Proteolysis of annexin VI labelled with 8-azido-ATP,
purication by afnity chromatography on ATP-agarose, and uores-
cence studies. Mol Cell Biochem 181: 11-20
Bandorowicz-Pikula J, Awasthi YC (1997) Interaction of annexins IV and
VI with ATP. An alternative mechanism by which a cellular function
of these calcium- and membrane-binding proteins is regulated. FEBS
Lett 409: 300-306
Bandorowicz-Pikula J, Pikula S (1998) Modulation of annexin VI-driven
aggregation of phosphatidylserine liposomes by ATP. Biochimie 80:
613-620
Bandorowicz J, Pikuła S, Sobota A (1992) Annexins IV (p32) and VI (p68)
interact with erythrocyte membrane in a calcium-dependent manner.
Biochim Biophys Acta. 1105: 201-206
Bandorowicz-Pikula J, Sikorski AF, Bialkowska K, Sobota A (1996) Inter-
action of annexins IV and VI with phosphatidylserine in the presence
of Ca2+: monolayer and proteolytic study. Mol Membr Biol 13: 241-250
Bandorowicz-Pikuła J, Wrzosek A, Pikuła S, Awasthi YC (1997a) Fluo-
rescence spectroscopic studies on interactions between liver annexin
VI and nucleotides - a possible role for a tryptophan residue. Eur J
Biochem 248: 238-244
Bandorowicz-Pikula J, Wrzosek A, Makowski P, Pikula S (1997b) The
relationship between the binding of ATP and calcium to annexin IV.
Effect of nucleotide on the calcium-dependent interaction of annexin
with phosphatidylserine. Mol Membr Biol 14: 179-186
Bandorowicz-Pikuła J, Wrzosek A, Danieluk M, Pikula S, Buchet R (1999)
ATP-Binding site of annexin VI characterized by photochemical re-
lease of nucleotide and infrared difference spectroscopy. Biochem Bio-
phys Res Commun 263: 775-779
Bandorowicz-Pikula J, Buchet R, Pikula S (2001) Annexins as nucleotide-
binding proteins: facts and speculations. Bioessays 23: 170-178
Bandorowicz-Pikula J, Kirilenko A, van Deursen R, Golczak M, Kühnel
M, Lancelin JM, Pikula S, Buchet R (2003) A putative consensus se-
quence for the nucleotide-binding site of annexin A6. Biochemistry 42:
9137-9146
Bandorowicz-Pikuła J, Woś M, Pikuła S (2012) Do annexins participate
in lipid messenger mediated intracellular signaling? A question revis-
ited. Mol Membr Biol 29: 229-242
Bottini M, Mebarek S, Anderson KL, Strzelecka-Kiliszek A, Bozycki L,
Simão AMS, Bolean M, Ciancaglini P, Bandorowicz-Pikula J, Pikula S,
Magne D, Volkmann N, Hanein D, Millán JL, Buchet R (2018) Matrix
vesicles from chondrocytes and osteoblasts: Their biogenesis, proper-
ties, functions and biomimetic models. Biochim Biophys Acta 1862:
532-546
Boye TL, Maeda K, Pezeshkian W, Sønder SL, Haeger SC, Gerke V, Si-
monsen AC, Nylandsted J (2017) Annexin A4 and A6 induce mem-
brane curvature and constriction during cell membrane repair. Nat
Commun 8: 1623
Buzhynskyy N, Golczak M, Lai-Kee-Him J, Lambert O, Tessier B, Gou-
nou C, Bérat R, Simon A, Granier T, Chevalier JM, Mazères S, Ban-
dorowicz-Pikula J, Pikula S, Brisson AR (2009) Annexin-A6 presents
two modes of association with phospholipid membranes. A combined
QCM-D, AFM and cryo-TEM study. J Struct Biol 168: 107-116
Cairns R, Alvarez-Guaita A, Martínez-Saludes I, Wason SJ, Hanh J, Na-
garajan SR, Hosseini-Beheshti E, Monastyrskaya K, Hoy AJ, Buechler
C, Enrich C, Rentero C, Grewal T (2017) Role of hepatic Annexin A6 in
fatty acid-induced lipid droplet formation. Exp Cell Res 358: 397-410
Cairns R, Fischer AW, Blanco-Munoz P, Alvarez-Guaita A, Meneses-Salas
E, Egert A, Buechler C, Hoy AJ, Heeren J, Enrich C, Rentero C, Grewal
T (2018) Altered hepatic glucose homeostasis in AnxA6-KO mice fed a
high-fat diet. PLoS One 13: e0201310
Cmoch A, Strzelecka-Kiliszek A, Palczewska M, Groves P, Pikula S (2011)
Matrix vesicles isolated from mineralization-competent Saos-2 cells
are selectively enriched with annexins and S100 proteins. Biochem
Biophys Res Commun 412: 683-687
Cornely R, Rentero C, Enrich C, Grewal T, Gaus K (2011) Annexin A6 is
an organizer of membrane microdomains to regulate receptor localiza-
tion and signalling. IUBMB Life 63: 1009-1017
Cornely R, Pollock AH, Rentero C, Norris SE, Alvarez-Guaita A, Grewal
T, Mitchell T, Enrich C, Moss SE, Parton RG, Rossy J, Gaus K (2016)
Annexin A6 regulates interleukin-2-mediated T-cell proliferation. Im-
munol Cell Biol 94: 543-553
Creutz CE (1992) The annexins and exocytosis. Science 258: 924-931
Cubells L, Vilà de Muga S, Tebar F, Wood P, Evans R, Ingelmo-Torres M,
Calvo M, Gaus K, Pol A, Grewal T, Enrich C (2007) Annexin A6-in-
duced alterations in cholesterol transport and caveolin export from the
Golgi complex. Trafc 8: 1568-1589
Cubells L, Vilà de Muga S, Tebar F, Bonventre JV, Balsinde J, Pol A, Gre-
wal T, Enrich C (2008) Annexin A6-induced inhibition of cytoplasmic
phospholipase A2 is linked to caveolin-1 export from the Golgi. J Biol
Chem 283: 10174-10183
Danieluk M, Pikuła S, Bandorowicz-Pikuła J (1999) Annexin VI interacts
with adenine nucleotides and their analogs. Biochimie 81: 717-726
Demonbreun AR, Quattrocelli M, Bareeld DY, Allen MV, Swanson KE,
McNally EM (2016) An actin-dependent annexin complex mediates
plasma membrane repair in muscle. J Cell Biol 213: 705-718
Domon M, Matar G, Strzelecka-Kiliszek A, Bandorowicz-Pikula J, Pikula
S, Besson F (2010) Interaction of annexin A6 with cholesterol rich mem-
branes is pH-dependent and mediated by the sterol OH. J Colloid In-
terf Sci 346: 436-441
Domon M, Besson F, Bandorowicz-Pikula J, Pikula S (2011) Annexin A6
is recruited into lipid rafts of Niemann-Pick type C disease broblasts
in a Ca2+-dependent manner. Biochem Biophys Res Commun 405: 192-
196
Domon M, Nasir MN, Matar G, Pikula S, Besson F, Bandorowicz-Pikula
J (2012) Annexins as organizers of cholesterol- and spingomyelin-en-
riched membrane microdomains in Niemann-Pick type C disease. Cell
Mol Life Sci 69: 1773-1785
Domon MM, Besson F, Tylki-Szymanska A, Bandorowicz-Pikula J, Pikula
S (2013a) Interaction of AnxA6 with isolated and articial lipid micro-
domains; importance of lipid composition and calcium content. Molec
Biosyst 9: 668-676
Domon MM, Nasir MN, Pikula S, Besson F (2013b) Inuence of the
524-VAAEIL-529 sequence of annexins A6 in their interfacial behavior
and interaction with lipid monolayers. J Colloid Interf Sci 403: 99-104
194 www.postepybiochemii.pl
Enrich C, Rentero C, de Muga SV, Reverter M, Mulay V, Wood P, Koese
M, Grewal T (2011) Annexin A6-linking Ca2+ signaling with choles-
terol transport. Biochim Biophys Acta 1813: 935-947
Enrich C, Rentero C, Grewal T (2017) Annexin A6 in the liver: from the
endocytic compartment to cellular physiology. Biochim Biophys Acta
1864: 933-946
Filipek A, Wojda U (1996) p30, a novel protein target of mouse calcyclin
(S100A6). Biochem J 320: 585-587
Filipek A, Gerke V, Weber K, Kuźnicki J (1991) Characterization of the
cell-cycle-regulated protein calcyclin from Ehrlich ascites tumor cells.
Identication of two binding proteins obtained by Ca2+-dependent af-
nity chromatography. Eur J Biochem 195: 795-800
Filipek A, Wojda U, Leśniak W (1995) Interaction of calcyclin and its
cyanogen bromide fragments with annexin II and glyceraldehyde
3-phosphate dehydrogenase. Int J Biochem Cell Biol 27: 1123-1131
García-Melero A, Reverter M, Hoque M, Meneses-Salas E, Koese M, Con-
way JR, Johnsen CH, Alvarez-Guaita A, Morales-Paytuvi F, Elmaghra-
bi YA, Pol A, Tebar F, Murray RZ, Timpson P, Enrich C, Grewal T,
Rentero C (2016) Annexin A6 and late endosomal cholesterol modu-
late integrin recycling and cell migration. J Biol Chem 291: 1320-1335
Gerke V, Creutz CE, Moss SE (2005) Annexins: linking Ca2+ signaling to
membrane dynamics. Nat Rev Mol Cell Biol 6: 449-461
Golczak M, Kicinska A, Bandorowicz-Pikula J, Buchet R, Szewczyk A, Pi-
kula S (2001) Acidic pH-induced folding of annexin VI is a prerequisite
for its insertion into lipid bilayers and formation of ion channels by the
protein molecules. FASEB J 15: 1083-1085
Golczak M, Kirilenko A, Bandorowicz-Pikula J, Desbat B, Pikula S (2004)
Structure of human annexin a6 at the air-water interface and in a mem-
brane-bound state. Biophys J 87: 1215-1226
Grewal T, Wason SJ, Enrich C, Rentero C (2016) Annexins - insights from
knockout mice. Biol Chem 397: 1031-1053
Grewal T, Hoque M, Conway JRW, Reverter M, Wahba M, Beevi SS,
Timpson P, Enrich C, Rentero C (2017) Annexin A6 - A multifunctional
scaffold in cell motility. Cell Adh Migr 11: 288-304
Hayes MJ, Rescher U, Gerke V, Moss SE (2004) Annexin-actin interac-
tions. Trafc 5: 571-576
Hoque M, Rentero C, Cairns R, Tebar F, Enrich C, Grewal T (2014) Annex-
ins - scaffolds modulating PKC localization and signaling. Cell Signal
26: 1213-1225
Kapustin AN, Shanahan CM (2016) Emerging roles for vascular smooth
muscle cell exosomes in calcication and coagulation. J Physiol 594:
2905-2914
Kirilenko A, Golczak M, Pikula S, Buchet R, Bandorowicz-Pikula J (2002)
GTP-induced membrane binding and ion channel activity of annexin
VI: is annexin VI a GTP biosensor? Biophys J 82: 2737-2745
Kirilenko A, Pikula S, Bandorowicz-Pikula J (2006) Effects of mutagenesis
of W343 in human annexin A6 isoform 1 on its interaction with GTP:
nucleotide-induced oligomer formation and ion channel activity. Bio-
chemistry 45: 4965-4973
Kodavali PK, Dudkiewicz M, Pikuła S, Pawłowski K (2014) Bioinformat-
ics analysis of bacterial annexins-putative ancestral relatives of eukar-
yotic annexins. PLoS One 9: e85428
Koese M, Rentero C, Kota BP, Hoque M, Cairns R, Wood P, Vilà de Muga
S, Reverter M, Alvarez-Guaita A, Monastyrskaya K, et al. (2013) An-
nexin A6 is a scaffold for PKCα to promote EGFR inactivation. Onco-
gene 32: 2858-5872
Lauritzen SP, Boye TL, Nylandsted J (2015) Annexins are instrumental
for efcient plasma membrane repair in cancer cells. Semin Cell Dev
Biol 45: 32-38
Minashima T, Kirsch T (2018) Annexin A6 regulates catabolic events in ar-
ticular chondrocytes via the modulation of NF-κB and Wnt/ß-catenin
signaling. PLoS One 13: e0197690
Monastyrskaya K, Babiychuk EB, Hostettler A, Wood P, Grewal T, Drae-
ger A (2009) Plasma membrane-associated annexin A6 reduces Ca2+
entry by stabilizing the cortical actin cytoskeleton. J Biol Chem 284:
17227-17242
Morgan RO, Martin-Almedina S, Iglesias JM, Gonzalez-Florez MI, Fer-
nandez MP (2004) Evolutionary perspective on annexin calcium-bind-
ing domains. Biochim Biophys Acta 1742: 133-140
Morgan RO, Martin-Almedina S, Garcia M, Jhoncon-Kooyip J, Fernandez
MP (2006) Deciphering function and mechanism of calcium-binding
proteins from their evolutionary imprints. Biochim Biophys Acta 1763:
1238-1249
Musiol A, Gran S, Ehrhardt C, Ludwig S, Grewal T, Gerke V, Rescher U
(2013) Annexin A6-balanced late endosomal cholesterol controls inu-
enza a replication and propagation. MBio 4: e00608-613
Pikula S (2003) Acidic pH-induced ion channels formed by annexin
VI – transformation from soluble to membrane integral protein. In:
Annexins: Biological Importance and Annexin-Related Pathologies
(Bandorowicz-Pikula J, ed.), pp. 182-195, Kluwer Academic/Plenum
Publishers, New York, NY & Landes Bioscience, Georgetown, TX
Podszywalow-Bartnicka P, Strzelecka-Kiliszek A, Bandorowicz-Pikula J,
Pikula S (2007) Calcium- and proton-dependent relocation of annexin
A6 in Jurkat T cells stimulated for interleukin-2 secretion. Acta Bio-
chim Pol 54: 261-271
Podszywalow-Bartnicka P, Kosiorek M, Piwocka K, Sikora E, Zablocki K,
Pikula S (2010) Role of annexin A6 isoforms in catecholamine secretion
by PC12 cells: distinct inuence on calcium response. J Cell Biochem
111 168-178
Potez S, Luginbühl M, Monastyrskaya K, Hostettler A, Draeger A, Ba-
biychuk EB (2011) Tailored protection against plasmalemmal injury by
annexins with different Ca2+ sensitivities. J Biol Chem 286: 17982-17991
Qi H, Liu S, Guo C, Wang J, Greenaway FT, Sun MZ (2015) Role of an-
nexin A6 in cancer. Oncol Lett 10: 1947-1952
Raouf R, Lolignier S, Sexton JE, Millet Q, Santana-Varela S, Biller A, Fuller
AM, Pereira V, Choudhary JS, Collins MO, Moss SE, Lewis R, Tordo
J, Henckaerts E, Linden M, Wood JN (2018) Inhibition of somatosen-
sory mechanotransduction by annexin A6. Sci Signal 11: doi: 10.1126/
scisignal.aao2060
Rentero C, Blanco-Muñoz P, Meneses-Salas E, Grewal T, Enrich C (2018)
Annexins - coordinators of cholesterol homeostasis in endocytic path-
ways. Int J Mol Sci 9: doi: 10.3390/ijms19051444
Reverter M, Rentero C, de Muga SV, Alvarez-Guaita A, Mulay V, Cairns
R, Wood P, Monastyrskaya K, Pol A, Tebar F, Blasi J, Grewal T, Enrich
C (2011) Cholesterol transport from late endosomes to the Golgi reg-
ulates t-SNARE trafcking, assembly, and function. Mol Biol Cell 22:
4108-4102
Schloer S, Pajonczyk D, Rescher U (2018) Annexins in translational re-
search: hidden treasures to be found. Int J Mol Sci 19: doi: 10.3390/
ijms19061781
Sobota A, Bandorowicz J, Jezierski A, Sikorski AF (1993) The effect of an-
nexin IV and VI on the uidity of phosphatidylserine/phosphatidyl-
choline bilayers studied with the use of 5-deoxylstearate spin label.
FEBS Lett 315: 178-182
Strzelecka-Kiliszek A, Buszewska ME, Podszywalow-Bartnicka P, Pikula
S, Otulak K, Buchet R, Bandorowicz-Pikula J (2008) Calcium- and pH-
dependent localization of annexin A6 isoforms in Balb/3T3 broblasts
reecting their potential participation in vesicular transport. J Cell Bio-
chem 104: 418-434
Swaggart KA, Demonbreun AR, Vo AH, Swanson KE, Kim EY, Fahren-
bach JP, Holley-Cuthrell J, Eskin A, Chen Z, Squire K, Heydemann
A, Palmer AA, Nelson SF, McNally EM (2014) Annexin A6 modies
muscular dystrophy by mediating sarcolemmal repair. Proc Natl Acad
Sci USA 111: 6004-6009
Sztolsztener ME, Strzelecka-Kiliszek A, Pikula S, Tylki-Szymanska A,
Bandorowicz-Pikula J (2010) Cholesterol as a factor regulating intracel-
lular localization of annexin A6 in Niemann-Pick type C human bro-
blasts. Arch Biochem Biophys 493: 221-233
Sztolsztener ME, Dobrzyn A, Pikula S, Tylki-Szymanska A, Bandorowicz-
Pikula J (2012) Impaired dynamics of late endosome/lysosome com-
partment in human Niemann-Pick type C skin broblasts carrying
mutation in NPC1 gene. Molec BioSyst 8: 1197-1205
Postępy Biochemii 64 (2–3) 2018 195
Aneksyna A6, białko wiążące cholesterol i nukleotydy, uczestniczące
w naprawie błon biologicznych i w transporcie pęcherzykowym
Joanna Bandorowicz-Pikuła*, Agnieszka Kinga Seliga
Pracownia Metabolizmu Komórki, Instytut Biologii Doświadczalnej im. Marcelego Nenckiego PAN, ul. Pasteura 3, 02-093 Warszawa
*e-mail: j.bandorowicz-pikula@nencki.gov.pl
Słowa kluczowe: aneksyna A6, cholesterol, naprawa błon biologicznych, transport pęcherzykowy
STRESZCZENIE
Aneksyny, rodzina białek wiążących jony wapnia i błony biologiczne, były badane w Instytucie Biologii Doświadczalnej im. Marcelego
Nenckiego w Warszawie od wczesnych lat 90. XX wieku. Szczególną uwagę poświęcono strukturze aneksyn, potencjalnym ligandom tych
białek oraz ich funkcji, np. w procesie biomineralizacji zachodzącym w normie i w stanach patologicznych. Wyniki badań prowadzonych w
wielu laboratoriach na świecie wskazują, że aneksyny odgrywają bardzo ważną rolę w organizacji błon biologicznych. W tym artykule prze-
glądowym opisujemy jednego z największych pod względem masy cząsteczkowej przedstawiciela rodziny aneksyn, aneksynę A6 (AnxA6),
białko wykazujące zdolność wiązania się z cholesterolem i nukleotydami oraz zmieniające przepuszczalność błony dla jonów w odpowiedzi
na obniżenie wewnątrzkomórkowego pH. Dodatkowo, opisano funkcje AnxA6, takie jak udział w tworzeniu mikrodomen błonowych, w
naprawie uszkodzeń błony plazmatycznej oraz w regulacji transportu pęcherzykowego.
Tebar F, Gelabert-Baldrich M, Hoque M, Cairns R, Rentero C, Pol A, Gre-
wal T, Enrich C (2014) Annexins and endosomal signaling. Methods
Enzymol 535: 55-74
Woś M, Szczepanowska J, Pikuła S, Tylki-Szymańska A, Zabłocki K,
Bandorowicz-Pikuła J (2016) Mitochondrial dysfunction in broblasts
derived from patients with Niemann-Pick type C disease. Arch Bio-
chem Biophys 593: 50-59
Woś M, Komiażyk M, Pikuła S, Tylki-Szymańska A, Bandorowicz-Pikuła
J (2018) Activation of mTOR kinase and GSK-3β accompanies abnor-
mal accumulation of cholesterol in broblasts from Niemann-Pick
type C patients. J Cell Biochem in press
... On [76], bone [77], and skeletal muscle [78]. ...
Article
Full-text available
The stable gastric pentadecapeptide BPC 157 counteracts various venous occlusion-induced syndromes. Summarized are all these arguments, in the Robert's cytoprotection concept, to substantiate the resolution of different major vessel occlusion disturbances, in particular ischemia-reperfusion injury following the Pringle maneuver and Budd-Chiari syndrome, which was obtained by BPC 157 therapy. Conceptually, there is a new point, namely, endothelium maintenance to epithelium maintenance (the recruitment of collateral blood vessels to compensate for vessel occlusion and reestablish blood flow or bypass the occluded or ruptured vessel). In this paper, we summarize the evidence of the native cytoprotective gastric pentadecapeptide BPC 157, which is stable in the human gastric juice, is a membrane stabilizer and counteracts gut-leaky syndrome. As a particular target, it is distinctive from the standard peptide growth factors, involving particular molecular pathways and controlling VEGF and NO pathways. In the early 1990s, BPC 157 appeared as a late outbreak of the Robert's and Szabo's cytoprotection-organoprotection concept, like the previous theoretical/practical breakthrough in the 1980s and the brain-gut axis and gut-brain axis. As the time went on, with its reported effects, it is likely most useful theory practical implementation and justification. Meantime, several reviews suggest that BPC 157, which does not have a lethal dose, has profound cytoprotective activity, used to be demonstrated in ulcerative colitis and multiple sclerosis trials. Likely, it may bring the theory to practical application, starting with the initial argument, no degradation in human gastric juice for more than 24 h, and thereby, the therapeutic effectiveness (including via a therapeutic per-oral regimen) and pleiotropic beneficial effects.
Article
Full-text available
The mitochondrial large-conductance calcium-activated potassium channel (mitoBKCa) is located in the inner mitochondrial membrane and seems to play a crucial role in cytoprotection. The mitoBKCa channel is regulated by many modulators, including activators, such as calcium ions and inhibitors, such as heme and its oxidized form hemin. Heme/hemin binds to the heme-binding motif (CXXCH) located between two RCK domains present in the mitochondrial matrix. In the present study, we used the patch-clamp technique in the outside-out configuration to record the activity of mitoBKCa channels. This allowed for the application of channel modulators to the intermembrane-space side of the mitoBKCa. We found that hemin applied in this configuration inhibits the activity of mitoBKCa. In addition, we proved that the observed hemin effect is specific and it is not due to its interaction with the inner mitochondrial membrane. Our data suggest the existence of a new potential heme/hemin binding site in the structure of the mitoBKCa channel located on the mitochondrial intermembrane space side, which could constitute a new way for the regulation of mitoBKCa channel activity.
Article
Full-text available
Ca ²⁺ -activated K ⁺ channels of intermediate conductance (IK) are frequently overexpressed in breast cancer (BC) cells, while IK channel depletion reduces BC cell proliferation and tumorigenesis. This raises the question, of whether and mechanistically how IK activity interferes with the metabolic activity and energy consumption rates, which are fundamental for rapidly growing cells. Using BC cells obtained from MMTV-PyMT tumor-bearing mice, we show that both, glycolysis and mitochondrial ATP-production are reduced in cells derived from IK-deficient breast tumors. Loss of IK altered the sub-/cellular K ⁺ - and Ca ²⁺ - homeostasis and mitochondrial membrane potential, ultimately resulting in reduced ATP-production and metabolic activity. Consequently, we find that BC cells lacking IK upregulate AMP-activated protein kinase activity to induce autophagy compensating the glycolytic and mitochondrial energy shortage. Our results emphasize that IK by modulating cellular Ca ²⁺ - and K ⁺ -dynamics contributes to the remodeling of metabolic pathways in cancer. Thus, targeting IK channel might disturb the metabolic activity of BC cells and reduce malignancy.
Article
Full-text available
Mitochondrial potassium channels control potassium influx into the mitochondrial matrix and thus regulate mitochondrial membrane potential, volume, respiration, and synthesis of reactive oxygen species (ROS). It has been found that pharmacological activation of mitochondrial potassium channels during ischemia/reperfusion (I/R) injury activates cytoprotective mechanisms resulting in increased cell survival. In cancer cells, the inhibition of these channels leads to increased cell death. Therefore, mitochondrial potassium channels are intriguing targets for the development of new pharmacological strategies. In most cases, however, the substances that modulate the mitochondrial potassium channels have a few alternative targets in the cell. This may result in unexpected or unwanted effects induced by these compounds. In our review, we briefly present the various classes of mitochondrial potassium (mitoK) channels and describe the chemical compounds that modulate their activity. We also describe examples of the multidirectional activity of the activators and inhibitors of mitochondrial potassium channels.
Article
ATP synthase (F1Fo) is a rotary molecular engine that harnesses energy from electrochemical-gradients across the inner mitochondrial membrane for ATP synthesis. Despite the accepted tenet that F1Fo transports exclusively H⁺, our laboratory has demonstrated that, in addition to H⁺, F1Fo ATP synthase transports a significant fraction of ΔΨm-driven charge as K⁺ to synthesize ATP. Herein, we utilize a computational modeling approach as a proof of principle of the feasibility of the core mechanism underlying the enhanced ATP synthesis, and to explore its bioenergetic consequences. A minimal model comprising the ‘core’ mechanism constituted by ATP synthase, driven by both proton (PMF) and potassium motive force (KMF), respiratory chain, adenine nucleotide translocator, Pi carrier, and K⁺/H⁺ exchanger (KHEmito) was able to simulate enhanced ATP synthesis and respiratory fluxes determined experimentally with isolated heart mitochondria. This capacity of F1Fo ATP synthase confers mitochondria with a significant energetic advantage compared to K⁺ transport through a channel not linked to oxidative phosphorylation (OxPhos). The K⁺-cycling mechanism requires a KHEmito that exchanges matrix K⁺ for intermembrane space H⁺, leaving PMF as the overall driving energy of OxPhos, in full agreement with the standard chemiosmotic mechanism. Experimental data of state 4➔3 energetic transitions, mimicking low to high energy demand, could be reproduced by an integrated computational model of mitochondrial function that incorporates the ‘core’ mechanism. Model simulations display similar behavior compared to the experimentally observed changes in ΔΨm, mitochondrial K⁺ uptake, matrix volume, respiration, and ATP synthesis during the energetic transitions at physiological pH and K⁺ concentration. The model also explores the role played by KHEmito in modulating the energetic performance of mitochondria. The results obtained support the available experimental evidence on ATP synthesis driven by K⁺ and H⁺ transport through the F1Fo ATP synthase.
Article
Full-text available
Cystic fibrosis is a hereditary disease that mainly affects secretory organs in humans. It is caused by mutations in the gene encoding CFTR with the most common phenylalanine deletion at position 508. CFTR is an anion channel mainly conducting Cl− across the apical membranes of many different epithelial cells, the impairment of which causes dysregulation of epithelial fluid secretion and thickening of the mucus. This, in turn, leads to the dysfunction of organs such as the lungs, pancreas, kidney and liver. The CFTR protein is mainly localized in the plasma membrane; however, there is a growing body of evidence that it is also present in the intracellular organelles such as the endosomes, lysosomes, phagosomes and mitochondria. Dysfunction of the CFTR protein affects not only the ion transport across the epithelial tissues, but also has an impact on the proper functioning of the intracellular compartments. The review aims to provide a summary of the present state of knowledge regarding CFTR localization and function in intracellular compartments, the physiological role of this localization and the consequences of protein dysfunction at cellular, epithelial and organ levels. An in-depth understanding of intracellular processes involved in CFTR impairment may reveal novel opportunities in pharmacological agents of cystic fibrosis.
Article
Full-text available
Potassium ions can cross both the outer and inner mitochondrial membranes by means of multiple routes. A few potassium-permeable ion channels exist in the outer membrane, while in the inner membrane, a multitude of different potassium-selective and potassium-permeable channels mediate K+ uptake into energized mitochondria. In contrast, potassium is exported from the matrix thanks to an H+/K+ exchanger whose molecular identity is still debated. Among the K+ channels of the inner mitochondrial membrane, the most widely studied is the ATP-dependent potassium channel, whose pharmacological activation protects cells against ischemic damage and neuronal injury. In this review, we briefly summarize and compare the different hypotheses regarding the molecular identity of this patho-physiologically relevant channel, taking into account the electrophysiological characteristics of the proposed components. In addition, we discuss the characteristics of the other channels sharing localization to both the plasma membrane and mitochondria.
Article
Full-text available
Mitochondria are bioenergetic organelles with a plethora of fundamental functions ranging from metabolism and ATP production to modulation of signaling events leading to cell survival or cell death. Ion channels located in the outer and inner mitochondrial membranes critically control mitochondrial function and, as a consequence, also cell fate. Opening or closure of mitochondrial ion channels allow the fine-tuning of mitochondrial membrane potential, ROS production, and function of the respiratory chain complexes. In this review, we critically discuss the intracellular regulatory factors that affect channel activity in the inner membrane of mitochondria and, indirectly, contribute to cell death. These factors include various ligands, kinases, second messengers, and lipids. Comprehension of mitochondrial ion channels regulation in cell death pathways might reveal new therapeutic targets in mitochondria-linked pathologies like cancer, ischemia, reperfusion injury, and neurological disorders.
Article
Full-text available
A large number of diverse mechanisms that lead to cytoprotection have been described to date. Perhaps, not surprisingly, the role of mitochondria in these phenomena is notable. In addition to being metabolic centers, due to their role in cell catabolism, ATP synthesis, and biosynthesis these organelles are triggers and/or end-effectors of a large number of signaling pathways. Their role in the regulation of the intrinsic apoptotic pathway, calcium homeostasis, and reactive oxygen species signaling is well documented. In this review, we aim to characterize the prospects of influencing cytoprotective mitochondrial signaling routes by natural substances of plant origin, namely, flavonoids (e.g., flavanones, flavones, flavonols, flavan-3-ols, anthocyanidins, and isoflavones). Flavonoids are a family of widely distributed plant secondary metabolites known for their beneficial effects on human health and are widely applied in traditional medicine. Their pharmacological characteristics include antioxidative, anticarcinogenic, anti-inflammatory, antibacterial, and antidiabetic properties. Here, we focus on presenting mitochondria-mediated cytoprotection against various insults. Thus, the role of flavonoids as antioxidants and modulators of antioxidant cellular response, apoptosis, mitochondrial biogenesis, autophagy, and fission and fusion is reported. Finally, an emerging field of flavonoid-mediated changes in the activity of mitochondrial ion channels and their role in cytoprotection is outlined.
Article
Full-text available
Mitochondrial ion channels are emerging oncological targets, as modulation of these ion-transporting proteins may impact on mitochondrial membrane potential, efficiency of oxidative phosphorylation and reactive oxygen production. In turn, these factors affect the release of cytochrome c, which is the point of no return during mitochondrial apoptosis. Many of the currently used chemotherapeutics induce programmed cell death causing damage to DNA and subsequent activation of p53-dependent pathways that finally leads to cytochrome c release from the mitochondrial inter-membrane space. The view is emerging, as summarized in the present review, that ion channels located in this organelle may account in several cases for the resistance that cancer cells can develop against classical chemotherapeutics, by preventing drug-induced apoptosis. Thus, pharmacological modulation of these channel activities might be beneficial to fight chemo-resistance of different types of cancer cells.
Article
Full-text available
Mechanically activated, slowly adapting currents in sensory neurons have been linked to noxious mechanosensation. The conotoxin NMB-1 (noxious mechanosensation blocker-1) blocks such currents and inhibits mechanical pain. Using a biotinylated form of NMB-1 in mass spectrometry analysis, we identified 67 binding proteins in sensory neurons and a sensory neuron–derived cell line, of which the top candidate was annexin A6, a membrane-associated calcium-binding protein. Annexin A6–deficient mice showed increased sensitivity to mechanical stimuli. Sensory neurons from these mice showed increased activity of the cation channel Piezo2, which mediates a rapidly adapting mechano-gated current linked to proprioception and touch, and a decrease in mechanically activated, slowly adapting currents. Conversely, overexpression of annexin A6 in sensory neurons inhibited rapidly adapting currents that were partially mediated by Piezo2. Furthermore, overexpression of annexin A6 in sensory neurons attenuated mechanical pain in a mouse model of osteoarthritis, a disease in which mechanically evoked pain is particularly problematic. These data suggest that annexin A6 can be exploited to inhibit chronic mechanical pain.
Article
Full-text available
The vertebrate annexin superfamily (AnxA) consists of 12 members of a calcium (Ca2+) and phospholipid binding protein family which share a high structural homology. In keeping with this hallmark feature, annexins have been implicated in the Ca2+-controlled regulation of a broad range of membrane events. In this review, we identify and discuss several themes of annexin actions that hold a potential therapeutic value, namely, the regulation of the immune response and the control of tissue homeostasis, and that repeatedly surface in the annexin activity profile. Our aim is to identify and discuss those annexin properties which might be exploited from a translational science and specifically, a clinical point of view.
Article
Full-text available
Annexin A6 (AnxA6) is expressed in articular chondrocytes at levels higher than in other mesenchymal cell types. However, the role of AnxA6 in articular chondrocytes is not known. Here we show that complete lack of AnxA6 functions resulted in increased ß-catenin activation in Wnt3a-treated murine articular chondrocytes, whereas AnxA6 expressing articular chondrocytes showed decreased ß-catenin activation. High expression of AnxA6 in human articular chondrocytes showed the highest inhibition of Wnt/ß-catenin signaling. Inhibition of Wnt/ß-catenin signaling activity by AnxA6 together with cytosolic Ca²⁺ was achieved by interfering with the plasma membrane association of the Wnt signaling complex. AnxA6 also affected the cross-talk between Wnt/ß-catenin signaling and NF-κB signaling by decreasing ß-catenin activity and increasing NF-κB activity in Wnt3a-, interleukin-1beta (IL-1ß)-, and combined Wnt3a/IL-1ß-treated cells. Wnt3a treatment increased the mRNA levels of catabolic markers (cyclooxygenase-2, interleukin-6, inducible nitric oxide synthase) to a much lesser degree than IL-1ß treatment in human articular chondrocytes, and decreased the mRNA levels of matrix metalloproteinase-13 (MMP-13) and articular cartilage markers (aggrecan, type II collagen). Furthermore, Wnt3a decreased the mRNA levels of catabolic markers and MMP-13 in IL-1ß-treated human articular chondrocytes. High expression of AnxA6 resulted in decreased mRNA levels of catabolic markers, and increased MMP-13 and articular cartilage marker mRNA levels in Wnt3a-treated human articular chondrocytes, whereas leading to increased mRNA levels of catabolic markers and MMP-13 in human articular chondrocytes treated with IL-1ß, or combined Wnt3a and IL-1ß. Our findings define a novel role for AnxA6 in articular chondrocytes via its modulation of Wnt/ß-catenin and NF-κB signaling activities and the cross-talk between these two signaling pathways.
Article
Full-text available
The spatiotemporal regulation of calcium (Ca2+) storage in late endosomes (LE) and lysosomes (Lys) is increasingly recognized to influence a variety of membrane trafficking events, including endocytosis, exocytosis, and autophagy. Alterations in Ca2+ homeostasis within the LE/Lys compartment are implicated in human diseases, ranging from lysosomal storage diseases (LSDs) to neurodegeneration and cancer, and they correlate with changes in the membrane binding behaviour of Ca2+-binding proteins. This also includes Annexins (AnxA), which is a family of Ca2+-binding proteins participating in membrane traffic and tethering, microdomain organization, cytoskeleton interactions, Ca2+ signalling, and LE/Lys positioning. Although our knowledge regarding the way Annexins contribute to LE/Lys functions is still incomplete, recruitment of Annexins to LE/Lys is greatly influenced by the availability of Annexin bindings sites, including acidic phospholipids, such as phosphatidylserine (PS) and phosphatidic acid (PA), cholesterol, and phosphatidylinositol (4,5)-bisphosphate (PIP2). Moreover, the cytosolic portion of LE/Lys membrane proteins may also, directly or indirectly, determine the recruitment of Annexins to LE. Strikingly, within LE/Lys, AnxA1, A2, A6, and A8 differentially contribute to cholesterol transport along the endocytic route, in particular, cholesterol transfer between LE and other compartments, positioning Annexins at the centre of major pathways mediating cellular cholesterol homeostasis. Underlying mechanisms include the formation of membrane contact sites (MCS) and intraluminal vesicles (ILV), as well as the modulation of LE-cholesterol transporter activity. In this review, we will summarize the current understanding how Annexins contribute to influence LE/Lys membrane transport and associated functions.
Article
Full-text available
Annexins are a family of proteins that bind to phospholipids in a calcium-dependent manner. Earlier studies implicated Annexin A6 (AnxA6) to inhibit secretion and participate in the organization of the extracellular matrix (ECM). We recently showed that elevated AnxA6 levels significantly reduced secretion of the ECM protein fibronectin (FN). Since FN is directly linked to the ability of cells to migrate, this prompted us to investigate the role of AnxA6 in cell migration. Upregulation of AnxA6 in several cell models was associated with reduced cell migration in wound healing, individual cell tracking as well as transwell and 3D-migration/invasion assays. The reduced ability of AnxA6 expressing cells to migrate was associated with decreased cell surface expression of αVβ3 and α5β1 integrins, both FN receptors. Mechanistically, we found that elevated AnxA6 levels interfered with syntaxin-6 (Stx6)-dependent recycling of integrins to the cell surface. AnxA6 overexpression caused mislocalization and accumulation of Stx6 and integrins in recycling endosomes (RE) while siRNA-mediated AnxA6 knockdown did not modify the trafficking of integrins. Given our recent findings that inhibition of cholesterol export from late endosomes (LE) inhibits Stx6-dependent integrin recycling, and that elevated AnxA6 levels cause LE-cholesterol accumulation, we propose that AnxA6 and blockage of LE-cholesterol transport is critical for endosomal function required for Stx6-mediated recycling of integrins in cell migration.
Article
Annexin A6 (AnxA6) belongs to a highly conserved protein family characterized by their calcium (Ca(2+)) -dependent binding to phospholipids. Over the years, immunohistochemistry, subcellular fractionations, and live cell microscopy established that AnxA6 is predominantly found at the plasma membrane and endosomal compartments. In these locations, AnxA6 acts as a multifunctional scaffold protein, recruiting signaling proteins, modulating cholesterol and membrane transport and influencing actin dynamics. These activities enable AnxA6 to contribute to the formation of multifactorial protein complexes and membrane domains relevant in signal transduction, cholesterol homeostasis and endo-/exocytic membrane transport. Hence, AnxA6 has been implicated in many biological processes, including cell proliferation, survival, differentiation, inflammation, but also membrane repair and viral infection. More recently, we and others identified roles for AnxA6 in cancer cell migration and invasion. This review will discuss how the multiple scaffold functions may enable AnxA6 to modulate migratory cell behavior in health and disease.
Article
Annexin A6 (AnxA6) belongs to the conserved annexin family — a group of Ca2 +-dependent membrane binding proteins. AnxA6 is the largest of all annexins and highly expressed in smooth muscle, hepatocytes, endothelial cells and cardiomyocytes. Upon activation, AnxA6 binds to negatively charged phospholipids in a wide range of intracellular localizations, in particular the plasma membrane, late endosomes/pre-lysosomes, but also synaptic vesicles and sarcolemma. In these cellular sites, AnxA6 is believed to contribute to the organization of membrane microdomains, such as cholesterol-rich lipid rafts and confer multiple regulatory functions, ranging from vesicle fusion, endocytosis and exocytosis to programmed cell death and muscle contraction. Growing evidence supports that Ca2 + and Ca2 +-binding proteins control endocytosis and autophagy. Their regulatory role seems to operate at the level of the signalling pathways that initiate autophagy or at later stages, when autophagosomes fuse with endolysosomal compartments. The convergence of the autophagic and endocytic vesicles to lysosomes shares several features that depend on Ca2 + originating from lysosomes/late endosomes and seems to depend on proteins that are subsequently activated by this cation. However, the involvement of Ca2 + and its effector proteins in these autophagic and endocytic stages still remains poorly understood. Although AnxA6 makes up almost 0.25% of total protein in the liver, little is known about its function in hepatocytes. Within the endocytic route, we identified AnxA6 in endosomes and autophagosomes of hepatocytes. Hence, AnxA6 and possibly other annexins might represent new Ca2 + effectors that regulate converging steps of autophagy and endocytic trafficking in hepatocytes. This article is part of a Special Issue entitled: ECS Meeting edited by Claus Heizmann, Joachim Krebs and Jacques Haiech.
Article
Annexins are a highly conserved protein family that bind to phospholipids in a calcium (Ca2+) -dependent manner. Studies with purified annexins, as well as overexpression and knockdown approaches identified multiple functions predominantly linked to their dynamic and reversible membrane binding behaviour. However, most annexins are found at multiple locations and interact with numerous proteins. Furthermore, similar membrane binding characteristics, overlapping localizations and shared interaction partners have complicated identification of their precise functions. To gain insight into annexin function in vivo, mouse models deficient of annexin A1 (AnxA1), A2, A4, A5, A6 and A7 have been generated. Interestingly, with the exception of one study, all mice strains lacking one or even two annexins are viable and develop normally. This suggested redundancy within annexins, but examining these knockout (KO) -strains under stress conditions revealed striking phenotypes, identifying underlying mechanisms specific for individual annexins, often supporting Ca2+ homeostasis and membrane transport being central for annexin biology. On the other hand, mice lacking AnxA1 or A2 show extracellular functions relevant in health and disease that appear independent of membrane trafficking or Ca2+ signaling. This review will summarize the mechanistic insights gained from studies utilizing mouse models lacking members of the annexin family.
Article
Vascular smooth muscle cell (VSMC) phenotypic conversion from a contractile to «synthetic» state contributes to vascular pathologies including restenosis, atherosclerosis and vascular calcification. We have recently found that the secretion of exosomes is a feature of “synthetic” VSMCs and that exosomes are novel players in vascular repair processes as well as pathological vascular thrombosis and calcification. Pro-inflammatory cytokines and growth factors as well as mineral imbalance stimulate exosome secretion by VSMCs, most likely by the activation of sphingomyelin phosphodiesterase 3 (SMPD3) and cytoskeletal remodeling. Calcium stress induces dramatic changes in VSMC exosome composition and accumulation of phosphatidylserine (PS), Annexin A6 and MMP2, which converts exosomes into a nidus for calcification. In addition, by presenting PS, VSMC exosomes can also provide the catalytic surface for the activation of coagulation factors. Recent data showing that VSMC exosomes are loaded with proteins and miRNA regulating cell adhesion and migration highlight VSMC exosomes as potentially important communication messengers in vascular repair. Thus, the identification of signalling pathways regulating VSMC exosome secretion, including activation of SMPD3 and cytoskeletal rearrangements, opens up novel avenues for a deeper understanding of vascular remodeling processes. This article is protected by copyright. All rights reserved