ArticlePDF Available

Modulation of Cell Death in Yeast by the Bcl-2 Family of Proteins

Authors:

Abstract and Figures

Bcl-2 family members are regulators of cell death. The precise biochemical properties of these proteins are unclear although intrafamily protein-protein association is thought to be involved. To elucidate structure-activity relationships among Bcl-2 proteins and identify the pathways in which they act, an inducible death suppressor assay was developed in yeast. Only Bax and Bak killed yeast via a process that did not require interleukin-1beta-converting enzyme-like proteases. Bax/Bak lethality was suppressed by coexpression of Bcl-2 family members that are anti-apoptotic in vertebrates, namely Bcl-xL, Bcl-2, Mcl-1, and A1. Furthermore, Bcl-xL and Bcl-2 suppressed Bax toxicity by distinct mechanisms in yeast. Bad, Bcl-xS, and Ced-9 lacked suppressor activity. These inactive proteins bound to anti-apoptotic members of the Bcl-2 family but not to Bax or Bak. In contrast, most Bcl-2 family proteins that attenuated death bound to Bax and Bak. However, two mutants of Bcl-xL suppressed Bax-induced cell death while having no Bax binding activity. Therefore, Bcl-xL functions independently of Bax binding, perhaps by interacting with a common target or promoting a pathway that antagonizes Bax. Thus, the pathways downstream of Bax and Bcl-xL may be conserved between vertebrates and yeast. This suppressor assay could be used to isolate components of these pathways.
Structure-activity relationships for Bcl-xL and Bcl-2 binding to Bax and suppression of Bax lethality in yeast. Panel A, yeasts were transformed with Bax and an additional plasmid encoding LexA, LexA-Bcl-xL, or a LexA fusion of a mutated Bcl-xL. Growth was assessed after 22 h by measuring cell density at OD 660 and is expressed as Growth Index (see " Experimental Procedures " for details). The binding of the LexA fusion proteins to VP16-Bax was determined using the two-hybrid assay. Colonies that turned blue within 1 h were scored as positive (), whereas those that were still white at 24 h were considered negative (). Three mutants were negative at 1 h but weakly positive at 3 h when present as LexA fusions but were negative at 24 h when present as VP16 fusions. These clones were scored as having marginal Bax binding activity (). The structures of the various Bcl-xL mutants are shown at the left. The major Bcl-2 homology (BH) domains as well as the transmembrane region (TM) are indicated by shaded boxes. The numbers over the constructs refer to the respective amino acids in the full-length Bcl-xL sequence that define particular mutants. In one mutant, XF15, amino acids 26 – 82 were deleted and replaced with the sequence AAAAVAAAA (amino acid single letter code) (A 4 VA 4 ). The data shown are representative of three independent experiments. Panel B, the same series of VP16-Bax binding and suppression experiments were performed for Bcl-2 and a number of its mutants. Details of the growth and two-hybrid assays are as for panel A, as are domain nomenclature and amino acid numbering. In one Bcl-2 mutant, BF6, residues 32– 87 were deleted and replaced with four alanines (A 4 ).
… 
Content may be subject to copyright.
Modulation of Cell Death in Yeast by the Bcl-2 Family of Proteins*
(Received for publication, February 27, 1997)
Weikang Tao‡§, Cornelia Kurschner‡, and James I. Morgan‡
From the Department of Developmental Neurobiology, St. Jude Children’s Research Hospital, Memphis, Tennessee 38105
and the §Department of Biochemistry and Molecular Biology, University of Medicine and Dentistry of New Jersey-
Graduate School of Biochemical Sciences, Newark, New Jersey 07103
Bcl-2 family members are regulators of cell death. The
precise biochemical properties of these proteins are un-
clear although intrafamily protein-protein association
is thought to be involved. To elucidate structure-activity
relationships among Bcl-2 proteins and identify the
pathways in which they act, an inducible death suppres-
sor assay was developed in yeast. Only Bax and Bak
killed yeast via a process that did not require interleu-
kin-1
b
-converting enzyme-like proteases. Bax/Bak le-
thality was suppressed by coexpression of Bcl-2 family
members that are anti-apoptotic in vertebrates, namely
Bcl-xL, Bcl-2, Mcl-1, and A1. Furthermore, Bcl-xL and
Bcl-2 suppressed Bax toxicity by distinct mechanisms in
yeast. Bad, Bcl-xS, and Ced-9 lacked suppressor activity.
These inactive proteins bound to anti-apoptotic mem-
bers of the Bcl-2 family but not to Bax or Bak. In con-
trast, most Bcl-2 family proteins that attenuated death
bound to Bax and Bak. However, two mutants of Bcl-xL
suppressed Bax-induced cell death while having no Bax
binding activity. Therefore, Bcl-xL functions independ-
ently of Bax binding, perhaps by interacting with a com-
mon target or promoting a pathway that antagonizes
Bax. Thus, the pathways downstream of Bax and Bcl-xL
may be conserved between vertebrates and yeast. This
suppressor assay could be used to isolate components of
these pathways.
Cell death is a highly regulated process involving interac-
tions among extracellular molecules, intracellular signal trans-
duction pathways, and resident suicide/rescue programs (1, 2).
Studies in Caenorhabditis elegans have pointed to a cell suicide
pathway that includes several molecules that have homologs in
vertebrates (3). Central among these death-regulating proteins
is Ced-9, which suppresses programmed cell death in C. el-
egans (4). Bcl-2, the vertebrate homolog of Ced-9, was identified
independently through its translocation in many B cell follicu-
lar lymphomas in man (5, 6). Bcl-2 inhibits cell death in various
circumstances in vertebrate cells and functionally substitutes
for Ced-9 in C. elegans (7, 8). Subsequent investigations have
identified a number of proteins in vertebrates that are struc-
turally related to Bcl-2 (7). These proteins constitute a family,
the members of which share a number of regions of homology,
termed BH1 (Bcl-2 homology), BH2, and BH3 domains (9, 10).
Some of these Bcl-2 related proteins, such as Bcl-xL, also pre-
vent cell death, whereas others, such as Bax, provoke cell
elimination. The biochemical and biophysical mechanisms that
confer these properties on the Bcl-2 family of proteins remain
enigmatic, although recent structural data suggest that they
may be pore-forming proteins (11). However, Bcl-2 and many of
its related proteins can participate in homo- and heteromeric
complexes, and it has been suggested that the activity of pro-
apoptotic members of the Bcl-2 family is neutralized by their
association with anti-apoptotic members (12, 13).
There are a number of caveats in the interpretation of the
role of Bcl-2 family members in the regulation of cell death in
vertebrates. First, the effects of Bcl-2 family members are often
assessed in models where cell death is triggered by an exoge-
nous means such as growth factor withdrawal or addition of a
toxin or virus to the culture medium. While this is more rele-
vant to the physiological situation, it adds a level of ambiguity
as to whether the effects are mediated through intrafamily
interactions. Second, a given cell type may already express the
gene of interest as well as other known and potentially un-
known members of the Bcl-2 family. Thus, there is uncertainty
as to precisely which proteins interact to produce the observed
effect. Finally, Bcl-2 family members are differentially ex-
pressed, and bcl-2- and bcl-x-null mice have distinct pheno-
types (14, 15). Together, these data imply that the various
Bcl-2-like proteins have functional differences. Indeed, there
are indications that Bcl-xL need not dimerize with Bax to
suppress cell killing and that Bcl-2 and Bcl-xL have differential
activities in some assays (14–18). Thus, from the mechanistic
standpoint, there is a need for a model in which the role of Bcl-2
family members in cell death can be determined without the
foregoing ambiguities.
Recently, several studies reported that the expression of Bax
is lethal in the budding yeast, Saccharomyces cerevisiae (19
21). This is despite the fact that yeasts express no identifiable
members of the Bcl-2 family and are not known to undergo
programmed cell death. This afforded the opportunity to de-
velop a suppressor assay in which the ability of Bcl-2 family
members to attenuate Bax killing could be determined quanti-
tatively. Moreover, since the model is in essence the same as a
two-hybrid system, protein-protein association can be assessed
simultaneously.
It is shown that Bcl-2, Bcl-xL, Mcl-1, and A1 can suppress
death induced by Bax and Bak in yeast, whereas Ced-9, Bad,
and Bcl-xS are inactive. Bcl-2 and Bcl-xL do not have identical
structure-activity relationships for death suppression in yeast,
indicating that they have distinct modes of action. Moreover,
several Bcl-xL mutants suppressed Bax lethality without bind-
ing to Bax, indicating that Bcl-xL can function by a mechanism
other than direct association with Bax. This implies that mech-
anisms downstream of Bcl-xL and Bax are conserved in yeast
and that this system could be used to isolate these molecules.
* This work was supported in part by National Institutes of Health
Cancer Support CORE Grant P30 CA21765 and by the American Leb-
anese Syrian Associated Charities. The costs of publication of this
article were defrayed in part by the payment of page charges. This
article must therefore be hereby marked “advertisement” in accordance
with 18 U.S.C. Section 1734 solely to indicate this fact.
To whom correspondence should be addressed: Dept. of Develop-
mental Neurobiology, St. Jude Children’s Research Hospital, 332 N.
Lauderdale, Memphis, TN 38105. Tel.: 901-495-2256; Fax: 901-495-
3143; E-mail: jim.morgan@stjude.org.
THE JOURNAL OF BIOLOGICAL CHEMISTRY Vol. 272, No. 24, Issue of June 13, pp. 15547–15552, 1997
© 1997 by The American Society for Biochemistry and Molecular Biology, Inc. Printed in U.S.A.
This paper is available on line at http://www.jbc.org 15547
by guest on April 19, 2017http://www.jbc.org/Downloaded from
EXPERIMENTAL PROCEDURES
Yeast Strains, Growth, and Transformation—The S. cerevisiae strain
S260 (ura3 trp1) contained a genomic LEXA-operator-LACZ fusion
reporter gene and was described previously (22). Yeast growth, main-
tenance, and transformations were as described (23).
Yeast Expression Constructs—Fusion proteins with the LexA DNA
binding domain were constructed in the yeast expression plasmid,
Y.LexA (22), which carries the S. cerevisiae TRP1 gene as a selectable
marker. Fusion proteins with the VP16 transcriptional activation do-
main were generated in pSD.10a, which harbors the URA3 selection
marker (24). Constructs lacking heterologous fusion sequences were
made in pSD.10a after deletion of the VP16 codons.
LexA and VP16 fusions of murine Bax, Bcl-2, and A1 were reported
previously (22). cDNAs encoding murine Bcl-x, Bak, and Bad were
obtained by reverse transcription polymerase chain reaction using
mouse brain RNA and polymerase chain reaction primers based upon
published sequences. Murine mcl-1 cDNA was isolated in a yeast two-
hybrid screening for Bax-binding proteins.
1
ACed-9 cDNA was a gift
from Dr. M. O. Hengartner, and plasmid p996 containing a crmA cDNA
(25) was a gift from Dr. D. Pickup. Truncation and deletion mutants
were generated by polymerase chain reaction. The sequences of all
constructs were verified.
In Vitro Translation Constructs—For in vitro transcription/transla-
tion reactions, cDNAs encoding full-length Bcl-xL and two of its mu-
tants (XF14 and XF15) were inserted into the vector pT7
b
plink (24). A
full-length murine bax cDNA was cloned in pT7
b
plink-TagN (22). In
this construct, an epitope tag derived from human c-MYC protein is
fused to the N terminus of Bax. This epitope is recognized by the
monoclonal antibody 9E10.
Coimmunoprecipitation—Proteins were translated in vitro as de-
scribed (22). MYC-tagged Bax was translated in the absence of [
35
S]me-
thionine (Amersham Life Science, Inc.), and untagged Bcl-xL and its
mutants XF14 and XF15 were translated in the presence of [
35
S]methi-
onine. For coimmunoprecipitations, 2
m
l of the Bax-MYC translation
reaction were mixed with 10
m
l of the Bcl-xL, XF14, or XF15 translation
reactions, respectively. Incubation and washing steps were performed
as described (26) in NETgel buffer with 0.2% Nonidet P-40. The precip-
itating antibody was 9E10 (Santa Cruz Biotechnology, Inc.). Protein
A-Sepharose CL-4B (Sigma) was used to precipitate the immune
complexes.
Yeast Two-hybrid Analysis—S260 was transformed with two plas-
mids encoding a LexA fusion construct and a VP16 hybrid. Transform-
ants were grown and assayed for
b
-galactosidase activity as described
(22). The development of blue color in the yeast colonies was monitored
for 24 h.
Yeast Growth Assay—S260 was (co)transformed with expression
plasmids encoding Bcl-2 family members. Selective media containing
2% glucose was inoculated with a single colony of transformants and
incubated overnight at 30 °C. Subsequently, cells were washed three
times with H
2
O. Typically, 20 ml of selective medium (with 2% galac-
tose) was inoculated with 2.56 310
7
cells, and incubation was contin-
ued. Samples were taken at different time points, and cell density was
measured by determining the OD at 660 nm. To compare results
from different experiments, a growth index (GI)
2
was devised.
(OD
660/22 h
2OD
660/0 h
(cells containing Bax or Bak and the test
protein))/(OD660/22 h 2OD
660/0 h
(cells containing Bax or Bak and LexA or
VP16)).
Immunoblotting—Yeast cells were lysed mechanically as described
(26). Proteins were separated on 15% SDS-polyacrylamide gels (30
m
g/lane) and transferred to nitrocellulose membranes. Immunostaining
was performed in Tris-buffered saline containing 1% fetal calf serum at
room temperature. Membranes were incubated with a rabbit anti-
murine Bax polyclonal antibody, 13686E (1:1000 dilution) (Pharmin-
gen), followed by horseradish peroxidase-conjugated goat anti-rabbit
immunoglobulin antiserum (1:1000) (Amersham), both for 2 h. Immu-
noblots were developed using diaminobenzidine as chromogen (27). For
Bcl-xL immunostaining, the membrane was stripped of bound antibody,
reprobed with a monoclonal anti-Bcl-x antibody, B22620 (1:250) (Trans-
duction Laboratories). The immunoblots were developed using the en-
hanced chemiluminescence method (Amersham).
RESULTS
Identification of Members of the Bcl-2 Family That Are Le-
thal in Yeast—Since Bax kills yeast (19–21), additional mem-
bers of the Bcl-2 family (Bcl-2, Bcl-xL, Bcl-xS, A1, Mcl-1, Bad,
Bak, and Ced-9) were tested for this property. Besides Bax,
only Bak was lethal, although it was consistently less potent
(Table I). Moreover, like native Bax, the LexA and VP16 fusion
proteins of Bax and Bak used in the two-hybrid assay were also
lethal (Fig. 1, Aand B, and Table I), making it possible to
correlate binding activity with biological activity in subsequent
studies.
Suppression of Bax- and Bak-induced Lethality by Bcl-2
Family Members—To quantitatively determine the suppres-
sion of Bax/Bak toxicity in yeast, cDNAs encoding LexA or
VP16 fusions of various Bcl-2 family members were coex-
pressed with either Bax or Bak. Coexpression of the Bcl-xL
fusion proteins generated for the two-hybrid system inhibited
Bax and Bak toxicity (Fig. 1Aand Table I). In addition, LexA
fusions of Bcl-2, Mcl-1, and A1 inhibited both Bax and Bak
killing (Fig. 2 and Table I). Bcl-xS, Bad, and Ced-9 did not
attenuate Bax or Bak lethality (Table I). To ensure that the
results were not artifacts of LexA or VP16 fusion proteins,
these data were confirmed using native (unfused) protein se-
quences. Both native Bcl-2 (GI, 7.4) and native Bcl-xL (GI, 8.0)
suppressed the killing elicited by native Bax (GI, 1.0).
Relationship between the Anti-death Activity of Bcl-2 Family
Members and Their Binding to Bax and Bak—In the yeast
two-hybrid assay, Bcl-xL, Bcl-2, Mcl-1, and A1 all bound to Bax
and Bak, whereas Bcl-xS, Bad, and Ced-9 did not (Table I).
Therefore, anti-death activity was associated with the ability to
bind to Bax or Bak. Proteins such as Ced-9, Bcl-xS, and Bad
that did not bind to Bax/Bak did not inhibit killing, whereas
proteins that could associate, such as Bcl-xL and Mcl-1, were
inhibitory. Since the lack of death suppressor activity of Ced-9
was unexpected, its association with anti-apoptotic members of
the Bcl-2 family was determined. LexA-Ced-9 (as well as LexA-
Bcl-xS and LexA-Bad) bound to Bcl-xL, Bcl-2, and A1 (data not
shown). Therefore, Ced-9 had the binding and activity profiles
of proteins such as Bcl-xS rather than Bcl-2.
Mutations in Bcl-xL Dissociate Bax Binding from Death Sup-
pressor Activity—To further pursue the relationship between
heterodimerization and biological activity, a series of trunca-
tion and deletion mutations of Bcl-2 and Bcl-xL were made.
1
C. Kurschner and J. I. Morgan, unpublished data.
2The abbreviations used are: GI, growth index; ICE, interleukin-1
b
-
converting enzyme; TM, transmembrane.
TABLE I
Relationship between heterodimerization and suppression of cell death for Bcl-2 family members in yeast
S260 yeast were cotransformed with either VP16-fused Bax or Bak and one of the indicated Bcl-2 family members fused to LexA. Association
between the proteins was determined by the two-hybrid assay. 1, blue colony within 1 h; 2, no blue colony after 24 h. The death-suppressive
activities of the Bcl-2 family members were determined in the yeast growth assay. GI values are indicated. ND, not done; NA, not applicable. The
data shown are representative of three independent experiments.
Y.LexA Bcl-2 Bcl-xL Bcl-xS A1 Mcl-1 Bad Ced-9 Bax Bak
Bax
Binding 2 1 1 2 1 1 2211
Suppression 1.00 5.18 5.55 1.01 3.60 8.05 0.82 1.10 NA ND
Bak
Binding 2 1 1 2 1 1 2211
Suppression 1.00 29.15 29.87 0.97 27.75 28.44 1.24 1.12 0.13 NA
Structure-Activity Relationships of Bcl-2 Family in Yeast15548
by guest on April 19, 2017http://www.jbc.org/Downloaded from
When coexpressed with Bax, the majority of these mutants
failed to bind to Bax and did not suppress death (Fig. 2, Aand
B). However, two Bcl-xL mutants, XF14 and XF15, were active
in suppression of Bax toxicity but either bound weakly or not
all in the two-hybrid assay (Fig. 2A).
To ensure that these data were not a product of the LexA
sequences, unfused XF14 and XF15 were examined for their
suppressive effects on native Bax toxicity and for coimmuno-
precipitation with MYC-tagged Bax. Both unfused XF14 (GI,
8.7) and XF15 (GI, 8.8) were potent suppressors of Bax lethality
(GI, 1.0). For coimmunoprecipitation, the proteins were in vitro
translated in the presence of [
35
S]methionine, whereas a MYC
epitope-tagged Bax (MYC-Bax) was translated in the absence
of the radionuclide. After appropriate mixing and incubation,
Bcl-xL was specifically immunoprecipitated with MYC-Bax but
not the MYC epitope alone (Fig. 3). In contrast, neither XF14
nor XF15 was precipitated specifically with MYC-Bax and the
anti-MYC monoclonal antibody (Fig. 3). XF15 showed no evi-
dence of Bax binding. XF14 gave a relatively high background
immunoprecipitation that was not augmented by the presence
of MYC-Bax, indicating that it does not bind to Bax. This
analysis confirms a prior report using these and similar con-
structs (28).
Bcl-xL and Bcl-xL Mutants Do Not Alter Bax Expression—To
preclude the possibility that coexpression of Bcl-xL or the mu-
tants either reduced the expression or promoted the degrada-
tion of Bax, steady-state protein levels were determined by
FIG.2. Structure-activity relationships for Bcl-xL and Bcl-2
binding to Bax and suppression of Bax lethality in yeast. Panel A,
yeasts were transformed with Bax and an additional plasmid encoding
LexA, LexA-Bcl-xL, or a LexA fusion of a mutated Bcl-xL. Growth was
assessed after 22 h by measuring cell density at OD
660
and is expressed
as Growth Index (see “Experimental Procedures” for details). The bind-
ing of the LexA fusion proteins to VP16-Bax was determined using the
two-hybrid assay. Colonies that turned blue within 1 h were scored as
positive (1), whereas those that were still white at 24 h were considered
negative (2). Three mutants were negative at 1 h but weakly positive at
3 h when present as LexA fusions but were negative at 24 h when
present as VP16 fusions. These clones were scored as having marginal
Bax binding activity (6). The structures of the various Bcl-xL mutants
are shown at the left. The major Bcl-2 homology (BH) domains as well
as the transmembrane region (TM) are indicated by shaded boxes. The
numbers over the constructs refer to the respective amino acids in the
full-length Bcl-xL sequence that define particular mutants. In one
mutant, XF15, amino acids 2682 were deleted and replaced with the
sequence AAAAVAAAA (amino acid single letter code) (A
4
VA
4
). The
data shown are representative of three independent experiments. Panel
B, the same series of VP16-Bax binding and suppression experiments
were performed for Bcl-2 and a number of its mutants. Details of the
growth and two-hybrid assays are as for panel A, as are domain no-
menclature and amino acid numbering. In one Bcl-2 mutant, BF6,
residues 32–87 were deleted and replaced with four alanines (A
4
).
FIG.1. Bcl-xL suppresses Bax-induced lethality in yeast. A,
yeasts were transformed with plasmids encoding LexA, LexA-Bax,
LexA-Bax and Bcl-xL, or LexA-Bax and VP16. Cell density was deter-
mined at various time intervals by measuring OD
660
.B, yeasts were
transformed with plasmids encoding VP16, Bax, Bax and LexA, or Bax
and LexA-Bcl-xL. Growth was assessed at various times by measuring
OD
660
. The data are representative curves that were repeated a mini-
mum of three times.
Structure-Activity Relationships of Bcl-2 Family in Yeast 15549
by guest on April 19, 2017http://www.jbc.org/Downloaded from
immunoblotting. Fig. 4Ashows the immunoblot analysis of Bax
and Bcl-xL proteins in yeast. Cells were cotransformed with
constructs encoding LexA-Bax and one of unfused Bcl-xL mu-
tants, XF14 or XF15. An ;45-kDa LexA-Bax fusion protein
accumulated to maximal levels within 12 h after galactose
induction. Expression of ;26-kDa Bcl-xL, ;20-kDa XF14, and
;21-kDa XF15 immunoreactive bands were also detected 12 h
after galactose induction. None of the anti-death proteins al-
tered the levels or time course of LexA-Bax expression (Fig.
4A). Additional Bax-reactive proteins of lower molecular mass
were also induced with similar kinetics to LexA-Bax. The levels
of these proteins were also unaffected by Bcl-xL expression
(Fig. 4A). It is unclear whether these bands are proteolytic
fragments of LexA-Bax or incomplete transcription/translation
products. No such proteins were observed when unfused Bax
was expressed with Bcl-xL in yeast (Fig. 4B), suggesting that
the additional bands do not represent selective processing of
Bax sequences.
Whereas expression of Bax is lethal, yeast transformed with
the Bax plasmid did grow if cultured for longer periods (data
not shown). This phenomenon is important in that it has im-
plications for the use of this system as a suppressor screening
assay. The growth effect could arise in a number of ways. First,
a mutation in the vector could either inhibit expression of Bax
or render it biologically inactive. Second, yeast may produce
their own suppressor of Bax or lose a target of Bax. To test
these possibilities, yeasts were cotransformed with Bax and
Bcl-xL and subjected to immunoblot analysis after 100 h of
culture. Fig. 4Bshows that cells containing Bax and LexA
plasmids no longer expressed immunoreactive Bax. However,
an ;21-kDa Bax was still present in the cells containing Bax
and LexA-Bcl-xL. Thus, mutations are selected for in Bax-
expressing yeast that result in a loss of Bax expression. In the
presence of Bcl-xL, this selective pressure is absent, and high
(normally lethal) levels of full-length Bax are expressed. This
effect must be considered when using this suppressor assay to
identify proteins that functionally interact with Bax/Bak.
Bax Toxicity May Not Be Mediated by ICE-like Proteases—
Since Bax and Bcl-xL may function independently, the mech-
anisms that mediate Bax toxicity were investigated. Another
study has established that Bax lethality in yeast is not associ-
ated with DNA laddering (21), although it may involve prote-
olysis. Indeed, proteases belonging to the interleukin-1
b
-con-
verting enzyme (ICE) subfamily (recently termed caspases)
have been implicated in cell death in phylogenetically dispar-
ate species (2). A search of the yeast genome for the consensus
active site of ICE proteases (QACRG) yielded no hits, suggest-
ing that these enzymes could not mediate death in yeast. To
further examine this point, yeasts were cotransformed with
Bax and CrmA, an inhibitor of ICE proteases that is derived
from cowpox virus (25). CrmA did not rescue Bax toxicity and
alone had no effect upon yeast growth (data not shown). To-
gether, the data indicate that ICE-like proteases do not medi-
ate Bax lethality in yeast.
Bcl-2 and Bcl-xL Have Distinct Structure-Activity Relation-
ships in Yeast—Several studies have suggested that Bcl-2 and
Bcl-xL have distinct properties (15, 17, 18). Therefore, the
structure-activity relationships for Bax dimerization and sup-
pression were determined for the two molecules in yeast. Two
regions of Bcl-2 and Bcl-xL distinguished the biological prop-
erties of the two proteins. We confirm that the transmembrane
(TM) domain of Bcl-2 is not essential for suppression of Bax
toxicity in yeast and its elimination does not affect Bax binding
(see mutant BF3 in Fig. 2B) (21). However, elimination of the
TM domain in Bcl-xL leads to both the loss of Bax binding and
suppressor activity (see mutant XF3 in Fig. 2A). As shown
above, deletion of the putative loop region in Bcl-xL (mutants
FIG.3. Coimmunoprecipitation of Bcl-xL and Bcl-xL mutants
with MYC-tagged Bax. Either unlabeled MYC-tagged Bax (MYC-Bax)
or the MYC epitope (MYC) were incubated with [
35
S]methionine-la-
beled Bcl-xL, XF14, or XF15 (see “Experimental Procedures” for de-
tails). Subsequently, immunoprecipitation was carried out using the
monoclonal anti-MYC antibody 9E10. Bcl-xL showed a low but detect-
able background precipitation with the MYC epitope alone. However,
Bcl-xL showed markedly enhanced immunoprecipitation in the pres-
ence of MYC-Bax, indicating specific association between Bcl-xL and
Bax. Mutant XF15 was not precipitated by either the MYC epitope or
MYC-Bax, indicating a lack of association of this mutant with Bax.
Mutant XF14 gave a relatively high background with MYC epitope
alone, but unlike Bcl-xL, this signal was not augmented by MYC-Bax.
M
r
(K), molecular mass in kilodaltons.
FIG.4. Evaluation of the levels of Bcl-2 family members in
yeast by immunoblotting. A, yeasts were cotransformed with LexA-
Bax and a plasmid encoding VP16, Bcl-xL, XF14, or XF15. Transform-
ants were switched to galactose medium, and the levels of the various
proteins were determined by sequential immunoblotting as described
under “Experimental Procedures.” The position of full-length LexA-Bax
is indicated. Several inducible lower molecular mass species are also
evident. The upper panel shows the immunoblot of LexA-Bax, whereas
the lower panel depicts the same blot reprobed for Bcl-xL or its two
mutants. The positions of Bcl-xL, XF14, and XF15 are indicated at the
right, and molecular mass is at the left. Coexpression of Bcl-xL or the
two mutants did not affect the level or processing of any of the Bax-
reactive bands. B, yeasts were cotransformed with VP16 and LexA, Bax
and LexA-Bcl-xL, or Bax and LexA. Transformants were switched to
galactose medium for 100 h, and extracts were immunoblotted for Bax
as described under “Experimental Procedures.” Unlike LexA-Bax, na-
tive Bax gave a single band at the appropriate molecular mass. Bax was
only detectable in cultures that coexpressed Bax with LexA-Bcl-xL.
Structure-Activity Relationships of Bcl-2 Family in Yeast15550
by guest on April 19, 2017http://www.jbc.org/Downloaded from
XF14 and XF15) (11) results in a loss of Bax binding but
retention of Bax suppressor activity (Fig. 2A). Whereas an
equivalent mutation in Bcl-2 (BF6) retained suppressor activ-
ity, it still bound well to Bax (Fig. 2B). These data suggest that
the loop region may be important for the interaction of Bcl-xL
with Bax, whereas the equivalent domain in Bcl-2 is not. To-
gether, the results indicate that whereas Bcl-2 and Bcl-xL,
both, can suppress Bax toxicity in yeast, they may not do so in
an identical manner.
DISCUSSION
As in vertebrate cell death models, members of the Bcl-2
family can be grouped into three functional classes in yeast.
The first group comprises proteins, such as Bax and Bak, which
are lethal per se. The second group includes Bcl-2, Bcl-xL,
Mcl-1, and A1, which bind to and suppress Bax and Bak lethal-
ity. The third group bind to anti- but not pro-apoptotic mem-
bers of the Bcl-2 family. These proteins, which include Bad,
Bcl-xS, and the C. elegans Ced-9, are functionally inactive in
terms of direct killing or death suppression. In vertebrates,
proteins such as Bad and Bcl-xS are not considered to be lethal
per se but rather are thought to potentiate killing by binding to
anti-apoptotic members of the Bcl-2 family (29, 30). Thus,
mammalian Bcl-2 family members have the same spectrum of
biological activities in yeast as they do in vertebrate cells.
One unexpected result was that whereas Ced-9 did bind to
anti-apoptotic members of the Bcl-2 family, it did not bind to
Bax and did not suppress Bax/Bak killing. Although we can
find no study that has used Ced-9 to rescue death in a verte-
brate cell, it is the presumed homolog of Bcl-2 (8) and was
expected to suppress killing. However, the properties of Ced-9
are more akin to those of Bcl-xS than they are to Bcl-2. It is
conceivable that Ced-9 binds specifically to a C. elegans hom-
olog of Bax, although no such gene has been identified. How-
ever, the possibility exists that family members such as Ced-9
and Bcl-xS might exert their functions through mechanisms
other than intrafamily binding.
Genetic analysis in C. elegans has shown that Ced-9 acts via
Ced-4, a protein of unknown function, and Ced-3, an ICE-like
cysteine protease (4). It is possible that Ced-9, and by implica-
tion Bcl-xS and Bad, might act by binding to Ced-4 or related
proteins in vertebrates. However, whereas many studies have
implicated cysteine proteases in cell death in vertebrates (2), it
is unlikely that ICE-like proteases mediate Bcl-2 family effects
in yeast. First, Bax lethality is not inhibited by coexpression of
the ICE protease inhibitor, CrmA. Second, no consensus se-
quence for the active site of ICE proteases has been found in
the yeast genome. Therefore, Bax lethality may not involve the
activation of ICE-like proteases in yeast. Indeed, Bax killing
has been shown to be independent of this class of proteases in
at least one vertebrate model (31).
Analysis of the structural requirements of Bcl-xL for sup-
pression of Bax toxicity in yeast revealed that deletions at both
the N and C termini eliminated biological activity. In one
mutant, XF3, deletion of the last 22 amino acids, which in-
cluded the TM domain, resulted in loss of suppressor activity.
This result suggests that membrane targeting is essential for
the protective effect of Bcl-xL in yeast. However, the equivalent
domain in Bcl-2 is not required for rescue (19), indicating that
these two related proteins may have divergent mechanisms of
action. Such a notion is underscored by the distinct phenotypes
of mice that lack functional bcl-2 or bcl-x alleles and by the
unique protective effects of Bcl-xL in some cell death models
(14, 15, 17, 18). Whereas the TM domain is thought to be
necessary for the activity of Bcl-xL in vertebrate cells, the
situation for Bcl-2 is controversial (12, 32, 33). However, the
recent demonstration that Bcl-xL has structural similarities to
the pore-forming subunit of diphtheria toxin suggests that its
biological function may occur at or within cellular membranes
(11). Therefore it is conceivable that there are proteins in yeast
that can dock Bcl-2, but not Bcl-xL, to membranes thereby
obviating the requirement for a TM domain. Bcl-2 and Bcl-xL
can also be discriminated by their structure-activity relation-
ship for binding to Bax. Mutants of Bcl-xL that had the puta-
tive loop domain deleted (XF14 and XF15) did not bind Bax,
whereas an equivalent deletion in Bcl-2 (BF6) did bind in the
two-hybrid assay. Since the loop regions of Bcl-2 and Bcl-xL are
not conserved, these data may be an indication that these
domains can selectively modify dimerization, although they
may not be part of the dimerization interface.
Dimerization between Bcl-2 family members is considered
central to their biological activity. Indeed, in one model it is the
stoichiometry of various pro- to anti-apoptotic Bcl-2 family
members that is supposed to determine cell fate (34). However,
the view that dimerization is the sole determinant of activity
has been questioned. For example, some Bcl-xL mutants that
failed to associate with Bax still rescued 7080% of Sindbis
virus-induced cell death (16). However, it could be argued that
Sindbis virus-triggered cell death may not be mediated by Bax.
Indeed, there is evidence of Bax-independent cell death path-
ways in bax-null mice (35). Recently, some Bcl-xL mutations
were described that had reduced, or no, binding activity to Bax
but that rescued vertebrate cells from death triggered by IL-3
deprivation (11, 28). Therefore we compared the activity of
these deletions with other Bcl-xL and Bcl-2 mutants as sup-
pressors of Bax toxicity in yeast. All full-length Bcl-2 family
members and mutants that bound to Bax inhibited killing,
whereas those that did not bind were inactive. However, the
two internal deletion mutants of Bcl-xL, XF14 and XF15, that
did not bind to Bax were potent suppressors. This indicates
that dimerization is not essential for Bcl-xL to suppress Bax
lethality in yeast.
The observation that Bcl-xL can antagonize Bax killing in
yeast independent of heterodimerization has several important
implications. First, it argues against the effects of Bax in yeast
as being nonspecific toxicity that can be attenuated by any
Bax-binding protein. Second, it suggests that Bax and Bcl-xL
either interact with, or compete for, a common downstream
target or pathway in yeast. Alternatively, they could influence
antagonistic mechanisms. Third, the data suggest that Bax and
Bcl-xL act upon mechanisms that have been conserved from
yeast to mammals, although these processes may only have
been adapted for the control of cell elimination in multicellular
organisms. The assay described here can be used as a genetic
suppressor screen to identify potential components of this path-
way in yeast and vertebrates.
Acknowledgments—We thank Dr. Craig B. Thompson for helpful
discussions and Dr. Steven Dalton for providing the yeast expression
plasmids pSD.10a and Y.LexA, in vitro translation plasmids, and the S.
cerevisiae reporter strain S260.
REFERENCES
1. Ellis, R. E., Yuan, J., and Horvitz, H. R. (1991) Annu. Rev. Cell Biol. 7,
663–698
2. Hale, A. J., Smith, C. A., Sutherland, L. C., Stoneman, V. E. A., Longthorne, V.
L., Culhane, A. C., and Williams, G. T. (1996) Eur. J. Biochem. 236, 1–26
3. Miura, M., and Yuan, J. (1996) Curr. Top. Dev. Biol. 32, 139–174
4. Hengartner, M., Ellis, R., and Horvitz, H. (1992) Nature 356, 494–499
5. Tsujimoto, Y., Cossman, J., Jaffe, E., and Croce, C. (1985) Science 228,
1440–1443
6. Tsujimoto, Y., and Croce, C. R. (1986) Proc. Natl. Acad. Sci. U. S. A. 83,
5214–5218
7. Reed, J. (1994) J. Cell Biol. 124, 1–6
8. Hengartner, M. O., and Horvitz, H. R. (1994) Cell 76, 665–676
9. Williams, G. T., and Smith, C. A. (1993) Cell 74, 777–779
10. Zha, H., Aime´-Sempe´, C., Sato, T., and Reed, J. C. (1996) J. Biol. Chem. 271,
7440–7444
11. Muchmore, S. M., Sattler, M., Liang, H., Meadows, R. P., Harlan, J. E., Yoon,
H. P., Nettesheim, D., Chang, B. S., Thompson, C. B., Wong, S.-L., Ng, S.-C.,
Structure-Activity Relationships of Bcl-2 Family in Yeast 15551
by guest on April 19, 2017http://www.jbc.org/Downloaded from
and Fesik, S. W. (1996) Nature 381, 335–341
12. Oltvai, Z. N., Milliman, C. L., and Korsmeyer, S. J. (1993) Cell 74, 609–619
13. Sato, T., Hanada, M., Bodrug, S., Irie, S., Iwama, N., Boise, L. H., Thompson,
C. B., Golemis, E., Fong, L., Wang, H.-G., and Reed, J. C. (1994) Proc. Natl.
Acad. Sci. U. S. A. 91, 9238–9242
14. Nakayama, K.-I., Nakayama, K., Negishi, I., Kuida, K., Shinkai, Y., Louie, M.
C., Fields, L. E., Lucas, P. J., Stewart, V., Alt, F. W., and Loh, D. Y. (1993)
Science 261, 1584–1588
15. Motoyama, N., Wang, F., Roth, K. A., Sawa, H., Nakayama, K.-I., Nakayama,
K., Negishi, I., Senju, S., Zhang, Q., Fujii, S., and Loh, D. Y. (1995) Science
267, 1506–1509
16. Cheng, E. H.-Y., Levine, B., Boise, L. H., Thompson, C. B., and Hardwick, J. M.
(1996) Nature 379, 554–556
17. Tuscano, J. M., Druey, K. M., Riva, A., Pena, J., Thompson, C. B., and Kehrl,
J. H. (1996) Blood 88, 1359–1364
18. Gottschalk, A. R., Boise, L. H., Thompson, C. B., and Quintans, J. (1994) Proc.
Natl. Acad. Sci. U. S. A. 91, 7350–7354
19. Hanada, M., Aime´-Sempe´, C., Sato, T., and Reed, J., C. (1995) J. Biol. Chem.
270, 11962–11969
20. Greenhalf, W., Stephan, C., and Chaudhuri, B. (1996) FEBS Lett. 380,
169–175
21. Zha, H., Fisk, H. A., Yaffe, M. P., Mahajan, N., Herman, B., and Reed, J. C.
(1996) Mol. Cell. Biol. 16, 6494–6508
22. Kurschner, C., and Morgan, J. I. (1996) Mol. Brain Res. 37, 249–258
23. Kurschner, C., and Morgan, J. I. (1995) Mol. Cell. Biol. 15, 246–254
24. Dalton, S., and Treisman, R. (1992) Cell 68, 597–612
25. Ray, C., Black, R., Kronheim, S., Greenstreet, T., Sleath, P., Salvesen, G., and
Pickup, D. (1992) Cell 69, 597–604
26. Sambrook, J., Fritsch, E. F., and Maniatis, T. (1989) in Molecular Cloning: A
Laboratory Manual, 2nd Ed., Cold Spring Harbor Laboratory, Cold Spring
Harbor, NY
27. Gallagher, S., and Hurrel, J. G. R. (1993) in Current Protocols in Molecular
Biology, pp. 10.8.1–10.8.14, John Wiley & Sons, Inc., NY
28. Chang, B. S., Minn, A. J., Muchmore, S. W., Fesik, S. W., and Thompson, C. B.
(1997) EMBO J. 16, 968–977
29. Yang, E., Zha, J., Jockel, J., Boise, L. H., Thompson, C. B., and Korsmeyer, S.
J. (1995) Cell 80, 285–291
30. Boise, L. H., Gonzalez-Garcia, M., Postema, C. E., Ding, L., Lindsten, T.,
Turka, L. A., Mao, X., Nunez, G., and Thompson, C. B. (1993) Cell 74,
597–608
31. Xiang, J., Chao, D. T., and Korsmeyer, S. J. (1996) Proc. Natl. Acad. Sci.
U. S. A. 93, 14559 –14563
32. Borner, C., Martinou, I., Mattmann, C., Irmler, M., Schaere, E., and Martinou,
J.-C. (1994) J. Cell Biol. 126, 1059–1068
33. Hunter, J. J., Bond, B. L., and Parslow, T. G. (1996) Mol. Cell. Biol. 16,
877–883
34. Oltvai, Z. N., and Korsmeyer, S. J. (1994) Cell 79, 189–192
35. Knudson, C. M., Tung, K. S. K., Tourtellotte, W. G., Brown, G. A. J., and
Korsmeyer, S. J. (1995) Science 270, 96–98
Structure-Activity Relationships of Bcl-2 Family in Yeast15552
by guest on April 19, 2017http://www.jbc.org/Downloaded from
Weikang Tao, Cornelia Kurschner and James I. Morgan
Modulation of Cell Death in Yeast by the Bcl-2 Family of Proteins
doi: 10.1074/jbc.272.24.15547
1997, 272:15547-15552.J. Biol. Chem.
http://www.jbc.org/content/272/24/15547Access the most updated version of this article at
Alerts:
When a correction for this article is posted When this article is cited
to choose from all of JBC's e-mail alertsClick here
http://www.jbc.org/content/272/24/15547.full.html#ref-list-1
This article cites 33 references, 17 of which can be accessed free at
by guest on April 19, 2017http://www.jbc.org/Downloaded from
... Persuasive evidence that pro-apoptotic members of the Bcl-2 family also act on m itochondria is derived from several studies in the unicellular eukaryotes Saccharomyces cerevisiae and Schizosaccharomyces pomhe. In both yeasts, ectopic expression of Bax or Bak is toxic, while expression of Bcl-2 or Bcl-XL is not (Greenhalf et al., 1996;Ink et al., 1997;Jurgensmeier et al., 1997;Tao et al., 1997;. The genome of S. cerevisiae has been completely sequenced (Nature, 1997) and appears not to contain Bcl-2 hom ologues, suggesting that the pro-apoptotic m oleules have an intrinsic activity and are not exerting their effect through neutralisation of an anti-apoptotic homologue. ...
... pombe results in cytotoxicity, w ith sim ilar phenotypes in each case (Greenhalf et al., 1996;Ink et al., 1997;Jurgensmeier et al., 1997;Tao et al., 1997;. Although this killing could m erely represent some form of 'non-specific' toxicity, expression of either Bax/Bak m utant proteins that are unable to induce apoptosis in mammalian cells, or of antiapoptotic Bcl-2 family m em bers (including CED-9), does not kill yeasts suggesting that the observed cytotoxicity has some relevance to Bax/Bak function in m am m alian cells. ...
Thesis
The nematode worm Caenorhabditis elegans has proved to be an illuminating model for programmed cell death. Three principal genes are involved in nematode PCD, ced-3, ced-4 and ced-9: each has proved to have vertebrate homologues that are critical regulators of vertebrate apoptosis. Until recently, the molecular functions of CED-4 and CED-9 were largely obscure. I have used the yeast Schizosaccharomyces pombe as a naive model system in which to assay the function of the pro-apoptotic protein CED-4. Expression of wild-type ced-4 is toxic to S. pombe, while expression of the point mutant I258N, which gives rise to a ced-4-null phenotype in the worm, has no effect, suggesting that the toxicity is the result of a bona fide activity of CED-4. Mutation of the putative nucleotide-binding P-loop motif of CED-4 also eliminates the lethal phenotype, demonstrating for the first time the importance of this domain for CED-4 function. The anti-apoptotic protein CED-9 is able to rescue S. pombe from CED-4- induced lethality: the most parsimonious explanation for this observation is that CED-9 directly binds and inhibits CED-4. This is confirmed by immunogold labelling of CED-4 visualised by electron microscopy: CED-4 expressed alone is nuclear, but when co-expressed with CED-9 it is found on mitochondrial and ER membranes, the presumptive location of CED-9. The physical interaction between CED-4 and CED-9 is further confirmed by yeast two-hybrid analysis. In addition, the cloning and characterisation of two C. elegans homologues of baculovirus Inhibitor of Apoptosis Proteins (IAPs) is described. Cellular IAP homologues are found in Drosophila and humans and influence apoptosis in both organisms. The C. elegans IAP homologues do not inhibit the activity of CED-3 or CED-4 in S. pombe. Knockout of their activity in vivo by means of RNA-mediated inhibition reveals no obvious cell death-related phenotype.
... ;https://doi.org/10.1101https://doi.org/10. /2023 Subsequently, another research that involved the heterologous expression of both murine Bax and Bcl-xL in yeast cells, the cell density was measured with OD660 at several time points, the data revealed that Bcl-xL inhibit the Bax-induced toxicity (Tao et al, 1997). However, these studies have no evidence to define the effect caused by mammalian Bcl-2 family proteins as apoptosis. ...
Preprint
Endothelin convert enzyme-1(ECE-1) plays a significant role in cardiovascular development including four isoforms with unclear function. Therefore, we are interested in studying the function of ECE-1 isoforms through mitochondria due to the high correlation between congenital heart diseases (CHDs) and apoptosis. Since the expression of human Bax and Bcl-xL in budding yeast (Saccharomyces cerevisiae) results in similar effects in mammalian cells, a yeast system was generated for mimicking human Bax-induced apoptosis and the expression of human ECE-1 isoforms was involved. The correlation between Bax-induced growth defect and the candidates of apoptosis via mitochondrial pathway was preliminarily investigated in this system. Furthermore, the phenotypes of ECE-1 isoforms have been identified through yeast growth defect. Individual ECE-1 isoform does not affect yeast growth but act as enhancers for the Bax-induced growth defect. ECE-1c is the strongest enhancer that affect the expression of candidates of outer membrane translocases. This study indicates that ECE-1 might play an important role in inducing apoptosis and we speculate these findings are possible to provide new perspectives with clarifying the mechanism of CHDs.
... No Bcl2 family homologs are constitutively expressed in yeast, but still Bax can kill S.cerevisae and Bcl2 inhibit this death 28 . Thus yeast has been an interesting model to study the constitutive cellular machineries and partners common in Eukaryotes that are implicated in cell death. ...
Article
Full-text available
In this work, we have explored the subcellular localization of Bcl2, a major antiapoptotic protein. In U251 glioma cells, we found that Bcl2 is localized mainly in the ER and is translocated to MAM and mitochondria upon induction of apoptosis; this mitochondrial transfer was not restricted to the demonstrator cell line, even if cell-specific modulations exist. We found that the Bcl2/mitochondria interaction is controlled by TOM20, a protein that belongs to the protein import machinery of the mitochondrial outer membrane. The expression of a small domain of interaction of TOM20 with Bcl2 potentiates its anti-apoptotic properties, which suggests that the Bcl2-TOM20 interaction is proapoptotic. The role of MAM and TOM20 in Bcl2 apoptotic mitochondrial localization and function has been confirmed in a yeast model in which the ER-mitochondria encounter structure (ERMES) complex (required for MAM stability in yeast) has been disrupted. Bcl2-TOM20 interaction is thus an additional player in the control of apoptosis.
... Bak can induce apoptosis in various mammalian cells and a decrease in Bak protein expression was reported in primary colorectal adenocarcinomas and gastric adenocarcinomas when compared with the normal tissue [25,26]. Since Bak and Bax can form a heterodimer with anti-apoptotic Bcl-2 and Bcl-xL and induce apoptosis, Bak maybe a tumor suppressor in some cancers [27,28]. Therefore, reagents that affect the expression of Bak or Bax can control cancer cell growth. ...
Article
Full-text available
Background: Lactoferrin is a glycoprotein with antimicrobial, antioxidant, immune-modulating, antiviral, and most importantly anticancer properties. In the present study, the effect of lactoferrin on breast cancer cell growth and the expression of Bax and Bak genes are evaluated. Materials and Methods: MCF7 cells were cultured in a 96-well plate with 1×105 cells in each well. Different lactoferrin concentrations of 0, 50, 300, 600, and 800 μg/mL were added to each well in three replicates and the well was incubated for 24 hours. After treatment, cell survival was measured using the MTT assay. To determine the level of expression of Bax and Bak genes, the cells were treated with lactoferrin concentrations of 0, 50, and 800 μg/mL in 2 replicates for 24 hours. Then RNA extraction was performed and cDNA was synthesized immediately and the expression of the genes in the presence of beta-actin reference gene and cyber-green fluorescence color was investigated with real-time reactions. Results: The cells viability in lactoferrin concentrations of 0, 50, 300, 500 and 800 μg/μL were 100%, 94%, 83%, 62%, and 32%, respectively. The expression level of the Bax gene at a concentration of 50 μg increased by 2.71 times and in 800 μg concentration decreased by 0.88 times. Also, the expression level of the Bak gene at concentrations of 50 and 800 μg increased by 1.23 and 1.0 fold, respectively. Statistical analysis of the data indicated that the expression levels of two genes at a concentration of 50 μg/mL of lactoferrin significantly increased (P<0.01), compared to the control. The significance level in this study was set at < 0.05. Conclusion: In this study, lactoferrin showed a growth inhibitory effect on breast cancer cells and increased the expression of Bax and Bak genes involved in apoptosis at a concentration of 50 µg/mL.
... The heterologous expression of mammalian pro-apoptotic genes (ced-4, bax, bak) provokes mitochondrial dysfunction and the death of S. cerevisiae and S. pombe cells [93,95,[98][99][100][101]. Several authors reported that this induced-cytotoxicity was characterized by typical phenotypic markers of apoptosis [95,101], identical to those observed in the pseudo-apoptotic mutant cdc48 S565G [92], and other features, such as release of cytochrome c [102]. Moreover, as observed in mammals, coexpression of Bcl-2 or Bcl-xL with Bax abolishes the pseudo-apoptotic phenotype and the cell death [101][102][103][104][105][106]. Since on the one hand, Bax cytotoxicity is increased under respiratory condition [107], and, on the other hand, yeast mutants lacking superoxide dismutase were partially rescued by expression of Bcl-2 [42, 108], it is tempting to speculate that a mitochondrial ROS production is involved in the pseudo-apoptotic process. ...
... Several authors reported that this induced-cytotoxicity was characterized by typical phenotypic markers of apoptosis (Ink, et al., 1997;Ligr, et al., 1998), identical to those observed in the pseudo-apoptotic mutant cdc48 S565G (Madeo, et al., 1997), and other features, such as release of cytochrome c (Manon et al., 1997). Moreover, as observed in mammals, coexpression of Bcl-2 or Bcl-XL with Bax abolishes the pseudo-apoptotic phenotype and the cell death (Jürgensmeier et al., 1997;Ligr, et al., 1998;Manon, et al., 1997;Matsuyama et al., 1998;Tao et al., 1997;Zha et al., 1996a). Since on the one hand, Bax cytotoxicity is increased under respiratory condition (Priault et al., 1999), and, on the other hand, yeast mutants lacking superoxide dismutase were partially rescued by expression of Bcl-2 Longo, et al., 1997), it is tempting to speculate that a mitochondrial ROS production is involved in the bax-induced cell death process. ...
... Several authors reported that this induced-cytotoxicity was characterized by typical phenotypic markers of apoptosis (Ink, et al., 1997;Ligr, et al., 1998), identical to those observed in the pseudo-apoptotic mutant cdc48 S565G (Madeo, et al., 1997), and other features, such as release of cytochrome c (Manon et al., 1997). Moreover, as observed in mammals, coexpression of Bcl-2 or Bcl-XL with Bax abolishes the pseudo-apoptotic phenotype and the cell death (Jürgensmeier et al., 1997;Ligr, et al., 1998;Manon, et al., 1997;Matsuyama et al., 1998;Tao et al., 1997;Zha et al., 1996a). Since on the one hand, Bax cytotoxicity is increased under respiratory condition (Priault et al., 1999), and, on the other hand, yeast mutants lacking superoxide dismutase were partially rescued by expression of Bcl-2 Longo, et al., 1997), it is tempting to speculate that a mitochondrial ROS production is involved in the bax-induced cell death process. ...
... Despite this, animals lacking BAX are viable 9 . BAK, which is closely related to BAX in assayed in vitro systems [10][11][12] , displays widespread tissue distribution similar to BAX. BAK-deficient mice also show normal development, suggesting BAK has redundant functions with other proapoptotic BCL-2 family members 13 . ...
Article
Full-text available
Intrinsic apoptosis relies on the ability of the BCL-2 family to induce the formation of pores on the outer mitochondrial membrane. Previous studies have shown that both BAX and BAK are essential during murine embryogenesis, and reports in human cancer cell lines identified non-canonical roles for BAX and BAK in mitochondrial fission during apoptosis. BAX and BAK function in human brain development remains elusive due to the lack of appropriate model systems. Here, we generated BAX/BAK double knockout human-induced pluripotent stem cells (hiPSCs), hiPSC-derived neural progenitor cells (hNPCs), neural rosettes, and cerebral organoids to uncover the effects of BAX and BAK deletion in an in vitro model of early human brain development. We found that BAX and BAK-deficient cells have abnormal mitochondrial morphology and give rise to aberrant cortical structures. We suggest crucial functions for BAX and BAK during human development, including maintenance of homeostatic mitochondrial morphology, which is crucial for proper development of progenitors and neurons of the cortex. Human pluripotent stem cell-derived systems can be useful platforms to reveal novel functions of the apoptotic machinery in neural development.
Article
Programmed cell death (PCD¹) is a tightly regulated process which is required for survival and proper development of all cellular life. Despite this ubiquity, the precise molecular underpinnings of PCD have been primarily characterized in animals. Attempts to expand our understanding of this process in fungi have proven difficult as core regulators of animal PCD are apparently absent in fungal genomes, with the notable exception of a class of proteins referred to as inhibitors of apoptosis proteins (IAPs²). These proteins are characterized by the conservation of a distinct Baculovirus IAP Repeat (BIR³) domain and animal IAPs are known to regulate a number of processes, including cellular death, development, organogenesis, immune system maturation, host-pathogen interactions and more. IAP homologs are broadly conserved throughout the fungal kingdom, but our understanding of both their mechanism and role in fungal development/virulence is still unclear. In this review, we provide a broad and comparative overview of IAP function across taxa, with a particular focus on fungal processes regulated by IAPs. Furthermore, their putative modes of action in the absence of canonical interactors will be discussed.
Thesis
Caspases are cysteine proteases whose activity is required for the execution of cell death in all multicellular organisms. The morphology of Drosophila cells undergoing cell death is identical to that observed in other organisms, suggesting that Drosophila also have a caspase-containing cell death machinery; however, until recently no Drosophila caspases had been identified. Here I describe the first cloning and characterisation of a Drosophila caspase. I have used a degenerate PCR approach to clone a novel Drosophila melanogaster caspase, which I called drICE. drICE contains all the residues required for caspase activity. Expression of full-length drICE sensitises the S2 Drosophila cell-line to induction of apoptosis by etoposide and cycloheximide treatment; expression of an N-terminally truncated form of drICE, p30drICE, mimicking the proteolytic removal of the prodomain during caspase activation, induces apoptosis in S2 cells. This requires p30drICE caspase activity. drICE is proteolytically activated during S2 cell apoptosis as would be predicted if drICE is part of the Drosophila cell death machinery. drICE auto-processes when expressed in E. coli, and the resulting protein has DEVD-cleaving substrate specificity. The purified active enzyme has two subunits and cleaves lamin DmO and baculovirus p35 in vitro. Lysates of S2 cells undergoing apoptosis contain a caspase activity that can cleave p35, lamin DmO and drICE in vitro and can activate chromatin condensation in added HeLa nuclei. drICE is required for all these activities, suggesting that it has both a role as a necessary 'effector' caspase and as an upstream caspase: whether this is an 'amplifier' or an 'apical' role is unclear. The cloning of drICE described in this thesis should allow genetic screens to identify either upstream regulators or downstream targets of drICE; these in turn should illuminate caspase function in general.
Article
Full-text available
Both rapid B-cell proliferation and programmed cell death (PCD) occur during the differentiation and selection of B cells within the germinal center. To help elucidate the role of Bcl-x in B-cell antigen selection and PCD within the germinal center, we examined its expression in defined B-cell populations and by immunochemistry of tonsil tissue. Purified B-cell fractions enriched for centrocytes express high amounts of Bcl-x and relatively low amounts of Bcl-2, whereas fractions enriched for centroblasts lack significant levels of both proteins. Consistent with this observation, immunocytochemistry localized Bcl-x within cells scattered throughout the germinal center. Stimulation of tonsil B cells with either CD40 or Staphylococcus aureus Cowan increase bcl-x mRNA and protein levels. Treatment of a cell line with a germinal center phenotype (RAMOS) or the tonsillar B-cell centroblast fraction with CD40 rapidly increased Bcl-x levels and partially rescued B cells from PCD. These data suggest that Bcl-x rather than Bcl-2 may rescue centrocytes during selection in the germinal center.
Article
Full-text available
Most members of the Bcl-2 protein family of apoptosis regulating proteins contain two evolutionarily conserved domains, termed BH1 and BH2. Both BH1 and BH2 in the Bcl-2 protein are required for its function as an inhibitor of cell death and for heterodimerization with the proapoptotic protein Bax. In this report, we mapped the region in Bax required for heterodimerization with Bcl-2 and homodimerization with Bax, using yeast two-hybrid and in vitro protein-protein interaction assays. Neither the BH1 nor the BH2 domain of Bax was required for binding to the wild-type Bcl-2 and Bax proteins. Moreover, Bax (ΔBH1) and Bax (ΔBH2) mutant proteins bound efficiently to themselves and each other, further confirming the lack of requirement for BH1 and BH2 for Bax/Bax homodimerization. Bax/Bax homodimerization was not dependent on the inclusion of the NH-terminal 58 amino acids of the Bax protein in each dimerization partner, unlike Bcl-2/Bcl-2 homodimers which involve head-to-tail interactions between the region of Bcl-2 where BH1 and BH2 resides, and an NH-terminal domain in Bcl-2 that contains another domain BH4 which is conserved among antiapoptotic members of the Bcl-2 family. Similarly, heterodimerization with Bcl-2 occurred without the NH-terminal domain of either Bax or Bcl-2, suggesting a tail-to-tail interaction. The essential region in Bax required for both homodimerization with Bax and heterodimerization with Bcl-2 was mapped to residues 59-101. This region in Bax contains a stretch of 15 amino acids that is highly homologous in several members of the Bcl-2 protein family, suggesting the existence of a novel functional domain which we have termed BH3. Deletion of this 15-amino acid region abolished the ability of Bax to dimerize with itself and to heterodimerize with Bcl-2. The findings suggest that the structural features of Bax and Bcl-2 that allow them to participate in homo- and heterodimerization phenomena are markedly different, despite their amino-acid sequence similarity.
Article
Bcl-2 is a mitochondrial- and perinuclear-associated protein that prolongs the lifespan of a variety of cell types by interfering with programmed cell death (apoptosis). Bcl-2 seems to function in an antioxidant pathway, and it is believed that membrane attachment mediated by a COOH-terminal hydrophobic tail is required for its full activity. To identify critical regions in bcl-2 alpha for subcellular localization, activity, and/or interaction with other proteins, we created, by site-directed mutagenesis, various deletion, truncation, and point mutations. We show here that membrane attachment is not required for the survival activity of bcl-2 alpha. A truncation mutant of bcl-2 alpha lacking the last 33 amino acids (T3.1) including the hydrophobic COOH terminus shows full activity in blocking apoptosis of nerve growth factor-deprived sympathetic neurons or TNF-alpha-treated L929 fibroblasts. Confocal microscopy reveals that the T3 mutant departs into the extremities of neurites in neurons and filopodias in fibroblasts. Consistently, T3 is predominantly detected in the soluble fraction by Western blotting, and is not inserted into microsomes after in vitro transcription/translation. We further provide evidence for motifs (S-N and S-II) at the NH2 and COOH terminus of bcl-2, which are crucial for its activity.
Article
We used a yeast genetic screen to isolate cDNAs that encode a protein, SRF accessory protein-1 (SAP-1), that is recruited to the c-fos serum response element (SRE) as part of a ternary complex that includes serum response factor (SRF). SAP-1 requires DNA-bound SRF for ternary complex formation and makes extensive DNA contacts to the 5′ side of SRF, but does not bind DNA autonomously. Ternary complex formation by SAP-1 requires only the DNA-binding domain of SRF, which can be replaced by that of the related yeast protein MCM1. We isolated cDNAs encoding two forms of SAP-1 protein, SAP-1a and SAP-1b, which differ at their C termini. Both SAP-1 proteins contain three regions of striking homology with the elk-1 protein, including an N-terminal ets domain. Ternary complex formation by SAP-1 requires both the ets domain and a second conserved region 50 amino acids to its C-terminal side. SAP-1 has similar DNA binding properties to the previously characterized HeLa cell protein .
Article
In mammalian cells, the Bcl-2 and Bcl-x(L) proteins suppress programmed cell death whereas the topographically similar Bax protein accelerates the apoptotic process. Recently published data suggest that expression of the human Bax-α gene is lethal for the yeast can be overcome by co-expressing Bcl-2 or Bcl-x(L). Our findings corroborate these results. However, we find that although Bax induction invariably stops cell growth under all circumstances, it does not lead to death in ‘petite’ cell. Petites cannot respire because they lack functional mitochondria. It seems that in ‘grande’ cells, which do possess normal mitochondrial DNA, nutritional limitation is critical for increased mortality. Surprisingly, murine Bcl-2 lacking the membrane anchor of human Bcl-2 has no effect on grande cells, but can efficiently rescue petites in rich medium. It has been suggested that the C-terminal membrane anchor of human Bcl-2 may have a crucial role in rescuing apoptosis in mammalian cells. When murine Bcl-2 is fused to the membrane anchor of yeast mitochondrial Mas70 protein, the Bcl-2 variant mBcl-2-mma rescues not only petites but also grandes, just like human Bcl-x(L). The rescuing ability of Bcl-x(L), which contains its own membrane anchor, surpasses that of mBcl-2-mma. Our results indicate that the process involving Bax-induced growth inhibition followed by possible lethality, and the rescuing effect of Bcl-2 or Bcl-x(L) is linked to yeast mitochondrial function. We propose a model which is consistent with these observations.
Article
Cowpox virus effectively inhibits inflammatory responses against viral infection in the chick embryo. This study demonstrates that one of the viral genes necessary for this inhibition, the crmA gene (a cytokine response modifier gene), encodes a serpin that is a specific inhibitor of the interleukin-1 beta converting enzyme. This serpin can prevent the proteolytic activation of interleukin-1 beta, thereby suppressing an interleukin-1 beta response to infection. However, the modification of this single cytokine response is not sufficient to inhibit inflammatory responses. This suggests that cowpox virus encodes several cytokine response modifiers that act together to inhibit the release of pro-inflammatory cytokines in response to infection. These viral countermeasures to host defenses against infection may contribute significantly to the pathology associated with poxvirus infections.
Article
The gene ced-9 of the nematode Caenorhabditis elegans acts to protect cells from programmed cell death. A mutation that abnormally activates ced-9 prevents the cell deaths that occur during normal C. elegans development. Conversely, mutations that inactivate ced-9 cause cells that normally live to undergo programmed cell death; these mutations result in embryonic lethality, indicating that ced-9 function is essential for development. The ced-9 gene functions by negatively regulating the activities of other genes that are required for the process of programmed cell death.