029-030 genetika implantacije (14)

  • Upload
    cf94

  • View
    217

  • Download
    0

Embed Size (px)

Citation preview

  • 8/13/2019 029-030 genetika implantacije (14)

    1/19

    Molecular Mechanisms of Trophoblast Survival:From Implantation to Birth

    Andrea Jurisicova,* Jacqui Detmar and Isabella Caniggia

    INTRODUCTIONProgrammed cell death (PCD) is an

    evolutionarily conserved processthat has been documented to play a

    vital role during development inmany diverse species including

    mammals, amphibians, and in-sects. It is commonly known that

    cell death is required for adjustingthe number of cells and for remov-

    ing unnecessary organs such as thetadpole tail during amphibian

    metamorphosis, disintegration oflarval organs during insect meta-

    morphosis, elimination of sex-spe-cific organs such as the Mullerian

    duct in males or the Wolffian duct infemales, as well as formation of the

    digits in mammals (reviewed in Ja-cobson et al., 1997). Cell death is

    also a driving force behind cellularsculpturing of embryonic struc-

    tures, such as the creation of the

    proamniotic cavity and the forma-

    tion of tubular structures (Coucou-

    vanis and Martin, 1995) including

    neural tube closure (Weil et al.,

    1997) and development of the earcanal (Leon et al., 2004), tooth re-

    modeling (Matalova et al., 2004),

    and selection of immune cells (Vaux

    and Korsmeyer, 1999; Ranger et

    al., 2001), as well as elimination of

    misplaced, injured, or otherwise

    dangerous cells such as primordial

    germ cells (Molyneaux and Wylie,

    2004). While this programmed cell

    death is not always apoptotic in na-

    ture, as both necrotic-like and au-

    tophagic-like death have been ob-

    served to occur in some organs

    (Chautan et al., 1999; Baehrecke,2003), it is always tightly regulated

    by conserved molecular pathways

    that guard the decision-making and

    execution of cell death machinery

    (Assuncao Guimaraes and Linden,2004; Levine and Klionsky, 2004).

    Thus, it is obvious that a misbal-ance in cell death can have profound

    developmental consequences with awide spectrum of phenotypes, rang-

    ing from mild developmental delay,retardation, induction of congenital

    malformations, and defects or even

    death of the embryo. In such cases,either too little or too much deathcan have detrimental consequences.

    Developmental cell death also ap-

    pears to be evolutionarily conserved,as embryos from invertebrates such

    as C. elegans and Drosophila up tomammals express and utilize similar

    molecules to activate, execute, andfinalize cell death, although the com-

    plexity and redundancy in mamma-lian system is far more explicit (Meier

    et al., 2000; Twomey and McCarthy,2005).

    The core cell death pathways havebeen extensively reviewed else-

    where (Danial and Korsmeyer,2004; Tran et al., 2004; Kroemer

    and Martin, 2005; Strasser, 2005),and thus are not discussed here. In-

    stead, this review focuses on our cur-rent understanding of these regula-

    tory pathways in the blastocyst, witha focus on the trophoblast lineage of

    the developing placenta both prior toand after implantation.

    Cell Death Prior to

    Implantation

    Successful pre- and postimplanta-tion embryonic development de-

    Fetal development depends upon a coordinated series of events in boththe embryo and in the supporting placenta. The initial event in placenta-tion is appropriate lineage allocation of stem cells followed by the forma-tion of a spheroidal trophoblastic shell surrounding the embryo, facilitat-ing implantation into the uterine stroma and exclusion of oxygenatedmaternal blood. In mammals, cellular proliferation, differentiation, anddeath accompany early placental development. Programmed cell death isa critical driving force behind organ sculpturing and eliminating abnormal,misplaced, nonfunctional, or harmful cells in the embryo proper, althoughvery little is known about its physiological function during placental de-

    velopment. This review summarizes current knowledge of the cell deathpatterns and molecular pathways governing the survival of cells within theblastocyst, with a focus on the trophoblast lineage prior to and afterimplantation. Particular emphasis is given to human placental develop-ment in the context of normal and pathological conditions. As molecularpathways in humans are poorly elucidated, we have also included anoverview of pertinent genetic animal models displaying defects in tropho-blast survival. Birth Defects Research (Part C) 75:262280, 2005. 2006 Wiley-Liss, Inc.

    Andrea JurisicovaandIsabella Caniggiaare from the Department of Obstetrics and Gynecology, University of Toronto, Mount SinaiHospital, and the Department of Physiology, University of Toronto, Toronto, Ontario, Canada.Jacqui Detmar is from the Institute of Medical Studies, University of Toronto, Toronto, Ontario, Canada.

    *Correspondence to: A. Jurisicova, SLRI876, Mount Sinai Hospital, 600 University Avenue, Toronto, Ontario, Canada, M5G 1X5.E-mail: [email protected]

    Published online in Wiley InterScience (www.interscience.wiley.com). DOI: 10.1002/bdrc.20053

    REVIEW

    Birth Defects Research (Part C) 75:262280 (2005)

    2006 Wiley-Liss, Inc.

  • 8/13/2019 029-030 genetika implantacije (14)

    2/19

    pends on the sequential implemen-

    tation of a certain genetic program,

    which is profoundly influenced not

    only by the genetic endowment ofthe gametes, but also by interac-

    tions with the surrounding environ-

    ment. For one in 10 couples, failure

    to achieve a pregnancy remains amajor problem. Clinical results of

    assisted reproductive technology(ART) procedures in North America

    suggest that clinical pregnancy is

    achieved in only 30% of patientsundergoing this treatment. This low

    rate of reproductive fitness is not

    restricted to infertility patients,since in the general population up

    to 40% of conceptions may be lost

    before they are clinically recog-

    nized as pregnancy (Hertig et al.,

    1959). The cellular and molecularreasons behind such a high rate of

    failure are unclear; however, lowdevelopmental competence of con-

    ceived embryos is believed to be

    the most likely culprit. While it iswell recognized that a significant

    proportion of human embryos dis-

    play a variety of cellular defects, in-cluding embryo fragmentation

    and/or cleavage arrest, abnormal

    cavitation, depletion of cells, or ab-normal lineage allocation, the un-

    derlying molecular mechanisms re-

    sponsible for these deficiencies areunclear.

    Successful development of mam-malian preimplantation embryos cul-

    minates in the first major step to-ward embryonic differentiation, that

    being the formation of the trophecto-

    derm (TE) and the inner cell mass(ICM) at the blastocyst stage. The

    polar trophectoderm, a fluid-trans-porting epithelium, is the precursor

    of the majority of the extraembry-

    onic placental tissues (Kunath et al.,

    2004). On the other hand, cells of theICM, comprising approximately onefourth of the total cell count in the

    human expanded blastocyst (Hardy

    et al., 1989; Hardy, 1997), contrib-ute to the epiblast (e.g., embryo

    proper) and are represented by em-bryonic stem cell population (ES).

    The third embryonic lineage, estab-

    lished in fully expanded blastocystsand located on the surface of the ICM

    layer, called the primitive endoderm(PE), represents a population of cells

    that will give rise to another ex-

    traembryonic tissue called parietal

    and visceral endoderm (Kunath et

    al., 2005). Murine stem cells for allthese lineages have been success-

    fully established and a subset of ge-

    netic pathways, responsible for the

    original allocation of these lineages,

    has also been elucidated (Ralstonand Rossant, 2005). However, the

    contribution of cell death to develop-ment at this stage has been poorly

    explored.

    There are two windows for celldeath susceptibility during preim-

    plantation development: early cleav-

    age stage, frequently described asembryonic arrest with or without cel-

    lular fragmentation (reviewed in Ju-

    risicova and Acton, 2004), and apop-totic-like death coinciding with

    formation of the blastocyst (Hardy,1997; Pampfer and Donnay, 1999).

    While cell death at the blastocyststage has been documented for

    over 20 years, its regulation and

    function in early developing em-bryos remain poorly understood.

    Cells exhibiting characteristic fea-

    tures of apoptosis have been ob-served in blastocysts of several

    mammalian species (Handyside,

    1986; Papaioannou and Ebert,1988; Knijn et al., 2003), including

    human (Hardy et al., 1989); this

    cellular phenomenon has been ob-served in embryos developing in

    vivo, but appears to be elevated byconditions used during in vitro cul-

    ture (Jurisicova et al., 1998; Gjor-ret et al., 2003).

    The presence of dead and dying

    cells has been confirmed in blasto-cysts from a number of mammalian

    species through the use of severaltechniques, implicating apoptosis

    as a mode of death during blasto-

    cyst formation (reviewed in Hardy,

    1997, 1999). Positive terminaltransferase mediated dUTP DNAEnd Labeling (TUNEL) labeling, in-

    dicative of fragmented DNA as well

    as condensed chromatin morphol-ogy, in a subset of cells in the

    mouse blastocyst has confirmedthat peak levels of PCD in murine

    embryos developing in vivo occur at

    97 hr postcoitum (Handyside,1986). Culture of murine preim-

    plantation embryos in vitro fromzygote until blastocyst stage in

    mice elevates cell death indices

    (percentage of dead cells), with

    death restricted predominantly to

    cells of the ICM lineage (Brison andSchultz, 1997; Hardy, 1997). This

    is even more evident in embryos of

    different mammalian species that

    have been conceived, manipulated,

    and cultured in vitro (Jurisicova etal., 1998; Gjorret et al., 2003; Hao

    et al., 2004a). Under these condi-tions, death occurs in both the ICM

    and in the TE lineage (reviewed in

    Hardy, 1997). The susceptibility todeath of these two different lin-

    eages (i.e., ICM and TE) are likely

    influenced by the mode of fertiliza-tion, as both human and murine

    blastocysts obtained byin vitrofer-

    tilization (IVF) display trophecto-dermal cell death (Jurisicova et al.,

    1998, 1999), while murine blasto-cysts conceived in vivo, but cul-

    tured in vitro, have almost exclu-sive localization of dead cells to the

    ICM (Hardy, 1997).

    Based on cell counts of spare hu-man embryos donated to research,

    we have observed that the cell

    death index (CDI) contributes tothe loss of approximately 15% of

    cells at the blastocyst stage (Jurisi-

    cova et al., 1999). In most of theembryos, cell death appeared ran-

    dom, affecting both the ICM and

    trophectoderm; however, a sub-population of blastocysts exhibited

    depletion of the majority of cellscomprising the ICM, as evidenced

    by extensive chromatin condensa-tion and/or fragmented DNA (see

    Fig. 1). This could likely result in the

    total elimination of the embryoniclineage destined to form the fetus,

    leaving only precursor cells for theplacenta. Such blastocysts may ini-

    tiate implantation, but would prob-

    ably create only extraembryonic

    tissues with an empty embryonicsac, a condition clinically termed a

    blighted ovum. Lack of an ICM

    has been previously described in

    human embryos cultured in vitro(Winston et al., 1991; Desai et al.,

    1997), and was attributed to theabnormal allocation of cells to the

    ICM and TE lineages. We propose

    that in some embryos, cells of theICM become established but are

    rapidly eliminated via PCD in theearly developing blastocyst. Since

    cells of the TE have extremely well-

    MOLECULAR MECHANISMS OF TROPHOBLAST SURVIVAL 263

    Birth Defects Research (Part C) 75:262280, (2005)

  • 8/13/2019 029-030 genetika implantacije (14)

    3/19

    developed phagocytotic capabilities

    (Drake and Rodger, 1987), apopto-tic bodies produced by dying cells of

    the ICM are most likely engulfedand degraded very rapidly (Hardy,

    1997). Therefore, the remainingcells of the failing blastocyst would

    not be negatively affected by cellu-lar debris produced by the dead

    cells following secondary necrosis.It is interesting to note that the lev-

    els of cell death in human blasto-cysts were previously observed to

    be elevated in the ICM of high-grade embryos in comparison to

    low grade embryos, suggesting

    that apoptosis may play a regula-tory role in the maintenance of cellnumber during normal healthy em-

    bryo development (Hardy et al.,2003).

    Further examination of CDI anddistribution of dead cells identified

    an additional subgroup of human

    blastocysts characterized by exces-sive activation of the cell death cas-

    cade in both ICM and TE lineages(Fig. 1). These blastocysts would

    likely be unable to initiate and/orsustain postimplantation develop-

    ment and, therefore, would not be

    clinically recognized as a preg-nancy. However, they may contrib-

    ute to the pool of biochemicalpregnancies, which are frequently

    observed after IVF (Simon et al.,1999). In these cases, patients

    manifest mildly elevated humanchorionic gonadotrophin (hCG) and

    have delayed menstrual period.However, the hCG, which reflects

    the activity of cells from the tropho-blast lineage, declines within a few

    days thereafter, followed by men-strual period, indicating loss of

    pregnancy. The incidence of cell

    death in the human blastocyst hasadditionally been shown to be di-rectly correlated with early embryo

    quality (Hardy et al., 1989; Stoneet al., 2005), and deregulated cell

    death very likely contributes to lowdevelopmental potential of human

    embryos.

    Growth Factors, Nutritional

    Needs, and Cell Death

    In contrast to the in vitro environ-ment, the in vivo microenviron-

    ment provides additional growth

    factors and cytokine mediated ef-fectors that positively influence

    embryo development and growth.Thus, embryo development is con-

    trolled not only by innate factors,but also through autocrine and

    paracrine effectors (reviewed byBrison, 2000; Hardy and Spanos,

    2002). Differences between cleav-age rates (Bowman and McLaren,

    1970) and levels of apoptosis (Bri-son and Schultz, 1997) in in vivo

    and in vitro cultured mouse em-bryos suggest that the maternal

    environment is exerting a paracrine

    effect on the preimplantation stageembryo. Studies of in vitro culturedembryos have further demon-

    strated positive autocrine effects,either through reducing culture vol-

    ume or by grouped embryo culture(Lane and Gardner, 1992; Brison

    and Schultz, 1997). Supplementa-

    tion of culture media with variousgrowth factors has resulted in ele-

    vated rates of blastocyst formationaccompanied by increased cell

    number, likely resulting from sup-pression of cell death. Transform-

    Figure 1. Schematic drawing of mammalian embryodevelopment from morula to blastocyst (TE, trophectoderm; ICM, inner cell mass;PE, primitive endoderm). Three patterns of cell death in human blastocyst observed under in vitro culture aredescribed. Representativephotomicrographs are shown for expanded human blastocysts at day 7 that were analyzed for cell death by assessing chromatin status(DNA stain in blue) and fragmentation of DNA (TUNEL labeling in red). Normal nuclei have diffuse pale-blue staining and ovalmorphology, while nuclei with condensed chromatin, indicative of dead cells, exhibit bright white staining and disintegrate into smallround nuclear blebs. Cell death index (CDI) reflects the proportion of cells in the embryo displaying hallmarks of apoptosis.

    264 JURISICOVA ET AL.

    Birth Defects Research (Part C) 75:262280, (2005)

  • 8/13/2019 029-030 genetika implantacije (14)

    4/19

    ing growth factor-(TGF-) (Brison

    and Schultz, 1997), platelet acti-

    vating factor (PAF) (ONeill, 1998),

    and insulin-like growth factor-1(IGF-1) (Brison, 2000) have been

    shown to decrease the incidence of

    apoptotic cells in mouse and human

    blastocysts, while tumor necrosisfactor (TNF-) has been shown to

    exert the opposite effect in rat(Pampfer et al., 1997) and mouse

    embryos (Wuu et al., 1999; Fabian

    et al., 2004). Animal model in vitrostudies demonstrated that addition

    of IGF-1 to the culture medium re-

    sulted in a significant decrease in

    the number of apoptotic cells perembryo, mostly in cells of the ICM

    lineage (Makarevich and Markkula,

    2002), while supplementation with

    growth hormone exerted similar ef-fects targeting the TE (Kolle et al.,

    2002, 2003). We and others haveexplored the biological effects of

    these growth factors in human pre-

    implantation embryos with similaroutcomes, suggesting conservation

    of these molecular pathways. The

    percentage of apoptotic nuclei inhuman blastocysts was decreased

    by approximately 50% upon addi-

    tion of IGF-1 to culture (Spanos etal., 2000). Addition of granulocyte-

    macrophage colony stimulating

    factor (GM-CSF) to human embryocultures increased blastocyst for-

    mation rates two-fold (Sjoblom etal., 2002) and decreased the per-

    centage of apoptotic nuclei, whileincreasing the number of viable

    ICM cells (Sjoblom et al., 2002).

    Given these data, we focused ourefforts on analysis of the growth

    hormone pathway. Human em-bryos express growth hormone re-

    ceptor and respond to it in a similar

    way as murine embryos, exhibiting

    a higher number of cells allocatedto both ICM and TE lineages (Di Be-

    rardino, 2004). Collectively, thesedata further support the important

    role of autocrine and paracrine me-diators in human embryonic cell

    death and development (Hardy and

    Spanos, 2002).While the effects of cytokines on

    embryo growth and developmenthave been known for several years,

    the downstream effects on gene

    expression in the context of embry-onic cell death remain relatively un-

    explored. The inhibiting activity of

    TGF- in murine blastocysts was re-

    cently linked to the antiapoptotic

    action of the inhibitor of apoptosis(IAP) molecule, survivin, and is me-

    diated by activation of the phos-

    phatidylinositol-3 kinase (PI3K)

    signaling pathway (Kawamura etal., 2003, 2005). The PI3K/Akt sig-

    nal transduction pathway is a well-known mediator of growth promot-

    ing and cell survival signals (Gross

    et al., 2005). Recent reports sug-

    gest that both PI3K and Akt exhibitan apical staining pattern in tro-

    phectoderm cells, and inhibition of

    Akt activity resulted in reduced in-sulin-stimulated glucose uptake,

    leading to a significant delay in

    blastocyst hatching (Riley et al.,

    2005). The site of expression ofthese prosurvival signaling mole-

    cules appears to coincide with theexpression of Death receptor

    5/Killer and its ligand Trail (Riley et

    al., 2004), raising a possibility that

    these molecules may work in onesignaling pathway.

    The exact effector death pathway

    used by IGF to suppress cell deathin ICM is less understood. IGF-I ex-

    posure appears to change the ratioof Bax/Bcl-2, and this shift in bal-

    ance between pro- and antiapop-

    totic Bcl-2 family members wasproposed to contribute to de-

    creased apoptosis (Kolle et al.,2002). However, preimplantation

    embryos exposed to high concen-

    trations of IGF-1 or insulin also un-dergo extensive apoptosis of the

    ICM, and deficiency in Bax, or ex-posure to inhibitors of either

    caspase or ceramide synthase ac-

    tivity, prevents this event (Chi etal., 2000). Furthermore, it has

    been proposed that the embryo-

    toxic insult caused by high IGF-1levels may be responsible for the

    high incidence of pregnancy lossseen in women with polycystic

    ovary syndrome (Chi et al., 2000),as these patients exhibit insulin re-

    sistance and show elevated levels

    of IGF-1 due to insufficient produc-tion of IGF-1 binding protein

    (Conover, 1992).On the other hand, imbalance in

    glucose metabolism has been

    linked to excessive activation ofseveral death pathways in preim-

    plantation embryos. Cell death in-duced by elevated exposure to glu-

    cose in vitro downregulates glucose

    transporters, leading to a drop inintraembryonic free glucose (Chi et

    al., 2002) and triggering massive

    apoptosis in the ICM lineage (Pam-

    pfer et al., 1997). This pathwayalso involves excessive accumula-

    tion of Bax (Moley et al., 1998) andcleaved caspase 3/6 (Hinck et al.,

    2001, 2003), as well as promotes

    activation of the p53 dependent cell

    death pathway (Keim et al., 2001).Disruption of any of these pathways

    by gene knockout or biochemical

    inhibition approaches partially res-cues cell death triggered by intra-

    cellular glucose starvation (Keim et

    al., 2001). Moreover, the diabetic

    mouse

    in vivo model caused by de-struction of islet cells by streptoco-

    zin treatment results in recapitula-tion of these embryonic phenotypes

    (Chi et al., 2000, 2002).

    Given these data, hence the

    question arises, what is the func-tion of cell death during early em-

    bryo development? While several

    speculations were put forward,very little experimental data exist

    to support these theories. Mostlikely, it is a means to eliminate

    cells with abnormal genetic com-

    plements, reduced developmentalpotential (Hardy, 1999), and/or de-

    fects in lineage allocation. The reg-ulation of cell death at these early

    stages is further thought to be crit-

    ical to later development of theconceptus (Moley, 2001). The exis-

    tence of this link was confirmed by aseries of elegant experiments that

    induced cell death during preimplan-

    tation embryo development by dis-rupting glucose metabolism. Both in

    vitro and in vivo exposure to hyper-

    glycemic environment results in in-creased apoptosis observed in the

    blastocyst, leading to an increasednumber of malformations and con-

    genital abnormalities such as micro-gnathia, omphalocele, and cranial

    and neural tube defects; these ab-

    normalities could be prevented byablation of the cell death initiator,

    Bax (Moley et al., 1998; Chi et al.,

    2000). At least one report in humansalso indicates that transfer of em-

    bryos with increased cellular frag-mentation, previously linked to the

    MOLECULAR MECHANISMS OF TROPHOBLAST SURVIVAL 265

    Birth Defects Research (Part C) 75:262280, (2005)

  • 8/13/2019 029-030 genetika implantacije (14)

    5/19

  • 8/13/2019 029-030 genetika implantacije (14)

    6/19

    bryonic carcinoma cell lines of

    solely embryonic or trophectoder-

    mal developmental potential were

    transplanted into the blastocelecavity of mouse blastocysts indi-

    cated that cells capable of forming

    TE are preferentially eliminated by

    PCD (Pierce et al., 1989). Extracel-lular hydrogen peroxide generated

    via polyamine oxidation was impli-cated as a mediator of PCD in the

    blastocyst; however, genes re-

    sponsible for the activation or exe-cution of this death are unknown

    (Pierce et al., 1991). Thus, it is

    tempting to speculate that PARP-1is a key regulator of cell death in

    ICM cells that have potential to be-

    come TE lineage. In wild-type em-

    bryos, PARP-1 would be activated

    in the response to DNA damagecaused by hydrogen peroxide in the

    subset of ICM considering their TEfate due to their low antioxidant

    status perhaps, which would lead to

    their elimination. ICM cells of em-bryos lacking PARP-1 fail to die

    when they switch the fate, leading

    to formation of trophoblast lineagein vitro as well as in vivo produced

    carcinoma tumors (Masutani et al.,

    2001). This surveillance pathwaymay not be functional in humans,

    as human ES cells readily differen-

    tiate into the trophoblast lineageunder in vitro conditions without

    any genetic manipulation (Xu et al.,2002).

    Myeloid cell leukemia-1 (Mcl-1) isa prosurvival Bcl-2 family member

    facilitating proper differentiation of

    stem cells (Opferman et al., 2003,2005). Since Mcl-1 has been pro-

    posed to work upstream of otherBcl-2 family members (Nijhawan et

    al., 2003), it is not surprising that

    embryonic disruption of this mole-

    cule leads to preimplantation le-thality in mice, well before themidgestational death of embryos

    caused by Bcl-x deletion (Mo-

    toyama et al., 1995). While blasto-cysts lacking Mcl-1 could be de-

    tected at day 4 in the reproductivetract in vivo, these embryos failed

    to implant and were unable to pro-

    duce viable blastocyst outgrowthsin vitro (Rinkenberger et al., 2000),

    suggesting a compromised TE lin-eage viability. No overt increase in

    apoptosis was observed in these

    blastocysts and embryonic loss

    could not be rescued by the disrup-tion of Bax or p53, indicating that

    Mcl-1 may not act through theseapoptotic pathways and/or may

    have other cellular function(s). Wehave previously observed that an-

    other proapoptotic multichannelBcl-2 family member, Mtd/Bok, is

    abundantly expressed in murine

    blastocyst, particularly in the TElineage (Jurisicova et al., 2000), a

    finding that will be discussed laterin the context of human placenta.

    As the preferred Bcl-2 interactivepartner of Mtd is Mcl-1 (Hsu et al.,

    1997), it is possible that deletion ofthis particular cell death gene may

    be able to rescue Mcl-1 lethality inthe early developing embryo.

    Defender against apoptosis-1(Dad-1) is an evolutionarily highly

    conserved enzyme, which catalyzesthe transfer of pre-assembled high

    mannose oligosaccharides ontospecific asparagine residues of nas-

    cent polypeptides (Sanjay et al.,1998). This enzyme is capable of

    interacting with and modifyingMcl-1 function (Makishima et al.,

    2000). The functional importanceof this interaction can be indirectly

    shown from results of gene target-ing experiments, as disruption of

    this gene leads to embryonic lethal-ity at the blastocyst stage (Nishii et

    al., 1999; Brewster et al., 2000),phenocopying the Mcl-1 knockout

    mouse. Dad-1 mutants, however,display an increased rate of cell

    death in the ICM both in vitro and in

    vivo, suggesting that this enzymemay have additional modification

    targets besides Mcl-1.

    THE PLACENTA AND FETAL

    PROGRAMMING

    In the last decade, substantial evi-dence has supported the idea that a

    compromised in utero environmentcan influence health during postna-

    tal life. Barker (1992) introduced

    the concept of fetal programming,which proposes that a number of

    organ structures and functions un-dergo programming during embry-

    onic and fetal life. This develop-mental programming determines

    the physiologic and metabolic setpoints, which will ultimately cue re-

    sponses to the environment in the

    adult. Adverse in utero conditions,

    such as placental insufficiency, in-

    adequate maternal nutrition, and

    altered maternal stress hormone

    profiles lead to developmental ad-

    aptations by the embryo/fetus that

    readjust these set points (reviewedby Lau and Rogers, 2004; Wobus

    and Boheler, 2005). These adaptive

    measures ostensibly have short-

    term benefits to the embryo and fe-

    tus, but these changes to the ge-

    netically-determined body plan

    may confer a discordant physiology

    on the adult, leading to increased

    risk of disease. It has been repeat-

    edly shown in human populations

    that growth-compromised fetuses

    resulting from intrauterine growth

    restriction (IUGR) following placen-tal insufficiency are at increased

    risk for adverse short- and long-

    term outcomes, such as hyperten-

    sion, obesity, and type II diabetes,

    that can extend into adult life

    (Barker, 1992; Bernstein et al.,

    2000; Clausson et al., 2001). There

    is a rapidly-growing body of evi-

    dence suggesting that the placenta

    is involved in fetal programming

    (reviewed by Godfrey, 2002; Bas-

    chat, 2004), with particular empha-

    sis on the impact of placental insuf-ficiency on cardiac development

    (reviewed by Law and Shiell, 1996;

    Huxley et al., 2000), neural devel-

    opment (Blair and Stanley, 1988;

    Mallard et al., 1999; Bui et al.,

    2002) and endocrinology (reviewed

    by Kanaka-Gantenbein et al.,

    2003). Proper establishment of the

    placenta at these times allows the

    fetus to grow and attain its devel-

    opmental potential in the third tri-

    mester, in preparation for postnatal

    life. Investigations into how normal

    placental cell death assists in shap-

    ing placental architecture and func-

    tion will provide us with information

    to better understand how aberrant

    or insufficient cell death in this tran-

    sient organ contributes to overall

    fetal, infant, and adult health. Ad-

    ditionally, the study of animal mod-

    els of placentation provides useful

    clues and allows us profound in-

    sight into the regulation and pat-

    terning of cell death in the mamma-

    lian placenta.

    MOLECULAR MECHANISMS OF TROPHOBLAST SURVIVAL 267

    Birth Defects Research (Part C) 75:262280, (2005)

  • 8/13/2019 029-030 genetika implantacije (14)

    7/19

    Human Placental

    Development

    The placenta is a life-sustaining

    though transient organ, which medi-

    ates the physiological exchange be-

    tween mother and fetus. During

    human placental development, un-

    differentiated mononuclear cytotro-

    phoblast (CT) cells reside in two dif-

    ferent types of highly specialized

    chorionic villi: floating and anchoring

    villi. These highly proliferative, yet,

    undifferentiated trophoblast cells fol-

    low two separate pathways of differ-

    entiation (Cross et al., 1994). In

    floating villi, these cells have the

    unique ability to fuse and form the

    multinucleated syncytiotrophoblast

    (ST) layer, which functions as the

    natural physical barrier between

    maternal and fetal cells, and be-

    cause it is directly bathed in ma-

    ternal blood, it mediates gas and

    nutrient exchange for the devel-

    oping fetus. This layer is subjected

    to continuous renewal, as aged

    nuclei together with small

    amounts of cytoplasm are re-

    leased into the maternal circula-

    tion as membrane-sealed struc-

    tures termed syncytial knots,

    and are eventually believed to be

    cleared by maternal lung macro-

    phages. This dynamic process,

    also known as trophoblast turn-

    over, is necessary, as the multinu-

    cleated ST cells are highly differ-

    entiated and, as such, unable to

    regenerate (Huppertz and King-

    dom, 2004).

    In anchoring villi, proliferative

    CT cells differentiate and invade

    deeply into the endometrium

    reaching the first third of the ad-

    jacent myometrium. This popula-

    tion of migratory/invasive cells,also known as extravillous tropho-

    blasts (EVT), have the unique fea-

    ture of remodeling the pregnant

    endometrium and its vasculature.

    Endovascular invasion within the

    uterine wall is necessary, since

    the remodeling of the maternal

    spiral arteries generates a low re-

    sistance vascular system that per-

    mits continuous and adequate

    blood flow to the growing fetus

    (Aplin, 1991).

    Cell Death in the Human

    Placenta

    During normal placentation, a bal-

    ance between proliferation, differen-

    tiation, and apoptosis is mandatoryfor trophoblast cells homeostasis, fa-

    cilitating maintenance of normalplacental villi functions. While pro-

    liferation and differentiation of tro-phoblast cells have been exten-

    sively examined, only in the past

    decade have studies begun to ad-dress the importance of apoptosis

    during normal and abnormal pla-

    cental development.Numerous studies have reported

    that during pregnancy, the rate of

    cell death is low in the first trimes-ter of pregnancy and mostly re-

    stricted to the chorionic villous cy-totrophoblasts, followed by anincrease with advancing gestation

    and a shift to the syncytium (Smith

    et al., 1997; Gruslin et al., 2001).

    ST cells, and to a lesser extent CTcells, express the prosurvival Bcl-2

    family members Bcl-2 and Mcl-1

    (Huppertz et al., 1998), while theproapoptotic proteins Bax and p53

    have been found in cytotropho-

    blasts and mesenchymal cells ofthe chorionic villi throughout gesta-

    tion (Ratts et al., 2000). This

    unique spatial distribution of pro-and antiapoptotic molecules be-

    tween the adjacent cell layers hasbeen postulated to confine the ap-

    optotic events to a subset of spe-cific nuclei within the renewing syn-

    cytium, thereby preventing the

    death of the entire layer. Interest-ingly, a large number of apoptotic

    nuclei in syncytial knots show de-creased expression of the anti-ap-

    optotic Bcl-2 and Mcl-1 (Huppertz

    et al., 1998). Downstream events

    of death execution and syncytialknot formation appear to be medi-ated by Caspase 3 (Huppertz et al.,

    1999). Thus, it is not surprising that

    the expression of the X-linked in-hibitor of apoptosis (Xiap), a pro-

    survival inhibitor of apoptotic pro-

    tein known to interfere withexecutor caspase activation, was

    found to decrease with advancinggestation, and it inversely corre-

    lated with increased trophoblast

    apoptosis found at term (Gruslin etal., 2001). Others have reported

    abundant Bcl-2 expression in con-

    junction with apoptotic nuclei in

    syncytiotrophoblast overlying fi-brin-containing fibrinoid deposition

    (Ratts et al., 2000). Since this ex-

    tracellular matrix molecule is in-

    volved in reparative and degenera-

    tive processes within the humanplacenta, Bcl-2 may be involved in

    these events (Marzioni et al.,1998). More recently, studies have

    highlighted the importance of cell

    death in regulating the events in-volved in trophoblast cell differenti-

    ation towards both syncytial forma-

    tion and extravillous trophoblastcells remodeling of the spiral arter-

    ies. It is now established that apo-

    ptotic cell death in the developing

    placenta is associated with tropho-

    blast cell turnover, enabling re-newal of syncytium (Huppertz et

    al., 1999). It has been postulatedthat apoptosis is initiated in the vil-

    lous trophoblasts facilitating fusion,

    but is delayed in the ST layer, beingonly finalized prior to the extrusion

    of apoptotic nuclei in the form of

    syncytial knots (Huppertz et al.,1998). The balance between tro-

    phoblast differentiation/fusion and

    extrusion of aging material istightly regulated, i.e., fusion of un-

    differentiated cells occurs within

    two to four days, while syncytial fu-sion of nuclei and their extrusion

    requires three to four weeks (Hup-pertz and Kingdom, 2004). This

    constant process of cell fusion andturnover represents a unique bio-

    logical example of fine tuning of the

    apoptotic cascade. Recent in vitrostudies suggest that caspase 8

    plays a major role in driving the firstevents of trophoblast turnover, as

    its inhibition using antisense oligo-

    nucleotides and/or caspase-spe-

    cific inhibitors impairs cell fusion(Black et al., 2004).Emerging evidence suggests that

    the events associated with tropho-

    blast invasion into the spiral arter-ies are in part also mediated by ap-

    optosis. Using an in vitro coculturesystem of trophoblast cells and spi-

    ral arteries, it has been recently

    shown that the expression ofcleaved PARP, an apoptotic marker,

    localizes only to those arteries that

    exhibit trophoblast remodeling fol-lowing loss of endothelial cells

    268 JURISICOVA ET AL.

    Birth Defects Research (Part C) 75:262280, (2005)

  • 8/13/2019 029-030 genetika implantacije (14)

    8/19

    (Cartwright et al., 2002). The na-

    ture of the molecular cascade ofdeath activation in this model is

    less clear; however, a recent report

    suggests that the endothelial celldeath induced by trophoblasts dur-

    ing the process of remodeling ap-

    pears to be mediated via Fas/Fasligand interaction (Ashton et al.,2005).

    Oxygen as a Regulator of

    Trophoblast Fate

    Early embryonic development takesplace in a relatively hypoxic environ-

    ment. Oxygen electrode studieshave shown that at five to eight

    weeks gestation, oxygen tension is

    low (

    20 mm Hg, equivalent to 3%O2), while at 1012 weeks, when theintervillous space opens to maternal

    blood, oxygen levels increase to55

    mm Hg (810% O2) (Rodesch et al.,1992; Burton et al., 1999). The steep

    rise of oxygen tension experiencedby trophoblast cells has been found

    to be associated with increased ex-pression of genes associated with

    oxidative stress (Jauniaux et al.,2000). Increasing evidence suggests

    that oxygen is a key regulator for thecellular events occurring during early

    placentation. Low pO2levels supportproper trophoblast proliferation,

    while the increase of pO2 after theintervillous space opens to maternal

    blood is essential to finalize the pro-cess of trophoblast differentiation/

    invasion (Jaffe et al., 1997).The precise mechanisms by which

    oxygen modulates these develop-mental events are not fully under-

    stood; however, hypoxia-induciblefactor-1 (HIF-1), a master regulator

    of oxygen homeostasis (Semenza,

    2001a, 2001b; Kaelin, 2002), seemsto play a key role in this cellular re-sponse (Caniggia et al., 2000).

    In vitro studies have indicatedthat, hypoxia, one of the most phys-

    iologically relevant stimuli is able toinitiate apoptosis of term tropho-

    blasts (Nelson et al., 1999; Levy et

    al., 2000). Increased cell death interm primary isolated cells exposed

    to severe hypoxic conditions (15mm Hg) is accompanied by in-

    creased expression of the proapop-totic Bax and p53 and decreased ex-

    pression of Bcl-2 (Levy et al., 2000).

    Interestingly, addition of epidermalgrowth factor inhibits the hypoxia-

    induced trophoblast cell death (Levyet al., 2000). In the human placenta,

    the low oxygen-stimulatory effect oncell death appears to entail intrinsic

    (mitochondrial) rather than extrinsic(death receptor) pathways. This isconsistent with observations that re-

    active oxygen species do not medi-ate trophoblast cell apoptosis in-

    duced by TNF- (Smith et al., 1999).Recent evidence indicates that

    the effect of oxygen on trophoblastcell death is exquisitely dependent

    upon the pO2 levels. While tropho-blast cells exposed to both severe

    hypoxia (10 mm Hg) and to highoxygen levels (140 mm Hg) un-

    dergo apoptosis readily, cells ex-posed to oxygen levels ranging be-

    tween 15 and 40 mm Hg show a lowfrequency of apoptosis (Mackova et

    al., 2003). This is not surprising, ifone considers that during normal

    placental development the physio-logical oxygen levels that permit

    proper trophoblast differentiationrange between 15 and 55 mm Hg.

    In vitro studies using villous ex-plant cultures have demonstrated

    that severe hypoxia favors ne-

    crotic, as opposed to apoptotic,

    shedding of syncytiotrophoblasts(Huppertz et al., 2003). Hung et al.(2002) have proposed that inter-

    mittent placental perfusion leads toincreased oxidative stress, and

    have reported that hypoxia-reoxy-genation is a powerful inducer of

    classical events associated with ap-optosis, including mitochondrial cy-

    tochrome C release, caspase 3 ac-tivation, and PARP cleavage. Thus,

    rather than hypoxia, it is the localhypoxia-reoxygenation injury that

    may be more relevant to activationof cell death pathways in develop-

    ing placenta, particularly in theconditions associated with placen-

    tal insufficiencies.

    Cell Death in Physiological

    and Pathological

    Conditions of Placental

    Hypoxia

    Preeclampsia is an unpredictable

    and devastating disorder that man-ifests abruptly, and remains a ma-

    jor cause of maternal and perinatal

    morbidity and mortality complicat-

    ing 57% of pregnancies. This dis-

    ease, unique to humans, is charac-terized by a general maternal

    inflammatory response likely due to

    the release of placental factors,

    leading to the damage of variousorgans, including kidney, lungs,

    and heart, often resulting in life-long complications. A key feature

    that characterizes this disease is

    the excessive shedding and depor-tation of placental debris into the

    maternal circulation due to exces-

    sive placental trophoblast cell

    death (Knight et al., 1998; Redmanand Sargent, 2000). This is be-

    lieved to be the culprit of the ma-

    ternal endothelial injury leading to

    the onset of the clinical manifesta-tions including hypertension, pro-

    teinuria, and edema (Roberts et al.,1989; Broughton Pipkin and Rubin,

    1994). Although preeclampsia is

    the most intensively studied clinicalcondition in pregnancy, its cause

    remains elusive and numerous hy-

    potheses on its origin abound. It is,however, accepted that the pla-

    centa plays a key role in its patho-

    genesis, as its removal at deliveryresults in prompt resolution of this

    disease. A typical hallmark of pre-

    eclampsia is the lack of remodelingof the maternal spiral arteries by

    trophoblast cells, and this is be-lieved to be the cause for insuffi-

    cient uteroplacental circulationleading to placental hypoxia (Hung

    et al., 2002). Both relative and di-

    rect measures of utero-placentalblood flow (Lunell et al., 1979), as

    well as Doppler studies (Kingdomand Kaufmann, 1997), support the

    notion that this serious disease is

    associated with insufficient blood

    flow and placental hypoxia. The in-sufficient perfusion of the placenta

    often causes fetal growth restric-tion. Recent studies suggest that

    the rate of apoptosis is increased inpreeclampsia due to placental oxi-

    dative stress (Hung et al., 2002;

    Myatt and Cui, 2004). Thus, it isplausible that hypoxic stress may

    be responsible for the increased ap-optotic rates of syncytiotropho-

    blasts observed in preeclamptic

    placentae (Allaire et al., 2000). Thedeportation of placental debris into

    MOLECULAR MECHANISMS OF TROPHOBLAST SURVIVAL 269

    Birth Defects Research (Part C) 75:262280, (2005)

  • 8/13/2019 029-030 genetika implantacije (14)

    9/19

    the maternal circulation is believed

    to be the culprit of the maternal en-

    dothelial injury. These fragments

    are normally present in the plasmaof normal pregnant women; how-

    ever, the release of this placental

    debris is increased in preeclampsia,

    a likely consequence of excessiveplacental cell death.

    While it is now widely acceptedthat preeclampsia is associated

    with increased trophoblast cell

    death, the molecular pathways be-hind this deregulated process are

    unclear. Elevated cell death in pre-

    eclampsia has been detected both

    in villous trophoblast and in extra-villous trophoblast cells within an-

    choring villi at the fetal-maternal

    interface. Widespread cell death

    measured by TUNEL is detected inextravillous trophoblast cells within

    the uterine wall, and is accompa-nied by decreased expression of the

    prosurvival factor Bcl-2 (Di Be-

    rardino, 2004). More recently, ithas been proposed that excess of

    macrophages found in the placental

    bed of preeclamptic patients maycurtail extravillous trophoblast in-

    vasion by an apoptotic mechanism

    that involves TNF and tryptophandepletion (Reister et al., 2001).

    While no differences in the expres-

    sion of Bcl-2 family proteins, suchas Bax, Bcl-2, Bak, and Bcl-XL, are

    found between preeclampsia/IUGRand control placentae, elevated ex-

    pression of p53 and Fas, but notFasL, has been detected in villous

    trophoblast of preeclamptic placen-

    tae (Hsu et al., 2001; Levy et al.,2002). Only Ishihara et al. (2002)

    have reported decreased expres-

    sion of the pro-survival factor Bcl-2in syncytiotrophoblast of severe

    preeclamptic and IUGR placentae.

    Our recently published observa-tions have highlighted the impor-

    tance of Mtd/Bok, a proapoptoticBcl-2 family member, during both

    normal and abnormal placentation.Mtd/Bok (Mtd: Matador/Bok: Bcl-2

    ovarian killer) belongs to the mul-

    tidomain pore-forming subfamily ofproapoptotic Bcl-2 family mem-

    bers, including Bax and Bak (Hsu etal., 1997; Inohara et al., 1998).

    Mtd transcript is alternatively

    spliced and encodes for two proteinisoforms: Mtd-L that primarily in-

    teracts with Mcl-1, and Mtd-S re-

    sulting from fusion of BH3 and BH1

    domains, the function of which can-

    not be antagonized by any knownantiapoptotic Bcl-2 family member

    (Hsu and Hsueh, 1998). We identi-

    fied a third Mtd splice variant,

    Mtd-P, with a unique developmen-tal expression profile in normal hu-

    man placenta. The unique expres-sion and distribution of Mtd-P is a

    result of its response to oxygen, a

    master regulator of trophoblast dif-ferentiation (Soleymanlou et al.,

    2005b). Mtd-P, a C-terminal trun-

    cated protein resulting from exon II

    skipping, is a proapoptotic mole-cule involved in trophoblast cell

    death, regulating mitochondrial

    membrane potential. We deter-

    mined that the elevated expressionof this novel splice variant is unique

    to pregnancies complicated by se-vere early onset preeclampsia

    (Soleymanlou et al., 2005b). In-

    creased Mtd-P expression in pre-eclampsia may be causative of in-

    creased trophoblast cell death

    leading to excessive shedding ofplacenta debris observed in this

    disease. Interestingly, placentae

    obtained from normotensive con-trol subjects, as well as late-onset

    (term) preeclampsia, IUGR, or es-

    sential hypertensive subjects, didnot show any alteration in Mtd-P

    expression. The high Mtd-P expres-sion found in early severe, but not

    late, onset preeclampsia raises thepossibility that the classical defini-

    tion of preeclampsia might be too

    broad and might encompass differ-ent diseases. In support of this

    idea, Redman and Sargent (2000)have recently classified preeclamp-

    sia into two very distinct catego-

    ries: the placental and the maternal

    preeclampsia. They define the firsttype as being linked to placental al-teration following hypoxia insult,

    whereas the second type is associ-

    ated with a defect in the maternalvasculature and not in the placenta.

    It is also likely that different molec-

    ular pathways will be driving patho-physiological defects in preeclamp-

    tic placental tissue in these twodifferent subtypes.

    Placentae from high altitude

    pregnancies represent a uniquenatural model of chronic placental

    hypoxia (Fig. 2). Because of the

    reduction in maternal arterial ox-

    ygen pressure, these placentae

    are exposed to reduced uteropla-cental oxygenation (Zamudio,

    2003). In pregnancies at high al-

    titude (2700 m), intrauterine

    growth is restricted and the inci-dence of preeclampsia is elevated

    two- to four-fold (Palmer et al.,1999; Zamudio, 2003), support-

    ing the widespread belief that pla-

    cental hypoxia is associated withpreeclampsia. As such, the high

    altitude model represents a natu-

    ral laboratory to study preeclamp-sia. Using high-throughput func-

    tional genomics, we have recently

    reported a striking similarity in

    global gene expression between

    low oxygen-treated explants, highaltitude placentae, and placentae

    from preeclamptic pregnancies,indicating that changes in global

    gene expression in preeclamptic

    placentae are due to reduced ox-ygenation (Soleymanlou et al.,

    2005a).

    Pregnancies at high altitude alsoshow incomplete remodeling of the

    spiral arteries, similar to that re-

    ported in preeclampsia (Tissot vanPatot et al., 2003). Histomorpho-

    logical studies have shown that

    high altitude placentae are sub-jected to unique changes including

    increased villous vascularization,increased capillary density, and

    thinning of villous membrane,events believed to ensure adequate

    oxygen delivery to the fetus. More-

    over, high altitude placentae alsoexhibit reduced perisyncytial fibrin-

    type fibrinoid deposition when com-pared to placentae from lower alti-

    tude pregnancies (Mayhew et al.,

    2002; Zamudio, 2003). The signif-

    icance of reduced fibrin depositionis unknown. It is yet unclearwhether this change is related to an

    altered trophoblast apoptotic rheo-

    stat affecting trophoblast turnover.Although studies of cell death in in

    vitro and in vivo pathological mod-els of placental hypoxia are avail-

    able, little is known about the apo-

    ptotic adaptation/changes thatoccur in high-altitude placentae.

    We have recently found that alti-tude-induced chronic hypoxia leads

    to increased expression of the pro-

    270 JURISICOVA ET AL.

    Birth Defects Research (Part C) 75:262280, (2005)

  • 8/13/2019 029-030 genetika implantacije (14)

    10/19

    tector Mcl-1L and decreased ex-

    pression of the proapoptotic Mtd-Pand Mcl1-S. In addition, high alti-

    tude placentae appear to have adecreased rate of trophoblast turn-

    over, as identified by decreasedsyncytin expression (Caniggia et

    al., 2004). Hence, in pregnanciesfrom high altitude-induced chronic

    hypoxia, a molecular shift towardsinhibition of trophoblast cell death

    occurs, and this may protect/slow

    down trophoblast cell turnover anddeath.

    THE MOUSE PLACENTA AS

    A MODEL

    Recent comparative studies be-

    tween mouse and human placentaehave revealed striking similarities

    in cellular mechanisms and tissue

    framework, providing justificationfor using the mouse as a suitable

    animal model for studies of humanplacentation. At day 6 of mouse de-

    velopment (two days after implan-tation), two diploid populations of

    trophoblast derivatives are estab-lished: the proximal extraembry-

    onic ectoderm and the distal ecto-

    placental cone. Between days 7.5

    and 9.5, the placenta undergoes

    major morphological remodeling,resulting in the formation of three

    distinct cellular layers, each withunique morphology and function

    (Rossant, 2001). The fusion of the

    allantois with the chorion and sub-

    sequent branching establishes anetwork of small canals collectively

    referred to as the labyrinth. This

    layer is responsible for gas and nu-trient exchange, and thus bears a

    similarity to floating chorionic villi in

    humans (Cross, 2000). The spon-

    giotrophoblast layer is distal to thelabyrinth and consists of variable-sized, hormone-producing cells

    with the capability of differentiating

    into glycogen cells. Due to the ex-pression of several gene markers,

    as well as to their spatial distribu-

    tion, the cells of this layer are con-sidered to be analogous to human

    column CT cells (Hemberger andCross, 2001). The two most distal

    trophoblast cells in the mouse pla-

    centa are glycogen cells, a cell typeof distinct morphology but un-

    known function, and giant cells.

    Both are invasive cell types and are

    in direct contact with the maternal

    decidua; as such, these cells areanalogous to human EVT cells

    (Cross, 2000). Giant cells have dis-tinctively large, polyploid nuclei,

    which very efficiently produce sev-

    eral key regulatory, luteotropic,and lactogenic hormones, as well

    as angiogenic factors (Cross,

    2000). To further support the use of

    the mouse as a model organism forhuman placentation, recent reports

    have established that a subset of

    specialized murine giant cells is in-

    vasive and capable of remodelingmaternal spiral arteries (Cross etal., 2002). This is identical to the

    events that occur during human

    placentation, where spiral arteriesof the placental bed are invaded by

    EVT cells. During invasion of thehuman spiral arteries, the endothe-

    lial and smooth muscle layers are

    dislodged and replaced by EVTcells. The presence of homologous

    cell types and cellular behaviorshighlight the utility of the mouse

    placenta as a suitable model for

    Figure 2. Putative model of trophoblast cell death in physiological and pathological models of human placental hypoxia. In pregnanciesfrom high altitude, chronic hypoxia regulates the placental Mcl-1/Mtd rheostat via alteration of splicing, favoring trophoblast cellsurvival in contrast to preeclampsia, where increased trophoblast demise and turnover is observed.

    MOLECULAR MECHANISMS OF TROPHOBLAST SURVIVAL 271

    Birth Defects Research (Part C) 75:262280, (2005)

  • 8/13/2019 029-030 genetika implantacije (14)

    11/19

    study of normal, human placenta-

    tion. Such a model is highly attrac-tive, as it allows for genetic manip-

    ulation and induction of variouspathological conditions.

    While it is now clear that apopto-sis plays a functional role in placen-

    tal tissue morphogenesis, the un-derlying mechanisms coordinating

    cell death have not been studied inthis context. The localization and

    extent of cell death in the rodent

    placenta and how this correlates tocell death patterns in human pla-centa remain largely unknown;

    however, some sporadic in vivo(Zybina et al., 2000; Mu et al.,

    2002) and in vitro (Goncalves et al.,2003) observations have surfaced

    as supplementary data while inves-

    tigating other events during pla-centation. In order to overcome

    this dearth of information regardingcell death during placental develop-

    ment, we are currently finalizingsystematic analyses of the tempo-

    ral and spatial distribution of dying

    cells throughout gestation in mouseplacentae obtained after natural

    matings, and these results aresummarized in Figure 3. Prelimi-

    nary analyses of these tissues indi-cate the presence of sporadic cell

    death in both the labyrinth andspongiotrophoblast layers, which

    likely reflects normal cell turnover(Detmar et al., 2004), similar to

    that reported for ST turnover in hu-

    mans (Huppertz et al., 1998). Inaddition, more extensive, orga-nized death of giant cells is ob-

    served over gestation, resulting inalmost entire elimination of this cell

    type at term. Furthermore, startingat approximately embryonic day

    15.5, fetal glycogen cells and ma-ternal decidual cells undergo orga-

    nized cell death at the maternal-fetal interface (Detmar et al.,

    2004), creating focal perforations

    that may facilitate delivery of theconceptus. Additionally, we have

    observed that the extent of cell

    death in murine placentae is influ-enced by genetic background, sug-

    gesting either the presence of ge-netic modifiers that regulate cell

    death susceptibility, or that regu-late other physiological factors,

    such as nutrition and hypoxia, influ-encing the delicate balance be-

    tween anti- and proapoptotic path-ways (Detmar et al., 2004). Similar

    variability in the severity and pen-

    etrance of embryonic phenotypescaused by the disruption of celldeath-associated genes have been

    described for caspase-3 (Kuida etal., 1996; Woo et al., 1998) Rho

    (Humphries et al., 2001), and Daxx(Michaelson et al., 1999).

    Genetic Models of Cell

    Death in Mouse Placenta

    Tumor suppressor genes encode

    for proteins that are involved in cellcycle regulation and cellular differ-

    Figure 3.Schematic and histological representations of cell death patterns in murine placenta. Diagrammatic transverse sections ofday 15.5 (A) and day 18.5 (B) murine placentae at midline, after in situ TUNEL staining, showing sporadic cell death patterns in mostplacental layers except the maternal decidua, where regionalized pockets of dying maternal decidual and fetal glycogen cells can befound, and in the giant cell layer, which exhibits organized cell death over gestation, resulting in diminished numbers of this cell typeat day 18.5. Accompanying histomicrographs represent 5-m sections of mouse placentae after TUNEL staining, showing isolatedtrophoblast giant cell (C) and labyrinthine trophoblast cell death (D) in day 15.5 placenta, and whole regions of maternal decidual andfetal glycogen cell death in day 18.5 placenta (E). TUNEL-positive cells are stained brown and sections are counterstained withhematoxylin.

    272 JURISICOVA ET AL.

    Birth Defects Research (Part C) 75:262280, (2005)

  • 8/13/2019 029-030 genetika implantacije (14)

    12/19

    entiation; such proteins are often

    referred to as gatekeepers,inhib-

    iting or permitting cell cycle pro-

    gression, depending on the internaland external status of the cell. A

    number of these genessuch as

    p53have been implicated in pro-

    moting apoptosis in various celltypes, as this outcome, or cell se-

    nescence, is usually consideredpreferable to the propagation of a

    cell with uncontrolled proliferative

    potential. The mouse placenta ex-presses a number of these tumor

    suppressors, including p53, retino-

    blastoma (Rb), and phosphatase

    and tensin homolog (PTEN).The p53 tumor suppressor is a

    ubiquitously-expressed transcrip-

    tion factor that is activated by DNA

    damage (DSa-Eipper et al., 2001;Katayama et al., 2002) and other

    cellular stressors, such as hypoxia(Long et al., 1997; Lee et al.,

    2005). The primary function of this

    protein is to induce cells to undergoG1 arrest (Xiong et al., 1993; el-

    Deiry et al., 1994) or apoptosis by

    activating the appropriate targetgenes. In addition to its transcrip-

    tional activity, a novel p53-medi-

    ated apoptotic pathway was re-cently described, suggesting that

    p53 translocates to the mitochon-

    dria, where it induces permeabiliza-tion of mitochondria via interaction

    with Bcl-2 and BH3-only familymembers (Chipuk et al., 2004,

    2005). The transcriptional and ap-optotic potential of p53 in placental

    cells have been demonstrated in

    both rodent and human models.p53 has been shown to upregulate

    Bax and downregulate Bcl-2 in hu-

    man trophoblast cultured under hy-poxic conditions (Levy et al., 2000).

    In addition, p53 has exhibited in-

    volvement in triggering the apopto-tic pathway in early, proliferating

    trophoblast cells with compromisedgenomic stability (Lim and Hasty,

    1996; Weiss et al., 2000). In vivostudies of the effects of DNA-dam-

    age-inducing agents on rat placen-

    tation revealed that cells within thelabyrinth appear most susceptible

    to p53-mediated apoptosis, as evi-denced by increased levels of active

    caspase-3 (Katayama et al., 2002;

    Yamauchi et al., 2004). Lastly, a re-cent report demonstrates that the

    loss of p53 in differentiating murineTS cells renders them not only re-

    sistant to DNA-damage-induced

    apoptosis, but also allows them tobypass the critical G1 checkpoint

    (Soloveva and Linzer, 2004). This is

    supported by the report that ex-

    traembryonic cells, but not embry-onic cells, are resistant to p53-de-

    pendent cell death after DNAdamage (Heyer et al., 2000). p53-

    deficient mice are viable, but cells

    derived from these animals demon-strate higher proliferation rates in

    culture (Tsukada et al., 1993);

    however, the growth dynamics of

    trophoblast-derived cells were notassessed in this study. In light of

    the above reports, it may be worth-

    while to revisit the p53 knockout

    placentae for overall trophoblastresistance to hypoxia- and DNA-

    damage-induced apoptosis, and toassess p53-mediated apoptosis

    patterns in differentiating tropho-

    blast cell subpopulations.The Rb protein is also a tumor

    suppressor, with a prominent role

    in cell cycle regulation. The phos-phorylation state of Rb correlates

    with its functional capacity: phos-

    phorylation is maximal at the startof S phase (i.e., in proliferating

    cells) and lowest after mitosis and

    entry in G1(i.e., in quiescent cells);hence, the hypophosphorylated

    form of Rb suppresses cell prolifer-ation. It was recently demonstrated

    that hypophosphorylated Rb pre-dominates in murine TS cells de-

    prived of Fgf-4 (a required growth

    factor for TS cells) and hyperphos-phorylated Rb is found in actively

    growing TS cells (Soloveva and Lin-

    zer, 2004); however, all forms of Rbprotein were observed to decrease in

    differentiating giant cells in vitro (So-

    loveva and Linzer, 2004). Initialstudies producing Rb knockout mice

    revealed that these mice die be-tween embryonic days 13.5 and

    15.5, displaying abnormalities inerythropoiesis and central nervous

    system due to excessive apoptosis,

    failed differentiation, and disruptionof the cell cycle (Clarke et al., 1992;

    Jacks et al., 1992; Lee et al., 1992).Additionally, it was later reported

    that Rb mutant placentae exhibit in-

    creased numbers of trophoblast cellsin the labyrinth, leading to disrupted

    architecture of this placental layer

    and resulting in decreased placental

    vascularization and transport (Wu et

    al., 2003). These data indicate thatRb mutant placentae appear to have

    an unprogrammed proliferation de-

    fect within the labyrinthine layer, off-

    setting the balance between the celldeath and proliferation pathways. In

    order to determine whether the em-bryonic phenotype was secondary to

    a placental defect, tetraploid aggre-

    gation and conditional knockout ap-proaches were employed to provide

    Rb-deficient embryos with wild-type

    placentae. Under these conditions,

    the knockout embryos were carriedto term, but died shortly after birth

    (Wu et al., 2003), suggesting that

    the majority of embryonic pheno-

    types in Rb-deficient embryos arecaused by a dysfunctional placenta

    and are therefore, not due to a cell-autonomous Rb requirement by the

    embryo. Rb family members bridge

    the cell death and cycle pathways bydirectly repressing E2F/Dp transcrip-

    tion factors (Trimarchi and Lees,

    2002). Recently, it was reported thatDp1 knockout embryos die by em-

    bryonic day 12.5 and demonstrate

    increased rates of apoptosis in earlyextraembryonic cells (Kohn et al.,

    2003). Dp1 deficiency might effect

    this increased cell death by prevent-ing Rb-mediated repression of

    p19ARF (a p53 stabilizer) or p73 (ap53 homolog with apoptotic func-

    tions).PTEN is a tumor suppressor that

    has been shown to inhibit cell mi-

    gration, cell spreading, and focaladhesion formation through inter-

    actions with focal adhesion kinase(Tamura et al., 1998; Gu et al.,

    1999). Additionally, PTEN has been

    shown to negatively regulate phos-

    phoinositide-dependent kinase 1(PDK1), which phosphorylates andactivates protein kinase B (PKB/

    Akt1), triggering a well-established

    survival pathway (Staal and Hart-ley, 1988; Dudek et al., 1997).

    PTEN has been shown to be ubiqui-

    tously expressed in embryonic day7.5 mouse embryos, and its inacti-

    vation in the mouse causes embry-onic lethality at day 9.5 (Stambolic

    et al., 1998; Suzuki et al., 1998).

    Knockout embryos demonstrate re-gions of increased proliferation, in-

    MOLECULAR MECHANISMS OF TROPHOBLAST SURVIVAL 273

    Birth Defects Research (Part C) 75:262280, (2005)

  • 8/13/2019 029-030 genetika implantacije (14)

    13/19

    cluding the allantois, the expansion

    of which is hypothesized to inhibit

    chorioallantoic fusion and there-

    fore, causes embryonic death (Su-zuki et al., 1998). Interestingly,

    murine trophoblast and endothelial

    cells have been demonstrated to

    express PKB/Akt1, and the conse-quences of PKB/Akt1-deficiency are

    placental hypotrophy, with reduceddecidual basalis, glycogen-contain-

    ing spongiotrophoblast, and vascu-

    larization (Yang et al., 2003). Thesedefects indicate a deficiency in pla-

    centation, leading to the observed

    fetal growth reduction, neonatal le-

    thality, and diminished life span af-ter exposure to genotoxic stress

    (Yang et al., 2003). Lastly, it has

    also been shown that PKB/Akt1 sig-

    naling is involved in differentiationof murine TS cells to giant cells in

    vitro (Kamei et al., 2002). Since gi-ant cells in the mouse placenta are

    involved in the production of nu-

    merous protein and steroid hor-

    mones, it is possible that reducedplacental and fetal growth may be

    due to the impaired differentiation

    of this cell type.Murine Bruce/Apollon (baculovi-

    rus inhibitor of apoptosis repeat-containing (BIR) ubiquitin-conju-

    gating enzyme) is a large

    (528-kDa) protein that has both anN-terminal BIR domain and a C-ter-

    minal ubiquitin-conjugating do-main (UBC); these regions provide

    Bruce with both antiapoptotic

    (Hauser et al., 1998; Chen et al.,1999; Bartke et al., 2004) and ubiq-

    uitinoylation capabilities (Bartke etal., 2004; Hao et al., 2004b). There

    are several inhibitors of apoptosis

    (IAP) proteins, including Bruce, andthese proteins function as cell death

    antagonists by suppressing pro-

    apoptotic proteins such as Smac/Diablo and active caspase-9 (Bartke

    et al., 2004; Hao et al., 2004b). TheBruce gene is highly conserved, as

    human APOLLON shares 92% iden-tity with Bruce, and has been dem-

    onstrated to confer chemotherapeu-

    tic resistance to certain cancer cells(Chen et al., 1999). Lotz et al.

    (2004) reported that Bruce is ex-pressed predominantly in the diploid

    trophoblast of the placenta, including

    the labyrinthine layer and the spon-giotrophoblast; reduced Bruce ex-

    pression was observed in trophoblastgiant cells. Analysis of earlier devel-

    opmental stages revealed that Bruce

    can also be detected in the chorion,within the cells of the ectoplacental

    cone and in early trophoblast giant

    cells, as well as in the late gastrula

    stage embryo (Hitz et al., 2005; Renet al., 2005). Three separate groups

    of investigators have producedBruce knockout mice, with two

    groups observing a trophoblast pro-

    liferation-related phenotype and no

    alterations in cell death (Hitz et al.,2005; Lotz et al., 2004). The third

    group also observed proliferation de-

    fects but additionally reported in-creased rates of apoptosis in Bruce-

    deficient placentae (Ren et al.,

    2005). Loss of Bruce leads to embry-

    onic and/or perinatal growth reduc-tion and lethality, which can likely be

    attributed to the observed placentaldefects. Proliferation was severely

    reduced in the spongiotrophoblast

    layer (Hitz et al., 2005; Lotz et al.,2004), and was diminished in the

    labyrinth (Lotz et al., 2004). Addi-

    tionally, up regulation of Bax, Bak,and caspase-2 in mutant trophoblast

    cells and activation of the mitochon-

    drial cell death cascade in embryonicfibroblasts obtained from mutant

    embryos was also detected (Ren et

    al., 2005). Moreover, p53 expressionwas also demonstrated to be ele-

    vated in Bruce-deficient placentae,particularly in spongiotrophoblast

    cells, and silencing of p53 and Bruce

    expression in cell lines resulted in im-proved cell viability (Ren et al.,

    2005). Thus, p53 appears to acts asa downstream effector of Bruce, and

    in the absence of Bruce, mitochon-

    drially-mediated apoptosis ensues.The conflicting results of these stud-

    ies is perhaps, not so unexpected in

    hindsight. Bruce is a chimeric mole-cule, with both ubiquitinoylation and

    apoptosis-inhibiting capabilities, un-derscoring the multifunctional na-

    ture of the Bruce protein. Yeast ho-mologs of Bruce have likewise been

    shown to be involved in cell division

    (Uren et al., 1999; Silke and Vaux,2001). On the other hand, as previ-

    ously stated, Bruce is also clearly as-sociated with the apoptotic pathway,

    and further molecular analysis into

    the nature of this enigmaticyet ex-citingmolecule in the right cellular

    context is required, in order to fur-ther elucidate the function of Bruce

    during mammalian placentation.

    Daxx (Fas death domain-associ-ated protein) was initially re-

    ported as a highly conserved pro-

    tein that associated with the

    intracellular domain of Fas andenhanced Fas-mediated apoptosis

    in overexpression studies (Yang etal., 1997). As is the case with

    Bruce, Daxx is a multifunctional

    protein with seemingly contradic-

    tory functions. It has been shownto be involved in both extrinsic

    (TGF--mediated) and intrinsic

    (p53-dependent DNA damage)apoptosis pathways (Gostissa et

    al., 2004; Chang et al., 2005). In

    addition to these cell death func-

    tions, Daxx is also capable of tran-scriptionally repressing CRE,

    E2F1, Sp1, NF-B, and the andro-gen receptor (Ecsedy et al., 2003;

    Chang et al., 2005), demonstrat-

    ing further potential in modulatingcellular behavior. Daxx is ex-

    pressed in a number of murine (Su

    et al., 2002) and human tissues,including placenta (Annotation:

    GDS1096, GDS181, Geo profiles

    at www.ncbi.nlm.nih.gov). Daxxdeficiency in mice leads to embry-

    onic lethality by day 9.5 and both

    embryonic and extraembryoniclineages are diminished in com-

    parison to wild-type littermates,marked by increased rates of ap-

    optosis by day 7.5 and day 8.5(Michaelson et al., 1999). This en-

    hanced apoptosis was unex-

    pected, since Daxx had, untilthen, only been associated with

    promoting apoptotic events in the

    cell; however, further investiga-tion revealed that Daxx mutant ES

    cells also had elevated rates of ap-

    optosis. It was later revealed thata similar Daxx deletion triggered

    cell death by stimulating the JNK/p38-Bim-Bax pathway, leading to

    the activation of caspase-9 andcaspase-3 (Song and Lee, 2004).

    Lastly, Daxx has recently been re-

    ported to play a role in viral pro-tection, as Daxx-null fibroblast

    cell lines demonstrated enhancedviral gene expression compared to

    Daxx-complemented cells (Greger

    et al., 2005). Therefore, it is in-triguing to speculate that in addi-

    274 JURISICOVA ET AL.

    Birth Defects Research (Part C) 75:262280, (2005)

  • 8/13/2019 029-030 genetika implantacije (14)

    14/19

    tion to the role of Daxx as a regu-

    lator of cell death in the placenta,

    it may also be involved in protect-

    ing the trophoblast and hence, the

    fetus, from viral invasion.

    Prostaglandin F2 receptor (FP)

    is a G-protein-coupled receptor

    that has been shown to induce theapoptosis cascade by activating

    caspase-8 in luteal cells (Li et al.,

    2001). Interestingly, FP lacks an

    intracytoplasmic region possess-

    ing any of the traditional death or

    caspase-recruiting domains that

    characterize other, typical death

    associated receptors. Prostaglan-

    din F2 receptor is highly ex-

    pressed in the uterus (Sugimoto

    et al., 1994), and has also been

    shown to be expressed in the

    mouse (Su et al., 2004) and hu-man (Su et al., 2002) placenta.

    Homozygous deletion of FP re-

    sulted in developmentally normal

    and viable mice, but FP-deficient

    females fail to deliver their fetuses

    at term, which eventually died in

    utero and were resorbed (Sugi-

    moto et al., 1997). No changes

    were detected in mutant placental

    and decidual weights, nor were

    there any disparities in placental

    cell death patterns; however, ele-

    vated decidual cell death overgestation was noted (Mu et al.,

    2002). Further characterization of

    phenotype revealed alterations of

    decidual cell death patterns once

    postterm fetuses were catego-

    rized as either live or dead. Ele-

    vated decidual cell death in dead

    fetuses was associated with alter-

    ations in the Bax/Bcl-2 ratio and

    increased active caspase-3 levels

    (Mu et al., 2003). Given these ob-

    servations, it was hypothesized

    that decidual cell death was nec-

    essary for normal term delivery of

    the conceptus, and that a Bax:

    Bcl-2 rheostatis involved in reg-

    ulating apoptosis in the postterm

    placenta (Mu et al., 2003). On the

    contrary, upregulation of Bcl-2

    was reported in the decidua of

    abortion-prone mice, perhaps

    serving as a compensatory or pro-

    tective mechanism (Bertoja et al.,

    2005), especially considering the

    premature deliveryof these pre-

    term placentae.

    As a well-spent day

    brings happy sleep, so life

    well used brings happy

    death (Leonardo Da Vinci)

    The functional meaning of cellulardeath in the context of tissue sur-

    vival is beautifully reflected in thiswise observation. This is particu-

    larly true for the life of the tropho-blast cells as the proper oxygen-

    ated environment brings ameaningful death, while improper

    oxygenation, characteristic featureof placental pathologies, leads to an

    accelerated death which often re-sults in a harmful death. In sum-

    mary, cell death plays a key role inthe appropriate maturation of the

    placenta, and pro- and antiapop-

    totic expression patterns can bemanipulated in the fetal-placental-maternal unit in order to optimize

    the uterine environment for healthygestation. However, deregulation

    of these pathways has clear nega-tive long-term developmental con-

    sequences for both the mother and

    the fetus, resulting in serioushealth threats that may ultimately

    lead to death.

    REFERENCES

    Allaire AD, Ballenger KA, Wells SR, et al.2000. Placental apoptosis in preeclamp-sia. Obstet Gynecol 96:271276.

    Aplin JD. 1991.Implantation, trophoblastdifferentiation and haemochorial pla-centation: mechanistic evidence in vivoand in vitro. J Cell Sci 99(Pt 4):681692.

    Ashton SV, Whitley GS, Dash PR, et al.2005. Uterine spiral artery remodel-ing involves endothelial apoptosis in-duced by extravillous trophoblaststhrough Fas/FasL interactions. Arterio-scler Thromb Vasc Biol 25:102108.

    Assuncao Guimaraes C, Linden R. 2004.Programmed cell deaths. Apoptosisand

    alternative deathstyles. Eur J Biochem271:16381650.Baehrecke EH. 2003. Autophagic pro-

    grammed cell death in Drosophila. CellDeath Differ 10:940945.

    Barker DJ. 1992. Thefetal origins of adulthypertension. J Hypertens Suppl 10:S39S44.

    Bartke T, Pohl C, Pyrowolakis G, JentschS. 2004. Dual role of BRUCE as an an-tiapoptotic IAP and a chimeric E2/E3ubiquitin ligase. Mol Cell 14:801811.

    Baschat AA. 2004. Fetal responsesto pla-cental insufficiency: an update. BJOG111:10311041.

    Bernstein IM, Horbar JD, Badger GJ, etal. 2000. Morbidity andmortality among

    very-low-birth-weight neonates with in-trauterine growth restriction. The Ver-mont Oxford Network. Am J ObstetGynecol 182(Pt 1):198206.

    Bertoja AZ, Zenclussen ML, WollenbergI, et al. 2005. Upregulation of Bcl-2 atthe foetal-maternal interface from miceundergoing abortion. Scand J Immu-

    nol 61:492

    502.Black S, Kadyrov M, Kaufmann P, et al.2004. Syncytial fusion of human tro-phoblast depends on caspase 8. CellDeath Differ 11:9098.

    Blair E, Stanley FJ. 1988. Intrapartumasphyxia: a rarecause of cerebral palsy.J Pediatr 112:515519.

    Bowman P, McLaren A. 1970. Cleavagerate of mouse embryos in vivo and invitro. J Embryol Exp Morphol 24:203207.

    Brewster JL, Martin SL, Toms J, et al.2000.Deletion of Dad1 in mice inducesan apoptosis-associated embryonicdeath. Genesis 26:271278.

    Brison DR, Schultz RM. 1997. Apoptosisduring mouse blastocyst formation: ev-idence for a role for survival factorsincluding transforming growth factoralpha. Biol Reprod 56:10881096.

    Brison DR. 2000. Apoptosis in mamma-lian preimplantation embryos: regula-tion by survival factors. Hum Fertil(Camb) 3:3647.

    Broughton Pipkin F, RubinPC. 1994.Pre-eclampsiathe disease of theories. BrMed Bull 50:381396.

    Brown EJ, Baltimore D. 2000. ATR dis-ruption leads to chromosomal fragmen-tation and early embryonic lethality.Genes Dev 14:397402.

    Bui BV, Rees SM, Loeliger M, et al. 2002.Altered retinal function and structureafter chronic placental insufficiency. In-vest Ophthalmol Vis Sci 43:805812.

    Burton GJ, Jauniaux E, Watson AL. 1999.Maternal arterial connections to the pla-cental intervillous space during the firsttrimester of human pregnancy: theBoyd collection revisited. Am J ObstetGynecol 181:718724.

    Caniggia I, Mostachfi H, Winter J, et al.2000. Hypoxia-inducible factor-1 me-diates the biological effects of oxygenon human trophoblast differentiationthrough TGFbeta(3). J Clin Invest 105:577587.

    Caniggia IWY, Soleymanlou N, Chijiiwa

    M, et al. 2004. Evidence for decreasedapoptotic mediated trophoblast turn-over in high altitude placentae. Asilo-mar, CA. International Federation ofPlacental Association, Annual Meeting.

    Cartwright JE, Kenny LC, Dash PR, et al.2002. Trophoblast invasion of spiralarteries: a novel in vitro model. Pla-centa 23:232235.

    Chang CC, Lin DY, Fang HI, et al. 2005.Daxx mediates the small ubiquitin-likemodifier-dependent transcriptional re-pression of Smad4. J Biol Chem 280:1016410173.

    Chautan M, Chazal G, Cecconi F, et al.1999. Interdigital cell death can occur

    MOLECULAR MECHANISMS OF TROPHOBLAST SURVIVAL 275

    Birth Defects Research (Part C) 75:262280, (2005)

  • 8/13/2019 029-030 genetika implantacije (14)

    15/19

    through a necrotic and caspase-inde-pendent pathway. CurrBiol 9:967970.

    Chawengsaksophak K, de Graaff W, Ros-sant J, et al. 2004. Cdx2 is essentialfor axial elongation in mouse develop-ment. Proc Natl Acad Sci USA101:76417645.

    Chen Z, Naito M, Hori S, et al. 1999. A

    human IAP-family gene, apollon, ex-pressed in human brain cancer cells.Biochem Biophys Res Commun 264:847854.

    ChiMM, Schlein AL, Moley KH. 2000. Highinsulin-likegrowth factor 1 (IGF-1) andinsulin concentrations trigger apopto-sis in the mouse blastocyst via down-regulation of the IGF-1 receptor.Endocrinology 141:47844792.

    Chi MM, Hoehn A, Moley KH. 2002. Met-abolic changes in the glucose-inducedapoptoticblastocyst suggest alterationsin mitochondrial physiology. Am JPhysiol Endocrinol Metab 283:E226E232.

    Chipuk JE, Kuwana T, Bouchier-Hayes

    L, et al. 2004. Direct activation of Baxby p53 mediates mitochondrial mem-brane permeabilization and apoptosis.Science 303:10101014.

    Chipuk JE, Bouchier-Hayes L, Kuwana T,et al. 2005. PUMA couples the nuclearand cytoplasmic proapoptotic functionof p53. Science 309:17321735.

    Clarke AR, Maandag ER, van Roon M, etal. 1992. Requirement for a functionalRb-1 gene in murine development. Na-ture 359:328330.

    Clausson B, Gardosi J, Francis A, Cnat-tingius S. 2001. Perinatal outcome inSGA births defined by customised ver-sus population-based birthweight stan-

    dards. BJOG 108:830

    834.ConoverCA. 1992. Potentiationof insulin-

    like growth factor (IGF) action by IGF-binding protein-3: studies of underlyingmechanism. Endocrinology 130:31913199.

    Coucouvanis E, Martin GR. 1995. Sig-nals for death and survival:a two-stepmechanism for cavitation in the ver-tebrate embryo. Cell 83:279287.

    Cross JC, Werb Z, Fisher SJ. 1994. Im-plantation and the placenta: keypiecesof the development puzzle. Science266:15081518.

    Cross JC. 2000. Geneticinsights into tro-phoblast differentiation and placentalmorphogenesis. Semin Cell Dev Biol11:105113.

    Cross JC, Hemberger M, Lu Y, et al. 2002.Trophoblastfunctions,angiogenesis andremodeling of the maternal vascula-ture in the placenta. Mol Cell Endocri-nol 187:207212.

    DSa-Eipper C, Leonard JR, Putcha G, etal. 2001. DNA damage-induced neu-ral precursor cell apoptosis requiresp53and caspase 9 but neither Bax norcaspase 3. Development 128:137146.

    Danial NN, Korsmeyer SJ. 2004. Celldeath: critical control points. Cell 116:205219.

    de Klein A, Muijtjens M, van Os R, et al.2000. Targeted disruption of the cell-

    cycle checkpoint gene ATRleads to earlyembryonic lethality in mice. Curr Biol10:479482.

    Desai N, Kinzer D, Loeb A, Goldfarb J.1997. Use of synthetic serum substi-tuteand alpha-minimum essential me-dium forthe extendedculture of humanembryos to the blastocyst stage. Hum

    Reprod 12:328

    335.Detmar J, Shang Y, Taniuchi Y, et al.2004. A study of cell death gene ex-pression patterns in murine placentathroughout gestation. February 38,Meeting on Apoptosis in Development,Keystone Symposia, Keystone, CO.

    Di Berardino TJA and Jurisicova A. 2004.Theimportance of growthhormone forhuman pre-implantation embryo de-velopment, 50th Annual Meeting of Ca-nadian Fertility and Andrology Society,November 2427. Jasper, Alberta.

    Drake BL, Rodger JC. 1987. Phagocyticproperties of cultured murine tropho-blast. Placenta 8:129139.

    Dudek H, Datta SR, Franke TF, et al.1997. Regulation of neuronal survivalby the serine-threonine protein kinaseAkt. Science 275:661665.

    Ebner T, Yaman C, Moser M, et al. 2001.Embryo fragmentation in vitro and itsimpact on treatment and pregnancyoutcome. Fertil Steril 76:281285.

    Ecsedy JA,Michaelson JS,Leder P. 2003.Homeodomain-interacting protein ki-nase 1 modulates Daxx localization,phosphorylation, and transcriptionalac-tivity. Mol Cell Biol 23:950960.

    el-Deiry WS, Harper JW, OConnor PM,et al. 1994. WAF1/CIP1 is induced inp53-mediated G1 arrest and apopto-sis. Cancer Res 54:11691174.

    FabianD, Ilkova G,Rehak P, et al. 2004.Inhibitory effect of IGF-I on inducedapoptosis in mousepreimplantationem-bryos cultured in vitro. Theriogenol-ogy 61:745755.

    Gjorret JO, Knijn HM, Dieleman SJ, et al.2003. Chronology of apoptosis in bo-vine embryos produced in vivo and invitro. Biol Reprod 69:11931200.

    Godfrey KM. 2002. The role of the pla-centa in fetal programming-a review.Placenta 23(Suppl A):S20S27.

    Goncalves CR, Antonini S, Vianna-Mor-gante AM, et al. 2003. Developmentalchanges in the ploidy of mouse im-planting trophoblast cells in vitro. His-

    tochem Cell Biol 119:189198.Gostissa M, Morelli M, Mantovani F, et al.

    2004. The transcriptional repressorhDaxx potentiates p53-dependent ap-optosis. J BiolChem 279:4801348023.

    Greger JG, Katz RA,IshovAM, et al.2005.The cellular protein daxx interacts withavian sarcoma virus integrase and vi-ral DNA to repress viral transcription.J Virol 79:46104618.

    Gross VS, Hess M, Cooper GM. 2005.Mouse embryonic stem cells and pre-implantation embryos require signal-ing through the phosphatidylinositol3-kinase pathway to suppress apopto-sis. Mol Reprod Dev 70:324332.

    Gruslin A, Qiu Q, Tsang BK. 2001. X-linked inhibitor of apoptosis protein ex-pressionand the regulation of apoptosisduring human placental development.Biol Reprod 64:12641272.

    Gu J, Tamura M, Pankov R, et al. 1999.Shc and FAK differentially regulate cellmotility and directionality modulated

    by PTEN. J Cell Biol 146:389

    403.Guermah M, Ge K, Chiang CM, RoederRG. 2003. The TBN protein, which isessential for early embryonic mousedevelopment, is an inducible TAFII im-plicated in adipogenesis. Mol Cell 12:9911001.

    Handyside AH. 1986. Cell division anddeath in the mouse blastocyst beforeimplantation. Rouxs Arch Dev Biol 195:519526.

    Hao Y, Lai L, Mao J, et al. 2004a. Apo-ptosis in parthenogenetic preimplan-tation porcine embryos. Biol Reprod70:16441649.

    Hao Y, SekineK, Kawabata A, etal. 2004b.Apollon ubiquitinates SMAC and

    caspase-9, and has an essential cyto-protection function. Nat CellBiol 6:849860.

    Hardy K, Handyside AH, Winston RM.1989. Thehuman blastocyst: cell num-ber, death and allocation during latepreimplantation development in vitro.Development 107:597604.

    Hardy K. 1997. Cell death in the mam-malian blastocyst. Mol Hum Reprod3:919925.

    Hardy K. 1999. Apoptosis in the humanembryo. Rev Reprod 4:125134.

    Hardy K, Spanos S. 2002. Growth factorexpression and function in the humanand mouse preimplantation embryo. J

    Endocrinol 172:221

    236.HardyK, StarkJ, WinstonRM. 2003. Main-

    tenance of the inner cell mass in hu-man blastocysts from fragmentedembryos. Biol Reprod 68:11651169.

    Hauser HP, Bardroff M, Pyrowolakis G,Jentsch S. 1998. A giant ubiquitin-con-

    jugating enzyme related to IAP apo-ptosis inhibitors. J Cell Biol 141:14151422.

    Hemberger M, Cross JC. 2001. Genesgoverning placental development.Trends Endocrinol Metab 12:162168.

    Hemberger M, Nozaki T, Winterhager E,et al. 2003. Parp1-deficiency inducesdifferentiation of ES cells into tropho-blast derivatives. Dev Biol 257:371381.

    Hertig AT, Rock J, Adams EC, MenkinMC. 1959. Thirty-four fertilized humanova, good, bad and indifferent, recov-ered from 210 women of known fertil-ity; a study of biologic wastage in earlyhuman pregnancy. Pediatrics 23(Pt 2):202211.

    Heyer BS, MacAuley A, Behrendtsen O,Werb Z. 2000. Hypersensitivity to DNAdamage leads to increased apoptosisduring early mouse development.Genes Dev 14:20722084.

    Hinck L, Van Der Smissen P, Heuster-preute M, et al. 2001. Identification ofcaspase-3 and caspase-activated de-

    276 JURISICOVA ET AL.

    Birth Defects Research (Part C) 75:262280, (2005)

  • 8/13/2019 029-030 genetika implantacije (14)

    16/19

    oxyribonuclease in rat blastocysts andtheir implication in the induction of chro-matin degradation (but notnuclear frag-mentation) by highglucose. Biol Reprod64:555562.

    Hinck L, Thissen JP, De Hertogh R. 2003.Identification of caspase-6 in rat blas-tocysts and its implication in the in-

    duction of apoptosis by high glucose.Biol Reprod 68:18081812.Hitz C, Vogt-Weisenhorn D, Ruiz P, et al.

    2005. Progressive loss of the spongio-trophoblast layer of Birc6/Bruce mu-tants results in embryonic lethality.Genesis 42:91103.

    Hsu SY, Kaipia A, McGee E, et al. 1997.Bok is a pro-apoptotic Bcl-2 proteinwith restricted expression in reproduc-tive tissues and heterodimerizes withselective anti-apoptotic Bcl-2 familymembers. Proc Natl Acad Sci USA 94:1240112406.

    Hsu SY, Hsueh AJ. 1998. A splicing vari-ant of the Bcl-2 member Bok with a

    truncated BH3 domain induces apo-ptosis but