ArticlePDF Available

Cleavage of MCM2 licensing protein fosters senescence in human keratinocytes

Taylor & Francis
Cell Cycle
Authors:
  • Atomwise

Abstract

In eukaryotic cells, MCM, the minichromosome maintenance proteins, form a heterohexamer during G(1) phase in the cell cycle and constitute a DNA helicase activity at the onset of replication. MCM proteins are downregulated and dissociated from chromatin when cells exit the cell cycle. MCM proteins are upregulated frequently in a variety of dysplastic and cancer cells. To delineate the role of MCM in esophageal epithelial biology, we determined the MCM family gene expression during cellular senescence, immortalization, differentiation and apoptosis. All of the MCM2-7 proteins appeared to be downregulated in primary human esophageal keratinocytes upon replicative senescence. Their expression was restored by ectopic expression of a catalytic subunit of human telomerase, resulting in immortalization. Interestingly, we found a reciprocal induction of a novel MCM2-related protein fragment upon cell growth inhibition associated with senescence, contact inhibition or terminal differentiation, but not apoptosis. Epitope mapping of this MCM2-related fragment suggested the lack of amino- and carboxyl-terminal regions, including one of the putative nuclear localization signals and the ATPase domain, the MCM box. The absence of multiple MCM2 transcripts implied a possible posttranslational molecular cleavage in generation of the MCM2-related fragment, and a potential functional role in the regulation of the activity of the MCM protein complex.
Full Terms & Conditions of access and use can be found at
https://www.tandfonline.com/action/journalInformation?journalCode=kccy20
Cell Cycle
ISSN: 1538-4101 (Print) 1551-4005 (Online) Journal homepage: https://www.tandfonline.com/loi/kccy20
Cleavage of MCM2 licensing protein fosters
senescence in human keratinocytes
Hideki Harada, Hiroshi Nakagawa, Munenori Takaoka, James Lee, Meenhard
Herlyn, J. Allan Diehl & Anil K. Rustgi
To cite this article: Hideki Harada, Hiroshi Nakagawa, Munenori Takaoka, James Lee, Meenhard
Herlyn, J. Allan Diehl & Anil K. Rustgi (2008) Cleavage of MCM2 licensing protein fosters
senescence in human keratinocytes, Cell Cycle, 7:22, 3534-3538, DOI: 10.4161/cc.7.22.7043
To link to this article: https://doi.org/10.4161/cc.7.22.7043
Published online: 15 Nov 2008.
Submit your article to this journal
Article views: 68
Citing articles: 10 View citing articles
© 2008 LANDES BIOSCIENCE. DO NOT DISTRIBUTE.
[Cell Cycle 7:22, 3534-3538; 15 November 2008]; ©2008 Landes Bioscience
3534 Cell Cycle 2008; Vol. 7 Issue 22
In eukaryotic cells, MCM, the minichromosome maintenance
proteins, form a heterohexamer during G1 phase in the cell cycle
and constitute a DNA helicase activity at the onset of replication.
MCM proteins are downregulated and dissociated from chromatin
when cells exit the cell cycle. MCM proteins are upregulated
frequently in a variety of dysplastic and cancer cells. To delineate
the role of MCM in esophageal epithelial biology, we determined
the MCM family gene expression during cellular senescence,
immortalization, differentiation and apoptosis. All of the MCM2-7
proteins appeared to be downregulated in primary human esopha-
geal keratinocytes upon replicative senescence. Their expression
was restored by ectopic expression of a catalytic subunit of human
telomerase, resulting in immortalization. Interestingly, we found
a reciprocal induction of a novel MCM2-related protein fragment
upon cell growth inhibition associated with senescence, contact
inhibition or terminal differentiation, but not apoptosis. Epitope
mapping of this MCM2-related fragment suggested the lack of
amino- and carboxyl-terminal regions, including one of the puta-
tive nuclear localization signals and the ATPase domain, the MCM
box. The absence of multiple MCM2 transcripts implied a possible
posttranslational molecular cleavage in generation of the MCM2-
related fragment, and a potential functional role in the regulation
of the activity of the MCM protein complex.
Introduction
DNA replication occurs in a precise fashion during eukaryotic
cell division. This tight control is orchestrated by many regulatory
molecules, including members of the minichromosome mainte-
nance gene family, designated as MCM. Initially, MCM proteins are
recruited to sites of DNA replication and interact with each other,
forming the MCM2-7 complex during G1 phase.1 This complex has
helicase activity and facilitates DNA replication.2,3 Therefore, the
MCM proteins are essential for proliferating cells. In fact, MCM
proteins are upregulated frequently in a variety of dysplastic and
cancer cells.4-6
Normal human epithelial cells are limited in their prolifera-
tive capacity and eventually undergo differentiation, senescence or
apoptosis. In this context, deregulated cells have mechanisms to
suppress such processes, thereby resulting in unlimited cell prolifera-
tion, termed immortalization. Immortalization of human esophageal
epithelial cells can be achieved by the ectopic expression of human
telomerase, hTERT.7 Importantly, these cells maintain cell cycle
checkpoints such as p16INK4a/pRb and p14ARF/p53/p21WAF1.7
When oncogenic Ha-Ras is expressed ectopically in immortalized
human esophageal epithelial cells, these cells undergo senescence,
accompanied by upregulation of p16INK4a and hypophosphory-
lated pRb.8 To understand the molecular mechanisms underlying
constrained cellular growth arrest induced by senescence or differ-
entiation, we used normal human esophageal epithelial cells (EPC2)
and their derivative immortalized cells (EPC2-hTERT). We found
that MCM2 was cleaved in senescence, while cancer cells prevented
MCM2 from being cleaved. These results might indicate that the
cleavage of MCM2 plays a critical role in cellular growth arrest
induced by senescence or differentiation.
Results
A novel MCM2-related protein fragment is induced upon
replicative senescence through a post transcriptional mechanism.
We have previously carried out extensive characterization of EPC2
primary normal human esophageal epithelial cells.7 EPC2 cells
cease proliferation as they undergo replicative senescence by 44 PD
with an induction of p16INK4a protein and senescence-associated
β-galactosidase activity. Retrovirus-mediated stable transduction of a
catalytic subunit of telomerase (hTERT) in the presenescent EPC2
cells (42 PD) permitted the cells to reenter the cell cycle and resulted
in immortalization.7 The MCM family proteins are associated with
cell proliferation.9 In agreement, all of the examined MCM family
members were found downregulated as EPC2 cells underwent
senescence as documented by a reduced phosphorylation level of
pRB protein (Fig. 2A). By contrast, their expression was reversed
*Correspondence to: Anil K. Rustgi; 600 CRB; University of Pennsylvania; 415
Curie Blvd.; Philadelphia, Pennsylvania 19104 USA; Tel.: 215.898.0154; Fax:
215.573.5412; Email: anil2@mail.med.upenn.edu
Submitted: 08/29/08; Accepted: 09/18/08
Previously published online as a Cell Cycle E-publication:
http://www.landesbioscience.com/journals/cc/article/7043
Report
Cleavage of MCM2 licensing protein fosters senescence in human
keratinocytes
Hideki Harada,1,3,† Hiroshi Nakagawa,1,3,† Munenori Takaoka,1,3 James Lee,1,3 Meenhard Herlyn,4 J. Alan Diehl5,6 and
Anil K. Rustgi1-3,*
1Gastroenterology Division and Department of Medicine; 2Department of Genetics; 3Abramson Cancer Center; 4Wistar Institute; 5Department of Cancer Biology; 6Abramson
Family Cancer Research Institute; University of Pennsylvania; Philadelphia, Pennsylvania USA
Co-first authors
Key words: MCM2, senescence, differentiation, esophageal keratinocytes
© 2008 LANDES BIOSCIENCE. DO NOT DISTRIBUTE.
MCM2 and cellular senescence
www.landesbioscience.com Cell Cycle 3535
upon hTERT transduction (Fig. 2A). Interestingly, a novel band was
induced reciprocally in senescing cells (Fig. 2A). This molecular mass
of approximately 55 kDa was detected by an anti-MCM2 antibody
raised against synthetic peptides corresponding to amino acid resi-
dues 131–150 of the MCM2 protein (Fig. 1), and thus designated
as a MCM2-related fragment. Since there is no known protein with
substantive similarity to this peptide sequence, we hypothesized that
the MCM2-related fragment may represent a novel splicing variant
or posttranslational cleavage of MCM2, and that its expression is
associated with the status of cell proliferation.
To determine a possible splicing variant, quantitative PCR was
performed by employing three independent sets of primers and a
TaqMan® probe, in which the MCM2 exon 3–4 boundary region is
specifically detected by the sequence encoding amino acids 131–150
of MCM2. Figure 2B demonstrates that all of the TaqMan® assays
detected a reduction in MCM2 mRNA expression upon cellular
senescence in parallel with the protein level. In addition, Northern
blotting with a full length MCM2 cDNA probe detected a single
transcript only throughout the PDs of EPC2 cells with or without
hTERT expression (data not shown), although the sensitivity of
detecting low copy mRNA species was not as great as that by PCR.
These observations argue against the idea that a unique splicing
variant is expressed upon cellular senescence.
MCM2-related protein fragment is also induced upon contact
inhibition and terminal differentiation, but not apoptosis. We next
tested whether or not the MCM2-related fragment may be induced
by cellular conditions other than senescence and detected by other
MCM2 antibodies raised against different epitopes within the
amino-terminal or carboxyl-terminal regions (Fig. 1). First, EPC2
cells were grown in monolayer culture until they reach an overly
confluent status. Contact inhibition was confirmed by decreased cell
proliferation and pRB phosphorylation (Fig. 3B). As shown in Figure
3A, the MCM2-related fragment was detected progressively by only
the anti-MCM2 antibody recognizing amino acid residues 131–150
in post confluent day 3 through day 11 while downregulation of the
full length MCM2 protein was detected consistently by all of the
antibodies in postconfluent cells (Fig. 3A). Thus, the MCM2-related
fragment was thought to lack both amino- and carboxyl-termini of
the full length MCM2 protein as denoted in Figure 1.
Such an induction of the MCM2-related fragment was observed
also in hTERT-immortalized EPC2 cells upon contact inhibition
(Fig. 3C and data not shown). Of note, the MCM2-related fragment
was expressed weakly in subconfluent EPC2 cells, where subpopula-
tion cells undergoes spontaneous senescence in primary culture (Figs.
3A and 4A).
Next, EPC2 and EPC2-hTERT cells were treated with calcium
chloride to induce terminal differentiation as indicated by induction
of involucrin (Fig. 4). Again, the full length MCM2 was down-
regulated and the MCM2-related fragment was induced, indicating
that the MCM2-related fragment can be induced upon cell growth
inhibition associated with cellular senescence, contact inhibition or
terminal differentiation. Finally, cells were exposed to actinomycin
D or gamma-irradiation. While apoptosis was induced within 12–24
hours as indicated by fragmentation of caspase 3, the MCM2-related
fragment was not detected (Fig. 5 and data not shown). Importantly,
the full length MCM2 level was not altered by apoptosis. These data
indicate that the MCM2 protein undergoes a molecular shift from
the full-length form to a short form upon chronic, but not acute,
induction of cell growth inhibition or cell cycle arrest.
MCM2 protein expression may be regulated through post-
translational modification. The above data suggested that the full
length MCM2 protein may be cleaved to generate the MCM2-
related fragment upon cell growth inhibition. However, the lack of
both amino- and carboxyl-terminal regions (Fig. 1) precluded us
from determining whether the MCM2-related fragment is derived
from the full-length form of MCM2 protein. Nonetheless, we
expressed ectopically an amino-terminally Flag-epitope tagged full-
length MCM2 protein in EPC2-hTERT cells. Flag-tagged MCM2
was expressed constitutively under the retroviral LTR promoter.
Interestingly, contact inhibition or terminal differentiation reduced
the expression of the Flag-tagged MCM2 (Figs. 3C and 4B),
suggesting that MCM2 protein may undergo degradation and/or
cleavage although the resulting putative MCM2-related fragment per
se could not be detected by an anti-Flag antibody.
Figure 1. Schematic representation of the MCM2 protein and a novel MCM2-related fragment. Horizontal bars indicate the primary structure of the 120
kDa MCM2 protein and putative molecular mass of approximately 55 kDa (MCM2-RF), sharing an epitope recognized by an antibody directed against
amino acids 131–150 of human MCM2. Failure of detection of the MCM2-RF by antibodies directed against peptides containing amino acids 1–50 or
amino acids 805–904 (see Figs. 3–5), indicated by horizontal arrow, predicted the molecular region of MCM2-RF. MCM2-RF is likely to contain one of
two putative nuclear-targeting sequences (NTS) and a Zn2+ finger motif, but not the MCM box, encompassing Walker A/B ATPase motifs, evolutionarily
conserved in the MCM2-7 family. For quantitative determination of MCM2 mRNA, TaqMan® Gene Expression Assays were carried out detecting transcripts
derived from exonic boundaries of the MCM2 gene, indicated by open vertical bars. Note that the TaqMan® assay detecting the exon 3–4 overlaps the
gene region encoding amino acids 131–150.
© 2008 LANDES BIOSCIENCE. DO NOT DISTRIBUTE.
MCM2 and cellular senescence
3536 Cell Cycle 2008; Vol. 7 Issue 22
Our data with MCM2 mRNA quantitation with three indepen-
dent probe sets (Fig. 2B) did not support the possibility of de novo
transcription of an alternative spliced form to account for the novel
MCM2-related fragment. Although post-translational cleavage of
MCM2 protein is suggested, neither the precise cleavage site(s) nor
the responsible proteases were identified in the present study. First,
in silico analysis using the peptide cutter program10 failed to identify
promising cleavage sites for MCM2 compatible with the predicted
cleavage pattern (Fig. 1) based upon the distribution of epitopes for
the antibodies we used. Second, the lack of either amino- or carboxyl
terminal region in the MCM2 related fragment hampered the use of
epitope-tagged protein to be expressed ectopically. In fact, amino-
terminally Flag-epitope tagged MCM2 only disappeared following
differentiation (Fig. 3C). Finally, a low protein yield from the senes-
cent or terminally differentiated cells limited affinity purification
with an antibody. Amongst the MCM family members, MCM3 has
been reported to undergo proteolytic cleavage mediated by Caspase-
3.11 Consistent with that notion, MCM2 remained uncleaved upon
caspase-3 activation (Fig. 5).
Squamous epithelial cells, or keratinocytes, are distinguished
by their proliferation-differentiation gradient and renewal during
homeostasis and tissue regeneration. Such cells slough that is trig-
gered by apoptosis and senescence prior to renewal. While the
expression of MCMs is decreased, if not lost, during differentiation
in somatic cells,12 it is unclear to what extent changes in their struc-
tures and functions impact upon differentiation. We have discovered
In aggregate, the MCM2 protein may give rise to a putative
cleaved form designated as a MCM2-related fragment, lacking both
amino- and carboxyl-terminal regions as well as the MCM box of
MCM2 (Fig. 1) upon cellular senescence, contact inhibition or
terminal differentiation, but not apoptosis.
Discussion
MCM2-7 proteins are related to each other and form a complex
for DNA synthesis initiation. The MCM complex binds origins of
DNA replication during the late M/early G1 phases of the cell cycle.
Through the action of S phase protein kinases, the MCM complexes
unwind the double-stranded DNA at the origin, with subsequent
recruitment of DNA polymerases and initiation of DNA synthesis.
They then disengage from replication origins, so DNA replication
cannot be reinitiated. MCMs are targets of S phase checkpoints.
That being said, in premalignant and malignant conditions, the
MCMs are often expressed abnormally, and have been advocated
as serving as diagnostic biomarkers to distinguish between normal
cells and malignant cells, and in cancers themselves as potential
therapeutic targets. In fact, we have observed marked upregulation
of MCM2 mRNA and its 120-kDa protein product in primary
esophageal tumor tissues as well as actively proliferating esophageal
cell lines (Fig. 2 and unpublished observations). Nevertheless, by
virtue of MCM dysregulation, chromosomal aberrations accrue
during tumorigenesis; loss of MCM proteins triggers DNA damage
and genomic instability.
Figure 2. Reciprocal expression of MCM2 protein and the MCM2 fragment with senescence and immortalization of primary human esophageal keratino-
cytes. (A) Western blotting demonstrates progressive downregulation of examined MCM2-7 family proteins, including the 120 kDa MCM2 (solid arrow)
in EPC2 cells undergoing replicative senescence by 42 population doublings (PD) while they were reinduced variably upon immortalization of EPC2 by
hTERT. By contrast, an MCM2 fragment (open arrow) was reciprocally induced upon senescence and suppressed by immortalization when Western blot
was carried out with an MCM2 antibody directed against amino acids 131–150 of MCM (Fig. 1). Cellular senescence was accompanied by a reduction
in the phosphorylation level of RB protein. MCM2, but not the MCM2 fragment, was detected in TE3 esophageal cancer cells, an esophageal squamous
cell carcinoma cell line. β-actin was used as a loading control. Note that protein yield from presenescent cells (42PD) was low and only 1 μg of protein
was loaded while other lanes were loaded with 10 μg of protein. Nonetheless, the MCM2 fragment was detectable at 42PD. (B) MCM2 mRNA level was
determined by TaqMan® Gene Expression Assays at indicated population doublings using a set of sequence specific primers and a probe detecting different
exonic junctions of the MCM2 gene transcript as described in Figure 1.
© 2008 LANDES BIOSCIENCE. DO NOT DISTRIBUTE.
MCM2 and cellular senescence
www.landesbioscience.com Cell Cycle 3537
instructions. The MCM2 mRNA was determined by TaqMan® Gene
Expression Assays (Applied Biosystems) using three independent
sets of primers and the probes (Hs00170472_m1 for exons 2–3,
Hs01091568_g1 for exons 3–4 and Hs01091564_m1 for exons
13–14), targeting different exon boundaries of the MCM2 transcript
(MN_004526.2). SYBR green reagent (Applied Biosystems) was
used to quantitate mRNA for GAPDH as an internal control. All
PCR reactions were performed in triplicate. The relative expression
level of MCM2 mRNA was calculated by normalizing to GAPDH
that while all of the MCM family members were
markedly downregulated during cellular senescence
(Fig. 2) and differentiation (data not shown), there
is the emergence of a unique fragmented MCM2
protein that may arise due to cleavage, and lacks both
the amino- and carboxyl domains as well as the MCM
box. It is conceivable that this fragment is responsible
for preventing DNA replication during periods of
prolonged or terminal differentiation, and/or during
senescence. In a recent study, Komamura-Kohno Y, et
al., analyzed the MCM2 protein biochemically through
partial digestion with trypsin.13 They demonsrated
that MCM2 fragments derived from the C-terminal
region inhibit DNA helicase activity through their
ability to bind to ssDNA. By contrast, two fragments
(148–441 and 442–676) from the central region were
responsible chiefly for the interaction between MCM2
and MCM4.13 Thus, the MCM2 fragment identified
in our study may prevent assembly of the MCM2-7
complex. It is also quite possible that fragments of
MCM2 might confer different effects in comparison
to full-length MCM2, depending upon subcellular
localization based upon solubility, or lack of it, as has
been reported in mouse ovarian oocytes,14 implying
there may be posttranslational modification of MCM2
in association with different functional properties.
Materials and Methods
Cell culture, retroviral vectors and retroviral trans-
duction. Normal human esophageal epithelial cells
(EPC2) and their hTERT-immortalized derivative
(EPC2-hTERT) were described previously.7 A full-
length human MCM2 cDNA was generated by reverse
transcription-polymerase chain reaction (RT-PCR)
using total RNA extracted from EPC2 cells as a
template. The PCR product, tagged with BamHI and
XhoI sites at the 5' and 3' ends, respectively, was
cloned into the pCMV FLAG-tagging mammalian
expression vector (Stratagene). For stable expression,
FLAG-tagged human MCM2 coding sequence was
further subcloned into the pBabe puromycin-resistant
retrovirus vector.15 Retrovirus production and infec-
tion were carried out as described previously.7,16
In brief, retroviral expression vectors were trans-
fected into Phoenix-Ampho packaging cells and the
virus-containing culture medium supernatants were
collected 48 and 72 hours post-transfection. EPC2-
hTERT cells were infected with the virus in a 6-well
plate (0.5 x 105 cells/well), followed by drug selection with
0.5 μg/mL of puromycin for 5 days.
RNA extraction, cDNA preparation and real-time PCR. Briefly,
total RNA was isolated from cells with the RNeasy Mini Kit (Qiagen,
Inc., Valencia, CA). DNaseI treatment was done on column and
cDNA was synthesized with the SuperscriptTM First Strand Synthesis
System (Invitrogen, Carlsbad, CA). Real-time PCR was performed
using the ABI PRISM® 7000 Sequence Detection System (Applied
Biosystems, Foster City, CA), according to the manufacturer’s
Figure 3. Contact inhibition of human primary esophageal keratinocytes is associated with
reduction in the full-length MCM2 protein and induction of the MCM2 fragment through a
posttranscriptional mechanism. (A) Western blotting using anti-MCM2 antibodies recogniz-
ing different epitopes as depicted in Figure 1 detects changes in expression of full-length
MCM2 (solid arrow) and an MCM2-related fragment (open arrow) in EPC2 cells harvested
at various cell densities. In (B), contact inhibition was documented by a reduction in cell pro-
liferation. 3H-thymidine incorporation was adjusted by the amount of protein from the whole
cell lysate used for Western blotting to detect the hyper-phosphorylated form of RB protein
shown in the insert. In (A and B), days elapsed after 100% confluent status are indicated in
the parentheses. (C) EPC2-hTERT cells stably transduced with amino-terminally Flag-epitope
tagged MCM2 protein (Flag-MCM2) or a control empty vector (puro) were harvested at
90% cell density or 3 days after reaching 100% confluency and subjected to Western blot-
ting with the indicated antibody. Note that anti-Flag antibody detected a reduced level of
exogenously expressed epitope-tagged full-length MCM2 protein but not MCM2 protein that
lacks the amino-terminal sequence (Fig. 1). β-actin was used as a loading control.
© 2008 LANDES BIOSCIENCE. DO NOT DISTRIBUTE.
MCM2 and cellular senescence
3538 Cell Cycle 2008; Vol. 7 Issue 22
Figure 5. Caspase-3 activation does not affect MCM2 protein. EPC2-hTERT
cells were treated with or without 2 μg/ml actinomycin D (Act. D) for 5 hours
(A) or exposed to indicated amount of γ-ray for 24 hours (B) and subjected
to Western blotting to detect MCM2 (solid arrow), full-size (open triangle) or
fragmented (solid triangle) forms of caspase-3 (Casp.-3), and Phospho-Chk2
(P-Chk2). β-actin was used as a loading control.
6. Laskey R. The Croonian Lecture 2001 hunting the antisocial cancer cell: MCM proteins
and their exploitation. Philos Trans R Soc Lond B Biol Sci 2005; 360:1119-32.
7. Harada H, Nakagawa H, Oyama K, Takaoka M, Andl CD, Jacobmeier B, von Werder A,
Enders GH, Opitz OG, Rustgi AK. Telomerase induces immortalization of human esopha-
geal keratinocytes without p16INK4a inactivation. Mol Cancer Res 2003; 1:729-38.
8. Takaoka M, Harada H, Deramaudt TB, Oyama K, Andl CD, Johnstone CN, Rhoades B,
Enders GH, Opitz OG, Nakagawa H. Ha-Ras(G12V) induces senescence in primary and
immortalized human esophageal keratinocytes with p53 dysfunction. Oncogene 2004;
23:6760-8.
9. Stoeber K, Tlsty TD, Happerfield L, Thomas GA, Romanov S, Bobrow L, Williams ED,
Williams GH. DNA replication licensing and human cell proliferation. J Cell Sci 2001;
114:2027-41.
10. Gasteiger E, Gattiker A, Hoogland C, Ivanyi I, Appel RD, Bairoch A. ExPASy: The
proteomics server for in-depth protein knowledge and analysis. Nucleic Acids Res 2003;
31:3784-8.
11. Schwab BL, Leist M, Knippers R, Nicotera P. Selective proteolysis of the nuclear replication
factor MCM3 in apoptosis. Exp Cell Res 1998; 238:415-21.
12. Barkley LR, Hong HK, Kingsbury SR, James M, Stoeber K, Williams GH. Cdc6 is a rate-
limiting factor for proliferative capacity during HL60 cell differentiation. Exp Cell Res
2007; 313:3789-99.
13. Komamura-Kohno Y, Tanaka R, Omori A, Kohno T, Ishimi Y. Biochemical characterization
of fragmented human MCM2. Febs J 2008; 275:727-38.
14. Swiech L, Kisiel K, Czolowska R, Zientarski M, Borsuk E. Accumulation and dynamics of
proteins of the MCM family during mouse oogenesis and the first embryonic cell cycle. Int
J Dev Biol 2007; 51:283-95.
15. Morgenstern JP, Land H. Advanced mammalian gene transfer: high titre retroviral vectors
with multiple drug selection markers and a complementary helper-free packaging cell line.
Nucleic Acids Res 1990; 18:3587-96.
16. Andl CD, Mizushima T, Nakagawa H, Oyama K, Harada H, Chruma K, Herlyn M, Rustgi AK.
Epidermal growth factor receptor mediates increased cell proliferation, migration, and aggrega-
tion in esophageal keratinocytes in vitro and in vivo. J Biol Chem 2003; 278:1824-30.
mRNA expression level. Data were analyzed using ABI PRISM®
7000 sequence detection system software (Applied Biosystems).
Immunoprecipitation and western blot analysis. 10 μg protein
from whole-cell extracts was separated by SDS-PAGE and transferred
to Immobilon-P membranes (Millipore). The membranes were incu-
bated with anti-MCM2 (A300-191A) (amino acid residues 1–50)
(Bethyl laboratories, Montgomery, TX), anti-MCM2 (559542)
(amino acid residues 131–150) (BD Pharmingen), anti-MCM2
(clone 6A8) (amino acid residues 805–904) (Novus Biologicals,
Littleton, CO), anti-MCM3 (BD Pharmingen), anti-MCM4 (BD
Pharmingen), anti-MCM5 (BD Pharmingen), anti-MCM6 (BD
Pharmingen), anti-MCM7 (BD Pharmingen), anti-FLAG M2
(Sigma), AC-74 against β-actin (Sigma), and DMA1A + DM1B
against tubulin (Neo Markers). Staining was detected by using
ECL Plus (Amersham Pharmacia biotech). The FLAG-tagged
proteins were immunoprecipitated using FLAG Tagged Protein
Immunoprecipitation Kit (Sigma).
Acknowledgements
This research is supported by NCI grant PO1-CA098101 (M.H.,
J.A. H.N, A.K.R), the NIH/NIDDK Center for Molecular Studies
in Digestive and Liver Diseases P30-DK50306, the Morphology
Core, Molecular Biology Core and Cell Culture Core facilities, and
NIH grant R01-DK077005 (H.N.).
References
1. Kearsey SE, Labib K. MCM proteins: evolution, properties and role in DNA replication.
Biochim Biophys Acta 1998; 1398:113-36.
2. Maiorano D, Lutzmann M, Mechali M. MCM proteins and DNA replication. Curr Opin
Cell Biol 2006; 18:130-6.
3. Forsburg SL. Eukaryotic MCM proteins: beyond replication initiation. Microbiol Mol Biol
Rev 2004; 68:109-31.
4. Alison MR, Hunt T, Forbes SJ. Minichromosome maintenance (MCM) proteins may be
pre-cancer markers. Gut 2002; 50:290-1.
5. Freeman A, Morris LS, Mills AD, Stoeber K, Laskey RA, Williams GH, Coleman N.
Minichromosome maintenance proteins as biological markers of dysplasia and malignancy.
Clin Cancer Res 1999; 5:2121-32.
Figure 4. Terminal differentiation in primary human esophageal keratinocytes
is associated with a reciprocal expression of MCM2 and the MCM2 frag-
ment. Subconfluent EPC2 cells (A) and EPC2-hTERT cells stably expressing
Flag-epitope tagged MCM2 (B) were treated for up to 72 hours with 1.8 mM
CaCl2 to induce terminal differentiation which was documented by Western
blotting for involucrin. Indicated anti-MCM2 antibodies and anti-Flag anti-
body detected full size MCM2 (solid arrow) or a MCM2 fragment (open
arrow). β-actin was used as a loading control.
... It has been reported that chronic DNA replication stress reduces the replicative lifespan of mouse cells by p53-dependent microRNA-assisted MCM2-7 down-regulation (Bai et al., 2016). It has been reported that MCM2 is cleaved to produce a 55-kDa protein in senescent keratinocytes (Harada et al., 2008). MCM2-7 proteins are largely localized in nuclei to bind to the replication origins in mammalian cells. ...
... We showed by western blotting analysis that protein levels of MCM2, MCM3, MCM4 and MCM6 decrease at near-senescence (Fig. 2, Supplementary Fig. S2, S3 and S4), although the mode of the change varies significantly among different cell culture series, perhaps due to differences in cell conditions. MCM2 is reportedly cleaved to generate a 55-kDa fragment lacking the amino terminus and the carboxylterminal half region of full-length MCM2 in senescent keratinocytes (Fig. 6) (Harada et al., 2008). Our data show that in WI38 cells, MCM2 is specifically fragmented among MCM2-7, and various MCM2 fragments larger than 55 kDa are produced. ...
Article
Cellular aging is characterized by the loss of DNA replication capability and is mainly brought about by various changes in chromatin structure. Here, we examined changes in MCM2–7 proteins, which act as a replicative DNA helicase, during aging of human WI38 fibroblasts at the single-cell level. We used nuclear accumulation of p21 as a marker of senescent cells, and examined changes in MCM2–7 by western blot analysis. First, we found that senescent cells are enriched for cells with a DNA content higher than 4N. Second, the levels of MCM2, MCM3, MCM4 and MCM6 proteins decreased in senescent cells. Third, cytoplasmic localization of MCM2 and MCM7 was observed in senescent cells, from an analysis of MCM2–7 except for MCM5. Consistent with this finding, fragmented MCM2 was predominant in these cells. These age-dependent changes in MCM2–7, a protein complex that directly affects cellular DNA replication, may play a critical role in cellular senescence.
... The MCM complex is a double-hexamer ring structure of six MCM proteins, MCM2-MCM7, and its expression is tightly regulated at both the initiation and elongation stages of eukaryotic DNA replication (21). Elevated expression of MCM subunits is associated with cell transformation, whereas MCM subunit downregulation and cleavage are linked to reduced growth potential and induction of senescence in the epithelium, including in esophageal epithelial cells (22)(23)(24). Treating esophageal epithelial cells with the MCM complex inhibitor ciprofloxacin (25) reduced epithelial proliferation in vitro and improved histopathologic features in a murine model of EoE. Collectively, through comprehensive proteomic analysis, we identify in situ upregulation of the MCM complex in EoE, demonstrate that inhibiting this complex with ciprofloxacin counteracts BZH, and propose that ciprofloxacin and other MCM inhibitors may be therapeutic for EoE. ...
Article
Full-text available
Eosinophilic esophagitis (EoE) is a chronic gastrointestinal disorder characterized by food antigen-driven eosinophilic inflammation and hyperproliferation of esophageal mucosa. By utilizing a large-scale, proteomic screen of esophageal biopsies, we aimed to uncover molecular drivers of the disease. Proteomic analysis by liquid chromatography-tandem mass spectrometry identified 402 differentially expressed proteins (DEPs) that correlated with the EoE transcriptome. Immune cell-related proteins were among the most highly upregulated DEPs in EoE compared to controls, whereas proteins linked to epithelial differentiation were primarily downregulated. Notably, in the inflamed esophageal tissue, all six subunits of the minichromosome maintenance (MCM) complex, a DNA helicase essential for genomic DNA replication, were significantly upregulated at the gene and protein levels. Furthermore, treating esophageal epithelial cells with a known inhibitor of the MCM complex (ciprofloxacin) blocked esophageal epithelial proliferation. In a murine model of EoE driven by overexpression of IL-13, ciprofloxacin treatment decreased basal zone thickness and reduced dilated intercellular spaces by blocking the transition of the epithelial cell through the S-phase of the cell cycle. Collectively, a broad-spectrum proteomic screen has identified the involvement of the MCM complex in EoE and has highlighted MCM inhibitors as potential therapeutic agents for the disease.
... The 2D esophageal cancer cell line is also an important tool for EAC and ESCC esophagus. Telomerase-immortalized human esophageal epithelial cell lines have been useful in studying esophageal proliferation, differentiation, and malignant transformation [24][25][26][27]. Organotypic 3D culture (OTC) is a form of tissue engineering in which esophageal epithelium is cultured at the air-liquid interface in a liquid medium containing fibroblasts and type 1 collagen [28]. ...
Article
Full-text available
Organoids have been developed in the last decade as a new research tool to simulate organ cell biology and disease. Compared to traditional 2D cell lines and animal models, experimental data based on esophageal organoids are more reliable. In recent years, esophageal organoids derived from multiple cell sources have been established, and relatively mature culture protocols have been developed. Esophageal inflammation and cancer are two directions of esophageal organoid modeling, and organoid models of esophageal adenocarcinoma, esophageal squamous cell carcinoma, and eosinophilic esophagitis have been established. The properties of esophageal organoids, which mimic the real esophagus, contribute to research in drug screening and regenerative medicine. The combination of organoids with other technologies, such as organ chips and xenografts, can complement the deficiencies of organoids and create entirely new research models that are more advantageous for cancer research. In this review, we will summarize the development of tumor and non-tumor esophageal organoids, the current application of esophageal organoids in disease modeling, regenerative medicine, and drug screening. We will also discuss the future prospects of esophageal organoids.
... Fig 1D), when G1 arrest (Suppl. Fig 135 1E), nor markers of senescence such as loss of MCM2 (Harada et al, 2008), were observed 136 (Suppl. Fig 1F). ...
Preprint
Full-text available
Cells are equipped to defend themselves from invading pathogens through sensors such as cGAS, which upon binding DNA induces type I interferon (IFN) expression. Whilst IFNs are crucial for limiting viral infection and activating adaptive immunity, uncontrolled production causes excessive inflammation and autoimmunity. cGAS binds DNA of both pathogenic and cellular origin and its activity is therefore tightly regulated. This is particularly apparent during mitosis, where cGAS association with chromatin following nuclear membrane dissolution and phosphorylation by mitotic kinases negatively regulate enzymatic activity. Here we describe a novel mechanism by which DNA sensing and other innate immune pathways are regulated during cell division, dependent on cyclin dependent kinases (CDK) 4 and 6. Inhibition of CDK4/6 using chemical inhibitors, shRNA-mediated depletion, or overexpression of cellular CDK4/6 inhibitor p16INK4a, greatly enhanced DNA- or cGAMP-induced expression of cytokines and IFN-stimulated genes (ISG). Mechanistically, CDK4/6-dependent inhibition mapped downstream of cytoplasmic signalling events including STING and IRF3 phosphorylation, limiting innate immune induction at the level of IFNβ mRNA expression. This regulation was universal, occurring in primary and transformed cells of human and murine origin, and broad, as IFNβ expression was inhibited in a CDK4/6-dependent manner downstream of multiple pattern recognition receptors. Together these findings demonstrate that host innate responses are limited by multiple mechanisms during cell division, thus defining cellular replication as an innate immune privileged process that may be necessary to avoid aberrant self-recognition and autoimmunity.
... Similarly, Yasin et al. showed that 28% of BCs in vitro expressed MCM2 (Yasin et al. 2010). MCM2 is a DNA replication licensing factor and is expressed at all phases of the proliferative cell cycle, but its expression is down-regulated in the quiescent and senescent cells (Stoeber et al. 2001;Harada et al. 2008;Chen et al. 2004) The expression of MCM2 can identify cycling cells as well as those with proliferative potential (Stoeber et al. 2001) compared to Ki-67 which can only detect the former. Therefore, MCM2 is considered as a more sensitive marker of proliferative capacity than Ki-67. ...
Conference Paper
Introduction: Focal cortical dysplasia (FCD) is a malformation of cortical development that is a frequent cause of multidrug resistant paediatric epilepsy. FCD type IIb (FCDIIb) is characterised by a population of unique abnormally enlarged cells known as balloon cells (BCs). The understanding of the molecular abnormalities underlying FCDIIb is poor. It is unclear if BCs are the key pathological cell or if there are other types of cells that are important in the pathogenesis of the disease. Interactions between such cell types are also unknown. Methods: Bioinformatic analyses of gene expression data were used to identify networks of genes that could characterise FCD and to identify putative diagnostic biomarkers. These markers were validated at the protein level by immunohistochemistry. Furthermore, various techniques allowed for the investigation of possible signalling between different cell types in FCDIIb. Results: Several of the gene networks and biomarkers were differentially expressed between FCDIIb and control. Analysis of one particular network showed that some components were expressed in BCs but others were expressed in a potentially novel cell population that was only present in FCDIIb. The importance of cellular heterogeneity of FCDIIb was further supported by the finding that BCs may have a unique senescence-associated secretome that could be involved in cell-to-cell communications. Conclusion: A genomic characterisation of FCD has provided a better understanding of the cellular abnormalities in FCDIIb from a molecular perspective. In addition to the identification of several differentially expressed gene networks, the results also provide an interesting insight into cell-to-cell communications between different cell types in FCDIIb, which could be important in driving the pathogenesis of the disease.
Article
Speckle-type pox virus and zinc finger (POZ) protein (SPOP), a substrate recognition receptor for the cullin-3/RING ubiquitin E3 complex, leads to the ubiquitination of >40 of its target substrates. Since a variety of point mutations in the substrate-binding domain of SPOP have been identified in cancers, including prostate and endometrial cancers, the pathological roles of those cancer-associated SPOP mutants have been extensively elucidated. In this study, we evaluated the cellular functions of wild-type SPOP in non-cancerous human keratinocyte-derived HaCaT cells expressing wild-type SPOP gene. SPOP knockdown using siRNA in HaCaT cells dramatically reduced cell growth and arrested their cell cycles at G1/S phase. The expression of DNA replication licensing factors CDT1 and CDC6 in HaCaT cells drastically decreased on SPOP knockdown as their translation was inhibited. CDT1 and CDC6 downregulation induced p21 expression without p53 activation. Our results suggest that SPOP is essential for DNA replication licensing in non-cancerous keratinocyte HaCaT cells.
Chapter
DNA replication, the process of copying one double stranded DNA molecule to form two identical copies, is highly conserved at the mechanistic level across evolution. Interesting in its own right as a fascinating feat of biochemical regulation and coordination, DNA replication is at the heart of modern advances in molecular biology. An understanding of the process at both the biological and chemical level is essential to developing new techniques in molecular biology. Insights into the process at the molecular level provide opportunities to modulate and intervene in replication. Rapidly dividing cells need to replicate their DNA prior to division, and targeting components of the replication process is a potentially powerful strategy in cancer treatment. Conversely, ageing may be associated with loss of replication activity and restoring it to cells may moderate some of the diseases associated with old age. Replication is, therefore, fundamental to a huge range of molecular biological and biochemical applications, and provides many potential targets for drug design. The fast pace of replication research, particularly in providing new structural insights, has outdated the majority of available texts. This learned, yet accessible, book contains the latest research written by those conducting it. It examines conserved themes providing a biological background for biochemical, chemical and pharmaceutical studies of this huge and exciting field. Rather than simply "itemising" the replication steps and the proteins involved, replication is tackled from a novel perspective. The book provides logical groupings of processes based upon biochemical similarities. The emphasis on mechanisms and the relationship between structure and function targets the chapters towards biochemists and biological chemists as well as molecular and cell biologists. The book highlights new insights into the replication process, from the assembly of pre-replication complexes, through polymerisation mechanisms, to considering replication in the context of chromatin and chromosomes. It also covers mitochondrial DNA replication, and includes archaeal paradigms, which are proving increasingly relevant to the study of replication in higher eukaryotes. Exciting potential drug targets in DNA replication are discussed, particularly in the context of treating malaria and cancer.
Article
Full-text available
There is no direct evidence supporting that SDS is a carcinogen, so to investigate this fact, we used HaCaT keratinocytes as a model of human epidermal cells. To reveal the candidate proteins and/or pathways characterizing the SDS impact on HaCaT, we proposed comparative proteoinformatics pipeline. For protein extraction, the performance of two sample preparation protocols was assessed: 0.2% SDS-based solubilization combined with the 1DE-gel concentration (Protocol 1) and osmotic shock (Protocol 2). As a result, in SDS-exposed HaCaT cells, Protocol 1 revealed 54 differentially expressed proteins (DEPs) involved in the disease of cellular proliferation (DOID:14566), whereas Protocol 2 found 45 DEPs of the same disease ID. The ‘skin cancer’ term was a single significant COSMIC term for Protocol 1 DEPs, including those involved in double-strand break repair pathway (BIR, GO:0000727). Considerable upregulation of BIR-associated proteins MCM3, MCM6, and MCM7 was detected. The eightfold increase in MCM6 level was verified by reverse transcription qPCR. Thus, Protocol 1 demonstrated high effectiveness in terms of the total number and sensitivity of MS identifications in HaCaT cell line proteomic analysis. The utility of Protocol 1 was confirmed by the revealed upregulation of cancer-associated MCM6 in HaCaT keratinocytes induced by non-toxic concentration of SDS. Data are available via ProteomeXchange with identifier PXD035202.
Article
Three-dimensional (3D) organoids are a novel tool to model epithelial cell biology and human diseases of the esophagus. 3D organoid culture systems have been utilized to investigate the pathobiology of esophageal cancer, including both squamous cell carcinoma and adenocarcinoma. Additional organoid-based approaches for study of esophageal development and benign esophageal diseases have provided key insights into esophageal keratinocyte differentiation and mucosal regeneration. These investigations have implications for the identification of esophageal cancer stem cells, as well as the potential to halt malignant progression through induction of differentiation pathways. Patient-derived organoids (PDOs) from human tissue samples allow for unique and faithful in vitro modeling of esophageal cancers, and provide an exciting platform for investigation into personalized medicine and targeted treatment approaches, as well as new models for understanding therapy resistance and recurrent disease. Future directions include high-throughput genomic screening using PDOs, and study of tumor-microenvironmental interactions through co-culture with immune and stromal cells and novel extracellular matrix complexes.
Article
Dermal fibroblast is one of the major constitutive cells of skin and plays a central role in skin senescence. The replicative senescence of fibroblasts may cause skin aging, bad wound healing, skin diseases and even cancer. In this study, a label-free quantitative proteomic approach was employed to analyzing the serial passaged human skin fibroblast (CCD-1079Sk) cells, resulting in 3371 proteins identified. Of which, 280 proteins were significantly changed in early passage (6 passages, P6), middle passage (12 passages, P12) and late passage (21 passages, P21), with a time-dependent decrease or increase tendency. Bioinformatic analysis demonstrated that the chromosome condensin complex, including structural maintenance of chromosomes protein 2 (SMC2) and structural maintenance of chromosomes protein 4 (SMC4), were down-regulated in the serially passaged fibroblast cells. The qRT-PCR and Western Blot experiments confirmed that the expression of these two proteins were significantly down-regulated in a time-dependent manner in the subculture of human skin fibroblasts (HSFb cells). In summary, we used serially passaged human skin fibroblast cells coupled with quantitative proteomic approach to profile the protein expression pattern in the temporal progress of replicative senescence in HSFb cells and revealed that the down-regulation of the chromosome condensin complex subunits, such as SMC2 and SMC4, may play an important role in the fibroblast senescence.
Article
Full-text available
Epidermal growth factor receptor (EGFR) overexpression is observed in a number of malignancies, especially those of esophageal squamous cell origin. However, little is known about the biological functions of EGFR in primary esophageal squamous epithelial cells. Using newly established primary human esophageal squamous epithelial cells as a platform, we overexpressed EGFR through retroviral transduction and established novel three-dimensional organotypic cultures. Additionally, EGFR was targeted in a cell type- and tissue-specific fashion to the esophageal epithelium in transgenic mice. EGFR overexpression in primary esophageal keratinocytes resulted in the biochemical activation of Akt and STAT pathways and induced enhanced cell migration and cell aggregation. When established in organotypic culture, EGFR-overexpressing cells had evidence of epithelial cell hyperproliferation and hyperplasia. These effects were also observed in EGFR-overexpressing transgenic mice and the esophageal cell lines established thereof. In particular, EGFR-induced effects upon aggregation appear to be mediated through the relocalization of p120 from the cytoplasm to the membrane and increased interaction with E-cadherin. EGFR modulates cell migration through the up-regulation of matrix metalloproteinase 1. Taken together, the functional effects of EGFR overexpression help to explain its role in the initiating steps of esophageal squamous carcinogenesis.
Article
Full-text available
The ExPASy (the Expert Protein Analysis System) World Wide Web server (http://www.expasy.org), is provided as a service to the life science community by a multidisciplinary team at the Swiss Institute of Bioinformatics (SIB). It provides access to a variety of databases and analytical tools dedicated to proteins and proteomics. ExPASy databases include SWISS-PROT and TrEMBL, SWISS-2DPAGE, PROSITE, ENZYME and the SWISS-MODEL repository. Analysis tools are available for specific tasks relevant to proteomics, similarity searches, pattern and profile searches, post-translational modification prediction, topology prediction, primary, secondary and tertiary structure analysis and sequence alignment. These databases and tools are tightly interlinked: a special emphasis is placed on integration of database entries with related resources developed at the SIB and elsewhere, and the proteomics tools have been designed to read the annotations in SWISS-PROT in order to enhance their predictions. ExPASy started to operate in 1993, as the first WWW server in the field of life sciences. In addition to the main site in Switzerland, seven mirror sites in different continents currently serve the user community.
Article
Full-text available
Normal human somatic cells have a finite life span and undergo replicative senescence after a limited number of cell divisions. Erosion of telomeric DNA has emerged as a key factor in senescence, which is antagonized during cell immortalization and transformation. To clarify the involvement of telomerase in the immortalization of keratinocytes, catalytic subunit of telomerase (hTERT) expression was restored in normal human esophageal epithelial cells (EPC2). EPC2-hTERT cells overcame senescence and were immortalized without p16INK4a genetic or epigenetic alterations. p16INK4a was expressed at moderate levels and remained functional as evidenced by induction with UV treatment and binding to cyclin-dependent kinase 4 and 6. There were no mutations in the p53 gene, and p53 was functionally intact. Importantly, senescence could be activated in the immortalized EPC2-hTERT cells by overexpression of oncogenic H-ras or p16INK4a. Furthermore, the EPC2-hTERT cells yielded basal cell hyperplasia in an innovative organotypic culture system in contrast to a normal epithelium from parental cells. These comprehensive results indicate that the expression of telomerase induces immortalization of normal human esophageal keratinocytes without inactivation of p16INK4a/pRb pathway or abrogation of the p53 pathway.
Article
Full-text available
Oncogenic Ras induces premature senescence in primary cells. Such an oncogene-induced senescence involves activation of tumor suppressor genes that provide a checkpoint mechanism against malignant transformation. In mouse, the ARF-p53 pathway mediates Ha-Ras(G12V)-induced senescence, and p19(ARF-/-) and p53(-/-) cells undergo transformation upon Ras activation. In addition, mouse cells, unlike human cells, express constitutively active telomerase and have long telomeres. However, it is unclear how Ras activation affects human cells of epithelial origin with p53 mutation and/or telomerase activation. In order to address this question, Ha-Ras(G12V) was expressed ectopically in primary as well as hTERT-immortalized human esophageal keratinocytes stably expressing dominant-negative p53 mutants. In human esophageal keratinocytes, we found that Ha-Ras(G12V) induced senescence regardless of p53 status and telomerase activation. Ras activation resulted in changes of cellular morphology, activation of senescence-associated beta-galactosidase, and suppression of cell proliferation, all coupled with reduction in the hyperphosphorylated form of the retinoblastoma protein (pRb). Furthermore, Ha-Ras(G12V) upregulated p16(INK4a) and downregulated cyclin-dependent kinase Cdk4 in human esophageal keratinocytes. Thus, Ras-mediated senescence may involve distinct mechanisms between human and mouse cells. Inactivation of the pRb pathway may be necessary for Ras to overcome senescence and transform human esophageal epithelial cells.
Article
We report the development of an advanced system for transfer and expression of exogenous genes in mammalian cells based on Moloney murine leukemia virus (Mo MuLV). Extensive deletion/mutagenesis analysis to identify cis-acting signals involved in virus transmission has led to the design of a family of novel, highly efficient retroviral vectors and a partner helper-free packaging cell line. The pBabe retroviral vector constructs transmit inserted genes at high titres and express them from the Mo MuLV Long Terminal Repeat (LTR). Each of these vectors has been constructed with one of four different dominantly acting selectable markers, allowing the growth of infected mammalian cells in the presence of G418, hygromycin B, bleomycin/phleomycin or puromycin, respectively. The high titre ecotropic helper free packaging cell line, omega E, was designed in conjunction with the pBabe vectors to reduce the risk of generation of wild type Mo MuLV via homologous recombination events. The omega E cell line was generated with separate gagpol and ecotropic env expression constructs with minimal sequence overlap and decreased sequence homology achieved by 'codon wobbling'. Homologous env coding sequences were deleted from the pBabe vectors without diminishing recombinant vector titre. Together, the pBabe vectors and omega E cell line should prove useful in experiments where highest frequencies of gene transfer, or concomitant expression of several different genes within a single cell are required with minimal risk of helper virus contamination.
Article
Cleavage of specific protein subsets is a key event in the execution of apoptosis. Protein degradation may serve for the structural alterations that result in cell self-destruction, but it may also function as a switch in the decisions between apoptosis and necrosis or apoptosis and cell proliferation. Here, we show that MCM3, but not other members of the Mcm family of replicative proteins, is cleaved early in several models of apoptosis. Cleavage of MCM3 can be prevented by caspase inhibitors, and it does not occur when cells are forced to undergo necrosis by energy deprivation. We propose that active destruction of MCM3 inactivates the Mcm complex and serves to prevent untimely DNA replication events during the execution of the cell death program.
Article
Dysplasia, an intermediate stage in the progression from normal tissue to neoplasia, is defined morphologically by a loss of normal orientation between epithelial cells, with changes in cellular and nuclear shape and size. However, little is known about the functional properties of dysplastic cells, including their replicative state, largely due to a lack of available biological markers. We have used novel antibodies against minichromosome maintenance (MCM) proteins to examine the proliferative status of a range of histological lesions and to characterize dysplastic cells in functional terms. Immunoperoxidase staining was used to localize the MCM proteins, components of the prereplicative complex that is essential for initiating eukaryotic DNA replication. These proteins are down-regulated in cells undergoing differentiation or quiescence and, thus, serve as specific markers for proliferating cells. In normal and some reactive tissues, MCM expression was present only in restricted proliferative compartments, consistent with our published findings in the uterine cervix. In dysplastic and malignant tissues, in contrast, MCM proteins were expressed in the majority of cells, extending to surface layers of dysplastic stratified epithelia. In carcinomas, the frequency of expression of MCM proteins showed an inverse correlation with the degree of tumor differentiation. Thus, we suggest that dysplastic cells may be characterized in functional terms as remaining in cell cycle, due to deregulation of normal controls over cell proliferation. Antibodies against MCM proteins have potential clinical applications, for example, in the assessment of tumor prognosis in histological sections and the identification of proliferating cells in clinical samples using biochemical or cytological assays.
Article
The convergence point of growth regulatory pathways that control cell proliferation is the initiation of genome replication, the core of which is the assembly of pre-replicative complexes resulting in chromatin being "licensed" for DNA replication in the subsequent S phase. We have analysed regulation of the pre-replicative complex proteins ORC, Cdc6, and MCM in cycling and non-proliferating quiescent, differentiated and replicative senescent human cells. Moreover, a human cell-free DNA replication system has been exploited to study the replicative capacity of nuclei and cytosolic extracts prepared from these cells. These studies demonstrate that downregulation of the Cdc6 and MCM constituents of the replication initiation pathway is a common downstream mechanism for loss of proliferative capacity in human cells. Furthermore, analysis of MCM protein expression in self-renewing, stable and permanent human tissues shows that the three classes of tissue have developed very different growth control strategies with respect to replication licensing. Notably, in breast tissue we found striking differences between the proportion of mammary acinar cells that express MCM proteins and those labelled with conventional proliferation markers, raising the intriguing possibility that progenitor cells of some tissues are held in a prolonged G1 phase or "in-cycle arrest". We conclude that biomarkers for replication-licensed cells detect, in addition to actively proliferating cells, cells with growth potential, a concept that has major implications for developmental and cancer biology.