ArticlePDF Available

A microRNA polycistron as a potential human oncogene

Authors:

Abstract and Figures

To date, more than 200 microRNAs have been described in humans; however, the precise functions of these regulatory, non-coding RNAs remains largely obscure. One cluster of microRNAs, the mir-17-92 polycistron, is located in a region of DNA that is amplified in human B-cell lymphomas. Here we compared B-cell lymphoma samples and cell lines to normal tissues, and found that the levels of the primary or mature microRNAs derived from the mir-17-92 locus are often substantially increased in these cancers. Enforced expression of the mir-17-92 cluster acted with c-myc expression to accelerate tumour development in a mouse B-cell lymphoma model. Tumours derived from haematopoietic stem cells expressing a subset of the mir-17-92 cluster and c-myc could be distinguished by an absence of apoptosis that was otherwise prevalent in c-myc-induced lymphomas. Together, these studies indicate that non-coding RNAs, specifically microRNAs, can modulate tumour formation, and implicate the mir-17-92 cluster as a potential human oncogene.
Content may be subject to copyright.
A microRNA polycistron as a potential human
oncogene
Lin He
1
*, J. Michael Thomson
2
*, Michael T. Hemann
1
, Eva Hernando-Monge
4
, David Mu
1
, Summer Goodson
2
,
Scott Powers
1
, Carlos Cordon-Cardo
4
, Scott W. Lowe
1
, Gregory J. Hannon
1
& Scott M. Hammond
2,3
To date, more than 200 microRNAs have been described in
humans; however, the precise functions of these regulatory, non-
coding RNAs remains largely obscure. One cluster of microRNAs,
the mir-1792 polycistron, is located in a region of DNA that is
amplified in human B-cell lymphomas
1
. Here we compared B-cell
lymphoma samples and cell lines to normal tissues, and found that
the levels of the primary or mature microRNAs derived from the
mir-1792 locus are often substantially increased in these cancers.
Enforced expression of the mir-1792 cluster acted with c-myc
expression to accelerate tumour development in a mouse B-cell
lymphoma model. Tumours derived from haematopoietic stem
cells expressing a subset of the mir-1792 cluster and c-myc could
be distinguished by an absence of apoptosis that was otherwise
prevalent in c-myc-induced lymphomas. Together, these studies
indicate that non-coding RNAs, specifically microRNAs, can
modulate tumour formation, and implicate the mir-1792 cluster
as a potential human oncogene.
MicroRNAs (miRNAs) have emerged relatively recently as a new
class of small, non-coding RNAs that regulate gene expression.
Nascent primary miRNA transcripts (pri-miRNAs) are processed
sequentially by two RNase III enzymes, Drosha and Dicer
2,3
, to yield
mature miRNAs, ranging from 18 to 24 nucleotides (nt) in length.
miRNAs are incorporated into the RNA interference (RNAi) effector
complex, RISC, and target specific messenger RNAs for translational
repression or mRNA cleavage
4–6
. Although hundreds of miRNAs
have been cloned and/or predicted, only a handful have been
functionally characterized. For example, lin-4 and let-7 regulate the
timing of larval development in C. elegans
7,8
. Left/right asymmetric
gene expression in C. elegans chemosensory neurons is controlled by
lsy-6 and miR-273 (refs 9, 10). Bantam stimulates cell growth and
prevents apoptosis in Drosophila
11
, and miR-181 potentiates B-cell
differentiation in mammals
12
. These findings, in combination with
computational target predictions, are consistent with miRNAs regu-
lating a broad spectrum of physiological and developmental
processes.
Microarray-based expression studies have indicated specific
alterations in human miRNA expression profiles that correlate with
particular tumour phenotypes (J.M.T. and S.M.H., unpublished
data). Among those that show altered expression, the mir-1792
cistron is located at 13q31, a genomic locus that is amplified in cases
of diffuse large B-cell lymphoma, follicular lymphoma, mantle cell
lymphoma, primary cutaneous B-cell lymphoma and several other
tumour types
1,13
. There are only two annotated genes in the epicentre
of this amplicon, c13orf25 and GPC5. Previous studies have shown
that c13orf25 is the only one of the two genes for which increased
expression correlates with the presence of the amplicon
1
. Therefore,
c13orf25 had been implicated as a target of the 13q31 amplicon
1
.Itis
unlikely that c13orf25 actually encodes a protein, as predicted open
reading frames (ORFs) encode only short peptides (,70 amino
acids), which are not conserved in closely related species. Instead, the
c13orf25 transcript appears to be the functional precursor of a series
of seven microRNAs: miR-17-5p, miR-17-3p, miR-18, miR-19a,
miR-20, miR-19b-1 and miR-92-1 (Fig. 1a). Additionally, this cluster
is related to the homologous mir-106a92 cluster on chromosome X
and the mir-106b25 cluster on chromosome 7 (ref. 14; Fig. 1a).
Alignment of the human c13orf25 locus and its murine orthologue
revealed extensive sequence conservation only within the mir-1792
polycistron and its immediate flanking sequence. Several of the
expressed-sequence-tags (ESTs) derived from c13orf25 and its
mouse orthologue terminate at the 3 0end of mir-1792 cluster,
consistent with the presence of a Drosha processing site at this
location (Fig. 1b).
A principal consequence of 13q31-q32 amplification could be
an increase in the abundance of mature miRNA species from the
mir-1792 cluster. We acquired four cell lines previously described as
carrying amplifications in the 13q31-q32 region
1
and confirmed the
gene dosage increase at the c13orf25 locus in three of those cell lines
using quantitative polymerase chain reaction (PCR) analysis.
The abundance of 191 mature miRNAs was assessed in these four
cell lines and compared to normal B-cells, and to five leukaemia and
lymphoma cell lines lacking the amplicon (Fig. 1c and Supplemen-
tary Fig. 1). Using a significance analysis of microarrays (SAM)
analysis
15
, we identified six miRNAs for which high-level expression
correlated with increased gene dosage of c13orf25 (see Supple-
mentary Table 1). Five were from the mir-1792 cistron, and
the sixth, miR-106a, was identified as a probable result of cross-
hybridization to miR-17-5p, from which it differs at only two
terminal nucleotides (Fig. 1c). This hypothesis is supported by the
observation that the mir-106a92 locus does not show copy number
alterations in these cell lines (not shown). In each cell line, expression
levels correlated with the copy number of the mir-1792 locus (Fig.
1c, lower panel).
We also examined the expression of pri-mir-1792 in a series of
human tumour samples comprising both lymphomas and colorectal
carcinomas. Of 46 lymphoma samples, including 13 diffuse large
B-cell lymphomas and 6 follicular lymphomas, we saw significant
(greater than fivefold) overexpression of pri-mir-1792 in 65% of the
samples. Considering all of the B-cell lymphoma samples analysed,
the average increase in pri-miRNA expression was about tenfold
(Fig. 1d). In contrast, colorectal carcinomas rarely showed over-
expression of the pri-miRNA. Increases in expression from this locus
were less common (15% of samples showed greater than fivefold
LETTERS
1
Cold Spring Harbor Laboratory, Watson School of Biological Sciences, 1 Bungtown Road, Cold Spring Harbor, New York 11724, USA.
2
Department of Cell and Developmental
Biology and
3
Lineberger Comprehensive Cancer Center, University of North Carolina, Chapel Hill, North Carolina 27599, USA.
4
Memorial Sloan-Kettering Cancer Center, Division
of Molecular Pathology, Memorial Sloan-Kettering Cancer Center, 1275 York Ave, New York, New York 10021, USA.
*These authors contributed equally to this work.
Vol 435|9 June 2005|doi:10.1038/nature03552
828 © 2005 Nature Publishing Group
Figure 1 |The mir-1792 cluster shows increased expression in B-cell
lymphoma samples and cell lines. a, Genomic organization of three
polycistronic miRNA clusters is shown. There are five paralogous groups
located in three homologous clusters (mir-17–92,mir-106a–92 and
mir-106b–25) with a conserved order: miR-17-5p/miR-106a/miR-106b;
miR-18;miR-19a/miR-19b-1/miR-19b-2;miR-20/miR-93;andmiR-92-1/
miR-92-2/miR-25 (yellow boxes, pre-miRNAs; purple boxes, mature
miRNAs). b, The level of conservation between human and mouse
homologues is represented using an mVista plot
28
. Two alternative isoforms
have been detected for the human gene, and these are shown schematically
1
(dark blue, exons; light blue, introns; orange, the mir-1792 cluster).
c, MicroRNA expression levels in cell lines carrying the 13q31-q32 amplicon
(including Karpas 1718, OCI-Ly4 and OCI-Ly7) were compared to those in
leukaemia and lymphoma cell lines lacking this genetic lesion, and to normal
B-cells isolated from cortical blood (top panel). We included in this analysis
the OCI-Ly8 cell line, which has previously been identified as a cell line
carrying the 13q31-32 amplicon, but showed no gene dosage increase at the
c13orf25 locus in our study. Normalized one-channel measurements for 191
human miRNAs were hierarchically clustered for all miRNA genes
represented on the array. An excerpt of the data is shown, and the full cluster
analysis is presented in Supplementary Fig. 1. The expression map node that
correlates with the amplification is shown. The let-7 cluster node is also
shown for comparison (middle panel). In the cell lines examined, expression
levels of the mature microRNAs from the mir-1792 polycistron correlate
with the copy number at the mir-1792 locus (bottom panel). d, The level of
mir-1792 pri-miRNA was determined by real-time quantitative PCR in 46
lymphomas and 47 colorectal carcinomas, and compared to levels found in
corresponding normal tissues from five individuals. In cand d, error bars
indicate standard deviation (s.d.).
NATURE|Vol 435|9 June 2005 LETTERS
829
© 2005 Nature Publishing Group
upregulation), and the degree of overexpression was substantially
lower (Fig. 1d).
Considered together, our data prompted the hypothesis that mir-
1792 might contribute to tumour development. To test this idea
directly, we used a mouse model of human B-cell lymphoma.
Transgenic animals carrying a c-myc oncogene, driven by the
immunoglobulin heavy-chain enhancer (Em), develop B-cell lym-
phomas by 4–6 months of age
16
. Similarly, haematopoietic stem cells
(HSCs) derived from fetal livers of Em-myc transgenic mice generate
B-cell lymphomas with comparable latency when transplanted into
lethally irradiated recipients
17–20
(Fig. 2a). We therefore infected
Em-myc/þHSCs with a murine stem cell virus (MSCV) retrovirus
that directs expression of a truncated cluster comprising mir-1719b-1
(hereafter mir-1719b), the vertebrate-specific portion of the
mir-1792 miRNA cistron (Fig. 1a). This virus also contained a
green fluorescent protein (GFP) transgene, allowing us to follow
infected stem cells in vitro and in vivo (Fig. 2a). Mice reconstituted
with Em-myc/þHSCs carrying a control MSCV vector developed
lymphomas after the expected latency (3–6 months), with incomplete
penetrance (Fig. 2b). Similarly, we examined .40 animals reconsti-
tuted with Em-myc/þHSCs expressing subsets of 96 different, single
microRNAs (see Supplementary Table 2). Although we did not
confirm miRNA overexpression for every case, we did not observe
any significantly accelerated onset of disease. In contrast, 100% of
the animals co-expressing the mir-1719b polycistron and c-myc
developed leukaemias at an average of 51 days following transplan-
tation (s.d. ^13 days, P,0.0001 compared with MSCV controls
using the logrank test), and died of B-cell lymphomas at an average age
of 65 days (s.d. ^13 days, P,0.0001 compared to MSCV controls;
Fig. 2b). In all but one case, primary lymphomas could be
visualized by virtue of the linked GFP marker (Fig. 2c and Table
1). The mature miRNAs from the mir-1719b cluster show high-
level expression in these tumours, compared with miRNAs from the
paralogous mir-106a92 locus, and also have similar mir-1719b
expression levels compared to the Karpas 1718 lymphoma cell line,
which has increased c13orf25 gene dosage (see Supplementary
Fig. 2).
The full mir-1792 cistron was also tested in a small cohort of
animals. Although similar results were obtained compared to those
animals reconstituted with HSCs expressing mir-1719b, studies in
cell lines indicated that the construct used to express the entire cluster
gave lower levels of mature miRNAs. We therefore focused most of
our study on the truncated mir-1719b cluster. In these ongoing
studies, we have yet to find any individual miRNA from the
Figure 2 |Overexpression of the mir-1719b cluster accelerates
c-myc-induced lymphomagenesis in mice. a, Schematic representation of
the adoptive transfer protocol using Em-myc HSCs. b, Mice reconstituted
with HSCs expressing mir-1719b in an MSCV retroviral vector (MSCV
mir-1719b) or infected with a control MSCV virus were monitored by
blood smear analysis starting 5 weeks after transplantation. The Kaplan-
Meier curves represent the percentage of leukaemia-free survival or overall
survival. c, External GFP imaging of tumour-bearing mice with Em-myc/mir-
1719b or Em-myc/MSCV shows the overall distribution of tumour cells.
Em-myc/mir-1719b tumours show a more disseminated phenotype
compared with control tumours. These animals are representative of their
genotype.
LETTERS NATURE|Vol 435|9 June 2005
830 © 2005 Nature Publishing Group
mir-1719b cluster that can accelerate tumour formation to the
extent seen with the intact polycistron (not shown).
The Em-myc/mir-1719b lymphomas are true malignancies rather
than hyperplasias, because primary tumour cells, when transplanted
into C57B6/J recipients, induce B-cell lymphomas in 2–3 weeks
that result in lethality after 4–5 weeks (data not shown). The
secondary tumours show pathological features indistinguishable
from the original tumours, and retain tumorigenic potential after
two additional rounds of serial transplantation (data not shown).
Therefore, a microRNA cluster can accelerate Em-myc induced
tumorigenesis in mice.
The pathological hallmarks of Em-myc/mir-1719b mosaic animals
included massive enlargement of lymph nodes, splenic hyperplasia,
infiltration of the thymus by lymphoma cells, and leukaemia (Fig. 2c).
Animals with advanced lymphomas showed extramedullary haema-
topoiesis due to functional failure of the bone marrow. Furthermore,
6 out of 14 animals showed hind limb paralysis, associated with
substantial tumour growth at the lumbar node. Tumours resulting
from combined c-myc and mir-1719b expression consistently
invaded visceral organs outside the lymphoid compartment, includ-
ing liver, lung and occasionally kidney (Figs 2c, 3b and Table 1).
Additionally, Em-myc/mir-1719b lymphomas show a high mitotic
index without extensive apoptosis (Fig. 3a). This contrasts with the
Em-myc/MSCV tumours lacking the microRNA cluster, which show a
high degree of apoptosis (Fig. 3a). These findings indicate that
cooperation between Em-myc and mir-1719b gives rise to highly
malignant, disseminated lymphomas capable of evading normal
apoptotic responses to inappropriate proliferation.
Em-myc-induced lymphomas originate from the B-lymphoid line-
age. However, the developmental characteristics of these tumour cells
are not stage-specific, as they can resemble either mature B cells or
pre-B cells. To examine the cell lineage of the Em-myc/mir-1719b
lymphomas, we assessed the expression of cell-surface markers,
including the B-cell-specific marker B220 and the T-cell-specific
markers CD4 and CD8a. All tumours were found to be of B-cell
origin, staining positive for B220 and negative for both CD4 and
CD8a (Fig. 3c and Table 1). We next analysed these tumours for
CD19 and IgM expression to distinguish pre-B from mature B cells.
With one exception, Em-myc/mir-1719b lymphomas were derived
purely from the pre-B cell lineage (low to absent Thy1.2 staining,
CD19
þ
B220
þ
IgM
2
) (Table 1), suggesting that overexpression of
mir-1719b strongly favours transformation of B-cell progenitors
under our experimental conditions.
Exactly how overproduction of mir-1719b might promote onco-
genesis is unclear. At a minimum, studies of tumour pathology
suggest that increased expression of this cluster mitigates the pro-
apoptotic response to elevated myc expression in vivo. Furthermore,
we have previously shown that this miRNA cistron is highly
expressed in embryonic stem cells, with expression levels decreasing
during embryonic development in mice
21
. It is therefore tempting to
speculate that these miRNAs promote ‘stem cell’ properties or specify
characteristics of early developmental lineages. A detailed, mechan-
istic understanding of how this non-coding RNA cluster acts as an
oncogene is at present hampered by the lack of a validated biochemi-
cal strategy for identifying miRNA targets.
Previous circumstantial evidence has indicated the potential
involvement of a number of miRNAs in tumorigenesis. Although
miRNAs only represent 1% of the mammalian genome, more than
50% of miRNA genes are located within regions associated with
amplification, deletion and translocation in cancer
22
. Expression
studies of various tumour types have also revealed specific alterations
in miRNA profiles
22–25
. For example, mir-15 and mir-16 are fre-
quently deleted and/or downregulated in B-cell chronic lymphocytic
leukaemia
26
, miR-143 and miR-145 show decreased abundance in
colorectal neoplasia
25
, and miR-155 and its non-coding RNA host
gene, BIC, are upregulated 100-fold in Burkitt’s lymphoma
patients
24
. Here, we have shown that one miRNA polycistron is not
only the subject of tumour-specific amplification, but that it is also
overexpressed in tumours and tumour cell lines, and can act as an
oncogene in vivo. Our results indicate that non-coding RNAs might
act as integral parts of the molecular architecture of oncogene and
tumour suppressor networks. Such oncogenic microRNAs might be
designated ‘oncomiRs, with mir-1792 being oncomiR-1.
METHODS
miRNA expression profiling. Five micrograms of total RNA was labelled with
RNA ligase and a Cy3-conjugated dinucleotide, and hybridized to custom
oligonucleotide microarrays as described in ref. 21. Cy3 median intensity values
Table 1 |Phenotypic analysis of a subset of Em-myc/mir-1719b tumours
Animal GFP Immunophenotyping Cell type Ki67 staining Apoptosis* Pathological features
1þB220
þ
, Thy1.2
2
, IgM
2
,
CD19
þ
, CD4
2
, CD8
2
Pre-B cell þþ(70–80%) Low Tumour cells invaded liver and spleen;
mild infiltrations observed in lung
and kidney; spleen enlarged; hindlimb
paralysis
2þB220
þ
, Thy1.2
low
, IgM
2
,
CD19
þ
, CD4
2
, CD8
2
Pre-B cell þþ(80–90%) Low Tumour cells invaded liver, lung and
spleen; spleen enlarged
3þB220
þ
, Thy1.2
2
, IgM
2
,
CD19
þ
, CD4
2
, CD8
2
Pre-B cell þþ(80–90%) Low Tumour cells invaded liver, lung and
spleen; spleen enlarged
4þB220
þ
, Thy1.2
low
, IgM
2
,
CD19
þ
, CD4
2
, CD8
2
Pre-B cell þþ(70–80%) Low Tumour cells invaded liver and spleen;
spleen enlarged
5þB220
þ
, Thy1,2
2
, IgM
2
,
CD19
þ
, CD4
2
, CD8
2
Pre-B cell þþ(80–90%) Slightly less
than control
Highly disseminated lymphoma; tumour
cells invaded liver, spleen, lung and
kidney; spleen enlarged
6þB220
þ
, Thy1,2
2
, IgM
2
,
CD19
þ
, CD4
2
, CD8
2
Pre-B cell þþ(80–90%) Low Highly disseminated lymphoma; tumour
cells invaded liver, spleen, lung and
kidney; spleen enlarged
7þB220
þ
, Thy1.2
2
, IgM
2
,
CD19
þ
, CD4
2
, CD8
2
Pre-B cell þþ(70–80%) Low Spleen enlarged; mild infiltration of
tumour cells into liver only
8þB220
þ
, Thy1.2
low
, IgM
2
,
CD19
þ
, CD4
2
, CD8
2
and
B220
þ
, Thy1.2
low
, IgM
2
,
CD19
þ
, CD4
2
, CD8
2
Pre-B cell and
mature B cell
þþ(70–80%) Low Highly disseminated lymphoma; tumour
cells invaded liver, spleen, lung and
kidney; enlarged spleen
92ND ND þþ(80–90%) Low Tumour cells invaded liver, lung and
spleen; enlarged spleen
*Levels of apoptosis in Em-myc/mir-1719b lymphomas are compared to control Em-myc/MSCV lymphomas on the basis of haemotoxylin and eosin staining, and TUNEL staining.
†The two clones of tumour cells with different cell type specificity might reflect independent transformation events or maturation of a single primary clone.
ND, not determined.
NATURE|Vol 435|9 June 2005 LETTERS
831
© 2005 Nature Publishing Group
were filtered to remove data points that did not exceed background levels by
twofold. Values were log
2
-transformed and median-centred by array. Clustering
was performed with Cluster (Stanford University), using values that were
median-centred by gene. Dendrograms and expression maps were generated
using Treeview (Stanford).
Cell lines. The measurement of miRNA abundance was carried out using the
following human cell lines: Karpas 1718 (derived from splenic lymphoma with
villous lymphocytes, provided by A. Karpas), OCI-Ly4, OCI-Ly7 and OCI-Ly8
(derived from diffuse large B-cell lymphoma, provided by R. Dalla-Favera). The
cell lines lacking the 13q31-q32 amplicon were Raji (B-cell, derived from
Burkitt’s lymphoma, ATCC); Namalwa (B-cell, derived from Burkitt’s lym-
phoma, ATCC); HG 1125 (EBV-transformed human lymphoblastoid, provided
by B. Stillman); Manca (lymphoblast-like, derived from chronic myelogenous
leukaemia); Jurkat; proliferating B-cells (spleenic B-cells isolated from a C57B6/
Ly5.2 mouse and stimulated to proliferate in culture with lipopolysaccharide,
provided by I. Weissman); and normal B cells (derived from cord blood,
Cambrex).
PCR and copy number analysis. Tumour samples were obtained from the
Cooperative Human Tissue Network (http://www-chtn.ims.nci.nih.gov). Cor-
responding normal tissue RNA from five individuals was purchased from
Biochain Institute Inc. For fluorogenic real-time PCR, primers that amplify
mir-1792 pri-miRNA and b-actin mRNA (control), and probes were designed
using Primer Express software (V.2): mir-1792 forward primer, 30-CAGTAAA
GGTAAGGAGAGCTCAATCTG-50; reverse primer, 30-CATACAACCACTAA
GCTAAAGAATAATCTGA-50;mir-1792 probe, (6-FAM)-TGGAAATAAGATC
ATCATGCCCACTTGAGAC-(TAMRA); b-actin forward primer, 30-GCAAAG
ACCTGTACGCCAACA-50; reverse primer, 30-TGCATCCTGTCGGCAATG-50;
b-actin probe, (6-FAM)-TGGCGGCACCACCATGTACC-(TAMRA). The ratios
of RNA species detected by mir-1792 primers and b-actin primers in each RNA
sample were determined in triplicate by reverse-transcription, quantitative PCR
Figure 3 |Pathological and immunological analysis of lymphomas produced
by cooperation between mir-1719b and c-myc.a, Haemotoxylin and eosin
(H&E), Ki67, B220 and TUNEL staining of Em-myc/mir-1719b lymph node
tumours. The ‘starry sky’ morphology is a hallmark of cell clusters
undergoing apoptosis (black arrows). Scale bar, 10
m
m. b, Invasion of
visceral organs (liver, spleen, lung and kidney) by Em-myc/mir-1719b
tumour cells, shown by H&E and B220 staining. Invasion was observed both
perivascularly and parenchymally in liver. Scale bar, 50
m
m.
c, Immunophenotyping of Em-myc/mir-1719b lymphomas. Tumour cells
stained positively for the B-cell-specific marker B220, but not for the
T-cell-specific markers CD4, CD8a and Thy1.2. Tumour cells bore cellular
characteristics of pre-B cells, staining positively for CD19 but not for IgM, a
marker of mature B-cells.
LETTERS NATURE|Vol 435|9 June 2005
832 © 2005 Nature Publishing Group
using an ABI 7900HT Taqman sequence detector, following the ‘standard curve’
method. All values were subsequently normalized to the averaged ratio of the five
corresponding normal samples. For DNA copy number determination using the
ABI 7900HT sequence detector, we performed quantitative PCR analysis using
the same mir-1792 primer set described above, and normalized the data against
a reference probe corresponding to chromosomal region 6p22 (forward primer,
30-GGTCTCTATTTGCACTTGGCTGAT-50; reverse primer, 30-TTTTCA
TTGTTGACCAAGCTAGACA-50; probe, (6-FAM)-TAGGGCATACTGCCTG
CATATTTCCTGCT-(TAMRA)) or a b-actin probe. The reported values rep-
resent the ratios of DNA copy number at the mir-1792 locus to the normal
reference probe.
Adotptive transfer of Em-myc HSCs. Fetal liver-derived HSCs were isolated
from Em-myc/þmouse embryos at embryonic day (E)13.5–E15.5, and were
transduced with MSCV alone or MSCV expressing the mir-1719b cluster. To
exclude the possibility that the observed acceleration of lymphomagenesis was
due to insertional mutagenesis, independent experiments were carried out using
fetal liver cells isolated from eight Em-myc/þembryos. Cells from each isolation
were separately infected with MSCV mir-1719b or control MSCV. The MSCV
retroviral vector used in our studies contains a PGK-puromycin-IRES-GFP
(PIG) cassette
18
. Infection rates, assessed by fluorescence-activated cell
sorting (FACS), were typically 40% of bulk fetal liver cells. HSCs infected with
MSCV-mir1719b-PIG and MSCV-PIG (control) were subsequently trans-
planted into 6–8-week-old, lethally irradiated C57BL/6 recipient mice
17
. Tumour
onset was monitored by blood smear analysis, and tumour samples were either
collected into 4% paraformaldehyde for histopathological studies, or prepared as
single-cell suspensions for FACS.
We also carried out a screen of 96 miRNAs, to look for miRNA(s) that
accelerate Myc-induced lymphomagenesis. In this experiment, each pre-miRNA
(and ,50 bp of flanking sequence 50and 3 0of the pre-miRNA) was cloned
downstream of the cytomegalovirus (CMV) promoter in a MSCV vector
containing SV40–GFP. Eight individual MSCV constructs, each overexpressing
a specific miRNA, were pooled at equal concentrations. Twelve pools of DNA
were used individually to produce virus to infect Em-myc/þfetal liver cells for
adoptive transfer into at least three recipient animals, as described above.
Recipient animals were monitored for tumour growth for at least six months.
For those that developed lymphoma, tumour cells were prepared from the
enlarged lymph nodes and then subjected to FACS analysis for GFP expression.
Histopathology. Tissue samples were fixed in 4% paraformaldehyde, embedded
in paraffin, sectioned into 5-
m
m slices, and stained with haemotoxylin and eosin.
For Ki67 detection (rabbit anti-Ki67 antibody, NovoCastra), representative
sections were deparaffinized, rehydrated in graded alcohols, and processed
using the avidin–biotin immunoperoxidase method. Sections were then sub-
jected to antigen retrieval by microwave oven treatment using standard
procedures. Diaminobenzidine was used as the chromogen and haematoxylin
as the nuclear counterstain. For B220 immunohistochemistry, a rat anti-mouse
CD45R/B220 antibody (clone RA3-6B2, BD Biosciences Pharmingen) was used;
pretreatment for antigen retrieval was not required. Analysis of the rate of
apoptotis by TUNEL assay was performed according to a published protocol
27
.
Received 15 February; accepted 16 March 2005.
1. Ota, A. et al. Identification and characterization of a novel gene, C13orf25,asa
target for 13q31-q32 amplification in malignant lymphoma. Cancer Res. 64,
3087–-3095 (2004).
2. Lee, Y. et al. The nuclear RNase III Drosha initiates microRNA processing.
Nature 425, 415–-419 (2003).
3. Bernstein, E., Caudy, A. A., Hammond, S. M. & Hannon, G. J. Role for a
bidentate ribonuclease in the initiation step of RNA interference. Nature 409,
363–-366 (2001).
4. Bartel, D. P. MicroRNAs: genomics, biogenesis, mechanism, and function. Cell
116, 281–-297 (2004).
5. Ambros, V. The functions of animal microRNAs. Nature 431, 350–-355 (2004).
6. He, L. & Hannon, G. J. MicroRNAs: small RNAs with a big role in gene
regulation. Nature Rev. Genet. 5, 522–-531 (2004).
7. Ruvkun, G. & Giusto, J. The Caenorhabditis elegans heterochronic gene lin-14
encodes a nuclear protein that forms a temporal developmental switch. Nature
338, 313–-319 (1989).
8. Ambros, V. A hierarchy of regulatory genes controls a larva-to-adult
developmental switch in C. elegans.Cell 57, 49–-57 (1989).
9. Chang, S., Johnston, R. J. Jr, Frokjaer-Jensen, C., Lockery, S. & Hobert, O.
MicroRNAs act sequentially and asymmetrically to control chemosensory
laterality in the nematode. Nature 430, 785–-789 (2004).
10. Johnston, R. J. & Hobert, O. A microRNA controlling left/right neuronal
asymmetry in Caenorhabditis elegans.Nature 426, 845–-849 (2003).
11. Brennecke, J., Hipfner, D. R., Stark, A., Russell, R. B. & Cohen, S. M. bantam
encodes a developmentally regulated microRNA that controls cell proliferation
and regulates the proapoptotic gene hid in Drosophila.Cell 113, 25–-36 (2003).
12. Chen, C. Z., Li, L., Lodish, H. F. & Bartel, D. P. MicroRNAs modulate
hematopoietic lineage differentiation. Science 303, 83–-86 (2004).
13. Knuutila, S. et al. DNA copy number amplifications in human neoplasms:
review of comparative genomic hybridization studies. Am. J. Pathol. 152,
1107–-1123 (1998).
14. Tanzer, A. & Stadler, P. F. Molecular evolution of a microRNA cluster. J. Mol.
Biol. 339, 327–-335 (2004).
15. Tusher, V. G., Tibshirani, R. & Chu, G. Significance analysis of microarrays
applied to the ionizing radiation response. Proc. Natl Acad. Sci. USA 98,
5116–-5121 (2001).
16. Adams, J. M. et al. The c-myc oncogene driven by immunoglobulin enhancers
induces lymphoid malignancy in transgenic mice. Nature 318, 533–-538 (1985).
17. Schmitt, C. A. et al. Dissecting p53 tumor suppressor functions in vivo.Cancer
Cell 1, 289–-298 (2002).
18. Hemann, M. T. et al. An epi-allelic series of p53 hypomorphs created by stable
RNAi produces distinct tumor phenotypes in vivo.Nature Genet. 33, 396–-400
(2003).
19. Hemann, M. T. et al. Suppression of tumorigenesis by the p53 target PUMA.
Proc. Natl Acad. Sci. USA 101, 9333–-9338 (2004).
20. Wendel, H. G. et al. Survival signalling by Akt and eIF4E in oncogenesis and
cancer therapy. Nature 428, 332–-337 (2004).
21. Thomson, J. M., Parker, J., Perou, C. M. & Hammond, S. M. A custom
microarray plateform for analysis of microRNA gene expression. Nature
Methods 1, 47–-53 (2004).
22. Calin, G. A. et al. Human microRNA genes are frequently located at fragile sites
and genomic regions involved in cancers. Proc. Natl Acad. Sci. USA 101,
2999–-3004 (2004).
23. Calin, G. A. et al. MicroRNA profiling reveals distinct signatures in B cell
chronic lymphocytic leukemias. Proc. Natl Acad. Sci. USA 101, 11755–-11760
(2004).
24. Metzler, M., Wilda, M., Busch, K., Viehmann, S. & Borkhardt, A. High
expression of precursor microRNA-155/BIC RNA in children with Burkitt
lymphoma. Genes Chromosom. Cancer 39, 167–-169 (2004).
25. Michael, M. Z., O’Connor, S. M., van Holst Pellekaan, N. G., Young, G. P. &
James, R. J. Reduced accumulation of specific microRNAs in colorectal
neoplasia. Mol. Cancer Res. 1, 882–-891 (2003).
26. Calin, G. A. et al. Frequent deletions and down-regulation of micro- RNA genes
miR15 and miR16 at 13q14 in chronic lymphocytic leukemia. Proc. Natl Acad. Sci.
USA 99, 15524–-15529 (2002).
27. Di Cristofano, A., De Acetis, M., Koff, A., Cordon-Cardo, C. & Pandolfi, P. P.
Pten and p27
KIP1
cooperate in prostate cancer tumor suppression in the mouse.
Nature Genet. 27, 222–-224 (2001).
28. Mayor, C. et al. VISTA: visualizing global DNA sequence alignments of
arbitrary length. Bioinformatics 16, 1046–-1047 (2000).
Supplementary Information is linked to the online version of the paper at
www.nature.com/nature.
Acknowledgements We thank members of the Hannon, Lowe and Hammond
laboratories for discussions and input. We also thank Z. Xuan, N. Chen, N. Sheth
and R. Sachidanandam for bioniformatic analysis. C. Perou and J. Leib provided
advice and support for microarray methodologies, and A. Barnes and B. Boone
gave assistance with mouse tissue procurement. We are grateful to H. Wendel,
C. Scott, C. Marsden and C. Rosenthal, R. Karni, P. Moody and R. Whitaker, who
provided advice and technical support. F. Slack coined the oncomiR
nomenclature. L.H. and M.T.H. are Fellows of the Helen Hay Whitney
Foundation. S.W.L. and C.C.-C. are supported by a program project grant from
the NCI. G.J.H is supported by an Innovator Award from the US Army Breast
Cancer Research Program and by grants from the DOD and NIH. S.M.H. is a
General Motors Cancer Research Foundation Scholar, and J.M.T is a Frederick
Gardner Cottrell Postdoctoral Fellow.
Author Information Microarray data have been deposited in NCBI-GEO under
accession numbers GSM45026–GSM45065 and GSE-2399. Reprints and
permissions information is available at npg.nature.com/reprintsandpermissions.
The authors declare no competing financial interests. Correspondence and
requests for materials should be addressed to G.J.H. (hannon@cshl.edu) or
S.M.H. (hammond@med.unc.edu).
NATURE|Vol 435|9 June 2005 LETTERS
833
© 2005 Nature Publishing Group
... Numerous miRNAs have been found to have links to certain types of cancer [11,12]. Three observations made during the early stages of miRNA research suggested that they may play a role in human cancer. ...
Article
Full-text available
MicroRNAs (miRNAs) are small RNA molecules that regulate more than 30% of genes in humans. Recent studies have revealed that miRNAs play a crucial role in tumorigenesis. Large sets of miRNAs in human tumors are under-expressed compared to normal tissues. Furthermore, experiments have shown that interference with miRNA processing enhances tumorigenesis. Multiple studies have documented the causal role of miRNAs in cancer, and miRNA-based anticancer therapies are currently being developed. This review primarily focuses on two key points: (1) miRNAs and their role in human cancer and (2) the regulation of tumor suppressors by miRNAs. The review discusses (a) the regulation of the tumor suppressor p53 by miRNA, (b) the critical role of the miR-144/451 cluster in regulating the Itch-p63-Ago2 pathway, and (c) the regulation of PTEN by miRNAs. Future research and the perspectives of miRNA in cancer are also discussed. Understanding these pathways will open avenues for therapeutic interventions targeting miRNA regulation.
... Earlier studies demonstrated that miR-17-5p can act either as a tumor suppressor or a promoter, contingent upon the cellular context and the specific target mRNAs involved [70][71][72][73]. In Non-small cell lung carcinoma (NSCLC) and cervical, miR-17-5p is downregulated, whereas it is upregulated in hepatocellular, gastric, colorectal, breast, bladder, esophageal cancer, and pancreatic [74][75][76][77][78][79][80][81][82]. ...
Article
Full-text available
While intensity-modulated radiation therapy-based comprehensive therapy increases outcomes, cancer patients still have a low five-year survival rate and a high recurrence rate. The primary factor contributing to cancer patients’ poor prognoses is radiation resistance. A class of endogenous non-coding RNAs, known as microRNAs (miRNAs), controls various biological processes in eukaryotes. These miRNAs influence tumor cell growth, death, migration, invasion, and metastasis, which controls how human carcinoma develops and spreads. The correlation between the unbalanced expression of miRNAs and the prognosis and sensitivity to radiation therapy is well-established. MiRNAs have a significant impact on the regulation of DNA repair, the epithelial-to-mesenchymal transition (EMT), and stemness in the tumor radiation response. But because radio resistance is a complicated phenomena, further research is required to fully comprehend these mechanisms. Radiation response rates vary depending on the modality used, which includes the method of delivery, radiation dosage, tumor stage and grade, confounding medical co-morbidities, and intrinsic tumor microenvironment. Here, we summarize the possible mechanisms through which miRNAs contribute to human tumors’ resistance to radiation.
... As a result, under typical circumstances, the feedback loop between the miR-17-92 cluster and E2F offers a way to maintain regular cell cycle progression. But miR-17-92 overexpression, which is prevalent in a number of tumours, shatters the feedback loop that encourages cell division [43]. ...
Article
Full-text available
Oral cancer (OC) is among the most common malignancies in the world. Despite advances in therapy, the worst-case scenario for OC remains metastasis, with a 50% survival rate. Therefore, it is critical to comprehend the pathophysiology of the condition and to create diagnostic and treatment plans for OC. The development of high-throughput genome sequencing has revealed that over 90% of the human genome encodes non-coding transcripts, or transcripts that do not code for any proteins. This paper describes the function of these different kinds of non-coding RNAs (ncRNAs) in OC as well as their intriguing therapeutic potential. The onset and development of OC, as well as treatment resistance, are linked to dysregulated ncRNA expression. These ncRNAs’ potentially significant roles in diagnosis and prognosis have been suggested by their differing expression in blood or saliva. We have outlined every promising feature of ncRNAs in the treatment of OC in this study.
... This was supported by data suggesting at least 50% of miRNAs are located at disturbed genomic sites involved in cancer [41]. Demonstrated with the 13q31 region found to be amplified in B-cell lymphomas which includes the miR-17-92 cluster of seven miRNAs resulting in a 10-fold increase in pri-miRNA levels in 65% of B-Cell lymphoma samples [42]. In parallel, the TGCT-related miRNAs exhibiting increased expression, such as miR-371a-3p, are found in the 19q13.42 ...
Preprint
Full-text available
MicroRNAs, short non-protein coding RNAs, are overexpressed in GCTs. Circulating levels of germ cell tumor (GCT)- associated miRNAs, such as miR-371a-3p, can be utilized as efficient and cost-effective alternatives in diagnosing and managing patients presenting with GCTs. This quality of miRNAs has demonstrated favorable performance characteristics as a reliable blood-based biomarker with high diagnostic accuracy compared to current serum tumor markers (STMs) including α-fetoprotein (AFP), beta human chorionic gonadotropin (β-hCG), and lactate dehydrogenase (LDH). The conventional STMs exhibit limited specificity and sensitivity. Potential clinical implications of miRNAs include impact on de-escalating or intensifying treatment, detecting recurrence at earlier stages, and lessening the necessity of cross-sectional imaging or invasive tissue biopsy for non-teratomatous GCTs. Here we also highlight the outstanding issues that must be addressed prior to clinical implementation. Standards for measuring circulating miRNAs and determining ideal cutoff values are essential for integration into current clinical guidelines.
... 41 These clusters exhibit coexpression and target multiple mRNA transcripts and coordinate the regulation of related cellular pathways. 42,43 Nine clusters with 2 downregulated and 7 upregulated miRs were identified in our study. The downregulated clusters contain miR-212/132 and miR-125a/let-7e/99b, while the upregulated clusters include miR-431/127/434/136, miR-412/409/369/410/541/412, miR-411/379/380, miR-485/382/668, miR-154/496a, miR-543/ 666, and miR-493/337. ...
Article
Full-text available
Background and Objectives Micro-RNAs (miRNAs) are critical for regulating the expression of genes in multiple neurodegenerative diseases, but miRNAs have not been investigated in spinocerebellar ataxia type 2 (SCA2). SCA2, a dominantly inherited progressive neurodegenerative polyglutamine (polyQ) disease, is caused by a CAG repeat expansion in the ataxin-2 (ATXN2) gene. In this study, we determined miRNA transcriptomes in SCA2-BAC-ATXN2[Q72] transgenic mice. Methods We assessed the expression of miRNAs in SCA2 transgenic mouse cerebella using the HiSeq Illumina sequencer. We used the miRNA target filter tool in Qiagen Ingenuity Pathway Analysis (IPA) to identify target genes of differentially expressed miRNAs (DEmiRs) within in the SCA2 mouse transcriptomes and then performed pathway analyses. Results Our analysis revealed significant changes in the expression levels of multiple miRNAs in mice with SCA2. We identified 81 DEmiRs in mice with SCA2, with 52 miRNAs upregulated and 29 miRNAs downregulated after onset of rotarod deficit. Subsequent IPA processing enabled us to establish connections between these DEmiRs and specific biological regulatory functions. Furthermore, by using the IPA miRNA target filter, we identified target genes of DEmiRs in the SCA2-BAC-ATXN2[Q72] transcriptome data set and demonstrated their significant impact on several biological functional and disease pathways. Discussion Our study establishes the role of both DEmiRs and their targets in SCA2 pathogenesis. By expressing mutant ATXN2 under the control of its endogenous regulatory elements in the SCA2-BAC-ATXN2[Q72] mouse model, we identified a set of DEmiRs that are shared across multiple neurodegenerative diseases including other SCAs, Alzheimer disease (AD), Parkinson disease (PD), and amyotrophic lateral sclerosis (ALS). There was a significant overlap of both DEmiRs and their targets of BAC-ATXN2[Q72] transcriptomes in dysregulated pathways that characterize SCA2. This observation also extended to dysregulated pathways in ALS, AD, and PD. DEmiRs identified in this study may represent therapeutic targets for neurodegeneration or lead to biomarkers for characterizing various neurodegenerative diseases.
... This was supported by data suggesting that at least 50% of miRNAs are located at disturbed genomic sites involved in cancer [41]. This has been demonstrated with the 13q31 region found to be amplified in B-cell lymphomas, which includes the miR-17-92 cluster of seven miRNAs, resulting in a 10-fold increase in pri-miRNA levels in 65% of B-cell lymphoma samples [42]. In parallel, the TGCT-related miRNAs exhibiting increased expression, such as miR-371a-3p, are found in the 19q13.42 ...
Article
Full-text available
Simple Summary The diagnosis and management of testicular germ cell tumors (TGCTs) currently relies on the use of unreliable biomarkers—conventional serum tumor markers (STMs). Variable levels of elevation in serum samples among patients and cancer types make the use of conventional STMs unfavorable for dependable diagnosis and clinical management of TGCTs. This review of recent studies suggests germ cell tumor associated miRNAs, such as miR-371a-3p, can be utilized as not only a reliable but cost-effective and convenient blood-based biomarker. However, before widespread clinical implementation, the development of a protocol for sample collection, analysis, and interpretation is necessary to prevent overtreatment and guide the management of patients. Abstract MicroRNAs, short non-protein coding RNAs, are overexpressed in GCTs. Circulating levels of germ cell tumor (GCT)-associated miRNAs, such as miR-371a-3p, can be utilized as efficient and cost-effective alternatives in diagnosing and managing patients presenting with GCTs. This quality of miRNAs has demonstrated favorable performance characteristics as a reliable blood-based biomarker with high diagnostic accuracy compared to current serum tumor markers (STMs), including α-fetoprotein (AFP), beta human chorionic gonadotropin (β-hCG), and lactate dehydrogenase (LDH). The conventional STMs exhibit limited specificity and sensitivity. Potential clinical implications of miRNAs include impact on de-escalating or intensifying treatment, detecting recurrence at earlier stages, and lessening the necessity of cross-sectional imaging or invasive tissue biopsy for non-teratomatous GCTs. Here, we also highlight the outstanding issues that must be addressed prior to clinical implementation. Standards for measuring circulating miRNAs and determining ideal cutoff values are essential for integration into current clinical guidelines.
Chapter
MicroRNAs (miRNAs) are tiny RNA molecules that, while not directly involved in the protein translation process, indirectly regulate protein production by targeting specific genes or messenger RNAs (mRNAs). Some miRNAs play crucial roles in various human diseases like cancer, diabetes, viral infections, and cardiovascular issues. Understanding the regulatory relationships between genes and miRNAs is essential for identifying potential drug targets for these serious diseases. This chapter introduces a multiobjective miRNA module detection algorithm designed to identify groups of miRNAs associated with different cancer types. The algorithm optimizes two objective functions simultaneously, based on changes in miRNA coexpression patterns across different conditions and functional similarities within miRNA pairs. The non-dominated sorting genetic algorithm-II (NSGA-II) is employed for optimization. The technique’s superiority over existing algorithms in identifying microRNA markers is demonstrated through experiments based on several real-life miRNA expression datasets. The chapter also explores the biological significance of the mRNA targets of the identified miRNA markers.
Article
Objectives: MiR-1253, miR-504-5p, miR-26a-5p, miR-21-3p, and miR495-3p have been identified as driver mutation-specific microRNAs (miRNAs) in human non-small-cell lung cancer. We examined the expression of these miRNAs in human lung adenocarcinoma cell lines and investigated whether the miRNAs truly show specific driver mutations. Material and Methods: We clarified the status of EGFR and KRAS mutations reported in the literature (2 cell lines) and those identified by DNA sequence analysis (7 cell lines) in human lung adenocarcinoma cell lines, and analyzed the expression of the above driver mutation-specific miRNAs in all 9 cell lines. Results: MiR-504-5p, miR-26a-5p, miR-21-3p, and miR495-3p did not show any expression patterns related to driver mutations in any of the human lung adenocarcinoma cell lines analyzed. On the basis of cytomorphological characteristics, we could not determine the association between driver mutation and morphological changes. Discussion: The results of this study indicate that the changes in miR-504-5p, miR-26a-5p, miR-21-3p, and miR495-3p were not directly caused by driver mutations that occurred in cancer cells. However, these changes may indicate driver mutation-led transformations in the tumor tissue environment by driver mutation, suggesting that the miRNAs are products of peritumor noncancer cells rather than tumor cells.
Article
Full-text available
A large number of tiny noncoding RNAs have been cloned and named microRNAs (miRs). Recently, we have reported that miR-15a and miR-16a, located at 13q14, are frequently deleted andor down-regulated in patients with B cell chronic lymphocytic leuke-mia, a disorder characterized by increased survival. To further investigate the possible involvement of miRs in human cancers on a genome-wide basis, we have mapped 186 miRs and compared their location to the location of previous reported nonrandom genetic alterations. Here, we show that miR genes are frequently located at fragile sites, as well as in minimal regions of loss of heterozygosity, minimal regions of amplification (minimal ampli-cons), or common breakpoint regions. Overall, 98 of 186 (52.5%) of miR genes are in cancer-associated genomic regions or in fragile sites. Moreover, by Northern blotting, we have shown that several miRs located in deleted regions have low levels of expression in cancer samples. These data provide a catalog of miR genes that may have roles in cancer and argue that the full complement of miRs in a genome may be extensively involved in cancers.
Article
Full-text available
The genetic bases underlying prostate tumorigenesis are poorly understood. Inactivation of the tumor-suppressor gene PTEN and lack of p27KIP1 expression have been detected in most advanced prostate cancers1, 2. But mice deficient for Cdkn1b (encoding p27Kip1) do not develop prostate cancer3, 4, 5. PTEN activity leads to the induction of p27KIP1 expression, which in turn can negatively regulate the transition through the cell cycle6. Thus, the inactivation of p27KIP1 may be epistatic to PTEN in the control of the cell cycle. Here we show that the concomitant inactivation of one Pten allele and one or both Cdkn1b alleles accelerates spontaneous neoplastic transformation and incidence of tumors of various histological origins. Cell proliferation, but not cell survival, is increased in Pten +/-/Cdkn1b -/- mice. Moreover, Pten +/-/Cdkn1b -/- mice develop prostate carcinoma at complete penetrance within three months from birth. These cancers recapitulate the natural history and pathological features of human prostate cancer. Our findings reveal the crucial relevance of the combined tumor-suppressive activity of Pten and p27Kip1 through the control of cell-cycle progression.
Article
Full-text available
This review summarizes reports of recurrent DNA sequence copy number amplifications in human neoplasms detected by comparative genomic hybridization. Some of the chromosomal areas with recurrent DNA copy number amplifications (amplicons) of 1p22-p31, 1p32-p36, 1q, 2p13-p16, 2p23-p25, 2q31-q33, 3q, 5p, 6p12-pter, 7p12-p13, 7q11.2, 7q21-q22, 8p11-p12, 8q, 11q13-q14, 12p, 12q13-q21, 13q14, 13q22-qter, 14q13-q21, 15q24-qter, 17p11.2-p12, 17q12-q21, 17q22-qter, 18q, 19p13.2-pter, 19cen-q13.3, 20p11.2-p12, 20q, Xp11.2-p21, and Xp11-q13 and genes therein are presented in more detail. The paper with more than 150 references and two tables can be accessed from our web site http://www.helsinki.fi/lglvwww/CMG.html. The data will be updated biannually until the year 2001.
Article
Full-text available
VISTA is a program for visualizing global DNA sequence alignments of arbitrary length. It has a clean output, allowing for easy identification of similarity, and is easily configurable, enabling the visualization of alignments of various lengths at different levels of resolution. It is currently available on the web, thus allowing for easy access by all researchers. Availability: VISTA server is available on the web at http://www-gsd.lbl.gov/vista. The source code is available upon request. Contact: vista{at}lbl.gov
Article
Nature Reviews Genetics 5, 522–531 (2004)In figure 2, the orientation of some RNA structures was incorrect. The corrected version is shown below.
Article
Cell proliferation, cell death, and pattern formation are coordinated in animal development. Although many proteins that control cell proliferation and apoptosis have been identified, the means by which these effectors are linked to the patterning machinery remain poorly understood. Here, we report that the bantam gene of Drosophila encodes a 21 nucleotide microRNA that promotes tissue growth. bantam expression is temporally and spatially regulated in response to patterning cues. bantam microRNA simultaneously stimulates cell proliferation and prevents apoptosis. We identify the pro-apoptotic gene hid as a target for regulation by bantam miRNA, providing an explanation for bantam's anti-apoptotic activity.
Article
MicroRNAs (miRNAs), an abundant class of ~22 nucleotide non-coding RNAs, are thought to play an important regulatory role in animal and plant development at the posttranscriptional level. Many miRNAs cloned from mouse bone marrow cells are differentially regulated in various hematopoietic lineages, suggesting that they might influence hematopoietic lineage differentiation. Some human miRNAs are linked to leukemias: the miR-15a/miR-16 locus is frequently deleted or down-regulated in patients with B-cell chronic lymphocytic leukemia and miR-142 is at a translocation site found in a case of aggressive B-cell leukemia. miR-181, a miRNA upregulated only in the B cell lineage of mouse bone marrow cells, promotes B cell differentiation and inhibits production of CD8⁺ T cells when expressed in hematopoietic stem/progenitor cells. In contrast miR-142s inhibits production of both CD4⁺ and CD8⁺ T cells and does not affect B cells. Collectively, these results indicate that microRNAs are components of the molecular circuitry controlling mouse hematopoiesis and suggest that other microRNAs have similar regulatory roles during other facets of vertebrate development. Singapore-MIT Alliance (SMA)
Article
The heterochronic genes lin-4, lin-14, lin-28, and lin-29 control the timing of specific postembryonic developmental events in C. elegans. The experiments described here examine how these four genes interact to control a particular stage-specific event of the lateral hypodermal cell lineages. This event, termed the "larva-to-adult switch" (L/A switch), involves several coordinate changes in the behavior of hypodermal cells at the fourth molt: cessation of cell division, formation of adult (instead of larval) cuticle, cell fusion, and cessation of the molting cycle. The phenotypes of multiply mutant strains suggest a model wherein the L/A switch is controlled by the stage-specific activity of a regulatory hierarchy: At early stages of wild-type development, lin-14 and lin-28 inhibit lin-29 and thus prevent switching. Later, lin-4 inhibits lin-14 and lin-28, allowing activation of lin-29, which in turn triggers the switch in the L4 stage. lin-29 may activate the L/A switch by regulating genes that control cell division, differentiation, and stage-specific gene expression in hypodermal cells.
Article
During wild-type development, a protein product of the Caenorhabditis elegans heterochronic gene lin-14 is localized to nuclei of specific somatic cells in embryos and early larvae, but is absent in late larvae and adult soma. Gain-of-function lin-14 mutations cause the level of lin-14 protein to remain high throughout development, resulting in developmental reiterations of early cell lineages. The normal down-regulation of the lin-14 nuclear protein level encodes a temporal switch between early and late cell fates.
Article
Transgenic mice bearing the cellular myc oncogene coupled to the immunoglobulin mu or kappa enhancer frequently develop a fatal lymphoma within a few months of birth. Since the tumours represent represent both immature and mature B lymphocytes, constitutive c-myc expression appears to be highly leukaemogenic at several stages of B-cell maturation. These myc mice should aid study of lymphoma development, B-cell ontogeny and immunoglobulin regulation.