ArticlePDF Available

Adult somatic stem cells in the human parasite Schistosoma mansoni

Authors:

Abstract and Figures

Schistosomiasis is among the most prevalent human parasitic diseases, affecting more than 200 million people worldwide. The aetiological agents of this disease are trematode flatworms (Schistosoma) that live and lay eggs within the vasculature of the host. These eggs lodge in host tissues, causing inflammatory responses that are the primary cause of morbidity. Because these parasites can live and reproduce within human hosts for decades, elucidating the mechanisms that promote their longevity is of fundamental importance. Although adult pluripotent stem cells, called neoblasts, drive long-term homeostatic tissue maintenance in long-lived free-living flatworms (for example, planarians), and neoblast-like cells have been described in some parasitic tapeworms, little is known about whether similar cell types exist in any trematode species. Here we describe a population of neoblast-like cells in the trematode Schistosoma mansoni. These cells resemble planarian neoblasts morphologically and share their ability to proliferate and differentiate into derivatives of multiple germ layers. Capitalizing on available genomic resources and RNA-seq-based gene expression profiling, we find that these schistosome neoblast-like cells express a fibroblast growth factor receptor orthologue. Using RNA interference we demonstrate that this gene is required for the maintenance of these neoblast-like cells. Our observations indicate that adaptation of developmental strategies shared by free-living ancestors to modern-day schistosomes probably contributed to the success of these animals as long-lived obligate parasites. We expect that future studies deciphering the function of these neoblast-like cells will have important implications for understanding the biology of these devastating parasites.
Content may be subject to copyright.
Adult somatic stem cells in the human parasite, Schistosoma
mansoni
James J. Collins III1,2, Bo Wang1,3, Bramwell G. Lambrus1, Marla Tharp1, Harini Iyer1, and
Phillip A. Newmark1,2,*
1Howard Hughes Medical Institute and Department of Cell and Developmental Biology, University
of Illinois at Urbana-Champaign, Urbana, IL 61801, USA
2Neuroscience Program, University of Illinois at Urbana-Champaign, Urbana, IL 61801, USA
3Institute for Genomic Biology, University of Illinois at Urbana-Champaign, Urbana, IL 61801,
USA
Summary
Schistosomiasis is among the most prevalent human parasitic diseases, affecting more than 200
million people worldwide1. The etiological agents of this disease are trematode flatworms
(
Schistosoma
) that live and lay eggs within the vasculature of the host. These eggs lodge in host
tissues, causing inflammatory responses that are the primary cause of morbidity. Because these
parasites can live and reproduce within human hosts for decades2, elucidating the mechanisms that
promote their longevity is of fundamental importance. Although adult pluripotent stem cells,
called neoblasts, drive long-term homeostatic tissue maintenance in long-lived free-living
flatworms3,4 (e.g., planarians), and neoblast-like cells have been described in some parasitic
tapeworms5, little is known about whether similar cell types exist in any trematode species. Here,
we describe a population of neoblast-like cells in the trematode
Schistosoma mansoni
. These cells
resemble planarian neoblasts morphologically and share their ability to proliferate and
differentiate into derivatives of multiple germ layers. Capitalizing on available genomic
resources6,7 and RNAseq-based gene expression profiling, we find that these schistosome
neoblast-like cells express a
fibroblast growth factor receptor
ortholog. Using RNA interference
we demonstrate that this gene is required for the maintenance of these neoblast-like cells. Our
observations suggest that adaptation of developmental strategies shared by free-living ancestors to
modern-day schistosomes likely contributed to the success of these animals as long-lived obligate
parasites. We expect that future studies deciphering the function of these neoblast-like cells will
have important implications for understanding the biology of these devastating parasites.
Although classic studies of cell proliferation in
Schistosoma
focused on reproductive
tissues8,9, Den Hollander and Erasmus did note occasional “undifferentiated” somatic cells
that incorporated tritiated thymidine in adult parasites. Encouraged that these cells could
*Corresponding author: pnewmark@life.illinois.edu.
Full Methods and any associated references are available in the online version of the paper.
Supplementary Information is linked to the online version of the paper at www.nature.com/nature
Author Contributions J.J.C., B.W., B.G.L., M.T., H.I., performed experiments. J.J.C. and B.W. analyzed data. J.J.C. and P.A.N.
designed the study and wrote the paper.
Author Information Experiments with and care of vertebrate animals were performed in accordance with protocols approved by the
Institutional Animal Care and Use Committee (IACUC) of the University of Illinois at Urbana-Champaign (protocol approval number
10035). RNAseq analyses have been deposited in the NCBI Gene Expression Omnibus (Accession number: GSE42757). The authors
declare no competing financial interests.
NIH Public Access
Author Manuscript
Nature
. Author manuscript; available in PMC 2013 August 28.
Published in final edited form as:
Nature
. 2013 February 28; 494(7438): 476–479. doi:10.1038/nature11924.
NIH-PA Author Manuscript NIH-PA Author Manuscript NIH-PA Author Manuscript
represent neoblast-like stem cells, we treated adult
S. mansoni
with the thymidine analogue
5-ethynyl-2'-deoxyuridine (EdU)10 to examine the distribution of S-phase cells in the
parasite (Fig. 1a,b). In addition to the expected incorporation in the highly proliferative
reproductive organs (testes, ovaries, and vitellaria) (Fig. 1a,b and Supplementary Fig. 1), we
observed a population of EdU+ cells throughout the soma of male and female parasites (Fig.
1a–d). Similar distributions of EdU-incorporating cells were observed whether parasites
were given EdU during
in vitro
culture or
in vivo
by intraperitoneal injection of
schistosome-infected mice. Analogous to the neoblasts in free-living flatworms11,12, these
proliferating somatic cells (PSCs) were restricted to the mesenchyme of male and female
worms (Fig. 1c,d), not associated with reproductive organs, and were often found in clusters
near the intestine (Supplementary Fig. 2a). We also observed a conspicuous population of
PSCs adjacent to the ventral sucker (Supplementary Fig. 2b). PSCs traversed the cell cycle:
they initially expressed the cell cycle-associated transcript
histone h2b
(Supplementary Fig.
3a–c) and progressed to M-phase within 24 hours following an EdU pulse (Supplementary
Fig. 3d).
Neoblasts are the only proliferating somatic cells in planarians4,11 and they possess a
distinct morphology; they are round-to-ovoid mesenchymal cells with a high nuclear-
tocytoplasmic ratio, a large nucleolus, and they often extend a cytoplasmic projection3,11,13.
To determine if PSCs share similarities with planarian neoblasts, we examined these cells by
dissociating male tissues devoid of germ cells (Fig. 1e). In these preparations we observed a
number of distinct differentiated cell types that failed to incorporate EdU, including cells
with a low nuclear-to-cytoplasmic ratio, neuron-like cells, and ciliated cells (Fig. 1f). By
contrast, we found that EdU incorporation was restricted to a neoblast-like population of
cells with scant cytoplasm (n=136/137 cells) and often a prominent nucleolus (Fig. 1f). We
also inspected PSCs within the mesenchyme using EdU to label nuclei and fluorescent in
situ hybridization (FISH) to detect
histone h2b
mRNA in the cytoplasm of proliferative
cells. Consistent with our results from tissue macerates, EdU+ cells possess a narrow rim of
cytoplasm surrounding their nucleus, and these cells often display a cytoplasmic projection
(Fig. 1g). These observations highlight morphological similarities between proliferating
cells in schistosomes and planarian neoblasts.
Previous studies have exploited the sensitivity of planarian neoblasts to γ-irradiation as a
means to identify neoblast-enriched transcripts14–16. Using this strategy to identify PSC-
expressed genes, we exposed parasites to various dosages of γ-irradiation and determined
that 100–200 Gy were sufficient to block EDU incorporation (Fig. 2a). Because of their high
ratio of somatic tissue to reproductive tissue and their large number of PSCs relative to
female worms (compare insets in Fig. 1a and 1b), our remaining studies, unless otherwise
noted, focused on male parasites. By comparing the transcriptional profiles of irradiated and
non-irradiated parasites by RNAseq (Fig. 2b), we identified 128 genes with significantly
down-regulated expression (≥ 2-fold, p < 0.05) 48 hours post-irradiation (Fig. 2c and
Supplementary Table 1). Highlighting the efficacy of this approach to identify transcripts
specific to proliferating cells, we found that genes expressed in differentiated tissues, such as
the intestine (
Sm
-
cathepsin B
17), were unaffected by irradiation. By contrast, our list of
down-regulated genes was enriched for factors involved in the cell cycle (Supplementary
Fig.4 and Supplementary Table 1).
In addition to identifying cell cycle-associated factors, systematic comparison of irradiation-
sensitive genes with neoblast-enriched transcripts14,15,18,19 uncovered a number of
interesting similarities (Supplementary Table 1). For instance, homologues of genes known
to regulate planarian neoblasts such as
p53
, a sox-family transcription factor, fibroblast
growth factor receptors, and
argonaute2
14,20–22, were significantly down-regulated in
irradiated schistosomes (Supplementary Fig. 4 and Supplementary Table 1). Another
Collins et al. Page 2
Nature
. Author manuscript; available in PMC 2013 August 28.
NIH-PA Author Manuscript NIH-PA Author Manuscript NIH-PA Author Manuscript
distinctive feature of neoblasts3,14,22, and the somatic stem cells of other invertebrates23, is
that they often express post-transcriptional regulators associated with germline development
(e.g.,
vasa, piwi, tudor, and nanos
). Although
vasa-like
genes have been reported in
Schistosoma
, no true
vasa
orthologue has been indentified24. Similarly,
piwi
and
tudor
genes
appear to be absent from schistosomes (data not shown). However, we identified a
nanos
orthologue (
Sm-nanos-2
) that was down-regulated in somatic tissue following irradiation
(Supplementary Fig. 4, and Supplementary Table 1). Since these genes represented potential
regulators of PSC behavior and could serve as useful markers for these cells, we examined
their expression by whole-mount in situ hybridization (WISH). We detected
Sm-ago2-1
,
Sm-nanos-2
, and
Sm-fgfrA
transcripts in cells scattered throughout the mesenchyme (Fig.
2d and Supplementary Fig. 5) in a pattern similar to that of cells incorporating EdU (Fig.
1A). This mesenchymal expression was radiation sensitive (Fig. 2d), suggesting these genes
are expressed in proliferating cells. Consistent with this idea, we found that following an
EdU pulse, >99% of EdU-incorporating somatic cells also expressed
Sm-fgfrA
(Fig. 2e).
To determine if PSCs are stem cells, we assessed their ability both to self-renew and to
produce differentiated cell types. To examine self-renewal, we administered sequential
pulses of EdU and 5-Bromo-2'-deoxyuridine (BrdU) to parasites
in vitro
. Because nearly all
PSCs that incorporate EdU are
Sm-fgfrA
+, the ability of EdU+ cells to incorporate BrdU in
subsequent cell cycles would suggest that
fgfrA
+ PSCs self-renew (i.e., divide and produce
more
fgfrA
+ PSCs). For these experiments we chose a chase period of 44 hours, since this
time frame should give many EdU+ PSCs sufficient time to divide (Supplementary Fig. 3d).
Consistent with PSCs possessing the capacity for self-renewal, we find that 41% of cells that
initially incorporate EdU are BrdU+ 3 days following an initial EdU pulse (Fig. 3a).
Furthermore, we observed that many EdU+ cells were distributed in pairs, or “doublets”
(Fig. 3a); we suggest a majority of these doublets are the products of cell division
(Supplementary Discussion and Supplementary Fig. 6). In these EdU+ doublets, a
disproportionately large fraction displayed asymmetric BrdU incorporation (i.e. one nucleus
is EdU+BrdU+, while the other is EdU+BrdU) (Fig. 3b and Supplementary Discussion).
This observation suggests that division progeny have an asymmetric capacity to proliferate.
Whether this represents stem cell-like asymmetric division or temporal differences in the
ability of these cells to reenter the cell cycle requires further experimentation. Nevertheless,
these data are consistent with PSCs (or some PSC subpopulation) being capable of self-
renewal.
To examine the capacity of PSCs to differentiate, we performed EdU pulse-chase
experiments
in vivo
. For these experiments, schistosome-infected mice were injected with
EdU and the distribution of EdU+ cells was monitored at early (D1) and late (D7) time
points (Fig. 3c). We successfully used this pulse-chase approach to monitor the
differentiation of schistosome germ cells (Supplementary Fig. 7). Visualizing the syncytial
epithelium of the schistosome intestine at D1, we did not observe EdU+ intestinal nuclei in
male or female parasites (Fig. 3d, 0 EdU+/3151 DAPI+ nuclei, 14 mixed sex parasites, n=5
mice), confirming that cells in the intestine do not proliferate. Following a 7-day chase,
however, ~2.5% of the intestinal nuclei were EdU+ (Fig. 3e, 56 EdU+/2189 DAPI+ nuclei,
10 mixed sex parasites, n=3 mice). This observation suggests that cells initially labeling
with EdU have the capacity to migrate into the intestine and differentiate into new intestinal
cells. Similarly, we were able to monitor the differentiation of new cells in the body wall
muscles. At D1 no EdU+ nuclei were observed in the male body wall musculature (Fig. 3f, 0
EdU+/1882 DAPI+ nuclei, 13 male parasites, n=6 mice), whereas at D7 ~10% of the muscle
cell nuclei were EdU+ (Fig. 3g, 55 EdU+/584 DAPI+ nuclei, 6 male parasites, n=3 mice).
Since virtually all cells that initially incorporate EdU are
Sm-fgfrA
+, we suggest that these
double-positive cells are likely to represent the only source of new intestinal and muscle
cells and, thus, represent a collectively multipotent population of neoblast-like stem cells.
Collins et al. Page 3
Nature
. Author manuscript; available in PMC 2013 August 28.
NIH-PA Author Manuscript NIH-PA Author Manuscript NIH-PA Author Manuscript
Whether all
Sm-fgfrA
+ PSCs are multipotent or whether they exist as lineage-restricted
progenitors remains unclear.
While progress has been made in identifying transcriptional14 and post-transcriptional22
regulators of planarian neoblasts, little is known about the signal transduction networks
functioning within these cells. Since the expression of FGF receptor family members in
proliferative cells is conserved between planarians14,21 and schistosomes, we speculated that
FGF signaling could regulate these cells in
S. mansoni
. To examine this idea, we disrupted
Sm-fgfrA
in
in vitro
-cultured adult parasites using RNA interference (Supplementary Fig.
8). We found that inhibition of
Sm-fgfrA
resulted in reduced EdU incorporation (Fig. 4a,b
and Supplementary Table 2) and down-regulation of cell cycle-associated transcripts (Fig.
4c and Supplementary Fig. 8). To resolve whether this effect is due to reduced cell
proliferation or a failure to maintain neoblast-like cells, we monitored the expression of PSC
markers
Sm-ago2-1
and
Sm-nanos-2
in
Sm-fgfrA(RNAi)
parasites.
Sm-fgfrA
RNAi
treatment resulted in a dramatic reduction in the number of cells expressing
Sm-nanos-2
(Fig. 2c) as well as significantly reduced mRNA levels for
Sm-ago2-1
and
Sm-nanos-2
(Supplementary Fig. 8b). Together, these results suggest that
Sm-fgfrA
promotes the long-
term maintenance of neoblast-like cells in
S. mansoni
. FGF signaling is known to influence
multiple processes, such as cell proliferation, differentiation, and survival; furthermore, it
plays key roles in various stem cell populations25. Our results suggest a conserved role for
FGF signaling in controlling stem cell behavior in these parasites and demonstrate the
feasibility of using RNAi to abrogate adult gene expression and manipulate neoblast-like
cells in
S. mansoni
.
Adult schistosomes can modulate growth in response to host immune signals26 and male-
female pairing status2,27 and they can regenerate damaged tissues following sub-lethal doses
of the anti-schistosomal drug praziquantel28. These observations reveal the developmental
plasticity of schistosomes, and suggest that these parasites can utilize distinct developmental
programs in response to a range of external stimuli. Future studies characterizing the role of
neoblast-like cells in diverse contexts could address long-standing gaps in our knowledge of
schistosome biology and may reveal novel therapeutic strategies for treating and eliminating
schistosomiasis.
Methods
Parasite Acquisition and Culture
Adult
S. mansoni
(6–8 weeks post-infection) were obtained from infected mice by hepatic
portal vein perfusion31 with 37°C DMEM (Mediatech, Manassas, VA) plus 5% Fetal Calf
Serum (FBS, Hyclone/Thermo Scientific Logan, UT). Parasites were rinsed several times in
DMEM + 5% FBS and cultured (37°C/5% CO2) in Basch's Medium 16932 and 1×
Antibiotic-Antimycotic (Gibco/Life Technologies, Carlsbad, CA 92008). Media was
changed every 1–3 days.
EdU labeling
For
in vitro
labeling, parasites were cultured in Basch's Medium 169 supplemented with 10
μM EdU (Invitrogen, Carlsbad, CA) diluted from a 10 mM stock in DMSO. Unless
otherwise noted, animals were pulsed for 18–24 hours. For
in vivo
labeling, schistosome-
infected mice (6–8 weeks post-infection) were given a single intraperitoneal injection (100–
200 mg EdU/kg bodyweight) with 5 mg/ml EdU dissolved in PBS and then harvested at
various time points after injection.
Collins et al. Page 4
Nature
. Author manuscript; available in PMC 2013 August 28.
NIH-PA Author Manuscript NIH-PA Author Manuscript NIH-PA Author Manuscript
In situ hybridization
Male and female parasites were separated by incubation (2–3 minutes) in a 0.25% solution
of the anaesthetic ethyl 3-aminobenzoate methanesulfonate (Sigma-Aldrich, St. Louis, MO)
dissolved in Basch's Medium 169 or Phosphate Buffered Saline (PBS). Relaxed parasites
were then killed in a 0.6 M solution of MgCl2 and fixed for 4.5 hrs in 4% Formaldehyde
dissolved in PBSTx (PBS + 0.3% Triton X-100). Following fixation, parasites were
dehydrated in MeOH and stored at −20°C. Samples were rehydrated by incubation in 1:1
MeOH:PBSTx followed by incubation in PBSTx. Rehydrated samples were bleached for 1–
2 hours in formamide bleaching solution (0.5% Formamide, 0.5% SSC, and 1.2% H2O2),
rinsed with PBSTx, treated with Proteinase K (2–10 μg/mL, Invitrogen, Carlsbad, CA) for
20–30 minutes at room temperature and post-fixed for 10–15 minutes in 4% formaldehyde
in PBSTx. Samples were hybridized at 52–55°C and otherwise processed as previously
described29,33. Plasmids used for riboprobe synthesis were generated as described
previously29 using oligonucleotide primers listed in Supplementary Table 3.
Immunofluorescence, histological staining, and EdU detection
Parasites were relaxed, killed, fixed, dehydrated and rehydrated as described above and
bleached in 6% H2O2 dissolved in PBS for 0.5–2 h. Dehydration and bleaching were
omitted for samples labeled with phalloidin. Samples were then treated with Proteinase K
and post-fixed as described above. Immunofluorescence, lectin, and phalloidin staining were
performed as described previously30. Rabbit anti-Phospho-Histone H3 Ser10 (anti-pH3)
(D2C8, Cell Signaling, Danvers, MA), rhodamine-conjugated sWGA (Vector Laboratories
Burlingame, CA), and Alexa Fluor 568 phalloidin (Invitrogen, Carlsbad, CA), were used at
1:1000, 1:100, and 1:100, respectively. EdU detection was performed essentially as
previously described10,34 with 100 μM Alexa Fluor 488 or Alexa Fluor 594 azide
conjugates. All imaging was performed as described previously29,30. To quantify intestinal
cell differentiation, the number of EdU+ and DAPI+ intestinal nuclei were determined from
12 consecutive confocal sections imaged from the intestine. To quantify muscle cell
differentiation, the number of EdU+ and DAPI+ nuclei were determined from 4 to 9
consecutive confocal sections through the dorsal muscle layer of male parasites.
Tissue dissociation and EdU detection
Following an overnight pulse with 10 μM EdU, the heads and testes of adult male
S.
mansoni
were removed and the remaining tissue added to dissociation solution (Hanks
Balanced Salt Solution with 3.5 × Trypsin-EDTA (from 10× stock, Sigma-Aldrich, St.
Louis, MO)) and minced with a razor blade. These tissue fragments were incubated in ~4
mL of dissociation solution for 45–60 min at room temperature on a rocker and gentle
pipetting was used to break up large tissue fragments. This mixture was passed over two sets
of cell strainers (100 and 40 μm, BD, Franklin Lakes, New Jersey) and dissociated cells
were collected by centrifugation (250 × g for 5 m). Pelleted cells were fixed in 4%
formaldhyde in PBS for 30 min, spotted on Superfrost Plus microscope slides (Fisher
Scientific), permeabilized for 30 min with PBSTx, and EdU was detected as described above
with 10 μM Alexa Fluor 488 azide.
To quantify the ratio of EdU+ to total DAPI+ nuclei in RNAi knockdowns, 8 male parasites
were processed as above and tiled images of EdU and DAPI labeling were captured on a
Zeiss LSM 710 (Plan-Apochromat 20×/0.8). Numbers of EdU+ and DAPI+ nuclei were
quantified using the Image-based Tool for Counting Nuclei (ITCN) plugin for ImageJ35.
Collins et al. Page 5
Nature
. Author manuscript; available in PMC 2013 August 28.
NIH-PA Author Manuscript NIH-PA Author Manuscript NIH-PA Author Manuscript
γ-irradiation and transcriptional profiling
Parasites (D43 post-infection) were harvested from mice, suspended in Basch medium 169,
and exposed to 200 Gy of γ-irradiation using a Gammacell-220 Excel with a Co60 source
(Nordion, Ottawa, ON, Canada). Control parasites were mock irradiated. Parasites were
cultured in Basch Medium 169 and 48 hours post-irradiation males were separated from
female parasites using ethyl 3-aminobenzoate methanesulfonate. Following separation, the
head and testes of males were removed and purified total RNA was prepared from the
remaining tissue from pools of 14–18 parasites using Trizol (Invitrogen, Carlsbad, CA) and
DNase treatment (DNA-free RNA Kit, Zymo Research, Irvine, CA). Three independent
biological replicates were performed for both control and irradiated experimental groups.
Individually tagged libraries for RNAseq were prepared (TruSeq RNAseq Sample Prep Kit,
Illumina, San Diego, CA), pooled in a single lane, and 100 bp reads were generated using an
Illumina HiSeq2000. Library preparation and Illumina sequencing were performed at the
W.M. Keck Center for Comparative and Functional Genomics. The resulting reads were
mapped to the annotated
S. mansoni
genome6 (v5.0) and differences in gene expression
were determined using CLC Genomics Workbench (CLC bio, Aarhus, Denmark). Statistical
enrichment of Gene Ontology terms was determined in CLC Genomics Workbench using a
hyper geometric test that is similar to the GOstat test described in previous studies36. To
examine similarities between proteins encoded from irradiation-sensitive transcripts in
S.
mansoni
and genes expressed in planarian neoblasts, we compared our schistosome dataset
with both neoblast-enriched and “whole” transcriptomes14,15,18,19 using standalone
tBLASTn. Schistosome proteins sharing no similarity to translated planarian mRNAs (e-
value cut-off > 1e-5) were omitted from analysis. Assignment of whether protein pairs were
orthologous, homologous, paralogous, or unrelated was assessed manually on an individual
basis. Data and evidence supporting protein similarity is provided in Supplementary Table 1.
EdU/BrdU double labeling
Parasites labeled with 10 μM EdU and BrdU were fixed in Methacarn (6:3:1
methanol:chloroform:glacial acetic acid) or processed for in situ hybridization. Following a
45 min 2N HCl treatment, EdU was detected and parasites were processed for anti-BrdU
immunofluorescence (anti-BrdU 1:500, clone MoBU, Invitrogen). We observed no cross-
reactivity between this antibody and EdU.
To quantify the level of BrdU/EdU overlap and measure center-to-center distances between
nuclei, 3D confocal stacks from EdU/BrdU labeled animals were resampled to give isotropic
voxels, and subjected to Gaussian filtering and background-subtraction. Labeled nuclei were
segmented with Imaris (Bitplane Inc., South Winsor, CT) using parameters empirically
determined to minimize the need for manual corrections; typically, fewer than 5% of the
total nuclei required correction. The 3D coordinates of the nuclei were exported and
analyzed with MATLAB. Overlapping EdU and BrdU labeled nuclei were defined as nuclei
with center-to-center distances < 1 nuclear size (~4 um). Statistical analyses were performed
in Origin (OriginLab, Northampton, MA).
RNA interference
Although procedures have been previously described37,38, RNAi experiments with adult
parasites were based on methods optimized for schistosomula39. Briefly,
in vitro
cultured
parasites were soaked with 20–30 μg of dsRNA freshly added on days 1–3 and every 5–6
days thereafter. As a negative control, animals were soaked with dsRNA synthesized from
the
ccdB
and
camR
-containing insert of pJC53.229. dsRNA synthesis was performed as
previously described29. Sequences used to generate dsRNAs are provided in Supplementary
Fig. 9. To measure mRNA levels, total RNA from control and knockdown parasites (~8
male posterior somatic fragments) was reverse transcribed (iScript cDNA Synthesis Kit,
Collins et al. Page 6
Nature
. Author manuscript; available in PMC 2013 August 28.
NIH-PA Author Manuscript NIH-PA Author Manuscript NIH-PA Author Manuscript
Bio-Rad, Hercules, CA) and quantitative real time PCR was performed on an Applied
Biosystems Step One Plus instrument using GoTaq qPCR Master Mix with SYBR green
(Promega, Madison, WI). Transcript levels were normalized to the mRNA levels of
proteasome subunit beta type-4
(smp_056500). Relative quantities were calculated using the
ΔΔCt calculation in the Step One Plus software. Oligonucleotide primer sequences are
listed in Supplementary Table 3.
Supplementary Material
Refer to Web version on PubMed Central for supplementary material.
Acknowledgments
We thank Rachel Roberts-Galbraith, Melanie Issigonis, and Labib Rouhana for comments on the manuscript; Ryan
King for sharing the
Cathepsin B
plasmid and unpublished protocols; and Alvaro Hernandez for assistance with
Illumina sequencing. Schistosome-infected mice were provided by the NIAID Schistosomiasis Resource Center and
the Biomedical Research Institute (Rockville, MD) through NIAID Contract N01-A1-30026. This work was
supported by: NIH F32 HD062124 (J.J.C.) and NIH R21 AI099642 (P.A.N.). P.A.N. is an investigator of the
Howard Hughes Medical Institute.
References
1. Chitsulo L, Engels D, Montresor A, Savioli L. The global status of schistosomiasis and its control.
Acta Trop. 2000; 77:41–51. [PubMed: 10996119]
2. Basch, PF. Schistosomes: Development, Reproduction, and Host Relations. Oxford University
Press; 1991.
3. Wagner DE, Wang IE, Reddien PW. Clonogenic neoblasts are pluripotent adult stem cells that
underlie planarian regeneration. Science. 2011; 332:811–816. [PubMed: 21566185]
4. Newmark PA, Sánchez Alvarado A. Not your father's planarian: a classic model enters the era of
functional genomics. Nat Rev Genet. 2002; 3:210–219. [PubMed: 11972158]
5. Brehm K.
Echinococcus multilocularis
as an experimental model in stem cell research and
molecular host-parasite interaction. Parasitology. 2010; 137:537–555. [PubMed: 19961652]
6. Protasio AV, et al. A systematically improved high quality genome and transcriptome of the human
blood fluke
Schistosoma mansoni
. PLoS Negl Trop Dis. 2012; 6:e1455. [PubMed: 22253936]
7. Berriman M, et al. The genome of the blood fluke
Schistosoma mansoni
. Nature. 2009; 460:352–
358. [PubMed: 19606141]
8. Den Hollander JE, Erasmus DA.
Schistosoma mansoni
: DNA synthesis in males and females from
mixed and single-sex infections. Parasitology. 1984; 88(Pt 3):463–476. [PubMed: 6739132]
9. Nollen PM, Floyd RD, Kolzow RG, Deter DL. The timing of reproductive cell development and
movement in
Schistosoma mansoni
,
S. japonicum
, and
S. haematobium
, using techniques of
autoradiography and transplantation. J Parasitol. 1976; 62:227–231. [PubMed: 1263031]
10. Salic A, Mitchison TJ. A chemical method for fast and sensitive detection of DNA synthesis in
vivo. Proc Natl Acad Sci U S A. 2008; 105:2415–2420. [PubMed: 18272492]
11. Newmark PA, Sánchez Alvarado A. Bromodeoxyuridine specifically labels the regenerative stem
cells of planarians. Dev Biol. 2000; 220:142–153. [PubMed: 10753506]
12. Forsthoefel DJ, Park AE, Newmark PA. Stem cell-based growth, regeneration, and remodeling of
the planarian intestine. Dev Biol. 2011; 356:445–459. [PubMed: 21664348]
13. Baguñá J, Romero R. Quantitative analysis of cell types during growth, degrowth and regeneration
in the planarians
Dugesia mediterranea
and
Dugesia tigrina
. Hydrobiologia. 1981; 84:181–194.
14. Wagner DE, Ho JJ, Reddien PW. Genetic regulators of a pluripotent adult stem cell system in
planarians identified by RNAi and clonal analysis. Cell Stem Cell. 2012; 10:299–311. [PubMed:
22385657]
15. Solana J, et al. Defining the molecular profile of planarian pluripotent stem cells using a
combinatorial RNAseq, RNA interference and irradiation approach. Genome Biol. 2012; 13:R19.
[PubMed: 22439894]
Collins et al. Page 7
Nature
. Author manuscript; available in PMC 2013 August 28.
NIH-PA Author Manuscript NIH-PA Author Manuscript NIH-PA Author Manuscript
16. Eisenhoffer GT, Kang H, Sánchez Alvarado A. Molecular analysis of stem cells and their
descendants during cell turnover and regeneration in the planarian
Schmidtea mediterranea
. Cell
Stem Cell. 2008; 3:327–339. [PubMed: 18786419]
17. Caffrey CR, McKerrow JH, Salter JP, Sajid M. Blood 'n' guts: an update on schistosome digestive
peptidases. Trends Parasitol. 2004; 20:241–248. [PubMed: 15105025]
18. Onal P, et al. Gene expression of pluripotency determinants is conserved between mammalian and
planarian stem cells. EMBO J. 2012; 31:2755–2769. [PubMed: 22543868]
19. Labbe RM, et al. A comparative transcriptomic analysis reveals conserved features of stem cell
pluripotency in planarians and mammals. Stem Cells. 2012; 30:1734–1745. [PubMed: 22696458]
20. Pearson BJ, Sánchez Alvarado A. A planarian p53 homolog regulates proliferation and self-
renewal in adult stem cell lineages. Development. 2010; 137:213–221. [PubMed: 20040488]
21. Ogawa K, et al. Planarian fibroblast growth factor receptor homologs expressed in stem cells and
cephalic ganglions. Dev Growth Differ. 2002; 44:191–204. [PubMed: 12060069]
22. Rouhana L, Shibata N, Nishimura O, Agata K. Different requirements for conserved post-
transcriptional regulators in planarian regeneration and stem cell maintenance. Dev Biol. 2010;
341:429–443. [PubMed: 20230812]
23. Juliano C, Wessel G. Versatile germline genes. Science. 2010; 329:640–641. [PubMed: 20689009]
24. Skinner DE, et al. Vasa-Like DEAD-Box RNA Helicases of
Schistosoma mansoni
. PLoS Negl
Trop Dis. 2012; 6:e1686. [PubMed: 22720105]
25. Lanner F, Rossant J. The role of FGF/Erk signaling in pluripotent cells. Development. 2010;
137:3351–3360. [PubMed: 20876656]
26. Davies SJ, et al. Modulation of blood fluke development in the liver by hepatic CD4+
lymphocytes. Science. 2001; 294:1358–1361. [PubMed: 11701932]
27. Severinghaus AE. Sex Studies on
Schistosoma japonicum
. Quarterly Journal of Microscopical
Science. 1928; 71:653–702.
28. Shaw MK, Erasmus DA.
Schistosoma mansoni:
structural damage and tegumental repair after in
vivo treatment with praziquantel. Parasitology. 1987; 94(Pt 2):243–254. [PubMed: 3108831]
29. Collins JJ III, et al. Genome-Wide Analyses Reveal a Role for Peptide Hormones in Planarian
Germline Development. PLoS Biol. 2010; 8:e1000509. [PubMed: 20967238]
30. Collins JJ III, King RS, Cogswell A, Williams DL, Newmark PA. An atlas for
Schistosoma
mansoni
organs and life-cycle stages using cell type-specific markers and confocal microscopy.
PLoS Negl Trop Dis. 2011; 5:e1009. [PubMed: 21408085]
31. Lewis F. Schistosomiasis. Curr Protoc Immunol. 2001; Chapter 19(Unit 19):11.
32. Basch PF. Cultivation of
Schistosoma mansoni
in vitro. I. Establishment of cultures from cercariae
and development until pairing. J Parasitol. 1981; 67:179–185. [PubMed: 7241277]
33. Cogswell AA, Collins JJ III, Newmark PA, Williams DL. Whole mount in situ hybridization
methodology for
Schistosoma mansoni
. Mol Biochem Parasitol. 2011; 178:46–50. [PubMed:
21397637]
34. Neef AB, Luedtke NW. Dynamic metabolic labeling of DNA in vivo with arabinosyl nucleosides.
Proc Natl Acad Sci U S A. 2011; 108:20404–20409. [PubMed: 22143759]
35. Abramoff MD, Magelhaes PJ, Ram SJ. Image Processing with ImageJ. Biophotonics International.
2004; 11:36–42.
36. Falcon S, Gentleman R. Using GOstats to test gene lists for GO term association. Bioinformatics.
2007; 23:257–258. [PubMed: 17098774]
37. Skelly PJ, Da'dara A, Harn DA. Suppression of
cathepsin B
expression in
Schistosoma mansoni
by
RNA interference. Int J Parasitol. 2003; 33:363–369. [PubMed: 12705930]
38. Boyle JP, Wu XJ, Shoemaker CB, Yoshino TP. Using RNA interference to manipulate endogenous
gene expression in
Schistosoma mansoni
sporocysts. Mol Biochem Parasitol. 2003; 128:205–215.
[PubMed: 12742587]
39. Stefanic S, et al. RNA Interference in
Schistosoma mansoni
Schistosomula: Selectivity, Sensitivity
and Operation for Larger-Scale Screening. PLoS Negl Trop Dis. 2010; 4:e850. [PubMed:
20976050]
Collins et al. Page 8
Nature
. Author manuscript; available in PMC 2013 August 28.
NIH-PA Author Manuscript NIH-PA Author Manuscript NIH-PA Author Manuscript
40. McVeigh P, et al. Discovery of multiple neuropeptide families in the phylum Platyhelminthes. Int J
Parasitol. 2009; 39:1243–1252. [PubMed: 19361512]
41. Faghiri Z, et al. The tegument of the human parasitic worm
Schistosoma mansoni
as an excretory
organ: the surface aquaporin
SmAQP
is a lactate transporter. PLoS One. 2010; 5:e10451.
[PubMed: 20454673]
Collins et al. Page 9
Nature
. Author manuscript; available in PMC 2013 August 28.
NIH-PA Author Manuscript NIH-PA Author Manuscript NIH-PA Author Manuscript
Figure 1. Proliferation of somatic cells in adult schistosomes
a–b, EdU labeling in (a) male and (b) female parasites.
c–d, Distribution of mesenchymal PSCs in (c) male and (d) female parasites. Phalloidin
staining for actin shows male enteric and dorso-ventral muscles and female enteric and
uterine muscles.
e, Strategy to characterize PSC morphology.
f, The morphology of EdU and EdU+ cells. Arrowhead indicates a nucleolus.
g, FISH for
histone h2b
with EdU labeling. Arrowhead indicates a cytoplasmic projection.
(a–d, g) are confocal projections; (a–b) are derived from tiled stacks. Scale bars: (a–b) 500
μm, (c–d) 20 μm, (f–g) 5 μm.
Collins et al. Page 10
Nature
. Author manuscript; available in PMC 2013 August 28.
NIH-PA Author Manuscript NIH-PA Author Manuscript NIH-PA Author Manuscript
Figure 2. Transcriptional profiling identifies genes expressed in proliferative cells
a, EdU incorporation is abrogated at D3 following irradiation.
b, Strategy to identify PSC-expressed genes.
c, Volcano plot showing expression differences in control versus irradiated parasites. n = 3
for each group.
d, WISH for various transcripts in unirradiated and D5 post-irradiation parasites. n > 3
parasites.
e, EdU labeling and FISH for
Sm-fgfrA
. 1988/2000 EdU+ PSCs were
Sm-fgfrA
+ following
a 20–22 hour pulse (n = 20 male parasites). (a, e) are confocal projections. Scale bars: (a, e)
20 μm, (d) 100 μm.
Collins et al. Page 11
Nature
. Author manuscript; available in PMC 2013 August 28.
NIH-PA Author Manuscript NIH-PA Author Manuscript NIH-PA Author Manuscript
Figure 3. PSCs self-renew and differentiate
a, EdU-BrdU double labeling. Arrowheads, EdU+BrdU+ nuclei. Arrows, EdU+ “doublets”.
b, Percentage (±s.d.) EdU+ doublets (green) that are BrdU-BrdU (top), BrdU+-BrdU+
(middle, BrdU is magenta), or BrdU+-BrdU (Bottom). n = 21 parasites.
c, Strategy to monitor cellular differentiation.
d–g, EdU and sWGA labeling showing EdU+ cells in (d,e) male intestine or (f,g) dorsal
musculature at (d,f) D1 and (e,g) D7 following a pulse. (d,e) Insets, intestinal basal surface
(dashed lines) and lumen (green). Arrowheads, EdU+ (e) intestinal cells or (g) muscle cells.
Images are confocal projections. Scale bars: (a,d–g) 20 μm (b) 5 μm.
Collins et al. Page 12
Nature
. Author manuscript; available in PMC 2013 August 28.
NIH-PA Author Manuscript NIH-PA Author Manuscript NIH-PA Author Manuscript
Figure 4. Sm-fgfrA is required for the maintenance of somatic stem cells
a, EdU labeling and DIC images in control and
Sm-fgfrA(RNAi)
at RNAi D17.
b, Percentage EdU+ nuclei/total nuclei in dissociated tissues from control(RNAi) (n = 3002
nuclei) and
Sm-fgfrA(RNAi)
(n = 3642 nuclei) parasites. Error bars, 95% confidence
intervals, p < 0.0001 χ2.
c, WISH for
histone h2b
(top row) and
nanos2
(bottom row) transcripts in control (left
column) versus
Sm-fgfrA(RNAi)
(right column) parasites at RNAi D20-21. n > 5 parasites/
experiment. Scale bars: (a) 100 μm (c) 200 μm.
Collins et al. Page 13
Nature
. Author manuscript; available in PMC 2013 August 28.
NIH-PA Author Manuscript NIH-PA Author Manuscript NIH-PA Author Manuscript

Supplementary resource (1)

... Adult or somatic SCs are a type of cell that can alter or differentiate into any type of specialized cell to repair and rebuild tissue in all regions of your body. They have been isolated from a wide variety of tissues, including bone marrow, fat tissue and within the brain their role is supposed to be one of maintaining homeostasis and preventing tissue dysfunction (Collins et al., 2013). Some studies in recent years have emphasized their application for various therapeutic purposes such as degenerative disorders and traumas. ...
Chapter
Zoology: Advancements and Research Trends is a comprehensive compilation of cutting-edge research and insights in the field of Zoology. This book has been meticulously curated to serve as an invaluable resource for students, researchers, and professionals who are keen to explore the latest advancements and emerging trends in various sub-disciplines of Zoology, including toxicology, pharmacology, and general zoology. As an Assistant Professor in the Department of Zoology, Wildlife, and Fisheries at the University of Agriculture, Faisalabad, I, Dr. Muhammad Umar Ijaz, have dedicated over a decade to advancing our understanding of animal physiology, cell biology, toxicology and pharmacology. My extensive experience in these fields, coupled with my broader expertise in general zoology, has provided me with a profound appreciation for the complex interplay between living organisms and their environments. This book reflects my commitment to bringing together a diverse array of topics that highlight the breadth and depth of contemporary zoological research. Zoology: Advancements and Research Trends is not just a collection of research topics but a reflection of the collaborative spirit that drives scientific progress. The diverse expertise of the contributing authors ensures that this book offers a well-rounded perspective on the current trends and future directions in Zoology. I hope that this book will serve as a valuable reference for those in academia and industry, providing insights that will inspire future research and contribute to the ongoing dialogue in the field. It is my sincere belief that the knowledge shared within these pages will help shape the future of Zoology, ultimately contributing to the betterment of our understanding and stewardship of the natural world. I would like to express my gratitude to all the contributors, reviewers, and colleagues whose efforts have made this book possible. I also extend my thanks to the students and researchers who continue to push the boundaries of what we know and what we can achieve in this dynamic field.
... A further interesting question raised by our study are the mechanistic causes of the frequent genome rearrangements in Schmidtea. Both free-living and parasitic flatworms survive gammairradiation well beyond lethal doses in vertebrates [81][82][83][84] , which implies the existence of efficient double-strand break repair pathways that are also known to mediate Robertsonian translocations [85][86][87] . The enrichment of LTR/Gypsy and LINE/R2 elements near the synteny breakpoints might indicate the role of retrotransposons as templates for strand invasion during the repair of double-strand breaks. ...
Article
Full-text available
The planarian Schmidtea mediterranea is being studied as a model species for regeneration, but the assembly of planarian genomes remains challenging. Here, we report a high-quality haplotype-phased, chromosome-scale genome assembly of the sexual S2 strain of S. mediterranea and high-quality chromosome-scale assemblies of its three close relatives, S. polychroa, S. nova, and S. lugubris. Using hybrid gene annotations and optimized ATAC-seq and ChIP-seq protocols for regulatory element annotation, we provide valuable genome resources for the planarian research community and a first comparative perspective on planarian genome evolution. Our analyses reveal substantial divergence in protein-coding sequences and regulatory regions but considerable conservation within promoter and enhancer annotations. We also find frequent retrotransposon-associated chromosomal inversions and interchromosomal translocations within the genus Schmidtea and, remarkably, independent and nearly complete losses of ancestral metazoan synteny in Schmidtea and two other flatworm groups. Overall, our results suggest that platyhelminth genomes can evolve without syntenic constraints.
... In terms of insight into the life cycle of schistosomes, two stem cell populations have been identified as crucial for the completion of the schistosome life cycle (Wendt and Collins 2016;You et al. 2021a). Germinal cells are totipotent stem cells that are involved in the clonal expansion of the asexual stage sporocysts in molluscan hosts and neoblasts, which are pluripotent stem cells identified in adult worms (Collins et al. 2013). Two distinct types of stem cell populations (somatic stem cells and germinal cells) have been identified in schistosomula; these two cell types are responsible for somatic tissue differentiation and the production of germ cells (sperm and oocytes), respectively, in adult schistosomes in the definitive host. ...
Article
Full-text available
Schistosomiasis is a major cause of morbidity in the world and almost 800 million people worldwide are at risk for schistosomiasis; it is second only to malaria as a major infectious disease. Globally, it is estimated that the disease affects more than 250 million people in 78 countries of the world and is responsible for some 280,000–500,000 deaths each year. The three major schistosomes infecting humans are Schistosoma mansoni, S. japonicum, and S. haematobium. This chapter covers a wide range of aspects of schistosomiasis, including basic biology of the parasites, epidemiology, immunopathology, treatment, control, vaccines, and genomics/proteomics. In this chapter, the reader will understand the significant toll this disease takes in terms of mortality and morbidity. A description of the various life stages of schistosomes is presented, which will be informative for both those unfamiliar with the disease and experienced scientists. Clinical and public health aspects are addressed that cover acute and chronic disease, diagnosis, current treatment regimens and alternative drugs, and schistosomiasis control programs. A brief overview of genomics and proteomics is included that details recent advances in the field that will help scientists investigate the molecular biology of schistosomes. The reader will take away an appreciation for general aspects of schistosomiasis and the current research advances.
... In Schistosoma mansoni, nanos role in the maintenance, proliferation and develop-. Available at: http://ijpa.tums.ac.ir ment of the germinative cells has been established as some workers showed that nanos is expressed in germinal cells of sporocysts and the somatic stem cell as well as the vitelline cells in the adult schistosomes (24,25,26). It has been shown that nanos is essential for the reproductive organ development of both male and female S. japonicum particularly the testes, vitelline glans and ovaries (27,28). ...
Article
Full-text available
Background We aimed to evaluate the differential expression of nanos and ago genes in the protoscoleces, germinal layer, the neck, and the sucker regions of adult Echinococcus granulosus. Methods The study was conducted in 2018 at the Research Center for Hydatid Disease in Iran, Kerman University of Medical Sciences, Kerman, Iran. In the present study E. granulosus protoscoleces were cultured in a di-phasic medium to obtain strobilated worms. The strobilated worms were harvested and using a sterile razor blade, the neck region was separated. In the molecular study the neck sections were compared with the tissues derived from the suckers from the same worm. The primers were specifically designed for RT-qPCR on nanos and ago. The germinative cells were isolated from the cyst germinal layer and cultured in DMEM for further molecular studies. The Immunohisto-chemical profile was designed to explore the nature of nanos protein in the strobilated worms. Differences between and within groups were statistically assessed relative to the protoscoleces. Results An increasing nanos gene expressions were found in sucker, neck, cells and germinal layer in comparison to the protoscoleces. The expression of ago gene was decreased in sucker, cell and germinal layer, and increased in the neck region in comparison to the protoscoleces. The results showed that both genes were expressed in all developmental stages of E. granulosus. Conclusion nanos and ago genes were differentially expressed at different developmental stages of E. granulosus and may contribute to differentiation of the parasite.
Article
CRISPR genome editing is actively used for schistosomes and other flukes. The ability to genetically manipulate these flatworms enables deeper investigation of their (patho)biological nature. CRISPR gene knockout (KO) demonstrated that a liver fluke growth mediator contributes to disease progression. Genome safe harbor sites have been predicted in Schistosoma mansoni and targeted for transgene insertion. CRISPR-based diagnosis has been demonstrated for infection with schistosomes and Opisthorchis viverrini. This review charts the progress, and the state of play, and posits salient questions for the field to address. Derivation of heritably transgenic loss-of-function or gain-of-function lines is the next milestone.
Article
Full-text available
Schistosomes are blood-dwelling parasitic flatworms that rely on a syncytial surface coat, known as the tegument, for long-term survival and immune evasion in the blood of their human hosts. Previous studies have shown that cells within the tegumental syncytium are perpetually turned over and renewed by somatic stem cells called neoblasts. Yet, little is known about this renewal process on a molecular level. Here, we characterized a Kruppel-like factor 4 ( klf4 ) using a combination of bulk and single cell RNAseq approaches and demonstrate that klf4 is essential for the maintenance of a specific tegumental lineage, resulting in the loss of a subpopulation of molecularly-unique tegument cells. Thus, klf4 is critical for maintaining the balance between different tegumental progenitor pools, thereby fine-tuning the molecular composition of the mature tegument. Understanding these distinct tegumental cell populations is expected to provide insights into parasite defense mechanisms and suggest new avenues for therapeutics.
Article
Full-text available
Schistosomes are parasitic flatworms responsible for the neglected tropical disease schistosomiasis, causing devastating morbidity and mortality in the developing world. The parasites are protected by a skin-like tegument, and maintenance of this tegument is controlled by a schistosome ortholog of the tumor suppressor TP53. To understand mechanistically how p53–1 controls tegument production, we identified a cyclin dependent kinase inhibitor homolog (cki) that was co-expressed with p53–1. RNA interference of cki resulted in a hyperproliferation phenotype, that, in combination with p53–1 RNA interference yielded abundant tumor-like growths, indicating that cki and p53–1 are bona fide tumor suppressors in Schistosoma mansoni. Interestingly, cki homologs are widely present throughout parasitic flatworms but evidently absent from their free-living ancestors, suggesting this cki homolog came from an ancient horizontal gene transfer event. This in turn implies that the evolution of parasitism in flatworms may have been aided by a highly unusual means of metazoan genetic inheritance.
Article
Full-text available
Flatworms are known for their remarkable regenerative ability, one which depends on totipotent cells known as germinative cells in cestodes. Depletion of germinative cells with hydroxyurea (HU) affects the regeneration of the parasite. Here, we studied the reduction and recovery of germinative cells in T. crassiceps cysticerci after HU treatment (25 mM and 40 mM of HU for 6 days) through in vitro assays. Viability and morphological changes were evaluated. The recovery of cysticerci’s mobility and morphology was evaluated at 3 and 6 days, after 6 days of treatment. The number of proliferative cells was evaluated using EdU. Our results show morphological changes in the size, shape, and number of evaginated cysticerci at the 40 mM dose. The mobility of cysticerci was lower after 6 days of HU treatment at both concentrations. On days 3 and 6 of recovery after 25 mM of HU treatment, a partial recovery of the proliferative cells was observed. Proteomic and Gene Ontology analyses identified modifications in protein groups related to DNA binding, DNA damage, glycolytic enzymes, cytoskeleton, skeletal muscle, and RNA binding.
Article
Full-text available
Schistosoma mansoniis responsible for the neglected tropical disease schistosomiasis that affects 210 million people in 76countries. Here we present analysis of the 363 megabase nuclear genome of the blood fluke. It encodes at least 11,809 genes,with an unusual intron size distribution, and new families of micro-exon genes that undergo frequent alternative splicing. Asthe first sequenced flatworm, and a representative of the Lophotrochozoa, it offers insights into early events in the evolutionof the animals, including the development of a body pattern with bilateral symmetry, and the development of tissues intoorgans. Our analysis has been informed by the need to find new drug targets. The deficits in lipid metabolism that makeschistosomes dependent on the host are revealed, and the identification of membrane receptors, ion channels and more than300 proteases provide new insights into the biology of the life cycle and new targets. Bioinformatics approaches haveidentified metabolic chokepoints, and a chemogenomic screen has pinpointed schistosome proteins for which existing drugsmay be active. The information generated provides an invaluable resource for the research community to develop muchneeded new control tools for the treatment and eradication of this important and neglected disease.
Article
Full-text available
Genome sequences are available for the human blood flukes, Schistosoma japonicum, S. mansoni and S. haematobium. Functional genomic approaches could aid in identifying the role and importance of these newly described schistosome genes. Transgenesis is established for functional genomics in model species, which can lead to gain- or loss-of-functions, facilitate vector-based RNA interference, and represents an effective forward genetics tool for insertional mutagenesis screens. Progress toward routine transgenesis in schistosomes might be expedited if germ cells could be reliably localized in cultured schistosomes. Vasa, a member of the ATP-dependent DEAD-box RNA helicase family, is a prototypic marker of primordial germ cells and the germ line in the Metazoa. Using bioinformatics, 33 putative DEAD-box RNA helicases exhibiting conserved motifs that characterize helicases of this family were identified in the S. mansoni genome. Moreover, three of the helicases exhibited vasa-like sequences; phylogenetic analysis confirmed the three vasa-like genes-termed Smvlg1, Smvlg2, and Smvlg3-were members of the Vasa/PL10 DEAD-box subfamily. Transcripts encoding Smvlg1, Smvlg2, and Smvlg3 were cloned from cDNAs from mixed sex adult worms, and quantitative real time PCR revealed their presence in developmental stages of S. mansoni with elevated expression in sporocysts, adult females, eggs, and miracidia, with strikingly high expression in the undeveloped egg. Whole mount in situ hybridization (WISH) analysis revealed that Smvlg1, Smvlg2 and Smvlg3 were transcribed in the posterior ovary where the oocytes mature. Germ cell specific expression of schistosome vasa-like genes should provide an informative landmark for germ line transgenesis of schistosomes, etiologic agents of major neglected tropical diseases.
Article
Full-text available
A method of tissue maceration (dissociation) of planarian tissues into single cells was used to characterize the basic cell types in the planarians Dugesia mediterranea and Dugesia tigrina, and to determine the total cell number and distribution of cell types during growth, degrowth and regeneration. Using this method, 13 basic cell types have been determined for both species. The total number of cells increases with body length and volume whereas the distribution of cell types is only slightly affected. Growth and degrowth occur mainly through changes in total cell number leaving cell distribution only moderately affected. During regeneration, an increase in neoblast density in the blastema followed later on by increases in nerve cells are the more significant changes detected. These results are discussed in relation to mechanisms of cell renewal, blastema formation and maintenance of tissue polarity.
Article
The strong regenerative capacity of planarians is considered to reside in the totipotent somatic stem cell called the 'neoblast'. However, the signal systems regulating the differentiation/growth/migration of stem cells remain unclear. The fibroblast growth factor (FGF)/FGF receptor (FGFR) system is thought to mediate various developmental events in both vertebrates and invertebrates. We examined the molecular structures and expression of DjFGFR1 and DjFGFR2 , two planarian genes closely related to other animal FGFR genes. DjFGFR1 and DjFGFR2 proteins contain three and two immunoglobulin-like domains, respectively, in the extracellular region and a split tyrosine kinase domain in the intracellular region. Expression of DjFGFR1 and DjFGFR2 was observed in the cephalic ganglion and mesenchymal space in intact planarians. In regenerating planarians, accumulation of DjFGFR1 -expressing cells was observed in the blastema and in fragments regenerating either a pharynx or a brain. In X-ray-irradiated planarians, which had lost regenerative capacity, the number of DjFGFR1 -expressing cells in the mesenchymal space decreased markedly. These results suggest that the DjFGFR1 protein may be involved in the signal systems controlling such aspects of planarian regeneration as differentiation/growth/migration of stem cells.
Article
With Plates 43-6 and 1 Text-figure. FOREWORD. THE experiments outlined in this paper were begun at the suggestion of the writer in 1924 in the laboratories of biology and of parasitology of the Peking Union Medical College, Rockefeller Foundation. The problem was begun with the hearty co-operation of Dr. C. U. Lee, who directed most of the mammalian infections, and who personally autopsied about one-half of the infected hamsters in the recovery of the flukes. The records of these autopsies are not included in this paper, although they are preserved as valuable data. Dr. Lee's medical knowledge was here an invaluable aid, for it was essential to note accurately the condition of the host, and especially the cause of death when the hamster died before it was planned to sacrifice it. The writer is furthermore indebted to Dr. Lee for the later preservation of material with special fixatives, without which certain cytological features could not have been deter-mined. This material will be described more fully at a later date. A large measure of appreciation is due to Dr. Henry E. Meleney, who, with the kind permission of Dr. E. C. Faust, Professor of Parasitology, absent on leave, placed at our disposal the facilities of the laboratories of this department, lent every assistance, and gave many valuable suggestions. During a part of the trying summer period when the writer's duties called him from Peking, Dr. Meleney personally super-vised the care of the infected animals, examining them daily and performing such autopsies as were necessary.
Article
When do germ cells establish their separate, independent identity during animal development?
Article
Abramoff, M.D., Magelhaes, P.J., Ram, S.J. "Image Processing with ImageJ". Biophotonics International, volume 11, issue 7, pp. 36-42, 2004.
Article
Many long-lived species of animals require the function of adult stem cells throughout their lives. However, the transcriptomes of stem cells in invertebrates and vertebrates have not been compared, and consequently, ancestral regulatory circuits that control stem cell populations remain poorly defined. In this study, we have used data from high-throughput RNA sequencing to compare the transcriptomes of pluripotent adult stem cells from planarians with the transcriptomes of human and mouse pluripotent embryonic stem cells. From a stringently defined set of 4,432 orthologs shared between planarians, mice and humans, we identified 123 conserved genes that are ≥5-fold differentially expressed in stem cells from all three species. Guided by this gene set, we used RNAi screening in adult planarians to discover novel stem cell regulators, which we found to affect the stem cell-associated functions of tissue homeostasis, regeneration, and stem cell maintenance. Examples of genes that disrupted these processes included the orthologs of TBL3, PSD12, TTC27, and RACK1. From these analyses, we concluded that by comparing stem cell transcriptomes from diverse species, it is possible to uncover conserved factors that function in stem cell biology. These results provide insights into which genes comprised the ancestral circuitry underlying the control of stem cell self-renewal and pluripotency.