Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 22
Filter
Add more filters











Publication year range
1.
bioRxiv ; 2024 Sep 03.
Article in English | MEDLINE | ID: mdl-39282438

ABSTRACT

The Transient Receptor Potential superfamily of proteins (TRPs) form cation channels that are abundant in animal sensory systems. Amongst TRPs, the Melastatin-related subfamily (TRPMs) is composed of members that respond to temperature, pH, sex hormones, and various other stimuli. Some TRPMs exhibit enriched expression in gonads of vertebrate and invertebrate species, but their contributions to germline development remain to be determined. We identified twenty-one potential TRPMs in the planarian flatworm Schmidtea mediterranea and analyzed their anatomical distribution of expression by whole-mount in situ hybridization. Enriched expression of two TRPMs ( Smed-TRPM-c and Smed-TRPM-l ) was detected in testis, whereas eight TRPM genes had detectable expression in patterns representative of neuronal and/or sensory cell types. Functional analysis of TRPM homologs by RNA-interference (RNAi) revealed that disruption of Smed-TRPM-c expression results in reduced sperm development, indicating a role for this receptor in supporting spermatogenesis. Smed-TRPM-l RNAi did not result in a detectable phenotype, but it increased sperm development deficiencies when combined with Smed-TRPM-c RNAi. Fluorescence in situ hybridization revealed expression of Smed-TRPM-c in early spermatogenic cells within testes, suggesting cell-autonomous regulatory functions in germ cells for this gene. In addition, Smed-TRPM-c RNAi resulted in reduced numbers of presumptive germline stem cell clusters in asexual planarians, suggesting that Smed-TRPM-c supports establishment, maintenance, and/or expansion of spermatogonial germline stem cells. While further research is needed to identify the factors that trigger Smed-TRPM-c activity, these findings reveal one of few known examples for TRPM function in direct regulation of sperm development.

2.
Mol Biol Evol ; 40(6)2023 06 01.
Article in English | MEDLINE | ID: mdl-37288606

ABSTRACT

Differential regulation of gene expression has produced the astonishing diversity of life on Earth. Understanding the origin and evolution of mechanistic innovations for control of gene expression is therefore integral to evolutionary and developmental biology. Cytoplasmic polyadenylation is the biochemical extension of polyadenosine at the 3'-end of cytoplasmic mRNAs. This process regulates the translation of specific maternal transcripts and is mediated by the Cytoplasmic Polyadenylation Element-Binding Protein family (CPEBs). Genes that code for CPEBs are amongst a very few that are present in animals but missing in nonanimal lineages. Whether cytoplasmic polyadenylation is present in non-bilaterian animals (i.e., sponges, ctenophores, placozoans, and cnidarians) remains unknown. We have conducted phylogenetic analyses of CPEBs, and our results show that CPEB1 and CPEB2 subfamilies originated in the animal stem lineage. Our assessment of expression in the sea anemone, Nematostella vectensis (Cnidaria), and the comb jelly, Mnemiopsis leidyi (Ctenophora), demonstrates that maternal expression of CPEB1 and the catalytic subunit of the cytoplasmic polyadenylation machinery (GLD2) is an ancient feature that is conserved across animals. Furthermore, our measurements of poly(A)-tail elongation reveal that key targets of cytoplasmic polyadenylation are shared between vertebrates, cnidarians, and ctenophores, indicating that this mechanism orchestrates a regulatory network that is conserved throughout animal evolution. We postulate that cytoplasmic polyadenylation through CPEBs was a fundamental innovation that contributed to animal evolution from unicellular life.


Subject(s)
Ctenophora , Sea Anemones , Animals , Phylogeny , Polyadenylation , Ctenophora/genetics , Sea Anemones/genetics
3.
Methods Mol Biol ; 2364: 199-216, 2022.
Article in English | MEDLINE | ID: mdl-34542855

ABSTRACT

Spermatogenesis is one of the most dramatic cellular differentiation events observed in animals. In particular, spermiogenesis (the final stage of spermatogenesis) involves extensive shedding of cytoplasmic organelles, dramatic nuclear rearrangements, and assembly of long flagellar structures. In planarian flatworms, the spherical nucleus present in round spermatids elongates to produce the filamentous nucleus of mature sperm. Newly formed cortical microtubules participate in cytoskeletal rearrangements observed during spermiogenesis and remain present in sperm. In addition, a pair of flagella assemble at one end of each spermatid in a process that likely involves de novo formation of centrioles. This chapter includes a brief introduction to planarian spermatogenesis and current tools for the analysis of molecular players in this process. Step-by-step protocols for isolating and imaging spermatogenic cells are provided with enough detail to be carried out by newcomers to the field who would like to study this unique organism in the laboratory.


Subject(s)
Planarians , Animals , Cell Nucleus , Male , Microtubules , Morphogenesis , Spermatids , Spermatogenesis , Spermatozoa
4.
Front Cell Dev Biol ; 9: 777951, 2021.
Article in English | MEDLINE | ID: mdl-34901022

ABSTRACT

Detection of chemical stimuli is crucial for living systems and also contributes to quality of life in humans. Since loss of olfaction becomes more prevalent with aging, longer life expectancies have fueled interest in understanding the molecular mechanisms behind the development and maintenance of chemical sensing. Planarian flatworms possess an unsurpassed ability for stem cell-driven regeneration that allows them to restore any damaged or removed part of their bodies. This includes anteriorly-positioned lateral flaps known as auricles, which have long been thought to play a central role in chemotaxis. The contribution of auricles to the detection of positive chemical stimuli was tested in this study using Girardia dorotocephala, a North American planarian species known for its morphologically prominent auricles. Behavioral experiments staged under laboratory conditions revealed that removal of auricles by amputation leads to a significant decrease in the ability of planarians to find food. However, full chemotactic capacity is observed as early as 2 days post-amputation, which is days prior from restoration of auricle morphology, but correlative with accumulation of ciliated cells in the position of auricle regeneration. Planarians subjected to x-ray irradiation prior to auricle amputation were unable to restore auricle morphology, but were still able to restore chemotactic capacity. These results indicate that although regeneration of auricle morphology requires stem cells, some restoration of chemotactic ability can still be achieved in the absence of normal auricle morphology, corroborating with the initial observation that chemotactic success is reestablished 2-days post-amputation in our assays. Transcriptome profiles of excised auricles were obtained to facilitate molecular characterization of these structures, as well as the identification of genes that contribute to chemotaxis and auricle development. A significant overlap was found between genes with preferential expression in auricles of G. dorotocephala and genes with reduced expression upon SoxB1 knockdown in Schmidtea mediterranea, suggesting that SoxB1 has a conserved role in regulating auricle development and function. Models that distinguish between possible contributions to chemotactic behavior obtained from cellular composition, as compared to anatomical morphology of the auricles, are discussed.

5.
Dev Biol ; 477: 191-204, 2021 09.
Article in English | MEDLINE | ID: mdl-34090925

ABSTRACT

Development of sperm requires microtubule-based movements that drive assembly of a compact head and flagellated tails. Much is known about how flagella are built given their shared molecular core with motile cilia, but less is known about the mechanisms that shape the sperm head. The Kinesin Superfamily Protein 3A (KIF3A) pairs off with a second motor protein (KIF3B) and the Kinesin Associated Protein 3 (KAP3) to form Heterotrimeric Kinesin II. This complex drives intraflagellar transport (IFT) along microtubules during ciliary assembly. We show that KIF3A and KAP3 orthologs in Schmidtea mediterranea are required for axonemal assembly and nuclear elongation during spermiogenesis. Expression of Smed-KAP3 is enriched during planarian spermatogenesis with transcript abundance peaking in spermatocyte and spermatid cells. Disruption of Smed-kif3A or Smed-KAP3 expression by RNA-interference results in loss of spermatozoa and accumulation of unelongated spermatids. Confocal microscopy of planarian testis lobes stained with alpha-tubulin antibodies revealed that spermatids with disrupted Kinesin II function fail to assemble flagella, and visualization with 4',6-diamidino-2-phenylindole (DAPI) revealed reduced nuclear elongation. Disruption of Smed-kif3A or Smed-KAP3 expression also resulted in edema, reduced locomotion, and loss of epidermal cilia, which corroborates with somatic phenotypes previously reported for Smed-kif3B. These findings demonstrate that heterotrimeric Kinesin II drives assembly of cilia and flagella, as well as rearrangements of nuclear morphology in developing sperm. Prolonged activity of heterotrimeric Kinesin II in manchette-like structures with extended presence during spermiogenesis is hypothesized to result in the exaggerated nuclear elongation observed in sperm of turbellarians and other lophotrochozoans.


Subject(s)
Kinesins/physiology , Planarians/cytology , Sperm Tail/physiology , Spermatogenesis/physiology , Animals , Cell Nucleus/ultrastructure , Cytoskeletal Proteins/physiology , Gene Knockdown Techniques , Kinesins/chemistry , Kinesins/genetics , Male , RNA Interference , Sperm Head/ultrastructure , Sperm Tail/ultrastructure
6.
Dev Growth Differ ; 62(6): 423-437, 2020 Aug.
Article in English | MEDLINE | ID: mdl-32359074

ABSTRACT

Motile cilia propel directed cell movements and sweep fluids across the surface of tissues. Orthologs of Dynein Assembly Factor with WD Repeat Domains 1 (DAW1) support normal ciliary beating by enhancing delivery of dynein complexes to axonemal microtubules. DAW1 mutations in vertebrates result in multiple developmental abnormalities and early or prenatal lethality, complicating functional assessment of DAW1 in adult structures. Planarian flatworms maintain cellular homeostasis and regenerate through differentiation of adult pluripotent stem cells, and systemic RNA-interference (RNAi) can be induced to analyze gene function at any point after birth. A single ortholog of DAW1 was identified in the genome of the planarian Schmidtea mediterranea (Smed-daw1). Smed-DAW1 is composed of eight WD repeats, which are 55% identical to the founding member of this protein family (Chlamydomonas reinhardtii ODA16) and 58% identical to human DAW1. Smed-daw1 is expressed in the planarian epidermis, protonephridial excretory system, and testes, all of which contain cells functionally dependent on motile cilia. Smed-daw1 RNAi resulted in locomotion defects and edema, which are phenotypes characteristic of multiciliated epidermis and protonephridial dysfunction, respectively. Changes in abundance or length of motile cilia were not observed at the onset of phenotypic manifestations upon Smed-daw1 RNAi, corroborating with studies showing that DAW-1 loss of function leads to aberrant movement of motile cilia in other organisms, rather than loss of cilia per se. However, extended RNAi treatments did result in shorter epidermal cilia and decreased abundance of ciliated protonephridia, suggesting that Smed-daw1 is required for homeostatic maintenance of these structures in flatworms.


Subject(s)
Cilia/metabolism , Dyneins/metabolism , Planarians/cytology , Planarians/metabolism , WD40 Repeats , Animals , Cilia/genetics , Dyneins/genetics , Planarians/genetics , WD40 Repeats/genetics
7.
Mol Biol Cell ; 30(17): 2155-2170, 2019 08 01.
Article in English | MEDLINE | ID: mdl-31141462

ABSTRACT

Cilia are microtubule-based structures that protrude from the apical surface of cells to mediate motility, transport, intracellular signaling, and environmental sensing. Tau tubulin kinases (TTBKs) destabilize microtubules by phosphorylating microtubule-associated proteins (MAPs) of the MAP2/Tau family, but also contribute to the assembly of primary cilia during embryogenesis. Expression of TTBKs is enriched in testicular tissue, but their relevance to reproductive processes is unknown. We identified six TTBK homologues in the genome of the planarian Schmidtea mediterranea (Smed-TTBK-a, -b, -c, -d, -e, and -f), all of which are preferentially expressed in testes. Inhibition of TTBK paralogues by RNA interference (RNAi) revealed a specific requirement for Smed-TTBK-d in postmeiotic regulation of spermatogenesis. Disrupting expression of Smed-TTBK-d results in loss of spermatozoa, but not spermatids. In the soma, Smed-TTBK-d RNAi impaired the function of multiciliated epidermal cells in propelling planarian movement, as well as the osmoregulatory function of protonephridia. Decreased density and structural defects of motile cilia were observed in the epidermis of Smed-TTBK-d(RNAi) by phase contrast, immunofluorescence, and transmission electron microscopy. Altogether, these results demonstrate that members of the TTBK family of proteins are postmeiotic regulators of sperm development and also contribute to the formation of motile cilia in the soma.


Subject(s)
Cilia/physiology , Protein Serine-Threonine Kinases/metabolism , Spermatogenesis/physiology , Animals , Cilia/metabolism , Male , Microtubule-Associated Proteins/metabolism , Phosphorylation , Planarians/genetics , Planarians/metabolism , Protein Serine-Threonine Kinases/physiology , RNA Interference , Regeneration/genetics , Signal Transduction , Tubulin/genetics
8.
Dev Biol ; 433(2): 433-447, 2018 01 15.
Article in English | MEDLINE | ID: mdl-28774726

ABSTRACT

Planarian flatworms are popular models for the study of regeneration and stem cell biology in vivo. Technical advances and increased availability of genetic information have fueled the discovery of molecules responsible for stem cell pluripotency and regeneration in flatworms. Unfortunately, most of the planarian research performed worldwide utilizes species that are not natural habitants of North America, which limits their availability to newcomer laboratories and impedes their distribution for educational activities. In order to circumvent these limitations and increase the genetic information available for comparative studies, we sequenced the transcriptome of Girardia dorotocephala, a planarian species pandemic and commercially available in North America. A total of 254,802,670 paired sequence reads were obtained from RNA extracted from intact individuals, regenerating fragments, as well as freshly excised auricles of a clonal line of G. dorotocephala (MA-C2), and used for de novo assembly of its transcriptome. The resulting transcriptome draft was validated through functional analysis of genetic markers of stem cells and their progeny in G. dorotocephala. Akin to orthologs in other planarian species, G. dorotocephala Piwi1 (GdPiwi1) was found to be a robust marker of the planarian stem cell population and GdPiwi2 an essential component for stem cell-driven regeneration. Identification of G. dorotocephala homologs of the early stem cell descendent marker PROG-1 revealed a family of lysine-rich proteins expressed during epithelial cell differentiation. Sequences from the MA-C2 transcriptome were found to be 98-99% identical to nucleotide sequences from G. dorotocephala populations with different chromosomal number, demonstrating strong conservation regardless of karyotype evolution. Altogether, this work establishes G. dorotocephala as a viable and accessible option for analysis of gene function in North America.


Subject(s)
Argonaute Proteins/genetics , Genes, Helminth , Helminth Proteins/genetics , Planarians/genetics , Stem Cells/cytology , Transcriptome , Animals , Argonaute Proteins/physiology , Biomarkers , Cloning, Organism , Helminth Proteins/biosynthesis , Homeostasis/genetics , Multigene Family , RNA Interference , RNA, Double-Stranded/administration & dosage , RNA, Double-Stranded/genetics , RNA, Messenger/biosynthesis , RNA, Messenger/genetics , Regeneration/genetics , Reproduction, Asexual , Sequence Analysis, RNA
9.
Mol Reprod Dev ; 84(12): 1271-1284, 2017 12.
Article in English | MEDLINE | ID: mdl-29095551

ABSTRACT

Chaperonin-containing Tail-less complex polypeptide 1 (CCT) is a highly conserved, hetero-oligomeric complex that ensures proper folding of actin, tubulin, and regulators of mitosis. Eight subunits (CCT1-8) make up this complex, and every subunit has a homolog expressed in the testes and somatic tissue of the planarian flatworm Schmidtea mediterranea. Gene duplications of four subunits in the genomes of S. mediterranea and other planarian flatworms created paralogs to CCT1, CCT3, CCT4, and CCT8 that are expressed exclusively in the testes. Functional analyses revealed that each CCT subunit expressed in the S. mediterranea soma is essential for homeostatic integrity and survival, whereas sperm elongation defects were observed upon knockdown of each individual testis-specific paralog (Smed-cct1B; Smed-cct3B; Smed-cct4A; and Smed-cct8B), regardless of potential redundancy with paralogs expressed in both testes and soma (Smed-cct1A; Smed-cct3A; Smed-cct4B; and Smed-cct8A). Yet, no detriment was observed in the number of adult somatic stem cells (neoblasts) that maintain differentiated tissue in planarians. Thus, expression of all eight CCT subunits is required to execute the essential functions of the CCT complex. Furthermore, expression of the somatic paralogs in planarian testes is not sufficient to complete spermatogenesis when testis-specific paralogs are knocked down, suggesting that the evolution of chaperonin subunits may drive changes in the development of sperm structure and that correct CCT subunit stoichiometry is crucial for spermiogenesis.


Subject(s)
Chaperonin Containing TCP-1/metabolism , Helminth Proteins/metabolism , Planarians/metabolism , Spermatogenesis/physiology , Testis/metabolism , Animals , Chaperonin Containing TCP-1/genetics , Helminth Proteins/genetics , Male , Planarians/cytology , Planarians/genetics , Protein Isoforms/genetics , Protein Isoforms/metabolism , Testis/cytology
10.
Dev Biol ; 426(1): 43-55, 2017 06 01.
Article in English | MEDLINE | ID: mdl-28434803

ABSTRACT

Cytoplasmic polyadenylation is a mechanism of mRNA regulation prevalent in metazoan germ cells; it is largely dependent on Cytoplasmic Polyadenylation Element Binding proteins (CPEBs). Two CPEB homologs were identified in the planarian Schmidtea mediterranea. Smed-CPEB1 is expressed in ovaries and yolk glands of sexually mature planarians, and required for oocyte and yolk gland development. In contrast, Smed-CPEB2 is expressed in the testes and the central nervous system; its function is required for spermatogenesis as well as non-autonomously for development of ovaries and accessory reproductive organs. Transcriptome analysis of CPEB knockdown animals uncovered a comprehensive collection of molecular markers for reproductive structures in S. mediterranea, including ovaries, testes, yolk glands, and the copulatory apparatus. Analysis by RNA interference revealed contributions for a dozen of these genes during oogenesis, spermatogenesis, or capsule formation. We also present evidence suggesting that Smed-CPEB2 promotes translation of Neuropeptide Y-8, a prohormone required for planarian sexual maturation. These findings provide mechanistic insight into potentially conserved processes of germ cell development, as well as events involved in capsule deposition by flatworms.


Subject(s)
Germ Cells/cytology , Oogenesis/physiology , Ovary/growth & development , Planarians/anatomy & histology , Planarians/growth & development , Spermatogenesis/physiology , mRNA Cleavage and Polyadenylation Factors/genetics , Animals , Cell Differentiation/genetics , Female , Gene Expression Profiling , Ovary/metabolism , Polyadenylation , RNA Interference , RNA, Small Interfering/genetics , Receptors, Neuropeptide Y/biosynthesis , Receptors, Neuropeptide Y/genetics , Sexual Maturation/genetics , Sexual Maturation/physiology , mRNA Cleavage and Polyadenylation Factors/biosynthesis
11.
Dev Cell ; 37(3): 226-37, 2016 05 09.
Article in English | MEDLINE | ID: mdl-27165555

ABSTRACT

Differentiation of pluripotent stem cells (PSCs) requires transposon silencing throughout the process. PIWIs, best known as key factors in germline transposon silencing, are also known to act in somatic differentiation of planarian PSCs (neoblasts). However, how PIWIs control the latter process remains elusive. Here, using Dugesia japonica, we show that a nuclear PIWI, DjPiwiB, was bound to PIWI-interacting RNAs (generally key mediators of PIWI-dependent transposon silencing), and was detected in not only neoblasts but also their descendant somatic cells, which do not express piwi. In contrast, cytoplasmic DjPiwiA and DjPiwiC were detected only in neoblasts, in accord with their transcription there. DjPiwiB was indispensable for regeneration, but dispensable for transposon silencing in neoblasts. However, transposons were derepressed at the onset of differentiation in DjPiwiB-knockdown planarians. Thus, DjPiwiB appears to be inherited by descendant somatic cells of neoblasts to ensure transposon silencing in those cells, which are unable to produce PIWI proteins.


Subject(s)
Argonaute Proteins/metabolism , Cell Differentiation , Cell Nucleus/metabolism , DNA Transposable Elements/genetics , Inheritance Patterns/genetics , Planarians/cytology , Planarians/genetics , Pluripotent Stem Cells/metabolism , Animals , Base Sequence , Gene Silencing , Immunohistochemistry , Models, Biological , RNA, Small Interfering/metabolism
12.
PLoS Genet ; 12(5): e1006030, 2016 05.
Article in English | MEDLINE | ID: mdl-27149082

ABSTRACT

Few animals are known to lay eggs in the absence of ovulation or copulation, as it is presumably energetically wasteful and subjected to negative selection. Characterization of Smed-boule, a member of the DAZ family of germline RNA-binding proteins, revealed that egg capsule (or capsule) production and deposition occurs independently of the presence of gametes in the planarian flatworm Schmidtea mediterranea. Reduction of Smed-boule expression by RNA-interference (RNAi) causes ablation of spermatogonial stem cells and the inability of ovarian germline stem cells to undergo oogenesis. Although animals subjected to Smed-boule RNAi lose their gametes and become sterile, they continue to lay egg capsules. Production of sterile capsules is even observed in virgin Smed-boule(RNAi) and control planarians maintained in complete isolation, demonstrating that egg production in S. mediterranea occurs independently of ovulation, fertilization, or mating. Evidence suggests that this is a conserved feature amongst Platyhelminthes, and therefore relevant to the pathology and dissemination of parasitic flatworms. These findings demonstrate that Smed-boule functions at different stages during male and female germline stem cell development, and also demonstrate that egg capsule production by planarian flatworms occurs independently of signals produced by mating or ova.


Subject(s)
Cell Differentiation/genetics , Cell Proliferation/genetics , Ovum/growth & development , Planarians/genetics , Animals , Female , Fertilization/genetics , Gene Expression Regulation, Developmental , Germ Cells/growth & development , Germ Cells/metabolism , Male , Ovulation/genetics , Ovum/metabolism , Planarians/growth & development , RNA Interference , Regeneration/genetics , Signal Transduction , Stem Cells
13.
Stem Cells Int ; 2016: 6947395, 2016.
Article in English | MEDLINE | ID: mdl-26681954

ABSTRACT

The ability to regenerate complex structures is broadly represented in both plant and animal kingdoms. Although regenerative abilities vary significantly amongst metazoans, cumulative studies have identified cellular events that are broadly observed during regenerative events. For example, structural damage is recognized and wound healing initiated upon injury, which is followed by programmed cell death in the vicinity of damaged tissue and a burst in proliferation of progenitor cells. Sustained proliferation and localization of progenitor cells to site of injury give rise to an assembly of differentiating cells known as the regeneration blastema, which fosters the development of new tissue. Finally, preexisting tissue rearranges and integrates with newly differentiated cells to restore proportionality and function. While heterogeneity exists in the basic processes displayed during regenerative events in different species-most notably the cellular source contributing to formation of new tissue-activation of conserved molecular pathways is imperative for proper regulation of cells during regeneration. Perhaps the most fundamental of such molecular processes entails chromatin rearrangements, which prime large changes in gene expression required for differentiation and/or dedifferentiation of progenitor cells. This review provides an overview of known contributions to regenerative processes by noncoding RNAs and chromatin-modifying enzymes involved in epigenetic regulation.

14.
Methods Mol Biol ; 1365: 323-38, 2016.
Article in English | MEDLINE | ID: mdl-26498794

ABSTRACT

Planarian flatworms have become an important system for the study of stem cell behavior and regulation in vivo. These organisms are able to regenerate any part of their body upon damage or amputation. A crucial cellular event in the process of planarian regeneration is the migration of pluripotent stem cells (known as neoblasts) to the site of injury. Here we describe two approaches for analyzing migration of planarian stem cells to an area where these have been ablated by localized X-ray irradiation. The first approach involves immunolabeling of mitotic neoblasts, while the second is based on tracing stem cells and their progeny after BrdU incorporation. The use of planarians in studies of cell motility is suitable for the identification of factors that influence stem cell migration in vivo and is amenable to RNA interference or pharmacological screening.


Subject(s)
Bromodeoxyuridine/metabolism , Cell Movement , Fluorescent Antibody Technique/methods , Planarians/cytology , Pluripotent Stem Cells/cytology , Pluripotent Stem Cells/metabolism , Animals , Pluripotent Stem Cells/radiation effects , X-Rays
15.
Development ; 141(13): 2592-601, 2014 Jul.
Article in English | MEDLINE | ID: mdl-24903754

ABSTRACT

The well-known regenerative abilities of planarian flatworms are attributed to a population of adult stem cells called neoblasts that proliferate and differentiate to produce all cell types. A characteristic feature of neoblasts is the presence of large cytoplasmic ribonucleoprotein granules named chromatoid bodies, the function of which has remained largely elusive. This study shows that histone mRNAs are a common component of chromatoid bodies. Our experiments also demonstrate that accumulation of histone mRNAs, which is typically restricted to the S phase of eukaryotic cells, is extended during the cell cycle of neoblasts. The planarian PIWI homologs SMEDWI-1 and SMEDWI-3 are required for proper localization of germinal histone H4 (gH4) mRNA to chromatoid bodies. The association between histone mRNA and chromatoid body components extends beyond gH4 mRNA, since transcripts of other core histone genes were also found in these structures. Additionally, piRNAs corresponding to loci of every core histone type have been identified. Altogether, this work provides evidence that links PIWI proteins and chromatoid bodies to histone mRNA regulation in planarian stem cells. The molecular similarities between neoblasts and undifferentiated cells of other organisms raise the possibility that PIWI proteins might also regulate histone mRNAs in stem cells and germ cells of other metazoans.


Subject(s)
Argonaute Proteins/metabolism , Cytoplasmic Granules/metabolism , Histones/genetics , Planarians/metabolism , RNA, Double-Stranded/biosynthesis , RNA, Messenger/metabolism , Animals , Argonaute Proteins/genetics , Blotting, Northern , Bromodeoxyuridine , Fluorescent Antibody Technique , Histones/metabolism , In Situ Hybridization, Fluorescence , Oligonucleotides/genetics , Planarians/genetics , RNA Interference , RNA, Messenger/genetics , Reverse Transcriptase Polymerase Chain Reaction
16.
Dev Dyn ; 242(6): 718-30, 2013 Jun.
Article in English | MEDLINE | ID: mdl-23441014

ABSTRACT

BACKGROUND: The ability to assess gene function is essential for understanding biological processes. Currently, RNA interference (RNAi) is the only technique available to assess gene function in planarians, in which it has been induced by means of injection of double-stranded RNA (dsRNA), soaking, or ingestion of bacteria expressing dsRNA. RESULTS: We describe a simple and robust RNAi protocol, involving in vitro synthesis of dsRNA that is fed to the planarians. Advantages of this protocol include the ability to produce dsRNA from any vector without subcloning, resolution of ambiguities in quantity and quality of input dsRNA, as well as time and ease of application. We have evaluated the logistics of inducing RNAi in planarians using this methodology in careful detail, from the ingestion and processing of dsRNA in the intestine, to timing and efficacy of knockdown in neoblasts, germline, and soma. We also present systematic comparisons of effects of amount, frequency, and mode of dsRNA delivery. CONCLUSIONS: This method gives robust and reproducible results and is amenable to high-throughput studies. Overall, this RNAi methodology provides a significant advance by combining the strengths of current protocols available for dsRNA delivery in planarians and has the potential to benefit RNAi methods in other systems.


Subject(s)
Gene Expression Regulation, Developmental , Planarians/genetics , RNA Interference , RNA, Double-Stranded/genetics , Animals , Bacteria/genetics , Developmental Biology/methods , Genetic Techniques , Genetic Vectors , Phenotype , Reproducibility of Results
17.
Development ; 139(6): 1083-94, 2012 Mar.
Article in English | MEDLINE | ID: mdl-22318224

ABSTRACT

Planarian flatworms contain a population of adult stem cells (neoblasts) that proliferate and generate cells of all tissues during growth, regeneration and tissue homeostasis. A characteristic feature of neoblasts is the presence of chromatoid bodies, large cytoplasmic ribonucleoprotein (RNP) granules morphologically similar to structures present in the germline of many organisms. This study aims to reveal the function, and identify additional components, of planarian chromatoid bodies. We uncover the presence of symmetrical dimethylarginine (sDMA) on chromatoid body components and identify the ortholog of protein arginine methyltransferase PRMT5 as the enzyme responsible for sDMA modification in these proteins. RNA interference-mediated depletion of planarian PRMT5 results in defects in homeostasis and regeneration, reduced animal size, reduced number of neoblasts, fewer chromatoid bodies and increased levels of transposon and repetitive-element transcripts. Our results suggest that PIWI family member SMEDWI-3 is one sDMA-containing chromatoid body protein for which methylation depends on PRMT5. Additionally, we discover an RNA localized to chromatoid bodies, germinal histone H4. Our results reveal new components of chromatoid bodies and their function in planarian stem cells, and also support emerging studies indicative of sDMA function in stabilization of RNP granules and the Piwi-interacting RNA pathway.


Subject(s)
Adult Stem Cells/metabolism , Arginine/analogs & derivatives , Cytoplasmic Granules/chemistry , Helminth Proteins/metabolism , Planarians/cytology , Planarians/metabolism , Protein-Arginine N-Methyltransferases/metabolism , Adult Stem Cells/chemistry , Adult Stem Cells/ultrastructure , Animals , Arginine/metabolism , Base Sequence , Cell Differentiation , Helminth Proteins/genetics , Histones , Interspersed Repetitive Sequences/genetics , Methylation , Planarians/genetics , Protein-Arginine N-Methyltransferases/biosynthesis , RNA Interference , RNA, Small Interfering/genetics , Regeneration/genetics , Ribonucleoproteins/genetics , Ribonucleoproteins/metabolism , Sequence Analysis, RNA
18.
J Neurosci ; 32(4): 1383-94, 2012 Jan 25.
Article in English | MEDLINE | ID: mdl-22279223

ABSTRACT

The astrocyte brain fatty acid binding protein (Fabp7) has previously been shown to have a coordinated diurnal regulation of mRNA and protein throughout mouse brain, and an age-dependent decline in protein expression within synaptoneurosomal fractions. Mechanisms that control time-of-day changes in expression and trafficking Fabp7 to the perisynaptic process are not known. In this study, we confirmed an enrichment of Fabp7 mRNA and protein in the astrocytic perisynaptic compartment, and observed a diurnal change in the intracellular distribution of Fabp7 mRNA in molecular layers of hippocampus. Northern blotting revealed a coordinated time-of-day-dependent oscillation for the Fabp7 mRNA poly(A) tail throughout murine brain. Cytoplasmic polyadenylation element-binding protein 1 (CPEB1) regulates subcellular trafficking and translation of synaptic plasticity-related mRNAs. Here we show that Fabp7 mRNA coimmunoprecipitated with CPEB1 from primary mouse astrocyte extracts, and its 3'UTR contains phylogenetically conserved cytoplasmic polyadenylation elements (CPEs) capable of regulating translation of reporter mRNAs during Xenopus oocyte maturation. Given that Fabp7 expression is confined to astrocytes and neural progenitors in adult mouse brain, the synchronized cycling pattern of Fabp7 mRNA is a novel discovery among known CPE-regulated transcripts. These results implicate circadian, sleep, and/or metabolic control of CPEB-mediated subcellular trafficking and localized translation of Fabp7 mRNA in the tripartite synapse of mammalian brain.


Subject(s)
Astrocytes/metabolism , Circadian Rhythm/physiology , Fatty Acid-Binding Proteins/metabolism , Nerve Tissue Proteins/metabolism , RNA, Messenger/metabolism , Synapses/metabolism , Animals , Base Sequence , Cells, Cultured , Fatty Acid-Binding Protein 7 , Female , Hippocampus/metabolism , Hippocampus/physiology , Male , Mice , Mice, Inbred C57BL , Molecular Sequence Data , Polyadenylation/physiology , Protein Transport/physiology , Subcellular Fractions/metabolism , Subcellular Fractions/physiology , Synapses/physiology , Xenopus
19.
Dev Biol ; 341(2): 429-43, 2010 May 15.
Article in English | MEDLINE | ID: mdl-20230812

ABSTRACT

Planarian regeneration depends on the presence and precise regulation of pluripotent adult somatic stem cells named neoblasts, which differentiate to replace cells of any missing tissue. A characteristic feature of neoblasts is the presence of large perinuclear nonmembranous organelles named "chromatoid bodies", which are comparable to ribonucleoprotein structures found in germ cells of organisms across different phyla. In order to better understand regulation of gene expression in neoblasts, and potentially the function and composition of chromatoid bodies, we characterized homologues to known germ and soma ribonucleoprotein granule components from other organisms and analyzed their function during regeneration of the planarian Dugesia japonica. Expression in neoblasts was detected for 49 of 55 analyzed genes, highlighting the prevalence of post-transcriptional regulation in planarian stem cells. RNAi-mediated knockdown of two factors [ago-2 and bruli] lead to loss of neoblasts, and consequently loss of regeneration, corroborating with results previously reported for a bruli ortholog in the planarian Schmidtea mediterranea (Guo et al., 2006). Conversely, depletion mRNA turnover factors [edc-4 or upf-1], exoribonucleases [xrn-1 or xrn-2], or DEAD box RNA helicases [Djcbc-1 or vas-1] inhibited planarian regeneration, but did not reduce neoblast proliferation or abundance. We also found that depletion of cap-dependent translation initiation factors eIF-3A or eIF-2A interrupted cell cycle progression outside the M-phase of mitosis. Our results show that a set of post-transcriptional regulators is required to maintain the stem cell identity in neoblasts, while another facilitates proper differentiation. We propose that planarian neoblasts maintain pluripotency by employing mechanisms of post-transcriptional regulation exhibited in germ cells and early development of most metazoans.


Subject(s)
Gene Expression Regulation, Developmental , Planarians/cytology , Planarians/metabolism , Animals , Planarians/genetics , RNA Interference , Ribonucleoproteins/metabolism
20.
Dev Growth Differ ; 52(1): 27-41, 2010 Jan.
Article in English | MEDLINE | ID: mdl-20078652

ABSTRACT

Freshwater planarians, Plathelminthes, have been an intriguing model animal of regeneration studies for more than 100 years. Their robust regenerative ability is one of asexual reproductive capacity, in which complete animals develop from tiny body fragments within a week. Pluripotent adult somatic stem cells, called neoblasts, assure this regenerative ability. Neoblasts give rise to not only all types of somatic cells, but also germline cells. During the last decade, several experimental techniques for the analysis of planarian neoblasts at the molecular level, such as in situ hybridization, RNAi and fluorescence activated cell sorting, have been established. Moreover, information about genes involved in maintenance and differentiation of neoblasts has been accumulated. One of the molecular features of neoblasts is the expression of many RNA regulators, which are involved in germline development in other animals, such as vasa and piwi family genes. In this review, we introduce physiological and molecular features of the neoblast, and discuss how germline genes regulate planarian neoblasts and what differences exist between neoblasts and germline cells.


Subject(s)
Cell Differentiation , Planarians/cytology , Pluripotent Stem Cells/cytology , Animals , Gene Expression Regulation, Developmental , Genes, Helminth/genetics , Microscopy, Electron, Transmission , Models, Biological , Planarians/genetics , Planarians/physiology , Pluripotent Stem Cells/metabolism , Pluripotent Stem Cells/ultrastructure , Regeneration/genetics , Regeneration/physiology
SELECTION OF CITATIONS
SEARCH DETAIL