Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 45
Filtrar
1.
Development ; 150(18)2023 09 15.
Artículo en Inglés | MEDLINE | ID: mdl-37681295

RESUMEN

The planarian Schmidtea mediterranea is a well-established model of adult regeneration, which is dependent on a large population of adult stem cells called neoblasts. Upon amputation, planarians undergo transcriptional wounding programs and coordinated stem cell proliferation to give rise to missing tissues. Interestingly, the Wnt signaling pathway is key to guiding what tissues are regenerated, yet less known are the transcriptional regulators that ensure proper activation and timing of signaling pathway components. Here, we have identified an aristaless-like homeobox transcription factor, alx-3, that is enriched in a population of putative neural-fated progenitor cells at homeostasis, and is also upregulated in stem cells and muscle cells at anterior-facing wounds upon amputation. Knockdown of alx-3 results in failure of head regeneration and patterning defects in amputated tail fragments. alx-3 is required for the expression of several early wound-induced genes, including the Wnt inhibitor notum, which is required to establish anterior polarity during regeneration. Together, these findings reveal a role for alx-3 as an early wound-response transcriptional regulator in both muscle cells and stem cells that is required for anterior regeneration by promoting a low-Wnt environment.


Asunto(s)
Planarias , Animales , Planarias/genética , Genes Homeobox , Regulación de la Expresión Génica , Células Madre , Vía de Señalización Wnt/genética , Interferencia de ARN
2.
Proc Natl Acad Sci U S A ; 118(16)2021 04 20.
Artículo en Inglés | MEDLINE | ID: mdl-33859045

RESUMEN

The extracellular matrix (ECM) provides a precise physical and molecular environment for cell maintenance, self-renewal, and differentiation in the stem cell niche. However, the nature and organization of the ECM niche is not well understood. The adult freshwater planarian Schmidtea mediterranea maintains a large population of multipotent stem cells (neoblasts), presenting an ideal model to study the role of the ECM niche in stem cell regulation. Here we tested the function of 165 planarian homologs of ECM and ECM-related genes in neoblast regulation. We identified the collagen gene family as one with differential effects in promoting or suppressing proliferation of neoblasts. col4-1, encoding a type IV collagen α-chain, had the strongest effect. RNA interference (RNAi) of col4-1 impaired tissue maintenance and regeneration, causing tissue regression. Finally, we provide evidence for an interaction between type IV collagen, the discoidin domain receptor, and neuregulin-7 (NRG-7), which constitutes a mechanism to regulate the balance of symmetric and asymmetric division of neoblasts via the NRG-7/EGFR pathway.


Asunto(s)
Colágeno Tipo IV/genética , Planarias/genética , Planarias/metabolismo , Animales , Diferenciación Celular/genética , Linaje de la Célula/genética , Colágeno Tipo IV/metabolismo , Matriz Extracelular/genética , Matriz Extracelular/metabolismo , Homeostasis , Colágenos no Fibrilares/metabolismo , Regeneración , Transducción de Señal , Células Madre/citología , Células Madre/metabolismo
3.
BMC Biol ; 21(1): 227, 2023 10 20.
Artículo en Inglés | MEDLINE | ID: mdl-37864247

RESUMEN

BACKGROUND: The flatworm planarian, Schmidtea mediterranea, has a large population of adult stem cells (ASCs) that replace any cell type during tissue turnover or regeneration. How planarian ASCs (called neoblasts) manage self-renewal with the ability to produce daughter cells of different cell lineages (multipotency) is not well understood. Chromatin remodeling complexes ultimately control access to DNA regions of chromosomes and together with specific transcription factors determine whether a gene is transcribed in a given cell type. Previous work in planarians determined that RNAi of core components of the BAF chromatin remodeling complex, brg1 and smarcc2, caused increased ASCs and failed regeneration, but how these cellular defects arise at the level of gene regulation in neoblasts is unknown. RESULTS: Here, we perform ATAC and RNA sequencing on purified neoblasts, deficient for the BAF complex subunits brg-1 and smarcc2. The data demonstrate that the BAF complex promotes chromatin accessibility and facilitates transcription at target loci, as in other systems. Interestingly, we find that the BAF complex enables access to genes known to be required for the generation of mesoderm- and ectoderm-derived lineages, including muscle, parenchymal cathepsin, neural, and epithelial lineages. BAF complex knockdowns result in disrupted differentiation into these cell lineages and functional consequences on planarian regeneration and tissue turnover. Notably, we did not detect a role for the BAF complex in neoblasts making endodermal lineages. CONCLUSIONS: Our study provides functional insights into how the BAF complex contributes to cell fate decisions in planarian ASCs in vivo.


Asunto(s)
Planarias , Animales , Planarias/genética , Ensamble y Desensamble de Cromatina , Ectodermo , Factores de Transcripción/genética , Factores de Transcripción/metabolismo , Células Madre/metabolismo , Diferenciación Celular/genética
4.
EMBO Rep ; 22(3): e50292, 2021 03 03.
Artículo en Inglés | MEDLINE | ID: mdl-33511776

RESUMEN

Epimorphic regeneration commonly relies on the activation of reserved stem cells to drive new cell production. The planarian Schmidtea mediterranea is among the best regenerators in nature, thanks to its large population of adult stem cells, called neoblasts. While neoblasts have long been known to drive regeneration, whether a subset of neoblasts is reserved for this purpose is unknown. Here, we revisit the idea of reserved neoblasts by approaching neoblast heterogeneity from a regulatory perspective. By implementing a new fluorescence-activated cell sorting strategy in planarians, we identify a population of neoblasts defined by low transcriptional activity. These RNAlow neoblasts are relatively slow-cycling at homeostasis and undergo a morphological regeneration response characterized by cell growth at 48 h post-amputation. At this time, RNAlow neoblasts proliferate in a TOR-dependent manner. Additionally, knockdown of the tumour suppressor Lrig-1, which is enriched in RNAlow neoblasts, results in RNAlow neoblast growth and hyperproliferation at homeostasis, and ultimately delays regeneration. We propose that slow-cycling RNAlow neoblasts represent a regeneration-reserved neoblast population.


Asunto(s)
Planarias , Animales , Homeostasis , Planarias/genética , Células Madre
5.
PLoS Genet ; 16(2): e1008613, 2020 02.
Artículo en Inglés | MEDLINE | ID: mdl-32078629

RESUMEN

The extracellular matrix (ECM) is important for maintaining the boundaries between tissues. This role is particularly critical in the stem cell niche, as pre-neoplastic or cancerous stem cells must pass these boundaries in order to invade into the surrounding tissue. Here, we examine the role of the ECM as a regulator of the stem cell compartment in the planarian Schmidtea mediterranea, a highly regenerative, long-lived organism with a large population of adult stem cells. We identify two EGF repeat-containing genes, megf6 and hemicentin, with identical knockdown phenotypes. We find that megf6 and hemicentin are needed to maintain the structure of the basal lamina, and in the absence of either gene, pluripotent stem cells migrate ectopically outside of their compartment and hyper-proliferate, causing lesions in the body wall muscle. These muscle lesions and ectopic stem cells are also associated with ectopic gut branches, which protrude from the normal gut towards the dorsal side of the animal. Interestingly, both megf6 and hemicentin knockdown worms are capable of regenerating tissue free of both muscle lesions and ectopic cells, indicating that these genes are dispensable for regeneration. These results provide insight into the role of planarian ECM in restricting the stem cell compartment, and suggest that signals within the compartment may act to suppress stem cell hyperproliferation.


Asunto(s)
Células Madre Adultas/fisiología , Genes de Helminto/fisiología , Platelmintos/fisiología , Células Madre Pluripotentes/fisiología , Nicho de Células Madre/genética , Animales , Animales Modificados Genéticamente , Movimiento Celular/genética , Matriz Extracelular/metabolismo , Técnicas de Silenciamiento del Gen , Proteínas del Helminto/genética , Proteínas del Helminto/metabolismo , Platelmintos/citología , Regeneración/genética
6.
Proc Natl Acad Sci U S A ; 117(32): 19310-19320, 2020 08 11.
Artículo en Inglés | MEDLINE | ID: mdl-32727892

RESUMEN

Fat, Fat-like, and Dachsous family cadherins are giant proteins that regulate planar cell polarity (PCP) and cell adhesion in bilaterians. Their evolutionary origin can be traced back to prebilaterian species, but their ancestral function(s) are unknown. We identified Fat-like and Dachsous cadherins in Hydra, a member of phylum Cnidaria a sister group of bilaterian. We found Hydra does not possess a true Fat homolog, but has homologs of Fat-like (HyFatl) and Dachsous (HyDs) that localize at the apical membrane of ectodermal epithelial cells and are planar polarized perpendicular to the oral-aboral axis of the animal. Using a knockdown approach we found that HyFatl is involved in local cell alignment and cell-cell adhesion, and that reduction of HyFatl leads to defects in tissue organization in the body column. Overexpression and knockdown experiments indicate that the intracellular domain (ICD) of HyFatl affects actin organization through proline-rich repeats. Thus, planar polarization of Fat-like and Dachsous cadherins has ancient, prebilaterian origins, and Fat-like cadherins have ancient roles in cell adhesion, spindle orientation, and tissue organization.


Asunto(s)
Cadherinas/metabolismo , Polaridad Celular , Hydra/citología , Animales , Cadherinas/genética , Adhesión Celular , Hydra/clasificación , Hydra/genética , Hydra/metabolismo , Filogenia , Huso Acromático/genética , Huso Acromático/metabolismo
7.
Semin Cell Dev Biol ; 87: 37-44, 2019 03.
Artículo en Inglés | MEDLINE | ID: mdl-29758350

RESUMEN

Pigment cells serve a variety of important uses across the animal kingdom, and in many species can change and regenerate throughout the lifetime of the organism. The functions of these cells, as well as their origins in both embryonic development and adult regeneration, are not fully understood. Here, we review advances in the study of pigment cells in the freshwater planarian, a model system for stem cell biology and regeneration. Freshwater planarians produce at least three pigment types to generate brown eye and body colouration: melanin, porphyrin, and ommochrome. The body pigments of planarians are produced and contained by a specialized, highly dendritic cell type located in the subepidermal parenchymal space. This cell type is specifically ablated following intense light exposure, a characteristic which has been exploited to discover the gene expression and regeneration of planarian pigment cells. Regenerating pigment cells progress through an immature state marked by upregulation of pigment synthesis genes before differentiating into mature pigment cells; these two states are dynamically regulated in homeostasis to maintain constant body pigmentation. The transcription factors Albino, FoxF-1, and Ets-1, as well as an FGFR-like molecule, are required for proper maintenance of the pigment lineage in both regeneration and homeostasis. These discoveries set the stage for research into external signals that regulate the pigment lineage, as well as possible functions for pigment cells in planarians, including the extra-ocular light response. These insights will address outstanding questions about the evolutionarily-conserved biology of pigment cells.


Asunto(s)
Planarias/genética , Platelmintos/crecimiento & desarrollo , Animales , Linaje de la Célula , Pigmentación
8.
Dev Biol ; 465(2): 144-156, 2020 09 15.
Artículo en Inglés | MEDLINE | ID: mdl-32697972

RESUMEN

The zebrafish model organism has been of exceptional utility for the study of vertebrate development and disease through the application of tissue-specific labelling and overexpression of genes carrying patient-derived mutations. However, there remains a need for a binary expression system that is both non-toxic and not silenced over animal generations by DNA methylation. The Q binary expression system derived from the fungus Neurospora crassa is ideal, because the consensus binding site for the QF transcription factor lacks CpG dinucleotides, precluding silencing by CpG-meditated methylation. To optimize this system for zebrafish, we systematically tested several variants of the QF transcription factor: QF full length; QF2, which lacks the middle domain; QF2w, which is an attenuated version of QF2; and chimeric QFGal4. We found that full length QF and QF2 were strongly toxic to zebrafish embryos, QF2w was mildly toxic, and QFGal4 was well tolerated, when injected as RNA or expressed ubiquitously from stable transgenes. In addition, QFGal4 robustly activated a Tg(QUAS:GFPNLS) reporter transgene. To increase the utility of this system, we also modified the QF effector sequence termed QUAS, which consists of five copies of the QF binding site. Specifically, we decreased both the CpG dinucleotide content, as well as the repetitiveness of QUAS, to reduce the risk of transgene silencing via CpG methylation. Moreover, these modifications to QUAS removed leaky QF-independent neural expression that we detected in the original QUAS sequence. To demonstrate the utility of our QF optimizations, we show how the Q-system can be used for lineage tracing using a Cre-dependent Tg(ubi:QFGal4-switch) transgene. We also demonstrate that QFGal4 can be used in transient injections to tag and label endogenous genes by knocking in QFGal4 into sox2 and ubiquitin C genes.


Asunto(s)
Animales Modificados Genéticamente , Expresión Génica , Neurospora crassa/genética , Proteínas Protozoarias , Factores de Transcripción , Pez Cebra , Animales , Animales Modificados Genéticamente/genética , Animales Modificados Genéticamente/metabolismo , Proteínas Protozoarias/genética , Proteínas Protozoarias/metabolismo , Factores de Transcripción/genética , Factores de Transcripción/metabolismo , Pez Cebra/genética , Pez Cebra/metabolismo
9.
J Virol ; 94(22)2020 10 27.
Artículo en Inglés | MEDLINE | ID: mdl-32907972

RESUMEN

Freshwater planarians, flatworms from order Tricladida, are experimental models of stem cell biology and tissue regeneration. An aspect of their biology that remains less well studied is their relationship with viruses that may infect them. In this study, we identified a taxon of monosegmented double-stranded RNA (dsRNA) viruses in five planarian species, including the well-characterized model Schmidtea mediterranea Sequences for the S. mediterranea virus (abbreviated SmedTV for S. mediterranea tricladivirus) were found in public transcriptome data from multiple institutions, indicating that SmedTV is prevalent in S. mediterranea lab colonies, though without causing evident disease. The presence of SmedTV in discrete cells was shown through in situ hybridization methods for detecting the viral RNA. SmedTV-staining cells were found to be concentrated in neural structures (eyes and brain) but were also scattered in other worm tissues as well. In contrast, few SmedTV-staining cells were seen in stem cell compartments (also consistent with RNA sequencing data) or early blastema tissue. RNA interference (RNAi) targeted to the SmedTV sequence led to apparent cure of infection, though effects on worm health or behavior were not observed. Efforts to transmit SmedTV horizontally through microinjection were unsuccessful. Based on these findings, we conclude that SmedTV infects S. mediterranea in a persistent manner and undergoes vertical transmission to progeny worms during serial passage in lab colonies. The utility of S. mediterranea as a regeneration model, coupled with the apparent capacity of SmedTV to evade normal host immune/RNAi defenses under standard conditions, argues that further studies are warranted to explore this newly recognized virus-host system.IMPORTANCE Planarians are freshwater flatworms, related more distantly to tapeworms and flukes, and have been developed as models to study the molecular mechanisms of stem cell biology and tissue regeneration. These worms live in aquatic environments, where they are likely to encounter a variety of viruses, bacteria, and eukaryotic organisms with pathogenic potential. How the planarian immune system has evolved to cope with these potential pathogens is not well understood, and only two types of planarian viruses have been described to date. Here, we report discovery and inaugural studies of a novel taxon of dsRNA viruses in five different planarian species. The virus in the best-characterized model species, Schmidtea mediterranea, appears to persist long term in that host while avoiding endogenous antiviral or RNAi mechanisms. The S. mediterranea virus-host system thus seems to offer opportunity for gaining new insights into host defenses and their evolution in an important lab model.


Asunto(s)
Virus ARN Bicatenario/clasificación , Virus ARN Bicatenario/genética , Virus ARN Bicatenario/metabolismo , Planarias/virología , Platelmintos/virología , Animales , Virus ARN Bicatenario/aislamiento & purificación , Evolución Molecular , Agua Dulce , Hibridación in Situ , Planarias/fisiología , Interferencia de ARN , ARN Bicatenario , Análisis de Secuencia de ARN , Células Madre , Transcriptoma
10.
Development ; 144(24): 4540-4551, 2017 12 15.
Artículo en Inglés | MEDLINE | ID: mdl-29158443

RESUMEN

Many pigment cells acquire unique structural properties and gene expression profiles during animal development. The underlying differentiation pathways have been well characterized in cells formed during embryogenesis, such as the neural crest-derived melanocyte. However, much less is known about the developmental origins of pigment cells produced in adult organisms during tissue homeostasis and repair. Here we report a lineage analysis of ommochrome- and porphyrin-producing cells in the brown, freshwater planarian Schmidtea mediterranea Using an RNA-sequencing approach, we identified two classes of markers expressed in sequential fashion when new pigment cells are generated during regeneration or in response to pigment cell ablation. We also report roles for FOXF-1 and ETS-1 transcription factors, as well as for an FGFR-like molecule, in the specification and maintenance of this cell type. Together, our results provide insights into mechanisms of adult pigment cell development in the strikingly colorful Platyhelminthes phylum.


Asunto(s)
Factores de Transcripción Forkhead/genética , Pigmentación/genética , Planarias/crecimiento & desarrollo , Proteína Proto-Oncogénica c-ets-1/genética , Regeneración/fisiología , Animales , Secuencia de Bases , Diferenciación Celular/genética , Linaje de la Célula , Fenotiazinas/metabolismo , Porfirinas/biosíntesis , Interferencia de ARN , ARN Interferente Pequeño/genética , Análisis de Secuencia de ARN , Células Madre/citología , Transcripción Genética/genética
11.
PLoS Genet ; 13(7): e1006874, 2017 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-28686611

RESUMEN

Regeneration requires the precise integration of cues that initiate proliferation, direct differentiation, and ultimately re-pattern tissues to the proper size and scale. Yet how these processes are integrated with wounding responses remains relatively unknown. The freshwater planarian, Schmidtea mediterranea, is an ideal model to study the stereotyped proliferative and transcriptional responses to injury due to its high capacity for regeneration. Here, we characterize the effector of the Hippo signalling cascade, yorkie, during planarian regeneration and its role in restricting early injury responses. In yki(RNAi) regenerating animals, wound responses are hyper-activated such that both stem cell proliferation and the transcriptional wound response program are heighted and prolonged. Using this observation, we also uncovered novel wound-induced genes by RNAseq that were de-repressed in yki(RNAi) animals compared with controls. Additionally, we show that yki(RNAi) animals have expanded epidermal and muscle cell populations, which we hypothesize are the increased sources of wound-induced genes. Finally, we show that in yki(RNAi) animals, the sensing of the size of an injury by eyes or the pharynx is not appropriate, and the brain, gut, and midline cannot remodel or scale correctly to the size of the regenerating fragment. Taken together, our results suggest that yki functions as a key molecule that can integrate multiple aspects of the injury response including proliferation, apoptosis, injury-induced transcription, and patterning.


Asunto(s)
Tipificación del Cuerpo/genética , Diferenciación Celular/genética , Proteínas Nucleares/genética , Regeneración/genética , Animales , Apoptosis/genética , Proliferación Celular/genética , Ojo/crecimiento & desarrollo , Regulación del Desarrollo de la Expresión Génica , Proteínas Nucleares/biosíntesis , Faringe/crecimiento & desarrollo , Planarias/genética , Planarias/crecimiento & desarrollo , Transducción de Señal , Células Madre/metabolismo , Cola (estructura animal)/crecimiento & desarrollo
12.
Dev Biol ; 433(2): 374-383, 2018 01 15.
Artículo en Inglés | MEDLINE | ID: mdl-29291981

RESUMEN

The adult brain of the planarian Schmidtea mediterranea (a freshwater flatworm) is a dynamic structure with constant cell turnover as well as the ability to completely regenerate de novo. Despite this, function and pattern is achieved in a reproducible manner from individual to individual in terms of the correct spatial and temporal production of specific neuronal subtypes. Although several signaling molecules have been found to be key to scaling and cell turnover, the mechanisms by which specific neural subtypes are specified remain largely unknown. Here we performed a 6 day RNAseq time course on planarians that were regenerating either 0, 1, or 2 heads in order to identify novel regulators of brain regeneration. Focusing on transcription factors, we identified a TCF/LEF factor, Smed-tcf1, which was required to correctly pattern the dorsal-lateral cell types of the regenerating brain. The most severely affected neurons in Smed-tcf1(RNAi) animals were the dorsal GABAergic neurons, which failed to regenerate, leading to an inability of the animals to phototaxis away from light. Together, Smed-tcf1 is a critical regulator, required to pattern the dorsal-lateral region of the regenerating planarian brain.


Asunto(s)
Proteínas del Helminto/fisiología , Regeneración Nerviosa/fisiología , Proteínas del Tejido Nervioso/fisiología , Neuronas/fisiología , Planarias/fisiología , Factores de Transcripción TCF/fisiología , Animales , Neuronas GABAérgicas/fisiología , Ganglios de Invertebrados/fisiología , Regulación de la Expresión Génica , Genes de Helminto , Estudios de Asociación Genética , Cabeza/fisiología , Proteínas del Helminto/genética , Regeneración Nerviosa/genética , Especificidad de Órganos , Fototaxis , Planarias/genética , Cola (estructura animal)/fisiología , Transcriptoma
13.
Development ; 141(6): 1197-208, 2014 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-24523458

RESUMEN

During adult homeostasis and regeneration, the freshwater planarian must accomplish a constant balance between cell proliferation and cell death, while also maintaining proper tissue and organ size and patterning. How these ordered processes are precisely modulated remains relatively unknown. Here we show that planarians use the downstream effector of the Hippo signaling cascade, yorkie (yki; YAP in vertebrates) to control a diverse set of pleiotropic processes in organ homeostasis, stem cell regulation, regeneration and axial patterning. We show that yki functions to maintain the homeostasis of the planarian excretory (protonephridial) system and to limit stem cell proliferation, but does not affect the differentiation process or cell death. Finally, we show that Yki acts synergistically with WNT/ß-catenin signaling to repress head determination by limiting the expression domains of posterior WNT genes and that of the WNT-inhibitor notum. Together, our data show that yki is a key gene in planarians that integrates stem cell proliferation control, organ homeostasis, and the spatial patterning of tissues.


Asunto(s)
Planarias/crecimiento & desarrollo , Planarias/fisiología , Transactivadores/fisiología , Células Madre Adultas/citología , Células Madre Adultas/fisiología , Animales , Tipificación del Cuerpo , Diferenciación Celular , Proliferación Celular , Homeostasis , Planarias/genética , Interferencia de ARN , Regeneración/genética , Regeneración/fisiología , Transactivadores/antagonistas & inhibidores , Transactivadores/genética , Vía de Señalización Wnt
14.
Development ; 140(17): 3577-88, 2013 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-23903188

RESUMEN

In contrast to most adult organisms, freshwater planarians can regenerate any injured body part, including their entire nervous system. This allows for the analysis of genes required for both the maintenance and regeneration of specific neural subtypes. In addition, the loss of specific neural subtypes may uncover previously unknown behavioral roles for that neural population in the context of the adult animal. Here we show that two homeodomain transcription factor homologs, Smed-lhx1/5-1 and Smed-pitx, are required for the maintenance and regeneration of serotonergic neurons in planarians. When either lhx1/5-1 or pitx was knocked down by RNA interference, the expression of multiple canonical markers for serotonergic neurons was lost. Surprisingly, the loss of serotonergic function uncovered a role for these neurons in the coordination of motile cilia on the ventral epidermis of planarians that are required for their nonmuscular gliding locomotion. Finally, we show that in addition to its requirement in serotonergic neurons, Smed-pitx is required for proper midline patterning during regeneration, when it is required for the expression of the midline-organizing molecules Smed-slit in the anterior and Smed-wnt1 in the posterior.


Asunto(s)
Proteínas con Homeodominio LIM/metabolismo , Factores de Transcripción Paired Box/metabolismo , Planarias/fisiología , Regeneración/fisiología , Neuronas Serotoninérgicas/fisiología , Animales , Secuencia de Bases , Teorema de Bayes , Biología Computacional , Secuenciación de Nucleótidos de Alto Rendimiento , Hibridación in Situ , Proteínas con Homeodominio LIM/genética , Funciones de Verosimilitud , Microscopía por Video , Modelos Genéticos , Datos de Secuencia Molecular , Factores de Transcripción Paired Box/genética , Filogenia , Interferencia de ARN
15.
Development ; 140(23): 4691-702, 2013 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-24173799

RESUMEN

In contrast to most well-studied model organisms, planarians have a remarkable ability to completely regenerate a functional nervous system from a pluripotent stem cell population. Thus, planarians provide a powerful model to identify genes required for adult neurogenesis in vivo. We analyzed the basic helix-loop-helix (bHLH) family of transcription factors, many of which are crucial for nervous system development and have been implicated in human diseases. However, their potential roles in adult neurogenesis or central nervous system (CNS) function are not well understood. We identified 44 planarian bHLH homologs, determined their patterns of expression in the animal and assessed their functions using RNAi. We found nine bHLHs expressed in stem cells and neurons that are required for CNS regeneration. Our analyses revealed that homologs of coe, hes (hesl-3) and sim label progenitors in intact planarians, and following amputation we observed an enrichment of coe(+) and sim(+) progenitors near the wound site. RNAi knockdown of coe, hesl-3 or sim led to defects in CNS regeneration, including failure of the cephalic ganglia to properly pattern and a loss of expression of distinct neuronal subtype markers. Together, these data indicate that coe, hesl-3 and sim label neural progenitor cells, which serve to generate new neurons in uninjured or regenerating animals. Our study demonstrates that this model will be useful to investigate how stem cells interpret and respond to genetic and environmental cues in the CNS and to examine the role of bHLH transcription factors in adult tissue regeneration.


Asunto(s)
Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/metabolismo , Regulación del Desarrollo de la Expresión Génica , Neurogénesis/fisiología , Planarias/metabolismo , Regeneración/fisiología , Animales , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/biosíntesis , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/genética , Sistema Nervioso Central/crecimiento & desarrollo , Sistema Nervioso Central/metabolismo , Estudio de Asociación del Genoma Completo , Datos de Secuencia Molecular , Neuronas/metabolismo , Planarias/embriología , Planarias/genética , Transducción de Señal , Células Madre/metabolismo
16.
Dev Biol ; 373(2): 442-52, 2013 Jan 15.
Artículo en Inglés | MEDLINE | ID: mdl-23123964

RESUMEN

Freshwater planarians are flatworms of the Lophotrochozoan superphylum and are well known for their regenerative abilities, which rely on a large population of pluripotent adult stem cells. However, the mechanisms by which planarians maintain a precise population of adult stem cells while balancing proliferation and cell death, remain to be elucidated. Here we have identified, characterized, and functionally tested the core Retinoblastoma (Rb) pathway components in planarian adult stem cell biology. The Rb pathway is an ancient and conserved mechanism of proliferation control from plants to animals and is composed of three core components: an Rb protein, and a transcription factor heterodimer of E2F and DP proteins. Although the planarian genome contains all components of the Rb pathway, we found that they have undergone gene loss from the ancestral state, similar to other species in their phylum. The single Rb homolog (Smed-Rb) was highly expressed in planarian stem cells and was required for stem cell maintenance, similar to the Rb-homologs p107 and p130 in vertebrates. We show that planarians and their phylum have undergone the most severe reduction in E2F genes observed thus far, and the single remaining E2F was predicted to be a repressive-type E2F (Smed-E2F4-1). Knockdown of either Smed-E2F4-1 or its dimerization partner Dp (Smed-Dp) by RNAi resulted in temporary hyper-proliferation. Finally, we showed that known Rb-interacting genes in other systems, histone deacetylase 1 and cyclinD (Smed-HDAC1; Smed-cycD), were similar to Rb in expression and phenotypes when knocked down by RNAi, suggesting that these established interactions with Rb may also be conserved in planarians. Together, these results showed that planarians use the conserved components of the Rb tumor suppressor pathway to control proliferation and cell survival.


Asunto(s)
Agua Dulce , Planarias/citología , Planarias/metabolismo , Proteína de Retinoblastoma/metabolismo , Células Madre/citología , Células Madre/metabolismo , Animales , División Celular/genética , Linaje de la Célula/genética , Proliferación Celular , Supervivencia Celular/genética , Ciclina D/metabolismo , Factores de Transcripción E2F/genética , Factores de Transcripción E2F/metabolismo , Embrión no Mamífero/metabolismo , Evolución Molecular , Regulación del Desarrollo de la Expresión Génica , Técnicas de Silenciamiento del Gen , Histona Desacetilasa 1/metabolismo , Homeostasis/genética , Modelos Biológicos , Fenotipo , Planarias/embriología , Planarias/genética , Interferencia de ARN , Regeneración/genética , Proteína de Retinoblastoma/genética , Transducción de Señal/genética
17.
Mol Cell Proteomics ; 11(9): 681-91, 2012 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-22653920

RESUMEN

The freshwater planarian Schmidtea mediterranea has been used in research for over 100 years, and is an emerging stem cell model because of its capability of regenerating large portions of missing body parts. Exteriorly, planarians are covered in mucous secretions of unknown composition, implicated in locomotion, predation, innate immunity, and substrate adhesion. Although the planarian genome has been sequenced, it remains mostly unannotated, challenging both genomic and proteomic analyses. The goal of the current study was to annotate the proteome of the whole planarian and its mucous fraction. The S. mediterranea proteome was analyzed via mass spectrometry by using multidimensional protein identification technology with whole-worm tryptic digests. By using a proteogenomics approach, MS data were searched against an in silico translated planarian transcript database, and by using the Swiss-Prot BLAST algorithm to identify proteins similar to planarian queries. A total of 1604 proteins were identified. The mucous subproteome was defined through analysis of a mucous trail fraction and an extract obtained by treating whole worms with the mucolytic agent N-acetylcysteine. Gene Ontology analysis confirmed that the mucous fractions were enriched with secreted proteins. The S. mediterranea proteome is highly similar to that predicted for the trematode Schistosoma mansoni associated with intestinal schistosomiasis, with the mucous subproteome particularly highly conserved. Remarkably, orthologs of 119 planarian mucous proteins are present in human mucosal secretions and tear fluid. We suggest planarians have potential to be a model system for the characterization of mucous protein function and relevant to parasitic flatworm infections and diseases underlined by mucous aberrancies, such as cystic fibrosis, asthma, and other lung diseases.


Asunto(s)
Proteínas del Helminto/análisis , Planarias/química , Proteoma , Animales , Bases de Datos de Proteínas , Perfilación de la Expresión Génica , Proteínas del Helminto/genética , Humanos , Espectrometría de Masas , Moco/química , Planarias/genética , Proteómica , Lágrimas/química
18.
Cell Rep ; 43(1): 113621, 2024 01 23.
Artículo en Inglés | MEDLINE | ID: mdl-38165802

RESUMEN

Cell corpse removal is a critical component of both development and homeostasis throughout the animal kingdom. Extensive research has revealed many of the mechanisms involved in corpse removal, typically involving engulfment and digestion by another cell; however, the dynamics of cell corpse clearance in adult tissues remain unclear. Here, we track cell death in the adult planarian Schmidtea mediterranea and find that, following light-induced cell death, pigment cell corpses transit to the gut and are excreted from the animal. Gut phagocytes, previously only known to phagocytose food, are required for pigment cells to enter the gut lumen. Finally, we show that the planarian ortholog of ced-12/engulfment and cell motility (ELMO) is required for corpse phagocytosis and removal through the gut. In total, we present a mechanism of cell clearance in an adult organism involving transit of dead cells to the gut, transport into the gut by phagocytes, and physical excretion of debris.


Asunto(s)
Proteínas de Caenorhabditis elegans , Planarias , Animales , Caenorhabditis elegans/metabolismo , Proteínas de Caenorhabditis elegans/metabolismo , Transducción de Señal/fisiología , Apoptosis/fisiología , Planarias/metabolismo , Fagocitosis/fisiología , Cadáver
19.
Dev Biol ; 365(2): 458-69, 2012 May 15.
Artículo en Inglés | MEDLINE | ID: mdl-22445864

RESUMEN

Multicellular organisms are equipped with cellular mechanisms that enable them to replace differentiated cells lost to normal physiological turnover, injury, and for some such as planarians, even amputation. This process of tissue homeostasis is generally mediated by adult stem cells (ASCs), tissue-specific stem cells responsible for maintaining anatomical form and function. To do so, ASCs must modulate the balance between cell proliferation, i.e. in response to nutrients, and that of cell death, i.e. in response to starvation or injury. But how these two antagonistic processes are coordinated remains unclear. Here, we explore the role of the core components of the TOR pathway during planarian tissue homeostasis and regeneration and identified an essential function for TORC1 in these two processes. RNAi-mediated silencing of TOR in intact animals resulted in a significant increase in cell death, whereas stem cell proliferation and stem cell maintenance were unaffected. Amputated animals failed to increase stem cell proliferation after wounding and displayed defects in tissue remodeling. Together, our findings suggest two distinct roles for TORC1 in planarians. TORC1 is required to modulate the balance between cell proliferation and cell death during normal cell turnover and in response to nutrients. In addition, it is required to initiate appropriate stem cell proliferation during regeneration and for proper tissue remodeling to occur to maintain scale and proportion.


Asunto(s)
Apoptosis , Proliferación Celular , Homeostasis/fisiología , Planarias/fisiología , Regeneración/fisiología , Factores de Transcripción/fisiología , Células Madre Adultas/citología , Células Madre Adultas/fisiología , Animales , Homeostasis/genética , Planarias/citología , Planarias/genética , Interferencia de ARN , Regeneración/genética , Factores de Transcripción/genética
20.
Development ; 137(2): 213-21, 2010 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-20040488

RESUMEN

The functions of adult stem cells and tumor suppressor genes are known to intersect. However, when and how tumor suppressors function in the lineages produced by adult stem cells is unknown. With a large population of stem cells that can be manipulated and studied in vivo, the freshwater planarian is an ideal system with which to investigate these questions. Here, we focus on the tumor suppressor p53, homologs of which have no known role in stem cell biology in any invertebrate examined thus far. Planaria have a single p53 family member, Smed-p53, which is predominantly expressed in newly made stem cell progeny. When Smed-p53 is targeted by RNAi, the stem cell population increases at the expense of progeny, resulting in hyper-proliferation. However, ultimately the stem cell population fails to self-renew. Our results suggest that prior to the vertebrates, an ancestral p53-like molecule already had functions in stem cell proliferation control and self-renewal.


Asunto(s)
Células Madre Adultas/citología , Células Madre Adultas/metabolismo , Linaje de la Célula/fisiología , Proteínas del Helminto/metabolismo , Planarias/citología , Planarias/metabolismo , Proteína p53 Supresora de Tumor/metabolismo , Animales , Citometría de Flujo , Proteínas del Helminto/genética , Inmunohistoquímica , Hibridación in Situ , Interferencia de ARN
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA