Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 36
Filter
Add more filters











Publication year range
1.
G3 (Bethesda) ; 14(8)2024 Aug 07.
Article in English | MEDLINE | ID: mdl-38865472

ABSTRACT

The heterochronic genes of the nematode Caenorhabditis elegans control the succession of postembryonic developmental events. The 4 core heterochronic genes lin-14, lin-28, hbl-1, and lin-41 act in a sequence to specify cell fates specific to each of the 4 larval stages. It was previously shown that lin-14 has 2 activities separated in time that promote L1 and L2 developmental events, respectively. Using the auxin-inducible degron system, we find that lin-28 and hbl-1 each have 2 activities that control L2 and L3 events which are also separated in time. Relative to events they control, both lin-28 and hbl-1 appear to act just prior to or concurrently with events of the L2. Relative to each other, lin-28 and hbl-1 appear to act simultaneously. By contrast, the lin-14 activity controlling L2 events precedes those of lin-28 and hbl-1 controlling the same events, suggesting that lin-14's regulation of lin-28 is responsible for the delay. Likewise, the activities of lin-28 and hbl-1 controlling L3 fates act well in advance of those fates, suggesting a similar regulatory gap. lin-41 acts early in the L3 to affect fates of the L4, although it was not possible to determine whether it too has 2 temporally separated activities. We also uncovered a feedback phenomenon that prevents the reactivation of heterochronic gene activity late in development after it has been downregulated. This study places the heterochronic gene activities into a timeline of postembryonic development relative to one another and to the developmental events whose timing they control.


Subject(s)
Caenorhabditis elegans Proteins , Caenorhabditis elegans , Gene Expression Regulation, Developmental , Transcription Factors , Caenorhabditis elegans/genetics , Caenorhabditis elegans/growth & development , Animals , Caenorhabditis elegans Proteins/genetics , Caenorhabditis elegans Proteins/metabolism , Transcription Factors/metabolism , Transcription Factors/genetics , Larva , DNA-Binding Proteins , Nuclear Proteins , Repressor Proteins
2.
Genetics ; 225(4)2023 Dec 06.
Article in English | MEDLINE | ID: mdl-37788363

ABSTRACT

The heterochronic genes of Caenorhabditis elegans comprise the best-studied pathway controlling the timing of tissue and organ formation in an animal. To begin to understand the evolution of this pathway and the significance of the relationships among its components, we characterized 11 Caenorhabditis briggsae orthologs of C. elegans heterochronic genes. Using CRISPR/Cas9, we made a variety of alleles and found that several mutant phenotypes differ in significant ways from those of C. elegans. Although most mutant orthologs displayed defects in developmental timing, their phenotypes could differ in which stages were affected, the penetrance and expressivity of the phenotypes, or by having additional pleiotropies that were not obviously connected to developmental timing. However, when examining pairwise epistasis and synergistic relationships, we found those paralleled the known relationships between their C. elegans orthologs, suggesting that the arrangements of these genes in functional modules are conserved, but the modules' relationships to each other and/or to their targets has drifted since the time of the species' last common ancestor. Furthermore, our investigation has revealed a relationship between this pathway to other aspects of the animal's growth and development, including gonad development, which is relevant to both species.


Subject(s)
Caenorhabditis elegans Proteins , Caenorhabditis , Animals , Caenorhabditis elegans/genetics , Caenorhabditis/genetics , Caenorhabditis elegans Proteins/genetics
3.
Cureus ; 14(11): e31134, 2022 Nov.
Article in English | MEDLINE | ID: mdl-36479402

ABSTRACT

Background As regulations governing appropriate resident supervision increase, it has become increasingly difficult to provide residents with the appropriate level of autonomy during their training years. The "Attending of the Day" describes an experiential teaching method that provides a balance between learners' autonomy and appropriate supervision. Methodology Each day one member of the inpatient team is selected as the "Attending of the Day," or "The Pretending." She or he then performs the typical duties of the teaching faculty, from medical decision-making regarding patient care to educating other team members during rounds. "The Pretending" is directly supervised by the clinical faculty. Results Using the grounded theory methodology, we analyzed 935 anonymous evaluations from students and residents over 14 years, leading to the identification of the following three major themes: created an enabling learning environment, provided autonomy, and improved confidence. These results led to the inclusion of the technique as part of the Back to Bedside initiative, which was rated as an essential tool in building confidence and autonomy by 75% of the participants in the 2018 Accreditation Council for Graduate Medical Education's Back to Bedside residents' well-being survey. Recently, the Jacobs School of Medicine launched the Moments of Excellence in Education: Recognition and Inspiration (MEE:RI) program which gives students a way to recognize exemplary moments of teaching they encounter. The "Attending of the Day" method received recognition as a transformative experience in students' medical education. Conclusions The "Attending of the Day" is the first innovative experiential learning technique that allows learners of all levels in both Undergraduate Medical Education (UME) and Graduate Medical Education (GME) to practice and assess autonomy. This innovation suggests that residents and students are looking for opportunities to challenge themselves. "The Pretending" allows them to experience those challenges in an empowering learning environment while they gradually build their confidence on the path to achieving progressive autonomy.

4.
Genetics ; 220(2)2022 02 04.
Article in English | MEDLINE | ID: mdl-34739048

ABSTRACT

The auxin-inducible degradation system in C. elegans allows for spatial and temporal control of protein degradation via heterologous expression of a single Arabidopsis thaliana F-box protein, transport inhibitor response 1 (AtTIR1). In this system, exogenous auxin (Indole-3-acetic acid; IAA) enhances the ability of AtTIR1 to function as a substrate recognition component that adapts engineered degron-tagged proteins to the endogenous C. elegans E3 ubiquitin ligases complex [SKR-1/2-CUL-1-F-box (SCF)], targeting them for degradation by the proteosome. While this system has been employed to dissect the developmental functions of many C. elegans proteins, we have found that several auxin-inducible degron (AID)-tagged proteins are constitutively degraded by AtTIR1 in the absence of auxin, leading to undesired loss-of-function phenotypes. In this manuscript, we adapt an orthogonal auxin derivative/mutant AtTIR1 pair [C. elegans AID version 2 (C.e.AIDv2)] that transforms the specificity of allosteric regulation of TIR1 from IAA to one that is dependent on an auxin derivative harboring a bulky aryl group (5-Ph-IAA). We find that a mutant AtTIR1(F79G) allele that alters the ligand-binding interface of TIR1 dramatically reduces ligand-independent degradation of multiple AID*-tagged proteins. In addition to solving the ectopic degradation problem for some AID-targets, the addition of 5-Ph-IAA to culture media of animals expressing AtTIR1(F79G) leads to more penetrant loss-of-function phenotypes for AID*-tagged proteins than those elicited by the AtTIR1-IAA pairing at similar auxin analog concentrations. The improved specificity and efficacy afforded by the mutant AtTIR1(F79G) allele expand the utility of the AID system and broaden the number of proteins that can be effectively targeted with it.


Subject(s)
Arabidopsis Proteins , Arabidopsis , Caenorhabditis elegans Proteins , F-Box Proteins , Animals , Arabidopsis/genetics , Arabidopsis/metabolism , Arabidopsis Proteins/genetics , Caenorhabditis elegans/genetics , Caenorhabditis elegans/metabolism , Caenorhabditis elegans Proteins/genetics , Caenorhabditis elegans Proteins/metabolism , F-Box Proteins/genetics , F-Box Proteins/metabolism , Indoleacetic Acids/metabolism
5.
J Phys Condens Matter ; 32(35): 355402, 2020 Apr 24.
Article in English | MEDLINE | ID: mdl-32330909

ABSTRACT

In this report, we present results of high-pressure experiments probing the melt line of zirconium (Zr) up to 37 GPa. This investigation has determined that temperature versus laser power curves provide an accurate method to determine melt temperatures. When this information is combined with the onset of diffuse scattering, which is associated with the melt process, we demonstrate the ability to accurately determine the melt boundary. This presents a reliable method for rapid determination of melt boundary and agrees well with other established techniques for such measurements, as reported in previous works on Zr.

6.
J Phys Condens Matter ; 32(12): 12LT02, 2020 Mar 20.
Article in English | MEDLINE | ID: mdl-31796651

ABSTRACT

Zirconium (Zr) has properties conducive to nuclear applications and exhibits complex behavior at high pressure with respect to the effects of impurities, deviatoric stress, kinetics, and grain growth which makes it scientifically interesting. Here, we present experimental results on the 300 K equation of state of ultra-high purity Zr obtained using the diamond-anvil cell coupled with synchrotron-based x-ray diffraction and electrical resistance measurements. Based on quasi-hydrostatic room-temperature compression in helium to pressure P = 69.4(2) GPa, we constrain the bulk modulus and its pressure derivative of body-centered cubic (bcc) ß-Zr to be K = 224(2) GPa and K' = 2.6(1) at P = 37.0(1) GPa. A Monte Carlo approach was developed to accurately quantify the uncertainties in K and K'. In the Monte Carlo simulations, both the unit-cell volume and pressure vary according to their experimental uncertainty. Our high-pressure studies do not indicate additional isostructural volume collapse in the bcc phase of Zr in the 56-58 GPa pressure range.

7.
Contact Dermatitis ; 82(3): 137-146, 2020 Mar.
Article in English | MEDLINE | ID: mdl-31617582

ABSTRACT

BACKGROUND: High-resolution magic angle spinning (HRMAS) is a nuclear magnetic resonance (NMR) technique that enables the characterization of metabolic phenotypes/metabolite profiles of cells, tissues, and organs, under both normal and pathological conditions, without resorting to time-consuming extraction techniques. OBJECTIVES: To assess the impact of chemical skin sensitizers vs non-sensitizers on the metabolome of three-dimensional reconstructed human epidermis (RHE) by HRMAS NMR. METHODS: Based on the SENS-IS assay, 12 skin sensitizers and five non-sensitizing chemicals were investigated and applied on EpiSkin RHE at the published maximal non-irritating concentrations under the conditions of the test. The metabolome of RHE samples was then analyzed by HRMAS NMR. RESULTS: A total of 32 different metabolites were identified; 20 of these were quantified for all samples. Statistical univariate analysis showed that the tissue content of most measured metabolites (with the exception of acetate and glucose) was different in the untreated, treated with non-sensitizers, and treated with sensitizers samples. In RHE samples in contact with sensitizing chemicals, concentrations of 18 metabolites were significantly decreased. Alanine and tyrosine could not discriminate between sensitizer- and non-sensitizer-treated groups. A multivariate partial least-squares-discriminant analysis was performed on the two treated groups, discriminating sensitizing and non-sensitizing chemicals with a very good R2Y value of 0.87 and a good Q2Y value of 0.70. CONCLUSIONS: Data suggest that HRMAS NMR could be used to monitor the impact of chemicals, skin allergens vs non-sensitizers, on the metabolome of three-dimensional RHE.


Subject(s)
Allergens/toxicity , Dermatitis, Allergic Contact/metabolism , Epidermis/metabolism , Magnetic Resonance Spectroscopy/methods , Metabolome , Biomarkers/metabolism , Dermatitis, Allergic Contact/etiology , Discriminant Analysis , Humans , Multivariate Analysis , Pilot Projects , Skin Irritancy Tests
8.
FASEB J ; 33(3): 3291-3303, 2019 03.
Article in English | MEDLINE | ID: mdl-30423261

ABSTRACT

The RNA-binding protein LIN28 is known to regulate cell fate, tissue growth, and pluripotency; however, a unified understanding of its role at the cellular level has not been achieved. Here, we address its developmental activity in mammalian postnatal neurogenesis. Constitutive expression of LIN28 in progenitor cells of the mouse subventricular zone (SVZ) caused several distinct effects: 1) the number of differentiated neurons in the olfactory bulb was dramatically reduced, whereas the relative abundance of 2 neuronal subtypes was significantly altered, 2) the population of proliferating neural progenitors in the SVZ was reduced, whereas the proportion of neuroblasts was increased, and 3) the number of astrocytes was reduced, occasionally causing them to appear early. Thus, LIN28 acts at a poststem cell/predifferentiation step, and its continuous expression caused a precocious phenotype unlike in other experimental systems. Furthermore, for the first time in a vertebrate system, we separate the majority of the biologic role of LIN28 from its known activity of blocking the microRNA let-7 by using a circular RNA sponge. We find that although LIN28 has a multifaceted role in the number and types of cells produced during postnatal neurogenesis, it appears that its action through let-7 is responsible for only a fraction of these effects.-Romer-Seibert, J. S., Hartman, N. W., Moss, E. G. The RNA-binding protein LIN28 controls progenitor and neuronal cell fate during postnatal neurogenesis.


Subject(s)
Neurogenesis/physiology , Neurons/cytology , Neurons/metabolism , RNA-Binding Proteins/metabolism , Aging/genetics , Aging/metabolism , Aging/pathology , Animals , Astrocytes/cytology , Astrocytes/metabolism , Caenorhabditis elegans/genetics , Caenorhabditis elegans/growth & development , Caenorhabditis elegans/metabolism , Cell Count , Cell Differentiation , Cell Line , Gene Expression Regulation, Developmental , Lateral Ventricles/cytology , Lateral Ventricles/metabolism , Mice , MicroRNAs/antagonists & inhibitors , MicroRNAs/genetics , MicroRNAs/metabolism , Models, Neurological , Neural Stem Cells/cytology , Neural Stem Cells/metabolism , Olfactory Bulb/cytology , Olfactory Bulb/metabolism , RNA/genetics , RNA/metabolism , RNA, Circular , RNA-Binding Proteins/genetics
9.
Genetics ; 205(1): 251-262, 2017 01.
Article in English | MEDLINE | ID: mdl-27815363

ABSTRACT

In normal development, the order and synchrony of diverse developmental events must be explicitly controlled. In the nematode Caenorhabditis elegans, the timing of larval events is regulated by hierarchy of proteins and microRNAs (miRNAs) known as the heterochronic pathway. These regulators are organized in feedforward and feedback interactions to form a robust mechanism for specifying the timing and execution of cell fates at successive stages. One member of this pathway is the RNA binding protein LIN-28, which promotes pluripotency and cell fate decisions in successive stages. Two genetic circuits control LIN-28 abundance: it is negatively regulated by the miRNA lin-4, and positively regulated by the transcription factor LIN-14 through a mechanism that was previously unknown. In this report, we used animals that lack lin-4 to elucidate LIN-14's activity in this circuit. We demonstrate that three let-7 family miRNAs-miR-48, miR-84, and miR-241-inhibit lin-28 expression. Furthermore, we show genetically that these miRNAs act between lin-14 and lin-28, and that they comprise the pathway by which lin-14 positively regulates lin-28 We also show that the lin-4 family member mir-237, also regulates early cell fates. Finally, we show that the expression of these miRNAs is directly inhibited by lin-14 activity, making them the first known targets of lin-14 that act in the heterochronic pathway.


Subject(s)
Caenorhabditis elegans Proteins/genetics , Caenorhabditis elegans/genetics , Nuclear Proteins/genetics , Repressor Proteins/genetics , Animals , Caenorhabditis elegans Proteins/metabolism , Cell Differentiation/genetics , Gene Expression Regulation, Developmental , Larva/genetics , MicroRNAs/genetics , MicroRNAs/metabolism , Nuclear Proteins/metabolism , RNA-Binding Proteins/genetics , Repressor Proteins/metabolism , Transcription Factors/genetics
10.
Chem Res Toxicol ; 29(7): 1172-8, 2016 07 18.
Article in English | MEDLINE | ID: mdl-27281158

ABSTRACT

Chemical modification of epidermal proteins by skin sensitizers is the molecular event which initiates the induction of contact allergy. However, not all chemical skin allergens react directly as haptens with epidermal proteins but need either a chemical (prehaptens) or metabolic (prohaptens) activation step to become reactive. Cinnamyl alcohol has been considered a model prohapten, as this skin sensitizer has no intrinsic reactivity. Therefore, the prevailing theory is that cinnamyl alcohol is enzymatically oxidized into the protein-reactive cinnamaldehyde, which is the sensitizing agent. Knowing that reconstructed human epidermis (RHE) models have been demonstrated to be quite similar to the normal human epidermis in terms of metabolic enzymes, use of RHE may be useful to investigate the in situ metabolism/activation of cinnamyl alcohol, particularly when coupled with high-resolution magic angle spinning nuclear magnetic resonance. Incubation of carbon-13 substituted cinnamyl derivatives with RHE did not result in the formation of cinnamaldehyde. The metabolites formed suggest the formation of an epoxy-alcohol and an allylic sulfate as potential electrophiles. These data suggest that cinnamyl alcohol is inducing skin sensitization through a route independent of the one involving cinnamaldehyde and should therefore be considered as a skin sensitizer on its own.


Subject(s)
Propanols/metabolism , Skin/metabolism , Carbon-13 Magnetic Resonance Spectroscopy , Humans , Propanols/pharmacology , Proton Magnetic Resonance Spectroscopy , Skin/drug effects
11.
Development ; 142(9): 1616-27, 2015 May 01.
Article in English | MEDLINE | ID: mdl-25922525

ABSTRACT

Neural progenitor cells (NPCs) have distinct proliferation capacities at different stages of brain development. Lin28 is an RNA-binding protein with two homologs in mice: Lin28a and Lin28b. Here we show that Lin28a/b are enriched in early NPCs and their expression declines during neural differentiation. Lin28a single-knockout mice show reduced NPC proliferation, enhanced cell cycle exit and a smaller brain, whereas mice lacking both Lin28a alleles and one Lin28b allele display similar but more severe phenotypes. Ectopic expression of Lin28a in mice results in increased NPC proliferation, NPC numbers and brain size. Mechanistically, Lin28a physically and functionally interacts with Imp1 (Igf2bp1) and regulates Igf2-mTOR signaling. The function of Lin28a/b in NPCs could be attributed, at least in part, to the regulation of their mRNA targets that encode Igf1r and Hmga2. Thus, Lin28a and Lin28b have overlapping functions in temporally regulating NPC proliferation during early brain development.


Subject(s)
Brain/embryology , Cell Proliferation/physiology , DNA-Binding Proteins/metabolism , Gene Expression Regulation, Developmental/physiology , Neural Stem Cells/physiology , RNA-Binding Proteins/metabolism , Animals , Brain/cytology , Bromodeoxyuridine , DNA-Binding Proteins/genetics , Electroporation , Gene Expression Regulation, Developmental/genetics , HMGA2 Protein/metabolism , Immunoprecipitation , Mice , Mice, Knockout , RNA-Binding Proteins/genetics , Real-Time Polymerase Chain Reaction
12.
Wiley Interdiscip Rev Dev Biol ; 3(5): 365-77, 2014.
Article in English | MEDLINE | ID: mdl-25124757

ABSTRACT

UNLABELLED: In certain instances we can witness cells controlling the sequence of their behaviors as they divide and differentiate. Striking examples occur in the nervous systems of animals where the order of differentiated cell types can be traced to internal changes in their progenitors. Elucidating the molecular mechanisms underlying such cell fate succession has been of interest for its role in generating cell type diversity and proper tissue structure. Another well-studied instance of developmental timing occurs in the larva of the nematode Caenorhabditis elegans, where the heterochronic gene pathway controls the succession of a variety of developmental events. In each case, the identification of molecules involved and the elucidation of their regulatory relationships is ongoing, but some important factors and dynamics have been revealed. In particular, certain homologs of worm heterochronic factors have been shown to work in neural development, alerting us to possible connections among these systems and the possibility of universal components of timing mechanisms. These connections also cause us to consider whether cell-intrinsic timing is more widespread, regardless of whether multiple differentiated cell types are produced in any particular order. For further resources related to this article, please visit the WIREs website. CONFLICT OF INTEREST: The authors have declared no conflicts of interest for this article.


Subject(s)
Gene Expression Regulation, Developmental , Neurogenesis , Animals , MicroRNAs/genetics , RNA-Binding Proteins/genetics , RNA-Binding Proteins/metabolism , Transcription Factors/genetics , Transcription Factors/metabolism
13.
Biochim Biophys Acta ; 1838(10): 2607-14, 2014 Oct.
Article in English | MEDLINE | ID: mdl-24998360

ABSTRACT

A diminution in the order of membrane lipids, which occurs during apoptosis, has been shown to correlate with increased membrane susceptibility to hydrolysis by secretory phospholipase A2. Studies with artificial membranes, however, have demonstrated that the relationship between membrane order and hydrolysis is more complex than suggested thus far by cell studies. To better resolve this relationship, this study focused on comparisons between increasing temperature and calcium ionophore as means of decreasing membrane order in S49 cells. Although these two treatments caused comparable changes in apparent membrane order as detected by steady-state fluorescence measurements, only ionophore treatment enhanced phospholipase activity. Experiments with exogenously-added phosphatidylserine indicated that the difference was not due to the presence of that anionic phospholipid in the outer membrane leaflet. Instead, analysis of the equilibration kinetics of various cationic membrane probes revealed that the difference could relate to the spacing of membrane lipids. Specifically, ionophore treatment increased that spacing while temperature only affected overall membrane order and fluidity. To consider the possibility that the distinction with ionophore might relate to the actin cytoskeleton, cells were stained with phalloidin and imaged via confocal microscopy. Ionophore caused disruption of actin fibers while increased temperature did not. This apparent connection between membrane hydrolysis and the cytoskeleton was further corroborated by examining the relationship among these events during apoptosis stimulated by thapsigargin.


Subject(s)
Calcium Ionophores/pharmacology , Cell Membrane/enzymology , Hot Temperature , Ionomycin/pharmacology , Membrane Fluidity/drug effects , Phospholipases A2, Secretory/metabolism , Actin Cytoskeleton/metabolism , Animals , Cell Line, Tumor , Mice , Phalloidine/pharmacology , Phospholipids/metabolism , Poisons/pharmacology
14.
Curr Top Dev Biol ; 105: 153-80, 2013.
Article in English | MEDLINE | ID: mdl-23962842

ABSTRACT

Molecular mechanisms control the timing, sequence, and synchrony of developmental events in multicellular organisms. In Caenorhabditis elegans, these mechanisms are revealed through the analysis of mutants with "heterochronic" defects: cell division or differentiation patterns that occur in the correct lineage, but simply at the wrong time. Subsets of cells in these mutants thus express temporal identities normally restricted to a different life stage. A seminal finding arising from studies of the heterochronic genes was the discovery of miRNAs; these tiny miRNAs are now a defining feature of the pathway. A series of sequentially expressed miRNAs guide larval transitions through stage-specific repression of key effector molecules. The wild-type lineage patterns are executed as discrete modules programmed between temporal borders imposed by the molting cycles. How these successive events are synchronized with the oscillatory molting cycle is just beginning to come to light. Progression through larval stages can be specifically, yet reversibly, halted in response to environmental cues, including nutrient availability. Here too, heterochronic genes and miRNAs play key roles. Remarkably, developmental arrest can, in some cases, either mask or reveal timing defects associated with mutations. In this chapter, we provide an overview of how the C. elegans heterochronic gene pathway guides developmental transitions during continuous and interrupted larval development.


Subject(s)
Caenorhabditis elegans/growth & development , Gene Expression Regulation, Developmental/physiology , MicroRNAs/metabolism , Models, Biological , Molting/physiology , Morphogenesis/physiology , Animals , Caenorhabditis elegans/genetics , Gene Expression Regulation, Developmental/genetics , Larva/growth & development , MicroRNAs/genetics , Morphogenesis/genetics , Time Factors
15.
Stem Cells ; 31(8): 1563-73, 2013 Aug.
Article in English | MEDLINE | ID: mdl-23666760

ABSTRACT

LIN28A/B are RNA binding proteins implicated by genetic association studies in human growth and glucose metabolism. Mice with ectopic over-expression of Lin28a have shown related phenotypes. Here, we describe the first comprehensive analysis of the physiologic consequences of Lin28a and Lin28b deficiency in knockout (KO) mice. Lin28a/b-deficiency led to dwarfism starting at different ages, and compound gene deletions showed a cumulative dosage effect on organismal growth. Conditional gene deletion at specific developmental stages revealed that fetal but neither neonatal nor adult deficiency resulted in growth defects and aberrations in glucose metabolism. Tissue-specific KO mice implicated skeletal muscle-deficiency in the abnormal programming of adult growth and metabolism. The effects of Lin28b KO could be rescued by Tsc1 haplo-insufficiency in skeletal muscles. Our data implicate fetal expression of Lin28a/b in the regulation of life-long effects on metabolism and growth, and demonstrate that fetal Lin28b acts at least in part via mTORC1 signaling.


Subject(s)
DNA-Binding Proteins/physiology , Glucose/metabolism , RNA-Binding Proteins/physiology , Animals , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , Dwarfism/genetics , Dwarfism/metabolism , Female , Fetus/metabolism , Gene Expression , Glucose/genetics , Growth and Development , Male , Mice , Mice, Inbred BALB C , Mice, Knockout , RNA-Binding Proteins/genetics , RNA-Binding Proteins/metabolism , Sex Factors , Signal Transduction
16.
Stem Cells ; 31(5): 1001-9, 2013 May.
Article in English | MEDLINE | ID: mdl-23378032

ABSTRACT

Overexpression of LIN28A is associated with human germ cell tumors and promotes primordial germ cell (PGC) development from embryonic stem cells in vitro and in chimeric mice. Knockdown of Lin28a inhibits PGC development in vitro, but how constitutional Lin28a deficiency affects the mammalian reproductive system in vivo remains unknown. Here, we generated Lin28a knockout (KO) mice and found that Lin28a deficiency compromises the size of the germ cell pool in both males and females by affecting PGC proliferation during embryogenesis. Interestingly however, in Lin28a KO males, the germ cell pool partially recovers during postnatal expansion, while fertility remains impaired in both males and females mated to wild-type mice. Embryonic overexpression of let-7, a microRNA negatively regulated by Lin28a, reduces the germ cell pool, corroborating the role of the Lin28a/let-7 axis in regulating the germ lineage.


Subject(s)
Fertility/physiology , Germ Cells/physiology , RNA-Binding Proteins/physiology , Age Factors , Animals , Cell Differentiation/physiology , Female , Germ Cells/cytology , Male , Mice , Mice, Knockout , MicroRNAs/biosynthesis , RNA-Binding Proteins/biosynthesis , RNA-Binding Proteins/genetics , Spermatogenesis/physiology , Testis/cytology , Testis/physiology
17.
PLoS Genet ; 8(3): e1002588, 2012.
Article in English | MEDLINE | ID: mdl-22457637

ABSTRACT

lin-28 is a conserved regulator of cell fate succession in animals. In Caenorhabditis elegans, it is a component of the heterochronic gene pathway that governs larval developmental timing, while its vertebrate homologs promote pluripotency and control differentiation in diverse tissues. The RNA binding protein encoded by lin-28 can directly inhibit let-7 microRNA processing by a novel mechanism that is conserved from worms to humans. We found that C. elegans LIN-28 protein can interact with four distinct let-7 family pre-microRNAs, but in vivo inhibits the premature accumulation of only let-7. Surprisingly, however, lin-28 does not require let-7 or its relatives for its characteristic promotion of second larval stage cell fates. In other words, we find that the premature accumulation of mature let-7 does not account for lin-28's precocious phenotype. To explain let-7's role in lin-28 activity, we provide evidence that lin-28 acts in two steps: first, the let-7-independent positive regulation of hbl-1 through its 3'UTR to control L2 stage-specific cell fates; and second, a let-7-dependent step that controls subsequent fates via repression of lin-41. Our evidence also indicates that let-7 functions one stage earlier in C. elegans development than previously thought. Importantly, lin-28's two-step mechanism resembles that of the heterochronic gene lin-14, and the overlap of their activities suggests a clockwork mechanism for developmental timing. Furthermore, this model explains the previous observation that mammalian Lin28 has two genetically separable activities. Thus, lin-28's two-step mechanism may be an essential feature of its evolutionarily conserved role in cell fate succession.


Subject(s)
Caenorhabditis elegans Proteins/genetics , Caenorhabditis elegans , Cell Differentiation/genetics , Larva , Repressor Proteins/genetics , 3' Untranslated Regions/genetics , Animals , Caenorhabditis elegans/genetics , Caenorhabditis elegans/growth & development , Caenorhabditis elegans Proteins/metabolism , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , Gene Expression Regulation, Developmental , Larva/genetics , Larva/growth & development , MicroRNAs , Nuclear Proteins/genetics , Nuclear Proteins/metabolism , Transcription Factors/genetics , Transcription Factors/metabolism
18.
Biochim Biophys Acta ; 1818(5): 1196-204, 2012 May.
Article in English | MEDLINE | ID: mdl-22266334

ABSTRACT

Normal human lymphocytes resisted the hydrolytic action of secretory phospholipase A(2) but became susceptible to the enzyme following treatment with a calcium ionophore, ionomycin. To test the hypothesis that this susceptibility requires exposure of the anionic lipid phosphatidylserine on the external face of the cell membrane, experiments were repeated with a human Burkitt's lymphoma cell line (Raji cells). In contrast to normal lymphocytes or S49 mouse lymphoma cells, most of the Raji cells (83%) did not translocate phosphatidylserine to the cell surface upon treatment with ionomycin. Those few that did display exposed phosphatidylserine were hydrolyzed immediately upon addition of phospholipase A(2). Interestingly, the remaining cells were also completely susceptible to the enzyme but were hydrolyzed at a slower rate and after a latency of about 100s. In contradistinction to the defect in phosphatidylserine translocation, Raji cells did display other physical membrane changes upon ionomycin treatment that may be relevant to hydrolysis by phospholipase A(2). These changes were detected by merocyanine 540 and trimethylammonium diphenylhexatriene fluorescence and were common among normal lymphocytes, S49 cells, and Raji cells. The levels of these latter effects corresponded well with the relative rates of hydrolysis among the three cell lines. These results suggested that while phosphatidylserine enhances the rate of cell membrane hydrolysis by secretory phospholipase A(2), it is not an absolute requirement. Other physical properties such as membrane order contribute to the level of membrane susceptibility to the enzyme independent of phosphatidylserine.


Subject(s)
Cell Membrane/metabolism , Lymphocytes/enzymology , Phosphatidylserines/metabolism , Phospholipases A2, Secretory/metabolism , Phospholipid Transfer Proteins/biosynthesis , Animals , Biological Transport, Active/physiology , Cell Line, Tumor , Cell Membrane/genetics , Gene Expression Regulation, Enzymologic/physiology , Humans , Hydrolysis , Lymphocytes/cytology , Mice , Phosphatidylserines/genetics , Phospholipases A2, Secretory/genetics , Phospholipid Transfer Proteins/genetics
19.
Development ; 137(6): 891-900, 2010 Mar.
Article in English | MEDLINE | ID: mdl-20179095

ABSTRACT

LIN28 is an RNA-binding protein that is expressed in many developing tissues. It can block let-7 (Mirlet7) microRNA processing and help promote pluripotency. We have observed LIN28 expression in the developing mouse neural tube, colocalizing with SOX2, suggesting a role in neural development. To better understand its normal developmental function, we investigated LIN28 activity during neurogliogenesis in vitro, where the succession of neuronal to glial cell fates occurs as it does in vivo. LIN28 expression was high in undifferentiated cells, and was downregulated rapidly upon differentiation. Constitutive LIN28 expression caused a complete block of gliogenesis and an increase in neurogenesis. LIN28 expression was compatible with neuronal differentiation and did not increase proliferation. LIN28 caused significant changes in gene expression prior to any effect on let-7, notably on Igf2. Furthermore, a mutant LIN28 that permitted let-7 accumulation was still able to completely block gliogenesis. Thus, at least two biological activities of LIN28 are genetically separable and might involve distinct mechanisms. LIN28 can differentially promote and inhibit specific fates and does not function exclusively by blocking let-7 family microRNAs. Importantly, the role of LIN28 in cell fate succession in vertebrate cells is analogous to its activity as a developmental timing regulator in C. elegans.


Subject(s)
Cell Differentiation/genetics , MicroRNAs/physiology , Neuroglia/physiology , RNA-Binding Proteins/physiology , Animals , Cell Count , Cell Proliferation , Cells, Cultured , Conserved Sequence/genetics , Conserved Sequence/physiology , Embryo, Mammalian , Gene Expression Regulation, Developmental , Mice , MicroRNAs/metabolism , Neural Tube/cytology , Neural Tube/embryology , Neural Tube/metabolism , Neurogenesis/genetics , Neuroglia/cytology , Neuroglia/metabolism , Protein Structure, Tertiary/physiology , RNA-Binding Proteins/chemistry , RNA-Binding Proteins/genetics , RNA-Binding Proteins/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL