Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 41
Filter
1.
Development ; 151(13)2024 Jul 01.
Article in English | MEDLINE | ID: mdl-38884356

ABSTRACT

Neural crest cells are a stem cell population unique to vertebrate embryos that retains broad multi-germ layer developmental potential through neurulation. Much remains to be learned about the genetic and epigenetic mechanisms that control the potency of neural crest cells. Here, we examine the role that epigenetic readers of the BET (bromodomain and extra terminal) family play in controlling the potential of pluripotent blastula and neural crest cells. We find that inhibiting BET activity leads to loss of pluripotency at blastula stages and a loss of neural crest at neurula stages. We compare the effects of HDAC (an eraser of acetylation marks) and BET (a reader of acetylation) inhibition and find that they lead to similar cellular outcomes through distinct effects on the transcriptome. Interestingly, loss of BET activity in cells undergoing lineage restriction is coupled to increased expression of genes linked to pluripotency and prolongs the competence of initially pluripotent cells to transit to a neural progenitor state. Together these findings advance our understanding of the epigenetic control of pluripotency and the formation of the vertebrate neural crest.


Subject(s)
Neural Crest , Animals , Neural Crest/cytology , Neural Crest/metabolism , Epigenesis, Genetic , Gene Expression Regulation, Developmental , Xenopus Proteins/metabolism , Xenopus Proteins/genetics , Xenopus laevis/embryology , Blastula/metabolism , Blastula/cytology , Cell Differentiation , Pluripotent Stem Cells/cytology , Pluripotent Stem Cells/metabolism , Transcriptome/genetics
2.
Development ; 151(3)2024 Feb 01.
Article in English | MEDLINE | ID: mdl-38345109

ABSTRACT

The field of developmental biology has declined in prominence in recent decades, with off-shoots from the field becoming more fashionable and highly funded. This has created inequity in discovery and opportunity, partly due to the perception that the field is antiquated or not cutting edge. A 'think tank' of scientists from multiple developmental biology-related disciplines came together to define specific challenges in the field that may have inhibited innovation, and to provide tangible solutions to some of the issues facing developmental biology. The community suggestions include a call to the community to help 'rebrand' the field, alongside proposals for additional funding apparatuses, frameworks for interdisciplinary innovative collaborations, pedagogical access, improved science communication, increased diversity and inclusion, and equity of resources to provide maximal impact to the community.


Subject(s)
Developmental Biology
3.
Development ; 150(4)2023 02 15.
Article in English | MEDLINE | ID: mdl-36789951

ABSTRACT

Monoclonal antibodies are powerful and versatile tools that enable the study of proteins in diverse contexts. They are often utilized to assist with identification of subcellular localization and characterization of the function of target proteins of interest. However, because there can be considerable sequence diversity between orthologous proteins in Xenopus and mammals, antibodies produced against mouse or human proteins often do not recognize Xenopus counterparts. To address this issue, we refined existing mouse monoclonal antibody production protocols to generate antibodies against Xenopus proteins of interest. Here, we describe several approaches for the generation of useful mouse anti-Xenopus antibodies to multiple Xenopus proteins and their validation in various experimental approaches. These novel antibodies are now available to the research community through the Developmental Study Hybridoma Bank (DSHB).


Subject(s)
Antibodies, Monoclonal , Xenopus Proteins , Animals , Mice , Hybridomas , Xenopus laevis , Xenopus Proteins/genetics
4.
Semin Cell Dev Biol ; 138: 36-44, 2023 03 30.
Article in English | MEDLINE | ID: mdl-35534333

ABSTRACT

Neural crest cells are central to vertebrate development and evolution, endowing vertebrates with a "new head" that resulted in morphological, physiological, and behavioral features that allowed vertebrates to become active predators. One remarkable feature of neural crest cells is their multi-germ layer potential that allows for the formation of both ectodermal (pigmentation, peripheral glia, sensory neurons) and mesenchymal (connective tissue, cartilage/bone, dermis) cell types. Understanding the cellular and evolutionary origins of this broad cellular potential in the neural crest has been a long-standing focus for developmental biologists. Here, we review recent work that has demonstrated that neural crest cells share key features with pluripotent blastula stem cells, including expression of the Yamanaka stem cell factors (Oct3/4, Klf4, Sox2, c-Myc). These shared features suggest that pluripotency is either retained in the neural crest from blastula stages or subsequently reactivated as the neural crest forms. We highlight the cellular and molecular parallels between blastula stem cells and neural crest cells and discuss the work that has led to current models for the cellular origins of broad potential in the crest. Finally, we explore how these themes can provide new insights into how and when neural crest cells and pluripotency evolved in vertebrates and the evolutionary relationship between these populations.


Subject(s)
Neural Crest , Pluripotent Stem Cells , Animals , Neural Crest/metabolism , Vertebrates/genetics , Ectoderm , Pluripotent Stem Cells/metabolism , Gene Expression Regulation, Developmental , Biological Evolution
5.
Dev Biol ; 505: 34-41, 2024 Jan.
Article in English | MEDLINE | ID: mdl-37890713

ABSTRACT

Neural crest cells are a stem cell population unique to vertebrates that give rise to a diverse array of derivatives, including much of the peripheral nervous system, pigment cells, cartilage, mesenchyme, and bone. Acquisition of these cells drove the evolution of vertebrates and defects in their development underlies a broad set of neurocristopathies. Moreover, studies of neural crest can inform differentiation protocols for pluripotent stem cells and regenerative medicine applications. Xenopus embryos are an important system for studies of the neural crest and have provided numerous insights into the signals and transcription factors that control the formation and later lineage diversification of these stem cells. Pluripotent animal pole explants are a particularly powerful tool in this system as they can be cultured in simple salt solution and instructed to give rise to any cell type including the neural crest. Here we report a protocol for small molecule-mediated induction of the neural crest state from blastula stem cells and validate it using transcriptome analysis and grafting experiments. This is an powerful new tool for generating this important cell type that will facilitate future studies of neural crest development and mutations and variants linked to neurocristopathies.


Subject(s)
Neural Crest , Pluripotent Stem Cells , Animals , Neural Crest/metabolism , Xenopus laevis/genetics , Blastula/metabolism , Cell Differentiation
6.
BMC Genomics ; 23(1): 723, 2022 Oct 23.
Article in English | MEDLINE | ID: mdl-36273135

ABSTRACT

BACKGROUND: During embryogenesis, the developmental potential of initially pluripotent cells becomes progressively restricted as they transit to lineage restricted states. The pluripotent cells of Xenopus blastula-stage embryos are an ideal system in which to study cell state transitions during developmental decision-making, as gene expression dynamics can be followed at high temporal resolution. RESULTS: Here we use transcriptomics to interrogate the process by which pluripotent cells transit to four different lineage-restricted states: neural progenitors, epidermis, endoderm and ventral mesoderm, providing quantitative insights into the dynamics of Waddington's landscape. Our findings provide novel insights into why the neural progenitor state is the default lineage state for pluripotent cells and uncover novel components of lineage-specific gene regulation. These data reveal an unexpected overlap in the transcriptional responses to BMP4/7 and Activin signaling and provide mechanistic insight into how the timing of signaling inputs such as BMP are temporally controlled to ensure correct lineage decisions. CONCLUSIONS: Together these analyses provide quantitative insights into the logic and dynamics of developmental decision making in early embryos. They also provide valuable lineage-specific time series data following the acquisition of specific lineage states during development.


Subject(s)
Gene Expression Regulation, Developmental , Transcriptome , Mesoderm , Endoderm/metabolism , Activins/genetics , Activins/metabolism , Cell Differentiation/genetics , Xenopus Proteins/genetics , Xenopus Proteins/metabolism
7.
Development ; 145(15)2018 08 08.
Article in English | MEDLINE | ID: mdl-30002130

ABSTRACT

The neural crest, a progenitor population that drove vertebrate evolution, retains the broad developmental potential of the blastula cells it is derived from, even as neighboring cells undergo lineage restriction. The mechanisms that enable these cells to preserve their developmental potential remain poorly understood. Here, we explore the role of histone deacetylase (HDAC) activity in this process in Xenopus We show that HDAC activity is essential for the formation of neural crest, as well as for proper patterning of the early ectoderm. The requirement for HDAC activity initiates in naïve blastula cells; HDAC inhibition causes loss of pluripotency gene expression and blocks the ability of blastula stem cells to contribute to lineages of the three embryonic germ layers. We find that pluripotent naïve blastula cells and neural crest cells are both characterized by low levels of histone acetylation, and show that increasing HDAC1 levels enhance the ability of blastula cells to be reprogrammed to a neural crest state. Together, these findings elucidate a previously uncharacterized role for HDAC activity in establishing the neural crest stem cell state.


Subject(s)
Histone Deacetylase 1/metabolism , Neural Crest/embryology , Neural Crest/enzymology , Xenopus Proteins/metabolism , Xenopus laevis/embryology , Acetylation , Animals , Biomarkers/metabolism , Blastula/cytology , Blastula/metabolism , Cell Lineage/drug effects , Cell Lineage/genetics , Embryo, Nonmammalian/drug effects , Embryo, Nonmammalian/metabolism , Gene Expression Regulation, Developmental/drug effects , Histone Deacetylase Inhibitors/pharmacology , Histones/metabolism , Neural Crest/drug effects , Pluripotent Stem Cells/cytology , Pluripotent Stem Cells/drug effects , Pluripotent Stem Cells/metabolism , Xenopus laevis/genetics
10.
Dev Biol ; 444(2): 50-61, 2018 12 15.
Article in English | MEDLINE | ID: mdl-30144418

ABSTRACT

The neural crest is a stem cell population unique to vertebrate embryos that gives rise to derivatives from multiple embryonic germ layers. The molecular underpinnings of potency that govern neural crest potential are highly conserved with that of pluripotent blastula stem cells, suggesting that neural crest cells may have evolved through retention of aspects of the pluripotency gene regulatory network (GRN). A striking difference in the regulatory factors utilized in pluripotent blastula cells and neural crest cells is the deployment of different sub-families of Sox transcription factors; SoxB1 factors play central roles in the pluripotency of naïve blastula and ES cells, whereas neural crest cells require SoxE function. Here we explore the shared and distinct activities of these factors to shed light on the role that this molecular hand-off of Sox factor activity plays in the genesis of neural crest and the lineages derived from it. Our findings provide evidence that SoxB1 and SoxE factors have both overlapping and distinct activities in regulating pluripotency and lineage restriction in the embryo. We hypothesize that SoxE factors may transiently replace SoxB1 factors to control pluripotency in neural crest cells, and then poise these cells to contribute to glial, chondrogenic and melanocyte lineages at stages when SoxB1 factors promote neuronal progenitor formation.


Subject(s)
SOXB1 Transcription Factors/genetics , SOXE Transcription Factors/genetics , Animals , Blastula/metabolism , Gene Expression Regulation, Developmental/genetics , Gene Regulatory Networks/genetics , Germ Layers/metabolism , Neural Crest/metabolism , Pluripotent Stem Cells/metabolism , SOXB1 Transcription Factors/metabolism , SOXE Transcription Factors/metabolism , Transcription Factors/physiology , Xenopus Proteins/genetics , Xenopus laevis/embryology , Xenopus laevis/genetics
11.
Nature ; 497(7449): 374-7, 2013 May 16.
Article in English | MEDLINE | ID: mdl-23676755

ABSTRACT

An ambitious goal in biology is to understand the behaviour of cells during development by imaging-in vivo and with subcellular resolution-changes of the embryonic structure. Important morphogenetic movements occur throughout embryogenesis, but in particular during gastrulation when a series of dramatic, coordinated cell movements drives the reorganization of a simple ball or sheet of cells into a complex multi-layered organism. In Xenopus laevis, the South African clawed frog and also in zebrafish, cell and tissue movements have been studied in explants, in fixed embryos, in vivo using fluorescence microscopy or microscopic magnetic resonance imaging. None of these methods allows cell behaviours to be observed with micrometre-scale resolution throughout the optically opaque, living embryo over developmental time. Here we use non-invasive in vivo, time-lapse X-ray microtomography, based on single-distance phase contrast and combined with motion analysis, to examine the course of embryonic development. We demonstrate that this powerful four-dimensional imaging technique provides high-resolution views of gastrulation processes in wild-type X. laevis embryos, including vegetal endoderm rotation, archenteron formation, changes in the volumes of cavities within the porous interstitial tissue between archenteron and blastocoel, migration/confrontation of mesendoderm and closure of the blastopore. Differential flow analysis separates collective from relative cell motion to assign propulsion mechanisms. Moreover, digitally determined volume balances confirm that early archenteron inflation occurs through the uptake of external water. A transient ectodermal ridge, formed in association with the confrontation of ventral and head mesendoderm on the blastocoel roof, is identified. When combined with perturbation experiments to investigate molecular and biomechanical underpinnings of morphogenesis, our technique should help to advance our understanding of the fundamentals of development.


Subject(s)
Gastrulation/physiology , X-Ray Microtomography/methods , Xenopus laevis/embryology , Animals , Biological Evolution , Cell Movement , Endoderm/embryology , Head/embryology , Imaging, Three-Dimensional/instrumentation , Imaging, Three-Dimensional/methods , Mesoderm/embryology , Morphogenesis , Movement , Rotation , Time Factors , X-Ray Microtomography/instrumentation , Xenopus laevis/anatomy & histology
12.
Dev Biol ; 389(1): 2-12, 2014 May 01.
Article in English | MEDLINE | ID: mdl-24321819

ABSTRACT

The neural crest and craniofacial placodes are two distinct progenitor populations that arise at the border of the vertebrate neural plate. This border region develops through a series of inductive interactions that begins before gastrulation and progressively divide embryonic ectoderm into neural and non-neural regions, followed by the emergence of neural crest and placodal progenitors. In this review, we describe how a limited repertoire of inductive signals-principally FGFs, Wnts and BMPs-set up domains of transcription factors in the border region which establish these progenitor territories by both cross-inhibitory and cross-autoregulatory interactions. The gradual assembly of different cohorts of transcription factors that results from these interactions is one mechanism to provide the competence to respond to inductive signals in different ways, ultimately generating the neural crest and cranial placodes.


Subject(s)
Body Patterning , Ectoderm/embryology , Neural Crest/embryology , Neural Plate/embryology , Animals , Bone Morphogenetic Proteins/genetics , Bone Morphogenetic Proteins/metabolism , Ectoderm/cytology , Ectoderm/metabolism , Epidermal Growth Factor/genetics , Epidermal Growth Factor/metabolism , Gene Expression Regulation, Developmental , Humans , Neural Crest/cytology , Neural Crest/metabolism , Neural Plate/cytology , Neural Plate/metabolism , Wnt Proteins/genetics , Wnt Proteins/metabolism
13.
Dev Biol ; 361(2): 313-25, 2012 Jan 15.
Article in English | MEDLINE | ID: mdl-22119055

ABSTRACT

The neural crest (NC) is a population of multipotent stem cell-like progenitors that arise at the neural plate border in vertebrates and migrate extensively before giving rise to diverse derivatives. A number of components of the neural crest gene regulatory network (NC-GRN) are used reiteratively to control multiple steps in the development of these cells. It is therefore important to understand the mechanisms that control the distinct function of reiteratively used factors in different cellular contexts, and an important strategy for doing so is to identify and characterize the regulatory factors they interact with. Here we report that the LIM adaptor protein, LMO4, is a Slug/Snail interacting protein that is essential for NC development. LMO4 is expressed in NC forming regions of the embryo, as well as in the central nervous system and the cranial placodes. LMO4 is necessary for normal NC development as morpholino-mediated knockdown of this factor leads to loss of NC precursor formation at the neural plate border. Misexpression of LMO4 leads to ectopic expression of some neural crest markers, but a reduction in the expression of others. LMO4 binds directly to Slug and Snail, but not to other components of the NC-GRN and can modulate Slug-mediated neural crest induction, suggesting a mechanistic link between these factors. Together these findings implicate LMO4 as a critical component of the NC-GRN and shed new light on the control of Snail family repressors.


Subject(s)
Adaptor Proteins, Signal Transducing/metabolism , LIM Domain Proteins/metabolism , Neural Crest/embryology , Neural Crest/metabolism , Xenopus Proteins/metabolism , Xenopus/embryology , Xenopus/metabolism , Adaptor Proteins, Signal Transducing/genetics , Animals , Embryo, Nonmammalian/metabolism , Gene Expression Regulation, Developmental , Histone Deacetylases/metabolism , LIM Domain Proteins/genetics , Protein Binding , Protein Structure, Tertiary , Snail Family Transcription Factors , Transcription Factors/chemistry , Transcription Factors/metabolism , Xenopus/genetics , Xenopus Proteins/genetics
14.
Dev Biol ; 366(1): 10-21, 2012 Jun 01.
Article in English | MEDLINE | ID: mdl-22583479

ABSTRACT

Neural crest cells are a population of multipotent stem cell-like progenitors that arise at the neural plate border in vertebrates, migrate extensively, and give rise to diverse derivatives such as melanocytes, craniofacial cartilage and bone, smooth muscle, peripheral and enteric neurons and glia. The neural crest gene regulatory network (NC-GRN) includes a number of key factors that are used reiteratively to control multiple steps in the development of neural crest cells, including the acquisition of stem cell attributes. It is therefore essential to understand the mechanisms that control the distinct functions of such reiteratively used factors in different cellular contexts. The context-dependent control of neural crest specification is achieved through combinatorial interaction with other factors, post-transcriptional and post-translational modifications, and the epigenetic status and chromatin state of target genes. Here we review the current understanding of the NC-GRN, including the role of the neural crest specifiers, their links to the control of "stemness," and their dynamic context-dependent regulation during the formation of neural crest progenitors.


Subject(s)
Embryonic Induction , Gene Regulatory Networks , Multipotent Stem Cells , Neural Crest , Animals , Cell Differentiation , Cell Lineage/genetics , Epigenesis, Genetic , Multipotent Stem Cells/cytology , Multipotent Stem Cells/physiology , Neural Crest/cytology , Neural Crest/embryology , Neural Crest/physiology , Protein Processing, Post-Translational , RNA Processing, Post-Transcriptional , Vertebrates
15.
bioRxiv ; 2023 Sep 29.
Article in English | MEDLINE | ID: mdl-37808794

ABSTRACT

SoxB1 transcription factors (Sox2/3) are well known for their role in early neural fate specification in the embryo, but little is known about functional roles for SoxB1 factors in non-neural ectodermal cell types, such as the neural plate border (NPB). Using Xenopus laevis , we set out to determine if SoxB1 transcription factors have a regulatory function in NPB formation. Herein, we show that SoxB1 factors are necessary for NPB formation, and that prolonged SoxB1 factor activity blocks the transition from a NPB to a neural crest state. Using ChIP-seq we demonstrate that Sox3 is enriched upstream of NPB genes in early NPB cells and, surprisingly, in blastula stem cells. Depletion of SoxB1 factors in blastula stem cells results in downregulation of NPB genes. Finally, we identify Pou5f3 factors as a potential SoxB1 partners in regulating the formation of the NPB and show their combined activity is needed to maintain NPB gene expression. Together, these data identify a novel role for SoxB1 factors in the establishment and maintenance of the NPB, in part through partnership with Pou5f3 factors.

16.
bioRxiv ; 2023 Dec 22.
Article in English | MEDLINE | ID: mdl-38187687

ABSTRACT

The neural crest is vertebrate-specific stem cell population that helped drive the origin and evolution of the vertebrate clade. A distinguishing feature of these stem cells is their multi-germ layer potential, which has drawn developmental and evolutionary parallels to another stem cell population-pluripotent embryonic stem cells (animal pole cells or ES cells) of the vertebrate blastula. Here, we investigate the evolutionary origins of neural crest potential by comparing neural crest and pluripotency gene regulatory networks (GRNs) in both jawed ( Xenopus ) and jawless (lamprey) vertebrates. Through comparative gene expression analysis and transcriptomics, we reveal an ancient evolutionary origin of shared regulatory factors between neural crest and pluripotency GRNs that dates back to the last common ancestor of extant vertebrates. Focusing on the key pluripotency factor pou5 (formerly oct4), we show that the lamprey genome encodes a pou5 ortholog that is expressed in animal pole cells, as in jawed vertebrates, but is absent from the neural crest. However, gain-of-function experiments show that both lamprey and Xenopus pou5 enhance neural crest formation, suggesting that pou5 was lost from the neural crest of jawless vertebrates. Finally, we show that pou5 is required for neural crest specification in jawed vertebrates and that it acquired novel neural crest-enhancing activity after evolving from an ancestral pou3 -like clade that lacks this functionality. We propose that a pluripotency-neural crest GRN was assembled in stem vertebrates and that the multi-germ layer potential of the neural crest evolved by deploying this regulatory program.

17.
Proc Natl Acad Sci U S A ; 106(33): 13667-72, 2009 Aug 18.
Article in English | MEDLINE | ID: mdl-19666616

ABSTRACT

A transition metal complex targeted for the inhibition of a subset of zinc finger transcription factors has been synthesized and tested in Xenopus laevis. A Co(III) Schiff base complex modified with a 17-bp DNA sequence is designed to selectively inhibit Snail family transcription factors. The oligonucleotide-conjugated Co(III) complex prevents Slug, Snail, and Sip1 from binding their DNA targets whereas other transcription factors are still able to interact with their target DNA. The attachment of the oligonucleotide to the Co(III) complex increases specificity 150-fold over the unconjugated complex. Studies demonstrate that neither the oligo, or the Co(III) Schiff base complex alone, are sufficient for inactivation of Slug at concentrations that the conjugated complex mediates inhibition. Slug, Snail, and Sip1 have been implicated in the regulation of epithelial-to-mesenchymal transition in development and cancer. A complex targeted to inactivate their transcriptional activity could prove valuable as an experimental tool and a cancer therapeutic.


Subject(s)
Cobalt/chemistry , Gene Expression Regulation, Developmental , Schiff Bases/pharmacology , Transcription Factors/chemistry , Animals , Circular Dichroism , Cobalt/pharmacology , Homeodomain Proteins/antagonists & inhibitors , Homeodomain Proteins/metabolism , Magnetic Resonance Spectroscopy , Protein Structure, Secondary , Repressor Proteins/antagonists & inhibitors , Repressor Proteins/metabolism , Schiff Bases/chemistry , Snail Family Transcription Factors , Temperature , Transcription Factors/antagonists & inhibitors , Transcription Factors/metabolism , Xenopus Proteins/antagonists & inhibitors , Xenopus Proteins/metabolism , Xenopus laevis , Zinc Fingers
19.
Curr Opin Genet Dev ; 17(4): 326-31, 2007 Aug.
Article in English | MEDLINE | ID: mdl-17651964

ABSTRACT

Substantial progress has been made in defining the regulatory factors involved in generating multipotent neural crest cells at the neural plate border of vertebrate embryos, controlling the onset of their migratory behavior, and directing their differentiation into one of a diverse array of derivatives. Growing evidence suggests that these factors function as a complex network, in some cases displaying overlapping functions and cross-regulatory interactions. Mechanisms are emerging for how some of these regulatory components are controlled post-translationally and the extent to which their activities are conserved across species.


Subject(s)
Cell Differentiation/genetics , Cell Movement/genetics , Gene Expression Regulation, Developmental , Neural Crest/metabolism , Animals , Cell Differentiation/physiology , Cell Movement/physiology , Ectoderm/cytology , Ectoderm/metabolism , Models, Biological , Neural Crest/cytology , Snail Family Transcription Factors , Transcription Factors/genetics , Transcription Factors/physiology
20.
Nat Chem ; 13(7): 683-691, 2021 07.
Article in English | MEDLINE | ID: mdl-34155376

ABSTRACT

Mammalian oocytes undergo major changes in zinc content and localization to be fertilized, the most striking being the rapid exocytosis of over 10 billion zinc ions in what are known as zinc sparks. Here, we report that fertilization of amphibian Xenopus laevis eggs also initiates a zinc spark that progresses across the cell surface in coordination with dynamic calcium waves. This zinc exocytosis is accompanied by a newly recognized loss of intracellular manganese. Synchrotron-based X-ray fluorescence and analytical electron microscopy reveal that zinc and manganese are sequestered in a system of cortical granules that are abundant at the animal pole. Through electron-nuclear double-resonance studies, we rule out Mn2+ complexation with phosphate or nitrogenous ligands in intact eggs, but the data are consistent with a carboxylate coordination environment. Our observations suggest that zinc and manganese fluxes are a conserved feature of fertilization in vertebrates and that they function as part of a physiological block to polyspermy.


Subject(s)
Fertilization/physiology , Metals, Heavy/metabolism , Ovum/metabolism , Xenopus laevis/metabolism , Animals , Embryo, Nonmammalian/metabolism , Embryo, Nonmammalian/ultrastructure , Exocytosis/physiology , Fertilization/drug effects , Metals, Heavy/pharmacology , Ovum/drug effects , Ovum/ultrastructure
SELECTION OF CITATIONS
SEARCH DETAIL