Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 18 de 18
Filter
Add more filters










Publication year range
1.
Biol Open ; 12(1)2023 01 01.
Article in English | MEDLINE | ID: mdl-36524613

ABSTRACT

Stem cells cycle between periods of quiescence and proliferation to promote tissue health. In Drosophila ovaries, quiescence to proliferation transitions of follicle stem cells (FSCs) are exquisitely feeding-dependent. Here, we demonstrate feeding-dependent induction of follicle cell differentiation markers, eyes absent (Eya) and castor (Cas) in FSCs, a patterning process that does not depend on proliferation induction. Instead, FSCs extend micron-scale cytoplasmic projections that dictate Eya-Cas patterning. We identify still life and sickie as necessary and sufficient for FSC projection growth and Eya-Cas induction. Our results suggest that sequential, interdependent events establish long-term differentiation patterns in follicle cell precursors, independently of FSC proliferation induction.


Subject(s)
Drosophila Proteins , Ovary , Animals , Female , Ovary/metabolism , Drosophila/metabolism , Drosophila melanogaster/metabolism , Drosophila Proteins/genetics , Drosophila Proteins/metabolism , Cell Division , Cell Differentiation
2.
J Microbiol Biol Educ ; 22(3)2021 Dec.
Article in English | MEDLINE | ID: mdl-34880963

ABSTRACT

A call for the integration of research experiences into all biology curricula has been a major goal for educational reform efforts nationally. Course-based undergraduate research experiences (CUREs) have been the predominant method of accomplishing this, but their associated costs and complex design can limit their wide adoption. In 2020, the COVID-19 pandemic forced programs to identify unique ways to still provide authentic research experiences while students were virtual. We report here a complete guide for the successful implementation of a semester-long virtual CURE that uses Drosophila behavioral assays to explore the connection between pain and addiction with the use of an at-home "lab-in-a-box." Individual components were piloted across three semesters and launched as a 100-level introductory course with 19 students. We found that this course increased science identity and successfully improved key research competencies as per the Undergraduate Research Student Self-Assessment (URSSA) survey. This course is ideal for flipped classrooms ranging from introductory to upper-level biology/neuroscience courses and can be integrated directly into the lecture period without the need for building a new course. Given the low cost, recent comfort with virtual learning environments, and current proliferation of flipped classrooms following the 2020 pandemic, this curriculum could serve as an ideal project-based active-learning tool for equitably increasing access to authentic research experiences.

3.
J Pancreat Cancer ; 7(1): 39-47, 2021.
Article in English | MEDLINE | ID: mdl-34235374

ABSTRACT

Background: Sonic Hedgehog (Shh) is a tightly regulated membrane-associated morphogen and a known driver of tumorigenesis in pancreatic ductal adenocarcinoma (PDAC). After processing, Shh remains at the plasma membrane of Shh producing cells, thereby limiting its distribution and signal strength. In PDAC, the release of Shh from tumor cells is necessary to promote a tumor-permissive microenvironment. Mechanisms regulating Shh sequestration and/or release from tumor cells to signal distant stromal cells are not well known. Previously, our laboratory demonstrated that the Drosophila transmembrane protein Boi, sequesters Hh at the membrane of Hh-producing cells. In response to dietary cholesterol or in the absence of boi, Hh is constitutively released to promote proliferation in distant cells. In this study, we investigated the conservation of this mechanism in mammals by exploring the role of the human boi homolog, CDON, in PDAC. Methods: Using PDAC cell-lines BxPC-3, Capan-2, and MIA PaCa-2, along with normal pancreatic epithelial cells (PDEC), we investigated Shh expression via Immunoblot and real-time, quantitative polymerase chain reaction in addition to Shh release via enzyme-linked immunoassay following cholesterol treatment and/or transfection with either RNA interference to reduce CDON expression or with human CDON to increase expression. Results: Consistent with our Boi model, CDON suppresses Shh release, which is alleviated in response to dietary cholesterol. However, over-expressing CDON suppresses cholesterol-mediated Shh release in some PDAC contexts, which may be relative to the mutational burden of the cells. Conclusion: Identifying mechanisms that either sequester or stimulate Shh release from the tumor cell membrane may provide new avenues to reduce signaling between the tumor and its surrounding environment, which may restrain tumor development.

4.
STAR Protoc ; 2(2): 100592, 2021 06 18.
Article in English | MEDLINE | ID: mdl-34169286

ABSTRACT

We have outlined the approach of visualizing autophagy specifically in the epithelial follicle stem cells of the Drosophila ovary using the LysoTracker dye. The advantage of using this protocol is that it details several techniques, including ovary dissection, immunofluorescence, and western blotting, that positively identify autophagy changes in a very small population of cells. One of the limitations of this protocol is that it needs to be combined with other genetic manipulations and positive markers of the autophagy pathway. For complete details on the use and execution of this protocol, please refer to Singh et al., (2018).


Subject(s)
Autophagy , Cell Tracking , Ovarian Follicle/cytology , Stem Cells/cytology , Animals , Drosophila , Epithelial Cells/cytology , Female , Microscopy, Confocal
5.
AIMS Public Health ; 8(2): 352-368, 2021.
Article in English | MEDLINE | ID: mdl-34017897

ABSTRACT

Despite the clinically proven benefits of the human papillomavirus (HPV) vaccine in preventing cervical and other HPV-associated cancers, vaccination coverage has been suboptimal among adolescents and young adults in the United States (US), particularly among racial and ethnic minority adolescents. Historical legacies, combined with current racial/ethnic disparities in healthcare, may contribute to suboptimal uptake and completion of the HPV vaccine in part through differing levels of trust in doctors and healthcare institutions. The purpose of this narrative review was to characterize trust and its role in decision making about HPV vaccine uptake among US racial and ethnic minorities. We conducted a literature search using the PubMed database, and our search terms yielded 1176 articles. We reviewed 41 full-text articles for eligibility and included 20 articles in this review. These studies used varied measures of trust or mistrust and assessed trust in not only doctors/healthcare providers, but also other sources including pharmaceutical companies, media, and clergy. Our review findings revealed generally high levels of trust in doctors and healthcare providers, but less so in pharmaceutical companies. Mistrust of either healthcare providers, government agencies or pharmaceutical companies was consistently associated with less favorable attitudes and lower vaccine uptake. The downstream effects of mistrust may occur through selected health beliefs regarding the perceived efficacy and safety of the vaccine. Minority groups were more likely to report trust in family members, religious organizations, and media sources compared to their white counterparts. Decision making about vaccine uptake is a multilayered process that involves comparing the perceived benefits of the vaccine against its perceived risks. Understanding how trusted sources can effectively harness the tools of social and traditional media to increase knowledge and awareness may help combat misinformation about the HPV vaccine and improve engagement with diverse communities.

6.
Cancer Cell ; 38(4): 567-583.e11, 2020 10 12.
Article in English | MEDLINE | ID: mdl-32976774

ABSTRACT

Oncogenic transformation alters lipid metabolism to sustain tumor growth. We define a mechanism by which cholesterol metabolism controls the development and differentiation of pancreatic ductal adenocarcinoma (PDAC). Disruption of distal cholesterol biosynthesis by conditional inactivation of the rate-limiting enzyme Nsdhl or treatment with cholesterol-lowering statins switches glandular pancreatic carcinomas to a basal (mesenchymal) phenotype in mouse models driven by KrasG12D expression and homozygous Trp53 loss. Consistently, PDACs in patients receiving statins show enhanced mesenchymal features. Mechanistically, statins and NSDHL loss induce SREBP1 activation, which promotes the expression of Tgfb1, enabling epithelial-mesenchymal transition. Evidence from patient samples in this study suggests that activation of transforming growth factor ß signaling and epithelial-mesenchymal transition by cholesterol-lowering statins may promote the basal type of PDAC, conferring poor outcomes in patients.


Subject(s)
Biosynthetic Pathways/genetics , Carcinoma, Pancreatic Ductal/genetics , Cholesterol, LDL/biosynthesis , Pancreatic Neoplasms/genetics , Transforming Growth Factor beta/genetics , 3-Hydroxysteroid Dehydrogenases/genetics , 3-Hydroxysteroid Dehydrogenases/metabolism , Animals , Atorvastatin/pharmacology , Carcinoma, Pancreatic Ductal/drug therapy , Carcinoma, Pancreatic Ductal/metabolism , Cell Differentiation/drug effects , Cell Differentiation/genetics , Cell Line, Tumor , Epithelial-Mesenchymal Transition/drug effects , Epithelial-Mesenchymal Transition/genetics , Gene Expression Profiling , Gene Expression Regulation, Neoplastic/drug effects , Humans , Hydroxymethylglutaryl-CoA Reductase Inhibitors/pharmacology , Kaplan-Meier Estimate , Mice, Inbred C57BL , Mice, Knockout , Pancreatic Neoplasms/drug therapy , Pancreatic Neoplasms/metabolism , Signal Transduction/genetics , Transforming Growth Factor beta/metabolism , Xenograft Model Antitumor Assays/methods
7.
Biol Open ; 9(7)2020 07 21.
Article in English | MEDLINE | ID: mdl-32580972

ABSTRACT

Polymerization of metabolic enzymes into micron-scale assemblies is an emerging mechanism for regulating their activity. CTP synthase (CTPS) is an essential enzyme in the biosynthesis of the nucleotide CTP and undergoes regulated and reversible assembly into large filamentous structures in organisms from bacteria to humans. The purpose of these assemblies is unclear. A major challenge to addressing this question has been the inability to abolish assembly without eliminating CTPS protein. Here we demonstrate that a recently reported point mutant in CTPS, Histidine 355A (H355A), prevents CTPS filament assembly in vivo and dominantly inhibits the assembly of endogenous wild-type CTPS in the Drosophila ovary. Expressing this mutant in ovarian germline cells, we show that disruption of CTPS assembly in early stage egg chambers reduces egg production. This effect is exacerbated in flies fed the glutamine antagonist 6-diazo-5-oxo-L-norleucine, which inhibits de novo CTP synthesis. These findings introduce a general approach to blocking the assembly of polymerizing enzymes without eliminating their catalytic activity and demonstrate a role for CTPS assembly in supporting egg production, particularly under conditions of limited glutamine metabolism.This article has an associated First Person interview with the first author of the paper.


Subject(s)
Carbon-Nitrogen Ligases/metabolism , Drosophila/physiology , Germ Cells/metabolism , Protein Multimerization , Reproduction , Animals , Carbon-Nitrogen Ligases/chemistry , Carbon-Nitrogen Ligases/genetics , Energy Metabolism , Fluorescent Antibody Technique , Gene Expression , Glutamine/metabolism , Mutation
8.
PLoS Genet ; 15(7): e1008218, 2019 Jul.
Article in English | MEDLINE | ID: mdl-31295252
9.
10.
Dev Cell ; 46(6): 720-734.e6, 2018 09 24.
Article in English | MEDLINE | ID: mdl-30197240

ABSTRACT

Egg production declines with age in many species, a process linked with stem cell loss. Diet-dependent signaling has emerged as critical for stem cell maintenance during aging. Follicle stem cells (FSCs) in the Drosophila ovary are exquisitely responsive to diet-induced signals including Hedgehog (Hh) and insulin-IGF signaling (IIS), entering quiescence in the absence of nutrients and initiating proliferation rapidly upon feeding. Although highly proliferative FSCs generally exhibit an extended lifespan, we find that constitutive Hh signaling drives FSC loss and premature sterility despite high proliferative rates. This occurs due to Hh-mediated induction of autophagy in FSCs via a Ptc-dependent, Smo-independent mechanism. Hh-dependent autophagy increases during aging, triggering FSC loss and consequent reproductive arrest. IIS is necessary and sufficient to suppress Hh-induced autophagy, promoting a stable proliferative state. These results suggest that opposing action of diet-responsive IIS and Hh signals determine reproductive lifespan by modulating the proliferation-autophagy balance in FSCs during aging.


Subject(s)
Autophagy , Cell Proliferation , Drosophila Proteins/metabolism , Drosophila melanogaster/physiology , Hedgehog Proteins/metabolism , Insulin/pharmacology , Ovarian Follicle/cytology , Stem Cells/cytology , Animals , Drosophila Proteins/genetics , Drosophila melanogaster/drug effects , Female , Hedgehog Proteins/genetics , Hypoglycemic Agents/pharmacology , Insulin-Like Growth Factor I/genetics , Insulin-Like Growth Factor I/metabolism , Male , Ovarian Follicle/drug effects , Ovarian Follicle/metabolism , Receptors, Cell Surface/genetics , Receptors, Cell Surface/metabolism , Signal Transduction/drug effects , Smoothened Receptor/genetics , Smoothened Receptor/metabolism , Stem Cell Niche/drug effects , Stem Cells/drug effects , Stem Cells/metabolism
11.
Biochemistry ; 55(11): 1663-72, 2016 Mar 22.
Article in English | MEDLINE | ID: mdl-26836402

ABSTRACT

Histone modification plays a major role in regulating gene transcription and ensuring the healthy development of an organism. Numerous studies have suggested that histones are dynamically modified during developmental events to control gene expression levels in a temporal and spatial manner. However, the study of histone acetylation dynamics using currently available techniques is hindered by the difficulty of simultaneously measuring acetylation of the numerous potential sites of modification present in histones. Here, we present a methodology that allows us to combine mass spectrometry-based histone analysis with Drosophila developmental genetics. Using this system, we characterized histone acetylation patterns during multiple developmental stages of the fly. Additionally, we utilized this analysis to characterize how treatments with pharmacological agents or environmental changes such as γ-irradiation altered histone acetylation patterns. Strikingly, γ-irradiation dramatically increased the level of acetylation at H3K18, a site linked to DNA repair via nonhomologous end joining. In mutant fly strains deficient in DNA repair proteins, however, this increase in the level of H3K18 acetylation was lost. These results demonstrate the efficacy of our combined mass spectrometry system with a Drosophila model system and provide interesting insight into the changes in histone acetylation during development, as well as the effects of both pharmacological and environmental agents on global histone acetylation.


Subject(s)
DNA Repair , Drosophila Proteins/metabolism , Gamma Rays , Histones/metabolism , Transcription, Genetic/radiation effects , Acetylation , Animals , Drosophila Proteins/genetics , Drosophila melanogaster , Histones/genetics , Mutation
12.
Genetics ; 199(4): 935-57, 2015 Apr.
Article in English | MEDLINE | ID: mdl-25680813

ABSTRACT

In many tissues, the presence of stem cells is inferred by the capacity of the tissue to maintain homeostasis and undergo repair after injury. Isolation of self-renewing cells with the ability to generate the full array of cells within a given tissue strongly supports this idea, but the identification and genetic manipulation of individual stem cells within their niche remain a challenge. Here we present novel methods for marking and genetically altering epithelial follicle stem cells (FSCs) within the Drosophila ovary. Using these new tools, we define a sequential multistep process that comprises transitioning of FSCs from quiescence to proliferation. We further demonstrate that integrins are cell-autonomously required within FSCs to provide directional signals that are necessary at each step of this process. These methods may be used to define precise roles for specific genes in the sequential events that occur during FSC division after a period of quiescence.


Subject(s)
Drosophila melanogaster/genetics , Gene Expression Profiling/methods , Genome, Insect , Integrins/metabolism , Ovarian Follicle/cytology , Stem Cells/metabolism , Animals , Cell Proliferation , Drosophila melanogaster/cytology , Female , Integrins/genetics , Male , Stem Cells/cytology , Stem Cells/physiology
13.
EMBO Rep ; 15(11): 1184-91, 2014 Nov.
Article in English | MEDLINE | ID: mdl-25223282

ABSTRACT

The enzyme CTP synthase (CTPS) dynamically assembles into macromolecular filaments in bacteria, yeast, Drosophila, and mammalian cells, but the role of this morphological reorganization in regulating CTPS activity is controversial. During Drosophila oogenesis, CTPS filaments are transiently apparent in ovarian germline cells during a period of intense genomic endoreplication and stockpiling of ribosomal RNA. Here, we demonstrate that CTPS filaments are catalytically active and that their assembly is regulated by the non-receptor tyrosine kinase DAck, the Drosophila homologue of mammalian Ack1 (activated cdc42-associated kinase 1), which we find also localizes to CTPS filaments. Egg chambers from flies deficient in DAck or lacking DAck catalytic activity exhibit disrupted CTPS filament architecture and morphological defects that correlate with reduced fertility. Furthermore, ovaries from these flies exhibit reduced levels of total RNA, suggesting that DAck may regulate CTP synthase activity. These findings highlight an unexpected function for DAck and provide insight into a novel pathway for the developmental control of an essential metabolic pathway governing nucleotide biosynthesis.


Subject(s)
Carbon-Nitrogen Ligases/metabolism , Drosophila Proteins/metabolism , Drosophila/metabolism , Oogenesis , Protein-Tyrosine Kinases/metabolism , Animals , Carbon-Nitrogen Ligases/genetics , Drosophila/genetics , Drosophila/physiology , Drosophila Proteins/genetics , Female , Ovary/metabolism , Protein Transport , Protein-Tyrosine Kinases/genetics , RNA/metabolism
14.
J Cell Biol ; 201(5): 741-57, 2013 May 27.
Article in English | MEDLINE | ID: mdl-23690177

ABSTRACT

A healthy diet improves adult stem cell function and delays diseases such as cancer, heart disease, and neurodegeneration. Defining molecular mechanisms by which nutrients dictate stem cell behavior is a key step toward understanding the role of diet in tissue homeostasis. In this paper, we elucidate the mechanism by which dietary cholesterol controls epithelial follicle stem cell (FSC) proliferation in the fly ovary. In nutrient-restricted flies, the transmembrane protein Boi sequesters Hedgehog (Hh) ligand at the surface of Hh-producing cells within the ovary, limiting FSC proliferation. Upon feeding, dietary cholesterol stimulates S6 kinase-mediated phosphorylation of the Boi cytoplasmic domain, triggering Hh release and FSC proliferation. This mechanism enables a rapid, tissue-specific response to nutritional changes, tailoring stem cell divisions and egg production to environmental conditions sufficient for progeny survival. If conserved in other systems, this mechanism will likely have important implications for studies on molecular control of stem cell function, in which the benefits of low calorie and low cholesterol diets are beginning to emerge.


Subject(s)
Cell Proliferation/drug effects , Cholesterol, Dietary/pharmacology , Drosophila Proteins/metabolism , Drosophila melanogaster/drug effects , Hedgehog Proteins/metabolism , Ovarian Follicle/drug effects , Stem Cells/drug effects , Animals , Carrier Proteins/metabolism , Carrier Proteins/physiology , Drosophila Proteins/physiology , Drosophila melanogaster/metabolism , Female , Ovarian Follicle/cytology , Ovarian Follicle/metabolism , Ovary/cytology , Ovary/drug effects , Ovary/metabolism , Phosphorylation , Ribosomal Protein S6 Kinases/metabolism , Stem Cells/cytology , Stem Cells/metabolism
15.
J Cell Biol ; 191(5): 943-52, 2010 Nov 29.
Article in English | MEDLINE | ID: mdl-21098113

ABSTRACT

Stem cells depend on signals from cells within their microenvironment, or niche, as well as factors secreted by distant cells to regulate their maintenance and function. Here we show that Boi, a Hedgehog (Hh)-binding protein, is a novel suppressor of proliferation of follicle stem cells (FSCs) in the Drosophila ovary. Hh is expressed in apical cells, distant from the FSC niche, and diffuses to reach FSCs, where it promotes FSC proliferation. We show that Boi is expressed in apical cells and exerts its suppressive effect on FSC proliferation by binding to and sequestering Hh on the apical cell surface, thereby inhibiting Hh diffusion. Our studies demonstrate that cells distant from the local niche can regulate stem cell function through ligand sequestration, a mechanism that likely is conserved in other epithelial tissues.


Subject(s)
Carrier Proteins/metabolism , Cell Proliferation , Drosophila Proteins/metabolism , Drosophila/metabolism , Hedgehog Proteins/metabolism , Ovarian Follicle/cytology , Stem Cells/cytology , Animals , Animals, Genetically Modified , Carrier Proteins/genetics , Drosophila Proteins/genetics , Embryo, Nonmammalian/metabolism , Female , Hedgehog Proteins/genetics , Ovarian Follicle/metabolism , Stem Cells/metabolism
16.
PLoS One ; 4(12): e8106, 2009 Dec 01.
Article in English | MEDLINE | ID: mdl-19956620

ABSTRACT

BACKGROUND: The coordinated action of genes that control patterning, cell fate determination, cell size, and cell adhesion is required for proper wing formation in Drosophila. Defects in any of these basic processes can lead to wing aberrations, including blisters. The xenicid mutation was originally identified in a screen designed to uncover regulators of adhesion between wing surfaces [1]. PRINCIPAL FINDINGS: Here, we demonstrate that expression of the betaPS integrin or the patterning protein Engrailed are not affected in developing wing imaginal discs in xenicid mutants. Instead, expression of the homeotic protein Ultrabithorax (Ubx) is strongly increased in xenicid mutant cells. CONCLUSION: Our results suggest that upregulation of Ubx transforms cells from a wing blade fate to a haltere fate, and that the presence of haltere cells within the wing blade is the primary defect leading to the adult wing phenotypes observed.


Subject(s)
Alleles , Drosophila Proteins/genetics , Drosophila melanogaster/genetics , Repressor Proteins/genetics , Sequence Deletion , Wings, Animal/pathology , Animals , Cell Lineage , Cell Size , Clone Cells , Drosophila Proteins/metabolism , Drosophila melanogaster/cytology , Homeodomain Proteins/genetics , Homeodomain Proteins/metabolism , Integrins/metabolism , Phenotype , Protein Transport , Transcription Factors/genetics , Transcription Factors/metabolism , Up-Regulation/genetics
17.
J Cell Biol ; 182(4): 801-15, 2008 Aug 25.
Article in English | MEDLINE | ID: mdl-18725542

ABSTRACT

Adult stem cells are maintained in specialized microenvironments called niches, which promote self-renewal and prevent differentiation. In this study, we show that follicle stem cells (FSCs) in the Drosophila melanogaster ovary rely on cues that are distinct from those of other ovarian stem cells to establish and maintain their unique niche. We demonstrate that integrins anchor FSCs to the basal lamina, enabling FSCs to maintain their characteristic morphology and position. Integrin-mediated FSC anchoring is also essential for proper development of differentiating prefollicle cells that arise from asymmetrical FSC divisions. Our results support a model in which FSCs contribute to the formation and maintenance of their own niche by producing the integrin ligand, laminin A (LanA). Together, LanA and integrins control FSC proliferation rates, a role that is separable from their function in FSC anchoring. Importantly, LanA-integrin function is not required to maintain other ovarian stem cell populations, demonstrating that distinct pathways regulate niche-stem cell communication within the same organ.


Subject(s)
Cell Polarity , Drosophila melanogaster/cytology , Integrins/metabolism , Ovarian Follicle/cytology , Stem Cells/cytology , Actins/metabolism , Animals , Cell Proliferation , Cell Shape , Drosophila melanogaster/genetics , Female , Genes, Insect , Laminin/metabolism , Models, Biological , Mutation/genetics , Protein Transport , Time Factors
18.
Development ; 133(14): 2627-38, 2006 Jul.
Article in English | MEDLINE | ID: mdl-16775001

ABSTRACT

The Src family protein tyrosine kinases (SFKs) are crucial regulators of cellular morphology. In Drosophila, Src64 controls complex morphological events that occur during oogenesis. Recent studies have identified key Src64-dependent mechanisms that regulate actin cytoskeletal dynamics during the growth of actin-rich ring canals, which act as intercellular bridges between germ cells. By contrast, the molecular mechanisms that regulate Src64 activity levels and potential roles for Src64 in additional morphological events in the ovary have not been defined. In this report, we demonstrate that regulation of Src64 by Drosophila C-terminal-Src Kinase (Csk) contributes to the packaging of germline cysts by overlying somatic follicle cells during egg chamber formation. These results uncover novel roles for both Csk and Src64 in a dynamic event that involves adhesion, communication between cell types and control of cell motility. Strikingly, Src64 and Csk function in the germline to control packaging, not in migrating follicle cells, suggesting novel functions for this signaling cassette in regulating dynamic adhesion. In contrast to the role played by Csk in the regulation of Src64 activity during packaging, Csk is dispensable for ring canal growth control, indicating that distinct mechanisms control Src64 activity during different morphological events.


Subject(s)
Drosophila melanogaster/cytology , Drosophila melanogaster/embryology , Gene Expression Regulation, Developmental , Germ Cells/physiology , Morphogenesis , Animals , Animals, Genetically Modified , CSK Tyrosine-Protein Kinase , Cell Differentiation , Cell Shape , Drosophila Proteins , Drosophila melanogaster/genetics , Drosophila melanogaster/metabolism , Enzyme Activation , Female , Fertility/physiology , Germ Cells/cytology , Mutation , Oocytes/cytology , Oocytes/physiology , Ovary/anatomy & histology , Ovary/metabolism , Ovary/pathology , Protein-Tyrosine Kinases , Proto-Oncogene Proteins , src Homology Domains , src-Family Kinases
SELECTION OF CITATIONS
SEARCH DETAIL
...