Network


Latest external collaboration on country level. Dive into details by clicking on the dots.

Hotspot


Dive into the research topics where Tony DeFalco is active.

Publication


Featured researches published by Tony DeFalco.


PLOS Genetics | 2012

Temporal transcriptional profiling of somatic and germ cells reveals biased lineage priming of sexual fate in the fetal mouse gonad.

Samantha Jameson; Anirudh Natarajan; Jonah Cool; Tony DeFalco; Danielle M. Maatouk; Lindsey Mork; Steven C. Munger; Blanche Capel

The divergence of distinct cell populations from multipotent progenitors is poorly understood, particularly in vivo. The gonad is an ideal place to study this process, because it originates as a bipotential primordium where multiple distinct lineages acquire sex-specific fates as the organ differentiates as a testis or an ovary. To gain a more detailed understanding of the process of gonadal differentiation at the level of the individual cell populations, we conducted microarrays on sorted cells from XX and XY mouse gonads at three time points spanning the period when the gonadal cells transition from sexually undifferentiated progenitors to their respective sex-specific fates. We analyzed supporting cells, interstitial/stromal cells, germ cells, and endothelial cells. This work identified genes specifically depleted and enriched in each lineage as it underwent sex-specific differentiation. We determined that the sexually undifferentiated germ cell and supporting cell progenitors showed lineage priming. We found that germ cell progenitors were primed with a bias toward the male fate. In contrast, supporting cells were primed with a female bias, indicative of the robust repression program involved in the commitment to XY supporting cell fate. This study provides a molecular explanation reconciling the female default and balanced models of sex determination and represents a rich resource for the field. More importantly, it yields new insights into the mechanisms by which different cell types in a single organ adopt their respective fates.


Annual Review of Cell and Developmental Biology | 2009

Gonad Morphogenesis in Vertebrates: Divergent Means to a Convergent End

Tony DeFalco; Blanche Capel

A critical element of successful sexual reproduction is the generation of sexually dimorphic adult reproductive organs, the testis and ovary, which produce functional gametes. Examination of different vertebrate species shows that the adult gonad is remarkably similar in its morphology across different phylogenetic classes. Surprisingly, however, the cellular and molecular programs employed to create similar organs are not evolutionarily conserved. We highlight the mechanisms used by different vertebrate model systems to generate the somatic architecture necessary to support gametogenesis. In addition, we examine the different vertebrate patterns of germ cell migration from their site of origin to colonize the gonad and highlight their roles in sex-specific morphogenesis. We also discuss the plasticity of the adult gonad and consider how different genetic and environmental conditions can induce transitions between testis and ovary morphology.


Developmental Biology | 2011

Two distinct origins for Leydig cell progenitors in the fetal testis.

Tony DeFalco; Satoru Takahashi; Blanche Capel

During the differentiation of the mammalian embryonic testis, two compartments are defined: the testis cords and the interstitium. The testis cords give rise to the adult seminiferous tubules, whereas steroidogenic Leydig cells and other less well characterized cell types differentiate in the interstitium (the space between testis cords). Although the process of testis cord formation is essential for male development, it is not entirely understood. It has been viewed as a Sertoli-cell driven process, but growing evidence suggests that interstitial cells play an essential role during testis formation. However, little is known about the origin of the interstitium or the molecular and cellular diversity within this early stromal compartment. To better understand the process of mammalian gonad differentiation, we have undertaken an analysis of developing interstitial/stromal cells in the early mouse testis and ovary. We have discovered molecular heterogeneity in the interstitium and have characterized new markers of distinct cell types in the gonad: MAFB, C-MAF, and VCAM1. Our results show that at least two distinct progenitor lineages give rise to the interstitial/stromal compartment of the gonad: the coelomic epithelium and specialized cells along the gonad-mesonephros border. We demonstrate that both these populations give rise to interstitial precursors that can differentiate into fetal Leydig cells. Our analysis also reveals that perivascular cells migrate into the gonad from the mesonephric border along with endothelial cells and that these vessel-associated cells likely represent an interstitial precursor lineage. This study highlights the cellular diversity of the interstitial cell population and suggests that complex cell-cell interactions among cells in the interstitium are involved in testis morphogenesis.


Proceedings of the National Academy of Sciences of the United States of America | 2011

Vascular-mesenchymal cross-talk through Vegf and Pdgf drives organ patterning

Jonah Cool; Tony DeFalco; Blanche Capel

The initiation of de novo testis cord organization in the fetal gonad is poorly understood. Endothelial cell migration into XY gonads initiates testis morphogenesis. However, neither the signals that regulate vascularization of the gonad nor the mechanisms through which vessels affect tissue morphogenesis are known. Here, we show that Vegf signaling is required for gonad vascularization and cord morphogenesis. We establish that interstitial cells express Vegfa and respond, by proliferation, to endothelial migration. In the absence of vasculature, four-dimensional imaging of whole organs revealed that interstitial proliferation is reduced and prevents formation of wedge-like structures that partition the gonad into cord-forming domains. Antagonizing vessel maturation also reduced proliferation. However, proliferation of mesenchymal cells was rescued by the addition of PDGF-BB. These results suggest a pathway that integrates initiation of vascular development and testis cord morphogenesis, and lead to a model in which undifferentiated mesenchyme recruits blood vessels, proliferates in response, and performs a primary function in the morphogenesis and patterning of the developing organ.


Cell Reports | 2015

Macrophages Contribute to the Spermatogonial Niche in the Adult Testis

Tony DeFalco; Sarah J. Potter; Alyna V. Williams; Brittain Waller; Matthew J. Kan; Blanche Capel

The testis produces sperm throughout the male reproductive lifespan by balancing self-renewal and differentiation of spermatogonial stem cells (SSCs). Part of the SSC niche is thought to lie outside the seminiferous tubules of the testis; however, specific interstitial components of the niche that regulate spermatogonial divisions and differentiation remain undefined. We identified distinct populations of testicular macrophages, one of which lies on the surface of seminiferous tubules, in close apposition to areas of tubules enriched for undifferentiated spermatogonia. These macrophages express spermatogonial proliferation- and differentiation-inducing factors, such as colony-stimulating factor 1 (CSF1) and enzymes involved in retinoic acid (RA) biosynthesis. We show that transient depletion of macrophages leads to a disruption in spermatogonial differentiation. These findings reveal an unexpected role for macrophages in the spermatogonial niche in the testis and raise the possibility that macrophages play previously unappreciated roles in stem/progenitor cell regulation in other tissues.


Proceedings of the National Academy of Sciences of the United States of America | 2014

Yolk-sac–derived macrophages regulate fetal testis vascularization and morphogenesis

Tony DeFalco; Indrashis Bhattacharya; Alyna V. Williams; Dustin M. Sams; Blanche Capel

Significance A main requisite for organogenesis is the integration of vascular networks, which not only deliver oxygen and nutrients but also serve instructive roles in organ patterning that determine how organs develop into their final structure and function in the adult. Our previous work showed that vascularization is essential to induce formation of the primitive cord structures that give rise to the seminiferous tubules of the adult testis. Here we show that fetal macrophages are required to remodel the vasculature and refine organ compartments during differentiation of the gonad. This study reveals an underappreciated and likely vital role for macrophages in fetal organogenesis that may be relevant to the development of many organs. Organogenesis of the testis is initiated when expression of Sry in pre-Sertoli cells directs the gonad toward a male-specific fate. The cells in the early bipotential gonad undergo de novo organization to form testis cords that enclose germ cells inside tubules lined by epithelial Sertoli cells. Although Sertoli cells are a driving force in the de novo formation of testis cords, recent studies in mouse showed that reorganization of the vasculature and of interstitial cells also play critical roles in testis cord morphogenesis. However, the mechanism driving reorganization of the vasculature during fetal organogenesis remained unclear. Here we demonstrate that fetal macrophages are associated with nascent gonadal and mesonephric vasculature during the initial phases of testis morphogenesis. Macrophages mediate vascular reorganization and prune errant germ cells and somatic cells after testis architecture is established. We show that gonadal macrophages are derived from primitive yolk-sac hematopoietic progenitors and exhibit hallmarks of M2 activation status, suggestive of angiogenic and tissue remodeling functions. Depletion of macrophages resulted in impaired vascular reorganization and abnormal cord formation. These findings reveal a previously unappreciated role for macrophages in testis morphogenesis and suggest that macrophages are an intermediary between neovascularization and organ architecture during fetal organogenesis.


Developmental Biology | 2013

Constitutive activation of NOTCH1 signaling in Sertoli cells causes gonocyte exit from quiescence

Thomas Garcia; Tony DeFalco; Blanche Capel; Marie Claude Hofmann

Notch signaling components have long been detected in Sertoli and germ cells in the developing and mature testis. However, the role of this pathway in testis development and spermatogenesis remains unknown. Using reporter mice expressing green fluorescent protein following Notch receptor activation, we found that Notch signaling was active in Sertoli cells at various fetal, neonatal, and adult stages. Since Notch signaling specifies stem cell fate in many developing and mature organ systems, we hypothesized that maintenance and differentiation of gonocytes and/or spermatogonial stem cells would be modulated through this pathway in Sertoli cells. To this end, we generated mutant mice constitutively expressing the active, intracellular domain of NOTCH1 (NICD1) in Sertoli cells. We found that mutant Sertoli cells were morphologically normal before and after birth, but presented a number of functional changes that drastically affected gonocyte numbers and physiology. We observed aberrant exit of gonocytes from mitotic arrest, migration toward cord periphery, and premature differentiation before birth. These events, presumably unsupported by the cellular microenvironment, were followed by gonocyte apoptosis and near complete disappearance of the gonocytes by day 2 after birth. Molecular analysis demonstrated that these effects are correlated with a dysregulation of Sertoli-expressed genes that are required for germ cell maintenance, such as Cyp26b1 and Gdnf. Taken together, our results demonstrate that Notch signaling is active in Sertoli cells throughout development and that proper regulation of Notch signaling in Sertoli cells is required for the maintenance of gonocytes in an undifferentiated state during fetal development.


Biology of Reproduction | 2013

Testosterone Levels Influence Mouse Fetal Leydig Cell Progenitors Through Notch Signaling

Tony DeFalco; Anirudh Saraswathula; Anaı̈s Briot; M. Luisa Iruela-Arispe; Blanche Capel

ABSTRACT Leydig cells are the steroidogenic lineage of the mammalian testis that produces testosterone, a key hormone required throughout male fetal and adult life for virilization and spermatogenesis. Both fetal and adult Leydig cells arise from a progenitor population in the testis interstitium but are thought to be lineage-independent of one another. Genetic evidence indicates that Notch signaling is required during fetal life to maintain a balance between differentiated Leydig cells and their progenitors, but the elusive progenitor cell type and ligands involved have not been identified. In this study, we show that the Notch pathway signals through the ligand JAG1 in perivascular interstitial cells during fetal life. In the early postnatal testis, we show that circulating levels of testosterone directly affect Notch signaling, implicating a feedback role for systemic circulating factors in the regulation of progenitor cells. Between Postnatal Days 3 and 21, as fetal Leydig cells disappear from the testis and are replaced by adult Leydig cells, the perivascular population of interstitial cells active for Notch signaling declines, consistent with distinct regulation of adult Leydig progenitors.


Wiley Interdisciplinary Reviews-Developmental Biology | 2012

Testis formation in the fetal mouse: dynamic and complex de novo tubulogenesis.

Jonah Cool; Tony DeFalco; Blanche Capel

Soon after Sry initiates male sex determination, cells in XY gonads undergo an unusual process of de novo cord formation that results in the organization of Sertoli cells into epithelial tubules enclosing germ cells and partitioning mesenchymal cells and vasculature to the interstitial space of the testis. Recent experiments investigating this dynamic process in four dimensions have begun to shed new light on the collective interactions of multiple cell types during morphogenesis of testis cords. WIREs Dev Biol 2012 doi: 10.1002/wdev.62


Endocrinology | 2014

Lactoferrin-iCre: A New Mouse Line to Study Uterine Epithelial Gene Function

Takiko Daikoku; Yuya Ogawa; Jumpei Terakawa; Akiyo Ogawa; Tony DeFalco; Sudhansu K. Dey

Transgenic animal models are valuable for studying gene function in various tissue compartments. Mice with conditional deletion of genes in the uterus using the Cre-loxP system serve as powerful tools to study uterine biology. The uterus is comprised of 3 major tissue types: myometrium, stroma, and epithelium. Proliferation and differentiation in each uterine cell type are differentially regulated by ovarian hormones, resulting in spatiotemporal control of gene expression. Therefore, examining gene function in each uterine tissue type will provide more meaningful information regarding uterine biology during pregnancy and disease states. Although currently available Cre mouse lines have been very useful in exploring functions of specific genes in uterine biology, overlapping expression of these Cre lines in more than 1 tissue type and in other reproductive organs sometimes makes interpretation of results difficult. In this article, we report the generation of a new iCre knock-in mouse line, in which iCre is expressed from endogenous lactoferrin (Ltf) promoter. Ltf-iCre mice primarily direct recombination in the uterine epithelium in adult females and in immature females after estrogen treatment. These mice will allow for specific interrogation of gene function in the mature uterine epithelium, providing a helpful tool to uncover important aspects of uterine biology.

Collaboration


Dive into the Tony DeFalco's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar

Sarah J. Potter

Cincinnati Children's Hospital Medical Center

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Andrew J. Kim

Cincinnati Children's Hospital Medical Center

View shared research outputs
Top Co-Authors

Avatar

Deepti Lava Kumar

Cincinnati Children's Hospital Medical Center

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Alyna V. Williams

Cincinnati Children's Hospital Medical Center

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge