Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 38
Filtrar
1.
Development ; 149(1)2022 01 01.
Artigo em Inglês | MEDLINE | ID: mdl-34989394

RESUMO

Fluid secretion by exocrine glandular organs is essential to the survival of mammals. Each glandular unit within the body is uniquely organized to carry out its own specific functions, with failure to establish these specialized structures resulting in impaired organ function. Here, we review glandular organs in terms of shared and divergent architecture. We first describe the structural organization of the diverse glandular secretory units (the end-pieces) and their fluid transporting systems (the ducts) within the mammalian system, focusing on how tissue architecture corresponds to functional output. We then highlight how defects in development of end-piece and ductal architecture impacts secretory function. Finally, we discuss how knowledge of exocrine gland structure-function relationships can be applied to the development of new diagnostics, regenerative approaches and tissue regeneration.


Assuntos
Glândulas Exócrinas/anatomia & histologia , Morfogênese , Animais , Glândulas Exócrinas/embriologia , Glândulas Exócrinas/fisiologia , Humanos
2.
Development ; 149(11)2022 06 01.
Artigo em Inglês | MEDLINE | ID: mdl-35575097

RESUMO

The uterine luminal epithelium folds characteristically in mammals, including humans, horses and rodents. Improper uterine folding in horses results in pregnancy failure, but the precise function of folds remains unknown. Here, we uncover dynamic changes in the 3D uterine folding pattern during early pregnancy with the entire lumen forming pre-implantation transverse folds along the mesometrial-antimesometrial axis. Using a time course, we show that transverse folds are formed before embryo spacing, whereas implantation chambers form as the embryo begins attachment. Thus, folds and chambers are two distinct structures. Transverse folds resolve to form a flat implantation region, after which an embryo arrives at its center to attach and form the post-implantation chamber. Our data also suggest that the implantation chamber facilitates embryo rotation and its alignment along the uterine mesometrial-antimesometrial axis. Using WNT5A- and RBPJ-deficient mice that display aberrant folds, we show that embryos trapped in longitudinal folds display misalignment of the embryo-uterine axes, abnormal chamber formation and defective post-implantation morphogenesis. These mouse models with disrupted uterine folding provide an opportunity to understand uterine structure-based mechanisms that are crucial for implantation and pregnancy success. This article has an associated 'The people behind the papers' interview.


Assuntos
Implantação do Embrião , Útero , Animais , Embrião de Mamíferos , Epitélio , Feminino , Cavalos , Humanos , Mamíferos , Camundongos , Gravidez
3.
Proc Natl Acad Sci U S A ; 119(35): e2123267119, 2022 08 30.
Artigo em Inglês | MEDLINE | ID: mdl-35994660

RESUMO

The pregnant uterus is an immunologically rich organ, with dynamic changes in the inflammatory milieu and immune cell function underlying key stages of pregnancy. Recent studies have implicated dysregulated expression of the interleukin-1 (IL-1) family cytokine, IL-33, and its receptor, ST2, in poor pregnancy outcomes in women, including recurrent pregnancy loss, preeclampsia, and preterm labor. How IL-33 supports pregnancy progression in vivo is not well understood. Here, we demonstrate that maternal IL-33 signaling critically regulates uterine tissue remodeling and immune cell function during early pregnancy in mice. IL-33-deficient dams exhibit defects in implantation chamber formation and decidualization, and abnormal vascular remodeling during early pregnancy. These defects coincide with delays in early embryogenesis, increased resorptions, and impaired fetal and placental growth by late pregnancy. At a cellular level, myometrial fibroblasts, and decidual endothelial and stromal cells, are the main IL-33+ cell types in the uterus during decidualization and early placentation, whereas ST2 is expressed by uterine immune populations associated with type 2 immune responses, including ILC2s, Tregs, CD4+ T cells, M2- and cDC2-like myeloid cells, and mast cells. Early pregnancy defects in IL-33-deficient dams are associated with impaired type 2 cytokine responses by uterine lymphocytes and fewer Arginase-1+ macrophages in the uterine microenvironment. Collectively, our data highlight a regulatory network, involving crosstalk between IL-33-producing nonimmune cells and ST2+ immune cells at the maternal-fetal interface, that critically supports pregnancy progression in mice. This work has the potential to advance our understanding of how IL-33 signaling may support optimal pregnancy outcomes in women.


Assuntos
Interleucina-33 , Placenta , Placentação , Útero , Animais , Decídua/irrigação sanguínea , Decídua/citologia , Decídua/crescimento & desenvolvimento , Decídua/imunologia , Feminino , Feto/imunologia , Proteína 1 Semelhante a Receptor de Interleucina-1/metabolismo , Interleucina-33/deficiência , Interleucina-33/imunologia , Linfócitos/imunologia , Linfócitos/metabolismo , Camundongos , Placenta/imunologia , Placenta/metabolismo , Gravidez , Útero/irrigação sanguínea , Útero/crescimento & desenvolvimento , Útero/imunologia , Útero/metabolismo
4.
Biol Reprod ; 110(6): 1175-1190, 2024 Jun 12.
Artigo em Inglês | MEDLINE | ID: mdl-38713674

RESUMO

Uterine muscle contractility is essential for reproductive processes including sperm and embryo transport, and during the uterine cycle to remove menstrual effluent. Even still, uterine contractions have primarily been studied in the context of preterm labor. This is partly due to a lack of methods for studying the uterine muscle contractility in the intact organ. Here, we describe an imaging-based method to evaluate mouse uterine contractility of both the longitudinal and circular muscles in the cycling stages and in early pregnancy. By transforming the image-based data into three-dimensional spatiotemporal contractility maps, we calculate waveform characteristics of muscle contractions, including amplitude, frequency, wavelength, and velocity. We report that the native organ is highly contractile during the progesterone-dominant diestrus stage of the cycle when compared to the estrogen-dominant proestrus and estrus stages. We also observed that during the first phase of uterine embryo movement when clustered embryos move toward the middle of the uterine horn, contractions are dynamic and non-uniform between different segments of the uterine horn. In the second phase of embryo movement, contractions are more uniform and rhythmic throughout the uterine horn. Finally, in Lpar3-/- uteri, which display faster embryo movement, we observe global and regional increases in contractility. Our method provides a means to understand the wave characteristics of uterine smooth muscle in response to modulators and in genetic mutants. Better understanding uterine contractility in the early pregnancy stages is critical for the advancement of artificial reproductive technologies and a possibility of modulating embryo movement during clinical embryo transfers.


Assuntos
Contração Uterina , Feminino , Animais , Contração Uterina/fisiologia , Gravidez , Camundongos , Útero/fisiologia , Ciclo Estral/fisiologia
5.
Mol Hum Reprod ; 30(6)2024 May 30.
Artigo em Inglês | MEDLINE | ID: mdl-38788747

RESUMO

Uterine glands are branched, tubular structures whose secretions are essential for pregnancy success. It is known that pre-implantation glandular expression of leukemia inhibitory factor (LIF) is crucial for embryo implantation; however, the contribution of uterine gland structure to gland secretions, such as LIF, is not known. Here, we use mice deficient in estrogen receptor 1 (ESR1) signaling to uncover the role of ESR1 signaling in gland branching and the role of a branched structure in LIF secretion and embryo implantation. We observed that deletion of ESR1 in neonatal uterine epithelium, stroma, and muscle using the progesterone receptor PgrCre causes a block in uterine gland development at the gland bud stage. Embryonic epithelial deletion of ESR1 using a Müllerian duct Cre line, Pax2Cre, displays gland bud elongation but a failure in gland branching. Reduction of ESR1 in adult uterine epithelium using the lactoferrin-Cre (LtfCre) displays normally branched uterine glands. Unbranched glands from Pax2Cre Esr1flox/flox uteri fail to express glandular pre-implantation Lif, preventing implantation chamber formation and embryo alignment along the uterine mesometrial-antimesometrial axis. In contrast, branched glands from LtfCre Esr1flox/flox uteri display reduced expression of ESR1 and glandular Lif resulting in delayed implantation chamber formation and embryo-uterine axes alignment but mice deliver a normal number of pups. Finally, pre-pubertal unbranched glands in control mice express Lif in the luminal epithelium but fail to express Lif in the glandular epithelium, even in the presence of estrogen. These data strongly suggest that branched glands are necessary for pre-implantation glandular Lif expression for implantation success. Our study is the first to identify a relationship between the branched structure and secretory function of uterine glands and provides a framework for understanding how uterine gland structure-function contributes to pregnancy success.


Assuntos
Implantação do Embrião , Receptor alfa de Estrogênio , Fator Inibidor de Leucemia , Útero , Animais , Feminino , Implantação do Embrião/fisiologia , Útero/metabolismo , Camundongos , Fator Inibidor de Leucemia/metabolismo , Fator Inibidor de Leucemia/genética , Receptor alfa de Estrogênio/metabolismo , Receptor alfa de Estrogênio/genética , Gravidez , Camundongos Knockout , Transdução de Sinais
6.
Hum Reprod ; 39(5): 1023-1041, 2024 May 02.
Artigo em Inglês | MEDLINE | ID: mdl-38511208

RESUMO

STUDY QUESTION: How does ovarian stimulation (OS), which is used to mature multiple oocytes for ART procedures, impact the principal cellular compartments and transcriptome of the human endometrium in the periovulatory and mid-secretory phases? SUMMARY ANSWER: During the mid-secretory window of implantation, OS alters the abundance of endometrial immune cells, whereas during the periovulatory period, OS substantially changes the endometrial transcriptome and impacts both endometrial glandular and immune cells. WHAT IS KNOWN ALREADY: Pregnancies conceived in an OS cycle are at risk of complications reflective of abnormal placentation and placental function. OS can alter endometrial gene expression and immune cell populations. How OS impacts the glandular, stromal, immune, and vascular compartments of the endometrium, in the periovulatory period as compared to the window of implantation, is unknown. STUDY DESIGN, SIZE, DURATION: This prospective cohort study carried out between 2020 and 2022 included 25 subjects undergoing OS and 25 subjects in natural menstrual cycles. Endometrial biopsies were performed in the proliferative, periovulatory, and mid-secretory phases. PARTICIPANTS/MATERIALS, SETTING, METHODS: Blood samples were processed to determine serum estradiol and progesterone levels. Both the endometrial transcriptome and the principal cellular compartments of the endometrium, including glands, stroma, immune, and vasculature, were evaluated by examining endometrial dating, differential gene expression, protein expression, cell populations, and the three-dimensional structure in endometrial tissue. Mann-Whitney U tests, unpaired t-tests or one-way ANOVA and pairwise multiple comparison tests were used to statistically evaluate differences. MAIN RESULTS AND THE ROLE OF CHANCE: In the periovulatory period, OS induced high levels of differential gene expression, glandular-stromal dyssynchrony, and an increase in both glandular epithelial volume and the frequency of endometrial monocytes/macrophages. In the window of implantation during the mid-secretory phase, OS induced changes in endometrial immune cells, with a greater frequency of B cells and a lower frequency of CD4 effector T cells. LARGE SCALE DATA: The data underlying this article have been uploaded to the Genome Expression Omnibus/National Center for Biotechnology Information with accession number GSE220044. LIMITATIONS, REASONS FOR CAUTION: A limited number of subjects were included in this study, although the subjects within each group, natural cycle or OS, were homogenous in their clinical characteristics. The number of subjects utilized was sufficient to identify significant differences; however, with a larger number of subjects and additional power, we may detect additional differences. Another limitation of the study is that proliferative phase biopsies were collected in natural cycles, but not in OS cycles. Given that the OS cycle subjects did not have known endometrial factor infertility, and the comparisons involved subjects who had a similar and robust response to stimulation, the findings are generalizable to women with a normal response to OS. WIDER IMPLICATIONS OF THE FINDINGS: OS substantially altered the periovulatory phase endometrium, with fewer transcriptomic and cell type-specific changes in the mid-secretory phase. Our findings show that after OS, the endometrial microenvironment in the window of implantation possesses many more similarities to that of a natural cycle than does the periovulatory endometrium. Further investigation of the immune compartment and the functional significance of this cellular compartment under OS conditions is warranted. STUDY FUNDING/COMPETING INTERESTS: Research reported in this publication was supported by the National Institute of Allergy and Infectious Diseases (R01AI148695 to A.M.B. and N.C.D.), Eunice Kennedy Shriver National Institute of Child Health and Human Development (R01HD109152 to R.A.), and the March of Dimes (5-FY20-209 to R.A.). The content is solely the responsibility of the authors and does not necessarily represent the official views of the National Institutes of Health or March of Dimes. All authors declare no conflict of interest.


Assuntos
Endométrio , Indução da Ovulação , Transcriptoma , Humanos , Feminino , Endométrio/metabolismo , Adulto , Microambiente Celular , Estudos Prospectivos , Estradiol/sangue , Implantação do Embrião/fisiologia , Progesterona/sangue , Progesterona/metabolismo , Gravidez , Ciclo Menstrual
7.
FASEB J ; 37(7): e22983, 2023 07.
Artigo em Inglês | MEDLINE | ID: mdl-37249327

RESUMO

In mammals, the endometrium undergoes dynamic changes in response to estrogen and progesterone to prepare for blastocyst implantation. Two distinct types of endometrial epithelial cells, the luminal (LE) and glandular (GE) epithelial cells play different functional roles during this physiological process. Previously, we have reported that Notch signaling plays multiple roles in embryo implantation, decidualization, and postpartum repair. Here, using the uterine epithelial-specific Ltf-iCre, we showed that Notch1 signaling over-activation in the endometrial epithelium caused dysfunction of the epithelium during the estrous cycle, resulting in hyper-proliferation. During pregnancy, it further led to dysregulation of estrogen and progesterone signaling, resulting in infertility in these animals. Using 3D organoids, we showed that over-activation of Notch1 signaling increased the proliferative potential of both LE and GE cells and reduced the difference in transcription profiles between them, suggesting disrupted differentiation of the uterine epithelium. In addition, we demonstrated that both canonical and non-canonical Notch signaling contributed to the hyper-proliferation of GE cells, but only the non-canonical pathway was involved with estrogen sensitivity in the GE cells. These findings provided insights into the effects of Notch1 signaling on the proliferation, differentiation, and function of the uterine epithelium. This study demonstrated the important roles of Notch1 signaling in regulating hormone response and differentiation of endometrial epithelial cells and provides an opportunity for future studies in estrogen-dependent diseases, such as endometriosis.


Assuntos
Progesterona , Útero , Animais , Feminino , Camundongos , Gravidez , Proliferação de Células , Implantação do Embrião/fisiologia , Endométrio/metabolismo , Epitélio/metabolismo , Estrogênios/farmacologia , Estrogênios/metabolismo , Progesterona/farmacologia , Progesterona/metabolismo , Útero/metabolismo
8.
Development ; 147(24)2020 12 21.
Artigo em Inglês | MEDLINE | ID: mdl-33158924

RESUMO

How a mammalian embryo determines and arrives at its attachment site has been studied for decades, but our understanding of this process is far from complete. Using confocal imaging and image analysis, we evaluate embryo location along the longitudinal oviductal-cervical axis of murine uteri. Our analysis reveals three distinct pre-implantation phases: embryo entry, unidirectional movement of embryo clusters and bidirectional scattering and spacing of embryos. We show that unidirectional clustered movement is facilitated by a mechanical stimulus of the embryo and is regulated by adrenergic uterine smooth muscle contractions. Embryo scattering, on the other hand, depends on embryo-uterine communication reliant on the LPAR3 signaling pathway and is independent of adrenergic muscle contractions. Finally, we demonstrate that uterine implantation sites in mice are neither random nor predetermined but are guided by the number of embryos entering the uterine lumen. These studies have implications for understanding how embryo-uterine communication is key to determining an optimal implantation site necessary for the success of a pregnancy.


Assuntos
Implantação do Embrião/genética , Contração Muscular/genética , Receptores de Ácidos Lisofosfatídicos/genética , Contração Uterina/genética , Animais , Desenvolvimento Embrionário/genética , Tubas Uterinas/crescimento & desenvolvimento , Feminino , Humanos , Camundongos , Movimento/fisiologia , Músculo Liso/crescimento & desenvolvimento , Gravidez , Transdução de Sinais/genética , Útero/crescimento & desenvolvimento
9.
Mol Hum Reprod ; 29(2)2023 01 31.
Artigo em Inglês | MEDLINE | ID: mdl-36579867

RESUMO

Pre-implantation embryo movement is crucial to pregnancy success, but the role of ovarian hormones in modulating embryo movement is not understood. We ascertain the effects of altered hormonal environment on embryo location using two delayed implantation mouse models: natural lactational diapause (ND); and artificially induced diapause (AD), a laboratory version of ND generated by ovary removal and provision of supplemental progesterone (P4). Previously, we showed that embryos in a natural pregnancy (NP) first display unidirectional clustered movement, followed by bidirectional scattering and spacing movement. In the ND model, we discovered that embryos are present as clusters near the oviductal-uterine junction for ∼24 h longer than NP, followed by locations consistent with a unidirectional scattering and spacing movement. Intriguingly, the AD model resembles embryo location in NP and not ND. When measuring serum hormone levels, unlike the popular paradigm of reduced estrogen (E2) levels in diapause, we observed that E2 levels are comparable across NP, ND and AD. P4 levels are reduced in ND and highly increased in AD when compared to NP. Further, exogenous administration of E2 or P4 modifies embryo location during the unidirectional phase, while E2 treatment also affects embryo location in the bidirectional phase. Taken together, our data suggest that embryo movement can be modulated by both P4 and E2. Understanding natural hormonal adaptation in diapause provides an opportunity to determine key players that regulate embryo location, thus impacting implantation success. This knowledge can be leveraged to understand pregnancy survival and implantation success in hormonally altered conditions in the clinic.


Assuntos
Implantação do Embrião , Estradiol , Gravidez , Feminino , Camundongos , Animais , Estradiol/farmacologia , Progesterona/farmacologia , Desenvolvimento Embrionário , Útero
10.
FASEB J ; 35(2): e21209, 2021 02.
Artigo em Inglês | MEDLINE | ID: mdl-33222288

RESUMO

Though endometriosis and infertility are clearly associated, the pathophysiological mechanism remains unclear. Previous work has linked endometrial ARID1A loss to endometriosis-related endometrial non-receptivity. Here, we show in mice that ARID1A binds and regulates transcription of the Foxa2 gene required for endometrial gland function. Uterine-specific deletion of Arid1a compromises gland development and diminishes Foxa2 and Lif expression. Deletion of Arid1a with Ltf-iCre in the adult mouse endometrial epithelium preserves the gland development while still compromising the gland function. Mice lacking endometrial epithelial Arid1a are severely sub-fertile due to defects in implantation, decidualization, and endometrial receptivity from disruption of the LIF-STAT3-EGR1 pathway. FOXA2 is also reduced in the endometrium of women with endometriosis in correlation with diminished ARID1A, and both ARID1A and FOXA2 are reduced in nonhuman primates induced with endometriosis. Our findings describe a role for ARID1A in the endometrial epithelium supporting early pregnancy establishment through the maintenance of gland function.


Assuntos
Proteínas de Ligação a DNA/metabolismo , Implantação do Embrião , Endométrio/metabolismo , Fatores de Transcrição/metabolismo , Adulto , Animais , Proteínas de Ligação a DNA/genética , Feminino , Fator 3-beta Nuclear de Hepatócito/genética , Fator 3-beta Nuclear de Hepatócito/metabolismo , Humanos , Fator Inibidor de Leucemia/genética , Fator Inibidor de Leucemia/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Gravidez , Fatores de Transcrição/genética
11.
Development ; 143(24): 4749-4754, 2016 12 15.
Artigo em Inglês | MEDLINE | ID: mdl-27836961

RESUMO

Although much is known about the embryo during implantation, the architecture of the uterine environment in which the early embryo develops is not well understood. We employed confocal imaging in combination with 3D analysis to identify and quantify dynamic changes to the luminal structure of murine uterus in preparation for implantation. When applied to mouse mutants with known implantation defects, this method detected striking peri-implantation abnormalities in uterine morphology that cannot be visualized by histology. We revealed 3D organization of uterine glands and found that they undergo a stereotypical reorientation concurrent with implantation. Furthermore, we extended this technique to generate a 3D rendering of the cycling human endometrium. Analyzing the uterine and embryo structure in 3D for different genetic mutants and pathological conditions will help uncover novel molecular pathways and global structural changes that contribute to successful implantation of an embryo.


Assuntos
Blastocisto/ultraestrutura , Implantação do Embrião/fisiologia , Embrião de Mamíferos/ultraestrutura , Endométrio/ultraestrutura , Útero/ultraestrutura , Animais , Embrião de Mamíferos/diagnóstico por imagem , Endométrio/diagnóstico por imagem , Endométrio/fisiologia , Feminino , Humanos , Imageamento Tridimensional , Camundongos , Camundongos Endogâmicos C57BL , Útero/diagnóstico por imagem , Útero/fisiologia , Proteína Wnt-5a/genética
12.
J Cell Sci ; 129(6): 1271-82, 2016 Mar 15.
Artigo em Inglês | MEDLINE | ID: mdl-26826184

RESUMO

Meiotic progression requires exquisitely coordinated translation of maternal messenger (m)RNA that has accumulated during oocyte growth. A major regulator of this program is the cytoplasmic polyadenylation element binding protein 1 (CPEB1). However, the temporal pattern of translation at different meiotic stages indicates the function of additional RNA binding proteins (RBPs). Here, we report that deleted in azoospermia-like (DAZL) cooperates with CPEB1 to regulate maternal mRNA translation. Using a strategy that monitors ribosome loading onto endogenous mRNAs and a prototypic translation target, we show that ribosome loading is induced in a DAZL- and CPEB1-dependent manner, as the oocyte reenters meiosis. Depletion of the two RBPs from oocytes and mutagenesis of the 3' untranslated regions (UTRs) demonstrate that both RBPs interact with the Tex19.1 3' UTR and cooperate in translation activation of this mRNA. We observed a synergism between DAZL and cytoplasmic polyadenylation elements (CPEs) in the translation pattern of maternal mRNAs when using a genome-wide analysis. Mechanistically, the number of DAZL proteins loaded onto the mRNA and the characteristics of the CPE might define the degree of cooperation between the two RBPs in activating translation and meiotic progression.


Assuntos
Oócitos/citologia , Oócitos/metabolismo , Oogênese , Biossíntese de Proteínas , RNA Mensageiro/metabolismo , Proteínas de Ligação a RNA/metabolismo , Fatores de Transcrição/metabolismo , Fatores de Poliadenilação e Clivagem de mRNA/metabolismo , Animais , Feminino , Regulação da Expressão Gênica no Desenvolvimento , Masculino , Meiose , Camundongos Endogâmicos C57BL , RNA Mensageiro/genética , Proteínas de Ligação a RNA/genética , Fatores de Transcrição/genética , Fatores de Poliadenilação e Clivagem de mRNA/genética
13.
J Cell Sci ; 129(13): 2493-9, 2016 07 01.
Artigo em Inglês | MEDLINE | ID: mdl-27199373

RESUMO

Mouse ovarian germ cells enter meiosis in a wave that propagates from anterior to posterior, but little is known about contribution of germ cells to initiation or propagation of meiosis. In a Ror2 mutant with diminished germ cell number and migration, we find that overall timing of meiotic initiation is delayed at the population level. We use chemotherapeutic depletion to exclude a profoundly reduced number of germ cells as a cause for meiotic delay. We rule out sex reversal or failure to specify somatic support cells as contributors to the meiotic phenotype. Instead, we find that anomalies in the distribution of germ cells as well as gonad shape in mutants contribute to aberrant initiation of meiosis. Our analysis supports a model of meiotic initiation via diffusible signal(s), excludes a role for germ cells in commencing the meiotic wave and furnishes the first phenotypic demonstration of the wave of meiotic entry. Finally, our studies underscore the importance of considering germ cell migration defects while studying meiosis to discern secondary effects resulting from positioning versus primary meiotic entry phenotypes.


Assuntos
Células Germinativas/metabolismo , Gônadas/patologia , Meiose/genética , Receptores Órfãos Semelhantes a Receptor Tirosina Quinase/genética , Animais , Contagem de Células , Movimento Celular/genética , Forma Celular/genética , Feminino , Células Germinativas/crescimento & desenvolvimento , Células Germinativas/patologia , Gônadas/crescimento & desenvolvimento , Camundongos , Mutação , Ovário/crescimento & desenvolvimento , Ovário/metabolismo , Ovário/patologia , Transdução de Sinais/genética
14.
Dev Biol ; 403(1): 69-79, 2015 Jul 01.
Artigo em Inglês | MEDLINE | ID: mdl-25889274

RESUMO

Quantitative analysis of tissues and organs can reveal large-scale patterning as well as the impact of perturbations and aging on biological architecture. Here we develop tools for imaging of single cells in intact organs and computational approaches to assess spatial relationships in 3D. In the mouse ovary, we use nuclear volume of the oocyte to read out quiescence or growth of oocyte-somatic cell units known as follicles. This in-ovary quantification of non-growing follicle dynamics from neonate to adult fits a mathematical function, which corroborates the model of fixed oocyte reserve. Mapping approaches show that radial organization of folliculogenesis established in the newborn ovary is preserved through adulthood. By contrast, inter-follicle clustering increases during aging with different dynamics depending on size. These broadly applicable tools can reveal high dimensional phenotypes and age-related architectural changes in other organs. In the adult mouse pancreas, we find stochastic radial organization of the islets of Langerhans but evidence for localized interactions among the smallest islets.


Assuntos
Imageamento Tridimensional/métodos , Ilhotas Pancreáticas/fisiologia , Oócitos/fisiologia , Folículo Ovariano/fisiologia , Análise de Célula Única/métodos , Envelhecimento , Algoritmos , Animais , Feminino , Ilhotas Pancreáticas/ultraestrutura , Camundongos , Camundongos Endogâmicos C57BL , Oócitos/ultraestrutura , Folículo Ovariano/ultraestrutura
15.
PLoS Genet ; 9(2): e1003206, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-23437001

RESUMO

Danforth's short tail mutant (Sd) mouse, first described in 1930, is a classic spontaneous mutant exhibiting defects of the axial skeleton, hindgut, and urogenital system. We used meiotic mapping in 1,497 segregants to localize the mutation to a 42.8-kb intergenic segment on chromosome 2. Resequencing of this region identified an 8.5-kb early retrotransposon (ETn) insertion within the highly conserved regulatory sequences upstream of Pancreas Specific Transcription Factor, 1a (Ptf1a). This mutation resulted in up to tenfold increased expression of Ptf1a as compared to wild-type embryos at E9.5 but no detectable changes in the expression levels of other neighboring genes. At E9.5, Sd mutants exhibit ectopic Ptf1a expression in embryonic progenitors of every organ that will manifest a developmental defect: the notochord, the hindgut, and the mesonephric ducts. Moreover, at E 8.5, Sd mutant mice exhibit ectopic Ptf1a expression in the lateral plate mesoderm, tail bud mesenchyme, and in the notochord, preceding the onset of visible defects such as notochord degeneration. The Sd heterozygote phenotype was not ameliorated by Ptf1a haploinsufficiency, further suggesting that the developmental defects result from ectopic expression of Ptf1a. These data identify disruption of the spatio-temporal pattern of Ptf1a expression as the unifying mechanism underlying the multiple congenital defects in Danforth's short tail mouse. This striking example of an enhancer mutation resulting in profound developmental defects suggests that disruption of conserved regulatory elements may also contribute to human malformation syndromes.


Assuntos
Desenvolvimento Embrionário/genética , Mutagênese Insercional/genética , Retroelementos/genética , Fatores de Transcrição , Animais , Proteínas de Ligação a DNA/genética , Proteínas de Ligação a DNA/metabolismo , Regulação da Expressão Gênica no Desenvolvimento/genética , Humanos , Mesoderma/anormalidades , Mesoderma/crescimento & desenvolvimento , Camundongos , Pâncreas/anormalidades , Pâncreas/crescimento & desenvolvimento , Medula Espinal/anormalidades , Medula Espinal/crescimento & desenvolvimento , Cauda/anatomia & histologia , Fatores de Transcrição/genética , Fatores de Transcrição/metabolismo
16.
PLoS Genet ; 8(8): e1002866, 2012.
Artigo em Inglês | MEDLINE | ID: mdl-22876201

RESUMO

Normal development of the respiratory system is essential for survival and is regulated by multiple genes and signaling pathways. Both Tbx4 and Tbx5 are expressed throughout the mesenchyme of the developing lung and trachea; and, although multiple genes are known to be required in the epithelium, only Fgfs have been well studied in the mesenchyme. In this study, we investigated the roles of Tbx4 and Tbx5 in lung and trachea development using conditional mutant alleles and two different Cre recombinase transgenic lines. Loss of Tbx5 leads to a unilateral loss of lung bud specification and absence of tracheal specification in organ culture. Mutants deficient in Tbx4 and Tbx5 show severely reduced lung branching at mid-gestation. Concordant with this defect, the expression of mesenchymal markers Wnt2 and Fgf10, as well as Fgf10 target genes Bmp4 and Spry2, in the epithelium is downregulated. Lung branching undergoes arrest ex vivo when Tbx4 and Tbx5 are both completely lacking. Lung-specific Tbx4 heterozygous;Tbx5 conditional null mice die soon after birth due to respiratory distress. These pups have small lungs and show severe disruptions in tracheal/bronchial cartilage rings. Sox9, a master regulator of cartilage formation, is expressed in the trachea; but mesenchymal cells fail to condense and consequently do not develop cartilage normally at birth. Tbx4;Tbx5 double heterozygous mutants show decreased lung branching and fewer tracheal cartilage rings, suggesting a genetic interaction. Finally, we show that Tbx4 and Tbx5 interact with Fgf10 during the process of lung growth and branching but not during tracheal/bronchial cartilage development.


Assuntos
Pulmão/metabolismo , Transdução de Sinais/genética , Proteínas com Domínio T/genética , Traqueia/metabolismo , Proteínas Adaptadoras de Transdução de Sinal , Animais , Biomarcadores/metabolismo , Proteína Morfogenética Óssea 4/genética , Proteína Morfogenética Óssea 4/metabolismo , Cartilagem/anatomia & histologia , Cartilagem/embriologia , Cartilagem/metabolismo , Embrião de Mamíferos , Feminino , Fator 10 de Crescimento de Fibroblastos/genética , Fator 10 de Crescimento de Fibroblastos/metabolismo , Regulação da Expressão Gênica no Desenvolvimento , Peptídeos e Proteínas de Sinalização Intracelular , Pulmão/anatomia & histologia , Pulmão/embriologia , Proteínas de Membrana/genética , Proteínas de Membrana/metabolismo , Mesoderma/anatomia & histologia , Mesoderma/embriologia , Mesoderma/metabolismo , Camundongos , Morfogênese/genética , Técnicas de Cultura de Órgãos , Proteínas Serina-Treonina Quinases , Proteínas com Domínio T/deficiência , Traqueia/anatomia & histologia , Traqueia/embriologia , Proteína Wnt2
17.
Dev Dyn ; 243(8): 1037-45, 2014 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-24753105

RESUMO

BACKGROUND: The noncanonical Wnt receptor and tyrosine kinase Ror2 has been associated with recessive Robinow syndrome (RRS) and dominant brachydactyly type B1. The phenotypes of mouse mutants implicate Ror2 in the development of the heart, lungs, bone, and craniofacial structures, which are affected in RRS. Following a recently identified role of Ror2 in the migration of mouse primordial germ cells, we extensively characterized its expression throughout the fetal internal reproductive system and the postnatal ductal system. RESULTS: We show that Ror2 gene products are present in the germ cells and somatic cells of the testis and the ovary of both the mouse and human fetus. In reproductive tract structures, we find that Ror2 is expressed in the mesonephros, developing Wolffian and Müllerian ducts, and later in their derivatives, the epididymal epithelium and uterine epithelium. CONCLUSIONS: This study sets the stage to explore function for this tyrosine kinase receptor in novel regions of expression in the developing reproductive system in both mouse and human.


Assuntos
Proteínas Proto-Oncogênicas/metabolismo , Receptores Órfãos Semelhantes a Receptor Tirosina Quinase/metabolismo , Proteínas Wnt/metabolismo , Animais , Feminino , Células Germinativas/citologia , Células da Granulosa , Humanos , Masculino , Camundongos , Ductos Paramesonéfricos/citologia , Ovário/citologia , Proteínas Proto-Oncogênicas/genética , Receptores Órfãos Semelhantes a Receptor Tirosina Quinase/genética , Células de Sertoli/citologia , Testículo/citologia , Proteínas Wnt/genética , Proteína Wnt-5a , Ductos Mesonéfricos/citologia
18.
Dev Dyn ; 243(5): 629-39, 2014 May.
Artigo em Inglês | MEDLINE | ID: mdl-24347445

RESUMO

BACKGROUND: The short stature homeodomain transcription factors SHOX and SHOX2 play key roles in limb formation. To gain more insight into genes regulated by Shox2 during limb development, we analyzed expression profiles of WT and Shox2-/- mouse embryonic limbs and identified the T-Box transcription factor Tbx4 as a potential downstream target. Tbx4 is known to exert essential functions in skeletal and muscular hindlimb development. In humans, haploinsufficiency of TBX4 causes small patella syndrome, a skeletal dysplasia characterized by anomalies of the knee, pelvis, and foot. RESULTS: Here, we demonstrate an inhibitory regulatory effect of Shox2 on Tbx4 specifically in the forelimbs. We also show that Tbx4 activates Shox2 expression in fore- and hindlimbs, suggesting Shox2 as a feedback modulator of Tbx4. Using EMSA studies, we find that Tbx4/TBX4 is able to bind to distinct T-box binding sites within the mouse and human Shox2/SHOX2 promoter. CONCLUSIONS: Our data identifies Tbx4 as a novel transcriptional activator of Shox2 during murine fore- and hindlimb development. Tbx4 is also regulated by Shox2 specifically in the forelimb bud possibly via a feedback mechanism. These data extend our understanding of the role and regulation of Tbx4 and Shox2 in limb development and limb associated diseases.


Assuntos
Embrião de Mamíferos/embriologia , Membro Posterior/embriologia , Proteínas de Homeodomínio/metabolismo , Organogênese/fisiologia , Proteínas com Domínio T/metabolismo , Animais , Proteínas de Homeodomínio/genética , Humanos , Camundongos , Camundongos Knockout , Proteínas com Domínio T/genética
19.
Blood ; 120(13): 2562-72, 2012 Sep 27.
Artigo em Inglês | MEDLINE | ID: mdl-22855605

RESUMO

The allantois is the embryonic precursor of the umbilical cord in mammals and is one of several embryonic regions, including the yolk sac and dorsal aorta, that undergoes vasculogenesis, the de novo formation of blood vessels. Despite its importance in establishing the chorioallantoic placenta and umbilical circulation, the allantois frequently is overlooked in embryologic studies. Nonetheless, recent studies demonstrate that vasculogenesis, vascular remodeling, and angiogenesis are essential allantois functions in the establishment of the chorioallantoic placenta. Here, we review blood vessel formation in the murine allantois, highlighting the expression of genes and involvement of pathways common to vasculogenesis or angiogenesis in other parts of the embryo. We discuss experimental techniques available for manipulation of the allantois that are unavailable for yolk sac or dorsal aorta, and review how this system has been used as a model system to discover new genes and mechanisms involved in vessel formation. Finally, we discuss the potential of the allantois as a model system to provide insights into disease and therapeutics.


Assuntos
Alantoide/irrigação sanguínea , Alantoide/embriologia , Vasos Sanguíneos/crescimento & desenvolvimento , Modelos Animais , Neovascularização Fisiológica , Alantoide/metabolismo , Animais , Camundongos
20.
bioRxiv ; 2024 May 21.
Artigo em Inglês | MEDLINE | ID: mdl-38766130

RESUMO

Endometrial stromal cell decidualization is required for pregnancy success. Although this process is integral to fertility, many of the intricate molecular mechanisms contributing to decidualization remain undefined. One pathway that has been implicated in endometrial stromal cell decidualization in humans in vitro is the Hippo signaling pathway. Two previously conducted studies showed that the effectors of the Hippo signaling pathway, YAP1 and WWTR1, were required for decidualization of primary stromal cells in culture. To investigate the in vivo role of YAP1 and WWTR1 in decidualization and pregnancy initiation, we generated a Progesterone Cre mediated partial double knockout (pdKO) of Yap1 and Wwtr1. Female pdKOs exhibited subfertility, a compromised decidualization response, partial interruption in embryo transport, blunted endometrial receptivity, delayed implantation and subsequent embryonic development, and a unique transcriptional profile. Bulk mRNA sequencing revealed aberrant maternal remodeling evidenced by significant alterations in extracellular matrix proteins at 7.5 days post-coitus in pdKO dams and enrichment for terms associated with fertility-compromising diseases like pre-eclampsia and endometriosis. Our results indicate a required role for YAP1 and WWTR1 for successful mammalian uterine function and pregnancy success.

SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA