Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 258
Filtrar
Mais filtros

Base de dados
Tipo de documento
Intervalo de ano de publicação
1.
Cell ; 186(15): 3138-3142, 2023 07 20.
Artigo em Inglês | MEDLINE | ID: mdl-37478817

RESUMO

Despite tremendous diversity, Asian Americans in STEM are grouped and viewed as a homogeneous monolith, facing stereotypes and disparities. We propose solutions that include disaggregating the Asian American grouping and recognizing the diverse individual ethnic subgroups that comprise Americans of Asian ancestry to implement change within the STEM field.


Assuntos
Asiático , Humanos , Estados Unidos
2.
Differentiation ; 138: 100791, 2024.
Artigo em Inglês | MEDLINE | ID: mdl-38941819

RESUMO

A Wt1 conditional deletion, nuclear red fluorescent protein (RFP) reporter allele was generated in the mouse by gene targeting in embryonic stem cells. Upon Cre-mediated recombination, a deletion allele is generated that expresses RFP in a Wt1-specific pattern. RFP expression was detected in embryonic and adult tissues known to express Wt1, including the kidney, mesonephros, and testis. In addition, RFP expression and WT1 co-localization was detected in the adult uterine stroma and myometrium, suggesting a role in uterine function. Crosses with Wnt7a-Cre transgenic mice that express Cre in the Müllerian duct epithelium activate Wt1-directed RFP expression in the epithelium of the oviduct but not the stroma and myometrium of the uterus. This new mouse strain should be a useful resource for studies of Wt1 function and marking Wt1-expressing cells.


Assuntos
Alelos , Proteínas Luminescentes , Camundongos Transgênicos , Proteína Vermelha Fluorescente , Proteínas WT1 , Animais , Camundongos , Proteínas WT1/genética , Proteínas WT1/metabolismo , Proteínas Luminescentes/genética , Proteínas Luminescentes/metabolismo , Feminino , Genes Reporter , Masculino , Deleção de Genes
3.
Biol Reprod ; 110(6): 1077-1085, 2024 Jun 12.
Artigo em Inglês | MEDLINE | ID: mdl-38641547

RESUMO

Volumetric data provide unprecedented structural insight to the reproductive tract and add vital anatomical context to the relationships between organs. The morphology of the female reproductive tract in non-avian reptiles varies between species, corresponding to a broad range of reproductive modes and providing valuable insight to comparative investigations of reproductive anatomy. However, reproductive studies in reptilian models, such as the brown anole studied here, have historically relied on histological methods to understand the anatomy. While these methods are highly effective for characterizing the cell types present in each organ, histological methods lose the 3D relationships between images and leave the architecture of the organ system poorly understood. We present the first comprehensive volumetric analyses of the female brown anole reproductive tract using two non-invasive, non-destructive imaging modalities: micro-computed tomography (microCT) and optical coherence tomography (OCT). Both are specialized imaging technologies that facilitate high-throughput imaging and preserve three-dimensional information. This study represents the first time that microCT has been used to study all reproductive organs in this species and the very first time that OCT has been applied to this species. We show how the non-destructive volumetric imaging provided by each modality reveals anatomical context including orientation and relationships between reproductive organs of the anole lizard. In addition to broad patterns of morphology, both imaging modalities provide the high resolution necessary to capture details and key anatomical features of each organ. We demonstrate that classic histological features can be appreciated within whole-organ architecture in volumetric imaging using microCT and OCT, providing the complementary information necessary to understand the relationships between tissues and organs in the reproductive system. This side-by-side imaging analysis using microCT and OCT allows us to evaluate the specific advantages and limitations of these two methods for the female reptile reproductive system.


Assuntos
Genitália Feminina , Lagartos , Tomografia de Coerência Óptica , Microtomografia por Raio-X , Animais , Feminino , Microtomografia por Raio-X/métodos , Microtomografia por Raio-X/veterinária , Tomografia de Coerência Óptica/métodos , Tomografia de Coerência Óptica/veterinária , Lagartos/anatomia & histologia , Genitália Feminina/diagnóstico por imagem , Genitália Feminina/anatomia & histologia , Imageamento Tridimensional/métodos , Imageamento Tridimensional/veterinária
4.
Differentiation ; 2023 Nov 07.
Artigo em Inglês | MEDLINE | ID: mdl-37957094

RESUMO

FGF5 functions as a negative regulator of the hair cycle in mammals. It is expressed in the outer root sheath of hair follicles during the late anagen phase of the hair cycle. It functions as a signaling molecule, mediating the transition of the anagen growth phase to catagen regression phase of the hair cycle. Spontaneous and engineered FGF5 mutations in mammalian animal models result in long hair phenotypes. In humans, inherited FGF5 mutations result in trichomegaly (long eyelashes). Knockdown of fgf5 in zebrafish embryos results in inner ear alterations. Alterations in FGF5 expression are also associated with various human pathologies.

5.
Differentiation ; 130: 28-31, 2023.
Artigo em Inglês | MEDLINE | ID: mdl-36543010

RESUMO

The International Society of Differentiation was born from the First International Conference on Cell Differentiation conceived by D.V. and held in Nice, France in 1971. The conference also resulted in the creation of the journal of the Society named Differentiation. The Society advocates for the field of differentiation through the journal Differentiation, organizing and supporting international scientific conferences, honoring scientific achievements, and supporting trainees.


Assuntos
Diferenciação Celular , Sociedades Científicas , Sociedades Científicas/história
6.
BMC Biol ; 21(1): 101, 2023 05 04.
Artigo em Inglês | MEDLINE | ID: mdl-37143038

RESUMO

BACKGROUND: Through the evolution of novel wing structures, bats (Order Chiroptera) became the only mammalian group to achieve powered flight. This achievement preceded the massive adaptive radiation of bats into diverse ecological niches. We investigate some of the developmental processes that underlie the origin and subsequent diversification of one of the novel membranes of the bat wing: the plagiopatagium, which connects the fore- and hind limb in all bat species. RESULTS: Our results suggest that the plagiopatagium initially arises through novel outgrowths from the body flank that subsequently merge with the limbs to generate the wing airfoil. Our findings further suggest that this merging process, which is highly conserved across bats, occurs through modulation of the programs controlling the development of the periderm of the epidermal epithelium. Finally, our results suggest that the shape of the plagiopatagium begins to diversify in bats only after this merging has occurred. CONCLUSIONS: This study demonstrates how focusing on the evolution of cellular processes can inform an understanding of the developmental factors shaping the evolution of novel, highly adaptive structures.


Assuntos
Quirópteros , Animais , Voo Animal , Mamíferos , Desenvolvimento Embrionário , Asas de Animais
7.
PLoS Genet ; 16(6): e1008808, 2020 06.
Artigo em Inglês | MEDLINE | ID: mdl-32497036

RESUMO

Metastasis is responsible for 90% of human cancer mortality, yet it remains a challenge to model human cancer metastasis in vivo. Here we describe mouse models of high-grade serous ovarian cancer, also known as high-grade serous carcinoma (HGSC), the most common and deadliest human ovarian cancer type. Mice genetically engineered to harbor Dicer1 and Pten inactivation and mutant p53 robustly replicate the peritoneal metastases of human HGSC with complete penetrance. Arising from the fallopian tube, tumors spread to the ovary and metastasize throughout the pelvic and peritoneal cavities, invariably inducing hemorrhagic ascites. Widespread and abundant peritoneal metastases ultimately cause mouse deaths (100%). Besides the phenotypic and histopathological similarities, mouse HGSCs also display marked chromosomal instability, impaired DNA repair, and chemosensitivity. Faithfully recapitulating the clinical metastases as well as molecular and genomic features of human HGSC, this murine model will be valuable for elucidating the mechanisms underlying the development and progression of metastatic ovarian cancer and also for evaluating potential therapies.


Assuntos
Antineoplásicos/farmacologia , Cistadenocarcinoma Seroso/genética , Neoplasias Ovarianas/patologia , Neoplasias Peritoneais/genética , Animais , Antineoplásicos/uso terapêutico , Linhagem Celular Tumoral , Instabilidade Cromossômica , Cistadenocarcinoma Seroso/tratamento farmacológico , Cistadenocarcinoma Seroso/secundário , RNA Helicases DEAD-box/genética , Reparo do DNA , Modelos Animais de Doenças , Resistencia a Medicamentos Antineoplásicos/genética , Ensaios de Seleção de Medicamentos Antitumorais/métodos , Estudos de Viabilidade , Feminino , Humanos , Camundongos , Camundongos Knockout , Mutação , Gradação de Tumores , Metástase Neoplásica/genética , Neoplasias Ovarianas/tratamento farmacológico , Neoplasias Ovarianas/genética , PTEN Fosfo-Hidrolase/genética , Neoplasias Peritoneais/tratamento farmacológico , Neoplasias Peritoneais/secundário , Cultura Primária de Células , Ribonuclease III/genética , Proteína Supressora de Tumor p53/genética
8.
Differentiation ; 127: 1-11, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-36041259

RESUMO

The cervical and anterior thoracic regions of mammals generally exhibit similar vertebral numbers and identities along the anterior-posterior axis. The position of the forelimbs along the axial skeleton is also generally conserved. In contrast, the number of lumbar and sacral vertebrae and pelvic position exhibit more variation, correlating with posture and locomotion. The molecular mechanisms that lead to these conserved and variable axial skeletal patterns between species are not fully understood. Here we use a human HOXB1-9 transgene to complement a HoxB1-9 deficiency in the mouse. In TgHOXB1-9 mice, human HOXB1, B2, B3, and B4 (HOXB1-4) genes were expressed in mouse embryos in patterns similar to mouse Hoxb1-4 genes. Human transgene expression rescued the cervical and anterior thoracic vertebral patterning defects of HoxB1-9Δ/Δ mice. In addition, the posterior shift in forelimb position of HoxB1-9Δ/Δ mice was rescued by the transgene. Interestingly, the position of the lumbar-sacral transition in both TgHOXB1-9; HoxB1-9Δ/Δ and TgHOXB1-9; HoxB1-9+/+ mice was altered from six lumbar and four sacral vertebrae found in wild-type controls to five lumbar and five sacral vertebrae. The change in the position of the lumbar-sacral transition consequently altered the position of the pelvis. In contrast to the conserved expression of human HOXB1-4 genes in TgHOXB1-9 mouse embryos, the anterior border of human HOXB9 expression in the neural tube and paraxial mesoderm was shifted posteriorly by 2-3 somites compared to the anterior boundary of endogenous Hoxb9 expression. These findings suggest that conservation and variation in Hoxb/HOXB expression contributes to conserved and species-specific vertebral pattern and limb position.


Assuntos
Genes Homeobox , Somitos , Animais , Osso e Ossos , Regulação da Expressão Gênica no Desenvolvimento/genética , Proteínas de Homeodomínio/genética , Humanos , Mamíferos/genética , Mamíferos/metabolismo , Camundongos , Camundongos Transgênicos , Coluna Vertebral/metabolismo , Fatores de Transcrição/genética
9.
Proc Natl Acad Sci U S A ; 115(33): 8382-8387, 2018 08 14.
Artigo em Inglês | MEDLINE | ID: mdl-30061417

RESUMO

In mammals, the developing reproductive tract primordium of male and female fetuses consists of the Wolffian duct and the Müllerian duct (MD), two epithelial tube pairs surrounded by mesenchyme. During male development, mesenchyme-epithelia interactions mediate MD regression to prevent its development into a uterus, oviduct, and upper vagina. It is well established that transforming growth factor-ß family member anti-Müllerian hormone (AMH) secreted from the fetal testis and its type 1 and 2 receptors expressed in MD mesenchyme regulate MD regression. However, little is known about the molecular network regulating downstream actions of AMH signaling. To identify potential AMH-induced genes and regulatory networks controlling MD regression in a global nonbiased manner, we examined transcriptome differences in MD mesenchyme between males (AMH signaling on) and females (AMH signaling off) by RNA-seq analysis of purified fetal MD mesenchymal cells. This analysis found 82 genes up-regulated in males during MD regression and identified Osterix (Osx)/Sp7, a key transcriptional regulator of osteoblast differentiation and bone formation, as a downstream effector of AMH signaling during MD regression. Osx/OSX was expressed in a male-specific pattern in MD mesenchyme during MD regression. OSX expression was lost in mutant males without AMH signaling. In addition, transgenic mice ectopically expressing human AMH in females induced a male pattern of Osx expression. Together, these results indicate that AMH signaling is necessary and sufficient for Osx expression in the MD mesenchyme. In addition, MD regression was delayed in Osx-null males, identifying Osx as a factor that regulates MD regression.


Assuntos
Hormônio Antimülleriano/fisiologia , Ductos Paramesonéfricos/fisiologia , Transdução de Sinais/fisiologia , Fator de Transcrição Sp7/fisiologia , Animais , Perfilação da Expressão Gênica , Masculino , Camundongos , Camundongos Endogâmicos C57BL , beta Catenina/fisiologia
10.
Differentiation ; 112: 1-6, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-31830612

RESUMO

Defective anorectal and urogenital malformations are some of the most severe congenital anomalies encountered in children. Only a few molecular cues have been identified in early formation of the female urogenital system. Here we describe a novel long non-coding RNA molecule known as Leat1 (long non-coding RNA, EphrinB2 associated transcript 1). This lncRNA is syntenic with EfnB2 (which encodes EphrinB2) and expressed during embryonic development of the genital tubercle. While lncRNAs have varied functions, many are known to regulate their neighbouring genes. Eph/Ephrin bidirectional signaling molecules mediate many patterning pathways in early embryonic development, including cloacal septation and urethral development. Here we investigate the role of Leat1 and its possible regulation of EphrinB2 during development of the female reproductive tract. We show that a loss of Leat1 leads to reduced EfnB2 expression in the developing female genital tubercle, reduced anogenital distance and decreased fertility.


Assuntos
Efrina-B2/genética , Organogênese/genética , RNA Longo não Codificante/genética , Anormalidades Urogenitais/genética , Animais , Embrião de Mamíferos , Desenvolvimento Embrionário/genética , Feminino , Regulação da Expressão Gênica no Desenvolvimento , Humanos , Infertilidade Feminina/genética , Infertilidade Feminina/patologia , Camundongos
11.
Dev Dyn ; 249(11): 1377-1386, 2020 11.
Artigo em Inglês | MEDLINE | ID: mdl-32767478

RESUMO

BACKGROUND: The uterus is the location where multiple events occur that are required for the start of new life in mammals. The adult uterus contains endometrial or uterine glands that are essential for female fertility. In the mouse, uterine glands are located in the lateral and antimesometrial regions of the uterine horn. Previous three-dimensional (3D)-imaging of the adult uterus, its glands, and implanting embryos has been performed by multiple groups, using fluorescent microscopy. Adenogenesis, the formation of uterine glands, initiates after birth. Recently, we created a 3D-staging system of mouse uterine gland development at postnatal time points, using light sheet fluorescent microscopy. Here, using a similar approach, we examine the morphological changes in the epithelium of the perinatal mouse uterus. RESULTS: The uterine epithelium exhibits dorsoventral (mesometrial-antimesometrial) patterning as early as 3 days after birth (P3), marked by the presence of the dorsally positioned developing uterine rail. Uterine gland buds are present beginning at P4. Novel morphological epithelial structures, including a ventral ridge and uterine segments were identified. CONCLUSIONS: The perinatal mouse uterine luminal epithelium develops dorsal-ventral morphologies at 3 to 4 days postpartum. Between 5 and 6 days postpartum uterine epithelial folds form, defining alternating left-right segments.


Assuntos
Embrião de Mamíferos/embriologia , Organogênese , Útero/embriologia , Animais , Epitélio/embriologia , Feminino , Criaturas Lendárias , Camundongos
12.
Dev Dyn ; 249(11): 1394-1403, 2020 11.
Artigo em Inglês | MEDLINE | ID: mdl-32745301

RESUMO

BACKGROUND: Precise manipulation of gene expression with temporal and spatial control is essential for functional analysis and determining cell lineage relationships in complex biological systems. The cyclic recombinase (Cre)-loxP system is commonly used for gene manipulation at desired times and places. However, specificity is dependent on the availability of tissue- or cell-specific regulatory elements used in combination with Cre. Here, we present CreLite, an optogenetically controlled Cre system using red light in developing zebrafish embryos. RESULTS: Cre activity is disabled by splitting Cre and fusing with the Arabidopsis thaliana red light-inducible binding partners, PhyB and PIF6. Upon red light illumination, the PhyB-CreC and PIF6-CreN fusion proteins come together in the presence of the cofactor phycocyanobilin (PCB) to restore Cre activity. Red light exposure of zebrafish embryos harboring a Cre-dependent multicolor fluorescent protein reporter injected with CreLite mRNAs and PCB resulted in Cre activity as measured by the generation of multispectral cell labeling in several different tissues. CONCLUSIONS: Our data show that CreLite can be used for gene manipulations in whole embryos or small groups of cells at different developmental stages, and suggests CreLite may also be useful for temporal and spatial control of gene expression in cell culture, ex vivo organ culture, and other animal models.


Assuntos
Arabidopsis , Integrases , Optogenética , Plantas Geneticamente Modificadas , Arabidopsis/genética , Arabidopsis/metabolismo , Integrases/genética , Integrases/metabolismo , Plantas Geneticamente Modificadas/genética , Plantas Geneticamente Modificadas/metabolismo
13.
Dev Biol ; 455(1): 1-9, 2019 11 01.
Artigo em Inglês | MEDLINE | ID: mdl-31301298

RESUMO

Anti-Müllerian hormone (AMH) is a member of the Transforming Growth Factor-ß family of secreted signaling proteins. AMH is expressed in Sertoli cells of the fetal and adult testes and granulosa cells of the postnatal ovary. AMH is required for the regression of the Müllerian ducts in mammalian fetuses during male differentiation. AMH signals through its Type II receptor, AMHR2. AMHR2 is expressed in mesenchyme adjacent to the Müllerian ducts, and in Sertoli, Leydig, and granulosa cells. Although AMH and AMHR2 genes have been identified in numerous vertebrate species, spontaneous or engineered mutations or variants have been found or created in only a few mammals and teleost fishes. AMH or AMHR2 mutations in mammals lead to the development of Persistent Müllerian Duct Syndrome (PMDS), a recessive condition in which affected males are fully virilized but retain Müllerian duct-derived tissues, including a uterus and oviducts, and in human and dog, undescended testes. Amh mutant female mice had accelerated ovarian primordial follicle recruitment, suggesting a role for AMH in regulating germ cells. amh and amhr2 mutations have also been experimentally generated in various teleost fishes. Depending on the fish species, loss of AMH signaling results in infertility, germ cell tumors, or male-to-female sex reversal. Here we compare the spectrum of phenotypes caused by AMH and AMHR2 mutations in a variety of vertebrate species. There are both common and unique phenotypes between species, highlighting the range of biological processes regulated by AMH signaling.


Assuntos
Hormônio Antimülleriano/genética , Transtorno 46,XY do Desenvolvimento Sexual/genética , Mutação , Receptores de Peptídeos/genética , Receptores de Fatores de Crescimento Transformadores beta/genética , Animais , Hormônio Antimülleriano/metabolismo , Transtorno 46,XY do Desenvolvimento Sexual/metabolismo , Feminino , Humanos , Masculino , Fenótipo , Receptores de Peptídeos/metabolismo , Receptores de Fatores de Crescimento Transformadores beta/metabolismo , Reprodução/genética , Especificidade da Espécie , Vertebrados/classificação , Vertebrados/metabolismo
14.
Genesis ; 57(11-12): e23335, 2019 11.
Artigo em Inglês | MEDLINE | ID: mdl-31513344

RESUMO

Dead-End 1 (DND1) encodes an RNA binding protein critical for viable primordial germ cells in vertebrates. When introduced into cancer cell lines, DND1 suppresses cell proliferation and enhances apoptosis. However, the molecular function of mammalian wild-type DND1 has mostly been studied in cell lines and not verified in the organism. To facilitate study of wild-type DND1 function in mammalian systems, we generated a novel transgenic mouse line, LSL-FM-DND1 flox/+ , which conditionally expresses genetically engineered, FLAG-tagged and myc-tagged DND1 in a cell type-specific manner. We report that FLAG-myc-DND1 is indeed expressed in specific tissues of the mouse when LSL-FM-DND1 flox/+ is combined with mouse strains expressing Cre-recombinase. LSL-FM-DND1 flox/+ mice are fertile with no overt health effects. We expressed FLAG-myc-DND1 in the pancreas and found that chronic, ectopic expression of FLAG-myc-DND1 led to increase in fasting glucose levels in older mice. Thus, this novel LSL-FM-DND1 flox/+ mouse strain will facilitate studies on the biological and molecular function of wild-type DND1.


Assuntos
Proteínas de Neoplasias/genética , Proteínas de Neoplasias/metabolismo , Animais , Linhagem Celular , Feminino , Células Germinativas/metabolismo , Humanos , Integrases , Masculino , Camundongos , Camundongos Transgênicos , RNA Mensageiro/genética , Proteínas de Ligação a RNA/metabolismo
15.
Dev Biol ; 442(2): 301-314, 2018 10 15.
Artigo em Inglês | MEDLINE | ID: mdl-30118662

RESUMO

Disruption of endometrial gland formation or function can cause female infertility. Formation of endometrial glands via tubulogenesis of luminal epithelial cells requires the establishment and maintenance of cell polarity and cell adhesion. The FERM domain-containing protein Merlin coordinates epithelial cell polarity and cell adhesion and is critical for epithelial tissue function in the skin and kidney. We now demonstrate a requirement for Merlin in endometrial gland development. Conditional deletion of Merlin in the endometrium results in female infertility caused by the absence of gland formation. Interestingly, we observed glandular epithelial markers within discrete groups of cells in the Merlin-deficient luminal epithelium. Wnt signaling, a pathway necessary for endometrial gland development is maintained in Merlin-deficient endometrium, suggesting the glandular fate program is active. Instead, we observe increased levels of apical actin and markers indicative of high membrane tension on the basal surface of the Merlin-deficient luminal epithelium. These findings suggest that the structural integrity of the luminal epithelium during gland formation is required for appropriate endometrial tubulogenesis and tissue function. Moreover, our work implicates Merlin-dependent regulation of mechanical tension in the proper formation of endometrial gland architecture and function.


Assuntos
Endométrio/crescimento & desenvolvimento , Neurofibromina 2/fisiologia , Animais , Adesão Celular/fisiologia , Polaridade Celular/fisiologia , Proteínas de Ligação a DNA/metabolismo , Proteínas de Ligação a DNA/fisiologia , Endométrio/citologia , Endométrio/metabolismo , Endométrio/patologia , Células Epiteliais/metabolismo , Feminino , Hibridização In Situ , Infertilidade Feminina , Camundongos , Camundongos Endogâmicos C57BL , Morfogênese/fisiologia , Neurofibromina 2/deficiência , Neurofibromina 2/genética , Neurofibromina 2/metabolismo , Transdução de Sinais , Fatores de Transcrição/metabolismo , Fatores de Transcrição/fisiologia
16.
Development ; 142(11): 2069-79, 2015 Jun 01.
Artigo em Inglês | MEDLINE | ID: mdl-25977363

RESUMO

Lhx1 encodes a LIM homeobox transcription factor that is expressed in the primitive streak, mesoderm and anterior mesendoderm of the mouse embryo. Using a conditional Lhx1 flox mutation and three different Cre deleters, we demonstrated that LHX1 is required in the anterior mesendoderm, but not in the mesoderm, for formation of the head. LHX1 enables the morphogenetic movement of cells that accompanies the formation of the anterior mesendoderm, in part through regulation of Pcdh7 expression. LHX1 also regulates, in the anterior mesendoderm, the transcription of genes encoding negative regulators of WNT signalling, such as Dkk1, Hesx1, Cer1 and Gsc. Embryos carrying mutations in Pcdh7, generated using CRISPR-Cas9 technology, and embryos without Lhx1 function specifically in the anterior mesendoderm displayed head defects that partially phenocopied the truncation defects of Lhx1-null mutants. Therefore, disruption of Lhx1-dependent movement of the anterior mesendoderm cells and failure to modulate WNT signalling both resulted in the truncation of head structures. Compound mutants of Lhx1, Dkk1 and Ctnnb1 show an enhanced head truncation phenotype, pointing to a functional link between LHX1 transcriptional activity and the regulation of WNT signalling. Collectively, these results provide comprehensive insight into the context-specific function of LHX1 in head formation: LHX1 enables the formation of the anterior mesendoderm that is instrumental for mediating the inductive interaction with the anterior neuroectoderm and LHX1 also regulates the expression of factors in the signalling cascade that modulate the level of WNT activity.


Assuntos
Embrião de Mamíferos/metabolismo , Cabeça/embriologia , Proteínas com Homeodomínio LIM/metabolismo , Fatores de Transcrição/metabolismo , Animais , Caderinas/metabolismo , Endoderma/citologia , Endoderma/metabolismo , Deleção de Genes , Regulação da Expressão Gênica no Desenvolvimento , Camadas Germinativas/citologia , Camadas Germinativas/metabolismo , Proteínas com Homeodomínio LIM/genética , Camundongos Knockout , Modelos Biológicos , Mutação , Fenótipo , Transdução de Sinais , Fatores de Transcrição/genética , Proteínas Wnt/metabolismo
17.
Mol Reprod Dev ; 85(5): 397-405, 2018 05.
Artigo em Inglês | MEDLINE | ID: mdl-29543367

RESUMO

Endometrial or uterine glands secrete substances essential for uterine receptivity to the embryo, implantation, conceptus survival, and growth. Adenogenesis is the process of gland formation within the stroma of the uterus. In the mouse, uterine gland formation initiates at postnatal day (P) 5. Uterine gland morphology is poorly understood because it is primarily based on two-dimensional (2D) histology. To more fully describe uterine gland morphogenesis, we generated three-dimensional (3D) models of postnatal uterine glands from P0 to P21, based on volumetric imaging using light sheet microscopy. At birth (P0), there were no glands. At P8, we found bud- and teardrop-shaped epithelial invaginations. By P11, the forming glands were elongated epithelial tubes. By P21, the elongated tubes had a sinuous morphology. These morphologies are homogeneously distributed along the anterior-posterior axis of the uterus. To facilitate uterine gland analyses, we propose a novel 3D staging system of uterine gland morphology during development in the prepubertal mouse. We define five uterine gland stages: Stage 1: bud; Stage 2: teardrop; Stage 3: elongated; Stage 4: sinuous; and Stage 5: primary branches. This staging system provides a standardized key to assess and quantify prepubertal uterine gland morphology that can be used for studies of uterine gland development and pathology. In addition, our studies suggest that gland formation initiation occurs during P8 and P11. However, between P11 and P21 gland formation initiation stops and all glands elongate and become sinuous. We also found that the mesometrial epithelium develops a unique morphology we term the uterine rail.


Assuntos
Endométrio/citologia , Endométrio/crescimento & desenvolvimento , Maturidade Sexual/fisiologia , Animais , Epitélio/crescimento & desenvolvimento , Feminino , Camundongos
18.
J Bone Miner Metab ; 36(4): 410-419, 2018 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-28770354

RESUMO

Sex-determining region Y (Sry)-box (Sox)9 is required for chondrogenesis as a transcriptional activator of genes related to chondrocyte proliferation, differentiation, and cartilage-specific extracellular matrix. Although there have been studies investigating the Sox9-dependent transcriptional complexes, not all their components have been identified. In the present study, we demonstrated that thyroid hormone receptor-associated protein (THRAP)3 is a component of a SOX9 transcriptional complex by liquid chromatography mass spectrometric analysis of FLAG-tagged Sox9-binding proteins purified from FLAG-HA-tagged Sox9 knock-in mice. Thrap3 knockdown in ATDC5 chondrogenic cells increased the expression of Collagen type II alpha 1 chain (Col2a1) without affecting Sox9 expression. THRAP3 and SOX9 overexpression reduced Col2a1 levels to a greater degree than overexpression of SOX9 alone. The negative regulation of SOX9 transcriptional activity by THRAP3 was mediated by interaction between the proline-, glutamine-, and serine-rich domain of SOX9 and the innominate domain of THRAP3. These results indicate that THRAP3 negatively regulates SOX9 transcriptional activity as a cofactor of a SOX9 transcriptional complex during chondrogenesis.


Assuntos
Condrogênese , Proteínas de Ligação a DNA/metabolismo , Proteínas Nucleares/metabolismo , Fatores de Transcrição SOX9/metabolismo , Fatores de Transcrição/metabolismo , Transcrição Gênica , Animais , Núcleo Celular/metabolismo , Condrócitos/citologia , Condrócitos/metabolismo , Técnicas de Introdução de Genes , Lâmina de Crescimento/metabolismo , Células HEK293 , Humanos , Camundongos Endogâmicos C57BL , Ligação Proteica
19.
PLoS Genet ; 11(8): e1005398, 2015 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-26317994

RESUMO

Variation among individuals is a prerequisite of evolution by natural selection. As such, identifying the origins of variation is a fundamental goal of biology. We investigated the link between gene interactions and variation in gene expression among individuals and species using the mammalian limb as a model system. We first built interaction networks for key genes regulating early (outgrowth; E9.5-11) and late (expansion and elongation; E11-13) limb development in mouse. This resulted in an Early (ESN) and Late (LSN) Stage Network. Computational perturbations of these networks suggest that the ESN is more robust. We then quantified levels of the same key genes among mouse individuals and found that they vary less at earlier limb stages and that variation in gene expression is heritable. Finally, we quantified variation in gene expression levels among four mammals with divergent limbs (bat, opossum, mouse and pig) and found that levels vary less among species at earlier limb stages. We also found that variation in gene expression levels among individuals and species are correlated for earlier and later limb development. In conclusion, results are consistent with the robustness of the ESN buffering among-individual variation in gene expression levels early in mammalian limb development, and constraining the evolution of early limb development among mammalian species.


Assuntos
Extremidades/embriologia , Regulação da Expressão Gênica no Desenvolvimento/genética , Redes Reguladoras de Genes/genética , Botões de Extremidades/embriologia , Animais , Evolução Biológica , Quirópteros/genética , Simulação por Computador , Extremidades/crescimento & desenvolvimento , Expressão Gênica/genética , Variação Genética/genética , Botões de Extremidades/citologia , Botões de Extremidades/crescimento & desenvolvimento , Camundongos , Gambás/genética , Seleção Genética , Suínos/genética
20.
BMC Evol Biol ; 17(1): 86, 2017 03 23.
Artigo em Inglês | MEDLINE | ID: mdl-28335721

RESUMO

BACKGROUND: From bat wings to whale flippers, limb diversification has been crucial to the evolutionary success of mammals. We performed the first transcriptome-wide study of limb development in multiple species to explore the hypothesis that mammalian limb diversification has proceeded through the differential expression of conserved shared genes, rather than by major changes to limb patterning. Specifically, we investigated the manner in which the expression of shared genes has evolved within and among mammalian species. RESULTS: We assembled and compared transcriptomes of bat, mouse, opossum, and pig fore- and hind limbs at the ridge, bud, and paddle stages of development. Results suggest that gene expression patterns exhibit larger variation among species during later than earlier stages of limb development, while within species results are more mixed. Consistent with the former, results also suggest that genes expressed at later developmental stages tend to have a younger evolutionary age than genes expressed at earlier stages. A suite of key limb-patterning genes was identified as being differentially expressed among the homologous limbs of all species. However, only a small subset of shared genes is differentially expressed in the fore- and hind limbs of all examined species. Similarly, a small subset of shared genes is differentially expressed within the fore- and hind limb of a single species and among the forelimbs of different species. CONCLUSIONS: Taken together, results of this study do not support the existence of a phylotypic period of limb development ending at chondrogenesis, but do support the hypothesis that the hierarchical nature of development translates into increasing variation among species as development progresses.


Assuntos
Regulação da Expressão Gênica no Desenvolvimento , Mamíferos/classificação , Mamíferos/genética , Animais , Evolução Biológica , Extremidades/anatomia & histologia , Extremidades/crescimento & desenvolvimento , Extremidades/fisiologia , Mamíferos/anatomia & histologia , Mamíferos/crescimento & desenvolvimento , Transcriptoma , Asas de Animais
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA