Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 9 de 9
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Proc Natl Acad Sci U S A ; 121(6): e2309466121, 2024 Feb 06.
Artigo em Inglês | MEDLINE | ID: mdl-38300866

RESUMO

Congenital anomalies of the lower genitourinary (LGU) tract are frequently comorbid due to genetically linked developmental pathways, and are among the most common yet most socially stigmatized congenital phenotypes. Genes involved in sexual differentiation are prime candidates for developmental anomalies of multiple LGU organs, but insufficient prospective screening tools have prevented the rapid identification of causative genes. Androgen signaling is among the most influential modulators of LGU development. The present study uses SpDamID technology in vivo to generate a comprehensive map of the pathways actively regulated by the androgen receptor (AR) in the genitalia in the presence of the p300 coactivator, identifying wingless/integrated (WNT) signaling as a highly enriched AR-regulated pathway in the genitalia. Transcription factor (TF) hits were then assayed for sexually dimorphic expression at two critical time points and also cross-referenced to a database of clinically relevant copy number variations to identify 252 TFs exhibiting copy variation in patients with LGU phenotypes. A subset of 54 TFs was identified for which LGU phenotypes are statistically overrepresented as a proportion of total observed phenotypes. The 252 TF hitlist was then subjected to a functional screen to identify hits whose silencing affects genital mesenchymal growth rates. Overlap of these datasets results in a refined list of 133 TFs of both functional and clinical relevance to LGU development, 31 of which are top priority candidates, including the well-documented renal progenitor regulator, Sall1. Loss of Sall1 was examined in vivo and confirmed to be a powerful regulator of LGU development.


Assuntos
Variações do Número de Cópias de DNA , Sistema Urinário , Humanos , Estudos Prospectivos , Androgênios/metabolismo , Genitália/metabolismo , Sistema Urinário/metabolismo , Fatores de Transcrição/genética , Fatores de Transcrição/metabolismo
2.
Mol Ther Nucleic Acids ; 32: 937-948, 2023 Jun 13.
Artigo em Inglês | MEDLINE | ID: mdl-37346979

RESUMO

Dominant missense mutations in DNAJB6, a co-chaperone of HSP70, cause limb girdle muscular dystrophy (LGMD) D1. No treatments are currently available. Two isoforms exist, DNAJB6a and DNAJB6b, each with distinct localizations in muscle. Mutations reside in both isoforms, yet evidence suggests that DNAJB6b is primarily responsible for disease pathogenesis. Knockdown treatment strategies involving both isoforms carry risk, as DNAJB6 knockout is embryonic lethal. We therefore developed an isoform-specific knockdown approach using morpholinos. Selective reduction of each isoform was achieved in vitro in primary mouse myotubes and human LGMDD1 myoblasts, as well as in vivo in mouse skeletal muscle. To assess isoform specific knockdown in LGMDD1, we created primary myotube cultures from a knockin LGMDD1 mouse model. Using mass spectrometry, we identified an LGMDD1 protein signature related to protein homeostasis and myofibril structure. Selective reduction of DNAJB6b levels in LGMDD1 myotubes corrected much of the proteomic disease signature toward wild type levels. Additional in vivo functional data is required to determine if selective reduction of DNAJB6b is a viable therapeutic target for LGMDD1.

3.
PNAS Nexus ; 2(1): pgac300, 2023 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-36712925

RESUMO

Sexually dimorphic outgrowth and differentiation of the embryonic genital tubercles (GTs) give rise to the penis in males and the clitoris in females. Defects in androgen production or in response to androgen signaling can lead to various congenital penile anomalies in both mice and humans. Due to lack of a high-throughput screening system, identification of crucial regulators of GT sexual differentiation has been slow. To overcome this research barrier, we isolated embryonic GT mesenchymal (GTme) cells to model genitalia growth and differentiation in vitro. Using either a mechanical or fluorescence-activated cell sorting-assisted purification method, GTme cells were isolated and assayed for their proliferation using a microscopy and image analysis system, on a single cell level over time. Male and female GTme cells inherently exhibit different cellular dynamics, consistent with their in-vivo behaviors. This system allows for the rapid quantitative analyses of numerous drug treatments, and enables the discovery of potential genetic modulators of GT morphogenesis on a large scale. Using this system, we completed a 438-compound library screen and identified 82 kinase inhibitor hits. In mice, in-utero exposure to one such candidate kinase inhibitor, Cediranib, resulted in embryos with severe genitalia defects, especially in males. Gene silencing by RNAi was optimized in this system, laying the foundation for future larger-scale genetic screenings. These findings demonstrate the power of this novel high-throughput system to rapidly and successfully identify modulators of genitalia growth and differentiation, expanding the toolbox for the study of functional genomics and environmental factors.

4.
JCI Insight ; 6(17)2021 09 08.
Artigo em Inglês | MEDLINE | ID: mdl-34292881

RESUMO

Retinoic acid (RA) signaling has long been speculated to regulate embryo implantation, because many enzymes and proteins responsible for maintaining RA homeostasis and transducing RA signals are tightly regulated in the endometrium during this critical period. However, due to a lack of genetic data, it was unclear whether RA signaling is truly required for implantation and which specific RA signaling cascades are at play. Herein we utilize a genetic murine model that expresses a dominant-negative form of RA receptor (RAR) specifically in female reproductive organs to show that functional RA signaling is fundamental to female fertility, particularly implantation and decidualization. Reduction in RA signaling activity severely affects the ability of the uterus to achieve receptive status and decidualize, partially through dampening follistatin expression and downstream activin B/bone morphogenetic protein 2 signaling. To confirm translational relevance of these findings to humans, human endometrial stromal cells (hESCs) were treated with a pan-RAR antagonist to show that in vitro decidualization is impaired. RNA interference perturbation of individual RAR transcripts in hESCs revealed that RARα in particular was essential for proper decidualization. These data provide direct functional evidence that uterine RAR-mediated RA signaling was crucial for mammalian embryo implantation, and its disruption led to failure of uterine receptivity and decidualization, resulting in severely compromised fertility.


Assuntos
DNA/genética , Endométrio/metabolismo , Mutação , Receptores do Ácido Retinoico/genética , Útero/metabolismo , Animais , Diferenciação Celular , Células Cultivadas , Análise Mutacional de DNA , Endométrio/citologia , Feminino , Masculino , Camundongos , Camundongos Mutantes , Receptores do Ácido Retinoico/metabolismo , Transdução de Sinais , Útero/citologia
5.
Dis Model Mech ; 13(8)2020 08 18.
Artigo em Inglês | MEDLINE | ID: mdl-32571845

RESUMO

Wnt/ß-catenin signaling has an essential role in eye development. Faulty regulation of this pathway results in ocular malformations, owing to defects in cell-fate determination and differentiation. Herein, we show that disruption of Maz, the gene encoding Myc-associated zinc-finger transcription factor, produces developmental eye defects in mice and humans. Expression of key genes involved in the Wnt cascade, Sfrp2, Wnt2b and Fzd4, was significantly increased in mice with targeted inactivation of Maz, resulting in abnormal peripheral eye formation with reduced proliferation of the progenitor cells in the region. Paradoxically, the Wnt reporter TCF-Lef1 displayed a significant downregulation in Maz-deficient eyes. Molecular analysis indicates that Maz is necessary for the activation of the Wnt/ß-catenin pathway and participates in the network controlling ciliary margin patterning. Copy-number variations and single-nucleotide variants of MAZ were identified in humans that result in abnormal ocular development. The data support MAZ as a key contributor to the eye comorbidities associated with chromosome 16p11.2 copy-number variants and as a transcriptional regulator of ocular development.


Assuntos
Proteínas de Ligação a DNA/metabolismo , Anormalidades do Olho/metabolismo , Olho/metabolismo , Fatores de Transcrição/metabolismo , Adolescente , Adulto , Animais , Apoptose , Proliferação de Células , Pré-Escolar , Variações do Número de Cópias de DNA , Proteínas de Ligação a DNA/genética , Olho/patologia , Anormalidades do Olho/genética , Anormalidades do Olho/patologia , Feminino , Dosagem de Genes , Regulação da Expressão Gênica no Desenvolvimento , Predisposição Genética para Doença , Humanos , Lactente , Masculino , Camundongos Endogâmicos C57BL , Camundongos Knockout , Morfogênese , Fenótipo , Polimorfismo de Nucleotídeo Único , Fatores de Transcrição/genética , Via de Sinalização Wnt , Adulto Jovem
6.
Proc Natl Acad Sci U S A ; 116(39): 19541-19551, 2019 09 24.
Artigo em Inglês | MEDLINE | ID: mdl-31501330

RESUMO

Failure of embryo implantation accounts for a significant percentage of female infertility. Exquisitely coordinated molecular programs govern the interaction between the competent blastocyst and the receptive uterus. Decidualization, the rapid proliferation and differentiation of endometrial stromal cells into decidual cells, is required for implantation. Decidualization defects can cause poor placentation, intrauterine growth restriction, and early parturition leading to preterm birth. Decidualization has not yet been systematically studied at the genetic level due to the lack of a suitable high-throughput screening tool. Herein we describe the generation of an immortalized human endometrial stromal cell line that uses yellow fluorescent protein under the control of the prolactin promoter as a quantifiable visual readout of the decidualization response (hESC-PRLY cells). Using this cell line, we performed a genome-wide siRNA library screen, as well as a screen of 910 small molecules, to identify more than 4,000 previously unrecognized genetic and chemical modulators of decidualization. Ontology analysis revealed several groups of decidualization modulators, including many previously unappreciated transcription factors, sensory receptors, growth factors, and kinases. Expression studies of hits revealed that the majority of decidualization modulators are acutely sensitive to ovarian hormone exposure. Gradient treatment of exogenous factors was used to identify EC50 values of small-molecule hits, as well as verify several growth factor hits identified by the siRNA screen. The high-throughput decidualization reporter cell line and the findings described herein will aid in the development of patient-specific treatments for decidualization-based recurrent pregnancy loss, subfertility, and infertility.


Assuntos
Decídua/metabolismo , Endométrio/metabolismo , Infertilidade Feminina/metabolismo , Blastocisto/metabolismo , Diferenciação Celular/genética , Diferenciação Celular/fisiologia , Linhagem Celular , Decídua/fisiologia , Implantação do Embrião/fisiologia , Endométrio/patologia , Feminino , Fertilidade , Estudo de Associação Genômica Ampla , Humanos , Infertilidade Feminina/genética , Gravidez , Prolactina/metabolismo , RNA Interferente Pequeno/genética , Receptores de Progesterona/genética , Células Estromais/metabolismo , Útero/metabolismo , Útero/patologia
7.
Differentiation ; 110: 1-7, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-31521888

RESUMO

Fertilization requires the physical combination of gametes, and terrestrial mammals necessitated the evolution of genitalia capable of successfully completing the fertilization process in a non-aqueous environment. Thus, the male mammalian external genitalia evolved as an outgrowth from the body, an appendage sufficient to fertilize eggs housed deep inside the female. In this way, sexual dimorphism of mammalian genitalia became highly pronounced. This highly complex evolutionary divergence both from aqueous fertilization, as well as divergence between the sexes of terrestrial mammals, required exquisitely coordinated, novel patterns of gene expression to regulate the spatial and temporal events governing external genitalia development. Recent studies delineating the genetic regulation of external genitalia development, largely focusing on development of the murine genital tubercle, have vastly enlightened the field of reproductive developmental biology. Murine homologs of human genes have been selectively deleted in the mouse, either in the whole body or using tissue-specific and temporally-specific genetic drivers. The defects in outgrowth and urethral tubularization subsequent to the deletion of specific genes in the developing murine external genitalia delineates which genes are required in which compartments and at what times. This review details how these murine genetic models have created a somewhat modest but rapidly growing library of knowledge detailing the spatial-temporal genetic regulation of external genitalia development.


Assuntos
Regulação da Expressão Gênica no Desenvolvimento/genética , Genitália/crescimento & desenvolvimento , Organogênese/fisiologia , Transdução de Sinais/genética , Animais , Proteínas Hedgehog/metabolismo , Humanos , Mamíferos
8.
Proc Natl Acad Sci U S A ; 115(8): E1849-E1858, 2018 02 20.
Artigo em Inglês | MEDLINE | ID: mdl-29432158

RESUMO

Genitourinary (GU) birth defects are among the most common yet least studied congenital malformations. Congenital anomalies of the kidney and urinary tract (CAKUTs) have high morbidity and mortality rates and account for ∼30% of structural birth defects. Copy number variation (CNV) mapping revealed that 16p11.2 is a hotspot for GU development. The only gene covered collectively by all of the mapped GU-patient CNVs was MYC-associated zinc finger transcription factor (MAZ), and MAZ CNV frequency is enriched in nonsyndromic GU-abnormal patients. Knockdown of MAZ in HEK293 cells results in differential expression of several WNT morphogens required for normal GU development, including Wnt11 and Wnt4. MAZ knockdown also prevents efficient transition into S phase, affects transcription of cell-cycle regulators, and abrogates growth of human embryonic kidney cells. Murine Maz is ubiquitously expressed, and a CRISPR-Cas9 mouse model of Maz deletion results in perinatal lethality with survival rates dependent on Maz copy number. Homozygous loss of Maz results in high penetrance of CAKUTs, and Maz is haploinsufficient for normal bladder development. MAZ, once thought to be a simple housekeeping gene, encodes a dosage-sensitive transcription factor that regulates urogenital development and contributes to both nonsyndromic congenital malformations of the GU tract as well as the 16p11.2 phenotype.


Assuntos
Proteínas de Ligação a DNA/metabolismo , Fatores de Transcrição/metabolismo , Bexiga Urinária/anormalidades , Anormalidades Urogenitais/genética , Animais , Adesão Celular , Cromossomos Humanos Par 16 , Proteínas de Ligação a DNA/genética , Deleção de Genes , Regulação da Expressão Gênica no Desenvolvimento , Técnicas de Silenciamento de Genes , Células HEK293 , Humanos , Masculino , Camundongos , Regiões Promotoras Genéticas , Interferência de RNA , RNA Interferente Pequeno , Fatores de Transcrição/genética , Transcrição Gênica , Anormalidades Urogenitais/patologia
9.
Proc Natl Acad Sci U S A ; 114(19): 4981-4986, 2017 05 09.
Artigo em Inglês | MEDLINE | ID: mdl-28439006

RESUMO

The spectrum of congenital anomalies affecting either the upper tract (kidneys and ureters) or lower tract (reproductive organs) of the genitourinary (GU) system are fundamentally linked by the developmental origin of multiple GU tissues, including the kidneys, gonads, and reproductive ductal systems: the intermediate mesoderm. Although ∼31% of DiGeorge/del22q11.2 syndrome patients exhibit GU defects, little focus has been placed on the molecular etiology of GU defects in this syndrome. Among del22q11.2 patients exhibiting GU anomalies, we have mapped the smallest relevant region to only five genes, including CRKLCRKL encodes a src-homology adaptor protein implicated in mediating tyrosine kinase signaling, and is expressed in the developing GU-tract in mice and humans. Here we show that Crkl mutant embryos exhibit gene dosage-dependent growth restriction, and homozygous mutants exhibit upper GU defects at a microdissection-detectable rate of 23%. RNA-sequencing revealed that 52 genes are differentially regulated in response to uncoupling Crkl from its signaling pathways in the developing kidney, including a fivefold up-regulation of Foxd1, a known regulator of nephron progenitor differentiation. Additionally, Crkl heterozygous adult males exhibit cryptorchidism, lower testis weight, lower sperm count, and subfertility. Together, these data indicate that CRKL is intimately involved in normal development of both the upper and lower GU tracts, and disruption of CRKL contributes to the high incidence of GU defects associated with deletion at 22q11.2.


Assuntos
Proteínas Adaptadoras de Transdução de Sinal/metabolismo , Cromossomos Humanos Par 22/metabolismo , Regulação da Expressão Gênica no Desenvolvimento , Genitália , Proteínas Nucleares/metabolismo , Sistema Urinário , Proteínas Adaptadoras de Transdução de Sinal/genética , Animais , Cromossomos Humanos Par 22/genética , Feminino , Genitália/anormalidades , Genitália/embriologia , Humanos , Masculino , Camundongos , Camundongos Knockout , Proteínas Nucleares/genética , Sistema Urinário/anormalidades , Sistema Urinário/embriologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...