Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 19 de 19
Filtrar
Mais filtros

Base de dados
Tipo de documento
Intervalo de ano de publicação
1.
Pediatr Res ; 93(3): 604-611, 2023 02.
Artigo em Inglês | MEDLINE | ID: mdl-36280708

RESUMO

BACKGROUND: Preclinical data demonstrate that opioids modulate brain reward signaling through an inflammatory cascade, but this relationship has yet to be studied in opioid-exposed neonates. METHODS: Saliva samples of 54 opioid-exposed and sex- and age-matched non-exposed neonates underwent transcriptomic analysis of inflammatory and reward genes. A subset of 22 neonates underwent brain magnetic resonance imaging (MRI) to evaluate white matter injury commonly associated with inflammatory response. Gene expression and brain MRI were compared between opioid- and non-exposed neonates and further stratified by sex and pharmacotherapy need. RESULTS: Opioid-exposed females regardless of pharmacotherapy need had higher expression of inflammatory genes than their male counterparts, with notable differences in the expression of CCL2 and CXCL1 in females requiring pharmacotherapy (p = 0.01 and 0.06, respectively). Opioid-exposed males requiring pharmacotherapy had higher expression of DRD2 than exposed females (p = 0.07), validating our prior research. Higher expression of IL1ß, IL6, TNFα, and IL10 was seen in opioid-exposed neonates with T1 white matter hyperintensity (WMH) compared to exposed neonates without WMH (p < 0.05). CONCLUSION: Prenatal opioid exposure may promote inflammation resulting in changes in reward signaling and white matter injury in the developing brain, with unique sex-specific effects. The actions of opioids through non-neuronal pathways need further investigation. IMPACT: Opioid-exposed neonates are at risk for punctate T1 white matter hyperintensity (WMH). Females carry a greater propensity for WMH. Salivary transcriptomic data showed significantly higher expression of inflammatory genes in opioid-exposed neonates with WMH than those without WMH, irrespective of pharmacotherapy need. Adding to prior studies, our findings suggest that prenatal opioid exposure may modulate white matter injury and reward signaling through a pro-inflammatory process that is sex specific. This novel study highlights the short-term molecular and structural effects of prenatal opioids and the need to elucidate the long-term impact of prenatal opioid exposure.


Assuntos
Lesões Encefálicas , Substância Branca , Recém-Nascido , Feminino , Gravidez , Masculino , Humanos , Substância Branca/diagnóstico por imagem , Substância Branca/patologia , Analgésicos Opioides/efeitos adversos , Projetos Piloto , Encéfalo , Imageamento por Ressonância Magnética/métodos , Lesões Encefálicas/patologia
2.
J Pediatr ; 214: 60-65.e2, 2019 11.
Artigo em Inglês | MEDLINE | ID: mdl-31474426

RESUMO

OBJECTIVES: To evaluate salivary biomarkers that elucidate the molecular mechanisms by which in utero opioid exposure exerts sex-specific effects on select hypothalamic and reward genes driving hyperphagia, a hallmark symptom of infants suffering from neonatal opioid withdrawal syndrome (NOWS). STUDY DESIGN: We prospectively collected saliva from 50 newborns born at ≥34 weeks of gestational age with prenatal opioid exposure and 50 sex- and gestational age-matched infants without exposure. Saliva underwent transcriptomic analysis for 4 select genes involved in homeostatic and hedonic feeding regulation (neuropeptide Y2 receptor [NPY2R], proopiomelanocortin [POMC], leptin receptor [LEPR], dopamine type 2 receptor [DRD2]). Normalized gene expression data were stratified based on sex and correlated with feeding volume on day of life 7 and length of stay in infants with NOWS requiring pharmacotherapy. RESULTS: Expression of DRD2, a hedonistic/reward regulator, was significantly higher in male newborns compared with female newborns with NOWS (Δ threshold cycle 10.8 ± 3.8 vs 13.9 ± 3.7, P = .01). In NOWS requiring pharmacotherapy expression of leptin receptor, an appetite suppressor, was higher in male subjects than female subjects (Δ threshold cycle 8.4 ± 2.5 vs 12.4 ± 5.1, P = .05), DRD2 expression significantly correlated with intake volume on day of life 7 (r = 0.58, P = .02), and expression of NPY2R, an appetite regulator, negatively correlated with length of stay (r = -0.24, P = .05). CONCLUSIONS: Prenatal opioid exposure exerts sex-dependent effects on hypothalamic feeding regulatory genes with clinical correlations. Neonatal salivary gene expression analyses may predict hyperphagia, severity of withdrawal state, and length of stay in infants with NOWS.


Assuntos
Analgésicos Opioides/efeitos adversos , Expressão Gênica , Hiperfagia/etiologia , Síndrome de Abstinência Neonatal/genética , Saliva/química , Estudos de Casos e Controles , Feminino , Perfilação da Expressão Gênica , Marcadores Genéticos , Humanos , Recém-Nascido , Masculino , Síndrome de Abstinência Neonatal/complicações , Projetos Piloto , Pró-Opiomelanocortina/genética , Estudos Prospectivos , Receptores de Dopamina D2/genética , Receptores para Leptina/genética , Receptores de Neuropeptídeo Y/genética , Índice de Gravidade de Doença , Fatores Sexuais
3.
PLoS Genet ; 8(8): e1002906, 2012.
Artigo em Inglês | MEDLINE | ID: mdl-22916036

RESUMO

Germline mutations of the Liver Kinase b1 (LKB1/STK11) tumor suppressor gene have been linked to Peutz-Jeghers Syndrome (PJS), an autosomal-dominant, cancer-prone disorder in which patients develop neoplasms in several organs, including the oviduct, ovary, and cervix. We have conditionally deleted Lkb1 in Müllerian duct mesenchyme-derived cells of the female reproductive tract and observed expansion of the stromal compartment and hyperplasia and/or neoplasia of adjacent epithelial cells throughout the reproductive tract with paratubal cysts and adenomyomas in oviducts and, eventually, endometrial cancer. Examination of the proliferation marker phospho-histone H3 and mammalian Target Of Rapamycin Complex 1 (mTORC1) pathway members revealed increased proliferation and mTORC1 activation in stromal cells of both the oviduct and uterus. Treatment with rapamycin, an inhibitor of mTORC1 activity, decreased tumor burden in adult Lkb1 mutant mice. Deletion of the genes for Tuberous Sclerosis 1 (Tsc1) or Tsc2, regulators of mTORC1 that are downstream of LKB1 signaling, in the oviductal and uterine stroma phenocopies some of the defects observed in Lkb1 mutant mice, confirming that dysregulated mTORC1 activation in the Lkb1-deleted stroma contributes to the phenotype. Loss of PTEN, an upstream regulator of mTORC1 signaling, along with Lkb1 deletion significantly increased tumor burden in uteri and induced tumorigenesis in the cervix and vagina. These studies show that LKB1/TSC1/TSC2/mTORC1 signaling in mesenchymal cells is important for the maintenance of epithelial integrity and suppression of carcinogenesis in adjacent epithelial cells. Because similar changes in the stromal population are also observed in human oviductal/ovarian adenoma and endometrial adenocarcinoma patients, we predict that dysregulated mTORC1 activity by upstream mechanisms similar to those described in these model systems contributes to the pathogenesis of these human diseases.


Assuntos
Adenoma/genética , Neoplasias do Endométrio/genética , PTEN Fosfo-Hidrolase/genética , Proteínas Serina-Treonina Quinases/genética , Proteínas/genética , Proteínas Supressoras de Tumor/genética , Proteínas Quinases Ativadas por AMP , Adenoma/tratamento farmacológico , Adenoma/patologia , Animais , Colo do Útero/efeitos dos fármacos , Colo do Útero/metabolismo , Colo do Útero/patologia , Modelos Animais de Doenças , Neoplasias do Endométrio/tratamento farmacológico , Neoplasias do Endométrio/patologia , Endométrio/efeitos dos fármacos , Endométrio/metabolismo , Endométrio/patologia , Feminino , Deleção de Genes , Regulação Neoplásica da Expressão Gênica/efeitos dos fármacos , Humanos , Alvo Mecanístico do Complexo 1 de Rapamicina , Células-Tronco Mesenquimais/efeitos dos fármacos , Células-Tronco Mesenquimais/metabolismo , Células-Tronco Mesenquimais/patologia , Camundongos , Complexos Multiproteicos , Ovário/efeitos dos fármacos , Ovário/metabolismo , Ovário/patologia , Oviductos/efeitos dos fármacos , Oviductos/metabolismo , Oviductos/patologia , PTEN Fosfo-Hidrolase/deficiência , Proteínas Serina-Treonina Quinases/deficiência , Proteínas/antagonistas & inibidores , Transdução de Sinais/efeitos dos fármacos , Sirolimo/administração & dosagem , Serina-Treonina Quinases TOR , Proteína 1 do Complexo Esclerose Tuberosa , Proteína 2 do Complexo Esclerose Tuberosa , Microambiente Tumoral/efeitos dos fármacos , Microambiente Tumoral/genética , Proteínas Supressoras de Tumor/deficiência
4.
Carcinogenesis ; 35(3): 546-53, 2014 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-24170201

RESUMO

Epithelial ovarian cancer presents mostly with serous, endometrioid or mucinous histology but is treated as a single disease. The development of histotype-specific therapy has been challenging because of the relative lack of studies attributing disrupted pathways to a distinct histotype differentiation. mTOR activation is frequently associated with poor prognosis in serous ovarian cancer, which is the most common and most deadly histotype. However, the mechanisms dysregulating mTOR in the pathogenesis of ovarian cancer are unknown. We detected copy number loss and correlated lower expression levels of LKB1, TSC1, TSC2 and PTEN tumor suppressor genes for upstream regulators of mTOR activity in up to 80% in primary ovarian serous tumor databases, with LKB1 allelic loss-predominant. Reduced LKB1 protein was usually associated with increased mTOR activity in both serous ovarian cancer cell lines and primary tumors. Conditional deletion of Lkb1 in murine ovarian surface epithelial (OSE) cells caused papillary hyperplasia and shedding but not tumors. Simultaneous deletion of Lkb1 and Pten, however, led to development of high-grade ovarian serous histotype tumors with 100% penetrance that expressed WT1, ERα, PAX8, TP53 and cytokeratin 8, typical markers used in the differential diagnosis of serous ovarian cancer. Neither hysterectomy nor salpingectomy interfered with progression of ovarian tumorigenesis, suggesting that neither uterine nor Fallopian tube epithelial cells were contributing to tumorigenesis. These results implicate LKB1 loss in the OSE in the pathogenesis of serous ovarian cancer and provide a compelling rationale for investigating the therapeutic potential of targeting LKB1 signaling in patients with this deadly disease.


Assuntos
Genes Supressores de Tumor , Neoplasias Ovarianas/genética , PTEN Fosfo-Hidrolase/genética , Proteínas Serina-Treonina Quinases/genética , Quinases Proteína-Quinases Ativadas por AMP , Feminino , Humanos , Neoplasias Ovarianas/patologia
5.
Hum Mol Genet ; 21(20): 4394-405, 2012 Oct 15.
Artigo em Inglês | MEDLINE | ID: mdl-22791749

RESUMO

Male patients with Peutz-Jeghers syndrome (PJS) have defective spermatogenesis and are at increased risk of developing Sertoli cell tumors. Mutations in the Liver Kinase B1 (LKB1/STK11) gene are associated with the pathogenesis of PJS and have been identified in non-PJS patients with sporadic testicular cancers. The mechanisms controlled by LKB1 signaling in Sertoli cell functions and testicular biology have not been described. We have conditionally deleted the Lkb1 gene (Lkb1(cko)) in somatic testicular cells to define the molecular mechanisms involved in the development of the testicular phenotype observed in PJS patients. Focal vacuolization in some of the seminiferous tubules was observed in 4-week-old mutant testes but germ cell development appeared to be normal. However, similar to PJS patients, we observed progressive germ cell loss and Sertoli cell only tubules in Lkb1(cko) testes from mice older than 10 weeks, accompanied by defects in Sertoli cell polarity and testicular junctional complexes and decreased activation of the MAP/microtubule affinity regulating and focal adhesion kinases. Suppression of AMP kinase and activation of mammalian target of rapamycin (mTOR) signaling were also observed in Lkb1(cko) testes. Loss of Tsc1 or Tsc2 copies the progressive Lkb1(cko) phenotype, suggesting that dysregulated activation of mTOR contributes to the pathogenesis of the Lkb1(cko) testicular phenotype. Pten(cko) mice had a normal testicular phenotype, which could be explained by the comparative lack of mTOR activation detected. These studies describe the importance of LKB1 signaling in testicular biology and the possible molecular mechanisms driving the pathogenesis of the testicular defects observed in PJS patients.


Assuntos
Proteínas Quinases Ativadas por AMP/genética , Polaridade Celular/genética , Proteínas Serina-Treonina Quinases/genética , Células de Sertoli/metabolismo , Transdução de Sinais , Espermatogênese/genética , Serina-Treonina Quinases TOR/genética , Proteínas Supressoras de Tumor/genética , Proteínas Quinases Ativadas por AMP/metabolismo , Animais , Masculino , Camundongos , Proteínas Serina-Treonina Quinases/metabolismo , Serina-Treonina Quinases TOR/metabolismo , Proteína 1 do Complexo Esclerose Tuberosa , Proteínas Supressoras de Tumor/metabolismo
6.
Mol Hum Reprod ; 20(11): 1126-34, 2014 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-25189766

RESUMO

Uterine leiomyomata are the most common tumors found in the female reproductive tract. Despite the high prevalence and associated morbidities of these benign tumors, little is known about the molecular basis of uterine leiomyoma development and progression. Loss of the Tuberous Sclerosis 2 (TSC2) tumor suppressor has been proposed as a mechanism important for the etiology of uterine leiomyomata based on the Eker rat model. However, conflicting evidence showing increased TSC2 expression has been reported in human uterine leiomyomata, suggesting that TSC2 might not be involved in the pathogenesis of this disorder. We have produced mice with conditional deletion of the Tsc2 gene in the myometria to determine whether loss of TSC2 leads to leiomyoma development in murine uteri. Myometrial hyperplasia and increased collagen deposition was observed in Tsc2(cKO) mice compared with control mice, but no leiomyomata were detected by post-natal week 24. Increased signaling activity of mammalian target of rapamycin complex 1, which is normally repressed by TSC2, was also detected in the myometria of Tsc2(cKO) mice. Treatment of the mutant mice with rapamycin significantly inhibited myometrial expansion, but treatment with the progesterone receptor modulator, mifepristone, did not. The ovaries of the Tsc2(cKO) mice appeared normal, but half the mice were infertile and most of the other half became infertile after a single litter, which was likely due to oviductal blockage. Our study shows that although TSC2 loss alone does not lead to leiomyoma development, it does lead to myometrial hyperplasia and fibrosis.


Assuntos
Ductos Paramesonéfricos/patologia , Miométrio/patologia , Proteínas Supressoras de Tumor/genética , Animais , Feminino , Fertilidade/genética , Fibrose/genética , Deleção de Genes , Hiperplasia/genética , Leiomioma/genética , Leiomioma/patologia , Alvo Mecanístico do Complexo 1 de Rapamicina , Mesoderma/patologia , Camundongos , Complexos Multiproteicos/metabolismo , Sirolimo/farmacologia , Serina-Treonina Quinases TOR/metabolismo , Proteína 2 do Complexo Esclerose Tuberosa
7.
Carcinogenesis ; 34(4): 893-901, 2013 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-23276799

RESUMO

Epithelial ovarian cancer is a heterogeneous disease that is subdivided into five major histotypes but the mechanisms driving their differentiation are not clear. Mutations in adenomatous polyposis coli (APC) and ß-catenin are commonly observed in the human ovarian endometrioid adenocarcinoma (OEA) patients. However, the mechanisms subsequent to APC deletion in ovarian tumorigenesis have not been well characterized. We have conditionally deleted APC in the murine ovarian surface epithelium (OSE) and showed that its loss leads to development of epithelial inclusion cysts. High-grade OEAs with tightly packed villoglandular histology were observed in older APC-deleted mice. Phosphatase and tensin homolog (PTEN) expression was elevated in the early lesions but lost after progression to the more advanced tumors. Knockdown of APC or expression of a gain-of-function ß-catenin similarly induced human OSE cells to develop tumors with endometrioid histology in xenografts. Expression of HOXA10 was induced in both the advanced APC-deleted murine tumors and in the tumor xenografts of human OSE cells with knocked-down APC. These results show that reduced APC activity is sufficient to induce formation of epithelial inclusion cysts and support OEA development and suggest that induced HOXA10 expression and loss of PTEN are key mechanisms driving endometrioid histotype differentiation and progression.


Assuntos
Polipose Adenomatosa do Colo/genética , Carcinoma Endometrioide/genética , Carcinoma Endometrioide/patologia , Proteínas de Homeodomínio/genética , Neoplasias Epiteliais e Glandulares/genética , Neoplasias Ovarianas/genética , PTEN Fosfo-Hidrolase/genética , Adenocarcinoma/genética , Animais , Carcinoma Endometrioide/metabolismo , Carcinoma Epitelial do Ovário , Diferenciação Celular , Transformação Celular Neoplásica/genética , Progressão da Doença , Feminino , Proteínas Homeobox A10 , Humanos , Camundongos , Camundongos Knockout , Transplante de Neoplasias , Neoplasias Epiteliais e Glandulares/patologia , Neoplasias Ovarianas/patologia , Ovário/patologia , Transplante Heterólogo , Células Tumorais Cultivadas , Via de Sinalização Wnt/genética , beta Catenina/genética
8.
Sci Rep ; 12(1): 19368, 2022 11 12.
Artigo em Inglês | MEDLINE | ID: mdl-36371454

RESUMO

Placentas of obese women have low mitochondrial ß-oxidation of fatty acids (FA) and accumulate lipids in late pregnancy. This creates a lipotoxic environment, impairing placental efficiency. We hypothesized that placental FA metabolism is impaired in women with obesity from early pregnancy. We assessed expression of key regulators of FA metabolism in first trimester placentas of lean and obese women. Maternal fasting triglyceride and insulin levels were measured in plasma collected at the time of procedure. Expression of genes associated with FA oxidation (FAO; ACOX1, CPT2, AMPKα), FA uptake (LPL, LIPG, MFSD2A), FA synthesis (ACACA) and storage (PLIN2) were significantly reduced in placentas of obese compared to lean women. This effect was exacerbated in placentas of male fetuses. Placental ACOX1 protein was higher in women with obesity and correlated with maternal circulating triglycerides. The PPARα pathway was enriched for placental genes impacted by obesity, and PPARα antagonism significantly reduced 3H-palmitate oxidation in 1st trimester placental explants. These results demonstrate that obesity and hyperlipidemia impact placental FA metabolism as early as 7 weeks of pregnancy.


Assuntos
Metabolismo dos Lipídeos , Placenta , Gravidez , Feminino , Masculino , Humanos , Placenta/metabolismo , Metabolismo dos Lipídeos/genética , Primeiro Trimestre da Gravidez , PPAR alfa/genética , PPAR alfa/metabolismo , Obesidade/metabolismo , Ácidos Graxos/metabolismo , Triglicerídeos/metabolismo
9.
J Clin Endocrinol Metab ; 106(12): 3526-3535, 2021 11 19.
Artigo em Inglês | MEDLINE | ID: mdl-34333643

RESUMO

CONTEXT: An increase in maternal insulin resistance (IR) during pregnancy is essential for normal fetal growth. The mechanisms underlying this adaptation are poorly understood. Placental factors are believed to instigate and maintain these changes, as IR decreases shortly after delivery. Methylation of placental gene loci that are common targets for miRNAs are associated with maternal IR. OBJECTIVE: We hypothesized that placental miRNAs targeting methylated loci are associated with maternal IR during late pregnancy. METHODS: We collected placentas from 132 elective cesarean sections and fasting blood samples at delivery to estimate maternal homeostasis model assessment of insulin resistance (HOMA-IR). Placental miRNA expression was measured via whole genome small-RNA sequencing in a subset of 40 placentas selected by maternal pre-gravid body mass index (BMI) and neonatal adiposity. Five miRNAs correlated with maternal HOMA-IR and previously identified as targeting methylated genes were selected for validation in all 132 placenta samples via RT-qPCR. Multiple regression adjusted for relevant clinical variables. RESULTS: Median maternal age was 27.5 years, with median pre-pregnancy BMI of 24.7 kg/m2, and median HOMA-IR of 2.9. Among the 5 selected miRNA, maternal HOMA-IR correlated with the placental expression of miRNA-371b-3p (r = 0.25; P = 0.008) and miRNA-3940-3p (r = 0.32; P = 0.0004) across the 132 individuals. After adjustment for confounding variables, placental miRNA-3940-3p expression remained significantly associated with HOMA-IR (ß = 0.16; P = 0.03). CONCLUSION: Placental miRNA-3940-3p was associated with maternal IR at delivery. This placental miRNA may have an autocrine or paracrine effect-regulating placental genes involved in modulating maternal IR.


Assuntos
Biomarcadores/metabolismo , Índice de Massa Corporal , Redes Reguladoras de Genes , Resistência à Insulina , MicroRNAs/genética , Placenta/metabolismo , Trofoblastos/metabolismo , Adulto , Feminino , Seguimentos , Perfilação da Expressão Gênica , Humanos , Gravidez , Prognóstico
10.
Biol Reprod ; 82(2): 422-32, 2010 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-19794154

RESUMO

Sertoli and germ cell interactions are essential for spermatogenesis and, thus, male fertility. Sertoli cells provide a specialized microenvironment for spermatogonial stem cells to divide, allowing both self-renewal and spermatogenesis. In the present study, we used mice with a conditional activated allele of the beta-catenin gene (Ctnnb1(tm1Mmt)(/+)) in Sertoli cells expressing Cre recombinase driven by the anti-Müllerian hormone (AMH; also known as Müllerian-inhibiting substance) type II receptor promoter (Amhr2(tm3(cre)Bhr)(/+)) to show that constitutively activated beta-catenin leads to their continuous proliferation and compromised differentiation. Compared to controls, Sertoli cells in mature mutant mice continue to express high levels of both AMH and glial cell-derived neurotrophic factor (GDNF), which normally are expressed only in immature Sertoli cells. We also show evidence that LiCl treatment, which activates endogenous nuclear beta-catenin activity, regulates both AMH and GDNF expression at the transcriptional level. The epididymides were devoid of sperm in the Amhr2(tm3(cre)Bhr)(/+);Ctnnb1(tm1Mmt)(/+) mice at all ages examined. We show that the mutant mice are infertile because of defective differentiation of germ cells and increased apoptosis, both of which are characteristic of GDNF overexpression in Sertoli cells. Constitutive activation of beta-catenin in Amhr2-null mice showed the same histology, suggesting that the phenotype was the result of persistent overexpression of GDNF. These results show that dysregulated wingless-related MMTV integration site/beta-catenin signaling in Sertoli cells inhibits their postnatal differentiation, resulting in increased germ cell apoptosis and infertility.


Assuntos
Diferenciação Celular/fisiologia , Células de Sertoli/citologia , Transdução de Sinais/fisiologia , Espermatogênese/fisiologia , Proteínas Wnt/fisiologia , beta Catenina/fisiologia , Animais , Hormônio Antimülleriano/genética , Apoptose/genética , Diferenciação Celular/genética , Expressão Gênica , Fator Neurotrófico Derivado de Linhagem de Célula Glial/genética , Infertilidade Masculina/genética , Integrases/genética , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Mutação , Regiões Promotoras Genéticas/genética , Receptores de Peptídeos/genética , Receptores de Fatores de Crescimento Transformadores beta/genética , Células de Sertoli/química , Células de Sertoli/fisiologia , Testículo/embriologia , Testículo/crescimento & desenvolvimento , beta Catenina/análise , beta Catenina/genética
11.
Front Pediatr ; 8: 618553, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-33575231

RESUMO

Non-invasive techniques to monitor and diagnose neonates, particularly those born prematurely, are a long-sought out goal of Newborn Medicine. In recent years, technical advances, combined with increased assay sensitivity, have permitted the high-throughput analysis of multiple biomarkers simultaneously from a single sample source. Multiplexed transcriptomic and proteomic platforms, along with more comprehensive assays such as RNASeq, allow for interrogation of ongoing physiology and pathology in unprecedented ways. In the fragile neonatal population, saliva is an ideal biofluid to assess clinical status serially and offers many advantages over more invasively obtained blood samples. Importantly, saliva samples are amenable to analysis on emerging proteomic and transcriptomic platforms, even at quantitatively limited volumes. However, biomarker targets are often degraded in human saliva, and as a mixed source biofluid containing both human and microbial targets, saliva presents unique challenges for the investigator. Here, we provide insight into technical considerations and protocol optimizations developed in our laboratory to quantify and discover neonatal salivary biomarkers with improved reproducibility and reliability. We will detail insights learned from years of experimentation on neonatal saliva within our laboratory ranging from salivary collection techniques to processing to downstream analyses, highlighting the need for consistency in approach and a global understanding of both the potential benefits and limitations of neonatal salivary biomarker analyses. Importantly, we will highlight the need for robust and stringent research in this population to provide the field with standardized approaches and workflows to impact neonatal care successfully.

12.
Am J Speech Lang Pathol ; 29(2S): 1022-1029, 2020 07 10.
Artigo em Inglês | MEDLINE | ID: mdl-32650666

RESUMO

Purpose Successful oral feeding and speech emergence are dependent upon the coordination of shared oral muscles and facial nerves. We aimed to determine if the speech-associated genes, forkhead box P2 (FOXP2), contactin-associated protein-like 2 (CNTNAP2), glutamate receptor, ionotropic, N-methyl D-aspartate 2A (GRIN2A), and neurexin 1, were detectable in neonatal saliva and could predict feeding outcomes in premature newborns. Method In this prospective, observational, preliminary study, saliva collected from 51 premature infants (gestational ages: 30-34 6/7 weeks) at different stages of oral feeding development underwent gene expression analysis. Binary (+/-) expression profiles were explored and examined in relation to days to achieve full oral feeds. Results GRIN2A and neurexin 1 rarely amplified in neonatal saliva and were not informative. Infants who amplified FOXP2 but not CNTNAP2 at the start of oral feeds achieved oral feeding success 3.20 (95% CI [-2.5, 8.9]) days sooner than other gene combinations. Conclusions FOXP2 and CNTNAP2 may be informative in predicting oral feeding outcomes in newborns. Salivary analysis at the start of oral feeding trials may inform feeding outcomes in this population and warrants further investigation.


Assuntos
Biomarcadores , Recém-Nascido Prematuro , Fala , Ingestão de Alimentos , Idade Gestacional , Humanos , Lactente , Transtornos da Nutrição do Lactente , Recém-Nascido , Estudos Prospectivos
14.
Endocrinology ; 153(1): 404-16, 2012 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-22128018

RESUMO

Tumors develop with dysregulated activation of mammalian target of rapamycin (mTOR), the kinase activity of which is kept in an inactive state by a tumor suppressor dimer containing tuberous sclerosis 1 (TSC1) and TSC2. We examined whether conditional deletion of TSC1 by a knock-in allele of the anti-Müllerian hormone type 2 receptor (Amhr2) driving Cre expression and subsequent activation of mTOR in granulosa cells and in oviductal and uterine stromal cells affects fertility in female mice. Increased phosphorylation of ribosomal protein S6, a downstream target of activated mTOR, was observed in all AMHR2-expressing tissues examined, indicating loss of TSC1 activity. TSC1 deletion in granulosa cells led to the detection of significantly fewer primordial follicles in mutant mice at 12 wk, suggesting premature ovarian insufficiency, which might be related to the significantly increased time mutant mice spent in estrus. Although the number of good-quality ovulated oocytes was not significantly different compared with controls, there was a significantly higher number of degenerated oocytes after normal and superovulation, suggesting compromised oocyte quality, as well. Natural mating also showed severalfold higher numbers of degenerate bodies in the mutants that collected in bilateral swellings resembling hydrosalpinges that formed in all mice examined because of occlusion of the proximal oviduct. Attempts to transfer control embryos into mutant uteri also failed, indicating that implantation was compromised. Endometrial epithelial cells continued to proliferate, and quantitative RT-PCR showed that mucin 1 expression persisted during the window of implantation in mutant uteri, without any changes in progesterone receptor mRNA expression, suggesting a mechanism that does not involve disrupted estradiol-regulated progesterone receptor expression. Homozygous deletion of TSC1 in reproductive tract somatic tissues of mice rendered females completely infertile, which is likely due to these pleiotropic effects on follicle recruitment, oviductal development, and blastocyst implantation.


Assuntos
Infertilidade Feminina/genética , Proteínas Supressoras de Tumor/deficiência , Animais , Sequência de Bases , Primers do DNA/genética , Implantação do Embrião/genética , Implantação do Embrião/fisiologia , Endométrio/fisiopatologia , Feminino , Técnicas de Introdução de Genes , Infertilidade Feminina/patologia , Infertilidade Feminina/fisiopatologia , Masculino , Camundongos , Camundongos da Linhagem 129 , Camundongos Endogâmicos C57BL , Camundongos Knockout , Camundongos Transgênicos , Ductos Paramesonéfricos/patologia , Oócitos/patologia , Oócitos/fisiologia , Ovário/patologia , Ovário/fisiopatologia , Gravidez , Receptores de Peptídeos/genética , Receptores de Peptídeos/fisiologia , Receptores de Fatores de Crescimento Transformadores beta/genética , Receptores de Fatores de Crescimento Transformadores beta/fisiologia , Serina-Treonina Quinases TOR/fisiologia , Proteína 1 do Complexo Esclerose Tuberosa , Proteínas Supressoras de Tumor/genética , Proteínas Supressoras de Tumor/fisiologia
15.
PLoS One ; 6(6): e20715, 2011.
Artigo em Inglês | MEDLINE | ID: mdl-21695255

RESUMO

Despite the fact that epithelial ovarian cancers are the leading cause of death from gynecological cancer, very little is known about the pathophysiology of the disease. Mutations in the WNT and PI3K pathways are frequently observed in the human ovarian endometrioid adenocarcinomas (OEAs). However, the role of WNT/ß-catenin and PTEN/AKT signaling in the etiology and/or progression of this disease is currently unclear. In this report we show that mice with a gain-of-function mutation in ß-catenin that leads to dysregulated nuclear accumulation of ß-catenin expression in the ovarian surface epithelium (OSE) cells develop indolent, undifferentiated tumors with both mesenchymal and epithelial characteristics. Combining dysregulated ß-catenin with homozygous deletion of PTEN in the OSE resulted in development of significantly more aggressive tumors, which was correlated with inhibition of p53 expression and cellular senescence. Induced expression of both mTOR kinase, a master regulator of proliferation, and phosphorylation of its downstream target, S6Kinase was also observed in both the indolent and aggressive mouse tumors, as well as in human OEA with nuclear ß-catenin accumulation. Ectopic allotransplants of the mouse ovarian tumor cells with a gain-of-function mutation in ß-catenin and PTEN deletion developed into tumors with OEA histology, the growth of which were significantly inhibited by oral rapamycin treatment. These studies demonstrate that rapamycin might be an effective therapeutic for human ovarian endometrioid patients with dysregulated Wnt/ß-catenin and Pten/PI3K signaling.


Assuntos
Carcinoma Endometrioide/enzimologia , Neoplasias Ovarianas/enzimologia , PTEN Fosfo-Hidrolase/metabolismo , Serina-Treonina Quinases TOR/metabolismo , Proteínas Wnt/metabolismo , beta Catenina/metabolismo , Animais , Carcinoma Endometrioide/patologia , Morte Celular/efeitos dos fármacos , Proliferação de Células/efeitos dos fármacos , Senescência Celular/efeitos dos fármacos , Feminino , Deleção de Genes , Humanos , Camundongos , Camundongos Endogâmicos C57BL , Transplante de Neoplasias , Neoplasias Ovarianas/patologia , Transdução de Sinais/efeitos dos fármacos , Sirolimo/farmacologia , Carga Tumoral/efeitos dos fármacos , Proteína Supressora de Tumor p53/metabolismo
16.
Reprod Sci ; 16(4): 347-56, 2009 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-19087973

RESUMO

Activation of the aryl hydrocarbon receptor (AHR) by polycyclic aromatic hydrocarbons (PAH), a ubiquitous class of environmental and occupational biohazards, accelerates germ cell depletion in female mice during prenatal and postnatal life. Like AHR, BAX is also functionally required for PAH to kill oocytes. Here, we show that PAH upregulates ovarian expression of not just Bax but a large cassette of proapoptotic genes that function at multiple steps of the cell death signaling pathway. We further show that ovarian expression of p53 and several proapoptotic genes that are known transcriptional targets of p53 are increased by PAH treatment, and that mice lacking functional p53 are resistant to the ovotoxic effects of in vivo PAH exposure. This study provides further mechanistic insights into how PAH accelerate oocyte depletion in females and adds p53 to the list of genes whose functional importance to PAH-induced ovotoxicity has been demonstrated by gene knockout technology.


Assuntos
Apoptose/genética , Expressão Gênica/efeitos dos fármacos , Folículo Ovariano/efeitos dos fármacos , Ovário/efeitos dos fármacos , Hidrocarbonetos Policíclicos Aromáticos/toxicidade , Proteína Supressora de Tumor p53/genética , Animais , Northern Blotting , DNA/análise , Feminino , Perfilação da Expressão Gênica , Imuno-Histoquímica , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Mutação , Oócitos/efeitos dos fármacos , Oócitos/fisiologia , Ovário/química , Ovário/metabolismo , Receptores do Fator de Necrose Tumoral , Transdução de Sinais/fisiologia , Proteína Supressora de Tumor p53/deficiência , Proteína Supressora de Tumor p53/fisiologia , Proteína X Associada a bcl-2/genética
17.
Biol Reprod ; 77(4): 658-65, 2007 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-17582011

RESUMO

During early gestation in invasively implanting species, the uterine stromal compartment undergoes dramatic remodeling, defined by the differentiation of stromal fibroblast cells into decidual cells. Lipid signaling molecules from a number of pathways are well-established functional components of this decidualization reaction. Because of a correlation in the events that transpire in the uterus during early implantation with known functions of bioactive sphingolipid metabolites established from studies in other organ systems, we hypothesized that uterine sphingolipid metabolism would change during implantation. By a combination of Northern blot, Western blot, and immunohistochemical analyses, we establish that enzymes at each of the major catalytic steps in the sphingolipid cascade become transcriptionally up-regulated in the uterus during decidualization. Each of the enzymes analyzed was up-regulated from Days of Pregnancy (DOP) 4.5-7.5. When comparing embryo-induced decidualization (decidual) with mechanically induced decidualization (deciduomal), sphingomyelin phosphodiesterase 1 (Smpd1) mRNA and sphingosine kinase 1 (SPHK1) protein were shown to be dually regulated in the endometrium by both maternal and embryonic factors. As measured by the diacyl glycerol kinase assay, ceramide levels rose in parallel with Smpd1 gene expression, suggesting that elevated transcription of sphingolipid enzymes results in heightened catalytic activity of the pathway. Altogether, these findings place sphingolipids on a growing list of lipid signaling molecules that become increasingly present at the maternal-embryonic interface.


Assuntos
Decídua/enzimologia , Implantação do Embrião/genética , Regulação da Expressão Gênica no Desenvolvimento , Regulação Enzimológica da Expressão Gênica , Fosfotransferases (Aceptor do Grupo Álcool)/genética , Esfingolipídeos/metabolismo , Esfingomielina Fosfodiesterase/genética , Animais , Feminino , Camundongos , Fosfotransferases (Aceptor do Grupo Álcool)/análise , Fosfotransferases (Aceptor do Grupo Álcool)/metabolismo , Gravidez , Esfingomielina Fosfodiesterase/análise , Esfingomielina Fosfodiesterase/metabolismo , Células Estromais/enzimologia , Regulação para Cima , Útero/enzimologia
18.
Differentiation ; 75(2): 93-9, 2007 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-17316379

RESUMO

A recent commentary in this journal by Byskov et al. (2005) claims that, despite published results from numerous independent lines of investigation from our laboratory and others, there does not "exist any evidence for neo-folliculogenesis in the adult mammalian ovary." While we agree with Byskov et al. that our work represents a radical departure from the age-old dogma that mammalian females permanently lose the capacity for oocyte and follicle production during the perinatal period, careful examination of all of the available data leaves no doubt that adult female mammals retain the capacity for oogenesis and folliculogenesis. These findings do not change the fact that exhaustion of the oocyte pool occurs with advancing chronological age--a process responsible for driving the menopause in women--but rather question the basic mechanism underlying age-related ovarian failure. In this regard, studies of aging male mice have demonstrated that testicular atrophy is associated with a dramatic decline in the number, activity and quality of germline stem cells that maintain spermatogenesis during adulthood (Zhang et al., 2006). Therefore, to the contrary of the opinion of Byskov et al. that such a process would be "considered exceptional among stem cells," it is certainly reasonable to hypothesize that a similar deterioration of female germline stem cell function underlies the decline in oocyte quality and the onset of ovarian failure in aging females. Further, while we accept that a departure from conventional thinking can take years to gain widespread acceptance, we feel this resistance to change should not be construed as the sole means to voice opinions about the validity of our data or the maturity of our principal conclusion.


Assuntos
Diferenciação Celular , Oócitos/citologia , Oogênese/fisiologia , Óvulo/citologia , Envelhecimento/fisiologia , Animais , Feminino , Células Germinativas/fisiologia , Camundongos , Ovário/anatomia & histologia , Células-Tronco/fisiologia
19.
Biol Reprod ; 74(3): 569-76, 2006 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-16319286

RESUMO

Signaling mechanisms coordinating uterine angiogenesis and tissue remodeling during decidualization are not completely understood. Prostanoid signaling is thought to play a functionally important role in each of these events. In the present study, we demonstrate that the subfamily of G-protein-coupled receptors that binds and becomes activated by the terminal signaling lipid in the sphingolipid pathway, sphingosine-1-phosphate (S1P), were expressed during uterine decidualization. Three of the five known S1P receptors, termed endothelial differentiation genes (Edg; Edg1, Edg3, and Edg5) were upregulated in the uterine deciduum from Day of Pregnancy (DOP) 4.5 to 7.5, while Edg6 and Edg8 expression remained unchanged. Consistent with angiogenesis in general during decidualization, we believe EDG1 and EDG5 to be regulated by the embryo because no microvascular expression for these receptors was observed in oil-induced deciduomas. Observed expression of EDG1 and EDG5 showed a similar expression pattern to that previously reported for prostaglandin-endoperoxide synthase 2 (PTGS2), transitioning from the sublumenal stromal compartment in the antimesometrial pole (DOP 5) to the microvasculature of the mesometrial pole (DOP 7). Furthermore, these two receptors colocalized with PTGS2 at three additional sites at the maternal:fetal interface throughout pregnancy. Treatment of cultured predecidualized stromal cells with S1P resulted in upregulation of Ptgs2 mRNA and PTGS2 protein, but not the downstream enzyme prostacyclin synthase. These combined results suggest the existence of a link between the sphingolipid and prostanoid signaling pathways in uterine physiology, and that, based on their expression pattern, S1P receptors function to coordinate uterine mesometrial angiogenesis during the implantation phase of early gestation.


Assuntos
Ciclo-Oxigenase 2/genética , Decídua/fisiologia , Neovascularização Fisiológica/fisiologia , Prenhez/fisiologia , Receptores de Lisoesfingolipídeo/genética , Receptores de Lisoesfingolipídeo/metabolismo , Animais , Ciclo-Oxigenase 2/metabolismo , Decídua/irrigação sanguínea , Decídua/citologia , Feminino , Regulação da Expressão Gênica no Desenvolvimento , Masculino , Camundongos , Camundongos Endogâmicos ICR , Gravidez , RNA Mensageiro/metabolismo , Transdução de Sinais/fisiologia , Células Estromais/fisiologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA