Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 39
Filtrar
Mais filtros

Base de dados
Tipo de documento
Intervalo de ano de publicação
1.
J Mammary Gland Biol Neoplasia ; 26(3): 263-276, 2021 09.
Artigo em Inglês | MEDLINE | ID: mdl-34617201

RESUMO

We previously showed that dietary trans-10, cis-12 conjugated linoleic acid (10,12 CLA) stimulates estrogen-independent mammary growth in young ovariectomized mice. Here we investigated the effects of in utero or postnatal exposure to cis-9, trans-11 (9,11 CLA) and 10,12 CLA on postnatal development of the mammary gland and its responsiveness to ovarian steroids. In the first experiment we fed dams different CLA prior to and during gestation, then cross fostered female pups onto control fed dams prior to assessing the histomorphology of their mammary glands. Pregnant dams in the second experiment were similarly exposed to CLA, after which their female pups were ovariectomized then treated with 17ß-estradiol (E), progesterone (P) or E + P for 5 days. In a third experiment, mature female mice were fed different CLA for 28 days prior to ovariectomy, then treated with E, P or E + P. Our data indicate that 10,12 CLA modifies the responsiveness of the mammary glands to E or E + P when exposure occurs either in utero, or postnatally. These findings underline the sensitivity of the mammary glands to dietary fatty acids and reinforce the potential for maternal nutrition to impact postnatal development of the mammary glands and their risk for developing cancer.


Assuntos
Gorduras na Dieta/efeitos adversos , Ácidos Linoleicos Conjugados/efeitos adversos , Glândulas Mamárias Animais/crescimento & desenvolvimento , Exposição Materna/efeitos adversos , Efeitos Tardios da Exposição Pré-Natal/etiologia , Animais , Biomarcadores/metabolismo , Estrogênios/metabolismo , Feminino , Glândulas Mamárias Animais/metabolismo , Camundongos , Camundongos Endogâmicos BALB C , Gravidez , Efeitos Tardios da Exposição Pré-Natal/metabolismo , Progesterona/metabolismo
2.
BMC Genomics ; 22(1): 118, 2021 Feb 12.
Artigo em Inglês | MEDLINE | ID: mdl-33581720

RESUMO

BACKGROUND: The homologous recombination (HR) pathway is largely inactive in early embryos prior to the first cell division, making it difficult to achieve targeted gene knock-ins. The homology-mediated end joining (HMEJ)-based strategy has been shown to increase knock-in efficiency relative to HR, non-homologous end joining (NHEJ), and microhomology-mediated end joining (MMEJ) strategies in non-dividing cells. RESULTS: By introducing gRNA/Cas9 ribonucleoprotein complex and a HMEJ-based donor template with 1 kb homology arms flanked by the H11 safe harbor locus gRNA target site, knock-in rates of 40% of a 5.1 kb bovine sex-determining region Y (SRY)-green fluorescent protein (GFP) template were achieved in Bos taurus zygotes. Embryos that developed to the blastocyst stage were screened for GFP, and nine were transferred to recipient cows resulting in a live phenotypically normal bull calf. Genomic analyses revealed no wildtype sequence at the H11 target site, but rather a 26 bp insertion allele, and a complex 38 kb knock-in allele with seven copies of the SRY-GFP template and a single copy of the donor plasmid backbone. An additional minor 18 kb allele was detected that looks to be a derivative of the 38 kb allele resulting from the deletion of an inverted repeat of four copies of the SRY-GFP template. CONCLUSION: The allelic heterogeneity in this biallelic knock-in calf appears to have resulted from a combination of homology directed repair, homology independent targeted insertion by blunt-end ligation, NHEJ, and rearrangement following editing of the gRNA target site in the donor template. This study illustrates the potential to produce targeted gene knock-in animals by direct cytoplasmic injection of bovine embryos with gRNA/Cas9, although further optimization is required to ensure a precise single-copy gene integration event.


Assuntos
Sistemas CRISPR-Cas , Zigoto , Animais , Bovinos/genética , Repetições Palindrômicas Curtas Agrupadas e Regularmente Espaçadas , Reparo do DNA por Junção de Extremidades , Feminino , Edição de Genes , Técnicas de Introdução de Genes , Masculino
3.
J Nutr ; 150(8): 2120-2130, 2020 08 01.
Artigo em Inglês | MEDLINE | ID: mdl-32510141

RESUMO

BACKGROUND: Folate is essential for DNA synthesis, DNA repair, cell proliferation, development, and morphogenesis. Folic acid (FA) is a nutritional supplement used to fortify human diets. OBJECTIVES: We investigated the effects of dietary FA on early mammary gland (MG) development and hyperplasia. METHODS: Study 1: nulliparous female FVB wild-type (WT) mice were fed control (Con; 2 mg FA/kg), deficient (Def; 0 mg FA/kg), excess (Ex; 5 mg FA/kg), or super excess (S-Ex; 20 mg FA/kg) diets for 8 wk before mating to WT or heterozygous FVB/N-Tg[mouse mammary tumor virus long terminal repeat (MMTV)-polyomavirus middle T antigen (PyVT)]634Mul/J (MMTV-PyMT+/-) transgenic males. Dams were fed these diets until they weaned WT or MMTV-PyMT+/- pups, which were fed the dam's diet from postnatal day (PND) 21 to 42. Tissues were collected from female progeny at PNDs 1, 21, and 42. Study 2: Con or Def diets were fed to WT intact females and males from PND 21 to 56, or to ovariectomized females from PND 21 to 77; tissues were collected at PND 56 or 77. Growth of all offspring, development of MGs, MG hyperplasia, supramammary lymph nodes, thymus and spleen, cell proliferation, and expression of MG growth factors were measured. RESULTS: Study 1: Ex or S-Ex did not affect postnatal MG development or hyperplasia. The rate of isometric MG growth (PND 1-21) was reduced by 69% in Def female progeny (P < 0.0001). Similarly, hyperplastic growth in MGs of Def MMTV-PyMT+/- offspring was 18% of Con (P < 0.05). The Def diet reduced supramammary lymph node size by 20% (P < 0.0001) and increased MG insulin-like growth factor 2 mRNA by 200% (P < 0.05) and protein by 130%-150% (P < 0.05). Study 2: the Def diet did not affect MG growth, but it did reduce supramammary lymph node size (P < 0.05), spleen weight (P < 0.001), and thymic medulla area (P < 0.05). CONCLUSIONS: In utero and postnatal folate deficiency reduced the isometric development of the MGs and early MG hyperplasia. Postnatal folate deficiency reduced the development of lymphatic tissues.


Assuntos
Deficiência de Ácido Fólico , Ácido Fólico/administração & dosagem , Linfonodos/efeitos dos fármacos , Linfonodos/crescimento & desenvolvimento , Glândulas Mamárias Animais/efeitos dos fármacos , Glândulas Mamárias Animais/crescimento & desenvolvimento , Animais , Dieta , Feminino , Masculino , Camundongos , Ovariectomia
4.
BMC Nephrol ; 20(1): 66, 2019 02 25.
Artigo em Inglês | MEDLINE | ID: mdl-30803434

RESUMO

BACKGROUND: Autosomal dominant polycystic kidney disease (ADPKD) is the most common hereditary kidney disease and is characterized by gradual cyst growth and expansion, increase in kidney volume with an ultimate decline in kidney function leading to end stage renal disease (ESRD). Given the decades long period of stable kidney function while cyst growth occurs, it is important to identify those patients who will progress to ESRD. Recent data from our and other laboratories have demonstrated that metabolic reprogramming may play a key role in cystic epithelial proliferation resulting in cyst growth in ADPKD. Height corrected total kidney volume (ht-TKV) accurately reflects cyst burden and predicts future loss of kidney function. We hypothesize that specific plasma metabolites will correlate with eGFR and ht-TKV early in ADPKD, both predictors of disease progression, potentially indicative of early physiologic derangements of renal disease severity. METHODS: To investigate the predictive role of plasma metabolites on eGFR and/or ht-TKV, we used a non-targeted GC-TOF/MS-based metabolomics approach on hypertensive ADPKD patients in the early course of their disease. Patient data was obtained from the HALT-A randomized clinical trial at baseline including estimated glomerular filtration rate (eGFR) and measured ht-TKV. To identify individual metabolites whose intensities are significantly correlated with eGFR and ht-TKV, association analyses were performed using linear regression with each metabolite signal level as the primary predictor variable and baseline eGFR and ht-TKV as the continuous outcomes of interest, while adjusting for covariates. Significance was determined by Storey's false discovery rate (FDR) q-values to correct for multiple testing. RESULTS: Twelve metabolites significantly correlated with eGFR and two triglycerides significantly correlated with baseline ht-TKV at FDR q-value < 0.05. Specific significant metabolites, including pseudo-uridine, indole-3-lactate, uric acid, isothreonic acid, and creatinine, have been previously shown to accumulate in plasma and/or urine in both diabetic and cystic renal diseases with advanced renal insufficiency. CONCLUSIONS: This study identifies metabolic derangements in early ADPKD which may be prognostic for ADPKD disease progression. CLINICAL TRIAL: HALT Progression of Polycystic Kidney Disease (HALT PKD) Study A; Clinical www.clinicaltrials.gov identifier: NCT00283686; first posted January 30, 2006, last update posted March 19, 2015.


Assuntos
Rim , Rim Policístico Autossômico Dominante , Insuficiência Renal , Adulto , Creatinina/sangue , Progressão da Doença , Feminino , Humanos , Indóis/sangue , Rim/metabolismo , Rim/patologia , Testes de Função Renal/métodos , Testes de Função Renal/estatística & dados numéricos , Estudos Longitudinais , Masculino , Tamanho do Órgão , Gravidade do Paciente , Rim Policístico Autossômico Dominante/sangue , Rim Policístico Autossômico Dominante/diagnóstico , Rim Policístico Autossômico Dominante/fisiopatologia , Valor Preditivo dos Testes , Prognóstico , Pseudouridina/sangue , Insuficiência Renal/sangue , Insuficiência Renal/diagnóstico , Insuficiência Renal/etiologia , Ácido Úrico/sangue
5.
Am J Physiol Renal Physiol ; 315(6): F1855-F1868, 2018 12 01.
Artigo em Inglês | MEDLINE | ID: mdl-30280600

RESUMO

Research into metabolic reprogramming in cancer has become commonplace, yet this area of research has only recently come of age in nephrology. In light of the parallels between cancer and autosomal dominant polycystic kidney disease (ADPKD), the latter is currently being studied as a metabolic disease. In clear cell renal cell carcinoma (RCC), which is now considered a metabolic disease, we and others have shown derangements in the enzyme arginosuccinate synthase 1 (ASS1), resulting in RCC cells becoming auxotrophic for arginine and leading to a new therapeutic paradigm involving reducing extracellular arginine. Based on our earlier finding that glutamine pathways are reprogrammed in ARPKD, and given the connection between arginine and glutamine synthetic pathways via citrulline, we investigated the possibility of arginine reprogramming in ADPKD. We now show that, in a remarkable parallel to RCC, ASS1 expression is reduced in murine and human ADPKD, and arginine depletion results in a dose-dependent compensatory increase in ASS1 levels as well as decreased cystogenesis in vitro and ex vivo with minimal toxicity to normal cells. Nontargeted metabolomics analysis of mouse kidney cell lines grown in arginine-deficient versus arginine-replete media suggests arginine-dependent alterations in the glutamine and proline pathways. Thus, depletion of this conditionally essential amino acid by dietary or pharmacological means, such as with arginine-degrading enzymes, may be a novel treatment for this disease.


Assuntos
Arginina/metabolismo , Proliferação de Células , Metabolismo Energético , Rim/metabolismo , Rim Policístico Autossômico Dominante/metabolismo , Animais , Arginina/deficiência , Arginina/farmacologia , Argininossuccinato Sintase/genética , Argininossuccinato Sintase/metabolismo , Proliferação de Células/efeitos dos fármacos , Células Cultivadas , Modelos Animais de Doenças , Metabolismo Energético/efeitos dos fármacos , Feminino , Predisposição Genética para Doença , Humanos , Rim/efeitos dos fármacos , Rim/patologia , Masculino , Metabolômica/métodos , Camundongos Knockout , Fenótipo , Rim Policístico Autossômico Dominante/genética , Rim Policístico Autossômico Dominante/patologia , Receptores de Superfície Celular/deficiência , Receptores de Superfície Celular/genética , Transdução de Sinais , Canais de Cátion TRPP/deficiência , Canais de Cátion TRPP/genética
6.
Breast Cancer Res ; 19(1): 102, 2017 Sep 02.
Artigo em Inglês | MEDLINE | ID: mdl-28865492

RESUMO

BACKGROUND: Stat1 gene-targeted knockout mice (129S6/SvEvTac-Stat1 tm1Rds) develop estrogen receptor-positive (ER+), luminal-type mammary carcinomas at an advanced age. There is evidence for both host environment as well as tumor cell-intrinsic mechanisms to initiate tumorigenesis in this model. In this report, we summarize details of the systemic and mammary pathology at preneoplastic and tumor-bearing time points. In addition, we investigate tumor progression in the 129:Stat1 -/- host compared with wild-type 129/SvEv, and we describe the immune cell reaction to the tumors. METHODS: Mice housed and treated according to National Institutes of Health guidelines and Institutional Animal Care and Use Committee-approved methods were evaluated by histopathology, and their tissues were subjected to immunohistochemistry with computer-assisted quantitative image analysis. Tumor cell culture and conditioned media from cell culture were used to perform macrophage (RAW264.7) cell migration assays, including the 129:Stat1 -/--derived SSM2 cells as well as control Met1 and NDL tumor cells and EpH4 normal cells. RESULTS: Tumorigenesis in 129:Stat1 -/- originates from a population of FoxA1+ large oval pale cells that initially appear and accumulate along the mammary ducts in segments or regions of the gland prior to giving rise to mammary intraepithelial neoplasias. Progression to invasive carcinoma is accompanied by a marked local stromal and immune cell response composed predominantly of T cells and macrophages. In conditioned media experiments, cells derived from 129:Stat1 -/- tumors secrete both chemoattractant and chemoinhibitory factors, with greater attraction in the extracellular vesicular fraction and inhibition in the soluble fraction. The result appears to be recruitment of the immune reaction to the periphery of the tumor, with exclusion of immune cell infiltration into the tumor. CONCLUSIONS: 129:Stat1 -/- is a unique model for studying the critical origins and risk reduction strategies in age-related ER+ breast cancer. In addition, it can be used in preclinical trials of hormonal and targeted therapies as well as immunotherapies.


Assuntos
Neoplasias da Mama/etiologia , Neoplasias da Mama/metabolismo , Fenótipo , Receptores de Estrogênio/metabolismo , Fator de Transcrição STAT1/deficiência , Fatores Etários , Animais , Neoplasias da Mama/mortalidade , Neoplasias da Mama/patologia , Quimiotaxia de Leucócito/imunologia , Modelos Animais de Doenças , Feminino , Humanos , Imuno-Histoquímica , Incidência , Macrófagos/imunologia , Macrófagos/metabolismo , Macrófagos/patologia , Neoplasias Mamárias Experimentais , Camundongos , Camundongos da Linhagem 129 , Camundongos Knockout
7.
J Dairy Sci ; 99(1): 875-83, 2016 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-26506542

RESUMO

The mammary gland (MG) is one of a few organs that undergoes most of its growth after birth. Much of this development occurs concurrently with specific reproductive states, such that the ultimate goal of milk synthesis and secretion is coordinated with the nutritional requirements of the neonate. Central to the reproductive-MG axis is its endocrine regulation, and pivotal to this regulation is the ovarian secretion of estrogen (E). Indeed, it is widely accepted that estrogens are essential for growth of the MG to occur, both for ductal elongation during puberty and for alveolar development during gestation. As the factors regulating MG development continually come to light from the fields of developmental biology, lactation physiology, and breast cancer research, a growing body of evidence serves as a reminder that the MG are not as exclusively dependent on estrogens as might have been thought. The objective of this review is to summarize the state of information regarding our understanding of how estrogen (E) has been implicated as the key regulator of MG development, and to highlight some of the alternative E-independent mechanisms that have been discovered. In particular, we review our findings that dietary trans-10,cis-12 conjugated linoleic acid promotes ductal elongation and that the combination of progesterone (P) and prolactin (PRL) can stimulate branching morphogenesis in the absence of E. Ultimately, these examples stand as a healthy challenge to the question of just how important estrogens are for MG development. Answers to this question, in turn, increase our understanding of MG development across all mammals and the ways in which it can affect milk production.


Assuntos
Glândulas Mamárias Animais/crescimento & desenvolvimento , Ração Animal/análise , Animais , Dieta/veterinária , Estrogênios/fisiologia , Feminino , Lactação , Ácidos Linoleicos Conjugados/administração & dosagem , Glândulas Mamárias Animais/fisiologia , Leite/metabolismo , Morfogênese , Progesterona/metabolismo , Progesterona/fisiologia , Prolactina/fisiologia , Ruminantes
8.
Proc Natl Acad Sci U S A ; 109(40): 16294-9, 2012 Oct 02.
Artigo em Inglês | MEDLINE | ID: mdl-22988119

RESUMO

Lifetime breast cancer risk reflects an unresolved combination of early life factors including diet, body mass index, metabolic syndrome, obesity, and age at first menses. In parallel, the onset of allometric growth by the mammary glands around puberty is widely held to be estrogen (E)-dependent. Here we report that several physiological changes associated with metabolic syndrome in response to a diet supplemented with the trans-10, cis-12 isomer of conjugated linoleic acid lead to ovary-independent allometric growth of the mammary ducts. The E-independence of this diet-induced growth was highlighted by the fact that it occurred both in male mice and with pharmacological inhibition of either E receptor function or E biosynthesis. Reversal of the metabolic phenotype with the peroxisome proliferator-activated receptor-γ agonist rosiglitazone abrogated diet-induced mammary growth. A role for hyperinsulinemia and increased insulin-like growth factor-I receptor (IGF-IR) expression during mammary growth induced by the trans-10, cis-12 isomer of conjugated linoleic acid was confirmed by its reversal upon pharmacological inhibition of IGF-IR function. Diet-stimulated ductal growth also increased mammary tumorigenesis in ovariectomized polyomavirus middle T-antigen mice. Our data demonstrate that diet-induced metabolic dysregulation, independently of ovarian function, stimulates allometric growth within the mammary glands via an IGF-IR-dependent mechanism.


Assuntos
Ração Animal/análise , Ácidos Linoleicos Conjugados/farmacologia , Glândulas Mamárias Animais/efeitos dos fármacos , Glândulas Mamárias Animais/crescimento & desenvolvimento , Síndrome Metabólica/dietoterapia , Animais , Western Blotting , Corticosterona/sangue , Primers do DNA/genética , Ácidos Graxos/análise , Feminino , Técnicas Histológicas , Insulina/sangue , Fator de Crescimento Insulin-Like I/análise , Análise dos Mínimos Quadrados , Ácidos Linoleicos Conjugados/uso terapêutico , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Transgênicos , Reação em Cadeia da Polimerase , Rosiglitazona , Tiazolidinedionas
9.
Anim Genet ; 45(1): 74-86, 2014 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-24350599

RESUMO

The actions of prolactin (PRL) are mediated by both long (LF) and short isoforms (SF) of the PRL receptor (PRLR). Here, we report on a genetic and functional analysis of the porcine PRLR (pPRLR) SF. Three single nucleotide polymorphisms (SNPs) within exon 11 of the pPRLR-SF give rise to four amino acid haplotypes of the intracellular domain. We identified the dimorphic insertion of a short interspersed repetitive DNA element (PRE-1) along with 32 SNPs and four other insertion/deletion sites within the 3' untranslated region (UTR) of pPRLR-SF. The PRE-1 element reduced protein translation in vitro by 75%, whereas the combination of 10 SNPs and one insertion/deletion decreased translation by 50%. Full-length cDNAs for all four haplotypes of pPRLR-SF were cloned behind the elongation factor 1-alpha promoter and functionally analyzed in vitro. None of the haplotypes could initiate transcription from the ß-casein promoter, whereas all four were dominant negatives against PRL-activation of the pPRLR-LF. Two of the haplotypes completely inhibited pPRLR-LF activity at a four-fold excess, whereas the others required a six-fold excess to impart the same effect. The ligand binding affinities of the pPRLR-SF haplotypes did not differ. Expression of the pPRLR-SF increased linearly during gestation in the endometrium and was hormonally regulated in a tissue-specific manner in the mammary glands and uterus. In conclusion, we identified a PRE-1 and other SNPs in the pPRLR-SF 3' UTR that reduce protein expression and four haplotypes of the pPRLR-SF that suppress pPRLR-LF signaling and may differentially impact the phenotypic effects of PRL in vivo.


Assuntos
Regiões 3' não Traduzidas , Polimorfismo de Nucleotídeo Único , Receptores da Prolactina/genética , Animais , Sequência de Bases , Cruzamento , Éxons , Feminino , Haplótipos , Mutação INDEL , Sequências Repetitivas Dispersas , Dados de Sequência Molecular , Isoformas de Proteínas/genética , Suínos
10.
J Neuroendocrinol ; 36(7): e13385, 2024 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-38586906

RESUMO

The conserved and multifaceted functions of prolactin (PRL) are coordinated through varied distribution and expression of its cell-surface receptor (PRLR) across a range of tissues and physiological states. The resultant heterogeneous expression of PRLR mRNA and protein across different organs and cell types supports a wide range of PRL-regulated processes including reproduction, lactation, development, and homeostasis. Genetic variation within the PRLR gene also accounts for several phenotypes impacting agricultural production and human pathology. The goal of this review is to highlight the many elements that control differential expression of the PRLR across tissues, and the various phenotypes that exist across species due to variation in the PRLR gene.


Assuntos
Regulação da Expressão Gênica , Variação Genética , Receptores da Prolactina , Receptores da Prolactina/genética , Receptores da Prolactina/metabolismo , Humanos , Animais , Especificidade da Espécie , Especificidade de Órgãos , Prolactina/metabolismo , Prolactina/genética , Transcrição Gênica/fisiologia
11.
Sci Rep ; 12(1): 7627, 2022 05 10.
Artigo em Inglês | MEDLINE | ID: mdl-35538091

RESUMO

A long intergenic non-coding RNA (lincRNA#1) is overexpressed in the horn bud region of polled (hornless) bovine fetuses, suggesting a potential role in horn bud suppression. Genome editing was used to test whether the absence of this sequence was associated with the horned phenotype. Two gRNAs with high mutation efficiencies targeting the 5' and the 3' regions flanking the lincRNA#1 sequence were co-injected with Cas9 as ribonucleoprotein complexes into bovine zygotes (n = 121) 6 h post insemination. Of the resulting blastocysts (n = 31), 84% had the expected 3.7 kb deletion; of these embryos with the 3.7 kb deletions, 88% were biallelic knockouts. Thirty-nine presumptive edited 7-day blastocysts were transferred to 13 synchronized recipient cows resulting in ten pregnancies, five with embryos heterozygous for the dominant PC POLLED allele at the POLLED locus, and five with the recessive pp genotype. Eight (80%) of the resulting fetuses were biallelic lincRNA#1 knockouts, with the remaining two being mosaic. RT-qPCR analysis was used to confirm the absence of lincRNA#1 expression in knockout fetuses. Phenotypic and histological analysis of the genotypically (PCp) POLLED, lincRNA#1 knockout fetuses revealed similar morphology to non-edited, control polled fetuses, indicating the absence of lincRNA#1 alone does not result in a horned phenotype.


Assuntos
Cornos , RNA Longo não Codificante , Alelos , Animais , Bovinos , Feminino , Heterozigoto , Fenótipo , Gravidez , RNA Longo não Codificante/genética
12.
Endocrinology ; 163(3)2022 03 01.
Artigo em Inglês | MEDLINE | ID: mdl-34918063

RESUMO

Successful lactation and the risk for developing breast cancer depend on growth and differentiation of the mammary gland (MG) epithelium that is regulated by ovarian steroids (17ß-estradiol [E] and progesterone [P]) and pituitary-derived prolactin (PRL). Given that the MG of pigs share histomorphogenic features present in the normal human breast, we sought to define the transcriptional responses within the MG of pigs following exposure to all combinations of these hormones. Hormone-ablated female pigs were administered combinations of E, medroxyprogesterone 17-acetate (source of P), and either haloperidol (to induce PRL) or 2-bromo-α-ergocryptine. We subsequently monitored phenotypic changes in the MG including mitosis, receptors for E and P (ESR1 and PGR), level of phosphorylated STAT5 (pSTAT5), and the frequency of terminal ductal lobular unit (TDLU) subtypes; these changes were then associated with all transcriptomic changes. Estrogen altered the expression of approximately 20% of all genes that were mostly associated with mitosis, whereas PRL stimulated elements of fatty acid metabolism and an inflammatory response. Several outcomes, including increased pSTAT5, highlighted the ability of E to enhance PRL action. Regression of transcriptomic changes against several MG phenotypes revealed 1669 genes correlated with proliferation, among which 29 were E inducible. Additional gene expression signatures were associated with TDLU formation and the frequency of ESR1 or PGR. These data provide a link between the hormone-regulated genome and phenome of the MG in a species having a complex histoarchitecture like that in the human breast, and highlight an underexplored synergy between the actions of E and PRL during MG development.


Assuntos
Estrogênios/fisiologia , Glândulas Mamárias Animais/crescimento & desenvolvimento , Progesterona/fisiologia , Prolactina/fisiologia , Porco Miniatura/fisiologia , Transcriptoma/fisiologia , Animais , Bromocriptina/administração & dosagem , Sinergismo Farmacológico , Estradiol/administração & dosagem , Receptor alfa de Estrogênio/análise , Receptor alfa de Estrogênio/genética , Estrogênios/deficiência , Feminino , Haloperidol/administração & dosagem , Glândulas Mamárias Animais/química , Glândulas Mamárias Animais/efeitos dos fármacos , Acetato de Medroxiprogesterona/administração & dosagem , Modelos Animais , Morfogênese/efeitos dos fármacos , Morfogênese/genética , Ovariectomia , Progesterona/deficiência , Prolactina/deficiência , Receptores de Progesterona/análise , Receptores de Progesterona/genética , Suínos , Transcriptoma/efeitos dos fármacos
13.
Front Genet ; 13: 1072853, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-36561310

RESUMO

Milk production by dairy cows is sensitive to increased levels of stress hormones such as glucocorticoids (GC) that also regulate the transcription of several genes required for milk synthesis. Whereas previous studies identified that an exogenous GC such as dexamethasone (DEX) transiently suppresses milk yield in several species without any pronounced effect on milk protein or fat percentage, the mechanism underlying this effect has not been established. In this study we sought to establish changes within the mammary glands of non-pregnant dairy cows in their second lactation (n = 3-4; 648-838 kg) following a single dose of exogenous DEX. Changes in the udder were monitored by serial biopsy of alternating quarters, concurrent with quarter-level monitoring of milk yield and composition. Dexamethasone increased serum glucose levels from 12-36 h (p <0 .05), reduced milk yield from 12-48 h (p <0 .05), increased % milk protein content at 24 h post-DEX, and transiently decreased both milk lactose and α-lactalbumin content, while not altering the level of milk fat. After 72 h, all aspects of milk production had returned to pre-treatment levels. Transcriptomic changes in the mammary glands in response to DEX were identified by RNA sequencing followed by differential gene expression analysis. Coincident with the milk yield and composition changes was the differential expression of 519 and 320 genes at 12 and 24 h after DEX (adjusted p <0 .05), respectively, with the return of all gene expression to baseline levels by 72 h. Among the transcriptomic changes in response to DEX, there was notable downregulation of elements in the lactose synthesis pathway, specifically AQP3, GALE and LALBA (α-lactalbumin) at 12 h, and sustained downregulation of LALBA at 24 h. One gene in the pathway, UGP2, was upregulated at 12-24 h post-DEX. This work supports the hypothesis that there is a direct relationship between the response to DEX and the concurrent suppression of milk yield due to the reduced synthesis of α-lactalbumin and lactose by the mammary epithelium. The ability of glucocorticoids to modulate the homeorrhetic requirements for glucose during stressful states concurrent with immune activation bears significance for dairy animals as well as a broad range of lactating mammals.

14.
Annu Rev Anim Biosci ; 9: 453-478, 2021 02 16.
Artigo em Inglês | MEDLINE | ID: mdl-33186503

RESUMO

Genetically engineered (GE) livestock were first reported in 1985, and yet only a single GE food animal, the fast-growing AquAdvantage salmon, has been commercialized. There are myriad interconnected reasons for the slow progress in this once-promising field, including technical issues, the structure of livestock industries, lack of public research funding and investment, regulatory obstacles, and concern about public opinion. This review focuses on GE livestock that have been produced and documents the difficulties that researchers and developers have encountered en route. Additionally, the costs associated with delayed commercialization of GE livestock were modeled using three case studies: GE mastitis-resistant dairy cattle, genome-edited porcine reproductive and respiratory syndrome virus-resistant pigs, and the AquAdvantage salmon. Delays of 5 or 10 years in the commercialization of GE livestock beyond the normative 10-year GE product evaluation period were associated with billions of dollars in opportunity costs and reduced global food security.


Assuntos
Animais Geneticamente Modificados , Engenharia Genética/legislação & jurisprudência , Engenharia Genética/veterinária , Animais , Bovinos , Feminino , Gado/genética , Mastite Bovina/genética , Mastite Bovina/prevenção & controle , Síndrome Respiratória e Reprodutiva Suína/genética , Síndrome Respiratória e Reprodutiva Suína/prevenção & controle , Salmão/genética , Salmão/crescimento & desenvolvimento , Suínos , Fatores de Tempo
15.
Kidney360 ; 1(5): 376-388, 2020 May 28.
Artigo em Inglês | MEDLINE | ID: mdl-35224510

RESUMO

BACKGROUND: Kidney cancer (or renal cell carcinoma, RCC) is the sixth most common malignancy in the United States and is increasing in incidence. Despite new therapies, including targeted therapies and immunotherapies, most RCCs are resistant to treatment. Thus, several laboratories have been evaluating new approaches to therapy, both with single agents as well as combinations. Although we have previously shown efficacy of the dual PAK4/nicotinamide phosphoribosyltransferase (NAMPT) inhibitor KPT-9274, and the immune checkpoint inhibitors (CPI) have shown utility in the clinic, there has been no evaluation of this combination either clinically or in an immunocompetent animal model of kidney cancer. METHODS: In this study, we use the renal cell adenocarcinoma (RENCA) model of spontaneous murine kidney cancer. Male BALB/cJ mice were injected subcutaneously with RENCA cells and, after tumors were palpable, they were treated with KPT-9274 and/or anti-programmed cell death 1 (PDCD1; PD1) antibody for 21 days. Tumors were measured and then removed at animal euthanasia for subsequent studies. RESULTS: We demonstrate a significant decrease in allograft growth with the combination treatment of KPT-9274 and anti-PD1 antibody without significant weight loss by the animals. This is associated with decreased (MOUSE) Naprt expression, indicating dependence of these tumors on NAMPT in parallel to what we have observed in human RCC. Histology of the tumors showed substantial necrosis regardless of treatment condition, and flow cytometry of antibody-stained tumor cells revealed that the enhanced therapeutic effect of KPT-9274 and anti-PD1 antibody was not driven by infiltration of T cells into tumors. CONCLUSIONS: This study highlights the potential of the RENCA model for evaluating immunologic responses to KPT-9274 and checkpoint inhibitor (CPI) and suggests that therapy with this combination could improve efficacy in RCC beyond what is achievable with CPI alone.


Assuntos
Carcinoma de Células Renais , Neoplasias Renais , Animais , Proteínas Reguladoras de Apoptose/farmacologia , Carcinoma de Células Renais/tratamento farmacológico , Proliferação de Células , Neoplasias Renais/tratamento farmacológico , Masculino , Camundongos , Nicotinamida Fosforribosiltransferase
16.
Sci Rep ; 10(1): 4203, 2020 03 06.
Artigo em Inglês | MEDLINE | ID: mdl-32144367

RESUMO

Autosomal dominant polycystic kidney disease (ADPKD) is one of the most common inherited monogenic disorders, characterized by a progressive decline in kidney function due in part to the formation of fluid-filled cysts. While there is one FDA-approved therapy, it is associated with potential adverse effects, and all other clinical interventions are largely supportive. Insights into the cellular pathways underlying ADPKD have revealed striking similarities to cancer. Moreover, several drugs originally developed for cancer have shown to ameliorate cyst formation and disease progression in animal models of ADPKD. These observations prompted us to develop a high-throughput screening platform of cancer drugs in a quest to repurpose them for ADPKD. We screened ~8,000 compounds, including compounds with oncological annotations, as well as FDA-approved drugs, and identified 155 that reduced the viability of Pkd1-null mouse kidney cells with minimal effects on wild-type cells. We found that 109 of these compounds also reduced in vitro cyst growth of Pkd1-null cells cultured in a 3D matrix. Moreover, the result of the cyst assay identified therapeutically relevant compounds, including agents that interfere with tubulin dynamics and reduced cyst growth without affecting cell viability. Because it is known that several ADPKD therapies with promising outcomes in animal models failed to be translated to human disease, our platform also incorporated the evaluation of compounds in a panel of primary ADPKD and normal human kidney (NHK) epithelial cells. Although we observed differences in compound response amongst ADPKD and NHK cell preparation, we identified 18 compounds that preferentially affected the viability of most ADPKD cells with minimal effects on NHK cells. Our study identifies attractive candidates for future efficacy studies in advanced pre-clinical models of ADPKD.


Assuntos
Rim Policístico Autossômico Dominante/metabolismo , Acrilamidas/farmacologia , Aminopiridinas/farmacologia , Animais , Linhagem Celular , Sobrevivência Celular/efeitos dos fármacos , Reposicionamento de Medicamentos/métodos , Células Epiteliais/citologia , Células Epiteliais/efeitos dos fármacos , Células Epiteliais/metabolismo , Humanos , Rim/citologia , Rim/metabolismo , Camundongos , Proteína Quinase C/genética , Proteína Quinase C/metabolismo , Transdução de Sinais/efeitos dos fármacos
17.
Nat Biotechnol ; 38(2): 225-232, 2020 02.
Artigo em Inglês | MEDLINE | ID: mdl-31591551

RESUMO

Genome editing followed by reproductive cloning was previously used to produce two hornless dairy bulls. We crossed one genome-edited dairy bull, homozygous for the dominant PC Celtic POLLED allele, with horned cows (pp) and obtained six heterozygous (PCp) polled calves. The calves had no horns and were otherwise healthy and phenotypically unremarkable. We conducted whole-genome sequencing of all animals using an Illumina HiSeq4000 to achieve ~20× coverage. Bioinformatics analyses revealed the bull was a compound heterozygote, carrying one naturally occurring PC Celtic POLLED allele and an allele containing an additional introgression of the homology-directed repair donor plasmid along with the PC Celtic allele. These alleles segregated in the offspring of this bull, and inheritance of either allele produced polled calves. No other unintended genomic alterations were observed. These data can be used to inform conversations in the scientific community, with regulatory authorities and with the public around 'intentional genomic alterations' and future regulatory actions regarding genome-edited animals.


Assuntos
Bovinos/genética , Edição de Genes , Genoma , Alelos , Animais , Sequência de Bases , Cruzamento , Quimerismo , Feminino , Feto/fisiologia , Loci Gênicos , Genótipo , Cornos , Masculino , Fenótipo , Filogenia , Plasmídeos/genética , Polimorfismo de Nucleotídeo Único/genética
18.
Nat Biotechnol ; 38(2): 245, 2020 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-31992864

RESUMO

An amendment to this paper has been published and can be accessed via a link at the top of the paper.

19.
J Mammary Gland Biol Neoplasia ; 13(1): 3-11, 2008 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-18204889

RESUMO

Around 80 years ago researchers first established that the pituitary gland regulates mammary gland function as demonstrated by the ability of its extracts to promote both mammogenesis and lactogenesis in animal models. Little did they realize that in fact two hormones, prolactin (PRL) and growth hormone (GH), were contributing to these effects. By the mid 1930s PRL had been purified as a distinct lactogen, while the galactopoietic effect of GH was confirmed after its purification in the 1940s. Interest in these hormones initially centered about their potential for increasing milk production, while in the latter half of the twentieth century it became obvious that these hormones also had the potential to influence mammary cancer development. During the past 50 years large strides have been made into understanding how these hormones signal to, and within, cells of the mammary gland, paralleling rapid developments in the fields of cellular and molecular biology. In compiling this review we have summarized the progress that has been made to date regarding roles for these hormones in the mammary gland, with a goal of ensuring that some of the seminal literature is not diluted or forgotten. In doing so it is clear that there are lessons to be learned from past experiences, where new methods and technologies will continue to present exciting new opportunities to revisit lingering questions regarding these fascinating hormones and this fascinating organ.


Assuntos
Hormônio do Crescimento/história , Hormônio do Crescimento/metabolismo , Lactose/metabolismo , Glândulas Mamárias Animais/metabolismo , Glândulas Mamárias Humanas/metabolismo , Prolactina/história , Prolactina/metabolismo , Animais , Galactose/metabolismo , História do Século XX , História do Século XXI , Humanos , Glândulas Mamárias Animais/citologia , Glândulas Mamárias Animais/crescimento & desenvolvimento , Glândulas Mamárias Humanas/citologia , Glândulas Mamárias Humanas/crescimento & desenvolvimento
20.
Domest Anim Endocrinol ; 33(3): 313-34, 2007 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-16905289

RESUMO

Prolactin (PRL) regulates various functions in pigs including reproduction, mammary development and lactation. We used 5'-rapid amplification of cDNA ends (5'-RACE) to clone three full-length alleles of the porcine PRL receptor (pPRLR) from Landrace (alleles LR2 and LR4) and Yucatan miniature (MP) pigs, corresponding to the A and B alleles previously reported to be associated with reproductive traits. When expressed in Chinese hamster ovary (CHO-K1) cells, all three pPRLRs transduced differentiation signals to a beta-casein promoter with the same effectiveness, where human growth hormone (hGH) and porcine PRL (pPRL) were more effective ligands than ovine PRL (oPRL). The pPRLR had a lower binding affinity for oPRL than pPRL while binding affinity for hGH was not different between the three pPRLR variants. The pPRLRs primarily localized to the cytoplasm with perinuclear concentration. In conclusion, we have cloned three allelic variants of the pPRLR and have functionally characterized these as different from the hPRLR. However, our data do not support the proposal that allelic variation of the pPRLR confers functional differences in vivo.


Assuntos
Receptores da Prolactina/genética , Porco Miniatura/genética , Alelos , Sequência de Aminoácidos , Animais , Sequência de Bases , Northern Blotting/veterinária , Células CHO , Clonagem Molecular , Cricetinae , Cricetulus , Feminino , Variação Genética , Hormônio do Crescimento Humano/metabolismo , Hormônio do Crescimento Humano/farmacologia , Imuno-Histoquímica , Microscopia de Fluorescência , Dados de Sequência Molecular , Prolactina/metabolismo , Prolactina/farmacologia , Regiões Promotoras Genéticas , RNA/química , RNA/genética , Técnica de Amplificação ao Acaso de DNA Polimórfico/veterinária , Receptores da Prolactina/biossíntese , Receptores da Prolactina/metabolismo , Transdução de Sinais , Suínos , Porco Miniatura/metabolismo , Transfecção/veterinária
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA