Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 17 de 17
Filtrar
1.
Aging Cell ; : e14150, 2024 Apr 04.
Artículo en Inglés | MEDLINE | ID: mdl-38576084

RESUMEN

Hutchinson-Gilford Progeria syndrome (HGPS) is a lethal premature aging disorder caused by a de novo heterozygous mutation that leads to the accumulation of a splicing isoform of Lamin A termed progerin. Progerin expression deregulates the organization of the nuclear lamina and the epigenetic landscape. Progerin has also been observed to accumulate at low levels during normal aging in cardiovascular cells of adults that do not carry genetic mutations linked with HGPS. Therefore, the molecular mechanisms that lead to vascular dysfunction in HGPS may also play a role in vascular aging-associated diseases, such as myocardial infarction and stroke. Here, we show that HGPS patient-derived vascular smooth muscle cells (VSMCs) recapitulate HGPS molecular hallmarks. Transcriptional profiling revealed cardiovascular disease remodeling and reactive oxidative stress response activation in HGPS VSMCs. Proteomic analyses identified abnormal acetylation programs in HGPS VSMC replication fork complexes, resulting in reduced H4K16 acetylation. Analysis of acetylation kinetics revealed both upregulation of K16 deacetylation and downregulation of K16 acetylation. This correlates with abnormal accumulation of error-prone nonhomologous end joining (NHEJ) repair proteins on newly replicated chromatin. The knockdown of the histone acetyltransferase MOF recapitulates preferential engagement of NHEJ repair activity in control VSMCs. Additionally, we find that primary donor-derived coronary artery vascular smooth muscle cells from aged individuals show similar defects to HGPS VSMCs, including loss of H4K16 acetylation. Altogether, we provide insight into the molecular mechanisms underlying vascular complications associated with HGPS patients and normative aging.

2.
Adv Sci (Weinh) ; 10(26): e2302611, 2023 09.
Artículo en Inglés | MEDLINE | ID: mdl-37400371

RESUMEN

Lymphangioleiomyomatosis (LAM) is a rare disease involving cystic lung destruction by invasive LAM cells. These cells harbor loss-of-function mutations in TSC2, conferring hyperactive mTORC1 signaling. Here, tissue engineering tools are employed to model LAM and identify new therapeutic candidates. Biomimetic hydrogel culture of LAM cells is found to recapitulate the molecular and phenotypic characteristics of human disease more faithfully than culture on plastic. A 3D drug screen is conducted, identifying histone deacetylase (HDAC) inhibitors as anti-invasive agents that are also selectively cytotoxic toward TSC2-/- cells. The anti-invasive effects of HDAC inhibitors are independent of genotype, while selective cell death is mTORC1-dependent and mediated by apoptosis. Genotype-selective cytotoxicity is seen exclusively in hydrogel culture due to potentiated differential mTORC1 signaling, a feature that is abrogated in cell culture on plastic. Importantly, HDAC inhibitors block invasion and selectively eradicate LAM cells in vivo in zebrafish xenografts. These findings demonstrate that tissue-engineered disease modeling exposes a physiologically relevant therapeutic vulnerability that would be otherwise missed by conventional culture on plastic. This work substantiates HDAC inhibitors as possible therapeutic candidates for the treatment of patients with LAM and requires further study.


Asunto(s)
Neoplasias Pulmonares , Linfangioleiomiomatosis , Animales , Humanos , Linfangioleiomiomatosis/tratamiento farmacológico , Linfangioleiomiomatosis/genética , Linfangioleiomiomatosis/metabolismo , Neoplasias Pulmonares/metabolismo , Inhibidores de Histona Desacetilasas/farmacología , Inhibidores de Histona Desacetilasas/uso terapéutico , Ingeniería de Tejidos , Pez Cebra , Diana Mecanicista del Complejo 1 de la Rapamicina
3.
iScience ; 25(11): 105316, 2022 Nov 18.
Artículo en Inglés | MEDLINE | ID: mdl-36254158

RESUMEN

The severe acute respiratory syndrome coronavirus-2 (SARS-CoV-2) spike glycoprotein (S) binds to angiotensin-converting enzyme 2 (ACE2) to mediate membrane fusion via two distinct pathways: 1) a surface, serine protease-dependent or 2) an endosomal, cysteine protease-dependent pathway. In this study, we found that SARS-CoV-2 S has a wider protease usage and can also be activated by TMPRSS13 and matrix metalloproteinases (MMPs). We found that MMP-2 and MMP-9 played roles in SARS-CoV-2 S cell-cell fusion and TMPRSS2- and cathepsin-independent viral entry in cells expressing high MMP levels. MMP-dependent viral entry required cleavage at the S1/S2 junction in viral producer cells, and differential processing of variants of concern S dictated its usage; the efficiently processed Delta S preferred metalloproteinase-dependent entry when available, and less processed Omicron S was unable to us metalloproteinases for entry. As MMP-2/9 are released during inflammation, they may play roles in S-mediated cytopathic effects, tropism, and disease outcome.

4.
Cell Rep ; 40(1): 111048, 2022 07 05.
Artículo en Inglés | MEDLINE | ID: mdl-35793620

RESUMEN

Tuberous sclerosis complex (TSC) is a multisystem tumor-forming disorder caused by loss of TSC1 or TSC2. Renal manifestations predominately include cysts and angiomyolipomas. Despite a well-described monogenic etiology, the cellular pathogenesis remains elusive. We report a genetically engineered human renal organoid model that recapitulates pleiotropic features of TSC kidney disease in vitro and upon orthotopic xenotransplantation. Time course single-cell RNA sequencing demonstrates that loss of TSC1 or TSC2 affects multiple developmental processes in the renal epithelial, stromal, and glial compartments. First, TSC1 or TSC2 ablation induces transitional upregulation of stromal-associated genes. Second, epithelial cells in the TSC1-/- and TSC2-/- organoids exhibit a rapamycin-insensitive epithelial-to-mesenchymal transition. Third, a melanocytic population forms exclusively in TSC1-/- and TSC2-/- organoids, branching from MITF+ Schwann cell precursors. Together, these results illustrate the pleiotropic developmental consequences of biallelic inactivation of TSC1 or TSC2 and offer insight into TSC kidney lesion pathogenesis.


Asunto(s)
Esclerosis Tuberosa , Humanos , Riñón/patología , Organoides/patología , Esclerosis Tuberosa/genética , Proteína 1 del Complejo de la Esclerosis Tuberosa/genética , Proteína 2 del Complejo de la Esclerosis Tuberosa , Proteínas Supresoras de Tumor/genética
5.
Stem Cell Reports ; 13(6): 1111-1125, 2019 12 10.
Artículo en Inglés | MEDLINE | ID: mdl-31813826

RESUMEN

Human pluripotent stem cells (hPSCs) are an essential cell source in tissue engineering, studies of development, and disease modeling. Efficient, broadly amenable protocols for rapid lineage induction of hPSCs are of great interest in the stem cell biology field. We describe a simple, robust method for differentiation of hPSCs into mesendoderm in defined conditions utilizing single-cell seeding (SCS) and BMP4 and Activin A (BA) treatment. BA treatment was readily incorporated into existing protocols for chondrogenic and endothelial progenitor cell differentiation, while fine-tuning of BA conditions facilitated definitive endoderm commitment. After prolonged differentiation in vitro or in vivo, BA pretreatment resulted in higher mesoderm and endoderm levels at the expense of ectoderm formation. These data demonstrate that SCS with BA treatment is a powerful method for induction of mesendoderm that can be adapted for use in mesoderm and endoderm differentiation.


Asunto(s)
Diferenciación Celular/genética , Mesodermo/citología , Mesodermo/metabolismo , Células Madre Pluripotentes/citología , Células Madre Pluripotentes/metabolismo , Transcripción Genética , Activinas/farmacología , Proteína Morfogenética Ósea 4/farmacología , Técnicas de Cultivo de Célula , Diferenciación Celular/efectos de los fármacos , Células Cultivadas , Endodermo/citología , Endodermo/metabolismo , Perfilación de la Expresión Génica , Humanos , Células Madre Pluripotentes/efectos de los fármacos , Análisis de la Célula Individual , Teratoma/etiología , Factores de Tiempo , Transcriptoma
6.
Adv Mater ; 31(7): e1806214, 2019 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-30589121

RESUMEN

Cell behavior is highly dependent upon microenvironment. Thus, to identify drugs targeting metastatic cancer, screens need to be performed in tissue mimetic substrates that allow cell invasion and matrix remodeling. A novel biomimetic 3D hydrogel platform that enables quantitative analysis of cell invasion and viability at the individual cell level is developed using automated data acquisition methods with an invasive lung disease (lymphangioleiomyomatosis, LAM) characterized by hyperactive mammalian target of rapamycin complex 1 (mTORC1) signaling as a model. To test the lung-mimetic hydrogel platform, a kinase inhibitor screen is performed using tuberous sclerosis complex 2 (TSC2) hypomorphic cells, identifying Cdk2 inhibition as a putative LAM therapeutic. The 3D hydrogels mimic the native niche, enable multiple modes of invasion, and delineate phenotypic differences between healthy and diseased cells, all of which are critical to effective drug screens of highly invasive diseases including lung cancer.


Asunto(s)
Movimiento Celular/efectos de los fármacos , Evaluación Preclínica de Medicamentos/instrumentación , Hidrogeles , Neoplasias Pulmonares/tratamiento farmacológico , Modelos Biológicos , Animales , Antineoplásicos/farmacología , Automatización de Laboratorios , Materiales Biomiméticos , Movimiento Celular/fisiología , Supervivencia Celular/efectos de los fármacos , Células Cultivadas , Humanos , Células Madre Pluripotentes Inducidas/efectos de los fármacos , Células Madre Pluripotentes Inducidas/metabolismo , Pulmón/efectos de los fármacos , Pulmón/metabolismo , Pulmón/patología , Neoplasias Pulmonares/metabolismo , Neoplasias Pulmonares/patología , Ensayo de Materiales , Fosfotransferasas/antagonistas & inhibidores , Ratas , Proteína 2 del Complejo de la Esclerosis Tuberosa/metabolismo
7.
Cancer Discov ; 8(11): 1376-1389, 2018 11.
Artículo en Inglés | MEDLINE | ID: mdl-30115703

RESUMEN

Deep sequencing has revealed that epigenetic modifiers are the most mutated genes in acute myeloid leukemia (AML). Thus, elucidating epigenetic dysregulation in AML is crucial to understand disease mechanisms. Here, we demonstrate that metal response element binding transcription factor 2/polycomblike 2 (MTF2/PCL2) plays a fundamental role in the polycomb repressive complex 2 (PRC2) and that its loss elicits an altered epigenetic state underlying refractory AML. Unbiased systems analyses identified the loss of MTF2-PRC2 repression of MDM2 as central to, and therefore a biomarker for, refractory AML. Thus, immature MTF2-deficient CD34+CD38- cells overexpress MDM2, thereby inhibiting p53 that leads to chemoresistance due to defects in cell-cycle regulation and apoptosis. Targeting this dysregulated signaling pathway by MTF2 overexpression or MDM2 inhibitors sensitized refractory patient leukemic cells to induction chemotherapeutics and prevented relapse in AML patient-derived xenograft mice. Therefore, we have uncovered a direct epigenetic mechanism by which MTF2 functions as a tumor suppressor required for AML chemotherapeutic sensitivity and identified a potential therapeutic strategy to treat refractory AML.Significance: MTF2 deficiency predicts refractory AML at diagnosis. MTF2 represses MDM2 in hematopoietic cells and its loss in AML results in chemoresistance. Inhibiting p53 degradation by overexpressing MTF2 in vitro or by using MDM2 inhibitors in vivo sensitizes MTF2-deficient refractory AML cells to a standard induction-chemotherapy regimen. Cancer Discov; 8(11); 1376-89. ©2018 AACR. See related commentary by Duy and Melnick, p. 1348 This article is highlighted in the In This Issue feature, p. 1333.


Asunto(s)
Daunorrubicina/farmacología , Resistencia a Antineoplásicos , Leucemia Mieloide Aguda/tratamiento farmacológico , Complejo Represivo Polycomb 2/metabolismo , Proteínas Proto-Oncogénicas c-mdm2/metabolismo , Animales , Antibióticos Antineoplásicos/farmacología , Humanos , Leucemia Mieloide Aguda/inmunología , Leucemia Mieloide Aguda/metabolismo , Leucemia Mieloide Aguda/patología , Ratones , Ratones Endogámicos NOD , Ratones SCID , Complejo Represivo Polycomb 2/antagonistas & inhibidores , Complejo Represivo Polycomb 2/genética , Proteínas Proto-Oncogénicas c-mdm2/antagonistas & inhibidores , Proteínas Proto-Oncogénicas c-mdm2/genética , Células Tumorales Cultivadas , Proteína p53 Supresora de Tumor/genética , Proteína p53 Supresora de Tumor/metabolismo , Ensayos Antitumor por Modelo de Xenoinjerto
8.
Phytother Res ; 32(4): 705-714, 2018 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-29377302

RESUMEN

Perturbations to extravillous trophoblast (EVT) cell migration and invasion are associated with the development of placenta-mediated diseases. Phytochemicals found in the lowbush blueberry plant (Vaccinium angustifolium) have been shown to influence cell migration and invasion in models of tumorigenesis and noncancerous, healthy cells, however never in EVT cells. We hypothesized that the phenolic compounds present in V. angustifolium leaf extract promote trophoblast migration and invasion. Using the HTR-8/SVneo human EVT cell line and Boyden chamber assays, the influence of V. angustifolium leaf extract (0 to 2 × 104  ng/ml) on trophoblast cell migration (n = 4) and invasion (n = 4) was determined. Cellular proliferation and viability were assessed using immunoreactivity to Ki67 (n = 3) and trypan blue exclusion assays (n = 3), respectively. At 20 ng/ml, V. angustifolium leaf extract increased HTR-8/SVneo cell migration and invasion (p < .01) and did not affect cell proliferation or viability. Chlorogenic acid was identified as a major phenolic compound of the leaf extract and the most active compound. Evidence from Western blot analysis (n = 3) suggests that the effects of the leaf extract and chlorogenic acid on trophoblast migration and invasion are mediated through an adenosine monophosphate-activated protein (AMP) kinase-dependent mechanism. Further investigations examining the potential therapeutic applications of this natural health product extract and its major chemical compounds in the context of placenta-mediated diseases are warranted.


Asunto(s)
Arándanos Azules (Planta)/química , Movimiento Celular/efectos de los fármacos , Extractos Vegetales/química , Hojas de la Planta/química , Trofoblastos/metabolismo
9.
Cancer Res ; 77(20): 5491-5502, 2017 10 15.
Artículo en Inglés | MEDLINE | ID: mdl-28830860

RESUMEN

Lymphangioleiomyomatosis (LAM) is a progressive destructive neoplasm of the lung associated with inactivating mutations in the TSC1 or TSC2 tumor suppressor genes. Cell or animal models that accurately reflect the pathology of LAM have been challenging to develop. Here, we generated a robust human cell model of LAM by reprogramming TSC2 mutation-bearing fibroblasts from a patient with both tuberous sclerosis complex (TSC) and LAM (TSC-LAM) into induced pluripotent stem cells (iPSC), followed by selection of cells that resemble those found in LAM tumors by unbiased in vivo differentiation. We established expandable cell lines under smooth muscle cell (SMC) growth conditions that retained a patient-specific genomic TSC2+/- mutation and recapitulated the molecular and functional characteristics of pulmonary LAM cells. These include multiple indicators of hyperactive mTORC1 signaling, presence of specific neural crest and SMC markers, expression of VEGF-D and female sex hormone receptors, reduced autophagy, and metabolic reprogramming. Intriguingly, the LAM-like features of these cells suggest that haploinsufficiency at the TSC2 locus contributes to LAM pathology, and demonstrated that iPSC reprogramming and SMC lineage differentiation of somatic patient cells with germline mutations was a viable approach to generate LAM-like cells. The patient-derived SMC lines we have developed thus represent a novel cellular model of LAM that can advance our understanding of disease pathogenesis and develop therapeutic strategies against LAM. Cancer Res; 77(20); 5491-502. ©2017 AACR.


Asunto(s)
Linfangioleiomiomatosis/genética , Linfangioleiomiomatosis/patología , Miocitos del Músculo Liso/fisiología , Células Madre Pluripotentes/fisiología , Animales , Proliferación Celular/fisiología , Femenino , Haploinsuficiencia , Humanos , Ratones , Ratones Endogámicos NOD , Ratones SCID , Miocitos del Músculo Liso/metabolismo , Miocitos del Músculo Liso/patología , Células Madre Pluripotentes/metabolismo , Células Madre Pluripotentes/patología
10.
Placenta ; 42: 1-8, 2016 06.
Artículo en Inglés | MEDLINE | ID: mdl-27238707

RESUMEN

INTRODUCTION: Discriminating between placentally-mediated fetal growth restriction and constitutionally-small fetuses is a challenge in obstetric practice. Placental growth factor (PlGF), measurable in the maternal circulation, may have this discriminatory capacity. METHODS: Plasma PlGF was measured in women presenting with suspected fetal growth restriction (FGR; ultrasound fetal abdominal circumference <10th percentile for gestational age) at sites in Canada, New Zealand and the United Kingdom. When available, placenta tissue underwent histopathological examination for lesions indicating placental dysfunction, blinded to PlGF and clinical outcome. Lesions were evaluated according to pre-specified severity criteria and an overall severity grade was assigned (0-3, absent to severe). Low PlGF (concentration <5th percentile for gestational age) to identify placental FGR (severity grade≥2) was assessed and compared with routine parameters for fetal assessment. For all cases, the relationship between PlGF and the sampling-to-delivery interval was determined. RESULTS: Low PlGF identified placental FGR with an area under the receiver-operator characteristic curve of 0.96 [95% CI 0.93-0.98], 98.2% [95% CI 90.5-99.9] sensitivity and 75.1% [95% CI 67.6-81.7] specificity. Negative and positive predictive values were 99.2% [95% CI 95.4-99.9] and 58.5% [95% CI 47.9-68.6], respectively. Low PlGF outperformed gestational age, abdominal circumference and umbilical artery resistance index in predicting placental FGR. Very low PlGF (<12 pg/mL) was associated with shorter sampling-to-delivery intervals than normal PlGF (13 vs. 29.5 days, P < 0.0001). DISCUSSION: Low PlGF identifies small fetuses with significant underlying placental pathology and is a promising tool for antenatal discrimination of FGR from fetuses who are constitutionally-small.


Asunto(s)
Retardo del Crecimiento Fetal/diagnóstico , Factor de Crecimiento Placentario/sangre , Placenta/patología , Insuficiencia Placentaria/diagnóstico , Adulto , Biomarcadores/sangre , Parto Obstétrico , Femenino , Retardo del Crecimiento Fetal/etiología , Retardo del Crecimiento Fetal/patología , Humanos , Insuficiencia Placentaria/patología , Embarazo , Ultrasonografía Prenatal , Adulto Joven
11.
J Reprod Immunol ; 110: 89-101, 2015 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-25725501

RESUMEN

The success of pregnancy is dependent on the precise regulation of the immune response within the utero-placental environment. Rats are beginning to be widely used as a model for human immune-related pregnancy complications. However, our knowledge of immune cells and cytokine localization in the rat utero-placental tissue is limited. The current study aimed to localize the immune cell populations, including uterine natural killer (uNK) cells, neutrophils, and macrophages within the rat utero-placental unit at two crucial gestational ages, gestational days 15.5 and 18.5. In addition, we characterized the distribution of the cytokines TNFα, IFNγ, and IL-10 in the utero-placental regions at both the above-mentioned gestational ages. Our study has demonstrated co-localization TNFα and IFNγ with uNK cells in perivascular regions of the rat mesometrial triangle at both gestational ages. Neutrophils and IL-10-positive cells were localized at the maternal-fetal interface and in the spiral artery lumen of the rat mesometrial triangle at both gestational ages. TNFα and IL-10 demonstrated a temporal change in the localization from GD15.5 to GD18.5, which coincides with the leading edge of trophoblast invasion into the mesometrial triangle. The current study furthers our knowledge of the localization of uterine immune cells and relevant cytokines, and provides a base from which to research the function of these immune cells and cytokines during rat pregnancy as a model to study human immune-related pregnancy complications.


Asunto(s)
Citocinas/inmunología , Edad Gestacional , Neutrófilos/inmunología , Placenta/inmunología , Embarazo/inmunología , Útero/inmunología , Animales , Femenino , Humanos , Neutrófilos/citología , Placenta/citología , Ratas , Útero/citología
12.
Am J Reprod Immunol ; 74(1): 1-11, 2015 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-25472023

RESUMEN

The process of uterine spiral artery remodeling in the first trimester of human pregnancy is an essential part of establishing adequate blood perfusion of the placenta that will allow optimal nutrient/waste exchange to meet fetal demands during later development. Key regulators of spiral artery remodeling are the uterine natural killer cells and the invasive extravillous trophoblasts. The functions of these cells as well as regulation of their activation states and temporal regulation of their localization within the uterine tissue are beginning to be known. In this review, we discuss the roles of these two cell lineages in arterial remodeling events, their interaction/influence on one another and the outcomes of altered temporal, and spatial regulation of these cells in pregnancy complications.


Asunto(s)
Células Asesinas Naturales/inmunología , Trofoblastos/inmunología , Arteria Uterina/metabolismo , Útero/irrigación sanguínea , Remodelación Vascular/inmunología , Animales , Femenino , Humanos , Células Asesinas Naturales/citología , Ratones , Placenta/irrigación sanguínea , Embarazo , Complicaciones del Embarazo , Embarazo de Alto Riesgo , Receptores de Células Asesinas Naturales/inmunología , Trofoblastos/citología , Útero/citología , Útero/inmunología
13.
Hum Reprod Update ; 21(2): 228-48, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-25376587

RESUMEN

BACKGROUND: Emerging evidence from clinical and epidemiological studies suggests that dietary polyphenols play an important role in the prevention of chronic diseases, including cancer, cardiovascular disease, diabetes and neurodegenerative disorders. Although these beneficial health claims are supported by experimental data for many subpopulation groups, some studies purport that excessive polyphenol consumption may have negative health effects in other subpopulations. The ever-growing interest and public awareness surrounding the potential benefits of natural health products and polyphenols, in addition to their widespread availability and accessibility through nutritional supplements and fortified foods, has led to increased consumption throughout gestation. Therefore, understanding the implications of polyphenol intake on obstetrical health outcomes is of utmost importance with respect to safe consumption during pregnancy. METHODS: Using relevant keywords, a literature search was performed to gather information regarding polyphenol pharmacology and the molecular mechanisms by which polyphenols exert their biological effects. The primary focus of this paper is to understand the relevance of these findings in the context of reproductive physiology and medicine. RESULTS: Evidence from both in vitro experiments and in vivo studies using animals and humans demonstrates that polyphenols regulate key targets related to oxidative stress, inflammation and advanced glycation end products. Although the majority of these studies have been conducted in the context of chronic diseases, such as cancer and diabetes, several of the key targets influenced by polyphenols are also related to a variety of obstetrical complications, including pre-eclampsia, intrauterine growth restriction and preterm birth. Polyphenols have also been shown to influence fertility and sexual development, fetal health and the bioavailability of nutrients. CONCLUSIONS: Further research leading to a thorough understanding of the physiological roles and potential clinical value that polyphenol consumption may play in pregnancy is urgently needed to help inform patient safety.


Asunto(s)
Estrés Oxidativo , Polifenoles/farmacología , Salud Reproductiva , Animales , Femenino , Humanos , Inflamación/metabolismo , Ratones , Polifenoles/efectos adversos , Polifenoles/química , Preeclampsia/metabolismo , Embarazo , Nacimiento Prematuro/metabolismo
14.
Int J Mol Sci ; 15(9): 16153-85, 2014 Sep 12.
Artículo en Inglés | MEDLINE | ID: mdl-25222554

RESUMEN

Appropriate in utero growth is essential for offspring development and is a critical contributor to long-term health. Fetal growth is largely dictated by the availability of nutrients in maternal circulation and the ability of these nutrients to be transported into fetal circulation via the placenta. Substrate flux across placental gradients is dependent on the accessibility and activity of nutrient-specific transporters. Changes in the expression and activity of these transporters is implicated in cases of restricted and excessive fetal growth, and may represent a control mechanism by which fetal growth rate attempts to match availability of nutrients in maternal circulation. This review provides an overview of placenta nutrient transport with an emphasis on macro-nutrient transporters. It highlights the changes in expression and activity of these transporters associated with common pregnancy pathologies, including intrauterine growth restriction, macrosomia, diabetes and obesity, as well as the potential impact of maternal diet. Molecular signaling pathways linking maternal nutrient availability and placenta nutrient transport are discussed. How sexual dimorphism affects fetal growth strategies and the placenta's response to an altered intrauterine environment is considered. Further knowledge in this area may be the first step in the development of targeted interventions to help optimize fetal growth.


Asunto(s)
Placenta/fisiología , Aminoácidos/metabolismo , Animales , Colesterol/metabolismo , Diabetes Gestacional/metabolismo , Diabetes Gestacional/patología , Ácidos Grasos/metabolismo , Femenino , Retardo del Crecimiento Fetal/metabolismo , Retardo del Crecimiento Fetal/patología , Glucosa/metabolismo , Proteínas Facilitadoras del Transporte de la Glucosa/metabolismo , Humanos , Intercambio Materno-Fetal , Obesidad/metabolismo , Obesidad/patología , Embarazo
15.
Dev Dyn ; 243(5): 640-51, 2014 May.
Artículo en Inglés | MEDLINE | ID: mdl-24868594

RESUMEN

BACKGROUND: Over the past decade, the Ste20-like kinase SLK, has been implicated in several signaling processes. SLK repression has been shown to impair cell cycle kinetics and inhibit FAK-mediated cell migration. Here, using a gene trapped allele, we have generated mice expressing a truncated form of the SLK kinase. RESULTS: Our results show that an SLK-LacZ fusion protein is expressed in embryonic stem cells and in embryos throughout development. We find that the SLK-LacZ fusion protein is less efficient at phosphorylating substrates resulting in reduced cell proliferation within the embryos and angiogenic defects in the placentae of the homozygous mutant animals at embryonic day (E) 12.5. This results in marked developmental defects and apoptotic lesions in the embryos by E14.5. CONCLUSIONS: Homozygotes expressing the SLK-LacZ fusion protein present with an embryonic lethal phenotype occurring between E12.5 and E14.5. Overall, we demonstrate a requirement for SLK kinase activity in the developing embryo and placenta.


Asunto(s)
Embrión de Mamíferos/enzimología , Desarrollo Embrionario/fisiología , Placenta/enzimología , Proteínas Gestacionales/metabolismo , Proteínas Serina-Treonina Quinasas/metabolismo , Animales , Embrión de Mamíferos/citología , Femenino , Ratones , Ratones Transgénicos , Placenta/citología , Embarazo , Proteínas Gestacionales/genética , Proteínas Serina-Treonina Quinasas/genética
16.
Biochem Cell Biol ; 87(6): 905-18, 2009 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-19935876

RESUMEN

Intercellular adhesion molecule-1 (ICAM-1) plays an important role in the immune system, enabling the interactions between effector cells and target cells. It is also known to be involved in tumor growth and metastasis. Its expression is transcriptionally regulated by several proinflammatory cytokines including IFN-gamma, which induces ICAM-1 transcription via the JAK-STAT signaling pathway in a Stat1-dependent fashion. The ICAM-1 promoter contains several cis-active regulatory elements including 2 Ets binding sites (EBSs) located at positions -158 and -138 relatively to the AUG, which were previously shown to play a role in the constitutive activity of the ICAM-1 promoter. In the present study, we have determined whether the EBSs are also involved in the regulation of ICAM-1 gene transcription by pro-inflammatory cytokines. Transient transfection assays were performed with reporter genes containing ICAM-1 promoter constructions cloned upstream from the firefly luciferase gene. Site-specific mutations of the EBS diminished the promoter activity stimulated by IFN-gamma, although the IFN-gamma responsive element (pIgammaRE), which binds Stat1, was intact. Stimulation of the transcriptional activity following IFN-gamma treatment was significantly reduced when both EBSs were inactivated. Co-immunoprecipitation experiments provided evidence of a physical interaction involving Ets1 and Stat1. In COS-1 and HEK 293 cells cotransfected with CFP-Stat1 and YFP-Ets fusion protein, fluorescence resonance energy transfer experiments confirmed the close proximity of these 2 proteins in living cells following treatment with IFN-gamma.


Asunto(s)
Molécula 1 de Adhesión Intercelular/metabolismo , Proteína Proto-Oncogénica c-ets-1/metabolismo , Factor de Transcripción STAT1/metabolismo , Transcripción Genética , Animales , Secuencia de Bases , Células COS , Chlorocebus aethiops , Humanos , Molécula 1 de Adhesión Intercelular/genética , Interferón gamma/metabolismo , Datos de Secuencia Molecular , Regiones Promotoras Genéticas , Proteína Proto-Oncogénica c-ets-1/genética , Proteínas Recombinantes de Fusión/genética , Proteínas Recombinantes de Fusión/metabolismo , Factor de Transcripción STAT1/genética
17.
DNA Cell Biol ; 28(11): 561-6, 2009 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-19630533

RESUMEN

Primary endothelial cells are largely recognized as hard-to-transfect cells. We have been using a double-pulse electroporation technique to efficiently insert genetic material into human umbilical vein endothelial cell (HUVEC). Previously, this technique has been successfully used on hard-to-transfect monocytic cells. Using a conventional electroporation device, we have tested this protocol on HUVECs and compared it with conventional transfection techniques. The average transfection efficiency was up to 68% as measured by the ability of the cells to efficiently express the red fluorophore of the tdTomato gene. Similar results were obtained in human aortic endothelial cells and human microvascular endothelial cells. This technique does not require any particular expensive device, specific medium, or reagent, and the results we obtained so far exceed those of any other previous protocol. This is therefore an affordable and efficient transfection technique that opens new avenues in vascular endothelial research.


Asunto(s)
Electroporación/métodos , Células Endoteliales/metabolismo , Transfección/métodos , Aorta/citología , Aorta/metabolismo , Células Cultivadas , Electroporación/economía , Humanos , Microvasos/citología , Microvasos/metabolismo , Transfección/economía
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA