Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 36
Filter
Add more filters










Publication year range
1.
PNAS Nexus ; 3(6): pgae207, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38894879

ABSTRACT

Placental System L amino acid transporter activity is decreased in pregnancies complicated by intrauterine growth restriction (IUGR) and increased in fetal overgrowth. However, it is unknown if changes in the expression/activity of placental Large Neutral Amino Acid Transporter Small Subunit 1 (Slc7a5/LAT1) are mechanistically linked to placental function and fetal growth. We hypothesized that trophoblast-specific Slc7a5 overexpression increases placental transport of essential amino acids, activates the placental mechanistic target of rapamycin (mTOR) signaling, and promotes fetal growth in mice. Using lentiviral transduction of blastocysts with a Slc7a5 transgene, we achieved trophoblast-specific overexpression of Slc7a5 (Slc7a5 OX) with increased fetal (+27%) and placental weights (+10%). Trophoblast-specific Slc7a5 overexpression increased trophoblast plasma membrane (TPM) LAT1 protein abundance and TPM System L transporter (+53%) and System A transporter activity (+ 21%). Slc7a5 overexpression also increased transplacental transport of leucine (+ 85%) but not of the System A tracer, 14C-methylamino isobutyric acid, in vivo. Trophoblast-specific overexpression of Slc7a5 activated placental mTORC1, as assessed by increased (+44%) phosphorylation of S6 ribosomal protein (Ser 235/236), and mTORC2 as indicated by phosphorylation of PKCα-Tyr-657 (+47%) and Akt-Ser 473 (+96%). This is the first demonstration that placental transport of essential amino acids is mechanistically linked to fetal growth. The decreased placental System L activity in human IUGR and the increased placental activity of this transporter in some cases of fetal overgrowth may directly contribute to the development of these pregnancy complications.

2.
Reprod Sci ; 2024 May 22.
Article in English | MEDLINE | ID: mdl-38777947

ABSTRACT

Preeclampsia (PE) is a leading cause of maternal and fetal mortality and morbidity. While placental dysfunction is a core underlying issue, the pathogenesis of this disorder is thought to differ between early-onset (EOPE) and late-onset (LOPE) subtypes. As recent reports suggest that small extracellular vesicles (sEVs) contribute to the development of PE, we have compared systemic sEV concentrations between normotensive, EOPE, and LOPE pregnancies. To circumvent lengthy isolation techniques and intermediate filtration steps, a streamlined approach was developed to evaluate circulating plasma sEVs from maternal plasma. Polymer-based precipitation and purification were used to isolate total systemic circulating maternal sEVs, free from bias toward specific surface marker expression or extensive subpurification. Immediate Nanoparticle Tracking Analysis (NTA) of freshly isolated sEV samples afforded a comprehensive analysis that can be completed within hours, avoiding confounding freeze-thaw effects of particle aggregation and degradation.Rather than exosomal subpopulations, our findings indicate a significant elevation in the total number of circulating maternal sEVs in patients with EOPE. This streamlined approach also preserves sEV-bound protein and microRNA (miRNA) that can be used for potential biomarker analysis. This study is one of the first to demonstrate that maternal plasma sEVs harbor full-length hypoxia inducible factor 1 alpha (HIF-1α) protein, with EOPE sEVs carrying higher levels of HIF-1α compared to control sEVs. The detection of HIF-1α and its direct signaling partner microRNA-210 (miR-210) within systemic maternal sEVs lays the groundwork for identifying how sEV signaling contributes to the development of preeclampsia. When taken together, our quantitative and qualitative results provide compelling evidence to support the translational potential of streamlined sEV analysis for future use in the clinical management of patients with EOPE.

3.
J Pharmacol Exp Ther ; 388(2): 367-375, 2024 01 17.
Article in English | MEDLINE | ID: mdl-37918856

ABSTRACT

Organophosphates cause hyperstimulation of the central nervous system, leading to extended seizures, convulsions, and brain damage. Sarin is a highly toxic organophosphate nerve agent that has been employed in several terrorist attacks. The prolonged toxicity of sarin may be enhanced by the neuroinflammatory response initiated by the inflammasome, caspase involvement, and generation/release of proinflammatory cytokines. Since neurodegeneration and neuroinflammation are prevalent in sarin-exposed animals, we were interested in evaluating the capacity of quinolyl-valyl-O-methylaspartyl-[-2,6-difluorophenoxy]-methyl ketone (Q-VD-OPh), a pan caspase inhibitor to attenuate neuroinflammation following sarin exposure. To test this hypothesis, sarin-exposed C57BL/6 mice were treated with Q-VD-OPh or negative control quinolyl-valyl-O-methylglutamyl-[-2,6-difluorophenoxy]-methyl ketone, sacrificed at 2- and 14-day time points, followed by removal of the amygdala and hippocampus. A Bio-Rad 23-Plex cytokine analysis was completed on each tissue. The results suggest that exposure to sarin induced a dramatic increase in interleukin-1ß and 6 other cytokines and a decrease in 2 of the 23 cytokines at 2 days in the amygdala compared with controls. Q-VD-OPh attenuated these changes at the 2-day time point. At 14 days, six of these cytokines were still significantly different from controls. Hippocampus was less affected at both time points. Diazepam, a neuroprotective drug against nerve agents, caused an increase in several cytokines but did not have a synergistic effect with Q-VD-OPh. Treatment of sarin exposure with apoptosis inhibitors appears to be a worthwhile approach for further testing as a comprehensive counteragent against organophosphate exposure. SIGNIFICANCE STATEMENT: A pan inhibitor of caspases (Q-VD-OPh) was proposed as a potential antidote for sarin-induced neuroinflammation by reducing the level of inflammation via inflammasome caspase inhibition. Q-VD-OPh added at 30 minutes post-sarin exposure attenuated the inflammatory response of a number of cytokines and chemokines in the amygdala and hippocampus, two brain regions sensitive to organophosphate exposure. Apoptotic marker reduction at 2 and 14 days further supports further testing of inhibitors of apoptosis as a means to lessen extended organophosphate toxicity in the brain.


Subject(s)
Amino Acid Chloromethyl Ketones , Nerve Agents , Quinolines , Sarin , Mice , Animals , Sarin/toxicity , Caspase Inhibitors/pharmacology , Caspase Inhibitors/therapeutic use , Neuroinflammatory Diseases , Inflammasomes , Mice, Inbred C57BL , Seizures/chemically induced , Seizures/drug therapy , Brain , Cytokines , Nerve Agents/pharmacology , Caspases , Inflammation/chemically induced , Inflammation/drug therapy , Organophosphates/pharmacology , Ketones/adverse effects
4.
Clin Sci (Lond) ; 137(21): 1651-1664, 2023 11 15.
Article in English | MEDLINE | ID: mdl-37861075

ABSTRACT

The System L amino acid transporter, particularly the isoform Large Neutral Amino Acid Transporter Small Subunit 1 (LAT1) encoded by SLC7A5, is believed to mediate the transfer of essential amino acids in the human placenta. Placental System L amino acid transporter expression and activity is decreased in pregnancies complicated by IUGR and increased in fetal overgrowth. However, it remains unknown if changes in the expression of LAT1 are mechanistically linked to System L amino acid transport activity. Here, we combined overexpression approaches with protein analysis and functional studies in cultured primary human trophoblast (PHT) cells to test the hypothesis that SLC7A5 overexpression increases the uptake of essential amino acids and activates mTOR signaling in PHT cells. Overexpression of SLC7A5 resulted in a marked increase in protein expression of LAT1 in the PHT cells microvillous plasma membrane and System L amino acid transporter activity. Moreover, mTOR signaling was activated, and System A amino acid transporter activity increased following SLC7A5 overexpression, suggesting coordination of trophoblast amino transporter expression and activity to ensure balanced nutrient flux to the fetus. This is the first report showing that overexpression of LAT1 is sufficient to increase the uptake of essential amino acids in PHT cells, which activates mTOR, a master regulator of placental function. The decreased placental System L activity in human IUGR and the increased placental activity of this transporter system in some cases of fetal overgrowth may directly contribute to changes in fetal amino acid availability and altered fetal growth in these pregnancy complications.


Subject(s)
Diabetes, Gestational , Trophoblasts , Female , Humans , Pregnancy , Amino Acids/metabolism , Amino Acids, Essential/metabolism , Diabetes, Gestational/metabolism , Fetal Macrosomia/metabolism , Large Neutral Amino Acid-Transporter 1/genetics , Large Neutral Amino Acid-Transporter 1/metabolism , Membrane Transport Proteins/metabolism , Placenta/metabolism , TOR Serine-Threonine Kinases/metabolism , Trophoblasts/metabolism
5.
Front Physiol ; 14: 1137058, 2023.
Article in English | MEDLINE | ID: mdl-37089425

ABSTRACT

Preeclampsia is a pregnancy-specific condition and a leading cause of maternal and fetal morbidity and mortality. It is thought to occur due to abnormal placental development or dysfunction, because the only known cure is delivery of the placenta. Several clinical risk factors are associated with an increased incidence of preeclampsia including chronic hypertension, diabetes, autoimmune conditions, kidney disease, and obesity. How these comorbidities intersect with preeclamptic etiology, however, is not well understood. This may be due to the limited number of animal models as well as the paucity of studies investigating the impact of these comorbidities. This review examines the current mouse models of chronic hypertension, pregestational diabetes, and obesity that subsequently develop preeclampsia-like symptoms and discusses how closely these models recapitulate the human condition. Finally, we propose an avenue to expand the development of mouse models of preeclampsia superimposed on chronic comorbidities to provide a strong foundation needed for preclinical testing.

6.
Hypertension ; 80(6): 1363-1374, 2023 06.
Article in English | MEDLINE | ID: mdl-36987911

ABSTRACT

BACKGROUND: Placental disorders contribute to pregnancy complications, including preeclampsia and fetal growth restriction (FGR), but debate regarding their specific pathobiology persists. Our objective was to apply transcriptomics with weighted gene correlation network analysis to further clarify the placental dysfunction in these conditions. METHODS: We performed RNA sequencing with weighted gene correlation network analysis using human placental samples (n=30), separated into villous tissue and decidua basalis, and clinically grouped as follows: (1) early-onset preeclampsia (EOPE)+FGR (n=7); (2) normotensive, nonanomalous preterm FGR (n=5); (2) EOPE without FGR (n=8); (4) spontaneous idiopathic preterm birth (n=5) matched for gestational age; and (5) uncomplicated term births (n=5). Our data was compared with RNA sequencing data sets from public databases (GSE114691, GSE148241, and PRJEB30656; n=130 samples). RESULTS: We identified 14 correlated gene modules in our specimens, of which most were significantly correlated with birthweight and maternal blood pressure. Of the 3 network modules consistently predictive of EOPE±FGR across data sets, we prioritized a coexpression gene group enriched for hypoxia-response and metabolic pathways for further investigation. Cluster analysis based on transcripts from this module and the glycolysis/gluconeogenesis metabolic pathway consistently distinguished a subset of EOPE±FGR samples with an expression signature suggesting modified tissue bioenergetics. We demonstrated that the expression ratios of LDHA/LDHB and PDK1/GOT1 could be used as surrogate indices for the larger panels of genes in identifying this subgroup. CONCLUSIONS: We provide novel evidence for a molecular subphenotype consistent with a glycolytic metabolic shift that occurs more frequently but not universally in placental specimens of EOPE±FGR.


Subject(s)
Placenta Diseases , Pre-Eclampsia , Premature Birth , Humans , Pregnancy , Infant, Newborn , Female , Placenta/metabolism , Fetal Growth Retardation , Transcriptome , Pre-Eclampsia/metabolism , Premature Birth/metabolism , Placenta Diseases/metabolism
7.
Immunohorizons ; 6(6): 334-343, 2022 06 13.
Article in English | MEDLINE | ID: mdl-35697477

ABSTRACT

Cardiac resident macrophages facilitate electrical conduction by interacting with cardiomyocytes via connexin-43 (Cx43) hemichannels. Cx43 is critical for impulse propagation and coordination between muscle contractions. Cardiomyocyte electrophysiology can be altered when coupled with noncardiomyocyte cell types such as M2c tissue-resident macrophages. Using cocultures of murine HL-1 cardiomyocytes and RAW 264.7 macrophages, we examined the hypothesis that cytokine signals, TGF-ß1 and IL-10, upregulate Cx43 expression at points of contact between the two cell types. These cytokine signals maintain the macrophages in an M2c anti-inflammatory phenotype, mimicking cardiac resident macrophages. The electrophysiology of cardiomyocytes was examined using di-8-ANEPPS potentiometric dye, which reflects a change in membrane potential. Greater fluorescence intensity of di-8-ANEPPS occurred in areas where macrophages interacted with cardiomyocytes. Suppressor of cytokine signaling 3 (SOCS3) peptide mimetic downregulated fluorescence of this membrane potentiometric stain. Cx43 expression in cocultures was confirmed by fluorescence microscopy and flow cytometry. Confocal images of these interactions demonstrate the Cx43 hemichannel linkages between the cardiomyocytes and macrophages. These results suggest that TGF-ß1 and IL-10 upregulate Cx43 hemichannels, thus enhancing macrophage-cardiomyocyte coupling, raising the cellular resting membrane potential and leading to a more excitatory cardiomyocyte.


Subject(s)
Connexin 43 , Myocytes, Cardiac , Animals , Connexin 43/genetics , Connexin 43/metabolism , Gap Junctions/metabolism , Interleukin-10/metabolism , Macrophages/metabolism , Membrane Potentials , Mice , Transforming Growth Factor beta/metabolism , Transforming Growth Factor beta1/metabolism
8.
Clin Sci (Lond) ; 135(17): 2049-2066, 2021 09 17.
Article in English | MEDLINE | ID: mdl-34406367

ABSTRACT

Fetal growth restriction (FGR) is a complication of pregnancy that reduces birth weight, markedly increases infant mortality and morbidity and is associated with later-life cardiometabolic disease. No specific treatment is available for FGR. Placentas of human FGR infants have low abundance of sodium-coupled neutral amino acid transporter 2 (Slc38a2/SNAT2), which supplies the fetus with amino acids required for growth. We determined the mechanistic role of placental Slc38a2/SNAT2 deficiency in the development of restricted fetal growth, hypothesizing that placenta-specific Slc38a2 knockdown causes FGR in mice. Using lentiviral transduction of blastocysts with a small hairpin RNA (shRNA), we achieved 59% knockdown of placental Slc38a2, without altering fetal Slc38a2 expression. Placenta-specific Slc38a2 knockdown reduced near-term fetal and placental weight, fetal viability, trophoblast plasma membrane (TPM) SNAT2 protein abundance, and both absolute and weight-specific placental uptake of the amino acid transport System A tracer, 14C-methylaminoisobutyric acid (MeAIB). We also measured human placental SLC38A2 gene expression in a well-defined term clinical cohort and found that SLC38A2 expression was decreased in late-onset, but not early-onset FGR, compared with appropriate for gestational age (AGA) control placentas. The results demonstrate that low placental Slc38a2/SNAT2 causes FGR and could be a target for clinical therapies for late-onset FGR.


Subject(s)
Amino Acid Transport System A/deficiency , Fetal Development , Fetal Growth Retardation/metabolism , Placenta/metabolism , Placentation , Amino Acid Transport System A/genetics , Animals , Case-Control Studies , Female , Fetal Growth Retardation/genetics , Fetal Growth Retardation/physiopathology , Gene Expression Regulation, Developmental , Gene Knockdown Techniques , Placenta/physiopathology , Pregnancy , Prospective Studies , RNA Interference
9.
Front Physiol ; 12: 681632, 2021.
Article in English | MEDLINE | ID: mdl-34276401

ABSTRACT

Preeclampsia (PE) is a multisystemic, pregnancy-specific disorder and a leading cause of maternal and fetal death. PE is also associated with an increased risk for chronic morbidities later in life for mother and offspring. Abnormal placentation or placental function has been well-established as central to the genesis of PE; yet much remains to be determined about the factors involved in the development of this condition. Despite decades of investigation and many clinical trials, the only definitive treatment is parturition. To better understand the condition and identify potential targets preclinically, many approaches to simulate PE in mice have been developed and include mixed mouse strain crosses, genetic overexpression and knockout, exogenous agent administration, surgical manipulation, systemic adenoviral infection, and trophoblast-specific gene transfer. These models have been useful to investigate how biological perturbations identified in human PE are involved in the generation of PE-like symptoms and have improved the understanding of the molecular mechanisms underpinning the human condition. However, these approaches were characterized by a wide variety of physiological endpoints, which can make it difficult to compare effects across models and many of these approaches have aspects that lack physiological relevance to this human disorder and may interfere with therapeutic development. This report provides a comprehensive review of mouse models that exhibit PE-like symptoms and a proposed standardization of physiological characteristics for analysis in murine models of PE.

10.
Sci Rep ; 9(1): 2742, 2019 02 26.
Article in English | MEDLINE | ID: mdl-30808910

ABSTRACT

The placenta is an essential organ that is formed during pregnancy and its proper development is critical for embryonic survival. While several animal models have been shown to exhibit some of the pathological effects present in human preeclampsia, these models often do not represent the physiological aspects that have been identified. Hypoxia-inducible factor 1 alpha (Hif-1α) is a necessary component of the cellular oxygen-sensing machinery and has been implicated as a major regulator of trophoblast differentiation. Elevated levels of Hif-1α in the human placenta have been linked to the development of pregnancy-associated disorders, such as preeclampsia and fetal growth restriction. As oxygen regulation is a critical determinant for placentogenesis, we determined the effects of constitutively active Hif-1α, specifically in trophoblasts, on mouse placental development in vivo. Our research indicates that prolonged expression of trophoblast-specific Hif-1α leads to a significant decrease in fetal birth weight. In addition, we noted significant physiological alterations in placental differentiation that included reduced branching morphogenesis, alterations in maternal and fetal blood spaces, and failure to remodel the maternal spiral arteries. These placental alterations resulted in subsequent maternal hypertension with parturitional resolution and maternal kidney glomeruloendotheliosis with accompanying proteinuria, classic hallmarks of preeclampsia. Our findings identify Hif-1α as a critical molecular mediator of placental development and indicate that prolonged expression of Hif-1α, explicitly in placental trophoblasts causes maternal pathology and establishes a mouse model that significantly recapitulates the physiological and pathophysiological characteristics of preeclampsia with fetal growth restriction.


Subject(s)
Fetal Growth Retardation/pathology , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Kidney/pathology , Placenta/pathology , Placentation , Pre-Eclampsia/pathology , Trophoblasts/metabolism , Animals , Female , Fetal Growth Retardation/metabolism , Kidney/metabolism , Male , Mice , Mice, Inbred C57BL , Mice, Inbred ICR , Placenta/metabolism , Pre-Eclampsia/metabolism , Pregnancy
11.
Int J Stem Cells ; 11(1): 111-120, 2018 May 30.
Article in English | MEDLINE | ID: mdl-29699384

ABSTRACT

BACKGROUND: Transforming growth factor beta (TGF-ß) signaling has been shown to control a large number of critical cellular actions such as cell death, differentiation, and development and has been implicated as a major regulator of placental function. SM10 cells are a mouse placental progenitor cell line, which has been previously shown to differentiate into nutrient transporting, labyrinthine-like cells upon treatment with TGF-ß. However, the signal transduction pathway activated by TGF-ß to induce SM10 progenitor differentiation has yet to be fully investigated. MATERIALS AND METHODS: In this study the SM10 labyrinthine progenitor cell line was used to investigate TGF-ß induced differentiation. Activation of the TGF-ß pathway and the ability of TGF-ß to induce differentiation were investigated by light microscopy, luciferase assays, and Western blot analysis. RESULTS AND CONCLUSIONS: In this report, we show that three isoforms of TGF-ß have the ability to terminally differentiate SM10 cells, whereas other predominant members of the TGF-ß superfamily, Nodal and Activin A, do not. Additionally, we have determined that TGF-ß induced Smad2 phosphorylation can be mediated via the ALK-5 receptor with subsequent transactivation of the Activin response element. Our studies identify an important regulatory signaling pathway in SM10 progenitor cells that is involved in labyrinthine trophoblast differentiation.

12.
Theriogenology ; 107: 115-126, 2018 Feb.
Article in English | MEDLINE | ID: mdl-29145065

ABSTRACT

The placenta is a complex and essential organ composed largely of fetal-derived cells, including several different trophoblast subtypes that work in unison to support nutrient transport to the fetus during pregnancy. Abnormal placental development can lead to pregnancy-associated disorders that often involve metabolic dysfunction. The scope of dysregulated metabolism during placental development may not be fully representative of the in vivo state in defined culture systems, such as cell lines or isolated primary cells. Thus, assessing metabolic function in intact placental tissue would provide a better assessment of placental metabolism. In this study, we describe a methodology for assaying glycolytic function in structurally-intact mouse placental tissue, ex vivo, without culturing or tissue dissociation, that more closely resembles the in vivo state. Additionally, we present data highlighting sex-dependent differences of two mouse strains (C57BL/6 and ICR) in the pre-hypertrophic (E14.5) and hypertrophic (E18.5) placenta. These data establish a foundation for investigation of metabolism throughout gestation and provides a comprehensive assessment of glycolytic function during placental development.


Subject(s)
Glucose/metabolism , Glycolysis/physiology , Placenta/physiology , Animals , Female , Fetal Development , Mice , Mice, Inbred C57BL , Mice, Inbred ICR , Pregnancy
13.
Stem Cells Dev ; 26(11): 808-817, 2017 06 01.
Article in English | MEDLINE | ID: mdl-28335680

ABSTRACT

Placental abnormalities can cause Pregnancy-Associated Disorders, including preeclampsia, intrauterine growth restriction, and placental insufficiency, resulting in complications for both the mother and fetus. Trophoblast cells within the labyrinthine layer of the placenta facilitate the exchange of nutrients, gases, and waste between mother and fetus; therefore, the development of this cell layer is critical for fetal development. As trophoblast cells differentiate, it is assumed their metabolism changes with their energy requirements. We hypothesize that proper regulation of trophoblast metabolism is a key component of normal placental development; therefore, we examined the role of AMP-activated kinase (AMPK, PRKAA1/2), a sensor of cellular energy status. Our previous studies have shown that AMPK knockdown alters both trophoblast differentiation and nutrient transport. In this study, AMPKα1/2 shRNA was used to investigate the metabolic effects of AMPK knockdown on SM10 placental labyrinthine progenitor cells before and after differentiation. Extracellular flux analysis confirmed that AMPK knockdown was sufficient to reduce trophoblast glycolysis, mitochondrial respiration, and ATP coupling efficiency. A reduction in AMPK in differentiated trophoblasts also resulted in increased mitochondrial volume. These data indicate that a reduction in AMPK disrupts cellular metabolism in both progenitors and differentiated placental trophoblasts. This disruption correlates to abortive trophoblast differentiation that may contribute to the development of Pregnancy-Associated Disorders.


Subject(s)
AMP-Activated Protein Kinases/metabolism , Cell Differentiation , Chorionic Villi/metabolism , Energy Metabolism , Gene Knockdown Techniques , Stem Cells/cytology , Stem Cells/enzymology , Adenosine Triphosphate/metabolism , Animals , Cell Line , Cell Respiration , Cell Shape , Cell Size , Female , Glycolysis , Mice , Mitochondria/metabolism , Organelle Size , Pregnancy , Protons
14.
Exp Suppl ; 107: 73-79, 2016.
Article in English | MEDLINE | ID: mdl-27812977

ABSTRACT

AMPK is important in numerous physiological systems but plays a vital role in embryonic and placental development. The placenta is a unique organ that is the essential lifeline between the mother and baby during pregnancy and gestation. During placental development, oxygen concentrations are very low until cells differentiate to establish the appropriate lineages that take on new functions required for placental and embryonic survival. Balancing the oxygen regulatory environment with the demands for energy and need to maintain metabolism during this process places AMPK at the center of maintaining placental cellular homeostasis as it integrates and responds to numerous complex stimuli. AMPK plays a critical role in sensing metabolic and energy changes. Once activated, it turns on pathways that produce energy and shuts down catabolic processes. AMPK coordinates cell growth, differentiation, and nutrient transport to maintain cell survival. Appropriate regulation of AMPK is essential for normal placental and embryonic development, and its dysregulation may lead to pregnancy-associated disorders such as intrauterine growth restriction, placental insufficiency, or preeclampsia.


Subject(s)
AMP-Activated Protein Kinases/genetics , Oxygen/metabolism , Placenta/enzymology , Placentation/genetics , Stem Cells/enzymology , AMP-Activated Protein Kinases/metabolism , Cell Differentiation , Cell Proliferation , Female , Fetal Growth Retardation/enzymology , Fetal Growth Retardation/etiology , Fetal Growth Retardation/genetics , Fetal Growth Retardation/pathology , Gene Expression Regulation , Humans , Placenta/cytology , Placental Insufficiency/enzymology , Placental Insufficiency/etiology , Placental Insufficiency/genetics , Placental Insufficiency/pathology , Pre-Eclampsia/enzymology , Pre-Eclampsia/etiology , Pre-Eclampsia/genetics , Pre-Eclampsia/pathology , Pregnancy , Pregnancy Complications/enzymology , Pregnancy Complications/genetics , Pregnancy Complications/pathology , Protein Subunits/genetics , Protein Subunits/metabolism , Signal Transduction , Stem Cells/pathology
15.
Stem Cells Dev ; 25(13): 959-74, 2016 07 01.
Article in English | MEDLINE | ID: mdl-27168216

ABSTRACT

The placenta is an organ that is formed transiently during pregnancy, and appropriate placental development is necessary for fetal survival and growth. Proper differentiation of the labyrinthine layer of the placenta is especially crucial, as it establishes the fetal-maternal interface that is involved in physiological exchange processes. Although previous studies have indicated the importance of inhibitor of differentiation/inhibitor of DNA binding-2 (Id2) helix-loop-helix transcriptional regulator in mediating cell differentiation, the ability of Id2 to regulate differentiation toward the labyrinthine (transport) lineage of the placenta has yet to be determined. In the current study, we have generated labyrinthine trophoblast progenitor cells with increased (SM10-Id2) or decreased (SM10-Id2-shRNA) Id2 expression and determined the effect on TGF-ß-induced differentiation. Our Id2 overexpression and knockdown analyses indicate that Id2 mediates TGF-ß-induced morphological differentiation of labyrinthine trophoblast cells, as Id2 overexpression prevents differentiation and Id2 knockdown results in differentiation. Thus, our data indicate that Id2 is an important molecular mediator of labyrinthine trophoblast differentiation. An understanding of the regulators of trophoblast progenitor differentiation toward the labyrinthine lineage may offer insights into events governing pregnancy-associated disorders, such as placental insufficiency, fetal growth restriction, and preeclampsia.


Subject(s)
Cell Differentiation , Inhibitor of Differentiation Protein 2/metabolism , Placenta/cytology , Stem Cells/cytology , Animals , Biomarkers/metabolism , Cell Differentiation/drug effects , Cell Differentiation/genetics , Cell Line , Cell Shape/drug effects , Clone Cells , Down-Regulation/drug effects , Female , Gene Knockdown Techniques , Inhibitor of Differentiation Protein 2/genetics , Mice , Pregnancy , RNA, Messenger/genetics , RNA, Messenger/metabolism , Stem Cells/drug effects , Transcription, Genetic/drug effects , Transforming Growth Factor beta/pharmacology
16.
J Cell Death ; 8: 1-7, 2015.
Article in English | MEDLINE | ID: mdl-25922583

ABSTRACT

Apoptosis is physiological cell death required for the cellular maintenance of homeostasis, and caspases play a major role in the execution of this process. Numerous disorders occur when levels of apoptosis within an organism are excessive, and several studies have explored the possibility of using caspase inhibitors to prevent these disorders. Q-VD-OPh (quinolyl-valyl-O-methylaspartyl-[2,6-difluorophenoxy]-methyl ketone), a novel pan caspase inhibitor, has been used because of its efficacy to inhibit apoptosis at low concentrations, its ability to cross the blood-brain barrier, as well as being nontoxic in vivo. This review examines Q-VD-OPh's ability to inhibit apoptosis in several animal models of human disease.

17.
Theriogenology ; 82(7): 1043-8, 2014 Oct 15.
Article in English | MEDLINE | ID: mdl-25110063

ABSTRACT

The placenta is a unique and highly complex organ that develops only during pregnancy and is essential for growth and survival of the developing fetus. The placenta provides the vital exchange of gases and wastes, the necessary nutrients for fetal development, acts as immune barrier that protects against maternal rejection, and produces numerous hormones and growth factors that promote fetal maturity to regulate pregnancy until parturition. Abnormal placental development is a major underlying cause of pregnancy-associated disorders that often result in preterm birth. Defects in placental stem cell propagation, growth, and differentiation are the major factors that affect embryonic and fetal well-being and dramatically increase the risk of pregnancy complications. Understanding the processes that regulate placentation is important in determining the underlying factors behind abnormal placental development. The ability to manipulate genes in a placenta-specific manner provides a unique tool to analyze development and eliminates potentially confounding results that can occur with traditional gene knockouts. Trophoblast stem cells and mouse embryos are not overly amenable to traditional gene transfer techniques. Most viral vectors, however, have a low infection rate and often lead to mosaic transgenesis. Although the traditional method of embryo transfer is intrauterine surgical implantation, the methodology reported here, combining lentiviral blastocyst infection and nonsurgical embryo transfer, leads to highly efficient and placental-specific gene transfer. Numerous advantages of our optimized procedures include increased investigator safety, a reduction in animal stress, rapid and noninvasive embryo transfer, and higher a rate of pregnancy and live birth.


Subject(s)
Embryo Transfer/veterinary , Gene Expression Regulation, Developmental/physiology , Gene Transfer Techniques , Placenta/metabolism , Animals , Blastocyst/physiology , Embryo Transfer/methods , Female , Green Fluorescent Proteins/metabolism , Male , Mice , Mice, Inbred C57BL , Pregnancy
18.
Stem Cells Dev ; 23(23): 2921-30, 2014 Dec 01.
Article in English | MEDLINE | ID: mdl-25003940

ABSTRACT

The placenta is a transient organ that develops upon the initiation of pregnancy and is essential for embryonic development and fetal survival. The rodent placenta consists of distinct lineages and includes cell types that are analogous to those that make up the human placenta. Trophoblast cells within the labyrinth layer, which lies closest to the fetus, fuse and come in contact with maternal blood, thus facilitating nutrient and waste exchange between the mother and the baby. Abnormalities of the placenta may occur as a result of cellular stress and have been associated with pregnancy-associated disorders: such as preeclampsia, intrauterine growth restriction, and placental insufficiency. Cellular stress has also been shown to alter proliferation and differentiation rates of trophoblast cells. This stress response is important for cell survival and ensures continued placental functionality. AMP-activated protein kinase is an important sensor of cellular metabolism and stress. To study the role of AMPK in the trophoblast cells, we used RNA interference to simultaneously knockdown levels of both the AMPK alpha isoforms, AMPKα1 and AMPKα2. SM10 trophoblast progenitor cells were transduced with AMPKα1/2 shRNA and stable clones were established to analyze the effects of AMPK knockdown on important cellular functions. Our results indicate that a reduction in AMPK levels causes alterations in cell morphology, growth rate, and nutrient transport, thus identifying an important role for AMPK in the regulation of placental trophoblast differentiation.


Subject(s)
AMP-Activated Protein Kinases/metabolism , Cell Differentiation/physiology , Cell Proliferation/physiology , Stem Cells/enzymology , Trophoblasts/enzymology , AMP-Activated Protein Kinases/genetics , Animals , Gene Knockdown Techniques , Mice , Stem Cells/cytology , Trophoblasts/cytology
19.
Cell Physiol Biochem ; 31(2-3): 257-76, 2013.
Article in English | MEDLINE | ID: mdl-23466888

ABSTRACT

BACKGROUND/AIMS: Chelerythrine [CET], a protein kinase C [PKC] inhibitor, is a prop-apoptotic BH3-mimetic binding to BH1-like motifs of Bcl-2 proteins. CET action was examined on PKC phosphorylation-dependent membrane transporters (Na+/K+ pump/ATPase [NKP, NKA], Na+-K+-2Cl+ [NKCC] and K+-Cl- [KCC] cotransporters, and channel-supported K+ loss) in human lens epithelial cells [LECs]. METHODS: K+ loss and K+ uptake, using Rb+ as congener, were measured by atomic absorption/emission spectrophotometry with NKP and NKCC inhibitors, and Cl- replacement by NO3ˉ to determine KCC. 3H-Ouabain binding was performed on a pig renal NKA in the presence and absence of CET. Bcl-2 protein and NKA sequences were aligned and motifs identified and mapped using PROSITE in conjunction with BLAST alignments and analysis of conservation and structural similarity based on prediction of secondary and crystal structures. RESULTS: CET inhibited NKP and NKCC by >90% (IC50 values ~35 and ~15 µM, respectively) without significant KCC activity change, and stimulated K+ loss by ~35% at 10-30 µM. Neither ATP levels nor phosphorylation of the NKA α1 subunit changed. 3H-ouabain was displaced from pig renal NKA only at 100 fold higher CET concentrations than the ligand. Sequence alignments of NKA with BH1- and BH3-like motifs containing pro-survival Bcl-2 and BclXl proteins showed more than one BH1-like motif within NKA for interaction with CET or with BH3 motifs. One NKA BH1-like motif (ARAAEILARDGPN) was also found in all P-type ATPases. Also, NKA possessed a second motif similar to that near the BH3 region of Bcl-2. CONCLUSION: Findings support the hypothesis that CET inhibits NKP by binding to BH1-like motifs and disrupting the α1 subunit catalytic activity through conformational changes. By interacting with Bcl-2 proteins through their complementary BH1- or BH3-like-motifs, NKP proteins may be sensors of normal and pathological cell functions, becoming important yet unrecognized signal transducers in the initial phases of apoptosis. CET action on NKCC1 and K+ channels may involve PKC-regulated mechanisms; however, limited sequence homologies to BH1-like motifs cannot exclude direct effects.


Subject(s)
Anti-Bacterial Agents/pharmacology , Apoptosis/drug effects , Benzophenanthridines/pharmacology , Proto-Oncogene Proteins c-bcl-2/chemistry , Sodium-Potassium-Exchanging ATPase/metabolism , Adenosine Triphosphate/metabolism , Amino Acid Motifs , Amino Acid Sequence , Animals , Cell Line , Humans , Molecular Sequence Data , Ouabain/pharmacology , Phosphorylation , Potassium/metabolism , Protein Structure, Tertiary , Proto-Oncogene Proteins c-bcl-2/metabolism , Rubidium/metabolism , Sequence Alignment , Sodium-Potassium-Chloride Symporters/chemistry , Sodium-Potassium-Chloride Symporters/metabolism , Sodium-Potassium-Exchanging ATPase/antagonists & inhibitors , Swine , Tritium/chemistry
20.
J RNAi Gene Silencing ; 8: 470-8, 2012.
Article in English | MEDLINE | ID: mdl-23316259

ABSTRACT

AMP-activated protein kinase (AMPK) is a master metabolic regulator that responds to the AMP: ATP ratio and promotes ATP production when the cell is low on energy. There are two isoforms of the catalytic alpha subunit, AMPKα1 and AMPKα2. Here, we describe the production of a small interfering RNA (siRNA) and a short hairpin RNA (shRNA) targeting both catalytic isoforms of AMPK in human, mouse, and rat. Multiple loop sequences were tested to generate the most effective shRNA. The shRNA causes significant knockdown of both isoforms of AMPKα in mouse and human cells. The shRNA effectively knocked down AMPKα1 and AMPKα2 protein levels, compared to a five basepair mismatch-control shRNA in mouse fibroblast NIH3T3 cells and significantly knocked down AMPKα1 (63%) and AMPKα2 (72%) levels compared to control in human embryonic kidney cells, HEK293s. The shRNA also causes a significant reduction in AMPK activity, measured as phosphorylation of acetyl-CoA carboxylase (ACC), a direct phosphorylation target. While the protein levels of total ACC remained the same between the AMPKα1and α2 shRNA and control shRNA-treated cells, there was a 41% reduction in phospho-ACC protein levels. The generation of this AMPKα1and α2 shRNA can be used to stably knock down protein levels and activity of both catalytic isoforms of AMPK in different species to assess function.

SELECTION OF CITATIONS
SEARCH DETAIL
...