Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 6 de 6
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Biol Reprod ; 110(5): 950-970, 2024 May 09.
Artigo em Inglês | MEDLINE | ID: mdl-38330185

RESUMO

Research on the biology of fetal-maternal barriers has been limited by access to physiologically relevant cells, including trophoblast cells. In this study, we describe the development of a human term placenta-derived cytotrophoblast immortalized cell line (hPTCCTB) derived from the basal plate. Human-term placenta-derived cytotrophoblast immortalized cell line cells are comparable to their primary cells of origin in terms of morphology, marker expression, and functional responses. We demonstrate that these can transform into syncytiotrophoblast and extravillous trophoblasts. We also compared the hPTCCTB cells to immortalized chorionic trophoblasts (hFM-CTC), trophoblasts of the chorionic plate, and BeWo cells, choriocarcinoma cell lines of conventional use. Human-term placenta-derived cytotrophoblast immortalized cell line and hFM-CTCs displayed more similarity to each other than to BeWos, but these differ in syncytialization ability. Overall, this study (1) demonstrates that the immortalized hPTCCTB generated are cells of higher physiological relevance and (2) provides a look into the distinction between the spatially distinct placental and fetal barrier trophoblasts cells, hPTCCTB and hFM-CTC, respectively.


Assuntos
Placenta , Trofoblastos , Humanos , Trofoblastos/citologia , Trofoblastos/fisiologia , Feminino , Gravidez , Placenta/citologia , Placenta/fisiologia , Linhagem Celular
2.
Front Immunol ; 14: 1196453, 2023.
Artigo em Inglês | MEDLINE | ID: mdl-37600782

RESUMO

Background: Fetal inflammatory response mediated by the influx of immune cells and activation of pro-inflammatory transcription factor NF-κB in feto-maternal uterine tissues is the major determinant of infection-associated preterm birth (PTB, live births < 37 weeks of gestation). Objective: To reduce the incidence of PTB by minimizing inflammation, extracellular vesicles (EVs) were electroporetically engineered to contain anti-inflammatory cytokine interleukin (IL)-10 (eIL-10), and their efficacy was tested in an ascending model of infection (vaginal administration of E. coli) induced PTB in mouse models. Study design: EVs (size: 30-170 nm) derived from HEK293T cells were electroporated with recombinant IL-10 at 500 volts and 125 Ω, and 6 pulses to generate eIL-10. eIL-10 structural characters (electron microscopy, nanoparticle tracking analysis, ExoView [size and cargo content] and functional properties (co-treatment of macrophage cells with LPS and eIL-10) were assessed. To test efficacy, CD1 mice were vaginally inoculated with E. coli (1010CFU) and subsequently treated with either PBS, eIL-10 (500ng) or Gentamicin (10mg/kg) or a combination of eIL-10+gentamicin. Fetal inflammatory response in maternal and fetal tissues after the infection or treatment were conducted by suspension Cytometer Time of Flight (CyTOF) using a transgenic mouse model that express red fluorescent TdTomato (mT+) in fetal cells. Results: Engineered EVs were structurally and functionally stable and showed reduced proinflammatory cytokine production from LPS challenged macrophage cells in vitro. Maternal administration of eIL-10 (10 µg/kg body weight) crossed feto-maternal barriers to delay E. coli-induced PTB to deliver live pups at term. Delay in PTB was associated with reduced feto-maternal uterine inflammation (immune cell infiltration and histologic chorioamnionitis, NF-κB activation, and proinflammatory cytokine production). Conclusions: eIL-10 administration was safe, stable, specific, delayed PTB by over 72 hrs and delivered live pups. The delivery of drugs using EVs overcomes the limitations of in-utero fetal interventions. Protecting IL-10 in EVs eliminates the need for the amniotic administration of recombinant IL-10 for its efficacy.


Assuntos
Vesículas Extracelulares , Interleucina-10 , Complicações Infecciosas na Gravidez , Animais , Feminino , Humanos , Camundongos , Gravidez , Citocinas , Modelos Animais de Doenças , Escherichia coli , Feto , Células HEK293 , Interleucina-10/farmacologia , Lipopolissacarídeos , NF-kappa B , Nascimento Prematuro , Proteínas Recombinantes/farmacologia , Inflamação , Complicações Infecciosas na Gravidez/tratamento farmacológico
3.
Placenta ; 135: 33-42, 2023 04.
Artigo em Inglês | MEDLINE | ID: mdl-36913807

RESUMO

INTRODUCTION: During pregnancy, the growth of the fetus is supported by the exchange of nutrients, waste, and other molecules between maternal and fetal circulations in the utero-placental unit. Nutrient transfer, in particular, is mediated by solute transporters such as solute carrier (SLC) and adenosine triphosphate-binding cassette (ABC) proteins. While nutrient transport has been extensively studied in the placenta, the role of human fetal membranes (FM), which was recently reported to have a role in drug transport, in nutrient uptake remains unknown. OBJECTIVES: This study determined nutrient transport expression in human FM and FM cells and compared expression with placental tissues and BeWo cells. METHODS: RNA sequencing (RNA-Seq) of placental and FM tissues and cells was done. Genes of major solute transporter groups, such as SLC and ABC, were identified. Proteomic analysis of cell lysates was performed via nano-liquid chromatography-tandem mass spectrometry (nanoLC-MS/MS) to confirm expression at a protein level. RESULTS: We determined that FM tissues and cells derived from the fetal membrane tissues express nutrient transporter genes, and their expression is similar to that seen in the placenta or BeWo cells. In particular, transporters involved in macronutrient and micronutrient transfer were identified in both placental and FM cells. Consistent with RNA-Seq findings, carbohydrate transporters (3), vitamin transport-related proteins (8), amino acid transporters (21), fatty acid transport-related proteins (9), cholesterol transport-related proteins (6) and nucleoside transporters (3) were identified in BeWo and FM cells, with both groups sharing similar nutrient transporter expression. CONCLUSION: This study determined the expression of nutrient transporters in human FMs. This knowledge is the first step in improving our understanding of nutrient uptake kinetics during pregnancy. Functional studies are required to determine the properties of nutrient transporters in human FMs.


Assuntos
Placenta , Espectrometria de Massas em Tandem , Gravidez , Feminino , Humanos , Placenta/metabolismo , Proteômica , Proteínas de Transporte , Sistemas de Transporte de Aminoácidos/metabolismo , Nutrientes
4.
Clin Sci (Lond) ; 136(22): 1591-1614, 2022 11 30.
Artigo em Inglês | MEDLINE | ID: mdl-36250628

RESUMO

Oxidative stress (OS) induced activation of p38 mitogen-activated kinase (MAPK) and cell fate from p38 signaling was tested using the human fetal membrane's amnion epithelial cells (AEC). We created p38 KO AEC using the CRISPR/Cas9 approach and tested cell fate in response to OS on an AEC-free fetal membrane extracellular matrix (ECM). Screening using image CyTOF indicated OS causing epithelial-mesenchymal transition (EMT). Further testing revealed p38 deficiency prevented AEC senescence, EMT, cell migration, and inflammation. To functionally validate in vitro findings, fetal membrane-specific conditional KO (cKO) mice were developed by injecting Cre-recombinase encoded exosomes intra-amniotically into p38αloxP/loxP mice. Amnion membranes from p38 cKO mice had reduced senescence, EMT, and increased anti-inflammatory IL-10 compared with WT animals. Our study suggested that overwhelming activation of p38 in response to OS inducing risk exposures can have an adverse impact on cells, cause cell invasion, inflammation, and ECM degradation detrimental to tissue homeostasis.


Assuntos
Mitógenos , Proteínas Quinases p38 Ativadas por Mitógeno , Humanos , Camundongos , Animais , Proteínas Quinases p38 Ativadas por Mitógeno/metabolismo , Células Epiteliais/metabolismo , Âmnio , Inflamação/metabolismo
5.
J Assoc Physicians India ; 70(9): 11-12, 2022 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-36082888

RESUMO

BACKGROUND: Worldwide, obesity has nearly tripled since 1975 and has become a major healthcare challenge today. Intermittent fasting (IF) is gaining popularity as a weight loss strategy in recent times. This study aimed to study the role of IF as a modern-day weight-loss strategy in obese adults through a real-world pilot experiment conducted at a nutrition clinic in Mumbai. METHODOLOGY: To understand the effects of IF on weight loss, 32 overweight/obese [body mass index (BMI) ≥23 kg/m2] adults from a nutrition clinic in Mumbai, were assigned consecutively to an IF plan and followed up for 3 months. Their demographic, anthropometric, and dietary assessments were done pre- and post-intervention. Qualitative interviews were done at the end of the study to record the participants' overall well-being, experience, and sustainability of IF. RESULTS: 56% of study participants were males and their mean age was 35.6 ± 8.9 years. 65.6% of participants were able to maintain 14-16 hours of fasting and 53% managed all 7 days of IF. Analysis of post-intervention data showed a significant reduction in mean body weight (88.5 ± 19 to 83.8 ± 17.6 kg), waist circumference (M: 108.2 ± 11.3 to 103.6 ± 4.4 cm, F: 98.9 ± 8.8 to 93.3 ± 3.3 cm), BMI (31.4 ± 5.3 to 29.6 ± 5.1 kg/m2), daily calories (1782 ± 237 to 1388 ± 243 kcal/day), carbohydrate intake (267 ± 18.4 to 164 ± 4.0 g/day), and an increase in protein intake (39 ± 11 to 55 ± 11 g/day). Participants reported positive experiences of practicing IF such as improved fitness, sleep cycle, and adoption of healthy eating habits. CONCLUSION: The study demonstrates that IF could aid in weight loss and adoption of a healthier lifestyle.


Assuntos
Jejum , Obesidade , Adulto , Índice de Massa Corporal , Feminino , Humanos , Masculino , Obesidade/terapia , Sobrepeso , Redução de Peso
6.
Life Sci ; 307: 120867, 2022 Oct 15.
Artigo em Inglês | MEDLINE | ID: mdl-35940219

RESUMO

BACKGROUND: Environmental exposure to toxicants is a major risk factor for spontaneous preterm birth (PTB, <37 weeks). Toxicants and drugs administered to patients are metabolized primarily by the cytochrome P450 (CYP450) system. Along with the adult and fetal liver, the placenta, a critical feto-maternal interface organ, expresses CYP450 enzymes that metabolize these xenobiotics. However, the contribution of the fetal membranes, another tissue of the feto-maternal interface, to the expression of CYP450 enzymes and the detoxification of xenobiotics remains unknown. AIMS: This study characterized CYP450 expression and determined the functional activity of CYP450 enzymes in fetal membranes. MAIN METHODS: RNA sequencing (RNA-Seq) of placental and fetal membrane tissues and cells was done. Differential expressions of CYP450 genes were compared and validated via reverse transcription-quantitative polymerase chain reaction (RT-qPCR) between the two tissues. The functional activity of major CYP450 enzymes was determined using a fluorophore-based enzymatic assay in the presence and absence of their corresponding inhibitors. KEY FINDINGS: With the exception of genes that regulate cholesterol metabolism, the expression profile of CYP450 genes was similar between placental and fetal membranes tissues/cells. RT-qPCR analysis confirmed these findings with significant levels of mRNA for major CYP450 genes being detectable in amnion epithelial cells (AECs) and chorion trophoblasts cells (CTCs). Biochemical analyses revealed significant CYP450 enzymatic activities that were sensitive to specific inhibitors for both AECs and CTCs, suggesting that the genes were expressed as functional enzymes. SIGNIFICANCE: This is the first study to determine global expression of CYP450 enzymes in fetal membranes which may play a role in xenobiotic metabolism during pregnancy. Given that many women are exposed to environmental toxins or require medications during pregnancy, a better understanding of their role in metabolism is required to develop safer therapeutics and prevent adverse outcomes.


Assuntos
Nascimento Prematuro , Xenobióticos , Adulto , Colesterol/metabolismo , Sistema Enzimático do Citocromo P-450/genética , Sistema Enzimático do Citocromo P-450/metabolismo , Membranas Extraembrionárias/química , Membranas Extraembrionárias/metabolismo , Feminino , Humanos , Recém-Nascido , Placenta/metabolismo , Gravidez , RNA Mensageiro/metabolismo , Xenobióticos/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...