Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 102
Filtrar
1.
PLoS One ; 19(3): e0300461, 2024.
Artigo em Inglês | MEDLINE | ID: mdl-38512862

RESUMO

Polycystic ovary syndrome (PCOS), a common endocrine and metabolic disorder affecting women in their reproductive years. Emerging evidence suggests that the maternal-fetal immune system is crucial for proper pregnancy. However, whether immune function is altered at the end of pregnancy in PCOS women and the underlying molecular mechanisms is currently unexplored. Herein, the basic maternal immune system was investigated (n = 136 in the control group; n = 103 in the PCOS group), and whole-transcriptome sequencing was carried out to quantify the mRNAs, miRNAs, and lncRNAs expression levels in fetal side placental tissue of women with PCOS. GO, KEGG, and GSEA analysis were employed for functional enrichment analysis. The process of identifying hub genes was conducted utilizing the protein-protein interaction network. CIBERSORT and Connectivity Map were deployed to determine immune cell infiltration and predict potential drugs, respectively. A network of mRNA-miRNA-lncRNA was constructed and then validated by qRT-PCR. First, red blood cell count, neutrophil count, lymphocyte count, hypersensitive C-reactive protein, and procalcitonin were significantly elevated, while placental growth factor was hindered in PCOS women. We identified 308 DEmRNAs, 77 DEmiRNAs, and 332 DElncRNAs in PCOS samples. Functional enrichment analysis revealed that there were significant changes observed in terms of the immune system, especially the chemokine pathway. Eight genes, including FOS, JUN, EGR1, CXCL10, CXCR1, CXCR2, CXCL11, and CXCL8, were considered as hub genes. Furthermore, the degree of infiltration of neutrophils was dramatically decreased in PCOS tissues. In total, 57 ceRNA events were finally obtained, and immune-related ceRNA networks were validated. Some potential drug candidates, such as enalapril and RS-100329, could have a function in PCOS therapy. This study represents the inaugural attempt to evaluate the immune system at the end of pregnancy and placental ceRNA networks in PCOS, indicating alterations in the chemokine pathway, which may impact fetal and placental growth, and provides new therapy targets.


Assuntos
MicroRNAs , Síndrome do Ovário Policístico , RNA Longo não Codificante , Humanos , Feminino , Gravidez , Síndrome do Ovário Policístico/genética , Síndrome do Ovário Policístico/metabolismo , 60414 , Placenta/metabolismo , Fator de Crescimento Placentário/genética , MicroRNAs/genética , MicroRNAs/metabolismo , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Quimiocinas/genética , RNA Longo não Codificante/genética , Redes Reguladoras de Genes
2.
Aging (Albany NY) ; 15(19): 10117-10132, 2023 10 02.
Artigo em Inglês | MEDLINE | ID: mdl-37787982

RESUMO

OBJECTIVE: Placental growth factor (PlGF), an important polypeptide hormone, plays an important regulatory role in various physiological processes. Observational studies have shown that PlGF is associated with the risk of coronary heart disease (CHD). However, the causal association between PlGF and CHD is unclear at present. This study aimed to investigate the causal association between genetically predicted PlGF levels and CHD. METHODS: Single nucleotide polymorphisms (SNPs) associated with PlGF were selected as instrumental variables (IVs) to evaluate the causal association between genetically predicted circulating PlGF levels and CHD risk by two-sample Mendelian randomization (MR). RESULTS: Inverse variance weighted (IVW) analysis showed that there was a suggestive causal association between genetically predicted PlGF level and the risk of CHD (OR = 0.79, 95% CI: 0.66-0.95, P = 0.011) overall. In addition, PlGF levels had a significant negative causal association with the risk of myocardial infarction (OR = 0.83, 95% CI: 0.72-0.95, P = 0.007). A negative correlation trend was found between PlGF level and the risk of angina pectoris (OR = 0.89, 95% CI: 0.79-1.01, P = 0.067). In addition, PlGF levels had a significant negative association with the risk of unstable angina pectoris (OR = 0.78, 95% CI: 0.64-0.94, P = 0.008). PlGF levels were negatively correlated with CHD events with suggestive significance (OR = 0.89, 95% CI: 0.80-0.99, P = 0.046). CONCLUSION: Genetically predicted circulating PlGF levels are causally associated with the risk of CHD, especially acute coronary syndrome, and PlGF is a potential therapeutic target for CHD.


Assuntos
Doença das Coronárias , Infarto do Miocárdio , Feminino , Humanos , Fator de Crescimento Placentário/genética , Análise da Randomização Mendeliana , Doença das Coronárias/genética , Angina Pectoris , Polimorfismo de Nucleotídeo Único , Estudo de Associação Genômica Ampla
3.
Taiwan J Obstet Gynecol ; 62(3): 457-460, 2023 May.
Artigo em Inglês | MEDLINE | ID: mdl-37188454

RESUMO

OBJECTIVE: We present low-level mosaic trisomy 9 at amniocentesis associated with a positive non-invasive prenatal testing (NIPT) for trisomy 9, maternal uniparental disomy (UPD) 9, intrauterine growth restriction (IUGR) and a favorable fetal outcome in a pregnancy. CASE REPORT: A 41-year-old, gravida 3, para 0, woman underwent amniocentesis at 18 weeks of gestation because of NIPT at 10 weeks of gestation suspicious of trisomy 9 in the fetus. This pregnancy was conceived by in vitro fertilization (IVF). Amniocentesis revealed a karyotype of 47,XY,+9 [2]/46,XY[23]. Simultaneous array comparative genomic hybridization (aCGH) analysis on the DNA extracted from uncultured amniocytes revealed arr (1-22) × 2, (X,Y) × 1 and detected no genomic imbalance. Polymorphic DNA marker analysis showed maternal uniparental heterodisomy 9 in the amniocytes. Prenatal ultrasound was normal. The woman was referred for genetic counseling at 22 weeks of gestation. The soluble fms-like tyrosine kinase (sFlt)/placental growth factor (PlGF) = 13.1 (normal < 38). There was no gestational hypertension. Continuing the pregnancy was advised. No repeat amniocentesis was performed because of persistent irregular contractions. IUGR was noted. A 2156-g phenotypically normal baby was delivered at 37 weeks of gestation. The cord blood and umbilical cord had a karyotype of 46,XY (40/40 cells). The placenta had a karyotype of 47,XY,+9 (40/40 cells). The parental karyotypes were normal. Quantitative fluorescence polymerase chain reaction (QF-PCR) on the DNA extracted from parental bloods, cord blood, umbilical cord and placenta revealed maternal uniparental heterodisomy 9 in cord blood and umbilical cord, and trisomy 9 of maternal origin in placenta. When follow-up at age three months, the neonate was normal in development and phenotype. The buccal mucosal cells had 3% (3/101 cells) mosaicism for trisomy 9 by interphase fluorescent in situ hybridization (FISH) analysis. CONCLUSION: Mosaic trisomy 9 at prenatal diagnosis should alert the possibility of UPD 9 and include a UPD 9 testing. Low-level mosaic trisomy 9 at amniocentesis can be associated with UPD 9 and a favorable fetal outcome.


Assuntos
Amniocentese , Dissomia Uniparental , Gravidez , Feminino , Humanos , Dissomia Uniparental/diagnóstico , Dissomia Uniparental/genética , Retardo do Crescimento Fetal/diagnóstico , Retardo do Crescimento Fetal/genética , Hibridização in Situ Fluorescente , Hibridização Genômica Comparativa , Fator de Crescimento Placentário/genética , Trissomia/diagnóstico , Trissomia/genética , Feto , Mosaicismo
4.
Neurobiol Aging ; 124: 11-17, 2023 04.
Artigo em Inglês | MEDLINE | ID: mdl-36680854

RESUMO

The vascular endothelial growth factor (VEGF) family of genes has been implicated in the clinical development of Alzheimer's Disease (AD). A previous study identified associations between gene expression of VEGF family members in the prefrontal cortex and cognitive performance and AD pathology. This study explored if those associations were also observed in the blood. Consistent with previous observations in brain tissue, higher blood gene expression of placental growth factor (PGF) was associated with a faster rate of memory decline (p=0.04). Higher protein abundance of FMS-related receptor tyrosine kinase 4 (FLT4) in blood was associated with biomarker levels indicative of lower amyloid and tau pathology, opposite the direction observed in brain. Also, higher gene expression of VEGFB in blood was associated with better baseline memory (p=0.008). Notably, we observed that higher gene expression of VEGFB in blood was associated with lower expression of VEGFB in the brain (r=-0.19, p=0.02). Together, these results suggest that the VEGFB, FLT4, and PGF alterations in the AD brain may be detectable in the blood compartment.


Assuntos
Doença de Alzheimer , Disfunção Cognitiva , Humanos , Feminino , Fator A de Crescimento do Endotélio Vascular/genética , Fator A de Crescimento do Endotélio Vascular/metabolismo , Fator de Crescimento Placentário/genética , Fatores de Crescimento do Endotélio Vascular , Doença de Alzheimer/genética , Doença de Alzheimer/patologia , Biomarcadores , Cognição , Peptídeos beta-Amiloides , Proteínas tau/genética
5.
Ophthalmic Genet ; 44(2): 133-138, 2023 04.
Artigo em Inglês | MEDLINE | ID: mdl-36193031

RESUMO

BACKGROUND: Primary congenital glaucoma (PCG) is characterized by developmental abnormalities of the anterior chamber angle. Although several genes have been associated with PCG, pathogenic mutations could only be detected in about 20% of Chinese patients. GLC3B (1p36.2-36.1) and GLC3C (14q24.3) loci were previously identified in PCG pedigrees via linkage analysis. However, no causative genes were reported in these loci. This study was designed to search for novel PCG-related genes in these genetic regions. MATERIALS AND METHODS: DNA samples from 100 PCG patients and 200 normal controls were pooled and sequenced using a customized panel of 133 positional candidate genes located around GLC3B and GLC3C loci (±1Mb). PCG-related genes were prioritized by the distribution of variants between patients and controls. Confirmation of selected variants and co-segregation analysis were performed using Sanger sequencing. RESULTS: Patient and control group contained 116 and 147 rare variants respectively after screening. Three genes (ZC2HC1C, VPS13D, and PGF) were prioritized according to the distribution of variants between the two groups. Rare variants of PGF were only identified in PCG patients. CONCLUSIONS: To the best of our knowledge, this is the first study aiming at exploring novel PCG-related genes at GLC3B and GLC3C loci. Our preliminary results suggest that there are potential associations between ZC2HC1C, VPS13D, PGF, and PCG. However, larger cohort studies and functional assays are required to provide further evidence for the proposed genotype-phenotype association.


Assuntos
Glaucoma , Sequenciamento de Nucleotídeos em Larga Escala , Fator de Crescimento Placentário , Humanos , População do Leste Asiático , Glaucoma/genética , Glaucoma/congênito , Mutação , Proteínas/genética , Fator de Crescimento Placentário/genética
6.
Bull Exp Biol Med ; 174(1): 174-178, 2022 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-36437340

RESUMO

We studied regulation of the expression of placental growth factor (PlGF) that plays an important role in the trophoblast cells functions and reduced production of which by the placenta is associated with gestational complications. PlGF expression is regulated by transcription factors whose activity is controlled by sumoylation, which is also necessary for the formation of an adequate cellular response to hypoxia. Increased sumoylation and reduced expression of some miRNA targeted to transcription factors VEGF, GCM-1, and UBC9 conjugating SUMO with targets protein were detected in the placenta. Correlations were revealed between changes in the expression of miR-423-3p and miR-652-3p, the level of SUMO 1-4 and UBC9 in the placenta, reduced concentration of PlGF, and increased sFlt-1/PlGF ratio in the blood of pregnant women with early-onset preeclampsia, which attests to the presence of a regulatory mechanism along the axis of miR-652-3p/SUMO-2/3/4/UBC9/GCM-1/PlGF.


Assuntos
MicroRNAs , Gravidez , Feminino , Humanos , Fator de Crescimento Placentário/genética , MicroRNAs/genética
7.
Nat Commun ; 13(1): 6292, 2022 10 22.
Artigo em Inglês | MEDLINE | ID: mdl-36272973

RESUMO

Pancreatic ductal adenocarcinoma (PDAC) has a poor 5-year overall survival rate. Patients with PDAC display limited benefits after undergoing chemotherapy or immunotherapy modalities. Herein, we reveal that chemotherapy upregulates placental growth factor (PlGF), which directly activates cancer-associated fibroblasts (CAFs) to induce fibrosis-associated collagen deposition in PDAC. Patients with poor prognosis have high PIGF/VEGF expression and an increased number of PIGF/VEGF receptor-expressing CAFs, associated with enhanced collagen deposition. We also develop a multi-paratopic VEGF decoy receptor (Ate-Grab) by fusing the single-chain Fv of atezolizumab (anti-PD-L1) to VEGF-Grab to target PD-L1-expressing CAFs. Ate-Grab exerts anti-tumor and anti-fibrotic effects in PDAC models via the PD-L1-directed PlGF/VEGF blockade. Furthermore, Ate-Grab synergizes with gemcitabine by relieving desmoplasia. Single-cell RNA sequencing identifies that a CD141+ CAF population is reduced upon Ate-Grab and gemcitabine combination treatment. Overall, our results elucidate the mechanism underlying chemotherapy-induced fibrosis in PDAC and highlight a combinatorial therapeutic strategy for desmoplastic cancers.


Assuntos
Antineoplásicos , Fibroblastos Associados a Câncer , Carcinoma Ductal Pancreático , Neoplasias Pancreáticas , Anticorpos de Cadeia Única , Feminino , Humanos , Fibroblastos Associados a Câncer/metabolismo , Fator A de Crescimento do Endotélio Vascular/metabolismo , Fator de Crescimento Placentário/genética , Fator de Crescimento Placentário/metabolismo , Anticorpos de Cadeia Única/metabolismo , Neoplasias Pancreáticas/patologia , Carcinoma Ductal Pancreático/genética , Receptores de Fatores de Crescimento do Endotélio Vascular/metabolismo , Antineoplásicos/farmacologia , Fibrose , Neoplasias Pancreáticas
8.
Int J Mol Sci ; 23(17)2022 Sep 04.
Artigo em Inglês | MEDLINE | ID: mdl-36077527

RESUMO

The abnormal implantation of the trophoblast during the first trimester of pregnancy precedes the appearance of the clinical manifestations of preeclampsia (PE), which is a hypertensive disorder of pregnancy. In a previous study, which was carried out in a murine model of PE that was induced by NG-nitro-L-arginine methyl ester (L-NAME), we observed that the intravenous administration of fibroblast growth factor 2 (FGF2) had a hypotensive effect, improved the placental weight gain and attenuated the fetal growth restriction, and the morphological findings that were induced by L-NAME in the evaluated tissues were less severe. In this study, we aimed to determine the effect of FGF2 administration on the placental gene expression of the vascular endothelial growth factor (VEGFA), VEGF receptor 2 (VEGFR2), placental growth factor, endoglin (ENG), superoxide dismutase 1 (SOD1), catalase (CAT), thioredoxin (TXN), tumor protein P53 (P53), BCL2 apoptosis regulator, Fas cell surface death receptor (FAS), and caspase 3, in a Sprague Dawley rat PE model, which was induced by L-NAME. The gene expression was determined by a real-time polymerase chain reaction using SYBR green. Taking the vehicle or the L-NAME group as a reference, there was an under expression of placental VEGFA, VEGFR2, ENG, P53, FAS, SOD1, CAT, and TXN genes in the group of L-NAME + FGF2 (p < 0.05). The administration of FGF2 in the murine PE-like model that was induced by L-NAME reduced the effects that were generated by proteinuria and the increased BP, as well as the response of the expression of genes that participate in angiogenesis, apoptosis, and OS. These results have generated valuable information regarding the identification of molecular targets for PE and provide new insights for understanding PE pathogenesis.


Assuntos
Fator 2 de Crescimento de Fibroblastos , Pré-Eclâmpsia , Animais , Modelos Animais de Doenças , Feminino , Fator 2 de Crescimento de Fibroblastos/farmacologia , Expressão Gênica , Humanos , NG-Nitroarginina Metil Éster/efeitos adversos , Placenta/metabolismo , Fator de Crescimento Placentário/genética , Fator de Crescimento Placentário/metabolismo , Pré-Eclâmpsia/induzido quimicamente , Pré-Eclâmpsia/tratamento farmacológico , Pré-Eclâmpsia/genética , Gravidez , Ratos , Ratos Sprague-Dawley , Superóxido Dismutase-1/genética , Proteína Supressora de Tumor p53/metabolismo , Fator A de Crescimento do Endotélio Vascular/metabolismo
9.
J Biol Chem ; 298(9): 102378, 2022 09.
Artigo em Inglês | MEDLINE | ID: mdl-35970387

RESUMO

Placental growth factor (PlGF) belongs to the vascular endothelial growth factor (VEGF) family of proteins that participate in angiogenesis and vasculogenesis. Anti-VEGF therapy has become the standard treatment for ocular angiogenic disorders in ophthalmological practice. However, there is emerging evidence that anti-VEGF treatment may increase the risk of atrophy of the retinal pigment epithelium (RPE), which is important for the homeostasis of retinal tissue. Whereas the cytoprotective role of VEGF family molecules, particularly that of VEGF A (VEGFA) through its receptor VEGF receptor-2 (VEGFR-2), has been recognized, the physiological role of PlGF in the retina is still unknown. In this study, we explored the role of PlGF in the RPE using PlGF-knockdown RPE cells generated by retrovirus-based PlGF-shRNA transduction. We show that VEGFA reduced apoptosis induced by serum starvation in RPE cells, whereas the antiapoptotic effect of VEGFA was abrogated by VEGFR-2 knockdown. Furthermore, PlGF knockdown increased serum starvation-induced cell apoptosis and unexpectedly reduced the protein level of VEGFR-2 in the RPE. The antiapoptotic effect of VEGFA was also diminished in PlGF-knockdown RPE cells. In addition, we found that glycogen synthase kinase 3 activity was involved in proteasomal degradation of VEGFR-2 in RPE cells and inactivated by PlGF via AKT phosphorylation. Overall, the present data demonstrate that PlGF is crucial for RPE cell viability and that PlGF supports VEGFA/VEGFR-2 signaling by stabilizing the VEGFR-2 protein levels through glycogen synthase kinase 3 inactivation.


Assuntos
Células Epiteliais , Quinase 3 da Glicogênio Sintase , Fator de Crescimento Placentário , Receptor 2 de Fatores de Crescimento do Endotélio Vascular , Células Epiteliais/metabolismo , Quinase 3 da Glicogênio Sintase/genética , Quinase 3 da Glicogênio Sintase/metabolismo , Humanos , Epitélio Pigmentado Ocular/citologia , Fator de Crescimento Placentário/genética , Fator de Crescimento Placentário/metabolismo , Proteínas Proto-Oncogênicas c-akt , RNA Interferente Pequeno , Fator A de Crescimento do Endotélio Vascular/genética , Fator A de Crescimento do Endotélio Vascular/metabolismo , Receptor 2 de Fatores de Crescimento do Endotélio Vascular/genética , Receptor 2 de Fatores de Crescimento do Endotélio Vascular/metabolismo
10.
Artigo em Inglês | MEDLINE | ID: mdl-35490598

RESUMO

The interest on the endocannabinoid system (ECS) in human reproduction has grown due to its involvement in placenta development, which led to growing concerns over pregnant cannabis consumer's impact on pregnancy outcome. The endocannabinoids anandamide (AEA) and 2-arachidonoylglycerol (2-AG) modulate placental trophoblast proliferation and apoptosis. However, their role on other placentation events such as angiogenesis and invasion are unknown. Using the human extravillous trophoblast HTR-8/SVneo cells, a well-accepted model of first trimester extravillous trophoblast (EVT), this study aims to investigate whether AEA and 2-AG can modulate the expression of angiogenesis- and invasion-related factors. Transcript analysis of angiogenic factors of the vascular endothelial growth factor (VEGF) and matrix metalloproteinase (MMP) protein family demonstrated the ability of AEA to increase VEGF-C and VEGFR3 expression via cannabinoid receptors CB1 and CB2 while the placental growth factor (PlGF) was increased through CB1. Moreover, an increase in VEGFR1, sFLT1, VEGFR2, MMP-2 and TIMP-1 independent of cannabinoid receptor activation was verified. However, 2-AG only increased PlGF transcript through CB1/CB2 activation. Both endocannabinoids stimulated HTR8/SVneo endothelial-like tube formation. As for the wound healing assay, only 2-AG was able to increase the percentage of wound closure. Moreover, the data demonstrated that both AEA and 2-AG, via cannabinoid receptors, activated the STAT3 signaling pathway. Distinct effects were observed on transcription factor HIF-1α and AKT phosphorylation that decreased with both endocannabinoids. Although different angiogenic and migration factors are affected the results obtained in this work showcase once more the ability of the endocannabinoids to modulate key processes in placental physiology.


Assuntos
Endocanabinoides , Receptor 1 de Fatores de Crescimento do Endotélio Vascular , Indutores da Angiogênese/metabolismo , Indutores da Angiogênese/farmacologia , Ácidos Araquidônicos , Movimento Celular , Endocanabinoides/metabolismo , Endocanabinoides/farmacologia , Feminino , Glicerídeos , Humanos , Placenta/metabolismo , Fator de Crescimento Placentário/genética , Fator de Crescimento Placentário/metabolismo , Placentação , Alcamidas Poli-Insaturadas , Gravidez , Receptores de Canabinoides/metabolismo , Trofoblastos/metabolismo , Fator A de Crescimento do Endotélio Vascular/genética , Fator A de Crescimento do Endotélio Vascular/metabolismo , Receptor 1 de Fatores de Crescimento do Endotélio Vascular/metabolismo
11.
Taiwan J Obstet Gynecol ; 61(2): 359-363, 2022 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-35361402

RESUMO

OBJECTIVE: We present diagnosis and molecular cytogenetic characterization of mosaic ring chromosome 21 [r(21)]. CASE REPORT: A 17-year-old, gravida 2, para 1, woman underwent amniocentesis at 17 weeks of gestation because of an abnormal result of the first-trimester maternal serum screening for Down syndrome with a free ß-hCG level of 1.736 multiples of the median (MoM), a pregnancy associated plasma protein-A (PAPP-A) level of 0.275 MoM, a placental growth factor (PlGF) level of 0.281 MoM, a Down syndrome risk of 1:222 and a preeclampsia risk of 1:175. Cytogenetic analysis of cultured amniocytes revealed the result of 46,XX,r(21) (p12q22.3)[19]/45,XX,-21[13]. Array comparative genomic hybridization (aCGH) analysis of cultured amniocytes revealed the result of arr [GRCh37] 21q11.2q22.2 (15,485,008-40,625,594) × 1∼2, 21q22.2q22.3 (40,703,792-46,682,184) × 2∼3, 21q22.3 (46,761,631-48,084,156) × 1, consistent with mosaic monosomy 21 and r(21) (p12q22.3). The pregnancy was subsequently terminated, and a malformed fetus was delivered with low-set ears and hypotelorism. Postnatal cytogenetic analysis revealed a karyotype of 46,XX,r(21) (p12q22.3)[30]/45,XX,-21[8]/46,XX,idic r(21) (p12q22.3)[2] in the cord blood, 46,XX,r(21) (p12q22.3)[34]/45,XX,-21[6] in the skin, 46,XX,r(21) (p12q22.3)[37]/45,XX,-21[3] in the umbilical cord and 46,XX,dup(21) (q22.2q22.3)[32]/46,XX,r(21) (p12q22.3)[8] in the placenta. aCGH analysis of cord blood revealed the result of arr 21q11.2q22.2 (15,499,847-40,662,581) × 2.3, arr 21q22.2q22.3 (40,703,792-46,682,184) × 3.6, arr 21q22.3 (46,761,632-48,090,317) × 1, consistent with mosaic duplication of 21q11.2-q22.2 and 21q22.2-q22.3, and a 1.33-Mb 21q22.3 deletion encompassing the genes of COL18A1, SLC19A1, PCBP3, COL6A1, COL6A2, FTCD, LSS, MCM3AP, YBEY, PCNT, DIP2A, S100B and PRMT2. CONCLUSION: Mosaic r(21) at amniocentesis may be associated with monosomy 21, idic r(21) and dup(21), and low PAPP-A and low PlGF in the first-trimester maternal serum screening.


Assuntos
Transtornos Cromossômicos , Cromossomos em Anel , Adolescente , Transtornos Cromossômicos/diagnóstico , Transtornos Cromossômicos/genética , Hibridização Genômica Comparativa , Análise Citogenética , Feminino , Humanos , Fator de Crescimento Placentário/genética , Gravidez , Primeiro Trimestre da Gravidez , Proteína Plasmática A Associada à Gravidez/genética , Diagnóstico Pré-Natal
12.
Mol Cancer ; 21(1): 80, 2022 03 19.
Artigo em Inglês | MEDLINE | ID: mdl-35305647

RESUMO

BACKGROUND: Previous studies have shown that the N6-methyladenosine (m6A) modification enhances the binding ability of mRNAs/long noncoding RNAs (lncRNAs) to microRNAs (miRNAs), but the impact of this modification on the competitive endogenous RNA (ceRNA) function of circular RNAs (circRNAs) is unclear. METHODS: We used a human circRNA microarray to detect the expression profiles of circRNAs in 3 pairs of cancer and paracancerous tissues from patients with colorectal cancer (CRC) and 3 pairs of peripheral blood specimens from patients with CRC and healthy individuals. The circRNAs highly expressed in both peripheral blood and tumour tissues of patients with CRC, including circALG1, were screened. A quantitative reverse-transcription polymerase chain reaction (qRT-PCR) analysis of an expanded sample size was performed to detect the expression level of circALG1 in peripheral blood and tumour tissues of patients with CRC and determine its correlation with clinicopathological features, and circRNA loop-forming validation and stability assays were then conducted. Transwell assays and a nude mouse cancer metastasis model were used to study the function of circALG1 in CRC and the role of altered m6A modification levels on the regulation of circALG1 function. qRT-PCR, western blot (WB), Transwell, RNA-binding protein immunoprecipitation (RIP), RNA antisense purification (RAP), and dual-luciferase reporter gene assays were performed to analyse the ceRNA mechanism of circALG1 and the effect of the m6A modification of circALG1 on the ceRNA function of this circRNA. RESULTS: CircALG1 was highly expressed in both the peripheral blood and tumour tissues of patients with CRC and was closely associated with CRC metastasis. CircALG1 overexpression promoted the migration and invasion of CRC cells, and circALG1 silencing and reduction of the circALG1 m6A modification level inhibited CRC cell migration and invasion. In vivo experiments further confirmed the prometastatic role of circALG1 in CRC. Further mechanistic studies showed that circALG1 upregulated the expression of placental growth factor (PGF) by binding to miR-342-5p and that m6A modification enhanced the binding of circALG1 to miR-342-5p and promoted its ceRNA function. CONCLUSION: M6A modification enhances the binding ability of circALG1 to miR-342-5p to promote the ceRNA function of circALG1, and circALG1 could be a potential therapeutic target in and a prognostic marker for CRC.


Assuntos
Neoplasias Colorretais , MicroRNAs , Animais , Feminino , Humanos , Camundongos , Adenosina/análogos & derivados , Linhagem Celular Tumoral , Movimento Celular/genética , Proliferação de Células/genética , Neoplasias Colorretais/patologia , Regulação Neoplásica da Expressão Gênica , MicroRNAs/genética , MicroRNAs/metabolismo , Fator de Crescimento Placentário/genética , Fator de Crescimento Placentário/metabolismo , RNA Circular/genética
13.
J Nutr Biochem ; 104: 108977, 2022 06.
Artigo em Inglês | MEDLINE | ID: mdl-35248701

RESUMO

The aim of this study was to investigate certain parameters regarding the maternal-fetal outcomes in a diet-induced obesity model. Obese, glucose-intolerant females who were exposed to a high-fat diet prior to pregnancy had lower placental efficiency and lower birth weight pups compared to the controls. Simple linear regression analyses showed that maternal obesity disrupts the proportionality between maternal and fetal outcomes during pregnancy. Maternal obesity is correlated with fetal outcomes, perhaps because of problems with hormonal signaling and exacerbation of inflammation in the maternal metabolic environment. The maternal obese phenotype altered the thickness of the placental layer, the transport of fatty acids, and the expression of growth factors. For example, lower expression of epidermal growth factor receptor (EGFR) mRNA in the obesity-prone group may have contributed to the rupture of the placental layers, leading to adverse fetal outcomes. Furthermore, maintenance of maternal glucose homeostasis and overexpression of placental growth factor (PGF) in the obesity-resistant group likely protected the placenta and fetuses from morphological and functional damage.


Assuntos
Dieta Hiperlipídica , Obesidade Materna , Animais , Dieta Hiperlipídica/efeitos adversos , Feminino , Desenvolvimento Fetal , Retardo do Crescimento Fetal/genética , Glucose/metabolismo , Humanos , Camundongos , Obesidade/metabolismo , Fenótipo , Placenta/metabolismo , Fator de Crescimento Placentário/genética , Fator de Crescimento Placentário/metabolismo , Gravidez
14.
Lab Invest ; 102(3): 312-319, 2022 03.
Artigo em Inglês | MEDLINE | ID: mdl-34764437

RESUMO

Spinal fusion is an effective treatment for low back pain and typically applied with prosthetic fixation devices. Spinal fusion can be improved by transplantation of mesenchymal stem cells (MSCs) into the paraspinal muscle. However, in contrast to the direct contribution of MSCs to spinal fusion, the indirect effects of MSCs on spinal infusion have not been studied and were thus addressed here. The correlation between the outcome of spinal fusion and the local macrophage number, polarization and the levels of placental growth factor (PlGF) in patients was analyzed. MSCs were genetically modified to overexpress PlGF, and its effects on macrophage proliferation and polarization were analyzed in vitro in a transwell co-culture system, as well as in vivo in a mouse model for spinal fusion, for which the cells were bilaterally injected into paravertebral muscles of the mouse lumbar spine. The effects on spinal fusion were assessed by microcomputed tomography and a custom four-point bending apparatus for structural bending stiffness. Local macrophages were analyzed by flow cytometry. We found that posterior spinal fusion could be improved by PlGF-expressing MSCs, compared to the control MSCs, evident by significant improvement of bone bridging of the targeted vertebrae. Mechanistically, PlGF-expressing MSCs appeared to attract macrophages and induce their M2 polarization, which in turn promotes the bone formation. Together, our data suggest that PlGF-expressing MSCs may improve spinal fusion through macrophage recruitment and polarization.


Assuntos
Ativação de Macrófagos , Macrófagos/metabolismo , Transplante de Células-Tronco Mesenquimais/métodos , Células-Tronco Mesenquimais/metabolismo , Fator de Crescimento Placentário/metabolismo , Fusão Vertebral/métodos , Adulto , Animais , Diferenciação Celular , Movimento Celular , Células Cultivadas , Feminino , Humanos , Macrófagos/citologia , Masculino , Células-Tronco Mesenquimais/citologia , Camundongos Endogâmicos C57BL , Pessoa de Meia-Idade , Osteogênese , Fator de Crescimento Placentário/genética , Microtomografia por Raio-X , Adulto Jovem
15.
Cell Mol Life Sci ; 78(23): 7663-7679, 2021 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-34698882

RESUMO

Endothelial cells (ECs) within the microvasculature of brown adipose tissue (BAT) are important in regulating the plasticity of adipocytes in response to increased metabolic demand by modulating the angiogenic response. However, the mechanism of EC-adipocyte crosstalk during this process is not completely understood. We used RNA sequencing to profile microRNAs derived from BAT ECs of obese mice and identified an anti-angiogenic microRNA, miR-409-3p. MiR-409-3p overexpression inhibited EC angiogenic properties; whereas, its inhibition had the opposite effects. Mechanistic studies revealed that miR-409-3p targets ZEB1 and MAP4K3. Knockdown of ZEB1/MAP4K3 phenocopied the angiogenic effects of miR-409-3p. Adipocytes co-cultured with conditioned media from ECs deficient in miR-409-3p showed increased expression of BAT markers, UCP1 and CIDEA. We identified a pro-angiogenic growth factor, placental growth factor (PLGF), released from ECs in response to miR-409-3p inhibition. Deficiency of ZEB1 or MAP4K3 blocked the release of PLGF from ECs and PLGF stimulation of 3T3-L1 adipocytes increased UCP1 expression in a miR-409-3p dependent manner. MiR-409-3p neutralization improved BAT angiogenesis, glucose and insulin tolerance, and energy expenditure in mice with diet-induced obesity. These findings establish miR-409-3p as a critical regulator of EC-BAT crosstalk by modulating a ZEB1-MAP4K3-PLGF signaling axis, providing new insights for therapeutic intervention in obesity.


Assuntos
Tecido Adiposo Marrom/patologia , Resistência à Insulina , MicroRNAs/genética , Neovascularização Patológica/patologia , Fator de Crescimento Placentário/metabolismo , Proteínas Serina-Treonina Quinases/metabolismo , Homeobox 1 de Ligação a E-box em Dedo de Zinco/metabolismo , Tecido Adiposo Marrom/metabolismo , Animais , Células Endoteliais/metabolismo , Células Endoteliais/patologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Neovascularização Patológica/metabolismo , Fator de Crescimento Placentário/genética , Proteínas Serina-Treonina Quinases/genética , Transdução de Sinais , Homeobox 1 de Ligação a E-box em Dedo de Zinco/genética
16.
Front Immunol ; 12: 736153, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-34659227

RESUMO

Diabetic foot disease (DFD) is a common and serious complication for diabetes and is characterized with impaired angiogenesis. In addition to the well-defined role of vascular endothelial growth factor (VEGF) -A and its defect in the pathogenesis of DFD, another VEGF family member, placental growth factor (PlGF), was also recently found to alter expression pattern in the DFD patients with undetermined mechanisms. This question was thus addressed in the current study. We detected attenuated PlGF upregulation in a mouse DFD model. In addition, the major cell types at the wound to express the unique PlGF receptor, VEGF receptor 1 (VEGFR1), were macrophages and endothelial cells. To assess how PlGF regulates DFD-associated angiogenesis, we injected recombinant PlGF and depleted VEGF1R specifically in macrophages by local injection of an adeno-associated virus (AAV) carrying siRNA for VEGFR1 under a macrophage-specific CD68 promoter. We found that the angiogenesis and recovery of the DFD were both improved by PlGF injection. The PlGF-induced improvement in angiogenesis and the recovery of skin injury were largely attenuated by macrophage-specific depletion of VEGF1R, likely resulting from reduced macrophage number and reduced M2 polarization. Together, our data suggest that reduced PlGF compromises angiogenesis in DFD at least partially through macrophages.


Assuntos
Pé Diabético/metabolismo , Pé/irrigação sanguínea , Macrófagos/metabolismo , Neovascularização Patológica , Fator de Crescimento Placentário/metabolismo , Indutores da Angiogênese/farmacologia , Animais , Dependovirus/genética , Pé Diabético/tratamento farmacológico , Pé Diabético/genética , Pé Diabético/patologia , Modelos Animais de Doenças , Regulação para Baixo , Células Endoteliais/metabolismo , Células Endoteliais/patologia , Feminino , Fibroblastos/metabolismo , Fibroblastos/patologia , Vetores Genéticos , Macrófagos/efeitos dos fármacos , Masculino , Camundongos Endogâmicos C57BL , Fator de Crescimento Placentário/genética , Fator de Crescimento Placentário/farmacologia , Interferência de RNA , Receptor 1 de Fatores de Crescimento do Endotélio Vascular/genética , Receptor 1 de Fatores de Crescimento do Endotélio Vascular/metabolismo , Cicatrização
17.
Bioengineered ; 12(1): 6629-6642, 2021 12.
Artigo em Inglês | MEDLINE | ID: mdl-34516333

RESUMO

Clear cell renal cell carcinoma (ccRCC) is an aggressive malignancy with a poor prognosis. Therefore, investigating the molecular mechanism of ccRCC is important for ccRCC treatment. Here, we aimed to explore the effect of the long non-coding RNA ARAP1-AS1/miR-361-3p/PGF axis on ccRCC. The expression of lncRNA ARAP1-AS1, miR-361-3p, and placental growth factor (PGF) in ccRCC cells was verified by real-time quantitative PCR (RT-qPCR). The influence of the ARAP1-AS1/miR-361-3p/PGF axis on ccRCC cells was identified using the Cell Counting Kit-8 (CCK-8) assay, colony formation assay, flow cytometry, and wound healing assay. The interaction between ARAP1-AS1, miR-361-3p, and PGF was confirmed by bioinformatics analysis and luciferase assay. The results showed that the levels of ARAP1-AS1 and PGF increased in ccRCC cells, while miR-361-3p expression decreased. Cell functional experiments showed that cell proliferation and migration were inhibited by silencing ARAP1-AS1 or PGF, while miR-361-3p inhibitor or PGF overexpression could relieve the inhibitory effect of silencing ARAP1-AS1 on ccRCC cells. Moreover, ARAP1-AS1 sponges miR-361-3p to increase PGF expression. In conclusion, our study revealed that ARAP1-AS1 enhanced the malignancy of ccRCC cells by regulating the miR-361-3p/PGF axis.


Assuntos
Carcinoma de Células Renais , Neoplasias Renais , MicroRNAs/genética , Fator de Crescimento Placentário/genética , RNA Longo não Codificante/genética , Carcinoma de Células Renais/genética , Carcinoma de Células Renais/metabolismo , Carcinoma de Células Renais/patologia , Linhagem Celular Tumoral , Movimento Celular/genética , Proliferação de Células/genética , Humanos , Rim/metabolismo , Rim/patologia , Neoplasias Renais/genética , Neoplasias Renais/metabolismo , Neoplasias Renais/patologia , MicroRNAs/metabolismo , Fator de Crescimento Placentário/metabolismo , RNA Longo não Codificante/metabolismo
18.
Placenta ; 114: 22-28, 2021 10.
Artigo em Inglês | MEDLINE | ID: mdl-34418751

RESUMO

INTRODUCTION: Preeclampsia is associated with reduced pro-angiogenic Placental Growth Factor (PlGF) and increased levels of anti-angiogenic soluble FMS like tyrosine kinase-1 (sFlt-1). We have previously shown that sFlt-1 secretion is positively regulated via the Epidermal Growth Factor Receptor (EGFR) and mitochondrial respiration pathways. We assessed whether these pathways also regulate endothelial and placental secretion of PlGF. METHODS: Primary cytotrophoblast cells and primary human umbilical vein endothelial cells (HUVECs) were treated with EGFR inhibitor gefitinib, or small molecules that inhibit down-stream pathways of the receptor: U0126, PD98059 (ERK/MEK pathway inhibitors), ZM336372 (JAK/STAT inhibitor) or AG490 (JAK inhibitor). We inhibited mitochondrial respiration in primary cytotrophoblasts using mitochondrial complex inhibitors rotenone (complex I), antimycin (complex III) or oligomycin (complex IV). We then measured PlGF secretion in the condition media. RESULTS: Three inhibitors of the EGFR pathway significantly increased PlGF secretion: gefitinib (p = 0.03), AG490 (p < 0.0001) and U0126 (p = 0.03) in primary cytotrophoblasts, while PD98059 reduced PlGF secretion (p = 0.002). In the same cells, neither gefitinib or UO126 altered PlGF mRNA expression, but AG490 significantly increased its expression (p = 0.02). Primary endothelial cell PlGF secretion was significantly reduced when treated with PD98059 and U0126 while ZM336372 had no effect. Rotenone significantly reduced cytotrophoblast PlGF secretion (p = 0.0005). Neither antimycin (p = 0.9) or oligomycin (p = 0.9) had an effect. DISCUSSION: We have shown that PlGF secretion from primary cytotrophoblast and HUVECs is altered by inhibiting EGFR signaling and potentially mitochondrial respiration, coincident with reduced sFlt-1 secretion. This suggests that common pathways are regulating both pro and anti-angiogenic molecules that are changed in association with preeclampsia and provides insight into the pathogenesis of this serious disease.


Assuntos
Fator de Crescimento Epidérmico/metabolismo , Regulação da Expressão Gênica , Células Endoteliais da Veia Umbilical Humana/metabolismo , Fator de Crescimento Placentário/metabolismo , Placenta/metabolismo , Pré-Eclâmpsia/metabolismo , Transdução de Sinais/genética , Adulto , Inibidores Enzimáticos/farmacologia , Fator de Crescimento Epidérmico/genética , Feminino , Gefitinibe/farmacologia , Células Endoteliais da Veia Umbilical Humana/efeitos dos fármacos , Humanos , Placenta/efeitos dos fármacos , Fator de Crescimento Placentário/genética , Gravidez , Inibidores de Proteínas Quinases/farmacologia , Transdução de Sinais/efeitos dos fármacos , Trofoblastos/metabolismo
19.
Sci Rep ; 11(1): 16344, 2021 08 11.
Artigo em Inglês | MEDLINE | ID: mdl-34381074

RESUMO

Placenta growth factor (PlGF) is a pro-inflammatory angiogenic mediator that promotes many pathologies including diabetic complications and atherosclerosis. Widespread endothelial dysfunction precedes the onset of these conditions. As very little is known of the mechanism(s) controlling PlGF expression in pathology we investigated the role of hyperglycaemia in the regulation of PlGF production in endothelial cells. Hyperglycaemia stimulated PlGF secretion in cultured primary endothelial cells, which was suppressed by IGF-1-mediated PI3K/Akt activation. Inhibition of PI3K activity resulted in significant PlGF mRNA up-regulation and protein secretion. Similarly, loss or inhibition of Akt activity significantly increased basal PlGF expression and prevented any further PlGF secretion in hyperglycaemia. Conversely, constitutive Akt activation blocked PlGF secretion irrespective of upstream PI3K activity demonstrating that Akt is a central regulator of PlGF expression. Knock-down of the Forkhead box O-1 (FOXO1) transcription factor, which is negatively regulated by Akt, suppressed both basal and hyperglycaemia-induced PlGF secretion, whilst FOXO1 gain-of-function up-regulated PlGF in vitro and in vivo. FOXO1 association to a FOXO binding sequence identified in the PlGF promoter also increased in hyperglycaemia. This study identifies the PI3K/Akt/FOXO1 signalling axis as a key regulator of PlGF expression and unifying pathway by which PlGF may contribute to common disorders characterised by endothelial dysfunction, providing a target for therapy.


Assuntos
Células Endoteliais/fisiologia , Proteína Forkhead Box O1/genética , Hiperglicemia/genética , Fator de Crescimento Placentário/genética , Transdução de Sinais/genética , Regulação para Cima/genética , Animais , Células Cultivadas , Endotélio Vascular/fisiologia , Células Endoteliais da Veia Umbilical Humana , Humanos , Camundongos , Fosfatidilinositol 3-Quinases/genética , Regiões Promotoras Genéticas/genética , Proteínas Proto-Oncogênicas c-akt/genética , Transcrição Gênica/genética , Ativação Transcricional/genética
20.
Gene ; 791: 145724, 2021 Jul 30.
Artigo em Inglês | MEDLINE | ID: mdl-34010703

RESUMO

Intravitreal anti-vascular endothelial growth factor agents are the gold standard treatment of ocular neovascular diseases. However, their short-term efficacy implies frequent intravitreal injections. Gene therapy has the ability to provide longer duration of the therapeutic effect. We have previously described the effectiveness of the self-replicating episomal vector, pEPito, in long-term gene expression in mouse retina. In this study, we evaluated different constructs to overexpress pigment epithelium-derived factor (PEDF), an angiogenesis inhibitor, and simultaneously, to silence placental growth factor (PlGF), a key player in neovascularization. We employed the human cytomegalovirus promoter to drive the expression of PEDF and PlGF shRNA, in conjunction with cis-acting ribozymes, using pEPito as expressing vector. Our results demonstrated that the non-viral systems were able to efficiently promote a sustained increase of the PEDF: PlGF ratio in the mice retina, decreased in pathological conditions. This innovative approach could open avenues for the development of new therapeutic strategies.


Assuntos
Terapia Genética/métodos , Retina/metabolismo , Neovascularização Retiniana/metabolismo , Inibidores da Angiogênese/metabolismo , Animais , Linhagem Celular , Proteínas do Olho/genética , Proteínas do Olho/metabolismo , Feminino , Expressão Gênica/genética , Regulação da Expressão Gênica/genética , Regulação da Expressão Gênica/fisiologia , Vetores Genéticos , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Neovascularização Patológica/genética , Neovascularização Patológica/metabolismo , Neovascularização Patológica/fisiopatologia , Fatores de Crescimento Neural/genética , Fatores de Crescimento Neural/metabolismo , Fator de Crescimento Placentário/genética , Fator de Crescimento Placentário/metabolismo , Retina/fisiologia , Neovascularização Retiniana/genética , Serpinas/genética , Serpinas/metabolismo , Fator A de Crescimento do Endotélio Vascular/genética
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...