Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 66
Filtrar
1.
Arterioscler Thromb Vasc Biol ; 43(6): e190-e209, 2023 06.
Artigo em Inglês | MEDLINE | ID: mdl-37051927

RESUMO

BACKGROUND: Abnormal placental angiogenesis is an important cause of fetal intrauterine growth restriction (IUGR), but its underlying mechanisms and therapies remain unclear. Adenosine and its mediated signaling has been reported to be associated with the development of angiogenesis. However, whether the adenosine-related signaling plays a role in modulating angiogenesis in placenta and the IUGR pregnancy outcomes remains unclear. METHODS: The angiogenesis and adenosine signaling expressions in normal and IUGR placentas were detected in different species. And the role of adenosine in regulating IUGR pregnancy outcomes was evaluated using diet-induced IUGR mouse model. Molecular mechanisms underlying adenosine-induced angiogenesis were investigated by in vitro angiogenesis assays and in vivo Matrigel plug assays. RESULTS: Here, we demonstrated poor angiogenesis and low adenosine concentration and downregulated expression of its receptor A2a (ADORA2A [adenosine A2a receptor]) in IUGR placenta. Additionally, the beneficial effects of adenosine in improving IUGR pregnancy outcomes were revealed in a diet-induced IUGR mouse model. Moreover, adenosine was found to effectively improve adenosine signaling and angiogenesis in IUGR mice placenta. Mechanistically, by using angiogenesis assays in vitro and in vivo, adenosine was shown to activate ADORA2A to promote the phosphorylation of Stat3 (signal transducer and activator of transcription 3) and Akt (protein kinase B), resulting in increased Ang (angiogenin)-dependent angiogenesis. CONCLUSIONS: Collectively, this study uncovers an unexpected mechanism of promoting placental angiogenesis by adenosine-ADORA2A signaling and advances the translation of this signaling as a prognostic indicator and therapeutic target in IUGR treatment.


Assuntos
Placenta , Proteínas Proto-Oncogênicas c-akt , Animais , Feminino , Humanos , Camundongos , Gravidez , Retardo do Crescimento Fetal/induzido quimicamente , Proteínas Proto-Oncogênicas c-akt/metabolismo , Receptor A2A de Adenosina/metabolismo , Fator de Transcrição STAT3/metabolismo
2.
Ann Vasc Surg ; 2024 Sep 27.
Artigo em Inglês | MEDLINE | ID: mdl-39343376

RESUMO

BACKGROUND: Chronic limb threatening ischemia (CLTI) carries a significant risk for amputation especially in diabetic patients with poor options for revascularization. Phase I trials have demonstrated efficacy of allogeneic mesenchymal stromal cells (MSC) in treating diabetic CLTI. Vertebral bone adherent mesenchymal stromal cells (vBA-MSC) are derived from vertebral bodies of deceased organ donors which offer the distinct advantage of providing a 1,000x greater yield compared to that of living donor bone aspiration. This study describes the effects of intramuscular injection of allogenic vBA-MSC in promoting limb perfusion and muscle recovery in a diabetic CLTI mouse model. METHODS: A CLTI mouse model was created through unilateral ligation of the femoral artery in male polygenic diabetic TALLYHO mice. Treated mice were injected with vBA-MSC into the gracilis muscle of the ischemic limb 7 days post ligation. Gastrocnemius or tibialis muscle was assessed post-mortem for fibrosis by collagen staining, capillary density via immunohistochemistry, and mRNA by quantitative real time PCR. Laser Doppler perfusion imaging and plantar flexion muscle testing were performed to quantify changes in limb perfusion and muscle function. RESULTS: Compared to vehicle control, treated mice demonstrated indicators of muscle recovery including decreased fibrosis, increased perfusion, muscle torque, and angiogenesis. PCR analysis of muscle obtained 7- and 30-days post vBA-MSC injection showed an upregulation in expression of MyoD1 (p = 0.03) and MyH3 (p = 0.008) mRNA representing muscle regeneration, VEGF-A (p = 0.002 ; p = 0.004) signifying angiogenesis as well as IL-10 (p < 0.001), T regulatory cell marker Foxp3 (p = 0.04), and M2-biased macrophage marker Mrc1 (CD206) (p = 0.02). CONCLUSIONS: These findings indicate human allogeneic vBA-MSC ameliorate ischemic muscle damage and rescue muscle function. These results in a murine model will enable further studies to develop potential therapies for diabetic CLTI patients.

3.
Indian J Clin Biochem ; 39(4): 529-538, 2024 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-39346711

RESUMO

Breast cancer is the leading cause of cancer-related mortality among women worldwide. MicroRNAs (miRNAs), short non-coding RNAs, have been implicated in cancer-related processes such as tumor development, metastasis, angiogenesis, and drug resistance. Circulating miRNA-373 demonstrates higher relative exosomal serum levels in breast cancer patients compared to healthy women, making it a potential non-invasive biomarker. Separately, vascular endothelial growth factor (VEGF) is crucial for angiogenesis, and is elevated in breast cancer. In this case-control study, we aimed to investigate the diagnostic accuracy of miRNA-373 and VEGF as biomarkers for early-stage breast cancer detection. Serum samples were collected from 120 participants, comprising 30 breast cancer patients, 30 benign breast tumor patients, and 60 healthy controls, over the period of April 2022 to January 2023. MiRNA-373 expression was analyzed by reverse transcription-quantitative PCR with GAPDH normalisation, while VEGF levels in serum samples were measured by ELISA. Receiver operating characteristic (ROC) curve analysis was performed to evaluate the diagnostic performance of both biomarkers. MiRNA-373 expression (∆Ct) differed significantly between the three groups (breast cancer: - 12.20 ± 1.11; benign tumors: - 12.79 ± 1.09; controls: - 13.64 ± 0.93). ROC analysis revealed moderate discriminative power for miRNA-373 (specificity = 76.7%; sensitivity = 70.0%; AUC = 0.839) and excellent discriminative power for VEGF (specificity = 85.0%; sensitivity = 90.0%; AUC = 0.944) in distinguishing early-stage breast cancer patients from healthy controls. In summary, this study demonstrates the promising potential of miRNA-373 as an early diagnostic biomarker for breast cancer detection, requiring further validation in larger cohorts. Our findings also reinforce the diagnostic value of circulating VEGF levels for breast cancer screening. Supplementary Information: The online version contains supplementary material available at 10.1007/s12291-023-01174-9.

4.
Int Endod J ; 56(10): 1270-1283, 2023 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-37461231

RESUMO

AIM: Angiogenesis contributes to the development of apical periodontitis, periodontitis, and other oral pathologies; however, it remains unclear how this process is triggered. The aim was to evaluate whether lipopolysaccharide (LPS) from Porphyromonas endodontalis and Porphyromonas gingivalis induced angiogenesis-related effects in vitro via TLR2 and TLR4. METHODOLOGY: Porphyromonas endodontalis LPS (ATCC 35406 and clinical isolate) was purified with TRIzol, whereas P. gingivalis LPS was obtained commercially. The effects of the different LPS (24 h) in endothelial cell migration were analysed by Transwell assays, following quantification in an optical microscope (40×). The effects of LPS on FAK Y397 phosphorylation were assessed by Western blotting. Angiogenesis in vitro was determined in an endothelial tube formation assay (14 h) in Matrigel in the absence or presence of either LPS. IL-6 and VEGF-A levels were determined in cell supernatants, following 24 h treatment with LPS, and measured in multiplex bead immunoassay. The involvement of TLR2 and TLR4 was assessed with blocking antibodies. The statistical analysis was performed using STATA 12® (StataCorp LP). RESULTS: The results revealed that P. endodontalis LPS, but not P. gingivalis LPS, stimulated endothelial cell migration. Pre-treatment with anti-TLR2 and anti-TLR4 antibodies prevented P. endodontalis LPS-induced cell migration. P. endodontalis LPS promoted FAK phosphorylation on Y397, as observed by an increased p-FAK/FAK ratio. Both P. gingivalis and P. endodontalis LPS (ATCC 35406) induced endothelial tube formation in a TLR-2 and -4-dependent manner, as shown by using blocking antibodies, however, only TLR2 blocking decreased tube formation induced by P. endodontalis (clinical isolate). Moreover, all LPS induced IL-6 and VEGF-A synthesis in endothelial cells. TLR2 and TLR4 were required for IL-6 induction by P. endodontalis LPS (ATCC 35406), while only TLR4 was involved in IL-6 secretion by the other LPS. Finally, VEGF-A synthesis did not require TLR signalling. CONCLUSION: Porphyromonas endodontalis and P. gingivalis LPS induced angiogenesis via TLR2 and TLR4. Collectively, these data contribute to understanding the role of LPS from Porphyromonas spp. in angiogenesis and TLR involvement.


Assuntos
Lipopolissacarídeos , Receptor 2 Toll-Like , Lipopolissacarídeos/farmacologia , Receptor 2 Toll-Like/metabolismo , Porphyromonas gingivalis/metabolismo , Porphyromonas endodontalis/metabolismo , Fator A de Crescimento do Endotélio Vascular , Células Endoteliais/metabolismo , Anticorpos Bloqueadores , Interleucina-6 , Receptor 4 Toll-Like/metabolismo
5.
Circulation ; 139(2): 226-242, 2019 01 08.
Artigo em Inglês | MEDLINE | ID: mdl-30586702

RESUMO

BACKGROUND: Atherosclerotic occlusions decrease blood flow to the lower limbs, causing ischemia and tissue loss in patients with peripheral artery disease (PAD). No effective medical therapies are currently available to induce angiogenesis and promote perfusion recovery in patients with severe PAD. Clinical trials aimed at inducing vascular endothelial growth factor (VEGF)-A levels, a potent proangiogenic growth factor to induce angiogenesis, and perfusion recovery were not successful. Alternate splicing in the exon-8 of VEGF-A results in the formation of VEGFxxxa (VEGF165a) and VEGFxxxb (VEGF165b) isoforms with existing literature focusing on VEGF165b's role in inhibiting vascular endothelial growth factor receptor 2-dependent angiogenesis. However, we have recently shown that VEGF165b blocks VEGF-A-induced endothelial vascular endothelial growth factor receptor 1 (VEGFR1) activation in ischemic muscle to impair perfusion recovery. Because macrophage-secreted VEGF165b has been shown to decrease angiogenesis in peripheral artery disease, and macrophages were well known to play important roles in regulating ischemic muscle vascular remodeling, we examined the role of VEGF165b in regulating macrophage function in PAD. METHODS: Femoral artery ligation and resection were used as an in vivo preclinical PAD model, and hypoxia serum starvation was used as an in vitro model for PAD. Experiments including laser-Doppler perfusion imaging, adoptive cell transfer to ischemic muscle, immunoblot analysis, ELISAs, immunostainings, flow cytometry, quantitative polymerase chain reaction analysis, and RNA sequencing were performed to determine a role of VEGF165b in regulating macrophage phenotype and function in PAD. RESULTS: First, we found increased VEGF165b expression with increased M1-like macrophages in PAD versus non-PAD (controls) muscle biopsies. Next, using in vitro hypoxia serum starvation, in vivo pre clinical PAD models, and adoptive transfer of VEGF165b-expressing bone marrow-derived macrophages or VEGFR1+/- bone marrow-derived macrophages (M1-like phenotype), we demonstrate that VEGF165b inhibits VEGFR1 activation to induce an M1-like phenotype that impairs ischemic muscle neovascularization. Subsequently, we found S100A8/S100A9 as VEGFR1 downstream regulators of macrophage polarization by RNA-Seq analysis of hypoxia serum starvation-VEGFR1+/+ versus hypoxia serum starvation-VEGFR1+/- bone marrow-derived macrophages. CONCLUSIONS: In our current study, we demonstrate that increased VEGF165b expression in macrophages induces an antiangiogenic M1-like phenotype that directly impairs angiogenesis. VEGFR1 inhibition by VEGF165b results in S100A8/S100A9-mediated calcium influx to induce an M1-like phenotype that impairs ischemic muscle revascularization and perfusion recovery.


Assuntos
Calgranulina A/metabolismo , Calgranulina B/metabolismo , Células Endoteliais/metabolismo , Isquemia/metabolismo , Macrófagos/metabolismo , Músculo Esquelético/irrigação sanguínea , Neovascularização Fisiológica , Doença Arterial Periférica/metabolismo , Fator A de Crescimento do Endotélio Vascular/metabolismo , Animais , Sinalização do Cálcio , Células Cultivadas , Modelos Animais de Doenças , Células Endoteliais/patologia , Humanos , Isquemia/patologia , Isquemia/fisiopatologia , Macrófagos/patologia , Camundongos da Linhagem 129 , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , Camundongos Knockout , Comunicação Parácrina , Doença Arterial Periférica/patologia , Doença Arterial Periférica/fisiopatologia , Fenótipo , Fator A de Crescimento do Endotélio Vascular/genética , Receptor 1 de Fatores de Crescimento do Endotélio Vascular/genética , Receptor 1 de Fatores de Crescimento do Endotélio Vascular/metabolismo
6.
Circulation ; 135(8): 786-805, 2017 02 21.
Artigo em Inglês | MEDLINE | ID: mdl-27899395

RESUMO

BACKGROUND: During adult life, blood vessel formation is thought to occur via angiogenic processes involving branching from existing vessels. An alternate proposal suggests that neovessels form from endothelial progenitors able to assemble the intimal layers. We here aimed to define vessel-resident endothelial progenitors in vivo in a variety of tissues in physiological and pathological situations such as normal aorta, lungs, and wound healing, tumors, and placenta, as well. METHODS: Based on protein expression levels of common endothelial markers using flow cytometry, 3 subpopulations of endothelial cells could be identified among VE-Cadherin+ and CD45- cells. RESULTS: Lineage tracing by using Cdh5creERt2/Rosa-YFP reporter strategy demonstrated that the CD31-/loVEGFR2lo/intracellular endothelial population was indeed an endovascular progenitor (EVP) of an intermediate CD31intVEGFR2lo/intracellular transit amplifying (TA) and a definitive differentiated (D) CD31hiVEGFR2hi/extracellular population. EVP cells arose from vascular-resident beds that could not be transferred by bone marrow transplantation. Furthermore, EVP displayed progenitor-like status with a high proportion of cells in a quiescent cell cycle phase as assessed in wounds, tumors, and aorta. Only EVP cells and not TA and D cells had self-renewal capacity as demonstrated by colony-forming capacity in limiting dilution and by transplantation in Matrigel plugs in recipient mice. RNA sequencing revealed prominent gene expression differences between EVP and D cells. In particular, EVP cells highly expressed genes related to progenitor function including Sox9, Il33, Egfr, and Pdfgrα. Conversely, D cells highly expressed genes related to differentiated endothelium including Ets1&2, Gata2, Cd31, Vwf, and Notch. The RNA sequencing also pointed to an essential role of the Sox18 transcription factor. The role of SOX18 in the differentiation process was validated by using lineage-tracing experiments based on Sox18CreERt2/Rosa-YFP mice. Besides, in the absence of functional SOX18/SOXF, EVP progenitors were still present, but TA and D populations were significantly reduced. CONCLUSIONS: Our findings support an entirely novel endothelial hierarchy, from EVP to TA to D, as defined by self-renewal, differentiation, and molecular profiling of an endothelial progenitor. This paradigm shift in our understanding of vascular-resident endothelial progenitors in tissue regeneration opens new avenues for better understanding of cardiovascular biology.


Assuntos
Células Endoteliais/metabolismo , Células-Tronco/metabolismo , Animais , Antígenos CD/metabolismo , Aorta/metabolismo , Aorta/patologia , Transplante de Medula Óssea , Caderinas/metabolismo , Diferenciação Celular , Células Endoteliais/citologia , Endotélio Vascular/citologia , Feminino , Antígenos Comuns de Leucócito/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Neovascularização Fisiológica , Placenta/metabolismo , Placenta/patologia , Molécula-1 de Adesão Celular Endotelial a Plaquetas/metabolismo , Gravidez , Fatores de Transcrição SOXF/metabolismo , Células-Tronco/citologia , Receptor 2 de Fatores de Crescimento do Endotélio Vascular/metabolismo , Ferimentos e Lesões/patologia , Ferimentos e Lesões/terapia
7.
Int Wound J ; 15(2): 274-282, 2018 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-29239111

RESUMO

This study aimed to investigate the effects of gallium-aluminum-arsenium (GaAlAs) (670 nm) laser therapy on neoangiogenesis and fibroplasia during tissue remodelling. Forty male Wistar rats underwent cutaneous surgery and were divided into 2 experimental groups: the Control and Laser group (9 mW, 670 nm, 0.031 W/cm2 , 4 J/cm2 ). After 14, 21, 28, and 35 days, the animals were euthanised. Descriptive and quantitative analyses were performed in sections stained with haematoxylin-eosin and Sirius Red, respectively. The amounts of VEGF+ and CD31+ cells were evaluated by immunohistochemistry and histomorphometric analysis, respectively. Statistical analysis was performed using the Mann-Whitney, Friedman, and Spearman correlation test, P < 0.05. The collagen expression was significantly higher in the laser group compared with the control group on days 14 and 21 after the creation of the skin wound (P = 0.008; P = 0.016) and in the control group between 14 and 28 and 14 and 35 days (P = 0.001; P = 0.007). There were more blood vessels in three periods of the study only in the (Laser) treated group, with statistical significance at day 14 (P = 0.016). There was no statistically significant difference in VEGF+ cell count in the different experimental groups throughout the study, although a positive correlation was shown with the area of collagen on days 14 and 28 (P = 0.037). Laser treatment had a positive effect in the late course of healing, particularly with regards to collagen expression and the number of newly formed vessels. VEGF+ cells were present in both experimental groups, and VEGF appeared to influence fibroplasia in the treated group.


Assuntos
Colágeno/efeitos da radiação , Lasers Semicondutores/uso terapêutico , Terapia com Luz de Baixa Intensidade/métodos , Pele/efeitos da radiação , Cicatrização/fisiologia , Ferimentos e Lesões/tratamento farmacológico , Ferimentos e Lesões/radioterapia , Alumínio/uso terapêutico , Animais , Colágeno/efeitos dos fármacos , Gálio/uso terapêutico , Masculino , Modelos Animais , Ratos , Ratos Wistar
8.
Circulation ; 134(24): 1973-1990, 2016 Dec 13.
Artigo em Inglês | MEDLINE | ID: mdl-27780851

RESUMO

BACKGROUND: The transcription factor GATA2 orchestrates the expression of many endothelial-specific genes, illustrating its crucial importance for endothelial cell function. The capacity of this transcription factor in orchestrating endothelial-important microRNAs (miRNAs/miR) is unknown. METHODS: Endothelial GATA2 was functionally analyzed in human endothelial cells in vitro. Endogenous short interfering RNA-mediated knockdown and lentiviral-based overexpression were applied to decipher the capacity of GATA2 in regulating cell viability and capillary formation. Next, the GATA2-dependent miR transcriptome was identified by using a profiling approach on the basis of quantitative real-time polymerase chain reaction. Transcriptional control of miR promoters was assessed via chromatin immunoprecipitation, luciferase promoter assays, and bisulfite sequencing analysis of sites in proximity. Selected miRs were modulated in combination with GATA2 to identify signaling pathways at the angiogenic cytokine level via proteome profiler and enzyme-linked immunosorbent assays. Downstream miR targets were identified via bioinformatic target prediction and luciferase reporter gene assays. In vitro findings were translated to a mouse model of carotid injury in an endothelial GATA2 knockout background. Nanoparticle-mediated delivery of proangiogenic miR-126 was tested in the reendothelialization model. RESULTS: GATA2 gain- and loss-of-function experiments in human umbilical vein endothelial cells identified a key role of GATA2 as master regulator of multiple endothelial functions via miRNA-dependent mechanisms. Global miRNAnome-screening identified several GATA2-regulated miRNAs including miR-126 and miR-221. Specifically, proangiogenic miR-126 was regulated by GATA2 transcriptionally and targeted antiangiogenic SPRED1 and FOXO3a contributing to GATA2-mediated formation of normal vascular structures, whereas GATA2 deficiency led to vascular abnormalities. In contrast to GATA2 deficiency, supplementation with miR-126 normalized vascular function and expression profiles of cytokines contributing to proangiogenic paracrine effects. GATA2 silencing resulted in endothelial DNA hypomethylation leading to induced expression of antiangiogenic miR-221 by GATA2-dependent demethylation of a putative CpG island in the miR-221 promoter. Mechanistically, a reverted GATA2 phenotype by endogenous suppression of miR-221 was mediated through direct proangiogenic miR-221 target genes ICAM1 and ETS1. In a mouse model of carotid injury, GATA2 was reduced, and systemic supplementation of miR-126-coupled nanoparticles enhanced miR-126 availability in the carotid artery and improved reendothelialization of injured carotid arteries in vivo. CONCLUSIONS: GATA2-mediated regulation of miR-126 and miR-221 has an important impact on endothelial biology. Hence, modulation of GATA2 and its targets miR-126 and miR-221 is a promising therapeutic strategy for treatment of many vascular diseases.


Assuntos
Doenças das Artérias Carótidas/terapia , Fator de Transcrição GATA2/metabolismo , MicroRNAs/uso terapêutico , Regiões 3' não Traduzidas , Proteínas Adaptadoras de Transdução de Sinal , Animais , Antagomirs/metabolismo , Sequência de Bases , Doenças das Artérias Carótidas/patologia , Modelos Animais de Doenças , Proteína Forkhead Box O3/antagonistas & inibidores , Proteína Forkhead Box O3/genética , Proteína Forkhead Box O3/metabolismo , Fator de Transcrição GATA2/antagonistas & inibidores , Fator de Transcrição GATA2/genética , Vetores Genéticos/genética , Vetores Genéticos/metabolismo , Células Endoteliais da Veia Umbilical Humana , Humanos , Molécula 1 de Adesão Intercelular/química , Molécula 1 de Adesão Intercelular/genética , Molécula 1 de Adesão Intercelular/metabolismo , Peptídeos e Proteínas de Sinalização Intracelular/química , Peptídeos e Proteínas de Sinalização Intracelular/genética , Peptídeos e Proteínas de Sinalização Intracelular/metabolismo , Lentivirus/genética , Masculino , Proteínas de Membrana/química , Proteínas de Membrana/genética , Proteínas de Membrana/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , MicroRNAs/antagonistas & inibidores , MicroRNAs/metabolismo , Nanopartículas/química , Interferência de RNA , RNA Interferente Pequeno/metabolismo , Alinhamento de Sequência
9.
Circ Res ; 117(4): 321-32, 2015 Jul 31.
Artigo em Inglês | MEDLINE | ID: mdl-25966710

RESUMO

RATIONALE: Oxidative stress is an important contributing factor in several human pathologies ranging from atherosclerosis to cancer progression; however, the mechanisms underlying tissue protection from oxidation products are poorly understood. Oxidation of membrane phospholipids, containing the polyunsaturated fatty acid docosahexaenoic acid, results in the accumulation of an end product, 2-(ω-carboxyethyl)pyrrole (CEP), which was shown to have proangiogenic and proinflammatory functions. Although CEP is continuously accumulated during chronic processes, such as tumor progression and atherosclerosis, its level during wound healing return to normal when the wound is healed, suggesting the existence of a specific clearance mechanism. OBJECTIVE: To identify the cellular and molecular mechanism for CEP clearance. METHODS AND RESULTS: Here, we show that macrophages are able to bind, scavenge, and metabolize carboxyethylpyrrole derivatives of proteins but not structurally similar ethylpyrrole derivatives, demonstrating the high specificity of the process. F4/80(hi) and M2-skewed macrophages are much more efficient at CEP binding and scavenging compared with F4/80(lo) and M1-skewed macrophages. Depletion of macrophages leads to increased CEP accumulation in vivo. CEP binding and clearance are dependent on 2 receptors expressed by macrophages, CD36 and toll-like receptor 2. Although knockout of each individual receptor results in diminished CEP clearance, the lack of both receptors almost completely abrogates macrophages' ability to scavenge CEP derivatives of proteins. CONCLUSIONS: Our study demonstrates the mechanisms of recognition, scavenging, and clearance of pathophysiologically active products of lipid oxidation in vivo, thereby contributing to tissue protection against products of oxidative stress.


Assuntos
Antígenos CD36/metabolismo , Lipoproteínas LDL/metabolismo , Macrófagos Peritoneais/metabolismo , Estresse Oxidativo , Pirróis/metabolismo , Receptor 2 Toll-Like/metabolismo , Animais , Antígenos de Diferenciação/metabolismo , Apolipoproteínas E/deficiência , Apolipoproteínas E/genética , Aterosclerose/genética , Aterosclerose/metabolismo , Antígenos CD36/deficiência , Antígenos CD36/genética , Modelos Animais de Doenças , Células HEK293 , Humanos , Macrófagos Peritoneais/imunologia , Melanoma Experimental/metabolismo , Melanoma Experimental/patologia , Camundongos Endogâmicos C57BL , Camundongos Knockout , Neovascularização Fisiológica , Fenótipo , Interferência de RNA , Transdução de Sinais , Fatores de Tempo , Receptor 2 Toll-Like/deficiência , Receptor 2 Toll-Like/genética , Transfecção , Carga Tumoral , Cicatrização
10.
J Urol ; 195(6): 1936-46, 2016 06.
Artigo em Inglês | MEDLINE | ID: mdl-26812302

RESUMO

PURPOSE: We examined whether and how Sac-1004, a vascular leakage blocker, would restore erectile function in an animal model of diabetic erectile dysfunction. MATERIALS AND METHODS: Eight-week-old C57BL/6J mice were used. Diabetes was induced by intraperitoneal injection of streptozotocin. Eight weeks after diabetes induction the animals were divided into 6 groups, including controls, diabetic mice that received repeat intracavernous injections of phosphate buffered saline (20 µl) on days -3 and 0, and diabetic mice that received repeat intracavernous injections of Sac-1004 on days -3 and 0 (1, 2, 5 and 10 µg, respectively, in 20 µl phosphate buffered saline). One week after injection erectile function was measured by cavernous nerve stimulation. The penis was then harvested for histological examinations and Western blot analysis. RESULTS: Local delivery of Sac-1004 in the corpus cavernosum restored erectile function in diabetic mice. The highest erectile response was noted at a dose of 5 µg with a response comparable to that in the control group. Sac-1004 significantly increased cavernous endothelial and smooth muscle contents, and induced endothelial nitric oxide synthase phosphorylation (Ser1177). Sac-1004 decreased extravasation of oxidized low density lipoprotein by restoring endothelial cell-cell junction proteins and pericyte content. Sac-1004 also promoted tube formation in primary cultured mouse cavernous endothelial cells in vitro. Sac-1004 mediated cavernous angiogenesis and erectile function recovery was abolished by inhibiting angiopoietin-1-Tie2 signaling with soluble Tie2 antibody. CONCLUSIONS: With the effects of angiogenesis and antipermeability Sac-1004 reestablishes structural and functional cavernous sinusoids. This is highly promising for future treatment of erectile dysfunction from vascular causes.


Assuntos
Indutores da Angiogênese/uso terapêutico , Diabetes Mellitus Experimental/complicações , Disfunção Erétil/tratamento farmacológico , Saponinas/uso terapêutico , Indutores da Angiogênese/farmacologia , Animais , Diabetes Mellitus Experimental/induzido quimicamente , Disfunção Erétil/etiologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Neovascularização Fisiológica/efeitos dos fármacos , Saponinas/farmacologia , Estreptozocina , Resultado do Tratamento
11.
Arterioscler Thromb Vasc Biol ; 35(2): 348-57, 2015 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-25425620

RESUMO

OBJECTIVE: Therapeutic angiogenesis is a promising strategy for treating ischemia. Our previous work showed that endogenous endothelial tissue factor (TF) expression induces intracrine signaling and switches-on angiogenesis in microvascular endothelial cells (mECs). We have hypothesized that activated mECs could exert a further paracrine regulation through the release of TF-rich microvascular endothelial microparticles (mEMPs) and induce neovascularization of ischemic tissues. APPROACH AND RESULTS: Here, we describe for the first time that activated mECs are able to induce reparative neovascularization in ischemic zones by releasing TF-rich microparticles. We show in vitro and in vivo that mEMPs released by both wild-type and TF-upregulated-mECs induce angiogenesis and collateral vessel formation, whereas TF-poor mEMPs derived from TF-silenced mECs are not able to trigger angiogenesis. Isolated TF-bearing mEMPs delivered to nonperfused adductor muscles in a murine hindlimb ischemia model enhance collateral flow and capillary formation evidenced by MRI. TF-bearing mEMPs increase angiogenesis operating via paracrine regulation of neighboring endothelial cells, signaling through the ß1-integrin pathway Rac1-ERK1/2-ETS1 and triggering CCL2 (chemokine [C-C motif] ligand 2) production to form new and competent mature neovessels. CONCLUSIONS: These findings demonstrate that TF-rich mEMPs released by microvascular endothelial cells can overcome the consequences of arterial occlusion and tissue ischemia by promoting postischemic neovascularization and tissue reperfusion.


Assuntos
Micropartículas Derivadas de Células/transplante , Circulação Colateral , Células Endoteliais/transplante , Isquemia/cirurgia , Microvasos/metabolismo , Músculo Esquelético/irrigação sanguínea , Músculo Esquelético/metabolismo , Neovascularização Fisiológica , Tromboplastina/metabolismo , Animais , Linhagem Celular , Micropartículas Derivadas de Células/metabolismo , Micropartículas Derivadas de Células/patologia , Modelos Animais de Doenças , Células Endoteliais/metabolismo , Células Endoteliais/patologia , Membro Posterior , Humanos , Integrina beta1/metabolismo , Isquemia/genética , Isquemia/metabolismo , Isquemia/patologia , Isquemia/fisiopatologia , Angiografia por Ressonância Magnética , Masculino , Camundongos Nus , Microvasos/patologia , Microvasos/fisiopatologia , Músculo Esquelético/patologia , Comunicação Parácrina , Interferência de RNA , Transdução de Sinais , Tromboplastina/genética , Fatores de Tempo , Técnicas de Cultura de Tecidos , Transfecção
12.
Circulation ; 129(21): 2144-57, 2014 May 27.
Artigo em Inglês | MEDLINE | ID: mdl-24710033

RESUMO

BACKGROUND: Bronchopulmonary dysplasia and emphysema are life-threatening diseases resulting from impaired alveolar development or alveolar destruction. Both conditions lack effective therapies. Angiogenic growth factors promote alveolar growth and contribute to alveolar maintenance. Endothelial colony-forming cells (ECFCs) represent a subset of circulating and resident endothelial cells capable of self-renewal and de novo vessel formation. We hypothesized that resident ECFCs exist in the developing lung, that they are impaired during arrested alveolar growth in experimental bronchopulmonary dysplasia, and that exogenous ECFCs restore disrupted alveolar growth. METHODS AND RESULTS: Human fetal and neonatal rat lungs contain ECFCs with robust proliferative potential, secondary colony formation on replating, and de novo blood vessel formation in vivo when transplanted into immunodeficient mice. In contrast, human fetal lung ECFCs exposed to hyperoxia in vitro and neonatal rat ECFCs isolated from hyperoxic alveolar growth-arrested rat lungs mimicking bronchopulmonary dysplasia proliferated less, showed decreased clonogenic capacity, and formed fewer capillary-like networks. Intrajugular administration of human cord blood-derived ECFCs after established arrested alveolar growth restored lung function, alveolar and lung vascular growth, and attenuated pulmonary hypertension. Lung ECFC colony- and capillary-like network-forming capabilities were also restored. Low ECFC engraftment and the protective effect of cell-free ECFC-derived conditioned media suggest a paracrine effect. Long-term (10 months) assessment of ECFC therapy showed no adverse effects with persistent improvement in lung structure, exercise capacity, and pulmonary hypertension. CONCLUSIONS: Impaired ECFC function may contribute to arrested alveolar growth. Cord blood-derived ECFC therapy may offer new therapeutic options for lung diseases characterized by alveolar damage.


Assuntos
Proliferação de Células/efeitos dos fármacos , Células Endoteliais/fisiologia , Oxigênio/toxicidade , Alvéolos Pulmonares/efeitos dos fármacos , Alvéolos Pulmonares/cirurgia , Transplante de Células-Tronco/métodos , Animais , Animais Recém-Nascidos , Células Cultivadas , Células Endoteliais/transplante , Feto , Células Endoteliais da Veia Umbilical Humana/fisiologia , Células Endoteliais da Veia Umbilical Humana/transplante , Humanos , Camundongos , Camundongos Endogâmicos NOD , Camundongos Knockout , Camundongos SCID , Alvéolos Pulmonares/lesões , Ratos , Ratos Nus , Ratos Sprague-Dawley
13.
Circulation ; 129(17): 1770-80, 2014 Apr 29.
Artigo em Inglês | MEDLINE | ID: mdl-24503951

RESUMO

BACKGROUND: Chloride intracellular channel 4 (CLIC4) is highly expressed in the endothelium of remodeled pulmonary vessels and plexiform lesions of patients with pulmonary arterial hypertension. CLIC4 regulates vasculogenesis through endothelial tube formation. Aberrant CLIC4 expression may contribute to the vascular pathology of pulmonary arterial hypertension. METHODS AND RESULTS: CLIC4 protein expression was increased in plasma and blood-derived endothelial cells from patients with idiopathic pulmonary arterial hypertension and in the pulmonary vascular endothelium of 3 rat models of pulmonary hypertension. CLIC4 gene deletion markedly attenuated the development of chronic hypoxia-induced pulmonary hypertension in mice. Adenoviral overexpression of CLIC4 in cultured human pulmonary artery endothelial cells compromised pulmonary endothelial barrier function and enhanced their survival and angiogenic capacity, whereas CLIC4 shRNA had an inhibitory effect. Similarly, inhibition of CLIC4 expression in blood-derived endothelial cells from patients with idiopathic pulmonary arterial hypertension attenuated the abnormal angiogenic behavior that characterizes these cells. The mechanism of CLIC4 effects involves p65-mediated activation of nuclear factor-κB, followed by stabilization of hypoxia-inducible factor-1α and increased downstream production of vascular endothelial growth factor and endothelin-1. CONCLUSION: Increased CLIC4 expression is an early manifestation and mediator of endothelial dysfunction in pulmonary hypertension.


Assuntos
Canais de Cloreto/fisiologia , Endotélio Vascular/fisiopatologia , Hipertensão Pulmonar/fisiopatologia , Proteínas Mitocondriais/fisiologia , Artéria Pulmonar/fisiopatologia , Animais , Células Cultivadas , Canais de Cloreto/genética , Endotélio Vascular/citologia , Hipertensão Pulmonar Primária Familiar , Humanos , Hipertensão Pulmonar/genética , Subunidade alfa do Fator 1 Induzível por Hipóxia/fisiologia , Masculino , Camundongos , Camundongos Endogâmicos , Camundongos Knockout , Proteínas Mitocondriais/genética , Músculo Liso Vascular/citologia , Músculo Liso Vascular/fisiopatologia , Artéria Pulmonar/citologia , Ratos , Ratos Sprague-Dawley , Fator de Transcrição RelA/fisiologia
14.
Arterioscler Thromb Vasc Biol ; 34(2): 408-18, 2014 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-24202301

RESUMO

OBJECTIVE: CTX0E03 (CTX) is a clinical-grade human neural stem cell (hNSC) line that promotes angiogenesis and neurogenesis in a preclinical model of stroke and is now under clinical development for stroke disability. We evaluated the therapeutic activity of intramuscular CTX hNSC implantation in murine models of hindlimb ischemia for potential translation to clinical studies in critical limb ischemia. APPROACH AND RESULTS: Immunodeficient (CD-1 Fox(nu/nu)) mice acutely treated with hNSCs had overall significantly increased rates and magnitude of recovery of surface blood flow (laser Doppler), limb muscle perfusion (fluorescent microspheres, P<0.001), and capillary and small arteriole densities in the ischemic limb (fluorescence immunohistochemistry, both P<0.001) when compared with the vehicle-treated group. Hemodynamic and anatomic improvements were dose related and optimal at a minimum dose of 3×10(5) cells. Dose-dependent improvements in blood flow and increased vessel densities by hNSC administration early after ischemia were confirmed in immunocompetent CD-1 and streptozotocin-induced diabetic mice, together with marked reductions in the incidence of necrotic toes (P<0.05). Delayed administration of hNSCs, 7 days after occlusion, produced restorative effects when comparable with acute treatment of 35 days after hindlimb ischemia. Histological studies in hindlimb ischemia immunocompetent mice for the first 7 days after treatment revealed short-term hNSC survival, transient elevation of early host muscle inflammatory, and angiogenic responses and acceleration of myogenesis. CONCLUSIONS: hNSC therapy represents a promising treatment option for critical limb ischemia.


Assuntos
Pé Diabético/cirurgia , Isquemia/cirurgia , Músculo Esquelético/irrigação sanguínea , Neovascularização Fisiológica , Células-Tronco Neurais/transplante , Animais , Arteríolas/fisiopatologia , Velocidade do Fluxo Sanguíneo , Capilares/fisiopatologia , Linhagem Celular , Sobrevivência Celular , Pé Diabético/imunologia , Pé Diabético/fisiopatologia , Modelos Animais de Doenças , Regulação da Expressão Gênica , Membro Posterior , Humanos , Imunocompetência , Isquemia/genética , Isquemia/imunologia , Isquemia/fisiopatologia , Fluxometria por Laser-Doppler , Camundongos , Camundongos Knockout , Camundongos Nus , Células-Tronco Neurais/imunologia , Fluxo Sanguíneo Regional , Fatores de Tempo
15.
Circulation ; 128(1): 60-71, 2013 Jul 02.
Artigo em Inglês | MEDLINE | ID: mdl-23723256

RESUMO

BACKGROUND: Hypoandrogenemia is associated with an increased risk of ischemic diseases. Because actions of androgens are exerted through androgen receptor (AR) activation, we studied hind-limb ischemia in AR knockout mice to elucidate the role of AR in response to ischemia. METHODS AND RESULTS: Both male and female AR knockout mice exhibited impaired blood flow recovery, more cellular apoptosis, and a higher incidence of autoamputation after ischemia. In ex vivo and in vivo angiogenesis studies, AR-deficient vascular endothelial cells showed reduced angiogenic capability. In ischemic limbs of AR knockout mice, reductions in the phosphorylation of the Akt protein kinase and endothelial nitric oxide synthase were observed despite a robust increase in hypoxia-inducible factor 1α and vascular endothelial cell growth factor (VEGF) gene expression. In in vitro studies, siRNA-mediated ablation of AR in vascular endothelial cells blunted VEGF-stimulated phosphorylation of Akt and endothelial nitric oxide synthase. Immunoprecipitation experiments documented an association between AR and kinase insert domain protein receptor that promoted the recruitment of downstream signaling components. CONCLUSIONS: These results document a physiological role of AR in sex-independent angiogenic potency and provide evidence of novel cross-talk between the androgen/AR signaling and VEGF/kinase insert domain protein receptor signaling pathways.


Assuntos
Isquemia/fisiopatologia , Neovascularização Fisiológica/fisiologia , Receptores Androgênicos/genética , Receptores Androgênicos/metabolismo , Receptor 2 de Fatores de Crescimento do Endotélio Vascular/metabolismo , Cotos de Amputação/patologia , Animais , Apoptose/fisiologia , Capilares/fisiologia , Feminino , Feminização/genética , Feminização/metabolismo , Membro Posterior/irrigação sanguínea , Membro Posterior/patologia , Células Endoteliais da Veia Umbilical Humana , Humanos , Isquemia/metabolismo , Isquemia/patologia , Masculino , Camundongos , Camundongos Knockout , Músculo Esquelético/irrigação sanguínea , Músculo Esquelético/patologia , Óxido Nítrico Sintase Tipo III/metabolismo , Fosfatidilinositol 3-Quinases/metabolismo , Proteínas Proto-Oncogênicas c-akt/metabolismo , Proteínas Proto-Oncogênicas pp60(c-src)/metabolismo , Receptor Cross-Talk/fisiologia , Transdução de Sinais/fisiologia
16.
Arterioscler Thromb Vasc Biol ; 33(8): 1943-51, 2013 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-23723373

RESUMO

OBJECTIVE: Toll-like receptor 2 (TLR2) inhibition by function blocking antibodies (ABs) is associated with enhanced preservation of endothelial cell function during vascular disease. In the present study, we investigated the capacity of TLR2-blocking ABs to modulate the angiogenic response of endothelial cells in vitro and in vivo. APPROACH AND RESULTS: Incubation of endothelial cells with mono- or polyclonal anti-TLR2 ABs resulted in increased tube formation, sprouting, and migration of endothelial cells compared with controls. In a mouse model of hindlimb ischemia, using TLR2-deficient or anti-TLR2 AB-treated wild-type mice resulted in increased new capillary formation and enhanced reperfusion. The effects of anti-TLR2 ABs were similar to those exerted by stromal cell-derived factor-1, and we show that anti-TLR2 ABs yet not TLR2 ligands lead to comparable activation of extracellular signal-regulated kinase1/2 and AKT but not p38 mitogen-activated protein kinase as activation of the CXCR4 canonical signal transduction pathways by stromal cell-derived factor-1. Immunoprecipitation of TLR2 revealed that anti-TLR2 ABs initiate an association of TLR2 with CXCR4 and mitogen-activated protein kinase activation. The proangiogenic properties of anti-TLR2 ABs were abolished by both G-protein inhibition and CXCR4 knockdown in endothelial cells. CONCLUSIONS: Our results provide evidence for a proangiogenic effect of TLR2-blocking ABs on endothelial cells in vitro and in vivo. They identify a novel molecular mechanism linking TLR2 to angiogenic processes that is independent from the activation of inflammatory cascades and further support the concept of a beneficial effect of TLR2 inhibition for endothelial cell function in vascular disease.


Assuntos
Anticorpos Bloqueadores/farmacologia , Sistema de Sinalização das MAP Quinases/imunologia , Neovascularização Fisiológica/imunologia , Doença Arterial Periférica/imunologia , Receptores CXCR4/metabolismo , Receptor 2 Toll-Like/imunologia , Animais , Células Cultivadas , Quimiocina CXCL12/metabolismo , Células Endoteliais/citologia , Células Endoteliais/imunologia , Células Endoteliais/metabolismo , Membro Posterior/irrigação sanguínea , Isquemia/imunologia , Isquemia/metabolismo , Isquemia/fisiopatologia , Camundongos , Camundongos Knockout , Músculo Esquelético/irrigação sanguínea , Doença Arterial Periférica/metabolismo , Doença Arterial Periférica/fisiopatologia , Proteínas Proto-Oncogênicas c-akt/metabolismo , RNA Interferente Pequeno/genética , Receptores CXCR4/genética , Receptores CXCR4/imunologia , Receptor 2 Toll-Like/genética , Receptor 2 Toll-Like/metabolismo , Proteínas Quinases p38 Ativadas por Mitógeno/metabolismo
17.
Prog Urol ; 24 Suppl 1: S20-5, 2014 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-24950928

RESUMO

Ischemia-reperfusion (IR) injury can negatively influence the short- and long-term outcomes of kidney transplantation because it promotes acute tubular necrosis and tissue scarring and activates innate alloimmunity. The adaptive responses to IR are centrally involved in reducing tissue damage but can also be deleterious when they activate programmed cell death and inflammation. The HIF-1α-mediated angiogenic responses following IR at early and late stages are complex and poorly understood. The early stages of IR seem to be associated with an antiangiogenic response, whereas the hypoxia that follows IR at later stages may activate angiogenic factors such as vascular endothelial growth factor (VEGF) and may be beneficial by stabilizing the microvasculature and favoring local blood supply. In addition to HIF-1α, new players in angiogenesis, including mTOR and the unfolded protein response, may lead to innovative therapeutic strategies for treating patients with ischemia- and reperfusion-associated tissue inflammation and organ dysfunction.


Assuntos
Transplante de Rim , Rim/irrigação sanguínea , Neovascularização Patológica/etiologia , Traumatismo por Reperfusão/complicações , Endotélio Vascular , Humanos
18.
Acta Med Philipp ; 58(10): 49-64, 2024.
Artigo em Inglês | MEDLINE | ID: mdl-38939420

RESUMO

Background: Current medical management of endometriosis leads to suppression of ovulation and will not be helpful for women with endometriosis who are desirous of pregnancy. Thus, drugs that can both treat endometriosis and its associated infertility are highly warranted. Objective: Anti-angiogenic agents are potential drugs for patients with endometriosis and infertility. Among these drugs, dopamine agonist (DA) is promising since it does not interfere with ovulation, is safe, and not teratogenic. The aim of the study is to determine the efficacy and safety of DA for improving reproductive outcomes in women with endometriosis and infertility. Methods: A qualitative narrative review was done from inception to July 31, 2022 using the appropriate MeSH terms in PubMed, Cochrane Database of Systematic Reviews, the Cochrane Central Register of Controlled Trials, ClinicalTrial.gov, and World Health Organization International Clinical Trials Registry Platform. Date analysis was through qualitative analysis and synthesis of researches and their outcome measures. Results: No studies used the core outcomes for trials evaluating treatments for infertility associated with endometriosis. All the included articles in the review supported the possible anti-angiogenic effects of DA on the vascular endothelial growth factor [VEGF] /VEGF receptor system. The use of DA does not have an effect on ovulation and menstrual cyclicity. Studies on safety profile of DA were consistent with existing data. Conclusion: Most of studies reviewed demonstrated that DA were effective in reducing endometriotic lesions. However, further research is required to establish whether this anti-angiogenic effect can improve reproductive outcomes in women with endometriosis-associated infertility.

19.
J Ethnopharmacol ; 334: 118531, 2024 Nov 15.
Artigo em Inglês | MEDLINE | ID: mdl-38971343

RESUMO

ETHNOPHARMACOLOGICAL RELEVANCE: Ginseng (Panax ginseng C. A. Mey) is a common traditional Chinese medicine used for anti-inflammation, anti-apoptosis, anti-oxidative stress, and neuroprotection. Ginsenosides Rg1, the main active components isolated from ginseng, may be a feasible therapy for spinal cord injury (SCI). AIMS OF THE STUDY: SCI causes endothelial cell death and blood vessel rupture, ultimately resulting in long-term neurological impairment. As a result, encouraging spinal angiogenesis may be a feasible therapy for SCI. This investigation aimed to validate the capacity of ginsenoside Rg1 in stimulating angiogenesis within the spinal cord. MATERIALS AND METHODS: Rats with SCI were injected intraperitoneally with ginsenoside Rg1. The effectiveness of ginsenoside Rg1 was assessed using the motor function score and the motor-evoked potential (MEP). Immunofluorescence techniques were applied to identify the spinal cord's angiogenesis. Angiogenic factors were examined through Western Blot (WB) and Immunohistochemistry. Oxygen-glucose deprivation (OGD) was employed to establish the hypoxia-ischemia model in vitro, and astrocytes (As) were given ginsenoside Rg1 and co-cultured with spinal cord microvascular endothelial cells (SCMECs). Immunofluorescence, wound healing test, and tube formation assay were used to identify the co-cultured SCMECs' activity. Finally, network pharmacology analysis and siRNA transfection were applied to verify the mechanism of ginsenoside Rg1 promoting angiogenesis. RESULTS: The rats with SCI treated with ginsenoside Rg1 indicated more significant functional recovery, more pronounced angiogenesis, and higher levels of angiogenic factor expression. In vitro, the co-culture system with ginsenoside Rg1 intervention improved SCMECs' capacity for proliferating, migrating, and forming tubes, possibly by promoting the expression of vascular endothelial growth factor (VEGF) in As via the janus kinase 2 (JAK2)/signal transducer and activator of transcription 3 (STAT3) signaling pathway. CONCLUSION: Ginsenoside Rg1 can regulate As to promote angiogenesis, which may help to understand the mechanism of promoting SCI recovery.


Assuntos
Astrócitos , Ginsenosídeos , Janus Quinase 2 , Neovascularização Fisiológica , Ratos Sprague-Dawley , Fator de Transcrição STAT3 , Transdução de Sinais , Traumatismos da Medula Espinal , Animais , Ginsenosídeos/farmacologia , Traumatismos da Medula Espinal/tratamento farmacológico , Fator de Transcrição STAT3/metabolismo , Janus Quinase 2/metabolismo , Transdução de Sinais/efeitos dos fármacos , Astrócitos/efeitos dos fármacos , Astrócitos/metabolismo , Masculino , Neovascularização Fisiológica/efeitos dos fármacos , Ratos , Células Endoteliais/efeitos dos fármacos , Células Endoteliais/metabolismo , Medula Espinal/efeitos dos fármacos , Medula Espinal/metabolismo , Células Cultivadas , Indutores da Angiogênese/farmacologia , Técnicas de Cocultura , Angiogênese
20.
Eur J Obstet Gynecol Reprod Biol ; 300: 268-277, 2024 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-39053087

RESUMO

Monitoring and timing of delivery in preterm preeclampsia and fetal growth restriction is one of the biggest challenges in Obstetrics. Finding the optimal time of delivery of these fetuses usually involves a trade-off between the severity of the disease and prematurity. So far, most clinical guidelines recommend the use of a combination between clinical, laboratory and ultrasound markers to guide the time of delivery. Angiogenic biomarkers, especially placental growth factor (PlGF) and soluble fms-like tyrosine kinase-1 (sFlt-1), have gained significant attention in recent years for their potential role in the prediction and diagnosis of placenta-related disorders including preeclampsia and fetal growth restriction. Another potential clinical application of the angiogenic biomarkers is for the differential diagnosis of patients with chronic kidney disease, as this condition shares similar clinical features with preeclampsia. Consequently, angiogenic biomarkers have been advocated as tools for monitoring and deciding the optimal time of the delivery of fetuses affected by placental dysfunction. In this clinical opinion, we critically review the available literature on PlGF and sFlt-1 for the surveillance and time of the delivery in fetuses affected by preterm preeclampsia and fetal growth restriction. Moreover, we explore the use of angiogenic biomarkers for the differentiation between chronic kidney disease and superimposed preeclampsia.


Assuntos
Biomarcadores , Retardo do Crescimento Fetal , Fator de Crescimento Placentário , Pré-Eclâmpsia , Receptor 1 de Fatores de Crescimento do Endotélio Vascular , Humanos , Feminino , Gravidez , Pré-Eclâmpsia/diagnóstico , Pré-Eclâmpsia/sangue , Retardo do Crescimento Fetal/diagnóstico , Retardo do Crescimento Fetal/sangue , Biomarcadores/sangue , Fator de Crescimento Placentário/sangue , Receptor 1 de Fatores de Crescimento do Endotélio Vascular/sangue
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA