Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 1.330
Filter
1.
J BUON ; 26(3): 663-669, 2021.
Article in English | MEDLINE | ID: mdl-34268918

ABSTRACT

PURPOSE: To estimate the expression and clinical significance of miR-139-5p and fibroblast growth factor 2 (FGF2) in ovarian cancer (OC). METHODS: Of the 198 female patients undergoing surgical treatment in our hospital, 101 patients with ovarian tumor resection were allocated in a study group and 97 with ovarian resection for benign lesions were allocated in a control group. MiR-139-5p and FGF2 expression was quantified, and associations between miR-139-5p and FGF2 and clinicopathological features of OC were analyzed, as well as their diagnostic performances (receiver operating characteristic (ROC) curve). RESULTS: The study group presented lower miR-139-5p level and higher FGF2 level (both p<0.05). Significant associations of miR-139-5p and FGF2 with tumor differentiation and clinical stage were noted in OC (p<0.05). MiR-139-5p was reversely associated with clinical stage and positively associated with tumor differentiation (p<0.05), FGF2 was positively correlated with clinical stage and negatively correlated with tumor differentiation (p<0.05). The overall survival in the study group was 70.41%. The survival in high miR-139-5p expression group and low FGF2 expression group improved remarkably (p<0.05). The area under the curve (AUC) of combined detection (0.91) was higher than that of single detection. CONCLUSION: MiR-139-5p shows a decreased expression and FGF-2 shows an increased expression in OC, and they are associated with clinical stage and tumor differentiation. Combined detection of miR-139-5p and FGF-2 contributes to the diagnosis and treatment of OC, and is an available biomarker for the diagnosis and prognosis of patients.


Subject(s)
Fibroblast Growth Factor 2/biosynthesis , MicroRNAs/biosynthesis , Ovarian Neoplasms/metabolism , Female , Fibroblast Growth Factor 2/physiology , Humans , MicroRNAs/physiology , Middle Aged
2.
Iran J Immunol ; 18(2): 103-110, 2021 06.
Article in English | MEDLINE | ID: mdl-34190691

ABSTRACT

BACKGROUND: Interleukin (IL)-17A possesses biological activities to promote vascular endothelial cell migration and microvessel development. OBJECTIVE: To clarify which angiogenic factors are involved in IL-17A-modified angiogenesis-related functions of vascular endothelial cell migration and microtube development or not. METHODS: The potential contribution of various angiogenic stimulators to in vitro angiogenic activities of IL-17A was assessed with both modified Boyden Chemotaxicell chamber assay and in vitro angiogenesis assay. RESULTS: The addition of a neutralizing antibody (Ab) for hepatocyte growth factor (HGF), basic fibroblast growth factor (bFGF) or vascular endothelial growth factor (VEGF)-A to the upper and lower compartments in a modified Boyden Chemotaxicell chamber significantly attenuated human dermal microvascular endothelial cell (HMVEC) migration elicited by IL-17A. Moreover, IL-17A-induced capillary-like microvessel development in human umbilical vein endothelial cell (HUVEC) and human dermal fibroblast (HDF) co-culture system was significantly impaired by a neutralizing Ab against HGF, bFGF, VEGF-A, cysteine-x-cysteine ligand 8 (CXCL8)/IL-8 or cysteine-x-cysteine (CXC) chemokine receptor (CXCR)-2. CONCLUSION: Our findings demonstrate the involvement of HGF, bFGF, VEGF-A and/or CXCL8/IL-8, to various degrees, in migration and microvessel development of vascular endothelial cells mediated by IL-17A.


Subject(s)
Endothelial Cells/drug effects , Fibroblast Growth Factor 2/physiology , Hepatocyte Growth Factor/physiology , Interleukin-17/pharmacology , Neovascularization, Physiologic/drug effects , Vascular Endothelial Growth Factor A/physiology , Capillaries/physiology , Cell Movement/drug effects , Cells, Cultured , Endothelial Cells/physiology , Humans , Interleukin-8/physiology , Neovascularization, Physiologic/physiology
3.
Int J Mol Sci ; 22(9)2021 Apr 29.
Article in English | MEDLINE | ID: mdl-33946884

ABSTRACT

Triple-negative breast cancer (TNBC) is an aggressive breast tumor subtype characterized by poor clinical outcome. In recent years, numerous advancements have been made to better understand the biological landscape of TNBC, though appropriate targets still remain to be determined. In the present study, we have determined that the expression levels of FGF2 and S100A4 are higher in TNBC with respect to non-TNBC patients when analyzing "The Invasive Breast Cancer Cohort of The Cancer Genome Atlas" (TCGA) dataset. In addition, we have found that the gene expression of FGF2 is positively correlated with S100A4 in TNBC samples. Performing quantitative PCR, Western blot, CRISPR/Cas9 genome editing, promoter studies, immunofluorescence analysis, subcellular fractionation studies, and ChIP assays, we have also demonstrated that FGF2 induces in TNBC cells the upregulation and secretion of S100A4 via FGFR1, along with the ERK1/2-AKT-c-Rel transduction signaling. Using conditioned medium from TNBC cells stimulated with FGF2, we have also ascertained that the paracrine activation of the S100A4/RAGE pathway triggers angiogenic effects in vascular endothelial cells (HUVECs) and promotes the migration of cancer-associated fibroblasts (CAFs). Collectively, our data provide novel insights into the action of the FGF2/FGFR1 axis through S100A4 toward stimulatory effects elicited in TNBC cells.


Subject(s)
Fibroblast Growth Factor 2/physiology , Neoplasm Proteins/physiology , Receptor, Fibroblast Growth Factor, Type 1/physiology , S100 Calcium-Binding Protein A4/physiology , Signal Transduction/physiology , Triple Negative Breast Neoplasms/physiopathology , Antigens, Neoplasm/physiology , Cell Movement/drug effects , Culture Media, Conditioned/pharmacology , Female , Fibroblast Growth Factor 2/pharmacology , Fibroblasts/pathology , Gene Expression Regulation, Neoplastic/physiology , Human Umbilical Vein Endothelial Cells , Humans , Mitogen-Activated Protein Kinases/physiology , Neovascularization, Pathologic/physiopathology , Paracrine Communication , Protein Kinase Inhibitors/pharmacology , Proto-Oncogene Proteins c-rel/physiology , Receptor, Fibroblast Growth Factor, Type 1/antagonists & inhibitors , Receptor, Fibroblast Growth Factor, Type 1/genetics , Signal Transduction/drug effects , Triple Negative Breast Neoplasms/blood supply , Tumor Cells, Cultured
4.
J Surg Res ; 259: 261-270, 2021 03.
Article in English | MEDLINE | ID: mdl-33162101

ABSTRACT

BACKGROUND: Keloid is a benign fibroproliferative tumor of the skin caused by abnormal wound healing process after skin injury. Long noncoding RNAs have been reported to be involved in the development of keloid. However, the role and mechanism of nuclear enriched abundant transcript 1 (NEAT1) in keloid are still unknown. METHODS: Quantitative real-time polymerase chain reaction was performed to detect the expression of NEAT1, miR-196b-5p, and fibroblast growth factor 2 (FGF2). Western blot was conducted to measure the levels of collagen I, α-smooth muscle actin, fibronectin, and FGF2. Cell Counting Kit-8 assay and transwell assay were used to evaluate cell viability and migration, respectively. Dual-luciferase reporter assay was conducted to verify the targeting relationship between miR-196b-5p and NEAT1 or FGF2. RESULTS: NEAT1 was increased and miR-196b-5p was decreased in keloid tissues and fibroblasts. NEAT1 knockdown or miR-196b-5p overexpression suppressed cell viability, migration, and extracellular matrix (ECM) component production in keloid fibroblasts. MiR-196 b-5p was a target of NEAT1, and NEAT1 overexpression reversed the effect of miR-196b-5p on keloid fibroblast progression. Moreover, we found that miR-196b-5p directly targeted FGF2. FGF2 knockdown suppressed keloid fibroblast viability, migration, and ECM protein production. FGF2 overexpression abolished the effect of miR-196b-5p overexpression on keloid fibroblast development. CONCLUSIONS: NEAT1 silencing suppressed cell viability, migration, and ECM expression in keloid fibroblasts by regulating miR-196b-5p/FGF2 axis, indicating a promising strategy for keloid treatment.


Subject(s)
Fibroblast Growth Factor 2/physiology , Keloid/pathology , MicroRNAs/physiology , RNA, Long Noncoding/physiology , Cell Movement , Cell Survival , Cells, Cultured , Extracellular Matrix Proteins/biosynthesis , Fibroblasts/physiology , Humans , Keloid/metabolism
5.
Neuropharmacology ; 170: 108070, 2020 06 15.
Article in English | MEDLINE | ID: mdl-32229140

ABSTRACT

The majority of the fibroblast growth factor receptor 1-serotonin 1 A receptor (FGFR1-5-HT1AR) heterocomplexes in the hippocampus appeared to be located mainly in the neuronal networks and a relevant target for antidepressant drugs. Through a neurochemical and electrophysiological analysis it was therefore tested in the current study if astrocytic FGFR1-5-HT1AR heterocomplexes also exist in hippocampus. They may modulate the structure and function of astroglia in the hippocampus leading to possible changes in the gamma oscillations. Localization of hippocampal FGFR1-5-HT1AR heterocomplexes in astrocytes was found using in situ proximity ligation assay combined with immunohistochemistry using glial fibrillary acidic protein (GFAP) immunoreactivity as a marker for astroglia. Acute i.c.v. treatment with 8-OH-DPAT alone or together with basic fibroblast growth factor (FGF2) significantly increased FGFR1-5-HT1AR heterocomplexes in the GFAP positive cells, especially in the polymorphic layer of the dentate gyrus (PoDG) but also in the CA3 area upon combined treatment. No other hippocampal regions were studied. Also, structural plasticity changes were observed in the astrocytes, especially in the PoDG region, upon these pharmacological treatments. They may also be of relevance for enhancing the astroglial volume transmission with increased modulation of the neuronal networks in the regions studied. The effects of combined FGF2 and 5-HT agonist treatments on gamma oscillations point to a significant antagonistic interaction in astroglial FGFR1-5-HT1AR heterocomplexes that may contribute to counteraction of the 5-HT1AR-mediated decrease of gamma oscillations. This article is part of the special issue entitled 'Serotonin Research: Crossing Scales and Boundaries'.


Subject(s)
Astrocytes/physiology , Fibroblast Growth Factor 2/physiology , Gamma Rhythm/physiology , Hippocampus/physiology , Receptor, Fibroblast Growth Factor, Type 1/physiology , Receptor, Serotonin, 5-HT1A/physiology , Serotonin/physiology , Animals , Astrocytes/drug effects , Gamma Rhythm/drug effects , Hippocampus/drug effects , Male , Organ Culture Techniques , Rats , Rats, Sprague-Dawley , Receptor, Fibroblast Growth Factor, Type 1/agonists , Serotonin 5-HT1 Receptor Agonists/pharmacology
6.
Hum Gene Ther ; 31(5-6): 286-296, 2020 03.
Article in English | MEDLINE | ID: mdl-32013585

ABSTRACT

Osteonecrosis of the femoral head (ONFH) is a common and disabling joint disease. Although there is no clear consensus on the complex pathogenic mechanism of ONFH, trauma, abuse of glucocorticoids, and alcoholism are implicated in its etiology. The therapeutic strategies are still limited, and the clinical outcomes are not satisfactory. Mesenchymal stem cells (MSCs) have been shown to exert a positive impact on ONFH in preclinical experiments and clinical trials. The beneficial properties of MSCs are due, at least in part, to their ability to home to the injured tissue, secretion of paracrine signaling molecules, and multipotentiality. Nevertheless, the regenerative capacity of transplanted cells is impaired by the hostile environment of necrotic tissue in vivo, limiting their clinical efficacy. Recently, genetic engineering has been introduced as an attractive strategy to improve the regenerative properties of MSCs in the treatment of early-stage ONFH. This review summarizes the function of several genes used in the engineering of MSCs for the treatment of ONFH. Further, current challenges and future perspectives of genetic manipulation of MSCs are discussed. The notion of genetically engineered MSCs functioning as a "factory" that can produce a significant amount of multipotent and patient-specific therapeutic product is emphasized.


Subject(s)
Femur Head Necrosis/genetics , Femur Head Necrosis/therapy , Genetic Therapy/methods , Mesenchymal Stem Cells/physiology , Animals , Chemokines/physiology , Fibroblast Growth Factor 2/physiology , Genetic Engineering , Hepatocyte Growth Factor/physiology , Humans , Hypoxia-Inducible Factor 1, alpha Subunit/physiology , Intercellular Signaling Peptides and Proteins/physiology , Osteogenesis , Vascular Endothelial Growth Factor A/physiology
7.
Sci Rep ; 10(1): 739, 2020 01 20.
Article in English | MEDLINE | ID: mdl-31959818

ABSTRACT

Human urine is a non-invasive source of renal stem cells with regeneration potential. Urine-derived renal progenitor cells were isolated from 10 individuals of both genders and distinct ages. These renal progenitors express pluripotency-associated proteins- TRA-1-60, TRA-1-81, SSEA4, C-KIT and CD133, as well as the renal stem cell markers -SIX2, CITED1, WT1, CD24 and CD106. The transcriptomes of all SIX2+ renal progenitors clustered together, and distinct from the human kidney biopsy-derived epithelial proximal cells (hREPCs). Stimulation of the urine-derived renal progenitor cells (UdRPCs) with the GSK3ß-inhibitor (CHIR99021) induced differentiation. Transcriptome and KEGG pathway analysis revealed upregulation of WNT-associated genes- AXIN2, JUN and NKD1. Protein interaction network identified JUN- a downstream target of the WNT pathway in association with STAT3, ATF2 and MAPK1 as a putative negative regulator of self-renewal. Furthermore, like pluripotent stem cells, self-renewal is maintained by FGF2-driven TGFß-SMAD2/3 pathway. The urine-derived renal progenitor cells and the data presented should lay the foundation for studying nephrogenesis in human.


Subject(s)
Cell Self Renewal/genetics , Cell Self Renewal/physiology , Fibroblast Growth Factor 2/physiology , Kidney/cytology , Pluripotent Stem Cells/physiology , Signal Transduction/genetics , Signal Transduction/physiology , Transforming Growth Factor beta/physiology , Urine/cytology , Wnt Signaling Pathway/genetics , Wnt Signaling Pathway/physiology , Cell Differentiation/genetics , Female , Fibroblast Growth Factor 2/metabolism , Gene Expression , Humans , Male , Pluripotent Stem Cells/metabolism , Transcriptome , Transforming Growth Factor beta/metabolism
8.
J Neurosci ; 40(1): 12-21, 2020 01 02.
Article in English | MEDLINE | ID: mdl-31896560

ABSTRACT

Over the last 50 years, the concept of stress has evolved significantly, and our understanding of the underlying neurobiology has expanded dramatically. Rather than consider stress biology to be relevant only under unusual and threatening conditions, we conceive of it as an ongoing, adaptive process of assessing the environment, coping with it, and enabling the individual to anticipate and deal with future challenges. Though much remains to be discovered, the fundamental neurocircuitry that underlies these processes has been broadly delineated, key molecular players have been identified, and the impact of this system on neuroplasticity has been well established. More recently, we have come to appreciate the critical interaction between the brain and the rest of the body as it pertains to stress responsiveness. Importantly, this system can become overloaded due to ongoing environmental demands on the individual, be they physical, physiological, or psychosocial. The impact of this overload is deleterious to brain health, and it results in vulnerability to a range of brain disorders, including major depression and cognitive deficits. Thus, stress biology is one of the best understood systems in affective neuroscience and is an ideal target for addressing the pathophysiology of many brain-related diseases. The story we present began with the discovery of glucocorticoid receptors in hippocampus and has extended to other brain regions in both animal models and the human brain with the further discovery of structural and functional adaptive plasticity in response to stressful and other experiences.


Subject(s)
Brain/physiology , Glucocorticoids/physiology , Mood Disorders/physiopathology , Stress, Physiological/physiology , Stress, Psychological/physiopathology , Adaptation, Physiological/physiology , Animals , Endocannabinoids/physiology , Epigenesis, Genetic , Feedback, Physiological , Fibroblast Growth Factor 2/physiology , Fibroblast Growth Factor 2/therapeutic use , Gene Expression Regulation/physiology , Hormones/physiology , Humans , Hypothalamo-Hypophyseal System/physiology , Intercellular Signaling Peptides and Proteins/physiology , Life Change Events , Models, Neurological , Models, Psychological , Mood Disorders/etiology , Mood Disorders/psychology , Nerve Tissue Proteins/physiology , Neuronal Plasticity , Pituitary-Adrenal System/physiology , Psychophysiology , Receptors, Cell Surface/physiology , Social Determinants of Health
9.
Gene ; 725: 144143, 2020 01 30.
Article in English | MEDLINE | ID: mdl-31629816

ABSTRACT

Atherosclerosis is a common cardiovascular disorder and is characterized by damage of endothelial cells, cell inflammation, hyper-proliferation of vascular smooth muscle cells and the accumulation of extracellular lipids and fibrous tissues. In this study, we firstly examined the expression level of long intergenic non-protein coding RNA, regulator of reprogramming (linc-ROR) in homocysteine (Hcy)-stimulated human aortic smooth muscle cells (HASMCs), and then looked into the potential molecular signaling axis of linc-ROR in regulating the proliferation and migration of HASMCs. Hcy promoted HASMC proliferation and up-regulated linc-ROR expression. Functional studies showed that linc-ROR exerted enhanced actions on the proliferation and migration of HASMCs. In addition, linc-ROR acted as a competing endogenous RNA for miR-195-5p and repressed the miR-195-5p expression in HASMCs. Linc-ROR was up-regulated the miR-195-3p was down-regulated in the plasma from CAD patients when compared to normal controls. Furthermore, fibroblast growth factor 2 (FGF2) was identified as a target of miR-195-5p and was negatively regulated by miR-195-5p in HASMCs. The rescue experiments revealed that linc-ROR-mediated HASMC proliferation and migration may be via regulating miR-195-5p/FGF2 axis. Linc-ROR inhibition blocked the miR-195-5p/FGF2 signaling in Hcy-treated HASMCs, and this effect may also involve in the miR-195-5p/FGF2 axis. To summarize, the data of the present study identified the up-regulation of linc-ROR in Hcy-stimulated HASMCs, and further mechanistic functional studies revealed that linc-ROR promoted HASMC proliferation and migration via regulating miR-195-5p/FGF2 axis. The present study provided the novel actions of linc-ROR in regulating HASMC proliferation and migration, which may be related to the pathophysiology of atherosclerosis.


Subject(s)
Fibroblast Growth Factor 2/metabolism , MicroRNAs/metabolism , RNA, Long Noncoding/metabolism , Aorta/cytology , Aorta/drug effects , Aorta/metabolism , Atherosclerosis/genetics , Atherosclerosis/metabolism , Atherosclerosis/pathology , Autophagy/physiology , Cell Line, Tumor , Cell Movement/physiology , Cell Proliferation/drug effects , Cell Proliferation/physiology , Endothelial Cells/drug effects , Endothelial Cells/metabolism , Fibroblast Growth Factor 2/genetics , Fibroblast Growth Factor 2/physiology , Homocysteine/pharmacology , Humans , MicroRNAs/genetics , Myocytes, Smooth Muscle/cytology , Myocytes, Smooth Muscle/drug effects , Myocytes, Smooth Muscle/metabolism , RNA, Long Noncoding/biosynthesis , RNA, Long Noncoding/genetics , Signal Transduction
10.
Life Sci ; 242: 117213, 2020 Feb 01.
Article in English | MEDLINE | ID: mdl-31881228

ABSTRACT

Acute respiratory distress syndrome (ARDS) is a multifactorial, inflammatory lung injury disease with high morbidity and mortality. However, the underlying pathogenic mechanism remains unknown. In this study, lipopolysaccharide (LPS)-stimulated alveolar epithelial cells were used to mimic the inflammatory pathogenesis of ARDS in vitro. We here investigated the role of miR-424 in LPS-stimulated alveolar epithelial cells and found it to be substantially downregulated. Overexpression of miR-424 inhibited apoptosis and inflammation in LPS-stimulated alveolar epithelial cells, and the miR-424 inhibitor exhibited the opposite effect. A bioinformatic analysis revealed a potential binding site of miR-424 in the 3'-UTR of fibroblast growth factor 2 (FGF2). A luciferase reporter assay suggested that miR-424 targeted FGF2 in alveolar epithelial cells. The level of FGF2 protein was inhibited by miR-424 mimic, whereas was significantly upregulated after miR-424 suppression in LPS-stimulated alveolar epithelial cells. MiR-424 also exhibited the protective role in LPS-induced apoptosis and inflammation by directly targeting FGF2 via the NF-κB pathway. In conclusion, our results demonstrate that miR-424 had a protective role in LPS-induced apoptosis and inflammation of alveolar epithelial cells by targeting FGF2 via regulating NF-κB pathway. This might contribute novel evidence to help identify a therapeutic target for treating ARDS.


Subject(s)
A549 Cells/metabolism , Apoptosis/drug effects , Fibroblast Growth Factor 2/physiology , Inflammation/physiopathology , Lipopolysaccharides/pharmacology , MicroRNAs/metabolism , NF-kappa B/metabolism , Pulmonary Alveoli/metabolism , Respiratory Mucosa/metabolism , Signal Transduction , A549 Cells/physiology , Apoptosis/physiology , Blotting, Western , Fibroblast Growth Factor 2/metabolism , Fluorescent Antibody Technique , Humans , Inflammation/metabolism , MicroRNAs/physiology , Pulmonary Alveoli/cytology , Pulmonary Alveoli/physiology , Real-Time Polymerase Chain Reaction , Respiratory Mucosa/cytology , Respiratory Mucosa/physiology , Signal Transduction/physiology
11.
Arch Oral Biol ; 109: 104574, 2020 Jan.
Article in English | MEDLINE | ID: mdl-31585238

ABSTRACT

Fibroblast growth factors (FGFs) are growth factors that play an important role in tooth development, repair, and regeneration. Of the FGF families, basic fibroblast growth factor (bFGF) has been the most frequently investigated in dentistry. Numerous studies have reported advantages of bFGF, while others did not find any additional benefit. This review gives a comprehensive summary of the potential role of bFGF in dental pulp wound healing and regeneration in connection with cell proliferation and differentiation, angiogenesis, and neural differentiation from both in vitro and in vivo studies. Furthermore, the possible underlying mechanisms associated with bFGF in promoting dental pulp wound healing are discussed in this review.


Subject(s)
Dental Pulp/physiology , Fibroblast Growth Factor 2/physiology , Animals , Cell Differentiation , Cell Proliferation , Humans , Neovascularization, Physiologic , Odontogenesis , Regeneration , Wound Healing
12.
Med Sci Monit ; 25: 7191-7201, 2019 Sep 25.
Article in English | MEDLINE | ID: mdl-31551405

ABSTRACT

BACKGROUND Disruption of the blood-brain barrier (BBB) is a mechanism in the pathogenesis of traumatic brain injury. Basic fibroblast growth factor (bFGF) is expressed in angiogenesis, neurogenesis, and neuronal survival. This study aimed to investigate the role of bFGF in vitro in human brain microvascular endothelial cells (HBMECs) challenged by oxygen-glucose deprivation/reperfusion (OGD/R). MATERIAL AND METHODS HBMECs were cultured in glucose-free medium and an environment with <0.5% oxygen in an anaerobic chamber. Immunocytochemistry, Western blot, and quantitative reverse transcription-polymerase chain reaction (qRT-PCR) were used to measure the protein and mRNA expression levels of bFGF, tight junction, adherens junction, apoptotic proteins, and matrix metalloproteinases (MMPs). The effects of bFGF on the viability of HBMECs was evaluated using the cell counting kit-8 (CCK-8) assay. Cell apoptosis was evaluated using the TUNEL assay, and endothelial permeability was quantified using a transwell migration assay with fluorescein isothiocyanate (FITC) conjugated with dextran. The effects of bFGF were evaluated following inhibition of fibroblast growth factor receptor 1 (FGFR1) with PD173074 and inhibition of ERK with PD98059. RESULTS Following OGD/R of HBMECs, bFGF significantly reduced cell permeability and apoptosis and significantly inhibited the down-regulation of the expressions of proteins associated with tight junctions, adherens junctions, apoptosis and matrix metalloproteinases (MMPs). The effects of bFGF were mediated by the activation of FGFR1 and ERK, as they were blocked by FGFR1 and ERK inhibitors. CONCLUSIONS Permeability and apoptosis of HBMECs challenged by OGD/R were reduced by bFGF by activation of the FGFR1 and the ERK pathway.


Subject(s)
Capillary Permeability/physiology , Endothelial Cells/drug effects , Fibroblast Growth Factor 2/metabolism , Apoptosis/drug effects , Blood-Brain Barrier/metabolism , Blood-Brain Barrier/pathology , Brain/pathology , Cell Survival/drug effects , Endothelial Cells/physiology , Fibroblast Growth Factor 2/physiology , Glucose/metabolism , Humans , Hypoxia/metabolism , MAP Kinase Signaling System/drug effects , Oxygen/metabolism , Primary Cell Culture/methods , Receptor, Fibroblast Growth Factor, Type 1/metabolism , Signal Transduction/physiology
13.
Behav Brain Res ; 374: 112113, 2019 11 18.
Article in English | MEDLINE | ID: mdl-31381976

ABSTRACT

BACKGROUND: Loss of fibroblast growth factor 2 (FGF-2) is responsible for the development of an increased number of dopaminergic (DA) neurons in the murine substantia nigra pars compacta (SNpc). Furthermore, dysregulation of its expression patterns within the central nervous system (CNS) is associated with behavioral abnormalities in mice. Until now, the contributions of the individual FGF-2 isoforms (one low (LMW) and two high molecular weight (HMW) isoforms) in the CNS are elusive. METHODS: To unravel the specific effects of FGF-2 isoforms, we compared three knockout mouse lines, one only deficient for LMW, one deficient for HMW and another lacking both isoforms, regarding DA neuronal development. With this regard, three time points of ontogenic development of the SNpc were stereologically investigated. Furthermore, behavioral aspects were analyzed in young adult mice, supplemented by corticosterone measurements. RESULTS: Juvenile mice lacking either LMW or HMW develop equal supernumerary DA neuron numbers in the SNpc. Compensatory increased LMW expression is observed in animals lacking HMW. Meanwhile, no knockout mouse line demonstrated changes in anxiety-like behavior, stress susceptibility, or locomotor behavior. CONCLUSIONS: Both FGF-2 isoforms crucially influence DA neuronal development in the murine SNpc. However, absence of LMW or HMW alone alters neither anxiety-like nor locomotor behavior, or stress susceptibility. Therefore, FGF-2 is not a determinant and causative factor for behavioral alterations alone, but probably in combination with appropriate conditions, like environmental or genetic factors.


Subject(s)
Dopaminergic Neurons/metabolism , Fibroblast Growth Factor 2/metabolism , Pars Compacta/metabolism , Animals , Anxiety/metabolism , Anxiety/physiopathology , Dopaminergic Neurons/physiology , Fibroblast Growth Factor 2/physiology , Fibroblast Growth Factors/metabolism , Fibroblast Growth Factors/physiology , Locomotion/physiology , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Neurogenesis , Protein Isoforms/genetics , Substantia Nigra/metabolism
14.
Theriogenology ; 139: 90-97, 2019 Nov.
Article in English | MEDLINE | ID: mdl-31400551

ABSTRACT

Basic fibroblast growth factor (bFGF) plays a pivotal role in prompting ovarian follicular development and angiogenesis as well as inhibiting atresia. In the chicken, high laying performance depends largely on efficient healthy development of ovarian follicles. Moreover, rapid growth of oocytes resulted from abundant yolk deposition via blood circulation and intra-ovarian interactions among somatic and germ cells. The major components of yolk mass consist of very low density lipoprotein (VLDL) and vitellogenin that are taken up by maturing oocytes via VLDL receptor (VLDLR)-mediated endocytosis from blood capillaries in the theca layer and gaps between granulosa cells. Here we used immunofluorescence, BrdU, TUNEL, Western bolt and RT-qPCR methods to investigate effects of bFGF on growth and yolk deposition of chicken prehierarchical follicles. Results showed that VLDLR was mainly expressed in the granulosa cells of the prehierarchical and preovulatory follicles, and its expression declined with follicle growth. Moreover, bFGF caused a dose-dependent promoting effect on growth of small white follicles and this effect was inhibited by SU5402 (an FGFR1 antagonist). Proliferation of follicular theca externa cells was accelerated by bFGF via FGFR1-AKT signaling, coupled with augmented angiogenesis and up-regulated p-ERK expression in granulosa cells. After combined inhibition of FGFR1 and PPARγ, we found that PPARγ could also suppress VLDLR expression in granulosa cells. These results indicate that bFGF facilitated growth and yolk deposition in chicken prehierarchical follicles through promoting proliferation and angiogenesis in theca layers, and also through down-regulating VLDLR expression in granulosa cells.


Subject(s)
Chickens/physiology , Egg Yolk/physiology , Fibroblast Growth Factor 2/physiology , Ovarian Follicle/growth & development , Animals , Female , Fibroblast Growth Factor 2/metabolism , Oocytes/growth & development , Oocytes/metabolism
15.
Eur Rev Med Pharmacol Sci ; 22(24): 8866-8876, 2018 12.
Article in English | MEDLINE | ID: mdl-30575929

ABSTRACT

OBJECTIVE: The aim of this study was to investigate whether hypoxia in vivo can induce hypoxic pulmonary hypertension by inhibiting the activation of FGF2 by miR-203. MATERIALS AND METHODS: We established a rat model of hypoxic pulmonary hypertension (HPH), and measured the right ventricular systolic pressure (RVSP) and right ventricular hypertrophy (right ventricular hypertrophy index). The ventricular hypertrophy index (RVHI) was calculated and HE staining of the lung tissue of HPH rats was performed. We extracted pulmonary arterial smooth muscle cells (PASMCs) from rats and identified them by immunofluorescence assay. The expression of miR-203 in hypoxic PASMCs was detected by quantitative Real time-polymerase chain reaction (qRT-PCR). The proliferation and migration of PASMCs were detected by EDU (5-Ethynyl-2'-deoxyuridine), cell counting kit-8 (CCK-8) and scratch assay, respectively. Dual Luciferase reporting assay and Western blot were used to detect the binding of miR-203 and FGF2. RESULTS: The results of qRT-PCR showed that miR-203 expression in rat PASMCs was significantly lower than that in normoxia control group at 24 h and 48 h after hypoxic treatment. EDU, CCK8 and scratch test results showed that proliferation and migration ability of PASMCs were weakened after overexpression of miR-203, and vice versa. Dual Luciferase reporter gene assays and Western blot experiments showed that miR-203 could target and combine with FGF2 to inhibit its expression. In vivo experiments showed that low expression of FGF2 could lead to decreased RVSP and RVHI, decreased FGF2 protein levels, and decreased WT% and (PM+FM)% in hypoxia-treated rats. CONCLUSIONS: Hypoxia in vivo is involved in the development of HPH by inhibiting the activation of FGF2 by miR-203. Meanwhile, specific inhibition of FGF2 can reduce hypoxia-induced pulmonary hypertension and improve pulmonary vascular remodeling.


Subject(s)
Fibroblast Growth Factor 2/antagonists & inhibitors , Hypertension, Pulmonary/etiology , Hypoxia/complications , MicroRNAs/physiology , Animals , Cells, Cultured , Fibroblast Growth Factor 2/physiology , Male , Myocytes, Smooth Muscle/physiology , Pulmonary Artery/cytology , Rats , Rats, Sprague-Dawley
16.
Dev Dyn ; 247(11): 1186-1198, 2018 11.
Article in English | MEDLINE | ID: mdl-30295986

ABSTRACT

BACKGROUND: The tissue-specific transcriptional programs during normal development require tight control by distal cis-regulatory elements, such as enhancers, with specific DNA sequences recognized by transcription factors, coactivators, and chromatin remodeling enzymes. Gata3 is a sequence-specific DNA-binding transcription factor that regulates formation of multiple tissues and organs, including inner ear, lens, mammary gland, T-cells, urogenital system, and thyroid gland. In the eye, Gata3 has a highly restricted expression domain in the posterior part of the lens vesicle; however, the underlying regulatory mechanisms are unknown. RESULTS: Here we describe the identification of a novel bipartite Gata3 lens-specific enhancer located ∼18 kb upstream from its transcriptional start site. We also found that a 5-kb Gata3 promoter possesses low activity in the lens. The bipartite enhancer contains arrays of AP-1, Ets-, and Smad1/5-binding sites as well as binding sites for lens-associated DNA-binding factors. Transient transfection studies of the promoter with the bipartite enhancer showed enhanced activation by BMP4 and FGF2. CONCLUSIONS: These studies identify a novel distal enhancer of Gata3 with high activity in lens and indicate that BMP and FGF signaling can up-regulate expression of Gata3 in differentiating lens fiber cells through the identified Gata3 enhancer and promoter elements. Developmental Dynamics 247:1186-1198, 2018. © 2018 The Authors. Developmental Dynamics published by Wiley Periodicals, Inc. on behalf of American Association of Anatomists.


Subject(s)
Enhancer Elements, Genetic , GATA3 Transcription Factor/genetics , Lens, Crystalline/embryology , Animals , Binding Sites , Bone Morphogenetic Protein 4/physiology , DNA-Binding Proteins , Fibroblast Growth Factor 2/physiology , GATA3 Transcription Factor/chemistry , GATA3 Transcription Factor/metabolism , Mice , Promoter Regions, Genetic , Transcriptional Activation
17.
Biomed Pharmacother ; 108: 1282-1288, 2018 Dec.
Article in English | MEDLINE | ID: mdl-30372829

ABSTRACT

Keloid is a fibrous benign tumor of the skin caused by increased fibroblast proliferation and overproduction of extracellular matrix (ECM) in the dermis. Several miRNAs exhibit critical roles in regulating keloid development. This is study aimed to investigate the effects and mechanisms of miR-203 in keloid fibroblasts. The miR-203 expression was detected by qRT-PCR; The cell viability was measured by MTT assay; The cell proliferation was measured by BrdU assay; The cell invasion was measured by Transwell assay; The protein expression was detected by Western blot; The target relationship between miR-203 and mRNA was measured by dual-luciferase assay. We found that miR-203 was significantly downregulated in both keloid tissues and keloid fibroblasts from keloid patients. MiR-203 overexpression in vitro led to a significant decrease of proliferation, invasion, and ECM production in keloid fibroblasts, whereas miR-203 inhibition induced the opposite results. A dual-luciferase reporter assay identified early growth response 1 (EGR1) and fibroblast growth factor 2 (FGF2) as targets of miR-203. EGR1 and FGF2 were overexpressed in keloid fibroblasts and negatively regulated by miR-203. Furthermore, overexpression of EGR1 and FGF2 partially attenuated the suppressive effect of miR-203 on the proliferation, invasion, and ECM production of keloid fibroblasts. In conclusion, we demonstrated for the first time that miR-203 decreased the proliferation, invasion, and ECM production of keloid fibroblasts by repressing EGR1 and FGF2 expression, suggesting a potential role of miR-203 in preventing and treating keloids.


Subject(s)
Early Growth Response Protein 1/genetics , Extracellular Matrix Proteins/biosynthesis , Fibroblast Growth Factor 2/genetics , Keloid/pathology , MicroRNAs/physiology , 3' Untranslated Regions , Adult , Cell Proliferation , Cells, Cultured , Early Growth Response Protein 1/physiology , Female , Fibroblast Growth Factor 2/physiology , Fibroblasts/physiology , Humans , Keloid/metabolism , Male , Middle Aged
18.
Chin Med J (Engl) ; 131(17): 2089-2096, 2018 Sep 05.
Article in English | MEDLINE | ID: mdl-30127219

ABSTRACT

BACKGROUND: Fibroblasts were the main seed cells in the studies of tissue engineering of the pelvic floor ligament. Basic fibroblast growth factor (bFGF) and epidermal growth factor (EGF) were widely studied but at various concentrations. This study aimed to optimize the concentrations of combined bFGF and EGF by evaluating their effects on proliferation and collagen secretion of fibroblasts. METHODS: Fibroblasts were differentiated from rat adipose mesenchymal stem cells (ADSCs). Flow cytometry and immunohistochemistry were used for cell identification. The growth factors were applied at concentrations of 0, 1, 10, and 100 ng/ml as three groups: (1) bFGF alone, (2) EGF alone, and (3) bFGF mixed with EGF. Cell proliferation was evaluated by Cell Counting Kit-8 assays. Expression of Type I and III collagen (Col-I and Col-III) mRNAs was evaluated by real-time quantitative reverse transcription-polymerase chain reaction. Statistical analysis was performed with SPSS software and GraphPad Prism using one-way analysis of variance and multiple t-test. RESULTS: ADSCs were successfully isolated from rat adipose tissue as identified by expression of typical surface markers CD29, CD44, CD90, and CD45 in flow cytometry. Fibroblasts induced from ADSC, compared with ADSCs, were with higher mRNA expression levels of Col I and Col III (F = 1.29, P = 0.0390). bFGF, EGF, and the mixture of bFGF with EGF can enhanced fibroblasts proliferation, and the concentration of 10 ng/ml of the mixture of bFGF with EGF displayed most effectively (all P < 0.05). The expression levels of Col-I and Col-III mRNAs in fibroblasts displayed significant increases in the 10 ng/ml bFGF combined with EGF group (all P < 0.05). CONCLUSIONS: The optimal concentration of both bFGF and EGF to promote cell proliferation and collagen expression in fibroblasts was 10 ng/ml at which fibroblasts grew faster and secreted more Type I and III collagens into the extracellular matrix, which might contribute to the stability of the pelvic floor microenvironment.


Subject(s)
Cell Proliferation , Collagen/metabolism , Epidermal Growth Factor/physiology , Fibroblast Growth Factor 2/physiology , Fibroblasts/physiology , Animals , Cells, Cultured , Pelvic Floor , Rats , Regeneration
19.
Gen Comp Endocrinol ; 263: 72-79, 2018 07 01.
Article in English | MEDLINE | ID: mdl-29626449

ABSTRACT

Growth factors: vascular endothelial growth factor A (VEGF-A) and fibroblast growth factor (FGF-2) were reported to affect normal physiological reproductive processes in human, domestic and free living animals. Moreover, some reports suggest that VEGF-A and FGF-2 may be directly involved in the control of the annual reproductive cycle of seasonally breeding animals but detailed knowledge is still missing. Our study aimed to demonstrate the expression of mRNA and protein for both factors in the tissues of testis and epididymis (caput, corpus, cauda) at different periods of the year (March, June, November, December) in European bison as a model of seasonally breeding animal. Results suggest, that VEGF-A expression was more pronounced in testis than in epididymis and the highest expression was noted in December and June. Surprisingly, the highest protein accumulation was observed in June at the same level in all tissues analyzed. On the other hand, the highest FGF-2 mRNA expression was noted in testis in June and in epididymis in March. However, no differences in protein expression of FGF-2 were found between analyzed groups. The results indicate that both factors are necessary for proper functioning of the reproductive system and their levels differ seasonally. Perhaps, it is linked to increased need of these factors in the testis as well as epididymis during preparation for the reproductive functions. Moreover, VEGF-A and FGF-2 not only may regulate reproductive functions by affecting vascularization and cell nutrition, but it also may be possible that they possess protective functions by stabilizing the reproductive cells. Therefore, obtained results provide new insight into mechanisms underlying seasonal breeding of the male European bison.


Subject(s)
Bison , Fibroblast Growth Factor 2/physiology , Reproduction , Seasons , Vascular Endothelial Growth Factor A/physiology , Animals , Bison/genetics , Bison/metabolism , Epididymis/metabolism , Fibroblast Growth Factor 2/genetics , Fibroblast Growth Factor 2/metabolism , Gene Expression Regulation , Male , Testis/metabolism , Vascular Endothelial Growth Factor A/genetics , Vascular Endothelial Growth Factor A/metabolism
20.
Sheng Li Xue Bao ; 69(6): 843-851, 2017 Dec 25.
Article in Chinese | MEDLINE | ID: mdl-29270600

ABSTRACT

Neutrophils are one of the most abundant leukocytes present in the human blood circulation system, which could provide continuous immune surveillance. Recent studies have shown that neutrophils are closely related to angiogenesis. Neutrophils could release various cytokines, which regulate the angiogenic process by affecting the growth and migration of endothelial cells directly or indirectly. In the present review, the regulatory effects of neutrophils on angiogenic process and mechanisms are analyzed and summarized, which would provide clues for the treatment of related diseases using neutrophils as the targets in the future.


Subject(s)
Neovascularization, Physiologic/physiology , Neutrophils/physiology , Animals , Fibroblast Growth Factor 2/physiology , Humans , Matrix Metalloproteinase 9/physiology , Oncostatin M/physiology , Vascular Endothelial Growth Factor A/physiology
SELECTION OF CITATIONS
SEARCH DETAIL