Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 43
Filter
Add more filters

Publication year range
1.
J Mol Cell Cardiol ; 130: 23-35, 2019 05.
Article in English | MEDLINE | ID: mdl-30858037

ABSTRACT

OBJECTIVE: To analyze the effects of miR-455-3p-1 and its possible mechanisms in pulmonary arterial hypertension (PAH). METHODS: A microarray assay was used to examine the expressed genes between normal and PAH. The expressed genes in PAH was assessed by qRT-PCR. The targeted interaction between miRNAs and FGF7 was confirmed using a dual luciferase reporter assay. A CCK-8 assay and cell count were used to analyze the pulmonary artery smooth muscle cells (PASMCs) activity and proliferation level, respectively. Apoptotic PASMCs were detected by flow cytometry. In addition, the mRNA and protein expression levels of RAS/ERK signaling pathway were determined by qRT-PCR and a Western blot assay, respectively. A PAH rat model was used to identify the effects of miR-455-3p-1 in vivo. RESULTS: FGF7 was upregulated in PAH. MiR-455-3p-1 was downregulated in PAH. MiR-455-3p-1 targeted FGF7. MiR-455-3p-1 decreased the expression of FGF7. Moreover, the effect of FGF7 on PASMCs was suppressed by miR-455-3p-1. MiR-455-3p-1 upregulation was associated with reduced mRNA and protein levels of core RAS/ERK signal genes, suggesting the inhibition of the RAS/ERK pathway. Furthermore, miR-455-3p-1 upregulation improved the RVSP, mPAP, ratio of RV/LV + S, CO and RV function of PAH rat model in vivo. CONCLUSION: Our findings illustrate a role for miR-455-3p-1 in modulating FGF7-RAS/ERK signaling and suggest that an agomir of miR-455-3p-1 could inhibit the proliferation of PASMCs and mitigate PAH in vivo.


Subject(s)
Fibroblast Growth Factor 7/biosynthesis , Gene Expression Regulation , MAP Kinase Signaling System , MicroRNAs/metabolism , Oncogene Protein p21(ras)/metabolism , Pulmonary Arterial Hypertension/metabolism , Animals , Cell Line , Disease Models, Animal , Humans , Male , Pulmonary Arterial Hypertension/pathology , Rats
2.
J Cell Biochem ; 120(8): 13792-13801, 2019 08.
Article in English | MEDLINE | ID: mdl-31017714

ABSTRACT

Choroidal neovascularization (CNV), a characteristic of age-related macular degeneration, is an underlying cause of severe vision loss among elderly patients. Fibroblast growth factor (FGF) is suggested to exert an important role in the pathogenesis of CNV. However, the molecular mechanisms governing this event are not fully elucidated. Herein, we identified the potential role of FGF7 in CNV. To examine the roles of FGF7 in the progression of CNV, rat CNV models were established and treated with small interfering RNA (siRNA) against FGF7 or FGF7 overexpression, followed by identification of expression of FGF7 in the CNV modeled rats. Next, proliferation and migration, and in vitro tube formation of human umbilical vein endothelial cells, as well as expression of vascular endothelial growth factor (VEGF) and transforming growth factor-beta 2 (TGF-ß2) were evaluated. CNV led to upregulated FGF7 expression. Cells in the presence of FGF7 siRNA showed suppressed proliferation, migration, and tube formation, along with downregulated VEGF and TGF-ß2 expression. Taken together, functional suppression of FGF7 inhibited the onset of CNV, ultimately highlighting a novel therapeutic target for suppressing CNV progression.


Subject(s)
Choroidal Neovascularization , Fibroblast Growth Factor 7 , Gene Silencing , Lasers, Excimer/adverse effects , RNA, Small Interfering , Animals , Choroidal Neovascularization/etiology , Choroidal Neovascularization/genetics , Choroidal Neovascularization/metabolism , Choroidal Neovascularization/pathology , Disease Models, Animal , Fibroblast Growth Factor 7/biosynthesis , Fibroblast Growth Factor 7/genetics , Male , RNA, Small Interfering/genetics , RNA, Small Interfering/pharmacology , Rats
3.
Proc Natl Acad Sci U S A ; 113(13): 3621-6, 2016 Mar 29.
Article in English | MEDLINE | ID: mdl-26976597

ABSTRACT

Influenza can cause acute lung injury. Because immune responses often play a role, antivirals may not ensure a successful outcome. To identify pathogenic mechanisms and potential adjunctive therapeutic options, we compared the extent to which avian influenza A/H5N1 virus and seasonal influenza A/H1N1 virus impair alveolar fluid clearance and protein permeability in an in vitro model of acute lung injury, defined the role of virus-induced soluble mediators in these injury effects, and demonstrated that the effects are prevented or reduced by bone marrow-derived multipotent mesenchymal stromal cells. We verified the in vivo relevance of these findings in mice experimentally infected with influenza A/H5N1. We found that, in vitro, the alveolar epithelium's protein permeability and fluid clearance were dysregulated by soluble immune mediators released upon infection with avian (A/Hong Kong/483/97, H5N1) but not seasonal (A/Hong Kong/54/98, H1N1) influenza virus. The reduced alveolar fluid transport associated with down-regulation of sodium and chloride transporters was prevented or reduced by coculture with mesenchymal stromal cells. In vivo, treatment of aged H5N1-infected mice with mesenchymal stromal cells increased their likelihood of survival. We conclude that mesenchymal stromal cells significantly reduce the impairment of alveolar fluid clearance induced by A/H5N1 infection in vitro and prevent or reduce A/H5N1-associated acute lung injury in vivo. This potential adjunctive therapy for severe influenza-induced lung disease warrants rapid clinical investigation.


Subject(s)
Acute Lung Injury/prevention & control , Influenza A Virus, H5N1 Subtype/pathogenicity , Influenza, Human/complications , Mesenchymal Stem Cells/physiology , Orthomyxoviridae Infections/complications , Acute Lung Injury/etiology , Acute Lung Injury/physiopathology , Angiotensin I/biosynthesis , Animals , Body Fluids/physiology , Coculture Techniques , Cystic Fibrosis Transmembrane Conductance Regulator/metabolism , Cytokines/biosynthesis , Female , Fibroblast Growth Factor 7/biosynthesis , Humans , Inflammation Mediators/metabolism , Mesenchymal Stem Cell Transplantation , Mice , Mice, Inbred BALB C , Orthomyxoviridae Infections/therapy , Permeability , Pulmonary Alveoli/physiopathology , Sodium-Potassium-Exchanging ATPase/metabolism
4.
Am J Respir Cell Mol Biol ; 57(2): 162-173, 2017 08.
Article in English | MEDLINE | ID: mdl-28296468

ABSTRACT

Repair of the lung epithelium after injury is a critical component for resolution; however, the processes necessary to drive epithelial resolution are not clearly defined. Published data demonstrate that Foxp3+ regulatory T cells (Tregs) enhance alveolar epithelial proliferation after injury, and Tregs in vitro directly promote type II alveolar epithelial cell (AT2) proliferation, in part by a contact-independent mechanism. Therefore, we sought to determine the contribution of Treg-specific expression of a growth factor that is known to be important in lung repair, keratinocyte growth factor (kgf). The data demonstrate that Tregs express kgf and that Treg-specific expression of kgf regulates alveolar epithelial proliferation during the resolution phase of acute lung injury and in a model of regenerative alveologenesis in vivo. In vitro experiments demonstrate that AT2 cells cocultured with Tregs lacking kgf have decreased rates of proliferation compared with AT2 cells cocultured with wild-type Tregs. Moreover, Tregs isolated from lung tissue and grown in culture express higher levels of two growth factors that are important for lung repair (kgf and amphiregulin) compared with Tregs isolated from splenic tissue. Lastly, Tregs isolated from human lung tissue can be stimulated ex vivo to induce kgf expression. This study reveals mechanisms by which Tregs direct tissue-reparative effects during resolution after acute lung injury, further supporting the emerging role of Tregs in tissue repair.


Subject(s)
Alveolar Epithelial Cells/cytology , Fibroblast Growth Factor 7/physiology , T-Lymphocytes, Regulatory/metabolism , Acute Lung Injury/chemically induced , Acute Lung Injury/immunology , Acute Lung Injury/metabolism , Acute Lung Injury/pathology , Adoptive Transfer , Alveolar Epithelial Cells/pathology , Amphiregulin/biosynthesis , Amphiregulin/genetics , Animals , Cell Division , Coculture Techniques , Diphtheria Toxin/toxicity , Fibroblast Growth Factor 7/biosynthesis , Fibroblast Growth Factor 7/genetics , Forkhead Transcription Factors/analysis , Gene Expression Regulation/immunology , Humans , Lipopolysaccharides/toxicity , Lung/cytology , Lymphocyte Depletion , Male , Mice , Mice, 129 Strain , Mice, Inbred C57BL , Mice, Transgenic , Pneumonectomy , Postoperative Complications/immunology , Postoperative Complications/metabolism , Postoperative Complications/pathology , T-Lymphocytes, Regulatory/classification , T-Lymphocytes, Regulatory/immunology , T-Lymphocytes, Regulatory/transplantation
5.
Microbiol Immunol ; 61(7): 264-271, 2017 Jul.
Article in English | MEDLINE | ID: mdl-28543309

ABSTRACT

Pulmonary emphysema impairs quality of life and increases mortality. It has previously been shown that administration of adenovirus vector expressing murine keratinocyte growth factor (KGF) before elastase instillation prevents pulmonary emphysema in mice. We therefore hypothesized that therapeutic administration of KGF would restore damage to lungs caused by elastase instillation and thus improve pulmonary function in an animal model. KGF expressing adenovirus vector, which prevented bleomycin-induced pulmonary fibrosis in a previous study, was constructed. Adenovirus vector (1.0 × 109 plaque-forming units) was administered intratracheally one week after administration of elastase into mouse lungs. One week after administration of KGF-vector, exercise tolerance testing and blood gas analysis were performed, after which the lungs were removed under deep anesthesia. KGF-positive pneumocytes were more numerous, surfactant protein secretion in the airspace greater and mean linear intercept of lungs shorter in animals that had received KGF than in control animals. Unexpectedly, however, arterial blood oxygenation was worse in the KGF group and maximum running speed, an indicator of exercise capacity, had not improved after KGF in mice with elastase-induced emphysema, indicating that KGF-expressing adenovirus vector impaired pulmonary function in these mice. Notably, vector lacking KGF-expression unit did not induce such impairment, implying that the KGF expression unit itself may cause the damage to alveolar cells. Possible involvement of the CAG promoter used for KGF expression in impairing pulmonary function is discussed.


Subject(s)
Adenoviridae/genetics , Emphysema/therapy , Fibroblast Growth Factor 7/biosynthesis , Fibroblast Growth Factor 7/genetics , Adenoviridae/metabolism , Alveolar Epithelial Cells/drug effects , Alveolar Epithelial Cells/pathology , Animals , Bleomycin/pharmacology , DNA, Viral/genetics , Disease Models, Animal , Emphysema/chemically induced , Emphysema/physiopathology , Fibroblast Growth Factor 7/administration & dosage , Genetic Therapy , Genetic Vectors/genetics , Genetic Vectors/metabolism , Male , Mice , Mice, Inbred BALB C , Pancreatic Elastase , Promoter Regions, Genetic , Pulmonary Fibrosis/drug therapy , Pulmonary Fibrosis/virology , Pulmonary Surfactant-Associated Protein D/metabolism
6.
An Acad Bras Cienc ; 89(4): 2707-2717, 2017.
Article in English | MEDLINE | ID: mdl-29267793

ABSTRACT

6-Gingerol is the major active constituent of ginger. In the current study, we aimed to investigate the mechanisms underlying the effects of 6-Gingerol on hair growth. Mice were randomly divided into five groups; after hair depilation (day 0), mice were treated with saline, or different concentrations of 6-Gingerol for 11 days. The histomorphological characteristics of the growing hair follicles were examined after hematoxylin and eosin staining. The results indicated that 6-Gingerol significantly suppressed hair growth compared with that in the control group. And choose the concentration of 6-Gingerol at 1 mg/mL to treated with mice. Moreover, 6-Gingerol (1 mg/mL) significantly reduced hair re-growth ratio, hair follicle number, and hair follicle length, which were associated with increased expression of MMP2 and MMP9. Furthermore, the growth factors, such as EGF, KGF, VEGF, IGF-1 and TGF-ß participate in the hair follicle cycle regulation and regulate hair growth. We then measured the concentrations of them using ELISA assays, and the results showed that 6-Gingerol decreased EGF, KGF, VEGF, and IGF-1 concentrations, and increased TGF-ß concentration. Thus, this study showed that 6-Gingerol might act as a hair growth suppressive drug via induction of MMP2 and MMP9 expression, which could interfere with the hair cycle.


Subject(s)
Catechols/pharmacology , Fatty Alcohols/pharmacology , Hair Follicle/drug effects , Matrix Metalloproteinase 2/biosynthesis , Matrix Metalloproteinase 9/biosynthesis , Plant Extracts/pharmacology , Animals , Enzyme Induction , Female , Fibroblast Growth Factor 7/biosynthesis , Hair Follicle/pathology , Insulin-Like Growth Factor I/biosynthesis , Male , Mice , Mice, Inbred C57BL , Random Allocation , Transforming Growth Factor beta/biosynthesis , Vascular Endothelial Growth Factor A/biosynthesis
7.
Am J Physiol Lung Cell Mol Physiol ; 308(2): L118-29, 2015 Jan 15.
Article in English | MEDLINE | ID: mdl-25398985

ABSTRACT

Respiratory distress syndrome (RDS) and bronchopulmonary dysplasia remain the leading causes of preterm infant morbidity, mortality, and lifelong disability. Research to improve outcomes requires translational large animal models for RDS. Preterm pigs delivered by caesarian section at gestation days (GD) 98, 100, 102, and 104 were provided 24 h of neonatal intensive care, monitoring (pulse oximetry, blood gases, serum biomarkers, radiography), and nutritional support, with or without intubation and mechanical ventilation (MV; pressure control ventilation with volume guarantee). Spontaneous development of RDS and mortality without MV are inversely related with GD at delivery and correspond with inadequacy of tidal volume and gas exchange. GD 98 and 100 pigs have consolidated lungs, immature alveolar architecture, and minimal surfactant protein-B expression, and MV is essential at GD 98. Although GD 102 pigs had some alveoli lined by pneumocytes and surfactant was released in response to MV, blood gases and radiography revealed limited recruitment 1-2 h after delivery, and mortality at 24 h was 66% (35/53) with supplemental oxygen provided by a mask and 69% (9/13) with bubble continuous positive airway pressure (8-9 cmH2O). The lungs at GD 104 had higher densities of thin-walled alveoli that secreted surfactant, and MV was not essential. Between GD 98 and 102, preterm pigs have ventilation inadequacies and risks of RDS that mimic those of preterm infants born during the saccular phase of lung development, are compatible with standards of neonatal intensive care, and are alternative to fetal nonhuman primates and lambs.


Subject(s)
Bronchopulmonary Dysplasia/pathology , Disease Models, Animal , Lung/embryology , Pulmonary Alveoli/embryology , Respiratory Distress Syndrome, Newborn/pathology , Swine , Animals , Biomarkers , Female , Fibroblast Growth Factor 7/biosynthesis , Humans , Infant, Newborn , Infant, Premature , Intensive Care, Neonatal , Male , Pregnancy , Pulmonary Alveoli/diagnostic imaging , Pulmonary Surfactant-Associated Protein B/biosynthesis , Radiography , Respiration, Artificial
8.
Eur Arch Otorhinolaryngol ; 272(10): 2689-96, 2015 Oct.
Article in English | MEDLINE | ID: mdl-25138153

ABSTRACT

We reported previously that keratinocyte growth factor (KGF), a mesenchymal cell-derived paracrine growth factor, plays an important role in middle ear cholesteatoma formation, which is characterized by marked proliferation of epithelial cells. Here, we investigated whether KGF, the main factor that induces cholesteatoma, overexpression in vivo results in the formation of cholesteatoma. Flag-hKGF cDNA driven by CMV14 promoter was transfected through electroporation into the external auditory canal (EAC) of rats once (short-term model) or five times on every fourth day (long-term model). Ears transfected with empty vector were used as controls. Successful transfection of plasmids into epithelial and stromal cells was confirmed by Flag immunohistochemistry. In the short-term model, the intensity of KGF protein was the strongest in hKGF transfected ear at day 4. KGF expression induced epithelial cell proliferation, reaching a peak level at day 4 and then decreased later, while in the long-term model, KGF expression in the EAC led to middle ear cholesteatoma formation. In conclusion, we described here a new experimental model of human middle ear cholesteatoma, and demonstrated that KGF and KGF receptor paracrine action play an essential role in middle ear cholesteatoma formation in an in vivo model.


Subject(s)
Cholesteatoma, Middle Ear/genetics , DNA/genetics , Fibroblast Growth Factor 7/genetics , Gene Expression Regulation , Animals , Cholesteatoma, Middle Ear/metabolism , Disease Models, Animal , Fibroblast Growth Factor 7/biosynthesis , Humans , Immunohistochemistry , Male , Rats , Rats, Sprague-Dawley
9.
Appl Microbiol Biotechnol ; 98(9): 3933-45, 2014 May.
Article in English | MEDLINE | ID: mdl-24463717

ABSTRACT

Three forms of recombinant human keratinocyte growth factor 1 (rhKGF1) with or without the native signal peptide or a 23-amino acid truncation were expressed in Spodoptera frugiperda 9 (Sf9) cells by designing with insect codon usage. Immunoblotting demonstrated that these rhKGF1 proteins were recognized by a human anti-KGF1 antibody. The multiplicity of infection and timing of harvest had a significant effect on protein yield, protein quality, and cytotoxicity. Our results indicated that the native signal peptide directed KGF1 secretion from insect cells, reaching a maximum at 60 h postinfection. Although secretion of rhKGF1194 was less efficient than that of rhKGF1163 and rhKGF1140, protein secretion is an attractive pathway for simple purification of biologically active rhKGF1 at a high yield. Moreover, the sizes of rhKGF1194 and rhKGF1163 were similar (20 kDa), suggesting that the signal peptide may be recognized and removed in Sf9 cells. A 3-[4,5-dimethylthiazol-2-yl]-2,5 diphenyl tetrazolium bromide assay was used to analyze the biological function of rhKGF1, indicating that the three forms of rhKGF1 had a similar mitogenic function in BaF3 cells. Furthermore, to elucidate the effect of rhKGF1 on cytoprotection of liver cells, we used KGF1 pretreatment of an acute liver injury model. The results indicated that rhKGF1 prevented necrosis and apoptosis of CCl4-treated HL7702 cells in vitro and in vivo. These results suggest that KGF1 may be a candidate therapeutic drug for acute liver injury.


Subject(s)
Fibroblast Growth Factor 7/biosynthesis , Gene Expression , Hepatocytes/drug effects , Hepatocytes/physiology , Animals , Cell Line , Cytoprotection , Fibroblast Growth Factor 7/chemistry , Fibroblast Growth Factor 7/genetics , Fibroblast Growth Factor 7/metabolism , Humans , Molecular Sequence Data , Molecular Weight , Protein Sorting Signals , Recombinant Proteins/biosynthesis , Recombinant Proteins/chemistry , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Sequence Analysis, DNA , Spodoptera
10.
Cell Tissue Res ; 353(1): 117-22, 2013 Jul.
Article in English | MEDLINE | ID: mdl-23579552

ABSTRACT

Efficient cell expansion is a basic requirement for obtaining clinically relevant numbers of mesenchymal stem cells designed for cell-based therapies or tissue-engineering application. Previous studies have demonstrated that mesenchymal stem cells (MSC) cultivated under reduced atmospheric oxygen concentrations (2.5% O2) possess enhanced proliferation potential and can maintain their differentiation properties. We have analyzed the oxygen-dependent cytokine expression of human MSC derived from umbilical cord and attempted to link the results to the proliferation and differentiation capacities of these cells. By quantitative reverse transcription plus the polymerase chain reaction and by protein microarray, we measured the gene expression and intracellular protein concentration of several growth factors and growth factor receptors. Fibroblast growth factor-7, two growth factor receptors (vascular endothelial growth factor receptor 2 and stem cell factor receptor), and two growth-factor-binding proteins (insulin-like growth-factor-binding proteins 3 and 6) were over-expressed under hypoxic conditions, indicating that their signaling pathways participate in cell proliferation. On the other hand, typical differentiation factors such as bone morphogenetic protein-4, endothelial growth factor, and tissue growth factor-ß1 were absent in cells cultivated under hypoxic and normoxic conditions. The absolute concentration of some intracellular cytokines was also measured for the first time under hypoxia and normoxia. Our results in combination with previous findings indicate that enhanced proliferation potential and a maintained undifferentiated cell state can be ascribed to the oxygen-dependent expression of a set of cytokines. This knowledge might help in the understanding of MSC physiology and in the achievement of directed cell fate of MSC for clinical application.


Subject(s)
Cell Hypoxia/physiology , Mesenchymal Stem Cells/metabolism , Oxygen/metabolism , Umbilical Cord/metabolism , Bone Morphogenetic Protein 4/deficiency , Cell Differentiation , Cell Proliferation , Cells, Cultured , Endothelial Growth Factors/deficiency , Fibroblast Growth Factor 7/biosynthesis , Fibroblast Growth Factor 7/metabolism , Gene Expression , Humans , Insulin-Like Growth Factor Binding Protein 3/biosynthesis , Insulin-Like Growth Factor Binding Protein 3/metabolism , Insulin-Like Growth Factor Binding Protein 6/biosynthesis , Insulin-Like Growth Factor Binding Protein 6/metabolism , Proto-Oncogene Proteins c-kit/biosynthesis , Proto-Oncogene Proteins c-kit/metabolism , Transforming Growth Factor beta1/biosynthesis , Umbilical Cord/cytology , Vascular Endothelial Growth Factor Receptor-2/biosynthesis , Vascular Endothelial Growth Factor Receptor-2/metabolism
11.
Thorax ; 67(6): 496-501, 2012 Jun.
Article in English | MEDLINE | ID: mdl-22106021

ABSTRACT

BACKGROUND: Bone-marrow derived mesenchymal stem cells (MSCs) reduce the severity of evolving acute lung injury (ALI), but their ability to repair the injured lung is not clear. A study was undertaken to determine the potential for MSCs to enhance repair after ventilator-induced lung injury (VILI) and elucidate the mechanisms underlying these effects. METHODS: Anaesthetised rats underwent injurious ventilation which produced severe ALI. Following recovery, they were given an intravenous injection of MSCs (2×10(6) cells) or vehicle immediately and a second dose 24 h later. The extent of recovery following VILI was assessed after 48 h. Subsequent experiments examined the potential for non-stem cells and for the MSC secretome to enhance VILI repair. The contribution of specific MSC-secreted mediators was then examined in a wound healing model. RESULTS: MSC therapy enhanced repair following VILI. MSCs enhanced restoration of systemic oxygenation and lung compliance, reduced total lung water, decreased lung inflammation and histological lung injury and restored lung structure. They attenuated alveolar tumour necrosis factor α concentrations while increasing concentrations of interleukin 10. These effects were not seen with non-stem cells (ie, rat fibroblasts). MSC-secreted products also enhanced lung repair and attenuated the inflammatory response following VILI. The beneficial effect of the MSC secretome on repair of pulmonary epithelial wounds was attenuated by prior depletion of keratinocyte growth factor. CONCLUSION: MSC therapy enhances lung repair following VILI via a paracrine mechanism that may be keratinocyte growth factor-dependent.


Subject(s)
Interleukin-10/biosynthesis , Mesenchymal Stem Cell Transplantation , Tumor Necrosis Factor-alpha/drug effects , Ventilator-Induced Lung Injury/surgery , Animals , Disease Models, Animal , Fibroblast Growth Factor 7/biosynthesis , Injections, Intravenous , Male , Mesenchymal Stem Cells , Rats , Rats, Sprague-Dawley , Treatment Outcome , Ventilator-Induced Lung Injury/metabolism , Wound Healing
12.
Proc Natl Acad Sci U S A ; 106(18): 7553-8, 2009 May 05.
Article in English | MEDLINE | ID: mdl-19383786

ABSTRACT

The possibility that Vgamma2Vdelta2 T effector cells can confer protection against pulmonary infectious diseases has not been tested. We have recently demonstrated that single-dose (E)-4-hydroxy-3-methyl-but-2-enyl pyrophosphate (HMBPP) plus IL-2 treatment can induce prolonged accumulation of Vgamma2Vdelta2 T effector cells in lungs. Here, we show that a delayed HMBPP/IL-2 administration after inhalational Yersinia pestis infection induced marked expansion of Vgamma2Vdelta2 T cells but failed to control extracellular plague bacterial replication/infection. Surprisingly, despite the absence of infection control, expansion of Vgamma2Vdelta2 T cells after HMBPP/IL-2 treatment led to the attenuation of inhalation plague lesions in lungs. Consistently, HMBPP-activated Vgamma2Vdelta2 T cells accumulated and localized in pulmonary interstitials surrounding small blood vessels and airway mucosa in the lung tissues with no or mild plague lesions. These infiltrating Vgamma2Vdelta2 T cells produced FGF-7, a homeostatic mediator against tissue damages. In contrast, control macaques treated with glucose plus IL-2 or glucose alone exhibited severe hemorrhages and necrosis in most lung lobes, with no or very few Vgamma2Vdelta2 T cells detectable in lung tissues. The findings are consist with the paradigm that circulating Vgamma2Vdelta2 T cells can traffic to lungs for homeostatic protection against tissue damages in infection.


Subject(s)
Interleukin-2/administration & dosage , Lung/immunology , Organophosphates/administration & dosage , Plague/immunology , Pneumonia, Bacterial/immunology , Receptors, Antigen, T-Cell, gamma-delta/immunology , T-Lymphocytes/drug effects , Yersinia pestis , Animals , Cell Movement , Disease Models, Animal , Fibroblast Growth Factor 7/biosynthesis , Homeostasis , Lung/microbiology , Lung/pathology , Macaca , Plague/pathology , Pneumonia, Bacterial/pathology , Respiratory Mucosa/immunology , Respiratory Mucosa/microbiology , Respiratory Mucosa/pathology , T-Lymphocytes/immunology
13.
J Pharmacol Sci ; 115(2): 230-4, 2011.
Article in English | MEDLINE | ID: mdl-21258175

ABSTRACT

It has been reported that pantothenic acid (vitamin B5) and panthenol, an alcohol derivative of pantothenic acid, have beneficial moisturizing effects on the skin. However, few studies have investigated the mechanism of action of pantothenic acid on skin tissues. We tried to clarify the role of pantothenic acid on skin function by using keratinocytes and fibroblasts. The depletion of pantothenic acid from the culture medium suppressed keratinocyte proliferation and promoted differentiation. Moreover, pantothenic acid depletion decreased the synthesis of keratinocyte growth factor and procollagen 4a2 in fibroblasts. These results suggest that pantothenic acid is essential for maintaining keratinocyte proliferation and differentiation.


Subject(s)
Cell Proliferation , Collagen/biosynthesis , Fibroblast Growth Factor 7/biosynthesis , Fibroblasts/metabolism , Keratinocytes/cytology , Pantothenic Acid/deficiency , Pantothenic Acid/physiology , Animals , Cell Cycle , Cell Differentiation , Cells, Cultured , Fibroblasts/physiology , Humans , Keratinocytes/metabolism , Mice
14.
Ann Clin Transl Neurol ; 8(7): 1508-1514, 2021 07.
Article in English | MEDLINE | ID: mdl-34053190

ABSTRACT

Schwannomas are benign neoplasms that can cause gain- and loss-of-function neurological phenotypes, including severe, intractable pain. To investigate the molecular mechanisms underlying schwannoma-associated pain we compared the RNA sequencing profile of painful and non-painful schwannomas from NF2 patients. Distinct segregation of painful and non-painful tumors by gene expression patterns was observed. Differential expression analysis showed the upregulation of fibroblast growth factor 7 (FGF7) in painful schwannomas. Behavioral support for this finding was observed using a xenograft human NF2-schwannoma model in nude mice. In this model, over-expression of FGF7 in intra-sciatically implanted NF2 tumor cells generated pain behavior compared with controls.


Subject(s)
Fibroblast Growth Factor 7/genetics , Neurilemmoma/genetics , Neurofibromatosis 2/genetics , Pain/genetics , Sequence Analysis, RNA/methods , Transcriptome/genetics , Animals , Cell Line, Tumor , Female , Fibroblast Growth Factor 7/biosynthesis , Humans , Male , Mice , Mice, Nude , Neurilemmoma/metabolism , Neurilemmoma/pathology , Neurofibromatosis 2/metabolism , Neurofibromatosis 2/pathology , Pain/metabolism , Pain/pathology , Sciatic Neuropathy/genetics , Sciatic Neuropathy/metabolism , Sciatic Neuropathy/pathology , Xenograft Model Antitumor Assays/methods
15.
Biochem Biophys Res Commun ; 386(1): 21-5, 2009 Aug 14.
Article in English | MEDLINE | ID: mdl-19497301

ABSTRACT

The development and characterization of an enhanced composite skin substitute based on collagen and poly(epsilon-caprolactone) are reported. Considering the features of excellent biocompatibility, easy-manipulated property and exempt from cross-linking related toxicity observed in the 1:20 biocomposites, skin substitutes were developed by seeding human single-donor keratinocytes and fibroblasts alone on both sides of the 1:20 biocomposite to allow for separation of two cell types and preserving cell signals transmission via micro-pores with a porosity of 28.8 +/- 16.1 microm. The bi-layered skin substitute exhibited both differentiated epidermis and fibrous dermis in vitro. Less Keratinocyte Growth Factor production was measured in the co-cultured skin model compared to fibroblast alone condition indicating a favorable microenvironment for epidermal homeostasis. Moreover, fast wound closure, epidermal differentiation, and abundant dermal collagen deposition were observed in composite skin in vivo. In summary, the beneficial characteristics of the new skin substitutes exploited the potential for pharmaceutical screening and clinical application.


Subject(s)
Collagen/chemistry , Polyesters/chemistry , Skin, Artificial , Animals , Coculture Techniques , Fibroblast Growth Factor 7/biosynthesis , Fibroblasts/metabolism , Fibroblasts/physiology , Humans , Keratinocytes/metabolism , Keratinocytes/physiology , Materials Testing , Mice , Mice, Nude , Porosity , Tissue Donors
16.
Lasers Med Sci ; 24(6): 885-91, 2009 Nov.
Article in English | MEDLINE | ID: mdl-18600291

ABSTRACT

The effects of laser phototherapy on the release of growth factors by human gingival fibroblasts were studied in vitro. Cells from a primary culture were irradiated twice (6 h interval), with continuous diode laser [gallium-aluminum-arsenium (GaAlAs), 780 nm, or indium-gallium-aluminum-phosphide (InGaAlP),_660 nm] in punctual and contact mode, 40 mW, spot size 0.042 cm(2), 3 J/cm(2) and 5 J/cm(2) (3 s and 5 s, respectively). Positive [10% fetal bovine serum (FBS)] and negative (1%FBS) controls were not irradiated. Production of keratinocyte growth factor (KGF) and basic fibroblast growth factor (bFGF) was quantified by enzyme-linked immunosorbent assay (ELISA). The data were statistically compared by analysis of variance (ANOVA) followed by Tukey's test (P

Subject(s)
Fibroblast Growth Factors/biosynthesis , Gingiva/metabolism , Gingiva/radiation effects , Low-Level Light Therapy , Phototherapy , Animals , Cattle , Cells, Cultured , Culture Media , Fibroblast Growth Factor 2/biosynthesis , Fibroblast Growth Factor 7/biosynthesis , Fibroblasts/metabolism , Fibroblasts/radiation effects , Gingiva/cytology , Gingivoplasty , Humans , Wound Healing/physiology , Wound Healing/radiation effects
17.
PLoS Med ; 5(1): e19, 2008 Jan 29.
Article in English | MEDLINE | ID: mdl-18232728

ABSTRACT

BACKGROUND: Important support functions, including promotion of tumor growth, angiogenesis, and invasion, have been attributed to the different cell types populating the tumor stroma, i.e., endothelial cells, cancer-associated fibroblasts, pericytes, and infiltrating inflammatory cells. Fibroblasts have long been recognized inside carcinomas and are increasingly implicated as functional participants. The stroma is prominent in cervical carcinoma, and distinguishable from nonmalignant tissue, suggestive of altered (tumor-promoting) functions. We postulated that pharmacological targeting of putative stromal support functions, in particular those of cancer-associated fibroblasts, could have therapeutic utility, and sought to assess the possibility in a pre-clinical setting. METHODS AND FINDINGS: We used a genetically engineered mouse model of cervical carcinogenesis to investigate platelet-derived growth factor (PDGF) receptor signaling in cancer-associated fibroblasts and pericytes. Pharmacological blockade of PDGF receptor signaling with the clinically approved kinase inhibitor imatinib slowed progression of premalignant cervical lesions in this model, and impaired the growth of preexisting invasive carcinomas. Inhibition of stromal PDGF receptors reduced proliferation and angiogenesis in cervical lesions through a mechanism involving suppression of expression of the angiogenic factor fibroblast growth factor 2 (FGF-2) and the epithelial cell growth factor FGF-7 by cancer-associated fibroblasts. Treatment with neutralizing antibodies to the PDGF receptors recapitulated these effects. A ligand trap for the FGFs impaired the angiogenic phenotype similarly to imatinib. Thus PDGF ligands expressed by cancerous epithelia evidently stimulate PDGFR-expressing stroma to up-regulate FGFs, promoting angiogenesis and epithelial proliferation, elements of a multicellular signaling network that elicits functional capabilities in the tumor microenvironment. CONCLUSIONS: This study illustrates the therapeutic benefits in a mouse model of human cervical cancer of mechanism-based targeting of the stroma, in particular cancer-associated fibroblasts. Drugs aimed at stromal fibroblast signals and effector functions may prove complementary to conventional treatments targeting the overt cancer cells for a range of solid tumors, possibly including cervical carcinoma, the second most common lethal malignancy in women worldwide, for which management remains poor.


Subject(s)
Antineoplastic Agents/therapeutic use , Carcinoma/blood supply , Neoplasm Proteins/physiology , Neovascularization, Pathologic/drug therapy , Paracrine Communication/physiology , Piperazines/therapeutic use , Platelet-Derived Growth Factor/physiology , Protein Kinase Inhibitors/therapeutic use , Pyrimidines/therapeutic use , Receptors, Platelet-Derived Growth Factor/physiology , Uterine Cervical Neoplasms/blood supply , Animals , Antineoplastic Agents/pharmacology , Benzamides , Carcinoma/drug therapy , Carcinoma/physiopathology , Carcinoma/virology , Epithelial Cells/metabolism , Estradiol/administration & dosage , Estradiol/toxicity , Female , Fibroblast Growth Factor 2/biosynthesis , Fibroblast Growth Factor 2/genetics , Fibroblast Growth Factor 2/physiology , Fibroblast Growth Factor 7/biosynthesis , Fibroblast Growth Factor 7/genetics , Fibroblast Growth Factor 7/physiology , Fibroblasts/drug effects , Fibroblasts/enzymology , Fibroblasts/metabolism , Human papillomavirus 16 , Humans , Imatinib Mesylate , Mice , Mice, Inbred Strains , Mice, Transgenic , Neoplasm Proteins/antagonists & inhibitors , Neoplasm Proteins/biosynthesis , Neoplasm Proteins/genetics , Neovascularization, Pathologic/physiopathology , Paracrine Communication/drug effects , Pericytes/metabolism , Piperazines/pharmacology , Platelet-Derived Growth Factor/antagonists & inhibitors , Platelet-Derived Growth Factor/genetics , Protein Kinase Inhibitors/pharmacology , Proto-Oncogene Proteins c-kit/biosynthesis , Proto-Oncogene Proteins c-kit/genetics , Pyrimidines/pharmacology , Receptors, Platelet-Derived Growth Factor/antagonists & inhibitors , Receptors, Platelet-Derived Growth Factor/genetics , Stromal Cells/drug effects , Stromal Cells/enzymology , Stromal Cells/metabolism , Uterine Cervical Neoplasms/drug therapy , Uterine Cervical Neoplasms/physiopathology , Uterine Cervical Neoplasms/virology
18.
Cancer Res ; 66(14): 7326-33, 2006 Jul 15.
Article in English | MEDLINE | ID: mdl-16849583

ABSTRACT

Prostate gland development and growth requires both androgen action and epithelial-stromal communications. In fact, androgen signaling through the androgen receptor (AR) may be important in both stromal and epithelial cells of the prostate. Because interaction of AR with the coactivator, Hic-5/ARA55, results in enhanced androgen-induced transcription, we analyzed Hic-5/ARA55 expression in prostate tissue sections from normal human donors and prostate cancer patients. In each sample, Hic-5/ARA55 expression was confined to the stromal compartment of the prostate. Furthermore, a prostate stromal cell line, WPMY-1 cells, expresses Hic-5/ARA55, which is localized both at focal adhesion complexes and within the soluble cytoplasmic compartment. The ability of Hic-5/ARA55 to shuttle between the nuclear and cytoplasmic compartments was revealed on inhibition of nuclear export with leptomycin B. Small interfering RNA ablation experiments established endogenous Hic-5/ARA55 as a coactivator for both viral and endogenous cellular AR-regulated genes. Finally, the mechanism of Hic-5/ARA55 coactivator activity in WPMY-1 cells was revealed by chromatin immunoprecipitation analysis that showed its androgen-dependent recruitment to the promoter of the stromal androgen-responsive keratinocyte growth factor gene. These data provide the first demonstration of a stromal-specific AR coactivator that has an effect on an androgen-regulated growth factor that is essential for stromal/epithelial cell communication in the prostate.


Subject(s)
Cytoskeletal Proteins/biosynthesis , DNA-Binding Proteins/biosynthesis , Prostate/metabolism , Prostatic Neoplasms/metabolism , Amino Acid Sequence , Animals , Cell Line , Cell Nucleus/metabolism , Cytoplasm/metabolism , Cytoskeletal Proteins/genetics , Cytoskeletal Proteins/metabolism , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , Fibroblast Growth Factor 7/biosynthesis , Fibroblast Growth Factor 7/genetics , Humans , Intracellular Signaling Peptides and Proteins , LIM Domain Proteins , Male , Mice , Molecular Sequence Data , Promoter Regions, Genetic , Prostate/cytology , Prostate/physiology , Prostatic Neoplasms/genetics , Prostatic Neoplasms/pathology , Receptors, Androgen/genetics , Receptors, Androgen/metabolism , Sequence Alignment , Stromal Cells/cytology , Stromal Cells/metabolism , Stromal Cells/physiology , Transcriptional Activation
19.
Enzyme Microb Technol ; 111: 12-20, 2018 Apr.
Article in English | MEDLINE | ID: mdl-29421032

ABSTRACT

Keratinocyte Growth Factor (KGF) is a paracrine-acting, epithelial mitogen that plays a prominent role in the regeneration of damaged epithelial tissues. In spite of different attempts to produce recombinant human KGF in many organisms, including bacteria, mammalian cells, plant cells and insect cells; production of recombinant form suffers from lower yields and recovery relative to other recombinant proteins of similar size and properties. Due to many advantages of Pichia pastoris expression systems for producing industrial enzymes and pharmaceutical proteins, in this study P. pastoris was chosen as a host for KGF expression. For preparing human KGF coding sequence, MCF-7 cell line was treated with 1,25-Dihydroxy vitamin D3 for inducing the expression of KGF. The coding sequence of 23N-terminal truncated KGF form was amplified using RT-PCR technique and then cloned into the yeast expression vector in frame with the yeast α-factor secretion signal. The recombinant plasmid was integrated into Pichia pastoris strain X-33 genome. Western blotting and Mass Spectrometry demonstrated that recombinant human KGF (rhKGF) was correctly expressed after methanol induction and secreted into the media. The recombinant protein was purified from the media by heparin affinity chromatography. MTT assay showed that the purified rhKGF had a proliferative effect on NIH3T3 and A549 cell lines. In addition, protective effect of recombinant KGF was assessed in A549 cell line after irradiation. The results showed that the recombinant protein was biologically active. Finally, the effect of recombinant KGF was investigated on proliferation of MCF-7 cell line and its response to radiation. The results showed that pre-treatment of KGF have a protective effect on MCF-7 cell line after irradiation.


Subject(s)
Fibroblast Growth Factor 7/biosynthesis , Fibroblast Growth Factor 7/genetics , Pichia/genetics , Pichia/metabolism , Radiation-Protective Agents/pharmacology , A549 Cells , Animals , Cell Proliferation/drug effects , Cloning, Molecular , Fibroblast Growth Factor 7/pharmacology , Humans , MCF-7 Cells , Mice , NIH 3T3 Cells , Radiation Tolerance/drug effects , Recombinant Proteins/biosynthesis , Recombinant Proteins/genetics , Recombinant Proteins/pharmacology
20.
Physiol Rep ; 6(16): e13794, 2018 08.
Article in English | MEDLINE | ID: mdl-30155985

ABSTRACT

TGF beta is a multifunctional cytokine that is important in the pathogenesis of pulmonary fibrosis. The ability of TGF beta to stimulate smooth muscle actin and extracellular matrix gene expression in fibroblasts is well established. In this report, we evaluated the effect of TGF beta on the expression of HGF, FGF7 (KGF), and FGF10, important growth and survival factors for the alveolar epithelium. These growth factors are important for maintaining type II cells and for restoration of the epithelium after lung injury. Under conditions of normal serum supplementation or serum withdrawal TGF beta inhibited fibroblast expression of HGF, FGF7, and FGF10. We confirmed these observations with genome wide RNA sequencing of the response of control and IPF fibroblasts to TGF beta. In general, gene expression in IPF fibroblasts was similar to control fibroblasts. Reduced expression of HGF, FGF7, and FGF10 is another means whereby TGF beta impairs epithelial healing and promotes fibrosis after lung injury.


Subject(s)
Fibroblasts/drug effects , Idiopathic Pulmonary Fibrosis/pathology , Intercellular Signaling Peptides and Proteins/biosynthesis , Transforming Growth Factor beta/pharmacology , Aged , Aged, 80 and over , Cells, Cultured , Culture Media, Serum-Free , Female , Fibroblast Growth Factor 10/biosynthesis , Fibroblast Growth Factor 10/genetics , Fibroblast Growth Factor 7/biosynthesis , Fibroblast Growth Factor 7/genetics , Fibroblasts/metabolism , Gene Expression Regulation/drug effects , Hepatocyte Growth Factor/biosynthesis , Hepatocyte Growth Factor/genetics , Humans , Idiopathic Pulmonary Fibrosis/metabolism , Intercellular Signaling Peptides and Proteins/genetics , Lung/metabolism , Male , Middle Aged , RNA, Messenger/genetics , Transforming Growth Factor beta/physiology
SELECTION OF CITATIONS
SEARCH DETAIL