Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 113
Filter
Add more filters

Country/Region as subject
Publication year range
1.
Biochem Biophys Res Commun ; 692: 149321, 2024 Jan 15.
Article in English | MEDLINE | ID: mdl-38056156

ABSTRACT

Spinal cord injury (SCI) can cause severe and permanent neurological damage, and neuronal apoptosis could inhibit functional recovery of damaged spinal cord greatly. Human umbilical cord mesenchymal stem cells (hUC-MSCs) have great potential to repair SCI because of a series of advantages, including inhibition of neuronal apoptosis and multiple differentiation. The former may play an important role. However, the detailed regulatory mechanism associated with the inhibition of neuronal apoptosis after hUC-MSCs administration has not been elucidated. In this study, proteomics analysis of precious human cerebrospinal fluid (CSF) samples collected from SCI subjects receiving hUC-MSCs delivery indicated that hepatocyte growth factor (HGF) is largely involved in SCI repair. Furthermore, overexpression of HGF derived from hUC-MSCs could decrease reactive oxygen species to prevent neuron apoptosis to the maximum, and thus lead to significant recovery of spinal cord dysfunction. Moreover, HGF could promote phosphorylation of Akt/FoxO3a pathway to decrease reactive oxygen species to reduce neuron apoptosis. For the first time, our research revealed that HGF secreted by hUC-MSCs inhibits neuron apoptosis by phosphorylation of Akt/FoxO3a to repair SCI. This study provides important clues associated with drug selection for the effective treatment of SCI in humans.


Subject(s)
Mesenchymal Stem Cell Transplantation , Mesenchymal Stem Cells , Spinal Cord Injuries , Humans , Hepatocyte Growth Factor/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Phosphorylation , Reactive Oxygen Species/metabolism , Umbilical Cord , Apoptosis , Spinal Cord Injuries/metabolism
2.
J Periodontal Res ; 59(3): 565-575, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38240289

ABSTRACT

BACKGROUND AND OBJECTIVE: Clinical studies found high levels of hepatocyte growth factor (HGF) expression in patients with periodontitis. Studies suggest that HGF plays an important role in periodontitis, is involved in inflammation, and modulates alveolar bone integrity in periodontitis. This study aims to investigate the effects and mechanisms of HGF in the progression of experimental periodontitis. METHODS: We used silk thread ligation to induce periodontitis in HGF-overexpressing transgenic (HGF-Tg) and wild-type C57BL/6J mice. The effects of HGF overexpression on alveolar bone destruction were assessed by microcomputed tomography imaging at baseline and on days 7, 14, 21, and 28. We analyzed the cytokines (IL-6 and TNF-α) and lymphocytes in periodontitis tissues by enzyme-linked immunosorbent assay and flow cytometry. The effects of HGF on alveolar bone destruction were further tested by quantifying the systemic bone metabolism markers CTXI and PINP and by RNA sequencing for the signaling pathways involved in bone destruction. Western blotting and immunohistochemistry were performed to further elucidate the involved signaling pathways. RESULTS: We found that experimental periodontitis increased HGF production in periodontitis tissues; however, the effects of HGF overexpression were inconsistent with disease progression. In the early stage of periodontitis, periodontal inflammation and alveolar bone destruction were significantly lower in HGF-Tg mice than in wild-type mice. In the late stage, HGF-Tg mice showed higher inflammatory responses and progressively aggravated bone destruction with continued stimulation of inflammation. We identified the IL-17/RANKL/TRAF6 pathway as a signaling pathway involved in the HGF effects on the progression of periodontitis. CONCLUSION: HGF plays divergent effects in the progression of experimental periodontitis and accelerates osteoclastic activity and bone destruction in the late stage of inflammation.


Subject(s)
Alveolar Bone Loss , Hepatocyte Growth Factor , Mice, Inbred C57BL , Mice, Transgenic , Periodontitis , X-Ray Microtomography , Animals , Hepatocyte Growth Factor/metabolism , Periodontitis/metabolism , Periodontitis/pathology , Mice , Alveolar Bone Loss/metabolism , Alveolar Bone Loss/diagnostic imaging , Alveolar Bone Loss/pathology , Disease Models, Animal , Disease Progression , Tumor Necrosis Factor-alpha/metabolism , Interleukin-6/metabolism , Signal Transduction , Male , Enzyme-Linked Immunosorbent Assay
3.
J Cell Mol Med ; 26(18): 4745-4755, 2022 09.
Article in English | MEDLINE | ID: mdl-35922965

ABSTRACT

Mesenchymal stem cell (MSC) therapy is considered a new treatment for a wide range of diseases and injuries, but challenges remain, such as poor survival, homing and engraftment rates, thus limiting the therapeutic efficacy of the transplanted MSCs. Many strategies have been developed to enhance the therapeutic efficacy of MSCs, such as preconditioning, co-transplantation with graft materials and gene modification. Hepatocyte growth factor (HGF) is secreted by MSCs, which plays an important role in MSC therapy. It has been reported that the modification of the HGF gene is beneficial to the therapeutic efficacy of MSCs, including diseases of the heart, lung, liver, urinary system, bone and skin, lower limb ischaemia and immune-related diseases. This review focused on studies involving HGF/MSCs both in vitro and in vivo. The characteristics of HGF/MSCs were summarized, and the mechanisms of their improved therapeutic efficacy were analysed. Furthermore, some insights are provided for HGF/MSCs' clinical application based on our understanding of the HGF gene and MSC therapy.


Subject(s)
Mesenchymal Stem Cell Transplantation , Mesenchymal Stem Cells , Hepatocyte Growth Factor/genetics , Hepatocyte Growth Factor/metabolism , Hepatocytes/metabolism , Lung/metabolism , Mesenchymal Stem Cells/metabolism
4.
Cancer Sci ; 113(4): 1292-1304, 2022 Apr.
Article in English | MEDLINE | ID: mdl-35092134

ABSTRACT

MET, the receptor for the hepatocyte growth factor (HGF), is strongly associated with resistance to tyrosine kinase inhibitors, key drugs that are used in the therapy of non-small cell lung cancer. MET contains 11 potential N-glycosylation sites, but the site-specific roles of these N-glycans have not been elucidated. We report herein that these N-glycans regulate the proteolytic processing of MET and HGF-induced MET signaling, and that this regulation is site specific. Inhibitors of N-glycosylation were found to suppress the processing and trafficking of endogenous MET in H1975 and EBC-1 lung cancer cells and exogenous MET in CHO-K1 cells. We purified the recombinant extracellular domain of human MET and determined the site-specific N-glycan structures and occupancy using mass spectrometry. The results indicated that most sites were fully glycosylated and that the dominant population was the complex type. To examine the effects of the deletion of N-glycans of MET, we prepared endogenous MET knockout Flp-In CHO cells and transfected them with a series of N-glycan-deletion mutants of MET. The results showed that several N-glycans are implicated in the processing of MET. The findings also suggested that the N-glycans of the SEMA domain of MET positively regulate HGF signaling, and the N-glycans of the region other than the SEMA domain negatively regulate HGF signaling. Processing, cell surface expression, and signaling were significantly suppressed in the case of the all-N-glycan-deletion mutant. The overall findings suggest that N-glycans of MET affect the status and the function of the receptor in a site-specific manner.


Subject(s)
Carcinoma, Non-Small-Cell Lung , Lung Neoplasms , Animals , Cricetinae , Cricetulus , Glycosylation , Hepatocyte Growth Factor/metabolism , Humans , Lung Neoplasms/drug therapy , Proto-Oncogene Proteins c-met
5.
Mol Carcinog ; 61(6): 537-548, 2022 06.
Article in English | MEDLINE | ID: mdl-35319795

ABSTRACT

Transglutaminase 2 (TG2) is an important mesothelioma cancer cell survival protein. However, the mechanism whereby TG2 maintains mesothelioma cell survival is not well understood. We present studies showing that TG2 drives hepatocyte growth factor (HGF)-dependent MET receptor signaling to maintain the aggressive mesothelioma cancer phenotype. TG2 increases HGF and MET messenger RNA and protein levels to enhance MET signaling. TG2 inactivation reduces MET tyrosine kinase activity to reduce cancer cell spheroid formation, invasion and migration. We also confirm that HGF/MET signaling is a biologically important mediator of TG2 action. Reducing MET level using genetic methods or treatment with MET inhibitors reduces spheroid formation, invasion and migration and this is associated with reduced MEK1/2 and ERK1/2. In addition, MEK1/2 and ERK1/2 inhibitors suppress the cancer phenotype. Moreover, MET knockout mesothelioma cells form 10-fold smaller tumors compared to wild-type cells and these tumors display reduced MET, MEK1/2, and ERK1/2 activity. These findings suggest that TG2 maintains HGF and MET levels in cultured mesothelioma cells and tumors to drive HGF/MET, MEK1/2, and ERK1/2 signaling to maintain the aggressive mesothelioma cancer phenotype.


Subject(s)
Hepatocyte Growth Factor , Mesothelioma, Malignant , Mesothelioma , Protein Glutamine gamma Glutamyltransferase 2 , Cell Movement , Hepatocyte Growth Factor/genetics , Hepatocyte Growth Factor/metabolism , Humans , Mesothelioma/genetics , Mesothelioma/pathology , Phenotype , Protein Glutamine gamma Glutamyltransferase 2/genetics , Protein Glutamine gamma Glutamyltransferase 2/metabolism
6.
Int J Mol Sci ; 23(3)2022 Jan 27.
Article in English | MEDLINE | ID: mdl-35163371

ABSTRACT

Vascular endothelial cells express glycoprotein 130 (gp130), which is utilized as a signaling receptor by cytokines in the interleukin-6 (IL-6) family. Several IL-6 family cytokines can be found in the circulatory system during physiological or pathological conditions, and may influence endothelial function and response. This study evaluated and compared the cellular and molecular responses induced by IL-6 family cytokines in human endothelial cells. A proteomic analysis showed that IL-6 family cytokines induce the release of a range of proteins from endothelial cells, such as C-C motif chemokine ligand 23, hepatocyte growth factor, and IL-6. Pathway analysis indicated that gp130-signaling in endothelial cells regulates several functions related to angiogenesis and immune cell recruitment. The present investigation also disclosed differences and similarities between different IL-6 family cytokines in their ability to induce protein release and regulate gene expression and intracellular signaling, in regards to which oncostatin M showed the most pronounced effect. Further, this study showed that soluble gp130 preferentially blocks trans-signaling-induced responses, but does not affect responses induced by classic signaling. In conclusion, IL-6 family cytokines induce both specific and overlapping molecular responses in endothelial cells, and regulate genes and proteins involved in angiogenesis and immune cell recruitment.


Subject(s)
Cytokines/metabolism , Human Umbilical Vein Endothelial Cells/metabolism , Interleukin-6/metabolism , Cytokine Receptor gp130/metabolism , Gene Expression Regulation , Humans , Interleukin-6/genetics , MAP Kinase Signaling System , Phosphorylation , Receptors, Interleukin-6/metabolism , STAT3 Transcription Factor/metabolism , Signal Transduction , Transcription, Genetic
7.
J Biol Chem ; 295(2): 481-503, 2020 01 10.
Article in English | MEDLINE | ID: mdl-31744884

ABSTRACT

Considering the role of proto-oncogene c-Met (c-Met) in oncogenesis, we examined the effects of the metastasis suppressor, N-myc downstream-regulated gene-1 (NDRG1), and two NDRG1-inducing thiosemicarbazone-based agents, Dp44mT and DpC, on c-Met expression in DU145 and Huh7 cells. NDRG1 silencing without Dp44mT and DpC up-regulated c-Met expression, demonstrating that NDRG1 modulates c-Met levels. Dp44mT and DpC up-regulated NDRG1 by an iron-dependent mechanism and decreased c-Met levels, c-Met phosphorylation, and phosphorylation of its downstream effector, GRB2-associated binding protein 1 (GAB1). However, incubation with Dp44mT and DpC after NDRG1 silencing or silencing of the receptor tyrosine kinase inhibitor, mitogen-inducible gene 6 (MIG6), decreased c-Met and its phosphorylation, suggesting NDRG1- and MIG6-independent mechanism(s). Lysosomal inhibitors rescued the Dp44mT- and DpC-mediated c-Met down-regulation in DU145 cells. Confocal microscopy revealed that lysosomotropic agents and the thiosemicarbazones significantly increased co-localization between c-Met and lysosomal-associated membrane protein 2 (LAMP2). Moreover, generation of c-Met C-terminal fragment (CTF) and its intracellular domain (ICD) suggested metalloprotease-mediated cleavage. In fact, Dp44mT increased c-Met CTF while decreasing the ICD. Dp44mT and a γ-secretase inhibitor increased cellular c-Met CTF levels, suggesting that Dp44mT induces c-Met CTF levels by increasing metalloprotease activity. The broad metalloprotease inhibitors, EDTA and batimastat, partially prevented Dp44mT-mediated down-regulation of c-Met. In contrast, the ADAM inhibitor, TIMP metallopeptidase inhibitor 3 (TIMP-3), had no such effect, suggesting c-Met cleavage by another metalloprotease. Notably, Dp44mT did not induce extracellular c-Met shedding that could decrease c-Met levels. In summary, the thiosemicarbazones Dp44mT and DpC effectively inhibit oncogenic c-Met through lysosomal degradation and metalloprotease-mediated cleavage.


Subject(s)
Antineoplastic Agents/pharmacology , Down-Regulation/drug effects , Lysosomes/drug effects , Proto-Oncogene Proteins c-met/genetics , Thiosemicarbazones/pharmacology , Cell Line, Tumor , Gene Expression Regulation, Neoplastic/drug effects , Humans , Lysosomes/genetics , Lysosomes/metabolism , Neoplasms/drug therapy , Neoplasms/genetics , Neoplasms/metabolism , Proteolysis/drug effects , Proto-Oncogene Mas , Proto-Oncogene Proteins c-met/metabolism
8.
J Reprod Dev ; 67(2): 89-97, 2021 Apr 21.
Article in English | MEDLINE | ID: mdl-33455972

ABSTRACT

Depletion of hepatocyte growth factor (HGF) or mesenchymal-epithelial transition factor (c-Met) in mice leads to fetal lethality and placental maldevelopment. However, the dynamic change pattern of HGF/c-Met signaling during placental development and its involvement in the early differentiation of trophoblasts remain to be elucidated. In this study, using in situ hybridization assay, we elaborately demonstrated the spatial-temporal expression of Hgf and c-Met in mouse placenta from E5.5, the very early stage after embryonic implantation, to E12.5, when the placental structure is well developed. The concentration of the soluble form of c-Met (sMet) in maternal circulation peaked at E10.5. By utilizing the induced differentiation model of mouse trophoblast stem cells (mTSCs), we found that HGF significantly promoted mTSC differentiation into syncytiotrophoblasts (STBs) and invasive parietal trophoblast giant cells (PTGCs). Interestingly, sMet efficiently reversed the effect of HGF on mTSC differentiation. These findings indicate that HGF/c-Met signaling participates in regulating placental trophoblast cell fate at the early differentiation stage and that sMet acts as an endogenous antagonist in this aspect.


Subject(s)
Hepatocyte Growth Factor/metabolism , Placenta/metabolism , Proto-Oncogene Proteins c-met/metabolism , Trophoblasts/metabolism , Animals , Biomarkers/metabolism , Cell Differentiation , Cell Lineage , Cell Proliferation , Female , Fibroblasts/metabolism , Gene Expression Profiling , Gene Expression Regulation, Developmental , In Situ Hybridization , Mice , Placentation , Pregnancy , Signal Transduction , Stem Cells/cytology , Time Factors
9.
Pediatr Surg Int ; 37(12): 1743-1753, 2021 Dec.
Article in English | MEDLINE | ID: mdl-34448078

ABSTRACT

PURPOSE: Total parental nutrition (TPN) causes gastrointestinal mucosal atrophy. The present study investigated the effects of hepatocyte growth factor (HGF) on the intestinal mucosal atrophy induced by TPN. METHODS: Rats underwent jugular vein catheterization and were divided into four groups: oral feeding (OF), TPN alone (TPN), TPN plus low-dose HGF (0.3 mg/kg/day; TPNLH), and TPN plus high-dose HGF (1.0 mg/kg/day; TPNHH). On day 7, rats were euthanized, and the small intestine was harvested and evaluated histologically. The expression of c-MET, a receptor of HGF, and nutrition transporter protein were evaluated using quantitative polymerase chain reaction. RESULTS: The jejunal villus height (VH) and absorptive mucosal surface area in the TPNHH group were significantly higher than in the TPN group (p < 0.05). The VH in the ileum showed the same trend only in the TPNHH group, albeit without statistical significance. The crypt cell proliferation rate (CCPR) of the jejunum in both HGF-treated groups was significantly higher than in the TPN group (p < 0.01). The expression of c-MET and transporter protein in all TPN-treated groups was decreased compared with that in the OF group. CONCLUSION: HGF attenuated TPN-associated intestinal mucosal atrophy by increasing the villus height, which was associated with an increase in CCPR.


Subject(s)
Hepatocyte Growth Factor , Parenteral Nutrition, Total , Animals , Atrophy , Intestinal Mucosa/pathology , Jejunum , Parenteral Nutrition, Total/adverse effects , Rats
10.
Breast Cancer Res ; 22(1): 30, 2020 03 18.
Article in English | MEDLINE | ID: mdl-32188473

ABSTRACT

BACKGROUND: Breast cancer is rare in men, but management is focused on tumor characteristics commonly found in female breast cancer. The tumor microenvironment of male breast cancer is less well understood, and insight may improve male breast cancer management. The hepatocyte growth factor (HGF)/c-MET axis and the stromal cell-derived factor-1 (CXCL12)/C-X-C chemokine receptor type 4 (CXCR4) axis are prognostic in women with breast cancer. We aimed to investigate these factors in male breast cancer and correlate them with patient survival. METHODS: From 841 Dutch males with breast cancer who were enrolled in the EORTC 10085/TBCRC/BIG/NABCG International Male Breast Cancer Program (NCT01101425) and diagnosed between 1990 and 2010, archival primary tumor samples were collected. Tissue microarrays were constructed with 3 cores per sample and used for immunohistochemical analysis of HGF, c-MET, CXCL12, and CXCR4. Overall survival (OS) of the patients without metastases (M0) was analyzed using the Kaplan-Meier method. The value of the markers regarding OS was determined using univariable and multivariable Cox regression analyses, providing hazard ratios (HRs) and 95% confidence intervals (95% CIs). RESULTS: Of 720 out of 841 patients, sufficient tissue was available for analysis; 487 out of 720 patients had M0 disease. Patients with high HGF expression and high CXCL12 expression had a superior OS (low vs high expression of both markers, 7.5 vs 13.0 years, hazard ratio [HR] 0.64, 95% CI 0.49-0.84, P = 0.001 [HGF]; 9.1 vs 15.3 years, HR 0.63, 95% CI 0.45-0.87, P = 0.005 [CXCL12]). Multivariate analysis identified HGF as an independent predictor for OS (HR 0.64, 95% CI 0.47-0.88, P = 0.001). CONCLUSIONS: HGF and CXCL12 tumor expression appear to identify male breast cancer patients with a relatively good prognosis. Possibly, this could support male breast cancer-specific management strategies in the future.


Subject(s)
Biomarkers, Tumor/metabolism , Breast Neoplasms, Male/mortality , Chemokine CXCL12/metabolism , Hepatocyte Growth Factor/metabolism , Tumor Microenvironment , Aged , Breast Neoplasms, Male/metabolism , Breast Neoplasms, Male/pathology , Humans , Male , Middle Aged , Prognosis , Retrospective Studies , Signal Transduction , Survival Rate
11.
Int J Mol Sci ; 21(8)2020 Apr 11.
Article in English | MEDLINE | ID: mdl-32290402

ABSTRACT

Unlike in normal epithelium, dysregulated overactivation of various proteases have been reported in cancers. Degradation of pericancerous extracellular matrix leading to cancer cell invasion by matrix metalloproteases is well known evidence. On the other hand, several cell-surface proteases, including type II transmembrane serine proteases (TTSPs), also induce progression through activation of growth factors, protease activating receptors and other proteases. Hepatocyte growth factor (HGF) known as a multifunctional growth factor that upregulates cancer cell motility, invasiveness, proliferative, and anti-apoptotic activities through phosphorylation of MET (a specific receptor of HGF). HGF secreted as inactive zymogen (pro-HGF) from cancer associated stromal fibroblasts, and the proteolytic activation by several TTSPs including matriptase and hepsin is required. The activation is strictly regulated by HGF activator inhibitors (HAIs) in physiological condition. However, downregulation is frequently observed in cancers. Indeed, overactivation of MET by upregulation of matriptase and hepsin accompanied by the downregulation of HAIs in urological cancers (prostate cancer, renal cell carcinoma, and bladder cancer) are also reported, a phenomenon observed in cancer cells with malignant phenotype, and correlated with poor prognosis. In this review, we summarized current reports focusing on TTSPs, HAIs, and MET signaling axis in urological cancers.


Subject(s)
Membrane Proteins/metabolism , Proto-Oncogene Proteins c-met/metabolism , Serine Proteases/metabolism , Urologic Neoplasms/etiology , Urologic Neoplasms/metabolism , Animals , Cell Line, Tumor , Cell Membrane/metabolism , Disease Susceptibility , Enzyme Activation , Hepatocyte Growth Factor/metabolism , Humans , Ligands , Protein Binding , Protein Interaction Domains and Motifs , Signal Transduction , Urologic Neoplasms/pathology
12.
Int J Mol Sci ; 21(2)2020 Jan 09.
Article in English | MEDLINE | ID: mdl-31936623

ABSTRACT

Melanoma is one of the most lethal and malignant cancers and its incidence is increasing worldwide, and Japan is not an exception. Although there are numerous therapeutic options for melanoma, the prognosis is still poor once it has metastasized. The main concern after removal of a primary melanoma is whether it has metastasized, and early detection of metastatic melanoma would be effective in improving the prognosis of patients. Thus, it is very important to identify reliable methods to detect metastases as early as possible. Although many prognostic biomarkers (mainly for metastases) of melanoma have been reported, there are very few effective for an early diagnosis. Serum and urinary biomarkers for melanoma diagnosis have especially received great interest because of the relative ease of sample collection and handling. Several serum and urinary biomarkers appear to have significant potential both as prognostic indicators and as targets for future therapeutic methods, but still there are no efficient serum and urinary biomarkers for early detection, accurate diagnosis and prognosis, efficient monitoring of the disease and reliable prediction of survival and recurrence. Levels of 5-S-cysteinyldopa (5SCD) in the serum or urine as biomarkers of melanoma have been found to be significantly elevated earlier and to reflect melanoma progression better than physical examinations, laboratory tests and imaging techniques, such as scintigraphy and echography. With recent developments in the treatment of melanoma, studies reporting combinations of 5SCD levels and new applications for the treatment of melanoma are gradually increasing. This review summarizes the usefulness of 5SCD, the most widely used and well-known melanoma marker in the serum and urine, compares 5SCD and other useful markers, and finally its application to other fields.


Subject(s)
Biomarkers, Tumor/metabolism , Cysteinyldopa/metabolism , Melanoma/metabolism , Skin Neoplasms/metabolism , Cysteinyldopa/blood , Drug Monitoring , Humans , Melanoma/blood , Melanoma/pathology , Metabolome , Skin Neoplasms/blood , Skin Neoplasms/pathology
13.
J Cell Physiol ; 234(4): 4256-4266, 2019 04.
Article in English | MEDLINE | ID: mdl-30191983

ABSTRACT

Research that pertains to the molecular mechanisms involved in retinal pigment epithelial (RPE) development can significantly contribute to cell therapy studies. The effects of periocular mesenchymal cells on the expansion of RPE cells remain elusive. We have examined the possible proliferative role of hepatocyte growth factor (HGF) as a mesenchymal cell secretory factor against human embryonic stem cell derived RPE (hESC-RPE). We found that the conditioned medium of human mesenchymal stem cells from apical papilla and/or exogenous HGF promoted proliferation of the hESC-RPE cells as single cells and cell sheets, in addition to rabbit RPE sheets in vitro. Blockage of HGF signaling by HGF receptor inhibitor, PHA-665752, inhibited proliferation of hESC-RPE cells. However, differentiation of hESCs and human-induced pluripotent stem cells to a rostral fate and eye-field specification was unaffected by HGF. Our in vivo analysis showed HGF expression in periocular mesenchymal cells after optic cup formation in chicken embryos. Administration of HGF receptor inhibitor at this developmental stage in chicken embryos led to reduced eye size and disorganization of the RPE sheet. These findings suggested that HGF administration could be beneficial for obtaining higher numbers of hESC-RPE cells in human preclinical and clinical trials.


Subject(s)
Cell Proliferation , Epithelial Cells/metabolism , Hepatocyte Growth Factor/metabolism , Human Embryonic Stem Cells/metabolism , Mesenchymal Stem Cells/metabolism , Paracrine Communication , Retinal Pigment Epithelium/metabolism , Adolescent , Animals , Cell Differentiation , Chick Embryo , Culture Media, Conditioned/metabolism , Eye/embryology , Eye/metabolism , Humans , Proto-Oncogene Proteins c-met/metabolism , Rabbits , Secretory Pathway , Signal Transduction , Young Adult
14.
J Cell Biochem ; 120(3): 3637-3650, 2019 03.
Article in English | MEDLINE | ID: mdl-30242894

ABSTRACT

Mesenchymal stem cells (MSCs) protect the endothelial barrier complex and survival, implicated in the pathogenesis of acute lung injury (ALI) via paracrine hepatocyte growth factor (HGF). However, the mechanism of HGF in endothelial regulation remains unclear. Here, we introduced a coculture protocol of pulmonary microvascular endothelial cells (PMVECs) and overexpression of the HGF gene of MSCs (MSC-HGF). Immunofluorescence and endothelial permeability analysis revealed that MSC-HGF protected endothelial tight junction protein occludin expression and attenuated cellular permeability as well as endothelial apoptosis. To investigate the novel mechanism mammalian TOR (mTOR)/ signal transducer and activator of transcription 3 (STAT-3) signaling in HGF protective effects against endothelial barrier and apoptosis, we used recombinant mouse HGF in endothelial cells. In addition, we used mTOR inhibitor rapamycin to inhibit the mTOR pathway. Our study demonstrated that rapamycin decreased the protective effects of HGF on the endothelium by decreasing tight junction protein occludin expression and cell proliferation, and raising lipopolysaccharide (LPS)-induced endothelial permeability, endothelial cell injury factors ET-1 and vWF. Similarly, the protective effects of HGF on reducing endothelial barrier and apoptosis were weakened when PMVECs were treated with the STAT-3 inhibitor S3I-201. Moreover, mTOR/STAT-3 were activated by HGF demonstrated as raising mTOR (Ser2448) and STAT3 (Ser727) phosphorylation proteins, leading to endothelial barrier improvement and survival. Reversely, rapamycin or S3I-201 inhibited mTOR/STAT-3 activation. Taken together, our findings highlight that the activation of the mTOR/STAT-3 pathway provides novel mechanistic insights into MSC-secreted HGF protection against LPS-induced vascular endothelial permeability dysfunction and apoptosis, which contributes to decreasing microvascular loss and lung injury.


Subject(s)
Apoptosis/drug effects , Capillary Permeability/drug effects , Endothelial Cells/metabolism , Hepatocyte Growth Factor/biosynthesis , Lipopolysaccharides/toxicity , Mesenchymal Stem Cells/metabolism , STAT3 Transcription Factor/metabolism , Signal Transduction/drug effects , TOR Serine-Threonine Kinases/metabolism , Animals , Endothelial Cells/pathology , Mesenchymal Stem Cells/pathology , Mice
15.
Growth Factors ; 37(3-4): 105-119, 2019 08.
Article in English | MEDLINE | ID: mdl-31452434

ABSTRACT

Nowadays, transplantation of human mesenchymal stem cells (MSCs) has emerged as a potential cellular therapy for liver cirrhosis. Hepatocyte growth factor (HGF) plays an important role in the regeneration of the liver. The objective of the study was to investigate the therapeutic effect of HGF-transfected human umbilical cord blood-derived MSCs on dimethylnitrosamine (DMN)-induced liver fibrosis in rats. HGF-transfected MSCs were transplanted into rats with DMN-induced liver fibrosis. H2O2-induced cytotoxicity, apoptosis and intracellular reactive oxygen species were reduced in HGF-transfected MSCs in HGF-transfected MSCs. Pro-apoptotic proteins, such as cleaved poly (ADP-ribose) polymerase and cleaved caspase-3, were decreased in HGF-transfected MSCs. Biochemical analysis showed that the levels of aspartate aminotransferase and alanine aminotransferase were decreased after transplantation of HGF-transfected MSCs in rat fibrosis. Trichrome staining showed that HGF-transfected MSCs reduced liver damage. Taken together, our study indicated that HGF-transfected MSCs have therapeutic effects on DMN-induced liver fibrosis in rats.


Subject(s)
Cell- and Tissue-Based Therapy/methods , Hepatocyte Growth Factor/genetics , Hepatocyte Growth Factor/metabolism , Liver Cirrhosis/pathology , Mesenchymal Stem Cell Transplantation/methods , Alanine Transaminase/metabolism , Animals , Apoptosis/physiology , Aspartate Aminotransferases/metabolism , Caspase 3/metabolism , Cells, Cultured , Dimethylnitrosamine/toxicity , Fetal Blood/cytology , Humans , Hydrogen Peroxide/metabolism , Liver Cirrhosis/chemically induced , Male , Mesenchymal Stem Cells/metabolism , Poly(ADP-ribose) Polymerases/metabolism , Rats , Rats, Sprague-Dawley , Transfection
16.
Osteoporos Int ; 30(2): 441-449, 2019 Feb.
Article in English | MEDLINE | ID: mdl-30306221

ABSTRACT

We explored relations between serum hepatocyte growth factor (HGF), disease activity, osteoproliferation, and bone mineral density (BMD) in ankylosing spondylitis (AS), in comparison with healthy controls. HGF was increased especially in male AS patients and smokers and associated with both lower BMD and more chronic radiographic changes in the spine. INTRODUCTION: Ankylosing spondylitis (AS) is characterized by both osteoproliferation and increased bone loss. Biomarkers are requested to predict the processes. The aims of this study were to compare serum levels of hepatocyte growth factor (HGF), matrix metalloproteinase-3 (MMP-3), and vascular endothelial growth factor (VEGF) in AS patients with healthy controls (HC) and to explore the associations with disease activity, osteoproliferation, and bone mineral density (BMD). METHODS: Serum from AS patients (modified NY-criteria) and HC was analyzed for HGF, MMP-3, and VEGF with ELISA. Disease activity parameters were collected. Osteoproliferation was assessed with modified Stoke Ankylosing Spondylitis Spine Score (mSASSS) and BMD was measured in femoral neck. RESULTS: Totally, 204 AS patients and 80 sex and age matched HC were included. Serum HGF was higher in the AS patients compared with the HC, whereas serum MMP-3 and VEGF were not. Serum HGF was also higher in smokers and in the male AS patients positively correlated with age, BASMI, and mSASSS, and negatively correlated with BMD. The biomarkers were all positively associated with ESR, CRP, and WBC. In multiple linear regression analysis serum HGF remained associated with higher mSASSS and lower BMD, after adjusting for age, sex, CRP, smoking, and body mass index. CONCLUSIONS: Serum HGF was increased in male AS patients and associated with higher mSASSS and lower BMD. In addition, serum HGF was positively associated with risk factors for osteoproliferation such as age, CRP and smoking. HGF could be a potential biomarker of importance for the bone metabolism in AS. TRIAL REGISTRATION: NCT00858819.


Subject(s)
Hepatocyte Growth Factor/blood , Osteogenesis/physiology , Osteoporosis/etiology , Spondylitis, Ankylosing/complications , Absorptiometry, Photon/methods , Adult , Aging/blood , Biomarkers/blood , Bone Density/physiology , Case-Control Studies , Female , Femur Neck/physiopathology , Humans , Male , Matrix Metalloproteinase 3/blood , Middle Aged , Osteoporosis/diagnosis , Osteoporosis/physiopathology , Severity of Illness Index , Spondylitis, Ankylosing/blood , Spondylitis, Ankylosing/pathology , Spondylitis, Ankylosing/physiopathology , Vascular Endothelial Growth Factor A/blood
17.
Exp Eye Res ; 184: 30-37, 2019 07.
Article in English | MEDLINE | ID: mdl-30978346

ABSTRACT

A patient with bilateral diffuse uveal melanocytic proliferation (BDUMP) associated with endometrial cancer was treated with plasmapheresis, but failed therapy with progressive serous retinal detachment. We collected plasma before and after plasmapheresis therapy. Our goal was to determine if the cultured melanocyte elongation and proliferation (CMEP) factor and hepatocyte growth factor (HGF) was present in the IgG enriched fraction and understand why our patient failed plasmapheresis therapy. Melanocytes were cultured for 3-5 days in the presence of control medium, unfractionated pre-plasmapheresis BDUMP medium, IgG enriched or IgG depleted BDUMP medium, or unfractionated post-plasmapheresis BDUMP medium. Subretinal fluid was collected from patients with BDUMP and control retinal detachments and analyzed by electropheresis with immunoblotting. Medium with unfractionated BDUMP plasma stimulated melanocyte growth 1.4-1.5 fold compared to control medium on days 3-5 (p < 0.001 for all). Both IgG enriched and IgG depleted BDUMP medium mildly increased melanocyte growth 1.3 fold (p < 0.05 for enriched, p < 0.01 for depleted) compared to control. In comparison, unfractionated BDUMP medium caused a 1.7-fold increase in melanocyte growth, which was significantly more than the enriched (p < 0.01) and depleted (p < 0.05) fractions. Pre-plasmapheresis and post-plasmapheresis unfractionated BDUMP medium equally stimulated melanocyte growth 1.7-fold (p < 0.05) compared to control. HGF was present in IgG depleted, pre-plasmapheresis, and post-plasmapheresis samples, but absent in the IgG enriched fraction. There was no enrichment of IgG in the subretinal fluid from eyes with BDUMP. In conclusion, CMEP factor is not concentrated in the IgG enriched plasma fraction in our patient who failed plasmapheresis therapy. HGF levels have no correlation with melanocyte growth. Because plasmapheresis preferentially removes immunoglobulins from the plasma, our patient responded poorly to plasmapheresis treatment with worsening retinal detachment.


Subject(s)
Adenocarcinoma, Clear Cell/pathology , Endometrial Neoplasms/pathology , Intercellular Signaling Peptides and Proteins/blood , Melanocytes/pathology , Paraneoplastic Syndromes, Ocular/pathology , Uvea/pathology , Adenocarcinoma, Clear Cell/blood , Adenocarcinoma, Clear Cell/therapy , Aged , Cell Proliferation , Cells, Cultured , Electrophoresis, Polyacrylamide Gel , Endometrial Neoplasms/blood , Endometrial Neoplasms/therapy , Female , Fluorescein Angiography , Humans , Immunoblotting , Multimodal Imaging , Paraneoplastic Syndromes, Ocular/blood , Paraneoplastic Syndromes, Ocular/therapy , Plasmapheresis , Subretinal Fluid , Treatment Failure
18.
J Biochem Mol Toxicol ; 33(6): e22316, 2019 Jun.
Article in English | MEDLINE | ID: mdl-30897285

ABSTRACT

The system of hepatocyte growth factor (HGF) and its receptor c-Met plays a critical role in tumor invasive growth and metastasis. The mortality rate of colorectal cancer (CRC), one of the most commonly diagnosed malignancies, is increased by it gradual development into metastasis, most frequently in the liver. Overexpression of c-Met, the protein tyrosine kinase receptor for the HCF/scatter factor, has been implicated in the progression and metastasis of human colorectal carcinoma. In this study, we aimed to investigate the role of c-Met in CRC liver metastasis and illustrate the clinical impact of regulating HGF/c-Met signaling in patients with CRC liver metastasis. We found that (I) higher levels of c-Met expression (mRNA and Protein) in CRC liver metastasis than primary CRC by assessing the patient tissue samples; (II) a positive correlation of c-Met expression with tumor stages of CRC liver metastasis, as well as c-Met expression in CRC, live metastasis concurred with regional lymph node metastasis; (III) the clinical impact of downregulation of HGF/c-Met signaling on the reduction of proliferation and invasion in CRC liver metastasis. Therefore, we demonstrate that the regulation of HGF/c-Met pathways may be a promising strategy in the treatment of patients with CRC liver metastasis.


Subject(s)
Colorectal Neoplasms , Gene Expression Regulation, Neoplastic , Hepatocyte Growth Factor/biosynthesis , Liver Neoplasms , Liver/metabolism , Proto-Oncogene Proteins c-met/biosynthesis , Aged , Aged, 80 and over , Colorectal Neoplasms/metabolism , Colorectal Neoplasms/mortality , Colorectal Neoplasms/pathology , Colorectal Neoplasms/therapy , Female , Humans , Liver/pathology , Liver Neoplasms/metabolism , Liver Neoplasms/mortality , Liver Neoplasms/secondary , Liver Neoplasms/therapy , Lymphatic Metastasis , Male , Middle Aged , Neoplasm Invasiveness
19.
Int J Mol Sci ; 20(10)2019 May 22.
Article in English | MEDLINE | ID: mdl-31121879

ABSTRACT

Hepatocyte growth factor (HGF) and transforming growth factor ß1 (TGFß1) are biological stimuli of the micro-environment which affect bone metastasis phenotype through transcription factors, but their influence on the growth is scarcely known. In a xenograft model prepared with 1833 bone metastatic cells, derived from breast carcinoma cells, we evaluated mice survival and Twist and Snail expression and localization after competitive inhibition of HGF with NK4, or after blockade of TGFß1-type I receptor (RI) with SB431542: in the latter condition HGF was also measured. To explain the in vivo data, in 1833 cells treated with SB431542 plus TGFß1 we measured HGF formation and the transduction pathway involved. Altogether, HGF seemed relevant for bone-metastatic growth, being hampered by NK4 treatment, which decreased Twist more than Snail in the metastasis bulk. TGFß1-RI blockade enhanced HGF in metastasis and adjacent bone marrow, while reducing prevalently Snail expression at the front and bulk of bone metastasis. The HGF accumulation in 1833 cells depended on an auxiliary signaling pathway, triggered by TGFß1 under SB431542, which interfered in the transcription of HGF activator inhibitor type 1 (HAI-1) downstream of TGFß-activated kinase 1 (TAK1): HGF stimulated Twist transactivation. In conclusion, the impairment of initial outgrowth with NK4 seemed therapeutically promising more than SB431542 chemotherapy; a functional correlation between Twist and Snail in bone metastasis seemed to be influenced by the biological stimuli of the micro-environment, and the targeting of these phenotype biomarkers might inhibit metastasis plasticity and colonization, even if it would be necessary to consider the changes of HGF levels in bone metastases undergoing TGFß1-RI blockade.


Subject(s)
Benzamides/therapeutic use , Bone Neoplasms/drug therapy , Bone Neoplasms/secondary , Breast Neoplasms/pathology , Dioxoles/therapeutic use , Hepatocyte Growth Factor/antagonists & inhibitors , Receptor, Transforming Growth Factor-beta Type I/antagonists & inhibitors , Animals , Bone Neoplasms/pathology , Breast Neoplasms/drug therapy , Cell Line, Tumor , Epithelial-Mesenchymal Transition , Female , Humans , Mice
20.
Bull Exp Biol Med ; 166(3): 339-343, 2019 Jan.
Article in English | MEDLINE | ID: mdl-30627913

ABSTRACT

We studied effects of semaphorin 3A, keratinocyte growth factor (KGF), hepatocyte growth factor (HGF), and their combinations on the proliferative activity of cortical (cTEC1-2) and medullary (mTEC3-10) thymus epithelium cell lines. Semaphorin 3A inhibited the proliferative activity of epithelial cells, while HGF and KGF, in contrast, exerted a stimulating effect. The effect of KGF and semaphorin 3A on different cell lines depended on the expression of receptors for these two factors. When the combination of two factors was used, semaphorin 3A was able to neutralize the stimulating effect of HGF and KGF. It can be assumed that semaphorin 3A synthesized in the thymus stroma, can act as a functional antagonist of HGF and KGF and have an inhibitory effect when these drugs are administered into the body for the therapeutic purpose of restoring thymus functions.


Subject(s)
Cell Proliferation/drug effects , Epithelial Cells/drug effects , Fibroblast Growth Factor 7/pharmacology , Hepatocyte Growth Factor/pharmacology , RNA, Messenger/genetics , Semaphorin-3A/pharmacology , Animals , Cell Line , Drug Combinations , Epithelial Cells/cytology , Epithelial Cells/metabolism , Fibroblast Growth Factor 7/genetics , Fibroblast Growth Factor 7/metabolism , Gene Expression Regulation , Hepatocyte Growth Factor/genetics , Hepatocyte Growth Factor/metabolism , Mice , Nerve Tissue Proteins/genetics , Nerve Tissue Proteins/metabolism , Neuropilin-1/genetics , Neuropilin-1/metabolism , Protein Isoforms/genetics , Protein Isoforms/metabolism , RNA, Messenger/metabolism , Receptor Protein-Tyrosine Kinases/genetics , Receptor Protein-Tyrosine Kinases/metabolism , Receptor, Fibroblast Growth Factor, Type 2/genetics , Receptor, Fibroblast Growth Factor, Type 2/metabolism , Receptors, Cell Surface/genetics , Receptors, Cell Surface/metabolism , Semaphorin-3A/genetics , Semaphorin-3A/metabolism , Signal Transduction , Thymus Gland/cytology , Thymus Gland/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL