Your browser doesn't support javascript.
loading
Montrer: 20 | 50 | 100
Résultats 1 - 20 de 2.222
Filtrer
1.
Stem Cell Res Ther ; 15(1): 166, 2024 Jun 13.
Article de Anglais | MEDLINE | ID: mdl-38867276

RÉSUMÉ

BACKGROUND: Hypertrophic scarring results from myofibroblast differentiation and persistence during wound healing. Currently no effective treatment for hypertrophic scarring exists however, autologous fat grafting has been shown to improve scar elasticity, appearance, and function. The aim of this study was to understand how paracrine factors from adipose tissues and adipose-derived stromal cells (ADSC) affect fibroblast to myofibroblast differentiation. METHODS: The transforming growth factor-ß1 (TGF-ß1) induced model of myofibroblast differentiation was used to test the effect of conditioned media from adipose tissue, ADSC or lipid on the proportion of fibroblasts and myofibroblasts. RESULTS: Adipose tissue conditioned media inhibited the differentiation of fibroblasts to myofibroblasts but this inhibition was not observed following treatment with ADSC or lipid conditioned media. Hepatocyte growth factor (HGF) was readily detected in the conditioned medium from adipose tissue but not ADSC. Cells treated with HGF, or fortinib to block HGF, demonstrated that HGF was not responsible for the inhibition of myofibroblast differentiation. Conditioned media from adipose tissue was shown to reduce the proportion of myofibroblasts when added to fibroblasts previously treated with TGF-ß1, however, conditioned media treatment was unable to significantly reduce the proportion of myofibroblasts in cell populations isolated from scar tissue. CONCLUSIONS: Cultured ADSC or adipocytes have been the focus of most studies, however, this work highlights the importance of considering whole adipose tissue to further our understanding of fat grafting. This study supports the use of autologous fat grafts for scar treatment and highlights the need for further investigation to determine the mechanism.


Sujet(s)
Tissu adipeux , Différenciation cellulaire , Facteur de croissance des hépatocytes , Myofibroblastes , Facteur de croissance transformant bêta-1 , Myofibroblastes/métabolisme , Myofibroblastes/effets des médicaments et des substances chimiques , Myofibroblastes/cytologie , Facteur de croissance transformant bêta-1/pharmacologie , Facteur de croissance transformant bêta-1/métabolisme , Tissu adipeux/cytologie , Tissu adipeux/métabolisme , Différenciation cellulaire/effets des médicaments et des substances chimiques , Milieux de culture conditionnés/pharmacologie , Humains , Facteur de croissance des hépatocytes/pharmacologie , Facteur de croissance des hépatocytes/métabolisme , Communication paracrine/effets des médicaments et des substances chimiques , Phénotype , Cellules cultivées , Fibroblastes/métabolisme , Fibroblastes/effets des médicaments et des substances chimiques , Fibroblastes/cytologie , Adipocytes/métabolisme , Adipocytes/cytologie , Adipocytes/effets des médicaments et des substances chimiques , Cellules stromales/métabolisme , Cellules stromales/cytologie , Cellules stromales/effets des médicaments et des substances chimiques
2.
Nature ; 630(8015): 158-165, 2024 Jun.
Article de Anglais | MEDLINE | ID: mdl-38693268

RÉSUMÉ

The liver has a unique ability to regenerate1,2; however, in the setting of acute liver failure (ALF), this regenerative capacity is often overwhelmed, leaving emergency liver transplantation as the only curative option3-5. Here, to advance understanding of human liver regeneration, we use paired single-nucleus RNA sequencing combined with spatial profiling of healthy and ALF explant human livers to generate a single-cell, pan-lineage atlas of human liver regeneration. We uncover a novel ANXA2+ migratory hepatocyte subpopulation, which emerges during human liver regeneration, and a corollary subpopulation in a mouse model of acetaminophen (APAP)-induced liver regeneration. Interrogation of necrotic wound closure and hepatocyte proliferation across multiple timepoints following APAP-induced liver injury in mice demonstrates that wound closure precedes hepatocyte proliferation. Four-dimensional intravital imaging of APAP-induced mouse liver injury identifies motile hepatocytes at the edge of the necrotic area, enabling collective migration of the hepatocyte sheet to effect wound closure. Depletion of hepatocyte ANXA2 reduces hepatocyte growth factor-induced human and mouse hepatocyte migration in vitro, and abrogates necrotic wound closure following APAP-induced mouse liver injury. Together, our work dissects unanticipated aspects of liver regeneration, demonstrating an uncoupling of wound closure and hepatocyte proliferation and uncovering a novel migratory hepatocyte subpopulation that mediates wound closure following liver injury. Therapies designed to promote rapid reconstitution of normal hepatic microarchitecture and reparation of the gut-liver barrier may advance new areas of therapeutic discovery in regenerative medicine.


Sujet(s)
Défaillance hépatique aigüe , Régénération hépatique , Animaux , Femelle , Humains , Mâle , Souris , Acétaminophène/pharmacologie , Lignage cellulaire , Mouvement cellulaire/effets des médicaments et des substances chimiques , Prolifération cellulaire/effets des médicaments et des substances chimiques , Lésions hépatiques dues aux substances/anatomopathologie , Modèles animaux de maladie humaine , Facteur de croissance des hépatocytes/métabolisme , Facteur de croissance des hépatocytes/pharmacologie , Hépatocytes/cytologie , Hépatocytes/effets des médicaments et des substances chimiques , Hépatocytes/métabolisme , Hépatocytes/anatomopathologie , Foie/cytologie , Foie/effets des médicaments et des substances chimiques , Foie/anatomopathologie , Défaillance hépatique aigüe/anatomopathologie , Défaillance hépatique aigüe/induit chimiquement , Régénération hépatique/effets des médicaments et des substances chimiques , Souris de lignée C57BL , Nécrose/induit chimiquement , Médecine régénérative , Analyse de l'expression du gène de la cellule unique , Cicatrisation de plaie
3.
Expert Opin Investig Drugs ; 33(7): 713-720, 2024 Jul.
Article de Anglais | MEDLINE | ID: mdl-38783527

RÉSUMÉ

INTRODUCTION: Spinal cord injury (SCI) is a condition in which the spinal cord parenchyma is damaged by various factors. The mammalian central nervous system has been considered unable to regenerate once damaged, but recent progress in basic research has gradually revealed that injured neural cells can indeed regenerate. Drug therapy using novel agents is being actively investigated as a new treatment for SCI. One notable treatment method is regeneration therapy using hepatocyte growth factors (HGF). AREA COVERED: HGF has pluripotent neuroregenerative actions, as indicated by its neuroprotective and regenerative effects on the microenvironment and damaged cells, respectively. This review examines these effects in various phases of SCI, from basic research to clinical studies, and the application of this treatment to other diseases. EXPERT OPINION: In regenerative medicine for SCI, drug therapies have tended to be more likely to be developed compared to cell replacement treatment. Nevertheless, there are still challenges to be addressed for these clinical applications due to a wide variety of pathology and animal experimental models of basic study, but HGF could be an effective treatment for SCI with expanded application.


Sujet(s)
Facteur de croissance des hépatocytes , Neuroprotecteurs , Médecine régénérative , Traumatismes de la moelle épinière , Traumatismes de la moelle épinière/traitement médicamenteux , Traumatismes de la moelle épinière/physiopathologie , Facteur de croissance des hépatocytes/pharmacologie , Facteur de croissance des hépatocytes/métabolisme , Animaux , Humains , Neuroprotecteurs/pharmacologie , Neuroprotecteurs/administration et posologie , Médecine régénérative/méthodes , Modèles animaux de maladie humaine , Régénération nerveuse/effets des médicaments et des substances chimiques , Développement de médicament
4.
Tissue Cell ; 87: 102326, 2024 Apr.
Article de Anglais | MEDLINE | ID: mdl-38442547

RÉSUMÉ

BACKGROUND: Transplantation of mesenchymal stem cells (MSCs) is a newly developed strategy for treating acute liver failure (ALF). Nonetheless, the low survival rate of MSCs after transplantation and their poor homing to damaged tissues limit the clinical application of MSCs. The research assessed whether hypoxic preconditioning (HPC) can improve the biological activity of human amniotic mesenchymal stem cells (hA-MSCs), promote their homing ability to the liver of mice with ALF, and influence liver tissue repair. METHODS: Flow cytometry, CCK8, Transwell, and Western blotting assays were conducted to assess the effects of hypoxic preconditioning on the phenotype, proliferation, and migration of hA-MSCs and the changes in the c-Met and CXCR4 gene expression levels were studied. To evaluate the effects of the transplantation of hypoxic preconditioning of hA-MSCs on the homing and repair of D-galactosamine (D-GalN)/LPS-induced ALF, the mechanism was elucidated by adding c-Met, CXCR4-specific blockers (SU11274 and AMD3100). RESULTS: After hypoxia pretreatment (1% oxygen volume fraction), hA-MSCs maintained the morphological characteristics of adherence and vortex colony growth and showed high CD44, CD90, and CD105 and low CD31, CD34, and CD45 expression levels. Hypoxic preconditioning of hA-MSCs significantly increased their proliferation and migration and highly expressed the c-Met and CXCR4 genes. In vivo and in vitro, this migration-promoting effect was suppressed by the c-Met specific blocker SU11274. In the acute liver failure mouse model, the HGF expression level was considerably elevated in the liver than that in the serum, lungs and kidneys. The transplantation of hypoxic preconditioned hA-MSCs introduced a remarkable improvement in the liver function and survival rate of mice with ALF and enhanced the anti-apoptosis ability of liver cells. The anti-apoptotic enhancing effect of hypoxic preconditioning was suppressed by the c-Met specific blocker SU11274. Hypoxic hA-MSCs administration was observed to have considerably increased the fluorescent cells in the liver than that recorded after administering normal oxygen-hA-MSCs. The number of hepatic fluorescent cells decreased remarkably after adding the c-Met inhibitor SU11274, compared to that recorded after hypoxic pretreatment, whereas the effect of c-Met inhibitor SU11274 on normal oxygen-hA-MSCs was not significant. CONCLUSIONS: Hypoxic preconditioning depicted no impact on the morphology and phenotype features of the human amniotic mesenchymal stem cells, but it can promote their proliferation, migration, anti-apoptotic effect, and homing rate and improve the repair of acute liver failure, which might be mediated by the HGF/c-Met signaling axis.


Sujet(s)
Indoles , Défaillance hépatique aigüe , Transplantation de cellules souches mésenchymateuses , Cellules souches mésenchymateuses , Pipérazines , Sulfonamides , Humains , Souris , Animaux , Défaillance hépatique aigüe/thérapie , Défaillance hépatique aigüe/métabolisme , Hypoxie/métabolisme , Oxygène/métabolisme , Prolifération cellulaire , Facteur de croissance des hépatocytes/métabolisme , Facteur de croissance des hépatocytes/pharmacologie
5.
Cell Mol Life Sci ; 81(1): 28, 2024 Jan 12.
Article de Anglais | MEDLINE | ID: mdl-38212428

RÉSUMÉ

Although amplifications and mutations in receptor tyrosine kinases (RTKs) act as bona fide oncogenes, in most cancers, RTKs maintain moderate expression and remain wild-type. Consequently, cognate ligands control many facets of tumorigenesis, including resistance to anti-RTK therapies. Herein, we show that the ligands for the RTKs MET and RON, HGF and HGFL, respectively, are synthesized as inactive precursors that are activated by cellular proteases. Our newly generated HGF/HGFL protease inhibitors could overcome both de novo and acquired cetuximab resistance in colorectal cancer (CRC). Conversely, HGF overexpression was necessary and sufficient to induce cetuximab resistance and loss of polarity. Moreover, HGF-induced cetuximab resistance could be overcome by the downstream MET inhibitor, crizotinib, and upstream protease inhibitors. Additionally, HAI-1, an endogenous inhibitor of HGF proteases, (i) was downregulated in CRC, (ii) exhibited increased genomic methylation that correlated with poor prognosis, (iii) HAI-1 expression correlated with cetuximab response in a panel of cancer cell lines, and (iv) exogenous addition of recombinant HAI-1 overcame cetuximab resistance in CC-HGF cells. Thus, we describe a targetable, autocrine HAI-1/Protease/HGF/MET axis in cetuximab resistance in CRC.


Sujet(s)
Tumeurs colorectales , Transduction du signal , Humains , Cétuximab/pharmacologie , Protéines proto-oncogènes c-met/génétique , Protéines proto-oncogènes c-met/métabolisme , Résistance aux médicaments antinéoplasiques/génétique , Tumeurs colorectales/traitement médicamenteux , Tumeurs colorectales/génétique , Tumeurs colorectales/métabolisme , Inhibiteurs de protéases/pharmacologie , Peptide hydrolases/métabolisme , Lignée cellulaire tumorale , Facteur de croissance des hépatocytes/génétique , Facteur de croissance des hépatocytes/métabolisme , Facteur de croissance des hépatocytes/pharmacologie
6.
Radiother Oncol ; 190: 109984, 2024 Jan.
Article de Anglais | MEDLINE | ID: mdl-37926332

RÉSUMÉ

BACKGROUND & AIM: Irradiation of the salivary glands during head and neck cancer treatment induces cellular senescence in response to DNA damage and contributes to radiation-induced hyposalivation by affecting the salivary gland stem/progenitor cell (SGSC) niche. Cellular senescence, such as that induced by radiation, is a state of cell-cycle arrest, accompanied by an altered pro-inflammatory secretome known as the senescence-associated secretory phenotype (SASP) with potential detrimental effects on the surrounding microenvironment. We hypothesized that the pro-regenerative properties of mesenchymal stem cells (MSCs) may attenuate cellular senescence post-irradiation. Therefore, here we evaluated the effects of adipose-derived MSCs (ADSCs) on the radiation-induced response of salivary gland organoids (SGOs). METHODS: Proteomic analyses to identify soluble mediators released by ADSCs co-cultured with SGOS revealed secretion of hepatocyte growth factor (HGF) in ADSCs, suggesting a possible role in the stem cell crosstalk. Next, the effect of recombinant HGF in the culture media of ex vivo grown salivary gland cells was tested in 2D monolayers and 3D organoid models. RESULTS: Treatment with HGF robustly increased salivary gland cell proliferation. Importantly, HGF supplementation post-irradiation enhanced proliferation at lower doses of radiation (0, 3, 7 Gy), but not at higher doses (10, 14 Gy) where most cells stained positive for senescence-associated beta-galactosidase. Furthermore, HGF had no effect on the senescence-associated secretory phenotype (SASP) of irradiated SGOs, suggesting there may be compensatory proliferation by cell-division competent cells instead of a reversal of cellular senescence after irradiation. CONCLUSION: ADSCs may positively influence radiation recovery through HGF secretion and can promote the ex vivo expansion of salivary gland stem/progenitor cells to enhance the effects of co-transplanted SGSC.


Sujet(s)
Facteur de croissance des hépatocytes , Cellules souches mésenchymateuses , Humains , Facteur de croissance des hépatocytes/pharmacologie , Protéomique , Glandes salivaires , Vieillissement de la cellule/effets des radiations , Prolifération cellulaire
7.
Macromol Biosci ; 24(3): e2300356, 2024 Mar.
Article de Anglais | MEDLINE | ID: mdl-37877161

RÉSUMÉ

Small cell lung cancer (SCLC) is one of lethal cancers resulting in very low 5-year-survival rate. Although its clinical treatment largely relies on chemotherapy, SCLC cell physiology in three-dimenstional (3D) matrix has been less explored. In this work, the tumor microenvironment is reconstructed with decellularized porcine pulmonary extracellular matrix (dECM) with hyaluronic acid. To modulate matrix stiffness, the methacrylate groups are introduced into both dECM and hyaluronic acid, followed by photocrosslinking with photoinitiator. The stiffness of the resulting dECM-based hydrogel covers the stiffness of normal or cancerous tissue with varying dECM content. The proliferation and cancer stem cell marker expression of encapsulated SCLC cells are promoted in a compliant hydrogel matrix, which has a low shear modulus similar to that of the normal tissue. The hepatocyte growth factor (HGF) that induces SCLC cell invasion and chemoresistance markedly increases invasiveness and gene expression levels of CD44 and Sox2 in the hydrogel matrix. In addition, HGF treatment causes higher resistance against anticancer drugs (cisplatin and paclitaxel) in the 3D microenvironment. These findings indicate that malignant SCLC can be recapitulated in a pulmonary dECM-based matrix.


Sujet(s)
Tumeurs du poumon , Carcinome pulmonaire à petites cellules , Animaux , Suidae , Hydrogels/pharmacologie , Hydrogels/métabolisme , Matrice extracellulaire/métabolisme , Matrice extracellulaire décellularisée , Carcinome pulmonaire à petites cellules/métabolisme , Acide hyaluronique/pharmacologie , Facteur de croissance des hépatocytes/pharmacologie , Facteur de croissance des hépatocytes/métabolisme , Tumeurs du poumon/traitement médicamenteux , Tumeurs du poumon/métabolisme , Ingénierie tissulaire/méthodes , Microenvironnement tumoral
8.
Anticancer Agents Med Chem ; 24(1): 30-38, 2024.
Article de Anglais | MEDLINE | ID: mdl-37957870

RÉSUMÉ

BACKGROUND: The biological behavior of cells changes after they develop drug resistance, and the degree of resistance will be affected by the tumor microenvironment. In this study, we aimed to study the effects of M2 macrophages on gefitinib resistance. METHODS: We polarized THP-1 cells into M0 and M2 macrophages, and conducted various experiments to investigate the effects of M2 macrophages on gefitinib resistance in lung cancer. RESULTS: We found that M2 macrophages promote gefitinib resistance in HCC827 and PC9 cells. In addition, we used ELISA to measure the secretion level of HGF. HGF secretion levels were significantly increased in M2 macrophages. Exogenous HGF remarkably increased the proliferation and invasion in HCC827 and PC9 cells. However, the addition of anti-HGF antibodies abolished the proliferation and invasion of both HCC827 and PC9 cells promoted by M2 macrophages. Furthermore, M2 macrophages or exogenous HGF significantly increased the expression of p-met and p-ERK in HCC827 and PC9 cells, while anti-HGF antibodies diminished the expression of p-met and p-ERK by neutralizing HGF in M2 macrophages. CONCLUSION: Our results revealed that M2 macrophages promote gefitinib resistance by activating ERK and HGF/c-met signaling pathways in HCC827 and PC9 cells. Our findings provide a new therapeutic strategy for gefitinib resistance in lung cancer.


Sujet(s)
Antinéoplasiques , Carcinome pulmonaire non à petites cellules , Tumeurs du poumon , Humains , Géfitinib/pharmacologie , Tumeurs du poumon/anatomopathologie , Carcinome pulmonaire non à petites cellules/anatomopathologie , Macrophages associés aux tumeurs/métabolisme , Macrophages associés aux tumeurs/anatomopathologie , Récepteurs ErbB/métabolisme , Quinazolines/pharmacologie , Résistance aux médicaments antinéoplasiques , Lignée cellulaire tumorale , Transduction du signal , Antinéoplasiques/pharmacologie , Antinéoplasiques/usage thérapeutique , Microenvironnement tumoral , Facteur de croissance des hépatocytes/pharmacologie , Facteur de croissance des hépatocytes/métabolisme , Facteur de croissance des hépatocytes/usage thérapeutique
9.
J Biomed Mater Res B Appl Biomater ; 112(1): e35317, 2024 01.
Article de Anglais | MEDLINE | ID: mdl-37584376

RÉSUMÉ

Tissue-engineered blood vessel substitutes have been developed due to the lack of suitable small-diameter vascular grafts. Xenogeneic extracellular matrix (ECM) scaffolds have the potential to provide an ideal source for off-the-shelf vascular grafts. In this study, porcine carotid arteries were used to develop ECM scaffolds by decellularization and coating with heparin and hepatocyte growth factor (HGF). After decellularization, cellular and nucleic materials were successfully removed with preservation of the main compositions (collagen, elastin, and basement membrane) of the native ECM. The ultimate tensile strength, suture strength, and burst pressure were significantly increased after cross-linking. Pore size distribution analysis revealed a porous structure within ECM scaffolds with a high distribution of pores larger than 10 µm. Heparinized scaffolds exhibited sustained release of heparin in vitro and showed potent anticoagulant activity by prolonging activated partial thromboplastin time. The scaffolds showed an enhanced HGF binding capacity as well as a constant release of HGF as a result of heparin modification. When implanted subcutaneously in rats, the modified scaffolds revealed good biocompatibility with enzyme degradation resistance, mitigated immune response, and anti-calcification. In conclusion, heparinized and HGF-coated acellular porcine carotid arteries may be a promising biological scaffold for tissue-engineered vascular grafts.


Sujet(s)
Facteur de croissance des hépatocytes , Structures d'échafaudage tissulaires , Suidae , Rats , Animaux , Structures d'échafaudage tissulaires/composition chimique , Facteur de croissance des hépatocytes/pharmacologie , Facteur de croissance des hépatocytes/analyse , Ingénierie tissulaire , Artères carotides/composition chimique , Prothèse vasculaire , Héparine/pharmacologie , Héparine/composition chimique , Matrice extracellulaire/composition chimique
10.
J Pediatr Surg ; 59(4): 627-633, 2024 Apr.
Article de Anglais | MEDLINE | ID: mdl-38160183

RÉSUMÉ

BACKGROUND: We evaluated the effect of recombinant human hepatocyte growth factor (rh-HGF) on intestinal adaptation in a rat model of short-bowel syndrome (SBS). METHODS: Sprague-Dawley rats underwent jugular vein catheterization for continuous total parenteral nutrition (TPN) and 90 % small bowel resection. The animals were divided into 3 groups: TPN/SBS (control group, n = 7), TPN/SBS/intravenous recombinant human hepatocyte growth factor (HGF) (0.3 mg/kg/day) (HGF group, n = 7), and TPN/SBS/intravenous c-Met inhibitor (0.3 mg/kg/day) (anti-HGF group, n = 5). On day 7, rats were euthanized and histologically evaluated. Serum diamine oxidase (S-DAO) levels were evaluated using an enzyme-linked immunosorbent assay. The nutrient transporter and glucagon-like peptide-2 (GLP-2) receptor expression were evaluated using real-time polymerase chain reaction. RESULTS: The jejunal and ileal villus heights were higher and the S-DAO concentrations significantly higher (p = 0.04) in the HGF group than in the control and anti-HGF groups. The sodium-dependent glucose transporter 1 expression in the HGF group was significantly higher than in the control group and significantly suppressed in the anti-HGF group (p < 0.01). The peptide transporter 1 expression in the jejunum was higher in the HGF group than in the other groups and significantly suppressed in the anti-HGF group (p < 0.01). The GLP-2 receptor expression in the jejunum was higher in the HGF group than the other groups, and it was significantly suppressed in the anti-HGF group (p < 0.01). These jejunal results regarding nutrient transporter an GLP-2 receptor were not found in the ileum. CONCLUSIONS: The administration of rh-HGF appears to be more effective in the jejunum than in the ileum. TYPE OF STUDY: Experimental Research. LEVEL OF EVIDENCE: N/A.


Sujet(s)
Jéjunum , Syndrome de l'intestin court , Animaux , Humains , Rats , Adaptation physiologique , Modèles animaux de maladie humaine , Récepteur du peptide-2 similaire au glucagon/métabolisme , Facteur de croissance des hépatocytes/pharmacologie , Muqueuse intestinale/métabolisme , Intestins/anatomopathologie , Jéjunum/anatomopathologie , Rat Sprague-Dawley , Syndrome de l'intestin court/métabolisme
11.
ACS Appl Bio Mater ; 6(12): 5252-5263, 2023 Dec 18.
Article de Anglais | MEDLINE | ID: mdl-37955977

RÉSUMÉ

The surface modification of biologically active factors on tissue-engineering vascular scaffold fails to fulfill the mechanical property and bioactive compounds' sustained release in vivo and results in the inhibition of tissue regeneration of small-diameter vascular grafts in vascular replacement therapies. In this study, biodegradable poly(ε-caprolactone) (PCL) was applied for scaffold preparation, and poly(ethylene glycol) (PG) hydrogel was used to load heparin and hepatocyte growth factor (HGF). In vitro analysis demonstrated that the PCL scaffold could inhibit the heparin release from the PG hydrogel, and the PG hydrogel could inhibit heparin release during the process of PCL degradation. Finally, it results in sustained release of HGF and heparin from the PCL-PG-HGF scaffold. The mechanical property of this hybrid scaffold improved after being coated with the PG hydrogel. In addition, the PCL-PG-HGF scaffold illustrated no inflammatory lesions, organ damage, or biological toxicity in all primary organs, with rapid organization of the endothelial cell layer, smooth muscle regeneration, and extracellular matrix formation. These results indicated that the PCL-PG-HGF scaffold is biocompatible and provides a microenvironment in which a tissue-engineered vascular graft with anticoagulant properties allows regeneration of vascular tissue (Scheme 1). Such findings confirm the feasibility of creating hydrogel scaffolds coated with bioactive factors to prepare novel vascular grafts.


Sujet(s)
Matériaux biocompatibles , Facteur de croissance des hépatocytes , Facteur de croissance des hépatocytes/pharmacologie , Préparations à action retardée/pharmacologie , Matériaux biocompatibles/pharmacologie , Polyéthylène glycols/pharmacologie , Hydrogels/pharmacologie , Héparine/pharmacologie
12.
Biochem Biophys Res Commun ; 682: 371-380, 2023 11 19.
Article de Anglais | MEDLINE | ID: mdl-37844446

RÉSUMÉ

The overexpression of hepatic growth factor(HGF) is one of the important reasons for the development of gefitinib resistance in EGFR-sensitive mutant lung adenocarcinoma cells. Targeting the HGF receptor MET through endocytosis inhibition or degradation induction has been proposed as a potential strategy to overcome this resistance. However, the effectiveness of this approach remains needs to be evaluated. In this study, we observed that MET receptors undergo persistent endocytosis but rarely enter the degradation pathway in HGF-overexpressing cells. We showed that MET endocytosis can be inhibited by using gene silence method or MET inhibitors. CHC or DNM2 gene silence slightly increases the sensitivity of resistant cells to gefitinib without affecting MET activity, while GRB2 gene silence can simultaneously inhibit MET endocytosis and reduce MET activity, resulting in a significant reversal effect of gefitinib resistance. Similarly, MET inhibitors significantly reverse drug resistance, accompanied by simultaneous inhibition of MET endocytosis and activity, highlighting the importance of both endocytosis and activity in HGF-induced gefitinib resistance. Additionally, we demonstrated that promoting MET degradation through deubiquitinase (USP8 or USP32) gene silence is another effective method for reversing drug resistance. Overall, our findings suggest that targeting MET receptor endocytosis and degradation is an attractive strategy for overcoming HGF-induced gefitinib resistance in EGFR-sensitive mutant lung adenocarcinoma.


Sujet(s)
Adénocarcinome pulmonaire , Tumeurs du poumon , Humains , Géfitinib/pharmacologie , Tumeurs du poumon/traitement médicamenteux , Tumeurs du poumon/génétique , Tumeurs du poumon/métabolisme , Quinazolines/pharmacologie , Récepteurs ErbB/métabolisme , Facteur de croissance des hépatocytes/génétique , Facteur de croissance des hépatocytes/pharmacologie , Facteur de croissance des hépatocytes/métabolisme , Résistance aux médicaments antinéoplasiques/génétique , Lignée cellulaire tumorale , Adénocarcinome pulmonaire/traitement médicamenteux , Adénocarcinome pulmonaire/génétique , Endocytose , Inhibiteurs de protéines kinases/pharmacologie
13.
Growth Factors ; 41(3): 130-139, 2023 08.
Article de Anglais | MEDLINE | ID: mdl-37398999

RÉSUMÉ

This study aimed to assess the efficacy of hepatocyte growth factor (HGF)-transfected adipose-derived mesenchymal stem cell (ADSC) transplantation in the injured vocal folds (VFs) of canines. A lentiviral vector encoding HGF was successfully produced via Gateway cloning, which was used to infect ADSCs. Four weeks after transoral laser microsurgery (type II) with CO2 laser, the beagles of each group were injected with HGF-transfected ADSCs or uninfected ADSCs into VFs. The results showed that the retention of HGF-transfected ADSCs in the VFs persisted about three months post-injection. The VFs in the HGF-transfected ADSCs group exhibited a closer-to-normal structure with less collagen deposition and higher amounts of hyaluronic acid (HA) in the third month. The short microvilli in the HGF-transfected ADSCs group showed a dense and uniform distribution. These results revealed that HGF-transfected ADSC is a potential treatment option for injured VFs.


Sujet(s)
Transplantation de cellules souches mésenchymateuses , Animaux , Chiens , Transplantation de cellules souches mésenchymateuses/méthodes , Plis vocaux/chirurgie , Facteur de croissance des hépatocytes/génétique , Facteur de croissance des hépatocytes/pharmacologie
14.
Hear Res ; 434: 108786, 2023 07.
Article de Anglais | MEDLINE | ID: mdl-37192594

RÉSUMÉ

Loss of sensory hair cells from exposure to certain licit drugs, such as aminoglycoside antibiotics, can result in permanent hearing damage. Exogenous application of the neurotrophic molecule hepatocyte growth factor (HGF) promotes neuronal cell survival in a variety of contexts, including protecting hair cells from aminoglycoside ototoxicity. HGF itself is not an ideal therapeutic due to a short half-life and limited blood-brain barrier permeability. MM-201 is a chemically stable, blood-brain barrier permeable, synthetic HGF mimetic that serves as a functional ligand to activate the HGF receptor and its downstream signaling cascade. We previously demonstrated that MM-201 robustly protects zebrafish lateral line hair cells from aminoglycoside ototoxicity. Here, we examined the ability of MM-201 to protect mammalian sensory hair cells from aminoglycoside damage to further evaluate MM-201's clinical potential. We found that MM-201 exhibited dose-dependent protection from neomycin and gentamicin ototoxicity in mature mouse utricular explants. MM-201's protection was reduced following inhibition of mTOR, a downstream target of HGF receptor activation, implicating the activation of endogenous intracellular substrates by MM-201 as critical for the observed protection. We then asked if MM-201 altered the bactericidal properties of aminoglycosides. Using either plate or liquid growth assays we found that MM-201 did not alter the bactericidal efficacy of aminoglycoside antibiotics at therapeutically relevant concentrations. We therefore assessed the protective capacity of MM-201 in an in vivo mouse model of kanamycin ototoxicity. In contrast to our in vitro data, MM-201 did not attenuate kanamycin ototoxicity in vivo. Further, we found that MM-201 was ototoxic to mice across the dose range tested here. These data suggest species- and tissue-specific differences in otoprotective capacity. Next generation HGF mimetics are in clinical trials for neurodegenerative diseases and show excellent safety profiles, but neither preclinical studies nor clinical trials have examined hearing loss as a potential consequence of pharmaceutical HGF activation. Further research is needed to determine the consequences of systemic MM-201 application on the auditory system.


Sujet(s)
Aminosides , Ototoxicité , Souris , Animaux , Aminosides/toxicité , Protéines proto-oncogènes c-met/pharmacologie , Danio zébré , Facteur de croissance des hépatocytes/pharmacologie , Antibactériens/toxicité , Mort cellulaire , Kanamycine/toxicité , Mammifères
15.
J Sex Med ; 20(6): 749-755, 2023 05 26.
Article de Anglais | MEDLINE | ID: mdl-37037785

RÉSUMÉ

BACKGROUND: Structural alterations of the penis, including cavernosal apoptosis and fibrosis, induce venous leakage into the corpus cavernosum or cavernosal veno-occlusive dysfunction, a key pathophysiology associated with erectile dysfunction after radical prostatectomy. We hypothesized that the effect of JNK inhibitors on reducing apoptosis and hepatocyte growth factor (HGF) on inducing tissue regeneration could be another treatment mechanism of erectile dysfunction after radical prostatectomy. AIM: To investigate whether JNK inhibition combined with intracavernosal administration of HGF can completely preserve cavernosal veno-occlusive function (CVOF) in a rat model of erectile dysfunction induced via bilateral cavernosal nerve crush injury (CNCI). METHODS: A total of 42 male Sprague-Dawley rats were randomly assigned to sham control (group S), CNCI (group I), and CNCI treated with a combination of JNK inhibitor and HGF (group J + H) for 5 weeks after surgery. OUTCOMES: Rats in each group were evaluated via dynamic infusion cavernosometry (DIC), caspase-3 activity assay, Masson trichrome staining, immunohistochemical staining of α-smooth muscle actin, and immunoblotting at 5 weeks after surgery. RESULTS: Regarding CVOF, group I showed decreased papaverine response, increased maintenance, and drop rates of DIC when compared with group S. Group J + H showed significant improvement in the 3 DIC parameters vs group I. No differences in the 3 DIC parameters were found between group J + H and group S. Regarding the structural integrity of the corpus cavernosum, group I showed increased caspase-3 activity, decreased smooth muscle (SM):collagen ratio, decreased SM content, decreased protein expression of PECAM-1, and decreased phosphorylation of c-Jun and c-Met. Group J + H showed significant attenuation in histologic and molecular derangement as compared with group I. There were no differences in caspase-3 activity, SM content, SM:collagen ratio, PECAM-1 protein expression, c-Jun phosphorylation, and c-Met phosphorylation between groups J + H and S. CLINICAL IMPLICATIONS: Our results suggest that antiapoptotic and regenerative therapy for the corpus cavernosum is a potential mechanism of penile rehabilitation after radical prostatectomy. STRENGTHS AND LIMITATIONS: This study provides evidence that combination treatment of JNK inhibitor and HGF preserves erectile function by restoring corporal SM and endothelium. However, additional human studies are needed to confirm the clinical effect. CONCLUSION: Chronic treatment with JNK inhibitor and HGF may preserve CVOF to levels comparable to sham control by preserving the structural integrity of the corpus cavernosum and so represents a potential therapeutic option for preventing the development of cavernosal veno-occlusive dysfunction.


Sujet(s)
Dysfonctionnement érectile , Traumatismes du système nerveux , Animaux , Humains , Mâle , Rats , Caspase-3 , Modèles animaux de maladie humaine , Dysfonctionnement érectile/traitement médicamenteux , Dysfonctionnement érectile/étiologie , Facteur de croissance des hépatocytes/pharmacologie , Facteur de croissance des hépatocytes/usage thérapeutique , Érection du pénis , Pénis/innervation , Antigènes CD31 , Rat Sprague-Dawley
16.
Pharmacol Res ; 188: 106640, 2023 02.
Article de Anglais | MEDLINE | ID: mdl-36627004

RÉSUMÉ

Inflammation resolution is an active process that involves cellular events such as apoptosis and efferocytosis, which are key steps in the restoration of tissue homeostasis. Hepatocyte growth factor (HGF) is a growth factor mostly produced by mesenchymal-origin cells and has been described to act via MET receptor tyrosine kinase. The HGF/MET axis is essential for determining the progression and severity of inflammatory and immune-mediated disorders. Here, we investigated the effect of blocking the HGF/MET signalling pathway by PF-04217903 on the resolution of established models of neutrophilic inflammation. In a self-resolving model of gout induced by MSU crystals, HGF expression on periarticular tissue peaked at 12 h, the same time point that neutrophils reach their maximal accumulation in the joints. The HGF/MET axis was activated in this model, as demonstrated by increased levels of MET phosphorylation in neutrophils (Ly6G+ cells). In addition, the number of neutrophils was reduced in the knee exudate after PF-04217903 treatment, an effect accompanied by increased neutrophil apoptosis and efferocytosis and enhanced expression of Annexin A1, a key molecule for inflammation resolution. Reduced MPO activity, IL-1ß and CXCL1 levels were also observed in periarticular tissue. Importantly, PF-04217903 reduced the histopathological score and hypernociceptive response. Similar findings were obtained in LPS-induced neutrophilic pleurisy. In human neutrophils, the combined use of LPS and HGF increased MET phosphorylation and provided a prosurvival signal, whereas blocking MET with PF-04217903 induced caspase-dependent neutrophil apoptosis. Taken together, these data demonstrate that blocking HGF/MET signalling may be a potential therapeutic strategy for inducing the resolution of neutrophilic inflammatory responses.


Sujet(s)
Facteur de croissance des hépatocytes , Granulocytes neutrophiles , Humains , Facteur de croissance des hépatocytes/métabolisme , Facteur de croissance des hépatocytes/pharmacologie , Facteur de croissance des hépatocytes/usage thérapeutique , Lipopolysaccharides/pharmacologie , Inflammation/métabolisme , Apoptose , Protéines proto-oncogènes c-met/métabolisme , Homéostasie
17.
Pediatr Surg Int ; 39(1): 80, 2023 Jan 11.
Article de Anglais | MEDLINE | ID: mdl-36631569

RÉSUMÉ

PURPOSE: Nowadays, the standard therapy for patients with short bowel syndrome is parenteral nutrition (PN). Various growth factors have been tested to achieve weaning from prolonged PN administration. We evaluated the effect of hepatocyte growth factor (HGF) on structural intestinal adaptation and cell proliferation in a rat model of SBS. METHODS: Thirty Sprague-Dawley rats were divided into three groups; group A rats (sham) underwent bowel transection, group B rats underwent a 75% bowel resection, and group C rats underwent the same procedure but were treated postoperatively with HGF. Histopathologic parameters of intestinal adaptation were determined, while microarray and rt-PCR analyses of ileal RNA were also performed. RESULTS: Treatment with HGF resulted in significant increase in body weight, while the jejunal and ileal villus height and crypt depth were increased in HGF rats (36%, p < 0.05 and 27%, p < 0.05 respectively). Enterocyte proliferation was also significantly increased in HGF rats (21% p < 0.05). Microarray and quantitative rt-PCR analyses showed that the genes hgfac, rac 1, cdc42, and akt 1 were more than twofold up-regulated after HGF treatment. CONCLUSION: HGF emerges as a growth factor that enhances intestinal adaptation. The future use of HGF may potentially reduce the requirement for PN in SBS patients.


Sujet(s)
Adaptation physiologique , Facteur de croissance des hépatocytes , Syndrome de l'intestin court , Animaux , Rats , Modèles animaux de maladie humaine , Facteur de croissance des hépatocytes/pharmacologie , Facteur de croissance des hépatocytes/usage thérapeutique , Muqueuse intestinale/métabolisme , Intestins/anatomopathologie , Modèles théoriques , Rat Sprague-Dawley , Syndrome de l'intestin court/traitement médicamenteux , Syndrome de l'intestin court/métabolisme
18.
Neurotherapeutics ; 20(2): 431-451, 2023 03.
Article de Anglais | MEDLINE | ID: mdl-36538176

RÉSUMÉ

All types of dementia, including Alzheimer's disease, are debilitating neurodegenerative conditions marked by compromised cognitive function for which there are few effective treatments. Positive modulation of hepatocyte growth factor (HGF)/MET, a critical neurotrophic signaling system, may promote neuronal health and function, thereby addressing neurodegeneration in dementia. Here, we evaluate a series of novel small molecules for their ability to (1) positively modulate HGF/MET activity, (2) induce neurotrophic changes and protect against neurotoxic insults in primary neuron culture, (3) promote anti-inflammatory effects in vitro and in vivo, and (4) reverse cognitive deficits in animal models of dementia. Through screening studies, the compound now known as fosgonimeton-active metabolite (fosgo-AM) was identified by use of immunocytochemistry to be the most potent positive modulator of HGF/MET and was selected for further testing. Primary hippocampal neurons treated with fosgo-AM showed enhanced synaptogenesis and neurite outgrowth, supporting the neurotrophic effects of positive modulators of HGF/MET. Additionally, fosgo-AM protected against neurotoxic insults in primary cortical neuron cultures. In vivo, treatment with fosgo-AM rescued cognitive deficits in the rat scopolamine amnesia model of dementia. Although fosgo-AM demonstrated several procognitive effects in vitro and in vivo, a prodrug strategy was used to enhance the pharmacological properties of fosgo-AM, resulting in the development of fosgonimeton (ATH-1017). The effect of fosgonimeton on cognition was confirmed in a lipopolysaccharide (LPS)-induced neuroinflammatory mouse model of dementia. Together, the results of these studies support the potential of positive modulators of HGF/MET to be used as novel therapeutics and suggest the drug candidate fosgonimeton might protect against neurodegeneration and be therapeutic in the management of Alzheimer's disease and other types of dementia.


Sujet(s)
Maladie d'Alzheimer , Facteur de croissance des hépatocytes , Animaux , Souris , Rats , Maladie d'Alzheimer/traitement médicamenteux , Maladie d'Alzheimer/génétique , Maladie d'Alzheimer/métabolisme , Facteur de croissance des hépatocytes/métabolisme , Facteur de croissance des hépatocytes/pharmacologie , Hippocampe , Neurones/métabolisme , Transduction du signal
19.
Cancer Med ; 12(5): 5809-5820, 2023 03.
Article de Anglais | MEDLINE | ID: mdl-36416133

RÉSUMÉ

BACKGROUND: Entrectinib is an effective drug for treating solid tumors with NTRK gene rearrangement and non-small cell lung cancer (NSCLC) with ROS1 gene rearrangement. However, its efficacy is limited by tolerance and acquired resistance, the mechanisms of which are not fully understood. The growth factors produced by the tumor microenvironment, including hepatocyte growth factor (HGF) produced by tumor-associated fibroblasts, critically affect the sensitivity to targeted drugs. METHODS: We investigated whether growth factors that can be produced by the microenvironment affect sensitivity of NTRK1-rearranged colon cancer KM12SM cells and ROS1-rearranged NSCLC HCC78 cells to entrectinib both in vitro and in vivo. RESULTS: Among the growth factors assessed, HGF most potently induced entrectinib resistance in KM12SM and HCC78 cells by activating its receptor MET. HGF-induced entrectinib resistance was reversed by the active-HGF-specific macrocyclic peptide HiP-8 and the MET kinase inhibitor capmatinib in vitro. In addition, HGF-producing fibroblasts promoted entrectinib resistance in vitro (culture model) and in vivo (subcutaneous tumor model). The use of capmatinib circumvented entrectinib resistance in a subcutaneous tumor model inoculated with KM12SM and HGF-producing fibroblasts. CONCLUSION: Our findings suggest that growth factors in the tumor microenvironment, such as HGF, may induce resistance to entrectinib in tumors with NTRK1 or ROS1 rearrangements. Our results further suggest that optimally co-administering inhibitors of resistance-inducing growth factors may maximize the therapeutic efficacy of entrectinib.


Sujet(s)
Antinéoplasiques , Carcinome pulmonaire non à petites cellules , Tumeurs du poumon , Humains , Antinéoplasiques/usage thérapeutique , Carcinome pulmonaire non à petites cellules/traitement médicamenteux , Carcinome pulmonaire non à petites cellules/génétique , Facteur de croissance des hépatocytes/génétique , Facteur de croissance des hépatocytes/pharmacologie , Tumeurs du poumon/anatomopathologie , Inhibiteurs de protéines kinases/pharmacologie , Inhibiteurs de protéines kinases/usage thérapeutique , Protein-tyrosine kinases/génétique , Protéines proto-oncogènes/génétique , Récepteurs à activité tyrosine kinase , Microenvironnement tumoral
20.
Int J Radiat Oncol Biol Phys ; 116(3): 640-654, 2023 07 01.
Article de Anglais | MEDLINE | ID: mdl-36586496

RÉSUMÉ

PURPOSE: Cancer-associated fibroblasts (CAFs) are an integral part of the tumor microenvironment (TME), which is involved in therapy resistance. This study aimed to investigate the role of CAFs in radiosensitivity of breast cancer cells. METHODS AND MATERIALS: The CAFs were isolated from the breast cancer tissues, and the conditioned medium was collected to culture breast cancer cells. Radiation-induced DNA damage was evaluated by immunofluorescence and western blotting. Cytokine array and enzyme-linked immunosorbent assay were performed to analyze the secretion of cytokines and growth factors. An in vitro clonogenic survival assay and in vivo xenograft mouse model were performed to determine the radiosensitivity of breast cancer cells. Finally, the expression of hepatocyte growth factor (HGF) and c-Met in the breast cancer tissues were detected by immunohistochemistry. RESULTS: The CAFs were found to secrete HGF to activate the c-Met signaling pathway, which induced epithelial-to-mesenchymal transition, growth, and radioresistance of breast cancer cells. Furthermore, radiation was observed to enhance HGF secretion by CAFs and increase c-Met expression in breast cancer cells, which led to HGF/c-Met signaling pathway activation. Moreover, radiation-induced tumor necrosis factor α (TNFα) secretion by breast cancer cells promoted CAF proliferation and HGF secretion. Additionally, HGF and c-Met high expression were associated with worse recurrence-free survival in patients with breast cancer who had received radiation therapy. CONCLUSIONS: The findings revealed that HGF and TNFα are critical for the crosstalk between breast cancer cells and CAFs in the TME and that the HGF/c-Met signaling pathway is a promising therapeutic target for radiosensitizing breast cancer.


Sujet(s)
Tumeurs du sein , Fibroblastes associés au cancer , Humains , Animaux , Souris , Femelle , Facteur de nécrose tumorale alpha/métabolisme , Facteur de croissance des hépatocytes/génétique , Facteur de croissance des hépatocytes/métabolisme , Facteur de croissance des hépatocytes/pharmacologie , Transduction du signal , Protéines proto-oncogènes c-met , Tumeurs du sein/anatomopathologie , Prolifération cellulaire , Lignée cellulaire tumorale , Fibroblastes/métabolisme , Microenvironnement tumoral
SÉLECTION CITATIONS
DÉTAIL DE RECHERCHE