Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 838
Filter
1.
Zhongguo Zhong Yao Za Zhi ; 49(15): 4188-4196, 2024 Aug.
Article in Chinese | MEDLINE | ID: mdl-39307751

ABSTRACT

Based on the hepatocyte growth factor(HGF)/phosphatidylinositol 3-kinase(PI3K)/protein kinase B(Akt) signaling axis, this study investigated the therapeutic effect of Paeoniae Radix Rubra and Aconiti Lateralis Radix Praeparata(PRR-ALRP) her-bal pair on acute-on-chronic liver failure(ACLF) rats and its impact on hepatocellular regeneration. The rat model of ACLF was constructed by subcutaneous and tail vein injection of bovine serum albumin combined with intraperitoneal injection of lipopolysaccharides(LPS)+D-galactosamine(D-GalN). The rats were divided into normal control(NC) group, model(vehicle) group, PRR-ALRP(5.85 g·kg~(-1)) group, and hepatocyte growth factor granules(HGFG, 4.05 g·kg~(-1)) group. Hematoxylin-eosin(HE) staining was used to observe pathological changes in rat liver tissues. Serum alanine aminotransferase(ALT), aspartate transaminase(AST), and total bilirubin(TBIL) were detected using an automatic biochemical analyzer. The levels of interleukin-1ß(IL-1ß) and tumor necrosis factor-α(TNF-α) inflammatory factors were detected by ELISA. Immunofluorescence staining was used to detect the positive expression of proliferating cell nuclear antigen(PCNA), antigen identified by monoclonal antibody(Ki67), and cell cycle protein B1(CyclinB1). Real-time fluorescence-based quantitative polymerase chain reaction(RT-qPCR) and Western blot were used to detect the mRNA and protein expression levels of HGF, growth factor receptor-bound protein 1(Gab1), PI3K, Akt, phosphorylated PI3K(p-PI3K), and phosphorylated Akt(p-Akt). The results showed that compared with the vehicle group, the PRR-ALRP group had reduced liver tissue pathological scores, improved liver function, and reduced inflammatory response, with enhanced PCNA, Ki67, and CyclinB1 fluorescence expression. Furthermore, compared with the model group, the PRR-ALRP group showed upregulated expression of HGF and Gab1 proteins, as well as activation of PI3K and Akt phosphorylation. These findings suggest that PRR-ALRP herbal pair exerts anti-liver failure effects by alleviating hepatocyte inflammatory damage and promoting hepatocellular regeneration, and its specific regulatory mechanism may be related to the activation of the HGF/PI3K/Akt signaling pathway.


Subject(s)
Aconitum , Drugs, Chinese Herbal , Hepatocyte Growth Factor , Paeonia , Phosphatidylinositol 3-Kinases , Proto-Oncogene Proteins c-akt , Rats, Sprague-Dawley , Signal Transduction , Animals , Hepatocyte Growth Factor/genetics , Hepatocyte Growth Factor/metabolism , Rats , Proto-Oncogene Proteins c-akt/metabolism , Proto-Oncogene Proteins c-akt/genetics , Drugs, Chinese Herbal/administration & dosage , Drugs, Chinese Herbal/pharmacology , Signal Transduction/drug effects , Male , Aconitum/chemistry , Phosphatidylinositol 3-Kinases/metabolism , Phosphatidylinositol 3-Kinases/genetics , Paeonia/chemistry , Liver Regeneration/drug effects , Acute-On-Chronic Liver Failure/drug therapy , Acute-On-Chronic Liver Failure/genetics , Acute-On-Chronic Liver Failure/metabolism , Liver/drug effects , Liver/metabolism , Humans , Hepatocytes/drug effects , Hepatocytes/metabolism
2.
Aging Cell ; 23(10): e14337, 2024 Oct.
Article in English | MEDLINE | ID: mdl-39297318

ABSTRACT

In response to peroxynitrite (ONOO-) generation, myogenic stem satellite cell activator HGF (hepatocyte growth factor) undergoes nitration of tyrosine residues (Y198 and Y250) predominantly on fast IIa and IIx myofibers to lose its binding to the signaling receptor c-met, thereby disturbing muscle homeostasis during aging. Here we show that rat anti-HGF monoclonal antibody (mAb) 1H41C10, which was raised in-house against a synthetic peptide FTSNPEVRnitroY198EV, a site well-conserved in mammals, functions to confer resistance to nitration dysfunction on HGF. 1H41C10 was characterized by recognizing both nitrated and non-nitrated HGF with different affinities as revealed by Western blotting, indicating that the paratope of 1H41C10 may bind to the immediate vicinity of Y198. Subsequent experiments showed that 1H41C10-bound HGF resists peroxynitrite-induced nitration of Y198. A companion mAb-1H42F4 presented similar immuno-reactivity, but did not protect Y198 nitration, and thus served as the control. Importantly, 1H41C10-HGF also withstood Y250 nitration to retain c-met binding and satellite cell activation functions in culture. The Fab region of 1H41C10 exerts resistivity to Y250 nitration possibly due to its localization in the immediate vicinity to Y250, as supported by an additional set of experiments showing that the 1H41C10-Fab confers Y250-nitration resistance which the Fc segment does not. Findings highlight the in vitro preventive impact of 1H41C10 on HGF nitration-dysfunction that strongly impairs myogenic stem cell dynamics, potentially pioneering cogent strategies for counteracting or treating age-related muscle atrophy with fibrosis (including sarcopenia and frailty) and the therapeutic application of investigational HGF drugs.


Subject(s)
Hepatocyte Growth Factor , Muscular Atrophy , Animals , Hepatocyte Growth Factor/metabolism , Muscular Atrophy/metabolism , Rats , Aging , Mice , Peroxynitrous Acid/metabolism , Peroxynitrous Acid/pharmacology , Satellite Cells, Skeletal Muscle/metabolism , Humans
3.
Front Cell Dev Biol ; 12: 1462841, 2024.
Article in English | MEDLINE | ID: mdl-39291265

ABSTRACT

Bronchopulmonary dysplasia (BPD) is fundamentally characterized by the arrest of lung development and abnormal repair mechanisms, which result in impaired development of the alveoli and microvasculature. Hepatocyte growth factor (HGF), secreted by pulmonary mesenchymal and endothelial cells, plays a pivotal role in the promotion of epithelial and endothelial cell proliferation, branching morphogenesis, angiogenesis, and alveolarization. HGF exerts its beneficial effects on pulmonary vascular development and alveolar simplification primarily through two pivotal pathways: the stimulation of neovascularization, thereby enriching the pulmonary microvascular network, and the inhibition of the epithelial-mesenchymal transition (EMT), which is crucial for maintaining the integrity of the alveolar structure. We discuss HGF and its receptor c-Met, interact with various growth factors throughout the process of lung development and BPD, and form a signaling network with HGF as a hub, which plays the pivotal role in orchestrating and integrating epithelial, endothelial and mesenchymal.

4.
Stem Cells ; 2024 Sep 12.
Article in English | MEDLINE | ID: mdl-39269318

ABSTRACT

Transplanted mesenchymal stem cells (MSCs) can significantly aid in repairing spinal cord injuries (SCI) by migrating to and settling at the injury site. However, this process is typically inefficient, as only a small fraction of MSCs successfully reach the target lesion area. During SCI, the increased expression and secretion of hepatocyte growth factor (HGF) act as a chemoattractant that guides MSC migration. Nonetheless, the precise mechanisms by which HGF influences MSC migration are not fully understood. This study focused on unraveling the molecular pathways that drive MSC migration towards the SCI site in response to HGF. It was found that HGF can activate ß-catenin signaling in MSCs either by phosphorylating LRP6 or by suppressing GSK3ß phosphorylation through the AKT and ERK1/2 pathways, or by enhancing the expression and nuclear translocation of TCF4. This activation leads to elevated Nedd9 expression, which promotes focal adhesion formation and F-actin polymerization, facilitating chemotactic migration. Transplanting MSCs during peak HGF expression in injured tissues substantially improves nerve regeneration, reduces scarring, and enhances hind limb mobility. Additionally, prolonging HGF release can further boost MSC migration and engraftment, thereby amplifying regenerative outcomes. However, inhibiting HGF/Met or interfering with ß-catenin or Nedd9 signaling significantly impairs MSC engraftment, obstructing tissue repair and functional recovery. Together, these findings provide a theoretical basis and practical strategy for MSC transplantation therapy in SCI, highlighting the specific molecular mechanisms by which HGF regulates ß-catenin signaling in MSCs, ultimately triggering their chemotactic migration.

5.
Int J Mol Sci ; 25(16)2024 Aug 22.
Article in English | MEDLINE | ID: mdl-39201787

ABSTRACT

Non-small cell lung cancer (NSCLC) is characterized by several molecular alterations that contribute to its development and progression. These alterations include the epidermal growth factor receptor (EGFR), anaplastic lymphoma kinase (ALK), human epidermal growth factor receptor 2 (HER2), and mesenchymal-epithelial transition factor (c-MET). Among these, the hepatocyte growth factor (HGF)/c-MET signaling pathway plays a crucial role in NSCLC. In spite of this, the involvement of the HGF/c-MET signaling axis in remodeling the tumor microenvironment (TME) remains relatively unexplored. This review explores the biological functions of the HGF/c-MET signaling pathway in both normal and cancerous cells, examining its multifaceted roles in the NSCLC tumor microenvironment, including tumor cell proliferation, migration and invasion, angiogenesis, and immune evasion. Furthermore, we summarize the current progress and clinical applications of MET-targeted therapies in NSCLC and discuss future research directions, such as the development of novel MET inhibitors and the potential of combination immunotherapy.


Subject(s)
Carcinoma, Non-Small-Cell Lung , Hepatocyte Growth Factor , Lung Neoplasms , Proto-Oncogene Proteins c-met , Signal Transduction , Tumor Microenvironment , Humans , Carcinoma, Non-Small-Cell Lung/metabolism , Carcinoma, Non-Small-Cell Lung/pathology , Proto-Oncogene Proteins c-met/metabolism , Hepatocyte Growth Factor/metabolism , Lung Neoplasms/metabolism , Lung Neoplasms/pathology , Animals
6.
Int J Mol Sci ; 25(15)2024 Aug 04.
Article in English | MEDLINE | ID: mdl-39126076

ABSTRACT

Alcoholic liver disease (ALD) is a form of hepatic inflammation. ALD is mediated by gut leakiness. This study evaluates the anti-inflammatory effects of ASCs overexpressing interferon-beta (ASC-IFN-ß) on binge alcohol-induced liver injury and intestinal permeability. In vitro, ASCs were transfected with a non-viral vector carrying the human IFN-ß gene, which promoted hepatocyte growth factor (HGF) secretion in the cells. To assess the potential effects of ASC-IFN-ß, C57BL/6 mice were treated with three oral doses of binge alcohol and were administered intraperitoneal injections of ASC-IFN-ß. Mice treated with binge alcohol and administered ASC-IFN-ß showed reduced liver injury and inflammation compared to those administered a control ASC. Analysis of intestinal tissue from ethanol-treated mice administered ASC-IFN-ß also indicated decreased inflammation. Additionally, fecal albumin, blood endotoxin, and bacterial colony levels were reduced, indicating less gut leakiness in the binge alcohol-exposed mice. Treatment with HGF, but not IFN-ß or TRAIL, mitigated the ethanol-induced down-regulation of cell death and permeability in Caco-2 cells. These results demonstrate that ASCs transfected with a non-viral vector to induce IFN-ß overexpression have protective effects against binge alcohol-mediated liver injury and gut leakiness via HGF.


Subject(s)
Ethanol , Interferon-beta , Liver Diseases, Alcoholic , Mesenchymal Stem Cells , Mice, Inbred C57BL , Permeability , Animals , Humans , Interferon-beta/metabolism , Liver Diseases, Alcoholic/metabolism , Liver Diseases, Alcoholic/pathology , Liver Diseases, Alcoholic/genetics , Mice , Mesenchymal Stem Cells/metabolism , Ethanol/adverse effects , Caco-2 Cells , Hepatocyte Growth Factor/metabolism , Hepatocyte Growth Factor/genetics , Male , Adipose Tissue/metabolism , Liver/metabolism , Liver/pathology , Intestinal Mucosa/metabolism , Intestinal Mucosa/drug effects , Intestinal Mucosa/pathology
7.
Biomedicines ; 12(8)2024 Aug 20.
Article in English | MEDLINE | ID: mdl-39200370

ABSTRACT

Hereditary neurodegenerative diseases (hNDDs) such as Alzheimer's, Parkinson's, Huntington's disease, and others are primarily characterized by their progressive nature, severely compromising both the cognitive and motor abilities of patients. The underlying genetic component in hNDDs contributes to disease risk, creating a complex genetic landscape. Considering the fact that growth factors play crucial roles in regulating cellular processes, such as proliferation, differentiation, and survival, they could have therapeutic potential for hNDDs, provided appropriate dosing and safe delivery approaches are ensured. This article presents a detailed overview of growth factors, and explores their therapeutic potential in treating hNDDs, emphasizing their roles in neuronal survival, growth, and synaptic plasticity. However, challenges such as proper dosing, delivery methods, and patient variability can hinder their clinical application.

8.
Int J Mol Sci ; 25(13)2024 Jul 04.
Article in English | MEDLINE | ID: mdl-39000441

ABSTRACT

Although inhibitors targeting the PD1/PD-L1 immune checkpoint are showing comparably good outcomes, a significant percentage of head and neck squamous cell carcinoma (HNSCC) patients do not respond to treatment. Apart from using different treatment strategies, another possibility would be to target other immune checkpoints operating in these non-responding tumors. To obtain an overview of which checkpoint ligands are expressed on HNSCC tumor cells and if these ligands are affected by HGF/MET signaling, we used mRNA sequencing and antibody-based techniques for identifying checkpoint ligands in six HNSCC tumor cell lines. Furthermore, we compared our results to mRNA sequencing data. From the checkpoint ligands we investigated, VISTA was expressed the highest at the RNA level and was also the most ubiquitously expressed. PD-L2 and B7-H3 were expressed comparably lower and were not present in all cell lines to the same extent. B7-H4, however, was only detectable in the Detroit 562 cell line. Concerning the effect of HGF on the ligand levels, PD-L2 expression was enhanced with HGF stimulation, whereas other checkpoint ligand levels decreased with stimulation. B7-H4 levels in the Detroit 562 cell line drastically decreased with HGF stimulation. This is of interest because both the checkpoint ligand and the growth factor are reported to be connected to epithelial-mesenchymal transition in the literature.


Subject(s)
Gene Expression Regulation, Neoplastic , Head and Neck Neoplasms , Hepatocyte Growth Factor , Immune Checkpoint Proteins , Proto-Oncogene Proteins c-met , Signal Transduction , Squamous Cell Carcinoma of Head and Neck , Humans , Proto-Oncogene Proteins c-met/metabolism , Proto-Oncogene Proteins c-met/genetics , Squamous Cell Carcinoma of Head and Neck/metabolism , Squamous Cell Carcinoma of Head and Neck/genetics , Squamous Cell Carcinoma of Head and Neck/pathology , Squamous Cell Carcinoma of Head and Neck/immunology , Hepatocyte Growth Factor/metabolism , Hepatocyte Growth Factor/genetics , Cell Line, Tumor , Immune Checkpoint Proteins/metabolism , Immune Checkpoint Proteins/genetics , Head and Neck Neoplasms/metabolism , Head and Neck Neoplasms/genetics , Head and Neck Neoplasms/pathology , Programmed Cell Death 1 Ligand 2 Protein/metabolism , Programmed Cell Death 1 Ligand 2 Protein/genetics , B7-H1 Antigen/metabolism , B7-H1 Antigen/genetics , B7 Antigens/metabolism , B7 Antigens/genetics
9.
Adv Sci (Weinh) ; 11(36): e2404882, 2024 Sep.
Article in English | MEDLINE | ID: mdl-39049677

ABSTRACT

The limited therapeutic strategies available for stroke leave many patients disabled for life. This study assessed the potential of programmed death-ligand 1 (PD-L1) and hepatocyte growth factor (HGF)-engineered mesenchymal stem cell-derived exosomes (EXO-PD-L1-HGF) in enhancing neurological recovery post-stroke. EXO-PD-L1-HGF, which efficiently endocytosed into target cells, significantly diminishes the H2O2-induced neurotoxicity and increased the antiapoptotic proteins in vitro. EXO-PD-L1-HGF attenuates inflammation by inhibiting T-cell proliferation and increasing the number of CD8+CD122+IL-10+ regulatory T cells. Intravenous injection of EXO-PD-L1-HGF could target stromal cell-derived factor-1α (SDF-1α+) cells over the peri-infarcted area of the ischemic brain through CXCR4 upregulation and accumulation in neuroglial cells post-stroke. EXO-PD-L1-HGF facilitates endogenous nestin+ neural progenitor cell (NPC)-induced neurogenesis via STAT3-FOXO3 signaling cascade, which plays a pivotal role in cell survival and neuroprotection, thereby mitigating infarct size and enhancing neurological recovery in a murine stroke model. Moreover, increasing populations of the immune-regulatory CD19+IL-10+ and CD8+CD122+IL-10+ cells, together with reducing populations of proinflammatory cells, created an anti-inflammatory microenvironment in the ischemic brain. Thus, innovative approaches employing EXO-PD-L1-HGF intervention, which targets SDF-1α+ expression, modulates the immune system, and enhances the activation of resident nestin+ NPCs, might significantly alter the brain microenvironment and create a niche conducive to inducing neuroplastic regeneration post-stroke.


Subject(s)
B7-H1 Antigen , Disease Models, Animal , Exosomes , Forkhead Box Protein O3 , Hepatocyte Growth Factor , Mesenchymal Stem Cells , Neuronal Plasticity , STAT3 Transcription Factor , Signal Transduction , Stroke , Animals , Mice , Exosomes/metabolism , Mesenchymal Stem Cells/metabolism , B7-H1 Antigen/metabolism , Stroke/metabolism , Forkhead Box Protein O3/metabolism , Forkhead Box Protein O3/genetics , STAT3 Transcription Factor/metabolism , Hepatocyte Growth Factor/metabolism , Hepatocyte Growth Factor/genetics , Male , Mice, Inbred C57BL
10.
Cell Signal ; 121: 111295, 2024 Sep.
Article in English | MEDLINE | ID: mdl-38996955

ABSTRACT

Calpain2 is a conventional member of the non-lysosomal calpain protease family that has been shown to affect the dynamics of focal and cell-cell adhesions by proteolyzing the components of adhesion complexes. Here, we inactivated calpain2 using CRISPR/Cas9 in epithelial MDCK cells. We show that depletion of calpain2 has multiple effects on cell morphology and function. Calpain2-depleted cells develop epithelial shape, however, they cover a smaller area, and cell clusters are more compact. Inactivation of calpain2 enhanced restoration of transepithelial electrical resistance after calcium switch, decreased cell migration, and delayed cell scattering induced by HGF/SF. In addition, calpain2 depletion prevented morphological changes induced by ERK2 overexpression. Interestingly, proteolysis of several calpain2 targets, including E-cadherin, ß-catenin, talin, FAK, and paxillin, was not discernibly affected by calpain2 depletion. Taken together, these data suggest that calpain2 regulates the stability of cell-cell and cell-substratum adhesions indirectly without affecting the proteolysis of these adhesion complexes.


Subject(s)
Calpain , Cell Adhesion , Epithelial Cells , Animals , Dogs , beta Catenin/metabolism , Cadherins/metabolism , Calcium/metabolism , Calpain/metabolism , Cell Movement , CRISPR-Cas Systems , Epithelial Cells/metabolism , Epithelial Cells/cytology , Hepatocyte Growth Factor/metabolism , Madin Darby Canine Kidney Cells , Mitogen-Activated Protein Kinase 1/metabolism , Proteolysis
11.
Biomed Pharmacother ; 177: 117039, 2024 Aug.
Article in English | MEDLINE | ID: mdl-38955085

ABSTRACT

T-cell acute lymphoblastic leukemia (T-ALL) is a malignant hematological disorder characterized by an increased proliferation of immature T lymphocytes precursors. T-ALL treatment includes chemotherapy with strong side effects, and patients that undergo relapse display poor prognosis. Although cell-intrinsic oncogenic pathways are well-studied, the tumor microenvironment, like inflammatory cellular and molecular components is less explored in T-ALL. We sought to determine the composition of the inflammatory microenvironment induced by T-ALL, and its role in T-ALL progression. We show in two mouse T-ALL cell models that T-ALLs enhance blood neutrophils and resident monocytes, accompanied with a plasmatic acute secretion of inflammatory molecules. Depleting neutrophils using anti-Ly6G treatment or resident monocytes by clodronate liposomes treatment does not modulate plasmatic inflammatory molecule secretion and mice survival. However, inhibiting the secretion of inflammatory molecules by microenvironment with NECA, an agonist of adenosine receptors, diminishes T-ALL progression enhancing mouse survival. We uncovered Hepatocyte Growth Factor (HGF), T-ALL-driven and the most decreased molecule with NECA, as a potential therapeutic target in T-ALL. Altogether, we identified a signature of inflammatory molecules that can potentially be involved in T-ALL evolution and uncovered HGF/cMET pathway as important to target for limiting T-ALL progression.


Subject(s)
Disease Progression , Hepatocyte Growth Factor , Precursor T-Cell Lymphoblastic Leukemia-Lymphoma , Tumor Microenvironment , Animals , Hepatocyte Growth Factor/metabolism , Precursor T-Cell Lymphoblastic Leukemia-Lymphoma/pathology , Precursor T-Cell Lymphoblastic Leukemia-Lymphoma/drug therapy , Precursor T-Cell Lymphoblastic Leukemia-Lymphoma/metabolism , Mice , Mice, Inbred C57BL , Cell Line, Tumor , Inflammation/pathology , Inflammation/drug therapy , Inflammation/metabolism , Inflammation Mediators/metabolism , Neutrophils/metabolism , Neutrophils/drug effects , Monocytes/drug effects , Monocytes/metabolism , Monocytes/pathology
12.
Sci Rep ; 14(1): 14892, 2024 06 28.
Article in English | MEDLINE | ID: mdl-38937503

ABSTRACT

Accurate screening of COVID-19 infection status for symptomatic patients is a critical public health task. Although molecular and antigen tests now exist for COVID-19, in resource-limited settings, screening tests are often not available. Furthermore, during the early stages of the pandemic tests were not available in any capacity. We utilized an automated machine learning (ML) approach to train and evaluate thousands of models on a clinical dataset consisting of commonly available clinical and laboratory data, along with cytokine profiles for patients (n = 150). These models were then further tested for generalizability on an out-of-sample secondary dataset (n = 120). We were able to develop a ML model for rapid and reliable screening of patients as COVID-19 positive or negative using three approaches: commonly available clinical and laboratory data, a cytokine profile, and a combination of the common data and cytokine profile. Of the tens of thousands of models automatically tested for the three approaches, all three approaches demonstrated > 92% sensitivity and > 88 specificity while our highest performing model achieved 95.6% sensitivity and 98.1% specificity. These models represent a potential effective deployable solution for COVID-19 status classification for symptomatic patients in resource-limited settings and provide proof-of-concept for rapid development of screening tools for novel emerging infectious diseases.


Subject(s)
COVID-19 , Cytokines , Machine Learning , Humans , COVID-19/diagnosis , Cytokines/blood , SARS-CoV-2/isolation & purification , SARS-CoV-2/immunology , Mass Screening/methods , Male , Female , Sensitivity and Specificity , Middle Aged , Adult , Aged
13.
Gels ; 10(6)2024 Jun 16.
Article in English | MEDLINE | ID: mdl-38920948

ABSTRACT

A novel scaffold design has been created to enhance tissue engineering and regenerative medicine by optimizing the controlled, prolonged release of Hepatocyte Growth Factor (HGF), a powerful chemoattractant for endogenous mesenchymal stem cells. We present a new stacked scaffold that is made up of three different fibrin gel layers, each of which has HGF integrated into the matrix. The design attempts to preserve HGF's regenerative properties for long periods of time, which is necessary for complex tissue regeneration. These multi-layered fibrin gels have been mechanically evaluated using rheometry, and their degradation behavior has been studied using D-Dimer ELISA. Understanding the kinetics of HGF release from this novel scaffold configuration is essential for understanding HGF's long-term sustained bioactivity. A range of cell-based tests were carried out to verify the functionality of HGF following extended incorporation. These tests included 2-photon microscopy using phalloidin staining to examine cellular morphology, SEM analysis for scaffold-cell interactions, and scratch and scatter assays to assess migration and motility. The analyses show that the novel stacking scaffold promotes vital cellular processes for tissue regeneration in addition to supporting HGF's bioactivity. This scaffold design was developed for in situ tissue engineering. Using the body as a bioreactor, the scaffold should recruit mesenchymal stem cells from their niche, thus combining the regenerative abilities of HGF and MSCs to promote tissue remodeling and wound repair.

14.
Curr Mol Med ; 2024 Jun 24.
Article in English | MEDLINE | ID: mdl-38918984

ABSTRACT

BACKGROUND: Restructuring of dermal microcapillaries is one of the hallmarks of plaque psoriasis. To control the proliferation of vascular endothelial cells, vascular endothelial growth factor (VEGF) promotes the remodeling of the existing blood vessels and angiogenesis. OBJECTIVE: This study aimed to explain the lowering protein and mRNA levels of VEGF in lesional skin of patients with severe psoriasis (the Psoriasis Area and Severity Index, PASI > 25). METHODS: Using the method of qPCR, we assessed the expression of VEGF mRNA in lesional and nonlesional psoriatic skin. Using ELISA, we also compared the levels of VEGF in skin homogenates of psoriasis patients and healthy volunteers. RESULTS: We found that the exacerbation of psoriasis induced VEGF on mRNA and protein levels 12 and 20 times, respectively. We also confirmed a strong correlation between VEGF and PASI score in patients with PASI < 25. In addition, we showed that several factors, namely HGF, HNRPD, and sFLT1 interfere with the biosynthesis of VEGF in skin lesions of patients with PASI > 25%. CONCLUSION: Thus, using VEGF as a biomarker to monitor the disease shall be done cautiously in patients with severe psoriasis.

15.
Cancer Manag Res ; 16: 431-444, 2024.
Article in English | MEDLINE | ID: mdl-38751848

ABSTRACT

Purpose: Pancreatic ductal adenocarcinoma (PDAC) is extremely malignant and rapidly progresses. The overall response rate of PDAC to current treatment methods is still unsatisfactory. Thus, identifying novel targets and clarifying the underlying mechanisms associated with PDAC progression may potentially offer additional treatment strategies. AHNAK2 is aberrantly expressed in a variety of tumors and exerts pro-tumorigenic effects. However, the biological role of AHNAK2 in PDAC remains poorly understood. Methods: The expression of AHNAK2 in PDAC and paired non-tumor tissues was detected by immunohistochemistry (IHC) and quantitative real-time polymerase chain reaction (qRT-PCR). Lentivirus knockdown was performed to investigate the impact of AHNAK2 on the biological function of pancreatic cancer cells. The subcutaneous cell-derived xenograft (CDX) model and the KPC spontaneous mouse model with AHNAK2 silencing were used to observe the effects of AHNAK2 on tumor growth and prognosis. The expression of c-MET at protein level in response to HGF treatment was assessed using western blot. Results: Our results demonstrated that AHNAK2 was highly expressed in PDAC clinical samples and associated with poor prognosis. Knockdown of AHNAK2 significantly inhibited the proliferation, migration, and invasion of pancreatic cancer cells. AHNAK2 knockdown or knockout resulted in tumor growth suppression and prolonged survival in mice with PDAC. In addition, AHNAK2 and c-MET expression levels showed a significant positive correlation at the post-transcriptional level. Mechanistically, AHNAK2 promoted tumor progression by preventing c-MET degradation and persistently activating the HGF/c-MET signaling pathway. Conclusion: Overall, our study revealed that AHNAK2 plays an important role in PDAC progression by modulating the c-MET signaling pathway, and targeting AHNAK2 may be an effective therapeutic strategy for PDAC.

16.
Heliyon ; 10(9): e30520, 2024 May 15.
Article in English | MEDLINE | ID: mdl-38756586

ABSTRACT

Persistent HGF/Met signaling drives tumor growth and dissemination. Proteoglycans within the tumor microenvironment might control HGF availability and signaling by affecting its accessibility to Met (HGF receptor), likely defining whether acute or sustained HGF/Met signaling cues take place. Given that betaglycan (BG, also known as type III TGFß receptor or TGFBR3), a multi-faceted proteoglycan TGFß co-receptor, can be found within the tumor microenvironment, we addressed its hypothetical role in oncogenic HGF signaling. We found that HGF/Met promotes lung cancer and endothelial cells migration via PI3K and mTOR. This effect was enhanced by recombinant soluble betaglycan (solBG) via a mechanism attributable to its glycosaminoglycan chains, as a mutant without them did not modulate HGF effects. Moreover, soluble betaglycan extended the effect of HGF-induced phosphorylation of Met, Akt, and Erk, and membrane recruitment of the RhoGEF P-Rex1. Data-mining analysis of lung cancer patient datasets revealed a significant correlation between high MET receptor, HGF, and PREX1 expression and reduced patient survival. Soluble betaglycan showed biochemical interaction with HGF and, together, they increased tumor growth in immunocompetent mice. In conclusion, the oncogenic properties of the HGF/Met pathway are enhanced and sustained by GAG-containing soluble betaglycan.

17.
Int J Mol Sci ; 25(10)2024 May 17.
Article in English | MEDLINE | ID: mdl-38791500

ABSTRACT

Lymphedema is a disorder that leads to excessive swelling due to lymphatic insufficiency, resulting in the accumulation of protein-rich interstitial fluid. Primary lymphedema predominantly impacts the lower extremities and is frequently linked to hereditary factors. This condition is known to be associated with variants in several genes, such as FOXC2, FLT4, and SOX18. However, many cases remain unexplained, suggesting undiscovered gene associations. This study describes a novel mutation in the hepatocyte growth factor (HGF) gene, a previously hypothesized candidate for lymphedema pathogenesis. This mutation was identified in affected members of a multigenerational family presenting with primary leg lymphedema, consistent with an autosomal dominant inheritance pattern.


Subject(s)
Hepatocyte Growth Factor , Lymphedema , Mutation , Adult , Female , Humans , Male , Middle Aged , Hepatocyte Growth Factor/genetics , Hepatocyte Growth Factor/metabolism , Lymphedema/genetics , Pedigree
18.
Phytomedicine ; 129: 155630, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38678952

ABSTRACT

BACKGROUND: Premature ovarian insufficiency (POI) is a tricky puzzle in the field of female reproductive medicine. Bushen Huoxue recipe (BHR), a traditional Chinese medicine compound based on the combination of kidney-tonifying and blood-activating functions, has shown excellent efficacy in improving female irregular menstruation, POI, and infertility. However, the potential mechanism of BHR in POI treatment has not yet been elucidated. Bone marrow mesenchymal stem cells (BMSCs), a type of pluripotent stem cells, have received increasing attention for their significant role in improving ovarian function and restoring fertility in women with POI. PURPOSE: This study aimed to evaluate the therapeutic effect of BHR in POI mice and explore its potential mechanism. METHODS: A POI mouse model was established with a single intraperitoneal injection of 120 mg/kg cyclophosphamide (CTX). Distilled water, BHR, or dehydroepiandrosterone was administered via gavage for 28 consecutive days. The effect of BHR on ovarian function in POI mice was evaluated by assessing the estrous cycle, ovarian morphology, follicular development, hormone levels, and angiogenesis. The proportion of BMSCs in bone marrow, peripheral blood, and ovary was analyzed via flow cytometry, and the level of molecules mediating migration and homing in ovary was measured. Cell viability assays, scratch healing assays and transwell migration assays were performed to explore the effect of BHR on BMSCs proliferation and migration in vitro, and its potential mechanism was explored. RESULTS: BHR significantly ameliorated estrous cycle disorders, hormone disorders, ovarian morphology, ovarian microvascular formation, and ovarian reserve in POI mice. Meanwhile, the number of BMSCs number in the bone marrow, peripheral blood, and ovary was apparently increased. Of note, BHR increased the level of hepatocyte growth factor (HGF)/cellular mesenchymal epithelial transition factor (cMET) and stromal cell-derived factor-1(SDF-1)/CXC chemokine receptor 4 (CXCR4) in the ovaries of POI mice. Moreover, BHR treatment promoted BMSCs proliferation and migration in vitro, with a significant increase in the level of proliferating cell nuclear antigen, cMET, and CXCR4. CONCLUSIONS: BHR effectively restored ovarian reserve, ovarian function, and ovarian angiogenesis in CTX-induced POI mice. In addition, BHR promoted BMSCs proliferation, migration, and homing to the ovary, which was mediated by the SDF-1/CXCR4 and HGF/cMET signaling axis. Finally, the amelioration of ovarian reserve and ovarian function in CTX-induced POI mice by BHR may be related to its promotion of endogenous BMSCs proliferation and homing.


Subject(s)
Cell Proliferation , Disease Models, Animal , Drugs, Chinese Herbal , Mesenchymal Stem Cells , Ovary , Primary Ovarian Insufficiency , Animals , Female , Drugs, Chinese Herbal/pharmacology , Primary Ovarian Insufficiency/drug therapy , Mesenchymal Stem Cells/drug effects , Ovary/drug effects , Cell Proliferation/drug effects , Mice , Cyclophosphamide , Estrous Cycle/drug effects , Cell Movement/drug effects
19.
Biomed Pharmacother ; 175: 116621, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38677244

ABSTRACT

BACKGROUND: Extracellular neutrophil extracellular traps (NETs) play an important role in acute lung injury (ALI), but their mechanisms are still unclear. The aim of this study is to explore the effects of NETs on endothelial glycocalyx/HGF/cMET pathway and ferroptosis in ALI and elucidate their potential mechanisms. METHODS: Plasma was collected from healthy and sepsis patients to test for differences in neutrophil elastase (NE) expression of NETs components. In addition, LPS-ALI mice and endothelial cell injury models were established, and NETs were disrupted by siPAD4 (a driver gene for NETs) and sivelestat (an inhibitor of the NETs component) in the mice and by sivelestat in the endothelial cell injury models, and the effects of NETs on the SDC-1/HS/HGF/cMET pathway were studied. To verify the relationship between NETs and ferroptosis, Fer1, a ferroptosis inhibitor, was added as a positive control to observe the effect of NETs on ferroptosis indicators. RESULTS: The expression level of NE was significantly higher in the plasma of sepsis patients. In ALI mice, intervention in the generation of NETs reduced pulmonary vascular permeability, protected the integrity of SDC-1/HS and promoted the downstream HGF/cMET pathway. In addition, sivelestat also improved the survival rate of mice, decreased the serious degree of ferroptosis. In the endothelial cells, the results were consistent with those of the ALI mice. CONCLUSION: The study indicates that inhibiting the production of NETs can protect the normal conduction of the SDC-1/HS/HGF/cMET signalling pathway and reduce the severity of ferroptosis.


Subject(s)
Acute Lung Injury , Endothelial Cells , Extracellular Traps , Ferroptosis , Lipopolysaccharides , Mice, Inbred C57BL , Signal Transduction , Syndecan-1 , Animals , Ferroptosis/drug effects , Ferroptosis/physiology , Humans , Lipopolysaccharides/pharmacology , Male , Mice , Acute Lung Injury/pathology , Acute Lung Injury/metabolism , Endothelial Cells/metabolism , Endothelial Cells/drug effects , Endothelial Cells/pathology , Extracellular Traps/metabolism , Extracellular Traps/drug effects , Signal Transduction/drug effects , Syndecan-1/metabolism , Sepsis/metabolism , Sepsis/pathology , Female , Middle Aged , Human Umbilical Vein Endothelial Cells/metabolism , Human Umbilical Vein Endothelial Cells/drug effects , Disease Models, Animal
20.
Neurotherapeutics ; 21(4): e00350, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38599894

ABSTRACT

Positive modulation of hepatocyte growth factor (HGF) signaling may represent a promising therapeutic strategy for Alzheimer's disease (AD) based on its multimodal neurotrophic, neuroprotective, and anti-inflammatory effects addressing the complex pathophysiology of neurodegeneration. Fosgonimeton is a small-molecule positive modulator of the HGF system that has demonstrated neurotrophic and pro-cognitive effects in preclinical models of dementia. Herein, we evaluate the neuroprotective potential of fosgonimeton, or its active metabolite, fosgo-AM, in amyloid-beta (Aß)-driven preclinical models of AD, providing mechanistic insight into its mode of action. In primary rat cortical neurons challenged with Aß (Aß1-42), fosgo-AM treatment significantly improved neuronal survival, protected neurite networks, and reduced tau hyperphosphorylation. Interrogation of intracellular events indicated that cortical neurons treated with fosgo-AM exhibited a significant decrease in mitochondrial oxidative stress and cytochrome c release. Following Aß injury, fosgo-AM significantly enhanced activation of pro-survival effectors ERK and AKT, and reduced activity of GSK3ß, one of the main kinases involved in tau hyperphosphorylation. Fosgo-AM also mitigated Aß-induced deficits in Unc-like kinase 1 (ULK1) and Beclin-1, suggesting a potential effect on autophagy. Treatment with fosgo-AM protected cortical neurons from glutamate excitotoxicity, and such effects were abolished in the presence of an AKT or MEK/ERK inhibitor. In vivo, fosgonimeton administration led to functional improvement in an intracerebroventricular Aß25-35 rat model of AD, as it significantly rescued cognitive function in the passive avoidance test. Together, our data demonstrate the ability of fosgonimeton to counteract mechanisms of Aß-induced toxicity. Fosgonimeton is currently in clinical trials for mild-to-moderate AD (NCT04488419; NCT04886063).


Subject(s)
Alzheimer Disease , Amyloid beta-Peptides , Neurons , Neuroprotective Agents , Animals , Amyloid beta-Peptides/toxicity , Amyloid beta-Peptides/metabolism , Alzheimer Disease/drug therapy , Alzheimer Disease/metabolism , Rats , Neuroprotective Agents/pharmacology , Neuroprotective Agents/therapeutic use , Neurons/drug effects , Neurons/metabolism , Peptide Fragments/toxicity , Disease Models, Animal , Rats, Sprague-Dawley , Cells, Cultured , Oxidative Stress/drug effects , Oxidative Stress/physiology , Cerebral Cortex/drug effects , Cerebral Cortex/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL