Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 3.195
Filter
1.
Proc Natl Acad Sci U S A ; 121(20): e2312855121, 2024 May 14.
Article in English | MEDLINE | ID: mdl-38713626

ABSTRACT

The immune landscape of bladder cancer progression is not fully understood, and effective therapies are lacking in advanced bladder cancer. Here, we visualized that bladder cancer cells recruited neutrophils by secreting interleukin-8 (IL-8); in turn, neutrophils played dual functions in bladder cancer, including hepatocyte growth factor (HGF) release and CCL3highPD-L1high super-immunosuppressive subset formation. Mechanistically, c-Fos was identified as the mediator of HGF up-regulating IL-8 transcription in bladder cancer cells, which was central to the positive feedback of neutrophil recruitment. Clinically, compared with serum IL-8, urine IL-8 was a better biomarker for bladder cancer prognosis and clinical benefit of immune checkpoint blockade (ICB). Additionally, targeting neutrophils or hepatocyte growth factor receptor (MET) signaling combined with ICB inhibited bladder cancer progression and boosted the antitumor effect of CD8+ T cells in mice. These findings reveal the mechanism by which tumor-neutrophil cross talk orchestrates the bladder cancer microenvironment and provide combination strategies, which may have broad impacts on patients suffering from malignancies enriched with neutrophils.


Subject(s)
Disease Progression , Interleukin-8 , Neutrophils , Tumor Microenvironment , Urinary Bladder Neoplasms , Urinary Bladder Neoplasms/pathology , Urinary Bladder Neoplasms/metabolism , Urinary Bladder Neoplasms/immunology , Tumor Microenvironment/immunology , Humans , Neutrophils/immunology , Neutrophils/metabolism , Animals , Mice , Interleukin-8/metabolism , Cell Line, Tumor , Hepatocyte Growth Factor/metabolism , CD8-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/metabolism , B7-H1 Antigen/metabolism , Immune Checkpoint Inhibitors/pharmacology , Immune Checkpoint Inhibitors/therapeutic use , Female , Male , Neutrophil Infiltration
2.
Eur J Med Res ; 29(1): 270, 2024 May 04.
Article in English | MEDLINE | ID: mdl-38704575

ABSTRACT

BACKGROUND: This study aims to investigate the effects of a conditioned medium (CM) from human umbilical cord mesenchymal stem cells (HuMSCs) cultivated in gelatin sponge (GS-HuMSCs-CM) on hair growth in a mouse model. METHODS: CM was collected from the HuMSCs cultivated in a monolayer or in a gelatin sponge. Vascular endothelial growth factor (VEGF), insulin-like growth factor-1 (IGF-1), keratinocyte growth factor (KGF), and hepatocyte growth factor (HGF) levels in CMs were measured by enzyme-linked immunosorbent assays (ELISAs). A hair loss model by a C57 BL/6J mouse was prepared. The effects of GS-HuMSCs-CM and HuMSCs on hair regrowth in mice were investigated by intradermal injection in the depilated back skin with normal saline (NS) as the control. The time for hair regrowth and full covering in depilated areas was observed, and the hair growth was evaluated histologically and by grossly measuring hair length and diameter. RESULTS: Compared with monolayer cultured cells, the three-dimensional (3D) culture of HuMSCs in gelatin sponge drastically increased VEGF, IGF-1, KGF, and HGF production. GS-HuMSCs-CM and HuMSCs injection both promoted hair regeneration in mice, while GS-HuMSCs-CM presented more enhanced effects in hair length, hair diameter, and growth rate. GS-HuMSCs-CM significantly promoted angiogenesis in injected skin areas, which might also contribute to faster hair regrowth. CONCLUSION: GS-HuMSCs-CM exerted significant effects on inducing hair growth and promoted skin angiogenesis in C57BL/6J mice.


Subject(s)
Hair , Insulin-Like Growth Factor I , Mesenchymal Stem Cells , Umbilical Cord , Animals , Mesenchymal Stem Cells/cytology , Mesenchymal Stem Cells/drug effects , Mesenchymal Stem Cells/metabolism , Humans , Culture Media, Conditioned/pharmacology , Mice , Umbilical Cord/cytology , Hair/growth & development , Hair/drug effects , Insulin-Like Growth Factor I/metabolism , Vascular Endothelial Growth Factor A/metabolism , Hepatocyte Growth Factor/metabolism , Gelatin/chemistry , Tissue Scaffolds/chemistry , Mice, Inbred C57BL , Cells, Cultured , Fibroblast Growth Factor 7/metabolism
3.
J Diabetes Res ; 2024: 5574968, 2024.
Article in English | MEDLINE | ID: mdl-38800586

ABSTRACT

Islet transplantation (ITx) is an established and safe alternative to pancreas transplantation for type 1 diabetes mellitus (T1DM) patients. However, most ITx recipients lose insulin independence by 3 years after ITx due to early graft loss, such that multiple donors are required to achieve insulin independence. In the present study, we investigated whether skeletal myoblast cells could be beneficial for promoting angiogenesis and maintaining the differentiated phenotypes of islets. In vitro experiments showed that the myoblast cells secreted angiogenesis-related cytokines (vascular endothelial growth factor (VEGF), hepatocyte growth factor (HGF), and stromal-derived factor-1α (SDF-1α)), contributed to maintenance of differentiated islet phenotypes, and enhanced islet cell insulin secretion capacity. To verify these findings in vivo, we transplanted islets alone or with myoblast cells under the kidney capsule of streptozotocin-induced diabetic mice. Compared with islets alone, the group bearing islets with myoblast cells had a significantly lower average blood glucose level. Histological examination revealed that transplants with islets plus myoblast cells were associated with a significantly larger insulin-positive area and significantly higher number of CD31-positive microvessels compared to islets alone. Furthermore, islets cotransplanted with myoblast cells showed JAK-STAT signaling activation. Our results suggest two possible mechanisms underlying enhancement of islet graft function with myoblast cells cotransplantation: "indirect effects" mediated by angiogenesis and "direct effects" of myoblast cells on islets via the JAK-STAT cascade. Overall, these findings suggest that skeletal myoblast cells enhance the function of transplanted islets, implying clinical potential for a novel ITx procedure involving myoblast cells for patients with diabetes.


Subject(s)
Diabetes Mellitus, Experimental , Insulin , Islets of Langerhans Transplantation , Myoblasts, Skeletal , Neovascularization, Physiologic , Animals , Islets of Langerhans Transplantation/methods , Diabetes Mellitus, Experimental/metabolism , Myoblasts, Skeletal/transplantation , Myoblasts, Skeletal/metabolism , Mice , Male , Insulin/metabolism , Hepatocyte Growth Factor/metabolism , Mice, Inbred C57BL , Vascular Endothelial Growth Factor A/metabolism , Islets of Langerhans/metabolism , Islets of Langerhans/blood supply , Chemokine CXCL12/metabolism , Blood Glucose/metabolism , Diabetes Mellitus, Type 1/metabolism , Diabetes Mellitus, Type 1/physiopathology , Diabetes Mellitus, Type 1/surgery , Signal Transduction , Insulin Secretion , Cell Differentiation
4.
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 , Pedigree , Humans , Lymphedema/genetics , Hepatocyte Growth Factor/genetics , Hepatocyte Growth Factor/metabolism , Female , Male , Middle Aged , Adult
5.
Nat Commun ; 15(1): 3165, 2024 Apr 11.
Article in English | MEDLINE | ID: mdl-38605010

ABSTRACT

The mechanisms of bifurcation, a key step in thyroid development, are largely unknown. Here we find three zebrafish lines from a forward genetic screening with similar thyroid dysgenesis phenotypes and identify a stop-gain mutation in hgfa and two missense mutations in met by positional cloning from these zebrafish lines. The elongation of the thyroid primordium along the pharyngeal midline was dramatically disrupted in these zebrafish lines carrying a mutation in hgfa or met. Further studies show that MAPK inhibitor U0126 could mimic thyroid dysgenesis in zebrafish, and the phenotypes are rescued by overexpression of constitutively active MEK or Snail, downstream molecules of the HGF/Met pathway, in thyrocytes. Moreover, HGF promotes thyrocyte migration, which is probably mediated by downregulation of E-cadherin expression. The delayed bifurcation of the thyroid primordium is also observed in thyroid-specific Met knockout mice. Together, our findings reveal that HGF/Met is indispensable for the bifurcation of the thyroid primordium during thyroid development mediated by downregulation of E-cadherin in thyrocytes via MAPK-snail pathway.


Subject(s)
Hepatocyte Growth Factor , Thyroid Dysgenesis , Animals , Mice , Hepatocyte Growth Factor/genetics , Hepatocyte Growth Factor/metabolism , Zebrafish/genetics , Zebrafish/metabolism , Cadherins/genetics , Thyroid Dysgenesis/genetics , Proto-Oncogene Proteins c-met/genetics , Proto-Oncogene Proteins c-met/metabolism
6.
Int J Biol Macromol ; 268(Pt 1): 131560, 2024 May.
Article in English | MEDLINE | ID: mdl-38631570

ABSTRACT

Triple-negative breast cancer (TNBC) is a subtype of breast cancer with a poor prognosis. Cancer-associated fibroblasts (CAFs) play a critical role in regulating TNBC tumor development. This study aimed to identify and characterize a specific subtype of CAFs associated with TNBC. Initially, using high-throughput bulk transcriptomic data in two cohorts, we identified three CAF-related subtypes (CS1, CS2, CS3) in TNBC samples. These three CAFs subtypes were closely linked to the tumor microenvironment. The CS1 subtype exhibited a relatively immune-rich microenvironment and a favourable prognosis, whereas the CS3 subtype displayed an immune-deprived tumor microenvironment and an unfavourable prognosis. Through WGCNA analysis, POSTN was identified as a key biomarker for CAFs associated with TNBC. Then, POSTN+CAFs was identified and characterized. Both POSTN and POSTN+CAFs showed significant positive correlations with stromal molecules HGF and MET at both the transcriptional and protein levels. Specifically co-localized with CAFs in the tumor stromal area, POSTN, produced by POSTN+CAFs, could modulate the HGF-MET axis, serving as a bypass activation pathway to regulate tumor cell proliferation in response to EGFR inhibitor and MET inhibitor. This study underscores the significance of POSTN and POSTN+CAFs as crucial targets for the diagnosis and treatment of TNBC.


Subject(s)
Cancer-Associated Fibroblasts , Cell Adhesion Molecules , Gene Expression Regulation, Neoplastic , Proto-Oncogene Proteins c-met , Triple Negative Breast Neoplasms , Tumor Microenvironment , Triple Negative Breast Neoplasms/pathology , Triple Negative Breast Neoplasms/metabolism , Triple Negative Breast Neoplasms/genetics , Humans , Cancer-Associated Fibroblasts/metabolism , Cancer-Associated Fibroblasts/pathology , Cell Adhesion Molecules/metabolism , Cell Adhesion Molecules/genetics , Female , Proto-Oncogene Proteins c-met/metabolism , Proto-Oncogene Proteins c-met/genetics , Hepatocyte Growth Factor/metabolism , Hepatocyte Growth Factor/genetics , Cell Proliferation , Biomarkers, Tumor/metabolism , Cell Line, Tumor , Prognosis
7.
Anticancer Res ; 44(5): 1973-1981, 2024 May.
Article in English | MEDLINE | ID: mdl-38677740

ABSTRACT

BACKGROUND/AIM: A role for cold-shock domain (CSD) proteins in abnormal cell proliferation has been suggested in the literature. The aim of this study was to investigate the effect of hepatocyte growth factor (HGF)-induced up-regulation of CSD protein A (CSDA) expression on vascular endothelial growth factor (VEGF) expression and its role in gastric cancer cell invasion and proliferation. MATERIALS AND METHODS: We assessed effects on two gastric cancer cell lines using reverse transcription-polymerase chain reaction, western blotting, 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide (MTT) assays, and CSDA knockdown with short hairpin RNA. RESULTS: Hepatocyte growth factor (HGF) elevates CSDA levels in gastric cancer cell lines. To elucidate the mechanism by which HGF prompts CSDA expression and its impact on vascular endothelial growth factor (VEGF), we applied the Mitogen Activated Protein Kinase (MAPK) inhibitor PD098059 and conducted analyses using western blot. Following the administration of PD098059, a reduction in the protein levels of HGF-stimulated VEGF was observed. Additionally, silencing of CSDA resulted in diminished levels of both VEGF and phosphorylated extracellular signal-regulated kinase (ERK). The suppression of CSDA also led to reduced HGF-induced cell proliferation and diminished invasive capabilities in vitro. Furthermore, our research pinpointed a potential activator protein-1 (AP-1) binding site within the VEGF promoter zone, validating its activity via chromatin immunoprecipitation assays. Electrophoretic mobility shift assays further disclosed that HGF-induced CSDA augmentation correlates with an increase in AP-1 binding to VEGF. CONCLUSION: CSDA is crucial for the proliferation of gastric cancer cells, and the inhibition of this protein could impede the advancement of gastric cancer.


Subject(s)
Cell Proliferation , Hepatocyte Growth Factor , Proteinase Inhibitory Proteins, Secretory , Stomach Neoplasms , Vascular Endothelial Growth Factor A , Humans , Stomach Neoplasms/pathology , Stomach Neoplasms/metabolism , Stomach Neoplasms/genetics , Hepatocyte Growth Factor/metabolism , Hepatocyte Growth Factor/genetics , Vascular Endothelial Growth Factor A/metabolism , Vascular Endothelial Growth Factor A/genetics , Cell Line, Tumor , Gene Expression Regulation, Neoplastic/drug effects , Neoplasm Metastasis , Cell Movement/drug effects , Neoplasm Invasiveness
8.
Immunol Cell Biol ; 102(5): 381-395, 2024.
Article in English | MEDLINE | ID: mdl-38629182

ABSTRACT

Resident macrophages of various mammalian organs are characterized by several distinctive features in their gene expression profile and phenotype, including involvement in the regulation of organ functions, as well as reduced sensitivity to proinflammatory activation factors. The reasons for the formation of such a specific phenotype remain the subject of intensive research. Some papers emphasize the role of the origin of organ macrophages. Other studies indicate that monocytes that develop in the red bone marrow are also able to form resident macrophages with a phenotype characteristic of a particular organ, but this requires appropriate microenvironmental conditions. In this article, we studied the possibility of differentiation of monocyte-derived macrophages into cells with a Kupffer-like phenotype under the influence of the main stromal components of Kupffer cells macrophage niche: Ito cells, liver sinusoid endotheliocytes and hepatocyte growth factor (HGF). It was found that Kupffer cells are characterized by several features, including increased expression of transcription factors Spic and Id3, as well as MUP family genes, Clusterin and Ngp genes. In addition, Kupffer cells were characterized by a higher proliferative activity. The expression of marker genes of Kupffer cells (i.e. Id3, Spic, Marco and Timd4) increased in monocyte-derived macrophages during coculture with Ito cells, liver sinusoid endothelial cells, macrophage colony-stimulating factor and HGF cells only by 3 days. However, the expression level of these genes was always higher in Kupffer cells. In addition, a complete coincidence of the expressed gene profile in monocyte-derived macrophages and Kupffer cells did not occur even after 3 days of culturing.


Subject(s)
Cell Differentiation , Cellular Microenvironment , Kupffer Cells , Macrophages , Phenotype , Kupffer Cells/metabolism , Kupffer Cells/cytology , Macrophages/metabolism , Animals , Monocytes/metabolism , Monocytes/cytology , Hepatocyte Growth Factor/metabolism , Endothelial Cells/metabolism , Coculture Techniques , Humans , Cell Proliferation , Cells, Cultured , Liver/cytology , Liver/metabolism , Mice
9.
Sci Signal ; 17(834): eadj6603, 2024 Apr 30.
Article in English | MEDLINE | ID: mdl-38687825

ABSTRACT

The localization, number, and function of postsynaptic AMPA-type glutamate receptors (AMPARs) are crucial for synaptic plasticity, a cellular correlate for learning and memory. The Hippo pathway member WWC1 is an important component of AMPAR-containing protein complexes. However, the availability of WWC1 is constrained by its interaction with the Hippo pathway kinases LATS1 and LATS2 (LATS1/2). Here, we explored the biochemical regulation of this interaction and found that it is pharmacologically targetable in vivo. In primary hippocampal neurons, phosphorylation of LATS1/2 by the upstream kinases MST1 and MST2 (MST1/2) enhanced the interaction between WWC1 and LATS1/2, which sequestered WWC1. Pharmacologically inhibiting MST1/2 in male mice and in human brain-derived organoids promoted the dissociation of WWC1 from LATS1/2, leading to an increase in WWC1 in AMPAR-containing complexes. MST1/2 inhibition enhanced synaptic transmission in mouse hippocampal brain slices and improved cognition in healthy male mice and in male mouse models of Alzheimer's disease and aging. Thus, compounds that disrupt the interaction between WWC1 and LATS1/2 might be explored for development as cognitive enhancers.


Subject(s)
Hippocampus , Intracellular Signaling Peptides and Proteins , Neuronal Plasticity , Phosphoproteins , Protein Serine-Threonine Kinases , Receptors, AMPA , Animals , Protein Serine-Threonine Kinases/metabolism , Protein Serine-Threonine Kinases/genetics , Male , Humans , Receptors, AMPA/metabolism , Receptors, AMPA/genetics , Mice , Neuronal Plasticity/physiology , Hippocampus/metabolism , Hippo Signaling Pathway , Serine-Threonine Kinase 3 , Signal Transduction , Memory/physiology , Tumor Suppressor Proteins/metabolism , Tumor Suppressor Proteins/genetics , Hepatocyte Growth Factor/metabolism , Mice, Inbred C57BL , Alzheimer Disease/metabolism , Phosphorylation , Neurons/metabolism
10.
Shock ; 61(6): 951-960, 2024 Jun 01.
Article in English | MEDLINE | ID: mdl-38598838

ABSTRACT

ABSTRACT: Objectives: Puerarin, the principal active constituent extracted from Pueraria, is believed to confer protection against sepsis-induced lung injury. The study aimed to elucidate the role and mechanism of Mst1/ERS in puerarin-mediated protection against acute lung injury (ALI). Methods: Monolayer vascular endothelial cell permeability was assessed by gauging the paracellular flow of FITC-dextran 40,000 (FD40). ELISA was employed for the quantification of inflammatory cytokines. Identification of target proteins was conducted through western blotting. Histological alterations and apoptosis were scrutinized using hematoxylin-eosin staining and TUNEL staining, respectively. The ultrastructure of the endoplasmic reticulum was observed via transmission electron microscopy. Results: Puerarin significantly protected mice from LPS-induced ALI, reducing lung interstitial width, neutrophil and lymphocyte infiltration, pulmonary interstitial and alveolar edema, and lung apoptosis. Puerarin treatment also markedly attenuated levels of TNF-α and IL-1ß in both alveolar lavage fluid and serum. Furthermore, puerarin significantly attenuated LPS-induced increases in Mst1, GRP78, CHOP, and Caspase12 protein expression and blunted LPS-induced decrease in ZO-1 protein expression in lung tissues. Puerarin obviously reduced endoplasmic reticulum expansion and vesiculation. Similarly, puerarin significantly mitigated the LPS-induced reduction in HUVEC cell viability and ZO-1 expression. Puerarin also attenuated LPS-induced increase in apoptosis, TNF-α and IL-1ß, FD40 flux, and Mst1, GRP78, CHOP, and Caspase12 expression in HUVEC cells. Nevertheless, the inhibitory impact of puerarin on vascular endothelial cell injury, lung injury, and endoplasmic reticulum stress (ERS) was diminished by Mst1 overexpression. Conclusion: These findings demonstrated that the Mst1/ERS signaling pathway played a pivotal role in the development of LPS-induced vascular endothelial cell dysfunction and ALI. Puerarin exhibited the ability to attenuate LPS-induced vascular endothelial cell dysfunction and ALI by inhibiting the Mst1/ERS signaling pathway.


Subject(s)
Acute Lung Injury , Endoplasmic Reticulum Chaperone BiP , Endoplasmic Reticulum Stress , Isoflavones , Signal Transduction , Acute Lung Injury/drug therapy , Acute Lung Injury/metabolism , Acute Lung Injury/prevention & control , Isoflavones/pharmacology , Isoflavones/therapeutic use , Animals , Mice , Signal Transduction/drug effects , Male , Endoplasmic Reticulum Stress/drug effects , Humans , Hepatocyte Growth Factor/metabolism , Lipopolysaccharides/toxicity , Proto-Oncogene Proteins/metabolism , Apoptosis/drug effects , Mice, Inbred C57BL , Human Umbilical Vein Endothelial Cells/metabolism , Human Umbilical Vein Endothelial Cells/drug effects
11.
Tissue Cell ; 87: 102326, 2024 Apr.
Article in English | MEDLINE | ID: mdl-38442547

ABSTRACT

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.


Subject(s)
Indoles , Liver Failure, Acute , Mesenchymal Stem Cell Transplantation , Mesenchymal Stem Cells , Piperazines , Sulfonamides , Humans , Mice , Animals , Liver Failure, Acute/therapy , Liver Failure, Acute/metabolism , Hypoxia/metabolism , Oxygen/metabolism , Cell Proliferation , Hepatocyte Growth Factor/metabolism , Hepatocyte Growth Factor/pharmacology
12.
Oncogene ; 43(19): 1431-1444, 2024 May.
Article in English | MEDLINE | ID: mdl-38485737

ABSTRACT

MET amplification/mutations are important targetable oncogenic drivers in NSCLC, however, acquired resistance is inevitable and the majority of patients with targetable MET alterations fail to respond to MET tyrosine kinase inhibitors (TKIs). Furthermore, MET amplification is among the most common mediators of TKI resistance. As such, novel therapies to target MET pathway and overcome MET TKI resistance are clearly needed. Here we show that the epithelial-mesenchymal transition (EMT) transcription factor, TWIST1 is a key downstream mediator of HGF/MET induced resistance through suppression of p27 and targeting TWIST1 can overcome resistance. We found that TWIST1 is overexpressed at the time of TKI resistance in multiple MET-dependent TKI acquired resistance PDX models. We have shown for the first time that MET directly stabilized the TWIST protein leading to TKI resistance and that TWIST1 was required for MET-driven lung tumorigenesis as well as could induce MET TKI resistance when overexpressed. TWIST1 mediated MET TKI resistance through suppression of p27 expression and genetic or pharmacologic inhibition of TWIST1 overcame TKI resistance in vitro and in vivo. Our findings suggest that targeting TWIST1 may be an effective therapeutic strategy to overcome resistance in MET-driven NSCLC as well as in other oncogene driven subtypes in which MET amplification is the resistance mechanism.


Subject(s)
Drug Resistance, Neoplasm , Hepatocyte Growth Factor , Lung Neoplasms , Nuclear Proteins , Protein Kinase Inhibitors , Proto-Oncogene Proteins c-met , Twist-Related Protein 1 , Twist-Related Protein 1/genetics , Twist-Related Protein 1/metabolism , Proto-Oncogene Proteins c-met/genetics , Proto-Oncogene Proteins c-met/metabolism , Humans , Lung Neoplasms/genetics , Lung Neoplasms/pathology , Lung Neoplasms/drug therapy , Lung Neoplasms/metabolism , Drug Resistance, Neoplasm/genetics , Hepatocyte Growth Factor/genetics , Hepatocyte Growth Factor/metabolism , Nuclear Proteins/genetics , Nuclear Proteins/metabolism , Animals , Mice , Protein Kinase Inhibitors/pharmacology , Protein Kinase Inhibitors/therapeutic use , Signal Transduction/genetics , Carcinoma, Non-Small-Cell Lung/genetics , Carcinoma, Non-Small-Cell Lung/pathology , Carcinoma, Non-Small-Cell Lung/drug therapy , Carcinoma, Non-Small-Cell Lung/metabolism , Epithelial-Mesenchymal Transition/genetics , Cell Line, Tumor , Gene Expression Regulation, Neoplastic , Oncogenes/genetics , Xenograft Model Antitumor Assays
13.
Cancer Lett ; 588: 216780, 2024 Apr 28.
Article in English | MEDLINE | ID: mdl-38462033

ABSTRACT

Breast cancer is the most common malignancy among women, posing a formidable health challenge worldwide. In this complex landscape, the c-MET (cellular-mesenchymal epithelial transition factor) receptor tyrosine kinase (RTK), also recognized as the hepatocyte growth factor (HGF) receptor (HGFR), emerges as a prominent protagonist, displaying overexpression in nearly 50% of breast cancer cases. Activation of c-MET by its ligand, HGF, secreted by neighboring mesenchymal cells, contributes to a cascade of tumorigenic processes, including cell proliferation, metastasis, angiogenesis, and immunosuppression. While c-MET inhibitors such as crizotinib, capmatinib, tepotinib and cabozantinib have garnered FDA approval for non-small cell lung cancer (NSCLC), their potential within breast cancer therapy is still undetermined. This comprehensive review embarks on a journey through structural biology, multifaceted functions, and intricate signaling pathways orchestrated by c-MET across cancer types. Furthermore, we highlight the pivotal role of c-MET-targeted therapies in breast cancer, offering a clinical perspective on this promising avenue of intervention. In this pursuit, we strive to unravel the potential of c-MET as a beacon of hope in the fight against breast cancer, unveiling new horizons for therapeutic innovation.


Subject(s)
Breast Neoplasms , Carcinoma, Non-Small-Cell Lung , Lung Neoplasms , Female , Humans , Carcinoma, Non-Small-Cell Lung/pathology , Breast Neoplasms/drug therapy , Breast Neoplasms/genetics , Breast Neoplasms/pathology , Lung Neoplasms/metabolism , Hepatocyte Growth Factor/metabolism , Proto-Oncogene Proteins c-met/metabolism , Signal Transduction
14.
Cell Rep ; 43(4): 114002, 2024 Apr 23.
Article in English | MEDLINE | ID: mdl-38547126

ABSTRACT

The dysfunction of matriptase, a membrane-anchored protease, is highly related to the progression of skin and breast cancers. Epidermal growth factor (EGF)-induced matriptase activation and cancer invasion are known but with obscure mechanisms. Here, we demonstrate a vesicular-trafficking-mediated interplay between matriptase and EGF signaling in cancer promotion. We found that EGF induces matriptase to undergo endocytosis together with the EGF receptor, followed by acid-induced activation in endosomes. Activated matriptase is then secreted extracellularly on exosomes to catalyze hepatocyte growth factor precursor (pro-HGF) cleavage, resulting in autocrine HGF/c-Met signaling. Matriptase-induced HGF/c-Met signaling represents the second signal wave of EGF, which promotes cancer cell scattering, migration, and invasion. These findings demonstrate a role of vesicular trafficking in efficient activation and secretion of membrane matriptase and a reciprocal regulation of matriptase and EGF signaling in cancer promotion, providing insights into the physiological functions of vesicular trafficking and the molecular pathological mechanisms of skin and breast cancers.


Subject(s)
Breast Neoplasms , Neoplasm Invasiveness , Serine Endopeptidases , Signal Transduction , Animals , Female , Humans , Mice , Breast Neoplasms/metabolism , Breast Neoplasms/pathology , Cell Line, Tumor , Cell Movement , Endocytosis , Endosomes/metabolism , Epidermal Growth Factor/metabolism , ErbB Receptors/metabolism , Exosomes/metabolism , Hepatocyte Growth Factor/metabolism , Protein Precursors , Proto-Oncogene Proteins c-met/metabolism , Serine Endopeptidases/metabolism , Skin Neoplasms/pathology , Skin Neoplasms/metabolism
15.
Nat Commun ; 15(1): 1231, 2024 Feb 09.
Article in English | MEDLINE | ID: mdl-38336745

ABSTRACT

Androgen deprivation therapy (ADT) targeting androgen/androgen receptor (AR)- signaling pathways is the main therapy for advanced prostate cancer (PCa). However, ADT eventually fails in most patients who consequently develop castration-resistant prostate cancer (CRPC). While more potent AR antagonists and blockers for androgen synthesis were developed to improve clinical outcomes, they also show to induce more diverse CRPC phenotypes. Specifically, the AR- and neuroendocrine-null PCa, DNPC, occurs in abiraterone and enzalutamide-treated patients. Here, we uncover that current ADT induces aberrant HGF/MET signaling activation that further elevates Wnt/ß-catenin signaling in human DNPC samples. Co-activation of HGF/MET and Wnt/ß-catenin axes in mouse prostates induces DNPC-like lesions. Single-cell RNA sequencing analyses identify increased expression and activity of XPO1 and ribosomal proteins in mouse DNPC-like cells. Elevated expression of XPO1 and ribosomal proteins is also identified in clinical DNPC specimens. Inhibition of XPO1 and ribosomal pathways represses DNPC growth in both in vivo and ex vivo conditions, evidencing future therapeutic targets.


Subject(s)
Androgens , Prostatic Neoplasms, Castration-Resistant , Male , Humans , Mice , Animals , Androgens/metabolism , Prostatic Neoplasms, Castration-Resistant/metabolism , Androgen Antagonists/pharmacology , beta Catenin/metabolism , Active Transport, Cell Nucleus , Wnt Signaling Pathway , Ribosomal Proteins/metabolism , Receptors, Androgen/genetics , Receptors, Androgen/metabolism , Cell Line, Tumor , Hepatocyte Growth Factor/metabolism
16.
Cell Mol Life Sci ; 81(1): 28, 2024 Jan 12.
Article in English | MEDLINE | ID: mdl-38212428

ABSTRACT

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.


Subject(s)
Colorectal Neoplasms , Signal Transduction , Humans , Cetuximab/pharmacology , Proto-Oncogene Proteins c-met/genetics , Proto-Oncogene Proteins c-met/metabolism , Drug Resistance, Neoplasm/genetics , Colorectal Neoplasms/drug therapy , Colorectal Neoplasms/genetics , Colorectal Neoplasms/metabolism , Protease Inhibitors/pharmacology , Peptide Hydrolases/metabolism , Cell Line, Tumor , Hepatocyte Growth Factor/genetics , Hepatocyte Growth Factor/metabolism , Hepatocyte Growth Factor/pharmacology
17.
Vasa ; 53(2): 120-128, 2024 Apr.
Article in English | MEDLINE | ID: mdl-38205733

ABSTRACT

Background: Hepatocyte growth factor (HGF) is a pleiotropic cytokine mainly produced by mesenchymal cells. After endothelial damage by oxidized low-density lipoprotein (LDL), HGF is produced and released into the circulation in response. Due to this mechanism HGF has been proposed as possible clinical biomarker for clinical as well as subclinical atherosclerosis. Patients and methods: The conducted study is an observational, single centre, cohort study, including 171 patients with at least one cardiovascular risk factor or already established cardiovascular disease (CVD). Each patient underwent 3D plaque volumetry of the carotid and femoral arteries as well as physical examination and record of the medical history. Additionally, plasma HGF and further laboratory parameters like high sensitivity C-reactive protein and LDL-cholesterol were determined. Results: 169 patients were available for statistical analysis. In bivariate correlation, HGF showed a highly significant correlation with total plaque volume (TPV, r=0.48; p<0.001). In receiver operating characteristic (ROC) analysis for high TPV, HGF showed an area under the curve (AUC) of 0.68 (CI 95%: 0.59-0.77, p<0.001) with a sensitivity of 78% and a specificity of 52% to predict high TPV at a cut-off of 959 ng/ml. In the ROC-analysis for the presence of CVD, HGF demonstrated an AUC of 0.65 (95% CI 0.55-0.73; p=0.01) with a sensitivity of 77% and a specificity of 52%. Conclusions: Higher plasma levels of HGF are associated with higher atherosclerotic plaque volume as measured by 3D-ultrasound.


Subject(s)
Atherosclerosis , Hepatocyte Growth Factor , Humans , Atherosclerosis/diagnostic imaging , Cardiovascular Diseases , Cohort Studies , Hepatocyte Growth Factor/metabolism , Plaque, Atherosclerotic/complications , Risk Factors
18.
Mol Syst Biol ; 20(3): 187-216, 2024 Mar.
Article in English | MEDLINE | ID: mdl-38216754

ABSTRACT

Chronic liver diseases are worldwide on the rise. Due to the rapidly increasing incidence, in particular in Western countries, metabolic dysfunction-associated steatotic liver disease (MASLD) is gaining importance as the disease can develop into hepatocellular carcinoma. Lipid accumulation in hepatocytes has been identified as the characteristic structural change in MASLD development, but molecular mechanisms responsible for disease progression remained unresolved. Here, we uncover in primary hepatocytes from a preclinical model fed with a Western diet (WD) an increased basal MET phosphorylation and a strong downregulation of the PI3K-AKT pathway. Dynamic pathway modeling of hepatocyte growth factor (HGF) signal transduction combined with global proteomics identifies that an elevated basal MET phosphorylation rate is the main driver of altered signaling leading to increased proliferation of WD-hepatocytes. Model-adaptation to patient-derived hepatocytes reveal patient-specific variability in basal MET phosphorylation, which correlates with patient outcome after liver surgery. Thus, dysregulated basal MET phosphorylation could be an indicator for the health status of the liver and thereby inform on the risk of a patient to suffer from liver failure after surgery.


Subject(s)
Carcinoma, Hepatocellular , Fatty Liver , Liver Neoplasms , Humans , Phosphorylation , Phosphatidylinositol 3-Kinases/metabolism , Hepatocytes/metabolism , Hepatocyte Growth Factor/metabolism , Fatty Liver/metabolism , Liver Neoplasms/pathology
19.
Stem Cells Dev ; 33(3-4): 89-103, 2024 Feb.
Article in English | MEDLINE | ID: mdl-38164089

ABSTRACT

Mesenchymal stem cells (MSCs) directly differentiate into neurons and endothelial cells after transplantation, and their secretome has considerable potential for treating brain injuries. Previous studies have suggested that the effects of MSCs priming with exposure to hypoxia, cytokines, growth factors, or chemical agents could optimize the paracrine potency and therapeutic potential of MSCs. Studies have suggested that thrombin-primed Wharton's Jelly-derived mesenchymal stem cells (Th.WJ-MSCs) significantly enhance the neuroprotective beneficial effects of naive MSCs in brain injury such as hypoxic-ischemic brain injury (HIE) and intraventricular hemorrhage (IVH). This study aimed to characterize WJ-MSCs in terms of stem cell markers, differentiation, cell proliferation, and paracrine factors by comparing naive and Th.WJ-MSCs. We demonstrated that compared with naive MSCs, Th.MSCs significantly enhanced the neuroprotective effects in vitro. Moreover, we identified differentially expressed proteins in the conditioned media of naive and Th.WJ-MSCs by liquid chromatography-tandem mass spectrometry analysis. Secretome analysis of the conditioned medium of WJ-MSCs revealed that such neuroprotective effects were mediated by paracrine effects with secretomes of Th.WJ-MSCs, and hepatocyte growth factor was identified as a key paracrine mediator. These results can be applied further in the preclinical and clinical development of effective and safe cell therapeutics for brain injuries such as HIE and IVH.


Subject(s)
Brain Injuries , Mesenchymal Stem Cells , Neuroprotective Agents , STAT3 Transcription Factor , Wharton Jelly , Humans , Hepatocyte Growth Factor/metabolism , Neuroprotective Agents/pharmacology , Thrombin/pharmacology , Thrombin/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Endothelial Cells/metabolism , Cells, Cultured , Signal Transduction , Cell Differentiation , Immunologic Factors/metabolism , Brain Injuries/metabolism , Cell Proliferation
20.
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
SELECTION OF CITATIONS
SEARCH DETAIL
...