Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 25.212
Filter
2.
Nat Commun ; 15(1): 5010, 2024 Jun 12.
Article in English | MEDLINE | ID: mdl-38866762

ABSTRACT

Primary human hepatocyte (PHH) transplantation is a promising alternative to liver transplantation, whereby liver function could be restored by partial repopulation of the diseased organ with healthy cells. However, currently PHH engraftment efficiency is low and benefits are not maintained long-term. Here we refine two male mouse models of human chronic and acute liver diseases to recapitulate compromised hepatocyte proliferation observed in nearly all human liver diseases by overexpression of p21 in hepatocytes. In these clinically relevant contexts, we demonstrate that transient, yet robust expression of human hepatocyte growth factor and epidermal growth factor in the liver via nucleoside-modified mRNA in lipid nanoparticles, whose safety was validated with mRNA-based COVID-19 vaccines, drastically improves PHH engraftment, reduces disease burden, and improves overall liver function. This strategy may overcome the critical barriers to clinical translation of cell therapies with primary or stem cell-derived hepatocytes for the treatment of liver diseases.


Subject(s)
Hepatocyte Growth Factor , Hepatocytes , Nanoparticles , RNA, Messenger , Animals , Hepatocytes/metabolism , Hepatocytes/transplantation , Humans , Mice , Male , RNA, Messenger/metabolism , RNA, Messenger/genetics , Nanoparticles/chemistry , Hepatocyte Growth Factor/metabolism , Hepatocyte Growth Factor/genetics , Disease Models, Animal , Liver/metabolism , Epidermal Growth Factor/metabolism , Cell- and Tissue-Based Therapy/methods , COVID-19/therapy , Liver Diseases/therapy , Liver Diseases/metabolism , Liver Diseases/genetics , Cell Proliferation , SARS-CoV-2/genetics , Liposomes
3.
Drug Des Devel Ther ; 18: 1933-1945, 2024.
Article in English | MEDLINE | ID: mdl-38831868

ABSTRACT

Introduction: Vascular ulcers constitute a serious global public health problem, responsible for causing a significant social and economic impact due to their recurrent, disabling nature and the need for prolonged therapies to cure them. Objective: To evaluate the use and efficacy of the rhEGF in the epithelialization of patients with a diagnosis of CEAP stage 6 venous insufficiency, in the two regimes of the health system in Colombia, the contributive (equivalent to a health system where citizens with payment capacity contribute a percentage of their salary) and the subsidized (equivalent to a health system where the state covers the vulnerable population and low socioeconomic level) versus the other treatments used. Methodology: Observational, descriptive, retrospective, multicenter study, in which 105 medical records with 139 ulcers were reviewed, in 2 centers, one belonging to the subsidized system and the other to the contributive system in Colombia. Results: The association with the epithelialization variable of the different treatment groups for ulcers according to the application of the mixed effect model test, for both regimes was for the Biologicals (EC 34.401/p = 0.000), Bioactive Agents (Hydrogels) (EC 24.735/p = 0.005) groups; for the rest of the treatment groups, the results were neither associated nor statistically significant. Conclusion: Intra- and perilesional therapy with rhEGF expands the therapeutic spectrum in patients with venous ulcers, regardless of the type of health system in which it will be applied, shortening the healing time and reaching a possible therapeutic goal, which according to this study there is an association with epithelialization regardless of the regime applied.


Subject(s)
Varicose Ulcer , Humans , Colombia , Varicose Ulcer/drug therapy , Varicose Ulcer/economics , Retrospective Studies , Male , Female , Middle Aged , Epidermal Growth Factor , Recombinant Proteins/economics , Recombinant Proteins/therapeutic use , Recombinant Proteins/administration & dosage , Aged
4.
Sci Signal ; 17(839): eade8041, 2024 Jun 04.
Article in English | MEDLINE | ID: mdl-38833528

ABSTRACT

A long-standing question in the field of signal transduction is how distinct signaling pathways interact with each other to control cell behavior. Growth factor receptors and G protein-coupled receptors (GPCRs) are the two major signaling hubs in eukaryotes. Given that the mechanisms by which they signal independently have been extensively characterized, we investigated how they may cross-talk with each other. Using linear ion trap mass spectrometry and cell-based biophysical, biochemical, and phenotypic assays, we found at least three distinct ways in which epidermal growth factor affected canonical G protein signaling by the Gi-coupled GPCR CXCR4 through the phosphorylation of Gαi. Phosphomimicking mutations in two residues in the αE helix of Gαi (tyrosine-154/tyrosine-155) suppressed agonist-induced Gαi activation while promoting constitutive Gßγ signaling. Phosphomimicking mutations in the P loop (serine-44, serine-47, and threonine-48) suppressed Gi activation entirely, thus completely segregating growth factor and GPCR pathways. As expected, most of the phosphorylation events appeared to affect intrinsic properties of Gαi proteins, including conformational stability, nucleotide binding, and the ability to associate with and to release Gßγ. However, one phosphomimicking mutation, targeting the carboxyl-terminal residue tyrosine-320, promoted mislocalization of Gαi from the plasma membrane, a previously uncharacterized mechanism of suppressing GPCR signaling through G protein subcellular compartmentalization. Together, these findings elucidate not only how growth factor and chemokine signals cross-talk through the phosphorylation-dependent modulation of Gαi but also how such cross-talk may generate signal diversity.


Subject(s)
GTP-Binding Protein alpha Subunits, Gi-Go , Receptors, CXCR4 , Signal Transduction , Phosphorylation , Humans , HEK293 Cells , GTP-Binding Protein alpha Subunits, Gi-Go/metabolism , GTP-Binding Protein alpha Subunits, Gi-Go/genetics , GTP-Binding Protein alpha Subunits, Gi-Go/chemistry , Receptors, CXCR4/metabolism , Receptors, CXCR4/genetics , Epidermal Growth Factor/metabolism , Epidermal Growth Factor/genetics , Receptors, G-Protein-Coupled/metabolism , Receptors, G-Protein-Coupled/genetics , Animals
5.
Crit Care Explor ; 6(7): e1109, 2024 Jul 01.
Article in English | MEDLINE | ID: mdl-38922318

ABSTRACT

IMPORTANCE: COVID-19 may injure the kidney tubules via activation of inflammatory host responses and/or direct viral infiltration. Most studies of kidney injury in COVID-19 lacked contemporaneous controls or measured kidney biomarkers at a single time point. OBJECTIVES: To better understand mechanisms of acute kidney injury in COVID-19, we compared kidney outcomes and trajectories of tubular injury, viability, and function in prospectively enrolled critically ill adults with and without COVID-19. DESIGN, SETTING, AND PARTICIPANTS: The COVID-19 Host Response and Outcomes study prospectively enrolled patients admitted to ICUs in Washington State with symptoms of lower respiratory tract infection, determining COVID-19 status by nucleic acid amplification on arrival. MAIN OUTCOMES AND MEASURES: We evaluated major adverse kidney events (MAKE) defined as a doubling of serum creatinine, kidney replacement therapy, or death, in 330 patients after inverse probability weighting. In the 181 patients with available biosamples, we determined trajectories of urine kidney injury molecule-1 (KIM-1) and epithelial growth factor (EGF), and urine:plasma ratios of endogenous markers of tubular secretory clearance. RESULTS: At ICU admission, the mean age was 55 ± 16 years; 45% required mechanical ventilation; and the mean serum creatinine concentration was 1.1 mg/dL. COVID-19 was associated with a 70% greater occurrence of MAKE (relative risk 1.70; 95% CI, 1.05-2.74) and a 741% greater occurrence of KRT (relative risk 7.41; 95% CI, 1.69-32.41). The biomarker cohort had a median of three follow-up measurements. Urine EGF, secretory clearance ratios, and estimated glomerular filtration rate (eGFR) increased over time in the COVID-19 negative group but remained unchanged in the COVID-19 positive group. In contrast, urine KIM-1 concentrations did not significantly change over the course of the study in either group. CONCLUSIONS: Among critically ill adults, COVID-19 is associated with a more protracted course of proximal tubular dysfunction and reduced eGFR despite similar degrees of kidney injury.


Subject(s)
Acute Kidney Injury , COVID-19 , Critical Illness , Hepatitis A Virus Cellular Receptor 1 , Humans , COVID-19/physiopathology , Middle Aged , Male , Acute Kidney Injury/etiology , Acute Kidney Injury/virology , Female , Prospective Studies , Aged , Hepatitis A Virus Cellular Receptor 1/analysis , Hepatitis A Virus Cellular Receptor 1/metabolism , SARS-CoV-2 , Adult , Biomarkers/blood , Biomarkers/urine , Kidney Tubules/pathology , Kidney Tubules/physiopathology , Creatinine/blood , Creatinine/urine , Intensive Care Units , Washington/epidemiology , Epidermal Growth Factor/blood , Epidermal Growth Factor/urine , Renal Replacement Therapy
6.
Cell Mol Life Sci ; 81(1): 272, 2024 Jun 20.
Article in English | MEDLINE | ID: mdl-38900158

ABSTRACT

We addressed the heteromerization of the epidermal growth factor receptor (EGFR) with G-protein coupled receptors (GPCR) on the basis of angiotensin-II-receptor-subtype-1(AT1R)-EGFR interaction as proof-of-concept and show its functional relevance during synergistic nuclear information transfer, beyond ligand-dependent EGFR transactivation. Following in silico modelling, we generated EGFR-interaction deficient AT1R-mutants and compared them to AT1R-wildtype. Receptor interaction was assessed by co-immunoprecipitation (CoIP), Förster resonance energy transfer (FRET) and fluorescence-lifetime imaging microscopy (FLIM). Changes in cell morphology, ERK1/2-phosphorylation (ppERK1/2), serum response factor (SRF)-activation and cFOS protein expression were determined by digital high content microscopy at the single cell level. FRET, FLIM and CoIP confirmed the physical interaction of AT1R-wildtype with EGFR that was strongly reduced for the AT1R-mutants. Responsiveness of cells transfected with AT1R-WT or -mutants to angiotensin II or EGF was similar regarding changes in cell circularity, ppERK1/2 (direct and by ligand-dependent EGFR-transactivation), cFOS-expression and SRF-activity. By contrast, the EGFR-AT1R-synergism regarding these parameters was completely absent for in the interaction-deficient AT1R mutants. The results show that AT1R-EGFR heteromerisation enables AT1R-EGFR-synergism on downstream gene expression regulation, modulating the intensity and the temporal pattern of nuclear AT1R/EGFR-information transfer. Furthermore, remote EGFR transactivation, via ligand release or cytosolic tyrosine kinases, is not sufficient for the complete synergistic control of gene expression.


Subject(s)
Cell Nucleus , ErbB Receptors , Receptor, Angiotensin, Type 1 , ErbB Receptors/metabolism , Humans , Receptor, Angiotensin, Type 1/metabolism , Receptor, Angiotensin, Type 1/genetics , Cell Nucleus/metabolism , Fluorescence Resonance Energy Transfer , Phosphorylation , Receptors, G-Protein-Coupled/metabolism , Receptors, G-Protein-Coupled/genetics , Cell Membrane/metabolism , Angiotensin II/metabolism , Angiotensin II/pharmacology , Epidermal Growth Factor/metabolism , HEK293 Cells , Protein Binding , Serum Response Factor/metabolism , Serum Response Factor/genetics
7.
Cells ; 13(9)2024 Apr 30.
Article in English | MEDLINE | ID: mdl-38727302

ABSTRACT

We have previously shown that the transmembrane protein ODZ1 promotes cytoskeletal remodeling of glioblastoma (GBM) cells and invasion of the surrounding parenchyma through the activation of a RhoA-ROCK pathway. We also described that GBM cells can control the expression of ODZ1 through transcriptional mechanisms triggered by the binding of IL-6 to its receptor and a hypoxic environment. Epidermal growth factor (EGF) plays a key role in the invasive capacity of GBM. However, the molecular mechanisms that enable tumor cells to acquire the morphological changes to migrate out from the tumor core have not been fully characterized. Here, we show that EGF is able to induce the expression of ODZ1 in primary GBM cells. We analyzed the levels of the EGF receptor (EGFR) in 20 GBM primary cell lines and found expression in 19 of them by flow cytometry. We selected two cell lines that do or do not express the EGFR and found that EGFR-expressing cells responded to the EGF ligand by increasing ODZ1 at the mRNA and protein levels. Moreover, blockade of EGF-EGFR binding by Cetuximab, inhibition of the p38 MAPK pathway, or Additionally, the siRNA-mediated knockdown of MAPK11 (p38ß MAPK) reduced the induction of ODZ1 in response to EGF. Overall, we show that EGF may activate an EGFR-mediated signaling pathway through p38ß MAPK, to upregulate the invasion factor ODZ1, which may initiate morphological changes for tumor cells to invade the surrounding parenchyma. These data identify a new candidate of the EGF-EGFR pathway for novel therapeutic approaches.


Subject(s)
Epidermal Growth Factor , ErbB Receptors , Glioblastoma , Tenascin , Up-Regulation , Humans , Cell Line, Tumor , Epidermal Growth Factor/pharmacology , Epidermal Growth Factor/metabolism , ErbB Receptors/metabolism , Gene Expression Regulation, Neoplastic/drug effects , Glioblastoma/pathology , Glioblastoma/metabolism , Glioblastoma/genetics , Neoplasm Invasiveness , Signal Transduction/drug effects , Up-Regulation/drug effects , Up-Regulation/genetics , Nerve Tissue Proteins/genetics , Nerve Tissue Proteins/metabolism , Tenascin/genetics , Tenascin/metabolism
8.
Hematology ; 29(1): 2356292, 2024 Dec.
Article in English | MEDLINE | ID: mdl-38785187

ABSTRACT

OBJECTIVES: This study aims to investigate the role of excessive Protein Tyrosine Phosphatase Non-Receptor Type 21 (PTPN21) in the proliferation of Acute Lymphoblastic Leukemia (ALL) cells with EGF stimulation. METHODS: PTPN21 was overexpressed in ALL cell lines by lentiviral transfection. Apoptosis was assayed by Annexin V/7-AAD staining. The proliferation and cell cycle of EGF-treated ALL cells were assessed by MTT and Ki-67/7-AAD staining respectively. The phosphorylation of Src tyrosine kinase and mediators of distinct MAPK pathways were assessed by Western blot. RESULTS: Overexpression of PTPN21 had minimal effect on the apoptosis of ALL cells, but significantly promoted the proliferation and cell cycle progression of ALL cells stimulated with EGF. The activity of Src tyrosine kinase and the MAPK pathways was elevated. Inhibition of MAPK pathways by specific inhibitors mitigated this pro-proliferative effect of excessive PTPN21 on EGF-stimulated ALL cells. CONCLUSION: PTPN21 may facilitate ALL progression by promoting cell proliferation via the Src/MAPK signaling pathways.


Subject(s)
Cell Proliferation , Epidermal Growth Factor , MAP Kinase Signaling System , Precursor Cell Lymphoblastic Leukemia-Lymphoma , Protein Tyrosine Phosphatases, Non-Receptor , Humans , Apoptosis/drug effects , Cell Line, Tumor , Cell Proliferation/drug effects , Epidermal Growth Factor/pharmacology , MAP Kinase Signaling System/drug effects , Precursor Cell Lymphoblastic Leukemia-Lymphoma/pathology , Precursor Cell Lymphoblastic Leukemia-Lymphoma/metabolism , Precursor Cell Lymphoblastic Leukemia-Lymphoma/genetics , Protein Tyrosine Phosphatases, Non-Receptor/genetics , Protein Tyrosine Phosphatases, Non-Receptor/metabolism
9.
Mol Cell ; 84(11): 2152-2165.e5, 2024 Jun 06.
Article in English | MEDLINE | ID: mdl-38781971

ABSTRACT

A disintegrin and metalloprotease 17 (ADAM17) is a membrane-tethered protease that triggers multiple signaling pathways. It releases active forms of the primary inflammatory cytokine tumor necrosis factor (TNF) and cancer-implicated epidermal growth factor (EGF) family growth factors. iRhom2, a rhomboid-like, membrane-embedded pseudoprotease, is an essential cofactor of ADAM17. Here, we present cryoelectron microscopy (cryo-EM) structures of the human ADAM17/iRhom2 complex in both inactive and active states. These reveal three regulatory mechanisms. First, exploiting the rhomboid-like hallmark of TMD recognition, iRhom2 interacts with the ADAM17 TMD to promote ADAM17 trafficking and enzyme maturation. Second, a unique iRhom2 extracellular domain unexpectedly retains the cleaved ADAM17 inhibitory prodomain, safeguarding against premature activation and dysregulated proteolysis. Finally, loss of the prodomain from the complex mobilizes the ADAM17 protease domain, contributing to its ability to engage substrates. Our results reveal how a rhomboid-like pseudoprotease has been repurposed during evolution to regulate a potent membrane-tethered enzyme, ADAM17, ensuring the fidelity of inflammatory and growth factor signaling.


Subject(s)
ADAM17 Protein , Cryoelectron Microscopy , Signal Transduction , ADAM17 Protein/metabolism , ADAM17 Protein/genetics , Humans , HEK293 Cells , Carrier Proteins/metabolism , Carrier Proteins/genetics , Inflammation/metabolism , Inflammation/genetics , Proteolysis , Tumor Necrosis Factor-alpha/metabolism , Tumor Necrosis Factor-alpha/genetics , Protein Domains , Protein Binding , Intercellular Signaling Peptides and Proteins/metabolism , Intercellular Signaling Peptides and Proteins/genetics , Epidermal Growth Factor/metabolism , Epidermal Growth Factor/genetics , Intracellular Signaling Peptides and Proteins
10.
Bioconjug Chem ; 35(6): 750-757, 2024 Jun 19.
Article in English | MEDLINE | ID: mdl-38815180

ABSTRACT

Site-specific installation of non-natural functionality onto proteins has enabled countless applications in biotechnology, chemical biology, and biomaterials science. Though the N-terminus is an attractive derivatization location, prior methodologies targeting this site have suffered from low selectivity, a limited selection of potential chemical modifications, and/or challenges associated with divergent protein purification/modification steps. In this work, we harness the atypically split VidaL intein to simultaneously N-functionalize and purify homogeneous protein populations in a single step. Our method─referred to as VidaL-tagged expression and protein ligation (VEPL)─enables modular and scalable production of N-terminally modified proteins with native bioactivity. Demonstrating its flexibility and ease of use, we employ VEPL to combinatorially install 4 distinct (multi)functional handles (e.g., biotin, alkyne, fluorophores) to the N-terminus of 4 proteins that span three different classes: fluorescent (Enhanced Green Fluorescent Protein, mCherry), enzymatic (ß-lactamase), and growth factor (epidermal growth factor). Moving forward, we anticipate that VEPL's ability to rapidly generate and isolate N-modified proteins will prove useful across the growing fields of applied chemical biology.


Subject(s)
Inteins , Green Fluorescent Proteins/chemistry , Green Fluorescent Proteins/metabolism , Green Fluorescent Proteins/genetics , beta-Lactamases/metabolism , beta-Lactamases/chemistry , Luminescent Proteins/chemistry , Epidermal Growth Factor/metabolism , Epidermal Growth Factor/chemistry , Red Fluorescent Protein , Proteins/chemistry
11.
Pigment Cell Melanoma Res ; 37(4): 514-529, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38705722

ABSTRACT

Epidermal melanin unit integrity is crucial for skin homeostasis and pigmentation. Epidermal growth factor (EGF) receptor (EGFR) is a pivotal player in cell growth, wound healing, and maintaining skin homeostasis. However, its influence on skin pigmentation is relatively unexplored. This study investigates the impact and underlying mechanisms of EGFR inhibitors on skin pigmentation. We evaluated EGF and EGFR expression in various skin cells using quantitative real-time PCR, Western blot, and immunofluorescence. EGF and EGFR were predominantly expressed in epidermal keratinocytes, and treatment with the EGFR tyrosine kinase inhibitors (EGFR-TKIs) gefitinib and PD153035 significantly increased stem cell factor (SCF) and endothelin-1 (ET-1) expression in cultured keratinocytes. Enhanced melanocyte migration and proliferation were observed in co-culture, as evidenced by time-lapse live imaging and single-cell tracking assays. Furthermore, topical application of gefitinib to guinea pig dorsal skin induced increased pigmentation and demonstrated efficacy in mitigating rhododendrol-induced leukoderma. Suppression of EGF signaling indirectly enhanced skin pigmentation by upregulating SCF and ET-1 in epidermal keratinocytes. This novel mechanism highlights the pivotal role of EGF signaling in regulating skin pigmentation, and topical EGFR-TKI therapy at an appropriate dose may be a promising approach for depigmentation disorder management.


Subject(s)
ErbB Receptors , Gefitinib , Hypopigmentation , Keratinocytes , Melanins , Melanocytes , Protein Kinase Inhibitors , ErbB Receptors/metabolism , Animals , Melanins/metabolism , Melanins/biosynthesis , Humans , Protein Kinase Inhibitors/pharmacology , Melanocytes/drug effects , Melanocytes/metabolism , Melanocytes/pathology , Keratinocytes/drug effects , Keratinocytes/metabolism , Keratinocytes/pathology , Hypopigmentation/pathology , Hypopigmentation/drug therapy , Gefitinib/pharmacology , Guinea Pigs , Skin Pigmentation/drug effects , Stem Cell Factor/metabolism , Epidermis/drug effects , Epidermis/pathology , Epidermis/metabolism , Cell Proliferation/drug effects , Cell Movement/drug effects , Endothelin-1/metabolism , Epidermal Growth Factor/metabolism , Epidermal Growth Factor/pharmacology , Quinazolines
12.
Int J Biol Macromol ; 270(Pt 1): 132365, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38750850

ABSTRACT

This study strategically incorporates epidermal growth factor (EGF) and keratinocyte growth factor (KGF) within a hyaluronic acid (HA) hydrogel to enhance corneal wound healing. The controlled release of EGF and KGF from the HA hydrogel is engineered to promote the regeneration of both the epithelial and stromal layers. Specifically, EGF plays a pivotal role in the regeneration of the epithelial layer, while KGF exhibits efficacy in the regeneration of the stromal layer. The combination of these growth factors facilitates efficient regeneration of each layer and demonstrates the capability to modulate each other's regenerative effects. The interplay between EGF and KGF provides an understanding of their cooperative influence on the dynamics of corneal wound healing. The results of this study contribute to the development of advanced strategies for corneal wound management and offer insights into the complex process of corneal regeneration.


Subject(s)
Epidermal Growth Factor , Fibroblast Growth Factor 7 , Hyaluronic Acid , Hydrogels , Wound Healing , Hyaluronic Acid/chemistry , Hyaluronic Acid/pharmacology , Epidermal Growth Factor/pharmacology , Wound Healing/drug effects , Hydrogels/chemistry , Hydrogels/pharmacology , Animals , Humans , Cornea/drug effects , Cornea/metabolism , Corneal Injuries/drug therapy , Corneal Injuries/metabolism , Rabbits
13.
Arch Oral Biol ; 164: 105989, 2024 Aug.
Article in English | MEDLINE | ID: mdl-38718466

ABSTRACT

OBJECTIVE: To investigate the expression and secretion of epidermal growth factor (EGF) in major and minor salivary gland tissues of human subjects and to examine the potential influence of sex and age on EGF expression and secretion. DESIGN: Saliva samples from the oral cavity at rest and after citric acid stimulation, as well as serum samples, were collected from 150 healthy subjects, and the concentrations of EGF were measured with enzyme-linked immunosorbent assay (ELISA) and compared. The expression of EGF mRNA and protein in normal salivary gland tissues was measured by real-time polymerase chain reaction (RT-PCR), Western blot (WB), and immunohistochemistry (IHC). RESULTS: The EGF concentration in acid-stimulated saliva was significantly higher than that in resting saliva (P < 0.001), and significantly higher than that in serum (P < 0.001). No sex difference was observed in EGF levels of whole saliva and serum, whereas the EGF levels in saliva and serum were decreased with age (P < 0.001 and P < 0.001, respectively). The EGF concentration and compound secretion rate (CSR) in resting submandibular glands saliva were significantly higher than those in resting parotid glands saliva (P = 0.002 and P < 0.001, respectively). The EGF was expressed in all major and minor salivary glands and ranked in order of submandibular, parotid, sublingual, and labial glands. CONCLUSION: All salivary glands have the function of secreting EGF, and the submandibular gland is the main source of salivary EGF. Aging is a factor influencing the expression and secretion of EGF.


Subject(s)
Blotting, Western , Enzyme-Linked Immunosorbent Assay , Epidermal Growth Factor , Immunohistochemistry , Real-Time Polymerase Chain Reaction , Saliva , Salivary Glands , Humans , Female , Male , Epidermal Growth Factor/metabolism , Adult , Saliva/metabolism , Saliva/chemistry , Middle Aged , Salivary Glands/metabolism , Sex Factors , Aged , Age Factors , RNA, Messenger/metabolism , Adolescent , Submandibular Gland/metabolism , Salivary Glands, Minor/metabolism , Citric Acid/metabolism
14.
Cell Mol Biol (Noisy-le-grand) ; 70(5): 289-294, 2024 May 27.
Article in English | MEDLINE | ID: mdl-38814200

ABSTRACT

Collagen sponge and epidermal growth factor (EGF) promote wound healing. However, the effect of collagen sponge combined with EGF in repairing maxillofacial head and neck wounds remains unclear. The rats were divided into 3 groups, including experimental group 1 (Vaseline gauze+EGF), experimental group 2 (collagen sponge+EGF) with control group (Vaseline+normal saline), and maxillofacial head and neck wounds were simulated. Wound pathological morphology was detected by HE staining; wound EGF, IL-1ß, IL-6 along with TNF-α contents by ELISA and MMP1 level by western blot. At 7 and 14 days after treatment, wound healing rate of two experimental groups was higher than that of control group, and that of experimental group 2 presented higher than that of experimental group 1. Compared with control group, experimental group 1 had significantly fewer inflammatory cells in the wound tissue, local erythrocyte spillage outside the vascular walls, more collagen deposition and more granulation tissue. Compared with experimental group 1, inflammatory cells in wound tissues of experimental group 2 were significantly reduced, the collagen tissues were visible and arranged, and the growth of the wound granulation tissue was obvious. IL-1ß, IL-6 along with TNF-α levels in two experimental groups presented lower than control group, and EGF level was higher. More importantly, in contrast to experimental group 1, IL-1ß, IL-6 along with TNF-α in experimental group 2 presented lower, and EGF level presented higher. At 14 days after treatment, MMP1 level in two experimental groups was lower than control group. In contrast to experimental group 1, MMP1 level in experimental group 2 was lower. In summary, collagen sponge combined with EGF for the first time significantly improved the healing speed of maxillofacial head and neck wounds and reduced the scar left after wound healing.


Subject(s)
Collagen , Epidermal Growth Factor , Matrix Metalloproteinase 1 , Rats, Sprague-Dawley , Tumor Necrosis Factor-alpha , Wound Healing , Animals , Epidermal Growth Factor/pharmacology , Epidermal Growth Factor/metabolism , Wound Healing/drug effects , Collagen/metabolism , Matrix Metalloproteinase 1/metabolism , Male , Tumor Necrosis Factor-alpha/metabolism , Interleukin-6/metabolism , Rats , Interleukin-1beta/metabolism , Granulation Tissue/drug effects , Granulation Tissue/pathology
15.
Mol Biol Rep ; 51(1): 633, 2024 May 09.
Article in English | MEDLINE | ID: mdl-38724835

ABSTRACT

BACKGROUND: Radiation therapy is utilized for treatment of localized prostate cancer. Nevertheless, cancerous cells frequently develop radiation resistance. While higher radiation doses have not always been effective, radiosensitizers have been extensively studied for their ability to enhance the cytotoxic effects of radiation. So, this study aims to evaluate the possible radiosensitization effects of docetaxel (DTX) and silver nanoparticles (SNP) in LNCaP cells. METHODS: The cytotoxic effects of DTX, SNP and 2 Gy of X-Ray radiation treatments were assessed in human LNCaP cell line using the MTT test after 24 h. Moreover, the effects of DTX, SNP and radiation on Epidermal growth factor (EGF), Caspase 3, inducible nitric oxide synthase and E-cadherin gene expression were analyzed using the Real-time PCR method. The level of Hydrogen peroxide (H2O2), an oxidative stress marker, was also detected 24 h after various single and combined treatments. RESULTS: The combinations of SNP (in low toxic concentration) and/or DTX (0.25× IC50 and 0.5 × IC50 concentrations for triple and double combinations respectively) with radiation induced significant cytotoxicity in LNCaP cells in comparison to monotherapies. These cytotoxic effects were associated with the downregulation of EGF mRNA. Additionally, H2O2 levels increased after Radiation + SNP + DTX triple combination and double combinations including Radiation + SNP and Radiation + DTX versus single treatments. The triple combination treatment also increased Caspase 3 and and E-cadherin mRNA levels in compared to single treatments in LNCaP cells. CONCLUSION: Our results indicate that the combination of SNP and DTX with radiation induces significant anti-cancer effects. Upregulation of Caspase 3 and E-cadherin gene expression, and decreased mRNA expression level of EGF may be exerted specifically by use of this combination versus single treatments.


Subject(s)
Docetaxel , Metal Nanoparticles , Prostatic Neoplasms , Radiation-Sensitizing Agents , Silver , Humans , Docetaxel/pharmacology , Male , Silver/pharmacology , Prostatic Neoplasms/radiotherapy , Prostatic Neoplasms/drug therapy , Prostatic Neoplasms/metabolism , Prostatic Neoplasms/genetics , Cell Line, Tumor , Radiation-Sensitizing Agents/pharmacology , Oxidative Stress/drug effects , Oxidative Stress/radiation effects , Hydrogen Peroxide/pharmacology , Cell Survival/drug effects , Cell Survival/radiation effects , Caspase 3/metabolism , Caspase 3/genetics , Antineoplastic Agents/pharmacology , Epidermal Growth Factor/metabolism , Epidermal Growth Factor/pharmacology , Gene Expression Regulation, Neoplastic/drug effects , Gene Expression Regulation, Neoplastic/radiation effects , Apoptosis/drug effects , Apoptosis/radiation effects , Cadherins/metabolism , Cadherins/genetics
16.
J Ovarian Res ; 17(1): 97, 2024 May 08.
Article in English | MEDLINE | ID: mdl-38720330

ABSTRACT

The epidermal growth factor (EGF)-like factors, comprising amphiregulin (AREG), betacellulin (BTC), and epiregulin (EREG), play a critical role in regulating the ovulatory process. Pentraxin 3 (PTX3), an essential ovulatory protein, is necessary for maintaining extracellular matrix (ECM) stability during cumulus expansion. The aim of this study was to investigate the impact of EGF-like factors, AREG, BTC, and EREG on the expression and production of PTX3 in human granulosa-lutein (hGL) cells and the molecular mechanisms involved. Our results demonstrated that AREG, BTC, and EREG could regulate follicular function by upregulating the expression and increasing the production of PTX3 in both primary (obtained from 20 consenting patients undergoing IVF treatment) and immortalized hGL cells. The upregulation of PTX3 expression was primarily facilitated by the activation of the extracellular signal-regulated kinase 1 and 2 (ERK1/2) signaling pathway, induced by these EGF-like factors. In addition, we found that the upregulation of PTX3 expression triggered by the EGF-like factors was completely reversed by either pretreatment with the epidermal growth factor receptor (EGFR) inhibitor, AG1478, or knockdown of EGFR, suggesting that EGFR is crucial for activating the ERK1/2 signaling pathway in hGL cells. Overall, our findings indicate that AREG, BTC, and EREG may modulate human cumulus expansion during the periovulatory stage through the upregulation of PTX3.


Subject(s)
Amphiregulin , Betacellulin , C-Reactive Protein , Epiregulin , Luteal Cells , Serum Amyloid P-Component , Up-Regulation , Female , Humans , Amphiregulin/metabolism , Amphiregulin/genetics , Betacellulin/metabolism , C-Reactive Protein/metabolism , C-Reactive Protein/genetics , Epidermal Growth Factor/metabolism , Epidermal Growth Factor/pharmacology , Epiregulin/metabolism , Epiregulin/genetics , ErbB Receptors/metabolism , Luteal Cells/metabolism , MAP Kinase Signaling System , Serum Amyloid P-Component/metabolism , Serum Amyloid P-Component/genetics
17.
BMC Oral Health ; 24(1): 527, 2024 May 03.
Article in English | MEDLINE | ID: mdl-38702671

ABSTRACT

BACKGROUND: This study aimed to assess and compare the concentrations of growth factors, white blood cells (WBCs), and platelets in injectable platelet-rich fibrin (i-PRF) derived from people with healthy periodontal conditions and those with chronic periodontitis. METHODS: Venous blood samples were obtained from 30 patients diagnosed with chronic periodontitis (test group) and 30 participants with healthy periodontal conditions (control group). The i-PRF was then acquired from centrifuged blood. The growth factors (VEGF, IGF-1, TGF-ß1, PDGF-BB and EGF) released from the i-PRF samples were compared between groups with ELISA testing. The amounts of WBCs and platelets were also compared. RESULTS: No significant differences in the concentrations of growth factors were found between the groups (the mean values for the control and test groups were, respectively: IGF: 38.82, 42.46; PDGF: 414.25, 466.28; VEGF: 375.69, 412.18; TGF-ß1: 21.50, 26.21; EGF: 138.62, 154.82). The test group exhibited a significantly higher WBC count than the control group (8.80 vs. 6.60, respectively). However, the platelet count did not show a statistically significant difference between the groups (control group 242.0 vs. test group 262.50). No significant correlation was observed between WBC count and growth factor level in either group. CONCLUSIONS: The growth factor levels in i-PRFs did not exhibit significant difference between the two groups. This suggests that the levels of these growth factors may be unaffected by the periodontal disease.


Subject(s)
Chronic Periodontitis , Insulin-Like Growth Factor I , Intercellular Signaling Peptides and Proteins , Platelet-Rich Fibrin , Transforming Growth Factor beta1 , Vascular Endothelial Growth Factor A , Humans , Chronic Periodontitis/blood , Pilot Projects , Male , Female , Adult , Middle Aged , Vascular Endothelial Growth Factor A/blood , Insulin-Like Growth Factor I/analysis , Intercellular Signaling Peptides and Proteins/blood , Intercellular Signaling Peptides and Proteins/analysis , Transforming Growth Factor beta1/blood , Epidermal Growth Factor/blood , Epidermal Growth Factor/analysis , Leukocyte Count , Becaplermin/blood , Case-Control Studies , Blood Platelets/metabolism , Injections
18.
Methods Mol Biol ; 2800: 75-87, 2024.
Article in English | MEDLINE | ID: mdl-38709479

ABSTRACT

Enzymatic ascorbate peroxidase (APEX) tagging allows for high-resolution, three-dimensional protein distribution analyses in cells and tissues. This chapter describes the application of APEX-tagging to visualize the trafficking of the epidermal growth factor receptor (EGFR) during epidermal growth factor-mediated receptor activation. Here, we describe the preparation of cells, methods to validate the stimulation of the EGFR, and visualization of the APEX-resolved distribution of the EGFR in the transmission electron microscope.


Subject(s)
ErbB Receptors , Microscopy, Electron, Transmission , Protein Transport , Humans , Ascorbate Peroxidases/chemistry , Ascorbate Peroxidases/metabolism , Epidermal Growth Factor/metabolism , ErbB Receptors/metabolism , Microscopy, Electron, Transmission/methods
19.
Cell Mol Biol (Noisy-le-grand) ; 70(5): 243-247, 2024 May 27.
Article in English | MEDLINE | ID: mdl-38814208

ABSTRACT

Burns are the most severe type of trauma, and the resulting ischemia and hypoxia damage can promote the dysfunction and even failure of tissues and organs throughout the body, endangering patients' life safety. Recombinant human growth hormone (rhGH) has the functions of promoting protein synthesis to reverse negative nitrogen balance, accelerating wound healing, and improving immune function, which is widely used in the treatment of burns. However, the exact mechanism and pathway of rhGH's action is not yet fully understood. In this study, we observed the wound repair effect of recombinant human growth hormone (rhGH) on burned mice and further analyzed the mechanism of action, which can provide more comprehensive reference opinions for clinical practice. First, by establishing a burn mouse model and and intervening with different doses of rhGH, we found that the wound healing capacity of mice was significantly enhanced and the inflammatory and oxidative stress responses were obviously alleviated, confirming the excellent promotion of wound repair and anti-inflammatory and antioxidant effects of rhGH. Subsequently, we found that the expression of p-ERK1/2/ERK1/2, EGF, TGF-ß, and VEGF proteins was elevated in the traumatic tissues of mice after rhGH intervention, suggesting that the pathway of action of rhGH might be related to the activation of ERK pathway to promote the regeneration of traumatic capillaries.


Subject(s)
Burns , Human Growth Hormone , MAP Kinase Signaling System , Neovascularization, Physiologic , Recombinant Proteins , Wound Healing , Animals , Burns/drug therapy , Burns/pathology , Wound Healing/drug effects , MAP Kinase Signaling System/drug effects , Recombinant Proteins/pharmacology , Mice , Human Growth Hormone/pharmacology , Humans , Neovascularization, Physiologic/drug effects , Male , Oxidative Stress/drug effects , Disease Models, Animal , Vascular Endothelial Growth Factor A/metabolism , Transforming Growth Factor beta/metabolism , Mice, Inbred C57BL , Epidermal Growth Factor/pharmacology , Angiogenesis
20.
Biosensors (Basel) ; 14(5)2024 Apr 23.
Article in English | MEDLINE | ID: mdl-38785686

ABSTRACT

Combinatorial drug therapy has emerged as a critically important strategy in medical research and patient treatment and involves the use of multiple drugs in concert to achieve a synergistic effect. This approach can enhance therapeutic efficacy while simultaneously mitigating adverse side effects. However, the process of identifying optimal drug combinations, including their compositions and dosages, is often a complex, costly, and time-intensive endeavor. To surmount these hurdles, we propose a novel microfluidic device capable of simultaneously generating multiple drug concentration gradients across an interlinked array of culture chambers. This innovative setup allows for the real-time monitoring of live cell responses. With minimal effort, researchers can now explore the concentration-dependent effects of single-agent and combination drug therapies. Taking neural stem cells (NSCs) as a case study, we examined the impacts of various growth factors-epithelial growth factor (EGF), platelet-derived growth factor (PDGF), and fibroblast growth factor (FGF)-on the differentiation of NSCs. Our findings indicate that an overdose of any single growth factor leads to an upsurge in the proportion of differentiated NSCs. Interestingly, the regulatory effects of these growth factors can be modulated by the introduction of additional growth factors, whether singly or in combination. Notably, a reduced concentration of these additional factors resulted in a decreased number of differentiated NSCs. Our results affirm that the successful application of this microfluidic device for the generation of multi-drug concentration gradients has substantial potential to revolutionize drug combination screening. This advancement promises to streamline the process and accelerate the discovery of effective therapeutic drug combinations.


Subject(s)
High-Throughput Screening Assays , Neural Stem Cells , Neural Stem Cells/drug effects , Humans , Cell Differentiation , Lab-On-A-Chip Devices , Platelet-Derived Growth Factor , Epidermal Growth Factor , Drug Evaluation, Preclinical , Drug Combinations , Fibroblast Growth Factors
SELECTION OF CITATIONS
SEARCH DETAIL
...