Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 5.187
Filter
1.
Sci Rep ; 14(1): 22778, 2024 10 01.
Article in English | MEDLINE | ID: mdl-39354061

ABSTRACT

Histopathologically, oral squamous cell carcinoma (OSCC) consists of well-defined interfaces with adjacent non-cancerous epithelium. Previously, we found that SCC tissues expressed higher levels of specific proteins at this interface. Ladinin-1 (LAD1) is one of the specific molecules that has increased expressions in cancer fronts; however, its function in OSCC is unknown. Therefore, this study aimed to elucidate the function of LAD1 in human OSCC cells. LAD1 was localized on the actin arc at the distal periphery of cell clusters in the OSCC cell lines HSC-2, HSC-3, and HSC-4. When LAD1 was knocked down, cellular migration was repressed in wound scratch assays but was reversed in three-dimensional collagen gel invasion assays. Characteristic LAD1 localization along actin arcs forming the leading edge of migrating cells was diminished with loss of filopodia formation and ruffling in knockdown cells, in which the expression levels of cell motility-related genes-p21-activated kinase 1 (PAK1) and caveolin-1 (CAV1)-were upregulated and downregulated, respectively. LAD1 expression was also associated with the downregulation of vimentin and increased histological differentiation of OSCC. These results suggest that LAD1 is involved in actin dynamics during filopodia and lamellipodia formation, and in maintaining the epithelial phenotype of OSCC cells.


Subject(s)
Actins , Carcinoma, Squamous Cell , Cell Movement , Mouth Neoplasms , Humans , Actins/metabolism , Carcinoma, Squamous Cell/metabolism , Carcinoma, Squamous Cell/pathology , Carcinoma, Squamous Cell/genetics , Caveolin 1/metabolism , Caveolin 1/genetics , Cell Line, Tumor , Epithelial Cells/metabolism , Epithelial Cells/pathology , Gene Expression Regulation, Neoplastic , Mouth Neoplasms/pathology , Mouth Neoplasms/metabolism , Mouth Neoplasms/genetics , p21-Activated Kinases/metabolism , p21-Activated Kinases/genetics , Phenotype , Vimentin/metabolism , Vimentin/genetics
2.
Sci Rep ; 14(1): 20553, 2024 09 04.
Article in English | MEDLINE | ID: mdl-39232048

ABSTRACT

The plasma membrane protein caveolin-1 (CAV-1) regulates signaling by inhibiting a wide range of kinases and other enzymes. Our previous study demonstrated that the downregulation of CAV-1 in psoriatic epidermal cells contributes to inflammation by enhancing JAK/STAT signaling, cell proliferation, and chemokine production. Administration of the CAV-1 scaffolding domain (CSD) peptide suppressed imiquimod (IMQ)-induced psoriasis-like dermatitis. To identify an optimal therapeutic peptide derived from CAV-1, we have compared the efficacy of CSD and subregions of CSD that have been modified to make them water soluble. We refer to these modified peptides as sCSD, sA, sB, and sC. In IMQ-induced psoriasis-like dermatitis, while all four peptides showed major beneficial effects, sB caused the most significant improvements of skin phenotype and number of infiltrating cells, comparable or superior to the effects of sCSD. Phosphorylation of STAT3 was also inhibited by sB. Furthermore, sB suppressed angiogenesis both in vivo in the dermis of IMQ-induced psoriasis mice and in vitro by blocking the ability of conditioned media derived from CAV-1-silenced keratinocytes to inhibit tube formation by HUVEC. In conclusion, sB had similar or greater beneficial effects than sCSD not only by cytokine suppression but by angiogenesis inhibition adding to its ability to target psoriatic inflammation.


Subject(s)
Caveolin 1 , Cytokines , Imiquimod , Neovascularization, Pathologic , Psoriasis , STAT3 Transcription Factor , Psoriasis/drug therapy , Psoriasis/chemically induced , Psoriasis/pathology , Psoriasis/metabolism , Caveolin 1/metabolism , Animals , Mice , Cytokines/metabolism , Humans , STAT3 Transcription Factor/metabolism , Neovascularization, Pathologic/drug therapy , Neovascularization, Pathologic/metabolism , Peptides/pharmacology , Peptides/chemistry , Skin/drug effects , Skin/metabolism , Skin/pathology , Keratinocytes/drug effects , Keratinocytes/metabolism , Disease Models, Animal , Water/chemistry , Solubility , Human Umbilical Vein Endothelial Cells/drug effects , Angiogenesis
3.
Medicine (Baltimore) ; 103(22): e38367, 2024 May 31.
Article in English | MEDLINE | ID: mdl-39259075

ABSTRACT

This study aimed to decipher the interaction between CD26 and caveolin-1, key proteins involved in cell signaling and linked to various diseases. Using computational methods, we predicted their binding conformations and assessed stability through 100 ns molecular dynamics (MD) simulations. We identified two distinct binding conformations (con1 and con4), with con1 exhibiting superior stability. In con1, specific amino acids in CD26, namely GLU237, TYR241, TYR248, and ARG147, were observed to engage in interactions with the F-J chain of Caveolin-1, establishing hydrogen bonds and cation or π-π interactions. Meanwhile, in con4, CD26 amino acids ARG253, LYS250, and TYR248 interacted with the J chain of Caveolin-1 via hydrogen bonds, cation-π interactions, and π-π interactions. Virtual screening also revealed potential small-molecule modulators, including Crocin, Poliumoside, and Canagliflozin, that could impact this interaction. Additionally, predictive analyses were conducted on the potential bioactivity, drug-likeness, and ADMET properties of these three compounds. These findings offer valuable insights into the binding mechanism, paving the way for new therapeutic strategies. However, further validation is required before clinical application. In summary, we provide a detailed understanding of the CD26 and caveolin-1 interaction, identifying key amino acids and potential modulators, essential for developing targeted therapies.


Subject(s)
Amino Acids , Caveolin 1 , Dipeptidyl Peptidase 4 , Molecular Dynamics Simulation , Humans , Amino Acids/metabolism , Caveolin 1/metabolism , Dipeptidyl Peptidase 4/metabolism , Hydrogen Bonding , Protein Binding , Protein Conformation
4.
Int J Nanomedicine ; 19: 9091-9107, 2024.
Article in English | MEDLINE | ID: mdl-39258003

ABSTRACT

Purpose: Castration Resistant Prostate Cancer (CRPC) is characterized by poor prognosis and limited therapeutic options. AgNPs functionalized with glucose (G-AgNPs) were observed cytotoxic to CRPC cell lines (PC-3 and Du-145) and not LNCaP. This study aims to evaluate AgNPs and G-AgNPs' uptake mechanisms in these cells and understand their role in the selective effect against CRPC cells. Methods: Uptake of AgNPs and G-AgNPs was assessed through transmission electron microscopy (TEM). A microRNA (miRNAs) analysis approach was used to uncover the main molecular differences responsible for the endocytic mechanisms' regulation. Caveolin (Cav) 1 and 2 mRNA and protein levels were assessed in the three cell lines. Caveolae-dependent endocytosis was inhibited with genistein or siCav1- and siCav2- in PC-3 and Du-145 and resazurin assay was used to evaluate viability after AgNPs and G-AgNPs administration. Caveolae-dependent endocytosis was induced with Cav1+ and Cav2+ plasmids in LNCaP, resazurin assay was used to evaluate viability after AgNPs and G-AgNPs administration and TEM to assess their location. Results: AgNPs and G-AgNPs were not uptaked by LNCaP. miRNA analysis revealed 37 upregulated and 90 downregulated miRNAs. Functional enrichment analysis of miRNAs' targets resulted in enrichment of terms related to endocytosis and caveolae. We observed that Cav1 and Cav2 are not expressed in LNCaP. Inhibiting caveolae-dependent endocytosis in Du-145 and PC-3 led to a significative reduction of cytotoxic capacity of AgNPs and G-AgNPs and induction of caveolae-dependent endocytosis in LNCaP lead to a significative increase as well as their uptake by cells. Conclusion: This study shows the potential of these AgNPs as a new therapeutic approach directed to CRPC patients, uncovers caveolae-dependent endocytosis as the uptake mechanism of these AgNPs and highlights deregulation of Cav1 and Cav2 expression as a key difference in hormone sensitive and resistant PCa cells which may be responsible for drug resistance.


Subject(s)
Caveolae , Caveolin 1 , Endocytosis , Metal Nanoparticles , MicroRNAs , Prostatic Neoplasms, Castration-Resistant , Silver , Male , Humans , Endocytosis/drug effects , Endocytosis/physiology , Prostatic Neoplasms, Castration-Resistant/metabolism , Caveolae/metabolism , Caveolae/drug effects , Silver/chemistry , Silver/pharmacology , Silver/pharmacokinetics , Caveolin 1/metabolism , Caveolin 1/genetics , Metal Nanoparticles/chemistry , Cell Line, Tumor , MicroRNAs/metabolism , MicroRNAs/genetics , Cell Survival/drug effects , Caveolin 2/metabolism , Caveolin 2/genetics , Antineoplastic Agents/pharmacology , PC-3 Cells
5.
Turk Neurosurg ; 34(5): 833-839, 2024.
Article in English | MEDLINE | ID: mdl-39087290

ABSTRACT

AIM: To observe changes in the serum levels of visinin-like protein-1 (VILIP-1), caveolin-1 (Cav-1) and neuron-specific enolase (NSE) after glioma resection. MATERIAL AND METHODS: Consecutive 14 glioma patients with different histologic grade and 14 age and gender-matched healthy subjects were included in this pilot study. From the patients serum samples were taken in preoperative and on day 2 and 10 of postoperative periods. Healthy subjects provided serum sample once. The serum changes of three proteins were evaluated by ELISA. The results were compared between preoperative and postoperative periods and between patients and controls. RESULTS: Preoperative serum levels of VILIP-1 (p=0.008) and Cav-1 (p=0.012) were significantly higher in the patients. Mean serum levels of VILIP-1 (p=0.002) and Cav-1 (p=0.013) again were significantly higher than those of the controls. NSE did not show significant changes compared to controls in none of the periods. There was a steady decline regarding all three molecules from preoperative to postoperative day 10. However, statistical comparisons did not reveal any significant difference with respect the decline in any molecule. Significant positive correlation was detected between preoperative serum levels of VILIP-1 and Cav-1 (p=0.00001) in the patients and the controls (p=0.0000). CONCLUSION: This pilot study suggested that Cav-1 and particularly VILIP-1 may be used as a valuable serum biomarker for follow-up and for early detection of recurrence in high-grade gliomas. Future studies including larger cohort of patients with homogeneous group of glioma is required.


Subject(s)
Caveolin 1 , Glioma , Neurocalcin , Phosphopyruvate Hydratase , Supratentorial Neoplasms , Humans , Caveolin 1/blood , Phosphopyruvate Hydratase/blood , Pilot Projects , Male , Glioma/surgery , Glioma/blood , Female , Middle Aged , Adult , Supratentorial Neoplasms/surgery , Supratentorial Neoplasms/blood , Neurocalcin/blood , Biomarkers, Tumor/blood , Aged , Postoperative Period
6.
Pathol Res Pract ; 262: 155552, 2024 Oct.
Article in English | MEDLINE | ID: mdl-39180803

ABSTRACT

Glaucoma is a degenerative disease characterized by retinal ganglion cell (RGC) death and visual impairment caused by elevated intraocular pressure (IOP). Elevated IOP can activate microglia, which participate in ganglion cell injury. Based on the study of caveolin-1 (Cav-1) in glaucoma, we aimed to explore the effect and mechanism of Cav-1 on RGC apoptosis in mice with acute ocular hypertension (AOH). AOH mice were established, and Cav-1 was intravitreally injected. Retinal microglia and RGCs were isolated from neonatal mice. TUNEL staining, hematoxylin-eosin staining, immunohistochemistry, flow cytometry, PCR and western blotting were used to observe the effect of Cav-1 on RGCs and mouse retinas. The thickness of the whole retina and the inner retinal sublayer decreased significantly, retinal cell apoptosis increased after AOH injury, and Cav-1 treatment reversed the effect of AOH injury. In addition, Cav-1 treatment promoted the conversion of proinflammatory M1 microglia to anti-inflammatory M2 microglia. Microglia and RGCs were isolated from neonatal mice. Cav-1 protects RGCs from OGD/R-induced injury by changing the polarization status of retinal microglia in vitro. Further studies revealed that Cav-1 activated the Akt/PTEN signaling pathway and inhibited TLR4. Our study provides evidence that Cav-1 may be a promising therapeutic target for glaucoma.


Subject(s)
Caveolin 1 , Glaucoma , PTEN Phosphohydrolase , Proto-Oncogene Proteins c-akt , Retinal Ganglion Cells , Signal Transduction , Toll-Like Receptor 4 , Animals , Retinal Ganglion Cells/pathology , Retinal Ganglion Cells/metabolism , Retinal Ganglion Cells/drug effects , Caveolin 1/metabolism , Signal Transduction/physiology , PTEN Phosphohydrolase/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Mice , Glaucoma/metabolism , Glaucoma/pathology , Toll-Like Receptor 4/metabolism , Mice, Inbred C57BL , Apoptosis/drug effects , Microglia/metabolism , Microglia/pathology , Disease Models, Animal
7.
Oncogene ; 43(37): 2768-2780, 2024 Sep.
Article in English | MEDLINE | ID: mdl-39138263

ABSTRACT

Peritoneal metastasis is one of the most common risk factors contributing to the poor prognosis of gastric cancer. We previously reported that extracellular vesicles from gastric cancer cells could facilitate peritoneal metastasis. However, their impact on gastric cancer-induced peritoneal metastasis under hypoxic conditions remains unclear. This study aims to elucidate how hypoxia-resistant gastric cancer cell-derived extracellular vesicles affect the peritoneal metastasis of normoxic gastric cancer cells. Proteomic analysis revealed elevated levels of Caveolin1 and Laminin ß2 in hypoxia-resistant gastric cancer cells and their corresponding extracellular vesicles. Importantly, Caveolin1 was found to play a central role in mediating Laminin ß2 sorting into extracellular vesicles derived from hypoxia-resistant gastric cancer cells, and subsequently, extracellular vesicle-associated Laminin ß2 promoted peritoneal metastasis in normoxic gastric cancer cells by activating the AKT pathway. Further investigation confirmed that Caveolin1 activation by Rho-related Coiled-coil kinase 1-mediated phosphorylation of Y14 residue is a key factor facilitating Laminin ß2 sorting into extracellular vesicles. Moreover, Y14 phosphorylated- Caveolin1 enhanced Laminin ß2 sorting by activating Rab11. Finally, our study demonstrated that a combined assessment of plasma extracellular vesicle-associated Caveolin1 and extracellular vesicle-associated Laminin ß2 could provide an accurate predictive tool for peritoneal metastasis occurrence in gastric cancer.


Subject(s)
Caveolin 1 , Extracellular Vesicles , Peritoneal Neoplasms , Stomach Neoplasms , rab GTP-Binding Proteins , rho-Associated Kinases , Stomach Neoplasms/pathology , Stomach Neoplasms/metabolism , Stomach Neoplasms/genetics , Humans , Caveolin 1/metabolism , Caveolin 1/genetics , Peritoneal Neoplasms/secondary , Peritoneal Neoplasms/metabolism , Animals , rho-Associated Kinases/metabolism , Extracellular Vesicles/metabolism , Mice , rab GTP-Binding Proteins/metabolism , rab GTP-Binding Proteins/genetics , Cell Line, Tumor , Signal Transduction , Male , Female
8.
J Biol Chem ; 300(9): 107705, 2024 Sep.
Article in English | MEDLINE | ID: mdl-39178948

ABSTRACT

The cell signaling molecules nitric oxide (NO) and Ca2+ regulate diverse biological processes through their closely coordinated activities directed by signaling protein complexes. However, it remains unclear how dynamically the multicomponent protein assemblies behave within the signaling complexes upon the interplay between NO and Ca2+ signals. Here we demonstrate that TRPC5 channels activated by the stimulation of G-protein-coupled ATP receptors mediate Ca2+ influx, that triggers NO production from endothelial NO synthase (eNOS), inducing secondary activation of TRPC5 via cysteine S-nitrosylation and eNOS in vascular endothelial cells. Mutations in the caveolin-1-binding domains of TRPC5 disrupt its association with caveolin-1 and impair Ca2+ influx and NO production, suggesting that caveolin-1 serves primarily as the scaffold for TRPC5 and eNOS to assemble into the signal complex. Interestingly, during ATP receptor activation, eNOS is dissociated from caveolin-1 and in turn directly associates with TRPC5, which accumulates at the plasma membrane dependently on Ca2+ influx and calmodulin. This protein reassembly likely results in a relief of eNOS from the inhibitory action of caveolin-1 and an enhanced TRPC5 S-nitrosylation by eNOS localized in the proximity, thereby facilitating the secondary activation of Ca2+ influx and NO production. In isolated rat aorta, vasodilation induced by acetylcholine was significantly suppressed by the TRPC5 inhibitor AC1903. Thus, our study provides evidence that dynamic remodeling of the protein assemblies among TRPC5, eNOS, caveolin-1, and calmodulin determines the ensemble of Ca2+ mobilization and NO production in vascular endothelial cells.


Subject(s)
Calcium , Caveolin 1 , Nitric Oxide Synthase Type III , Nitric Oxide , TRPC Cation Channels , Nitric Oxide Synthase Type III/metabolism , Caveolin 1/metabolism , Caveolin 1/genetics , TRPC Cation Channels/metabolism , TRPC Cation Channels/genetics , Animals , Nitric Oxide/metabolism , Humans , Rats , Calcium/metabolism , Male , Calcium Signaling/physiology , Endothelial Cells/metabolism , Signal Transduction , Feedback, Physiological , HEK293 Cells
9.
Biochem Biophys Res Commun ; 733: 150586, 2024 Nov 12.
Article in English | MEDLINE | ID: mdl-39197200

ABSTRACT

The modified cell-penetrating peptide Pas2r12 can deliver antibodies (IgG, 150 kDa) and enhanced green fluorescent protein (EGFP1, 27 kDa) into the cytosol through caveolae-dependent endocytosis. In this study, we determined the effect of Caveolin-1 overexpression on the cytosolic delivery of EGFP by Pas2r12. Three types of Caveolin-1 overexpressing strains were isolated, including Cav1L (low), Cav1M (medium), and Cav1H (high), using HEK293 as the parent cell line. We found that the number of caveolae on the surface of the Caveolin-1-overexpressing strains was similar to that of HEK293. We examined the cytosolic delivery rate of EGFP by Pas2r12. In the Cav1L and Cav1M cells, there was little change compared with HEK293; however, in Cav1H, the rate was significantly decreased. Moreover, the amount of EGFP uptake into the cells (total intracellular EGFP) showed an increasing trend in Cav1H compared with HEK293. These results indicate that in Cav1H, the amount of EGFP uptake into the cells increases, whereas the cytosolic delivery rate of EGFP decreases. This suggests that high overexpression of Caveolin-1 inhibits the transition of EGFP from endosomes to the cytosol.


Subject(s)
Caveolin 1 , Cell-Penetrating Peptides , Cytosol , Green Fluorescent Proteins , Caveolin 1/metabolism , Caveolin 1/genetics , Humans , Green Fluorescent Proteins/metabolism , Green Fluorescent Proteins/genetics , Cytosol/metabolism , HEK293 Cells , Cell-Penetrating Peptides/metabolism , Cell-Penetrating Peptides/chemistry , Endocytosis , Protein Transport , Caveolae/metabolism
10.
Kidney Int ; 106(3): 356-358, 2024 Sep.
Article in English | MEDLINE | ID: mdl-39174195

ABSTRACT

Jiang et al. show that zinc finger FYVE-type containing 21, a Rab5 effector in glomerular endothelial cells is involved in the maintenance of glomerular filtration barrier homeostasis through the stabilization of activated endothelial nitric oxide synthase on subcellular vesicles. The study demonstrates that zinc finger FYVE-type containing 21 could modulate the levels of caveolin-1 in glomerular endothelial cells using vesicle-based trafficking, thereby supporting endothelial nitric oxide synthase activity. The authors provide evidence that decreased zinc finger FYVE-type containing 21 expression in glomerular endothelial cells could play a role in aging-related glomerular filtration barrier dysfunction.


Subject(s)
Aging , Caveolin 1 , Endothelial Cells , Nitric Oxide Synthase Type III , Aging/metabolism , Aging/physiology , Humans , Nitric Oxide Synthase Type III/metabolism , Caveolin 1/metabolism , Endothelial Cells/metabolism , Endothelial Cells/pathology , Animals , Glomerular Filtration Barrier/metabolism , Kidney Glomerulus/pathology , Kidney Glomerulus/physiopathology , Kidney Glomerulus/metabolism , Kidney/physiopathology , Kidney/metabolism , Endothelium, Vascular/physiopathology , Endothelium, Vascular/metabolism
11.
Cancer Lett ; 598: 217130, 2024 Aug 28.
Article in English | MEDLINE | ID: mdl-39089666

ABSTRACT

PURPOSE: Cholesterol metabolism reprograming has been acknowledged as a novel feature of cancers. Pancreatic ductal adenocarcinoma (PDAC) is a cancer with a high demand of cholesterol for rapid growth. The underlying mechanism of how cholesterol metabolism homestasis are disturbed in PDAC is explored. EXPERIMENTAL DESIGN: The relevance between PDAC and cholesterol was confirmed in TCGA database. The expression and clinical association were discovered in TCGA and GEO datasets. Knockdown and overexpression of AGFG1 was adopted to perform function studies. RNA sequencing, cholesterol detection, transmission electron microscope, co-immunoprecipitation, and immunofluorescence et al. were utilized to reveal the underlying mechanism. RESULTS: AGFG1 was identified as one gene positively correlated with cholesterol metabolism in PDAC as revealed by bioinformatics analysis. AGFG1 expression was then found associated with poor prognosis in PDAC. AGFG1 knockdown led to decreased proliferation of tumor cells both in vitro and in vivo. By RNA sequencing, we found AGFG1 upregulated expression leads to enhanced intracellular cholesterol biosynthesis. AGFG1 knockdown suppressed cholesterol biosynthesis and an accumulation of cholesterol in the ER. Mechanistically, we confirmed that AGFG1 interacted with CAV1 to relocate cholesterol for the proceeding of cholesterol biosynthesis, therefore causing disorders in intracellular cholesterol metabolism. CONCLUSIONS: Our study demonstrates the tumor-promoting role of AGFG1 by disturbing cholesterol metabolism homestasis in PDAC. Our study has present a new perspective on cancer therapeutic approach based on cholerstrol metabolism in PDAC.


Subject(s)
Carcinoma, Pancreatic Ductal , Cell Proliferation , Cholesterol , Homeostasis , Pancreatic Neoplasms , Humans , Cholesterol/metabolism , Cholesterol/biosynthesis , Carcinoma, Pancreatic Ductal/pathology , Carcinoma, Pancreatic Ductal/metabolism , Carcinoma, Pancreatic Ductal/genetics , Pancreatic Neoplasms/pathology , Pancreatic Neoplasms/metabolism , Pancreatic Neoplasms/genetics , Animals , Cell Line, Tumor , Mice , Gene Expression Regulation, Neoplastic , Disease Progression , Prognosis , Caveolin 1/genetics , Caveolin 1/metabolism , Mice, Nude , Male
12.
J Biochem Mol Toxicol ; 38(8): e23785, 2024 Aug.
Article in English | MEDLINE | ID: mdl-39051181

ABSTRACT

An arteriovenous fistula (AVF) is the preferred vascular access for hemodialysis in uremic patients, yet its dysfunction poses a significant clinical challenge. Venous stenosis, primarily caused by venous neointimal hyperplasia, is a key factor in the failure of vascular access. During vascular access dysfunction, endothelial cells (ECs) transform mechanical stimuli into intracellular signals and interact with vascular smooth muscle cells. Tanshinone IIA, an important compound derived from Salvia miltiorrhiza, has been widely used to treat cardiovascular diseases. However, its role in modulating ECs under uremic conditions remains incompletely understood. In this research, ECs were exposed to sodium tanshinone IIA sulfonate (STS) and subjected to shear stress and uremic conditions. The results indicate that STS can reduce the suppressive effects on the expression of NF-κB p65, JNK and Collagen I in uremia-induced ECs. Moreover, the downregulation of NF-κB p65, JNK and Collagen I can be enhanced through the inhibition of ERK1/2 and the upregulation of Caveolin-1. These findings suggest that tanshinone IIA may improve EC function under uremic conditions by targeting the Caveolin-1/ERK1/2 pathway, presenting tanshinone IIA as a potential therapeutic agent against AVF immaturity caused by EC dysfunction.


Subject(s)
Abietanes , Caveolin 1 , Uremia , Uremia/metabolism , Uremia/drug therapy , Uremia/pathology , Humans , Abietanes/pharmacology , Abietanes/therapeutic use , Caveolin 1/metabolism , MAP Kinase Signaling System/drug effects , Collagen Type I/metabolism , Transcription Factor RelA/metabolism , Endothelial Cells/drug effects , Endothelial Cells/metabolism , Endothelial Cells/pathology , Human Umbilical Vein Endothelial Cells/metabolism , Human Umbilical Vein Endothelial Cells/drug effects , Phenanthrenes
13.
PLoS One ; 19(7): e0305222, 2024.
Article in English | MEDLINE | ID: mdl-38959243

ABSTRACT

BACKGROUND: Currently, there are few treatment-predictive and prognostic biomarkers in triple-negative breast cancer (TNBC). Caveolin-1 (CAV1) is linked to chemoresistance and several important processes involved in tumor progression and metastasis, such as epithelial-mesenchymal transition (EMT). Herein, we report that high CAV1 gene expression is an independent factor of poor prognosis in TNBC. METHODS: CAV1 gene expression was compared across different molecular features (e.g., PAM50 subtypes). CAV1 expression was assessed in relation to clinical outcomes using Cox regression adjusted for clinicopathological predictors. Differential gene expression and gene set enrichment analyses were applied to compare high- and low-expressing CAV1 tumors. Tumor microenvironment composition of high- and low-expressing CAV1 tumors was estimated using ECOTYPER. Tumor tissue microarrays were used to evaluate CAV1 protein levels in stromal and malignant cells. RESULTS: In the SCAN-B (n = 525) and GSE31519 (n = 327) cohorts, patients with CAV1-high tumors had an increased incidence of early recurrence adjusted HR 1.78 (95% CI 1.12-2.81) and 2.20 (95% CI 1.39-3.47), respectively. In further analysis, high CAV1 gene expression was associated with a molecular profile indicating altered metabolism, neovascularization, chemoresistance, EMT, suppressed immune response, and active tumor microenvironment. Protein levels of CAV1 in malignant and stromal cells were not correlated with CAV1 gene expression. CONCLUSION: CAV1 gene expression in TNBC is a biomarker that merits further investigation in clinical trials and as a therapeutic target.


Subject(s)
Caveolin 1 , Drug Resistance, Neoplasm , Triple Negative Breast Neoplasms , Tumor Microenvironment , Humans , Caveolin 1/genetics , Caveolin 1/metabolism , Triple Negative Breast Neoplasms/genetics , Triple Negative Breast Neoplasms/pathology , Triple Negative Breast Neoplasms/metabolism , Tumor Microenvironment/genetics , Female , Drug Resistance, Neoplasm/genetics , Middle Aged , Gene Expression Regulation, Neoplastic , RNA, Messenger/genetics , RNA, Messenger/metabolism , Prognosis , Biomarkers, Tumor/genetics , Biomarkers, Tumor/metabolism , Epithelial-Mesenchymal Transition/genetics , Aged
14.
Nanotechnology ; 35(41)2024 Jul 25.
Article in English | MEDLINE | ID: mdl-39008958

ABSTRACT

The rise of gene therapy has solved many diseases that cannot be effectively treated by conventional methods. Gene vectors is very important to protect and deliver the therapeutic genes to the target site. Polyethyleneimine (PEI) modified with mannitol could enhance the gene transfection efficiency reported by our group previously. In order to further control and improve the effective gene release to action site, disulfide bonds were introduced into mannitol-modified PEI to construct new non-viral gene vectors PeiSM. The degrees of mannitol linking with disulfide bonds were screened. Among them, moderate mannitol-modified PEI with disulfide bonds showed the best transfection efficiency, and significantly enhanced long-term systemic transgene expression for 72 hin vivoeven at a single dose administration, and could promote caveolae-mediated uptake through up-regulating the phosphorylation of caveolin-1 and increase the loaded gene release from the nanocomplexes in high glutathione intracellular environment. This functionalized gene delivery system can be used as an potential and safe non-viral nanovector for further gene therapy.


Subject(s)
Genetic Vectors , Glutathione , Polyethyleneimine , Transfection , Polyethyleneimine/chemistry , Transfection/methods , Glutathione/metabolism , Glutathione/chemistry , Animals , Humans , Genetic Vectors/chemistry , Genetic Vectors/genetics , Mannitol/chemistry , Mice , Caveolin 1/metabolism , Caveolin 1/genetics , Genetic Therapy/methods , Gene Transfer Techniques , Disulfides/chemistry
15.
Arterioscler Thromb Vasc Biol ; 44(9): 2053-2068, 2024 09.
Article in English | MEDLINE | ID: mdl-38989581

ABSTRACT

BACKGROUND: In early atherosclerosis, circulating LDLs (low-density lipoproteins) traverse individual endothelial cells by an active process termed transcytosis. The CANTOS trial (Canakinumab Antiinflammatory Thrombosis Outcome Study) treated advanced atherosclerosis using a blocking antibody for IL-1ß (interleukin-1ß); this significantly reduced cardiovascular events. However, whether IL-1ß regulates early disease, particularly LDL transcytosis, remains unknown. METHODS: We used total internal reflection fluorescence microscopy to quantify transcytosis by human coronary artery endothelial cells exposed to IL-1ß. To investigate transcytosis in vivo, we injected wild-type and knockout mice with IL-1ß and LDL to visualize acute LDL deposition in the aortic arch. RESULTS: Exposure to picomolar concentrations of IL-1ß induced transcytosis of LDL but not of albumin by human coronary artery endothelial cells. Surprisingly, expression of the 2 known receptors for LDL transcytosis, ALK-1 (activin receptor-like kinase-1) and SR-BI (scavenger receptor BI), was unchanged or decreased. Instead, IL-1ß increased the expression of the LDLR (LDL receptor); this was unexpected because LDLR is not required for LDL transcytosis. Overexpression of LDLR had no effect on basal LDL transcytosis. However, knockdown of LDLR abrogated the effect of IL-1ß on transcytosis rates while the depletion of Cav-1 (caveolin-1) did not. Since LDLR was necessary but overexpression had no effect, we reasoned that another player must be involved. Using public RNA sequencing data to curate a list of Rab (Ras-associated binding) GTPases affected by IL-1ß, we identified Rab27a. Overexpression of Rab27a alone had no effect on basal transcytosis, but its knockdown prevented induction by IL-1ß. This was phenocopied by depletion of the Rab27a effector JFC1 (synaptotagmin-like protein 1). In vivo, IL-1ß increased LDL transcytosis in the aortic arch of wild-type but not Ldlr-/- or Rab27a-deficient mice. The JFC1 inhibitor nexinhib20 also blocked IL-1ß-induced LDL accumulation in the aorta. CONCLUSIONS: IL-1ß induces LDL transcytosis by a distinct pathway requiring LDLR and Rab27a; this route differs from basal transcytosis. We speculate that induction of transcytosis by IL-1ß may contribute to the acceleration of early disease.


Subject(s)
Coronary Vessels , Endothelial Cells , Interleukin-1beta , Lipoproteins, LDL , Mice, Knockout , Receptors, LDL , Signal Transduction , Transcytosis , rab GTP-Binding Proteins , Interleukin-1beta/metabolism , Animals , Humans , Receptors, LDL/genetics , Receptors, LDL/metabolism , rab GTP-Binding Proteins/metabolism , rab GTP-Binding Proteins/genetics , Endothelial Cells/metabolism , Endothelial Cells/drug effects , Lipoproteins, LDL/metabolism , Coronary Vessels/metabolism , Coronary Vessels/drug effects , Cells, Cultured , Mice, Inbred C57BL , Caveolin 1/metabolism , Caveolin 1/genetics , Aortic Diseases/metabolism , Aortic Diseases/genetics , Aortic Diseases/pathology , Disease Models, Animal , Aorta, Thoracic/metabolism , Aorta, Thoracic/drug effects , Atherosclerosis/metabolism , Atherosclerosis/genetics , Atherosclerosis/pathology , Male , Mice
16.
Proc Natl Acad Sci U S A ; 121(30): e2319267121, 2024 Jul 23.
Article in English | MEDLINE | ID: mdl-39008679

ABSTRACT

Migrasomes, vesicular organelles generated on the retraction fibers of migrating cells, play a crucial role in migracytosis, mediating intercellular communication. The cargoes determine the functional specificity of migrasomes. Migrasomes harbor numerous intraluminal vesicles, a pivotal component of their cargoes. The mechanism underlying the transportation of these intraluminal vesicles to the migrasomes remains enigmatic. In this study, we identified that Rab10 and Caveolin-1 (CAV1) mark the intraluminal vesicles in migrasomes. Transport of Rab10-CAV1 vesicles to migrasomes required the motor protein Myosin Va and adaptor proteins RILPL2. Notably, the phosphorylation of Rab10 by the kinase LRRK2 regulated this process. Moreover, CSF-1 can be transported to migrasomes through this mechanism, subsequently fostering monocyte-macrophage differentiation in skin wound healing, which served as a proof of the physiological importance of this transporting mechanism.


Subject(s)
Caveolin 1 , Cell Movement , rab GTP-Binding Proteins , rab GTP-Binding Proteins/metabolism , rab GTP-Binding Proteins/genetics , Humans , Caveolin 1/metabolism , Caveolin 1/genetics , Macrophages/metabolism , Phosphorylation , Adaptor Proteins, Signal Transducing/metabolism , Adaptor Proteins, Signal Transducing/genetics , Animals , Myosin Type V/metabolism , Myosin Type V/genetics , Leucine-Rich Repeat Serine-Threonine Protein Kinase-2/metabolism , Leucine-Rich Repeat Serine-Threonine Protein Kinase-2/genetics , Mice , Myosin Heavy Chains/metabolism , Myosin Heavy Chains/genetics , Biological Transport , Wound Healing/physiology , Organelles/metabolism
17.
Mol Biol Rep ; 51(1): 829, 2024 Jul 22.
Article in English | MEDLINE | ID: mdl-39037581

ABSTRACT

BACKGROUND: The roles of Caveolin-1 (Cav-1) and the Wnt/ß-catenin signaling pathways in cerebral ischemia-reperfusion (I/R) injury are well established. The translocation of ß-catenin into the nucleus is critical for regulating neuronal apoptosis, repair, and neurogenesis within the ischemic brain. It has been reported that the scaffold domain of Caveolin-1 (Cav-1) (residues 95-98) interacts with ß-catenin (residues 330-337). However, the specific contribution of the Cav-1/ß-catenin complex to I/R injury remains unknown. METHODS AND RESULTS: To investigate the mechanism underlying the involvement of the Cav-1/ß-catenin complex in the subcellular translocation of ß-catenin and its subsequent effects on cerebral I/R injury, we treated ischemic brains with ASON (Cav-1 antisense oligodeoxynucleotides) or FTVT (a competitive peptide antagonist of the Cav-1 and ß-catenin interaction). Our study demonstrated that the binding of Cav-1 to ß-catenin following I/R injury prevented the nuclear accumulation of ß-catenin. Treatment with ASON or FTVT after I/R injury significantly increased the levels of nuclear ß-catenin. Furthermore, ASON reduced the phosphorylation of ß-catenin at Ser33, Ser37, and Thr41, which contributes to its proteasomal degradation, while FTVT increased phosphorylation at Tyr333, which is associated with its nuclear translocation. CONCLUSIONS: The above results indicate that the formation of the Cav-1/ß-catenin complex anchors ß-catenin in the cytoplasm following I/R injury. Additionally, both ASON and FTVT treatments attenuated neuronal death in ischemic brains. Our study suggests that targeting the interaction between Cav-1 and ß-catenin serve as a novel therapeutic strategy to protect against neuronal damage during cerebral injury.


Subject(s)
Caveolin 1 , Cell Nucleus , Neurons , Reperfusion Injury , beta Catenin , beta Catenin/metabolism , Animals , Reperfusion Injury/metabolism , Caveolin 1/metabolism , Caveolin 1/genetics , Neurons/metabolism , Neurons/pathology , Cell Nucleus/metabolism , Male , Rats , Brain Ischemia/metabolism , Brain Ischemia/pathology , Apoptosis , Wnt Signaling Pathway , Rats, Sprague-Dawley , Protein Binding , Protein Transport , Cell Death
18.
Trends Neurosci ; 47(8): 651-664, 2024 Aug.
Article in English | MEDLINE | ID: mdl-38972795

ABSTRACT

Caveolins are a family of transmembrane proteins located in caveolae, small lipid raft invaginations of the plasma membrane. The roles of caveolin-enriched lipid rafts are diverse, and include mechano-protection, lipid homeostasis, metabolism, transport, and cell signaling. Caveolin-1 (Cav-1) and other caveolins were described in endothelial cells and later in other cell types of the central nervous system (CNS), including neurons, astrocytes, oligodendrocytes, microglia, and pericytes. This pancellular presence of caveolins demands a better understanding of their functional roles in each cell type. In this review we describe the various functions of Cav-1 in the cells of normal and pathological brains. Several emerging preclinical findings suggest that Cav-1 could represent a potential therapeutic target in brain disorders.


Subject(s)
Caveolins , Central Nervous System , Humans , Animals , Caveolins/metabolism , Central Nervous System/metabolism , Central Nervous System/physiology , Caveolin 1/metabolism , Neurons/metabolism , Neurons/physiology
19.
Cell Calcium ; 123: 102928, 2024 Nov.
Article in English | MEDLINE | ID: mdl-39003871

ABSTRACT

As the uncontrolled entry of calcium ions (Ca2+) through plasmalemmal calcium channels is a cell death trigger, the conjecture is here raised that mitigating such an excess of Ca2+ entry should rescue from death the vulnerable neurons in neurodegenerative diseases (NDDs). However, this supposition has failed in some clinical trials (CTs). Thus, a recent CT tested whether isradipine, a blocker of the Cav1 subtype of voltage-operated calcium channels (VOCCs), exerted a benefit in patients with Parkinson's disease (PD); however, outcomes were negative. This is one more of the hundreds of CTs done under the principle of one-drug-one-target, that have failed in Alzheimer's disease (AD) and other NDDs during the last three decades. As there are myriad calcium channels to let Ca2+ ions gain the cell cytosol, it seems reasonable to predict that blockade of Ca2+ entry through a single channel may not be capable of preventing the Ca2+ flood of cells by the uncontrolled Ca2+ entry. Furthermore, as Ca2+ signaling is involved in the regulation of myriad functions in different cell types, it seems also reasonable to guess that a therapy should be more efficient by targeting different cells with various drugs. Here, we propose to mitigate Ca2+ entry by the simultaneous partial blockade of three quite different subtypes of plasmalemmal calcium channels that is, the Cav1 subtype of VOCCs, the Orai1 store-operated calcium channel (SOCC), and the purinergic P2X7 calcium channel. All three channels are expressed in both microglia and neurons. Thus, by targeting the three channels with a combination of three drug blockers we expect favorable changes in some of the pathogenic features of NDDs, namely (i) to mitigate Ca2+ entry into microglia; (ii) to decrease the Ca2+-dependent microglia activation; (iii) to decrease the sustained neuroinflammation; (iv) to decrease the uncontrolled Ca2+ entry into neurons; (v) to rescue vulnerable neurons from death; and (vi) to delay disease progression. In this review we discuss the arguments underlying our triad hypothesis in the sense that the combination of three repositioned medicines targeting Cav1, Orai1, and P2X7 calcium channels could boost neuroprotection and delay the progression of AD and other NDDs.


Subject(s)
ORAI1 Protein , Receptors, Purinergic P2X7 , Humans , Animals , ORAI1 Protein/metabolism , Receptors, Purinergic P2X7/metabolism , Calcium/metabolism , Neuroprotection/drug effects , Calcium Channel Blockers/pharmacology , Calcium Channel Blockers/therapeutic use , Caveolin 1/metabolism , Neuroprotective Agents/pharmacology , Neuroprotective Agents/therapeutic use , Neurodegenerative Diseases/metabolism , Neurodegenerative Diseases/drug therapy , Neurodegenerative Diseases/pathology , Calcium Channels/metabolism
20.
Arch Dermatol Res ; 316(6): 330, 2024 Jun 05.
Article in English | MEDLINE | ID: mdl-38837051

ABSTRACT

Lichen planopilaris (LPP) and frontal fibrosing alopecia (FFA) are primary cicatricial alopecia that cause a major impact on quality of life due to irreversible hair loss and symptoms as itching, burning and pain. They are characterized by permanent loss of hair follicle stem cells (HFSCs) by pathomechanisms still poorly understood, resulting in poor efficacy of currently available treatments. Caveolae are flask-shaped lipid rafts invaginated within the plasma membrane of multiple cell types. Although their role in the HF physiology and pathophysiology is relatively unknown, we have previously demonstrated that the primary structural component of caveolae (caveolin-1 or Cav1) is upregulated in FFA. Thus, we propose to investigate the expression and localization of caveolae-associated structural proteins (Cav1, Cav2, and Cavin-1) and HFSCs (identified by K15) in both LPP and FFA. We analyzed 4 patients with LPP biopsied in affected and non-affected (NA) scalp, 4 patients with FFA biopsied in affected scalp and 4 healthy controls. Affected scalp of LPP and FFA demonstrated increased levels of Cav1 and Cavin-1 compared with HC and LPP-NA. Moreover, Cav1, Cav2 and Cavin1 all exhibit high colocalization with K15 and their expression appears to be negatively correlated, supporting the hypothesis that these proteins are important players in LPP/FFA and may serve as therapeutic targets in future treatments.


Subject(s)
Alopecia , Caveolae , Caveolin 1 , Hair Follicle , Lichen Planus , Up-Regulation , Humans , Alopecia/pathology , Alopecia/metabolism , Hair Follicle/pathology , Hair Follicle/metabolism , Lichen Planus/metabolism , Lichen Planus/pathology , Middle Aged , Female , Caveolin 1/metabolism , Male , Caveolae/metabolism , Scalp/pathology , Adult , Keratin-15/metabolism , Aged , Biopsy , Fibrosis , Stem Cells/metabolism , Stem Cells/pathology , RNA-Binding Proteins/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL