Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 78
Filter
Add more filters










Publication year range
1.
Sci Adv ; 10(9): eadj3551, 2024 Mar.
Article in English | MEDLINE | ID: mdl-38427741

ABSTRACT

Pancreatic ductal adenocarcinoma (PDAC) is characterized by its nutrient-scavenging ability, crucial for tumor progression. Here, we investigated the roles of caveolae-mediated endocytosis (CME) in PDAC progression. Analysis of patient data across diverse datasets revealed a strong association of high caveolin-1 (Cav-1) expression with higher histologic grade, the most aggressive PDAC molecular subtypes, and worse clinical outcomes. Cav-1 loss markedly promoted longer overall and tumor-free survival in a genetically engineered mouse model. Cav-1-deficient tumor cell lines exhibited significantly reduced proliferation, particularly under low nutrient conditions. Supplementing cells with albumin rescued the growth of Cav-1-proficient PDAC cells, but not in Cav-1-deficient PDAC cells under low glutamine conditions. In addition, Cav-1 depletion led to significant metabolic defects, including decreased glycolytic and mitochondrial metabolism, and downstream protein translation signaling pathways. These findings highlight the crucial role of Cav-1 and CME in fueling pancreatic tumorigenesis, sustaining tumor growth, and promoting survival through nutrient scavenging.


Subject(s)
Carcinoma, Pancreatic Ductal , Pancreatic Neoplasms , Mice , Animals , Humans , Caveolae/metabolism , Caveolae/pathology , Pancreatic Neoplasms/pathology , Endocytosis , Carcinoma, Pancreatic Ductal/genetics , Carcinoma, Pancreatic Ductal/metabolism , Signal Transduction , Cell Line, Tumor
2.
Adv Healthc Mater ; 12(32): e2302094, 2023 Dec.
Article in English | MEDLINE | ID: mdl-37827986

ABSTRACT

Gene therapy based on miRNAs has broad application prospects in the treatment of tumors. However, due to degradation and ineffective release during intracellular transport, current gene delivery vectors used for miRNAs limited their actual transfection efficiency. This study develops a novel nonviral vector PEI-SPDP-Man (PSM) that can simultaneously target cellular uptake pathways and intracellular responsive release for miR-34a. PSM is synthesized by connected mannitol (Man) to branched polyethylenimine (PEI) using a disulfide bond. The prepared PSM/miR-34a gene delivery system can induce and enter to tumor cells through caveolae-mediated endocytosis to reduce the degradation of miR-34a in lysosomes. The disulfide bond is sensed at high concentration of glutathione (GSH) in the tumor cells and miR-34a is released, thereby reducing the expression of Bcl-2 and CD44 to suppress the proliferation and invasion of tumor cells. In vitro and in vivo experiments show that through the targeted cellular uptake and the efficient release of miR-34a, an effective antitumor and antimetastasis profiles for the treatment of orthotopic triple negative breast cancer (TNBC) are achieved. This strategy of controlling intracellular transport pathways by targeting cellular uptake pathways in the gene therapy is an approach that could be developed for highly effective cancer therapy.


Subject(s)
MicroRNAs , Triple Negative Breast Neoplasms , Humans , Triple Negative Breast Neoplasms/genetics , Triple Negative Breast Neoplasms/therapy , Triple Negative Breast Neoplasms/pathology , Cell Line, Tumor , Polymers , Caveolae/metabolism , Caveolae/pathology , MicroRNAs/metabolism , Gene Transfer Techniques , Endocytosis , Disulfides , Cell Proliferation
3.
JAMA Neurol ; 79(8): 808-816, 2022 08 01.
Article in English | MEDLINE | ID: mdl-35696196

ABSTRACT

Importance: Immune-mediated rippling muscle disease (iRMD) is a rare myopathy characterized by wavelike muscle contractions (rippling) and percussion- or stretch-induced muscle mounding. A serological biomarker of this disease is lacking. Objective: To describe a novel autoantibody biomarker of iRMD and report associated clinicopathological characteristics. Design, Setting, and Participants: This retrospective cohort study evaluated archived sera from 10 adult patients at tertiary care centers at the Mayo Clinic, Rochester, Minnesota, and Brigham & Women's Hospital, Boston, Massachusetts, who were diagnosed with iRMD by neuromuscular specialists in 2000 and 2021, based on the presence of electrically silent percussion- or stretch-induced muscle rippling and percussion-induced rapid muscle contraction with or without muscle mounding and an autoimmune basis. Sera were evaluated for a common biomarker using phage immunoprecipitation sequencing. Myopathology consistent with iRMD was documented in most patients. The median (range) follow-up was 18 (1-30) months. Exposures: Diagnosis of iRMD. Main Outcomes and Measures: Detection of a common autoantibody in serum of patients sharing similar clinical and myopathological features. Results: Seven male individuals and 3 female individuals with iRMD were identified (median [range] age at onset, 60 [18-76] years). An IgG autoantibody specific for caveolae-associated protein 4 (cavin-4) was identified in serum of patients with iRMD using human proteome phage immunoprecipitation sequencing. Immunoassays using recombinant cavin-4 confirmed cavin-4 IgG seropositivity in 8 of 10 patients with iRMD. Results for healthy and disease-control individuals (n = 241, including myasthenia gravis and immune-mediated myopathies) were cavin-4 IgG seronegative. Six of the 8 individuals with cavin-4 IgG were male, and the median (range) age was 60 (18-76) years. Initial symptoms included rippling of lower limb muscles in 5 of 8 individuals or all limb muscles in 2 of 8 sparing bulbar muscles, fatigue in 9 of 10, mild proximal weakness in 3 of 8, and isolated myalgia in 1 of 8, followed by development of diffuse rippling. All patients had percussion-induced muscle rippling and half had percussion- or stretch-induced muscle mounding. Four of the 10 patients had proximal weakness. Plasma creatine kinase was elevated in all but 1 patient. Six of the 10 patients underwent malignancy screening; cancer was detected prospectively in only 1. Muscle biopsy was performed in 7 of the 8 patients with cavin-4 IgG; 6 of 6 specimens analyzed immunohistochemically revealed a mosaic pattern of sarcolemmal cavin-4 immunoreactivity. Three of 6 patients whose results were seropositive and who received immunotherapy had complete resolution of symptoms, 1 had mild improvement, and 2 had no change. Conclusions and Relevance: The findings indicate that cavin-4 IgG may be the first specific serological autoantibody biomarker identified in iRMD. Depletion of cavin-4 expression in muscle biopsies of patients with iRMD suggests the potential role of this autoantigen in disease pathogenesis.


Subject(s)
Muscular Diseases , Myasthenia Gravis , Adult , Aged , Autoantibodies , Biomarkers , Caveolae/metabolism , Caveolae/pathology , Female , Humans , Immunoglobulin G , Male , Middle Aged , Muscular Diseases/metabolism , Myasthenia Gravis/diagnosis , Retrospective Studies
4.
Biomed Pharmacother ; 153: 113282, 2022 Sep.
Article in English | MEDLINE | ID: mdl-35750009

ABSTRACT

Multiple pathophysiological pathways are activated during the process of myocardial injury. Various cardioprotective strategies protect the myocardium from ischemia, infarction, and ischemia/reperfusion (I/R) injury through different targets, yet the clinical translation remains limited. Caveolae and its structure protein, caveolins, have been suggested as a bridge to transmit damage-preventing signals and mediate the protection of ultrastructure in cardiomyocytes under pathological conditions. In this review, we first briefly introduce caveolae and caveolins. Then we review the cardioprotective strategies mediated by caveolins through various pathophysiological pathways. Finally, some possible research directions are proposed to provide future experiments and clinical translation perspectives targeting caveolin based on the investigative evidence.


Subject(s)
Caveolins , Myocardial Reperfusion Injury , Caveolae/metabolism , Caveolae/pathology , Caveolae/ultrastructure , Caveolin 1/metabolism , Caveolins/metabolism , Humans , Ischemia/metabolism , Myocardial Reperfusion Injury/metabolism , Myocardium/pathology
5.
Nanoscale Horiz ; 7(7): 779-789, 2022 06 27.
Article in English | MEDLINE | ID: mdl-35703339

ABSTRACT

Nano-tumor interactions are fundamental for cancer nanotherapy, and the cross-talk of nanomedicines with the extracellular matrix (ECM) is increasingly considered essential. Here, we specifically investigate the nano-ECM interactivity using drug-free nanoparticulates (NPs) and highly metastatic cancer cells as models. We discover with surprise that NPs closely bind to specific types of ECM components, namely, retraction fibers (RFs) and migrasomes, which are located at the rear of tumor cells during their migration. This interaction is observed to alter cell morphology, limit cell motion range and change cell adhesion. Importantly, NPs are demonstrated to inhibit tumor cell removal in vitro, and their anti-metastasis potential is preliminarily confirmed in vivo. Mechanically, the NPs are found to coat and form a rigid shell on the surface of migrasomes and retraction fibers via interaction with lipid raft/caveolae substructures. In this way, NPs block the recognition, endocytosis and elimination of migrasomes by their surrounding tumor cells. Thereby, NPs interfere with the cell-ECM interaction and reduce the promotion effect of migrasomes on cell movement. Additionally, NPs trigger alteration of the expression of proteins related to cell-cell adhesion and cytoskeleton organization, which also restricts cell migration. In summary, all the findings here provide a potential target for anti-tumor metastasis nanomedicines.


Subject(s)
Extracellular Matrix , Neoplasms , Caveolae/pathology , Cell Adhesion , Cell Movement , Endocytosis , Extracellular Matrix/metabolism , Extracellular Matrix/pathology , Humans
6.
J Biol Chem ; 298(6): 102005, 2022 06.
Article in English | MEDLINE | ID: mdl-35513070

ABSTRACT

Caveolae are invaginated membrane domains that provide mechanical strength to cells in addition to being focal points for the localization of signaling molecules. Caveolae are formed through the aggregation of caveolin-1 or -3 (Cav1/3), membrane proteins that assemble into multifunctional complexes with the help of caveola-associated protein cavin-1. In addition to its role in the formation of caveolae, cavin-1, also called polymerase I and transcript release factor, is further known to promote ribosomal RNA transcription in the nucleus. However, the mechanistic link between these functions is not clear. Here, we found that deforming caveolae by subjecting cells to mild osmotic stress (150-300 mOsm) changes levels of GAPDH, Hsp90, and Ras only when Cav1/cavin-1 levels are reduced, suggesting a link between caveola deformation and global protein expression. We show that this link may be due to relocalization of cavin-1 to the nucleus upon caveola deformation. Cavin-1 relocalization is also seen when Cav1-Gαq contacts change upon stimulation. Furthermore, Cav1 and cavin-1 levels have been shown to have profound effects on cytosolic RNA levels, which in turn impact the ability of cells to form stress granules and RNA-processing bodies (p-bodies) which sequester and degrade mRNAs, respectively. Our studies here using a cavin-1-knockout cell line indicate adaptive changes in cytosolic RNA levels but a reduced ability to form stress granules. Taken together, our findings suggest that caveolae, through release of cavin-1, communicate extracellular cues to the cell interior to impact transcriptional and translational.


Subject(s)
Caveolae , Caveolin 1 , Protein Biosynthesis , RNA-Binding Proteins , Transcription, Genetic , Caveolae/metabolism , Caveolae/pathology , Caveolin 1/genetics , Caveolin 1/metabolism , Cell Line , Gene Knockout Techniques , Membrane Proteins/metabolism , RNA, Messenger/metabolism , RNA-Binding Proteins/genetics , RNA-Binding Proteins/metabolism , Signal Transduction
7.
Mol Oncol ; 16(1): 289-306, 2022 01.
Article in English | MEDLINE | ID: mdl-34716958

ABSTRACT

Isocitrate dehydrogenase 1 (IDH1) is a key enzyme in cellular metabolism. IDH1 mutation (IDH1mut) is the most important genetic alteration in lower grade glioma, whereas glioblastoma (GB), the most common malignant brain tumor, often has wild-type IDH1 (IDH1wt). Although there is no effective treatment yet for neither IDH1wt nor IDHmut GB, it is important to note that the survival span of IDH1wt GB patients is significantly shorter than those with IDH1mut GB. Thus, understanding IDH1wt GB biology and developing effective molecular-targeted therapies is of paramount importance. Fatty acid-binding protein 7 (FABP7) is highly expressed in GB, and its expression level is negatively correlated with survival in malignant glioma patients; however, the underlying mechanisms of FABP7 involvement in tumor proliferation are still unknown. In this study, we demonstrate that FABP7 is highly expressed and localized in nuclei in IDH1wt glioma. Wild-type FABP7 (FABP7wt) overexpression in IDH1wt U87 cells increased cell proliferation rate, caveolin-1 expression, and caveolae/caveosome formation. In addition, FABP7wt overexpression increased the levels of H3K27ac on the caveolin-1 promoter through controlling the nuclear acetyl-CoA level via the interaction with ACLY. Consistent results were obtained using a xenograft model transplanted with U87 cells overexpressing FABP7. Interestingly, in U87 cells with mutant FABP7 overexpression, both in vitro and in vivo phenotypes shown by FABP7wt overexpression were disrupted. Furthermore, IDH1wt patient GB showed upregulated caveolin-1 expression, increased levels of histone acetylation, and increased levels of acetyl-CoA compared with IDH1mut patient GB. Taken together, these data suggest that nuclear FABP7 is involved in cell proliferation of GB through caveolae function/formation regulated via epigenetic regulation of caveolin-1, and this mechanism is critically important for IDH1wt tumor biology.


Subject(s)
Brain Neoplasms , Glioblastoma , Glioma , Acetyl Coenzyme A/genetics , Acetyl Coenzyme A/metabolism , Brain Neoplasms/pathology , Caveolae/metabolism , Caveolae/pathology , Caveolin 1/genetics , Caveolin 1/metabolism , Cell Line, Tumor , Cell Proliferation/genetics , Epigenesis, Genetic , Fatty Acid-Binding Protein 7/genetics , Fatty Acid-Binding Protein 7/metabolism , Glioblastoma/genetics , Glioma/metabolism , Humans , Isocitrate Dehydrogenase/genetics , Mutation/genetics , Tumor Suppressor Proteins/metabolism
8.
Eur J Endocrinol ; 185(6): 841-854, 2021 Nov 10.
Article in English | MEDLINE | ID: mdl-34643546

ABSTRACT

OBJECTIVE: CAV1 encodes caveolin-1, a major protein of plasma membrane microdomains called caveolae, involved in several signaling pathways. Caveolin-1 is also located at the adipocyte lipid droplet. Heterozygous pathogenic variants of CAV1 induce rare heterogeneous disorders including pulmonary arterial hypertension and neonatal progeroid syndrome. Only one patient was previously reported with a CAV1 homozygous pathogenic variant, associated with congenital generalized lipodystrophy (CGL3). We aimed to further delineate genetic transmission, clinical, metabolic, and cellular characteristics of CGL3. DESIGN/METHODS: In a large consanguineous kindred referred for CGL, we performed next-generation sequencing, as well as clinical, imagery, and metabolic investigations. We studied skin fibroblasts from the index case and the previously reported patient with CGL3. RESULTS: Four patients, aged 8 months to 18 years, carried a new homozygous p.(His79Glnfs*3) CAV1 variant. They all displayed generalized lipodystrophy since infancy, insulin resistance, low HDL-cholesterol, and/or high triglycerides, but no pulmonary hypertension. Two patients also presented at the age of 15 and 18 years with dysphagia due to achalasia, and one patient had retinitis pigmentosa. Heterozygous parents and relatives (n = 9) were asymptomatic, without any metabolic abnormality. Patients' fibroblasts showed a complete loss of caveolae and no protein expression of caveolin-1 and its caveolin-2 and cavin-1 partners. Patients' fibroblasts also displayed insulin resistance, increased oxidative stress, and premature senescence. CONCLUSIONS: The CAV1 null variant investigated herein leads to an autosomal recessive congenital lipodystrophy syndrome. Loss of caveolin-1 and/or caveolae induces specific manifestations including achalasia which requires specific management. Overlapping phenotypic traits between the different CAV1-related diseases require further studies.


Subject(s)
Caveolin 1/genetics , Esophageal Achalasia/genetics , Lipodystrophy, Congenital Generalized/genetics , Adolescent , Caveolae/pathology , Caveolae/ultrastructure , Caveolin 1/metabolism , Caveolin 2/metabolism , Cellular Senescence , Child , Child, Preschool , Consanguinity , Dyslipidemias/metabolism , Esophageal Achalasia/pathology , Female , Fibroblasts/pathology , Fibroblasts/ultrastructure , Homozygote , Humans , Infant , Lipodystrophy, Congenital Generalized/metabolism , Lipodystrophy, Congenital Generalized/pathology , Male , Microscopy, Electron, Transmission , Oxidative Stress , Pedigree , RNA-Binding Proteins/metabolism
9.
Histol Histopathol ; 36(10): 1085-1092, 2021 Oct.
Article in English | MEDLINE | ID: mdl-34672358

ABSTRACT

BACKGROUND: Hepatocellular carcinoma (HCC) is among the most malignant tumors with high recurrence and low 5-year survival rate. Lipid metabolism is essential in tumor metastasis, although how altered lipid metabolism promotes HCC progression has not been well elucidated. Fat Storage Inducing Transmembrane Protein 2 (FITM2) is a gene involved in lipid homeostasis and cytoskeletal organization; however, its role in regulating tumor biological behavior has not been evaluated. METHODS: In this study, immunohistochemistry was performed to evaluate the expression of FITM2 in HCC. Univariate and multivariate analysis was performed to identify the prognostic factors. RNA interference wound healing and transwell experiments were performed to analyze the biological role of FITM2. Western blot analysis was performed to investigate the potential downstream signaling. RESULTS: The results revealed that FITM2 was highly expressed in the intratumoral tissues of HCC. Expression of intratumoral FITM2 was associated with microvascular invasion. FITM2 is an independent risk factor of HCC disease-free survival and overall survival. In vitro studies revealed that knockdown of FITM2 significantly inhibited the migration ability of HCC cells. FITM2 promotes HCC cell migration by regulating the expression of caveolin-1 and promoting the formation of caveolae. These results indicate that high intratumoral expression of FITM2 is associated with poor HCC prognosis, which may be applied to develop a new adjuvant therapy.


Subject(s)
Carcinoma, Hepatocellular/genetics , Carcinoma, Hepatocellular/pathology , Caveolae/pathology , Cell Movement/genetics , Membrane Proteins/genetics , Adult , Aged , Aged, 80 and over , Cell Line, Tumor , Female , Humans , Immunohistochemistry , Liver Neoplasms , Male , Membrane Proteins/biosynthesis , Middle Aged , Neoplasm Invasiveness/genetics , Neoplasm Invasiveness/pathology , Prognosis , RNA Interference , Signal Transduction/genetics , Survival Analysis , Wound Healing
10.
Bull Exp Biol Med ; 171(3): 393-398, 2021 Jul.
Article in English | MEDLINE | ID: mdl-34297295

ABSTRACT

We studied ultrastructure and vesicular structures in endothelial cells of myocardial micro-vessels in burn patients. Electron microscopy revealed a significant decrease in volume density of vesicular structures in the endotheliocytes of myocardial capillaries in patients with burn septicotoxemia. The observed structural signs of endothelial dysfunction revealed in this category of patients can be a promising area for further research and for the development of methods of pathogenetic correction of myocardial disorders in the case of burn injury.


Subject(s)
Burns/pathology , Capillaries/ultrastructure , Endothelial Cells/ultrastructure , Myocardium/ultrastructure , Sepsis/pathology , Adult , Autopsy , Burns/complications , Capillaries/pathology , Caveolae/pathology , Caveolae/ultrastructure , Endothelial Cells/pathology , Female , Humans , Male , Microscopy, Electron , Middle Aged , Myocardium/pathology , Sepsis/complications , Transport Vesicles/pathology , Transport Vesicles/ultrastructure
11.
Cancer Metastasis Rev ; 39(2): 505-517, 2020 06.
Article in English | MEDLINE | ID: mdl-32474691

ABSTRACT

Caveolae are bulb-like invaginations made up of two essential structural proteins, caveolin-1 and cavins, which are abundantly present at the plasma membrane of vertebrate cells. Since their discovery more than 60 years ago, the function of caveolae has been mired in controversy. The last decade has seen the characterization of new caveolae components and regulators together with the discovery of additional cellular functions that have shed new light on these enigmatic structures. Early on, caveolae and/or caveolin-1 have been involved in the regulation of several parameters associated with cancer progression such as cell migration, metastasis, angiogenesis, or cell growth. These studies have revealed that caveolin-1 and more recently cavin-1 have a dual role with either a negative or a positive effect on most of these parameters. The recent discovery that caveolae can act as mechanosensors has sparked an array of new studies that have addressed the mechanobiology of caveolae in various cellular functions. This review summarizes the current knowledge on caveolae and their role in cancer development through their activity in membrane tension buffering. We propose that the role of caveolae in cancer has to be revisited through their response to the mechanical forces encountered by cancer cells during tumor mass development.


Subject(s)
Caveolae/pathology , Neoplasms/pathology , Animals , Caveolae/metabolism , Caveolin 1/metabolism , Cell Membrane/metabolism , Cell Membrane/pathology , Humans , Mechanotransduction, Cellular , Neoplasms/metabolism
12.
Cancer Metastasis Rev ; 39(2): 415-433, 2020 06.
Article in English | MEDLINE | ID: mdl-32358634

ABSTRACT

Caveolae are specialised and dynamic plasma membrane subdomains, involved in many cellular functions including endocytosis, signal transduction, mechanosensing and lipid storage, trafficking, and metabolism. Two protein families are indispensable for caveola formation and function, namely caveolins and cavins. Mutations of genes encoding these caveolar proteins cause serious pathological conditions such as cardiomyopathies, skeletal muscle diseases, and lipodystrophies. Deregulation of caveola-forming protein expression is associated with many types of cancers including prostate cancer. The distinct function of secretion of the prostatic fluid, and the unique metabolic phenotype of prostate cells relying on lipid metabolism as a main bioenergetic pathway further suggest a significant role of caveolae and caveolar proteins in prostate malignancy. Accumulating in vitro, in vivo, and clinical evidence showed the association of caveolin-1 with prostate cancer grade, stage, metastasis, and drug resistance. In contrast, cavin-1 was found to exhibit tumour suppressive roles. Studies on prostate cancer were the first to show the distinct function of the caveolar proteins depending on their localisation within the caveolar compartment or as cytoplasmic or secreted proteins. In this review, we summarise the roles of caveola-forming proteins in prostate cancer and the potential of exploiting them as therapeutic targets or biological markers.


Subject(s)
Caveolae/metabolism , Caveolins/metabolism , Membrane Proteins/metabolism , Prostatic Neoplasms, Castration-Resistant/metabolism , Prostatic Neoplasms/metabolism , RNA-Binding Proteins/metabolism , Animals , Caveolae/pathology , Humans , Male , Prostatic Neoplasms/pathology , Prostatic Neoplasms, Castration-Resistant/pathology
13.
Am J Pathol ; 189(9): 1846-1862, 2019 09.
Article in English | MEDLINE | ID: mdl-31199921

ABSTRACT

The mammalian target of rapamycin (mTOR) and associated phosphatidylinositol 3-kinase/AKT/mTOR signaling pathway is commonly up-regulated in cancer, including bladder cancer. mTOR complex 2 (mTORC2) is a major regulator of bladder cancer cell migration and invasion, but the mechanisms by which mTORC2 regulates these processes are unclear. A discovery mass spectrometry and reverse-phase protein array-based proteomics dual approach was used to identify novel mTORC2 phosphoprotein targets in actively invading cancer cells. mTORC2 targets included focal adhesion kinase, proto-oncogene tyrosine-protein kinase Src, and caveolin-1 (Cav-1), among others. Functional testing shows that mTORC2 regulates Cav-1 localization and dynamic phosphorylation of Cav-1 on Y14. Regulation of Cav-1 activity by mTORC2 also alters the abundance of caveolae, which are specialized lipid raft invaginations of the plasma membrane associated with cell signaling and membrane compartmentalization. Our results demonstrate a unique role for mTORC2-mediated regulation of caveolae formation in actively migrating cancer cells.


Subject(s)
Caveolae/pathology , Caveolin 1/metabolism , Cell Movement , Mechanistic Target of Rapamycin Complex 2/metabolism , TOR Serine-Threonine Kinases/metabolism , Urinary Bladder Neoplasms/pathology , Adult , Aged , Aged, 80 and over , Caveolae/metabolism , Caveolin 1/antagonists & inhibitors , Caveolin 1/genetics , Female , Gene Expression Regulation, Neoplastic , Humans , Male , Mechanistic Target of Rapamycin Complex 2/genetics , Middle Aged , Phosphorylation , Prognosis , Proto-Oncogene Mas , RNA, Small Interfering/genetics , Survival Rate , TOR Serine-Threonine Kinases/genetics , Tumor Cells, Cultured , Urinary Bladder Neoplasms/genetics , Urinary Bladder Neoplasms/metabolism
14.
Sci Rep ; 9(1): 6696, 2019 04 30.
Article in English | MEDLINE | ID: mdl-31040342

ABSTRACT

Caveolae are plasma membrane invaginations enriched with high cholesterol and sphingolipid content; they also contain caveolin proteins in their structure. Endothelial nitric oxide synthase (eNOS), an enzyme that synthesizes nitric oxide (NO) by converting L-arginine to L-citrulline, is highly concentrated in plasma membrane caveolae. Hypertension is associated with decreased NO production and impaired endothelium-dependent relaxation. Understanding the molecular mechanisms that follow hypertension is important. For this study, we hypothesized that spontaneously hypertensive rat (SHR) vessels should have a smaller number of caveolae, and that the caveolae structure should be disrupted in these vessels. This should impair the eNOS function and diminish NO bioavailability. Therefore, we aimed to investigate caveolae integrity and density in SHR aortas and mesenteric arteries and the role played by caveolae in endothelium-dependent relaxation. We have been able to show the presence of caveolae-like structures in SHR aortas and mesenteric arteries. Increased phenylephrine-induced contractile response after treatment with dextrin was related to lower NO release. In addition, impaired acetylcholine-induced endothelium-dependent relaxation could be related to decreased caveolae density in SHR vessels. The most important finding of this study was that cholesterol depletion with dextrin induced eNOS phosphorylation at Serine1177 (Ser1177) and boosted reactive oxygen species (ROS) production in normotensive rat and SHR vessels, which suggested eNOS uncoupling. Dextrin plus L-NAME or BH4 decreased ROS production in aorta and mesenteric arteries supernatant's of both SHR and normotensive groups. Human umbilical vein endothelial cells (HUVECs) treated with dextrin confirmed eNOS uncoupling, as verified by the reduced eNOS dimer/monomer ratio. BH4, L-arginine, or BH4 plus L-arginine inhibited eNOS monomerization. All these results showed that caveolae structure and integrity are essential for endothelium-dependent relaxation. Additionally, a smaller number of caveolae is associated with hypertension. Finally, caveolae disruption promotes eNOS uncoupling in normotensive and hypertensive rat vessels and in HUVECs.


Subject(s)
Caveolae/pathology , Endothelium, Vascular/physiopathology , Hypertension/physiopathology , Mesenteric Arteries/pathology , Reactive Oxygen Species/metabolism , Acetylcholine/pharmacology , Animals , Aorta/metabolism , Aorta/pathology , Caveolae/metabolism , Caveolae/ultrastructure , Human Umbilical Vein Endothelial Cells , Humans , Hypertension/metabolism , Male , Mesenteric Arteries/drug effects , Mesenteric Arteries/metabolism , Nitric Oxide/metabolism , Nitric Oxide Synthase Type III/metabolism , Phenylephrine/pharmacology , Rats, Inbred SHR , Rats, Wistar , Vasodilation/drug effects , Vasodilator Agents/pharmacology
15.
Annu Rev Cell Dev Biol ; 34: 111-136, 2018 10 06.
Article in English | MEDLINE | ID: mdl-30296391

ABSTRACT

The plasma membrane of eukaryotic cells is not a simple sheet of lipids and proteins but is differentiated into subdomains with crucial functions. Caveolae, small pits in the plasma membrane, are the most abundant surface subdomains of many mammalian cells. The cellular functions of caveolae have long remained obscure, but a new molecular understanding of caveola formation has led to insights into their workings. Caveolae are formed by the coordinated action of a number of lipid-interacting proteins to produce a microdomain with a specific structure and lipid composition. Caveolae can bud from the plasma membrane to form an endocytic vesicle or can flatten into the membrane to help cells withstand mechanical stress. The role of caveolae as mechanoprotective and signal transduction elements is reviewed in the context of disease conditions associated with caveola dysfunction.


Subject(s)
Caveolae/metabolism , Cell Membrane/genetics , Transport Vesicles/genetics , Caveolae/chemistry , Caveolae/pathology , Cell Membrane/chemistry , Endocytosis/genetics , Humans , Signal Transduction/genetics , Stress, Mechanical , Structure-Activity Relationship , Transport Vesicles/chemistry
16.
PLoS One ; 13(10): e0205306, 2018.
Article in English | MEDLINE | ID: mdl-30346954

ABSTRACT

A range of cellular functions have been attributed to caveolae, flask-like invaginations of the plasma membrane. Here, we have used RNA-seq to achieve quantitative transcriptional profiling of primary embryonic fibroblasts from caveolin 1 knockout mice (CAV1-/- MEFs), and thereby to gain hypothesis-free insight into how these cells respond to the absence of caveolae. Components of the extracellular matrix were decisively over-represented within the set of genes displaying altered expression in CAV1-/- MEFs when compared to congenic wild-type controls. This was confirmed biochemically and by imaging for selected examples. Up-regulation of components of the extracellular matrix was also observed in a second cell line, NIH-3T3 cells genome edited to delete CAV1. Up-regulation of components of the extracellular matrix was detected in vivo by assessing collagen deposition and compliance of CAV1-/- lungs. We discuss the implications of these findings in terms of the cellular function of caveolae.


Subject(s)
Caveolin 1/genetics , Extracellular Matrix Proteins/genetics , Extracellular Matrix/metabolism , Fibroblasts/metabolism , Lung/metabolism , Animals , Caveolae/metabolism , Caveolae/pathology , Caveolin 1/deficiency , Collagen/genetics , Collagen/metabolism , Embryo, Mammalian , Extracellular Matrix/chemistry , Extracellular Matrix Proteins/metabolism , Fibroblasts/chemistry , Fibroblasts/pathology , Gene Editing , Gene Expression Profiling , Gene Expression Regulation , Heterozygote , Homozygote , Lung/pathology , Mice , Mice, Inbred C57BL , Mice, Knockout , NIH 3T3 Cells , Primary Cell Culture , Sequence Analysis, RNA , Transcription, Genetic
17.
Auton Neurosci ; 213: 60-70, 2018 09.
Article in English | MEDLINE | ID: mdl-30005741

ABSTRACT

Cholesterol rich membrane invaginations, caveolae, have important roles in various cellular activities, one of them being signal transduction. This signaling pathway seems to be affected during various bladder disorders and the current study aimed to elucidate the plausible involvement of caveolae mediated signal transduction during cyclophosphamide induced cystitis. Furthermore, the urothelial cholinergic part of ATP-evoked contractions and its possible link to caveolae were investigated. Cholinergic, as well as purinergic, contractile responses in rat urinary bladders were examined using a classic organ bath set-up with full-thickness strip preparations or a whole bladder model that enabled luminal administration of substances. Furthermore, sub groups with and without urothelium were examined. The expression of caveolin-1 was also tested using western blot and immunofluorescence. Caveolae cholesterol depletion by methyl-ß-cyclodextrin entailed a significant decrease of ATP-evoked bladder contractility. Interestingly, after muscarinic blockade the ATP induced contractions were significantly reduced in the same manner. Furthermore, this atropine-sensitive part of ATP-evoked responses was absent in denuded as well as inflamed bladders. A tendency towards a reduced expression of caveolin-1 was observed in rats with experimental cystitis. The cholinergic part of ATP-induced contractile responses seemed to be affected by urothelium denudation as well as caveolae depletion. Removing one of these structures nullifies the effect of the other, suggesting an important interaction between the urothelium and the caveolar structures. These effects are absent in inflamed animals and might be one pathophysiological aspect behind BPS/IC.


Subject(s)
Caveolae/metabolism , Cystitis/metabolism , Receptors, Cholinergic/metabolism , Receptors, Purinergic/metabolism , Urinary Bladder/metabolism , Urothelium/metabolism , Adenosine Triphosphate/metabolism , Animals , Caveolae/drug effects , Caveolae/pathology , Caveolin 1/metabolism , Cyclophosphamide , Cystitis/pathology , Disease Models, Animal , Muscle Contraction/drug effects , Muscle Contraction/physiology , Rats, Sprague-Dawley , Signal Transduction , Tissue Culture Techniques , Urinary Bladder/drug effects , Urinary Bladder/pathology , Urological Agents/pharmacology , Urothelium/drug effects , Urothelium/pathology
18.
Am J Chin Med ; 45(8): 1667-1682, 2017.
Article in English | MEDLINE | ID: mdl-29132216

ABSTRACT

Caveolin-1, the marker protein of membranal caveolae, is not only involved in cholesterol regulation, but also participates in the cleavage of amyloid [Formula: see text]-protein precursor (APP) and the generation of [Formula: see text]-amyloid peptide. It has been reported to be tightly related with Tau. In our previous studies, curcumin has been confirmed to play a neuroprotective role in Alzheimer's disease (AD), but its effects on Caveolin-1, Tau and their correlation, and the mechanism is still unknown. As such, in the present study, N2a/WT cells, N2a/APP695swe cell and six-month-old APP/PS1 double transgenic mice were enrolled. After curcumin treatment, the expression of Caveolin-1, Tau and their relationship was detected, and the potential mechanisms were explored. The results showed that in the N2a/APP695swe cells, curcumin not only decreased the number of caveolae, but also made their membrane to be thinner; and curcumin could decreased the expression of phosphorylated Tau (P-Tau(ser404)/Tau) and Caveolin-1 ([Formula: see text]), but the expression of phosphorylated GSK-3[Formula: see text] (P-GSK-3[Formula: see text]/GSK-3[Formula: see text] was increased ([Formula: see text]). In APP/PS1 transgenic mice, the same results were observed. Taken together, our data suggest that curcumin may play an important role in AD via reducing Caveolin-1, inactivating GSK-3[Formula: see text] and inhibiting the abnormal excessive phosphorylation of Tau, which will provide a new theory for AD treatment with curcumin.


Subject(s)
Alzheimer Disease/genetics , Alzheimer Disease/metabolism , Caveolin 1/physiology , Curcumin/pharmacology , Down-Regulation/drug effects , Glycogen Synthase Kinase 3/genetics , Glycogen Synthase Kinase 3/metabolism , tau Proteins/metabolism , Alzheimer Disease/drug therapy , Amyloid beta-Peptides/metabolism , Amyloid beta-Protein Precursor/genetics , Amyloid beta-Protein Precursor/metabolism , Animals , Brain/metabolism , Caveolae/drug effects , Caveolae/pathology , Caveolin 1/genetics , Caveolin 1/metabolism , Cells, Cultured , Curcumin/therapeutic use , Gene Expression/drug effects , Mice, Transgenic , Molecular Targeted Therapy , Phosphorylation/drug effects , Phytotherapy , tau Proteins/genetics
19.
Biochem Soc Trans ; 45(4): 999-1006, 2017 08 15.
Article in English | MEDLINE | ID: mdl-28710286

ABSTRACT

Tetraspanins are ubiquitous membrane proteins that induce local membrane curvature and hence co-ordinate cell-to-cell contacts. This review highlights their role in inflammation, which requires control of the nano-architecture of attachment sites between endothelial cells and leukocytes. The active role of endothelial cells in preparing for transmigration of leukocytes and determining the severity of an inflammation is often underscored. A clear hint to endothelial pre-activation is their ability to protrude clustered adhesion proteins upward prior to leukocyte contact. The elevation of molecular adhesive platforms toward the blood stream is crucially dependent on tetraspanins. In addition, leukocytes require tetraspanins for their activation. The example of the B-cell receptor is referenced in some detail here, since it provides deeper insights into the receptor-coreceptor interplay. To lift the role of tetraspanins from an abstract model of inflammation toward a player of clinical significance, two pathologies are analyzed for the known contributions of tetraspanins. The recent publication of the first crystal structure of a full-length tetraspanin revealed a cholesterol-binding site, which provides a strong link to the pathophysiological condition of atherosclerosis. Dysregulation of the inflammatory cascade in autoimmune diseases by endothelial cells is exemplified by the involvement of tetraspanins in multiple sclerosis.


Subject(s)
Autoimmune Diseases/metabolism , Cell Membrane/metabolism , Endothelial Cells/metabolism , Inflammation/metabolism , Leukocytes/metabolism , Models, Biological , Tetraspanins/metabolism , Animals , Autoimmune Diseases/blood , Autoimmune Diseases/immunology , Autoimmune Diseases/pathology , Caveolae/immunology , Caveolae/metabolism , Caveolae/pathology , Caveolae/ultrastructure , Cell Adhesion , Cell Membrane/immunology , Cell Membrane/pathology , Cell Membrane/ultrastructure , Endothelial Cells/immunology , Endothelial Cells/pathology , Endothelial Cells/ultrastructure , Humans , Inflammation/blood , Inflammation/immunology , Inflammation/pathology , Leukocytes/immunology , Leukocytes/pathology , Leukocytes/ultrastructure , Microvilli/immunology , Microvilli/metabolism , Microvilli/pathology , Microvilli/ultrastructure , Protein Conformation , Receptors, Antigen, B-Cell/agonists , Receptors, Antigen, B-Cell/chemistry , Receptors, Antigen, B-Cell/metabolism , Signal Transduction , Tetraspanins/chemistry , Transendothelial and Transepithelial Migration
20.
Sci Rep ; 7: 44638, 2017 03 14.
Article in English | MEDLINE | ID: mdl-28291255

ABSTRACT

AS-1, the TIR/BB loop mimetic, plays a protective role in cardiac ischemia/reperfusion (I/R) but the molecular mechanism remains unclear. The muscle specific caveolin3 (Cav-3) and the caveolae have been found to be critical for cardioprotection. This study aimed to evaluate our hypothesis that caveolae and Cav-3 are essential for AS-1-induced cardioprotection against myocardial I/R injury. To address these issues, we analyzed the involvement of Cav-3 in AS-1 mediated cardioprotection both in vivo and in vitro. We demonstrate that AS-1 administration significantly decreased infarct size, improved cardiac function after myocardial I/R and modulated membrane caveolae and Cav-3 expression in the myocardium. For in vitro studies, AS-1 treatment prevented Cav-3 re-distribution induced by H/R injury. In contrast, disruption of caveolae by MCD treatment or Cav-3 knockdown abolished the protection against H/R-induced myocytes injury by AS-1. Our findings reveal that AS-1 attenuates myocardial I/R injury through caveolae and Cav-3 dependent mechanism.


Subject(s)
Cardiotonic Agents/pharmacology , Caveolae/drug effects , Caveolin 3/genetics , Myocardial Reperfusion Injury/drug therapy , Myocytes, Cardiac/drug effects , Peptidomimetics/pharmacology , Animals , Apoptosis/drug effects , Apoptosis/genetics , Caveolae/metabolism , Caveolae/pathology , Caveolin 3/antagonists & inhibitors , Caveolin 3/metabolism , Cell Line , Echocardiography , Gene Expression , Male , Mice , Mice, Inbred C57BL , Myocardial Reperfusion Injury/diagnostic imaging , Myocardial Reperfusion Injury/genetics , Myocardial Reperfusion Injury/metabolism , Myocardium/metabolism , Myocardium/pathology , Myocytes, Cardiac/metabolism , Myocytes, Cardiac/pathology , RNA, Small Interfering/genetics , RNA, Small Interfering/metabolism , Rats , Rats, Wistar
SELECTION OF CITATIONS
SEARCH DETAIL
...