Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 2.213
Filter
1.
Mol Med ; 30(1): 76, 2024 Jun 05.
Article in English | MEDLINE | ID: mdl-38840067

ABSTRACT

BACKGROUND: Advanced glycation end product-modified low-density lipoprotein (AGE-LDL) is related to inflammation and the development of atherosclerosis. Additionally, it has been demonstrated that receptor for advanced glycation end products (RAGE) has a role in the condition known as calcific aortic valve disease (CAVD). Here, we hypothesized that the AGE-LDL/RAGE axis could also be involved in the pathophysiological mechanism of CAVD. METHODS: Human aortic valve interstitial cells (HAVICs) were stimulated with AGE-LDL following pre-treatment with or without interleukin 37 (IL-37). Low-density lipoprotein receptor deletion (Ldlr-/-) hamsters were randomly allocated to chow diet (CD) group and high carbohydrate and high fat diet (HCHFD) group. RESULTS: AGE-LDL levels were significantly elevated in patients with CAVD and in a hamster model of aortic valve calcification. Our in vitro data further demonstrated that AGE-LDL augmented the expression of intercellular cell adhesion molecule-1 (ICAM-1), interleukin-6 (IL-6) and alkaline phosphatase (ALP) in a dose-dependent manner through NF-κB activation, which was attenuated by nuclear factor kappa-B (NF-κB) inhibitor Bay11-7082. The expression of RAGE was augmented in calcified aortic valves, and knockdown of RAGE in HAVICs attenuated the AGE-LDL-induced inflammatory and osteogenic responses as well as NF-κB activation. IL-37 suppressed inflammatory and osteogenic responses and NF-κB activation in HAVICs. The vivo experiment also demonstrate that supplementation with IL-37 inhibited valvular inflammatory response and thereby suppressed valvular osteogenic activities. CONCLUSIONS: AGE-LDL promoted inflammatory responses and osteogenic differentiation through RAGE/NF-κB pathway in vitro and aortic valve lesions in vivo. IL-37 suppressed the AGE-LDL-induced inflammatory and osteogenic responses in vitro and attenuated aortic valve lesions in a hamster model of CAVD.


Subject(s)
Aortic Valve Stenosis , Aortic Valve , Calcinosis , Glycation End Products, Advanced , Lipoproteins, LDL , NF-kappa B , Osteogenesis , Receptor for Advanced Glycation End Products , Signal Transduction , Animals , Aortic Valve/metabolism , Aortic Valve/pathology , Glycation End Products, Advanced/metabolism , NF-kappa B/metabolism , Humans , Calcinosis/metabolism , Calcinosis/pathology , Calcinosis/genetics , Receptor for Advanced Glycation End Products/metabolism , Receptor for Advanced Glycation End Products/genetics , Aortic Valve Stenosis/metabolism , Aortic Valve Stenosis/etiology , Aortic Valve Stenosis/pathology , Cricetinae , Osteogenesis/drug effects , Male , Lipoproteins, LDL/metabolism , Disease Models, Animal , Female , Middle Aged , Glycated Proteins
2.
Scand Cardiovasc J ; 58(1): 2353070, 2024 May 09.
Article in English | MEDLINE | ID: mdl-38757904

ABSTRACT

Objectives: The role of diabetes mellitus as a risk factor for the development of calcific aortic valve disease has not been fully clarified. Aortic valve interstitial cells (VICs) have been suggested to be crucial for calcification of the valve. Induced calcification in cultured VICs is a good in vitro model for aortic valve calcification. The purpose of this study was to investigate whether increased glucose levels increase experimentally induced calcification in cultured human VICs. Design: VICs were isolated from explanted calcified aortic valves after valve replacement. Osteogenic medium induced calcification of cultured VICs at different glucose levels (5, 15, and 25 mM). Calcium deposits were visualized using Alizarin Red staining and measured spectrophotometrically. Results: The higher the glucose concentration, the lower the level of calcification. High glucose (25 mM) reduced calcification by 52% compared with calcification at a physiological (5 mM) glucose concentration (correlation and regression analysis: r = -0.55, p = .025 with increased concentration of glucose). Conclusions: In vitro hyperglycemia-like conditions attenuated calcification in VICs. High glucose levels may trigger a series of events that secondarily stimulate calcification of VICs in vivo.


Subject(s)
Aortic Valve Stenosis , Aortic Valve , Calcinosis , Glucose , Hyperglycemia , Humans , Aortic Valve/pathology , Aortic Valve/metabolism , Aortic Valve/surgery , Calcinosis/pathology , Calcinosis/metabolism , Cells, Cultured , Glucose/metabolism , Hyperglycemia/metabolism , Aortic Valve Stenosis/pathology , Aortic Valve Stenosis/metabolism , Aortic Valve Stenosis/surgery , Male , Middle Aged , Aged , Female , Dose-Response Relationship, Drug , Osteogenesis/drug effects
3.
Commun Biol ; 7(1): 577, 2024 May 16.
Article in English | MEDLINE | ID: mdl-38755434

ABSTRACT

Pseudoxanthoma elasticum (PXE) is a rare disease characterized by ectopic calcification, however, despite the widely spread effect of pro/anti-calcifying systemic factors associated with this genetic metabolic condition, it is not known why elastic fibers in the same patient are mainly fragmented or highly mineralized in clinically unaffected (CUS) and affected (CAS) skin, respectively. Cellular morphology and secretome are investigated in vitro in CUS and CAS fibroblasts. Here we show that, compared to CUS, CAS fibroblasts exhibit: a) differently distributed and organized focal adhesions and stress fibers; b) modified cell-matrix interactions (i.e., collagen gel retraction); c) imbalance between matrix metalloproteinases and tissue inhibitor of metalloproteinases; d) differentially expressed pro- and anti-calcifying proteoglycans and elastic-fibers associated glycoproteins. These data emphasize that in the development of pathologic mineral deposition fibroblasts play an active role altering the stability of elastic fibers and of the extracellular matrix milieu creating a local microenvironment guiding the level of matrix remodeling at an extent that may lead to degradation (in CUS) or to degradation and calcification (in CAS) of the elastic component. In conclusion, this study contributes to a better understanding of the mechanisms of the mineral deposition that can be also associated with several inherited or age-related diseases (e.g., diabetes, atherosclerosis, chronic kidney diseases).


Subject(s)
Calcinosis , Elastin , Fibroblasts , Pseudoxanthoma Elasticum , Pseudoxanthoma Elasticum/metabolism , Pseudoxanthoma Elasticum/pathology , Pseudoxanthoma Elasticum/genetics , Humans , Elastin/metabolism , Fibroblasts/metabolism , Fibroblasts/pathology , Calcinosis/metabolism , Calcinosis/pathology , Dermis/metabolism , Dermis/pathology , Middle Aged , Female , Male , Adult , Cells, Cultured , Extracellular Matrix/metabolism , Elastic Tissue/metabolism , Elastic Tissue/pathology
4.
Sci Rep ; 14(1): 12222, 2024 05 28.
Article in English | MEDLINE | ID: mdl-38806601

ABSTRACT

Calcification of aortic valve leaflets is a growing mortality threat for the 18 million human lives claimed globally each year by heart disease. Extensive research has focused on the cellular and molecular pathophysiology associated with calcification, yet the detailed composition, structure, distribution and etiological history of mineral deposition remains unknown. Here transdisciplinary geology, biology and medicine (GeoBioMed) approaches prove that leaflet calcification is driven by amorphous calcium phosphate (ACP), ACP at the threshold of transformation toward hydroxyapatite (HAP) and cholesterol biomineralization. A paragenetic sequence of events is observed that includes: (1) original formation of unaltered leaflet tissues: (2) individual and coalescing 100's nm- to 1 µm-scale ACP spherules and cholesterol crystals biomineralizing collagen fibers and smooth muscle cell myofilaments; (3) osteopontin coatings that stabilize ACP and collagen containment of nodules preventing exposure to the solution chemistry and water content of pumping blood, which combine to slow transformation to HAP; (4) mm-scale nodule growth via ACP spherule coalescence, diagenetic incorporation of altered collagen and aggregation with other ACP nodules; and (5) leaflet diastole and systole flexure causing nodules to twist, fold their encasing collagen fibers and increase stiffness. These in vivo mechanisms combine to slow leaflet calcification and establish previously unexplored hypotheses for testing novel drug therapies and clinical interventions as viable alternatives to current reliance on surgical/percutaneous valve implants.


Subject(s)
Aortic Valve , Calcinosis , Calcium Phosphates , Collagen , Osteopontin , Calcium Phosphates/metabolism , Humans , Aortic Valve/metabolism , Aortic Valve/pathology , Osteopontin/metabolism , Calcinosis/metabolism , Calcinosis/prevention & control , Collagen/metabolism , Durapatite/metabolism , Durapatite/chemistry , Aortic Valve Stenosis/metabolism , Aortic Valve Stenosis/pathology , Cholesterol/metabolism
5.
J Mater Chem B ; 12(18): 4509-4520, 2024 May 08.
Article in English | MEDLINE | ID: mdl-38647022

ABSTRACT

One of the key challenges in diagnosing thyroid cancer lies in the substantial percentage of indeterminate diagnoses of thyroid nodules that have undergone ultrasound-guided fine-needle aspiration (FNA) biopsy for cytological evaluation. This delays the definitive diagnosis and treatment plans. We recently demonstrated that hydroxyapatite microcalcifications (MCs) aspirated from thyroid nodules may aid nodule diagnosis based on their composition. In particular, Zn-enriched MCs have emerged as potential cancer biomarkers. However, a pertinent question remains: is the elevated Zn content within MCs a consequence of cancer, or do the Zn-enriched MCs encourage tumorigenesis? To address this, we treated the human thyroid cancer cell line MDA-T32 with synthetic MC analogs comprising hydroxyapatite crystals with varied pathologically relevant Zn fractions and assessed the cellular response. The MC analogs exhibited an irregular surface morphology similar to FNA MCs observed in cancerous thyroid nodules. These MC analogs displayed an inverse relationship between Zn fraction and crystallinity, as shown by X-ray diffractometry. The zeta potential of the non-Zn-bearing hydroxyapatite crystals was negative, which decreased once Zn was incorporated into the crystal. The MC analogs were not cytotoxic. The cellular response to exposure to these crystals was evaluated in terms of cell migration, proliferation, the tendency of the cells to form multicellular spheroids, and the expression of cancer markers. Our findings suggest that, if thyroid MCs play a role in promoting cancerous behavior in vivo, it is likely a result of the interplay of crystallinity with Zn and carbonate fractions in MCs.


Subject(s)
Calcinosis , Thyroid Neoplasms , Zinc , Humans , Thyroid Neoplasms/pathology , Thyroid Neoplasms/metabolism , Zinc/chemistry , Calcinosis/pathology , Calcinosis/metabolism , Carbonates/chemistry , Crystallization , Cell Proliferation/drug effects , Cell Line, Tumor , Durapatite/chemistry
6.
Biochim Biophys Acta Mol Basis Dis ; 1870(5): 167171, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38631411

ABSTRACT

Patients with advanced chronic kidney disease (CKD) have elevated circulating calcium × phosphate product levels and exhibit soft tissue calcification. Besides the cardiovascular system, calcification is commonly observed in the cornea in CKD patients on hemodialysis. Cardiovascular calcification is a cell-mediated, highly regulated process, and we hypothesized that a similar regulatory mechanism is implicated in corneal calcification with the involvement of corneal epithelial cells (CECs). We established a mouse model of CKD-associated corneal calcification by inducing CKD in DBA/2J mice with an adenine and high phosphate diet. CKD was associated with aorta and corneal calcification as detected by OsteoSense staining and corneal Ca measurement (1.67-fold elevation, p < 0.001). In vitro, excess phosphate and Ca induced human CEC calcification in a dose-dependent and synergistic manner, without any influence on cell viability. High phosphate and Ca-containing osteogenic medium (OM; 2.5 mmol/L excess phosphate and 0.6 mmol/L excess Ca over control) increased the protein expression of Runx2 and induced its nuclear translocation. OM increased the expression of the bone-specific Ca-binding protein osteocalcin (130-fold increase, p < 0.001). Silencing of Runx2 attenuated OM-induced CEC calcification. Immunohistology revealed upregulation of Runx2 and overlapping between the Runx2 and the Alizarin red positive areas of calcification in the cornea of CKD mice. This work sheds light on the mechanism of CKD-induced corneal calcification and provides tools to test calcification inhibitors for the prevention of this detrimental process.


Subject(s)
Calcinosis , Calcium , Core Binding Factor Alpha 1 Subunit , Osteoblasts , Phosphates , Renal Insufficiency, Chronic , Animals , Core Binding Factor Alpha 1 Subunit/metabolism , Core Binding Factor Alpha 1 Subunit/genetics , Renal Insufficiency, Chronic/pathology , Renal Insufficiency, Chronic/metabolism , Renal Insufficiency, Chronic/complications , Mice , Humans , Osteoblasts/metabolism , Osteoblasts/pathology , Phosphates/metabolism , Calcium/metabolism , Calcinosis/pathology , Calcinosis/metabolism , Epithelium, Corneal/pathology , Epithelium, Corneal/metabolism , Male , Mice, Inbred DBA , Epithelial Cells/metabolism , Epithelial Cells/pathology , Disease Models, Animal , Phenotype
7.
Cardiovasc Toxicol ; 24(5): 499-512, 2024 May.
Article in English | MEDLINE | ID: mdl-38589550

ABSTRACT

Calcific aortic valve stenosis (CAVS) is characterized by increasing inflammation and progressive calcification in the aortic valve leaflets and is a major cause of death in the aging population. This study aimed to identify the inflammatory proteins involved in CAVS and provide potential therapeutic targets. We investigated the observational and causal associations of 92 inflammatory proteins, which were measured using affinity-based proteomic assays. Firstly, the case-control cohort identified differential proteins associated with the occurrence and progression of CAVS. Subsequently, we delved into exploring the causal impacts of these associated proteins through Mendelian randomization. This involved utilizing genetic instruments derived from cis-protein quantitative loci identified in genome-wide association studies, encompassing a cohort of over 400,000 individuals. Finally, we investigated the gene transcription and protein expression levels of inflammatory proteins by single-cell and immunohistochemistry analysis. Multivariate logistic regression and spearman's correlation analysis showed that five proteins showed a significant positive correlation with disease severity. Mendelian randomization showed that elevated levels of two proteins, namely, matrix metallopeptidase-1 (MMP1) and sirtuin 2 (SIRT2), were associated with an increased risk of CAVS. Immunohistochemistry and single-cell transcriptomes showed that expression levels of MMP1 and SIRT2 at the tissue and cell levels were significantly higher in calcified valves than in non-calcified control valves. These findings indicate that MMP1 and SIRT2 are causally related to CAVS and open up the possibility for identifying novel therapeutic targets.


Subject(s)
Aortic Valve Stenosis , Aortic Valve , Aortic Valve/pathology , Biomarkers , Calcinosis , Inflammation Mediators , Matrix Metalloproteinase 1 , Mendelian Randomization Analysis , Proteomics , Humans , Aortic Valve Stenosis/metabolism , Aortic Valve Stenosis/blood , Aortic Valve Stenosis/pathology , Aortic Valve Stenosis/genetics , Calcinosis/genetics , Calcinosis/metabolism , Calcinosis/blood , Calcinosis/pathology , Aortic Valve/metabolism , Male , Female , Aged , Case-Control Studies , Biomarkers/blood , Inflammation Mediators/metabolism , Inflammation Mediators/blood , Matrix Metalloproteinase 1/genetics , Matrix Metalloproteinase 1/metabolism , Middle Aged , Risk Factors , Severity of Illness Index , Aged, 80 and over , Genetic Predisposition to Disease , Blood Proteins/genetics , Blood Proteins/analysis , Phenotype
8.
Cancer Biol Med ; 21(5)2024 Apr 09.
Article in English | MEDLINE | ID: mdl-38605478

ABSTRACT

OBJECTIVE: Mammographic calcifications are a common feature of breast cancer, but their molecular characteristics and treatment implications in hormone receptor-positive (HR+)/human epidermal growth factor receptor 2-negative (HER2-) breast cancer remain unclear. METHODS: We retrospectively collected mammography records of an HR+/HER2- breast cancer cohort (n = 316) with matched clinicopathological, genomic, transcriptomic, and metabolomic data. On the basis of mammographic images, we grouped tumors by calcification status into calcification-negative tumors, tumors with probably benign calcifications, tumors with calcification of low-moderate suspicion for maligancy and tumors with calcification of high suspicion for maligancy. We then explored the molecular characteristics associated with each calcification status across multiple dimensions. RESULTS: Among the different statuses, tumors with probably benign calcifications exhibited elevated hormone receptor immunohistochemical staining scores, estrogen receptor (ER) pathway activation, lipid metabolism, and sensitivity to endocrine therapy. Tumors with calcifications of high suspicion for malignancy had relatively larger tumor sizes, elevated lymph node metastasis incidence, Ki-67 staining scores, genomic instability, cell cycle pathway activation, and may benefit from cyclin-dependent kinase 4 and 6 (CDK4/6) inhibitors. CONCLUSIONS: Our research established links between tumor calcifications and molecular features, thus proposing potential precision treatment strategies for HR+/HER2- breast cancer.


Subject(s)
Breast Neoplasms , Calcinosis , Mammography , Receptor, ErbB-2 , Receptors, Estrogen , Humans , Female , Breast Neoplasms/pathology , Breast Neoplasms/metabolism , Breast Neoplasms/therapy , Receptor, ErbB-2/metabolism , Receptors, Estrogen/metabolism , Middle Aged , Retrospective Studies , Calcinosis/pathology , Calcinosis/metabolism , Receptors, Progesterone/metabolism , Aged , Adult , Biomarkers, Tumor/metabolism , Biomarkers, Tumor/genetics
9.
J Cell Mol Med ; 28(7): e18235, 2024 Apr.
Article in English | MEDLINE | ID: mdl-38509735

ABSTRACT

Kidney stone, one of the oldest known diseases, has plagued humans for centuries, consistently imposing a heavy burden on patients and healthcare systems worldwide due to their high incidence and recurrence rates. Advancements in endoscopy, imaging, genetics, molecular biology and bioinformatics have led to a deeper and more comprehensive understanding of the mechanism behind nephrolithiasis. Kidney stone formation is a complex, multi-step and long-term process involving the transformation of stone-forming salts from free ions into asymptomatic or symptomatic stones influenced by physical, chemical and biological factors. Among the various types of kidney stones observed in clinical practice, calcareous nephrolithiasis is currently the most common and exhibits the most intricate formation mechanism. Extensive research suggests that calcareous nephrolithiasis primarily originates from interstitial subepithelial calcified plaques and/or calcified blockages in the openings of collecting ducts. These calcified plaques and blockages eventually come into contact with urine in the renal pelvis, serving as a nidus for crystal formation and subsequent stone growth. Both pathways of stone formation share similar mechanisms, such as the drive of abnormal urine composition, involvement of oxidative stress and inflammation, and an imbalance of stone inhibitors and promoters. However, they also possess unique characteristics. Hence, this review aims to provide detailed description and present recent discoveries regarding the formation processes of calcareous nephrolithiasis from two distinct birthplaces: renal interstitium and tubule lumen.


Subject(s)
Calcinosis , Kidney Calculi , Humans , Kidney Medulla/metabolism , Kidney Calculi/complications , Kidney Calculi/metabolism , Calcinosis/metabolism , Endoscopy , Inflammation/metabolism
10.
J Neurooncol ; 167(3): 455-465, 2024 May.
Article in English | MEDLINE | ID: mdl-38446374

ABSTRACT

PURPOSE: Meningiomas are the most common type of brain tumors and are generally benign, but malignant atypical meningiomas and anaplastic meningiomas frequently recur with poor prognosis. The metabolism of meningiomas is little known, so few effective treatment options other than surgery and radiation are available, and the targets for treatment of recurrence are not well defined. The Aim of this paper is to find the therapeutic target. METHODS: The effects of bone morphogenetic protein (BMP) signal inhibitor (K02288) and upstream regulator Gremlin2 (GREM2) on meningioma's growth and senescence were examined. In brief, we examined as follows: 1) Proliferation assay by inhibiting BMP signaling. 2) Comprehensive analysis of forced expression GREM2.3) Correlation between GREM2 mRNA expression and proliferation marker in 87 of our clinical samples. 4) Enrichment analysis between GREM2 high/low expressed groups using RNA-seq data (42 cases) from the public database GREIN. 5) Changes in metabolites and senescence markers associated with BMP signal suppression. RESULTS: Inhibitors of BMP receptor (BMPR1A) and forced expression of GREM2 shifted tryptophan metabolism from kynurenine/quinolinic acid production to serotonin production in malignant meningiomas, reduced NAD + /NADH production, decreased gene cluster expression involved in oxidative phosphorylation, and caused decrease in ATP. Finally, malignant meningiomas underwent cellular senescence, decreased proliferation, and eventually formed psammoma bodies. Reanalyzed RNA-seq data of clinical samples obtained from GREIN showed that increased expression of GREM2 decreased the expression of genes involved in oxidative phosphorylation, similar to our experimental results. CONCLUSIONS: The GREM2-BMPR1A-tryptophan metabolic pathway in meningiomas is a potential new therapeutic target.


Subject(s)
Bone Morphogenetic Proteins , Calcinosis , Meningeal Neoplasms , Meningioma , Signal Transduction , Humans , Meningioma/metabolism , Meningioma/pathology , Meningioma/genetics , Meningeal Neoplasms/metabolism , Meningeal Neoplasms/pathology , Meningeal Neoplasms/genetics , Bone Morphogenetic Proteins/metabolism , Bone Morphogenetic Proteins/genetics , Calcinosis/pathology , Calcinosis/metabolism , Calcinosis/genetics , Cell Proliferation , Cellular Senescence , Intercellular Signaling Peptides and Proteins/metabolism , Intercellular Signaling Peptides and Proteins/genetics
11.
Adv Sci (Weinh) ; 11(20): e2307319, 2024 May.
Article in English | MEDLINE | ID: mdl-38502885

ABSTRACT

The senescence of aortic valve interstitial cells (VICs) plays a critical role in the progression of calcific aortic valve disease (CAVD). However, the precise mechanisms underlying the senescence of VICs remain unclear, demanding the identification of a novel target to mitigate this process. Previous studies have highlighted the anti-aging potential of morusin. Thus, this study aimed to explore the therapeutic potential of morusin in CAVD. Cellular experiments reveal that morusin effectively suppresses cellular senescence and cause a shift toward osteogenic differentiation of VICs in vitro. Mechanistically, morusin activate the Nrf2-mediated antiaging signaling pathway by downregulating CCND1 expression and aiding Keap1 degradation through Trim 25. This activation lead to the upregulated expression of antioxidant genes, thus reducing reactive oxygen species production and thereby preventing VIC osteogenic differentiation. In vivo experiments in ApoE-/- mice on a high-fat Western diet demonstrate the positive effect of morusin in mitigating aortic valve calcification. These findings emphasize the antiaging properties of morusin and its potential as a therapeutic agent for CAVD.


Subject(s)
Aortic Valve Stenosis , Aortic Valve , Calcinosis , Cellular Senescence , NF-E2-Related Factor 2 , Signal Transduction , Animals , Aortic Valve/metabolism , Aortic Valve/pathology , Mice , Cellular Senescence/drug effects , NF-E2-Related Factor 2/metabolism , NF-E2-Related Factor 2/genetics , Calcinosis/metabolism , Calcinosis/genetics , Signal Transduction/drug effects , Aortic Valve Stenosis/metabolism , Aortic Valve Stenosis/genetics , Aortic Valve Stenosis/pathology , Disease Models, Animal , Cyclin D1/metabolism , Cyclin D1/genetics , Male , Transcription Factors/metabolism , Transcription Factors/genetics , Osteogenesis/drug effects , Humans , Mice, Inbred C57BL
12.
J Cell Physiol ; 239(5): e31211, 2024 May.
Article in English | MEDLINE | ID: mdl-38304971

ABSTRACT

Cataract, a leading cause of blindness, is characterised by lens opacification. Type 2 diabetes is associated with a two- to fivefold higher prevalence of cataracts. The risk of cataract formation increases with the duration of diabetes and the severity of hyperglycaemia. Hydroxyapatite deposition is present in cataractous lenses that could be the consequence of osteogenic differentiation and calcification of lens epithelial cells (LECs). We hypothesised that hyperglycaemia might promote the osteogenic differentiation of human LECs (HuLECs). Osteogenic medium (OM) containing excess phosphate and calcium with normal (1 g/L) or high (4.5 g/L) glucose was used to induce HuLEC calcification. High glucose accelerated and intensified OM-induced calcification of HuLECs, which was accompanied by hyperglycaemia-induced upregulation of the osteogenic markers Runx2, Sox9, alkaline phosphatase and osteocalcin, as well as nuclear translocation of Runx2. High glucose-induced calcification was abolished in Runx2-deficient HuLECs. Additionally, high glucose stabilised the regulatory alpha subunits of hypoxia-inducible factor 1 (HIF-1), triggered nuclear translocation of HIF-1α and increased the expression of HIF-1 target genes. Gene silencing of HIF-1α or HIF-2α attenuated hyperglycaemia-induced calcification of HuLECs, while hypoxia mimetics (desferrioxamine, CoCl2) enhanced calcification of HuLECs under normal glucose conditions. Overall, this study suggests that high glucose promotes HuLEC calcification via Runx2 and the activation of the HIF-1 signalling pathway. These findings may provide new insights into the pathogenesis of diabetic cataracts, shedding light on potential factors for intervention to treat this sight-threatening condition.


Subject(s)
Calcinosis , Cataract , Core Binding Factor Alpha 1 Subunit , Glucose , Hyperglycemia , Hypoxia-Inducible Factor 1 , Lens, Crystalline , Humans , Alkaline Phosphatase/metabolism , Alkaline Phosphatase/genetics , Calcinosis/etiology , Calcinosis/metabolism , Calcinosis/pathology , Cataract/etiology , Cataract/metabolism , Cataract/pathology , Cell Differentiation/drug effects , Cells, Cultured , Core Binding Factor Alpha 1 Subunit/metabolism , Core Binding Factor Alpha 1 Subunit/genetics , Epithelial Cells/metabolism , Epithelial Cells/drug effects , Epithelial Cells/pathology , Glucose/metabolism , Hyperglycemia/complications , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Hypoxia-Inducible Factor 1, alpha Subunit/genetics , Lens, Crystalline/metabolism , Lens, Crystalline/pathology , Osteocalcin/metabolism , Osteocalcin/genetics , Signal Transduction , SOX9 Transcription Factor/metabolism , SOX9 Transcription Factor/genetics , Hypoxia-Inducible Factor 1/genetics , Hypoxia-Inducible Factor 1/metabolism
13.
Br J Pharmacol ; 181(12): 1843-1856, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38378175

ABSTRACT

BACKGROUND AND PURPOSE: Our previous studies have found that andrographolide (AGP) alleviates calcific aortic valve disease (CAVD), but the underlying mechanism is unclear. This study explores the molecular target and signal mechanisms of AGP in inhibiting CAVD. EXPERIMENTAL APPROACH: The anti-calcification effects of the aortic valve with AGP treatment were evaluated by alizarin red staining in vitro and ultrasound and histopathological assessment of a high-fat (HF)-fed ApoE-/- mouse valve calcification model. A correlation between the H3 histone lactylation (H3Kla) and calcification was detected. Molecular docking and surface plasmon resonance (SPR) experiments were further used to confirm p300 as a target for AGP. Overexpression (oe) and silencing (si) of p300 were used to verify the inhibitory effect of AGP targeting p300 on the H3Kla in vitro and ex vivo. KEY RESULTS: AGP significantly inhibited calcium deposition in valve interstitial cells (VICs) and ameliorated aortic valve calcification. The multi-omics analysis revealed the glycolysis pathway involved in CAVD, indicating that AGP interfered with lactate production by regulating lactate dehydrogenase A (LDHA). In addition, lactylation, a new post-translational modification, was shown to have a role in promoting aortic valve calcification. Furthermore, H3Kla and H3K9la site were shown to correlate with Runx2 expression inhibition by AGP treatment. Importantly, we found that p300 transferase was the molecular target of AGP in inhibiting H3Kla. CONCLUSIONS AND IMPLICATIONS: Our findings, for the first time, demonstrated that AGP alleviates calcification by interfering with H3Kla via p300, which might be a powerful drug to prevent CAVD.


Subject(s)
Aortic Valve Stenosis , Aortic Valve , Calcinosis , Diterpenes , Histones , Animals , Humans , Male , Mice , Aortic Valve/pathology , Aortic Valve/metabolism , Aortic Valve/drug effects , Aortic Valve Stenosis/drug therapy , Aortic Valve Stenosis/metabolism , Aortic Valve Stenosis/pathology , Calcinosis/metabolism , Calcinosis/drug therapy , Calcinosis/pathology , Diterpenes/pharmacology , Diterpenes/chemistry , E1A-Associated p300 Protein/metabolism , E1A-Associated p300 Protein/antagonists & inhibitors , Histones/metabolism , p300-CBP Transcription Factors/metabolism , p300-CBP Transcription Factors/antagonists & inhibitors
14.
BMC Cardiovasc Disord ; 24(1): 128, 2024 Feb 28.
Article in English | MEDLINE | ID: mdl-38418967

ABSTRACT

OBJECTIVE: Calcific aortic valve disease (CAVD) is the leading cause of angina, heart failure, and death from aortic stenosis. However, the molecular mechanisms of its progression, especially the complex disease-related transcriptional regulatory mechanisms, remain to be further elucidated. METHODS: This study used porcine valvular interstitial cells (PVIC) as a model. We used osteogenic induced medium (OIM) to induce calcium deposition in PVICs to calcify them, followed by basic fibroblast growth factor (bFGF) treatment to inhibit calcium deposition. Transcriptome sequencing was used to study the mRNA expression profile of PVICs and its related transcriptional regulation. We used DaPars to further examine alternative polyadenylation (APA) between different treatment groups. RESULTS: We successfully induced calcium deposition of PVICs through OIM. Subsequently, mRNA-seq was used to identify differentially expressed mRNAs for three different treatments: control, OIM-induced and OIM-induced bFGF treatment. Global APA events were identified in the OIM and bFGF treatment groups by bioinformatics analysis. Finally, it was discovered and proven that catalase (CAT) is one of the potential targets of bFGF-induced APA regulation. CONCLUSION: We described a global APA change in a calcium deposition model related to CAVD. We revealed that transcriptional regulation of the CAT gene may contribute to bFGF-induced calcium deposition inhibition.


Subject(s)
Aortic Valve Stenosis , Aortic Valve/pathology , Calcinosis , Swine , Animals , Aortic Valve Stenosis/metabolism , Aortic Valve/metabolism , Calcium/metabolism , Fibroblast Growth Factor 2/genetics , Fibroblast Growth Factor 2/pharmacology , Polyadenylation , Cells, Cultured , Calcinosis/metabolism , RNA, Messenger/genetics , RNA, Messenger/metabolism
15.
Int J Mol Sci ; 25(4)2024 Feb 14.
Article in English | MEDLINE | ID: mdl-38396969

ABSTRACT

Calcific aortic valve disease (CAVD) is characterized by the fibrosis and mineralization of the aortic valve, which leads to aortic stenosis and heart failure. At the cellular level, this is due to the osteoblastic-like differentiation of valve interstitial cells (VICs), resulting in the calcification of the tissue. Unfortunately, human VICs are not readily available to study CAVD pathogenesis and the implicated mechanisms in vitro; however, adipose-derived stromal/stem cells (ASCs), carrying the patient's specific genomic features, have emerged as a promising cell source to model cardiovascular diseases due to their multipotent nature, availability, and patient-specific characteristics. In this study, we describe a comprehensive transcriptomic analysis of tissue-engineered, scaffold-free, ASC-embedded mineralized tissue sheets using bulk RNA sequencing. Bioinformatic and gene set enrichment analyses revealed the up-regulation of genes associated with the organization of the extracellular matrix (ECM), suggesting that the ECM could play a vital role in the enhanced mineralization observed in these tissue-engineered ASC-embedded sheets. Upon comparison with publicly available gene expression datasets from CAVD patients, striking similarities emerged regarding cardiovascular diseases and ECM functions, suggesting a potential link between ECM gene expression and CAVDs pathogenesis. A matrisome-related sub-analysis revealed the ECM microenvironment promotes the transcriptional activation of the master gene runt-related transcription factor 2 (RUNX2), which is essential in CAVD development. Tissue-engineered ASC-embedded sheets with enhanced mineralization could be a valuable tool for research and a promising avenue for the identification of more effective aortic valve replacement therapies.


Subject(s)
Aortic Valve Disease , Aortic Valve Stenosis , Aortic Valve/pathology , Calcinosis , Humans , Aortic Valve Stenosis/metabolism , Calcinosis/metabolism , Aortic Valve Disease/metabolism , Stem Cells/metabolism , Cells, Cultured
16.
Int J Mol Sci ; 25(4)2024 Feb 14.
Article in English | MEDLINE | ID: mdl-38396954

ABSTRACT

Runx2 (runt related transcription factor 2) is an essential transcription factor for osteoblast proliferation and differentiation. Uridine diphosphate (UDP)-N-acetylgalactosamine (GalNAc): polypeptide GalNAc-transferase 3 (Galnt3) prevents proteolytic processing of fibroblast growth factor 23 (Fgf23), which is a hormone that regulates the serum level of phosphorus. Runx2 and Galnt3 were expressed in osteoblasts and osteocytes, and Fgf23 expression was restricted to osteocytes in bone. Overexpression and knock-down of Runx2 upregulated and downregulated, respectively, the expressions of Galnt3 and Fgf23, and Runx2 directly regulated the transcriptional activity of Galnt3 in reporter assays. The expressions of Galnt3 and Fgf23 in osteoblast-specific Runx2 knockout (Runx2fl/flCre) mice were about half those in Runx2fl/fl mice. However, the serum levels of phosphorus and intact Fgf23 in Runx2fl/flCre mice were similar to those in Runx2fl/fl mice. The trabecular bone volume was increased during aging in both male and female Galnt3-/- mice, but the osteoid was reduced. The markers for bone formation and resorption in Galnt3-/- mice were similar to the control in both sexes. Galnt3-/- mice exhibited hyperphosphatemia and hypercalcemia, and the intact Fgf23 was about 40% that of wild-type mice. These findings indicated that Runx2 regulates the expressions of Galnt3 and Fgf23 and that Galnt3 decelerates the mineralization of osteoid by stabilizing Fgf23.


Subject(s)
Calcification, Physiologic , Calcinosis , N-Acetylgalactosaminyltransferases , Osteoblasts , Animals , Female , Male , Mice , Calcinosis/metabolism , Core Binding Factor Alpha 1 Subunit/genetics , Fibroblast Growth Factors/metabolism , N-Acetylgalactosaminyltransferases/genetics , N-Acetylgalactosaminyltransferases/metabolism , Osteoblasts/metabolism , Phosphorus , Polypeptide N-acetylgalactosaminyltransferase
17.
Circulation ; 149(20): 1578-1597, 2024 May 14.
Article in English | MEDLINE | ID: mdl-38258575

ABSTRACT

BACKGROUND: Calcification of the aortic valve leads to increased leaflet stiffness and consequently results in the development of calcific aortic valve disease (CAVD). However, the underlying molecular and cellular mechanisms of calcification remain unclear. Here, we identified a novel aortic valve calcification-associated PIWI-interacting RNA (piRNA; AVCAPIR) that increases valvular calcification and promotes CAVD progression. METHODS: Using piRNA sequencing, we identified piRNAs contributing to the pathogenesis of CAVD that we termed AVCAPIRs. High-cholesterol diet-fed ApoE-/- mice with AVCAPIR knockout were used to examine the role of AVCAPIR in aortic valve calcification (AVC). Gain- and loss-of-function assays were conducted to determine the role of AVCAPIR in the induced osteogenic differentiation of human valvular interstitial cells. To dissect the mechanisms underlying AVCAPIR-elicited procalcific effects, we performed various analyses, including an RNA pulldown assay followed by liquid chromatography-tandem mass spectrometry, methylated RNA immunoprecipitation sequencing, and RNA sequencing. RNA pulldown and RNA immunoprecipitation assays were used to study piRNA interactions with proteins. RESULTS: We found that AVCAPIR was significantly upregulated during AVC and exhibited potential diagnostic value for CAVD. AVCAPIR deletion markedly ameliorated AVC in high-cholesterol diet-fed ApoE-/- mice, as shown by reduced thickness and calcium deposition in the aortic valve leaflets, improved echocardiographic parameters (decreased peak transvalvular jet velocity and mean transvalvular pressure gradient, as well as increased aortic valve area), and diminished levels of osteogenic markers (Runx2 and Osterix) in aortic valves. These results were confirmed in osteogenic medium-induced human valvular interstitial cells. Using unbiased protein-RNA screening and molecular validation, we found that AVCAPIR directly interacts with FTO (fat mass and obesity-associated protein), subsequently blocking its N6-methyladenosine demethylase activity. Further transcriptomic and N6-methyladenosine modification epitranscriptomic screening followed by molecular validation confirmed that AVCAPIR hindered FTO-mediated demethylation of CD36 mRNA transcripts, thus enhancing CD36 mRNA stability through the N6-methyladenosine reader IGF2BP1 (insulin-like growth factor 2 mRNA binding protein 1). In turn, the AVCAPIR-dependent increase in CD36 stabilizes its binding partner PCSK9 (proprotein convertase subtilisin/kexin type 9), a procalcific gene, at the protein level, which accelerates the progression of AVC. CONCLUSIONS: We identified a novel piRNA that induced AVC through an RNA epigenetic mechanism and provide novel insights into piRNA-directed theranostics in CAVD.


Subject(s)
Aortic Valve Stenosis , Aortic Valve , Calcinosis , RNA, Small Interfering , Animals , Calcinosis/metabolism , Calcinosis/genetics , Calcinosis/pathology , Aortic Valve/metabolism , Aortic Valve/pathology , Aortic Valve/abnormalities , Humans , Mice , Aortic Valve Stenosis/metabolism , Aortic Valve Stenosis/genetics , Aortic Valve Stenosis/pathology , RNA, Small Interfering/metabolism , RNA, Small Interfering/genetics , Male , Osteogenesis , Mice, Inbred C57BL , Mice, Knockout , Disease Models, Animal , Aortic Valve Disease/metabolism , Aortic Valve Disease/genetics , Aortic Valve Disease/pathology , Piwi-Interacting RNA
18.
Nat Commun ; 15(1): 557, 2024 Jan 16.
Article in English | MEDLINE | ID: mdl-38228638

ABSTRACT

Calcific aortic valve disease is a prevalent cardiovascular disease with no available drugs capable of effectively preventing its progression. Hence, an efficient drug delivery system could serve as a valuable tool in drug screening and potentially enhance therapeutic efficacy. However, due to the rapid blood flow rate associated with aortic valve stenosis and the lack of specific markers, achieving targeted drug delivery for calcific aortic valve disease has proved to be challenging. Here we find that protease-activated-receptor 2 (PAR2) expression is up-regulated on the plasma membrane of osteogenically differentiated valvular interstitial cells. Accordingly, we develop a magnetic nanocarrier functionalized with PAR2-targeting hexapeptide for dual-active targeting drug delivery. We show that the nanocarriers effectively deliver XCT790-an anti-calcification drug-to the calcified aortic valve under extra magnetic field navigation. We demonstrate that the nano-cargoes consequently inhibit the osteogenic differentiation of valvular interstitial cells, and alleviate aortic valve calcification and stenosis in a high-fat diet-fed low-density lipoprotein receptor-deficient (Ldlr-/-) mouse model. This work combining PAR2- and magnetic-targeting presents an effective targeted drug delivery system for treating calcific aortic valve disease in a murine model, promising future clinical translation.


Subject(s)
Aortic Valve Stenosis , Calcinosis , Mice , Animals , Aortic Valve/metabolism , Aortic Valve Stenosis/drug therapy , Osteogenesis , Calcinosis/drug therapy , Calcinosis/metabolism , Cells, Cultured , Magnetic Phenomena
19.
Am J Pathol ; 194(4): 539-550, 2024 Apr.
Article in English | MEDLINE | ID: mdl-37517686

ABSTRACT

This review focuses on technologies at the core of calcific aortic valve disease (CAVD) and drug target research advancement, including transcriptomics, proteomics, and molecular imaging. We examine how bulk RNA sequencing and single-cell RNA sequencing have engendered organismal genomes and transcriptomes, promoting the analysis of tissue gene expression profiles and cell subpopulations, respectively. We bring into focus how the field is also largely influenced by increasingly accessible proteome profiling techniques. In unison, global transcriptional and protein expression analyses allow for increased understanding of cellular behavior and pathogenic pathways under pathologic stimuli including stress, inflammation, low-density lipoprotein accumulation, increased calcium and phosphate levels, and vascular injury. We also look at how direct investigation of protein signatures paves the way for identification of targetable pathways for pharmacologic intervention. Here, we note that imaging techniques, once a clinical diagnostic tool for late-stage CAVD, have since been refined to address a clinical need to identify microcalcifications using positron emission tomography/computed tomography and even detect in vivo cellular events indicative of early stage CAVD and map the expression of identified proteins in animal models. Together, these techniques generate a holistic approach to CAVD investigation, with the potential to identify additional novel regulatory pathways.


Subject(s)
Aortic Valve Stenosis , Aortic Valve/pathology , Calcinosis , Animals , Aortic Valve/metabolism , Aortic Valve Stenosis/genetics , Aortic Valve Stenosis/metabolism , Aortic Valve Stenosis/pathology , Gene Expression Profiling , Calcinosis/genetics , Calcinosis/metabolism
20.
Curr Opin Clin Nutr Metab Care ; 27(1): 77-86, 2024 01 01.
Article in English | MEDLINE | ID: mdl-37650693

ABSTRACT

PURPOSE OF REVIEW: Calcific aortic valve disease (CAVD), the most common cause of aortic stenosis (AS), is characterized by slowly progressive fibrocalcific remodelling of the valve cusps. Once symptomatic, severe AS is associated with poor survival unless surgical or transcatheter valve replacement is performed. Unfortunately, no pharmacological interventions have been demonstrated to alter the natural history of CAVD. Lipoprotein(a) [Lp(a)], a low-density lipoprotein-like particle, has been implicated in the pathophysiology of CAVD. RECENT FINDINGS: The mechanisms by which Lp(a) results in CAVD are not well understood. However, the oxidized phospholipids carried by Lp(a) are considered a crucial mediator of the disease process. An increasing number of studies demonstrate a causal association between plasma Lp(a) levels and frequency of AS and need for aortic valve replacement, which is independent of inflammation, as measured by plasma C-reactive protein levels. However, not all studies show an association between Lp(a) and increased progression of calcification in individuals with established CAVD. SUMMARY: Epidemiologic, genetic, and Mendelian randomization studies have collectively suggested that Lp(a) is a causal risk factor for CAVD. Whether Lp(a)-lowering can prevent initiation or slow progression of CAVD remains to be demonstrated.


Subject(s)
Aortic Valve Stenosis , Calcinosis , Humans , Aortic Valve/surgery , Aortic Valve/metabolism , Aortic Valve Stenosis/epidemiology , Aortic Valve Stenosis/etiology , Aortic Valve Stenosis/surgery , Calcinosis/genetics , Calcinosis/metabolism , Lipoprotein(a)/genetics , Lipoprotein(a)/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...