Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 27
Filter
1.
J Mol Cell Cardiol ; 179: 18-29, 2023 06.
Article in English | MEDLINE | ID: mdl-36967106

ABSTRACT

BACKGROUND: Calcific aortic stenosis (CAS) is more prevalent, occurs earlier, progresses faster and has worse outcomes in patients with chronic kidney disease (CKD). The uremic toxin indoxyl sulfate (IS) is powerful predictor of cardiovascular mortality in these patients and a strong promoter of ectopic calcification whose role in CAS remains poorly studied. The objective of this study was to evaluate whether IS influences the mineralization of primary human valvular interstitial cells (hVICs) from the aortic valve. METHODS: Primary hVICs were exposed to increasing concentrations of IS in osteogenic medium (OM). The hVICs' osteogenic transition was monitored by qRT-PCRs for BMP2 and RUNX2 mRNA. Cell mineralization was assayed using the o-cresolphthalein complexone method. Inflammation was assessed by monitoring NF-κB activation using Western blots as well as IL-1ß, IL-6 and TNF-α secretion by ELISAs. Small interfering RNA (siRNA) approaches enabled us to determine which signaling pathways were involved. RESULTS: Indoxyl-sulfate increased OM-induced hVICs osteogenic transition and calcification in a concentration-dependent manner. This effect was blocked by silencing the receptor for IS (the aryl hydrocarbon receptor, AhR). Exposure to IS promoted p65 phosphorylation, the blockade of which inhibited IS-induced mineralization. Exposure to IS promoted IL-6 secretion by hVICs, a phenomenon blocked by silencing AhR or p65. Incubation with an anti-IL-6 antibody neutralized IS's pro-calcific effects. CONCLUSION: IS promotes hVIC mineralization through AhR-dependent activation of the NF-κB pathway and the subsequent release of IL-6. Further research should seek to determine whether targeting inflammatory pathways can reduce the onset and progression of CKD-related CAS.


Subject(s)
Aortic Valve Stenosis , Calcinosis , Humans , Aortic Valve/metabolism , NF-kappa B/metabolism , Aortic Valve Stenosis/metabolism , Interleukin-6/pharmacology , Indican/pharmacology , Indican/metabolism , Osteogenesis , Receptors, Aryl Hydrocarbon/metabolism , Calcinosis/metabolism , Cells, Cultured , Cell Differentiation , RNA, Small Interfering/metabolism , Sulfates/metabolism , Sulfates/pharmacology
2.
Kidney Int ; 99(6): 1382-1391, 2021 06.
Article in English | MEDLINE | ID: mdl-33647324

ABSTRACT

In chronic kidney disease (CKD), calcium-sensing receptor (CaSR) expression and function have been extensively studied in parathyroid tissue and vascular tissues. To examine whether similar changes occurred in other tissues, we measured total and surface CaSR expression in monocytes of patients with various stages of CKD and healthy volunteers respectively in cross-sectional studies. We further explored in vitro the impact of uremic serum on CaSR expression in monocytes (U937 and THP-1 cell lines), and whether human peripheral blood mononuclear cells or U937 and THP-1 monocytes might modify vascular calcium deposition in rat carotid arteries in vitro. CKD was associated with a decrease in peripheral blood mononuclear cell CaSR expression both in total and at the monocyte surface alone (43% and 34%, respectively in CKD stages 4-5). This decrease was associated with a reduction in the ability of monocytes to inhibit vascular calcification in vitro. Pretreatment with the calcimimetic NPSR568 of peripheral blood mononuclear cells isolated from patients with CKD significantly improved monocyte capacity to reduce carotid calcification in vitro. The fewer peripheral blood mononuclear cells expressing cell surface CaSR, the more calcimimetic treatment enhanced the decrease of carotid calcium content. Thus, we demonstrate that monocyte CaSR expression is decreased in patients with CKD and provide in vitro evidence for a potential role of this decrease in the promotion of vascular calcification. Hence, targeting this alteration or following monocyte CaSR expression as an accessible marker might represent a promising therapeutic strategy in CKD-associated arterial calcification.


Subject(s)
Monocytes , Receptors, Calcium-Sensing , Renal Insufficiency, Chronic , Vascular Calcification , Animals , Calcium , Cross-Sectional Studies , Humans , Leukocytes, Mononuclear , Rats , Vascular Calcification/etiology , Vascular Calcification/prevention & control
3.
J Mol Cell Cardiol ; 129: 2-12, 2019 04.
Article in English | MEDLINE | ID: mdl-30769016

ABSTRACT

INTRODUCTION AND AIMS: Calcific aortic valve disease (CAVD) is the most common heart valve disease in western countries. It has been reported that activation of the calcium-sensing receptor(CaSR) expressed by vascular smooth muscle cells prevents vascular calcification. However, to date, the CaSR's expression and function in cardiac valves have not been studied. The present study sought to evaluate the presence of the CaSR within human valvular interstitial cells (hVICs), assess the CaSR's functionality, and ascertain its involvement in hVIC calcification. METHODS AND RESULTS: Data from Western blot, flow cytometry and immunocytochemistry experiments demonstrated that primary hVICs express the CaSR. The receptor was functional, since the incubation of hVICs with the calcimimetic R-568 significantly increased Ca2+-induced ERK1/2 phosphorylation, and exposure to the calcilytic NPS2143 reduced ERK1/2 activation. A reduction in endogenous CaSR expression by hVICs (using siRNA) was associated with significantly lower levels of Ca2+-induced mineralization (quantified using Alizarin Red staining). Similar data were obtained after the pharmacological inhibition of CaSR activity by the calcilytic NPS2143. In contrast, overexpression of a functional CaSR amplified Ca2+-induced calcification. Pharmacological activation of the CaSR with the calcimimetic R-568 showed similar effects. CaSR's procalcific properties are associated with increased osteogenic transition (as characterized by elevated mRNA expression of bone morphogenetic protein 2 and osterix), and reduced the expression of the calcification inhibitor osteopontin. Histological analysis of 12 human aortic tricuspid valves showed that CaSR expression was greater in calcified areas than in non-calcified areas. These data were confirmed by Western blots. CONCLUSIONS: To the best of our knowledge, this study is the first to have demonstrated that hVICs express a functional CaSR. Taken as a whole, our data suggest that activation of the CaSR expressed by hVICs might be a key promoter of CAVD progression.


Subject(s)
Aortic Valve Stenosis/metabolism , Aortic Valve/metabolism , Aortic Valve/pathology , Calcinosis/metabolism , Receptors, Calcium-Sensing/metabolism , Aortic Valve Stenosis/pathology , Calcinosis/pathology , Calcium/metabolism , Down-Regulation , Humans , Minerals/metabolism , Osteogenesis , Receptors, Calcium-Sensing/genetics , Tricuspid Valve/metabolism
4.
Biochim Biophys Acta ; 1853(9): 2158-67, 2015 Sep.
Article in English | MEDLINE | ID: mdl-25701758

ABSTRACT

The inverse correlation between dietary calcium intake and the risk of colorectal cancer (CRC) is well known, but poorly understood. Expression of the calcium-sensing receptor (CaSR), a calcium-binding G protein-coupled receptor is downregulated in CRC leading us to hypothesize that the CaSR has tumor suppressive roles in the colon. The aim of this study was to understand whether restoration of CaSR expression could reduce the malignant phenotype in CRC. In human colorectal tumors, expression of the CaSR negatively correlated with proliferation markers whereas loss of CaSR correlated with poor tumor differentiation and reduced apoptotic potential. In vivo, dearth of CaSR significantly increased expression of proliferation markers and decreased levels of differentiation and apoptotic markers in the colons of CaSR/PTH double knock-out mice confirming the tumor suppressive functions of CaSR. In vitro CRC cells stably overexpressing wild-type CaSR showed significant reduction in proliferation, as well as increased differentiation and apoptotic potential. The positive allosteric modulator of CaSR, NPS R-568 further enhanced these effects, whereas treatment with the negative allosteric modulator, NPS 2143 inhibited these functions. Interestingly, the dominant-negative mutant (R185Q) was able to abrogate these effects. Our results demonstrate a critical tumor suppressive role of CaSR in the colon. Restoration of CaSR expression and function is linked to regulation of the balance between proliferation, differentiation, and apoptosis and provides a rationale for novel strategies in CRC therapy.


Subject(s)
Colorectal Neoplasms/metabolism , Colorectal Neoplasms/prevention & control , Receptors, Calcium-Sensing/metabolism , Receptors, G-Protein-Coupled/metabolism , Tumor Suppressor Proteins/metabolism , Amino Acid Substitution , Aniline Compounds/pharmacology , Animals , Apoptosis/drug effects , Apoptosis/genetics , Caco-2 Cells , Cell Differentiation/drug effects , Cell Differentiation/genetics , Cell Proliferation/drug effects , Cell Proliferation/genetics , Colorectal Neoplasms/genetics , Colorectal Neoplasms/pathology , Female , Gene Expression Regulation, Neoplastic/drug effects , Gene Expression Regulation, Neoplastic/genetics , Humans , Male , Mice , Mice, Knockout , Mutation, Missense , Naphthalenes/pharmacology , Phenethylamines , Propylamines , Receptors, Calcium-Sensing/antagonists & inhibitors , Receptors, Calcium-Sensing/genetics , Receptors, G-Protein-Coupled/antagonists & inhibitors , Receptors, G-Protein-Coupled/genetics , Tumor Suppressor Proteins/antagonists & inhibitors , Tumor Suppressor Proteins/genetics
6.
J Cell Physiol ; 230(12): 2927-35, 2015 Dec.
Article in English | MEDLINE | ID: mdl-25899466

ABSTRACT

Chronic kidney disease (CKD) is characterized by vascular remodeling and the retention of uremic toxins, several of which are independently associated with the high cardiovascular mortality rate in CKD patients. Whether the association between these uremic toxins and cardiovascular mortality is due to induction of vascular dysfunction and resulting vascular remodeling remains to be determined. This study evaluates the effects of para-cresyl sulfate (PCS), a newly identified uremic toxin, on vascular function and remodeling. PCS acutely induced oxidative stress in both endothelial and vascular smooth muscle cells, with a maximal effect at 0.15 mM, corresponding to the mean "uremic" concentration found in dialysis patients. PCS significantly increased within 30 min phenylephrine-induced contraction of mouse thoracic aorta, through direct activation of rho-kinase, independently of oxidative stress induction, as demonstrated by the capacity of rho-kinase inhibitor Y-27632 to abolish this effect. After exposure of the aorta to PCS for 48 h, we observed inward eutrophic remodeling, a hallmark of uremic vasculopathy characterized by a reduction of the area of both lumen and media, with unchanged media/lumen ratio. In conclusion, elevated PCS concentrations such as those observed in CKD patients, by promoting both vascular dysfunction and vascular remodeling, may contribute to the development of hypertension and to cardiovascular mortality in CKD.


Subject(s)
Aorta, Thoracic/drug effects , Cresols/toxicity , Sulfuric Acid Esters/toxicity , Vascular Remodeling/drug effects , Vasoconstriction/drug effects , Animals , Aorta, Thoracic/metabolism , Aorta, Thoracic/physiopathology , Cells, Cultured , Dose-Response Relationship, Drug , Female , Human Umbilical Vein Endothelial Cells/drug effects , Human Umbilical Vein Endothelial Cells/metabolism , Humans , In Vitro Techniques , Mice, Inbred C57BL , Muscle, Smooth, Vascular/drug effects , Muscle, Smooth, Vascular/metabolism , Muscle, Smooth, Vascular/physiopathology , Myocytes, Smooth Muscle/drug effects , Myocytes, Smooth Muscle/metabolism , Oxidative Stress/drug effects , Signal Transduction/drug effects , Time Factors , Tissue Culture Techniques , rho-Associated Kinases/metabolism
7.
Cardiovasc Res ; 119(13): 2355-2367, 2023 Oct 24.
Article in English | MEDLINE | ID: mdl-37517061

ABSTRACT

AIMS: Inflammatory cytokines play a critical role in the progression of calcific aortic valve disease (CAVD), for which there is currently no pharmacological treatment. The aim of this study was to test the hypothesis that interleukin-8 (IL-8), known to be involved in arterial calcification, also promotes aortic valve calcification (AVC) and to evaluate whether pharmacologically blocking the IL-8 receptor, CXC motif chemokine receptor 2 (CXCR2), could be effective in preventing AVC progression. METHODS AND RESULTS: A cohort of 195 patients (median age 73, 74% men) diagnosed with aortic valve stenosis (severe in 16.9% of cases) were prospectively followed by CT for a median time of 2.6 years. A Cox proportional hazards regression analysis indicated that baseline IL-8 serum concentrations were associated with rapid progression of AVC, defined as an annualized change in the calcification score by CT ≥ 110 AU/year, after adjustment for age, gender, bicuspid anatomy, and baseline disease severity. In vitro, exposure of primary human aortic valvular interstitial cells (hVICs) to 15 pg/mL IL-8 induced a two-fold increase in inorganic phosphate (Pi)-induced calcification. IL-8 promoted NFκB pathway activation, MMP-12 expression, and elastin degradation in hVICs exposed to Pi. These effects were prevented by SCH527123, an antagonist of CXCR2. The expression of CXCR2 was confirmed in hVICs and samples of aortic valves isolated from patients with CAVD, in which the receptor was mainly found in calcified areas, along with MMP-12 and a degraded form of elastin. Finally, in a rat model of chronic kidney disease-associated CAVD, SCH527123 treatment (1 mg/kg/day given orally for 11 weeks) limited the decrease in aortic cusp separation, the increase in maximal velocity of the transaortic jet, and the increase in aortic mean pressure gradient measured by echocardiography, effects that were associated with a reduction in hydroxyapatite deposition and MMP-12 expression in the aortic valves. CONCLUSION: Overall, these results highlight, for the first time, a significant role for IL-8 in the progression of CAVD by promoting calcification via a CXCR2- and MMP-12-dependent mechanism that leads to elastin degradation, and identify CXCR2 as a promising therapeutic target for the treatment of CAVD.

8.
Sci Rep ; 11(1): 7464, 2021 04 02.
Article in English | MEDLINE | ID: mdl-33811249

ABSTRACT

Chronic kidney disease (CKD) worsens ischemic stroke severity in both patients and animals. In mice, these poorer functional outcomes are associated with decreased brain activity of AMP-activated protein kinase (AMPK), a molecule that recently emerged as a potential therapeutic target for ischemic stroke. The antidiabetic drug metformin, a well-known activator of AMPK, has improved stroke outcomes in diabetic patients with normal renal function. We investigated whether chronic metformin pre-conditioning can rescue AMPK activity and prevent stroke damage in non-diabetic mice with CKD. Eight-week-old female C57BL/6J mice were assigned to CKD or SHAM groups. CKD was induced through right kidney cortical electrocautery, followed by left total nephrectomy. Mice were then allocated to receive metformin (200 mg/kg/day) or vehicle for 5 weeks until stroke induction by transient middle cerebral artery occlusion (tMCAO). The infarct volumes were lower in CKD mice exposed to metformin than in vehicle-treated CKD mice 24 h after tMCAO. Metformin pre-conditioning of CKD mice improved their neurological score, grip strength, and prehensile abilities. It also enhanced AMPK activation, reduced apoptosis, increased neuron survival and decreased microglia/macrophage M1 signature gene expression as well as CKD-induced activation of the canonical NF-κB pathway in the ischemic lesions of CKD mice.


Subject(s)
Metformin/therapeutic use , Renal Insufficiency, Chronic/complications , Renal Insufficiency, Chronic/drug therapy , Stroke/drug therapy , Stroke/prevention & control , Adenylate Kinase/metabolism , Animals , Apoptosis/drug effects , Body Weight , Brain Infarction/blood , Brain Infarction/complications , Brain Infarction/drug therapy , Brain Infarction/genetics , Enzyme Activation/drug effects , Female , Gene Expression Regulation , Gliosis/blood , Gliosis/complications , Gliosis/drug therapy , Infarction, Middle Cerebral Artery/blood , Infarction, Middle Cerebral Artery/complications , Infarction, Middle Cerebral Artery/drug therapy , Infarction, Middle Cerebral Artery/genetics , Ischemic Preconditioning , Macrophages/drug effects , Macrophages/pathology , Metformin/blood , Metformin/pharmacology , Mice, Inbred C57BL , Microglia/drug effects , Microglia/pathology , Models, Biological , NF-kappa B/metabolism , Neurons/drug effects , Neurons/pathology , Renal Insufficiency, Chronic/blood , Renal Insufficiency, Chronic/genetics , Stroke/genetics
9.
Am J Pathol ; 175(6): 2416-29, 2009 Dec.
Article in English | MEDLINE | ID: mdl-19893034

ABSTRACT

Recent data suggest that the signal transducer and activator of transcription (STAT)5 contributes to differentiation and growth of mast cells. It has also been described that constitutively phosphorylated STAT5 (pSTAT5) plays a pro-oncogenic role in various myeloid neoplasms. We examined the expression of pSTAT5 in neoplastic mast cells in systemic mastocytosis and asked whether the disease-related oncoprotein KIT D816V is involved in STAT5 activation. As assessed by immunohistochemistry using the anti-pSTAT5 antibody AX1, neoplastic mast cells were found to display pSTAT5 in all SM patients examined (n = 40). Expression of pSTAT5 was also demonstrable in the KIT D816V-positive mast cell leukemia cell line HMC-1. Using various staining-protocols, pSTAT5 was found to be located in both the cytoplasmic and nuclear compartment of mast cells. To define the functional role of KIT D816V in STAT5-activation, Ba/F3 cells with doxycycline-inducible expression of KIT D816V were used. In these cells, induction of KIT D816V resulted in an increased expression of pSTAT5 without substantial increase in total STAT5. Moreover, the KIT D816V-targeting kinase-inhibitor PKC412 was found to counteract expression of pSTAT5 in HMC-1 cells as well as doxycycline-induced expression of pSTAT5 in Ba/F3 cells. Finally, a dominant negative STAT5-construct was found to inhibit growth of HMC-1 cells. Together, our data show that neoplastic mast cells express cytoplasmic and nuclear pSTAT5, that KIT D816V promotes STAT5-activation, and that STAT5-activation contributes to growth of neoplastic mast cells.


Subject(s)
Mast Cells/metabolism , Mastocytosis, Systemic/genetics , Mastocytosis, Systemic/metabolism , Proto-Oncogene Proteins c-kit/genetics , STAT5 Transcription Factor/metabolism , Adult , Aged , Blotting, Western , Cell Separation , Electrophoretic Mobility Shift Assay , Female , Flow Cytometry , Humans , Immunohistochemistry , Male , Middle Aged , Mutation
10.
Blood ; 112(6): 2463-73, 2008 Sep 15.
Article in English | MEDLINE | ID: mdl-18579792

ABSTRACT

The D816V-mutated variant of Kit triggers multiple signaling pathways and is considered essential for malignant transformation in mast cell (MC) neoplasms. We here describe that constitutive activation of the Stat5-PI3K-Akt-cascade controls neoplastic MC development. Retrovirally transduced active Stat5 (cS5(F)) was found to trigger PI3K and Akt activation, and to transform murine bone marrow progenitors into tissue-infiltrating MCs. Primary neoplastic Kit D816V(+) MCs in patients with mastocytosis also displayed activated Stat5, which was found to localize to the cytoplasm and to form a signaling complex with PI3K, with consecutive Akt activation. Finally, the knock-down of either Stat5 or Akt activity resulted in growth inhibition of neoplastic Kit D816V(+) MCs. These data suggest that a downstream Stat5-PI3K-Akt signaling cascade is essential for Kit D816V-mediated growth and survival of neoplastic MCs.


Subject(s)
MAP Kinase Signaling System , Mastocytosis, Systemic/pathology , Phosphatidylinositol 3-Kinases/metabolism , Proto-Oncogene Proteins c-kit/physiology , STAT5 Transcription Factor/metabolism , Animals , Bone Marrow Cells , Case-Control Studies , Cell Proliferation , Hematopoietic Stem Cells , Humans , Leukemic Infiltration , Mice , Mutation, Missense , Proto-Oncogene Proteins c-akt/metabolism , Proto-Oncogene Proteins c-kit/genetics
11.
Exp Cell Res ; 315(12): 2072-80, 2009 Jul 15.
Article in English | MEDLINE | ID: mdl-19285978

ABSTRACT

Breast cancer is the most frequent form of cancer in women, with the highest incidence of metastasis to the bone. The reason for the preferential destination to the bone is believed to be due to chemoattractant factors released during bone resorption, which act on the cancer cells facilitating their metastasis. One of the factors released during osteolysis that may mediate breast cancer bone localization is Ca2+. Here, we show that extracellular Ca2+ (Ca2+(o)) acting via the calcium-sensing receptor (CaSR), greatly promotes the migration of bone-preferring breast cancer cells. In Boyden Chamber and Scratch Wound migration assays, an increase in breast cancer cell migration was observed at 2.5 mM and 5 mM Ca2+(o) compared to basal levels for three of the four breast cancer cell lines tested. However, a significantly greater migratory response was observed for the highly bone metastatic MDA-MB-231 cells, compared to the MCF7 and T47D, which have a lower metastatic potential in vivo. The BT474 cells, which do not metastasize to the bone, did not respond to elevated concentrations of Ca2+(o) in the migration assays. Inhibition of either ERK1/2 MAPK or phospholipase Cbeta (PLCbeta) led to an abolition of the Ca2+(o)-induced migration, implicating these pathways in the migratory response. Knockdown of the CaSR by siRNA resulted in an inhibition of the Ca2+(o)-induced migration, demonstrating the involvement of this receptor in the effect. These results suggest that the activation of the CaSR by elevated Ca2+(o) concentrations, such as those found near resorbing bone, produces an especially strong chemoattractant effect on bone metastatic breast cancer cells toward the Ca2+-rich environment.


Subject(s)
Breast Neoplasms/metabolism , Calcium Signaling/physiology , Calcium/metabolism , Extracellular Space/metabolism , Receptors, Calcium-Sensing/physiology , Bone Neoplasms/secondary , Breast Neoplasms/pathology , Butadienes/pharmacology , Cell Line, Tumor , Chemotaxis , Estrenes/pharmacology , Extracellular Signal-Regulated MAP Kinases/antagonists & inhibitors , Extracellular Signal-Regulated MAP Kinases/metabolism , Female , Humans , Nitriles/pharmacology , Phospholipase C beta/antagonists & inhibitors , Phospholipase C beta/metabolism , Pyrrolidinones/pharmacology
12.
Toxins (Basel) ; 11(9)2019 09 12.
Article in English | MEDLINE | ID: mdl-31547340

ABSTRACT

Cardiovascular disease (CVD) is an important cause of death in patients with chronic kidney disease (CKD), and cardiovascular calcification (CVC) is one of the strongest predictors of CVD in this population. Cardiovascular calcification results from complex cellular interactions involving the endothelium, vascular/valvular cells (i.e., vascular smooth muscle cells, valvular interstitial cells and resident fibroblasts), and monocyte-derived macrophages. Indeed, the production of pro-inflammatory cytokines and oxidative stress by monocyte-derived macrophages is responsible for the osteogenic transformation and mineralization of vascular/valvular cells. However, monocytes/macrophages show the ability to modify their phenotype, and consequently their functions, when facing environmental modifications. This plasticity complicates efforts to understand the pathogenesis of CVC-particularly in a CKD setting, where both uraemic toxins and CKD treatment may affect monocyte/macrophage functions and thereby influence CVC. Here, we review (i) the mechanisms by which each monocyte/macrophage subset either promotes or prevents CVC, and (ii) how both uraemic toxins and CKD therapies might affect these monocyte/macrophage functions.


Subject(s)
Calcinosis/immunology , Cardiomyopathies/immunology , Macrophages , Monocytes , Renal Insufficiency, Chronic/immunology , Animals , Humans
13.
Sci Rep ; 9(1): 6432, 2019 04 23.
Article in English | MEDLINE | ID: mdl-31015533

ABSTRACT

Ischemic stroke is highly prevalent in chronic kidney disease (CKD) patients and has been associated with a higher risk of neurological deterioration and in-hospital mortality. To date, little is known about the processes by which CKD worsens ischemic stroke. This work aimed to investigate the cellular and molecular mechanism associated with ischemic stroke severity in an in vivo model of CKD. CKD was induced through right kidney cortical electrocautery in 8-week-old female C57BL/6 J mice followed by left total nephrectomy. Transient middle cerebral artery occlusion (tMCAO) was performed 6 weeks after left nephrectomy. Twenty-four hours after tMCAO, the infarct volumes were significantly wider in CKD than in SHAM mice. CKD mice displayed decreased neuroscore, impaired ability to remain on rotarod device, weaker muscular strength and decreased prehensile score. Apoptosis, neuronal loss, glial cells recruitment and microglia/macrophages M1 signature genes CD32, CD86, IL-1ß, IL-6, MCP1 and iNOS were significantly increased within ischemic lesions of CKD mice. This effect was associated with decreased AMP kinase phosphorylation and increased activation of the NFΚB pathway. Pharmacological targeting of AMP kinase activity, which is known to block microglia/macrophages M1 polarization, appears promising to improve stroke recovery in CKD.


Subject(s)
Brain Ischemia/physiopathology , Kidney Cortex/metabolism , Muscle Weakness/physiopathology , Neurons/metabolism , Renal Insufficiency, Chronic/physiopathology , Stroke/physiopathology , Adenylate Kinase/genetics , Adenylate Kinase/metabolism , Animals , Antigens, CD/genetics , Antigens, CD/metabolism , Apoptosis/genetics , Brain Ischemia/complications , Brain Ischemia/genetics , Brain Ischemia/metabolism , Chemokine CCL2/genetics , Chemokine CCL2/metabolism , Disease Models, Animal , Electrocoagulation , Female , Gene Expression Regulation , Humans , Interleukin-1beta/genetics , Interleukin-1beta/metabolism , Interleukin-6/genetics , Interleukin-6/metabolism , Kidney Cortex/pathology , Mice , Mice, Inbred C57BL , Muscle Weakness/complications , Muscle Weakness/genetics , Muscle Weakness/metabolism , NF-kappa B/genetics , NF-kappa B/metabolism , Neuroglia/metabolism , Neuroglia/pathology , Neurons/pathology , Renal Insufficiency, Chronic/complications , Renal Insufficiency, Chronic/genetics , Renal Insufficiency, Chronic/metabolism , Rotarod Performance Test , Severity of Illness Index , Stroke/complications , Stroke/genetics , Stroke/metabolism
14.
Joint Bone Spine ; 84(2): 175-181, 2017 Mar.
Article in English | MEDLINE | ID: mdl-27346252

ABSTRACT

OBJECTIVES: We assessed calcium-sensing receptor (CaSR) expression in monocytes isolated from synovial fluid of patients with different types of rheumatisms and explored whether CaSR expression was related to the inflammatory nature of synovial fluid. METHODS: Forty-one patients were included: osteoarthritis (n=10), microcristallin rheumatisms (n=10), rheumatoid arthritis (n=12) and other inflammatory rheumatisms (n=9). Surface and total CaSR expressions in monocytes isolated from synovial fluid and blood were assessed by flow cytometry analysis. U937 cells were cultured during 24hours in presence of cell-free synovial fluids. RESULTS: Every monocyte population tested express the CaSR intra- and extracellularly. Whereas similar pattern of CaSR expression exist in monocyte isolated from blood or synovial fluids, our results indicate that higher CaSR expression levels can be observed in monocytes from synovial fluids than in circulating monocytes. In both populations of monocytes, surface and total CaSR expressions were found to be significantly increased in patients with osteoarthritis compared to rheumatoid arthritis. Similar data were obtained when U937 cells were incubated with cell-free synovial fluids from osteoarthritis patients. Still present, this effect was significantly lowered when "inflammatory" synovial fluids were introduced in culture. CONCLUSIONS: Our results indicate that CaSR expression in synovial derived monocytes is higher in osteoarthritis than in inflammatory rheumatisms and that CaSR expression is modulated by the nature of the synovial fluid. Given the role played by monocytes in the pathogenesis of chronic rheumatisms, monocytes could be interesting therapeutic targets via the CaSR.


Subject(s)
Arthritis/metabolism , Monocytes/metabolism , Receptors, Calcium-Sensing/biosynthesis , Synovial Fluid/metabolism , Adult , Aged , Aged, 80 and over , Cross-Sectional Studies , Female , Flow Cytometry , Humans , Male , Middle Aged , Osteoarthritis/metabolism , Pilot Projects , U937 Cells , Young Adult
15.
J Tissue Eng Regen Med ; 11(2): 382-389, 2017 02.
Article in English | MEDLINE | ID: mdl-24919776

ABSTRACT

In 2000, Masquelet reported a long bone reconstruction technique using an induced membrane formed around a polymethylmethacrylate (PMMA) spacer placed in the defect with appropriate stabilization followed by secondary bone graft after PMMA removal. This reconstruction procedure allows rapid and safe bone reformation for septic, traumatic, neoplastic or congenital bone defects. A rat model of the Masquelet technique was developed to further characterize the biological activities of this induced membrane. Our model allows healing of a critical-sized femoral defect (8 mm) by means of this procedure over a period of 18 weeks. Comparison of induced membranes obtained 3, 4, 5 and 6 weeks after PMMA insertion indicated that this tissue changes over time. Several mineralization spots and bone cells were observed in contact with the PMMA, when assessed by Alizarin Red, Von Kossa, Alkaline phosphatase and Tartrate-resistant acid phosphatase staining of the membranes. CTR (calcitonin receptor)- and RANK (Receptor Activator of Nuclear factor Kappa B)- positive mononuclear cells were detected in the induced membrane, confirming the presence of osteoclasts in this tissue. These cells were observed in a thin, highly cellular layer in the induced membrane in contact with the PMMA. Together, these findings suggest that the membrane is able to promote osteointegration of autologous corticocancellous bone grafts during the Masquelet technique by creating local conditions that may be favourable to graft bone remodelling and osteointegration. Copyright © 2014 John Wiley & Sons, Ltd.


Subject(s)
Bone Regeneration , Bone and Bones/pathology , Membranes, Artificial , Osteoclasts/cytology , Plastic Surgery Procedures , Animals , Bone Cements , Bone Transplantation , Femur/pathology , Leukocytes, Mononuclear/metabolism , Male , Polymethyl Methacrylate/chemistry , Rats , Rats, Sprague-Dawley , Receptor Activator of Nuclear Factor-kappa B/metabolism , Receptors, Calcitonin/metabolism , Surgical Procedures, Operative , Time Factors , Transplantation, Autologous , Wound Healing
16.
Oncotarget ; 8(34): 56460-56472, 2017 Aug 22.
Article in English | MEDLINE | ID: mdl-28915604

ABSTRACT

INTRODUCTION AND AIMS: Osteolytic bone metastases are observed in advanced cases of breast cancer. In vitro data suggest that the activity of the calcium-sensing receptor (CaSR) expressed by metastatic cells could potentiate their osteolytic potential. This study aimed to demonstrate in vivo the involvement of the CaSR in breast cancer cells osteolytic potential and to identify potential targets linked to CaSR activity. METHODS AND RESULTS: MDA-MB-231 stably transfected with plasmids containing either a full-length wild-type CaSR (CaSR-WT), or a functionally inactive dominant negative mutant (CaSR-DN) or an empty vector (EV) were intratibially injected into Balb/c-Nude mice. X-ray analysis performed 19 days after injection showed a dramatic increase of osteolytic lesions in mice injected with CaSR-WT-transfected cells as compared to mice injected with EV- or CaSR-DN-transfected cells. This was associated with decreased BV/TV ratio and increased tumor burden. Epiregulin, an EGF-like ligand, was identified by a DNA microarray as a possible candidate involved in CaSR-mediated osteolysis. Indeed, in vitro, CaSR overexpression increased both epiregulin expression and secretion as compared to EV- or CaSR-DN-transfected cells. Increased epiregulin expression was also detected in osteolytic bone lesions from mice injected with CaSR-WT-transfected MDA-MB-231. In vitro, exposure of osteoblastic cells (HOB and SaOS2) to exogenous epiregulin significantly decreased OPG mRNA expression. Exposure of osteoblastic cells to conditioned media prepared from CaSR-WT-transfected cells also decreased OPG expression. This effect was partially blocked after addition of an anti-epiregulin antibody. CONCLUSIONS: Overexpression of a functional CaSR in metastatic breast cancer cells dramatically amplifies their osteolytic potential through epiregulin-mediated OPG downregulation.

17.
Cell Signal ; 14(10): 869-78, 2002 Oct.
Article in English | MEDLINE | ID: mdl-12135708

ABSTRACT

Erythropoietin (Epo)-induced glycosylphosphatidylinositol (GPI) hydrolysis was previously described to be correlated with phospholipase C-gamma 2 (PLC-gamma2) activation. Here, we analyzed the involvement of phosphatidylinositol (PtdIns) 3-kinase in GPI hydrolysis through PLC-gamma2 tyrosine phosphorylation in response to Epo in FDC-P1 cells transfected with a wild type (WT) erythropoietin-receptor (Epo-R). We showed that phosphatidylinositol 3-kinase (PtdIns 3-kinase) inhibitor LY294002 inhibits Epo-induced hydrolysis of endogenous GPI and Epo-induced PLC-gamma2 tyrosine phosphorylation in a dose-dependent manner. Wortmannin, another PtdIns 3-kinase inhibitor, also suppressed Epo-induced PLC-gamma2 tyrosine phosphorylation. We also present evidence that PLC-gamma2 translocation to the membrane fraction on Epo stimulation is completely inhibited by LY294002. Upon Epo stimulation, the tyrosine-phosphorylated PLC-gamma2 was found to be associated with the tyrosine-phosphorylated Grb2-associated binder (GAB)2, SHC and SHP2 proteins. LY294002 cell preincubation did not affect GAB2, SHC and SHP2 tyrosine phosphorylation but inhibited the binding of PLC-gamma2 to GAB2 and SHP2. Taken together, these results show that PtdIns 3-kinase controls Epo-induced GPI hydrolysis through PLC-gamma2.


Subject(s)
Adaptor Proteins, Signal Transducing , Adaptor Proteins, Vesicular Transport , Cell Membrane/enzymology , Erythrocytes/enzymology , Erythroid Precursor Cells/enzymology , Erythropoietin/metabolism , Glycosylphosphatidylinositols/metabolism , Isoenzymes/metabolism , Phosphatidylinositol 3-Kinases/metabolism , Type C Phospholipases/metabolism , Animals , Cell Differentiation/drug effects , Cell Differentiation/physiology , Cell Division/drug effects , Cell Division/physiology , Cell Membrane/drug effects , Cells, Cultured , Dose-Response Relationship, Drug , Enzyme Inhibitors/pharmacology , Erythropoietin/genetics , Helminth Proteins/metabolism , Humans , Hydrolysis/drug effects , Phospholipase C gamma , Phosphoproteins/metabolism , Phosphorylation , Protein Binding/physiology , Protein Transport/drug effects , Protein Transport/physiology , Proteins/metabolism , Shc Signaling Adaptor Proteins , Src Homology 2 Domain-Containing, Transforming Protein 1 , Subcellular Fractions , Transfection , Tyrosine/metabolism
18.
Endocrinology ; 156(6): 1965-74, 2015 Jun.
Article in English | MEDLINE | ID: mdl-25763635

ABSTRACT

Vascular calcification (VC) is a degenerative disease that contributes to cardiovascular morbidity and mortality. A negative relationship has been demonstrated between VC and calcium sensing receptor (CaSR) expression in the vasculature. Of interest, vitamin D response elements, which allow responsiveness to 1,25-dihydroxyvitamin D3 [1,25(OH)2D3], are present in the promoters of the CaSR gene. We hypothesized that 1,25(OH)2D3, by modulating CaSR expression in vascular smooth muscle cells (VSMCs), might protect against VC. Human VSMCs were exposed to increasing concentrations of 1,25(OH)2D3 (0.01-10 nmol/L) in noncalcifying (1.8 mmol/L) or procalcifying Ca(2+)0 condition (5.0 mmol/L). Using quantitative RT-PCR and Western blotting we observed a significant increase in both CaSR mRNA and protein levels after exposure to 1.0 nmol/L 1,25(OH)2D3. This effect was associated with a maximal increase in CaSR expression at the cell surface after 48 hours of 1,25(OH)2D3 treatment, as assessed by flow cytometry. Down-regulation of the vitamin D receptor by small interfering RNA abolished these effects. In the procalcifying condition, 1.0 nmol/L 1,25(OH)2D3 blocked the Ca(2+)0-induced decrease in total and surface CaSR expression and protected against mineralization. Down-regulation of CaSR expression by CaSR small interfering RNA abolished this protective effect. 1,25(OH)2D3 concentrations of 0.5 and 5.0 nmol/L were also effective, but other (0.01, 0.1, and 10 nmol/L) concentrations did not modify CaSR expression and human VSMC mineralization. In conclusion, these findings suggest that nanomolar concentrations of 1,25(OH)2D3 induce a CaSR-dependent protection against VC. Both lower and higher concentrations are either ineffective or may even promote VC. Whether this also holds true in the clinical setting requires further study.


Subject(s)
Calcitriol/pharmacology , Muscle, Smooth, Vascular/cytology , Myocytes, Smooth Muscle/cytology , Myocytes, Smooth Muscle/drug effects , Receptors, Calcium-Sensing/metabolism , Blotting, Western , Cells, Cultured , Female , Flow Cytometry , Humans , Male , Reverse Transcriptase Polymerase Chain Reaction
19.
Cardiovasc Res ; 101(2): 256-65, 2014 Feb 01.
Article in English | MEDLINE | ID: mdl-24217682

ABSTRACT

AIMS: Vascular calcification (VC) contributes to morbidity and mortality in patients with chronic kidney disease (CKD). Allosteric modulators of the calcium (Ca)-sensing receptor (CaSR) may slow the progression of VC in CKD patients either by reducing serum parathyroid hormone (PTH), Ca, and phosphate levels or by a direct effect on the vessel wall. The aim of this study was to examine the effects of calcimimetics on CaSR expression, cell phenotype, and mineral deposition in human vascular smooth muscle cells (h-VSMCs). METHODS AND RESULTS: Primary h-VSMCs were exposed for 14 days to increasing concentrations of Ca(2+) (from 1.8 to 5 mmol/L) in the presence or absence of calcimimetics R-568 or AMG 641 (0.1 µmol/L). Mineralization was detected by Alizarin red staining, and the cell phenotype was assessed using immunocytochemistry and qRT-PCR. CaSR expression was evaluated using flow cytometry. Short- and long-term exposure (1 day to 14 days) of h-VSMCs to calcimimetics promoted CaSR protein transport from the endoplasmic reticulum to the plasma membrane with enhanced CaSR expression on the cell surface, together with an increase in total cell CaSR expression due to enhanced biosynthesis. In pro-mineralizing conditions, exposure to calcimimetics counteracted the Ca(2+)-dependent reduction of CaSR expression, decreased matrix collagen secretion, and mineral deposition by ~90%. These effects involved CaSR activation since it could be inhibited by CaSR siRNA, but not scrambled siRNA. CONCLUSIONS: The calcimimetic-dependent increase in biosynthesis and activation of the CaSR in h-VSMCs probably play a key role in the protection against calcium-induced VC.


Subject(s)
Aniline Compounds/pharmacology , Biphenyl Compounds/pharmacology , Calcimimetic Agents/pharmacology , Calcium/metabolism , Muscle, Smooth, Vascular/drug effects , Myocytes, Smooth Muscle/drug effects , Phenethylamines/pharmacology , Receptors, Calcium-Sensing/drug effects , Vascular Calcification/prevention & control , Cell Membrane/drug effects , Cell Membrane/metabolism , Cells, Cultured , Collagen Type I/metabolism , Endoplasmic Reticulum/drug effects , Endoplasmic Reticulum/metabolism , Humans , Muscle, Smooth, Vascular/metabolism , Myocytes, Smooth Muscle/metabolism , Phenotype , Propylamines , Protein Transport , RNA Interference , Receptors, Calcium-Sensing/genetics , Receptors, Calcium-Sensing/metabolism , Time Factors , Transfection , Up-Regulation , Vascular Calcification/genetics , Vascular Calcification/metabolism
20.
Arthritis Res Ther ; 16(5): 412, 2014 Aug 19.
Article in English | MEDLINE | ID: mdl-25134967

ABSTRACT

INTRODUCTION: Human circulating monocytes express the calcium-sensing receptor (CaSR) and are involved in atherosclerosis. This study investigated the potential association between vascular calcification in rheumatoid arthritis (RA) and CaSR expression in circulating monocytes. METHODS: In this cross-sectional study, 50 RA patients were compared to 25 control subjects matched for age and gender. Isolation of peripheral blood mononuclear cells and flow cytometry analysis were performed to study the surface and total CaSR expression in circulating monocytes. Coronary artery calcium (CAC) and abdominal aortic calcification (AAC) scores were evaluated by computed tomography and an association between these scores and the surface and/or total CaSR expression in circulating monocytes in RA patients was investigated. RESULTS: The two groups were similar in terms of age (RA: 60.9 ± 8.3 years, versus controls: 59.6 ± 5.3 years) and gender (RA: 74.0% females versus 72.0% females). We did not find a higher prevalence and greater burden of CAC or AAC in RA patients versus age- and gender-matched controls. When compared with control subjects, RA patients did not exhibit greater total CaSR (101.6% ± 28.8 vs. 99.9% ± 22.0) or surface CaSR (104.6% ± 20.4 vs. 99.9% ± 13.7) expression, but total CaSR expression in circulating monocytes was significantly higher in RA patients with severe CAC (Agatston score ≥ 200, n = 11) than in patients with mild-to-moderate CAC (1 to 199, n = 21) (P = 0.01). CONCLUSIONS: This study demonstrates for the first time that total CaSR expression in human circulating monocytes is increased in RA patients with severe coronary artery calcification.


Subject(s)
Arthritis, Rheumatoid/metabolism , Calcinosis/metabolism , Coronary Artery Disease/metabolism , Monocytes/metabolism , Receptors, Calcium-Sensing/metabolism , Aged , Arthritis, Rheumatoid/blood , Arthritis, Rheumatoid/complications , Calcinosis/complications , Coronary Artery Disease/complications , Cross-Sectional Studies , Female , Flow Cytometry , Humans , Logistic Models , Male , Middle Aged , Multivariate Analysis , Severity of Illness Index
SELECTION OF CITATIONS
SEARCH DETAIL