Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 38
Filter
Add more filters










Publication year range
1.
Ann Biol Clin (Paris) ; 81(6): 585-590, 2024 02 24.
Article in English | MEDLINE | ID: mdl-38391163

ABSTRACT

The recreational use of nitrous oxide (N2O) is an emerging public health issue. Chronic N2O abuse may result in various clinical symptoms, encompassing neurological, psychiatric and cardiovascular outcomes. Despite the difficulties for the laboratory investigation of N2O intoxication, there is currently no guidelines in France to help both clinicians and biologists use appropriate biomarkers for the diagnosis and monitoring of patients with clinical symptoms potentially related to N2O intoxication. A multi-disciplinary Working Group, carried out under the auspices of the French Society of Clinical Biology (SFBC) and in collaboration with the French Societies of Emergency Medicine (SFMU), Analytical Toxicology (SFTA), Hemostasis and Thrombosis (SFTH), Vitamins and Biofactors (SFVB), and the French Federation of Neurology (FFN), was recently implemented to elaborate practical guidelines. The methodology of the Working Group is based on the critical analysis of the literature, and raising concerns and objectives are grouped into five working packages. The present manuscript primarily aims to expound upon the methodology and objectives of the ongoing SFBC Working Group on N2O.


Subject(s)
Nitrous Oxide , Substance-Related Disorders , Humans , Nitrous Oxide/toxicity , Biomarkers , France , Vitamin B 12
2.
Pharmaceuticals (Basel) ; 16(11)2023 Oct 24.
Article in English | MEDLINE | ID: mdl-38004380

ABSTRACT

Bone fracture healing is a complex biological process involving four phases coordinated over time: hematoma formation, granulation tissue formation, bony callus formation, and bone remodelling. Bone fractures represent a significant health problem, particularly among the elderly population and patients with comorbidities. Therapeutic strategies proposed to treat such fractures include the use of autografts, allografts, and tissue engineering strategies. It has been shown that bone morphogenetic protein 2 (BMP-2) has a therapeutic potential to enhance fracture healing. Despite the clinical efficacy of BMP-2 in osteoinduction and bone repair, adverse side effects and complications have been reported. Therefore, in this in vitro study, we propose the use of a disaccharide compound (DP2) to improve the mineralisation process. We first evaluated the effect of DP2 on primary human osteoblasts (HOb), and then investigated the mechanisms involved. Our findings showed that (i) DP2 improved osteoblast differentiation by inducing alkaline phosphatase activity, osteopontin, and osteocalcin expression; (ii) DP2 induced earlier in vitro mineralisation in HOb cells compared to BMP-2 mainly by earlier activation of Runx2; and (iii) DP2 is internalized in HOb cells and activates the protein kinase C signalling pathway. Consequently, DP2 is a potential therapeutical candidate molecule for bone fracture repair.

3.
Sci Total Environ ; 905: 167089, 2023 Dec 20.
Article in English | MEDLINE | ID: mdl-37717745

ABSTRACT

OBJECTIVE: Systematic screening for congenital hypothyroidism by heel-stick sampling has revealed unexpected heterogeneity in the geographic distribution of newborn thyroid-stimulating hormone concentrations in Picardy, France. We explored a possible relationship with environmental pollutants. METHODS: Zip code geolocation data from mothers of newborns without congenital hypothyroidism born in 2021 were linked to ecological data for a set of airborne (particulate matter with a diameter of 2.5 µm or less [PM2.5] or 10 µm or less [PM10]) and tap-water (nitrate and perchlorate ions and atrazine) pollutants. Statistical associations between mean exposure levels during the third trimester of pregnancy and Thyroid-stimulating hormone (TSH) concentrations in 6249 newborns (51 % male) were investigated using linear regression models. RESULTS: Median neonatal TSH concentration (interquartile range, IQR) was 1.7 (1-2.8) mIU/L. An increase of one IQR in prenatal exposure to perchlorate ions (3.6 µg/L), nitrate ions (19.2 mg/L), PM2.5 (3.7 µg/m3) and PM10 (3.4 µg/m3), were associated with increases in TSH concentrations of 2.30 % (95 % CI: 0.95-3.66), 5.84 % (95 % CI: 2.81-8.87), 13.44 % (95 % CI: 9.65-17.28) and 6.26 % (95 % CI: 3.01-9.56), respectively. CONCLUSIONS: Prenatal exposure to perchlorate and nitrate ions in tap water and to airborne PM over the third trimester of pregnancy was significantly associated with increased neonatal TSH concentrations.


Subject(s)
Air Pollutants , Air Pollution , Congenital Hypothyroidism , Environmental Pollutants , Prenatal Exposure Delayed Effects , Water Pollutants , Humans , Pregnancy , Infant, Newborn , Female , Male , Child, Preschool , Thyrotropin , Perchlorates , Nitrates , Particulate Matter/analysis , Water , Environmental Exposure
4.
Int J Mol Sci ; 23(11)2022 May 31.
Article in English | MEDLINE | ID: mdl-35682871

ABSTRACT

The endothelium has a fundamental role in the cardiovascular complications of coronavirus disease 2019 (COVID-19). Infection with severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) particularly affects endothelial cells. The virus binds to the angiotensin-converting enzyme 2 (ACE-2) receptor (present on type 2 alveolar cells, bronchial epithelial cells, and endothelial cells), and induces a cytokine storm. The cytokines tumor necrosis factor alpha, interleukin-1 beta, and interleukin-6 have particular effects on endothelial cells-leading to endothelial dysfunction, endothelial cell death, changes in tight junctions, and vascular hyperpermeability. Under normal conditions, apoptotic endothelial cells are removed into the bloodstream. During COVID-19, however, endothelial cells are detached more rapidly, and do not regenerate as effectively as usual. The loss of the endothelium on the luminal surface abolishes all of the vascular responses mediated by the endothelium and nitric oxide production in particular, which results in greater contractility. Moreover, circulating endothelial cells infected with SARS-CoV-2 act as vectors for viral dissemination by forming clusters that migrate into the circulation and reach distant organs. The cell clusters and the endothelial dysfunction might contribute to the various thromboembolic pathologies observed in COVID-19 by inducing the formation of intravascular microthrombi, as well as by triggering disseminated intravascular coagulation. Here, we review the contributions of endotheliopathy and endothelial-cell-derived extracellular vesicles to the pathogenesis of COVID-19, and discuss therapeutic strategies that target the endothelium in patients with COVID-19.


Subject(s)
COVID-19 , Vascular Diseases , COVID-19/complications , Cytokines/metabolism , Endothelial Cells/metabolism , Endothelium, Vascular/metabolism , Humans , SARS-CoV-2 , Vascular Diseases/metabolism
5.
Front Cell Dev Biol ; 10: 823450, 2022.
Article in English | MEDLINE | ID: mdl-35356285

ABSTRACT

Background: Vascular calcification (VC) is a cardiovascular complication associated with a high mortality rate among patients with diseases such as atherosclerosis and chronic kidney disease. During VC, vascular smooth muscle cells (VSMCs) undergo an osteogenic switch and secrete a heterogeneous population of extracellular vesicles (EVs). Recent studies have shown involvement of EVs in the inflammation and oxidative stress observed in VC. We aimed to decipher the role and mechanism of action of macrophage-derived EVs in the propagation of inflammation and oxidative stress on VSMCs during VC. Methods: The macrophage murine cell line RAW 264.7 treated with lipopolysaccharide (LPS-EK) was used as a cellular model for inflammatory and oxidative stress. EVs secreted by these macrophages were collected by ultracentrifugation and characterized by transmission electron microscopy, cryo-electron microscopy, nanoparticle tracking analysis, and the analysis of acetylcholinesterase activity, as well as that of CD9 and CD81 protein expression by western blotting. These EVs were added to a murine VSMC cell line (MOVAS-1) under calcifying conditions (4 mM Pi-7 or 14 days) and calcification assessed by the o-cresolphthalein calcium assay. EV protein content was analyzed in a proteomic study and EV cytokine content assessed using an MSD multiplex immunoassay. Results: LPS-EK significantly decreased macrophage EV biogenesis. A 24-h treatment of VSMCs with these EVs induced both inflammatory and oxidative responses. LPS-EK-treated macrophage-derived EVs were enriched for pro-inflammatory cytokines and CAD, PAI-1, and Saa3 proteins, three molecules involved in inflammation, oxidative stress, and VC. Under calcifying conditions, these EVs significantly increase the calcification of VSMCs by increasing osteogenic markers and decreasing contractile marker expression. Conclusion: Our results show that EVs derived from LPS-EK-treated-macrophages are able to induce pro-inflammatory and pro-oxidative responses in surrounding cells, such as VSMCs, thus aggravating the VC process.

6.
Front Endocrinol (Lausanne) ; 12: 739287, 2021.
Article in English | MEDLINE | ID: mdl-34690932

ABSTRACT

Background: Obesity is a major public health problem of our time as a risk factor for cardiometabolic disease and the available pharmacological tools needed to tackle the obesity pandemic are insufficient. Neurotensin (NTS) is a 13 amino acid peptide, which is derived from a larger precursor hormone called proneurotensin or Long Form NTS (LF NTS). NTS modulates neuro-transmitter release in the central system nervous, and facilitates intestinal fat absorption in the gastrointestinal tract. Mice lacking LF NTS are protected from high fat diet (HFD) induced obesity, hepatic steatosis and glucose intolerance. In humans, increased levels of LF NTS strongly and independently predict the development of obesity, diabetes mellitus, cardiovascular disease and mortality. With the perspective to develop therapeutic tools to neutralize LF NTS, we developed a monoclonal antibody, specifically inhibiting the function of the LF NTS (LF NTS mAb). This antibody was tested for the effects on body weight, metabolic parameters and behavior in mice made obese by high-fat diet. Methods: C57bl/6j mice were subjected to high-fat diet (HFD) until they reached an obesity state, then food was switched to chow. Mice were treated with either PBS (control therapy) or LF NTS mAb at the dose of 5 mg/kg once a week (i.v.). Mice weight, plasma biochemical analysis, fat and muscle size and distribution and behavioral tests were performed during the losing weight period and the stabilization period. Results: Obese mice treated with the LF NTS mAb lost weight significantly faster than the control treated group. LF NTS mAb treatment also resulted in smaller fat depots, increased fecal cholesterol excretion, reduced liver fat and larger muscle fiber size. Moreover, mice on active therapy were also less stressed, more curious and more active, providing a possible explanation to their weight loss. Conclusion: Our results demonstrate that in mice subjected to HFD-induced obesity, a blockade of LF NTS with a monoclonal antibody results in reduced body weight, adipocyte volume and increased muscle fiber size, possibly explained by beneficial effects on behavior. The underlying mechanisms as well as any future role of LF NTS mAb as an anti-obesity agent warrants further studies.


Subject(s)
Antibodies, Monoclonal/pharmacology , Behavior, Animal/drug effects , Diet, High-Fat/adverse effects , Neurotensin/immunology , Obesity/drug therapy , Weight Loss/drug effects , Adipose Tissue/drug effects , Adipose Tissue/metabolism , Animals , Anti-Obesity Agents/pharmacology , Anti-Obesity Agents/therapeutic use , Antibodies, Monoclonal/therapeutic use , Male , Mice , Muscle, Skeletal/drug effects , Muscle, Skeletal/metabolism , Obesity/etiology , Obesity/metabolism
7.
J Pers Med ; 11(9)2021 Sep 08.
Article in English | MEDLINE | ID: mdl-34575675

ABSTRACT

BACKGROUND: Fabry disease (FD) is an X-linked lysosomal disease due to a deficiency in the activity of the lysosomal α-galactosidase A (GalA), a key enzyme in the glycosphingolipid degradation pathway. FD is a complex disease with a poor genotype-phenotype correlation. FD could involve kidney, heart or central nervous system impairment that significantly decreases life expectancy. The advent of omics technologies offers the possibility of a global, integrated and systemic approach well-suited for the exploration of this complex disease. MATERIALS AND METHODS: Sixty-six plasmas of FD patients from the French Fabry cohort (FFABRY) and 60 control plasmas were analyzed using liquid chromatography and mass spectrometry-based targeted metabolomics (188 metabolites) along with the determination of LysoGb3 concentration and GalA enzymatic activity. Conventional univariate analyses as well as systems biology and machine learning methods were used. RESULTS: The analysis allowed for the identification of discriminating metabolic profiles that unambiguously separate FD patients from control subjects. The analysis identified 86 metabolites that are differentially expressed, including 62 Glycerophospholipids, 8 Acylcarnitines, 6 Sphingomyelins, 5 Aminoacids and 5 Biogenic Amines. Thirteen consensus metabolites were identified through network-based analysis, including 1 biogenic amine, 2 lysophosphatidylcholines and 10 glycerophospholipids. A predictive model using these metabolites showed an AUC-ROC of 0.992 (CI: 0.965-1.000). CONCLUSION: These results highlight deep metabolic remodeling in FD and confirm the potential of omics-based approaches in lysosomal diseases to reveal clinical and biological associations to generate pathophysiological hypotheses.

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.
Front Cell Dev Biol ; 8: 589761, 2020.
Article in English | MEDLINE | ID: mdl-33330469

ABSTRACT

OBJECTIVE: Vascular calcification (VC) is an active process during which vascular smooth muscle cells (VSMCs) undergo an osteogenic switch and release extracellular vesicles (EVs). In turn, the EVs serve as calcification foci via interaction with type 1 collagen (COL1). We recently showed that a specific, six-amino-acid repeat (GFOGER) in the sequence of COL1 was involved in the latter's interaction with integrins expressed on EVs. Our main objective was to test the GFOGER ability to inhibit VC. APPROACH: We synthesized the GFOGER peptide and tested its ability to inhibit the inorganic phosphate (Pi)-induced calcification of VSMCs and aortic rings. Using mass spectrometry, we studied GFOGER's effect on the protein composition of EVs released from Pi-treated VSMCs. RESULTS: Calcification of mouse VSMCs (MOVAS-1 cells), primary human VSMCs, and rat aortic rings was lower in the presence of GFOGER than with Pi alone (with relative decreases of 66, 58, and 91%, respectively; p < 0.001 for all) (no effect was observed with the scramble peptide GOERFG). A comparative proteomic analysis of EVs released from MOVAS-1 cells in the presence or absence of Pi highlighted significant differences in EVs' protein content. Interestingly, the expression of some of the EVs' proteins involved in the calcification process (such as osteogenic markers, TANK-binding kinase 1, and casein kinase II) was diminished in the presence of GFOGER peptide (data are available via ProteomeXchange with identifier PXD018169∗). The decrease of osteogenic marker expression observed in the presence of GFOGER was confirmed by q-RT-PCR analysis. CONCLUSION: GFOGER peptide reduces vascular calcification by modifying the protein content of the subsequently released EVs, in particular by decreasing osteogenicswitching in VSMCs.

10.
Toxins (Basel) ; 12(12)2020 Dec 21.
Article in English | MEDLINE | ID: mdl-33371311

ABSTRACT

Vascular calcification (VC) is a cardiovascular complication associated with a high mortality rate, especially in patients with diabetes, atherosclerosis or chronic kidney disease (CKD). In CKD patients, VC is associated with the accumulation of uremic toxins, such as indoxyl sulphate or inorganic phosphate, which can have a major impact in vascular remodeling. During VC, vascular smooth muscle cells (VSMCs) undergo an osteogenic switch and secrete extracellular vesicles (EVs) that are heterogeneous in terms of their origin and composition. Under physiological conditions, EVs are involved in cell-cell communication and the maintenance of cellular homeostasis. They contain high levels of calcification inhibitors, such as fetuin-A and matrix Gla protein. Under pathological conditions (and particularly in the presence of uremic toxins), the secreted EVs acquire a pro-calcifying profile and thereby act as nucleating foci for the crystallization of hydroxyapatite and the propagation of calcification. Here, we review the most recent findings on the EVs' pathophysiological role in VC, the impact of uremic toxins on EV biogenesis and functions, the use of EVs as diagnostic biomarkers and the EVs' therapeutic potential in CKD.


Subject(s)
Extracellular Vesicles/metabolism , Renal Insufficiency, Chronic/metabolism , Toxins, Biological/metabolism , Uremia/metabolism , Animals , Biomarkers/metabolism , Extracellular Vesicles/drug effects , Humans , Renal Insufficiency, Chronic/complications , Toxins, Biological/toxicity , Uremia/complications , Vascular Calcification/etiology , Vascular Calcification/metabolism
11.
Pharmaceuticals (Basel) ; 12(4)2019 Oct 27.
Article in English | MEDLINE | ID: mdl-31717866

ABSTRACT

There is an urgent need to propose effective treatments for Alzheimer's disease (AD). Although the origin of the disease is poorly understood, several therapeutic options have been proposed. The new therapeutic approaches targeting biometal-mediated neurodegenerative pathways appear to be interesting ones. As a continuation of our preceding studies, two novel series of advanced glycation endproducts (AGE)/advanced lipid peroxidation endproducts (ALE) inhibitors have been developed as multifunctional scavengers. This extended work allowed us to highlight the new hydroxypyridinone-diamine hybrid IIa-3 bearing a C4 alkyl linker between the two pharmacophores. This derivative exhibited preserved potent capacities to trap reactive carbonyl species (vicinal diamine function) as well as reactive oxygen species and transition metals (hydroxypyridinone moiety) in comparison with previously described lead compound 1. In addition, its good predicted absorption, distribution, metabolism and excretion (ADME) properties were correlated with a better efficacy to inhibit in vitro methylglyoxal-induced apoptosis in neuronal-like PC12 cells. This new promising agent revealed improved druglikeness and ability to prevent biometal-mediated oxidative and carbonyl stress amplification involved in AD pathogenesis.

12.
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
13.
Cancer Lett ; 444: 147-161, 2019 03 01.
Article in English | MEDLINE | ID: mdl-30583074

ABSTRACT

Overall survival of patients with metastatic non-small cell lung cancer (NSCLC) has significantly improved with platinum-based salt treatments and recently with targeted therapies and immunotherapies. However, treatment failure occurs due to acquired or emerging tumor resistance. We developed a monoclonal antibody against the proform of neurotensin (LF-NTS mAb) that alters the homeostasis of tumors overexpressing NTSR1. Neurotensin is frequently overexpressed along with its high affinity receptor (NTSR1) in tumors from epithelial origins. This ligand/receptor complex contributes to the progression of many tumor types by activation of the cellular effects involved in tumor progression (proliferation, survival, migration, and invasion). We demonstrate that LF-NTS mAb operates on the plasticity of tumor cells overexpressing NTSR1 and lowers their aggressiveness. The mAb enables the restoration of platinum-based therapies responsiveness, while also decreasing metastatic processes. Efficacy dosage with long-term treatment showed no obvious adverse events, while demonstrating improvement in the performance status. Our data suggests that LF-NTS mAb is an ideal candidate to be safely added to the conventional standard of care in order to improve its efficacy.


Subject(s)
Adenocarcinoma of Lung/drug therapy , Antibodies, Monoclonal/pharmacology , Carcinoma, Non-Small-Cell Lung/drug therapy , Cisplatin/pharmacology , Drug Resistance, Neoplasm , Neurotensin/antagonists & inhibitors , Receptors, Neurotensin/antagonists & inhibitors , Adenocarcinoma of Lung/immunology , Adenocarcinoma of Lung/metabolism , Adenocarcinoma of Lung/pathology , Animals , Antineoplastic Agents/pharmacology , Apoptosis , Carcinoma, Non-Small-Cell Lung/immunology , Carcinoma, Non-Small-Cell Lung/metabolism , Carcinoma, Non-Small-Cell Lung/pathology , Cell Proliferation , Female , Follow-Up Studies , Humans , Lung Neoplasms/drug therapy , Lung Neoplasms/immunology , Lung Neoplasms/metabolism , Lung Neoplasms/pathology , Male , Mice , Mice, Inbred C57BL , Mice, Nude , Neurotensin/immunology , Neurotensin/metabolism , Prognosis , Receptors, Neurotensin/immunology , Receptors, Neurotensin/metabolism , Retrospective Studies , Survival Rate , Tumor Cells, Cultured , Xenograft Model Antitumor Assays
14.
Toxins (Basel) ; 10(10)2018 10 07.
Article in English | MEDLINE | ID: mdl-30301260

ABSTRACT

Endothelial dysfunction in uremia can result in cell-to-cell junction loss and increased permeability, contributing to cardiovascular diseases (CVD) development. This study evaluated the impact of the uremic milieu on endothelial morphology and cell junction's proteins. We evaluated (i) serum levels of inflammatory biomarkers in a cohort of chronic kidney disease (CKD) patients and the expression of VE-cadherin and Zonula Occludens-1 (ZO-1) junction proteins on endothelial cells (ECs) of arteries removed from CKD patients during renal transplant; (ii) ECs morphology in vitro under different uremic conditions, and (iii) the impact of uremic toxins p-cresyl sulfate (PCS), indoxyl sulfate (IS), and inorganic phosphate (Pi) as well as of total uremic serum on VE-cadherin and ZO-1 gene and protein expression in cultured ECs. We found that the uremic arteries had lost their intact and continuous endothelial morphology, with a reduction in VE-cadherin and ZO-1 expression. In cultured ECs, both VE-cadherin and ZO-1 protein expression decreased, mainly after exposure to Pi and uremic serum groups. VE-cadherin mRNA expression was reduced while ZO-1 was increased after exposure to PCS, IS, Pi, and uremic serum. Our findings show that uremia alters cell-to-cell junctions leading to an increased endothelial damage. This gives a new perspective regarding the pathophysiological role of uremia in intercellular junctions and opens new avenues to improve cardiovascular outcomes in CKD patients.


Subject(s)
Antigens, CD/metabolism , Cadherins/metabolism , Endothelial Cells/physiology , Intercellular Junctions/physiology , Renal Artery/physiopathology , Renal Insufficiency, Chronic/physiopathology , Uremia/physiopathology , Zonula Occludens-1 Protein/metabolism , Cell Line , Cresols/pharmacology , Endothelial Cells/drug effects , Female , Humans , Indican/pharmacology , Male , Middle Aged , Phosphates/pharmacology , Renal Artery/metabolism , Renal Insufficiency, Chronic/blood , Sulfuric Acid Esters/pharmacology , Toxins, Biological/pharmacology , Uremia/blood
15.
Glia ; 66(8): 1709-1723, 2018 08.
Article in English | MEDLINE | ID: mdl-29624734

ABSTRACT

Neuroinflammation and iron accumulation are hallmarks of a variety of adult neurodegenerative diseases. In Sanfilippo syndrome (mucopolysaccharidosis type III, MPSIII, a pediatric neurodegenerative disease that shares some features with adult neurodegenerative diseases), the progressive accumulation of heparan sulfate oligosaccharides (HSOs) induces microglia and astrocytes to produce pro-inflammatory cytokines leading to severe neuroinflammation. The objectives of the present study were (1) to measure the local iron concentration and to assess iron metabolism in the brain of a MPSIIIB murine model and (2) to identify the brain cells involved in this accumulation. We found that iron accumulation in MPSIIIB mice primarily affected the cerebral cortex where hepcidin levels were higher than in wild-type mice, and increased with aging. This increase was correlated with low expression of ferroportin 1 (FPN1), and thus brain iron retention. Moreover, we showed in vitro that HSOs are directly responsible for the production of hepcidin and the relative decrease in FPN1 expression when added to cultures of microglia and, to a lesser extent, to cultures of astrocytes. In contrast, no significant differences were observed in neurons. Hepcidin induction results from activation of the TLR4 pathway and STAT3 signaling, and leads to iron retention within microglia. Our results show that microglia have a key role in cerebral hepcidin overexpression and thus in the brain iron accumulation observed in the MPSIIIB model.


Subject(s)
Brain/metabolism , Iron/metabolism , Microglia/metabolism , Mucopolysaccharidosis III/metabolism , Animals , Astrocytes/metabolism , Mice, Knockout , Neurodegenerative Diseases/metabolism , Neurons/metabolism
16.
Arterioscler Thromb Vasc Biol ; 37(7): 1391-1401, 2017 07.
Article in English | MEDLINE | ID: mdl-28522698

ABSTRACT

OBJECTIVE: Cardiovascular diseases constitute the leading cause of mortality worldwide. Calcification of the vessel wall is associated with cardiovascular morbidity and mortality in patients having many diseases, including diabetes mellitus, atherosclerosis, and chronic kidney disease. Vascular calcification is actively regulated by inductive and inhibitory mechanisms (including vascular smooth muscle cell adaptation) and results from an active osteogenic process. During the calcification process, extracellular vesicles (also known as matrix vesicles) released by vascular smooth muscle cells interact with type I collagen and then act as nucleating foci for calcium crystallization. Our primary objective was to identify new, natural molecules that inhibit the vascular calcification process. APPROACH AND RESULTS: We have found that oligogalacturonic acids (obtained by the acid hydrolysis of polygalacturonic acid) reduce in vitro inorganic phosphate-induced calcification of vascular smooth muscle cells by 80% and inorganic phosphate-induced calcification of isolated rat aortic rings by 50%. A specific oligogalacturonic acid with a degree of polymerization of 8 (DP8) was found to inhibit the expression of osteogenic markers and, thus, prevent the conversion of vascular smooth muscle cells into osteoblast-like cells. We also evidenced in biochemical and immunofluorescence assays a direct interaction between matrix vesicles and type I collagen via the GFOGER sequence (where single letter amino acid nomenclature is used, O=hydroxyproline) thought to be involved in interactions with several pairs of integrins. CONCLUSIONS: DP8 inhibits vascular calcification development mainly by inhibition of osteogenic marker expression but also partly by masking the GFOGER sequence-thereby, preventing matrix vesicles from binding to type I collagen.


Subject(s)
Aortic Diseases/prevention & control , Calcium/metabolism , Cell Transdifferentiation/drug effects , Collagen Type I/metabolism , Muscle, Smooth, Vascular/drug effects , Myocytes, Smooth Muscle/drug effects , Oligosaccharides/pharmacology , Osteogenesis/drug effects , Vascular Calcification/prevention & control , Animals , Aorta, Thoracic/drug effects , Aorta, Thoracic/metabolism , Aorta, Thoracic/pathology , Aortic Diseases/metabolism , Aortic Diseases/pathology , Binding Sites , Biomarkers/metabolism , Cells, Cultured , Crystallization , Dose-Response Relationship, Drug , Extracellular Vesicles/drug effects , Extracellular Vesicles/metabolism , Male , Mice , Muscle, Smooth, Vascular/metabolism , Muscle, Smooth, Vascular/pathology , Myocytes, Smooth Muscle/metabolism , Myocytes, Smooth Muscle/pathology , Protein Binding , Protein Interaction Domains and Motifs , Rats, Wistar , Signal Transduction/drug effects , Tissue Culture Techniques , Vascular Calcification/metabolism , Vascular Calcification/pathology
17.
Hepatology ; 65(6): 1851-1864, 2017 06.
Article in English | MEDLINE | ID: mdl-28152568

ABSTRACT

The development of different cell culture models has greatly contributed to increased understanding of the hepatitis C virus (HCV) life cycle. However, it is still challenging to grow HCV clinical isolates in cell culture. If overcome, this would open new perspectives to study HCV biology, including drug-resistant variants emerging with new antiviral therapies. In this study we hypothesized that this hurdle could be due to the presence of inhibitory factors in patient serum. Combining polyethylene glycol precipitation, iodixanol gradient, and size-exclusion chromatography, we obtained from HCV-seronegative sera a purified fraction enriched in inhibitory factors. Mass spectrometric analysis identified apolipoprotein(a) (apo[a]) as a potential inhibitor of HCV entry. Apo(a) consists of 10 kringle IV domains (KIVs), one kringle V domain, and an inactive protease domain. The 10 KIVs are present in a single copy with the exception of KIV type 2 (KIV2 ), which is encoded in a variable number of tandemly repeated copies, giving rise to numerous apo(a) size isoforms. In addition, apo(a) covalently links to the apolipoprotein B component of a low-density lipoprotein through a disulfide bridge to form lipoprotein(a). Using a recombinant virus derived from the JFH1 strain, we confirmed that plasma-derived and recombinant lipoprotein(a) as well as purified recombinant apo(a) variants were able to specifically inhibit HCV by interacting with infectious particles. Our results also suggest that small isoforms are less inhibitory than the large ones. Finally, we observed that the lipoprotein moiety of HCV lipoviroparticles was essential for inhibition, whereas functional lysine-binding sites in KIV7 , KIV8 , and KIV10 were not required. CONCLUSIONS: Our results identify apo(a) as an additional component of the lipid metabolism modulating HCV infection. (Hepatology 2017;65:1851-1864).


Subject(s)
Hepacivirus/metabolism , Hepatitis C/therapy , Lipoprotein(a)/pharmacology , Blotting, Western , Cell Survival/drug effects , Cells, Cultured , Chromatography, Liquid/methods , Hepacivirus/drug effects , Hepatitis C/blood , Hepatocytes/drug effects , Hepatocytes/metabolism , Humans , Immunoprecipitation , Lysine/metabolism , Protein Binding , Sensitivity and Specificity , Structure-Activity Relationship
18.
Toxins (Basel) ; 8(11)2016 11 15.
Article in English | MEDLINE | ID: mdl-27854278

ABSTRACT

N-methyl-2-pyridone-5-carboxamide (2PY, a major metabolite of nicotinamide, NAM) was recently identified as a uremic toxin. Recent interventional trials using NAM to treat high levels of phosphorus in end-stage renal disease have highlighted new potential uremic toxicities of 2PY. In the context of uremia, the accumulation of 2PY could be harmful-perhaps by inhibiting poly (ADP-ribose) polymerase-1 activity. Here, we review recently published data on 2PY's metabolism and toxicological profile.


Subject(s)
Niacinamide/analogs & derivatives , Niacinamide/metabolism , Toxins, Biological/metabolism , Uremia/metabolism , Animals , Humans , Renal Insufficiency, Chronic/metabolism
19.
Eur J Med Chem ; 122: 702-722, 2016 Oct 21.
Article in English | MEDLINE | ID: mdl-27451257

ABSTRACT

An important part of pathogenesis of Alzheimer's disease (AD) is attributed to the contribution of AGE (Advanced Glycation Endproducts) and ALE (Advanced Lipid peroxidation Endproducts). In order to attenuate the progression of AD, we designed a new type of molecules that consist of two trapping parts for reactive carbonyl species (RCS) and reactive oxygen species (ROS), precursors of AGE and ALE, respectively. These molecules also chelate transition metals, the promoters of ROS formation. In this paper, synthesis of the new AGE/ALE inhibitors and evaluation of their physicochemical and biological properties (carbonyl trapping capacity, antioxidant activity, Cu(2+)-chelating capacity, cytotoxicity and protective effect against in vitro MGO-induced apoptosis in the model AD cell-line PC12) are described. It is found that compounds 40b and 51e possess promising therapeutic potentials for treating AD.


Subject(s)
Alzheimer Disease/drug therapy , Diamines/chemistry , Diamines/pharmacology , Glycation End Products, Advanced/antagonists & inhibitors , Lipid Peroxidation/drug effects , Animals , Apoptosis/drug effects , Cell Line, Tumor , Cell Survival/drug effects , Copper/chemistry , Diamines/therapeutic use , Malondialdehyde/metabolism , PC12 Cells , Pyruvaldehyde/metabolism , Rats , Reactive Oxygen Species/metabolism
20.
J Am Soc Nephrol ; 27(2): 354-70, 2016 Feb.
Article in English | MEDLINE | ID: mdl-26311460

ABSTRACT

Advanced glycation end products (AGEs), a heterogeneous group of compounds formed by nonenzymatic glycation reactions between reducing sugars and amino acids, lipids, or DNA, are formed not only in the presence of hyperglycemia, but also in diseases associated with high levels of oxidative stress, such as CKD. In chronic renal failure, higher circulating AGE levels result from increased formation and decreased renal clearance. Interactions between AGEs and their receptors, including advanced glycation end product-specific receptor (RAGE), trigger various intracellular events, such as oxidative stress and inflammation, leading to cardiovascular complications. Although patients with CKD have a higher burden of cardiovascular disease, the relationship between AGEs and cardiovascular disease in patients with CKD is not fully characterized. In this paper, we review the various deleterious effects of AGEs in CKD that lead to cardiovascular complications and the role of these AGEs in diabetic nephropathy. We also discuss potential pharmacologic approaches to circumvent these deleterious effects by reducing exogenous and endogenous sources of AGEs, increasing the breakdown of existing AGEs, or inhibiting AGE-induced inflammation. Finally, we speculate on preventive and therapeutic strategies that focus on the AGE-RAGE axis to prevent vascular complications in patients with CKD.


Subject(s)
Glycation End Products, Advanced/toxicity , Renal Insufficiency, Chronic/complications , Renal Insufficiency, Chronic/metabolism , Uremia/chemically induced , Uremia/complications , Cardiovascular Diseases/etiology , Humans , Inflammation/etiology , Oxidative Stress , Uremia/therapy
SELECTION OF CITATIONS
SEARCH DETAIL
...