Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 20 de 32
1.
Cell Death Dis ; 13(12): 1046, 2022 12 15.
Article En | MEDLINE | ID: mdl-36522309

Interleukin-8 (IL-8/CXCL8) is a pro-angiogenic and pro-inflammatory chemokine that plays a role in cancer development. Non-small cell lung carcinoma (NSCLC) produces high amounts of IL-8, which is associated with poor prognosis and resistance to chemo-radio and immunotherapy. However, the signaling pathways that lead to IL-8 production in NSCLC are unresolved. Here, we show that expression and release of IL-8 are regulated autonomously by TRAIL death receptors in several squamous and adenocarcinoma NSCLC cell lines. NSCLC constitutively secrete IL-8, which could be further enhanced by glucose withdrawal or by treatment with TRAIL or TNFα. In A549 cells, constitutive and inducible IL-8 production was dependent on NF-κB and MEK/ERK MAP Kinases. DR4 and DR5, known regulators of these signaling pathways, participated in constitutive and glucose deprivation-induced IL-8 secretion. These receptors were mainly located intracellularly. While DR4 signaled through the NF-κB pathway, DR4 and DR5 both regulated the ERK-MAPK and Akt pathways. FADD, caspase-8, RIPK1, and TRADD also regulated IL-8. Analysis of mRNA expression data from patients indicated that IL-8 transcripts correlated with TRAIL, DR4, and DR5 expression levels. Furthermore, TRAIL receptor expression levels also correlated with markers of angiogenesis and neutrophil infiltration in lung squamous carcinoma and adenocarcinoma. Collectively, these data suggest that TRAIL receptor signaling contributes to a pro-tumorigenic inflammatory signature associated with NSCLC.


Adenocarcinoma , Carcinoma, Non-Small-Cell Lung , Lung Neoplasms , Humans , Receptors, TNF-Related Apoptosis-Inducing Ligand/genetics , Receptors, TNF-Related Apoptosis-Inducing Ligand/metabolism , Carcinoma, Non-Small-Cell Lung/pathology , Interleukin-8/genetics , Interleukin-8/metabolism , NF-kappa B/metabolism , TNF-Related Apoptosis-Inducing Ligand/metabolism , Cell Line, Tumor , Lung Neoplasms/pathology , Glucose , Apoptosis
2.
Int J Mol Sci ; 23(14)2022 Jul 15.
Article En | MEDLINE | ID: mdl-35887177

The surgically induced remission of liver disease represents a model to investigate the signalling processes that trigger the development of nonalcoholic steatohepatitis with the aim of identifying novel therapeutic targets. We recruited patients with severe obesity with or without nonalcoholic steatohepatitis and obtained liver and plasma samples before and after laparoscopic sleeve gastrectomy for immunoblotting, immunocytochemical, metabolomic, transcriptomic and epigenetic analyses. Functional studies were performed in HepG2 cells and primary hepatocytes. Surgery was associated with a decrease in the inflammatory response and revealed the role of mitogen-activated protein kinases. Nonalcoholic steatohepatitis was associated with an increased glutaminolysis-induced production of α-ketoglutarate and the hyperactivation of mammalian target of rapamycin complex 1. These changes were crucial for adenosine monophosphate-activated protein kinase/mammalian target of rapamycin-driven pathways that modulated hepatocyte survival by coordinating apoptosis and autophagy and affected methylation-related epigenomic remodelling enzymes. Hepatic transcriptome signatures and differentially methylated genomic regions distinguished patients with and without steatohepatitis. Our results suggest that the increased glutaminolysis-induced α-ketoglutarate production and the mammalian target of rapamycin complex 1 dysregulation play a crucial role in the inefficient adaptive responses leading to steatohepatitis in obesity.


Laparoscopy , Non-alcoholic Fatty Liver Disease , Obesity, Morbid , Gastrectomy/methods , Humans , Ketoglutaric Acids , Non-alcoholic Fatty Liver Disease/complications , Non-alcoholic Fatty Liver Disease/genetics , Obesity, Morbid/surgery , TOR Serine-Threonine Kinases
3.
J Hepatol ; 2021 May 04.
Article En | MEDLINE | ID: mdl-33961941

BACKGROUND & AIMS: A holistic insight on the relationship between obesity and metabolic dysfunction-associated fatty liver disease is an unmet clinical need. Omics investigations can be used to investigate the multifaceted role of altered mitochondrial pathways to promote nonalcoholic steatohepatitis, a major risk factor for liver disease-associated death. There are no specific treatments but remission via surgery might offer an opportunity to examine the signaling processes that govern the complex spectrum of chronic liver diseases observed in extreme obesity. We aim to assess the emerging relationship between metabolism, methylation and liver disease. METHODS: We tailed the flow of information, before and after steatohepatitis remission, from biochemical, histological, and multi-omics analyses in liver biopsies from patients with extreme obesity and successful bariatric surgery. Functional studies were performed in HepG2 cells and primary hepatocytes. RESULTS: The reversal of hepatic mitochondrial dysfunction and the control of oxidative stress and inflammatory responses revealed the regulatory role of mitogen-activated protein kinases. The reversible metabolic rearrangements leading to steatohepatitis increased the glutaminolysis-induced production of α-ketoglutarate and the hyperactivation of mammalian target of rapamycin complex 1. These changes were crucial for the adenosine monophosphate-activated protein kinase/mammalian target of rapamycin-driven pathways that modulated hepatocyte survival by coordinating apoptosis and autophagy. The signaling activity of α-ketoglutarate and the associated metabolites also affected methylation-related epigenomic remodeling enzymes. Integrative analysis of hepatic transcriptome signatures and differentially methylated genomic regions distinguished patients with and without steatohepatitis. CONCLUSION: We provide evidence supporting the multifaceted potential of the increased glutaminolysis-induced α-ketoglutarate production and the mammalian target of rapamycin complex 1 dysregulation as a conceivable source of the inefficient adaptive responses leading to steatohepatitis. LAY SUMMARY: Steatohepatitis is a frequent and threatening complication of extreme obesity without specific treatment. Omics technologies can be used to identify therapeutic targets. We highlight increased glutaminolysis-induced α-ketoglutarate production as a potential source of signals promoting and exacerbating steatohepatitis.

4.
Sci Rep ; 11(1): 1343, 2021 01 14.
Article En | MEDLINE | ID: mdl-33446705

Patients with morbid obesity frequently present non-alcoholic fatty liver (NAFL) and non-alcoholic steatohepatitis (NASH) associated with pro-atherogenic alterations. Laparoscopic sleeve gastrectomy (LSG) is an effective treatment for weight reduction, and for the remission of hepatic alterations. Using 1H-nuclear magnetic resonance (1H-NMR), we investigated the effects of LSG on lipoprotein and glycoprotein profile in patients with morbid obesity and liver disease. We included 154 patients with morbid obesity (49 non-NASH, 54 uncertain NASH, 51 definite NASH). A blood sample was obtained before surgery and, in patients with definite NASH, one year after surgery. Patients with NASH had increased concentrations of medium and small VLDL particles, VLDL and IDL cholesterol concentrations, IDL, LDL, and HDL triglyceride concentrations, and elevated glycoprotein levels. These changes were more marked in patients with type 2 diabetes mellitus. LSG produced significant decreases in the concentration of VLDL particles, VLDL cholesterol and triglycerides, an increase in the concentration LDL particles and LDL cholesterol concentrations, and a decrease in protein glycation. We conclude that patients with obesity and NASH had significant alterations in circulating levels of lipoproteins and glycoproteins that were associated with the severity of the disease. Most of these changes were reversed post-LSG.


Diabetes Mellitus, Type 2 , Glycoproteins/blood , Laparoscopy , Lipoproteins/blood , Non-alcoholic Fatty Liver Disease , Obesity, Morbid , Adult , Diabetes Mellitus, Type 2/blood , Diabetes Mellitus, Type 2/surgery , Female , Humans , Male , Middle Aged , Non-alcoholic Fatty Liver Disease/blood , Non-alcoholic Fatty Liver Disease/surgery , Obesity, Morbid/blood , Obesity, Morbid/surgery , Retrospective Studies
5.
J Nutr Biochem ; 89: 108559, 2021 03.
Article En | MEDLINE | ID: mdl-33264665

BACKGROUND: Obesity is a chronic progressive disease with several metabolic alterations. Nonalcoholic fatty liver disease (NAFLD) is an important comorbidity of obesity that can progress to nonalcoholic steatohepatitis (NASH), cirrhosis or hepatocarcinoma. This study aimed at clarifying the molecular mechanisms underlying the metabolic alterations in hepatic and adipose tissue during high-fat high-sucrose diet-induced NAFLD development in mice. METHODS: Twenty-four male mice (C57BL/6J) were randomly allocated into 3 groups (n = 8 mice per group) to receive a chow diet, a high-fat diet (HFD), or a high-fat high-sucrose diet (HF-HSD) for 20 weeks. At sacrifice, liver and adipose tissue were obtained for histopathological, metabolomic, and protein expression analyses. RESULTS: HF-HSD (but not HFD) was associated with NASH and increased oxidative stress. These animals presented an inhibition of hepatic autophagy and alterations in AMP-activated protein kinase/mammalian target of rapamycin activity. We also observed that the ability of metabolic adaptation was adversely affected by the increase of damaged mitochondria. NASH development was associated with changes in adipose tissue dynamics and increased amounts of saturated fatty acids, monounsaturated fatty acids and polyunsaturated fatty acids in visceral adipose tissue. CONCLUSION: HF-HSD led to a metabolic blockage and impaired hepatic mitochondria turnover. In addition, the continuous accumulation of fatty acids produced adipose tissue dysfunction and hepatic fat accumulation that favored the progression to NASH.


Adipose Tissue/metabolism , Diet, High-Fat/adverse effects , Dietary Sucrose/adverse effects , Liver/metabolism , Non-alcoholic Fatty Liver Disease/metabolism , AMP-Activated Protein Kinases/metabolism , Adipose Tissue/pathology , Animals , Autophagy , Disease Models, Animal , Fatty Acids/metabolism , Intra-Abdominal Fat/metabolism , Liver/pathology , Liver Cirrhosis/metabolism , Male , Mice , Mice, Inbred C57BL , Mitochondria/metabolism , Non-alcoholic Fatty Liver Disease/pathology , Obesity/metabolism , TOR Serine-Threonine Kinases/metabolism
6.
Aging (Albany NY) ; 12(20): 20001-20023, 2020 10 26.
Article En | MEDLINE | ID: mdl-33104522

Injection of tissues with senescent cells induces changes that mimic aging, and this process is delayed in mice engineered to eliminate senescent cells, which secrete proinflammatory cytokines, including C-C motif chemokine ligand 2 (Ccl2). Circulating levels of Ccl2 correlate with age, but the impact of Ccl2 on tissue homeostasis has not been established. We generated an experimental model by crossbreeding mice overexpressing Ccl2 with progeroid mice bearing a mutation in the lamin A (Lmna) gene. Wild-type animals and progeroid mice that do not overexpress Ccl2 were used as controls. Ccl2 overexpression decreased the lifespan of the progeroid mice and induced the dysregulation of glycolysis, the citric acid cycle and one-carbon metabolism in skeletal muscle, driving dynamic changes in energy metabolism and DNA methylation. This impact on cellular bioenergetics was associated with mitochondrial alterations and affected cellular metabolism, autophagy and protein synthesis through AMPK/mTOR pathways. The data revealed the ability of Ccl2 to promote death in mice with accelerated aging, which supports its putative use as a biomarker of an increased senescent cell burden and for the assessment of the efficacy of interventions aimed at extending healthy aging.


Cellular Senescence , Chemokine CCL2/metabolism , Energy Metabolism , Mitochondria, Muscle/metabolism , Muscle, Skeletal/metabolism , Progeria/metabolism , Animals , Animals, Genetically Modified , Autophagy , Chemokine CCL2/genetics , DNA Methylation , Disease Models, Animal , Lamin Type A/genetics , Longevity , Male , Mice, Inbred C57BL , Mice, Mutant Strains , Mitochondria, Muscle/genetics , Mitochondria, Muscle/pathology , Mitochondrial Dynamics , Muscle, Skeletal/pathology , Mutation , Progeria/genetics , Progeria/pathology , Signal Transduction , Up-Regulation
7.
Sci Rep ; 10(1): 11954, 2020 07 20.
Article En | MEDLINE | ID: mdl-32686726

Chemokine (C-C motif) ligand 2 (CCL2) has been associated with chronic metabolic diseases. We aimed to investigate whether Ccl2 gene overexpression is involved in the regulation of signaling pathways in metabolic organs. Biochemical and histological analyses were used to explore tissue damage in cisgenic mice that overexpressed the Ccl2 gene. Metabolites from energy and one-carbon metabolism in liver and muscle extracts were measured by targeted metabolomics. Western blot analysis was used to explore the AMP-activated protein kinase (AMPK) and mammalian target of rapamycin pathways. Ccl2 overexpression resulted in steatosis, decreased AMPK activity and altered mitochondrial dynamics in the liver. These changes were associated with decreased oxidative phosphorylation and alterations in the citric acid cycle and transmethylation. In contrast, AMPK activity and its downstream mediators were increased in muscle, where we observed an increase in oxidative phosphorylation and increased concentrations of different metabolites associated with ATP synthesis. In conclusion, Ccl2 overexpression induces distinct metabolic alterations in the liver and muscle that affect mitochondrial dynamics and the regulation of energy sensors involved in cell homeostasis. These data suggest that CCL2 may be a therapeutic target in metabolic diseases.


Chemokine CCL2/genetics , Energy Metabolism , Gene Expression , Liver/metabolism , Muscles/metabolism , Animals , Autophagy , Biopsy , Chemokine CCL2/metabolism , Male , Metabolic Networks and Pathways , Mice , Models, Biological , Organ Specificity , Phenotype , Signal Transduction
8.
Aliment Pharmacol Ther ; 51(3): 374-387, 2020 02.
Article En | MEDLINE | ID: mdl-31825539

BACKGROUND: Obesity can influence hepatic mitochondrial function, and cause non-alcoholic steatohepatitis (NASH). Diagnosis and follow-up rely on invasive liver biopsy so blood-based markers are urgently required. AIM: To investigate whether values of circulating metabolites from energy and one-carbon (1-C) metabolism may: (a) reflect hepatic mitochondrial flexibility failure and (b) act as NASH biomarkers. METHODS: Patients with severe obesity undergoing bariatric surgery (n = 270) were investigated using quantitative targeted plasma metabolomics. Comparisons were with non-obese controls without liver disease (n = 50). Obese patients with NASH (n = 53) and without NASH (n = 130) representing extreme groups of liver disease were assessed to test the diagnostic ability of the measured circulating metabolites. Paired liver biopsy and plasma samples from NASH patients were available 1 year post-surgery and were evaluated to monitor metabolomic changes with liver damage resolution. RESULTS: We identified correlations between human liver metabolism and obesity. High-plasma α-ketoglutarate (α-KG) and lactate concentrations in NASH patients indicating citric acid cycle replenishment via glutaminolysis might also be a crucial point in NASH onset. Plasma measurements of α-KG, ß-hydroxybutyrate, pyruvate and oxaloacetate reduced the uncertainty in clinical diagnosis of NASH [area under receiver operating characteristic curve (AUC) of 0.826] and predicted NASH resolution without ambiguity (AUC of 0.999). CONCLUSION: Changes in plasma mitochondrial metabolites appear to be associated with NASH. These metabolic responses may be dynamically remodelled following resolution of liver damage through massive weight loss.


Biomarkers/blood , Metabolome , Non-alcoholic Fatty Liver Disease/blood , Non-alcoholic Fatty Liver Disease/complications , Obesity, Morbid/blood , Obesity, Morbid/complications , Adult , Bariatric Surgery , Biomarkers/metabolism , Biopsy , Female , Humans , Intraoperative Period , Liver/metabolism , Liver/pathology , Male , Metabolomics/methods , Middle Aged , Mitochondria, Liver/metabolism , Mitochondria, Liver/pathology , Non-alcoholic Fatty Liver Disease/diagnosis , Non-alcoholic Fatty Liver Disease/pathology , Obesity, Morbid/surgery , Weight Loss/physiology
9.
Cytokine ; 126: 154923, 2020 02.
Article En | MEDLINE | ID: mdl-31739217

Chemokines, particularly chemokine (C-C- motif) ligand 2 (CCL2), control leukocyte migration into the wall of the artery and regulate the traffic of inflammatory cells. CCL2 is bound to functional receptors (CCR2), but also to atypical chemokine receptors (ACKRs), which do not induce cell migration but can modify chemokine gradients. Whether atherosclerosis alters CCL2 function by influencing the expression of these receptors remains unknown. In a necropsy study, we used immunohistochemistry to explore where and to what extent CCL2 and related receptors are present in diseased arteries that caused the death of men with coronary artery disease compared with unaffected arteries. CCL2 was marginally detected in normal arteries but was more frequently found in the intima. The expression of CCL2 and related receptors was significantly increased in diseased arteries with relative differences among the artery layers. The highest relative increases were those of CCL2 and ACKR1. CCL2 expression was associated with a significant predictive value of atherosclerosis. Findings suggest the need for further insight into receptor specificity or activity and the interplay among chemokines. CCL2-associated conventional and atypical receptors are overexpressed in atherosclerotic arteries, and these may suggest new potential therapeutic targets to locally modify the overall anti-inflammatory response.


Atherosclerosis/pathology , Chemokine CCL2/metabolism , Coronary Artery Disease/pathology , Duffy Blood-Group System/metabolism , Receptors, CCR2/metabolism , Receptors, Cell Surface/metabolism , Adult , Aged , Humans , Inflammation/pathology , Male , Middle Aged , Prognosis , Receptors, Chemokine/metabolism
10.
Antioxidants (Basel) ; 8(7)2019 Jul 10.
Article En | MEDLINE | ID: mdl-31295833

We investigated alterations in the levels of the antioxidant paraoxonase-1 (PON1) and the lipoprotein profile (analyzed by nuclear magnetic resonance) in patients with lung cancer (LC) or head and neck cancer (HNC), and the effects produced thereon by radiotherapy (RT). We included 33 patients with LC and 28 patients with HNC. Before irradiation, and one month after completion of RT, blood samples were obtained. The control group was composed of 50 healthy subjects. Patients had significantly lower serum PON1 activity and concentration before RT than the control group. PON1-related variables were good predictors of the presence of LC or HNC, with analytical sensitivities and specificities greater than 80%. Patients showed a significant increase in the number of particles of all subclasses of very-low-density lipoproteins (large, medium and small). However, these changes were not maintained when adjusted for age, sex, and other clinical and demographic variables. Irradiation was associated with a significant increase in PON1 concentration and, only in patients with HNC, with an increase in high-density lipoprotein-cholesterol concentration. Our results suggest that determinations of the levels of PON1-related variables may constitute good biomarkers for the evaluation of these diseases. Studies with a larger number of patients are needed to fully confirm this hypothesis.

11.
Metabolism ; 99: 81-89, 2019 10.
Article En | MEDLINE | ID: mdl-31279739

BACKGROUND & AIMS: Hepatic alterations, such as in non-alcoholic fatty liver disease (NAFLD) and non-alcoholic steatohepatitis (NASH) are frequently associated with obesity. To investigate the molecular mechanisms of these alterations and to identify molecules that could be used as potential therapeutic targets, we investigated the modulation of hepatic indices of oxidative stress and inflammation in obese patients undergoing laparoscopic sleeve gastrectomy (LSG). METHODS: Patients (n = 436) attending our obesity clinic underwent LSG for weight loss. We obtained a diagnostic intraoperative liver biopsy, and a sub-cohort (n = 120) agreed to a 1-year follow-up that included donation of blood samples and additional liver biopsies. Selected key molecules in blood and liver tissue were used to investigate the hepatic alterations in obesity, and their response to LSG. RESULTS: One year post-surgery, the prevalence of diabetes, dyslipidemia and hypertension decreased significantly. LSG improved liver histology features in all patients. Improvement was greater in severe cases of NAFLD including those with steatohepatitis, bridging fibrosis or cirrhosis. Significant pre-surgery differences in plasma, and liver markers of oxidative stress and inflammation (including chemokine C-C motif ligand 2, paraoxonase-1, galectin-3, and sonic hedgehog) were observed between patients with, and those without, NASH; post-surgery indicated consistent improvements in these parameters. CONCLUSION: Our study shows that the histology and liver function of patients with morbid obesity are significantly improved after LSG via mechanisms that involve the reduction of oxidative stress and inflammatory processes. These data encourage the use of LSG as a therapeutic option to improve, or resolve, NAFLD.


Gastrectomy/methods , Inflammation/therapy , Non-alcoholic Fatty Liver Disease/surgery , Oxidative Stress , Adult , Biomarkers , Biopsy , Female , Humans , Inflammation/surgery , Laparoscopy , Male , Middle Aged , Non-alcoholic Fatty Liver Disease/pathology , Non-alcoholic Fatty Liver Disease/therapy , Obesity, Morbid/surgery , Treatment Outcome
12.
Biochim Biophys Acta Mol Basis Dis ; 1865(6): 1555-1566, 2019 06 01.
Article En | MEDLINE | ID: mdl-30905786

The risk of non-alcoholic fatty liver disease increases with obesity. Vulnerability to oxidative stress and/or inflammation represents a crucial step in non-alcoholic fatty liver disease progression through abnormal metabolic responses. In this study, we investigated the role of CCL2 gene ablation in mice that were double deficient in low density lipoprotein receptor and in paraoxonase-1. Mass spectrometry methods were used to assess the liver metabolic response in mice fed either regular chow or a high-fat diet. Dietary fat caused liver steatosis, oxidative stress and the accumulation of pro-inflammatory macrophages in the livers of double deficient mice. We observed alterations in energy metabolism-related pathways and in metabolites associated with the methionine cycle and the glutathione reduction pathway. This metabolic response was associated with impaired autophagy. Conversely, when we established CCL2 deficiency, histologic features of fatty liver disease were abrogated, hepatic liver oxidative stress decreased, and anti-inflammatory macrophage marker expression levels increased. These changes were associated with the normalization of metabolic disturbances and increased lysosome-associated membrane protein 2, expression, which suggests enhanced chaperone-mediated autophagy. This study demonstrates that CCL2 is a key molecule for the development of metabolic and histological alterations in the liver of mice sensitive to the development of hyperlipidemia and hepatic steatosis, a finding with potential to identify new therapeutic targets in liver diseases.


Aryldialkylphosphatase/genetics , Chemokine CCL2/genetics , Hyperlipidemias/genetics , Lipoproteins, LDL/metabolism , Non-alcoholic Fatty Liver Disease/genetics , Receptors, LDL/genetics , Animals , Aryldialkylphosphatase/deficiency , Autophagy/genetics , Chemokine CCL2/deficiency , Diet, High-Fat/adverse effects , Energy Metabolism/genetics , Gene Expression Regulation , Glutathione/metabolism , Hyperlipidemias/etiology , Hyperlipidemias/metabolism , Hyperlipidemias/pathology , Liver/metabolism , Liver/pathology , Lysosomal-Associated Membrane Protein 2/genetics , Lysosomal-Associated Membrane Protein 2/metabolism , Macrophages/metabolism , Macrophages/pathology , Male , Metabolome/genetics , Methionine/metabolism , Mice , Mice, Inbred C57BL , Mice, Knockout , Non-alcoholic Fatty Liver Disease/etiology , Non-alcoholic Fatty Liver Disease/metabolism , Non-alcoholic Fatty Liver Disease/pathology , Oxidative Stress , Receptors, LDL/deficiency , Signal Transduction
13.
PLoS One ; 13(11): e0207474, 2018.
Article En | MEDLINE | ID: mdl-30444915

The aims of this study were to investigate changes in energy balance-associated metabolites associated with radiotherapy in patients with breast cancer, and to relate these changes to the clinical and pathological response-to-treatment. We studied 151 women with breast cancer who received radiotherapy following surgical excision of the tumor. Blood was obtained before and after the irradiation procedure. The control group was composed of 44 healthy women with a similar age distribution to that of the patients. We analyzed the concentrations of metabolites involved in glycolysis, citric acid cycle and amino acid metabolism using targeted quantitative metabolomics. Post-surgery, pre-radiotherapy, patients had major alterations in the serum concentrations of products of glycolysis, citric acid cycle and amino acid metabolism. The strongest alterations were decreases in serine, leucine and isoleucine concentrations. Alterations in metabolite levels were partially, or totally, reversed after irradiation; the concentrations of serine, leucine and isoleucine approached equivalence to those of the control group. Estrogen receptor-positive patients were those with lower concentrations, while triple negative patients had higher concentrations of these amino acids. The normalization of the amino acids serine, leucine and isoleucine concentrations could be clinically relevant because the normalization of these energy-balance metabolites would suggest that residual micro-metastatic disease had been effectively diminished by the radiotherapy, and may be an indicator of its efficacy.


Amino Acids/metabolism , Citric Acid Cycle , Glycolysis , Triple Negative Breast Neoplasms , Adult , Aged , Aged, 80 and over , Female , Humans , Middle Aged , Triple Negative Breast Neoplasms/metabolism , Triple Negative Breast Neoplasms/pathology , Triple Negative Breast Neoplasms/therapy
14.
Breast ; 42: 142-149, 2018 Dec.
Article En | MEDLINE | ID: mdl-30296647

Alterations in the circulating levels of trace elements have been observed in breast cancer (BC) patients. However, the relationships between these alterations and the metabolic and clinical consequences of BC are unknown. The treatment-of-choice of BC is surgery followed by radiation therapy (RT). The present study was aimed at investigating: 1) the concentrations of several trace elements in BC patients, and their relationships with the intrinsic molecular subtypes of tumors; 2) the toxicological effect of RT. We studied 49 women with BC who were scheduled to receive RT following excision of the tumor. Plasma samples were obtained before and after the irradiation procedure. The control group was composed of 49 healthy women. Patients had significantly lower pre-RT concentrations of B, Cu, and Zn, and significantly higher concentrations of Sr than the control group. Irradiation was associated with a striking increase in plasma B concentrations, while Cu, Fe, Sr and Zn concentrations were not significantly different from pre-RT levels, albeit Sr and Zn showed non-significant trends towards increases. The plasma concentrations of B, Cu, Fe, Sr, and Zn were associated with the tumor expression of hormone receptors, epidermal growth factor receptor 2, Ki67 antigen, as well as dermatitis and asthenia, all of which represent the main toxicological responses to RT.


Biomarkers, Tumor/blood , Breast Neoplasms/metabolism , Trace Elements/blood , Adult , Biomarkers, Tumor/metabolism , Breast Neoplasms/blood , Breast Neoplasms/drug therapy , Breast Neoplasms/pathology , Case-Control Studies , Female , Humans , Middle Aged , Neoadjuvant Therapy , Trace Elements/metabolism
15.
Mediators Inflamm ; 2018: 2760272, 2018.
Article En | MEDLINE | ID: mdl-30327580

Peripheral artery disease (PAD) is a common disease affecting 20-25% of population over 60 years old. Early diagnosis is difficult because symptoms only become evident in advanced stages of the disease. Inflammation, impaired metabolism, and mitochondrial dysfunction predispose to PAD, which is normally associated with other highly prevalent and related conditions, such as diabetes, dyslipidemia, and hypertension. We have measured energy-balance-associated metabolite concentrations in the plasma of PAD patients segregated by the severity of the disease and in plasma of healthy volunteers using a quantitative and targeted metabolomic approach. We found relevant associations between several metabolites (3-hydroxybutirate, aconitate, (iso)citrate, glutamate, and serine) with markers of oxidative stress and inflammation. Metabolomic profiling also revealed that (iso)citrate and glutamate are metabolites with high ability to discriminate between healthy participants and PAD patients without symptoms. Collectively, our data suggest that metabolomics provide significant information on the pathogenesis of PAD and useful biomarkers for the diagnosis and assessment of progression.


Peripheral Arterial Disease/blood , Aryldialkylphosphatase/metabolism , Biomarkers/blood , Chemokine CCL2/metabolism , Cross-Sectional Studies , Energy Metabolism/physiology , Humans , Metabolomics , Oxidative Stress , Peripheral Arterial Disease/diagnosis , Peripheral Arterial Disease/metabolism
16.
Clin Biochem ; 61: 1-6, 2018 Nov.
Article En | MEDLINE | ID: mdl-30165052

BACKGROUND: Acute renal failure in patients with sepsis is associated with high mortality. Studies have highlighted alterations in serum paraoxonase-1 in severe infections. However, the published literature has no insight into the clinical evolution of these parameters in patients with sepsis and acute renal failure treated with extra-renal depuration techniques. METHODS: We studied 25 patients with sepsis and acute renal failure who were treated with continuous renal-replacement therapy. Blood for laboratory analyses was collected at days 0, 1, 2, 5, 7, and 10. We measured serum paraoxonase-1 activity and concentration, lipid profile, aminotransferase activities, pH, and lactate, urea, creatinine and C-reactive protein concentrations. Values were compared with those of 50 healthy individuals. RESULTS: Patients with sepsis and acute renal failure had lower serum paraoxonase-1 activity, lower high-density lipoprotein cholesterol concentrations, and higher serum paraoxonase-1 concentrations than the control group. We found a significant inverse correlation between serum paraoxonase-1 concentrations and the Acute Physiology And Chronic Health Evaluation II score in survivors as well as non-survivors, and a significant inverse correlation between serum paraoxonase-1 concentrations and the Sequential Organ Failure Assessment score only in survivors. Extra-renal depuration techniques produced a further increase in this enzyme related to the duration of treatment, and to serum urea concentration. CONCLUSION: Our results show an inverse relationship between the concentration of paraoxonase-1 and the disease severity of patients with renal failure caused by septic shock. These results highlight relationships between paraoxonase-1 and infectious diseases and sepsis, with insights into potential clinical evolution of treatment.


Acute Kidney Injury/therapy , Aryldialkylphosphatase/blood , Kidney/physiopathology , Renal Replacement Therapy , Shock, Septic/physiopathology , Acute Kidney Injury/blood , Acute Kidney Injury/etiology , Acute Kidney Injury/physiopathology , Aged , Biomarkers/blood , Cholesterol/blood , Cholesterol, HDL/blood , Cohort Studies , Female , Hospitals, University , Humans , Intensive Care Units , Length of Stay , Longitudinal Studies , Male , Middle Aged , Prospective Studies , Renal Replacement Therapy/adverse effects , Severity of Illness Index , Spain , Urea/blood
17.
Aging Cell ; 17(4): e12772, 2018 08.
Article En | MEDLINE | ID: mdl-29740925

Metformin, the first drug chosen to be tested in a clinical trial aimed to target the biology of aging per se, has been clinically exploited for decades in the absence of a complete understanding of its therapeutic targets or chemical determinants. We here outline a systematic chemoinformatics approach to computationally predict biomolecular targets of metformin. Using several structure- and ligand-based software tools and reference databases containing 1,300,000 chemical compounds and more than 9,000 binding sites protein cavities, we identified 41 putative metformin targets including several epigenetic modifiers such as the member of the H3K27me3-specific demethylase subfamily, KDM6A/UTX. AlphaScreen and AlphaLISA assays confirmed the ability of metformin to inhibit the demethylation activity of purified KDM6A/UTX enzyme. Structural studies revealed that metformin might occupy the same set of residues involved in H3K27me3 binding and demethylation within the catalytic pocket of KDM6A/UTX. Millimolar metformin augmented global levels of H3K27me3 in cultured cells, including reversion of global loss of H3K27me3 occurring in premature aging syndromes, irrespective of mitochondrial complex I or AMPK. Pharmacological doses of metformin in drinking water or intraperitoneal injection significantly elevated the global levels of H3K27me3 in the hepatic tissue of low-density lipoprotein receptor-deficient mice and in the tumor tissues of highly aggressive breast cancer xenograft-bearing mice. Moreover, nondiabetic breast cancer patients receiving oral metformin in addition to standard therapy presented an elevated level of circulating H3K27me3. Our biocomputational approach coupled to experimental validation reveals that metformin might directly regulate the biological machinery of aging by targeting core chromatin modifiers of the epigenome.


Enzyme Inhibitors/pharmacology , Histone Demethylases/antagonists & inhibitors , Metformin/pharmacology , Nuclear Proteins/antagonists & inhibitors , Animals , Biocatalysis , Dose-Response Relationship, Drug , Enzyme Inhibitors/chemistry , Female , Histone Demethylases/metabolism , Humans , Ligands , Metformin/chemistry , Mice , Mice, Knockout , Models, Molecular , Molecular Structure , Neoplasms, Experimental/drug therapy , Neoplasms, Experimental/metabolism , Nuclear Proteins/metabolism
18.
J Trace Elem Med Biol ; 48: 8-15, 2018 Jul.
Article En | MEDLINE | ID: mdl-29773198

The metabolic alterations associated with obesity include mineral dysregulation. Essential trace elements are nutrients with a relevant function in a large number of cellular processes and multiple roles in the correct functioning of metabolic enzymes. Paraoxonase-1 (PON1) is an antioxidant and anti-inflammatory enzyme that is compromised in obesity. In the present study, the potential alterations in trace elements in morbidly obese women were assessed in relation to serum PON1 activity and concentration, as well as to other obesity-related comorbidities such as diabetes mellitus and fatty liver. We recruited 41 morbidly obese women and 51 control individuals. The serum concentrations of 30 elements, PON1 paraoxonase and lactonase activities, and PON1 concentration were measured. We observed significant alterations in the levels of As, Ba, Cu, Ca, Fe, Mg, Na, Se, Sr, and Zn in obese women; some of them (As, Ca, Cr, Cu, Mg, and Se) being significantly correlated with serum PON1 values. The most relevant changes were observed in the concentrations of As, Sr and Mg, the last of which was also significantly associated with diabetes mellitus. The current results raise the possibility that increased ingestion and/or storage of a number of trace elements may be factors predisposing to obesity-related comorbidities and metabolic alterations.


Aryldialkylphosphatase/blood , Obesity, Morbid/blood , Trace Elements/blood , Adult , Female , Humans , Middle Aged , Young Adult
19.
Arch Med Res ; 49(2): 119-122, 2018 02.
Article En | MEDLINE | ID: mdl-29699809

This study provides preliminary information on the usefulness of measuring serum paraoxonase-1 (PON1) concentration and activity (and other inflammatory markers) to predict tumor recurrence in patients with urinary bladder cancer. We studied a total of 39 hospitalized patients in whom the diagnosis of urinary bladder cancer was confirmed by transurethral resection. After five years of follow-up, 29 patients presented with tumor recurrence. As control subjects, we also studied 61 healthy subjects and a further 132 hospitalized patients who had a urinary catheter-related infection due to causes other than cancer. Results showed that urinary bladder patients had lower serum PON1 concentration and activity, and higher chemokine (C-C motif) ligand 2, C-reactive protein, and procalcitonin concentrations than the control individuals. Patients with tumor recurrence had significantly lower serum PON1 concentration than patients without tumor recurrence. The mean area under the curve of the receiver operating characteristics plot for serum PON1 concentration in discriminating patients with and those without tumor recurrence was 0.755 and the best combination of sensitivity and specificity was obtained at PON1 = 100 mg/L (0.72 and 0.80, respectively). Establishing this value as a cut-off, positive predictive value was = 0.91, and negative predictive value was = 0.50. These results suggest that the measurement of serum PON1 concentration may be a high-sensitivity marker of tumor recurrence in urinary bladder cancer patients.


Aryldialkylphosphatase/blood , Biomarkers, Tumor/blood , Neoplasm Recurrence, Local/blood , Neoplasm Recurrence, Local/diagnosis , Urinary Bladder Neoplasms/blood , Urinary Bladder Neoplasms/diagnosis , C-Reactive Protein/analysis , Chemokines, C/blood , Follow-Up Studies , Humans , Procalcitonin/blood , ROC Curve , Sensitivity and Specificity
20.
Biol Trace Elem Res ; 186(1): 74-84, 2018 Nov.
Article En | MEDLINE | ID: mdl-29525848

Oxidative stress and inflammation are candidate mechanisms to explain the potential role of exposure to metals and reduced activity of paraoxonase-1 (PON1) in age-related diseases. Both may be risk factors contributing to atherosclerosis. In the present study, inductively coupled mass spectrometry was used to explore multiple trace elements, while in-house methods were employed to measure PON1-related variables in patients with lower extremity artery disease (LEAD). Healthy controls were matched for sex, age, body weight, and relevant genotype variants. Serum concentrations of As, Ba, Cu, and Sr were higher in patients than those in controls, with a strong predictive ability to discriminate between groups. Differences in serum Pb, Cd, and Zn were negligible. Serum Cu increased when the disease was more severe, but a negative trend was noted for serum As, B, Ba, and Zn. The only variable associated with ankle-brachial index was serum Zn. Serum PON1 activity was significantly lower in LEAD patients. When the ability of serum trace elements to modulate PON1 activity was explored, the analysis revealed a unique association with serum Zn. The current results strongly suggest that Zn may have a protective effect in non-coronary atherosclerosis and indicate that this element may exert its anti-inflammatory and antioxidant functions through interactions with PON1 activity. These findings deserve confirmation and further research. In particular, the periodic evaluation of serum trace elements and the prescription of Zn supplements are easy measures to implement and that can improve the treatment of patients with LEAD.


Anti-Inflammatory Agents, Non-Steroidal/metabolism , Antioxidants/metabolism , Aryldialkylphosphatase/metabolism , Atherosclerosis/metabolism , Trace Elements/metabolism , Aged , Anti-Inflammatory Agents, Non-Steroidal/blood , Anti-Inflammatory Agents, Non-Steroidal/therapeutic use , Antioxidants/therapeutic use , Atherosclerosis/blood , Atherosclerosis/drug therapy , Female , Humans , Lower Extremity , Male , Mass Spectrometry , Middle Aged , Trace Elements/blood , Trace Elements/therapeutic use
...