Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 19 de 19
Filter
2.
Diabetes Metab ; 48(5): 101353, 2022 09.
Article in English | MEDLINE | ID: mdl-35487478

ABSTRACT

AIM: This study investigated whether rare, deleterious variants in monogenic diabetes-genes are associated with early-onset type 2 diabetes (T2D). METHODS: A nested case-control study was designed from 9712 Italian patients with T2D. Individuals with age at diabetes onset ≤35 yrs (n = 300; cases) or ≥65 yrs (n = 300; controls) were selected and screened for variants in 27 monogenic diabetes-genes by targeted resequencing. Rare (minor allele frequency-MAF <1%) and possibly deleterious variants were collectively tested for association with early-onset T2D. The association of a genetic risk score (GRS) based on 17 GWAS-SNPs for T2D was also tested. RESULTS: When all rare variants were considered together, each increased the risk of early-onset T2D by 65% (allelic OR =1.64, 95% CI: 1.08-2.48, p = 0.02). Effects were similar when the 600 study participants were stratified according to their place of recruitment (Central-Southern Italy, 182 cases vs. 142 controls, or Rome urban area, 118 vs. 158, p for heterogeneity=0.53). Progressively less frequent variants showed increasingly stronger effects in the risk of early-onset T2D for those with MAF <0.001% (OR=6.34, 95% CI: 1.87-22.43, p = 0.003). One unit of T2D-GRS significantly increased the risk of early-onset T2D (OR 1.09, 95% CI: 1.01-1.18; p = 0.02). This association was stronger among rare variants carriers as compared to non-carriers (p = 0.02). CONCLUSION: Rare variants in monogenic-diabetes genes are associated with an increased risk of early-onset T2D, and interact with common T2D susceptibility variants in shaping it. These findings might help develop prediction tools to identify individuals at high risk of developing T2D in early adulthood.


Subject(s)
Diabetes Mellitus, Type 2 , Adult , Case-Control Studies , Diabetes Mellitus, Type 2/epidemiology , Diabetes Mellitus, Type 2/genetics , Gene Frequency , Genetic Predisposition to Disease , Humans , Polymorphism, Single Nucleotide
3.
J Clin Endocrinol Metab ; 107(3): 668-684, 2022 02 17.
Article in English | MEDLINE | ID: mdl-34718610

ABSTRACT

CONTEXT: Genes causing familial forms of diabetes mellitus are only partially known. OBJECTIVE: We set out to identify the genetic cause of hyperglycemia in multigenerational families with an apparent autosomal dominant form of adult-onset diabetes not due to mutations in known monogenic diabetes genes. METHODS: Existing whole-exome sequencing (WES) data were used to identify exonic variants segregating with diabetes in 60 families from the United States and Italy. Functional studies were carried out in vitro (transduced MIN6-K8 cells) and in vivo (Caenorhabditis elegans) to assess the diabetogenic potential of 2 variants in the malate dehydrogenase 2 (MDH2) gene linked with hyperglycemia in 2 of the families. RESULTS: A very rare mutation (p.Arg52Cys) in MDH2 strongly segregated with hyperglycemia in 1 family from the United States. An infrequent MDH2 missense variant (p.Val160Met) also showed disease cosegregation in a family from Italy, although with reduced penetrance. In silico, both Arg52Cys and Val160Met were shown to affect MDH2 protein structure and function. In transfected HepG2 cells, both variants significantly increased MDH2 enzymatic activity, thereby decreasing the NAD+/NADH ratio-a change known to affect insulin signaling and secretion. Stable expression of human wild-type MDH2 in MIN6-K8 cell lines enhanced glucose- and GLP-1-stimulated insulin secretion. This effect was blunted by the Cys52 or Met160 substitutions. Nematodes carrying equivalent changes at the orthologous positions of the mdh-2 gene showed impaired glucose-stimulated insulin secretion. CONCLUSION: Our findings suggest a central role of MDH2 in human glucose homeostasis and indicate that gain of function variants in this gene may be involved in the etiology of familial forms of diabetes.


Subject(s)
Blood Glucose/metabolism , Hyperglycemia/genetics , Malate Dehydrogenase/genetics , Adult , Aged , Aged, 80 and over , Animals , Animals, Genetically Modified , Blood Glucose/analysis , Caenorhabditis elegans , Caenorhabditis elegans Proteins/genetics , Caenorhabditis elegans Proteins/metabolism , Case-Control Studies , Cell Line, Tumor , DNA Mutational Analysis , Female , Gain of Function Mutation , Humans , Hyperglycemia/blood , Insulin/analysis , Insulin/metabolism , Insulin Secretion/genetics , Islets of Langerhans , Malate Dehydrogenase/metabolism , Male , Mice , Middle Aged , Models, Animal , Primary Cell Culture , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Exome Sequencing
4.
Diabetes ; 69(4): 771-783, 2020 04.
Article in English | MEDLINE | ID: mdl-31974142

ABSTRACT

The cardiovascular benefits of fibrates have been shown to be heterogeneous and to depend on the presence of atherogenic dyslipidemia. We investigated whether genetic variability in the PPARA gene, coding for the pharmacological target of fibrates (PPAR-α), could be used to improve the selection of patients with type 2 diabetes who may derive cardiovascular benefit from addition of this treatment to statins. We identified a common variant at the PPARA locus (rs6008845, C/T) displaying a study-wide significant influence on the effect of fenofibrate on major cardiovascular events (MACE) among 3,065 self-reported white subjects treated with simvastatin and randomized to fenofibrate or placebo in the ACCORD-Lipid trial. T/T homozygotes (36% of participants) experienced a 51% MACE reduction in response to fenofibrate (hazard ratio 0.49; 95% CI 0.34-0.72), whereas no benefit was observed for other genotypes (P interaction = 3.7 × 10-4). The rs6008845-by-fenofibrate interaction on MACE was replicated in African Americans from ACCORD (N = 585, P = 0.02) and in external cohorts (ACCORD-BP, ORIGIN, and TRIUMPH, total N = 3059, P = 0.005). Remarkably, rs6008845 T/T homozygotes experienced a cardiovascular benefit from fibrate even in the absence of atherogenic dyslipidemia. Among these individuals, but not among carriers of other genotypes, fenofibrate treatment was associated with lower circulating levels of CCL11-a proinflammatory and atherogenic chemokine also known as eotaxin (P for rs6008845-by-fenofibrate interaction = 0.003). The GTEx data set revealed regulatory functions of rs6008845 on PPARA expression in many tissues. In summary, we have found a common PPARA regulatory variant that influences the cardiovascular effects of fenofibrate and that could be used to identify patients with type 2 diabetes who would derive benefit from fenofibrate treatment, in addition to those with atherogenic dyslipidemia.


Subject(s)
Diabetes Mellitus, Type 2/genetics , Dyslipidemias/drug therapy , Fenofibrate/therapeutic use , Hypolipidemic Agents/therapeutic use , PPAR alpha/genetics , Polymorphism, Single Nucleotide , Chemokines/blood , Diabetes Mellitus, Type 2/blood , Diabetes Mellitus, Type 2/complications , Diabetes Mellitus, Type 2/drug therapy , Dyslipidemias/blood , Dyslipidemias/complications , Dyslipidemias/genetics , Female , Genotype , Humans , Lipids/blood , Male , Middle Aged , Pharmacogenetics , Treatment Outcome
5.
Acta Diabetol ; 56(5): 515-523, 2019 May.
Article in English | MEDLINE | ID: mdl-30656436

ABSTRACT

AIMS: Maturity Onset Diabetes of the Young (MODY) is a monogenic form of diabetes with autosomal dominant inheritance pattern. The diagnosis of MODY and its subtypes is based on genetic testing. Our aim was investigating MODY by means of next-generation sequencing in the Tunisian population. METHODS: We performed a targeted sequencing of 27 genes known to cause monogenic diabetes in 11 phenotypically suspected Tunisian patients. We retained genetic variants passing filters of frequency in public databases as well as their probable effects on protein structures and functions evaluated by bioinformatics prediction tools. RESULTS: Five heterozygous variants were found in four patients. They include two mutations in HNF1A and GCK that are the causative genes of the two most prevalent MODY subtypes described in the literature. Other possible mutations, including novel frameshift and splice-site variants were identified in ABCC8 gene. CONCLUSIONS: Our study is the first to investigate the clinical application of targeted next-generation sequencing for the diagnosis of MODY in Africa. The combination of this approach with a filtering/prioritization strategy made a step towards the identification of MODY mutations in the Tunisian population.


Subject(s)
Diabetes Mellitus, Type 2/genetics , High-Throughput Nucleotide Sequencing , Adult , Diabetes Mellitus, Type 2/diagnosis , Female , Frameshift Mutation , Genetic Testing , Hepatocyte Nuclear Factor 1-alpha/genetics , Heterozygote , Humans , Male , Mutation , Pedigree , Phenotype , Tunisia
6.
Diabetes ; 67(1): 137-145, 2018 01.
Article in English | MEDLINE | ID: mdl-28993341

ABSTRACT

Multigenerational diabetes of adulthood is a mostly overlooked entity, simplistically lumped into the large pool of type 2 diabetes. The general aim of our research in the past few years is to unravel the genetic causes of this form of diabetes. Identifying among families with multigenerational diabetes those who carry mutations in known monogenic diabetes genes is the first step to then allow us to concentrate on remaining pedigrees in which to unravel new diabetes genes. Targeted next-generation sequencing of 27 monogenic diabetes genes was carried out in 55 family probands and identified mutations verified among their relatives by Sanger sequencing. Nine variants (in eight probands) survived our filtering/prioritization strategy. After likelihood of causality assessment by established guidelines, six variants were classified as "pathogenetic/likely pathogenetic" and two as "of uncertain significance." Combining present results with our previous data on the six genes causing the most common forms of maturity-onset diabetes of the young allows us to infer that 23.6% of families with multigenerational diabetes of adulthood carry mutations in known monogenic diabetes genes. Our findings indicate that the genetic background of hyperglycemia is unrecognized in the vast majority of families with multigenerational diabetes of adulthood. These families now become the object of further research aimed at unraveling new diabetes genes.


Subject(s)
Diabetes Mellitus, Type 2/genetics , High-Throughput Nucleotide Sequencing/methods , Adult , Aged , Basic Helix-Loop-Helix Transcription Factors/genetics , Female , Germinal Center Kinases , Hepatocyte Nuclear Factor 1-alpha/genetics , Hepatocyte Nuclear Factor 1-beta/genetics , Hepatocyte Nuclear Factor 4/genetics , Homeodomain Proteins/genetics , Humans , Hyperglycemia/genetics , Male , Middle Aged , Mutation/genetics , Pedigree , Protein Serine-Threonine Kinases/genetics , Trans-Activators/genetics
7.
Nephrol Dial Transplant ; 32(10): 1718-1722, 2017 Oct 01.
Article in English | MEDLINE | ID: mdl-27448670

ABSTRACT

BACKGROUND: UMOD variability has been associated at a genome-wide level of statistical significance with glomerular filtration rate (GFR) in Swedish individuals with type 2 diabetes (T2D; n = 4888). Whether this finding is extensible also to diabetic patients from other populations deserves further study. Our aim was to investigate the relationship between UMOD variability and GFR in patients with T2D from Italy. METHODS: Genotyping of the single nucleotide polymorphism (SNP) rs12917707 at the UMOD locus has been carried out in 3087 individuals from four independent Italian cohorts of patients with T2D by TaqMan allele discrimination. RESULTS: In none of the four study cohorts was rs12917707 significantly associated with GFR (P > 0.05 for all). Similar results were obtained when the four samples were pooled and analyzed together (ß = 0.83, P = 0.19). Such effect was strikingly smaller than that previously reported in Swedish patients (P for heterogeneity = 1.21 × 10-7). CONCLUSIONS: The previously reported strong association between rs12917707 and GFR in diabetic patients from Sweden is not observed in Italian diabetic patients, thus clearly pointing to a heterogeneous effect across the two different samples. This suggests that UMOD is a strong genetic determinant of kidney function in patients with T2D in some, but not all, populations.


Subject(s)
Diabetes Mellitus, Type 2/ethnology , Glomerular Filtration Rate , Polymorphism, Single Nucleotide , Uromodulin/genetics , White People/genetics , Alleles , Cohort Studies , Diabetes Mellitus, Type 2/genetics , Diabetes Mellitus, Type 2/physiopathology , Female , Genotype , Humans , Italy/epidemiology , Male , Middle Aged , Sweden/epidemiology
8.
Endocrine ; 54(1): 38-46, 2016 Oct.
Article in English | MEDLINE | ID: mdl-26956846

ABSTRACT

The high mortality risk of patients with type 2 diabetes mellitus may well be explained by the several comorbidities and/or complications. Also the intrinsic genetic component predisposing to diabetes might have a role in shaping the risk of diabetes-related mortality. Among type 2 diabetes mellitus SNPs, rs1801282 is of particular interest because (i) it is harbored by peroxisome proliferator-activated receptor-γ2 (PPARγ2), which is the target for thiazolidinediones which are used as antidiabetic drugs, decreasing all-cause mortality in type 2 diabetes mellitus, and (ii) it is associated with insulin resistance and related traits, risk factors for overall mortality in type 2 diabetes mellitus. We investigated the role of PPARγ2 P12A, according to a dominant model (PA + AA vs. PP individuals) on incident all-cause mortality in three cohorts of type 2 diabetes mellitus, comprising a total of 1672 patients (462 deaths) and then performed a meta-analysis of ours and all available published data. In the three cohorts pooled and analyzed together, no association between PPARγ2 P12A and all-cause mortality was observed (HR 1.02, 95 % CI 0.79-1.33). Similar results were observed after adjusting for age, sex, smoking habits, and BMI (HR 1.09, 95 % CI 0.83-1.43). In a meta-analysis of ours and all studies previously published (n = 3241 individuals; 666 events), no association was observed between PPARγ2 P12A and all-cause mortality (HR 1.07, 95 % CI 0.85-1.33). Results from our individual samples as well as from our meta-analysis suggest that the PPARγ2 P12A does not significantly affect all-cause mortality in patients with type 2 diabetes mellitus.


Subject(s)
Diabetes Mellitus, Type 2/genetics , Insulin Resistance/genetics , PPAR gamma/genetics , Polymorphism, Single Nucleotide , Diabetes Mellitus, Type 2/mortality , Genetic Association Studies , Genetic Predisposition to Disease , Genotype , Humans , Phenotype
9.
Atherosclerosis ; 242(1): 334-9, 2015 Sep.
Article in English | MEDLINE | ID: mdl-26253791

ABSTRACT

OBJECTIVE: Genes that modulate insulin sensitivity may also be involved in shaping the risk of coronary artery disease (CAD). The relatively common TRIB3 Q84R polymorphism (rs2295490) has been associated with abnormal insulin signaling, endothelial dysfunction, insulin resistance, and pro-atherogenic phenotypes. The aim of our study was to investigate the association between low-frequency TRIB3 coding variants and CAD in patients with type 2 diabetes (T2D). METHODS: Three case-control studies for CAD from Italy and US were analyzed, for a total of 1565 individuals, all with type 2 diabetes. Infrequent variants were identified by re-sequencing TRIB3 exons in 140 "extreme cases" and 140 "super-controls" and then genotyped in all study subjects. RESULTS: TRIB3 infrequent variants (n = 8), considered according to a collapsing rare variants framework, were significantly associated with CAD in diabetic patients from Italy (n = 700, OR = 0.43, 95% CI 0.20-0.91; p = 0.027), but not from the US (n = 865, OR = 1.22, 95% CI 0.69-2.18; p = 0.49). In the Italian sets, the association was especially strong among individuals who also carried the common R84 variant. CONCLUSION: Although preliminary, our finding suggests a role of TRIB3 low-frequency variants on CAD among Italian patients with T2D. Further studies are needed to address the role of TRIB3 infrequent variants in other populations of both European and non-European ancestries.


Subject(s)
Cell Cycle Proteins/genetics , Coronary Artery Disease/genetics , Diabetes Mellitus, Type 2/genetics , Diabetic Angiopathies/genetics , Polymorphism, Single Nucleotide , Protein Serine-Threonine Kinases/antagonists & inhibitors , Repressor Proteins/genetics , Aged , Case-Control Studies , Coronary Artery Disease/diagnosis , Coronary Artery Disease/epidemiology , Diabetes Mellitus, Type 2/diagnosis , Diabetes Mellitus, Type 2/epidemiology , Diabetic Angiopathies/diagnosis , Diabetic Angiopathies/epidemiology , Exons , Female , Gene Frequency , Genetic Association Studies , Genetic Predisposition to Disease , Humans , Italy , Logistic Models , Male , Middle Aged , Odds Ratio , Phenotype , Protein Serine-Threonine Kinases/genetics , Risk Factors , United States/epidemiology
10.
Am J Hum Genet ; 97(1): 177-85, 2015 Jul 02.
Article in English | MEDLINE | ID: mdl-26073777

ABSTRACT

Diabetes mellitus is a highly heterogeneous disorder encompassing several distinct forms with different clinical manifestations including a wide spectrum of age at onset. Despite many advances, the causal genetic defect remains unknown for many subtypes of the disease, including some of those forms with an apparent Mendelian mode of inheritance. Here we report two loss-of-function mutations (c.1655T>A [p.Leu552(∗)] and c.280G>A [p.Asp94Asn]) in the gene for the Adaptor Protein, Phosphotyrosine Interaction, PH domain, and leucine zipper containing 1 (APPL1) that were identified by means of whole-exome sequencing in two large families with a high prevalence of diabetes not due to mutations in known genes involved in maturity onset diabetes of the young (MODY). APPL1 binds to AKT2, a key molecule in the insulin signaling pathway, thereby enhancing insulin-induced AKT2 activation and downstream signaling leading to insulin action and secretion. Both mutations cause APPL1 loss of function. The p.Leu552(∗) alteration totally abolishes APPL1 protein expression in HepG2 transfected cells and the p.Asp94Asn alteration causes significant reduction in the enhancement of the insulin-stimulated AKT2 and GSK3ß phosphorylation that is observed after wild-type APPL1 transfection. These findings-linking APPL1 mutations to familial forms of diabetes-reaffirm the critical role of APPL1 in glucose homeostasis.


Subject(s)
Adaptor Proteins, Signal Transducing/genetics , Diabetes Mellitus/genetics , Models, Molecular , Mutation, Missense/genetics , Adaptor Proteins, Signal Transducing/chemistry , Adult , Aged , Female , Hep G2 Cells , Humans , Immunoblotting , Insulin/metabolism , Italy , Male , Middle Aged , Pedigree , Proto-Oncogene Proteins c-akt/metabolism , United States
11.
Diabetes ; 64(7): 2658-63, 2015 Jul.
Article in English | MEDLINE | ID: mdl-25677913

ABSTRACT

Single nucleotide polymorphism (SNP) rs10911021 at the glutamate-ammonia ligase (GLUL) locus has been associated with an increased risk of coronary heart disease in individuals with type 2 diabetes. The effect of this SNP on mortality was investigated among 1,242 white subjects with type 2 diabetes from the Joslin Kidney Study (JKS) (n = 416) and the Gargano Mortality Study (GMS) (n = 826). During a mean follow-up of 12.8 ± 5.8 and 7.5 ± 2.2 years, respectively, a total of 215 and 164 deaths were observed in the two studies. In both cohorts, the all-cause mortality rate significantly increased with the number of rs10911021 risk alleles, with allelic hazard ratios (HRs) of 1.32 (95% CI 1.07-1.64, P = 0.01), 1.30 (1.10-1.69, P = 0.04), and 1.32 (1.12-1.55, P = 0.0011), respectively, in the JKS, the GMS, and the two studies combined. These associations were not affected by adjustment for possible confounders. In the JKS, for which data on causes of death were available, the HR for cardiovascular mortality was 1.51 (1.12-2.04, P = 0.0077) as opposed to 1.15 (0.84-1.55, P = 0.39) for mortality from noncardiovascular causes. These findings point to SNP rs10911021 as an independent modulator of mortality in patients with type 2 diabetes and, together with the previous observation, suggest that this results from an effect of this variant on cardiovascular risk.


Subject(s)
Diabetes Mellitus, Type 2/genetics , Glutamate-Ammonia Ligase/genetics , Polymorphism, Single Nucleotide , Adult , Aged , Cause of Death , Diabetes Mellitus, Type 2/mortality , Female , Humans , Male , Middle Aged
12.
Diabetes ; 63(9): 3135-40, 2014 Sep.
Article in English | MEDLINE | ID: mdl-24947357

ABSTRACT

This study tried to replicate in a large sample of white patients with type 2 diabetes (T2D) from Italy a previously reported association of the IRS1 G972R polymorphism with failure to oral antidiabetes drugs (OAD). A total of 2,409 patients from four independent studies were investigated. Case subjects (n = 1,193) were patients in whom, because of uncontrolled diabetes (i.e., HbA1c >8%), insulin therapy had been added either on, or instead of, maximal or near-maximal doses of OAD, mostly metformin and sulfonylureas; control subjects (n = 1,216) were patients with HbA1c <8% in the absence of insulin therapy. The IRS1 G972R polymorphism was typed by TaqMan allele discrimination. In all samples, individuals carrying the IRS1 R972 risk variant tended to be more frequent among case than control subjects, though reaching statistical significance only in one case. As no IRS1 G972R-by-study sample interaction was observed, data from the four samples were analyzed together; a significant association was observed (allelic odds ratio [OR] 1.30, 95% CI 1.03-1.63). When our present data were meta-analyzed with those obtained in a previous study, an overall R972 allelic OR of 1.37 (1.12-1.69) was observed. This study confirms in a large and ethnically homogeneous sample that IRS1 G972R polymorphism is associated with failure to OAD among patients with T2D.


Subject(s)
Diabetes Mellitus, Type 2/drug therapy , Hypoglycemic Agents/therapeutic use , Insulin Receptor Substrate Proteins/genetics , Polymorphism, Single Nucleotide , Aged , Diabetes Mellitus, Type 2/genetics , Humans , Italy , Male , Metformin/therapeutic use , Middle Aged , Mutation, Missense , Odds Ratio , Sulfonylurea Compounds/therapeutic use , White People/genetics
13.
Oncotarget ; 5(4): 978-92, 2014 Feb 28.
Article in English | MEDLINE | ID: mdl-24657851

ABSTRACT

The key role of the Wnt/ß-catenin signaling in colorectal cancer (CRC) insurgence and progression is now recognized and several therapeutic strategies targeting this pathway are currently in developing. Wnt/ß-catenin signaling not only dominates the early stages of sporadic colorectal cancer (SCC), but could also represent the connection between inflammatory bowel diseases (IBD) and increased risk of developing SCC. The knowledge on the sequential molecular events of Wnt-signaling cascade in IBD and during colorectal carcinogenesis, might provide new diagnostic/prognostic markers and could be helpful for optimizing the treatment protocols, thus improving the efficacy of Wnt-targeting therapies. We performed a comparative evaluation of the expression of some crucial molecules participating to Wnt signaling in an animal model of chemically-induced CRC and in human tissues obtained from patients suffering from IBD or at sequential stages of SCC. Specifically, we analyzed upstream events of Wnt signaling including ß-catenin nuclear translocation and loss of E-cadherin and APC functions, and downstream events including c-Myc and Cyclin-D1 expression. We demonstrated that these crucial components of the Wnt/ß-catenin pathway, when evaluated by immunohistochemistry using a multiparametric approach that includes the analyses of both expression and localization, could be potent markers for diagnosis, prevention and therapy in IBD and SCC, also possessing a predictive value for responsiveness to Wnt-targeting therapies. Furthermore, we showed that the animal model of chemically-induced CRC mimics the molecular events of Wnt signaling during IBD and SCC development in humans and may therefore be suitable for testing chemopreventive or therapeutic drugs targeting this pathway.


Subject(s)
Colorectal Neoplasms/metabolism , Inflammatory Bowel Diseases/metabolism , Wnt Signaling Pathway , beta Catenin/metabolism , Adenomatous Polyposis Coli Protein/metabolism , Animals , Cadherins/metabolism , Carcinogenesis/metabolism , Carcinogenesis/pathology , Colorectal Neoplasms/diagnosis , Colorectal Neoplasms/pathology , Colorectal Neoplasms/therapy , Cyclin D1/metabolism , Disease Models, Animal , Disease Progression , Humans , Inflammatory Bowel Diseases/diagnosis , Inflammatory Bowel Diseases/pathology , Inflammatory Bowel Diseases/therapy , Intestinal Mucosa/metabolism , Intestinal Mucosa/pathology , Male , Paraffin Embedding , Proto-Oncogene Proteins c-myc/metabolism , Rats , Signal Transduction
14.
Atherosclerosis ; 226(1): 140-5, 2013 Jan.
Article in English | MEDLINE | ID: mdl-23107043

ABSTRACT

OBJECTIVE: Insulin resistance (IR) and cardiovascular disease (CVD) share a common soil. We investigated the combined role of single nucleotide polymorphisms (SNPs) affecting insulin signaling (ENPP1 K121Q, rs1044498; IRS1 G972R, rs1801278; TRIB3 Q84R, rs2295490) on CVD, age at myocardial infarction (MI), in vivo insulin sensitivity and in vitro insulin-stimulated nitric oxide synthase (NOS) activity. DESIGN AND SETTING: 1. We first studied, incident cardiovascular events (a composite endpoint comprising myocardial infarction-MI, stroke and cardiovascular death) in 733 patients (2186 person-years, 175 events). 2. In a replication attempt, age at MI was tested in 331 individuals. 3. OGTT-derived insulin sensitivity index (ISI) was assessed in 829 individuals with fasting glucose <126 mg/dl. 4. NOS activity was measured in 40 strains of human vein endothelial cells (HUVECs). RESULTS: 1. Risk variants jointly predicted cardiovascular events (HR = 1.181; p = 0.0009) and, when added to clinical risk factors, significantly improved survival C-statistics; they also allowed a significantly correct reclassification (by net reclassification index) in the whole sample (135/733 individuals) and, even more, in obese patients (116/204 individuals). 2. Risk variants were jointly associated with age at MI (p = 0.006). 3. A significant association was also observed with ISI (p = 0.02). 4. Finally, risk variants were jointly associated with insulin-stimulated NOS activity in HUVECs (p = 0.009). CONCLUSIONS: Insulin signaling genes variants jointly affect cardiovascular disease, very likely by promoting whole body and endothelium-specific insulin resistance. Further studies are needed to address whether their genotyping help identify very high-risk patients who need specific and/or more aggressive preventive strategies.


Subject(s)
Cardiovascular Diseases/genetics , Endothelium, Vascular/metabolism , Insulin Resistance/genetics , Insulin/genetics , Signal Transduction/genetics , Cells, Cultured , Cross-Sectional Studies , Endothelial Cells/metabolism , Female , Humans , Male , Prospective Studies
15.
Nephrol Dial Transplant ; 27(12): 4411-3, 2012 Dec.
Article in English | MEDLINE | ID: mdl-22622453

ABSTRACT

BACKGROUND: We investigated whether the coronary artery disease (CAD) locus on chromosome 9p21 (as represented by single nucleotide polymorphism rs2383206) is associated with low estimated glomerular filtration rate (eGFR) or increased urinary albumin excretion in patients with Type 2 diabetes mellitus (T2DM). METHODS: Four samples, including a total of 3167 patients, were studied. The presence of low eGFR (<60 mL/min/1.73m(2)) was estimated from serum creatinine by means of the Modification of Diet in Renal Disease Study equation. Increased urinary albumin excretion was defined as an albumin-creatinine ratio (ACR) ≥2.5 mg/mmol in men and ≥3.5 mg/mmol in women. RESULTS: No association was found between rs2383206 and low eGFR or increased ACR in each sample as well as in a pooled analysis (overall odds ratio = 1.07, 95% confidence interval 0.94-1.22, P = 0.31 and overall odds ratio = 1.00, 95% confidence interval 0.90-1.12, P = 0.95, respectively). No interaction was observed between rs2383206 and poor glycemic control [HbA1c was above the median in the pooled sample (7.7%) in modulating eGFR or ACR (P for interaction = 0.42 and 0.90, respectively)]. CONCLUSION: Variability at the 9p21 CAD locus is unlikely to play a role in modulating susceptibility to kidney dysfunction in patients with T2DM.


Subject(s)
Albuminuria/genetics , Chromosomes, Human, Pair 9/genetics , Coronary Artery Disease/genetics , Coronary Artery Disease/physiopathology , Diabetes Mellitus, Type 2/physiopathology , Diabetic Angiopathies/genetics , Diabetic Angiopathies/physiopathology , Glomerular Filtration Rate/genetics , Kidney/physiopathology , Female , Humans , Male , Polymorphism, Single Nucleotide
16.
Biochim Biophys Acta ; 1822(6): 1004-18, 2012 Jun.
Article in English | MEDLINE | ID: mdl-22387884

ABSTRACT

Acidic tumor microenvironment and Wnt/ß-catenin pathway activation have been recognized as two crucial events associated with the initiation and progression of cancer. The aim of this study was to clarify the molecular mechanisms underlying the anti-proliferative effects of atrial natriuretic peptide (ANP) as well as to investigate the relationship between the cellular pH and the Wnt/ß-catenin signaling in cancer cells.To pursue our aims, we conducted investigations in DHD/K12/Trb rat colon adenocarcinoma cells. Intracellular pH was measured by Confocal Laser Scanning Microscopy (CLSM) using the lysosensor Green DND-189 probe. Expression of crucial molecules in the Wnt/ß-catenin signaling pathway was analyzed by CLSM, western blot, and real time PCR. Measurements of activation (phosphorylation state) of Akt, ERK1/2, and p38MAPKinase were performed by Reverse-Phase Protein Microarray Analysis (RPMA).We showed that ANP triggered a NHE-1-mediated increase of the intracellular acidity, inhibiting the Wnt/ß-catenin signaling simultaneously. Moreover, we observed that the Wnt1a, a Wnt signaling activator, affected the intracellular pH in an opposite fashion. Results from the comparative analysis of ANP and EIPA (a NHE-1 specific inhibitor) showed that these two molecules affect both the intracellular acidification and the Wnt/ß-catenin signaling cascade. Specifically, ANP acts on the upstream of the cascade, through a Frizzled-mediated activation, while EIPA does on the downstream.We show for the first time that the Akt activity might be a relevant molecular event linking the NHE-1-regulated intracellular pH and the Wnt/ß-catenin signaling. This provides evidence for a cross-talk between the intracellular alkalinization and the Wnt signaling in tumor cells.


Subject(s)
Atrial Natriuretic Factor/pharmacology , Colorectal Neoplasms/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Sodium-Hydrogen Exchangers/metabolism , Wnt Signaling Pathway , Adenocarcinoma/metabolism , Adenocarcinoma/pathology , Amiloride/analogs & derivatives , Amiloride/pharmacology , Animals , Breast Neoplasms/metabolism , Breast Neoplasms/pathology , Breast Neoplasms/physiopathology , Cell Line, Tumor , Cell Proliferation , Colorectal Neoplasms/pathology , Colorectal Neoplasms/physiopathology , Female , Gene Expression Regulation, Neoplastic , Humans , Hydrogen-Ion Concentration , MAP Kinase Signaling System , Phosphorylation , Rats , Sodium-Hydrogen Exchanger 1 , Sodium-Hydrogen Exchangers/antagonists & inhibitors , beta Catenin/metabolism , p38 Mitogen-Activated Protein Kinases/metabolism
17.
Curr Cancer Drug Targets ; 11(5): 572-85, 2011 Jun.
Article in English | MEDLINE | ID: mdl-21486216

ABSTRACT

An innovative approach for cancer therapy implies the use of drugs covalently conjugated to macromolecular carriers that specifically target molecules over-expressed on tumor cells. This drug delivery strategy may allow a controlled release of the drug and a high targeting selectivity on tumor cells, increasing drug cytotoxicity and decreasing its undesirable side effects. We provide in vitro and in vivo preclinical data on the antitumor efficacy of ONCOFID™-S, a new bioconjugate of hyaluronic acid (HA) with SN-38 (the CPT11 active metabolite), that support the validity of the drug delivery strategy implying the use of HA as macromolecular carrier of antineoplastic drugs, an approach based on the over-expression of its target CD44 (the receptor for HA-mediated motility) in a wide variety of cancers. We show that ONCOFID™-S exerts a strong in vitro anti-proliferative activity on CD44 over-expressing rat DHD/K12/trb colon adenocarcinoma cells, as well as on gastric, breast, oesophageal, ovarian and lung human cancer cells, higher than that exerted by unconjugated SN-38. We also demonstrated the in vivo anti-tumor efficacy of locoregional treatment with ONCOFID™-S on two pre-clinical models of colorectal cancer (CRC) in BDIX rats: a) syngeneic model of subcutaneous tumor; b) syngeneic model of metastatic tumor induced by injection of cells into the peritoneal cavity, mimicking the clinical situation of peritoneal carcinomatosis. Specifically, in the latter model ONCOFID™-S is able to dramatically reduce all parameters indicative of a poor prognosis in peritoneal metastatization of CRC without any myelotoxicity or mesothelial inflammation. We propose this CD44-targeted therapeutic strategy for locoregional treatment of peritoneal carcinomatosis from CRC, against which systemic chemotherapy results almost inefficient.


Subject(s)
Camptothecin/analogs & derivatives , Carcinoma/drug therapy , Drug Carriers/therapeutic use , Drug Delivery Systems/methods , Hyaluronan Receptors/metabolism , Hyaluronic Acid/analogs & derivatives , Peritoneal Neoplasms/drug therapy , Abdominal Wall/pathology , Animals , Antineoplastic Agents, Phytogenic/administration & dosage , Antineoplastic Agents, Phytogenic/adverse effects , Antineoplastic Agents, Phytogenic/chemistry , Antineoplastic Agents, Phytogenic/therapeutic use , Camptothecin/administration & dosage , Camptothecin/adverse effects , Camptothecin/chemistry , Camptothecin/therapeutic use , Carcinoma/metabolism , Carcinoma/pathology , Carcinoma/secondary , Cell Line, Tumor , Cell Proliferation/drug effects , Colorectal Neoplasms/drug therapy , Colorectal Neoplasms/metabolism , Colorectal Neoplasms/pathology , Drug Carriers/adverse effects , Drug Delivery Systems/adverse effects , Drug Evaluation, Preclinical , Humans , Hyaluronic Acid/adverse effects , Hyaluronic Acid/therapeutic use , Inhibitory Concentration 50 , Injections, Intraperitoneal , Irinotecan , Male , Neoplasm Transplantation , Peritoneal Neoplasms/metabolism , Peritoneal Neoplasms/pathology , Peritoneal Neoplasms/secondary , Random Allocation , Rats , Rats, Inbred Strains
18.
Atherosclerosis ; 216(1): 157-60, 2011 May.
Article in English | MEDLINE | ID: mdl-21353221

ABSTRACT

OBJECTIVE: SNP rs2943641 near the insulin receptor substrate 1 (IRS1) gene has been found to be associated with type 2 diabetes (T2D) and insulin-resistance in genome-wide association studies. We investigated whether this SNP is associated with cardiovascular risk factors and coronary artery disease (CAD) among diabetic individuals. METHODS: SNP rs2943641 was typed in 2133 White T2D subjects and tested for association with BMI, serum HDL cholesterol and triglycerides, hypertension history, and CAD risk. RESULTS: HDL cholesterol decreased by 1mg/dl (p = 0.004) and serum triglycerides increased by 6 mg/dl (p = 0.016) for each copy of the insulin-resistance allele. Despite these effects, no association was found with increased CAD risk (OR = 1.00, 95% CI 0.88-1.13). CONCLUSIONS: The insulin-resistance and T2D locus near the IRS1 gene is a determinant of lower HDL cholesterol among T2D subjects. However, this effect is small and does not translate into a detectable increase in CAD risk in this population.


Subject(s)
Cardiovascular Diseases/genetics , Cholesterol, HDL/blood , Coronary Artery Disease/genetics , Diabetes Mellitus, Type 2/genetics , Insulin Receptor Substrate Proteins/genetics , Insulin Resistance/genetics , Polymorphism, Single Nucleotide , Triglycerides/blood , Biomarkers/blood , Body Mass Index , Boston , Cardiovascular Diseases/blood , Cardiovascular Diseases/physiopathology , Case-Control Studies , Coronary Artery Disease/blood , Coronary Artery Disease/physiopathology , Diabetes Mellitus, Type 2/blood , Diabetes Mellitus, Type 2/complications , Diabetes Mellitus, Type 2/physiopathology , Female , Genetic Predisposition to Disease , Humans , Hypertension/genetics , Italy , Logistic Models , Male , Odds Ratio , Phenotype , Risk Assessment , Risk Factors
19.
BMC Immunol ; 9: 17, 2008 Apr 18.
Article in English | MEDLINE | ID: mdl-18423004

ABSTRACT

BACKGROUND: Besides few data concerning the antiseptic properties against a range of microbial agents and the anti-inflammatory potential both in vitro and in vivo, little is known about the influence of Eucalyptus oil (EO) extract on the monocytic/macrophagic system, one of the primary cellular effectors of the immune response against pathogen attacks. The activities of this natural extract have mainly been recognized through clinical experience, but there have been relatively little scientific studies on its biological actions. Here we investigated whether EO extract is able to affect the phagocytic ability of human monocyte derived macrophages (MDMs) in vitro and of rat peripheral blood monocytes/granulocytes in vivo in absence or in presence of immuno-suppression induced by the chemotherapeutic agent 5-fluorouracil (5-FU). METHODS: Morphological activation of human MDMs was analysed by scanning electron microscopy. Phagocytic activity was tested: i) in vitro in EO treated and untreated MDMs, by confocal microscopy after fluorescent beads administration; ii) in vivo in monocytes/granulocytes from peripheral blood of immuno-competent or 5-FU immuno-suppressed rats, after EO oral administration, by flow cytometry using fluorescein-labelled E. coli. Cytokine release by MDMs was determined using the BD Cytometric Bead Array human Th1/Th2 cytokine kit. RESULTS: EO is able to induce activation of MDMs, dramatically stimulating their phagocytic response. EO-stimulated internalization is coupled to low release of pro-inflammatory cytokines and requires integrity of the microtubule network, suggesting that EO may act by means of complement receptor-mediated phagocytosis. Implementation of innate cell-mediated immune response was also observed in vivo after EO administration, mainly involving the peripheral blood monocytes/granulocytes. The 5-FU/EO combined treatment inhibited the 5-FU induced myelotoxicity and raised the phagocytic activity of the granulocytic/monocytic system, significantly decreased by the chemotherapic. CONCLUSION: Our data, demonstrating that Eucalyptus oil extract is able to implement the innate cell-mediated immune response, provide scientific support for an additional use of this plant extract, besides those concerning its antiseptic and anti-inflammatory properties and stimulate further investigations also using single components of this essential oil. This might drive development of a possible new family of immuno-regulatory agents, useful as adjuvant in immuno-suppressive pathologies, in infectious disease and after tumour chemotherapy.


Subject(s)
Immunity, Innate/drug effects , Lymphocyte Activation/drug effects , Oils, Volatile/pharmacology , Phagocytosis/drug effects , Phagocytosis/immunology , Animals , Cell Culture Techniques , Cytokines/immunology , Cytokines/metabolism , Eucalyptus/immunology , Eucalyptus Oil , Fluorouracil , Humans , Leukocytes, Mononuclear/drug effects , Leukocytes, Mononuclear/immunology , Macrophages/drug effects , Macrophages/immunology , Microscopy, Confocal , Monoterpenes/immunology , Monoterpenes/pharmacology , Myeloablative Agonists , Nocodazole/pharmacology , Rats , Tubulin Modulators/pharmacology
SELECTION OF CITATIONS
SEARCH DETAIL
...