1
|
Mirabelli M, Chiefari E, Arcidiacono B, Salatino A, Pascarella A, Morelli M, Credendino SC, Brunetti FS, Di Vito A, Greco A, Huin V, Nicoletti F, Pierantoni GM, Fedele M, Aguglia U, Foti DP, Brunetti A. HMGA1 deficiency: a pathogenic link between tau pathology and insulin resistance. EBioMedicine 2025; 115:105700. [PMID: 40233659 PMCID: PMC12019291 DOI: 10.1016/j.ebiom.2025.105700] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2024] [Revised: 03/29/2025] [Accepted: 04/01/2025] [Indexed: 04/17/2025] Open
Abstract
BACKGROUND Growing evidence links tau-related neurodegeneration with insulin resistance and type 2 diabetes (T2D), though the underlying mechanisms remain unclear. Our previous research identified HMGA1 as crucial for insulin receptor (INSR) expression, with defects in the HMGA1 gene associated with insulin resistance and T2D. Here, we explore HMGA1 deficiency as a potential contributor to tauopathies, such as Alzheimer's disease (AD), and its connection to insulin resistance. METHODS Immunoblot analyses, protein-DNA interaction studies, ChIP-qPCR, and reporter gene assays were conducted in human and mouse neuronal cell models. Tau immunohistochemistry, behavioural studies, and brain glucose metabolism were analysed in Hmga1-knockout mice. Additionally, a case-control study investigated the relationship between HMGA1 and tau pathology in patients with tauopathy, carrying or not the HMGA1 rs146052672 variant, known to reduce HMGA1 protein levels and increase the risk of insulin resistance and T2D. FINDINGS We show that HMGA1 regulates tau protein expression primarily through the specific repression of MAPT gene transcription. In both human neuronal cells and primary mouse neurons, tau mRNA and protein levels were inversely correlated with HMGA1 expression. This inverse relationship was further confirmed in the brain of Hmga1-knockout mice, where tau was overexpressed, INSR was downregulated, and brain glucose uptake was impaired. Additionally, the rs146052672 variant was more common in patients with tauopathy (12/69, 17.4%) than in controls (10/200, 5.0%) (p = 0.001), and carriers of this variant exhibited more severe disease progression and poorer therapeutic outcomes. INTERPRETATION These findings suggest that HMGA1 deficiency may drive tau pathology, linking tauopathies to insulin resistance and providing new insights into the relationship between metabolic and neurodegenerative disorders. Furthermore, our observation that over 17% of individuals with tauopathy exhibit a deficit in HMGA1 protein production could have significant clinical implications, potentially guiding the development of therapeutic strategies targeting this specific defect. FUNDING See acknowledgements section.
Collapse
Affiliation(s)
- Maria Mirabelli
- Department of Health Sciences, University of Catanzaro "Magna Græcia", Catanzaro, Italy
| | - Eusebio Chiefari
- Department of Health Sciences, University of Catanzaro "Magna Græcia", Catanzaro, Italy
| | - Biagio Arcidiacono
- Department of Health Sciences, University of Catanzaro "Magna Græcia", Catanzaro, Italy
| | - Alessandro Salatino
- Department of Health Sciences, University of Catanzaro "Magna Græcia", Catanzaro, Italy
| | - Angelo Pascarella
- Department of Medical and Surgical Sciences, University of Catanzaro "Magna Græcia", Catanzaro, Italy
| | - Maurizio Morelli
- Department of Medical and Surgical Sciences, University of Catanzaro "Magna Græcia", Catanzaro, Italy
| | - Sara C Credendino
- Department of Molecular Medicine and Medical Biotechnology, University of Naples "Federico II", Naples, Italy
| | - Francesco S Brunetti
- Department of Health Sciences, University of Catanzaro "Magna Græcia", Catanzaro, Italy
| | - Anna Di Vito
- Department of Experimental and Clinical Medicine, University of Catanzaro "Magna Græcia", Catanzaro, Italy
| | - Adelaide Greco
- Interdepartmental Centre of Veterinary Radiology, University of Naples "Federico II", Naples, Italy
| | - Vincent Huin
- University of Lille, Inserm, CHU-Lille, Lille Neuroscience & Cognition, UMR-S1172, Team Alzheimer & Tauopathies, F-59000, Lille, France
| | - Ferdinando Nicoletti
- Department of Physiology and Pharmacology, University of Rome "Sapienza", Rome, Italy; IRCCS Neuromed, Pozzilli, Italy
| | - Giovanna M Pierantoni
- Department of Molecular Medicine and Medical Biotechnology, University of Naples "Federico II", Naples, Italy
| | - Monica Fedele
- Institute of Experimental Endocrinology and Oncology, CNR, Naples, Italy
| | - Umberto Aguglia
- Department of Medical and Surgical Sciences, University of Catanzaro "Magna Græcia", Catanzaro, Italy
| | - Daniela P Foti
- Department of Experimental and Clinical Medicine, University of Catanzaro "Magna Græcia", Catanzaro, Italy.
| | - Antonio Brunetti
- Department of Health Sciences, University of Catanzaro "Magna Græcia", Catanzaro, Italy.
| |
Collapse
|
2
|
Yin S, Song G, Gao N, Gao H, Zeng Q, Lu P, Zhang Q, Xu K, He J. Identifying Genetic Architecture of Carcass and Meat Quality Traits in a Ningxiang Indigenous Pig Population. Genes (Basel) 2023; 14:1308. [PMID: 37510213 PMCID: PMC10378861 DOI: 10.3390/genes14071308] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2023] [Revised: 06/16/2023] [Accepted: 06/19/2023] [Indexed: 07/30/2023] Open
Abstract
Ningxiang pig is a breed renowned for its exceptional meat quality, but it possesses suboptimal carcass traits. To elucidate the genetic architecture of meat quality and carcass traits in Ningxiang pigs, we assessed heritability and executed a genome-wide association study (GWAS) concerning carcass length, backfat thickness, meat color parameters (L.LD, a.LD, b.LD), and pH at two postmortem intervals (45 min and 24 h) within a Ningxiang pig population. Heritability estimates ranged from moderate to high (0.30~0.80) for carcass traits and from low to high (0.11~0.48) for meat quality traits. We identified 21 significant SNPs, the majority of which were situated within previously documented QTL regions. Furthermore, the GRM4 gene emerged as a pleiotropic gene that correlated with carcass length and backfat thickness. The ADGRF1, FKBP5, and PRIM2 genes were associated with carcass length, while the NIPBL gene was linked to backfat thickness. These genes hold the potential for use in selective breeding programs targeting carcass traits in Ningxiang pigs.
Collapse
Affiliation(s)
- Shishu Yin
- College of Animal Science and Technology, Hunan Agricultural University, Changsha 410128, China
| | - Gang Song
- College of Animal Science and Technology, Hunan Agricultural University, Changsha 410128, China
- Laboratory of Animal Nutrition Physiology and Metabolism, The Chinese Academy of Sciences, The Institute of Subtropical Agriculture, Changsha 410128, China
| | - Ning Gao
- College of Animal Science and Technology, Hunan Agricultural University, Changsha 410128, China
| | - Hu Gao
- College of Animal Science and Technology, Hunan Agricultural University, Changsha 410128, China
- Laboratory of Animal Nutrition Physiology and Metabolism, The Chinese Academy of Sciences, The Institute of Subtropical Agriculture, Changsha 410128, China
| | - Qinghua Zeng
- College of Animal Science and Technology, Hunan Agricultural University, Changsha 410128, China
| | - Peng Lu
- Center of Ningxiang Animal Husbandry and Fishery Affairs, Ningxiang 410625, China
| | - Qin Zhang
- College of Animal Science and Technology, Hunan Agricultural University, Changsha 410128, China
- College of Animal Science and Technology, China Agricultural University, Beijing 100091, China
| | - Kang Xu
- Laboratory of Animal Nutrition Physiology and Metabolism, The Chinese Academy of Sciences, The Institute of Subtropical Agriculture, Changsha 410128, China
| | - Jun He
- College of Animal Science and Technology, Hunan Agricultural University, Changsha 410128, China
| |
Collapse
|
3
|
Iuliano S, Greco EA, Mirabelli M, Chiefari E, Caroleo P, Puccio L, Giuliano S, Foti DP, Brunetti A, Aversa A. Predicting the response to SGLT-2 inhibitors as add-on therapy to multiple day injection insulin with glycated albumin: a pilot study. Minerva Endocrinol (Torino) 2022; 47:379-387. [PMID: 35103458 DOI: 10.23736/s2724-6507.22.03691-0] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/11/2023]
Abstract
BACKGROUND Achieving optimal glycemic targets is the main therapeutic goal in patients with type 2 diabetes (T2D) mellitus. HbA1c is the reference biomarker for monitoring glycemic control; however, in specific conditions affecting erythrocyte turnover or in patients on multiple daily injection (MDI) insulin regimens, the determination of glycated albumin (GA) may be preferable. Sodium-glucose cotransporter-2 (SGLT-2) inhibitors represent a novel class of antidiabetic drugs that lower plasma glucose concentrations quickly, with insulin-independent mechanisms. Herein, we explored the role of GA in predicting the short-term response to SGLT-2 inhibitors as add-on to MDI insulin. METHODS Sixteen patients with long-standing, poorly controlled T2D on MDI insulin starting an SGLT-2 inhibitor were subjected to plasma GA and HbA1c measurements at 30 days intervals for up to 3 months in order to examine the temporal changes of these glycemic biomarkers. RESULTS At the end of the study, grossly coincident with the life span of erythrocytes, a significant decrease in median HbA1c was observed, (from 8.7 [range: 8.2-9.3%] at baseline to 7.2 [range: 7.0-7.9%]), with the advantage of less insulin dose requirements. However, significant, and incremental reductions in median GA determinations could be already evident after 30 days (-3.5 [range: -7.5, -2.5%]) and 60 days (-6.4 [range: -10.5, -4.7%]) from the start of SGLT-2 inhibitor treatment and persisted for up to 3 months (-8.6 [range: -12.1, 6.1%]). The decrements of HbA1c observed at the 3-month visit were highly correlated with the concurrent absolute reductions of plasma GA (ρ=0.550, P=0.027), whereas a borderline significance could be demonstrated with reference to reductions in plasma GA at 30 and 60 days. CONCLUSIONS Although limited by the small number of participants, these preliminary findings suggest that GA, rather than HbA1c, could represent a useful and reliable biomarker in T2D to monitor the early glucose-lowering effects of antidiabetic drugs with rapid onset of action, such as SGLT-2 inhibitors and MDI insulin.
Collapse
Affiliation(s)
- Stefano Iuliano
- Department of Experimental and Clinical Medicine, The Magna Græcia University of Catanzaro, Catanzaro, Italy
| | - Emanuela A Greco
- Department of Health Sciences, The Magna Græcia University of Catanzaro, Catanzaro, Italy
| | - Maria Mirabelli
- Department of Health Sciences, The Magna Græcia University of Catanzaro, Catanzaro, Italy
| | - Eusebio Chiefari
- Department of Health Sciences, The Magna Græcia University of Catanzaro, Catanzaro, Italy
| | | | | | - Stefania Giuliano
- Department of Health Sciences, The Magna Græcia University of Catanzaro, Catanzaro, Italy
| | - Daniela P Foti
- Department of Experimental and Clinical Medicine, The Magna Græcia University of Catanzaro, Catanzaro, Italy
| | - Antonio Brunetti
- Department of Health Sciences, The Magna Græcia University of Catanzaro, Catanzaro, Italy -
| | - Antonio Aversa
- Department of Experimental and Clinical Medicine, The Magna Græcia University of Catanzaro, Catanzaro, Italy
| |
Collapse
|
4
|
Saed L, Jeleń A, Mirowski M, Sałagacka-Kubiak A. Prognostic Significance of HMGA1 Expression in Lung Cancer Based on Bioinformatics Analysis. Int J Mol Sci 2022; 23:ijms23136933. [PMID: 35805937 PMCID: PMC9266824 DOI: 10.3390/ijms23136933] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2022] [Revised: 06/18/2022] [Accepted: 06/18/2022] [Indexed: 12/15/2022] Open
Abstract
High-mobility group protein 1 (HMGA1) participates in the processes of DNA transcription, replication, recombination, and repair. The HMGA1 gene is expressed abundantly during embryogenesis and is reactivated during carcinogenesis. HMGA1 gene expression has been associated with a high degree of malignancy, metastatic tendency, and poor survival in breast, colon, ovary, and pancreatic cancers. However, its prognostic significance in lung cancer remains unclear. Using publicly available data, HMGA1 was shown to be overexpressed in both small and non-small lung tumors, with higher expression compared to both the adjacent non-malignant lung tissues and non-tumor lung tissues of healthy individuals. Elevated HMGA1 expression could result from lowered HMGA1 methylation and was connected with some clinicopathological features like sex, age, and stage of the disease. The high HMGA1 expression level was connected with shorter overall and first progression survival time among lung adenocarcinoma patients, but not lung squamous cell carcinoma patients. HMGA1 could interact with proteins involved in cellular senescence and cell cycle control (TP53, RB1, RPS6KB1, and CDK1), transcription regulation (EP400 and HMGA2), chromatin assembly and remodeling (LMNB1), and cholesterol and isoprene biosynthesis (HMGCR and INSIG1). Taken together, HMGA1 overexpression could be an essential element of lung carcinogenesis and a prognostic feature in lung cancer.
Collapse
|
5
|
Insulin Resistance and Cancer: In Search for a Causal Link. Int J Mol Sci 2021; 22:ijms222011137. [PMID: 34681797 PMCID: PMC8540232 DOI: 10.3390/ijms222011137] [Citation(s) in RCA: 76] [Impact Index Per Article: 19.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2021] [Revised: 10/11/2021] [Accepted: 10/12/2021] [Indexed: 12/12/2022] Open
Abstract
Insulin resistance (IR) is a condition which refers to individuals whose cells and tissues become insensitive to the peptide hormone, insulin. Over the recent years, a wealth of data has made it clear that a synergistic relationship exists between IR, type 2 diabetes mellitus, and cancer. Although the underlying mechanism(s) for this association remain unclear, it is well established that hyperinsulinemia, a hallmark of IR, may play a role in tumorigenesis. On the other hand, IR is strongly associated with visceral adiposity dysfunction and systemic inflammation, two conditions which favor the establishment of a pro-tumorigenic environment. Similarly, epigenetic modifications, such as DNA methylation, histone modifications, and non-coding RNA, in IR states, have been often associated with tumorigenesis in numerous types of human cancer. In addition to these observations, it is also broadly accepted that gut microbiota may play an intriguing role in the development of IR-related diseases, including type 2 diabetes and cancer, whereas potential chemopreventive properties have been attributed to some of the most commonly used antidiabetic medications. Herein we provide a concise overview of the most recent literature in this field and discuss how different but interrelated molecular pathways may impact on tumor development.
Collapse
|
6
|
Imbroisi Filho R, Ochioni AC, Esteves AM, Leandro JGB, Demaria TM, Sola-Penna M, Zancan P. Western diet leads to aging-related tumorigenesis via activation of the inflammatory, UPR, and EMT pathways. Cell Death Dis 2021; 12:643. [PMID: 34162829 PMCID: PMC8222293 DOI: 10.1038/s41419-021-03929-9] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2021] [Revised: 06/10/2021] [Accepted: 06/11/2021] [Indexed: 12/11/2022]
Abstract
Among the principal causative factors for the development of complications related to aging is a diet rich in fats and sugars, also known as the Western diet. This diet advocates numerous changes that might increase the susceptibility to initiate cancer and/or to create a tissue microenvironment more conducive to the growth of malignant cells, thus favoring the progression of cancer and metastasis. Hypercaloric diets in general lead to oxidative stress generating reactive oxygen species and induce endoplasmic reticulum stress. Our results demonstrate that mice bearing tumors fed with a Western diet presented bigger tumor mass with increased insulin sensitivity in these tissues. Several markers of insulin signaling, such as AKT phosphorylation and mTOR pathway, are promoted in tumors of Western diet-fed animals. This process is associated with increased macrophage infiltration, activation of unfolded protein response pathway, and initiation of epithelial-mesenchymal transition (EMT) process in these tumor tissues. Summing up, we propose that the Western diet accelerates the aging-related processes favoring tumor development.
Collapse
MESH Headings
- Age Factors
- Animals
- Cell Line, Tumor
- Cell Proliferation
- Diet, High-Fat/adverse effects
- Diet, Western/adverse effects
- Epithelial-Mesenchymal Transition/drug effects
- Female
- Gene Expression Regulation, Neoplastic
- Inflammation Mediators/metabolism
- Melanoma, Experimental/genetics
- Melanoma, Experimental/immunology
- Melanoma, Experimental/metabolism
- Melanoma, Experimental/pathology
- Mice, Inbred C57BL
- Receptor, Insulin/genetics
- Receptor, Insulin/metabolism
- Signal Transduction
- Skin Neoplasms/genetics
- Skin Neoplasms/immunology
- Skin Neoplasms/metabolism
- Skin Neoplasms/pathology
- Stearoyl-CoA Desaturase/genetics
- Stearoyl-CoA Desaturase/metabolism
- Time Factors
- Tumor Burden
- Tumor Microenvironment
- Unfolded Protein Response/genetics
- Mice
Collapse
Affiliation(s)
- Ricardo Imbroisi Filho
- Laboratório de Oncobiologia Molecular, Departamento de Biotecnologia Farmacêutica, Faculdade de Farmácia, Universidade Federal Do Rio de Janeiro, Rio de Janeiro, 21941-902, Brazil
| | - Alan C Ochioni
- Laboratório de Oncobiologia Molecular, Departamento de Biotecnologia Farmacêutica, Faculdade de Farmácia, Universidade Federal Do Rio de Janeiro, Rio de Janeiro, 21941-902, Brazil
| | - Amanda M Esteves
- Laboratório de Oncobiologia Molecular, Departamento de Biotecnologia Farmacêutica, Faculdade de Farmácia, Universidade Federal Do Rio de Janeiro, Rio de Janeiro, 21941-902, Brazil
| | - João G B Leandro
- Laboratório de Enzimologia e Controle do Metabolismo, Departamento de Biotecnologia Farmacêutica, Faculdade de Farmácia, Universidade Federal Do Rio de Janeiro, Rio de Janeiro, 21941-902, Brazil
| | - Thainá M Demaria
- Laboratório de Enzimologia e Controle do Metabolismo, Departamento de Biotecnologia Farmacêutica, Faculdade de Farmácia, Universidade Federal Do Rio de Janeiro, Rio de Janeiro, 21941-902, Brazil
| | - Mauro Sola-Penna
- Laboratório de Enzimologia e Controle do Metabolismo, Departamento de Biotecnologia Farmacêutica, Faculdade de Farmácia, Universidade Federal Do Rio de Janeiro, Rio de Janeiro, 21941-902, Brazil
| | - Patricia Zancan
- Laboratório de Oncobiologia Molecular, Departamento de Biotecnologia Farmacêutica, Faculdade de Farmácia, Universidade Federal Do Rio de Janeiro, Rio de Janeiro, 21941-902, Brazil.
| |
Collapse
|
7
|
Mirabelli M, Chiefari E, Tocci V, Caroleo P, Giuliano S, Greco E, Luque RM, Puccio L, Foti DP, Aversa A, Brunetti A. Clinical Effectiveness and Safety of Once-Weekly GLP-1 Receptor Agonist Dulaglutide as Add-On to Metformin or Metformin Plus Insulin Secretagogues in Obesity and Type 2 Diabetes. J Clin Med 2021; 10:jcm10050985. [PMID: 33801192 PMCID: PMC7957905 DOI: 10.3390/jcm10050985] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2021] [Revised: 02/12/2021] [Accepted: 02/19/2021] [Indexed: 12/13/2022] Open
Abstract
AIMS AND METHODS The aim of this monocentric retrospective observational study was to evaluate the 18-month safety and effectiveness of GLP-1 receptor agonist (GLP-1 RA) dulaglutide (DU) 1.5 mg/once weekly as an add-on to metformin (MET) or MET plus conventional insulin secretagogues in a study cohort with excess body weight and type 2 diabetes (T2D). Comparative efficacy versus liraglutide (LIRA) 1.2-1.8 mg/once daily in a study sample naïve to GLP-1 RAs, frequency matching for age, gender, T2D duration, degree of glycemic impairment, cardiovascular comorbidities, and medications, was addressed as a secondary aim. Clinical and biochemical data for efficacy outcomes and information on drug discontinuation due to adverse events (AEs) were collected from digital records. RESULTS Initial analysis included 126 overweight and obese T2D patients (48.4% females). Out of these, 13 discontinued DU due to moderate-severe gastrointestinal AEs after a mean follow-up of 6 (4 standard deviations (SD)) months, while 65 completed 18 months of continuous therapy. At 6 months, there was a significant mean HbA1c reduction of -0.85% (1.17 SD) with respect to baseline values (p < 0.001), which remained stable during 18 months follow-up. These results were accompanied by a moderate weight loss sustained over time, with a mean reduction of -2.0% (4.3 SD) at 6 months and -1.3% (4.8 SD) at 18 months (p = 0.091). At univariate analysis, a negative correlation between baseline body mass index (BMI) and risk of drug discontinuation due to gastrointestinal AEs was observed. The protective effect of obesity against drug discontinuation was confirmed by logistic regression analysis. Neither gender, nor age, nor T2D duration, nor concomitant conventional insulin secretagogue use, nor switching to DU from other GLP-1 RAs influenced its long-term effectiveness. However, higher baseline HbA1c values emerged as predictors of clinically relevant efficacy outcomes, either in terms of HbA1c reduction ≥ 0.5% or body weight loss ≥ 5%. The efficacy outcomes were corroborated by head-to-head comparison with LIRA, a GLP-1 RA with durable beneficial effects on glycemic control and body weight in real-world experiences. With the advantage of once-weekly administration, at 18-month follow-up, a significantly larger fraction of patients on DU therapy reached glycemic targets (HbA1c ≤ 7.0%) when compared to those on LIRA: from 14.8% at baseline (both groups) to 64.8% with DU and 42.6% with LIRA (p = 0.033). CONCLUSIONS Although limited by a retrospective design and lack of constant up-titration for LIRA to the highest dose, these findings indicate that the beneficial responses to DU on a background of MET or MET plus insulin secretagogues are durable, especially in the presence of obesity and greater HbA1c impairment.
Collapse
Affiliation(s)
- Maria Mirabelli
- Department of Health Sciences, University “Magna Græcia” of Catanzaro, 88100 Catanzaro, Italy; (M.M.); (E.C.); (E.G.); (D.P.F.)
| | - Eusebio Chiefari
- Department of Health Sciences, University “Magna Græcia” of Catanzaro, 88100 Catanzaro, Italy; (M.M.); (E.C.); (E.G.); (D.P.F.)
| | - Vera Tocci
- Unit of Endocrinology, Azienda Ospedaliera Mater-Domini, 88100 Catanzaro, Italy; (V.T.); (S.G.); (A.A.)
| | - Patrizia Caroleo
- Unit of Endocrinology and Diabetes, Azienda Ospedaliera Pugliese-Ciaccio, 88100 Catanzaro, Italy; (P.C.); (L.P.)
| | - Stefania Giuliano
- Unit of Endocrinology, Azienda Ospedaliera Mater-Domini, 88100 Catanzaro, Italy; (V.T.); (S.G.); (A.A.)
| | - Emanuela Greco
- Department of Health Sciences, University “Magna Græcia” of Catanzaro, 88100 Catanzaro, Italy; (M.M.); (E.C.); (E.G.); (D.P.F.)
| | - Raul Miguel Luque
- Department of Cell Biology, Physiology and Immunology, University of Córdoba, 14071 Córdoba, Spain;
| | - Luigi Puccio
- Unit of Endocrinology and Diabetes, Azienda Ospedaliera Pugliese-Ciaccio, 88100 Catanzaro, Italy; (P.C.); (L.P.)
| | - Daniela Patrizia Foti
- Department of Health Sciences, University “Magna Græcia” of Catanzaro, 88100 Catanzaro, Italy; (M.M.); (E.C.); (E.G.); (D.P.F.)
| | - Antonio Aversa
- Unit of Endocrinology, Azienda Ospedaliera Mater-Domini, 88100 Catanzaro, Italy; (V.T.); (S.G.); (A.A.)
- Department of Clinical and Experimental Medicine, University “Magna Græcia” of Catanzaro, 88100 Catanzaro, Italy
| | - Antonio Brunetti
- Department of Health Sciences, University “Magna Græcia” of Catanzaro, 88100 Catanzaro, Italy; (M.M.); (E.C.); (E.G.); (D.P.F.)
- Unit of Endocrinology, Azienda Ospedaliera Mater-Domini, 88100 Catanzaro, Italy; (V.T.); (S.G.); (A.A.)
- Correspondence: ; Tel.: +39-0961-3694368; Fax: +39-0961-3694147
| |
Collapse
|
8
|
Kase NG, Gretz Friedman E, Brodman M, Kang C, Gallagher EJ, LeRoith D. The midlife transition and the risk of cardiovascular disease and cancer Part I: magnitude and mechanisms. Am J Obstet Gynecol 2020; 223:820-833. [PMID: 32497614 DOI: 10.1016/j.ajog.2020.05.051] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2020] [Revised: 05/09/2020] [Accepted: 05/28/2020] [Indexed: 12/20/2022]
Abstract
Heart disease and cancer are the leading causes of death in the United States. In women, the clinical appearance of both entities-coronary heart disease and cancer (breast, endometrium, and ovary)-escalate during the decades of the midlife transition encompassing the menopause. In addition to the impact of aging, during the interval between the age of 40 and 65 years, the pathophysiologic components of metabolic syndrome also emerge and accelerate. These include visceral adiposity (measured as waist circumference), hypertension, diabetes, and dyslipidemia. Osteoporosis, osteoarthritis, sarcopenia, depression, and even cognitive decline and dementia appear, and most, if not all, are considered functionally related. Two clinical reports confirm the interaction linking the emergence of disease: endometrial cancer and metabolic syndrome. One describes the discovery of unsuspected endometrial cancer in a large series of elective hysterectomies performed in aged and metabolically susceptible populations. The other is from the Women's Health Initiative Observational Study, which found a positive interaction between endometrial cancer and metabolic syndrome regardless of the presence or absence of visceral adiposity. Both provide additional statistical support for the long-suspected causal interaction among the parallel but variable occurrence of these common entities-visceral obesity, heart disease, diabetes, cancer, and the prevalence of metabolic syndrome. Therefore, 2 critical clinical questions require analysis and answers: 1: Why do chronic diseases of adulthood-metabolic, cardiovascular, endocrine-and, in women, cancers of the breast and endometrium (tissues and tumors replete with estrogen receptors) emerge and their incidence trajectories accelerate during the postmenopausal period when little or no endogenous estradiol is available, and yet the therapeutic application of estrogen stimulates their appearance? 2: To what extent should identification of these etiologic driving forces require modification of the gynecologist's responsibilities in the care of our patients in the postreproductive decades of the female life cycle? Part l of this 2-part set of "expert reviews" defines the dimensions, gravity, and interactive synergy of each clinical challenge gynecologists face while caring for their midlife (primarily postmenopausal) patients. It describes the clinically identifiable, potentially treatable, pathogenic mechanisms driving these threats to quality of life and longevity. Part 2 (accepted, American Journal of Obstetrics & Gynecology) identifies 7 objectives of successful clinical care, offers "triage" prioritization targets, and provides feasible opportunities for insertion of primary preventive care initiatives. To implement these goals, a reprogrammed, repurposed office visit is described.
Collapse
Affiliation(s)
- Nathan G Kase
- Department of Obstetrics, Gynecology, and Reproductive Science, Icahn School of Medicine at Mount Sinai, New York, NY; Division of Endocrinology, Diabetes, and Bone Diseases, Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, NY.
| | - Elissa Gretz Friedman
- Department of Obstetrics, Gynecology, and Reproductive Science, Icahn School of Medicine at Mount Sinai, New York, NY
| | - Michael Brodman
- Department of Obstetrics, Gynecology, and Reproductive Science, Icahn School of Medicine at Mount Sinai, New York, NY
| | - Chifei Kang
- Division of Endocrinology, Diabetes, and Bone Diseases, Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, NY
| | - Emily J Gallagher
- Division of Endocrinology, Diabetes, and Bone Diseases, Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, NY
| | - Derek LeRoith
- Division of Endocrinology, Diabetes, and Bone Diseases, Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, NY
| |
Collapse
|
9
|
Bovolini A, Garcia J, Andrade MA, Duarte JA. Metabolic Syndrome Pathophysiology and Predisposing Factors. Int J Sports Med 2020; 42:199-214. [PMID: 33075830 DOI: 10.1055/a-1263-0898] [Citation(s) in RCA: 175] [Impact Index Per Article: 35.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Metabolic syndrome (MetS) is a cluster of cardiometabolic risk factors with high prevalence among adult populations and elevated costs for public health systems worldwide. Despite the lack of consensus regarding the syndrome definition and diagnosis criteria, it is characterized by the coexistence of risk factors such as abdominal obesity, atherogenic dyslipidemia, elevated blood pressure, a prothrombotic and pro-inflammatory state, insulin resistance (IR), and higher glucose levels, factors indubitably linked to an increased risk of developing chronic conditions, such as type 2 diabetes (T2D) and cardiovascular disease (CVD). The syndrome has a complex and multifaceted origin not fully understood; however, it has been strongly suggested that sedentarism and unbalanced dietary patterns might play a fundamental role in its development. The purpose of this review is to provide an overview from the syndrome epidemiology, costs, and main etiological traits from its relationship with unhealthy diet patterns and sedentary lifestyles.
Collapse
Affiliation(s)
| | - Juliana Garcia
- Centre for the Research and Technology of Agro-Environmental and Biological Sciences, University of Trás-os-Montes and Alto Douro, Vila Real
| | | | - José Alberto Duarte
- CIAFEL Faculty of Sport, University of Porto, Porto.,University Institute of Health Sciences (IUCS), Rua Central de Gandra, 1317 4585-116 Gandra Paredes, Portugal
| |
Collapse
|
10
|
Greco M, Chiefari E, Accattato F, Corigliano DM, Arcidiacono B, Mirabelli M, Liguori R, Brunetti FS, Pullano SA, Scorcia V, Fiorillo AS, Foti DP, Brunetti A. MicroRNA-1281 as a Novel Circulating Biomarker in Patients With Diabetic Retinopathy. Front Endocrinol (Lausanne) 2020; 11:528. [PMID: 32849308 PMCID: PMC7417427 DOI: 10.3389/fendo.2020.00528] [Citation(s) in RCA: 36] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/06/2020] [Accepted: 06/29/2020] [Indexed: 01/10/2023] Open
Abstract
Objective: Recently, the role of circulating miRNAs as non-invasive biomarkers for the identification and monitoring of diabetes microvascular complications has emerged. Herein, we aimed to: identify circulating miRNAs differentially expressed in patients with and without diabetic retinopathy (DR); examine their predictive value; and understand their pathogenic impact. Methods: Pooled serum samples from randomly selected matched patients with type 2 diabetes, either with or without DR, were used for initial serum miRNA profiling. Validation of the most relevant miRNAs was thereafter conducted by RT-qPCR in an extended sample of patients with DR and matched controls. Results: Following miRNA profiling, 43 miRNAs were significantly up- or down-regulated in patients with DR compared with controls. After individual validation, 5 miRNAs were found significantly overexpressed in patients with DR. One of them, miR-1281, was the most up-regulated and appeared to be specifically related to DR. Furthermore, secreted levels of miR-1281 were increased in high glucose-cultured retinal cells, and there was evidence of a potential link between glucose-induced miR-1281 up-regulation and DR. Conclusion: Our findings suggest miR-1281 as a circulating biomarker of DR. Also, they highlight the pathogenic significance of miR-1281, providing insights for a new potential target in treating DR.
Collapse
Affiliation(s)
- Marta Greco
- Department of Health Sciences, University of Catanzaro “Magna Græcia,”Catanzaro, Italy
| | - Eusebio Chiefari
- Department of Health Sciences, University of Catanzaro “Magna Græcia,”Catanzaro, Italy
| | - Francesca Accattato
- Department of Health Sciences, University of Catanzaro “Magna Græcia,”Catanzaro, Italy
| | | | - Biagio Arcidiacono
- Department of Health Sciences, University of Catanzaro “Magna Græcia,”Catanzaro, Italy
| | - Maria Mirabelli
- Department of Health Sciences, University of Catanzaro “Magna Græcia,”Catanzaro, Italy
| | - Rossella Liguori
- Department of Health Sciences, University of Catanzaro “Magna Græcia,”Catanzaro, Italy
| | - Francesco S. Brunetti
- Department of Health Sciences, University of Catanzaro “Magna Græcia,”Catanzaro, Italy
| | - Salvatore A. Pullano
- Department of Health Sciences, University of Catanzaro “Magna Græcia,”Catanzaro, Italy
| | - Vincenzo Scorcia
- Department of Medical and Surgical Sciences, University of Catanzaro “Magna Græcia,”Catanzaro, Italy
| | - Antonino S. Fiorillo
- Department of Health Sciences, University of Catanzaro “Magna Græcia,”Catanzaro, Italy
| | - Daniela P. Foti
- Department of Health Sciences, University of Catanzaro “Magna Græcia,”Catanzaro, Italy
| | - Antonio Brunetti
- Department of Health Sciences, University of Catanzaro “Magna Græcia,”Catanzaro, Italy
| |
Collapse
|
11
|
Mediterranean Diet Nutrients to Turn the Tide against Insulin Resistance and Related Diseases. Nutrients 2020; 12:nu12041066. [PMID: 32290535 PMCID: PMC7230471 DOI: 10.3390/nu12041066] [Citation(s) in RCA: 160] [Impact Index Per Article: 32.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2020] [Revised: 04/06/2020] [Accepted: 04/10/2020] [Indexed: 12/11/2022] Open
Abstract
Insulin resistance (IR), defined as an attenuated biological response to circulating insulin, is a fundamental defect in obesity and type 2 diabetes (T2D), and is also linked to a wide spectrum of pathological conditions, such as non-alcoholic fatty liver disease (NAFLD), cognitive impairment, endothelial dysfunction, chronic kidney disease (CKD), polycystic ovary syndrome (PCOS), and some endocrine tumors, including breast cancer. In obesity, the unbalanced production of pro- and anti-inflammatory adipocytokines can lead to the development of IR and its related metabolic complications, which are potentially reversible through weight-loss programs. The Mediterranean diet (MedDiet), characterized by high consumption of extra-virgin olive oil (EVOO), nuts, red wine, vegetables and other polyphenol-rich elements, has proved to be associated with greater improvement of IR in obese individuals, when compared to other nutritional interventions. Also, recent studies in either experimental animal models or in humans, have shown encouraging results for insulin-sensitizing nutritional supplements derived from MedDiet food sources in the modulation of pathognomonic traits of certain IR-related conditions, including polyunsaturated fatty acids from olive oil and seeds, anthocyanins from purple vegetables and fruits, resveratrol from grapes, and the EVOO-derived, oleacein. Although the pharmacological properties and clinical uses of these functional nutrients are still under investigation, the molecular mechanism(s) underlying the metabolic benefits appear to be compound-specific and, in some cases, point to a role in gene expression through an involvement of the nuclear high-mobility group A1 (HMGA1) protein.
Collapse
|
12
|
Mirabelli M, Chiefari E, Caroleo P, Arcidiacono B, Corigliano DM, Giuliano S, Brunetti FS, Tanyolaç S, Foti DP, Puccio L, Brunetti A. Long-Term Effectiveness of Liraglutide for Weight Management and Glycemic Control in Type 2 Diabetes. INTERNATIONAL JOURNAL OF ENVIRONMENTAL RESEARCH AND PUBLIC HEALTH 2019; 17:ijerph17010207. [PMID: 31892206 PMCID: PMC6981922 DOI: 10.3390/ijerph17010207] [Citation(s) in RCA: 32] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/27/2019] [Revised: 12/17/2019] [Accepted: 12/24/2019] [Indexed: 12/15/2022]
Abstract
Background: Liraglutide is the first glucagon-like peptide-1 receptor agonist (GLP-1 RA) based on the human GLP-1 sequence, with potential weight loss benefits, approved for the treatment of type 2 diabetes (T2D) mellitus. Herein, we aimed to assess the 5-year effectiveness of Liraglutide in the management of weight and glycometabolic control in a Southern Italian cohort of overweight/obese T2D patients, who were naïve to GLP-1 RAs. Patients and Methods: Forty overweight or obese patients treated with Liraglutide at doses up to 1.8 mg/day, in combination with one or more oral antidiabetic agents, were retrospectively assessed at baseline, during, and after 60 months of continuous therapy. Results: After 5 years of Liraglutide treatment, body weight decreased from 92.1 ± 20.5 kg to 87.3 ± 20.0 Kg (p < 0.001), with a mean reduction of 5.0 ± 7.0 Kg and a body mass index (BMI) decrement of −2.0 ± 3.1 Kg/m2. On Spearman’s univariate analysis, change in body weight was correlated with female gender and baseline BMI. Hemoglobin A1c (HbA1c) decreased from 7.9 ± 0.9% at baseline to 7.0 ± 0.7% at the end of the study period (p < 0.001), followed by a significant reduction in fasting plasma glucose. No significant differences emerged in other biochemical parameters, despite a trend toward improvement in lipid profile. Notwithstanding encouraging effects on several markers of cardiovascular disease (CVD), increments in the 5- and 10-year risk for the first atherosclerotic cardiovascular event were documented, as four incident cases of myocardial infarction. Conclusions: Prolonging treatment with Liraglutide can lead to durable benefits in relation to weight and glycemic control, with a greater impact on women. These results extend and corroborate previous observations, suggesting that gender per se may modulate the response to Liraglutide. Despite favorable effects on some established CVD risks factors, the long-term role of Liraglutide in primary prevention of CVD in patients with T2D remains controversial.
Collapse
Affiliation(s)
- Maria Mirabelli
- Department of Health Sciences, University “Magna Græcia” of Catanzaro, 88100 Catanzaro, Italy; (M.M.); (E.C.); (B.A.); (D.M.C.); (S.G.); (F.S.B.); (D.P.F.)
| | - Eusebio Chiefari
- Department of Health Sciences, University “Magna Græcia” of Catanzaro, 88100 Catanzaro, Italy; (M.M.); (E.C.); (B.A.); (D.M.C.); (S.G.); (F.S.B.); (D.P.F.)
| | - Patrizia Caroleo
- Complex Operative Structure Endocrinology-Diabetology, Hospital Pugliese-Ciaccio, 88100 Catanzaro, Italy; (P.C.); (L.P.)
| | - Biagio Arcidiacono
- Department of Health Sciences, University “Magna Græcia” of Catanzaro, 88100 Catanzaro, Italy; (M.M.); (E.C.); (B.A.); (D.M.C.); (S.G.); (F.S.B.); (D.P.F.)
| | - Domenica Maria Corigliano
- Department of Health Sciences, University “Magna Græcia” of Catanzaro, 88100 Catanzaro, Italy; (M.M.); (E.C.); (B.A.); (D.M.C.); (S.G.); (F.S.B.); (D.P.F.)
| | - Stefania Giuliano
- Department of Health Sciences, University “Magna Græcia” of Catanzaro, 88100 Catanzaro, Italy; (M.M.); (E.C.); (B.A.); (D.M.C.); (S.G.); (F.S.B.); (D.P.F.)
| | - Francesco Saverio Brunetti
- Department of Health Sciences, University “Magna Græcia” of Catanzaro, 88100 Catanzaro, Italy; (M.M.); (E.C.); (B.A.); (D.M.C.); (S.G.); (F.S.B.); (D.P.F.)
| | - Sinan Tanyolaç
- Division of Endocrinology and Metabolism, Department of Internal Medicine, School of Medicine, Biruni University, 34010 Istanbul, Turkey;
| | - Daniela Patrizia Foti
- Department of Health Sciences, University “Magna Græcia” of Catanzaro, 88100 Catanzaro, Italy; (M.M.); (E.C.); (B.A.); (D.M.C.); (S.G.); (F.S.B.); (D.P.F.)
| | - Luigi Puccio
- Complex Operative Structure Endocrinology-Diabetology, Hospital Pugliese-Ciaccio, 88100 Catanzaro, Italy; (P.C.); (L.P.)
| | - Antonio Brunetti
- Department of Health Sciences, University “Magna Græcia” of Catanzaro, 88100 Catanzaro, Italy; (M.M.); (E.C.); (B.A.); (D.M.C.); (S.G.); (F.S.B.); (D.P.F.)
- Correspondence: ; Tel.: +39-0961-3694368; Fax: +39-0961-3694147
| |
Collapse
|
13
|
Visconti F, Quaresima P, Chiefari E, Caroleo P, Arcidiacono B, Puccio L, Mirabelli M, Foti DP, Di Carlo C, Vero R, Brunetti A. First Trimester Combined Test (FTCT) as a Predictor of Gestational Diabetes Mellitus. INTERNATIONAL JOURNAL OF ENVIRONMENTAL RESEARCH AND PUBLIC HEALTH 2019; 16:ijerph16193654. [PMID: 31569431 PMCID: PMC6801433 DOI: 10.3390/ijerph16193654] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/02/2019] [Revised: 09/25/2019] [Accepted: 09/26/2019] [Indexed: 01/27/2023]
Abstract
Background—The first trimester combined test (FTCT) is an effective screening tool to estimate the risk of fetal aneuploidy. It is obtained by the combination of maternal age, ultrasound fetal nuchal translucency (NT) measurement, and the maternal serum markers free β-human chorionic gonadotropin (β-hCG) and pregnancy-associated plasma protein A (PAPP-A). However, conflicting data have been reported about the association of FTCT, β-hCG, or PAPP-A with the subsequent diagnosis of gestational diabetes mellitus (GDM). Research design and methods—2410 consecutive singleton pregnant women were retrospectively enrolled in Calabria, Southern Italy. All participants underwent examinations for FTCT at 11–13 weeks (plus 6 days) of gestation, and screening for GDM at 16–18 and/or 24–28 weeks of gestation, in accordance with current Italian guidelines and the International Association Diabetes Pregnancy Study Groups (IADPSG) glycemic cut-offs. Data were examined by univariate and logistic regression analyses. Results—1814 (75.3%) pregnant women were normal glucose tolerant, while 596 (24.7%) were diagnosed with GDM. Spearman univariate analysis demonstrated a correlation between FTCT values and subsequent GDM diagnosis (ρ = 0.048, p = 0.018). The logistic regression analysis showed that women with a FTCT <1:10000 had a major GDM risk (p = 0.016), similar to women with a PAPP-A <1 multiple of the expected normal median (MoM, p = 0.014). Conversely, women with β-hCG ≥2.0 MoM had a reduced risk of GDM (p = 0.014). Conclusions—Our findings indicate that GDM susceptibility increases with fetal aneuploidy risk, and that FTCT and its related maternal serum parameters can be used as early predictors of GDM.
Collapse
Affiliation(s)
- Federica Visconti
- Unit of Obstetrics and Gynecology, Department of Medical and Surgical Sciences, University "Magna Græcia" of Catanzaro, Viale Europa, 88100 Catanzaro, Italy.
| | - Paola Quaresima
- Unit of Obstetrics and Gynecology, Department of Medical and Surgical Sciences, University "Magna Græcia" of Catanzaro, Viale Europa, 88100 Catanzaro, Italy.
| | - Eusebio Chiefari
- Department of Health Sciences, University "Magna Græcia" of Catanzaro, 88100 Catanzaro, Italy.
| | - Patrizia Caroleo
- Complex Operative Structure Endocrinology-Diabetology, Hospital Pugliese-Ciaccio, 88100 Catanzaro, Italy.
| | - Biagio Arcidiacono
- Department of Health Sciences, University "Magna Græcia" of Catanzaro, 88100 Catanzaro, Italy.
| | - Luigi Puccio
- Complex Operative Structure Endocrinology-Diabetology, Hospital Pugliese-Ciaccio, 88100 Catanzaro, Italy.
| | - Maria Mirabelli
- Department of Health Sciences, University "Magna Græcia" of Catanzaro, 88100 Catanzaro, Italy.
| | - Daniela P Foti
- Department of Health Sciences, University "Magna Græcia" of Catanzaro, 88100 Catanzaro, Italy.
| | - Costantino Di Carlo
- Unit of Obstetrics and Gynecology, Department of Medical and Surgical Sciences, University "Magna Græcia" of Catanzaro, Viale Europa, 88100 Catanzaro, Italy.
| | - Raffaella Vero
- Complex Operative Structure Endocrinology-Diabetology, Hospital Pugliese-Ciaccio, 88100 Catanzaro, Italy.
| | - Antonio Brunetti
- Department of Health Sciences, University "Magna Græcia" of Catanzaro, 88100 Catanzaro, Italy.
| |
Collapse
|
14
|
Jiao H, Zang Y, Zhang M, Zhang Y, Wang Y, Wang K, Price RA, Li WD. Genome-Wide Interaction and Pathway Association Studies for Body Mass Index. Front Genet 2019; 10:404. [PMID: 31118946 PMCID: PMC6504780 DOI: 10.3389/fgene.2019.00404] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2018] [Accepted: 04/12/2019] [Indexed: 11/24/2022] Open
Abstract
Objective: We investigated gene interactions (epistasis) for body mass index (BMI) in a European-American adult female cohort via genome-wide interaction analyses (GWIA) and pathway association analyses. Methods: Genome-wide pairwise interaction analyses were carried out for BMI in 493 extremely obese cases (BMI > 35 kg/m2) and 537 never-overweight controls (BMI < 25 kg/m2). To further validate the results, specific SNPs were selected based on the GWIA results for haplotype-based association studies. Pathway-based association analyses were performed using a modified Gene Set Enrichment Algorithm (GSEA) (GenGen program) to further explore BMI-related pathways using our genome wide association study (GWAS) data set, GIANT, ENGAGE, and DIAGRAM Consortia. Results: The EXOC4-1q23.1 interaction was associated with BMI, with the most significant epistasis between rs7800006 and rs10797020 (P = 2.63 × 10-11). In the pathway-based association analysis, Tob1 pathway showed the most significant association with BMI (empirical P < 0.001, FDR = 0.044, FWER = 0.040). These findings were further validated in different populations. Conclusion: Genome-wide pairwise SNP-SNP interaction and pathway analyses suggest that EXOC4 and TOB1-related pathways may contribute to the development of obesity.
Collapse
Affiliation(s)
- Hongxiao Jiao
- Research Center of Basic Medical Sciences, Tianjin Medical University, Tianjin, China
| | - Yong Zang
- Department of Genetics, College of Basic Medical Sciences, Tianjin Medical University, Tianjin, China
| | - Miaomiao Zhang
- Department of Genetics, College of Basic Medical Sciences, Tianjin Medical University, Tianjin, China
| | - Yuan Zhang
- Department of Genetics, College of Basic Medical Sciences, Tianjin Medical University, Tianjin, China
| | - Yaogang Wang
- College of Public Health, Tianjin Medical University, Tianjin, China
| | - Kai Wang
- Raymond G. Perelman Center for Cellular and Molecular Therapeutics, Children’s Hospital of Philadelphia, Philadelphia, PA, United States
- Laboratory Medicine, Department of Pathology, University of Pennsylvania, Philadelphia, PA, United States
| | - R. Arlen Price
- Department of Psychiatry, Center for Neurobiology and Behavior, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, United States
| | - Wei-Dong Li
- Department of Genetics, College of Basic Medical Sciences, Tianjin Medical University, Tianjin, China
| |
Collapse
|
15
|
Mirabelli M, Chiefari E, Caroleo P, Vero R, Brunetti FS, Corigliano DM, Arcidiacono B, Foti DP, Puccio L, Brunetti A. Long-Term Effectiveness and Safety of SGLT-2 Inhibitors in an Italian Cohort of Patients with Type 2 Diabetes Mellitus. J Diabetes Res 2019; 2019:3971060. [PMID: 31781664 PMCID: PMC6875368 DOI: 10.1155/2019/3971060] [Citation(s) in RCA: 26] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/11/2019] [Accepted: 10/14/2019] [Indexed: 02/06/2023] Open
Abstract
BACKGROUND SGLT-2 (sodium-glucose cotransporter-2) inhibitors are a novel class of oral hypoglycemic agents for the management of type 2 diabetes mellitus (T2DM). Herein, we aimed to assess the long-term effectiveness and safety of SGLT-2 inhibitors in a Southern Italy population of subjects affected by T2DM. PATIENTS AND METHODS 408 diabetic patients treated with one of the three SGLT-2 inhibitors currently available in Italy (dapagliflozin, empagliflozin, and canagliflozin), either alone or in combination with other antidiabetic drugs, were retrospectively assessed at baseline, during, and after 18 months of continuous therapy. RESULTS Treatment with SGLT-2 inhibitors resulted in a median decrease in HbA1c of 0.9%, with a percentage of decrement of 12 in relation to the baseline value, followed by a significant reduction (P < 0.001) in fasting plasma glucose. Variations in HbA1c occurred independently of the baseline clinical or biochemical characteristics. In addition, treatment with SGLT-2 inhibitors reduced body weight (P < 0.008) and decreased diastolic blood pressure (P = 0.004). With regard to safety outcomes, 66 patients out of 91 stopped SGLT-2 inhibitors during follow-up because of chronic or recurring genital infections, while the rest experienced other adverse events, such as urinary tract infections, polyuria, nausea, hypotension, dizziness, acute coronary event, worsening of glycemic control status, and rapid deterioration of renal function. CONCLUSION In our patients' population, the glycometabolic effects of SGLT-2 inhibitors were durable and comparable to those observed in multicenter randomized controlled trials. This notwithstanding safety concerns must be raised regarding the frequent occurrence of genitourinary infections and the risk of a rapid decline of renal function in patients with evidence of volume depletion and/or receiving other medications which can adversely affect kidney function.
Collapse
Affiliation(s)
- Maria Mirabelli
- Department of Health Sciences, University “Magna Græcia” of Catanzaro, Catanzaro, Italy
| | - Eusebio Chiefari
- Department of Health Sciences, University “Magna Græcia” of Catanzaro, Catanzaro, Italy
| | - Patrizia Caroleo
- Complex Operative Structure Endocrinology-Diabetology, Hospital Pugliese-Ciaccio, Catanzaro, Italy
| | - Raffaella Vero
- Complex Operative Structure Endocrinology-Diabetology, Hospital Pugliese-Ciaccio, Catanzaro, Italy
| | | | | | - Biagio Arcidiacono
- Department of Health Sciences, University “Magna Græcia” of Catanzaro, Catanzaro, Italy
| | - Daniela Patrizia Foti
- Department of Health Sciences, University “Magna Græcia” of Catanzaro, Catanzaro, Italy
| | - Luigi Puccio
- Complex Operative Structure Endocrinology-Diabetology, Hospital Pugliese-Ciaccio, Catanzaro, Italy
| | - Antonio Brunetti
- Department of Health Sciences, University “Magna Græcia” of Catanzaro, Catanzaro, Italy
| |
Collapse
|
16
|
Liu Y, Zheng L, Kong H, Wang Q, Tian X. HMGA1 variant IVS5-13insC is associated with insulin resistance and type 2 diabetes: an updated meta-analysis. Afr Health Sci 2018; 18:865-872. [PMID: 30766549 PMCID: PMC6354845 DOI: 10.4314/ahs.v18i4.4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022] Open
Abstract
Background High-mobility group A1 (HMGA1) polymorphism has been suspected as a gene variant associated with type 2 diabetes (T2D). However, conflicting outcomes have been reported. Objective This meta-analysis aimed to predict the association between the HMGA1 variant IVS5-13insC and T2D. Methods Statistical analyses were performed using Stata/SE 12.0 software. Results A total of 11 case-control studies in 6 articles were included. Results suggested that the HMGA1 variant IVS5-13insC was associated with an increased risk of insulin resistance (OR = 0.61, 95% CI 0.56 to 0.66, P < 0.0001), T2D (OR = 0.67, 95% CI 0.61 to 0.73, P < 0.0001), particularly for Caucasians with increased risks of T2D (OR = 0.56, 95% CI 0.49 to 0.65, P < 0.0001) compared with wild-type subjects. Conclusion This meta-analysis indicated that the HMGA1 variant IVS5-13insC can be a risk factor of T2D development, particularly among Caucasians. Significant risks were also found (Asian: OR = 0.74, 95% CI: 0.63 to 0.86, P < 0.0001, Hispanic-American: OR = 0.81, 95% CI: 0.65 to 1.01, P < 0.0001) in non-Caucasian population. However, ethnical studies should be conducted to reveal whether the HMGA1 variant IVS5-13insC is associated with an increased risk of T2D.
Collapse
|
17
|
Vella V, Milluzzo A, Scalisi NM, Vigneri P, Sciacca L. Insulin Receptor Isoforms in Cancer. Int J Mol Sci 2018; 19:ijms19113615. [PMID: 30453495 PMCID: PMC6274710 DOI: 10.3390/ijms19113615] [Citation(s) in RCA: 71] [Impact Index Per Article: 10.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2018] [Revised: 11/05/2018] [Accepted: 11/13/2018] [Indexed: 12/17/2022] Open
Abstract
The insulin receptor (IR) mediates both metabolic and mitogenic effects especially when overexpressed or in clinical conditions with compensatory hyperinsulinemia, due to the metabolic pathway resistance, as obesity diabetes. In many cancers, IR is overexpressed preferentially as IR-A isoform, derived by alternative splicing of exon 11. The IR-A overexpression, and the increased IR-A:IR-B ratio, are mechanisms that promote the mitogenic response of cancer cells to insulin and IGF-2, which is produced locally by both epithelial and stromal cancer cells. In cancer IR-A, isoform predominance may occur for dysregulation at both mRNA transcription and post-transcription levels, including splicing factors, non-coding RNAs and protein degradation. The mechanisms that regulate IR isoform expression are complex and not fully understood. The IR isoform overexpression may play a role in cancer cell stemness, in tumor progression and in resistance to target therapies. From a clinical point of view, the IR-A overexpression in cancer may be a determinant factor for the resistance to IGF-1R target therapies for this issue. IR isoform expression in cancers may have the meaning of a predictive biomarker and co-targeting IGF-1R and IR-A may represent a new more efficacious treatment strategy.
Collapse
Affiliation(s)
- Veronica Vella
- Department of Clinical and Experimental Medicine, Endocrinology Section, University of Catania Medical School, Garibaldi-Nesima Hospital, via Palermo 636, 95122 Catania, Italy.
- School of Human and Social Science, University "Kore" of Enna, 94100 Enna, Italy.
| | - Agostino Milluzzo
- Department of Clinical and Experimental Medicine, Endocrinology Section, University of Catania Medical School, Garibaldi-Nesima Hospital, via Palermo 636, 95122 Catania, Italy.
| | - Nunzio Massimo Scalisi
- Department of Clinical and Experimental Medicine, Endocrinology Section, University of Catania Medical School, Garibaldi-Nesima Hospital, via Palermo 636, 95122 Catania, Italy.
| | - Paolo Vigneri
- Department of Clinical and Experimental Medicine, University of Catania Medical School, Center of Experimental Oncology and Hematology, A.O.U. Policlinico Vittorio Emanuele, via Santa Sofia, 78, 95123 Catania, Italy.
| | - Laura Sciacca
- Department of Clinical and Experimental Medicine, Endocrinology Section, University of Catania Medical School, Garibaldi-Nesima Hospital, via Palermo 636, 95122 Catania, Italy.
| |
Collapse
|
18
|
Kang C, LeRoith D, Gallagher EJ. Diabetes, Obesity, and Breast Cancer. Endocrinology 2018; 159:3801-3812. [PMID: 30215698 PMCID: PMC6202853 DOI: 10.1210/en.2018-00574] [Citation(s) in RCA: 141] [Impact Index Per Article: 20.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/13/2018] [Accepted: 09/05/2018] [Indexed: 12/13/2022]
Abstract
The rates of obesity and diabetes are increasing worldwide, whereas the age of onset for both obesity and diabetes are decreasing steadily. Obesity and diabetes are associated with multiple factors that contribute to the increased risk of a number of different cancers, including breast cancer. These factors are hyperinsulinemia, elevated IGFs, hyperglycemia, dyslipidemia, adipokines, inflammatory cytokines, and the gut microbiome. In this review, we discuss the current understanding of the complex signaling pathways underlying these multiple factors involved in the obesity/diabetes-breast cancer link, with a focus particularly on the roles of the insulin/IGF system and dyslipidemia in preclinical breast cancer models. We review some of the therapeutic strategies to target these metabolic derangements in cancer. Future research directions and potential therapeutic strategies are also discussed.
Collapse
Affiliation(s)
- Chifei Kang
- Division of Endocrinology, Diabetes and Bone Disease, Icahn School of Medicine at Mount Sinai, New York, New York
| | - Derek LeRoith
- Division of Endocrinology, Diabetes and Bone Disease, Icahn School of Medicine at Mount Sinai, New York, New York
| | - Emily J Gallagher
- Division of Endocrinology, Diabetes and Bone Disease, Icahn School of Medicine at Mount Sinai, New York, New York
| |
Collapse
|
19
|
Cross-talk among HMGA1 and FoxO1 in control of nuclear insulin signaling. Sci Rep 2018; 8:8540. [PMID: 29867121 PMCID: PMC5986867 DOI: 10.1038/s41598-018-26968-3] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2017] [Accepted: 05/21/2018] [Indexed: 12/27/2022] Open
Abstract
As a mediator of insulin-regulated gene expression, the FoxO1 transcription factor represents a master regulator of liver glucose metabolism. We previously reported that the high-mobility group AT-hook 1 (HMGA1) protein, a molecular switch for the insulin receptor gene, functions also as a downstream target of the insulin receptor signaling pathway, representing a critical nuclear mediator of insulin function. Here, we investigated whether a functional relationship existed between FoxO1 and HMGA1, which might help explain insulin-mediated gene transcription in the liver. To this end, as a model study, we investigated the canonical FoxO1-HMGA1-responsive IGFBP1 gene, whose hepatic expression is regulated by insulin. By using a conventional GST-pull down assay combined with co-immunoprecipitation and Fluorescence Resonance Energy Transfer (FRET) analyses, we provide evidence of a physical interaction between FoxO1 and HMGA1. Further investigation with chromatin immunoprecipitation, confocal microscopy, and Fluorescence Recovery After Photobleaching (FRAP) technology indicated a functional significance of this interaction, in both basal and insulin-stimulated states, providing evidence that, by modulating FoxO1 transactivation, HMGA1 is essential for FoxO1-induced IGFBP1 gene expression, and thereby a critical modulator of insulin-mediated FoxO1 regulation in the liver. Collectively, our findings highlight a novel FoxO1/HMGA1-mediated mechanism by which insulin may regulate gene expression and metabolism.
Collapse
|
20
|
Comment on Li et al. HMGA1: A novel predisposing gene for acute myocardial infarction. Int J Cardiol 2018; 256:38. [DOI: 10.1016/j.ijcard.2018.01.109] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/18/2018] [Accepted: 01/23/2018] [Indexed: 11/18/2022]
|
21
|
Arcidiacono B, Chiefari E, Messineo S, Bilotta FL, Pastore I, Corigliano DM, Foti DP, Brunetti A. HMGA1 is a novel transcriptional regulator of the FoxO1 gene. Endocrine 2018; 60:56-64. [PMID: 29052178 PMCID: PMC5845622 DOI: 10.1007/s12020-017-1445-8] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/12/2017] [Accepted: 09/27/2017] [Indexed: 12/14/2022]
Abstract
PURPOSE The forkhead transcription factor (FoxO1) is a master transcriptional regulator of fundamental cellular processes ranging from cell proliferation and differentiation to inflammation and metabolism. However, despite its relevance, the mechanism(s) underlying FoxO1 gene regulation are largely unknown. We have previously shown that the chromatin factor high-mobility group A1 (HMGA1) plays a key role in the transcriptional regulation of glucose-responsive genes, including some that are involved in FoxO1-mediated glucose metabolism. Here we investigated the impact of HMGA1 on FoxO1 gene expression. METHODS FoxO1 protein and gene expression studies were performed by Western blot analysis combined with qRT-PCR of material from human cultured cells and EBV-transformed lymphoblasts, and from primary cultured hepatocytes from wild-type and Hmga1 -/- mice. Reporter gene assays and chromatin immunoprecipitation for binding of HMGA1 to the endogenous FoxoO1 locus were performed in cells overexpressing HMGA1 and in cells pretreated with siRNA targeting HMGA1. RESULTS HMGA1 increased FoxO1 mRNA and protein expression in vitro, in cultured HepG2 and HEK-293 cells by binding FoxO1 gene promoter, thereby activating FoxO1 gene transcription. Forced expression of HMGA1 in primary cultured hepatocytes from Hmga1 -/- mice and in EBV-transformed lymphoblasts from subjects with reduced expression of endogenous HMGA1 increased FoxO1 mRNA and protein levels. CONCLUSION These findings may contribute to the understanding of FoxO1 gene regulation and its role in metabolism.
Collapse
Affiliation(s)
- Biagio Arcidiacono
- Department of Health Sciences, University "Magna Græcia" of Catanzaro, Viale Europa (Località Germaneto), 88100, Catanzaro, Italy
| | - Eusebio Chiefari
- Department of Health Sciences, University "Magna Græcia" of Catanzaro, Viale Europa (Località Germaneto), 88100, Catanzaro, Italy
| | - Sebastiano Messineo
- Department of Health Sciences, University "Magna Græcia" of Catanzaro, Viale Europa (Località Germaneto), 88100, Catanzaro, Italy
| | - Francesco L Bilotta
- Department of Health Sciences, University "Magna Græcia" of Catanzaro, Viale Europa (Località Germaneto), 88100, Catanzaro, Italy
| | - Ida Pastore
- Department of Health Sciences, University "Magna Græcia" of Catanzaro, Viale Europa (Località Germaneto), 88100, Catanzaro, Italy
| | - Domenica M Corigliano
- Department of Health Sciences, University "Magna Græcia" of Catanzaro, Viale Europa (Località Germaneto), 88100, Catanzaro, Italy
| | - Daniela P Foti
- Department of Health Sciences, University "Magna Græcia" of Catanzaro, Viale Europa (Località Germaneto), 88100, Catanzaro, Italy
| | - Antonio Brunetti
- Department of Health Sciences, University "Magna Græcia" of Catanzaro, Viale Europa (Località Germaneto), 88100, Catanzaro, Italy.
| |
Collapse
|
22
|
Zhang Q, Chen L, Zhao Z, Wu Y, Zhong J, Wen G, Cao R, Zu X, Liu J. HMGA1 Mediated High-Glucose-Induced Vascular Smooth Muscle Cell Proliferation in Diabetes Mellitus: Association Between PI3K/Akt Signaling and HMGA1 Expression. DNA Cell Biol 2018; 37:389-397. [PMID: 29634420 DOI: 10.1089/dna.2017.3957] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022] Open
Abstract
High-mobility group protein A1 (HMGA1), an architectural transcription factor, was found to regulate multiple gene expression in mammals. Recent studies firmly indicate an association between HMGA1 and type 2 diabetes. However, the presence and function of HMGA1 in diabetic vasculopathy has not been substantiated. in this study, we first determined the HMGA1 changes in aorta tissue of diabetic rats. In streptozotocin-induced diabetic rats, a higher level of blood glucose and plasma lipids, an increase of intima-media thickness, and a significant upregulation and accumulation of HMGA1, mainly in the nucleus and around the nuclear membrane of vascular smooth muscle cells (VSMCs), were detected. In vitro, high glucose increased HMGA1 expression and promoted proliferation of VSMCs, which could be blunted by Wortmannin and LY294002, inhibitors of PI3K/Akt pathway, and specificity protein 1 (SP1) siRNA. Moreover, knockdown of HMGA1 could weaken the upregulation of cyclin D1 accompanied by high-glucose-induced HMGA1 in VSMCs. Taken together, these findings demonstrate the vital role of PI3K/Akt-SP1-HMGA1 pathway in high-glucose-induced VSMCs proliferation.
Collapse
Affiliation(s)
- Qinghai Zhang
- Department of Metabolism and Endocrinology, The First Affiliated Hospital, University of South China , Hengyang, Hunan, P.R. China
| | - Ling Chen
- Department of Metabolism and Endocrinology, The First Affiliated Hospital, University of South China , Hengyang, Hunan, P.R. China
| | - Zhibo Zhao
- Department of Metabolism and Endocrinology, The First Affiliated Hospital, University of South China , Hengyang, Hunan, P.R. China
| | - Ying Wu
- Department of Metabolism and Endocrinology, The First Affiliated Hospital, University of South China , Hengyang, Hunan, P.R. China
| | - Jing Zhong
- Department of Metabolism and Endocrinology, The First Affiliated Hospital, University of South China , Hengyang, Hunan, P.R. China
| | - Gebo Wen
- Department of Metabolism and Endocrinology, The First Affiliated Hospital, University of South China , Hengyang, Hunan, P.R. China
| | - Renxian Cao
- Department of Metabolism and Endocrinology, The First Affiliated Hospital, University of South China , Hengyang, Hunan, P.R. China
| | - Xuyu Zu
- Department of Metabolism and Endocrinology, The First Affiliated Hospital, University of South China , Hengyang, Hunan, P.R. China
| | - Jianghua Liu
- Department of Metabolism and Endocrinology, The First Affiliated Hospital, University of South China , Hengyang, Hunan, P.R. China
| |
Collapse
|
23
|
Pastore I, Chiefari E, Vero R, Brunetti A. Postpartum glucose intolerance: an updated overview. Endocrine 2018; 59:481-494. [PMID: 28808874 DOI: 10.1007/s12020-017-1388-0] [Citation(s) in RCA: 46] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/23/2017] [Accepted: 07/28/2017] [Indexed: 12/19/2022]
Abstract
The prevalence of type 2 diabetes mellitus has increased worldwide over the past three decades, as a consequence of the more westernized lifestyle, which is responsible for the increasing obesity rate in the modern adult's life. Concomitant with this increase there has been a gradual rise in the overall prevalence of gestational diabetes mellitus, a condition that strongly predisposes to overt diabetes later in life. Many women with previous gestational diabetes mellitus show glucose intolerance in the early postpartum period. Although the best screening strategy for postpartum glucose intolerance is still debated, numerous evidences indicate that identification of these women at this time is of critical importance, as efforts to initiate early intensive lifestyle modification, including hypocaloric diet and physical activity, and to ameliorate the metabolic profile of these high-risk subjects can prevent or delay the onset of type 2 diabetes mellitus. Nevertheless, less than one fifth of women attend the scheduled postpartum screening following gestational diabetes mellitus and they are at increased risk to develop type 2 diabetes mellitus later in their lives. Unsatisfying results have also come from early intervention strategies and tools that have been developed during the last few years to help improving the rate of adherence to postpartum glycemic testing, thereby indicating that more effective strategies are needed to improve women's participation in postpartum screening.
Collapse
Affiliation(s)
- Ida Pastore
- Department of Health Sciences, University "Magna Græcia" of Catanzaro, Viale Europa (Loc. Germaneto), Catanzaro, 88100, Italy
| | - Eusebio Chiefari
- Department of Health Sciences, University "Magna Græcia" of Catanzaro, Viale Europa (Loc. Germaneto), Catanzaro, 88100, Italy
| | - Raffaella Vero
- Complex Operative Structure Endocrinology-Diabetology, Hospital Pugliese-Ciaccio, Catanzaro, 88100, Italy
| | - Antonio Brunetti
- Department of Health Sciences, University "Magna Græcia" of Catanzaro, Viale Europa (Loc. Germaneto), Catanzaro, 88100, Italy.
| |
Collapse
|
24
|
Chiefari E, Foti DP, Sgarra R, Pegoraro S, Arcidiacono B, Brunetti FS, Greco M, Manfioletti G, Brunetti A. Transcriptional Regulation of Glucose Metabolism: The Emerging Role of the HMGA1 Chromatin Factor. Front Endocrinol (Lausanne) 2018; 9:357. [PMID: 30034366 PMCID: PMC6043803 DOI: 10.3389/fendo.2018.00357] [Citation(s) in RCA: 41] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/22/2018] [Accepted: 06/13/2018] [Indexed: 02/06/2023] Open
Abstract
HMGA1 (high mobility group A1) is a nonhistone architectural chromosomal protein that functions mainly as a dynamic regulator of chromatin structure and gene transcription. As such, HMGA1 is involved in a variety of fundamental cellular processes, including gene expression, epigenetic regulation, cell differentiation and proliferation, as well as DNA repair. In the last years, many reports have demonstrated a role of HMGA1 in the transcriptional regulation of several genes implicated in glucose homeostasis. Initially, it was proved that HMGA1 is essential for normal expression of the insulin receptor (INSR), a critical link in insulin action and glucose homeostasis. Later, it was demonstrated that HMGA1 is also a downstream nuclear target of the INSR signaling pathway, representing a novel mediator of insulin action and function at this level. Moreover, other observations have indicated the role of HMGA1 as a positive modulator of the Forkhead box protein O1 (FoxO1), a master regulatory factor for gluconeogenesis and glycogenolysis, as well as a positive regulator of the expression of insulin and of a series of circulating proteins that are involved in glucose counterregulation, such as the insulin growth factor binding protein 1 (IGFBP1), and the retinol binding protein 4 (RBP4). Thus, several lines of evidence underscore the importance of HMGA1 in the regulation of glucose production and disposal. Consistently, lack of HMGA1 causes insulin resistance and diabetes in humans and mice, while variations in the HMGA1 gene are associated with the risk of type 2 diabetes and metabolic syndrome, two highly prevalent diseases that share insulin resistance as a common pathogenetic mechanism. This review intends to give an overview about our current knowledge on the role of HMGA1 in glucose metabolism. Although research in this field is ongoing, many aspects still remain elusive. Future directions to improve our insights into the pathophysiology of glucose homeostasis may include epigenetic studies and the use of "omics" strategies. We believe that a more comprehensive understanding of HMGA1 and its networks may reveal interesting molecular links between glucose metabolism and other biological processes, such as cell proliferation and differentiation.
Collapse
Affiliation(s)
- Eusebio Chiefari
- Department of Health Sciences, University “Magna Græcia” of Catanzaro, Catanzaro, Italy
| | - Daniela P. Foti
- Department of Health Sciences, University “Magna Græcia” of Catanzaro, Catanzaro, Italy
| | - Riccardo Sgarra
- Department of Life Sciences, University of Trieste, Trieste, Italy
| | - Silvia Pegoraro
- Department of Life Sciences, University of Trieste, Trieste, Italy
| | - Biagio Arcidiacono
- Department of Health Sciences, University “Magna Græcia” of Catanzaro, Catanzaro, Italy
| | - Francesco S. Brunetti
- Department of Medical and Surgical Sciences, University “Magna Græcia” of Catanzaro, Catanzaro, Italy
| | - Manfredi Greco
- Department of Clinical and Experimental Medicine, University “Magna Græcia” of Catanzaro, Catanzaro, Italy
| | | | - Antonio Brunetti
- Department of Health Sciences, University “Magna Græcia” of Catanzaro, Catanzaro, Italy
- *Correspondence: Antonio Brunetti
| |
Collapse
|
25
|
De Rosa S, Arcidiacono B, Chiefari E, Brunetti A, Indolfi C, Foti DP. Type 2 Diabetes Mellitus and Cardiovascular Disease: Genetic and Epigenetic Links. Front Endocrinol (Lausanne) 2018; 9:2. [PMID: 29387042 PMCID: PMC5776102 DOI: 10.3389/fendo.2018.00002] [Citation(s) in RCA: 227] [Impact Index Per Article: 32.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/27/2017] [Accepted: 01/03/2018] [Indexed: 12/14/2022] Open
Abstract
Type 2 diabetes mellitus (DM) is a common metabolic disorder predisposing to diabetic cardiomyopathy and atherosclerotic cardiovascular disease (CVD), which could lead to heart failure through a variety of mechanisms, including myocardial infarction and chronic pressure overload. Pathogenetic mechanisms, mainly linked to hyperglycemia and chronic sustained hyperinsulinemia, include changes in metabolic profiles, intracellular signaling pathways, energy production, redox status, increased susceptibility to ischemia, and extracellular matrix remodeling. The close relationship between type 2 DM and CVD has led to the common soil hypothesis, postulating that both conditions share common genetic and environmental factors influencing this association. However, although the common risk factors of both CVD and type 2 DM, such as obesity, insulin resistance, dyslipidemia, inflammation, and thrombophilia, can be identified in the majority of affected patients, less is known about how these factors influence both conditions, so that efforts are still needed for a more comprehensive understanding of this relationship. The genetic, epigenetic, and environmental backgrounds of both type 2 DM and CVD have been more recently studied and updated. However, the underlying pathogenetic mechanisms have seldom been investigated within the broader shared background, but rather studied in the specific context of type 2 DM or CVD, separately. As the precise pathophysiological links between type 2 DM and CVD are not entirely understood and many aspects still require elucidation, an integrated description of the genetic, epigenetic, and environmental influences involved in the concomitant development of both diseases is of paramount importance to shed new light on the interlinks between type 2 DM and CVD. This review addresses the current knowledge of overlapping genetic and epigenetic aspects in type 2 DM and CVD, including microRNAs and long non-coding RNAs, whose abnormal regulation has been implicated in both disease conditions, either etiologically or as cause for their progression. Understanding the links between these disorders may help to drive future research toward an integrated pathophysiological approach and to provide future directions in the field.
Collapse
Affiliation(s)
- Salvatore De Rosa
- Department of Medical and Surgical Sciences, Magna Græcia University of Catanzaro, Catanzaro, Italy
| | - Biagio Arcidiacono
- Department of Health Sciences, Magna Græcia University of Catanzaro, Catanzaro, Italy
| | - Eusebio Chiefari
- Department of Health Sciences, Magna Græcia University of Catanzaro, Catanzaro, Italy
| | - Antonio Brunetti
- Department of Health Sciences, Magna Græcia University of Catanzaro, Catanzaro, Italy
- *Correspondence: Antonio Brunetti, ; Ciro Indolfi, ; Daniela P. Foti,
| | - Ciro Indolfi
- Department of Medical and Surgical Sciences, Magna Græcia University of Catanzaro, Catanzaro, Italy
- *Correspondence: Antonio Brunetti, ; Ciro Indolfi, ; Daniela P. Foti,
| | - Daniela P. Foti
- Department of Health Sciences, Magna Græcia University of Catanzaro, Catanzaro, Italy
- *Correspondence: Antonio Brunetti, ; Ciro Indolfi, ; Daniela P. Foti,
| |
Collapse
|
26
|
Arcidiacono B, Chiefari E, Laria AE, Messineo S, Bilotta FL, Britti D, Foti DP, Foryst-Ludwig A, Kintscher U, Brunetti A. Expression of matrix metalloproteinase-11 is increased under conditions of insulin resistance. World J Diabetes 2017; 8:422-428. [PMID: 28989568 PMCID: PMC5612832 DOI: 10.4239/wjd.v8.i9.422] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/30/2017] [Revised: 04/04/2017] [Accepted: 05/05/2017] [Indexed: 02/05/2023] Open
Abstract
AIM To investigate matrix metalloproteinase-11 (MMP-11) expression in adipose tissue dysfunction, using in vitro and in vivo models of insulin resistance.
METHODS Culture of mouse 3T3-L1 preadipocytes were induced to differentiation into mature 3T3-L1 adipocytes. Cellular insulin resistance was induced by treating differentiated cultured adipocytes with hypoxia and/or tumor necrosis factor (TNF)-α, and transcriptional changes were analyzed in each condition thereafter. For the in vivo studies, MMP-11 expression levels were measured in white adipose tissue (WAT) from C57BL/6J mice that underwent low fat diet or high-fat feeding in order to induce obesity and obesity-related insulin resistance. Statistical analysis was carried out with GraphPad Prism Software.
RESULTS MMP-11 mRNA expression levels were significantly higher in insulin resistant 3T3-L1 adipocytes compared to control cells (1.46 ± 0.49 vs 0.83 ± 0.21, respectively; P < 0.00036). The increase in MMP-11 expression was observed even in the presence of TNF-α alone (3.79 ± 1.11 vs 1 ± 0.17, P < 0.01) or hypoxia alone (1.79 ± 0.7 vs 0.88 ± 0.1, P < 0.00023). The results obtained in in vitro experiments were confirmed in the in vivo model of insulin resistance. In particular, MMP-11 mRNA was upregulated in WAT from obese mice compared to lean mice (5.5 ± 2.8 vs 1.1 ± 0.7, respectively; P < 3.72E-08). The increase in MMP-11 levels in obese mice was accompanied by the increase in typical markers of fibrosis, such as collagen type VI alpha 3 (Col6α3), and fibroblast-specific protein 1.
CONCLUSION Our results indicate that dysregulation of MMP-11 expression is an early process in the adipose tissue dysfunction, which leads to obesity and obesity-related insulin resistance.
Collapse
Affiliation(s)
- Biagio Arcidiacono
- Department of Health Sciences, University “Magna Græcia” of Catanzaro, 88100 Catanzaro, Italy
| | - Eusebio Chiefari
- Department of Health Sciences, University “Magna Græcia” of Catanzaro, 88100 Catanzaro, Italy
| | - Anna Elisa Laria
- Department of Health Sciences, University “Magna Græcia” of Catanzaro, 88100 Catanzaro, Italy
| | - Sebastiano Messineo
- Department of Health Sciences, University “Magna Græcia” of Catanzaro, 88100 Catanzaro, Italy
| | | | - Domenico Britti
- Department of Health Sciences, University “Magna Græcia” of Catanzaro, 88100 Catanzaro, Italy
| | - Daniela Patrizia Foti
- Department of Health Sciences, University “Magna Græcia” of Catanzaro, 88100 Catanzaro, Italy
| | - Anna Foryst-Ludwig
- Institute of Pharmacology, Center for Cardiovascular Research, 10117 Berlin, Germany
| | - Ulrich Kintscher
- Institute of Pharmacology, Center for Cardiovascular Research, 10117 Berlin, Germany
| | - Antonio Brunetti
- Department of Health Sciences, University “Magna Græcia” of Catanzaro, 88100 Catanzaro, Italy
| |
Collapse
|
27
|
Zhang S, Lei R, Wu J, Shan J, Hu Z, Chen L, Ren X, Yao L, Wang J, Wang X. Role of high mobility group A1 and body mass index in the prognosis of patients with breast cancer. Oncol Lett 2017; 14:5719-5726. [PMID: 29113200 PMCID: PMC5661362 DOI: 10.3892/ol.2017.6963] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2016] [Accepted: 03/09/2017] [Indexed: 12/20/2022] Open
Abstract
The high mobility group A1 (HMGA1) protein is associated with poor prognosis in patients with a wide range of cancers. However, the affect of HMGA1 on the risk of mortality from breast cancer (BC) has not been fully characterized. In the present retrospective multiple center study, the HMGA1 expression level was determined by performing immunohistochemistry on surgical tissue samples of 273 BC specimens from the Second Affiliated Hospital of Zhejiang University (Zhejiang, China) and 310 BCs from the National Engineering Center for Biochip (Shanghai, China). Kaplan-Meier analysis and Cox proportional hazard model were employed to analyze the survivability. HMGA1 expression was significantly associated with tumor histological degree and body mass index (BMI). However, HMGA1 expression showed no prognostic value in patients with BC. Combined evaluation of HMGA1 expression and high BMI (≥24 kg/m2) predicted worse overall survival of BC. Therefore, HMGA1 and BMI were considered to serve synergistic roles in the development and progression of BC, and combined evaluation of HMGA1 expression and high BMI may be an effective marker in predicting poor prognosis of BC patients.
Collapse
Affiliation(s)
- Shizhen Zhang
- Department of Surgical Oncology, Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang 310009, P.R. China.,Department of Cancer Institute, Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang 310009, P.R. China
| | - Rui Lei
- Department of Plastic Surgery, First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang 310003, P.R. China
| | - Jingjing Wu
- Department of Pathology, Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang 310009, P.R. China
| | - Jinlan Shan
- Department of Surgical Oncology, Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang 310009, P.R. China.,Department of Cancer Institute, Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang 310009, P.R. China
| | - Zujian Hu
- Department of Breast Surgery, Hangzhou Traditional Chinese Medical Hospital, Hangzhou, Zhejiang 310000, P.R. China
| | - Lirong Chen
- Department of Cancer Institute, Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang 310009, P.R. China.,Department of Pathology, Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang 310009, P.R. China
| | - Xingchang Ren
- Department of Pathology, Hangzhou Traditional Chinese Medical Hospital, Hangzhou, Zhejiang 310000, P.R. China
| | - Lifang Yao
- Department of Pathology, Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang 310009, P.R. China
| | - Jian Wang
- Department of Surgical Oncology, Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang 310009, P.R. China.,Department of Cancer Institute, Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang 310009, P.R. China
| | - Xiaochen Wang
- Department of Surgical Oncology, Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang 310009, P.R. China.,Department of Cancer Institute, Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang 310009, P.R. China
| |
Collapse
|
28
|
Effect of Mediterranean Diet and Antioxidant Formulation in Non-Alcoholic Fatty Liver Disease: A Randomized Study. Nutrients 2017; 9:nu9080870. [PMID: 28805669 PMCID: PMC5579663 DOI: 10.3390/nu9080870] [Citation(s) in RCA: 96] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2017] [Revised: 07/20/2017] [Accepted: 08/08/2017] [Indexed: 12/12/2022] Open
Abstract
Non-alcoholic fatty liver disease (NAFLD) is the most common liver disease worldwide, characterized by liver fatty acid accumulation and fibrosis, not due to excessive alcohol consumption. Notably, nutritional habits have been reported to be implicated in the onset and severity of the hepatic damage, while the Mediterranean diet has shown beneficial effects on NAFLD. Free radicals and oxidative stress were suggested to be involved in the pathogenesis and progression of NAFLD, and several data highlighted the efficacy of antioxidant supplementation in its treatment. The aim of this study was to compare the effects of the Mediterranean diet, with or without an antioxidant complex supplement, in overweight patients suffering from NAFLD. In this prospective study, fifty Caucasian overweight patients were randomized into three groups (Groups A-C). A personalized moderately hypocaloric Mediterranean diet was prescribed to all patients included in the A and B groups. In addition to the diet, Group B was administered antioxidant supplementation daily and for the period of six months. Group C did not have any type of treatment. The study proved that the Mediterranean diet alone or in association with the antioxidant complex improved anthropometric parameters, lipid profile and reduced hepatic fat accumulation and liver stiffness. However, Group B patients, in which the diet was associated with antioxidant intake, showed not only a significant improvement in insulin sensitivity, but also a more consistent reduction of anthropometric parameters when compared with Group A patients. Taken together, these results support the benefit of antioxidant supplementation in overweight patients with NAFLD.
Collapse
|
29
|
Santuario-Facio SK, Cardona-Huerta S, Perez-Paramo YX, Trevino V, Hernandez-Cabrera F, Rojas-Martinez A, Uscanga-Perales G, Martinez-Rodriguez JL, Martinez-Jacobo L, Padilla-Rivas G, Muñoz-Maldonado G, Gonzalez-Guerrero JF, Valero-Gomez J, Vazquez-Guerrero AL, Martinez-Rodriguez HG, Barboza-Quintana A, Barboza-Quintana O, Garza-Guajardo R, Ortiz-Lopez R. A New Gene Expression Signature for Triple Negative Breast Cancer Using Frozen Fresh Tissue before Neoadjuvant Chemotherapy. Mol Med 2017; 23:101-111. [PMID: 28474731 PMCID: PMC5469719 DOI: 10.2119/molmed.2016.00257] [Citation(s) in RCA: 38] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2016] [Accepted: 04/17/2017] [Indexed: 12/31/2022] Open
Abstract
Triple-negative breast cancer (TNBC) is an aggressive subtype of breast cancer tumors. Comparisons between TNBC and non-triple negative breast cancer (nTNBC) may help to differentiate key components involved in TNBC neoplasms. The purpose of the study was to analyze the expression profile of TNBC versus nTNBC tumors in a homogeneous population from northeastern Mexico. A prospective study of 50 patients was conducted (25 TNBC and 25 nTNBC). Clinic parameters were equally distributed for TNBC and nTNBC: age at diagnosis (51 vs 47 years, p=0.1), glucose levels (107 mg/dl vs 104 mg/dl, p=0.64), and body mass index (28 vs 29, p=0.14), respectively. Core biopsies were collected for histopathological diagnosis and gene expression analyses. Total RNA was isolated and expression profiling was performed. 40 genes showed differential expression pattern in TNBC tumors. Among these, 9 over-expressed genes (PRKX/PRKY, UGT8, HMGA1, LPIN1, HAPLN3, and ANKRD11), and one under-expressed (ANX9) gene are involved in general metabolism. Based on this biochemical peculiarity, and the over-expression of BCL11A and FOXC1 (involved in tumor growth and metastasis, respectively) we validated by qPCR the expression profile of 7 genes out of the signature. In this report, a new gene signature for TNBC is proposed. To our knowledge, this is the first TNBC signature which describes genes involved in general metabolism. The findings may be pertinent for Mexican patients and require to be evaluated in further ethnic groups and populations.
Collapse
Affiliation(s)
- Sandra K Santuario-Facio
- Universidad Autonoma de Nuevo Leon, Centro de Investigación y Desarrollo en Ciencias de la Salud, Monterrey, Nuevo Leon, Mexico
- Tecnologico de Monterrey, Escuela Nacional de Medicina, Monterrey, Nuevo Leon, Mexico
| | - Servando Cardona-Huerta
- Tecnologico de Monterrey, Hospital San Jose, Centro de Cancer de Mama, Monterrey, Nuevo Leon, Mexico
| | - Yadira X Perez-Paramo
- Pharmaceutical Sciences Department, College of Pharmacy, Washington State University, Spokane, Washington, United States of America
| | - Victor Trevino
- Tecnológico de Monterrey, Escuela Nacional de Medicina, Grupo de Investigacion en Bioinformatica, Monterrey, Nuevo Leon, Mexico
| | - Francisco Hernandez-Cabrera
- Universidad Autonoma de Nuevo Leon, Facultad de Ciencias Fisico Matematicas, Centro Investigación en Ciencias Físico Matemáticas, Monterrey, Nuevo Leon, Mexico
| | - Augusto Rojas-Martinez
- Universidad Autonoma de Nuevo Leon, Centro de Investigación y Desarrollo en Ciencias de la Salud, Monterrey, Nuevo Leon, Mexico
- Tecnologico de Monterrey, Escuela Nacional de Medicina, Monterrey, Nuevo Leon, Mexico
| | - Grecia Uscanga-Perales
- Universidad Autonoma de Nuevo Leon, Facultad de Medicina, Departamento de Bioquimica y Medicina Molecular, Monterrey, Nuevo Leon, Mexico
| | - Jorge L Martinez-Rodriguez
- Tecnologico de Monterrey, Hospital San Jose, Centro de Cancer de Mama, Monterrey, Nuevo Leon, Mexico
- Universidad Autonoma de Nuevo Leon, Hospital Universitario Dr. Jose Eleuterio Gonzalez, Centro Universitario Contra el Cancer, Servicio de Oncologia, Monterrey, Nuevo Leon, Mexico
| | - Lizeth Martinez-Jacobo
- Universidad Autonoma de Nuevo Leon, Facultad de Medicina, Departamento de Bioquimica y Medicina Molecular, Monterrey, Nuevo Leon, Mexico
| | - Gerardo Padilla-Rivas
- Universidad Autonoma de Nuevo Leon, Facultad de Medicina, Departamento de Bioquimica y Medicina Molecular, Monterrey, Nuevo Leon, Mexico
| | - Gerardo Muñoz-Maldonado
- Universidad Autonoma de Nuevo Leon, Hospital Universitario Dr. Jose Eleuterio Gonzalez, Servicio de Cirugia General, Monterrey, Nuevo Leon, Mexico
| | - Juan Francisco Gonzalez-Guerrero
- Universidad Autonoma de Nuevo Leon, Hospital Universitario Dr. Jose Eleuterio Gonzalez, Centro Universitario Contra el Cancer, Servicio de Oncologia, Monterrey, Nuevo Leon, Mexico
| | - Javier Valero-Gomez
- Tecnologico de Monterrey, Hospital San Jose, Centro de Cancer de Mama, Monterrey, Nuevo Leon, Mexico
| | - Ana L Vazquez-Guerrero
- Tecnologico de Monterrey, Hospital San Jose, Centro de Cancer de Mama, Monterrey, Nuevo Leon, Mexico
| | - Herminia G Martinez-Rodriguez
- Universidad Autonoma de Nuevo Leon, Facultad de Medicina, Departamento de Bioquimica y Medicina Molecular, Monterrey, Nuevo Leon, Mexico
| | - Alvaro Barboza-Quintana
- Tecnologico de Monterrey, Hospital San Jose, Centro de Cancer de Mama, Monterrey, Nuevo Leon, Mexico
| | - Oralia Barboza-Quintana
- Universidad Autonoma de Nuevo Leon, Hospital Universitario Dr. Jose Eleuterio Gonzalez, Servicio de Anatomia Patologica y Citopatologia, Monterrey, Nuevo Leon, Mexico
| | - Raquel Garza-Guajardo
- Universidad Autonoma de Nuevo Leon, Hospital Universitario Dr. Jose Eleuterio Gonzalez, Servicio de Anatomia Patologica y Citopatologia, Monterrey, Nuevo Leon, Mexico
| | - Rocio Ortiz-Lopez
- Universidad Autonoma de Nuevo Leon, Centro de Investigación y Desarrollo en Ciencias de la Salud, Monterrey, Nuevo Leon, Mexico
- Tecnologico de Monterrey, Escuela Nacional de Medicina, Monterrey, Nuevo Leon, Mexico
| |
Collapse
|
30
|
Chiefari E, Ventura V, Capula C, Randazzo G, Scorcia V, Fedele M, Arcidiacono B, Nevolo MT, Bilotta FL, Vitiello M, Palmieri C, Gulletta E, Fusco A, Foti D, Vero R, Brunetti A. A polymorphism of HMGA1 protects against proliferative diabetic retinopathy by impairing HMGA1-induced VEGFA expression. Sci Rep 2016; 6:39429. [PMID: 27991577 PMCID: PMC5171873 DOI: 10.1038/srep39429] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2016] [Accepted: 11/22/2016] [Indexed: 12/18/2022] Open
Abstract
Diabetic retinopathy (DR) is a major complication of diabetes mellitus, and is the leading cause of blindness in working-age people. Usually, DR progresses from the asymptomatic non-proliferative DR that does not significantly alter vision, to proliferative DR (PDR), which can result in aberrant retinal neovessel formation and blindness. The High-Mobility-Group A1 (HMGA1) protein is a transcriptional master regulator of numerous genes, including metabolic and inflammatory genes, which, by modulating the expression of angiogenic factors, may induce retinal neovascularization, a hallmark of PDR. Herein, we examined the relationship between HMGA1 rs139876191 variant and DR. Results revealed that patients with type 2 diabetes, who were carriers of the HMGA1 rs139876191 variant had a significantly lower risk of developing PDR, compared to non-carrier diabetic patients. From a mechanistic point of view, our findings indicated that, by adversely affecting HMGA1 protein expression and function, the HMGA1 rs139876191 variant played a key role in this protective mechanism by downregulating the expression of vascular endothelial growth factor A (VEGFA), a major activator of neovascularization in DR. These data provide new insights into the pathogenesis and progression of DR, and may offer opportunities for discovering novel biomarkers and therapeutic targets for diagnosis, prevention and treatment of PDR.
Collapse
Affiliation(s)
- Eusebio Chiefari
- Department of Health Sciences, University “Magna Græcia” of Catanzaro, Catanzaro, Italy
| | - Valeria Ventura
- Department of Health Sciences, University “Magna Græcia” of Catanzaro, Catanzaro, Italy
| | - Carmelo Capula
- Operative Unit of Endocrinology and Diabetes, Hospital Pugliese-Ciaccio, Catanzaro, Italy
| | - Giorgio Randazzo
- Department of Clinical and Experimental Medicine, University “Magna Græcia” of Catanzaro, Catanzaro, Italy
| | - Vincenzo Scorcia
- Department of Clinical and Experimental Medicine, University “Magna Græcia” of Catanzaro, Catanzaro, Italy
| | - Monica Fedele
- Institute of Experimental Endocrinology and Oncology, CNR, Napoli, Italy
| | - Biagio Arcidiacono
- Department of Health Sciences, University “Magna Græcia” of Catanzaro, Catanzaro, Italy
| | - Maria Teresa Nevolo
- Department of Health Sciences, University “Magna Græcia” of Catanzaro, Catanzaro, Italy
| | | | - Michela Vitiello
- Institute of Experimental Endocrinology and Oncology, CNR, Napoli, Italy
| | - Camillo Palmieri
- Department of Clinical and Experimental Medicine, University “Magna Græcia” of Catanzaro, Catanzaro, Italy
| | - Elio Gulletta
- Department of Health Sciences, University “Magna Græcia” of Catanzaro, Catanzaro, Italy
| | - Alfredo Fusco
- Institute of Experimental Endocrinology and Oncology, CNR, Napoli, Italy
- Department of Molecular Medicine and Medical Biotechnologies, University of Naples “Federico II”, Napoli, Italy
| | - Daniela Foti
- Department of Health Sciences, University “Magna Græcia” of Catanzaro, Catanzaro, Italy
| | - Raffaella Vero
- Operative Unit of Endocrinology and Diabetes, Hospital Pugliese-Ciaccio, Catanzaro, Italy
| | - Antonio Brunetti
- Department of Health Sciences, University “Magna Græcia” of Catanzaro, Catanzaro, Italy
| |
Collapse
|
31
|
Vigneri R, Goldfine ID, Frittitta L. Insulin, insulin receptors, and cancer. J Endocrinol Invest 2016; 39:1365-1376. [PMID: 27368923 DOI: 10.1007/s40618-016-0508-7] [Citation(s) in RCA: 158] [Impact Index Per Article: 17.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/26/2016] [Accepted: 06/23/2016] [Indexed: 12/13/2022]
Abstract
Insulin is a major regulator of cell metabolism but, in addition, is also a growth factor. Insulin effects in target cells are mediated by the insulin receptor (IR), a transmembrane protein with enzymatic (tyrosine kinase) activity. The insulin receptor, however, is represented by a heterogeneous family of proteins, including two different IR isoforms and also hybrid receptors resulting from the IR hemireceptor combination with a hemireceptor of the cognate IGF-1 receptor. These different receptors may bind insulin and its analogs with different affinity and produce different biologic effects. Since many years, it is known that many cancer cells require insulin for optimal in vitro growth. Recent data indicate that: (1) insulin stimulates growth mainly via its own receptor and not the IGF-1 receptor; (2) in many cancer cells, the IR is overexpressed and the A isoform, which has a predominant mitogenic effect, is more represented than the B isoform. These characteristics provide a selective growth advantage to malignant cells when exposed to insulin. For this reason, all conditions of hyperinsulinemia, both endogenous (prediabetes, metabolic syndrome, obesity, type 2 diabetes before pancreas exhaustion and polycystic ovary syndrome) and exogenous (type 1 diabetes) will increase the risk of cancer. Cancer-related mortality is also increased in patients exposed to hyperinsulinemia but other factors, related to the different diseases, may also contribute. The complexity of the diseases associated with hyperinsulinemia and their therapies does not allow a precise evaluation of the cancer-promoting effect of hyperinsulinemia, but its detrimental effect on cancer incidence and mortality is well documented.
Collapse
Affiliation(s)
- R Vigneri
- Endocrinology, Garibaldi-Nesima Medical Center, Via Palermo 636, 95122, Catania, Italy.
- Department of Clinical and Experimental Medicine, University of Catania, Catania, Italy.
- Humanitas, Catania Cancer Center, Catania, Italy.
- CNR, Institute of Bioimages and Biostructures, Catania, Italy.
| | - I D Goldfine
- University of California, San Francisco, CA, USA
| | - L Frittitta
- Endocrinology, Garibaldi-Nesima Medical Center, Via Palermo 636, 95122, Catania, Italy
- Department of Clinical and Experimental Medicine, University of Catania, Catania, Italy
| |
Collapse
|
32
|
De Rosa S, Chiefari E, Salerno N, Ventura V, D'Ascoli GL, Arcidiacono B, Ambrosio G, Bilotta FL, Torella D, Foti D, Indolfi C, Brunetti A. HMGA1 is a novel candidate gene for myocardial infarction susceptibility. Int J Cardiol 2016; 227:331-334. [PMID: 27839822 DOI: 10.1016/j.ijcard.2016.11.088] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/02/2016] [Accepted: 11/06/2016] [Indexed: 10/20/2022]
Abstract
BACKGROUND Acute Myocardial infarction (AMI), a leading cause of morbidity and mortality worldwide, is a dreadful acute complication of coronary atherosclerosis. Type 2 diabetes mellitus (T2DM) is associated with an increased risk of developing AMI. The architectural transcription factor high-mobility-group AT-hook 1 (HMGA1) has been involved in atherosclerosis, plaque formation, inflammation, and in the pathogenesis of insulin resistance and T2DM. An association of the HMGA1 rs146052672 variant with T2DM has been recently reported. Thus, our aim was to evaluate whether this variant was also associated with AMI. METHODS AND RESULTS In a case-control study from Calabria (Southern Italy), we enrolled 254 consecutive, unrelated, patients with first diagnosis of AMI, and 508 age, sex-matched controls. Genotyping of the rs146052672 was performed using the TaqMan allelic discrimination method. We found that this variant was present in 7.9% of AMI patients and in 3.1% of controls (p=0.003). Multiple logistic regression confirmed that the rs146052672 was significantly associated with AMI (OR=2.54; p=0.002), and this association was independent of classical cardiovascular risk factors such as gender, hypertension, obesity and T2DM (for all, p<0.05). CONCLUSIONS Our findings demonstrate that a relationship exists between the HMGA1 rs146052672 variant and AMI, suggesting that defects at the HMGA1 locus may play a pathogenetic role in AMI, in the absence of T2DM and other cardiovascular risk factors.
Collapse
Affiliation(s)
- Salvatore De Rosa
- Division of Cardiology, Department of Medical and Surgical Sciences, "Magna Graecia" University, Catanzaro, Italy
| | - Eusebio Chiefari
- Department of Health Sciences, University "Magna Græcia" of Catanzaro, 88100 Catanzaro, Italy
| | - Nadia Salerno
- Division of Cardiology, Department of Medical and Surgical Sciences, "Magna Graecia" University, Catanzaro, Italy
| | - Valeria Ventura
- Department of Health Sciences, University "Magna Græcia" of Catanzaro, 88100 Catanzaro, Italy
| | - Greta Luana D'Ascoli
- Division of Cardiology, Department of Medical and Surgical Sciences, "Magna Graecia" University, Catanzaro, Italy
| | - Biagio Arcidiacono
- Department of Health Sciences, University "Magna Græcia" of Catanzaro, 88100 Catanzaro, Italy
| | - Giuseppe Ambrosio
- Division of Cardiology, Department of Medical and Surgical Sciences, "Magna Graecia" University, Catanzaro, Italy
| | | | - Daniele Torella
- Division of Cardiology, Department of Medical and Surgical Sciences, "Magna Graecia" University, Catanzaro, Italy
| | - Daniela Foti
- Department of Health Sciences, University "Magna Græcia" of Catanzaro, 88100 Catanzaro, Italy
| | - Ciro Indolfi
- Division of Cardiology, Department of Medical and Surgical Sciences, "Magna Graecia" University, Catanzaro, Italy; URT-CNR, Department of Medicine, Consiglio Nazionale delle Ricerche of IFC, Catanzaro, Italy.
| | - Antonio Brunetti
- Department of Health Sciences, University "Magna Græcia" of Catanzaro, 88100 Catanzaro, Italy.
| |
Collapse
|
33
|
Messineo S, Laria AE, Arcidiacono B, Chiefari E, Luque Huertas RM, Foti DP, Brunetti A. Cooperation between HMGA1 and HIF-1 Contributes to Hypoxia-Induced VEGF and Visfatin Gene Expression in 3T3-L1 Adipocytes. Front Endocrinol (Lausanne) 2016; 7:73. [PMID: 27445976 PMCID: PMC4921468 DOI: 10.3389/fendo.2016.00073] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/08/2016] [Accepted: 06/14/2016] [Indexed: 12/28/2022] Open
Abstract
The architectural transcription factor high-mobility group AT-hook 1 (HMGA1) is a chromatin regulator with implications in several biological processes, including tumorigenesis, inflammation, and metabolism. Previous studies have indicated a role for this factor in promoting the early stages of adipogenesis, while inhibiting adipocyte terminal differentiation, and decreasing fat mass. It has been demonstrated that hypoxia - through the hypoxia-inducible factor 1 (HIF-1) - plays a major role in triggering changes in the adipose tissue of the obese, leading to inhibition of adipocyte differentiation, adipose cell dysfunction, inflammation, insulin resistance, and type 2 diabetes. To examine the possible cooperation between HMGA1 and HIF-1, herein, we investigated the role of HMGA1 in the regulation of Visfatin and VEGF, two genes normally expressed in adipose cells, which are both responsive to hypoxia. We demonstrated that HMGA1 enhanced Visfatin and VEGF gene expression in human embryonic kidney (HEK) 293 cells in hypoxic conditions, whereas HMGA1 knockdown in differentiated 3T3-L1 adipocytes reduced these effects. Reporter gene analysis showed that Visfatin and VEGF transcriptional activity was increased by the addition of either HMGA1 or HIF-1 and even further by the combination of both factors. As demonstrated by chromatin immunoprecipitation in intact cells, HMGA1 directly interacted with the VEGF gene, and this interaction was enhanced in hypoxic conditions. Furthermore, as indicated by co-immunoprecipitation studies, HMGA1 and HIF-1 physically interacted with each other, supporting the notion that this association may corroborate a functional link between these factors. Therefore, our findings provide evidence for molecular cross-talk between HMGA1 and HIF-1, and this may be important for elucidating protein and gene networks relevant to obesity.
Collapse
Affiliation(s)
- Sebastiano Messineo
- Department of Health Sciences, University “Magna Græcia” of Catanzaro, Catanzaro, Italy
| | - Anna Elisa Laria
- Department of Health Sciences, University “Magna Græcia” of Catanzaro, Catanzaro, Italy
| | - Biagio Arcidiacono
- Department of Health Sciences, University “Magna Græcia” of Catanzaro, Catanzaro, Italy
| | - Eusebio Chiefari
- Department of Health Sciences, University “Magna Græcia” of Catanzaro, Catanzaro, Italy
| | - Raúl M. Luque Huertas
- Department of Cell Biology, Physiology and Immunology, Instituto Maimónides de Investigación Biomédica de Córdoba (IMIBIC), Hospital Universitario Reina Sofía (HURS), CIBERobn and ceiA3, University of Córdoba, Córdoba, Spain
| | - Daniela P. Foti
- Department of Health Sciences, University “Magna Græcia” of Catanzaro, Catanzaro, Italy
| | - Antonio Brunetti
- Department of Health Sciences, University “Magna Græcia” of Catanzaro, Catanzaro, Italy
| |
Collapse
|
34
|
Han X, Cao Y, Wang K, Zhu G. HMGA1 facilitates tumor progression through regulating Wnt/β-catenin pathway in endometrial cancer. Biomed Pharmacother 2016; 82:312-8. [PMID: 27470368 DOI: 10.1016/j.biopha.2016.05.004] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2016] [Revised: 05/02/2016] [Accepted: 05/02/2016] [Indexed: 10/21/2022] Open
Abstract
Recent studies have identified a unique role for high mobility group protein A1 (HMGA1) as a major regulator of tumor progression and in diverse tumor models. Emerging evidences indicate that overexpressed HMGA1 facilitates multiple malignant phenotypes of cancer cells, however, the oncogenic activities of HMGA1 in endometrial cancer (EC) remains elusive. Here we showed that HMGA1 was more frequently expressed in human EC tissues compared to non-tumor tissues. Elevated HMGA1 was significantly associated with advanced clinical stage. Wound-healing assay and transwell assay showed that HMGA1 can positively regulate cell migration and invasion. Mechanistically, luciferase reporter assay and Western blotting assay demonstrated that activation of Wnt/β-catenin pathway contributed to the oncogenic activity of HMGA1. Taken together, our data reveal that HMGA1 may function as an oncogene and modulate EC cell migration and invasion by activating Wnt/β-catenin pathway, implying that suppression of HMGA1 might be a potential therapeutic strategy for EC.
Collapse
Affiliation(s)
- Xiuxia Han
- Department of Gynecology and Obstetrics, The People's Hospital of Dongying, Dongying 257091, PR China.
| | - Yanhua Cao
- Department of Gynecology and Obstetrics, The People's Hospital of Dongying, Dongying 257091, PR China
| | - Kun Wang
- Department of Operating Rooms, The People's Hospital of Dongying, Dongying 257091, PR China
| | - Guiping Zhu
- Department of Gynecology and Obstetrics, Central Hospital of Shengli Oil Field, Dongying 257034, PR China
| |
Collapse
|
35
|
Spichiger C, Torres-Farfan C, Galdames HA, Mendez N, Alonso-Vazquez P, Richter HG. Gestation under chronic constant light leads to extensive gene expression changes in the fetal rat liver. Physiol Genomics 2015; 47:621-33. [DOI: 10.1152/physiolgenomics.00023.2015] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2015] [Accepted: 09/22/2015] [Indexed: 12/19/2022] Open
Abstract
Recent reports account for altered metabolism in adult offspring from pregnancy subjected to abnormal photoperiod, suggesting fetal programming of liver physiology. To generate a pipeline of subsequent mechanistic experiments addressing strong candidate genes, here we investigated the effects of constant gestational light on the fetal liver transcriptome. At 10 days of gestation, dams were randomized in two groups ( n = 7 each): constant light (LL) and normal photoperiod (12 h light/12 h dark; LD). At 18 days of gestation, RNA was isolated from the fetal liver and subjected to DNA microarray (Affymetrix platform for 28,000 genes). Selected differential mRNAs were validated by quantitative PCR (qPCR), while integrated transcriptional changes were analyzed with Ingenuity Pathway Analysis and other bioinformatics tools. Comparison of LL relative to LD fetal liver led to the following findings. Significant differential expression was found for 3,431 transcripts (1,960 upregulated and 1,471 downregulated), with 393 of them displaying ≥ 1.5-fold change. We validated 27 selected transcripts by qPCR, which displayed fold-change values highly correlated with microarray ( r2 = 0.91). Different markers of nonalcoholic fatty liver disease were either upregulated (e.g., Ndn and Pnpla3) or downregulated (e.g., Gnmt, Bhmt1/2, Sult1a1, Mpo, and Mat1a). Diverse pathways were altered, including hematopoiesis, coagulation cascade, complement system, and carbohydrate and lipid metabolism. The microRNAs 7a-1, 431, 146a, and 153 were upregulated, while the abundant hepatic miRNA 122 was downregulated. Constant gestational light induced extensive modification of the fetal liver transcriptome. A number of differentially expressed transcripts belong to fundamental functional pathways, potentially contributing to long-term liver disease.
Collapse
Affiliation(s)
- Carlos Spichiger
- Laboratory of Developmental Chronobiology, Institute of Anatomy, Histology and Pathology, Faculty of Medicine, Universidad Austral de Chile, Valdivia, Chile
| | - Claudia Torres-Farfan
- Laboratory of Developmental Chronobiology, Institute of Anatomy, Histology and Pathology, Faculty of Medicine, Universidad Austral de Chile, Valdivia, Chile
| | - Hugo A. Galdames
- Laboratory of Developmental Chronobiology, Institute of Anatomy, Histology and Pathology, Faculty of Medicine, Universidad Austral de Chile, Valdivia, Chile
| | - Natalia Mendez
- Laboratory of Developmental Chronobiology, Institute of Anatomy, Histology and Pathology, Faculty of Medicine, Universidad Austral de Chile, Valdivia, Chile
| | - Pamela Alonso-Vazquez
- Laboratory of Developmental Chronobiology, Institute of Anatomy, Histology and Pathology, Faculty of Medicine, Universidad Austral de Chile, Valdivia, Chile
| | - Hans G. Richter
- Laboratory of Developmental Chronobiology, Institute of Anatomy, Histology and Pathology, Faculty of Medicine, Universidad Austral de Chile, Valdivia, Chile
| |
Collapse
|
36
|
Hara K, Kadowaki T, Odawara M. Genes associated with diabetes: potential for novel therapeutic targets? Expert Opin Ther Targets 2015; 20:255-67. [DOI: 10.1517/14728222.2016.1098618] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/30/2023]
Affiliation(s)
- Kazuo Hara
- Division of Diabetes, Endocrinology and Metabolism, Tokyo Medical University, 6-7-1Nishi-shinjuku, Shinjuku-ku, Tokyo 160-0023, Japan
| | - Takashi Kadowaki
- Department of Diabetes and Metabolic Diseases, Graduate School of Medicine, University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo 113-8655, Japan
| | - Masato Odawara
- Division of Diabetes, Endocrinology and Metabolism, Tokyo Medical University, 6-7-1Nishi-shinjuku, Shinjuku-ku, Tokyo 160-0023, Japan
| |
Collapse
|
37
|
HMGA1 overexpression in adipose tissue impairs adipogenesis and prevents diet-induced obesity and insulin resistance. Sci Rep 2015; 5:14487. [PMID: 26411793 PMCID: PMC4585969 DOI: 10.1038/srep14487] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2015] [Accepted: 08/12/2015] [Indexed: 01/14/2023] Open
Abstract
High-Mobility-Group-A1 (HMGA1) proteins are non-histone proteins that regulate chromatin structure and gene expression during embryogenesis, tumourigenesis and immune responses. In vitro studies suggest that HMGA1 proteins may be required to regulate adipogenesis. To examine the role of HMGA1 in vivo, we generated transgenic mice overexpressing HMGA1 in adipose tissues. HMGA1 transgenic mice showed a marked reduction in white and brown adipose tissue mass that was associated with downregulation of genes involved in adipogenesis and concomitant upregulation of preadipocyte markers. Reduced adipogenesis and decreased fat mass were not associated with altered glucose homeostasis since HMGA1 transgenic mice fed a regular-chow diet exhibited normal glucose tolerance and insulin sensitivity. However, when fed a high-fat diet, overexpression of HMGA1 resulted in decreased body-weight gain, reduced fat mass, but improved insulin sensitivity and glucose tolerance. Although HMGA1 transgenic mice exhibited impaired glucose uptake in adipose tissue due to impaired adipogenesis, the increased glucose uptake observed in skeletal muscle may account for the improved glucose homeostasis. Our results indicate that HMGA1 plays an important function in the regulation of white and brown adipogenesis in vivo and suggests that impaired adipocyte differentiation and decreased fat mass is not always associated with impaired whole-body glucose homeostasis.
Collapse
|
38
|
Bianco A, Chiefari E, Nobile CGA, Foti D, Pavia M, Brunetti A. The Association between HMGA1 rs146052672 Variant and Type 2 Diabetes: A Transethnic Meta-Analysis. PLoS One 2015; 10:e0136077. [PMID: 26296198 PMCID: PMC4546600 DOI: 10.1371/journal.pone.0136077] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2015] [Accepted: 07/29/2015] [Indexed: 12/16/2022] Open
Abstract
The high-mobility group A1 (HMGA1) gene has been previously identified as a potential novel candidate gene for susceptibility to insulin resistance and type 2 diabetes (T2D) mellitus. For this reason, several studies have been conducted in recent years examining the association of the HMGA1 gene variant rs146052672 (also designated IVS5-13insC) with T2D. Because of non-univocal data and non-overlapping results among laboratories, we conducted the current meta-analysis with the aim to yield a more precise and reliable conclusion for this association. Using predetermined inclusion criteria, MEDLINE, PubMed, Web of Science, Scopus, Google Scholar and Embase were searched for all relevant available literature published until November 2014. Two of the authors independently evaluated the quality of the included studies and extracted the data. Values from the single studies were combined to determine the meta-analysis pooled estimates. Heterogeneity and publication bias were also examined. Among the articles reviewed, five studies (for a total of 13,789 cases and 13,460 controls) met the predetermined criteria for inclusion in this meta-analysis. The combined adjusted odds ratio estimates revealed that the rs146052672 variant genotype had an overall statistically significant effect on increasing the risk of development of T2D. As most of the study subjects were Caucasian, further studies are needed to establish whether the association of this variant with an increased risk of T2D is generalizable to other populations. Also, in the light of this result, it would appear to be highly desirable that further in-depth investigations should be undertaken to elucidate the biological significance of the HMGA1 rs146052672 variant.
Collapse
Affiliation(s)
- Aida Bianco
- Department of Health Sciences, University “Magna Græcia” of Catanzaro, Catanzaro, Italy
| | - Eusebio Chiefari
- Department of Health Sciences, University “Magna Græcia” of Catanzaro, Catanzaro, Italy
| | - Carmelo G. A. Nobile
- Department of Health Sciences, University “Magna Græcia” of Catanzaro, Catanzaro, Italy
| | - Daniela Foti
- Department of Health Sciences, University “Magna Græcia” of Catanzaro, Catanzaro, Italy
| | - Maria Pavia
- Department of Health Sciences, University “Magna Græcia” of Catanzaro, Catanzaro, Italy
| | - Antonio Brunetti
- Department of Health Sciences, University “Magna Græcia” of Catanzaro, Catanzaro, Italy
| |
Collapse
|
39
|
Chiefari E, Capula C, Vero A, Oliverio R, Puccio L, Liguori R, Pullano V, Greco M, Foti D, Tirinato D, Vero R, Brunetti A. Add-On Treatment with Liraglutide Improves Glycemic Control in Patients with Type 2 Diabetes on Metformin Therapy. Diabetes Technol Ther 2015; 17:468-74. [PMID: 25844858 DOI: 10.1089/dia.2014.0412] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
BACKGROUND Liraglutide is a glucagon-like peptide-1 receptor analog recently approved for the treatment of type 2 diabetes mellitus (T2DM). We aimed to assess the efficacy and safety of liraglutide versus glimepiride, as adjunct treatments to metformin, in achieving glycemic control in Italian patients with T2DM uncontrolled by metformin alone. SUBJECTS AND METHODS One hundred seventy-nine diabetes patients treated with metformin plus liraglutide (1.8 mg) or glimepiride (4 mg) were retrospectively assessed at baseline, during, and after 18 months of continuous therapy. RESULTS Treatment with liraglutide resulted in mean decreases in hemoglobin A1c (HbA1c) of -1.4%, when compared with glimepiride (-0.4%) (P < 0.001), and was followed by a significant reduction (P < 0.001) in fasting plasma glucose. Variations in HbA1c occurred independently from weight loss, which was significantly reduced (P < 0.001) in liraglutide-treated patients. The percentage of subjects reaching HbA1c levels below 7% or ≤ 6.5% was significantly different between the two treated groups (P < 0.001). Treatment with liraglutide reduced waist circumference (WC) (P < 0.001) and decreased both systolic and diastolic blood pressure (BP) (P < 0.001). It is interesting that the study also showed the impact of female gender in predicting a better glycemic response to liraglutide (P = 0.028). CONCLUSIONS Liraglutide was more effective than glimepiride in reducing HbA1c levels in treated patients with T2DM. This was evident in both genders, but particularly in women. Furthermore, liraglutide reduced body weight, WC, and BP, which are critical risk factors for cardiovascular disease.
Collapse
Affiliation(s)
- Eusebio Chiefari
- 1 Department of Health Sciences, University "Magna Græcia" of Catanzaro , Catanzaro, Italy
| | - Carmelo Capula
- 2 Complex Operative Structure of Endocrinology-Diabetology, Pugliese-Ciaccio Hospital , Catanzaro, Italy
| | - Ada Vero
- 2 Complex Operative Structure of Endocrinology-Diabetology, Pugliese-Ciaccio Hospital , Catanzaro, Italy
| | - Rosa Oliverio
- 1 Department of Health Sciences, University "Magna Græcia" of Catanzaro , Catanzaro, Italy
| | - Luigi Puccio
- 2 Complex Operative Structure of Endocrinology-Diabetology, Pugliese-Ciaccio Hospital , Catanzaro, Italy
| | - Rossella Liguori
- 1 Department of Health Sciences, University "Magna Græcia" of Catanzaro , Catanzaro, Italy
| | - Vittorio Pullano
- 2 Complex Operative Structure of Endocrinology-Diabetology, Pugliese-Ciaccio Hospital , Catanzaro, Italy
| | - Manfredi Greco
- 3 Department of Experimental and Clinical Medicine, University "Magna Græcia" of Catanzaro , Catanzaro, Italy
| | - Daniela Foti
- 1 Department of Health Sciences, University "Magna Græcia" of Catanzaro , Catanzaro, Italy
| | | | - Raffaella Vero
- 2 Complex Operative Structure of Endocrinology-Diabetology, Pugliese-Ciaccio Hospital , Catanzaro, Italy
| | - Antonio Brunetti
- 1 Department of Health Sciences, University "Magna Græcia" of Catanzaro , Catanzaro, Italy
| |
Collapse
|
40
|
Arnoldo L, Sgarra R, Chiefari E, Iiritano S, Arcidiacono B, Pegoraro S, Pellarin I, Brunetti A, Manfioletti G. A novel mechanism of post-translational modulation of HMGA functions by the histone chaperone nucleophosmin. Sci Rep 2015; 5:8552. [PMID: 25711412 PMCID: PMC4339810 DOI: 10.1038/srep08552] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2014] [Accepted: 01/12/2015] [Indexed: 02/07/2023] Open
Abstract
High Mobility Group A are non-histone nuclear proteins that regulate chromatin plasticity and accessibility, playing an important role both in physiology and pathology. Their activity is controlled by transcriptional, post-transcriptional, and post-translational mechanisms. In this study we provide evidence for a novel modulatory mechanism for HMGA functions. We show that HMGAs are complexed in vivo with the histone chaperone nucleophosmin (NPM1), that this interaction requires the histone-binding domain of NPM1, and that NPM1 modulates both DNA-binding affinity and specificity of HMGAs. By focusing on two human genes whose expression is directly regulated by HMGA1, the Insulin receptor (INSR) and the Insulin-like growth factor-binding protein 1 (IGFBP1) genes, we demonstrated that occupancy of their promoters by HMGA1 was NPM1-dependent, reflecting a mechanism in which the activity of these cis-regulatory elements is directly modulated by NPM1 leading to changes in gene expression. HMGAs need short stretches of AT-rich nucleosome-free regions to bind to DNA. Therefore, many putative HMGA binding sites are present within the genome. Our findings indicate that NPM1, by exerting a chaperoning activity towards HMGAs, may act as a master regulator in the control of DNA occupancy by these proteins and hence in HMGA-mediated gene expression.
Collapse
Affiliation(s)
- Laura Arnoldo
- Department of Life Sciences, University of Trieste, Trieste, 34127, Italy
| | - Riccardo Sgarra
- Department of Life Sciences, University of Trieste, Trieste, 34127, Italy
| | - Eusebio Chiefari
- Department of Health Sciences, University "Magna Græcia" of Catanzaro, Catanzaro, 88100, Italy
| | - Stefania Iiritano
- Department of Health Sciences, University "Magna Græcia" of Catanzaro, Catanzaro, 88100, Italy
| | - Biagio Arcidiacono
- Department of Health Sciences, University "Magna Græcia" of Catanzaro, Catanzaro, 88100, Italy
| | - Silvia Pegoraro
- Department of Life Sciences, University of Trieste, Trieste, 34127, Italy
| | - Ilenia Pellarin
- Department of Life Sciences, University of Trieste, Trieste, 34127, Italy
| | - Antonio Brunetti
- Department of Health Sciences, University "Magna Græcia" of Catanzaro, Catanzaro, 88100, Italy
| | | |
Collapse
|
41
|
Capula C, Chiefari E, Vero A, Foti DP, Brunetti A, Vero R. Prevalence and predictors of postpartum glucose intolerance in Italian women with gestational diabetes mellitus. Diabetes Res Clin Pract 2014; 105:223-30. [PMID: 24931701 DOI: 10.1016/j.diabres.2014.05.008] [Citation(s) in RCA: 59] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/24/2013] [Revised: 04/25/2014] [Accepted: 05/18/2014] [Indexed: 02/07/2023]
Abstract
AIMS To determine the prevalence of both prediabetes and type 2 diabetes mellitus (T2DM) by postpartum oral glucose tolerance test (ppOGTT) in Italian women diagnosed with gestational diabetes mellitus (GDM), and identify antepartum predictors of glucose intolerance. METHODS Retrospective study of 454 Caucasian women that underwent a 75g OGTT between 6 and 12 weeks postpartum in Calabria (Southern Italy) between 2004 and 2012. Prediabetes and T2DM were diagnosed according to the American Diabetes Association (ADA) criteria. Data were examined by univariate analysis and multiple regression analysis. RESULTS 290 women (63.9%) were normal, 146 (32.1%) had prediabetes (85 impaired fasting glycemia; 61 impaired glucose tolerance), and 18 (4.0%) had T2DM. Of the continuous variables, pre-pregnancy body mass index (BMI), age at pregnancy, fasting plasma glucose (FPG) at gravid OGTT, and week at diagnosis of GDM were associated with prediabetes and T2DM, whereas the parity was associated with T2DM only. For categorical traits, pre-pregnancy BMI ≥ 25 and previous diagnosis of polycystic ovary syndrome (PCOS) emerged as the strongest predictors of prediabetes whereas the strongest predictors of T2DM were FPG ≥ 100 mg/dl (5.6 mmol/l) at GDM diagnosis and pre-pregnancy BMI ≥ 25. Moreover, FPG at GDM screening was a good predictor of T2DM after receiver-operating-characteristic analysis. CONCLUSIONS Our findings confirm the high prevalence of glucose intolerance in the early postpartum period in women with previous GDM. PCOS emerges as a new strong antepartum predictor of prediabetes.
Collapse
Affiliation(s)
- Carmelo Capula
- Complex Operative Structure of Endocrinology-Diabetology, Pugliese-Ciaccio Hospital, 88100 Catanzaro, Italy
| | - Eusebio Chiefari
- Department of Health Sciences, University "Magna Græcia" of Catanzaro, 88100 Catanzaro, Italy
| | - Anna Vero
- Complex Operative Structure of Endocrinology-Diabetology, Pugliese-Ciaccio Hospital, 88100 Catanzaro, Italy
| | - Daniela P Foti
- Department of Health Sciences, University "Magna Græcia" of Catanzaro, 88100 Catanzaro, Italy
| | - Antonio Brunetti
- Department of Health Sciences, University "Magna Græcia" of Catanzaro, 88100 Catanzaro, Italy.
| | - Raffaella Vero
- Complex Operative Structure of Endocrinology-Diabetology, Pugliese-Ciaccio Hospital, 88100 Catanzaro, Italy
| |
Collapse
|
42
|
Zhang J, Zhao Y, Xu C, Hong Y, Lu H, Wu J, Chen Y. Association between serum free fatty acid levels and nonalcoholic fatty liver disease: a cross-sectional study. Sci Rep 2014; 4:5832. [PMID: 25060337 PMCID: PMC5376058 DOI: 10.1038/srep05832] [Citation(s) in RCA: 136] [Impact Index Per Article: 12.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2014] [Accepted: 07/07/2014] [Indexed: 02/06/2023] Open
Abstract
High serum free fatty acid (FFA) levels are associated with metabolic syndrome (MS). This study aimed to assess the association of fasting serum FFAs with nonalcoholic fatty liver disease (NAFLD) in a Chinese population. A total of 840 subjects fulfilled the diagnostic criteria of NAFLD and 331 healthy control participants were enrolled in this cross-sectional study. Fasting serum FFA levels and other clinical and laboratory parameters were measured. NAFLD patients had significantly higher serum FFA levels than controls (P < 0.001). Serum FFA levels were significantly and positively correlated with parameters of MS, inflammation indexes, and markers of hepatocellular damage. Elevated serum FFA levels were found in NAFLD subjects with individual components of MS (obesity, hypertriglyceridaemia, and hyperglycaemia). Stepwise regression showed that serum FFA levels were an independent factor predicting advanced fibrosis (FIB-4 ≥ 1.3) in NAFLD patients. Serum FFA levels correlated with NAFLD and could be used as an indicator for predicting advanced fibrosis in NAFLD patients.
Collapse
Affiliation(s)
- Juanwen Zhang
- Department of Laboratory Medicine, The First Affiliated Hospital, College of Medicine, Zhejiang University, 79 Qingchun Road, Hangzhou 310003, China
- These authors contributed equally to this work
| | - Ying Zhao
- Department of Laboratory Medicine, The First Affiliated Hospital, College of Medicine, Zhejiang University, 79 Qingchun Road, Hangzhou 310003, China
- These authors contributed equally to this work
| | - Chengfu Xu
- Department of Gastroenterology, The First Affiliated Hospital, College of Medicine, Zhejiang University, 79 Qingchun Road, Hangzhou 310003, China
| | - Yani Hong
- Department of Laboratory Medicine, The First Affiliated Hospital, College of Medicine, Zhejiang University, 79 Qingchun Road, Hangzhou 310003, China
| | - Huanle Lu
- Department of Laboratory Medicine, The First Affiliated Hospital, College of Medicine, Zhejiang University, 79 Qingchun Road, Hangzhou 310003, China
| | - Jianping Wu
- Department of Laboratory Medicine, The First Affiliated Hospital, College of Medicine, Zhejiang University, 79 Qingchun Road, Hangzhou 310003, China
| | - Yu Chen
- Department of Laboratory Medicine, The First Affiliated Hospital, College of Medicine, Zhejiang University, 79 Qingchun Road, Hangzhou 310003, China
| |
Collapse
|
43
|
Brunetti A, Chiefari E, Foti D. Recent advances in the molecular genetics of type 2 diabetes mellitus. World J Diabetes 2014; 5:128-140. [PMID: 24748926 PMCID: PMC3990314 DOI: 10.4239/wjd.v5.i2.128] [Citation(s) in RCA: 86] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/19/2013] [Revised: 12/28/2013] [Accepted: 01/20/2014] [Indexed: 02/05/2023] Open
Abstract
Type 2 diabetes mellitus (T2DM) is a complex disease in which both genetic and environmental factors interact in determining impaired β-cell insulin secretion and peripheral insulin resistance. Insulin resistance in muscle, liver and fat is a prominent feature of most patients with T2DM and obesity, resulting in a reduced response of these tissues to insulin. Considerable evidence has been accumulated to indicate that heredity is a major determinant of insulin resistance and T2DM. It is believed that, among individuals destined to develop T2DM, hyperinsulinemia is the mechanism by which the pancreatic β-cell initially compensates for deteriorating peripheral insulin sensitivity, thus ensuring normal glucose tolerance. Most of these people will develop T2DM when β-cells fail to compensate. Despite the progress achieved in this field in recent years, the genetic causes of insulin resistance and T2DM remain elusive. Candidate gene association, linkage and genome-wide association studies have highlighted the role of genetic factors in the development of T2DM. Using these strategies, a large number of variants have been identified in many of these genes, most of which may influence both hepatic and peripheral insulin resistance, adipogenesis and β-cell mass and function. Recently, a new gene has been identified by our research group, the HMGA1 gene, whose loss of function can greatly raise the risk of developing T2DM in humans and mice. Functional genetic variants of the HMGA1 gene have been associated with insulin resistance syndromes among white Europeans, Chinese individuals and Americans of Hispanic ancestry. These findings may represent new ways to improve or even prevent T2DM.
Collapse
|
44
|
Arcidiacono B, Iiritano S, Chiefari E, Brunetti FS, Gu G, Foti DP, Brunetti A. Cooperation between HMGA1, PDX-1, and MafA is Essential for Glucose-Induced Insulin Transcription in Pancreatic Beta Cells. Front Endocrinol (Lausanne) 2014; 5:237. [PMID: 25628604 PMCID: PMC4292585 DOI: 10.3389/fendo.2014.00237] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/23/2014] [Accepted: 12/18/2014] [Indexed: 01/03/2023] Open
Abstract
The high-mobility group AT-hook 1 (HMGA1) protein is a nuclear architectural factor that can organize chromatin structures. It regulates gene expression by controlling the formation of stereospecific multiprotein complexes called "enhanceosomes" on the AT-rich regions of target gene promoters. Previously, we reported that defects in HMGA1 caused decreased insulin receptor expression and increased susceptibility to type 2 diabetes mellitus in humans and mice. Interestingly, mice with disrupted HMGA1 gene had significantly smaller islets and decreased insulin content in their pancreata, suggesting that HMGA1 may have a direct role in insulin transcription and secretion. Herein, we investigate the regulatory roles of HMGA1 in insulin transcription. We provide evidence that HMGA1 physically interacts with PDX-1 and MafA, two critical transcription factors for insulin gene expression and beta-cell function, both in vitro and in vivo. We then show that the overexpression of HMGA1 significantly improves the transactivating activity of PDX-1 and MafA on human and mouse insulin promoters, while HMGA1 knockdown considerably decreased this transactivating activity. Lastly, we demonstrate that high glucose stimulus significantly increases the binding of HMGA1 to the insulin (INS) gene promoter, suggesting that HMGA1 may act as a glucose-sensitive element controlling the transcription of the INS gene. Together, our findings provide evidence that HMGA1, by regulating PDX-1- and MafA-induced transactivation of the INS gene promoter, plays a critical role in pancreatic beta-cell function and insulin production.
Collapse
Affiliation(s)
- Biagio Arcidiacono
- Department of Health Sciences, University “Magna Græcia” of Catanzaro, Catanzaro, Italy
| | - Stefania Iiritano
- Department of Health Sciences, University “Magna Græcia” of Catanzaro, Catanzaro, Italy
| | - Eusebio Chiefari
- Department of Health Sciences, University “Magna Græcia” of Catanzaro, Catanzaro, Italy
| | - Francesco S. Brunetti
- Department of Medical and Surgical Sciences, University “Magna Græcia” of Catanzaro, Catanzaro, Italy
| | - Guoqiang Gu
- Department of Cell and Developmental Biology, Center of Stem Cell Biology, Vanderbilt Medical Center, Nashville, TN, USA
| | - Daniela Patrizia Foti
- Department of Health Sciences, University “Magna Græcia” of Catanzaro, Catanzaro, Italy
| | - Antonio Brunetti
- Department of Health Sciences, University “Magna Græcia” of Catanzaro, Catanzaro, Italy
- *Correspondence: Antonio Brunetti, Department of Health Sciences, University “Magna Græcia” of Catanzaro, Viale Europa (Località Germaneto), Catanzaro 88100, Italy e-mail:
| |
Collapse
|
45
|
Chiefari E, Arcidiacono B, Possidente K, Iiritano S, Ventura V, Pandolfo R, Brunetti FS, Greco M, Foti D, Brunetti A. Transcriptional regulation of the HMGA1 gene by octamer-binding proteins Oct-1 and Oct-2. PLoS One 2013; 8:e83969. [PMID: 24367622 PMCID: PMC3867479 DOI: 10.1371/journal.pone.0083969] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2013] [Accepted: 11/19/2013] [Indexed: 01/20/2023] Open
Abstract
The High-Mobility Group AT-Hook 1 (HMGA1) protein is an architectural transcription factor that binds to AT-rich sequences in the promoter region of DNA and functions as a specific cofactor for gene activation. Previously, we demonstrated that HMGA1 is a key regulator of the insulin receptor (INSR) gene and an important downstream target of the INSR signaling cascade. Moreover, from a pathogenic point of view, overexpression of HMGA1 has been associated with human cancer, whereas functional variants of the HMGA1 gene have been recently linked to type 2 diabetes mellitus and metabolic syndrome. However, despite of this biological and pathological relevance, the mechanisms that control HMGA1 gene expression remain unknown. In this study, to define the molecular mechanism(s) that regulate HMGA1 gene expression, the HMGA1 gene promoter was investigated by transient transfection of different cell lines, either before or after DNA and siRNA cotransfections. An octamer motif was identified as an important element of transcriptional regulation of this gene, the interaction of which with the octamer transcription factors Oct-1 and Oct-2 is crucial in modulating HMGA1 gene and protein expression. Additionally, we demonstrate that HMGA1 binds its own promoter and contributes to its transactivation by Oct-2 (but not Oct-1), supporting its role in an auto-regulatory circuit. Overall, our results provide insight into the transcriptional regulation of the HMGA1 gene, revealing a differential control exerted by both Oct-1 and Oct-2. Furthermore, they consistently support the hypothesis that a putative defect in Oct-1 and/or Oct-2, by affecting HMGA1 expression, may cause INSR dysfunction, leading to defects of the INSR signaling pathway.
Collapse
Affiliation(s)
- Eusebio Chiefari
- Department of Health Sciences, University “Magna Græcia” of Catanzaro, Viale Europa (Loc. Germaneto), Catanzaro, Italy
| | - Biagio Arcidiacono
- Department of Health Sciences, University “Magna Græcia” of Catanzaro, Viale Europa (Loc. Germaneto), Catanzaro, Italy
| | - Katiuscia Possidente
- Department of Health Sciences, University “Magna Græcia” of Catanzaro, Viale Europa (Loc. Germaneto), Catanzaro, Italy
| | - Stefania Iiritano
- Department of Health Sciences, University “Magna Græcia” of Catanzaro, Viale Europa (Loc. Germaneto), Catanzaro, Italy
| | - Valeria Ventura
- Department of Health Sciences, University “Magna Græcia” of Catanzaro, Viale Europa (Loc. Germaneto), Catanzaro, Italy
| | - Rosantony Pandolfo
- Department of Health Sciences, University “Magna Græcia” of Catanzaro, Viale Europa (Loc. Germaneto), Catanzaro, Italy
| | - Francesco Saverio Brunetti
- Department of Medical and Surgical Sciences, University “Magna Græcia” of Catanzaro, Viale Europa (Loc. Germaneto), Catanzaro, Italy
| | - Manfredi Greco
- Department of Experimental and Clinical Medicine, University “Magna Græcia” of Catanzaro, Viale Europa (Loc. Germaneto), Catanzaro, Italy
| | - Daniela Foti
- Department of Health Sciences, University “Magna Græcia” of Catanzaro, Viale Europa (Loc. Germaneto), Catanzaro, Italy
| | - Antonio Brunetti
- Department of Health Sciences, University “Magna Græcia” of Catanzaro, Viale Europa (Loc. Germaneto), Catanzaro, Italy
| |
Collapse
|
46
|
Pullinger CR, Goldfine ID, Tanyolaç S, Movsesyan I, Faynboym M, Durlach V, Chiefari E, Foti DP, Frost PH, Malloy MJ, Brunetti A, Kane JP. Evidence that an HMGA1 gene variant associates with type 2 diabetes, body mass index, and high-density lipoprotein cholesterol in a Hispanic-American population. Metab Syndr Relat Disord 2013; 12:25-30. [PMID: 24148075 DOI: 10.1089/met.2013.0086] [Citation(s) in RCA: 44] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
BACKGROUND High-mobility group AT-hook 1 (HMGA1) is an important regulator of the insulin receptor gene. We have previously shown in three populations of white European ancestry that the HMGA1 gene variant rs146052672 (also designated IVS5-13insC) is associated with type 2 diabetes mellitus (T2DM). The aim of this study was to measure the frequency of this variant and to determine the degree of the association with T2DM and other features of the metabolic syndrome in a replication cohort of Hispanic Americans. METHODS This was a retrospective cohort study of well-characterized Hispanic-American participants analyzed in the Genomic Resource in Atherosclerosis (GRA) (Cardiovascular Research Institute, University of California, San Francisco). A total of 1144 individuals were studied, 320 of whom had T2DM. We examined associations of the rs146052672 SNP with T2DM, plasma lipids, lipoproteins, and body mass index (BMI). RESULTS In this Hispanic-American cohort, the HMGA1 rs146052672 minor allele (C-insertion) frequency (MAF) was 21.4% with a carrier frequency of 37.4%, considerably higher than we previously observed among GRA white Europeans (MAF 3.1%). The prevalence of the IVS5-13insC variant was significantly higher in those with T2DM compared to controls [42.2% vs. 35.5%; odds ratio (OR) 1.44 95% confidence interval (CI) 1.09-1.90, P=0.011). The variant was also associated with BMI (positively, P=0.045) and plasma high-density lipoprotein cholesterol (HDL-C) (negatively, P=0.047). CONCLUSIONS As we saw previously among white Europeans, a functional HMGA1 variant was associated with T2DM in individuals of Hispanic-American ethnicity and was present at a much higher frequency.
Collapse
Affiliation(s)
- Clive R Pullinger
- 1 Cardiovascular Research Institute and Department of Physiological Nursing, University of California , San Francisco, California
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
47
|
Zhang R, Wang J, Yang R, Sun J, Chen R, Luo H, Liu D, Cai D. Effects of Pro12Ala polymorphism in peroxisome proliferator-activated receptor-γ2 gene on metabolic syndrome risk: a meta-analysis. Gene 2013; 535:79-87. [PMID: 24012868 DOI: 10.1016/j.gene.2013.07.087] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2013] [Revised: 07/10/2013] [Accepted: 07/23/2013] [Indexed: 12/22/2022]
Abstract
BACKGROUND Associations between peroxisome proliferator-activated receptor γ2 (PPARγ2) gene polymorphism and metabolic syndrome risk remained controversial and ambiguous. Thus, we performed a meta-analysis to assess the association between Pro12Ala polymorphism in PPARγ2 gene and metabolic syndrome susceptibility. METHODS An electronic literature search was conducted on Medline, OVID, Cochrane Library database, and the China National Knowledge Internet up to March 2013. Odds ratios (ORs) with 95% confidence intervals (CIs) were used to calculate the strength of association in the fixed or random effects model. RESULTS Ten studies involving a total of 4456 cases and 10343 controls were included in this meta-analysis. No statistical evidence of association was found between Pro12Ala polymorphism and metabolic syndrome risk in all genetic models (homozygote model: OR=0.83, 95% CI=0.62-1.12; heterozygote model: OR=1.04, 95% CI=0.94-1.14; dominant model: OR=1.02, 95% CI=0.93-1.12; recessive model: OR=0.83, 95% CI=0.62-1.11). No statistical evidence of significant association was observed when stratified by ethnicity, definition of metabolic syndrome, source of control groups and quality score of the selected articles. All in all, the results did not support a major role of the Pro12Ala variant of the PPARγ2 gene in metabolic syndrome risk. CONCLUSIONS This meta-analysis suggested that the effect of Pro12Ala polymorphism in PPARγ2 gene may not be related to metabolic syndrome as an entity. However, Pro12Ala may affect the single component of metabolic syndrome. A large, well designed study is required to more adequately assess the role for Pro12Ala polymorphism on metabolic syndrome.
Collapse
Affiliation(s)
- Ruyi Zhang
- Department of Endocrinology, Southern Medical University, Zhujiang Hospital, 253# Industry Road, 510282 Guangzhou, Guangdong, China.
| | - Jiao Wang
- Department of Endocrinology, Southern Medical University, Zhujiang Hospital, 253# Industry Road, 510282 Guangzhou, Guangdong, China.
| | - Rui Yang
- Department of Endocrinology, Southern Medical University, Zhujiang Hospital, 253# Industry Road, 510282 Guangzhou, Guangdong, China.
| | - Jia Sun
- Department of Endocrinology, Southern Medical University, Zhujiang Hospital, 253# Industry Road, 510282 Guangzhou, Guangdong, China.
| | - Rongping Chen
- Department of Endocrinology, Southern Medical University, Zhujiang Hospital, 253# Industry Road, 510282 Guangzhou, Guangdong, China.
| | - Haizhao Luo
- Department of Endocrinology, Southern Medical University, Zhujiang Hospital, 253# Industry Road, 510282 Guangzhou, Guangdong, China.
| | - Duan Liu
- Department of Endocrinology, Southern Medical University, Zhujiang Hospital, 253# Industry Road, 510282 Guangzhou, Guangdong, China.
| | - Dehong Cai
- Department of Endocrinology, Southern Medical University, Zhujiang Hospital, 253# Industry Road, 510282 Guangzhou, Guangdong, China.
| |
Collapse
|