1
|
Pagkali A, Makris A, Brofidi K, Agouridis AP, Filippatos TD. Pathophysiological Mechanisms and Clinical Associations of Non-Alcoholic Fatty Pancreas Disease. Diabetes Metab Syndr Obes 2024; 17:283-294. [PMID: 38283640 PMCID: PMC10813232 DOI: 10.2147/dmso.s397643] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/09/2023] [Accepted: 12/29/2023] [Indexed: 01/30/2024] Open
Abstract
Non-Alcoholic Fatty Pancreas disease (NAFPD), characterized by fat accumulation in pancreatic tissue, is an emerging clinical entity. However, the clinical associations, the underlying molecular drivers, and the pathophysiological mechanisms of NAFPD have not yet been characterized in detail. The NAFPD spectrum not only includes infiltration and accumulation of fat within and between pancreatic cells but also involves several inflammatory processes, dysregulation of physiological metabolic pathways, and hormonal defects. A deeper understanding of the underlying molecular mechanisms is key to correlate NAFPD with clinical entities including non-alcoholic fatty liver disease, metabolic syndrome, diabetes mellitus, atherosclerosis, as well as pancreatic cancer and pancreatitis. The aim of this review is to examine the pathophysiological mechanisms of NAFPD and to assess the possible causative/predictive risk factors of NAFPD-related clinical syndromes.
Collapse
Affiliation(s)
- Antonia Pagkali
- School of Medicine, National and Kapodistrian University of Athens, Athens, Greece
| | - Anastasios Makris
- School of Medicine, National and Kapodistrian University of Athens, Athens, Greece
| | - Kalliopi Brofidi
- Department of Internal Medicine, School of Medicine, University of Crete, Heraklion, Greece
| | - Aris P Agouridis
- School of Medicine, European University Cyprus, Nicosia, Cyprus
- Department of Internal Medicine, German Oncology Center, Limassol, Cyprus
| | | |
Collapse
|
2
|
Mancinetti F, Xenos D, De Fano M, Mazzieri A, Porcellati F, Boccardi V, Mecocci P. Diabetes-Alzheimer's connection in older age: SGLT2 inhibitors as promising modulators of disease pathways. Ageing Res Rev 2023; 90:102018. [PMID: 37481164 DOI: 10.1016/j.arr.2023.102018] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2023] [Revised: 07/16/2023] [Accepted: 07/18/2023] [Indexed: 07/24/2023]
Abstract
Late-onset Alzheimer's disease (LOAD) is the most frequent cause of dementia in older persons. Subjects affected by type 2 diabetes mellitus (T2DM) are at higher risk of vascular disease, cognitive decline, and dementia. LOAD has many characteristics shared with impaired insulin signaling pathways, and substantial evidence has demonstrated a pivotal role in dysregulated glucose metabolism in its pathogenesis. Recent studies have shown that some anti-diabetic drugs, other than regulating the metabolism of peripheral tissues, can also modulate the brain's metabolism, reduce inflammation, and have a direct neuroprotective effect. Sodium-glucose cotransporter-2 inhibitors (SGLT2i) are a newer class with many pleiotropic effects that may have strong neuroprotective potential. After a summary of the principal "anti-diabetic" drugs acting as suitable candidates in treating LOAD, this narrative review explored the potential role of SGLT2i on cognition from pre-clinical to clinical studies.
Collapse
Affiliation(s)
- Francesca Mancinetti
- Institute of Gerontology and Geriatrics, Department of Medicine and Surgery, University of Perugia, Italy
| | - Dionysios Xenos
- Institute of Gerontology and Geriatrics, Department of Medicine and Surgery, University of Perugia, Italy
| | - Michelantonio De Fano
- Institute of Internal Medicine, Endocrinology and Metabolism, Department of Medicine and Surgery, University of Perugia, Italy
| | - Alessio Mazzieri
- Institute of Internal Medicine, Endocrinology and Metabolism, Department of Medicine and Surgery, University of Perugia, Italy
| | - Francesca Porcellati
- Institute of Internal Medicine, Endocrinology and Metabolism, Department of Medicine and Surgery, University of Perugia, Italy
| | - Virginia Boccardi
- Institute of Gerontology and Geriatrics, Department of Medicine and Surgery, University of Perugia, Italy.
| | - Patrizia Mecocci
- Institute of Gerontology and Geriatrics, Department of Medicine and Surgery, University of Perugia, Italy; Division of Clinical Geriatrics, NVS Department, Karolinska Institutet, Stockholm, Sweden
| |
Collapse
|
3
|
Hearing loss drug discovery and medicinal chemistry: Current status, challenges, and opportunities. PROGRESS IN MEDICINAL CHEMISTRY 2022; 61:1-91. [PMID: 35753714 DOI: 10.1016/bs.pmch.2022.05.001] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
Abstract
Hearing loss is a severe high unmet need condition affecting more than 1.5 billion people globally. There are no licensed medicines for the prevention, treatment or restoration of hearing. Prosthetic devices, such as hearing aids and cochlear implants, do not restore natural hearing and users struggle with speech in the presence of background noise. Hearing loss drug discovery is immature, and small molecule approaches include repurposing existing drugs, combination therapeutics, late-stage discovery optimisation of known chemotypes for identified molecular targets of interest, phenotypic tissue screening and high-throughput cell-based screening. Hearing loss drug discovery requires the integration of specialist therapeutic area biology and otology clinical expertise. Small molecule drug discovery projects in the global clinical portfolio for hearing loss are here collated and reviewed. An overview is provided of human hearing, inner ear anatomy, inner ear delivery, types of hearing loss and hearing measurement. Small molecule experimental drugs in clinical development for hearing loss are reviewed, including their underpinning biology, discovery strategy and activities, medicinal chemistry, calculated physicochemical properties, pharmacokinetics and clinical trial status. SwissADME BOILED-Egg permeability modelling is applied to the molecules reviewed, and these results are considered. Non-small molecule hearing loss assets in clinical development are briefly noted in this review. Future opportunities in hearing loss drug discovery for human genomics and targeted protein degradation are highlighted.
Collapse
|
4
|
Katsiki N, Dimitriadis GD, Mikhailidis DP. Serum Uric Acid and Diabetes: From Pathophysiology to Cardiovascular Disease. Curr Pharm Des 2021; 27:1941-1951. [PMID: 33397230 DOI: 10.2174/1381612827666210104124320] [Citation(s) in RCA: 42] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2020] [Accepted: 11/13/2020] [Indexed: 11/22/2022]
Abstract
Hyperuricemia, has been traditionally related to nephrolithiasis and gout. However, it has also been associated with the development of type 2 diabetes mellitus (T2DM) and cardiometabolic and cardiovascular diseases. Pathophysiologically, elevated serum uric acid (SUA) levels may be associated with abnormal lipid and glucose metabolism. In this narrative review, we consider the associations between hyperuricemia, hyperglycemia, atherosclerosis and thrombosis. Furthermore, we comment on the available evidence linking elevated SUA levels with the incidence and outcomes of coronary heart disease, stroke, peripheral artery disease and non-alcoholic fatty liver in subjects with T2DM. The effects of antidiabetic drugs (e.g. metformin, pioglitazone, sulfonylureas, dipeptidyl peptidase 4 inhibitors, glucagon-like peptide-1 receptor agonists, sodium-glucose cotransporter 2 inhibitors and insulin) on SUA concentrations are also reviewed.
Collapse
Affiliation(s)
- Niki Katsiki
- First Department of Internal Medicine, Diabetes Center, Division of Endocrinology and Metabolism, AHEPA University Hospital, Thessaloniki, Greece
| | | | - Dimitri P Mikhailidis
- Department of Clinical Biochemistry, Royal Free Hospital campus, University College London Medical School, University College London (UCL), London NW3 2QG, United Kingdom
| |
Collapse
|
5
|
Zhu J, Sun L, Yang J, Fan J, Tse LA, Li Y. Genetic Predisposition to Type 2 Diabetes and Insulin Levels Is Positively Associated With Serum Urate Levels. J Clin Endocrinol Metab 2021; 106:e2547-e2556. [PMID: 33770169 DOI: 10.1210/clinem/dgab200] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/03/2021] [Indexed: 12/26/2022]
Abstract
PURPOSE Previous epidemiological evidence showed that type 2 diabetes (T2D) is related with gout. However, the causality and the direction of this association are still not definitely elucidated. We investigated bidirectional associations of T2D and glycemic traits with serum urate concentrations and gout using a Mendelian randomization approach. METHODS Summary statistics from the large-scale genomewide association studies conducted for T2D (Ncase = 62 892, Ncontrol = 596 424), fasting glucose (N = 133 010), fasting insulin (N = 133 010), hemoglobin A1c (N = 123 665), homeostasis model assessment of insulin resistance (N = 46 186), urate (N = 110 347), and gout (Ncase = 2115, Ncontrol = 67 259) among participants of European ancestry were analyzed. For each trait of interest, independent genomewide significant (P < 5 × 10-8) single nucleotide polymorphisms were selected as instrumental variables. The inverse-variance weighted method was used for the primary analyses. RESULTS Genetic predisposition to higher risk of T2D [beta = 0.042; 95% confidence interval (CI) = 0.016-0.068; P = 0.002] and higher levels of fasting insulin (beta = 0.756; 95% CI = 0.408-1.102; P = 1.96e-05) were significantly associated with increased serum urate concentrations. Moreover, we found suggestively significant evidence supporting a causal role of fasting insulin on risk of developing gout (odds ratio = 3.06; 95% CI = 0.88-10.61; P = 0.078). In the reverse direction analysis, genetic predisposition to both urate and gout were not associated with T2D or any of 4 glycemic traits being investigated. CONCLUSIONS This study provides supportive evidence on causal associations of T2D and fasting insulin with serum urate concentrations and a suggestive association of fasting insulin with risk of gout. Future research is required to examine the underlying biological mechanisms on such relationships.
Collapse
Affiliation(s)
- Jiahao Zhu
- Department of Epidemiology and Health Statistics, School of Public Health, Hangzhou Medical College, Hangzhou, China
| | - Lingling Sun
- Department of Orthopaedics, the Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| | - Jing Yang
- Zhuji People's Hospital of Zhejiang Province, Zhuji Affiliated Hospital of Shaoxing University, Zhuji, China
| | - Jiayao Fan
- Department of Epidemiology and Health Statistics, School of Public Health, Hangzhou Medical College, Hangzhou, China
| | - Lap Ah Tse
- JC School of Public Health and Primary Care, the Chinese University of Hong Kong, New Territories, Hong Kong
| | - Yingjun Li
- Department of Epidemiology and Health Statistics, School of Public Health, Hangzhou Medical College, Hangzhou, China
| |
Collapse
|
6
|
Modulation of Insulin Sensitivity by Insulin-Degrading Enzyme. Biomedicines 2021; 9:biomedicines9010086. [PMID: 33477364 PMCID: PMC7830943 DOI: 10.3390/biomedicines9010086] [Citation(s) in RCA: 35] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2020] [Revised: 01/14/2021] [Accepted: 01/15/2021] [Indexed: 12/15/2022] Open
Abstract
Insulin-degrading enzyme (IDE) is a highly conserved and ubiquitously expressed metalloprotease that degrades insulin and several other intermediate-size peptides. For many decades, IDE had been assumed to be involved primarily in hepatic insulin clearance, a key process that regulates availability of circulating insulin levels for peripheral tissues. Emerging evidence, however, suggests that IDE has several other important physiological functions relevant to glucose and insulin homeostasis, including the regulation of insulin secretion from pancreatic β-cells. Investigation of mice with tissue-specific genetic deletion of Ide in the liver and pancreatic β-cells (L-IDE-KO and B-IDE-KO mice, respectively) has revealed additional roles for IDE in the regulation of hepatic insulin action and sensitivity. In this review, we discuss current knowledge about IDE’s function as a regulator of insulin secretion and hepatic insulin sensitivity, both evaluating the classical view of IDE as an insulin protease and also exploring evidence for several non-proteolytic functions. Insulin proteostasis and insulin sensitivity have both been highlighted as targets controlling blood sugar levels in type 2 diabetes, so a clearer understanding the physiological functions of IDE in pancreas and liver could led to the development of novel therapeutics for the treatment of this disease.
Collapse
|
7
|
Albiero M, Bonora BM, Fadini GP. Diabetes pharmacotherapy and circulating stem/progenitor cells. State of the art and evidence gaps. Curr Opin Pharmacol 2020; 55:151-156. [PMID: 33271409 DOI: 10.1016/j.coph.2020.10.019] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2020] [Revised: 10/22/2020] [Accepted: 10/23/2020] [Indexed: 12/13/2022]
Abstract
Diabetes is burdened with the development of several end-organ complications leading to excess mortality. Though the causes of such organ damage are far from being clarified, diabetes has been redefined as a disease of impaired damage control, wherein ongoing damage is not adequately compensated by activation of repair processes. Bone marrow-derived hematopoietic stem/progenitor cells (HSPCs) and their descendants endothelial progenitor cells (EPCs) have been extensively studied as major players in tissue homeostasis as well as biomarkers of diabetic complication risk. Thus, strategies to raise the levels of circulating HSPCs/EPCs have attracted interest for their potential to modify the future risk of complications. We herein discuss state-of-the-art of the effects exerted by diabetes pharmacotherapy on such cell populations. Further, we highlight which outstanding questions remain to be addressed for a more comprehensive understanding of this topic.
Collapse
Affiliation(s)
- Mattia Albiero
- Department of Medicine, University of Padova, 35128 Padova, Italy; Veneto Institute of Molecular Medicine, 35128 Padova, Italy
| | - Benedetta Maria Bonora
- Department of Medicine, University of Padova, 35128 Padova, Italy; Veneto Institute of Molecular Medicine, 35128 Padova, Italy
| | - Gian Paolo Fadini
- Department of Medicine, University of Padova, 35128 Padova, Italy; Veneto Institute of Molecular Medicine, 35128 Padova, Italy.
| |
Collapse
|
8
|
Di Pino A, DeFronzo RA. Insulin Resistance and Atherosclerosis: Implications for Insulin-Sensitizing Agents. Endocr Rev 2019; 40:1447-1467. [PMID: 31050706 PMCID: PMC7445419 DOI: 10.1210/er.2018-00141] [Citation(s) in RCA: 231] [Impact Index Per Article: 38.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/15/2018] [Accepted: 10/18/2018] [Indexed: 12/12/2022]
Abstract
Patients with type 2 diabetes mellitus (T2DM) are at high risk for macrovascular complications, which represent the major cause of mortality. Despite effective treatment of established cardiovascular (CV) risk factors (dyslipidemia, hypertension, procoagulant state), there remains a significant amount of unexplained CV risk. Insulin resistance is associated with a cluster of cardiometabolic risk factors known collectively as the insulin resistance (metabolic) syndrome (IRS). Considerable evidence, reviewed herein, suggests that insulin resistance and the IRS contribute to this unexplained CV risk in patients with T2DM. Accordingly, CV outcome trials with pioglitazone have demonstrated that this insulin-sensitizing thiazolidinedione reduces CV events in high-risk patients with T2DM. In this review the roles of insulin resistance and the IRS in the development of atherosclerotic CV disease and the impact of the insulin-sensitizing agents and of other antihyperglycemic medications on CV outcomes are discussed.
Collapse
Affiliation(s)
- Antonino Di Pino
- Diabetes Division, University of Texas Health Science Center and Texas Diabetes Institute, San Antonio, Texas
| | - Ralph A DeFronzo
- Diabetes Division, University of Texas Health Science Center and Texas Diabetes Institute, San Antonio, Texas
| |
Collapse
|
9
|
Bae J, Kim G, Lee YH, Lee BW, Kang ES, Cha BS. Differential Effects of Thiazolidinediones and Dipeptidyl Peptidase-4 Inhibitors on Insulin Resistance and β-Cell Function in Type 2 Diabetes Mellitus: A Propensity Score-Matched Analysis. Diabetes Ther 2019; 10:149-158. [PMID: 30506494 PMCID: PMC6349276 DOI: 10.1007/s13300-018-0541-y] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/07/2018] [Indexed: 02/02/2023] Open
Abstract
INTRODUCTION Comparisons of the glycemic durability between thiazolidinediones (TZDs) and dipeptidyl peptidase-4 (DPP-4) inhibitors remain insufficient. This study aimed to find clues for the differences in glycemic durability between TZDs and DPP-4 inhibitors by comparing the insulin resistance and β-cell function among patients using these agents. METHODS A total of 241 patients with type 2 diabetes mellitus (T2DM) treated with either pioglitazone (a TZD) or DPP-4 inhibitors as combination therapy with metformin for at least 1 year were analyzed. A propensity score based on the patients' baseline characteristics and glycated hemoglobin (HbA1c) was used to match them. Indices for insulin resistance and secretory function of β-cells, namely the homeostasis model assessment of insulin resistance (HOMA-IR) or β-cells (HOMA-β), were calculated and compared. Multiple regression analysis was performed to find the independent variables correlated with β-cell function or insulin resistance. RESULTS Evaluation of the data from 168 matched patients with T2DM showed that TZD users had significantly better insulin sensitivity compared with DPP-4 inhibitor users (HOMA-IR 2.3 ± 1.9 vs. 3.5 ± 3.2, p = 0.003). Conversely, DPP-4 inhibitor users secreted more insulin than TZD users (HOMA-β 45.7 ± 31.6 vs. 61.4 ± 49.5, p = 0.016). Multiple linear regression analysis showed that these agents were independently associated with both insulin resistance and β-cell function. CONCLUSION TZD users showed significantly better insulin sensitivity, whereas DPP-4 inhibitor users secreted more insulin from β-cells under similar glycemic control.
Collapse
Affiliation(s)
- Jaehyun Bae
- Division of Endocrinology and Metabolism, Department of Internal Medicine, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Gyuri Kim
- Division of Endocrinology and Metabolism, Department of Internal Medicine, Yonsei University College of Medicine, Seoul, Republic of Korea
- Division of Endocrinology and Metabolism, Department of Medicine, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Republic of Korea
| | - Yong-Ho Lee
- Division of Endocrinology and Metabolism, Department of Internal Medicine, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Byung-Wan Lee
- Division of Endocrinology and Metabolism, Department of Internal Medicine, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Eun Seok Kang
- Division of Endocrinology and Metabolism, Department of Internal Medicine, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Bong-Soo Cha
- Division of Endocrinology and Metabolism, Department of Internal Medicine, Yonsei University College of Medicine, Seoul, Republic of Korea.
| |
Collapse
|
10
|
Khajebishak Y, Payahoo L, Alivand M, Alipour B. Punicic acid: A potential compound of pomegranate seed oil in Type 2 diabetes mellitus management. J Cell Physiol 2018; 234:2112-2120. [PMID: 30317607 DOI: 10.1002/jcp.27556] [Citation(s) in RCA: 37] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2018] [Accepted: 09/13/2018] [Indexed: 12/15/2022]
Abstract
Diabetes is one of the most prevalent diseases in the worldwide. Type 2 diabetes mellitus (T2DM), the most common form of the disease, has become a serious threat to public health and is a growing burden on global economies. Due to the unexpected adverse effects of antidiabetic medicines, the use of nutraceuticals as a complementary therapy has drawn extensive attention by investigators. In this issue, a novel nutraceutical, Punicic acid (PA)-the main ingredient of pomegranate seed oil (PSO) that has potential therapeutic effects in T2DM-has been investigated. PA is a peroxisome proliferator-activated receptor gamma agonist, and unlike synthetic ligands, such as thiazolidinediones, it has no side effects. PA exerts antidiabetic effects via various mechanisms, such as reducing inflammatory cytokines, modulating glucose homeostasis, and antioxidant properties. In this review, we discussed the potential therapeutic effects of PSO and PA and represented the related mechanisms involved in the management of T2DM.
Collapse
Affiliation(s)
- Yaser Khajebishak
- Nutrition Research Center, Drug Applied Research Center, Student Research Committee, Department of Nutrition, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Laleh Payahoo
- Nutrition Research Center, Drug Applied Research Center, Student Research Committee, Department of Nutrition, Tabriz University of Medical Sciences, Tabriz, Iran.,Department of Nutrition, Maraghe University of Medical Sciences, Maraghe, Iran
| | - Mohammadreza Alivand
- Department of Medical Genetics, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Beitollah Alipour
- Department of Community Nutrition, Faculty of Nutrition, Tabriz University of Medical Sciences, Tabriz, Iran
| |
Collapse
|
11
|
Paciello F, Fetoni AR, Rolesi R, Wright MB, Grassi C, Troiani D, Paludetti G. Pioglitazone Represents an Effective Therapeutic Target in Preventing Oxidative/Inflammatory Cochlear Damage Induced by Noise Exposure. Front Pharmacol 2018; 9:1103. [PMID: 30349478 PMCID: PMC6187064 DOI: 10.3389/fphar.2018.01103] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2018] [Accepted: 09/10/2018] [Indexed: 12/18/2022] Open
Abstract
Recent progress in hearing loss research has provided strong evidence for the imbalance of cellular redox status and inflammation as common predominant mechanisms of damage affecting the organ of Corti including noise induced hearing loss. The discovery of a protective molecule acting on both mechanisms is challenging. The thiazolidinediones, a class of antidiabetic drugs including pioglitazone and rosiglitazone, have demonstrated diverse pleiotrophic effects in many tissues where they exhibit anti-inflammatory, anti-proliferative, tissue protective effects and regulators of redox balance acting as agonist of peroxisome proliferator-activated receptors (PPARs). They are members of the family of ligand regulated nuclear hormone receptors that are also expressed in several cochlear cell types, including the outer hair cells. In this study, we investigated the protective capacity of pioglitazone in a model of noise-induced hearing loss in Wistar rats and the molecular mechanisms underlying this protective effects. Specifically, we employed a formulation of pioglitazone in a biocompatible thermogel providing rapid, uniform and sustained inner ear drug delivery via transtympanic injection. Following noise exposure (120 dB, 10 kHz, 1 h), different time schedules of treatment were employed: we explored the efficacy of pioglitazone given immediately (1 h) or at delayed time points (24 and 48 h) after noise exposure and the time course and extent of hearing recovery were assessed. We found that pioglitazone was able to protect auditory function at the mid-high frequencies and to limit cell death in the cochlear basal/middle turn, damaged by noise exposure. Immunofluorescence and western blot analysis provided evidence that pioglitazone mediates both anti-inflammatory and anti-oxidant effects by decreasing NF-κB and IL-1β expression in the cochlea and opposing the oxidative damage induced by noise insult. These results suggest that intratympanic pioglitazone can be considered a valid therapeutic strategy for attenuating noise-induced hearing loss and cochlear damage, reducing inflammatory signaling and restoring cochlear redox balance.
Collapse
Affiliation(s)
- Fabiola Paciello
- Fondazione Policlinico Universitario A. Gemelli, IRCCS, Rome, Italy.,Institute of Otolaryngology, Università Cattolica del Sacro Cuore, Rome, Italy.,Institute of Cell Biology and Neurobiology, CNR, Rome, Italy
| | - Anna Rita Fetoni
- Fondazione Policlinico Universitario A. Gemelli, IRCCS, Rome, Italy.,Institute of Otolaryngology, Università Cattolica del Sacro Cuore, Rome, Italy.,Institute of Cell Biology and Neurobiology, CNR, Rome, Italy
| | - Rolando Rolesi
- Fondazione Policlinico Universitario A. Gemelli, IRCCS, Rome, Italy
| | | | - Claudio Grassi
- Fondazione Policlinico Universitario A. Gemelli, IRCCS, Rome, Italy.,Institute of Human Physiology, Università Cattolica del Sacro Cuore, Rome, Italy
| | - Diana Troiani
- Institute of Human Physiology, Università Cattolica del Sacro Cuore, Rome, Italy
| | - Gaetano Paludetti
- Fondazione Policlinico Universitario A. Gemelli, IRCCS, Rome, Italy.,Institute of Otolaryngology, Università Cattolica del Sacro Cuore, Rome, Italy
| |
Collapse
|
12
|
Anagnostis P, Siolos P, Christou K, Gkekas NK, Kosmidou N, Athyros VG, Karagiannis A. The effect of antidiabetic medications on the cardiovascular system: a critical appraisal of current data. Hormones (Athens) 2018; 17:83-95. [PMID: 29858866 DOI: 10.1007/s42000-018-0017-5] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/27/2017] [Accepted: 01/23/2018] [Indexed: 02/07/2023]
Abstract
Both type 1 and type 2 diabetes are associated with increased risk for cardiovascular disease (CVD) events. This risk seems to be reduced by achievement of euglycemia. However, after the withdrawal of rosiglitazone from the market, the question arose as to whether this risk concerns simply a matter of euglycemia or the distinct role played by each antidiabetic drug with respect to its effect on CVD risk. To address this issue, many studies have been published during the last decade involving old and new antidiabetic agents, which however yielded contradictory results. Briefly, metformin is still considered safe and confers a beneficial effect on CVD risk. Conflicting data exist as concerns sulfonylureas, although the second and third generation representatives are regarded as relatively safe. Pioglitazone use seems to be associated with a reduction in CVD risk, whereas the dipeptidyl-dipeptidase-4 inhibitors (DPP-4i), lixisenatide and exenatide-LAR [from the category of glucagon-like-peptide-1 receptor (GLP-1R) agonists], confer a neutral effect. Two other GLP-1R agonists, liraglutide and semaglutide, as well as the sodium-glucose transporter-2 (SGLT2)-inhibitors, empagliflozin and cangliflozin, have shown an additional effect on CVD risk reduction, although their safety is in doubt. Insulin analogues and newer long-acting compounds are also safe for the cadiovascular system. The aim of this narrative review is to present and critically analyse the current data for each antidiabetic drug category with regard to their effect on CVD risk.
Collapse
Affiliation(s)
- Panagiotis Anagnostis
- Police Medical Center of Thessaloniki, Thessaloniki, Greece.
- Second Propedeutic Department of Internal Medicine, Medical School, Aristotle University of Thessaloniki, Hippokration Hospital, Sarantaporou 10, 546 40, Thessaloniki, Greece.
| | - Pavlos Siolos
- Police Medical Center of Thessaloniki, Thessaloniki, Greece
| | | | - Nifon K Gkekas
- Police Medical Center of Thessaloniki, Thessaloniki, Greece
| | | | - Vasilios G Athyros
- Second Propedeutic Department of Internal Medicine, Medical School, Aristotle University of Thessaloniki, Hippokration Hospital, Sarantaporou 10, 546 40, Thessaloniki, Greece
| | - Asterios Karagiannis
- Second Propedeutic Department of Internal Medicine, Medical School, Aristotle University of Thessaloniki, Hippokration Hospital, Sarantaporou 10, 546 40, Thessaloniki, Greece
| |
Collapse
|
13
|
Almon E, Khoury T, Drori A, Gingis-Velitski S, Alon S, Chertkoff R, Mushkat M, Shaaltiel Y, Ilan Y. An oral administration of a recombinant anti-TNF fusion protein is biologically active in the gut promoting regulatory T cells: Results of a phase I clinical trial using a novel oral anti-TNF alpha-based therapy. J Immunol Methods 2017; 446:21-29. [PMID: 28392436 DOI: 10.1016/j.jim.2017.03.023] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2017] [Revised: 03/30/2017] [Accepted: 03/30/2017] [Indexed: 02/07/2023]
Abstract
BACKGROUND An orally administered BY-2 plant cell-expressed recombinant anti-TNF fusion protein (PRX-106) consists of the soluble form of the human TNF receptor (TNFR) fused to the Fc component of a human IgG1 domain. Aim This study aim at determining the safety and the immune modulatory effect of an oral administration of PRX-106 in humans. METHODS Three different doses (2, 8 or 16mg/day) of PRX-106 were orally administered for five consecutive days in 14 healthy volunteered participants. Subjects were followed for safety parameters and for an effect on T lymphocytes subsets and cytokine levels. RESULTS An oral administration of PRX-106 was safe and well tolerated. The PK study showed that PRX106 is not absorbed. No effect on white blood cells and lymphocytes counts were noted. A dose dependent effect was noted on systemic lymphocytes. The oral administration of all three dosages was associated with an increase in CD4+CD25+ and CD8+CD25+ subset of suppressor lymphocytes. A marked increase in CD4+CD25+FoxP3 regulatory T cells was noted in the 8mg treated group. In addition, NKT regulatory cells, CD3+CD69+ and CD4+CD62 lymphocyte subsets increased with treatment. No changes in serum TNF alpha were observed. CONCLUSION An oral administration of the non-absorbable recombinant anti-TNF fusion protein, PRX-106, is safe, not associated with immune suppression, while inducing a favorable anti-inflammatory immune modulation. The PRX-106 may provide a safe orally administered effective anti-TNF alpha-based immune therapy for inflammatory bowel diseases and non-alcoholic steatohepatitis, as well as other autoimmune, TNF-mediated diseases.
Collapse
Affiliation(s)
| | - Tawfik Khoury
- Gastroenterology and Liver Units, Department of Medicine, Hadassah Hebrew University Medical Center, Jerusalem, Israel
| | - Ariel Drori
- Gastroenterology and Liver Units, Department of Medicine, Hadassah Hebrew University Medical Center, Jerusalem, Israel
| | | | | | | | - Mordechai Mushkat
- Gastroenterology and Liver Units, Department of Medicine, Hadassah Hebrew University Medical Center, Jerusalem, Israel
| | | | - Yaron Ilan
- Gastroenterology and Liver Units, Department of Medicine, Hadassah Hebrew University Medical Center, Jerusalem, Israel.
| |
Collapse
|
14
|
Katsiki N, Mikhailidis DP. Comment on Inzucchi et al. Pioglitazone Prevents Diabetes in Patients With Insulin Resistance and Cerebrovascular Disease. Diabetes Care 2016;39:1684-1692. Diabetes Care 2017; 40:e46. [PMID: 28325803 DOI: 10.2337/dc16-2301] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/03/2023]
Affiliation(s)
- Niki Katsiki
- Second Propedeutic Department of Internal Medicine, School of Medicine, Aristotle University of Thessaloniki, Hippokration Hospital, Thessaloniki, Greece
| | - Dimitri P Mikhailidis
- Department of Clinical Biochemistry, Royal Free Hospital Campus, University College London Medical School, University College London, London, U.K.
| |
Collapse
|
15
|
Kumar Singh A, Tripathi AC, Tewari A, Chawla V, Saraf SK. Design and microwave facilitated green synthesis of 2-[4-(3-carboxymethyl, methoxy carbonylmethyl-2,4-dioxo and 4-oxo-2-thioxo-thiazolidin-5-ylidenemethyl)-phenoxy]-2 and 3-methyl propionic acid ethyl ester derivatives: a novel structural class of antidyslipidemic agents. Med Chem Res 2017. [DOI: 10.1007/s00044-017-1875-0] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/28/2023]
|
16
|
Affiliation(s)
- Christos V Rizos
- a Department of Internal Medicine, School of Medicine , University of Ioannina , Ioannina , Greece
| | - Moses S Elisaf
- a Department of Internal Medicine, School of Medicine , University of Ioannina , Ioannina , Greece
| |
Collapse
|
17
|
Rizos CV, Kei A, Elisaf MS. The current role of thiazolidinediones in diabetes management. Arch Toxicol 2016; 90:1861-81. [DOI: 10.1007/s00204-016-1737-4] [Citation(s) in RCA: 47] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2016] [Accepted: 04/28/2016] [Indexed: 12/17/2022]
|
18
|
Ding F, Qiu J, Li Q, Hu J, Song C, Han C, He H, Wang J. Effects of rosiglitazone on proliferation and differentiation of duck preadipocytes. In Vitro Cell Dev Biol Anim 2015; 52:174-81. [PMID: 26487429 DOI: 10.1007/s11626-015-9958-z] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2015] [Accepted: 09/10/2015] [Indexed: 12/23/2022]
Abstract
Rosiglitazone (RSG), one member of the thiazolidinediones (TZDs), is a type of anti-diabetic drug in diabetic humans and animal models, whose function remains unknown in waterfowl. In this study, effects of RSG on duck preadipocyte differentiation were investigated. We detected cell viability using CCK method and measured the mRNA expression of key genes and protein contents involved in preadipocyte differentiation via qRT-PCR and ELISA kits, respectively. Lipid accumulation was determined via Oil Red O staining extraction, and lipolysis was measured by free fatty acid release in the culture medium. Results showed that high concentrations of RSG (50, 100 μM) significantly decreased cell viability. RSG (0-10 μM) enhanced preadipocyte differentiation in a dose-dependent manner and thus promoted lipid accumulation. With increasing RSG concentrations, cellular lipid content gradually decreased and preadipocyte differentiation was suppressed. mRNA expression of key genes involved in preadipocyte differentiation including FAS, ACC, SCD1, LPL, PLIN, SREBP1c, and ATGL were significantly upregulated by RSG, and the protein content of FAS, ACC, and ATGL were also increased in response to RSG. Meanwhile, RSG exposure increased free fatty acid release in the culture medium. Similar results were obtained in response to RSG plus oleate that was used to induce cell differentiation. These findings suggest that RSG does not promote duck preadipocyte viability, but it does induce duck preadipocyte differentiation, which might influence both lipogenesis and lipolysis pathways.
Collapse
Affiliation(s)
- Fang Ding
- Farm Animal Genetic Resources Exploration and Innovation Key Laboratory of Sichuan Province, Sichuan Agricultural University, Wenjiang, Sichuan, 611130, China
- Suzhou Institute of Systems Medicine, Center of System Medicine, Chinese Academy of Medical Sciences, Suzhou, Jiangsu, 215123, China
| | - Jiamin Qiu
- Farm Animal Genetic Resources Exploration and Innovation Key Laboratory of Sichuan Province, Sichuan Agricultural University, Wenjiang, Sichuan, 611130, China
| | - Qingqing Li
- Farm Animal Genetic Resources Exploration and Innovation Key Laboratory of Sichuan Province, Sichuan Agricultural University, Wenjiang, Sichuan, 611130, China
| | - Jiwei Hu
- Farm Animal Genetic Resources Exploration and Innovation Key Laboratory of Sichuan Province, Sichuan Agricultural University, Wenjiang, Sichuan, 611130, China
| | - Chenling Song
- Farm Animal Genetic Resources Exploration and Innovation Key Laboratory of Sichuan Province, Sichuan Agricultural University, Wenjiang, Sichuan, 611130, China
| | - Chunchun Han
- Farm Animal Genetic Resources Exploration and Innovation Key Laboratory of Sichuan Province, Sichuan Agricultural University, Wenjiang, Sichuan, 611130, China
| | - Hua He
- Farm Animal Genetic Resources Exploration and Innovation Key Laboratory of Sichuan Province, Sichuan Agricultural University, Wenjiang, Sichuan, 611130, China
| | - Jiwen Wang
- Farm Animal Genetic Resources Exploration and Innovation Key Laboratory of Sichuan Province, Sichuan Agricultural University, Wenjiang, Sichuan, 611130, China.
| |
Collapse
|
19
|
Mavian AA, Miller S, Henry RR. Managing Type 2 Diabetes: Balancing HbA1cand Body Weight. Postgrad Med 2015; 122:106-17. [DOI: 10.3810/pgm.2010.05.2148] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023]
|
20
|
Katsiki N, Mikhailidis DP, Nair DR. The effects of antiepileptic drugs on vascular risk factors: A narrative review. Seizure 2014; 23:677-84. [DOI: 10.1016/j.seizure.2014.05.011] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2013] [Revised: 05/21/2014] [Accepted: 05/24/2014] [Indexed: 12/13/2022] Open
|
21
|
Yang HM, Kim BK, Kim JY, Kwon YW, Jin S, Lee JE, Cho HJ, Lee HY, Kang HJ, Oh BH, Park YB, Kim HS. PPARγ modulates vascular smooth muscle cell phenotype via a protein kinase G-dependent pathway and reduces neointimal hyperplasia after vascular injury. Exp Mol Med 2013; 45:e65. [PMID: 24287871 PMCID: PMC3849568 DOI: 10.1038/emm.2013.112] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2013] [Accepted: 08/01/2013] [Indexed: 01/28/2023] Open
Abstract
Vascular smooth muscle cells (VSMCs) undergo phenotypic changes in response to vascular injury such as angioplasty. Protein kinase G (PKG) has an important role in the process of VSMC phenotype switching. In this study, we examined whether rosiglitazone, a peroxisome proliferator-activated receptor (PPAR)-γ agonist, could modulate VSMC phenotype through the PKG pathway to reduce neointimal hyperplasia after angioplasty. In vitro experiments showed that rosiglitazone inhibited the phenotype change of VSMCs from a contractile to a synthetic form. The platelet-derived growth factor (PDGF)-induced reduction of PKG level was reversed by rosiglitazone treatment, resulting in increased PKG activity. This increased activity of PKG resulted in phosphorylation of vasodilator-stimulated phosphoprotein at serine 239, leading to inhibited proliferation of VSMCs. Interestingly, rosiglitazone did not change the level of nitric oxide (NO) or cyclic guanosine monophosphate (cGMP), which are upstream of PKG, suggesting that rosiglitazone influences PKG itself. Chromatin immunoprecipitation assays for the PKG promoter showed that the activation of PKG by rosiglitazone was mediated by the increased binding of Sp1 on the promoter region of PKG. In vivo experiments showed that rosiglitazone significantly inhibited neointimal formation after balloon injury. Immunohistochemistry staining for calponin and thrombospondin showed that this effect of rosiglitazone was mediated by modulating VSMC phenotype. Our findings demonstrate that rosiglitazone is a potent modulator of VSMC phenotype, which is regulated by PKG. This activation of PKG by rosiglitazone results in reduced neointimal hyperplasia after angioplasty. These results provide important mechanistic insight into the cardiovascular-protective effect of PPARγ.
Collapse
Affiliation(s)
- Han-Mo Yang
- National Research Laboratory for Stem Cell Niche, Seoul National University College of Medicine, Seoul, Korea
- Department of Internal Medicine, Seoul National University Hospital, Seoul, Korea
- Innovative Research Institute for Cell Therapy, Seoul National University Hospital, Seoul, Korea
| | - Baek-Kyung Kim
- National Research Laboratory for Stem Cell Niche, Seoul National University College of Medicine, Seoul, Korea
- Innovative Research Institute for Cell Therapy, Seoul National University Hospital, Seoul, Korea
| | - Ju-Young Kim
- National Research Laboratory for Stem Cell Niche, Seoul National University College of Medicine, Seoul, Korea
- Innovative Research Institute for Cell Therapy, Seoul National University Hospital, Seoul, Korea
| | - Yoo-Wook Kwon
- National Research Laboratory for Stem Cell Niche, Seoul National University College of Medicine, Seoul, Korea
- Innovative Research Institute for Cell Therapy, Seoul National University Hospital, Seoul, Korea
| | - Sooryeonhwa Jin
- National Research Laboratory for Stem Cell Niche, Seoul National University College of Medicine, Seoul, Korea
- Innovative Research Institute for Cell Therapy, Seoul National University Hospital, Seoul, Korea
| | - Joo-Eun Lee
- National Research Laboratory for Stem Cell Niche, Seoul National University College of Medicine, Seoul, Korea
- Innovative Research Institute for Cell Therapy, Seoul National University Hospital, Seoul, Korea
| | - Hyun-Jai Cho
- National Research Laboratory for Stem Cell Niche, Seoul National University College of Medicine, Seoul, Korea
- Department of Internal Medicine, Seoul National University Hospital, Seoul, Korea
- Innovative Research Institute for Cell Therapy, Seoul National University Hospital, Seoul, Korea
| | - Hae-Young Lee
- National Research Laboratory for Stem Cell Niche, Seoul National University College of Medicine, Seoul, Korea
- Department of Internal Medicine, Seoul National University Hospital, Seoul, Korea
- Innovative Research Institute for Cell Therapy, Seoul National University Hospital, Seoul, Korea
| | - Hyun-Jae Kang
- National Research Laboratory for Stem Cell Niche, Seoul National University College of Medicine, Seoul, Korea
- Department of Internal Medicine, Seoul National University Hospital, Seoul, Korea
- Innovative Research Institute for Cell Therapy, Seoul National University Hospital, Seoul, Korea
| | - Byung-Hee Oh
- National Research Laboratory for Stem Cell Niche, Seoul National University College of Medicine, Seoul, Korea
- Department of Internal Medicine, Seoul National University Hospital, Seoul, Korea
- Innovative Research Institute for Cell Therapy, Seoul National University Hospital, Seoul, Korea
| | - Young-Bae Park
- National Research Laboratory for Stem Cell Niche, Seoul National University College of Medicine, Seoul, Korea
- Department of Internal Medicine, Seoul National University Hospital, Seoul, Korea
- Innovative Research Institute for Cell Therapy, Seoul National University Hospital, Seoul, Korea
| | - Hyo-Soo Kim
- National Research Laboratory for Stem Cell Niche, Seoul National University College of Medicine, Seoul, Korea
- Department of Internal Medicine, Seoul National University Hospital, Seoul, Korea
- Innovative Research Institute for Cell Therapy, Seoul National University Hospital, Seoul, Korea
- Molecular Medicine and Biopharmaceutical Sciences, Seoul National University, Seoul, Korea
| |
Collapse
|
22
|
Rizos CV, Liberopoulos EN, Tellis K, DiNicolantonio JJ, Tselepis AD, Elisaf MS. Combining Rosuvastatin With Angiotensin-Receptor Blockers of Different PPARγ-Activating Capacity. Angiology 2013; 66:36-42. [DOI: 10.1177/0003319713512556] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
The effects of combining angiotensin-receptor blockers of different peroxisome proliferator-activated receptor γ-activating capacity with rosuvastatin on high-density lipoprotein (HDL) subfractions and associated enzymes in patients with mixed dyslipidemia, hypertension, and prediabetes were assessed. Patients (n = 151) were randomly allocated to rosuvastatin (10 mg/d) plus telmisartan 80 mg/d (RT group, n = 52) or irbesartan 300 mg/d (RI group, n = 48) or olmesartan 20 mg/d (RO group, n = 51). Total and intermediate HDL cholesterol (HDL-C) levels did not change in any group. Large HDL-C increased, while small HDL-C decreased significantly in all the groups ( P = not significant between the groups). The mass of HDL lipoprotein-associated phospholipase A2 (HDL-Lp-PLA2) and the activities of paraoxonase 1 remained unchanged in all the groups. However, HDL-Lp-PLA2 activity increased only in the RT group (+21.4%; P < .01 vs baseline) and did not change in the RI (−4.3%; P = .005 vs RT group) and RO (+3.2%; P = .01 vs RT) groups. In conclusion, only the combination of rosuvastatin with telmisartan increased the possibly antiatherosclerotic HDL-Lp-PLA2 activity.
Collapse
Affiliation(s)
- Christos V. Rizos
- Department of Internal Medicine, School of Medicine, University of Ioannina, Ioannina, Greece
| | | | - Kostas Tellis
- Laboratory of Biochemistry, School of Chemistry, University of Ioannina, Ioannina, Greece
| | - James J. DiNicolantonio
- Mid America Heart Institute, Saint Luke’s Hospital, Kansas City, MO, USA
- Wegmans Pharmacy, Ithaca, NY, USA
| | - Alexandros D. Tselepis
- Laboratory of Biochemistry, School of Chemistry, University of Ioannina, Ioannina, Greece
| | - Moses S. Elisaf
- Department of Internal Medicine, School of Medicine, University of Ioannina, Ioannina, Greece
| |
Collapse
|
23
|
Katsiki N, Karagiannis A, Athyros VG, Mikhailidis DP. Hyperuricaemia. J Cardiovasc Med (Hagerstown) 2013; 14:397-402. [DOI: 10.2459/jcm.0b013e3283595adc] [Citation(s) in RCA: 59] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
|
24
|
Rizzo M, Avogaro A, Montalto G, Rizvi AA. Non-glycemic effects of pioglitazone and incretin-based therapies. Expert Opin Ther Targets 2013; 17:739-42. [PMID: 23691979 DOI: 10.1517/14728222.2013.804067] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
Abstract
Atherosclerosis and cardiovascular events are highly prevalent and represent the major cause of mortality in patients with type 2 diabetes. Therefore, there is significant interest in the non-glycemic properties of anti-diabetic agents, particularly on those that are effective on cardiovascular risk factors. Thiazolidinediones and incretin-based therapies (IBTs) represent some of the most recent treatment options approved for the management of type 2 diabetes; these agents have shown important glycemic effects, as well as a number of non-glycemic effects. The latter include those on body weight, inflammation, hypertension and dyslipidemia, thus impacting the different components of the metabolic syndrome. Pioglitazone has been shown to significantly reduce cardiovascular adverse outcomes, while preliminary data on IBTs are very encouraging as well. Although highlighting the non-glycemic effects of pioglitazone and incretin-based therapies is of potential significance, clinical practice and patient care must be based largely on evidence-based medicine. Therefore, definitive opinions will await additional data from ongoing studies evaluating the effects of both GLP-1 agonists and DPP-4 inhibitors on cardiovascular morbidity and mortality.
Collapse
|
25
|
Rizos CV, Elisaf MS. Metformin and cancer. Eur J Pharmacol 2013; 705:96-108. [PMID: 23499688 DOI: 10.1016/j.ejphar.2013.02.038] [Citation(s) in RCA: 116] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2012] [Revised: 02/16/2013] [Accepted: 02/24/2013] [Indexed: 12/13/2022]
Abstract
Type 2 diabetes mellitus is a rising cause of cardiovascular morbidity and mortality. A number of studies have also identified diabetic patients as having increased risk for the development of cancer. Metformin is a widely prescribed antidiabetic drug with an established efficacy coupled with a favorable safety profile and low cost. An increasing number of studies have associated metformin treatment with a decrease of cancer risk. Moreover, metformin has also been associated with improved outcomes in cancer patients. These possible pleiotropic effects of metformin may establish metformin as a cancer prevention and treatment option. However, any favorable effects of metformin on cancer are not always corroborated by clinical trials. Larger studies are expected to better investigate the possible antineoplastic effects of metformin.
Collapse
Affiliation(s)
- Christos V Rizos
- Department of Internal Medicine, School of Medicine, University of Ioannina, Ioannina, Greece
| | | |
Collapse
|
26
|
Köttgen A, Albrecht E, Teumer A, Vitart V, Krumsiek J, Hundertmark C, Pistis G, Ruggiero D, O'Seaghdha CM, Haller T, Yang Q, Tanaka T, Johnson AD, Kutalik Z, Smith AV, Shi J, Struchalin M, Middelberg RPS, Brown MJ, Gaffo AL, Pirastu N, Li G, Hayward C, Zemunik T, Huffman J, Yengo L, Zhao JH, Demirkan A, Feitosa MF, Liu X, Malerba G, Lopez LM, van der Harst P, Li X, Kleber ME, Hicks AA, Nolte IM, Johansson A, Murgia F, Wild SH, Bakker SJL, Peden JF, Dehghan A, Steri M, Tenesa A, Lagou V, Salo P, Mangino M, Rose LM, Lehtimäki T, Woodward OM, Okada Y, Tin A, Müller C, Oldmeadow C, Putku M, Czamara D, Kraft P, Frogheri L, Thun GA, Grotevendt A, Gislason GK, Harris TB, Launer LJ, McArdle P, Shuldiner AR, Boerwinkle E, Coresh J, Schmidt H, Schallert M, Martin NG, Montgomery GW, Kubo M, Nakamura Y, Tanaka T, Munroe PB, Samani NJ, Jacobs DR, Liu K, D'Adamo P, Ulivi S, Rotter JI, Psaty BM, Vollenweider P, Waeber G, Campbell S, Devuyst O, Navarro P, Kolcic I, Hastie N, Balkau B, Froguel P, Esko T, Salumets A, Khaw KT, Langenberg C, Wareham NJ, Isaacs A, Kraja A, Zhang Q, Wild PS, Scott RJ, Holliday EG, Org E, Viigimaa M, Bandinelli S, Metter JE, Lupo A, Trabetti E, Sorice R, Döring A, Lattka E, Strauch K, Theis F, Waldenberger M, Wichmann HE, Davies G, Gow AJ, Bruinenberg M, Stolk RP, Kooner JS, Zhang W, Winkelmann BR, Boehm BO, Lucae S, Penninx BW, Smit JH, Curhan G, Mudgal P, Plenge RM, Portas L, Persico I, Kirin M, Wilson JF, Mateo Leach I, van Gilst WH, Goel A, Ongen H, Hofman A, Rivadeneira F, Uitterlinden AG, Imboden M, von Eckardstein A, Cucca F, Nagaraja R, Piras MG, Nauck M, Schurmann C, Budde K, Ernst F, Farrington SM, Theodoratou E, Prokopenko I, Stumvoll M, Jula A, Perola M, Salomaa V, Shin SY, Spector TD, Sala C, Ridker PM, Kähönen M, Viikari J, Hengstenberg C, Nelson CP, Meschia JF, Nalls MA, Sharma P, Singleton AB, Kamatani N, Zeller T, Burnier M, Attia J, Laan M, Klopp N, Hillege HL, Kloiber S, Choi H, Pirastu M, Tore S, Probst-Hensch NM, Völzke H, Gudnason V, Parsa A, Schmidt R, Whitfield JB, Fornage M, Gasparini P, Siscovick DS, Polašek O, Campbell H, Rudan I, Bouatia-Naji N, Metspalu A, Loos RJF, van Duijn CM, Borecki IB, Ferrucci L, Gambaro G, Deary IJ, Wolffenbuttel BHR, Chambers JC, März W, Pramstaller PP, Snieder H, Gyllensten U, Wright AF, Navis G, Watkins H, Witteman JCM, Sanna S, Schipf S, Dunlop MG, Tönjes A, Ripatti S, Soranzo N, Toniolo D, Chasman DI, Raitakari O, Kao WHL, Ciullo M, Fox CS, Caulfield M, Bochud M, Gieger C. Genome-wide association analyses identify 18 new loci associated with serum urate concentrations. Nat Genet 2013; 45:145-54. [PMID: 23263486 PMCID: PMC3663712 DOI: 10.1038/ng.2500] [Citation(s) in RCA: 607] [Impact Index Per Article: 50.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2012] [Accepted: 11/27/2012] [Indexed: 12/13/2022]
Abstract
Elevated serum urate concentrations can cause gout, a prevalent and painful inflammatory arthritis. By combining data from >140,000 individuals of European ancestry within the Global Urate Genetics Consortium (GUGC), we identified and replicated 28 genome-wide significant loci in association with serum urate concentrations (18 new regions in or near TRIM46, INHBB, SFMBT1, TMEM171, VEGFA, BAZ1B, PRKAG2, STC1, HNF4G, A1CF, ATXN2, UBE2Q2, IGF1R, NFAT5, MAF, HLF, ACVR1B-ACVRL1 and B3GNT4). Associations for many of the loci were of similar magnitude in individuals of non-European ancestry. We further characterized these loci for associations with gout, transcript expression and the fractional excretion of urate. Network analyses implicate the inhibins-activins signaling pathways and glucose metabolism in systemic urate control. New candidate genes for serum urate concentration highlight the importance of metabolic control of urate production and excretion, which may have implications for the treatment and prevention of gout.
Collapse
Affiliation(s)
- Anna Köttgen
- Renal Division, Freiburg University Hospital, Freiburg, Germany.
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
27
|
Khan S, Khan S, Imran M, Pillai KK, Akhtar M, Najmi AK. Effects of pioglitazone and vildagliptin on coagulation cascade in diabetes mellitus – targeting thrombogenesis. Expert Opin Ther Targets 2013; 17:627-39. [DOI: 10.1517/14728222.2013.764991] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
|
28
|
Giampietro L, D’Angelo A, Giancristofaro A, Ammazzalorso A, De Filippis B, Fantacuzzi M, Linciano P, Maccallini C, Amoroso R. Synthesis and structure–activity relationships of fibrate-based analogues inside PPARs. Bioorg Med Chem Lett 2012; 22:7662-6. [DOI: 10.1016/j.bmcl.2012.09.111] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2012] [Revised: 09/26/2012] [Accepted: 09/29/2012] [Indexed: 02/02/2023]
|
29
|
Galle J, Kleophas W, Dellanna F, Schmid VHR, Forkel C, Dikta G, Krajewski V, Fuchs W, Forst T, Pfützner A. Comparison of the Effects of Pioglitazone versus Placebo when Given in Addition to Standard Insulin Treatment in Patients with Type 2 Diabetes Mellitus Requiring Hemodialysis: Results from the PIOren Study. NEPHRON EXTRA 2012; 2:104-14. [PMID: 22739963 PMCID: PMC3383235 DOI: 10.1159/000337334] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
BACKGROUND Patients with type 2 diabetes mellitus and advanced kidney disease are usually treated with insulin. However, the prolonged pharmacokinetic insulin profile in patients with delayed renal insulin elimination impairs a successful therapy. Due to its hepatic metabolism, pioglitazone is a potential candidate for additional administration. The aim of this study was to investigate the effect of pioglitazone versus placebo on total daily insulin requirements and several pleiotropic factors in type 2 diabetes patients requiring hemodialysis. METHODS The effect of pioglitazone (30 mg) versus placebo was explored in this prospective, randomized, double-blind parallel multicenter phase II study analyzing data from 36 patients with type 2 diabetes mellitus currently under hemodialysis (25 male, 11 female, aged 69.2 ± 7.9 years, baseline HbA1c 7.6 ± 0.9%). The most important efficacy parameters collected before dialysis and after an overnight fast at baseline and after 6 months were: total daily insulin dose, HbA1c, fasting blood glucose, adiponectin, HDL, LDL, triglycerides, NT-proBNP, and ultrafiltrate volume. RESULTS Application of pioglitazone resulted in a significant decrease of the daily insulin dose by 35% versus baseline (placebo: -10%, n.s.), improvement in HbA1c (-0.60 ± 0.87%, p = 0.015; placebo: 0.21 ± 1.1%, n.s.) and adiponectin (7.33 ± 4.80 mg/l, p < 0.001; placebo: -1.37 ± 2.56 mg/l, n.s.). Slight improvements or no changes were seen with fasting glucose, triglycerides, HDL, LDL and NT-proBNP. There was no indication of increased hypoglycemia risk and volume overload by the addition of pioglitazone. CONCLUSIONS Addition of pioglitazone to insulin in patients with late-stage kidney failure requiring hemodialysis is a well-tolerated treatment option that improves glycemic control with simultaneous insulin-sparing potential.
Collapse
Affiliation(s)
- Jan Galle
- Klinik für Nephrologie und Dialyseverfahren, Klinikum Lüdenscheid, Lüdenscheid, Germany
| | | | | | | | | | | | | | | | | | | |
Collapse
|
30
|
Katsiki N, Yovos JG, Gotzamani-Psarrakou A, Karamitsos DT. Adipokines and vascular risk in type 2 diabetes mellitus. Angiology 2012; 62:601-4. [PMID: 21990548 DOI: 10.1177/0003319711409201] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Affiliation(s)
- Niki Katsiki
- First Propedeutic Department of Internal Medicine, AHEPA University Hospital, Aristotle University of Thessaloniki, Thessaloniki, Greece
| | | | | | | |
Collapse
|
31
|
Zhang L, Li L, Yang M, Liu H, Yang G. Elevated circulating vaspin levels were decreased by rosiglitazone therapy in T2DM patients with poor glycemic control on metformin alone. Cytokine 2011; 56:399-402. [PMID: 21802961 DOI: 10.1016/j.cyto.2011.07.003] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2011] [Revised: 05/16/2011] [Accepted: 07/05/2011] [Indexed: 11/28/2022]
|
32
|
Katsiki N, Elisaf M. Multifactorial treatment for improvement of renal function and cardiovascular risk: an ATTEMPT for patients with metabolic syndrome and chronic kidney disease. Curr Med Res Opin 2011; 27:1669-72. [PMID: 21718096 DOI: 10.1185/03007995.2011.596410] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
|
33
|
Agouridis AP, Filippatos TD, Tsimihodimos V, Elisaf MS. Combinations of ezetimibe with nonstatin drug regimens affecting lipid metabolism. Expert Rev Cardiovasc Ther 2011; 9:355-66. [PMID: 21438815 DOI: 10.1586/erc.11.4] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
In this article we discuss the available data on the effects of combined therapy of ezetimibe with agents affecting lipid metabolism other than statins. We consider studies evaluating the effects of combined therapy of ezetimibe with bile acid sequestrants, fenofibrate, niacin, n-3 fatty acids, plant sterols, orlistat, metformin, acarbose and glitazones. Combination of ezetimibe with bile acid sequestrants (especially colesevelam) was shown to have additional effects on lipid parameters in patients with hyperlipidemia. Combination of ezetimibe with fenofibrate may be a good approach to improve the overall lipid profile of patients with mixed hyperlipidemia. The addition of ezetimibe to niacin-based therapy can be useful for high-risk patients with dyslipidemia who are not achieving their assigned treatment goals. For patients who cannot tolerate statins there are useful combinations of ezetimibe with other drugs affecting lipid metabolism. These combinations improve many metabolic parameters, but more trials should be carried out to reach more robust conclusions about their effects on cardiovascular disease prevention.
Collapse
Affiliation(s)
- Aris P Agouridis
- Department of Internal Medicine Medical School, University of Ioannina, Ioannina, Greece
| | | | | | | |
Collapse
|
34
|
Papanas N, Katsiki N, Hatzitolios AI, Maltezos E. Pioglitazone: a valuable component of combination therapy for type 2 diabetes mellitus. Expert Opin Pharmacother 2011; 12:1457-61. [DOI: 10.1517/14656566.2011.568477] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
|
35
|
Rizos CV, Liberopoulos EN, Tellis CC, Florentin M, Elisaf MS, Tselepis AD. Combining Rosuvastatin with Sartans of Different Peroxisome Proliferator-Activated Receptor-γ Activating Capacity Is Not Associated with Different Changes in Low-Density Lipoprotein Subfractions and Plasma Lipoprotein-Associated Phospholipase A2. Metab Syndr Relat Disord 2011; 9:217-23. [DOI: 10.1089/met.2010.0120] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Affiliation(s)
- Christos V. Rizos
- Department of Internal Medicine, School of Medicine, University of Ioannina, Ioannina, Greece
| | | | - Constantinos C. Tellis
- Laboratory of Biochemistry, School of Chemistry, University of Ioannina, Ioannina, Greece
| | - Matilda Florentin
- Department of Internal Medicine, School of Medicine, University of Ioannina, Ioannina, Greece
| | - Moses S. Elisaf
- Department of Internal Medicine, School of Medicine, University of Ioannina, Ioannina, Greece
| | - Alexandros D. Tselepis
- Laboratory of Biochemistry, School of Chemistry, University of Ioannina, Ioannina, Greece
| |
Collapse
|
36
|
Soskić SS, Dobutović BD, Sudar EM, Obradović MM, Nikolić DM, Zarić BL, Stojanović SD, Stokić EJ, Mikhailidis DP, Isenović ER. Peroxisome proliferator-activated receptors and atherosclerosis. Angiology 2011; 62:523-34. [PMID: 21467121 DOI: 10.1177/0003319711401012] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
The peroxisome proliferator-activated receptors (PPARs) represent the family of 3 nuclear receptor isoforms-PPARα, -γ, and -δ/β, which are encoded by different genes. As lipid sensors, they are primarily involved in regulation of lipid metabolism and subsequently in inflammation and atherosclerosis. Atherosclerosis considers accumulation of the cells and extracellular matrix in the vessel wall leading to the formation of atherosclerotic plaque, atherothrombosis, and other vascular complications. Besides existence of natural ligands for PPARs, their more potent synthetic ligands are fibrates and thiazolidindiones. Future investigations should now focus on the mechanisms of PPARs activation, which might present new approaches involved in the antiatherosclerotic effects revealed in this review. In addition, in this review we are presenting latest data from recent performed clinical studies which have focus on novel approach to PPARs agonists as potential therapeutic agents in the treatment of complex disease such as atherosclerosis.
Collapse
Affiliation(s)
- Sanja S Soskić
- Laboratory for Radiobiology and Molecular Genetics, Institute Vinca, University of Belgrade, Belgrade, Serbia
| | | | | | | | | | | | | | | | | | | |
Collapse
|
37
|
Vikram A, Jena G, Ramarao P. Pioglitazone attenuates prostatic enlargement in diet-induced insulin-resistant rats by altering lipid distribution and hyperinsulinaemia. Br J Pharmacol 2011; 161:1708-21. [PMID: 20726985 DOI: 10.1111/j.1476-5381.2010.00994.x] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
BACKGROUND AND PURPOSE Increased incidence of benign prostatic hyperplasia among insulin-resistant individuals suggests a role for hyperinsulinaemia in prostatic enlargement. We have already reported increased cell proliferation and enlargement of prostate gland in insulin-resistant rats. The present study aimed to elucidate the molecular mechanisms underlying the reversal of prostatic enlargement in insulin-resistant rats by the peroxisome proliferator-activated receptor γ agonist pioglitazone. EXPERIMENTAL APPROACH Sprague-Dawley rats were fed a normal pellet or a high-fat diet for 12 weeks with or without pioglitazone (20 mg·kg(-1)). Subgroups of animals fed different diets were castrated. Effects of dietary manipulation and pioglitazone were measured on insulin sensitivity, lipid distribution, cell proliferation and apoptosis. KEY RESULTS A high-fat diet led to the accumulation of fat in non-adipose tissues, insulin resistance, compensatory hyperinsulinaemia and prostatic enlargement in rats. Pioglitazone treatment altered fat distribution, improved insulin sensitivity and normalized lipid and insulin level in rats on the high-fat diet. The improved metabolic parameters led to decreased cellular proliferation and increased apoptosis in the prostate gland. High-fat diet feeding and pioglitazone treatment did not change plasma testosterone levels. However, significant prostatic atrophy was observed in castrated rats irrespective of dietary intervention. CONCLUSIONS AND IMPLICATIONS Our results show a previously unexplored therapeutic potential of pioglitazone for prostatic enlargement under insulin-resistant condition and further suggest that targeting distribution of lipid from non-adipose tissue to adipose tissue and insulin signalling could be new strategies for the treatment of benign prostatic hyperplasia.
Collapse
Affiliation(s)
- Ajit Vikram
- Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research, S. A. S. Nagar, Mohali, Punjab, India
| | | | | |
Collapse
|
38
|
Yang Y, Yang G. Rosiglitazone regulates IL-6-stimulated lipolysis in porcine adipocytes. Biochem Cell Biol 2011; 88:853-60. [PMID: 20921996 DOI: 10.1139/o10-116] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
Interleukin (IL)-6, a proinflammatory cytokine, stimulates adipocyte lipolysis and induces insulin resistance in obese and diabetic subjects. However, the effects of the anti-diabetic drug rosiglitazone on IL-6-stimulated lipolysis and the underlying molecular mechanism are largely unknown. In this study, we demonstrated that rosiglitazone suppressed IL-6-stimulated lipolysis in differentiated porcine adipocytes by inactivation of extracellular signal-related kinase (ERK). Meanwhile, rosiglitazone enhanced the lipolysis response of adipocytes to isoprenaline. In addition, rosiglitazone significantly reversed IL-6-induced down-regulation of several genes such as perilipin A, peroxisome proliferators activated receptor gamma (PPARγ), and fatty acid synthetase, as well as the up-regulation of IL-6 mRNA. However, mRNA expression of PPARγ coactivator-1 alpha (PCG-1α) was enhanced by rosiglitazone in IL-6-stimulated adipocytes. These results indicate that rosiglitazone suppresses IL-6-stimulated lipolysis in porcine adipocytes through multiple molecular mechanisms.
Collapse
Affiliation(s)
- Yongqing Yang
- College of Life Science, Shanxi Normal University, Linfen, Shanxi Province 041000, People's Republic of China.
| | | |
Collapse
|
39
|
Chatterjee PK. Hepatic inflammation and insulin resistance in pre-diabetes - further evidence for the beneficial actions of PPAR-gamma agonists and a role for SOCS-3 modulation. Br J Pharmacol 2010; 160:1889-91. [PMID: 20649587 DOI: 10.1111/j.1476-5381.2010.00739.x] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/14/2023] Open
Abstract
Pre-diabetes is a condition affecting increasing numbers of the population who find themselves caught in the grey area between normal glucose regulation and diabetes mellitus and who experience impaired glucose tolerance or fasting glucose. The ability of thiozolidinediones (TZDs) to ameliorate the clinical signs of diabetes mellitus is well-known but there is also emerging evidence for the benefits of PPAR-gamma agonists in pre-diabetes. In this issue of the British Journal of Pharmacology, Collino and colleagues report that pioglitazone can reduce hepatic inflammation and insulin resistance in rats administered a high cholesterol and fructose diet. Furthermore, pioglitazone reduced the expression of suppressor of cytokine signalling (SOCS)-3 - considered to be a key link between inflammation and insulin resistance. Although much work remains to be performed in fully understanding how TZDs modulate the cellular mechanisms which underlie pre-diabetes, these findings provide preliminary evidence that administration of TZDs to pre-diabetics could be beneficial.
Collapse
Affiliation(s)
- Prabal K Chatterjee
- Centre for Biomedical and Health Science Research, School of Pharmacy and Biomolecular Sciences, University of Brighton, Brighton, UK.
| |
Collapse
|
40
|
Abstract
Polycystic ovary syndrome (PCOS) is characterised by anovulation, hyperandrogenism, and polycystic ovaries. Fifty percent of patients fulfil the criteria for the metabolic syndrome. The pathogenesis of PCOS may be looked as a vicious cycle involving hyperandrogenemia, central obesity, and insulin resistance and medical treatment should aim at breaking this cycle. Lifestyle intervention, oral contraceptives, and insulin sensitises such as metformin are the most commonly used treatment modalities. The thiazolidinediones rosiglitazone and pioglitazone were recently applied as insulin sensitising treatment in patients with PCOS. Thiazolidinediones activate the transcription of genes that affect glucose and lipid metabolism mediating decreased free fatty acid levels and decreased visceral fat mass. In the present article, we give an overview of the diverse effects of glitazone treatment in patients with PCOS. We review the studies comparing glitazone versus metformin and oral contraceptive treatment and recommend which patients should be treated with glitazones in daily praxis.
Collapse
Affiliation(s)
- Dorte Glintborg
- Department of Endocrinology and Metabolism, Odense University Hospital, Odense C, Denmark.
| | | |
Collapse
|
41
|
Abstract
Diabetes mellitus is an established risk factor for cardiovascular disease and the leading cause of end-stage renal disease in the Western World. Thiazolidinediones (TZDs) represent a class of antidiabetic agents that exert their glucose-lowering effects by reducing insulin resistance, through stimulation of a type of nuclear receptor, called peroxisome proliferator-activated receptor-γ. Apart from improving glycemic control, TZDs were shown to exert beneficial effects on several components of the metabolic syndrome and cardiovascular risk markers. Furthermore, background and human studies have shown that TZDs reduce urinary albumin and protein excretion and interfere with most of the pathogenentic pathways involved in the development and progression of diabetic nephropathy. On the other hand, currently used TZDs have side effects, most important of which is fluid retention leading to wait gain and heart failure deterioration. With regards to cardiovascular outcomes, the anticipated benefit of TZDs was demonstrated for pioglitazone, whereas a series of previous meta-analyses linking rosiglitazone treatment with increased risk of myocardial infarction and cardiovascular death raised uncertainty around the cardiovascular safety of rosiglitazone. This article will discuss the effects of TZDs on established and emerging cardiovascular risk factors, the data on possible beneficial renal effects of these compounds, and the existing evidence from large-scale clinical trials and meta-analyses on their effects on cardiovascular outcomes, aiming to provide an overview of the cardio- and renoprotective properties of these drugs.
Collapse
Affiliation(s)
- Pantelis A Sarafidis
- Section of Nephrology and Hypertension, 1st Department of Medicine, AHEPA Hospital, Aristotle University of Thessaloniki, Greece
| | | | | |
Collapse
|
42
|
Abstract
BACKGROUND AND AIM Pioglitazone has diverse multiple effects on metabolic and inflammatory processes that have the potential to influence cardiovascular disease pathophysiology at various points in the disease process, including atherogenesis, plaque inflammation, plaque rupture, haemostatic disturbances and microangiopathy. RESULTS Linking the many direct and indirect effects on the vasculature to the reduction in key macrovascular outcomes reported with pioglitazone in patients with type 2 diabetes presents a considerable challenge. However, recent large-scale clinical cardiovascular imaging studies are beginning to provide some mechanistic insights, including a potentially important role for improvements in high-density lipoprotein cholesterol with pioglitazone. In addition to a role in prevention, animal studies also suggest that pioglitazone may minimize damage and improve recovery during and after ischaemic cardio- and cerebrovascular events. DESIGN AND METHODS In this review, we consider potential cardiovascular protective mechanisms of pioglitazone by linking preclinical data and clinical cardiovascular outcomes guided by insights from recent imaging studies. CONCLUSION Pioglitazone may influence CVD pathophysiology at multiple points in the disease process, including atherogenesis, plaque inflammation, plaque rupture and haemostatic disturbances (i.e. thrombus/embolism formation), as well as microangiopathy.
Collapse
Affiliation(s)
- E Erdmann
- Department of Medicine, Heart Center, University of Cologne, Cologne, Germany.
| | | |
Collapse
|
43
|
Abstract
The global burden of metabolic disease demands that we develop new therapeutic strategies. Many of these approaches may center on manipulating the behavior of adipocytes, which contribute directly and indirectly to a host of disease processes including obesity and type 2 diabetes. One way to achieve this goal will be to alter key transcriptional pathways in fat cells, such as those regulating glucose uptake, lipid handling, or adipokine secretion. In this review, we look at what is known about how adipocytes govern their physiology at the gene expression level, and discuss novel ways that we can accelerate our understanding of this area.
Collapse
Affiliation(s)
- Evan Rosen
- Beth Israel Deaconess Medical Center, Division of Endocrinology/CLS743, Boston, MA 02215, USA.
| | | | | |
Collapse
|
44
|
Site-specific modulation of white adipose tissue lipid metabolism by oleoyl-estrone and/or rosiglitazone in overweight rats. Naunyn Schmiedebergs Arch Pharmacol 2010; 381:339-48. [DOI: 10.1007/s00210-010-0495-8] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2009] [Accepted: 02/05/2010] [Indexed: 10/19/2022]
|
45
|
Kadoglou NPE, Tsanikidis H, Kapelouzou A, Vrabas I, Vitta I, Karayannacos PE, Liapis CD, Sailer N. Effects of rosiglitazone and metformin treatment on apelin, visfatin, and ghrelin levels in patients with type 2 diabetes mellitus. Metabolism 2010; 59:373-9. [PMID: 19815243 DOI: 10.1016/j.metabol.2009.08.005] [Citation(s) in RCA: 53] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/17/2009] [Revised: 07/29/2009] [Accepted: 08/07/2009] [Indexed: 12/13/2022]
Abstract
Visfatin, ghrelin, and apelin are the most recently identified adipocytokines; but their response to insulin-sensitizing agents is poorly clarified. We aimed to assess the differential effects of either rosiglitazone or metformin monotherapy on the aforementioned adipocytokines in patients with type 2 diabetes mellitus (T2DM). One hundred T2DM patients (30 men, 70 women), with poor glycemic control (glycosylated hemoglobin >6.5%) while taking 850 mg of metformin daily, were enrolled. All participants were randomized to receive either adjunctive therapy with rosiglitazone (8 mg/d, n = 50) or the maximum dose (2550 mg/d) of metformin (MET group, n = 50). Anthropometric parameters, glycemic and lipid profile, high-sensitivity CRP (hs-CRP), insulin resistance (homeostasis model assessment of insulin resistance index [HOMA-IR]), visfatin, ghrelin, and apelin were assessed at baseline and after 14 weeks of therapy. Both rosiglitazone and metformin led to similar, significant improvement in glycemic profile and apelin levels, whereas lipid parameters, fat mass, and visfatin remained almost unaffected (P > .05). Insulin resistance was significantly attenuated in both groups, but to a lesser degree in the MET group (P = .045). Rosiglitazone-treated patients experienced a significant decrease in hs-CRP and systolic blood pressure compared with baseline values and those of the MET group (P < .05). Besides, rosiglitazone treatment considerably increased plasma ghrelin (3.74 +/- 1.52 ng/mL) in comparison with either baseline (P = .034) or metformin monotherapy values (-2.23 +/- 1.87 ng/mL, P = .008). On the other hand, the MET group, rather than the rosiglitazone group, had decreased body mass index (-0.79 +/- 0.47 vs 0.56 kg/m(2), P = .009). The aforementioned changes in apelin and ghrelin were independently associated with HOMA-IR changes. Both rosiglitazone and metformin favorably changed glycemic indexes and apelin levels. The addition of rosiglitazone seemed to confer greater benefits in ghrelin, hs-CRP, systolic blood pressure, and HOMA-IR regulation than metformin monotherapy. Although these results reflect improvement in cardiovascular risk profile, the overall clinical importance of insulin sensitizers must be further assessed.
Collapse
Affiliation(s)
- Nikolaos P E Kadoglou
- First Department of Internal Medicine, Hippokratio General Hospital of Thessaloniki, Greece.
| | | | | | | | | | | | | | | |
Collapse
|
46
|
Toblli JE, Ferrini MG, Cao G, Vernet D, Angerosa M, Gonzalez-Cadavid NF. Antifibrotic effects of pioglitazone on the kidney in a rat model of type 2 diabetes mellitus. Nephrol Dial Transplant 2009; 24:2384-91. [PMID: 19297362 PMCID: PMC2727296 DOI: 10.1093/ndt/gfp103] [Citation(s) in RCA: 53] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2008] [Accepted: 02/19/2009] [Indexed: 11/13/2022] Open
Abstract
BACKGROUND Recent evidence suggests that treatment of type 2 diabetes with thiazolidinediones [peroxisome proliferator-activated receptor-gamma (PPAR-gamma) agonists], ameliorates glomerulosclerosis and tubulointerstitial fibrosis in the rat kidney. In the current work, we have investigated whether these drugs, and specifically pioglitazone (PGT), act by preventing fibrosis and kidney dysfunction mainly through antioxidant and anti-inflammatory effects, independently of glycaemic control. METHODS Male 2- to 3-month-old obese Zucker fa/fa (OZR) and ZDF fa/fa rats (ZDFR), and their control the lean Zucker rat (LZR), were used. Diabetic rats were given either a low dose (0.6 mg/kg/day) or a high dose (12 mg/ kg/day) of PGT in the chow for 2 or 4-5 months. Glycaemia, blood pressure, creatinine clearance and proteinuria were determined, and the underlying histopathology was defined with markers of fibrosis, glomerular damage, oxidative stress and inflammation by immunohistochemistry/ quantitative image analysis in tissue sections, and western blots and ad hoc assays in fresh tissue. RESULTS PGT at low doses given for 4-5 months considerably reduced blood pressure, proteinuria and creatinine clearance. This was associated with amelioration of renal tissue damage and fibrosis, evidenced by the glomerulosclerosis, tubulointerstitial fibrosis, tubular atrophy and podocyte injury indexes, and of oxidative stress and inflammation, as shown by the decrease in the respective markers, although glycaemia remained high and obesity was not affected. CONCLUSIONS These results indicate that low doses of PGT ameliorate renal fibrosis and preserve renal function in this animal model of metabolic syndrome, independently of glycaemic control or effects on body weight.
Collapse
Affiliation(s)
- Jorge E. Toblli
- Laboratory of Experimental Medicine, Hospital Alemán, Buenos Aires, Argentina
| | | | - Gabriel Cao
- Laboratory of Experimental Medicine, Hospital Alemán, Buenos Aires, Argentina
| | | | - Margarita Angerosa
- Laboratory of Experimental Medicine, Hospital Alemán, Buenos Aires, Argentina
| | | |
Collapse
|
47
|
Rizos CV, Elisaf MS, Mikhailidis DP, Liberopoulos EN. How safe is the use of thiazolidinediones in clinical practice? Expert Opin Drug Saf 2009; 8:15-32. [PMID: 19236215 DOI: 10.1517/14740330802597821] [Citation(s) in RCA: 114] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
BACKGROUND Thiazolidinediones (TZDs) are widely used antidiabetic drugs with proven efficacy regarding mainly surrogate markers of diabetes management. However, efficacy on surrogate markers may not always translate into benefits in clinical outcomes. Thiazolidinediones are usually well tolerated; however, their use may be associated with several adverse effects. The first TZD, troglitazone, was withdrawn from the market owing to serious hepatotoxicity. However, this does not seem to be the case with newer TZDs. OBJECTIVE The aim of the present review is to discuss the safety profile of this drug class. METHODS We searched PubMed up to July 2008 using relevant keywords. CONCLUSIONS Common side effects associated with TZDs include edema, weight gain, macular edema and heart failure. Moreover, they may cause hypoglycemia when combined with other antidiabetic drugs as well as decrease hematocrit and hemoglobin levels. Increased bone fracture risk is another TZD-related side effect. Thiazolidinediones tend to increase serum low density lipoprotein cholesterol levels, with rosiglitazone having a more pronounced effect compared with pioglitazone. Moreover, rosiglitazone increases low density lipoprotein particle concentration in contrast to pioglitazone where a decrease is observed. Rosiglitazone has been associated with an increase in myocardial infarction incidence. On the other hand, pioglitazone may reduce cardiovascular events. Overall, TZDs are valuable drugs for diabetes management but physicians should keep in mind that they are associated with several adverse events, the most prominent of which is heart failure.
Collapse
Affiliation(s)
- C V Rizos
- Department of Internal Medicine, University of Ioannina, School of Medicine, Ioannina 45110, Greece.
| | | | | | | |
Collapse
|
48
|
Kaimal R, Song X, Yan B, King R, Deng R. Differential modulation of farnesoid X receptor signaling pathway by the thiazolidinediones. J Pharmacol Exp Ther 2009; 330:125-34. [PMID: 19369578 DOI: 10.1124/jpet.109.151233] [Citation(s) in RCA: 36] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/24/2023] Open
Abstract
Thiazolidinediones (TZD), including troglitazone, rosiglitazone, and pioglitazone, are agonists of peroxisome proliferator-activated receptor (PPAR)-gamma and belong to a class of insulin-sensitizing drugs for type 2 diabetes mellitus. However, member-specific, PPARgamma-independent activities and toxicity have been reported, especially for troglitazone. Currently, the underlying mechanisms are not fully understood. In this study, we demonstrated that troglitazone but not rosiglitazone or pioglitazone modulated expression of farnesoid X receptor (FXR) target genes bile salt export pump (BSEP) and small heterodimer partner (SHP) in Huh-7 cells. More specifically, troglitazone acted as a partial agonist of FXR to weakly increase BSEP and SHP expression but functioned as a potent antagonist to significantly suppress bile acid-induced expression. Consistent with the finding, troglitazone partially induced but markedly antagonized bile acid-mediated BSEP promoter transactivation. However, such modulating effects were not detected with rosiglitazone or pioglitazone. Using the crystal structure of ligand-bound FXR ligand binding domain (LBD), molecular docking predicted that troglitazone, but not rosiglitazone or pioglitazone, could form a stable complex with FXR LBD. The specific alpha-tocopherol side chain of troglitazone significantly contributed to the formation of such a stable complex through extensive interactions with FXR LBD. The docking model was further validated by functional analyses of a series of docking-guided FXR mutants. In summary, the data demonstrated that troglitazone, but not rosiglitazone or pioglitazone, was an FXR modulator and potently antagonized bile acid-induced expression of FXR target genes. Such differential modulation of FXR signaling pathway by TZDs may represent one of the mechanisms for member-specific, PPARgamma-independent activities and toxicity.
Collapse
Affiliation(s)
- Rajani Kaimal
- Department of Biomedical and Pharmaceutical Sciences, Center for Pharmacogenomics and Molecular Therapy, College of Pharmacy, University of Rhode Island, 41 Lower College Rd., Kingston, RI 02881, USA
| | | | | | | | | |
Collapse
|
49
|
Aigner E, Bachofner N, Klein K, De Geyter C, Hohla F, Patsch W, Datz C. Retinol-binding protein 4 in polycystic ovary syndrome--association with steroid hormones and response to pioglitazone treatment. J Clin Endocrinol Metab 2009; 94:1229-35. [PMID: 19158194 DOI: 10.1210/jc.2008-2156] [Citation(s) in RCA: 41] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/26/2023]
Abstract
CONTEXT Polycystic ovary syndrome (PCOS) is frequently associated with insulin resistance. OBJECTIVE The aim of the study was to investigate a putative role of the adipokines retinol-binding protein 4 (RBP4), adiponectin, and visfatin in a cohort of patients with PCOS and their response to treatment with pioglitazone. DESIGN AND SETTING We conducted a randomized, controlled, double-blind study at a tertiary referral center. PATIENTS AND INTERVENTIONS Forty premenopausal women with PCOS were allocated to receive treatment with either pioglitazone (30 mg/d) or a placebo for a period of 3 months. MAIN OUTCOME MEASURES Serum concentrations of RBP4, adiponectin, and visfatin were determined along with metabolic and hormonal parameters before and after treatment. RESULTS Serum adiponectin concentrations were higher after treatment with pioglitazone (P = 0.003), whereas RBP4 levels tended to decrease (P = 0.06), and visfatin concentrations remained unchanged. We found RBP4 serum concentrations at baseline to be positively correlated with serum levels of testosterone (R = 0.446; P = 0.005), 17-OH progesterone (R = 0.345, P = 0.037), and dehydroepiandrosterone sulfate (R = 0.347; P = 0.041). However, these correlations were abolished after treatment with pioglitazone. Patients with high RBP4 levels had significantly higher hirsutism scores (P = 0.038 before and P = 0.034 after treatment). In contrast, serum adiponectin concentrations were related to parameters of impaired glucose metabolism, and no significant associations were detected for visfatin. CONCLUSIONS Our results suggest that RBP4 may contribute to endocrine changes and to the phenotypic manifestation of patients with PCOS because higher RBP4 concentrations are associated with higher androgen levels and higher clinical hirsutism scores independently of pioglitazone treatment. The molecular involvement of RBP4 in human steroid metabolism requires further clarification.
Collapse
Affiliation(s)
- Elmar Aigner
- Department of Internal Medicine, General Hospital Oberndorf/Salzburg, Oberndorf, Austria
| | | | | | | | | | | | | |
Collapse
|
50
|
Kim W, Egan JM. The role of incretins in glucose homeostasis and diabetes treatment. Pharmacol Rev 2008; 60:470-512. [PMID: 19074620 DOI: 10.1124/pr.108.000604] [Citation(s) in RCA: 573] [Impact Index Per Article: 33.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Incretins are gut hormones that are secreted from enteroendocrine cells into the blood within minutes after eating. One of their many physiological roles is to regulate the amount of insulin that is secreted after eating. In this manner, as well as others to be described in this review, their final common raison d'être is to aid in disposal of the products of digestion. There are two incretins, known as glucose-dependent insulinotropic peptide (GIP) and glucagon-like peptide-1 (GLP-1), that share many common actions in the pancreas but have distinct actions outside of the pancreas. Both incretins are rapidly deactivated by an enzyme called dipeptidyl peptidase 4 (DPP4). A lack of secretion of incretins or an increase in their clearance are not pathogenic factors in diabetes. However, in type 2 diabetes (T2DM), GIP no longer modulates glucose-dependent insulin secretion, even at supraphysiological (pharmacological) plasma levels, and therefore GIP incompetence is detrimental to beta-cell function, especially after eating. GLP-1, on the other hand, is still insulinotropic in T2DM, and this has led to the development of compounds that activate the GLP-1 receptor with a view to improving insulin secretion. Since 2005, two new classes of drugs based on incretin action have been approved for lowering blood glucose levels in T2DM: an incretin mimetic (exenatide, which is a potent long-acting agonist of the GLP-1 receptor) and an incretin enhancer (sitagliptin, which is a DPP4 inhibitor). Exenatide is injected subcutaneously twice daily and its use leads to lower blood glucose and higher insulin levels, especially in the fed state. There is glucose-dependency to its insulin secretory capacity, making it unlikely to cause low blood sugars (hypoglycemia). DPP4 inhibitors are orally active and they increase endogenous blood levels of active incretins, thus leading to prolonged incretin action. The elevated levels of GLP-1 are thought to be the mechanism underlying their blood glucose-lowering effects.
Collapse
Affiliation(s)
- Wook Kim
- National Institute on Aging, National Institutes of Health, 5600 Nathan Shock Drive, Baltimore, MD 21224, USA
| | | |
Collapse
|