1
|
Antoniadou C, Gavriilidis E, Ritis K, Tsilingiris D. Anemia in diabetes mellitus: Pathogenetic aspects and the value of early erythropoietin therapy. Metabol Open 2025; 25:100344. [PMID: 39886103 PMCID: PMC11780985 DOI: 10.1016/j.metop.2024.100344] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2024] [Accepted: 12/30/2024] [Indexed: 02/01/2025] Open
Abstract
Anemia is a frequent, yet increasingly recognized, comorbidity in diabetes mellitus (DM), with prevalence often driven by multifactorial mechanisms. Hematinic deficiencies, common in this population, may arise from associated comorbidities or medications, such as metformin, as well as other drugs commonly employed for DM-related conditions. Among contributing factors, diabetic kidney disease (DKD) plays a pivotal role, with anemia developing more frequently and being more pronounced in earlier stages, than in CKD of other causes. This enhanced susceptibility stems primarily from the combined impact of impaired renal oxygen sensing and deficient erythropoietin (EPO) production linked to tubulointerstitial fibrosis. Additional mechanisms comprise glomerular dysfunction, shortened erythrocyte lifespan, uremia-induced bone marrow suppression, and increased bleeding risk. DM is also recognized as a chronic low-grade inflammatory condition, with its inflammatory burden driving iron maldistribution, suppression of erythropoiesis, and resistance to EPO. The diagnostic approach of anemia in DM mirrors that in the general population. Addressing modifiable causes such as hematinic deficiencies, and other chronic conditions, such as DKD and bone marrow disorders, is paramount. In total, the underlying pathophysiology of anemia in DM primarily reflects a state of absolute or relative EPO deficiency and/or diminished bone marrow responsiveness, effectively corresponding to 'anemia of chronic disease. Early initiation of EPO therapy, even in DM patients without overt DKD, may mitigate disease progression and improve outcomes. Future research should focus on diabetes-specific strategies integrating optimal EPO use, potentially implementing targeted management of renal and inflammatory contributors to anemia.
Collapse
Affiliation(s)
- Christina Antoniadou
- First Department of Internal Medicine, University Hospital of Alexandroupolis, Democritus University of Thrace, Alexandroupolis, Greece
- Laboratory of Molecular Hematology, Department of Medicine, Democritus University of Thrace, Alexandroupolis, Greece
| | - Efstratios Gavriilidis
- First Department of Internal Medicine, University Hospital of Alexandroupolis, Democritus University of Thrace, Alexandroupolis, Greece
- Laboratory of Molecular Hematology, Department of Medicine, Democritus University of Thrace, Alexandroupolis, Greece
| | - Konstantinos Ritis
- First Department of Internal Medicine, University Hospital of Alexandroupolis, Democritus University of Thrace, Alexandroupolis, Greece
- Laboratory of Molecular Hematology, Department of Medicine, Democritus University of Thrace, Alexandroupolis, Greece
| | - Dimitrios Tsilingiris
- First Department of Internal Medicine, University Hospital of Alexandroupolis, Democritus University of Thrace, Alexandroupolis, Greece
- Laboratory of Molecular Hematology, Department of Medicine, Democritus University of Thrace, Alexandroupolis, Greece
| |
Collapse
|
2
|
Kounatidis D, Vallianou NG, Rebelos E, Kouveletsou M, Kontrafouri P, Eleftheriadou I, Diakoumopoulou E, Karampela I, Tentolouris N, Dalamaga M. The Many Facets of PPAR-γ Agonism in Obesity and Associated Comorbidities: Benefits, Risks, Challenges, and Future Directions. Curr Obes Rep 2025; 14:19. [PMID: 39934485 DOI: 10.1007/s13679-025-00612-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 02/03/2025] [Indexed: 02/13/2025]
Abstract
PURPOSE OF REVIEW Obesity is strongly associated with cardiometabolic disorders and certain malignancies, emphasizing the key role of adipose tissue in human health. While incretin mimetics have shown effectiveness in glycemic control and weight loss, a holistic strategy for combating obesity and associated comorbidities remains elusive. This review explores peroxisome proliferator-activated receptor gamma (PPAR-γ) agonism as a potential therapeutic approach, highlighting its benefits, addressing its limitations, and outlining future directions for developing more effective treatment strategies. RECENT FINDINGS Both natural and synthetic PPAR-γ agonists hold significant therapeutic potential as insulin sensitizers, while also demonstrating anti-inflammatory properties and playing a critical role in regulating lipid metabolism. However, the clinical use of natural agonists is limited by poor bioavailability, while synthetic agents like thiazolidinediones are associated with adverse effects, including fluid retention, weight gain, and bone loss. Current research is focused on developing modified, tissue-specific PPAR-γ agonists, as well as dual PPAR-α/PPAR-γ agonists, with improved safety profiles to mitigate these side effects. Nanotechnology-based drug delivery systems also hold promise for enhancing bioavailability and therapeutic efficacy. Furthermore, the transformative potential of machine learning and artificial intelligence offers opportunities to accelerate advancements in this field. PPAR-γ agonists exhibit significant potential in addressing metabolic syndrome, cardiovascular disease, and cancer. However, their clinical use is restricted by safety concerns and suboptimal pharmacokinetics. Innovations in modified PPAR-γ agonists, nanotechnology-based delivery systems, and computational tools hold promise for creating safer and more effective therapeutic options for obesity and its associated disorders.
Collapse
Affiliation(s)
- Dimitris Kounatidis
- Diabetes Center, First Department of Propaedeutic Internal Medicine, Laiko General Hospital, Medical School, National and Kapodistrian University of Athens, 11527, Athens, Greece.
| | - Natalia G Vallianou
- First Department of Internal Medicine, Sismanogleio General Hospital, 15126, Athens, Greece
| | - Eleni Rebelos
- Diabetes Center, First Department of Propaedeutic Internal Medicine, Laiko General Hospital, Medical School, National and Kapodistrian University of Athens, 11527, Athens, Greece
| | - Marina Kouveletsou
- Diabetes Center, First Department of Propaedeutic Internal Medicine, Laiko General Hospital, Medical School, National and Kapodistrian University of Athens, 11527, Athens, Greece
| | - Paraskevi Kontrafouri
- Diabetes Center, First Department of Propaedeutic Internal Medicine, Laiko General Hospital, Medical School, National and Kapodistrian University of Athens, 11527, Athens, Greece
| | - Ioanna Eleftheriadou
- Diabetes Center, First Department of Propaedeutic Internal Medicine, Laiko General Hospital, Medical School, National and Kapodistrian University of Athens, 11527, Athens, Greece
| | - Evanthia Diakoumopoulou
- Diabetes Center, First Department of Propaedeutic Internal Medicine, Laiko General Hospital, Medical School, National and Kapodistrian University of Athens, 11527, Athens, Greece
| | - Irene Karampela
- Second Department of Critical Care, Attikon General University Hospital, Medical School, National and Kapodistrian University of Athens, 12462, Athens, Greece
| | - Nikolaos Tentolouris
- Diabetes Center, First Department of Propaedeutic Internal Medicine, Laiko General Hospital, Medical School, National and Kapodistrian University of Athens, 11527, Athens, Greece
| | - Maria Dalamaga
- Department of Biological Chemistry, Medical School, National and Kapodistrian University of Athens, 11527, Athens, Greece
| |
Collapse
|
3
|
Mansoori A, Sahranavard T, Hosseini ZS, Soflaei SS, Emrani N, Nazar E, Gharizadeh M, Khorasanchi Z, Effati S, Ghamsary M, Ferns G, Esmaily H, Mobarhan MG. Prediction of type 2 diabetes mellitus using hematological factors based on machine learning approaches: a cohort study analysis. Sci Rep 2023; 13:663. [PMID: 36635303 PMCID: PMC9837189 DOI: 10.1038/s41598-022-27340-2] [Citation(s) in RCA: 32] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2022] [Accepted: 12/30/2022] [Indexed: 01/13/2023] Open
Abstract
Type 2 Diabetes Mellitus (T2DM) is a significant public health problem globally. The diagnosis and management of diabetes are critical to reduce the diabetes complications including cardiovascular disease and cancer. This study was designed to assess the potential association between T2DM and routinely measured hematological parameters. This study was a subsample of 9000 adults aged 35-65 years recruited as part of Mashhad stroke and heart atherosclerotic disorder (MASHAD) cohort study. Machine learning techniques including logistic regression (LR), decision tree (DT) and bootstrap forest (BF) algorithms were applied to analyze data. All data analyses were performed using SPSS version 22 and SAS JMP Pro version 13 at a significant level of 0.05. Based on the performance indices, the BF model gave high accuracy, precision, specificity, and AUC. Previous studies suggested the positive relationship of triglyceride-glucose (TyG) index with T2DM, so we considered the association of TyG index with hematological factors. We found this association was aligned with their results regarding T2DM, except MCHC. The most effective factors in the BF model were age and WBC (white blood cell). The BF model represented a better performance to predict T2DM. Our model provides valuable information to predict T2DM like age and WBC.
Collapse
Affiliation(s)
- Amin Mansoori
- International UNESCO Center for Health-Related Basic Sciences and Human Nutrition, Mashhad University of Medical Sciences, Mashhad, 99199-91766, Iran
- Department of Applied Mathematics, Ferdowsi University of Mashhad, Mashhad, Iran
- Department of Biostatistics, School of Health, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Toktam Sahranavard
- International UNESCO Center for Health-Related Basic Sciences and Human Nutrition, Mashhad University of Medical Sciences, Mashhad, 99199-91766, Iran
| | | | - Sara Saffar Soflaei
- International UNESCO Center for Health-Related Basic Sciences and Human Nutrition, Mashhad University of Medical Sciences, Mashhad, 99199-91766, Iran
| | - Negar Emrani
- Student Research Committee, School of Medicine, Mashhad University of Medical Science, Mashhad, Iran
| | - Eisa Nazar
- International UNESCO Center for Health-Related Basic Sciences and Human Nutrition, Mashhad University of Medical Sciences, Mashhad, 99199-91766, Iran
- Student Research Committee, Department of Biostatistics, School of Health, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Melika Gharizadeh
- Student Research Committee, School of Paramedical Sciences, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Zahra Khorasanchi
- Student Research Committee, School of Medicine, Mashhad University of Medical Science, Mashhad, Iran
- Department of Nutrition, School of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Sohrab Effati
- Department of Applied Mathematics, Ferdowsi University of Mashhad, Mashhad, Iran
| | - Mark Ghamsary
- School of Public Health, Loma Linda University, Loma Linda, CA, USA
| | - Gordon Ferns
- Division of Medical Education, Brighton and Sussex Medical School, Brighton, UK
| | - Habibollah Esmaily
- Social Determinants of Health Research Center, Mashhad University of Medical Sciences, Mashhad, Iran.
- Department of Biostatistics, School of Health, Mashhad University of Medical Sciences, Mashhad, Iran.
| | - Majid Ghayour Mobarhan
- International UNESCO Center for Health-Related Basic Sciences and Human Nutrition, Mashhad University of Medical Sciences, Mashhad, 99199-91766, Iran.
| |
Collapse
|
4
|
Matsubayashi Y, Yoshida A, Suganami H, Oe M, Sato T, Yaguchi Y, Fujihara K, Yamada T, Tanaka S, Kaku K, Sone H. Predictors of haemoglobin levels and of changes in these levels, focusing on anaemia and polycythaemia after administration of the sodium-glucose cotransporter-2 inhibitor tofogliflozin. Diabetes Obes Metab 2022; 24:2469-2473. [PMID: 35979908 PMCID: PMC9825934 DOI: 10.1111/dom.14836] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/02/2022] [Revised: 08/05/2022] [Accepted: 08/10/2022] [Indexed: 01/11/2023]
Affiliation(s)
- Yasuhiro Matsubayashi
- Department of Hematology, Endocrinology and Metabolism, Faculty of MedicineNiigata UniversityNiigataJapan
| | - Alkihiro Yoshida
- Department of Hematology, Endocrinology and Metabolism, Faculty of MedicineNiigata UniversityNiigataJapan
- Kowa Company, LtdTokyoJapan
| | | | - Momoko Oe
- Department of Hematology, Endocrinology and Metabolism, Faculty of MedicineNiigata UniversityNiigataJapan
- Kowa Company, LtdTokyoJapan
| | - Takaaki Sato
- Department of Hematology, Endocrinology and Metabolism, Faculty of MedicineNiigata UniversityNiigataJapan
| | - Yuta Yaguchi
- Department of Hematology, Endocrinology and Metabolism, Faculty of MedicineNiigata UniversityNiigataJapan
| | - Kazuya Fujihara
- Department of Hematology, Endocrinology and Metabolism, Faculty of MedicineNiigata UniversityNiigataJapan
| | - Takaho Yamada
- Department of Hematology, Endocrinology and Metabolism, Faculty of MedicineNiigata UniversityNiigataJapan
| | - Shiro Tanaka
- Department of Clinical BiostatisticsGraduate School of Medicine Kyoto University
| | | | - Hirohito Sone
- Department of Hematology, Endocrinology and Metabolism, Faculty of MedicineNiigata UniversityNiigataJapan
| |
Collapse
|
5
|
Michalak SS, Wolny-Rokicka E, Nowakowska E, Michalak M, Gil L. Clinical Implications of the Coexistence of Anemia and Diabetes Mellitus in the Elderly Population. J Diabetes Res 2021; 2021:8745968. [PMID: 34708130 PMCID: PMC8545586 DOI: 10.1155/2021/8745968] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/30/2021] [Revised: 08/23/2021] [Accepted: 09/30/2021] [Indexed: 11/18/2022] Open
Abstract
Diabetes mellitus (DM) and also anemia are common in the elderly and have a negative impact on the clinical outcomes of patients. The coexistence of anemia and DM seems to be insufficiently recognized; therefore, the aim of our study is to analyze the incidence and clinical consequences of this coexistence, including mortality, in the population of people aged ≥60. A retrospective study was conducted on 981 primary care clinic patients aged ≥60 during 2013-2014. The prevalence of coexistence of DM and anemia (defined in accordance with WHO) and data on the incidence of comorbidities, hospitalization, medical procedures, and all-cause mortality were analyzed. In the study population, 25% had DM, while 5.4% had both DM and anemia. Peripheral artery disease (PAD) was found in 48 patients (4.89%) of the entire study population, more often in men (p < 0.001). Diabetic patients with anemia compared to nonanemic diabetics had more comorbidities (median 4 (4, 5) vs. 3 (2-4); p < 0.001)-PAD more often (p = 0.004), more hospitalization (median 2 (0-11) vs. 0 (0-11); p < 0.001), and more frequent medical procedures (e.g., percutaneous coronary intervention (p < 0.001), coronary artery bypass surgery (p = 0.027), arteriography (p < 0.001), and bypass surgery or endovascular treatments of lower limb ischemia (p < 0.001)). The cumulative survival of patients with both DM and anemia vs. nonanemic diabetics at 36 months was 86.4% vs. 99.3% (p < 0.001). A multivariate logistic regression model showed anemia to be a significant risk factor for death in diabetic patients (p = 0.013). Patients with both DM and anemia have more comorbidities than nonanemic diabetic patients; they are more often hospitalized, require medical procedures more frequently, and are at a higher risk of death. Effective treatment of anemia in patients with DM is advisable and may well improve the prognosis of patients.
Collapse
Affiliation(s)
- S. S. Michalak
- Department of Pharmacology and Toxicology, Collegium Medicum, University of Zielona Gora, Zielona Gora, Poland
| | - E. Wolny-Rokicka
- Department of Radiotherapy, Multidisciplinary Hospital, Gorzow Wielkopolski, Poland
| | - E. Nowakowska
- Department of Pharmacology and Toxicology, Collegium Medicum, University of Zielona Gora, Zielona Gora, Poland
| | - M. Michalak
- Department of Computer Science and Statistics, Poznan University of Medical Sciences, Poznan, Poland
| | - L. Gil
- Department of Hematology and Bone Marrow Transplantation, Poznan University of Medical Sciences, Poznan, Poland
| |
Collapse
|
6
|
Della Pepa G, Russo M, Vitale M, Carli F, Vetrani C, Masulli M, Riccardi G, Vaccaro O, Gastaldelli A, Rivellese AA, Bozzetto L. Pioglitazone even at low dosage improves NAFLD in type 2 diabetes: clinical and pathophysiological insights from a subgroup of the TOSCA.IT randomised trial. Diabetes Res Clin Pract 2021; 178:108984. [PMID: 34311022 DOI: 10.1016/j.diabres.2021.108984] [Citation(s) in RCA: 51] [Impact Index Per Article: 12.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/09/2021] [Revised: 07/17/2021] [Accepted: 07/21/2021] [Indexed: 02/07/2023]
Abstract
AIMS Non-Alcoholic Fatty Liver Disease (NAFLD) and type 2 diabetes (T2D) share pathophysiological mechanisms and possible therapeutic strategies. We evaluated the effects of 1-year treatment with pioglitazone or sulphonylureas on indirect indices of NAFLD in people with T2D and the role of insulin-resistance and glucotoxicity in determining these effects. METHODS Patients with T2D (n = 195) aged 50-75 years, poorly controlled with metformin 2 g/day, were randomly allocated to add-on pioglitazone (n = 98) or sulphonylureas (n = 97) within the TOSCA.IT trial. Plasma insulin, glucose, and liver enzymes were measured at baseline and after 1-year. Indirect indices of NAFLD (Liver Fat Equation [LFE], Hepatic Steatosis Index [HSI], and Index of NASH [ION]), and insulin resistance (HOMA-IR, Visceral Adiposity Index [VAI] and adipose tissue Insulin Resistance [ADIPO-IR]) were calculated. RESULTS Indices of NAFLD improved after pioglitazone, but not after sulphonylureas; differences between changes (1-year minus baseline) were respectively: -1.76 ± 3.84 vs. 0.28 ± 3.75 for LFE; -1.35 ± 2.78 vs. -0.27 ± 2.63 for HSI; -9.75 ± 43 vs. 3.24 ± 31 for ION; p < 0.05 for all. Indices of insulin resistance decreased after pioglitazone, but not after sulphonylureas: -0.95 ± 4.57 vs. 0.37 ± 3.34 for HOMA-IR, p = 0.032; -1.25 ± 4.11 vs. 1.36 ± 5.43 for ADIPO-IR, p = 0.001; -0.53 ± 1.88 vs. 0.03 ± 2.36 for VAI, p = 0.074. Changes in NAFLD indices were similar with different doses of pioglitazone (15, 30, or 45 mg/day), and were independent of blood glucose control. CONCLUSIONS One-year treatment with pioglitazone even at low dosage significantly improved liver steatosis and inflammation, systemic and adipose tissue insulin resistance in patients with T2D. The beneficial effects of pioglitazone on NAFLD were independent of blood glucose control.
Collapse
Affiliation(s)
- Giuseppe Della Pepa
- Department of Clinical Medicine and Surgery, University of Naples Federico II, Naples, Italy
| | - Marco Russo
- Cardiometabolic Risk Unit, Institute of Clinical Physiology, CNR, Pisa, Italy; University of Siena, Siena, Italy
| | - Marilena Vitale
- Department of Clinical Medicine and Surgery, University of Naples Federico II, Naples, Italy
| | - Fabrizia Carli
- Cardiometabolic Risk Unit, Institute of Clinical Physiology, CNR, Pisa, Italy
| | - Claudia Vetrani
- Department of Clinical Medicine and Surgery, University of Naples Federico II, Naples, Italy
| | - Maria Masulli
- Department of Clinical Medicine and Surgery, University of Naples Federico II, Naples, Italy
| | - Gabriele Riccardi
- Department of Clinical Medicine and Surgery, University of Naples Federico II, Naples, Italy
| | - Olga Vaccaro
- Department of Pharmacy, University of Naples Federico II, Naples, Italy
| | - Amalia Gastaldelli
- Cardiometabolic Risk Unit, Institute of Clinical Physiology, CNR, Pisa, Italy.
| | - Angela A Rivellese
- Department of Clinical Medicine and Surgery, University of Naples Federico II, Naples, Italy.
| | - Lutgarda Bozzetto
- Department of Clinical Medicine and Surgery, University of Naples Federico II, Naples, Italy
| |
Collapse
|
7
|
Lambert J, Saliba J, Calderon C, Sii-Felice K, Salma M, Edmond V, Alvarez JC, Delord M, Marty C, Plo I, Kiladjian JJ, Soler E, Vainchenker W, Villeval JL, Rousselot P, Prost S. PPARγ agonists promote the resolution of myelofibrosis in preclinical models. J Clin Invest 2021; 131:136713. [PMID: 33914703 DOI: 10.1172/jci136713] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2020] [Accepted: 04/22/2021] [Indexed: 12/14/2022] Open
Abstract
Myelofibrosis (MF) is a non-BCR-ABL myeloproliferative neoplasm associated with poor outcomes. Current treatment has little effect on the natural history of the disease. MF results from complex interactions between (a) the malignant clone, (b) an inflammatory context, and (c) remodeling of the bone marrow (BM) microenvironment. Each of these points is a potential target of PPARγ activation. Here, we demonstrated the therapeutic potential of PPARγ agonists in resolving MF in 3 mouse models. We showed that PPARγ agonists reduce myeloproliferation, modulate inflammation, and protect the BM stroma in vitro and ex vivo. Activation of PPARγ constitutes a relevant therapeutic target in MF, and our data support the possibility of using PPARγ agonists in clinical practice.
Collapse
Affiliation(s)
- Juliette Lambert
- Division of Innovative Therapies, CEA/DRF/François Jacob Biology Institute, UMR1184 IMVA-HB/IDMIT, Université Paris-Saclay, Fontenay-aux-Roses, France.,Department of Hematology and Oncology, Centre Hospitalier de Versailles, Le Chesnay, France.,Opale Carnot Institute, Paris, France
| | - Joseph Saliba
- Division of Innovative Therapies, CEA/DRF/François Jacob Biology Institute, UMR1184 IMVA-HB/IDMIT, Université Paris-Saclay, Fontenay-aux-Roses, France
| | - Carolina Calderon
- Division of Innovative Therapies, CEA/DRF/François Jacob Biology Institute, UMR1184 IMVA-HB/IDMIT, Université Paris-Saclay, Fontenay-aux-Roses, France.,Opale Carnot Institute, Paris, France
| | - Karine Sii-Felice
- Division of Innovative Therapies, CEA/DRF/François Jacob Biology Institute, UMR1184 IMVA-HB/IDMIT, Université Paris-Saclay, Fontenay-aux-Roses, France
| | - Mohammad Salma
- Institut de Génétique Moléculaire de Montpellier, University of Montpellier, CNRS, Montpellier, France.,Université de Paris, Laboratory of Excellence GR-Ex, Paris, France
| | - Valérie Edmond
- INSERM, UMR1287, Université Paris-Saclay, Gustave Roussy, Villejuif, France
| | - Jean-Claude Alvarez
- Département de Pharmacologie-Toxicologie, Hôpitaux Universitaires Paris Ile-de-France Ouest, AP-HP, Hôpital Raymond-Poincaré, FHU Sepsis, Garches, France.,MasSpecLab, Plateforme de spectrométrie de masse, INSERM U-1173, Université Paris-Saclay (Versailles Saint-Quentin-en-Yvelines), UFR des sciences de la santé, Montigny-le-Bretonneux, France
| | - Marc Delord
- Recherche Clinique, Centre Hospitalier de Versailles, Le Chesnay, France
| | - Caroline Marty
- INSERM, UMR1287, Université Paris-Saclay, Gustave Roussy, Villejuif, France
| | - Isabelle Plo
- INSERM, UMR1287, Université Paris-Saclay, Gustave Roussy, Villejuif, France
| | - Jean-Jacques Kiladjian
- Opale Carnot Institute, Paris, France.,Université de Paris, AP-HP, Hôpital Saint-Louis, Centre d'Investigations Cliniques CIC 1427, INSERM, Paris, France
| | - Eric Soler
- Institut de Génétique Moléculaire de Montpellier, University of Montpellier, CNRS, Montpellier, France.,Université de Paris, Laboratory of Excellence GR-Ex, Paris, France
| | | | - Jean-Luc Villeval
- INSERM, UMR1287, Université Paris-Saclay, Gustave Roussy, Villejuif, France
| | - Philippe Rousselot
- Division of Innovative Therapies, CEA/DRF/François Jacob Biology Institute, UMR1184 IMVA-HB/IDMIT, Université Paris-Saclay, Fontenay-aux-Roses, France.,Department of Hematology and Oncology, Centre Hospitalier de Versailles, Le Chesnay, France.,Opale Carnot Institute, Paris, France.,Université Paris-Saclay (Versailles Saint-Quentin-en-Yvelines), UFR des sciences de la santé, Montigny-le-Bretonneux, France
| | - Stéphane Prost
- Division of Innovative Therapies, CEA/DRF/François Jacob Biology Institute, UMR1184 IMVA-HB/IDMIT, Université Paris-Saclay, Fontenay-aux-Roses, France.,Opale Carnot Institute, Paris, France
| |
Collapse
|
8
|
Jarrar YB, Jarrar Q, Abaalkhail SJ, Moh'd Kalloush H, Naser W, Zihlif M, Al Shhab M, El Madani A, Jamous Y, Lee SJ. Molecular toxicological alterations in the mouse hearts induced by sub-chronic thiazolidinedione drugs administration. Fundam Clin Pharmacol 2021; 36:143-149. [PMID: 33969534 DOI: 10.1111/fcp.12694] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2021] [Revised: 05/03/2021] [Accepted: 05/05/2021] [Indexed: 12/12/2022]
Abstract
Thiazolidinediones are well-known anti-diabetic drugs. However, they are not widely used due to their cardiotoxic effects. Therefore, in this study, we aimed to determine the molecular toxicological alterations induced in the mouse hearts after thiazolidinedione administration. Balb/c mice received doses clinically equivalent to those given to humans of the most commonly used thiazolidinediones, pioglitazone, and rosiglitazone for 30 days. After that, RNA samples were isolated from the hearts. The mRNA expression of cytochrome (cyp) p450 genes that synthesize the cardiotoxic 20-hydroxyeicosatetraenoic acid (20-HETE) in addition to 92 cardiotoxicity biomarker genes were analyzed using quantitative polymerase chain reaction array technique. The analysis demonstrated that thiazolidinediones caused a significant upregulation (p < 0.5) of the mRNA expression of cyp1a1, cyp4a12, itpr1, ccl7, ccr1, and b2 m genes. In addition, thiazolidinediones caused a significant (p < 0.05) downregulation of the mRNA expression of adra2a, bsn, col15a1, fosl1, Il6, bpifa1, plau, and reg3b genes. The most affected gene was itpr1 gene, which was upregulated by pioglitazone and rosiglitazone by sevenfold and 3.5-fold, respectively. In addition, pioglitazone caused significant upregulation of (p < 0.05) hamp, ppbp, psma2, sik1, timp1, and ucp1 genes, which were not affected significantly (p > 0.05) by rosiglitazone administration. In conclusion, this study showed that thiazolidinediones induce toxicological molecular alterations in the mouse hearts, such as the induction of cyp450s that synthesize 20-HETE, chemokine activation, inflammatory responses, blood clotting, and oxidative stress. These findings may help us understand the mechanism of cardiotoxicity involved in thiazolidinedione administration.
Collapse
Affiliation(s)
| | - Qais Jarrar
- Department of Pharmaceutical Science, Al-Isra'a University, Amman, Jordan
| | - Sara J Abaalkhail
- Department of Pharmacy, Al-Zaytoonah University of Jordan, Amman, Jordan
| | | | - Wisam Naser
- Department of Pharmacy, Al-Zaytoonah University of Jordan, Amman, Jordan
| | - Malek Zihlif
- Department of Pharmacology, Faculty of Medicine, The University of Jordan, Amman, Jordan
| | - Mohammad Al Shhab
- Department of Pharmacology, Faculty of Medicine, The University of Jordan, Amman, Jordan
| | - Abdulla El Madani
- Department of Pharmacy, Al-Zaytoonah University of Jordan, Amman, Jordan
| | - Yahya Jamous
- Nanotechnology Centre, King AbdulAziz City of Science and Technology, Riyadh, Saudi Arabia
| | - Su-Jun Lee
- Department of Pharmacology, College of Medicine, Inje University, Busan, South Korea
| |
Collapse
|
9
|
Yorke E, Barnes N, Akpalu J, Boafo E, Amoah AB. Predictors of anemia in Type 2 diabetes mellitus without renal disease. NIGERIAN JOURNAL OF MEDICINE 2021. [DOI: 10.4103/njm.njm_131_21] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/04/2022] Open
|
10
|
Ida S, Kaneko R, Imataka K, Okubo K, Shirakura Y, Azuma K, Fujiwara R, Murata K. Effects of Antidiabetic Drugs on Muscle Mass in Type 2 Diabetes Mellitus. Curr Diabetes Rev 2021; 17:293-303. [PMID: 32628589 DOI: 10.2174/1573399816666200705210006] [Citation(s) in RCA: 32] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/14/2020] [Revised: 06/16/2020] [Accepted: 06/21/2020] [Indexed: 11/22/2022]
Abstract
BACKGROUND When considering the administration of glucagon-like peptide-1 receptor agonists (GLP-1RAs), sodium-glucose cotransporter-2 (SGLT2) inhibitors, or metformin, it is important to understand their weight loss effect as well as the degree of muscle loss caused by each drug in clinical practice. OBJECTIVE To comparatively examine the effects of GLP-1RAs and oral antidiabetic drugs, including SGLT2 inhibitors and metformin, on muscle mass and body weight in patients with type 2 diabetes via a network meta-analysis of randomized controlled trials. METHODS We included randomized controlled trials evaluating the effects of antidiabetic drugs on muscle mass and body weight. Mean difference (MD) and 95% confidence intervals (CIs) were calculated using a random-effects network meta-analysis. RESULTS Of the studies identified, 18 randomized controlled trials (1, 363 subjects) satisfied the eligibility criteria. In all studies, the effects of these drugs on fat-free mass (FFM) were evaluated. Therefore, FFM, which is used as an alternative index of muscle mass, was included in the study. Semaglutide (MD: -1.68, 95% CI: -2.84 to -0.52), dapagliflozin (-0.53, -0.93 to -0.13), and canagliflozin (-0.90, -1.73 to -0.07) showed a significant decrease in FFM compared with the placebo. Metformin did not show a significant decrease in FFM compared with the placebo. When compared with the placebo, semaglutide, dapagliflozin, ipragliflozin, and canagliflozin showed a significant weight loss. CONCLUSION Although semaglutide, dapaglifrozin, and canagliflozin have a large weight loss effect, it is important to pay attention to muscle loss because a decrease in FFM was observed.
Collapse
Affiliation(s)
- Satoshi Ida
- Department of Diabetes and Metabolism, Ise Red Cross Hospital, Mie, Japan
| | - Ryutaro Kaneko
- Department of Diabetes and Metabolism, Ise Red Cross Hospital, Mie, Japan
| | - Kanako Imataka
- Department of Diabetes and Metabolism, Ise Red Cross Hospital, Mie, Japan
| | - Kaoru Okubo
- Department of Diabetes and Metabolism, Ise Red Cross Hospital, Mie, Japan
| | | | - Kentaro Azuma
- Department of Diabetes and Metabolism, Ise Red Cross Hospital, Mie, Japan
| | - Ryoko Fujiwara
- Department of Diabetes and Metabolism, Ise Red Cross Hospital, Mie, Japan
| | - Kazuya Murata
- Department of Diabetes and Metabolism, Ise Red Cross Hospital, Mie, Japan
| |
Collapse
|
11
|
Abstract
G Protein-coupled receptor 120 (GPR120; fatty acid receptor 4, FFAR4) and PPARγ agonists both lead to anti-inflammatory and insulin sensitizing effects despite signalling through distinct pathways. We recently reported the overarching idea that these two pathways are interactive. Specifically, treatment of obese mice with the PPARγ agonist rosiglitazone (a thiazolidinedione, TZD) in combination with the GPR120 agonist compound A synergistically improves glucose tolerance and insulin sensitivity. We have deconvoluted the mechanisms underlying this feed-forward effect in the study. Taken together, our study shows that low dose TZD administration, in combination with GPR120 agonists, produces additive beneficial effects on glucose tolerance and insulin sensitivity without the undesirable adverse effects of TZD. Our study suggests potential value of combination PPARγ and GPR120 agonists to treat metabolic disease.
Collapse
Affiliation(s)
- Vivian A. Paschoal
- Touchstone Diabetes Center, Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Da Young Oh
- Touchstone Diabetes Center, Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, TX, USA
| |
Collapse
|
12
|
Donnelly LA, Dennis JM, Coleman RL, Sattar N, Hattersley AT, Holman RR, Pearson ER. Risk of Anemia With Metformin Use in Type 2 Diabetes: A MASTERMIND Study. Diabetes Care 2020; 43:2493-2499. [PMID: 32801130 PMCID: PMC7510037 DOI: 10.2337/dc20-1104] [Citation(s) in RCA: 34] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/11/2020] [Accepted: 07/13/2020] [Indexed: 02/03/2023]
Abstract
OBJECTIVE To evaluate the association between metformin use and anemia risk in type 2 diabetes, and the time-course for this, in a randomized controlled trial (RCT) and real-world population data. RESEARCH DESIGN AND METHODS Anemia was defined as a hemoglobin measure of <11 g/dL. In the RCTs A Diabetes Outcome Progression Trial (ADOPT; n = 3,967) and UK Prospective Diabetes Study (UKPDS; n = 1,473), logistic regression was used to model anemia risk and nonlinear mixed models for change in hematological parameters. In the observational Genetics of Diabetes Audit and Research in Tayside Scotland (GoDARTS) population (n = 3,485), discrete-time failure analysis was used to model the effect of cumulative metformin exposure on anemia risk. RESULTS In ADOPT, compared with sulfonylureas, the odds ratio (OR) (95% CI) for anemia was 1.93 (1.10, 3.38) for metformin and 4.18 (2.50, 7.00) for thiazolidinediones. In UKPDS, compared with diet, the OR (95% CI) was 3.40 (1.98, 5.83) for metformin, 0.96 (0.57, 1.62) for sulfonylureas, and 1.08 (0.62, 1.87) for insulin. In ADOPT, hemoglobin and hematocrit dropped after metformin initiation by 6 months, with no further decrease after 3 years. In UKPDS, hemoglobin fell by 3 years in the metformin group compared with other treatments. At years 6 and 9, hemoglobin was reduced in all treatment groups, with no greater difference seen in the metformin group. In GoDARTS, each 1 g/day of metformin use was associated with a 2% higher annual risk of anemia. CONCLUSIONS Metformin use is associated with early risk of anemia in individuals with type 2 diabetes, a finding consistent across two RCTs and replicated in one real-world study. The mechanism for this early fall in hemoglobin is uncertain, but given the time course, is unlikely to be due to vitamin B12 deficiency alone.
Collapse
Affiliation(s)
- Louise A Donnelly
- Population Health & Genomics, School of Medicine, University of Dundee, Dundee, U.K
| | - John M Dennis
- Institute of Biomedical & Clinical Science, University of Exeter Medical School, Royal Devon & Exeter Hospital, Exeter, U.K
| | - Ruth L Coleman
- Institute of Cardiovascular and Medicine Sciences, University of Glasgow, Glasgow, U.K
| | - Naveed Sattar
- Diabetes Trials Unit, Radcliffe Department of Medicine, University of Oxford, Oxford, U.K
| | - Andrew T Hattersley
- Institute of Biomedical & Clinical Science, University of Exeter Medical School, Royal Devon & Exeter Hospital, Exeter, U.K
| | - Rury R Holman
- Institute of Cardiovascular and Medicine Sciences, University of Glasgow, Glasgow, U.K
| | - Ewan R Pearson
- Population Health & Genomics, School of Medicine, University of Dundee, Dundee, U.K.
| |
Collapse
|
13
|
Paschoal VA, Walenta E, Talukdar S, Pessentheiner AR, Osborn O, Hah N, Chi TJ, Tye GL, Armando AM, Evans RM, Chi NW, Quehenberger O, Olefsky JM, Oh DY. Positive Reinforcing Mechanisms between GPR120 and PPARγ Modulate Insulin Sensitivity. Cell Metab 2020; 31:1173-1188.e5. [PMID: 32413335 PMCID: PMC7337476 DOI: 10.1016/j.cmet.2020.04.020] [Citation(s) in RCA: 45] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/25/2020] [Revised: 04/06/2020] [Accepted: 04/27/2020] [Indexed: 12/14/2022]
Abstract
G protein-coupled receptor 120 (GPR120) and PPARγ agonists each have insulin sensitizing effects. But whether these two pathways functionally interact and can be leveraged together to markedly improve insulin resistance has not been explored. Here, we show that treatment with the PPARγ agonist rosiglitazone (Rosi) plus the GPR120 agonist Compound A leads to additive effects to improve glucose tolerance and insulin sensitivity, but at lower doses of Rosi, thus avoiding its known side effects. Mechanistically, we show that GPR120 is a PPARγ target gene in adipocytes, while GPR120 augments PPARγ activity by inducing the endogenous ligand 15d-PGJ2 and by blocking ERK-mediated inhibition of PPARγ. Further, we used macrophage- (MKO) or adipocyte-specific GPR120 KO (AKO) mice to show that GRP120 has anti-inflammatory effects via macrophages while working with PPARγ in adipocytes to increase insulin sensitivity. These results raise the prospect of a safer way to increase insulin sensitization in the clinic.
Collapse
Affiliation(s)
- Vivian A Paschoal
- Touchstone Diabetes Center, Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA; Division of Endocrinology and Metabolism, Department of Medicine, University of California, San Diego, La Jolla, CA 92093, USA
| | - Evelyn Walenta
- Division of Endocrinology and Metabolism, Department of Medicine, University of California, San Diego, La Jolla, CA 92093, USA
| | - Saswata Talukdar
- Division of Endocrinology and Metabolism, Department of Medicine, University of California, San Diego, La Jolla, CA 92093, USA; Merck & Co., Inc., SSF, 630 Gateway Boulevard, South San Francisco, CA 94080, USA
| | - Ariane R Pessentheiner
- Division of Endocrinology and Metabolism, Department of Medicine, University of California, San Diego, La Jolla, CA 92093, USA
| | - Olivia Osborn
- Division of Endocrinology and Metabolism, Department of Medicine, University of California, San Diego, La Jolla, CA 92093, USA
| | - Nasun Hah
- Gene Expression Laboratory, Salk Institute for Biological Studies, La Jolla, CA 92037, USA
| | - Tyler J Chi
- Division of Endocrinology and Metabolism, Department of Medicine, University of California, San Diego, La Jolla, CA 92093, USA
| | - George L Tye
- Division of Endocrinology and Metabolism, Department of Medicine, University of California, San Diego, La Jolla, CA 92093, USA; Case Western Reserve University School of Medicine, 2109 Adelbert Road, Cleveland, OH 44106, USA
| | - Aaron M Armando
- Department of Pharmacology, University of California, San Diego, La Jolla, CA 92093, USA
| | - Ronald M Evans
- Gene Expression Laboratory, Salk Institute for Biological Studies, La Jolla, CA 92037, USA; Howard Hughes Medical Institute, Salk Institute for Biological Studies, La Jolla, CA 92037, USA
| | - Nai-Wen Chi
- Division of Endocrinology and Metabolism, Department of Medicine, University of California, San Diego, La Jolla, CA 92093, USA; VA San Diego Healthcare System, San Diego, CA, USA
| | - Oswald Quehenberger
- Department of Pharmacology, University of California, San Diego, La Jolla, CA 92093, USA
| | - Jerrold M Olefsky
- Division of Endocrinology and Metabolism, Department of Medicine, University of California, San Diego, La Jolla, CA 92093, USA.
| | - Da Young Oh
- Touchstone Diabetes Center, Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA; Division of Endocrinology and Metabolism, Department of Medicine, University of California, San Diego, La Jolla, CA 92093, USA.
| |
Collapse
|
14
|
Wee YH, Anpalahan M. The Role of Older Age in Normocytic Anaemia in Type 2 Diabetes Mellitus. Curr Aging Sci 2019; 12:76-83. [PMID: 31244445 DOI: 10.2174/1874609812666190627154316] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2019] [Revised: 06/05/2019] [Accepted: 06/11/2019] [Indexed: 12/18/2022]
Abstract
BACKGROUND Older age has a significant association with anaemia. However, this has not been adequately investigated in the context of specific comorbidities such as Type 2 Diabetes Mellitus (T2DM). OBJECTIVES To investigate the role of age in Normocytic Anaemia (NCA) and the adverse outcomes of NCA in T2DM. METHODS Patients with NCA, either unexplained or related to Chronic Kidney Disease (CKD), were recruited from a diabetic clinic over six months. Anaemia was defined as a haemoglobin(Hb) < 130g/l for men and <120g/l for women. The relevant data were obtained by interviewing patients and review of medical records. Patients were followed for 12 months for pre-defined adverse outcomes. RESULTS Of the 354 patients assessed, 203 were included (mean age 63.12 ± 13.62 years, males 49.8%). The prevalence of NCA was 24% (49). Older age had a significant univariate association with NCA (p < 0.001) and this remained significant (adjusted Odds Ratio (OR) 1.24, 95% CI 1.16- 5.29) after adjusting for estimated Glomerular Filtration Rate (eGFR) < 60ml/min/1.73m2, albuminuria and other potential confounders. Adjusting for eGFR as a continuous variable also confirmed this significant association (OR1.15, 95% CI 1.10-7.01). In the subgroup of patients aged ≥ 75 years, only older age was significantly associated with NCA. The incidence of all-cause mortality and composite cardiovascular/cerebrovascular events was similar in the anaemic and nonanaemic groups. CONCLUSION NCA is common in T2DM and has a significant association with older age independent of CKD. The anaemia is mild in most patients and appears to have a benign course.
Collapse
Affiliation(s)
- Yen H Wee
- Department of Aged Care and Rehabilitation, General Medicine, Alfred Health, Melbourne, Australia.,Department of General Medicine, Eastern Health, Melbourne, Australia
| | - Mahesan Anpalahan
- Department of General Medicine, Eastern Health, Melbourne, Australia.,Department of Medicine, Monash University, Melbourne, Australia.,Department of Medicine, The University of Melbourne, Melbourne, Australia
| |
Collapse
|
15
|
Abdul-Ghani M, Migahid O, Megahed A, Singh R, Fawaz M, DeFronzo RA, Jayyousi A. Pioglitazone prevents the increase in plasma ketone concentration associated with dapagliflozin in insulin-treated T2DM patients: Results from the Qatar Study. Diabetes Obes Metab 2019; 21:705-709. [PMID: 30259621 DOI: 10.1111/dom.13546] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/18/2018] [Revised: 09/19/2018] [Accepted: 09/23/2018] [Indexed: 11/28/2022]
Abstract
Because of the unique mechanism of action of sodium-glucose co-transport inhibitors (SGLT2i), which is independent of insulin secretion and insulin action, members of this class of drugs effectively lower plasma glucose concentration when used in combination with all other antidiabetic agents, including insulin. Increased plasma ketone concentration has been reported in association with SGLT2i initiation, which, under certain clinical conditions, has developed into diabetic ketoacidosis. The daily insulin dose often is reduced at the time of initiating SGLT2i therapy in insulin-treated patients to avoid hypoglycaemia. However, reduction of insulin dose can increase the risk of ketoacidosis. In the present study, we examined the effect of the addition of dapagliflozin plus pioglitazone on plasma ketone concentration in insulin-treated T2DM patients and compared the results to the effect of dapagliflozin alone. A total of 18 poorly controlled, insulin-treated T2DM participants in the Qatar Study received dapagliflozin (10 mg) plus pioglitazone (30 mg), and 10 poorly controlled non-insulin-treated T2DM patients received dapagliflozin (10 mg) alone for 4 months. Dapagliflozin plus pioglitazone produced a robust decrease in HbA1c (-1.4%) and resulted in a 50% reduction in daily insulin dose, from 133 to 66 units, while dapagliflozin alone caused a 0.8% reduction in HbA1c. Dapagliflozin caused a four-fold increase in fasting plasma ketone concentration, while the combination of pioglitazone plus dapagliflozin was not associated with a significant increase (0.13 vs 0.15 mM) in plasma ketone concentration or in risk of hypoglycaemia. These results demonstrate that the addition of pioglitazone to dapagliflozin prevents the increase in plasma ketone concentration associated with SGLT2i therapy.
Collapse
Affiliation(s)
- Muhammad Abdul-Ghani
- Academic Health System, Hamad General Hospital, Doha, Qatar
- Division of Diabetes, University of Texas Health Science Center at San Antonio, San Antonio, Texas
| | - Osama Migahid
- Academic Health System, Hamad General Hospital, Doha, Qatar
| | - Ayman Megahed
- Academic Health System, Hamad General Hospital, Doha, Qatar
| | - Rajvir Singh
- Academic Health System, Hamad General Hospital, Doha, Qatar
| | - Mohammad Fawaz
- Academic Health System, Hamad General Hospital, Doha, Qatar
| | - Ralph A DeFronzo
- Division of Diabetes, University of Texas Health Science Center at San Antonio, San Antonio, Texas
| | - Amin Jayyousi
- Academic Health System, Hamad General Hospital, Doha, Qatar
| |
Collapse
|
16
|
Rehani PR, Iftikhar H, Nakajima M, Tanaka T, Jabbar Z, Rehani RN. Safety and Mode of Action of Diabetes Medications in comparison with 5-Aminolevulinic Acid (5-ALA). J Diabetes Res 2019; 2019:4267357. [PMID: 31781665 PMCID: PMC6874935 DOI: 10.1155/2019/4267357] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/27/2019] [Accepted: 09/24/2019] [Indexed: 12/12/2022] Open
Abstract
5-Aminolevulinic acid (5-ALA) is a delta amino acid naturally present in every living cell of the human body. 5-ALA is produced in the mitochondria as the first product of the porphyrin synthesis pathway and composes heme; exogenously supplemented 5-ALA helps in upregulating mitochondrial functions. Mitochondrial dysfunction has been associated with the pathophysiology of diabetes mellitus. Thus, in this review, we evaluate the mechanisms of action and adverse effects of common medications used to treat type 2 diabetes mellitus as well as 5-ALA including its mechanism and possible use in diabetes management.
Collapse
|
17
|
Abstract
Background Pioglitazone is a promising compound for treatment of core autism spectrum disorder (ASD) symptoms as it targets multiple relevant pathways, including immune system alterations. Objective This pilot study aimed to elucidate the maximum tolerated dose, safety, preliminary evidence of efficacy, and appropriate outcome measures in autistic children ages 5–12 years old. Methods We conducted a 16-week prospective cohort, single blind, single arm, 2-week placebo run-in, dose-finding study of pioglitazone. Twenty-five participants completed treatment. A modified dose finding method was used to determine safety and dose response among three dose levels: 0.25 mg/kg, 0.5 mg/kg, and 0.75 mg/kg once daily. Results Maximum tolerated dose: there were no serious adverse events (SAEs) and as such the maximum tolerated dose within the range tested was 0.75 mg/Kg once daily. Safety: overall, pioglitazone was well tolerated. Two participants discontinued intervention due to perceived non-efficacy and one due to the inability to tolerate interim blood work. Three participants experienced mild neutropenia. Early evidence of efficacy: statistically significant improvement was observed in social withdrawal, repetitive behaviors, and externalizing behaviors as measured by the Aberrant Behavior Checklist (ABC), Child Yale-Brown Obsessive Compulsive Scale (CY-BOCS), and Repetitive Behavior Scale–Revised (RBS-R). Forty-six percent of those enrolled were deemed to be global responders. Conclusions and relevance Pioglitazone is well-tolerated and shows a potential signal in measures of social withdrawal, repetitive, and externalizing behaviors. Randomized controlled trials using the confirmed dose are warranted. Trial registration ClinicalTrials.gov, NCT01205282. Registration date: September 20, 2010. Electronic supplementary material The online version of this article (10.1186/s13229-018-0241-5) contains supplementary material, which is available to authorized users.
Collapse
|
18
|
Sohn JH, Kim JI, Jeon YG, Park J, Kim JB. Effects of Three Thiazolidinediones on Metabolic Regulation and Cold-Induced Thermogenesis. Mol Cells 2018; 41:900-908. [PMID: 30145862 PMCID: PMC6199571 DOI: 10.14348/molcells.2018.0294] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2018] [Revised: 08/02/2018] [Accepted: 08/03/2018] [Indexed: 12/14/2022] Open
Abstract
Insulin resistance is closely associated with metabolic diseases such as type 2 diabetes, dyslipidemia, hypertension and atherosclerosis. Thiazolidinediones (TZDs) have been developed to ameliorate insulin resistance by activation of peroxisome proliferator-activated receptor (PPAR) γ. Although TZDs are synthetic ligands for PPARγ, metabolic outcomes of each TZD are different. Moreover, there are lack of head-to-head comparative studies among TZDs in the aspect of metabolic outcomes. In this study, we analyzed the effects of three TZDs, including lobeglitazone (Lobe), rosiglitazone (Rosi), and pioglitazone (Pio) on metabolic and thermogenic regulation. In adipocytes, Lobe more potently stimulated adipogenesis and insulin-dependent glucose uptake than Rosi and Pio. In the presence of pro-inflammatory stimuli, Lobe efficiently suppressed expressions of pro-inflammatory genes in macrophages and adipocytes. In obese and diabetic db/db mice, Lobe effectively promoted insulin-stimulated glucose uptake and suppressed pro-inflammatory responses in epididymal white adipose tissue (EAT), leading to improve glucose intolerance. Compared to other two TZDs, Lobe enhanced beige adipocyte formation and thermogenic gene expression in inguinal white adipose tissue (IAT) of lean mice, which would be attributable to cold-induced thermogenesis. Collectively, these comparison data suggest that Lobe could relieve insulin resistance and enhance thermogenesis at low-concentration conditions where Rosi and Pio are less effective.
Collapse
Affiliation(s)
- Jee Hyung Sohn
- National Creative Research Initiatives Center for Adipose Tissue Remodeling, Institute of Molecular Biology and Genetics, Department of Biological Sciences, Seoul National University, Seoul 08826,
Korea
| | - Jong In Kim
- National Creative Research Initiatives Center for Adipose Tissue Remodeling, Institute of Molecular Biology and Genetics, Department of Biological Sciences, Seoul National University, Seoul 08826,
Korea
| | - Yong Geun Jeon
- National Creative Research Initiatives Center for Adipose Tissue Remodeling, Institute of Molecular Biology and Genetics, Department of Biological Sciences, Seoul National University, Seoul 08826,
Korea
| | - Jeu Park
- National Creative Research Initiatives Center for Adipose Tissue Remodeling, Institute of Molecular Biology and Genetics, Department of Biological Sciences, Seoul National University, Seoul 08826,
Korea
| | - Jae Bum Kim
- National Creative Research Initiatives Center for Adipose Tissue Remodeling, Institute of Molecular Biology and Genetics, Department of Biological Sciences, Seoul National University, Seoul 08826,
Korea
| |
Collapse
|
19
|
Sahay M, Kalra S, Badani R, Bantwal G, Bhoraskar A, Das AK, Dhorepatil B, Ghosh S, Jeloka T, Khandelwal D, Latif ZA, Nadkar M, Pathan MF, Saboo B, Sahay R, Shimjee S, Shrestha D, Siyan A, Talukdar SH, Tiwaskar M, Unnikrishnan AG. Diabetes and Anemia: International Diabetes Federation (IDF) - Southeast Asian Region (SEAR) position statement. Diabetes Metab Syndr 2017; 11 Suppl 2:S685-S695. [PMID: 28483426 DOI: 10.1016/j.dsx.2017.04.026] [Citation(s) in RCA: 48] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/14/2017] [Accepted: 04/27/2017] [Indexed: 02/07/2023]
Abstract
Anemia is often associated with diabetes mellitus and is known to intensify the risk of developing diabetes-related microvascular and macrovascular complications. There is paucity in understanding of co-existence of these conditions, especially in Southeast Asian countries. Iron and/or erythropoietin deficiencies are the major causes of anemia in diabetes, and diabetic kidney disease plays a key role. Patients with diabetes need to be screened for anemia along with other risk factors and anemia should be corrected appropriately to improve overall clinical outcomes. This position statement aims to provide a comprehensive overview and an algorithm for appropriate management of anemia in patients with diabetes.
Collapse
Affiliation(s)
| | | | | | | | | | - A K Das
- Pondicherry Institute of Medical Sciences, Puducherry, India
| | | | | | | | | | | | - Milind Nadkar
- Seth G.S. Medical College & KEM Hospital, Mumbai, India
| | | | - Banshi Saboo
- Dia Care - Diabetes Care and Hormone Clinic, Ambawadi, Ahmedabad, India
| | | | | | | | - Ali Siyan
- Maldivian Diabetes Society, Male, Maldives
| | | | | | | |
Collapse
|
20
|
Davidson MA, Mattison DR, Azoulay L, Krewski D. Thiazolidinedione drugs in the treatment of type 2 diabetes mellitus: past, present and future. Crit Rev Toxicol 2017; 48:52-108. [PMID: 28816105 DOI: 10.1080/10408444.2017.1351420] [Citation(s) in RCA: 70] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Thiazolidinedione (TZD) drugs used in the treatment of type 2 diabetes mellitus (T2DM) have proven effective in improving insulin sensitivity, hyperglycemia, and lipid metabolism. Though well tolerated by some patients, their mechanism of action as ligands of peroxisome proliferator-activated receptors (PPARs) results in the activation of several pathways in addition to those responsible for glycemic control and lipid homeostasis. These pathways, which include those related to inflammation, bone formation, and cell proliferation, may lead to adverse health outcomes. As treatment with TZDs has been associated with adverse hepatic, cardiovascular, osteological, and carcinogenic events in some studies, the role of TZDs in the treatment of T2DM continues to be debated. At the same time, new therapeutic roles for TZDs are being investigated, with new forms and isoforms currently in the pre-clinical phase for use in the prevention and treatment of some cancers, inflammatory diseases, and other conditions. The aims of this review are to provide an overview of the mechanism(s) of action of TZDs, a review of their safety for use in the treatment of T2DM, and a perspective on their current and future therapeutic roles.
Collapse
Affiliation(s)
- Melissa A Davidson
- a Faculty of Health Sciences , University of Ottawa , Ottawa , Canada.,b McLaughlin Centre for Population Health Risk Assessment , Ottawa , Canada
| | - Donald R Mattison
- b McLaughlin Centre for Population Health Risk Assessment , Ottawa , Canada.,c Risk Sciences International , Ottawa , Canada
| | - Laurent Azoulay
- d Center for Clinical Epidemiology , Lady Davis Research Institute, Jewish General Hospital , Montreal , Canada.,e Department of Oncology , McGill University , Montreal , Canada
| | - Daniel Krewski
- a Faculty of Health Sciences , University of Ottawa , Ottawa , Canada.,b McLaughlin Centre for Population Health Risk Assessment , Ottawa , Canada.,c Risk Sciences International , Ottawa , Canada.,f Faculty of Medicine , University of Ottawa , Ottawa , Canada
| |
Collapse
|
21
|
Goltsman I, Khoury EE, Winaver J, Abassi Z. Does Thiazolidinedione therapy exacerbate fluid retention in congestive heart failure? Pharmacol Ther 2016; 168:75-97. [PMID: 27598860 DOI: 10.1016/j.pharmthera.2016.09.007] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
The ever-growing global burden of congestive heart failure (CHF) and type 2 diabetes mellitus (T2DM) as well as their co-existence necessitate that anti-diabetic pharmacotherapy will modulate the cardiovascular risk inherent to T2DM while complying with the accompanying restrictions imposed by CHF. The thiazolidinedione (TZD) family of peroxisome proliferator-activated receptor γ (PPARγ) agonists initially provided a promising therapeutic option in T2DM owing to anti-diabetic efficacy combined with pleiotropic beneficial cardiovascular effects. However, the utility of TZDs in T2DM has declined in the past decade, largely due to concomitant adverse effects of fluid retention and edema formation attributed to salt-retaining effects of PPARγ activation on the nephron. Presumably, the latter effects are potentially deleterious in the context of pre-existing fluid retention in CHF. However, despite a considerable body of evidence on mechanisms responsible for TZD-induced fluid retention suggesting that this class of drugs is rightfully prohibited from use in CHF patients, there is a paucity of experimental and clinical studies that investigate the effects of TZDs on salt and water homeostasis in the CHF setting. In an attempt to elucidate whether TZDs actually exacerbate the pre-existing fluid retention in CHF, our review summarizes the pathophysiology of fluid retention in CHF. Moreover, we thoroughly review the available data on TZD-induced fluid retention and proposed mechanisms in animals and patients. Finally, we will present recent studies challenging the common notion that TZDs worsen renal salt and water retention in CHF.
Collapse
Affiliation(s)
- Ilia Goltsman
- Department of Physiology, Biophysics and Systems Biology, The Bruce Rappaport, Rappaport Faculty of Medicine, Technion, Haifa, Israel
| | - Emad E Khoury
- Department of Physiology, Biophysics and Systems Biology, The Bruce Rappaport, Rappaport Faculty of Medicine, Technion, Haifa, Israel
| | - Joseph Winaver
- Department of Physiology, Biophysics and Systems Biology, The Bruce Rappaport, Rappaport Faculty of Medicine, Technion, Haifa, Israel
| | - Zaid Abassi
- Department of Physiology, Biophysics and Systems Biology, The Bruce Rappaport, Rappaport Faculty of Medicine, Technion, Haifa, Israel; Department of Laboratory Medicine, Rambam Human Health Care Campus, Haifa, Israel.
| |
Collapse
|
22
|
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]
|
23
|
Ramot Y, Mastrofrancesco A, Camera E, Desreumaux P, Paus R, Picardo M. The role of PPARγ-mediated signalling in skin biology and pathology: new targets and opportunities for clinical dermatology. Exp Dermatol 2016; 24:245-51. [PMID: 25644500 DOI: 10.1111/exd.12647] [Citation(s) in RCA: 66] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/21/2015] [Indexed: 12/19/2022]
Abstract
Peroxisome proliferator-activated receptors (PPARs) are ligand-activated transcription factors that modulate the expression of multiple different genes involved in the regulation of lipid, glucose and amino acid metabolism. PPARs and cognate ligands also regulate important cellular functions, including cell proliferation and differentiation, as well as inflammatory responses. This includes a role in mediating skin and pilosebaceous unit homoeostasis: PPARs appear to be essential for maintaining skin barrier permeability, inhibit keratinocyte cell growth, promote keratinocyte terminal differentiation and regulate skin inflammation. They also may have protective effects on human hair follicle (HFs) epithelial stem cells, while defects in PPARγ-mediated signalling may promote the death of these stem cells and thus facilitate the development of cicatricial alopecia (lichen planopilaris). Overall, however, selected PPARγ modulators appear to act as hair growth inhibitors that reduce the proliferation and promote apoptosis of hair matrix keratinocytes. The fact that commonly prescribed PPARγ-modulatory drugs of the thiazolidine-2,4-dione class can exhibit a battery of adverse cutaneous effects underscores the importance of distinguishing beneficial from clinically undesired cutaneous activities of PPARγ ligands and to better understand on the molecular level how PPARγ-regulated cutaneous lipid metabolism and PPARγ-mediated signalling impact on human skin physiology and pathology. Surely, the therapeutic potential that endogenous and exogenous PPARγ modulators may possess in selected skin diseases, ranging from chronic inflammatory hyperproliferative dermatoses like psoriasis and atopic dermatitis, via scarring alopecia and acne can only be harnessed if the complexities of PPARγ signalling in human skin and its appendages are systematically dissected.
Collapse
Affiliation(s)
- Yuval Ramot
- Department of Dermatology, Hadassah - Hebrew University Medical Center, Jerusalem, Israel
| | | | | | | | | | | |
Collapse
|
24
|
Youssef J, Badr M. Peroxisome Proliferator-Activated Receptors Features, Functions, and Future. NUCLEAR RECEPTOR RESEARCH 2015. [DOI: 10.11131/2015/101188] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/15/2023] Open
|
25
|
Prost S, Relouzat F, Spentchian M, Ouzegdouh Y, Saliba J, Massonnet G, Beressi JP, Verhoeyen E, Raggueneau V, Maneglier B, Castaigne S, Chomienne C, Chrétien S, Rousselot P, Leboulch P. Erosion of the chronic myeloid leukaemia stem cell pool by PPARγ agonists. Nature 2015; 525:380-3. [PMID: 26331539 DOI: 10.1038/nature15248] [Citation(s) in RCA: 226] [Impact Index Per Article: 22.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2014] [Accepted: 07/28/2015] [Indexed: 12/19/2022]
Abstract
Whether cancer is maintained by a small number of stem cells or is composed of proliferating cells with approximate phenotypic equivalency is a central question in cancer biology. In the stem cell hypothesis, relapse after treatment may occur by failure to eradicate cancer stem cells. Chronic myeloid leukaemia (CML) is quintessential to this hypothesis. CML is a myeloproliferative disorder that results from dysregulated tyrosine kinase activity of the fusion oncoprotein BCR-ABL. During the chronic phase, this sole genetic abnormality (chromosomal translocation Ph(+): t(9;22)(q34;q11)) at the stem cell level causes increased proliferation of myeloid cells without loss of their capacity to differentiate. Without treatment, most patients progress to the blast phase when additional oncogenic mutations result in a fatal acute leukaemia made of proliferating immature cells. Imatinib mesylate and other tyrosine kinase inhibitors (TKIs) that target the kinase activity of BCR-ABL have improved patient survival markedly. However, fewer than 10% of patients reach the stage of complete molecular response (CMR), defined as the point when BCR-ABL transcripts become undetectable in blood cells. Failure to reach CMR results from the inability of TKIs to eradicate quiescent CML leukaemia stem cells (LSCs). Here we show that the residual CML LSC pool can be gradually purged by the glitazones, antidiabetic drugs that are agonists of peroxisome proliferator-activated receptor-γ (PPARγ). We found that activation of PPARγ by the glitazones decreases expression of STAT5 and its downstream targets HIF2α and CITED2, which are key guardians of the quiescence and stemness of CML LSCs. When pioglitazone was given temporarily to three CML patients in chronic residual disease in spite of continuous treatment with imatinib, all of them achieved sustained CMR, up to 4.7 years after withdrawal of pioglitazone. This suggests that clinically relevant cancer eradication may become a generally attainable goal by combination therapy that erodes the cancer stem cell pool.
Collapse
Affiliation(s)
- Stéphane Prost
- CEA, Institute of Emerging Diseases and Innovative Therapies (iMETI), F-92265 Fontenay-aux-Roses, France
| | - Francis Relouzat
- CEA, Institute of Emerging Diseases and Innovative Therapies (iMETI), F-92265 Fontenay-aux-Roses, France
| | - Marc Spentchian
- Département de biologie médicale, Hôpital Mignot, F-78150 Le Chesnay, France
| | - Yasmine Ouzegdouh
- CEA, Institute of Emerging Diseases and Innovative Therapies (iMETI), F-92265 Fontenay-aux-Roses, France
| | - Joseph Saliba
- CEA, Institute of Emerging Diseases and Innovative Therapies (iMETI), F-92265 Fontenay-aux-Roses, France
| | - Gérald Massonnet
- Unité de Biologie Cellulaire, UMR-S-940 Institut Universitaire d'Hématologie, Hôpital Saint Louis, F-75010 Paris, France
| | - Jean-Paul Beressi
- Service d'Endocrinologie et de Diabétologie, Hôpital Mignot, F-78150 Le Chesnay, France
| | - Els Verhoeyen
- CIRI, International Center for Infectiology Research, EVIR team, Inserm, U1111, CNRS, UMR5308, Université de Lyon-1, ENS de Lyon, 69007 Lyon, France.,Inserm, U895, Centre de Médecine Moléculaire (C3M), équipe 3, 06204 Nice, France
| | - Victoria Raggueneau
- Laboratoire d'hématologie, Centre Hospitalier de Versailles, F-78150 Le Chesnay, France
| | - Benjamin Maneglier
- Unité de Pharmacologie, Service de Biologie Médicale, Centre Hospitalier de Versailles, F-78150 Le Chesnay, France
| | - Sylvie Castaigne
- Service d'Hématologie et d'Oncologie, Hôpital Mignot, Université Versailles Saint-Quentin-en-Yvelines, F-78150 Le Chesnay, France
| | - Christine Chomienne
- Unité de Biologie Cellulaire, UMR-S-940 Institut Universitaire d'Hématologie, Hôpital Saint Louis, F-75010 Paris, France
| | - Stany Chrétien
- CEA, Institute of Emerging Diseases and Innovative Therapies (iMETI), F-92265 Fontenay-aux-Roses, France.,Inserm, Institute of Emerging Diseases and Innovative Therapies (iMETI), F-92265 Fontenay-aux-Roses, France
| | - Philippe Rousselot
- Unité de Biologie Cellulaire, UMR-S-940 Institut Universitaire d'Hématologie, Hôpital Saint Louis, F-75010 Paris, France.,Service d'Hématologie et d'Oncologie, Hôpital Mignot, Université Versailles Saint-Quentin-en-Yvelines, F-78150 Le Chesnay, France
| | - Philippe Leboulch
- CEA, Institute of Emerging Diseases and Innovative Therapies (iMETI), F-92265 Fontenay-aux-Roses, France.,Genetics Division, Brigham &Women's Hospital and Harvard Medical School, Boston, Massachussetts 02115, USA.,Hematology Division, Ramathibodi Hospital and Mahidol University, 10400 Bangkok, Thailand
| |
Collapse
|
26
|
|
27
|
Abbasi P, Shamsasenjan K, Movassaghpour Akbari AA, Akbarzadehlaleh P, Dehdilani N, Ejtehadifar M. The Effect of Baicalin as A PPAR Activator on Erythroid Differentiation of CD133(+)Hematopoietic Stem Cells in Umbilical Cord Blood. CELL JOURNAL 2015; 17:15-26. [PMID: 25870831 PMCID: PMC4393663 DOI: 10.22074/cellj.2015.508] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/01/2013] [Accepted: 01/14/2014] [Indexed: 11/04/2022]
Abstract
OBJECTIVE The peroxisome proliferator-activated receptors (PPARs) are a group of nu- clear receptor proteins whose functions as transcription factors regulate gene expres- sions. PPARs play essential roles in the regulation of cellular differentiation, development, and metabolism (carbohydrate, lipid, protein), and tumorigenesis of higher organisms. This study attempts to determine the effect of baicalin, a PPARγ activator, on erythroid differentiation of cluster of differentiation 133(+)(CD133(+)) cord blood hematopoietic stem cells (HSCs). MATERIALS AND METHODS In this experimental study, in order to investigate the effects of the PPARγ agonists baicalin and troglitazone on erythropoiesis, we isolated CD133(+) cells from human umbilical cord blood using the MACS method. Isolated cells were cultured in erythroid-inducing medium with or without various amounts of the two PPARγ activa- tors (baicalin and troglitazone). Erythroid differentiation of CD133(+)cord blood HSCs were assessed using microscopic morphology analysis, flow cytometric analysis of erythroid surface markers transferrin receptor (TfR) and glycophorin A (GPA) and bycolony forming assay. RESULTS Microscopic and flow cytometric analysis revealed the erythroid differentiation of CD133(+)cord blood HSCs under applied erythroid inducing conditions. Our flow cytometric data showed that the TfR and GPA positive cell population diminished significantly in the presence of either troglitazone or baicalin. The suppression of erythroid differentiation in response to PPARγ agonists was dose-dependent. Erythroid colony-forming ability of HSC decreased significantly after treatment with both PPARγ agonists but troglitazone had a markedly greater effect. CONCLUSION Our results have demonstrated that PPARγ agonists modulate erythroid dif- ferentiation of CD133(+)HSCs, and therefore play an important role in regulation of normal erythropoiesis under physiologic conditions. Thus, considering the availability and applica- tion of this herbal remedy for treatment of a wide range of diseases, the inhibitory effect of baicalin on erythropoiesis should be noted.
Collapse
Affiliation(s)
- Parvaneh Abbasi
- Hematology and Oncology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Karim Shamsasenjan
- Hematology and Oncology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran ; Blood Transfusion Research Center, High Institute for Research and Education in Transfusion Medicine, Tehran, Iran
| | | | - Parvin Akbarzadehlaleh
- Department of Pharmaceutical Biotechnology, Faculty of Pharmacy, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Nima Dehdilani
- Blood Transfusion Research Center, High Institute for Research and Education in Transfusion Medicine, Tehran, Iran
| | - Mostafa Ejtehadifar
- Department of Pharmaceutical Biotechnology, Faculty of Pharmacy, Tabriz University of Medical Sciences, Tabriz, Iran
| |
Collapse
|
28
|
Lin KD, Lee MY, Feng CC, Chen BK, Yu ML, Shin SJ. Residual effect of reductions in red blood cell count and haematocrit and haemoglobin levels after 10-month withdrawal of pioglitazone in patients with Type 2 diabetes. Diabet Med 2014; 31:1341-1349. [PMID: 24797920 DOI: 10.1111/dme.12481] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/05/2013] [Revised: 02/24/2014] [Accepted: 04/28/2014] [Indexed: 12/22/2022]
Abstract
AIM To investigate the recovery of thiazolidinedione-induced body weight gain and haematopoietic changes after stopping pioglitazone treatment in patients with Type 2 diabetes. METHODS This retrospective cohort study included 214 patients divided into three groups according to pioglitazone treatment status. The first study arm included patients who received pioglitazone for 38 months then interrupted this for 10 months (pioglitazone-interruption group). The second arm consisted of patients who received pioglitazone throughout the 48 months (pioglitazone-continuous group); the third arm included patients who had never received pioglitazone therapy (control group). RESULTS Red blood cell count and haematocrit and haemoglobin levels decreased significantly, while body weight increased in the two pioglitazone-treated groups as compared with the control group at 38 months. Multivariate regression analysis showed that the reductions in red blood cell count/haemoglobin levels were associated with pioglitazone use. In the pioglitazone-interruption group, no recoveries of red blood cells, or haematocrit or haemoglobin levels were observed after stopping pioglitazone for 10 months compared with the pioglitazone-continuous group, but body weight gain decreased to a level that was significantly lower than that in the pioglitazone-continuous group and did not differ significantly from the control group. CONCLUSION In this study, we observed a reversal of body weight gain but no recoveries in red blood cells or haematocrit or haemoglobin levels after stopping pioglitazone for 10 months in patients treated with pioglitazone for 38 months. This finding should prompt a reconsideration of the sustained effect of thiazolidinediones on the haematopoietic system in patients with Type 2 diabetes.
Collapse
Affiliation(s)
- K-D Lin
- The Graduate Institute of Medicine, College of Medicine, Kaohsiung Medical University, Columbia, SC, USA; Division of Endocrinology and Metabolism, Columbia, SC, USA
| | | | | | | | | | | |
Collapse
|
29
|
Abstract
Type 2 diabetes is caused by insulin resistance coupled with an inability to produce enough insulin to control blood glucose, and thiazolidinediones (TZDs) are the only current antidiabetic agents that function primarily by increasing insulin sensitivity. However, despite clear benefits in glycemic control, this class of drugs has recently fallen into disuse due to concerns over side effects and adverse events. Here we review the clinical data and attempt to balance the benefits and risks of TZD therapy. We also examine potential mechanisms of action for the beneficial and harmful effects of TZDs, mainly via agonism of the nuclear receptor PPARγ. Based on critical appraisal of both preclinical and clinical studies, we discuss the prospect of harnessing the insulin sensitizing effects of PPARγ for more effective, safe, and potentially personalized treatments of type 2 diabetes.
Collapse
Affiliation(s)
- Raymond E Soccio
- Division of Endocrinology, Diabetes, and Metabolism, Department of Medicine, Department of Genetics, and The Institute for Diabetes, Obesity, and Metabolism, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Eric R Chen
- Division of Endocrinology, Diabetes, and Metabolism, Department of Medicine, Department of Genetics, and The Institute for Diabetes, Obesity, and Metabolism, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Mitchell A Lazar
- Division of Endocrinology, Diabetes, and Metabolism, Department of Medicine, Department of Genetics, and The Institute for Diabetes, Obesity, and Metabolism, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA 19104, USA.
| |
Collapse
|
30
|
Trevest K, Treadway H, Hawkins-van der Cingel G, Bailey C, Abdelhafiz AH. Prevalence and determinants of anemia in older people with diabetes attending an outpatient clinic: a cross-sectional audit. Clin Diabetes 2014; 32:158-62. [PMID: 25646941 PMCID: PMC4220597 DOI: 10.2337/diaclin.32.4.158] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/17/2023]
Abstract
This cross-sectional audit investigates the prevalence and determinants of anemia in older people with diabetes attending an outpatient clinic. Anemia was found to be highly prevalent, affecting 59% of patients. Older age and longer duration of diabetes were the main predictors of anemia, whereas the presence of chronic kidney disease was a mediator rather than a direct cause.
Collapse
|
31
|
Angelousi A, Larger E. Anaemia, a common but often unrecognized risk in diabetic patients: a review. DIABETES & METABOLISM 2014; 41:18-27. [PMID: 25043174 DOI: 10.1016/j.diabet.2014.06.001] [Citation(s) in RCA: 53] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Received: 05/28/2014] [Revised: 06/20/2014] [Accepted: 06/21/2014] [Indexed: 12/14/2022]
Abstract
Anaemia in patients with diabetes, both type 1 and type 2, is a frequent clinical finding. The mechanisms of anaemia are multifactorial and often not very well understood. Iatrogenic causes, including oral antidiabetic drugs, ACE inhibitors and ARBs, and renal insufficiency are the major causes of anaemia in patients with type 2 diabetes. In patients with type 1, the cause is often an associated autoimmune disease, and screening for autoimmune gastritis, pernicious anaemia, Hashimoto's thyroiditis, coeliac disease and Addison's disease is recommended. Other rare causes - including G6PD deficiency, microangiopathic haemolytic anaemia and thiamine-responsive megaloblastic anaemia - should be suspected in young patients or when the classical causes are excluded. Early detection and recognition of the cause(s) of anaemia in patients with diabetes could help to prevent other clinical manifestations as well as the complications of diabetes.
Collapse
Affiliation(s)
- A Angelousi
- Service de diabétologie, GH Cochin-Hôtel Dieu, Paris, France; 6, rue Andromachis, 12135 Athens, Greece
| | - E Larger
- Service de diabétologie, GH Cochin-Hôtel Dieu, Paris, France; Université Paris-René-Descartes, Paris, France.
| |
Collapse
|
32
|
Grossman C, Dovrish Z, Koren-Morag N, Bornstein G, Leibowitz A. Diabetes mellitus with normal renal function is associated with anaemia. Diabetes Metab Res Rev 2014; 30:291-6. [PMID: 24170527 DOI: 10.1002/dmrr.2491] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/09/2013] [Revised: 09/22/2013] [Accepted: 10/21/2013] [Indexed: 12/20/2022]
Abstract
BACKGROUND Anaemia is a common complication of diabetes mellitus (DM), usually related to renal failure. There is scarce information as to the levels of haemoglobin (Hb) and the rate of anaemia in diabetic patients with normal renal function. We, therefore, evaluated haemoglobin levels and the rate of anaemia in diabetic subjects with normal renal functions [estimated glomerular filtration rate (eGFR) > 60 mL/min]. METHODS The charts of 9250 subjects who attended the Institute of Periodic Medical Examinations at the Chaim Sheba Medical Center for a routine yearly check-up were reviewed. Four hundred and forty-five subjects with type 2 DM and normal renal function were indentified and compared with those without DM who were routinely examined at the same time. Subjects' electronic records were used to build a biochemical and clinical database. RESULTS Mean haemoglobin levels were lower in subjects with DM than in those without (14.2 vs. 14.7 g/dL, respectively; p < 0.001). Anaemia was observed in 48 (10.8%) subjects in the diabetic group and in only 12 (2.7%) in the nondiabetic group (p < 0.001). Multivariate analysis revealed that age, gender, history of gastrointestinal disease, use of beta blockers, renal function and DM were independent determinants of haemoglobin levels. After adjustment for age, gender, history of gastrointestinal tract diseases and renal function, DM remained a significant determinant of anaemia with an odds ratio of 2.15 (confidence interval: 1.07-4.31). CONCLUSIONS Anaemia is more common in diabetic patients even when eGFR > 60 mL/min.
Collapse
Affiliation(s)
- Chagai Grossman
- Rheumatology Unit, The Chaim Sheba Medical Center, Tel-Hashomer, Israel; Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| | | | | | | | | |
Collapse
|
33
|
Regulation of diet-induced adipose tissue and systemic inflammation by salicylates and pioglitazone. PLoS One 2013; 8:e82847. [PMID: 24376593 PMCID: PMC3871540 DOI: 10.1371/journal.pone.0082847] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2013] [Accepted: 10/29/2013] [Indexed: 11/19/2022] Open
Abstract
It is increasingly accepted that chronic inflammation participates in obesity-induced insulin resistance and type 2 diabetes (T2D). Salicylates and thiazolidinediones (TZDs) both have anti-inflammatory and anti-hyperglycemic properties. The present study compared the effects of these drugs on obesity-induced inflammation in adipose tissue (AT) and AT macrophages (ATMs), as well as the metabolic and immunological phenotypes of the animal models. Both drugs improved high fat diet (HFD)-induced insulin resistance. However, salicylates did not affect AT and ATM inflammation, whereas Pioglitazone improved these parameters. Interestingly, HFD and the drug treatments all modulated systemic inflammation as assessed by changes in circulating immune cell numbers and activation states. HFD increased the numbers of circulating white blood cells, neutrophils, and a pro-inflammatory monocyte subpopulation (Ly6Chi), whereas salicylates and Pioglitazone normalized these cell numbers. The drug treatments also decreased circulating lymphocyte numbers. These data suggest that obesity induces systemic inflammation by regulating circulating immune cell phenotypes and that anti-diabetic interventions suppress systemic inflammation by normalizing circulating immune phenotypes.
Collapse
|
34
|
Abstract
Over the last few years a number of important drugs like rofecoxib, rosiglitazone, gatifloxacin (in diabetics) have lost their position in disease management. The newest controversy revolves around Pioglitazone, a thiozolidindione, which improves insulin sensitivity and is reputed to have cardioprotective actions, but is riddled with several controversies related to weight gain, distal fractures of long bones, recent reports of bladder cancer and others. There are now new groups of drugs, which have been introduced with stringent FDA approval. These include DPP-4 inhibitors, GLP-1 analogues and bromocriptine (old wine in a new bottle). Early in 2013 we are also looking at the launch of another new agent - SGLT-2 inhibitors. These newer agents are associated with not only a significant glucose lowering effect but also positive extra-glycaemic benefits principally in the areas of hypoglycaemia and weight gain. This raises a very important question - do we really need such a controversial agent when such a plethora of agents are available to us with possibly better metabolic profile than pioglitazone? This review addresses this highly contentious area dissecting the pros and cons as we see it.
Collapse
|
35
|
Hughes AD, Park C, March K, Coady E, Khir A, Chaturvedi N, Thom SAM. A randomized placebo controlled double blind crossover study of pioglitazone on left ventricular diastolic function in type 2 diabetes. Int J Cardiol 2013; 167:1329-32. [DOI: 10.1016/j.ijcard.2012.03.179] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/25/2011] [Revised: 03/04/2012] [Accepted: 03/31/2012] [Indexed: 02/05/2023]
|
36
|
Valsamakis G, Lois K, Kumar S, Mastorakos G. Metabolic and other effects of pioglitazone as an add-on therapy to metformin in the treatment of polycystic ovary syndrome (PCOS). Hormones (Athens) 2013; 12:363-78. [PMID: 24121378 DOI: 10.1007/bf03401302] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
Insulin resistance is a key pathogenic defect of the clustered metabolic disturbances seen in polycystic ovary syndrome (PCOS). Metformin is an insulin sensitizer acting in the liver and the peripheral tissues that ameliorates the metabolic and reproductive defects in PCOS. In addition, pioglitazone is an insulin sensitizer used in diabetes mellitus type 2 (T2DM), improving insulin resistance (IR) in adipose tissue and muscles. In T2DM, these drugs are also used as a combined treatment due to their "add-on effect" on insulin resistance. Although the beneficial role of troglitazone (a member of the thiazolidinediones (TZDs) family) in PCOS has been shown in the past, currently only pioglitazone is available in the market. A few small randomized controlled trials have directly compared the effectiveness of pioglitazone in women with PCOS, while there are a limited number of small studies that support the beneficial metabolic add-on effect of pioglitazone on metformin-treated PCOS women as compared to metformin or pioglitazone monotherapy. These findings suggest a potentially promising role for combined pioglitazone/metformin treatment in the management of PCOS in metformin-resistant patients. In view of recent concerns regarding pioglitazone usage and its associated health risk, we aim to compare the pros and cons of each drug regarding their metabolic and other hormonal effects in women with PCOS and to explore the possible beneficial effect of combined therapy in certain cases, taking into consideration the teratogenic effect of pioglitazone. Finally, we discuss the need for a randomized controlled trial that will evaluate the metabolic and other hormonal effects of combined metformin/pioglitazone treatment in PCOS with selective treatment targets.
Collapse
Affiliation(s)
- Georgios Valsamakis
- Warwickshire Institute for the Study of Diabetes, Endocrinology and Metabolism (WISDEM), University Hospitals Coventry and Warwickshire, Warwick Medical School, Coventry, UK, Endocrine Unit, Aretaieion University Hospital, National and Kapodistrian University of Athens Medical School, Athens, Greece
| | | | | | | |
Collapse
|
37
|
Yau H, Rivera K, Lomonaco R, Cusi K. The future of thiazolidinedione therapy in the management of type 2 diabetes mellitus. Curr Diab Rep 2013; 13:329-41. [PMID: 23625197 DOI: 10.1007/s11892-013-0378-8] [Citation(s) in RCA: 93] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Since their approval, thiazolidinediones (TZDs) have been used extensively as insulin-sensitizers for the management of type 2 diabetes mellitus (T2DM). Activation of peroxisomal proliferator-activated receptor gamma (PPARγ) nuclear receptors by TZDs leads to a vast spectrum of metabolic and antiinflammatory effects. In the past decade, clinicians and scientists across the fields of metabolism, diabetes, liver disease (NAFLD), atherosclerosis, inflammation, infertility, and even cancer have had high hopes about the potential for TZDs to treat many of these diseases. However, an increasing awareness about undesirable "off-target" effects of TZDs have made us rethink their role and be more cautious about the long-term benefits and risks related to their use. This review examines the most relevant work on the benefits and risks associated with TZD treatment, with a focus on the only PPARγ agonist currently available (pioglitazone), aiming to offer the reader a balanced overview about the current and future role of TZDs in the management of insulin-resistant states and T2DM.
Collapse
Affiliation(s)
- Hanford Yau
- Division of Endocrinology, Diabetes and Metabolism, Department of Medicine, The University of Florida North Florida/South Georgia Veterans Health System, Gainesville, Florida, USA
| | | | | | | |
Collapse
|
38
|
Sridhar S, Walia R, Sachdeva N, Bhansali A. Effect of pioglitazone on testosterone in eugonadal men with type 2 diabetes mellitus: a randomized double-blind placebo-controlled study. Clin Endocrinol (Oxf) 2013; 78:454-9. [PMID: 22816533 DOI: 10.1111/j.1365-2265.2012.04510.x] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/25/2012] [Revised: 06/19/2012] [Accepted: 07/17/2012] [Indexed: 01/22/2023]
Abstract
OBJECTIVES Pioglitazone is an insulin sensitizer used for the management of type 2 diabetes mellitus (T2DM). It has been shown to reduce testosterone level in patients with polycystic ovarian syndrome. However, its effect on testosterone in men has not been studied. RESEARCH DESIGN AND METHODS A randomized, double-blind, placebo-controlled trial with 6 months follow-up. Fifty (25 in each group) eugonadal men (well virilized and total testosterone ≥ 12 nm) with T2DM, aged 30-55 year and HbA1c of ≤ 7.5% were randomly assigned to receive pioglitazone 30 mg per day or placebo along with existing glimepiride and metformin therapy. RESULTS As compared to placebo, 6 months of pioglitazone therapy in patients with T2DM resulted in significant reduction in mean total testosterone level (16.1 to 14.9 vs 17.1 to 17.0 nm; P = 0.031), calculated free testosterone (P = 0.001) and bioavailable testosterone (P = 0.000) despite significant increase in sex hormone-binding globulin (P = 0.000). Plasma androstenedione (∆(4) ) level increased (1.5 to 1.9 vs 1.7 to 1.7 ng/ml; P = 0.051) following pioglitazone therapy. The decrease in testosterone was independent of change in body weight, body fat and HbA1c. CONCLUSION Pioglitazone therapy significantly decreases total, free and bioavailable testosterone in eugonadal men with T2DM. The effects of these alterations need to be determined by further long-term studies.
Collapse
Affiliation(s)
- Subbiah Sridhar
- Department of Endocrinology, Post Graduate Institute of Medical Education and Research, Chandigarh, India
| | | | | | | |
Collapse
|
39
|
Grunberger G. Will PPAR-γ agonist therapy still have a role in diabetes management in 2013? ACTA ACUST UNITED AC 2013. [DOI: 10.2217/dmt.12.81] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
|
40
|
Abstract
Over the last few years a number of important drugs like rofecoxib, Rosiglitazone, and Gatifloxacin (in diabetics) have lost their position in disease management. The newest controversy revolves around Pioglitazone, a thiozolidindione, which improves insulin sensitivity and is reputed to have cardioprotective actions, but is riddled with several controversies related to weight gain, distal fractures of long bones, recent reports of bladder cancer and others. There are now new groups of drugs, which have been introduced with stringent FDA approval. These include DPP-4 inhibitors, GLP-1 analogues and bromocriptine (old wine in a new bottle). Early in 2013 we are also looking at the launch of another new agent--SGLT-2 inhibitors. These newer agents are associated with not only a significant glucose lowering effect but also positive extra-glycemic benefits principally in the areas of hypoglycemia and weight gain. This raises a very important question--do we really need such a controversial agent when such a plethora of agents are available to us with possibly better metabolic profile than Pioglitazone? This review addresses this highly contentious area dissecting the pros and cons as we see it.
Collapse
|
41
|
Viswanathan V, Mohan V, Subramani P, Parthasarathy N, Subramaniyam G, Manoharan D, Sundaramoorthy C, Gnudi L, Karalliedde J, Viberti G. Effect of spironolactone and amiloride on thiazolidinedione-induced fluid retention in South Indian patients with type 2 diabetes. Clin J Am Soc Nephrol 2012. [PMID: 23184569 DOI: 10.2215/cjn.06330612] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
BACKGROUND AND OBJECTIVES Thiazolidinediones (pioglitazone and rosiglitazone) induce renal epithelial sodium channel (ENaC)-mediated sodium reabsorption, resulting in plasma volume (PV) expansion. Incidence and long-term management of fluid retention induced by thiazolidinediones remain unclear. DESIGN, SETTING, PARTICIPANTS, & MEASUREMENTS In a 4-week run-in period, rosiglitazone, 4 mg twice daily, was added to a background anti-diabetic therapy in 260 South Indian patients with type 2 diabetes mellitus. Patients with PV expansion (absolute reduction in hematocrit in run-in, ≥1.5 percentage points) entered a randomized, placebo-controlled study to evaluate effects of amiloride and spironolactone on attenuating rosiglitazone-induced fluid retention. Primary endpoint was change in hematocrit in each diuretic group versus placebo (control group). RESULTS Of the 260 patients, 70% (n=180) had PV expansion. These 180 patients (70% male; mean age, 47.8 years [range, 30-80 years]) were randomly assigned to rosiglitazone, 4 mg twice daily, plus spironolactone, 50 mg once daily; rosiglitazone, 4 mg twice daily, plus amiloride, 10 mg once daily; or rosiglitazone, 4 mg twice daily, plus placebo for 24 weeks. Hematocrit continued to decrease significantly in control and spironolactone groups (mean absolute change, -1.2 [P=0.01] and -0.7 [P=0.02] percentage points, respectively), suggesting continued PV expansion. No change occurred with amiloride (mean change, 0.0 percentage points). Amiloride, but not spironolactone, was superior to control (mean hematocrit difference [95% confidence interval] relative to control, 1.27 [0.21-2.55] and 0.49 [-0.79-1.77] percentage points [P=0.04 and P=0.61], respectively). CONCLUSIONS Prevalence of rosiglitazone-induced fluid retention in South Indian patients with type 2 diabetes is high. Amiloride, a direct ENaC blocker, but not spironolactone, prevented protracted fluid retention in these patients.
Collapse
Affiliation(s)
- Vijay Viswanathan
- Department of Diabetes, M.V. Hospital for Diabetes and Prof M. Viswanathan Diabetes Research Centre, Chennai, India
| | | | | | | | | | | | | | | | | | | |
Collapse
|
42
|
Pishvaian MJ, Marshall JL, Wagner AJ, Hwang JJ, Malik S, Cotarla I, Deeken JF, He AR, Daniel H, Halim AB, Zahir H, Copigneaux C, Liu K, Beckman RA, Demetri GD. A phase 1 study of efatutazone, an oral peroxisome proliferator-activated receptor gamma agonist, administered to patients with advanced malignancies. Cancer 2012; 118:5403-13. [PMID: 22570147 PMCID: PMC3726261 DOI: 10.1002/cncr.27526] [Citation(s) in RCA: 54] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2011] [Revised: 12/13/2011] [Accepted: 02/09/2012] [Indexed: 01/20/2023]
Abstract
BACKGROUND Efatutazone (CS-7017), a novel peroxisome proliferator-activated receptor gamma (PPARγ) agonist, exerts anticancer activity in preclinical models. The authors conducted a phase 1 study to determine the recommended phase 2 dose, safety, tolerability, and pharmacokinetics of efatutazone. METHODS Patients with advanced solid malignancies and no curative therapeutic options were enrolled to receive a given dose of efatutazone, administered orally (PO) twice daily for 6 weeks, in a 3 + 3 intercohort dose-escalation trial. After the third patient, patients with diabetes mellitus were excluded. Efatutazone dosing continued until disease progression or unacceptable toxicity, with measurement of efatutazone pharmacokinetics and plasma adiponectin levels. RESULTS Thirty-one patients received efatutazone at doses ranging from 0.10 to 1.15 mg PO twice daily. Dose escalation stopped when maximal impact on PPARγ-related biomarkers had been reached before any protocol-defined maximum-tolerated dose level. On the basis of a population pharmacokinetic/pharmacodynamic analysis, the recommended phase 2 dose was 0.5 mg PO twice daily. A majority of patients experienced peripheral edema (53.3%), often requiring diuretics. Three episodes of dose-limiting toxicities, related to fluid retention, were noted in the 0.10-, 0.25-, and 1.15-mg cohorts. Of 31 treated patients, 27 were evaluable for response. A sustained partial response (PR; 690 days on therapy) was observed in a patient with myxoid liposarcoma. Ten additional patients had stable disease (SD) for ≥60 days. Exposures were approximately dose proportional, and adiponectin levels increased after 4 weeks of treatment at all dose levels. Immunohistochemistry of archived specimens demonstrated that PPARγ and retinoid X receptor expression levels were significantly greater in patients with SD for ≥60 days or PR (P = .0079), suggesting a predictive biomarker. CONCLUSIONS Efatutazone demonstrates acceptable tolerability with evidence of disease control in patients with advanced malignancies.
Collapse
Affiliation(s)
- Michael J Pishvaian
- Lombardi Comprehensive Cancer Center, Developmental Therapeutics Program, Georgetown University Medical Center, Washington, DC 20007, USA.
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
43
|
Pini M, Rhodes DH, Castellanos KJ, Cabay RJ, Grady EF, Fantuzzi G. Rosiglitazone improves survival and hastens recovery from pancreatic inflammation in obese mice. PLoS One 2012; 7:e40944. [PMID: 22815875 PMCID: PMC3397967 DOI: 10.1371/journal.pone.0040944] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2012] [Accepted: 06/15/2012] [Indexed: 02/06/2023] Open
Abstract
Obesity increases severity of acute pancreatitis (AP) by unclear mechanisms. We investigated the effect of the PPAR-gamma agonist rosiglitazone (RGZ, 0.01% in the diet) on severity of AP induced by administration of IL-12+ IL-18 in male C57BL6 mice fed a low fat (LFD) or high fat diet (HFD), under the hypothesis that RGZ would reduce disease severity in HFD-fed obese animals. In both LFD and HFD mice without AP, RGZ significantly increased body weight and % fat mass, with significant upregulation of adiponectin and suppression of erythropoiesis. In HFD mice with AP, RGZ significantly increased survival and hastened recovery from pancreatic inflammation, as evaluated by significantly improved pancreatic histology, reduced saponification of visceral adipose tissue and less severe suppression of erythropoiesis at Day 7 post-AP. This was associated with significantly lower circulating and pancreas-associated levels of IL-6, Galectin-3, osteopontin and TIMP-1 in HFD + RGZ mice, particularly at Day 7 post-AP. In LFD mice with AP, RGZ significantly worsened the degree of intrapancreatic acinar and fat necrosis as well as visceral fat saponification, without affecting other parameters of disease severity or inflammation. Induction of AP lead to major suppression of adiponectin levels at Day 7 in both HFD and HFD + RGZ mice. In conclusion, RGZ prevents development of severe AP in obese mice even though it significantly increases adiposity, indicating that obesity can be dissociated from AP severity by improving the metabolic and inflammatory milieu. However, RGZ worsens selective parameters of AP severity in LFD mice.
Collapse
Affiliation(s)
- Maria Pini
- Department of Kinesiology and Nutrition, University of Illinois at Chicago, Chicago, Illinois, United States of America
| | - Davina H. Rhodes
- Department of Kinesiology and Nutrition, University of Illinois at Chicago, Chicago, Illinois, United States of America
| | - Karla J. Castellanos
- Department of Kinesiology and Nutrition, University of Illinois at Chicago, Chicago, Illinois, United States of America
| | - Robert J. Cabay
- Department of Pathology, University of Illinois at Chicago, Chicago, Illinois, United States of America
| | - Eileen F. Grady
- Department of Surgery, University of California San Francisco, San Francisco, California, United States of America
| | - Giamila Fantuzzi
- Department of Kinesiology and Nutrition, University of Illinois at Chicago, Chicago, Illinois, United States of America
- * E-mail:
| |
Collapse
|
44
|
Campeau PM, Astapova O, Martins R, Bergeron J, Couture P, Hegele RA, Leff T, Gagné C. Clinical and molecular characterization of a severe form of partial lipodystrophy expanding the phenotype of PPARγ deficiency. J Lipid Res 2012; 53:1968-78. [PMID: 22750678 DOI: 10.1194/jlr.p025437] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/27/2023] Open
Abstract
Familial partial lipodystrophy (FPLD) is characterized by abnormal fat distribution and a metabolic syndrome with hypertriglyceridemia. We identified a family with a severe form of FPLD3 with never-reported clinical features and a novel mutation affecting the DNA binding domain of PPARγ (E157D). Apart from the lipodystrophy and severe metabolic syndrome, individuals presented musculoskeletal and hematological issues. E157D heterozygotes had a muscular habitus yet displayed muscle weakness and myopathy. Also, E157D heterozygotes presented multiple cytopenias and a susceptibility to autoimmune disease. In vitro studies showed that the E157D mutation does not decrease the receptor's affinity to classical PPAR response elements or its responsiveness to a PPARγ agonist, yet it severely reduces its target gene transcription. Microarray experiments demonstrated a decreased activation of a wide array of genes, including genes involved in the PPAR response, the immune response, hematopoiesis, and metabolism in muscle. In addition, a subset of genes with cryptic PPAR response elements was activated. In summary, we describe a large family with a novel PPARγ mutation, which extends the clinical phenotype of FPLD3 to include muscular, immune, and hematological features. Together, our results support the role of PPARγ in controlling homeostasis of multiple systems beyond lipid metabolism.
Collapse
Affiliation(s)
- Philippe M Campeau
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX, USA.
| | | | | | | | | | | | | | | |
Collapse
|
45
|
Abstract
INTRODUCTION Thiazolidinediones (TZDs) initially showed great promise as unique receptor-mediated oral therapy for type 2 diabetes, but a host of serious side effects, primarily cardiovascular, have limited their utility. It is crucial at this point to perform a risk-benefit analysis to determine what role TZDs should play in our current treatment of type 2 diabetes and where the future of this class of drugs is headed. AREAS COVERED This review provides a comprehensive overview of the literature from 2000 onward reporting the known side effects of rosiglitazone and pioglitazone, with commentary on the quality of the data available, putative mechanism of each side effect and clinical significance. Finally, a perspective on the future of the TZDs as a class is provided. EXPERT OPINION The current TZDs are first-generation, non-specific activators of peroxisome proliferator-activated receptor (PPAR) gamma, resulting in a wide array of deleterious side effects that currently limit their use. However, the development of highly targeted selective PPAR gamma modulators (SPPARγMs) and dual PPAR gamma/alpha agonists is on the horizon.
Collapse
Affiliation(s)
- Jacqueline Kung
- Tufts University, Division of Endocrinology, Diabetes and Metabolism, 800 Washington St #268, Boston, MA 02111, USA.
| | | |
Collapse
|
46
|
Higashide T, Murotani E, Sugiyama K. Cystoid macular edema associated with oral antineoplastic agent s-1 in a patient with diabetic retinopathy. Case Rep Ophthalmol 2012; 3:91-5. [PMID: 22548042 PMCID: PMC3339688 DOI: 10.1159/000337491] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022] Open
Abstract
A 60-year-old man with neovascular glaucoma due to diabetic retinopathy received an intravitreal injection of 1.25 mg bevacizumab (IVB) followed by extensive panretinal photocoagulation in the right eye. The anterior segment neovascularization regressed within 10 days after IVB. One and a half months later, the patient underwent gastrectomy for stage IIIb gastric cancer. Two months later, he was started on S-1 orally (100 mg/day for 48, 26, and 32 consecutive days in the first, second, and third treatment cycle, respectively). The interval between the first and second treatment cycle was 20 days and between the second and third cycle it was 24 days. The patient developed anemia and diarrhea. At the end of the second S-1 cycle, cystoid macular edema developed in the right eye, although diabetic retinopathy and neovascular glaucoma were stable. Macular edema persisted for 5 months despite another IVB, and disappeared 3 months after termination of S-1 therapy. The time course of the magnitude of macular edema correlated well with the severity of anemia. The macular edema was possibly associated with anemia, which is a major side effect of S-1. Further studies are warranted to investigate the relationship between anemia and macular edema in patients with diabetic retinopathy.
Collapse
Affiliation(s)
- Tomomi Higashide
- Department of Ophthalmology and Visual Science, Kanazawa University Graduate School of Medical Science, Kanazawa, Japan
| | | | | |
Collapse
|
47
|
Bolignano D, Zoccali C. Glitazones in chronic kidney disease: potential and concerns. Nutr Metab Cardiovasc Dis 2012; 22:167-175. [PMID: 22364889 DOI: 10.1016/j.numecd.2011.11.005] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/18/2011] [Revised: 11/07/2011] [Accepted: 11/13/2011] [Indexed: 11/17/2022]
Abstract
AIMS Glitazones rank now among the most used hypoglycemic agents in patients with type-2 diabetes. This systematic review focuses on the cardiovascular and renal outcomes in chronic kidney disease (CKD) patients treated with these drugs. DATA SYNTHESIS Data from randomized clinical trials and a meta-analysis indicate that glitazones (particularly rosiglitazone) may increase the risk of myocardial infarction, heart failure and cardiovascular death in type-2 diabetics. Observational studies looking at survival and cardiovascular outcomes in diabetic patients with kidney failure show controversial results. Studies in experimental models and clinical studies suggest that glitazones may have favorable effects on renal disease progression, because these drugs coherently reduce urinary albumin excretion and proteinuria in diabetic and non-diabetic nephropathies. No clinical trial based on clinical end-points like kidney failure has until now tested the effect of glitazones on the evolution of chronic renal failure in these patients. CONCLUSIONS Whether the use of glitazones has a positive or a negative impact upon major cardiovascular and renal outcomes in diabetic patients remains an open, unanswered question. Specific studies are needed to assess the efficacy and safety of glitazones in a high risk population like type-2 diabetics with chronic kidney disease.
Collapse
Affiliation(s)
- D Bolignano
- CNR-IBIM, Clinical Epidemiology and Physiopathology of Renal Diseases and Hypertension of Reggio Calabria, Reggio Calabria, Italy
| | | |
Collapse
|
48
|
Sengupta P, Nandi U, Pal TK. Development of safety profile evaluating pharmacokinetics, pharmacodynamics and toxicity of a combination of pioglitazone and olmesartan medoxomil in Wistar albino rats. Regul Toxicol Pharmacol 2012; 62:7-15. [DOI: 10.1016/j.yrtph.2011.12.008] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2011] [Revised: 12/07/2011] [Accepted: 12/12/2011] [Indexed: 10/14/2022]
|
49
|
Aimaiti A, Wufuer M, Wang YH, Saiyiti M, Cui L, Yusufu A. Can bisphenol A diglycidyl ether (BADGE) administration prevent steroid-induced femoral head osteonecrosis in the early stage? Med Hypotheses 2011; 77:282-5. [PMID: 21620573 DOI: 10.1016/j.mehy.2011.04.036] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2010] [Revised: 04/13/2011] [Accepted: 04/28/2011] [Indexed: 12/01/2022]
Abstract
Steroid-induced osteonecrosis of the femoral head (ONFH) is associated with increase of intraosseous pressure caused by elevating of adipogenesis and fat cell hypertrophy in the bone marrow, which subsequently decreases the blood flow in the femoral head and finally resulting in bone ischemia. The early femoral head-preserving method has mainly focused on the conventional core decompression procedure. However, it only achieves a slight decrease in intra-medullary pressure with limited clinical outcome. The crucial point in prevention is to achieve a thorough decompression of intra-medullary pressure and improvement of microcirculation of the femoral head. Bisphenol-A-diglycidyl ether (BADGE), an antagonism of PPAR-γ(Peroxisome proliferator-activated receptor gamma), has been shown to successfully reverse bone marrow adipogenesis and fat cell hypertrophy, enhances proliferation of osteoblasts, inhibit osteoclastogenesis. Therefore, we hypothesized that BADGE administration may be an appropriate novel method for the prevention of early stage steroid-induced ONFH.
Collapse
Affiliation(s)
- Abudusaimi Aimaiti
- Department of Micro-Reconstructive Surgery, The First Teaching Hospital of Xinjiang Medical University, Urumqi 830054, PR China
| | | | | | | | | | | |
Collapse
|
50
|
Schöndorf T, Musholt PB, Hohberg C, Forst T, Lehmann U, Fuchs W, Löbig M, Müller J, Pfützner A. The fixed combination of pioglitazone and metformin improves biomarkers of platelet function and chronic inflammation in type 2 diabetes patients: results from the PIOfix study. J Diabetes Sci Technol 2011; 5:426-32. [PMID: 21527115 PMCID: PMC3125938 DOI: 10.1177/193229681100500233] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
BACKGROUND Type 2 diabetes mellitus (T2DM) is characterized by a proinflammatory and procoagulant condition. This study investigates the impact of a pioglitazone plus metformin therapy on biomarkers of inflammation and platelet activation in comparison to a treatment with glimepiride plus metformin. METHODS The study was designed as a multicenter, randomized, double-blinded two-arm trial. Patients with T2DM and dyslipidemia under metformin monotherapy with hemoglobin A1c value between 6.5% and 9.0% were eligible for trial participation. Blood was drawn at baseline and after 24 weeks of treatment from patients of five centers. Markers of inflammation and thrombocyte function (soluble CD40 ligand, thromboxane, vWillebrand factor, adhesion molecules, clotting reaction) were evaluated subsequently in a central laboratory. RESULTS A total of 46 patients were included in the final analyses. Mean (± standard deviation) age was 58.5 ± 9.0 years (13 women, 33 men; disease duration 6.3 ± 5.0 years; body mass index 32.0 ± 4.8 kg/m(2)). A total of 25 patients were treated with pioglitazone plus metformin, and 21 patients were in the glimepiride arm. There was a significant decline of E-selectin (-3.7 ± 4.8 ng/ml, p < .001 versus baseline), vWillebrand factor (-19.5 ± 32.0%, p < .05), and high-sensitivity C-reactive protein concentrations (-1.08 ± 0.91 mg/liter, p < .05) in the metformin + pioglitazone arm only (metformin + glimepiride, -0.5 ± 3.4 ng/ml, +1.4 ± 33.2%, + 0.08 ± 0.72 mg/liter, respectively, all not significant). Also, all other surrogate markers for platelet function and inflammation showed slight improvements in the metformin + pioglitazone arm but not in the metformin + glimepiride arm. CONCLUSIONS The fixed metformin + pioglitazone combination treatment showed an overall improvement of laboratory surrogate markers, indicating improvement of platelet function and of chronic systemic inflammation, which was not seen with metformin + glimepiride.
Collapse
Affiliation(s)
- Thomas Schöndorf
- Institute for Clinical Research and DevelopmentMainz, Germany
- University of Cologne, Medical CenterCologne, Germany
- University of Applied SciencesRheinbach, Germany
| | - Petra B Musholt
- Institute for Clinical Research and DevelopmentMainz, Germany
| | - Cloth Hohberg
- Institute for Clinical Research and DevelopmentMainz, Germany
| | - Thomas Forst
- Institute for Clinical Research and DevelopmentMainz, Germany
| | | | | | - Mirjam Löbig
- Institute for Clinical Research and DevelopmentMainz, Germany
| | | | - Andreas Pfützner
- Institute for Clinical Research and DevelopmentMainz, Germany
- University of Applied SciencesBingen, Germany
| |
Collapse
|