1
|
Zhao Y, Yue R. White adipose tissue in type 2 diabetes and the effect of antidiabetic drugs. Diabetol Metab Syndr 2025; 17:116. [PMID: 40186308 PMCID: PMC11969724 DOI: 10.1186/s13098-025-01678-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/20/2024] [Accepted: 03/19/2025] [Indexed: 04/07/2025] Open
Abstract
White adipose tissue (WAT) is highly flexible and was previously considered a passive location for energy storage. Its endocrine function has been established for several years, earning it the title of an "endocrine organ" due to its ability to secrete many adipokines that regulate metabolism. WAT is one of the core tissues that influence insulin sensitivity. Its dysfunction enhances insulin resistance and type 2 diabetes (T2D) progression. However, T2D may cause WAT dysfunction, including changes in distribution, metabolism, adipocyte hypertrophy, inflammation, aging, and adipokines and free fatty acid levels, which may exacerbate insulin resistance. This review used PubMed to search WAT dysfunction in T2D and the effects of these changes on insulin resistance. Additionally, we described and discussed the effects of antidiabetic drugs, including insulin therapy, sulfonylureas, metformin, glucose-like peptide-1 receptor agonists, thiazolidinediones, and sodium-dependent glucose transporters-2 inhibitors, on WAT parameters under T2D conditions.
Collapse
Affiliation(s)
- Yixuan Zhao
- Chengdu University of Traditional Chinese Medicine, Hospital of Chengdu, University of Traditional Chinese Medicine, No. 39 Shi-er-qiao Road, Chengdu, Sichuan Province, 610072, P. R. China
| | - Rensong Yue
- Chengdu University of Traditional Chinese Medicine, Hospital of Chengdu, University of Traditional Chinese Medicine, No. 39 Shi-er-qiao Road, Chengdu, Sichuan Province, 610072, P. R. China.
| |
Collapse
|
2
|
Civelek E, Karaman EF, Özden S, Büyükpınarbaşılı N, Uydeş Doğan BS, Kaleli Durman D. Evaluation of the effects of pioglitazone on perivascular adipose tissue function, properties, and structure in a rat model of type-2 diabetes. Can J Physiol Pharmacol 2025; 103:12-28. [PMID: 39361973 DOI: 10.1139/cjpp-2024-0084] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/05/2024]
Abstract
Perivascular adipose tissue (PVAT) plays an important role in many physiological and pathological processes, such as regulation of vascular tone. The aim of this study is to evaluate the effects of pioglitazone on functional, structural, and biochemical properties of PVAT in an experimental model of type-2 diabetes (T2DM). T2DM was induced by high-fat-diet/low-dose-streptozotocin in rats, and pioglitazone (20 mg/kg/p.o.) was administered for 6 weeks. Changes in biochemical parameters, PVAT-mass, vascular-reactivity in thoracic-aorta, as well as PVAT adipocytokine and PPARG-expression levels, and histopathology were evaluated. Pioglitazone administration improved blood glucose and lipid profiles in T2DM. Pioglitazone did not change the anticontractile effect of PVAT on aortic contractile reactivity and besides, had no influence on endothelium-dependent and -independent relaxation responses. Pioglitazone administration increased PVAT-mass and tumor necrotizing factor-α levels, while adiponectin, leptin, and interleukin-6 levels were unchanged. Also, a prominent increase was observed in PPARG-expression in T2DM-Pio group. Moreover, pioglitazone decreased liver steatosis, aortic wall thickening, and myocardial damage, whereas increased adipocyte size and adiposity in PVAT. Overall, pioglitazone treatment changed the mass and in part the inflammatory profile of PVAT but did not modify vasoreactivity in T2DM. This study provides novel findings in relationship with the adipogenic effect of pioglitazone and PVAT function.
Collapse
MESH Headings
- Animals
- Pioglitazone/pharmacology
- Pioglitazone/therapeutic use
- Diabetes Mellitus, Type 2/drug therapy
- Diabetes Mellitus, Type 2/metabolism
- Diabetes Mellitus, Type 2/pathology
- Diabetes Mellitus, Type 2/physiopathology
- Adipose Tissue/drug effects
- Adipose Tissue/metabolism
- Adipose Tissue/pathology
- Male
- Rats
- Hypoglycemic Agents/pharmacology
- PPAR gamma/metabolism
- Rats, Wistar
- Diabetes Mellitus, Experimental/drug therapy
- Diabetes Mellitus, Experimental/physiopathology
- Diabetes Mellitus, Experimental/metabolism
- Diabetes Mellitus, Experimental/pathology
- Blood Glucose/drug effects
- Blood Glucose/metabolism
- Aorta, Thoracic/drug effects
- Aorta, Thoracic/pathology
- Aorta, Thoracic/metabolism
- Aorta, Thoracic/physiopathology
- Adipokines/metabolism
- Disease Models, Animal
Collapse
Affiliation(s)
- Erkan Civelek
- Istanbul University, Faculty of Pharmacy, Department of Pharmacology, Istanbul, Turkey
- Istanbul University, Graduate School of Health Sciences, Istanbul, Turkey
| | - Ecem Fatma Karaman
- Istanbul University, Faculty of Pharmacy, Department of Pharmaceutical Toxicology, Istanbul, Turkey
- Biruni University, Faculty of Pharmacy, Department of Pharmaceutical Toxicology, Istanbul, Turkey
| | - Sibel Özden
- Istanbul University, Faculty of Pharmacy, Department of Pharmaceutical Toxicology, Istanbul, Turkey
| | - Nur Büyükpınarbaşılı
- Bezmialem Vakif University, Faculty of Medicine, Department of Medical Pathology, Istanbul, Turkey
| | - B Sönmez Uydeş Doğan
- Istanbul University, Faculty of Pharmacy, Department of Pharmacology, Istanbul, Turkey
| | - Deniz Kaleli Durman
- Istanbul University, Faculty of Pharmacy, Department of Pharmacology, Istanbul, Turkey
| |
Collapse
|
3
|
Oo TT, Pratchayasakul W, Chattipakorn K, Siri-Angkul N, Choovuthayakorn J, Charumporn T, Ongnok B, Arunsak B, Chunchai T, Kongkaew A, Songtrai S, Kaewsuwan S, Chattipakorn N, Chattipakorn S. Cyclosorus Terminans Extract Alleviates Neuroinflammation in Insulin Resistant Rats. Mol Neurobiol 2024; 61:4879-4890. [PMID: 38148371 DOI: 10.1007/s12035-023-03883-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2023] [Accepted: 12/12/2023] [Indexed: 12/28/2023]
Abstract
High-fat diet consumption for an extended period causes obesity, systemic metabolic disturbance, and brain insulin resistance, resulting in neuroinflammation. Although the beneficial effect of Cyclosorus terminans extract on obesity-related insulin resistance has been demonstrated, little is known about how it affects neuroinflammation and brain insulin resistance in obese rats. Male Wistar rats were given either a normal diet (ND, n = 6) or a high-fat diet (HFD, n = 24) for a total of 14 weeks. At the beginning of the week, 13 rats in the ND group were given vehicle orally for 2 weeks, while rats on HFD diets were randomized to one of four groups and given either vehicle, 100 mg/kg/day of Cyclosorus terminans extract, 200 mg/kg/day of Cyclosorus terminans extract, or 20 mg/kg/day of pioglitazone orally for 2 weeks. After the experimental period, blood and brain samples were taken to assess metabolic and brain parameters. HFD-fed rats had obesity, systemic and brain insulin resistance, brain inflammation, microglial and astrocyte hyperactivity, and brain necroptosis. Treatment with 200 mg/kg/day of Cyclosorus terminans extract and pioglitazone equally attenuated obesity, insulin resistance, brain insulin dysfunction, and neuroinflammation in insulin resistant rats. Our findings suggest that Cyclosorus terminans extract may hold promise as a therapeutic agent for insulin resistance and neuroinflammation in obese conditions.
Collapse
Affiliation(s)
- Thura Tun Oo
- Neurophysiology Unit, Cardiac Electrophysiology Research and Training Center, Faculty of Medicine, Chiang Mai University, Chiang Mai, Thailand
- Center of Excellence in Cardiac Electrophysiology Research, Chiang Mai University, Chiang Mai, Thailand
| | - Wasana Pratchayasakul
- Neurophysiology Unit, Cardiac Electrophysiology Research and Training Center, Faculty of Medicine, Chiang Mai University, Chiang Mai, Thailand
- Center of Excellence in Cardiac Electrophysiology Research, Chiang Mai University, Chiang Mai, Thailand
- Cardiac Electrophysiology Unit, Department of Physiology, Faculty of Medicine, Chiang Mai University, Chiang Mai, Thailand
| | - Kenneth Chattipakorn
- Neurophysiology Unit, Cardiac Electrophysiology Research and Training Center, Faculty of Medicine, Chiang Mai University, Chiang Mai, Thailand
- Center of Excellence in Cardiac Electrophysiology Research, Chiang Mai University, Chiang Mai, Thailand
| | - Natthapat Siri-Angkul
- Neurophysiology Unit, Cardiac Electrophysiology Research and Training Center, Faculty of Medicine, Chiang Mai University, Chiang Mai, Thailand
- Center of Excellence in Cardiac Electrophysiology Research, Chiang Mai University, Chiang Mai, Thailand
- Cardiac Electrophysiology Unit, Department of Physiology, Faculty of Medicine, Chiang Mai University, Chiang Mai, Thailand
| | - Jirachaya Choovuthayakorn
- Neurophysiology Unit, Cardiac Electrophysiology Research and Training Center, Faculty of Medicine, Chiang Mai University, Chiang Mai, Thailand
- Center of Excellence in Cardiac Electrophysiology Research, Chiang Mai University, Chiang Mai, Thailand
| | - Thanapat Charumporn
- Neurophysiology Unit, Cardiac Electrophysiology Research and Training Center, Faculty of Medicine, Chiang Mai University, Chiang Mai, Thailand
- Center of Excellence in Cardiac Electrophysiology Research, Chiang Mai University, Chiang Mai, Thailand
| | - Benjamin Ongnok
- Neurophysiology Unit, Cardiac Electrophysiology Research and Training Center, Faculty of Medicine, Chiang Mai University, Chiang Mai, Thailand
- Center of Excellence in Cardiac Electrophysiology Research, Chiang Mai University, Chiang Mai, Thailand
- Cardiac Electrophysiology Unit, Department of Physiology, Faculty of Medicine, Chiang Mai University, Chiang Mai, Thailand
| | - Busarin Arunsak
- Neurophysiology Unit, Cardiac Electrophysiology Research and Training Center, Faculty of Medicine, Chiang Mai University, Chiang Mai, Thailand
- Center of Excellence in Cardiac Electrophysiology Research, Chiang Mai University, Chiang Mai, Thailand
| | - Titikorn Chunchai
- Neurophysiology Unit, Cardiac Electrophysiology Research and Training Center, Faculty of Medicine, Chiang Mai University, Chiang Mai, Thailand
- Center of Excellence in Cardiac Electrophysiology Research, Chiang Mai University, Chiang Mai, Thailand
| | - Aphisek Kongkaew
- Research Administration Section, Faculty of Medicine, Chiang Mai University, Chiang Mai, Thailand
| | - Sujinda Songtrai
- Department of Pharmacognosy and Pharmaceutical Botany, Faculty of Pharmaceutical Sciences, Prince of Songkla University, Songkhla, Thailand
- Phytomedicine and Pharmaceutical Biotechnology Excellence Center, Faculty of Pharmaceutical Sciences, Prince of Songkla University, Songkhla, Thailand
| | - Sireewan Kaewsuwan
- Department of Pharmacognosy and Pharmaceutical Botany, Faculty of Pharmaceutical Sciences, Prince of Songkla University, Songkhla, Thailand
- Phytomedicine and Pharmaceutical Biotechnology Excellence Center, Faculty of Pharmaceutical Sciences, Prince of Songkla University, Songkhla, Thailand
| | - Nipon Chattipakorn
- Neurophysiology Unit, Cardiac Electrophysiology Research and Training Center, Faculty of Medicine, Chiang Mai University, Chiang Mai, Thailand
- Center of Excellence in Cardiac Electrophysiology Research, Chiang Mai University, Chiang Mai, Thailand
- Cardiac Electrophysiology Unit, Department of Physiology, Faculty of Medicine, Chiang Mai University, Chiang Mai, Thailand
| | - Siriporn Chattipakorn
- Neurophysiology Unit, Cardiac Electrophysiology Research and Training Center, Faculty of Medicine, Chiang Mai University, Chiang Mai, Thailand.
- Center of Excellence in Cardiac Electrophysiology Research, Chiang Mai University, Chiang Mai, Thailand.
- Department of Oral Biology and Diagnostic Science, Faculty of Dentistry, Chiang Mai University, Chiang Mai, Thailand.
| |
Collapse
|
4
|
Margolis LM, Wilson MA, Drummer DJ, Carrigan CT, Murphy NE, Allen JT, Dawson MA, Mantzoros CS, Young AJ, Pasiakos SM. Pioglitazone does not enhance exogenous glucose oxidation or metabolic clearance rate during aerobic exercise in men under acute high-altitude exposure. Am J Physiol Regul Integr Comp Physiol 2024; 327:R25-R34. [PMID: 38682243 PMCID: PMC11381008 DOI: 10.1152/ajpregu.00064.2024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2024] [Revised: 04/09/2024] [Accepted: 04/23/2024] [Indexed: 05/01/2024]
Abstract
Insulin insensitivity decreases exogenous glucose oxidation and metabolic clearance rate (MCR) during aerobic exercise in unacclimatized lowlanders at high altitude (HA). Whether use of an oral insulin sensitizer before acute HA exposure enhances exogenous glucose oxidation is unclear. This study investigated the impact of pioglitazone (PIO) on exogenous glucose oxidation and glucose turnover compared with placebo (PLA) during aerobic exercise at HA. With the use of a randomized crossover design, native lowlanders (n = 7 males, means ± SD, age: 23 ± 6 yr, body mass: 84 ± 11 kg) consumed 145 g (1.8 g/min) of glucose while performing 80 min of steady-state (1.43 ± 0.16 V̇o2 L/min) treadmill exercise at HA (460 mmHg; [Formula: see text] 96.6 mmHg) following short-term (5 days) use of PIO (15 mg oral dose per day) or PLA (microcrystalline cellulose pill). Substrate oxidation and glucose turnover were determined using indirect calorimetry and stable isotopes ([13C]glucose and 6,6-[2H2]glucose). Exogenous glucose oxidation was not different between PIO (0.31 ± 0.03 g/min) and PLA (0.32 ± 0.09 g/min). Total carbohydrate oxidation (PIO: 1.65 ± 0.22 g/min, PLA: 1.68 ± 0.32 g/min) or fat oxidation (PIO: 0.10 ± 0.0.08 g/min, PLA: 0.09 ± 0.07 g/min) was not different between treatments. There was no treatment effect on glucose rate of appearance (PIO: 2.46 ± 0.27, PLA: 2.43 ± 0.27 mg/kg/min), disappearance (PIO: 2.19 ± 0.17, PLA: 2.20 ± 0.22 mg/kg/min), or MCR (PIO: 1.63 ± 0.37, PLA: 1.73 ± 0.40 mL/kg/min). Results from this study indicate that PIO is not an effective intervention to enhance exogenous glucose oxidation or MCR during acute HA exposure. Lack of effect with PIO suggests that the etiology of glucose metabolism dysregulation during acute HA exposure may not result from insulin resistance in peripheral tissues.NEW & NOTEWORTHY Short-term (5 days) use of the oral insulin sensitizer pioglitazone does not alter circulating glucose or insulin responses to enhance exogenous glucose oxidation during steady-state aerobic exercise in young healthy men under simulated acute (8 h) high-altitude (460 mmHg) conditions. These results indicate that dysregulations in glucose metabolism in native lowlanders sojourning at high altitude may not be due to insulin resistance at peripheral tissue.
Collapse
Affiliation(s)
- Lee M Margolis
- United States Army Research Institute of Environmental Medicine, Natick, Massachusetts, United States
| | - Marques A Wilson
- United States Army Research Institute of Environmental Medicine, Natick, Massachusetts, United States
| | - Devin J Drummer
- United States Army Research Institute of Environmental Medicine, Natick, Massachusetts, United States
- Oak Ridge Institute for Science and Education, Belcamp, Maryland, United States
| | - Christopher T Carrigan
- United States Army Research Institute of Environmental Medicine, Natick, Massachusetts, United States
| | - Nancy E Murphy
- United States Army Research Institute of Environmental Medicine, Natick, Massachusetts, United States
| | - Jillian T Allen
- United States Army Research Institute of Environmental Medicine, Natick, Massachusetts, United States
| | - M Alan Dawson
- United States Army Research Institute of Environmental Medicine, Natick, Massachusetts, United States
| | - Christos S Mantzoros
- Department of Medicine, Beth-Israel Deaconess Medical Center, Harvard Medical School, Boston, Massachusetts, United States
- Department of Medicine, Veterans Affairs Boston Healthcare System, Boston, Massachusetts, United States
| | - Andrew J Young
- United States Army Research Institute of Environmental Medicine, Natick, Massachusetts, United States
- Oak Ridge Institute for Science and Education, Belcamp, Maryland, United States
| | - Stefan M Pasiakos
- Office of Dietary Supplements, National Institutes of Health, Bethesda, Maryland, United States
| |
Collapse
|
5
|
Ertuglu LA, Deger SM, Alsouqi A, Hung A, Gamboa J, Mambungu C, Sha F, Siew E, Abumrad NN, Ikizler TA. A randomized controlled pilot trial of anakinra and pioglitazone for protein metabolism in patients on maintenance haemodialysis. J Cachexia Sarcopenia Muscle 2024; 15:401-411. [PMID: 38178557 PMCID: PMC10834322 DOI: 10.1002/jcsm.13395] [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] [Received: 05/10/2023] [Revised: 09/17/2023] [Accepted: 11/02/2023] [Indexed: 01/06/2024] Open
Abstract
BACKGROUND Chronic inflammation and insulin resistance are highly prevalent in patients on maintenance haemodialysis (MHD) and are strongly associated with protein energy wasting. We conducted a pilot, randomized, placebo-controlled trial of recombinant human interleukin-1 receptor antagonist (IL-1ra) and pioglitazone to explore the safety, feasibility and efficacy for insulin-mediated protein metabolism in patients undergoing MHD. METHODS Twenty-four patients were randomized to receive IL-1ra, pioglitazone or placebo for 12 weeks. Changes in serum inflammatory markers and insulin-mediated protein synthesis, breakdown and net balance in the whole-body and skeletal muscle compartments were assessed using hyperinsulinaemic-hyperaminoacidemic clamp technique at baseline and Week 12. RESULTS Among 24 patients, median (interquartile range) age was 51 (40, 61), 79% were African American and 21% had diabetes mellitus. All patients initiated on intervention completed the study, and no serious adverse events were observed. There was a statistically significant decrease in serum high-sensitivity C-reactive protein in the pioglitazone group compared with placebo, but not in the IL-1ra group. No significant differences in the changes of whole-body or skeletal muscle protein synthesis, breakdown and net balance were found between the groups. CONCLUSIONS In this pilot study, there were no statistically significant effects of 12 weeks of IL-1ra or pioglitazone on protein metabolism in patients on MHD. CLINICALTRIALS gov registration: NCT02278562.
Collapse
Affiliation(s)
- Lale A Ertuglu
- Department of Medicine, Division of Nephrology and Hypertension, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Serpil Muge Deger
- Department of Nephrology, Faculty of Medicine, Dokuz Eylul University, Izmir, Turkey
| | - Aseel Alsouqi
- Department of Medicine, Division of Nephrology and Hypertension, Vanderbilt University Medical Center, Nashville, TN, USA
- Now with Department of Medicine, Division of Hematology and Oncology, University of Pittsburgh Medical Center, Pittsburgh, PA, USA
| | - Adriana Hung
- Department of Medicine, Division of Nephrology and Hypertension, Vanderbilt University Medical Center, Nashville, TN, USA
- Vanderbilt Center Kidney Disease, Vanderbilt University Medical Center, Nashville, TN, USA
- Veterans Administration Tennessee Valley Healthcare System, Nashville, TN, USA
| | - Jorge Gamboa
- Department of Medicine, Division of Clinical Pharmacology, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Cindy Mambungu
- Department of Medicine, Division of Nephrology and Hypertension, Vanderbilt University Medical Center, Nashville, TN, USA
- Vanderbilt Center Kidney Disease, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Feng Sha
- Department of Medicine, Division of Nephrology and Hypertension, Vanderbilt University Medical Center, Nashville, TN, USA
- Vanderbilt Center Kidney Disease, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Edward Siew
- Department of Medicine, Division of Nephrology and Hypertension, Vanderbilt University Medical Center, Nashville, TN, USA
- Vanderbilt Center Kidney Disease, Vanderbilt University Medical Center, Nashville, TN, USA
- Veterans Administration Tennessee Valley Healthcare System, Nashville, TN, USA
| | - Naji N Abumrad
- Department of Surgery, Vanderbilt University Medical Center, Nashville, TN, USA
| | - T Alp Ikizler
- Department of Medicine, Division of Nephrology and Hypertension, Vanderbilt University Medical Center, Nashville, TN, USA
- Vanderbilt Center Kidney Disease, Vanderbilt University Medical Center, Nashville, TN, USA
- Veterans Administration Tennessee Valley Healthcare System, Nashville, TN, USA
| |
Collapse
|
6
|
Tonello R, Silveira Prudente A, Hoon Lee S, Faith Cohen C, Xie W, Paranjpe A, Roh J, Park CK, Chung G, Strong JA, Zhang JM, Berta T. Single-cell analysis of dorsal root ganglia reveals metalloproteinase signaling in satellite glial cells and pain. Brain Behav Immun 2023; 113:401-414. [PMID: 37557960 PMCID: PMC10530626 DOI: 10.1016/j.bbi.2023.08.005] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/07/2023] [Revised: 07/14/2023] [Accepted: 08/06/2023] [Indexed: 08/11/2023] Open
Abstract
Satellite glial cells (SGCs) are among the most abundant non-neuronal cells in dorsal root ganglia (DRGs) and closely envelop sensory neurons that detect painful stimuli. However, little is still known about their homeostatic activities and their contribution to pain. Using single-cell RNA sequencing (scRNA-seq), we were able to obtain a unique transcriptional profile for SGCs. We found enriched expression of the tissue inhibitor metalloproteinase 3 (TIMP3) and other metalloproteinases in SGCs. Small interfering RNA and neutralizing antibody experiments revealed that TIMP3 modulates somatosensory stimuli. TIMP3 expression decreased after paclitaxel treatment, and its rescue by delivery of a recombinant TIMP3 protein reversed and prevented paclitaxel-induced pain. We also established that paclitaxel directly impacts metalloproteinase signaling in cultured SGCs, which may be used to identify potential new treatments for pain. Therefore, our results reveal a metalloproteinase signaling pathway in SGCs for proper processing of somatosensory stimuli and potential discovery of novel pain treatments.
Collapse
Affiliation(s)
- Raquel Tonello
- Pain Research Center, Department of Anesthesiology, University of Cincinnati Medical Center, Cincinnati, OH, USA
| | - Arthur Silveira Prudente
- Pain Research Center, Department of Anesthesiology, University of Cincinnati Medical Center, Cincinnati, OH, USA
| | - Sang Hoon Lee
- Pain Research Center, Department of Anesthesiology, University of Cincinnati Medical Center, Cincinnati, OH, USA
| | - Cinder Faith Cohen
- Pain Research Center, Department of Anesthesiology, University of Cincinnati Medical Center, Cincinnati, OH, USA
| | - Wenrui Xie
- Pain Research Center, Department of Anesthesiology, University of Cincinnati Medical Center, Cincinnati, OH, USA
| | - Aditi Paranjpe
- Bioinformatics Collaborative Services, Division of Biomedical Informatics, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA
| | - Jueun Roh
- Department of Physiology, Gachon Pain Center, College of Medicine, Gachon University, Incheon 21936, Republic of Korea
| | - Chul-Kyu Park
- Department of Physiology, Gachon Pain Center, College of Medicine, Gachon University, Incheon 21936, Republic of Korea
| | - Gehoon Chung
- Department of Oral Physiology, School of Dentistry, Seoul National University, Seoul, Republic of Korea
| | - Judith A Strong
- Pain Research Center, Department of Anesthesiology, University of Cincinnati Medical Center, Cincinnati, OH, USA
| | - Jun-Ming Zhang
- Pain Research Center, Department of Anesthesiology, University of Cincinnati Medical Center, Cincinnati, OH, USA
| | - Temugin Berta
- Pain Research Center, Department of Anesthesiology, University of Cincinnati Medical Center, Cincinnati, OH, USA.
| |
Collapse
|
7
|
Longo S, Rizza S, Federici M. Microbiota-gut-brain axis: relationships among the vagus nerve, gut microbiota, obesity, and diabetes. Acta Diabetol 2023:10.1007/s00592-023-02088-x. [PMID: 37058160 DOI: 10.1007/s00592-023-02088-x] [Citation(s) in RCA: 45] [Impact Index Per Article: 22.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/21/2023] [Accepted: 03/22/2023] [Indexed: 04/15/2023]
Abstract
AIMS The purpose of this review is to explore the interconnected pathways of the microbiota-gut-brain axis (MGBA), focusing on the roles of the vagus nerve and glucagon like peptide-1 in appetite control, and in the development of obesity and diabetes. METHODS Type 2 diabetes mellitus (T2DM) and obesity are metabolic disorders whose prevalence has significantly increased in recent decades and is expected to increase every year, to pandemic proportions. These two pathologies often coexist and have substantial public health implications. The term "diabesity" defines the pathophysiological connection between overweight and T2DM. The gut microbiota affects many aspects of the host. Beyond the regulation of intestinal functions and the activation of immune responses, the gut microbiota plays a role in central nervous system functions (i.e., mood, and psychiatric conditions associated with stress and memory) and is a central regulator of metabolism and appetite. RESULTS The MGBA involves pathways such as the autonomic and enteric nervous systems, the hypothalamic- pituitary-adrenal axis, the immune system, enteroendocrine cells, and microbial metabolites. Notably, the vagus nerve plays an essential role in eating behavior by modulating appetite and learning nutritional preferences. CONCLUSIONS Because of its enteroendocrine cell-mediated interaction with the gut microbiota, the vagus nerve may provide a potential pathway through which gut microorganisms influence host feeding behavior and metabolic control of physiological and pathological conditions.
Collapse
Affiliation(s)
- Susanna Longo
- Department of Systems Medicine, University of Rome Tor Vergata, Via Montpellier 1, 00133, Rome, Italy
| | - Stefano Rizza
- Department of Systems Medicine, University of Rome Tor Vergata, Via Montpellier 1, 00133, Rome, Italy
| | - Massimo Federici
- Department of Systems Medicine, University of Rome Tor Vergata, Via Montpellier 1, 00133, Rome, Italy.
| |
Collapse
|
8
|
McClelland TJ, Fowler AJ, Davies TW, Pearse R, Prowle J, Puthucheary Z. Can pioglitazone be used for optimization of nutrition in critical illness? A systematic review. JPEN J Parenter Enteral Nutr 2023; 47:459-475. [PMID: 36700419 DOI: 10.1002/jpen.2481] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2022] [Revised: 01/13/2023] [Accepted: 01/23/2023] [Indexed: 01/27/2023]
Abstract
BACKGROUND Skeletal muscle wasting is a determinant of physical disability in survivors of critical illness. Intramuscular bioenergetic failure, altered substrate metabolim, and inflammation are likely underpinning mechanisms. We examined the effect of pioglitazone, a peroxisome proliferator-activated receptor γ agonist, on muscle-related outcomes in adults. METHODS We included randomized controlled trials in which pioglitazone was administered (no dose/dosage restrictions) and muscle-related outcomes were reported. We searched MEDLINE, CENTRAL, EMBASE, CINAHL, and trial registries. Risk of bias was assessed using RoB 2. Primary outcomes were physical function and symptoms, muscle mass and function, or body composition and muscular compositional change. Secondary outcomes included muscle insulin sensitivity, mitochondrial effects, and intramuscular inflammation. RESULTS Fourteen studies over 19 publications (n = 474 patients) were included. Lean body mass was unaffected in three studies (n = 126) and increased by 1.8-1.92 kg in two studies (P = 0.02 and 0.003, respectively; n = 48). Pioglitazone was associated with increased peripheral insulin sensitivity (+23%-72%, standardized mean difference of 0.97 from trial start point to end point [95% CI, 0.36-1.58; n = 213]). Treatment reduced intramuscular tumor necrosis factor-α (TNF-α) levels (-30%; P = 0.02; n = 29), with mixed effects on serum TNF-α and intramyocellular lipid concentrations. Treatment increased intramuscular markers of adenosine triphosphate (ATP) biosynthesis (ATP5A [+33%, P ≤ 0.05], ETFA [+60%, P ≤ 0.05], and CX6B1 [+ 33%, P = 0.01] [n = 24]), PGC1α and PGC1β messenger RNA expression (P < 0.05; n = 26), and AMPK phosphorylation (+38%, P < 0.05; n = 26). These data have low-quality evidence profiles owing to risk of bias. CONCLUSIONS Pioglitazone therapy increases skeletal muscle insulin sensitivity and can decrease intramuscular inflammation.
Collapse
Affiliation(s)
- Thomas J McClelland
- William Harvey Research Institute, Barts and The London School of Medicine & Dentistry, Queen Mary University of London, London, UK
| | - Alexander J Fowler
- William Harvey Research Institute, Barts and The London School of Medicine & Dentistry, Queen Mary University of London, London, UK.,Adult Critical Care Unit, Royal London Hospital, London, UK
| | - Thomas W Davies
- William Harvey Research Institute, Barts and The London School of Medicine & Dentistry, Queen Mary University of London, London, UK.,Adult Critical Care Unit, Royal London Hospital, London, UK
| | - Rupert Pearse
- William Harvey Research Institute, Barts and The London School of Medicine & Dentistry, Queen Mary University of London, London, UK.,Adult Critical Care Unit, Royal London Hospital, London, UK
| | - John Prowle
- William Harvey Research Institute, Barts and The London School of Medicine & Dentistry, Queen Mary University of London, London, UK.,Adult Critical Care Unit, Royal London Hospital, London, UK
| | - Zudin Puthucheary
- William Harvey Research Institute, Barts and The London School of Medicine & Dentistry, Queen Mary University of London, London, UK.,Adult Critical Care Unit, Royal London Hospital, London, UK
| |
Collapse
|
9
|
Jotwani ML, Wu Z, Lunde CE, Sieberg CB. The missing mechanistic link: Improving behavioral treatment efficacy for pediatric chronic pain. FRONTIERS IN PAIN RESEARCH 2022; 3:1022699. [PMID: 36313218 PMCID: PMC9614027 DOI: 10.3389/fpain.2022.1022699] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2022] [Accepted: 09/26/2022] [Indexed: 11/07/2022] Open
Abstract
Pediatric chronic pain is a significant global issue, with biopsychosocial factors contributing to the complexity of the condition. Studies have explored behavioral treatments for pediatric chronic pain, but these treatments have mixed efficacy for improving functional and psychological outcomes. Furthermore, the literature lacks an understanding of the biobehavioral mechanisms contributing to pediatric chronic pain treatment response. In this mini review, we focus on how neuroimaging has been used to identify biobehavioral mechanisms of different conditions and how this modality can be used in mechanistic clinical trials to identify markers of treatment response for pediatric chronic pain. We propose that mechanistic clinical trials, utilizing neuroimaging, are warranted to investigate how to optimize the efficacy of behavioral treatments for pediatric chronic pain patients across pain types and ages.
Collapse
Affiliation(s)
- Maya L. Jotwani
- Department of Psychiatry and Behavioral Sciences, Biobehavioral Pain Innovations Lab, Boston Children's Hospital, Boston, MA, United States
- Pain and Affective Neuroscience Center, Department of Anesthesiology, Critical Care, Pain Medicine, Boston Children's Hospital, Boston, MA, United States
| | - Ziyan Wu
- Department of Psychiatry and Behavioral Sciences, Biobehavioral Pain Innovations Lab, Boston Children's Hospital, Boston, MA, United States
- Pain and Affective Neuroscience Center, Department of Anesthesiology, Critical Care, Pain Medicine, Boston Children's Hospital, Boston, MA, United States
- Department of Psychiatry, Harvard Medical School, Boston, MA, United States
| | - Claire E. Lunde
- Department of Psychiatry and Behavioral Sciences, Biobehavioral Pain Innovations Lab, Boston Children's Hospital, Boston, MA, United States
- Pain and Affective Neuroscience Center, Department of Anesthesiology, Critical Care, Pain Medicine, Boston Children's Hospital, Boston, MA, United States
- Nuffield Department of Women's and Reproductive Health, Medical Sciences Division, University of Oxford, Oxford, United Kingdom
| | - Christine B. Sieberg
- Department of Psychiatry and Behavioral Sciences, Biobehavioral Pain Innovations Lab, Boston Children's Hospital, Boston, MA, United States
- Pain and Affective Neuroscience Center, Department of Anesthesiology, Critical Care, Pain Medicine, Boston Children's Hospital, Boston, MA, United States
- Department of Psychiatry, Harvard Medical School, Boston, MA, United States
| |
Collapse
|
10
|
Nguyen NN, Ho DS, Nguyen HS, Ho DKN, Li HY, Lin CY, Chiu HY, Chen YC. Preadmission use of antidiabetic medications and mortality among patients with COVID-19 having type 2 diabetes: A meta-analysis. Metabolism 2022; 131:155196. [PMID: 35367460 PMCID: PMC8970613 DOI: 10.1016/j.metabol.2022.155196] [Citation(s) in RCA: 43] [Impact Index Per Article: 14.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/30/2021] [Revised: 03/10/2022] [Accepted: 03/28/2022] [Indexed: 12/16/2022]
Abstract
BACKGROUND Diabetes is an independent predictor of poor outcomes in patients with COVID-19. We compared the effects of the preadmission use of antidiabetic medications on the in-hospital mortality of patients with COVID-19 having type 2 diabetes. METHODS A systematic search of PubMed, EMBASE, Scopus and Web of Science databases was performed to include studies (except case reports and review articles) published until November 30, 2021. We excluded papers regarding in-hospital use of antidiabetic medications. We used a random-effects meta-analysis to calculate the pooled OR (95% CI) and performed a sensitivity analysis to confirm the robustness of the meta-analyses. MAIN FINDINGS We included 61 studies (3,061,584 individuals), which were rated as having low risk of bias. The OR (95% CI) indicated some medications protective against COVID-related death, including metformin [0.54 (0.47-0.62), I2 86%], glucagon-like peptide-1 receptor agonist (GLP-1RA) [0.51 (0.37-0.69), I2 85%], and sodium-glucose transporter-2 inhibitor (SGLT-2i) [0.60 (0.40-0.88), I2 91%]. Dipeptidyl peptidase-4 inhibitor (DPP-4i) [1.23 (1.07-1.42), I2 82%] and insulin [1.70 (1.33-2.19), I2 97%] users were more likely to die during hospitalization. Sulfonylurea, thiazolidinedione, and alpha-glucosidase inhibitor were mortality neutral [0.92 (95% CI 0.83-1.01, I2 44%), 0.90 (95% CI 0.71-1.14, I2 46%), and 0.61 (95% CI 0.26-1.45, I2 77%), respectively]. The sensitivity analysis indicated that our findings were robust. CONCLUSIONS Metformin, GLP-1RA, and SGLT-2i were associated with lower mortality rate in patients with COVID-19 having type 2 diabetes. DPP-4i and insulin were linked to increased mortality. Sulfonylurea, thiazolidinedione, and alpha-glucosidase inhibitors were mortality neutral. These findings can have a large impact on the clinicians' decisions amid the COVID-19 pandemic.
Collapse
Affiliation(s)
- Nam Nhat Nguyen
- College of Medicine, Taipei Medical University, Taipei, Taiwan
| | - Dung Si Ho
- Department of Geriatric Medicine, Faculty of Medicine, Pham Ngoc Thach University of Medicine, Ho Chi Minh City, Viet Nam; Department of Pulmonology, Thong Nhat Hospital, Ho Chi Minh City, Viet Nam
| | - Hung Song Nguyen
- College of Medicine, Taipei Medical University, Taipei, Taiwan; Wellcome Trust Major Overseas Program, Oxford University Clinical Research Unit, Ho Chi Minh City, Viet Nam
| | - Dang Khanh Ngan Ho
- School of Nutrition and Health Sciences, College of Nutrition, Taipei Medical University, Taipei, Taiwan
| | - Hung-Yuan Li
- Department of Internal Medicine, National Taiwan University Hospital, Taipei, Taiwan
| | - Chia-Yuan Lin
- Department of Family Medicine, Taipei Medical University Hospital, Taipei, Taiwan
| | - Hsiao-Yean Chiu
- School of Nursing, College of Nursing, Taipei Medical University, Taipei, Taiwan; Research Center of Sleep Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan
| | - Yang-Ching Chen
- Department of Family Medicine, Taipei Medical University Hospital, Taipei, Taiwan; Department of Family Medicine, School of Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan.
| |
Collapse
|
11
|
Impact of untreated diabetes and COVID-19-related diabetes on severe COVID-19. Heliyon 2022; 8:e08801. [PMID: 35079646 PMCID: PMC8776352 DOI: 10.1016/j.heliyon.2022.e08801] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2021] [Revised: 01/06/2022] [Accepted: 01/17/2022] [Indexed: 01/08/2023] Open
Abstract
Diabetes is a common comorbidity in patients with coronavirus disease (COVID-19) and contributes significantly to COVID-19 severity. We aimed to investigate the association between diabetic status and severe COVID-19. This prospective study included all COVID-19 patients admitted to our hospital, who were divided into four groups according to their diabetic status: no diabetes, treated diabetes, untreated diabetes, and COVID-19-related diabetes. Severe COVID-19 was defined as a condition that required the use of a ventilator. Of the 114 patients included in this study, 26 had severe COVID-19. The adjusted odds ratio (OR; 95% confidence interval [CI]) for severe COVID-19 was significantly higher in the treated diabetes, untreated diabetes, and COVID-19-related diabetes groups than in the no diabetes group (OR: 5.9, 95% CI [1.2–27.9]; OR 12.6, 95% CI [1.8–86.4]; and OR: 9.3, 95% [1.1–81.4], respectively). Findings from this study showed that the risk of severe COVID-19 was increased in treated diabetes, untreated diabetes, and COVID-19-related diabetes compared to no diabetes. Furthermore, the OR for severe COVID-19 was greater in untreated diabetes and COVID-19-related diabetes than in treated diabetes.
Collapse
|
12
|
Casagrande V, Federici M, Menghini R. TIMP3 involvement and potentiality in the diagnosis, prognosis and treatment of diabetic nephropathy. Acta Diabetol 2021; 58:1587-1594. [PMID: 34181080 PMCID: PMC8542557 DOI: 10.1007/s00592-021-01766-y] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/26/2021] [Accepted: 06/14/2021] [Indexed: 11/28/2022]
Abstract
Diabetic kidney disease, one of the most severe complications associated with diabetes, is characterized by albuminuria, glomerulosclerosis and progressive loss of renal function. Loss of TIMP3, an Extracellular matrix-bound protein, is a hallmark of diabetic nephropathy in human and mouse models, suggesting its pivotal role in renal diseases associated to diabetes. There is currently no specific therapy for diabetic nephropathy, and the ability to restore high TIMP3 activity specifically in the kidney may represent a potential therapeutic strategy for the amelioration of renal injury under conditions in which its reduction is directly related to the disease. Increasing evidence shows that diabetic nephropathy is also regulated by epigenetic mechanisms, including noncoding RNA. This review recapitulates the pathological, diagnostic and therapeutic potential roles of TIMP3 and the noncoding RNA (microRNA, long noncoding RNA) related to its expression, in the progression of diabetic nephropathy.
Collapse
Affiliation(s)
- Viviana Casagrande
- Departments of Systems Medicine, University of Rome "Tor Vergata", Rome, Italy
| | - Massimo Federici
- Departments of Systems Medicine, University of Rome "Tor Vergata", Rome, Italy
- Center for Atherosclerosis, Department of Medical Sciences, Policlinico Tor Vergata University, Rome, Italy
| | - Rossella Menghini
- Departments of Systems Medicine, University of Rome "Tor Vergata", Rome, Italy.
| |
Collapse
|
13
|
Luong TV, Pedersen MGB, Kjærulff MLBG, Madsen S, Lauritsen KM, Tolbod LP, Søndergaard E, Gormsen LC. Ischemic heart failure mortality is not predicted by cardiac insulin resistance but by diabetes per se and coronary flow reserve: A retrospective dynamic cardiac 18F-FDG PET study. Metabolism 2021; 123:154862. [PMID: 34375646 DOI: 10.1016/j.metabol.2021.154862] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/16/2021] [Revised: 07/09/2021] [Accepted: 07/28/2021] [Indexed: 01/01/2023]
Abstract
BACKGROUND & AIMS The connection between peripheral insulin resistance (IR) and coronary artery disease is well-established. Both are major risk factors for the development of ischemic cardiomyopathy potentially leading to heart failure (HF). Whether cardiac IR also impacts overall survival and morbidity is still debated. We therefore aimed to test if cardiac IR predicts mortality and major cardiovascular events (MACE) in patients with HF scheduled for cardiac viability testing before revascularization. METHODS This retrospective study included 131 patients with a clinical diagnosis of ischemic HF (114 (87%) male, 33 (25%) with diabetes) referred to a viability Rubidium-82 (perfusion) and dynamic 18F-Fluorodeoxyglucose (metabolism) positron emission tomography combined with computed tomography prior to a potential revascularization procedure. Cardiac IR was assessed by myocardial glucose uptake (MGU) in a remote (non-scarred) area of the left ventricle during a hyperinsulinemic-euglycemic clamp (1mIE/kg/min). RESULTS MGU correlated with skeletal muscle glucose uptake (p < 0.001) and whole-body glucose uptake (M-value) (p < 0.001), whereas no association was observed for individuals with diabetes. MGU did not predict the risk of death or MACE. However, both overt diabetes and reduced coronary flow reserve predicted overall survival. CONCLUSION Even though diabetes and related small-vessel disease is associated with increased mortality, cardiac IR per se does not predict cardiovascular morbidity and mortality.
Collapse
Affiliation(s)
- Thien Vinh Luong
- Department of Nuclear Medicine & PET-Centre, Aarhus University Hospital, Palle Juul-Jensens Boulevard 165, 8200 Aarhus N, Denmark; Steno Diabetes Center Aarhus, Aarhus University Hospital, Hedeager 3, 8200 Aarhus N, Denmark.
| | | | - Mette Louise Blouner Gram Kjærulff
- Department of Nuclear Medicine & PET-Centre, Aarhus University Hospital, Palle Juul-Jensens Boulevard 165, 8200 Aarhus N, Denmark; Steno Diabetes Center Aarhus, Aarhus University Hospital, Hedeager 3, 8200 Aarhus N, Denmark
| | - Simon Madsen
- Department of Nuclear Medicine & PET-Centre, Aarhus University Hospital, Palle Juul-Jensens Boulevard 165, 8200 Aarhus N, Denmark
| | - Katrine Meyer Lauritsen
- Steno Diabetes Center Aarhus, Aarhus University Hospital, Hedeager 3, 8200 Aarhus N, Denmark
| | - Lars Poulsen Tolbod
- Department of Nuclear Medicine & PET-Centre, Aarhus University Hospital, Palle Juul-Jensens Boulevard 165, 8200 Aarhus N, Denmark
| | - Esben Søndergaard
- Steno Diabetes Center Aarhus, Aarhus University Hospital, Hedeager 3, 8200 Aarhus N, Denmark
| | - Lars Christian Gormsen
- Department of Nuclear Medicine & PET-Centre, Aarhus University Hospital, Palle Juul-Jensens Boulevard 165, 8200 Aarhus N, Denmark
| |
Collapse
|
14
|
Pioglitazone Reverses Markers of Islet Beta-Cell De-Differentiation in db/db Mice While Modulating Expression of Genes Controlling Inflammation and Browning in White Adipose Tissue from Insulin-Resistant Mice and Humans. Biomedicines 2021; 9:biomedicines9091189. [PMID: 34572374 PMCID: PMC8470788 DOI: 10.3390/biomedicines9091189] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2021] [Revised: 08/21/2021] [Accepted: 09/02/2021] [Indexed: 12/12/2022] Open
Abstract
Obesity, insulin resistance, and type 2 diabetes contribute to increased morbidity and mortality in humans. The db/db mouse is an important mouse model that displays many key features of the human disease. Herein, we used the drug pioglitazone, a thiazolidinedione with insulin-sensitizing properties, to investigate blood glucose levels, indicators of islet β-cell health and maturity, and gene expression in adipose tissue. Oral administration of pioglitazone lowered blood glucose levels in db/db mice with a corresponding increase in respiratory quotient, which indicates improved whole-body carbohydrate utilization. In addition, white adipose tissue from db/db mice and from humans treated with pioglitazone showed increased expression of glycerol kinase. Both db/db mice and humans given pioglitazone displayed increased expression of UCP-1, a marker typically associated with brown adipose tissue. Moreover, pancreatic β-cells from db/db mice treated with pioglitazone had greater expression of insulin and Nkx6.1 as well as reduced abundance of the de-differentiation marker Aldh1a3. Collectively, these findings indicate that four weeks of pioglitazone therapy improved overall metabolic health in db/db mice. Our data are consistent with published reports of human subjects administered pioglitazone and with analysis of human adipose tissue taken from subjects treated with pioglitazone. In conclusion, the current study provides evidence that pioglitazone restores key markers of metabolic health and also showcases the utility of the db/db mouse to understand mechanisms associated with human metabolic disease and interventions that provide therapeutic benefit.
Collapse
|
15
|
Tripathy D, Solis-Herrera C, Ryder RE. Cardioprotective Effects of Pioglitazone in Type 2 Diabetes. Diabetes Spectr 2021; 34:243-247. [PMID: 34511850 PMCID: PMC8387608 DOI: 10.2337/ds20-0078] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
Antidiabetic medications that improve glycemic control as well as cardiovascular outcomes will be the mainstay of treatment for type 2 diabetes moving forward. This article reviews the beneficial effects of the thiazolidinedione pioglitazone of ameliorating hyperglycemia and improving cardiovascular risk factors. While the newer sodium-glucose cotransporter 2 inhibitor and glucagon-like peptide 1 receptor agonist drug classes have confirmed cardiovascular benefits, pioglitazone also has been shown to reduce major adverse cardiovascular events, in both people with type 2 diabetes and nondiabetic subjects with insulin resistance. Adverse effects associated with pioglitazone can be mitigated by its use at a lower dose and in combination with antidiabetic agents from other drug classes.
Collapse
Affiliation(s)
- Devjit Tripathy
- Department of Medicine, Diabetes Division, University of Texas Health Science Center, San Antonio, TX
- Audie L. Murphy VA Hospital, South Texas Veterans Heath Care System, Foundation for Advancement of Veteran’s Health and Research (FAVHR), San Antonio, TX
- Corresponding author: Devjit Tripathy,
| | - Carolina Solis-Herrera
- Department of Medicine, Diabetes Division, University of Texas Health Science Center, San Antonio, TX
| | | |
Collapse
|
16
|
Codella R, Della Guardia L, Terruzzi I, Solini A, Folli F, Varoni EM, Carrassi A, Luzi L. Physical activity as a proxy to ameliorate inflammation in patients with type 2 diabetes and periodontal disease at high cardiovascular risk. Nutr Metab Cardiovasc Dis 2021; 31:2199-2209. [PMID: 34099361 DOI: 10.1016/j.numecd.2021.04.022] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/04/2021] [Revised: 04/19/2021] [Accepted: 04/24/2021] [Indexed: 12/17/2022]
Abstract
While the beneficial impact of physical activity has been ascertained in a variety of pathological scenarios, including diabetes and low-grade systemic inflammation, its potential remains still putative for periodontal health. Periodontal disease has been associated with inflammatory systemic alterations, which share a common denominator with type 2 diabetes mellitus and cardiovascular disease. Physical exercise, along with nutritional counseling, is a cornerstone in the treatment and prevention of type 2 diabetes, also able to reduce the prevalence of periodontal disease and cardiovascular risk. In addition, considering the higher incidence of periodontitis in patients with type 2 diabetes compared to healthy controls, the fascinating research question would be whether physical activity could relieve the inflammatory pressure exerted by the combination of these two diseases. This multi-disciplinary viewpoint discusses available literature in order to argument the hypothesis of a "three-way relationship" linking diabetes, periodontitis, and physical activity.
Collapse
Affiliation(s)
- Roberto Codella
- Department of Biomedical Sciences for Health, Università Degli Studi di Milano, Milano, Italy; Department of Endocrinology, Nutrition and Metabolic Diseases, IRCCS MultiMedica, Milan, Italy
| | - Lucio Della Guardia
- Department of Biomedical Sciences for Health, Università Degli Studi di Milano, Milano, Italy
| | - Ileana Terruzzi
- Department of Biomedical Sciences for Health, Università Degli Studi di Milano, Milano, Italy; Department of Endocrinology, Nutrition and Metabolic Diseases, IRCCS MultiMedica, Milan, Italy
| | - Anna Solini
- Department of Surgical, Medical, Molecular and Critical Area Pathology, Università di Pisa, Pisa, Italy
| | - Franco Folli
- Endocrinology and Metabolism, Department of Health Science, Università Degli Studi di Milano, Milano, Italy
| | - Elena Maria Varoni
- Department of Biomedical, Surgical and Dental Sciences, Università degli Studi di Milano, Milano, Italy
| | - Antonio Carrassi
- Department of Biomedical, Surgical and Dental Sciences, Università degli Studi di Milano, Milano, Italy
| | - Livio Luzi
- Department of Biomedical Sciences for Health, Università Degli Studi di Milano, Milano, Italy; Department of Endocrinology, Nutrition and Metabolic Diseases, IRCCS MultiMedica, Milan, Italy.
| |
Collapse
|
17
|
Bignotto M, Dei Cas M, Paroni R, Bianco E, Zermiani P, Gangale MG, Zadro V, Maregatti M, Piagnani A, Russo A, Baldassarre D, Folli F, Battezzati PM, Zuin M. CA.ME.LI.A. An epidemiological study on the prevalence of CArdiovascular, MEtabolic, LIver and Autoimmune diseases in Northern Italy. Nutr Metab Cardiovasc Dis 2021; 31:1416-1426. [PMID: 33814235 DOI: 10.1016/j.numecd.2021.02.001] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/27/2020] [Revised: 01/29/2021] [Accepted: 02/03/2021] [Indexed: 12/12/2022]
Abstract
BACKGROUND AND AIMS CA.ME.LI.A (CArdiovascular risks, MEtabolic syndrome, LIver and Autoimmune disease) is a cross-sectional, epidemiological study performed between 2009-2011 in Abbiategrasso (Milan, Italy) to estimate the prevalence of cardiovascular risk factors, metabolic syndrome, liver and autoimmune diseases in the general adult population. This report focuses on the description and presentation of baseline characteristics of the population. METHODS AND RESULTS Citizens were randomly selected from the city electoral registers (n = 30903), yielding a sample of 2554 subjects (M = 1257, F = 1297; age, 47 ± 15 yrs; range 18-77 yrs). Men had higher prevalence of overweight or obesity (60.8% vs 41.6%; p < 0.0001) and greater thickness of visceral adipose tissue (40 ± 19 vs 27 ± 17 mm; p < 0.0001); no gender difference was found in subcutaneous adipose tissue thickness. Men also showed higher levels of serum triglycerides, γ-GT, fasting blood glucose, insulin and Homa-IR Index, while HDL, CRP, and prevalence of elevated (>5.0 mg/L) CRP were lower. Compared to normal weight men, risk-ratio (RR) of CRP elevation was 1.32 (ns) in overweight and 2.68 (p < 0.0001) in obese subjects. The corresponding figures in females were 2.68 (p < 0.0001) and 5.18 (p < 0.0001). Metabolic syndrome was more frequent in men (32.7% vs 14.5%; RR: 2.24, p < 0.0001). Interadventitia common carotid artery diameter was higher in men and increased with age and BMI. CONCLUSIONS The present study reports on the overall characteristics of a large population from Northern Italy. It aims to identify the associations among cardiovascular risk factors to prevent their development and progression, improve healthy lifestyle and identify subjects liable to pharmacological interventions.
Collapse
Affiliation(s)
- Monica Bignotto
- Liver and Gastroenterology Unit, Department of Health Sciences, Universita' degli Studi di Milano, Milan, Italy
| | - Michele Dei Cas
- Clinical Biochemistry and Mass Spectrometry, Department of Health Sciences, Universita' degli Studi di Milano, Milan, Italy
| | - Rita Paroni
- Clinical Biochemistry and Mass Spectrometry, Department of Health Sciences, Universita' degli Studi di Milano, Milan, Italy
| | - Elena Bianco
- Liver and Gastroenterology Unit, Department of Health Sciences, Universita' degli Studi di Milano, Milan, Italy
| | - Paola Zermiani
- Liver and Gastroenterology Unit, Department of Health Sciences, Universita' degli Studi di Milano, Milan, Italy
| | - Maria G Gangale
- ASST Ovest Milanese, via Papa Giovanni Paolo II, Legnano, Milan, Italy
| | - Valentina Zadro
- Liver and Gastroenterology Unit, Department of Health Sciences, Universita' degli Studi di Milano, Milan, Italy
| | - Margherita Maregatti
- Liver and Gastroenterology Unit, Department of Health Sciences, Universita' degli Studi di Milano, Milan, Italy
| | - Alessandra Piagnani
- Liver and Gastroenterology Unit, Department of Health Sciences, Universita' degli Studi di Milano, Milan, Italy
| | - Antonio Russo
- Epidemiology Unit, Agency for Health Protection of Milan, Corso Italia 19, 20122, Milan, Italy
| | - Damiano Baldassarre
- Department of Medical Biotechnology and Translational Medicine, Università degli Studi di Milano, Milan, Italy; Centro Cardiologico Monzino, IRCCS, Milan, Italy
| | - Franco Folli
- Endocrinology and Metabolism, Department of Health Sciences, Universita' degli Studi di Milano, Milan, Italy; ASST Santi Paolo e Carlo, University Hospital San Paolo, via A. Di Rudini', Milan, Italy.
| | - Pier Maria Battezzati
- Liver and Gastroenterology Unit, Department of Health Sciences, Universita' degli Studi di Milano, Milan, Italy; ASST Santi Paolo e Carlo, University Hospital San Paolo, via A. Di Rudini', Milan, Italy
| | - Massimo Zuin
- Liver and Gastroenterology Unit, Department of Health Sciences, Universita' degli Studi di Milano, Milan, Italy; ASST Santi Paolo e Carlo, University Hospital San Paolo, via A. Di Rudini', Milan, Italy.
| |
Collapse
|
18
|
Oz M, Lorke DE, Kabbani N. A comprehensive guide to the pharmacologic regulation of angiotensin converting enzyme 2 (ACE2), the SARS-CoV-2 entry receptor. Pharmacol Ther 2021; 221:107750. [PMID: 33275999 PMCID: PMC7854082 DOI: 10.1016/j.pharmthera.2020.107750] [Citation(s) in RCA: 27] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2020] [Revised: 11/18/2020] [Accepted: 11/19/2020] [Indexed: 02/06/2023]
Abstract
The recent emergence of coronavirus disease-2019 (COVID-19) as a global pandemic has prompted scientists to address an urgent need for defining mechanisms of disease pathology and treatment. Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), the causative agent for COVID-19, employs angiotensin converting enzyme 2 (ACE2) as its primary target for cell surface attachment and likely entry into the host cell. Thus, understanding factors that may regulate the expression and function of ACE2 in the healthy and diseased body is critical for clinical intervention. Over 66% of all adults in the United States are currently using a prescription drug and while earlier findings have focused on possible upregulation of ACE2 expression through the use of renin angiotensin system (RAS) inhibitors, mounting evidence suggests that various other widely administered drugs used in the treatment of hypertension, heart failure, diabetes mellitus, hyperlipidemias, coagulation disorders, and pulmonary disease may also present a varied risk for COVID-19. Specifically, we summarize mechanisms on how heparin, statins, steroids and phytochemicals, besides their established therapeutic effects, may also interfere with SARS-CoV-2 viral entry into cells. We also describe evidence on the effect of several vitamins, phytochemicals, and naturally occurring compounds on ACE2 expression and activity in various tissues and disease models. This comprehensive review aims to provide a timely compendium on the potential impact of commonly prescribed drugs and pharmacologically active compounds on COVID-19 pathology and risk through regulation of ACE2 and RAS signaling.
Collapse
Key Words
- adam17, a disintegrin and metalloprotease 17
- ace, angiotensin i converting enzyme
- ace-inh., angiotensin i converting enzyme inhibitor
- ampk, amp-activated protein kinase
- ang-ii, angiotensin ii
- arb, angiotensin ii type 1-receptor blocker
- ards, acute respiratory distress syndrome
- at1-r, angiotensin ii type 1-receptor
- βarb, β-adrenergic receptor blockers
- bk, bradykinin
- ccb, calcium channel blockers
- ch25h, cholesterol-25-hydroxylase
- copd, chronic obstructive lung disease
- cox, cyclooxygenase
- covid-19, coronavirus disease-2019
- dabk, [des-arg9]-bradykinin
- erk, extracellular signal-regulated kinase
- 25hc, 25-hydroxycholesterol
- hs, heparan sulfate
- hspg, heparan sulfate proteoglycan
- ibd, inflammatory bowel disease
- map, mitogen-activated protein
- mers, middle east respiratory syndrome
- mrb, mineralocorticoid receptor blocker
- nos, nitric oxide synthase
- nsaid, non-steroid anti-inflammatory drug
- ras, renin-angiotensin system
- sars-cov, severe acute respiratory syndrome coronavirus
- sh, spontaneously hypertensive
- s protein, spike protein
- sirt1, sirtuin 1
- t2dm, type 2 diabetes mellitus
- tcm, traditional chinese medicine
- tmprss2, transmembrane protease, serine 2
- tnf, tumor necrosis factor
- ufh, unfractionated heparin
Collapse
Affiliation(s)
- Murat Oz
- Department of Pharmacology and Therapeutics, Faculty of Pharmacy, Kuwait University, Safat 13110, Kuwait.
| | - Dietrich Ernst Lorke
- Department of Anatomy and Cellular Biology, College of Medicine and Health Sciences, Khalifa University, Abu Dhabi, United Arab Emirates; Center for Biotechnology, Khalifa University of Science and Technology, Abu Dhabi, United Arab Emirates
| | - Nadine Kabbani
- School of Systems Biology, George Mason University, Fairfax, VA 22030, USA
| |
Collapse
|
19
|
Li G, Chen Z, Lv Z, Li H, Chang D, Lu J. Diabetes Mellitus and COVID-19: Associations and Possible Mechanisms. Int J Endocrinol 2021; 2021:7394378. [PMID: 33859687 PMCID: PMC8025139 DOI: 10.1155/2021/7394378] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/16/2020] [Revised: 08/02/2020] [Accepted: 03/11/2021] [Indexed: 01/08/2023] Open
Abstract
Coronavirus disease 2019 (COVID-19) is a recently emerged disease with formidable infectivity and high mortality. Emerging data suggest that diabetes is one of the most prevalent comorbidities in patients with COVID-19. Although their causal relationship has not yet been investigated, preexisting diabetes can be considered as a risk factor for the adverse outcomes of COVID-19. Proinflammatory state, attenuation of the innate immune response, possibly increased level of ACE2, along with vascular dysfunction, and prothrombotic state in people with diabetes probably contribute to higher susceptibility for SARS-CoV-2 infection and worsened prognosis. On the other hand, activated inflammation, islet damage induced by virus infection, and treatment with glucocorticoids could, in turn, result in impaired glucose regulation in people with diabetes, thus working as an amplification loop to aggravate the disease. Therefore, glycemic management in people with COVID-19, especially in those with severe illness, is of considerable importance. The insights may help to reduce the fatality in the effort against COVID-19.
Collapse
Affiliation(s)
- Gerui Li
- Department of Geratology, Zhongnan Hospital of Wuhan University, Wuhan 430071, Hubei, China
| | - Ze Chen
- Department of Cardiology, Zhongnan Hospital of Wuhan University, Wuhan 430071, Hubei, China
| | - Zhan Lv
- Department of Geratology, Zhongnan Hospital of Wuhan University, Wuhan 430071, Hubei, China
| | - Hang Li
- Department of Geratology, Zhongnan Hospital of Wuhan University, Wuhan 430071, Hubei, China
| | - Danqi Chang
- Department of Geriatrics, Renmin Hospital of Wuhan University, Wuhan 430060, Hubei, China
| | - Jinping Lu
- Department of Geratology, Zhongnan Hospital of Wuhan University, Wuhan 430071, Hubei, China
| |
Collapse
|
20
|
Fiorentino TV, Monroy A, Kamath S, Sotero R, Cas MD, Daniele G, Chavez AO, Abdul-Ghani M, Hribal ML, Sesti G, Tripathy D, DeFronzo RA, Folli F. Pioglitazone corrects dysregulation of skeletal muscle mitochondrial proteins involved in ATP synthesis in type 2 diabetes. Metabolism 2021; 114:154416. [PMID: 33137378 DOI: 10.1016/j.metabol.2020.154416] [Citation(s) in RCA: 24] [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: 08/02/2020] [Revised: 10/27/2020] [Accepted: 10/28/2020] [Indexed: 12/25/2022]
Abstract
CONTEXT In this study, we aimed to identify the determinants of mitochondrial dysfunction in skeletal muscle (SKLM) of subjects with type 2 diabetes (T2DM), and to evaluate the effect of pioglitazone (PIO) on SKLM mitochondrial proteome. METHODS Two different groups of adults were studied. Group I consisted of 8 individuals with normal glucose tolerance (NGT) and 8 with T2DM, subjected to SKLM mitochondrial proteome analysis by 2D-gel electrophoresis followed by mass spectrometry-based protein identification. Group II included 24 individuals with NGT and 24 with T2DM, whose SKLM biopsies were subjected to immunoblot analysis. Of the 24 subjects with T2DM, 20 were randomized to receive placebo or PIO (15 mg daily) for 6 months. After 6 months of treatment, SKLM biopsy was repeated. RESULTS Mitochondrial proteomic analysis on Group I revealed that several mitochondrial proteins involved in oxidative metabolism were differentially expressed between T2DM and NGT groups, with a downregulation of ATP synthase alpha chain (ATP5A), electron transfer flavoprotein alpha-subunit (ETFA), cytochrome c oxidase subunit VIb isoform 1 (CX6B1), pyruvate dehydrogenase protein X component (ODPX), dihydrolipoamide dehydrogenase (DLDH), dihydrolipoamide-S-succinyltransferase (DLST), and mitofilin, and an up-regulation of hydroxyacyl-CoA-dehydrogenase (HCDH), 3,2-trans-enoyl-CoA-isomerase (D3D2) and delta3,5-delta2,4-dienoyl-CoA-isomerase (ECH1) in T2DM as compared to NGT subjects. By immunoblot analysis on SKLM lysates obtained from Group II we confirmed that, in comparison to NGT subjects, those with T2DM exhibited lower protein levels of ATP5A (-30%, P = 0.006), ETFA (-50%, P = 0.02), CX6B1 (-30%, P = 0.03), key factors for ATP biosynthesis, and of the structural protein mitofilin (-30%, P = 0.01). T2DM was associated with a reduced abundance of the enzymes involved in the Krebs cycle DLST and ODPX (-20%, P ≤ 0.05) and increased levels of HCDH and ECH1, enzymes implicated in the fatty acid catabolism (+30%, P ≤ 0.05). In subjects with type 2 diabetes treated with PIO for 6 months we found a restored SKLM protein abundance of ATP5A, ETFA, CX6B1, and mitofilin. Moreover, protein levels of HCDH and ECH1 were reduced by -10% and - 15% respectively (P ≤ 0.05 for both) after PIO treatment. CONCLUSION Type 2 diabetes is associated with reduced levels of mitochondrial proteins involved in oxidative phosphorylation and an increased abundance of enzymes implicated in fatty acid catabolism in SKLM. PIO treatment is able to improve SKLM mitochondrial proteomic profile in subjects with T2DM.
Collapse
Affiliation(s)
- Teresa Vanessa Fiorentino
- Department of Medical and Surgical Sciences, University Magna Graecia of Catanzaro, Catanzaro, Italy; Division of Diabetes, Department of Medicine, University of Texas Health Science Center at San Antonio, San Antonio, TX, United States of America
| | - Adriana Monroy
- Division of Diabetes, Department of Medicine, University of Texas Health Science Center at San Antonio, San Antonio, TX, United States of America; Oncology, General Hospital of Mexico, Mexico City, Mexico
| | - Subash Kamath
- Division of Diabetes, Department of Medicine, University of Texas Health Science Center at San Antonio, San Antonio, TX, United States of America
| | - Rosa Sotero
- Department of Medical and Surgical Sciences, University Magna Graecia of Catanzaro, Catanzaro, Italy
| | - Michele Dei Cas
- Clinical Biochemistry and Mass Spectrometry Laboratory, Department of Health Science, University of Milan, Milan, Italy
| | - Giuseppe Daniele
- Division of Diabetes, Department of Medicine, University of Texas Health Science Center at San Antonio, San Antonio, TX, United States of America
| | - Alberto O Chavez
- Department of Medical and Surgical Sciences, University Magna Graecia of Catanzaro, Catanzaro, Italy
| | - Muhammad Abdul-Ghani
- Division of Diabetes, Department of Medicine, University of Texas Health Science Center at San Antonio, San Antonio, TX, United States of America
| | - Marta Letizia Hribal
- Department of Medical and Surgical Sciences, University Magna Graecia of Catanzaro, Catanzaro, Italy
| | - Giorgio Sesti
- Department of Clinical and Molecular Medicine, University of Rome-Sapienza, Rome, Italy
| | - Devjit Tripathy
- Division of Diabetes, Department of Medicine, University of Texas Health Science Center at San Antonio, San Antonio, TX, United States of America
| | - Ralph A DeFronzo
- Division of Diabetes, Department of Medicine, University of Texas Health Science Center at San Antonio, San Antonio, TX, United States of America
| | - Franco Folli
- Division of Diabetes, Department of Medicine, University of Texas Health Science Center at San Antonio, San Antonio, TX, United States of America; Endocrinology and Metabolism, Department of Health Science, University of Milan, Diabetologia e Malattie Metaboliche, Aziende Socio Sanitarie Territoriali Santi Paolo e Carlo, Milan, Italy.
| |
Collapse
|
21
|
Dandona P, Dhindsa S, Ghanim H, Saad F. Mechanisms underlying the metabolic actions of testosterone in humans: A narrative review. Diabetes Obes Metab 2021; 23:18-28. [PMID: 32991053 DOI: 10.1111/dom.14206] [Citation(s) in RCA: 38] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/16/2020] [Revised: 09/13/2020] [Accepted: 09/25/2020] [Indexed: 12/20/2022]
Abstract
The role of testosterone in improving sexual symptoms in men with hypogonadism is well known. However, recent studies indicate that testosterone plays an important role in several metabolic functions in males. Multiple PubMed searches were conducted with the use of the terms testosterone, insulin sensitivity, obesity, type 2 diabetes, anaemia, bone density, osteoporosis, fat mass, lean mass and body composition. This narrative review is focused on detailing the mechanisms that underlie the metabolic aspects of testosterone therapy in humans. Testosterone enhances insulin sensitivity in obese men with hypogonadism by decreasing fat mass, increasing lean mass, decreasing free fatty acids and suppressing inflammation. At a cellular level, testosterone increases the expression of insulin receptor β subunit, insulin receptor substrate-1, protein kinase B and glucose transporter type 4 in adipose tissue and adenosine 5'-monophosphate-activated protein kinase expression and activity in skeletal muscle. Observational studies show that long-term therapy with testosterone prevents progression from prediabetes to diabetes and improves HbA1c. Testosterone increases skeletal muscle satellite cell activator, fibroblast growth factor-2 and decreases expression of the muscle growth suppressors, myostatin and myogenic regulatory factor 4. Testosterone increases haematocrit by suppressing hepcidin and increasing expression of ferroportin along with that of transferrin receptor and plasma transferrin concentrations. Testosterone also increases serum osteocalcin concentrations, which may account for its anabolic actions on bone. In conclusion, testosterone exerts a series of potent metabolic effects, which include insulin sensitization, maintenance and growth of the skeletal muscle, suppression of adipose tissue growth and maintenance of erythropoiesis and haematocrit.
Collapse
Affiliation(s)
- Paresh Dandona
- Division of Endocrinology, Diabetes and Metabolism, State University of New York at Buffalo, Williamsville, New York, USA
| | - Sandeep Dhindsa
- Division of Endocrinology, Diabetes and Metabolism, State University of New York at Buffalo, Williamsville, New York, USA
- Division of Endocrinology, Diabetes and Metabolism, Saint Louis University, St. Louis, Missouri, USA
| | - Husam Ghanim
- Division of Endocrinology, Diabetes and Metabolism, State University of New York at Buffalo, Williamsville, New York, USA
| | - Farid Saad
- Research Department, Gulf Medical University, Ajman, UAE
| |
Collapse
|
22
|
Type 2 diabetes subgroups and potential medication strategies in relation to effects on insulin resistance and beta-cell function: A step toward personalised diabetes treatment? Mol Metab 2020; 46:101158. [PMID: 33387681 PMCID: PMC8085543 DOI: 10.1016/j.molmet.2020.101158] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/01/2020] [Revised: 11/16/2020] [Accepted: 12/27/2020] [Indexed: 02/07/2023] Open
Abstract
Background Type 2 diabetes is a syndrome defined by hyperglycaemia that is the result of various degrees of pancreatic β-cell failure and reduced insulin sensitivity. Although diabetes can be caused by multiple metabolic dysfunctions, most patients are defined as having either type 1 or type 2 diabetes. Recently, Ahlqvist and colleagues proposed a new method of classifying patients with adult-onset diabetes, considering the heterogenous metabolic phenotype of the disease. This new classification system could be useful for more personalised treatment based on the underlying metabolic disruption of the disease, although to date no prospective intervention studies have generated data to support such a claim. Scope of Review In this review, we first provide a short overview of the phenotype and pathogenesis of type 2 diabetes and discuss the current and new classification systems. We then review the effects of different anti-diabetic medication classes on insulin sensitivity and β-cell function and discuss future treatment strategies based on the subgroups proposed by Ahlqvist et al. Major Conclusions The proposed novel type 2 diabetes subgroups provide an interesting concept that could lead to a better understanding of the pathophysiology of the broad group of type 2 diabetes, paving the way for personalised treatment choices based on understanding the root cause of the disease. We conclude that the novel subgroups of adult-onset diabetes would benefit from anti-diabetic medications that take into account the main pathophysiology of the disease and thereby prevent end-organ damage. However, we are only beginning to address the personalised treatment of type 2 diabetes, and studies investigating the effects of current and novel drugs in subgroups with different metabolic phenotypes are needed to develop personalised treatment of the syndrome Novel subgroups of type 2 diabetes provide a concept that could lead to a better understanding of its pathophysiology. Treatment strategies would benefit from anti-diabetic medications that influence the main pathophysiology of diabetes. Here, we review different anti-diabetic medications classes affecting insulin sensitivity and β-cell function. We suggest that future treatment strategies could benefit by taking into account subgroups provided by Ahlqvist et al.
Collapse
|
23
|
ACE2, angiotensin 1-7 and skeletal muscle: review in the era of COVID-19. Clin Sci (Lond) 2020; 134:3047-3062. [PMID: 33231620 PMCID: PMC7687025 DOI: 10.1042/cs20200486] [Citation(s) in RCA: 38] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2020] [Revised: 10/30/2020] [Accepted: 11/03/2020] [Indexed: 12/12/2022]
Abstract
Angiotensin converting enzyme-2 (ACE2) is a multifunctional transmembrane protein recently recognised as the entry receptor of the virus causing COVID-19. In the renin–angiotensin system (RAS), ACE2 cleaves angiotensin II (Ang II) into angiotensin 1-7 (Ang 1-7), which is considered to exert cellular responses to counteract the activation of the RAS primarily through a receptor, Mas, in multiple organs including skeletal muscle. Previous studies have provided abundant evidence suggesting that Ang 1-7 modulates multiple signalling pathways leading to protection from pathological muscle remodelling and muscle insulin resistance. In contrast, there is relatively little evidence to support the protective role of ACE2 in skeletal muscle. The potential contribution of endogenous ACE2 to the regulation of Ang 1-7-mediated protection of these muscle pathologies is discussed in this review. Recent studies have suggested that ACE2 protects against ageing-associated muscle wasting (sarcopenia) through its function to modulate molecules outside of the RAS. Thus, the potential association of sarcopenia with ACE2 and the associated molecules outside of RAS is also presented herein. Further, we introduce the transcriptional regulation of muscle ACE2 by drugs or exercise, and briefly discuss the potential role of ACE2 in the development of COVID-19.
Collapse
|
24
|
Muniangi-Muhitu H, Akalestou E, Salem V, Misra S, Oliver NS, Rutter GA. Covid-19 and Diabetes: A Complex Bidirectional Relationship. Front Endocrinol (Lausanne) 2020; 11:582936. [PMID: 33133024 PMCID: PMC7578412 DOI: 10.3389/fendo.2020.582936] [Citation(s) in RCA: 55] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/13/2020] [Accepted: 09/08/2020] [Indexed: 01/08/2023] Open
Abstract
Covid-19 is a recently-emerged infectious disease caused by the novel severe acute respiratory syndrome coronavirus SARS-CoV2. SARS-CoV2 differs from previous coronavirus infections (SARS and MERS) due to its high infectivity (reproduction value, R0, typically 2-4) and pre- or asymptomatic transmission, properties that have contributed to the current global Covid-19 pandemic. Identified risk factors for disease severity and death from SARS-Cov2 infection include older age, male sex, diabetes, obesity and hypertension. The reasons for these associations are still largely obscure. Evidence is also emerging that SARS-CoV2 infection exacerbates the underlying pathophysiology of hyperglycemia in people with diabetes. Here, we discuss potential mechanisms through which diabetes may affect the risk of more severe outcomes in Covid-19 and, additionally, how diabetic emergencies and longer term pathology may be aggravated by infection with the virus. We consider roles for the immune system, the observed phenomenon of microangiopathy in severe Covid-19 infection and the potential for direct viral toxicity on metabolically-relevant tissues including pancreatic beta cells and targets of insulin action.
Collapse
Affiliation(s)
- Hermine Muniangi-Muhitu
- Section of Cell Biology and Functional Genomics, Division of Diabetes, Endocrinology and Metabolism, Department of Metabolism, Digestion and Reproduction, Imperial College London, London, United Kingdom
| | - Elina Akalestou
- Section of Cell Biology and Functional Genomics, Division of Diabetes, Endocrinology and Metabolism, Department of Metabolism, Digestion and Reproduction, Imperial College London, London, United Kingdom
| | - Victoria Salem
- Section of Endocrinology, Division of Diabetes, Endocrinology and Metabolism, Department of Metabolism, Digestion and Reproduction, Imperial College London, London, United Kingdom
| | - Shivani Misra
- Section of Metabolic Medicine, Division of Diabetes, Endocrinology and Metabolism, Department of Metabolism, Digestion and Reproduction, Imperial College London, London, United Kingdom
| | - Nicholas S. Oliver
- Section of Metabolic Medicine, Division of Diabetes, Endocrinology and Metabolism, Department of Metabolism, Digestion and Reproduction, Imperial College London, London, United Kingdom
| | - Guy A. Rutter
- Section of Cell Biology and Functional Genomics, Division of Diabetes, Endocrinology and Metabolism, Department of Metabolism, Digestion and Reproduction, Imperial College London, London, United Kingdom
- Lee Kong Chian School of Medicine, Nan Yang Technological University, Singapore, Singapore
| |
Collapse
|
25
|
Pal R, Banerjee M. Comment on "Is the type of diabetes treatment relevant to outcome of COVID-19?". J Diabetes 2020; 12:705-707. [PMID: 32459882 PMCID: PMC7283857 DOI: 10.1111/1753-0407.13069] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/17/2020] [Accepted: 05/22/2020] [Indexed: 11/28/2022] Open
Affiliation(s)
- Rimesh Pal
- Department of EndocrinologyPost Graduate Institute of Medical Education and ResearchChandigarhIndia
| | - Mainak Banerjee
- Department of EndocrinologyInstitute of Post Graduate Medical Education and ResearchKolkataIndia
| |
Collapse
|
26
|
Jeong IK, Yoon KH, Lee MK. Diabetes and COVID-19: Global and regional perspectives. Diabetes Res Clin Pract 2020; 166:108303. [PMID: 32623038 PMCID: PMC7332438 DOI: 10.1016/j.diabres.2020.108303] [Citation(s) in RCA: 31] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/31/2020] [Revised: 06/19/2020] [Accepted: 06/29/2020] [Indexed: 02/07/2023]
Abstract
The coronavirus disease-2019 (COVID-19) has been designated as a highly contagious infectious disease caused by the severe acute respiratory syndrome coronavirus-2 (SARS-CoV-2) since December 2019, when an outbreak of pneumonia cases emerged in Wuhan, China. The COVID-19 pandemic has led to a global health crisis, devastating the social, economic and political aspects of life. Many clinicians, health professionals, scientists, organizations, and governments have actively defeated COVID-19 and shared their experiences of the SARS-CoV2. Diabetes is one of the major risk factors for fatal outcomes from COVID-19. Patients with diabetes are vulnerable to infection because of hyperglycemia; impaired immune function; vascular complications; and comorbidities such as hypertension, dyslipidemia, and cardiovascular disease. In addition, angiotensin-converting enzyme 2 (ACE2) is a receptor for SARS-CoV-2 in the human body. Hence, the use of angiotensin-directed medications in patients with diabetes requires attention. The severity and mortality from COVID-19 was significantly higher in patients with diabetes than in those without. Thus, the patients with diabetes should take precautions during the COVID-19 pandemic. Therefore, we review the current knowledge of COVID-19 including the global and regional epidemiology, virology, impact of diabetes on COVID-19, treatment of COVID-19, and standard of care in the management of diabetes during this critical period.
Collapse
Affiliation(s)
- In-Kyung Jeong
- Department of Endocrinology and Metabolism, Kyung Hee University School of Medicine, Kyung Hee University Hospital at Gangdong, Seoul, Republic of Korea.
| | - Kun Ho Yoon
- Division of Endocrinology and Metabolism, Department of Internal Medicine, College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea.
| | - Moon Kyu Lee
- Division of Endocrinology and Metabolism, Department of Internal Medicine, Soonchunghyang University College of Medicine, Gumi, Republic of Korea.
| |
Collapse
|
27
|
Baidya A, Singh SK, Bajaj S, Zargar AH, Singh P, Das S, Shankar A. Diabetes and COVID-19: A Review. J ASEAN Fed Endocr Soc 2020; 35:40-48. [PMID: 33442168 PMCID: PMC7784172 DOI: 10.15605/jafes.035.01.06] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2020] [Accepted: 05/22/2020] [Indexed: 01/08/2023] Open
Abstract
Coronavirus Disease 2019 (COVID-19) is an emerging disease and since its first identification in Wuhan, China, in December 2019, there has been a rapid increase in cases and deaths across the world. COVID-19 has been shown to have an immense impact in infected persons with diabetes, worsening their outcome, especially in elderly, smokers, obese, those having CVD, CKD, poor glycemic control and long duration of diabetes. In this review we summarize the current understanding of `the impact of COVID-19 on diabetes and discusses the pathophysiological mechanisms and management of diabetes and its complication in this scenario.
Collapse
Affiliation(s)
- Arjun Baidya
- Department of Endocrinology, Nil Ratan Sircar Medical College, Kolkata, India
| | | | - Sarita Bajaj
- Department of Medicine, Motilal Nehru Medical College, Allahabad, India
| | - Abdul Hamid Zargar
- Centre for Diabetes and Endocrine Care, National Highway Gulshan Nagar Srinagar, Jammu and Kashmir, India
| | - Parminder Singh
- Department of Endocrinology, Dayanand Medical College and Hospital, Ludhiana, India
| | | | | |
Collapse
|
28
|
Pal R, Bhadada SK. Should anti-diabetic medications be reconsidered amid COVID-19 pandemic? Diabetes Res Clin Pract 2020; 163:108146. [PMID: 32283128 PMCID: PMC7151403 DOI: 10.1016/j.diabres.2020.108146] [Citation(s) in RCA: 89] [Impact Index Per Article: 17.8] [Reference Citation Analysis] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/03/2020] [Accepted: 04/04/2020] [Indexed: 12/03/2022]
Affiliation(s)
- Rimesh Pal
- Department of Endocrinology, Post Graduate Institute of Medical Education and Research, Chandigarh 160012, India
| | - Sanjay K Bhadada
- Department of Endocrinology, Post Graduate Institute of Medical Education and Research, Chandigarh 160012, India.
| |
Collapse
|
29
|
Schinzari F, Tesauro M, Campia U, Cardillo C. Increased fractalkine and vascular dysfunction in obesity and in type 2 diabetes. Effects of oral antidiabetic treatment. Vascul Pharmacol 2020; 128-129:106676. [PMID: 32224233 DOI: 10.1016/j.vph.2020.106676] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2019] [Revised: 02/23/2020] [Accepted: 03/20/2020] [Indexed: 12/29/2022]
Abstract
Activation of fractalkine and other chemokines plays an important role in atherogenesis and, in conjunction with endothelial dysfunction, promotes premature vascular damage in obesity and diabetes. We hypothesized that increased circulating fractalkine coexists with impaired vasomotor function in metabolically healthy or unhealthy obesity, and that treatment with antidiabetic drugs may impact these abnormalities in type 2 diabetes. Compared to lean subjects, in both obese groups the vasodilator responses to acetylcholine and sodium nitroprusside were impaired (both P < .001); ETA-receptor blockade resulted in greater vasodilation (both P < .001); and plasma levels of fractalkine, E-selectin and monocyte chemoattractant protein (MCP)-1 were increased (all P < .05). In diabetic patients, oral antidiabetic drugs (glyburide, metformin or pioglitazone) reduced circulating levels fractalkine and E-selectin (both P < .05), without affecting vascular responses (all P > .05). Our findings indicate that insulin resistant states are associated with elevated atherogenic chemokines and impaired vascular reactivity. Antidiabetic treatment results in lower circulating fractalkine, which may provide cardiovascular benefits.
Collapse
Affiliation(s)
| | - Manfredi Tesauro
- Department of Internal Medicine, Università Tor Vergata, Roma, Italy
| | - Umberto Campia
- Vascular Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Carmine Cardillo
- Policlinico A. Gemelli IRCCS, Roma, Italy; Department of Internal Medicine, Università Cattolica del Sacro Cuore, Roma, Italy.
| |
Collapse
|
30
|
Bansal G, Thanikachalam PV, Maurya RK, Chawla P, Ramamurthy S. An overview on medicinal perspective of thiazolidine-2,4-dione: A remarkable scaffold in the treatment of type 2 diabetes. J Adv Res 2020; 23:163-205. [PMID: 32154036 PMCID: PMC7052407 DOI: 10.1016/j.jare.2020.01.008] [Citation(s) in RCA: 44] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2019] [Revised: 01/07/2020] [Accepted: 01/18/2020] [Indexed: 12/26/2022] Open
Abstract
TZDs, an important pharmacophore in the treatment of diabetes. Various analog-based synthetic strategies and biological significance are discussed. Clinical studies using TZDs along with other antidiabetic agents are also highlighted. SAR has been discussed to suggest the interactions between derivatives and receptor sites. Pyrazole, chromone, and acid-based TZDs can be considered as potential lead molecules.
Diabetes or diabetes mellitus is a complex or polygenic disorder, which is characterized by increased levels of glucose (hyperglycemia) and deficiency in insulin secretion or resistance to insulin over an elongated period in the liver and peripheral tissues. Thiazolidine-2,4-dione (TZD) is a privileged scaffold and an outstanding heterocyclic moiety in the field of drug discovery, which provides various opportunities in exploring this moiety as an antidiabetic agent. In the past few years, various novel synthetic approaches had been undertaken to synthesize different derivatives to explore them as more potent antidiabetic agents with devoid of side effects (i.e., edema, weight gain, and bladder cancer) of clinically used TZD (pioglitazone and rosiglitazone). In this review, an effort has been made to summarize the up to date research work of various synthetic strategies for TZD derivatives as well as their biological significance and clinical studies of TZDs in combination with other category as antidiabetic agents. This review also highlights the structure-activity relationships and the molecular docking studies to convey the interaction of various synthesized novel derivatives with its receptor site.
Collapse
Key Words
- ADDP, 1,1′-(Azodicarbonyl)dipiperidine
- AF, activation factor
- ALP, alkaline phosphatase
- ALT, alanine transaminase
- AST, aspartate transaminase
- Boc, Butyloxycarbonyl
- DBD, DNA-binding domain
- DCM, dichloromethane
- DM, diabetes mellitus
- DMF, dimethylformamide
- DMSO, dimethyl sulfoxide
- DNA, deoxyribonucleic acid
- Diabetes
- E, Entgegen
- ECG, electrocardiogram
- FDA, food and drug administration
- FFA, free fatty acid
- GAL4, Galactose transporter type
- GLUT4, glucose transporter type 4
- GPT, glutamic pyruvic transaminase
- HCl, Hydrochloric Acid
- HDL, high-density lipoprotein
- HEK, human embryonic kidney
- HEp-2, Human epithelial type 2
- HFD, high-fat diet
- IDF, international diabetes federation
- IL-β, interlukin-beta
- INS-1, insulin-secreting cells
- K2CO3, Potassium carbonate
- KOH, potassium hydroxide
- LBD, ligand-binding domain
- LDL, low-density lipoprotein
- MDA, malondialdehyde
- NA, nicotinamide
- NBS, N-bromosuccinimide
- NFκB, nuclear factor kappa-B
- NO, nitric oxide
- NaH, Sodium Hydride
- OGTT, oral glucose tolerance test
- PDB, protein data bank
- PPAR, peroxisome-proliferator activated receptor
- PPAR-γ
- PPRE, peroxisome proliferator response element
- PTP1B, protein-tyrosine phosphatase 1B
- Pd, Palladium
- Pioglitazone
- QSAR, quantitative structure-activity relationship
- RXR, retinoid X receptor
- Rosiglitazone
- SAR, structure-activity relationship
- STZ, streptozotocin
- T2DM, type 2 diabetes mellitus
- TFA, trifluoroacetic acid
- TFAA, trifluoroacetic anhydride
- TG, triglycerides
- THF, tetrahydrofuran
- TNF-α, tumor necrosis factor-alpha
- TZD, thiazolidine-2,4-dione
- Thiazolidine-2,4-diones
- WAT, white adipose tissue
- Z, Zusammen
- i.m, Intramuscular
- mCPBA, meta-chloroperoxybenzoic acid
Collapse
Affiliation(s)
- Garima Bansal
- Department of Pharmaceutical Chemistry, ISF College of Pharmacy, Ghal Kalan, Moga, Punjab 142001, India
| | - Punniyakoti Veeraveedu Thanikachalam
- Department of Pharmaceutical Chemistry, ISF College of Pharmacy, Ghal Kalan, Moga, Punjab 142001, India.,GRT Institute of Pharmaceutical Education and Research, GRT Mahalakshmi Nagar, Tiruttani, India
| | - Rahul K Maurya
- Department of Pharmaceutical Chemistry, ISF College of Pharmacy, Ghal Kalan, Moga, Punjab 142001, India.,Amity Institute of Pharmacy, Amity University Uttar Pradesh, Lucknow Campus, India
| | - Pooja Chawla
- Department of Pharmaceutical Chemistry, ISF College of Pharmacy, Ghal Kalan, Moga, Punjab 142001, India
| | - Srinivasan Ramamurthy
- College of Pharmacy and Health Sciences, University of Science and Technology of Fujairah, United Arab Emirates
| |
Collapse
|
31
|
de-Lima-Júnior JC, Rodovalho S, Van de Sande-Lee S, Monfort-Pires M, Rachid B, Cintra RM, Ramos CD, Cendes F, Folli F, Velloso LA. Effect of pioglitazone treatment on brown adipose tissue volume and activity and hypothalamic gliosis in patients with type 2 diabetes mellitus: a proof-of-concept study. Acta Diabetol 2019; 56:1333-1339. [PMID: 31506721 DOI: 10.1007/s00592-019-01418-2] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/29/2019] [Accepted: 08/29/2019] [Indexed: 02/06/2023]
Abstract
AIMS This study aimed to evaluate the effect of pioglitazone on brown adipose tissue function and hypothalamic gliosis in humans. Brown adipose tissue and the hypothalamus are regarded as important potential pharmacological targets to metabolic diseases, and defining the impact of current therapies on their structure and/or function could provide therapeutic advance in this field. METHODS Six patients with type 2 diabetes were treated for 24 weeks with pioglitazone 30 mg/day as an add-on therapy. Brown adipose tissue glucose uptake and volume were determined using 18F-FDG PET/CT scans; hypothalamic gliosis was determined using MRI scans; blood was collected for hormone and biochemistry measurements. All tests were performed at inclusion and six months after pioglitazone introduction. RESULTS Pioglitazone treatment led to a significant 3% body mass increase. There were neither changes in cold-induced brown adipose tissue glucose uptake and volume nor changes in hypothalamic gliosis. CONCLUSIONS This is a proof-of-concept study that provides clinical evidence for a lack of action of a thiazolidinedione, pioglitazone, to promote homogeneous and measurable changes in brown adipose tissue volume and also in hypothalamic gliosis after 6 months of treatment.
Collapse
Affiliation(s)
- José C de-Lima-Júnior
- Laboratory of Cell Signaling, Department of Internal Medicine, University of Campinas (UNICAMP), Campinas, São Paulo, Brazil
- Obesity and Comorbidities Research Center, Campinas, São Paulo, Brazil
| | - Sylka Rodovalho
- Laboratory of Cell Signaling, Department of Internal Medicine, University of Campinas (UNICAMP), Campinas, São Paulo, Brazil
- Obesity and Comorbidities Research Center, Campinas, São Paulo, Brazil
| | - Simone Van de Sande-Lee
- Laboratory of Cell Signaling, Department of Internal Medicine, University of Campinas (UNICAMP), Campinas, São Paulo, Brazil
- Department of Internal Medicine, Federal University of Santa Catarina (UFSC), Florianópolis, SC, Brazil
| | - Milena Monfort-Pires
- Laboratory of Cell Signaling, Department of Internal Medicine, University of Campinas (UNICAMP), Campinas, São Paulo, Brazil
- Obesity and Comorbidities Research Center, Campinas, São Paulo, Brazil
| | - Briana Rachid
- Laboratory of Cell Signaling, Department of Internal Medicine, University of Campinas (UNICAMP), Campinas, São Paulo, Brazil
- Obesity and Comorbidities Research Center, Campinas, São Paulo, Brazil
| | - Riobaldo M Cintra
- Obesity and Comorbidities Research Center, Campinas, São Paulo, Brazil
| | - Celso D Ramos
- Department of Radiology, University of Campinas, Campinas, São Paulo, 13084-970, Brazil
| | - Fernando Cendes
- Neuroimaging Laboratory, Department of Neurology, University of Campinas, Campinas, Brazil
| | - Franco Folli
- Obesity and Comorbidities Research Center, Campinas, São Paulo, Brazil
- School of Medicine, Endocrinology and Metabolism Dipartimento di Scienze Della Salute, Universita' degli Studi di Milano, Milan, Italy
- Departmental Unit of Diabetes and Metabolic Disorders, Azienda Socio-Sanitaria Santi Paolo e Carlo, Via A. Di Rudini, 8, 20143, Milan, Italy
| | - Lício A Velloso
- Laboratory of Cell Signaling, Department of Internal Medicine, University of Campinas (UNICAMP), Campinas, São Paulo, Brazil.
- Obesity and Comorbidities Research Center, Campinas, São Paulo, Brazil.
| |
Collapse
|
32
|
Guadagnini D, Rocha GZ, Santos A, Assalin HB, Hirabara SM, Curi R, Oliveira AG, Prada PO, Saad MJA. Microbiota determines insulin sensitivity in TLR2-KO mice. Life Sci 2019; 234:116793. [PMID: 31465735 DOI: 10.1016/j.lfs.2019.116793] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2019] [Revised: 08/17/2019] [Accepted: 08/25/2019] [Indexed: 01/04/2023]
Abstract
INTRODUCTION Environmental factors have a key role in the control of gut microbiota and obesity. TLR2 knockout (TLR2-/-) mice in some housing conditions are protected from diet-induced insulin resistance. However, in our housing conditions these animals are not protected from diet-induced insulin-resistance. AIM The aim of the present study was to investigate the influence of our animal housing conditions on the gut microbiota, glucose tolerance and insulin sensitivity in TLR2-/- mice. MATERIAL AND METHODS The microbiota was investigated by metagenomics, associated with hyperinsulinemic euglycemic clamp and GTT associated with insulin signaling through immunoblotting. RESULTS The results showed that TLR2-/- mice in our housing conditions presented a phenotype of metabolic syndrome characterized by insulin resistance, glucose intolerance and increase in body weight. This phenotype was associated with differences in microbiota in TLR2-/- mice that showed a decrease in the Proteobacteria and Bacteroidetes phyla and an increase in the Firmicutesphylum, associated with and in increase in the Oscillospira and Ruminococcus genera. Furthermore there is also an increase in circulating LPS and subclinical inflammation in TLR2-/-. The molecular mechanism that account for insulin resistance was an activation of TLR4, associated with ER stress and JNK activation. The phenotype and metabolic behavior was reversed by antibiotic treatment and reproduced in WT mice by microbiota transplantation. CONCLUSIONS Our data show, for the first time, that the intestinal microbiota can induce insulin resistance and obesity in an animal model that is genetically protected from these processes.
Collapse
Affiliation(s)
- Dioze Guadagnini
- Department of Internal Medicine-FCM, University of Campinas-UNICAMP, Campinas, SP, Brazil
| | - Guilherme Zweig Rocha
- Department of Internal Medicine-FCM, University of Campinas-UNICAMP, Campinas, SP, Brazil
| | - Andrey Santos
- Department of Internal Medicine-FCM, University of Campinas-UNICAMP, Campinas, SP, Brazil
| | - Heloisa Balan Assalin
- Department of Internal Medicine-FCM, University of Campinas-UNICAMP, Campinas, SP, Brazil
| | - Sandro Massao Hirabara
- Interdisciplinary Post-Graduate Program in Health Science, Cruzeiro do Sul University, Sao Paulo, Brazil
| | - Rui Curi
- Interdisciplinary Post-Graduate Program in Health Science, Cruzeiro do Sul University, Sao Paulo, Brazil
| | - Alexandre Gabarra Oliveira
- Department of Internal Medicine-FCM, University of Campinas-UNICAMP, Campinas, SP, Brazil.; Department of Physical Education, Biosciences Institute, São Paulo State University (UNESP), Rio Claro, SP, Brazil
| | - Patricia O Prada
- Graduate Program in Nutritional and Sport Sciences and Metabolism, School of Applied Sciences, University of Campinas- UNICAMP, Campinas, Brazil
| | - Mario J A Saad
- Department of Internal Medicine-FCM, University of Campinas-UNICAMP, Campinas, SP, Brazil..
| |
Collapse
|
33
|
Plaza A, Merino B, Del Olmo N, Ruiz-Gayo M. The cholecystokinin receptor agonist, CCK-8, induces adiponectin production in rat white adipose tissue. Br J Pharmacol 2019; 176:2678-2690. [PMID: 31012948 DOI: 10.1111/bph.14690] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2018] [Revised: 03/27/2019] [Accepted: 04/02/2019] [Indexed: 12/16/2022] Open
Abstract
BACKGROUND AND PURPOSE A cholecystokinin (CCK) system has been identified in white adipose tissue (WAT). Nevertheless, the endocrine actions of CCK on WAT remain unknown. Our goal was to investigate the role of CCK in regulating the production of adiponectin, an adipokine expressed in WAT, which is pivotal in preserving energy homeostasis. EXPERIMENTAL APPROACH The effect of the bioactive CCK fragment CCK-8 on adiponectin production was studied both in vivo and in vitro. CCK-8 effects were characterized in rats treated with selective CCK1 and CCK2 receptor antagonists as well as in pre-adipocytes carrying the selective silencing of either CCK1 or CCK2 receptors. The influence of insulin on CCK-8 responses was also analysed. KEY RESULTS In WAT, CCK-8 increased plasma adiponectin levels and the expression of the adiponectin gene (Adipoq). In pre-adipocytes, CCK-8 up-regulated adiponectin production. CCK-8 effects were abolished by L-365,260, a selective CCK2 receptor antagonist. CCK2 receptor knockdown also abolished the effects of CCK-8 in pre-adipocytes. Moreover, in vitro CCK-8 effects were blocked by triciribine, a specific inhibitor of protein kinase B (Akt) and by the PPARγ antagonist T0070907. Silencing the expression of the insulin receptor inhibited CCK-8-induced Adipoq expression in pre-adipocytes. Furthermore, insulin potentiated the effect of CCK-8. CONCLUSION AND IMPLICATIONS CCK-8 stimulates adiponectin production in WAT by acting on CCK2 receptors, through a mechanism involving both Akt and PPARγ. Moreover, CCK-8 actions are only observed in the presence of insulin. Our results could have translational value in the design of new insulin-sensitizing therapies.
Collapse
Affiliation(s)
- Adrián Plaza
- Departamento de Ciencias Farmacéuticas y de la Salud, Facultad de Farmacia, Universidad CEU San Pablo, Madrid, Spain
| | - Beatriz Merino
- Departamento de Ciencias Farmacéuticas y de la Salud, Facultad de Farmacia, Universidad CEU San Pablo, Madrid, Spain
| | - Nuria Del Olmo
- Departamento de Ciencias Farmacéuticas y de la Salud, Facultad de Farmacia, Universidad CEU San Pablo, Madrid, Spain
| | - Mariano Ruiz-Gayo
- Departamento de Ciencias Farmacéuticas y de la Salud, Facultad de Farmacia, Universidad CEU San Pablo, Madrid, Spain
| |
Collapse
|
34
|
Sartore G, Chilelli NC, Seraglia R, Ragazzi E, Marin R, Roverso M, Cosma C, Vaccaro O, Burlina S, Lapolla A. Long-term effect of pioglitazone vs glimepiride on lipoprotein oxidation in patients with type 2 diabetes: a prospective randomized study. Acta Diabetol 2019; 56:505-513. [PMID: 30740640 DOI: 10.1007/s00592-018-01278-2] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/13/2018] [Accepted: 12/14/2018] [Indexed: 01/07/2023]
Abstract
AIMS Type 2 diabetes (DM2) is associated to oxidative modifications of high-density lipoproteins (HDL), which can interfere with their function. Pioglitazone has proved effective in raising HDL cholesterol (HDL-C) and lowering small dense low-density lipoprotein (LDL), but no clinical studies have examined its effect on lipoprotein oxidation in patients with DM2. METHODS We assessed the effect of pioglitazone vs glimepiride after 1 year on HDL oxidation, expressed as relative abundance of peptides containing Met112O in ApoA-I (oxApoA-I) estimated by mass spectrometry (MALDI/TOF/TOF), in 95 patients with DM2. The oxLDL and AGE were quantified by ELISA. RESULTS Patients receiving pioglitazone showed a significant increase in the concentration of ApoA-I (Δ = 7.2 ± 14.8 mg/dL, p < 0.02) and a reduction in oxApoA-I (Δ = - 1.0 ± 2.6%, p < 0.02); this reduction was not significantly different from glimepiride. oxLDL showed a slight, but not significant increase in both treatment groups. Regression analysis showed a correlation between ΔoxApoA-I and ΔAGE (r = 0.30; p = 0.007) in all patients, while both of these parameters were unrelated to changes in HbA1c, HDL-C, duration of illness, or use of statins. CONCLUSIONS Long-term treatment with pioglitazone was effective in reducing the oxidation of HDL, but not LDL in patients with DM2, while glimepiride didn't. This finding seems to be associated to the change of glyco-oxidation status, not to any improvement in glycemic control or lipid profile. TRIAL REGISTRATION NCT00700856, ClinicalTrials.gov Registered June 18, 2008.
Collapse
Affiliation(s)
- Giovanni Sartore
- Diabetology and Dietetics, Department of Medicine (DIMED), University of Padova, via Giustiniani, 2, 35100, Padua, Italy
| | - Nino Cristiano Chilelli
- Diabetology and Dietetics, Department of Medicine (DIMED), University of Padova, via Giustiniani, 2, 35100, Padua, Italy.
| | - Roberta Seraglia
- National Research Council-Institute for Energy and Interphases, Padua, Italy
| | - Eugenio Ragazzi
- Department of Pharmaceutical and Pharmacological Sciences, University of Padova, Padua, Italy
| | - Raffaella Marin
- Lipid Laboratory, Department of Medicine (DIMED), University of Padova, Padua, Italy
| | - Marco Roverso
- National Research Council-Institute for Energy and Interphases, Padua, Italy
| | - Chiara Cosma
- Department of Laboratory Medicine, University of Padova, Padua, Italy
| | - Olga Vaccaro
- Department of Clinical Medicine and Surgery, Federico II University, Naples, Italy
| | - Silvia Burlina
- Diabetology and Dietetics, Department of Medicine (DIMED), University of Padova, via Giustiniani, 2, 35100, Padua, Italy
| | - Annunziata Lapolla
- Diabetology and Dietetics, Department of Medicine (DIMED), University of Padova, via Giustiniani, 2, 35100, Padua, Italy
| |
Collapse
|
35
|
Guo VY, Cao B, Cai C, Cheng KKY, Cheung BMY. Fetuin-A levels and risk of type 2 diabetes mellitus: a systematic review and meta-analysis. Acta Diabetol 2018; 55:87-98. [PMID: 29127490 DOI: 10.1007/s00592-017-1068-9] [Citation(s) in RCA: 40] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/04/2017] [Accepted: 10/23/2017] [Indexed: 12/31/2022]
Abstract
AIMS Fetuin-A has been linked to insulin resistance and obesity. Its role in the pathogenesis of type 2 diabetes (T2DM) has also been discussed. We aimed to investigate the prospective association of fetuin-A and the risk of T2DM in a systematic review and meta-analysis. METHODS A systematic search of studies from the MEDLINE, EMBASE, Pubmed and Web of Science using fetuin-A, diabetes and various synonyms was conducted up to June 5, 2017. Relevant studies were extracted by two reviewers independently. The quality of studies was assessed using Newcastle-Ottawa scales. Overall estimates were pooled using fixed effect with inverse variance meta-analysis. Subgroup analyses by gender, study population, techniques of assessing fetuin-A, diabetes ascertainment methods, follow-up duration and measures of association were conducted. RESULTS Seven studies comprising a total of 11,497 individuals and 2176 cases of T2DM were included in the systematic review and meta-analysis. Overall, one SD increment of fetuin-A level was associated with a 23% greater risk of incident T2DM (RR: 1.23, 95% CI 1.16-1.31). No significant heterogeneity or publication bias was found. The association was relatively stable across different subgroups. However, the association seemed only evident in women, but not in men. CONCLUSIONS Higher circulating fetuin-A levels were associated with increased risk of T2DM. However, the causality deserved further analysis.
Collapse
Affiliation(s)
- Vivian Yawei Guo
- Department of Family Medicine and Primary Care, Faculty of Medicine, The University of Hong Kong, 3F, Ap Lei Chau Clinic, 161 Main Street, Ap Lei Chau, Hong Kong.
| | - Bing Cao
- Department of Biomedical Sciences, City University of Hong Kong, Kowloon, Hong Kong
| | - Chunyan Cai
- Biostatistics/Epidemiology/Research Design Core, Center for Clinical and Translational Sciences, The University of Texas Health Science Center at Houston, Houston, TX, USA
| | - Kenneth King-Yip Cheng
- Department of Health Technology and Informatics, The Hong Kong Polytechnic University, Kowloon, Hong Kong
| | - Bernard Man Yung Cheung
- Department of Medicine, Faculty of Medicine, The University of Hong Kong, Hong Kong Island, Hong Kong
| |
Collapse
|
36
|
Chang YH, Hwu DW, Chang DM, An LW, Hsieh CH, Lee YJ. Renal function preservation with pioglitazone or with basal insulin as an add-on therapy for patients with type 2 diabetes mellitus. Acta Diabetol 2017; 54:561-568. [PMID: 28331986 DOI: 10.1007/s00592-017-0983-0] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/17/2016] [Accepted: 03/11/2017] [Indexed: 01/11/2023]
Abstract
AIMS Clinical outcome may differ owing to the distinct pharmacological characteristics of insulin sensitizers and insulin. This study was performed to compare the metabolic and renal function changes with add-on pioglitazone treatment versus basal insulin in patients with type 2 diabetes mellitus (DM) in whom sulfonylurea and metformin regimens failed. METHODS Patients who were consecutively managed in the diabetes comprehensive program with add-on pioglitazone or detemir/glargine treatment for at least 2 years following sulfonylurea and metformin treatment failure were included. RESULTS A total of 1002 patients were enrolled (pioglitazone: 559, detemir: 264, glargine: 179). After propensity score matching, there were 105 patients with matchable baseline characteristics in each group. After a mean of 3.5 years of follow-up, the pioglitazone group showed a greater HbA1c reduction than the detemir group and the glargine group. Despite patients in all three groups exhibiting significant body weight gain, those in the pioglitazone group and the glargine group showed greater body weight increases than the patients in the detemir group (2.1, 1.6 and 0.8 kg, respectively, p < 0.05). Interestingly, Cox regression analysis indicated that patients under detemir or glargine treatment had a higher probability of CKD progression as compared with the pioglitazone group, with hazard ratios of 2.63 (95% CI 1.79-3.88) and 3.13 (95% CI 2.01-4.87), respectively. CONCLUSIONS Our study first showed that treatment with both pioglitazone and basal insulin improved glycemic control, while only pioglitazone treatment was observed to be advantageous in terms of preserving renal function when used as an add-on therapy for patients with type 2 DM in whom sulfonylurea and metformin regimens failed.
Collapse
Affiliation(s)
- Yu-Hung Chang
- Department of Internal Medicine, Lee's Endocrinology Clinic, # 130 Min-Tzu Rd, Pingtung, 90000, Taiwan.
| | - Der-Wei Hwu
- Department of Internal Medicine, Lee's Endocrinology Clinic, # 130 Min-Tzu Rd, Pingtung, 90000, Taiwan
- Graduate Institute of Clinical Medicine, Kaohsiung Medical University, Kaohsiung, Taiwan
| | - Dao-Ming Chang
- Department of Internal Medicine, Lee's Endocrinology Clinic, # 130 Min-Tzu Rd, Pingtung, 90000, Taiwan
| | - Ling-Wang An
- Department of Medical Education, Beijing Ruijing Diabetes Hospital, Beijing, 100078, China
| | - Chang-Hsun Hsieh
- Division of Endocrinology and Metabolism, Department of Internal MedicineNational Defense Medical Center, Tri-Service General Hospital, Taipei, Taiwan
| | - Yau-Jiunn Lee
- Department of Internal Medicine, Lee's Endocrinology Clinic, # 130 Min-Tzu Rd, Pingtung, 90000, Taiwan
| |
Collapse
|
37
|
Garbossa SG, Folli F. Vitamin D, sub-inflammation and insulin resistance. A window on a potential role for the interaction between bone and glucose metabolism. Rev Endocr Metab Disord 2017; 18:243-258. [PMID: 28409320 DOI: 10.1007/s11154-017-9423-2] [Citation(s) in RCA: 63] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Vitamin D is a key hormone involved in the regulation of calcium/phosphorous balance and recently it has been implicated in the pathogenesis of sub-inflammation, insulin resistance and obesity. The two main forms of vitamin D are cholecalciferol (Vitamin D3) and ergocalciferol (Vitamin D2): the active form (1,25-dihydroxyvitamin D) is the result of two hydroxylations that take place in liver, kidney, pancreas and immune cells. Vitamin D increases the production of some anti-inflammatory cytokines and reduces the release of some pro-inflammatory cytokines. Low levels of Vitamin D are also associated with an up-regulation of TLRs expression and a pro-inflammatory state. Regardless of the effect on inflammation, Vitamin D seems to directly increase insulin sensitivity and secretion, through different mechanisms. Considering the importance of low grade chronic inflammation in metabolic syndrome, obesity and diabetes, many authors hypothesized the involvement of this nutrient/hormone in the pathogenesis of these diseases. Vitamin D status could alter the balance between pro and anti-inflammatory cytokines and thus affect insulin action, lipid metabolism and adipose tissue function and structure. Numerous studies have shown that Vitamin D concentrations are inversely associated with pro-inflammatory markers, insulin resistance, glucose intolerance and obesity. Interestingly, some longitudinal trials suggested also an inverse association between vitamin D status and incident type 2 diabetes mellitus. However, vitamin D supplementation in humans showed controversial effects: with some studies demonstrating improvements in insulin sensitivity, glucose and lipid metabolism while others showing no beneficial effect on glycemic control and on inflammation. In conclusion, although the evidences of a significant role of Vitamin D on inflammation, insulin resistance and insulin secretion in the pathogenesis of obesity, metabolic syndrome and type 2 diabetes, its potential function in treatment and prevention of type 2 diabetes mellitus is unclear. Encouraging results have emerged from Vitamin D supplementation trials on patients at risk of developing diabetes and further studies are needed to fully explore and understand its clinical applications.
Collapse
Affiliation(s)
- Stefania Giuliana Garbossa
- Department of Internal Medicine, Azienda Socio Sanitaria Territoriale Santi Paolo e Carlo, Ospedale San Paolo, Via A. Di Rudini 8, Milan, 20142, Italy
- Program in Nutritional Science, Universitá degli Studi di Milano, Milan, Italy
| | - Franco Folli
- Department of Internal Medicine, Azienda Socio Sanitaria Territoriale Santi Paolo e Carlo, Ospedale San Paolo, Via A. Di Rudini 8, Milan, 20142, Italy.
- Endocrinology and Metabolism, Department of Health Science, Universitá degli Studi di Milano, San Paolo Hospital, Room #737, Via A. Di Rudiní 8, 20142, Milan, Italy.
- Department of Medicine, Diabetes Division, University of Texas Health System, 7703 Floyd Curl Drive, San Antonio, 78229, TX, USA.
| |
Collapse
|
38
|
McEniery CM, Wilkinson IB, Johansen NB, Witte DR, Singh-Manoux A, Kivimaki M, Tabak AG, Brunner EJ, Shipley MJ. Nondiabetic Glucometabolic Status and Progression of Aortic Stiffness: The Whitehall II Study. Diabetes Care 2017; 40:599-606. [PMID: 28122839 PMCID: PMC5360278 DOI: 10.2337/dc16-1773] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/16/2016] [Accepted: 01/06/2017] [Indexed: 02/03/2023]
Abstract
OBJECTIVE Aortic stiffness is an important predictor of future morbidity and mortality. Diabetes is associated with increased aortic stiffness, but the importance of nondiabetic glucometabolic status for accelerated aortic stiffening is unclear. We tested the hypothesis that adverse glucometabolic status is associated with accelerated aortic stiffening in individuals without diabetes, independently of known risk factors for arterial stiffening. RESEARCH DESIGN AND METHODS Glucometabolic status and other cardiovascular risk factors were assessed at baseline in 2008-09, and carotid femoral pulse wave velocity (cfPWV) at baseline and follow-up in 2012-13, in 4,386 participants without diabetes of the Whitehall II Study. RESULTS The mean age of the cohort at cfPWV baseline was 60 years, and 74% were male. cfPWV increased from (mean ± SE) 8.30 ± 0.03 to 8.98 ± 0.04 m/s during 4 years of follow-up. At baseline, cfPWV was associated with fasting and 2-h postload glucose, HbA1c, and HOMA-insulin resistance (HOMA-IR). HbA1c and HOMA-IR were associated with progression of cfPWV after adjusting for physiological confounders and cardiovascular risk factors. A 1 SD higher HbA1c and HOMA-IR were associated with greater increases in cfPWV (0.11 m/s per 5 years [95% CI 0.04, 0.18], P = 0.003 and 0.09 m/s per 5 years [0.01, 0.17], P = 0.03, respectively). Additional adjustment for BMI weakened the association with HOMA-IR but not with HbA1c. CONCLUSIONS HbA1c is independently associated with accelerated progression of aortic stiffness in individuals without diabetes. These findings suggest that long-term glucometabolic status, even in individuals without diabetes, could be an important target for preventative strategies against vascular aging.
Collapse
Affiliation(s)
- Carmel M McEniery
- Division of Experimental Medicine and Immunotherapeutics, University of Cambridge, Cambridge, U.K.
| | - Ian B Wilkinson
- Division of Experimental Medicine and Immunotherapeutics, University of Cambridge, Cambridge, U.K
| | - Nanna B Johansen
- Steno Diabetes Center A/S, Gentofte, Denmark.,Danish Diabetes Academy, Odense, Denmark
| | - Daniel R Witte
- Danish Diabetes Academy, Odense, Denmark.,Department of Public Health, Aarhus University, Aarhus, Denmark
| | - Archana Singh-Manoux
- Research Department of Epidemiology and Public Health, University College London, London, U.K
| | - Mika Kivimaki
- Research Department of Epidemiology and Public Health, University College London, London, U.K
| | - Adam G Tabak
- Research Department of Epidemiology and Public Health, University College London, London, U.K.,1st Department of Medicine, Faculty of Medicine, Semmelweis University, Budapest, Hungary
| | - Eric J Brunner
- Research Department of Epidemiology and Public Health, University College London, London, U.K
| | - Martin J Shipley
- Research Department of Epidemiology and Public Health, University College London, London, U.K
| |
Collapse
|
39
|
Saad MJA, Santos A, Prada PO. Linking Gut Microbiota and Inflammation to Obesity and Insulin Resistance. Physiology (Bethesda) 2017; 31:283-93. [PMID: 27252163 DOI: 10.1152/physiol.00041.2015] [Citation(s) in RCA: 460] [Impact Index Per Article: 57.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Obesity and insulin resistance are the major predisposing factors to comorbidities, such as Type 2 diabetes, nonalcoholic fatty liver disease, cardiovascular and neurodegenerative diseases, and several types of cancer. The prevalence of obesity is still increasing worldwide and now affects a large number of individuals. Here, we review the role of the gut microbiota in the pathophysiology of insulin resistance/obesity. The human intestine is colonized by ∼100 trillion bacteria, which constitute the gut microbiota. Studies have shown that lean and overweight rodents and humans may present differences in the composition of their intestinal flora. Over the past 10 years, data from different sources have established a causal link between the intestinal microbiota and obesity/insulin resistance. It is important to emphasize that diet-induced obesity promotes insulin resistance by mechanisms independent and dependent on gut microbiota. In this review, we present several mechanisms that contribute to explaining the link between intestinal flora and insulin resistance/obesity. The LPS from intestinal flora bacteria can induce a chronic subclinical inflammatory process and obesity, leading to insulin resistance through activation of TLR4. The reduction in circulating SCFA may also have an essential role in the installation of reduced insulin sensitivity and obesity. Other mechanisms include effects of bile acids, branched-chain amino acids (BCAA), and some other lesser-known factors. In the near future, this area should open new therapeutic avenues for obesity/insulin resistance and its comorbidities.
Collapse
Affiliation(s)
- M J A Saad
- Department of Internal Medicine, State University of Campinas (UNICAMP), Campinas, Brazil; and
| | - A Santos
- Department of Internal Medicine, State University of Campinas (UNICAMP), Campinas, Brazil; and
| | - P O Prada
- Department of Internal Medicine, State University of Campinas (UNICAMP), Campinas, Brazil; and School of Applied Sciences, State University of Campinas (UNICAMP), Limeira, Brazil
| |
Collapse
|
40
|
Zanotto TM, Quaresma PGF, Guadagnini D, Weissmann L, Santos AC, Vecina JF, Calisto K, Santos A, Prada PO, Saad MJA. Blocking iNOS and endoplasmic reticulum stress synergistically improves insulin resistance in mice. Mol Metab 2016; 6:206-218. [PMID: 28180062 PMCID: PMC5279911 DOI: 10.1016/j.molmet.2016.12.005] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/07/2016] [Revised: 12/02/2016] [Accepted: 12/12/2016] [Indexed: 12/12/2022] Open
Abstract
OBJECTIVE Recent data show that iNOS has an essential role in ER stress in obesity. However, whether iNOS is sufficient to account for obesity-induced ER stress and Unfolded Protein Response (UPR) has not yet been investigated. In the present study, we used iNOS knockout mice to investigate whether high-fat diet (HFD) can still induce residual ER stress-associated insulin resistance. METHODS For this purpose, we used the intraperitoneal glucose tolerance test (GTT), euglycemic-hyperinsulinemic clamp, western blotting and qPCR in liver, muscle, and adipose tissue of iNOS KO and control mice on HFD. RESULTS The results of the present study demonstrated that, in HFD fed mice, iNOS-induced alteration in insulin signaling is an essential mechanism of insulin resistance in muscle, suggesting that iNOS may represent an important target that could be blocked in order to improve insulin sensitivity in this tissue. However, in liver and adipose tissue, the insulin resistance induced by HFD was only partially dependent on iNOS, and, even in the presence of genetic or pharmacological blockade of iNOS, a clear ER stress associated with altered insulin signaling remained evident in these tissues. When this ER stress was blocked pharmacologically, insulin signaling was improved, and a complete recovery of glucose tolerance was achieved. CONCLUSIONS Taken together, these results reinforce the tissue-specific regulation of insulin signaling in obesity, with iNOS being sufficient to account for insulin resistance in muscle, but in liver and adipose tissue ER stress and insulin resistance can be induced by both iNOS-dependent and iNOS-independent mechanisms.
Collapse
Key Words
- AKT, Protein kinase B
- ATF6, activating transcription factor 6
- Blocking
- ER, endoplasmic reticulum
- Endoplasmic reticulum stress
- GAPDH, glyceraldehyde 3-phosphate dehydrogenase
- GTT, glucose tolerance test
- HFD, high-fat diet
- IKK, kappa α/β kinase
- IRE1, inositol requiring enzyme 1
- ITT, insulin tolerance test
- Improving
- Insulin resistance
- JNK, c-JunN-terminal kinase
- NO, nitric oxide
- PERK, protein kinase RNA-like ER kinase
- UPR, unfolded protein response
- iNOS
- iNOS, inducible nitric oxide synthase
- qPCR, real time PCR
Collapse
Affiliation(s)
- Tamires M Zanotto
- Department of Internal Medicine, State University of Campinas (UNICAMP), Campinas, SP, Brazil; Department of Medical Clinics, Obesity and Comorbidities Research Center (O.C.R.C.), State University of Campinas (UNICAMP), Campinas, SP, Brazil
| | - Paula G F Quaresma
- Department of Internal Medicine, State University of Campinas (UNICAMP), Campinas, SP, Brazil; Department of Medical Clinics, Obesity and Comorbidities Research Center (O.C.R.C.), State University of Campinas (UNICAMP), Campinas, SP, Brazil
| | - Dioze Guadagnini
- Department of Internal Medicine, State University of Campinas (UNICAMP), Campinas, SP, Brazil
| | - Lais Weissmann
- Department of Internal Medicine, State University of Campinas (UNICAMP), Campinas, SP, Brazil
| | - Andressa C Santos
- Department of Internal Medicine, State University of Campinas (UNICAMP), Campinas, SP, Brazil
| | - Juliana F Vecina
- Department of Internal Medicine, State University of Campinas (UNICAMP), Campinas, SP, Brazil; Department of Medical Clinics, Obesity and Comorbidities Research Center (O.C.R.C.), State University of Campinas (UNICAMP), Campinas, SP, Brazil
| | - Kelly Calisto
- Department of Internal Medicine, State University of Campinas (UNICAMP), Campinas, SP, Brazil; Department of Medical Clinics, Obesity and Comorbidities Research Center (O.C.R.C.), State University of Campinas (UNICAMP), Campinas, SP, Brazil
| | - Andrey Santos
- Department of Internal Medicine, State University of Campinas (UNICAMP), Campinas, SP, Brazil
| | - Patrícia O Prada
- Department of Internal Medicine, State University of Campinas (UNICAMP), Campinas, SP, Brazil; School of Applied Sciences, State University of Campinas (UNICAMP), Limeira, SP, Brazil; Department of Medical Clinics, Obesity and Comorbidities Research Center (O.C.R.C.), State University of Campinas (UNICAMP), Campinas, SP, Brazil
| | - Mario J A Saad
- Department of Internal Medicine, State University of Campinas (UNICAMP), Campinas, SP, Brazil; Department of Medical Clinics, Obesity and Comorbidities Research Center (O.C.R.C.), State University of Campinas (UNICAMP), Campinas, SP, Brazil.
| |
Collapse
|
41
|
Kaji H. Adipose Tissue‐Derived Plasminogen Activator Inhibitor‐1 Function and Regulation. Compr Physiol 2016; 6:1873-1896. [DOI: 10.1002/cphy.c160004] [Citation(s) in RCA: 57] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
|
42
|
Anderson JR, Mortimer K, Pang L, Smith KM, Bailey H, Hodgson DB, Shaw DE, Knox AJ, Harrison TW. Evaluation of the PPAR-γ Agonist Pioglitazone in Mild Asthma: A Double-Blind Randomized Controlled Trial. PLoS One 2016; 11:e0160257. [PMID: 27560168 PMCID: PMC4999189 DOI: 10.1371/journal.pone.0160257] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2016] [Accepted: 07/15/2016] [Indexed: 11/18/2022] Open
Abstract
Background Peroxisome proliferator-activated receptor gamma (PPAR-γ) is a nuclear receptor that modulates inflammation in models of asthma. To determine whether pioglitazone improves measures of asthma control and airway inflammation, we performed a single-center randomized, double-blind, placebo-controlled, parallel-group trial. Methods Sixty-eight participants with mild asthma were randomized to 12 weeks pioglitazone (30 mg for 4 weeks, then 45 mg for 8 weeks) or placebo. The primary outcome was the adjusted mean forced expiratory volume in one second (FEV1) at 12 weeks. The secondary outcomes were mean peak expiratory flow (PEF), scores on the Juniper Asthma Control Questionnaire (ACQ) and Asthma Quality of Life Questionnaire (AQLQ), fractional exhaled nitric oxide (FeNO), bronchial hyperresponsiveness (PD20), induced sputum counts, and sputum supernatant interferon gamma-inducible protein-10 (IP-10), vascular endothelial growth factor (VEGF), monocyte chemotactic protein-1 (MCP-1), and eosinophil cationic protein (ECP) levels. Study recruitment was closed early after considering the European Medicines Agency’s reports of a potential increased risk of bladder cancer with pioglitazone treatment. Fifty-five cases were included in the full analysis (FA) and 52 in the per-protocol (PP) analysis. Results There was no difference in the adjusted FEV1 at 12 weeks (-0.014 L, 95% confidence interval [CI] -0.15 to 0.12, p = 0.84) or in any of the secondary outcomes in the FA. The PP analysis replicated the FA, with the exception of a lower evening PEF in the pioglitazone group (-21 L/min, 95% CI -39 to -4, p = 0.02). Conclusions We found no evidence that treatment with 12 weeks of pioglitazone improved asthma control or airway inflammation in mild asthma. Trial Registration ClinicalTrials.gov NCT01134835
Collapse
Affiliation(s)
- J. R. Anderson
- Nottingham Respiratory Research Unit, University of Nottingham, Clinical Sciences Building, City Hospital, Hucknall Road, Nottingham, NG5 1PB, United Kingdom
| | - K. Mortimer
- Liverpool School of Tropical Medicine, Liverpool, UK and Aintree University Hospital NHS Foundation Trust, Fazakerley, United Kingdom
- * E-mail:
| | - L. Pang
- Nottingham Respiratory Research Unit, University of Nottingham, Clinical Sciences Building, City Hospital, Hucknall Road, Nottingham, NG5 1PB, United Kingdom
| | - K. M Smith
- Nottingham Respiratory Research Unit, University of Nottingham, Clinical Sciences Building, City Hospital, Hucknall Road, Nottingham, NG5 1PB, United Kingdom
| | - H. Bailey
- Nottingham Respiratory Research Unit, University of Nottingham, Clinical Sciences Building, City Hospital, Hucknall Road, Nottingham, NG5 1PB, United Kingdom
| | - D. B. Hodgson
- Nottingham Respiratory Research Unit, University of Nottingham, Clinical Sciences Building, City Hospital, Hucknall Road, Nottingham, NG5 1PB, United Kingdom
| | - D. E. Shaw
- Nottingham Respiratory Research Unit, University of Nottingham, Clinical Sciences Building, City Hospital, Hucknall Road, Nottingham, NG5 1PB, United Kingdom
| | - A. J. Knox
- Nottingham Respiratory Research Unit, University of Nottingham, Clinical Sciences Building, City Hospital, Hucknall Road, Nottingham, NG5 1PB, United Kingdom
| | - T. W. Harrison
- Nottingham Respiratory Research Unit, University of Nottingham, Clinical Sciences Building, City Hospital, Hucknall Road, Nottingham, NG5 1PB, United Kingdom
| |
Collapse
|
43
|
Jarczok MN, Koenig J, Li J, Mauss D, Hoffmann K, Schmidt B, Fischer JE, Thayer JF. The Association of Work Stress and Glycemic Status Is Partially Mediated by Autonomic Nervous System Function: Cross-Sectional Results from the Mannheim Industrial Cohort Study (MICS). PLoS One 2016; 11:e0160743. [PMID: 27532642 PMCID: PMC4988666 DOI: 10.1371/journal.pone.0160743] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2014] [Accepted: 07/25/2016] [Indexed: 11/26/2022] Open
Abstract
BACKGROUND Work stress is associated with an increased risk of pre-diabetes, Type 2 diabetes, and inflammation, as well as decreased autonomic nervous system function as measured, for example, via heart rate variability. We investigated the extent to which the association between work stress and glycemic status is mediated by vagally-mediated heart rate variability (vmHRV) and/or inflammation. METHODS Cross-sectional data from the Mannheim Industrial Cohort Study (MICS) with 9,937 participants were analyzed. The root mean squared successive differences (RMSSD) from long-term heart rate monitoring during work and night time periods was used to index vmHRV. Fasting plasma glucose and glycosylated hemoglobin were assessed to determine glycemic status. High sensitive C-reactive protein levels were observed as a measure of systemic inflammation and the Effort-Reward-Imbalance scale was used to evaluate work stress. Mediation models were adjusted for age, sex, and occupational status, and estimations were bootstrapped (5,000 replications). RESULTS Effort-Reward-Imbalance was significantly negatively associated with RMSSD and both glycosylated hemoglobin and fasting plasma glucose during both work and night time periods. Effort-Reward-Imbalance was observed to have a significant direct effect on glycosylated hemoglobin and significant indirect effects, through RMSSD, on both glycemic measures during both time periods. Introducing C-reactive protein as a further mediator to the model did not alter the indirect effects observed. C-reactive protein, as an exclusive mediator, was observed to have smaller direct and indirect effects on the glycemic measures as compared to when Effort-Reward-Imbalance was included in the model. CONCLUSIONS Our results suggest that the association between work stress and glycemic status is partially mediated through vmHRV independent of systemic inflammation as measured by C-reactive protein. We conclude that work stress may be an additional factor that promotes development of hyperglycemic-metabolic states. If supported by prospective evidence, these results may lead to new approaches for primary prevention of hyperglycemia in the workplace.
Collapse
Affiliation(s)
- Marc N. Jarczok
- Mannheim Institute of Public Health, Social and Preventive Medicine, Mannheim Medical Faculty, Heidelberg University, Ludolf–Krehl–Strasse 7–11, 68167, Mannheim, Germany
- Institute of Medical Psychology, Center for Psychosocial Medicine, UniversityHospital Heidelberg, Heidelberg, Germany, Bergheimer Strasse 20, 69115, Heidelberg, Germany
| | - Julian Koenig
- The Ohio State University, Department of Psychology, 1835 Neil Avenue, 43210, Columbus, Ohio, United States of America
| | - Jian Li
- Institute of Occupational, Social, and Environmental Medicine, Center for Health and Society, Faculty of Medicine, University of Düsseldorf, Universitätsstrasse 1, 40225 Düsseldorf, Germany
| | - Daniel Mauss
- Department of Occupational Medicine Allianz SE, Königinstrasse 28 80802, Munich, Germany
| | - Kristina Hoffmann
- Mannheim Institute of Public Health, Social and Preventive Medicine, Mannheim Medical Faculty, Heidelberg University, Ludolf–Krehl–Strasse 7–11, 68167, Mannheim, Germany
| | - Burkhard Schmidt
- Mannheim Institute of Public Health, Social and Preventive Medicine, Mannheim Medical Faculty, Heidelberg University, Ludolf–Krehl–Strasse 7–11, 68167, Mannheim, Germany
| | - Joachim E. Fischer
- Mannheim Institute of Public Health, Social and Preventive Medicine, Mannheim Medical Faculty, Heidelberg University, Ludolf–Krehl–Strasse 7–11, 68167, Mannheim, Germany
| | - Julian F. Thayer
- The Ohio State University, Department of Psychology, 1835 Neil Avenue, 43210, Columbus, Ohio, United States of America
| |
Collapse
|
44
|
Andreozzi F, Raciti GA, Nigro C, Mannino GC, Procopio T, Davalli AM, Beguinot F, Sesti G, Miele C, Folli F. The GLP-1 receptor agonists exenatide and liraglutide activate Glucose transport by an AMPK-dependent mechanism. J Transl Med 2016; 14:229. [PMID: 27473212 PMCID: PMC4967343 DOI: 10.1186/s12967-016-0985-7] [Citation(s) in RCA: 64] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2016] [Accepted: 07/20/2016] [Indexed: 01/11/2023] Open
Abstract
Aims/hypothesis Potentiation of glucose-induced insulin secretion is the main mechanism of exenatide (EXE) antidiabetic action, however, increased glucose utilization by peripheral tissues has been also reported. We here studied the effect of EXE on glucose uptake by skeletal muscle cells. Methods 2-deoxy-glucose (2DG) uptake and intracellular signal pathways were measured in rat L6 skeletal muscle myotubes exposed to 100 nmol/l EXE for up to 48 h. Mechanisms of EXE action were explored by inhibiting AMPK activity with compound C (CC, 40 μmol/l) or siRNAs (2 μmol/l). Results Time course experiments show that EXE increases glucose uptake up to 48 h achieving its maximal effect, similar to that induced by insulin, after 20 min (2- vs 2.5-fold-increase, respectively). Differently from insulin, EXE does not stimulate: (i) IR β-subunit- and IRS1 tyrosine phosphorylation and binding to p85 regulatory subunit of PI-3kinase; (ii) AKT activation; and (iii) ERK1/2 and JNK1/2 phosphorylation. Conversely, EXE increases phosphorylation of α-subunit of AMPK at Thr172 by 2.5-fold (p < 0.01). Co-incubation of EXE and insulin does not induce additive effects on 2DG-uptake. Inhibition of AMPK with CC, and reduction of AMPK protein expression by siRNA, completely abolish EXE-induced 2DG-uptake. Liraglutide, another GLP-1 receptor agonist, also stimulates AMPK phosphorylation and 2DG-uptake. Moreover, EXE stimulates 2DG-uptake also by L6 myotubes rendered insulin-resistant with methylglyoxal. Finally, EXE also induces glucose transporter Glut-4 translocation to the plasma membrane. Conclusions/interpretation In L6 myotubes, EXE and liraglutide increase glucose uptake in an insulin-independent manner by activating AMPK. Electronic supplementary material The online version of this article (doi:10.1186/s12967-016-0985-7) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Francesco Andreozzi
- Department of Medical and Surgical Sciences, University of Catanzaro "Magna-Graecia", Catanzaro, Italy. .,Division of Diabetes, Department of Medicine, University of Texas Health Science Center, San Antonio, TX, USA.
| | - Gregory Alexander Raciti
- Institute of Experimental Endocrinology and Oncology "G. Salvatore", National Council of Research, Naples, Italy.,Department of Translational Medical Sciences, University of Naples "Federico II", Naples, Italy
| | - Cecilia Nigro
- Institute of Experimental Endocrinology and Oncology "G. Salvatore", National Council of Research, Naples, Italy.,Department of Translational Medical Sciences, University of Naples "Federico II", Naples, Italy
| | - Gaia Chiara Mannino
- Department of Medical and Surgical Sciences, University of Catanzaro "Magna-Graecia", Catanzaro, Italy
| | - Teresa Procopio
- Department of Medical and Surgical Sciences, University of Catanzaro "Magna-Graecia", Catanzaro, Italy
| | - Alberto M Davalli
- Department of Medicine Endocrinology Unit, Ospedale San Raffaele, Milan, Italy
| | - Francesco Beguinot
- Institute of Experimental Endocrinology and Oncology "G. Salvatore", National Council of Research, Naples, Italy.,Department of Translational Medical Sciences, University of Naples "Federico II", Naples, Italy
| | - Giorgio Sesti
- Department of Medical and Surgical Sciences, University of Catanzaro "Magna-Graecia", Catanzaro, Italy
| | - Claudia Miele
- Institute of Experimental Endocrinology and Oncology "G. Salvatore", National Council of Research, Naples, Italy.,Department of Translational Medical Sciences, University of Naples "Federico II", Naples, Italy
| | - Franco Folli
- Division of Diabetes, Department of Medicine, University of Texas Health Science Center, San Antonio, TX, USA. .,Department of Internal Medicine, University of Campinas, Campinas, SP, Brazil.
| |
Collapse
|
45
|
Dreymueller D, Ludwig A. Considerations on inhibition approaches for proinflammatory functions of ADAM proteases. Platelets 2016; 28:354-361. [PMID: 27460023 DOI: 10.1080/09537104.2016.1203396] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Proteases of the disintegrin and metalloproteinase (ADAM) family mediate the proteolytic shedding of various surface molecules including cytokine precursors, adhesion molecules, growth factors, and receptors. Within the vasculature ADAM10 and ADAM17 regulate endothelial permeability, transendothelial leukocyte migration, and the adhesion of leukocytes and platelets. In vivo studies show that both proteases are implicated in several inflammatory pathologies, for example, edema formation, leukocyte infiltration, and thrombosis. However, both proteases also contribute to developmental and regenerative processes. Thus, although ADAMs can be regarded as valuable drug targets in many aspects, the danger of severe side effects is clearly visible. To circumvent these side effects, traditional inhibition approaches have to be improved to target ADAMs at the right time in the right place. Moreover, the inhibitors need to be more selective for the target protease and if possible also for the substrate. Antibodies recognizing the active conformation of ADAMs or small molecules blocking exosites of ADAM proteases may represent inhibitors with the desired selectivities.
Collapse
Affiliation(s)
- Daniela Dreymueller
- a Institute of Pharmacology and Toxicology , RWTH Aachen University , Aachen , Germany
| | - Andreas Ludwig
- a Institute of Pharmacology and Toxicology , RWTH Aachen University , Aachen , Germany
| |
Collapse
|
46
|
Stirban AO, Andjelkovic M, Heise T, Nosek L, Fischer A, Gastaldelli A, Herz M. Aleglitazar, a dual peroxisome proliferator-activated receptor-α/γ agonist, improves insulin sensitivity, glucose control and lipid levels in people with type 2 diabetes: findings from a randomized, double-blind trial. Diabetes Obes Metab 2016; 18:711-5. [PMID: 26663152 DOI: 10.1111/dom.12620] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/03/2015] [Revised: 08/02/2015] [Accepted: 11/30/2015] [Indexed: 12/01/2022]
Abstract
The present single-centre, randomized, double-blind, placebo-controlled phase II study investigated the effect of the balanced dual peroxisome proliferator-activated receptor-α/γ agonist aleglitazar on whole-body and liver insulin sensitivity, β-cell function and other components of cardiometabolic syndrome after 16 weeks of treatment in patients with type 2 diabetes inadequately controlled with metformin monotherapy who received once-daily 150 µg aleglitazar or matching placebo as add-on therapy to metformin. Baseline and 16-week assessments included a two-step hyperinsulinaemic-euglycaemic clamp, followed by a hyperglycaemic clamp, as well as evaluation of glycated haemoglobin (HbA1c), lipids and safety variables. The primary endpoint was change in whole-body insulin sensitivity (M-value) from baseline compared with placebo, derived from the second clamp step. M-value improved significantly from baseline with aleglitazar (n = 16) compared with placebo (n = 24; p = 0.05 for difference between arms). We found statistically significant treatment differences with aleglitazar versus placebo in fasting hepatic insulin resistance index (p = 0.01), and in total glucose disposal (p = 0.03) at the second insulin infusion step. Aleglitazar treatment resulted in significant improvements in HbA1c and lipids and was well tolerated.
Collapse
Affiliation(s)
| | | | | | | | | | | | - M Herz
- F. Hoffmann-La Roche Ltd, Basel, Switzerland
| |
Collapse
|
47
|
Finding novel distinctions between the sAPPα-mediated anabolic biochemical pathways in Autism Spectrum Disorder and Fragile X Syndrome plasma and brain tissue. Sci Rep 2016; 6:26052. [PMID: 27212113 PMCID: PMC4876513 DOI: 10.1038/srep26052] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2015] [Accepted: 04/25/2016] [Indexed: 02/07/2023] Open
Abstract
Autism spectrum disorder (ASD) and Fragile X syndrome (FXS) are developmental disorders. No validated blood-based biomarkers exist for either, which impedes bench-to-bedside approaches. Amyloid-β (Aβ) precursor protein (APP) and metabolites are usually associated with Alzheimer’s disease (AD). APP cleavage by α-secretase produces potentially neurotrophic secreted APPα (sAPPα) and the P3 peptide fragment. β-site APP cleaving enzyme (BACE1) cleavage produces secreted APPβ (sAPPβ) and intact Aβ. Excess Aβ is potentially neurotoxic and can lead to atrophy of brain regions such as amygdala in AD. By contrast, amygdala is enlarged in ASD but not FXS. We previously reported elevated levels of sAPPα in ASD and FXS vs. controls. We now report elevated plasma Aβ and total APP levels in FXS compared to both ASD and typically developing controls, and elevated levels of sAPPα in ASD and FXS vs. controls. By contrast, plasma and brain sAPPβ and Aβ were lower in ASD vs. controls but elevated in FXS plasma vs. controls. We also detected age-dependent increase in an α-secretase in ASD brains. We report a novel mechanistic difference in APP pathways between ASD (processing) and FXS (expression) leading to distinct APP metabolite profiles in these two disorders. These novel, distinctive biochemical differences between ASD and FXS pave the way for blood-based biomarkers for ASD and FXS.
Collapse
|
48
|
Vu A, Kosmiski LA, Beitelshees AL, Prigeon R, Sidhom MS, Bredbeck B, Predhomme J, Deininger KM, Aquilante CL. Pharmacodynamic Effects of Low-Dose Pioglitazone in Patients with the Metabolic Syndrome without Diabetes Mellitus. Pharmacotherapy 2016; 36:252-62. [DOI: 10.1002/phar.1713] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022]
Affiliation(s)
- Anh Vu
- Skaggs School of Pharmacy and Pharmaceutical Sciences; University of Colorado; Aurora Colorado
| | | | | | - Ronald Prigeon
- College of Medicine; University of Maryland; Baltimore Maryland
| | - Maha S. Sidhom
- Skaggs School of Pharmacy and Pharmaceutical Sciences; University of Colorado; Aurora Colorado
| | - Brooke Bredbeck
- Skaggs School of Pharmacy and Pharmaceutical Sciences; University of Colorado; Aurora Colorado
| | - Julie Predhomme
- Skaggs School of Pharmacy and Pharmaceutical Sciences; University of Colorado; Aurora Colorado
| | - Kimberly M. Deininger
- Skaggs School of Pharmacy and Pharmaceutical Sciences; University of Colorado; Aurora Colorado
| | - Christina L. Aquilante
- Skaggs School of Pharmacy and Pharmaceutical Sciences; University of Colorado; Aurora Colorado
| |
Collapse
|
49
|
Shankar SS, Shankar RR, Railkar RA, Beals CR, Steinberg HO, Kelley DE. Early Clinical Detection of Pharmacologic Response in Insulin Action in a Nondiabetic Insulin-Resistant Population. Curr Ther Res Clin Exp 2015; 77:83-9. [PMID: 26543510 PMCID: PMC4589823 DOI: 10.1016/j.curtheres.2015.08.001] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 08/07/2015] [Indexed: 11/16/2022] Open
Abstract
Background Insulin resistance heightens the risk for type 2 diabetes mellitus and cardiovascular disease. Amelioration of insulin resistance may reduce this risk. The thiazolidinedone class of insulin sensitizers improves insulin action in individuals with insulin-resistant diabetes and nondiabetic individuals. However, there are few reports on the time of onset of such effects independent of reversal of glucotoxicity. Objective The goal of our study was to test whether the thiazolidinedione pioglitazone has prominent early metabolic effects that can be detected in an obese, nondiabetic, insulin-resistant population. Methods We conducted a randomized, double-blind, placebo-controlled, parallel-group trial in men with nondiabetic insulin resistance using a hyperinsulinemic euglycemic clamp technique (at low and high doses of insulin at 10 and 40 mU/m2/min, respectively). The patients were given 30 mg daily oral pioglitazone or placebo for 28 days. Patients underwent a baseline clamp before initiation of treatment, and again at 14 and 28 days of treatment. Results Compared with placebo, under high-dose hyperinsulinemia, pioglitazone led to significant increases in glucose disposal rates (GDR) of 1.29 mg/kg/min (90% CI, 0.43–2.15; 39%; P=0.008) that were detectable at 2 weeks of treatment and persisted at 4 weeks of treatment. Under low-dose hyperinsulinemia, significant increases in GDR of 0.40 mg/kg/min (90% CI, 0.17–0.62; 95%; P=0.003) were observed at 4 weeks of treatment. These responses were accompanied by robust suppression of free fatty acids under hyperinsulinemic conditions, and by significant increases in circulating basal total adiponectin at 2 and 4 weeks of treatment. Conclusions Significant changes in insulin action across multiple insulin-sensitive tissues can be detected within 2 weeks of initiation of insulin-sensitizing therapy with pioglitazone in obese patients with nondiabetic insulin resistance. ClinicalTrials.gov identifier: NCT01115712.
Collapse
Affiliation(s)
- Sudha S Shankar
- Merck Research Laboratories, Merck & Co., Inc., Kenilworth, NJ
| | - R Ravi Shankar
- Merck Research Laboratories, Merck & Co., Inc., Kenilworth, NJ
| | - Radha A Railkar
- Merck Research Laboratories, Merck & Co., Inc., Kenilworth, NJ
| | - Chan R Beals
- Merck Research Laboratories, Merck & Co., Inc., Kenilworth, NJ
| | | | - David E Kelley
- Merck Research Laboratories, Merck & Co., Inc., Kenilworth, NJ
| |
Collapse
|
50
|
Daniele G, Winnier D, Mari A, Bruder J, Fourcaudot M, Pengou Z, Tripathy D, Jenkinson C, Folli F. Sclerostin and Insulin Resistance in Prediabetes: Evidence of a Cross Talk Between Bone and Glucose Metabolism. Diabetes Care 2015; 38:1509-17. [PMID: 26084344 DOI: 10.2337/dc14-2989] [Citation(s) in RCA: 90] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/16/2014] [Accepted: 04/28/2015] [Indexed: 02/03/2023]
Abstract
OBJECTIVE A gene mutation of the Wnt/β-catenin signaling cascade is present in rare patients with the insulin resistance syndrome. Sclerostin is a circulating peptide inhibiting Wnt/β-catenin signaling. Our aims were to evaluate serum sclerostin in subjects with prediabetes and to analyze its relationship with insulin resistance and β-cell function. RESEARCH DESIGN AND METHODS We performed a cross-sectional study including 43 healthy normal glucose-tolerant (NGT) individuals and 79 individuals with impaired glucose regulation (IGR), which included subjects with impaired fasting glucose (IFG), impaired glucose tolerance (IGT), and combined IFG-IGT, undergoing oral glucose tolerance test (OGTT) and dual-energy X-ray absorptiometry. A subgroup of 18 with NGT and 30 with IGR also underwent a euglycemic-hyperinsulinemic clamp with tracer. RESULTS Sclerostin levels were higher in IGR compared with NGT (50.8 ± 2.4 vs. 38.7 ± 2.3 pmol/L; P = 0.01), positively correlated with HOMA-insulin resistance (IR) (r = 0.62; P < 0.001), and negatively correlated with insulin-mediated total body glucose disposal (r = -0.40; P < 0.001). Fasting endogenous glucose production (EGP) and hepatic and adipose tissue insulin resistance indexes were positively correlated with sclerostin levels (r = 0.48, r = 0.62, and r = 0.61, respectively; P < 0.001). Fasting and OGTT insulin clearance were inversely correlated with sclerostin serum levels (r = -0.52 and r = -0.44, respectively; both P < 0.001). Sclerostin levels were not correlated with β-cell function parameters. In multiple linear regression analysis, the addition of sclerostin levels to the traditional risk factors for insulin resistance improved the r(2) associated with HOMA-IR (r(2) change: 0.055; F change: 28.893; P = 0.001) and insulin-mediated total body glucose disposal (r(2) change: 0.059; F change: 4.938; P = 0.033). CONCLUSIONS Sclerostin levels are increased in individuals with prediabetes and correlated with insulin resistance in skeletal muscle, liver, and adipose tissue. The correlation between sclerostin and insulin clearance at fasting state and during OGTT is novel; thus, studies are needed to explore the potential causal relationship.
Collapse
Affiliation(s)
- Giuseppe Daniele
- Department of Medicine, Diabetes Division, University of Texas Health Science Center at San Antonio, San Antonio, TX
| | - Deidre Winnier
- Department of Medicine, Diabetes Division, University of Texas Health Science Center at San Antonio, San Antonio, TX
| | - Andrea Mari
- Institute of Neuroscience, National Research Council, Padua, Italy
| | - Jan Bruder
- Department of Medicine, University of Texas Health Science Center at San Antonio, San Antonio, TX
| | - Marcel Fourcaudot
- Department of Medicine, Diabetes Division, University of Texas Health Science Center at San Antonio, San Antonio, TX
| | - Zuo Pengou
- Department of Medicine, Diabetes Division, University of Texas Health Science Center at San Antonio, San Antonio, TX
| | - Devjit Tripathy
- Department of Medicine, Diabetes Division, University of Texas Health Science Center at San Antonio, San Antonio, TX
| | - Christopher Jenkinson
- Department of Medicine, Diabetes Division, University of Texas Health Science Center at San Antonio, San Antonio, TX
| | - Franco Folli
- Department of Medicine, Diabetes Division, University of Texas Health Science Center at San Antonio, San Antonio, TX Departamento de Clinica Medica, Faculdade de Ciencias Medicas, Obesity and Comorbidities Research Center, Universidade Estadual de Campinas, Campinas, Brazil
| |
Collapse
|