1
|
Frangie Machado M, Shunk T, Hansen G, Harvey C, Fulford B, Hauf S, Schuh O, Kaldas M, Arcaroli E, Ortiz J, De Gaetano J. Clinical Effects of Glucagon-Like Peptide-1 Agonist Use for Weight Loss in Women With Polycystic Ovary Syndrome: A Scoping Review. Cureus 2024; 16:e66691. [PMID: 39262529 PMCID: PMC11389649 DOI: 10.7759/cureus.66691] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2024] [Accepted: 08/12/2024] [Indexed: 09/13/2024] Open
Abstract
Glucagon-like peptide-1 (GLP-1) is a gastrointestinal regulatory hormone that stimulates insulin release from the pancreas. While GLP-1 receptor agonists (GLP-1 RAs) have traditionally been utilized to address insulin resistance, their potential application in treating polycystic ovary syndrome (PCOS) has recently garnered attention. This study aimed to investigate the therapeutic efficacy of GLP-1 RAs use for weight loss in women diagnosed with PCOS. We conducted a scoping review following the Joanna Briggs Institute (JBI) methodology and adhering to the Preferred Reporting Items for Systematic Reviews and Meta-Analyses (PRISMA) guidelines. Our investigation delved into the clinical effects experienced by women of diverse racial and ethnic backgrounds with PCOS who were prescribed GLP-1 RAs for weight loss. Peer-reviewed articles from Ovid Medline, Web of Science, CINAHL, Cochrane CENTRAL, SCOPUS, and ClinicalTrials.gov spanning from 2012 to 2023 were scrutinized. After eliminating duplicates, 811 articles were identified, and ultimately, eight met the eligibility criteria for inclusion. All studies were published in English and exhibited wide geographic diversity. The included studies uniformly reported reductions in weight and body mass index (BMI) among patients who were prescribed GLP-1 RAs, specifically liraglutide or exenatide. Additionally, evidence pointed towards improvements in anthropometric parameters (MF1) (including total body weight, BMI, reduction in waist circumference, and total fat percentage), glucose homeostasis, cardiovascular inflammatory markers (midregional pro-atrial natriuretic peptide (MR-proANP) and mid-regional pro-adrenomedullin (MR-proADM)), rates of pregnancy, and menstrual regulation. However, findings regarding the impact of GLP-1 RAs on lipid profiles were inconsistent. Although some short-term adverse effects were noted, long-term effects of GLP-1 RAs use remain undetermined. GLP-1 RA use demonstrated promising clinical outcomes for women with PCOS, including reduced BMI, improved metabolic parameters, menstrual regularity, and increased rates of natural pregnancy. While the current evidence is encouraging, further research is warranted to elucidate both short- and long-term adverse effects of GLP-1 RA therapy for PCOS.
Collapse
Affiliation(s)
- Melissa Frangie Machado
- Family Medicine, Nova Southeastern University Dr. Kiran C. Patel College of Osteopathic Medicine, Tampa, USA
| | - Taylor Shunk
- Family Medicine, Nova Southeastern University Dr. Kiran C. Patel College of Osteopathic Medicine, Tampa, USA
| | - Grace Hansen
- Family Medicine, Nova Southeastern University Dr. Kiran C. Patel College of Osteopathic Medicine, Tampa, USA
| | - Charles Harvey
- Family Medicine, Nova Southeastern University Dr. Kiran C. Patel College of Osteopathic Medicine, Tampa, USA
| | - Baylee Fulford
- Family Medicine, Nova Southeastern University Dr. Kiran C. Patel College of Osteopathic Medicine, Tampa, USA
| | - Shane Hauf
- Family Medicine, Nova Southeastern University Dr. Kiran C. Patel College of Osteopathic Medicine, Tampa, USA
| | - Olivia Schuh
- Family Medicine, Nova Southeastern University Dr. Kiran C. Patel College of Osteopathic Medicine, Tampa, USA
| | - Matthew Kaldas
- Family Medicine, Nova Southeastern University Dr. Kiran C. Patel College of Osteopathic Medicine, Tampa, USA
| | - Elena Arcaroli
- Family Medicine, Nova Southeastern University Dr. Kiran C. Patel College of Osteopathic Medicine, Tampa, USA
| | - Justin Ortiz
- Family Medicine, Nova Southeastern University Dr. Kiran C. Patel College of Osteopathic Medicine, Tampa, USA
| | | |
Collapse
|
2
|
Lo CH, O’Connor LM, Loi GWZ, Saipuljumri EN, Indajang J, Lopes KM, Shirihai OS, Grinstaff MW, Zeng J. Acidic Nanoparticles Restore Lysosomal Acidification and Rescue Metabolic Dysfunction in Pancreatic β-Cells under Lipotoxic Conditions. ACS NANO 2024; 18:15452-15467. [PMID: 38830624 PMCID: PMC11192035 DOI: 10.1021/acsnano.3c09206] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/23/2023] [Revised: 05/09/2024] [Accepted: 05/16/2024] [Indexed: 06/05/2024]
Abstract
Type 2 diabetes (T2D), a prevalent metabolic disorder lacking effective treatments, is associated with lysosomal acidification dysfunction, as well as autophagic and mitochondrial impairments. Here, we report a series of biodegradable poly(butylene tetrafluorosuccinate-co-succinate) polyesters, comprising a 1,4-butanediol linker and varying ratios of tetrafluorosuccinic acid (TFSA) and succinic acid as components, to engineer lysosome-acidifying nanoparticles (NPs). The synthesized NPs are spherical with diameters of ≈100 nm and have low polydispersity and good stability. Notably, TFSA NPs, which are composed entirely of TFSA, exhibit the strongest degradation capability and superior acidifying properties. We further reveal significant downregulation of lysosomal vacuolar (H+)-ATPase subunits, which are responsible for maintaining lysosomal acidification, in human T2D pancreatic islets, INS-1 β-cells under chronic lipotoxic conditions, and pancreatic tissues of high-fat-diet (HFD) mice. Treatment with TFSA NPs restores lysosomal acidification, autophagic function, and mitochondrial activity, thereby improving the pancreatic function in INS-1 cells and HFD mice with lipid overload. Importantly, the administration of TFSA NPs to HFD mice reduces insulin resistance and improves glucose clearance. These findings highlight the therapeutic potential of lysosome-acidifying TFSA NPs for T2D.
Collapse
Affiliation(s)
- Chih Hung Lo
- Lee
Kong Chian School of Medicine, Nanyang Technological University, Singapore 308232, Singapore
| | - Lance M. O’Connor
- College
of Biological Sciences, University of Minnesota, Minneapolis, Minnesota 55455, United States
| | - Gavin Wen Zhao Loi
- Lee
Kong Chian School of Medicine, Nanyang Technological University, Singapore 308232, Singapore
| | | | - Jonathan Indajang
- Meinig
School of Biomedical Engineering, Cornell
University, Ithaca, New York 14853, United States
| | - Kaitlynn M. Lopes
- Department
of Chemistry, Boston University, Boston, Massachusetts 02215, United States
| | - Orian S. Shirihai
- Division
of Endocrinology, Department of Medicine, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, California 90045, United States
- Department
of Molecular and Medical Pharmacology, University
of California, Los Angeles, Los
Angeles, California 90095, United States
| | - Mark W. Grinstaff
- Department
of Chemistry, Boston University, Boston, Massachusetts 02215, United States
- Department
of Biomedical Engineering, Boston University, Boston, Massachusetts 02215, United States
| | - Jialiu Zeng
- Lee
Kong Chian School of Medicine, Nanyang Technological University, Singapore 308232, Singapore
| |
Collapse
|
3
|
Kovács P, Kitka T, Bali ZK, Nagy LV, Bodó A, Kovács-Öller T, Péterfi Z, Hernádi I. Chemogenetic inhibition of the lateral hypothalamus effectively reduces food intake in rats in a translational proof-of-concept study. Sci Rep 2024; 14:11402. [PMID: 38762561 PMCID: PMC11102470 DOI: 10.1038/s41598-024-62014-1] [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: 02/12/2024] [Accepted: 05/13/2024] [Indexed: 05/20/2024] Open
Abstract
Despite the therapeutic potential of chemogenetics, the method lacks comprehensive preclinical validation, hindering its progression to human clinical trials. We aimed to validate a robust but simple in vivo efficacy assay in rats which could support chemogenetic drug discovery by providing a quick, simple and reliable animal model. Key methodological parameters such as adeno-associated virus (AAV) serotype, actuator drug, dose, and application routes were investigated by measuring the food-intake-reducing effect of chemogenetic inhibition of the lateral hypothalamus (LH) by hM4D(Gi) designer receptor stimulation. Subcutaneous deschloroclozapine in rats transfected with AAV9 resulted in a substantial reduction of food-intake, comparable to the efficacy of exenatide. We estimated that the effect of deschloroclozapine lasts 1-3 h post-administration. AAV5, oral administration of deschloroclozapine, and clozapine-N-oxide were also effective but with slightly less potency. The strongest effect on food-intake occurred within the first 30 min after re-feeding, suggesting this as the optimal experimental endpoint. This study demonstrates that general chemogenetic silencing of the LH can be utilized as an optimal, fast and reliable in vivo experimental model for conducting preclinical proof-of-concept studies in order to validate the in vivo effectiveness of novel chemogenetic treatments. We also hypothesize based on our results that universal LH silencing with existing and human translatable genetic neuroengineering techniques might be a viable strategy to affect food intake and influence obesity.
Collapse
Affiliation(s)
- Péter Kovács
- VRG Therapeutics, Füvészkert utca 3., Budapest, 1083, Hungary
| | - Tamás Kitka
- VRG Therapeutics, Füvészkert utca 3., Budapest, 1083, Hungary
| | - Zsolt Kristóf Bali
- Grastyán Endre Translational Research Centre, University of Pécs, 6 Ifjúság str., Pécs, 7624, Hungary.
- Translational Neuroscience Research Group, Centre for Neuroscience, Szentágothai Research Centre, University of Pécs, 20 Ifjúság str., Pécs, 7624, Hungary.
| | - Lili Veronika Nagy
- Grastyán Endre Translational Research Centre, University of Pécs, 6 Ifjúság str., Pécs, 7624, Hungary
- Translational Neuroscience Research Group, Centre for Neuroscience, Szentágothai Research Centre, University of Pécs, 20 Ifjúság str., Pécs, 7624, Hungary
- Department of Neurobiology, Faculty of Sciences, University of Pécs, 6 Ifjúság str., Pécs, 7624, Hungary
| | - Angelika Bodó
- Grastyán Endre Translational Research Centre, University of Pécs, 6 Ifjúság str., Pécs, 7624, Hungary
- Translational Neuroscience Research Group, Centre for Neuroscience, Szentágothai Research Centre, University of Pécs, 20 Ifjúság str., Pécs, 7624, Hungary
| | - Tamás Kovács-Öller
- Department of Neurobiology, Faculty of Sciences, University of Pécs, 6 Ifjúság str., Pécs, 7624, Hungary
- Histology and Light Microscopy Core Facility, Szentágothai Research Centre, University of Pécs, 20 Ifjúság str., Pécs, 7624, Hungary
| | - Zalán Péterfi
- VRG Therapeutics, Füvészkert utca 3., Budapest, 1083, Hungary
| | - István Hernádi
- Grastyán Endre Translational Research Centre, University of Pécs, 6 Ifjúság str., Pécs, 7624, Hungary
- Translational Neuroscience Research Group, Centre for Neuroscience, Szentágothai Research Centre, University of Pécs, 20 Ifjúság str., Pécs, 7624, Hungary
- Department of Neurobiology, Faculty of Sciences, University of Pécs, 6 Ifjúság str., Pécs, 7624, Hungary
- Institute of Physiology, Medical School, University of Pécs, 12 Szigeti út, Pécs, 7624, Hungary
| |
Collapse
|
4
|
Cristelo C, Nunes R, Pinto S, Marques JM, Gama FM, Sarmento B. Targeting β Cells with Cathelicidin Nanomedicines Improves Insulin Function and Pancreas Regeneration in Type 1 Diabetic Rats. ACS Pharmacol Transl Sci 2023; 6:1544-1560. [PMID: 37854630 PMCID: PMC10580391 DOI: 10.1021/acsptsci.3c00218] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2023] [Indexed: 10/20/2023]
Abstract
Type 1 diabetes (T1D) is an incurable condition with an increasing incidence worldwide, in which the hallmark is the autoimmune destruction of pancreatic insulin-producing β cells. Cathelicidin-based peptides have been shown to improve β cell function and neogenesis and may thus be relevant while developing T1D therapeutics. In this work, a cathelicidin-derived peptide, LLKKK18, was loaded in poly(lactic-co-glycolic acid) (PLGA) nanoparticles (NPs), surface-functionalized with exenatide toward a GLP-1 receptor, aiming the β cell-targeted delivery of the peptide. The NPs present a mean size of around 100 nm and showed long-term stability, narrow size distribution, and negative ζ-potential (-10 mV). The LLKKK18 association efficiency and loading were 62 and 2.9%, respectively, presenting slow and sustained in vitro release under simulated physiologic fluids. Glucose-stimulated insulin release in the INS-1E cell line was observed in the presence of the peptide. In addition, NPs showed a strong association with β cells from isolated rat islets. After administration to diabetic rats, NPs induced a significant reduction of the hyperglycemic state, an improvement in the pancreatic insulin content, and glucose tolerance. Also remarkable, a considerable increase in the β cell mass in the pancreas was observed. Overall, this novel and versatile nanomedicine showed glucoregulatory ability and can pave the way for the development of a new generation of therapeutic approaches for T1D treatment.
Collapse
Affiliation(s)
- Cecília Cristelo
- i3S
− Instituto de Investigação e Inovação
em Saúde, Universidade do Porto, Porto 4200-135, Portugal
- Centro
de Engenharia Biológica, Universidade
do Minho, Campus de Gualtar, Braga 4710-057, Portugal
- ICBAS
− Instituto de Ciências Biomédicas Abel Salazar, Universidade do Porto, Porto 4050-313, Portugal
| | - Rute Nunes
- i3S
− Instituto de Investigação e Inovação
em Saúde, Universidade do Porto, Porto 4200-135, Portugal
- IUCS-CESPU, Instituto
Universitário de Ciências
da Saúde, Gandra 4585-116, Portugal
| | - Soraia Pinto
- i3S
− Instituto de Investigação e Inovação
em Saúde, Universidade do Porto, Porto 4200-135, Portugal
- ICBAS
− Instituto de Ciências Biomédicas Abel Salazar, Universidade do Porto, Porto 4050-313, Portugal
| | - Joana Moreira Marques
- i3S
− Instituto de Investigação e Inovação
em Saúde, Universidade do Porto, Porto 4200-135, Portugal
- Faculdade
de Farmácia, Universidade do Porto, Porto 4099-002, Portugal
| | - Francisco Miguel Gama
- Centro
de Engenharia Biológica, Universidade
do Minho, Campus de Gualtar, Braga 4710-057, Portugal
| | - Bruno Sarmento
- i3S
− Instituto de Investigação e Inovação
em Saúde, Universidade do Porto, Porto 4200-135, Portugal
- IUCS-CESPU, Instituto
Universitário de Ciências
da Saúde, Gandra 4585-116, Portugal
| |
Collapse
|
5
|
Xie Z, Enkhjargal B, Nathanael M, Wu L, Zhu Q, Zhang T, Tang J, Zhang JH. Exendin-4 Preserves Blood-Brain Barrier Integrity via Glucagon-Like Peptide 1 Receptor/Activated Protein Kinase-Dependent Nuclear Factor-Kappa B/Matrix Metalloproteinase-9 Inhibition After Subarachnoid Hemorrhage in Rat. Front Mol Neurosci 2022; 14:750726. [PMID: 35002615 PMCID: PMC8733623 DOI: 10.3389/fnmol.2021.750726] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2021] [Accepted: 11/26/2021] [Indexed: 11/15/2022] Open
Abstract
In this study, we investigated the role of Exendin-4 (Ex-4), a glucagon-like peptide 1 receptor (GLP-1R) agonist, in blood-brain barrier (BBB) disruption after subarachnoid hemorrhage (SAH) in rats. The endovascular perforation model of SAH was performed in Sprague-Dawley rats. Ex-4 was intraperitoneally injected 1 h after SAH induction. To elucidate the underlying molecular mechanism, small interfering ribonucleic acid (siRNA) for GLP-1R and Dorsomorphin, a specific inhibitor of adenosine monophosphate-activated protein kinase (AMPK), were intracerebroventricularly injected 48 h before induction of SAH correspondingly. Immunofluorescence results supported GLP-1R expressed on the endothelial cells of microvessels in the brain after SAH. Administration of Ex-4 significantly reduced brain water content and Evans blue extravasation in both hemispheres, which improved neurological scores at 24 h after SAH. In the mechanism study, Ex-4 treatment significantly increased the expression of GLP-1R, p-AMPK, IκB-α, Occludin, and Claudin-5, while the expression of p-nuclear factor-kappa B (NF-κB) p65, matrix metalloproteinase-9 (MMP-9), and albumin was significantly decreased. The effects of Ex-4 were reversed by the intervention of GLP-1R siRNA or Dorsomorphin, respectively. In conclusion, Ex-4 could preserve the BBB integrity through GLP-1R/AMPK-dependent NF-κB/MMP-9 inhibition after SAH, which should be further investigated as a potential therapeutic target in SAH.
Collapse
Affiliation(s)
- Zhiyi Xie
- Department of Neurosurgery, The Second Affiliated Hospital, Zhejiang University, Hangzhou, China.,Department of Physiology and Pharmacology, Loma Linda University, Loma Linda, CA, United States
| | - Budbazar Enkhjargal
- Department of Physiology and Pharmacology, Loma Linda University, Loma Linda, CA, United States
| | - Matei Nathanael
- Department of Physiology and Pharmacology, Loma Linda University, Loma Linda, CA, United States
| | - Lingyun Wu
- Department of Physiology and Pharmacology, Loma Linda University, Loma Linda, CA, United States
| | - Qiquan Zhu
- Department of Physiology and Pharmacology, Loma Linda University, Loma Linda, CA, United States
| | - Tongyu Zhang
- Department of Physiology and Pharmacology, Loma Linda University, Loma Linda, CA, United States
| | - Jiping Tang
- Department of Physiology and Pharmacology, Loma Linda University, Loma Linda, CA, United States
| | - John H Zhang
- Department of Physiology and Pharmacology, Loma Linda University, Loma Linda, CA, United States
| |
Collapse
|
6
|
Bhatt M, Shende P. Modulated approaches for strategic transportation of proteins and peptides via ocular route. J Drug Deliv Sci Technol 2021. [DOI: 10.1016/j.jddst.2021.102835] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
|
7
|
Wang L, Liu X, Yang W, Lai J, Yu X, Liu J, Gao X, Ming J, Ma K, Xu J, Tian Z, He Q, Ji Q. Comparison of Blood Glucose Variability Between Exenatide and Biphasic Insulin Aspart 30 in Chinese Participants with Type 2 Diabetes Inadequately Controlled with Metformin Monotherapy: A Multicenter, Open-Label, Randomized Trial. Diabetes Ther 2020; 11:2313-2328. [PMID: 32856226 PMCID: PMC7509011 DOI: 10.1007/s13300-020-00904-z] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/02/2020] [Indexed: 12/21/2022] Open
Abstract
INTRODUCTION To compare blood glucose variability (GV) in Chinese participants with type 2 diabetes mellitus (T2DM) whose blood glucose levels are inadequately controlled with metformin monotherapy after twice-daily exenatide or biphasic insulin aspart 30 (BIAsp30). METHODS In this 16-week multicenter, randomized clinical trial, 104 participants were randomized 1:1 to receive exenatide (exenatide group) or BIAsp30 (BIAsp30 group) twice daily. All participants continued metformin treatment. The primary outcome was the change in GV as measured by a continuous glucose monitoring system (CGMS) from baseline to 16 weeks. RESULTS At 16 weeks, both the Exenatide and BIAsp30 groups effectively decreased mean glucose (MG), but neither group changed the mean amplitude of glycemic excursion (MAGE), largest amplitude of glycemic excursion (LAGE), mean of daily difference (MODD), or standard deviation of blood glucose (SDBG). The decrease in 2-h post-breakfast glucose excursions was greater in the Exenatide group compared to the BIAsp30 group, with a least square (LS) mean difference [95% CI] of (1.58 [0.53, 2.63]). Exenatide also significantly reduced 2-h post-lunch glucose excursion compared to BIAsp30 (LS mean difference [95% CI], 1.19 [0.18, 2.20]). The Exenatide group had significantly reduced body weight and body mass index (BMI), while the BIAsp30 group had increased weight and had no change in BMI. Both treatments were well tolerated with no serious hypoglycemic events and with fewer identified hypoglycemic events in the Exenatide group than in the BIAsp30 group (5.77% vs. 17.31%, P < 0.01). CONCLUSION Although there was no difference in change of GV between Exenatide and BIAsp30, exenatide provided more improvement in postprandial glucose excursion and weight control, without increasing the risk of hypoglycemia in Chinese patients with T2DM whose blood glucose was inadequately controlled with metformin. These findings may provide new options for patients who choose further hypoglycemic treatment, especially in patients with obesity who have large postprandial plasma glucose excursions. TRIAL REGISTRATION ClinicalTrials.gov indentifier: NCT02449603.
Collapse
Affiliation(s)
- Li Wang
- Department of Endocrinology, Xijing Hospital, Fourth Military Medical University, Xi'an, China
| | - Xiangyang Liu
- Department of Endocrinology, Xijing Hospital, Fourth Military Medical University, Xi'an, China
| | - Wenjuan Yang
- Department of Endocrinology, Shaanxi Aerospace Hospital, Xi'an, Shaanxi, China
| | - Jingbo Lai
- Department of Endocrinology, Xijing Hospital, Fourth Military Medical University, Xi'an, China
| | - Xinwen Yu
- Department of Endocrinology, Xijing Hospital, Fourth Military Medical University, Xi'an, China
| | - Jianrong Liu
- Department of Endocrinology, Xi'an Chang an Hospital, Xi'an, Shaanxi, China
| | - Xiling Gao
- Department of Endocrinology, Yan'an People's Hospital, Yan'an, Shaanxi, China
| | - Jie Ming
- Department of Endocrinology, Xijing Hospital, Fourth Military Medical University, Xi'an, China
| | - Kaiyan Ma
- Department of Endocrinology, Shangluo Central Hospital, Shangluo, Shaanxi, China
| | - Jing Xu
- Department of Endocrinology, Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi, China
| | - Zhufang Tian
- Department of Endocrinology, Xi'an Central Hospital, Xi'an, Shaanxi, China
| | - Qingzhen He
- Department of Endocrinology, Xi'an Gaoxin Hospital, Xi'an, Shaanxi, China
| | - Qiuhe Ji
- Department of Endocrinology, Xijing Hospital, Fourth Military Medical University, Xi'an, China.
| |
Collapse
|
8
|
Neo CWY, Ciaramicoli LM, Soetedjo AAP, Teo AKK, Kang NY. A new perspective of probe development for imaging pancreatic beta cell in vivo. Semin Cell Dev Biol 2020; 103:3-13. [PMID: 32057664 DOI: 10.1016/j.semcdb.2020.01.009] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2019] [Revised: 01/20/2020] [Accepted: 01/28/2020] [Indexed: 12/23/2022]
Abstract
Beta cells assume a fundamental role in maintaining blood glucose homeostasis through the secretion of insulin, which is contingent on both beta cell mass and function, in response to elevated blood glucose levels or secretagogues. For this reason, evaluating beta cell mass and function, as well as scrutinizing how they change over time in a diabetic state, are essential prerequisites in elucidating diabetes pathophysiology. Current clinical methods to measure human beta cell mass and/or function are largely lacking, indirect and sub-optimal, highlighting the continued need for noninvasive in vivo beta cell imaging technologies such as optical imaging techniques. While numerous probes have been developed and evaluated for their specificity to beta cells, most of them are more suited to visualize beta cell mass rather than function. In this review, we highlight the distinction between beta cell mass and function, and the importance of developing more probes to measure beta cell function. Additionally, we also explore various existing probes that can be employed to measure beta cell mass and function in vivo, as well as the caveats in probe development for in vivo beta cell imaging.
Collapse
Affiliation(s)
- Claire Wen Ying Neo
- Stem Cells and Diabetes Laboratory, Institute of Molecular and Cell Biology (IMCB), A*STAR, Singapore, 138673, Singapore; Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 117596, Singapore
| | - Larissa Miasiro Ciaramicoli
- Department of Advanced Materials Science, Pohang University of Science and Technology (POSTECH), Pohang, 37673, Republic of Korea
| | - Andreas Alvin Purnomo Soetedjo
- Stem Cells and Diabetes Laboratory, Institute of Molecular and Cell Biology (IMCB), A*STAR, Singapore, 138673, Singapore
| | - Adrian Kee Keong Teo
- Stem Cells and Diabetes Laboratory, Institute of Molecular and Cell Biology (IMCB), A*STAR, Singapore, 138673, Singapore; Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 117596, Singapore.
| | - Nam-Young Kang
- Department of Creative IT Engineering, Pohang University of Science and Technology (POSTECH), 77 Cheongam-ro, Namgu, C5 Building, Room 203, Pohang, Kyungbuk, 37673, Republic of Korea.
| |
Collapse
|
9
|
Liu Y, Wang Q, Nie D, Zhang Y, Wang Z, Zhang Y. Novel Modified GLP-1 Derivatives with Prolonged Glucose-Lowering Ability In Vivo. Int J Pept Res Ther 2019. [DOI: 10.1007/s10989-019-09991-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
|
10
|
Preparation of [ 177Lu]Lu-DOTA-Ahx-Lys40-Exendin-4 for radiotherapy of insulinoma: a detailed insight into the radiochemical intricacies. Nucl Med Biol 2019; 78-79:31-40. [PMID: 31731177 DOI: 10.1016/j.nucmedbio.2019.11.003] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2019] [Revised: 10/11/2019] [Accepted: 11/07/2019] [Indexed: 01/05/2023]
Abstract
INTRODUCTION [177Lu]Lu-DOTA-Ahx-Lys40-Exendin-4 ([177Lu]Lu-DOTA-Exendin-4) is a potential agent for radiotherapy of insulinomas owing to its specificity towards GLP-1 (Glucagon like peptide-1) receptors over-expressed on such cancers. The objective of the present study is to optimize the various radiochemistry parameters for the consistent formulation of the agent with high radiolabeling yield using carrier added [177Lu]LuCl3 and also to evaluate its biological behaviour in small animal model. METHODS In order to optimize the radiolabeling parameters, DOTA-Exendin-4 was radiolabeled with [177Lu]LuCl3 in two different buffer systems (sodium acetate and HEPES) at three different temperatures (45, 65 and 95 °C) using three different ligand to metal ratios (3:1, 4:1 and 5:1). The radiolabeled peptide was characterized by both paper chromatography and HPLC. The effect of addition of three different radio-protectors on complexation yield was also studied. Bio-distribution studies were carried out in healthy Swiss mice to evaluate the pharmacokinetic behaviour of the radiolabeled peptide as well as to determine the in vivo specificity of the radiotracer towards GLP-1 receptors (blocking studies). Urine and kidney lysate of the animals were analyzed at various post-administration time-points in order to determine the in vivo stability of the radiolabeled peptide. RESULTS The [177Lu]Lu-DOTA-Exendin-4 complex could be prepared consistently with >95% radiolabeling yield using the optimized reaction conditions. Bio-distribution studies revealed early accumulation of [177Lu]Lu-DOTA-Exendin-4 in pancreas along with fast clearance via renal pathway. Significantly high accumulation of the radiotracer was observed in kidneys. Analyses of urine and kidney lysate of the animals revealed in vivo stability of [177Lu]Lu-DOTA-Exendin-4. Blocking studies showed displacement of significant amount of radiotracer from GLP-1 receptor-positive organs such as, pancreas and lungs (p <0.05) in presence of unlabeled peptide, indicating the specificity of the radiolabeled preparation towards GLP-1 receptors. CONCLUSIONS Present study shows that [177Lu]Lu-DOTA-Exendin-4 could be formulated for radiotherapeutic application with high radiochemical purity and adequate in vivo stability using [177Lu]LuCl3 produced via direct neutron irradiation. ADVANCES IN KNOWLEDGE AND IMPLICATIONS FOR PATIENT CARE Findings of the present study will be helpful in preparing the patient dose of [177Lu]Lu-labeled Exendin for radiotherapy of insulinoma using carrier added [177Lu]LuCl3, produced in a medium flux reactor, without the requirement of post-labeling purification.
Collapse
|
11
|
Wang J, Huang J, Li W, Tang S, Sun J, Zhang X, Liu J, Yi B, Liu J, Zhang X, Yang Q, Yang X, Yang S, Yang G, Zhang H. Polyethylene glycol loxenatide (PEX168) in subjects with renal impairment: A pharmacokinetic study. Br J Clin Pharmacol 2019; 85:2714-2720. [PMID: 31396983 PMCID: PMC6955414 DOI: 10.1111/bcp.14091] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2019] [Revised: 05/13/2019] [Accepted: 07/26/2019] [Indexed: 12/29/2022] Open
Abstract
Aims Type 2 diabetes mellitus (T2DM) is commonly complicated by renal impairment. Polyethylene glycol loxenatide (PEX168) is a novel long‐acting glucagon‐like peptide‐1 receptor agonist for T2DM. PEX168 pharmacokinetics was studied to identify requirements for dose‐modification in T2DM complicated by renal impairment. Methods This was a single‐centre, open‐labelled, parallel‐group, single‐dose, phase I clinical trial of patients with mild and moderate renal impairment, and with or without T2DM. Age‐, sex‐ and body mass index‐matched subjects with normal renal function, and with or without T2DM were recruited as controls. Subjects received a single abdominal subcutaneous injection of PEX168 200 μg. Pharmacokinetic samples were taken at 0, 24, 48, 72, 96, 120, 144, 216, 312, 480, 648 and 720 hours. Results Twenty‐three patients were included in the pharmacokinetics analysis. Vz/F and CL/F were lower in the moderate impairment group than in the other groups. The mean t1/2 (163 hours) in the moderate impairment group was prolonged compared to the mild impairment (117 hours) and normal (121 hours) groups. AUC0–inf increased by 13 and 100.7% in patients with mild and moderate renal impairment, respectively. Most adverse events were mild gastrointestinal disorders, with only 1 serious adverse event observed. Conclusion A single dose of 200 μg of PEX168 was in general well tolerated in patients with renal impairment. The in vivo clearance rate of PEX168 in patients with moderate renal impairment is slower than in patients with mild renal impairment and normal renal function and dose adjustment might be required (http://ClinicalTrials.org #NCT02467790).
Collapse
Affiliation(s)
- Jianwen Wang
- Department of Nephrology, The Third Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Jie Huang
- Center of Clinical Pharmacology, The Third Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Wei Li
- Department of Nephrology, The Third Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Shiqi Tang
- Department of Nephrology, The Third Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Jian Sun
- Department of Nephrology, The Third Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Xianming Zhang
- Department of Nephrology, The Third Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Jun Liu
- Department of Nephrology, The Third Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Bin Yi
- Department of Nephrology, The Third Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Jishi Liu
- Department of Nephrology, The Third Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Xingfei Zhang
- Center of Clinical Pharmacology, The Third Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Qian Yang
- Center of Clinical Pharmacology, The Third Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Xiaoyan Yang
- Center of Clinical Pharmacology, The Third Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Shuang Yang
- Center of Clinical Pharmacology, The Third Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Guoping Yang
- Center of Clinical Pharmacology, The Third Xiangya Hospital, Central South University, Changsha, Hunan, China.,Research Center of Drug Clinical Evaluation of Central South University, Changsha, Hunan, China.,Xiangya School of Pharmaceutical Sciences, Central South University, Changsha, Hunan, China
| | - Hao Zhang
- Department of Nephrology, The Third Xiangya Hospital, Central South University, Changsha, Hunan, China
| |
Collapse
|
12
|
Exenatide Reduces Graft Injury in a Rat Transplantation Model Using Kidneys Donated after Cardiac Death. Transplant Proc 2019; 51:2116-2123. [PMID: 31303407 DOI: 10.1016/j.transproceed.2019.04.034] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2018] [Revised: 04/04/2019] [Accepted: 04/22/2019] [Indexed: 11/21/2022]
Abstract
Besides being used in the therapy of type 2 diabetes, exenatide reduces cerebral ischemia-reperfusion (I/R) injury. We evaluated the potential effects of exenatide on inhibition of apoptosis in kidney grafts donated after cardiac death and on reduction of I/R injury after kidney transplantation (KTx) in a rat model. We used a rat syngeneic KTx model with kidney grafts obtained after cardiac death, and apoptosis was detected in the graft before KTx. Graft function, rat survival, morphologic examination, and activation of inflammatory molecules were analyzed after KTx. By the end of the cold storage, exenatide pretreatment donors had significantly reduced caspase pathway activation, terminal deoxynucleotidyl transferase dUTP nick-end labeling--positive cells, release of mitochondrial porin proteins into the cytosol, and expression of cleaved caspase-3 and poly (ADP-ribose) polymerase in kidney grafts. Exenatide pretreatment improved renal function survival rate with lower scores of acute tubular necrosis, infiltrating macrophages, and interstitial fibrosis as well as reduced messenger RNA expression of inflammatory mediators (tumor necrosis factor α, interleukin-6, interleukin-1β, and intercellular adhesion molecule-1) after KTx. Our study showed that exenatide reduced I/R injury in kidneys donated after cardiac death in a rat transplantation model and improved recipient survival and graft function.
Collapse
|
13
|
Seth PP, Tanowitz M, Bennett CF. Selective tissue targeting of synthetic nucleic acid drugs. J Clin Invest 2019; 129:915-925. [PMID: 30688661 DOI: 10.1172/jci125228] [Citation(s) in RCA: 70] [Impact Index Per Article: 11.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023] Open
Abstract
Antisense oligonucleotides (ASOs) are chemically synthesized nucleic acid analogs designed to bind to RNA by Watson-Crick base pairing. Following binding to the targeted RNA, the ASO perturbs RNA function by promoting selective degradation of the targeted RNA, altering RNA intermediary metabolism, or disrupting function of the RNA. Most antisense drugs are chemically modified to enhance their pharmacological properties and for passive targeting of the tissues of therapeutic interest. Recent advances in selective tissue targeting have resulted in a newer generation of ASO drugs that are more potent and better tolerated than previous generations, spawning renewed interest in identifying selective ligands that enhance targeted delivery of ASOs to tissues.
Collapse
|
14
|
Wei W, Ehlerding EB, Lan X, Luo QY, Cai W. Molecular imaging of β-cells: diabetes and beyond. Adv Drug Deliv Rev 2019; 139:16-31. [PMID: 31378283 DOI: 10.1016/j.addr.2018.06.022] [Citation(s) in RCA: 37] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2017] [Revised: 04/27/2018] [Accepted: 06/26/2018] [Indexed: 02/09/2023]
Abstract
Since diabetes is becoming a global epidemic, there is a great need to develop early β-cell specific diagnostic techniques for this disorder. There are two types of diabetes (i.e., type 1 diabetes mellitus (T1DM) and type 2 diabetes mellitus (T2DM)). In T1DM, the destruction of pancreatic β-cells leads to reduced insulin production or even absolute insulin deficiency, which consequently results in hyperglycemia. Actually, a central issue in the pathophysiology of all types of diabetes is the relative reduction of β-cell mass (BCM) and/or impairment of the function of individual β-cells. In the past two decades, scientists have been trying to develop imaging techniques for noninvasive measurement of the viability and mass of pancreatic β-cells. Despite intense scientific efforts, only two tracers for positron emission tomography (PET) and one contrast agent for magnetic resonance (MR) imaging are currently under clinical evaluation. β-cell specific imaging probes may also allow us to precisely and specifically visualize transplanted β-cells and to improve transplantation outcomes, as transplantation of pancreatic islets has shown promise in treating T1DM. In addition, some of these probes can be applied to the preoperative detection of hidden insulinomas as well. In the present review, we primarily summarize potential tracers under development for imaging β-cells with a focus on tracers for PET, SPECT, MRI, and optical imaging. We will discuss the advantages and limitations of the various imaging probes and extend an outlook on future developments in the field.
Collapse
|
15
|
Abstract
In the 1990s it was discovered that the enzyme dipeptidyl peptidase-4 (DPP-4) inactivates the incretin hormones glucagon-like peptide-1 (GLP-1) and glucose-dependent insulinotropic polypeptide (GIP). DPP-4 inhibition results in raised levels of the two incretin hormones which in turn result in lowering of circulating glucose through stimulation of insulin secretion and inhibition of glucagon secretion. Since then, several small orally available molecules have been developed with DPP-4 inhibitory action. Early studies in the 1990s showed that the DPP-4 inhibitors improve glycemia in animals. Subsequent clinical studies during the 2000s showed a glucose-lowering action of DPP-4 inhibitors also in human subjects with type 2 diabetes. This action was seen when DPP-4 inhibitors were used both as monotherapy and as add-on to other therapies, i.e., metformin, sulfonylureas, tiazolidinediones or exogenous insulin. The DPP-4 inhibitors were also found to have a low risk of adverse events, including hypoglycemia. Five of the DPP-4 inhibitors (sitagliptin, vildagliptin, alogliptin, saxagliptin and linagliptin) were approved by regulatory authorities and entered the market between 2006 and 2013. DPP-4 inhibitors have thereafter undergone long-term cardiovascular outcome trials, showing non-inferiority for risk of major acute cardiovascular endpoints. Also the risk of other potential adverse events is low in these long-term studies. DPP-4 inhibitors are at present included in guidelines as a glucose-lowering concept both as monotherapy and in combination therapies. This article summarizes the development of the DPP-4 inhibition concept from its early stages in the 1990s. The article underscores that the development has its basis in scientific studies on pathophysiology of type 2 diabetes and the importance of targeting the islet dysfunction, that the development has been made possible through academic science in collaboration with the research-oriented pharmaceutical industry, and that the development of a novel concept takes time and requires focused efforts, persistence and long-term perserverance.
Collapse
|
16
|
Exendin-4 attenuates neuronal death via GLP-1R/PI3K/Akt pathway in early brain injury after subarachnoid hemorrhage in rats. Neuropharmacology 2017; 128:142-151. [PMID: 28986282 DOI: 10.1016/j.neuropharm.2017.09.040] [Citation(s) in RCA: 68] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2017] [Revised: 09/06/2017] [Accepted: 09/26/2017] [Indexed: 02/05/2023]
Abstract
Neuronal apoptosis is considered to be a crucial therapeutic target against early brain injury (EBI) after subarachnoid hemorrhage (SAH). Emerging evidence indicates that Exendin-4 (Ex-4), a glucagon-like peptide 1 receptor (GLP-1R) agonist, plays a neuroprotective role in cerebrovascular disease. This study was conducted in order to verify the neuroprotective role of EX-4 in EBI after SAH in rats. The endovascular perforation model of SAH was performed in Sprague-Dawley rats (n = 153). Ex-4 was intraperitoneally injected 1 h after SAH induction in the rats (SAH + Ex-4). To elucidate the underlying molecular mechanism, small interfering ribonucleic acid (siRNA) for GLP-1R and a specific inhibitor of PI3K, LY294002, were injected intracerebroventricularly into SAH + Ex-4 rats before induction of SAH (n = 6 per group). SAH grading evaluation, immunohistochemistry, Western blots, neurobehavioral assessment, and Fluoro-Jade C (FJC) staining experiments were performed. Expression of GLP-1R was significantly increased and mainly expressed in neurons at 24 h after SAH induction. Administration of Ex-4 significantly improved both short- and long-term neurobehavior in SAH + Ex-4 group compared to SAH + Vehicle group after SAH. Ex-4 treatment significantly increased the expression of GLP-1R, PI3K, p-Akt, Bcl-xl, and Bcl-2, while at the same time was found to decrease expression of Bax in the brain. Effects of Ex-4 were reversed by the intervention of GLP-1R siRNA and LY294002 in SAH + Ex-4+GLP-1R siRNA and SAH + Ex-4+LY294002 groups, respectively. In conclusion, the neuroprotective effect of Ex-4 in EBI after SAH was mediated by attenuation of neuronal apoptosis via GLP-1R/PI3K/Akt signaling pathway, therefore EX-4 should be further investigated as a potential therapeutic agent in stroke patients.
Collapse
|
17
|
Pharmacokinetics and Preliminary Pharmacodynamics of Single- and Multiple-dose Lyophilized Recombinant Glucagon-like Peptide-1 Receptor Agonist (rE-4) in Chinese Patients with Type 2 Diabetes Mellitus. Clin Drug Investig 2017; 37:1107-1115. [DOI: 10.1007/s40261-017-0569-1] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023]
|
18
|
Wu J, Williams GR, Branford-White C, Li H, Li Y, Zhu LM. Liraglutide-loaded poly(lactic-co-glycolic acid) microspheres: Preparation and in vivo evaluation. Eur J Pharm Sci 2016; 92:28-38. [DOI: 10.1016/j.ejps.2016.06.018] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/25/2015] [Revised: 06/19/2016] [Accepted: 06/21/2016] [Indexed: 01/10/2023]
|
19
|
Thiebaud N, Llewellyn-Smith IJ, Gribble F, Reimann F, Trapp S, Fadool DA. The incretin hormone glucagon-like peptide 1 increases mitral cell excitability by decreasing conductance of a voltage-dependent potassium channel. J Physiol 2016; 594:2607-28. [PMID: 26931093 PMCID: PMC4865572 DOI: 10.1113/jp272322] [Citation(s) in RCA: 38] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2016] [Accepted: 02/25/2016] [Indexed: 02/06/2023] Open
Abstract
Key points The gut hormone called glucagon‐like peptide 1 (GLP‐1) is a strong moderator of energy homeostasis and communication between the peripheral organs and the brain. GLP‐1 signalling occurs in the brain; using a newly developed genetic reporter line of mice, we have discovered GLP‐synthesizing cells in the olfactory bulb. GLP‐1 increases the firing frequency of neurons (mitral cells) that encode olfactory information by decreasing activity of voltage‐dependent K channels (Kv1.3). Modifying GLP‐1 levels, either therapeutically or following the ingestion of food, could alter the excitability of neurons in the olfactory bulb in a nutrition or energy state‐dependent manner to influence olfactory detection or metabolic sensing. The results of the present study uncover a new function for an olfactory bulb neuron (deep short axon cells, Cajal cells) that could be capable of modifying mitral cell activity through the release of GLP‐1. This might be of relevance for the action of GLP‐1 mimetics now widely used in the treatment of diabetes.
Abstract The olfactory system is intricately linked with the endocrine system where it may serve as a detector of the internal metabolic state or energy homeostasis in addition to its classical function as a sensor of external olfactory information. The recent development of transgenic mGLU‐yellow fluorescent protein mice that express a genetic reporter under the control of the preproglucagon reporter suggested the presence of the gut hormone, glucagon‐like peptide (GLP‐1), in deep short axon cells (Cajal cells) of the olfactory bulb and its neuromodulatory effect on mitral cell (MC) first‐order neurons. A MC target for the peptide was determined using GLP‐1 receptor binding assays, immunocytochemistry for the receptor and injection of fluorescence‐labelled GLP‐1 analogue exendin‐4. Using patch clamp recording of olfactory bulb slices in the whole‐cell configuration, we report that GLP‐1 and its stable analogue exendin‐4 increase the action potential firing frequency of MCs by decreasing the interburst interval rather than modifying the action potential shape, train length or interspike interval. GLP‐1 decreases Kv1.3 channel contribution to outward currents in voltage clamp recordings as determined by pharmacological blockade of Kv1.3 or utilizing mice with Kv1.3 gene‐targeted deletion as a negative control. Because fluctuations in GLP‐1 concentrations monitored by the olfactory bulb can modify the firing frequency of MCs, olfactory coding could change depending upon nutritional or physiological state. As a regulator of neuronal activity, GLP‐1 or its analogue may comprise a new metabolic factor with a potential therapeutic target in the olfactory bulb (i.e. via intranasal delivery) for controlling an imbalance in energy homeostasis. The gut hormone called glucagon‐like peptide 1 (GLP‐1) is a strong moderator of energy homeostasis and communication between the peripheral organs and the brain. GLP‐1 signalling occurs in the brain; using a newly developed genetic reporter line of mice, we have discovered GLP‐synthesizing cells in the olfactory bulb. GLP‐1 increases the firing frequency of neurons (mitral cells) that encode olfactory information by decreasing activity of voltage‐dependent K channels (Kv1.3). Modifying GLP‐1 levels, either therapeutically or following the ingestion of food, could alter the excitability of neurons in the olfactory bulb in a nutrition or energy state‐dependent manner to influence olfactory detection or metabolic sensing. The results of the present study uncover a new function for an olfactory bulb neuron (deep short axon cells, Cajal cells) that could be capable of modifying mitral cell activity through the release of GLP‐1. This might be of relevance for the action of GLP‐1 mimetics now widely used in the treatment of diabetes.
Collapse
Affiliation(s)
- Nicolas Thiebaud
- The Florida State University, Department of Biological Science, Program in Neuroscience, Tallahassee, FL, USA
| | - Ida J Llewellyn-Smith
- Cardiovascular Medicine and Human Physiology, School of Medicine, Flinders University, Bedford Park, SA, Australia
| | - Fiona Gribble
- Cambridge Institute for Medical Research, Addenbrooke's Hospital, Cambridge, UK
| | - Frank Reimann
- Cambridge Institute for Medical Research, Addenbrooke's Hospital, Cambridge, UK
| | - Stefan Trapp
- Centre for Cardiovascular and Metabolic Neuroscience, Department of Neuroscience, Physiology, and Pharmacology, University College London, London, UK.,Department of Surgery and Cancer, Imperial College London, London, UK
| | - Debra Ann Fadool
- The Florida State University, Department of Biological Science, Program in Neuroscience, Tallahassee, FL, USA.,The Florida State University, Institute of Molecular Biophysics, Tallahassee, FL, USA
| |
Collapse
|
20
|
Szcześniak P, Październiok-Holewa A, Klimczak U, Stecko S. Synthesis of β- and γ-Hydroxy α-Amino Acids via Enzymatic Kinetic Resolution and Cyanate-to-Isocyanate Rearrangement. J Org Chem 2014; 79:11700-13. [DOI: 10.1021/jo502026a] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023]
Affiliation(s)
- Piotr Szcześniak
- Institute
of Organic Chemistry, Polish Academy of Sciences, Kasprzaka 44/52, 01-224 Warsaw, Poland
| | - Agnieszka Październiok-Holewa
- Department
of Organic Chemistry, Bioorganic Chemistry and Biotechnology, Silesian University of Technology, Krzywoustego 4, 44-100 Gliwice, Poland
| | - Urszula Klimczak
- Institute
of Organic Chemistry, Polish Academy of Sciences, Kasprzaka 44/52, 01-224 Warsaw, Poland
| | - Sebastian Stecko
- Institute
of Organic Chemistry, Polish Academy of Sciences, Kasprzaka 44/52, 01-224 Warsaw, Poland
| |
Collapse
|
21
|
Gutiérrez-Salmeán G, Ortiz-Vilchis P, Vacaseydel CM, Rubio-Gayosso I, Meaney E, Villarreal F, Ramírez-Sánchez I, Ceballos G. Acute effects of an oral supplement of (-)-epicatechin on postprandial fat and carbohydrate metabolism in normal and overweight subjects. Food Funct 2014; 5:521-7. [PMID: 24458104 DOI: 10.1039/c3fo60416k] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/15/2023]
Abstract
Postprandial hyperglycemia, in particular when accompanied by excessive hypertriglyceridemia, is associated with increased cardiovascular risk, mainly in overweight or obese subjects, as it favors oxidative stress, systemic inflammation and endothelial dysfunction. Thus, treatments that favorably modulate metabolism by reducing steep increases in postprandial serum glucose and triglycerides, are of considerable interest. Evidence suggests that (-)-epicatechin (EPI) is responsible for reductions in cardiometabolic risk associated with chocolate consumption; these effects may be associated with favorable effects of EPI on postprandial metabolism. The aims of this study were to assess the effects of EPI on postprandial metabolism in normal-weight and overweight/obese subjects. Twenty adult volunteers (normal and overweight) underwent oral metabolic tolerance tests in the absence and presence of oral EPI (1 mg kg(-1)). Metabolic responses were examined using indirect calorimetry and determining blood glucose and triglycerides at 0, 2 and 4 hours after metabolic load ingestion. Results show that EPI increased postprandial lipid catabolism, as evidenced by a significant decrease in the respiratory quotient, which implies an increase in fat oxidation. The effect was associated with significantly lower postprandial plasma glucose and triglycerides concentrations. The effects were more prominent in overweight subjects. In conclusion, EPI modulates postprandial metabolism by enhancing lipid oxidation accompanied by reductions in glycemia and triglyceridemia.
Collapse
Affiliation(s)
- Gabriela Gutiérrez-Salmeán
- Laboratorio de Investigación Integral Cardiometabólica, Escuela Superior de Medicina, Instituto Politécnico Nacional, Plan de San Luis y Díaz Mirón s/n. Casco de Santo Tomás, Miguel Hidalgo. C.P. 11340, Mexico City, Mexico.
| | | | | | | | | | | | | | | |
Collapse
|
22
|
Yang X, Li Y, Wang Y, Zheng X, Kong W, Meng F, Zhou Z, Liu C, Li Y, Gong M. Long-Acting GLP-1 Analogue in V-Shaped Conformation by Terminal Polylysine Modifications. Mol Pharm 2014; 11:4092-9. [PMID: 25243635 DOI: 10.1021/mp5002685] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/21/2023]
Affiliation(s)
- Xue Yang
- Department of Pharmacy, Tianjin Traditional Medicine University, Tianjin, China
| | - Ying Li
- Tianjin Neurological
Institute, Tianjin Medical University General Hospital, Tianjin, China
| | - Yuli Wang
- Tianjin Institute of Pharmaceutical Research, Tianjin, China
| | - Xuemin Zheng
- Tianjin Institute of Pharmaceutical Research, Tianjin, China
| | - Weiling Kong
- Tianjin Institute of Pharmaceutical Research, Tianjin, China
| | - Fancui Meng
- Tianjin Institute of Pharmaceutical Research, Tianjin, China
| | - Zhixing Zhou
- Tianjin Institute of Pharmaceutical Research, Tianjin, China
| | - Changxiao Liu
- Tianjin Institute of Pharmaceutical Research, Tianjin, China
| | - Ying Li
- Tianjin Neurological
Institute, Tianjin Medical University General Hospital, Tianjin, China
| | - Min Gong
- Tianjin Institute of Pharmaceutical Research, Tianjin, China
| |
Collapse
|
23
|
Selvaraju RK, Velikyan I, Asplund V, Johansson L, Wu Z, Todorov I, Shively J, Kandeel F, Eriksson B, Korsgren O, Eriksson O. Pre-clinical evaluation of [(68)Ga]Ga-DO3A-VS-Cys(40)-Exendin-4 for imaging of insulinoma. Nucl Med Biol 2014; 41:471-6. [PMID: 24857864 DOI: 10.1016/j.nucmedbio.2014.03.017] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2013] [Revised: 02/07/2014] [Accepted: 03/10/2014] [Indexed: 12/26/2022]
Abstract
INTRODUCTION Insulinoma is the most common form of pancreatic endocrine tumors responsible for hyperinsulinism in adults. These tumors overexpress glucagon like peptide-1 (GLP-1) receptor, and biologically stable GLP-1 analogs have therefore been proposed as potential imaging agents. Here, we evaluate the potential of a positron emission tomography (PET) tracer, [(68)Ga]Ga-DO3A-VS-Cys(40)-Exendin-4, for imaging and quantification of GLP-1 receptors (GLP-1R) in insulinoma. METHODS [(68)Ga]Ga-DO3A-VS-Cys(40)-Exendin-4 was evaluated for binding to GLP-1R by in vitro autoradiography binding studies in INS-1 tumor from xenografts. In vivo biodistribution was investigated in healthy control mice, INS-1 xenografted and PANC1 xenografted immunodeficient mice at two different doses of peptide: 2.5μg/kg (baseline) and 100μg/kg (block). In vivo imaging of [(68)Ga]Ga-DO3A-VS-Cys(40)-Exendin-4 in xenografted mice was evaluated by small animal PET/CT using a direct comparison with the clinically established insulinoma marker [(11)C]5-hydroxy-tryptophan ([(11)C]5-HTP). RESULTS GLP-1 receptor density could be quantified in INS-1 tumor biopsies. [(68)Ga]Ga-DO3A-VS-Cys(40)-Exendin-4 showed significant uptake (p≤0.05) in GLP1-R positive tissues such as INS-1 tumor, lungs and pancreas upon comparison between baseline and blocking studies. In vivo imaging showed concordant results with higher tumor-to-muscle ratio in INS-1 xenografted mice compared with [(11)C]5-HTP. CONCLUSION [(68)Ga]Ga-DO3A-VS-Cys(40)-Exendin-4 has high affinity and specificity for GLP-1R expressed on insulinoma in vitro and in vivo.
Collapse
Affiliation(s)
- Ram Kumar Selvaraju
- Preclinical PET Platform (PPP), Department of Medicinal Chemistry, Uppsala University, SE-751 83 Uppsala, Sweden.
| | - Irina Velikyan
- Preclinical PET Platform (PPP), Department of Medicinal Chemistry, Uppsala University, SE-751 83 Uppsala, Sweden; Department of Radiology, Oncology and Radiation Sciences, Uppsala University, SE-751 85 Uppsala, Sweden; PET Centre, Centre for Medical Imaging, Uppsala University Hospital, SE-751 85 Uppsala, Sweden
| | - Veronika Asplund
- Preclinical PET Platform (PPP), Department of Medicinal Chemistry, Uppsala University, SE-751 83 Uppsala, Sweden
| | - Lars Johansson
- Department of Radiology, Oncology and Radiation Sciences, Uppsala University, SE-751 85 Uppsala, Sweden; AstraZeneca R&D, SE-431 83 Mölndal, Sweden
| | - Zhanhong Wu
- Beckman Research Institute of the City of Hope, Duarte, CA 91010, USA
| | - Ivan Todorov
- Beckman Research Institute of the City of Hope, Duarte, CA 91010, USA
| | - Jack Shively
- Beckman Research Institute of the City of Hope, Duarte, CA 91010, USA
| | - Fouad Kandeel
- Beckman Research Institute of the City of Hope, Duarte, CA 91010, USA
| | - Barbro Eriksson
- Department of Medical Sciences, Uppsala University Hospital, SE-751 85 Uppsala, Sweden
| | - Olle Korsgren
- Department of Immunology, Genetics and Pathology, Uppsala University, SE-751 85 Uppsala, Sweden
| | - Olof Eriksson
- Preclinical PET Platform (PPP), Department of Medicinal Chemistry, Uppsala University, SE-751 83 Uppsala, Sweden
| |
Collapse
|
24
|
Nalin L, Selvaraju RK, Velikyan I, Berglund M, Andréasson S, Wikstrand A, Rydén A, Lubberink M, Kandeel F, Nyman G, Korsgren O, Eriksson O, Jensen-Waern M. Positron emission tomography imaging of the glucagon-like peptide-1 receptor in healthy and streptozotocin-induced diabetic pigs. Eur J Nucl Med Mol Imaging 2014; 41:1800-10. [PMID: 24643781 DOI: 10.1007/s00259-014-2745-3] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2013] [Accepted: 02/21/2014] [Indexed: 01/13/2023]
Abstract
PURPOSE The glucagon-like peptide-1 receptor (GLP-1R) has been proposed as a target for molecular imaging of beta cells. The feasibility of non-invasive imaging and quantification of GLP-1R in pancreas using the positron emission tomography (PET) tracer [(68)Ga]Ga-DO3A-VS-Cys(40)-Exendin-4 in non-diabetic and streptozotocin (STZ)-induced diabetic pigs treated with insulin was investigated. METHODS Non-diabetic (n = 4) and STZ-induced diabetic pigs (n = 3) from the same litter were examined. Development of diabetes was confirmed by blood glucose values, clinical examinations and insulin staining of pancreatic sections post mortem. Tissue perfusion in the pancreas and kidneys was evaluated by [(15)O]water PET/computed tomography (CT) scans. The in vivo receptor specificity of [(68)Ga]Ga-DO3A-VS-Cys(40)-Exendin-4 was assessed by administration of either tracer alone or by competition with 3-6.5 μg/kg of Exendin-4. Volume of distribution and occupancy in the pancreas were quantified with a single tissue compartment model. RESULTS [(15)O]water PET/CT examinations showed reduced perfusion in the pancreas and kidneys in diabetic pigs. [(68)Ga]Ga-DO3A-VS-Cys(40)-Exendin-4 uptake in the pancreas of both non-diabetic and diabetic pigs was almost completely abolished by co-injection of unlabeled Exendin-4 peptide. [(68)Ga]Ga-DO3A-VS-Cys(40)-Exendin-4 uptake did not differ between non-diabetic and diabetic pigs. In all animals, administration of the tracer resulted in an immediate increase in the heart rate (HR). CONCLUSION Pancreatic uptake of [(68)Ga]Ga-DO3A-VS-Cys(40)-Exendin-4 was not reduced by destruction of beta cells in STZ-induced diabetic pigs.
Collapse
Affiliation(s)
- Lovisa Nalin
- Department of Clinical Sciences, Faculty of Veterinary Medicine and Animal Science, Swedish University of Agricultural Sciences, Uppsala, Sweden
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
25
|
Han J, Sun L, Chu Y, Li Z, Huang D, Zhu X, Qian H, Huang W. Design, Synthesis, and Biological Activity of Novel Dicoumarol Glucagon-like Peptide 1 Conjugates. J Med Chem 2013; 56:9955-68. [DOI: 10.1021/jm4017448] [Citation(s) in RCA: 55] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Affiliation(s)
- Jing Han
- Center of Drug Discovery,
State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing 210009, P. R. China
| | - Lidan Sun
- Center of Drug Discovery,
State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing 210009, P. R. China
| | - Yingying Chu
- Center of Drug Discovery,
State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing 210009, P. R. China
| | - Zheng Li
- Center of Drug Discovery,
State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing 210009, P. R. China
| | - Dandan Huang
- Center of Drug Discovery,
State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing 210009, P. R. China
| | - Xiaoyun Zhu
- Center of Drug Discovery,
State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing 210009, P. R. China
| | - Hai Qian
- Center of Drug Discovery,
State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing 210009, P. R. China
| | - Wenlong Huang
- Center of Drug Discovery,
State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing 210009, P. R. China
| |
Collapse
|
26
|
Lee J, Jung DW, Kim WH, Um JI, Yim SH, Oh WK, Williams DR. Development of a highly visual, simple, and rapid test for the discovery of novel insulin mimetics in living vertebrates. ACS Chem Biol 2013; 8:1803-14. [PMID: 23725454 DOI: 10.1021/cb4000162] [Citation(s) in RCA: 50] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Diabetes mellitus is a global epidemic with major impacts on human health and society. Drug discovery for diabetes can be facilitated by the development of a rapid, vertebrate-based screen for identifying new insulin mimetic compounds. Our study describes the first development of a zebrafish-based system based on direct monitoring of glucose flux and validated for identifying novel anti-diabetic drugs. Our system utilizes a fluorescent-tagged glucose probe in an experimentally convenient 96-well plate format. To validate our new system, we identified compounds that can induce glucose uptake via activity-guided fractionation of the inner shell from the Japanese Chestnut (Castanea crenata). The best performing compound, UP3.2, was identified as fraxidin and validated as a novel insulin mimetic using a mammalian adipocyte system. Additional screening using sets of saponin- and triazine-based compounds was undertaken to further validate this assay, which led to the discovery of triazine PP-II-A03 as a novel insulin mimetic. Moreover, we demonstrate that our zebrafish-based system allows concomitant toxicological analysis of anti-diabetic drug candidates. Thus, we have developed a rapid and inexpensive vertebrate model that can enhance diabetes drug discovery by preselecting hits from chemical library screens, before testing in relatively expensive rodent assays.
Collapse
Affiliation(s)
| | | | | | | | | | - Won Keun Oh
- Korea Bioactive Natural Material
Bank, College of Pharmacy, Seoul National University, Seoul 151-742, Republic of Korea
| | | |
Collapse
|
27
|
Li Y, Shao M, Zheng X, Kong W, Zhang J, Gong M. Self-assembling peptides improve the stability of glucagon-like peptide-1 by forming a stable and sustained complex. Mol Pharm 2013; 10:3356-65. [PMID: 23859692 DOI: 10.1021/mp4001734] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
The multiple physiological characterizations of glucagon-like peptide-1 (GLP-1) make it a promising drug candidate for the treatment of T2DM. However, the short half-life of GLP-1 limits its clinical utility. Self-assembling peptides are presumed to wrap GLP-1 peptide, and this helps to prolong the stability of GLP-1 consequently. The aim of this study was to investigate whether self-assembling peptides could be applied to prolong the half-life of GLP-1 as sustained release preparations. In this study, five different self-assembling peptides were employed. The formation of the complexes was monitored using gel filtration and mass spectrometry and was simulated by Molecular Dynamics. Stabilization, insulin secretion stimulation, and glucose tolerance tests were performed to investigate the physiological characteristics retained by GLP-1 following complex formation with self-assembling peptides. Our findings revealed that, among the five different self-assembling peptides tested, complex of Pep-1 and GLP-1 exhibited a remarkable extension in the half-life of GLP-1. In addition, the experimental animals treated with a GLP-1/Pep-1 complex exhibited better blood glucose clearance activity over a greater duration of time than the animals treated with GLP-1 alone. Based on our results, an adjustment of the Pep-1 and GLP-1 ratios is presumed to be able to control the half-life of GLP-1 (e.g., medium-acting and long-acting). Collectively, the findings in this study suggest that the self-assembling peptide Pep-1 could serve as a powerful drug preparation tool to extend the short half-life of therapeutic peptides.
Collapse
Affiliation(s)
- Ying Li
- Tianjin Neurological Institute, Tianjin Medical University General Hospital, China
| | | | | | | | | | | |
Collapse
|
28
|
Novel fatty chain-modified glucagon-like peptide-1 conjugates with enhanced stability and prolonged in vivo activity. Biochem Pharmacol 2013; 86:297-308. [DOI: 10.1016/j.bcp.2013.05.012] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2013] [Revised: 05/14/2013] [Accepted: 05/14/2013] [Indexed: 11/21/2022]
|
29
|
Abstract
The story of oxytocin (OXT) began long ago in evolutionary terms with its recognition as a classical neurohypophyseal hormone important for lactation and uterine contraction. With the recent discovery of its local actions in the brain, its previously-unappreciated diverse functions in regulating social behaviors and metabolic physiology are emerging. In light of metabolic control, OXT has been shown to induce feeding restriction and body weight lowering through acting on brain regulatory regions, in particular the hypothalamus. Studies from pharmacologic interventions and genetic manipulations demonstrated that OXT can play significant roles in affecting glucose metabolism as well as insulin secretion and lipolysis, many of those functions being regulated both centrally and peripherally. Also excitingly, recent therapeutic success was obtained in clinical endeavor showing that OXT nasal spray effectively induced weight loss and metabolic improvement in human patients with obesity, thus further indicating OXT as a tangible drug target for treating obesity and metabolic complications. In addition to the native form, OXT-derived analogues have been found effective in inducing body weight control and glucose balance. Altogether, all recent advances in studying OXT and metabolic regulation has promoted a promising foundation for the therapeutic strategy of developing innovative OXT peptidyl drugs for the treatment of obesity and related metabolic diseases.
Collapse
|
30
|
Zhang H, Wu C, Chen Q, Chen X, Xu Z, Wu J, Cai D. Treatment of obesity and diabetes using oxytocin or analogs in patients and mouse models. PLoS One 2013; 8:e61477. [PMID: 23700406 PMCID: PMC3658979 DOI: 10.1371/journal.pone.0061477] [Citation(s) in RCA: 149] [Impact Index Per Article: 12.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2012] [Accepted: 03/09/2013] [Indexed: 01/06/2023] Open
Abstract
Obesity is important for the development of type-2 diabetes as a result of obesity-induced insulin resistance accompanied by impaired compensation of insulin secretion from pancreatic beta cells. Here, based on a randomized pilot clinical trial, we report that intranasal oxytocin administration over an 8-week period led to effective reduction of obesity and reversal of related prediabetic changes in patients. Using mouse models, we further systematically evaluated whether oxytocin and its analogs yield therapeutic effects against prediabetic or diabetic disorders regardless of obesity. Our results showed that oxytocin and two analogs including [Ser4, Ile8]-oxytocin or [Asu1,6]-oxytocin worked in mice to reverse insulin resistance and glucose intolerance prior to reduction of obesity. In parallel, using streptozotocin-induced diabetic mouse model, we found that treatment with oxytocin or its analogs reduced the magnitude of glucose intolerance through improving insulin secretion. The anti-diabetic effects of oxytocin and its analogs in these animal models can be produced similarly whether central or peripheral administration was used. In conclusion, oxytocin and its analogs have multi-level effects in improving weight control, insulin sensitivity and insulin secretion, and bear potentials for being developed as therapeutic peptides for obesity and diabetes.
Collapse
Affiliation(s)
- Hai Zhang
- Department of Molecular Pharmacology, Diabetes Research Center, Institute of Aging, Albert Einstein College of Medicine, Bronx, New York, United States of America
| | - Chenguang Wu
- Department of Medicine, Endocrine Division, the Affiliated People’s Hospital of Jiangsu University, Jiangsu Province, Zhenjiang, P. R. China
| | - Qiaofen Chen
- Department of Medicine, Endocrine Division, the Affiliated People’s Hospital of Jiangsu University, Jiangsu Province, Zhenjiang, P. R. China
| | - Xiaoluo Chen
- Department of Medicine, Endocrine Division, the Affiliated People’s Hospital of Jiangsu University, Jiangsu Province, Zhenjiang, P. R. China
| | - Zhigang Xu
- Department of Medicine, Endocrine Division, the Affiliated People’s Hospital of Jiangsu University, Jiangsu Province, Zhenjiang, P. R. China
| | - Jing Wu
- Department of Medicine, Endocrine Division, the Affiliated People’s Hospital of Jiangsu University, Jiangsu Province, Zhenjiang, P. R. China
| | - Dongsheng Cai
- Department of Molecular Pharmacology, Diabetes Research Center, Institute of Aging, Albert Einstein College of Medicine, Bronx, New York, United States of America
- * E-mail:
| |
Collapse
|
31
|
Darout E, Robinson RP, McClure KF, Corbett M, Li B, Shavnya A, Andrews MP, Jones CS, Li Q, Minich ML, Mascitti V, Guimarães CRW, Munchhof MJ, Bahnck KB, Cai C, Price DA, Liras S, Bonin PD, Cornelius P, Wang R, Bagdasarian V, Sobota CP, Hornby S, Masterson VM, Joseph RM, Kalgutkar AS, Chen Y. Design and Synthesis of Diazatricyclodecane Agonists of the G-Protein-Coupled Receptor 119. J Med Chem 2012; 56:301-19. [DOI: 10.1021/jm301626p] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/29/2023]
Affiliation(s)
- Etzer Darout
- Departments of Medicinal Chemistry,
Discovery Biology, Drug Metabolism, and Pharmaceutical Sciences, Pfizer Worldwide Research and Development, 620 Memorial
Drive, Cambridge, Massachusetts 02139, United States
| | - Ralph P. Robinson
- Departments of Medicinal Chemistry,
Discovery Biology, Drug Metabolism, and Pharmaceutical Sciences, Pfizer Worldwide Research and Development, Eastern
Point Road, Groton, Connecticut 06340, United States
| | - Kim F. McClure
- Departments of Medicinal Chemistry,
Discovery Biology, Drug Metabolism, and Pharmaceutical Sciences, Pfizer Worldwide Research and Development, 620 Memorial
Drive, Cambridge, Massachusetts 02139, United States
| | - Matthew Corbett
- Departments of Medicinal Chemistry,
Discovery Biology, Drug Metabolism, and Pharmaceutical Sciences, Pfizer Worldwide Research and Development, Eastern
Point Road, Groton, Connecticut 06340, United States
| | - Bryan Li
- Departments of Medicinal Chemistry,
Discovery Biology, Drug Metabolism, and Pharmaceutical Sciences, Pfizer Worldwide Research and Development, Eastern
Point Road, Groton, Connecticut 06340, United States
| | - Andrei Shavnya
- Departments of Medicinal Chemistry,
Discovery Biology, Drug Metabolism, and Pharmaceutical Sciences, Pfizer Worldwide Research and Development, Eastern
Point Road, Groton, Connecticut 06340, United States
| | - Melissa P. Andrews
- Departments of Medicinal Chemistry,
Discovery Biology, Drug Metabolism, and Pharmaceutical Sciences, Pfizer Worldwide Research and Development, Eastern
Point Road, Groton, Connecticut 06340, United States
| | - Christopher S. Jones
- Departments of Medicinal Chemistry,
Discovery Biology, Drug Metabolism, and Pharmaceutical Sciences, Pfizer Worldwide Research and Development, Eastern
Point Road, Groton, Connecticut 06340, United States
| | - Qifang, Li
- Departments of Medicinal Chemistry,
Discovery Biology, Drug Metabolism, and Pharmaceutical Sciences, Pfizer Worldwide Research and Development, Eastern
Point Road, Groton, Connecticut 06340, United States
| | - Martha L. Minich
- Departments of Medicinal Chemistry,
Discovery Biology, Drug Metabolism, and Pharmaceutical Sciences, Pfizer Worldwide Research and Development, Eastern
Point Road, Groton, Connecticut 06340, United States
| | - Vincent Mascitti
- Departments of Medicinal Chemistry,
Discovery Biology, Drug Metabolism, and Pharmaceutical Sciences, Pfizer Worldwide Research and Development, Eastern
Point Road, Groton, Connecticut 06340, United States
| | - Cristiano R. W. Guimarães
- Departments of Medicinal Chemistry,
Discovery Biology, Drug Metabolism, and Pharmaceutical Sciences, Pfizer Worldwide Research and Development, 620 Memorial
Drive, Cambridge, Massachusetts 02139, United States
| | - Michael J. Munchhof
- Departments of Medicinal Chemistry,
Discovery Biology, Drug Metabolism, and Pharmaceutical Sciences, Pfizer Worldwide Research and Development, Eastern
Point Road, Groton, Connecticut 06340, United States
| | - Kevin B. Bahnck
- Departments of Medicinal Chemistry,
Discovery Biology, Drug Metabolism, and Pharmaceutical Sciences, Pfizer Worldwide Research and Development, Eastern
Point Road, Groton, Connecticut 06340, United States
| | - Cuiman Cai
- Departments of Medicinal Chemistry,
Discovery Biology, Drug Metabolism, and Pharmaceutical Sciences, Pfizer Worldwide Research and Development, Eastern
Point Road, Groton, Connecticut 06340, United States
| | - David A. Price
- Departments of Medicinal Chemistry,
Discovery Biology, Drug Metabolism, and Pharmaceutical Sciences, Pfizer Worldwide Research and Development, 620 Memorial
Drive, Cambridge, Massachusetts 02139, United States
| | - Spiros Liras
- Departments of Medicinal Chemistry,
Discovery Biology, Drug Metabolism, and Pharmaceutical Sciences, Pfizer Worldwide Research and Development, 620 Memorial
Drive, Cambridge, Massachusetts 02139, United States
| | - Paul D. Bonin
- Departments of Medicinal Chemistry,
Discovery Biology, Drug Metabolism, and Pharmaceutical Sciences, Pfizer Worldwide Research and Development, Eastern
Point Road, Groton, Connecticut 06340, United States
| | - Peter Cornelius
- Departments of Medicinal Chemistry,
Discovery Biology, Drug Metabolism, and Pharmaceutical Sciences, Pfizer Worldwide Research and Development, Eastern
Point Road, Groton, Connecticut 06340, United States
| | - Ruduan Wang
- Departments of Medicinal Chemistry,
Discovery Biology, Drug Metabolism, and Pharmaceutical Sciences, Pfizer Worldwide Research and Development, Eastern
Point Road, Groton, Connecticut 06340, United States
| | - Victoria Bagdasarian
- Departments of Medicinal Chemistry,
Discovery Biology, Drug Metabolism, and Pharmaceutical Sciences, Pfizer Worldwide Research and Development, Eastern
Point Road, Groton, Connecticut 06340, United States
| | - Colleen P. Sobota
- Departments of Medicinal Chemistry,
Discovery Biology, Drug Metabolism, and Pharmaceutical Sciences, Pfizer Worldwide Research and Development, Eastern
Point Road, Groton, Connecticut 06340, United States
| | - Sam Hornby
- Departments of Medicinal Chemistry,
Discovery Biology, Drug Metabolism, and Pharmaceutical Sciences, Pfizer Worldwide Research and Development, Eastern
Point Road, Groton, Connecticut 06340, United States
| | - Victoria M. Masterson
- Departments of Medicinal Chemistry,
Discovery Biology, Drug Metabolism, and Pharmaceutical Sciences, Pfizer Worldwide Research and Development, Eastern
Point Road, Groton, Connecticut 06340, United States
| | - Reena M. Joseph
- Departments of Medicinal Chemistry,
Discovery Biology, Drug Metabolism, and Pharmaceutical Sciences, Pfizer Worldwide Research and Development, Eastern
Point Road, Groton, Connecticut 06340, United States
| | - Amit S. Kalgutkar
- Departments of Medicinal Chemistry,
Discovery Biology, Drug Metabolism, and Pharmaceutical Sciences, Pfizer Worldwide Research and Development, 620 Memorial
Drive, Cambridge, Massachusetts 02139, United States
| | - Yue Chen
- Departments of Medicinal Chemistry,
Discovery Biology, Drug Metabolism, and Pharmaceutical Sciences, Pfizer Worldwide Research and Development, Eastern
Point Road, Groton, Connecticut 06340, United States
| |
Collapse
|
32
|
Cao Z, Li Y, Tang L, Xu W, Liu C, Zhang J, Gong M. Formation of cyclic structure at amino-terminus of glucagon-like peptide-1 exhibited a prolonged half-life in vivo. Diabetes Res Clin Pract 2012; 96:362-70. [PMID: 22284602 DOI: 10.1016/j.diabres.2012.01.010] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/12/2011] [Revised: 12/12/2011] [Accepted: 01/03/2012] [Indexed: 11/20/2022]
Abstract
The multiple physiological characterizations of glucagon-like peptide-1 (GLP-1) make it a promising drug candidate for the therapy of type 2 diabetes. However, the biological half-life of GLP-1 is short in vivo due to degradation by dipeptidyl peptidase-IV (DPP-IV) and renal clearance. The stabilization of GLP-1 is critical for its utility in drug development. In this study, several GLP-1 mutants containing an N-terminal cyclic conformation were prepared in that the existence of cyclic conformation is predicted to increase the stabilization of GLP-1 in vivo. In this study, the binding capacities of the mutants were determined, the stabilities of the mutants were investigated and the physiological functions of the mutants were compared with those of wild-type GLP-1 in animals. The results indicated that the mutant (GLP1N8) remarkably raised the half-life in vivo; it also showed better glucose tolerance and higher HbA(1c) reduction than GLP-1 and exendin-4 in rodents. These results suggest that the GLP-1 analog (GLP1N8) which contains an N-terminal cyclic structure might be utilized as possible potent anti-diabetic drugs in the treatment of type 2 diabetes mellitus.
Collapse
Affiliation(s)
- Zhenghong Cao
- Department of Pharmacy, Tianjin University of Traditional Medicine, China
| | | | | | | | | | | | | |
Collapse
|
33
|
Gutierrez AD, Balasubramanyam A. Dysregulation of glucose metabolism in HIV patients: epidemiology, mechanisms, and management. Endocrine 2012; 41:1-10. [PMID: 22134974 PMCID: PMC3417129 DOI: 10.1007/s12020-011-9565-z] [Citation(s) in RCA: 57] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/22/2011] [Accepted: 11/07/2011] [Indexed: 02/06/2023]
Abstract
HIV-infected patients on highly active antiretroviral therapy (HAART) have increased prevalence of a number of chronic metabolic disorders of multifactorial but unclear etiology. These include disorders of lipid metabolism with or without lipodystrophy, insulin resistance, and an increased prevalence of impaired glucose tolerance, diabetes mellitus, and cardiometabolic syndrome. While much attention has been focused on the lipid and cardiovascular disorders, few investigations have attempted to characterize the prevalence, incidence, etiology, mechanisms, and management of glycemic disorders in HIV patients. In this review, we have focused specifically on a comprehensive assessment of dysglycemia in the context of HIV infection and HAART.
Collapse
Affiliation(s)
- Absalon D. Gutierrez
- Translational Metabolism Unit, Diabetes and Endocrinology Research Center, Division of Diabetes, Endocrinology and Metabolism, Baylor College of Medicine; Houston, Texas, USA
| | - Ashok Balasubramanyam
- Translational Metabolism Unit, Diabetes and Endocrinology Research Center, Division of Diabetes, Endocrinology and Metabolism, Baylor College of Medicine; Houston, Texas, USA
- Endocrine Service, Ben Taub General Hospital, Houston, Texas, USA
| |
Collapse
|
34
|
Glucagon-like peptide-1 activates endothelial nitric oxide synthase in human umbilical vein endothelial cells. Acta Pharmacol Sin 2012; 33:75-81. [PMID: 22120969 DOI: 10.1038/aps.2011.149] [Citation(s) in RCA: 101] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Abstract
AIM To investigate the effects of glucagon-like peptide-1 (GLP-1) on endothelial NO synthase (eNOS) in human umbilical vein endothelial cells (HUVECs), and elucidate whether GLP-1 receptor (GLP-1R) and GLP-1(9-36) are involved in these effects. METHODS HUVECs were used. The activity of eNOS was measured with NOS assay kit. Phosphorylated and total eNOS proteins were detected using Western blot analysis. The level of eNOS mRNA was quantified with real-time RT-PCR. RESULTS Incubation of HUVECs with GLP-1 (50-5000 pmol/L) for 30 min significantly increased the activity of eNOS. Incubation of HUVECs with GLP-1 (500-5000 pmol/L) for 5 or 10 min increased eNOS phosphorylated at ser-1177. Incubation with GLP-1 (5000 pmol/L) for 48 h elevated the level of eNOS protein, did not affect the level of eNOS mRNA. GLP-1R agonists exenatide and GLP-1(9-36) at the concentration of 5000 pmol/L increased the activity, phosphorylation and protein level of eNOS. GLP-1R antagonist exendin(9-39) or DPP-4 inhibitor sitagliptin, which abolished GLP-1(9-36) formation, at the concentration of 5000 pmol/L partially blocked the effects of GLP-1 on eNOS. CONCLUSION GLP-1 upregulated the activity and protein expression of eNOS in HUVECs through the GLP-1R-dependent and GLP-1(9-36)-related pathways. GLP-1 may prevent or delay the formation of atherosclerosis in diabetes mellitus by improving the function of eNOS.
Collapse
|
35
|
Congreve M, Langmead CJ, Mason JS, Marshall FH. Progress in structure based drug design for G protein-coupled receptors. J Med Chem 2011; 54:4283-311. [PMID: 21615150 PMCID: PMC3308205 DOI: 10.1021/jm200371q] [Citation(s) in RCA: 166] [Impact Index Per Article: 11.9] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2011] [Indexed: 12/12/2022]
Affiliation(s)
- Miles Congreve
- Heptares Therapeutics Limited, BioPark, Welwyn Garden City, Hertfordshire, UK.
| | | | | | | |
Collapse
|
36
|
Li Y, Li X, Zheng X, Tang L, Xu W, Gong M. Disulfide bond prolongs the half-life of therapeutic peptide-GLP-1. Peptides 2011; 32:1400-7. [PMID: 21600946 DOI: 10.1016/j.peptides.2011.05.003] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/30/2011] [Revised: 05/04/2011] [Accepted: 05/04/2011] [Indexed: 11/19/2022]
Abstract
The multiple physiological characterization of glucagon-like peptide-1 (GLP-1) makes it a promising drug candidate for the therapy of type 2 diabetes. However, the half-life of GLP-1 is short in vivo due to rapid degradation by dipeptidyl peptidase-IV (DPP-IV) and renal clearance. This indicates that the stabilization of GLP-1 is critical for its utility in drug development. In this study, we developed a cluster of GLP-1 homodimeric analogs, which fused the mutated GLP-1 monomer by an intra-disulfide bridge. The stabilities of the GLP-1 homodimeric analogs were investigated and the physiological functions of the analogs were compared with those of wild-type GLP-1 in rats and human serum. Single dose glucose tolerance test was performed to investigate the administration frequency which satisfied the efficient glucose regulatory in rats. Multiple dose glucose tolerance tests were employed also to study the long-acting anti-diabetic activity of GLP-1 homodimeric analog. The results indicated that the GLP-1 homodimeric analog (hdGLP1G10C) remarkably raised the biological half-life of GLP-1; also HDGLP1G10C showed better glucose tolerance and higher HbA(1c) reduction than GLP-1 in rodents. Based upon the results in this study, it was suggested that hdGLP1G10C prolonged the stability of GLP-1 and retained the biological activity of GLP-1. The improved physiological characterization of hdGLP1G10C makes it as possible potent anti-diabetic drug in the treatment of type 2 diabetes mellitus.
Collapse
Affiliation(s)
- Ying Li
- School of Biosciences, University of Birmingham, United Kingdom
| | | | | | | | | | | |
Collapse
|
37
|
Li Y, Xu W, Tang L, Gong M, Zhang J. A novel GLP-1 analog exhibits potent utility in the treatment of type 2 diabetes with an extended half-life and efficient glucose clearance in vivo. Peptides 2011; 32:1408-14. [PMID: 21664938 DOI: 10.1016/j.peptides.2011.05.026] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/03/2011] [Revised: 05/26/2011] [Accepted: 05/26/2011] [Indexed: 11/24/2022]
Abstract
The multiple physiological characterizations of glucagon-like peptide-1 (GLP-1) make it a promising drug candidate for the therapy of type 2 diabetes. However, the half-life of GLP-1 is short in vivo due to degradation by dipeptidyl peptidase-IV (DPP-IV) and renal clearance. Therefore, the stabilization of GLP-1 is critical for its utility in drug development. Based on our previous research, a GLP-1 analog that contained an intra-disulfide bond exhibited a prolonged biological half-life. In this study, we improved upon previous analogs with a novel GLP-1 analog that contained a tryptophan cage-like sequence for an improved binding affinity to the GLP-1 receptor. The binding capacities and the stabilities of GLP715a were investigated, and the physiological functions of the GLP715a were compared to those of the wild-type GLP-1 in animals. The results demonstrated that the new GLP-1 analog (GLP715a) increased its biological half-life to approximately 48h in vivo; GLP715a also exhibited a higher binding affinity to the GLP-1 receptor than the wild-type GLP-1. The increased binding capacity of GLP715a to its receptor resulted in a quick response to glucose administration. The long-acting anti-diabetic property of GLP715a was revealed by its increased glucose tolerance, higher HbA(1c) reduction, more efficient glucose clearance and quicker insulin stimulation upon glucose administration compared to the wild-type GLP-1 in rodents. The improved physiological characterizations of GLP715a make it a possible potent anti-diabetic drug in the treatment of type 2 diabetes mellitus.
Collapse
Affiliation(s)
- Ying Li
- School of Bioscience, University of Birmingham, UK
| | | | | | | | | |
Collapse
|
38
|
Li Y, Zheng X, Tang L, Xu W, Gong M. GLP-1 analogs containing disulfide bond exhibited prolonged half-life in vivo than GLP-1. Peptides 2011; 32:1303-12. [PMID: 21515323 DOI: 10.1016/j.peptides.2011.04.001] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/08/2011] [Revised: 04/07/2011] [Accepted: 04/07/2011] [Indexed: 01/08/2023]
Abstract
The multiple physiological characterizations of glucagon-like peptide-1 (GLP-1) make it a promising drug candidate for the therapy of type 2 diabetes. However, the half-life of GLP-1 is short in vivo due to degradation by dipeptidyl peptidase-IV (DPP-IV) and renal clearance. This indicates that the stabilization of GLP-1 is critical for its utility in drug development. In this study, we developed a cluster of GLP-1 mutants containing an inter-disulfide bond that is predicted to increase the half-life of GLP-1 in vivo. Exendin-4 was also mutated with a disulfide bond similar to the GLP-1 analogs. In this study, the binding capacities of the mutants were determined, the stabilities of the mutants were investigated and the physiological functions of the mutants were compared with those of wild-type GLP-1 and exendin-4 in animals. The results indicated that the mutants remarkably raised the half-life in vivo; they also showed better glucose tolerance and higher HbA(1c) reduction than GLP-1 and exendin-4 in rodents. These results suggest that GLP-1 and exendin-4 mutants containing disulfide bonds might be utilized as possible potent anti-diabetic drugs in the treatment of type 2 diabetes mellitus.
Collapse
Affiliation(s)
- Ying Li
- School of Biosciences, University of Birmingham, UK
| | | | | | | | | |
Collapse
|
39
|
Zheng X, Li Y, Fu G, Gong M. Application of novel peptide (Pp1) improving the half-life of exendin-4 in vivo. Peptides 2011; 32:964-70. [PMID: 21334413 DOI: 10.1016/j.peptides.2011.02.009] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/14/2010] [Revised: 02/10/2011] [Accepted: 02/12/2011] [Indexed: 10/18/2022]
Abstract
AIMS The multiple physiological characterizations of exendin-4 make it as a promising drug candidate for the therapy of type 2 diabetes. Although the longer biological half-life offered the exendin-4 with excellent therapeutic potentials for the clinical utility of type 2 diabetes than glucagon-like peptide-1, the exendin-4 still did not free from the inconveniently frequent injections. Therefore, there are increasing requirements for the long-acting exendin-4. METHODS Pp1 regard as a novel exendin-4 protecting peptide, which are predicted to have the ability of increasing the stabilization of exendin-4 in vivo. Protecting peptide is able to form stable complex by non-covalent interaction with human exendin-4. RESULTS In this study, the stability of the exendin-4/Pp1 complex was investigated, and the physiological functions of it were analyzed. Results indicated that exendin-4/Pp1 complex remarkably raised the stabilization of exendin-4 in vivo; it also showed better glucose tolerance and higher HbA(1c) reduction than exendin-4 which was utilized chronically in rodents. CONCLUSION Based upon these results, it is suggested that an exendin-4/Pp1 complex might be utilized as a potent anti-diabetic drug in the treatment of type 2 diabetes mellitus.
Collapse
Affiliation(s)
- Xuemin Zheng
- Department of Pharmaceutical Analysis, Tianjin University of Traditional Chinese Medicine, Tianjin 300193, China
| | | | | | | |
Collapse
|
40
|
Thakur G, Pal K, Mitra A, Mukherjee S, Basak A, Rousseau D. Some Common Antidiabetic Plants of the Indian Subcontinent. FOOD REVIEWS INTERNATIONAL 2010. [DOI: 10.1080/87559129.2010.496024] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
|
41
|
The nucleus tractus solitarius: a portal for visceral afferent signal processing, energy status assessment and integration of their combined effects on food intake. Int J Obes (Lond) 2010; 33 Suppl 1:S11-5. [PMID: 19363500 DOI: 10.1038/ijo.2009.10] [Citation(s) in RCA: 124] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
For humans and animal models alike there is general agreement that the central nervous system processing of gastrointestinal (GI) signals arising from ingested food provides the principal determinant of the size of meals and their frequency. Despite this, relatively few studies are aimed at delineating the brain circuits, neurochemical pathways and intracellular signals that mediate GI-stimulation-induced intake inhibition. Two additional motivations to pursue these circuits and signals have recently arisen. First, the success of gastric-bypass surgery in obesity treatment is highlighting roles for GI signals such as glucagon-like peptide-1 (GLP-1) in intake and energy balance control. Second, accumulating data suggest that the intake-reducing effects of leptin may be mediated through an amplification of the intake-inhibitory effects of GI signals. Experiments reviewed show that: (1) the intake-suppressive effects of a peripherally administered GLP-1 receptor agonist is mediated by caudal brainstem neurons and that forebrain-hypothalamic neural processing is not necessary for this effect; (2) a population of medial nucleus tractus solitarius (NTS) neurons that are responsive to gastric distention is also driven by leptin; (3) caudal brainstem-targeted leptin amplifies the food-intake-inhibitory effects of gastric distention and intestinal nutrient stimulation; (4) adenosine monophosphate-activated protein kinase (AMPK) activity in NTS-enriched brain lysates is elevated by food deprivation and reduced by refeeding and (5) the intake-suppressive effect of hindbrain-directed leptin is reversed by elevating hindbrain AMPK activity. Overall, data support the view that the NTS and circuits within the hindbrain mediate the intake inhibition of GI signals, and that the effects of leptin on food intake result from the amplification of GI signal processing.
Collapse
|
42
|
Samsom M, Bharucha A, Gerich JE, Herrmann K, Limmer J, Linke R, Maggs D, Schirra J, Vella A, Wörle HJ, Göke B. Diabetes mellitus and gastric emptying: questions and issues in clinical practice. Diabetes Metab Res Rev 2009; 25:502-14. [PMID: 19610128 DOI: 10.1002/dmrr.974] [Citation(s) in RCA: 31] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
It is long known that both type 1 and type 2 diabetes can be associated with changes in gastric emptying; a number of publications have linked diabetes to delayed gastric emptying of variable severity and often with poor relationship to gastrointestinal symptomatology. In contrast, more recent studies have reported accelerated gastric emptying when adjusted for glucose concentration in patients with diabetes, indicating a reciprocal relationship between gastric emptying and ambient glucose concentrations. This review proposes that gastroparesis or gastroparesis diabeticorum, a severe condition characterized by a significant impairment of gastric emptying accompanied by severe nausea, vomiting, and malnutrition, is often overdiagnosed and not well contrasted with delays in gastric emptying. The article offers a clinically relevant definition of gastroparesis that should help differentiate this rare condition from (often asymptomatic) delays in gastric emptying. The fact that delayed gastric emptying can also be observed in non-diabetic individuals under experimental conditions in which hyperglycaemia is artificially induced suggests that a delay in gastric emptying rate when blood glucose concentrations are high is actually an appropriate physiological response to hyperglycaemia, slowing further increases in blood glucose. The article discusses the strengths and weaknesses of various methodologies for assessing gastric emptying, especially with respect to the diabetes population, and reviews newer diabetes therapies that decelerate the rate of gastric emptying. These therapies may be a beneficial tool in managing postprandial hyperglycaemia because they attenuate rapid surges in glucose concentrations by slowing the delivery of meal-derived glucose.
Collapse
Affiliation(s)
- Melvin Samsom
- University Medical Center St Radboud, Gastroenterology, Nijmegen, The Netherlands.
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
43
|
Jeong H, Kwon HJ, Kim MK. Hypoglycemic effect of Chlorella vulgaris intake in type 2 diabetic Goto-Kakizaki and normal Wistar rats. Nutr Res Pract 2009; 3:23-30. [PMID: 20016698 PMCID: PMC2788164 DOI: 10.4162/nrp.2009.3.1.23] [Citation(s) in RCA: 29] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2008] [Revised: 01/23/2009] [Accepted: 02/13/2009] [Indexed: 11/04/2022] Open
Abstract
The aim of this study was to examine the hypoglycemic effect of chlorella in 6 week-old type 2 diabetic Goto-Kakizaki (GK, n=30) rats and 6 week-old normal Wistar (n=30) rats. Animals were randomly assigned to 3 groups respectively, and were fed three different experimental diets containing 0%, 3% or 5% (w/w) chlorella for 8 weeks. In diabetic GK rats, the insulinogenic-indices were not significantly different among the groups. The concentrations of fasting plasma glucagon and hepatic triglyceride, and the insulin/glucagon ratios of the GK-3% chlorella and GK-5% chlorella groups were significantly lower than those of the GK-control group. The HOMA-index and the concentrations of fasting blood glucose and plasma insulin of the GK-3% chlorella and GK-5% chlorella groups were slightly lower than those of the GK-control group. In normal Wistar rats, the insulinogenic-indices were not significantly different among the normal groups, but that of the Wistar-5% chlorella group was slightly higher than the other groups. The concentrations of fasting blood glucose and plasma insulin, and the HOMA-index of the Wistar-5% chlorella group were a little higher, and the fasting plasma glucagon concentration and the insulin/glucagon ratio of the Wistar-5% chlorella group were significantly higher than those of the Wistar-control and Wistar-3% chlorella groups. In conclusion, this study shows that the glucose-stimulated insulin secretion was not affected by the intake of chlorella, which could be beneficial, however, in improving insulin sensitivity in type 2 diabetic GK and normal Wistar rats.
Collapse
Affiliation(s)
- Hyejin Jeong
- Department of Nutritional Science and Food Management, Ewha Womans University, 11-1 Daehyeon-dong, Seodaemun-gu, Seoul 120-750, Korea
| | | | | |
Collapse
|
44
|
Lage MJ, Fabunmi R, Boye KS, Misurski DA. Comparison of costs among patients with type 2 diabetes treated with exenatide or sitagliptin therapy. Adv Ther 2009; 26:217-29. [PMID: 19219409 DOI: 10.1007/s12325-009-0002-0] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2008] [Indexed: 10/21/2022]
Abstract
INTRODUCTION Exenatide (Byetta, Amylin Pharmaceuticals Inc., CA, USA) and sitagliptin (Januvia, Merck & Co, NJ, USA) are two antidiabetic agents recently approved by the US Food and Drug Administration. The purpose of this analysis was to compare costs among patients with type 2 diabetes (T2D) treated with either of these agents. METHODS Data with dates of service from September 1, 2005 through August 31, 2007, were obtained from a large US retrospective claims database. Intent-to-treat cohorts of adults diagnosed with T2D who began taking either exenatide (n=1885) or sitagliptin (n=2482) and did not use the alternate medication in the 6-month follow-up period were created. Six-month total medical costs were estimated using stepwise multivariate regressions. Six-month total diabetes-related medical costs, a component of total medical costs, were also estimated using stepwise multivariate regressions. In addition, other cost components were examined using either stepwise multivariate regressions or a two-part model that controlled for the probability of using the medical service. Smearing estimates were used to transform estimated log costs into costs. The analysis controlled for the potential impact of patient demographics, general health, prior resource use, comorbidities, and timing of treatment initiation. RESULTS Exenatide was associated with lower total 6-month direct medical costs ($9340 vs. $9995; P<0.0001), despite some component costs being slightly higher with exenatide: diabetes-related drug costs ($1765 vs. $1743; P=0.0062), diabetes-related medical costs ($4142 vs. $4002; P<0.0001), and emergency room costs ($43 vs. $29; P=0.0388). Exenatide was associated with lower outpatient costs ($4498 vs. $5942; P<0.0001). CONCLUSIONS Compared with the use of sitagliptin, exenatide was associated with lower total medical costs (difference of $655) despite higher total diabetes-related costs (difference of $140). As a result, there appears to be overall cost savings associated with the use of exenatide relative to sitagliptin.
Collapse
|
45
|
Hayes MR, Skibicka KP, Grill HJ. Caudal brainstem processing is sufficient for behavioral, sympathetic, and parasympathetic responses driven by peripheral and hindbrain glucagon-like-peptide-1 receptor stimulation. Endocrinology 2008; 149:4059-68. [PMID: 18420740 PMCID: PMC2488211 DOI: 10.1210/en.2007-1743] [Citation(s) in RCA: 150] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/17/2007] [Accepted: 04/03/2008] [Indexed: 12/11/2022]
Abstract
The effects of peripheral glucagon like peptide-1 receptor (GLP-1R) stimulation on feeding, gastric emptying, and energetic responses involve vagal transmission and central nervous system processing. Despite a lack of studies aimed at determining which central nervous system regions are critical for the GLP-1R response production, hypothalamic/forebrain processing is regarded as essential for these effects. Here the contribution of the caudal brainstem to the control of food intake, core temperature, heart rate, and gastric emptying responses generated by peripheral delivery of the GLP-1R agonist, exendin-4 (Ex-4), was assessed by comparing responses of chronic supracollicular decerebrate (CD) rats to those of pair-fed intact control rats. Responses driven by hindbrain intracerebroventricular (fourth i.c.v) delivery of Ex-4 were also evaluated. Intraperitoneal Ex-4 (1.2 and 3.0 microg/kg) suppressed glucose intake in both CD rats (5.0+/-1.2 and 4.4+/-1.1 ml ingested) and controls (9.4+/-1.5 and 7.7+/-0.8 ml ingested), compared with intakes after vehicle injections (13.1+/-2.5 and 13.2+/-1.7 ml ingested, respectively). Hindbrain ventricular Ex-4 (0.3 microg) also suppressed food intake in CD rats (4.7+/-0.6 ml ingested) and controls (11.0+/-2.9 ml ingested), compared with vehicle intakes (9.3+/-2.1 and 19.3+/-4.3 ml ingested, respectively). Intraperitoneal Ex-4 (0.12, 1.2, 2.4 microg/kg) reduced gastric emptying rates in a dose-related manner similarly for both CD and control rats. Hypothermia followed ip and fourth i.c.v Ex-4 in awake, behaving controls (0.6 and 1.0 C average suppression) and CD rats (1.5 and 2.5 C average suppression). Intraperitoneal Ex-4 triggered tachycardia in both control and CD rats. Results demonstrate that caudal brainstem processing is sufficient for mediating the suppression of intake, core temperature, and gastric emptying rates as well as tachycardia triggered by peripheral GLP-1R activation and also hindbrain-delivered ligand. Contrary to the literature, hypothalamic/forebrain processing and forebrain-caudal brainstem communication is not required for the observed responses.
Collapse
Affiliation(s)
- Matthew R Hayes
- Graduate Groups of Psychology and Neuroscience, University of Pennsylvania, 3720 Walnut Street, Philadelphia, Pennsylvania 19104, USA.
| | | | | |
Collapse
|
46
|
Ceriello A, Colagiuri S, Gerich J, Tuomilehto J. Guideline for management of postmeal glucose. Nutr Metab Cardiovasc Dis 2008; 18:S17-S33. [PMID: 18501571 DOI: 10.1016/j.numecd.2008.01.012] [Citation(s) in RCA: 84] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/14/2008] [Revised: 01/17/2008] [Accepted: 01/31/2008] [Indexed: 02/08/2023]
Abstract
An estimated 246 million people worldwide have diabetes. Diabetes is a leading cause of death in most developed countries, and is reaching epidemic proportions in many developing and newly industrialized nations. Poorly controlled diabetes is associated with the development of renal failure, vision loss, macrovascular diseases and amputations. Large controlled clinical trials have demonstrated that intensive treatment of diabetes can significantly decrease the development and/or progression of microvascular complications of diabetes. There appears to be no glycaemic threshold for reduction of diabetes complications; the lower the glycated haemoglobin (HbA1c), the lower the risk. The progressive relationship between plasma glucose levels and cardiovascular risk extends well below the diabetic threshold. Until recently, the predominant focus of therapy has been on lowering HbA1c levels, with a strong emphasis on fasting plasma glucose. Although control of fasting hyperglycaemia is necessary, it is usually insufficient to obtain optimal glycaemic control. A growing body of evidence suggests that reducing postmeal plasma glucose excursions is as important, or perhaps more important for achieving HbA1c goals. This guideline reviews the evidence on the harmful effects of elevated postmeal glucose and makes recommendations on its treatment, assessment and targets.
Collapse
Affiliation(s)
- Antonio Ceriello
- Warwick Medical School, Clinical Science Research Institute, Clinical Science Building, University Hospital -- Walsgrave Campus, Clifford Bridge Road, Coventry CV2 2DX, UK
| | | | | | | |
Collapse
|
47
|
Giannoukakis N. Ranirestat as a therapeutic aldose reductase inhibitor for diabetic complications. Expert Opin Investig Drugs 2008; 17:575-81. [DOI: 10.1517/13543784.17.4.575] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
|
48
|
Dov A, Abramovitch E, Warwar N, Nesher R. Diminished phosphodiesterase-8B potentiates biphasic insulin response to glucose. Endocrinology 2008; 149:741-8. [PMID: 17991719 DOI: 10.1210/en.2007-0968] [Citation(s) in RCA: 37] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
cAMP activates multiple signal pathways, crucial for the pancreatic beta-cells function and survival and is a major potentiator of insulin release. A family of phosphodiesterases (PDEs) terminate the cAMP signals. We examined the expression of PDEs in rat beta-cells and their role in the regulation of insulin response. Using RT-PCR and Western blot analyses, we identified PDE3A, PDE3B, PDE4B, PDE4D, and PDE8B in rat islets and in INS-1E cells and several possible splice variants of these PDEs. Specific depletion of PDE3A with small interfering (si) RNA (siPDE3A) led to a small (67%) increase in the insulin response to glucose in INS-1E cells but not rat islets. siPDE3A had no effect on the glucagon-like peptide-1 (10 nmol/liter) potentiated insulin response in rat islets. Depletion in PDE8B levels in rat islets using similar technology (siPDE8B) increased insulin response to glucose by 70%, the potentiation being of similar magnitude during the first and second phase insulin release. The siPDE8B-potentiated insulin response was further increased by 23% when glucagon-like peptide-1 was included during the glucose stimulus. In conclusion, PDE8B is expressed in a small number of tissues unrelated to glucose or fat metabolism. We propose that PDE8B, an 3-isobutyl-1-methylxanthine-insensitive cAMP-specific phosphodiesterase, could prove a novel target for enhanced insulin response, affecting a specific pool of cAMP involved in the control of insulin granule trafficking and exocytosis. Finally, we discuss evidence for functional compartmentation of cAMP in pancreatic beta-cells.
Collapse
MESH Headings
- 3',5'-Cyclic-AMP Phosphodiesterases/genetics
- 3',5'-Cyclic-AMP Phosphodiesterases/metabolism
- Animals
- Cell Line, Tumor
- Cyclic Nucleotide Phosphodiesterases, Type 3/genetics
- Cyclic Nucleotide Phosphodiesterases, Type 3/metabolism
- Diabetes Mellitus, Type 2/metabolism
- Diabetes Mellitus, Type 2/physiopathology
- Gene Expression Regulation, Enzymologic/drug effects
- Gene Expression Regulation, Enzymologic/physiology
- Glucagon-Like Peptide 1/metabolism
- Glucagon-Like Peptide 1/pharmacology
- Glucose/metabolism
- Glucose/pharmacology
- Insulin/metabolism
- Insulin Secretion
- Insulin-Secreting Cells/cytology
- Insulin-Secreting Cells/enzymology
- Insulin-Secreting Cells/metabolism
- Insulinoma
- Male
- Pancreatic Neoplasms
- Rats
- Rats, Wistar
Collapse
Affiliation(s)
- Avital Dov
- Endocrinology and Metabolism Service, Department of Medicine, Hadassah, The Hebrew University Medical Center, 91120, Jerusalem, Israel
| | | | | | | |
Collapse
|