1
|
Waterman HL, Moore MC, Smith MS, Farmer B, Yankey K, Scott M, Edgerton DS, Cherrington AD. Morning Engagement of Hepatic Insulin Receptors Improves Afternoon Hepatic Glucose Disposal and Storage. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.09.25.614969. [PMID: 39386695 PMCID: PMC11463395 DOI: 10.1101/2024.09.25.614969] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/12/2024]
Abstract
Glucose tolerance improves significantly upon consuming a second, identical meal later in the day (second meal phenomenon). We previously established that morning hyperinsulinemia primes the liver for increased afternoon hepatic glucose uptake (HGU). Although the route of insulin delivery is an important determinant of the mechanisms by which insulin regulates liver glucose metabolism (direct hepatic vs indirect insulin action), it is not known if insulin's delivery route affects the second meal response. To determine whether morning peripheral insulin delivery (as occurs clinically (subcutaneous)) can enhance afternoon HGU, conscious dogs were treated in the morning with insulin delivered via the portal vein, or peripherally (leg vein), while glucose was infused to maintain euglycemia. Consequently, arterial insulin levels increased similarly in both groups, but relative hepatic insulin deficiency occurred when insulin was delivered peripherally. In the afternoon, all animals were challenged with the same hyperinsulinemic-hyperglycemic clamp to simulate identical postprandial-like conditions. The substantial enhancement of HGU in the afternoon caused by morning portal vein insulin delivery was lost when insulin was delivered peripherally. This indicates that morning insulin does not cause the second meal phenomenon via its indirect actions on the liver, but rather through direct activation of hepatic insulin signaling.
Collapse
Affiliation(s)
- Hannah L Waterman
- Department of Molecular Physiology & Biophysics, Vanderbilt University School of Medicine
| | - Mary Courtney Moore
- Department of Molecular Physiology & Biophysics, Vanderbilt University School of Medicine
| | - Marta S Smith
- Department of Molecular Physiology & Biophysics, Vanderbilt University School of Medicine
| | - Ben Farmer
- Department of Molecular Physiology & Biophysics, Vanderbilt University School of Medicine
| | - Kalisha Yankey
- Department of Molecular Physiology & Biophysics, Vanderbilt University School of Medicine
| | - Melanie Scott
- Department of Molecular Physiology & Biophysics, Vanderbilt University School of Medicine
| | - Dale S Edgerton
- Department of Molecular Physiology & Biophysics, Vanderbilt University School of Medicine
| | - Alan D Cherrington
- Department of Molecular Physiology & Biophysics, Vanderbilt University School of Medicine
| |
Collapse
|
2
|
Yu Z, Yuan Y, Zhang J, Li Y, Wang Z, Wang Y, Duan Y, Zhou Y. Review of the lethal mechanism of insulin poisoning and the characteristic of forensic identification. Leg Med (Tokyo) 2024; 70:102478. [PMID: 38959585 DOI: 10.1016/j.legalmed.2024.102478] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2024] [Revised: 06/10/2024] [Accepted: 06/22/2024] [Indexed: 07/05/2024]
Abstract
Insulin, as the only hypoglycemic hormone in the body, plays a key role in blood sugar control. However, excessive insulin intake can lead to insulin poisoning and even death, which often occurs in clinical and forensic work. At present, some researches on insulin poisoning have been carried out at home and abroad, however, it seems that the mechanism and forensic characteristics of insulin poisoning are not clear and complete. Therefore, in this paper, we reviewed the potential mechanism of insulin poisoning, the methods of insulin detection and the forensic identification of poisoning cases, aiming at providing services for the forensic identification of insulin poisoning.
Collapse
Affiliation(s)
- Zhonghao Yu
- Department of Forensic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China.
| | - Yuhao Yuan
- Department of Forensic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China.
| | - Jiaxin Zhang
- Department of Forensic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China.
| | - Yiling Li
- Department of Forensic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China.
| | - Zhaoxuan Wang
- Department of Forensic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China.
| | - Yubei Wang
- Department of Forensic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China.
| | - Yijie Duan
- Department of Forensic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China.
| | - Yiwu Zhou
- Department of Forensic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China.
| |
Collapse
|
3
|
Wright JJ, Eskaros A, Windon A, Bottino R, Jenkins R, Bradley AM, Aramandla R, Philips S, Kang H, Saunders DC, Brissova M, Powers AC. Exocrine Pancreas in Type 1 and Type 2 Diabetes: Different Patterns of Fibrosis, Metaplasia, Angiopathy, and Adiposity. Diabetes 2024; 73:1140-1152. [PMID: 37881846 PMCID: PMC11189834 DOI: 10.2337/db23-0009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/04/2023] [Accepted: 07/18/2023] [Indexed: 10/27/2023]
Abstract
The endocrine and exocrine compartments of the pancreas are spatially related but functionally distinct. Multiple diseases affect both compartments, including type 1 diabetes (T1D), pancreatitis, cystic fibrosis, and pancreatic cancer. To better understand how the exocrine pancreas changes with age, obesity, and diabetes, we performed a systematic analysis of well-preserved tissue sections from the pancreatic head, body, and tail of organ donors with T1D (n = 20) or type 2 diabetes (T2D) (n = 25) and donors with no diabetes (ND; n = 74). Among ND donors, we found that the incidence of acinar-to-ductal metaplasia (ADM), angiopathy, and pancreatic adiposity increased with age, and ADM and adiposity incidence also increased with BMI. Compared with age- and sex-matched ND organs, T1D pancreata had greater rates of acinar atrophy and angiopathy, with fewer intralobular adipocytes. T2D pancreata had greater rates of ADM and angiopathy and a higher total number of T lymphocytes, but no difference in adipocyte number, compared with ND organs. Although total pancreatic fibrosis was increased in both T1D and T2D, the patterns were different, with periductal and perivascular fibrosis occurring more frequently in T1D pancreata and lobular and parenchymal fibrosis occurring more frequently in T2D. Thus, the exocrine pancreas undergoes distinct changes as individuals age or develop T1D or T2D. ARTICLE HIGHLIGHTS
Collapse
Affiliation(s)
- Jordan J. Wright
- Division of Diabetes, Endocrinology and Metabolism, Department of Medicine, Vanderbilt University Medical Center, Nashville, TN
- Veterans Affairs Tennessee Valley Healthcare System, Nashville, TN
| | - Adel Eskaros
- Division of Diabetes, Endocrinology and Metabolism, Department of Medicine, Vanderbilt University Medical Center, Nashville, TN
| | - Annika Windon
- Department of Pathology, Microbiology and Immunology, Vanderbilt University Medical Center, Nashville, TN
| | - Rita Bottino
- Imagine Islet Center, Imagine Pharma, Pittsburgh, PA
- Institute of Cellular Therapeutics, Allegheny-Singer Research Institute, Allegheny Health Network, Pittsburgh, PA
| | - Regina Jenkins
- Division of Diabetes, Endocrinology and Metabolism, Department of Medicine, Vanderbilt University Medical Center, Nashville, TN
| | - Amber M. Bradley
- Division of Diabetes, Endocrinology and Metabolism, Department of Medicine, Vanderbilt University Medical Center, Nashville, TN
| | - Radhika Aramandla
- Division of Diabetes, Endocrinology and Metabolism, Department of Medicine, Vanderbilt University Medical Center, Nashville, TN
| | - Sharon Philips
- Department of Biostatistics, Vanderbilt University Medical Center, Nashville, TN
| | - Hakmook Kang
- Department of Biostatistics, Vanderbilt University Medical Center, Nashville, TN
| | - Diane C. Saunders
- Division of Diabetes, Endocrinology and Metabolism, Department of Medicine, Vanderbilt University Medical Center, Nashville, TN
- Human Pancreas Analysis Program, Nashville, TN; Philadelphia, PA; and Gainesville, FL
| | - Marcela Brissova
- Division of Diabetes, Endocrinology and Metabolism, Department of Medicine, Vanderbilt University Medical Center, Nashville, TN
- Human Pancreas Analysis Program, Nashville, TN; Philadelphia, PA; and Gainesville, FL
| | - Alvin C. Powers
- Division of Diabetes, Endocrinology and Metabolism, Department of Medicine, Vanderbilt University Medical Center, Nashville, TN
- Veterans Affairs Tennessee Valley Healthcare System, Nashville, TN
- Human Pancreas Analysis Program, Nashville, TN; Philadelphia, PA; and Gainesville, FL
- Department of Molecular Physiology and Biophysics, Vanderbilt University School of Medicine, Nashville, TN
| |
Collapse
|
4
|
Koutny F, Wiemann D, Eckert A, Meyhöfer S, Fritsch M, Pappa A, Wiegand S, Weyer M, Wurm M, Weghuber D, Holl RW. Poorly controlled pediatric type 1 diabetes mellitus is a risk factor for metabolic dysfunction associated steatotic liver disease (MASLD): An observational study. J Pediatr Gastroenterol Nutr 2024; 78:1027-1037. [PMID: 38558281 DOI: 10.1002/jpn3.12194] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/09/2023] [Revised: 01/26/2024] [Accepted: 02/08/2024] [Indexed: 04/04/2024]
Abstract
OBJECTIVES Recent studies have suggested a link between type 1 diabetes mellitus (T1D) and metabolic dysfunction associated steatotic liver disease (MASLD) in children and adolescent, but longitudinal evidence is lacking. This study aimed to investigate the potential association between poorly controlled T1D and elevated alanine aminotransferase (ALT), serving as a proxy for MASLD in children and adolescents over time. METHODS The study included 32,325 children aged 2-17 years with T1D from Germany, Austria, and Switzerland who had undergone at least one assessment of liver enzyme levels recorded in the Diabetes-Patienten- Verlaufsdokumentation registry. Multivariable logistic and Cox regression models were calculated to show possible associations between T1D and elevated ALT values (>26 U/L in males, >22 U/L in females) as a proxy for MASLD. RESULTS Children with poorly controlled T1D (HbA1c > 11%) exhibited increased odds of elevated ALT values, after adjustment for age, sex, diabetes duration and overweight (odds ratio [OR] 2.54; 95% confidence interval [CI], 2.10-3.10; p < 0.01). This finding is substantiated by a longitudinal analysis, which reveals that inadequately controlled T1D was associated with a higher hazard ratio (HR) of elevated ALT values compared to children with controlled T1D over an observation period extending up to 5.5 (HR: 1.54; 95% CI, 1.19-2.01; p < 0.01). CONCLUSION In conclusion, the current study strongly links poorly controlled T1D in children and adolescents to MASLD irrespective of overweight. This association is not only present cross-sectionally but also increases over time. The study underscores the critical role of effective diabetes management in reducing the risk of MASLD in this population.
Collapse
Affiliation(s)
- Florian Koutny
- Department of Human Medicine, PhD Medical Science, Paracelsus Medical University, Salzburg, Austria
- Department of Internal Medicine 2, Gastroenterology and Hepatology and Rheumatology, Karl Landsteiner University of Health Sciences, University Hospital of St. Pölten, St. Pölten, Austria
| | - Dagobert Wiemann
- Department of Pediatrics, University of Magdeburg, Magdeburg, Germany
| | - Alexander Eckert
- Institute of Epidemiology and Medical Biometry, ZIBMT, University of Ulm, Germany, and German Center for Diabetes Research (DZD), Munich-Neuherberg, Germany
| | - Svenja Meyhöfer
- Institute for Endocrinology & Diabetes, University of Lübeck, Lübeck, Germany
- German Center for Diabetes Research (DZD), München-Neuherberg, Germany
- Department of Internal Medicine 1, Endocrinology & Diabetes, University of Lübeck, Lübeck, Germany
| | - Maria Fritsch
- Department of Pediatrics, Medical University of Graz, Austria
| | - Angeliki Pappa
- Department of Pediatric and Adolescent Medicine, University Hospital RWTH Aachen, Aachen, Germany
| | - Susanna Wiegand
- Department of Pediatric Endocrinology and Diabetes, Center for social-pediatric care, Charité, Germany
| | - Marc Weyer
- Kamillus-Klinik Internal Medicine, Asbach, Germany
| | - Michael Wurm
- Department of Paediatrics, St. Hedwigs Campus, University Children's Hospital Regensburg, Regensburg, Germany
| | - Daniel Weghuber
- Department of Human Medicine, PhD Medical Science, Paracelsus Medical University, Salzburg, Austria
| | - Reinhard W Holl
- Institute of Epidemiology and Medical Biometry, ZIBMT, University of Ulm, Germany, and German Center for Diabetes Research (DZD), Munich-Neuherberg, Germany
| |
Collapse
|
5
|
Estes SK, Shiota C, O'Brien TP, Printz RL, Shiota M. The impact of glucagon to support postabsorptive glucose flux and glycemia in healthy rats and its attenuation in male Zucker diabetic fatty rats. Am J Physiol Endocrinol Metab 2024; 326:E308-E325. [PMID: 38265288 PMCID: PMC11193518 DOI: 10.1152/ajpendo.00192.2023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/26/2023] [Revised: 01/19/2024] [Accepted: 01/19/2024] [Indexed: 01/25/2024]
Abstract
Hyperglucagonemia is a hallmark of type 2 diabetes (T2DM), yet the role of elevated plasma glucagon (P-GCG) to promote excessive postabsorptive glucose production and contribute to hyperglycemia in patients with this disease remains debatable. We investigated the acute action of P-GCG to safeguard/support postabsorptive endogenous glucose production (EGP) and euglycemia in healthy Zucker control lean (ZCL) rats. Using male Zucker diabetic fatty (ZDF) rats that exhibit the typical metabolic disorders of human T2DM, such as excessive EGP, hyperglycemia, hyperinsulinemia, and hyperglucagonemia, we examined the ability of hyperglucagonemia to promote greater rates of postabsorptive EGP and hyperglycemia. Euglycemic or hyperglycemic basal insulin (INS-BC) and glucagon (GCG-BC) clamps were performed in the absence or during an acute setting of glucagon deficiency (GCG-DF, ∼10% of basal), either alone or in combination with insulin deficiency (INS-DF, ∼10% of basal). Glucose appearance, disappearance, and cycling rates were measured using [2-3H] and [3-3H]-glucose. In ZCL rats, GCG-DF reduced the levels of hepatic cyclic AMP, EGP, and plasma glucose (PG) by 50%, 32%, and 50%, respectively. EGP fell in the presence GCG-DF and INS-BC, but under GCG-DF and INS-DF, EGP and PG increased two- and threefold, respectively. GCG-DF revealed the hyperglucagonemia present in ZDF rats lacked the ability to regulate hepatic intracellular cyclic AMP levels and glucose flux, since EGP and PG levels fell by only 10%. We conclude that the liver in T2DM suffers from resistance to all three major regulatory factors, glucagon, insulin, and glucose, thus leading to a loss of metabolic flexibility.NEW & NOTEWORTHY In postabsorptive state, basal plasma insulin (P-INS) and plasma glucose (PG) act dominantly to increase hepatic glucose cycling and reduce endogenous glucose production (EGP) and PG in healthy rats, which is only counteracted by the acute action of basal plasma glucagon (P-GCG) to support EGP and euglycemia. Hyperglucagonemia, a hallmark of type 2 diabetes (T2DM) present in Zucker diabetic fatty (ZDF) rats, is not the primary mediator of hyperglycemia and high EGP as commonly thought; instead, the liver is resistant to glucagon as well as insulin and glucose.
Collapse
Affiliation(s)
- Shanea K Estes
- Department of Molecular Physiology and Biophysics, Vanderbilt University School of Medicine, Nashville, Tennessee, United States
| | - Chiyo Shiota
- Department of Molecular Physiology and Biophysics, Vanderbilt University School of Medicine, Nashville, Tennessee, United States
| | - Tracy P O'Brien
- Department of Molecular Physiology and Biophysics, Vanderbilt University School of Medicine, Nashville, Tennessee, United States
| | - Richard L Printz
- Department of Medicine, Vanderbilt University School of Medicine, Nashville, Tennessee, United States
| | - Masakazu Shiota
- Department of Molecular Physiology and Biophysics, Vanderbilt University School of Medicine, Nashville, Tennessee, United States
| |
Collapse
|
6
|
Basset-Sagarminaga J, van de Weijer T, Iozzo P, Schrauwen P, Schrauwen-Hinderling V. Advances and challenges in measuring hepatic glucose uptake with FDG PET: implications for diabetes research. Diabetologia 2024; 67:407-419. [PMID: 38099962 DOI: 10.1007/s00125-023-06055-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/02/2023] [Accepted: 10/10/2023] [Indexed: 02/06/2024]
Abstract
The liver plays a crucial role in the control of glucose homeostasis and is therefore of great interest in the investigation of the development of type 2 diabetes. Hepatic glucose uptake (HGU) can be measured through positron emission tomography (PET) imaging with the tracer [18F]-2-fluoro-2-deoxy-D-glucose (FDG). HGU is dependent on many variables (e.g. plasma glucose, insulin and glucagon concentrations), and the metabolic state for HGU assessment should be chosen with care and coherence with the study question. In addition, as HGU is influenced by many factors, protocols and measurement conditions need to be standardised for reproducible results. This review provides insights into the protocols that are available for the measurement of HGU by FDG PET and discusses the current state of knowledge of HGU and its impairment in type 2 diabetes. Overall, a scanning modality that allows for the measurement of detailed kinetic information and influx rates (dynamic imaging) may be preferable to static imaging. The combination of FDG PET and insulin stimulation is crucial to measure tissue-specific insulin sensitivity. While the hyperinsulinaemic-euglycaemic clamp allows for standardised measurements under controlled blood glucose levels, some research questions might require a more physiological approach, such as oral glucose loading, with both advantages and complexities relating to fluctuations in blood glucose and insulin levels. The available approaches to address HGU hold great potential but await more systematic exploitation to improve our understanding of the mechanisms underlying metabolic diseases. Current findings from the investigation of HGU by FDG PET highlight the complex interplay between insulin resistance, hepatic glucose metabolism, NEFA levels and intrahepatic lipid accumulation in type 2 diabetes and obesity. Further research is needed to fully understand the underlying mechanisms and potential therapeutic targets for improving HGU in these conditions.
Collapse
Affiliation(s)
- Jeremy Basset-Sagarminaga
- Department of Nutrition and Movement Sciences, School of Nutrition and Translational Research in Metabolism (NUTRIM), Maastricht University, Maastricht, the Netherlands
| | - Tineke van de Weijer
- Department of Nutrition and Movement Sciences, School of Nutrition and Translational Research in Metabolism (NUTRIM), Maastricht University, Maastricht, the Netherlands
- Department of Radiology and Nuclear Medicine, Maastricht University Medical Center, Maastricht, the Netherlands
| | - Patricia Iozzo
- Institute of Clinical Physiology, National Research Council (CNR), Pisa, Italy
| | - Patrick Schrauwen
- Department of Nutrition and Movement Sciences, School of Nutrition and Translational Research in Metabolism (NUTRIM), Maastricht University, Maastricht, the Netherlands
| | - Vera Schrauwen-Hinderling
- Department of Nutrition and Movement Sciences, School of Nutrition and Translational Research in Metabolism (NUTRIM), Maastricht University, Maastricht, the Netherlands.
- Department of Radiology and Nuclear Medicine, Maastricht University Medical Center, Maastricht, the Netherlands.
- Institute for Clinical Diabetology, German Diabetes Center, Leibniz Institute for Diabetes Research at Heinrich Heine University, Düsseldorf, Germany.
| |
Collapse
|
7
|
Ziebarth J, Diedrich C, Schineider Machado C, Mara Mainardes R. Optimized LC-MS/MS method for quantifying insulin degludec and liraglutide in rat plasma and Tissues: Application in pharmacokinetics and biodistribution. J Chromatogr B Analyt Technol Biomed Life Sci 2024; 1234:124015. [PMID: 38266609 DOI: 10.1016/j.jchromb.2024.124015] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2023] [Revised: 01/05/2024] [Accepted: 01/15/2024] [Indexed: 01/26/2024]
Abstract
We present an ultra-performance liquid chromatography-tandem mass spectrometry (LC-MS/MS) method for simultaneous detection of insulin degludec (I-Deg) and liraglutide (LIRA) in rat plasma and tissues, characterized by its sensitivity and selectivity. Chromatographic separation was achieved using an Acquity UPLC BEH C18 column, leveraging a mobile phase of acetonitrile and water (both with 0.1 % formic acid) under gradient elution over a run time of 7.5 min. The mass spectrometer operated in positive electrospray ionization multiple reaction monitoring (MRM) mode, tracking transitions of m/z 1221.6 → 641.6 for I-Deg, m/z 938.7 → 1064.1 for LIRA, and m/z 1184.7 → 454.4 for the internal standard. Validation ranged from 5 to 100 ng/mL, exhibiting robust linearity (r2 > 0.99) and limits of detection (LOD) of 1.63-2.02 ng/mL for I-Deg and 0.96-1.62 ng/mL for LIRA. Limits of quantification (LOQ) were 2.38-4.76 ng/mL for I-Deg and 3.22-4.40 ng/mL for LIRA. Notably, no significant matrix effects were detected. Stability was confirmed under various conditions, and precision metrics (intraday RSD 1.68-8.05 % for I-Deg and 1.11-7.69 % for LIRA; interday RSD 1.39-8.61 % for I-Deg and 1.06-8.83 % for LIRA) alongside accuracy (90.5-114.9 % for I-Deg and 92.7-113.7 % for LIRA) were within acceptable ranges. The method was successfully applied to pharmacokinetic and biodistribution studies following simultaneous subcutaneous administration of LIRA and I-Deg in rats.
Collapse
Affiliation(s)
- Jeferson Ziebarth
- Laboratory of Nanostructured Formulations, Universidade Estadual do Centro-Oeste, Guarapuava, Brazil
| | - Camila Diedrich
- Laboratory of Nanostructured Formulations, Universidade Estadual do Centro-Oeste, Guarapuava, Brazil
| | | | - Rubiana Mara Mainardes
- Laboratory of Nanostructured Formulations, Universidade Estadual do Centro-Oeste, Guarapuava, Brazil; Department of Pharmacy, Universidade Estadual do Centro-Oeste, Guarapuava, Brazil.
| |
Collapse
|
8
|
Kurtzhals P, Østergaard S, Nishimura E, Kjeldsen T. Derivatization with fatty acids in peptide and protein drug discovery. Nat Rev Drug Discov 2023; 22:59-80. [PMID: 36002588 DOI: 10.1038/s41573-022-00529-w] [Citation(s) in RCA: 54] [Impact Index Per Article: 27.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/29/2022] [Indexed: 01/28/2023]
Abstract
Peptides and proteins are widely used to treat a range of medical conditions; however, they often have to be injected and their effects are short-lived. These shortcomings of the native structure can be addressed by molecular engineering, but this is a complex undertaking. A molecular engineering technology initially applied to insulin - and which has now been successfully applied to several biopharmaceuticals - entails the derivatization of peptides and proteins with fatty acids. Various protraction mechanisms are enabled by the specific characteristics and positions of the attached fatty acid. Furthermore, the technology can ensure a long half-life following oral administration of peptide drugs, can alter the distribution of peptides and may hold potential for tissue targeting. Due to the inherent safety and well-defined chemical nature of the fatty acids, this technology provides a versatile approach to peptide and protein drug discovery.
Collapse
|
9
|
Kishor Sharma J, Sharma D, Gupta A. A glorious past, dynamic present and a promising future: Insulin at 100. J R Coll Physicians Edinb 2022; 52:59-64. [DOI: 10.1177/14782715221088981] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022] Open
Abstract
It has been 100 years since insulin was discovered. Insulin therapy remains the cornerstone for the management of diabetes. Advances in human physiology, pathology, molecular biology, biotechnology, biomedical engineering and devices have added tremendously to the journey of one of the greatest discoveries of modern medicine. Epidemiology of diabetes has gone through a major shift in a century; many oral drugs have been introduced for the management of type 2 diabetes; glucose monitoring has also gone through a sea change with the availability of novel parameters like continuous glucose monitoring, as has insulin delivery, with the availability of insulin pumps and the artificial pancreas. In this article, we look into historical facts, challenges, limitations and future developments of insulin therapy.
Collapse
Affiliation(s)
| | | | - Amit Gupta
- Centre for Diabetes Care, Greater Noida, India
| |
Collapse
|
10
|
Physiologic Insulin Resensitization as a Treatment Modality for Insulin Resistance Pathophysiology. Int J Mol Sci 2022; 23:ijms23031884. [PMID: 35163806 PMCID: PMC8836751 DOI: 10.3390/ijms23031884] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2021] [Revised: 01/28/2022] [Accepted: 02/03/2022] [Indexed: 12/10/2022] Open
Abstract
Prevalence of type 2 diabetes increased from 2.5% of the US population in 1990 to 10.5% in 2018. This creates a major public health problem, due to increases in long-term complications of diabetes, including neuropathy, retinopathy, nephropathy, skin ulcers, amputations, and atherosclerotic cardiovascular disease. In this review, we evaluated the scientific basis that supports the use of physiologic insulin resensitization. Insulin resistance is the primary cause of type 2 diabetes. Insulin resistance leads to increasing insulin secretion, leading to beta-cell exhaustion or burnout. This triggers a cascade leading to islet cell destruction and the long-term complications of type 2 diabetes. Concurrent with insulin resistance, the regular bursts of insulin from the pancreas become irregular. This has been treated by the precise administration of insulin more physiologically. There is consistent evidence that this treatment modality can reverse the diabetes-associated complications of neuropathy, diabetic ulcers, nephropathy, and retinopathy, and that it lowers HbA1c. In conclusion, physiologic insulin resensitization has a persuasive scientific basis, significant treatment potential, and likely cost benefits.
Collapse
|
11
|
Retnakaran R, Zinman B. The ongoing evolution of basal insulin therapy over 100 years and its promise for the future. Diabetes Obes Metab 2022; 24 Suppl 1:17-26. [PMID: 34532950 DOI: 10.1111/dom.14552] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/29/2021] [Revised: 09/03/2021] [Accepted: 09/12/2021] [Indexed: 10/20/2022]
Abstract
The evolution of basal insulin therapy over the past 100 years since the discovery of insulin is a testimony to the biomedical bench-to-bedside process, wherein incremental advances in the basic sciences are progressively translated over time into a series of enhancements in clinical care, each building upon the success of its predecessors. The emergence of recombinant DNA technology and the resultant biosynthesis of human insulin in the 1980s provided the critical capacity to bioengineer designer insulin analogues with pharmacokinetic and pharmacodynamic properties that can better mimic, although not fully replicate, the effects of endogenous insulin secretion. Through these efforts, basal insulin therapy has progressed over this time from first-generation analogues (glargine U-100, detemir) to second-generation analogues (glargine U-300, degludec) to ultra-long-acting formulations that are suitable for administration once weekly (icodec). Each iteration in this progression has represented a step closer towards the goal of replicating the continuous secretion of insulin that normally comprises the basal output of the pancreatic beta-cells between meals, during episodes of fasting and overnight. However, it may be that we may have reached the achievable limit in the context of an "open-loop" approach, such that only with the addition of closed loop control will we be able to achieve physiologic basal insulin replacement. In this review, we will examine the evolution of basal insulin therapy over the past 100 years and its implications for patient care and outcomes in current practice and the future.
Collapse
Affiliation(s)
- Ravi Retnakaran
- Leadership Sinai Centre for Diabetes, Mount Sinai Hospital, Toronto, Ontario, Canada
- Division of Endocrinology, University of Toronto, Toronto, Ontario, Canada
- Lunenfeld-Tanenbaum Research Institute, Mount Sinai Hospital, Toronto, Ontario, Canada
| | - Bernard Zinman
- Leadership Sinai Centre for Diabetes, Mount Sinai Hospital, Toronto, Ontario, Canada
- Division of Endocrinology, University of Toronto, Toronto, Ontario, Canada
- Lunenfeld-Tanenbaum Research Institute, Mount Sinai Hospital, Toronto, Ontario, Canada
| |
Collapse
|
12
|
Lee SH, Yoon KH. A Century of Progress in Diabetes Care with Insulin: A History of Innovations and Foundation for the Future. Diabetes Metab J 2021; 45:629-640. [PMID: 34610718 PMCID: PMC8497924 DOI: 10.4093/dmj.2021.0163] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/15/2021] [Accepted: 08/08/2021] [Indexed: 12/15/2022] Open
Abstract
The year 2021 marks the 100th anniversary of the discovery of insulin, which has greatly changed the lives of people with diabetes and become a cornerstone of advances in medical science. A rapid bench-to-bedside application of the lifesaving pancreatic extract and its immediate commercialization was the result of a promising idea, positive drive, perseverance, and collaboration of Banting and colleagues. As one of the very few proteins isolated in a pure form at that time, insulin also played a key role in the development of important methodologies and in the beginning of various fields of modern science. Since its discovery, insulin has evolved continuously to optimize the care of people with diabetes. Since the 1980s, recombinant DNA technology has been employed to engineer insulin analogs by modifying their amino acid sequence, which has resulted in the production of insulins with various profiles that are currently used. However, unmet needs in insulin treatment still exist, and several forms of future insulins are under development. In this review, we discuss the past, present, and future of insulin, including a history of ceaseless innovations and collective intelligence. We believe that this story will be a solid foundation and an unerring guide for the future.
Collapse
Affiliation(s)
- Seung-Hwan Lee
- Division of Endocrinology and Metabolism, Department of Internal Medicine, Seoul St. Mary’s Hospital, College of Medicine, The Catholic University of Korea, Seoul, Korea
- Department of Medical Informatics, College of Medicine, The Catholic University of Korea, Seoul, Korea
| | - Kun-Ho Yoon
- Division of Endocrinology and Metabolism, Department of Internal Medicine, Seoul St. Mary’s Hospital, College of Medicine, The Catholic University of Korea, Seoul, Korea
- Department of Medical Informatics, College of Medicine, The Catholic University of Korea, Seoul, Korea
- Corresponding author: Kun-Ho Yoon, https://orcid.org/0000-0002-9109-2208, Division of Endocrinology and Metabolism, Department of Internal Medicine, Seoul St. Mary’s Hospital, College of Medicine, The Catholic University of Korea, 222 Banpodaero, Seocho-gu, Seoul 06591, Korea E-mail:
| |
Collapse
|
13
|
Scherer T, Sakamoto K, Buettner C. Brain insulin signalling in metabolic homeostasis and disease. Nat Rev Endocrinol 2021; 17:468-483. [PMID: 34108679 DOI: 10.1038/s41574-021-00498-x] [Citation(s) in RCA: 80] [Impact Index Per Article: 20.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 04/22/2021] [Indexed: 02/06/2023]
Abstract
Insulin signalling in the central nervous system regulates energy homeostasis by controlling metabolism in several organs and by coordinating organ crosstalk. Studies performed in rodents, non-human primates and humans over more than five decades using intracerebroventricular, direct hypothalamic or intranasal application of insulin provide evidence that brain insulin action might reduce food intake and, more importantly, regulates energy homeostasis by orchestrating nutrient partitioning. This Review discusses the metabolic pathways that are under the control of brain insulin action and explains how brain insulin resistance contributes to metabolic disease in obesity, the metabolic syndrome and type 2 diabetes mellitus.
Collapse
Affiliation(s)
- Thomas Scherer
- Division of Endocrinology and Metabolism, Department of Medicine III, Medical University of Vienna, Vienna, Austria.
| | - Kenichi Sakamoto
- Division of Endocrinology, Metabolism & Nutrition, Department of Medicine, Rutgers Robert Wood Johnson Medical School, New Brunswick, NJ, USA
| | - Christoph Buettner
- Division of Endocrinology, Metabolism & Nutrition, Department of Medicine, Rutgers Robert Wood Johnson Medical School, New Brunswick, NJ, USA.
| |
Collapse
|
14
|
Kurtzhals P, Nishimura E, Haahr H, Høeg-Jensen T, Johansson E, Madsen P, Sturis J, Kjeldsen T. Commemorating insulin's centennial: engineering insulin pharmacology towards physiology. Trends Pharmacol Sci 2021; 42:620-639. [PMID: 34148677 DOI: 10.1016/j.tips.2021.05.005] [Citation(s) in RCA: 29] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2021] [Revised: 05/13/2021] [Accepted: 05/18/2021] [Indexed: 01/14/2023]
Abstract
The life-saving discovery of insulin in Toronto in 1921 is one of the most impactful achievements in medical history, at the time being hailed as a miracle treatment for diabetes. The insulin molecule itself, however, is poorly amenable as a pharmacological intervention, and the formidable challenge of optimizing insulin therapy has been ongoing for a century. We review early academic insights into insulin structure and its relation to self-association and receptor binding, as well as recombinant biotechnology, which have all been seminal for drug design. Recent developments have focused on combining genetic and chemical engineering with pharmaceutical optimization to generate ultra-rapid and ultra-long-acting, tissue-selective, or orally delivered insulin analogs. We further discuss these developments and propose that future scientific efforts in molecular engineering include realizing the dream of glucose-responsive insulin delivery.
Collapse
Affiliation(s)
- Peter Kurtzhals
- Research and Development, Novo Nordisk A/S, Novo Allé, DK-2880 Bagsværd, Denmark.
| | - Erica Nishimura
- Research and Development, Novo Nordisk A/S, Novo Allé, DK-2880 Bagsværd, Denmark
| | - Hanne Haahr
- Research and Development, Novo Nordisk A/S, Novo Allé, DK-2880 Bagsværd, Denmark
| | - Thomas Høeg-Jensen
- Research and Development, Novo Nordisk A/S, Novo Allé, DK-2880 Bagsværd, Denmark
| | - Eva Johansson
- Research and Development, Novo Nordisk A/S, Novo Allé, DK-2880 Bagsværd, Denmark
| | - Peter Madsen
- Research and Development, Novo Nordisk A/S, Novo Allé, DK-2880 Bagsværd, Denmark
| | - Jeppe Sturis
- Research and Development, Novo Nordisk A/S, Novo Allé, DK-2880 Bagsværd, Denmark
| | - Thomas Kjeldsen
- Research and Development, Novo Nordisk A/S, Novo Allé, DK-2880 Bagsværd, Denmark
| |
Collapse
|
15
|
Kraft G, Coate KC, Smith M, Farmer B, Scott M, Cherrington AD, Edgerton DS. The Importance of the Mechanisms by Which Insulin Regulates Meal-Associated Liver Glucose Uptake in the Dog. Diabetes 2021; 70:1292-1302. [PMID: 33757993 PMCID: PMC8275892 DOI: 10.2337/db20-1271] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/18/2020] [Accepted: 03/18/2021] [Indexed: 12/17/2022]
Abstract
Hepatic glucose uptake (HGU) is critical for maintaining normal postprandial glucose metabolism. Insulin is clearly a key regulator of HGU, but the physiologic mechanisms by which it acts have yet to be established. This study sought to determine the mechanisms by which insulin regulates liver glucose uptake under postprandial-like conditions (hyperinsulinemia, hyperglycemia, and a positive portal vein-to-arterial glucose gradient). Portal vein insulin infusion increased hepatic insulin levels fivefold in healthy dogs. In one group (n = 7), the physiologic response was allowed to fully occur, while in another (n = 7), insulin's indirect hepatic effects, occurring secondary to its actions on adipose tissue, pancreas, and brain, were blocked. This was accomplished by infusing triglyceride (intravenous), glucagon (portal vein), and inhibitors of brain insulin action (intracerebroventricular) to prevent decreases in plasma free fatty acids or glucagon, while blocking increased hypothalamic insulin signaling for 4 h. In contrast to the indirect hepatic effects of insulin, which were previously shown capable of independently generating a half-maximal stimulation of HGU, direct hepatic insulin action was by itself able to fully stimulate HGU. This suggests that under hyperinsulinemic/hyperglycemic conditions insulin's indirect effects are redundant to direct engagement of hepatocyte insulin receptors.
Collapse
Affiliation(s)
- Guillaume Kraft
- Department of Molecular Physiology and Biophysics, Vanderbilt University School of Medicine, Nashville, TN
| | - Katie C Coate
- Department of Molecular Physiology and Biophysics, Vanderbilt University School of Medicine, Nashville, TN
| | - Marta Smith
- Department of Molecular Physiology and Biophysics, Vanderbilt University School of Medicine, Nashville, TN
| | - Ben Farmer
- Department of Molecular Physiology and Biophysics, Vanderbilt University School of Medicine, Nashville, TN
| | - Melanie Scott
- Department of Molecular Physiology and Biophysics, Vanderbilt University School of Medicine, Nashville, TN
| | - Alan D Cherrington
- Department of Molecular Physiology and Biophysics, Vanderbilt University School of Medicine, Nashville, TN
| | - Dale S Edgerton
- Department of Molecular Physiology and Biophysics, Vanderbilt University School of Medicine, Nashville, TN
| |
Collapse
|
16
|
Abstract
Although insulin therapy was already introduced one-hundred years ago, insulin formulations are still being refined to reduce the risk of hypoglycaemia and of other insulin side effects such as weight gain. This review summarises the available clinical data for some ongoing developments of new insulins and evaluates their potential for future insulin therapy. Once-weekly insulins will most likely be the next addition to the insulin armamentarium. First clinical studies indicate low peak-to-trough fluctuations with these insulins indicating the potential to achieve better glycaemic control or reduce hypoglycaemic events versus available basal insulins. Proof-of-concept has also been established for hepato-preferential and oral insulins; however, adverse effects and low bioavailability still need to be overcome. It will take much longer, before glucose-responsive "smart" insulins will be available. A first clinical study and numerous pre-clinical data show the potential, but also the challenges of designing an insulin that quickly reacts to blood glucose changes and prevents hypoglycaemia and pronounced hyperglycaemia. Nevertheless, it is reassuring that the search for better insulins has never stopped since its first use one-hundred years ago and is still ongoing. New developments have a high potential of further improving the safety and efficacy of insulin therapy in the future.
Collapse
|
17
|
Bottinelli C, Bévalot F, Cartiser N, Fanton L, Guitton J. Detection of insulins in postmortem tissues: an optimized workflow based on immunopurification and LC-MS/HRMS detection. Int J Legal Med 2021; 135:1813-1822. [PMID: 33932171 DOI: 10.1007/s00414-021-02598-9] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2020] [Accepted: 04/05/2021] [Indexed: 12/22/2022]
Abstract
Diabetes is a worldwide disease in perpetual expansion. Type 1 and sometimes type 2 diabetic patients require daily human insulin (HI) or analog administration. Easy access to insulins for insulin-treated diabetics, their relatives, and medical professionals can enable abuse for suicidal or homicidal purpose. However, demonstrating insulin overdose in postmortem blood is challenging. Tissue analyses are contributive, as insulins can accumulate before death or undergo only limited degradation. The present study describes an assay for HI and synthetic analogs (lispro, aspart, glulisine, detemir and degludec, glargine and its main metabolite (M1)) in liver, kidney, muscle, and injection site samples. It is based on a 5-step sample preparation (reduction of tissue sample size, homogenization, extraction, concentration, and immunopurification) associated with liquid chromatography coupled to high-resolution mass spectrometry (LC-MS/HRMS). Selectivity and limit of detection (LOD) for all target analogs were assessed in the above matrices. LOD was determined at 25 ng/g for HI and for analogs except detemir and degludec, where LOD was 50 ng/g in kidney and injection site samples and 80 ng/g in the liver and muscle. The method was applied to13 forensic cases in which insulin use was suspected.
Collapse
Affiliation(s)
- Charline Bottinelli
- LAT LUMTOX Laboratory, 32 Rue du 35ème Régiment d'Aviation 69500, Bron, France.
| | - Fabien Bévalot
- LAT LUMTOX Laboratory, 32 Rue du 35ème Régiment d'Aviation 69500, Bron, France
| | - Nathalie Cartiser
- Department of Forensic Medicine, Edouard Herriot Hospital, Hospices Civils de Lyon, Lyon, France
| | - Laurent Fanton
- Department of Forensic Medicine, Edouard Herriot Hospital, Hospices Civils de Lyon, Lyon, France.,Faculty of Medicine Lyon-Est, University of Lyon, UCBL1, Lyon, France
| | - Jérôme Guitton
- Toxicology Laboratory, ISPB Faculty of Pharmacy, University of Lyon, UCBL1, Lyon, France.,Pharmacology-Toxicology Laboratory, Lyon-Sud Hospital, Hospices Civils de Lyon, Lyon, France
| |
Collapse
|
18
|
Edgerton DS, Moore MC, Gregory JM, Kraft G, Cherrington AD. Importance of the route of insulin delivery to its control of glucose metabolism. Am J Physiol Endocrinol Metab 2021; 320:E891-E897. [PMID: 33813879 PMCID: PMC8238128 DOI: 10.1152/ajpendo.00628.2020] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/14/2020] [Revised: 03/24/2021] [Accepted: 03/26/2021] [Indexed: 12/12/2022]
Abstract
Pancreatic insulin secretion produces an insulin gradient at the liver compared with the rest of the body (approximately 3:1). This physiological distribution is lost when insulin is injected subcutaneously, causing impaired regulation of hepatic glucose production and whole body glucose uptake, as well as arterial hyperinsulinemia. Thus, the hepatoportal insulin gradient is essential to the normal control of glucose metabolism during both fasting and feeding. Insulin can regulate hepatic glucose production and uptake through multiple mechanisms, but its direct effects on the liver are dominant under physiological conditions. Given the complications associated with iatrogenic hyperinsulinemia in patients treated with insulin, insulin designed to preferentially target the liver may have therapeutic advantages.
Collapse
Affiliation(s)
- Dale S Edgerton
- Department of Molecular Physiology and Biophysics, Vanderbilt University School of Medicine, Nashville, Tennessee
| | - Mary C Moore
- Department of Molecular Physiology and Biophysics, Vanderbilt University School of Medicine, Nashville, Tennessee
| | - Justin M Gregory
- Department of Molecular Physiology and Biophysics, Vanderbilt University School of Medicine, Nashville, Tennessee
| | - Guillaume Kraft
- Department of Molecular Physiology and Biophysics, Vanderbilt University School of Medicine, Nashville, Tennessee
| | - Alan D Cherrington
- Department of Molecular Physiology and Biophysics, Vanderbilt University School of Medicine, Nashville, Tennessee
| |
Collapse
|
19
|
Lewis GF, Carpentier AC, Pereira S, Hahn M, Giacca A. Direct and indirect control of hepatic glucose production by insulin. Cell Metab 2021; 33:709-720. [PMID: 33765416 DOI: 10.1016/j.cmet.2021.03.007] [Citation(s) in RCA: 58] [Impact Index Per Article: 14.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/31/2021] [Revised: 02/23/2021] [Accepted: 03/05/2021] [Indexed: 01/08/2023]
Abstract
There is general agreement that the acute suppression of hepatic glucose production by insulin is mediated by both a direct and an indirect effect on the liver. There is, however, no consensus regarding the relative magnitude of these effects under physiological conditions. Extensive research over the past three decades in humans and animal models has provided discordant results between these two modes of insulin action. Here, we review the field to make the case that physiologically direct hepatic insulin action dominates acute suppression of glucose production, but that there is also a delayed, second order regulation of this process via extrahepatic effects. We further provide our views regarding the timing, dominance, and physiological relevance of these effects and discuss novel concepts regarding insulin regulation of adipose tissue fatty acid metabolism and central nervous system (CNS) signaling to the liver, as regulators of insulin's extrahepatic effects on glucose production.
Collapse
Affiliation(s)
- Gary F Lewis
- Departments of Medicine and Physiology, University of Toronto, Toronto, ON, Canada; Banting & Best Diabetes Centre, University of Toronto, Toronto, ON, Canada; Institute of Medical Sciences, University of Toronto, Toronto, ON, Canada.
| | - Andre C Carpentier
- Division of Endocrinology, Department of Medicine, Centre de Recherche du CHUS, Université de Sherbrooke, Sherbrooke, QC, Canada
| | - Sandra Pereira
- Centre for Addiction and Mental Health and Department of Physiology, University of Toronto, Toronto, ON, Canada
| | - Margaret Hahn
- Banting & Best Diabetes Centre, University of Toronto, Toronto, ON, Canada; Institute of Medical Sciences, University of Toronto, Toronto, ON, Canada; Centre for Addiction and Mental Health and Department of Psychiatry, University of Toronto, Toronto, ON, Canada
| | - Adria Giacca
- Departments of Medicine and Physiology, University of Toronto, Toronto, ON, Canada; Banting & Best Diabetes Centre, University of Toronto, Toronto, ON, Canada; Institute of Medical Sciences, University of Toronto, Toronto, ON, Canada
| |
Collapse
|
20
|
Dimitriadis GD, Maratou E, Kountouri A, Board M, Lambadiari V. Regulation of Postabsorptive and Postprandial Glucose Metabolism by Insulin-Dependent and Insulin-Independent Mechanisms: An Integrative Approach. Nutrients 2021; 13:E159. [PMID: 33419065 PMCID: PMC7825450 DOI: 10.3390/nu13010159] [Citation(s) in RCA: 87] [Impact Index Per Article: 21.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2020] [Revised: 12/18/2020] [Accepted: 12/24/2020] [Indexed: 12/18/2022] Open
Abstract
Glucose levels in blood must be constantly maintained within a tight physiological range to sustain anabolism. Insulin regulates glucose homeostasis via its effects on glucose production from the liver and kidneys and glucose disposal in peripheral tissues (mainly skeletal muscle). Blood levels of glucose are regulated simultaneously by insulin-mediated rates of glucose production from the liver (and kidneys) and removal from muscle; adipose tissue is a key partner in this scenario, providing nonesterified fatty acids (NEFA) as an alternative fuel for skeletal muscle and liver when blood glucose levels are depleted. During sleep at night, the gradual development of insulin resistance, due to growth hormone and cortisol surges, ensures that blood glucose levels will be maintained within normal levels by: (a) switching from glucose to NEFA oxidation in muscle; (b) modulating glucose production from the liver/kidneys. After meals, several mechanisms (sequence/composition of meals, gastric emptying/intestinal glucose absorption, gastrointestinal hormones, hyperglycemia mass action effects, insulin/glucagon secretion/action, de novo lipogenesis and glucose disposal) operate in concert for optimal regulation of postprandial glucose fluctuations. The contribution of the liver in postprandial glucose homeostasis is critical. The liver is preferentially used to dispose over 50% of the ingested glucose and restrict the acute increases of glucose and insulin in the bloodstream after meals, thus protecting the circulation and tissues from the adverse effects of marked hyperglycemia and hyperinsulinemia.
Collapse
Affiliation(s)
- George D. Dimitriadis
- Sector of Medicine, Medical School, National and Kapodistrian University of Athens, 15772 Athens, Greece
| | - Eirini Maratou
- Department of Clinical Biochemistry, Medical School, National and Kapodistrian University of Athens, 15772 Athens, Greece;
- Department of Clinical Biochemistry, Medical School, “Attikon” University Hospital, Rimini 1, 12462 Chaidari, Greece
| | - Aikaterini Kountouri
- Research Institute and Diabetes Center, 2nd Department of Internal Medicine, “Attikon” University Hospital, 1 Rimini Street, 12542 Haidari, Greece; (A.K.); (V.L.)
| | - Mary Board
- St. Hilda’s College, University of Oxford, Cowley, Oxford OX4 1DY, UK;
| | - Vaia Lambadiari
- Research Institute and Diabetes Center, 2nd Department of Internal Medicine, “Attikon” University Hospital, 1 Rimini Street, 12542 Haidari, Greece; (A.K.); (V.L.)
| |
Collapse
|
21
|
Dirnena-Fusini I, Åm MK, Fougner AL, Carlsen SM, Christiansen SC. Intraperitoneal insulin administration in pigs: effect on circulating insulin and glucose levels. BMJ Open Diabetes Res Care 2021; 9:9/1/e001929. [PMID: 33452058 PMCID: PMC7813410 DOI: 10.1136/bmjdrc-2020-001929] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/29/2020] [Revised: 11/26/2020] [Accepted: 01/02/2021] [Indexed: 11/28/2022] Open
Abstract
INTRODUCTION The effect of intraperitoneal insulin infusion has limited evidence in the literature. Therefore, the aim of the study was to investigate the pharmacokinetics and pharmacodynamics of different intraperitoneal insulin boluses. There is a lack of studies comparing the insulin appearance in the systemic circulation after intraperitoneal compared with subcutaneous insulin delivery. Thus, we also aimed for a comparison with the subcutaneous route. RESEARCH DESIGN AND METHODS Eight anesthetized, non-diabetic pigs were given three different intraperitoneal insulin boluses (2, 5 and 10 U). The order of boluses for the last six pigs was randomized. Endogenous insulin and glucagon release were suppressed by repeated somatostatin analog injections. The first pig was used to identify the infusion rate of glucose to maintain stable glucose values throughout the experiment. The estimated difference between insulin boluses was compared using two-way analysis of variance (GraphPad Prism V.8).In addition, a trial of three pigs which received subcutaneous insulin boluses was included for comparison with intraperitoneal insulin boluses. RESULTS Decreased mean blood glucose levels were observed after 5 and 10 U intraperitoneal insulin boluses compared with the 2 U boluses. No changes in circulating insulin levels were observed after the 2 and 5 U intraperitoneal boluses, while increased circulating insulin levels were observed after the 10 U intraperitoneal boluses. Subcutaneously injected insulin resulted in higher values of circulating insulin compared with the corresponding intraperitoneal boluses. CONCLUSIONS Smaller intraperitoneal boluses of insulin have an effect on circulating glucose levels without increasing insulin levels in the systemic circulation. By increasing the insulin bolus, a major increase in circulating insulin was observed, with a minor additive effect on circulating glucose levels. This is compatible with a close to 100% first-pass effect in the liver after smaller intraperitoneal boluses. Subcutaneous insulin boluses markedly increased circulating insulin levels.
Collapse
Affiliation(s)
- Ilze Dirnena-Fusini
- Department Clinical and Molecular Medicine, Faculty of Medicine and Health Sciences, Norwegian University of Science and Technology, Trondheim, Norway
| | - Marte Kierulf Åm
- Department Clinical and Molecular Medicine, Faculty of Medicine and Health Sciences, Norwegian University of Science and Technology, Trondheim, Norway
- Department of Endocrinology, St Olavs Hospital Universitetssykehuset i Trondheim, Trondheim, Norway
| | - Anders Lyngvi Fougner
- Department of Engineering Cybernetics, Faculty of Information Technology and Electrical Engineering, Norwegian University of Science and Technology, Trondheim, Norway
| | - Sven Magnus Carlsen
- Department Clinical and Molecular Medicine, Faculty of Medicine and Health Sciences, Norwegian University of Science and Technology, Trondheim, Norway
- Department of Endocrinology, St Olavs Hospital Universitetssykehuset i Trondheim, Trondheim, Norway
| | - Sverre Christian Christiansen
- Department Clinical and Molecular Medicine, Faculty of Medicine and Health Sciences, Norwegian University of Science and Technology, Trondheim, Norway
- Department of Endocrinology, St Olavs Hospital Universitetssykehuset i Trondheim, Trondheim, Norway
| |
Collapse
|
22
|
Karimi R, Cleven A, Elbarbry F, Hoang H. The Impact of Fasting on Major Metabolic Pathways of Macronutrients and Pharmacokinetics Steps of Drugs. Eur J Drug Metab Pharmacokinet 2020; 46:25-39. [PMID: 33151502 DOI: 10.1007/s13318-020-00656-y] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
In this review, we have investigated how fasting promotes an adaptive cross-talk between different hormones and metabolic pathways to supply and meet the body's daily energy demands. We highlight in biochemical terms and mechanisms how fasting impacts four metabolic pathways-glycogenolysis, gluconeogenesis, amino acid oxidation, and fatty acid β-oxidation-that are actively engaged in the metabolism of carbohydrates, proteins, and lipids. Fasting results in reduced insulin secretion and increased glucagon and epinephrine release to prevent or stimulate metabolic reaction(s). Fasting stimulates glycogenolysis, amino acid and glucose oxidation, aminotransferase reactions in skeletal muscle, and promotes gluconeogenesis and urea production in the liver. In addition, fasting promotes gene expression of lipid metabolism in skeletal muscle, the synthesis of ketone bodies in the liver, and intracellular hormone-sensitive lipase activity in adipose tissue. Furthermore, the impact of fasting on reducing cellular damage by mitochondrial reactive oxygen species is discussed. Lastly, we briefly describe the impact of fasting on the four steps of pharmacokinetics-the absorption, distribution, metabolism, and excretion of a few select drugs-with an emphasis on the elimination of drugs related to the cytochrome-P450 family of enzymes.
Collapse
Affiliation(s)
- Reza Karimi
- Pacific University Oregon School of Pharmacy, 222 SE 8th Avenue, HPC-Ste 451, Hillsboro, OR, 97123, USA.
| | - Anita Cleven
- Pacific University Oregon School of Pharmacy, 222 SE 8th Avenue, HPC-Ste 451, Hillsboro, OR, 97123, USA
| | - Fawzy Elbarbry
- Pacific University Oregon School of Pharmacy, 222 SE 8th Avenue, HPC-Ste 451, Hillsboro, OR, 97123, USA
| | - Huy Hoang
- Pacific University Oregon School of Pharmacy, 222 SE 8th Avenue, HPC-Ste 451, Hillsboro, OR, 97123, USA
| |
Collapse
|
23
|
Sherk VD, Vigers T, Pyle L, Snell-Bergeon JK, Nadeau KJ, Rickels MR, Miller KM, Greenbaum CJ, Shah VN. Acute Hyperinsulinemia Alters Bone Turnover in Women and Men With Type 1 Diabetes. JBMR Plus 2020; 4:e10389. [PMID: 32995692 PMCID: PMC7507374 DOI: 10.1002/jbm4.10389] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/20/2020] [Revised: 06/25/2020] [Accepted: 06/30/2020] [Indexed: 12/24/2022] Open
Abstract
Type 1 diabetes (T1D) increases fracture risk across the lifespan. The low bone turnover associated with T1D is thought to be related to glycemic control, but it is unclear whether peripheral hyperinsulinemia due to dependence on exogenous insulin has an independent effect on suppressing bone turnover. The purpose of this study was to test the bone turnover marker (BTM) response to acute hyperinsulinemia. Fifty‐eight adults aged 18 to 65 years with T1D over 2 years were enrolled at seven T1D Exchange Clinic Network sites. Participants had T1D diagnosis between age 6 months to 45 years. Participants were stratified based on their residual endogenous insulin secretion measured as peak C‐peptide response to a mixed meal tolerance test. BTMs (CTX, P1NP, sclerostin [SCL], osteonectin [ON], alkaline phosphatase [ALP], osteocalcin [OCN], osteoprotegerin [OPG], osteopontin [OPN], and IGF‐1) were assessed before and at the end of a 2‐hour hyperinsulinemic‐euglycemic clamp (HEC). Baseline ON (r = −0.30, p = .022) and OCN (r = −0.41, p = .002) were negatively correlated with age at T1D diagnosis, but baseline BTMs were not associated with HbA1c. During the HEC, P1NP decreased significantly (−14.5 ± 44.3%; p = .020) from baseline. OCN, ON, and IGF‐1 all significantly increased (16.0 ± 13.1%, 29.7 ± 31.7%, 34.1 ± 71.2%, respectively; all p < .001) during the clamp. The increase in SCL was not significant (7.3 ± 32.9%, p = .098), but the decrease in CTX (−12.4 ± 48.9, p = .058) neared significance. ALP and OPG were not changed from baseline (p = .23 and p = .77, respectively). Baseline ON and SCL were higher in men, but OPG was higher in women (all p ≤ .029). SCL was the only BTM that changed differently in women than men. There were no differences in baseline BTMs or change in BTMs between C‐peptide groups. Exogenous hyperinsulinemia acutely alters bone turnover, suggesting a need to determine whether strategies to promote healthy remodeling may protect bone quality in T1D. © 2020 American Society for Bone and Mineral Research © 2020 The Authors. JBMR Plus published by Wiley Periodicals LLC on behalf of American Society for Bone and Mineral Research.
Collapse
Affiliation(s)
- Vanessa D Sherk
- Department of OrthopedicsSchool of Medicine University of Colorado Anschutz Medical Campus Aurora CO USA
| | - Timothy Vigers
- Department of Biostatistics and Informatics Colorado School of Public Health University of Colorado Anschutz Medical Campus Aurora CO USA.,Department of Pediatrics, Section of EndocrinologySchool of Medicine University of Colorado Anschutz Medical Campus Aurora CO USA.,Barbara Davis Center for Diabetes University of Colorado Anschutz Medical Campus Aurora CO USA
| | - Laura Pyle
- Department of Biostatistics and Informatics Colorado School of Public Health University of Colorado Anschutz Medical Campus Aurora CO USA.,Department of Pediatrics, Section of EndocrinologySchool of Medicine University of Colorado Anschutz Medical Campus Aurora CO USA.,Barbara Davis Center for Diabetes University of Colorado Anschutz Medical Campus Aurora CO USA
| | - Janet K Snell-Bergeon
- Barbara Davis Center for Diabetes University of Colorado Anschutz Medical Campus Aurora CO USA
| | - Kristen J Nadeau
- Children's Hospital Colorado University of Colorado School of Medicine Aurora CO USA
| | - Michael R Rickels
- Institute for Diabetes, Obesity & Metabolism University of Pennsylvania Perelman School of Medicine Philadelphia PA USA
| | | | | | - Viral N Shah
- Barbara Davis Center for Diabetes University of Colorado Anschutz Medical Campus Aurora CO USA
| |
Collapse
|
24
|
Batista TM, Garcia-Martin R, Cai W, Konishi M, O'Neill BT, Sakaguchi M, Kim JH, Jung DY, Kim JK, Kahn CR. Multi-dimensional Transcriptional Remodeling by Physiological Insulin In Vivo. Cell Rep 2020; 26:3429-3443.e3. [PMID: 30893613 DOI: 10.1016/j.celrep.2019.02.081] [Citation(s) in RCA: 37] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2018] [Revised: 01/11/2019] [Accepted: 02/21/2019] [Indexed: 12/12/2022] Open
Abstract
Regulation of gene expression is an important aspect of insulin action but in vivo is intertwined with changing levels of glucose and counter-regulatory hormones. Here we demonstrate that under euglycemic clamp conditions, physiological levels of insulin regulate interrelated networks of more than 1,000 transcripts in muscle and liver. These include expected pathways related to glucose and lipid utilization, mitochondrial function, and autophagy, as well as unexpected pathways, such as chromatin remodeling, mRNA splicing, and Notch signaling. These acutely regulated pathways extend beyond those dysregulated in mice with chronic insulin deficiency or insulin resistance and involve a broad network of transcription factors. More than 150 non-coding RNAs were regulated by insulin, many of which also responded to fasting and refeeding. Pathway analysis and RNAi knockdown revealed a role for lncRNA Gm15441 in regulating fatty acid oxidation in hepatocytes. Altogether, these changes in coding and non-coding RNAs provide an integrated transcriptional network underlying the complexity of insulin action.
Collapse
Affiliation(s)
- Thiago M Batista
- Section of Integrative Physiology and Metabolism, Joslin Diabetes Center, Harvard Medical School, Boston, MA 02215, USA
| | - Ruben Garcia-Martin
- Section of Integrative Physiology and Metabolism, Joslin Diabetes Center, Harvard Medical School, Boston, MA 02215, USA
| | - Weikang Cai
- Section of Integrative Physiology and Metabolism, Joslin Diabetes Center, Harvard Medical School, Boston, MA 02215, USA
| | - Masahiro Konishi
- Section of Integrative Physiology and Metabolism, Joslin Diabetes Center, Harvard Medical School, Boston, MA 02215, USA
| | - Brian T O'Neill
- Section of Integrative Physiology and Metabolism, Joslin Diabetes Center, Harvard Medical School, Boston, MA 02215, USA; Division of Endocrinology and Metabolism, Fraternal Order of Eagles Diabetes Research Center, University of Iowa Carver College of Medicine, Iowa City, IA, USA
| | - Masaji Sakaguchi
- Section of Integrative Physiology and Metabolism, Joslin Diabetes Center, Harvard Medical School, Boston, MA 02215, USA; Department of Metabolic Medicine, Kumamoto University, 1-1-1 Honjo, Chuoku, Kumamoto 860-8556, Japan
| | - Jong Hun Kim
- Program in Molecular Medicine, Department of Medicine, University of Massachusetts Medical School, Worcester, MA 01655, USA; Department of Food Science and Biotechnology, Sungshin University, Seoul 01133, Republic of Korea
| | - Dae Young Jung
- Program in Molecular Medicine, Department of Medicine, University of Massachusetts Medical School, Worcester, MA 01655, USA
| | - Jason K Kim
- Program in Molecular Medicine, Department of Medicine, University of Massachusetts Medical School, Worcester, MA 01655, USA; Division of Endocrinology, Metabolism, and Diabetes, Department of Medicine, University of Massachusetts Medical School, Worcester, MA 01655, USA
| | - C Ronald Kahn
- Section of Integrative Physiology and Metabolism, Joslin Diabetes Center, Harvard Medical School, Boston, MA 02215, USA.
| |
Collapse
|
25
|
Ehrhardt N, Cui J, Dagdeviren S, Saengnipanthkul S, Goodridge HS, Kim JK, Lantier L, Guo X, Chen YDI, Raffel LJ, Buchanan TA, Hsueh WA, Rotter JI, Goodarzi MO, Péterfy M. Adiposity-Independent Effects of Aging on Insulin Sensitivity and Clearance in Mice and Humans. Obesity (Silver Spring) 2019; 27:434-443. [PMID: 30801985 PMCID: PMC6474357 DOI: 10.1002/oby.22418] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/12/2018] [Accepted: 11/21/2018] [Indexed: 02/07/2023]
Abstract
OBJECTIVE Aging is associated with impaired insulin sensitivity and increased prevalence of type 2 diabetes. However, it remains unclear whether aging-associated insulin resistance is due to increased adiposity or other age-related factors. To address this question, the impact of aging on insulin sensitivity was investigated independently of changes in body composition. METHODS Cohorts of mice aged 4 to 8 months ("young") and 18 to 27 months ("aged") exhibiting similar body composition were characterized for glucose metabolism on chow and high-fat diets. Insulin sensitivity was assessed by hyperinsulinemic-euglycemic clamp analyses. The relationship between aging and insulin resistance in humans was investigated in 1,250 nondiabetic Mexican Americans who underwent hyperinsulinemic-euglycemic clamps. RESULTS In mice with similar body composition, age had no detrimental effect on plasma glucose and insulin levels. While aging did not diminish glucose tolerance, hyperinsulinemic-euglycemic clamps demonstrated impaired insulin sensitivity and reduced insulin clearance in aged mice on chow and high-fat diets. Consistent with results in the mouse, age remained an independent determinant of insulin resistance after adjustment for body composition in Mexican American males. CONCLUSIONS This study demonstrates that in addition to altered body composition, adiposity-independent mechanisms also contribute to aging-associated insulin resistance in mice and humans.
Collapse
Affiliation(s)
- Nicole Ehrhardt
- Department of Basic Medical Sciences, Western University of Health Sciences, Pomona, CA 91766, USA
| | - Jinrui Cui
- Division of Endocrinology, Diabetes, and Metabolism, Department of Medicine, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA
| | - Sezin Dagdeviren
- Program in Molecular Medicine, University of Massachusetts Medical School, Worcester, MA 01655, USA
| | - Suchaorn Saengnipanthkul
- Program in Molecular Medicine, University of Massachusetts Medical School, Worcester, MA 01655, USA
| | - Helen S. Goodridge
- Board of Governors Regenerative Medicine Institute, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA
- Department of Biomedical Sciences, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA
| | - Jason K. Kim
- Program in Molecular Medicine, University of Massachusetts Medical School, Worcester, MA 01655, USA
- Division of Endocrinology, Metabolism, and Diabetes, Department of Medicine, University of Massachusetts Medical School, Worcester, MA 01655, USA
| | - Louise Lantier
- Department of Molecular Physiology and Biophysics, Vanderbilt University, Nashville, TN 37235, USA
| | - Xiuqing Guo
- Institute for Translational Genomics and Population Sciences, Los Angeles BioMedical Research Institute and Department of Pediatrics, Harbor-UCLA Medical Center, Torrance, CA 90502, USA
| | - Yii-Der I. Chen
- Institute for Translational Genomics and Population Sciences, Los Angeles BioMedical Research Institute and Department of Pediatrics, Harbor-UCLA Medical Center, Torrance, CA 90502, USA
| | - Leslie J. Raffel
- Department of Pediatrics, Division of Genetic and Genomic Medicine, University of California, Irvine, CA 92697, USA
| | - Thomas A. Buchanan
- Department of Physiology and Biophysics and Department of Medicine, Keck School of Medicine of USC, Los Angeles, CA 90033, USA
| | - Willa A. Hsueh
- Division of Endocrinology, Diabetes and Metabolism, Department of Medicine, Wexner Medical Center, The Ohio State University, Columbus, OH 43210, USA
| | - Jerome I. Rotter
- Institute for Translational Genomics and Population Sciences, Los Angeles BioMedical Research Institute and Department of Pediatrics, Harbor-UCLA Medical Center, Torrance, CA 90502, USA
| | - Mark O. Goodarzi
- Division of Endocrinology, Diabetes, and Metabolism, Department of Medicine, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA
- Department of Medicine, David Geffen School of Medicine at UCLA, Los Angeles, CA 90095, USA
| | - Miklós Péterfy
- Department of Basic Medical Sciences, Western University of Health Sciences, Pomona, CA 91766, USA
- Department of Biomedical Sciences, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA
- Department of Medicine, David Geffen School of Medicine at UCLA, Los Angeles, CA 90095, USA
| |
Collapse
|
26
|
Edgerton DS, Scott M, Farmer B, Williams PE, Madsen P, Kjeldsen T, Brand CL, Fledelius C, Nishimura E, Cherrington AD. Targeting insulin to the liver corrects defects in glucose metabolism caused by peripheral insulin delivery. JCI Insight 2019; 5:126974. [PMID: 30830873 DOI: 10.1172/jci.insight.126974] [Citation(s) in RCA: 31] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Abstract
Peripheral hyperinsulinemia resulting from subcutaneous insulin injection is associated with metabolic defects which include abnormal glucose metabolism. The first aim of this study was to quantify the impairments in liver and muscle glucose metabolism that occur when insulin is delivered via a peripheral vein compared to when it is given through its endogenous secretory route (the hepatic portal vein) in overnight fasted conscious dogs. The second aim was to determine if peripheral delivery of a hepato-preferential insulin analog could restore the physiologic response to insulin that occurs under meal feeding conditions. This study is the first to show that hepatic glucose uptake correlates with insulin's direct effects on the liver under hyperinsulinemic-hyperglycemic conditions. In addition, glucose uptake was equally divided between the liver and muscle when insulin was infused into the portal vein, but when it was delivered into a peripheral vein the percentage of glucose taken up by muscle was 4-times greater than that going to the liver, with liver glucose uptake being less than half of normal. These defects could not be corrected by adjusting the dose of peripheral insulin. On the other hand, hepatic and non-hepatic glucose metabolism could be fully normalized by a hepato-preferential insulin analog.
Collapse
Affiliation(s)
- Dale S Edgerton
- Vanderbilt University School of Medicine, Department of Molecular Physiology and Biophysics, Nashville, Tennessee, USA
| | - Melanie Scott
- Vanderbilt University School of Medicine, Department of Molecular Physiology and Biophysics, Nashville, Tennessee, USA
| | - Ben Farmer
- Vanderbilt University School of Medicine, Department of Molecular Physiology and Biophysics, Nashville, Tennessee, USA
| | - Phillip E Williams
- Vanderbilt University Medical Center, Division of Surgical Research, Nashville, Tennessee, USA
| | - Peter Madsen
- Research and Development, Novo Nordisk A/S, Novo Nordisk Park, Maaleov, Denmark
| | - Thomas Kjeldsen
- Research and Development, Novo Nordisk A/S, Novo Nordisk Park, Maaleov, Denmark
| | - Christian L Brand
- Research and Development, Novo Nordisk A/S, Novo Nordisk Park, Maaleov, Denmark
| | - Christian Fledelius
- Research and Development, Novo Nordisk A/S, Novo Nordisk Park, Maaleov, Denmark
| | - Erica Nishimura
- Research and Development, Novo Nordisk A/S, Novo Nordisk Park, Maaleov, Denmark
| | - Alan D Cherrington
- Vanderbilt University School of Medicine, Department of Molecular Physiology and Biophysics, Nashville, Tennessee, USA
| |
Collapse
|
27
|
Kowalski GM, Moore SM, Hamley S, Selathurai A, Bruce CR. The Effect of Ingested Glucose Dose on the Suppression of Endogenous Glucose Production in Humans. Diabetes 2017; 66:2400-2406. [PMID: 28684634 DOI: 10.2337/db17-0433] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/11/2017] [Accepted: 06/21/2017] [Indexed: 11/13/2022]
Abstract
Insulin clamp studies have shown that the suppressive actions of insulin on endogenous glucose production (EGP) are markedly more sensitive than for stimulating glucose disposal (Rd). However, clamp conditions do not adequately mimic postprandial physiological responses. Here, using the variable infusion dual-tracer approach, we used a threefold range of ingested glucose doses (25, 50, and 75 g) to investigate how physiological changes in plasma insulin influence EGP in healthy subjects. Remarkably, the glucose responses were similar for all doses tested, yet there was a dose-dependent increase in insulin secretion and plasma insulin levels. Nonetheless, EGP was suppressed with the same rapidity and magnitude (∼55%) across all doses. The progressive hyperinsulinemia, however, caused a dose-dependent increase in the estimated rates of Rd, which likely accounts for the lack of a dose effect on plasma glucose excursions. This suggests that after glucose ingestion, the body preferentially permits a transient and optimal degree of postprandial hyperglycemia to efficiently enhance insulin-induced changes in glucose fluxes, thereby minimizing the demand for insulin secretion. This may represent an evolutionarily conserved mechanism that not only reduces the secretory burden on β-cells but also avoids the potential negative consequences of excessive insulin release into the systemic arterial circulation.
Collapse
Affiliation(s)
- Greg M Kowalski
- Institute for Physical Activity and Nutrition, School of Exercise and Nutrition Science, Deakin University, Burwood, Victoria, Australia
| | - Samantha M Moore
- Institute for Physical Activity and Nutrition, School of Exercise and Nutrition Science, Deakin University, Burwood, Victoria, Australia
| | - Steven Hamley
- Institute for Physical Activity and Nutrition, School of Exercise and Nutrition Science, Deakin University, Burwood, Victoria, Australia
| | - Ahrathy Selathurai
- Institute for Physical Activity and Nutrition, School of Exercise and Nutrition Science, Deakin University, Burwood, Victoria, Australia
| | - Clinton R Bruce
- Institute for Physical Activity and Nutrition, School of Exercise and Nutrition Science, Deakin University, Burwood, Victoria, Australia
| |
Collapse
|
28
|
Kummer S, Klee D, Kircheis G, Friedt M, Schaper J, Häussinger D, Mayatepek E, Meissner T. Screening for non-alcoholic fatty liver disease in children and adolescents with type 1 diabetes mellitus: a cross-sectional analysis. Eur J Pediatr 2017; 176:529-536. [PMID: 28213828 DOI: 10.1007/s00431-017-2876-1] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/15/2016] [Revised: 02/04/2017] [Accepted: 02/08/2017] [Indexed: 12/13/2022]
Abstract
UNLABELLED The liver is intensely involved in glucose metabolism and is thereby closely related to diabetes pathophysiology. Adult patients with type 1 diabetes mellitus (DM) are at an increased risk for non-alcoholic fatty liver disease (NAFLD). Here, we studied the prevalence of NAFLD in a cohort of children and adolescents with type 1 DM in a tertiary care paediatric diabetes centre in Germany. We screened 93 children and adolescents with type 1 DM using ultrasound, laboratory investigations, and liver stiffness measurements (Fibroscan® [FS] and acoustic radiation force imaging [ARFI]). Of these, 82 (88.1%) had completely normal results in all examined aspects. Only one patient (1.1%) fulfilled the criteria as potential NAFLD with ALT > twice the upper limit of normal. Ten of the 93 patients (10.8%) showed any mild abnormality in at least one examined category including ALT, conventional ultrasounds and liver stiffness measurements. However, none of these ten fulfilled the NAFLD case definition criteria. Therefore, these slightly abnormal results were judged to be unspecific or at least of unknown significance in terms of NAFLD indication. CONCLUSION Compared to data from the general population, our results do not indicate a significantly increased prevalence of NAFLD in this cohort, and advocate against the systematic screening for NAFLD in paediatric type 1 DM. What is Known: • Non-alcoholic fatty liver disease (NAFLD) is common in adults with type 1 DM, and paediatric patients with type 1 DM in Egypt and Saudi Arabia. What is New: • Our results do not indicate a significantly increased prevalence of NAFLD in a cohort of children and adolescents with type 1 DM from Germany compared to prevalence data from the general population. • This finding advocates against the systematic screening for NAFLD in paediatric type 1 DM in western countries.
Collapse
Affiliation(s)
- Sebastian Kummer
- Department of General Pediatrics, Neonatology and Pediatric Cardiology, University Children's Hospital, Moorenstr. 5, 40225, Duesseldorf, Germany.
| | - Dirk Klee
- Department of Diagnostic and Interventional Radiology, Medical Faculty, University Hospital, Moorenstr. 5, 40225, Dusseldorf, Germany
| | - Gerald Kircheis
- Department of Gastroenterology, Hepatology and Infectious Diseases, University Hospital, Moorenstr. 5, 40225, Duesseldorf, Germany
| | - Michael Friedt
- Department of General Pediatrics, Neonatology and Pediatric Cardiology, University Children's Hospital, Moorenstr. 5, 40225, Duesseldorf, Germany
| | - Joerg Schaper
- Department of Diagnostic and Interventional Radiology, Medical Faculty, University Hospital, Moorenstr. 5, 40225, Dusseldorf, Germany
| | - Dieter Häussinger
- Department of Gastroenterology, Hepatology and Infectious Diseases, University Hospital, Moorenstr. 5, 40225, Duesseldorf, Germany
| | - Ertan Mayatepek
- Department of General Pediatrics, Neonatology and Pediatric Cardiology, University Children's Hospital, Moorenstr. 5, 40225, Duesseldorf, Germany
| | - Thomas Meissner
- Department of General Pediatrics, Neonatology and Pediatric Cardiology, University Children's Hospital, Moorenstr. 5, 40225, Duesseldorf, Germany
| |
Collapse
|
29
|
Edgerton DS, Kraft G, Smith M, Farmer B, Williams PE, Coate KC, Printz RL, O'Brien RM, Cherrington AD. Insulin's direct hepatic effect explains the inhibition of glucose production caused by insulin secretion. JCI Insight 2017; 2:e91863. [PMID: 28352665 DOI: 10.1172/jci.insight.91863] [Citation(s) in RCA: 79] [Impact Index Per Article: 9.9] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
Insulin can inhibit hepatic glucose production (HGP) by acting directly on the liver as well as indirectly through effects on adipose tissue, pancreas, and brain. While insulin's indirect effects are indisputable, their physiologic role in the suppression of HGP seen in response to increased insulin secretion is not clear. Likewise, the mechanisms by which insulin suppresses lipolysis and pancreatic α cell secretion under physiologic circumstances are also debated. In this study, insulin was infused into the hepatic portal vein to mimic increased insulin secretion, and insulin's indirect liver effects were blocked either individually or collectively. During physiologic hyperinsulinemia, plasma free fatty acid (FFA) and glucagon levels were clamped at basal values and brain insulin action was blocked, but insulin's direct effects on the liver were left intact. Insulin was equally effective at suppressing HGP when its indirect effects were absent as when they were present. In addition, the inhibition of lipolysis, as well as glucagon and insulin secretion, did not require CNS insulin action or decreased plasma FFA. This indicates that the rapid suppression of HGP is attributable to insulin's direct effect on the liver and that its indirect effects are redundant in the context of a physiologic increase in insulin secretion.
Collapse
Affiliation(s)
- Dale S Edgerton
- Department of Molecular Physiology and Biophysics, Vanderbilt University, Nashville, Tennessee, USA
| | - Guillaume Kraft
- Department of Molecular Physiology and Biophysics, Vanderbilt University, Nashville, Tennessee, USA
| | - Marta Smith
- Department of Molecular Physiology and Biophysics, Vanderbilt University, Nashville, Tennessee, USA
| | - Ben Farmer
- Department of Molecular Physiology and Biophysics, Vanderbilt University, Nashville, Tennessee, USA
| | - Phillip E Williams
- Division of Surgical Research, Vanderbilt University School of Medicine, Nashville, Tennessee, USA
| | - Katie C Coate
- Samford University, Department of Nutrition and Dietetics, Birmingham, Alabama, USA
| | - Richard L Printz
- Department of Molecular Physiology and Biophysics, Vanderbilt University, Nashville, Tennessee, USA
| | - Richard M O'Brien
- Department of Molecular Physiology and Biophysics, Vanderbilt University, Nashville, Tennessee, USA
| | - Alan D Cherrington
- Department of Molecular Physiology and Biophysics, Vanderbilt University, Nashville, Tennessee, USA
| |
Collapse
|
30
|
|
31
|
Naftalin RJ. A computer model simulating human glucose absorption and metabolism in health and metabolic disease states. F1000Res 2016; 5:647. [PMID: 27347379 PMCID: PMC4909112 DOI: 10.12688/f1000research.8299.1] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 03/31/2016] [Indexed: 12/16/2022] Open
Abstract
A computer model designed to simulate integrated glucose-dependent changes in splanchnic blood flow with small intestinal glucose absorption, hormonal and incretin circulation and hepatic and systemic metabolism in health and metabolic diseases e.g. non-alcoholic fatty liver disease, (NAFLD), non-alcoholic steatohepatitis, (NASH) and type 2 diabetes mellitus, (T2DM) demonstrates how when glucagon-like peptide-1, (GLP-1) is synchronously released into the splanchnic blood during intestinal glucose absorption, it stimulates superior mesenteric arterial (SMA) blood flow and by increasing passive intestinal glucose absorption, harmonizes absorption with its distribution and metabolism. GLP-1 also synergises insulin-dependent net hepatic glucose uptake (NHGU). When GLP-1 secretion is deficient post-prandial SMA blood flow is not increased and as NHGU is also reduced, hyperglycaemia follows. Portal venous glucose concentration is also raised, thereby retarding the passive component of intestinal glucose absorption. Increased pre-hepatic sinusoidal resistance combined with portal hypertension leading to opening of intrahepatic portosystemic collateral vessels are NASH-related mechanical defects that alter the balance between splanchnic and systemic distributions of glucose, hormones and incretins.The model reveals the latent contribution of portosystemic shunting in development of metabolic disease. This diverts splanchnic blood content away from the hepatic sinuses to the systemic circulation, particularly during the glucose absorptive phase of digestion, resulting in inappropriate increases in insulin-dependent systemic glucose metabolism. This hastens onset of hypoglycaemia and thence hyperglucagonaemia. The model reveals that low rates of GLP-1 secretion, frequently associated with T2DM and NASH, may be also be caused by splanchnic hypoglycaemia, rather than to intrinsic loss of incretin secretory capacity. These findings may have therapeutic implications on GLP-1 agonist or glucagon antagonist usage.
Collapse
Affiliation(s)
- Richard J Naftalin
- Departments of Physiology and Vascular Biology, BHF centre of research excellence, King's College London School of Medicine, London, UK
| |
Collapse
|
32
|
Shao J, Zaro JL, Shen WC. Tissue barriers and novel approaches to achieve hepatoselectivity of subcutaneously-injected insulin therapeutics. Tissue Barriers 2016; 4:e1156804. [PMID: 27358753 DOI: 10.1080/21688370.2016.1156804] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2016] [Revised: 02/09/2016] [Accepted: 02/11/2016] [Indexed: 10/22/2022] Open
Abstract
Current subcutaneously (s.c.)-injected insulin (INS) products result in a hyperinsulin exposure to peripheral tissues (skeletal muscle and adipose) while INS hardly accesses to liver after injection. This unphysiological distribution raises risks of hypoglycemia episode and causes weight gain after long term treatment. An ideal INS replacement therapy requires the distribution or action of exogenous INS to more closely mimic physiological INS in terms of its preferential hepatic action. However, there are 2 factors that limit the ability of s.c. injected INS to restore the liver: peripheral gradient in INS deficient diabetes patients: (1) the transport of INS in capillary endothelium and peripheral tissues from the injection site; and (2) peripheral INS receptor (IR) mediated INS degradation. In this review, the tissue barriers against efficient liver targeting of s.c. injected INS are discussed and current advances in developing hepatoselective insulin therapeutics are introduced.
Collapse
Affiliation(s)
- Juntang Shao
- Department of Pharmacology and Pharmaceutical Sciences, University of Southern California , Los Angeles, CA, USA
| | - Jennica L Zaro
- Department of Pharmacology and Pharmaceutical Sciences, University of Southern California , Los Angeles, CA, USA
| | - Wei-Chiang Shen
- Department of Pharmacology and Pharmaceutical Sciences, University of Southern California , Los Angeles, CA, USA
| |
Collapse
|
33
|
Affiliation(s)
- Dale S Edgerton
- Department of Molecular Physiology and Biophysics, Vanderbilt University School of Medicine, Nashville, TN
| | - Alan D Cherrington
- Department of Molecular Physiology and Biophysics, Vanderbilt University School of Medicine, Nashville, TN
| |
Collapse
|