1
|
Freyberg Z, Codario RA. Biological mechanisms of dopamine D 2-like receptor agonist therapy in diabetes. Front Endocrinol (Lausanne) 2025; 16:1532414. [PMID: 39906261 PMCID: PMC11791534 DOI: 10.3389/fendo.2025.1532414] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/21/2024] [Accepted: 01/07/2025] [Indexed: 02/06/2025] Open
Affiliation(s)
- Zachary Freyberg
- Department of Psychiatry, University of Pittsburgh, Pittsburgh, PA, United States
- Department of Cell Biology, University of Pittsburgh, Pittsburgh, PA, United States
| | - Ronald A. Codario
- Department of Medicine, Division of Endocrinology & Metabolism, University of Pittsburgh, Pittsburgh, PA, United States
- Department of Endocrinology, VA Pittsburgh Healthcare System, Pittsburgh, PA, United States
| |
Collapse
|
2
|
Vora V, Tirgar P, Raval K. Anti-Diabetic and Insulinotropic Effects of p-Anisic Acid in High-Fat Diet and Streptozotocin Induced Type-2 Diabetic Rats. Chem Biodivers 2024; 21:e202401575. [PMID: 39210671 DOI: 10.1002/cbdv.202401575] [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: 06/28/2024] [Revised: 08/25/2024] [Accepted: 08/30/2024] [Indexed: 09/04/2024]
Abstract
PURPOSE To evaluate the antidiabetic effect of p-anisic acid (p-AA) against type 2 diabetes mellitus (T2DM). METHODS Thirty-six male rats were utilized for the study. The animals were provided with the high-fat diet (HFD) and T2DM was induced through 35 mg/kg streptozotocin (STZ). Subsequently, the animals were allocated to 6 groups and subjected to the treatments for 4 weeks followed by a 2-week observation. Three treatment groups were administered with p-AA (25 mg/kg, 50 mg/kg or 100 mg/kg). Glibenclamide (3 mg/kg) was provided to the standard control group. The normal control and disease control groups were supplied with 0.1 % carboxymethyl cellulose (CMC). Subsequently, the effects of treatment on body weight (BW), blood glucose level (BGL), glycosylated haemoglobin (HbA1c), insulin, and lipid parameters were measured and the pancreas was isolated for histopathology. RESULTS A statistically significant (P-value<0.001) increase in BW was observed in the p-AA (50 mg/kg and 100 mg/kg) groups. The BGL decreased significantly (P-value<0.001) in p-AA groups. p-AA significantly (P-value<0.001) regulated the levels of HbA1c and serum insulin and lipid parameters. A notable improvement in the morphology of the pancreas was observed. CONCLUSION Treatment with p-AA exhibited significant antidiabetic effects against STZ-HFD-induced T2DM.
Collapse
Affiliation(s)
- Vishal Vora
- Research Scholar, School of Pharmacy, RK University, Rajkot, Gujarat, India
| | - Pravin Tirgar
- Director, School of Pharmacy, RK University, Rajkot, Gujarat, India
| | - Keval Raval
- Assistant Professor, Ramanbhai Patel College of Pharmacy, Charotar University of Science and Technology, CHARUSAT Campus, Changa, Anand, Gujarat, 388421, India
| |
Collapse
|
3
|
Ferrero E, Masini M, Carli M, Moscato S, Beffy P, Vaglini F, Mattii L, Corti A, Scarselli M, Novelli M, De Tata V. Dopamine-mediated autocrine inhibition of insulin secretion. Mol Cell Endocrinol 2024; 592:112294. [PMID: 38838763 DOI: 10.1016/j.mce.2024.112294] [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: 02/20/2024] [Revised: 05/15/2024] [Accepted: 06/01/2024] [Indexed: 06/07/2024]
Abstract
The aim of the present research was to explore the mechanisms underlying the role of dopamine in the regulation of insulin secretion in beta cells. The effect of dopamine on insulin secretion was investigated on INS 832/13 cell line upon glucose and other secretagogues stimulation. Results show that dopamine significantly inhibits insulin secretion stimulated by both glucose and other secretagogues, while it has no effect on the basal secretion. This effect requires the presence of dopamine during incubation with the various secretagogues. Both electron microscopy and immunohistochemistry indicate that in beta cells the D2 dopamine receptor is localized within the insulin granules. Blocking dopamine entry into the insulin granules by inhibiting the VMAT2 transporter with tetrabenazine causes a significant increase in ROS production. Our results confirm that dopamine plays an important role in the regulation of insulin secretion by pancreatic beta cells through a regulated and precise compartmentalization mechanisms.
Collapse
Affiliation(s)
| | | | | | - Stefania Moscato
- Department of Clinical and Experimental Medicine, Italy; Interdepartmental Research Centre "Nutraceuticals and Food for Health", Italy
| | | | | | - Letizia Mattii
- Department of Clinical and Experimental Medicine, Italy; Interdepartmental Research Centre "Nutraceuticals and Food for Health", Italy
| | | | | | | | - Vincenzo De Tata
- Department of Translational Research, Italy; CIME (Interdepartmental Centre of Electron Microscopy), University of Pisa, Pisa, Italy.
| |
Collapse
|
4
|
Hendricks SA, Paul MJ, Subramaniam Y, Vijayam B. A collectanea of food insulinaemic index: 2023. Clin Nutr ESPEN 2024; 63:92-104. [PMID: 38941186 DOI: 10.1016/j.clnesp.2024.06.017] [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: 02/12/2024] [Revised: 04/28/2024] [Accepted: 06/11/2024] [Indexed: 06/30/2024]
Abstract
BACKGROUND AND AIMS To systematically update and publish the lnsulinaemic Index (II) value compilation of food/beverages. METHODS A literature search identified around 400 scholarly articles published between inception and December 2023. II values were pooled according to the selection criteria of at least 10 healthy, non-diabetic subjects with normal BMI. In addition, the II reported should have been derived from incremental area under the curve (iAUC) calculation of the insulin concentration over time. The reference food used from the pooled articles were either glucose or bread. RESULTS The II of 629 food/beverage items were found from 80 distinct articles. This is almost a five-fold increase in the number of entries from a previous compilation in 2011. Furthermore, these articles originated from 32 different countries, and were cleaved into 25 food categories. The II values ranged from 1 to 209. The highest overall recorded II was for a soy milk-based infant formula while the lowest was for both acacia fibre and gin. Upon clustering to single food, the infant formula retained the highest II while both acacia fibre and gin maintained the lowest recording. As for mixed meal, a potato dish served with a beverage recorded the highest II while a type of taco served with a sweetener, vegetable and fruit had the lowest II. Our minimum and maximum II data values replace the entries reported by previous compilations. CONCLUSION Acknowledging some limitations, these data would facilitate clinical usage of II for various applications in research, clinical nutrition, clinical medicine, diabetology and precision medicine. Future studies concerning II should investigate standardisation of reference food, including glucose and the test food portion. Although this collectanea adds up new food/beverages II values, priority should be given to populate this database.
Collapse
Affiliation(s)
| | | | - Yuganeswary Subramaniam
- Surgical Department, Hospital Besar Pulau Pinang, Jalan Residensi, 10990 Georgetown, Pulau Pinang, Malaysia
| | - Bhuwaneswaran Vijayam
- Newcastle University Medicine Malaysia (NUMed), Iskandar Puteri, 79200 Johor, Malaysia; Regenerative Medicine Working Group, Newcastle University Medicine Malaysia (NUMed), 79200 Iskandar Puteri, Johor, Malaysia.
| |
Collapse
|
5
|
Bonifazi A, Ellenberger M, Farino ZJ, Aslanoglou D, Rais R, Pereira S, Mantilla-Rivas JO, Boateng CA, Eshleman AJ, Janowsky A, Hahn MK, Schwartz GJ, Slusher BS, Newman AH, Freyberg Z. Development of Novel Tools for Dissection of Central Versus Peripheral Dopamine D2-Like Receptor Signaling in Dysglycemia. Diabetes 2024; 73:1411-1425. [PMID: 38869519 PMCID: PMC11333378 DOI: 10.2337/db24-0175] [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: 03/04/2024] [Accepted: 05/23/2024] [Indexed: 06/14/2024]
Abstract
Dopamine (DA) D2-like receptors in both the central nervous system (CNS) and the periphery are key modulators of metabolism. Moreover, disruption of D2-like receptor signaling is implicated in dysglycemia. Yet, the respective metabolic contributions of CNS versus peripheral D2-like receptors, including D2 (D2R) and D3 (D3R) receptors, remain poorly understood. To address this, we developed new pharmacological tools, D2-like receptor agonists with diminished and delayed blood-brain barrier capability, to selectively manipulate D2R/D3R signaling in the periphery. We designated bromocriptine methiodide (BrMeI), a quaternary methiodide analog of D2R/D3R agonist and diabetes drug bromocriptine, as our lead compound based on preservation of D2R/D3R binding and functional efficacy. We then used BrMeI and unmodified bromocriptine to dissect relative contributions of CNS versus peripheral D2R/D3R signaling in treating dysglycemia. Systemic administration of bromocriptine, with unrestricted access to CNS and peripheral targets, significantly improved both insulin sensitivity and glucose tolerance in obese, dysglycemic mice in vivo. In contrast, metabolic improvements were attenuated when access to bromocriptine was restricted either to the CNS through intracerebroventricular administration or delayed access to the CNS via BrMeI. Our findings demonstrate that the coordinated actions of both CNS and peripheral D2-like receptors are required for correcting dysglycemia. Ultimately, the development of a first-generation of drugs designed to selectively target the periphery provides a blueprint for dissecting mechanisms of central versus peripheral DA signaling and paves the way for novel strategies to treat dysglycemia. ARTICLE HIGHLIGHTS
Collapse
Affiliation(s)
- Alessandro Bonifazi
- Medicinal Chemistry Section, National Institute on Drug Abuse, Intramural Research Program, National Institutes of Health, Baltimore, MD
| | - Michael Ellenberger
- Medicinal Chemistry Section, National Institute on Drug Abuse, Intramural Research Program, National Institutes of Health, Baltimore, MD
| | | | | | - Rana Rais
- Department of Neurology, Johns Hopkins Drug Discovery, The Johns Hopkins University School of Medicine, Baltimore, MD
| | - Sandra Pereira
- Centre for Addiction and Mental Health, Toronto, Ontario, Canada
- Department of Physiology, University of Toronto, Toronto, Ontario, Canada
| | | | - Comfort A. Boateng
- Medicinal Chemistry Section, National Institute on Drug Abuse, Intramural Research Program, National Institutes of Health, Baltimore, MD
| | - Amy J. Eshleman
- Research Service, Veterans Affairs Portland Health Care System, Portland, OR
- Departments of Behavioral Neuroscience and Psychiatry, Oregon Health & Science University, Portland, OR
| | - Aaron Janowsky
- Research Service, Veterans Affairs Portland Health Care System, Portland, OR
- Departments of Behavioral Neuroscience and Psychiatry, Oregon Health & Science University, Portland, OR
- Methamphetamine Abuse Research Center, Oregon Health & Science University, Portland, OR
| | - Margaret K. Hahn
- Centre for Addiction and Mental Health, Toronto, Ontario, Canada
- Institute of Medical Sciences, University of Toronto, Toronto, Ontario, Canada
- Department of Pharmacology, University of Toronto, Toronto, Ontario, Canada
- Department of Psychiatry, University of Toronto, Toronto, Ontario, Canada
- Banting & Best Diabetes Centre, Toronto, Ontario, Canada
| | - Gary J. Schwartz
- The Fleischer Institute for Diabetes and Metabolism, Albert Einstein College of Medicine, Bronx, NY
- Division of Endocrinology, Department of Medicine, Albert Einstein College of Medicine, Bronx, NY
| | - Barbara S. Slusher
- Department of Neurology, Johns Hopkins Drug Discovery, The Johns Hopkins University School of Medicine, Baltimore, MD
| | - Amy Hauck Newman
- Medicinal Chemistry Section, National Institute on Drug Abuse, Intramural Research Program, National Institutes of Health, Baltimore, MD
| | - Zachary Freyberg
- Department of Psychiatry, University of Pittsburgh, Pittsburgh, PA
- Department of Cell Biology, University of Pittsburgh, Pittsburgh, PA
| |
Collapse
|
6
|
Zhou L, Zhu R, Figueroa-Miranda G, Neis M, Offenhäusser A, Mayer D. Ratiometric electrochemical aptasensor with strand displacement for insulin detection in blood samples. Anal Chim Acta 2024; 1317:342823. [PMID: 39029996 DOI: 10.1016/j.aca.2024.342823] [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: 04/02/2024] [Revised: 05/30/2024] [Accepted: 06/03/2024] [Indexed: 07/21/2024]
Abstract
BACKGROUND Diabetes patients suffer either from insulin deficiency or resistance with a high risk of severe long-term complications, thus the quantitative assessment of insulin level is highly desired for diabetes surveillance and management. Utilizing insulin-capturing aptamers may facilitate the development of affordable biosensors however, their rigid G-quadruplex structures impair conformational changes of the aptamers and diminish the sensor signals. RESULTS Here we report on a ratiometric, electrochemical insulin aptasensor which is achieved by hybridization of an insulin-capturing aptamer and a partially complementary ssDNA to break the rigid G-quadruplex structures. To improve the durability of the aptasensor, the capturing aptamer was immobilized on gold electrodes via two dithiol-phosphoramidite functional groups while methoxy-polyethylene glycol thiol was used as a blocking molecule. The exposure of the sensor to insulin-containing solutions induced the dissociation of the hybridized DNA accompanied by a conformational rearrangement of the capturing aptamer back into a G-quadruplex structure. The reliability of sensor readout was improved by the adoption of an AND logic gate utilizing anthraquinone and methylene blue redox probes associated to the aptamer and complementary strand, respectively. Our aptasensor possessed an improved detection limit of 0.15 nM in comparison to aptasensors without strand displacement. SIGNIFICANCE The sensor was adapted for detection in real blood and is ready for future PoC diagnostics. The capability of monitoring the insulin level in an affordably manner can improve the treatment for an increasing number of patients in developed and developing nations. The utilization of low-cost and versatile aptamer receptors together with the engineering of ratiometric electrochemical signal recording has the potential to considerably advance the current insulin detection technology toward multi-analyte diabetes sensors.
Collapse
Affiliation(s)
- Lei Zhou
- Institute of Biological Information Processing, Bioelectronics (IBI-3), Forschungszentrum Jülich GmbH, Jülich, Germany; Faculty I, RWTH Aachen University, Aachen, Germany
| | - Ruifeng Zhu
- Institute of Biological Information Processing, Bioelectronics (IBI-3), Forschungszentrum Jülich GmbH, Jülich, Germany
| | - Gabriela Figueroa-Miranda
- Institute of Biological Information Processing, Bioelectronics (IBI-3), Forschungszentrum Jülich GmbH, Jülich, Germany
| | - Marc Neis
- Institute of Biological Information Processing, Bioelectronics (IBI-3), Forschungszentrum Jülich GmbH, Jülich, Germany
| | - Andreas Offenhäusser
- Institute of Biological Information Processing, Bioelectronics (IBI-3), Forschungszentrum Jülich GmbH, Jülich, Germany
| | - Dirk Mayer
- Institute of Biological Information Processing, Bioelectronics (IBI-3), Forschungszentrum Jülich GmbH, Jülich, Germany.
| |
Collapse
|
7
|
Zhang T, Wu H, Qiu C, Wang M, Wang H, Zhu S, Xu Y, Huang Q, Li S. Ultrasensitive Hierarchical AuNRs@SiO 2@Ag SERS Probes for Enrichment and Detection of Insulin and C-Peptide in Serum. Int J Nanomedicine 2024; 19:6281-6293. [PMID: 38919772 PMCID: PMC11198011 DOI: 10.2147/ijn.s462601] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2024] [Accepted: 05/28/2024] [Indexed: 06/27/2024] Open
Abstract
Introduction Insulin and C-peptide played crucial roles as clinical indicators for diabetes and certain liver diseases. However, there has been limited research on the simultaneous detection of insulin and C-peptide in trace serum. It is necessary to develop a novel method with high sensitivity and specificity for detecting insulin and C-peptide simultaneously. Methods A core-shell-satellites hierarchical structured nanocomposite was fabricated as SERS biosensor using a simple wet-chemical method, employing 4-MBA and DTNB for recognition and antibodies for specific capture. Gold nanorods (Au NRs) were modified with Raman reporter molecules and silver nanoparticles (Ag NPs), creating SERS tags with high sensitivity for detecting insulin and C-peptide. Antibody-modified commercial carboxylated magnetic bead@antibody served as the capture probes. Target materials were captured by probes and combined with SERS tags, forming a "sandwich" composite structure for subsequent detection. Results Under optimized conditions, the nanocomposite fabricated could be used to detect simultaneously for insulin and C-peptide with the detection limit of 4.29 × 10-5 pM and 1.76 × 10-10 nM in serum. The insulin concentration (4.29 × 10-5-4.29 pM) showed a strong linear correlation with the SERS intensity at 1075 cm-1, with high recoveries (96.4-105.3%) and low RSD (0.8%-10.0%) in detecting human serum samples. Meanwhile, the C-peptide concentration (1.76 × 10-10-1.76 × 10-3 nM) also showed a specific linear correlation with the SERS intensity at 1333 cm-1, with recoveries 85.4%-105.0% and RSD 1.7%-10.8%. Conclusion This breakthrough provided a novel, sensitive, convenient and stable approach for clinical diagnosis of diabetes and certain liver diseases. Overall, our findings presented a significant contribution to the field of biomedical research, opening up new possibilities for improved diagnosis and monitoring of diabetes and liver diseases.
Collapse
Affiliation(s)
- Tong Zhang
- Medical Technology School of Xuzhou Medical University, Xuzhou, Jiangsu, 221000, People’s Republic of China
- Department of Laboratory Medicine, Affiliated Hospital of Xuzhou Medical University, Xuzhou, Jiangsu, 221000, People’s Republic of China
- Chuzhou Center for Disease Control and Prevention, Chuzhou City, Anhui, 239000, People’s Republic of China
| | - Han Wu
- Medical Technology School of Xuzhou Medical University, Xuzhou, Jiangsu, 221000, People’s Republic of China
- Department of Laboratory Medicine, Affiliated Hospital of Xuzhou Medical University, Xuzhou, Jiangsu, 221000, People’s Republic of China
| | - Chenling Qiu
- Medical Technology School of Xuzhou Medical University, Xuzhou, Jiangsu, 221000, People’s Republic of China
- Department of Laboratory Medicine, Affiliated Hospital of Xuzhou Medical University, Xuzhou, Jiangsu, 221000, People’s Republic of China
| | - Mingxin Wang
- Medical Technology School of Xuzhou Medical University, Xuzhou, Jiangsu, 221000, People’s Republic of China
- Department of Laboratory Medicine, Affiliated Hospital of Xuzhou Medical University, Xuzhou, Jiangsu, 221000, People’s Republic of China
| | - Haiting Wang
- Medical Technology School of Xuzhou Medical University, Xuzhou, Jiangsu, 221000, People’s Republic of China
- Department of Laboratory Medicine, Affiliated Hospital of Xuzhou Medical University, Xuzhou, Jiangsu, 221000, People’s Republic of China
| | - Shunhua Zhu
- Medical Technology School of Xuzhou Medical University, Xuzhou, Jiangsu, 221000, People’s Republic of China
- Public Experimental Research Center of Xuzhou Medical University, Xuzhou City, Jiangsu, 221004, People’s Republic of China
| | - Yinhai Xu
- Department of Laboratory Medicine, Affiliated Hospital of Xuzhou Medical University, Xuzhou, Jiangsu, 221000, People’s Republic of China
| | - Qingli Huang
- Medical Technology School of Xuzhou Medical University, Xuzhou, Jiangsu, 221000, People’s Republic of China
- Public Experimental Research Center of Xuzhou Medical University, Xuzhou City, Jiangsu, 221004, People’s Republic of China
| | - Shibao Li
- Medical Technology School of Xuzhou Medical University, Xuzhou, Jiangsu, 221000, People’s Republic of China
- Department of Laboratory Medicine, Affiliated Hospital of Xuzhou Medical University, Xuzhou, Jiangsu, 221000, People’s Republic of China
| |
Collapse
|
8
|
Bonifazi A, Ellenberger M, Farino ZJ, Aslanoglou D, Rais R, Pereira S, Mantilla-Rivas JO, Boateng CA, Eshleman AJ, Janowsky A, Hahn MK, Schwartz GJ, Slusher BS, Newman AH, Freyberg Z. Development of novel tools for dissection of central versus peripheral dopamine D 2-like receptor signaling in dysglycemia. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.02.21.581451. [PMID: 38529497 PMCID: PMC10962703 DOI: 10.1101/2024.02.21.581451] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/27/2024]
Abstract
Dopamine (DA) D2-like receptors in both the central nervous system (CNS) and the periphery are key modulators of metabolism. Moreover, disruption of D2-like receptor signaling is implicated in dysglycemia. Yet, the respective metabolic contributions of CNS versus peripheral D2-like receptors including D2 (D2R) and D3 (D3R) receptors remain poorly understood. To address this, we developed new pharmacological tools, D2-like receptor agonists with diminished and delayed blood-brain barrier capability, to selectively manipulate D2R/D3R signaling in the periphery. We designated bromocriptine methiodide (BrMeI), a quaternary methiodide analogue of D2/3R agonist and diabetes drug bromocriptine, as our lead compound based on preservation of D2R/D3R binding and functional efficacy. We then used BrMeI and unmodified bromocriptine to dissect relative contributions of CNS versus peripheral D2R/D3R signaling in treating dysglycemia. Systemic administration of bromocriptine, with unrestricted access to CNS and peripheral targets, significantly improved both insulin sensitivity and glucose tolerance in obese, dysglycemic mice in vivo. In contrast, metabolic improvements were attenuated when access to bromocriptine was restricted either to the CNS through intracerebroventricular administration or delayed access to the CNS via BrMeI. Our findings demonstrate that the coordinated actions of both CNS and peripheral D2-like receptors are required for correcting dysglycemia. Ultimately, the development of a first-generation of drugs designed to selectively target the periphery provides a blueprint for dissecting mechanisms of central versus peripheral DA signaling and paves the way for novel strategies to treat dysglycemia.
Collapse
Affiliation(s)
- Alessandro Bonifazi
- Medicinal Chemistry Section, National Institute on Drug Abuse, Intramural Research Program, National Institutes of Health, Baltimore, MD, USA
| | - Michael Ellenberger
- Medicinal Chemistry Section, National Institute on Drug Abuse, Intramural Research Program, National Institutes of Health, Baltimore, MD, USA
| | - Zachary J. Farino
- Department of Psychiatry, University of Pittsburgh, Pittsburgh, PA, USA
| | | | - Rana Rais
- Department of Neurology, Johns Hopkins Drug Discovery, The Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Sandra Pereira
- Centre for Addiction and Mental Health, Toronto, ON, Canada
- Department of Physiology, University of Toronto, Toronto, ON, Canada
| | | | - Comfort A. Boateng
- Medicinal Chemistry Section, National Institute on Drug Abuse, Intramural Research Program, National Institutes of Health, Baltimore, MD, USA
| | - Amy J. Eshleman
- Research Service, VA Portland Health Care System, Portland, Oregon, USA
- Departments of Behavioral Neuroscience and Psychiatry, Oregon Health & Science University, Portland, OR, USA
| | - Aaron Janowsky
- Research Service, VA Portland Health Care System, Portland, Oregon, USA
- Departments of Behavioral Neuroscience and Psychiatry, Oregon Health & Science University, Portland, OR, USA
- Methamphetamine Abuse Research Center, Oregon Health & Science University, Portland, OR, USA
| | - Margaret K. Hahn
- Centre for Addiction and Mental Health, Toronto, ON, Canada
- Institute of Medical Sciences, University of Toronto, Toronto, ON, Canada
- Department of Pharmacology, University of Toronto, Toronto, ON, Canada
- Department of Psychiatry, University of Toronto, Toronto, ON, Canada
- Banting & Best Diabetes Centre, Toronto, ON, Canada
| | - Gary J. Schwartz
- The Fleischer Institute for Diabetes and Metabolism, Albert Einstein College of Medicine, Bronx, NY, USA
- Division of Endocrinology, Department of Medicine, Albert Einstein College of Medicine, Bronx, NY, USA
| | - Barbara S. Slusher
- Department of Neurology, Johns Hopkins Drug Discovery, The Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Amy Hauck Newman
- Medicinal Chemistry Section, National Institute on Drug Abuse, Intramural Research Program, National Institutes of Health, Baltimore, MD, USA
| | - Zachary Freyberg
- Department of Psychiatry, University of Pittsburgh, Pittsburgh, PA, USA
- Department of Cell Biology, University of Pittsburgh, Pittsburgh, PA, USA
- Lead Contact
| |
Collapse
|
9
|
Thoduvayil S, Weerakkody JS, Sundaram RVK, Topper M, Bera M, Coleman J, Li X, Mariappan M, Ramakrishnan S. Rapid Quantification of First and Second Phase Insulin Secretion Dynamics using an In vitro Platform for Improving Insulin Therapy. Cell Calcium 2023; 113:102766. [PMID: 37295201 PMCID: PMC10450995 DOI: 10.1016/j.ceca.2023.102766] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2023] [Revised: 06/01/2023] [Accepted: 06/02/2023] [Indexed: 06/12/2023]
Abstract
High-throughput quantification of the first- and second-phase insulin secretion dynamics is intractable with current methods. The fact that independent secretion phases play distinct roles in metabolism necessitates partitioning them separately and performing high-throughput compound screening to target them individually. We developed an insulin-nanoluc luciferase reporter system to dissect the molecular and cellular pathways involved in the separate phases of insulin secretion. We validated this method through genetic studies, including knockdown and overexpression, as well as small-molecule screening and their effects on insulin secretion. Furthermore, we demonstrated that the results of this method are well correlated with those of single-vesicle exocytosis experiments conducted on live cells, providing a quantitative reference for the approach. Thus, we have developed a robust methodology for screening small molecules and cellular pathways that target specific phases of insulin secretion, resulting in a better understanding of insulin secretion, which in turn will result in a more effective insulin therapy through the stimulation of endogenous glucose-stimulated insulin secretion.
Collapse
Affiliation(s)
- Sikha Thoduvayil
- Nanobiology Institute, Yale University School of Medicine, West Haven, CT, 06516 USA; Department of Pathology, Yale University School of Medicine, New Haven, CT, 06520 USA
| | - Jonathan S Weerakkody
- Nanobiology Institute, Yale University School of Medicine, West Haven, CT, 06516 USA; Department of Pathology, Yale University School of Medicine, New Haven, CT, 06520 USA
| | - Ramalingam Venkat Kalyana Sundaram
- Nanobiology Institute, Yale University School of Medicine, West Haven, CT, 06516 USA; Department of Cell Biology, Yale University School of Medicine, New Haven, CT, 06520 USA
| | - Mackenzie Topper
- Nanobiology Institute, Yale University School of Medicine, West Haven, CT, 06516 USA
| | - Manindra Bera
- Nanobiology Institute, Yale University School of Medicine, West Haven, CT, 06516 USA; Department of Cell Biology, Yale University School of Medicine, New Haven, CT, 06520 USA
| | - Jeff Coleman
- Nanobiology Institute, Yale University School of Medicine, West Haven, CT, 06516 USA; Department of Cell Biology, Yale University School of Medicine, New Haven, CT, 06520 USA
| | - Xia Li
- Nanobiology Institute, Yale University School of Medicine, West Haven, CT, 06516 USA; Department of Cell Biology, Yale University School of Medicine, New Haven, CT, 06520 USA
| | - Malaiyalam Mariappan
- Nanobiology Institute, Yale University School of Medicine, West Haven, CT, 06516 USA; Department of Cell Biology, Yale University School of Medicine, New Haven, CT, 06520 USA
| | - Sathish Ramakrishnan
- Nanobiology Institute, Yale University School of Medicine, West Haven, CT, 06516 USA; Department of Pathology, Yale University School of Medicine, New Haven, CT, 06520 USA.
| |
Collapse
|
10
|
Raval KY, Tirgar PR. A pre-clinical study to investigate the anti-diabetic potential of p-propoxybenzoic acid as a multi-target inhibitor in streptozotocin-nicotinamide induced type-2 diabetic rats. J Diabetes Metab Disord 2023; 22:571-580. [PMID: 37255789 PMCID: PMC10225424 DOI: 10.1007/s40200-022-01177-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/12/2022] [Accepted: 12/25/2022] [Indexed: 06/01/2023]
Abstract
Purpose The study was undertaken to evaluate the anti-diabetic potential of p-propoxybenzoic acid (p-PBA). Methods 36 Sprague-Dawley rats of either sex were utilized for the study. Animals were injected with nicotinamide (230 mg/kg) followed by streptozotocin (65 mg/kg) to induce Type-2 Diabetes (T2DM). Animals with blood glucose levels (BGL) over 200 mg/kg were allocated in six groups. Three groups were treated with p-PBA dose of 100 mg/kg, 200 mg/kg and 300 mg/kg respectively; standard control group was treated with 5 mg/kg glibenclamide, while the other two groups were considered as normal control and disease control group. Body weight (BW) and BGL were recorded on Day 0, Day 7, Day 14, and Day 28. Glycosylated hemoglobin (HbA1c), serum insulin levels and lipid profile were recorded on Day 28. Animals were euthanized on Day 28 and the pancreas was isolated for histopathological examination. Results Diabetic animals treated with p-PBA showed significant improvements in BW (P < 0.05) and BGL (P < 0.001) over 28 days. Levels of HbA1c (P < 0.05) and serum insulin (P < 0.001) were significantly regulated in animals treated with p-PBA. A significant decrease (P < 0.001) was observed in elevated levels of TC, TG, LDL cholesterol and VLDL cholesterol in animals treated with p-PBA. p-PBA significantly regulated the levels of HDL cholesterol (P < 0.001). A notable protective effect of p-PBA was observed through the histopathological examination of pancreas. Conclusion p-PBA can be characterized as a multi-target inhibiting anti-diabetic agent which can be evaluated against diabetic complications. Supplementary Information The online version contains supplementary material available at 10.1007/s40200-022-01177-y.
Collapse
Affiliation(s)
- Keval Y. Raval
- School of Pharmacy, R K University, Rajkot, Gujarat India
| | | |
Collapse
|
11
|
You-xiang C, Lin Z. Nomogram model for the risk of insulin resistance in obese children and adolescents based on anthropomorphology and lipid derived indicators. BMC Public Health 2023; 23:275. [PMID: 36750783 PMCID: PMC9906839 DOI: 10.1186/s12889-023-15181-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2022] [Accepted: 02/01/2023] [Indexed: 02/09/2023] Open
Abstract
OBJECTIVE This study aims to screen for measures and lipid-derived indicators associated with insulin resistance (IR) in obese children and adolescents and develop a nomogram model for predicting the risk of insulin resistance. METHODS A total of 404 eligible obese children and adolescents aged 10-17 years were recruited for this study from a summer camp between 2019 and 2021. The risk factors were screened using the least absolute shrinkage and selection operator (LASSO)-logistic regression model, and a nomogram model was developed. The diagnostic value of the model was evaluated by plotting the receiver operator characteristic curve and calculating the area under the curve. Internal validation was performed using the Bootstrap method, with 1000 self-samples to evaluate the model stability. The clinical applicability of the model was assessed by plotting the clinical decision curve. RESULTS On the basis of the LASSO regression analysis results, three lipid-related derivatives, TG/HDL-c, TC/HDL-c, and LDL-c/HDL-c, were finally included in the IR risk prediction model. The nomogram model AUC was 0.804 (95% CI: 0.760 to 0.849). Internal validation results show a C-Index of 0.799, and the mean absolute error between the predicted and actual risks of IR was 0.015. The results of the Hosmer-Lemeshow goodness-of-fit test show a good model prediction (χ2 = 9.523, P = 0.300). CONCLUSION Three early warning factors, TG/HDL-c, TC/HDL-c, and LDL-c/HDL-c, were screened, which can effectively predict the risk of developing IR in obese children and adolescents, and the nomogram model has an eligible diagnostic value.
Collapse
Affiliation(s)
- Cao You-xiang
- grid.443378.f0000 0001 0483 836XGraduate Department, Guangzhou Sport University, Guangzhou, Guangdong Province China
| | - Zhu Lin
- School of Sport & Health, Guangzhou Sport University, No. 1268, Guangzhou Avenue Middle, Tianhe District, Guangzhou City, Guangdong Province, China.
| |
Collapse
|
12
|
Freyberg Z, Gittes GK. Roles of Pancreatic Islet Catecholamine Neurotransmitters in Glycemic Control and in Antipsychotic Drug-Induced Dysglycemia. Diabetes 2023; 72:3-15. [PMID: 36538602 PMCID: PMC9797319 DOI: 10.2337/db22-0522] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/09/2022] [Accepted: 09/24/2022] [Indexed: 12/24/2022]
Abstract
Catecholamine neurotransmitters dopamine (DA) and norepinephrine (NE) are essential for a myriad of functions throughout the central nervous system, including metabolic regulation. These molecules are also present in the pancreas, and their study may shed light on the effects of peripheral neurotransmission on glycemic control. Though sympathetic innervation to islets provides NE that signals at local α-cell and β-cell adrenergic receptors to modify hormone secretion, α-cells and β-cells also synthesize catecholamines locally. We propose a model where α-cells and β-cells take up catecholamine precursors in response to postprandial availability, preferentially synthesizing DA. The newly synthesized DA signals in an autocrine/paracrine manner to regulate insulin and glucagon secretion and maintain glycemic control. This enables islets to couple local catecholamine signaling to changes in nutritional state. We also contend that the DA receptors expressed by α-cells and β-cells are targeted by antipsychotic drugs (APDs)-some of the most widely prescribed medications today. Blockade of local DA signaling contributes significantly to APD-induced dysglycemia, a major contributor to treatment discontinuation and development of diabetes. Thus, elucidating the peripheral actions of catecholamines will provide new insights into the regulation of metabolic pathways and may lead to novel, more effective strategies to tune metabolism and treat diabetes.
Collapse
Affiliation(s)
- Zachary Freyberg
- Department of Psychiatry, University of Pittsburgh, Pittsburgh, PA
- Department of Cell Biology, University of Pittsburgh, Pittsburgh, PA
| | - George K. Gittes
- Division of Pediatric Surgery, UPMC Children’s Hospital of Pittsburgh, Pittsburgh, PA
| |
Collapse
|
13
|
Emami J, Haghighi M, Rostami M, Minaiyan M. Development and validation of a new robust RP-HPLC method for simultaneous quantitation of insulin and pramlintide in non-invasive and smart glucose-responsive microparticles. Res Pharm Sci 2022; 17:594-611. [PMID: 36704426 PMCID: PMC9872181 DOI: 10.4103/1735-5362.359428] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2022] [Revised: 09/18/2022] [Accepted: 10/01/2022] [Indexed: 11/06/2022] Open
Abstract
Background and purpose Since insulin and pramlintide cooperate in glucose hemostasis, co-administration and quantitation of them in pharmaceutical preparations are imperative. A simple, rapid, sensitive, and isocratic RP-HPLC method was developed and validated for simultaneous quantitation of insulin and pramlintide in loading and in-vitro release studies of a glucose-responsive system to improve the control of hyperglycemic episodes in diabetic patients. Experimental approach The isocratic RP-HPLC separation was achieved on a C18 µ-Bondopak column (250 mm × 4.6 mm) using a mobile phase of water:acetonitrile:trifluoroacetic acid (65:35:0.1%) at a flow rate of 1 mL/min in an ambient temperature. Both proteins were detected using a UV detector at 214 nm. The method was validated for specificity, linearity, precision, accuracy, the limit of detection, the limit of quantification, and robustness. Findings/Results Linearity was obtained in the concentration range of 30 to 360 μg/mL for insulin and 1.5 to 12 μg/mL for pramlintide. The results were validated statistically and recovery studies confirmed the great accuracy and precision of the proposed method. The robustness of the method was also confirmed through small changes in pH, mobile phase composition, and flow rate. Conclusion and implications The method was found to be simple, specific, precise, and reproducible. It was applied for the determination of loading capacity, entrapment efficiency, and in-vitro release studies of insulin and pramlintide in a smart glucose-responsive microparticle. Co-delivery of insulin and pramlintide could be a new intervention in diabetes management and concurrent quantitation of these two proteins is, therefore, essential.
Collapse
Affiliation(s)
- Jaber Emami
- Department of Pharmaceutics, School of Pharmacy and Pharmaceutical Sciences, Isfahan University of Medical Sciences, Isfahan, I.R. Iran,Corresponding author: J. Emami Tel: +98-3137927111, Fax: +98-3136680011
| | - Maryam Haghighi
- Department of Pharmaceutics, School of Pharmacy and Pharmaceutical Sciences, Isfahan University of Medical Sciences, Isfahan, I.R. Iran
| | - Mahboobeh Rostami
- Department of Medical Chemistry, School of Pharmacy and Pharmaceutical Sciences, Isfahan University of Medical Sciences, Isfahan, I.R. Iran
| | - Mohsen Minaiyan
- Department of Pharmacology and Toxicology, School of Pharmacy and Pharmaceutical Sciences, Isfahan University of Medical Sciences, Isfahan, I.R. Iran
| |
Collapse
|
14
|
Aslanoglou D, Bertera S, Friggeri L, Sánchez-Soto M, Lee J, Xue X, Logan RW, Lane JR, Yechoor VK, McCormick PJ, Meiler J, Free RB, Sibley DR, Bottino R, Freyberg Z. Dual pancreatic adrenergic and dopaminergic signaling as a therapeutic target of bromocriptine. iScience 2022; 25:104771. [PMID: 35982797 PMCID: PMC9379584 DOI: 10.1016/j.isci.2022.104771] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2021] [Revised: 06/10/2022] [Accepted: 07/11/2022] [Indexed: 11/23/2022] Open
Abstract
Bromocriptine is approved as a diabetes therapy, yet its therapeutic mechanisms remain unclear. Though bromocriptine's actions have been mainly attributed to the stimulation of brain dopamine D2 receptors (D2R), bromocriptine also targets the pancreas. Here, we employ bromocriptine as a tool to elucidate the roles of catecholamine signaling in regulating pancreatic hormone secretion. In β-cells, bromocriptine acts on D2R and α2A-adrenergic receptor (α2A-AR) to reduce glucose-stimulated insulin secretion (GSIS). Moreover, in α-cells, bromocriptine acts via D2R to reduce glucagon secretion. α2A-AR activation by bromocriptine recruits an ensemble of G proteins with no β-arrestin2 recruitment. In contrast, D2R recruits G proteins and β-arrestin2 upon bromocriptine stimulation, demonstrating receptor-specific signaling. Docking studies reveal distinct bromocriptine binding to α2A-AR versus D2R, providing a structural basis for bromocriptine's dual actions on β-cell α2A-AR and D2R. Together, joint dopaminergic and adrenergic receptor actions on α-cell and β-cell hormone release provide a new therapeutic mechanism to improve dysglycemia.
Collapse
Affiliation(s)
- Despoina Aslanoglou
- Translational Neuroscience Program, Department of Psychiatry, University of Pittsburgh, Pittsburgh, PA, USA
| | - Suzanne Bertera
- Institute of Cellular Therapeutics, Allegheny Health Network Research Institute, Allegheny Health Network, Pittsburgh, PA, USA
| | - Laura Friggeri
- Department of Chemistry, Center for Structural Biology, Vanderbilt University, Nashville, TN, USA
| | - Marta Sánchez-Soto
- Molecular Neuropharmacology Section, National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD, USA
| | - Jeongkyung Lee
- Diabetes and Beta Cell Biology Center, Division of Endocrinology, Diabetes and Metabolism, Department of Medicine, University of Pittsburgh, Pittsburgh, PA, USA
| | - Xiangning Xue
- Department of Biostatistics, University of Pittsburgh, Pittsburgh, PA, USA
| | - Ryan W. Logan
- Department of Pharmacology and Experimental Therapeutics, Boston University School of Medicine, Boston, MA, USA
| | - J. Robert Lane
- Division of Physiology, Pharmacology and Neuroscience, School of Life Sciences, Queen’s Medical Centre, University of Nottingham, Nottingham, UK
- Centre of Membrane Protein and Receptors, Universities of Birmingham and Nottingham, Nottingham, UK
| | - Vijay K. Yechoor
- Diabetes and Beta Cell Biology Center, Division of Endocrinology, Diabetes and Metabolism, Department of Medicine, University of Pittsburgh, Pittsburgh, PA, USA
| | - Peter J. McCormick
- Centre for Endocrinology, William Harvey Research Institute, Bart’s and the London School of Medicine and Dentistry, Queen Mary, University of London, London, UK
| | - Jens Meiler
- Department of Chemistry, Center for Structural Biology, Vanderbilt University, Nashville, TN, USA
- Institute for Drug Discovery, Leipzig University Medical School, Leipzig, Germany
| | - R. Benjamin Free
- Molecular Neuropharmacology Section, National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD, USA
| | - David R. Sibley
- Molecular Neuropharmacology Section, National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD, USA
| | - Rita Bottino
- Institute of Cellular Therapeutics, Allegheny Health Network Research Institute, Allegheny Health Network, Pittsburgh, PA, USA
- Imagine Pharma, Pittsburgh, PA, USA
| | - Zachary Freyberg
- Translational Neuroscience Program, Department of Psychiatry, University of Pittsburgh, Pittsburgh, PA, USA
- Department of Cell Biology, University of Pittsburgh, PA, USA
| |
Collapse
|
15
|
Nur Zati Iwani AK, Jalaludin MY, Yahya A, Mansor F, Md Zain F, Hong JYH, Wan Mohd Zin RM, Mokhtar AH. TG: HDL-C Ratio as Insulin Resistance Marker for Metabolic Syndrome in Children With Obesity. Front Endocrinol (Lausanne) 2022; 13:852290. [PMID: 35370951 PMCID: PMC8965646 DOI: 10.3389/fendo.2022.852290] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/11/2022] [Accepted: 02/14/2022] [Indexed: 01/28/2023] Open
Abstract
Insulin resistance (IR) is an important variable in the diagnosis of metabolic syndrome (MetS). Currently, IR is not part of the existing pediatric definition of MetS, instead elevated fasting blood glucose (FBG) is measured as an indicator of hyperglycemia. Arguably, many obese children with severe IR are still able to regulate their FBG well. Hence, this study aimed to assess the utility of triglyceride-to-high-density lipoprotein cholesterol (TG : HDL-C) ratio as an IR marker in the modeling of pediatric MetS among children with obesity using structural equation modeling (SEM). A total of 524 blood samples from children with obesity (age 10-16 years old) were analyzed for FBG, lipids, insulin, leptin, and adiponectin. Both exploratory (EFA) and confirmatory factor analysis (CFA) were used to examine TG : HDL-C ratio as an IR marker in pediatric MetS. EFA shows that TG: HDL-C ratio (standardized factor loading = 0.904) groups together with homeostasis model assessment-estimated insulin resistance (HOMA-IR) (standardized factor loading = 0.664), indicating a strong correlation to the IR factor. Replacing FBG with TG: HDL-C ratio improved the modeling of MetS structure in children with obesity. Our MetS model of TG: HDL-C ratio as IR component shows comparable model fitness indices (goodness of fit, Akaike's information criterion, and Bayesian information criterion) with leptin:adiponectin ratio (platinum standard for adiposity:IR marker) model. The least model fit was seen when using FBG as an IR surrogate. TG : HDL-C ratio performed better as IR surrogate in MetS structures (standardized factor loading = 0.39) compared to FBG (standardized factor loading = 0.27). TG: HDL-C ratio may be considered as an IR component in pediatric MetS.
Collapse
Affiliation(s)
- Ahmad Kamil Nur Zati Iwani
- Endocrine and Metabolic Unit, Institute for Medical Research, Ministry of Health Malaysia, Setia Alam, Malaysia
| | - Muhammad Yazid Jalaludin
- Department of Pediatrics, Faculty of Medicine, University Malaya, Kuala Lumpur, Malaysia
- *Correspondence: Muhammad Yazid Jalaludin, ;
| | - Abqariyah Yahya
- Department of Social and Preventive Medicine, Faculty of Medicine, University Malaya, Kuala Lumpur, Malaysia
| | - Fazliana Mansor
- Endocrine and Metabolic Unit, Institute for Medical Research, Ministry of Health Malaysia, Setia Alam, Malaysia
| | - Fuziah Md Zain
- Department of Pediatrics, Hospital Putrajaya, Ministry of Health Malaysia, Putrajaya, Malaysia
| | - Janet Yeow Hua Hong
- Department of Pediatrics, Hospital Putrajaya, Ministry of Health Malaysia, Putrajaya, Malaysia
| | - Ruziana Mona Wan Mohd Zin
- Endocrine and Metabolic Unit, Institute for Medical Research, Ministry of Health Malaysia, Setia Alam, Malaysia
| | - Abdul Halim Mokhtar
- Department of Sports Medicine, Faculty of Medicine, University Malaya, Kuala Lumpur, Malaysia
| |
Collapse
|
16
|
Pereira LMB, Cali MP, Marchi RC, Pazin WM, Carlos RM. Luminescent imaging of insulin amyloid aggregation using a sensitive ruthenium-based probe in the red region. J Inorg Biochem 2021; 224:111585. [PMID: 34450412 DOI: 10.1016/j.jinorgbio.2021.111585] [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: 06/09/2021] [Revised: 08/13/2021] [Accepted: 08/15/2021] [Indexed: 10/20/2022]
Abstract
A sensitive and selective strategy to identify insulin fibrils remains a challenge for researchers in amyloid protein research. Thus, it is critical to detect, in vitro, the species generated during amyloid aggregation, particularly the fibrillar species. Here we demonstrate that the luminescent complex cis-[Ru(phen)2(3,4Apy)2]2+ (RuApy; phen = 1,10-phenanthroline; 3,4Apy = 3,4-diaminopyridine) is a rapid, low-cost alternative to in vitro detection of fibrillar insulin, using conventional optical techniques. The RuApy complex displays emission intensity enhancement at 655 nm when associated with insulin, which enables imaging of the conformational changes of the protein's self-aggregation. The complex shows high sensitivity to fibrillar insulin with a limit of detection of 0.85 μM and binding affinity of 12.40 ± 1.84 μM which is comparable to those of Thioflavin T and Congo red, with the advantage of minimizing background fluorescence, absorption of light by biomolecules, and light scattering from physiologic salts in the medium.
Collapse
Affiliation(s)
- Lorena M B Pereira
- Department of Chemistry, Universidade Federal de São Carlos, São Carlos, São Paulo 13565-905, Brazil
| | - Mariana P Cali
- Department of Chemistry, Universidade Federal de São Carlos, São Carlos, São Paulo 13565-905, Brazil
| | - Rafael C Marchi
- Department of Chemistry, Universidade Federal de São Carlos, São Carlos, São Paulo 13565-905, Brazil
| | - Wallance M Pazin
- Department of Physics, Universidade Estadual Paulista, Presidente Prudente, São Paulo 19060-900, Brazil
| | - Rose M Carlos
- Department of Chemistry, Universidade Federal de São Carlos, São Carlos, São Paulo 13565-905, Brazil.
| |
Collapse
|
17
|
Aslanoglou D, Bertera S, Sánchez-Soto M, Benjamin Free R, Lee J, Zong W, Xue X, Shrestha S, Brissova M, Logan RW, Wollheim CB, Trucco M, Yechoor VK, Sibley DR, Bottino R, Freyberg Z. Dopamine regulates pancreatic glucagon and insulin secretion via adrenergic and dopaminergic receptors. Transl Psychiatry 2021; 11:59. [PMID: 33589583 PMCID: PMC7884786 DOI: 10.1038/s41398-020-01171-z] [Citation(s) in RCA: 63] [Impact Index Per Article: 15.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/30/2020] [Revised: 10/13/2020] [Accepted: 10/26/2020] [Indexed: 01/14/2023] Open
Abstract
Dopamine (DA) and norepinephrine (NE) are catecholamines primarily studied in the central nervous system that also act in the pancreas as peripheral regulators of metabolism. Pancreatic catecholamine signaling has also been increasingly implicated as a mechanism responsible for the metabolic disturbances produced by antipsychotic drugs (APDs). Critically, however, the mechanisms by which catecholamines modulate pancreatic hormone release are not completely understood. We show that human and mouse pancreatic α- and β-cells express the catecholamine biosynthetic and signaling machinery, and that α-cells synthesize DA de novo. This locally-produced pancreatic DA signals via both α- and β-cell adrenergic and dopaminergic receptors with different affinities to regulate glucagon and insulin release. Significantly, we show DA functions as a biased agonist at α2A-adrenergic receptors, preferentially signaling via the canonical G protein-mediated pathway. Our findings highlight the interplay between DA and NE signaling as a novel form of regulation to modulate pancreatic hormone release. Lastly, pharmacological blockade of DA D2-like receptors in human islets with APDs significantly raises insulin and glucagon release. This offers a new mechanism where APDs act directly on islet α- and β-cell targets to produce metabolic disturbances.
Collapse
Affiliation(s)
- Despoina Aslanoglou
- grid.21925.3d0000 0004 1936 9000Translational Neuroscience Program, Department of Psychiatry, University of Pittsburgh, Pittsburgh, PA USA
| | - Suzanne Bertera
- grid.417046.00000 0004 0454 5075Institute of Cellular Therapeutics, Allegheny Health Network Research Institute, Allegheny Health Network, Pittsburgh, PA USA
| | - Marta Sánchez-Soto
- grid.94365.3d0000 0001 2297 5165Molecular Neuropharmacology Section, National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD USA
| | - R. Benjamin Free
- grid.94365.3d0000 0001 2297 5165Molecular Neuropharmacology Section, National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD USA
| | - Jeongkyung Lee
- grid.21925.3d0000 0004 1936 9000Division of Endocrinology, Diabetes & Metabolism, Department of Medicine, Diabetes and Beta Cell Biology Center, University of Pittsburgh, Pittsburgh, PA USA
| | - Wei Zong
- grid.21925.3d0000 0004 1936 9000Department of Biostatistics, University of Pittsburgh, Pittsburgh, PA USA
| | - Xiangning Xue
- grid.21925.3d0000 0004 1936 9000Department of Biostatistics, University of Pittsburgh, Pittsburgh, PA USA
| | - Shristi Shrestha
- grid.412807.80000 0004 1936 9916Division of Diabetes, Endocrinology, and Metabolism, Department of Medicine, Vanderbilt University Medical Center, Nashville, TN USA
| | - Marcela Brissova
- grid.412807.80000 0004 1936 9916Division of Diabetes, Endocrinology, and Metabolism, Department of Medicine, Vanderbilt University Medical Center, Nashville, TN USA
| | - Ryan W. Logan
- grid.21925.3d0000 0004 1936 9000Translational Neuroscience Program, Department of Psychiatry, University of Pittsburgh, Pittsburgh, PA USA ,grid.249880.f0000 0004 0374 0039Center for Systems Neurogenetics of Addiction, The Jackson Laboratory, Bar Harbor, ME USA
| | - Claes B. Wollheim
- grid.8591.50000 0001 2322 4988Department of Cell Physiology and Metabolism, University of Geneva, Geneva, Switzerland
| | - Massimo Trucco
- grid.417046.00000 0004 0454 5075Institute of Cellular Therapeutics, Allegheny Health Network Research Institute, Allegheny Health Network, Pittsburgh, PA USA ,grid.147455.60000 0001 2097 0344Department of Biological Sciences, Carnegie Mellon University, Pittsburgh, PA USA ,grid.166341.70000 0001 2181 3113College of Medicine, Drexel University, Philadelphia, PA USA
| | - Vijay K. Yechoor
- grid.21925.3d0000 0004 1936 9000Division of Endocrinology, Diabetes & Metabolism, Department of Medicine, Diabetes and Beta Cell Biology Center, University of Pittsburgh, Pittsburgh, PA USA
| | - David R. Sibley
- grid.94365.3d0000 0001 2297 5165Molecular Neuropharmacology Section, National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD USA
| | - Rita Bottino
- grid.417046.00000 0004 0454 5075Institute of Cellular Therapeutics, Allegheny Health Network Research Institute, Allegheny Health Network, Pittsburgh, PA USA ,grid.147455.60000 0001 2097 0344Department of Biological Sciences, Carnegie Mellon University, Pittsburgh, PA USA ,grid.166341.70000 0001 2181 3113College of Medicine, Drexel University, Philadelphia, PA USA
| | - Zachary Freyberg
- Translational Neuroscience Program, Department of Psychiatry, University of Pittsburgh, Pittsburgh, PA, USA. .,Department of Cell Biology, University of Pittsburgh, Pittsburgh, PA, USA.
| |
Collapse
|
18
|
Tapia-Alveal C, Olsen TR, Worgall TS. Personalized immunoglobulin aptamers for detection of multiple myeloma minimal residual disease in serum. Commun Biol 2020; 3:781. [PMID: 33335255 PMCID: PMC7747622 DOI: 10.1038/s42003-020-01515-x] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2020] [Accepted: 11/20/2020] [Indexed: 01/21/2023] Open
Abstract
Multiple myeloma (MM) is a neoplasm of plasma cells that secrete patient specific monoclonal immunoglobulins. A recognized problem in MM treatment is the early recognition of minimal residual disease (MRD), the major cause of relapse. Current MRD detection methods (multiparameter flow cytometry and next generation sequencing) are based on the analysis of bone marrow plasma cells. Both methods cannot detect extramedullary disease and are unsuitable for serial measurements. We describe the methodology to generate high affinity DNA aptamers that are specific to a patient’s monoclonal Fab region. Such aptamers are 2000-fold more sensitive than immunofixation electrophoresis and enabled detection and quantification of MRD in serum when conventional MRD methods assessed complete remission. The aptamer isolation process that requires small volumes of serum is automatable, and Fab specific aptamers are adaptable to multiple diagnostic formats including point-of-care devices. Tapia-Alveal, Olsen and Worgall develop a novel strategy for patient-specific multiple myeloma diagnostics platform using DNA aptamers. The high affinity DNA aptamers enabled detection of minimal residual disease (MRD) when conventional MRD methods assessed complete remission and are adaptable to multiple diagnostic formats including point-of-care devices.
Collapse
Affiliation(s)
- Claudia Tapia-Alveal
- Department of Pathology and Cell Biology, Columbia University Irving Medical Center, New York, NY, 10032, USA
| | - Timothy R Olsen
- Department of Pathology and Cell Biology, Columbia University Irving Medical Center, New York, NY, 10032, USA
| | - Tilla S Worgall
- Department of Pathology and Cell Biology, Columbia University Irving Medical Center, New York, NY, 10032, USA.
| |
Collapse
|
19
|
Farino ZJ, Morgenstern TJ, Maffei A, Quick M, De Solis AJ, Wiriyasermkul P, Freyberg RJ, Aslanoglou D, Sorisio D, Inbar BP, Free RB, Donthamsetti P, Mosharov EV, Kellendonk C, Schwartz GJ, Sibley DR, Schmauss C, Zeltser LM, Moore H, Harris PE, Javitch JA, Freyberg Z. New roles for dopamine D 2 and D 3 receptors in pancreatic beta cell insulin secretion. Mol Psychiatry 2020; 25:2070-2085. [PMID: 30626912 PMCID: PMC6616020 DOI: 10.1038/s41380-018-0344-6] [Citation(s) in RCA: 53] [Impact Index Per Article: 10.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/12/2018] [Revised: 09/17/2018] [Accepted: 12/10/2018] [Indexed: 01/11/2023]
Abstract
Although long-studied in the central nervous system, there is increasing evidence that dopamine (DA) has important roles in the periphery including in metabolic regulation. Insulin-secreting pancreatic β-cells express the machinery for DA synthesis and catabolism, as well as all five DA receptors. In these cells, DA functions as a negative regulator of glucose-stimulated insulin secretion (GSIS), which is mediated by DA D2-like receptors including D2 (D2R) and D3 (D3R) receptors. However, the fundamental mechanisms of DA synthesis, storage, release, and signaling in pancreatic β-cells and their functional relevance in vivo remain poorly understood. Here, we assessed the roles of the DA precursor L-DOPA in β-cell DA synthesis and release in conjunction with the signaling mechanisms underlying DA's inhibition of GSIS. Our results show that the uptake of L-DOPA is essential for establishing intracellular DA stores in β-cells. Glucose stimulation significantly enhances L-DOPA uptake, leading to increased DA release and GSIS reduction in an autocrine/paracrine manner. Furthermore, D2R and D3R act in combination to mediate dopaminergic inhibition of GSIS. Transgenic knockout mice in which β-cell D2R or D3R expression is eliminated exhibit diminished DA secretion during glucose stimulation, suggesting a new mechanism where D2-like receptors modify DA release to modulate GSIS. Lastly, β-cell-selective D2R knockout mice exhibit marked postprandial hyperinsulinemia in vivo. These results reveal that peripheral D2R and D3R receptors play important roles in metabolism through their inhibitory effects on GSIS. This opens the possibility that blockade of peripheral D2-like receptors by drugs including antipsychotic medications may significantly contribute to the metabolic disturbances observed clinically.
Collapse
Affiliation(s)
- Zachary J. Farino
- Department of Psychiatry, University of Pittsburgh, Pittsburgh, PA, USA
| | - Travis J. Morgenstern
- Department of Psychiatry, College of Physicians & Surgeons, Columbia University, New York, NY, USA
| | - Antonella Maffei
- Division of Endocrinology, Department of Medicine, College of Physicians & Surgeons, Columbia University, New York, NY, USA
| | - Matthias Quick
- Department of Psychiatry, College of Physicians & Surgeons, Columbia University, New York, NY, USA,Division of Molecular Therapeutics, New York State Psychiatric Institute, New York, NY, USA
| | - Alain J. De Solis
- Division of Molecular Genetics, Naomi Berrie Diabetes Center, Columbia University, New York, NY, USA
| | - Pattama Wiriyasermkul
- Department of Psychiatry, College of Physicians & Surgeons, Columbia University, New York, NY, USA,Current address: Department of Collaborative Research, Nara Medical University, Kashihara, Nara, Japan
| | - Robin J. Freyberg
- Department of Psychiatry, University of Pittsburgh, Pittsburgh, PA, USA
| | | | - Denise Sorisio
- Department of Psychiatry, University of Pittsburgh, Pittsburgh, PA, USA
| | - Benjamin P. Inbar
- Department of Psychiatry, College of Physicians & Surgeons, Columbia University, New York, NY, USA
| | - R. Benjamin Free
- Molecular Neuropharmacology Section, National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD, USA
| | - Prashant Donthamsetti
- Department of Psychiatry, College of Physicians & Surgeons, Columbia University, New York, NY, USA,Current address: Department of Molecular and Cell Biology, University of California, Berkeley, CA, USA
| | - Eugene V. Mosharov
- Department of Psychiatry, College of Physicians & Surgeons, Columbia University, New York, NY, USA,Division of Molecular Therapeutics, New York State Psychiatric Institute, New York, NY, USA,Department of Neurology, College of Physicians & Surgeons, Columbia University, New York, NY, USA
| | - Christoph Kellendonk
- Department of Psychiatry, College of Physicians & Surgeons, Columbia University, New York, NY, USA,Division of Molecular Therapeutics, New York State Psychiatric Institute, New York, NY, USA,Department of Pharmacology, College of Physicians & Surgeons, Columbia University, New York, NY, USA
| | - Gary J. Schwartz
- Departments of Medicine and Neuroscience, Albert Einstein College of Medicine, Bronx, NY, USA
| | - David R. Sibley
- Molecular Neuropharmacology Section, National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD, USA
| | - Claudia Schmauss
- Department of Psychiatry, College of Physicians & Surgeons, Columbia University, New York, NY, USA,Division of Molecular Therapeutics, New York State Psychiatric Institute, New York, NY, USA
| | - Lori M. Zeltser
- Division of Molecular Genetics, Naomi Berrie Diabetes Center, Columbia University, New York, NY, USA,Department of Pathology and Cell Biology, College of Physicians & Surgeons, Columbia University, New York, NY, USA
| | - Holly Moore
- Department of Psychiatry, College of Physicians & Surgeons, Columbia University, New York, NY, USA,Division of Integrative Neuroscience, New York State Psychiatric Institute, New York, NY, USA
| | - Paul E. Harris
- Division of Endocrinology, Department of Medicine, College of Physicians & Surgeons, Columbia University, New York, NY, USA
| | - Jonathan A. Javitch
- Department of Psychiatry, College of Physicians & Surgeons, Columbia University, New York, NY, USA,Division of Molecular Therapeutics, New York State Psychiatric Institute, New York, NY, USA,Department of Pharmacology, College of Physicians & Surgeons, Columbia University, New York, NY, USA
| | - Zachary Freyberg
- Department of Psychiatry, University of Pittsburgh, Pittsburgh, PA, USA. .,Department of Cell Biology, University of Pittsburgh, Pittsburgh, PA, USA.
| |
Collapse
|
20
|
Wei H, Zapata RC, Lopez-Valencia M, Aslanoglou D, Farino ZJ, Benner V, Osborn O, Freyberg Z, McCarthy MJ. Dopamine D 2 receptor signaling modulates pancreatic beta cell circadian rhythms. Psychoneuroendocrinology 2020; 113:104551. [PMID: 31884319 PMCID: PMC7787223 DOI: 10.1016/j.psyneuen.2019.104551] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/12/2019] [Revised: 11/14/2019] [Accepted: 12/13/2019] [Indexed: 01/01/2023]
Abstract
Antipsychotic drugs (APD) have clinically important, adverse effects on metabolism that limit their therapeutic utility. Pancreatic beta cells produce dopamine and express the D2 dopamine receptor (D2R). As D2R antagonists, APDs alter glucose-stimulated insulin secretion, indicating that dopamine likely plays a role in APD-induced metabolic dysfunction. Insulin secretion from beta cells is also modulated by the circadian clock. Disturbed circadian rhythms cause metabolic disturbances similar to those observed in APD-treated subjects. Given the importance of dopamine and circadian rhythms for beta cells, we hypothesized that the beta cell dopamine system and circadian clock interact and dually regulate insulin secretion, and that circadian manipulations may alter the metabolic impact of APDs. We measured circadian rhythms, insulin release, and the impact of dopamine upon these processes in beta cells using bioluminescent reporters. We then assessed the impact of circadian timing on weight gain and metabolic outcomes in mice treated with the APD sulpiride at the onset of light or dark. We found that molecular components of the dopamine system were rhythmically expressed in beta cells. D2R stimulation by endogenous dopamine or the agonist bromocriptine reduced circadian rhythm amplitude, and altered the temporal profile of insulin secretion. Sulpiride caused greater weight gain and hyperinsulinemia in mice when given in the dark phase compared to the light phase. D2R-acting drugs affect circadian-dopamine interactions and modulate beta cell metabolic function. These findings identify circadian timing as a novel and important mechanism underlying APD-induced metabolic dysfunction, offering new possibilities for therapeutic interventions.
Collapse
Affiliation(s)
- Heather Wei
- Psychiatry Service, VA San Diego Healthcare, San Diego, CA 92161, USA
| | - Rizaldy C. Zapata
- Department of Medicine, University of California, San Diego, San Diego, CA 92093, USA
| | | | - Despoina Aslanoglou
- Department of Psychiatry, University of Pittsburgh, Pittsburgh, PA 15213, USA
| | - Zachary J. Farino
- Department of Psychiatry, University of Pittsburgh, Pittsburgh, PA 15213, USA
| | - Valerie Benner
- Psychiatry Service, VA San Diego Healthcare, San Diego, CA 92161, USA
| | - Olivia Osborn
- Department of Medicine, University of California, San Diego, San Diego, CA 92093, USA
| | - Zachary Freyberg
- Department of Psychiatry, University of Pittsburgh, Pittsburgh, PA 15213, USA; Department of Cell Biology, University of Pittsburgh, Pittsburgh, PA 15213, USA.
| | - Michael J. McCarthy
- Psychiatry Service, VA San Diego Healthcare, San Diego, CA 92161, USA,Department of Psychiatry and Center for Circadian Biology, University of California, San Diego, San Diego, CA 92161, USA,Corresponding author at: VA San Diego Healthcare System, 3350 La Jolla Village Dr MC116A, San Diego, CA 92161 USA
| |
Collapse
|
21
|
Han F, Konkalmatt P, Mokashi C, Kumar M, Zhang Y, Ko A, Farino ZJ, Asico LD, Xu G, Gildea J, Zheng X, Felder RA, Lee REC, Jose PA, Freyberg Z, Armando I. Dopamine D 2 receptor modulates Wnt expression and control of cell proliferation. Sci Rep 2019; 9:16861. [PMID: 31727925 PMCID: PMC6856370 DOI: 10.1038/s41598-019-52528-4] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2019] [Accepted: 10/17/2019] [Indexed: 01/06/2023] Open
Abstract
The Wnt/β-catenin pathway is one of the most conserved signaling pathways across species with essential roles in development, cell proliferation, and disease. Wnt signaling occurs at the protein level and via β-catenin-mediated transcription of target genes. However, little is known about the underlying mechanisms regulating the expression of the key Wnt ligand Wnt3a or the modulation of its activity. Here, we provide evidence that there is significant cross-talk between the dopamine D2 receptor (D2R) and Wnt/β-catenin signaling pathways. Our data suggest that D2R-dependent cross-talk modulates Wnt3a expression via an evolutionarily-conserved TCF/LEF site within the WNT3A promoter. Moreover, D2R signaling also modulates cell proliferation and modifies the pathology in a renal ischemia/reperfusion-injury disease model, via its effects on Wnt/β-catenin signaling. Together, our results suggest that D2R is a transcriptional modulator of Wnt/β-catenin signal transduction with broad implications for health and development of new therapeutics.
Collapse
MESH Headings
- Animals
- Cell Proliferation
- Dependovirus/genetics
- Dependovirus/metabolism
- Disease Models, Animal
- Embryo, Mammalian
- Epithelial Cells/metabolism
- Epithelial Cells/pathology
- Gene Expression Regulation
- Gene Knockdown Techniques
- Genetic Vectors/chemistry
- Genetic Vectors/metabolism
- Humans
- Kidney Tubules, Proximal/metabolism
- Kidney Tubules, Proximal/pathology
- Male
- Mice
- Mice, Inbred C57BL
- Primary Cell Culture
- Promoter Regions, Genetic
- RNA, Small Interfering/genetics
- RNA, Small Interfering/metabolism
- Receptors, Dopamine D2/genetics
- Receptors, Dopamine D2/metabolism
- Reperfusion Injury/genetics
- Reperfusion Injury/metabolism
- Reperfusion Injury/pathology
- Signal Transduction
- Transfection
- Wnt3A Protein/genetics
- Wnt3A Protein/metabolism
- beta Catenin/genetics
- beta Catenin/metabolism
Collapse
Affiliation(s)
- Fei Han
- Department of Medicine, School of Medicine and Health Sciences, The George Washington University, Washington, DC, 20052, USA
- Kidney Disease Center, First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, China
| | - Prasad Konkalmatt
- Department of Medicine, School of Medicine and Health Sciences, The George Washington University, Washington, DC, 20052, USA
| | - Chaitanya Mokashi
- Department of Computational & Systems Biology, University of Pittsburgh, Pittsburgh, PA, 15213, USA
| | - Megha Kumar
- Department of Medicine, School of Medicine and Health Sciences, The George Washington University, Washington, DC, 20052, USA
| | - Yanrong Zhang
- Department of Medicine, School of Medicine and Health Sciences, The George Washington University, Washington, DC, 20052, USA
| | - Allen Ko
- Institute of Human Nutrition, College of Physicians & Surgeons, Columbia University, New York, NY, 10032, USA
| | - Zachary J Farino
- Department of Psychiatry, University of Pittsburgh, Pittsburgh, PA, 15213, USA
| | - Laureano D Asico
- Department of Medicine, School of Medicine and Health Sciences, The George Washington University, Washington, DC, 20052, USA
| | - Gaosi Xu
- Department of Medicine, School of Medicine and Health Sciences, The George Washington University, Washington, DC, 20052, USA
| | - John Gildea
- Department of Pathology, The University of Virginia, Charlottesville, VA, 22904, USA
| | - Xiaoxu Zheng
- Department of Medicine, School of Medicine and Health Sciences, The George Washington University, Washington, DC, 20052, USA
| | - Robin A Felder
- Department of Pathology, The University of Virginia, Charlottesville, VA, 22904, USA
| | - Robin E C Lee
- Department of Computational & Systems Biology, University of Pittsburgh, Pittsburgh, PA, 15213, USA
| | - Pedro A Jose
- Department of Medicine, School of Medicine and Health Sciences, The George Washington University, Washington, DC, 20052, USA
- Department of Pharmacology and Physiology, School of Medicine and Health Sciences, The George Washington University, Washington, DC, 20052, USA
| | - Zachary Freyberg
- Department of Psychiatry, University of Pittsburgh, Pittsburgh, PA, 15213, USA.
- Department of Cell Biology, University of Pittsburgh, Pittsburgh, PA, 15213, USA.
| | - Ines Armando
- Department of Medicine, School of Medicine and Health Sciences, The George Washington University, Washington, DC, 20052, USA.
| |
Collapse
|
22
|
Merriman C, Fu D. Down-regulation of the islet-specific zinc transporter-8 (ZnT8) protects human insulinoma cells against inflammatory stress. J Biol Chem 2019; 294:16992-17006. [PMID: 31591269 DOI: 10.1074/jbc.ra119.010937] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2019] [Revised: 10/04/2019] [Indexed: 12/31/2022] Open
Abstract
Zinc transporter-8 (ZnT8) primarily functions as a zinc-sequestrating transporter in the insulin-secretory granules (ISGs) of pancreatic β-cells. Loss-of-function mutations in ZnT8 are associated with protection against type-2 diabetes (T2D), but the protective mechanism is unclear. Here, we developed an in-cell ZnT8 assay to track endogenous ZnT8 responses to metabolic and inflammatory stresses applied to human insulinoma EndoC-βH1 cells. Unexpectedly, high glucose and free fatty acids did not alter cellular ZnT8 levels, but proinflammatory cytokines acutely, reversibly, and gradually down-regulated ZnT8. Approximately 50% of the cellular ZnT8 was localized to the endoplasmic reticulum (ER), which was the primary target of the cytokine-mediated ZnT8 down-regulation. Transcriptome profiling of cytokine-exposed β-cells revealed an adaptive unfolded protein response (UPR) including a marked immunoproteasome activation that coordinately degraded ZnT8 and insulin over a 1,000-fold cytokine concentration range. RNAi-mediated ZnT8 knockdown protected cells against cytokine cytotoxicity, whereas inhibiting immunoproteasomes blocked cytokine-induced ZnT8 degradation and triggered a transition of the adaptive UPR to cell apoptosis. Hence, cytokine-induced down-regulation of the ER ZnT8 level promotes adaptive UPR, acting as a protective mechanism that decongests the ER burden of ZnT8 to protect β-cells from proapoptotic UPR during chronic low-grade inflammation.
Collapse
Affiliation(s)
- Chengfeng Merriman
- Department of Physiology, Johns Hopkins School of Medicine, Baltimore, Maryland 21205
| | - Dax Fu
- Department of Physiology, Johns Hopkins School of Medicine, Baltimore, Maryland 21205
| |
Collapse
|
23
|
Abstract
Insulin is an important polypeptide hormone that regulates carbohydrate metabolism.
Collapse
Affiliation(s)
- Yixiao Shen
- Department of Food Science
- Shenyang Agricultural University
- Shenyang
- China
| | - Witoon Prinyawiwatkul
- School of Nutrition and Food Sciences
- Louisiana State University
- Agricultural Center
- Baton Rouge
- USA
| | - Zhimin Xu
- School of Nutrition and Food Sciences
- Louisiana State University
- Agricultural Center
- Baton Rouge
- USA
| |
Collapse
|
24
|
Aslanoglou D, George EW, Freyberg Z. Homogeneous Time-resolved Förster Resonance Energy Transfer-based Assay for Detection of Insulin Secretion. J Vis Exp 2018. [PMID: 29806846 DOI: 10.3791/57531] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
The detection of insulin secretion is critical for elucidating mechanisms of regulated secretion as well as in studies of metabolism. Though numerous insulin assays have existed for decades, the recent advent of homogeneous time-resolved Förster Resonance Energy Transfer (HTRF) technology has significantly simplified these measurements. This is a rapid, cost-effective, reproducible, and robust optical assay reliant upon antibodies conjugated to bright fluorophores with long lasting emission which facilitates time-resolved Förster Resonance Energy Transfer. Moreover, HTRF insulin detection is amenable for the development of high-throughput screening assays. Here we use HTRF to detect insulin secretion in INS-1E cells, a rat insulinoma-derived cell line. This allows us to estimate basal levels of insulin and their changes in response to glucose stimulation. In addition, we use this insulin detection system to confirm the role of dopamine as a negative regulator of glucose-stimulated insulin secretion (GSIS). In a similar manner, other dopamine D2-like receptor agonists, quinpirole, and bromocriptine, reduce GSIS in a concentration-dependent manner. Our results highlight the utility of the HTRF insulin assay format in determining the role of numerous drugs in GSIS and their pharmacological profiles.
Collapse
Affiliation(s)
| | | | - Zachary Freyberg
- Department of Psychiatry, University of Pittsburgh; Department of Cell Biology, University of Pittsburgh;
| |
Collapse
|
25
|
Carter SD, Mageswaran SK, Farino ZJ, Mamede JI, Oikonomou CM, Hope TJ, Freyberg Z, Jensen GJ. Distinguishing signal from autofluorescence in cryogenic correlated light and electron microscopy of mammalian cells. J Struct Biol 2017; 201:15-25. [PMID: 29078993 DOI: 10.1016/j.jsb.2017.10.009] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2017] [Revised: 10/21/2017] [Accepted: 10/23/2017] [Indexed: 01/09/2023]
Abstract
In cryogenic correlated light and electron microscopy (cryo-CLEM), frozen targets of interest are identified and located on EM grids by fluorescence microscopy and then imaged at higher resolution by cryo-EM. Whilst working with these methods, we discovered that a variety of mammalian cells exhibit strong punctate autofluorescence when imaged under cryogenic conditions (80 K). Autofluorescence originated from multilamellar bodies (MLBs) and secretory granules. Here we describe a method to distinguish fluorescent protein tags from these autofluorescent sources based on the narrower emission spectrum of the former. The method is first tested on mitochondria and then applied to examine the ultrastructural variability of secretory granules within insulin-secreting pancreatic beta-cell-derived INS-1E cells.
Collapse
Affiliation(s)
- Stephen D Carter
- Division of Biology, California Institute of Technology, Pasadena, CA 91125, USA
| | - Shrawan K Mageswaran
- Division of Biology, California Institute of Technology, Pasadena, CA 91125, USA
| | - Zachary J Farino
- Department of Psychiatry, University of Pittsburgh, Pittsburgh, PA 15213, USA
| | - João I Mamede
- Department of Cell and Molecular Biology, Northwestern University, Chicago, IL 60611, USA
| | | | - Thomas J Hope
- Department of Cell and Molecular Biology, Northwestern University, Chicago, IL 60611, USA
| | - Zachary Freyberg
- Department of Psychiatry, University of Pittsburgh, Pittsburgh, PA 15213, USA; Department of Cell Biology, University of Pittsburgh, PA 15213, USA.
| | - Grant J Jensen
- Division of Biology, California Institute of Technology, Pasadena, CA 91125, USA; Howard Hughes Medical Institute (HHMI), California Institute of Technology, Pasadena, CA 91125, USA.
| |
Collapse
|
26
|
Freyberg Z, Aslanoglou D, Shah R, Ballon JS. Intrinsic and Antipsychotic Drug-Induced Metabolic Dysfunction in Schizophrenia. Front Neurosci 2017; 11:432. [PMID: 28804444 PMCID: PMC5532378 DOI: 10.3389/fnins.2017.00432] [Citation(s) in RCA: 51] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2017] [Accepted: 07/13/2017] [Indexed: 12/12/2022] Open
Abstract
For decades, there have been observations demonstrating significant metabolic disturbances in people with schizophrenia including clinically relevant weight gain, hypertension, and disturbances in glucose and lipid homeostasis. Many of these findings pre-date the use of antipsychotic drugs (APDs) which on their own are also strongly associated with metabolic side effects. The combination of APD-induced metabolic changes and common adverse environmental factors associated with schizophrenia have made it difficult to determine the specific contributions of each to the overall metabolic picture. Data from drug-naïve patients, both from the pre-APD era and more recently, suggest that there may be an intrinsic metabolic risk associated with schizophrenia. Nevertheless, these findings remain controversial due to significant clinical variability in both psychiatric and metabolic symptoms throughout patients' disease courses. Here, we provide an extensive review of classic and more recent literature describing the metabolic phenotype associated with schizophrenia. We also suggest potential mechanistic links between signaling pathways associated with schizophrenia and metabolic dysfunction. We propose that, beyond its symptomatology in the central nervous system, schizophrenia is also characterized by pathophysiology in other organ systems directly related to metabolic control.
Collapse
Affiliation(s)
- Zachary Freyberg
- Department of Psychiatry, University of PittsburghPittsburgh, PA, United States
- Department of Cell Biology, University of PittsburghPittsburgh, PA, United States
| | - Despoina Aslanoglou
- Department of Psychiatry, University of PittsburghPittsburgh, PA, United States
| | - Ripal Shah
- Department of Psychiatry and Behavioral Sciences, Stanford UniversityStanford, CA, United States
| | - Jacob S. Ballon
- Department of Psychiatry and Behavioral Sciences, Stanford UniversityStanford, CA, United States
| |
Collapse
|
27
|
Dopamine D 2 receptors and the circadian clock reciprocally mediate antipsychotic drug-induced metabolic disturbances. NPJ SCHIZOPHRENIA 2017; 3:17. [PMID: 28560263 PMCID: PMC5441531 DOI: 10.1038/s41537-017-0018-4] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/04/2017] [Revised: 03/01/2017] [Accepted: 03/08/2017] [Indexed: 01/11/2023]
Abstract
Antipsychotic drugs are widely prescribed medications, used for numerous psychiatric illnesses. However, antipsychotic drugs cause serious metabolic side effects that can lead to substantial weight gain and increased risk for type 2 diabetes. While individual drugs differ, all antipsychotic drugs may cause these important side effects to varying degrees. Given that the single unifying property shared by these medications is blockade of dopamine D2 and D3 receptors, these receptors likely play a role in antipsychotic drug-induced metabolic side effects. Dopamine D2 and dopamine D3 receptors are expressed in brain regions critical for metabolic regulation and appetite. Surprisingly, these receptors are also expressed peripherally in insulin-secreting pancreatic beta cells. By inhibiting glucose-stimulated insulin secretion, dopamine D2 and dopamine D3 receptors are important mediators of pancreatic insulin release. Crucially, antipsychotic drugs disrupt this peripheral metabolic regulatory mechanism. At the same time, disruptions to circadian timing have been increasingly recognized as a risk factor for metabolic disturbance. Reciprocal dopamine and circadian signaling is important for the timing of appetitive/feeding behaviors and insulin release, thereby coordinating cell metabolism with caloric intake. In particular, circadian regulation of dopamine D2 receptor/dopamine D3 receptor signaling may play a critical role in metabolism. Therefore, we propose that antipsychotic drugs’ blockade of dopamine D2 receptor and dopamine D3 receptors in pancreatic beta cells, hypothalamus, and striatum disrupts the cellular timing mechanisms that regulate metabolism. Ultimately, understanding the relationships between the dopamine system and circadian clocks may yield critical new biological insights into mechanisms of antipsychotic drug action, which can then be applied into clinical practice.
Collapse
|
28
|
Tagit O, Hildebrandt N. Fluorescence Sensing of Circulating Diagnostic Biomarkers Using Molecular Probes and Nanoparticles. ACS Sens 2017; 2:31-45. [PMID: 28722447 DOI: 10.1021/acssensors.6b00625] [Citation(s) in RCA: 65] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
The interplay of photonics, nanotechnology, and biochemistry has significantly improved the identification and characterization of multiple types of biomarkers by optical biosensors. Great achievements in fluorescence-based technologies have been realized, for example, by the advancement of multiplexing techniques or the introduction of nanoparticles to biochemical and clinical research. This review presents a concise overview of recent advances in fluorescence sensing techniques for the detection of circulating disease biomarkers. Detection principles of representative approaches, including fluorescence detection using molecular fluorophores, quantum dots, and metallic and silica nanoparticles, are explained and illustrated by pertinent examples from the recent literature. Advanced detection technologies and material development play a major role in modern biosensing and consistently provide significant improvements toward robust, sensitive, and versatile platforms for early detection of circulating diagnostic biomarkers.
Collapse
Affiliation(s)
- Oya Tagit
- NanoBioPhotonics
(nanofret.com), Institute for Integrative Biology of the Cell (I2BC), Université Paris-Saclay, Université Paris-Sud, CNRS, CEA, 91405 Orsay, France
- Department
of Tumor Immunology, Radboud Institute for Molecular Life Sciences, Radboud University Medical Center, 6500 HB Nijmegen, The Netherlands
| | - Niko Hildebrandt
- NanoBioPhotonics
(nanofret.com), Institute for Integrative Biology of the Cell (I2BC), Université Paris-Saclay, Université Paris-Sud, CNRS, CEA, 91405 Orsay, France
| |
Collapse
|
29
|
Li WH. Probes for monitoring regulated exocytosis. Cell Calcium 2017; 64:65-71. [PMID: 28089267 DOI: 10.1016/j.ceca.2017.01.002] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2016] [Accepted: 01/07/2017] [Indexed: 12/12/2022]
Abstract
Regulated secretion is a fundamental cellular process that serves diverse functions in neurobiology, endocrinology, immunology, and numerous other aspects of animal physiology. In response to environmental or biological cues, cells release contents of secretory granules into an extracellular medium to communicate with or impact neighboring or distant cells through paracrine or endocrine signaling. To investigate mechanisms governing stimulus-secretion coupling, to better understand how cells maintain or regulate their secretory activity, and to characterize secretion defects in human diseases, probes for tracking various exocytotic events at the cellular or sub-cellular level have been developed over the years. This review summarizes different strategies and recent progress in developing optical probes for monitoring regulated secretion in mammalian cells.
Collapse
Affiliation(s)
- Wen-Hong Li
- Departments of Cell Biology and of Biochemistry, University of Texas Southwestern Medical Center, 6000 Harry Hines Blvd., Dallas, TX 75390-9039, United States.
| |
Collapse
|
30
|
Chhasatia R, Sweetman MJ, Harding FJ, Waibel M, Kay T, Thomas H, Loudovaris T, Voelcker NH. Non-invasive, in vitro analysis of islet insulin production enabled by an optical porous silicon biosensor. Biosens Bioelectron 2017; 91:515-522. [PMID: 28082240 DOI: 10.1016/j.bios.2017.01.004] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2016] [Revised: 12/27/2016] [Accepted: 01/03/2017] [Indexed: 12/16/2022]
Abstract
A label-free porous silicon (pSi) based, optical biosensor, using both an antibody and aptamer bioreceptor motif has been developed for the detection of insulin. Two parallel biosensors were designed and optimised independently, based on each bioreceptor. Both bioreceptors were covalently attached to a thermally hydrosilylated pSi surface though amide coupling, with unreacted surface area rendered stable and low fouling by incorporation of PEG moieties. The insulin detection ability of each biosensor was determined using interferometric reflectance spectroscopy, using a range of different media both with and without serum. Sensing performance was compared in terms of response value, response time and limit of detection (LOD) for each platform. In order to demonstrate the capability of the best performing biosensor to detect insulin from real samples, an in vitro investigation with the aptamer-modified surface was performed. This biosensor was exposed to buffer conditioned by glucose-stimulated human islets, with the result showing a positive response and a high degree of selectivity towards insulin capture. The obtained results correlated well with the ELISA used in the clinic for assaying glucose-stimulated insulin release from donor islets. We anticipate that this type of sensor can be applied as a rapid point-of-use biosensor to assess the quality of donor islets in terms of their insulin production efficiency, prior to transplantation.
Collapse
Affiliation(s)
- Rinku Chhasatia
- ARC Centre of Excellence in Convergent Bio-Nano Science and Technology, Future Industries Institute, University of South Australia, Mawson Lakes, South Australia 5095, Australia
| | - Martin J Sweetman
- Experimental Therapeutics Laboratory, Hanson Institute and Samson Institute, School of Pharmacy and Medical Sciences, University of South Australia, Adelaide, SA 5000, Australia
| | - Frances J Harding
- ARC Centre of Excellence in Convergent Bio-Nano Science and Technology, Future Industries Institute, University of South Australia, Mawson Lakes, South Australia 5095, Australia
| | - Michaela Waibel
- St. Vincent's Institute of Medical Research, Victoria 3065, Australia
| | - Tom Kay
- St. Vincent's Institute of Medical Research, Victoria 3065, Australia
| | - Helen Thomas
- St. Vincent's Institute of Medical Research, Victoria 3065, Australia
| | - Thomas Loudovaris
- St. Vincent's Institute of Medical Research, Victoria 3065, Australia
| | - Nicolas H Voelcker
- ARC Centre of Excellence in Convergent Bio-Nano Science and Technology, Future Industries Institute, University of South Australia, Mawson Lakes, South Australia 5095, Australia.
| |
Collapse
|