1
|
Dow CT. Proposing Bromo-Epi-Androsterone (BEA) for Stiff Person Syndrome (SPS). Microorganisms 2025; 13:824. [PMID: 40284660 PMCID: PMC12029804 DOI: 10.3390/microorganisms13040824] [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: 02/25/2025] [Revised: 03/23/2025] [Accepted: 03/24/2025] [Indexed: 04/29/2025] Open
Abstract
SPS is characterized by progressive spasmodic muscular rigidity. SPS is thought to be an autoimmune disease with a prominent feature of antibodies against glutamic acid decarboxylase (GAD). GAD is responsible for the enzymatic conversion of glutamic acid (glutamate) into the inhibitory neurotransmitter gamma-aminobutyric acid (GABA). Reduced GABA activity leads to increased excitability in the central nervous system, resulting in muscle rigidity and spasms characteristic of SPS. While SPS is rare, anti-GAD antibodies seen in SPS are also seen in the much more common autoimmune disease, type 1 diabetes (T1D). There is evolving research showing that the anti-GAD antibodies of T1D are produced in response to the presence of mycobacterial heat shock protein 65 (mHSP65), and the mHSP65 is produced in response to an occult infection by a bacterium, Mycobacterium avium subspecies Paratuberculosis (MAP). Humans are broadly exposed to MAP in food, water, and air. There are linear and conformational similarities between the epitopes of GAD and mHSP65. This article proposes that MAP is also an infectious trigger for SPS. Dehydroepiandrosterone (DHEA) is a principal component of the steroid metabolome; it plateaus in young adults and then steadily declines. Bromo-epi-androsterone (BEA) is a potent synthetic analog of DHEA; unlike DHEA, it is non-androgenic, non-anabolic, and an effective modulator of immune dysregulation. BEA is also an anti-infective agent and has been shown to benefit mycobacterial infections, including tuberculosis and leprosy. With the immune stabilizing capacity of BEA as well as its anti-mycobacterial properties, there is reason to believe that a randomized clinical trial with BEA may be beneficial for SPS.
Collapse
Affiliation(s)
- Coad Thomas Dow
- McPherson Eye Research Institute, University of Wisconsin-Madison, Madison, WI 53705, USA;
- Protibea Therapeutics, LLC., Naples, FL 34105, USA
| |
Collapse
|
2
|
Łaszczych D, Czernicka A, Łaszczych K. Targeting GABA signaling in type 1 diabetes and its complications- an update on the state of the art. Pharmacol Rep 2025; 77:409-424. [PMID: 39833509 DOI: 10.1007/s43440-025-00697-7] [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: 10/30/2024] [Revised: 01/04/2025] [Accepted: 01/13/2025] [Indexed: 01/22/2025]
Abstract
Type 1 diabetes (T1D) is an autoimmune disease that leads to the progressive destruction of insulin-producing β cells, resulting in lifelong insulin dependence and a range of severe complications. Beyond conventional glycemic control, innovative therapeutic strategies are needed to address the underlying disease mechanisms. Recent research has highlighted gamma-aminobutyric acid (GABA) as a promising therapeutic target for T1D due to its dual role in modulating both β cell survival and immune response within pancreatic islets. GABA signaling supports β cell regeneration, inhibits α cell hyperactivity, and promotes α-to-β cell transdifferentiation, contributing to improved islet function. Moreover, GABA's influence extends to mitigating T1D complications, including nephropathy, neuropathy, and retinopathy, as well as regulating central nervous system pathways involved in glucose metabolism. This review consolidates the latest advances in GABA-related T1D therapies, covering animal preclinical and human clinical studies and examining the therapeutic potential of GABA receptor modulation, combination therapies, and dietary interventions. Emphasis is placed on the translational potential of GABA-based approaches to enhance β cell viability and counteract autoimmune processes in T1D. Our findings underscore the therapeutic promise of GABA signaling modulation as a novel approach for T1D treatment and encourage further investigation into this pathway's role in comprehensive diabetes management.
Collapse
Affiliation(s)
- Dariusz Łaszczych
- Faculty of Medicine, Collegium Medicum, Nicolaus Copernicus University in Torun, Jagiellońska 13, 85-067, Bydgoszcz, Poland.
| | | | - Katarzyna Łaszczych
- Faculty of Pharmaceutical Sciences, Medical University of Silesia in Katowice, Jedności 8, Sosnowiec, 41-200, Poland
- Ziko Pharmacy, Plebiscytowa 39, Katowice, Poland
| |
Collapse
|
3
|
Barakat H, Aljutaily T. Role of γ-Aminobutyric Acid (GABA) as an Inhibitory Neurotransmitter in Diabetes Management: Mechanisms and Therapeutic Implications. Biomolecules 2025; 15:399. [PMID: 40149935 PMCID: PMC11940341 DOI: 10.3390/biom15030399] [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: 01/31/2025] [Revised: 03/03/2025] [Accepted: 03/07/2025] [Indexed: 03/29/2025] Open
Abstract
GABA (γ-Aminobutyric Acid), a well-established inhibitory neurotransmitter in the central nervous system, has garnered considerable interest for its potential role in diabetes management, particularly due to its presence in pancreatic islets. This review aims to explore the therapeutic role of GABA in diabetes management and its potential mechanisms for antidiabetic effects. Relevant studies were searched across databases such as PubMed and ScienceDirect, applying strict eligibility criteria focused on GABA administration methods and diabetic models. The collective results showed that the administration of GABA in diabetic models resulted in remarkable enhancements in glucose and insulin homeostasis, favorable modifications in lipid profiles, and amelioration of dysfunctions across neural, hepatic, renal, and cardiac systems. The findings from the literature demonstrated that GABAergic signaling within pancreatic tissues can significantly contribute to the stimulation of β cell proliferation through the facilitation of a sustained trans-differentiation process, wherein glucagon-secreting α cells are converted into insulin-secreting β-like cells. In addition, activated GABAergic signaling can trigger the initiation of the PI3K/AKT signaling pathway within pancreatic tissues, leading to improved insulin signaling and maintained glucose homeostasis. GABAergic signaling can further function within hepatic tissues, promoting inhibitory effects on the expression of genes related to gluconeogenesis and lipogenesis. Moreover, GABA may enhance gut microbiota diversity by attenuating gut inflammation, attributable to its anti-inflammatory and immunomodulatory properties. Furthermore, the neuroprotective effects of GABA play a significant role in ameliorating neural disorders associated with diabetes by facilitating a substantial reduction in neuronal apoptosis. In conclusion, GABA emerges as a promising candidate for an antidiabetic agent; however, further research is highly encouraged to develop a rigorously designed framework that comprehensively identifies and optimizes the appropriate dosages and intervention methods for effectively managing and combating diabetes.
Collapse
Affiliation(s)
- Hassan Barakat
- Department of Food Science and Human Nutrition, College of Agriculture and Food, Qassim University, Buraydah 51452, Saudi Arabia;
| | | |
Collapse
|
4
|
Jagomäe T, Velling S, Tikva TB, Maksimtšuk V, Gaur N, Reimets R, Kaasik A, Vasar E, Plaas M. GABA and GLP-1 receptor agonist combination therapy modifies diabetes and Langerhans islet cytoarchitecture in a rat model of Wolfram syndrome. Diabetol Metab Syndr 2025; 17:82. [PMID: 40050934 PMCID: PMC11887366 DOI: 10.1186/s13098-025-01651-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/06/2025] [Accepted: 02/25/2025] [Indexed: 03/09/2025] Open
Abstract
BACKGROUND AND AIM Wolfram syndrome (WS) is a rare autosomal disorder caused by WFS1 gene mutations, currently lacking approved treatments. Preclinical and clinical reports suggest that diabetes medications, such as glucagon-like peptide-1 receptor agonist (GLP1-RA), slow WS-related diabetes and neurodegeneration, improving patient outcomes. Gamma-aminobutyric acid (GABA) has crucial role in pancreatic islet function and blood glucose regulation. However, its specific role in WS diabetic pathophysiology has never been explored. The aim of this study was to enhance the therapeutic efficacy of liraglutide in mitigating the progression of diabetes associated with WS through supplementation with GABA. METHODS In this study, 5-month-old glucose intolerant WS rats and their wild-type littermates where daily treated with GABA (1 g/kg/day), liraglutide (0.4 mg/kg/day), or a combination of both. During the four-month experimental period, the diabetic phenotype was closely monitored using intraperitoneal glucose tolerance tests (IPGTT) and corresponding hormone measurements via enzyme-linked immunoassay. Following the treatments, immunohistochemical staining was performed to examine the morphology, cellular distribution, and health of Langerhans islets. RESULTS Unlike in conventional diabetes models, GABA monotherapy alone had no significant effect on the diabetic phenotype in WS rats. In contrast, liraglutide monotherapy effectively delayed diabetes progression. Remarkably, the combined therapy of GABA and liraglutide reversed the diabetic phenotype, significantly enhancing glucose homeostasis, as well as insulin and C-peptide secretion. The combined treatment also increased β-cell mass and corrected the pancreatic Langerhans intra-islet ratio of α-, β-, and δ-cells. As a result, the overall morphology and cytoarchitecture of the pancreatic islets were fully restored, suggesting a potential role for these agents in preserving islet integrity. Additionally, both liraglutide and combination therapy increased the number of GAD (glutamic acid decarboxylase) 65/67-positive β-cells in WS rats, indicating an improvement in general β-cell health. CONCLUSION GABA monotherapy had no significant effect on the diabetic phenotype in WS rats, while liraglutide monotherapy effectively delayed diabetes progression. However, the combination therapy of GABA and liraglutide demonstrated a markedly superior effect, not only reversing the diabetic phenotype but also significantly enhancing glucose homeostasis, insulin and C-peptide secretion, and β-cell mass. This combined treatment led to a restoration of Langerhans islet architecture, correction of the endocrine cell proportions, and a notable increase in GAD65/67-positive β-cells, indicating improved β-cell health and function. These findings provide strong evidence supporting the evaluation of GABA and GLP-1 RAs as a combination therapy in clinical trials. Their synergistic effects may offer enhanced β-cell protection, promote functional recovery, and uncover novel therapeutic pathways for treating patients with WS.
Collapse
Affiliation(s)
- Toomas Jagomäe
- Laboratory Animal Centre, Institute of Biomedicine and Translational Medicine, University of Tartu, 14b Ravila Street, 50411, Tartu, Estonia.
| | - Sandra Velling
- Laboratory Animal Centre, Institute of Biomedicine and Translational Medicine, University of Tartu, 14b Ravila Street, 50411, Tartu, Estonia
| | - Tessa Britt Tikva
- Laboratory Animal Centre, Institute of Biomedicine and Translational Medicine, University of Tartu, 14b Ravila Street, 50411, Tartu, Estonia
| | - Varvara Maksimtšuk
- Laboratory Animal Centre, Institute of Biomedicine and Translational Medicine, University of Tartu, 14b Ravila Street, 50411, Tartu, Estonia
| | - Nayana Gaur
- Laboratory Animal Centre, Institute of Biomedicine and Translational Medicine, University of Tartu, 14b Ravila Street, 50411, Tartu, Estonia
| | - Riin Reimets
- Laboratory Animal Centre, Institute of Biomedicine and Translational Medicine, University of Tartu, 14b Ravila Street, 50411, Tartu, Estonia
| | - Allen Kaasik
- Department of Pharmacology, Institute of Biomedicine and Translational Medicine, University of Tartu, 19 Ravila Street, 50411, Tartu, Estonia
| | - Eero Vasar
- Department of Physiology, Institute of Biomedicine and Translational Medicine, University of Tartu, 19 Ravila Street, 50411, Tartu, Estonia
| | - Mario Plaas
- Laboratory Animal Centre, Institute of Biomedicine and Translational Medicine, University of Tartu, 14b Ravila Street, 50411, Tartu, Estonia.
| |
Collapse
|
5
|
Liu C, Cheng L, Yang M, He Z, Jia Y, Xu L, Zhang Y. Screening for Safe and Efficient Monascus Strains with Functions of Lowering Blood Lipids, Blood Glucose, and Blood Pressure. Foods 2025; 14:835. [PMID: 40077539 PMCID: PMC11899137 DOI: 10.3390/foods14050835] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2025] [Revised: 02/07/2025] [Accepted: 02/10/2025] [Indexed: 03/14/2025] Open
Abstract
Monascus is a fungus widely used in food fermentation. This study employed microbial technology, combined with microscopic morphological observations and ITS sequence analysis, to isolate, purify, and identify 10 strains of red yeast mold from various Monascus products. After the HPLC detection of metabolic products, the M8 strain containing the toxic substance citrinin was excluded. Using the EWM-TOPSIS model, the remaining nine safe Monascus strains were evaluated for their inhibitory activities against pancreatic lipase, α-glucosidase, α-amylase, and the angiotensin-converting enzyme. The M2 strain with the highest comprehensive scores for lowering blood sugar, blood lipids, and blood pressure was selected. Its fermentation product at a concentration of 3 mg/mL had inhibition rates of 96.938%, 81.903%, and 72.215%, respectively. The contents of the blood lipid-lowering active substance Monacolin K and the blood sugar and blood pressure-lowering active substance GABA were 18.078 mg/g and 5.137 mg/g, respectively. This strain can be utilized for the biosynthesis of important active substances such as Monacolin K and GABA, as well as for the fermentation production of safe and effective functional foods to address health issues like high blood lipids, high blood sugar, and high blood pressure in people. This study also provides insights into the use of natural fungi to produce healthy foods for combating chronic diseases in humans.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Yuansong Zhang
- College of Sericulture, Textile and Biomass Science, Southwest University, Chongqing 400715, China; (C.L.); (L.C.); (M.Y.); (Z.H.); (Y.J.); (L.X.)
| |
Collapse
|
6
|
Alhamar G, Vinci C, Franzese V, Tramontana F, Le Goux N, Ludvigsson J, Nissim A, Strollo R. The role of oxidative post-translational modifications in type 1 diabetes pathogenesis. Front Immunol 2025; 16:1537405. [PMID: 40028329 PMCID: PMC11868110 DOI: 10.3389/fimmu.2025.1537405] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2024] [Accepted: 01/22/2025] [Indexed: 03/05/2025] Open
Abstract
The pathogenesis of type 1 diabetes (T1D) involves a complex interplay of genetic predisposition, immune processes, and environmental factors, leading to the selective destruction of pancreatic beta-cells by the immune system. Emerging evidence suggests that intrinsic beta-cell factors, including oxidative stress and post-translational modifications (PTM) of beta-cell antigens, may also contribute to their immunogenicity, shedding new light on the multifaceted pathogenesis of T1D. Over the past 30 years, neoepitopes generated by PTMs have been hypothesized to play a role in T1D pathogenesis, but their involvement has only been systematically investigated in recent years. In this review, we explored the interplay between oxidative PTMs, neoepitopes, and T1D, highlighting oxidative stress as a pivotal factor in immune system dysfunction, beta-cell vulnerability, and disease onset.
Collapse
Affiliation(s)
- Ghadeer Alhamar
- Department of Immunology and Microbiology, Dasman Diabetes Institute, Dasman, Kuwait
| | - Chiara Vinci
- Center for Diabetes Research, Université Libre de Bruxelles, Brussels, Belgium
| | - Valentina Franzese
- Department for the Promotion of Human Science and Quality of Life, San Raffaele Open University, Rome, Italy
- Department of Medicine, Fondazione Policlinico Universitario Campus Bio-Medico di Roma, Rome, Italy
- Department of Medicine, Università Campus Bio-Medico di Roma, Rome, Italy
| | - Flavia Tramontana
- Department of Medicine, Università Campus Bio-Medico di Roma, Rome, Italy
| | - Nelig Le Goux
- Biochemical Pharmacology, William Harvey Research Institute, Queen Mary University of London, London, United Kingdom
| | - Johnny Ludvigsson
- Crown Princess Victoria Children’s Hospital and Division of Pediatrics, Department of Biomedical and Clinical Sciences, Linköping University, Linköping, Sweden
| | - Ahuva Nissim
- Biochemical Pharmacology, William Harvey Research Institute, Queen Mary University of London, London, United Kingdom
| | - Rocky Strollo
- Department for the Promotion of Human Science and Quality of Life, San Raffaele Open University, Rome, Italy
- Department of Medicine, Fondazione Policlinico Universitario Campus Bio-Medico di Roma, Rome, Italy
| |
Collapse
|
7
|
Testone G, Sobolev AP, Lambreva MD, Aturki Z, Mele G, Lamprillo M, Magnanimi F, Serino G, Arnesi G, Giannino D. The molecular pathways leading to GABA and lactic acid accumulation in florets of organic broccoli rabe ( Brassica rapa subsp. sylvestris) stored as fresh or as minimally processed product. HORTICULTURE RESEARCH 2025; 12:uhae274. [PMID: 39830310 PMCID: PMC11739617 DOI: 10.1093/hr/uhae274] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 07/02/2024] [Accepted: 09/21/2024] [Indexed: 01/22/2025]
Abstract
In the context of organic farming, the introduction of a local product to wider markets and an evaluation of storage effects, metabolic and transcriptomic variations in two broccoli rabe genotypes from production cycles of two different years were studied by comparing florets of stored fresh (SF) and packaged (P) for 4 days with those harvested fresh from the field (H). Twenty-five hydrosoluble compounds, including amino acids, carbohydrates, and organic acids, were quantified by untargeted nuclear magnetic resonance (NMR). Principal component analysis produced a neat separation among the three commodity statuses with P being the most divergent and SF closer to H. In the packaged florets, carbohydrate levels dropped significantly (over -52%), while the levels of amino acids and organic acids varied. There was an increase in stress-responsive phenylalanine and valine (over 30%) and succinic and α-ketoglutaric acids (over 75%). Compound correlation analyses indicated a carbohydrate sink towards γ-aminobutyric acid (GABA) and lactic acid (LA) metabolism under hypoxic conditions in packaged florets. RNA-seq analysis revealed that over 4000 genes were differentially expressed in SF vs H and 8000 in P vs H. Several CAR and AA pathways were significantly enriched in S and even more significantly in P, when compared to H. A map of gene expression (175 genes) and metabolite contents (14 compounds) was constructed to elucidate the gene routes that lead to accumulation of GABA and LA, known for healthy properties, in P. WGCNA and promoter binding site analyses enabled the identification of transcription factors (bZIP, WRKY, ERF types), interactions, and targeted genes encoding key enzymes in GABA and LA accumulation.
Collapse
Affiliation(s)
- Giulio Testone
- Institute for Biological Systems, National Research Council (CNR), Via Salaria Km 29,300, 00015 Monterotondo, Rome, Italy
| | - Anatoly Petrovich Sobolev
- Institute for Biological Systems, National Research Council (CNR), Via Salaria Km 29,300, 00015 Monterotondo, Rome, Italy
| | - Maya Dimova Lambreva
- Institute for Biological Systems, National Research Council (CNR), Via Salaria Km 29,300, 00015 Monterotondo, Rome, Italy
| | - Zeineb Aturki
- Institute for Biological Systems, National Research Council (CNR), Via Salaria Km 29,300, 00015 Monterotondo, Rome, Italy
| | - Giovanni Mele
- Institute for Biological Systems, National Research Council (CNR), Via Salaria Km 29,300, 00015 Monterotondo, Rome, Italy
| | - Michele Lamprillo
- Institute for Biological Systems, National Research Council (CNR), Via Salaria Km 29,300, 00015 Monterotondo, Rome, Italy
| | - Francesco Magnanimi
- Department of Biology and Biotechnology, Sapienza Università di Roma, 00185 Rome, Italy
| | - Giovanna Serino
- Department of Biology and Biotechnology, Sapienza Università di Roma, 00185 Rome, Italy
| | - Giuseppe Arnesi
- Enza Zaden Italia, Strada Statale Aurelia km. 96.400, 01016 Tarquinia, Viterbo, Italy
| | - Donato Giannino
- Institute for Biological Systems, National Research Council (CNR), Via Salaria Km 29,300, 00015 Monterotondo, Rome, Italy
| |
Collapse
|
8
|
Mick GJ, McCormick KL. The role of GABA in type 1 diabetes. Front Endocrinol (Lausanne) 2024; 15:1453396. [PMID: 39619323 PMCID: PMC11604429 DOI: 10.3389/fendo.2024.1453396] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/23/2024] [Accepted: 10/22/2024] [Indexed: 12/13/2024] Open
Abstract
Gamma aminobutyric acid (GABA) is synthesized from glutamate by glutamic decarboxylase (GAD). The entero-pancreatic biology of GABA, which is produced by pancreatic islets, GAD-expressing microbiota, enteric immune cells, or ingested through diet, supports an essential physiologic role of GABA in the health and disease. Outside the central nervous system (CNS), GABA is uniquely concentrated in pancreatic β-cells. They express GAD65, which is a type 1 diabetes (T1D) autoantigen. Glutamate constitutes 10% of the amino acids in dietary protein and is preeminently concentrated in human milk. GABA is enriched in many foods, such as tomato and fermented cheese, and is an over-the-counter supplement. Selected microbiota in the midgut have the enzymatic capacity to produce GABA. Intestinal microbiota interact with gut-associated lymphoid tissue to maintain host defenses and immune tolerance, which are implicated in autoimmune disease. Although GABA is a widely known inhibitory neurotransmitter, oral GABA does not cross the blood brain barrier. Three diabetes-related therapeutic actions are ascribed to GABA, namely, increasing pancreatic β-cell content, attenuating excess glucagon and tamping down T-cell immune destruction. These salutary actions have been observed in numerous rodent diabetes models that usually employed high or near-continuous GABA doses. Clinical studies, to date, have identified positive effects of oral GABA on peripheral blood mononuclear cell cytokine release and plasma glucagon. Going forward, it is reassuring that oral GABA therapy has been well-tolerated and devoid of serious adverse effects.
Collapse
Affiliation(s)
- Gail J. Mick
- Department of Pediatrics, University of Alabama at Birmingham, Birmingham, AL, United States
| | | |
Collapse
|
9
|
Bourgeois S, Coenen S, Degroote L, Willems L, Van Mulders A, Pierreux J, Heremans Y, De Leu N, Staels W. Harnessing beta cell regeneration biology for diabetes therapy. Trends Endocrinol Metab 2024; 35:951-966. [PMID: 38644094 DOI: 10.1016/j.tem.2024.03.006] [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: 02/03/2024] [Revised: 03/21/2024] [Accepted: 03/21/2024] [Indexed: 04/23/2024]
Abstract
The pandemic scale of diabetes mellitus is alarming, its complications remain devastating, and current treatments still pose a major burden on those affected and on the healthcare system as a whole. As the disease emanates from the destruction or dysfunction of insulin-producing pancreatic β-cells, a real cure requires their restoration and protection. An attractive strategy is to regenerate β-cells directly within the pancreas; however, while several approaches for β-cell regeneration have been proposed in the past, clinical translation has proven challenging. This review scrutinizes recent findings in β-cell regeneration and discusses their potential clinical implementation. Hereby, we aim to delineate a path for innovative, targeted therapies to help shift from 'caring for' to 'curing' diabetes.
Collapse
Affiliation(s)
- Stephanie Bourgeois
- Genetics, Reproduction, and Development (GRAD), Beta Cell Neogenesis (BENE) Research Unit, Vrije Universiteit Brussel (VUB), 1090 Brussels, Belgium
| | - Sophie Coenen
- Genetics, Reproduction, and Development (GRAD), Beta Cell Neogenesis (BENE) Research Unit, Vrije Universiteit Brussel (VUB), 1090 Brussels, Belgium
| | - Laure Degroote
- Genetics, Reproduction, and Development (GRAD), Beta Cell Neogenesis (BENE) Research Unit, Vrije Universiteit Brussel (VUB), 1090 Brussels, Belgium
| | - Lien Willems
- Genetics, Reproduction, and Development (GRAD), Beta Cell Neogenesis (BENE) Research Unit, Vrije Universiteit Brussel (VUB), 1090 Brussels, Belgium
| | - Annelore Van Mulders
- Genetics, Reproduction, and Development (GRAD), Beta Cell Neogenesis (BENE) Research Unit, Vrije Universiteit Brussel (VUB), 1090 Brussels, Belgium
| | - Julie Pierreux
- Genetics, Reproduction, and Development (GRAD), Beta Cell Neogenesis (BENE) Research Unit, Vrije Universiteit Brussel (VUB), 1090 Brussels, Belgium
| | - Yves Heremans
- Genetics, Reproduction, and Development (GRAD), Beta Cell Neogenesis (BENE) Research Unit, Vrije Universiteit Brussel (VUB), 1090 Brussels, Belgium
| | - Nico De Leu
- Genetics, Reproduction, and Development (GRAD), Beta Cell Neogenesis (BENE) Research Unit, Vrije Universiteit Brussel (VUB), 1090 Brussels, Belgium; Endocrinology, Universiteit Ziekenhuis Brussel (UZ Brussel), 1090 Brussels, Belgium; Endocrinology, ASZ Aalst, 9300 Aalst, Belgium.
| | - Willem Staels
- Genetics, Reproduction, and Development (GRAD), Beta Cell Neogenesis (BENE) Research Unit, Vrije Universiteit Brussel (VUB), 1090 Brussels, Belgium; Pediatric Endocrinology, Department of Pediatrics, KidZ Health Castle, Universiteit Ziekenhuis Brussel (UZ Brussel), 1090 Brussels, Belgium.
| |
Collapse
|
10
|
Zhu L, Wang Z, Gao L, Chen X. Unraveling the Potential of γ-Aminobutyric Acid: Insights into Its Biosynthesis and Biotechnological Applications. Nutrients 2024; 16:2760. [PMID: 39203897 PMCID: PMC11357613 DOI: 10.3390/nu16162760] [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: 07/11/2024] [Revised: 08/09/2024] [Accepted: 08/16/2024] [Indexed: 09/03/2024] Open
Abstract
γ-Aminobutyric acid (GABA) is a widely distributed non-protein amino acid that serves as a crucial inhibitory neurotransmitter in the brain, regulating various physiological functions. As a result of its potential benefits, GABA has gained substantial interest in the functional food and pharmaceutical industries. The enzyme responsible for GABA production is glutamic acid decarboxylase (GAD), which catalyzes the irreversible decarboxylation of glutamate. Understanding the crystal structure and catalytic mechanism of GAD is pivotal in advancing our knowledge of GABA production. This article provides an overview of GAD's sources, structure, and catalytic mechanism, and explores strategies for enhancing GABA production through fermentation optimization, metabolic engineering, and genetic engineering. Furthermore, the effects of GABA on the physiological functions of animal organisms are also discussed. To meet the increasing demand for GABA, various strategies have been investigated to enhance its production, including optimizing fermentation conditions to facilitate GAD activity. Additionally, metabolic engineering techniques have been employed to increase the availability of glutamate as a precursor for GABA biosynthesis. By fine-tuning fermentation conditions and utilizing metabolic and genetic engineering techniques, it is possible to achieve higher yields of GABA, thus opening up new avenues for its application in functional foods and pharmaceuticals. Continuous research in this field holds immense promise for harnessing the potential of GABA in addressing various health-related challenges.
Collapse
Affiliation(s)
- Lei Zhu
- School of Biological Engineering, Dalian Polytechnic University, Dalian 116034, China;
| | - Zhefeng Wang
- Tianjin Institute of Industrial Biotechnology, Chinese Academy of Sciences, National Technology Innovation Center for Synthetic Biology, Tianjin 300308, China;
| | - Le Gao
- Tianjin Institute of Industrial Biotechnology, Chinese Academy of Sciences, National Technology Innovation Center for Synthetic Biology, Tianjin 300308, China;
| | - Xiaoyi Chen
- School of Biological Engineering, Dalian Polytechnic University, Dalian 116034, China;
| |
Collapse
|
11
|
Saravanan PB, Kalivarathan J, McClintock K, Mohammed S, Burch E, Morecock C, Liu J, Khan A, Levy MF, Kanak MA. Inflammatory and hypoxic stress-induced islet exosomes released during isolation are associated with poor transplant outcomes in islet autotransplantation. Am J Transplant 2024; 24:967-982. [PMID: 38364959 DOI: 10.1016/j.ajt.2024.02.011] [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: 09/05/2023] [Revised: 02/03/2024] [Accepted: 02/10/2024] [Indexed: 02/18/2024]
Abstract
Islets experience enormous stress during the isolation process, leading to suboptimal endocrine function after total pancreatectomy with islet autotransplantation (TPIAT). Our investigation focused on inducing isolation stress in islets ex vivo, where proinflammatory cytokines and hypoxia prompted the release of stress exosomes (exoS) sized between 50 and 200 nm. Mass spectrometry analysis revealed 3 distinct subgroups of immunogenic proteins within these exoS: damage-associated molecular patterns (DAMPs), chaperones, and autoantigens. The involvement of endosomal-sorting complex required for transport proteins including ras-associated binding proteins7A, ras-associated binding protein GGTA, vacuolar protein sorting associated protein 45, vacuolar protein sorting associated protein 26B, and the tetraspanins CD9 and CD63, in exoS biogenesis was confirmed through immunoblotting. Next, we isolated similar exoS from the islet infusion bags of TPIAT recipients (N = 20). The exosomes from infusion bags exhibited higher DAMP (heat shock protein family A [Hsp70] member 1B and histone H2B) levels, particularly in the insulin-dependent TPIAT group. Additionally, elevated DAMP protein levels in islet infusion bag exosomes correlated with increased insulin requirements (P = .010) and higher hemoglobin A1c levels 1-year posttransplant. A deeper exploration into exoS functionality revealed their potential to activate monocytes via the toll-like receptor 3/7: DAMP axis. This stimulation resulted in the induction of inflammatory phenotypes marked by increased levels of CD68, CD80, inducible nitric oxide synthase, and cyclooxygenase-2. This activation mechanism may impact the successful engraftment of transplanted islets.
Collapse
Affiliation(s)
- Prathab Balaji Saravanan
- Department of Surgery, School of Medicine, VCU, Richmond, Virginia, USA; VCU Hume-Lee Islet Cell Transplant Lab, VCU Health System, Richmond, Virginia, USA.
| | - Jagan Kalivarathan
- VCU Hume-Lee Islet Cell Transplant Lab, VCU Health System, Richmond, Virginia, USA
| | - Kaeden McClintock
- Department of Surgery, School of Medicine, VCU, Richmond, Virginia, USA
| | | | - Elijah Burch
- VCU Hume-Lee Islet Cell Transplant Lab, VCU Health System, Richmond, Virginia, USA
| | - Christiane Morecock
- Department of Biostatistics, School of Medicine, VCU, Richmond, Virginia, USA
| | - Jinze Liu
- Department of Biostatistics, School of Medicine, VCU, Richmond, Virginia, USA
| | - Aamir Khan
- Department of Surgery, School of Medicine, VCU, Richmond, Virginia, USA; VCU Hume-Lee Islet Cell Transplant Lab, VCU Health System, Richmond, Virginia, USA
| | - Marlon F Levy
- Department of Surgery, School of Medicine, VCU, Richmond, Virginia, USA; VCU Hume-Lee Islet Cell Transplant Lab, VCU Health System, Richmond, Virginia, USA
| | - Mazhar A Kanak
- Department of Surgery, School of Medicine, VCU, Richmond, Virginia, USA; VCU Hume-Lee Islet Cell Transplant Lab, VCU Health System, Richmond, Virginia, USA
| |
Collapse
|
12
|
Sutedja JC, de Liyis BG, Saraswati MR. Gamma-aminobutyric acid for delaying type 1 diabetes mellitus: an update. Ann Pediatr Endocrinol Metab 2024; 29:142-151. [PMID: 38956751 PMCID: PMC11220392 DOI: 10.6065/apem.2346184.092] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/11/2023] [Revised: 10/10/2023] [Accepted: 11/28/2023] [Indexed: 07/04/2024] Open
Abstract
The current gold-standard management of hyperglycemia in individuals with type 1 diabetes mellitus (T1DM) is insulin therapy. However, this therapy is associated with a high incidence of complications, and delaying the onset of this disease produces a substantially positive impact on quality of life for individuals with a predisposition to T1DM, especially children. This review aimed to assess the use of gamma-aminobutyric acid (GABA) to delay the onset of T1DM in children. GABA produces protective and proliferative effects in 2 ways, β cell and immune cell modulation. Various in vitro and in vivo studies have shown that GABA induces proliferation of β cells, increases insulin levels, inhibits β-cell apoptosis, and suppresses T helper 1 cell activity against islet antigens. Oral GABA is safe as no serious adverse effects were reported in any of the studies included in this review. These findings demonstrate promising results for the use of GABA treatment to delay T1DM, specifically in genetically predisposed children, through immunoregulatory effects and the ability to induce β-cell proliferation.
Collapse
Affiliation(s)
| | | | - Made Ratna Saraswati
- Division of Endocrinology and Metabolism, Department of Internal Medicine, Faculty of Medicine, Udayana University/Prof. IGNG Ngoerah General Hospital, Bali, Indonesia
| |
Collapse
|
13
|
Cui D, Feng X, Lei S, Zhang H, Hu W, Yang S, Yu X, Su Z. Pancreatic β-cell failure, clinical implications, and therapeutic strategies in type 2 diabetes. Chin Med J (Engl) 2024; 137:791-805. [PMID: 38479993 PMCID: PMC10997226 DOI: 10.1097/cm9.0000000000003034] [Citation(s) in RCA: 9] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2023] [Indexed: 04/06/2024] Open
Abstract
ABSTRACT Pancreatic β-cell failure due to a reduction in function and mass has been defined as a primary contributor to the progression of type 2 diabetes (T2D). Reserving insulin-producing β-cells and hence restoring insulin production are gaining attention in translational diabetes research, and β-cell replenishment has been the main focus for diabetes treatment. Significant findings in β-cell proliferation, transdifferentiation, pluripotent stem cell differentiation, and associated small molecules have served as promising strategies to regenerate β-cells. In this review, we summarize current knowledge on the mechanisms implicated in β-cell dynamic processes under physiological and diabetic conditions, in which genetic factors, age-related alterations, metabolic stresses, and compromised identity are critical factors contributing to β-cell failure in T2D. The article also focuses on recent advances in therapeutic strategies for diabetes treatment by promoting β-cell proliferation, inducing non-β-cell transdifferentiation, and reprograming stem cell differentiation. Although a significant challenge remains for each of these strategies, the recognition of the mechanisms responsible for β-cell development and mature endocrine cell plasticity and remarkable advances in the generation of exogenous β-cells from stem cells and single-cell studies pave the way for developing potential approaches to cure diabetes.
Collapse
Affiliation(s)
- Daxin Cui
- Molecular Medicine Research Center and Department of Endocrinology and Metabolism, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, China
| | - Xingrong Feng
- State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, China
| | - Siman Lei
- Clinical Translational Innovation Center, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, China
| | - Hongmei Zhang
- Molecular Medicine Research Center and Department of Endocrinology and Metabolism, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, China
| | - Wanxin Hu
- State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, China
| | - Shanshan Yang
- Molecular Medicine Research Center and Department of Endocrinology and Metabolism, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, China
| | - Xiaoqian Yu
- Molecular Medicine Research Center and Department of Endocrinology and Metabolism, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, China
| | - Zhiguang Su
- Molecular Medicine Research Center and Department of Endocrinology and Metabolism, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, China
- State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, China
- Clinical Translational Innovation Center, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, China
| |
Collapse
|
14
|
Hwang SM, Rahman MM, Go EJ, Kim YH, Park CK. Specific transcription factors Ascl1 and Lhx6 attenuate diabetic neuropathic pain by modulating spinal neuroinflammation and microglial activation in mice. Biomed Pharmacother 2024; 173:116392. [PMID: 38479183 DOI: 10.1016/j.biopha.2024.116392] [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: 10/13/2023] [Revised: 02/23/2024] [Accepted: 03/06/2024] [Indexed: 03/27/2024] Open
Abstract
Gamma-aminobutyric acid (GABA) neuronal system-related transcription factors (TFs) play a critical role in GABA production, and GABA modulates diabetic neuropathic pain (DNP). The present study investigated the therapeutic effects of intrathecal delivery of two TFs achaete-scute homolog 1 (Ascl1) and LIM homeobox protein 6 (Lhx6) in a mouse model of DNP and elucidated their underlying mechanisms. GABA-related specific TFs, including Ascl1, Lhx6, distal-less homeobox 1, distal-less homeobox 5, the Nkx2.1 homeobox gene, and the Nkx2.2 homeobox gene, were investigated under normal and diabetic conditions. Among these, the expression of Ascl1 and Lhx6 was significantly downregulated in mice with diabetes. Therefore, a single intrathecal injection of combined lenti-Ascl1/Lhx6 was performed. Intrathecal delivery of lenti-Ascl1/Lhx6 significantly relieved mechanical allodynia and heat hyperalgesia in mice with DNP. Ascl1/Lhx6 delivery also reduced microglial activation, decreased the levels of pro-inflammatory cytokines including tumor necrosis factor-α and interleukin (IL)-1β, increased the levels of anti-inflammatory cytokines including IL-4, IL-10, and IL-13, and reduced the activation of p38, c-Jun N-terminal kinase, and NF-κB in the spinal cord of mice with DNP, thereby reducing DNP. The results of this study suggest that intrathecal Ascl1/Lhx6 delivery attenuates DNP via upregulating spinal GABA neuronal function and inducing anti-inflammatory effects.
Collapse
Affiliation(s)
- Sung-Min Hwang
- Gachon Pain Center and Department of Physiology, Gachon University College of Medicine, Incheon 21999, Republic of Korea
| | - Md Mahbubur Rahman
- Gachon Pain Center and Department of Physiology, Gachon University College of Medicine, Incheon 21999, Republic of Korea
| | - Eun Jin Go
- Gachon Pain Center and Department of Physiology, Gachon University College of Medicine, Incheon 21999, Republic of Korea
| | - Yong Ho Kim
- Gachon Pain Center and Department of Physiology, Gachon University College of Medicine, Incheon 21999, Republic of Korea.
| | - Chul-Kyu Park
- Gachon Pain Center and Department of Physiology, Gachon University College of Medicine, Incheon 21999, Republic of Korea.
| |
Collapse
|
15
|
Turbitt J, Moffett RC, Brennan L, Johnson PRV, Flatt PR, McClenaghan NH, Tarasov AI. Molecular determinants and intracellular targets of taurine signalling in pancreatic islet β-cells. Acta Physiol (Oxf) 2024; 240:e14101. [PMID: 38243723 DOI: 10.1111/apha.14101] [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: 06/26/2023] [Revised: 12/26/2023] [Accepted: 01/08/2024] [Indexed: 01/21/2024]
Abstract
AIM Despite its abundance in pancreatic islets of Langerhans and proven antihyperglycemic effects, the impact of the essential amino acid, taurine, on islet β-cell biology has not yet received due consideration, which prompted the current studies exploring the molecular selectivity of taurine import into β-cells and its acute and chronic intracellular interactions. METHODS The molecular aspects of taurine transport were probed by exposing the clonal pancreatic BRIN BD11 β-cells and primary mouse and human islets to a range of the homologs of the amino acid (assayed at 2-20 mM), using the hormone release and imaging of intracellular signals as surrogate read-outs. Known secretagogues were employed to profile the interaction of taurine with acute and chronic intracellular signals. RESULTS Taurine transporter TauT was expressed in the islet β-cells, with the transport of taurine and homologs having a weak sulfonate specificity but significant sensitivity to the molecular weight of the transporter. Taurine, hypotaurine, homotaurine, and β-alanine enhanced insulin secretion in a glucose-dependent manner, an action potentiated by cytosolic Ca2+ and cAMP. Acute and chronic β-cell insulinotropic effects of taurine were highly sensitive to co-agonism with GLP-1, forskolin, tolbutamide, and membrane depolarization, with an unanticipated indifference to the activation of PKC and CCK8 receptors. Pre-culturing with GLP-1 or KATP channel inhibitors sensitized or, respectively, desensitized β-cells to the acute taurine stimulus. CONCLUSION Together, these data demonstrate the pathways whereby taurine exhibits a range of beneficial effects on insulin secretion and β-cell function, consistent with the antidiabetic potential of its dietary low-dose supplementation.
Collapse
Affiliation(s)
- Julie Turbitt
- School of Biomedical Sciences, Ulster University, Coleraine, UK
| | | | - Lorraine Brennan
- UCD Institute of Food and Health, UCD School of Agriculture and Food Science, University College Dublin, Dublin 4, Republic of Ireland
- UCD Conway Institute of Biomolecular and Biomedical Research, University College Dublin, Dublin 4, Republic of Ireland
| | - Paul R V Johnson
- Nuffield Department of Surgical Sciences, Oxford Centre for Diabetes, Endocrinology and Metabolism, University of Oxford, Churchill Hospital, Oxford, UK
- Oxford Biomedical Research Centre (OxBRC), Oxford, UK
| | - Peter R Flatt
- School of Biomedical Sciences, Ulster University, Coleraine, UK
| | - Neville H McClenaghan
- School of Biomedical Sciences, Ulster University, Coleraine, UK
- Department of Life Sciences, Atlantic Technological University, Sligo, Republic of Ireland
| | | |
Collapse
|
16
|
Farrim MI, Gomes A, Milenkovic D, Menezes R. Gene expression analysis reveals diabetes-related gene signatures. Hum Genomics 2024; 18:16. [PMID: 38326874 PMCID: PMC10851551 DOI: 10.1186/s40246-024-00582-z] [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: 09/04/2023] [Accepted: 02/01/2024] [Indexed: 02/09/2024] Open
Abstract
BACKGROUND Diabetes is a spectrum of metabolic diseases affecting millions of people worldwide. The loss of pancreatic β-cell mass by either autoimmune destruction or apoptosis, in type 1-diabetes (T1D) and type 2-diabetes (T2D), respectively, represents a pathophysiological process leading to insulin deficiency. Therefore, therapeutic strategies focusing on restoring β-cell mass and β-cell insulin secretory capacity may impact disease management. This study took advantage of powerful integrative bioinformatic tools to scrutinize publicly available diabetes-associated gene expression data to unveil novel potential molecular targets associated with β-cell dysfunction. METHODS A comprehensive literature search for human studies on gene expression alterations in the pancreas associated with T1D and T2D was performed. A total of 6 studies were selected for data extraction and for bioinformatic analysis. Pathway enrichment analyses of differentially expressed genes (DEGs) were conducted, together with protein-protein interaction networks and the identification of potential transcription factors (TFs). For noncoding differentially expressed RNAs, microRNAs (miRNAs) and long noncoding RNAs (lncRNAs), which exert regulatory activities associated with diabetes, identifying target genes and pathways regulated by these RNAs is fundamental for establishing a robust regulatory network. RESULTS Comparisons of DEGs among the 6 studies showed 59 genes in common among 4 or more studies. Besides alterations in mRNA, it was possible to identify differentially expressed miRNA and lncRNA. Among the top transcription factors (TFs), HIPK2, KLF5, STAT1 and STAT3 emerged as potential regulators of the altered gene expression. Integrated analysis of protein-coding genes, miRNAs, and lncRNAs pointed out several pathways involved in metabolism, cell signaling, the immune system, cell adhesion, and interactions. Interestingly, the GABAergic synapse pathway emerged as the only common pathway to all datasets. CONCLUSIONS This study demonstrated the power of bioinformatics tools in scrutinizing publicly available gene expression data, thereby revealing potential therapeutic targets like the GABAergic synapse pathway, which holds promise in modulating α-cells transdifferentiation into β-cells.
Collapse
Affiliation(s)
- M I Farrim
- CBIOS, Universidade Lusófona's Research Center for Biosciences & Health Technologies, Universidade Lusófona, Lisbon, Portugal
- Universidad de Alcalá, Escuela de Doctorado, Madrid, Spain
| | - A Gomes
- CBIOS, Universidade Lusófona's Research Center for Biosciences & Health Technologies, Universidade Lusófona, Lisbon, Portugal
| | - D Milenkovic
- Department of Nutrition, University of California Davis, Davis, USA
| | - R Menezes
- CBIOS, Universidade Lusófona's Research Center for Biosciences & Health Technologies, Universidade Lusófona, Lisbon, Portugal.
| |
Collapse
|
17
|
Cao Z, Chen H, Zhou C, Gong M, Li Y, Shao Y, Wu Y, Bao D. Exogenous γ-Aminobutyric Acid (GABA) Enhanced Response to Abiotic Stress in Hypsizygus marmoreus by Improving Mycelial Growth and Antioxidant Capacity. Metabolites 2024; 14:94. [PMID: 38392986 PMCID: PMC10890280 DOI: 10.3390/metabo14020094] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2023] [Revised: 01/19/2024] [Accepted: 01/21/2024] [Indexed: 02/25/2024] Open
Abstract
γ-Aminobutyric (GABA) acid is a nutrient and signaling molecule existing in many plants, participating in the regulation of metabolism and various physiological activities. Two strains of Hypsizygus marmoreus (a white variety and a brown variety) were investigated to study the impact of exogenous GABA on mycelial growth and the response to stress. Mycelial growth, microscopic morphology, antioxidant profile, and gad2 expression in H. marmoreu were investigated under salt, dehydration, or cold stress. The results indicated that 5 mM GABA stimulated mycelial growth under standard cultivation conditions, whereas GABA addition over 10 mM hindered the growth. Under salt, dehydration, or cold stress, treatment with 5 mM GABA significantly enhanced the mycelial growth rate and density of both H. marmoreus strains by promoting front hyphae branching. Meanwhile, the activities of key antioxidant enzymes such as peroxidase (POD), catalase (CAT), and ascorbate peroxidase (APX) were enhanced by GABA, thereby augmenting the defensive network against abiotic stress. Additionally, gad2 expression and GABA concentration were increased under abiotic stresses as a resistance regulation response. The exogenous addition of GABA strengthened the upregulation of gad2 expression and GABA production. These findings indicated that exogenously adding low concentrations of GABA effectively enhanced the mycelial growth and antioxidant profile of H. marmoreus, thereby improving its resistance against stresses.
Collapse
Affiliation(s)
- Zhi Cao
- School of Health Science and Engineering, University of Shanghai for Science and Technology, Shanghai 200093, China
- National Engineering Research Center of Edible Fungi, Key Laboratory of Applied Mycological Resources and Utilization of Ministry of Agriculture, Institute of Edible Fungi, Shanghai Academy of Agricultural Sciences, Shanghai 201403, China
| | - Hongyu Chen
- National Engineering Research Center of Edible Fungi, Key Laboratory of Applied Mycological Resources and Utilization of Ministry of Agriculture, Institute of Edible Fungi, Shanghai Academy of Agricultural Sciences, Shanghai 201403, China
| | - Chenli Zhou
- National Engineering Research Center of Edible Fungi, Key Laboratory of Applied Mycological Resources and Utilization of Ministry of Agriculture, Institute of Edible Fungi, Shanghai Academy of Agricultural Sciences, Shanghai 201403, China
| | - Ming Gong
- National Engineering Research Center of Edible Fungi, Key Laboratory of Applied Mycological Resources and Utilization of Ministry of Agriculture, Institute of Edible Fungi, Shanghai Academy of Agricultural Sciences, Shanghai 201403, China
| | - Yan Li
- National Engineering Research Center of Edible Fungi, Key Laboratory of Applied Mycological Resources and Utilization of Ministry of Agriculture, Institute of Edible Fungi, Shanghai Academy of Agricultural Sciences, Shanghai 201403, China
| | - Youran Shao
- National Engineering Research Center of Edible Fungi, Key Laboratory of Applied Mycological Resources and Utilization of Ministry of Agriculture, Institute of Edible Fungi, Shanghai Academy of Agricultural Sciences, Shanghai 201403, China
| | - Yingying Wu
- National Engineering Research Center of Edible Fungi, Key Laboratory of Applied Mycological Resources and Utilization of Ministry of Agriculture, Institute of Edible Fungi, Shanghai Academy of Agricultural Sciences, Shanghai 201403, China
| | - Dapeng Bao
- School of Health Science and Engineering, University of Shanghai for Science and Technology, Shanghai 200093, China
- National Engineering Research Center of Edible Fungi, Key Laboratory of Applied Mycological Resources and Utilization of Ministry of Agriculture, Institute of Edible Fungi, Shanghai Academy of Agricultural Sciences, Shanghai 201403, China
| |
Collapse
|
18
|
Schmidt MD, Ishahak M, Augsornworawat P, Millman JR. Comparative and integrative single cell analysis reveals new insights into the transcriptional immaturity of stem cell-derived β cells. BMC Genomics 2024; 25:105. [PMID: 38267908 PMCID: PMC10807170 DOI: 10.1186/s12864-024-10013-x] [Citation(s) in RCA: 12] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2023] [Accepted: 01/14/2024] [Indexed: 01/26/2024] Open
Abstract
Diabetes cell replacement therapy has the potential to be transformed by human pluripotent stem cell-derived β cells (SC-β cells). However, the precise identity of SC-β cells in relationship to primary fetal and adult β-cells remains unclear. Here, we used single-cell sequencing datasets to characterize the transcriptional identity of islets from in vitro differentiation, fetal islets, and adult islets. Our analysis revealed that SC-β cells share a core β-cell transcriptional identity with human adult and fetal β-cells, however SC-β cells possess a unique transcriptional profile characterized by the persistent expression and activation of progenitor and neural-biased gene networks. These networks are present in SC-β cells, irrespective of the derivation protocol used. Notably, fetal β-cells also exhibit this neural signature at the transcriptional level. Our findings offer insights into the transcriptional identity of SC-β cells and underscore the need for further investigation of the role of neural transcriptional networks in their development.
Collapse
Affiliation(s)
- Mason D Schmidt
- Division of Endocrinology, Metabolism and Lipid Research, Washington University School of Medicine, MSC 8127-057-08, 660 South Euclid Avenue, St. Louis, MO, 63110, USA
| | - Matthew Ishahak
- Division of Endocrinology, Metabolism and Lipid Research, Washington University School of Medicine, MSC 8127-057-08, 660 South Euclid Avenue, St. Louis, MO, 63110, USA
| | - Punn Augsornworawat
- Division of Endocrinology, Metabolism and Lipid Research, Washington University School of Medicine, MSC 8127-057-08, 660 South Euclid Avenue, St. Louis, MO, 63110, USA
- Department of Biomedical Engineering, Washington University in St. Louis, 1 Brookings Drive, St. Louis, MO, 63130, USA
- Department of Immunology, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok, 10700, Thailand
| | - Jeffrey R Millman
- Division of Endocrinology, Metabolism and Lipid Research, Washington University School of Medicine, MSC 8127-057-08, 660 South Euclid Avenue, St. Louis, MO, 63110, USA.
- Department of Biomedical Engineering, Washington University in St. Louis, 1 Brookings Drive, St. Louis, MO, 63130, USA.
| |
Collapse
|
19
|
Garcia-Gutierrez E, O’Mahony AK, Dos Santos RS, Marroquí L, Cotter PD. Gut microbial metabolic signatures in diabetes mellitus and potential preventive and therapeutic applications. Gut Microbes 2024; 16:2401654. [PMID: 39420751 PMCID: PMC11492678 DOI: 10.1080/19490976.2024.2401654] [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: 11/29/2023] [Revised: 07/08/2024] [Accepted: 09/03/2024] [Indexed: 10/19/2024] Open
Abstract
Diabetes mellitus can be subdivided into several categories based on origin and clinical characteristics. The most common forms of diabetes are type 1 (T1D), type 2 diabetes (T2D) and gestational diabetes mellitus (GDM). T1D and T2D are chronic diseases affecting around 537 million adults worldwide and it is projected that these numbers will increase by 12% over the next two decades, while GDM affects up to 30% of women during pregnancy, depending on diagnosis methods. These forms of diabetes have varied origins: T1D is an autoimmune disease, while T2D is commonly associated with, but not limited to, certain lifestyle patterns and GDM can result of a combination of genetic predisposition and pregnancy factors. Despite some pathogenic differences among these forms of diabetes, there are some common markers associated with their development. For instance, gut barrier impairment and inflammation associated with an unbalanced gut microbiota and their metabolites may be common factors in diabetes development and progression. Here, we summarize the microbial signatures that have been linked to diabetes, how they are connected to diet and, ultimately, the impact on metabolite profiles resulting from host-gut microbiota-diet interactions. Additionally, we summarize recent advances relating to promising preventive and therapeutic interventions focusing on the targeted modulation of the gut microbiota to alleviate T1D, T2D and GDM.
Collapse
Affiliation(s)
- Enriqueta Garcia-Gutierrez
- Food Biosciences Department, Teagasc Food Research Centre, Fermoy, Co. Cork, Ireland
- APC Microbiome Ireland, University College Cork, Co. Cork, Ireland
- VistaMilk SFI Research Centre, Fermoy, Co. Cork, Ireland
- Departamento de Ingeniería Agronómica, Instituto de Biotecnología Vegetal, ETSIA-Universidad Politécnica de Cartagena, Cartagena, Spain
| | - A. Kate O’Mahony
- Food Biosciences Department, Teagasc Food Research Centre, Fermoy, Co. Cork, Ireland
- APC Microbiome Ireland, University College Cork, Co. Cork, Ireland
- School of Microbiology, University College Cork, Co. Cork, Ireland
| | - Reinaldo Sousa Dos Santos
- Instituto de Investigación, Desarrollo e Innovación en Biotecnología Sanitaria de Elche (IDiBE), Universidad Miguel Hernández de Elche, Elche, Spain
- CIBER de Diabetes y Enfermedades Metabólicas Asociadas (CIBERDEM), Instituto de Salud Carlos III, Madrid, Spain
| | - Laura Marroquí
- Instituto de Investigación, Desarrollo e Innovación en Biotecnología Sanitaria de Elche (IDiBE), Universidad Miguel Hernández de Elche, Elche, Spain
- CIBER de Diabetes y Enfermedades Metabólicas Asociadas (CIBERDEM), Instituto de Salud Carlos III, Madrid, Spain
| | - Paul D. Cotter
- Food Biosciences Department, Teagasc Food Research Centre, Fermoy, Co. Cork, Ireland
- APC Microbiome Ireland, University College Cork, Co. Cork, Ireland
- VistaMilk SFI Research Centre, Fermoy, Co. Cork, Ireland
| |
Collapse
|
20
|
Wu W, Zhang J, Qiao Y, Ren L, Chen Z, Fu Y, Yang Z. Association of long-term benzodiazepine hypnotic use and prediabetes in US population: A cross-sectional analysis of national health and nutrition examination survey data. Medicine (Baltimore) 2023; 102:e35705. [PMID: 37960777 PMCID: PMC10637564 DOI: 10.1097/md.0000000000035705] [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: 07/31/2023] [Accepted: 09/28/2023] [Indexed: 11/15/2023] Open
Abstract
Benzodiazepine hypnotics' effects on glucose metabolism are seldom reported, and the association between long-term (≥4 weeks) benzodiazepine usage and prediabetes has not been studied. This study was aimed to investigate the association between benzodiazepine hypnotic usage for ≥ 3 months and the prevalence of prediabetes. We analyzed cross-sectional data from the National Health and Nutrition Examination Survey (NHANES) during 2005 to 2008, selecting adult participants without diabetes who used benzodiazepine hypnotics for at least 3 months or did not take any hypnotics. Individuals taking other hypnotics, antipsychotics, glucocorticoids, or hypoglycemic drugs were excluded. We defined prediabetes as an hemoglobin A1C (HbA1C) 5.7-6.4%, as suggested by the American Diabetes Association. Prescribed drug information was self-reported and checked by official interviewers, and HbA1C data in NHANES was recognized by the National Glycohemoglobin Standardization Program. We calculated the propensity score according to the covariates and adjusted it using multivariate logistic regression. Lower thresholds of HbA1C ≥ 5.5% or ≥ 5.3% were also analyzed. Among 4694 eligible participants, 38 received benzodiazepine hypnotics; using these hypnotics for ≥ 3 months was not significantly associated with the prevalence of prediabetes, as well as HbA1C ≥ 5.5% or ≥ 5.3%. Adjusted for propensity score, the respective odds ratios for prediabetes, HbA1C ≥ 5.5%, and HbA1C ≥ 5.3% were 1.09 (95% confidence interval [CI] 0.19-6.32), 0.83 (95% CI 0.22-3.13), and 1.22 (95% CI 0.3-4.93). No significant association was found between benzodiazepine hypnotic usage ≥ 3 months and the prevalence of prediabetes.
Collapse
Affiliation(s)
- Weizhen Wu
- Graduate School, Beijing University of Chinese Medicine, Beijing, P.R. China
| | - Junning Zhang
- Graduate School, Beijing University of Chinese Medicine, Beijing, P.R. China
| | - Yizhuo Qiao
- Graduate School, China Academy of Chinese Medical Science, Beijing, P.R. China
| | - Lijiang Ren
- Graduate School, Beijing University of Chinese Medicine, Beijing, P.R. China
| | - Zhe Chen
- Graduate School, Beijing University of Chinese Medicine, Beijing, P.R. China
| | - Yan Fu
- Xiyuan Hospital of China Academy of Chinese Medical Science, Beijing, P.R. China
| | - Zhixu Yang
- Xiyuan Hospital of China Academy of Chinese Medical Science, Beijing, P.R. China
| |
Collapse
|
21
|
Yu J, Yao X, Zhang X, Hao J. New insights of metabolite abnormalities in the thalamus of rats with iminodiproprionitrile-induced tic disorders. Front Neurosci 2023; 17:1201294. [PMID: 37841690 PMCID: PMC10570423 DOI: 10.3389/fnins.2023.1201294] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2023] [Accepted: 09/15/2023] [Indexed: 10/17/2023] Open
Abstract
Introduction This study aimed to investigate pathological changes in the "Glutamate (Glu)-γ-aminobutyric acid (GABA)" loop and apply widely targeted metabolomic analysis technology to comprehensively explore metabolite abnormalities/ in the thalamus of rats with tic disorders (TD). Methods Wistar rats were randomized into control, TD, and tiapride (Tia) groups. Iminodipropionitrile (IDPN) was used to induce TD in rats. The Tia group was administered tiapride. Neurotransmitter levels in the thalamus of rats in the three groups were measured using UPLC-3Q MS. And, the protein expression levels of Glu decarboxylase (GAD65/67) and GABA transporter protein (GAD-T) were measured using western blotting. The mRNA expression levels of these genes were evaluated using real-time polymerase chain reaction. Lastly, other metabolites in the thalamus were detected by widely targeted metabolomic analysis between TD and Control group rats. Results The Glu level, Glu/GABA ratio, and Asp level in the TD group were significantly higher (all p < 0.001) than those of the Control group, whereas the GABA and Gly levels were lower (p < 0.001 and p = 0.009, respectively). The Tia group exhibited a significant reduction in the Glu level (p = 0.001) compared with the TD group. The protein expression level of GAD67 in TD group was higher (p = 0.009) and the mRNA expression levels of GAD65, GAD67, and GAT-1 were lower (p < 0.05) than those of the Control group. The Tia group did not display any differences in GAD65, GAD67, or GAT-1 expression. Widely targeted metabolomic analysis revealed that 34 substances were abnornal between the TD and Control groups (9 upregulated and 25 downregulated). Neurosteroids (progesterone, corticosterone) exhibited distinct differences. Metabolite analysis using the Kyoto encyclopedia for genes and genomes indicated that the steroid hormone biosynthesis pathway may be involved in TD pathogenesis. Conclusion This study revealed metabolic abnormalities in the thalamus of rats with TD. The interaction between neurotransmitters and neurosteroid biosynthesis represents a new direction for future studies.
Collapse
Affiliation(s)
- Jingru Yu
- School of Medicine, Shaoxing University, Shaoxing, China
| | - Xuan Yao
- School of Medicine, Shaoxing University, Shaoxing, China
| | - Xin Zhang
- Xin Hua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Juanjuan Hao
- School of Medicine, Shaoxing University, Shaoxing, China
| |
Collapse
|
22
|
Sun Y, Mehmood A, Giampieri F, Battino MA, Chen X. Insights into the cellular, molecular, and epigenetic targets of gamma-aminobutyric acid against diabetes: a comprehensive review on its mechanisms. Crit Rev Food Sci Nutr 2023; 64:12620-12637. [PMID: 37694998 DOI: 10.1080/10408398.2023.2255666] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/12/2023]
Abstract
Diabetes is a metabolic disease due to impaired or defective insulin secretion and is considered one of the most serious chronic diseases worldwide. Gamma-aminobutyric acid (GABA) is a naturally occurring non-protein amino acid commonly present in a wide range of foods. A number of studies documented that GABA has good anti-diabetic potential. This review summarized the available dietary sources of GABA as well as animal and human studies on the anti-diabetic properties of GABA, while also discussing the underlying mechanisms. GABA may modulate diabetes through various pathways such as inhibiting the activities of α-amylase and α-glucosidase, promoting β-cell proliferation, stimulating insulin secretion from β-cells, inhibiting glucagon secretion from α-cells, improving insulin resistance and glucose tolerance, and increasing antioxidant and anti-inflammatory activities. However, further mechanistic studies on animals and human are needed to confirm the therapeutic effects of GABA against diabetes.
Collapse
Affiliation(s)
- Yu Sun
- School of Food and Biological Engineering, Jiangsu University, Zhenjiang, Jiangsu, P.R. China
- Institute of Food Physical Processing, Jiangsu University, Zhenjiang, Jiangsu, P.R. China
| | - Arshad Mehmood
- School of Food and Biological Engineering, Jiangsu University, Zhenjiang, Jiangsu, P.R. China
- Institute of Food Physical Processing, Jiangsu University, Zhenjiang, Jiangsu, P.R. China
| | - Francesca Giampieri
- Research Group on Food, Nutritional Biochemistry and Health, Universidad Europea del Atlántico, Santander, Spain
| | - Maurizio Antonio Battino
- International Joint Research Laboratory of Intelligent Agriculture and Agri-products Processing, Jiangsu University, Zhenjiang, Jiangsu, P.R. China
- Research Group on Food, Nutritional Biochemistry and Health, Universidad Europea del Atlántico, Santander, Spain
- Department of Clinical Sciences, Università Politecnica delle Marche, Ancona, Italy
| | - Xiumin Chen
- School of Food and Biological Engineering, Jiangsu University, Zhenjiang, Jiangsu, P.R. China
- Institute of Food Physical Processing, Jiangsu University, Zhenjiang, Jiangsu, P.R. China
- International Joint Research Laboratory of Intelligent Agriculture and Agri-products Processing, Jiangsu University, Zhenjiang, Jiangsu, P.R. China
| |
Collapse
|
23
|
Abstract
Despite major advances over the past decade, prevention and treatment of type 1 diabetes mellitus (T1DM) remain suboptimal, with large and unexplained variations in individual responses to interventions. The current classification schema for diabetes mellitus does not capture the complexity of this disease or guide clinical management effectively. One of the approaches to achieve the goal of applying precision medicine in diabetes mellitus is to identify endotypes (that is, well-defined subtypes) of the disease each of which has a distinct aetiopathogenesis that might be amenable to specific interventions. Here, we describe epidemiological, clinical, genetic, immunological, histological and metabolic differences within T1DM that, together, suggest heterogeneity in its aetiology and pathogenesis. We then present the emerging endotypes and their impact on T1DM prediction, prevention and treatment.
Collapse
Affiliation(s)
- Maria J Redondo
- Paediatric Diabetes & Endocrinology, Texas Children's Hospital, Baylor College of Medicine, Houston, TX, USA.
| | - Noel G Morgan
- Exeter Centre of Excellence for Diabetes Research (EXCEED), Department of Clinical and Biomedical and Science, University of Exeter Medical School, Exeter, UK
| |
Collapse
|
24
|
Тюренков ИН, Файбисович ТИ, Бакулин ДА. [Synergistic effects of GABA and hypoglycemic drugs]. PROBLEMY ENDOKRINOLOGII 2023; 69:61-69. [PMID: 37694868 PMCID: PMC10520901 DOI: 10.14341/probl13257] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/11/2023] [Accepted: 02/23/2023] [Indexed: 09/12/2023]
Abstract
Diabetes mellitus (DM) is the leading cause of premature death and disability. Despite a significant number of drugs, the effectiveness of therapy aimed at normalizing the level of glycemia and preventing complications does not fully satisfy doctors and patients. Therefore, the search for new approaches for the prevention and treatment of DM and its complications continues. Significant resources are used to develop new drugs, but recently the possibility of using «old» widely available drugs with newly discovered pleiotropic properties has been substantiated. These may include preparations of gammaaminobutyric acid (GABA) and agents that directly or indirectly activate GABAergic transmission, which have a pronounced pancreatic protective effect, which has been widely discussed in foreign literature over the past 10-15 years. However, there are few such publications in the domestic literature.It has been established that the content of GABA in β-cells in patients with type 1 and type 2 diabetes is reduced and this correlates with the severity of the disease. Genetic suppression of GABA receptors causes a significant decrease in the mass of β-cells and glucose-stimulated insulin secretion, which confirms the importance of GABA in ensuring glucose homeostasis and the advisability of replenishing the GABA deficiency in DM with its additional administration. It has been established that in animals with DM, GABA suppresses apoptosis and stimulates the regeneration of β-cells, increases β-cell mass and insulin production.Experimental data have been obtained indicating a synergistic effect of GABA when combined with glucagon-like peptide-1 (GLP-1) receptor agonists, DPP-4 inhibitors and sodium-glucose cotransporter 2 (SGLT-2) inhibitors, when a more pronounced pancreoprotective effect is observed, due to decrease in oxidative and nitrosative stress, inflammation, increase in the level of Klotho protein, Nrf-2 activity and antioxidant defense enzymes, suppression of NF-kB activity and expression of pro-inflammatory cytokines. As a result, all this leads to a decrease in apoptosis and death of β-cells, an increase in β-cell mass, insulin production and, at the same time, a decrease in glucagon levels and insulin resistance.The review substantiates the feasibility of using GABA and drugs with a positive GABAeric effect in combination with new generation antidiabetic agents: GLP-1 receptor agonists, DPP-4 inhibitors and SGLT-2 inhibitors in order to increase their antidiabetic potential.The search was carried out in the databases Pubmed, eLibrary, Medline. Keywords: diabetes mellitus, gamma-aminobutyric acid, glucagon-like peptide-1, GLP-1 receptor agonists, glucose-dependent insulinotropic peptide, dipeptidyl peptidase inhibitors, sodium-glucose cotransporter 2 inhibitors. The search was carried out from 2000 to 2022, but the review presents the results studies published mainly in the last 3 years, due to the requirements of the journal for the maximum amount of work and the number of sources.
Collapse
Affiliation(s)
| | | | - Д. А. Бакулин
- Волгоградский государственный медицинский университет
| |
Collapse
|
25
|
Tamarit-Rodriguez J. Metabolic Role of GABA in the Secretory Function of Pancreatic β-Cells: Its Hypothetical Implication in β-Cell Degradation in Type 2 Diabetes. Metabolites 2023; 13:697. [PMID: 37367856 DOI: 10.3390/metabo13060697] [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: 04/22/2023] [Revised: 05/22/2023] [Accepted: 05/24/2023] [Indexed: 06/28/2023] Open
Abstract
The stimulus-secretion coupling of a glucose-induced release is generally attributed to the metabolism of the hexose in the β-cells in the glycolytic pathway and the citric acid cycle. Glucose metabolism generates an increased cytosolic concentration of ATP and of the ATP/ADP ratio that closes the ATP-dependent K+-channel at the plasma membrane. The resultant depolarization of the β-cells opens voltage-dependent Ca2+-channels at the plasma membrane that triggers the exocytosis of insulin secretory granules. The secretory response is biphasic with a first and transient peak followed by a sustained phase. The first phase is reproduced by a depolarization of the β-cells with high extracellular KCl maintaining the KATP-channels open with diazoxide (triggering phase); the sustained phase (amplifying phase) depends on the participation of metabolic signals that remain to be determined. Our group has been investigating for several years the participation of the β-cell GABA metabolism in the stimulation of insulin secretion by three different secretagogues (glucose, a mixture of L-leucine plus L-glutamine, and some branched chain alpha-ketoacids, BCKAs). They stimulate a biphasic secretion of insulin accompanied by a strong suppression of the intracellular islet content of gamma-aminobutyric acid (GABA). As the islet GABA release simultaneously decreased, it was concluded that this resulted from an increased GABA shunt metabolism. The entrance of GABA into the shunt is catalyzed by GABA transaminase (GABAT) that transfers an amino group between GABA and alpha-ketoglutarate, resulting in succinic acid semialdehyde (SSA) and L-glutamate. SSA is oxidized to succinic acid that is further oxidized in the citric acid cycle. Inhibitors of GABAT (gamma-vinyl GABA, gabaculine) or glutamic acid decarboxylating activity (GAD), allylglycine, partially suppress the secretory response as well as GABA metabolism and islet ATP content and the ATP/ADP ratio. It is concluded that the GABA shunt metabolism contributes together with the own metabolism of metabolic secretagogues to increase islet mitochondrial oxidative phosphorylation. These experimental findings emphasize that the GABA shunt metabolism is a previously unrecognized anaplerotic mitochondrial pathway feeding the citric acid cycle with a β-cell endogenous substrate. It is therefore a postulated alternative to the proposed mitochondrial cataplerotic pathway(s) responsible for the amplification phase of insulin secretion. It is concluded the new postulated alternative suggests a possible new mechanism of β-cell degradation in type 2 (perhaps also in type 1) diabetes.
Collapse
|
26
|
Fogarty MJ. Inhibitory Synaptic Influences on Developmental Motor Disorders. Int J Mol Sci 2023; 24:ijms24086962. [PMID: 37108127 PMCID: PMC10138861 DOI: 10.3390/ijms24086962] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2023] [Revised: 04/05/2023] [Accepted: 04/06/2023] [Indexed: 04/29/2023] Open
Abstract
During development, GABA and glycine play major trophic and synaptic roles in the establishment of the neuromotor system. In this review, we summarise the formation, function and maturation of GABAergic and glycinergic synapses within neuromotor circuits during development. We take special care to discuss the differences in limb and respiratory neuromotor control. We then investigate the influences that GABAergic and glycinergic neurotransmission has on two major developmental neuromotor disorders: Rett syndrome and spastic cerebral palsy. We present these two syndromes in order to contrast the approaches to disease mechanism and therapy. While both conditions have motor dysfunctions at their core, one condition Rett syndrome, despite having myriad symptoms, has scientists focused on the breathing abnormalities and their alleviation-to great clinical advances. By contrast, cerebral palsy remains a scientific quagmire or poor definitions, no widely adopted model and a lack of therapeutic focus. We conclude that the sheer abundance of diversity of inhibitory neurotransmitter targets should provide hope for intractable conditions, particularly those that exhibit broad spectra of dysfunction-such as spastic cerebral palsy and Rett syndrome.
Collapse
Affiliation(s)
- Matthew J Fogarty
- Department of Physiology & Biomedical Engineering, Mayo Clinic, Rochester, MN 55902, USA
| |
Collapse
|
27
|
Kim K, Yoon H. Gamma-Aminobutyric Acid Signaling in Damage Response, Metabolism, and Disease. Int J Mol Sci 2023; 24:ijms24054584. [PMID: 36902014 PMCID: PMC10003236 DOI: 10.3390/ijms24054584] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2023] [Revised: 02/23/2023] [Accepted: 02/24/2023] [Indexed: 03/03/2023] Open
Abstract
Gamma-aminobutyric acid (GABA) plays a crucial role in signal transduction and can function as a neurotransmitter. Although many studies have been conducted on GABA in brain biology, the cellular function and physiological relevance of GABA in other metabolic organs remain unclear. Here, we will discuss recent advances in understanding GABA metabolism with a focus on its biosynthesis and cellular functions in other organs. The mechanisms of GABA in liver biology and disease have revealed new ways to link the biosynthesis of GABA to its cellular function. By reviewing what is known about the distinct effects of GABA and GABA-mediated metabolites in physiological pathways, we provide a framework for understanding newly identified targets regulating the damage response, with implications for ameliorating metabolic diseases. With this review, we suggest that further research is necessary to develop GABA's beneficial and toxic effects on metabolic disease progression.
Collapse
|
28
|
Yang Y, Ren L, Li W, Zhang Y, Zhang S, Ge B, Yang H, Du G, Tang B, Wang H, Wang J. GABAergic signaling as a potential therapeutic target in cancers. Biomed Pharmacother 2023; 161:114410. [PMID: 36812710 DOI: 10.1016/j.biopha.2023.114410] [Citation(s) in RCA: 25] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2023] [Revised: 02/09/2023] [Accepted: 02/15/2023] [Indexed: 02/23/2023] Open
Abstract
GABA is the most common inhibitory neurotransmitter in the vertebrate central nervous system. Synthesized by glutamic acid decarboxylase, GABA could specifically bind with two GABA receptors to transmit inhibition signal stimuli into cells: GABAA receptor and GABAB receptor. In recent years, emerging studies revealed that GABAergic signaling not only participated in traditional neurotransmission but was involved in tumorigenesis as well as regulating tumor immunity. In this review, we summarize the existing knowledge of the GABAergic signaling pathway in tumor proliferation, metastasis, progression, stemness, and tumor microenvironment as well as the underlying molecular mechanism. We also discussed the therapeutical advances in targeting GABA receptors to provide the theoretical basis for pharmacological intervention of GABAergic signaling in cancer treatment especially immunotherapy.
Collapse
Affiliation(s)
- Yihui Yang
- The State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Beijing 100050, China; Key Laboratory of Drug Target Research and Drug Screen, Institute of Materia Medica, Chinese Academy of Medical Science and Peking Union Medical College, Beijing 100050, China
| | - Liwen Ren
- The State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Beijing 100050, China; Key Laboratory of Drug Target Research and Drug Screen, Institute of Materia Medica, Chinese Academy of Medical Science and Peking Union Medical College, Beijing 100050, China
| | - Wan Li
- The State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Beijing 100050, China; Key Laboratory of Drug Target Research and Drug Screen, Institute of Materia Medica, Chinese Academy of Medical Science and Peking Union Medical College, Beijing 100050, China
| | - Yizhi Zhang
- The State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Beijing 100050, China; Key Laboratory of Drug Target Research and Drug Screen, Institute of Materia Medica, Chinese Academy of Medical Science and Peking Union Medical College, Beijing 100050, China
| | - Sen Zhang
- The State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Beijing 100050, China; Key Laboratory of Drug Target Research and Drug Screen, Institute of Materia Medica, Chinese Academy of Medical Science and Peking Union Medical College, Beijing 100050, China
| | - Binbin Ge
- The State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Beijing 100050, China; Key Laboratory of Drug Target Research and Drug Screen, Institute of Materia Medica, Chinese Academy of Medical Science and Peking Union Medical College, Beijing 100050, China
| | - Hong Yang
- The State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Beijing 100050, China; Key Laboratory of Drug Target Research and Drug Screen, Institute of Materia Medica, Chinese Academy of Medical Science and Peking Union Medical College, Beijing 100050, China
| | - Guanhua Du
- The State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Beijing 100050, China; Key Laboratory of Drug Target Research and Drug Screen, Institute of Materia Medica, Chinese Academy of Medical Science and Peking Union Medical College, Beijing 100050, China
| | - Bo Tang
- Department of Pancreatic Cancer, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Tianjin's Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, 300060, China
| | - Hongquan Wang
- Department of Pancreatic Cancer, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Tianjin's Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, 300060, China
| | - Jinhua Wang
- The State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Beijing 100050, China; Key Laboratory of Drug Target Research and Drug Screen, Institute of Materia Medica, Chinese Academy of Medical Science and Peking Union Medical College, Beijing 100050, China.
| |
Collapse
|
29
|
Heli Z, Hongyu C, Dapeng B, Yee Shin T, Yejun Z, Xi Z, Yingying W. Recent advances of γ-aminobutyric acid: Physiological and immunity function, enrichment, and metabolic pathway. Front Nutr 2022; 9:1076223. [PMID: 36618705 PMCID: PMC9813243 DOI: 10.3389/fnut.2022.1076223] [Citation(s) in RCA: 29] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2022] [Accepted: 11/28/2022] [Indexed: 12/24/2022] Open
Abstract
γ-aminobutyric acid (GABA) is a non-protein amino acid which naturally and widely occurs in animals, plants, and microorganisms. As the chief inhibitory neurotransmitter in the central nervous system of mammals, it has become a popular dietary supplement and has promising application in food industry. The current article reviews the most recent literature regarding the physiological functions, preparation methods, enrichment methods, metabolic pathways, and applications of GABA. This review sheds light on developing GABA-enriched plant varieties and food products, and provides insights for efficient production of GABA through synthetic biology approaches.
Collapse
Affiliation(s)
- Zhou Heli
- School of Health Science and Engineering, University of Shanghai for Science and Technology, Shanghai, China
| | - Chen Hongyu
- National Engineering Research Center of Edible Fungi, Key Laboratory of Applied Mycological Resources and Utilization of Ministry of Agriculture, Institute of Edible Fungi, Shanghai Academy of Agricultural Sciences, Shanghai, China
| | - Bao Dapeng
- School of Health Science and Engineering, University of Shanghai for Science and Technology, Shanghai, China,National Engineering Research Center of Edible Fungi, Key Laboratory of Applied Mycological Resources and Utilization of Ministry of Agriculture, Institute of Edible Fungi, Shanghai Academy of Agricultural Sciences, Shanghai, China
| | - Tan Yee Shin
- Faculty of Science and Mushroom Research Centre, Institute of Biological Sciences, University of Malaya, Kuala Lumpur, Malaysia
| | - Zhong Yejun
- State Key Laboratory of Food Science and Technology, Nanchang University, Nanchang, Jiangxi, China
| | - Zhang Xi
- BannerBio Nutraceuticals Inc., Shenzhen, China
| | - Wu Yingying
- National Engineering Research Center of Edible Fungi, Key Laboratory of Applied Mycological Resources and Utilization of Ministry of Agriculture, Institute of Edible Fungi, Shanghai Academy of Agricultural Sciences, Shanghai, China,*Correspondence: Wu Yingying,
| |
Collapse
|
30
|
Goode RA, Hum JM, Kalwat MA. Therapeutic Strategies Targeting Pancreatic Islet β-Cell Proliferation, Regeneration, and Replacement. Endocrinology 2022; 164:6836713. [PMID: 36412119 PMCID: PMC9923807 DOI: 10.1210/endocr/bqac193] [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: 09/09/2022] [Revised: 11/16/2022] [Accepted: 11/18/2022] [Indexed: 11/23/2022]
Abstract
Diabetes results from insufficient insulin production by pancreatic islet β-cells or a loss of β-cells themselves. Restoration of regulated insulin production is a predominant goal of translational diabetes research. Here, we provide a brief overview of recent advances in the fields of β-cell proliferation, regeneration, and replacement. The discovery of therapeutic targets and associated small molecules has been enabled by improved understanding of β-cell development and cell cycle regulation, as well as advanced high-throughput screening methodologies. Important findings in β-cell transdifferentiation, neogenesis, and stem cell differentiation have nucleated multiple promising therapeutic strategies. In particular, clinical trials are underway using in vitro-generated β-like cells from human pluripotent stem cells. Significant challenges remain for each of these strategies, but continued support for efforts in these research areas will be critical for the generation of distinct diabetes therapies.
Collapse
Affiliation(s)
- Roy A Goode
- Division of Biomedical Sciences, College of Osteopathic Medicine, Marian University, Indianapolis, IN, USA
| | - Julia M Hum
- Division of Biomedical Sciences, College of Osteopathic Medicine, Marian University, Indianapolis, IN, USA
| | - Michael A Kalwat
- Correspondence: Michael A. Kalwat, PhD, Lilly Diabetes Center of Excellence, Indiana Biosciences Research Institute, 1210 Waterway Blvd, Suite 2000, Indianapolis, IN 46202, USA. or
| |
Collapse
|