1
|
Okamoto N, Hoshikawa T, Honma Y, Chibaatar E, Ikenouchi A, Harada M, Yoshimura R. Effect modification of tumor necrosis factor-α on the kynurenine and serotonin pathways in major depressive disorder on type 2 diabetes mellitus. Eur Arch Psychiatry Clin Neurosci 2024; 274:1697-1707. [PMID: 37991535 PMCID: PMC11422469 DOI: 10.1007/s00406-023-01713-8] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/17/2023] [Accepted: 10/29/2023] [Indexed: 11/23/2023]
Abstract
Major depressive disorder (MDD) is strongly associated with type 2 diabetes mellitus (T2DM). The kynurenine and serotonin pathways, as well as chronic low-grade inflammation, are being considered potential links between them. MDD associated with T2DM is less responsive to treatment than that without T2DM; however, the underlying mechanism remains unknown. We aimed to investigate the effects of inflammatory cytokines on the kynurenine and serotonin pathways in patients with comorbid MDD and T2DM and those with only MDD. We recruited 13 patients with comorbid MDD and T2DM and 27 patients with only MDD. We measured interleukin-6 and tumor necrosis factor-α (TNF-α) levels as inflammatory cytokines and metabolites of the kynurenine pathway and examined the relationship between the two. TNF-α levels were significantly higher in patients with comorbid MDD and T2DM than in those with only MDD in univariate (p = 0.044) and multivariate (adjusted p = 0.036) analyses. TNF-α showed a statistically significant effect modification (interaction) with quinolinic acid/tryptophan and serotonin in patients from both groups (β = 1.029, adjusted p < 0.001; β = - 1.444, adjusted p = 0.047, respectively). Limitations attributed to the study design and number of samples may be present. All patients were Japanese with mild to moderate MDD; therefore, the generalizability of our findings may be limited. MDD with T2DM has more inflammatory depression components and activations of the kynurenine pathway by inflammatory cytokines than MDD without T2DM. Hence, administering antidepressants and anti-inflammatory drugs in combination may be more effective in patients with comorbid MDD and T2DM.
Collapse
Affiliation(s)
- Naomichi Okamoto
- Department of Psychiatry, University of Occupational and Environmental Health, 807-8555, Kitakyushu, Fukuoka, 8078555, Japan.
| | - Takashi Hoshikawa
- Department of Psychiatry, University of Occupational and Environmental Health, 807-8555, Kitakyushu, Fukuoka, 8078555, Japan
| | - Yuichi Honma
- Third Department of Internal Medicine, University of Occupational and Environmental Health, Fukuoka, Japan
| | - Enkhmurun Chibaatar
- Department of Psychiatry, University of Occupational and Environmental Health, 807-8555, Kitakyushu, Fukuoka, 8078555, Japan
| | - Atsuko Ikenouchi
- Department of Psychiatry, University of Occupational and Environmental Health, 807-8555, Kitakyushu, Fukuoka, 8078555, Japan
- Medical Center for Dementia, University Hospital, University of Occupational and Environmental Health, Fukuoka, Japan
| | - Masaru Harada
- Third Department of Internal Medicine, University of Occupational and Environmental Health, Fukuoka, Japan
| | - Reiji Yoshimura
- Department of Psychiatry, University of Occupational and Environmental Health, 807-8555, Kitakyushu, Fukuoka, 8078555, Japan
| |
Collapse
|
2
|
Campanale A, Inserra A, Comai S. Therapeutic modulation of the kynurenine pathway in severe mental illness and comorbidities: A potential role for serotonergic psychedelics. Prog Neuropsychopharmacol Biol Psychiatry 2024; 134:111058. [PMID: 38885875 DOI: 10.1016/j.pnpbp.2024.111058] [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: 01/31/2024] [Revised: 05/15/2024] [Accepted: 06/14/2024] [Indexed: 06/20/2024]
Abstract
Mounting evidence points towards a crucial role of the kynurenine pathway (KP) in the altered gut-brain axis (GBA) balance in severe mental illness (SMI, namely depression, bipolar disorder, and schizophrenia) and cardiometabolic comorbidities. Preliminary evidence shows that serotonergic psychedelics and their analogues may hold therapeutic potential in addressing the altered KP in the dysregulated GBA in SMI and comorbidities. In fact, aside from their effects on mood, psychedelics elicit therapeutic improvement in preclinical models of obesity, metabolic syndrome, and vascular inflammation, which are highly comorbid with SMI. Here, we review the literature on the therapeutic modulation of the KP in the dysregulated GBA in SMI and comorbidities, and the potential application of psychedelics to address the altered KP in the brain and systemic dysfunction underlying SMI and comorbidities. Psychedelics might therapeutically modulate the KP in the altered GBA in SMI and comorbidities either directly, via altering the metabolic pathway by influencing the rate-limiting enzymes of the KP and affecting the levels of available tryptophan, or indirectly, by affecting the gut microbiome, gut metabolome, metabolism, and the immune system. Despite promising preliminary evidence, the mechanisms and outcomes of the KP modulation with psychedelics in SMI and systemic comorbidities remain largely unknown and require further investigation. Several concerns are discussed surrounding the potential side effects of this approach in specific cohorts of individuals with SMI and systemic comorbidities.
Collapse
Affiliation(s)
| | - Antonio Inserra
- Department of Psychiatry, McGill University, Montreal, QC, Canada
| | - Stefano Comai
- Department of Psychiatry, McGill University, Montreal, QC, Canada; Department of Pharmaceutical and Pharmacological Sciences, University of Padova, PD, Italy.; IRCCS San Raffaele Scientific Institute, Milan, Italy; Department of Biomedical Sciences, University of Padua, Padua, Italy.
| |
Collapse
|
3
|
Saheki T, Imachi H, Fukunaga K, Sato S, Kobayashi T, Yoshimura T, Saheki N, Murao K. NMDA Suppresses Pancreatic ABCA1 Expression through the MEK/ERK/LXR Pathway in Pancreatic Beta Cells. Nutrients 2024; 16:2865. [PMID: 39275180 PMCID: PMC11396903 DOI: 10.3390/nu16172865] [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: 07/31/2024] [Revised: 08/21/2024] [Accepted: 08/26/2024] [Indexed: 09/16/2024] Open
Abstract
Dysfunction or loss of pancreatic β cells can cause insulin deficiency and impaired glucose regulation, resulting in conditions like type 2 diabetes. The ATP-binding cassette transporter A1 (ABCA1) plays a key role in the reverse cholesterol transport system, and its decreased expression is associated with pancreatic β cell lipotoxicity, resulting in abnormal insulin synthesis and secretion. Increased glutamate release can cause glucotoxicity in β cells, though the detailed mechanisms remain unclear. This study investigated the effect of N-methyl-D-aspartic acid (NMDA) on ABCA1 expression in INS-1 cells and primary pancreatic islets to elucidate the signaling mechanisms that suppress insulin secretion. Using Western blotting, microscopy, and biochemical analyses, we found that NMDA activated the mitogen-activated protein kinase (MEK)-dependent pathway, suppressing ABCA1 protein and mRNA expression. The MEK-specific inhibitor PD98059 restored ABCA1 promoter activity, indicating the involvement of the extracellular signal-regulated kinase (MEK/ERK) pathway. Furthermore, we identified the liver X receptor (LXR) as an effector transcription factor in NMDA regulation of ABCA1 transcription. NMDA treatment increased cholesterol and triglyceride levels while decreasing insulin secretion, even under high-glucose conditions. These effects were abrogated by treatment with PD98059. This study reveals that NMDA suppresses ABCA1 expression via the MEK/ERK/LXR pathway, providing new insights into the pathological suppression of insulin secretion in pancreatic β cells and emphasizing the importance of investigating the role of NMDA in β cell dysfunction.
Collapse
Affiliation(s)
- Takanobu Saheki
- Department of Endocrinology and Metabolism, Faculty of Medicine, Kagawa University, 1750-1, Ikenobe, Miki-cho, Kita-gun 761-0793, Japan; (H.I.); (K.F.); (S.S.); (T.K.); (T.Y.); (N.S.); (K.M.)
| | | | | | | | | | | | | | | |
Collapse
|
4
|
Yuan S, She D, Jiang S, Deng N, Peng J, Ma L. Endoplasmic reticulum stress and therapeutic strategies in metabolic, neurodegenerative diseases and cancer. Mol Med 2024; 30:40. [PMID: 38509524 PMCID: PMC10956371 DOI: 10.1186/s10020-024-00808-9] [Citation(s) in RCA: 37] [Impact Index Per Article: 37.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2023] [Accepted: 03/12/2024] [Indexed: 03/22/2024] Open
Abstract
The accumulation of unfolded or misfolded proteins within the endoplasmic reticulum (ER), due to genetic determinants and extrinsic environmental factors, leads to endoplasmic reticulum stress (ER stress). As ER stress ensues, the unfolded protein response (UPR), comprising three signaling pathways-inositol-requiring enzyme 1, protein kinase R-like endoplasmic reticulum kinase, and activating transcription factor 6 promptly activates to enhance the ER's protein-folding capacity and restore ER homeostasis. However, prolonged ER stress levels propels the UPR towards cellular demise and the subsequent inflammatory cascade, contributing to the development of human diseases, including cancer, neurodegenerative disorders, and diabetes. Notably, increased expression of all three UPR signaling pathways has been observed in these pathologies, and reduction in signaling molecule expression correlates with decreased proliferation of disease-associated target cells. Consequently, therapeutic strategies targeting ER stress-related interventions have attracted significant research interest. In this review, we elucidate the critical role of ER stress in cancer, metabolic, and neurodegenerative diseases, offering novel therapeutic approaches for these conditions.
Collapse
Affiliation(s)
- Siqi Yuan
- Department of Clinical Laboratory, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, 430022, Wuhan, China
| | - Dan She
- Department of Clinical Laboratory, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, 430022, Wuhan, China
| | - Shangming Jiang
- Department of Clinical Laboratory, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, 430022, Wuhan, China
| | - Nan Deng
- Department of Clinical Laboratory, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, 430022, Wuhan, China
| | - Jiayi Peng
- Department of Clinical Laboratory, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, 430022, Wuhan, China
| | - Ling Ma
- Department of Clinical Laboratory, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, 430022, Wuhan, China.
| |
Collapse
|
5
|
Lee C, Lee DK, Wei IA, Qiu TA, Rubakhin SS, Roper MG, Sweedler JV. Relations between Glucose and d-Amino Acids in the Modulation of Biochemical and Functional Properties of Rodent Islets of Langerhans. ACS OMEGA 2023; 8:47723-47734. [PMID: 38144114 PMCID: PMC10733910 DOI: 10.1021/acsomega.3c05983] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/13/2023] [Revised: 11/06/2023] [Accepted: 11/10/2023] [Indexed: 12/26/2023]
Abstract
The cell-to-cell signaling role of d-amino acids (d-AAs) in the mammalian endocrine system, particularly in the islets of Langerhans, has drawn growing interest for their potential involvement in modulating glucose metabolism. Previous studies found colocalization of serine racemase [produces d-serine (d-Ser)] and d-alanine (d-Ala) within insulin-secreting beta cells and d-aspartate (d-Asp) within glucagon-secreting alpha cells. Expressed in the islets, functional N-methyl-d-aspartate receptors are involved in the modulation of glucose-stimulated insulin secretion and have binding sites for several d-AAs. However, knowledge of the regulation of d-AA levels in the islets during glucose stimulation as well as the response of islets to different levels of extracellular d-AAs is limited. In this study, we determined the intracellular and extracellular levels of d-Ser, d-Ala, and d-Asp in cultures of isolated rodent islets exposed to different levels of extracellular glucose. We found that the intracellular levels of the enantiomers demonstrated large variability and, in general, were not affected by extracellular glucose levels. However, significantly lower levels of extracellular d-Ser and d-Ala were observed in the islet media supplemented with 20 mM concentration of glucose compared to the control condition utilizing 3 mM glucose. Glucose-induced oscillations of intracellular free calcium concentration ([Ca2+]i), a proxy for insulin secretion, were modulated by the exogenous application of d-Ser and d-Ala but not by their l-stereoisomers. Our results provide new insights into the roles of d-AAs in the biochemistry and function of pancreatic islets.
Collapse
Affiliation(s)
- Cindy
J. Lee
- Department
of Chemistry and the Beckman Institute, University of Illinois Urbana-Champaign, Urbana, Illinois 61801, United States
| | - Dong-Kyu Lee
- Department
of Chemistry and the Beckman Institute, University of Illinois Urbana-Champaign, Urbana, Illinois 61801, United States
| | - I-An Wei
- Department
of Chemistry and Biochemistry, Florida State
University, Tallahassee, Florida 32306, United States
| | - Tian A. Qiu
- Department
of Chemistry and the Beckman Institute, University of Illinois Urbana-Champaign, Urbana, Illinois 61801, United States
| | - Stanislav S. Rubakhin
- Department
of Chemistry and the Beckman Institute, University of Illinois Urbana-Champaign, Urbana, Illinois 61801, United States
| | - Michael G. Roper
- Department
of Chemistry and Biochemistry, Florida State
University, Tallahassee, Florida 32306, United States
| | - Jonathan V. Sweedler
- Department
of Chemistry and the Beckman Institute, University of Illinois Urbana-Champaign, Urbana, Illinois 61801, United States
| |
Collapse
|
6
|
Liu W, Li Y, Zhao T, Gong M, Wang X, Zhang Y, Xu L, Li W, Li Y, Jia J. The role of N-methyl-D-aspartate glutamate receptors in Alzheimer's disease: From pathophysiology to therapeutic approaches. Prog Neurobiol 2023; 231:102534. [PMID: 37783430 DOI: 10.1016/j.pneurobio.2023.102534] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2023] [Revised: 09/20/2023] [Accepted: 09/25/2023] [Indexed: 10/04/2023]
Abstract
N-Methyl-D-aspartate glutamate receptors (NMDARs) are involved in multiple physiopathological processes, including synaptic plasticity, neuronal network activities, excitotoxic events, and cognitive impairment. Abnormalities in NMDARs can initiate a cascade of pathological events, notably in Alzheimer's disease (AD) and even other neuropsychiatric disorders. The subunit composition of NMDARs is plastic, giving rise to a diverse array of receptor subtypes. While they are primarily found in neurons, NMDAR complexes, comprising both traditional and atypical subunits, are also present in non-neuronal cells, influencing the functions of various peripheral tissues. Furthermore, protein-protein interactions within NMDAR complexes has been linked with Aβ accumulation, tau phosphorylation, neuroinflammation, and mitochondrial dysfunction, all of which potentially served as an obligatory relay of cognitive impairment. Nonetheless, the precise mechanistic link remains to be fully elucidated. In this review, we provided an in-depth analysis of the structure and function of NMDAR, investigated their interactions with various pathogenic proteins, discussed the current landscape of NMDAR-based therapeutics, and highlighted the remaining challenges during drug development.
Collapse
Affiliation(s)
- Wenying Liu
- Innovation Center for Neurological Disorders and Department of Neurology, Xuanwu Hospital, Capital Medical University, National Clinical Research Center for Geriatric Diseases, PR China
| | - Yan Li
- Innovation Center for Neurological Disorders and Department of Neurology, Xuanwu Hospital, Capital Medical University, National Clinical Research Center for Geriatric Diseases, PR China
| | - Tan Zhao
- Innovation Center for Neurological Disorders and Department of Neurology, Xuanwu Hospital, Capital Medical University, National Clinical Research Center for Geriatric Diseases, PR China
| | - Min Gong
- Innovation Center for Neurological Disorders and Department of Neurology, Xuanwu Hospital, Capital Medical University, National Clinical Research Center for Geriatric Diseases, PR China
| | - Xuechu Wang
- Innovation Center for Neurological Disorders and Department of Neurology, Xuanwu Hospital, Capital Medical University, National Clinical Research Center for Geriatric Diseases, PR China
| | - Yue Zhang
- Innovation Center for Neurological Disorders and Department of Neurology, Xuanwu Hospital, Capital Medical University, National Clinical Research Center for Geriatric Diseases, PR China
| | - Lingzhi Xu
- Innovation Center for Neurological Disorders and Department of Neurology, Xuanwu Hospital, Capital Medical University, National Clinical Research Center for Geriatric Diseases, PR China; Beijing Key Laboratory of Geriatric Cognitive Disorders, PR China; Clinical Center for Neurodegenerative Disease and Memory Impairment, Capital Medical University, PR China; Center of Alzheimer's Disease, Beijing Institute of Brain Disorders, Collaborative Innovation Center for Brain Disorders, Capital Medical University, PR China; Key Laboratory of Neurodegenerative Diseases, Ministry of Education, Beijing 100053, PR China
| | - Wenwen Li
- Innovation Center for Neurological Disorders and Department of Neurology, Xuanwu Hospital, Capital Medical University, National Clinical Research Center for Geriatric Diseases, PR China; Beijing Key Laboratory of Geriatric Cognitive Disorders, PR China; Clinical Center for Neurodegenerative Disease and Memory Impairment, Capital Medical University, PR China; Center of Alzheimer's Disease, Beijing Institute of Brain Disorders, Collaborative Innovation Center for Brain Disorders, Capital Medical University, PR China; Key Laboratory of Neurodegenerative Diseases, Ministry of Education, Beijing 100053, PR China
| | - Yan Li
- Innovation Center for Neurological Disorders and Department of Neurology, Xuanwu Hospital, Capital Medical University, National Clinical Research Center for Geriatric Diseases, PR China; Beijing Key Laboratory of Geriatric Cognitive Disorders, PR China; Clinical Center for Neurodegenerative Disease and Memory Impairment, Capital Medical University, PR China; Center of Alzheimer's Disease, Beijing Institute of Brain Disorders, Collaborative Innovation Center for Brain Disorders, Capital Medical University, PR China; Key Laboratory of Neurodegenerative Diseases, Ministry of Education, Beijing 100053, PR China
| | - Jianping Jia
- Innovation Center for Neurological Disorders and Department of Neurology, Xuanwu Hospital, Capital Medical University, National Clinical Research Center for Geriatric Diseases, PR China; Beijing Key Laboratory of Geriatric Cognitive Disorders, PR China; Clinical Center for Neurodegenerative Disease and Memory Impairment, Capital Medical University, PR China; Center of Alzheimer's Disease, Beijing Institute of Brain Disorders, Collaborative Innovation Center for Brain Disorders, Capital Medical University, PR China; Key Laboratory of Neurodegenerative Diseases, Ministry of Education, Beijing 100053, PR China.
| |
Collapse
|
7
|
Pelligra A, Mrugala J, Griess K, Kirschner P, Nortmann O, Bartosinska B, Köster A, Krupenko NI, Gebel D, Westhoff P, Steckel B, Eberhard D, Herebian D, Belgardt BF, Schrader J, Weber APM, Krupenko SA, Lammert E. Pancreatic islet protection at the expense of secretory function involves serine-linked mitochondrial one-carbon metabolism. Cell Rep 2023; 42:112615. [PMID: 37294632 PMCID: PMC10592470 DOI: 10.1016/j.celrep.2023.112615] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2022] [Revised: 03/30/2023] [Accepted: 05/23/2023] [Indexed: 06/11/2023] Open
Abstract
Type 2 diabetes is characterized by insulin hypersecretion followed by reduced glucose-stimulated insulin secretion (GSIS). Here we show that acute stimulation of pancreatic islets with the insulin secretagogue dextrorphan (DXO) or glibenclamide enhances GSIS, whereas chronic treatment with high concentrations of these drugs reduce GSIS but protect islets from cell death. Bulk RNA sequencing of islets shows increased expression of genes for serine-linked mitochondrial one-carbon metabolism (OCM) after chronic, but not acute, stimulation. In chronically stimulated islets, more glucose is metabolized to serine than to citrate, and the mitochondrial ATP/ADP ratio decreases, whereas the NADPH/NADP+ ratio increases. Activating transcription factor-4 (Atf4) is required and sufficient to activate serine-linked mitochondrial OCM genes in islets, with gain- and loss-of-function experiments showing that Atf4 reduces GSIS and is required, but not sufficient, for full DXO-mediated islet protection. In sum, we identify a reversible metabolic pathway that provides islet protection at the expense of secretory function.
Collapse
Affiliation(s)
- Angela Pelligra
- Institute of Metabolic Physiology, Heinrich Heine University, 40225 Düsseldorf, Germany
| | - Jessica Mrugala
- Institute of Metabolic Physiology, Heinrich Heine University, 40225 Düsseldorf, Germany; Institute for Vascular and Islet Cell Biology, German Diabetes Center (DDZ), Leibniz Center for Diabetes Research at Heinrich Heine University, 40225 Düsseldorf, Germany; German Center for Diabetes Research (DZD e.V.), Helmholtz Zentrum München, 85764 Neuherberg, Germany
| | - Kerstin Griess
- Institute of Metabolic Physiology, Heinrich Heine University, 40225 Düsseldorf, Germany
| | - Philip Kirschner
- Institute of Metabolic Physiology, Heinrich Heine University, 40225 Düsseldorf, Germany
| | - Oliver Nortmann
- Institute of Metabolic Physiology, Heinrich Heine University, 40225 Düsseldorf, Germany
| | - Barbara Bartosinska
- Institute of Metabolic Physiology, Heinrich Heine University, 40225 Düsseldorf, Germany
| | - Andrea Köster
- Institute of Metabolic Physiology, Heinrich Heine University, 40225 Düsseldorf, Germany
| | - Natalia I Krupenko
- University of North Carolina (UNC) Nutrition Research Institute, UNC Chapel Hill, Chapel Hill, NC, USA
| | - Dominik Gebel
- Institute of Metabolic Physiology, Heinrich Heine University, 40225 Düsseldorf, Germany
| | - Philipp Westhoff
- Institute of Plant Biochemistry, Cluster of Excellence on Plant Science (CEPLAS), Heinrich Heine University, 40225 Düsseldorf, Germany; Cluster of Excellence on Plant Science (CEPLAS), Heinrich Heine University, 40225 Düsseldorf, Germany
| | - Bodo Steckel
- Department of Molecular Cardiology, Heinrich Heine University, 40225 Düsseldorf, Germany
| | - Daniel Eberhard
- Institute of Metabolic Physiology, Heinrich Heine University, 40225 Düsseldorf, Germany; Institute for Vascular and Islet Cell Biology, German Diabetes Center (DDZ), Leibniz Center for Diabetes Research at Heinrich Heine University, 40225 Düsseldorf, Germany
| | - Diran Herebian
- Department of General Pediatrics, Neonatology and Pediatric Cardiology, Medical Faculty and University Hospital Düsseldorf, Heinrich Heine University, 40225 Düsseldorf, Germany
| | - Bengt-Frederik Belgardt
- Institute for Vascular and Islet Cell Biology, German Diabetes Center (DDZ), Leibniz Center for Diabetes Research at Heinrich Heine University, 40225 Düsseldorf, Germany; German Center for Diabetes Research (DZD e.V.), Helmholtz Zentrum München, 85764 Neuherberg, Germany
| | - Jürgen Schrader
- Department of Molecular Cardiology, Heinrich Heine University, 40225 Düsseldorf, Germany
| | - Andreas P M Weber
- Institute of Plant Biochemistry, Cluster of Excellence on Plant Science (CEPLAS), Heinrich Heine University, 40225 Düsseldorf, Germany; Cluster of Excellence on Plant Science (CEPLAS), Heinrich Heine University, 40225 Düsseldorf, Germany
| | - Sergey A Krupenko
- University of North Carolina (UNC) Nutrition Research Institute, UNC Chapel Hill, Chapel Hill, NC, USA
| | - Eckhard Lammert
- Institute of Metabolic Physiology, Heinrich Heine University, 40225 Düsseldorf, Germany; Institute for Vascular and Islet Cell Biology, German Diabetes Center (DDZ), Leibniz Center for Diabetes Research at Heinrich Heine University, 40225 Düsseldorf, Germany; German Center for Diabetes Research (DZD e.V.), Helmholtz Zentrum München, 85764 Neuherberg, Germany.
| |
Collapse
|
8
|
Noguera Hurtado H, Gresch A, Düfer M. NMDA receptors - regulatory function and pathophysiological significance for pancreatic beta cells. Biol Chem 2023; 404:311-324. [PMID: 36626848 DOI: 10.1515/hsz-2022-0236] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2022] [Accepted: 11/29/2022] [Indexed: 01/11/2023]
Abstract
Due to its unique features amongst ionotropic glutamate receptors, the NMDA receptor is of special interest in the physiological context but even more as a drug target. In the pathophysiology of metabolic disorders, particularly type 2 diabetes mellitus, there is evidence that NMDA receptor activation contributes to disease progression by impairing beta cell function. Consequently, channel inhibitors are suggested for treatment, but up to now there are many unanswered questions about the signaling pathways NMDA receptors are interfering with in the islets of Langerhans. In this review we give an overview about channel structure and function with special regard to the pancreatic beta cells and the regulation of insulin secretion. We sum up which signaling pathways from brain research have already been transferred to the beta cell, and what still needs to be proven. The main focus is on the relationship between an over-stimulated NMDA receptor and the production of reactive oxygen species, the amount of which is crucial for beta cell function. Finally, pilot studies using NMDA receptor blockers to protect the islet from dysfunction are reviewed and future perspectives for the use of such compounds in the context of impaired glucose homeostasis are discussed.
Collapse
Affiliation(s)
- Héctor Noguera Hurtado
- Institute of Pharmaceutical and Medicinal Chemistry, Department of Pharmacology, University of Münster, Corrensstraße 48, D-48149 Münster, Germany
| | - Anne Gresch
- Institute of Pharmaceutical and Medicinal Chemistry, Department of Pharmacology, University of Münster, Corrensstraße 48, D-48149 Münster, Germany
| | - Martina Düfer
- Institute of Pharmaceutical and Medicinal Chemistry, Department of Pharmacology, University of Münster, Corrensstraße 48, D-48149 Münster, Germany
| |
Collapse
|
9
|
Huang XT, Xiong DY, Xiao JN, Deng L, Liu W, Tang SY. Kindlin-2 protects pancreatic β cells through inhibiting NLRP3 inflammasome activation in diabetic mice. Biochem Biophys Res Commun 2022; 614:1-8. [PMID: 35567938 DOI: 10.1016/j.bbrc.2022.04.131] [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: 04/21/2022] [Accepted: 04/27/2022] [Indexed: 11/02/2022]
Abstract
Diabetes mellitus has been a major public health problem worldwide, characterized by insulin resistance and dysfunction of β-cells. A previous study showed that Kindlin-2 loss in β-cells dramatically reduces insulin secretion and decreases β-cell mass, resulting in severe diabetes-like phenotypes. It suggests that Kindlin-2 in β-cells play an important role in regulating glucose homeostasis. However, the effect of Kindlin-2 on the function of β-cells under chronic hyperglycemia in diabetes has not been explored. Here we report that Kindlin-2 overexpression ameliorates diabetes and improves insulin secretion in mice induced by streptozocin. In contrast, Kindlin-2 insufficiency exacerbates diabetes and promotes β-cells dysfunction and inflammation in β-cells induced by a high-fat diet (HFD). In vitro, Kindlin-2 overexpression prevented high-glucose (HG)-induced dysfunction in β-cells. Kindlin-2 overexpression also decreased the expression of pro-inflammatory cytokines and NLRP3 inflammasome expression in β-cells exposed to HG. Furthermore, the loss of Kindlin-2 aggravates the expression of inflammatory cytokines and NLRP3 induced by HG in β-cells. Collectively, we demonstrate that Kindlin-2 protects against diabetes by inhibiting NLRP3 inflammasome activation.
Collapse
Affiliation(s)
- Xiao-Ting Huang
- Xiangya Nursing School, Central South University, Changsha, Hunan, 410013, China
| | - Da-Yan Xiong
- Xiangya Nursing School, Central South University, Changsha, Hunan, 410013, China
| | - Jin-Nan Xiao
- Xiangya Nursing School, Central South University, Changsha, Hunan, 410013, China
| | - Lang Deng
- Xiangya Nursing School, Central South University, Changsha, Hunan, 410013, China
| | - Wei Liu
- Xiangya Nursing School, Central South University, Changsha, Hunan, 410013, China.
| | - Si-Yuan Tang
- Xiangya Nursing School, Central South University, Changsha, Hunan, 410013, China.
| |
Collapse
|
10
|
Gresch A, Hurtado HN, Wörmeyer L, De Luca V, Wiggers R, Seebohm G, Wünsch B, Düfer M. Selective Inhibition of N-Methyl-d-aspartate Receptors with GluN2B Subunit Protects β Cells against Stress-Induced Apoptotic Cell Death. J Pharmacol Exp Ther 2021; 379:235-244. [PMID: 34593560 DOI: 10.1124/jpet.121.000807] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2021] [Accepted: 09/28/2021] [Indexed: 11/22/2022] Open
Abstract
Participation of N-methyl-d-aspartate (NMDA) receptors (NMDARs) in the failure of pancreatic β cells during development of type 2 diabetes mellitus is discussed. Our study investigates whether β cell mass and function can be preserved by selectively addressing the GluN2B subunit of the NMDAR. NMDAR activation by NMDA and its coagonist glycine moderately influenced electrical activity and Ca2+ handling in islet cells at a threshold glucose concentration (4-5 mM) without affecting glucose-mediated insulin secretion. Exposure of islet cells to NMDA/glycine or a glucolipotoxic milieu increased apoptosis by 5% and 8%, respectively. The GluN2B-specific NMDAR antagonist WMS-1410 (0.1 and 1 µM) partly protected against this. In addition, WMS-1410 completely prevented the decrease in insulin secretion of about 32% provoked by a 24-hour-treatment with NMDA/glycine. WMS-1410 eliminated NMDA-induced changes in the oxidation status of the islet cells and elevated the sensitivity of intracellular calcium to 15 mM glucose. By contrast, WMS-1410 did not prevent the decline in glucose-stimulated insulin secretion occurring after glucolipotoxic culture. This lack of effect was due to a decrease in insulin content to 18% that obviously could not be compensated by the preservation of cell mass or the higher percentage of insulin release in relation to insulin content. In conclusion, the negative effects of permanent NMDAR activation were effectively counteracted by WMS-1410 as well as the apoptotic cell death induced by high glucose and lipid concentrations. Modulation of NMDARs containing the GluN2B subunit is suggested to preserve β cell mass during development of type 2 diabetes mellitus. SIGNIFICANCE STATEMENT: Addressing NMDA receptors containing the GluN2B subunit in pancreatic islet cells has the potential to protect the β cell mass that progressively declines during the development of type 2 diabetes. Furthermore, this study shows that harmful effects of permanent NMDAR activation can be effectively counteracted by the compound WMS-1410, a selective modulator for NMDARs containing the GluN2B subunit.
Collapse
Affiliation(s)
- Anne Gresch
- Pharmaceutical and Medicinal Chemistry, Department of Pharmacology (A.G., H.N.H., L.W., V.D.L., R.W., M.D.), and Pharmaceutical and Medicinal Chemistry (B.W.), PharmaCampus, University of Münster, Münster, Germany; and Institute for Genetics of Heart Diseases (IfGH), Department of Cardiovascular Medicine, University Hospital Münster, Münster, Germany (G.S.)
| | - Héctor Noguera Hurtado
- Pharmaceutical and Medicinal Chemistry, Department of Pharmacology (A.G., H.N.H., L.W., V.D.L., R.W., M.D.), and Pharmaceutical and Medicinal Chemistry (B.W.), PharmaCampus, University of Münster, Münster, Germany; and Institute for Genetics of Heart Diseases (IfGH), Department of Cardiovascular Medicine, University Hospital Münster, Münster, Germany (G.S.)
| | - Laura Wörmeyer
- Pharmaceutical and Medicinal Chemistry, Department of Pharmacology (A.G., H.N.H., L.W., V.D.L., R.W., M.D.), and Pharmaceutical and Medicinal Chemistry (B.W.), PharmaCampus, University of Münster, Münster, Germany; and Institute for Genetics of Heart Diseases (IfGH), Department of Cardiovascular Medicine, University Hospital Münster, Münster, Germany (G.S.)
| | - Vivien De Luca
- Pharmaceutical and Medicinal Chemistry, Department of Pharmacology (A.G., H.N.H., L.W., V.D.L., R.W., M.D.), and Pharmaceutical and Medicinal Chemistry (B.W.), PharmaCampus, University of Münster, Münster, Germany; and Institute for Genetics of Heart Diseases (IfGH), Department of Cardiovascular Medicine, University Hospital Münster, Münster, Germany (G.S.)
| | - Rebekka Wiggers
- Pharmaceutical and Medicinal Chemistry, Department of Pharmacology (A.G., H.N.H., L.W., V.D.L., R.W., M.D.), and Pharmaceutical and Medicinal Chemistry (B.W.), PharmaCampus, University of Münster, Münster, Germany; and Institute for Genetics of Heart Diseases (IfGH), Department of Cardiovascular Medicine, University Hospital Münster, Münster, Germany (G.S.)
| | - Guiscard Seebohm
- Pharmaceutical and Medicinal Chemistry, Department of Pharmacology (A.G., H.N.H., L.W., V.D.L., R.W., M.D.), and Pharmaceutical and Medicinal Chemistry (B.W.), PharmaCampus, University of Münster, Münster, Germany; and Institute for Genetics of Heart Diseases (IfGH), Department of Cardiovascular Medicine, University Hospital Münster, Münster, Germany (G.S.)
| | - Bernhard Wünsch
- Pharmaceutical and Medicinal Chemistry, Department of Pharmacology (A.G., H.N.H., L.W., V.D.L., R.W., M.D.), and Pharmaceutical and Medicinal Chemistry (B.W.), PharmaCampus, University of Münster, Münster, Germany; and Institute for Genetics of Heart Diseases (IfGH), Department of Cardiovascular Medicine, University Hospital Münster, Münster, Germany (G.S.)
| | - Martina Düfer
- Pharmaceutical and Medicinal Chemistry, Department of Pharmacology (A.G., H.N.H., L.W., V.D.L., R.W., M.D.), and Pharmaceutical and Medicinal Chemistry (B.W.), PharmaCampus, University of Münster, Münster, Germany; and Institute for Genetics of Heart Diseases (IfGH), Department of Cardiovascular Medicine, University Hospital Münster, Münster, Germany (G.S.)
| |
Collapse
|
11
|
Scholz O, Otter S, Welters A, Wörmeyer L, Dolenšek J, Klemen MS, Pohorec V, Eberhard D, Mrugala J, Hamacher A, Koch A, Sanz M, Hoffmann T, Hogeback J, Herebian D, Klöcker N, Piechot A, Mayatepek E, Meissner T, Stožer A, Lammert E. Peripherally active dextromethorphan derivatives lower blood glucose levels by targeting pancreatic islets. Cell Chem Biol 2021; 28:1474-1488.e7. [PMID: 34118188 DOI: 10.1016/j.chembiol.2021.05.011] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2020] [Revised: 03/09/2021] [Accepted: 05/17/2021] [Indexed: 12/12/2022]
Abstract
Dextromethorphan (DXM) acts as cough suppressant via its central action. Cell-protective effects of this drug have been reported in peripheral tissues, making DXM potentially useful for treatment of several common human diseases, such as type 2 diabetes mellitus (T2DM). Pancreatic islets are among the peripheral tissues that positively respond to DXM, and anti-diabetic effects of DXM were observed in two placebo-controlled, randomized clinical trials in humans with T2DM. Since these effects were associated with central side effects, we here developed chemical derivatives of DXM that pass the blood-brain barrier to a significantly lower extent than the original drug. We show that basic nitrogen-containing residues block central adverse events of DXM without reducing its anti-diabetic effects, including the protection of human pancreatic islets from cell death. These results show how to chemically modify DXM, and possibly other morphinans, as to exclude central side effects, while targeting peripheral tissues, such as pancreatic islets.
Collapse
Affiliation(s)
- Okka Scholz
- Institute for Vascular and Islet Cell Biology, German Diabetes Center (DDZ), Leibniz Center for Diabetes Research at Heinrich Heine University, 40225 Düsseldorf, Germany; Institute of Metabolic Physiology, Heinrich Heine University, 40225 Düsseldorf, Germany; Center of Competence for Innovative Diabetes Therapy (KomIT), German Diabetes Center (DDZ), 40225 Düsseldorf, Germany; German Center for Diabetes Research (DZD e.V.), Helmholtz Zentrum München, 85764 Neuherberg, Germany
| | - Silke Otter
- Institute for Vascular and Islet Cell Biology, German Diabetes Center (DDZ), Leibniz Center for Diabetes Research at Heinrich Heine University, 40225 Düsseldorf, Germany; Institute of Metabolic Physiology, Heinrich Heine University, 40225 Düsseldorf, Germany; Center of Competence for Innovative Diabetes Therapy (KomIT), German Diabetes Center (DDZ), 40225 Düsseldorf, Germany
| | - Alena Welters
- Institute of Metabolic Physiology, Heinrich Heine University, 40225 Düsseldorf, Germany; Department of General Pediatrics, Neonatology and Pediatric Cardiology, Medical Faculty and University Hospital Düsseldorf, Heinrich Heine University, 40225 Düsseldorf, Germany
| | - Laura Wörmeyer
- Institute of Metabolic Physiology, Heinrich Heine University, 40225 Düsseldorf, Germany; Department of General Pediatrics, Neonatology and Pediatric Cardiology, Medical Faculty and University Hospital Düsseldorf, Heinrich Heine University, 40225 Düsseldorf, Germany
| | - Jurij Dolenšek
- Faculty of Medicine, University of Maribor, Taborska ulica 8, 2000 Maribor, Slovenia; Faculty of Natural Sciences and Mathematics, University of Maribor, Koroška cesta 160, 2000 Maribor, Slovenia
| | - Maša Skelin Klemen
- Faculty of Medicine, University of Maribor, Taborska ulica 8, 2000 Maribor, Slovenia
| | - Viljem Pohorec
- Faculty of Medicine, University of Maribor, Taborska ulica 8, 2000 Maribor, Slovenia
| | - Daniel Eberhard
- Institute for Vascular and Islet Cell Biology, German Diabetes Center (DDZ), Leibniz Center for Diabetes Research at Heinrich Heine University, 40225 Düsseldorf, Germany; Institute of Metabolic Physiology, Heinrich Heine University, 40225 Düsseldorf, Germany
| | - Jessica Mrugala
- Institute for Vascular and Islet Cell Biology, German Diabetes Center (DDZ), Leibniz Center for Diabetes Research at Heinrich Heine University, 40225 Düsseldorf, Germany; Institute of Metabolic Physiology, Heinrich Heine University, 40225 Düsseldorf, Germany; German Center for Diabetes Research (DZD e.V.), Helmholtz Zentrum München, 85764 Neuherberg, Germany
| | - Anna Hamacher
- Institute for Vascular and Islet Cell Biology, German Diabetes Center (DDZ), Leibniz Center for Diabetes Research at Heinrich Heine University, 40225 Düsseldorf, Germany; Institute of Metabolic Physiology, Heinrich Heine University, 40225 Düsseldorf, Germany
| | - Angela Koch
- Institute of Neuro- and Sensory Physiology, Medical Faculty and University Hospital Düsseldorf, Heinrich Heine University, 40225 Düsseldorf, Germany
| | - Miguel Sanz
- Center of Competence for Innovative Diabetes Therapy (KomIT), German Diabetes Center (DDZ), 40225 Düsseldorf, Germany; Taros Chemicals GmbH & Co. KG, 44227 Dortmund, Germany
| | - Torsten Hoffmann
- Center of Competence for Innovative Diabetes Therapy (KomIT), German Diabetes Center (DDZ), 40225 Düsseldorf, Germany; Taros Chemicals GmbH & Co. KG, 44227 Dortmund, Germany
| | - Jens Hogeback
- A&M Labor für Analytik und Metabolismusforschung Service GmbH, 50126 Bergheim, Germany
| | - Diran Herebian
- Department of General Pediatrics, Neonatology and Pediatric Cardiology, Medical Faculty and University Hospital Düsseldorf, Heinrich Heine University, 40225 Düsseldorf, Germany
| | - Nikolaj Klöcker
- Institute of Neuro- and Sensory Physiology, Medical Faculty and University Hospital Düsseldorf, Heinrich Heine University, 40225 Düsseldorf, Germany
| | - Alexander Piechot
- Center of Competence for Innovative Diabetes Therapy (KomIT), German Diabetes Center (DDZ), 40225 Düsseldorf, Germany; Taros Chemicals GmbH & Co. KG, 44227 Dortmund, Germany
| | - Ertan Mayatepek
- Department of General Pediatrics, Neonatology and Pediatric Cardiology, Medical Faculty and University Hospital Düsseldorf, Heinrich Heine University, 40225 Düsseldorf, Germany
| | - Thomas Meissner
- Department of General Pediatrics, Neonatology and Pediatric Cardiology, Medical Faculty and University Hospital Düsseldorf, Heinrich Heine University, 40225 Düsseldorf, Germany
| | - Andraž Stožer
- Faculty of Medicine, University of Maribor, Taborska ulica 8, 2000 Maribor, Slovenia
| | - Eckhard Lammert
- Institute for Vascular and Islet Cell Biology, German Diabetes Center (DDZ), Leibniz Center for Diabetes Research at Heinrich Heine University, 40225 Düsseldorf, Germany; Institute of Metabolic Physiology, Heinrich Heine University, 40225 Düsseldorf, Germany; Center of Competence for Innovative Diabetes Therapy (KomIT), German Diabetes Center (DDZ), 40225 Düsseldorf, Germany; German Center for Diabetes Research (DZD e.V.), Helmholtz Zentrum München, 85764 Neuherberg, Germany.
| |
Collapse
|
12
|
Activation of TREM-1 induces endoplasmic reticulum stress through IRE-1α/XBP-1s pathway in murine macrophages. Mol Immunol 2021; 135:294-303. [PMID: 33957479 DOI: 10.1016/j.molimm.2021.04.023] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2021] [Revised: 04/16/2021] [Accepted: 04/26/2021] [Indexed: 12/14/2022]
Abstract
Increasing evidence suggests that endoplasmic reticulum (ER) stress activates several pro-inflammatory signaling pathways in many diseases, including acute lung injury (ALI). We have reported that blocking triggering receptor expressed on myeloid cells 1 (TREM-1) protects against ALI by suppressing pulmonary inflammation in mice with ALI induced by lipopolysaccharides (LPS). However, the molecular mechanism underlying the TREM-1-induced pro-inflammatory microenvironment in macrophages remains unclearly. Herein, we aimed to determine whether TREM-1 regulates the inflammatory responses induced by LPS associated with ER stress activation. We found that the activation of TREM-1 by a monoclonal agonist antibody (anti-TREM-1) increased the mRNA and protein levels of IL-1β, TNF-α, and IL-6 in primary macrophages. Treatment of the anti-TREM-1 antibody increased the expression of ER stress markers (ATF6, PERK, IRE-1α, and XBP-1s) in primary macrophages. While pretreatment with 4-PBA, an inhibitor of ER stress, significantly inhibited the expression of ER stress markers and pro-inflammatory cytokines and reduced LDH release. Furthermore, inhibiting the activity of the IRE-1α/XBP-1s pathway by STF-083010 significantly mitigated the increased levels of IL-1β, TNF-α, and IL-6 in macrophages treated by the anti-TREM-1 antibody. XBP-1 silencing attenuated pro-inflammatory microenvironment evoked by activation of TREM-1. Besides, we found that blockade of TREM-1 with LR12 ameliorated ER stress induced by LPS in vitro and in vivo. In conclusion, we conclude that TREM-1 activation induces ER stress through the IRE-1α/XBP-1s pathway in macrophages, contributing to the pro-inflammatory microenvironment.
Collapse
|
13
|
Banerjee A, Mukherjee S, Maji BK. Worldwide flavor enhancer monosodium glutamate combined with high lipid diet provokes metabolic alterations and systemic anomalies: An overview. Toxicol Rep 2021; 8:938-961. [PMID: 34026558 PMCID: PMC8120859 DOI: 10.1016/j.toxrep.2021.04.009] [Citation(s) in RCA: 42] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2020] [Revised: 04/20/2021] [Accepted: 04/25/2021] [Indexed: 12/13/2022] Open
Abstract
Flavor enhancing high lipid diet acts as silent killer. Monosodium glutamate mixed with high lipid diet alters redox-status. Monosodium glutamate mixed with high lipid diet induces systemic anomalies.
In this fast-food era, people depend on ready-made foods and engage in minimal physical activities that ultimately change their food habits. Majorities of such foods have harmful effects on human health due to higher percentages of saturated fatty acids, trans-fatty acids, and hydrogenated fats in the form of high lipid diet (HLD). Moreover, food manufacturers add monosodium glutamate (MSG) to enhance the taste and palatability of the HLD. Both MSG and HLD induce the generation of reactive oxygen species (ROS) and thereby alter the redox-homeostasis to cause systemic damage. However, MSG mixed HLD (MH) consumption leads to dyslipidemia, silently develops non-alcoholic fatty liver disease followed by metabolic alterations and systemic anomalies, even malignancies, via modulating different signaling pathways. This comprehensive review formulates health care strategies to create global awareness about the harmful impact of MH on the human body and recommends the daily consumption of more natural foods rich in antioxidants instead of toxic ingredients to counterbalance the MH-induced systemic anomalies.
Collapse
|
14
|
Lockridge A, Gustafson E, Wong A, Miller RF, Alejandro EU. Acute D-Serine Co-Agonism of β-Cell NMDA Receptors Potentiates Glucose-Stimulated Insulin Secretion and Excitatory β-Cell Membrane Activity. Cells 2021; 10:E93. [PMID: 33430405 PMCID: PMC7826616 DOI: 10.3390/cells10010093] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2020] [Revised: 12/18/2020] [Accepted: 01/04/2021] [Indexed: 02/06/2023] Open
Abstract
Insulin-secreting pancreatic β-cells express proteins characteristic of D-serine regulated synapses, but the acute effect of D-serine co-agonism on its presumptive β-cell target, N-methyl D-aspartate receptors (NMDARs), is unclear. We used multiple models to evaluate glucose homeostasis and insulin secretion in mice with a systemic increase in D-serine (intraperitoneal injection or DAAO mutants without D-serine catabolism) or tissue-specific loss of Grin1-encoded GluN1, the D-serine binding NMDAR subunit. We also investigated the effects of D-serine ± NMDA on glucose-stimulated insulin secretion (GSIS) and β-cell depolarizing membrane oscillations, using perforated patch electrophysiology, in β-cell-containing primary isolated mouse islets. In vivo models of elevated D-serine correlated to improved blood glucose and insulin levels. In vitro, D-serine potentiated GSIS and β-cell membrane excitation, dependent on NMDAR activating conditions including GluN1 expression (co-agonist target), simultaneous NMDA (agonist), and elevated glucose (depolarization). Pancreatic GluN1-loss females were glucose intolerant and GSIS was depressed in islets from younger, but not older, βGrin1 KO mice. Thus, D-serine is capable of acute antidiabetic effects in mice and potentiates insulin secretion through excitatory β-cell NMDAR co-agonism but strain-dependent shifts in potency and age/sex-specific Grin1-loss phenotypes suggest that context is critical to the interpretation of data on the role of D-serine and NMDARs in β-cell function.
Collapse
Affiliation(s)
- Amber Lockridge
- Department of Integrative Biology and Physiology, University of Minnesota, Minneapolis, MN 55455, USA; (A.L.); (E.G.); (A.W.)
| | - Eric Gustafson
- Department of Integrative Biology and Physiology, University of Minnesota, Minneapolis, MN 55455, USA; (A.L.); (E.G.); (A.W.)
- Department of Neuroscience, University of Minnesota, Minneapolis, MN 55455, USA;
| | - Alicia Wong
- Department of Integrative Biology and Physiology, University of Minnesota, Minneapolis, MN 55455, USA; (A.L.); (E.G.); (A.W.)
| | - Robert F. Miller
- Department of Neuroscience, University of Minnesota, Minneapolis, MN 55455, USA;
| | - Emilyn U. Alejandro
- Department of Integrative Biology and Physiology, University of Minnesota, Minneapolis, MN 55455, USA; (A.L.); (E.G.); (A.W.)
| |
Collapse
|
15
|
Huang XT, Yang JX, Wang Z, Zhang CY, Luo ZQ, Liu W, Tang SY. Activation of N-methyl-D-aspartate receptor regulates insulin sensitivity and lipid metabolism. Theranostics 2021; 11:2247-2262. [PMID: 33500723 PMCID: PMC7797674 DOI: 10.7150/thno.51666] [Citation(s) in RCA: 29] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2020] [Accepted: 11/19/2020] [Indexed: 12/13/2022] Open
Abstract
RATIONALE Although significant progress has been made in understanding the mechanisms of steatosis and insulin resistance, the physiological functions of regulators in these processes remain largely elusive. Evidence has suggested that the glutamate/N-methyl-D-aspartic acid receptor (NMDAR) axis contributes to acute lung injury, pulmonary arterial hypertension, and diabetes, but the specific metabolic contribution of the glutamate/NMDAR axis is not clear. Here we provide data at the animal, cellular, and molecular levels to support the role of the glutamate/NMDAR axis as a therapeutic target for metabolic syndrome in obesity. Methods: We examined the glutamate level in the obese mouse induced by a high-fat diet (HFD) for 12 weeks. To assess the role of NMDAR in insulin sensitivity and lipid metabolism, we tested the effects of Memantine (an NMDAR antagonist) and NMDA (an NMDAR agonist) on mice fed with HFD or standard chow diet. The in vitros NMDAR roles were analyzed in hepatocytes and potential mechanisms involved in regulating lipid metabolism were investigated. Results: Glutamate was increased in the serum of HFD-treated mice. The NMDAR blockade by Memantine decreased the susceptibility to insulin resistance and hepatic steatosis in obese mice. NMDA treatment for 6 months induced obesity in mice, characterized by hyperglycemia, hyperlipidemia, insulin resistance, and pathological changes in the liver. We provided in vitro evidence demonstrating that NMDAR activation facilitated metabolic syndrome in obesity through promoting lipid accumulation. NMDAR inhibition attenuated lipid accumulation induced by palmitic acid. Mechanistically, NMDAR activation impaired fatty acid oxidation by reducing PPARα phosphorylation and activity. The PPARα activity reduction induced by NMDAR activation was reversibly mediated by ERK1/2 signaling. Conclusion: These findings revealed that targeting NMDAR might be a promising therapeutic strategy for metabolic syndrome in obesity.
Collapse
Affiliation(s)
- Xiao-Ting Huang
- Xiangya Nursing School, Central South University, Changsha, Hunan, China
| | - Jun-Xiao Yang
- Department of Orthopedics, Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Zun Wang
- Xiangya Nursing School, Central South University, Changsha, Hunan, China
| | - Chen-Yu Zhang
- Department of Physiology, School of Basic Medicine Science, Central South University, Changsha, Hunan, China
| | - Zi-Qiang Luo
- Department of Physiology, School of Basic Medicine Science, Central South University, Changsha, Hunan, China
| | - Wei Liu
- Xiangya Nursing School, Central South University, Changsha, Hunan, China
| | - Si-Yuan Tang
- Xiangya Nursing School, Central South University, Changsha, Hunan, China
| |
Collapse
|
16
|
Yang HH, Duan JX, Liu SK, Xiong JB, Guan XX, Zhong WJ, Sun CC, Zhang CY, Luo XQ, Zhang YF, Chen P, Hammock BD, Hwang SH, Jiang JX, Zhou Y, Guan CX. A COX-2/sEH dual inhibitor PTUPB alleviates lipopolysaccharide-induced acute lung injury in mice by inhibiting NLRP3 inflammasome activation. Theranostics 2020; 10:4749-4761. [PMID: 32308747 PMCID: PMC7163435 DOI: 10.7150/thno.43108] [Citation(s) in RCA: 141] [Impact Index Per Article: 28.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2019] [Accepted: 03/08/2020] [Indexed: 01/11/2023] Open
Abstract
Rationale: Dysregulation of arachidonic acid (ARA) metabolism results in inflammation; however, its role in acute lung injury (ALI) remains elusive. In this study, we addressed the role of dysregulated ARA metabolism in cytochromes P450 (CYPs) /cyclooxygenase-2 (COX-2) pathways in the pathogenesis of lipopolysaccharide (LPS)-induced ALI in mice. Methods: The metabolism of CYPs/COX-2-derived ARA in the lungs of LPS-induced ALI was investigated in C57BL/6 mice. The COX-2/sEH dual inhibitor PTUPB was used to establish the function of CYPs/COX-2 dysregulation in ALI. Primary murine macrophages were used to evaluate the underlying mechanism of PTUPB involved in the activation of NLRP3 inflammasome in vitro. Results: Dysregulation of CYPs/COX-2 metabolism of ARA occurred in the lungs and in primary macrophages under the LPS challenge. Decrease mRNA expression of Cyp2j9, Cyp2j6, and Cyp2j5 was observed, which metabolize ARA into epoxyeicosatrienoic acids (EETs). The expressions of COX-2 and soluble epoxide hydrolase (sEH), on the other hand, was significantly upregulated. Pre-treatment with the dual COX-2 and sEH inhibitor, PTUPB, attenuated the pathological injury of lung tissues and reduced the infiltration of inflammatory cells. Furthermore, PTUPB decreased the pro-inflammatory factors, oxidative stress, and activation of NACHT, LRR, and PYD domains-containing protein 3 (NLRP3) inflammasome in LPS-induced ALI mice. PTUPB pre-treatment remarkably reduced the activation of macrophages and NLRP3 inflammasome in vitro. Significantly, both preventive and therapeutic treatment with PTUPB improved the survival rate of mice receiving a lethal dose of LPS. Conclusion: The dysregulation of CYPs/COX-2 metabolized ARA contributes to the uncontrolled inflammatory response in ALI. The dual COX-2 and sEH inhibitor PTUPB exerts anti-inflammatory effects in treating ALI by inhibiting the NLRP3 inflammasome activation.
Collapse
Affiliation(s)
- Hui-Hui Yang
- Department of Physiology, Xiangya School of Medicine, Central South University, Changsha, Hunan 410078, China
| | - Jia-Xi Duan
- Department of Pulmonary and Critical Care Medicine, the Second Xiangya Hospital, Central South University, Changsha, Hunan 410011, China
- Research Unit of Respiratory Disease, Central South University, Changsha, Hunan 410011, China
- Hunan Diagnosis and Treatment Center of Respiratory Disease, Central South University, Changsha, Hunan 410011, China
| | - Shao-Kun Liu
- Department of Pulmonary and Critical Care Medicine, the Second Xiangya Hospital, Central South University, Changsha, Hunan 410011, China
- Research Unit of Respiratory Disease, Central South University, Changsha, Hunan 410011, China
- Hunan Diagnosis and Treatment Center of Respiratory Disease, Central South University, Changsha, Hunan 410011, China
| | - Jian-Bing Xiong
- Department of Physiology, Xiangya School of Medicine, Central South University, Changsha, Hunan 410078, China
| | - Xin-Xin Guan
- Department of Physiology, Xiangya School of Medicine, Central South University, Changsha, Hunan 410078, China
| | - Wen-Jing Zhong
- Department of Physiology, Xiangya School of Medicine, Central South University, Changsha, Hunan 410078, China
| | - Chen-Chen Sun
- Department of Physiology, Xiangya School of Medicine, Central South University, Changsha, Hunan 410078, China
| | - Chen-Yu Zhang
- Department of Physiology, Xiangya School of Medicine, Central South University, Changsha, Hunan 410078, China
| | - Xiao-Qin Luo
- Department of Physiology, Xiangya School of Medicine, Central South University, Changsha, Hunan 410078, China
| | - Yan-Feng Zhang
- Department of Physiology, Xiangya School of Medicine, Central South University, Changsha, Hunan 410078, China
| | - Ping Chen
- Department of Pulmonary and Critical Care Medicine, the Second Xiangya Hospital, Central South University, Changsha, Hunan 410011, China
- Research Unit of Respiratory Disease, Central South University, Changsha, Hunan 410011, China
- Hunan Diagnosis and Treatment Center of Respiratory Disease, Central South University, Changsha, Hunan 410011, China
| | - Bruce D. Hammock
- Department of Entomology and Nematology and UC Davis Comprehensive Cancer Center, University of California, Davis, One Shields Avenue, Davis, CA 95616, USA
| | - Sung Hee Hwang
- Department of Entomology and Nematology and UC Davis Comprehensive Cancer Center, University of California, Davis, One Shields Avenue, Davis, CA 95616, USA
| | - Jian-Xin Jiang
- State Key Laboratory of Trauma, Burns, and Combined Injury, Army Medical University, Chongqing, 400038, China
| | - Yong Zhou
- Department of Physiology, Xiangya School of Medicine, Central South University, Changsha, Hunan 410078, China
| | - Cha-Xiang Guan
- Department of Physiology, Xiangya School of Medicine, Central South University, Changsha, Hunan 410078, China
| |
Collapse
|
17
|
Huang XT, Liu W, Zhou Y, Sun M, Yang HH, Zhang CY, Tang SY. Galectin-1 ameliorates lipopolysaccharide-induced acute lung injury via AMPK-Nrf2 pathway in mice. Free Radic Biol Med 2020; 146:222-233. [PMID: 31711983 DOI: 10.1016/j.freeradbiomed.2019.11.011] [Citation(s) in RCA: 92] [Impact Index Per Article: 18.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/23/2019] [Revised: 11/01/2019] [Accepted: 11/07/2019] [Indexed: 12/30/2022]
Abstract
Inflammation and oxidative stress contribute to the progression of acute lung injury (ALI). Galectin-1 (Gal-1) has important anti-inflammatory properties in renal ischemia-reperfusion injury, arthritis, uveitis, and hepatitis. However, whether Gal-1 could protect against ALI is still poorly elucidated. The current study aimed to investigate the protective effects of Gal-1 against lipopolysaccharide (LPS)-induced ALI and the underlying mechanisms. Accordingly, we found that pretreatment with Gal-1 attenuated the lung tissue injury induced by LPS, with the recovery of lung function, protecting against the production of pro-inflammatory cytokines and oxidative stress. We also confirmed the therapeutic potential of Gal-1 on the survival rate of LPS-challenged mice. In vitro studies demonstrated the protective effects of exogenous Gal-1 through downregulating pro-inflammatory cytokines release and oxidative stress in primary macrophages challenged by LPS. In addition, Gal-1 suppressed TXNIP-NLRP3 inflammasome activation in ALI mice and LPS-treated primary macrophages partly through directly binding to the NLRP3 protein. Gal-1 alleviated LPS-induced lung injury via activation of Nrf-2, which may be associated with AMPK phosphorylation. Collectively, our experimental results firstly provided the support that Gal-1 effectively protected against LPS-induced ALI via suppression of inflammation response and oxidative stress, which were largely dependent on the upregulation of the Nrf2 pathway via phosphorylation of AMPK. These results suggest that Gal-1 could be a valuable therapeutic candidate in the treatment of ALI.
Collapse
Affiliation(s)
- Xiao-Ting Huang
- Xiangya Nursing School, Central South University, Changsha, Hunan, China
| | - Wei Liu
- Xiangya Nursing School, Central South University, Changsha, Hunan, China
| | - Yong Zhou
- Department of Physiology, Xiangya School of Medicine, Central South University, Changsha, Hunan, China
| | - Mei Sun
- Xiangya Nursing School, Central South University, Changsha, Hunan, China
| | - Hui-Hui Yang
- Department of Physiology, Xiangya School of Medicine, Central South University, Changsha, Hunan, China
| | - Chen-Yu Zhang
- Department of Physiology, Xiangya School of Medicine, Central South University, Changsha, Hunan, China
| | - Si-Yuan Tang
- Xiangya Nursing School, Central South University, Changsha, Hunan, China.
| |
Collapse
|
18
|
Hu R, Peng GQ, Ban DY, Zhang C, Zhang XQ, Li YP. High-Expression of Neuropilin 1 Correlates to Estrogen-Induced Epithelial-Mesenchymal Transition of Endometrial Cells in Adenomyosis. Reprod Sci 2020; 27:395-403. [PMID: 32046395 DOI: 10.1007/s43032-019-00035-2] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2018] [Accepted: 05/06/2019] [Indexed: 12/22/2022]
Abstract
Epithelial-mesenchymal transition (EMT) induced by estrogen contributes to the development of adenomyosis. However, the exact underlying mechanism remains mostly obscure. We hypothesized that a transmembrane glycoprotein neuropilin 1 (NRP1) was critical in the EMT induced by estrogen, accelerating the development of adenomyosis. We firstly investigated the expression pattern of NRP1 in endometrium samples from women with adenomyosis. We found that NRP1 expression was significantly increased in the endometrium of uterine adenomyosis, especially in the ectopic endometrium. To determine the role of NRP1 in the EMT in endometrial cells, we used an NRP1 overexpression retrovirus to up-regulate the NPR1 expression in human endometrial cells (HEC-1-A). Endometrial cells infected with NRP1 retroviruses showed a high expression of NRP1 and exerted a mesenchymal phenotype, characterized by down-regulation of E-cadherin and Occludin, up-regulation of α-SMA and N-cadherin, and enhanced migration. Then, we found that 17β-estradiol (E2) up-regulated the expression of NRP1 in endometrial cells in a dose-dependent manner, which was eliminated by raloxifene, a selective estrogen receptor inhibitor. Importantly, NRP1 shRNA significantly suppressed the EMT induced by E2 in endometrial cells. And NRP1 shRNA significantly inhibited the phosphorylation of Smad3 and restored the expressions of Slug and Snail1 mRNA. Collectively, these data highlight the possible role of NRP1 in the EMT in the development of adenomyosis and provide a potential therapeutic target for adenomyosis patients.
Collapse
Affiliation(s)
- Rong Hu
- Department of Obstetrics and Gynecology, Xiangya Hospital, Central South University, Changsha, 410008, Hunan, China
| | - Guo-Qing Peng
- Department of Obstetrics and Gynecology, Xiangya Hospital, Central South University, Changsha, 410008, Hunan, China
| | - De-Ying Ban
- Department of Obstetrics and Gynecology, Xiangya Hospital, Central South University, Changsha, 410008, Hunan, China
| | - Chun Zhang
- Department of Obstetrics and Gynecology, Xiangya Hospital, Central South University, Changsha, 410008, Hunan, China
| | - Xiao-Qiong Zhang
- Department of Obstetrics and Gynecology, Xiangya Hospital, Central South University, Changsha, 410008, Hunan, China
| | - Yan-Ping Li
- Department of Reproductive Medicine, Xiangya Hospital, Central South University, Changsha, 410008, Hunan, China.
| |
Collapse
|
19
|
Zhou Y, Zhang CY, Duan JX, Li Q, Yang HH, Sun CC, Zhang J, Luo XQ, Liu SK. Vasoactive intestinal peptide suppresses the NLRP3 inflammasome activation in lipopolysaccharide-induced acute lung injury mice and macrophages. Biomed Pharmacother 2020; 121:109596. [DOI: 10.1016/j.biopha.2019.109596] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2019] [Revised: 10/17/2019] [Accepted: 10/26/2019] [Indexed: 02/07/2023] Open
|
20
|
Du MY, Duan JX, Zhang CY, Yang HH, Guan XX, Zhong WJ, Liu YZ, Li ZM, Cheng YR, Zhou Y, Guan CX. Psoralen attenuates bleomycin-induced pulmonary fibrosis in mice through inhibiting myofibroblast activation and collagen deposition. Cell Biol Int 2020; 44:98-107. [PMID: 31329322 DOI: 10.1002/cbin.11205] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2019] [Accepted: 07/16/2019] [Indexed: 01/24/2023]
Abstract
Idiopathic pulmonary fibrosis (IPF) is a progressive disease characterized by excessive deposition of extracellular matrix (ECM) and chronic inflammation with limited therapeutic options. Psoralen, a major active component extracted from Psoralea corylifolia L. seed, has several biological effects. However, the role of psoralen in IPF is still unclear. Here, we hypothesized that psoralen played an essential role in IPF in the inhibition of fibroblast proliferation and inflammatory response. A murine model of IPF was established by injecting bleomycin (BLM) intratracheally, and psoralen was administered for 14 days from the 7th to 21st day after BLM injection. Our results demonstrated that psoralen treatment reduced body weight loss and improved the survival rate of mice with IPF. Histological and immunofluorescent examination showed that psoralen alleviated BLM-induced lung parenchymal inflammatory and fibrotic alteration. Furthermore, psoralen inhibited proliferation and collagen synthesis of mouse fibroblasts and partially reversed BLM-induced expression of α-smooth muscle actin at both the tissue and cell level. Moreover, psoralen decreased the expression of transforming growth factor-β1, interleukin-1β, and tumor necrosis factor-α in the lungs of BLM-stimulated mice. Our results reveale for the first time that psoralen exerts therapeutic effects against IPF in a BLM-induced murine model.
Collapse
Affiliation(s)
- Ming-Yuan Du
- Department of Physiology, Xiangya School of Medicine, Central South University, Changsha, Hunan, 410078, China.,Department of Vascular Surgery, the Second Xiangya Hospital, Central South University, Changsha, Hunan, 410011, China.,Vascular Disease Institute, Central South University, Changsha, Hunan, 410011, China
| | - Jia-Xi Duan
- Department of Pulmonary and Critical Care Medicine, the Second Xiangya Hospital, Central South University, Changsha, Hunan, 410011, China
| | - Chen-Yu Zhang
- Department of Physiology, Xiangya School of Medicine, Central South University, Changsha, Hunan, 410078, China
| | - Hui-Hui Yang
- Department of Physiology, Xiangya School of Medicine, Central South University, Changsha, Hunan, 410078, China
| | - Xin-Xin Guan
- Department of Physiology, Xiangya School of Medicine, Central South University, Changsha, Hunan, 410078, China
| | - Wen-Jing Zhong
- Department of Physiology, Xiangya School of Medicine, Central South University, Changsha, Hunan, 410078, China
| | - Yan-Zhe Liu
- Department of Physiology, Xiangya School of Medicine, Central South University, Changsha, Hunan, 410078, China
| | - Zi-Ming Li
- Department of Physiology, Xiangya School of Medicine, Central South University, Changsha, Hunan, 410078, China
| | - Yu-Rui Cheng
- Department of Physiology, Xiangya School of Medicine, Central South University, Changsha, Hunan, 410078, China
| | - Yong Zhou
- Department of Physiology, Xiangya School of Medicine, Central South University, Changsha, Hunan, 410078, China
| | - Cha-Xiang Guan
- Department of Physiology, Xiangya School of Medicine, Central South University, Changsha, Hunan, 410078, China
| |
Collapse
|
21
|
Song Z, Li S, Zhang C, Yuan L, Han L, Liu Y. The therapeutic effect of verapamil in lipopolysaccharide-induced acute lung injury. Biochem Biophys Res Commun 2019; 517:648-654. [PMID: 31395340 DOI: 10.1016/j.bbrc.2019.07.090] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2019] [Accepted: 07/24/2019] [Indexed: 01/15/2023]
Abstract
The objective of this study was to investigate the exact therapeutic effects of Verapamil on lipopolysaccharide (LPS)-induced acute lung injury (ALI) and the molecular mechanism involved, through using LPS-induced animal models as well as LPS-stimulated mouse primary peritoneal macrophages models. Our results demonstrated that Verapamil reduced LPS-induced pathological damage of the lung tissue, infiltration of inflammatory cells and the production of IL-1β, TNF-α, and MCP-1 in the serum. The MPO activity, MDA content, lung wet/dry ratio and LDH activity were also attenuated by Verapamil. In addition, Verapamil attenuated LPS-induced inflammatory cytokine production and oxidative stress in primary murine peritoneal macrophages in vitro. Moreover, we confirmed that NF-κB/NLRP3 pathway was involved in the therapeutic effect of Verapamil against LPS-induced injury in vivo and in vitro. In conclusion, these findings indicate that Verapamil has a therapeutic effect on LPS-induced ALI in mice. The mechanism may be related to the inhibition of NF-κB and NLRP3 signaling pathways. Verapamil may be a potential therapeutic agent for the treatment of ALI.
Collapse
Affiliation(s)
- Zhuohui Song
- Department of Physiology, Changzhi Medical College, Changzhi, Shanxi, China
| | - Shufen Li
- Department of Physiology, Changzhi Medical College, Changzhi, Shanxi, China
| | - Cuiying Zhang
- Department of Physiology, Changzhi Medical College, Changzhi, Shanxi, China
| | - Li Yuan
- Department of Physiology, Changzhi Medical College, Changzhi, Shanxi, China
| | - Lingna Han
- Department of Physiology, Changzhi Medical College, Changzhi, Shanxi, China
| | - Yan Liu
- Department of Physiology, Changzhi Medical College, Changzhi, Shanxi, China.
| |
Collapse
|
22
|
Huang XT, Liu W, Zhou Y, Sun M, Sun CC, Zhang CY, Tang SY. Endoplasmic reticulum stress contributes to NMDA-induced pancreatic β-cell dysfunction in a CHOP-dependent manner. Life Sci 2019; 232:116612. [PMID: 31260687 DOI: 10.1016/j.lfs.2019.116612] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2019] [Revised: 06/17/2019] [Accepted: 06/27/2019] [Indexed: 01/09/2023]
Abstract
AIMS Accumulating evidence suggest that endoplasmic reticulum (ER) stress is an important mechanism underlying the development of diabetes. We have reported that sustained treatment with N-methyl-d-aspartate (NMDA) results in apoptotic β-cell death and impairs insulin secretion. However, the molecular mechanism responsible for NMDA-induced β-cell dysfunction remains largely obscure. Thus, this study aimed to determine whether sustained activation of NMDA receptors (NMDARs) causes β-cell dysfunction through ER stress. MAIN METHODS Primary mouse islets and MIN6 mouse pancreatic β-cells were treated with NMDA for 24 h or high-glucose for 72 h. After the treatment, glucose-stimulated insulin secretion (GSIS) and the expression of ER stress markers were measured, respectively. In vivo, the expression of ER stress markers was measured in the pancreas of diabetic mice treated with or without NMDARs inhibitor Memantine. KEY FINDINGS NMDA treatment caused an increase in the expression of ER stress markers (ATF4, CHOP, GRP78, and Xbp1s) in primary islets. While, tauroursodeoxycholic acid (TUDCA), an inhibitor of ER stress, significantly attenuated NMDA-induced β-cell dysfunction, including the loss of glucose-stimulated insulin secretion and reduction of pancreas duodenum homeobox factor-1 (Pdx-1) mRNA expression, a transcription factor regulating insulin synthesis. Besides, NMDA-induced ER stress strongly promoted pro-inflammatory cytokines synthesis (IL-1β and TNF-α) in β cells. Interestingly, knockdown of CHOP attenuated β-cell dysfunction evoked by NMDA. Furthermore, we demonstrated that blockade of NMDARs ameliorated high-glucose-induced ER stress in vitro and in vivo. SIGNIFICANCE This study confirms that ER stress is actively involved in the activation of NMDARs-related β-cell dysfunction.
Collapse
Affiliation(s)
- Xiao-Ting Huang
- Xiangya Nursing School, Central South University, Changsha, Hunan, China
| | - Wei Liu
- Xiangya Nursing School, Central South University, Changsha, Hunan, China
| | - Yong Zhou
- Department of Physiology, Xiangya School of Medicine, Central South University, Changsha, Hunan, China
| | - Mei Sun
- Xiangya Nursing School, Central South University, Changsha, Hunan, China
| | - Chen-Chen Sun
- Department of Physiology, Xiangya School of Medicine, Central South University, Changsha, Hunan, China
| | - Chen-Yu Zhang
- Department of Physiology, Xiangya School of Medicine, Central South University, Changsha, Hunan, China
| | - Si-Yuan Tang
- Xiangya Nursing School, Central South University, Changsha, Hunan, China.
| |
Collapse
|
23
|
Wang C, Liu Z, Zhang P, Ma X, Che K, Wang Y. The differences in homeostasis model assessment values in type 2 diabetic patients with different lengths of history of diabetes. ARCHIVES OF ENDOCRINOLOGY AND METABOLISM 2019; 63:222-227. [PMID: 31066759 PMCID: PMC10522207 DOI: 10.20945/2359-3997000000134] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/06/2018] [Accepted: 03/17/2019] [Indexed: 11/23/2022]
Abstract
OBJECTIVE Type 2 diabetes (T2DM) is characterized by the progressive deterioration of pancreatic islet β-cell function over time and insulin resistance. Knowing more about the differences in pancreatic islet function in T2DM patients who have had diabetes for different lengths of time can help improve therapy for T2DM. SUBJECTS AND METHODS We conducted a cross-sectional study to compare islet β-cell function and insulin resistance in T2DM patients (n = 3,254) who had had diabetes for different lengths of time and those in normal controls (n = 794) using ANOVA and LSD analysis. RESULTS We found that compared with that in normal controls, HOMA-β in T2DM patients with a history of diabetes of less than 1 year was lower (approximately 52% of that of normal controls, p = 0.003), while HOMA-IR in these patients was higher (approximately 50% of that of normal controls, p = 0.007). Compared with that in other diabetic patients, HOMA-β in patients with a history of diabetes of more than 30 years was the lowest. HOMA-IR in patients with a history of diabetes of between 20 and 30 years was lower than that in other diabetic patients (p < 0.05). CONCLUSIONS There were obvious decreases in HOMA-β and increases in HOMA-IR in T2DM patients with a history of diabetes of less than 1 year compared with those in normal controls. Therefore, early screening and intervention for T2DM might help improve islet function and delay the progression of diabetes.
Collapse
Affiliation(s)
- Chen Wang
- Endocrinology DepartmentThe Affiliated Hospital of Qingdao UniversityQingdaoShandongChinaEndocrinology Department, The Affiliated Hospital of Qingdao University, Qingdao, Shandong, China
| | - Zaibo Liu
- Department of General SurgeryPeople’s Hospital of HaiyangYantaiShandongChinaDepartment of General Surgery, People’s Hospital of Haiyang, Yantai, Shandong, China
| | - Peng Zhang
- Department of Gastroenterology,The Affiliated Hospital of Qingdao UniversityQingdaoShandongChinaDepartment of Gastroenterology, The Affiliated Hospital of Qingdao University, Qingdao, Shandong, China
| | - Xiaolong Ma
- Endocrinology DepartmentThe Affiliated Hospital of Qingdao UniversityQingdaoShandongChinaEndocrinology Department, The Affiliated Hospital of Qingdao University, Qingdao, Shandong, China
| | - Kui Che
- Laboratory of Thyroid DiseaseThe Affiliated Hospital of Qingdao UniversityQingdaoShandongChinaLaboratory of Thyroid Disease, The Affiliated Hospital of Qingdao University, Qingdao, Shandong, China
| | - Yangang Wang
- Endocrinology DepartmentThe Affiliated Hospital of Qingdao UniversityQingdaoShandongChinaEndocrinology Department, The Affiliated Hospital of Qingdao University, Qingdao, Shandong, China
| |
Collapse
|
24
|
TAKAHASHI H, YOKOI N, SEINO S. Glutamate as intracellular and extracellular signals in pancreatic islet functions. PROCEEDINGS OF THE JAPAN ACADEMY. SERIES B, PHYSICAL AND BIOLOGICAL SCIENCES 2019; 95:246-260. [PMID: 31189778 PMCID: PMC6751295 DOI: 10.2183/pjab.95.017] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/15/2019] [Accepted: 03/08/2019] [Indexed: 05/25/2023]
Abstract
l-Glutamate is one of the most abundant amino acids in the body and is a constituent of proteins and a substrate in metabolism. It is well known that glutamate serves as a primary excitatory neurotransmitter and a critical neuromodulator in the brain. Recent studies have shown that in addition to its pivotal role in neural functions, glutamate plays many important roles in a variety of cellular functions, including those as intracellular and extracellular signals. In pancreatic islets, glutamate is now known to be required for the normal regulation of insulin secretion, such as incretin-induced insulin secretion. In this review, we primarily discuss the physiological and pathophysiological roles of glutamate as intracellular and extracellular signals in the functions of pancreatic islets.
Collapse
Affiliation(s)
- Harumi TAKAHASHI
- Division of Molecular and Metabolic Medicine, Department of Physiology and Cell Biology, Kobe University Graduate School of Medicine, Kobe, Hyogo, Japan
| | - Norihide YOKOI
- Division of Molecular and Metabolic Medicine, Department of Physiology and Cell Biology, Kobe University Graduate School of Medicine, Kobe, Hyogo, Japan
| | - Susumu SEINO
- Division of Molecular and Metabolic Medicine, Department of Physiology and Cell Biology, Kobe University Graduate School of Medicine, Kobe, Hyogo, Japan
| |
Collapse
|
25
|
Dadvar S, Ferreira DMS, Cervenka I, Ruas JL. The weight of nutrients: kynurenine metabolites in obesity and exercise. J Intern Med 2018; 284:519-533. [PMID: 30141532 DOI: 10.1111/joim.12830] [Citation(s) in RCA: 40] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Obesity ultimately results from an imbalance between energy intake and expenditure. However, in addition to their bioenergetic value, nutrients and their metabolites can function as important signalling molecules in energy homeostasis. Indeed, macronutrients and their metabolites can be direct regulators of metabolism through their actions on different organs. In turn, target organs can decide to use, store or transform the incoming nutrients depending on their physiological context and in coordination with other cell types. Tryptophan-kynurenine metabolites are an example of a family of compounds that can serve as systemic integrators of energy metabolism by signalling to different cell types. These include adipocytes, immune cells and muscle fibres, in addition to the well-known effects of kynurenine metabolites on the central nervous system. In the context of energy metabolism, several of the effects elicited by kynurenic acid are mediated by the G-protein-coupled receptor, GPR35. As GPR35 is expressed in tissues such as the adipose tissue, immune cells and the gastrointestinal tract, this receptor could be a potential therapeutic target for the treatment of obesity, diabetes and other metabolic diseases. In addition, metabolic disorders often coincide with states of chronic inflammation, which further highlights GPR35 as an integration node in conditions where inflammation skews metabolism. Defining the molecular interplay between different tissues in the regulation of energy homeostasis can help us understand interindividual variability in the response to nutrient intake and develop safe and efficient therapies to fight obesity and metabolic disease.
Collapse
Affiliation(s)
- S Dadvar
- Department of Physiology and Pharmacology, Molecular & Cellular Exercise Physiology, Karolinska Institutet, Biomedicum, Stockholm, Sweden
| | - D M S Ferreira
- Department of Physiology and Pharmacology, Molecular & Cellular Exercise Physiology, Karolinska Institutet, Biomedicum, Stockholm, Sweden
| | - I Cervenka
- Department of Physiology and Pharmacology, Molecular & Cellular Exercise Physiology, Karolinska Institutet, Biomedicum, Stockholm, Sweden
| | - J L Ruas
- Department of Physiology and Pharmacology, Molecular & Cellular Exercise Physiology, Karolinska Institutet, Biomedicum, Stockholm, Sweden
| |
Collapse
|
26
|
Sun GY, Yang HH, Guan XX, Zhong WJ, Liu YP, Du MY, Luo XQ, Zhou Y, Guan CX. Vasoactive intestinal peptide overexpression mediated by lentivirus attenuates lipopolysaccharide-induced acute lung injury in mice by inhibiting inflammation. Mol Immunol 2018; 97:8-15. [PMID: 29544087 DOI: 10.1016/j.molimm.2018.03.002] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2017] [Revised: 02/01/2018] [Accepted: 03/05/2018] [Indexed: 01/17/2023]
Abstract
Vasoactive intestinal peptide (VIP) is one of the most abundant neuropeptides in the lungs with various biological characters. We have reported that VIP inhibited the expressions of TREM-1 and IL-17A, which are involved in the initiation and amplification of inflammation in acute lung injury (ALI). However, the overall effect of VIP on ALI remains unknown. The aim of this study is to investigate the therapeutic effect of VIP mediated by lentivirus (Lenti-VIP) on lipopolysaccharide (LPS)-induced murine ALI. We found that the expression of intrapulmonary VIP peaked at day7 after the intratracheal injection of Lenti-VIP. Lenti-VIP increased the respiratory rate, lung compliance, and tidal volume, while decreased airway resistance in ALI mice, detected by Buxco system. Lenti-VIP significantly reduced inflammatory cell infiltration and maintained the integrity of the alveolar septa. Lenti-VIP also remarkably decreased the total protein level, the number of neutrophil and lactate dehydrogenase activity in the bronchoalveolar lavage fluid of LPS-induced ALI mice. In addition, Lenti-VIP down-regulated pro-inflammatory tumor necrosis factor (TNF)-α mRNA and protein expression, while up-regulated anti-inflammatory interleukin-10 mRNA and protein expression in lungs of ALI mice. Furthermore, we observed that VIP reduced the TNF-α expression in murine macrophages under LPS stimulation through protein kinase C and protein kinase A pathways. Together, our findings show that in vivo administration of lentivirus expressing VIP exerts a potent therapeutic effect on LPS-induced ALI in mice via inhibiting inflammation.
Collapse
Affiliation(s)
- Guo-Ying Sun
- Department of Physiology, Xiangya School of Medicine, Central South University, Changsha, Hunan 410078, China; School of Medicine, Hunan Normal University, Changsha, Hunan 410013, China
| | - Hui-Hui Yang
- Department of Physiology, Xiangya School of Medicine, Central South University, Changsha, Hunan 410078, China
| | - Xin-Xin Guan
- Department of Physiology, Xiangya School of Medicine, Central South University, Changsha, Hunan 410078, China
| | - Wen-Jing Zhong
- Department of Physiology, Xiangya School of Medicine, Central South University, Changsha, Hunan 410078, China
| | - Yong-Ping Liu
- Department of Physiology, Xiangya School of Medicine, Central South University, Changsha, Hunan 410078, China
| | - Ming-Yuan Du
- Department of Physiology, Xiangya School of Medicine, Central South University, Changsha, Hunan 410078, China
| | - Xiao-Qin Luo
- Department of Physiology, Xiangya School of Medicine, Central South University, Changsha, Hunan 410078, China
| | - Yong Zhou
- Department of Physiology, Xiangya School of Medicine, Central South University, Changsha, Hunan 410078, China.
| | - Cha-Xiang Guan
- Department of Physiology, Xiangya School of Medicine, Central South University, Changsha, Hunan 410078, China.
| |
Collapse
|
27
|
Nunemaker CS, Li C. Can NMDA Receptors Get β-Cells Toxically Excited? Endocrinology 2017; 158:3709-3710. [PMID: 29099959 PMCID: PMC5695841 DOI: 10.1210/en.2017-00832] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/12/2017] [Accepted: 09/18/2017] [Indexed: 11/19/2022]
Affiliation(s)
- Craig S. Nunemaker
- Diabetes Institute, Heritage College of Osteopathic
Medicine, Ohio University, Athens, Ohio 45701
- Department of Biomedical Sciences, Heritage College of
Osteopathic Medicine, Ohio University, Athens, Ohio 45701
| | - Chien Li
- Obesity Research, Novo Nordisk Research Center, Seattle,
Washington 98109
| |
Collapse
|