1
|
Romero JC, Tonapi SS, Parihar M, Loranc E, Miller HE, Lawrence LA, Bassani N, Robledo DG, Cao L, Nie J, Kanda K, Stoja A, Garcia N, Gorthi A, Stoveken BJ, Fan TWM, Cassel TA, Zha S, Lechleiter JD, Musi N, Dong LQ, Lane AN, Bishop AJR. Loss of CD98HC phosphorylation by ATM impairs antiporter trafficking and drives glutamate toxicity in Ataxia telangiectasia. Nat Commun 2025; 16:5109. [PMID: 40456742 PMCID: PMC12130354 DOI: 10.1038/s41467-025-60304-4] [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: 08/26/2024] [Accepted: 05/14/2025] [Indexed: 06/11/2025] Open
Abstract
Ataxia-telangiectasia is a rare genetic disorder characterized by neurological defects, immunodeficiency, cancer predisposition, radiosensitivity, decreased blood vessel integrity, and diabetes. ATM, the protein mutated in Ataxia-telangiectasia, responds to DNA damage and oxidative stress, but its functional relationship to the progressive clinical manifestation of this disorder is not understood. CD98HC chaperones cystine/glutamate and cationic/neutral amino acid antiporters to the cell membrane, and CD98HC phosphorylation by ATM accelerates membrane localization to acutely increase amino acid transport. Loss of ATM impacts tissues reliant on heterodimeric amino acid transporters relevant to Ataxia-telangiectasia phenotypes, such as endothelial cells (telangiectasia) and pancreatic α-cells (fatty liver and diabetes), with toxic glutamate accumulation. Bypassing the antiporters restores intracellular metabolic balance in ATM-deficient cells and mouse models. These findings provide insight into the long-known benefits of N-acetyl cysteine in Ataxia-telangiectasia cells beyond oxidative stress through removing glutamate excess by producing glutathione.
Collapse
Affiliation(s)
- July Carolina Romero
- Cell Systems and Anatomy, UT Health San Antonio, San Antonio, TX, USA
- Greehey Children's Cancer Research Institute, UT Health San Antonio, San Antonio, TX, USA
| | - Sonal S Tonapi
- Cell Systems and Anatomy, UT Health San Antonio, San Antonio, TX, USA
- Greehey Children's Cancer Research Institute, UT Health San Antonio, San Antonio, TX, USA
| | - Manish Parihar
- Cell Systems and Anatomy, UT Health San Antonio, San Antonio, TX, USA
- Greehey Children's Cancer Research Institute, UT Health San Antonio, San Antonio, TX, USA
| | - Eva Loranc
- Greehey Children's Cancer Research Institute, UT Health San Antonio, San Antonio, TX, USA
| | - Henry E Miller
- Greehey Children's Cancer Research Institute, UT Health San Antonio, San Antonio, TX, USA
| | - Liesl A Lawrence
- Cell Systems and Anatomy, UT Health San Antonio, San Antonio, TX, USA
- Greehey Children's Cancer Research Institute, UT Health San Antonio, San Antonio, TX, USA
| | - Nicklas Bassani
- Cell Systems and Anatomy, UT Health San Antonio, San Antonio, TX, USA
- Greehey Children's Cancer Research Institute, UT Health San Antonio, San Antonio, TX, USA
| | - Daniel G Robledo
- Greehey Children's Cancer Research Institute, UT Health San Antonio, San Antonio, TX, USA
| | - Lin Cao
- Greehey Children's Cancer Research Institute, UT Health San Antonio, San Antonio, TX, USA
| | - Jia Nie
- Barshop Institute for Longevity and Aging Studies, Health San Antonio, San Antonio, TX, USA
- Medicine, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - Kairi Kanda
- Cell Systems and Anatomy, UT Health San Antonio, San Antonio, TX, USA
- Greehey Children's Cancer Research Institute, UT Health San Antonio, San Antonio, TX, USA
| | - Aiola Stoja
- Cell Systems and Anatomy, UT Health San Antonio, San Antonio, TX, USA
- Greehey Children's Cancer Research Institute, UT Health San Antonio, San Antonio, TX, USA
| | - Natalia Garcia
- Cell Systems and Anatomy, UT Health San Antonio, San Antonio, TX, USA
- Greehey Children's Cancer Research Institute, UT Health San Antonio, San Antonio, TX, USA
| | - Aparna Gorthi
- Cell Systems and Anatomy, UT Health San Antonio, San Antonio, TX, USA
- Greehey Children's Cancer Research Institute, UT Health San Antonio, San Antonio, TX, USA
| | - Brian J Stoveken
- Cell Systems and Anatomy, UT Health San Antonio, San Antonio, TX, USA
| | - Teresa W-M Fan
- Toxicology and Cancer Biology, Markey Cancer Center, University of Kentucky, Lexington, KY, USA
| | - Teresa A Cassel
- Toxicology and Cancer Biology, Markey Cancer Center, University of Kentucky, Lexington, KY, USA
| | - Shan Zha
- Institute for Cancer Genetics, Vagelos College of Physicians and Surgeons, Columbia University, New York, NY, USA
| | - James D Lechleiter
- Cell Systems and Anatomy, UT Health San Antonio, San Antonio, TX, USA
- Center for Precision Medicine, UT Health San Antonio, San Antonio, TX, USA
| | - Nicolas Musi
- Barshop Institute for Longevity and Aging Studies, Health San Antonio, San Antonio, TX, USA
- Medicine, Cedars-Sinai Medical Center, Los Angeles, CA, USA
- Mays Cancer Center, UT Health San Antonio, San Antonio, TX, USA
| | - Lily Q Dong
- Cell Systems and Anatomy, UT Health San Antonio, San Antonio, TX, USA
| | - Andrew N Lane
- Toxicology and Cancer Biology, Markey Cancer Center, University of Kentucky, Lexington, KY, USA
| | - Alexander J R Bishop
- Cell Systems and Anatomy, UT Health San Antonio, San Antonio, TX, USA.
- Greehey Children's Cancer Research Institute, UT Health San Antonio, San Antonio, TX, USA.
- Barshop Institute for Longevity and Aging Studies, Health San Antonio, San Antonio, TX, USA.
- Mays Cancer Center, UT Health San Antonio, San Antonio, TX, USA.
- Nationwide Children's Hospital, Center for Childhood Cancer, Columbus, OH, USA.
- Department of Pediatrics, The Ohio State University College of Medicine, Columbus, OH, USA.
| |
Collapse
|
2
|
Zhang YY, Zhu DX, Wang MY, Yi YT, Feng YH, Zhou C, Li CJ, Liu F, Shen JF. Activation of NR2A-Wnt-TLR2 Signaling Axis in Satellite Glial Cells of the Dorsal Root Ganglion Contributes to Neuropathic Pain Induced by Nerve Injury in Diabetic Mice. Mol Neurobiol 2025; 62:8013-8037. [PMID: 39964585 DOI: 10.1007/s12035-025-04754-3] [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: 11/21/2024] [Accepted: 02/06/2025] [Indexed: 03/17/2025]
Abstract
Diabetic peripheral neuropathic pain (DPNP), a common diabetic mellitus (DM) complication, may result from the activation of satellite glial cells (SGCs) in the dorsal root ganglion (DRG), potentially enhancing peripheral sensitization. The N-methyl-D-aspartate receptor (NMDAR) subtype NR2A and Toll-like receptor (TLR)2 play key roles in neuroimmune interactions. However, their roles in SGCs of DRG and the precise mechanisms mediating peripheral sensitization in DPNP remain unclear. Here, we found that the expression of glial fibrillary acidic protein (GFAP), NR2A, and TLR2 in SGCs from DRG significantly increased under increased glucose and NMDA stimulation in vitro. Additionally, upregulation of interleukin (IL)-6 and nerve growth factor (NGF) was observed. Notably, lentivirus-induced NR2A knockdown (KD) and C29 (TLR2 inhibitor) significantly blocked the above SGCs changes induced by NMDA and increased glucose. Behavior tests showed mechanical and thermal sensitivities induced by sciatic nerve ligation (SNL) were more obvious in DM background related to streptozotocin (STZ) injection than non-DM background mice, which were significantly alleviated by NR2A conditional knockout (CKO) in SGCs and TLR2 KO. Moreover, immunofluorescence (IF) results revealed the co-expression of NR2A and TLR2 in neurons and SGCs in the DRG. Following SNL in DM mice, the upregulation of NR2A, TLR2, GFAP, β-catenin, p-GSK-3β, p-nuclear factor kappa (NF-κ)-B, IL-6, NGF, Bcl-2-associated X protein (Bax), and Caspase 3, and the significant downregulation of Bcl-2 were consistent with the changes observed after increased glucose and NMDA treatment. The upregulation of TLR2 was blocked by NR2A CKO and Wnt signal pathway inhibition. Additionally, the activation of SGCs, upregulated IL-6 as well as NGF secretion and increased apoptosis, associated with nerve injury in DM background were altered by TLR2 KO and NF-κB pathway inhibition. In conclusion, the activation of the NR2A-Wnt-TLR2 signaling axis mediated peripheral sensitization in the DRG by influencing SGCs' activation, and the synthesis and secretion of pro-inflammatory cytokines and NGF, promoting SGCs' apoptosis, thus exacerbating a peripheral nerve injury related-NP in DM background. Our study provided insights into the role of NR2A-Wnt-TLR2 signaling axis of SGCs in mediating the generation and maintenance of DPNP and suggested targeting this signaling axis may be a promising therapeutic approach for DPNP.
Collapse
Affiliation(s)
- Yan-Yan Zhang
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, National Center for Stomatology, West China School of Stomatology, Sichuan University, No. 14, Section 3, Renminnan Road, Chengdu, 610041, China
- Department of Prosthodontics, West China Hospital of Stomatology, Sichuan University, Chengdu, 610041, China
| | - De-Xin Zhu
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, National Center for Stomatology, West China School of Stomatology, Sichuan University, No. 14, Section 3, Renminnan Road, Chengdu, 610041, China
- Department of Prosthodontics, West China Hospital of Stomatology, Sichuan University, Chengdu, 610041, China
| | - Mu-Yun Wang
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, National Center for Stomatology, West China School of Stomatology, Sichuan University, No. 14, Section 3, Renminnan Road, Chengdu, 610041, China
- Department of Prosthodontics, West China Hospital of Stomatology, Sichuan University, Chengdu, 610041, China
| | - Ya-Ting Yi
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, National Center for Stomatology, West China School of Stomatology, Sichuan University, No. 14, Section 3, Renminnan Road, Chengdu, 610041, China
- Department of Orthodontics, West China Hospital of Stomatology, Sichuan University, Chengdu, 610041, China
| | - Yu-Heng Feng
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, National Center for Stomatology, West China School of Stomatology, Sichuan University, No. 14, Section 3, Renminnan Road, Chengdu, 610041, China
- Department of Prosthodontics, West China Hospital of Stomatology, Sichuan University, Chengdu, 610041, China
| | - Cheng Zhou
- Laboratory of Anesthesia and Critical Care Medicine, Translational Neuroscience Center, West China Hospital of Sichuan University, Chengdu, China
| | - Chun-Jie Li
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, National Center for Stomatology, West China School of Stomatology, Sichuan University, No. 14, Section 3, Renminnan Road, Chengdu, 610041, China
- Department of Head and Neck Oncology, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Fei Liu
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, National Center for Stomatology, West China School of Stomatology, Sichuan University, No. 14, Section 3, Renminnan Road, Chengdu, 610041, China
- Department of Prosthodontics, West China Hospital of Stomatology, Sichuan University, Chengdu, 610041, China
| | - Jie-Fei Shen
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, National Center for Stomatology, West China School of Stomatology, Sichuan University, No. 14, Section 3, Renminnan Road, Chengdu, 610041, China.
- Department of Prosthodontics, West China Hospital of Stomatology, Sichuan University, Chengdu, 610041, China.
| |
Collapse
|
3
|
Mohyeldin RH, Sharata EE, Fawzy MA, Attya ME, Welson NN, Rofaeil RR. Memantine abrogates testicular dysfunction induced by risperidone in rats with a potential role of ERK1/2-Nrf2-caspase-3 signaling pathway. Sci Rep 2025; 15:12914. [PMID: 40234489 PMCID: PMC12000432 DOI: 10.1038/s41598-025-94760-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2024] [Accepted: 03/17/2025] [Indexed: 04/17/2025] Open
Abstract
Psychosis is usually a substantial global burden with a prevalence of 0.4-2%. On the other hand, 50 million people are suffering from dementia, with dementia-related psychosis affecting approximately 25% of them. The current experiment aimed to investigate the effect of the anti-dementia drug memantine (MEM) on testicular damage and insulin resistance induced by the chronic administration of risperidone (RIS) in rats. Six groups of Wistar albino rats were designated as follows: control, MEM-5 (rats received MEM at 5 mg/kg/day, orally, for 4 weeks), MEM-10 (rats received MEM at 10 mg/kg/day, orally, for 4 weeks), RIS (rats were administered RIS at 2.5 mg/kg/day, orally, for 4 weeks), RIS + MEM-5 (rats received MEM at 5 mg/kg/day, orally, co-administered with RIS as in the RIS group for 4 weeks), and RIS + MEM-10 (rats received MEM at 10 mg/kg/day, orally, co-administered with RIS as in the RIS group for 4 weeks). The duration of the study was 28 days. Serum testosterone, resistin, and adiponectin concentrations were determined. The homeostatic model assessment of insulin resistance (HOMA-IR) was also evaluated. Oxidative stress, inflammatory markers, and immunoblotting of ERK1/2, and Nrf2 were quantified in testicular tissue together with histopathological evaluation and a caspase-3 immunohistochemical study. MEM co-administration increased adiponectin, serum testosterone, GSH, SOD, CAT, and Nrf2 expression while decreasing HOMA-IR, resistin, MDA, NOx, ERK1/2, IL-6, TNF-α, NFĸB, and caspase-3 expression. Furthermore, MEM ameliorated all measured parameters and histopathological changes that occurred in the RIS group in a dose-dependent manner. The primary outcomes were attained by attenuating oxidative stress, inflammation, and apoptosis in the testis caused by chronic RIS administration via regulation of the ERK1/2-Nrf2 signaling pathway. Targeting the ERK1/2-Nrf2 pathway is a potential strategy for addressing testicular injury.
Collapse
Affiliation(s)
- Reham H Mohyeldin
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Deraya University, Minia, 61111, Egypt
| | - Ehab E Sharata
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Deraya University, Minia, 61111, Egypt
| | - Michael Atef Fawzy
- Department of Biochemistry, Faculty of Pharmacy, Minia University, Minia, 61511, Egypt
| | - Mina Ezzat Attya
- Department of Pathology, Faculty of Medicine, Minia University, Minia, 61519, Egypt
| | - Nermeen N Welson
- Department of Forensic Medicine and Clinical Toxicology, Faculty of Medicine, Beni-Suef University, Beni Suef, 62514, Egypt
| | - Remon Roshdy Rofaeil
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Deraya University, Minia, 61111, Egypt.
- Department of Medical Pharmacology, Faculty of Medicine, Minia University, Minia, 61511, Egypt.
| |
Collapse
|
4
|
Ježek P. Physiological Fatty Acid-Stimulated Insulin Secretion and Redox Signaling Versus Lipotoxicity. Antioxid Redox Signal 2025; 42:566-622. [PMID: 39834189 DOI: 10.1089/ars.2024.0799] [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] [Indexed: 01/22/2025]
Abstract
Significance: Type 2 diabetes as a world-wide epidemic is characterized by the insulin resistance concomitant to a gradual impairment of β-cell mass and function (prominently declining insulin secretion) with dysregulated fatty acids (FAs) and lipids, all involved in multiple pathological development. Recent Advances: Recently, redox signaling was recognized to be essential for insulin secretion stimulated with glucose (GSIS), branched-chain keto-acids, and FAs. FA-stimulated insulin secretion (FASIS) is a normal physiological event upon postprandial incoming chylomicrons. This contrasts with the frequent lipotoxicity observed in rodents. Critical Issues: Overfeeding causes FASIS to overlap with GSIS providing repeating hyperinsulinemia, initiates prediabetic states by lipotoxic effects and low-grade inflammation. In contrast the protective effects of lipid droplets in human β-cells counteract excessive lipids. Insulin by FASIS allows FATP1 recruitment into adipocyte plasma membranes when postprandial chylomicrons come late at already low glycemia. Future Directions: Impaired states of pancreatic β-cells and peripheral organs at prediabetes and type 2 diabetes should be revealed, including the inter-organ crosstalk by extracellular vesicles. Details of FA/lipid molecular physiology are yet to be uncovered, such as complex phenomena of FA uptake into cells, postabsorptive inactivity of G-protein-coupled receptor 40, carnitine carrier substrate specificity, the role of carnitine-O-acetyltransferase in β-cells, and lipid droplet interactions with mitochondria. Antioxid. Redox Signal. 42, 566-622.
Collapse
Affiliation(s)
- Petr Ježek
- Department of Mitochondrial Physiology, No.75, Institute of Physiology of the Czech Academy of Sciences, Prague, Czech Republic
| |
Collapse
|
5
|
Chen L, Wang C, Qin L, Zhang H. Parkinson's disease and glucose metabolism impairment. Transl Neurodegener 2025; 14:10. [PMID: 39962629 PMCID: PMC11831814 DOI: 10.1186/s40035-025-00467-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2024] [Accepted: 01/02/2025] [Indexed: 02/21/2025] Open
Abstract
Parkinson's disease (PD) is the second most common neurodegenerative disorder. PD patients exhibit varying degrees of abnormal glucose metabolism throughout disease stages. Abnormal glucose metabolism is closely linked to the PD pathogenesis and progression. Key glucose metabolism processes involved in PD include glucose transport, glycolysis, the tricarboxylic acid cycle, oxidative phosphorylation, the pentose phosphate pathway, and gluconeogenesis. Recent studies suggest that glucose metabolism is a potential therapeutic target for PD. In this review, we explore the connection between PD and abnormal glucose metabolism, focusing on the underlying pathophysiological mechanisms. We also summarize potential therapeutic drugs related to glucose metabolism based on results from current cellular and animal model studies.
Collapse
Affiliation(s)
- Liangjing Chen
- Department of Neurology, Second Xiangya Hospital, Central South University, Changsha, 410011, China
- Clinical Medical Research Center for Stroke Prevention and Treatment of Hunan Province, Department of Neurology, Second Xiangya Hospital, Central South University, Changsha, 410011, China
| | - Chunyu Wang
- Department of Neurology, Second Xiangya Hospital, Central South University, Changsha, 410011, China
- Clinical Medical Research Center for Stroke Prevention and Treatment of Hunan Province, Department of Neurology, Second Xiangya Hospital, Central South University, Changsha, 410011, China
| | - Lixia Qin
- Department of Neurology, Second Xiangya Hospital, Central South University, Changsha, 410011, China.
- Clinical Medical Research Center for Stroke Prevention and Treatment of Hunan Province, Department of Neurology, Second Xiangya Hospital, Central South University, Changsha, 410011, China.
| | - Hainan Zhang
- Department of Neurology, Second Xiangya Hospital, Central South University, Changsha, 410011, China.
- Clinical Medical Research Center for Stroke Prevention and Treatment of Hunan Province, Department of Neurology, Second Xiangya Hospital, Central South University, Changsha, 410011, China.
| |
Collapse
|
6
|
Dumont KD, Jannig PR, Porsmyr-Palmertz M, Ruas JL. Constitutive loss of kynurenine-3-monooxygenase changes circulating kynurenine metabolites without affecting systemic energy metabolism. Am J Physiol Endocrinol Metab 2025; 328:E274-E285. [PMID: 39805032 DOI: 10.1152/ajpendo.00386.2024] [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: 10/02/2024] [Revised: 11/22/2024] [Accepted: 12/09/2024] [Indexed: 01/16/2025]
Abstract
Kynurenic acid (KYNA) and quinolinic acid (QUIN) are metabolites of the kynurenine pathway of tryptophan degradation with opposing biological activities in the central nervous system. In the periphery, KYNA is known to positively affect metabolic health, whereas the effects of QUIN remain less explored. Interestingly, metabolic stressors, including exercise and obesity, differentially change the balance between circulating KYNA and QUIN. Here, we hypothesized that chronically elevated levels of circulating KYNA and reduced levels of QUIN would manifest as differences in whole body energy metabolism. To test this, we used a mouse model lacking the enzyme kynurenine 3-monooxygenase (KMO), thus shunting kynurenine away from QUIN synthesis and toward KYNA production. KMO-deficient and wild-type littermate male and female mice were evaluated under chow and high-fat diets. Comprehensive kynurenine pathway metabolite profiling in plasma showed that the loss of KMO elicits robust changes in circulating levels of kynurenine metabolites. This included a 45-fold increase in kynurenine, a 26-fold increase in KYNA, and a 99% decrease in QUIN levels, depending on the diet. However, despite these changes, loss of KMO did not significantly impact whole body energy metabolism or change the transcriptomic profile of subcutaneous adipose tissue on either diet. With KMO inhibitors being considered therapeutic candidates for various disorders, this work shows that chronic systemic KMO inhibition does not have widespread metabolic effects. Our data also indicate that the beneficial effects of KYNA on metabolism may depend on its acute, intermittent elevation in circulation, akin to transient exercise-induced signals that mediate improved metabolic health.NEW & NOTEWORTHY The kynurenine pathway of tryptophan degradation is influenced by metabolic stressors: exercise raises circulating KYNA levels, while obesity is linked to increased QUIN. We investigated whether a mouse model lacking KMO-leading to increased circulating KYNA and decreased QUIN-would exhibit changes in energy metabolism. We found that energy metabolism was largely unaffected despite robust changes in circulating kynurenine metabolites, suggesting that systemic KMO inhibition may not have widespread metabolic effects.
Collapse
Affiliation(s)
- Kyle D Dumont
- Molecular and Cellular Exercise Physiology, Department of Physiology and Pharmacology, Karolinska Institutet, Stockholm, Sweden
| | - Paulo R Jannig
- Molecular and Cellular Exercise Physiology, Department of Physiology and Pharmacology, Karolinska Institutet, Stockholm, Sweden
| | - Margareta Porsmyr-Palmertz
- Molecular and Cellular Exercise Physiology, Department of Physiology and Pharmacology, Karolinska Institutet, Stockholm, Sweden
| | - Jorge L Ruas
- Molecular and Cellular Exercise Physiology, Department of Physiology and Pharmacology, Karolinska Institutet, Stockholm, Sweden
- Department of Pharmacology and Stanley and Judith Frankel Institute for Heart & Brain Health, University of Michigan Medical School, Ann Arbor, Michigan, United States
| |
Collapse
|
7
|
Kuo H, Lin C, Tsai S, Chen C, Lyu R, Chu C, Ro L, Liao M, Chang H, Weng Y, Hwang J. Blood metabolomic profile in patients with type 2 diabetes mellitus with diabetic peripheral neuropathic pain. J Diabetes Investig 2025; 16:246-256. [PMID: 39548809 PMCID: PMC11786186 DOI: 10.1111/jdi.14355] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/11/2024] [Revised: 10/17/2024] [Accepted: 11/01/2024] [Indexed: 11/18/2024] Open
Abstract
AIMS This study aimed to identify metabolic markers for diabetic peripheral neuropathic pain (DPNP) in patients with type 2 diabetes mellitus (T2DM). MATERIALS AND METHODS Blood metabolite levels in the amino acid, biogenic amine, sphingomyelin, phosphatidylcholine (PC), carnitines, and hexose classes were analyzed in nondiabetic control (n = 27), T2DM without DPNP (n = 58), and T2DM with DPNP (n = 29) using liquid chromatography tandem mass spectrometry. Variable importance projection (VIP) evaluation by partial least squares discriminant analysis was performed on clinical parameters and metabolites. RESULTS Sixteen variables with VIP > 1.0 (P < 0.05) were identified across all patient groups, and 5 variables were identified to discriminate between the two T2DM groups. DPNP patients showed elevated fasting blood glucose, glutamate, PC aa C36:1, lysoPC a C18:1, and lysoPC a C18:2, while low-density lipoprotein cholesterol, phenylalanine, and tryptophan were reduced. Glutamate, lysoPC a C18:1, and lysoPC a C18:2 discriminated T2DM with DPNP from those without DPNP with an AUC of 0.671. The AUC was improved to 0.765 when ratios of metabolite pairs were considered. INTERPRETATION Blood metabolites include glutamate, and phospholipid-related metabolites implicated in neuropathic pain may have the potential as biomarkers for DPNP. Further investigation is required to understand the mechanism of action of these altered metabolites in DPNP.
Collapse
Affiliation(s)
- Hung‐Chou Kuo
- Department of Neurology, Linkou Medical CenterChang Gung Memorial HospitalTaoyuan CityTaiwan
- College of MedicineChang Gung UniversityTaoyuan CityTaiwan
| | - Chia‐Ni Lin
- Department of Laboratory MedicineChang Gung Memorial HospitalTaoyuan CityTaiwan
- Department of Medical Biotechnology and Laboratory Science, College of MedicineChang Gung UniversityTaoyuan CityTaiwan
| | - Sung‐Sheng Tsai
- College of MedicineChang Gung UniversityTaoyuan CityTaiwan
- Division of Endocrinology and Metabolism, Department of Internal Medicine, Linkou Medical CenterChang Gung Memorial HospitalTaoyuan CityTaiwan
| | - Chiung‐Mei Chen
- Department of Neurology, Linkou Medical CenterChang Gung Memorial HospitalTaoyuan CityTaiwan
- College of MedicineChang Gung UniversityTaoyuan CityTaiwan
| | - Rong‐Kuo Lyu
- Department of Neurology, Linkou Medical CenterChang Gung Memorial HospitalTaoyuan CityTaiwan
- College of MedicineChang Gung UniversityTaoyuan CityTaiwan
| | - Chun‐Che Chu
- Department of Neurology, Linkou Medical CenterChang Gung Memorial HospitalTaoyuan CityTaiwan
- College of MedicineChang Gung UniversityTaoyuan CityTaiwan
| | - Long‐Sun Ro
- Department of Neurology, Linkou Medical CenterChang Gung Memorial HospitalTaoyuan CityTaiwan
- College of MedicineChang Gung UniversityTaoyuan CityTaiwan
| | - Ming‐Feng Liao
- Department of Neurology, Linkou Medical CenterChang Gung Memorial HospitalTaoyuan CityTaiwan
- College of MedicineChang Gung UniversityTaoyuan CityTaiwan
| | - Hong‐Shiu Chang
- Department of Neurology, Linkou Medical CenterChang Gung Memorial HospitalTaoyuan CityTaiwan
- College of MedicineChang Gung UniversityTaoyuan CityTaiwan
| | - Yi‐Ching Weng
- Department of Neurology, Linkou Medical CenterChang Gung Memorial HospitalTaoyuan CityTaiwan
- College of MedicineChang Gung UniversityTaoyuan CityTaiwan
| | - Jawl‐Shan Hwang
- College of MedicineChang Gung UniversityTaoyuan CityTaiwan
- Division of Endocrinology and Metabolism, Department of Internal Medicine, Linkou Medical CenterChang Gung Memorial HospitalTaoyuan CityTaiwan
| |
Collapse
|
8
|
Hepsomali P, Costabile A, Schoemaker M, Imakulata F, Allen P. Adherence to unhealthy diets is associated with altered frontal gamma-aminobutyric acid and glutamate concentrations and grey matter volume: preliminary findings. Nutr Neurosci 2025; 28:125-137. [PMID: 38794782 DOI: 10.1080/1028415x.2024.2355603] [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: 05/26/2024]
Abstract
OBJECTIVES Common mental disorders (CMD) are associated with impaired frontal excitatory/inhibitory (E/I) balance and reduced grey matter volume (GMV). Larger GMV (in the areas that are implicated in CMD-pathology) and improved CMD-symptomatology have been observed in individuals who adhere to high quality diets. Moreover, preclinical studies have shown altered neurometabolites (primarily gamma-aminobutyric acid: GABA and glutamate: GLU) in relation to diet quality. However, neurochemical correlates of diet quality and how these neurobiological changes are associated with CMD and with its transdiagnostic factor, rumination, is unknown in humans. Therefore, in this study, we examined the associations between diet quality and frontal cortex neuro-chemistry and structure, as well as CMD and rumination in humans. METHODS Thirty adults were classified into high and low diet quality groups and underwent 1H-MRS to measure medial prefrontal cortex (mPFC) metabolite concentrations and volumetric imaging to measure GMV. RESULTS Low (vs High) diet quality group had reduced mPFC-GABA and elevated mPFC-GLU concentrations, as well as reduced right precentral gyrus (rPCG) GMV. However, CMD and rumination were not associated with diet quality. Notably, we observed a significant negative correlation between rumination and rPCG-GMV and a marginally significant association between rumination and mPFC-GLU concentrations. There was also a marginally significant association between mPFC-GLU concentrations and rPCG-GMV. DISCUSSION Adhering to unhealthy dietary patterns may be associated with compromised E/I balance, and this could affect GMV, and subsequently, rumination.
Collapse
Affiliation(s)
- Piril Hepsomali
- School of Psychology and Clinical Language Sciences, University of Reading, Reading, UK
| | - Adele Costabile
- School of Life and Health Sciences, University of Roehampton, London, UK
| | | | | | - Paul Allen
- Department of Neuroimaging, Kings College London, Institute of Psychology and Neuroscience, London, UK
| |
Collapse
|
9
|
Khan SR, Van JAD, Xiangyu Z, Alexeeff SE, Razani B, Wheeler MB, Gunderson EP. Early Postpartum Metabolic Heterogeneity Among Women Who Progressed to Type 2 Diabetes After Gestational Diabetes: A Prospective Cohort. Diabetes Metab Res Rev 2025; 41:e70027. [PMID: 39810724 PMCID: PMC11733828 DOI: 10.1002/dmrr.70027] [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: 03/21/2024] [Revised: 12/09/2024] [Accepted: 12/16/2024] [Indexed: 01/16/2025]
Abstract
AIMS Gestational diabetes mellitus (GDM) poses a significant risk for developing type 2 diabetes mellitus (T2D) and exhibits heterogeneity. However, understanding the link between different types of post-GDM individuals without diabetes and their progression to T2D is crucial to advance personalised medicine approaches. MATERIALS AND METHODS We employed a discovery-based unsupervised machine learning clustering method to generate clustering models for analysing metabolomics, clinical, and biochemical datasets. For this analysis, we selected 225 women who later developed T2D during the 12-year follow-up period from the cohort of 1010 women who returned to a non-diabetic state at 6-9 weeks (study baseline) after a GDM pregnancy based on 2-h 75 g research OGTTs. The optimal model was selected by assessing Bayesian Information Criterion values, class separation performance, and the potential for clinically distinguishable clusters, accounting for participant prenatal and early postpartum characteristics. RESULTS The selected model comprises three clusters: pancreatic beta cell dysfunction (cluster-β: median HOMA-B 161.3 and median HOMA-IR 3.8), insulin-resistance (cluster-IR: median HOMA-B 630.5 and median HOMA-IR 16.8), and a mixed cluster (cluster-mixed: median HOMA-B 307.2 and median HOMA-IR 8.6). These clusters are distinguishable based on postpartum blood test parameters such as glucose tolerance, HOMA indices, and fasting lipid profiles including triglycerides, leptin, HDL-c, and adiponectin, as well as participant age and BMI. Metabolomic analysis identified unique molecular signatures for each cluster. However, the time to T2D onset was not statistically significant among the three clusters (p = 0.22). CONCLUSION This study enhances our understanding of the heterogeneity of early postpartum metabolic profiles that characterise the future onset of T2D diabetes in a diverse cohort of women with GDM, revealing insights into distinct mechanisms and personalised intervention strategies for the prevention of T2D.
Collapse
Affiliation(s)
- Saifur R. Khan
- Department of MedicineDivision of CardiologyUniversity of PittsburghPittsburghPennsylvaniaUSA
- Vascular Medicine InstituteUniversity of PittsburghPittsburghPennsylvaniaUSA
- VA Medical CenterPittsburghPennsylvaniaUSA
- Center for ImmunometabolismUniversity of PittsburghPittsburghPennsylvaniaUSA
| | - Julie A. D. Van
- Departments of Physiology and MedicineUniversity of TorontoTorontoCanada
| | - Zhang Xiangyu
- Department of MedicineDivision of CardiologyUniversity of PittsburghPittsburghPennsylvaniaUSA
- Vascular Medicine InstituteUniversity of PittsburghPittsburghPennsylvaniaUSA
- VA Medical CenterPittsburghPennsylvaniaUSA
- Center for ImmunometabolismUniversity of PittsburghPittsburghPennsylvaniaUSA
| | - Stacey E. Alexeeff
- Division of ResearchKaiser Permanente Northern CaliforniaPleasantonCaliforniaUSA
| | - Babak Razani
- Department of MedicineDivision of CardiologyUniversity of PittsburghPittsburghPennsylvaniaUSA
- Vascular Medicine InstituteUniversity of PittsburghPittsburghPennsylvaniaUSA
- VA Medical CenterPittsburghPennsylvaniaUSA
- Center for ImmunometabolismUniversity of PittsburghPittsburghPennsylvaniaUSA
| | - Michael B. Wheeler
- Departments of Physiology and MedicineUniversity of TorontoTorontoCanada
| | - Erica P. Gunderson
- Division of ResearchKaiser Permanente Northern CaliforniaPleasantonCaliforniaUSA
- Department of Health Systems ScienceKaiser Permanente Bernard J. Tyson School of MedicinePasadenaCaliforniaUSA
| |
Collapse
|
10
|
Guzmán TJ, Klöpper N, Gurrola-Díaz CM, Düfer M. Inhibition of mTOR prevents glucotoxicity-mediated increase of SA-beta-gal, p16 INK4a, and insulin hypersecretion, without restoring electrical features of mouse pancreatic islets. Biogerontology 2024; 25:819-836. [PMID: 38748336 PMCID: PMC11374829 DOI: 10.1007/s10522-024-10107-9] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2024] [Accepted: 04/16/2024] [Indexed: 09/05/2024]
Abstract
An over-activation of the mechanistic target of rapamycin (mTOR) pathway promotes senescence and age-related diseases like type 2 diabetes. Besides, the regenerative potential of pancreatic islets deteriorates with aging. Nevertheless, the role of mTOR on senescence promoted by metabolic stress in islet cells as well as its relevance for electrophysiological aspects is not yet known. Here, we investigated whether parameters suggested to be indicative for senescence are induced in vitro in mouse islet cells by glucotoxicity and if mTOR inhibition plays a protective role against this. Islet cells exhibit a significant increase (~ 76%) in senescence-associated beta-galactosidase (SA-beta-gal) activity after exposure to glucotoxicity for 72 h. Glucotoxicity does not markedly influence p16INK4a protein within 72 h, but p16INK4a levels increase significantly after a 7-days incubation period. mTOR inhibition with a low rapamycin concentration (1 nM) entirely prevents the glucotoxicity-mediated increase of SA-beta-gal and p16INK4a. At the functional level, reactive oxygen species, calcium homeostasis, and electrical activity are disturbed by glucotoxicity, and rapamycin fails to prevent this. In contrast, rapamycin significantly attenuates the insulin hypersecretion promoted by glucotoxicity by modifying the mRNA levels of Vamp2 and Snap25 genes, related to insulin exocytosis. Our data indicate an influence of glucotoxicity on pancreatic islet-cell senescence and a reduction of the senescence markers by mTOR inhibition, which is relevant to preserve the regenerative potential of the islets. Decreasing the influence of mTOR on islet cells exposed to glucotoxicity attenuates insulin hypersecretion, but is not sufficient to prevent electrophysiological disturbances, indicating the involvement of mTOR-independent mechanisms.
Collapse
Affiliation(s)
- Tereso J Guzmán
- Department of Pharmacology, Institute of Pharmaceutical and Medicinal Chemistry, University of Münster, Corrensstraße 48, 48149, Münster, Germany.
- Departamento de Biología Molecular y Genómica, Universidad de Guadalajara, Instituto de Investigación en Enfermedades Crónico-Degenerativas, Centro Universitario de Ciencias de la Salud, 44340, Guadalajara, Jalisco, México.
| | - Nina Klöpper
- Department of Pharmacology, Institute of Pharmaceutical and Medicinal Chemistry, University of Münster, Corrensstraße 48, 48149, Münster, Germany
| | - Carmen M Gurrola-Díaz
- Departamento de Biología Molecular y Genómica, Universidad de Guadalajara, Instituto de Investigación en Enfermedades Crónico-Degenerativas, Centro Universitario de Ciencias de la Salud, 44340, Guadalajara, Jalisco, México
| | - Martina Düfer
- Department of Pharmacology, Institute of Pharmaceutical and Medicinal Chemistry, University of Münster, Corrensstraße 48, 48149, Münster, Germany.
| |
Collapse
|
11
|
Bishop A, Romero JC, Tonapi S, Parihar M, Loranc E, Miller H, Lawrence L, Bassani N, Robledo D, Cao L, Nie J, Kanda K, Stoja A, Garcia N, Gorthi A, Stoveken B, Lane A, Fan T, Cassel T, Zha S, Musi N. ATM phosphorylation of CD98HC increases antiporter membrane localization and prevents chronic toxic glutamate accumulation in Ataxia telangiectasia. RESEARCH SQUARE 2024:rs.3.rs-4947457. [PMID: 39281865 PMCID: PMC11398575 DOI: 10.21203/rs.3.rs-4947457/v1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 09/18/2024]
Abstract
Ataxia telangiectasia (A-T) is a rare genetic disorder characterized by neurological defects, immunodeficiency, cancer predisposition, radiosensitivity, decreased blood vessel integrity, and diabetes. ATM, the protein mutated in A-T, responds to DNA damage and oxidative stress, but its functional relationship to the progressive clinical manifestation of A-T is not understood. CD98HC chaperones cystine/glutamate (xc -) and cationic/neutral amino acid (y+L) antiporters to the cell membrane, and CD98HC phosphorylation by ATM accelerates membrane localization to acutely increase amino acid transport. Loss of ATM impacts tissues reliant on SLC family antiporters relevant to A-T phenotypes, such as endothelial cells (telangiectasia) and pancreatic α-cells (fatty liver and diabetes) with toxic glutamate accumulation. Bypassing the antiporters restores intracellular metabolic balance both in ATM-deficient cells and mouse models. These findings provide new insight into the long-known benefits of N-acetyl cysteine to A-T cells beyond oxidative stress through removing excess glutamate by production of glutathione.
Collapse
Affiliation(s)
| | | | | | | | - Eva Loranc
- University of Texas Health at San Antonio
| | | | | | | | | | - Lin Cao
- University of Texas Health at San Antonio
| | - Jia Nie
- University of Texas Health at San Antonio
| | | | | | | | | | | | | | - Teresa Fan
- Center for Environmental and Systems Biochemistry, University of Kentucky
| | | | | | | |
Collapse
|
12
|
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
|
13
|
Galli A, Moretti S, Dule N, Di Cairano ES, Castagna M, Marciani P, Battaglia C, Bertuzzi F, Fiorina P, Pastore I, La Rosa S, Davalli A, Folli F, Perego C. Hyperglycemia impairs EAAT2 glutamate transporter trafficking and glutamate clearance in islets of Langerhans: implications for type 2 diabetes pathogenesis and treatment. Am J Physiol Endocrinol Metab 2024; 327:E27-E41. [PMID: 38690938 PMCID: PMC11390119 DOI: 10.1152/ajpendo.00069.2024] [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: 02/07/2024] [Revised: 04/18/2024] [Accepted: 04/21/2024] [Indexed: 05/03/2024]
Abstract
Pancreatic endocrine cells employ a sophisticated system of paracrine and autocrine signals to synchronize their activities, including glutamate, which controls hormone release and β-cell viability by acting on glutamate receptors expressed by endocrine cells. We here investigate whether alteration of the excitatory amino acid transporter 2 (EAAT2), the major glutamate clearance system in the islet, may occur in type 2 diabetes mellitus and contribute to β-cell dysfunction. Increased EAAT2 intracellular localization was evident in islets of Langerhans from T2DM subjects as compared with healthy control subjects, despite similar expression levels. Chronic treatment of islets from healthy donors with high-glucose concentrations led to the transporter internalization in vesicular compartments and reduced [H3]-d-glutamate uptake (65 ± 5% inhibition), phenocopying the findings in T2DM pancreatic sections. The transporter relocalization was associated with decreased Akt phosphorylation protein levels, suggesting an involvement of the phosphoinositide 3-kinase (PI3K)/Akt pathway in the process. In line with this, PI3K inhibition by a 100-µM LY294002 treatment in human and clonal β-cells caused the transporter relocalization in intracellular compartments and significantly reduced the glutamate uptake compared to control conditions, suggesting that hyperglycemia changes the trafficking of the transporter to the plasma membrane. Upregulation of the glutamate transporter upon treatment with the antibiotic ceftriaxone rescued hyperglycemia-induced β-cells dysfunction and death. Our data underscore the significance of EAAT2 in regulating islet physiology and provide a rationale for potential therapeutic targeting of this transporter to preserve β-cell survival and function in diabetes.NEW & NOTEWORTHY The glutamate transporter SLC1A2/excitatory amino acid transporter 2 (EAAT2) is expressed on the plasma membrane of pancreatic β-cells and controls islet glutamate clearance and β-cells survival. We found that the EAAT2 membrane expression is lost in the islets of Langerhans from type 2 diabetes mellitus (T2DM) patients due to hyperglycemia-induced downregulation of the phosphoinositide 3-kinase/Akt pathway and modification of its intracellular trafficking. Pharmacological rescue of EAAT2 expression prevents β-cell dysfunction and death, suggesting EAAT2 as a new potential target of intervention in T2DM.
Collapse
Affiliation(s)
- Alessandra Galli
- Laboratory of Molecular and Cellular Physiology, Department of Excellence of Pharmacological and Biomolecular Sciences, Università degli Studi di Milano, Milan, Italy
| | - Stefania Moretti
- Laboratory of Molecular and Cellular Physiology, Department of Excellence of Pharmacological and Biomolecular Sciences, Università degli Studi di Milano, Milan, Italy
| | - Nevia Dule
- Laboratory of Molecular and Cellular Physiology, Department of Excellence of Pharmacological and Biomolecular Sciences, Università degli Studi di Milano, Milan, Italy
| | - Eliana Sara Di Cairano
- Laboratory of Molecular and Cellular Physiology, Department of Excellence of Pharmacological and Biomolecular Sciences, Università degli Studi di Milano, Milan, Italy
| | - Michela Castagna
- Laboratory of Molecular and Cellular Physiology, Department of Excellence of Pharmacological and Biomolecular Sciences, Università degli Studi di Milano, Milan, Italy
| | - Paola Marciani
- Laboratory of Molecular and Cellular Physiology, Department of Excellence of Pharmacological and Biomolecular Sciences, Università degli Studi di Milano, Milan, Italy
| | - Cristina Battaglia
- Department of Medical Biotechnology and Translational Medicine, Università degli Studi di Milano, Milan, Italy
| | | | - Paolo Fiorina
- Department of Biomedical and Clinical Sciences "L. Sacco,"Università degli Studi di Milano, Milan, Italy
- Endocrinology Unit, ASST Fatebenefratelli-Sacco, Milan, Italy
| | - Ida Pastore
- Endocrinology Unit, ASST Fatebenefratelli-Sacco, Milan, Italy
| | - Stefano La Rosa
- Unit of Pathology, Department of Oncology, ASST Sette Laghi, Varese, Italy
- Department of Medicine and Technological Innovation, Università degli Studi dell'Insubria, Varese, Italy
| | - Alberto Davalli
- Diabetes and Endocrinology Unit, Department of Internal Medicine, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Franco Folli
- Department of Health Sciences, Università degli Studi di Milano, Milan, Italy
| | - Carla Perego
- Laboratory of Molecular and Cellular Physiology, Department of Excellence of Pharmacological and Biomolecular Sciences, Università degli Studi di Milano, Milan, Italy
| |
Collapse
|
14
|
Park H, Lee Y, Hwang J, Lee Y. Ultra-processed food consumption and increased risk of metabolic syndrome in Korean adults: A cross-sectional analysis of the KNHANES 2016-2020. Nutrition 2024; 122:112374. [PMID: 38430845 DOI: 10.1016/j.nut.2024.112374] [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: 10/09/2023] [Revised: 12/20/2023] [Accepted: 01/27/2024] [Indexed: 03/05/2024]
Abstract
OBJECTIVE This study aimed to investigate the association between ultra-processed food (UPF) intake and the risk for metabolic syndrome (MetS) in Korean adults. METHODS The study consisted of 22 688 Korean adults ≥19 y of age from the Korea National Health and Nutrition Examination Survey (KNHANES) 2016-2020. The NOVA classification categorizes foods according to the nature, extent, and purpose of industrial processing. MetS was defined based on the National Cholesterol Education Program Adult Treatment Panel III criteria and a modified waist circumference cut-off for Korean adults. We estimated the usual percent total food intake from UPFs. We used multivariate logistic regression to assess the association between UPFs and risk for MetS, adjusted for age, sex, education level, income level, smoking status, alcohol drinking, physical activity, and total energy intake. We further analyzed the association of UPFs with each component of MetS. RESULTS The median usual percent total food intake from UPFs was 22%, and the midpoint of intake ranged from 3% (quartile 1) to 48% (quartile 4). The group with the highest UPF consumption had a 19% higher risk for developing MetS than the lowest quartile of UPF consumption (odds ratio [OR],1.19; 95% confidence interval [CI], 1.06-1.33; Ptrend = 0.006). In analysis of the relationship between UPF intake and MetS components, a higher UPF was associated with an increased risk for hypertension (OR, 1.13; 95% CI, 1.01-1.26; Ptrend = 0.037) and abdominal obesity (OR, 1.19; 95% CI, 1.07-1.33; Ptrend = 0.001), but had no significant association with other components (hyperglycemia, hypertriacylglycerolmia, and low high-density lipoprotein cholesterol, all P > 0.05). CONCLUSION Higher UPF contribution to total daily food intake is associated with an increased risk for MetS, particularly with a higher risk for hypertension and abdominal obesity.
Collapse
Affiliation(s)
- Hansol Park
- Department of Food and Nutrition, Myongji University, Yongin, Korea
| | - Youngmi Lee
- Department of Food and Nutrition, Myongji University, Yongin, Korea
| | - Jinah Hwang
- Department of Food and Nutrition, Myongji University, Yongin, Korea
| | - Yujin Lee
- Department of Food and Nutrition, Myongji University, Yongin, Korea.
| |
Collapse
|
15
|
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
|
16
|
Wörmeyer L, Nortmann O, Hamacher A, Uhlemeyer C, Belgardt B, Eberhard D, Mayatepek E, Meissner T, Lammert E, Welters A. The N-Methyl-D-Aspartate Receptor Antagonist Dextromethorphan Improves Glucose Homeostasis and Preserves Pancreatic Islets in NOD Mice. Horm Metab Res 2024; 56:223-234. [PMID: 38168730 PMCID: PMC10901624 DOI: 10.1055/a-2236-8625] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/05/2024]
Abstract
For treatment of type 1 diabetes mellitus, a combination of immune-based interventions and medication to promote beta-cell survival and proliferation has been proposed. Dextromethorphan (DXM) is an N-methyl-D-aspartate receptor antagonist with a good safety profile, and to date, preclinical and clinical evidence for blood glucose-lowering and islet-cell-protective effects of DXM have only been provided for animals and individuals with type 2 diabetes mellitus. Here, we assessed the potential anti-diabetic effects of DXM in the non-obese diabetic mouse model of type 1 diabetes. More specifically, we showed that DXM treatment led to five-fold higher numbers of pancreatic islets and more than two-fold larger alpha- and beta-cell areas compared to untreated mice. Further, DXM treatment improved glucose homeostasis and reduced diabetes incidence by 50%. Our data highlight DXM as a novel candidate for adjunct treatment of preclinical or recent-onset type 1 diabetes.
Collapse
Affiliation(s)
- Laura Wörmeyer
- Department of General Pediatrics, Neonatology and Pediatric Cardiology, Medical Faculty and University Hospital Düsseldorf, Heinrich Heine University Düsseldorf, Düsseldorf, Germany
- Institute of Metabolic Physiology, Heinrich Heine University Düsseldorf, Düsseldorf, Germany
| | - Oliver Nortmann
- Department of General Pediatrics, Neonatology and Pediatric Cardiology, Medical Faculty and University Hospital Düsseldorf, Heinrich Heine University Düsseldorf, Düsseldorf, Germany
- Institute of Metabolic Physiology, Heinrich Heine University Düsseldorf, Düsseldorf, Germany
| | - Anna Hamacher
- Institute of Metabolic Physiology, Heinrich Heine University Düsseldorf, Düsseldorf, Germany
| | - Celina Uhlemeyer
- Institute for Vascular and Islet Cell Biology, German Diabetes Center (DDZ), Leibniz Center for Diabetes Research, Heinrich Heine University Düsseldorf, Düsseldorf, Germany
- German Center for Diabetes Research (DZD e.V.), Helmholtz Zentrum München Deutsches Forschungszentrum für Gesundheit und Umwelt, Neuherberg, Germany
| | - Bengt Belgardt
- Institute for Vascular and Islet Cell Biology, German Diabetes Center (DDZ), Leibniz Center for Diabetes Research, Heinrich Heine University Düsseldorf, Düsseldorf, Germany
- German Center for Diabetes Research (DZD e.V.), Helmholtz Zentrum München Deutsches Forschungszentrum für Gesundheit und Umwelt, Neuherberg, Germany
| | - Daniel Eberhard
- Institute of Metabolic Physiology, Heinrich Heine University Düsseldorf, Düsseldorf, Germany
| | - Ertan Mayatepek
- Department of General Pediatrics, Neonatology and Pediatric Cardiology, Medical Faculty and University Hospital Düsseldorf, Heinrich Heine University Düsseldorf, Düsseldorf, Germany
| | - Thomas Meissner
- Department of General Pediatrics, Neonatology and Pediatric Cardiology, Medical Faculty and University Hospital Düsseldorf, Heinrich Heine University Düsseldorf, Düsseldorf, Germany
| | - Eckhard Lammert
- Institute of Metabolic Physiology, Heinrich Heine University Düsseldorf, Düsseldorf, Germany
- Institute for Vascular and Islet Cell Biology, German Diabetes Center (DDZ), Leibniz Center for Diabetes Research, Heinrich Heine University Düsseldorf, Düsseldorf, Germany
- German Center for Diabetes Research (DZD e.V.), Helmholtz Zentrum München Deutsches Forschungszentrum für Gesundheit und Umwelt, Neuherberg, Germany
| | - Alena Welters
- Department of General Pediatrics, Neonatology and Pediatric Cardiology, Medical Faculty and University Hospital Düsseldorf, Heinrich Heine University Düsseldorf, Düsseldorf, Germany
- Institute for Vascular and Islet Cell Biology, German Diabetes Center (DDZ), Leibniz Center for Diabetes Research, Heinrich Heine University Düsseldorf, Düsseldorf, Germany
| |
Collapse
|
17
|
Hristov M, Nankova A, Andreeva-Gateva P. Alterations of the glutamatergic system in diabetes mellitus. Metab Brain Dis 2024; 39:321-333. [PMID: 37747631 DOI: 10.1007/s11011-023-01299-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] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/16/2023] [Accepted: 09/17/2023] [Indexed: 09/26/2023]
Abstract
Diabetes mellitus (DM) is a chronic disease characterized by elevated blood glucose levels caused by a lack of insulin production (type 1 diabetes) or insulin resistance (type 2 diabetes). It is well known that DM is associated with cognitive deficits and metabolic and neurophysiological changes in the brain. Glutamate is the main excitatory neurotransmitter in the central nervous system that plays a key role in synaptic plasticity, learning, and memory processes. An increasing number of studies have suggested that abnormal activity of the glutamatergic system is implicated in the pathophysiology of DM. Dysfunction of glutamatergic neurotransmission in the central nervous system can provide an important neurobiological substrate for many disorders. Magnetic resonance spectroscopy (MRS) is a non-invasive technique that allows a better understanding of the central nervous system factors by measuring in vivo the concentrations of brain metabolites within the area of interest. Here, we briefly review the MRS studies that have examined glutamate levels in the brain of patients with DM. The present article also summarizes the available data on abnormalities in glutamatergic neurotransmission observed in different animal models of DM. In addition, the role of gut microbiota in the development of glutamatergic alterations in DM is addressed. We speculate that therapeutic strategies targeting the glutamatergic system may be beneficial in the treatment of central nervous system-related changes in diabetic patients.
Collapse
Affiliation(s)
- Milen Hristov
- Department of Pharmacology and Toxicology, Faculty of Medicine, Medical University of Sofia, 2 "Zdrave" St, Sofia, 1431, Bulgaria.
| | - Anelia Nankova
- Department of Endocrinology, Faculty of Medicine, Medical University of Sofia, Sofia, 1431, Bulgaria
| | - Pavlina Andreeva-Gateva
- Department of Pharmacology and Toxicology, Faculty of Medicine, Medical University of Sofia, 2 "Zdrave" St, Sofia, 1431, Bulgaria
| |
Collapse
|
18
|
Oruc A, Oruc KY, Yanar K, Mengi M, Caglar A, Kurt BO, Altan M, Sonmez OF, Cakatay U, Uzun H, Simsek G. The Role of Glycogen Synthase Kinase-3β in the Zinc-Mediated Neuroprotective Effect of Metformin in Rats with Glutamate Neurotoxicity. Biol Trace Elem Res 2024; 202:233-245. [PMID: 37071257 DOI: 10.1007/s12011-023-03667-3] [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: 03/08/2023] [Accepted: 04/11/2023] [Indexed: 04/19/2023]
Abstract
Metformin has been suggested to have protective effects on the central nervous system, but the mechanism is unknown. The similarity between the effects of metformin and the inhibition of glycogen synthase kinase (GSK)-3β suggests that metformin may inhibit GSK-3β. In addition, zinc is an important element that inhibits GSK-3β by phosphorylation. In this study, we investigated whether the effects of metformin on neuroprotection and neuronal survival were mediated by zinc-dependent inhibition of GSK-3β in rats with glutamate-induced neurotoxicity. Forty adult male rats were divided into 5 groups: control, glutamate, metformin + glutamate, zinc deficiency + glutamate, and zinc deficiency + metformin + glutamate. Zinc deficiency was induced with a zinc-poor pellet. Metformin was orally administered for 35 days. D-glutamic acid was intraperitoneally administered on the 35th day. On the 38th day, neurodegeneration was examined histopathologically, and the effects on neuronal protection and survival were evaluated via intracellular S-100β immunohistochemical staining. The findings were examined in relation to nonphosphorylated (active) GSK-3β levels and oxidative stress parameters in brain tissue and blood. Neurodegeneration was increased (p < 0.05) in rats fed a zinc-deficient diet. Active GSK-3β levels were increased in groups with neurodegeneration (p < 0.01). Decreased neurodegeneration, increased neuronal survival (p < 0.01), decreased active GSK-3β (p < 0.01) levels and oxidative stress parameters, and increased antioxidant parameters were observed in groups treated with metformin (p < 0.01). Metformin had fewer protective effects on rats fed a zinc-deficient diet. Metformin may exert neuroprotective effects and increase S-100β-mediated neuronal survival by zinc-dependent inhibition of GSK-3β during glutamate neurotoxicity.
Collapse
Affiliation(s)
- Aykut Oruc
- Department of Physiology, Cerrahpaşa Medical Faculty, Istanbul University-Cerrahpaşa, Istanbul, Turkey.
| | - Kadriye Yagmur Oruc
- Department of Physiology, Cerrahpaşa Medical Faculty, Istanbul University-Cerrahpaşa, Istanbul, Turkey
- Department of Physiology, Faculty of Medicine, Istinye University, Istanbul, Turkey
| | - Karolin Yanar
- Department of Medical Biochemistry, Cerrahpaşa Medical Faculty, Istanbul University-Cerrahpaşa, Istanbul, Turkey
| | - Murat Mengi
- Department of Physiology, Medical Faculty, Namık Kemal University, Tekirdag, Turkey
| | - Aysel Caglar
- Department of Pathology, Bagcilar Training and Research Hospital, Istanbul, Turkey
| | - Bahar Ozturk Kurt
- Department of Biophysics, Cerrahpaşa Medical Faculty, Istanbul University-Cerrahpaşa, Istanbul, Turkey
| | - Mehmet Altan
- Department of Physiology, Cerrahpaşa Medical Faculty, Istanbul University-Cerrahpaşa, Istanbul, Turkey
| | - Osman Fuat Sonmez
- Department of Physiology, Cerrahpaşa Medical Faculty, Istanbul University-Cerrahpaşa, Istanbul, Turkey
| | - Ufuk Cakatay
- Department of Medical Biochemistry, Cerrahpaşa Medical Faculty, Istanbul University-Cerrahpaşa, Istanbul, Turkey
| | - Hafize Uzun
- Department of Medical Biochemistry, Faculty of Medicine, Istanbul Atlas University, Istanbul, Turkey
| | - Gonul Simsek
- Department of Physiology, Cerrahpaşa Medical Faculty, Istanbul University-Cerrahpaşa, Istanbul, Turkey
| |
Collapse
|
19
|
Cheng HP, Feng DD, Li XH, Gao LH, Qiu YJ, Liang XY, Zhou Y, Huang P, Shao M, Zhang YN, Chang YF, Fu JF, Huang YH, Liu W, Tang SY, Li C, Luo ZQ. NMDA receptor activation induces damage of alveolar type II cells and lung fibrogenesis through ferroptosis. BIOCHIMICA ET BIOPHYSICA ACTA. MOLECULAR CELL RESEARCH 2023; 1870:119535. [PMID: 37451346 DOI: 10.1016/j.bbamcr.2023.119535] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/11/2023] [Revised: 07/03/2023] [Accepted: 07/05/2023] [Indexed: 07/18/2023]
Abstract
Ferroptosis, a newly discovered type of regulated cell death, has been implicated in numerous human diseases. Idiopathic pulmonary fibrosis (IPF) is a progressive and ultimately fatal interstitial lung disease with poor prognosis and limited treatment options. Emerging evidence has linked ferroptosis and glutamate-determined cell fate which is considered a new light on the etiology of pulmonary fibrosis. Here, we observed that N-methyl d-aspartate receptor (NMDAR) activation promoted cell damage and iron deposition in MLE-12 cells in a dose-, time-, and receptor-dependent manner. This mediated substantial Ca2+ influx, upregulated the expression levels of nNOS and IRP1, and affected intracellular iron homeostasis by regulating the expression of iron transport-related proteins (i.e., TFR1, DMT1, and FPN). Excessive iron load promoted the continuous accumulation of total intracellular and mitochondrial reactive oxygen species, which ultimately led to ferroptosis. NMDAR inhibition reduced lung injury and pulmonary fibrosis in bleomycin-induced mice. Bleomycin stimulation upregulated the expression of NMDAR1, nNOS, and IRP1 in mouse lung tissues, which ultimately led to iron deposition via regulation of the expression of various iron metabolism-related genes. NMDAR activation initiated the pulmonary fibrosis process by inducing iron deposition in lung tissues and ferroptosis of alveolar type II cells. Our data suggest that NMDAR activation regulates the expression of iron metabolism-related genes by promoting calcium influx, increasing nNOS and IRP1 expression, and increasing iron deposition by affecting cellular iron homeostasis, ultimately leading to mitochondrial damage, mitochondrial dysfunction, and ferroptosis. NMDAR activation-induced ferroptosis of alveolar type II cells might be a key event to the initiation of pulmonary fibrosis.
Collapse
Affiliation(s)
- Hai-Peng Cheng
- Department of Physiology, Xiangya School of Medicine, Central South University, Changsha, Hunan, China; Department of Pathology, The Second Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Dan-Dan Feng
- Department of Physiology, Xiangya School of Medicine, Central South University, Changsha, Hunan, China
| | - Xiao-Hong Li
- Department of Pathology, The Second Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Li-Hua Gao
- Department of Physiology, Xiangya School of Medicine, Central South University, Changsha, Hunan, China
| | - Yu-Jia Qiu
- Department of Physiology, Xiangya School of Medicine, Central South University, Changsha, Hunan, China
| | - Xing-Yue Liang
- Department of Physiology, Xiangya School of Medicine, Central South University, Changsha, Hunan, China
| | - Yan Zhou
- Department of Physiology, Xiangya School of Medicine, Central South University, Changsha, Hunan, China
| | - Pu Huang
- Department of Physiology, Xiangya School of Medicine, Central South University, Changsha, Hunan, China
| | - Min Shao
- Department of Physiology, Xiangya School of Medicine, Central South University, Changsha, Hunan, China
| | - Yun-Na Zhang
- Department of Physiology, Xiangya School of Medicine, Central South University, Changsha, Hunan, China
| | - Yan-Fen Chang
- Department of Physiology, Xiangya School of Medicine, Central South University, Changsha, Hunan, China
| | - Jia-Feng Fu
- Department of Physiology, Xiangya School of Medicine, Central South University, Changsha, Hunan, China
| | - Yan-Hong Huang
- Department of Physiology, Xiangya School of Medicine, 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
| | - Chen Li
- Department of Physiology, Changzhi Medical College, Changzhi, Shanxi, China.
| | - Zi-Qiang Luo
- Department of Physiology, Xiangya School of Medicine, Central South University, Changsha, Hunan, China; Hunan Key Laboratory of Organ Fibrosis, Central South University, Changsha, Hunan, China.
| |
Collapse
|
20
|
Lee HJ, Yeom JW, Yun JH, Jang HB, Yoo M, Kim H, Koo SK, Lee H. Increased glutamate in type 2 diabetes in the Korean population is associated with increased plasminogen levels. J Diabetes 2023; 15:777-786. [PMID: 37314019 PMCID: PMC10509517 DOI: 10.1111/1753-0407.13429] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/26/2022] [Revised: 04/10/2023] [Accepted: 05/23/2023] [Indexed: 06/15/2023] Open
Abstract
BACKGROUND Glutamate is a major neurotransmitter, although it causes cytotoxicity and inflammation in nonneuronal organs. This study aimed to investigate the metabolic disorders in which glutamate, associated with type 2 diabetes onset, is induced in the liver. METHODS An analysis of Korean community-based Ansan-Ansung cohort study data as well as functional research using in vitro and mouse models were performed. RESULTS Groups with high plasma glutamate levels (T2, T3) had a significantly increased risk of diabetes incidence after 8 years, compared to the group with relatively low glutamate levels (T1). Analysis of the effect of glutamate on diabetes onset in vitro showed that glutamate induces insulin resistance by increasing glucose-related protein 78 (GRP78) and phosphoenolpyruvate carboxykinase (PEPCK) expression in SK-Hep-1 human liver cells. In addition, three different genes, FRMB4B, PLG, and PARD3, were significantly associated with glutamate and were identified via genome-wide association studies. Among glutamate-related genes, plasminogen (PLG) levels were most significantly increased in several environments in which insulin resistance was induced, and was also upregulated by glutamate. Glutamate-induced increase in PLG in liver cells was caused by metabotropic glutamate receptor 5 activation, and PLG levels were also upregulated after extracellular secretion. Moreover, glutamate increased the expression of plasminogen activator inhibitor-1 (PAI-1). Thus, extracellular secreted PLG cannot be converted to plasmin (fibrinolytic enzyme) by increased PAI-1. CONCLUSIONS Increased glutamate is closely associated with the development of diabetes, and it may cause metabolic disorders by inhibiting the fibrinolytic system, which plays an important role in determining blood clots, a hallmark of diabetes.
Collapse
Affiliation(s)
- Hyo Jung Lee
- Division of Endocrine and Kidney Disease Research, Department of Chronic Disease Convergence ResearchKorea National Institute of Health, Korea Disease Control and Prevention AgencyCheongju‐siChungcheongbuk‐doKorea
| | - Jeong Won Yeom
- Division of Endocrine and Kidney Disease Research, Department of Chronic Disease Convergence ResearchKorea National Institute of Health, Korea Disease Control and Prevention AgencyCheongju‐siChungcheongbuk‐doKorea
| | - Ji Ho Yun
- Division of Endocrine and Kidney Disease Research, Department of Chronic Disease Convergence ResearchKorea National Institute of Health, Korea Disease Control and Prevention AgencyCheongju‐siChungcheongbuk‐doKorea
| | - Han Byul Jang
- Division of Endocrine and Kidney Disease Research, Department of Chronic Disease Convergence ResearchKorea National Institute of Health, Korea Disease Control and Prevention AgencyCheongju‐siChungcheongbuk‐doKorea
| | - Min‐Gyu Yoo
- Division of Endocrine and Kidney Disease Research, Department of Chronic Disease Convergence ResearchKorea National Institute of Health, Korea Disease Control and Prevention AgencyCheongju‐siChungcheongbuk‐doKorea
| | - Hyo‐Jin Kim
- Division of Endocrine and Kidney Disease Research, Department of Chronic Disease Convergence ResearchKorea National Institute of Health, Korea Disease Control and Prevention AgencyCheongju‐siChungcheongbuk‐doKorea
| | - Soo Kyung Koo
- Division of Endocrine and Kidney Disease Research, Department of Chronic Disease Convergence ResearchKorea National Institute of Health, Korea Disease Control and Prevention AgencyCheongju‐siChungcheongbuk‐doKorea
| | - Hye‐Ja Lee
- Division of Endocrine and Kidney Disease Research, Department of Chronic Disease Convergence ResearchKorea National Institute of Health, Korea Disease Control and Prevention AgencyCheongju‐siChungcheongbuk‐doKorea
| |
Collapse
|
21
|
Li XH, Fu JJ, Shi XJ, Zhang YN, Shao M, Yue SJ, Li C, Luo ZQ. Sp1 mediated the inhibitory effect of glutamate on pulmonary surfactant synthesis. PLoS One 2023; 18:e0289530. [PMID: 37556489 PMCID: PMC10411742 DOI: 10.1371/journal.pone.0289530] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2022] [Accepted: 07/20/2023] [Indexed: 08/11/2023] Open
Abstract
BACKGROUND Studies have shown that the release of endogenous glutamate (Glu) participates in lung injury by activating N-methyl-D-aspartate receptor (NMDAR), but the mechanism is still unclear. This study was to investigate the effects and related mechanisms of Glu on the lipid synthesis of pulmonary surfactant (PS) in isolated rat lung tissues. METHODS The cultured lung tissues of adult SD rats were treated with Glu. The amount of [3H]-choline incorporation into phosphatidylcholine (PC) was detected. RT-PCR and Western blot were used to detect the changes of mRNA and protein expression of cytidine triphosphate: phosphocholine cytidylyltransferase alpha (CCTα), a key regulatory enzyme in PC biosynthesis. Western blot was used to detect the expression of NMDAR1, which is a functional subunit of NMDAR. Specific protein 1 (Sp1) expression plasmids were used. After transfected with Sp1 expression plasmids, the mRNA and protein levels of CCTα were detected by RT-PCR and Western blot in A549 cells. After treated with NMDA and MK-801, the mRNA and protein levels of Sp1 were detected by RT-PCR and Western blot in A549 cells. RESULTS Glu decreased the incorporation of [3H]-choline into PC in a concentration- and time- dependent manner. Glu treatment significantly reduced the mRNA and protein levels of CCTα in lungs. Glu treatment up-regulated NMDAR1 protein expression, and the NMDAR blocker MK-801 could partially reverse the reduction of [3H]-choline incorporation induced by Glu (10-4 mol/L) in lungs. After transfected with Sp1 plasmid for 30 h, the mRNA and protein expression levels of CCTα were increased and the protein expression of Sp1 was also up-regulated. After A549 cells were treated with NMDA, the level of Sp1 mRNA did not change significantly, but the expression of nucleus protein in Sp1 was significantly decreased, while the expression of cytoplasmic protein was significantly increased. However, MK-801could reverse these changes. CONCLUSIONS Glu reduced the biosynthesis of the main lipid PC in PS and inhibited CCTα expression by activating NMDAR, which were mediated by the inhibition of the nuclear translocation of Sp1 and the promoter activity of CCTα. In conclusion, NMDAR-mediated Glu toxicity leading to impaired PS synthesis may be a potential pathogenesis of lung injury.
Collapse
Affiliation(s)
- Xiao-Hong Li
- Department of Pathology, The Second Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Jie-Jun Fu
- Department of Physiology, Xiangya School of Medicine, Central South University, Changsha, Hunan, China
| | - Xiao-Juan Shi
- Department of Physiology, Xiangya School of Medicine, Central South University, Changsha, Hunan, China
| | - Yun-Na Zhang
- Department of Physiology, Xiangya School of Medicine, Central South University, Changsha, Hunan, China
| | - Min Shao
- Department of Physiology, Xiangya School of Medicine, Central South University, Changsha, Hunan, China
| | - Shao-Jie Yue
- Department of Pediatrics, Xiangya Hospital, Central South University, Changsha, China
| | - Chen Li
- Department of Physiology, Xiangya School of Medicine, Central South University, Changsha, Hunan, China
- Department of Physiology, Changzhi Medical College, Changzhi, Shanxi, China
| | - Zi-Qiang Luo
- Department of Physiology, Xiangya School of Medicine, Central South University, Changsha, Hunan, China
- Hunan Key Laboratory of Organ Fibrosis, Central South University, Changsha, Hunan, China
| |
Collapse
|
22
|
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
|
23
|
Pan F, Bu L, Wu K, Wang A, Xu X. PKD2/polycystin-2 inhibits LPS-induced acute lung injury in vitro and in vivo by activating autophagy. BMC Pulm Med 2023; 23:171. [PMID: 37198573 DOI: 10.1186/s12890-023-02449-w] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2022] [Accepted: 04/21/2023] [Indexed: 05/19/2023] Open
Abstract
Polycystin-2 (PC2), which is a transmembrane protein encoded by the PKD2 gene, plays an important role in kidney disease, but its role in lipopolysaccharide (LPS)-induced acute lung injury (ALI) is unclear. We overexpressed PKD2 in lung epithelial cells in vitro and in vivo and examined the role of PKD2 in the inflammatory response induced by LPS in vitro and in vivo. Overexpression of PKD2 significantly decreased production of the inflammatory factors TNF-α, IL-1β, and IL-6 in LPS-treated lung epithelial cells. Moreover, pretreatment with 3-methyladenine (3-MA), an autophagy inhibitor, reversed the inhibitory effect of PKD2 overexpression on the secretion of inflammatory factors in LPS-treated lung epithelial cells. We further demonstrated that overexpression of PKD2 could inhibit LPS-induced downregulation of the LC3BII protein levels and upregulation of SQSTM1/P62 protein levels in lung epithelial cells. Moreover, we found that LPS-induced changes in the lung wet/dry (W/D) weight ratio and levels of the inflammatory cytokines TNF-α, IL-6 and IL-1β in the lung tissue were significantly decreased in mice whose alveolar epithelial cells overexpressed PKD2. However, the protective effects of PKD2 overexpression against LPS-induced ALI were reversed by 3-MA pretreatment. Our study suggests that overexpression of PKD2 in the epithelium may alleviate LPS-induced ALI by activating autophagy.
Collapse
Affiliation(s)
- Fan Pan
- Department of Anesthesiology, Affiliated Shanghai Sixth People's Hospital, Shanghai Jiao Tong University, Shanghai, 200233, China
| | - Lina Bu
- Department of Anesthesiology, Affiliated Shanghai Sixth People's Hospital, Shanghai Jiao Tong University, Shanghai, 200233, China
| | - Kaixuan Wu
- Department of Anesthesiology, Affiliated Shanghai Sixth People's Hospital, Shanghai Jiao Tong University, Shanghai, 200233, China
| | - Aizhong Wang
- Department of Anesthesiology, Affiliated Shanghai Sixth People's Hospital, Shanghai Jiao Tong University, Shanghai, 200233, China.
| | - Xiaotao Xu
- Department of Anesthesiology, Affiliated Shanghai Sixth People's Hospital, Shanghai Jiao Tong University, Shanghai, 200233, China.
| |
Collapse
|
24
|
Calvani R, Picca A, Rodriguez-Mañas L, Tosato M, Coelho-Júnior HJ, Biancolillo A, Laosa O, Gervasoni J, Primiano A, Santucci L, Giampaoli O, Bourdel-Marchasson I, Regueme SC, Sinclair AJ, Urbani A, Landi F, Gambassi G, Marini F, Marzetti E. Amino Acid Profiles in Older Adults with Frailty: Secondary Analysis from MetaboFrail and BIOSPHERE Studies. Metabolites 2023; 13:metabo13040542. [PMID: 37110200 PMCID: PMC10147014 DOI: 10.3390/metabo13040542] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2023] [Revised: 04/07/2023] [Accepted: 04/09/2023] [Indexed: 04/29/2023] Open
Abstract
An altered amino acid metabolism has been described in frail older adults which may contribute to muscle loss and functional decline associated with frailty. In the present investigation, we compared circulating amino acid profiles of older adults with physical frailty and sarcopenia (PF&S, n = 94), frail/pre-frail older adults with type 2 diabetes mellitus (F-T2DM, n = 66), and robust non-diabetic controls (n = 40). Partial least squares discriminant analysis (PLS-DA) models were built to define the amino acid signatures associated with the different frailty phenotypes. PLS-DA allowed correct classification of participants with 78.2 ± 1.9% accuracy. Older adults with F-T2DM showed an amino acid profile characterized by higher levels of 3-methylhistidine, alanine, arginine, ethanolamine, and glutamic acid. PF&S and control participants were discriminated based on serum concentrations of aminoadipic acid, aspartate, citrulline, cystine, taurine, and tryptophan. These findings suggest that different types of frailty may be characterized by distinct metabolic perturbations. Amino acid profiling may therefore serve as a valuable tool for frailty biomarker discovery.
Collapse
Affiliation(s)
- Riccardo Calvani
- Fondazione Policlinico Universitario "Agostino Gemelli" IRCCS, 00168 Rome, Italy
- Department of Geriatrics and Orthopedics, Università Cattolica del Sacro Cuore, 00168 Rome, Italy
| | - Anna Picca
- Fondazione Policlinico Universitario "Agostino Gemelli" IRCCS, 00168 Rome, Italy
- Department of Medicine and Surgery, LUM University, 70010 Casamassima, Italy
| | - Leocadio Rodriguez-Mañas
- Servicio de Geriatría, Hospital Universitario de Getafe, 28905 Getafe, Spain
- Centro de Investigación Biomédica en Red "Fragilidad y Envejecimiento Saludable" (CIBERFES), Instituto de Salud Carlos III, 28029 Madrid, Spain
| | - Matteo Tosato
- Fondazione Policlinico Universitario "Agostino Gemelli" IRCCS, 00168 Rome, Italy
| | - Hélio José Coelho-Júnior
- Department of Geriatrics and Orthopedics, Università Cattolica del Sacro Cuore, 00168 Rome, Italy
| | - Alessandra Biancolillo
- Department of Physical and Chemical Sciences, Università degli Studi dell'Aquila, 67100 L'Aquila, Italy
| | - Olga Laosa
- Department of Medicine and Surgery, LUM University, 70010 Casamassima, Italy
- Geriatric Research Group, Biomedical Research Foundation at Getafe University Hospital, 28905 Getafe, Spain
| | - Jacopo Gervasoni
- Fondazione Policlinico Universitario "Agostino Gemelli" IRCCS, 00168 Rome, Italy
| | - Aniello Primiano
- Fondazione Policlinico Universitario "Agostino Gemelli" IRCCS, 00168 Rome, Italy
| | - Lavinia Santucci
- Fondazione Policlinico Universitario "Agostino Gemelli" IRCCS, 00168 Rome, Italy
| | - Ottavia Giampaoli
- Department of Chemistry, Sapienza Università di Roma, 00185 Rome, Italy
| | - Isabelle Bourdel-Marchasson
- Clinical Gerontology Department, Bordeaux University Hospital, 33000 Bordeaux, France
- CRMSB, CNRS UMR 5536, Université de Bordeaux, 33000 Bordeaux, France
| | - Sophie C Regueme
- CHU Bordeaux, Pole Gérontologie Clinique, 33000 Bordeaux, France
| | - Alan J Sinclair
- Foundation for Diabetes Research in Older People (fDROP), King's College, London WC2R 2LS, UK
| | - Andrea Urbani
- Fondazione Policlinico Universitario "Agostino Gemelli" IRCCS, 00168 Rome, Italy
- Department of Geriatrics and Orthopedics, Università Cattolica del Sacro Cuore, 00168 Rome, Italy
| | - Francesco Landi
- Fondazione Policlinico Universitario "Agostino Gemelli" IRCCS, 00168 Rome, Italy
- Department of Geriatrics and Orthopedics, Università Cattolica del Sacro Cuore, 00168 Rome, Italy
| | - Giovanni Gambassi
- Fondazione Policlinico Universitario "Agostino Gemelli" IRCCS, 00168 Rome, Italy
- Department of Geriatrics and Orthopedics, Università Cattolica del Sacro Cuore, 00168 Rome, Italy
| | - Federico Marini
- Department of Chemistry, Sapienza Università di Roma, 00185 Rome, Italy
| | - Emanuele Marzetti
- Fondazione Policlinico Universitario "Agostino Gemelli" IRCCS, 00168 Rome, Italy
- Department of Geriatrics and Orthopedics, Università Cattolica del Sacro Cuore, 00168 Rome, Italy
| |
Collapse
|
25
|
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
|
26
|
Al-Sayyar A, Hammad MM, Williams MR, Al-Onaizi M, Abubaker J, Alzaid F. Neurotransmitters in Type 2 Diabetes and the Control of Systemic and Central Energy Balance. Metabolites 2023; 13:384. [PMID: 36984824 PMCID: PMC10058084 DOI: 10.3390/metabo13030384] [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: 01/01/2023] [Revised: 02/10/2023] [Accepted: 02/13/2023] [Indexed: 03/08/2023] Open
Abstract
Efficient signal transduction is important in maintaining the function of the nervous system across tissues. An intact neurotransmission process can regulate energy balance through proper communication between neurons and peripheral organs. This ensures that the right neural circuits are activated in the brain to modulate cellular energy homeostasis and systemic metabolic function. Alterations in neurotransmitters secretion can lead to imbalances in appetite, glucose metabolism, sleep, and thermogenesis. Dysregulation in dietary intake is also associated with disruption in neurotransmission and can trigger the onset of type 2 diabetes (T2D) and obesity. In this review, we highlight the various roles of neurotransmitters in regulating energy balance at the systemic level and in the central nervous system. We also address the link between neurotransmission imbalance and the development of T2D as well as perspectives across the fields of neuroscience and metabolism research.
Collapse
Affiliation(s)
| | | | | | - Mohammed Al-Onaizi
- Dasman Diabetes Institute, Kuwait City 15462, Kuwait
- Department of Anatomy, Faculty of Medicine, Kuwait University, Kuwait City 13110, Kuwait
| | | | - Fawaz Alzaid
- Dasman Diabetes Institute, Kuwait City 15462, Kuwait
- Institut Necker Enfants Malades-INEM, Université Paris Cité, CNRS, INSERM, F-75015 Paris, France
| |
Collapse
|
27
|
Sanches JM, Zhao LN, Salehi A, Wollheim CB, Kaldis P. Pathophysiology of type 2 diabetes and the impact of altered metabolic interorgan crosstalk. FEBS J 2023; 290:620-648. [PMID: 34847289 DOI: 10.1111/febs.16306] [Citation(s) in RCA: 46] [Impact Index Per Article: 23.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2021] [Revised: 10/14/2021] [Accepted: 11/29/2021] [Indexed: 02/06/2023]
Abstract
Diabetes is a complex and multifactorial disease that affects millions of people worldwide, reducing the quality of life significantly, and results in grave consequences for our health care system. In type 2 diabetes (T2D), the lack of β-cell compensatory mechanisms overcoming peripherally developed insulin resistance is a paramount factor leading to disturbed blood glucose levels and lipid metabolism. Impaired β-cell functions and insulin resistance have been studied extensively resulting in a good understanding of these pathways but much less is known about interorgan crosstalk, which we define as signaling between tissues by secreted factors. Besides hormones and organokines, dysregulated blood glucose and long-lasting hyperglycemia in T2D is associated with changes in metabolism with metabolites from different tissues contributing to the development of this disease. Recent data suggest that metabolites, such as lipids including free fatty acids and amino acids, play important roles in the interorgan crosstalk during the development of T2D. In general, metabolic remodeling affects physiological homeostasis and impacts the development of T2D. Hence, we highlight the importance of metabolic interorgan crosstalk in this review to gain enhanced knowledge of the pathophysiology of T2D, which may lead to new therapeutic approaches to treat this disease.
Collapse
Affiliation(s)
| | - Li Na Zhao
- Department of Clinical Sciences, Lund University, Malmö, Sweden
| | - Albert Salehi
- Department of Clinical Sciences, Lund University, Malmö, Sweden
| | - Claes B Wollheim
- Department of Clinical Sciences, Lund University, Malmö, Sweden.,Department of Cell Physiology and Metabolism, University of Geneva, Geneva, Switzerland
| | - Philipp Kaldis
- Department of Clinical Sciences, Lund University, Malmö, Sweden
| |
Collapse
|
28
|
Tremmel DM, Mikat AE, Gupta S, Mitchell SA, Curran AM, Menadue JA, Odorico JS, Sackett SD. Validating expression of beta cell maturation-associated genes in human pancreas development. Front Cell Dev Biol 2023; 11:1103719. [PMID: 36846594 PMCID: PMC9945361 DOI: 10.3389/fcell.2023.1103719] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2022] [Accepted: 01/16/2023] [Indexed: 02/04/2023] Open
Abstract
The identification of genes associated with human pancreatic beta cell maturation could stimulate a better understanding of normal human islet development and function, be informative for improving stem cell-derived islet (SC-islet) differentiation, and facilitate the sorting of more mature beta cells from a pool of differentiated cells. While several candidate factors to mark beta cell maturation have been identified, much of the data supporting these markers come from animal models or differentiated SC-islets. One such marker is Urocortin-3 (UCN3). In this study, we provide evidence that UCN3 is expressed in human fetal islets well before the acquisition of functional maturation. When SC-islets expressing significant levels of UCN3 were generated, the cells did not exhibit glucose-stimulated insulin secretion, indicating that UCN3 expression is not correlated with functional maturation in these cells. We utilized our tissue bank and SC-islet resources to test an array of other candidate maturation-associated genes, and identified CHGB, G6PC2, FAM159B, GLUT1, IAPP and ENTPD3 as markers with expression patterns that correlate developmentally with the onset of functional maturation in human beta cells. We also find that human beta cell expression of ERO1LB, HDAC9, KLF9, and ZNT8 does not change between fetal and adult stages.
Collapse
Affiliation(s)
- Daniel M. Tremmel
- *Correspondence: Daniel M. Tremmel, ; Sara Dutton Sackett, ; Jon S. Odorico,
| | - Anna E. Mikat
- University of Wisconsin-Madison, Department of Surgery, Transplantation Division, Madison, WI, United States
| | - Sakar Gupta
- University of Wisconsin-Madison, Department of Surgery, Transplantation Division, Madison, WI, United States
| | - Samantha A. Mitchell
- University of Wisconsin-Madison, Department of Surgery, Transplantation Division, Madison, WI, United States
| | - Andrew M. Curran
- University of Wisconsin-Madison, Department of Surgery, Transplantation Division, Madison, WI, United States
| | - Jenna A. Menadue
- University of Wisconsin-Madison, Department of Surgery, Transplantation Division, Madison, WI, United States
| | - Jon S. Odorico
- *Correspondence: Daniel M. Tremmel, ; Sara Dutton Sackett, ; Jon S. Odorico,
| | - Sara Dutton Sackett
- *Correspondence: Daniel M. Tremmel, ; Sara Dutton Sackett, ; Jon S. Odorico,
| |
Collapse
|
29
|
Nguyen HD. Combination of Donepezil and Memantine Attenuated Cognitive Impairment Induced by Mixed Endocrine-Disrupting Chemicals: an In Silico Study. Neurotox Res 2022; 40:2072-2088. [PMID: 36367679 DOI: 10.1007/s12640-022-00591-7] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2022] [Revised: 09/06/2022] [Accepted: 10/08/2022] [Indexed: 11/13/2022]
Abstract
Little is known about the effects of endocrine-disrupting chemicals (EDCs) and the combination of memantine and donepezil on the pathogenesis of cognitive impairment. Here, we aimed to identify in silico the molecular mechanisms of the combination of memantine and donepezil that combat cognitive impairment induced by nine common EDCs using GeneMania, AutoDock Vina, Metascape, SwissADME, MIENTURNET, and miRNAsong. We observed that the mixture of memantine and donepezil had therapeutic effects on mixed EDC-induced cognitive impairment via five genes (TNF, ACHE, BAX, IL1B, and CASP3). With ACHE and TNF, donepezil and memantine both had a high docking score, respectively. The predominant connections among five mutual genes were physical interactions (77.6%). The major pathways associated with memantine and donepezil countering cognitive impairment generated by mixed EDCs were discovered to be "AGE-RAGE signaling pathway in diabetic complications," "pro-survival signaling of neuroprotectin D1," and "non-alcoholic fatty liver disease." The miRNAs and transcription factors implicated in memantine and donepezil protecting against mixed EDCs were hsa-miR-128-3p and hsa-miR-34a-5p, NFKB1, NFKB2, IRF8, and E2F4. The sponges' tertiary structure predictions for two major miRNAs were provided. The physicochemical and pharmacokinetic properties of memantine and donepezil highlighted the need for a therapeutic combination of these medications to treat cognitive impairment.
Collapse
Affiliation(s)
- Hai Duc Nguyen
- Department of Pharmacy, College of Pharmacy and Research Institute of Life and Pharmaceutical Sciences, Sunchon National University, Suncheon, 57922, Republic of Korea.
| |
Collapse
|
30
|
Targeting APLN/APJ restores blood-testis barrier and improves spermatogenesis in murine and human diabetic models. Nat Commun 2022; 13:7335. [PMID: 36443325 PMCID: PMC9705293 DOI: 10.1038/s41467-022-34990-3] [Citation(s) in RCA: 25] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2021] [Accepted: 11/14/2022] [Indexed: 11/29/2022] Open
Abstract
Type 2 diabetes mellitus is one of the most prevalent metabolic diseases presenting with systemic pathologies, including reproductive disorders in male diabetic patients. However, the molecular mechanisms that contributing to spermatogenesis dysfunction in diabetic patients have not yet been fully elucidated. Here, we perform STRT-seq to examine the transcriptome of diabetic patients' testes at single-cell resolution including all major cell types of the testis. Intriguingly, whereas spermatogenesis appears largely preserved, the gene expression profiles of Sertoli cells and the blood-testis barrier (BTB) structure are dramatically impaired. Among these deregulate pathways, the Apelin (APLN) peptide/Apelin-receptor (APJ) axis is hyper-activated in diabetic patients' testes. Mechanistically, APLN is produced locally by Sertoli cells upon high glucose treatment, which subsequently suppress the production of carnitine and repress the expression of cell adhesion genes in Sertoli cells. Together, these effects culminate in BTB structural dysfunction. Finally, using the small molecule APLN receptor antagonist, ML221, we show that blocking APLN/APJ significantly ameliorate the BTB damage and, importantly, improve functional spermatogenesis in diabetic db/db mice. We also translate and validate these findings in cultured human testes. Our findings identify the APLN/APJ axis as a promising therapeutic target to improve reproduction capacity in male diabetic patients.
Collapse
|
31
|
Huang X, Xiong D, Deng L, Liu W, Tang S. Prolonged activation of NMDA receptors induces dedifferentiation of islet β cells in mice. ZHONG NAN DA XUE XUE BAO. YI XUE BAN = JOURNAL OF CENTRAL SOUTH UNIVERSITY. MEDICAL SCIENCES 2022; 47:1182-1190. [PMID: 36411701 PMCID: PMC10930331 DOI: 10.11817/j.issn.1672-7347.2022.220128] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Subscribe] [Scholar Register] [Received: 03/09/2022] [Indexed: 06/16/2023]
Abstract
OBJECTIVES The β-cell dedifferentiation is one of the critical mechanisms in diabetic β-cell loss. Long-term activation of N-methyl-D-aspartate (NMDA) receptors plays an essential role in the development of diabetes, but the underlying mechanisms have not been fully elucidated. This study aims to investigate the effect of prolonged activation of NMDA receptors on islet β-cell dedifferentiation. METHODS Male C57BL/6 mice were randomly divided into a normal control group (control group) and an NMDA group. The mice in the NMDA group were intraperitoneally injected with NMDA (8 mg/kg body weight) and those in the control group were injected with the same volume of saline every day for 6 months. At the end of the 6 th month, glucose tolerance and enzyme linked immunosorbent assay (ELISA) were used to detect the function of islets, and pancreatic tissues were taken for immunofluorescence staining to detect the expressions of insulin, glucagon, and proliferating cell nuclear antigen (PCNA). Real-time PCR was used to detect the mRNA expression of pancreatic β cells, α cells, and islet progenitor cell markers.The primary islets were treated with NMDA to observe the effect of NMDA on the dedifferentiation of β cells. The nuclear factor kappa-B (NF-κB) inhibitor BAY 11-7082 was used at the cellular level via detecting insulin secretion and the expression of endocrine cell markers. RESULTS Compared with the control group, the mice in the NMDA group had higher blood glucose levels at each time point after glucose injection, and the area under the glucose tolerance curve was significantly increased ( P <0.05). The serum insulin content and insulin stimulatory index of the mice in the NMDA group were significantly lower than those in the control group at 30 min after glucose injection (both P <0.05). The double immunofluorescence staining for insulin and glucagon showed that the number of insulin-positive β cells in the pancreatic tissues of mice was significantly decreased after intraperitoneal injection of NMDA in mice for 6 months, while the number of glucagon-positive α cells was significantly increased. Real-time PCR results showed that β-cell markers ( Insulin , Pdx1 , Neurod1 , and Mafa ) were significantly down-regulated in mouse pancreatic tissues after intraperitoneal injection of NMDA for 6 months, while pancreatic progenitor cell markers ( Neurog3 , Gata6 , Hnf4a , Notch1, and Hes1 ) were significantly down-regulated; α-cell markers ( Glucagon , Arx , Irx2 , Mafb , Pou6f2 , Fev , Kcnj3, and Sv2b ) were significantly up-regulated. NMDA treatment of mouse primary islets for 48 h cause significant down-regulation of β-cell marker gene expression ( P <0.05 or P <0.01), accompanied by significant up-regulation of pancreatic progenitor cell markers and α-cell markers ( P <0.05, P <0.01 or P <0.001). The NF-κB inhibitor BAY 11-7082 significantly blocked the down-regulation of β-cell marker expression (all P <0.05) and the up-regulation of α-cell and pancreatic progenitor cell marker after NMDA treatment of islets for 48 h ( P <0.05 or P <0.01). CONCLUSIONS Prolonged activation of NMDA receptors induces islet β-cell dedifferentiation via regulating the NF-κB pathway.
Collapse
Affiliation(s)
- Xiaoting Huang
- Xiangya Nursing School, Central South University, Changsha 410013, China.
| | - Dayan Xiong
- Xiangya Nursing School, Central South University, Changsha 410013, China
| | - Lang Deng
- Xiangya Nursing School, Central South University, Changsha 410013, China
| | - Wei Liu
- Xiangya Nursing School, Central South University, Changsha 410013, China.
| | - Siyuan Tang
- Xiangya Nursing School, Central South University, Changsha 410013, China.
| |
Collapse
|
32
|
Neuritin Promotes Bone Marrow-Derived Mesenchymal Stem Cell Migration to Treat Diabetic Peripheral Neuropathy. Mol Neurobiol 2022; 59:6666-6683. [DOI: 10.1007/s12035-022-03002-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2022] [Accepted: 08/16/2022] [Indexed: 10/15/2022]
|
33
|
Pan X, Tao S, Tong N. Potential Therapeutic Targeting Neurotransmitter Receptors in Diabetes. Front Endocrinol (Lausanne) 2022; 13:884549. [PMID: 35669692 PMCID: PMC9163348 DOI: 10.3389/fendo.2022.884549] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/26/2022] [Accepted: 04/19/2022] [Indexed: 12/04/2022] Open
Abstract
Neurotransmitters are signaling molecules secreted by neurons to coordinate communication and proper function among different sections in the central neural system (CNS) by binding with different receptors. Some neurotransmitters as well as their receptors are found in pancreatic islets and are involved in the regulation of glucose homeostasis. Neurotransmitters can act with their receptors in pancreatic islets to stimulate or inhibit the secretion of insulin (β cell), glucagon (α cell) or somatostatin (δ cell). Neurotransmitter receptors are either G-protein coupled receptors or ligand-gated channels, their effects on blood glucose are mainly decided by the number and location of them in islets. Dysfunction of neurotransmitters receptors in islets is involved in the development of β cell dysfunction and type 2 diabetes (T2D).Therapies targeting different transmitter systems have great potential in the prevention and treatment of T2D and other metabolic diseases.
Collapse
Affiliation(s)
- Xiaohui Pan
- Department of Endocrinology and Metabolism, West China Hospital, Sichuan University, Chengdu, China
- Laboratory of Diabetes and Islet Transplantation, Center for Diabetes and Metabolism Research, West China Hospital, Sichuan University, Chengdu, China
| | - Shibing Tao
- Department of Endocrinology and Metabolism, West China Hospital, Sichuan University, Chengdu, China
- Department of Endocrinology, Ziyang First People’s Hospital, Ziyang, China
| | - Nanwei Tong
- Department of Endocrinology and Metabolism, West China Hospital, Sichuan University, Chengdu, China
- Laboratory of Diabetes and Islet Transplantation, Center for Diabetes and Metabolism Research, West China Hospital, Sichuan University, Chengdu, China
| |
Collapse
|
34
|
Liu Y, Luo Z, Liao Z, Wang M, Zhou Y, Luo S, Ding Y, Liu T, Cao C, Yue S. Effects of Excessive Activation of N-methyl-D-aspartic Acid Receptors in Neonatal Cardiac Mitochondrial Dysfunction Induced by Intrauterine Hypoxia. Front Cardiovasc Med 2022; 9:837142. [PMID: 35498024 PMCID: PMC9039344 DOI: 10.3389/fcvm.2022.837142] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2021] [Accepted: 03/04/2022] [Indexed: 11/13/2022] Open
Abstract
Intrauterine hypoxia is a common complication during pregnancy and could increase the risk of cardiovascular disease in offspring. However, the underlying mechanism is controversial. Memantine, an NMDA receptor antagonist, is reported to be a potential cardio-protective agent. We hypothesized that antenatal memantine treatment could prevent heart injury in neonatal offspring exposed to intrauterine hypoxia. Pregnant rats were exposed to gestational hypoxia or antenatal memantine treatment during late pregnancy. Newborns were then sacrificed to assess multiple parameters. The results revealed that Intrauterine hypoxia resulted in declining birth weight, heart weight, and an abnormally high heart weight/birth weight ratio. Furthermore, intrauterine hypoxia caused mitochondrial structural, functional abnormalities and decreased expression of DRP1, and upregulation of NMDAR1 in vivo. Antenatal memantine treatment,an NMDARs antagonist, improved these changes. In vitro, hypoxia increased the glutamate concentration and expression of NMDAR1. NMDAR activation may lead to similar changes in mitochondrial function, structure, and downregulation of DRP1 in vitro. Pharmacological blockade of NMDARs by the non-competitive NMDA antagonist MK-801 or knockdown of the glutamate receptor NR1 significantly attenuated the increased mitochondrial reactive oxygen species and calcium overload-induced by hypoxia exposure. These facts suggest that memantine could provide a novel and promising treatment for clinical use in intrauterine hypoxia during pregnancy to protect the cardiac mitochondrial function in the offspring. To our best knowledge, our research is the first study that shows intrauterine hypoxia can excessively activate cardiac NMDARs and thus cause mitochondrial dysfunction.
Collapse
Affiliation(s)
- Yang Liu
- Department of Neonatology, Xiangya Hospital, Central South University, Changsha, China
| | - Ziqiang Luo
- Department of Physiology, Xiangya School of Medicine, Central South University, Changsha, China
| | - Zhengchang Liao
- Department of Neonatology, Xiangya Hospital, Central South University, Changsha, China
| | - Mingjie Wang
- Department of Neonatology, Xiangya Hospital, Central South University, Changsha, China
| | - Yan Zhou
- Department of Physiology, Xiangya School of Medicine, Central South University, Changsha, China
| | - Siwei Luo
- Departments of Pediatrics and Neonatology, Children's Hospital of Fudan University, Shanghai, China
- Laboratory of Neonatal Diseases, National Children's Medical Center, National Commission of Health, Shanghai, China
| | - Ying Ding
- Department of Neonatology, Xiangya Hospital, Central South University, Changsha, China
| | - Teng Liu
- Department of Neonatology, Xiangya Hospital, Central South University, Changsha, China
| | - Chuangding Cao
- Department of Neonatology, Xiangya Hospital, Central South University, Changsha, China
| | - Shaojie Yue
- Department of Neonatology, Xiangya Hospital, Central South University, Changsha, China
- *Correspondence: Shaojie Yue
| |
Collapse
|
35
|
Noghani AE, Asadpour R, Saberivand A, Mazaheri Z, Hamidian G. Effect of NMDA receptor agonist and antagonist on spermatogonial stem cells proliferation in 2- and 3- dimensional culture systems. Mol Biol Rep 2022; 49:2197-2207. [PMID: 35000063 DOI: 10.1007/s11033-021-07041-1] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2021] [Accepted: 12/01/2021] [Indexed: 12/25/2022]
Abstract
BACKGROUND The main purpose of this study was to investigate the effect of D-serine (DS) and Dizocilpine (MK-801) on the proliferation of spermatogonial stem cells (SSCs) in two-dimensional (2D) and three-dimensional (3D) culture systems. METHODS AND RESULTS The SSCs of male NMRI mice were isolated by enzymatic digestion and cultured for two weeks. Then, the identity of SSCs was validated by anti-Plzf and anti-GFR-α1 antibodies via immunocytochemistry (ICC). The proliferation capacity of SSCs was evaluated by their culture on a layer of the decellularized testicular matrix (DTM) prepared from mouse testis, as well as two-dimensional (2D) with different mediums. After two weeks of the initiation of proliferation culture on 3D and 2D medium, the pre-meiotic at the mRNA and protein levels were evaluated via qRT-PCR and flow cytometry methods, respectively. The results showed that the proliferation rate of SSCs in 3D culture with 50 mM glutamic acid and 20 mM D-serine was significantly different from other groups after 14 days treatment. mRNA expression levels of promyelocytic leukemia zinc finger (Plzf) in 3D cultures supplemented by 20 mM D-serine and 50 mM glutamic acid were considerably higher than the 3D control group (p < 0.001). The flow cytometry analysis revealed that the amount of Plzf in the 2D-culture groups of SSCs with 20 mM MK-801 was considerably lower compared to the 2D-culture control group (p < 0.001). CONCLUSIONS This study indicated that decellularized testicular matrix supplemented with D-serine and glutamic acid could be considered a promising vehicle to support cells and provide an appropriate niche for the proliferation of SSCs.
Collapse
Affiliation(s)
| | - Reza Asadpour
- Department of Clinical Science, Faculty of Veterinary Medicine, University of Tabriz, Tabriz, Iran.
| | - Adel Saberivand
- Department of Clinical Science, Faculty of Veterinary Medicine, University of Tabriz, Tabriz, Iran
| | - Zohreh Mazaheri
- Basic Medical Science Research Center, Histogenotech Company, Tehran, Iran
| | - Gholamreza Hamidian
- Department of Basic Sciences, Faculty of Veterinary Medicine, University of Tabriz, Tabriz, Iran
| |
Collapse
|
36
|
Durán AM, Beeson WL, Firek A, Cordero-MacIntyre Z, De León M. Dietary Omega-3 Polyunsaturated Fatty-Acid Supplementation Upregulates Protective Cellular Pathways in Patients with Type 2 Diabetes Exhibiting Improvement in Painful Diabetic Neuropathy. Nutrients 2022; 14:nu14040761. [PMID: 35215418 PMCID: PMC8876723 DOI: 10.3390/nu14040761] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2021] [Revised: 02/04/2022] [Accepted: 02/08/2022] [Indexed: 12/12/2022] Open
Abstract
Background: Omega-3 polyunsaturated fatty acids (PUFAs) have been proposed to improve chronic neuroinflammatory diseases in peripheral and central nervous systems. For instance, docosahexaenoic acid (DHA) protects nerve cells from noxious stimuli in vitro and in vivo. Recent reports link PUFA supplementation to improving painful diabetic neuropathy (pDN) symptoms, but cellular mechanisms responsible for this therapeutic effect are not well understood. The objective of this study is to identify distinct cellular pathways elicited by dietary omega-3 PUFA supplementation in patients with type 2 diabetes mellitus (T2DM) affected by pDN. Methods: Forty volunteers diagnosed with type 2 diabetes were enrolled in the “En Balance-PLUS” diabetes education study. The volunteers participated in weekly lifestyle/nutrition education and daily supplementation with 1000 mg DHA and 200 mg eicosapentaenoic acid. The Short-Form McGill Pain Questionnaire validated clinical determination of baseline and post-intervention pain complaints. Laboratory and untargeted metabolomics analyses were conducted using blood plasma collected at baseline and after three months of participation in the dietary regimen. The metabolomics data were analyzed using random forest, hierarchical clustering, ingenuity pathway analysis, and metabolic pathway mapping. Results: The data show that metabolites involved in oxidative stress and glutathione production shifted significantly to a more anti-inflammatory state post supplementation. Example of these metabolites include cystathionine (+90%), S-methylmethionine (+9%), glycine cysteine-glutathione disulfide (+157%) cysteinylglycine (+19%), glutamate (−11%), glycine (+11%), and arginine (+13.4%). In addition, the levels of phospholipids associated with improved membrane fluidity such as linoleoyl-docosahexaenoyl-glycerol (18:2/22:6) (+253%) were significantly increased. Ingenuity pathway analysis suggested several key bio functions associated with omega-3 PUFA supplementation such as formation of reactive oxygen species (p = 4.38 × 10−4, z-score = −1.96), peroxidation of lipids (p = 2.24 × 10−5, z-score = −1.944), Ca2+ transport (p = 1.55 × 10−4, z-score = −1.969), excitation of neurons (p = 1.07 ×10−4, z-score = −1.091), and concentration of glutathione (p = 3.06 × 10−4, z-score = 1.974). Conclusion: The reduction of pro-inflammatory and oxidative stress pathways following dietary omega-3 PUFA supplementation is consistent with the promising role of these fatty acids in reducing adverse symptoms associated with neuroinflammatory diseases and painful neuropathy.
Collapse
Affiliation(s)
- Alfonso M. Durán
- Center for Health Disparities and Molecular Medicine, Department of Basic Sciences, Loma Linda University School of Medicine, Loma Linda, CA 92350, USA; (A.M.D.); (W.L.B.); (Z.C.-M.)
| | - W. Lawrence Beeson
- Center for Health Disparities and Molecular Medicine, Department of Basic Sciences, Loma Linda University School of Medicine, Loma Linda, CA 92350, USA; (A.M.D.); (W.L.B.); (Z.C.-M.)
- Center for Nutrition, Healthy Lifestyle and Disease Prevention, School of Public Health, Loma Linda University, Loma Linda, CA 92350, USA
| | - Anthony Firek
- Comparative Effectiveness and Clinical Outcomes Research Center, Riverside University Health System Medical Center, Moreno Valley, CA 92555, USA;
| | - Zaida Cordero-MacIntyre
- Center for Health Disparities and Molecular Medicine, Department of Basic Sciences, Loma Linda University School of Medicine, Loma Linda, CA 92350, USA; (A.M.D.); (W.L.B.); (Z.C.-M.)
- Center for Nutrition, Healthy Lifestyle and Disease Prevention, School of Public Health, Loma Linda University, Loma Linda, CA 92350, USA
| | - Marino De León
- Center for Health Disparities and Molecular Medicine, Department of Basic Sciences, Loma Linda University School of Medicine, Loma Linda, CA 92350, USA; (A.M.D.); (W.L.B.); (Z.C.-M.)
- Correspondence: ; Tel.: +1-909-558-9474
| |
Collapse
|
37
|
Lépine G, Fouillet H, Rémond D, Huneau JF, Mariotti F, Polakof S. A Scoping Review: Metabolomics Signatures Associated with Animal and Plant Protein Intake and Their Potential Relation with Cardiometabolic Risk. Adv Nutr 2021; 12:2112-2131. [PMID: 34229350 PMCID: PMC8634484 DOI: 10.1093/advances/nmab073] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2021] [Revised: 04/22/2021] [Accepted: 05/12/2021] [Indexed: 12/11/2022] Open
Abstract
The dietary shift from animal protein (AP) to plant protein (PP) sources is encouraged for both environmental and health reasons. For instance, PPs are associated with lower cardiovascular and diabetes risks compared with APs, although the underlying mechanisms mostly remain unknown. Metabolomics is a valuable tool for globally and mechanistically characterizing the impact of AP and PP intake, given its unique ability to provide integrated signatures and specific biomarkers of metabolic effects through a comprehensive snapshot of metabolic status. This scoping review is aimed at gathering and analyzing the available metabolomics data associated with PP- and AP-rich diets, and discusses the metabolic effects underlying these metabolomics signatures and their potential implication for cardiometabolic health. We selected 24 human studies comparing the urine, plasma, or serum metabolomes associated with diets with contrasted AP and PP intakes. Among the 439 metabolites reported in those studies as able to discriminate AP- and PP-rich diets, 46 were considered to provide a robust level of evidence, according to a scoring system, especially amino acids (AAs) and AA-related products. Branched-chain amino acids, aromatic amino acids (AAAs), glutamate, short-chain acylcarnitines, and trimethylamine-N-oxide, which are known to be related to an increased cardiometabolic risk, were associated with AP-rich diets, whereas glycine (rather related to a reduced risk) was associated with PP-rich diets. Tricarboxylic acid (TCA) cycle intermediates and products from gut microbiota AAA degradation were also often reported, but the direction of their associations differed across studies. Overall, AP- and PP-rich diets result in different metabolomics signatures, with several metabolites being plausible candidates to explain some of their differential associations with cardiometabolic risk. Additional studies specifically focusing on protein type, with rigorous intake control, are needed to better characterize the associated metabolic phenotypes and understand how they could mediate differential AP and PP effects on cardiometabolic risk.
Collapse
Affiliation(s)
- Gaïa Lépine
- Université Clermont Auvergne, INRAE, UMR 1019, Unité Nutrition Humaine, Clermont-Ferrand, France
- Université Paris-Saclay, AgroParisTech, INRAE, UMR PNCA, Paris, France
| | - Hélène Fouillet
- Université Paris-Saclay, AgroParisTech, INRAE, UMR PNCA, Paris, France
| | - Didier Rémond
- Université Clermont Auvergne, INRAE, UMR 1019, Unité Nutrition Humaine, Clermont-Ferrand, France
| | | | - François Mariotti
- Université Paris-Saclay, AgroParisTech, INRAE, UMR PNCA, Paris, France
| | - Sergio Polakof
- Université Clermont Auvergne, INRAE, UMR 1019, Unité Nutrition Humaine, Clermont-Ferrand, France
| |
Collapse
|
38
|
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
|
39
|
Transcriptomic signatures of treatment response to the combination of escitalopram and memantine or placebo in late-life depression. Mol Psychiatry 2021; 26:5171-5179. [PMID: 32382137 PMCID: PMC9922535 DOI: 10.1038/s41380-020-0752-2] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/16/2019] [Revised: 04/17/2020] [Accepted: 04/22/2020] [Indexed: 12/17/2022]
Abstract
Drugs that target glutamate neuronal transmission, such as memantine, offer a novel approach to the treatment of late-life depression, which is frequently comorbid with cognitive impairment. The results of our recently published double-blind, randomized, placebo-controlled trial of escitalopram or escitalopram/memantine in late-life depression with subjective memory complaints (NCT01902004) indicated no differences between treatments in depression remission, but additional benefits in cognition at 12-month follow-up with combination treatment. To identify pathways and biological functions uniquely induced by combination treatment that may explain cognitive improvements, we generated transcriptional profiles of remission compared with non-remission from whole blood samples. Remitters to escitalopram compared with escitalopram/memantine combination treatment display unique patterns of gene expression at baseline and 6 months after treatment initiation. Functional enrichment analysis demonstrates that escitalopram-based remission associates to functions related to cellular proliferation, apoptosis, and inflammatory response. Escitalopram/memantine-based remission, however, is characterized by processes related to cellular clearance, metabolism, and cytoskeletal dynamics. Both treatments modulate inflammatory responses, albeit via different effector pathways. Additional research is needed to understand the implications of these results in explaining the observed superior effects of combination treatment on cognition observed with prolonged treatment.
Collapse
|
40
|
Watkins JC, Evans RH, Bayés À, Booker SA, Gibb A, Mabb AM, Mayer M, Mellor JR, Molnár E, Niu L, Ortega A, Pankratov Y, Ramos-Vicente D, Rodríguez-Campuzano A, Rodríguez-Moreno A, Wang LY, Wang YT, Wollmuth L, Wyllie DJA, Zhuo M, Frenguelli BG. 21st century excitatory amino acid research: A Q & A with Jeff Watkins and Dick Evans. Neuropharmacology 2021; 198:108743. [PMID: 34363811 DOI: 10.1016/j.neuropharm.2021.108743] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
In 1981 Jeff Watkins and Dick Evans wrote what was to become a seminal review on excitatory amino acids (EAAs) and their receptors (Watkins and Evans, 1981). Bringing together various lines of evidence dating back over several decades on: the distribution in the nervous system of putative amino acid neurotransmitters; enzymes involved in their production and metabolism; the uptake and release of amino acids; binding of EAAs to membranes; the pharmacological action of endogenous excitatory amino acids and their synthetic analogues, and notably the actions of antagonists for the excitations caused by both nerve stimulation and exogenous agonists, often using pharmacological tools developed by Jeff and his colleagues, they provided a compelling account for EAAs, especially l-glutamate, as a bona fide neurotransmitter in the nervous system. The rest, as they say, is history, but far from being consigned to history, EAA research is in rude health well into the 21st Century as this series of Special Issues of Neuropharmacology exemplifies. With EAAs and their receptors flourishing across a wide range of disciplines and clinical conditions, we enter into a dialogue with two of the most prominent and influential figures in the early days of EAA research: Jeff Watkins and Dick Evans.
Collapse
Affiliation(s)
| | | | - Àlex Bayés
- Molecular Physiology of the Synapse Laboratory, Biomedical Research Institute Sant Pau, Barcelona, Spain and Universitat Autònoma de Barcelona, Barcelona, Spain
| | - Sam A Booker
- Simons Initiative for the Developing Brain, Centre for Discovery Brain Sciences, University of Edinburgh, Edinburgh, EH8 9XD, UK
| | - Alasdair Gibb
- Research Department of Neuroscience, Physiology & Pharmacology, University College London, Gower Street, London, WC1E 6BT, UK
| | - Angela M Mabb
- Neuroscience Institute, Georgia State University, Atlanta, GA, United States
| | - Mark Mayer
- Bldg 35A, Room 3D-904, 35A Convent Drive, NINDS, NIH, Bethesda, MD, 20892, USA
| | - Jack R Mellor
- School of Physiology, Pharmacology and Neuroscience, University of Bristol, Biomedical Sciences Building, University Walk, Bristol, BS8 1TD, UK
| | - Elek Molnár
- School of Physiology, Pharmacology and Neuroscience, University of Bristol, Biomedical Sciences Building, University Walk, Bristol, BS8 1TD, UK
| | - Li Niu
- Chemistry Department, University at Albany, SUNY, 1400 Washington Ave, Albany, NY, 12222, USA
| | - Arturo Ortega
- Department of Toxicology, Cinvestav, Mexico City, Mexico
| | - Yuriy Pankratov
- School of Life Sciences, University of Warwick, Coventry, CV4 7AL, UK
| | - David Ramos-Vicente
- Molecular Physiology of the Synapse Laboratory, Biomedical Research Institute Sant Pau, Barcelona, Spain and Universitat Autònoma de Barcelona, Barcelona, Spain
| | | | | | - Lu-Yang Wang
- Program in Neurosciences & Mental Health, SickKids Research Institute and Department of Physiology, University of Toronto, 555 University Ave, Toronto, Ontario, M5G 1X8, Canada
| | - Yu Tian Wang
- Department of Medicine, University of British Columbia, Vancouver, BC, V6T 2B5, Canada
| | - Lonnie Wollmuth
- Depts. of Neurobiology & Behavior and Biochemistry & Cell Biology, Center for Nervous System Disorders, Stony Brook University, Stony Brook, NY, 11794-5230, USA
| | - David J A Wyllie
- Simons Initiative for the Developing Brain, Centre for Discovery Brain Sciences, University of Edinburgh, Edinburgh, EH8 9XD, UK
| | - Min Zhuo
- Institute of Brain Research, Qingdao International Academician Park, Qingdao, 266000, China
| | | |
Collapse
|
41
|
Šterk M, Križančić Bombek L, Skelin Klemen M, Slak Rupnik M, Marhl M, Stožer A, Gosak M. NMDA receptor inhibition increases, synchronizes, and stabilizes the collective pancreatic beta cell activity: Insights through multilayer network analysis. PLoS Comput Biol 2021; 17:e1009002. [PMID: 33974632 PMCID: PMC8139480 DOI: 10.1371/journal.pcbi.1009002] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2020] [Revised: 05/21/2021] [Accepted: 04/26/2021] [Indexed: 12/15/2022] Open
Abstract
NMDA receptors promote repolarization in pancreatic beta cells and thereby reduce glucose-stimulated insulin secretion. Therefore, NMDA receptors are a potential therapeutic target for diabetes. While the mechanism of NMDA receptor inhibition in beta cells is rather well understood at the molecular level, its possible effects on the collective cellular activity have not been addressed to date, even though proper insulin secretion patterns result from well-synchronized beta cell behavior. The latter is enabled by strong intercellular connectivity, which governs propagating calcium waves across the islets and makes the heterogeneous beta cell population work in synchrony. Since a disrupted collective activity is an important and possibly early contributor to impaired insulin secretion and glucose intolerance, it is of utmost importance to understand possible effects of NMDA receptor inhibition on beta cell functional connectivity. To address this issue, we combined confocal functional multicellular calcium imaging in mouse tissue slices with network science approaches. Our results revealed that NMDA receptor inhibition increases, synchronizes, and stabilizes beta cell activity without affecting the velocity or size of calcium waves. To explore intercellular interactions more precisely, we made use of the multilayer network formalism by regarding each calcium wave as an individual network layer, with weighted directed connections portraying the intercellular propagation. NMDA receptor inhibition stabilized both the role of wave initiators and the course of waves. The findings obtained with the experimental antagonist of NMDA receptors, MK-801, were additionally validated with dextrorphan, the active metabolite of the approved drug dextromethorphan, as well as with experiments on NMDA receptor KO mice. In sum, our results provide additional and new evidence for a possible role of NMDA receptor inhibition in treatment of type 2 diabetes and introduce the multilayer network paradigm as a general strategy to examine effects of drugs on connectivity in multicellular systems.
Collapse
Affiliation(s)
- Marko Šterk
- Faculty of Medicine, University of Maribor, Maribor, Slovenia
- Faculty of Natural Sciences and Mathematics, University of Maribor, Maribor, Slovenia
| | | | | | - Marjan Slak Rupnik
- Faculty of Medicine, University of Maribor, Maribor, Slovenia
- Center for Physiology and Pharmacology, Medical University of Vienna, Vienna, Austria
- Alma Mater Europaea–ECM, Maribor, Slovenia
| | - Marko Marhl
- Faculty of Medicine, University of Maribor, Maribor, Slovenia
- Faculty of Natural Sciences and Mathematics, University of Maribor, Maribor, Slovenia
- Faculty of Education, University of Maribor, Maribor, Slovenia
| | - Andraž Stožer
- Faculty of Medicine, University of Maribor, Maribor, Slovenia
| | - Marko Gosak
- Faculty of Medicine, University of Maribor, Maribor, Slovenia
- Faculty of Natural Sciences and Mathematics, University of Maribor, Maribor, Slovenia
| |
Collapse
|
42
|
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
|
43
|
Alomar SY, Gheit REAE, Enan ET, El-Bayoumi KS, Shoaeir MZ, Elkazaz AY, Al Thagfan SS, Zaitone SA, El-Sayed RM. Novel Mechanism for Memantine in Attenuating Diabetic Neuropathic Pain in Mice via Downregulating the Spinal HMGB1/TRL4/NF-kB Inflammatory Axis. Pharmaceuticals (Basel) 2021; 14:ph14040307. [PMID: 33915770 PMCID: PMC8065430 DOI: 10.3390/ph14040307] [Citation(s) in RCA: 29] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2021] [Revised: 03/05/2021] [Accepted: 03/07/2021] [Indexed: 01/22/2023] Open
Abstract
Diabetic neuropathic pain (DNP) is a common diabetic complication that currently lacks an efficient therapy. The aim of the current work was to uncover the anti-allodynic and neuroprotective effects of memantine in a model of mouse diabetic neuropathy and its ameliorative effect on the high-mobility group box-1 (HMGB1)/toll-like receptor 4 (TLR4)/nuclear factor-k B (NF-kB) inflammatory axis. Diabetes was prompted by an alloxan injection (180 mg/kg) to albino mice. On the ninth week after diabetes induction, DNP was confirmed. Diabetic mice were randomly allocated to two groups (six mice each); a diabetes mellitus (DM) group and DM+memantine group (10 mg/kg, daily) for five weeks. DNP-related behaviors were assessed in terms of thermal hyperalgesia and mechanical allodynia by hot-plate and von Frey filaments. Enzyme-linked immunosorbent assay (ELISA) kits were used to measure the spinal glutamate, interleukin-1 beta (IL-1β), and tumor necrosis factor-α (TNF-α). The spinal levels of N-methyl-D-aspartate type 1 receptor (NMDAR1), HMGB1, TLR4, and phosphorylated NF-kB were assessed using Western blotting. Histopathological investigation of the spinal cord and sciatic nerves, together with the spinal cord ultrastructure, was employed for assessment of the neuroprotective effect. Memantine alleviated pain indicators in diabetic mice and suppressed excessive NMDAR1 activation, glutamate, and pro-inflammatory cytokine release in the spinal cord. The current study validated the ability of memantine to combat the HMGB1/TLR4/NF-kB axis and modulate overactive glutamate spinal transmission, corroborating memantine as an appealing therapeutic target in DNP.
Collapse
Affiliation(s)
- Suliman Y. Alomar
- Department of Zoology, College of Science, King Saud University, Riyadh 11495, Saudi Arabia
- Correspondence: (S.Y.A.); (S.A.Z.)
| | - Rehab E. Abo El Gheit
- Department of Physiology, Faculty of Medicine, Tanta University, Tanta 31527, Egypt;
| | - Eman T. Enan
- Department of Pathology, Faculty of Medicine, Mansoura University, Mansoura 35516, Egypt;
| | - Khaled S. El-Bayoumi
- Department of Human Anatomy and Embryology, Faculty of Medicine, Mansoura University, Mansoura 35516, Egypt;
| | - Mohamed Z. Shoaeir
- Department of Rheumatology and Rehabilitation, Al-Azhar Asyut Faculty of Medicine for Men, Assiut 71524, Egypt;
| | - Amany Y. Elkazaz
- Biochemistry and Molecular Biology Department, Faculty of Medicine, Suez Canal University, Ismailia 41522, Egypt;
- Biochemistry and Molecular Biology Department, Faculty of Medicine, Port-Said University, Port Said 42526, Egypt
| | - Sultan S. Al Thagfan
- Department of Clinical and Hospital Pharmacy, College of Pharmacy, Taibah University, Al Madinah Al Munawwarah 41311, Saudi Arabia;
| | - Sawsan A. Zaitone
- Department of Pharmacology & Toxicology, Faculty of Pharmacy, Suez Canal University, Ismailia 41522, Egypt
- Department of Pharmacology & Toxicology, Faculty of Pharmacy, University of Tabuk, Tabuk 71451, Saudi Arabia
- Correspondence: (S.Y.A.); (S.A.Z.)
| | - Rehab M. El-Sayed
- Department of Pharmacology & Toxicology, Faculty of Pharmacy, Sinai University, El-Arish, North Sinai 45511, Egypt;
| |
Collapse
|
44
|
Badrah M, Riad A, Kassem I, Boccuzzi M, Klugar M. Craniofacial pain in COVID-19 patients with diabetes mellitus: Clinical and laboratory description of 21 cases. J Med Virol 2021; 93:2616-2619. [PMID: 33570190 PMCID: PMC8013969 DOI: 10.1002/jmv.26866] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2020] [Revised: 01/25/2021] [Accepted: 02/09/2021] [Indexed: 12/19/2022]
Affiliation(s)
- Mai Badrah
- Department of Internal Medicine, Faculty of Medicine, Alexandria University, Alexandria, Egypt
| | - Abanoub Riad
- Department of Public Health, Faculty of Medicine, Masaryk University, Brno, Czech Republic.,Czech National Centre for Evidence-Based Healthcare and Knowledge Translation (Cochrane Czech Republic, Czech EBHC: JBI Centre of Excellence, Masaryk University GRADE Centre), Institute of Biostatistics and Analyses, Faculty of Medicine, Masaryk University, Brno, Czech Republic
| | - Islam Kassem
- Department of Oral and Maxillofacial Surgery, Faculty of Dentistry, Alexandria University, Alexandria, Egypt
| | | | - Miloslav Klugar
- Department of Public Health, Faculty of Medicine, Masaryk University, Brno, Czech Republic.,Czech National Centre for Evidence-Based Healthcare and Knowledge Translation (Cochrane Czech Republic, Czech EBHC: JBI Centre of Excellence, Masaryk University GRADE Centre), Institute of Biostatistics and Analyses, Faculty of Medicine, Masaryk University, Brno, Czech Republic
| |
Collapse
|
45
|
Chen X, Xiang X, Xie T, Chen Z, Mou Y, Gao Z, Xie X, Song M, Huang H, Gao Z, Chen M. Memantine protects blood-brain barrier integrity and attenuates neurological deficits through inhibiting nitric oxide synthase ser1412 phosphorylation in intracerebral hemorrhage rats: involvement of peroxynitrite-related matrix metalloproteinase-9/NLRP3 inflammasome activation. Neuroreport 2021; 32:228-237. [PMID: 33470757 PMCID: PMC7870044 DOI: 10.1097/wnr.0000000000001577] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2020] [Accepted: 10/05/2020] [Indexed: 01/08/2023]
Abstract
Memantine has demonstrated beneficial effects on several types of brain insults via therapeutic mechanisms mainly related to its activity as a receptor antagonist of N-methyl-d-aspartate. However, the influences of memantine on intracerebral hemorrhage (ICH) remain obscure. This research probed into the neurovascular protective mechanisms of memantine after ICH and its impacts on neuronal nitric oxide synthase (nNOS) ser1412 phosphorylation. ICH model was established by employing intrastriatal collagenase injection in rats. After modeling, rats were then allocated randomly into sham-operated (sham), vehicle-treated (ICH+V), and memantine-administrated (ICH+M) groups. Memantine (20 mg/kg/day) was intraperitoneally administered 30 min after ICH and thenceforth once daily. Rats were dedicated at 0.25, 6, 12, 24 h, 3 and 7 d post-ICH for measurement of corresponding indexes. Behavioral changes, brain edema, levels of nNOS ser1412 phosphorylation, peroxynitrite, matrix metalloproteinase (MMP)-9, NLRP3, IL-1β and numbers of dying neurons, as well as the cellular localization of gelatinolytic activity, were detected among the groups. Memantine improved the neurologic deficits and mitigated brain water content, levels of MMP-9, NLRP3, IL-1β and dying neurons. Additionally, treatment with memantine also reduced nNOS ser1412 phosphorylation and peroxynitrite formation compared with the ICH+V group at 24 h after ICH. In situ zymography simultaneously revealed that gelatinase activity was primarily colocalized with vessel walls and neurons. We concluded that memantine ameliorated blood-brain barrier disruption and neurologic dysfunction in an ICH rat model. The underlying mechanism might involve repression of nNOS ser1412 phosphorylation, as well as peroxynitrite-related MMP-9 and NLRP3 inflammasome activation.
Collapse
Affiliation(s)
- Xiaowei Chen
- Department of Neurosurgery, the First People’s Hospital of Jingmen City
| | - Xu Xiang
- Department of Neurosurgery, Jingmen Clinical Medical School, Hubei Minzu University, Hubei Province
| | - Teng Xie
- Department of Neurosurgery, the First People’s Hospital of Jingmen City
| | - Zhijun Chen
- Department of Neurosurgery, the First People’s Hospital of Jingmen City
| | - Yu Mou
- Department of Neurosurgery, Jingmen Clinical Medical School, Hubei Minzu University, Hubei Province
| | - Zixu Gao
- The Second Clinical Medical College of Nanchang University
| | - Xun Xie
- The Second Clinical Medical College of Nanchang University
| | - Min Song
- The Second Clinical Medical College of Nanchang University
| | - Hui Huang
- Department of Neurosurgery, The Second Affiliated Hospital of Nanchang University, Nanchang, Jiangxi Province, China
| | - Ziyun Gao
- Department of Neurosurgery, The Second Affiliated Hospital of Nanchang University, Nanchang, Jiangxi Province, China
| | - Min Chen
- The Second Clinical Medical College of Nanchang University
| |
Collapse
|
46
|
Xin Y, Zou L, Lang S. 4-Octyl itaconate (4-OI) attenuates lipopolysaccharide-induced acute lung injury by suppressing PI3K/Akt/NF-κB signaling pathways in mice. Exp Ther Med 2021; 21:141. [PMID: 33456508 PMCID: PMC7791918 DOI: 10.3892/etm.2020.9573] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2020] [Accepted: 11/17/2020] [Indexed: 12/12/2022] Open
Abstract
The progression of acute lung injury (ALI) is attributable to inflammation and oxidative stress. The cell-permeable itaconate analog 4-octyl itaconate (4-OI) provides protection against inflammatory responses and oxidative stress. However, whether 4-OI can protect against ALI remains poorly understood. The aim of this study was to explore the protective effects of 4-OI against LPS-induced ALI and the underlying mechanisms using hematoxylin and eosin (H&E) to observe lung morphology, ELISA and reverse transcription-quantitative PCR to measure the levels of IL-1β, TNF-α and IL-6 and western blotting to examine the levels of PI3K, Akt and NF-κB. The present study demonstrates that intraperitoneal administration of 4-OI (25 mg/kg) 2 h before lipopolysaccharide (LPS; 5 mg/kg) intratracheal injection significantly alleviated the lung tissue injury induced by LPS, reducing the production of proinflammatory cytokines and reactive oxygen species (ROS) in vivo. Furthermore, 4-OI and the antioxidant N-acetyl-L-cysteine markedly suppressed PI3K and Akt phosphorylation in LPS-treated RAW264.7 macrophage cells in vitro. Further study demonstrated that a pharmacological inhibitor of the phosphoinositide 3-kinase (PI3K)-Akt pathway, LY294002, inhibited the expression of NF-κB p65 in the nuclear fraction and decreased the production of inflammatory cytokines. Collectively, the experimental results of the present study provide evidence that 4-OI significantly decreased LPS-induced lung inflammation by suppressing ROS-mediated PI3K/Akt/NF-κB signaling pathways. These results suggest that 4-OI could be a valuable therapeutic drug in the treatment of ALI.
Collapse
Affiliation(s)
- Yan Xin
- Department of Anesthesiology, Changchun Maternity Hospital, Changchun, Jilin 130042, P.R. China
| | - Lili Zou
- Department of Anesthesiology, General Hospital of Ning Xia Medical University, Yin Chuan, Ningxia 750004, P.R. China
| | - Shuhui Lang
- Department of Anesthesiology, General Hospital of Ning Xia Medical University, Yin Chuan, Ningxia 750004, P.R. China
| |
Collapse
|
47
|
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
|
48
|
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
|
49
|
Lizarbe B, Campillo B, Guadilla I, López-Larrubia P, Cerdán S. Magnetic resonance assessment of the cerebral alterations associated with obesity development. J Cereb Blood Flow Metab 2020; 40:2135-2151. [PMID: 32703110 PMCID: PMC7585928 DOI: 10.1177/0271678x20941263] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/05/2020] [Revised: 05/28/2020] [Accepted: 06/14/2020] [Indexed: 12/15/2022]
Abstract
Obesity is a current threat to health care systems, affecting approximately 13% of the world's adult population, and over 18% children and adolescents. The rise of obesity is fuelled by inadequate life style habits, as consumption of diets rich in fats and sugars which promote, additionally, the development of associated comorbidities. Obesity results from a neuroendocrine imbalance in the cerebral mechanisms controlling food intake and energy expenditure, including the hypothalamus and the reward and motivational centres. Specifically, high-fat diets are known to trigger an early inflammatory response in the hypothalamus that precedes weight gain, is time-dependent, and eventually extends to the remaining appetite regulating regions in the brain. Multiple magnetic resonance imaging (MRI) and spectroscopy (MRS) methods are currently available to characterize different features of cerebral obesity, including diffusion weighted, T2 and volumetric imaging and 1H and 13C spectroscopic evaluations. In particular, consistent evidences have revealed increased water diffusivity and T2 values, decreased grey matter volumes, and altered metabolic profiles and fluxes, in the brain of animal models and in obese humans. This review provides an integrative interpretation of the physio-pathological processes associated with obesity development in the brain, and the MRI and MRS methods implemented to characterize them.
Collapse
Affiliation(s)
- Blanca Lizarbe
- Instituto de Investigaciones Biomédicas “Alberto Sols” CSIC/UAM, Madrid, Spain
| | - Basilio Campillo
- Instituto de Investigaciones Biomédicas “Alberto Sols” CSIC/UAM, Madrid, Spain
| | - Irene Guadilla
- Instituto de Investigaciones Biomédicas “Alberto Sols” CSIC/UAM, Madrid, Spain
| | | | - Sebastián Cerdán
- Instituto de Investigaciones Biomédicas “Alberto Sols” CSIC/UAM, Madrid, Spain
| |
Collapse
|
50
|
Diboun I, Ramanjaneya M, Majeed Y, Ahmed L, Bashir M, Butler AE, Abou-Samra AB, Atkin SL, Mazloum NA, Elrayess MA. Metabolic profiling of pre-gestational and gestational diabetes mellitus identifies novel predictors of pre-term delivery. J Transl Med 2020; 18:366. [PMID: 32972433 PMCID: PMC7517617 DOI: 10.1186/s12967-020-02531-5] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2020] [Accepted: 09/15/2020] [Indexed: 12/23/2022] Open
Abstract
Background Pregnant women with gestational diabetes mellitus (GDM) or type 2 diabetes mellitus (T2DM) are at increased risks of pre-term labor, hypertension and preeclampsia. In this study, metabolic profiling of blood samples collected from GDM, T2DM and control pregnant women was undertaken to identify potential diagnostic biomarkers in GDM/T2DM and compared to pregnancy outcome. Methods Sixty-seven pregnant women (21 controls, 32 GDM, 14 T2DM) in their second trimester underwent targeted metabolomics of plasma samples using tandem mass spectrometry with the Biocrates MxP® Quant 500 Kit. Linear regression models were used to identify the metabolic signature of GDM and T2DM, followed by generalized linear model (GLMNET) and Receiver Operating Characteristic (ROC) analysis to determine best predictors of GDM, T2DM and pre-term labor. Results The gestational age at delivery was 2 weeks earlier in T2DM compared to GDM and controls and correlated negatively with maternal HbA1C and systolic blood pressure and positively with serum albumin. Linear regression models revealed elevated glutamate and branched chain amino acids in GDM + T2DM group compared to controls. Regression models also revealed association of lower levels of triacylglycerols and diacylglycerols containing oleic and linoleic fatty acids with pre-term delivery. A generalized linear model ROC analyses revealed that that glutamate is the best predictors of GDM compared to controls (area under curve; AUC = 0.81). The model also revealed that phosphatidylcholine diacyl C40:2, arachidonic acid, glycochenodeoxycholic acid, and phosphatidylcholine acyl-alkyl C34:3 are the best predictors of GDM + T2DM compared to controls (AUC = 0.90). The model also revealed that the triacylglycerols C17:2/36:4 and C18:1/34:1 are the best predictors of pre-term delivery (≤ 37 weeks) (AUC = 0.84). Conclusions This study highlights the metabolite alterations in women in their second trimester with diabetes mellitus and identifies predictive indicators of pre-term delivery. Future studies to confirm these associations in other cohorts and investigate their functional relevance and potential utilization for targeted therapies are warranted.
Collapse
Affiliation(s)
| | - Manjunath Ramanjaneya
- Qatar Metabolic Institute, Hamad Medical Corporation, Doha, Qatar.,Translational Research Institute, Hamad Medical Corporation, Doha, Qatar
| | | | - Lina Ahmed
- Weill Cornell Medicine-Qatar, Doha, Qatar
| | - Mohammed Bashir
- Qatar Metabolic Institute, Hamad Medical Corporation, Doha, Qatar
| | - Alexandra E Butler
- Diabetes Research Center (DRC), Qatar Biomedical Research Institute (QBRI), Hamad Bin Khalifa University (HBKU), Qatar Foundation (QF), PO Box 34110, Doha, Qatar
| | | | - Stephen L Atkin
- Royal College of Surgeons in Ireland Bahrain, Adliya, Kingdom of Bahrain
| | | | | |
Collapse
|