1
|
Tao M, Zhang LL, Zhou GH, Wang C, Luo X. Inhibition of metabotropic glutamate receptor-5 alleviates hepatic steatosis by enhancing autophagy via activation of the AMPK signaling pathway. World J Gastroenterol 2025; 31:98852. [PMID: 39991675 PMCID: PMC11755260 DOI: 10.3748/wjg.v31.i7.98852] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/07/2024] [Revised: 12/08/2024] [Accepted: 12/26/2024] [Indexed: 01/20/2025] Open
Abstract
BACKGROUND The global prevalence of metabolic dysfunction-associated steatotic liver disease (MASLD) has continued to increase annually. Recent studies have indicated that inhibition of metabotropic glutamate receptor 5 (mGluR5) may alleviate hepatic steatosis. However, the precise mechanism warrants further exploration. AIM To investigate the potential mechanism by which mGluR5 attenuates hepatocyte steatosis in vitro and in vivo. METHODS Free fatty acids (FFAs)-stimulated HepG2 cells were treated with the mGluR5 antagonist MPEP and the mGluR5 agonist CHPG. Oil Red O staining and a triglyceride assay kit were used to evaluate lipid content. Western blot analysis was conducted to detect the expression of the autophagy-associated proteins p62 and LC3-II, as well as the expression of the key signaling molecules AMPK and ULK1, in the treated cells. To further elucidate the contributions of autophagy and AMPK, we used chloroquine (CQ) to inhibit autophagy and compound C (CC) to inhibit AMPK activity. In parallel, wild-type mice and mGluR5 knockout (KO) mice fed a normal chow diet or a high-fat diet (HFD) were used to evaluate the effect of mGluR5 inhibition in vivo. RESULTS mGluR5 inhibition by MPEP attenuated hepatocellular steatosis and increased LC3-II and p62 protein expression. The autophagy inhibitor CQ reversed the effects of MPEP. In addition, MPEP promoted AMPK and ULK1 expression in HepG2 cells exposed to FFAs. MPEP treatment led to the nuclear translocation of transcription factor EB, which is known to promote p62 expression. This effect was negated by the AMPK inhibitor CC. mGluR5 KO mice presented reduced body weight, improved glucose tolerance and reduced hyperlipidemia when fed a HFD. Additionally, the livers of HFD-fed mGluR5 KO mice presented increases in LC3-II and p62. CONCLUSION Our results suggest that mGluR5 inhibition promoted autophagy and reduced hepatocyte steatosis through activation of the AMPK signaling pathway. These findings reveal a new functional mechanism of mGluR5 as a target in the treatment of MASLD.
Collapse
Affiliation(s)
- Min Tao
- Department of Endocrinology, The Second Affiliated Hospital, Chongqing Medical University, Chongqing 400010, China
| | - Li-Li Zhang
- Department of Endocrinology, The Second Affiliated Hospital, Chongqing Medical University, Chongqing 400010, China
| | - Guang-Hong Zhou
- Department of Endocrinology, The Second Affiliated Hospital, Chongqing Medical University, Chongqing 400010, China
| | - Cong Wang
- Department of Endocrinology, The Second Affiliated Hospital, Chongqing Medical University, Chongqing 400010, China
| | - Xie Luo
- Department of Endocrinology, The Second Affiliated Hospital, Chongqing Medical University, Chongqing 400010, China
| |
Collapse
|
2
|
Fan H, Yan D, Fang X, Xiao L, Liang M, Wu H, Zhu G, Geng D, Liu Q. Low expression of GRM4 is associated with poor prognosis and tumor immune infiltration in glioma. Int J Neurosci 2024; 134:1674-1686. [PMID: 38164693 DOI: 10.1080/00207454.2023.2297646] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2023] [Revised: 12/15/2023] [Accepted: 12/16/2023] [Indexed: 01/03/2024]
Abstract
INTRODUCTION The metabotropic glutamate receptor 4 (mGlu4, GRM4) exhibits significant expression within the central nervous system (CNS) and has been implicated to be correlated with a poor prognosis. OBJECTIVE This study was aimed to elucidate the relationship between the expression profile of GRM4 and the prognosis of glioma patients. METHODS RNA-sequencing datasets from The Cancer Genome Atlas (TCGA), Gene Expression Omnibus (GEO), and China Glioma Genome Atlas (CGGA) repositories were used to evaluate the potential relationship. The value of clinical prognostic about GRM4 was assessed using clinical survival data from CGGA and TCGA. The GEPIA database was used to select genes like GRM4. PPI network was constructed by the database of (STRING), GO and KEGG analyses were performed. TargetScan, TarBase, miRDB, and starBase were used to explore miRNAs that could regulate GRM4 expression. EWAS Data Hub, MethSurv, and MEXPRESS were used for the analysis and relationship between DNA methylation and GRM4 expression and prognosis in glioma. TIMER2.0 and CAMOIP databases were used to assess the association between immune cell infiltration and GRM4. Human GBM cell lines were used to validate the function of GRM4. RESULTS Our study shows that GRM4 is under expressed among gliomas and accompanied by poorer OS. Multivariate analysis showed that low mRNA expression of GRM4 was an independent factor of prognostic for shorter OS in all glioma patients. MiR-1262 affects the malignant phenotype of gliomas through GRM4. Methylation of DNA plays an important role in the instruction of GRM4 expression, the methylation level of GRM4 in glioma tissue is higher in comparison to normal tissue, and the higher methylation level was accompanied with the worse prognosis. Further analysis showed that GRM4 mRNA expression in GBM linked negatively with common lymphoid progenitor, Macrophage M1, Macrophage, and T cell CD4+ Th2, but not with the tumor purity. Overexpression of GRM4 prevents the migration of human GBM cell lines in vitro. CONCLUSION GRM4 may have a substantial impact on the infiltration of immune cells and serve as a valuable prognostic biomarker in gliomas.
Collapse
Affiliation(s)
- Hai Fan
- Department of Neurosurgery, The First Affiliated Hospital of Xinjiang Medical University, Urumqi, China
| | - Dongming Yan
- Engineering Research Center of Tropical Medicine Innovation and Transformation, Ministry of Education, Hainan Medical University, Haikou, China
- Shishou City People's Hospital, Shishou, China
| | - Xingyue Fang
- Engineering Research Center of Tropical Medicine Innovation and Transformation, Ministry of Education, Hainan Medical University, Haikou, China
| | - Liumin Xiao
- Shishou City People's Hospital, Shishou, China
| | - Mengjie Liang
- Department of Clinical Laboratory, the Second Affiliated Hospital of Shihezi University School of Medicine/Hospital of Xinjiang Production and Construction Corps, Shihezi, China
| | - Haolin Wu
- International Center for Aging and Cancer (ICAC), Hainan Medical University, Haikou, China
| | - Guohua Zhu
- Department of Neurosurgery, The First Affiliated Hospital of Xinjiang Medical University, Urumqi, China
| | - Dangmurenjiafu Geng
- Department of Neurosurgery, The First Affiliated Hospital of Xinjiang Medical University, Urumqi, China
| | - Qibing Liu
- Engineering Research Center of Tropical Medicine Innovation and Transformation, Ministry of Education, Hainan Medical University, Haikou, China
- Department of Pharmacy, The First Affiliated Hospital of Hainan Medical University, Haikou, China
| |
Collapse
|
3
|
Mick GJ, McCormick KL. The role of GABA in type 1 diabetes. Front Endocrinol (Lausanne) 2024; 15:1453396. [PMID: 39619323 PMCID: PMC11604429 DOI: 10.3389/fendo.2024.1453396] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/23/2024] [Accepted: 10/22/2024] [Indexed: 12/13/2024] Open
Abstract
Gamma aminobutyric acid (GABA) is synthesized from glutamate by glutamic decarboxylase (GAD). The entero-pancreatic biology of GABA, which is produced by pancreatic islets, GAD-expressing microbiota, enteric immune cells, or ingested through diet, supports an essential physiologic role of GABA in the health and disease. Outside the central nervous system (CNS), GABA is uniquely concentrated in pancreatic β-cells. They express GAD65, which is a type 1 diabetes (T1D) autoantigen. Glutamate constitutes 10% of the amino acids in dietary protein and is preeminently concentrated in human milk. GABA is enriched in many foods, such as tomato and fermented cheese, and is an over-the-counter supplement. Selected microbiota in the midgut have the enzymatic capacity to produce GABA. Intestinal microbiota interact with gut-associated lymphoid tissue to maintain host defenses and immune tolerance, which are implicated in autoimmune disease. Although GABA is a widely known inhibitory neurotransmitter, oral GABA does not cross the blood brain barrier. Three diabetes-related therapeutic actions are ascribed to GABA, namely, increasing pancreatic β-cell content, attenuating excess glucagon and tamping down T-cell immune destruction. These salutary actions have been observed in numerous rodent diabetes models that usually employed high or near-continuous GABA doses. Clinical studies, to date, have identified positive effects of oral GABA on peripheral blood mononuclear cell cytokine release and plasma glucagon. Going forward, it is reassuring that oral GABA therapy has been well-tolerated and devoid of serious adverse effects.
Collapse
Affiliation(s)
- Gail J. Mick
- Department of Pediatrics, University of Alabama at Birmingham, Birmingham, AL, United States
| | | |
Collapse
|
4
|
Li JL, Zhu CH, Tian MM, Liu Y, Ma L, Tao LJ, Zheng P, Yu JQ, Liu N. Negative allosteric modulator of Group Ⅰ mGluRs: Recent advances and therapeutic perspective for neuropathic pain. Neuroscience 2024; 560:406-421. [PMID: 39368605 DOI: 10.1016/j.neuroscience.2024.10.004] [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: 07/13/2024] [Revised: 09/26/2024] [Accepted: 10/01/2024] [Indexed: 10/07/2024]
Abstract
Neuropathic pain (NP) is a widespread public health problem that existing therapeutic treatments cannot manage adequately; therefore, novel treatment strategies are urgently required. G-protein-coupled receptors are important for intracellular signal transduction, and widely participate in physiological and pathological processes, including pain perception. Group I metabotropic glutamate receptors (mGluRs), including mGluR1 and mGluR5, are predominantly implicated in central sensitization, which can lead to hyperalgesia and allodynia. Many orthosteric site antagonists targeting Group I mGluRs have been found to alleviate NP, but their poor efficacy, low selectivity, and numerous side effects limit their development in NP treatment. Here we reviewed the advantages of Group I mGluRs negative allosteric modulators (NAMs) over orthosteric site antagonists based on allosteric modulation mechanism, and the challenges and opportunities of Group I mGluRs NAMs in NP treatment. This article aims to elucidate the advantages and future development potential of Group I mGluRs NAMs in the treatment of NP.
Collapse
Affiliation(s)
- Jia-Ling Li
- School of Pharmacy, Ningxia Medical University, Yinchuan 750000, China
| | - Chun-Hao Zhu
- School of Pharmacy, Ningxia Medical University, Yinchuan 750000, China
| | - Miao-Miao Tian
- School of Pharmacy, Ningxia Medical University, Yinchuan 750000, China
| | - Yue Liu
- School of Pharmacy, Ningxia Medical University, Yinchuan 750000, China
| | - Lin Ma
- School of Pharmacy, Ningxia Medical University, Yinchuan 750000, China
| | - Li-Jun Tao
- Department of Pharmacy, People's Hospital of Ningxia Hui Autonomous Region, Yinchuan 750000, China
| | - Ping Zheng
- School of Pharmacy, Ningxia Medical University, Yinchuan 750000, China.
| | - Jian-Qiang Yu
- School of Pharmacy, Ningxia Medical University, Yinchuan 750000, China.
| | - Ning Liu
- School of Pharmacy, Ningxia Medical University, Yinchuan 750000, China; School of Basic Medical Science, Ningxia Medical University, Yinchuan 750000, China.
| |
Collapse
|
5
|
Niu N, Zhao R, Tian M, Zong W, Hou X, Liu X, Wang L, Wang L, Zhang L. Genomic Variants Associated with Haematological Parameters and T Lymphocyte Subpopulations in a Large White and Min Pig Intercross Population. Animals (Basel) 2024; 14:3140. [PMID: 39518863 PMCID: PMC11545393 DOI: 10.3390/ani14213140] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2024] [Revised: 10/07/2024] [Accepted: 10/29/2024] [Indexed: 11/16/2024] Open
Abstract
The breeding of disease-resistant pigs has consistently been a topic of significant interest and concern within the pig farming industry. The study of pig blood indicators has the potential to confer economic benefits upon the pig farming industry, whilst simultaneously providing valuable insights that can inform the study of human diseases. In this study, an F2 resource population of 489 individuals was generated through the intercrossing of Large White boars and Min pig sows. A total of 17 haematological parameters and T lymphocyte subpopulations were measured, including white blood cell count (WBC), lymphocyte count (LYM), lymphocyte count percentage (LYM%), monocyte count (MID), monocyte count percentage (MID%), neutrophilic granulocyte count (GRN), percentage of neutrophils (GRN%), mean platelet volume (MPV), platelet distribution width (PDW), platelet count (PLT), CD4+/CD8+, CD4+CD8+CD3+, CD4+CD8-CD3+, CD4-CD8+CD3+, CD4-CD8-CD3+, and CD3+. The Illumina PorcineSNP60 Genotyping BeadChip was obtained for all of the F2 animals. Subsequently, a genome-wide association study (GWAS) was conducted using the TASSEL 5.0 software to identify associated variants and candidate genes for the 17 traits. Significant association signals were identified for PCT and PLT on SSC7, with 1 and 11 significant SNP loci, respectively. A single nucleotide polymorphism (SNP) on SSC12 was identified as a significant predictor of the white blood cell (WBC) trait. Significant association signals were detected for the T lymphocyte subpopulations, namely CD4+/CD8+, CD4+CD8+CD3+, CD4+CD8-CD3+, and CD4-CD8+CD3+, with the majority of these signals observed on SSC7. The genes CLIC5, TRIM15, and SLC17A4 were identified as potential candidates for influencing CD4+/CD8+ and CD4-CD8+CD3+. A missense variant, c.2707 G>A, in the SLC17A4 gene has been demonstrated to be significantly associated with the CD4+/CD8+ and CD4-CD8+CD3+ traits. Three missense variants (c.425 A>C, c.500 C>T, and c.733 A>G) have been identified in the TRIM15 gene as being linked to the CD4+/CD8+ trait. Nevertheless, only c.425 A>C has been demonstrated to be significantly associated with CD4-CD8+CD3+. In the CLIC5 gene, one missense variant (c.957 T>C) has been identified as being associated with the CD4+/CD8+ and CD4-CD8+CD3+ traits. Additionally, significant association signals were observed for CD4+CD8+CD3+ and CD4+CD8-CD3+ on SSC2 and 5, respectively. Subsequently, a gene ontology (GO) enrichment analysis was conducted on all genes within the quantitative trait loci (QTL) intervals of platelet count, CD4+/CD8+, and CD4-CD8+CD3+. The MHC class II protein complex binding pathway was identified as the most significant pathway among the three immune traits. These results provide guidance for further research in the field of breeding disease-resistant pigs.
Collapse
Affiliation(s)
- Naiqi Niu
- State Key Laboratory of Animal Biotech Breeding, Institute of Animal Sciences, Chinese Academy of Agricultural Sciences (CAAS), Beijing 100193, China; (N.N.); (R.Z.); (W.Z.); (X.H.); (X.L.); (L.W.); (L.W.)
| | - Runze Zhao
- State Key Laboratory of Animal Biotech Breeding, Institute of Animal Sciences, Chinese Academy of Agricultural Sciences (CAAS), Beijing 100193, China; (N.N.); (R.Z.); (W.Z.); (X.H.); (X.L.); (L.W.); (L.W.)
| | - Ming Tian
- Institute of Animal Husbandry, Heilongjiang Academy of Agricultural Sciences, Harbin 150086, China;
| | - Wencheng Zong
- State Key Laboratory of Animal Biotech Breeding, Institute of Animal Sciences, Chinese Academy of Agricultural Sciences (CAAS), Beijing 100193, China; (N.N.); (R.Z.); (W.Z.); (X.H.); (X.L.); (L.W.); (L.W.)
| | - Xinhua Hou
- State Key Laboratory of Animal Biotech Breeding, Institute of Animal Sciences, Chinese Academy of Agricultural Sciences (CAAS), Beijing 100193, China; (N.N.); (R.Z.); (W.Z.); (X.H.); (X.L.); (L.W.); (L.W.)
| | - Xin Liu
- State Key Laboratory of Animal Biotech Breeding, Institute of Animal Sciences, Chinese Academy of Agricultural Sciences (CAAS), Beijing 100193, China; (N.N.); (R.Z.); (W.Z.); (X.H.); (X.L.); (L.W.); (L.W.)
| | - Ligang Wang
- State Key Laboratory of Animal Biotech Breeding, Institute of Animal Sciences, Chinese Academy of Agricultural Sciences (CAAS), Beijing 100193, China; (N.N.); (R.Z.); (W.Z.); (X.H.); (X.L.); (L.W.); (L.W.)
| | - Lixian Wang
- State Key Laboratory of Animal Biotech Breeding, Institute of Animal Sciences, Chinese Academy of Agricultural Sciences (CAAS), Beijing 100193, China; (N.N.); (R.Z.); (W.Z.); (X.H.); (X.L.); (L.W.); (L.W.)
| | - Longchao Zhang
- State Key Laboratory of Animal Biotech Breeding, Institute of Animal Sciences, Chinese Academy of Agricultural Sciences (CAAS), Beijing 100193, China; (N.N.); (R.Z.); (W.Z.); (X.H.); (X.L.); (L.W.); (L.W.)
| |
Collapse
|
6
|
He K, Zhao Z, Hu X, Li Y. NMDA Receptor Modulation in COVID-19-Associated Acute Respiratory Syndrome in both In Silico and In Vitro Approach. Appl Biochem Biotechnol 2024; 196:5354-5372. [PMID: 38157155 DOI: 10.1007/s12010-023-04813-2] [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] [Accepted: 12/19/2023] [Indexed: 01/03/2024]
Abstract
The normal function of the N-methyl D-aspartate receptors (NMDAR) in human lungs depends on precisely regulated synaptic glutamate levels. Pathophysiology of the lungs is brought on by the changes in homeostasis of glutamate in the synapsis that leads to abnormal NMDAR activity. Severe acute respiratory syndrome (SARS) primarily results in lung infections, particularly lung muscle stiffening, and NMDA receptor potentiation may increase calcium ion influx and support downstream signaling mechanisms. Hence, NMDAR modulators that depend on glutamate levels could be therapeutically useful medications with fewer unintended side effects. A compound called THP (tetrahydropalmatine) that amplifies Ca2+ influx and potentiates NMDA receptors has been identified in the current study. In asthmatic human airway smooth muscle (HASM) cells, THP regulates the NMDA receptor and helps in asthmatic ASM contraction, and the pharmacological stimulation of ASM depends on both brain and respiratory NMDA receptors. Glutamate potency is altered by this substance without any voltage-dependent side effects. Additionally, a GGPP (geranylgeranyl pyrophosphate)-dependent mechanism of THP reduced the production of pro-inflammatory cytokines in ASM. THP is distinctive in terms of its chemical makeup, functioning, and agonist concentration-dependent and allosteric modulatory activity. To treat COVID-19-related SARS, THP, or any future-related compounds will make good drug-like molecule candidates.
Collapse
Affiliation(s)
- Kun He
- Department of Emergency, Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Tongji Shanxi Hospital, Third Hospital of Shanxi Medical University, Taiyuan, 030032, China
- Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Zhiyong Zhao
- Department of Emergency, Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Tongji Shanxi Hospital, Third Hospital of Shanxi Medical University, Taiyuan, 030032, China
- Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Xuan Hu
- Department of Emergency, Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Tongji Shanxi Hospital, Third Hospital of Shanxi Medical University, Taiyuan, 030032, China
- Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Yuan Li
- Department of Respiratory Medicine, Shanxi Province Cancer Hospital, Shanxi Hospital Affiliated to Cancer Hospital, Chinese Academy of Medical Sciences, Cancer Hospital Affiliated to Shanxi Medical University, Taiyuan, 030013, China.
| |
Collapse
|
7
|
Weinstein N, Carlsen J, Schulz S, Stapleton T, Henriksen HH, Travnik E, Johansson PI. A Lifelike guided journey through the pathophysiology of pulmonary hypertension-from measured metabolites to the mechanism of action of drugs. Front Cardiovasc Med 2024; 11:1341145. [PMID: 38845688 PMCID: PMC11153715 DOI: 10.3389/fcvm.2024.1341145] [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: 11/19/2023] [Accepted: 04/12/2024] [Indexed: 06/09/2024] Open
Abstract
Introduction Pulmonary hypertension (PH) is a pathological condition that affects approximately 1% of the population. The prognosis for many patients is poor, even after treatment. Our knowledge about the pathophysiological mechanisms that cause or are involved in the progression of PH is incomplete. Additionally, the mechanism of action of many drugs used to treat pulmonary hypertension, including sotatercept, requires elucidation. Methods Using our graph-powered knowledge mining software Lifelike in combination with a very small patient metabolite data set, we demonstrate how we derive detailed mechanistic hypotheses on the mechanisms of PH pathophysiology and clinical drugs. Results In PH patients, the concentration of hypoxanthine, 12(S)-HETE, glutamic acid, and sphingosine 1 phosphate is significantly higher, while the concentration of L-arginine and L-histidine is lower than in healthy controls. Using the graph-based data analysis, gene ontology, and semantic association capabilities of Lifelike, led us to connect the differentially expressed metabolites with G-protein signaling and SRC. Then, we associated SRC with IL6 signaling. Subsequently, we found associations that connect SRC, and IL6 to activin and BMP signaling. Lastly, we analyzed the mechanisms of action of several existing and novel pharmacological treatments for PH. Lifelike elucidated the interplay between G-protein, IL6, activin, and BMP signaling. Those pathways regulate hallmark pathophysiological processes of PH, including vasoconstriction, endothelial barrier function, cell proliferation, and apoptosis. Discussion The results highlight the importance of SRC, ERK1, AKT, and MLC activity in PH. The molecular pathways affected by existing and novel treatments for PH also converge on these molecules. Importantly, sotatercept affects SRC, ERK1, AKT, and MLC simultaneously. The present study shows the power of mining knowledge graphs using Lifelike's diverse set of data analytics functionalities for developing knowledge-driven hypotheses on PH pathophysiological and drug mechanisms and their interactions. We believe that Lifelike and our presented approach will be valuable for future mechanistic studies of PH, other diseases, and drugs.
Collapse
Affiliation(s)
- Nathan Weinstein
- CAG Center for Endotheliomics, Copenhagen University Hospital, Rigshospitalet, Copenhagen, Denmark
| | - Jørn Carlsen
- CAG Center for Endotheliomics, Copenhagen University Hospital, Rigshospitalet, Copenhagen, Denmark
- Department of Cardiology, Copenhagen University Hospital, Rigshospitalet, Copenhagen, Denmark
| | - Sebastian Schulz
- The Novo Nordisk Foundation Center for Biosustainability, Technical University of Denmark, Kgs. Lyngby, Denmark
| | - Timothy Stapleton
- The Novo Nordisk Foundation Center for Biosustainability, Technical University of Denmark, Kgs. Lyngby, Denmark
| | - Hanne H. Henriksen
- CAG Center for Endotheliomics, Copenhagen University Hospital, Rigshospitalet, Copenhagen, Denmark
- Department of Clinical Immunology, Copenhagen University Hospital, Rigshospitalet, Copenhagen, Denmark
| | - Evelyn Travnik
- The Novo Nordisk Foundation Center for Biosustainability, Technical University of Denmark, Kgs. Lyngby, Denmark
| | - Pär Ingemar Johansson
- CAG Center for Endotheliomics, Copenhagen University Hospital, Rigshospitalet, Copenhagen, Denmark
- Department of Clinical Immunology, Copenhagen University Hospital, Rigshospitalet, Copenhagen, Denmark
| |
Collapse
|
8
|
Sun K, Li Z, Li W, Chi C, Wang M, Xu R, Gao Y, Li B, Sun Y, Liu R. Investigating the anti-atherosclerotic effects and potential mechanism of Dalbergia odorifera in ApoE-deficient mice using network pharmacology combined with metabolomics. J Pharm Biomed Anal 2024; 242:116017. [PMID: 38387125 DOI: 10.1016/j.jpba.2024.116017] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2023] [Revised: 01/14/2024] [Accepted: 02/04/2024] [Indexed: 02/24/2024]
Abstract
Dalbergia odorifera (DO) is a precious rosewood species in Southern Asia, and its heartwood is used in China as an official plant for invigorating blood circulation and eliminating stasis. This study aims to evaluate the efficacy of DO on atherosclerosis (AS), and further explore its active components and potential mechanisms. The apolipoprotein-E (ApoE)-deficient mice fed a high-fat diet were used as model animals, and the pathological changes in mice with or without DO treatment were compared to evaluate the pharmacodynamics of DO on AS. The mechanisms were preliminarily expounded by combining with metabolomics and network pharmacology. Moreover, the bioactive components and targets were assessed by cell experiments and molecular docking, respectively. Our findings suggested that DO significantly modulated blood lipid levels and alleviated intimal hyperplasia in atherosclerotic-lesioned mice, and the mechanisms may involve the regulation of 18 metabolites that changed during the progression of AS, thus affecting 3 major metabolic pathways and 3 major signaling pathways. Moreover, the interactions between 16 compounds with anti-proliferative effect and hub targets in the 3 signaling pathways were verified using molecular docking. Collectively, our findings preliminarily support the therapeutic effect of DO in atherosclerosis, meanwhile explore the active constituents and potential pharmacological mechanisms, which is conducive to its reasonable exploitation and utilization.
Collapse
Affiliation(s)
- Kang Sun
- School of Pharmacy, Key Laboratory of Molecular Pharmacology and Drug Evaluation (Yantai University), Ministry of Education, Collaborative Innovation Center of Advanced Drug Delivery System and Biotech Drugs in Universities of Shandong, Yantai University, Yantai, China
| | - Zongchao Li
- School of Pharmacy, Key Laboratory of Molecular Pharmacology and Drug Evaluation (Yantai University), Ministry of Education, Collaborative Innovation Center of Advanced Drug Delivery System and Biotech Drugs in Universities of Shandong, Yantai University, Yantai, China
| | - Wenjing Li
- School of Pharmacy, Key Laboratory of Molecular Pharmacology and Drug Evaluation (Yantai University), Ministry of Education, Collaborative Innovation Center of Advanced Drug Delivery System and Biotech Drugs in Universities of Shandong, Yantai University, Yantai, China
| | - Chenglin Chi
- School of Pharmacy, Key Laboratory of Molecular Pharmacology and Drug Evaluation (Yantai University), Ministry of Education, Collaborative Innovation Center of Advanced Drug Delivery System and Biotech Drugs in Universities of Shandong, Yantai University, Yantai, China
| | - Minjun Wang
- School of Pharmacy, Key Laboratory of Molecular Pharmacology and Drug Evaluation (Yantai University), Ministry of Education, Collaborative Innovation Center of Advanced Drug Delivery System and Biotech Drugs in Universities of Shandong, Yantai University, Yantai, China
| | - Ruoxuan Xu
- School of Pharmacy, Key Laboratory of Molecular Pharmacology and Drug Evaluation (Yantai University), Ministry of Education, Collaborative Innovation Center of Advanced Drug Delivery System and Biotech Drugs in Universities of Shandong, Yantai University, Yantai, China
| | - Yan Gao
- Shandong International Biotechnology Park Development Co., Ltd, Yantai, China
| | - Bing Li
- Shandong International Biotechnology Park Development Co., Ltd, Yantai, China
| | - Yiying Sun
- Shandong International Biotechnology Park Development Co., Ltd, Yantai, China
| | - Rongxia Liu
- School of Pharmacy, Key Laboratory of Molecular Pharmacology and Drug Evaluation (Yantai University), Ministry of Education, Collaborative Innovation Center of Advanced Drug Delivery System and Biotech Drugs in Universities of Shandong, Yantai University, Yantai, China.
| |
Collapse
|
9
|
Caridi M, Alborghetti M, Pellicelli V, Orlando R, Pontieri FE, Battaglia G, Arcella A. Metabotropic Glutamate Receptors Type 3 and 5 Feature the "NeuroTransmitter-type" of Glioblastoma: A Bioinformatic Approach. Curr Neuropharmacol 2024; 22:1923-1939. [PMID: 38509672 PMCID: PMC11284726 DOI: 10.2174/1570159x22666240320112926] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2023] [Revised: 08/11/2023] [Accepted: 09/01/2023] [Indexed: 03/22/2024] Open
Abstract
BACKGROUND Glioblastoma (GBM) represents an aggressive and common tumor of the central nervous system. The prognosis of GBM is poor, and despite a refined genetic and molecular characterization, pharmacological treatment is largely suboptimal. OBJECTIVE Contribute to defining a therapeutic line in GBM targeting the mGlu3 receptor in line with the principles of precision medicine. METHODS Here, we performed a computational analysis focused on the expression of type 3 and 5 metabotropic glutamate receptor subtypes (mGlu3 and mGlu5, respectively) in high- and low-grade gliomas. RESULTS The analysis allowed the identification of a particular high-grade glioma type, characterized by a high expression level of both receptor subtypes and by other markers of excitatory and inhibitory neurotransmission. This so-called neurotransmitter-GBM (NT-GBM) also shows a distinct immunological, metabolic, and vascularization gene signature. CONCLUSION Our findings might lay the groundwork for a targeted therapy to be specifically applied to this putative novel type of GBM.
Collapse
Affiliation(s)
- Matteo Caridi
- Division of Hematology and Clinical Immunology, Department of Medicine, University of Perugia, Perugia, Italy
| | - Marika Alborghetti
- Department of Neuroscience, Mental Health and Sensory Organs, Sapienza University of Rome, Rome, Italy
| | | | - Rosamaria Orlando
- Department of Physiology and Pharmacology, University Sapienza of Roma, Rome, Italy
- IRCCS Neuromed, Pozzilli, Italy
| | - Francesco Ernesto Pontieri
- Department of Neuroscience, Mental Health and Sensory Organs, Sapienza University of Rome, Rome, Italy
- IRCCS Fondazione Santa Lucia, Rome, Italy
| | - Giuseppe Battaglia
- Department of Physiology and Pharmacology, University Sapienza of Roma, Rome, Italy
- IRCCS Neuromed, Pozzilli, Italy
| | | |
Collapse
|
10
|
Rus A, López-Sánchez JA, Martínez-Martos JM, Ramírez-Expósito MJ, Molina F, Correa-Rodríguez M, Aguilar-Ferrándiz ME. Predictive Ability of Serum Amino Acid Levels to Differentiate Fibromyalgia Patients from Healthy Subjects. Mol Diagn Ther 2024; 28:113-128. [PMID: 37843759 DOI: 10.1007/s40291-023-00677-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/19/2023] [Indexed: 10/17/2023]
Abstract
BACKGROUND Fibromyalgia is a complex illness to diagnose and treat. OBJECTIVES To evaluate a broad range of circulating free amino acid (AA) levels in fibromyalgia patients as well as the ability of the AAs to differentiate fibromyalgia patients from healthy subjects. DESIGN We carried out a case-control study to evaluate AA levels in 62 patients with fibromyalgia and 78 healthy subjects. This study adheres to the STROBE guidelines. METHODS AAs content was assayed by HPLC in serum samples. The predictive value of AA levels in fibromyalgia was determined by receiver operating characteristic (ROC) curve and forward binary logistic regression analyses. RESULTS Fibromyalgia patients showed higher serum levels of aspartic acid, glutamic acid, aminoadipic acid, asparagine, histidine, 3-methyl-histidine, 5-methyl-histidine, glycine, threonine, taurine, tyrosine, valine, methionine, isoleucine, phenylalanine, leucine, ornithine, lysine, branched chain AAs (BCAAs), large neutral AAs, essential AAs (EAAs), non-essential AAs (NEAAs), basic AAs, EAAs/NEAAs ratio, phenylalanine/tyrosine ratio, and global arginine bioavailability ratio than the controls. Serum alanine levels were lower in patients than in controls. According to ROC analysis, most of these AAs may be good markers for differentiating individuals with fibromyalgia from healthy subjects. Results of logistic regression showed that the combination of glutamic acid, histidine, and alanine had the greatest predictive ability to diagnose fibromyalgia. CONCLUSIONS Our results show an imbalance in serum levels of most AAs in patients with fibromyalgia, which suggest a metabolic disturbance. The determination of serum levels of these AAs may aid in the diagnosis of fibromyalgia, in combination with clinical data of the patient.
Collapse
Affiliation(s)
- Alma Rus
- Department of Cell Biology, University of Granada, Avenida de la Fuentenueva, s/n, 18071, Granada, Spain
- Instituto de Investigación Biosanitaria ibs.GRANADA, 18012, Granada, Spain
| | - José Alberto López-Sánchez
- Department of Cell Biology, University of Granada, Avenida de la Fuentenueva, s/n, 18071, Granada, Spain
- Instituto de Investigación Biosanitaria ibs.GRANADA, 18012, Granada, Spain
- Department of Physical Therapy, Faculty of Health Sciences, University of Granada, Avenida de la Ilustración, 60, 18016, Granada, Spain
| | | | | | - Francisco Molina
- Department of Cell Biology, University of Granada, Avenida de la Fuentenueva, s/n, 18071, Granada, Spain
- Department of Physical Therapy, Faculty of Health Sciences, University of Granada, Avenida de la Ilustración, 60, 18016, Granada, Spain
| | - María Correa-Rodríguez
- Department of Nursing, Faculty of Health Sciences, University of Granada, Avenida de la Ilustración, 60, 18016, Granada, Spain.
| | - María Encarnación Aguilar-Ferrándiz
- Department of Cell Biology, University of Granada, Avenida de la Fuentenueva, s/n, 18071, Granada, Spain
- Department of Physical Therapy, Faculty of Health Sciences, University of Granada, Avenida de la Ilustración, 60, 18016, Granada, Spain
| |
Collapse
|
11
|
Lin G, Wang J, Yang YG, Zhang Y, Sun T. Advances in dendritic cell targeting nano-delivery systems for induction of immune tolerance. Front Bioeng Biotechnol 2023; 11:1242126. [PMID: 37877041 PMCID: PMC10593475 DOI: 10.3389/fbioe.2023.1242126] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2023] [Accepted: 09/25/2023] [Indexed: 10/26/2023] Open
Abstract
Dendritic cells (DCs) are the major specialized antigen-presenting cells (APCs), play a key role in initiating the body's immune response, maintain the balance of immunity. DCs can also induce immune tolerance by rendering effector T cells absent and anergy, and promoting the expansion of regulatory T cells. Induction of tolerogenic DCs has been proved to be a promising strategy for the treatment of autoimmune diseases, organ transplantation, and allergic diseases by various laboratory researches and clinical trials. The development of nano-delivery systems has led to advances in situ modulation of the tolerance phenotype of DCs. By changing the material composition, particle size, zeta-potential, and surface modification of nanoparticles, nanoparticles can be used for the therapeutic payloads targeted delivery to DCs, endowing them with great potential in the induction of immune tolerance. This paper reviews how nano-delivery systems can be modulated for targeted delivery to DCs and induce immune tolerance and reviews their potential in the treatment of autoimmune diseases, organ transplantation, and allergic diseases.
Collapse
Affiliation(s)
- Guojiao Lin
- Key Laboratory of Organ Regeneration and Transplantation of Ministry of Education, Institute of Immunology, The First Hospital, Jilin University, Changchun, China
- National-local Joint Engineering Laboratory of Animal Models for Human Diseases, Changchun, China
| | - Jialiang Wang
- Key Laboratory of Organ Regeneration and Transplantation of Ministry of Education, Institute of Immunology, The First Hospital, Jilin University, Changchun, China
- National-local Joint Engineering Laboratory of Animal Models for Human Diseases, Changchun, China
| | - Yong-Guang Yang
- Key Laboratory of Organ Regeneration and Transplantation of Ministry of Education, Institute of Immunology, The First Hospital, Jilin University, Changchun, China
- National-local Joint Engineering Laboratory of Animal Models for Human Diseases, Changchun, China
- International Center of Future Science, Jilin University, Changchun, China
| | - Yuning Zhang
- Key Laboratory of Organ Regeneration and Transplantation of Ministry of Education, Institute of Immunology, The First Hospital, Jilin University, Changchun, China
- National-local Joint Engineering Laboratory of Animal Models for Human Diseases, Changchun, China
| | - Tianmeng Sun
- Key Laboratory of Organ Regeneration and Transplantation of Ministry of Education, Institute of Immunology, The First Hospital, Jilin University, Changchun, China
- National-local Joint Engineering Laboratory of Animal Models for Human Diseases, Changchun, China
- International Center of Future Science, Jilin University, Changchun, China
- State Key Laboratory of Supramolecular Structure and Materials, Jilin University, Changchun, China
| |
Collapse
|
12
|
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
|
13
|
Fang W, Chen S, Jin X, Liu S, Cao X, Liu B. Metabolomics in aging research: aging markers from organs. Front Cell Dev Biol 2023; 11:1198794. [PMID: 37397261 PMCID: PMC10313136 DOI: 10.3389/fcell.2023.1198794] [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: 04/02/2023] [Accepted: 06/02/2023] [Indexed: 07/04/2023] Open
Abstract
Metabolism plays an important role in regulating aging at several levels, and metabolic reprogramming is the main driving force of aging. Due to the different metabolic needs of different tissues, the change trend of metabolites during aging in different organs and the influence of different levels of metabolites on organ function are also different, which makes the relationship between the change of metabolite level and aging more complex. However, not all of these changes lead to aging. The development of metabonomics research has opened a door for people to understand the overall changes in the metabolic level in the aging process of organisms. The omics-based "aging clock" of organisms has been established at the level of gene, protein and epigenetic modifications, but there is still no systematic summary at the level of metabolism. Here, we reviewed the relevant research published in the last decade on aging and organ metabolomic changes, discussed several metabolites with high repetition rate, and explained their role in vivo, hoping to find a group of metabolites that can be used as metabolic markers of aging. This information should provide valuable information for future diagnosis or clinical intervention of aging and age-related diseases.
Collapse
Affiliation(s)
- Weicheng Fang
- State Key Laboratory of Subtropical Silviculture, School of Forestry and Biotechnology, Zhejiang A&F University, Hangzhou, China
| | - Shuxin Chen
- State Key Laboratory of Subtropical Silviculture, School of Forestry and Biotechnology, Zhejiang A&F University, Hangzhou, China
| | - Xuejiao Jin
- State Key Laboratory of Subtropical Silviculture, School of Forestry and Biotechnology, Zhejiang A&F University, Hangzhou, China
| | - Shenkui Liu
- State Key Laboratory of Subtropical Silviculture, School of Forestry and Biotechnology, Zhejiang A&F University, Hangzhou, China
| | - Xiuling Cao
- State Key Laboratory of Subtropical Silviculture, School of Forestry and Biotechnology, Zhejiang A&F University, Hangzhou, China
| | - Beidong Liu
- State Key Laboratory of Subtropical Silviculture, School of Forestry and Biotechnology, Zhejiang A&F University, Hangzhou, China
- Department of Chemistry and Molecular Biology, University of Gothenburg, Gothenburg, Sweden
| |
Collapse
|
14
|
Zeng S, Tao M, Yuan L, Zhang L, Luo X. Inhibition of mGluR5 ameliorates lipid accumulation and inflammation in HepG2 cells. Biochem Biophys Res Commun 2023; 653:1-11. [PMID: 36842305 DOI: 10.1016/j.bbrc.2023.02.033] [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: 01/27/2023] [Revised: 02/08/2023] [Accepted: 02/14/2023] [Indexed: 02/21/2023]
Abstract
Nonalcoholic fatty liver disease (NAFLD) is a common chronic liver disease characterized by ectopic lipid accumulation in hepatocytes. To date, no specific drug has been approved for its treatment. Metabotropic glutamate receptor 5 (mGluR5) has been showed expressed in hepatocytes and related to some liver diseases such as alcoholic steatosis. However, the function of mGluR5 in NAFLD is not clear. This work aims to investigate the effect and potential mechanism of mGluR5 in NAFLD. We found that mGluR5 expression was increased in the livers of HFD-fed mice and in palmitate-treated HepG2 cells. Suppression of mGluR5 by the specific antagonist MPEP could ameliorate palmitate-induced lipid accumulation, whereas the mGluR5 agonist CHPG promoted lipid deposition in the cells. Knockdown of mGluR5 by small interfering RNA further demonstrated that inhibition of mGluR5 could reduce lipid accumulation. Furthermore, our results revealed that mGluR5 regulated lipid metabolism by increasing the gene expression of lipogenesis. Inflammatory factors and phosphorylation levels of NF-κB-p65 and JNK were also tested in treated hepatocytes. mGluR5 promoted the inflammatory reaction and JNK phosphorylation. Inhibition of JNK signaling by JNK-IN-8 rescued CHPG-induced lipogenesis and inflammation. This study showed mGluR5 regulated lipid accumulation and inflammation in palmitic acid-treated HepG2 cells via the JNK signaling pathway. mGluR5 might be a potential drug target for NAFLD.
Collapse
Affiliation(s)
- Shu Zeng
- Centre for Lipid Research & Key Laboratory of Molecular Biology for Infectious Diseases (Ministry of Education), Institute for Viral Hepatitis, Department of Infectious Diseases, The Second Affiliated Hospital, Chongqing Medical University, Chongqing, China; Department of Gastroenterology, Institute of Digestive Diseases of PLA, Cholestatic Liver Diseases Center, and Center for Metabolic Associated Fatty Liver Disease, The First Affiliated Hospital (Southwest Hospital) of Third Military Medical University (Army Medical University), Chongqing, China.
| | - Min Tao
- Department of Endocrinology, The Second Affiliated Hospital, Chongqing Medical University, Chongqing, China.
| | - Lei Yuan
- Department of Endocrinology, The Second Affiliated Hospital, Chongqing Medical University, Chongqing, China.
| | - Lili Zhang
- Department of Endocrinology, The Second Affiliated Hospital, Chongqing Medical University, Chongqing, China.
| | - Xie Luo
- Department of Endocrinology, The Second Affiliated Hospital, Chongqing Medical University, Chongqing, China.
| |
Collapse
|
15
|
Finamor F, Scarabelot VL, Medeiros LF, Stein DJ, da Silva MD, Callai E, Caumo W, de Souza A, Torres ILS. Involvement of GABAergic, glutamatergic, opioidergic, and brain-derived neurotrophic factor systems in the trigeminal neuropathic pain process. Neurosci Lett 2023; 793:136970. [PMID: 36402255 DOI: 10.1016/j.neulet.2022.136970] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2022] [Revised: 11/11/2022] [Accepted: 11/14/2022] [Indexed: 11/18/2022]
Abstract
Trigeminal neuropathic pain (TNP) is an intense pain condition characterized by hyperalgesia and allodynia; however, its neural mechanisms are not completely understood. Its management is complex, and studies that investigate its biochemical mechanisms are important for improving clinical approaches. This study aimed to evaluate the involvement of GABAergic, glutamatergic, and opioidergic systems and brain-derived neurotrophic factor (BDNF) levels in the TNP process in rats. TNP is induced by chronic constriction injury of the infraorbital nerve (CCI-ION). Nociceptive responses were evaluated using the facial von Frey test before and after the administration of GABAergic and opioidergic agonists and glutamatergic antagonists. The rats were divided into vehicle-treated control (C), sham-surgery (SS), and CCI-ION groups, and then subdivided into the vehicle (V)-treated SS-V and CCI-ION-V groups, SS-MK801 and CCI-ION-MK801, treated with the N-methyl-d-aspartate receptor selective antagonist MK801; SS-PB and CCI-ION-PB, treated with phenobarbital; SS-BZD and CCI-ION-BZD, treated with diazepam; SS-MOR and CCI-ION-MOR, treated with morphine. BDNF levels were evaluated in the cerebral cortex, brainstem, trigeminal ganglion, infraorbital branch of the trigeminal nerve, and serum. CCI-ION induced facial mechanical hyperalgesia. Phenobarbital and morphine reversed the hyperalgesia induced by CCI-ION, and the CCI-BZD group had an increased nociceptive threshold until 60 min. CCI-ION-GLU increased the nociceptive threshold at 60 min. Cerebral cortex and brainstem BDNF levels increased in the CCI-ION and SS groups. Only the CCI group presented high levels of BDNF in the trigeminal ganglion. Our data suggest the involvement of GABAergic, glutamatergic, and opioidergic systems and peripheral BDNF in the TNP process.
Collapse
Affiliation(s)
- Fabrício Finamor
- Nucleus of Pain Pharmacology and Neuromodulation. Hospital de Clínicas de Porto Alegre, RS, Brazil
| | - Vanessa Leal Scarabelot
- Nucleus of Pain Pharmacology and Neuromodulation. Hospital de Clínicas de Porto Alegre, RS, Brazil
| | - Liciane Fernandes Medeiros
- Nucleus of Pain Pharmacology and Neuromodulation. Hospital de Clínicas de Porto Alegre, RS, Brazil; Universidade La Salle, Canoas, RS, Brazil
| | - Dirson João Stein
- Nucleus of Pain Pharmacology and Neuromodulation. Hospital de Clínicas de Porto Alegre, RS, Brazil; School of Medicine, Universidade Federal do Rio Grande do Sul, Porto Alegre, RS, Brazil
| | - Morgana Duarte da Silva
- Nucleus of Pain Pharmacology and Neuromodulation. Hospital de Clínicas de Porto Alegre, RS, Brazil
| | - Etiane Callai
- Nucleus of Pain Pharmacology and Neuromodulation. Hospital de Clínicas de Porto Alegre, RS, Brazil
| | - Wolnei Caumo
- Nucleus of Pain Pharmacology and Neuromodulation. Hospital de Clínicas de Porto Alegre, RS, Brazil; School of Medicine, Universidade Federal do Rio Grande do Sul, Porto Alegre, RS, Brazil
| | - Andressa de Souza
- School of Medicine, Universidade Federal do Rio Grande do Sul, Porto Alegre, RS, Brazil
| | - Iraci L S Torres
- Nucleus of Pain Pharmacology and Neuromodulation. Hospital de Clínicas de Porto Alegre, RS, Brazil; School of Medicine, Universidade Federal do Rio Grande do Sul, Porto Alegre, RS, Brazil.
| |
Collapse
|
16
|
Group I Metabotropic Glutamate Receptors Modulate Motility and Enteric Neural Activity in the Mouse Colon. Biomolecules 2023; 13:biom13010139. [PMID: 36671524 PMCID: PMC9856182 DOI: 10.3390/biom13010139] [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: 11/18/2022] [Revised: 01/04/2023] [Accepted: 01/05/2023] [Indexed: 01/12/2023] Open
Abstract
Glutamate is the major excitatory neurotransmitter in the central nervous system, and there is evidence that Group-I metabotropic glutamate receptors (mGlu1 and mGlu5) have established roles in excitatory neurotransmission and synaptic plasticity. While glutamate is abundantly present in the gut, it plays a smaller role in neurotransmission in the enteric nervous system. In this study, we examined the roles of Group-I mGlu receptors in gastrointestinal function. We investigated the expression of Grm1 (mGlu1) and Grm5 (mGlu5) in the mouse myenteric plexus using RNAscope in situ hybridization. Live calcium imaging and motility analysis were performed on ex vivo preparations of the mouse colon. mGlu5 was found to play a role in excitatory enteric neurotransmission, as electrically-evoked calcium transients were sensitive to the mGlu5 antagonist MPEP. However, inhibition of mGlu5 activity did not affect colonic motor complexes (CMCs). Instead, inhibition of mGlu1 using BAY 36-7620 reduced CMC frequency but did not affect enteric neurotransmission. These data highlight complex roles for Group-I mGlu receptors in myenteric neuron activity and colonic function.
Collapse
|
17
|
Mao LM, Mathur N, Shah K, Wang JQ. Roles of metabotropic glutamate receptor 8 in neuropsychiatric and neurological disorders. INTERNATIONAL REVIEW OF NEUROBIOLOGY 2023; 168:349-366. [PMID: 36868634 PMCID: PMC10162486 DOI: 10.1016/bs.irn.2022.10.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Metabotropic glutamate (mGlu) receptors are G protein-coupled receptors. Among eight mGlu subtypes (mGlu1-8), mGlu8 has drawn increasing attention. This subtype is localized to the presynaptic active zone of neurotransmitter release and is among the mGlu subtypes with high affinity for glutamate. As a Gi/o-coupled autoreceptor, mGlu8 inhibits glutamate release to maintain homeostasis of glutamatergic transmission. mGlu8 receptors are expressed in limbic brain regions and play a pivotal role in modulating motivation, emotion, cognition, and motor functions. Emerging evidence emphasizes the increasing clinical relevance of abnormal mGlu8 activity. Studies using mGlu8 selective agents and knockout mice have revealed the linkage of mGlu8 receptors to multiple neuropsychiatric and neurological disorders, including anxiety, epilepsy, Parkinson's disease, drug addiction, and chronic pain. Expression and function of mGlu8 receptors in some limbic structures undergo long-lasting adaptive changes in animal models of these disorders, which may contribute to the remodeling of glutamatergic transmission critical for the pathogenesis and symptomatology of brain illnesses. This review summarizes the current understanding of mGlu8 biology and the possible involvement of the receptor in several common psychiatric and neurological disorders.
Collapse
Affiliation(s)
- Li-Min Mao
- Department of Biomedical Sciences, University of Missouri-Kansas City, School of Medicine, Kansas City, MO, United States
| | - Nirav Mathur
- Department of Anesthesiology, University of Missouri-Kansas City, School of Medicine, Kansas City, MO, United States
| | - Karina Shah
- Department of Biomedical Sciences, University of Missouri-Kansas City, School of Medicine, Kansas City, MO, United States
| | - John Q Wang
- Department of Biomedical Sciences, University of Missouri-Kansas City, School of Medicine, Kansas City, MO, United States; Department of Anesthesiology, University of Missouri-Kansas City, School of Medicine, Kansas City, MO, United States.
| |
Collapse
|
18
|
Zhai J, He X, Sun Y, Wan Z, Ji B, Liu S, Li S, Wang J. In silico binding affinity prediction for metabotropic glutamate receptors using both endpoint free energy methods and a machine learning-based scoring function. Phys Chem Chem Phys 2022; 24:18291-18305. [PMID: 35880533 PMCID: PMC9460939 DOI: 10.1039/d2cp01727j] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
Metabotropic glutamate receptors (mGluRs) play an important role in regulating glutamate signal pathways, which are involved in neuropathy and periphery homeostasis. mGluR4, which belongs to Group III mGluRs, is most widely distributed in the periphery among all the mGluRs. It has been proved that the regulation of this receptor is involved in diabetes, colorectal carcinoma and many other diseases. However, the application of structure-based drug design to identify small molecules to regulate the mGluR4 receptor is limited due to the absence of a resolved mGluR4 protein structure. In this work, we first built a homology model of mGluR4 based on a crystal structure of mGluR8, and then conducted hierarchical virtual screening (HVS) to identify possible active ligands for mGluR4. The HVS protocol consists of three hierarchical filters including Glide docking, molecular dynamic (MD) simulation and binding free energy calculation. We successfully prioritized active ligands of mGluR4 from a set of screening compounds using HVS. The predicted active ligands based on binding affinities can almost cover all the experiment-determined active ligands, with only one ligand missed. The correlation between the measured and predicted binding affinities is significantly improved for the MM-PB/GBSA-WSAS methods compared to the Glide docking method. More importantly, we have identified hotspots for ligand binding, and we found that SER157 and GLY158 tend to contribute to the selectivity of mGluR4 ligands, while ALA154 and ALA155 could account for the ligand selectivity to mGluR8. We also recognized other 5 key residues that are critical for ligand potency. The difference of the binding profiles between mGluR4 and mGluR8 can guide us to develop more potent and selective modulators. Moreover, we evaluated the performance of IPSF, a novel type of scoring function trained by a machine learning algorithm on residue-ligand interaction profiles, in guiding drug lead optimization. The cross-validation root-mean-square errors (RMSEs) are much smaller than those by the endpoint methods, and the correlation coefficients are comparable to the best endpoint methods for both mGluRs. Thus, machine learning-based IPSF can be applied to guide lead optimization, albeit the total number of actives/inactives are not big, a typical scenario in drug discovery projects.
Collapse
Affiliation(s)
- Jingchen Zhai
- Department of Pharmaceutical Sciences and Computational Chemical Genomics Screening Center, School of Pharmacy, University of Pittsburgh, Pittsburgh, PA 15261, USA.
| | - Xibing He
- Department of Pharmaceutical Sciences and Computational Chemical Genomics Screening Center, School of Pharmacy, University of Pittsburgh, Pittsburgh, PA 15261, USA.
| | - Yuchen Sun
- Department of Pharmaceutical Sciences and Computational Chemical Genomics Screening Center, School of Pharmacy, University of Pittsburgh, Pittsburgh, PA 15261, USA.
| | - Zhuoya Wan
- Department of Pharmaceutical Sciences and Center for Pharmacogenetics, School of Pharmacy, University of Pittsburgh, Pittsburgh, PA 15261, USA
| | - Beihong Ji
- Department of Pharmaceutical Sciences and Computational Chemical Genomics Screening Center, School of Pharmacy, University of Pittsburgh, Pittsburgh, PA 15261, USA.
| | - Shuhan Liu
- Department of Pharmaceutical Sciences and Computational Chemical Genomics Screening Center, School of Pharmacy, University of Pittsburgh, Pittsburgh, PA 15261, USA.
| | - Song Li
- Department of Pharmaceutical Sciences and Center for Pharmacogenetics, School of Pharmacy, University of Pittsburgh, Pittsburgh, PA 15261, USA
| | - Junmei Wang
- Department of Pharmaceutical Sciences and Computational Chemical Genomics Screening Center, School of Pharmacy, University of Pittsburgh, Pittsburgh, PA 15261, USA.
| |
Collapse
|
19
|
Luessen DJ, Conn PJ. Allosteric Modulators of Metabotropic Glutamate Receptors as Novel Therapeutics for Neuropsychiatric Disease. Pharmacol Rev 2022; 74:630-661. [PMID: 35710132 PMCID: PMC9553119 DOI: 10.1124/pharmrev.121.000540] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
Abstract
Metabotropic glutamate (mGlu) receptors, a family of G-protein-coupled receptors, have been identified as novel therapeutic targets based on extensive research supporting their diverse contributions to cell signaling and physiology throughout the nervous system and important roles in regulating complex behaviors, such as cognition, reward, and movement. Thus, targeting mGlu receptors may be a promising strategy for the treatment of several brain disorders. Ongoing advances in the discovery of subtype-selective allosteric modulators for mGlu receptors has provided an unprecedented opportunity for highly specific modulation of signaling by individual mGlu receptor subtypes in the brain by targeting sites distinct from orthosteric or endogenous ligand binding sites on mGlu receptors. These pharmacological agents provide the unparalleled opportunity to selectively regulate neuronal excitability, synaptic transmission, and subsequent behavioral output pertinent to many brain disorders. Here, we review preclinical and clinical evidence supporting the utility of mGlu receptor allosteric modulators as novel therapeutic approaches to treat neuropsychiatric diseases, such as schizophrenia, substance use disorders, and stress-related disorders. SIGNIFICANCE STATEMENT: Allosteric modulation of metabotropic glutamate (mGlu) receptors represents a promising therapeutic strategy to normalize dysregulated cellular physiology associated with neuropsychiatric disease. This review summarizes preclinical and clinical studies using mGlu receptor allosteric modulators as experimental tools and potential therapeutic approaches for the treatment of neuropsychiatric diseases, including schizophrenia, stress, and substance use disorders.
Collapse
|
20
|
Špirková A, Kovaříková V, Šefčíková Z, Pisko J, Kšiňanová M, Koppel J, Fabian D, Čikoš Š. Glutamate can act as a signaling molecule in mouse preimplantation embryos. Biol Reprod 2022; 107:916-927. [PMID: 35746896 PMCID: PMC9562114 DOI: 10.1093/biolre/ioac126] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2022] [Revised: 05/31/2022] [Accepted: 06/20/2022] [Indexed: 11/12/2022] Open
Abstract
Free amino acids are present in the natural environment of the preimplantation embryo, and their availability can influence early embryo development. Glutamic acid is one of the amino acids with highest concentrations in female reproductive fluids, and we investigated whether glutamic acid/glutamate can affect preimplantation embryo development by acting through cell membrane receptors. Using RT-PCR, we detected 15 ionotropic glutamate receptor transcripts and 8 metabotropic glutamate receptor transcripts in mouse ovulated oocytes and/or in vivo developed blastocysts. Using immunohistochemistry, we detected expression of two AMPA receptor subunits, three kainate receptor subunits and member 5 metabotropic glutamate receptor protein in blastocysts. Extracellular concentrations of glutamic acid starting at 5 mM impaired mouse blastocyst development, and this fact may be of great practical importance since glutamic acid and its salts (mainly monosodium glutamate) are widely used as food additives. Experiments with glutamate receptor agonists (in combination with gene expression analysis) revealed that specific AMPA receptors (formed from GRIA3 and/or GRIA4 subunits), kainate receptors (formed from GRIK 3 and GRIK 4 or GRIK 5 subunits) and GRM5 glutamate receptor were involved in this effect. The glutamic acid-induced effects were prevented or reduced by pre-treatment of blastocysts with AMPA, kainate and GRM5 receptor antagonists, further confirming the involvement of these receptor types. Our results show that glutamic acid can act as a signaling molecule in preimplantation embryos, exerting its effects through activation of cell membrane receptors.
Collapse
Affiliation(s)
- Alexandra Špirková
- Institute of Animal Physiology, Centre of Biosciences of the Slovak Academy of Sciences, Šoltésovej 4-6, 04001 Košice, Slovakia
| | - Veronika Kovaříková
- Institute of Animal Physiology, Centre of Biosciences of the Slovak Academy of Sciences, Šoltésovej 4-6, 04001 Košice, Slovakia
| | - Zuzana Šefčíková
- Institute of Animal Physiology, Centre of Biosciences of the Slovak Academy of Sciences, Šoltésovej 4-6, 04001 Košice, Slovakia
| | - Jozef Pisko
- Institute of Animal Physiology, Centre of Biosciences of the Slovak Academy of Sciences, Šoltésovej 4-6, 04001 Košice, Slovakia
| | - Martina Kšiňanová
- Institute of Molecular Physiology and Genetics, Centre of Biosciences, Slovak Academy of Sciences, Dúbravská cesta 9, 840 05 Bratislava, Slovakia
| | - Juraj Koppel
- Institute of Animal Physiology, Centre of Biosciences of the Slovak Academy of Sciences, Šoltésovej 4-6, 04001 Košice, Slovakia
| | - Dušan Fabian
- Institute of Animal Physiology, Centre of Biosciences of the Slovak Academy of Sciences, Šoltésovej 4-6, 04001 Košice, Slovakia
| | - Štefan Čikoš
- Institute of Animal Physiology, Centre of Biosciences of the Slovak Academy of Sciences, Šoltésovej 4-6, 04001 Košice, Slovakia
| |
Collapse
|
21
|
Blocking Metabotropic Glutamate Receptor Subtype 7 via the Venus Flytrap Domain Promotes a Chronic Stress-Resilient Phenotype in Mice. Cells 2022; 11:cells11111817. [PMID: 35681512 PMCID: PMC9180111 DOI: 10.3390/cells11111817] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2022] [Revised: 05/30/2022] [Accepted: 05/31/2022] [Indexed: 12/01/2022] Open
Abstract
Chronic psychosocial stress participates prominently in the etiology of various psychiatric conditions and comorbid somatic pathologies; however, suitable pharmacotherapy of these disorders is still of high medical need. During the last few decades, research on mGlu receptors advanced remarkably and much attention was given to the mGlu7 subtype. Here, genetic mGlu7 ablation, short-term pharmacological mGlu7 blockade, as well as siRNA-mediated knockdown of mGlu7 were shown to result in an acute anti-stress, antidepressant- and anxiolytic-like phenotype in mice. Moreover, we recently revealed a prominent stress-protective effect of genetic mGlu7 ablation also with respect to chronic psychosocial stress. In addition, we are able to demonstrate in the present study that the chronic pharmacological blockade of mGlu7 interferes with various chronic stress-induced alterations. For this, we used the chronic subordinate colony housing (CSC), a mouse model of chronic male subordination, in combination with chronic treatment with the mGlu7-selective orthosteric-like antagonist XAP044 (7-hydroxy-3-(4-iodophenoxy)-4H-chromen-4-one). Interestingly, XAP044 dose-dependently ameliorates hypothalamic–pituitary–adrenal axis dysfunctions, thymus atrophy, as well as the CSC-induced increase in innate anxiety. Taken together, our findings provide further evidence for the role of mGlu7 in chronic psychosocial stress-induced alterations and suggests the pharmacological blockade of mGlu7 as a promising therapeutic approach for the treatment of chronic stress-related pathologies in men.
Collapse
|
22
|
Zhang Y, Chu JMT, Wong GTC. Cerebral Glutamate Regulation and Receptor Changes in Perioperative Neuroinflammation and Cognitive Dysfunction. Biomolecules 2022; 12:biom12040597. [PMID: 35454185 PMCID: PMC9029551 DOI: 10.3390/biom12040597] [Citation(s) in RCA: 41] [Impact Index Per Article: 13.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2022] [Revised: 04/11/2022] [Accepted: 04/15/2022] [Indexed: 12/23/2022] Open
Abstract
Glutamate is the major excitatory neurotransmitter in the central nervous system and is intricately linked to learning and memory. Its activity depends on the expression of AMPA and NMDA receptors and excitatory amino transporters on neurons and glial cells. Glutamate transporters prevent the excess accumulation of glutamate in synapses, which can lead to aberrant synaptic signaling, excitotoxicity, or cell death. Neuroinflammation can occur acutely after surgical trauma and contributes to the development of perioperative neurocognitive disorders, which are characterized by impairment in multiple cognitive domains. In this review, we aim to examine how glutamate handling and glutamatergic function are affected by neuroinflammation and their contribution to cognitive impairment. We will first summarize the current data regarding glutamate in neurotransmission, its receptors, and their regulation and trafficking. We will then examine the impact of inflammation on glutamate handling and neurotransmission, focusing on changes in glial cells and the effect of cytokines. Finally, we will discuss these changes in the context of perioperative neuroinflammation and the implications they have for perioperative neurocognitive disorders.
Collapse
|
23
|
Montanari M, Martella G, Bonsi P, Meringolo M. Autism Spectrum Disorder: Focus on Glutamatergic Neurotransmission. Int J Mol Sci 2022; 23:ijms23073861. [PMID: 35409220 PMCID: PMC8998955 DOI: 10.3390/ijms23073861] [Citation(s) in RCA: 52] [Impact Index Per Article: 17.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2022] [Revised: 03/24/2022] [Accepted: 03/29/2022] [Indexed: 12/16/2022] Open
Abstract
Disturbances in the glutamatergic system have been increasingly documented in several neuropsychiatric disorders, including autism spectrum disorder (ASD). Glutamate-centered theories of ASD are based on evidence from patient samples and postmortem studies, as well as from studies documenting abnormalities in glutamatergic gene expression and metabolic pathways, including changes in the gut microbiota glutamate metabolism in patients with ASD. In addition, preclinical studies on animal models have demonstrated glutamatergic neurotransmission deficits and altered expression of glutamate synaptic proteins. At present, there are no approved glutamatergic drugs for ASD, but several ongoing clinical trials are currently focusing on evaluating in autistic patients glutamatergic pharmaceuticals already approved for other conditions. In this review, we provide an overview of the literature concerning the role of glutamatergic neurotransmission in the pathophysiology of ASD and as a potential target for novel treatments.
Collapse
Affiliation(s)
- Martina Montanari
- Laboratory of Neurophysiology and Plasticity, IRCCS Fondazione Santa Lucia, 00179 Rome, Italy; (M.M.); (G.M.)
- Department of Systems Neuroscience, University Tor Vergata, 00133 Rome, Italy
| | - Giuseppina Martella
- Laboratory of Neurophysiology and Plasticity, IRCCS Fondazione Santa Lucia, 00179 Rome, Italy; (M.M.); (G.M.)
| | - Paola Bonsi
- Laboratory of Neurophysiology and Plasticity, IRCCS Fondazione Santa Lucia, 00179 Rome, Italy; (M.M.); (G.M.)
- Correspondence: (P.B.); (M.M.)
| | - Maria Meringolo
- Laboratory of Neurophysiology and Plasticity, IRCCS Fondazione Santa Lucia, 00179 Rome, Italy; (M.M.); (G.M.)
- Correspondence: (P.B.); (M.M.)
| |
Collapse
|
24
|
Wan Z, Sun R, Liu YW, Li S, Sun J, Li J, Zhu J, Moharil P, Zhang B, Ren P, Ren G, Zhang M, Ma X, Dai S, Yang D, Lu B, Li S. Targeting metabotropic glutamate receptor 4 for cancer immunotherapy. SCIENCE ADVANCES 2021; 7:eabj4226. [PMID: 34890233 PMCID: PMC8664261 DOI: 10.1126/sciadv.abj4226] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/12/2021] [Accepted: 10/23/2021] [Indexed: 05/30/2023]
Abstract
In this study, we report a novel role of metabotropic glutamate receptor 4 (GRM4) in suppressing antitumor immunity. We revealed in three murine syngeneic tumor models (B16, MC38, and 3LL) that either genetic knockout (Grm4−/−) or pharmacological inhibition led to significant delay in tumor growth. Mechanistically, perturbation of GRM4 resulted in a strong antitumor immunity by promoting natural killer (NK), CD4+, and CD8+ T cells toward an activated, proliferative, and functional phenotype. Single-cell RNA sequencing and T cell receptor profiling further defined the clonal expansion and immune landscape changes in CD8+ T cells. We further showed that Grm4−/− intrinsically activated interferon-γ production in CD8+ T cells through cyclic adenosine 3′,5′-monophosphate (cAMP)/cAMP response element binding protein–mediated pathway. Our study appears to be of clinical significance as a signature of NKhigh-GRM4low and CD8high-GRM4low correlated with improved survival in patients with melanoma. Targeting GRM4 represents a new approach for cancer immunotherapy.
Collapse
Affiliation(s)
- Zhuoya Wan
- Center for Pharmacogenetics, Department of Pharmaceutical Sciences, School of Pharmacy, University of Pittsburgh, Pittsburgh, PA 15261, USA
| | - Runzi Sun
- Department of Immunology, University of Pittsburgh School of Medicine, Pittsburgh, PA 15261, USA
| | - Yang-Wuyue Liu
- Center for Pharmacogenetics, Department of Pharmaceutical Sciences, School of Pharmacy, University of Pittsburgh, Pittsburgh, PA 15261, USA
| | - Sihan Li
- Center for Pharmacogenetics, Department of Pharmaceutical Sciences, School of Pharmacy, University of Pittsburgh, Pittsburgh, PA 15261, USA
| | - Jingjing Sun
- Center for Pharmacogenetics, Department of Pharmaceutical Sciences, School of Pharmacy, University of Pittsburgh, Pittsburgh, PA 15261, USA
| | - Jiang Li
- Center for Pharmacogenetics, Department of Pharmaceutical Sciences, School of Pharmacy, University of Pittsburgh, Pittsburgh, PA 15261, USA
| | - Junjie Zhu
- Center for Pharmacogenetics, Department of Pharmaceutical Sciences, School of Pharmacy, University of Pittsburgh, Pittsburgh, PA 15261, USA
| | - Pearl Moharil
- Center for Pharmacogenetics, Department of Pharmaceutical Sciences, School of Pharmacy, University of Pittsburgh, Pittsburgh, PA 15261, USA
| | - Bei Zhang
- Center for Pharmacogenetics, Department of Pharmaceutical Sciences, School of Pharmacy, University of Pittsburgh, Pittsburgh, PA 15261, USA
| | - Pengfei Ren
- Center for Pharmacogenetics, Department of Pharmaceutical Sciences, School of Pharmacy, University of Pittsburgh, Pittsburgh, PA 15261, USA
| | - Guolian Ren
- Center for Pharmacogenetics, Department of Pharmaceutical Sciences, School of Pharmacy, University of Pittsburgh, Pittsburgh, PA 15261, USA
| | - Min Zhang
- Center for Pharmacogenetics, Department of Pharmaceutical Sciences, School of Pharmacy, University of Pittsburgh, Pittsburgh, PA 15261, USA
| | - Xiaochao Ma
- Center for Pharmacogenetics, Department of Pharmaceutical Sciences, School of Pharmacy, University of Pittsburgh, Pittsburgh, PA 15261, USA
| | - Shuangshuang Dai
- Center for Pharmacogenetics, Department of Pharmaceutical Sciences, School of Pharmacy, University of Pittsburgh, Pittsburgh, PA 15261, USA
| | - Da Yang
- Center for Pharmacogenetics, Department of Pharmaceutical Sciences, School of Pharmacy, University of Pittsburgh, Pittsburgh, PA 15261, USA
- University of Pittsburgh Cancer Institute, University of Pittsburgh, Pittsburgh, PA 15261, USA
| | - Binfeng Lu
- Department of Immunology, University of Pittsburgh School of Medicine, Pittsburgh, PA 15261, USA
- University of Pittsburgh Cancer Institute, University of Pittsburgh, Pittsburgh, PA 15261, USA
| | - Song Li
- Center for Pharmacogenetics, Department of Pharmaceutical Sciences, School of Pharmacy, University of Pittsburgh, Pittsburgh, PA 15261, USA
- University of Pittsburgh Cancer Institute, University of Pittsburgh, Pittsburgh, PA 15261, USA
| |
Collapse
|
25
|
Li L, Solvi C, Zhang F, Qi Z, Chittka L, Zhao W. Gut microbiome drives individual memory variation in bumblebees. Nat Commun 2021; 12:6588. [PMID: 34824201 PMCID: PMC8616916 DOI: 10.1038/s41467-021-26833-4] [Citation(s) in RCA: 42] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2021] [Accepted: 10/25/2021] [Indexed: 12/12/2022] Open
Abstract
The potential of the gut microbiome as a driver of individual cognitive differences in natural populations of animals remains unexplored. Here, using metagenomic sequencing of individual bumblebee hindguts, we find a positive correlation between the abundance of Lactobacillus Firm-5 cluster and memory retention on a visual discrimination task. Supplementation with the Firm-5 species Lactobacillus apis, but not other non-Firm-5 bacterial species, enhances bees' memory. Untargeted metabolomics after L. apis supplementation show increased LPA (14:0) glycerophospholipid in the haemolymph. Oral administration of the LPA increases long-term memory significantly. Based on our findings and metagenomic/metabolomic analyses, we propose a molecular pathway for this gut-brain interaction. Our results provide insights into proximate and ultimate causes of cognitive differences in natural bumblebee populations.
Collapse
Affiliation(s)
- Li Li
- State Key Laboratory of Food Science and Technology, School of Food Science and Technology, Jiangnan University, Wuxi, Jiangsu, 214122, China
| | - Cwyn Solvi
- Ecology and Genetics Research Unit, University of Oulu, Oulu, Finland
- School of Biological and Behavioural Sciences, Queen Mary University of London, London, E1 4NS, UK
| | - Feng Zhang
- Affiliated Hospital of Jiangnan University, Wuxi, Jiangsu, 214122, China
| | - Zhaoyang Qi
- State Key Laboratory of Food Science and Technology, School of Food Science and Technology, Jiangnan University, Wuxi, Jiangsu, 214122, China
| | - Lars Chittka
- School of Biological and Behavioural Sciences, Queen Mary University of London, London, E1 4NS, UK
| | - Wei Zhao
- State Key Laboratory of Food Science and Technology, School of Food Science and Technology, Jiangnan University, Wuxi, Jiangsu, 214122, China.
| |
Collapse
|
26
|
Hofmann CS, Carrington S, Keller AN, Gregory KJ, Niswender CM. Regulation and functional consequences of mGlu 4 RNA editing. RNA (NEW YORK, N.Y.) 2021; 27:1220-1240. [PMID: 34244459 PMCID: PMC8457003 DOI: 10.1261/rna.078729.121] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/01/2021] [Accepted: 07/01/2021] [Indexed: 06/13/2023]
Abstract
Metabotropic glutamate receptor 4 (mGlu4) is one of eight mGlu receptors within the Class C G protein-coupled receptor superfamily. mGlu4 is primarily localized to the presynaptic membrane of neurons where it functions as an auto and heteroreceptor controlling synaptic release of neurotransmitter. mGlu4 is implicated in numerous disorders and is a promising drug target; however, more remains to be understood about its regulation and pharmacology. Using high-throughput sequencing, we have validated and quantified an adenosine-to-inosine (A-to-I) RNA editing event that converts glutamine 124 to arginine in mGlu4; additionally, we have identified a rare but novel K129R site. Using an in vitro editing assay, we then validated the pre-mRNA duplex that allows for editing by ADAR enzymes and predicted its conservation across the mammalian species. Structural modeling of the mGlu4 protein predicts the Q124R substitution to occur in the B helix of the receptor that is critical for receptor dimerization and activation. Interestingly, editing of a receptor homodimer does not disrupt G protein activation in response to the endogenous agonist, glutamate. Using an assay designed to specifically measure heterodimer populations at the surface, however, we found that Q124R substitution decreased the propensity of mGlu4 to heterodimerize with mGlu2 and mGlu7 Our study is the first to extensively describe the extent and regulatory factors of RNA editing of mGlu4 mRNA transcripts. In addition, we have proposed a novel functional consequence of this editing event that provides insights regarding its effects in vivo and expands the regulatory capacity for mGlu receptors.
Collapse
MESH Headings
- Adenosine Deaminase/genetics
- Adenosine Deaminase/metabolism
- Amino Acid Sequence
- Animals
- Base Pairing
- Base Sequence
- Birds
- Cerebral Cortex/cytology
- Cerebral Cortex/metabolism
- Corpus Striatum/cytology
- Corpus Striatum/metabolism
- HEK293 Cells
- Hippocampus/cytology
- Hippocampus/metabolism
- Humans
- Models, Molecular
- Neurons/cytology
- Neurons/metabolism
- Nucleic Acid Conformation
- Point Mutation
- Protein Conformation, alpha-Helical
- Protein Conformation, beta-Strand
- RNA Editing
- RNA, Messenger/chemistry
- RNA, Messenger/genetics
- RNA, Messenger/metabolism
- RNA-Binding Proteins/genetics
- RNA-Binding Proteins/metabolism
- Rats
- Rats, Sprague-Dawley
- Receptors, Metabotropic Glutamate/chemistry
- Receptors, Metabotropic Glutamate/genetics
- Receptors, Metabotropic Glutamate/metabolism
- Reptiles
- Sequence Homology, Amino Acid
Collapse
Affiliation(s)
- Christopher S Hofmann
- Department of Pharmacology, Vanderbilt University, Nashville, Tennessee 37232, USA
- Warren Center for Neuroscience Drug Discovery, Vanderbilt University, Nashville, Tennessee 37232, USA
| | - Sheridan Carrington
- Department of Pharmacology, Vanderbilt University, Nashville, Tennessee 37232, USA
- Warren Center for Neuroscience Drug Discovery, Vanderbilt University, Nashville, Tennessee 37232, USA
| | - Andrew N Keller
- Department of Pharmacology and Drug Discovery Biology, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, Victoria 3052, Australia
| | - Karen J Gregory
- Department of Pharmacology and Drug Discovery Biology, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, Victoria 3052, Australia
| | - Colleen M Niswender
- Department of Pharmacology, Vanderbilt University, Nashville, Tennessee 37232, USA
- Warren Center for Neuroscience Drug Discovery, Vanderbilt University, Nashville, Tennessee 37232, USA
- Vanderbilt Kennedy Center, Vanderbilt University Medical Center, Nashville, Tennessee 37203, USA
| |
Collapse
|
27
|
Choi WM, Ryu T, Lee JH, Shim YR, Kim MH, Kim HH, Kim YE, Yang K, Kim K, Choi SE, Kim W, Kim SH, Eun HS, Jeong WI. Metabotropic Glutamate Receptor 5 in Natural Killer Cells Attenuates Liver Fibrosis by Exerting Cytotoxicity to Activated Stellate Cells. Hepatology 2021; 74:2170-2185. [PMID: 33932306 DOI: 10.1002/hep.31875] [Citation(s) in RCA: 36] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/22/2020] [Revised: 04/12/2021] [Accepted: 04/18/2021] [Indexed: 12/14/2022]
Abstract
BACKGROUND AND AIMS The important roles of glutamate and metabotropic glutamate receptor 5 (mGluR5) in HSCs have recently been reported in various liver diseases; however, the mechanism linking the glutamine/glutamate metabolism and mGluR5 in liver fibrosis remains unclear. Here, we report that mGluR5 activation in natural killer (NK) cells attenuates liver fibrosis through increased cytotoxicity and interferon-γ (IFN-γ) production in both mice and humans. APPROACH AND RESULTS Following 2-week injection of carbon tetrachloride (CCl4 ) or 5-week methionine-deficient and choline-deficient diet, liver fibrosis was more aggravated in mGluR5 knockout mice with significantly decreased frequency of NK cells compared with wild-type mice. Consistently, NK cell-specific mGluR5 knockout mice had aggravated CCl4 -induced liver fibrosis with decreased production of IFN-γ. Conversely, in vitro activation of mGluR5 in NK cells significantly increased the expression of anti-fibrosis-related genes including Ifng, Prf1 (perforin), and Klrk1 (killer cell lectin like receptor K1) and the production of IFN-γ through the mitogen-activated extracellular signal-regulated kinase/extracellular signal-related kinase pathway, contributing to the increased cytotoxicity against activated HSCs. However, we found that the uptake of glutamate was increased in activated HSCs, resulting in shortage of extracellular glutamate and reduced stimulation of mGluR5 in NK cells. Consequently, this could enable HSCs to evade NK cell cytotoxicity in advanced liver fibrosis. In vivo, pharmacologic activation of mGluR5 accelerated CCl4 -induced liver fibrosis regression by restoring NK cell cytotoxicity. In humans, mGluR5 activation enhanced the cytotoxicity of NK cells isolated from healthy donors, but not from patients with cirrhosis with significantly reduced mGluR5 expression in NK cells. CONCLUSIONS mGluR5 plays important roles in attenuating liver fibrosis by augmenting NK cell cytotoxicity, which could be used as a potential therapeutic target for liver fibrosis.
Collapse
Affiliation(s)
- Won-Mook Choi
- Laboratory of Liver Research, Graduate School of Medical Science and Engineering, KAIST, Daejeon, Republic of Korea.,Department of Gastroenterology, Liver Center, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Republic of Korea
| | - Tom Ryu
- Laboratory of Liver Research, Graduate School of Medical Science and Engineering, KAIST, Daejeon, Republic of Korea
| | - Jun-Hee Lee
- Laboratory of Liver Research, Graduate School of Medical Science and Engineering, KAIST, Daejeon, Republic of Korea
| | - Young-Ri Shim
- Laboratory of Liver Research, Graduate School of Medical Science and Engineering, KAIST, Daejeon, Republic of Korea
| | - Myung-Ho Kim
- Laboratory of Liver Research, Graduate School of Medical Science and Engineering, KAIST, Daejeon, Republic of Korea
| | - Hee-Hoon Kim
- Laboratory of Liver Research, Graduate School of Medical Science and Engineering, KAIST, Daejeon, Republic of Korea
| | - Ye Eun Kim
- Laboratory of Liver Research, Graduate School of Medical Science and Engineering, KAIST, Daejeon, Republic of Korea
| | - Keungmo Yang
- Laboratory of Liver Research, Graduate School of Medical Science and Engineering, KAIST, Daejeon, Republic of Korea
| | - Kyurae Kim
- Laboratory of Liver Research, Graduate School of Medical Science and Engineering, KAIST, Daejeon, Republic of Korea
| | - Sung Eun Choi
- Laboratory of Liver Research, Graduate School of Medical Science and Engineering, KAIST, Daejeon, Republic of Korea
| | - Won Kim
- Department of Internal Medicine, Seoul Metropolitan Government Seoul National University Boramae Medical Center, Seoul, Republic of Korea
| | - Seok-Hwan Kim
- Department of Surgery, College of Medicine, Chungnam National University, Daejeon, Republic of Korea
| | - Hyuk Soo Eun
- Department of Internal Medicine, School of Medicine, Chungnam National University, Daejeon, Republic of Korea
| | - Won-Il Jeong
- Laboratory of Liver Research, Graduate School of Medical Science and Engineering, KAIST, Daejeon, Republic of Korea.,Biomedical Research Center, KAIST, Daejeon, Republic of Korea
| |
Collapse
|
28
|
Glutamate triggers the expression of functional ionotropic and metabotropic glutamate receptors in mast cells. Cell Mol Immunol 2021; 18:2383-2392. [PMID: 32313211 PMCID: PMC8484602 DOI: 10.1038/s41423-020-0421-z] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2019] [Accepted: 03/16/2020] [Indexed: 02/07/2023] Open
Abstract
Mast cells are emerging as players in the communication between peripheral nerve endings and cells of the immune system. However, it is not clear the mechanism by which mast cells communicate with peripheral nerves. We previously found that mast cells located within healing tendons can express glutamate receptors, raising the possibility that mast cells may be sensitive to glutamate signaling. To evaluate this hypothesis, we stimulated primary mast cells with glutamate and showed that glutamate induced the profound upregulation of a panel of glutamate receptors of both the ionotropic type (NMDAR1, NMDAR2A, and NMDAR2B) and the metabotropic type (mGluR2 and mGluR7) at both the mRNA and protein levels. The binding of glutamate to glutamate receptors on the mast cell surface was confirmed. Further, glutamate had extensive effects on gene expression in the mast cells, including the upregulation of pro-inflammatory components such as IL-6 and CCL2. Glutamate also induced the upregulation of transcription factors, including Egr2, Egr3 and, in particular, FosB. The extensive induction of FosB was confirmed by immunofluorescence assessment. Glutamate receptor antagonists abrogated the responses of the mast cells to glutamate, supporting the supposition of a functional glutamate-glutamate receptor axis in mast cells. Finally, we provide in vivo evidence supporting a functional glutamate-glutamate receptor axis in the mast cells of injured tendons. Together, these findings establish glutamate as an effector of mast cell function, thereby introducing a novel principle for how cells in the immune system can communicate with nerve cells.
Collapse
|
29
|
Moldovan OL, Rusu A, Tanase C, Vari CE. Glutamate - A multifaceted molecule: Endogenous neurotransmitter, controversial food additive, design compound for anti-cancer drugs. A critical appraisal. Food Chem Toxicol 2021; 153:112290. [PMID: 34023459 DOI: 10.1016/j.fct.2021.112290] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2021] [Revised: 05/10/2021] [Accepted: 05/14/2021] [Indexed: 12/18/2022]
Abstract
One of the most widely used flavour enhancers in the food industry is monosodium glutamate (MSG). MSG consumption has been on an upward trend, worrying in terms of potential toxic effects. This review is focused on the long-term toxicity of MSG and the experimental evidence that supports it. The article's primary purpose was to survey recently published data regarding the consumption of MSG within safe limits. The administered doses in animal models are very varied and have given rise to controversy. Also, the paper comprises pathways to lower MSG toxicity and highlight other underexploited biological effects, as anti-cancer potential. The administration of MSG, combined with various compounds, has been shown benefit against toxic effects. Several recent studies have identified a possible mechanism that recommends MSG and some derivatives as potential anti-cancer agents. New anti-cancer compounds based on the glutamic acid structure must be studied and further exploited. International regulations require harmonization of safe doses of MSG based on current scientific studies. Replacing MSG with other umami flavour enhancers may be a safer alternative for human health in the future. The biological consequences of MSG consumption or therapeutical administration have not been fully deciphered yet.
Collapse
Affiliation(s)
- Octavia-Laura Moldovan
- Medicine and Pharmacy Doctoral School, George Emil Palade University of Medicine, Pharmacy, Science and Technology of Târgu Mureș, 540142, Târgu Mureș, Romania.
| | - Aura Rusu
- Pharmaceutical and Therapeutic Chemistry Department, Faculty of Pharmacy, George Emil Palade University of Medicine, Pharmacy, Science and Technology of Târgu Mureș, 540142, Târgu Mureș, Romania.
| | - Corneliu Tanase
- Pharmaceutical Botany Department, Faculty of Pharmacy, George Emil Palade University of Medicine, Pharmacy, Science and Technology of Târgu Mureș, 540142, Târgu Mureș, Romania.
| | - Camil-Eugen Vari
- Pharmacy and Clinical Pharmacy Department, Faculty of Pharmacy, George Emil Palade University of Medicine, Pharmacy, Science and Technology of Târgu Mureș, 540142, Târgu Mureș, Romania.
| |
Collapse
|
30
|
Szpręgiel I, Wrońska D, Kmiecik M, Pałka S, Kania BF. Glutamic Acid Decarboxylase Concentration Changes in Response to Stress and Altered Availability of Glutamic Acid in Rabbit ( Oryctolagus cuniculus) Brain Limbic Structures. Animals (Basel) 2021; 11:455. [PMID: 33572286 PMCID: PMC7915518 DOI: 10.3390/ani11020455] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2020] [Revised: 01/31/2021] [Accepted: 02/04/2021] [Indexed: 12/18/2022] Open
Abstract
Glutamic acid decarboxylase (GAD) is an enzyme that catalyses the formation of γ-aminobutyric acid (GABA), the most important inhibitory neurotransmitter, from glutamic acid (Glu), which is considered the most important excitatory transmitter in the central and peripheral nervous systems. GAD is a key enzyme that provides a balance between Glu and GABA concentration. Hence, it can be assumed that if the GAD executes the synthesis of GABA from Glu, it is important in the stress response, and thus also in triggering the emotional states of the body that accompany stress. The aim of the study was to investigate the concentration of the GAD in motivational structures in the brain of the rabbit (Oryctolagus cuniculus) under altered homeostatic conditions caused by stress and variable availability of Glu. Summarising, the experimental results clearly showed variable concentrations of GAD in the motivational structures of the rabbit brain. The highest concentration of GAD was found in the hypothalamus, which suggests a strong effect of Glu and GABA on the activity of this brain structure. The GAD concentrations in individual experimental groups depended to a greater extent on blocking the activity of glutamate receptors than on the effects of a single stress exposure. The results obtained clearly support the possibility that a rapid change in the concentration of GAD could shift bodily responses to quickly achieve homeostasis, especially in this species. Further studies are necessary to reveal the role of the Glu-GAD-GABA system in the modulation of stress situations as well as in body homeostasis.
Collapse
Affiliation(s)
- Izabela Szpręgiel
- Department of Animal Physiology and Endocrinology, Faculty of Animal Sciences, University of Agriculture in Krakow, Al. Mickiewicza 24/28, 30-059 Kraków, Poland;
| | - Danuta Wrońska
- Department of Animal Physiology and Endocrinology, Faculty of Animal Sciences, University of Agriculture in Krakow, Al. Mickiewicza 24/28, 30-059 Kraków, Poland;
| | - Michał Kmiecik
- Department of Genetics, Animal Breeding and Ethology, Faculty of Animal Sciences, University of Agriculture in Krakow, Al. Mickiewicza 24/28, 30-059 Kraków, Poland; (M.K.); (S.P.)
| | - Sylwia Pałka
- Department of Genetics, Animal Breeding and Ethology, Faculty of Animal Sciences, University of Agriculture in Krakow, Al. Mickiewicza 24/28, 30-059 Kraków, Poland; (M.K.); (S.P.)
| | - Bogdan F. Kania
- University Centre of Veterinary Medicine JU-AU, University of Agriculture in Kraków, Mickiewicza 24/28, 30-059 Kraków, Poland;
| |
Collapse
|
31
|
Xiao G, Lyu M, Li Z, Cao L, Liu X, Wang Y, He S, Chen Z, Du H, Feng Y, Wang J, Zhu Y. Restoration of early deficiency of axonal guidance signaling by guanxinning injection as a novel therapeutic option for acute ischemic stroke. Pharmacol Res 2021; 165:105460. [PMID: 33513357 DOI: 10.1016/j.phrs.2021.105460] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/16/2020] [Revised: 12/22/2020] [Accepted: 01/22/2021] [Indexed: 01/12/2023]
Abstract
Despite of its high morbidity and mortality, there is still a lack of effective treatment for ischemic stroke in part due to our incomplete understanding of molecular mechanisms of its pathogenesis. In this study, we demonstrate that SHH-PTCH1-GLI1-mediated axonal guidance signaling and its related neurogenesis, a central pathway for neuronal development, also plays a critical role in early stage of an acute stroke model. Specifically, in vivo, we evaluated the effect of GXNI on ischemic stroke mice via using the middle cerebral artery embolization model, and found that GXNI significantly alleviated cerebral ischemic reperfusion (I/R) injury by reducing the volume of cerebral infarction, neurological deficit score and cerebral edema, reversing the BBB permeability and histopathological changes. A combined approach of RNA-seq and network pharmacology analysis was used to reveal the underlying mechanisms of GXNI followed by RT-PCR, immunohistochemistry and western blotting validation. It was pointed out that axon guidance signaling pathway played the most prominent role in GXNI action with Shh, Ptch1, and Gli1 genes as the critical contributors in brain protection. In addition, GXNI markedly prevented primary cortical neuron cells from oxygen-glucose deprivation/reoxygenation damage in vitro, and promoted axon growth and synaptogenesis of damaged neurons, which further confirmed the results of in vivo experiments. Moreover, due to the inhibition of the SHH-PTCH1-GLI1 signaling pathway by cyclopropylamine, the effect of GXNI was significantly weakened. Hence, our study provides a novel option for the clinical treatment of acute ischemic stroke by GXNI via SHH-PTCH1-GLI1-mediated axonal guidance signaling, a neuronal development pathway previously considered for after-stroke recovery.
Collapse
Affiliation(s)
- Guangxu Xiao
- State Key Laboratory of Component-based Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Beihua South Road, JingHai District, Tianjin, 301617, China; Research and Development Center of TCM, Tianjin International Joint Academy of Biotechnology & Medicine, 220 Dongting Road, TEDA, Tianjin, 300457, China
| | - Ming Lyu
- State Key Laboratory of Component-based Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Beihua South Road, JingHai District, Tianjin, 301617, China; Research and Development Center of TCM, Tianjin International Joint Academy of Biotechnology & Medicine, 220 Dongting Road, TEDA, Tianjin, 300457, China; Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing, 100700, China
| | - Zhixiong Li
- State Key Laboratory of Component-based Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Beihua South Road, JingHai District, Tianjin, 301617, China; Research and Development Center of TCM, Tianjin International Joint Academy of Biotechnology & Medicine, 220 Dongting Road, TEDA, Tianjin, 300457, China
| | - Linghua Cao
- State Key Laboratory of Component-based Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Beihua South Road, JingHai District, Tianjin, 301617, China; Research and Development Center of TCM, Tianjin International Joint Academy of Biotechnology & Medicine, 220 Dongting Road, TEDA, Tianjin, 300457, China
| | - Xinyan Liu
- State Key Laboratory of Component-based Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Beihua South Road, JingHai District, Tianjin, 301617, China; Research and Development Center of TCM, Tianjin International Joint Academy of Biotechnology & Medicine, 220 Dongting Road, TEDA, Tianjin, 300457, China
| | - Yule Wang
- State Key Laboratory of Component-based Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Beihua South Road, JingHai District, Tianjin, 301617, China; Research and Development Center of TCM, Tianjin International Joint Academy of Biotechnology & Medicine, 220 Dongting Road, TEDA, Tianjin, 300457, China
| | - Shuang He
- State Key Laboratory of Component-based Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Beihua South Road, JingHai District, Tianjin, 301617, China; Research and Development Center of TCM, Tianjin International Joint Academy of Biotechnology & Medicine, 220 Dongting Road, TEDA, Tianjin, 300457, China
| | - Zihao Chen
- State Key Laboratory of Component-based Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Beihua South Road, JingHai District, Tianjin, 301617, China; Research and Development Center of TCM, Tianjin International Joint Academy of Biotechnology & Medicine, 220 Dongting Road, TEDA, Tianjin, 300457, China
| | - Hongxia Du
- State Key Laboratory of Component-based Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Beihua South Road, JingHai District, Tianjin, 301617, China; Research and Development Center of TCM, Tianjin International Joint Academy of Biotechnology & Medicine, 220 Dongting Road, TEDA, Tianjin, 300457, China
| | - Yuxin Feng
- State Key Laboratory of Component-based Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Beihua South Road, JingHai District, Tianjin, 301617, China; Research and Development Center of TCM, Tianjin International Joint Academy of Biotechnology & Medicine, 220 Dongting Road, TEDA, Tianjin, 300457, China
| | - Jigang Wang
- Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing, 100700, China
| | - Yan Zhu
- State Key Laboratory of Component-based Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Beihua South Road, JingHai District, Tianjin, 301617, China; Research and Development Center of TCM, Tianjin International Joint Academy of Biotechnology & Medicine, 220 Dongting Road, TEDA, Tianjin, 300457, China.
| |
Collapse
|
32
|
Lehn-Stefan A, Peter A, Machann J, Schick F, Randrianarisoa E, Heni M, Wagner R, Birkenfeld AL, Fritsche A, Häring HU, Staiger H, Stefan N. Elevated Circulating Glutamate Is Associated With Subclinical Atherosclerosis Independently of Established Risk Markers: A Cross-Sectional Study. J Clin Endocrinol Metab 2021; 106:e982-e989. [PMID: 33277657 DOI: 10.1210/clinem/dgaa898] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/29/2020] [Indexed: 12/20/2022]
Abstract
OBJECTIVE Elevated plasma glutamate levels are associated with an increased risk of cardiovascular disease (CVD). Because plasma glutamate levels are also strongly associated with visceral adiposity, nonalcoholic fatty liver disease, insulin resistance, and high circulating levels of branched-chain amino acids (BCAAs), it is unknown to what extent elevated circulating glutamate is an independent marker of an increased risk of atherosclerosis. METHODS Plasma levels of glutamate and BCAAs were measured in 102 individuals who were precisely phenotyped for body fat mass and distribution (magnetic resonance [MR] tomography), liver fat content (1H-MR spectroscopy), insulin sensitivity (oral glucose tolerance test and hyperinsulinemic, euglycemic clamp [N = 57]), and carotid intima media thickness (cIMT). RESULTS Plasma glutamate levels, adjusted for age, sex, body fat mass, and visceral fat mass, correlated positively with liver fat content and cIMT (all std β ≥ .22, all P ≤ .023) and negatively with insulin sensitivity (std β ≤ -.31, P ≤ .002). Glutamate levels also were associated with cIMT, independently of additional adjustment for liver fat content, insulin sensitivity and BCAAs levels (std β ≥ .24, P ≤ .02). Furthermore, an independent positive association of glutamate and interleukin-6 (IL-6) levels was observed (N = 50; std β = .39, P = .03). Although glutamate, adjusted for age, sex, body fat mass, and visceral fat mass, also correlated positively with cIMT in this subgroup (std β = .31, P = .02), after additional adjustment for the parameters liver fat content, insulin sensitivity, BCAAs, or IL-6 levels, adjustment for IL-6 most strongly attenuated this relationship (std β = .28, P = .05). CONCLUSIONS Elevated plasma glutamate levels are associated with increased cIMT, independently of established CVD risk factors, and this relationship may in part be explained by IL-6-associated subclinical inflammation.
Collapse
Affiliation(s)
- Angela Lehn-Stefan
- Institute of Diabetes Research and Metabolic Diseases (IDM) of the Helmholtz Centre Munich, Tübingen, Germany
- German Center for Diabetes Research (DZD), München-Neuherberg, Germany
| | - Andreas Peter
- Institute of Diabetes Research and Metabolic Diseases (IDM) of the Helmholtz Centre Munich, Tübingen, Germany
- German Center for Diabetes Research (DZD), München-Neuherberg, Germany
- Institute for Clinical Chemistry and Pathobiochemistry, Department for Diagnostic Laboratory Medicine, University Hospital of Tübingen, Tübingen, Germany
| | - Jürgen Machann
- Institute of Diabetes Research and Metabolic Diseases (IDM) of the Helmholtz Centre Munich, Tübingen, Germany
- German Center for Diabetes Research (DZD), München-Neuherberg, Germany
- Section of Experimental Radiology, Department of Diagnostic and Interventional Radiology, University of Tübingen, Tübingen, Germany
| | - Fritz Schick
- Institute of Diabetes Research and Metabolic Diseases (IDM) of the Helmholtz Centre Munich, Tübingen, Germany
- German Center for Diabetes Research (DZD), München-Neuherberg, Germany
- Section of Experimental Radiology, Department of Diagnostic and Interventional Radiology, University of Tübingen, Tübingen, Germany
| | - Elko Randrianarisoa
- Institute of Diabetes Research and Metabolic Diseases (IDM) of the Helmholtz Centre Munich, Tübingen, Germany
- German Center for Diabetes Research (DZD), München-Neuherberg, Germany
| | - Martin Heni
- Institute of Diabetes Research and Metabolic Diseases (IDM) of the Helmholtz Centre Munich, Tübingen, Germany
- German Center for Diabetes Research (DZD), München-Neuherberg, Germany
- Department of Internal Medicine IV, Division of Endocrinology, Diabetology and Nephrology, University of Tübingen, Tübingen, Germany
| | - Robert Wagner
- Institute of Diabetes Research and Metabolic Diseases (IDM) of the Helmholtz Centre Munich, Tübingen, Germany
- German Center for Diabetes Research (DZD), München-Neuherberg, Germany
- Department of Internal Medicine IV, Division of Endocrinology, Diabetology and Nephrology, University of Tübingen, Tübingen, Germany
| | - Andreas L Birkenfeld
- Institute of Diabetes Research and Metabolic Diseases (IDM) of the Helmholtz Centre Munich, Tübingen, Germany
- German Center for Diabetes Research (DZD), München-Neuherberg, Germany
- Department of Internal Medicine IV, Division of Endocrinology, Diabetology and Nephrology, University of Tübingen, Tübingen, Germany
| | - Andreas Fritsche
- Institute of Diabetes Research and Metabolic Diseases (IDM) of the Helmholtz Centre Munich, Tübingen, Germany
- German Center for Diabetes Research (DZD), München-Neuherberg, Germany
- Department of Internal Medicine IV, Division of Endocrinology, Diabetology and Nephrology, University of Tübingen, Tübingen, Germany
| | - Hans-Ulrich Häring
- Institute of Diabetes Research and Metabolic Diseases (IDM) of the Helmholtz Centre Munich, Tübingen, Germany
- German Center for Diabetes Research (DZD), München-Neuherberg, Germany
- Department of Internal Medicine IV, Division of Endocrinology, Diabetology and Nephrology, University of Tübingen, Tübingen, Germany
| | - Harald Staiger
- Institute of Diabetes Research and Metabolic Diseases (IDM) of the Helmholtz Centre Munich, Tübingen, Germany
- German Center for Diabetes Research (DZD), München-Neuherberg, Germany
- Institute of Pharmacological Sciences, Department of Pharmacy and Biochemistry, University of Tübingen, Tübingen, Germany
| | - Norbert Stefan
- Institute of Diabetes Research and Metabolic Diseases (IDM) of the Helmholtz Centre Munich, Tübingen, Germany
- German Center for Diabetes Research (DZD), München-Neuherberg, Germany
- Department of Internal Medicine IV, Division of Endocrinology, Diabetology and Nephrology, University of Tübingen, Tübingen, Germany
| |
Collapse
|
33
|
Ramadhani A, Sofro ZM, Partadiredja G. The effect of oral administration of monosodium glutamate on orofacial pain response and the estimated number of trigeminal ganglion sensory neurons of male Wistar rats. BIO WEB OF CONFERENCES 2021. [DOI: 10.1051/bioconf/20214105007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/14/2022] Open
Abstract
Monosodium glutamate (MSG) is a worldwide flavor enhancer. The excessive glutamate concentration in nerve tissue induces the death of nerve cells, known as excitotoxicity. In the orofacial region, the nerve cells’ death affects pain perception such as mechanical hyperalgesia and allodynia. The aim of the present study was to examine the pain response modification and the estimated total number of trigeminal ganglion sensory neurons after sub chronic oral administration of MSG. Twenty eight male Wistar rats, aged 6-8 weeks (100-150 grams) were divided into 4 groups: Control (2 mL NaCl 0.9%); 1 mg/gWB MSG; 2 mg/gWB MSG; 4 mg/gWB MSG groups. Daily oral administration of MSG was given for 30 days. The control group received NaCl per oral for the same period. The pin prick and air puff test were performed on days 1-2, days 41-42 and days 55-56. The number of trigeminal ganglion sensory neurons were estimated by the unbiased stereology method, using the approach of numerical density and organ volume reference. The results showed that the sub chronic oral administration of MSG does not modify either the orofacial pain response or the estimated total number of trigeminal ganglion sensory neurons. .
Collapse
|
34
|
Gregory KJ, Goudet C. International Union of Basic and Clinical Pharmacology. CXI. Pharmacology, Signaling, and Physiology of Metabotropic Glutamate Receptors. Pharmacol Rev 2021; 73:521-569. [PMID: 33361406 DOI: 10.1124/pr.119.019133] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
Metabotropic glutamate (mGlu) receptors respond to glutamate, the major excitatory neurotransmitter in the mammalian brain, mediating a modulatory role that is critical for higher-order brain functions such as learning and memory. Since the first mGlu receptor was cloned in 1992, eight subtypes have been identified along with many isoforms and splice variants. The mGlu receptors are transmembrane-spanning proteins belonging to the class C G protein-coupled receptor family and represent attractive targets for a multitude of central nervous system disorders. Concerted drug discovery efforts over the past three decades have yielded a wealth of pharmacological tools including subtype-selective agents that competitively block or mimic the actions of glutamate or act allosterically via distinct sites to enhance or inhibit receptor activity. Herein, we review the physiologic and pathophysiological roles for individual mGlu receptor subtypes including the pleiotropic nature of intracellular signal transduction arising from each. We provide a comprehensive analysis of the in vitro and in vivo pharmacological properties of prototypical and commercially available orthosteric agonists and antagonists as well as allosteric modulators, including ligands that have entered clinical trials. Finally, we highlight emerging areas of research that hold promise to facilitate rational design of highly selective mGlu receptor-targeting therapeutics in the future. SIGNIFICANCE STATEMENT: The metabotropic glutamate receptors are attractive therapeutic targets for a range of psychiatric and neurological disorders. Over the past three decades, intense discovery efforts have yielded diverse pharmacological tools acting either competitively or allosterically, which have enabled dissection of fundamental biological process modulated by metabotropic glutamate receptors and established proof of concept for many therapeutic indications. We review metabotropic glutamate receptor molecular pharmacology and highlight emerging areas that are offering new avenues to selectively modulate neurotransmission.
Collapse
Affiliation(s)
- Karen J Gregory
- Drug Discovery Biology, Monash Institute of Pharmaceutical Sciences and Department of Pharmacology, Monash University, Parkville, Victoria, Australia (K.J.G.) and Institut de Génomique Fonctionnelle (IGF), University of Montpellier, Centre National de la Recherche Scientifique (CNRS), Institut National de la Sante et de la Recherche Medicale (INSERM), Montpellier, France (C.G.)
| | - Cyril Goudet
- Drug Discovery Biology, Monash Institute of Pharmaceutical Sciences and Department of Pharmacology, Monash University, Parkville, Victoria, Australia (K.J.G.) and Institut de Génomique Fonctionnelle (IGF), University of Montpellier, Centre National de la Recherche Scientifique (CNRS), Institut National de la Sante et de la Recherche Medicale (INSERM), Montpellier, France (C.G.)
| |
Collapse
|
35
|
Zaghmi A, Dopico-López A, Pérez-Mato M, Iglesias-Rey R, Hervella P, Greschner AA, Bugallo-Casal A, da Silva A, Gutiérrez-Fernández M, Castillo J, Pérez FC, Gauthier MA. Sustained blood glutamate scavenging enhances protection in ischemic stroke. Commun Biol 2020; 3:729. [PMID: 33273696 PMCID: PMC7713697 DOI: 10.1038/s42003-020-01406-1] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2020] [Accepted: 10/21/2020] [Indexed: 12/20/2022] Open
Abstract
Stroke is a major cause of morbidity, mortality, and disability. During ischemic stroke, a marked and prolonged rise of glutamate concentration in the brain causes neuronal cell death. This study explores the protective effect of a bioconjugate form of glutamate oxaloacetate transaminase (hrGOT), which catalyzes the depletion of blood glutamate in the bloodstream for ~6 days following a single administration. When treated with this bioconjugate, a significant reduction of the infarct volume and a better retention of sensorimotor function was observed for ischemic rats compared to those treated with saline. Moreover, the equivalent dose of native hrGOT yielded similar results to the saline treated group for some tests. Targeting the bioconjugate to the blood-brain-barrier did not improve its performance. The data suggest that the bioconjugates draw glutamate out of the brain by displacing homeostasis between the different glutamate pools of the body.
Collapse
Affiliation(s)
- Ahlem Zaghmi
- Institut National de la Recherche Scientifique (INRS), EMT Research Center, Varennes, Qc, J3X 1S2, Canada
| | - Antonio Dopico-López
- Clinical Neuroscience Research Laboratory, Health Research Institute of Santiago de Compostela (IDIS), Santiago de Compostela, Spain
| | - María Pérez-Mato
- Clinical Neuroscience Research Laboratory, Health Research Institute of Santiago de Compostela (IDIS), Santiago de Compostela, Spain
- Neuroscience and Cerebrovascular Research Laboratory, Department of Neurology and Stroke Center, La Paz University Hospital, Neuroscience Area of IdiPAZ Health Research Institute, Universidad Autónoma de Madrid, Madrid, Spain
| | - Ramón Iglesias-Rey
- Clinical Neuroscience Research Laboratory, Health Research Institute of Santiago de Compostela (IDIS), Santiago de Compostela, Spain
| | - Pablo Hervella
- Clinical Neuroscience Research Laboratory, Health Research Institute of Santiago de Compostela (IDIS), Santiago de Compostela, Spain
| | - Andrea A Greschner
- Institut National de la Recherche Scientifique (INRS), EMT Research Center, Varennes, Qc, J3X 1S2, Canada
| | - Ana Bugallo-Casal
- Clinical Neuroscience Research Laboratory, Health Research Institute of Santiago de Compostela (IDIS), Santiago de Compostela, Spain
| | - Andrés da Silva
- Clinical Neuroscience Research Laboratory, Health Research Institute of Santiago de Compostela (IDIS), Santiago de Compostela, Spain
| | - María Gutiérrez-Fernández
- Neuroscience and Cerebrovascular Research Laboratory, Department of Neurology and Stroke Center, La Paz University Hospital, Neuroscience Area of IdiPAZ Health Research Institute, Universidad Autónoma de Madrid, Madrid, Spain
| | - José Castillo
- Clinical Neuroscience Research Laboratory, Health Research Institute of Santiago de Compostela (IDIS), Santiago de Compostela, Spain
| | - Francisco Campos Pérez
- Clinical Neuroscience Research Laboratory, Health Research Institute of Santiago de Compostela (IDIS), Santiago de Compostela, Spain.
| | - Marc A Gauthier
- Institut National de la Recherche Scientifique (INRS), EMT Research Center, Varennes, Qc, J3X 1S2, Canada.
| |
Collapse
|
36
|
Kim HH, Choi SE, Jeong WI. Oxidative stress and glutamate excretion in alcoholic steatosis: Metabolic synapse between hepatocyte and stellate cell. Clin Mol Hepatol 2020; 26:697-704. [PMID: 33053940 PMCID: PMC7641576 DOI: 10.3350/cmh.2020.0152] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/05/2020] [Accepted: 08/27/2020] [Indexed: 12/12/2022] Open
Abstract
Chronic alcohol consumption induces the development of alcoholic steatosis in the liver, which is one of the most widespread liver diseases worldwide. During general alcohol metabolism, hepatocytes generate mitochondria- and cytochrome P450 2E1 (CYP2E1)-mediated reactive oxygen species (ROS) whose accumulation elicits activation of the hepatic anti-oxidant system, including glutathione (GSH). However, chronic alcohol consumption decreases GSH generation through cysteine deficiency by suppressing the methionine cycle and trans-sulfuration system, whereas it turns on an alternative defense pathway, such as the xCT transporter, to compensate for GSH shortage. The xCT transporter mediates the uptake of cystine coupled to the efflux of glutamate, leading to an increase in blood glutamate. In response to the elevated glutamate in the liver, the expression of metabotropic glutamate receptor 5 (mGluR5) is up-regulated in hepatic stellate cells (HSCs) along with enhanced production of 2-arachidonoylglycerol, which in turn stimulates cannabinoid receptor 1 (CB1R) on neighboring hepatocytes to increase de novo lipogenesis. On the other hand, blockade of mGluR5 and CB1R attenuates alcoholic steatosis. Interestingly, although the increased expression of CYP2E1-mediated xCT and ROS generation are mainly observed at the perivenous area (zone 3), fat accumulation is mostly detected at hepatic zone 2. To resolve this discrepancy, this review summarizes recent advances on glutamate/mGluR5-derived alcoholic steatosis and zone-dependently different responses to alcohol intake. In addition, the bidirectional loop pathway and its unique metabolic synapse between hepatocytes and HSCs are discussed.
Collapse
Affiliation(s)
- Hee-Hoon Kim
- Lab of Liver Research, Graduate School of Medical Science and Engineering, Korea Advanced Institute of Science and Technology (KAIST), Daejeon, Korea
| | - Sung Eun Choi
- Lab of Liver Research, Graduate School of Medical Science and Engineering, Korea Advanced Institute of Science and Technology (KAIST), Daejeon, Korea
| | - Won-Il Jeong
- Lab of Liver Research, Graduate School of Medical Science and Engineering, Korea Advanced Institute of Science and Technology (KAIST), Daejeon, Korea
| |
Collapse
|
37
|
Cachope R, Pereda AE. Regulatory Roles of Metabotropic Glutamate Receptors on Synaptic Communication Mediated by Gap Junctions. Neuroscience 2020; 456:85-94. [PMID: 32619474 DOI: 10.1016/j.neuroscience.2020.06.034] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2020] [Revised: 06/19/2020] [Accepted: 06/22/2020] [Indexed: 12/18/2022]
Abstract
Variations of synaptic strength are thought to underlie forms of learning and can functionally reshape neural circuits. Metabotropic glutamate receptors play key roles in regulating the strength of chemical synapses. However, information within neural circuits is also conveyed via a second modality of transmission: gap junction-mediated synapses. We review here evidence indicating that metabotropic glutamate receptors also play important roles in the regulation of synaptic communication mediated by neuronal gap junctions, also known as 'electrical synapses'. Activity-driven interactions between metabotropic glutamate receptors and neuronal gap junctions can lead to long-term changes in the strength of electrical synapses. Further, the regulatory action of metabotropic glutamate receptors on neuronal gap junctions is not restricted to adulthood but is also of critical relevance during brain development and contributes to the pathological mechanisms that follow brain injury.
Collapse
Affiliation(s)
- Roger Cachope
- CHDI Foundation, USA; Dominick P. Purpura Department of Neuroscience, Albert Einstein College of Medicine, Bronx, NY, USA
| | - Alberto E Pereda
- Dominick P. Purpura Department of Neuroscience, Albert Einstein College of Medicine, Bronx, NY, USA.
| |
Collapse
|
38
|
van Sadelhoff JHJ, Wiertsema SP, Garssen J, Hogenkamp A. Free Amino Acids in Human Milk: A Potential Role for Glutamine and Glutamate in the Protection Against Neonatal Allergies and Infections. Front Immunol 2020; 11:1007. [PMID: 32547547 PMCID: PMC7270293 DOI: 10.3389/fimmu.2020.01007] [Citation(s) in RCA: 35] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2020] [Accepted: 04/28/2020] [Indexed: 12/12/2022] Open
Abstract
Breastfeeding is indicated to support neonatal immune development and to protect against neonatal infections and allergies. Human milk composition is widely studied in relation to these unique abilities, which has led to the identification of various immunomodulating components in human milk, including various bioactive proteins. In addition to proteins, human milk contains free amino acids (FAAs), which have not been well-studied. Of those, the FAAs glutamate and glutamine are by far the most abundant. Levels of these FAAs in human milk sharply increase during the first months of lactation, in contrast to most other FAAs. These unique dynamics are globally consistent, suggesting that their levels in human milk are tightly regulated throughout lactation and, consequently, that they might have specific roles in the developing neonate. Interestingly, free glutamine and glutamate are reported to exhibit immunomodulating capacities, indicating that these FAAs could contribute to neonatal immune development and to the unique protective effects of breastfeeding. This review describes the current understanding of the FAA composition in human milk. Moreover, it provides an overview of the effects of free glutamine and glutamate on immune parameters relevant for allergic sensitization and infections in early life. The data reviewed provide rationale to study the role of free glutamine and glutamate in human milk in the protection against neonatal allergies and infections.
Collapse
Affiliation(s)
- Joris H J van Sadelhoff
- Division of Pharmacology, Utrecht Institute for Pharmaceutical Sciences, Faculty of Science, Utrecht University, Utrecht, Netherlands
| | | | - Johan Garssen
- Division of Pharmacology, Utrecht Institute for Pharmaceutical Sciences, Faculty of Science, Utrecht University, Utrecht, Netherlands.,Danone Nutricia Research, Utrecht, Netherlands
| | - Astrid Hogenkamp
- Division of Pharmacology, Utrecht Institute for Pharmaceutical Sciences, Faculty of Science, Utrecht University, Utrecht, Netherlands
| |
Collapse
|
39
|
Fan YN, Li C, Huang L, Chen L, Tang Z, Han G, Liu Y. Characterization of Group I Metabotropic Glutamate Receptors in Rat and Human Adrenal Glands. Front Physiol 2020; 11:401. [PMID: 32536873 PMCID: PMC7267184 DOI: 10.3389/fphys.2020.00401] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2019] [Accepted: 04/03/2020] [Indexed: 11/13/2022] Open
Abstract
Glutamate and its receptors have been demonstrated to promote both basal and nicotine-evoked catecholamine release in bovine chromaffin cells. Multiple glutamate receptors, including metabotropic glutamate receptors (mGluRs), are found in the adrenal glands of several species, as well as in chromaffin cells. However, there is limited information available regarding the expression of glutamate metabotropic receptor (GRM)1-8 mRNAs and the detailed localization of group I mGluRs (mGluR1 and mGluR5) in the rat and human adrenal cortex and medulla. Therefore, we examined mRNA expression of GRM1-8 subunits using reverse transcription-polymerase chain reaction (RT-PCR) and the distribution of mGluR1 and mGluR5 by immunostaining. The results showed that the GRM1-8 mRNAs were expressed in both the cortex and medulla of rat and human adrenal glands with the exception of GRM1, which was not detectable in the rat adrenal cortex. Immunostaining of mGluR1 revealed that it was localized only in the adrenal medulla of rats but was present in both the adrenal cortex and medulla in humans. In the adrenal medulla, the central part of the adrenal glands, mGluR1 was detected in chromaffin cells but not in nerve fibers and ganglion cells. Immunoactivity of mGluR5 was visible in the capillary wall throughout the adrenal cortex and medulla in rat and human samples. Its immunoactivity was also observed in ganglion cells in the rat adrenal medulla. There was no mGluR5 immunoactivity detected in chromaffin cells and nerve fibers in the rat and human adrenal medulla. Using dissected rat adrenal medulla as a model, we found that treatment with a mGluR1 agonist activated extracellular signal-regulated kinase (ERK) 1/2 and increased the expression of tyrosine hydroxylase (TH), the rate-limiting enzyme of catecholamine synthesis. Moreover, these results showed that mGluR1 signaling was involved in hypoxia-induced upregulation of TH in the rat adrenal medulla. This study shows the expression of GRM1-8 mRNAs in rat and human adrenal glands and indicates that glutamate, through the activation of mGluRs, may play various physiological roles in the adrenal gland. Furthermore, mGluR1 may be involved in catecholamine biosynthesis by regulating TH, and mGluR5 may affect cortical and medullar hormone levels by regulating microvascular function.
Collapse
Affiliation(s)
- Ya-Nan Fan
- Henan Key Laboratory of Neural Regeneration and Repairment, The First Affiliated Hospital of Xinxiang Medical University, Henan Neurology Institute, Weihui, China
| | - Chaohong Li
- Henan Key Laboratory of Neural Regeneration and Repairment, The First Affiliated Hospital of Xinxiang Medical University, Henan Neurology Institute, Weihui, China
| | - Lu Huang
- Henan Key Laboratory of Neural Regeneration and Repairment, The First Affiliated Hospital of Xinxiang Medical University, Henan Neurology Institute, Weihui, China
| | - Lingyun Chen
- Operating Room, The First Affiliated Hospital of Xinxiang Medical University, Weihui, China
| | - Zhao Tang
- Department of Urology Surgery, The First Affiliated Hospital of Xinxiang Medical University, Weihui, China
| | - Guangye Han
- Department of Urology Surgery, The First Affiliated Hospital of Xinxiang Medical University, Weihui, China
| | - Yuzhen Liu
- Henan Key Laboratory of Neural Regeneration and Repairment, The First Affiliated Hospital of Xinxiang Medical University, Henan Neurology Institute, Weihui, China
| |
Collapse
|
40
|
Patel BS, Ravix J, Pabelick C, Prakash YS. Class C GPCRs in the airway. Curr Opin Pharmacol 2020; 51:19-28. [PMID: 32375079 DOI: 10.1016/j.coph.2020.04.002] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2020] [Revised: 03/17/2020] [Accepted: 04/02/2020] [Indexed: 02/07/2023]
Abstract
Understanding and targeting of GPCRs remain a critical aspect of airway pharmacology and therapeutics for diseases such as asthma or COPD. Most attention has been on the large Class A GPCRs towards improved bronchodilation and blunting of remodeling. Better known in the central or peripheral nervous system, there is increasing evidence that Class C GPCRs which include metabotropic glutamate and GABA receptors, the calcium sensing receptor, sweet/umami taste receptors and a number of orphan receptors, can contribute to airway structure and function. In this review, we will summarize current state of knowledge regarding the pharmacology of Class C GPCRs, their expression and potential functions in the airways, and the application of pharmacological agents targeting this group in the context of airway diseases.
Collapse
Affiliation(s)
- Brijeshkumar S Patel
- Department of Anesthesiologyand Perioperative Medicine, Mayo Clinic, Rochester, MN, United States
| | - Jovanka Ravix
- Department of Anesthesiologyand Perioperative Medicine, Mayo Clinic, Rochester, MN, United States
| | - Christina Pabelick
- Department of Anesthesiologyand Perioperative Medicine, Mayo Clinic, Rochester, MN, United States; Department of Physiology and Biomedical Engineering, Mayo Clinic, Rochester, MN, United States
| | - Y S Prakash
- Department of Anesthesiologyand Perioperative Medicine, Mayo Clinic, Rochester, MN, United States; Department of Physiology and Biomedical Engineering, Mayo Clinic, Rochester, MN, United States.
| |
Collapse
|
41
|
Eppler HB, Jewell CM. Biomaterials as Tools to Decode Immunity. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2020; 32:e1903367. [PMID: 31782844 PMCID: PMC7124992 DOI: 10.1002/adma.201903367] [Citation(s) in RCA: 31] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/27/2019] [Revised: 09/23/2019] [Indexed: 05/02/2023]
Abstract
The immune system has remarkable capabilities to combat disease with exquisite selectivity. This feature has enabled vaccines that provide protection for decades and, more recently, advances in immunotherapies that can cure some cancers. Greater control over how immune signals are presented, delivered, and processed will help drive even more powerful options that are also safe. Such advances will be underpinned by new tools that probe how immune signals are integrated by immune cells and tissues. Biomaterials are valuable resources to support this goal, offering robust, tunable properties. The growing role of biomaterials as tools to dissect immune function in fundamental and translational contexts is highlighted. These technologies can serve as tools to understand the immune system across molecular, cellular, and tissue length scales. A common theme is exploiting biomaterial features to rationally direct how specific immune cells or organs encounter a signal. This precision strategy, enabled by distinct material properties, allows isolation of immunological parameters or processes in a way that is challenging with conventional approaches. The utility of these capabilities is demonstrated through examples in vaccines for infectious disease and cancer immunotherapy, as well as settings of immune regulation that include autoimmunity and transplantation.
Collapse
Affiliation(s)
- Haleigh B Eppler
- Fischell Department of Bioengineering, 8278 Paint Brach Drive, College Park, MD, 20742, USA
- Biological Sciences Training Program, 1247 Biology Psychology Building, College Park, MD, 20742, USA
| | - Christopher M Jewell
- Fischell Department of Bioengineering, 8278 Paint Brach Drive, College Park, MD, 20742, USA
- Biological Sciences Training Program, 1247 Biology Psychology Building, College Park, MD, 20742, USA
- Robert E. Fischell Institute for Biomedical Devices, 8278 Paint Branch Drive, College Park, MD, 20742, USA
- United States Department of Veterans Affairs, VA Maryland Health Care System, 10. N Green Street, Baltimore, MD, 21201, USA
- Department of Microbiology and Immunology, University of Maryland Medical School, 685 West Baltimore Street, HSF-I Suite 380, Baltimore, MD, 21201, USA
- Marlene and Stewart Greenebaum Cancer Center, 22 South Greene Street, Baltimore, MD, 21201, USA
| |
Collapse
|
42
|
Glutamate receptors in domestication and modern human evolution. Neurosci Biobehav Rev 2020; 108:341-357. [DOI: 10.1016/j.neubiorev.2019.10.004] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2019] [Revised: 09/28/2019] [Accepted: 10/07/2019] [Indexed: 02/08/2023]
|
43
|
Zhang C, Jiang M, Wang WQ, Zhao SJ, Yin YX, Mi QJ, Yang MF, Song YQ, Sun BL, Zhang ZY. Selective mGluR1 Negative Allosteric Modulator Reduces Blood-Brain Barrier Permeability and Cerebral Edema After Experimental Subarachnoid Hemorrhage. Transl Stroke Res 2019; 11:799-811. [PMID: 31833035 DOI: 10.1007/s12975-019-00758-z] [Citation(s) in RCA: 43] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2019] [Revised: 10/21/2019] [Accepted: 11/19/2019] [Indexed: 12/14/2022]
Abstract
The blood-brain barrier (BBB) disruption leads to the vasogenic brain edema and contributes to the early brain injury (EBI) after subarachnoid hemorrhage (SAH). However, the mechanisms underlying the BBB damage following SAH are poorly understood. Here we reported that the neurotransmitter glutamate of cerebrospinal fluid (CSF) was dramatically increased in SAH patients with symptoms of cerebral edema. Using the rat SAH model, we found that SAH caused the increase of CSF glutamate level and BBB permeability in EBI, intracerebroventricular injection of exogenous glutamate deteriorated BBB damage and cerebral edema, while intraperitoneally injection of metabotropic glutamate receptor 1(mGluR1) negative allosteric modulator JNJ16259685 significantly attenuated SAH-induced BBB damage and cerebral edema. In an in vitro BBB model, we showed that glutamate increased monolayer permeability of human brain microvascular endothelial cells (HBMEC), whereas JNJ16259685 preserved glutamate-damaged BBB integrity in HBMEC. Mechanically, glutamate downregulated the level and phosphorylation of vasodilator-stimulated phosphoprotein (VASP), decreased the tight junction protein occludin, and increased AQP4 expression at 72 h after SAH. However, JNJ16259685 significantly increased VASP, p-VASP, and occludin, and reduced AQP level at 72 h after SAH. Altogether, our results suggest an important role of glutamate in disruption of BBB function and inhibition of mGluR1 with JNJ16259685 reduced BBB damage and cerebral edema after SAH.
Collapse
Affiliation(s)
- Cheng Zhang
- Key Lab of Cerebral Microcirculation of Shandong, First Medical University & Shandong Academy of Medical Sciences, Tai'an, 271016, Shandong, People's Republic of China
| | - Ming Jiang
- Department of Neurology, Shanghai Tongji Hospital, Tongji University School of Medicine, Shanghai, 200065, People's Republic of China
| | - Wei-Qi Wang
- Key Lab of Cerebral Microcirculation of Shandong, First Medical University & Shandong Academy of Medical Sciences, Tai'an, 271016, Shandong, People's Republic of China.,Medical College of Qingdao University, Qingdao, 266021, Shandong, People's Republic of China
| | - Shi-Jun Zhao
- Department of Neurology, Baotou Central Hospital, Baotou, 014040, People's Republic of China
| | - Yan-Xin Yin
- Department of Neurology, Shanghai Tongji Hospital, Tongji University School of Medicine, Shanghai, 200065, People's Republic of China
| | - Qiong-Jie Mi
- Key Lab of Cerebral Microcirculation of Shandong, First Medical University & Shandong Academy of Medical Sciences, Tai'an, 271016, Shandong, People's Republic of China
| | - Ming-Feng Yang
- Key Lab of Cerebral Microcirculation of Shandong, First Medical University & Shandong Academy of Medical Sciences, Tai'an, 271016, Shandong, People's Republic of China
| | - Yu-Qiang Song
- Department of Neurology, The Affiliated Hospital of Qingdao University, Qingdao, 266003, Shandong, People's Republic of China
| | - Bao-Liang Sun
- Key Lab of Cerebral Microcirculation of Shandong, First Medical University & Shandong Academy of Medical Sciences, Tai'an, 271016, Shandong, People's Republic of China.
| | - Zong-Yong Zhang
- Key Lab of Cerebral Microcirculation of Shandong, First Medical University & Shandong Academy of Medical Sciences, Tai'an, 271016, Shandong, People's Republic of China.
| |
Collapse
|
44
|
Wu H, Gao S, Terakawa S. Inhibitory effects of fucoidan on NMDA receptors and l-type Ca 2+ channels regulating the Ca 2+ responses in rat neurons. PHARMACEUTICAL BIOLOGY 2019; 57:1-7. [PMID: 30734636 PMCID: PMC6374951 DOI: 10.1080/13880209.2018.1548626] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 08/20/2018] [Revised: 10/19/2018] [Accepted: 11/09/2018] [Indexed: 06/09/2023]
Abstract
CONTEXT Fucoidan, a sulphated polysaccharide extracted from brown algae [Fucus vesiculosus Linn. (Fucaceae)], has multiple biological activities. OBJECTIVE The effects of fucoidan on Ca2+ responses of rat neurons and its probable mechanisms with focus on glutamate receptors were examined. MATERIALS AND METHODS The neurons isolated from the cortex and hippocampi of Wistar rats in postnatal day 1 were employed. The intracellular Ca2+ responses triggered by various stimuli were measured in vitro by Fura-2/AM. Fucoidan at 0.5 mg/mL or 1.5 mg/mL was applied for 3 min to determine its effects on Ca2+ responses. RT-PCR was used to determine the mRNA expression of neuron receptors treated with fucoidan at 0.5 mg/mL for 3 h. RESULTS The Ca2+ responses induced by NMDA were 100% suppressed by fucoidan, and those induced by Bay K8644 90% in the cortical neurons. However, fucoidan has no significant effect on the Ca2+ responses of cortical neurons induced by AMPA or quisqualate. Meanwhile, the Ca2+ responses of hippocampal neurons induced by glutamate, ACPD or adrenaline, showed only a slight decrease following fucoidan treatment. RT-PCR assays of cortical and hippocampal neurons showed that fucoidan treatment significantly decreased the mRNA expression of NMDA-NR1 receptor and the primer pair for l-type Ca2+ channels, PR1/PR2. DISCUSSION AND CONCLUSIONS Our data indicate that fucoidan suppresses the intracellular Ca2+ responses by selectively inhibiting NMDA receptors in cortical neurons and l-type Ca2+ channels in hippocampal neurons. A wide spectrum of fucoidan binding to cell membrane may be useful for designing a general purpose drug in future.
Collapse
MESH Headings
- 3-Pyridinecarboxylic acid, 1,4-dihydro-2,6-dimethyl-5-nitro-4-(2-(trifluoromethyl)phenyl)-, Methyl ester/pharmacology
- Animals
- Calcium/metabolism
- Calcium Channels, L-Type/metabolism
- Cells, Cultured
- Cerebellar Cortex/cytology
- Cerebellar Cortex/drug effects
- Excitatory Amino Acid Agonists/pharmacology
- Glutamic Acid/pharmacology
- Hippocampus/cytology
- Hippocampus/drug effects
- N-Methylaspartate/pharmacology
- Neurons/drug effects
- Neurons/metabolism
- Polysaccharides/pharmacology
- Rats
- Rats, Wistar
- Receptors, AMPA/metabolism
- Receptors, Glutamate/metabolism
- Receptors, N-Methyl-D-Aspartate/antagonists & inhibitors
- Receptors, N-Methyl-D-Aspartate/biosynthesis
- Receptors, N-Methyl-D-Aspartate/metabolism
Collapse
Affiliation(s)
- Hong Wu
- Laboratory of Cell Imaging, Henan University of Chinese Medicine, Zhengzhou, China;
- Photon Medical Research Center, Hamamatsu University School of Medicine, Hamamatsu, Japan
| | - Shuibo Gao
- Laboratory of Cell Imaging, Henan University of Chinese Medicine, Zhengzhou, China;
| | - Susumu Terakawa
- Photon Medical Research Center, Hamamatsu University School of Medicine, Hamamatsu, Japan
| |
Collapse
|
45
|
Choi WM, Kim HH, Kim MH, Cinar R, Yi HS, Eun HS, Kim SH, Choi YJ, Lee YS, Kim SY, Seo W, Lee JH, Shim YR, Kim YE, Yang K, Ryu T, Hwang JH, Lee CH, Choi HS, Gao B, Kim W, Kim SK, Kunos G, Jeong WI. Glutamate Signaling in Hepatic Stellate Cells Drives Alcoholic Steatosis. Cell Metab 2019; 30:877-889.e7. [PMID: 31474565 PMCID: PMC6834910 DOI: 10.1016/j.cmet.2019.08.001] [Citation(s) in RCA: 75] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/18/2019] [Revised: 06/15/2019] [Accepted: 08/01/2019] [Indexed: 12/12/2022]
Abstract
Activation of hepatocyte cannabinoid receptor-1 (CB1R) by hepatic stellate cell (HSC)-derived 2-arachidonoylglycerol (2-AG) drives de novo lipogenesis in alcoholic liver disease (ALD). How alcohol stimulates 2-AG production in HSCs is unknown. Here, we report that chronic alcohol consumption induced hepatic cysteine deficiency and subsequent glutathione depletion by impaired transsulfuration pathway. A compensatory increase in hepatic cystine-glutamate anti-porter xCT boosted extracellular glutamate levels coupled to cystine uptake both in mice and in patients with ALD. Alcohol also induced the selective expression of metabotropic glutamate receptor-5 (mGluR5) in HSCs where mGluR5 activation stimulated 2-AG production. Consistently, genetic or pharmacologic inhibition of mGluR5 or xCT attenuated alcoholic steatosis in mice via the suppression of 2-AG production and subsequent CB1R-mediated de novo lipogenesis. We conclude that a bidirectional signaling operates at a metabolic synapse between hepatocytes and HSCs through xCT-mediated glutamate-mGluR5 signaling to produce 2-AG, which induces CB1R-mediated alcoholic steatosis.
Collapse
Affiliation(s)
- Won-Mook Choi
- Laboratory of Liver Research, Graduate School of Medical Science and Engineering, KAIST, Daejeon 34141, Republic of Korea; Department of Gastroenterology, Liver Center, Asan Medical Center, University of Ulsan College of Medicine, Seoul 05505, Republic of Korea
| | - Hee-Hoon Kim
- Laboratory of Liver Research, Graduate School of Medical Science and Engineering, KAIST, Daejeon 34141, Republic of Korea
| | - Myung-Ho Kim
- Laboratory of Liver Research, Graduate School of Medical Science and Engineering, KAIST, Daejeon 34141, Republic of Korea
| | - Resat Cinar
- Laboratory of Physiologic Studies, National Institute on Alcohol Abuse and Alcoholism, National Institutes of Health, Bethesda, MD 20892, USA
| | - Hyon-Seung Yi
- Laboratory of Liver Research, Graduate School of Medical Science and Engineering, KAIST, Daejeon 34141, Republic of Korea; Department of Internal Medicine, Chungnam National University, School of Medicine, Daejeon 35015, Republic of Korea
| | - Hyuk Soo Eun
- Laboratory of Liver Research, Graduate School of Medical Science and Engineering, KAIST, Daejeon 34141, Republic of Korea; Department of Internal Medicine, Chungnam National University, School of Medicine, Daejeon 35015, Republic of Korea
| | - Seok-Hwan Kim
- Department of Surgery, Chungnam National University, College of Medicine, Daejeon 35015, Republic of Korea
| | - Young Jae Choi
- College of Pharmacy, Chungnam National University, Daejeon 34134, Republic of Korea
| | - Young-Sun Lee
- Laboratory of Liver Research, Graduate School of Medical Science and Engineering, KAIST, Daejeon 34141, Republic of Korea; Department of Internal Medicine, Korea University College of Medicine, Seoul 08308, Republic of Korea
| | - So Yeon Kim
- Laboratory of Liver Research, Graduate School of Medical Science and Engineering, KAIST, Daejeon 34141, Republic of Korea
| | - Wonhyo Seo
- Laboratory of Liver Research, Graduate School of Medical Science and Engineering, KAIST, Daejeon 34141, Republic of Korea; Laboratory of Liver Diseases, National Institute on Alcohol Abuse and Alcoholism, National Institutes of Health, Bethesda, MD 20892, USA
| | - Jun-Hee Lee
- Laboratory of Liver Research, Graduate School of Medical Science and Engineering, KAIST, Daejeon 34141, Republic of Korea
| | - Young-Ri Shim
- Laboratory of Liver Research, Graduate School of Medical Science and Engineering, KAIST, Daejeon 34141, Republic of Korea
| | - Ye Eun Kim
- Laboratory of Liver Research, Graduate School of Medical Science and Engineering, KAIST, Daejeon 34141, Republic of Korea
| | - Keungmo Yang
- Laboratory of Liver Research, Graduate School of Medical Science and Engineering, KAIST, Daejeon 34141, Republic of Korea
| | - Tom Ryu
- Laboratory of Liver Research, Graduate School of Medical Science and Engineering, KAIST, Daejeon 34141, Republic of Korea
| | - Jung Hwan Hwang
- Laboratory Animal Resource Center, Korea Research Institute of Bioscience and Biotechnology, Daejeon 34141, Republic of Korea
| | - Chul-Ho Lee
- Laboratory Animal Resource Center, Korea Research Institute of Bioscience and Biotechnology, Daejeon 34141, Republic of Korea
| | - Hueng-Sik Choi
- School of the Biological Sciences and Technology, Chonnam National University, Gwangju 61186, Republic of Korea
| | - Bin Gao
- Laboratory of Liver Diseases, National Institute on Alcohol Abuse and Alcoholism, National Institutes of Health, Bethesda, MD 20892, USA
| | - Won Kim
- Department of Internal Medicine, Seoul Metropolitan Government, Seoul National University Boramae Medical Center, Seoul 07061, Republic of Korea
| | - Sang Kyum Kim
- College of Pharmacy, Chungnam National University, Daejeon 34134, Republic of Korea
| | - George Kunos
- Laboratory of Physiologic Studies, National Institute on Alcohol Abuse and Alcoholism, National Institutes of Health, Bethesda, MD 20892, USA.
| | - Won-Il Jeong
- Laboratory of Liver Research, Graduate School of Medical Science and Engineering, KAIST, Daejeon 34141, Republic of Korea.
| |
Collapse
|
46
|
Yi H, Talmon G, Wang J. Glutamate in cancers: from metabolism to signaling. J Biomed Res 2019; 34:260-270. [PMID: 32594024 PMCID: PMC7386414 DOI: 10.7555/jbr.34.20190037] [Citation(s) in RCA: 39] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2019] [Accepted: 08/20/2019] [Indexed: 01/31/2023] Open
Abstract
Glutamine and glutamate are major bioenergy substrates for normal and cancer cell growth. Cancer cells need more biofuel than normal tissues for energy supply, anti-oxidation activity and biomass production. Genes related to metabolic chains in many cancers are somehow mutated, which makes cancer cells more glutamate dependent. Meanwhile, glutamate is an excitatory neurotransmitter for conducting signals through binding with different types of receptors in central neuron system. Interestingly, increasing evidences have shown involvement of glutamate signaling, guided through their receptors, in human malignancy. Dysregulation of glutamate transporters, such as excitatory amino acid transporter and cystine/glutamate antiporter system, also generates excessive extracellular glutamate, which in turn, activates glutamate receptors on cancer cells and results in malignant growth. These features make glutamate an attractive target for anti-cancer drug development with some glutamate targeted but blood brain barrier impermeable anti-psychosis drugs under consideration. We discussed the relevant progressions and drawbacks in this field herein.
Collapse
Affiliation(s)
- Haowei Yi
- Department of Genetics, Cell Biology and Anatomy
| | | | - Jing Wang
- Department of Genetics, Cell Biology and Anatomy
- Eppley Institute for Research in Cancer and Allied Diseases, University of Nebraska Medical Center, Omaha, NE 68198, USA
| |
Collapse
|
47
|
León-Navarro DA, Albasanz JL, Martín M. Functional Cross-Talk between Adenosine and Metabotropic Glutamate Receptors. Curr Neuropharmacol 2019; 17:422-437. [PMID: 29663888 PMCID: PMC6520591 DOI: 10.2174/1570159x16666180416093717] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2017] [Revised: 03/19/2018] [Accepted: 04/13/2018] [Indexed: 12/14/2022] Open
Abstract
Abstract: G-protein coupled receptors are transmembrane proteins widely expressed in cells and their transduction pathways are mediated by controlling second messenger levels through different G-protein interactions. Many of these receptors have been described as involved in the physiopathology of neurodegenerative diseases and even considered as potential targets for the design of novel therapeutic strategies. Endogenous and synthetic allosteric and orthosteric selective ligands are able to modulate GPCRs at both gene and protein expression levels and can also modify their physiological function. GPCRs that coexist in the same cells can homo- and heteromerize, therefore, modulating their function. Adenosine receptors are GPCRs which stimulate or inhibit adenylyl cyclase activity through Gi/Gs protein and are involved in the control of neurotransmitter release as glutamate. In turn, metabotropic glutamate receptors are also GPCRs which inhibit adenylyl cyclase or stimulate phospholipase C activities through Gi or Gq proteins, respectively. In recent years, evidence of crosstalk mechanisms be-tween different GPCRs have been described. The aim of the present review was to summarize the described mechanisms of interaction and crosstalking between adenosine and metabotropic glutamate receptors, mainly of group I, in both in vitro and in vivo systems, and their possible use for the design of novel ligands for the treatment of neurodegenerative diseases.
Collapse
Affiliation(s)
- David Agustín León-Navarro
- Departamento de Quimica Inorganica, Organica y Bioquimica. CRIB, Universidad de Castilla-La Mancha, Spain.,Facultad de Ciencias y Tecnologías Químicas, Avenida Camilo José Cela, 10, 13071 Ciudad Real, Spain
| | - José Luis Albasanz
- Departamento de Quimica Inorganica, Organica y Bioquimica. CRIB, Universidad de Castilla-La Mancha, Spain.,Facultad de Ciencias y Tecnologías Químicas, Avenida Camilo José Cela, 10, 13071 Ciudad Real, Spain.,Facultad de Medicina de Ciudad Real, Camino Moledores s/n. 13071 Ciudad Real, Spain
| | - Mairena Martín
- Departamento de Quimica Inorganica, Organica y Bioquimica. CRIB, Universidad de Castilla-La Mancha, Spain.,Facultad de Ciencias y Tecnologías Químicas, Avenida Camilo José Cela, 10, 13071 Ciudad Real, Spain.,Facultad de Medicina de Ciudad Real, Camino Moledores s/n. 13071 Ciudad Real, Spain
| |
Collapse
|
48
|
Liu YW, Zhao L, Zhou M, Wang H, Yang N, Dai SS. Transplantation with mGluR5 deficiency bone marrow displays antidepressant-like effect in C57BL/6J mice. Brain Behav Immun 2019; 79:114-124. [PMID: 30682501 DOI: 10.1016/j.bbi.2019.01.022] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/26/2018] [Revised: 12/17/2018] [Accepted: 01/21/2019] [Indexed: 12/19/2022] Open
Abstract
Antidepressant-like effects of metabotropic glutamate receptor 5 (mGluR5) have been verified by specific antagonists or whole body knock-out (KO) mice. Previous experiments indicate that blocking mGluR5 exerts antidepressant-like effects through neuronal mechanisms, like modulating NMDA receptor activity or 5-HT system. Here we found that transplanting bone marrow from mGluR5 KO mice to WT mice could also show antidepressant-like effects, which were confirmed by sucrose preference test and tail suspension test. Furthermore, mGluR5 deficiency dramatically inhibits cytokines release from bone marrow cells, such as IL-1β, TNF-α and IL-6, alleviating proinflammatory responses in LPS-induced depression model. In addition, inhibited cytokines could decrease the activation of brain endothelial cells in ERK-dependent manner. These data provide the evidence that blocking mGluR5 could improve depression through inhibiting peripheral immune responses, confirming the causal relationship between peripheral immune phenotype and brain behavior.
Collapse
Affiliation(s)
- Yang-Wuyue Liu
- Department of Biochemistry and Molecular Biology, Army Medical University, Chongqing 400038, PR China
| | - Li Zhao
- Department of Biochemistry and Molecular Biology, Army Medical University, Chongqing 400038, PR China
| | - Mi Zhou
- Department of Biochemistry and Molecular Biology, Army Medical University, Chongqing 400038, PR China
| | - Hao Wang
- Department of Neurosurgery, Daping Hospital, Army Medical University, Chongqing 400042, PR China
| | - Nan Yang
- Molecular Biology Center, State Key Laboratory of Trauma, Burn, and Combined Injury, Daping Hospital, Army Medical University, Chongqing 400042, PR China
| | - Shuang-Shuang Dai
- Department of Biochemistry and Molecular Biology, Army Medical University, Chongqing 400038, PR China; Molecular Biology Center, State Key Laboratory of Trauma, Burn, and Combined Injury, Daping Hospital, Army Medical University, Chongqing 400042, PR China.
| |
Collapse
|
49
|
Rigi-Ladiz MA, Baranzehi T, Hassanpour B, Ashraf MJ, Kordi-Tamandani DM. DNA methylation and expression status of glutamate receptor genes in patients with oral squamous cell carcinoma. Meta Gene 2019. [DOI: 10.1016/j.mgene.2019.100545] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/27/2022] Open
|
50
|
Chen ANY, Hellyer SD, Trinh PNH, Leach K, Gregory KJ. Identification of monellin as the first naturally derived proteinaceous allosteric agonist of metabotropic glutamate receptor 5. Basic Clin Pharmacol Toxicol 2019; 126 Suppl 6:104-115. [DOI: 10.1111/bcpt.13239] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2019] [Accepted: 04/10/2019] [Indexed: 12/20/2022]
Affiliation(s)
- Amy N. Y. Chen
- Drug Discovery Biology, Monash Institute of Pharmaceutical Sciences and Department of Pharmacology Monash University Parkville Victoria Australia
| | - Shane D. Hellyer
- Drug Discovery Biology, Monash Institute of Pharmaceutical Sciences and Department of Pharmacology Monash University Parkville Victoria Australia
| | - Phuc N. H. Trinh
- Drug Discovery Biology, Monash Institute of Pharmaceutical Sciences and Department of Pharmacology Monash University Parkville Victoria Australia
| | - Katie Leach
- Drug Discovery Biology, Monash Institute of Pharmaceutical Sciences and Department of Pharmacology Monash University Parkville Victoria Australia
| | - Karen J. Gregory
- Drug Discovery Biology, Monash Institute of Pharmaceutical Sciences and Department of Pharmacology Monash University Parkville Victoria Australia
| |
Collapse
|