1
|
Rashed MS, Abdelkarim EA, Elsamahy T, Sobhy M, El-Mesery HS, Salem A. Advances in cell-based biosensors: Transforming food flavor evaluation with novel approaches. Food Chem X 2025; 26:102336. [PMID: 40115496 PMCID: PMC11923814 DOI: 10.1016/j.fochx.2025.102336] [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: 01/08/2025] [Revised: 02/18/2025] [Accepted: 02/26/2025] [Indexed: 03/23/2025] Open
Abstract
Food flavor, a blend of taste and smell, is key to consumer acceptance and food quality. Traditional sensory and instrumental methods often fail to replicate human sensory responses. This review discusses the role of cell-based biosensors in flavor evaluation, showcasing their sensitivity, specificity, and rapid response. Using living cells like taste and olfactory cells, these biosensors surpass traditional approaches. Advancements include microelectrode array systems with taste receptor cells for real-time detection of bitter, sweet, and umami substances and improved cell immobilization technologies for detecting complex odorant profiles. Challenges such as signal stability, selective detection, cell cultivation, and scalability persist. However, integrating artificial intelligence and portable technologies could broaden their applications. With the potential to revolutionize sensory analysis, cell-based biosensors offer a sustainable, precise, and scalable approach to food flavor evaluation, bridging sensory perception with advanced analytical methods and driving innovation in food science.
Collapse
Affiliation(s)
- Mahmoud Said Rashed
- Food Science and Technology Department, Faculty of Agriculture, Alexandria University, Alexandria 21545, Egypt
| | - Esraa A Abdelkarim
- Food Control Department, Faculty of Veterinary Medicine, Zagazig University, Zagazig 44519, Egypt
| | | | - Mabrouk Sobhy
- Food Science and Technology Department, Faculty of Agriculture, Alexandria University, Alexandria 21545, Egypt
- School of Agricultural Engineering, Jiangsu University, Zhenjiang 212013, China
- School of Food and Biological Engineering, Jiangsu University, Zhenjiang 212013, China
| | - Hany S El-Mesery
- School of Energy and Power Engineering, Jiangsu University, Zhenjiang 212013, China
- Agricultural Engineering Research Institute, Agricultural Research Center, Dokki, 12611 Giza, Egypt
| | - Ali Salem
- Civil Engineering Department, Faculty of Engineering, Minia University, Minia 61111, Egypt
- Structural Diagnostics and Analysis Research Group, Faculty of Engineering and Information Technology, University of Pecs, Hungary
| |
Collapse
|
2
|
Dimiati H, Srikandi M, Haypheng T, Sovira N, Herdata HN, Bakhtiar B, Edward ED. Relationship between G protein level with left ventricular systolic function in children with acyanotic heart disease. NARRA J 2024; 4:e531. [PMID: 39280266 PMCID: PMC11391980 DOI: 10.52225/narra.v4i2.531] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 11/11/2023] [Accepted: 04/29/2024] [Indexed: 09/18/2024]
Abstract
Heart failure is a pediatric emergency caused by the heart's inability to adequately meet the body metabolic needs and the most common cause is congenital heart disease (CHD). The G protein is the most prominent family of membrane-bound protein known to act in major regulatory events of the cardiovascular system, one of which is heart failure. The aim of this study was to determine the level of G protein and its relationship with left ventricular systolic function in children with acyanotic CHD. A cross-sectional study was conducted in Dr. Zaionel Abidin Hospital, Banda Aceh, Indonesia. The patients aged 0 to 18 years and had acyanotic CHD diagnosis by echocardiography were included. Anthropometry measurement was performed according to standard WHO procedures and G protein level was measured using the ELISA method. The Chi-squared test was used to measure the relationship between G protein level and left ventricular systolic function. Out of a total of 38 children with acyanotic CHD, the mean level of G protein was 36.25 ng/mL and the mean of left ventricular systolic function was 73.1%. There was no relationship between G protein and left ventricular systolic function in children with acyanotic CHD. However, further study with a larger sample size and considering other variables are needed to confirm this finding.
Collapse
Affiliation(s)
- Herlina Dimiati
- Division of Cardiology, Department of Pediatrics, Faculty of Medicine, Universitas Syiah Kuala, Banda Aceh, Indonesia
| | - Mira Srikandi
- Department of Pediatrics, Faculty of Medicine, Universitas Syiah Kuala, Banda Aceh, Indonesia
| | - Te Haypheng
- Department of General Pediatric and Infectious Disease, National Pediatric Hospital, Ministry of Health, Phnom Penh, Cambodia
| | - Nora Sovira
- Division of Emergency and Intensive Care, Department of Pediatrics, Faculty of Medicine, Universitas Syiah Kuala, Banda Aceh, Indonesia
| | - Heru N Herdata
- Division of Hemato-oncology, Department of Pediatrics, Faculty of Medicine, Universitas Syiah Kuala, Banda Aceh, Indonesia
| | - Bakhtiar Bakhtiar
- Division of Pulmonology, Department of Pediatrics, Faculty of Medicine, Universitas Syiah Kuala, Banda Aceh, Indonesia
| | - Eka D Edward
- Division of Hemato-oncology, Department of Pediatrics, Faculty of Medicine, Universitas Syiah Kuala, Banda Aceh, Indonesia
| |
Collapse
|
3
|
Chang CC, Chen CH, Hsu SY, Leu S. Cardiomyocyte-specific overexpression of GPR22 ameliorates cardiac injury in mice with acute myocardial infarction. BMC Cardiovasc Disord 2024; 24:287. [PMID: 38816768 PMCID: PMC11138089 DOI: 10.1186/s12872-024-03953-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2024] [Accepted: 05/22/2024] [Indexed: 06/01/2024] Open
Abstract
BACKGROUND The activation of G protein-coupled receptors (GPCR) signaling by external stimuli has been implicated in inducing cardiac stress and stress responses. GPR22 is an orphan GPCR expressed in brains and hearts, while its expression level is associated with cardiovascular damage in diabetes. Previous studies have suggested a protective role of GPR22 in mechanical cardiac stress, as loss of its expression increases susceptibility to heart failure post-ventricular pressure overload. However, the involvement and underlying signaling of GPR22 in cardiac stress response to ischemic stress remains unexplored. METHODS In this study, we used cultured cells and a transgenic mouse model with cardiomyocyte-specific GPR22 overexpression to investigate the impact of ischemic stress on GPR22 expression and to elucidate its role in myocardial ischemic injury. Acute myocardial infarction (AMI) was induced by left coronary artery ligation in eight-week-old male GPR22 transgenic mice, followed by histopathological and biochemical examination four weeks post-AMI induction. RESULTS GPR22 expression in H9C2 and RL-14 cells, two cardiomyocyte cell lines, was decreased by cobalt chloride (CoCl2) treatment. Similarly, reduced expression of myocardial GPR22 was observed in mice with AMI. Histopathological examinations revealed a protective effect of GPR22 overexpression in attenuating myocardial infarction in mice with AMI. Furthermore, myocardial levels of Bcl-2 and activation of PI3K-Akt signaling were downregulated by ischemic stress and upregulated by GPR22 overexpression. Conversely, the expression levels of caspase-3 and phosphorylated ERK1/2 in the infarcted myocardium were downregulated with GPR22 overexpression. CONCLUSION Myocardial ischemic stress downregulates cardiac expression of GPR22, whereas overexpression of GPR22 in cardiomyocytes upregulates Akt signaling, downregulates ERK activation, and mitigates ischemia-induced myocardial injury.
Collapse
Affiliation(s)
- Chin-Chuan Chang
- Department of Nuclear Medicine, Kaohsiung Medical University Hospital, Kaohsiung, 80756, Taiwan
- School of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung, 80756, Taiwan
- Neuroscience Research Center, Kaohsiung Medical University, Kaohsiung, 80756, Taiwan
| | - Chih-Hung Chen
- Division of Hepato-Gastroenterology, Department of Internal Medicine, Kaohsiung Chang Gung Memorial Hospital, Chang Gung University College of Medicine, Kaohsiung, 83301, Taiwan
| | - Shu-Yuan Hsu
- Department of Anatomy, Graduate Institute of Biomedical Sciences, College of Medicine, Chang Gung University, Taoyuan, 33302, Taiwan
- Institute for Translational Research in Biomedicine, Kaohsiung Chang Gung Memorial Hospital, Kaohsiung, 83301, Taiwan
| | - Steve Leu
- Institute for Translational Research in Biomedicine, Kaohsiung Chang Gung Memorial Hospital, Kaohsiung, 83301, Taiwan.
- Department of Biotechnology, College of Life Science, Kaohsiung Medical University, Kaohsiung, 80756, Taiwan.
| |
Collapse
|
4
|
Jobe A, Vijayan R. Orphan G protein-coupled receptors: the ongoing search for a home. Front Pharmacol 2024; 15:1349097. [PMID: 38495099 PMCID: PMC10941346 DOI: 10.3389/fphar.2024.1349097] [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: 12/04/2023] [Accepted: 02/15/2024] [Indexed: 03/19/2024] Open
Abstract
G protein-coupled receptors (GPCRs) make up the largest receptor superfamily, accounting for 4% of protein-coding genes. Despite the prevalence of such transmembrane receptors, a significant number remain orphans, lacking identified endogenous ligands. Since their conception, the reverse pharmacology approach has been used to characterize such receptors. However, the multifaceted and nuanced nature of GPCR signaling poses a great challenge to their pharmacological elucidation. Considering their therapeutic relevance, the search for native orphan GPCR ligands continues. Despite limited structural input in terms of 3D crystallized structures, with advances in machine-learning approaches, there has been great progress with respect to accurate ligand prediction. Though such an approach proves valuable given that ligand scarcity is the greatest hurdle to orphan GPCR deorphanization, the future pairings of the remaining orphan GPCRs may not necessarily take a one-size-fits-all approach but should be more comprehensive in accounting for numerous nuanced possibilities to cover the full spectrum of GPCR signaling.
Collapse
Affiliation(s)
- Amie Jobe
- Department of Biology, College of Science, United Arab Emirates University, Al Ain, United Arab Emirates
| | - Ranjit Vijayan
- Department of Biology, College of Science, United Arab Emirates University, Al Ain, United Arab Emirates
- The Big Data Analytics Center, United Arab Emirates University, Al Ain, United Arab Emirates
- Zayed Bin Sultan Center for Health Sciences, United Arab Emirates University, Al Ain, United Arab Emirates
| |
Collapse
|
5
|
Lin TY, Mai QN, Zhang H, Wilson E, Chien HC, Yee SW, Giacomini KM, Olgin JE, Irannejad R. Cardiac contraction and relaxation are regulated by distinct subcellular cAMP pools. Nat Chem Biol 2024; 20:62-73. [PMID: 37474759 PMCID: PMC10746541 DOI: 10.1038/s41589-023-01381-8] [Citation(s) in RCA: 20] [Impact Index Per Article: 20.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2022] [Accepted: 06/08/2023] [Indexed: 07/22/2023]
Abstract
Cells interpret a variety of signals through G-protein-coupled receptors (GPCRs) and stimulate the generation of second messengers such as cyclic adenosine monophosphate (cAMP). A long-standing puzzle is deciphering how GPCRs elicit different physiological responses despite generating similar levels of cAMP. We previously showed that some GPCRs generate cAMP from both the plasma membrane and the Golgi apparatus. Here we demonstrate that cardiomyocytes distinguish between subcellular cAMP inputs to elicit different physiological outputs. We show that generating cAMP from the Golgi leads to the regulation of a specific protein kinase A (PKA) target that increases the rate of cardiomyocyte relaxation. In contrast, cAMP generation from the plasma membrane activates a different PKA target that increases contractile force. We further validated the physiological consequences of these observations in intact zebrafish and mice. Thus, we demonstrate that the same GPCR acting through the same second messenger regulates cardiac contraction and relaxation dependent on its subcellular location.
Collapse
Affiliation(s)
- Ting-Yu Lin
- Cardiovascular Research Institute, University of California, San Francisco, CA, USA
| | - Quynh N Mai
- Cardiovascular Research Institute, University of California, San Francisco, CA, USA
| | - Hao Zhang
- Cardiovascular Research Institute, University of California, San Francisco, CA, USA
- Department of Medicine, Division of Cardiology, University of California San Francisco, San Francisco, CA, USA
| | - Emily Wilson
- Cardiovascular Research Institute, University of California, San Francisco, CA, USA
- Department of Medicine, Division of Cardiology, University of California San Francisco, San Francisco, CA, USA
| | - Huan-Chieh Chien
- Department of Bioengineering and Therapeutic Sciences, University of California San Francisco, California, CA, USA
| | - Sook Wah Yee
- Department of Bioengineering and Therapeutic Sciences, University of California San Francisco, California, CA, USA
| | - Kathleen M Giacomini
- Department of Bioengineering and Therapeutic Sciences, University of California San Francisco, California, CA, USA
- Institute for Human Genetics, University of California San Francisco, San Francisco, CA, USA
| | - Jeffrey E Olgin
- Cardiovascular Research Institute, University of California, San Francisco, CA, USA
- Department of Medicine, Division of Cardiology, University of California San Francisco, San Francisco, CA, USA
| | - Roshanak Irannejad
- Cardiovascular Research Institute, University of California, San Francisco, CA, USA.
- Department of Biochemistry & Biophysics, University of California, San Francisco, CA, USA.
| |
Collapse
|
6
|
Bloxham CJ, Hulme KD, Fierro F, Fercher C, Pegg CL, O'Brien SL, Foster SR, Short KR, Furness SGB, Reichelt ME, Niv MY, Thomas WG. Cardiac human bitter taste receptors contain naturally occurring variants that alter function. Biochem Pharmacol 2024; 219:115932. [PMID: 37989413 DOI: 10.1016/j.bcp.2023.115932] [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: 07/13/2023] [Revised: 10/26/2023] [Accepted: 11/16/2023] [Indexed: 11/23/2023]
Abstract
Bitter taste receptors (T2R) are a subfamily of G protein-coupled receptors that enable humans to detect aversive and toxic substances. The ability to discern bitter compounds varies between individuals and is attributed mainly to naturally occurring T2R polymorphisms. T2Rs are also expressed in numerous non-gustatory tissues, including the heart, indicating potential contributions to cardiovascular physiology. In this study. T2Rs that have previously been identified in human cardiac tissues (T2Rs - 10, 14, 30, 31, 46 and 50) and their naturally occurring polymorphisms were functionally characterised. The ligand-dependent signaling responses of some T2R variants were completely abolished (T2R30 Leu252 and T2R46 Met228), whereas other receptor variants had moderate changes in their maximal response, but not potency, relative to wild type. Using a cAMP fluorescent biosensor, we reveal the productive coupling of T2R14, but not the T2R14 Phe201 variant, to endogenous Gαi. Modeling revealed that these variants resulted in altered interactions that generally affected ligand binding (T2R30 Leu252) or Gα protein interactions (T2R46 Met228 and T2R14 Phe201), rather than receptor structural stability. Interestingly, this study is the first to show a difference in signaling for T2R50 Tyr203 (rs1376251) which has been associated with cardiovascular disease. The observation of naturally occurring functional variation in the T2Rs with the greatest expression in the heart is important, as their discovery should prove useful in deciphering the role of T2Rs within the cardiovascular system.
Collapse
Affiliation(s)
- Conor J Bloxham
- School of Biomedical Sciences, Faculty of Medicine, University of Queensland, QLD, Australia; Regenerative Medicine in Cardiovascular Diseases, First Department of Medicine, Klinikum rechts der Isar, Technical University of Munich, Germany
| | - Katina D Hulme
- School of Chemistry and Molecular Biosciences, Faculty of Science, University of Queensland, QLD, Australia; Department of Medical Microbiology, Academic Medical Center, University of Amsterdam, Amsterdam, Netherlands
| | - Fabrizio Fierro
- Institute of Biochemistry, Food Science and Nutrition, The Hebrew University of Jerusalem, Israel
| | - Christian Fercher
- Australian Institute for Bioengineering and Nanotechnology, University of Queensland, QLD, Australia
| | - Cassandra L Pegg
- School of Chemistry and Molecular Biosciences, Faculty of Science, University of Queensland, QLD, Australia
| | - Shannon L O'Brien
- Institute of Metabolism and Systems Research, University of Birmingham, United Kingdom; Centre of Membrane Proteins and Receptors (COMPARE), Universities of Nottingham and Birmingham, Birmingham, United Kingdom
| | - Simon R Foster
- School of Biomedical Sciences, Faculty of Medicine, University of Queensland, QLD, Australia; QIMR Berghofer Medical Research Institute, Herston, QLD, Australia
| | - Kirsty R Short
- School of Chemistry and Molecular Biosciences, Faculty of Science, University of Queensland, QLD, Australia
| | - Sebastian G B Furness
- School of Biomedical Sciences, Faculty of Medicine, University of Queensland, QLD, Australia; Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, VIC, Australia
| | - Melissa E Reichelt
- School of Biomedical Sciences, Faculty of Medicine, University of Queensland, QLD, Australia
| | - Masha Y Niv
- Institute of Biochemistry, Food Science and Nutrition, The Hebrew University of Jerusalem, Israel
| | - Walter G Thomas
- School of Biomedical Sciences, Faculty of Medicine, University of Queensland, QLD, Australia.
| |
Collapse
|
7
|
Chda A, Bencheikh R. Flavonoids as G Protein-coupled Receptors Ligands: New Potential Therapeutic Natural Drugs. Curr Drug Targets 2023; 24:1346-1363. [PMID: 38037994 DOI: 10.2174/0113894501268871231127105219] [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: 06/24/2023] [Revised: 10/18/2023] [Accepted: 10/25/2023] [Indexed: 12/02/2023]
Abstract
G protein coupled receptors (GPCRs) are among the largest family of cell surface receptors found in the human genome. They govern a wide range of physiological responses in both health and diseases, making them one of the potential targeted surface receptors for pharmaceuticals. Flavonoids can modulate GPCRs activity by acting as allosteric ligands. They can either enhance or reduce the GPCR's effect. Emerging research shows that individual flavonoids or mixtures of flavonoids from plant extracts can have relevant pharmacological effects against a number of diseases, particularly by influencing GPCRs. In the present review, we are considering to give a comprehensive overview of flavonoids and related compounds that exhibit GPCRs activity and to further explore which beneficial structural features. Molecular docking was used to strengthen experimental evidence and describe flavonoid-GPCRs interactions at molecular level.
Collapse
Affiliation(s)
- Alae Chda
- Laboratory of Microbial Biotechnology and Bioactive Molecules (LM2BM), Faculty of Science and Technology, Sidi Mohamed Ben Abdellah University, Road of Immouzer, PO Box 2202, Fez, Morocco
- Higher Institute of Nursing and Health Techniques - Fez. Ministry of Health and Social Protection, Fez, Morocco
| | - Rachid Bencheikh
- Laboratory of Microbial Biotechnology and Bioactive Molecules (LM2BM), Faculty of Science and Technology, Sidi Mohamed Ben Abdellah University, Road of Immouzer, PO Box 2202, Fez, Morocco
| |
Collapse
|
8
|
CXCR7 ameliorates myocardial infarction as a β-arrestin-biased receptor. Sci Rep 2021; 11:3426. [PMID: 33564089 PMCID: PMC7873251 DOI: 10.1038/s41598-021-83022-5] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2020] [Accepted: 01/28/2021] [Indexed: 02/08/2023] Open
Abstract
Most seven transmembrane receptors (7TMRs) are G protein-coupled receptors; however, some 7TMRs evoke intracellular signals through β-arrestin as a biased receptor. As several β-arrestin-biased agonists have been reported to be cardioprotective, we examined the role of the chemokine receptor CXCR7 as a β-arrestin-biased receptor in the heart. Among 510 7TMR genes examined, Cxcr7 was the most abundantly expressed in the murine heart. Single-cell RNA-sequencing analysis revealed that Cxcr7 was abundantly expressed in cardiomyocytes and fibroblasts. Cardiomyocyte-specific Cxcr7 null mice showed more prominent cardiac dilatation and dysfunction than control mice 4 weeks after myocardial infarction. In contrast, there was no difference in cardiac phenotypes between fibroblast-specific Cxcr7-knockout mice and control mice even after myocardial infarction. TC14012, a specific agonist of CXCR7, significantly recruited β-arrestin to CXCR7 in CXCR7-expressing cells and activated extracellular signal-regulated kinase (ERK) in neonatal rat cardiomyocytes. Cxcr7 expression was significantly increased and ERK was activated in the border zone of the heart in control, but not Cxcr7 null mice. These results indicate that the abundantly expressed CXCR7 in cardiomyocytes may play a protective role in the heart as a β-arrestin-biased receptor and that CXCR7 may be a novel therapeutic target for myocardial infarction.
Collapse
|
9
|
Li X, Cai H, Ren X, He J, Tang J, Xie P, Wang N, Nie F, Lei L, Wang C, Li W, Ma J. Sandstorm weather is a risk factor for mortality in ischemic heart disease patients in the Hexi Corridor, northwestern China. ENVIRONMENTAL SCIENCE AND POLLUTION RESEARCH INTERNATIONAL 2020; 27:34099-34106. [PMID: 32557065 DOI: 10.1007/s11356-020-09616-0] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/10/2019] [Accepted: 06/04/2020] [Indexed: 06/11/2023]
Abstract
Ischemic heart disease (IHD) is one of the leading causes of mortality worldwide. Moreover, the effects of air pollution have been associated with several cardiovascular diseases (CVDs). The relationship between sandstorm weather and IHD is unknown. The Hexi Corridor is located in northwestern China and is a typical desert region comprising a large area of desert with a high incidence of sandstorms. This study aimed to explore the association between sandstorm weather and IHD-related mortality in this area. We acquired meteorological data of sandstorm weather from 2006 to 2015 from the Gansu Meteorological Bureau, and data regarding deaths due to IHD in five cities within the Hexi Corridor were collected from the death registration system of the Center for Disease Control of Gansu during the same period. Two other cities with few sandstorm events were selected as control regions. The time series method of the generalized additive model (GAM) was used to assess the association between sandstorm weather and IHD-related mortality in the Hexi Corridor. The results showed that the frequency of sandstorms in the Hexi Corridor was higher than that in the control regions (5.48% vs 1.64%, P < 0.01), and IHD-related mortality was correspondingly higher than that in the control regions (56.42/100,000 vs 45.62/100,000, P < 0.01). After stratification by gender, age, and urban/rural residence, a significant difference in IHD-related mortality was also noted (P < 0.05). Significant associations were found between sandstorm weather and IHD-related mortality, and the relative risk (RR) increased with an increasing number of days of sandstorm weather. According to the monthly and annual analyses, the mortality rate corresponded to sandstorm frequency. Our data suggest a positive association between sandstorm weather and IHD-related mortality in the Hexi Corridor of Gansu Province. The underlying mechanism requires further study.
Collapse
Affiliation(s)
- Xinghui Li
- Department of Cardiology, Gansu Provincial Hospital, Lanzhou, 730000, China
| | - Hui Cai
- Gansu Key Laboratory of Molecular Diagnostics and Precision Medicine for Surgical Oncology, Gansu Provincial Hospital, Lanzhou, 730000, China
| | - Xiaolan Ren
- Department of Prevention and Control of Chronic Non-communicable Diseases, Gansu Provincial Center for Disease Control and Prevention, Lanzhou, 730000, China
| | - Jin He
- Department of Cardiology, Gansu Provincial Hospital, Lanzhou, 730000, China
| | - Jia Tang
- Department of Infectious Diseases, Huashan Hospital of Fudan University, Shanghai, 200041, China
| | - Ping Xie
- Department of Cardiology, Gansu Provincial Hospital, Lanzhou, 730000, China
| | - Nan Wang
- Department of Cardiology, Gansu Provincial Hospital, Lanzhou, 730000, China
| | - Fangfei Nie
- Department of Cardiology, Gansu Provincial Hospital, Lanzhou, 730000, China
| | - Linfeng Lei
- Department of Cardiology, Gansu Provincial Hospital, Lanzhou, 730000, China
| | - Chenchen Wang
- Department of Cardiology, Gansu Provincial Hospital, Lanzhou, 730000, China
| | - Wenli Li
- Central Meteorological Station of Gansu Meteorological Bureau, Lanzhou, 730000, China
| | - Jing Ma
- Department of Endocrinology, Gansu Provincial Hospital, No. 204 Donggang West Road, Lanzhou, 730000, Gansu, China.
| |
Collapse
|
10
|
Bloxham CJ, Foster SR, Thomas WG. A Bitter Taste in Your Heart. Front Physiol 2020; 11:431. [PMID: 32457649 PMCID: PMC7225360 DOI: 10.3389/fphys.2020.00431] [Citation(s) in RCA: 28] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2020] [Accepted: 04/08/2020] [Indexed: 12/11/2022] Open
Abstract
The human genome contains ∼29 bitter taste receptors (T2Rs), which are responsible for detecting thousands of bitter ligands, including toxic and aversive compounds. This sentinel function varies between individuals and is underpinned by naturally occurring T2R polymorphisms, which have also been associated with disease. Recent studies have reported the expression of T2Rs and their downstream signaling components within non-gustatory tissues, including the heart. Though the precise role of T2Rs in the heart remains unclear, evidence points toward a role in cardiac contractility and overall vascular tone. In this review, we summarize the extra-oral expression of T2Rs, focusing on evidence for expression in heart; we speculate on the range of potential ligands that may activate them; we define the possible signaling pathways they activate; and we argue that their discovery in heart predicts an, as yet, unappreciated cardiac physiology.
Collapse
Affiliation(s)
- Conor J Bloxham
- School of Biomedical Sciences, Faculty of Medicine, The University of Queensland, St Lucia, QLD, Australia
| | - Simon R Foster
- Department of Biochemistry and Molecular Biology, Monash Biomedicine Discovery Institute, Monash University, Clayton, VIC, Australia
| | - Walter G Thomas
- School of Biomedical Sciences, Faculty of Medicine, The University of Queensland, St Lucia, QLD, Australia
| |
Collapse
|
11
|
Nath AK, Ma J, Chen ZZ, Li Z, Vitery MDC, Kelley ML, Peterson RT, Gerszten RE, Yeh JRJ. Genetic deletion of gpr27 alters acylcarnitine metabolism, insulin sensitivity, and glucose homeostasis in zebrafish. FASEB J 2019; 34:1546-1557. [PMID: 31914600 DOI: 10.1096/fj.201901466r] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2019] [Revised: 10/31/2019] [Accepted: 11/11/2019] [Indexed: 02/06/2023]
Abstract
G protein-coupled receptors (GPCRs) comprise the largest group of membrane receptors in eukaryotic genomes and collectively they regulate nearly all cellular processes. Despite the widely recognized importance of this class of proteins, many GPCRs remain understudied. G protein-coupled receptor 27 (Gpr27) is an orphan GPCR that displays high conservation during vertebrate evolution. Although, GPR27 is known to be expressed in tissues that regulate metabolism including the pancreas, skeletal muscle, and adipose tissue, its functions are poorly characterized. Therefore, to investigate the potential roles of Gpr27 in energy metabolism, we generated a whole body gpr27 knockout zebrafish line. Loss of gpr27 potentiated the elevation in glucose levels induced by pharmacological or nutritional perturbations. We next leveraged a mass spectrometry metabolite profiling platform to identify other potential metabolic functions of Gpr27. Notably, genetic deletion of gpr27 elevated medium-chain acylcarnitines, in particular C6-hexanoylcarnitine, C8-octanoylcarnitine, C9-nonanoylcarnitine, and C10-decanoylcarnitine, lipid species known to be associated with insulin resistance in humans. Concordantly, gpr27 deletion in zebrafish abrogated insulin-dependent Akt phosphorylation and glucose utilization. Finally, loss of gpr27 increased the expression of key enzymes in carnitine shuttle complex, in particular the homolog to the brain-specific isoform of CPT1C which functions as a hypothalamic energy senor. In summary, our findings shed light on the biochemical functions of Gpr27 by illuminating its role in lipid metabolism, insulin signaling, and glucose homeostasis.
Collapse
Affiliation(s)
- Anjali K Nath
- Division of Cardiovascular Medicine, Beth Israel Deaconess Medical Center, Boston, MA, USA.,Broad Institute, Cambridge, MA, USA
| | - Junyan Ma
- Cardiovascular Research Center, Massachusetts General Hospital, Charlestown, MA, USA.,Department of Medicine, Harvard Medical School, Boston, MA, USA
| | - Zsu-Zsu Chen
- Division of Cardiovascular Medicine, Beth Israel Deaconess Medical Center, Boston, MA, USA
| | - Zhuyun Li
- Cardiovascular Research Center, Massachusetts General Hospital, Charlestown, MA, USA
| | | | - Michelle L Kelley
- Cardiovascular Research Center, Massachusetts General Hospital, Charlestown, MA, USA
| | | | - Robert E Gerszten
- Division of Cardiovascular Medicine, Beth Israel Deaconess Medical Center, Boston, MA, USA.,Broad Institute, Cambridge, MA, USA
| | - Jing-Ruey J Yeh
- Cardiovascular Research Center, Massachusetts General Hospital, Charlestown, MA, USA.,Department of Medicine, Harvard Medical School, Boston, MA, USA
| |
Collapse
|
12
|
Serebryakova LI, Studneva IM, Ovchinnikov MV, Veselova OM, Molokoedov AS, Arzamastsev EV, Afanasyeva EY, Terekhova OA, Sidorova MV, Pisarenko OI. [Cardiometabolic efficacy and toxicological evaluation of a pharmacological galanin receptor agonist]. BIOMEDIT︠S︡INSKAI︠A︡ KHIMII︠A︡ 2019; 65:231-238. [PMID: 31258147 DOI: 10.18097/pbmc20196503231] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
Abstract
The goal of this study was to examine effects of a novel galanin receptor agonist GalR1-3 [bAla14, His15]-galanine 2-15 (G), obtained by automatic solid-phase synthesis, on the metabolic state of the area at risk and the size of acute myocardial infarction (MI) in rats in vivo and evaluate its toxicity in BALB /c mice. In anesthetized rats, regional ischemia was simulated by coronary artery occlusion and then coronary blood flow was restored. The peptide G was administered intravenously (i.v.) with a bolus after a period of regional ischemia in the dose range of 0.25-3.0 mg/kg. The sizes of MI and the activities of creatine kinase-MB (СK-MB) and lactate dehydrogenase (LDH) in blood plasma were estimated. The effect of administration of the optimal dose of G (1.0 mg/kg) on myocardial content of adenine nucleotides (AN), phosphocreatine (PCr), creatine (Cr) and lactate was studied. I.v. administration of G to rats at a dose of 1.0 mg/kg slightly affected hemodynamic parameters, but reduced MI size by 40% and decreased plasma LDH and CK-MB activity by the end of reperfusion compared to control. These effects were accompanied by a significant improvement in energy state of area at risk (AAR) - an increase in myocardial content of ATP, åAN, PCr and åCr, and combined with a decrease in myocardial lactate level compared with the control. Toxicity of peptide G was studied with a single intraperitoneal injection of 0.5-3.0% solution of the peptide substance to mice. The absence of signs of intoxication and death of animals after G injection in the maximum possible dose did not allow determining the value of the average lethal dose. The results indicate therapeutic potential of the peptide G for preventing myocardial ischemia and reperfusion injury and feasibility for further study of its pharmacological properties and mechanisms of action.
Collapse
Affiliation(s)
| | - I M Studneva
- National Medical Research Center for Cardiology, Moscow, Russia
| | - M V Ovchinnikov
- National Medical Research Center for Cardiology, Moscow, Russia
| | - O M Veselova
- National Medical Research Center for Cardiology, Moscow, Russia
| | - A S Molokoedov
- National Medical Research Center for Cardiology, Moscow, Russia
| | - E V Arzamastsev
- National Medical Research Center for Cardiology, Moscow, Russia
| | - E Yu Afanasyeva
- National Medical Research Center for Cardiology, Moscow, Russia
| | - O A Terekhova
- National Medical Research Center for Cardiology, Moscow, Russia
| | - M V Sidorova
- National Medical Research Center for Cardiology, Moscow, Russia
| | - O I Pisarenko
- National Medical Research Center for Cardiology, Moscow, Russia
| |
Collapse
|
13
|
Yang X, Feng L, Zhang Y, Shi Y, Liang S, Zhao T, Sun B, Duan J, Sun Z. Integrative analysis of methylome and transcriptome variation of identified cardiac disease-specific genes in human cardiomyocytes after PM 2.5 exposure. CHEMOSPHERE 2018; 212:915-926. [PMID: 30286548 DOI: 10.1016/j.chemosphere.2018.09.010] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/20/2018] [Revised: 07/29/2018] [Accepted: 09/02/2018] [Indexed: 06/08/2023]
Abstract
PM2.5 exposure is strongly linked to cardiac disease. Subtle epigenetic or transcriptional alterations induced by PM2.5 might contribute to pathogenesis and disease susceptibility of cardiac disease. It is still a major challenge to identify biological targets in human genetics. Human cardiomyocytes AC16 was chosen as cell model. Epigenetic effect of PM2.5 in AC16 was analyzed using Illumina HumanMethylation 450 K BeadChip. Meanwhile the transcriptomic profiling was performed by Affymetrix® microarray. PM2.5 induced genome wide variation of DNA methylation pattern, including differentially methylated CpGs in promoter region. Then gene ontology analysis demonstrated differentially methylated genes were significantly clustered in pathways in regulation of apoptotic process, cell death and metabolic pathways, or associated with ion binding and shuttling. Correlation of the methylome and transcriptome revealed a clear bias toward transcriptional suppression by hypermethylation or activation by hypomethylation. Identified 386 genes which exhibited both differential methylation and expression were functionally associated with pathways including cardiovascular system development, regulation of blood vessel size, vasculature development, p53 pathway, AC-modulating/inhibiting GPCRs pathway and cellular response to metal ion/inorganic substance. Disease ontology demonstrated their prominent role in cardiac diseases and identified 14 cardiac-specific genes (ANK2, AQP1 et al.). PPI network analysis revealed 6 novel genes (POLR2I, LEP, BRIX1, ADCY6, INSL3, RARS). Those genes were then verified by qRT-PCR. Thus, in AC16, PM2.5 alters the methylome and transcriptome of genes might be relevant for PM2.5-/heart-associated diseases. Result gives additional insight in PM2.5 relative cardiac diseases/associated genes and the potential mechanisms that contribute to PM2.5 related cardiac disease.
Collapse
Affiliation(s)
- Xiaozhe Yang
- Department of Toxicology and Sanitary Chemistry, School of Public Health, Capital Medical University, Beijing 100069, PR China; Beijing Key Laboratory of Environmental Toxicology, Capital Medical University, Beijing 100069, PR China
| | - Lin Feng
- Department of Toxicology and Sanitary Chemistry, School of Public Health, Capital Medical University, Beijing 100069, PR China; Beijing Key Laboratory of Environmental Toxicology, Capital Medical University, Beijing 100069, PR China
| | - Yannan Zhang
- Department of Toxicology and Sanitary Chemistry, School of Public Health, Capital Medical University, Beijing 100069, PR China; Beijing Key Laboratory of Environmental Toxicology, Capital Medical University, Beijing 100069, PR China
| | - Yanfeng Shi
- Department of Toxicology and Sanitary Chemistry, School of Public Health, Capital Medical University, Beijing 100069, PR China; Beijing Key Laboratory of Environmental Toxicology, Capital Medical University, Beijing 100069, PR China
| | - Shuang Liang
- Department of Toxicology and Sanitary Chemistry, School of Public Health, Capital Medical University, Beijing 100069, PR China; Beijing Key Laboratory of Environmental Toxicology, Capital Medical University, Beijing 100069, PR China
| | - Tong Zhao
- Department of Toxicology and Sanitary Chemistry, School of Public Health, Capital Medical University, Beijing 100069, PR China; Beijing Key Laboratory of Environmental Toxicology, Capital Medical University, Beijing 100069, PR China
| | - Baiyang Sun
- Department of Toxicology and Sanitary Chemistry, School of Public Health, Capital Medical University, Beijing 100069, PR China; Beijing Key Laboratory of Environmental Toxicology, Capital Medical University, Beijing 100069, PR China
| | - Junchao Duan
- Department of Toxicology and Sanitary Chemistry, School of Public Health, Capital Medical University, Beijing 100069, PR China; Beijing Key Laboratory of Environmental Toxicology, Capital Medical University, Beijing 100069, PR China.
| | - Zhiwei Sun
- Department of Toxicology and Sanitary Chemistry, School of Public Health, Capital Medical University, Beijing 100069, PR China; Beijing Key Laboratory of Environmental Toxicology, Capital Medical University, Beijing 100069, PR China.
| |
Collapse
|
14
|
Doty RL. Olfaction: Smell of Change in the Air. CEREBRUM : THE DANA FORUM ON BRAIN SCIENCE 2017; 2017:cer-10-17. [PMID: 30210659 PMCID: PMC6132042] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/02/2022]
Abstract
Every whiff you take not only brings a cloud of chemicals swirling up your nose, but matters to your experience of taste as well as smell. Scientists studying smell have not only provided compelling evidence that it's more sophisticated than previously thought, but believe that the sense of smell impacts our mood and behavior and has the potential to detect and treat some neurological disorders. Compared to other senses, smell has long been underappreciated, writes our author, but that is now beginning to change.
Collapse
|
15
|
Jovancevic N, Dendorfer A, Matzkies M, Kovarova M, Heckmann JC, Osterloh M, Boehm M, Weber L, Nguemo F, Semmler J, Hescheler J, Milting H, Schleicher E, Gelis L, Hatt H. Medium-chain fatty acids modulate myocardial function via a cardiac odorant receptor. Basic Res Cardiol 2017; 112:13. [PMID: 28116519 PMCID: PMC5258789 DOI: 10.1007/s00395-017-0600-y] [Citation(s) in RCA: 80] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/29/2016] [Accepted: 01/03/2017] [Indexed: 12/17/2022]
Abstract
Several studies have demonstrated the expression of odorant receptors (OR) in various human tissues and their involvement in different physiological and pathophysiological processes. However, the functional role of ORs in the human heart is still unclear. Here, we firstly report the functional characterization of an OR in the human heart. Initial next-generation sequencing analysis revealed the OR expression pattern in the adult and fetal human heart and identified the fatty acid-sensing OR51E1 as the most highly expressed OR in both cardiac development stages. An extensive characterization of the OR51E1 ligand profile by luciferase reporter gene activation assay identified 2-ethylhexanoic acid as a receptor antagonist and various structurally related fatty acids as novel OR51E1 ligands, some of which were detected at receptor-activating concentrations in plasma and epicardial adipose tissue. Functional investigation of the endogenous receptor was carried out by Ca2+ imaging of human stem cell-derived cardiomyocytes. Application of OR51E1 ligands induced negative chronotropic effects that depended on activation of the OR. OR51E1 activation also provoked a negative inotropic action in cardiac trabeculae and slice preparations of human explanted ventricles. These findings indicate that OR51E1 may play a role as metabolic regulator of cardiac function.
Collapse
Affiliation(s)
- Nikolina Jovancevic
- Department of Cell Physiology, Ruhr-University Bochum, 44801, Bochum, Germany.
| | - A Dendorfer
- Walter Brendel Centre of Experimental Medicine, Ludwig-Maximilians-University, 80336, Munich, Germany.,DZHK (German Center for Cardiovascular Research), Partner Site Munich Heart Alliance, Munich, Germany
| | - M Matzkies
- Institute for Neurophysiology, University of Cologne, 50931, Cologne, Germany
| | - M Kovarova
- Division of Pathobiochemistry and Clinical Chemistry, University of Tuebingen, 72076, Tuebingen, Germany
| | - J C Heckmann
- Department of Cell Physiology, Ruhr-University Bochum, 44801, Bochum, Germany
| | - M Osterloh
- Department of Cell Physiology, Ruhr-University Bochum, 44801, Bochum, Germany
| | - M Boehm
- Department of Cell Physiology, Ruhr-University Bochum, 44801, Bochum, Germany
| | - L Weber
- Department of Cell Physiology, Ruhr-University Bochum, 44801, Bochum, Germany
| | - F Nguemo
- Institute for Neurophysiology, University of Cologne, 50931, Cologne, Germany
| | - J Semmler
- Institute for Neurophysiology, University of Cologne, 50931, Cologne, Germany
| | - J Hescheler
- Institute for Neurophysiology, University of Cologne, 50931, Cologne, Germany
| | - H Milting
- Erich and Hanna Klessmann Institute, Clinic for Thoracic and Cardiovascular Surgery, Heart and Diabetes Center NRW, Ruhr-University Bochum, 32545, Bad Oeynhausen, Germany
| | - E Schleicher
- Division of Pathobiochemistry and Clinical Chemistry, University of Tuebingen, 72076, Tuebingen, Germany
| | - L Gelis
- Department of Cell Physiology, Ruhr-University Bochum, 44801, Bochum, Germany
| | - H Hatt
- Department of Cell Physiology, Ruhr-University Bochum, 44801, Bochum, Germany
| |
Collapse
|