1
|
Yasumatsu K, Nagai Y, Ueshima F. [Causes of taste hyposensitivity in daily life and health risks: including the taste of fatty acids]. Nihon Yakurigaku Zasshi 2025; 160:73-78. [PMID: 40024708 DOI: 10.1254/fpj.24092] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/04/2025]
Abstract
The sensory system detects the internal and external environment of the body and the stimulus trigger feedback loops toward the set point to maintain homeostasis, but if taste sensitivity has changed, we may consume more nutrients or loss of appetite. These can lead metabolic syndrome or malnutrition, which can lead to frailty. In this review, we examined which of the five basic tastes (sweet, umami, bitter, sour, and salty) is affected by aging. Next, we summarize the effects of oral bacteria and tongue coating on taste, which can cause problems such as bad breath and aspiration pneumonia. Even healthy people can change their taste sensitivity and pose health risks if they continue to eat certain taste substances on a daily basis. Furthermore, we summarize research from the discovery of the taste of fatty acids to the present, and discuss how the involvement of taste in food intake regulation contributes to homeostasis through a literature survey. Recently, a gut-brain circuit for fat preference has been identified. In the intestine, fatty acids are sensed by the same receptors as those in the taste buds of the tongue, and nutritional information is sent to the brain via the vagus nerve. It is very interesting that nerves that convey fatty acid-specific information have been discovered. In this way, taste system of the tongue and nutrition-sensing in the digestive tract are very similar, so we think it will be very meaningful to progress research by referring to each other.
Collapse
|
2
|
Peng YC, Wu J, He X, Dai J, Xia L, Valenzuela-Leon P, Tumas KC, Singh BK, Xu F, Ganesan S, Munir S, Calvo E, Huang R, Liu C, Long CA, Su XZ. NAD activates olfactory receptor 1386 to regulate type I interferon responses in Plasmodium yoelii YM infection. Proc Natl Acad Sci U S A 2024; 121:e2403796121. [PMID: 38809710 PMCID: PMC11161801 DOI: 10.1073/pnas.2403796121] [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: 02/27/2024] [Accepted: 04/26/2024] [Indexed: 05/31/2024] Open
Abstract
Olfactory receptors (Olfr) are G protein-coupled receptors that are normally expressed on olfactory sensory neurons to detect volatile chemicals or odorants. Interestingly, many Olfrs are also expressed in diverse tissues and function in cell-cell recognition, migration, and proliferation as well as immune responses and disease processes. Here, we showed that many Olfr genes were expressed in the mouse spleen, linked to Plasmodium yoelii genetic loci significantly, and/or had genome-wide patterns of LOD scores (GPLSs) similar to those of host Toll-like receptor genes. Expression of specific Olfr genes such as Olfr1386 in HEK293T cells significantly increased luciferase signals driven by IFN-β and NF-κB promoters, with elevated levels of phosphorylated TBK1, IRF3, P38, and JNK. Mice without Olfr1386 were generated using the CRISPR/Cas9 method, and the Olfr1386-/- mice showed significantly lower IFN-α/β levels and longer survival than wild-type (WT) littermates after infection with P. yoelii YM parasites. Inhibition of G protein signaling and P38 activity could affect cyclic AMP-responsive element promoter-driven luciferase signals and IFN-β mRNA levels in HEK293T cells expressing the Olfr1386 gene, respectively. Screening of malaria parasite metabolites identified nicotinamide adenine dinucleotide (NAD) as a potential ligand for Olfr1386, and NAD could stimulate IFN-β responses and phosphorylation of TBK1 and STAT1/2 in RAW264.7 cells. Additionally, parasite RNA (pRNA) could significantly increase Olfr1386 mRNA levels. This study links multiple Olfrs to host immune response pathways, identifies a candidate ligand for Olfr1386, and demonstrates the important roles of Olfr1386 in regulating type I interferon (IFN-I) responses during malaria parasite infections.
Collapse
Affiliation(s)
- Yu-chih Peng
- Laboratory of Malaria and Vector Research, National Institute of Allergy and Infectious Disease, NIH, Rockville, MD20852
| | - Jian Wu
- Laboratory of Malaria and Vector Research, National Institute of Allergy and Infectious Disease, NIH, Rockville, MD20852
| | - Xiao He
- Laboratory of Malaria and Vector Research, National Institute of Allergy and Infectious Disease, NIH, Rockville, MD20852
| | - Jin Dai
- RNA Viruses Section, Laboratory of Infectious Diseases, National Institute of Allergy and Infectious Diseases, NIH, Bethesda, MD20892
| | - Lu Xia
- Hunan Key Laboratory of Medical Genetics, Key Lab of Rare Pediatric Disease of Ministry of Education, School of Life Sciences, Central South University, Changsha, Hunan410083, People’s Republic of China
| | - Paola Valenzuela-Leon
- Laboratory of Malaria and Vector Research, National Institute of Allergy and Infectious Disease, NIH, Rockville, MD20852
| | - Keyla C. Tumas
- Laboratory of Malaria and Vector Research, National Institute of Allergy and Infectious Disease, NIH, Rockville, MD20852
| | - Brajesh K. Singh
- Laboratory of Malaria and Vector Research, National Institute of Allergy and Infectious Disease, NIH, Rockville, MD20852
| | - Fangzheng Xu
- Laboratory of Malaria and Vector Research, National Institute of Allergy and Infectious Disease, NIH, Rockville, MD20852
| | - Sundar Ganesan
- Biological Imaging Section, National Institute of Allergy and Infectious Diseases, NIH, Bethesda, MD20892
| | - Shirin Munir
- RNA Viruses Section, Laboratory of Infectious Diseases, National Institute of Allergy and Infectious Diseases, NIH, Bethesda, MD20892
| | - Eric Calvo
- Laboratory of Malaria and Vector Research, National Institute of Allergy and Infectious Disease, NIH, Rockville, MD20852
| | - Ruili Huang
- National Center for Advancing Translational Sciences, NIH, Bethesda, MD20892
| | - Chengyu Liu
- Transgenic Core Facility, National Heart, Lung, and Blood Institute, NIH, Bethesda, MD20892
| | - Carole A. Long
- Laboratory of Malaria and Vector Research, National Institute of Allergy and Infectious Disease, NIH, Rockville, MD20852
| | - Xin-zhuan Su
- Laboratory of Malaria and Vector Research, National Institute of Allergy and Infectious Disease, NIH, Rockville, MD20852
| |
Collapse
|
3
|
Posta E, Fekete I, Gyarmati E, Stündl L, Zold E, Barta Z. The Effects of Artificial Sweeteners on Intestinal Nutrient-Sensing Receptors: Dr. Jekyll or Mr. Hyde? Life (Basel) 2023; 14:10. [PMID: 38276259 PMCID: PMC10817473 DOI: 10.3390/life14010010] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2023] [Revised: 12/14/2023] [Accepted: 12/15/2023] [Indexed: 01/27/2024] Open
Abstract
The consumption of artificial and low-calorie sweeteners (ASs, LCSs) is an important component of the Western diet. ASs play a role in the pathogenesis of metabolic syndrome, dysbiosis, inflammatory bowel diseases (IBDs), and various inflammatory conditions. Intestinal nutrient-sensing receptors act as a crosstalk between dietary components, the gut microbiota, and the regulation of immune, endocrinological, and neurological responses. This narrative review aimed to summarize the possible effects of ASs and LCSs on intestinal nutrient-sensing receptors and their related functions. Based on the findings of various studies, long-term AS consumption has effects on the gut microbiota and intestinal nutrient-sensing receptors in modulating incretin hormones, antimicrobial peptides, and cytokine secretion. These effects contribute to the regulation of glucose metabolism, ion transport, gut permeability, and inflammation and modulate the gut-brain, and gut-kidney axes. Based on the conflicting findings of several in vitro, in vivo, and randomized and controlled studies, artificial sweeteners may have a role in the pathogenesis of IBDs, functional bowel diseases, metabolic syndrome, and cancers via the modulation of nutrient-sensing receptors. Further studies are needed to explore the exact mechanisms underlying their effects to decide the risk/benefit ratio of sugar intake reduction via AS and LCS consumption.
Collapse
Affiliation(s)
- Edit Posta
- GI Unit, Department of Infectology, Faculty of Medicine, University of Debrecen, Bartok Bela Street 2-26, 4031 Debrecen, Hungary; (E.G.); (Z.B.)
| | - Istvan Fekete
- Institute of Food Technology, Faculty of Agricultural and Food Sciences and Environmental Management, University of Debrecen, Böszörményi út 138, 4032 Debrecen, Hungary; (I.F.); (L.S.)
| | - Eva Gyarmati
- GI Unit, Department of Infectology, Faculty of Medicine, University of Debrecen, Bartok Bela Street 2-26, 4031 Debrecen, Hungary; (E.G.); (Z.B.)
- Doctoral School of Clinical Immunology and Allergology, Faculty of Medicine, University of Debrecen, Nagyerdei Blvd. 98, 4032 Debrecen, Hungary
| | - László Stündl
- Institute of Food Technology, Faculty of Agricultural and Food Sciences and Environmental Management, University of Debrecen, Böszörményi út 138, 4032 Debrecen, Hungary; (I.F.); (L.S.)
| | - Eva Zold
- Department of Clinical Immunology, Institute of Internal Medicine, Faculty of Medicine, University of Debrecen, Móricz Zsigmond Str. 22, 4032 Debrecen, Hungary;
| | - Zsolt Barta
- GI Unit, Department of Infectology, Faculty of Medicine, University of Debrecen, Bartok Bela Street 2-26, 4031 Debrecen, Hungary; (E.G.); (Z.B.)
| |
Collapse
|
4
|
Kouakou YI, Lee RJ. Interkingdom Detection of Bacterial Quorum-Sensing Molecules by Mammalian Taste Receptors. Microorganisms 2023; 11:1295. [PMID: 37317269 PMCID: PMC10221136 DOI: 10.3390/microorganisms11051295] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2023] [Revised: 05/11/2023] [Accepted: 05/13/2023] [Indexed: 06/16/2023] Open
Abstract
Bitter and sweet taste G protein-coupled receptors (known as T2Rs and T1Rs, respectively) were originally identified in type II taste cells on the tongue, where they signal perception of bitter and sweet tastes, respectively. Over the past ~15 years, taste receptors have been identified in cells all over the body, demonstrating a more general chemosensory role beyond taste. Bitter and sweet taste receptors regulate gut epithelial function, pancreatic β cell secretion, thyroid hormone secretion, adipocyte function, and many other processes. Emerging data from a variety of tissues suggest that taste receptors are also used by mammalian cells to "eavesdrop" on bacterial communications. These receptors are activated by several quorum-sensing molecules, including acyl-homoserine lactones and quinolones from Gram-negative bacteria such as Pseudomonas aeruginosa, competence stimulating peptides from Streptococcus mutans, and D-amino acids from Staphylococcus aureus. Taste receptors are an arm of immune surveillance similar to Toll-like receptors and other pattern recognition receptors. Because they are activated by quorum-sensing molecules, taste receptors report information about microbial population density based on the chemical composition of the extracellular environment. This review summarizes current knowledge of bacterial activation of taste receptors and identifies important questions remaining in this field.
Collapse
Affiliation(s)
- Yobouet Ines Kouakou
- Department of Otorhinolaryngology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA;
| | - Robert J. Lee
- Department of Otorhinolaryngology and Physiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| |
Collapse
|
5
|
Anti-Citrullinated Peptide Antibodies Control Oral Porphyromonas and Aggregatibacter species in Patients with Rheumatoid Arthritis. Int J Mol Sci 2022; 23:ijms232012599. [PMID: 36293451 PMCID: PMC9604485 DOI: 10.3390/ijms232012599] [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: 09/27/2022] [Revised: 10/14/2022] [Accepted: 10/18/2022] [Indexed: 11/29/2022] Open
Abstract
Oral microbiome changes take place at the initiation of rheumatoid arthritis (RA); however, questions remain regarding the oral microbiome at pre-RA stages in individuals with clinically suspect arthralgia (CSA). Two cross-sectional cohorts were selected including 84 Tatarstan women (15 early-RA as compared to individuals with CSA ranging from CSA = 0 [n = 22], CSA = 1 [n = 19], CSA = 2 [n = 11], and CSA ≥ 3 [n = 17]) and 42 women with established RA (median: 5 years from diagnosis [IQ: 2–11]). Amplicon sequence variants (ASVs) obtained from oral samples (16S rRNA) were analyzed for alpha and beta diversity along with the abundance at the genus level. A decrease in oral Porphyromonas sp. is observed in ACPA-positive individuals, and this predominates in early-RA patients as compared to non-RA individuals irrespective of their CSA score. In the RA-established cohort, Porphyromonas sp. and Aggregatibacter sp. reductions were associated with elevated ACPA levels. In contrast, no associations were reported when considering individual, genetic and clinical RA-associated factors. Oral microbiome changes related to the genera implicated in post-translational citrullination (Porphyromonas sp. and Aggregatibacter sp.) characterized RA patients with elevated ACPA levels, which supports that the role of ACPA in controlling the oral microbiome needs further evaluation.
Collapse
|
6
|
Abstract
Taste receptors are receptor proteins that detect ligands belonging to the 5 taste modalities: sweet, bitter, sour, salty, and umami. Taste receptors are not restricted to taste cells in taste buds; rather, they are distributed throughout the entire body. For example, solitary chemosensory cells (SCCs) and tuft cells express taste signal proteins and are present in several mucosae. In the airways, SCCs sense bacteria, allergens, viruses, and noxious stimuli and drive evasive behavior, neuroinflammation, and antibacterial responses. In the gut, tuft cells detect helminth infection and bacterial dysbiosis and initiate type II immune responses characterized by tissue remodeling. In the gingiva, SCCs detect oral pathogenic bacteria, evoke innate immune responses and release antimicrobial compounds in the epithelium, and regulate the microbiome composition. This review summarizes the most recent research on extragustatory taste receptors and their function in antibacterial defense. We also discuss how these findings have provided insights into the development of potential therapeutic strategies for mucosal bacterial infection and dental diseases.
Collapse
Affiliation(s)
- R. Xi
- Department of Cariology and Endodontics, Sichuan University, West China Hospital of Stomatology, Chengdu, China
- Department of Basic and Translational Sciences, School of Dental Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - X. Zheng
- Department of Cariology and Endodontics, Sichuan University, West China Hospital of Stomatology, Chengdu, China
| | - M. Tizzano
- Department of Basic and Translational Sciences, School of Dental Medicine, University of Pennsylvania, Philadelphia, PA, USA
| |
Collapse
|
7
|
Gao L, Cheng Z, Zhu F, Bi C, Shi Q, Chen X. The Oral Microbiome and Its Role in Systemic Autoimmune Diseases: A Systematic Review of Big Data Analysis. Front Big Data 2022; 5:927520. [PMID: 35844967 PMCID: PMC9277227 DOI: 10.3389/fdata.2022.927520] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2022] [Accepted: 05/23/2022] [Indexed: 12/30/2022] Open
Abstract
Introduction Despite decades of research, systemic autoimmune diseases (SADs) continue to be a major global health concern and the etiology of these diseases is still not clear. To date, with the development of high-throughput techniques, increasing evidence indicated a key role of oral microbiome in the pathogenesis of SADs, and the alterations of oral microbiome may contribute to the disease emergence or evolution. This review is to present the latest knowledge on the relationship between the oral microbiome and SADs, focusing on the multiomics data generated from a large set of samples. Methodology By searching the PubMed and Embase databases, studies that investigated the oral microbiome of SADs, including systemic lupus erythematosus (SLE), rheumatoid arthritis (RA), and Sjögren's syndrome (SS), were systematically reviewed according to the PRISMA guidelines. Results One thousand and thirty-eight studies were found, and 25 studies were included: three referred to SLE, 12 referred to RA, nine referred to SS, and one to both SLE and SS. The 16S rRNA sequencing was the most frequent technique used. HOMD was the most common database aligned to and QIIME was the most popular pipeline for downstream analysis. Alterations in bacterial composition and population have been found in the oral samples of patients with SAD compared with the healthy controls. Results regarding candidate pathogens were not always in accordance, but Selenomonas and Veillonella were found significantly increased in three SADs, and Streptococcus was significantly decreased in the SADs compared with controls. Conclusion A large amount of sequencing data was collected from patients with SAD and controls in this systematic review. Oral microbial dysbiosis had been identified in these SADs, although the dysbiosis features were different among studies. There was a lack of standardized study methodology for each study from the inclusion criteria, sample type, sequencing platform, and referred database to downstream analysis pipeline and cutoff. Besides the genomics, transcriptomics, proteomics, and metabolomics technology should be used to investigate the oral microbiome of patients with SADs and also the at-risk individuals of disease development, which may provide us with a better understanding of the etiology of SADs and promote the development of the novel therapies.
Collapse
Affiliation(s)
- Lu Gao
- Stomatology Hospital, School of Stomatology, Zhejiang University School of Medicine, Hangzhou, China
- Zhejiang Provincial Clinical Research Center for Oral Diseases, Hangzhou, China
- Key Laboratory of Oral Biomedical Research of Zhejiang Province, Cancer Center of Zhejiang University, Hangzhou, China
| | - Zijian Cheng
- Stomatology Hospital, School of Stomatology, Zhejiang University School of Medicine, Hangzhou, China
- Zhejiang Provincial Clinical Research Center for Oral Diseases, Hangzhou, China
- Key Laboratory of Oral Biomedical Research of Zhejiang Province, Cancer Center of Zhejiang University, Hangzhou, China
| | - Fudong Zhu
- Stomatology Hospital, School of Stomatology, Zhejiang University School of Medicine, Hangzhou, China
- Zhejiang Provincial Clinical Research Center for Oral Diseases, Hangzhou, China
- Key Laboratory of Oral Biomedical Research of Zhejiang Province, Cancer Center of Zhejiang University, Hangzhou, China
| | - Chunsheng Bi
- Stomatology Hospital, School of Stomatology, Zhejiang University School of Medicine, Hangzhou, China
- Zhejiang Provincial Clinical Research Center for Oral Diseases, Hangzhou, China
- Key Laboratory of Oral Biomedical Research of Zhejiang Province, Cancer Center of Zhejiang University, Hangzhou, China
| | - Qiongling Shi
- Stomatology Hospital, School of Stomatology, Zhejiang University School of Medicine, Hangzhou, China
- Zhejiang Provincial Clinical Research Center for Oral Diseases, Hangzhou, China
- Key Laboratory of Oral Biomedical Research of Zhejiang Province, Cancer Center of Zhejiang University, Hangzhou, China
| | - Xiaoyan Chen
- Stomatology Hospital, School of Stomatology, Zhejiang University School of Medicine, Hangzhou, China
- Zhejiang Provincial Clinical Research Center for Oral Diseases, Hangzhou, China
- Key Laboratory of Oral Biomedical Research of Zhejiang Province, Cancer Center of Zhejiang University, Hangzhou, China
- *Correspondence: Xiaoyan Chen
| |
Collapse
|