1
|
Gomes AS, Gélébart V, Félix RC, Cardoso JCR, Zimmermann F, Lai F, Power DM, Rønnestad I. Activation profile of the Atlantic salmon (Salmo salar) calcium-sensing receptor (Casr) by selected L-amino acids. Sci Rep 2025; 15:13236. [PMID: 40247003 PMCID: PMC12006508 DOI: 10.1038/s41598-025-97483-5] [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: 05/10/2024] [Accepted: 04/04/2025] [Indexed: 04/19/2025] Open
Abstract
In mammals, the calcium-sensing receptor (CaSR) is involved in nutrient sensing and modulated by several amino acids. In teleosts, sequence homologues of the mammalian CaSR have been described but their function in sensing amino acids remains elusive, including in Atlantic salmon (Salmo salar), an important aquaculture species. This study investigated the activation of Atlantic salmon Casr (asCasr)-mediated signaling pathways-Gq, Gi, and ERK1/2-by six selected L-amino acids (histidine, tryptophan, phenylalanine, isoleucine, leucine and valine) and by Ca2+. Using a Flp-In-HEK293 cell line stably expressing asCasr, we confirmed activation of all three pathways. L-histidine, L-phenylalanine, and L-tryptophan triggered Gi signaling independent of Ca²⁺. Notably, no Ca²⁺ concentrations induced Gi activation, but IP1 production increased in a concentration-dependent manner. L-histidine was the only amino acid to activate the Gq pathway without Ca²⁺, and this response was amplified by the presence of Ca²⁺. In the presence of 2.5 mM Ca²⁺, L-phenylalanine and L-tryptophan also activated Gq signaling in a concentration-dependent manner. Additionally, in the presence of 10 mM Ca²⁺, L-histidine, L-phenylalanine, and L-tryptophan triggered ERK phosphorylation. These findings establish asCasr as a functional homologue of mammalian CaSR, activated in a concentration-dependent manner by L-amino acids with an aromatic ring.
Collapse
Affiliation(s)
- Ana S Gomes
- Department of Biological Sciences, University of Bergen, Bergen, Norway.
- Institute of Marine Research, Tromsø, Norway.
| | - Virginie Gélébart
- Department of Biological Sciences, University of Bergen, Bergen, Norway
| | - Rute C Félix
- Centre of Marine Sciences (CCMAR/CIMAR), University of Algarve, Faro, Portugal
| | - João C R Cardoso
- Centre of Marine Sciences (CCMAR/CIMAR), University of Algarve, Faro, Portugal
| | | | - Floriana Lai
- Department of Biological Sciences, University of Bergen, Bergen, Norway
| | - Deborah M Power
- Centre of Marine Sciences (CCMAR/CIMAR), University of Algarve, Faro, Portugal
- International Research Center for Marine Biosciences, Ministry of Science and Technology and National Demonstration Center for Experimental Fisheries Science Education, Shanghai Ocean University, Shanghai, China
| | - Ivar Rønnestad
- Department of Biological Sciences, University of Bergen, Bergen, Norway.
| |
Collapse
|
2
|
Feng T, Ma C, Chen S, Zhuang H, Song S, Sun M, Yao L, Wang H, Liu Q, Yu C. Exploring novel Kokumi peptides in Agaricus bisporus: selection, identification, and tasting mechanism investigation through sensory evaluation and computer simulation analysis. Food Funct 2024; 15:2879-2894. [PMID: 38318946 DOI: 10.1039/d3fo05406c] [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: 02/07/2024]
Abstract
Agaricus bisporus contains amino acids associated with thickness and full-mouthfeel, making it a potential candidate for salt substitutes and flavor enhancers in various food applications. Kokumi peptides were isolated from the enzymatic digest of Agaricus bisporus using ultrafiltration nanofiltration, gel chromatographic separation, and RP-HPLC, coupled with sensory evaluation. Subsequently, the peptides, EWVPVTK and EYPPLGR, were selected for solid-phase synthesis based on molecular docking. Sensory analysis, including thresholds, time intensity, and dose-configuration relationships, indicated that EWVPVTK and EYPPLGR exhibited odor thresholds of 0.6021 mmol L-1 and 2.332 mmol L-1 in an aqueous solution. Molecular docking scores correlated with low sensory thresholds, signifying strong taste sensitivities. EWVPVTK, in particular, demonstrated a higher sense of richness at lower concentrations compared to EYPPLGR. Molecular docking and dynamics simulations elucidated that the interactions between Kokumi peptides and the CaSR receptor primarily involved hydrogen bonding, electrostatic interactions, and hydrophobic interactions. Both EWVPVTK and EYPPLGR exhibited stable binding to the CaSR receptor. Active binding sites were identified, with EWVPVTK interacting at Arg 66, Asp 216, Gln 245, and Asn 102, while EYPPLGR engaged with Ser 272, Gln 193, Glu 297, Ala-298, Tyr-2, and Agr-66 in hydrophilic interactions through hydrogen bonds. Notably, these two Kokumi peptides were found to be enriched in umami and sweet amino acids, underscoring their pivotal role in umami perception. This study not only identifies novel Kokumi peptides from Agaricus bisporus but also contributes theoretical foundations and insights for future studies in the realm of Kokumi peptides.
Collapse
Affiliation(s)
- Tao Feng
- School of Perfume and Aroma Technology, Shanghai Institute of Technology, 100 Haiquan Road, Shanghai 201418, People's Republic of China.
| | - Chenwei Ma
- School of Perfume and Aroma Technology, Shanghai Institute of Technology, 100 Haiquan Road, Shanghai 201418, People's Republic of China.
| | - Sha Chen
- College of Life Science and Technology, Xinjiang University, 666 Shengli Road, Xinjiang Urumqi 830000, People's Republic of China.
| | - Haining Zhuang
- School of Food and Tourism, Shanghai Urban Construction Vocational College, 2080 Nanting Road, Shanghai, 201415, People's Republic of China.
| | - Shiqing Song
- School of Perfume and Aroma Technology, Shanghai Institute of Technology, 100 Haiquan Road, Shanghai 201418, People's Republic of China.
| | - Min Sun
- School of Perfume and Aroma Technology, Shanghai Institute of Technology, 100 Haiquan Road, Shanghai 201418, People's Republic of China.
| | - Lingyun Yao
- School of Perfume and Aroma Technology, Shanghai Institute of Technology, 100 Haiquan Road, Shanghai 201418, People's Republic of China.
| | - Huatian Wang
- School of Perfume and Aroma Technology, Shanghai Institute of Technology, 100 Haiquan Road, Shanghai 201418, People's Republic of China.
| | - Qian Liu
- School of Perfume and Aroma Technology, Shanghai Institute of Technology, 100 Haiquan Road, Shanghai 201418, People's Republic of China.
| | - Chuang Yu
- School of Perfume and Aroma Technology, Shanghai Institute of Technology, 100 Haiquan Road, Shanghai 201418, People's Republic of China.
| |
Collapse
|
3
|
Zhan X, Wang D, Wang H, Chen H, Wu X, Li T, Qi J, Chen T, Wu D, Gao Y. Revitalizing Skin Repair: Unveiling the Healing Power of Livisin, a Natural Peptide Calcium Mimetic. Toxins (Basel) 2023; 16:21. [PMID: 38251238 PMCID: PMC10819626 DOI: 10.3390/toxins16010021] [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: 10/31/2023] [Revised: 11/30/2023] [Accepted: 12/25/2023] [Indexed: 01/23/2024] Open
Abstract
When the skin is damaged, accelerating the repair of skin trauma and promoting the recovery of tissue function are crucial considerations in clinical treatment. Previously, we isolated and identified an active peptide (livisin) from the skin secretion of the frog Odorrana livida. Livisin exhibited strong protease inhibitory activity, water solubility, and stability, yet its wound-healing properties have not yet been studied. In this study, we assessed the impact of livisin on wound healing and investigated the underlying mechanism contributing to its effect. Our findings revealed livisin effectively stimulated the migration of keratinocytes, with the underlying mechanisms involved the activation of CaSR as a peptide calcium mimetic. This activation resulted in the stimulation of the CaSR/E-cadherin/EGFR/ERK signaling pathways. Moreover, the therapeutic effects of livisin were partially reduced by blocking the CaSR/E-cadherin/EGFR/ERK signaling pathway. The interaction between livisin and CaSR was further investigated by molecular docking. Additionally, studies using a mouse full-thickness wound model demonstrated livisin could accelerate skin wound healing by promoting re-epithelialization and collagen deposition. In conclusion, our study provides experimental evidence supporting the use of livisin in skin wound healing, highlighting its potential as an effective therapeutic option.
Collapse
Affiliation(s)
- Xuehui Zhan
- Zhejiang Provincial Key Laboratory for Water Environment and Marine, Biological Resources Protection, College of Life and Environmental Science, Wenzhou University, Wenzhou 325035, China; (X.Z.); (D.W.); (H.W.); (T.L.); (J.Q.)
- School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou 325015, China; (H.C.); (X.W.)
| | - Danni Wang
- Zhejiang Provincial Key Laboratory for Water Environment and Marine, Biological Resources Protection, College of Life and Environmental Science, Wenzhou University, Wenzhou 325035, China; (X.Z.); (D.W.); (H.W.); (T.L.); (J.Q.)
| | - Hanfei Wang
- Zhejiang Provincial Key Laboratory for Water Environment and Marine, Biological Resources Protection, College of Life and Environmental Science, Wenzhou University, Wenzhou 325035, China; (X.Z.); (D.W.); (H.W.); (T.L.); (J.Q.)
- College of Animal Science and Technology, Huazhong Agricultural University, Wuhan 430070, China
| | - Hui Chen
- School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou 325015, China; (H.C.); (X.W.)
| | - Xinyi Wu
- School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou 325015, China; (H.C.); (X.W.)
| | - Tao Li
- Zhejiang Provincial Key Laboratory for Water Environment and Marine, Biological Resources Protection, College of Life and Environmental Science, Wenzhou University, Wenzhou 325035, China; (X.Z.); (D.W.); (H.W.); (T.L.); (J.Q.)
| | - Junmei Qi
- Zhejiang Provincial Key Laboratory for Water Environment and Marine, Biological Resources Protection, College of Life and Environmental Science, Wenzhou University, Wenzhou 325035, China; (X.Z.); (D.W.); (H.W.); (T.L.); (J.Q.)
| | - Tianbao Chen
- Natural Drug Discovery Group, School of Pharmacy, Queen’s University Belfast, Belfast BT7 1NN, UK;
| | - Di Wu
- School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou 325015, China; (H.C.); (X.W.)
| | - Yitian Gao
- Zhejiang Provincial Key Laboratory for Water Environment and Marine, Biological Resources Protection, College of Life and Environmental Science, Wenzhou University, Wenzhou 325035, China; (X.Z.); (D.W.); (H.W.); (T.L.); (J.Q.)
| |
Collapse
|
4
|
Centeno PP, Binmahfouz LS, Alghamdi K, Ward DT. Inhibition of the calcium-sensing receptor by extracellular phosphate ions and by intracellular phosphorylation. Front Physiol 2023; 14:1154374. [PMID: 37064904 PMCID: PMC10102455 DOI: 10.3389/fphys.2023.1154374] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2023] [Accepted: 03/20/2023] [Indexed: 04/03/2023] Open
Abstract
As both a sensor of extracellular calcium (Ca2+o) concentration and a key controller of Ca2+o homeostasis, one of the most interesting properties of the calcium-sensing receptor (CaR) is its sensitivity to, and modulation by, ions and small ligands other than Ca2+. There is emerging evidence that extracellular phosphate can act as a partial, non-competitive CaR antagonist to modulate parathyroid hormone (PTH) secretion, thus permitting the CaR to integrate mineral homeostasis more broadly. Interestingly, phosphorylation of certain intracellular CaR residues can also inhibit CaR responsiveness. Thus, negatively charged phosphate can decrease CaR activity both extracellularly (via association with arginine) and intracellularly (via covalent phosphorylation).
Collapse
Affiliation(s)
- Patricia P. Centeno
- Faculty of Biology, Medicine and Health, The University of Manchester, Manchester, United Kingdom
| | - Lenah S. Binmahfouz
- Faculty of Biology, Medicine and Health, The University of Manchester, Manchester, United Kingdom
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, King Abdulaziz University, Jeddah, Saudi Arabia
| | - Khaleda Alghamdi
- Faculty of Biology, Medicine and Health, The University of Manchester, Manchester, United Kingdom
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, King Abdulaziz University, Jeddah, Saudi Arabia
| | - Donald T. Ward
- Faculty of Biology, Medicine and Health, The University of Manchester, Manchester, United Kingdom
- *Correspondence: Donald T. Ward,
| |
Collapse
|
5
|
Brennan SC, Mun HC, Delbridge L, Kuchel PW, Conigrave AD. Temperature sensing by the calcium-sensing receptor. Front Physiol 2023; 14:1117352. [PMID: 36818436 PMCID: PMC9931745 DOI: 10.3389/fphys.2023.1117352] [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: 12/06/2022] [Accepted: 01/16/2023] [Indexed: 02/05/2023] Open
Abstract
Whether GPCRs support the sensing of temperature as well as other chemical and physical modalities is not well understood. Introduction: Extracellular Ca2+ concentration (Ca2+ o) modulates core body temperature and the firing rates of temperature-sensitive CNS neurons, and hypocalcemia provokes childhood seizures. However, it is not known whether these phenomena are mediated by Ca2+ o-sensing GPCRs, including the calcium-sensing receptor (CaSR). In favor of the hypothesis, CaSRs are expressed in hypothalamic regions that support core temperature regulation, and autosomal dominant hypocalcemia, due to CaSR activating mutations, is associated with childhood seizures. Methods: Herein, we tested whether CaSR-dependent signaling is temperature sensitive using an established model system, CaSR-expressing HEK-293 cells. Results: We found that the frequency of Ca2+ o-induced Ca2+ i oscillations but not the integrated response was linearly dependent on temperature in a pathophysiologically relevant range. Chimeric receptor analysis showed that the receptor's C-terminus is required for temperature-dependent modulation and experiments with the PKC inhibitor GF109203X and CaSR mutants T888A and T888M, which eliminate a key phosphorylation site, demonstrated the importance of repetitive phosphorylation and dephosphorylation. Discussion and Conclusion: CaSRs mediate temperature-sensing and the mechanism, dependent upon repetitive phosphorylation and dephosphorylation, suggests that GPCRs more generally contribute to temperature-sensing.
Collapse
Affiliation(s)
- Sarah C. Brennan
- School of Life and Environmental Sciences, Faculty of Science, The University of Sydney, Sydney, NSW, Australia,Charles Perkins Centre, University of Sydney, Sydney, NSW, Australia
| | - Hee-chang Mun
- Charles Perkins Centre, University of Sydney, Sydney, NSW, Australia
| | - Leigh Delbridge
- Department of Surgery, Mater Hospital, North Sydney, NSW, Australia
| | - Philip W. Kuchel
- School of Life and Environmental Sciences, Faculty of Science, The University of Sydney, Sydney, NSW, Australia
| | - Arthur D. Conigrave
- School of Life and Environmental Sciences, Faculty of Science, The University of Sydney, Sydney, NSW, Australia,Charles Perkins Centre, University of Sydney, Sydney, NSW, Australia,*Correspondence: Arthur D. Conigrave,
| |
Collapse
|
6
|
Gong Y, Yang B, Zhang D, Zhang Y, Tang Z, Yang L, Coate KC, Yin L, Covington BA, Patel RS, Siv WA, Sellick K, Shou M, Chang W, Danielle Dean E, Powers AC, Chen W. Hyperaminoacidemia induces pancreatic α cell proliferation via synergism between the mTORC1 and CaSR-Gq signaling pathways. Nat Commun 2023; 14:235. [PMID: 36646689 PMCID: PMC9842633 DOI: 10.1038/s41467-022-35705-4] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2021] [Accepted: 12/20/2022] [Indexed: 01/18/2023] Open
Abstract
Glucagon has emerged as a key regulator of extracellular amino acid (AA) homeostasis. Insufficient glucagon signaling results in hyperaminoacidemia, which drives adaptive proliferation of glucagon-producing α cells. Aside from mammalian target of rapamycin complex 1 (mTORC1), the role of other AA sensors in α cell proliferation has not been described. Here, using both genders of mouse islets and glucagon receptor (gcgr)-deficient zebrafish (Danio rerio), we show α cell proliferation requires activation of the extracellular signal-regulated protein kinase (ERK1/2) by the AA-sensitive calcium sensing receptor (CaSR). Inactivation of CaSR dampened α cell proliferation, which was rescued by re-expression of CaSR or activation of Gq, but not Gi, signaling in α cells. CaSR was also unexpectedly necessary for mTORC1 activation in α cells. Furthermore, coactivation of Gq and mTORC1 induced α cell proliferation independent of hyperaminoacidemia. These results reveal another AA-sensitive mediator and identify pathways necessary and sufficient for hyperaminoacidemia-induced α cell proliferation.
Collapse
Affiliation(s)
- Yulong Gong
- Department of Molecular Physiology & Biophysics, Vanderbilt University, 2215 Garland Ave, Nashville, TN, 37232, USA
- State Key Laboratory of Freshwater Ecology and Biotechnology, Institute of Hydrobiology, Chinese Academy of Sciences, Wuhan, Hubei, 430072, China
| | - Bingyuan Yang
- Department of Molecular Physiology & Biophysics, Vanderbilt University, 2215 Garland Ave, Nashville, TN, 37232, USA
| | - Dingdong Zhang
- Department of Molecular Physiology & Biophysics, Vanderbilt University, 2215 Garland Ave, Nashville, TN, 37232, USA
- College of Animal Science and Technology, Nanjing Agricultural University, Nanjing, 210095, China
| | - Yue Zhang
- Department of Molecular Physiology & Biophysics, Vanderbilt University, 2215 Garland Ave, Nashville, TN, 37232, USA
| | - Zihan Tang
- Department of Molecular Physiology & Biophysics, Vanderbilt University, 2215 Garland Ave, Nashville, TN, 37232, USA
| | - Liu Yang
- Department of Molecular Physiology & Biophysics, Vanderbilt University, 2215 Garland Ave, Nashville, TN, 37232, USA
| | - Katie C Coate
- Division of Diabetes, Endocrinology and Metabolism, Department of Medicine, Vanderbilt University Medical Center, 2215 Garland Ave, Nashville, TN, 37232, USA
| | - Linlin Yin
- Department of Molecular Physiology & Biophysics, Vanderbilt University, 2215 Garland Ave, Nashville, TN, 37232, USA
| | - Brittney A Covington
- Department of Molecular Physiology & Biophysics, Vanderbilt University, 2215 Garland Ave, Nashville, TN, 37232, USA
| | - Ravi S Patel
- Department of Molecular Physiology & Biophysics, Vanderbilt University, 2215 Garland Ave, Nashville, TN, 37232, USA
| | - Walter A Siv
- Division of Diabetes, Endocrinology and Metabolism, Department of Medicine, Vanderbilt University Medical Center, 2215 Garland Ave, Nashville, TN, 37232, USA
| | - Katelyn Sellick
- Division of Diabetes, Endocrinology and Metabolism, Department of Medicine, Vanderbilt University Medical Center, 2215 Garland Ave, Nashville, TN, 37232, USA
| | - Matthew Shou
- Division of Diabetes, Endocrinology and Metabolism, Department of Medicine, Vanderbilt University Medical Center, 2215 Garland Ave, Nashville, TN, 37232, USA
| | - Wenhan Chang
- University of California San Francisco and San Francisco VA Medical Center, San Francisco, CA, 94158, USA
| | - E Danielle Dean
- Department of Molecular Physiology & Biophysics, Vanderbilt University, 2215 Garland Ave, Nashville, TN, 37232, USA
- Division of Diabetes, Endocrinology and Metabolism, Department of Medicine, Vanderbilt University Medical Center, 2215 Garland Ave, Nashville, TN, 37232, USA
| | - Alvin C Powers
- Department of Molecular Physiology & Biophysics, Vanderbilt University, 2215 Garland Ave, Nashville, TN, 37232, USA
- Division of Diabetes, Endocrinology and Metabolism, Department of Medicine, Vanderbilt University Medical Center, 2215 Garland Ave, Nashville, TN, 37232, USA
- VA Tennessee Valley Healthcare System, Nashville, TN, 37212, USA
| | - Wenbiao Chen
- Department of Molecular Physiology & Biophysics, Vanderbilt University, 2215 Garland Ave, Nashville, TN, 37232, USA.
| |
Collapse
|
7
|
Sato H, Murakami S, Horii Y, Nishimura G, Iwai R, Goto M, Takahashi N. Upacicalcet Is a Novel Secondary Hyperparathyroidism Drug that Targets the Amino Acid Binding Site of Calcium-Sensing Receptor . Mol Pharmacol 2022; 102:183-195. [PMID: 36122913 DOI: 10.1124/molpharm.122.000522] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2022] [Accepted: 07/14/2022] [Indexed: 02/14/2025] Open
Abstract
The human calcium-sensing receptor (CaSR) is a G protein-coupled receptor that maintains extracellular Ca2+ homeostasis by regulating the secretion of parathyroid hormone. Upacicalcet is a novel positive allosteric modulator of CaSR that is used for the treatment of secondary hyperparathyroidism. In the present study, to clarify the binding site of upacicalcet to CaSR, we conducted binding studies and agonistic activity studies in HEK-293T cells expressing human CaSR (intact and mutant) and an in silico docking-simulation analysis. As a result, upacicalcet competed with L-tryptophan and was thought to affect the amino acid binding site. In addition, the effects of substitutions at the amino acid binding site on the binding abilities to upacicalcet as well as the effects on receptor function as measured using inositol-1 monophosphate accumulation were examined. Upacicalcet interacted with several CaSR residues that constitute the amino acid binding site. Based on these results, we performed an in silico analysis and obtained a binding mode, consistent with the in vitro study results. Our study revealed that upacicalcet is a novel secondary hyperparathyroidism drug that targets the amino acid binding site of CaSR. Upacicalcet is expected to become a new treatment option for secondary hyperparathyroidism because the binding site differs from that of conventional drugs; consequently, it may be effective for patients who are not sensitive to conventional drugs, and it may have a superior safety profile. SIGNIFICANCE STATEMENT: Upacicalcet interacts with several residues that constitute the amino acid binding site of the calcium-sensing receptor (CaSR) and shows a potent positive allosteric activity. This mechanism differs from those of conventional drugs. Therefore, upacicalcet can be regarded as a novel secondary hyperparathyroidism drug that acts on the amino acid binding site of CaSR.
Collapse
Affiliation(s)
- Hirofumi Sato
- Pharmaceuticals Research Laboratories, Sanwa Kagaku Kenkyusho Co., Ltd., Mie, Japan
| | - Sei Murakami
- Pharmaceuticals Research Laboratories, Sanwa Kagaku Kenkyusho Co., Ltd., Mie, Japan
| | - Yusuke Horii
- Pharmaceuticals Research Laboratories, Sanwa Kagaku Kenkyusho Co., Ltd., Mie, Japan
| | - Go Nishimura
- Pharmaceuticals Research Laboratories, Sanwa Kagaku Kenkyusho Co., Ltd., Mie, Japan
| | - Ryosuke Iwai
- Pharmaceuticals Research Laboratories, Sanwa Kagaku Kenkyusho Co., Ltd., Mie, Japan
| | - Moritaka Goto
- Pharmaceuticals Research Laboratories, Sanwa Kagaku Kenkyusho Co., Ltd., Mie, Japan
| | - Naoki Takahashi
- Pharmaceuticals Research Laboratories, Sanwa Kagaku Kenkyusho Co., Ltd., Mie, Japan
| |
Collapse
|
8
|
Wang D, Ye J, Shi R, Zhao B, Liu Z, Lin W, Liu X. Dietary protein and amino acid restriction: Roles in metabolic health and aging-related diseases. Free Radic Biol Med 2022; 178:226-242. [PMID: 34890767 DOI: 10.1016/j.freeradbiomed.2021.12.009] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/30/2021] [Revised: 12/03/2021] [Accepted: 12/04/2021] [Indexed: 12/13/2022]
Abstract
The prevalence of obesity is a worldwide phenomenon in all age groups and is associated with aging-related diseases such as type 2 diabetes, as well metabolic and cardiovascular diseases. The use of dietary restriction (DR) while avoiding malnutrition has many profound beneficial effects on aging and metabolic health, and dietary protein or specific amino acid (AA) restrictions, rather than overall calorie intake, are considered to play key roles in the effects of DR on host health. Whereas comprehensive reviews of the underlying mechanisms are limited, protein restriction and methionine (Met) restriction improve metabolic health and aging-related neurodegenerative diseases, and may be associated with FGF21, mTOR and autophagy, improved mitochondrial function and oxidative stress. Circulating branched-chain amino acids (BCAAs) are inversely correlated with metabolic health, and BCAAs and leucine (Leu) restriction promote metabolic homeostasis in rodents. Although tryptophan (Trp) restriction extends the lifespan of rodents, the Trp-restricted diet is reported to increase inflammation in aged mice, while severe Trp restriction has side effects such as anorexia. Furthermore, inadequate protein intake in the elderly increases the risk of muscle-centric health. Therefore, the restriction of specific AAs may be an effective and executable dietary manipulation for metabolic and aging-related health in humans, which warrants further investigation to elucidate the underlying mechanisms.
Collapse
Affiliation(s)
- Danna Wang
- College of Food Science and Engineering, Northwest A&F University, Yangling, China
| | - Jin Ye
- College of Food Science and Engineering, Northwest A&F University, Yangling, China
| | - Renjie Shi
- College of Food Science and Engineering, Northwest A&F University, Yangling, China
| | - Beita Zhao
- College of Food Science and Engineering, Northwest A&F University, Yangling, China
| | - Zhigang Liu
- College of Food Science and Engineering, Northwest A&F University, Yangling, China
| | - Wei Lin
- Department of Neurosurgery, Xijing Institute of Clinical Neuroscience, Xijing Hospital, Air Force Medical University, Xi'an, Shanxi, China.
| | - Xuebo Liu
- College of Food Science and Engineering, Northwest A&F University, Yangling, China.
| |
Collapse
|
9
|
Watkins JD, Koumanov F, Gonzalez JT. Protein- and Calcium-Mediated GLP-1 Secretion: A Narrative Review. Adv Nutr 2021; 12:2540-2552. [PMID: 34192748 PMCID: PMC8634310 DOI: 10.1093/advances/nmab078] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2021] [Revised: 03/31/2021] [Accepted: 05/25/2021] [Indexed: 02/06/2023] Open
Abstract
Glucagon-like peptide 1 (GLP-1) is an incretin hormone produced in the intestine that is secreted in response to nutrient exposure. GLP-1 potentiates glucose-dependent insulin secretion from the pancreatic β cells and promotes satiety. These important actions on glucose metabolism and appetite have led to widespread interest in GLP-1 receptor agonism. Typically, this involves pharmacological GLP-1 mimetics or targeted inhibition of dipeptidyl peptidase-IV, the enzyme responsible for GLP-1 degradation. However, nutritional strategies provide a widely available, cost-effective alternative to pharmacological strategies for enhancing hormone release. Recent advances in nutritional research have implicated the combined ingestion of protein and calcium with enhanced endogenous GLP-1 release, which is likely due to activation of receptors with high affinity and/or sensitivity for amino acids and calcium. Specifically targeting these receptors could enhance gut hormone secretion, thus providing a new therapeutic option. This narrative review provides an overview of the latest research on protein- and calcium-mediated GLP-1 release with an emphasis on human data, and a perspective on potential mechanisms that link potent GLP-1 release to the co-ingestion of protein and calcium. In light of these recent findings, potential future research directions are also presented.
Collapse
Affiliation(s)
- Jonathan D Watkins
- Centre for Nutrition, Exercise and Metabolism, Department for Health, University of Bath, Bath, United Kingdom
| | - Françoise Koumanov
- Centre for Nutrition, Exercise and Metabolism, Department for Health, University of Bath, Bath, United Kingdom
| | | |
Collapse
|
10
|
Ahmad R, Dalziel JE. G Protein-Coupled Receptors in Taste Physiology and Pharmacology. Front Pharmacol 2020; 11:587664. [PMID: 33390961 PMCID: PMC7774309 DOI: 10.3389/fphar.2020.587664] [Citation(s) in RCA: 106] [Impact Index Per Article: 21.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2020] [Accepted: 10/09/2020] [Indexed: 12/14/2022] Open
Abstract
Heterotrimeric G protein-coupled receptors (GPCRs) comprise the largest receptor family in mammals and are responsible for the regulation of most physiological functions. Besides mediating the sensory modalities of olfaction and vision, GPCRs also transduce signals for three basic taste qualities of sweet, umami (savory taste), and bitter, as well as the flavor sensation kokumi. Taste GPCRs reside in specialised taste receptor cells (TRCs) within taste buds. Type I taste GPCRs (TAS1R) form heterodimeric complexes that function as sweet (TAS1R2/TAS1R3) or umami (TAS1R1/TAS1R3) taste receptors, whereas Type II are monomeric bitter taste receptors or kokumi/calcium-sensing receptors. Sweet, umami and kokumi receptors share structural similarities in containing multiple agonist binding sites with pronounced selectivity while most bitter receptors contain a single binding site that is broadly tuned to a diverse array of bitter ligands in a non-selective manner. Tastant binding to the receptor activates downstream secondary messenger pathways leading to depolarization and increased intracellular calcium in TRCs, that in turn innervate the gustatory cortex in the brain. Despite recent advances in our understanding of the relationship between agonist binding and the conformational changes required for receptor activation, several major challenges and questions remain in taste GPCR biology that are discussed in the present review. In recent years, intensive integrative approaches combining heterologous expression, mutagenesis and homology modeling have together provided insight regarding agonist binding site locations and molecular mechanisms of orthosteric and allosteric modulation. In addition, studies based on transgenic mice, utilizing either global or conditional knock out strategies have provided insights to taste receptor signal transduction mechanisms and their roles in physiology. However, the need for more functional studies in a physiological context is apparent and would be enhanced by a crystallized structure of taste receptors for a more complete picture of their pharmacological mechanisms.
Collapse
Affiliation(s)
- Raise Ahmad
- Food Nutrition and Health Team, Food and Bio-based Products Group, AgResearch, Palmerston North, New Zealand
| | - Julie E Dalziel
- Food Nutrition and Health Team, Food and Bio-based Products Group, AgResearch, Palmerston North, New Zealand
| |
Collapse
|
11
|
Danger-Sensing/Patten Recognition Receptors and Neuroinflammation in Alzheimer's Disease. Int J Mol Sci 2020; 21:ijms21239036. [PMID: 33261147 PMCID: PMC7731137 DOI: 10.3390/ijms21239036] [Citation(s) in RCA: 42] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2020] [Revised: 11/24/2020] [Accepted: 11/25/2020] [Indexed: 02/06/2023] Open
Abstract
Fibrillar aggregates and soluble oligomers of both Amyloid-β peptides (Aβs) and hyperphosphorylated Tau proteins (p-Tau-es), as well as a chronic neuroinflammation are the main drivers causing progressive neuronal losses and dementia in Alzheimer’s disease (AD). However, the underlying pathogenetic mechanisms are still much disputed. Several endogenous neurotoxic ligands, including Aβs, and/or p-Tau-es activate innate immunity-related danger-sensing/pattern recognition receptors (PPRs) thereby advancing AD’s neuroinflammation and progression. The major PRR families involved include scavenger, Toll-like, NOD-like, AIM2-like, RIG-like, and CLEC-2 receptors, plus the calcium-sensing receptor (CaSR). This quite intricate picture stresses the need to identify the pathogenetically topmost Aβ-activated PRR, whose signaling would trigger AD’s three main drivers and their intra-brain spread. In theory, the candidate might belong to any PRR family. However, results of preclinical studies using in vitro nontumorigenic human cortical neurons and astrocytes and in vivo AD-model animals have started converging on the CaSR as the pathogenetically upmost PRR candidate. In fact, the CaSR binds both Ca2+ and Aβs and promotes the spread of both Ca2+ dyshomeostasis and AD’s three main drivers, causing a progressive neurons’ death. Since CaSR’s negative allosteric modulators block all these effects, CaSR’s candidacy for topmost pathogenetic PRR has assumed a growing therapeutic potential worth clinical testing.
Collapse
|
12
|
Moustafa EM, Moawed FSM, Abdel-Hamid GR. Icariin Promote Stem Cells Regeneration and Repair Acinar Cells in L-arginine / Radiation -Inducing Chronic Pancreatitis in Rats. Dose Response 2020; 18:1559325820970810. [PMID: 33192204 PMCID: PMC7607780 DOI: 10.1177/1559325820970810] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2020] [Accepted: 09/28/2020] [Indexed: 12/11/2022] Open
Abstract
Objective: Chronic Pancreatitis (CP) is a multifactorial disease. It was characterized by severe inflammation and acinar cell destruction. Thus, the present study was initiated to evaluating the ability of bone marrow-based mesenchymal stem cell (MSCs) combined with Icariin to restore and regenerate acinar cells in the pancreas of rats suffering chronic pancreatitis. Methods: Chronic pancreatitis was induced in rats via both L-arginine plus radiation, repeated L-arginine injection (2.5g/Kg body-weight, 1, 4,7,10,13,16,19 days), then, on day 21, rats were exposed to a single dose of gamma-radiation (6 Gy), which exacerbate injury of pancreatic acinar cells. One day after irradiation, rats were treated with either MSCs (1 × 107 /rat, once, tail vein injection) labeled PKH26 fluorescent linker dye and/or Icariin (100 mg/Kg, daily, orally) for 8 weeks. Results: Icariin promotes MSCs proliferation boosting its productivity in vitro. MSCs, and/or icariin treatments has regulated molecular factors TGF-β/PDGF and promoted the regeneration of pancreatic tissues by releasing PDX-1 and MafA involved in the recruitment of stem/progenitor cell in the tissue, and confirmed by histopathological examination. Moreover, a significant decrease in IL-8 and TNF-α cytokines with significant amelioration of myeloperoxidase activity were noted. As well as, reduction in MCP-1 and collagen type-1 levels along with Hedgehog signaling down-regulating expression in such cells, Patched-1, Smoothened, and GLi-1. Conclusion: The potent bioactive therapeutic Icariin combined with MSCs induces a significantly greater improvement, compared to each therapy alone.
Collapse
Affiliation(s)
| | - Fatma S M Moawed
- Department of Health Radiation Research, National Center for Radiation Research & Technology (NCRRT), Egyptian Atomic Energy Authority (EAEA), Cairo, Egypt
| | | |
Collapse
|
13
|
Schatz M, Saravanan S, d'Adesky ND, Bramlett H, Perez-Pinzon MA, Raval AP. Osteocalcin, ovarian senescence, and brain health. Front Neuroendocrinol 2020; 59:100861. [PMID: 32781196 DOI: 10.1016/j.yfrne.2020.100861] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/23/2020] [Revised: 07/20/2020] [Accepted: 08/05/2020] [Indexed: 12/19/2022]
Abstract
Menopause, an inevitable event in a woman's life, significantly increases risk of bone resorption and diseases such as Alzheimer's, vascular dementia, cardiac arrest, and stroke. The sole role of bones, as traditionally regarded, is to provide structural support for skeletal muscles and allow for ambulation, however this concept is becoming quickly outdated. New literature has emerged that suggests the bone cell-derived hormone osteocalcin (OCN) plays a pivotal role in cognition. OCN levels are correlated with bone mass density and bone turnover, and thus are strongly influenced by the changes associated with menopause. The goal of the current review is to discuss potential gaps in our knowledge of OCN and cognition, discrepancies in methods of OCN quantification, and therapies to enhance circulating OCN. A discussion on implementing exercise or low frequency vibration interventions at the menopausal transition to reduce risk and severity of neurological diseases and associated cognitive decline is included.
Collapse
Affiliation(s)
- Marc Schatz
- Department of Orthopaedic Surgery, Louisiana State University Health Sciences Center, New Orleans, LA 70112, USA
| | - Sharnikha Saravanan
- Peritz Scheinberg Cerebral Vascular Disease Research Laboratory, Department of Neurology, Miami, FL 33136, USA
| | - Nathan D d'Adesky
- Peritz Scheinberg Cerebral Vascular Disease Research Laboratory, Department of Neurology, Miami, FL 33136, USA
| | - Helen Bramlett
- Department of Neurological Surgery, Leonard M. Miller School of Medicine, University of Miami, Miami, FL 33136, USA; Bruce W. Carter Department of Veterans Affairs Medical Center, Miami, FL 33136, USA
| | - Miguel A Perez-Pinzon
- Peritz Scheinberg Cerebral Vascular Disease Research Laboratory, Department of Neurology, Miami, FL 33136, USA
| | - Ami P Raval
- Peritz Scheinberg Cerebral Vascular Disease Research Laboratory, Department of Neurology, Miami, FL 33136, USA.
| |
Collapse
|
14
|
Leach K, Hannan FM, Josephs TM, Keller AN, Møller TC, Ward DT, Kallay E, Mason RS, Thakker RV, Riccardi D, Conigrave AD, Bräuner-Osborne H. International Union of Basic and Clinical Pharmacology. CVIII. Calcium-Sensing Receptor Nomenclature, Pharmacology, and Function. Pharmacol Rev 2020; 72:558-604. [PMID: 32467152 PMCID: PMC7116503 DOI: 10.1124/pr.119.018531] [Citation(s) in RCA: 59] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
The calcium-sensing receptor (CaSR) is a class C G protein-coupled receptor that responds to multiple endogenous agonists and allosteric modulators, including divalent and trivalent cations, L-amino acids, γ-glutamyl peptides, polyamines, polycationic peptides, and protons. The CaSR plays a critical role in extracellular calcium (Ca2+ o) homeostasis, as demonstrated by the many naturally occurring mutations in the CaSR or its signaling partners that cause Ca2+ o homeostasis disorders. However, CaSR tissue expression in mammals is broad and includes tissues unrelated to Ca2+ o homeostasis, in which it, for example, regulates the secretion of digestive hormones, airway constriction, cardiovascular effects, cellular differentiation, and proliferation. Thus, although the CaSR is targeted clinically by the positive allosteric modulators (PAMs) cinacalcet, evocalcet, and etelcalcetide in hyperparathyroidism, it is also a putative therapeutic target in diabetes, asthma, cardiovascular disease, and cancer. The CaSR is somewhat unique in possessing multiple ligand binding sites, including at least five putative sites for the "orthosteric" agonist Ca2+ o, an allosteric site for endogenous L-amino acids, two further allosteric sites for small molecules and the peptide PAM, etelcalcetide, and additional sites for other cations and anions. The CaSR is promiscuous in its G protein-coupling preferences, and signals via Gq/11, Gi/o, potentially G12/13, and even Gs in some cell types. Not surprisingly, the CaSR is subject to biased agonism, in which distinct ligands preferentially stimulate a subset of the CaSR's possible signaling responses, to the exclusion of others. The CaSR thus serves as a model receptor to study natural bias and allostery. SIGNIFICANCE STATEMENT: The calcium-sensing receptor (CaSR) is a complex G protein-coupled receptor that possesses multiple orthosteric and allosteric binding sites, is subject to biased signaling via several different G proteins, and has numerous (patho)physiological roles. Understanding the complexities of CaSR structure, function, and biology will aid future drug discovery efforts seeking to target this receptor for a diversity of diseases. This review summarizes what is known to date regarding key structural, pharmacological, and physiological features of the CaSR.
Collapse
Affiliation(s)
- Katie Leach
- Drug Discovery Biology, Monash Institute of Pharmaceutical Science, Monash University, Parkville, Australia (K.L., T.M.J., A.N.K.); Nuffield Department of Women's & Reproductive Health (F.M.H.) and Academic Endocrine Unit, Radcliffe Department of Clinical Medicine (F.M.H., R.V.T.), University of Oxford, Oxford, United Kingdom; Department of Drug Design and Pharmacology, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark (T.C.M., H.B.-O.); Faculty of Biology, Medicine and Health, The University of Manchester, Manchester, United Kingdom (D.T.W.); Department of Pathophysiology and Allergy Research, Medical University of Vienna, Vienna, Austria (E.K.); Physiology, School of Medical Sciences and Bosch Institute (R.S.M.) and School of Life & Environmental Sciences, Charles Perkins Centre (A.D.C.), University of Sydney, Sydney, Australia; and School of Biosciences, Cardiff University, Cardiff, United Kingdom (D.R.)
| | - Fadil M Hannan
- Drug Discovery Biology, Monash Institute of Pharmaceutical Science, Monash University, Parkville, Australia (K.L., T.M.J., A.N.K.); Nuffield Department of Women's & Reproductive Health (F.M.H.) and Academic Endocrine Unit, Radcliffe Department of Clinical Medicine (F.M.H., R.V.T.), University of Oxford, Oxford, United Kingdom; Department of Drug Design and Pharmacology, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark (T.C.M., H.B.-O.); Faculty of Biology, Medicine and Health, The University of Manchester, Manchester, United Kingdom (D.T.W.); Department of Pathophysiology and Allergy Research, Medical University of Vienna, Vienna, Austria (E.K.); Physiology, School of Medical Sciences and Bosch Institute (R.S.M.) and School of Life & Environmental Sciences, Charles Perkins Centre (A.D.C.), University of Sydney, Sydney, Australia; and School of Biosciences, Cardiff University, Cardiff, United Kingdom (D.R.)
| | - Tracy M Josephs
- Drug Discovery Biology, Monash Institute of Pharmaceutical Science, Monash University, Parkville, Australia (K.L., T.M.J., A.N.K.); Nuffield Department of Women's & Reproductive Health (F.M.H.) and Academic Endocrine Unit, Radcliffe Department of Clinical Medicine (F.M.H., R.V.T.), University of Oxford, Oxford, United Kingdom; Department of Drug Design and Pharmacology, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark (T.C.M., H.B.-O.); Faculty of Biology, Medicine and Health, The University of Manchester, Manchester, United Kingdom (D.T.W.); Department of Pathophysiology and Allergy Research, Medical University of Vienna, Vienna, Austria (E.K.); Physiology, School of Medical Sciences and Bosch Institute (R.S.M.) and School of Life & Environmental Sciences, Charles Perkins Centre (A.D.C.), University of Sydney, Sydney, Australia; and School of Biosciences, Cardiff University, Cardiff, United Kingdom (D.R.)
| | - Andrew N Keller
- Drug Discovery Biology, Monash Institute of Pharmaceutical Science, Monash University, Parkville, Australia (K.L., T.M.J., A.N.K.); Nuffield Department of Women's & Reproductive Health (F.M.H.) and Academic Endocrine Unit, Radcliffe Department of Clinical Medicine (F.M.H., R.V.T.), University of Oxford, Oxford, United Kingdom; Department of Drug Design and Pharmacology, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark (T.C.M., H.B.-O.); Faculty of Biology, Medicine and Health, The University of Manchester, Manchester, United Kingdom (D.T.W.); Department of Pathophysiology and Allergy Research, Medical University of Vienna, Vienna, Austria (E.K.); Physiology, School of Medical Sciences and Bosch Institute (R.S.M.) and School of Life & Environmental Sciences, Charles Perkins Centre (A.D.C.), University of Sydney, Sydney, Australia; and School of Biosciences, Cardiff University, Cardiff, United Kingdom (D.R.)
| | - Thor C Møller
- Drug Discovery Biology, Monash Institute of Pharmaceutical Science, Monash University, Parkville, Australia (K.L., T.M.J., A.N.K.); Nuffield Department of Women's & Reproductive Health (F.M.H.) and Academic Endocrine Unit, Radcliffe Department of Clinical Medicine (F.M.H., R.V.T.), University of Oxford, Oxford, United Kingdom; Department of Drug Design and Pharmacology, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark (T.C.M., H.B.-O.); Faculty of Biology, Medicine and Health, The University of Manchester, Manchester, United Kingdom (D.T.W.); Department of Pathophysiology and Allergy Research, Medical University of Vienna, Vienna, Austria (E.K.); Physiology, School of Medical Sciences and Bosch Institute (R.S.M.) and School of Life & Environmental Sciences, Charles Perkins Centre (A.D.C.), University of Sydney, Sydney, Australia; and School of Biosciences, Cardiff University, Cardiff, United Kingdom (D.R.)
| | - Donald T Ward
- Drug Discovery Biology, Monash Institute of Pharmaceutical Science, Monash University, Parkville, Australia (K.L., T.M.J., A.N.K.); Nuffield Department of Women's & Reproductive Health (F.M.H.) and Academic Endocrine Unit, Radcliffe Department of Clinical Medicine (F.M.H., R.V.T.), University of Oxford, Oxford, United Kingdom; Department of Drug Design and Pharmacology, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark (T.C.M., H.B.-O.); Faculty of Biology, Medicine and Health, The University of Manchester, Manchester, United Kingdom (D.T.W.); Department of Pathophysiology and Allergy Research, Medical University of Vienna, Vienna, Austria (E.K.); Physiology, School of Medical Sciences and Bosch Institute (R.S.M.) and School of Life & Environmental Sciences, Charles Perkins Centre (A.D.C.), University of Sydney, Sydney, Australia; and School of Biosciences, Cardiff University, Cardiff, United Kingdom (D.R.)
| | - Enikö Kallay
- Drug Discovery Biology, Monash Institute of Pharmaceutical Science, Monash University, Parkville, Australia (K.L., T.M.J., A.N.K.); Nuffield Department of Women's & Reproductive Health (F.M.H.) and Academic Endocrine Unit, Radcliffe Department of Clinical Medicine (F.M.H., R.V.T.), University of Oxford, Oxford, United Kingdom; Department of Drug Design and Pharmacology, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark (T.C.M., H.B.-O.); Faculty of Biology, Medicine and Health, The University of Manchester, Manchester, United Kingdom (D.T.W.); Department of Pathophysiology and Allergy Research, Medical University of Vienna, Vienna, Austria (E.K.); Physiology, School of Medical Sciences and Bosch Institute (R.S.M.) and School of Life & Environmental Sciences, Charles Perkins Centre (A.D.C.), University of Sydney, Sydney, Australia; and School of Biosciences, Cardiff University, Cardiff, United Kingdom (D.R.)
| | - Rebecca S Mason
- Drug Discovery Biology, Monash Institute of Pharmaceutical Science, Monash University, Parkville, Australia (K.L., T.M.J., A.N.K.); Nuffield Department of Women's & Reproductive Health (F.M.H.) and Academic Endocrine Unit, Radcliffe Department of Clinical Medicine (F.M.H., R.V.T.), University of Oxford, Oxford, United Kingdom; Department of Drug Design and Pharmacology, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark (T.C.M., H.B.-O.); Faculty of Biology, Medicine and Health, The University of Manchester, Manchester, United Kingdom (D.T.W.); Department of Pathophysiology and Allergy Research, Medical University of Vienna, Vienna, Austria (E.K.); Physiology, School of Medical Sciences and Bosch Institute (R.S.M.) and School of Life & Environmental Sciences, Charles Perkins Centre (A.D.C.), University of Sydney, Sydney, Australia; and School of Biosciences, Cardiff University, Cardiff, United Kingdom (D.R.)
| | - Rajesh V Thakker
- Drug Discovery Biology, Monash Institute of Pharmaceutical Science, Monash University, Parkville, Australia (K.L., T.M.J., A.N.K.); Nuffield Department of Women's & Reproductive Health (F.M.H.) and Academic Endocrine Unit, Radcliffe Department of Clinical Medicine (F.M.H., R.V.T.), University of Oxford, Oxford, United Kingdom; Department of Drug Design and Pharmacology, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark (T.C.M., H.B.-O.); Faculty of Biology, Medicine and Health, The University of Manchester, Manchester, United Kingdom (D.T.W.); Department of Pathophysiology and Allergy Research, Medical University of Vienna, Vienna, Austria (E.K.); Physiology, School of Medical Sciences and Bosch Institute (R.S.M.) and School of Life & Environmental Sciences, Charles Perkins Centre (A.D.C.), University of Sydney, Sydney, Australia; and School of Biosciences, Cardiff University, Cardiff, United Kingdom (D.R.)
| | - Daniela Riccardi
- Drug Discovery Biology, Monash Institute of Pharmaceutical Science, Monash University, Parkville, Australia (K.L., T.M.J., A.N.K.); Nuffield Department of Women's & Reproductive Health (F.M.H.) and Academic Endocrine Unit, Radcliffe Department of Clinical Medicine (F.M.H., R.V.T.), University of Oxford, Oxford, United Kingdom; Department of Drug Design and Pharmacology, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark (T.C.M., H.B.-O.); Faculty of Biology, Medicine and Health, The University of Manchester, Manchester, United Kingdom (D.T.W.); Department of Pathophysiology and Allergy Research, Medical University of Vienna, Vienna, Austria (E.K.); Physiology, School of Medical Sciences and Bosch Institute (R.S.M.) and School of Life & Environmental Sciences, Charles Perkins Centre (A.D.C.), University of Sydney, Sydney, Australia; and School of Biosciences, Cardiff University, Cardiff, United Kingdom (D.R.)
| | - Arthur D Conigrave
- Drug Discovery Biology, Monash Institute of Pharmaceutical Science, Monash University, Parkville, Australia (K.L., T.M.J., A.N.K.); Nuffield Department of Women's & Reproductive Health (F.M.H.) and Academic Endocrine Unit, Radcliffe Department of Clinical Medicine (F.M.H., R.V.T.), University of Oxford, Oxford, United Kingdom; Department of Drug Design and Pharmacology, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark (T.C.M., H.B.-O.); Faculty of Biology, Medicine and Health, The University of Manchester, Manchester, United Kingdom (D.T.W.); Department of Pathophysiology and Allergy Research, Medical University of Vienna, Vienna, Austria (E.K.); Physiology, School of Medical Sciences and Bosch Institute (R.S.M.) and School of Life & Environmental Sciences, Charles Perkins Centre (A.D.C.), University of Sydney, Sydney, Australia; and School of Biosciences, Cardiff University, Cardiff, United Kingdom (D.R.)
| | - Hans Bräuner-Osborne
- Drug Discovery Biology, Monash Institute of Pharmaceutical Science, Monash University, Parkville, Australia (K.L., T.M.J., A.N.K.); Nuffield Department of Women's & Reproductive Health (F.M.H.) and Academic Endocrine Unit, Radcliffe Department of Clinical Medicine (F.M.H., R.V.T.), University of Oxford, Oxford, United Kingdom; Department of Drug Design and Pharmacology, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark (T.C.M., H.B.-O.); Faculty of Biology, Medicine and Health, The University of Manchester, Manchester, United Kingdom (D.T.W.); Department of Pathophysiology and Allergy Research, Medical University of Vienna, Vienna, Austria (E.K.); Physiology, School of Medical Sciences and Bosch Institute (R.S.M.) and School of Life & Environmental Sciences, Charles Perkins Centre (A.D.C.), University of Sydney, Sydney, Australia; and School of Biosciences, Cardiff University, Cardiff, United Kingdom (D.R.)
| |
Collapse
|
15
|
Chiarini A, Armato U, Hu P, Dal Prà I. CaSR Antagonist (Calcilytic) NPS 2143 Hinders the Release of Neuroinflammatory IL-6, Soluble ICAM-1, RANTES, and MCP-2 from Aβ-Exposed Human Cortical Astrocytes. Cells 2020; 9:cells9061386. [PMID: 32498476 PMCID: PMC7349863 DOI: 10.3390/cells9061386] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2020] [Revised: 05/29/2020] [Accepted: 05/29/2020] [Indexed: 02/07/2023] Open
Abstract
Available evidence shows that human cortical neurons’ and astrocytes’ calcium-sensing receptors (CaSRs) bind Amyloid-beta (Aβ) oligomers triggering the overproduction/oversecretion of several Alzheimer’s disease (AD) neurotoxins—effects calcilytics suppress. We asked whether Aβ•CaSR signaling might also play a direct pro-neuroinflammatory role in AD. Cortical nontumorigenic adult human astrocytes (NAHAs) in vitro were untreated (controls) or treated with Aβ25–35 ± NPS 2143 (a calcilytic) and any proinflammatory agent in their protein lysates and growth media assayed via antibody arrays, enzyme-linked immunosorbent assays (ELISAs), and immunoblots. Results show Aβ•CaSR signaling upregulated the synthesis and release/shedding of proinflammatory interleukin (IL)-6, intercellular adhesion molecule-1 (ICAM-1) (holoprotein and soluble [s] fragment), Regulated upon Activation, normal T cell Expressed and presumably Secreted (RANTES), and monocyte chemotactic protein (MCP)-2. Adding NPS 2143 (i) totally suppressed IL-6′s oversecretion while remarkably reducing the other agents’ over-release; and (ii) more effectively than Aβ alone increased over controls the four agents’ distinctive intracellular accumulation. Conversely, NPS 2143 did not alter Aβ-induced surges in IL-1β, IL-3, IL-8, and IL-16 secretion, consequently revealing their Aβ•CaSR signaling-independence. Finally, Aβ25–35 ± NPS 2143 treatments left unchanged MCP-1′s and TIMP-2′s basal expression. Thus, NAHAs Aβ•CaSR signaling drove four proinflammatory agents’ over-release that NPS 2143 curtailed. Therefore, calcilytics would also abate NAHAs’ Aβ•CaSR signaling direct impact on AD’s neuroinflammation.
Collapse
Affiliation(s)
- Anna Chiarini
- Human Histology and Embryology Section, Department of Surgery, Dentistry, Pediatrics and Gynecology, Medical School, University of Verona, Veneto, 37134 Verona, Italy; (U.A.); (P.H.)
- Correspondence: (A.C.); (I.D.P.); Tel.: +39-045-802-7646 (A.C.); +39-045-802-7161 (I.D.P)
| | - Ubaldo Armato
- Human Histology and Embryology Section, Department of Surgery, Dentistry, Pediatrics and Gynecology, Medical School, University of Verona, Veneto, 37134 Verona, Italy; (U.A.); (P.H.)
- Burns Department, Shenzhen Second People’s Hospital, University of Shenzhen, Shenzhen 518000, China
| | - Peng Hu
- Human Histology and Embryology Section, Department of Surgery, Dentistry, Pediatrics and Gynecology, Medical School, University of Verona, Veneto, 37134 Verona, Italy; (U.A.); (P.H.)
| | - Ilaria Dal Prà
- Human Histology and Embryology Section, Department of Surgery, Dentistry, Pediatrics and Gynecology, Medical School, University of Verona, Veneto, 37134 Verona, Italy; (U.A.); (P.H.)
- Burns Department, Shenzhen Second People’s Hospital, University of Shenzhen, Shenzhen 518000, China
- Correspondence: (A.C.); (I.D.P.); Tel.: +39-045-802-7646 (A.C.); +39-045-802-7161 (I.D.P)
| |
Collapse
|
16
|
Guo D, He H, Hou T. Purification and characterization of positive allosteric regulatory peptides of calcium sensing receptor (CaSR) from desalted duck egg white. Food Chem 2020; 325:126919. [PMID: 32387992 DOI: 10.1016/j.foodchem.2020.126919] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2019] [Revised: 04/21/2020] [Accepted: 04/25/2020] [Indexed: 02/07/2023]
Abstract
HPLC-ESI-MS/MS, molecular docking simulation and in situ single-pass intestinal perfusion (SPIP) study were used to identify, select, and confirm the binding affinities between peptides identified from desalted duck egg white peptides (DPs) and calcium sensing receptor (CaSR), respectively. F3 fraction from DPs possessed superior calcium binding activity (P < 0.05), and 16 peptides enriched aromatic amino acids and other 33 peptides were identified. FAE, FNE, INSW, FDPE and NFE presented well binding affinities with CaSR in molecular docking. Additionally, SPIP results showed that NFE and INSW significantly reduced the increased PTH levels by 45.8% and 48.8%, respectively (P < 0.05), and increased calcium percent absorption, calcium absorption rate constant (Ka) and calcium effective permeability (Peff) (P < 0.05), as well as up-regulated mRNA levels of CaSR (P < 0.05). Moreover, NFE and INSW could interact with the VFT domain of CaSR, which exhibited the potential activities in regulation of CaSR.
Collapse
Affiliation(s)
- Danjun Guo
- College of Food Science and Technology, Huazhong Agricultural University, Wuhan 430070, China
| | - Hui He
- College of Food Science and Technology, Huazhong Agricultural University, Wuhan 430070, China.
| | - Tao Hou
- College of Food Science and Technology, Huazhong Agricultural University, Wuhan 430070, China.
| |
Collapse
|
17
|
Jørgensen CV, Bräuner‐Osborne H. Pharmacology and physiological function of the orphan GPRC6A receptor. Basic Clin Pharmacol Toxicol 2020; 126 Suppl 6:77-87. [DOI: 10.1111/bcpt.13397] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2019] [Revised: 02/11/2020] [Accepted: 02/11/2020] [Indexed: 12/17/2022]
Affiliation(s)
- Christinna V. Jørgensen
- Department of Drug Design and Pharmacology Faculty of Health and Medical Sciences University of Copenhagen Copenhagen Denmark
| | - Hans Bräuner‐Osborne
- Department of Drug Design and Pharmacology Faculty of Health and Medical Sciences University of Copenhagen Copenhagen Denmark
| |
Collapse
|
18
|
Fonin AV, Darling AL, Kuznetsova IM, Turoverov KK, Uversky VN. Multi-functionality of proteins involved in GPCR and G protein signaling: making sense of structure-function continuum with intrinsic disorder-based proteoforms. Cell Mol Life Sci 2019; 76:4461-4492. [PMID: 31428838 PMCID: PMC11105632 DOI: 10.1007/s00018-019-03276-1] [Citation(s) in RCA: 46] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2019] [Revised: 08/05/2019] [Accepted: 08/12/2019] [Indexed: 12/21/2022]
Abstract
GPCR-G protein signaling system recognizes a multitude of extracellular ligands and triggers a variety of intracellular signaling cascades in response. In humans, this system includes more than 800 various GPCRs and a large set of heterotrimeric G proteins. Complexity of this system goes far beyond a multitude of pair-wise ligand-GPCR and GPCR-G protein interactions. In fact, one GPCR can recognize more than one extracellular signal and interact with more than one G protein. Furthermore, one ligand can activate more than one GPCR, and multiple GPCRs can couple to the same G protein. This defines an intricate multifunctionality of this important signaling system. Here, we show that the multifunctionality of GPCR-G protein system represents an illustrative example of the protein structure-function continuum, where structures of the involved proteins represent a complex mosaic of differently folded regions (foldons, non-foldons, unfoldons, semi-foldons, and inducible foldons). The functionality of resulting highly dynamic conformational ensembles is fine-tuned by various post-translational modifications and alternative splicing, and such ensembles can undergo dramatic changes at interaction with their specific partners. In other words, GPCRs and G proteins exist as sets of conformational/basic, inducible/modified, and functioning proteoforms characterized by a broad spectrum of structural features and possessing various functional potentials.
Collapse
Affiliation(s)
- Alexander V Fonin
- Laboratory of structural Dynamics, Stability and Folding of Proteins, Institute of Cytology, Russian Academy of Sciences, St. Petersburg, 194064, Russian Federation
| | - April L Darling
- Department of Molecular Medicine and USF Health Byrd Alzheimer's Research Institute, Morsani College of Medicine, University of South Florida, Tampa, FL, USA
| | - Irina M Kuznetsova
- Laboratory of structural Dynamics, Stability and Folding of Proteins, Institute of Cytology, Russian Academy of Sciences, St. Petersburg, 194064, Russian Federation
| | - Konstantin K Turoverov
- Laboratory of structural Dynamics, Stability and Folding of Proteins, Institute of Cytology, Russian Academy of Sciences, St. Petersburg, 194064, Russian Federation
- Department of Biophysics, Peter the Great St. Petersburg Polytechnic University, Polytechnicheskaya av. 29, St. Petersburg, 195251, Russian Federation
| | - Vladimir N Uversky
- Department of Molecular Medicine and USF Health Byrd Alzheimer's Research Institute, Morsani College of Medicine, University of South Florida, Tampa, FL, USA.
- Institute for Biological Instrumentation, Russian Academy of Sciences, Pushchino, Moscow, Russian Federation.
| |
Collapse
|
19
|
Dal Prà I, Armato U, Chiarini A. Family C G-Protein-Coupled Receptors in Alzheimer's Disease and Therapeutic Implications. Front Pharmacol 2019; 10:1282. [PMID: 31719824 PMCID: PMC6826475 DOI: 10.3389/fphar.2019.01282] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2019] [Accepted: 10/07/2019] [Indexed: 12/12/2022] Open
Abstract
Alzheimer’s disease (AD), particularly its sporadic or late-onset form (SAD/LOAD), is the most prevalent (96–98% of cases) neurodegenerative dementia in aged people. AD’s neuropathology hallmarks are intrabrain accumulation of amyloid-β peptides (Aβs) and of hyperphosphorylated Tau (p-Tau) proteins, diffuse neuroinflammation, and progressive death of neurons and oligodendrocytes. Mounting evidences suggest that family C G-protein-coupled receptors (GPCRs), which include γ-aminobutyric acid B receptors (GABABRs), metabotropic glutamate receptors (mGluR1-8), and the calcium-sensing receptor (CaSR), are involved in many neurotransmitter systems that dysfunction in AD. This review updates the available knowledge about the roles of GPCRs, particularly but not exclusively those expressed by brain astrocytes, in SAD/LOAD onset and progression, taking stock of their respective mechanisms of action and of their potential as anti-AD therapeutic targets. In particular, GABABRs prevent Aβs synthesis and neuronal hyperexcitability and group I mGluRs play important pathogenetic roles in transgenic AD-model animals. Moreover, the specific binding of Aβs to the CaSRs of human cortical astrocytes and neurons cultured in vitro engenders a pathological signaling that crucially promotes the surplus synthesis and release of Aβs and hyperphosphorylated Tau proteins, and also of nitric oxide, vascular endothelial growth factor-A, and proinflammatory agents. Concurrently, Aβs•CaSR signaling hinders the release of soluble (s)APP-α peptide, a neurotrophic agent and GABABR1a agonist. Altogether these effects progressively kill human cortical neurons in vitro and likely also in vivo. Several CaSR’s negative allosteric modulators suppress all the noxious effects elicited by Aβs•CaSR signaling in human cortical astrocytes and neurons thus safeguarding neurons’ viability in vitro and raising hopes about their potential therapeutic benefits in AD patients. Further basic and clinical investigations on these hot topics are needed taking always heed that activation of the several brain family C GPCRs may elicit divergent upshots according to the models studied.
Collapse
Affiliation(s)
- Ilaria Dal Prà
- Human Histology and Embryology Unit, University of Verona Medical School, Verona, Italy
| | - Ubaldo Armato
- Human Histology and Embryology Unit, University of Verona Medical School, Verona, Italy
| | - Anna Chiarini
- Human Histology and Embryology Unit, University of Verona Medical School, Verona, Italy
| |
Collapse
|
20
|
Capiod T, Barry Delongchamps N, Pigat N, Souberbielle JC, Goffin V. Do dietary calcium and vitamin D matter in men with prostate cancer? Nat Rev Urol 2019; 15:453-461. [PMID: 29765146 DOI: 10.1038/s41585-018-0015-z] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
Active surveillance (AS) is an attractive alternative to immediate treatment for men with low-risk prostate cancer. Thus, the identification of environmental factors that promote the progression of indolent disease towards aggressive stages is critical to optimize clinical management. Epidemiological studies suggest that calcium-rich diets contribute to an increased risk of developing prostate cancer and that vitamin D reduces this risk. However, the potential effect of these nutrients on the progression of early-stage prostate tumours is uncertain, as studies in this setting are scarce and have not provided unambiguous conclusions. By contrast, the results of a preclinical study from our own group demonstrate that a diet high in calcium dose-dependently accelerated the progression of early-stage prostate tumours and that dietary vitamin D prevented this effect. The extent to which the conclusions of preclinical and epidemiological studies support a role for calcium and vitamin D and the relevance of monitoring and adjustment of calcium and/or vitamin D intake in patients on AS require further investigation.
Collapse
Affiliation(s)
- Thierry Capiod
- Inserm Unit 1151, Institut Necker-Enfants Malades (INEM), Université Paris Descartes, Paris, France
| | - Nicolas Barry Delongchamps
- Inserm Unit 1151, Institut Necker-Enfants Malades (INEM), Université Paris Descartes, Paris, France.,Urology Department, Hôpital Cochin, Assistance Publique Hôpitaux de Paris, Paris, France
| | - Natascha Pigat
- Inserm Unit 1151, Institut Necker-Enfants Malades (INEM), Université Paris Descartes, Paris, France
| | - Jean-Claude Souberbielle
- Inserm Unit 1151, Institut Necker-Enfants Malades (INEM), Université Paris Descartes, Paris, France.,Physiology Department, Hôpital Necker-Enfants Malades, Assistance Publique Hôpitaux de Paris, Paris, France
| | - Vincent Goffin
- Inserm Unit 1151, Institut Necker-Enfants Malades (INEM), Université Paris Descartes, Paris, France.
| |
Collapse
|
21
|
Mun HC, Leach KM, Conigrave AD. L-Amino Acids Promote Calcitonin Release via a Calcium-Sensing Receptor: Gq/11-Mediated Pathway in Human C-Cells. Endocrinology 2019; 160:1590-1599. [PMID: 31127815 DOI: 10.1210/en.2018-00860] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/02/2018] [Accepted: 04/19/2019] [Indexed: 11/19/2022]
Abstract
Human calcitonin release is promoted by elevated extracellular Ca2+ (Ca2+o) concentration acting, at least in part, via the calcium-sensing receptor (CaSR). The CaSR is positively modulated by L-amino acids, including the aromatic amino acids L-phenylalanine (Phe) and L-tryptophan (Trp). To investigate the effect of L-amino acids on human calcitonin secretion, we selected thyroid TT cells and exposed them to various Ca2+o concentrations in the absence or presence of L-Phe, plasma-like mixtures of L-amino acids, or the clinically effective positive modulator (calcimimetic) cinacalcet. In the presence of L-Phe or plasma-like mixtures of amino acids, TT cells exhibited enhanced Ca2+o sensitivity in assays of calcitonin release and intracellular Ca2+ mobilization. Furthermore, the effect of elevated Ca2+o and L-Phe on calcitonin release was markedly suppressed by the calcilytic NPS-2143. These effects were dependent on CaSR-mediated activation of Gq/11 as revealed by the specific inhibitor YM-254890. The findings support the hypothesis that calcitonin release is stimulated by increases in plasma L-amino acid levels as well as elevated Ca2+o concentration. They also demonstrate that stimulated calcitonin release as well as basal levels of calcitonin secretion are mediated by a CaSR:Gq/11 signaling mechanism.
Collapse
Affiliation(s)
- Hee-Chang Mun
- School of Life and Environmental Sciences, Charles Perkins Centre (D17), University of Sydney, Sydney, New South Wales, Australia
| | - Katie M Leach
- Drug Discovery Biology and Department of Pharmacology, Monash Institute of Pharmaceutical Sciences, Parkville, Victoria, Australia
| | - Arthur D Conigrave
- School of Life and Environmental Sciences, Charles Perkins Centre (D17), University of Sydney, Sydney, New South Wales, Australia
| |
Collapse
|
22
|
Anract J, Baures M, Barry Delongchamps N, Capiod T. Microcalcifications, calcium-sensing receptor, and cancer. Cell Calcium 2019; 82:102051. [PMID: 31276858 DOI: 10.1016/j.ceca.2019.06.005] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2019] [Revised: 06/13/2019] [Accepted: 06/13/2019] [Indexed: 12/20/2022]
Abstract
Calcium stones and calculi are observed in numerous human tissues. They are the result of deposition of calcium salts and are due to high local calcium concentrations. Prostatic calculi are usually classified as endogenous or extrinsic stones. Endogenous stones are commonly caused by obstruction of the prostatic ducts around an enlarged prostate resulting from benign prostatic hyperplasia or from chronic inflammation. The latter occurs mainly around the urethra and is generally caused by reflux of urine into the prostate. Calcium concentrations higher than in the plasma at sites of infection may induce the chemotactic response that eventually leads to recruitment of inflammatory cells. The calcium sensing receptor (CaSR) may be crucial for this recruitment as its expression and activity are increased by cytokines such as IL-6 and high extracellular calcium concentrations, respectively. The links between calcium calculi, inflammation, calcium supplementation, and CaSR functions in prostate cancer patients will be discussed in this review.
Collapse
Affiliation(s)
- Julien Anract
- INSERM Unit 1151, Institut Necker Enfants Malades (INEM), Université Paris Descartes, Paris 75014, France; Urology Department, Hôpital Cochin, Assistance Publique Hôpitaux de Paris, Paris 75014, France
| | - Manon Baures
- INSERM Unit 1151, Institut Necker Enfants Malades (INEM), Université Paris Descartes, Paris 75014, France
| | - Nicolas Barry Delongchamps
- INSERM Unit 1151, Institut Necker Enfants Malades (INEM), Université Paris Descartes, Paris 75014, France; Urology Department, Hôpital Cochin, Assistance Publique Hôpitaux de Paris, Paris 75014, France
| | - Thierry Capiod
- INSERM Unit 1151, Institut Necker Enfants Malades (INEM), Université Paris Descartes, Paris 75014, France.
| |
Collapse
|
23
|
Modvig IM, Kuhre RE, Holst JJ. Peptone-mediated glucagon-like peptide-1 secretion depends on intestinal absorption and activation of basolaterally located Calcium-Sensing Receptors. Physiol Rep 2019; 7:e14056. [PMID: 31020803 PMCID: PMC6482282 DOI: 10.14814/phy2.14056] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2019] [Revised: 03/15/2019] [Accepted: 03/16/2019] [Indexed: 01/13/2023] Open
Abstract
Protein intake robustly stimulates the secretion of the incretin hormone, glucagon-like peptide-1 (GLP-1) but the molecular mechanisms involved are not well understood. In particular, it is unknown whether proteins stimulate secretion by activation of luminal or basolateral sensors. We characterized the mechanisms using a physiologically relevant model - the isolated perfused proximal rat small intestine. Intraluminal protein hydrolysates derived from meat (peptone; 50 mg/mL) increased GLP-1 secretion 2.3-fold (from a basal secretion of 110 ± 28 fmol/min). The sensory mechanisms underlying the response depended on di/tripeptide uptake through Peptide Transporter 1 (PepT1) and subsequent basolateral activation of the amino acid sensing receptor, Calcium-Sensing Receptor (CaSR), since inhibition of PepT1 as well as CaSR both attenuated the peptone-induced GLP-1 response. Supporting this, intraluminal peptones were absorbed efficiently by the perfused intestine (resulting in increased amino acid concentrations in the venous effluent) and infusion of amino acids robustly stimulated GLP-1 secretion. Inhibitors of voltage-gated L-type Ca2+ channels had no effect on secretion suggesting that peptone-mediated GLP-1 secretion is not mediated by L-cell depolarization with subsequent opening of these channels. Specific targeting of CaSR could serve as a target to stimulate the endogenous secretion of GLP-1.
Collapse
Affiliation(s)
- Ida M. Modvig
- Department of Biomedical SciencesNNF Center for Basic Metabolic ResearchFaculty of Health and Medical SciencesUniversity of CopenhagenCopenhagenDenmark
| | - Rune E. Kuhre
- Department of Biomedical SciencesNNF Center for Basic Metabolic ResearchFaculty of Health and Medical SciencesUniversity of CopenhagenCopenhagenDenmark
| | - Jens Juul Holst
- Department of Biomedical SciencesNNF Center for Basic Metabolic ResearchFaculty of Health and Medical SciencesUniversity of CopenhagenCopenhagenDenmark
| |
Collapse
|
24
|
Zhang X, Jin T, Shi N, Yao L, Yang X, Han C, Wen L, Du D, Szatmary P, Mukherjee R, Liu T, Xia Q, Criddle DN, Huang W, Chvanov M, Sutton R. Mechanisms of Pancreatic Injury Induced by Basic Amino Acids Differ Between L-Arginine, L-Ornithine, and L-Histidine. Front Physiol 2019; 9:1922. [PMID: 30697165 PMCID: PMC6341295 DOI: 10.3389/fphys.2018.01922] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2018] [Accepted: 12/20/2018] [Indexed: 02/05/2023] Open
Abstract
Pancreatic acinar cells require high rates of amino acid uptake for digestive enzyme synthesis, but excessive concentrations can trigger acute pancreatitis (AP) by mechanisms that are not well understood. We have used three basic natural amino acids L-arginine, L-ornithine, and L-histidine to determine mechanisms of amino acid-induced pancreatic injury and whether these are common to all three amino acids. Caffeine markedly inhibited necrotic cell death pathway activation in isolated pancreatic acinar cells induced by L-arginine, but not L-ornithine, whereas caffeine accelerated L-histidine-induced cell death. Both necroptosis inhibitors of RIPK1 and RIPK3 and a necroptosis activator/apoptosis inhibitor z-VAD increased cell death caused by L-histidine, but not L-arginine or L-ornithine. Cyclophilin D knock-out (Ppif-/-) significantly attenuated cell death induced by L-histidine, but not L-arginine, or L-ornithine. Allosteric modulators of calcium-sensing receptor (CaSR) and G-protein coupled receptor class C group 6 member A (GPRC6A) had inhibitory effects on cell death induced by L-arginine but not L-ornithine or L-histidine. We developed a novel amino acid-induced AP murine model with high doses of L-histidine and confirmed AP severity was significantly reduced in Ppif-/- vs. wild type mice. In L-arginine-induced AP neither Ppif-/-, caffeine, or allosteric modulators of CaSR or GPRC6A reduced pancreatic damage, even though CaSR inhibition with NPS-2143 significantly reduced pancreatic and systemic injury in caerulein-induced AP. These findings demonstrate marked differences in the mechanisms of pancreatic injury induced by different basic amino acids and suggest the lack of effect of treatments on L-arginine-induced AP may be due to conversion to L-ornithine in the urea cycle.
Collapse
Affiliation(s)
- Xiaoying Zhang
- Department of Integrated Traditional Chinese and Western Medicine, Sichuan Provincial Pancreatitis Centre and West China-Liverpool Biomedical Research Centre, West China Hospital, Sichuan University, Chengdu, China
- Liverpool Pancreatitis Study Group, Royal Liverpool University Hospital, Institute of Translational Medicine, University of Liverpool, Liverpool, United Kingdom
| | - Tao Jin
- Department of Integrated Traditional Chinese and Western Medicine, Sichuan Provincial Pancreatitis Centre and West China-Liverpool Biomedical Research Centre, West China Hospital, Sichuan University, Chengdu, China
- Liverpool Pancreatitis Study Group, Royal Liverpool University Hospital, Institute of Translational Medicine, University of Liverpool, Liverpool, United Kingdom
| | - Na Shi
- Department of Integrated Traditional Chinese and Western Medicine, Sichuan Provincial Pancreatitis Centre and West China-Liverpool Biomedical Research Centre, West China Hospital, Sichuan University, Chengdu, China
| | - Linbo Yao
- Department of Integrated Traditional Chinese and Western Medicine, Sichuan Provincial Pancreatitis Centre and West China-Liverpool Biomedical Research Centre, West China Hospital, Sichuan University, Chengdu, China
| | - Xinmin Yang
- Department of Integrated Traditional Chinese and Western Medicine, Sichuan Provincial Pancreatitis Centre and West China-Liverpool Biomedical Research Centre, West China Hospital, Sichuan University, Chengdu, China
| | - Chenxia Han
- Department of Integrated Traditional Chinese and Western Medicine, Sichuan Provincial Pancreatitis Centre and West China-Liverpool Biomedical Research Centre, West China Hospital, Sichuan University, Chengdu, China
| | - Li Wen
- Liverpool Pancreatitis Study Group, Royal Liverpool University Hospital, Institute of Translational Medicine, University of Liverpool, Liverpool, United Kingdom
| | - Dan Du
- West China-Washington Mitochondria and Metabolism Centre, West China Hospital of Sichuan University, Chengdu, China
| | - Peter Szatmary
- Liverpool Pancreatitis Study Group, Royal Liverpool University Hospital, Institute of Translational Medicine, University of Liverpool, Liverpool, United Kingdom
| | - Rajarshi Mukherjee
- Liverpool Pancreatitis Study Group, Royal Liverpool University Hospital, Institute of Translational Medicine, University of Liverpool, Liverpool, United Kingdom
| | - Tingting Liu
- Department of Integrated Traditional Chinese and Western Medicine, Sichuan Provincial Pancreatitis Centre and West China-Liverpool Biomedical Research Centre, West China Hospital, Sichuan University, Chengdu, China
| | - Qing Xia
- Department of Integrated Traditional Chinese and Western Medicine, Sichuan Provincial Pancreatitis Centre and West China-Liverpool Biomedical Research Centre, West China Hospital, Sichuan University, Chengdu, China
| | - David N. Criddle
- Department of Cellular and Molecular Physiology, University of Liverpool, Liverpool, United Kingdom
| | - Wei Huang
- Department of Integrated Traditional Chinese and Western Medicine, Sichuan Provincial Pancreatitis Centre and West China-Liverpool Biomedical Research Centre, West China Hospital, Sichuan University, Chengdu, China
| | - Michael Chvanov
- Department of Cellular and Molecular Physiology, University of Liverpool, Liverpool, United Kingdom
| | - Robert Sutton
- Liverpool Pancreatitis Study Group, Royal Liverpool University Hospital, Institute of Translational Medicine, University of Liverpool, Liverpool, United Kingdom
| |
Collapse
|
25
|
Ding KH, Cain M, Davis M, Bergson C, McGee-Lawrence M, Perkins C, Hardigan T, Shi X, Zhong Q, Xu J, Bollag WB, Hill W, Elsalanty M, Hunter M, Isales MC, Lopez P, Hamrick M, Isales CM. Amino acids as signaling molecules modulating bone turnover. Bone 2018; 115:15-24. [PMID: 29499416 PMCID: PMC6110952 DOI: 10.1016/j.bone.2018.02.028] [Citation(s) in RCA: 31] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/22/2017] [Revised: 02/25/2018] [Accepted: 02/26/2018] [Indexed: 10/17/2022]
Abstract
Except for the essential amino acids (AAs), much of the focus on adequate dietary protein intake has been on total nitrogen and caloric intake rather than AA composition. Recent data, however, demonstrate that "amino-acid sensing" can occur through either intracellular or extracellular nutrient-sensing mechanisms. In particular, members of the class 3 G-protein coupled receptor family, like the calcium-sensing receptor are known to preferentially bind specific AAs, which then modulate receptor activation by calcium ions and thus potentially impact bone turnover. In pursuing the possibility of direct nutrient effects on bone cells, we examined individual AA effects on osteoprogenitor/bone marrow stromal cells (BMSCs), a key target for bone anabolism. We demonstrate that BMSCs express both intracellular and extracellular nutrient sensing pathways and that AAs are required for BMSC survival. In addition, certain AA types, like members of the aromatic AAs, can potently stimulate increases in intracellular calcium and ERK phosphorylation/activation. Further, based on the in vitro data, we examined the effect of specific AAs on bone mass. To better evaluate the impact of specific AAs, we added these to a low-protein diet. Our data demonstrate that a low-protein diet itself is associated with a significant drop in bone mineral density (BMD) in the older mice, related, at least in part, to an increase in osteoclastic activity. This drop in BMD in mice on the low-protein diet is prevented by addition of AAs from the aromatic group. Taken together our data show that AAs function as specific and selective signaling molecules in bone cells.
Collapse
Affiliation(s)
- Ke-Hong Ding
- Institute for Regenerative and Reparative Medicine, Medical College of Georgia, Augusta University, USA; Department of Neuroscience and Regenerative Medicine, Medical College of Georgia, Augusta University, USA
| | - Michael Cain
- Department of Orthopaedic Surgery, Medical College of Georgia, Augusta University, USA
| | - Michael Davis
- Institute for Regenerative and Reparative Medicine, Medical College of Georgia, Augusta University, USA
| | - Clare Bergson
- Department of Pharmacology and Toxicology, Medical College of Georgia, Augusta University, USA
| | - Meghan McGee-Lawrence
- Institute for Regenerative and Reparative Medicine, Medical College of Georgia, Augusta University, USA; Department of Orthopaedic Surgery, Medical College of Georgia, Augusta University, USA; Department of Cellular Biology and Anatomy, Medical College of Georgia, Augusta University, USA
| | - Crystal Perkins
- Institute for Regenerative and Reparative Medicine, Medical College of Georgia, Augusta University, USA
| | - Trevor Hardigan
- Institute for Regenerative and Reparative Medicine, Medical College of Georgia, Augusta University, USA
| | - Xingming Shi
- Institute for Regenerative and Reparative Medicine, Medical College of Georgia, Augusta University, USA; Department of Neuroscience and Regenerative Medicine, Medical College of Georgia, Augusta University, USA
| | - Qing Zhong
- Institute for Regenerative and Reparative Medicine, Medical College of Georgia, Augusta University, USA; Department of Neuroscience and Regenerative Medicine, Medical College of Georgia, Augusta University, USA
| | - Jianrui Xu
- Institute for Regenerative and Reparative Medicine, Medical College of Georgia, Augusta University, USA; Department of Neuroscience and Regenerative Medicine, Medical College of Georgia, Augusta University, USA
| | - Wendy B Bollag
- Institute for Regenerative and Reparative Medicine, Medical College of Georgia, Augusta University, USA; Department of Medicine, Medical College of Georgia, Augusta University, USA; Department of Physiology, Medical College of Georgia, Augusta University, USA; Department of Cellular Biology and Anatomy, Medical College of Georgia, Augusta University, USA; Charlie Norwood VA Medical Center, School of Dental Medicine, Augusta, GA 30912, USA
| | - William Hill
- Institute for Regenerative and Reparative Medicine, Medical College of Georgia, Augusta University, USA; Department of Orthopaedic Surgery, Medical College of Georgia, Augusta University, USA; Department of Cellular Biology and Anatomy, Medical College of Georgia, Augusta University, USA; Charlie Norwood VA Medical Center, School of Dental Medicine, Augusta, GA 30912, USA
| | - Mohammed Elsalanty
- Department of Oral Biology, School of Dental Medicine, Augusta, GA 30912, USA
| | - Monte Hunter
- Institute for Regenerative and Reparative Medicine, Medical College of Georgia, Augusta University, USA; Department of Orthopaedic Surgery, Medical College of Georgia, Augusta University, USA
| | - Maria C Isales
- Institute for Regenerative and Reparative Medicine, Medical College of Georgia, Augusta University, USA
| | - Patricia Lopez
- Institute for Regenerative and Reparative Medicine, Medical College of Georgia, Augusta University, USA
| | - Mark Hamrick
- Institute for Regenerative and Reparative Medicine, Medical College of Georgia, Augusta University, USA; Department of Orthopaedic Surgery, Medical College of Georgia, Augusta University, USA; Department of Cellular Biology and Anatomy, Medical College of Georgia, Augusta University, USA
| | - Carlos M Isales
- Institute for Regenerative and Reparative Medicine, Medical College of Georgia, Augusta University, USA; Department of Neuroscience and Regenerative Medicine, Medical College of Georgia, Augusta University, USA; Department of Orthopaedic Surgery, Medical College of Georgia, Augusta University, USA; Department of Medicine, Medical College of Georgia, Augusta University, USA; Department of Cellular Biology and Anatomy, Medical College of Georgia, Augusta University, USA.
| |
Collapse
|
26
|
Amino Y, Wakabayashi H, Akashi S, Ishiwatari Y. Structural analysis and taste evaluation of γ-glutamyl peptides comprising sulfur-containing amino acids. Biosci Biotechnol Biochem 2018; 82:383-394. [DOI: 10.1080/09168451.2018.1436433] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/02/2023]
Abstract
Abstract
The structures, flavor-modifying effects, and CaSR activities of γ-glutamyl peptides comprising sulfur-containing amino acids were investigated. The chemical structures, including the linkage mode of the N-terminal glutamic acid, of γ-L-glutamyl-S-(2-propenyl)-L-cysteine (γ-L-glutamyl-S-allyl-L-cysteine) and its sulfoxide isolated from garlic were established by comparing their NMR spectra with those of authentic peptides prepared using chemical methods. Mass spectrometric analysis also enabled determination of the linkage modes in the glutamyl dipeptides by their characteristic fragmentation. In sensory evaluation, these peptides exhibited flavor-modifying effects (continuity) in umami solutions less pronounced but similar to that of glutathione. Furthermore, the peptides exhibited intrinsic flavor due to the sulfur-containing structure, which may be partially responsible for their flavor-modifying effects. In CaSR assays, γ-L-glutamyl-S-methyl-L-cysteinylglycine was most active, which indicates that the presence of a medium-sized aliphatic substituent at the second amino acid residue in γ-glutamyl peptides enhances CaSR activity.
Collapse
Affiliation(s)
- Yusuke Amino
- Institute for Innovation, Ajinomoto Co., Inc., Kawasaki, Japan
| | - Hidehiko Wakabayashi
- Institute of Food Sciences and Technologies, Ajinomoto Co., Inc., Kawasaki, Japan
| | - Satoko Akashi
- Institute for Innovation, Ajinomoto Co., Inc., Kawasaki, Japan
| | | |
Collapse
|
27
|
Gwynne RM, Ly KDKN, Parry LJ, Bornstein JC. Calcium Sensing Receptors Mediate Local Inhibitory Reflexes Evoked by L-Phenylalanine in Guinea Pig Jejunum. Front Physiol 2017; 8:991. [PMID: 29255423 PMCID: PMC5722837 DOI: 10.3389/fphys.2017.00991] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2017] [Accepted: 11/17/2017] [Indexed: 12/18/2022] Open
Abstract
Amino acids applied to the mucosa evoke inhibitory reflexes in guinea-pig jejunum, but the receptors involved in sensory transduction are still unclear. One promising candidate is the extracellular calcium sensing receptor (CaSR), which is expressed by mucosal enteroendocrine cells and is preferentially activated by aromatic L-amino acids. We tested this by applying various amino acids to the mucosa and recording the resulting inhibitory junction potentials (IJPs) in nearby circular smooth muscle via intracellular recording. The CaSR is stereospecific and L-Phenylalanine evoked a significantly larger response than D-Phenylalanine when both were applied to the same site. The same pattern was seen with L- and D-Tryptophan, another aromatic amino acid. The CaSR is preferentially activated by aromatic amino acids and responses to L-Leucine and L-Lysine were significantly lower than those to L-Phenylalanine applied to the same site. Responses to L-Phenylalanine were dose-dependently suppressed by blockade of the CaSR with NPS2143, a CaSR antagonist, and mimicked by mucosal application of cinacalcet, a CaSR agonist. Responses to cinacalcet had similar pharmacology to that of responses to L-Phenylalanine, in that each requires both P2 purinoreceptors and 5-HT receptors. L-Glutamate evoked IJPs similar to those produced by L-Phenylalanine and these were depressed by blockade of P2 receptors and 5-HT3 plus 5-HT4 receptors, but NPS2143 was ineffective. The AMPA receptor antagonists DNQX (10 μM) and CNQX (10 μM) reduced IJPs evoked by L-Glutamate by 88 and 79% respectively, but neither BAY367260 (mGluR5 antagonist) nor 2APV (NMDA antagonist) affected IJPs evoked by L-Glutamate. We conclude that local inhibitory reflexes evoked by aromatic L-amino acids in guinea pig jejunum involve activation of CaSRs which triggers release of ATP and 5-HT from the mucosa. L-Glutamate also evokes inhibitory reflexes, via a pathway that does not involve CaSRs. It is likely there are multiple receptors acting as sensory transducers for different luminal amino acids.
Collapse
Affiliation(s)
- Rachel M Gwynne
- Department of Physiology, University of Melbourne, Parkville, VIC, Australia
| | - Kenny D K N Ly
- Department of Physiology, University of Melbourne, Parkville, VIC, Australia
| | - Laura J Parry
- School of BioSciences, University of Melbourne, Parkville, VIC, Australia
| | - Joel C Bornstein
- Department of Physiology, University of Melbourne, Parkville, VIC, Australia
| |
Collapse
|
28
|
Julliard AK, Al Koborssy D, Fadool DA, Palouzier-Paulignan B. Nutrient Sensing: Another Chemosensitivity of the Olfactory System. Front Physiol 2017; 8:468. [PMID: 28747887 PMCID: PMC5506222 DOI: 10.3389/fphys.2017.00468] [Citation(s) in RCA: 49] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2017] [Accepted: 06/19/2017] [Indexed: 12/31/2022] Open
Abstract
Olfaction is a major sensory modality involved in real time perception of the chemical composition of the external environment. Olfaction favors anticipation and rapid adaptation of behavioral responses necessary for animal survival. Furthermore, recent studies have demonstrated that there is a direct action of metabolic peptides on the olfactory network. Orexigenic peptides such as ghrelin and orexin increase olfactory sensitivity, which in turn, is decreased by anorexigenic hormones such as insulin and leptin. In addition to peptides, nutrients can play a key role on neuronal activity. Very little is known about nutrient sensing in olfactory areas. Nutrients, such as carbohydrates, amino acids, and lipids, could play a key role in modulating olfactory sensitivity to adjust feeding behavior according to metabolic need. Here we summarize recent findings on nutrient-sensing neurons in olfactory areas and delineate the limits of our knowledge on this topic. The present review opens new lines of investigations on the relationship between olfaction and food intake, which could contribute to determining the etiology of metabolic disorders.
Collapse
Affiliation(s)
- A-Karyn Julliard
- Univ Lyon, Université Claude Bernard Lyon1, Centre de Recherche en Neurosciences de Lyon (CRNL), INSERM U1028/Centre National de la Recherche Scientifique UMR5292 Team Olfaction: From Coding to MemoryLyon, France
| | - Dolly Al Koborssy
- Department of Biological Science, Florida State UniversityTallahassee, FL, United States.,Program in Neuroscience, Florida State UniversityTallahassee, FL, United States
| | - Debra A Fadool
- Department of Biological Science, Florida State UniversityTallahassee, FL, United States.,Program in Neuroscience, Florida State UniversityTallahassee, FL, United States.,Institute of Molecular Biophysics, Florida State UniversityTallahassee, FL, United States
| | - Brigitte Palouzier-Paulignan
- Univ Lyon, Université Claude Bernard Lyon1, Centre de Recherche en Neurosciences de Lyon (CRNL), INSERM U1028/Centre National de la Recherche Scientifique UMR5292 Team Olfaction: From Coding to MemoryLyon, France
| |
Collapse
|
29
|
Ho YY, Nakato J, Mizushige T, Kanamoto R, Tanida M, Akiduki S, Ohinata K. l-Ornithine stimulates growth hormone release in a manner dependent on the ghrelin system. Food Funct 2017; 8:2110-2114. [PMID: 28513740 DOI: 10.1039/c7fo00309a] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
We found that intraduodenal administration of l-ornithine (l-Orn) stimulates growth hormone (GH) secretion in Wistar rats, and then investigated its mechanism. GH-releasing activity after intraduodenal administration of l-Orn was blocked by [d-Lys3]-GHRP-6, an antagonist of the ghrelin receptor; however, l-Orn (100 μM) has no affinity for the ghrelin receptor, suggesting that the GH-releasing activity of l-Orn is mediated via ghrelin release and activation of the ghrelin receptor. Intraduodenally administered l-Orn increased ghrelin mRNA expression in the duodenum but not in the stomach or hypothalamus. In addition, l-Orn-induced GH-releasing activity was inhibited by propranolol, an antagonist of β-adrenergic receptor, which is known to be coupled to ghrelin release. In conclusion, intraduodenally administered l-Orn stimulates GH secretion through the sympathetic nervous and ghrelin systems.
Collapse
Affiliation(s)
- Yee Yin Ho
- Division of Food Science and Biotechnology, Graduate School of Agriculture, Kyoto University, Gokasho Uji, Kyoto 611-0011, Japan.
| | - Junya Nakato
- Division of Food Science and Biotechnology, Graduate School of Agriculture, Kyoto University, Gokasho Uji, Kyoto 611-0011, Japan.
| | - Takafumi Mizushige
- Division of Food Science and Biotechnology, Graduate School of Agriculture, Kyoto University, Gokasho Uji, Kyoto 611-0011, Japan. and Research Unit for Physiological Chemistry, C-PIER, Kyoto University, Kyoto 606-8501, Japan
| | - Ryuhei Kanamoto
- Division of Food Science and Biotechnology, Graduate School of Agriculture, Kyoto University, Gokasho Uji, Kyoto 611-0011, Japan.
| | - Mamoru Tanida
- Department of Biomedical Sciences, College of Life Sciences, Ritsumeikan University, 1-1-1 Noji-kita, Kusatsu, Shiga 525-8577, Japan and Department of Physiology II Kanazawa Medical University, Uchinada, Ishikawa 920-0293, Japan
| | - Saori Akiduki
- Healthcare Products Development Center, Kyowa Hakko Bio Co., Ltd, Tsukuba, Ibaraki 305-0841, Japan
| | - Kousaku Ohinata
- Division of Food Science and Biotechnology, Graduate School of Agriculture, Kyoto University, Gokasho Uji, Kyoto 611-0011, Japan.
| |
Collapse
|
30
|
Abstract
The rising incidence of metabolic diseases worldwide has prompted renewed interest in the study of intermediary metabolism and cellular bioenergetics. The application of modern biochemical methods for quantitating fuel substrate metabolism with advanced mouse genetic approaches has greatly increased understanding of the mechanisms that integrate energy metabolism in the whole organism. Examination of the intermediary metabolism of skeletal cells has been sparked by a series of unanticipated observations in genetically modified mice that suggest the existence of novel endocrine pathways through which bone cells communicate their energy status to other centers of metabolic control. The recognition of this expanded role of the skeleton has in turn led to new lines of inquiry directed at defining the fuel requirements and bioenergetic properties of bone cells. This article provides a comprehensive review of historical and contemporary studies on the metabolic properties of bone cells and the mechanisms that control energy substrate utilization and bioenergetics. Special attention is devoted to identifying gaps in our current understanding of this new area of skeletal biology that will require additional research to better define the physiological significance of skeletal cell bioenergetics in human health and disease.
Collapse
Affiliation(s)
- Ryan C Riddle
- Department of Orthopaedic Surgery, The Johns Hopkins University, Baltimore, Maryland; and The Baltimore Veterans Administration Medical Center, Baltimore, Maryland
| | - Thomas L Clemens
- Department of Orthopaedic Surgery, The Johns Hopkins University, Baltimore, Maryland; and The Baltimore Veterans Administration Medical Center, Baltimore, Maryland
| |
Collapse
|
31
|
Conigrave AD. The Calcium-Sensing Receptor and the Parathyroid: Past, Present, Future. Front Physiol 2016; 7:563. [PMID: 28018229 PMCID: PMC5156698 DOI: 10.3389/fphys.2016.00563] [Citation(s) in RCA: 63] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2016] [Accepted: 11/07/2016] [Indexed: 12/13/2022] Open
Abstract
Parathyroid hormone (PTH) defends the extracellular fluid from hypocalcemia and has powerful and well-documented actions on the skeleton and renal tubular system. To achieve a satisfactory stable plasma calcium level, the secretion of PTH, and the resulting serum PTH level, is titrated carefully to the prevailing plasma ionized Ca2+ concentration via a Ca2+ sensing mechanism that mediates feedback inhibition of PTH secretion. Herein, I consider the properties of the parathyroid Ca2+ sensing mechanism, the identity of the Ca2+ sensor, the intracellular biochemical mechanisms that it controls, the manner of its integration with other components of the PTH secretion control mechanism, and its modulation by other nutrients. Together the well-established, recently elucidated, and yet-to-be discovered elements of the story constitute the past, present, and future of the parathyroid and its calcium-sensing receptor (CaSR).
Collapse
Affiliation(s)
- Arthur D Conigrave
- Faculties of Science and Medicine, School of Life and Environmental Sciences, Charles Perkins Centre, University of Sydney Sydney, NSW, Australia
| |
Collapse
|
32
|
Morris SM. Arginine Metabolism Revisited. J Nutr 2016; 146:2579S-2586S. [PMID: 27934648 DOI: 10.3945/jn.115.226621] [Citation(s) in RCA: 251] [Impact Index Per Article: 27.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2015] [Revised: 01/22/2016] [Accepted: 02/05/2016] [Indexed: 01/20/2023] Open
Abstract
Mammalian arginine metabolism is complex due to the expression of multiple enzymes that utilize arginine as substrate and to interactions or competition between specific enzymes involved in arginine metabolism. Moreover, cells may contain multiple intracellular arginine pools that are not equally accessible to all arginine metabolic enzymes, thus presenting additional challenges to more fully understanding arginine metabolism. At the whole-body level, arginine metabolism ultimately results in the production of a biochemically diverse range of products, including nitric oxide, urea, creatine, polyamines, proline, glutamate, agmatine, and homoarginine. Included in this group of compounds are the methylated arginines (e.g., asymmetric dimethylarginine), which are released upon degradation of proteins containing methylated arginine residues. Changes in arginine concentration also can regulate cellular metabolism and function via a variety of arginine sensors. Although much is known about arginine metabolism, elucidation of the physiologic or pathophysiologic roles for all of the pathways and their metabolites remains an active area of investigation, as exemplified by current findings highlighted in this review.
Collapse
Affiliation(s)
- Sidney M Morris
- Department of Microbiology and Molecular Genetics, University of Pittsburgh School of Medicine, Pittsburgh, PA
| |
Collapse
|
33
|
Protein/amino-acid modulation of bone cell function. BONEKEY REPORTS 2016; 5:827. [PMID: 28149508 PMCID: PMC5238414 DOI: 10.1038/bonekey.2016.58] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/16/2015] [Accepted: 06/24/2016] [Indexed: 01/07/2023]
Abstract
Nutrients (protein, carbohydrates and fats) have traditionally been thought of as fuels simply providing the energy for cellular metabolic activity. According to the classic view, if nutrients are available, then anabolic pathways are activated, and if nutrients are not available, catabolic pathways are activated. However, it is becoming increasingly clear that nutrient effects on bone cells (stem cells, osteoblasts and osteoclasts) are complex, some nutrients promote bone formation, whereas others interfere with bone formation or actually promote bone break down. At an organ level, nutrient intake can suppress bone breakdown and modulate the activity of the calcium/vitamin D/parathyroid hormone axis. At a cellular level, nutrient intake can impact cellular energetics either through a direct mechanism (binding or uptake of the nutrient into the cell) or indirect (by elevating nutrient-related hormones such as insulin, insulin-like growth factor 1 or incretin hormones). It is also becoming clear that within a nutrient class (for example, protein), individual components (that is, amino acids) can have markedly different effects on cell function and impact bone formation. The focus of this review will be on one nutrient class in particular, dietary protein. As the prevalence of inadequate dietary protein intake increases with age, these findings may have translational implications as to the optimal dietary protein content in the setting of age-associated bone loss.
Collapse
|
34
|
Geng Y, Mosyak L, Kurinov I, Zuo H, Sturchler E, Cheng TC, Subramanyam P, Brown AP, Brennan SC, Mun HC, Bush M, Chen Y, Nguyen TX, Cao B, Chang DD, Quick M, Conigrave AD, Colecraft HM, McDonald P, Fan QR. Structural mechanism of ligand activation in human calcium-sensing receptor. eLife 2016; 5. [PMID: 27434672 PMCID: PMC4977154 DOI: 10.7554/elife.13662] [Citation(s) in RCA: 170] [Impact Index Per Article: 18.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2015] [Accepted: 07/18/2016] [Indexed: 12/21/2022] Open
Abstract
Human calcium-sensing receptor (CaSR) is a G-protein-coupled receptor (GPCR) that maintains extracellular Ca2+ homeostasis through the regulation of parathyroid hormone secretion. It functions as a disulfide-tethered homodimer composed of three main domains, the Venus Flytrap module, cysteine-rich domain, and seven-helix transmembrane region. Here, we present the crystal structures of the entire extracellular domain of CaSR in the resting and active conformations. We provide direct evidence that L-amino acids are agonists of the receptor. In the active structure, L-Trp occupies the orthosteric agonist-binding site at the interdomain cleft and is primarily responsible for inducing extracellular domain closure to initiate receptor activation. Our structures reveal multiple binding sites for Ca2+ and PO43- ions. Both ions are crucial for structural integrity of the receptor. While Ca2+ ions stabilize the active state, PO43- ions reinforce the inactive conformation. The activation mechanism of CaSR involves the formation of a novel dimer interface between subunits. DOI:http://dx.doi.org/10.7554/eLife.13662.001 Calcium ions regulate many processes in the human body. The calcium-sensing receptor, called CaSR, is responsible for maintaining a stable level of calcium ions in the blood. This receptor can detect small changes in the concentration of calcium ions, and activates signalling events within the cell to restore the level of calcium ions back to normal. Abnormal activity of this receptor is associated with severe diseases in humans CaSR is found in the surface membrane of cells and belongs to a family of proteins called G-protein coupled receptors. Much of the protein extends out of the cell and interacts with calcium ions, phosphate ions and certain other molecules such as amino acids. However, it was not well understood how these small molecules bind to CaSR and how this activates the receptor. Geng et al. have now used a technique called X-ray crystallography to view the three-dimensional structure of the exterior domain of CaSR in its resting state and active state. These structures revealed that, contrary to expectations, calcium ions are not the main activator of the receptor. Instead, Geng et al. found that CaSR adopts an inactive state in the absence or presence of calcium ions, while the active state only forms when an amino acid is bound. Furthermore investigation showed that calcium ions are needed to stabilise the active form, while phosphate ions keep the inactive form stable. Geng et al. also identified the shape changes that must occur as CaSR transitions from its inactive to its active state. In particular, an amino acid binding to the exterior domain causes it to close like a venus flytrap, which is a crucial step in activating the receptor. Taken together, the findings show that the amino acids and calcium ions act jointly to fully activate CaSR. The next steps are to determine the structure of the entire receptor with and without its small molecule partners and to use these structures to design drugs that can alter CaSR’s activity in order to treat human diseases. DOI:http://dx.doi.org/10.7554/eLife.13662.002
Collapse
Affiliation(s)
- Yong Geng
- Department of Pharmacology, Columbia University, New York, United States.,Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, China
| | - Lidia Mosyak
- Department of Pharmacology, Columbia University, New York, United States
| | - Igor Kurinov
- Department of Chemistry and Chemical Biology, Cornell University, Ithaca, United States
| | - Hao Zuo
- Department of Pharmacology, Columbia University, New York, United States
| | - Emmanuel Sturchler
- Department of Molecular Therapeutics, The Scripps Translational Science Institute, Jupiter, United States
| | - Tat Cheung Cheng
- Department of Pharmacology, Columbia University, New York, United States
| | - Prakash Subramanyam
- Department of Physiology and Cellular Biophysics, Columbia University, New York, United States
| | - Alice P Brown
- School of Life and Environmental Sciences, University of Sydney, New South Wales, Australia
| | - Sarah C Brennan
- School of Life and Environmental Sciences, University of Sydney, New South Wales, Australia
| | - Hee-Chang Mun
- School of Life and Environmental Sciences, University of Sydney, New South Wales, Australia
| | - Martin Bush
- Department of Pharmacology, Columbia University, New York, United States
| | - Yan Chen
- Department of Pharmacology, Columbia University, New York, United States
| | - Trang X Nguyen
- Department of Psychiatry, Columbia University, New York, United States
| | - Baohua Cao
- Department of Pharmacology, Columbia University, New York, United States
| | - Donald D Chang
- Department of Physiology and Cellular Biophysics, Columbia University, New York, United States
| | - Matthias Quick
- Department of Psychiatry, Columbia University, New York, United States
| | - Arthur D Conigrave
- School of Life and Environmental Sciences, University of Sydney, New South Wales, Australia
| | - Henry M Colecraft
- Department of Physiology and Cellular Biophysics, Columbia University, New York, United States
| | - Patricia McDonald
- Department of Molecular Therapeutics, The Scripps Translational Science Institute, Jupiter, United States
| | - Qing R Fan
- Department of Pharmacology, Columbia University, New York, United States.,Department of Pathology and Cell Biology, Columbia University, New York, United States
| |
Collapse
|
35
|
Abstract
The present review examines the pig as a model for physiological studies in human subjects related to nutrient sensing, appetite regulation, gut barrier function, intestinal microbiota and nutritional neuroscience. The nutrient-sensing mechanisms regarding acids (sour), carbohydrates (sweet), glutamic acid (umami) and fatty acids are conserved between humans and pigs. In contrast, pigs show limited perception of high-intensity sweeteners and NaCl and sense a wider array of amino acids than humans. Differences on bitter taste may reflect the adaptation to ecosystems. In relation to appetite regulation, plasma concentrations of cholecystokinin and glucagon-like peptide-1 are similar in pigs and humans, while peptide YY in pigs is ten to twenty times higher and ghrelin two to five times lower than in humans. Pigs are an excellent model for human studies for vagal nerve function related to the hormonal regulation of food intake. Similarly, the study of gut barrier functions reveals conserved defence mechanisms between the two species particularly in functional permeability. However, human data are scant for some of the defence systems and nutritional programming. The pig model has been valuable for studying the changes in human microbiota following nutritional interventions. In particular, the use of human flora-associated pigs is a useful model for infants, but the long-term stability of the implanted human microbiota in pigs remains to be investigated. The similarity of the pig and human brain anatomy and development is paradigmatic. Brain explorations and therapies described in pig, when compared with available human data, highlight their value in nutritional neuroscience, particularly regarding functional neuroimaging techniques.
Collapse
|
36
|
Zoch ML, Clemens TL, Riddle RC. New insights into the biology of osteocalcin. Bone 2016; 82:42-9. [PMID: 26055108 PMCID: PMC4670816 DOI: 10.1016/j.bone.2015.05.046] [Citation(s) in RCA: 403] [Impact Index Per Article: 44.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/22/2015] [Revised: 05/01/2015] [Accepted: 05/13/2015] [Indexed: 12/19/2022]
Abstract
Osteocalcin is among the most abundant proteins in bone and is produced exclusively by osteoblasts. Initially believed to be an inhibitor of bone mineralization, recent studies suggest a broader role for osteocalcin that extends to the regulation of whole body metabolism, reproduction, and cognition. Circulating undercarboxylated osteocalcin, which is regulated by insulin, acts in a feed-forward loop to increase β-cell proliferation as well as insulin production and secretion, while skeletal muscle and adipose tissue respond to osteocalcin by increasing their sensitivity to insulin. Osteocalcin also acts in the brain to increase neurotransmitter production and in the testes to stimulate testosterone production. At least one putative receptor for osteocalcin, Gprc6a, is expressed by adipose, skeletal muscle, and the Leydig cells of the testes and appears to mediate osteocalcin's effects in these tissues. In this review, we summarize these new discoveries, which suggest that the ability of osteocalcin to function both locally in bone and as a hormone depends on a novel post-translational mechanism that alters osteocalcin's affinity for the bone matrix and bioavailability. This article is part of a Special Issue entitled Bone and diabetes.
Collapse
Affiliation(s)
- Meredith L Zoch
- Department of Orthopaedic Surgery, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Thomas L Clemens
- Department of Orthopaedic Surgery, Johns Hopkins University School of Medicine, Baltimore, MD, USA; Baltimore Veterans Administration Medical Center, Baltimore, MD, USA
| | - Ryan C Riddle
- Department of Orthopaedic Surgery, Johns Hopkins University School of Medicine, Baltimore, MD, USA; Baltimore Veterans Administration Medical Center, Baltimore, MD, USA.
| |
Collapse
|
37
|
Amino Y, Nakazawa M, Kaneko M, Miyaki T, Miyamura N, Maruyama Y, Eto Y. Structure–CaSR–Activity Relation of Kokumi γ-Glutamyl Peptides. Chem Pharm Bull (Tokyo) 2016; 64:1181-9. [DOI: 10.1248/cpb.c16-00293] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Affiliation(s)
| | - Masakazu Nakazawa
- Research Institute for Bioscience Products & Fine Chemicals, Ajinomoto Co., Inc
| | | | - Takashi Miyaki
- Institute of Food Research and Technologies, Ajinomoto Co., Inc
| | | | | | - Yuzuru Eto
- Institute for Innovation, Ajinomoto Co., Inc
| |
Collapse
|
38
|
El Refaey M, Watkins CP, Kennedy EJ, Chang A, Zhong Q, Ding KH, Shi XM, Xu J, Bollag WB, Hill WD, Johnson M, Hunter M, Hamrick MW, Isales CM. Oxidation of the aromatic amino acids tryptophan and tyrosine disrupts their anabolic effects on bone marrow mesenchymal stem cells. Mol Cell Endocrinol 2015; 410:87-96. [PMID: 25637715 PMCID: PMC4444384 DOI: 10.1016/j.mce.2015.01.034] [Citation(s) in RCA: 61] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/25/2014] [Revised: 01/23/2015] [Accepted: 01/23/2015] [Indexed: 02/06/2023]
Abstract
Age-induced bone loss is associated with greater bone resorption and decreased bone formation resulting in osteoporosis and osteoporosis-related fractures. The etiology of this age-induced bone loss is not clear but has been associated with increased generation of reactive oxygen species (ROS) from leaky mitochondria. ROS are known to oxidize/damage the surrounding proteins/amino acids/enzymes and thus impair their normal function. Among the amino acids, the aromatic amino acids are particularly prone to modification by oxidation. Since impaired osteoblastic differentiation from bone marrow mesenchymal stem cells (BMMSCs) plays a role in age-related bone loss, we wished to examine whether oxidized amino acids (in particular the aromatic amino acids) modulated BMMSC function. Using mouse BMMSCs, we examined the effects of the oxidized amino acids di-tyrosine and kynurenine on proliferation, differentiation and Mitogen-Activated Protein Kinase (MAPK) pathway. Our data demonstrate that amino acid oxides (in particular kynurenine) inhibited BMMSC proliferation, alkaline phosphatase expression and activity and the expression of osteogenic markers (Osteocalcin and Runx2). Taken together, our data are consistent with a potential pathogenic role for oxidized amino acids in age-induced bone loss.
Collapse
Affiliation(s)
- Mona El Refaey
- Institute for Regenerative and Reparative Medicine, Georgia Regents University, Augusta, GA, United States; Department of Neuroscience and Regenerative Medicine, Georgia Regents University, Augusta, GA, United States
| | - Christopher P Watkins
- Department of Pharmaceutical and Biomedical Sciences, College of Pharmacy University of Georgia, Athens, GA 30602, United States
| | - Eileen J Kennedy
- Department of Pharmaceutical and Biomedical Sciences, College of Pharmacy University of Georgia, Athens, GA 30602, United States
| | - Andrew Chang
- Institute for Regenerative and Reparative Medicine, Georgia Regents University, Augusta, GA, United States
| | - Qing Zhong
- Institute for Regenerative and Reparative Medicine, Georgia Regents University, Augusta, GA, United States; Department of Neuroscience and Regenerative Medicine, Georgia Regents University, Augusta, GA, United States
| | - Ke-Hong Ding
- Institute for Regenerative and Reparative Medicine, Georgia Regents University, Augusta, GA, United States; Department of Neuroscience and Regenerative Medicine, Georgia Regents University, Augusta, GA, United States
| | - Xing-ming Shi
- Institute for Regenerative and Reparative Medicine, Georgia Regents University, Augusta, GA, United States; Department of Neuroscience and Regenerative Medicine, Georgia Regents University, Augusta, GA, United States; Department of Orthopaedic Surgery, Georgia Regents University, Augusta, GA, United States
| | - Jianrui Xu
- Institute for Regenerative and Reparative Medicine, Georgia Regents University, Augusta, GA, United States; Department of Medicine, Georgia Regents University, Augusta, GA, United States
| | - Wendy B Bollag
- Institute for Regenerative and Reparative Medicine, Georgia Regents University, Augusta, GA, United States; Department of Physiology, Georgia Regents University, Augusta, GA, United States; Charlie Norwood VA Medical Center, Augusta, GA 30912, United States
| | - William D Hill
- Institute for Regenerative and Reparative Medicine, Georgia Regents University, Augusta, GA, United States; Department of Orthopaedic Surgery, Georgia Regents University, Augusta, GA, United States; Charlie Norwood VA Medical Center, Augusta, GA 30912, United States; Department of Cellular Biology and Anatomy, Georgia Regents University, Augusta, GA, United States
| | - Maribeth Johnson
- Department of Biostatistics, Georgia Regents University, Augusta, GA, United States
| | - Monte Hunter
- Department of Orthopaedic Surgery, Georgia Regents University, Augusta, GA, United States
| | - Mark W Hamrick
- Institute for Regenerative and Reparative Medicine, Georgia Regents University, Augusta, GA, United States; Department of Orthopaedic Surgery, Georgia Regents University, Augusta, GA, United States; Department of Cellular Biology and Anatomy, Georgia Regents University, Augusta, GA, United States
| | - Carlos M Isales
- Institute for Regenerative and Reparative Medicine, Georgia Regents University, Augusta, GA, United States; Department of Neuroscience and Regenerative Medicine, Georgia Regents University, Augusta, GA, United States; Department of Orthopaedic Surgery, Georgia Regents University, Augusta, GA, United States; Department of Medicine, Georgia Regents University, Augusta, GA, United States; Department of Cellular Biology and Anatomy, Georgia Regents University, Augusta, GA, United States.
| |
Collapse
|
39
|
Kendig DM, Hurst NR, Bradley ZL, Mahavadi S, Kuemmerle JF, Lyall V, DeSimone J, Murthy KS, Grider JR. Activation of the umami taste receptor (T1R1/T1R3) initiates the peristaltic reflex and pellet propulsion in the distal colon. Am J Physiol Gastrointest Liver Physiol 2014; 307:G1100-7. [PMID: 25324508 PMCID: PMC4254958 DOI: 10.1152/ajpgi.00251.2014] [Citation(s) in RCA: 40] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
Abstract
Intraluminal nutrients in the gut affect the peristaltic reflex, although the mechanism is not well defined. Recent evidence supports the presence of taste receptors and their signaling components in enteroendocrine cells, although their function is unclear. This study aimed to determine if nutrients modify colonic motility through activation of taste receptors. Colonic sections were immunostained for the umami taste receptor T1R1/T1R3, which mediates the response to umami ligands, such as monosodium glutamate (MSG), in taste cells. Ascending contraction, descending relaxation, and calcitonin gene-related peptide release were measured in three-chamber flat-sheet preparations of rat colon in response to MSG alone or with inosine 5'-monophosphate (IMP). Velocity of artificial fecal pellet propulsion was measured by video recording in guinea pig distal colon. T1R1/T1R3 receptors were present in enteroendocrine cells of colonic sections from human, rat, mouse, and guinea pig. MSG initiated ascending contraction and descending relaxation components of the peristaltic reflex and calcitonin gene-related peptide release in flat-sheet preparations. IMP augmented the MSG-induced effects, suggesting activation of T1R1/T1R3 receptors. In T1R1(-/-) mice, mucosal stroking, but not MSG, elicited a peristaltic reflex. Intraluminal perfusion of MSG enhanced the velocity of artificial fecal pellet propulsion, which was also augmented by IMP. Propulsion was also increased by l-cysteine, but not l-tryptophan, supporting a role of T1R1/T1R3 receptors. We conclude that T1R1/T1R3 activation by luminal MSG or l-cysteine elicits a peristaltic reflex and CGRP release and increases the velocity of pellet propulsion in distal colon. This mechanism may explain how nutrients regulate colonic propulsion.
Collapse
Affiliation(s)
- Derek M. Kendig
- 1Department of Physiology and Biophysics, Virginia Commonwealth University School of Medicine, Richmond, Virginia; and
| | - Norman R. Hurst
- 1Department of Physiology and Biophysics, Virginia Commonwealth University School of Medicine, Richmond, Virginia; and
| | - Zachary L. Bradley
- 1Department of Physiology and Biophysics, Virginia Commonwealth University School of Medicine, Richmond, Virginia; and
| | - Sunila Mahavadi
- 1Department of Physiology and Biophysics, Virginia Commonwealth University School of Medicine, Richmond, Virginia; and
| | - John F. Kuemmerle
- 1Department of Physiology and Biophysics, Virginia Commonwealth University School of Medicine, Richmond, Virginia; and ,2Department of Medicine, VCU Program in Enteric Neuromuscular Sciences, Virginia Commonwealth University School of Medicine, Richmond, Virginia
| | - Vijay Lyall
- 1Department of Physiology and Biophysics, Virginia Commonwealth University School of Medicine, Richmond, Virginia; and
| | - John DeSimone
- 1Department of Physiology and Biophysics, Virginia Commonwealth University School of Medicine, Richmond, Virginia; and
| | - Karnam S. Murthy
- 1Department of Physiology and Biophysics, Virginia Commonwealth University School of Medicine, Richmond, Virginia; and ,2Department of Medicine, VCU Program in Enteric Neuromuscular Sciences, Virginia Commonwealth University School of Medicine, Richmond, Virginia
| | - John R. Grider
- 1Department of Physiology and Biophysics, Virginia Commonwealth University School of Medicine, Richmond, Virginia; and ,2Department of Medicine, VCU Program in Enteric Neuromuscular Sciences, Virginia Commonwealth University School of Medicine, Richmond, Virginia
| |
Collapse
|
40
|
Abstract
Although the prevalence of main idiopathic forms of inflammatory bowel disease (IBD) has risen considerably over the last decades, their clinical features do not allow accurate prediction of prognosis, likelihood of disease progression, or response to specific therapy. Through a better understanding of the molecular pathways involved in IBD and the promise of more targeted therapies, the personalized approach to the management of IBD shows potential. To achieve this, there remains a significant need to better understand the disease process at cellular and molecular levels for any given individual with IBD. The complexity of biological functional networks behind the etiology of IBD highlights the need for their comprehensive analysis. In this, omics technologies can generate a systemic view of IBD pathogenesis on which to base novel, multiple pathway-integrated therapies. Omics sciences have just started to contribute here by generating gene, protein expression, metabolite data at global level and large scale, and more recently by offering new opportunities to explore gut functional ecology. In particular, there is much expectation regarding the putative role of the gut microbiome in IBD. No doubt it will provide additional insights and lead to the development of alternative, hopefully better, diagnostic, prognostic, and monitoring tools in the management of IBD. This review discusses perspectives of relevance to clinical translation with emphasis on gut microbial metabolic activities.
Collapse
|
41
|
Gu J, Law AYS, Yeung BHY, Wong CKC. Activation of gill Ca2+-sensing receptor as a protective pathway to reduce Ca2+-induced cytotoxicity. J Mol Endocrinol 2014; 53:155-64. [PMID: 25180251 DOI: 10.1530/jme-14-0060] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
The expression of the Ca(2) (+)-sensing receptor (Casr) in the endocrine gland known as the corpuscle of Stannius (CS) regulates the secretion of the hypocalcemic hormone stanniocalcin-1 (STC1) to inhibit gill Ca(2) (+) uptake. Although numerous studies have reported the branchial expression of Casr and Stc1, the functions of these proteins in gills have not been elucidated yet. On the basis of recent findings regarding the autocrine/paracrine functions of STC1 in mammalian models, we proposed the hypothesis that branchial CaSR has an in situ 'sensing' function to regulate STC1 that maintains local Ca(2) (+) homeostasis. In this study, we investigated Casr-mediated signaling and its regulation of Stc1 and cyclooxygenase-2 (Cox2) expression/function using a primary gill-cell culture model. The biochemical responses of gill cells isolated from Japanese eels to an increasing concentration of extracellular Ca(2) (+) (0.1-1 mM) were tested. This stimulation led to a transient increase in phosphatidylcholine-phospholipase C (PC-PLC) activity, followed by activation of ERK and inositol 1,4,5-trisphosphate-Ca(2) (+)/calmodulin-dependent protein kinase 2 (CaMK2) signaling pathways. Cotreatment with the calcimimetic R467 caused synergistic effects on Ca(2) (+)-stimulated PC-PLC activity, ERK signaling, and CaMK2 signaling. The activation of the CaSR-PLC-ERK pathway was associated with increased expression levels of Stc1 and Cox2 as confirmed by the inhibition of Erk using a chemical inhibitor, PD98059. Functionally, Ca(2) (+)/R-467 pretreatment was found to protect cells from thapsigargin-induced cell death. Inhibition of COX2 activity using NS398 abolished this protection, while transduction of STC1 lentiviral particles in the gill cells increased the protective effects. Collectively, our data revealed the expression of functional CaSR in gill tissues. The identification of the CaSR-STC1/COX2-mediated protective pathway in gill cells sheds light on a possible cellular protective mechanism against an increase in intracellular Ca(2) (+) levels associated with transepithelial Ca(2) (+) transport.
Collapse
Affiliation(s)
- J Gu
- Department of BiologyCroucher Institute for Environmental Sciences, Hong Kong Baptist University, Kowloon Tong, Hong Kong, China
| | - A Y S Law
- Department of BiologyCroucher Institute for Environmental Sciences, Hong Kong Baptist University, Kowloon Tong, Hong Kong, China
| | - B H Y Yeung
- Department of BiologyCroucher Institute for Environmental Sciences, Hong Kong Baptist University, Kowloon Tong, Hong Kong, China
| | - C K C Wong
- Department of BiologyCroucher Institute for Environmental Sciences, Hong Kong Baptist University, Kowloon Tong, Hong Kong, China
| |
Collapse
|
42
|
Goolam MA, Ward JH, Avlani VA, Leach K, Christopoulos A, Conigrave AD. Roles of intraloops-2 and -3 and the proximal C-terminus in signalling pathway selection from the human calcium-sensing receptor. FEBS Lett 2014; 588:3340-6. [PMID: 25080008 DOI: 10.1016/j.febslet.2014.07.022] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2014] [Revised: 07/03/2014] [Accepted: 07/16/2014] [Indexed: 12/24/2022]
Abstract
The calcium-sensing receptor (CaSR) couples to signalling pathways via intracellular loops 2 and 3, and the C-terminus. However, the requirements for signalling are largely undefined. We investigated the impacts of selected point mutations in iL-2 (F706A) and iL-3 (L797A and E803A), and a truncation of the C-terminus (R866X) on extracellular Ca(2+) (Ca(2+)o)-stimulated phosphatidylinositol-specific phospholipase-C (PI-PLC) and various other signalling responses. CaSR-mediated activation of PI-PLC was markedly attenuated in all four mutants and similar suppressions were observed for Ca(2+)o-stimulated ERK1/2 phosphorylation. Ca(2+)o-stimulated intracellular Ca(2+) (Ca(2+)i) mobilization, however, was relatively preserved for the iL-2 and iL-3 mutants and suppression of adenylyl cyclase was unaffected by either E803A or R866X. The CaSR selects for specific signalling pathways via the proximal C-terminus and key residues in iL-2, iL-3.
Collapse
Affiliation(s)
- Mahvash A Goolam
- School of Molecular Bioscience, University of Sydney, NSW 2006, Australia
| | - James H Ward
- School of Molecular Bioscience, University of Sydney, NSW 2006, Australia
| | - Vimesh A Avlani
- School of Molecular Bioscience, University of Sydney, NSW 2006, Australia
| | - Katie Leach
- Monash Institute of Pharmaceutical Sciences and Department of Pharmacology, Monash University, Parkville, Victoria 3052, Australia
| | - Arthur Christopoulos
- Monash Institute of Pharmaceutical Sciences and Department of Pharmacology, Monash University, Parkville, Victoria 3052, Australia
| | - Arthur D Conigrave
- School of Molecular Bioscience, University of Sydney, NSW 2006, Australia.
| |
Collapse
|
43
|
Emerging roles of the extracellular calcium-sensing receptor in nutrient sensing: control of taste modulation and intestinal hormone secretion. Br J Nutr 2014; 111 Suppl 1:S16-22. [PMID: 24382107 DOI: 10.1017/s0007114513002250] [Citation(s) in RCA: 40] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
The extracellular Ca-sensing receptor (CaSR) is a sensor for a number of key nutrients within the body, including Ca ions (Ca²⁺) and L-amino acids. The CaSR is expressed in a number of specialised cells within the gastrointestinal (GI) tract, and much work has been done to examine CaSR's role as a nutrient sensor in this system. This review article examines two emerging roles for the CaSR within the GI tract--as a mediator of kokumi taste modulation in taste cells and as a regulator of dietary hormone release in response to L-amino acids in the intestine.
Collapse
|
44
|
Tao YX, Liang XF. G Protein-Coupled Receptors as Regulators of Glucose Homeostasis and Therapeutic Targets for Diabetes Mellitus. PROGRESS IN MOLECULAR BIOLOGY AND TRANSLATIONAL SCIENCE 2014; 121:1-21. [DOI: 10.1016/b978-0-12-800101-1.00001-6] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
|
45
|
Wauson EM, Lorente-Rodríguez A, Cobb MH. Minireview: Nutrient sensing by G protein-coupled receptors. Mol Endocrinol 2013; 27:1188-97. [PMID: 23820899 DOI: 10.1210/me.2013-1100] [Citation(s) in RCA: 64] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023] Open
Abstract
G protein-coupled receptors (GPCRs) are membrane proteins that recognize molecules in the extracellular milieu and transmit signals inside cells to regulate their behaviors. Ligands for many GPCRs are hormones or neurotransmitters that direct coordinated, stereotyped adaptive responses. Ligands for other GPCRs provide information to cells about the extracellular environment. Such information facilitates context-specific decision making that may be cell autonomous. Among ligands that are important for cellular decisions are amino acids, required for continued protein synthesis, as metabolic starting materials and energy sources. Amino acids are detected by a number of class C GPCRs. One cluster of amino acid-sensing class C GPCRs includes umami and sweet taste receptors, GPRC6A, and the calcium-sensing receptor. We have recently found that the umami taste receptor heterodimer T1R1/T1R3 is a sensor of amino acid availability that regulates the activity of the mammalian target of rapamycin. This review focuses on an array of findings on sensing amino acids and sweet molecules outside of neurons by this cluster of class C GPCRs and some of the physiologic processes regulated by them.
Collapse
Affiliation(s)
- Eric M Wauson
- Department of Pharmacology, University of Texas Southwestern Medical Center at Dallas, Dallas, Texas 75390-9041, USA
| | | | | |
Collapse
|
46
|
Conigrave AD, Ward DT. Calcium-sensing receptor (CaSR): pharmacological properties and signaling pathways. Best Pract Res Clin Endocrinol Metab 2013; 27:315-31. [PMID: 23856262 DOI: 10.1016/j.beem.2013.05.010] [Citation(s) in RCA: 160] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/04/2023]
Abstract
In this article we consider the mechanisms by which the calcium-sensing receptor (CaSR) induces its cellular responses via the control (activation or inhibition) of signaling pathways. We consider key features of CaSR-mediated signaling including its control of the heterotrimeric G-proteins Gq/11, Gi/o and G12/13 and the downstream consequences recognizing that very few CaSR-mediated cell phenomena have been fully described. We also consider the manner in which the CaSR contributes to the formation of specific signaling scaffolds via peptide recognition sequences in its intracellular C-terminal along with the origins of its high level of cooperativity, particularly for Ca(2+)o, and its remarkable resistance to desensitization. We also consider the nature of the mechanisms by which the CaSR controls oscillatory and sustained Ca(2+)i mobilizing responses and inhibits or elevates cyclic adenosine monophosphate (cAMP) levels dependent on the cellular and signaling context. Finally, we consider the diversity of the receptor's ligands, ligand binding sites and broader compartment-dependent physiological roles leading to the identification of pronounced ligand-biased signaling for agonists including Sr(2+) and modulators including l-amino acids and the clinically effective calcimimetic cinacalcet. We note the implications of these findings for the development of new designer drugs that might target the CaSR in pathophysiological contexts beyond those established for the treatment of disorders of calcium metabolism.
Collapse
Affiliation(s)
- Arthur D Conigrave
- School of Molecular Bioscience, University of Sydney, NSW 2006, Australia.
| | | |
Collapse
|
47
|
Avlani VA, Ma W, Mun HC, Leach K, Delbridge L, Christopoulos A, Conigrave AD. Calcium-sensing receptor-dependent activation of CREB phosphorylation in HEK293 cells and human parathyroid cells. Am J Physiol Endocrinol Metab 2013; 304:E1097-104. [PMID: 23531616 DOI: 10.1152/ajpendo.00054.2013] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
In addition to its acute effects on hormone secretion, epithelial transport, and shape change, the calcium-sensing receptor (CaSR) modulates the expression of genes that control cell survival, proliferation, and differentiation as well as the synthesis of peptide hormones and enzymes. In the present study, we investigated the impacts of a CaSR agonist and several CaSR modulators on phosphorylation of transcription factor CREB residue Ser(133) in CaSR-expressing HEK293 (HEK-CaSR) cells and human adenomatous parathyroid cells. Elevated Ca(2+)o concentration had no effect on CREB phosphorylation (p-CREB) in control HEK293 cells but stimulated p-CREB in both HEK-CaSR cells and human parathyroid cells. In addition, p-CREB was stimulated by the positive modulator cinacalcet and inhibited by the negative modulator NPS 2143 in both CaSR-expressing cell types. Two positive modulators that bind in the receptor's Venus Fly Trap domain, l-phenylalanine and S-methylglutathione, had no effect on p-CREB in HEK-CaSR cells, demonstrating the existence of pronounced signaling bias. Analysis of the signaling pathways using specific inhibitors demonstrated that phosphoinositide-specific phospholipase C and conventional protein kinase C isoforms make major contributions to Ca(2+)o-induced p-CREB in both cell-types, suggesting key roles for Gq/11. In addition, in parathyroid cells but not HEK-CaSR cells, activation of p-CREB was dependent on Gi/o, demonstrating the existence of cell type-specific signaling.
Collapse
Affiliation(s)
- Vimesh A Avlani
- School of Molecular Bioscience, University of Sydney, Sydney, New South Wales, Australia
| | | | | | | | | | | | | |
Collapse
|
48
|
Vertebrate extracellular calcium-sensing receptor evolution: selection in relation to life history and habitat. COMPARATIVE BIOCHEMISTRY AND PHYSIOLOGY D-GENOMICS & PROTEOMICS 2012; 8:86-94. [PMID: 23321268 DOI: 10.1016/j.cbd.2012.12.004] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/26/2012] [Revised: 12/07/2012] [Accepted: 12/09/2012] [Indexed: 11/20/2022]
Abstract
Ionic calcium (Ca(2+)) supports essential functions within physiological systems, and consequently its concentration is homeostatically regulated within narrow bounds in the body fluids of animals through endocrine effects at ion-transporting osmoregulatory tissues. In vertebrates, extracellular Ca(2+) is detected at the cell surface by the extracellular calcium-sensing receptor (CaSR), a member of the G protein-coupled receptor (GPCR) superfamily. Interestingly, the taxonomic distribution of CaSRs is restricted to vertebrates, with some CaSR-like receptors apparently present in non-vertebrate chordates. Since bone is a known Ca(2+) storage site and is characteristically restricted to the vertebrate lineage, we hypothesized a functional association of CaSR with vertebrate skeleton that may have an ancient origin. Protein sequence alignment and phylogenetic analysis of vertebrate CaSRs and related GPCRs of the glutamate receptor-like family expose similarities and indel differences among these receptors, and reveal the evolutionary history of CaSRs. Evolutionary selection was tested statistically by evaluating the relationship between non-synonymous (replacement, dN) versus synonymous (silent, dS) amino acid substitution rates (as dN/dS) of protein-coding DNA sequences among branches of the estimated protein phylogeny. On a background of strong purifying selection (dN/dS<1) in the CaSR phylogeny, statistical evidence for adaptive evolution (dN/dS>1) was detected on some branches to major clades in the CaSR phylogeny, especially to the tetrapod vertebrate CaSRs and chordate CaSR-like branches. Testing also revealed overall purifying selection at the codon level. At some sites relaxation from strong purifying selection was seen, but evidence for adaptive evolution was not detected for individual sites. The results suggest purifying selection of CaSRs, and of adaptive evolution among some major vertebrate clades, reflecting clade specific differences in natural history and organismal biology, including skeletal involvement in calcium homeostasis.
Collapse
|
49
|
Kilberg MS, Balasubramanian M, Fu L, Shan J. The transcription factor network associated with the amino acid response in mammalian cells. Adv Nutr 2012; 3:295-306. [PMID: 22585903 PMCID: PMC3649461 DOI: 10.3945/an.112.001891] [Citation(s) in RCA: 99] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
Mammals exhibit multiple adaptive mechanisms that sense and respond to fluctuations in dietary nutrients. Consumption of reduced total dietary protein or a protein diet that is deficient in 1 or more of the essential amino acids triggers wide-ranging changes in feeding behavior and gene expression. At the level of individual cells, dietary protein deficiency is manifested as amino acid (AA) deprivation, which activates the AA response (AAR). The AAR is composed of a collection of signal transduction pathways that terminate in specific transcriptional programs designed to catalyze adaptation to the nutrient stress or, ultimately, undergo apoptosis. Independently of the AAR, endoplasmic reticulum stress activates 3 signaling pathways, collectively referred to as the unfolded protein response. The transcription factor activating transcription factor 4 is one of the terminal transcriptional mediators for both the AAR and the unfolded protein response, leading to a significant degree of overlap with regard to the target genes for these stress pathways. Over the past 5 y, research has revealed that the basic leucine zipper superfamily of transcription factors plays the central role in the AAR. Formation of both homo- and heterodimers among the activating transcription factor, CCAAT enhancer-binding protein, and FOS/JUN families of basic leucine zipper proteins forms the nucleus of a highly integrated transcription factor network that determines the initiation, magnitude, and duration of the cellular response to dietary protein or AA limitation.
Collapse
|
50
|
Gastrointestinal chemosensation: chemosensory cells in the alimentary tract. Histochem Cell Biol 2012; 138:13-24. [DOI: 10.1007/s00418-012-0954-z] [Citation(s) in RCA: 53] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/01/2012] [Indexed: 12/29/2022]
|