1
|
Ardecky R, Dengler DG, Harikumar KG, Abelman MM, Zou J, Kramer BA, Ganji SR, Olson S, Ly A, Puvvula N, Ma CT, Ramachandra R, Sergienko EA, Miller LJ. Structure-activity relationships of thiadiazole agonists of the human secretin receptor. SLAS DISCOVERY : ADVANCING LIFE SCIENCES R & D 2024; 29:100176. [PMID: 39122117 PMCID: PMC11439415 DOI: 10.1016/j.slasd.2024.100176] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/15/2024] [Revised: 08/01/2024] [Accepted: 08/06/2024] [Indexed: 08/12/2024]
Abstract
Agonists of the secretin receptor have potential applications for diseases of the cardiovascular, gastrointestinal, and metabolic systems, yet no clinically-active non-peptidyl agonists of this receptor have yet been developed. In the current work, we have identified a new small molecule lead compound with this pharmacological profile. We have prepared and characterized a systematic structure-activity series around this thiadiazole scaffold to better understand the molecular determinants of its activity. We were able to enhance the in vitro activity and to maintain the specificity of the parent compound. We found the most active candidate to be quite stable in plasma, although it was metabolized by hepatic microsomes. This chemical probe should be useful for in vitro studies and needs to be tested for in vivo pharmacological activity. This could be an important lead toward the development of a first-in-class orally active agonist of the secretin receptor, which could be useful for multiple disease states.
Collapse
Affiliation(s)
- Robert Ardecky
- From Conrad Prebys Center for Chemical Genomics, Sanford Burnham Prebys Medical Discovery Institute, La Jolla, CA, USA.
| | - Daniela G Dengler
- From Conrad Prebys Center for Chemical Genomics, Sanford Burnham Prebys Medical Discovery Institute, La Jolla, CA, USA
| | - Kaleeckal G Harikumar
- Department of Molecular Pharmacology and Experimental Therapeutics, Mayo Clinic, Scottsdale, AZ, USA
| | - Mathew M Abelman
- From Conrad Prebys Center for Chemical Genomics, Sanford Burnham Prebys Medical Discovery Institute, La Jolla, CA, USA
| | - Jiwen Zou
- From Conrad Prebys Center for Chemical Genomics, Sanford Burnham Prebys Medical Discovery Institute, La Jolla, CA, USA
| | - Bryan A Kramer
- From Conrad Prebys Center for Chemical Genomics, Sanford Burnham Prebys Medical Discovery Institute, La Jolla, CA, USA
| | - Santhi Reddy Ganji
- From Conrad Prebys Center for Chemical Genomics, Sanford Burnham Prebys Medical Discovery Institute, La Jolla, CA, USA
| | - Steve Olson
- From Conrad Prebys Center for Chemical Genomics, Sanford Burnham Prebys Medical Discovery Institute, La Jolla, CA, USA
| | - Alina Ly
- From Conrad Prebys Center for Chemical Genomics, Sanford Burnham Prebys Medical Discovery Institute, La Jolla, CA, USA
| | - Nikhil Puvvula
- From Conrad Prebys Center for Chemical Genomics, Sanford Burnham Prebys Medical Discovery Institute, La Jolla, CA, USA
| | - Chen-Ting Ma
- From Conrad Prebys Center for Chemical Genomics, Sanford Burnham Prebys Medical Discovery Institute, La Jolla, CA, USA
| | - Raghuveer Ramachandra
- From Conrad Prebys Center for Chemical Genomics, Sanford Burnham Prebys Medical Discovery Institute, La Jolla, CA, USA
| | - Eduard A Sergienko
- From Conrad Prebys Center for Chemical Genomics, Sanford Burnham Prebys Medical Discovery Institute, La Jolla, CA, USA.
| | - Laurence J Miller
- Department of Molecular Pharmacology and Experimental Therapeutics, Mayo Clinic, Scottsdale, AZ, USA.
| |
Collapse
|
2
|
Parker MJ, Lee H, Yao S, Irwin S, Hwang S, Belanger K, de Mare SW, Surgenor R, Yan L, Gee P, Morla S, Puyang X, Hsiao P, Zeng H, Zhu P, Korpal M, Dransfield P, Bolduc DM, Larsen NA. Identification of 2-Sulfonyl/Sulfonamide Pyrimidines as Covalent Inhibitors of WRN Using a Multiplexed High-Throughput Screening Assay. Biochemistry 2023; 62:2147-2160. [PMID: 37403936 PMCID: PMC10358344 DOI: 10.1021/acs.biochem.2c00599] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2022] [Revised: 01/03/2023] [Indexed: 07/06/2023]
Abstract
Werner syndrome protein (WRN) is a multifunctional enzyme with helicase, ATPase, and exonuclease activities that are necessary for numerous DNA-related transactions in the human cell. Recent studies identified WRN as a synthetic lethal target in cancers characterized by genomic microsatellite instability resulting from defects in DNA mismatch repair pathways. WRN's helicase activity is essential for the viability of these high microsatellite instability (MSI-H) cancers and thus presents a therapeutic opportunity. To this end, we developed a multiplexed high-throughput screening assay that monitors exonuclease, ATPase, and helicase activities of full-length WRN. This screening campaign led to the discovery of 2-sulfonyl/sulfonamide pyrimidine derivatives as novel covalent inhibitors of WRN helicase activity. The compounds are specific for WRN versus other human RecQ family members and show competitive behavior with ATP. Examination of these novel chemical probes established the sulfonamide NH group as a key driver of compound potency. One of the leading compounds, H3B-960, showed consistent activities in a range of assays (IC50 = 22 nM, KD = 40 nM, KI = 32 nM), and the most potent compound identified, H3B-968, has inhibitory activity IC50 ∼ 10 nM. These kinetic properties trend toward other known covalent druglike molecules. Our work provides a new avenue for screening WRN for inhibitors that may be adaptable to different therapeutic modalities such as targeted protein degradation, as well as a proof of concept for the inhibition of WRN helicase activity by covalent molecules.
Collapse
Affiliation(s)
- Mackenzie J. Parker
- H3 Biomedicine, Inc., 300 Technology Square, Suite 5, Cambridge, Massachusetts 02139, United States
| | - Hyelee Lee
- H3 Biomedicine, Inc., 300 Technology Square, Suite 5, Cambridge, Massachusetts 02139, United States
| | - Shihua Yao
- H3 Biomedicine, Inc., 300 Technology Square, Suite 5, Cambridge, Massachusetts 02139, United States
| | - Sean Irwin
- H3 Biomedicine, Inc., 300 Technology Square, Suite 5, Cambridge, Massachusetts 02139, United States
| | - Sunil Hwang
- H3 Biomedicine, Inc., 300 Technology Square, Suite 5, Cambridge, Massachusetts 02139, United States
| | - Kylie Belanger
- H3 Biomedicine, Inc., 300 Technology Square, Suite 5, Cambridge, Massachusetts 02139, United States
| | - Sofia Woo de Mare
- H3 Biomedicine, Inc., 300 Technology Square, Suite 5, Cambridge, Massachusetts 02139, United States
| | - Richard Surgenor
- H3 Biomedicine, Inc., 300 Technology Square, Suite 5, Cambridge, Massachusetts 02139, United States
| | - Lu Yan
- H3 Biomedicine, Inc., 300 Technology Square, Suite 5, Cambridge, Massachusetts 02139, United States
| | - Patricia Gee
- H3 Biomedicine, Inc., 300 Technology Square, Suite 5, Cambridge, Massachusetts 02139, United States
| | - Shravan Morla
- H3 Biomedicine, Inc., 300 Technology Square, Suite 5, Cambridge, Massachusetts 02139, United States
| | - Xiaoling Puyang
- H3 Biomedicine, Inc., 300 Technology Square, Suite 5, Cambridge, Massachusetts 02139, United States
| | - Peng Hsiao
- H3 Biomedicine, Inc., 300 Technology Square, Suite 5, Cambridge, Massachusetts 02139, United States
| | - Hao Zeng
- H3 Biomedicine, Inc., 300 Technology Square, Suite 5, Cambridge, Massachusetts 02139, United States
| | - Ping Zhu
- H3 Biomedicine, Inc., 300 Technology Square, Suite 5, Cambridge, Massachusetts 02139, United States
| | - Manav Korpal
- H3 Biomedicine, Inc., 300 Technology Square, Suite 5, Cambridge, Massachusetts 02139, United States
| | - Paul Dransfield
- H3 Biomedicine, Inc., 300 Technology Square, Suite 5, Cambridge, Massachusetts 02139, United States
| | - David M. Bolduc
- H3 Biomedicine, Inc., 300 Technology Square, Suite 5, Cambridge, Massachusetts 02139, United States
| | - Nicholas A. Larsen
- H3 Biomedicine, Inc., 300 Technology Square, Suite 5, Cambridge, Massachusetts 02139, United States
| |
Collapse
|
3
|
Finneran D, Li Q, Subbarayan MS, Joly-Amado A, Kamath S, Dengler DG, Gordon MN, Jackson MR, Morgan D, Bickford PC, Smith LH, Nash KR. Concentration and proteolysis of CX3CL1 may regulate the microglial response to CX3CL1. Glia 2023; 71:245-258. [PMID: 36106533 PMCID: PMC9772123 DOI: 10.1002/glia.24269] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2022] [Revised: 08/25/2022] [Accepted: 08/26/2022] [Indexed: 12/24/2022]
Abstract
Fractalkine (FKN) is a membrane-bound chemokine that can be cleaved by proteases such as ADAM 10, ADAM 17, and cathepsin S to generate soluble fragments. Studies using different forms of the soluble FKN yield conflicting results in vivo. These observations prompted us to investigate the function and pharmacology of two commonly used isoforms of FKN, a human full-length soluble FKN (sFKN), and a human chemokine domain only FKN (cdFKN). Both are prevalent in the literature and are often assumed to be functionally equivalent. We observed that recombinant sFKN and cdFKN exhibit similar potencies in a cell-based cAMP assay, but binding affinity for CX3CR1 was modestly different. There was a 10-fold difference in potency between sFKN and cdFKN when assessing their ability to stimulate β-arrestin recruitment. Interestingly, high concentrations of FKN, regardless of cleavage variant, were ineffective at reducing pro-inflammatory microglial activation and may induce a pro-inflammatory response. This effect was observed in mouse and rat primary microglial cells as well as microglial cell lines. The inflammatory response was exacerbated in aged microglia, which is known to exhibit age-related inflammatory phenotypes. We observed the same effects in Cx3cr1-/- primary microglia and therefore speculate that an alternative FKN receptor may exist. Collectively, these data provide greater insights into the function and pharmacology of these common FKN reagents, which may clarify conflicting reports and urge greater caution in the selection of FKN peptides for use in in vitro and in vivo studies and the interpretation of results obtained using these differing peptides.
Collapse
Affiliation(s)
- Dylan Finneran
- Department of Molecular Pharmacology and Physiology, Morsani College of Medicine, University of South Florida, 12901 Bruce B Downs Blvd, Tampa FL-33612, USA
- Michigan State University, Department of Translational Neuroscience, 400 Monroe Ave. NW, Grand Rapids, MI, United States
| | - Qingyou Li
- Department of Molecular Pharmacology and Physiology, Morsani College of Medicine, University of South Florida, 12901 Bruce B Downs Blvd, Tampa FL-33612, USA
| | - Meena S. Subbarayan
- Department of Molecular Pharmacology and Physiology, Morsani College of Medicine, University of South Florida, 12901 Bruce B Downs Blvd, Tampa FL-33612, USA
- Center for Excellence in Aging and Brain Repair, Department of Neurosurgery and Brain Repair, Morsani College of Medicine, University of South Florida, 12901 Bruce B Downs Blvd, Tampa FL-33612, USA
- Gladstone Institute of Neurological Disease, Gladstone Institutes, 1650 Owens St, San Francisco, CA 94158
| | - Aurelie Joly-Amado
- Department of Molecular Pharmacology and Physiology, Morsani College of Medicine, University of South Florida, 12901 Bruce B Downs Blvd, Tampa FL-33612, USA
| | - Siddharth Kamath
- Department of Molecular Pharmacology and Physiology, Morsani College of Medicine, University of South Florida, 12901 Bruce B Downs Blvd, Tampa FL-33612, USA
| | - Daniela G. Dengler
- Conrad Prebys Center for Chemical Genomics, Sandford Burnham Prebys Medical Discovery Institute, 10901 North Torrey Pines Road, La Jolla, CA 92037
| | - Marcia N. Gordon
- Michigan State University, Department of Translational Neuroscience, 400 Monroe Ave. NW, Grand Rapids, MI, United States
| | - Michael R. Jackson
- Conrad Prebys Center for Chemical Genomics, Sandford Burnham Prebys Medical Discovery Institute, 10901 North Torrey Pines Road, La Jolla, CA 92037
| | - Dave Morgan
- Michigan State University, Department of Translational Neuroscience, 400 Monroe Ave. NW, Grand Rapids, MI, United States
| | - Paula C. Bickford
- Department of Molecular Pharmacology and Physiology, Morsani College of Medicine, University of South Florida, 12901 Bruce B Downs Blvd, Tampa FL-33612, USA
- Center for Excellence in Aging and Brain Repair, Department of Neurosurgery and Brain Repair, Morsani College of Medicine, University of South Florida, 12901 Bruce B Downs Blvd, Tampa FL-33612, USA
- Research Service, James A Haley Veterans Hospital, 13000 Bruce B Downs Blvd, Tampa FL-33612, USA
| | - Layton H. Smith
- Conrad Prebys Center for Chemical Genomics, Sandford Burnham Prebys Medical Discovery Institute, 10901 North Torrey Pines Road, La Jolla, CA 92037
| | - Kevin R. Nash
- Department of Molecular Pharmacology and Physiology, Morsani College of Medicine, University of South Florida, 12901 Bruce B Downs Blvd, Tampa FL-33612, USA
| |
Collapse
|
4
|
Dengler DG, Sun Q, Harikumar KG, Miller LJ, Sergienko EA. Screening for positive allosteric modulators of cholecystokinin type 1 receptor potentially useful for management of obesity. SLAS DISCOVERY : ADVANCING LIFE SCIENCES R & D 2022; 27:384-394. [PMID: 35850480 PMCID: PMC9580343 DOI: 10.1016/j.slasd.2022.07.001] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/29/2022] [Revised: 07/11/2022] [Accepted: 07/12/2022] [Indexed: 06/15/2023]
Abstract
Obesity has become a prevailing health burden globally and particularly in the US. It is associated with many health problems, including cardiovascular disease, diabetes and poorer mental health. Hence, there is a high demand to find safe and effective therapeutics for sustainable weight loss. Cholecystokinin (CCK) has been implicated as one of the first gastrointestinal hormones to reduce overeating and suppress appetite by activating the type 1 cholecystokinin receptor (CCK1R). Several drug development campaigns have focused on finding CCK1R-specific agonists, which showed promising efficacy for reducing meal size and weight, but fell short on FDA approval, likely due to side effects associated with potent, long-lasting activation of CCK1Rs. Positive allosteric modulators (PAMs) without inherent agonist activity have been proposed to overcome the shortcomings of traditional, orthosteric agonists and restore CCK1R signaling in failing physiologic systems. However, drug discovery campaigns searching for such novel acting CCK1R agents remain limited. Here we report a high-throughput screening effort and the establishment of a testing funnel, which led to the identification of novel CCK1R modulators. We utilized IP-One accumulation to develop robust functional equilibrium assays tailored to either detect PAMs, agonists or non-specific activators. In addition, we established the CCK1R multiplex PAM assay as a novel method to evaluate functional selectivity capable of recording CCK1R-induced cAMP accumulation and β-arrestin recruitment in the same well. This selection and arrangement of methods enabled the discovery of three scaffolds, which we characterized and validated in an array of functional and binding assays. We found two hits incorporating a tetracyclic scaffold that significantly enhanced CCK signaling at CCK1Rs without intrinsically activating CCK1Rs in an overexpressing system. Our results demonstrate that a well-thought-out testing funnel can identify small molecules with a distinct pharmacological profile and provides an important milestone for the development of novel potential treatments of obesity.
Collapse
Affiliation(s)
- Daniela G Dengler
- Conrad Prebys Center for Chemical Genomics, Sanford Burnham Prebys Medical Discovery Institute, La Jolla, California, USA.
| | - Qing Sun
- Conrad Prebys Center for Chemical Genomics, Sanford Burnham Prebys Medical Discovery Institute, La Jolla, California, USA
| | - Kaleeckal G Harikumar
- Department of Molecular Pharmacology and Experimental Therapeutics, Mayo Clinic, Scottsdale, Arizona, USA
| | - Laurence J Miller
- Department of Molecular Pharmacology and Experimental Therapeutics, Mayo Clinic, Scottsdale, Arizona, USA.
| | - Eduard A Sergienko
- Conrad Prebys Center for Chemical Genomics, Sanford Burnham Prebys Medical Discovery Institute, La Jolla, California, USA.
| |
Collapse
|
5
|
Lopez JB, Chang CC, Kuo YM, Chan MF, Winn BJ. Oxytocin and secretin receptors - implications for dry eye syndrome and ocular pain. FRONTIERS IN OPHTHALMOLOGY 2022; 2:948481. [PMID: 38983562 PMCID: PMC11182124 DOI: 10.3389/fopht.2022.948481] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 05/19/2022] [Accepted: 07/11/2022] [Indexed: 07/11/2024]
Abstract
Dry eye syndrome, a form of ocular surface inflammation, and chronic ocular pain are common conditions impacting activities of daily living and quality of life. Oxytocin and secretin are peptide hormones that have been shown to synergistically reduce inflammation in various tissues and attenuate the pain response at both the neuron and brain level. The oxytocin receptor (OXTR) and secretin receptor (SCTR) have been found in a wide variety of tissues and organs, including the eye. We reviewed the current literature of in vitro experiments, animal models, and human studies that examine the anti-inflammatory and anti-nociceptive roles of oxytocin and secretin. This review provides an overview of the evidence supporting oxytocin and secretin as the basis for novel treatments of dry eye and ocular pain syndromes.
Collapse
Affiliation(s)
- Jacqueline B Lopez
- Department of Ophthalmology, University of California, San Francisco, San Francisco, CA, United States
| | - Chih-Chiun Chang
- Department of Ophthalmology, University of California, San Francisco, San Francisco, CA, United States
| | - Yien-Ming Kuo
- Department of Ophthalmology, University of California, San Francisco, San Francisco, CA, United States
| | - Matilda F Chan
- Department of Ophthalmology, University of California, San Francisco, San Francisco, CA, United States
- Department of Ophthalmology, Zuckerberg San Francisco General Hospital and Trauma Center, San Francisco, CA, United States
- Francis I. Proctor Foundation, University of California, San Francisco, San Francisco, CA, United States
| | - Bryan J Winn
- Department of Ophthalmology, University of California, San Francisco, San Francisco, CA, United States
- Surgical Service, San Francisco Veterans Affairs Medical Center, San Francisco, CA, United States
| |
Collapse
|
6
|
Milburn JE, Harikumar KG, Piper SJ, Raval S, Christopoulos A, Wootten D, Sexton PM, Miller LJ. Secretin Amino-Terminal Structure-Activity Relationships and Complementary Mutagenesis at the Site of Docking to the Secretin Receptor. Mol Pharmacol 2022; 101:400-407. [PMID: 35351821 PMCID: PMC11033956 DOI: 10.1124/molpharm.122.000502] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2022] [Accepted: 03/22/2022] [Indexed: 04/24/2024] Open
Abstract
Class B1 G protein-coupled receptors are activated by peptides, with amino-terminal regions critical for biologic activity. Although high resolution structures exist, understanding of key features of the peptide activation domain that drive signaling is limited. In the secretin receptor (SecR) structure, interactions are observed between peptide residues His1 and Ser2 and seventh transmembrane segment (TM7) receptor residue E373. We interrogated these interactions using systematic structure-activity analysis of peptide and receptor. His1 was critical for binding and cAMP responses, but its orientation was not critical, and substitution could independently modify affinity and efficacy. Ser2 was also critical, with all substitutions reducing peptide affinity and functional responses proportionally. Mutation of E373 to conserved acidic Asp (E373D), uncharged polar Gln (E373Q), or charge-reversed basic Arg (E373R) did not alter receptor expression, with all exhibiting secretin-dependent cAMP accumulation. All position 373 mutants displayed reduced binding affinities and cAMP potencies for many peptide analogs, although relative effects of position 1 peptides were similar whereas position 2 peptides exhibited substantial differences. The peptide including basic Lys in position 2 was active at SecR having acidic Glu in position 373 and at E373D while exhibiting minimal activity at those receptors in which an acidic residue is absent in this position (E373Q and E373R). In contrast, the peptide including acidic Glu in position 2 was equipotent with secretin at E373R while being much less potent than secretin at wild-type SecR and E373D. These data support functional importance of a charge-charge interaction between the amino-terminal region of secretin and the top of TM7. SIGNIFICANCE STATEMENT: This work refines our molecular understanding of the activation mechanisms of class B1 G protein-coupled receptors. The amino-terminal region of secretin interacts with the seventh transmembrane segment of its receptor with structural specificity and with a charge-charge interaction helping to drive functional activation.
Collapse
Affiliation(s)
- Juliana E Milburn
- Department of Molecular Pharmacology and Experimental Therapeutics, Mayo Clinic, Scottsdale, Arizona (J.E.M., K.G.H., S.R., L.J.M.) and Drug Discovery Biology and Australian Research Council Centre for Cryo-Electron Microscopy of Membrane Proteins, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, Australia (S.J.P., A.C., D.W., P.M.S.)
| | - Kaleeckal G Harikumar
- Department of Molecular Pharmacology and Experimental Therapeutics, Mayo Clinic, Scottsdale, Arizona (J.E.M., K.G.H., S.R., L.J.M.) and Drug Discovery Biology and Australian Research Council Centre for Cryo-Electron Microscopy of Membrane Proteins, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, Australia (S.J.P., A.C., D.W., P.M.S.)
| | - Sarah J Piper
- Department of Molecular Pharmacology and Experimental Therapeutics, Mayo Clinic, Scottsdale, Arizona (J.E.M., K.G.H., S.R., L.J.M.) and Drug Discovery Biology and Australian Research Council Centre for Cryo-Electron Microscopy of Membrane Proteins, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, Australia (S.J.P., A.C., D.W., P.M.S.)
| | - Sweta Raval
- Department of Molecular Pharmacology and Experimental Therapeutics, Mayo Clinic, Scottsdale, Arizona (J.E.M., K.G.H., S.R., L.J.M.) and Drug Discovery Biology and Australian Research Council Centre for Cryo-Electron Microscopy of Membrane Proteins, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, Australia (S.J.P., A.C., D.W., P.M.S.)
| | - Arthur Christopoulos
- Department of Molecular Pharmacology and Experimental Therapeutics, Mayo Clinic, Scottsdale, Arizona (J.E.M., K.G.H., S.R., L.J.M.) and Drug Discovery Biology and Australian Research Council Centre for Cryo-Electron Microscopy of Membrane Proteins, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, Australia (S.J.P., A.C., D.W., P.M.S.)
| | - Denise Wootten
- Department of Molecular Pharmacology and Experimental Therapeutics, Mayo Clinic, Scottsdale, Arizona (J.E.M., K.G.H., S.R., L.J.M.) and Drug Discovery Biology and Australian Research Council Centre for Cryo-Electron Microscopy of Membrane Proteins, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, Australia (S.J.P., A.C., D.W., P.M.S.)
| | - Patrick M Sexton
- Department of Molecular Pharmacology and Experimental Therapeutics, Mayo Clinic, Scottsdale, Arizona (J.E.M., K.G.H., S.R., L.J.M.) and Drug Discovery Biology and Australian Research Council Centre for Cryo-Electron Microscopy of Membrane Proteins, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, Australia (S.J.P., A.C., D.W., P.M.S.)
| | - Laurence J Miller
- Department of Molecular Pharmacology and Experimental Therapeutics, Mayo Clinic, Scottsdale, Arizona (J.E.M., K.G.H., S.R., L.J.M.) and Drug Discovery Biology and Australian Research Council Centre for Cryo-Electron Microscopy of Membrane Proteins, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, Australia (S.J.P., A.C., D.W., P.M.S.)
| |
Collapse
|
7
|
Schnabl K, Li Y, U-Din M, Klingenspor M. Secretin as a Satiation Whisperer With the Potential to Turn into an Obesity-curbing Knight. Endocrinology 2021; 162:6294014. [PMID: 34089599 DOI: 10.1210/endocr/bqab113] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/09/2021] [Indexed: 01/01/2023]
Abstract
The obesity pandemic requires effective preventative and therapeutic intervention strategies. Successful and sustained obesity treatment is currently limited to bariatric surgery. Modulating the release of gut hormones is considered promising to mimic bariatric surgery with its beneficial effects on food intake, body weight, and blood glucose levels. The gut peptide secretin was the first molecule to be termed a hormone; nevertheless, only recently has it been established as a legitimate anorexigenic peptide. In contrast to gut hormones that crosstalk with the brain either directly or by afferent neuronal projections, secretin mediates meal-associated brown fat thermogenesis to induce meal termination, thereby qualifying this physiological mechanism as an attractive, peripheral target for the treatment of obesity. In this perspective, it is of pivotal interest to deepen our as yet superficial knowledge on the physiological roles of secretin as well as meal-associated thermogenesis in energy balance and body weight regulation. Of note, the emerging differences between meal-associated thermogenesis and cold-induced thermogenesis must be taken into account. In fact, there is no correlation between these 2 entities. In addition, the investigation of potential effects of secretin in hedonic-driven food intake, bariatric surgery and chronic treatment using suitable application strategies to overcome pharmacokinetic limitations will provide further insight into its potential to influence energy balance. The aim of this article is to review the facts on secretin's metabolic effects, address prevailing gaps in our knowledge, and provide an overview on the opportunities and challenges of the therapeutic potential of secretin in body weight control.
Collapse
Affiliation(s)
- Katharina Schnabl
- Chair for Molecular Nutritional Medicine, TUM School of Life Sciences, Technical University of Munich, Freising, Germany
- EKFZ-Else Kröner Fresenius Center for Nutritional Medicine, Technical University of Munich, Freising, Germany
- ZIEL-Institute for Food & Health, Technical University of Munich, Freising, Germany
| | - Yongguo Li
- Chair for Molecular Nutritional Medicine, TUM School of Life Sciences, Technical University of Munich, Freising, Germany
- EKFZ-Else Kröner Fresenius Center for Nutritional Medicine, Technical University of Munich, Freising, Germany
| | - Mueez U-Din
- Turku PET Centre, Turku University Hospital, Turku, Finland
| | - Martin Klingenspor
- Chair for Molecular Nutritional Medicine, TUM School of Life Sciences, Technical University of Munich, Freising, Germany
- EKFZ-Else Kröner Fresenius Center for Nutritional Medicine, Technical University of Munich, Freising, Germany
- ZIEL-Institute for Food & Health, Technical University of Munich, Freising, Germany
| |
Collapse
|