1
|
Steinke I, Govindarajulu M, Pinky PD, Bloemer J, Yoo S, Ward T, Schaedig T, Young T, Wibowo FS, Suppiramaniam V, Amin RH. Selective PPAR-Delta/PPAR-Gamma Activation Improves Cognition in a Model of Alzheimer's Disease. Cells 2023; 12:1116. [PMID: 37190025 PMCID: PMC10136457 DOI: 10.3390/cells12081116] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2022] [Revised: 03/23/2023] [Accepted: 04/04/2023] [Indexed: 05/17/2023] Open
Abstract
Background: The continuously increasing association of Alzheimer's disease (AD) with increased mortality rates indicates an unmet medical need and the critical need for establishing novel molecular targets for therapeutic potential. Agonists for peroxisomal proliferator activating receptors (PPAR) are known to regulate energy in the body and have shown positive effects against Alzheimer's disease. There are three members of this class (delta, gamma, and alpha), with PPAR-gamma being the most studied, as these pharmaceutical agonists offer promise for AD because they reduce amyloid beta and tau pathologies, display anti-inflammatory properties, and improve cognition. However, they display poor brain bioavailability and are associated with several adverse side effects on human health, thus limiting their clinical application. Methods: We have developed a novel series of PPAR-delta and PPAR-gamma agonists in silico with AU9 as our lead compound that displays selective amino acid interactions focused upon avoiding the Tyr-473 epitope in the PPAR-gamma AF2 ligand binding domain. Results: This design helps to avoid the unwanted side effects of current PPAR-gamma agonists and improve behavioral deficits and synaptic plasticity while reducing amyloid-beta levels and inflammation in 3xTgAD animals. Conclusions: Our innovative in silico design of PPAR-delta/gamma agonists may offer new perspectives for this class of agonists for AD.
Collapse
Affiliation(s)
- Ian Steinke
- Department of Drug Discovery and Development, Auburn University, Auburn, AL 36879, USA
| | - Manoj Govindarajulu
- Department of Drug Discovery and Development, Auburn University, Auburn, AL 36879, USA
| | - Priyanka Das Pinky
- Department of Drug Discovery and Development, Auburn University, Auburn, AL 36879, USA
| | - Jenna Bloemer
- Department of Pharmaceutical and Biomedical Sciences, Touro College of Pharmacy, New York, NY 10027, USA
| | - Sieun Yoo
- Department of Drug Discovery and Development, Auburn University, Auburn, AL 36879, USA
| | - Tracey Ward
- Department of Pharmaceutical Sciences, Ferris State University, Big Rapids, MI 49307, USA
| | - Taylor Schaedig
- Department of Drug Discovery and Development, Auburn University, Auburn, AL 36879, USA
| | - Taylor Young
- Department of Drug Discovery and Development, Auburn University, Auburn, AL 36879, USA
| | - Fajar Setyo Wibowo
- Department of Drug Discovery and Development, Auburn University, Auburn, AL 36879, USA
| | - Vishnu Suppiramaniam
- Department of Drug Discovery and Development, Auburn University, Auburn, AL 36879, USA
- College of Science and Mathematics, Kennesaw State University, Kennesaw, GA 31044, USA
| | - Rajesh H. Amin
- Department of Drug Discovery and Development, Auburn University, Auburn, AL 36879, USA
| |
Collapse
|
2
|
Paterson NM, Al-Zubieri H, Barber MF. Diversification of CD1 Molecules Shapes Lipid Antigen Selectivity. Mol Biol Evol 2021; 38:2273-2284. [PMID: 33528563 PMCID: PMC8136489 DOI: 10.1093/molbev/msab022] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
Molecular studies of host-pathogen evolution have largely focused on the consequences of variation at protein-protein interaction surfaces. The potential for other microbe-associated macromolecules to promote arms race dynamics with host factors remains unclear. The cluster of differentiation 1 (CD1) family of vertebrate cell surface receptors plays a crucial role in adaptive immunity through binding and presentation of lipid antigens to T-cells. Although CD1 proteins present a variety of endogenous and microbial lipids to various T-cell types, they are less diverse within vertebrate populations than the related major histocompatibility complex (MHC) molecules. We discovered that CD1 genes exhibit a high level of divergence between simian primate species, altering predicted lipid-binding properties and T-cell receptor interactions. These findings suggest that lipid-protein conflicts have shaped CD1 genetic variation during primate evolution. Consistent with this hypothesis, multiple primate CD1 family proteins exhibit signatures of repeated positive selection at surfaces impacting antigen presentation, binding pocket morphology, and T-cell receptor accessibility. Using a molecular modeling approach, we observe that interspecies variation as well as single mutations at rapidly-evolving sites in CD1a drastically alter predicted lipid binding and structural features of the T-cell recognition surface. We further show that alterations in both endogenous and microbial lipid-binding affinities influence the ability of CD1a to undergo antigen swapping required for T-cell activation. Together these findings establish lipid-protein interactions as a critical force of host-pathogen conflict and inform potential strategies for lipid-based vaccine development.
Collapse
Affiliation(s)
- Nicole M Paterson
- Institute of Ecology and Evolution, University of Oregon, Eugene, OR 97403, USA.,Department of Chemistry and Biochemistry, University of Oregon, Eugene, OR 97403, USA
| | - Hussein Al-Zubieri
- Institute of Ecology and Evolution, University of Oregon, Eugene, OR 97403, USA.,Department of Chemistry and Biochemistry, University of Oregon, Eugene, OR 97403, USA
| | - Matthew F Barber
- Institute of Ecology and Evolution, University of Oregon, Eugene, OR 97403, USA.,Department of Biology, University of Oregon, Eugene, OR 97403, USA
| |
Collapse
|
3
|
Govindarajulu M, Pinky PD, Bloemer J, Ghanei N, Suppiramaniam V, Amin R. Signaling Mechanisms of Selective PPAR γ Modulators in Alzheimer's Disease. PPAR Res 2018; 2018:2010675. [PMID: 30420872 PMCID: PMC6215547 DOI: 10.1155/2018/2010675] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2018] [Revised: 08/31/2018] [Accepted: 09/13/2018] [Indexed: 01/22/2023] Open
Abstract
Alzheimer's disease (AD) is a chronic neurodegenerative disease characterized by abnormal protein accumulation, synaptic dysfunction, and cognitive impairment. The continuous increase in the incidence of AD with the aged population and mortality rate indicates the urgent need for establishing novel molecular targets for therapeutic potential. Peroxisome proliferator-activated receptor gamma (PPARγ) agonists such as rosiglitazone and pioglitazone reduce amyloid and tau pathologies, inhibit neuroinflammation, and improve memory impairments in several rodent models and in humans with mild-to-moderate AD. However, these agonists display poor blood brain barrier permeability resulting in inadequate bioavailability in the brain and thus requiring high dosing with chronic time frames. Furthermore, these dosing levels are associated with several adverse effects including increased incidence of weight gain, liver abnormalities, and heart failure. Therefore, there is a need for identifying novel compounds which target PPARγ more selectively in the brain and could provide therapeutic benefits without a high incidence of adverse effects. This review focuses on how PPARγ agonists influence various pathologies in AD with emphasis on development of novel selective PPARγ modulators.
Collapse
Affiliation(s)
- Manoj Govindarajulu
- Department of Drug Discovery and Development, Harrison School of Pharmacy, Auburn University, Auburn, AL, USA
| | - Priyanka D. Pinky
- Department of Drug Discovery and Development, Harrison School of Pharmacy, Auburn University, Auburn, AL, USA
| | - Jenna Bloemer
- Department of Drug Discovery and Development, Harrison School of Pharmacy, Auburn University, Auburn, AL, USA
| | - Nila Ghanei
- Department of Drug Discovery and Development, Harrison School of Pharmacy, Auburn University, Auburn, AL, USA
| | - Vishnu Suppiramaniam
- Department of Drug Discovery and Development, Harrison School of Pharmacy, Auburn University, Auburn, AL, USA
- Center for Neuroscience, Auburn University, Auburn, AL, USA
| | - Rajesh Amin
- Department of Drug Discovery and Development, Harrison School of Pharmacy, Auburn University, Auburn, AL, USA
- Center for Neuroscience, Auburn University, Auburn, AL, USA
| |
Collapse
|
4
|
Design, sythesis and evaluation of a series of 3- or 4-alkoxy substituted phenoxy derivatives as PPARs agonists. Oncotarget 2017; 8:20766-20783. [PMID: 28186999 PMCID: PMC5400543 DOI: 10.18632/oncotarget.15198] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2016] [Accepted: 01/25/2017] [Indexed: 11/25/2022] Open
Abstract
Peroxisome proliferators-activated receptors (PPARα, γ and δ) are potentially effective targets for Type 2 diabetes mellitus therapy. The severe effects of known glitazones and the successfully approved agents (saroglitazar and lobeglitazone) motivated us to study novelly potent PPARs drugs with improved safety profile. In this work, we received 15 carboxylic acids based on the combination principle to integrate the polar head of bezafibrate with the hydrophobic tail of pioglitazone. Another 12 tetrazoles based on the bioisosterism principle were obtained accordingly. Furthermore, in vitro PPARs transactivation assays on these 3- or 4-alkoxy substituted phenoxy derivatives afforded six compounds. Interactions and binding stability from the docking analysis and 20 ns molecular dynamic simulations confirmed the representative compounds to be suitable and plausible for PPARs pockets. The above-mentioned results demonstrated that the compounds may be used as reference for further optimization for enhanced PPARs activities and wide safety range.
Collapse
|
5
|
Zhang J, Liu X, Xie XB, Cheng XC, Wang RL. Multitargeted bioactive ligands for PPARs discovered in the last decade. Chem Biol Drug Des 2016; 88:635-663. [PMID: 27317624 DOI: 10.1111/cbdd.12806] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Revised: 05/20/2016] [Indexed: 12/13/2022]
Abstract
Type 2 diabetes took insulin resistance as the main clinical manifestation. PPARs have been reported to be the therapeutic targets of metabolic disorders, such as obesity, hypertension, diabetes, and cardiovascular disease. Previously, PPARγ agonist rosiglitazone was restricted in clinic due to cardiomyocytes infarction, weight gain, and other serious side-effects, which were mainly due to the single and selective PPARγ agonism. In recent years, multitarget-directed PPAR agonists with synergistic reaction as well as fewer side-effect have been the hot topic in designing promising agents. In this review, we updated and generalized the development of PPARγ partial agonists, PPARγ antagonists, PPARα/γ dual agonists, PPARδ partial agonists, PPARδ antagonists, PPARα/δ dual agonists, PPARγ/δ dual agonists, and PPARα/γ/δ pan-agonists published in recent decade. Most of these molecules were modified from known structures or came from high-throughput screening. Among these molecules, some were expected to be promising drugs against metabolic disorders, while others seemed to provide new insight for designing novel PPAR agents.
Collapse
Affiliation(s)
- Jun Zhang
- Tianjin Key Laboratory on Technologies Enabling Development of Clinical Therapeutics and Diagnostics (Theranostics), School of Pharmacy, Tianjin Medical University, Tianjin, China
| | - Xin Liu
- Tianjin Key Laboratory on Technologies Enabling Development of Clinical Therapeutics and Diagnostics (Theranostics), School of Pharmacy, Tianjin Medical University, Tianjin, China
| | - Xian-Bin Xie
- Tianjin Key Laboratory on Technologies Enabling Development of Clinical Therapeutics and Diagnostics (Theranostics), School of Pharmacy, Tianjin Medical University, Tianjin, China
| | - Xian-Chao Cheng
- Tianjin Key Laboratory on Technologies Enabling Development of Clinical Therapeutics and Diagnostics (Theranostics), School of Pharmacy, Tianjin Medical University, Tianjin, China.
| | - Run-Ling Wang
- Tianjin Key Laboratory on Technologies Enabling Development of Clinical Therapeutics and Diagnostics (Theranostics), School of Pharmacy, Tianjin Medical University, Tianjin, China
| |
Collapse
|
6
|
Gim HJ, Li H, Jeong JH, Lee SJ, Sung MK, Song MY, Park BH, Oh SJ, Ryu JH, Jeon R. Design, synthesis, and biological evaluation of a series of alkoxy-3-indolylacetic acids as peroxisome proliferator-activated receptor γ/δ agonists. Bioorg Med Chem 2015; 23:3322-36. [PMID: 25982078 DOI: 10.1016/j.bmc.2015.04.046] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2015] [Revised: 04/15/2015] [Accepted: 04/17/2015] [Indexed: 01/08/2023]
Abstract
A series of alkoxy-3-indolylacetic acid analogs has been discovered as peroxisome proliferator-activated receptor (PPAR) agonists. Structure-activity relationship study indicated that PPARα/γ/δ activities were dependent on the nature of the hydrophobic group, the attachment position of the alkoxy linker to the indole ring, and N-alkylation of indole nitrogen. Some compounds presented significant PPARγ/δ activity and molecular modeling suggested their putative binding modes in the ligand binding domain of PPARγ. Of these, compound 51 was selected for in vivo study via an evaluation of microsomal stability in mouse and human liver. Compound 51 lowered the levels of fasting blood glucose, insulin, and HbA1c without gain in body weight in db/db mice. When compound 51 was treated, hepatic triglycerides level and the size of adipocytes in white adipose tissue of db/db mice were also reduced as opposed to treatment with rosiglitazone. Taken together, compound 51 shows high potential warranting further studies in models for diabetes and related metabolic disorders and may be in use as a chemical tool for the understanding of PPAR biology.
Collapse
Affiliation(s)
- Hyo Jin Gim
- Research Center for Cell Fate Control, College of Pharmacy, Sookmyung Women's University, Chengpa-ro 47-gil 100, Yongsan-gu, Seoul 140-742, Republic of Korea
| | - Hua Li
- Research Center for Cell Fate Control, College of Pharmacy, Sookmyung Women's University, Chengpa-ro 47-gil 100, Yongsan-gu, Seoul 140-742, Republic of Korea
| | - Ji Hye Jeong
- Research Center for Cell Fate Control, College of Pharmacy, Sookmyung Women's University, Chengpa-ro 47-gil 100, Yongsan-gu, Seoul 140-742, Republic of Korea
| | - Su Jeong Lee
- Department of Food and Nutrition, Sookmyung Women's University, Chengpa-ro 47-gil 100, Yongsan-gu, Seoul 140-742, Republic of Korea
| | - Mi-Kyung Sung
- Department of Food and Nutrition, Sookmyung Women's University, Chengpa-ro 47-gil 100, Yongsan-gu, Seoul 140-742, Republic of Korea
| | - Mi-Young Song
- Department of Biochemistry, Chonbuk National University Medical School, Jeonju, Jeonbuk 561-756, Republic of Korea
| | - Byung-Hyun Park
- Department of Biochemistry, Chonbuk National University Medical School, Jeonju, Jeonbuk 561-756, Republic of Korea
| | - Soo Jin Oh
- Bio-Evaluation Center, Korea Research Institute Bioscience and Biotechnology (KRIBB), Ochang, Chungbuk 363-833, Republic of Korea
| | - Jae-Ha Ryu
- Research Center for Cell Fate Control, College of Pharmacy, Sookmyung Women's University, Chengpa-ro 47-gil 100, Yongsan-gu, Seoul 140-742, Republic of Korea
| | - Raok Jeon
- Research Center for Cell Fate Control, College of Pharmacy, Sookmyung Women's University, Chengpa-ro 47-gil 100, Yongsan-gu, Seoul 140-742, Republic of Korea.
| |
Collapse
|
7
|
Gao D, Zhang YL, Xu P, Lin YX, Yang FQ, Liu JH, Zhu HW, Xia ZN. In vitro evaluation of dual agonists for PPARγ/β from the flower of Edgeworthia gardneri (wall.) Meisn. JOURNAL OF ETHNOPHARMACOLOGY 2015; 162:14-19. [PMID: 25557029 DOI: 10.1016/j.jep.2014.12.034] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/17/2014] [Revised: 09/17/2014] [Accepted: 12/22/2014] [Indexed: 06/04/2023]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE In Tibet, the flower of Edgeworthia gardneri (Wall.) Meisn., locally named "Lvluohua, [symbols: see text]", has been traditionally used to treat diabetes mellitus for many years. AIM OF THIS STUDY To evaluate the activity of dual agonists for PPARγ/β from the flower of E.gardneri in vitro. MATERIALS AND METHODS HeLa cells were transiently co-transfected with the re-constructed plasmids of pBIND-PPARγ-LBD or pBIND-PPARβ-LBD and rL4.35. The activities of crude extracts, secondary fractions and compounds from the flower of E.gardneri were evaluated with the transfected cells. Rosiglitazone (at 0.5 μg/mL) and L-165041 (at 0.5 μg/mL) were used as the positive controls for PPARγ and PPARβ respectively. RESULTS The results demonstrated that n-hexane, ethyl acetate and n-butanol extracts from the flower of E.gardneri were able to significantly activate PPARγ and PPARβ respectively, and the activity of ethyl acetate extract was much better. We further observed that, among the 11 secondary fractions of ethyl acetate extract, the fr. 9 could activate PPARγ and PPARβ significantly. Moreover, umbelliferone (from fr.9) and pentadecanoic acid could activate PPARγ and PPARβ at the same time. CONCLUSIONS The extracts from the flower of E.gardneri could significantly activate PPARγ and PPARβ. Besides, umbelliferone and pentadecanoic acid isolated from the flower of E.gardneri were the new agonists for PPARγ and PPARβ.
Collapse
Affiliation(s)
- Die Gao
- College of Chemistry and Chemical Engineering, Chongqing University, Chongqing, 400030, China
| | - Yong-lan Zhang
- College of Chemistry and Chemical Engineering, Chongqing University, Chongqing, 400030, China
| | - Pan Xu
- College of Chemistry and Chemical Engineering, Chongqing University, Chongqing, 400030, China
| | - Ye-xin Lin
- College of Chemistry and Chemical Engineering, Chongqing University, Chongqing, 400030, China
| | - Feng-qing Yang
- College of Chemistry and Chemical Engineering, Chongqing University, Chongqing, 400030, China
| | - Jian-hui Liu
- College of Pharmacy and Bioengineering, Chongqing University of Technology, Chongqing 400054, China
| | - Hai-wen Zhu
- College of Chemistry and Chemical Engineering, Chongqing University, Chongqing, 400030, China
| | - Zhi-ning Xia
- College of Chemistry and Chemical Engineering, Chongqing University, Chongqing, 400030, China.
| |
Collapse
|
8
|
Bhaskar R, Mohanty B. Pesticides in mixture disrupt metabolic regulation: in silico and in vivo analysis of cumulative toxicity of mancozeb and imidacloprid on body weight of mice. Gen Comp Endocrinol 2014; 205:226-34. [PMID: 24530807 DOI: 10.1016/j.ygcen.2014.02.007] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/07/2014] [Accepted: 02/05/2014] [Indexed: 01/14/2023]
Abstract
Pesticides acting as endocrine disrupting chemicals disrupt the homeostasis of body metabolism. The present study elucidated that the low dose coexposure of thyroid disrupting dithiocarbamate fungicide mancozeb (MCZ) and neonicotinoid insecticide imidacloprid (IMI) during lactation increased the risk of body weight gain in mice later in life. Body weight gain has been linked to pesticide-induced hypothyroidism and hyperprolactinemia and alteration of lipid profiles. In vivo results were substantiated with in silico molecular docking (MD) analysis that predicted the binding affinity of pesticides with thyroid hormone receptors (TRα and TRβ) and peroxisome proliferator activated receptor gamma (PPARγ), the major nuclear receptors of peripheral fat metabolism. Binding potency of MCZ and IMI was compared with that of T3, and its antagonist ethylene thiourea (ETU) as well as PPARγ agonist (rosiglitazone) and antagonist (HL005). MD simulation predicted that both MCZ and IMI may compete with T3 for binding with TRs. Imidazole group of IMI formed hydrogen bonds with TRs like that of ETU. MCZ may compete with rosiglitazone and HL005 for PPARγ, but IMI showed no affinity. Thus while both MCZ and IMI could disrupt the TRs functioning, MCZ alone may affect PPARγ. Coexposure of pesticides decreased the plasma thyroid hormones and increased the cholesterol and triglyceride. Individual pesticide exposure in low dose might not exert the threshold response to affect the receptors signaling further to cause hormonal/metabolic impairment. Thus, cumulative response of the mixture of thyroid disrupting pesticides can disrupt metabolic regulation through several pathways and contribute to gain in body weight.
Collapse
Affiliation(s)
- Rakesh Bhaskar
- Department of Zoology, University of Allahabad, Allahabad 211002, India
| | - Banalata Mohanty
- Department of Zoology, University of Allahabad, Allahabad 211002, India.
| |
Collapse
|
9
|
Kozielewicz P, Paradowska K, Erić S, Wawer I, Zloh M. Insights into mechanism of anticancer activity of pentacyclic oxindole alkaloids of Uncaria tomentosa by means of a computational reverse virtual screening and molecular docking approach. MONATSHEFTE FUR CHEMIE 2014. [DOI: 10.1007/s00706-014-1212-y] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
|